Evaluation and Management of the Child and Adult With Fontan Circulation: A Scientific Statement From the American Heart Association
Abstract
It has been 50 years since Francis Fontan pioneered the operation that today bears his name. Initially designed for patients with tricuspid atresia, this procedure is now offered for a vast array of congenital cardiac lesions when a circulation with 2 ventricles cannot be achieved. As a result of technical advances and improvements in patient selection and perioperative management, survival has steadily increased, and it is estimated that patients operated on today may hope for a 30-year survival of >80%. Up to 70 000 patients may be alive worldwide today with Fontan circulation, and this population is expected to double in the next 20 years. In the absence of a subpulmonary ventricle, Fontan circulation is characterized by chronically elevated systemic venous pressures and decreased cardiac output. The addition of this acquired abnormal circulation to innate abnormalities associated with single-ventricle congenital heart disease exposes these patients to a variety of complications. Circulatory failure, ventricular dysfunction, atrioventricular valve regurgitation, arrhythmia, protein-losing enteropathy, and plastic bronchitis are potential complications of the Fontan circulation. Abnormalities in body composition, bone structure, and growth have been detected. Liver fibrosis and renal dysfunction are common and may progress over time. Cognitive, neuropsychological, and behavioral deficits are highly prevalent. As a testimony to the success of the current strategy of care, the proportion of adults with Fontan circulation is increasing. Healthcare providers are ill-prepared to tackle these challenges, as well as specific needs such as contraception and pregnancy in female patients. The role of therapies such as cardiovascular drugs to prevent and treat complications, heart transplantation, and mechanical circulatory support remains undetermined. There is a clear need for consensus on how best to follow up patients with Fontan circulation and to treat their complications. This American Heart Association statement summarizes the current state of knowledge on the Fontan circulation and its consequences. A proposed surveillance testing toolkit provides recommendations for a range of acceptable approaches to follow-up care for the patient with Fontan circulation. Gaps in knowledge and areas for future focus of investigation are highlighted, with the objective of laying the groundwork for creating a normal quality and duration of life for these unique individuals.
As ever-greater numbers of patients with single-ventricle types of congenital heart disease (CHD) survive, this unique population will require continued lifelong medical care, with important personal, societal, and financial impact. The Fontan palliation was introduced in 19681 and improved the survival of patients with all types of single-ventricle anatomy, including those with underdeveloped or absent right or left ventricles.2 The worldwide population of patients with Fontan circulation grew to an estimated 50 000 to 70 000 patients in 2018, with 40% of patients >18 years of age.3 The current estimate of 30-year survival after surgical Fontan completion is ≈85%.4–7 Despite this remarkable improvement in survival, ensuring these patients a normal quality and duration of life is not currently a reality as multiple organ system dysfunction progresses.
Many alterations in the general health of patients with Fontan circulation are related to the biophysics and physiology of their cardiac anatomy and surgery, in addition to psychosocial, neurocognitive, and mental health challenges, which are just being recognized. The hallmark of the Fontan circulation is a sustained, abnormally elevated central venous pressure combined with decreased cardiac output, especially during periods of increased demands, resulting in a cascade of physiological consequences. Clinical hazards faced by patients include progressive fatigue, heart failure, arrhythmias, and end-organ complications such as liver disease, in addition to anxiety and concern about their condition and future. Whereas multiple reports exist characterizing the condition of these patients, there is an important unmet need in the medical community for a more complete understanding of the pathophysiology, origins, and mechanisms of the development of end-organ consequences.8 Evidence-based treatment strategies and preventive management schema are limited, with a need for more widespread consensus within the healthcare community as to how best to serially monitor and manage these patients.
Why Is There a Need for a Statement?
Because patients with Fontan circulation are surviving into adulthood in greater numbers, their management is affected by a relative scarcity of medical knowledge of the pathophysiological origin and optimal care for their unique condition. Potential complications involve not only the heart but also multiple organ systems, including the liver, lungs, brain, bones, and lymphatic system. The end-organ consequences of the Fontan circulation are ubiquitous and usually progressive. Appropriate medical care for these patients requires coordinated input from pediatric and adult congenital cardiologists, as well as multiple subspecialists, many of whom are not familiar with the physiology or the end-organ consequences of the Fontan circulation. A focused effort to increase our knowledge about patients surviving single-ventricle palliative surgeries is therefore critically important at this time.
In a synthesis of the best current evidence and expertise into a single unified source, this American Heart Association (AHA) statement outlines the fundamental cardiovascular and extracardiac physiological challenges faced by the patient with Fontan circulation, defines our current understanding of the end-organ consequences, highlights knowledge gaps in need of further investigational research, and provides a rationale to support diagnostic and therapeutic best practices that will benefit this population as it continues to increase in number and age over the coming years.
Surgical Considerations
The Fontan circulation is based on the premise that a subpulmonary ventricular pump is not compulsory for venous return to cross the pulmonary vascular bed. Rather, pulmonary blood flow, and thus preload to the single ventricle, can be driven by moderately elevated central venous pressure when pulmonary vascular resistance is low enough to permit adequate forward flow under these hemodynamic circumstances. The evolution of the surgical techniques to achieve this unique circulation began with animal studies in the 1940s and culminated in the successful treatment of 2 patients with tricuspid atresia reported in 1971 by Francis Fontan and colleagues9 and the pioneering work by Guillermo Kreutzer and colleagues2 reported in 1973.
Since then, the Fontan procedure has undergone numerous refinements. The initial use of homograft valves was abandoned because of early calcification and pathway obstruction.10 Modifications such as the atrioventricular and atriopulmonary connections that attempted to harness the contractile power of a small right ventricle or the right atrium have been abandoned for total cavopulmonary types of connection. These more recent modifications eliminate massive atrial dilation and the associated energy losses and stasis.11,12 To further bypass the right atrium completely, Marcelletti and colleagues13 introduced a valveless extracardiac conduit between the inferior vena cava and the branch pulmonary artery (extracardiac total cavopulmonary connection [TCPC]; Figure 1) to obviate suture lines within the atrium. Without clear evidence as to which is superior, both lateral tunnel and extracardiac TCPCs are performed by surgeons today according to personal and institutional preferences. Although technically TCPC can be completed as a single-stage operation, because of the survival benefits initially demonstrated with staged palliation in hypoplastic left heart syndrome, nearly all Fontan procedures are currently performed in planned stages with a transitional superior cavopulmonary connection (either the bidirectional Glenn or hemi-Fontan connection).14
A further landmark in the evolution of the Fontan procedure was the introduction of a fenestration between the systemic venous return and the pulmonary venous atrium, which creates a controlled right-to-left shunt. Because fenestration augments ventricular preload and partially offloads systemic venous hypertension, some centers have routinely adopted fenestration for all Fontan procedures, whereas others have used it only in high-risk patients. The primary clinical benefit of a fenestration appears to be in reducing the amount and duration of pleural drainage after the Fontan procedure, thus decreasing the duration of hospital stay. Some centers proceed with planned anticipatory fenestration closure by interventional catheterization during late follow-up.15
Considerable investigative efforts have been devoted to discovering ways to further improve surgical technique and to optimize the circulation. Collaborating with engineers in Milan, Italy, Marc de Leval and coworkers16 used computational fluid dynamics to conceptualize the most optimal cavopulmonary connections to limit flow disturbance and to minimize power loss. Investigations have now demonstrated that the routine use of an 18- to 20-mm conduit for extracardiac TCPC with the avoidance of flow competition between the superior and inferior venous pathways can lead to the lowest power loss; thus, this conduit is a favorable construct.17,18 Through computational fluid dynamics modeling, a new modification using a Y graft to route the inferior venous return has been proposed to minimize power loss with the additional benefit of evenly distributing the hepatic venous flow to the lungs and thereby reducing the impetus for the development of pulmonary arteriovenous malformations (PAVMs).19 However, clinical results with this Y-graft Fontan have been mixed.20 Innovative combined surgical/interventional catheterization or hybrid procedures for Fontan completion have been proposed but adopted in only a few centers.21
While there is no standardized age or weight for proceeding from a superior cavopulmonary circulation to Fontan completion, most centers currently proceed when a patient is 2 to 4 years of age. However, the optimal age at surgery is now being reconsidered in some centers.22,23 Preoperative investigations have traditionally included a combination of echocardiography, cardiac catheterization, and computed tomography or cardiac magnetic resonance. However, with improved early surgical outcomes and better postoperative management, a more patient-specific algorithm has been adopted. In low-risk patients with acceptable ventricular and atrioventricular valve function and absence of residual lesions such as aortic or pulmonary arterial obstruction, some centers now proceed with a Fontan procedure on the basis of an assessment with echocardiography and cardiac magnetic resonance only. Cardiac catheterization can be replaced with an internal jugular line to measure the superior vena cava pressure.24
Many of the so-called Ten Commandments of requisite anatomic and physiological criteria for Fontan suitability published in 1978 by Choussat et al25 have been overturned or revised.26 Some of the unmodifiable characteristics, including chronic lung disease with pulmonary hypertension, severe ventricular systolic or diastolic dysfunction, and recalcitrant pulmonary vein stenosis, may result in a patient being unsuitable for a Fontan procedure. Other risk factors are now considered potentially modifiable. Moderate to severe atrioventricular or semilunar valve regurgitation may require repair or replacement, and branch pulmonary artery stenosis or hypoplasia can be improved by arterioplasty or stenting. Consideration may be given to placement of steroid-eluting epicardial leads at the time of the Fontan procedure in patients with preoperative atrial arrhythmias or underlying sinus or junctional bradycardia because the transvenous route for placement of pacemaker leads can be problematic.27
The short-term and medium-term outcomes after the Fontan procedure are excellent in the current era (Figure 2), with operative mortality approaching 1% and reported transplantation-free survival of 95% and 90% at 5 and 10 years, respectively.4,7,28,29 It is now estimated that the 30-year outcome of those undergoing the Fontan procedure today will be 85% survival.3 Advances in surgical techniques and perioperative care, including the adaptation of a staged protocol for univentricular palliation and better patient selection, have undoubtedly contributed to this remarkable success. Conversely, because the spectrum of ventricular hypoplasia is wide, efforts are currently underway to design creative surgical options in select patients for whom a relatively high-risk biventricular repair may be possible. Long-term outcomes for complex biventricular repairs will need to be compared with those for Fontan circulation, as outlined in this statement.
Survival Outcomes
The Fontan operation is approaching its fifth decade.1 The estimation of long-term outcomes of this population spans up to 35 years, with the ANZFR (Australia and New Zealand Fontan Registry) reporting a 62% survival over that time frame.30 Large variations in survival outcomes have been reported in the literature. These variations can readily be explained by the impact of era, surgical techniques, heterogeneity of physiological features in different forms of a single ventricle, and indications for surgery. The early experience with the Fontan procedure was characterized by evolution of the cavopulmonary connection type. The early atriopulmonary Fontan technique has been shown to be associated with worse long-term outcomes than the more recent versions of the operation, including the lateral tunnel and the extracardiac conduit.31 Likewise, the earlier Bjork modification, which consisted of placement of a conduit between the right atrial and the right hypoplastic ventricular chamber, also has been associated with worse survival. A report from the Mayo Clinic, where the Bjork modification was pioneered, describes a 30-year survival of 43%, in a population that includes these patients. In contrast, the ANZFR, which excludes the Bjork modification and includes an experience that started a decade later, reports a survival of 70% over the same era.6,30 Today, the estimates of 20-year survival for survivors of the Fontan procedure vary between 61% and 85%.4–7 An examination of the survival curves shows that there does not seem to be a sudden or sharp decline in the survival of this population, without acceleration in mortality.3 The attrition is remarkably constant, and the outcomes of the contemporary techniques of Fontan—the lateral tunnel and the extracardiac conduit—are superior to those observed with the first forms of Fontan. We may therefore be in a position to predict that the population operated on today is likely to have reasonably good survival of ≈85% at 30 years.3
We are also more aware of factors affecting outcomes. Having a single right ventricle supporting the Fontan circulation and, in particular, having hypoplastic left heart syndrome, does not adversely affect early survival to this stage, although patients with a hypoplastic left heart seem to be at higher risk for more complications.7,32–34 Male sex, presence of a common atrioventricular valve (AVV), older age at Fontan operation, elevated preoperative and early postoperative pulmonary artery pressures, concomitant surgery at the time of the Fontan, and prolonged pleural effusions after Fontan completion have all been associated with worse late survival.5,6,35–38 Patients developing late complications such as protein-losing enteropathy (PLE), thromboembolic events, or arrhythmias and those with a ventricular pacemaker will have a worse survival.8,39–42 Although survival of this population has improved, the burden of associated physiological constraints and morbidities has remained, with up to 50% having a major adverse event before reaching adulthood.33
An evaluation of survival of a population with Fontan circulation should also now include the impact of cardiac transplantation. Large variation has been observed across centers worldwide in terms of access to heart transplantation.43,44 In a recent report from the Pediatric Heart Network, the proportion of survivors having transplantation within 15 years of the Fontan varied from 0% and 21% across centers.44 In a population-based study in Australia and New Zealand, the overall rate of transplantation for this population remains low at <3 % of patients.43
Physiological Limitations
The Fontan circulation is characterized by the absence of an adequate subpulmonary ventricle. Essentially, the caval veins are connected without a pumping ventricle to the pulmonary arteries.45,46 The cavopulmonary connection effectively creates an upstream flow restriction across the pulmonary circulation, which dominates the circulation.47 The pulmonary circulation is interposed between the venous return of the body and the systemic ventricle, thereby imposing a precardiac flow limitation to the systemic venous system, causing upstream congestion and decreased downstream flow (Figure 3). These 2 features, elevated systemic venous pressure and chronically decreased output, are the root cause of the majority of the pathological impairments of the Fontan circulation.
The Venous Pathway
The normal subpulmonary ventricle (normally the right ventricle) keeps systemic venous and right atrial pressures low at rest and during exertion, accelerates pulmonary flow in a pulsatile manner, and increases cardiac output by both heart rate and augmented stroke volume. Pulmonary pressures can normally increase up to 50 mm Hg (or even more) during exercise, leading to recruitment of lung vessels.48 The normal subpulmonary ventricle will also allow some compensation in disease states such as pulmonary pathway obstruction or any lung disease that may result in increased pulmonary vascular resistance. The absence of a subpulmonary ventricle results in increased venous pressure at rest and at exercise, with a greater dependency on the skeletal and respiratory muscle pumps for preload augmentation during exercise. In addition, lack of pulsatility and marked attenuation of the normal high-pressure high-flow pulmonary arterial circulation during exercise occur. Consequently, there is less recruitment of collapsible pulmonary vessels, especially at low exercise levels. This results in diminished ventricular preload, with decreased volume-induced ventricular stretch downstream during exercise.
Excessive flow restriction in this passive flow system can occur at any level of the Fontan circuit or the lungs: the Fontan venous connections; the pulmonary arteries; the pulmonary capillary network, including precapillary sphincters; the pulmonary veins; or the venoatrial connection. These impediments include, but are not limited to, stenosis, hypoplasia, distortion, vasoconstriction, pulmonary vascular disease, loss or exclusion of large vessels or microvessels, turbulence or competitive flow collision, flow mismatch, and obstruction by external compression. Impediment at any level of this circuit can profoundly affect cardiac output in Fontan circulation, to a greater degree than in a 2-ventricle circulation.
The Ventricle
From even before birth, the single ventricle is subject to abnormal hemodynamic demands and morphological abnormalities that can predispose to systolic or diastolic dysfunction.49 Palliation of the single ventricle with an aortopulmonary shunt or pulmonary artery band in the neonatal period results in increased ventricular volume load, a condition caused by the presence of pulmonary and systemic circulations existing in parallel, with mixing within the single ventricle. Valvular regurgitation (atrioventricular or semilunar) can also add to the hemodynamic stress on the myocardium. As a result of intracardiac mixing, arterial oxygen saturation is low, and the myocardium is subject to a chronically hypoxic environment. A significant proportion of patients with a single ventricle have either morphological right or indeterminate anatomy.50 However, even in those with single left ventricle, normal geometry as in a 2-ventricle system is not preserved. Myocardial fiber arrangement in the single ventricle is often disturbed and myocardial fibrosis is present. As a consequence, ventricular energetics and the mechanics of contraction can be deranged, with ventricular systolic and diastolic dyssynchrony possible.51,52
At the time when the Fontan procedure is performed, the ventricle commonly evolves from being volume overloaded, overgrown, and overstretched to volume deprived with a smaller cavity and increased wall thickness.49,53 In the short term, this can create a ventricular mass-to-volume mismatch that can deleteriously influence both systolic and diastolic function. Shifting volume load and ventricular mass ratios early in the course of surgical reconstruction may also carry long-term implications for ventricular mechanics.54
During exercise, individuals with a normal heart increase their stroke volume by 20% to 50%; in contrast, such an exercise-induced increase may be completely absent in patients with Fontan circulation, most likely as a result of preload restriction.48 Chronic volume deprivation with lack of fiber stretch is known to decrease contractility and to increase muscle stiffness, resulting in increasing filling pressures.55 Moreover, abnormal ventriculo-vascular coupling may further compound ventricular stiffness.56,57
The Lungs
Throughout the first years of life for the child with a single ventricle, the pulmonary vasculature is subjected to multiple alterations likely to affect growth and function. During fetal life, pulmonary flow is altered from normal, and pulmonary vascular development can be affected. Neonatal palliation will frequently result in further alterations (usually an increase) in pulmonary blood flow, whereas the staged cavopulmonary connections will decrease pulmonary blood flow. Flow distribution to the lungs may not be uniform. As a result, acquired hypoplasia or vascular disease of the pulmonary vascular bed may occur. Stenosis resulting from ductal constriction, abnormal connections, or surgical scarring can further compromise the pulmonary architecture. The Fontan circulation itself imposes multiple abnormal physiological features onto the pulmonary vasculature such as chronically decreased volume of flow, minimally to mildly desaturated bronchial flow, increased aortopulmonary collateral flow, suboptimal mixing of inferior and superior caval flow streams, endothelial dysfunction, absence of pulsatility, and absence of episodic high flow and high pressure, as is normally seen during exercise, all required for exercise-associated vessel recruitment and vasodilation. In combination, these factors contribute to a burden of pulmonary vascular trauma, resulting in an elevated and increasing pulmonary vascular resistance, which can itself contribute to Fontan circulatory failure.58,59
Circulatory Failure and Ventricular Dysfunction
Heart Failure
A well-accepted definition of heart failure is the inability of the heart to meet resting and exercise demands at low filling pressures. By such a definition, essentially all patients with Fontan circulation have a physiological form of chronic heart failure from the first day after the operation, yet we know that this circulation sufficiently sustains life for decades. The defining thresholds for characterizing failure in a patient with Fontan circulation differ from other cardiovascular conditions. Within the context of physiological failures inherent in the Fontan circulation, some patients develop classic heart failure symptoms such as fluid retention and exercise intolerance. The consequences of progressive heart failure in the patient with Fontan circulation are substantial and result in cardiac and noncardiac morbidities that affect long-term survival and quality of life.
An important distinction to make is the presence of 2 categories of heart failure in the patient with Fontan circulation with a single ventricle: the classic ventricular pump failure phenomenon with its clinical manifestations and Fontan circulatory failure, which is unique as a physiological consequence of chronically elevated systemic venous pressure and low cardiac output. In other words, symptoms of heart failure may exist despite reasonable ventricular performance of the single ventricle, simply as a consequence of venous hypertension and its end-organ consequences inherent in the pathophysiology of every patient with Fontan circulation.
Within 15 years of the introduction of the first Fontan procedure, early survival improved markedly; however, a 1%/y continuous hazard for failure was present throughout the follow-up period.60 A recent report of patients who underwent the atriopulmonary Fontan indicates that only 45% of patients remain free of death, heart transplantation, or New York Heart Association class III to VI heart failure over a 28-year follow-up period.61 Evidence suggests that long-term survival has improved in patients who have undergone the lateral tunnel or extracardiac conduit. However, circulatory failure remains an important cause of death in this population.31 Clinical heart failure signs and symptoms are among the leading causes of hospital admission in the Fontan population, and once such heart failure is apparent, it is a strong predictor for worse outcomes.44,62 In a report from the Dutch CONCOR registry, mortality in the patient with Fontan circulation admitted with heart failure was 24% at 1 year and 35% at 3 years from presentation.63
Systolic or diastolic dysfunction is the underlying cause of circulatory failure in the general population, and the diagnosis and treatment of affected individuals are focused primarily on measures that evaluate and improve myocardial performance.64 In contrast, the causes of heart failure in the Fontan population are substantially more heterogeneous and complex. They result in varying phenotypes of heart failure that may represent complex interactions among variables such as systolic and diastolic ventricular performance, structural cardiac or valvular abnormalities, rhythm disorders, and the pathophysiology of passive venous flow through the pulmonary circulation.65
Ventricular Function
Systolic ventricular function is relatively preserved in the first decades after the Fontan procedure50 but declines over time.44 In the presence of atrial arrhythmias, ventricular dysfunction can improve once rhythm control is achieved.66 Systolic ventricular dysfunction is present in 40% to 60% of patients with Fontan circulation undergoing evaluation for heart transplantation.67,68 These patients often have the manifestations of chronic low cardiac output, including exercise intolerance, poor somatic growth, and fatigue. Systolic ventricular dysfunction can be difficult to assess in the patient with Fontan circulation with a non–left ventricular systemic ventricle because normal values of functional indexes for the right ventricle or the univentricular heart are not uniformly agreed on or readily available. Cardiac magnetic resonance imaging (MRI) is a standard and reliable means for providing serial information on ventricular performance, valvular function, and flow data in the patient with a single ventricle after Fontan.69 Although 2-dimensional echocardiography is limited in its ability to evaluate a morphologically abnormal ventricle, fair correlation was demonstrated between the 2-dimensional echocardiographic and cardiac MRI determinations of ejection fraction in a large multicenter study.70 Thus, 2-dimensional echocardiography may be adequate to monitor serial changes in ventricular function on a more frequent basis and may complement the application of serial cardiac MRI.
Similar to the adult noncongenital heart failure population, a substantial number of patients with Fontan circulation have evidence of heart failure with preserved ejection fraction. These patients manifest the signs and symptoms of venous congestion and its attendant consequences such as ascites, PLE, and liver cirrhosis, and they may exhibit growth failure as an indicator of low cardiac output.67,68 Diastolic dysfunction can be difficult to assess noninvasively in the single-ventricle population because of the lack of normal values for systemic right ventricles and univentricular hearts. Estimates of pressure-volume loop relationships indicate evidence for diastolic dysfunction early in the postoperative period after the Fontan operation.71 A volume challenge can often elicit “occult” diastolic dysfunction late after Fontan, highlighting the sensitivity of the single-ventricle heart to volume loads.72,73
Ventricular performance can be gauged through functional assessments such as exercise performance. Baseline exercise performance in patients with Fontan circulation is significantly impaired, with a mean maximal peak oxygen consumption of 60% predicted.74,75 In a large multicenter study, no relationship was found between ejection fraction and exercise performance, whereas a lower resting oxygen saturation, lower oxygen pulse, and chronotropic incompetence were associated with worse exercise performance, highlighting the importance of factors other than systolic ventricular function on exercise capacity.75 Key take-away points for heart failure and ventricular dysfunction are listed in Table 1.
Symptoms of heart failure include poor somatic growth, fluid retention including ascites, and exercise intolerance. |
Heart failure can be related to systolic ventricular dysfunction, with important diastolic ventricular dysfunction increasing with age, although symptomatic circulatory failure may occur independently of ventricular function. |
Potential contributors to heart failure include ventricular dysfunction, atrial tachycardia, valvar regurgitation, and volume-loading shunts. |
Cyanosis
The purpose of the Fontan operation is to separate the systemic from the pulmonic venous blood returns, yet some mixing still occurs. Systemic arterial oxygen saturation at rest in room air rarely achieves levels >95% and is commonly in the 90% to 95% range. There are a number of reasons for this mild degree of arterial oxygen desaturation. First, markedly deoxygenated coronary sinus vein blood typically drains into the pulmonary venous chamber and mixes with pulmonary venous blood. Second, nonpulsatile pulmonary arterial blood flow tends to gravitate to lower segments of the lung, whereas pulmonary aeration favors upper segments, resulting in a ventilation-perfusion mismatch. Third, a fenestration in the systemic venous pathway placed at the time of the Fontan operation may remain patent and persist for years, with variable degrees of right-to-left shunting.76,77 Similarly, small communications between the high-pressure systemic venous and lower-pressure pulmonary venous chambers may develop over time, either within the heart or through extracardiac venous collaterals.78,79 With exercise, arterial oxygen saturation often drops to <90% because each of the mechanisms for cyanosis may become exaggerated with activity and the relative magnitude of desaturated lower extremity venous return increases.
Some patients with Fontan circulation demonstrate notably low arterial oxygen saturations (<90%) at rest. This can be caused by the presence of a fenestration or the development of veno-venous collaterals with right-to-left shunting. Veno-venous collaterals may naturally form as a “pop-off” under conditions of normally anticipated systemic venous congestion and pulmonary artery pressure (10–15 mm Hg) or secondary to the development of unusually elevated pulmonary arterial pressures (>15 mm Hg).80 Effective catheter-based techniques are available to close such communications and to improve oxygen saturation.81 However, this may come at the expense of increased impedance to venous return, diminished cardiac output, and decreased oxygen delivery,82,83 all of which can negatively affect end-organ function and patient well-being.84 Decisions must therefore be made on an individual case-by-case basis as to the merits of fenestration closure or veno-venous collateral occlusion, weighing the tradeoff of cyanosis against the potential for deleterious hemodynamic effects.
Patients with Fontan circulation, notably those with heterotaxy syndrome, may develop PAVMs with intrapulmonary shunting and progressive cyanosis.85 This is believed to result from the absence of an essential hepatic factor reaching the pulmonary vascular bed, a circulating agent that mitigates the development of PAVM. The precise nature or biochemical makeup of this agent is currently unknown, but it may also be the missing agent in conditions such as hepatopulmonary syndrome and hereditary hemorrhagic telangiectasia.86 Preferential shunting of hepatic venous effluent away from one lung, as present in some forms of heterotaxy syndrome with venous anomalies, can lead to the development of PAVMs, whereas re-establishing hepatic venous effluent flow to the affected lung can result in resolution.87–89 Creation of a brachial or axillary arteriovenous fistula may also deliver the hepatic factor to an affected lung and resolve PAVMs.90 MRI-derived computational fluid dynamic studies can be helpful in designing cavopulmonary connections that allow equitable hepatic venous effluent flow between the lungs to prevent PAVMs and in designing surgical venous rerouting strategies to treat this condition.91,92
Because of the mild cyanosis seen in most patients with Fontan circulation, hemoglobin levels are typically elevated. Normal or low hemoglobin values, although perhaps well tolerated in the patient with normal circulation, may be inadequate for the patient with Fontan circulation. Hemoglobin production is essential as a proper compensatory response to the mild cyanosis seen. Hence, maintenance of good iron stores is important.93 An increased hemoglobin level increases blood viscosity and indicates the need for maintenance of proper fluid intake and hydration at all times.
AVV Regurgitation
Significant AVV regurgitation in patients with univentricular hearts was historically a contraindication for staged single-ventricle palliation.25,94 More frequently, AVV regurgitation develops insidiously during follow-up at any stage of single-ventricle palliation. A continuous decline in AVV function in patients with Fontan circulation with the passing of time has been reported in a subset of patients with Fontan circulation.95 Significant AVV regurgitation can eventually compromise the functioning of the Fontan circulation as a result of volume overload, ventricular dilatation, reduced ventricular contractility, and increased postcapillary and central venous pressures. Mechanisms of AVV regurgitation in a univentricular heart are multifactorial and include prior volume overload with a systemic-to-pulmonary shunt, annular dilation, and abnormal chordae and papillary muscles.96,97 Changes in loading conditions at different stages of surgery may lead to dysfunction of chordae and valvular apparatus, structures that may be inherently abnormal and prone to dysfunction from the very start. The tricuspid valve as the systemic AVV appears to be more prone to functional regurgitation than the mitral valve, with further dysfunction potential in those with a common AVV.98 Reduced ventricular function is also associated with the development of AVV regurgitation.
Medical management of patients with Fontan circulation with AVV regurgitation may include afterload reduction and diuretic therapy. These agents may be helpful, but a strong evidence base for their use is lacking. Afterload reduction therapy is often used empirically in patients with a single ventricle before Fontan at various stages of palliation, but there are no data from prospective studies to support the use of these agents. In a multicenter randomized trial, enalapril administration in infants with a single ventricle did not improve somatic growth, ventricular function, or severity of heart failure.99 Similarly, there are no data to support the use of afterload reduction to treat AVV regurgitation. Diuretics may be beneficial for patients with Fontan circulation with features of volume overload, but these agents should be used with caution because they can decrease cardiac output.
Application of volume-unloading staged procedures and various AVV repair techniques during the staged palliation made Fontan completion among those patients achievable. Despite continued improvement in outcomes for patients at higher risk, AVV repair remains a significant risk factor for long-term mortality after the Fontan procedure.100
Debate has been ongoing on the best timing of AVV repair in patients with a univentricular heart and on the need for AVV repair in patients with mild regurgitation. Valve repair in cases of moderate or severe regurgitation either at the time of planned surgery or between stages seems beneficial to prevent deterioration of ventricular function. Whereas surgical feasibility is well established and a variety of techniques have been described, few studies have investigated the midterm and long-term outcomes after AVV repair in patients with single-ventricle heart disease.101,102 Despite the known negative influence of AVV regurgitation on outcomes, recent studies have reported improved outcomes for patients with a single ventricle with successful AVV repair and good ventricular function.103 Key take-away points for AVV regurgitation are listed in Table 2.
Significant AVV regurgitation may be related to structural valvar abnormalities, particularly of the tricuspid valve, annular dilatation, or reduced ventricular contractility. |
Moderate or greater AVV regurgitation is a significant risk factor for long-term mortality after Fontan surgery. |
Diuretics and afterload-reducing medications for AVV regurgitation require careful monitoring to avoid preload depletion. |
AVV indicates atrioventricular valve.
Arrhythmias
Each anatomic substrate of single-ventricle CHD and each type of surgical modification of the Fontan operation may pose its own arrhythmia-producing challenge. The various suture lines of any of the surgical techniques interfere with atrial conduction. The negative impact of the development of tachycardia on outcomes for patients with Fontan circulation has been most clearly demonstrated for adults with atriopulmonary repairs. Among a series of 321 adult patients, those with prior atriopulmonary repairs and receiving diuretics who developed atrial tachycardia had a 6-fold increase in death or transplantation and 6-year mortality of >40%.62 Patients with Fontan circulation with atrial tachycardia are more likely to develop right atrial thrombus, heart failure, and ventricular dysfunction and to require hospitalization.104–108
Traditionally, sinus node dysfunction and atrial tachycardia have been attributed to injury to the sinus node or its arterial supply, atrial suture lines, atrial dilatation, and hypertrophy related to elevated atrial pressures.109 Important causes of late arrhythmia include the underlying genetics of the congenital defect with associated altered atrial tissue organization and programmed cell apoptosis, as well as the effects of neonatal cyanosis and cardiopulmonary bypass with injury to atrial and ventricular tissues, resulting in fibrosis and altered conduction properties. The method of reporting of the existence of arrhythmia varies significantly and is associated with the method of detection, which may include patient self-reporting, the need for intervention to terminate tachycardia or to implant a pacemaker, resting ECG, 24-hour ambulatory rhythm monitoring, or device interrogation. Finally, follow-up for >10 years after surgery is needed to assess the arrhythmia impact of one type of Fontan procedure versus another.
Table 3 summarizes the prevalence of various arrhythmias and pacemaker placement in different types of Fontan surgeries in studies reporting follow-up beyond 5 years.5,6,31,104,105,110–124 Contemporary surgical outcomes appear to have improved tachycardia outcomes.5,31,115 However, the duration of follow-up may not be adequate to make this conclusion because the median postoperative interval preceding the development of atrial tachycardia is 9 to 14 years and the time-related risk increases steadily thereafter.116,119,125 In the Australia/New Zealand study of 1006 patients, which reported a median follow-up of ≈10 years, supraventricular tachycardia was present in 10% of patients.5 In 2 large studies of adults with predominantly atriopulmonary repairs and follow-up extending from 15 to 30 years, freedom from supraventricular tachycardia at 25 years was ≈45%.116,117 Nonsustained and, less frequently, sustained ventricular tachycardia and sudden death are reported in 2% to 12% of patients; the time-related risk of sudden death appears to be 0.15%/y to 0.2%/y.5,6,110,118,124 The reported prevalence of implantation of defibrillators for secondary prevention is 1.4% to 2 % of patients.5,44,118
Arrhythmia | Atriopulmonary | Lateral Tunnel | Extracardiac Connection |
---|---|---|---|
Sinus node dysfunction, % | 30–62 | 18–44 | 15–44 |
Supraventricular tachycardia, % | 60–80 | 7–33 | 5–15 |
Pacemaker, % | 11–23 | 5–55 | 3–27 |
Automated implantable cardioverter-defibrillator, % | 1.4 | ||
Ventricular tachycardia or sudden death, % (%/y) | 2–10 (0.15–0.2) |
Risk factors for the late development of arrhythmias can be summarized from the previous studies and include older age at the time of primary Fontan, atriopulmonary-type repair, preoperative and early postoperative tachycardia, moderate or greater AVV regurgitation, and longer follow-up.5,101,116,117,123 Male sex, right atrial isomerism, and loss of sinus rhythm during follow-up are reported risk factors for arrhythmia in some studies.111,117,118,126–128
Assessing the importance of sinus rhythm and chronotropic incompetence, or the ability to appropriately increase the sinus rate with exertion, can be difficult.120,129 Sinus node dysfunction is common and is associated with longer follow-up, the atriopulmonary technique, and the later development of atrial tachycardia. Autonomic control of heart rate is impaired in patients with Fontan circulation, with lower heart rate variability and depressed baroreflex sensitivity.130,131 The increase in heart rate with exertion is an important means of augmenting cardiac output during periods of increased demand, and impaired responses have been reported in 62% of patients with Fontan circulation, the highest prevalence among adults with CHD.62 A combination of decreased heart reserve and peak oxygen consumption on exercise testing, or chronotropic incompetence, identifies patients at increased risk for early mortality.62,132–135 These studies emphasize the importance of serial exercise testing in older patients with Fontan circulation and highlight a potential role for consideration of early atrial rate-responsive pacing to augment the heart rate response to exertion.
Some surgical studies have reported that the presence of a pacemaker is a risk factor for death, transplantation, or Fontan failure.61,136 The Pediatric Heart Network has serially evaluated a cohort of young patients with Fontan surgeries, largely total cavopulmonary repairs.44,114,137 Pacemakers were required in 13% of patients, and pacing was reported to be associated with lower quality-of-life scores and lower ventricular ejection fraction, with no differences found in exercise performance.137 The indication for pacing (sinus bradycardia or atrioventricular block, congenital or postsurgical) and the type of pacing (atrial, ventricular, or dual chamber) are not reported or analyzed separately. Pacing without atrioventricular synchrony and rate optimization and pacing only the ventricle can be expected to negatively affect hemodynamic status in the patient with a single ventricle.138
Therapies for atrial tachycardia include antiarrhythmic medications, catheter ablation, atrial antibradycardia and antitachycardia pacing, and Fontan conversion with arrhythmia surgery.139 Patients with Fontan circulation presenting with arrhythmias are recommended to undergo assessment of associated hemodynamic abnormalities, with interventional therapy as indicated, in addition to anticoagulation; termination of tachycardia expeditiously is recommended.140,141 The use of antiarrhythmic medications, including β-blockers, sotalol, dofetilide, and amiodarone, is associated with a high recurrence of atrial tachycardia and, in 1 study, an increased risk of adverse events compared with ablation or surgery.142 Flecainide should be used with caution in patients with structural heart disease, and long-term use of amiodarone is associated with thyroid dysfunction.141 Catheter ablation of atrial reentry tachycardia in patients with atriopulmonary Fontan is associated with acute success rates of 54% to 94%, lower than in other forms of heart disease, and early recurrence of atrial tachycardia in ≈50% of patients.143–147
In the more contemporary lateral tunnel and extracardiac repairs, focal atrial tachycardia is present in at least 10% to 15% of patients with atrial tachycardia and is amenable to catheter ablation, although vascular access may be challenging.147–149 In patients with atriopulmonary Fontan repairs, atrial tachycardia is most commonly associated with marked atrial dilatation and hypertrophy, with impaired hemodynamics. An approach consisting of a Fontan conversion to an extracardiac Fontan combined with arrhythmia surgery has been shown to reduce arrhythmia recurrence and to improve hemodynamic status.125,139,150–155 In patients with Fontan surgeries undergoing intracardiac reoperations, surgical ablation for atrial tachycardia is recommended in the most recent guidelines and consensus statements.140,141 Currently, high-risk older patients presenting with significant arrhythmias and ascites, significant AVV regurgitation, or advanced ventricular dysfunction are likely to be considered for heart transplantation rather than Fontan conversion surgery.125,155,156
Future directions for reducing the incidence of arrhythmias include prophylactic interventions at the time of Fontan surgery, which may include placement of isthmus or right atrial cryoablative lesions, or atrial antibradycardia pacing lead implantation.157–161 Early and prompt intervention for residual hemodynamic issues is essential to limit hypertrophy and ventricular dysfunction.140,141 Obesity contributes to the development of atrial fibrillation and diastolic dysfunction and represents a modifiable risk factor to improve outcomes.162,163 Regular physical exercise may have a positive impact on autonomic nervous function, heart rate reserve, and, in turn, arrhythmia propensity and outcome, although this is speculative.120,131 Key take-away points for arrhythmias are listed in Table 4.
Atrial tachycardia develops in >50% of patients late after Fontan, although current surgical techniques may reduce this incidence. |
Development of atrial tachycardia in patients with Fontan repairs is associated with significant morbidity, including heart failure and atrial thrombosis, and requires urgent and aggressive therapy. |
Ventricular pacing may negatively affect cardiac output and lower quality-of-life scores. |
PLE and Plastic Bronchitis
Protein-Losing Enteropathy
PLE is the abnormal loss of serum proteins into the intestinal lumen. It occurs in 5% to 12% of individuals after Fontan palliation.39,44,164 Morbidity stemming from the disease is marked, and although improved in the current era, mortality remains high.164–166
The pathophysiology of PLE remains incompletely characterized, but several factors appear to contribute in varying combinations for each individual case. Recent investigation into lymphatic anatomy and function in patients with PLE has revealed an increase in abnormal lymphatic vessels throughout the intestinal tract.167 Lymphatic congestion, likely present in every patient with Fontan circulation, can lead to overflow or leak of these channels, resulting in spillage of lymph-rich material into the low-pressure intestinal tract and thus enteric protein loss. Lymphatic leak may be worsened by several factors, including the degree of pressure elevation in the systemic veins into which the lymphatic vessels must drain.167,168 Variability in the anatomic configuration of the lymphatic system may predispose to intestinal or bronchial lymphatic leakage when chronic venous congestion is present. For example, some individuals may have lymphatic channels and decompression pathways that run adjacent to the gut lumen or airway and, when in the presence of elevated venous pressure and lymphatic congestion, manifest spillage into these low-pressure passages. This may explain why only select patients with similar degrees of venous hypertension develop PLE or plastic bronchitis (PB) and others do not. Exaggeration of elevated pressure secondary to anatomic obstruction or cardiac dysfunction can accentuate the problem of luminal protein loss.164,169 Altered intestinal mucosal perfusion may also play a role. Chronically limited cardiac output demonstrated in patients with Fontan circulation can result in a state of persistently elevated systemic vascular resistance with redistribution of blood flow, along the lines of chronic heart failure. A selective increase in mesenteric vascular resistance may be present as part of flow redistribution. Increased mesenteric vascular resistance has been demonstrated in patients with Fontan circulation and is also increased in those with PLE.170 Furthermore, chronically low cardiac output leads to a proinflammatory state that may alter enterocyte membrane permeability, predisposing to PLE.171 Finally, a subset of patients with PLE have diminished HSPGs (heparan sulfate proteoglycans), a protein present in the enterocyte membrane that normally assists in enteral protein trafficking, and its absence can result in protein leakage.172 The overall result of these derangements is leakage or spillover of the intestinal lymphatics and loss of protein into the intestinal lumen. As a result of protein loss, serum oncotic pressure decreases, leading to systemic edema. When the intestinal wall becomes edematous, malabsorption occurs, and protein loss can worsen, leading to a vicious cycle of intestinal symptoms and edema often referred to as a PLE flare. These sporadic periods of worsening PLE often follow a viral infection, further suggesting that inflammation plays a role in bowel wall leak in this condition.
PLE often persists as a chronic condition for extended periods of months to years. Subclinical PLE may also exist with low levels of enteric protein loss that may not manifest as ostensible tissue edema but may nevertheless contribute to perturbations in somatic growth and development of abnormalities in the coagulation cascade, processes that depend on proper protein balance. Transient PLE has been identified in select patients with spontaneous remission and periods of wellness for extended periods of time. However, when sustained PLE is present, complications from chronic protein loss lead to significant systemic morbidity. Chronic diffuse edema leads to symptoms of abdominal and extremity swelling, as well as diminished tissue integrity and difficulty in wound healing.173 Coagulation abnormalities result from dysregulation of clotting mechanisms (eg, factor loss or altered production), with an increased risk for thromboembolic complications above and beyond the already increased risk present in every patient with Fontan circulation. Bone density can be profoundly diminished as a consequence of chronic hypocalcemia secondary to low levels of albumin, an essential protein for calcium transport.174 Linear growth failure occurs frequently.173 Immune abnormalities, including low immunoglobulin levels and lymphopenia with profoundly low CD4 T-cell counts, are typically present. Individuals with PLE have a greater likelihood and increased persistence of cutaneous virally mediated processes such as warts and molluscum contagiosum.175,176 Emerging data suggest that lymphopenia may be present in a substantial number of patients with Fontan circulation without clinically overt PLE and that lymphopenia worsens with greater duration from the Fontan operation. The process of chronic low-level lymphocyte loss resulting from enteric lymphatic congestion and intestinal spillage may explain this finding. Gastrointestinal bleeding can also occur in PLE, with resulting anemia worsening the clinical symptoms.177
The gold standard test to diagnose PLE is an elevated α-1 antitrypsin clearance in a 24-hour stool collection.178 Given the challenges in collecting adequate specimen samples for this test, it is also acceptable to base a PLE diagnosis on an elevated α-1 antitrypsin level in a single stool sample together with the presence of serum hypoalbuminemia and symptoms of edema without another identified cause. Tracking serum albumin periodically may detect laboratory-based subclinical albumin loss, act as a marker for subtle deterioration of hemodynamics and strain on a congested lymphatic system, and gauge nutritional status before the onset of full-blown clinical PLE. After a confirmed PLE diagnosis, evaluation for potentially correctable derangements in the Fontan circulation should be undertaken with appropriate assessments.
At this time, treatment of PLE is primarily symptomatic. Heart transplantation has been reported to be an effective strategy in several reports. However, the ravages of long-standing PLE can lead to chronically ill, poorly nourished, frail individuals, a common picture for those with PLE. These patients are often poor candidates for transplantation and may not survive the wait for an organ. The mainstay of symptomatic treatment is aimed at decreasing fluid overload, with aggressive diuresis often augmented by intravenous albumin replacement to replenish oncotic pressure and to drive fluid out of tissue and into the vascular space. Even after a flare is resolved, most individuals will require ongoing maintenance diuretics, and some will require ongoing long-term albumin replacement.173 In addition to being useful for sparing renal potassium losses with diuresis, spironolactone may improve symptoms via improved natriuresis or as an anti-inflammatory agent and is often included in a maintenance regimen.179,180 Dietary modifications, including a high-protein, low-fat diet with a greater-than-normal proportion of medium-chain triglycerides, may be considered as an adjunct to symptomatic therapy,181 although diet alone is typically insufficient to overcome the process. Diarrhea may be present primarily as a result of intestinal lymphatic leak or secondary to gut microbial overgrowth, with loperamide therapy anecdotally reported as helpful.182 Intravenous immunoglobulin may be administered to raise oncotic pressure in addition to replacing chronically low serum immunoglobulin. Although there are reports of PLE remission associated with intravenous immunoglobulin administration,183 this response to treatment is not widespread. Enteral corticosteroids, most often oral controlled-release budesonide, have been demonstrated to help maintain serum albumin levels and to decrease symptoms.184,185 Although helpful in select patients, steroid therapy can result in cushingoid features, hypertension, dyslipidemia, and suppression of the adrenocortical axis and may contribute to poor growth, poor tissue integrity, and bone demineralization. A strategy of a time-limited course of high-dose steroids with weaning to low maintenance levels can be considered. Phosphodiesterase-5 inhibitors can be used to lower Fontan pathway pressures and to decrease mesenteric vascular resistance, thereby potentially increasing normal lymphatic drainage and improving tissue perfusion, but their efficacy in PLE is not well established.186 Exogenous unfractionated heparin, either intravenous or subcutaneous, has been reported to induce remission, perhaps in the select subset of individuals with PLE complicated by abnormal or absent cell membrane heparin sulfate proteoglycan. A trial of heparin therapy may therefore be considered, but as with other therapies, if improvement of the disease state is not achieved in the time span of weeks to months, it should be discontinued.187,188 Dopamine infusion appears to induce remission of protein loss in a small series of patients bridged to transplantation.189
Invasive procedures may provide the best opportunity for true disease improvement and should be considered on a case-by-case basis. Some individuals with PLE improve with transcatheter fenestration creation, which reduces impedance to forward venous flow at the expense of increased cyanosis resulting from right-to-left shunting.169,190 Lymphatic interventional procedures aimed at interrupting lymphointestinal connections originating from the liver have achieved remission in a small number of patients with PLE and appear promising.167 Surgical innominate vein exclusion with rerouting of the innominate vein and the thoracic ductal return directly to the low-pressure pulmonary venous atrium has resulted in remission of PLE at the expense of increased cyanosis.191 Lymphatic pathway drainage-altering therapies may prove to be safe and effective strategies for management of PLE as these procedures evolve and more experience is broadly achieved. Currently, such therapies are most commonly considered in the patient with PLE deemed at high risk or thought not to be a candidate for heart transplantation. To date, only orthotopic heart transplantation has been demonstrated to consistently and effectively treat PLE, with the tradeoff of a relatively high, but in most cases acceptable, risk of mortality.192 The precise timing of referral for invasive procedures such as fenestration creation, catheter-based lymphatic intervention, surgical thoracic duct rerouting, and heart transplantation remains a challenge when balanced against the potential efficacy of medical management.
Plastic Bronchitis
PB is a disease characterized by the production of thick, tenacious casts within the airway lumen. It is reported to occur in <5% of individuals with Fontan circulation, although the prevalence of subclinical PB may be higher.166,193 It confers significant morbidity, most often chronic cough and hypoxemia, with expectoration of proteinaceous casts and increased risk of mortality.194 Fontan-associated PB is believed to be caused by the spillage of protein-rich lymph through lymphatic-to-bronchial communications.195–197 Both PLE and PB may be considered part of the clinical spectrum of lymphatic insufficiency associated with chronically elevated venous pressure. Lymphatic system overflow and congestion are in search of natural decompression. In select patients, perhaps related to an individual’s unique lymphatic vascular architecture, lymph decompresses into the airway lumen, causing PB. In PB, an airway inflammatory reaction, precipitated either via a concurrent respiratory infection or secondary to individual abnormalities of inflammatory response, results in fibrin cross-linking within the lymph, causing it to coagulate into a rubbery, solid cast.198 Bronchial casts can partially or completely obstruct the airway in which it forms and in severe cases may lead to severe hypoxemia, asphyxiation, and death.
PB is diagnosed after production of an airway cast either by patient expectoration or via bronchoscopic removal. Although not diagnostic, symptoms of chronic cough, wheezing, or respiratory symptoms not responsive to bronchodilator therapy in a patient with Fontan circulation may prompt consideration of possible PB. Imaging with MRI T2-weighted techniques that allow visualization of high-water-content structures may help define patterns of lymphatic vascular architecture within the chest or abdomen either before or shortly after the Fontan operation that may be more highly associated with PB or PLE.199
Initial treatment should target symptomatic therapies aimed at airway clearance. However, a vigorous investigation of the state of the Fontan circulation is warranted to rule out cardiovascular anatomic considerations. Similar to PLE, pulmonary vasodilator therapy and diuretic treatment, particularly with aldosterone inhibition, can be helpful.200 Treatment with bronchodilators and aggressive chest physiotherapy should be started for any patient regularly producing casts.200,201 Anti-inflammatory therapy with inhaled steroids may be considered, although the effectiveness of this therapy is unclear.202 Therapy with mucolytics such as dornase-α or N-acetylcysteine has been reported to be efficacious in improving cast mobilization in small series and may be considered.201 Nebulized and inhaled tissue plasminogen activator therapy can be very effective in clearing obstructive airway material in acutely symptomatic individuals because it targets the fibrin cross-links that lead to cast formation.203,204 Long-term therapy with tissue plasminogen activator for cast prevention has also been reported.200 In addition to medical therapy, dietary modifications that decrease lymphatic flow such as a low-fat diet have been associated with symptomatic improvement or resolution in some patients.205 Any significant hypoxemia, respiratory distress, or shock in a patient with PB should be treated with urgent bronchoscopy with the goal of clearing the obstructed airway.206 Until recently, the most effective therapy for PB was heart transplantation and elimination of the Fontan circulation. Survival after heart transplantation for PB is relatively favorable and commonly results in disease remission.207 Currently, catheter-based intervention to embolize and eliminate lympho-bronchial airway communications has resulted in durable remission of the disease with excellent outcomes, including low morbidity and mortality.197 Catheter-based occlusion of lymphatic leaks in PB can be lifesaving; however, such interventions do not influence the underlying deleterious pathophysiology of the Fontan circulation. Therapies targeting venous and lymphatic decongestion with diuretics and pulmonary vasodilators are often still necessary to optimize the circulation in hopes of preventing recurrence. Table 5 lists the various management and treatment strategies for PLE and PB. Key take-away points for PLE and PB are listed in Table 6.
Condition | Strategy | Objective | |
---|---|---|---|
Diagnostic evaluation | |||
PLE and PB | Cardiac imaging (echocardiography, MRI, or CT) and cardiac catheterization | Identify anatomic problems and measure hemodynamics (eg, cardiac output, central venous pressure) | |
PLE and PB | Lymphatic imaging* (T2-weighted MRI, lymphangiography) | Characterize state of lymphatic vasculature | Identify possible decompression channels into gut or airway lumen |
PLE and PB | ECG and Holter monitoring | Identify hemodynamically important arrhythmia | |
Medical management | |||
PLE and PB | Diuretics | Reduce venous and lymphatic congestion | Treat peripheral edema and ascites (PLE) |
PLE and PB | Aldosterone inhibition | May have direct effects on lymphatic decongestion | May improve ventricular function and treat heart failure |
PLE and PB | Pulmonary vasodilators (phosphodiesterase-5 inhibitors, endothelin-1 inhibitors) | Lower impedance to forward systemic venous flow through the pulmonary vascular circuit, thereby reducing lymphatic congestion | |
PLE | Heparin | Reduce intestinal membrane permeability | Reduce intestinal inflammation |
PLE | Corticosteroid (oral controlled-release budesonide) | Reduce intestinal inflammation and stabilize cell membranes | Restore integrity to intestinal permeability barrier |
PB | Aerosolized tissue plasminogen activator | Break down bronchial casts to ease expectoration or to reduce production | |
Interventional/surgical management | |||
PB | Thoracic duct ligation | Redirect lymphatic drainage away from airway lumen decompression channels | |
PLE and PB | Cardiac pacing | Maintain appropriate heart rate and cardiac output in conditions of heart block or sick sinus node disease | |
PLE and PB | Fontan baffle fenestration | Lower impedance to forward systemic venous flow | May increase cardiac output and oxygen delivery but at expense of cyanosis |
PLE and PB | Catheter-based lymphatic intervention | Identify pathways of lymphatic decompression into gut lumen or airway and allow potential occlusion of abnormal decompression channels | |
PLE | Surgical rerouting of innominate vein/thoracic duct to left atrium | Eliminate impedance to lymphatic drainage | Decompress lymphatic system into low-pressure atrium but at the expense of cyanosis |
PLE and PB | Fontan takedown to Glenn or shunt state | Alter hemodynamics to reduce lower body systemic venous pressure and to reduce lymphatic congestion | |
PLE and PB | Heart transplantation | Alter hemodynamics by reducing systemic venous hypertension and improving cardiac output |
CT indicates computed tomography; MRI, magnetic resonance imaging; PB, plastic bronchitis; and PLE, protein-losing enteropathy.
*
Evolving technology with limited application at this point but with anticipated expansion of use.
PLE and PB are clinical manifestations of chronic venous congestion-induced lymphatic insufficiency with a host of potentially life-threatening consequences. |
PLE may lead to edema and ascites, growth failure, decreased bone density, coagulation abnormalities, and lymphopenia and results in increased risk of death. |
Treatment strategies for PLE and PB include symptomatic management, attempts at improvement of circulatory hemodynamics, manipulation of the lymphatic system, and cardiac transplantation. |
PB indicates plastic bronchitis; and PLE, protein-losing enteropathy.
Liver Fibrosis and Cirrhosis
Liver fibrosis and cirrhosis are growing concerns for the patient with Fontan circulation. Liver injury resulting from cardiac dysfunction has long been recognized, with the earliest documentation occurring in 1833.208
Cardiac failure with circulatory congestion typically manifests as centrilobular hepatic congestion and necrosis, with activation of inflammation in some cases.209 Over time, particularly if within the context of repeated insults, fibrosis may progress to cardiac cirrhosis. Liver involvement in patients with Fontan circulation was brought into prominence after the description of chronic passive congestion, cardiac cirrhosis, hepatocellular carcinoma (HCC), and hepatic adenoma in a series of 9 postmortem patients, all of whom had Fontan-associated liver disease (FALD).210 Abnormalities of the liver can be seen early within the first 5 years after Fontan operation.211 Findings from elective surveillance liver biopsy indicate that all patients with Fontan circulation have some degree of liver fibrosis present and that the severity is related mostly to increasing age at biopsy and increasing time from Fontan operation.212
Pathogenesis
FALD is in part, if not predominantly, related to hepatic flow characteristics unique to the Fontan circulation.213 Infradiaphragmatic hemodynamics change profoundly from the moment the Fontan circulation is installed. Venous pressures rise; lymphatic overflow and congestion ensue; and cardiac output decreases from baseline. Exertional stress exaggerates these phenomena with precipitous increases in central venous pressures to levels ≥25 mm Hg when the individual is awake and active.214,215 The earliest manifestations of these profound hemodynamic changes are reflected in the universal acute increase in liver stiffness. Hepatic stiffness persists during late follow-up in the vast majority.216 The high central venous pressures mimic hepatic venous outlet obstruction, giving rise to arterialization of hepatic blood flow through the so-called hepatic arterial buffer response. The arterial buffer response has long been recognized as an intricate regulatory mechanism through which overall hepatic blood inflow is maintained through autoregulation of the 2 hepatic inflows: hepatic arterial flow (normally contributing ≥25% of inflow) and portal venous flow (normally ≈75% of hepatic inflow).217,218 Hypervascular nodules within the body of the liver are seen frequently during late follow-up but do not necessarily correlate with worse FALD.219 Increased volume of hepatic venous return is associated with a greater degree of fibrosis.220 This may be a consequence of hepatic arterialization associated with worsening fibrosis, thus leading to increased venous return.
Manifestations of FALD
Biological Parameters
Mild elevations in liver enzymes such as ALT (alanine aminotransferase) and AST (aspartate aminotransferase) are very common soon after the Fontan operation without any major ostensible evidence of clinical impact on hepatic synthetic or metabolic dysfunction. In a review of late follow-up, 84% of patients with Fontan circulation demonstrated at least 1 serological liver abnormality, elevated GGT (γ-glutamyl transferase) being the most common and the dominant abnormality in 70% of these cases.222 Of note, in addition to the liver, increased GGT may reflect overall antioxidant inadequacy and increased oxidative stress and may prove to be a biomarker of general instability in cardiovascular disease.223 Over time, there is progressive elevation of predominantly GGT and bilirubin. GGT, for example, increases in 80% of individuals over a 4-year follow-up period. Total bilirubin levels also increase with age. A rise in prothrombin time and international normalized ratio may be seen, as well as a decrease in platelet count, consistent with increased portal venous pressure and splenic consumption.
Imaging Features
Abdominal imaging, in particular as patients transition into the adolescent and adult years, is important in the patient with Fontan circulation.224 In a study of adults at a median of 26 years of age, hepatic imaging was abnormal in all patients, with a wide spectrum of findings (Figure 5).225 Ultrasound is relatively insensitive for detecting fibrotic changes, although it is able to provide hemodynamic information and gross structural information, which can be important in the serial evaluation of patients. Ultrasound identifies increased liver volumes in the early phases of congestive hepatopathy or diminished volumes associated with relative caudate lobe hypertrophy as fibrosis progresses to cirrhosis. As central venous pressures or trans-hepatic resistance increases, ultrasound can characterize a number of important findings: the presence or absence of anterograde portal venous flow, enlargement of the hepatic artery as arterialization of hepatic flow begins, and changes in celiac and mesenteric flow resistances from Doppler tracings.
Experience is growing in defining the utility of elastography in the evaluation of the degree of FALD present.226 This promises to be a valuable tool for serial evaluation of the progression of hepatic stiffness over time.227 Acoustic tissue imaging with application of elastography methodology provides additional ultrastructural information on liver stiffness.228 Shear-wave velocity in patients with Fontan circulation is ≈2.2±0.38 m/s compared with a normal value of 1.1±0.29 m/s. MRI liver elastography is also available and can provide detailed characterization of liver stiffness and additional structural information.229
Liver stiffness reflects the combination of liver fibrosis, venous congestion, and altered liver compliance patterns, all of which are present in FALD. Hence, distinguishing between these contributing factors can be a challenge.230 Hydration status influenced elastography measures of liver stiffness in a porcine model.231 Nevertheless, the composite measure of both processes, namely hepatic vascular congestion and liver tissue scarring, may prove to be of clinical value in the longitudinal characterization of FALD.
Computed tomography and MRI provide complementary, detailed, and accurate information about liver morphology and structure and vascular enhancement patterns.232,233 The liver is imaged in multiple consecutive time phases after contrast injection. This yields information on early enhancement in the arterial, portal venous, and late venous phases. Enhancement patterns are demonstrated in Figure 6. Enhancement characteristics of hepatic nodules are particularly useful in advanced cross-sectional imaging. Arterially hyperenhancing nodules, for example, usually represent focal nodular hyperplasia. On the other hand, HCC appears to be hypoenhanced in the hepatic phase; that is, late at ≈20 minutes after contrast injection. New and exciting trends in MRI that offer promising prospects to further liver characterization in FALD include T1 and T2 imaging, diffusion-weighted imaging, and metabolic imaging that incorporates hepatocellular function such as gadonexate disodium hepatocyte–specific agents.234
Histological Changes
In a report of liver biopsies performed as part of a surveillance program in 67 patients with Fontan circulation at mean age of 17±4.5 years, some degree of liver fibrosis and scarring was seen in all.212 A wide spectrum of histological changes is associated with FALD (Table 7). Various degrees of liver fibrosis can be seen on Masson trichrome staining (Figure 7). A variety of liver fibrosis scoring systems exist, developed primarily to grade various liver conditions unrelated to the unique pathophysiology of the Fontan circulation. Recently, a more objective quantification of fibrotic load that uses Sirius red staining of collagen has been proposed. This approach is promising in that it may help standardization of histological grading, which will allow comparative analyses to be applied more widely (Figure 8).235 Liver fibrosis is known to be reversible in some instances. It is unlikely, however, when greater degrees of collagen cross-linkage have occurred; that is, a maturation and strengthening of the collagen matrix that can be detected histologically by orcein positivity. The notion of discovering mechanisms for impairing or halting the progression of histological change in FALD or the potential for reverse remodeling when hemodynamic normality has been restored is of great interest.
Histological Features | Prevalence of Feature, % |
---|---|
Any bridging fibrosis | 45 |
Central vein to vein bridging fibrosis | 30 |
Central vein to portal tract fibrosis | 37 |
Portal to portal tract fibrosis | 10 |
Bile ductular reaction | 24 |
Cholestasis | 1 |
Lobular inflammation | 1 |
Portal inflammation | 15 |
Hepatocellular damage/necrosis | 4 |
Steatosis | 7 |
Hepatocellular Carcinoma
The most extreme manifestation of liver injury associated with the Fontan circulation is the development of HCC. This phenomenon occurs in addition to an overall increased risk of cancer in general in adult patients with repaired complex CHD.236 In a report of 33 patients diagnosed with HCC after a Fontan operation at a median of 30 years of age (range, 12–52 years), the prevalence was documented to be ≈1.3%.237 The prognosis, similar to HCC in other contexts, remains quite poor, with more than half of affected patients dying within 2 years of diagnosis. Those who are detected while asymptomatic appear to have a better prognosis because their treatment options are broader. The association of HCC with cirrhosis and varying degrees of fibrosis needs confirmation in patients with Fontan circulation. The question of whether HCC risk is increased with increasing severity of fibrosis in FALD or whether other factors come into play is unclear. HCC has been observed in patients as young as 12 years of age. Advanced treatment strategies include local resection, chemoembolization, radiofrequency ablation, heart-liver transplantation, or a palliative care approach.238
Clinical Impact of FALD
Liver disease in patients with Fontan circulation is most frequently indolent. Although fortunately quite rare, decompensated cirrhosis may indeed occur with manifestations of encephalopathy, variceal bleed, and portal gastropathy and enteropathy.239 Most often, however, there is a variable degree of fibrosis and compensated cirrhosis. An uncoupling exists between structural/morphological features observed on imaging and histology and liver functional status as defined by synthetic function of the liver. Therefore, imaging features do not necessarily predict immediate clinical outcomes. Thus, clinical decision-making is difficult because critical or functional reserve of the liver cannot currently be accurately ascertained and may be quite satisfactory despite imaging assessment and histological findings. This lack of data becomes especially relevant when major surgical procedures such as revision surgery to the Fontan circuit or cardiac transplantation are being planned. Under these circumstances, clinicians lean heavily on blunt tools that do not directly predict surgical outcomes and are not specific to FALD. These tools may include, for example, the VAST (varices, ascites, splenomegaly, or thrombocytopenia) score, the MELD-XI (Model for End-Stage Liver Disease Excluding INR [international normalized ratio]) score, the Child-Turcotte-Pugh score, or liver damage scores.240–242 More work is required in this area to develop Fontan circulation–specific tools to predict the impact of FALD on surgical outcomes.
Proposed Therapeutic Approaches
All patients with Fontan circulation should undergo regular hepatic screening with assessments that are age dependent and may include laboratory serum screening and imaging (see the Surveillance Strategies: Proposed Schema of How to Monitor the Patient With Fontan Circulation section). Serum α-fetoprotein is a valuable early biomarker for conversion from liver fibrosis to carcinoma and may be incorporated into a regular screening scheme in older patients. A marked or sudden increase in abnormalities of liver parameters should prompt hemodynamic assessment. All potentially important hemodynamic derangements such as significant AVV regurgitation, pathway obstruction, diastolic dysfunction, pulmonary vascular disease, or aortic arch obstruction should be carefully evaluated for possible interventional approaches. Creative surgical strategies such as hepatic vein exclusion, although potentially of short-term promise, are hitherto unproven as a good long-term tactic and may carry adverse consequences such as cyanosis. The use of pharmacological agents such as pulmonary vasodilator therapy, vascular decongestant agents such as diuretics, or antifibrotic therapies such as aldosterone inhibition or angiotensin-converting enzyme (ACE) inhibition is theoretically of potential benefit but as of yet is unproven to prevent the development or to halt the progression of FALD. Considering the ubiquity of FALD, evidence for progression with age, and overall growing concern, study of these strategies is warranted.
General liver preventive health should be advised to all patients with Fontan circulation. These include avoidance of liver toxins when possible, including alcohol and high-dose acetaminophen. The risks and benefits of potential hepatotoxic drugs such as amiodarone should be weighed carefully. Patients with Fontan circulation should be strongly advised to be vaccinated for hepatitis B, to avoid smoking, and to prevent overweight/obesity.243 End-stage hepatic disease with liver cirrhosis is generally associated with diminished systemic vascular resistance, although this has not been extensively studied in the Fontan circulation. Nevertheless, agents that lower systemic vascular resistance (eg, milrinone, dobutamine) should be used with caution because they may be ineffective or potentially harmful in the critically ill patient with cirrhosis. Liver cirrhosis and end-stage disease may profoundly influence liver performance, leading to an increase in serum ammonia and a decrease in circulating albumin because hepatic clearance and synthetic function are affected. Key take-away points for FALD are listed in Table 8.
Variable degrees of hepatomegaly, hepatic congestion, and liver fibrosis are common and may lead to cirrhosis, with elevations of bilirubin and liver enzymes typical. |
Patients with Fontan circulation should be counseled about avoiding hepatotoxins and adverse lifestyle, which may exacerbate the likelihood of liver disease. |
An increased risk of late hepatocellular carcinoma is recognized, indicating the importance of long-term liver surveillance. |
Renal Dysfunction
The kidneys receive a substantial proportion (≈20%–25%) of cardiac output at rest. Therefore, the circulatory abnormalities common in the patient with Fontan circulation will eventually affect the renal system.244 Other factors may contribute to renal dysfunction in patients with Fontan circulation. These may accrue in a cumulative manner and include multiple exposures to nephrotoxic medications, cardiopulmonary bypass runs (inflammation), intravenous iodinated contrast agents, and long-standing cyanosis resulting in cyanotic nephropathy.244–246 Cardiac surgery–associated acute kidney injury is well documented in children and adults after Fontan palliation and revision surgery and can be secondary to prolonged cardiopulmonary bypass times, resulting in hemoglobinuria, perioperative low cardiac output, and systemic hypotension.247–249 In adults who underwent Fontan revision, a higher preoperative creatinine level has been associated with a greater risk of decline in renal function postoperatively.250 Postoperative acute kidney injury occurs in up to 40% of children undergoing Fontan surgery and is associated with longer intensive care unit and overall hospital stay.251 However, the long-term effects of postoperative acute kidney injury on renal function later in life remain unclear.
Chronic kidney disease or renal dysfunction is evident after the Fontan operation once patients reach adolescence and early adulthood, and it often goes undetected. Measured renal function (glomerular filtration rate), as opposed to estimated glomerular filtration rate (eGFR), reveals abnormality in nearly 50%, with one-third of patients >18 years of age demonstrating albuminuria.252 In a younger cohort of children, abnormal renal function detected from eGFR as opposed to direct measures is evident in 10% of patients, which may likely reflect an underestimation of the degree of dysfunction.253
Studies focusing on long-term renal function and biomarkers for surveillance in the patient with Fontan circulation are limited.250,253–257 Consensus is lacking on which circulating and urine biomarkers are optimal to evaluate renal dysfunction as part of routine clinical surveillance. Decreased eGFR is associated with a poorer prognosis in the general adult CHD population.244 The presence of chronic kidney disease is often calculated with the MDRD (Modification of Diet in Renal Disease) equation,258 the Cockcroft-Gault equation,259 or the Chronic Kidney Disease Epidemiology Collaboration formula.260 The MDRD equation is well validated and adjusts for age-related changes in creatinine and cystatin C, with an eGFR >90 mL·min−1·1.73 m−2 considered normal for adults.258 Conversely, the Schwartz and CKiD (Chronic Kidney Disease in Children) formulas are used for children <18 years of age.261 Cystatin C, a protein encoded by CST3, is gaining recognition as a biomarker for renal and cardiovascular disease.256 Equations that include measurements of cystatin C may better predict outcomes compared with creatinine-based eGFR because of the reduced muscle mass noted in the Fontan population.253,256 However, further studies are needed to assess the accuracy and prognostic significance of different eGFRs in the Fontan population, especially in the presence of significant liver disease, which may alter levels of cystatin C.
Proteinuria and microalbuminuria are well-recognized markers of chronic glomerular injury. An increased prevalence of proteinuria and microalbuminuria is reported in cyanotic CHD.262,263 The prevalence is 10% to 37% in patients with Fontan circulation.6,255,264 Despite the lack of evidence supporting the use of ACE inhibitors in patients with Fontan circulation, some investigators noted the use of ACE inhibitors to be associated with a possible renal protective effect, with no patients with proteinuria among those on ACE inhibitors compared with 43% with proteinuria in those patients not on ACE inhibitors.254 Whether proteinuria and microalbuminuria are associated with adverse outcomes, as in other populations, remains unclear.
The renal resistive index is an area of growing interest as a marker for heart failure severity and end-organ damage in adult CHD.265,266 This sonographic index is used to assess renal arterial disease by measuring the peak systolic and end-diastolic velocity with a calculated index normal value of 0.60.266 A study identified that a high renal resistive index (≥0.81) reflects heart failure severity, hepatic and renal function (24-hour creatine clearance), glucose intolerance, and overall mortality in 280 patients with Fontan circulation.266 This noninvasively measured renal resistive index may be a useful biomarker to assess potential end-organ sequelae associated with Fontan physiology.
Serum biomarkers such as creatinine, blood urea nitrogen, and cystatin C are the measures that can be used for regular renal surveillance. Other studies have reported the use of urine biomarkers such as albumin-to-creatinine ratio, KIM-1 (kidney injury molecule 1), NGAL (neutrophil gelatinase-associated lipocalin), and NAG (N-acetyl-glucosaminidase), which may be useful to further distinguish the type of renal dysfunction such as tubulointerstitial injury.256,267 With the increased incidence of renal dysfunction and its unclear impact later in life, monitoring for renal dysfunction through assessments such as blood laboratory evaluations (eg, cystatin C) and assessments for proteinuria in early adolescence is reasonable, with nephrology consultation in the face of early evidence of chronic kidney disease.
Renal protective measures should be advised for all patients with Fontan circulation. These include recognition of the importance of staying hydrated, keeping blood pressure and blood sugar levels under control, avoiding smoking and excessive sodium intake, and preventing overweight/obesity. If chronic kidney disease develops, avoiding prolonged use of nonsteroidal anti-inflammatory drugs and minimizing contrast dye exposure are required because they can further impair renal function. Key take-away points for renal dysfunction are listed in Table 9.
Renal dysfunction is present in at least 10% of younger patients with Fontan circulation, with increasing prevalence in adolescence and adulthood. |
Decreased GFR, proteinuria, and albuminuria appear to develop and progress over time. |
Impaired renal function may often go unnoticed and should be evaluated in the older patient, in particular before invasive procedures or before anesthesia is administered. |
GFR indicates glomerular filtration rate.
Somatic Growth, Bone Health, and Physical Development
The musculoskeletal system is crucially important to long-term health in children who have undergone the Fontan operation. Although many patients are within normal limits for height and weight, deficits in linear height have been associated with diminished functional health status.268 Abnormalities in body composition, bone structure, and growth factors are present in children and adolescents after the Fontan operation.269–271 Little is known about the specific pathophysiological mechanism of how Fontan circulatory physiology affects this important and dynamic system, on top of what may be an abnormal substrate either from a genetic blueprint perspective or as additive to early factors present before the Fontan operation.
Lean muscle mass is known to be abnormally low in children and adolescents with Fontan physiology and is associated with decreased exercise performance.270,272 Vitamin D levels are also abnormally low, and in those with vitamin D deficiency, deficits in lean muscle mass are more profound.270,273 It is not known whether there is something unique about the Fontan circulation that affects the absorption of vitamin D, although one may speculate that alterations in gut circulation may affect essential element and vitamin absorption. Differences in renal perfusion and microcirculation that are present in the Fontan circulation may also affect vitamin D production in the kidney. Understanding the impact of the Fontan operation on vitamin D levels may have implications for simple therapies such as the timing of supplementation to help promote long-term muscle and bone development and to improve strength and physical functioning.
Normal development of cortical bone structure in childhood and adolescence is critical to the lifelong health of the musculoskeletal system. In healthy children, cortical bone thickness increases in association with the increase in lean muscle mass that occurs during normal growth and pubertal development.274,275 Any reduction in the development of cortical bone thickness places children at risk for lifelong osteopenia and associated complications such as an increased likelihood of bone fracture.276 Although linear growth delay has been well described in children who have undergone Fontan palliation,268,277,278 bone structure itself is abnormal in children and adolescents with Fontan physiology.269 In a cohort of 43 patients, virtually all measures of bone health were below the mean value for an age- and sex-matched control cohort. The deficits in periosteal circumference and cortical area persisted even after adjustment for muscle mass deficits, suggesting that altered bone structure, although exacerbated by muscle deficits, is itself a consequence of the Fontan circulation. In a physiological milieu characterized by low cardiac output and elevated central venous pressure, it may not be surprising that bone structure is abnormal, but a recognition of the timing of the abnormality would provide an opportunity to design an intervention to increase bone health before bone structure is set in late adolescence.
In a large series, the bone health of 210 patients with Fontan circulation presenting to a multidisciplinary specialized Fontan clinic was characterized.279 The mean age was 11.8±4.8 years; 90 were female; and 25 had evidence of PLE. Patients underwent dual-energy x-ray absorptiometry (Hologic, Inc, Bedford, MA) for evaluation of bone mineral density and laboratory blood tests to measure parathyroid hormone and vitamin D 25-OH levels. For patients with Fontan circulation without PLE, the mean height z score, bone mineral density z scores for height-adjusted whole body less head, and bone mineral density z scores for height-adjusted lumbar spine were all significantly lower than normal (z=−0.65, −1.18, and −0.22, respectively). For those 25 patients with PLE, z score values were even more dramatically diminished (z=−1.99, −2.4, and −1.29, respectively). Furthermore, parathyroid hormone values were abnormally elevated (>52 pg/mL) in 48% of subjects, and vitamin D was profoundly low (<20 ng/mL) in 15%. Height-adjusted whole body less head bone mineral density z scores correlated inversely with parathyroid hormone levels (P<0.01). These findings suggest that secondary hyperparathyroidism is common in children with Fontan circulation, perhaps related to the postulated alterations in calcium metabolism resulting from changes in renal perfusion or poor absorption at the gut level. Secondary hyperparathyroidism is a factor that may contribute to bone demineralization and poor growth in children with Fontan circulation. As is now recognized in adults with heart failure, parathyroid hormone itself may prove to be a valuable biomarker of heart failure and may be helpful in prognosticating outcomes in those with Fontan circulation.280
An additional finding of importance in the Fontan population is the relationship between the mediators of growth hormone and the degree of heart failure as characterized by circulating levels of serum BNP (brain natriuretic peptide).271 In a cohort of 41 children and young adults with Fontan physiology, a significant negative association was noted between both IGF-1 (insulin-like growth factor 1) and IGFBP-3 (insulin-like growth factor binding protein 3) and serum BNP level. A downregulation of growth in the setting of heart failure is a relationship that might be expected. The relationship also opens up the possibility of theoretically treating heart failure and optimizing Fontan circulatory hemodynamics to reverse the decline in growth factors with a goal of creating healthier bones and improving longitudinal growth. Further understanding of the temporal changes of these factors in relation to the Fontan operation itself could provide an opportunity to intervene earlier to promote both bone health and long-term linear growth.
Obesity in the Patient With Fontan Circulation
The longitudinal development of obesity is similar to that of the general population but poses particularly hazardous consequences for patients with Fontan circulation.281,282 At the time of Fontan surgery, ≈10% of patients are overweight or obese, increasing to 20% to 30% by 12 years of age and 38% to 50% in adulthood.281–284 Similar to other populations, childhood overweight or obesity markedly increases the risk for obesity in adulthood for the Fontan population.282 Obesity imposes uniquely negative factors for the Fontan circulation that contribute to heart failure: increased pulmonary vascular resistance, increased ventricular mass, diastolic and ultimately systolic ventricular dysfunction, atrial fibrillation, and elevated blood pressure.285–287 Obesity contributes to nonalcoholic liver disease and an increased risk of cancer development, including HCC.243,288–291 For the general population, recent data suggest that a body mass index of < 25 kg/m2 is optimal for cardiovascular health, which should also be a target for patients with Fontan circulation.292 The prevention of overweight and obesity may be a critically important and modifiable risk factor for the long-term health of the patient with Fontan circulation.163,287,292,293 Key take-away points for growth and development are listed in Table 10.
Patients with Fontan circulation tend to have short stature relative to the normal population. |
Decreased vitamin D levels, increased levels of parathyroid hormone, low lean muscle mass, and decreased cortical bone mineral density are identified in many patients with Fontan circulation. |
Overweight and obesity are present in up to 50% of adult patients, which contributes to increased morbidity. |
Brain and Neurocognitive Function
Neurological, neurodevelopmental, and behavioral abnormalities have emerged as important comorbidities in survivors of CHD.294–297 In general, risk factors for late psychomotor and neurodevelopmental problems in patients with complex CHD such as a single ventricle include both patient factors (eg, genetic abnormalities, low birth weight, preterm and early-term birth, low socioeconomic status) and medical factors (eg, certain bypass variables such as low hematocrit, perioperative seizures, longer hospital stays, and global morbidity).295,298–303
The potential causes of brain injury in CHD are cumulative and interactive. First, the same genetic abnormalities that cause heart disease can affect brain development.304 In utero, disturbed fetal cerebral hemodynamics can reduce cerebral oxygen delivery and alter brain maturation.305–311 Children with a single ventricle generally undergo multiple operations and cardiac catheterizations, with risks of hypoxic-ischemic injury, macroemboli, and microemboli. Those with hypoplastic left heart syndrome are particularly exposed to preoperative and postoperative hemodynamic instability and complications, longer use of anesthetic agents and sedatives, and prolonged hospital length of stay.312 Cyanosis and inadequate nutrition may also affect neurodevelopment.313,314 The prothrombotic state of individuals with a single ventricle, combined with the potential for right-to-left shunting and the propensity of patients with Fontan circulation for atrial arrhythmias, creates lifelong risk for chronic cerebrovascular microemboli and stroke.41,315,316
Brain MRI studies in fetuses and neonates with complex CHD have shown abnormalities of maturation, volumetric growth, and white matter brain tissue beginning in fetal life.305–311 Given the multitude of risk factors for central nervous system injury, it is not surprising that brain MRI studies in those with Fontan circulation show striking deviations from normal.317–322 In a study that included brain MRIs of 144 adolescents with Fontan circulation, abnormalities on routine anatomic MRI were found in 66% compared with 6% of 105 normal control subjects, with a predominance of focal/multifocal abnormalities.320 Among the 19 patients (13%) with MRI findings of prior stroke, 7 had not previously had a neurological diagnosis; that is, the stroke had been silent. Investigators report that the percentage of patients with single ventricles with brain injury, including nonacute ischemic changes, atrophy, and ventriculomegaly, accumulates throughout staged surgical reconstruction to Fontan.323 In a subset of 53 children in this study who underwent brain MRI 3 to 9 months after the Fontan procedure, periventricular leukomalacia was observed in 28%, definite generalized atrophy was seen in 4%, focal tissue loss and atrophy were noted in 19%, definite ventriculomegaly was observed in 13%, and acute or chronic intracranial hemorrhage was seen in 8%. In 128 preteens and adolescents with Fontan circulation compared with 48 healthy control subjects, investigators documented widespread, significant reductions in cortical and subcortical gray matter volumes and reduced cortical thicknesses on volumetric brain MRI.321 The lower volumes and thicknesses spanned the frontal, temporal, parietal, and occipital lobes and were seen throughout the subcortical gray matter. Finally, an analysis of white matter microstructure in 102 adolescents with Fontan circulation compared with 47 control subjects showed widespread white matter injury involving major fiber tracts involved in cognitive function. The extent of white matter microstructural change was directly associated with lower Full Scale IQ and lower cognitive processing speed.37
Numerous studies over the past 2 decades have identified a high prevalence of cognitive, neuropsychological, and behavioral deficits in adolescents with critical CHD, including patients with Fontan circulation.317,320,324–329 In general, patients exhibit a somewhat lower IQ, lower academic achievement in reading and math, and issues with visual-spatial skills, working memory (holding information and performing operations on it), and processing speed compared with the normative population. In a cross-sectional study of patients with Fontan circulation 10 to 19 years of age, Full Scale IQ (91.6±16.8), mean Reading Composite score (91.9±17.2), and mean Mathematics Composite score (92.0±22.9) were significantly lower than the expected population mean of 100±15.320 Scores on the Full Scale IQ, Reading Composite, and Math Composite were >1 SD below the population mean in 31%, 29%, and 35% of patients and >2 SDs below the expected population mean in 12%, 12%, and 19%, respectively.320 Similarly, scores on the General Memory Index of the Children’s Memory Scale were <1 and 2 SDs below the expected population mean in 34% and 18% of patients; scores on the Wechsler Memory Scale Composite were <1 and 2 SDs lower in 39% and 14% of patients.320 Many individuals have achievement that is lower than expected in view of their Full Scale IQ scores, consistent with a history of a diagnosis of learning disability in more than one-third of adolescents with Fontan circulation.320 As in most patients with CHD with complex disease, patients with Fontan circulation demonstrate particularly weak visual-spatial skills that may affect academic outcomes.300
Executive functioning challenges, attention-deficit/hyperactivity disorder, and social cognition problems have become increasingly recognized in those with Fontan circulation.330 Impairment in executive function is a striking characteristic of many adolescents who have undergone the Fontan procedure.320,331 In a study of 463 children and adolescents, 145 of whom had Fontan circulation, one-third of parents and teachers scored such individuals in the at-risk range for executive dysfunction on the Behavior Rating Inventory of Executive Function. In another study of 156 adolescents with Fontan circulation, one-third were diagnosed with attention-deficit/hyperactivity disorder.332 Executive dysfunction and attention-deficit/hyperactivity disorder are of particular concern because lower childhood self-control is predictive of adult well-being, independently of social class origins and IQ.333 Finally, adolescents who have undergone the Fontan procedure are at significant risk for impaired social cognition. Specifically, adolescents with Fontan circulation have worse scores on tests that assess their ability to interpret the emotions of other people, social situations, and relationships, as well as more difficulty in recognizing their own emotions. In addition, they show more autism-like behaviors than a referent population.320
Risk factors for adverse neurodevelopmental and behavioral outcomes are highly correlated. Those that have emerged as independent risk factors across multiple studies include the presence of genetic disorders, lower birth weight and gestational age, lower socioeconomic status and maternal education, longer total circulatory support time, a greater number of operations and catheterizations, longer hospital length of stay, and a greater number of complications. As the population with Fontan circulation ages, further research is needed to assess the translation of neurodevelopmental and behavioral findings in childhood and adolescence into neurocognitive function in adulthood.297
Recognition and management of the neuropsychological and behavioral impairments that affect many but not all patients with Fontan circulation have important implications for patient education, medical adherence, the transition from pediatric to adult healthcare systems, completion of higher education, and adult employment. Thus, a collaborative approach among patients, their families, the educational system, and both pediatric and adult healthcare teams is key to promoting well-being in the high-risk adult Fontan population. Characterization of abnormalities of brain structure, cognitive and neuropsychological ability, and behavior provides an opportunity not only to better elucidate the causes of disability but also to develop and implement strategies to optimize long-term outcomes. Awareness and recognition of neuropsychological and behavioral impairments in patients with Fontan circulation can facilitate counseling and increase access to diagnostic testing and educational resources. Periodic surveillance screening and evaluation for neurodevelopmental disabilities starting in early life and beyond are recommended.294 Identification of deficits allows appropriate therapies and education and creates opportunities to enhance academic, behavioral, psychosocial, and adaptive functioning, thus allowing each individual to achieve his or her optimal potential. Key take-away points for brain and neurocognitive function are listed in Table 11.
Brain MRI studies have identified widespread abnormalities in white matter microstructure associated with cognitive performance, as well as gray matter. |
Neurodevelopmental disabilities are common. |
Neurocognitive impairment impedes academic achievement in many patients, and evaluation in the early school years is needed to identify those who may benefit from targeted interventions |
MRI indicates magnetic resonance imaging.
Psychosocial Challenges and Advanced Care Planning
Psychosocial Challenges
Living with a chronic health condition carries potential psychosocial impact, and having undergone a Fontan procedure is certainly no exception. Compared with healthy peers, individuals with Fontan physiology face potential psychosocial challenges in childhood, adolescence, and adulthood. As a group, many children and adolescents with CHD are at risk of psychosocial maladjustment, particularly according to parental report.334 A recent study compared the psychiatric and psychosocial status of 156 patients (mean age, 15 years) who had undergone Fontan procedures with that of 111 healthy peers.332 The patient cohort had a significantly higher rate of lifetime psychiatric diagnosis (65% versus 22%); anxiety and attention-deficit/hyperactivity disorder were most common. Furthermore, as a group, patients had worse outcomes on measures of global psychosocial functioning, anxiety, depressive symptoms, posttraumatic stress, and disruptive behavior. Risk factors of poorer psychiatric/psychosocial outcomes include male sex, lower birth weight, longer duration of deep hypothermic circulatory arrest, and lower intelligence.
In adults, a meta-analysis of international studies using survey assessment of psychological distress revealed no consistent evidence of poorer outcomes among adults with CHD of various subtypes, although methodological heterogeneity was evident.335 The results of 3 North American studies, however, suggest that one-third of adults with CHD meet diagnostic criteria for a mood or anxiety disorder when clinical interviews are administered.336–338 Although depression has received much of the attention in the behavioral cardiology literature, adults with CHD are also at risk of elevated anxiety and posttraumatic stress disorder.339–341 Patients’ subjective health status has been more strongly associated with psychosocial outcomes than objective assessment,337,342–345 an important consideration for physicians faced with the symptomatic adult patient with Fontan circulation. Among adolescents and young adults with Fontan physiology, elevated symptoms of depression have been reported (28% with mild symptoms, 32% with moderate symptoms) and demonstrated to be a negative predictor of quality of life.346 Furthermore, more than half of patients endorsed worry about current health, employment, and living independently.347 Anxiety is common; an Italian study revealed that anxiety symptoms were more elevated than depressive symptoms and that more than half of the patients were considered to have possible anxiety.348 MRI of the brain in adolescents with a single ventricle reveals injury in select areas that control anxiety and depression, suggesting a structural basis for the functional deficits seen.349
Qualitative research offers a rich understanding of post-Fontan life. In-depth interviews with adolescents and young adults with Fontan circulation (17–32 years of age) revealed difficulties and challenges, as well as 3 positive themes: feeling happiness in being themselves (eg, feeling proud, mature, healthy, and special), focusing on possibilities (eg, managing physical restrictions and medical procedures), and being committed to life (eg, living with uncertainty and making the most out of life).350
Improving the understanding and management of cognitive and psychiatric outcomes in adults with CHD was identified as a research priority in a working group of the National Heart, Lung, and Blood Institute and the Adult Congenital Heart Association.351 Patients who have undergone Fontan repairs represent a subgroup of patients with CHD for whom increased research and clinical resources targeting psychosocial outcomes are paramount. This is a group for whom the question is typically when, not if, significant medical sequelae will present, and the complex interplay between physical and psychosocial outcomes warrants attention. Congenital cardiology teams are encouraged to be proactive in their approach to the growing cohort of patients with Fontan circulation by developing efficient ways to identify psychosocial maladjustment, collaborating with mental health professionals, and developing pediatric-based preventive approaches to encourage positive psychosocial adaptation.
Advanced Care Planning
Although the Fontan procedure has certainly extended life spans and mortality has largely shifted from the pediatric to adult setting, a mortality risk remains that is associated with the original Fontan procedure and a Fontan conversion. Research has suggested that many adolescents and adults with CHD, including those with complex disease, overestimate their life expectancy.352 Qualitative research revealed that adolescents and adults with Fontan circulation “made a pledge to themselves that they would stay alive as long as possible”350 yet also expressed uncertainty about the future and were aware of the possibility of shortened life expectancy. Thus, it is critical that patients, families, and healthcare providers are well prepared for the premature mortality facing individuals with Fontan circulation.
Advance directives provide the valuable opportunity for patients to decide whom they want to convey their preferences for medical and palliative care should they become unable to speak for themselves. These directives benefit not only patients nearing the end of their lives but also family members and health providers facing this difficult phase. Guidelines recommend completion of advance directives and addressing end-of-life matters as part of the routine care of adolescents and adults with CHD.353,354 Patients should be encouraged to complete advance directives “ideally at a time during which they are not morbidly ill or hospitalized, so that they can express their wishes in a less stressful setting.”353 Although many deaths among patients with Fontan physiology occur after a period of decline, some deaths occur unexpectedly. Thus, waiting for the end stage of disease to initiate advanced care planning is discouraged.
North American studies that surveyed adults with CHD in the outpatient setting revealed that <15% recalled discussing advanced care planning with their provider, only 5% to 21% had completed advance directives, and the majority preferred advanced care planning discussions to be initiated when healthy.355–358 On a scale from 0 to 10, with 10 being most important, the average rating for discussing advanced care planning with providers was 7, and participants preferred a median age of 18 years (range, 5–60 years) as the most appropriate age to initiate the discussions.358 Variables for timing of discussions include the patients’ expressed wishes and cognitive functioning, familiarity with the physician, and timing in relation to transitioning care to a new provider. Thus, these conversations should not be postponed until end-stage disease progression. Room for improvement in clinical care was similarly demonstrated in a study of adults with CHD, most with advanced disease who died during a hospital admission.359 Very few had documented end-of-life discussions. Attempted resuscitation was less likely among patients with documented end-of-life discussions and did not occur at all among patients referred to palliative care.
In summary, documentation of advanced care planning discussions and the completion of advance directives occur for a minority of adults with CHD who, as a group, value these discussions and wish for them to occur earlier in the disease course. A common provider-reported barrier to end-of-life discussions is being unable to reliably estimate life expectancy.357 However, challenges in prognostication can be shared directly with patients and families, and providers may be reassured to know that it is general information on the life expectancy of patients with their form of CHD that most patients are seeking.356,358
Recommendations for patients with Fontan circulation include normalizing conversations about advanced care planning and end-of-life care, scheduling separate visits to discuss these important matters with patients (and their chosen loved ones) with later-stage disease approaching the end of their lives, using patient-friendly language to discuss health expectations and prognoses, and exploring patient preferences for end-of-life care while recognizing that they might shift over time.360 Soliciting guidance from mental health professionals and providers experienced in how best to approach these conversations is important. When working with adolescents and young adults, it is important to be mindful of neurodevelopmental abilities, capacity for decision-making, and cultural background.354,360–362 Providers are encouraged to partner with palliative care providers to manage symptoms and patient and family distress.360 It is also important for healthcare teams to recognize their own emotional reactions that often accompany the death of a younger person in whose care they may have been involved for years or even decades.360 Key take-away points for psychological, mental health, and social challenges are listed in Table 12.
Mood and anxiety disorders are prevalent and contribute to impaired quality of life. |
Discussions about lifestyle, prognosis, and advanced care planning should be incorporated into healthcare visits in an age- and development-appropriate manner. |
Unique Aspects and Challenges in the Adult
Despite an extraordinary physiology that would not seem compatible with normal adult experiences, many adults with a Fontan palliation lead normal adult lives. However, the natural history of this physiology places the burden of many of the complications described in this statement on adults. The time-dependent nature of many of the cardiac, hepatic, renal, and other sequelae is such that they develop in the years and decades after completion of the Fontan, thus manifesting in adulthood. In addition, the superimposition of noncardiac medical issues occurs more frequently with age, and management of even simple medical problems may be complicated by the hemodynamic deficiencies of the Fontan circulation.
Many of the cardiac complications after the Fontan operation become more common with time and consequently are more likely to affect adult patients. Mortality is higher in patients with Fontan circulation than with most other types of CHD, the sole exception being Eisenmenger syndrome. In a study of mortality comparing patients with the normal expected life span in the United States, a 40-year-old patient with Fontan repair has a risk of death equivalent to that of a 75-year-old.363 Adults are also more likely to have the surgical histories associated with higher cardiac complication rates such as atriopulmonary or Bjork-type modifications of the Fontan surgery, later age at Fontan completion, and longer periods of time with volume-loading shunts or cyanosis. Emblematic of the progression of complications seen in adults with Fontan palliation is the experience described by the ANZFR, in which survival of those who were alive at 16 years of age to 30 years of age was 90% but fell to 80% at 40 years of age, with only 53% of patients in New York Heart Association class I heart failure at the time of last follow-up and only 41% without a serious adverse event by 40 years of age.364 A report from the Mayo Clinic describes the 40-year outcomes of >1000 patients who had undergone a Fontan operation and gives 10-, 20-, and 30-year survival rates of 74%, 61%, and 43%.6 The association of poor outcomes was reported as a variety of perioperative and operative features that have improved over time, and not surprisingly, mortality has improved in the more recent decades. However, their experience emphasizes the mortality facing adults with Fontan circulation. In another report from the Children’s Hospital of Philadelphia, two-thirds of patients who underwent Fontan surgery between 1992 and 2009 required catheter or surgical interventions in the ensuing 20 years, with many occurring in young adults.365 Hospitalizations are more frequent in adults with Fontan circulation than in adults with other CHDs.366
Fontan Conversion and Revision Surgery
Adults are the group of patients who have old-style atriopulmonary Fontan operations and are thus the population for whom surgical conversion remains a treatment option. The primary indications for Fontan conversion include difficult atrial arrhythmias, atrial clots, and hemodynamic derangements (eg, AVV regurgitation) that require surgical intervention. The procedure involves takedown of the atriopulmonary connection, reduction of the right atrium, creation of an extracardiac TCPC, and modifications of atrial arrhythmia surgery. A report of 140 patients who underwent Fontan conversion describes a freedom of atrial arrhythmias of 77% and survival free of death or transplantation at 5, 10, and 15 years of 90%, 84%, and 66%, respectively.125 Fontan conversion with arrhythmia surgery is successful in reducing arrhythmias and improving symptoms, but as more children are treated with lateral tunnel or TCPC repairs, the number of patients for whom a Fontan conversion is indicated is decreasing over time.6 With the more recent advent of extracardiac conduit–type Fontan operations, some have speculated that a small-diameter conduit (<16 mm), although satisfactory for a child, might increase inferior vena caval pressure and potentially exacerbate FALD when patients grow to adulthood.367,368 There are currently few data to refute or support this hypothesis; more elapsed time is needed before we can determine whether a relatively small extracardiac conduit may ultimately require future surgical revision or catheter-based intervention.369
Concomitant Medical Challenges in the Adult Patient
The coexistence of other medical or surgical problems is more typical in adulthood, and management of these problems may be complicated by underlying Fontan physiology. In 1 series, adult patients with Fontan palliations had more periprocedural complications (15%) than patients with CHD and a biventricular repair or patients without CHD.370 In an administrative study of costs of admissions for noncardiac reasons, adult patients with CHD in general had higher costs, longer stays overall, higher rates of intensive care unit stay, and higher mortality.371 There is a paucity of robust data on the impact of Fontan physiology on the outcomes of other medical problems. For example, the risk of cancer is increased in the adult with CHD and is a considerable challenge to manage in the patient with Fontan circulation.236 More data on the impact of Fontan physiology on the management of common conditions are needed to provide guidance on optimal management strategies. Care in adult CHD centers has been demonstrated to reduce mortality in patients with moderate to complex CHD.372 Evaluation and care by adult CHD specialists is of benefit in optimizing management strategies for the unique and complex aspects of caring for the adult with Fontan circulation. Guidelines for the general management of adults with CHD were updated for 2018.373
Contraception and Pregnancy
Women with Fontan circulation may desire contraception and may consider the possibility of pregnancy. Counseling on pregnancy can be a challenge because there are few data on the long-term cardiovascular impact of the circulatory changes of pregnancy, particularly volume overload, on Fontan physiology. Nevertheless, because of the strong natural desire to have a family, there is growing experience with women with Fontan circulation experiencing pregnancy. Maternal complications of pregnancy include arrhythmias, heart failure, and thromboembolic complications, which can occur in up to 25% of women. Strikingly high rates of fetal loss are demonstrated in the few series of women with Fontan circulation who achieved pregnancy. In a study of 50 women with 124 pregnancies, the miscarriage rate was nearly 55%.374 Women who are cyanotic are more likely to have miscarriage.375 In a meta-analysis of pregnancies reported in women with Fontan circulation, arrhythmias and heart failure were more common than in the normal populations or in those with simpler forms of CHD; fetal complications were dramatically higher, with up to 50% of pregnancies ending in miscarriage.376
Counseling women with Fontan circulation about pregnancy should take into account the cardiovascular status, hepatic status, thromboembolic risk, potential heritability of the CHD, and potential for a long-term decrease in cardiovascular function as a result of pregnancy. An important, but difficult, conversation concerns the life expectations for women with Fontan physiology, in terms of both morbidity and mortality. In a study of outcomes of preconception counseling of women with a Fontan palliation, most women agreed to participate; however, the majority chose to pursue pregnancy even after being given information about the higher rates of maternal and fetal complications.377
Women with Fontan circulation have higher rates of primary amenorrhea; thus, menarche may be delayed.378 For some women, assisted reproduction may be considered. The hormonal manipulations needed for various forms of assisted reproduction may be poorly tolerated by a woman with Fontan circulation because of the potential for thrombosis and blood volume shifts. Pregnancy surrogacy may be a desirable alternative for some women. Ethical considerations related to maternal longevity concerns are also present when the use of advanced technology-based assisted reproductive technologies is considered. Assisted reproduction also carries a risk of multiple embryos, which can produce an even greater hemodynamic burden on the pregnant woman with Fontan circulation.
Surveillance Strategies: Proposed Schema of How to Monitor the Patient With Fontan Circulation
Given the relatively high prevalence and possible progressive nature of end-organ dysfunction in those with Fontan circulation, in particular during the early formative years of organ growth and development during childhood, adolescence, and early adult life, a compelling argument can be made for performing serial surveillance testing.379
On the basis of expert consensus, the authors of this AHA statement believe the following:
•
For patients with Fontan circulation who appear clinically well, it is reasonable to offer surveillance testing that systematically evaluates cardiovascular and end-organ health periodically.
•
Individual patients, in particular those who are unwell, will have specific needs that determine the most appropriate testing required, and such testing should be individualized at the liberty of their healthcare provider.
•
At this time, there is an insufficient evidence base, as strictly defined by the AHA guidelines for developing and grading recommendations, to support recommendations for specific tests or for the frequency/interval of testing. Nevertheless, we believe that surveillance testing for cardiovascular and end-organ system consequences of the Fontan circulation is reasonable, clinically important, and to be encouraged as part of overall high-quality patient care.
•
Surveillance may be provided by individual practitioners or by specialty multidisciplinary Fontan/single-ventricle care clinics. Surveillance should be offered by healthcare providers and centers familiar with and experienced in the unique aspects of Fontan circulation care and comfortable with interpreting the results.
•
The optimal surveillance testing elements and frequency of testing are age related and change as the patient moves through childhood and adolescence and into adulthood, as organ system complications emerge, and as the rate of progression of dysfunction changes.
•
It is anticipated that practice patterns will, and should, change over the coming years as further collaborative study ensues and higher-quality evidence emerges to guide selection of the most appropriate surveillance tests to undertake.
Through development of consensus opinion by this AHA writing group, we propose a Fontan Circulation Surveillance Testing Toolkit (Tables 13–16). Cardiovascular system surveillance testing is already common practice. Cardiovascular assessment at set intervals in all age groups throughout life is a reasonable approach. Obtaining a good interval history and performing a careful physical examination cannot be replaced by any test. Findings such as progressive cyanosis, new murmur, elevated jugular venous distension, ascites, leg edema, or unexpected changes in weight during serial surveillance should be noted. Examples of specific cardiovascular tests and suggested surveillance intervals are provided in Table 13. Surveillance of end-organ status in the patient with Fontan circulation is a relatively new concept. Tests within this toolkit are proposed as a means of characterizing organ systems in the individual with a single ventricle and Fontan circulation.
Test | Child | Adolescent | Adult |
---|---|---|---|
Outpatient visit, including physical examination | Every 6–12 mo | Every 6–12 mo | Every 6–12 mo |
ECG | Every 6–12 mo | Every 6–12 mo | Every 6–12 mo |
Echocardiogram | Yearly | Yearly | Yearly |
Holter 24-hour monitor | Every 2–3 y | Every 1–2 y | Every 1–2 y |
Exercise stress test | Every 2–3 y* | Every 1–3 y | Every 1–2 y |
Serum BNP or NT-proBNP | Once in childhood | Every 1–3 y | Every 1–2 y |
Cardiac MRI | Once every 3 y | Every 2–3 y | Every 2–3 y |
CT angiography | As clinically indicated | As clinically indicated | As clinically indicated |
Cardiac catheterization | As clinically indicated | Once every 10 y | Once every 10 y |
BNP indicates brain natriuretic peptide; CT, computed tomography; MRI, magnetic resonance imaging; and NT-proBNP, N-terminal pro-B-type natriuretic peptide.
*
In young children unable to participate in a complete exercise stress test (bike or treadmill) with metabolic assessment, testing may include a 6-minute walk to assess for distance, duration, and oxygen saturation measures of exercise capacity.
Organ System | Basic | In-Depth | Investigational |
---|---|---|---|
Liver | CMP | PT/INR | Liver imaging via CT or MRI |
Platelet count | Serum FibroSure biomarkers | Liver elastography (ultrasound or MRI) | |
Serum GGT | Serum α-fetoprotein | Liver biopsy | |
Abdominal (liver) ultrasound | |||
Total serum cholesterol | |||
Kidney | Serum BUN, creatinine | Urinalysis albumin/creatinine ratio | Renal ultrasound with Doppler |
Serum cystatin C | Nuclear scan GFR | ||
Lymph | Serum albumin, total protein | Serum IgG | T2-weighted MRI lymphatic imaging |
Absolute lymphocyte count | Fecal α-1 antitrypsin level | ||
Endocrine/metabolic | Serum calcium | Parathyroid hormone | Serum insulin-like growth factor |
Vitamin D | Bone densitometry/DXA scan | ||
Tanner staging, developmental assessment | Nutritional evaluation and consultation | ||
Heme | CBC, Hgb, Hct | Serum iron | Coagulation factors |
TIBC | |||
Ferritin | |||
Lungs | … | Chest x-ray | Pulmonary function testing |
Neurological/psychological | Neurodevelopmental /cognitive testing† | Psychological evaluation and consultation | Brain MRI scanning |
BUN indicates blood urea nitrogen; CBC, complete blood cell count; CMP, comprehensive metabolic panel; CT, computed tomography; DXA, dual-energy x-ray absorptiometry; GFR, glomerular filtration rate; GGT, γ-glutamyl transferase; Hct, hematocrit; Hgb, hemoglobin; IgG, immunoglobulin G; INR, international normalized ratio; MRI, magnetic resonance imaging; PT, prothrombin time; and TIBC, total iron-binding capacity.
*
Our group expert consensus found it reasonable to consider surveillance testing every 3 to 4 years in the child (<12 years of age) with Fontan circulation.
†
Neurodevelopmental testing is in accordance with recommendations made by Marino et al.294
Organ System | Basic | In-Depth | Investigational |
---|---|---|---|
Liver | CMP | Serum FibroSure biomarkers | Liver biopsy |
Platelet count | Serum α-fetoprotein | ||
Serum GGT | Abdominal (liver) ultrasound | ||
PT/INR | Total serum cholesterol | ||
Liver imaging via CT or MRI | |||
Liver elastography (ultrasound or MRI) | |||
Kidney | Serum BUN, creatinine | Urinalysis albumin/creatinine ratio | Nuclear scan GFR |
Serum cystatin C | Renal ultrasound with Doppler | ||
Lymph | Serum albumin, total protein | Serum IgG | T2-weighted MRI lymphatic imaging |
Absolute lymphocyte count | Fecal α-1 antitrypsin level | ||
Endocrine/metabolic | Serum calcium | Parathyroid hormone | Serum insulin-like growth factor |
Vitamin D | Bone densitometry/DXA scan | ||
Tanner staging, developmental assessment | |||
Nutritional evaluation and consultation | |||
Heme | CBC, Hgb, Hct | Serum iron | Coagulation factors |
TIBC | |||
Ferritin | |||
Lungs | … | Chest x-ray | |
Pulmonary function testing | |||
Neurological/psychological | Neurodevelopmental/cognitive testing† | Psychological evaluation and consultation | Brain MRI scanning |
BUN indicates blood urea nitrogen; CBC, complete blood cell count; CMP, comprehensive metabolic panel; CT, computed tomography; DXA, dual-energy x-ray absorptiometry; GFR, glomerular filtration rate; GGT, γ-glutamyl transferase; Hct, hematocrit; Hgb, hemoglobin; IgG, immunoglobulin G; INR, international normalized ratio; MRI, magnetic resonance imaging; PT, prothrombin time; and TIBC, total iron-binding capacity.
*
Our group expert consensus found it reasonable to consider surveillance testing every 1 to 3 years in the adolescent (12–18 years of age) with Fontan circulation.
†
Neurodevelopmental testing is in accordance with recommendations made by Marino et al.294
Organ System | Basic | In-Depth | Investigational |
---|---|---|---|
Liver | CMP | Serum FibroSure biomarkers | Liver biopsy |
Platelet count | Serum α-fetoprotein | ||
Serum GGT | Liver imaging via CT or MRI | ||
PT/INR | Liver elastography (ultrasound or MRI) | ||
Total serum cholesterol | |||
Abdominal (liver) ultrasound | |||
Kidney | Serum BUN, creatinine | Urinalysis albumin/creatinine ratio | Nuclear scan GFR |
Serum cystatin C | Renal ultrasound with Doppler | ||
Lymph | Serum albumin, total protein | Serum IgG | T2-weighted MRI lymphatic imaging |
Absolute lymphocyte count | Fecal α-1 antitrypsin level | Lymphatic angiography | |
Endocrine/metabolic | Serum calcium | Parathyroid hormone | Serum insulin-like growth factor |
Vitamin D | Bone densitometry/DXA scan | ||
Nutritional evaluation and consultation | |||
Heme | CBC, Hgb, Hct | Serum iron | Coagulation factors |
TIBC | |||
Ferritin | |||
Lungs | Pulmonary function testing | Chest x-ray | |
Neurological/psychological | Psychological evaluation and consultation | Neurodevelopmental/cognitive testing† | Brain MRI scanning |
BUN indicates blood urea nitrogen; CBC, complete blood cell count; CMP, comprehensive metabolic panel; CT, computed tomography; DXA, dual-energy x-ray absorptiometry; GFR, glomerular filtration rate; GGT, γ-glutamyl transferase; Hct, hematocrit; Hgb, hemoglobin; IgG, immunoglobulin G; INR, international normalized ratio; MRI, magnetic resonance imaging; PT, prothrombin time; and TIBC, total iron-binding capacity.
*
Our group expert consensus found it reasonable to consider surveillance testing every 1 to 2 years in the adult (>18 years of age) with Fontan circulation.
†
Neurodevelopmental testing is in accordance with recommendations made by Marino et al.294
A variety of potential tests exist for characterizing the end-organ systems. As an evidence base for the most disease-specific and valuable tests emerges, and in order to assist healthcare practitioners who wish to test their patients at various levels of depth of surveillance, we stratify the various organ system tests as basic (fundamental and rudimentary level of assessment), in-depth (more detailed level of characterization), and investigational (possible or likely of value; however, greater experience and study may be necessary before widespread use can be suggested).
End-organ system surveillance testing frequency should be tailored to patient age. It is reasonable to consider surveillance testing every 3 to 4 years in childhood (<12 years of age; Table 14), every 1 to 3 years in adolescence (12–18 years of age; Table 15), and every 1 to 2 years in adulthood (>18 years of age; Table 16). Findings of surveillance testing may influence decisions concerning the next interval until testing is repeated.
Exercise: A Means for Assessment and as Therapy
Subnormal exercise capacity is extremely common in patients with Fontan circulation, with a wide variety of potential contributing factors. Cardiac limitations can include impaired chronotropic responses, inability to raise right-sided heart output on exertion because of the lack of a subpulmonary ventricle, and possible limitations of pulmonary vascular reserve. Resting and exertional hypoxemia may also be relevant (from a systemic to pulmonary venous chamber fenestration or venovenous collaterals). Extracardiac sources of limitation can include restrictive lung issues (eg, after thoracotomy), reduced skeletal muscle mass (sarcopenia), and physical deconditioning. In particular, sarcopenia is a common and underappreciated finding in patients,272 relevant to exercise intolerance.
Exercise as a Test
Cardiopulmonary exercise testing (CPET) can be particularly useful in patients with Fontan circulation to quantify (serially) maximum exercise capacity and to infer the reason(s) for any exercise limitation observed. On CPET, key parameters of interest include max to assess aerobic capacity and the ratio of to to assess respiratory efficiency and anaerobic threshold. Impaired CPET parameters are associated with increased risk of hospitalization in patients with Fontan circulation,62 and both maximal and submaximal exercise data appear to have prognostic value in this population.380 Deterioration in CPET parameters on serial study identifies a population at increased risk for later cardiovascular events.133 Other specific parameters can provide insights into respiratory reserve, stroke volume, and ventilation-perfusion matching. Detailed knowledge of contributors to exercise limitation in patients with Fontan circulation can suggest potential therapeutic strategies for consideration such as fenestration closure for excessive hypoxemia or atrial pacing for important chronotropic incompetence.
Invasive CPET testing has also been applied in patients with Fontan circulation. Although this involves the insertion of pulmonary and radial artery catheters, which can alter resting physiology, such studies have clearly shown (inter alia) rapid and dramatic increases in systemic venous pressures in exercising patients with Fontan circulation, often exceeding 30 mm Hg at relatively modest workloads.214 This phenomenon is associated with poor systemic oxygen delivery.
Because maximal exercise capacity is so often limited in those with Fontan circulation, ranges of normal CPET parameters for children and teenagers are now published (at age 12±3 years).381
Exercise Training and Rehabilitation
Exercise training may have particular benefits in patients with Fontan circulation for physical and psychological well-being. Given the importance of peripheral venous return, increased muscle mass may augment venous return and thus influence heart preload. Investigators have shown that leg muscle resistance training can significantly augment peripheral muscle mass, venous return, cardiac output, and max in adults with Fontan circulation.382 Similarly, given the importance of the thoracic bellows to drive inspiratory venous return to the heart, investigators have found that inspiratory muscle training (for 6 weeks) improves resting cardiac output and ventilatory efficiency during exercise.383 In contrast, results from aerobic training programs have been less encouraging.384 Further studies will be informative concerning the optimal type or combination, frequency, and duration of exercise training for patients with Fontan circulation.
Exercise training certainly appears safe for patients with Fontan circulation. A review of 23 articles on exercise training found only 3 adverse events in >200 participants, all of which were transient.385 When measured, quality of life improved with exercise training in the majority of studies. The main physical benefits documented were improved muscle strength and stroke volume. Key take-away points for exercise are listed in Table 17.
Reduced exercise tolerance is common and related to lack of subpulmonary pump, chronotropic incompetence, cyanosis, lung restriction, reduced skeletal muscle mass, and deconditioning. |
Serial cardiopulmonary exercise testing is useful to quantify exercise capacity and to identify changes in clinical status, allowing therapeutic changes. |
Exercise training, in particular specific strategies such as leg muscle resistance training, may prove to be very beneficial because it might improve cardiac output and quality of life. |
Management Strategies: Pharmacological
The aims of pharmacological therapy are to ameliorate the deleterious impact of Fontan physiology and to prevent and treat specific complications that arise. Given the physiological constraints of the Fontan circulation, mechanistic targets include systolic and diastolic ventricular function, ventricular remodeling, and pulmonary and systemic vascular resistance. Additional targets might address specific complications such as PLE, liver fibrosis, or thrombosis. The interrelationship between these targets is not completely understood. In addition, their relationship with events such as death or morbidities or with patient-level outcomes such as functional health status and quality of life is incompletely known. These issues and the heterogeneity and rarity of patients with Fontan circulation have made the assessment of the efficacy of pharmacological therapies particularly challenging. This has led to a poor evidence base, which has contributed to important clinical practice variation in pharmacological management.386 However, the availability of new agents, particularly pulmonary vasodilators and antithrombotics, has created an opportunity and imperative for increased and more efficient study of the Fontan population.
Given the rarity and extended timeline for events to occur and the nonspecific and poorly responsive measures of patient-level outcomes, the feasibility and validity of using objectively measured surrogate outcomes have been sought. The primary aim of the multi-institutional National Institutes of Health Pediatric Heart Network Fontan Cross-Sectional Study was to determine laboratory measures that would be significantly associated with functional health status for pediatric Fontan survivors.387 The study showed that variation in physical functioning summary scores was explained, albeit poorly, by medical conditions and long-term and current medical problems, not by cardiac and management characteristics.388 Likewise, laboratory measures of ventricular characteristics and function and exercise capacity showed poor associations, variously accounting for only ≈10% of the variation in physical functioning summary scores.389 Nonetheless, exercise capacity and exercise hemodynamics have become the outcome of interest for recent trials.
Pulmonary Vascular Resistance
The applicability of agents that modulate pulmonary vascular resistance has generated a great deal of interest for the Fontan population.390 Agents include phosphodiesterase-5 enzyme inhibitors (sildenafil, tadalfil), endothelin receptor antagonists (bosentan, ambrisentan), and prostacyclins (iloprost). A recent systematic review of pharmacological therapy for patients with Fontan circulation examined 9 studies with a total of 267 patients, 8 of which studied a pulmonary vasodilator: 4 studies with sildenafil, 3 with bosentan, and 1 with iloprost.391 Most of these studies were small and short term, but a trend toward a positive impact on exercise capacity and hemodynamics was noted. The largest study, a randomized placebo-controlled trial of bosentan in 75 patients treated for 14 weeks, showed an increase in peak oxygen consumption, exercise time, and functional class without serious adverse events.392 The authors concluded that although the results were promising, further study was necessary. After completion of a pharmacokinetic dose-finding study of udenafil (a relative long-acting phosphodiesterase-5 inhibitor) in 36 patients with Fontan circulation,393 a larger efficacy and safety placebo-controlled trial of treatment for 26 weeks of 400 patients is currently in progress (Fontan Udenafil Exercise Longitudinal Assessment Trial; URL: ClinicalTrials.gov. Unique identifier: NCT02741115), with a primary outcome of change in peak oxygen consumption. A 52-week open-label extension study will further assess safety outcomes (A Extension Study of Udenafil in Adolescents; URL: ClinicalTrials.gov. Unique identifier: NCT03013751). A placebo-controlled trial of 52 weeks of macitentan (an endothelin receptor antagonist) in 134 patients with Fontan circulation is underway (Clinical Study Assessing the Efficacy and Safety of Macitentan in Fontan-Palliated Subjects; URL: ClinicalTrials.gov. Unique identifier: NCT03153137), also with a primary outcome of change in peak oxygen consumption. These studies will inform the evidence base to support a clinical recommendation, weighing benefit, safety, and cost.
Thrombosis
Thrombosis is an important and unpredictable complication, not only after Fontan operation but also associated with each stage of single-ventricle palliation.394 It is an important cause of morbidity, particularly when it leads to pulmonary embolism or stroke, and contributes to mortality. The predominant predisposing factor for thrombosis is stasis of blood flow. Venous stasis is often evident in the cavopulmonary circuit in light of the absence of a pump for systemic venous return and pulmonary blood flow. Congenital or acquired anatomic areas of stasis may be present such as a hypoplastic left ventricle with poor inflow and outflow or the creation of a blind-ended pulmonary artery stump. A number of reports have described altered levels of clotting and fibrinolysis factors at all stages of palliation but below the clinical threshold for abnormality. However, these altered levels have been associated with features specific to Fontan palliation, including lower systemic oxygenation, low cardiac output, and hepatic congestion. Furthermore, some patients may have inherited or intrinsic alterations in thrombophilia factors. Most alterations in clotting factor levels likely reduce the reserve of the clotting system, whereby an inciting stimulus such as cardiopulmonary bypass or an indwelling central line increases the risk of both bleeding and thrombosis. The presence of further disturbance of the coagulation system such as through excessive factor loss through PLE or synthetic impairment resulting from severe liver fibrosis or cirrhosis increases the risk of thromboembolic complications.
A targeted approach to thromboprophylaxis is not yet feasible, although patients with a history of thrombosis are at increased risk of recurrence. Additional factors such as atriopulmonary Fontan connection type, Kawashima connection type, dilated atrium, presence of thrombogenic foreign material, ventricular dysfunction, arrhythmias, prolonged immobilization, and PLE have been variously reported. Although clinicians remain concerned about the potential for paradoxical emboli in patients with atrial fenestration, this has not been reported as a risk factor for thrombosis or thromboembolic events.
Both antiplatelet and anticoagulation agents have been used. Some thromboprophylaxis has been shown to be better than none, but the superiority of acetylsalicylic acid (ASA) versus warfarin remains unclear, as was noted in a meta-analysis of 20 observational studies.395 An underpowered randomized clinical trial of ASA versus warfarin failed to show a significant difference in the incidence of thrombosis over a 2-year period immediately after the Fontan procedure,396 although a secondary analysis showed that poorly controlled warfarin was associated with a high risk of thrombosis.397 A small trial of ASA versus warfarin showed greater deposition of material, presumed laminar thrombus, within the extracardiac conduit cavopulmonary connection for those on ASA.398 Current guidelines suggest that it is reasonable for all patients with Fontan circulation to receive ASA, with anticoagulation reserved for those with presumed risk factors or previous thrombosis or older patients.316 With either agent, an important residual risk of thrombosis remains; hence, there is an ongoing unmet need for better strategies.399 Newer direct oral anticoagulants are currently being studied in pediatric cardiac populations, including patients with Fontan circulation. They are likely to be an attractive alternative to warfarin, although their comparative efficacy and safety have yet to be determined. Key take-away points for thrombosis are listed in Table 18.
Patients are at increased risk for thromboembolic events |
Although the most effective means to prevent thromboembolic events are still unclear, some form of thromboprophylaxis, either an antiplatelet agent or anticoagulant, is warranted. |
Management Strategies: Mechanical Circulatory Support
The importance of mechanical circulatory support (MCS) as a potential therapy for a single functional ventricle is evident by the steady increase in an aging Fontan population and a shortage of donor organs for transplantation. The application of MCS for Fontan circulatory support remains controversial and poorly defined. At present, there is no MCS device specifically intended for use in this population. Existing devices designed for systemic circulatory support in biventricular circulations have been adapted for use in the single-ventricle Fontan circulation. Although success with existing devices has been reported, the experience is highly variable, generally limited to case reports, and outcomes are still relatively poor.400–408
Unfortunately, MCS device performance is commonly mismatched to the unique physiological needs of the Fontan circulation. Existing technology is better suited to ventricular pump failure in the patient with a single ventricle with Fontan circulation than to Fontan failure secondary to a lack of a subpulmonary ventricle and the circulatory deficiencies that follow. Anecdotal evidence indicates that systemic ventricular assist devices appear to be most effective when applied to a Fontan circulatory system in the setting of impaired ventricular systolic function. At late follow-up, however, most patients with Fontan circulation who have not reached end-stage failure have preserved systolic function.50 In the setting of preserved systolic function, the role of systemic MCS may be superfluous and may further congest the right-sided circulation. A cavopulmonary assist device would require a highly specialized low-pressure right-sided circulatory technology, which does not yet exist. From a theoretical perspective, an MCS technology targeted to support the lack of a subpulmonary ventricle could be of tremendous benefit, especially if applied early to preempt the progression of failure by providing a biventricular circulation. Early-stage research is underway to address this important need.409–411 At present, the use of commercially available MCS therapy is limited to end-stage Fontan circulation failure as a bridge to transplantation. The total artificial heart has been successfully applied to bridge a patient to heart transplantation.412 Use of MCS as destination therapy in patients with Fontan circulation has been contemplated but not yet reported.
Moving forward, it will become more important to establish criteria to categorize Fontan circulatory inefficiency as predominant left- or right-sided to inform which MCS technology can be best matched to the deficit. Left-sided failure (systolic dysfunction, low cardiac output) may be best served with a systemic ventricular assist device; right-sided failure (preserved systolic function, diastolic dysfunction, sequelae of systemic venous hypertension) may be best served with a cavopulmonary assist device. In some instances, both forms of support may be needed. Evaluative parameters should be developed to categorize the cause of Fontan failure so that a specific device can be selected to best match to the physiological deficit.65
Although exceedingly challenging, MCS provides a tremendous opportunity to cure single functional ventricle and to preempt the insidious progression of chronic Fontan circulatory failure. Research and development of these technologies is critical.413
Management Strategies: Heart Transplantation
Heart transplantation can be successfully performed among patients who have received the Fontan operation, but many controversies remain, particularly concerning questions of transplantation indications, contraindications, and the optimal timing of transplantation. Although a uniform set of criteria that determine transplantation eligibility for the patient with Fontan circulation would be desirable, current knowledge of the actual survival rates associated with most Fontan complications is too rudimentary to allow such consistency. The patient with Fontan circulation without major complications has a significantly better prognosis than heart transplantation would afford. With the onset of major Fontan-associated morbidities, the prognosis becomes more guarded. Renal dysfunction and hepatic dysfunction, which are common in long-term survivors, further modulate outcomes, but the impact of these factors is not fully understood. Thus, listing decisions are made in a center-specific manner, evaluating Fontan circulation–specific morbidities, as well as the status of ventricular systolic and diastolic function and AVV insufficiency, to estimate prognosis and to compare this with the expected prognosis of transplantation.
Heart transplantation is best performed at a center with substantial experience in transplantation in individuals with CHD, including patients with Fontan circulation. Of patients who are listed for transplantation, the most common diagnoses include PLE (40%), ventricular dysfunction (50%), and other diagnoses such as PB and arrhythmias (10%).414,415 These categories are not mutually exclusive, and these proportions are rough estimates. It is not unusual for patients with PLE to be listed for transplantation, but it is probably more common for transplantation to be reserved for those patients who no longer respond to medical or interventional options. Patients with Fontan circulation and ventricular systolic dysfunction are generally listed on the basis of the severity of heart failure symptoms, the ejection fraction, and the clinical trajectory. For other forms of morbidity or failure, the decisions about listing remain highly individualized.
The process by which cardiac donors are assigned to potential candidates is determined on a national level in the United States, with priority levels defined by the United Network for Organ Sharing, incorporating input from both clinicians and patients. For children, CHD is afforded prioritization, with eligibility for the highest level of priority (status 1A) available to patients with CHD who require a single inotropic agent as continuous infusion. This priority is available only for patients with dilated cardiomyopathy who have received MCS or require mechanical ventilation. This prioritization reflects the large number of patients with CHD who receive transplantation as children, recently estimated as 55% of the total infant transplantation population and 40% of those <1 year of age.416 It should be noted, however, that even this approach largely benefits only the subset of patients with Fontan circulation with reduced ventricular systolic function, who make up approximately half of those who require transplantation. It is of much less value for those with PLE or other forms of Fontan failure with preserved ventricular function.
For adults, patients with CHD make up a very small fraction of the total transplantation population, and the prioritization scheme reflects a lack of focus on this population. The priority criteria for adults are currently in evolution, with new criteria approved but not yet implemented.417 However, in both the old and new schemes, CHD is not associated with any elevation in priority status, and the factors that lead to priority (eg, placement of MCS, continuous infusion of inotropes in combination with pulmonary artery catheter) are not commonly used in Fontan circulatory failure, leading to ongoing challenges in obtaining donors for these patients even after they are listed for transplantation.
For those patients who receive heart transplants, outcomes in children are known to be excellent and have been shown to be improving in the current era compared with earlier results. The most robust data for this comes from the PHTS (Pediatric Heart Transplant Study), a multicenter database. Initial reports from the PHTS showed 1-year survival after transplantation of 76% for patients with Fontan circulation, significantly lower than the survival of 91% for control subjects without CHD (P=0.0004).418 More recently, outcomes in the PHTS for patients with Fontan circulation have improved, with a 1-year survival rate of 89%, which is not distinguishable from the control population.415 For adults, the picture is murkier. There are far fewer adult patients with Fontan circulation who undergo transplantation, and the results are not as well documented. The largest single-center experience includes 26 patients over 26 years, with 1-year survival of 65%.419 Inasmuch as a large portion of the mortality after transplantation in the patient with Fontan circulation is attributable to extracardiac morbidities and these morbidities progress over time, there is concern about whether the outcomes in adults will reach the levels demonstrated in children. With the growing number of patients with Fontan circulation reaching adulthood, this question will take on increasing importance in the next epoch of Fontan care. Given the level of specialization required to provide transplantation evaluation and care and the relatively small size of the population, patients with Fontan circulation in need of these services should be offered referral to specialized centers.
Gaps in Knowledge, Goals, and Next Steps
Surgical innovation has led to a pathway of survival for those born with a single ventricle. However, this success has created a population of individuals with increasing morbidity and medical needs. Healthcare resource use and expenditures for patients with a single ventricle are increasing and expected to continue to increase,107 with a paucity of evidence-based clinical care strategies currently in place to affect outcomes.
To meet the challenge of providing a normal quality and duration of life for patients with Fontan circulation, a number of gaps in knowledge need to be bridged and a series of objectives achieved.
Improved Understanding of the Basic Science and Biology of the Single Ventricle
To develop novel strategies, a better foundation of scientific understanding is necessary. A scientific summit was held at Stanford University in April 2018 in which a number of research objectives were summarized. Insights into the genetics of single-ventricle CHD are required. Models of single-ventricle CHD are necessary to elucidate the mechanisms of arrested ventricular development. Processes such as decreased cell proliferation, increased apoptosis, and mitotic cell cycle arrest and associated metabolic and mitochondrial changes in both the fully formed ventricle and the hypoplastic ventricle should be investigated. Exploring the common developmental pathways of the placental and fetal cardiovascular systems may offer clues to the origins of single-ventricle CHD. Investigation of the relationship between genotype and phenotype to elucidate mechanisms of heart failure and to guide counseling for individual families could be accelerated by the development of large clinical registries, modeling single ventricles with induced pluripotent stem cells, high-throughput screening for mechanisms of single-ventricle failure, and identification of biomarkers to serve as a plasma signature of myocardial events. Strategies using stem cell-based therapies to prevent or treat ventricular failure are being explored. Further work in computer modeling, 3D printing, and tissue engineering would help to drive the design and creation of vessels, heart valves, organs, and auxiliary pumps. A large-animal model of Fontan circulation would be of tremendous benefit in further experimental design of the circulation and new surgical and mechanical support therapies.
Improved Understanding of Fontan Physiology
To achieve progress in the care of this growing population, we need to deepen our understanding of the basic physiological mechanisms underlying this extraordinary circulation. Understanding the physiological “critical bottleneck” in the circulation (ventricular dysfunction, valvular dysfunction, Fontan cavopulmonary connection, pulmonary vasculature, etc) will allow a targeted therapeutic strategy, which may vary from patient to patient and within individual patients over time. We now understand how dilatation of the atrial chamber in the classic atriopulmonary Fontan is associated with atrial arrhythmias, thromboembolic events, and ultimately ventricular failure.420,421 We now also understand that patients with Fontan circulation live with decreased cardiac output and increased systemic vascular resistance, but it is still uncertain whether lowering systemic vascular resistance will be beneficial or deleterious to these patients.422 The decline of the Fontan circulation is often associated with decreased oxygen saturation, increased cardiac volumes, and altered mesenteric blood flow, but how these factors interact is unclear.44,423–425 We understand that patients who have adverse characteristics such as decreased ventricular function, distorted or small pulmonary arteries, AVV regurgitation, and arrhythmias will have a worse outcome. However, we do not yet know whether patients who do not have any such adverse features will be affected by rapid deterioration of their Fontan circulation and, if so, in what time frame. It is suspected that all patients are at risk to be impaired because of the gradual increase of their pulmonary vascular resistance and their decreased diastolic ventricular function, but the evidence for when and how these impairments occur is weak.58,426,427
Characterizing the State of the Fontan Circulation
The main parameters helping us to identify the quality of a cavopulmonary circulation are invasive measurements of central pulmonary artery pressures and end-diastolic pressure of the systemic ventricle. Unless grossly abnormal, these parameters have been poorly correlated with adverse outcomes. It is likely that these parameters are unreliable and highly variable even within individual patients because they are measured in only 1 instance when most patients are fasted, in a supine position, and frequently under sedation or general anesthesia. A more real-life, dynamic mode of characterization of the Fontan circulation that better reflects the natural, daily state is necessary.
Understanding the Mechanisms of End-Organ Damage
A large proportion of patients will incur hepatic and renal end-organ damage driven likely in combination by the excessive venous pressure and altered cardiac output characterizing this circulation.8 Differences from normal in baseline substrate organ state as a consequence of altered development may increase vulnerability to the rigors of the Fontan circulation and thus place specific organs at risk. The model of a different-from-normal organ structure at birth, with acquired, additive insult along the way, is likely the process in play in neurological injury and its clinical manifestations. Such a model may similarly apply for other organ systems in the single ventricle. The potential for reversibility of these organ system damages is still unknown. A deeper understanding of the pathophysiology of end-organ damage will allow the development of specific treatments that may mitigate or alter the trajectory of decline and further inform the optimal timing to offer heart transplantation.
Lymphatic Circulation
Emerging knowledge indicates that the lymphatic circulation is under significant duress in the Fontan circulation. Increased venous pressure leads to increased production in organs such as the liver, as well as impediment to drainage. New imaging techniques for the first time allow visualization of the severe degree of lymphatic channel abnormalities present.428 The most feared complications after Fontan operation—PLE, PB and, in terminal patients, ascites and edema—are clearly driven by the incapacity of the lymphatic circulation to drain this increased lymphatic flow.167,197 Lymphocytes are depleted and specific T-cell populations are perturbed in patients with Fontan circulation.176 Defining the pathophysiology behind the trajectory from mild lymphopenia, common and progressive after Fontan operation, to the life-threatening conditions of PLE or PB is important. Exploring the relationship between lymphatic congestion and end-organ dysfunction such as liver fibrosis and intestinal absorptive abnormalities that may be the source of nutritional and vitamin deficiencies would be of great value. Whether there are individual variations in the lymphatic circulation—inherent, acquired, or a combination—is unknown. Research should be focused on studying the capacity for the lymphatic circulation to sustain these abnormal flows. Mechanical or medical interventions for decompression or reducing production may affect the function of the lymphatic circulation and thus profoundly influence outcomes after Fontan operation.429–431
Individual Variation
There is clearly a subset of patients with Fontan circulation who are doing well from both the physiological and the psychosocial perspective. Discovering what factors contribute to resilience and sustained success is critically important. Despite the heterogeneity of this population, practitioners still try to establish management strategies that would fit the entire population. There seems to be little doubt that the variation in innate and acquired characteristics of these patients necessitates a more balanced approach to management and treatment. It is reasonable to believe that the characteristics of the circulation such as cyanosis and increased venous pressures, as well as lymphatic circulatory variation and organ system vulnerabilities, are tolerated in various ways by individual patients, but this possibility is still unexplored. Exploring the genetic basis of ostensible phenotypic variability is essential to understanding this individual variability.
Risk Stratification
There is a continuity spectrum of incremental health status between the perfectly well-appearing, well-functioning patient with Fontan circulation and progressive degradation leading to failure and risk of premature death. However, we are lacking appropriate metrics for characterization specific to the state of patients with this unique circulation. Such metrics would provide a capacity to stratify risk and to better gauge prognosis and the best time to initiate therapies. A validated risk stratification scheme could likely be based on a rich host of features such as morphological and functional characteristics, cardiopulmonary fitness indexes, biological parameters, and estimates of end-organ functionality.
Neurodevelopmental Outcomes, Quality of Life, and Socioeconomic Impact
Neurodevelopmental status and psychological function are some of the most important factors that influence outcome and quality of life in patients with Fontan circulation. Today, the determinants of only 30% of the variations in neurodevelopmental outcomes are elucidated.295,296 Psychosocial well-being is a major concern voiced by families of a patient with Fontan circulation. Mental health concerns affect not just the patient but also parents and siblings, with potential influence on family socioeconomics and quality of life. Mental health therapies for active problems and preventive measures along the therapeutic trajectory starting early in life may improve outcomes but have not yet been clearly identified. Social integration of those born with a single ventricle is a goal, with education, gainful employment, and creation of a meaningful, productive life. Strategies to achieve these goals must emerge as the population grows further into adulthood. Although major physiological changes may not yet be readily available, we should exert greater efforts at the earliest possible points of care to promote wellness through strategies of optimizing exercise, nutrition, and mental health—factors that may very well prove to be readily modifiable and could have great impact on improving quality of life.
Achieving these goals and objectives will require collaboration and the development of multicenter networks. Registries can provide a data resource that may facilitate answering many of the above questions and provide further insight into the design of clinical trials evaluating drugs, exercise regimens, or other interventions. Adherence to the proposed surveillance toolkit suggestions may lead to a reduction in clinical practice variability with learning and discovery of optimal clinical practice strategies. Finally, building a culture of research investigation among patients is important. Healthcare providers should convey to those with single-ventricle heart disease that the best path forward to wellness and a long and healthful life is evolving, and thus, their participation in discovery and investigational research endeavors is essential and should be encouraged. This approach can allow the best evolution of optimal treatment strategies moving forward.
Take-Away Message for the Patient, Parents, and Families
This statement characterizes the health profile of the patient with Fontan circulation. The detailed listing of many issues and challenges within the cardiovascular system and other organs may, by the wealth of information provided, overwhelm patients and their families. The authors of this AHA statement express concern that through the exhaustive articulation of these many challenges, we risk the possibility of painting a very depressing picture of the life of those born with a single ventricle. We therefore believe it is necessary to take a step back and appropriately frame the current situation for the patient with Fontan circulation.
An Extraordinary Story
The survival of patients undergoing the Fontan procedure has been an extraordinary success. At the outset, it was never suspected that patients with such life-threatening birth defects could live for such extended periods in such relatively good condition. A substantial number of patients are now entering their fifth decade and beyond.61,364 We had not predicted the burden that this circulation would have on the physical and psychological health of our patients because at first the focus was on survival. The fear of death remains the highest concern of patients and their families.432 Nevertheless, the extraordinary survival of most born with this condition today is a testimony to the perseverance of numerous practitioners, patients, and their families and should be considered an amazing achievement of modern cardiovascular medicine.
Characterizing the Condition Is an Essential Initial Step
Today, we are in the early phase of characterizing this circulation and its consequences. One should realize that we are reporting population averages with relatively small numbers in most of our investigational studies and that individual patient measures will be spread along a wide variation spectrum. As an example, it was previously suspected that the Fontan circulation was incompatible with a normal exercise capacity. It is now reported that a substantial proportion of patients have better than normal, normal, or near-normal exercise capacity.433 The same is likely possible to hold true for liver, kidney, and brain function. Characterization of the condition in every patient is therefore a necessary step to discover care pathways that can allow the individualization of management and improve personal well-being. This is the fundamental principle underlying our consensus on suggested schema for surveillance.
We Have Not Yet Started to Evaluate the Impact of Interventions
Interventional and preventive strategies of care are evolving for the patient with Fontan circulation. For example, we know that better psychological support of families from the early stages of life may improve psychological well-being of patients, their parents, and their siblings.434 Of great promise but not yet fully tested is the impact of cardiac rehabilitation programs, enhanced nutrition, and interventions aimed at improving neurodevelopmental outcomes. Programs targeting patient education and health self-awareness may be of great value. Trials testing various medications are underway with anticipation of identifying optimal drug therapies to enhance and sustain cardiovascular and end-organ wellness.392,435 Instead of a reactive approach to complications, an era of proactive health maintenance is emerging as experiences unfold and understandings improve.
A Rich and Fulfilling Life
As healthcare practitioners, we may often lose perspective and not appreciate that the life of our patients is not necessarily limited to their medical condition. Between hospitalizations, complications, and fears, our patients enjoy a rich and satisfying life. Although some studies show patients to have a decreased quality of life, others identify patients with Fontan circulation to have a quality of life similar to that of their peers.436–438 Even among some of the oldest patients, many attain higher levels of education, have jobs, and build their own families with satisfying and fulfilling lives.61
We are now entering a new phase in the management of patients born with single-ventricle CHD. Provided that patients undergo regular follow-up, adopt a healthy lifestyle, and are encouraged to participate in investigational clinical protocols and research, healthcare providers and patients can share an optimistic view for a brighter future.
Writing Group Member | Employment | Research Grant | Other Research Support | Speakers’ Bureau/ Honoraria | Expert Witness | Ownership Interest | Consultant/Advisory Board | Other |
---|---|---|---|---|---|---|---|---|
Jack Rychik | The Children’s Hospital of Philadelphia | None | None | None | None | None | None | None |
Yves d’Udekem | Royal Children’s Hospital, Melbourne (Australia) | Actelion (PI on a study; research support for the Australia and New Zealand Fontan Registry)* | None | None | Western Australia Health Authorities* | None | MSD*; Actelion* | None |
Andrew M. Atz | Medical University of South Carolina, MUSC Children’s Hospital | Pediatric Heart Network (principal investigator)*; Idea States Pediatric Clinical Trials Network (principal investigator)* | None | None | None | None | None | None |
David S. Celermajer | University of Sydney and Royal Prince Alfred Hospital (Australia) | Actelion (research support for a clinical trial in Fontan patients)* | None | None | None | None | None | None |
Barbara J. Deal | Northwestern Medical School, Feinberg School of Medicine | None | None | None | Has expert testimony to disclose* | None | None | None |
Michael A. Gatzoulis | Royal Brompton Hospital (United Kingdom) | None | None | None | None | None | None | None |
Marc H. Gewillig | University Hospitals Leuven (Belgium) | None | None | None | None | None | None | None |
Tain-Yen Hsia | Yale New Haven Children’s Hospital, Yale University | None | None | None | Irwin Mitchell LLP*; DAC Beachcroft LLP* | None | None | None |
Daphne T. Hsu | Albert Einstein College of Medicine/Children’s Hospital at Montefiore | None | None | None | None | None | None | None |
Adrienne H. Kovacs | Oregon Health & Science University Knight Cardiovascular Institute | None | None | None | None | None | None | None |
Bradley S. Marino | Ann and Robert H. Lurie Children’s Hospital of Chicago | AHA (Strategically Focused Research Network for Children)† | None | None | Medical expert* | None | None | None |
Brian W. McCrindle | The Hospital for Sick Children (Canada) | NIH (PI in the Pediatric Heart Network)†; Janssen (industry-funded clinical trial)*; Mezzion (participation in industry-sponsored clinical trial)† | None | Alexion* | None | None | Janssen* | The Hospital for Sick Children (staff pediatric cardiologist)† |
Jane W. Newburger | Boston Children’s Hospital, Harvard Medical School | NHLBI (site PI in Pediatric Heart Network, which has some studies on Fontan patients)* | None | None | None | None | None | None |
Nancy A. Pike | University of California Los Angeles School of Nursing | NIH/NINR (R01NR01643)† | None | None | None | None | None | None |
Mark Rodefeld | Indiana University School of Medicine | None | None | None | None | None | None | None |
David N. Rosenthal | Stanford University | None | Abbott*, Berlin Heart* (both funding support of ACTION network, for which he is a co-chair); Berlin Heart*, Medtronic* (both funding support of educational meeting) | None | None | None | None | None |
Kurt R. Schumacher | University of Michigan Congenital Heart Center | NIH-FDA (PI for an FDA-funded orphan r01 grant study of therapy for Fontan-associated plastic bronchitis)† | None | None | None | None | None | None |
Karen Stout | University of Washington | None | None | None | None | None | None | None |
Gruschen Veldtman | Cincinnati Children’s Hospital Medical Center, Children’s Heart Institute | United Therapeutics (lead investigator-initiated trial for supply of drug and placebo for research study)* | None | None | None | None | None | None |
Adel K. Younoszai | Denver Children’s Hospital, Children’s Hospital Colorado | None | None | None | None | None | None | None |
This table represents the relationships of writing group members that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all members of the writing group are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.
*
Modest.
†
Significant.
Reviewer | Employment | Research Grant | Other Research Support | Speakers’ Bureau/Honoraria | Expert Witness | Ownership Interest | Consultant/Advisory Board | Other |
---|---|---|---|---|---|---|---|---|
Thomas M. Bashore | Duke University Medical Center | None | None | None | None | None | None | None |
Frank Cetta | Mayo Clinic | None | None | None | None | None | None | None |
Daniel J. Penny | Baylor College of Medicine | None | None | None | None | None | None | None |
Edward P. Walsh | Children’s Hospital Boston | None | None | None | None | None | None | None |
This table represents the relationships of reviewers that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all reviewers are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.
Acknowledgments
The idea for creating and disseminating a current compendium of knowledge on the Fontan circulation arose from an international “think tank” group that met to deliberate on the condition of these patients and to consider next steps for improving their quality and duration of life. To honor Dr Francis Fontan, many of the authors of this statement met in Bordeaux, France in June 2016. Although Dr Fontan has since passed away, his spirit of innovation and passion for improving the lives of patients with congenital heart disease continue to inspire us and our community to move forward. The authors recognize and acknowledge the important contributions made in Bordeaux by Julie Cleuziou, MD; Xavier Iriart, MD; and Jean-Benoit Thambo, MD; which were essential to laying the foundations for this statement. They also recognize Benjamin Zielonka, MD, for his valuable assistance with organization of references for this work.
References
1.
Fontan F, Baudet E. Surgical repair of tricuspid atresia. Thorax. 1971;26:240–248. doi: 10.1136/thx.26.3.240
2.
Kreutzer G, Galíndez E, Bono H, De Palma C, Laura JP. An operation for the correction of tricuspid atresia. J Thorac Cardiovasc Surg. 1973;66:613–621.
3.
Schilling C, Dalziel K, Nunn R, Du Plessis K, Shi WY, Celermajer D, Winlaw D, Weintraub RG, Grigg LE, Radford DJ, Bullock A, Gentles TL, Wheaton GR, Hornung T, Justo RN, d’Udekem Y. The Fontan epidemic: population projections from the Australia and New Zealand Fontan Registry. Int J Cardiol. 2016;219:14–19. doi: 10.1016/j.ijcard.2016.05.035
4.
Khairy P, Fernandes SM, Mayer JE, Triedman JK, Walsh EP, Lock JE, Landzberg MJ. Long-term survival, modes of death, and predictors of mortality in patients with Fontan surgery. Circulation. 2008;117:85–92. doi: 10.1161/CIRCULATIONAHA.107.738559
5.
d’Udekem Y, Iyengar AJ, Galati JC, Forsdick V, Weintraub RG, Wheaton GR, Bullock A, Justo RN, Grigg LE, Sholler GF, Hope S, Radford DJ, Gentles TL, Celermajer DS, Winlaw DS. 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(suppl 1):S32–S38. doi: 10.1161/CIRCULATIONAHA.113.007764
6.
Pundi KN, Johnson JN, Dearani JA, Pundi KN, Li Z, Hinck CA, Dahl SH, Cannon BC, O’Leary PW, Driscoll DJ, Cetta F. 40-Year follow-up after the Fontan operation: long-term outcomes of 1,052 patients. J Am Coll Cardiol. 2015;66:1700–1710. doi: 10.1016/j.jacc.2015.07.065
7.
Downing TE, Allen KY, Glatz AC, Rogers LS, Ravishankar C, Rychik J, Faerber JA, Fuller S, Montenegro LM, Steven JM, Spray TL, Nicolson SC, Gaynor JW, Goldberg DJ. Long-term survival after the Fontan operation: twenty years of experience at a single center. J Thorac Cardiovasc Surg. 2017;154:243–253.e2. doi: 10.1016/j.jtcvs.2017.01.056
8.
Rychik J. The relentless effects of the Fontan paradox. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2016;19:37–43. doi: 10.1053/j.pcsu.2015.11.006
9.
Fontan F, Mounicot FB, Baudet E, Simonneau J, Gordo J, Gouffrant JM. “Correction” of tricuspid atresia: 2 cases “corrected” using a new surgical technic [in French]. Ann Chir Thorac Cardiovasc. 1971;10:39–47.
10.
de Leval MR. Evolution of the Fontan-Kreutzer procedure. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2010;13:91–95. doi: 10.1053/j.pcsu.2010.01.006
11.
de Leval MR, Deanfield JE. Four decades of Fontan palliation. Nat Rev Cardiol. 2010;7:520–527. doi: 10.1038/nrcardio.2010.99
12.
de Leval MR, Kilner P, Gewillig M, Bull C. Total cavopulmonary connection: a logical alternative to atriopulmonary connection for complex Fontan operations: experimental studies and early clinical experience. J Thorac Cardiovasc Surg. 1988;96:682–695.
13.
Marcelletti C, Corno A, Giannico S, Marino B. Inferior vena cava-pulmonary artery extracardiac conduit: a new form of right heart bypass. J Thorac Cardiovasc Surg. 1990;100:228–232.
14.
Jacobs ML, Norwood WI Fontan operation: influence of modifications on morbidity and mortality. Ann Thorac Surg. 1994;58:945–951.
15.
Lemler MS, Scott WA, Leonard SR, Stromberg D, Ramaciotti C. Fenestration improves clinical outcome of the Fontan procedure: a prospective, randomized study. Circulation. 2002;105:207–212.
16.
de Leval MR, Dubini G, Migliavacca F, Jalali H, Camporini G, Redington A, Pietrabissa R. Use of computational fluid dynamics in the design of surgical procedures: application to the study of competitive flows in cavo-pulmonary connections. J Thorac Cardiovasc Surg. 1996;111:502–513.
17.
Hsia TY, Migliavacca F, Pittaccio S, Radaelli A, Dubini G, Pennati G, de Leval M. Computational fluid dynamic study of flow optimization in realistic models of the total cavopulmonary connections. J Surg Res. 2004;116:305–313. doi: 10.1016/j.jss.2003.08.004
18.
Bove EL, de Leval MR, Migliavacca F, Guadagni G, Dubini G. Computational fluid dynamics in the evaluation of hemodynamic performance of cavopulmonary connections after the Norwood procedure for hypoplastic left heart syndrome. J Thorac Cardiovasc Surg. 2003;126:1040–1047. doi: 10.1016/S0022
19.
Martin MH, Feinstein JA, Chan FP, Marsden AL, Yang W, Reddy VM. Technical feasibility and intermediate outcomes of using a handcrafted, area-preserving, bifurcated Y-graft modification of the Fontan procedure. J Thorac Cardiovasc Surg. 2015;149:239–245.e1. doi: 10.1016/j.jtcvs.2014.08.058
20.
Hsia TY. Taming the Fontan with the Y-graft: a nod and a wink to the great Yu. J Thorac Cardiovasc Surg. 2016;151:1537–1539. doi: 10.1016/j.jtcvs.2016.03.027
21.
Galantowicz M, Cheatham JP. Fontan completion without surgery. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2004;7:48–55.
22.
Daley M, d'Udekem Y. In patients undergoing Fontan completion, does a younger age at operation result in better long-term exercise capacity and prognosis? Interact Cardiovasc Thorac Surg. 2019;28:301–305. doi: 10.1093/icvts/ivy219
23.
Bolin EH, Maskatia SA, Tate AL, Petit CJ. Older age at completion of Fontan procedure is associated with improved percentage of predicted maximum oxygen uptake. Tex Heart Inst J. 2015;42:333–340. doi: 10.14503/THIJ-14-4178
24.
Fogel MA, Pawlowski TW, Whitehead KK, Harris MA, Keller MS, Glatz AC, Zhu W, Shore D, Diaz LK, Rome JJ. Cardiac magnetic resonance and the need for routine cardiac catheterization in single ventricle patients prior to Fontan: a comparison of 3 groups: pre-Fontan CMR versus cath evaluation. J Am Coll Cardiol. 2012;60:1094–1102. doi: 10.1016/j.jacc.2012.06.021
25.
Choussat, A, Fontan, F, Besse, P, Vallot, F, Chauve, A. Selection criteria for Fontan's procedure. Anderson, R, Shinebourne, E, eds. In: Pediatric Cardiology. Edinburgh, UK: Churchill Livingstone; 1978:559–566.
26.
Stern HJ. Fontan “Ten Commandments” revisited and revised. Pediatr Cardiol. 2010;31:1131–1134. doi: 10.1007/s00246-010-9811-9
27.
Tomaske M, Gerritse B, Kretzers L, Pretre R, Dodge-Khatami A, Rahn M, Bauersfeld U. A 12-year experience of bipolar steroid-eluting epicardial pacing leads in children. Ann Thorac Surg. 2008;85:1704–1711. doi: 10.1016/j.athoracsur.2008.02.016
28.
Iyengar AJ, Winlaw DS, Galati JC, Celermajer DS, Wheaton GR, Gentles TL, Grigg LE, Weintraub RG, Bullock A, Justo RN, d’Udekem Y. Trends in Fontan surgery and risk factors for early adverse outcomes after Fontan surgery: the Australia and New Zealand Fontan Registry experience. J Thorac Cardiovasc Surg. 2014;148:566–575. doi: 10.1016/j.jtcvs.2013.09.074
29.
Tweddell JS, Nersesian M, Mussatto KA, Nugent M, Simpson P, Mitchell ME, Ghanayem NS, Pelech AN, Marla R, Hoffman GM. Fontan palliation in the modern era: factors impacting mortality and morbidity. Ann Thorac Surg. 2009;88:1291–1299. doi: 10.1016/j.athoracsur.2009.05.076
30.
Australia and New Zealand Fontan Registry: Report 2016. 2016:1–10. https://www.fontanregistry.com/annual-reports. Accessed June 9, 2019.
31.
d’Udekem Y, Iyengar AJ, Cochrane AD, Grigg LE, Ramsay JM, Wheaton GR, Penny DJ, Brizard CP. The Fontan procedure: contemporary techniques have improved long-term outcomes. Circulation. 2007;116(suppl):I157–I164. doi: 10.1161/CIRCULATIONAHA.106.676445
32.
Ohye RG, Schranz D, D’Udekem Y. Current therapy for hypoplastic left heart syndrome and related single ventricle lesions. Circulation. 2016;134:1265–1279. doi: 10.1161/CIRCULATIONAHA.116.022816
33.
Iyengar AJ, Winlaw DS, Galati JC, Wheaton GR, Gentles TL, Grigg LE, Justo RN, Radford DJ, Weintraub RG, Bullock A, Celermajer DS, d’Udekem Y; Australia and New Zealand Fontan Registry. The extracardiac conduit Fontan procedure in Australia and New Zealand: hypoplastic left heart syndrome predicts worse early and late outcomes. Eur J Cardiothorac Surg. 2014;46:465–473. doi: 10.1093/ejcts/ezu015
34.
Hirsch JC, Goldberg C, Bove EL, Salehian S, Lee T, Ohye RG, Devaney EJ. Fontan operation in the current era: a 15-year single institution experience. Ann Surg. 2008;248:402–410. doi: 10.1097/SLA.0b013e3181858286
35.
Burkhart HM, Dearani JA, Mair DD, Warnes CA, Rowland CC, Schaff HV, Puga FJ, Danielson GK. The modified Fontan procedure: early and late results in 132 adult patients. J Thorac Cardiovasc Surg. 2003;125:1252–1259.
36.
Driscoll DJ, Offord KP, Feldt RH, Schaff HV, Puga FJ, Danielson GK. Five- to fifteen-year follow-up after Fontan operation. Circulation. 1992;85:469–496.
37.
Gaynor JW, Bridges ND, Cohen MI, Mahle WT, Decampli WM, Steven JM, Nicolson SC, Spray TL. Predictors of outcome after the Fontan operation: is hypoplastic left heart syndrome still a risk factor? J Thorac Cardiovasc Surg. 2002;123:237–245.
38.
Mitchell ME, Ittenbach RF, Gaynor JW, Wernovsky G, Nicolson S, Spray TL. Intermediate outcomes after the Fontan procedure in the current era. J Thorac Cardiovasc Surg. 2006;131:172–180. doi: 10.1016/j.jtcvs.2005.08.047
39.
Mertens L, Hagler DJ, Sauer U, Somerville J, Gewillig M. Protein-losing enteropathy after the Fontan operation: an international multicenter study: PLE Study Group. J Thorac Cardiovasc Surg. 1998;115:1063–1073.
40.
Carins TA, Shi WY, Iyengar AJ, Nisbet A, Forsdick V, Zannino D, Gentles T, Radford DJ, Justo R, Celermajer DS, Bullock A, Winlaw D, Wheaton G, Grigg L, d’Udekem Y. Long-term outcomes after first-onset arrhythmia in Fontan physiology. J Thorac Cardiovasc Surg. 2016;152:1355–1363.e1. doi: 10.1016/j.jtcvs.2016.07.073
41.
Egbe AC, Connolly HM, McLeod CJ, Ammash NM, Niaz T, Yogeswaran V, Poterucha JT, Qureshi MY, Driscoll DJ. Thrombotic and embolic complications associated with atrial arrhythmia after Fontan operation: role of prophylactic therapy. J Am Coll Cardiol. 2016;68:1312–1319. doi: 10.1016/j.jacc.2016.06.056
42.
Bulic A, Zimmerman FJ, Ceresnak SR, Shetty I, Motonaga KS, Freter A, Trela AV, Hanisch D, Russo L, Avasarala K, Dubin AM. Ventricular pacing in single ventricles-A bad combination. Heart Rhythm. 2017;14:853–857. doi: 10.1016/j.hrthm.2017.03.035
43.
Shi WY, Yong MS, McGiffin DC, Jain P, Ruygrok PN, Marasco SF, Finucane K, Keogh A, d’Udekem Y, Weintraub RG, Konstantinov IE. Heart transplantation in Fontan patients across Australia and New Zealand. Heart. 2016;102:1120–1126. doi: 10.1136/heartjnl-2015-308848
44.
Atz AM, Zak V, Mahony L, Uzark K, D’agincourt N, Goldberg DJ, Williams RV, Breitbart RE, Colan SD, Burns KM, Margossian R, Henderson HT, Korsin R, Marino BS, Daniels K, McCrindle BW; Pediatric Heart Network Investigators. Longitudinal outcomes of patients with single ventricle after the Fontan procedure. J Am Coll Cardiol. 2017;69:2735–2744. doi: 10.1016/j.jacc.2017.03.582
45.
Gewillig M, Brown SC. The Fontan circulation after 45 years: update in physiology. Heart. 2016;102:1081–1086. doi: 10.1136/heartjnl-2015-307467
46.
Veldtman GR, Opotowsky AR, Wittekind SG, Rychik J, Penny DJ, Fogel M, Marino BS, Gewillig M. Cardiovascular adaptation to the Fontan circulation. Congenit Heart Dis. 2017;12:699–710. doi: 10.1111/chd.12526
47.
Gewillig M, Brown SC, Eyskens B, Heying R, Ganame J, Budts W, La Gerche A, Gorenflo M. The Fontan circulation: who controls cardiac output? Interact Cardiovasc Thorac Surg. 2010;10:428–433. doi: 10.1510/icvts.2009.218594
48.
La Gerche A, Gewillig M. What limits cardiac performance during exercise in normal subjects and in healthy Fontan patients? Int J Pediatr. 2010;2010:791291. doi: 10.1155/2010/791291
49.
Gewillig MH, Lundström UR, Deanfield JE, Bull C, Franklin RC, Graham TP, Wyse RK. Impact of Fontan operation on left ventricular size and contractility in tricuspid atresia. Circulation. 1990;81:118–127.
50.
Anderson PA, Sleeper LA, Mahony L, Colan SD, Atz AM, Breitbart RE, Gersony WM, Gallagher D, Geva T, Margossian R, McCrindle BW, Paridon S, Schwartz M, Stylianou M, Williams RV, Clark BJ; Pediatric Heart Network Investigators. Contemporary outcomes after the Fontan procedure: a Pediatric Heart Network multicenter study. J Am Coll Cardiol. 2008;52:85–98. doi: 10.1016/j.jacc.2008.01.074
51.
Rathod RH, Prakash A, Powell AJ, Geva T. Myocardial fibrosis identified by cardiac magnetic resonance late gadolinium enhancement is associated with adverse ventricular mechanics and ventricular tachycardia late after Fontan operation. J Am Coll Cardiol. 2010;55:1721–1728. doi: 10.1016/j.jacc.2009.12.036
52.
Ho PK, Lai CT, Wong SJ, Cheung YF. Three-dimensional mechanical dyssynchrony and myocardial deformation of the left ventricle in patients with tricuspid atresia after Fontan procedure. J Am Soc Echocardiogr. 2012;25:393–400. doi: 10.1016/j.echo.2012.01.003
53.
Rychik J, Jacobs ML. Norwood WI. Acute changes in left ventricular geometry after volume reduction operation. Ann Thorac Surg. 1995;60:1267–1273.
54.
Fogel MA, Weinberg PM, Chin AJ, Fellows KE, Hoffman EA. Late ventricular geometry and performance changes of functional single ventricle throughout staged Fontan reconstruction assessed by magnetic resonance imaging. J Am Coll Cardiol. 1996;28:212–221.
55.
Silverstein DM, Hansen DP, Ojiambo HP, Griswold HE. Left ventricular function in severe pure mitral stenosis as seen at the Kenyatta National Hospital. Am Heart J. 1980;99:727–733.
56.
Di Molfetta A, Iacobelli R, Guccione P, Di Chiara L, Rocchi M, Cobianchi Belisari F, Campanale M, Gagliardi MG, Filippelli S, Ferrari G, Amodeo A. Evolution of ventricular energetics in the different stages of palliation of hypoplastic left heart syndrome: a retrospective clinical study. Pediatr Cardiol. 2017;38:1613–1619. doi: 10.1007/s00246-017-1704-8
57.
Biglino G, Schievano S, Steeden JA, Ntsinjana H, Baker C, Khambadkone S, de Leval MR, Hsia TY, Taylor AM, Giardini A; Modeling of Congenital Hearts Alliance (MOCHA) Collaborative Group. Reduced ascending aorta distensibility relates to adverse ventricular mechanics in patients with hypoplastic left heart syndrome: noninvasive study using wave intensity analysis. J Thorac Cardiovasc Surg. 2012;144:1307–1313. doi: 10.1016/j.jtcvs.2012.08.028
58.
Mitchell MB, Campbell DN, Ivy D, Boucek MM, Sondheimer HM, Pietra B, Das BB, Coll JR. Evidence of pulmonary vascular disease after heart transplantation for Fontan circulation failure. J Thorac Cardiovasc Surg. 2004;128:693–702. doi: 10.1016/j.jtcvs.2004.07.013
59.
Goldstein BH, Connor CE, Gooding L, Rocchini AP. Relation of systemic venous return, pulmonary vascular resistance, and diastolic dysfunction to exercise capacity in patients with single ventricle receiving Fontan palliation. Am J Cardiol. 2010;105:1169–1175. doi: 10.1016/j.amjcard.2009.12.020
60.
Gentles TL, Mayer JE, Gauvreau K, Newburger JW, Lock JE, Kupferschmid JP, Burnett J, Jonas RA, Castañeda AR, Wernovsky G. Fontan operation in five hundred consecutive patients: factors influencing early and late outcome. J Thorac Cardiovasc Surg. 1997;114:376–391.
61.
Poh CL, Zannino D, Weintraub RG, Winlaw DS, Grigg LE, Cordina R, Hornung T, Bullock A, Justo RN, Gentles TL, Verrall C, du Plessis K, Celermajer DS, d’Udekem Y. Three decades later: the fate of the population of patients who underwent the atriopulmonary Fontan procedure. Int J Cardiol. 2017;231:99–104. doi: 10.1016/j.ijcard.2017.01.057
62.
Diller GP, Giardini A, Dimopoulos K, Gargiulo G, Müller J, Derrick G, Giannakoulas G, Khambadkone S, Lammers AE, Picchio FM, Gatzoulis MA, Hager A. Predictors of morbidity and mortality in contemporary Fontan patients: results from a multicenter study including cardiopulmonary exercise testing in 321 patients. Eur Heart J. 2010;31:3073–3083. doi: 10.1093/eurheartj/ehq356
63.
Zomer AC, Vaartjes I, van der Velde ET, de Jong HM, Konings TC, Wagenaar LJ, Heesen WF, Eerens F, Baur LH, Grobbee DE, Mulder BJ. Heart failure admissions in adults with congenital heart disease; risk factors and prognosis. Int J Cardiol. 2013;168:2487–2493. doi: 10.1016/j.ijcard.2013.03.003
64.
Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE, Drazner MH, Fonarow GC, Geraci SA, Horwich T, Januzzi JL, Johnson MR, Kasper EK, Levy WC, Masoudi FA, McBride PE, McMurray JJ, Mitchell JE, Peterson PN, Riegel B, Sam F, Stevenson LW, Tang WH, Tsai EJ, Wilkoff BL. 2013 ACCF/AHA guideline for the management of heart failure: executive summary: a report of the American College of Cardiology Foundation/American Heart Association Task Force on practice guidelines. Circulation. 2013;128:1810–1852. doi: 10.1161/CIR.0b013e31829e8807
65.
Book WM, Gerardin J, Saraf A, Marie Valente A, Rodriguez F Clinical phenotypes of Fontan failure: implications for management. Congenit Heart Dis. 2016;11:296–308. doi: 10.1111/chd.12368
66.
Poterucha JT, Egbe AC, Johnson JN, Niaz T, Wackel PL, Cannon BC, Eidem BW, Cetta F. Improved ventricular function after TEE-guided cardioversion of atrial arrhythmias in patients after the Fontan operation. Congenit Heart Dis. 2016;11:578–583. doi: 10.1111/chd.12339
67.
Miller JR, Simpson KE, Epstein DJ, Lancaster TS, Henn MC, Schuessler RB, Balzer DT, Shahanavaz S, Murphy JJ, Canter CE, Eghtesady P, Boston US. Improved survival after heart transplant for failed Fontan patients with preserved ventricular function. J Heart Lung Transplant. 2016;35:877–883. doi: 10.1016/j.healun.2016.02.005
68.
Griffiths ER, Kaza AK, Wyler von Ballmoos MC, Loyola H, Valente AM, Blume ED, del Nido P. Evaluating failing Fontans for heart transplantation: predictors of death. Ann Thorac Surg. 2009;88:558–563.
69.
Rathod RH, Prakash A, Kim YY, Germanakis IE, Powell AJ, Gauvreau K, Geva T. Cardiac magnetic resonance parameters predict transplantation-free survival in patients with Fontan circulation. Circ Cardiovasc Imaging. 2014;7:502–509. doi: 10.1161/CIRCIMAGING.113.001473
70.
Margossian R, Schwartz ML, Prakash A, Wruck L, Colan SD, Atz AM, Bradley TJ, Fogel MA, Hurwitz LM, Marcus E, Powell AJ, Printz BF, Puchalski MD, Rychik J, Shirali G, Williams R, Yoo SJ, Geva T; Pediatric Heart Network Investigators. Comparison of echocardiographic and cardiac magnetic resonance imaging measurements of functional single ventricular volumes, mass, and ejection fraction (from the Pediatric Heart Network Fontan Cross-Sectional Study). Am J Cardiol. 2009;104:419–428. doi: 10.1016/j.amjcard.2009.03.058
71.
Garofalo CA, Cabreriza SE, Quinn TA, Weinberg AD, Printz BF, Hsu DT, Quaegebeur JM, Mosca RS, Spotnitz HM. Ventricular diastolic stiffness predicts perioperative morbidity and duration of pleural effusions after the Fontan operation. Circulation. 2006;114(suppl):I56–I61. doi: 10.1161/CIRCULATIONAHA.105.001396
72.
Averin K, Hirsch R, Seckeler MD, Whiteside W, Beekman RH, Goldstein BH. Diagnosis of occult diastolic dysfunction late after the Fontan procedure using a rapid volume expansion technique. Heart. 2016;102:1109–1114. doi: 10.1136/heartjnl-2015-309042
73.
De Mey W, Cools B, Heying R, Budts W, Louw JJ, Boshoff DE, Brown SC, Gewillig M. Can a volume challenge pinpoint the limiting factor in a Fontan circulation? Acta Cardiol. 2015;70:536–542. doi: 10.2143/AC.70.5.3110514
74.
Driscoll DJ, Durongpisitkul K. Exercise testing after the Fontan operation. Pediatr Cardiol. 1999;20:57–59. doi: 10.1007/s002469900397
75.
Paridon SM, Mitchell PD, Colan SD, Williams RV, Blaufox A, Li JS, Margossian R, Mital S, Russell J, Rhodes J; Pediatric Heart Network Investigators. A cross-sectional study of exercise performance during the first 2 decades of life after the Fontan operation. J Am Coll Cardiol. 2008;52:99–107. doi: 10.1016/j.jacc.2008.02.081
76.
Bridges ND, Lock JE, Castaneda AR. Baffle fenestration with subsequent transcatheter closure: modification of the Fontan operation for patients at increased risk. Circulation. 1990;82:1681–1689.
77.
Gorla SR, Jhingoeri NK, Chakraborty A, Raja KR, Garg A, Sandhu S, Rosenkranz ER, Swaminathan S. Incidence and factors influencing the spontaneous closure of Fontan fenestration. Congenit Heart Dis. 2018;13:776–781. doi: 10.1111/chd.12652
78.
Heinemann M, Breuer J, Steger V, Steil E, Sieverding L, Ziemer G. Incidence and impact of systemic venous collateral development after Glenn and Fontan procedures. Thorac Cardiovasc Surg. 2001;49:172–178. doi: 10.1055/s-2001-14339
79.
Lluri G, Levi DS, Aboulhosn J. Systemic to pulmonary venous collaterals in adults with single ventricle physiology after cavopulmonary palliation. Int J Cardiol. 2015;189:159–163. doi: 10.1016/j.ijcard.2015.04.065
80.
Sugiyama H, Yoo SJ, Williams W, Benson LN. Characterization and treatment of systemic venous to pulmonary venous collaterals seen after the Fontan operation. Cardiol Young. 2003;13:424–430.
81.
Masura J, Bordacova L, Tittel P, Berden P, Podnar T. Percutaneous management of cyanosis in Fontan patients using Amplatzer occluders. Catheter Cardiovasc Interv. 2008;71:843–849.
82.
Mavroudis C, Zales VR, Backer CL, Muster AJ, Latson LA. Fenestrated Fontan with delayed catheter closure: effects of volume loading and baffle fenestration on cardiac index and oxygen delivery. Circulation. 1992;86(suppl):II85–II92.
83.
Hijazi ZM, Fahey JT, Kleinman CS, Kopf GS, Hellenbrand WE. Hemodynamic evaluation before and after closure of fenestrated Fontan. An acute study of changes in oxygen delivery. Circulation. 1992;86:196–202.
84.
Poterucha JT, Johnson JN, Taggart NW, Cabalka AK, Hagler DJ, Driscoll DJ, Cetta F. Embolization of veno-venous collaterals after the Fontan operation Is associated with decreased survival. Congenit Heart Dis. 2015;10:E230–E236. doi: 10.1111/chd.12276
85.
Srivastava D, Preminger T, Lock JE, Mandell V, Keane JF, Mayer JE, Kozakewich H, Spevak PJ. Hepatic venous blood and the development of pulmonary arteriovenous malformations in congenital heart disease. Circulation. 1995;92:1217–1222.
86.
Vettukattil JJ. Is the hepatic factor a miRNA that maintains the integrity of pulmonary microvasculature by inhibiting the vascular endothelial growth factor? Curr Cardiol Rev. 2017;13:244–250. doi: 10.2174/1573403X13666170510115628
87.
Shah MJ, Rychik J, Fogel MA, Murphy JD, Jacobs ML. Pulmonary AV malformations after superior cavopulmonary connection: resolution after inclusion of hepatic veins in the pulmonary circulation. Ann Thorac Surg. 1997;63:960–963.
88.
McElhinney DB, Kreutzer J, Lang P, Mayer JE, del Nido PJ, Lock JE. Incorporation of the hepatic veins into the cavopulmonary circulation in patients with heterotaxy and pulmonary arteriovenous malformations after a Kawashima procedure. Ann Thorac Surg. 2005;80:1597–1603. doi: 10.1016/j.athoracsur.2005.05.101
89.
Pike NA, Vricella LA, Feinstein JA, Black MD, Reitz BA. Regression of severe pulmonary arteriovenous malformations after Fontan revision and “hepatic factor” rerouting. Ann Thorac Surg. 2004;78:697–699. doi: 10.1016/j.athoracsur.2004.02.003
90.
McElhinney DB, Marshall AC, Lang P, Lock JE, Mayer JE Creation of a brachial arteriovenous fistula for treatment of pulmonary arteriovenous malformations after cavopulmonary anastomosis. Ann Thorac Surg. 2005;80:1604–1609. doi: 10.1016/j.athoracsur.2005.05.100
91.
Sundareswaran KS, de Zélicourt D, Sharma S, Kanter KR, Spray TL, Rossignac J, Sotiropoulos F, Fogel MA, Yoganathan AP. Correction of pulmonary arteriovenous malformation using image-based surgical planning. JACC Cardiovasc Imaging. 2009;2:1024–1030. doi: 10.1016/j.jcmg.2009.03.019
92.
de Zélicourt DA, Haggerty CM, Sundareswaran KS, Whited BS, Rossignac JR, Kanter KR, Gaynor JW, Spray TL, Sotiropoulos F, Fogel MA, Yoganathan AP. Individualized computer-based surgical planning to address pulmonary arteriovenous malformations in patients with a single ventricle with an interrupted inferior vena cava and azygous continuation. J Thorac Cardiovasc Surg. 2011;141:1170–1177. doi: 10.1016/j.jtcvs.2010.11.032
93.
Tomkiewicz-Pajak L, Plazak W, Kolcz J, Pajak J, Kopec G, Dluzniewska N, Olszowska M, Moryl-Bujakowska A, Podolec P. Iron deficiency and hematological changes in adult patients after Fontan operation. J Cardiol. 2014;64:384–389. doi: 10.1016/j.jjcc.2014.02.021
94.
Barber G, Helton JG, Aglira BA, Chin AJ, Murphy JD, Pigott JD, Norwood WI. The significance of tricuspid regurgitation in hypoplastic left-heart syndrome. Am Heart J. 1988;116(pt 1):1563–1567.
95.
King G, Gentles TL, Winlaw DS, Cordina R, Bullock A, Grigg LE, Alphonso N, Radford DJ, Zannino D, Buratto E, d’Udekem Y. Common atrioventricular valve failure during single ventricle palliation†. Eur J Cardiothorac Surg. 2017;51:1037–1043. doi: 10.1093/ejcts/ezx025
96.
Ohye RG, Gomez CA, Goldberg CS, Graves HL, Devaney EJ, Bove EL. Tricuspid valve repair in hypoplastic left heart syndrome. J Thorac Cardiovasc Surg. 2004;127:465–472. doi: 10.1016/j.jtcvs.2003.07.053
97.
Takahashi K, Inage A, Rebeyka IM, Ross DB, Thompson RB, Mackie AS, Smallhorn JF. Real-time 3-dimensional echocardiography provides new insight into mechanisms of tricuspid valve regurgitation in patients with hypoplastic left heart syndrome. Circulation. 2009;120:1091–1098. doi: 10.1161/CIRCULATIONAHA.108.809566
98.
Stamm C, Anderson RH, Ho SY. The morphologically tricuspid valve in hypoplastic left heart syndrome. Eur J Cardiothorac Surg. 1997;12:587–592.
99.
Hsu DT, Zak V, Mahony L, Sleeper LA, Atz AM, Levine JC, Barker PC, Ravishankar C, McCrindle BW, Williams RV, Altmann K, Ghanayem NS, Margossian R, Chung WK, Border WL, Pearson GD, Stylianou MP, Mital S; for the Pediatric Heart Network Investigators. Enalapril in infants with single ventricle: results of a multicenter randomized trial. Circulation. 2010;122:333–340. doi: 10.1161/CIRCULATIONAHA.109.927988
100.
Alsaied T, Bokma JP, Engel ME, Kuijpers JM, Hanke SP, Zuhlke L, Zhang B, Veldtman GR. Predicting long-term mortality after Fontan procedures: a risk score based on 6707 patients from 28 studies. Congenit Heart Dis. 2017;12:393–398. doi: 10.1111/chd.12468
101.
Mavroudis C, Stewart RD, Backer CL, Deal BJ, Young L, Franklin WH. Atrioventricular valve procedures with repeat Fontan operations: influence of valve pathology, ventricular function, and arrhythmias on outcome. Ann Thorac Surg. 2005;80:29–36.
102.
Honjo O, Mertens L, Van Arsdell GS. Atrioventricular valve repair in patients with single-ventricle physiology: mechanisms, techniques of repair, and clinical outcomes. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2011;14:75–84. doi: 10.1053/j.pcsu.2011.02.001
103.
Kotani Y, Chetan D, Atlin CR, Mertens LL, Jegatheeswaran A, Caldarone CA, Van Arsdell GS, Honjo O. Longevity and durability of atrioventricular valve repair in single-ventricle patients. Ann Thorac Surg. 2012;94:2061–2069. doi: 10.1016/j.athoracsur.2012.04.048
104.
Ghai A, Harris L, Harrison DA, Webb GD, Siu SC. Outcomes of late atrial tachyarrhythmias in adults after the Fontan operation. J Am Coll Cardiol. 2001;37:585–592.
105.
Giannakoulas G, Dimopoulos K, Yuksel S, Inuzuka R, Pijuan-Domenech A, Hussain W, Tay EL, Gatzoulis MA, Wong T. Atrial tachyarrhythmias late after Fontan operation are related to increase in mortality and hospitalization. Int J Cardiol. 2012;157:221–226. doi: 10.1016/j.ijcard.2010.12.049
106.
Yap SC, Harris L, Chauhan VS, Oechslin EN, Silversides CK. Identifying high risk in adults with congenital heart disease and atrial arrhythmias. Am J Cardiol. 2011;108:723–728. doi: 10.1016/j.amjcard.2011.04.021
107.
Collins RT, Doshi P, Onukwube J, Fram RY, Robbins JM. Risk factors for increased hospital resource utilization and in-hospital mortality in adults with single ventricle congenital heart disease. Am J Cardiol. 2016;118:453–462. doi: 10.1016/j.amjcard.2016.05.020
108.
Collins RT, Fram RY, Tang X, Robbins JM, St John Sutton M. Hospital utilization in adults with single ventricle congenital heart disease and cardiac arrhythmias. J Cardiovasc Electrophysiol. 2014;25:179–186. doi: 10.1111/jce.12294
109.
Guichard JB, Nattel S. Atrial cardiomyopathy: a useful notion in cardiac disease management or a passing fad? J Am Coll Cardiol. 2017;70:756–765. doi: 10.1016/j.jacc.2017.06.033
110.
Gelatt M, Hamilton RM, McCrindle BW, Gow RM, Williams WG, Trusler GA, Freedom RM. Risk factors for atrial tachyarrhythmias after the Fontan operation. J Am Coll Cardiol. 1994;24:1735–1741.
111.
Fishberger SB, Wernovsky G, Gentles TL, Gauvreau K, Burnett J, Mayer JE, Walsh EP. Factors that influence the development of atrial flutter after the Fontan operation. J Thorac Cardiovasc Surg. 1997;113:80–86.
112.
Cohen MI, Wernovsky G, Vetter VL, Wieand TS, Gaynor JW, Jacobs ML, Spray TL, Rhodes LA. Sinus node function after a systematically staged Fontan procedure. Circulation. 1998;98(suppl):II352–II358.
113.
Giannico S, Hammad F, Amodeo A, Michielon G, Drago F, Turchetta A, Di Donato R, Sanders SP. Clinical outcome of 193 extracardiac Fontan patients: the first 15 years. J Am Coll Cardiol. 2006;47:2065–2073. doi: 10.1016/j.jacc.2005.12.065
114.
Stephenson EA, Lu M, Berul CI, Etheridge SP, Idriss SF, Margossian R, Reed JH, Prakash A, Sleeper LA, Vetter VL, Blaufox AD; Pediatric Heart Network Investigators. Arrhythmias in a contemporary Fontan cohort: prevalence and clinical associations in a multicenter cross-sectional study. J Am Coll Cardiol. 2010;56:890–896. doi: 10.1016/j.jacc.2010.03.079
115.
Balaji S, Daga A, Bradley DJ, Etheridge SP, Law IH, Batra AS, Sanatani S, Singh AK, Gajewski KK, Tsao S, Singh HR, Tisma-Dupanovic S, Tateno S, Takamuro M, Nakajima H, Roos-Hesselink JW, Shah M. An international multicenter study comparing arrhythmia prevalence between the intracardiac lateral tunnel and the extracardiac conduit type of Fontan operations. J Thorac Cardiovasc Surg. 2014;148:576–581. doi: 10.1016/j.jtcvs.2013.08.070
116.
Quinton E, Nightingale P, Hudsmith L, Thorne S, Marshall H, Clift P, de Bono J. Prevalence of atrial tachyarrhythmia in adults after Fontan operation. Heart. 2015;101:1672–1677. doi: 10.1136/heartjnl-2015-307514
117.
Song MK, Bae EJ, Kwon BS, Kim GB, Noh CI, Choi JY, Kim WH, Lee JR, Kim YJ. Intra-atrial reentrant tachycardia in adult patients after Fontan operation. Int J Cardiol. 2015;187:157–163. doi: 10.1016/j.ijcard.2015.03.157
118.
Pundi KN, Pundi KN, Johnson JN, Dearani JA, Li Z, Driscoll DJ, Wackel PL, McLeod CJ, Cetta F, Cannon BC. Sudden cardiac death and late arrhythmias after the Fontan operation. Congenit Heart Dis. 2017;12:17–23. doi: 10.1111/chd.12401
119.
Bossers SS, Duppen N, Kapusta L, Maan AC, Duim AR, Bogers AJ, Hazekamp MG, van Iperen G, Helbing WA, Blom NA. Comprehensive rhythm evaluation in a large contemporary Fontan population. Eur J Cardiothorac Surg. 2015;48:833–840. doi: 10.1093/ejcts/ezu548
120.
Diller GP, Dimopoulos K, Okonko D, Uebing A, Broberg CS, Babu-Narayan S, Bayne S, Poole-Wilson PA, Sutton R, Francis DP, Gatzoulis MA. Heart rate response during exercise predicts survival in adults with congenital heart disease. J Am Coll Cardiol. 2006;48:1250–1256. doi: 10.1016/j.jacc.2006.05.051
121.
Lasa JJ, Glatz AC, Daga A, Shah M. Prevalence of arrhythmias late after the Fontan operation. Am J Cardiol. 2014;113:1184–1188. doi: 10.1016/j.amjcard.2013.12.025
122.
Cohen MI, Bridges ND, Gaynor JW, Hoffman TM, Wernovsky G, Vetter VL, Spray TL, Rhodes LA. Modifications to the cavopulmonary anastomosis do not eliminate early sinus node dysfunction. J Thorac Cardiovasc Surg. 2000;120:891–900. doi: 10.1067/mtc.2000.109708
123.
Weipert J, Noebauer C, Schreiber C, Kostolny M, Zrenner B, Wacker A, Hess J, Lange R. Occurrence and management of atrial arrhythmia after long-term Fontan circulation. J Thorac Cardiovasc Surg. 2004;127:457–464. doi: 10.1016/j.jtcvs.2003.08.054
124.
Nakamura Y, Yagihara T, Kagisaki K, Hagino I, Kobayashi J. Ventricular performance in long-term survivors after Fontan operation. Ann Thorac Surg. 2011;91:172–180. doi: 10.1016/j.athoracsur.2010.07.055
125.
Deal BJ, Costello JM, Webster G, Tsao S, Backer CL, Mavroudis C. Intermediate-term outcome of 140 consecutive Fontan conversions with arrhythmia operations. Ann Thorac Surg. 2016;101:717–724. doi: 10.1016/j.athoracsur.2015.09.017
126.
Loomba RS, Willes RJ, Kovach JR, Anderson RH. Chronic arrhythmias in the setting of heterotaxy: differences between right and left isomerism. Congenit Heart Dis. 2016;11:7–18. doi: 10.1111/chd.12288
127.
Loomba RS, Buelow MW, Aggarwal S, Arora RR, Kovach J, Ginde S. Arrhythmias in adults with congenital heart disease: what are risk factors for specific arrhythmias? Pacing Clin Electrophysiol. 2017;40:353–361. doi: 10.1111/pace.12983
128.
Balaji S, Shah M. Lack of influence of heterotaxy on arrhythmia prevalence after intracardiac versus extracardiac Fontan operations. J Thorac Cardiovasc Surg. 2015;149:1224. doi: 10.1016/j.jtcvs.2014.12.044
129.
Reybrouck T, Vangesselen S, Gewillig M. Impaired chronotropic response to exercise in children with repaired cyanotic congenital heart disease. Acta Cardiol. 2009;64:723–727. doi: 10.2143/AC.64.6.2044734
130.
Ohuchi H, Hasegawa S, Yasuda K, Yamada O, Ono Y, Echigo S. Severely impaired cardiac autonomic nervous activity after the Fontan operation. Circulation. 2001;104:1513–1518.
131.
Davos CH, Francis DP, Leenarts MF, Yap SC, Li W, Davlouros PA, Wensel R, Coats AJ, Piepoli M, Sreeram N, Gatzoulis MA. Global impairment of cardiac autonomic nervous activity late after the Fontan operation. Circulation. 2003;108:(suppl 1):II180–II185.
132.
Diller GP, Dimopoulos K, Okonko D, Li W, Babu-Narayan SV, Broberg CS, Johansson B, Bouzas B, Mullen MJ, Poole-Wilson PA, Francis DP, Gatzoulis MA. Exercise intolerance in adult congenital heart disease: comparative severity, correlates, and prognostic implication. Circulation. 2005;112:828–835. doi: 10.1161/CIRCULATIONAHA.104.529800
133.
Egbe AC, Driscoll DJ, Khan AR, Said SS, Akintoye E, Berganza FM, Connolly HM. Cardiopulmonary exercise test in adults with prior Fontan operation: the prognostic value of serial testing. Int J Cardiol. 2017;235:6–10. doi: 10.1016/j.ijcard.2017.02.140
134.
Ohuchi H, Negishi J, Noritake K, Hayama Y, Sakaguchi H, Miyazaki A, Kagisaki K, Yamada O. Prognostic value of exercise variables in 335 patients after the Fontan operation: a 23-year single-center experience of cardiopulmonary exercise testing. Congenit Heart Dis. 2015;10:105–116. doi: 10.1111/chd.12222
135.
Fernandes SM, Alexander ME, Graham DA, Khairy P, Clair M, Rodriguez E, Pearson DD, Landzberg MJ, Rhodes J. Exercise testing identifies patients at increased risk for morbidity and mortality following Fontan surgery. Congenit Heart Dis. 2011;6:294–303. doi: 10.1111/j.1747-0803.2011.00500.x
136.
Elder RW, McCabe NM, Veledar E, Kogon BE, Jokhadar M, Rodriguez FH, McConnell ME, Book WM. Risk factors for major adverse events late after Fontan palliation. Congenit Heart Dis. 2015;10:159–168. doi: 10.1111/chd.12212
137.
Williams RV, Travison T, Kaltman JR, Cecchin F, Colan SD, Idriss SF, Lu M, Margossian R, Reed JH, Silver ES, Stephenson EA, Vetter VL; Pediatric Heart Network Investigators. Comparison of Fontan survivors with and without pacemakers: a report from the Pediatric Heart Network Fontan Cross-Sectional Study. Congenit Heart Dis. 2013;8:32–39. doi: 10.1111/j.1747-0803.2012.00692.x
138.
Fishberger SB, Wernovsky G, Gentles TL, Gamble WJ, Gauvreau K, Burnett J, Mayer JE, Walsh EP. Long-term outcome in patients with pacemakers following the Fontan operation. Am J Cardiol. 1996;77:887–889.
139.
Brida M, Baumgartner H, Gatzoulis MA, Diller GP. Early mortality and concomitant procedures related to Fontan conversion: quantitative analysis. Int J Cardiol. 2017;236:132–137. doi: 10.1016/j.ijcard.2017.01.111
140.
Khairy P, Van Hare GF, Balaji S, Berul CI, Cecchin F, Cohen MI, Daniels CJ, Deal BJ, Dearani JA, Groot Nd, Dubin AM, Harris L, Janousek J, Kanter RJ, Karpawich PP, Perry JC, Seslar SP, Shah MJ, Silka MJ, Triedman JK, Walsh EP, Warnes CA. PACES/HRS expert consensus statement on the recognition and management of arrhythmias in adult congenital heart disease: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Heart Rhythm. 2014;11:e102–e165. doi: 10.1016/j.hrthm.2014.05.009
141.
Page RL, Joglar JA, Caldwell MA, Calkins H, Conti JB, Deal BJ, Estes NA, Field ME, Goldberger ZD, Hammill SC, Indik JH, Lindsay BD, Olshansky B, Russo AM, Shen WK, Tracy CM, Al-Khatib SM. 2015 ACC/AHA/HRS guideline for the management of adult patients with supraventricular tachycardia: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society [published correction appears in Circulation. 2016;13r:e234–e235]. Circulation. 2016;133:e506–e574. doi: 10.1161/CIR.0000000000000311
142.
Egbe AC, Connolly HM, Khan AR, Niaz T, Said SS, Dearani JA, Warnes CA, Deshmukh AJ, Kapa S, McLeod CJ. Outcomes in adult Fontan patients with atrial tachyarrhythmias. Am Heart J. 2017;186:12–20. doi: 10.1016/j.ahj.2016.12.015
143.
Yap SC, Harris L, Silversides CK, Downar E, Chauhan VS. Outcome of intra-atrial re-entrant tachycardia catheter ablation in adults with congenital heart disease: negative impact of age and complex atrial surgery. J Am Coll Cardiol. 2010;56:1589–1596. doi: 10.1016/j.jacc.2010.04.061
144.
Kannankeril PJ, Anderson ME, Rottman JN, Wathen MS, Fish FA. Frequency of late recurrence of intra-atrial reentry tachycardia after radiofrequency catheter ablation in patients with congenital heart disease. Am J Cardiol. 2003;92:879–881.
145.
Betts TR, Roberts PR, Allen SA, Salmon AP, Keeton BR, Haw MP, Morgan JM. Electrophysiological mapping and ablation of intra-atrial reentry tachycardia after Fontan surgery with the use of a noncontact mapping system. Circulation. 2000;102:419–425.
146.
Yap SC, Harris L, Downar E, Nanthakumar K, Silversides CK, Chauhan VS. Evolving electroanatomic substrate and intra-atrial reentrant tachycardia late after Fontan surgery. J Cardiovasc Electrophysiol. 2012;23:339–345. doi: 10.1111/j.1540-8167.2011.02202.x
147.
de Groot NM, Lukac P, Blom NA, van Kuijk JP, Pedersen AK, Hansen PS, Delacretaz E, Schalij MJ. Long-term outcome of ablative therapy of postoperative supraventricular tachycardias in patients with univentricular heart: a European multicenter study. Circ Arrhythm Electrophysiol. 2009;2:242–248. doi: 10.1161/CIRCEP.108.828137
148.
de Groot NM, Zeppenfeld K, Wijffels MC, Chan WK, Blom NA, Van der Wall EE, Schalij MJ. Ablation of focal atrial arrhythmia in patients with congenital heart defects after surgery: role of circumscribed areas with heterogeneous conduction. Heart Rhythm. 2006;3:526–535. doi: 10.1016/j.hrthm.2006.01.011
149.
Correa R, Walsh EP, Alexander ME, Mah DY, Cecchin F, Abrams DJ, Triedman JK. Transbaffle mapping and ablation for atrial tachycardias after Mustard, Senning, or Fontan operations. J Am Heart Assoc. 2013;2:e000325. doi: 10.1161/JAHA.113.000325
150.
Deal BJ, Mavroudis C, Backer CL, Buck SH, Johnsrude C. Comparison of anatomic isthmus block with the modified right atrial maze procedure for late atrial tachycardia in Fontan patients. Circulation. 2002;106:575–579.
151.
Hiramatsu T, Iwata Y, Matsumura G, Konuma T, Yamazaki K. Impact of Fontan conversion with arrhythmia surgery and pacemaker therapy. Eur J Cardiothorac Surg. 2011;40:1007–1010. doi: 10.1016/j.ejcts.2011.01.022
152.
Mossad EB, Motta P, Vener DF. Anesthetic considerations for adults undergoing Fontan conversion surgery. Anesthesiol Clin. 2013;31:405–419. doi: 10.1016/j.anclin.2012.12.005
153.
Poh CL, Cochrane A, Galati JC, Bullock A, Celermajer DS, Gentles T, du Plessis K, Winlaw DS, Hornung T, Finucane K, d’Udekem Y. Ten-year outcomes of Fontan conversion in Australia and New Zealand demonstrate the superiority of a strategy of early conversion. Eur J Cardiothorac Surg. 2016;49:530–535. doi: 10.1093/ejcts/ezv112
154.
Takahashi K, Fynn-Thompson F, Cecchin F, Khairy P, del Nido P, Triedman JK. Clinical outcomes of Fontan conversion surgery with and without associated arrhythmia intervention. Int J Cardiol. 2009;137:260–266. doi: 10.1016/j.ijcard.2008.06.074
155.
Said SM, Burkhart HM, Schaff HV, Cetta F, Driscoll DJ, Li Z, Oliver WC, Ammash NM, Dearani JA. Fontan conversion: identifying the high-risk patient. Ann Thorac Surg. 2014;97:2115–2121. doi: 10.1016/j.athoracsur.2014.01.083
156.
Mavroudis C, Deal BJ. Fontan conversion: literature review and lessons learned over 20 years. World J Pediatr Congenit Heart Surg. 2016;7:192–198. doi: 10.1177/2150135115623960
157.
Yang G, Du X, Ni B, Chen H, Qi R, Cai C, Fang Y, Yang B, Ju W, Zhang F, Li M, Gu K, Shao Y, Chen M. Prevention of postsurgical atrial tachycardia with a modified right atrial free wall incision. Heart Rhythm. 2015;12:1611–1618. doi: 10.1016/j.hrthm.2015.03.026
158.
Hosseinpour AR, Adsuar-Gómez A, González-Calle A, Pedrote A, Arana-Rueda E, García-Riesco L, Arce-León Á, Jiménez-Velasco A, Borrego-Domínguez JM, Ordóñez-Fernández A. A simple surgical technique to prevent atrial reentrant tachycardia in surgery for congenital heart disease. Interact Cardiovasc Thorac Surg. 2016;22:47–52. doi: 10.1093/icvts/ivv268
159.
Collins KK, Rhee EK, Delucca JM, Alexander ME, Bevilacqua LM, Berul CI, Walsh EP, Mayer JE, Jonas RA, del Nido PJ, Triedman JK. Modification to the Fontan procedure for the prophylaxis of intra-atrial reentrant tachycardia: short-term results of a prospective randomized blinded trial. J Thorac Cardiovasc Surg. 2004;127:721–729. doi: 10.1016/S0022
160.
Cohen MI, Rhodes LA, Spray TL, Gaynor JW. Efficacy of prophylactic epicardial pacing leads in children and young adults. Ann Thorac Surg. 2004;78:197–202. doi: 10.1016/j.athoracsur.2004.02.008
161.
Lim HG, Lee JR, Kim YJ. the effects of modification to lateral tunnel Fontan procedure for prophylactic arrhythmia surgery. Ann Thorac Surg. 2017;104:197–204. doi: 10.1016/j.athoracsur.2016.11.033
162.
Kossaify A, Nicolas N. Impact of overweight and obesity on left ventricular diastolic function and value of tissue Doppler echocardiography. Clin Med Insights Cardiol. 2013;7:43–50. doi: 10.4137/CMC.S11156
163.
Lau DH, Nattel S, Kalman JM, Sanders P. Modifiable risk factors and atrial fibrillation. Circulation. 2017;136:583–596. doi: 10.1161/CIRCULATIONAHA.116.023163
164.
John AS, Johnson JA, Khan M, Driscoll DJ, Warnes CA, Cetta F. Clinical outcomes and improved survival in patients with protein-losing enteropathy after the Fontan operation. J Am Coll Cardiol. 2014;64:54–62. doi: 10.1016/j.jacc.2014.04.025
165.
Meadows J, Jenkins K. Protein-losing enteropathy: integrating a new disease paradigm into recommendations for prevention and treatment. Cardiol Young. 2011;21:363–377. doi: 10.1017/S1047951111000102
166.
Allen KY, Downing TE, Glatz AC, Rogers LS, Ravishankar C, Rychik J, Fuller S, Montenegro LM, Steven JM, Spray TL, Nicolson SC, Gaynor JW, Goldberg DJ. Effect of Fontan-associated morbidities on survival with intact Fontan circulation. Am J Cardiol. 2017;119:1866–1871. doi: 10.1016/j.amjcard.2017.03.004
167.
Itkin M, Piccoli DA, Nadolski G, Rychik J, DeWitt A, Pinto E, Rome J, Dori Y. Protein-losing enteropathy in patients with congenital heart disease. J Am Coll Cardiol. 2017;69:2929–2937. doi: 10.1016/j.jacc.2017.04.023
168.
Meadows J, Gauvreau K, Jenkins K. Lymphatic obstruction and protein-losing enteropathy in patients with congenital heart disease. Congenit Heart Dis. 2008;3:269–276. doi: 10.1111/j.1747-0803.2008.00201.x
169.
Vyas H, Driscoll DJ, Cabalka AK, Cetta F, Hagler DJ. Results of transcatheter Fontan fenestration to treat protein losing enteropathy. Catheter Cardiovasc Interv. 2007;69:584–589. doi: 10.1002/ccd.21045
170.
Rychik J, Gui-Yang S. Relation of mesenteric vascular resistance after Fontan operation and protein-losing enteropathy. Am J Cardiol. 2002;90:672–674.
171.
Ostrow AM, Freeze H, Rychik J. Protein-losing enteropathy after Fontan operation: investigations into possible pathophysiologic mechanisms. Ann Thorac Surg. 2006;82:695–700. doi: 10.1016/j.athoracsur.2006.02.048
172.
Bode L, Freeze HH. Applied glycoproteomics: approaches to study genetic-environmental collisions causing protein-losing enteropathy. Biochim Biophys Acta. 2006;1760:547–559. doi: 10.1016/j.bbagen.2005.11.004
173.
Schumacher KR, Stringer KA, Donohue JE, Yu S, Shaver A, Caruthers RL, Zikmund-Fisher BJ, Fifer C, Goldberg C, Russell MW. Fontan-associated protein-losing enteropathy and plastic bronchitis. J Pediatr. 2015;166:970–977. doi: 10.1016/j.jpeds.2014.12.068
174.
Goldberg DJ, Dodds K, Avitabile CM, Glatz AC, Brodsky JL, Semeao EJ, Rand EB, Mancilla EE, Rychik J. Children with protein-losing enteropathy after the Fontan operation are at risk for abnormal bone mineral density. Pediatr Cardiol. 2012;33:1264–1268. doi: 10.1007/s00246-012-0290-z
175.
Magdo HS, Stillwell TL, Greenhawt MJ, Stringer KA, Yu S, Fifer CG, Russell MW, Schumacher KR. Immune abnormalities in Fontan protein-losing enteropathy: a case-control study. J Pediatr. 2015;167:331–337. doi: 10.1016/j.jpeds.2015.04.061
176.
Morsheimer MM, Rychik J, Forbes L, Dodds K, Goldberg DJ, Sullivan K, Heimall JR. Risk factors and clinical significance of lymphopenia in survivors of the Fontan procedure for single-ventricle congenital cardiac disease. J Allergy Clin Immunol Pract. 2016;4:491–496. doi: 10.1016/j.jaip.2015.11.034
177.
Vyas H, Driscoll DJ, Cetta F, Loftus CG, Connolly HM. Gastrointestinal bleeding and protein-losing enteropathy after the Fontan operation. Am J Cardiol. 2006;98:666–667. doi: 10.1016/j.amjcard.2006.03.049
178.
Florent C, Lévy VG, Bernier JJ. Protein losing enteropathy and alcoholic cirrhosis [author’s transl; in French]. Gastroenterol Clin Biol. 1982;6:68–72.
179.
Okano S, Sugimoto M, Takase M, Iseki K, Kajihama A, Azuma H. Effectiveness of high-dose spironolactone therapy in a patient with recurrent protein-losing enteropathy after the Fontan procedure. Intern Med. 2016;55:1611–1614. doi: 10.2169/internalmedicine.55.6303
180.
Grattan MJ, McCrindle BW. Recurrent exacerbations of protein-losing enteropathy after initiation of growth hormone therapy in a Fontan patient controlled with spironolactone. Congenit Heart Dis. 2010;5:165–167. doi: 10.1111/j.1747-0803.2009.00320.x
181.
Johnson JN, Driscoll DJ, O’Leary PW. Protein-losing enteropathy and the Fontan operation. Nutr Clin Pract. 2012;27:375–384. doi: 10.1177/0884533612444532
182.
Windram JD, Clift PF, Speakman J, Thorne SA. An unusual treatment for protein losing enteropathy. Congenit Heart Dis. 2011;6:253–256. doi: 10.1111/j.1747-0803.2011.00484.x
183.
Zaupper LB, Nielsen BW, Herlin T. Protein-losing enteropathy after the total cavopulmonary connection: impact of intravenous immunoglobulin. Congenit Heart Dis. 2011;6:624–629. doi: 10.1111/j.1747-0803.2011.00568.x
184.
Thacker D, Patel A, Dodds K, Goldberg DJ, Semeao E, Rychik J. Use of oral budesonide in the management of protein-losing enteropathy after the Fontan operation. Ann Thorac Surg. 2010;89:837–842. doi: 10.1016/j.athoracsur.2009.09.063
185.
Schumacher KR, Cools M, Goldstein BH, Ioffe-Dahan V, King K, Gaffney D, Russell MW. Oral budesonide treatment for protein-losing enteropathy in Fontan-palliated patients. Pediatr Cardiol. 2011;32:966–971. doi: 10.1007/s00246-011-0029-2
186.
Uzun O, Wong JK, Bhole V, Stumper O. Resolution of protein-losing enteropathy and normalization of mesenteric Doppler flow with sildenafil after Fontan. Ann Thorac Surg. 2006;82:e39–e40. doi: 10.1016/j.athoracsur.2006.08.043
187.
Donnelly JP, Rosenthal A, Castle VP, Holmes RD. Reversal of protein-losing enteropathy with heparin therapy in three patients with univentricular hearts and Fontan palliation. J Pediatr. 1997;130:474–478.
188.
Ryerson L, Goldberg C, Rosenthal A, Armstrong A. Usefulness of heparin therapy in protein-losing enteropathy associated with single ventricle palliation. Am J Cardiol. 2008;101:248–251. doi: 10.1016/j.amjcard.2007.08.029
189.
Friedland-Little JM, Gajarski RJ, Schumacher KR. Dopamine as a potential rescue therapy for refractory protein-losing enteropathy in Fontan-palliated patients. Pediatr Transplant. 2017;21:12925. doi: 10.1111/petr.12925
190.
Rychik J, Rome JJ, Jacobs ML. Late surgical fenestration for complications after the Fontan operation. Circulation. 1997;96:33–36.
191.
António M, Gordo A, Pereira C, Pinto F, Fragata I, Fragata J. Thoracic duct decompression for protein-losing enteropathy in failing Fontan circulation. Ann Thorac Surg. 2016;101:2370–2373. doi: 10.1016/j.athoracsur.2015.08.079
192.
Schumacher KR, Gossett J, Guleserian K, Naftel DC, Pruitt E, Dodd D, Carboni M, Lamour J, Pophal S, Zamberlan M, Gajarski RJ. Fontan-associated protein-losing enteropathy and heart transplant: a Pediatric Heart Transplant Study analysis. J Heart Lung Transplant. 2015;34:1169–1176. doi: 10.1016/j.healun.2015.03.022
193.
Caruthers RL, Kempa M, Loo A, Gulbransen E, Kelly E, Erickson SR, Hirsch JC, Schumacher KR, Stringer KA. Demographic characteristics and estimated prevalence of Fontan-associated plastic bronchitis. Pediatr Cardiol. 2013;34:256–261. doi: 10.1007/s00246-012-0430-5
194.
Schumacher KR, Singh TP, Kuebler J, Aprile K, O'Brien M and Blume ED. Risk factors and outcome of Fontan-associated plastic bronchitis: a case-control study. J Am Heart Assoc. 2014;3:e000865. doi: 10.1161/JAHA.114.000865
195.
Ezmigna DR, Morgan WJ, Witte MH, Brown MA. Lymphoscintigraphy in plastic bronchitis, a pediatric case report. Pediatr Pulmonol. 2013;48:515–518. doi: 10.1002/ppul.22673
196.
Dori Y, Keller MS, Rychik J, Itkin M. Successful treatment of plastic bronchitis by selective lymphatic embolization in a Fontan patient. Pediatrics. 2014;134:e590–e595. doi: 10.1542/peds.2013-3723
197.
Dori Y, Keller MS, Rome JJ, Gillespie MJ, Glatz AC, Dodds K, Goldberg DJ, Goldfarb S, Rychik J, Itkin M. Percutaneous lymphatic embolization of abnormal pulmonary lymphatic flow as treatment of plastic bronchitis in patients with congenital heart disease. Circulation. 2016;133:1160–1170. doi: 10.1161/CIRCULATIONAHA.115.019710
198.
Racz J, Mane G, Ford M, Schmidt L, Myers J, Standiford TJ, Schumacher KR, Fifer C, Russell MW, Stringer KA. Immunophenotyping and protein profiling of Fontan-associated plastic bronchitis airway casts. Ann Am Thorac Soc. 2013;10:98–107. doi: 10.1513/AnnalsATS.201209-080OC
199.
Biko DM, DeWitt AG, Pinto EM, Morrison RE, Johnstone JA, Griffis H, O’Byrne ML, Fogel MA, Harris MA, Partington SL, Whitehead KK, Saul D, Goldberg DJ, Rychik J, Glatz AC, Gillespie MJ, Rome JJ, Dori Y. MRI evaluation of lymphatic abnormalities in the neck and thorax after Fontan surgery: relationship with outcome. Radiology. 2019;291:774–780. doi: 10.1148/radiol.2019180877edit
200.
Avitabile CM, Goldberg DJ, Dodds K, Dori Y, Ravishankar C, Rychik J. A multifaceted approach to the management of plastic bronchitis after cavopulmonary palliation. Ann Thorac Surg. 2014;98:634–640. doi: 10.1016/j.athoracsur.2014.04.015
201.
Brooks K, Caruthers RL, Schumacher KR, Stringer KA. Pharmacotherapy challenges of Fontan-associated plastic bronchitis: a rare pediatric disease. Pharmacotherapy. 2013;33:922–934. doi: 10.1002/phar.1290
202.
Onoue Y, Adachi Y, Ichida F, Miyawaki T. Effective use of corticosteroid in a child with life-threatening plastic bronchitis after Fontan operation. Pediatr Int. 2003;45:107–109.
203.
Costello JM, Steinhorn D, McColley S, Gerber ME, Kumar SP. Treatment of plastic bronchitis in a Fontan patient with tissue plasminogen activator: a case report and review of the literature. Pediatrics. 2002;109:e67.
204.
Heath L, Ling S, Racz J, Mane G, Schmidt L, Myers JL, Tsai WC, Caruthers RL, Hirsch JC, Stringer KA. Prospective, longitudinal study of plastic bronchitis cast pathology and responsiveness to tissue plasminogen activator. Pediatr Cardiol. 2011;32:1182–1189. doi: 10.1007/s00246-011-0058-x
205.
Parikh K, Witte MH, Samson R, Teodori M, Carpenter JB, Lowe MC, Morgan W, Hardin C, Brown M, Naughton Y, Sinha S, Barber BJ. Successful treatment of plastic bronchitis with low fat diet and subsequent thoracic duct ligation in child with Fontan physiology. Lymphology. 2012;45:47–52.
206.
Jasinovic T, Kozak FK, Moxham JP, Chilvers M, Wensley D, Seear M, Campbell A, Ludemann JP. Casting a look at pediatric plastic bronchitis. Int J Pediatr Otorhinolaryngol. 2015;79:1658–1661. doi: 10.1016/j.ijporl.2015.07.011
207.
Gossett JG, Almond CS, Kirk R, Zangwill S, Richmond ME, Kantor PF, Tresler MA, Lenderman SM, Naftel DC, Matthews KL, Pahl E. Outcomes of cardiac transplantation in single-ventricle patients with plastic bronchitis: a multicenter study. J Am Coll Cardiol. 2013;61:985–986. doi: 10.1016/j.jacc.2012.10.042
208.
Kiernan F. The anatomy and physiology of the liver. Philosophical Trans Royal Coll London. 1833;123:711–770.
209.
Arcidi JM, Moore GW, Hutchins GM. Hepatic morphology in cardiac dysfunction: a clinicopathologic study of 1000 subjects at autopsy. Am J Pathol. 1981;104:159–166.
210.
Ghaferi AA, Hutchins GM. Progression of liver pathology in patients undergoing the Fontan procedure: chronic passive congestion, cardiac cirrhosis, hepatic adenoma, and hepatocellular carcinoma. J Thorac Cardiovasc Surg. 2005;129:1348–1352. doi: 10.1016/j.jtcvs.2004.10.005
211.
Agnoletti G, Ferraro G, Bordese R, Marini D, Gala S, Bergamasco L, Ferroni F, Calvo PL, Barletti C, Cisarò F, Longo F, Pace Napoleone C. Fontan circulation causes early, severe liver damage: should we offer patients a tailored strategy? Int J Cardiol. 2016;209:60–65. doi: 10.1016/j.ijcard.2016.02.041
212.
Goldberg DJ, Surrey LF, Glatz AC, Dodds K, O'Byrne ML, Lin HC, Fogel M, Rome JJ, Rand EB, Russo P, Rychik J. Hepatic fibrosis is universal following Fontan operation, and severity is associated with time from surgery: a liver biopsy and hemodynamic study. J Am Heart Assoc. 2017;6:e004809. doi: 10.1161/JAHA.116.004809
213.
Pundi K, Pundi KN, Kamath PS, Cetta F, Li Z, Poterucha JT, Driscoll DJ, Johnson JN. Liver disease in patients after the Fontan operation. Am J Cardiol. 2016;117:456–460. doi: 10.1016/j.amjcard.2015.11.014
214.
Navaratnam D, Fitzsimmons S, Grocott M, Rossiter HB, Emmanuel Y, Diller GP, Gordon-Walker T, Jack S, Sheron N, Pappachan J, Pratap JN, Vettukattil JJ, Veldtman G. Exercise-induced systemic venous hypertension in the Fontan circulation. Am J Cardiol. 2016;117:1667–1671. doi: 10.1016/j.amjcard.2016.02.042
215.
Masutani S, Kurishima C, Yana A, Kuwata S, Iwamoto Y, Saiki H, Ishido H, Senzaki H. Assessment of central venous physiology of Fontan circulation using peripheral venous pressure. J Thorac Cardiovasc Surg. 2017;153:912–920. doi: 10.1016/j.jtcvs.2016.11.061
216.
DiPaola FW, Schumacher KR, Goldberg CS, Friedland-Little J, Parameswaran A, Dillman JR. Effect of Fontan operation on liver stiffness in children with single ventricle physiology. Eur Radiol. 2017;27:2434–2442. doi: 10.1007/s00330-016-4614-x
217.
Lautt WW, Legare DJ, d’Almeida MS. Adenosine as putative regulator of hepatic arterial flow (the buffer response). Am J Physiol. 1985;248(pt 2):H331–H338. doi: 10.1152/ajpheart.1985.248.3.H331
218.
Lautt WW, Greenway CV. Hepatic venous compliance and role of liver as a blood reservoir. Am J Physiol. 1976;231:292–295.
219.
Bryant T, Ahmad Z, Millward-Sadler H, Burney K, Stedman B, Kendall T, Vettukattil J, Haw M, Salmon AP, Cope R, Hacking N, Breen D, Sheron N, Veldtman GR. Arterialised hepatic nodules in the Fontan circulation: hepatico-cardiac interactions. Int J Cardiol. 2011;151:268–272. doi: 10.1016/j.ijcard.2010.05.047
220.
Trusty PM, Wei Z, Rychik J, Russo PA, Surrey LF, Goldberg DJ, Fogel MA, Yoganathan AP. Impact of hemodynamics and fluid energetics on liver fibrosis after Fontan operation. J Thorac Cardiovasc Surg. 2018;156:267–275. doi: 10.1016/j.jtcvs.2018.02.078
221.
Daniels CJ, Bradley EA, Landzberg MJ, Aboulhosn J, Beekman RH, Book W, Gurvitz M, John A, John B, Marelli A, Marino BS, Minich LL, Poterucha JJ, Rand EB, Veldtman GR. Fontan-associated liver disease: proceedings from the American College of Cardiology Stakeholders Meeting, October 1 to 2, 2015, Washington DC. J Am Coll Cardiol. 2017;70:3173–3194.
222.
van Nieuwenhuizen RC, Peters M, Lubbers LJ, Trip MD, Tijssen JG, Mulder BJ. Abnormalities in liver function and coagulation profile following the Fontan procedure. Heart. 1999;82:40–46. doi: 10.1136/hrt.82.1.40
223.
Ndrepepa G, Kastrati A. Gamma-glutamyl transferase and cardiovascular disease. Ann Transl Med. 2016;4:481. doi: 10.21037/atm.2016.12.27
224.
Nandwana SB, Olaiya B, Cox K, Sahu A, Mittal P. Abdominal imaging surveillance in adult patients after Fontan procedure: risk of chronic liver disease and hepatocellular carcinoma. Curr Probl Diagn Radiol. 2018;47:19–22. doi: 10.1067/j.cpradiol.2017.04.002
225.
Wu FM, Kogon B, Earing MG, Aboulhosn JA, Broberg CS, John AS, Harmon A, Sainani NI, Hill AJ, Odze RD, Johncilla ME, Ukomadu C, Gauvreau K, Valente AM, Landzberg MJ; Alliance for Adult Research in Congenital Cardiology (AARCC) Investigators. Liver health in adults with Fontan circulation: a multicenter cross-sectional study. J Thorac Cardiovasc Surg. 2017;153:656–664. doi: 10.1016/j.jtcvs.2016.10.060
226.
Chen B, Schreiber RA, Human DG, Potts JE, Guttman OR. Assessment of liver stiffness in pediatric Fontan patients using transient elastography. Can J Gastroenterol Hepatol. 2016;2016:7125193. doi: 10.1155/2016/7125193
227.
Egbe A, Miranda WR, Connolly HM, Khan AR, Al-Otaibi M, Venkatesh SK, Simonetto D, Kamath P, Warnes C. Temporal changes in liver stiffness after Fontan operation: results of serial magnetic resonance elastography. Int J Cardiol. 2018;258:299–304. doi: 10.1016/j.ijcard.2018.01.108
228.
Mueller S, Sandrin L. Liver stiffness: a novel parameter for the diagnosis of liver disease. Hepat Med. 2010;2:49–67.
229.
Poterucha JT, Johnson JN, Qureshi MY, O’Leary PW, Kamath PS, Lennon RJ, Bonnichsen CR, Young PM, Venkatesh SK, Ehman RL, Gupta S, Smyrk TC, Dearani JA, Warnes CA, Cetta F. Magnetic resonance elastography: a novel technique for the detection of hepatic fibrosis and hepatocellular carcinoma after the Fontan operation. Mayo Clin Proc. 2015;90:882–894. doi: 10.1016/j.mayocp.2015.04.020
230.
Millonig G, Friedrich S, Adolf S, Fonouni H, Golriz M, Mehrabi A, Stiefel P, Pöschl G, Büchler MW, Seitz HK, Mueller S. Liver stiffness is directly influenced by central venous pressure. J Hepatol. 2010;52:206–210. doi: 10.1016/j.jhep.2009.11.018
231.
Hu JJ, Qureshi MY, Urban MW, Graham R, Yin M, Oommen S, Holst KA, Edgerton S, Vasconcelos L, Nenadic I, Cetta F; Wanek Program Pre-Clinical Pipeline. Ultrasound shear wave elastography as a measure of porcine hepatic disease in right heart dysfunction: a pilot study. Ultrasound Med Biol. 2018;44:2393–2399. doi: 10.1016/j.ultrasmedbio.2018.05.027
232.
Lewis MJ, Hecht E, Ginns J, Benton J, Prince M, Rosenbaum MS. Serial cardiac MRIs in adult Fontan patients detect progressive hepatic enlargement and congestion. Congenit Heart Dis. 2017;12:153–158. doi: 10.1111/chd.12422
233.
Hilscher MB, Johnson JN, Cetta F, Driscoll DJ, Poterucha JJ, Sanchez W, Connolly HM, Kamath PS. Surveillance for liver complications after the Fontan procedure. Congenit Heart Dis. 2017;12:124–132. doi: 10.1111/chd.12446
234.
Kolbe AB, Podberesky DJ, Zhang B, Towbin AJ. The impact of hepatocyte phase imaging from infancy to young adulthood in patients with a known or suspected liver lesion. Pediatr Radiol. 2015;45:354–365. doi: 10.1007/s00247-014-3160-4
235.
Surrey LF, Russo P, Rychik J, Goldberg DJ, Dodds K, O’Byrne ML, Glatz AC, Rand EB, Lin HC. Prevalence and characterization of fibrosis in surveillance liver biopsies of patients with Fontan circulation. Hum Pathol. 2016;57:106–115. doi: 10.1016/j.humpath.2016.07.006
236.
Gurvitz M, Ionescu-Ittu R, Guo L, Eisenberg MJ, Abrahamowicz M, Pilote L, Marelli AJ. Prevalence of cancer in adults with congenital heart disease compared with the general population. Am J Cardiol. 2016;118:1742–1750. doi: 10.1016/j.amjcard.2016.08.057
237.
Egbe AC, Poterucha JT, Warnes CA, Connolly HM, Baskar S, Ginde S, Clift P, Kogon B, Book WM, Walker N, Wagenaar L, Moe T, Oechslin E, Kay WA, Norris M, Gordon-Walker T, Dillman JR, Trout A, Anwar N, Hoskoppal A, Veldtman GR. Hepatocellular carcinoma after Fontan operation. Circulation. 2018;138:746–748. doi: 10.1161/CIRCULATIONAHA.117.032717
238.
Josephus Jitta D, Wagenaar LJ, Mulder BJ, Guichelaar M, Bouman D, van Melle JP. Three cases of hepatocellular carcinoma in Fontan patients: review of the literature and suggestions for hepatic screening. Int J Cardiol. 2016;206:21–26. doi: 10.1016/j.ijcard.2015.12.033
239.
Velpula M, Sheron N, Guha N, Salmon T, Hacking N, Veldtman GR. Direct measurement of porto-systemic gradient in a failing Fontan circulation. Congenit Heart Dis. 2011;6:175–178. doi: 10.1111/j.1747-0803.2010.00451.x
240.
Elder RW, McCabe NM, Hebson C, Veledar E, Romero R, Ford RM, Mahle WT, Kogon BE, Sahu A, Jokhadar M, McConnell ME, Book WM. Features of portal hypertension are associated with major adverse events in Fontan patients: the VAST study. Int J Cardiol. 2013;168:3764–3769. doi: 10.1016/j.ijcard.2013.06.008
241.
Assenza GE, Graham DA, Landzberg MJ, Valente AM, Singh MN, Bashir A, Fernandes S, Mortele KJ, Ukomadu C, Volpe M, Wu F. MELD-XI score and cardiac mortality or transplantation in patients after Fontan surgery. Heart. 2013;99:491–496. doi: 10.1136/heartjnl-2012-303347
242.
Camposilvan S, Milanesi O, Stellin G, Pettenazzo A, Zancan L, D’Antiga L. Liver and cardiac function in the long term after Fontan operation. Ann Thorac Surg. 2008;86:177–182. doi: 10.1016/j.athoracsur.2008.03.077
243.
Rutigliano I, Vinci R, De Filippo G, Mancini M, Stoppino L, d’Apolito M, Giardino I, Macarini L, Pettoello Mantovani M, Campanozzi A. Metabolic syndrome, hepatic steatosis, and cardiovascular risk in children. Nutrition. 2017;36:1–7. doi: 10.1016/j.nut.2016.10.017
244.
Dimopoulos K, Diller GP, Koltsida E, Pijuan-Domenech A, Papadopoulou SA, Babu-Narayan SV, Salukhe TV, Piepoli MF, Poole-Wilson PA, Best N, Francis DP, Gatzoulis MA. Prevalence, predictors, and prognostic value of renal dysfunction in adults with congenital heart disease. Circulation. 2008;117:2320–2328. doi: 10.1161/CIRCULATIONAHA.107.734921
245.
Gupte PA, Vaideeswar P, Kandalkar BM. Cyanotic nephropathy–a morphometric analysis. Congenit Heart Dis. 2014;9:280–285. doi: 10.1111/chd.12121
246.
Esch JJ, Salvin JM, Thiagarajan RR, Del Nido PJ, Rajagopal SK. Acute kidney injury after Fontan completion: risk factors and outcomes. J Thorac Cardiovasc Surg. 2015;150:190–197. doi: 10.1016/j.jtcvs.2015.04.011
247.
Taylor ML, Carmona F, Thiagarajan RR, Westgate L, Ferguson MA, del Nido PJ, Rajagopal SK. Mild postoperative acute kidney injury and outcomes after surgery for congenital heart disease. J Thorac Cardiovasc Surg. 2013;146:146–152. doi: 10.1016/j.jtcvs.2012.09.008
248.
Blinder JJ, Goldstein SL, Lee VV, Baycroft A, Fraser CD, Nelson D, Jefferies JL. Congenital heart surgery in infants: effects of acute kidney injury on outcomes. J Thorac Cardiovasc Surg. 2012;143:368–374. doi: 10.1016/j.jtcvs.2011.06.021
249.
Ricci Z, Di Nardo M, Iacoella C, Netto R, Picca S, Cogo P. Pediatric RIFLE for acute kidney injury diagnosis and prognosis for children undergoing cardiac surgery: a single-center prospective observational study. Pediatr Cardiol. 2013;34:1404–1408. doi: 10.1007/s00246-013-0662-z
250.
Sammour F, Haw M, Paisey J, Cope R, Herbertson M, Salmon T, Vettukattil J, Rogerson M, Karagkiozaki V, Veldtman G. Renal function of patients with a failing Fontan circuit undergoing total cavopulmonary revision surgery. Pediatr Cardiol. 2009;30:282–288. doi: 10.1007/s00246-008-9344-7
251.
Algaze CA, Koth AM, Faberowski LW, Hanley FL, Krawczeski CD, Axelrod DM. Acute kidney injury in patients undergoing the extracardiac Fontan operation with and without the use of cardiopulmonary bypass. Pediatr Crit Care Med. 2017;18:34–43. doi: 10.1097/PCC.0000000000000984
252.
Lee D, Levin A, Kiess M, Sexsmith G, Chakrabarti S, Barlow A, Human D, Grewal J. Chronic kidney damage in the adult Fontan population. Int J Cardiol. 2018;257:62–66. doi: 10.1016/j.ijcard.2017.11.118
253.
Sharma S, Ruebner RL, Furth SL, Dodds KM, Rychik J, Goldberg DJ. Assessment of kidney function in survivors following Fontan palliation. Congenit Heart Dis. 2016;11:630–636. doi: 10.1111/chd.12358
254.
Anne P, Du W, Mattoo TK, Zilberman MV. Nephropathy in patients after Fontan palliation. Int J Cardiol. 2009;132:244–247. doi: 10.1016/j.ijcard.2007.11.079
255.
Mori M, Aguirre AJ, Elder RW, Kashkouli A, Farris AB, Ford RM, Book WM. Beyond a broken heart: circulatory dysfunction in the failing Fontan. Pediatr Cardiol. 2014;35:569–579. doi: 10.1007/s00246-014-0881-y
256.
Opotowsky AR, Baraona FR, Mc Causland FR, Loukas B, Landzberg E, Landzberg MJ, Sabbisetti V, Waikar SS. Estimated glomerular filtration rate and urine biomarkers in patients with single-ventricle Fontan circulation. Heart. 2017;103:434–442. doi: 10.1136/heartjnl-2016-309729
257.
Blok IM, van Riel AC, Schuuring MJ, de Bruin-Bon RH, van Dijk AP, Hoendermis ES, Zwinderman AH, Mulder BJ, Bouma BJ. The role of cystatin C as a biomarker for prognosis in pulmonary arterial hypertension due to congenital heart disease. Int J Cardiol. 2016;209:242–247. doi: 10.1016/j.ijcard.2016.02.003
258.
Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation: Modification of Diet in Renal Disease Study Group. Ann Intern Med. 1999;130:461–470.
259.
Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16:31–41. doi: 10.1159/000180580
260.
Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF, Feldman HI, Kusek JW, Eggers P, Van Lente F, Greene T, Coresh J; CKD-EPI (Chronic Kidney Disease Epidemiology Collaboration). A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150:604–612.
261.
Zhu Y, Ye X, Zhu B, Pei X, Wei L, Wu J, Zhao W. Comparisons between the 2012 new CKD-EPI (Chronic Kidney Disease Epidemiology Collaboration) equations and other four approved equations. PLoS One. 2014;9:e84688. doi: 10.1371/journal.pone.0084688
262.
Burke JR, Glasgow EF, McCredie DA, Powell HR. Nephropathy in cyanotic congenital heart disease. Clin Nephrol. 1977;7:38–42.
263.
Perloff JK, Latta H, Barsotti P. Pathogenesis of the glomerular abnormality in cyanotic congenital heart disease. Am J Cardiol. 2000;86:1198–1204.
264.
Wilson TG, d’Udekem Y, Winlaw DS, Cordina RL, Celermajer DS, Wheaton GR, Bullock A, Gentles TL, Weintraub RG, Justo RN, Grigg LE, Radford DJ, Hardikar W, Cheung M, Cain TM, Rao P, Alexander SI, Ayer J, Verrall C, Du Plessis K, Chapman J, Rice K, Barry J, Zannino D, Iyengar AJ; Australian and New Zealand Fontan Registry. Hepatic and renal end-organ damage in the Fontan circulation: a report from the Australian and New Zealand Fontan Registry. Int J Cardiol. 2018;273:100–107. doi: 10.1016/j.ijcard.2018.07.118
265.
Ohuchi H, Ikado H, Noritake K, Miyazaki A, Yasuda K, Yamada O. Impact of central venous pressure on cardiorenal interactions in adult patients with congenital heart disease after biventricular repair. Congenit Heart Dis. 2013;8:103–110. doi: 10.1111/j.1747-0803.2012.00717.x
266.
Ohuchi H, Negishi J, Hayama Y, Miyazaki A, Shiraishi I, Ichikawa H. Renal resistive index reflects Fontan pathophysiology and predicts mortality. Heart. 2017;103:1631–1637. doi: 10.1136/heartjnl-2016-310812
267.
Rajpal S, Alshawabkeh L, Opotowsky AR. Current role of blood and urine biomarkers in the clinical care of adults with congenital heart disease. Curr Cardiol Rep. 2017;19:50. doi: 10.1007/s11886-017-0860-z
268.
Cohen MS, Zak V, Atz AM, Printz BF, Pinto N, Lambert L, Pemberton V, Li JS, Margossian R, Dunbar-Masterson C, McCrindle BW. Anthropometric measures after Fontan procedure: implications for suboptimal functional outcome. Am Heart J. 2010;160:1092–1098.e1. doi: 10.1016/j.ahj.2010.07.039
269.
Avitabile CM, Goldberg DJ, Zemel BS, Brodsky JL, Dodds K, Hayden-Rush C, Whitehead KK, Goldmuntz E, Rychik J, Leonard MB. Deficits in bone density and structure in children and young adults following Fontan palliation. Bone. 2015;77:12–16. doi: 10.1016/j.bone.2015.04.012
270.
Avitabile CM, Leonard MB, Zemel BS, Brodsky JL, Lee D, Dodds K, Hayden-Rush C, Whitehead KK, Goldmuntz E, Paridon SM, Rychik J, Goldberg DJ. Lean mass deficits, vitamin D status and exercise capacity in children and young adults after Fontan palliation. Heart. 2014;100:1702–1707. doi: 10.1136/heartjnl-2014-305723
271.
Avitabile CM, Leonard MB, Brodsky JL, Whitehead KK, Ravishankar C, Cohen MS, Gaynor JW, Rychik J, Goldberg DJ. Usefulness of insulin like growth factor 1 as a marker of heart failure in children and young adults after the Fontan palliation procedure. Am J Cardiol. 2015;115:816–820. doi: 10.1016/j.amjcard.2014.12.041
272.
Cordina R, O’Meagher S, Gould H, Rae C, Kemp G, Pasco JA, Celermajer DS, Singh N. Skeletal muscle abnormalities and exercise capacity in adults with a Fontan circulation. Heart. 2013;99:1530–1534. doi: 10.1136/heartjnl-2013-304249
273.
Holler F, Hannes T, Germund I, Emmel M, Hoyer-Kuhn H, Khalil M, Sreeram N, Udink Ten Cate FE. Low serum 25-hydroxyvitamin D levels and secondary hyperparathyroidism in Fontan patients. Cardiol Young. 2016;26:876–884. doi: 10.1017/S1047951115001444
274.
Wetzsteon RJ, Zemel BS, Shults J, Howard KM, Kibe LW, Leonard MB. Mechanical loads and cortical bone geometry in healthy children and young adults. Bone. 2011;48:1103–1108. doi: 10.1016/j.bone.2011.01.005
275.
Leonard MB, Elmi A, Mostoufi-Moab S, Shults J, Burnham JM, Thayu M, Kibe L, Wetzsteon RJ, Zemel BS. Effects of sex, race, and puberty on cortical bone and the functional muscle bone unit in children, adolescents, and young adults. J Clin Endocrinol Metab. 2010;95:1681–1689. doi: 10.1210/jc.2009-1913
276.
Liu D, Manske SL, Kontulainen SA, Tang C, Guy P, Oxland TR, McKay HA. Tibial geometry is associated with failure load ex vivo: a MRI, pQCT and DXA study. Osteoporos Int. 2007;18:991–997. doi: 10.1007/s00198-007-0325-0
277.
Vogt KN, Manlhiot C, Van Arsdell G, Russell JL, Mital S, McCrindle BW. Somatic growth in children with single ventricle physiology impact of physiologic state. J Am Coll Cardiol. 2007;50:1876–1883. doi: 10.1016/j.jacc.2007.07.050
278.
Cohen MI, Bush DM, Ferry RJ, Spray TL, Moshang T, Wernovsky G, Vetter VL. Somatic growth failure after the Fontan operation. Cardiol Young. 2000;10:447–457.
279.
Mancilla EE, Zielonka B, Brodsky J, Roizen J, Zemel B, Goldberg DJ, Dodds KM, Rychik J. Growth parameters, bone mineral density and secondary hyperparathyroidism after Fontan operation: results from the Multidisciplinary Clinic at the CHOP Single Ventricle Survivorship Program. Circulation. 2016;134:A19714. Abstract 19714.
280.
Morsy MS, Dishmon DA, Garg N, Weber KT. Secondary hyperparathyroidism in heart failure. Am J Med Sci. 2017;354:335–338. doi: 10.1016/j.amjms.2017.02.008
281.
Freud LR, Webster G, Costello JM, Tsao S, Rychlik K, Backer CL, Deal BJ. Growth and obesity among older single ventricle patients presenting for Fontan conversion. World J Pediatr Congenit Heart Surg. 2015;6:514–520. doi: 10.1177/2150135115598212
282.
Chung ST, Hong B, Patterson L, Petit CJ, Ham JN. High Overweight and Obesity in Fontan Patients: A 20-Year History. Pediatr Cardiol. 2016;37:192–200. doi: 10.1007/s00246-015-1265-7
283.
Pinto NM, Marino BS, Wernovsky G, de Ferranti SD, Walsh AZ, Laronde M, Hyland K, Dunn SO, Cohen MS. Obesity is a common comorbidity in children with congenital and acquired heart disease. Pediatrics. 2007;120:e1157–e1164. doi: 10.1542/peds.2007-0306
284.
Wellnitz K, Harris IS, Sapru A, Fineman JR, Radman M. Longitudinal development of obesity in the post-Fontan population. Eur J Clin Nutr. 2015;69:1105–1108. doi: 10.1038/ejcn.2015.68
285.
Lavie CJ, Messerli FH. Cardiovascular adaptation to obesity and hypertension. Chest. 1986;90:275–279.
286.
Lavie CJ, Pandey A, Lau DH, Alpert MA, Sanders P. Obesity and atrial fibrillation prevalence, pathogenesis, and prognosis: effects of weight loss and exercise. J Am Coll Cardiol. 2017;70:2022–2035. doi: 10.1016/j.jacc.2017.09.002
287.
Lui GK, Saidi A, Bhatt AB, Burchill LJ, Deen JF, Earing MG, Gewitz M, Ginns J, Kay JD, Kim YY, Kovacs AH, Krieger EV, Wu FM, Yoo SJ; on behalf of the American Heart Association Adult Congenital Heart Disease Committee of the Council on Clinical Cardiology and Council on Cardiovascular Disease in the Young; Council on Cardiovascular Radiology and Intervention; and Council on Quality of Care and Outcomes Research. Diagnosis and management of noncardiac complications in adults with congenital heart disease: a scientific statement from the American Heart Association. Circulation. 2017;136:e348–e392. doi: 10.1161/CIR.0000000000000535
288.
Berger NA. Young adult cancer: influence of the obesity pandemic. Obesity (Silver Spring). 2018;26:641–650. doi: 10.1002/oby.22137
289.
Steele CB, Thomas CC, Henley SJ, Massetti GM, Galuska DA, Agurs-Collins T, Puckett M, Richardson LC. Vital signs: trends in incidence of cancers associated with overweight and obesity–United States, 2005-2014. MMWR Morb Mortal Wkly Rep. 2017;66:1052–1058. doi: 10.15585/mmwr.mm6639e1
290.
Massetti GM, Dietz WH, Richardson LC. Excessive weight gain, obesity, and cancer: opportunities for clinical intervention. JAMA. 2017;318:1975–1976. doi: 10.1001/jama.2017.15519
291.
Loomba R, Yang HI, Su J, Brenner D, Barrett-Connor E, Iloeje U, Chen CJ. Synergism between obesity and alcohol in increasing the risk of hepatocellular carcinoma: a prospective cohort study. Am J Epidemiol. 2013;177:333–342. doi: 10.1093/aje/kws252
292.
Khan SS, Ning H, Wilkins JT, Allen N, Carnethon M, Berry JD, Sweis RN, Lloyd-Jones DM. Association of body mass index with lifetime risk of cardiovascular disease and compression of morbidity. JAMA Cardiol. 2018;3:280–287. doi: 10.1001/jamacardio.2018.0022
293.
GBD 2015 Obesity Collaborators, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, Lee A, Marczak L, Mokdad AH, Moradi-Lakeh M, Naghavi M, Salama JS, Vos T, Abate KH, Abbafati C, Ahmed MB, Al-Aly Z, Alkerwi A, Al-Raddadi R, Amare AT, Amberbir A, Amegah AK, Amini E, Amrock SM, Anjana RM, Ärnlöv J, Asayesh H, Banerjee A, Barac A, Baye E, Bennett DA, Beyene AS, Biadgilign S, Biryukov S, Bjertness E, Boneya DJ, Campos-Nonato I, Carrero JJ, Cecilio P, Cercy K, Ciobanu LG, Cornaby L, Damtew SA, Dandona L, Dandona R, Dharmaratne SD, Duncan BB, Eshrati B, Esteghamati A, Feigin VL, Fernandes JC, Fürst T, Gebrehiwot TT, Gold A, Gona PN, Goto A, Habtewold TD, Hadush KT, Hafezi-Nejad N, Hay SI, Horino M, Islami F, Kamal R, Kasaeian A, Katikireddi SV, Kengne AP, Kesavachandran CN, Khader YS, Khang YH, Khubchandani J, Kim D, Kim YJ, Kinfu Y, Kosen S, Ku T, Defo BK, Kumar GA, Larson HJ, Leinsalu M, Liang X, Lim SS, Liu P, Lopez AD, Lozano R, Majeed A, Malekzadeh R, Malta DC, Mazidi M, McAlinden C, McGarvey ST, Mengistu DT, Mensah GA, Mensink GBM, Mezgebe HB, Mirrakhimov EM, Mueller UO, Noubiap JJ, Obermeyer CM, Ogbo FA, Owolabi MO, Patton GC, Pourmalek F, Qorbani M, Rafay A, Rai RK, Ranabhat CL, Reinig N, Safiri S, Salomon JA, Sanabria JR, Santos IS, Sartorius B, Sawhney M, Schmidhuber J, Schutte AE, Schmidt MI, Sepanlou SG, Shamsizadeh M, Sheikhbahaei S, Shin MJ, Shiri R, Shiue I, Roba HS, Silva DAS, Silverberg JI, Singh JA, Stranges S, Swaminathan S, Tabarés-Seisdedos R, Tadese F, Tedla BA, Tegegne BS, Terkawi AS, Thakur JS, Tonelli M, Topor-Madry R, Tyrovolas S, Ukwaja KN, Uthman OA, Vaezghasemi M, Vasankari T, Vlassov VV, Vollset SE, Weiderpass E, Werdecker A, Wesana J, Westerman R, Yano Y, Yonemoto N, Yonga G, Zaidi Z, Zenebe ZM, Zipkin B, Murray CJL. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377:13–27.
294.
Marino BS, Lipkin PH, Newburger JW, Peacock G, Gerdes M, Gaynor JW, Mussatto KA, Uzark K, Goldberg CS, Johnson WH, Li J, Smith SE, Bellinger DC, Mahle WT; on behalf of the American Heart Association Congenital Heart Defects Committee, Council on Cardiovascular Disease in the Young, Council on Cardiovascular Nursing, and Stroke Council. Neurodevelopmental outcomes in children with congenital heart disease: evaluation and management: a scientific statement from the American Heart Association. Circulation. 2012;126:1143–1172. doi: 10.1161/CIR.0b013e318265ee8a
295.
Gaynor JW, Stopp C, Wypij D, Andropoulos DB, Atallah J, Atz AM, Beca J, Donofrio MT, Duncan K, Ghanayem NS, Goldberg CS, Hövels-Gürich H, Ichida F, Jacobs JP, Justo R, Latal B, Li JS, Mahle WT, McQuillen PS, Menon SC, Pemberton VL, Pike NA, Pizarro C, Shekerdemian LS, Synnes A, Williams I, Bellinger DC, Newburger JW; International Cardiac Collaborative on Neurodevelopment (ICCON) Investigators. Neurodevelopmental outcomes after cardiac surgery in infancy. Pediatrics. 2015;135:816–825. doi: 10.1542/peds.2014-3825
296.
International Cardiac Collaborative on Neurodevelopment (ICCON) Investigators. Impact of operative and postoperative factors on neurodevelopmental outcomes after cardiac operations. Ann Thorac Surg. 2016;102:843–849.
297.
Marelli A, Miller SP, Marino BS, Jefferson AL, Newburger JW. Brain in congenital heart disease across the lifespan: the cumulative burden of injury. Circulation. 2016;133:1951–1962. doi: 10.1161/CIRCULATIONAHA.115.019881
298.
Calderon J, Stopp C, Wypij D, DeMaso DR, Rivkin M, Newburger JW, Bellinger DC. Early-term birth in single-ventricle congenital heart disease after the Fontan procedure: neurodevelopmental and psychiatric outcomes. J Pediatr. 2016;179:96–103. doi: 10.1016/j.jpeds.2016.08.084
299.
Gaynor JW, Ittenbach RF, Gerdes M, Bernbaum J, Clancy RR, McDonald-McGinn DM, Zackai EH, Wernovsky G, Nicolson SC, Spray TL. Neurodevelopmental outcomes in preschool survivors of the Fontan procedure. J Thorac Cardiovasc Surg. 2014;147:1276–1282. doi: 10.1016/j.jtcvs.2013.12.019
300.
Bean Jaworski JL, White MT, DeMaso DR, Newburger JW, Bellinger DC, Cassidy AR. Visuospatial processing in adolescents with critical congenital heart disease: organization, integration, and implications for academic achievement. Child Neuropsychol. 2018;24:451–468. doi: 10.1080/09297049.2017.1283396
301.
Ravishankar C, Zak V, Williams IA, Bellinger DC, Gaynor JW, Ghanayem NS, Krawczeski CD, Licht DJ, Mahony L, Newburger JW, Pemberton VL, Williams RV, Sananes R, Cook AL, Atz T, Khaikin S, Hsu DT; Pediatric Heart Network Investigators. Association of impaired linear growth and worse neurodevelopmental outcome in infants with single ventricle physiology: a report from the Pediatric Heart Network Infant Single Ventricle Trial. J Pediatr. 2013;162:250–6.e2. doi: 10.1016/j.jpeds.2012.07.048
302.
Newburger JW, Sleeper LA, Bellinger DC, Goldberg CS, Tabbutt S, Lu M, Mussatto KA, Williams IA, Gustafson KE, Mital S, Pike N, Sood E, Mahle WT, Cooper DS, Dunbar-Masterson C, Krawczeski CD, Lewis A, Menon SC, Pemberton VL, Ravishankar C, Atz TW, Ohye RG, Gaynor JW; for the Pediatric Heart Network Investigators. Early developmental outcome in children with hypoplastic left heart syndrome and related anomalies: the Single Ventricle Reconstruction Trial. Circulation. 2012;125:2081–2091. doi: 10.1161/CIRCULATIONAHA.111.064113
303.
Mahle WT, Lu M, Ohye RG, William Gaynor J, Goldberg CS, Sleeper LA, Pemberton VL, Mussatto KA, Williams IA, Sood E, Krawczeski CD, Lewis A, Mirarchi N, Scheurer M, Pasquali SK, Pinto N, Jacobs JP, McCrindle BW, Newburger JW. A predictive model for neurodevelopmental outcome after the Norwood procedure. Pediatr Cardiol. 2013;34:327–333. doi: 10.1007/s00246-012-0450-1
304.
Homsy J, Zaidi S, Shen Y, Ware JS, Samocha KE, Karczewski KJ, DePalma SR, McKean D, Wakimoto H, Gorham J, Jin SC, Deanfield J, Giardini A, Porter GA, Kim R, Bilguvar K, López-Giráldez F, Tikhonova I, Mane S, Romano-Adesman A, Qi H, Vardarajan B, Ma L, Daly M, Roberts AE, Russell MW, Mital S, Newburger JW, Gaynor JW, Breitbart RE, Iossifov I, Ronemus M, Sanders SJ, Kaltman JR, Seidman JG, Brueckner M, Gelb BD, Goldmuntz E, Lifton RP, Seidman CE, Chung WK. De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science. 2015;350:1262–1266. doi: 10.1126/science.aac9396
305.
Limperopoulos C, Tworetzky W, McElhinney DB, Newburger JW, Brown DW, Robertson RL, Guizard N, McGrath E, Geva J, Annese D, Dunbar-Masterson C, Trainor B, Laussen PC, du Plessis AJ. Brain volume and metabolism in fetuses with congenital heart disease: evaluation with quantitative magnetic resonance imaging and spectroscopy. Circulation. 2010;121:26–33. doi: 10.1161/CIRCULATIONAHA.109.865568
306.
Hinton RB, Andelfinger G, Sekar P, Hinton AC, Gendron RL, Michelfelder EC, Robitaille Y, Benson DW. Prenatal head growth and white matter injury in hypoplastic left heart syndrome. Pediatr Res. 2008;64:364–369. doi: 10.1203/PDR.0b013e3181827bf4
307.
Clouchoux C, du Plessis AJ, Bouyssi-Kobar M, Tworetzky W, McElhinney DB, Brown DW, Gholipour A, Kudelski D, Warfield SK, McCarter RJ, Robertson RL, Evans AC, Newburger JW, Limperopoulos C. Delayed cortical development in fetuses with complex congenital heart disease. Cereb Cortex. 2013;23:2932–2943. doi: 10.1093/cercor/bhs281
308.
Sun L, Macgowan CK, Sled JG, Yoo SJ, Manlhiot C, Porayette P, Grosse-Wortmann L, Jaeggi E, McCrindle BW, Kingdom J, Hickey E, Miller S, Seed M. Reduced fetal cerebral oxygen consumption is associated with smaller brain size in fetuses with congenital heart disease. Circulation. 2015;131:1313–1323. doi: 10.1161/CIRCULATIONAHA.114.013051
309.
Lim JM, Kingdom T, Saini B, Chau V, Post M, Blaser S, Macgowan C, Miller SP, Seed M. Cerebral oxygen delivery is reduced in newborns with congenital heart disease. J Thorac Cardiovasc Surg. 2016;152:1095–1103. doi: 10.1016/j.jtcvs.2016.05.027
310.
Licht DJ, Shera DM, Clancy RR, Wernovsky G, Montenegro LM, Nicolson SC, Zimmerman RA, Spray TL, Gaynor JW, Vossough A. Brain maturation is delayed in infants with complex congenital heart defects. J Thorac Cardiovasc Surg. 2009;137:529–536. doi: 10.1016/j.jtcvs.2008.10.025
311.
Morton PD, Ishibashi N, Jonas RA. Neurodevelopmental abnormalities and congenital heart disease: insights into altered brain maturation. Circ Res. 2017;120:960–977. doi: 10.1161/CIRCRESAHA.116.309048
312.
Ohye RG, Sleeper LA, Mahony L, Newburger JW, Pearson GD, Lu M, Goldberg CS, Tabbutt S, Frommelt PC, Ghanayem NS, Laussen PC, Rhodes JF, Lewis AB, Mital S, Ravishankar C, Williams IA, Dunbar-Masterson C, Atz AM, Colan S, Minich LL, Pizarro C, Kanter KR, Jaggers J, Jacobs JP, Krawczeski CD, Pike N, McCrindle BW, Virzi L, Gaynor JW; Pediatric Heart Network Investigators. Comparison of shunt types in the Norwood procedure for single-ventricle lesions. N Engl J Med. 2010;362:1980–1992. doi: 10.1056/NEJMoa0912461
313.
Newburger JW, Silbert AR, Buckley LP, Fyler DC. Cognitive function and age at repair of transposition of the great arteries in children. N Engl J Med. 1984;310:1495–1499. doi: 10.1056/NEJM198406073102303
314.
Burch PT, Ravishankar C, Newburger JW, Lambert LM, Pemberton VL, Granger S, Floh AA, Anderson JB, Hill GD, Hill KD, Oster ME, Lewis AB, Schumacher KR, Zyblewski SC, Davies RR, Jacobs JP, Lai WW, Minich LL; Pediatric Heart Network Investigators. Assessment of growth 6 years after the Norwood procedure. J Pediatr. 2017;180:270–274.e6. doi: 10.1016/j.jpeds.2016.09.048
315.
Odegard KC, Zurakowski D, DiNardo JA, Castro RA, McGowan FX, Neufeld EJ, Laussen PC. Prospective longitudinal study of coagulation profiles in children with hypoplastic left heart syndrome from stage I through Fontan completion. J Thorac Cardiovasc Surg. 2009;137:934–941. doi: 10.1016/j.jtcvs.2008.09.031
316.
Giglia TM, Massicotte MP, Tweddell JS, Barst RJ, Bauman M, Erickson CC, Feltes TF, Foster E, Hinoki K, Ichord RN, Kreutzer J, McCrindle BW, Newburger JW, Tabbutt S, Todd JL, Webb CL; on behalf of the American Heart Association Congenital Heart Defects Committee of the Council on Cardiovascular Disease in the Young, Council on Cardiovascular and Stroke Nursing, Council on Epidemiology and Prevention, and Stroke Council. Prevention and treatment of thrombosis in pediatric and congenital heart disease: a scientific statement from the American Heart Association [published correction appears in Circulation. 2014;129:e23]. Circulation. 2013;128:2622–2703. doi: 10.1161/01.cir.0000436140.77832.7a
317.
Goldberg CS, Schwartz EM, Brunberg JA, Mosca RS, Bove EL, Schork MA, Stetz SP, Cheatham JP, Kulik TJ. Neurodevelopmental outcome of patients after the Fontan operation: a comparison between children with hypoplastic left heart syndrome and other functional single ventricle lesions. J Pediatr. 2000;137:646–652. doi: 10.1067/mpd.2000.108952
318.
Sarajuuri A, Jokinen E, Mildh L, Tujulin AM, Mattila I, Valanne L, Lönnqvist T. Neurodevelopmental burden at age 5 years in patients with univentricular heart. Pediatrics. 2012;130:e1636–e1646. doi: 10.1542/peds.2012-0486
319.
Sarajuuri A, Jokinen E, Puosi R, Eronen M, Mildh L, Mattila I, Valanne L, Lönnqvist T. Neurodevelopmental and neuroradiologic outcomes in patients with univentricular heart aged 5 to 7 years: related risk factor analysis. J Thorac Cardiovasc Surg. 2007;133:1524–1532. doi: 10.1016/j.jtcvs.2006.12.022
320.
Bellinger DC, Watson CG, Rivkin MJ, Robertson RL, Roberts AE, Stopp C, Dunbar-Masterson C, Bernson D, DeMaso DR, Wypij D, Newburger JW. Neuropsychological status and structural brain imaging in adolescents with single ventricle who underwent the Fontan procedure. J Am Heart Association. 2015;4:e002302. doi: 10.1161/JAHA.115.002302
321.
Watson CG, Stopp C, Wypij D, Newburger JW, Rivkin MJ. Reduced cortical volume and thickness and their relationship to medical and operative features in post-Fontan children and adolescents. Pediatr Res. 2017;81:881–890. doi: 10.1038/pr.2017.30
322.
du Plessis AJ, Chang AC, Wessel DL, Lock JE, Wernovsky G, Newburger JW, Mayer JE Cerebrovascular accidents following the Fontan operation. Pediatr Neurol. 1995;12:230–236.
323.
Fogel MA, Li C, Elci OU, Pawlowski T, Schwab PJ, Wilson F, Nicolson SC, Montenegro LM, Diaz L, Spray TL, Gaynor JW, Fuller S, Mascio C, Keller MS, Harris MA, Whitehead KK, Bethel J, Vossough A, Licht DJ. Neurological injury and cerebral blood flow in single ventricles throughout staged surgical reconstruction. Circulation. 2017;135:671–682. doi: 10.1161/CIRCULATIONAHA.116.021724
324.
Uzark K, Lincoln A, Lamberti JJ, Mainwaring RD, Spicer RL, Moore JW. Neurodevelopmental outcomes in children with Fontan repair of functional single ventricle. Pediatrics. 1998;101(pt 1):630–633.
325.
Wernovsky G, Stiles KM, Gauvreau K, Gentles TL, duPlessis AJ, Bellinger DC, Walsh AZ, Burnett J, Jonas RA, Mayer JE, Newburger JW. Cognitive development after the Fontan operation. Circulation. 2000;102:883–889.
326.
Mahle WT, Visconti KJ, Freier MC, Kanne SM, Hamilton WG, Sharkey AM, Chinnock RE, Jenkins KJ, Isquith PK, Burns TG, Jenkins PC. Relationship of surgical approach to neurodevelopmental outcomes in hypoplastic left heart syndrome. Pediatrics. 2006;117:e90–e97. doi: 10.1542/peds.2005-0575
327.
Rogers BT, Msall ME, Buck GM, Lyon NR, Norris MK, Roland JM, Gingell RL, Cleveland DC, Pieroni DR. Neurodevelopmental outcome of infants with hypoplastic left heart syndrome. J Pediatr. 1995;126:496–498.
328.
Kern JH, Hinton VJ, Nereo NE, Hayes CJ, Gersony WM. Early developmental outcome after the Norwood procedure for hypoplastic left heart syndrome. Pediatrics. 1998;102:1148–1152.
329.
Goldberg CS, Lu M, Sleeper LA, Mahle WT, Gaynor JW, Williams IA, Mussatto KA, Ohye RG, Graham EM, Frank DU, Jacobs JP, Krawczeski C, Lambert L, Lewis A, Pemberton VL, Sananes R, Sood E, Wechsler SB, Bellinger DC, Newburger JW; Pediatric Heart Network Investigators. Factors associated with neurodevelopment for children with single ventricle lesions. J Pediatr. 2014;165:490–496.e8. doi: 10.1016/j.jpeds.2014.
330.
Calderon J, Bellinger DC. Executive function deficits in congenital heart disease: why is intervention important? Cardiol Young. 2015;25:1238–1246. doi: 10.1017/S1047951115001134
331.
Cassidy AR, White MT, DeMaso DR, Newburger JW, Bellinger DC. Executive function in children and adolescents with critical cyanotic congenital heart disease. J Int Neuropsychol Soc. 2015;21:34–49. doi: 10.1017/S1355617714001027
332.
DeMaso DR, Calderon J, Taylor GA, Holland JE, Stopp C, White MT, Bellinger DC, Rivkin MJ, Wypij D, Newburger JW. Psychiatric disorders in adolescents with single ventricle congenital heart disease. Pediatrics. 2017;139:e20162241. doi: 10.1542/peds.2016-2241
333.
Moffitt TE, Arseneault L, Belsky D, Dickson N, Hancox RJ, Harrington H, Houts R, Poulton R, Roberts BW, Ross S, Sears MR, Thomson WM, Caspi A. A gradient of childhood self-control predicts health, wealth, and public safety. Proc Natl Acad Sci U S A. 2011;108:2693–2698. doi: 10.1073/pnas.1010076108
334.
Latal B, Helfricht S, Fischer JE, Bauersfeld U, Landolt MA. Psychological adjustment and quality of life in children and adolescents following open-heart surgery for congenital heart disease: a systematic review. BMC Pediatr. 2009;9:6. doi: 10.1186/1471-2431-9-6
335.
Jackson JL, Misiti B, Bridge JA, Daniels CJ, Vannatta K. Emotional functioning of adolescents and adults with congenital heart disease: a meta-analysis. Congenit Heart Dis. 2015;10:2–12. doi: 10.1111/chd.12178
336.
Horner T, Liberthson R, Jellinek MS. Psychosocial profile of adults with complex congenital heart disease. Mayo Clin Proc. 2000;75:31–36. doi: 10.4065/75.1.31
337.
Kovacs AH, Saidi AS, Kuhl EA, Sears SF, Silversides C, Harrison JL, Ong L, Colman J, Oechslin E, Nolan RP. Depression and anxiety in adult congenital heart disease: predictors and prevalence. Int J Cardiol. 2009;137:158–164. doi: 10.1016/j.ijcard.2008.06.042
338.
Bromberg JI, Beasley PJ, D’Angelo EJ, Landzberg M, DeMaso DR. Depression and anxiety in adults with congenital heart disease: a pilot study. Heart Lung. 2003;32:105–110. doi: 10.1067/mhl.2003.26
339.
Ferguson M, Kovacs AH. An integrated adult congenital heart disease psychology service. Congenit Heart Dis. 2016;11:444–451. doi: 10.1111/chd.12331
340.
Deng LX, Khan AM, Drajpuch D, Fuller S, Ludmir J, Mascio CE, Partington SL, Qadeer A, Tobin L, Kovacs AH, Kim YY. Prevalence and correlates of post-traumatic stress disorder in adults with congenital heart disease. Am J Cardiol. 2016;117:853–857. doi: 10.1016/j.amjcard.2015.11.065
341.
Eslami B. Correlates of posttraumatic stress disorder in adults with congenital heart disease. Congenit Heart Dis. 2017;12:357–363. doi: 10.1111/chd.12452
342.
Callus E, Utens EM, Quadri E, Ricci C, Carminati M, Giamberti A, Chessa M. The impact of actual and perceived disease severity on pre-operative psychological well-being and illness behaviour in adult congenital heart disease patients. Cardiol Young. 2014;24:275–282. doi: 10.1017/S1047951113000218
343.
van Rijen EH, Utens EM, Roos-Hesselink JW, Meijboom FJ, van Domburg RT, Roelandt JR, Bogers AJ, Verhulst FC. Medical predictors for psychopathology in adults with operated congenital heart disease. Eur Heart J. 2004;25:1605–1613. doi: 10.1016/j.ehj.2004.06.025
344.
Popelová J, Slavík Z, Skovránek J. Are cyanosed adults with congenital cardiac malformations depressed? Cardiol Young. 2001;11:379–384.
345.
Rose M, Köhler K, Köhler F, Sawitzky B, Fliege H, Klapp BF. Determinants of the quality of life of patients with congenital heart disease. Qual Life Res. 2005;14:35–43.
346.
Pike NA, Evangelista LS, Doering LV, Eastwood JA, Lewis AB, Child JS. Quality of life, health status, and depression: comparison between adolescents and adults after the Fontan procedure with healthy counterparts. J Cardiovasc Nurs. 2012;27:539–546. doi: 10.1097/JCN.0b013e31822ce5f6
347.
Pike NA, Evangelista LS, Doering LV, Koniak-Griffin D, Lewis AB, Child JS. Clinical profile of the adolescent/adult Fontan survivor. Congenit Heart Dis. 2011;6:9–17. doi: 10.1111/j.1747-0803.2010.00475.x
348.
Bordin G, Padalino MA, Perentaler S, Castaldi B, Maschietto N, Michieli P, Crepaz R, Frigo AC, Vida VL, Milanesi O. Clinical profile and quality of life of adult patients after the Fontan procedure. Pediatr Cardiol. 2015;36:1261–1269. doi: 10.1007/s00246-015-1156-y
349.
Pike NA, Roy B, Gupta R, Singh S, Woo MA, Halnon NJ, Lewis AB, Kumar R. Brain abnormalities in cognition, anxiety, and depression regulatory regions in adolescents with single ventricle heart disease. J Neurosci Res. 2018;96:1104–1118. doi: 10.1002/jnr.24215
350.
Berghammer MC, Brink E, Rydberg AM, Dellborg M, Ekman I. Committed to life: adolescents’ and young adults’ experiences of living with Fontan circulation. Congenit Heart Dis. 2015;10:403–412. doi: 10.1111/chd.12244
351.
Gurvitz M, Burns KM, Brindis R, Broberg CS, Daniels CJ, Fuller SM, Honein MA, Khairy P, Kuehl KS, Landzberg MJ, Mahle WT, Mann DL, Marelli A, Newburger JW, Pearson GD, Starling RC, Tringali GR, Valente AM, Wu JC, Califf RM. Emerging research directions in adult congenital heart disease: a report from an NHLBI/ACHA working group. J Am Coll Cardiol. 2016;67:1956–1964. doi: 10.1016/j.jacc.2016.01.062
352.
Reid GJ, Webb GD, Barzel M, McCrindle BW, Irvine MJ, Siu SC. Estimates of life expectancy by adolescents and young adults with congenital heart disease. J Am Coll Cardiol. 2006;48:349–355. doi: 10.1016/j.jacc.2006.03.041
353.
Warnes CA, Williams RG, Bashore TM, Child JS, Connolly HM, Dearani JA, Del Nido P, Fasules JW, Graham TP, Hijazi ZM, Hunt SA, King ME, Landzberg MJ, Miner PD, Radford MJ, Walsh EP, Webb GD. ACC/AHA 2008 guidelines for the management of adults with congenital heart disease: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Adults With Congenital Heart Disease). Circulation. 2008;118:2395–2451. doi: 10.1161/CIRCULATIONAHA.108.190811
354.
Sable C, Foster E, Uzark K, Bjornsen K, Canobbio MM, Connolly HM, Graham TP, Gurvitz MZ, Kovacs A, Meadows AK, Reid GJ, Reiss JG, Rosenbaum KN, Sagerman PJ, Saidi A, Schonberg R, Shah S, Tong E, Williams RG; on behalf of the American Heart Association Congenital Heart Defects Committee of the Council on Cardiovascular Disease in the Young, Council on Cardiovascular Nursing, Council on Clinical Cardiology, and Council on Peripheral Vascular Disease. Best practices in managing transition to adulthood for adolescents with congenital heart disease: the transition process and medical and psychosocial issues: a scientific statement from the American Heart Association. Circulation. 2011;123:1454–1485. doi: 10.1161/CIR.0b013e3182107c56
355.
Tobler D, Greutmann M, Colman JM, Greutmann-Yantiri M, Librach SL, Kovacs AH. Knowledge of and preference for advance care planning by adults with congenital heart disease. Am J Cardiol. 2012;109:1797–1800. doi: 10.1016/j.amjcard.2012.02.027
356.
Tobler D, Greutmann M, Colman JM, Greutmann-Yantiri M, Librach LS, Kovacs AH. End-of-life in adults with congenital heart disease: a call for early communication. Int J Cardiol. 2012;155:383–387. doi: 10.1016/j.ijcard.2010.10.050
357.
Greutmann M, Tobler D, Colman JM, Greutmann-Yantiri M, Librach SL, Kovacs AH. Facilitators of and barriers to advance care planning in adult congenital heart disease. Congenit Heart Dis. 2013;8:281–288. doi: 10.1111/chd.12025
358.
Deng LX, Gleason LP, Khan AM, Drajpuch D, Fuller S, Goldberg LA, Mascio CE, Partington SL, Tobin L, Kim YY, Kovacs AH. Advance care planning in adults with congenital heart disease: a patient priority. Int J Cardiol. 2017;231:105–109. doi: 10.1016/j.ijcard.2016.12.185
359.
Tobler D, Greutmann M, Colman JM, Greutmann-Yantiri M, Librach LS, Kovacs AH. End-of-life care in hospitalized adults with complex congenital heart disease: care delayed, care denied. Palliat Med. 2012;26:72–79. doi: 10.1177/0269216311407694
360.
Kovacs AH, Landzberg MJ, Goodlin SJ. Advance care planning and end-of-life management of adult patients with congenital heart disease. World J Pediatr Congenit Heart Surg. 2013;4:62–69. doi: 10.1177/2150135112464235
361.
American Academy of Pediatrics. Committee on Bioethics and Committee on Hospital Care: palliative care for children. Pediatrics. 2000;106(pt 1):351–357.
362.
Mccabe MA, Rushton CH, Glover J, Murray MG, Leikin S. Implications of the Patient Self-Determination Act: guidelines for involving adolescents in medical decision making. J Adolesc Health. 1996;19:319–324. doi: 10.1016/S1054-139X(96)00160-7
363.
Diller GP, Kempny A, Alonso-Gonzalez R, Swan L, Uebing A, Li W, Babu-Narayan S, Wort SJ, Dimopoulos K, Gatzoulis MA. Survival prospects and circumstances of death in contemporary adult congenital heart disease patients under follow-up at a large tertiary centre. Circulation. 2015;132:2118–2125. doi: 10.1161/CIRCULATIONAHA.115.017202
364.
Dennis M, Zannino D, du Plessis K, Bullock A, Disney PJS, Radford DJ, Hornung T, Grigg L, Cordina R, d’Udekem Y, Celermajer DS. Clinical outcomes in adolescents and adults after the Fontan procedure. J Am Coll Cardiol. 2018;71:1009–1017. doi: 10.1016/j.jacc.2017.12.054
365.
Downing TE, Allen KY, Goldberg DJ, Rogers LS, Ravishankar C, Rychik J, Fuller S, Montenegro LM, Steven JM, Gillespie MJ, Rome JJ, Spray TL, Nicolson SC, Gaynor JW, Glatz AC. Surgical and catheter-based reinterventions are common in long-term survivors of the Fontan operation. Circ Cardiovasc Interv. 2017;10:e004924. doi: 10.1161/CIRCINTERVENTIONS.116.004924
366.
Egbe A, Khan AR, Al-Otaibi M, Said SM, Connolly HM. Outcomes of hospitalization in adults with Fontan palliation: the Mayo Clinic experience. Am Heart J. 2018;198:115–122. doi: 10.1016/j.ahj.2017.12.012
367.
Itatani K, Miyaji K, Tomoyasu T, Nakahata Y, Ohara K, Takamoto S, Ishii M. Optimal conduit size of the extracardiac Fontan operation based on energy loss and flow stagnation. Ann Thorac Surg. 2009;88:565–572. doi: 10.1016/j.athoracsur.2009.04.109
368.
Cho S, Kim WH, Choi ES, Kwak JG, Chang HW, Hyun K, Lee CH. Outcomes after extracardiac Fontan procedure with a 16-mm polytetrafluoroethylene conduit. Eur J Cardiothorac Surg. 2018;53:269–275. doi: 10.1093/ejcts/ezx238
369.
Udink Ten Cate FEA, Trieschmann U, Germund I, Hannes T, Emmel M, Bennink G, Sreeram N. Stenting the Fontan pathway in paediatric patients with obstructed extracardiac conduits. Heart. 2017;103:1111–1116. doi: 10.1136/heartjnl-2016-310511
370.
Egbe AC, Khan AR, Ammash NM, Barbara DW, Oliver WC, Said SM, Akintoye E, Warnes CA, Connolly HM. Predictors of procedural complications in adult Fontan patients undergoing non-cardiac procedures. Heart. 2017;103:1813–1820. doi: 10.1136/heartjnl-2016-311039
371.
Seckeler MD, Thomas ID, Andrews J, Meziab O, Moe T, Heller E, Klewer SE. Higher cost of hospitalizations for non-cardiac diagnoses in adults with congenital heart disease. Pediatr Cardiol. 2018;39:437–444. doi: 10.1007/s00246-017-1770-y
372.
Mylotte D, Pilote L, Ionescu-Ittu R, Abrahamowicz M, Khairy P, Therrien J, Mackie AS, Marelli A. Specialized adult congenital heart disease care: the impact of policy on mortality. Circulation. 2014;129:1804–1812. doi: 10.1161/CIRCULATIONAHA.113.005817
373.
Stout KK, Daniels CJ, Aboulhosn JA, Bozkurt B, Broberg CS, Colman JM, Crumb SR, Dearani JA, Fuller S, Gurvitz M, Khairy P, Landzberg MJ, Saidi A, Valente AM, Van Hare GF. 2018 AHA/ACC guideline for the management of adults with congenital heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines [published correction appears in Circulation. 2019;139:e833–e834]. Circulation. 2019;139:e698–e800. doi: 10.1161/CIR.0000000000000603
374.
Cauldwell M, Steer PJ, Bonner S, Asghar O, Swan L, Hodson K, Head CEG, Jakes AD, Walker N, Simpson M, Bolger AP, Siddiqui F, English KM, Maudlin L, Abraham D, Sands AJ, Mohan AR, Curtis SL, Coats L, Johnson MR. Retrospective UK multicentre study of the pregnancy outcomes of women with a Fontan repair. Heart. 2018;104:401–406. doi: 10.1136/heartjnl-2017-311763
375.
Canobbio MM, Mair DD, van der Velde M, Koos BJ. Pregnancy outcomes after the Fontan repair. J Am Coll Cardiol. 1996;28:763–767.
376.
Garcia Ropero A, Baskar S, Roos Hesselink JW, Girnius A, Zentner D, Swan L, Ladouceur M, Brown N, Veldtman GR. Pregnancy in women with a Fontan circulation: a systematic review of the literature. Circ Cardiovasc Qual Outcomes. 2018;11:e004575. doi: 10.1161/CIRCOUTCOMES.117.004575
377.
Cauldwell M, Von Klemperer K, Uebing A, Swan L, Steer PJ, Babu-Narayan SV, Gatzoulis MA, Johnson MR. A cohort study of women with a Fontan circulation undergoing preconception counselling. Heart. 2016;102:534–540. doi: 10.1136/heartjnl-2015-308788
378.
Drenthen W, Pieper PG, Roos-Hesselink JW, van Lottum WA, Voors AA, Mulder BJ, van Dijk AP, Vliegen HW, Sollie KM, Moons P, Ebels T, van Veldhuisen DJ; ZAHARA Investigators. Pregnancy and delivery in women after Fontan palliation. Heart. 2006;92:1290–1294. doi: 10.1136/hrt.2005.085407
379.
Di Maria MV, Brown DW, Cetta F, Ginde S, Goldberg D, Menon SC, Phelps HM, Rychik J, Schumacher KR, Thrush P, Veldtman G, Wright G, Younoszai AK. Surveillance testing and preventive care after Fontan operation: a multi-institutional survey. Pediatr Cardiol. 2019;40:110–115. doi: 10.1007/s00246-018-1966-9
380.
Chen CA, Chen SY, Chiu HH, Wang JK, Chang CI, Chiu IS, Chen YS, Lu CW, Lin MT, Lue HC, Hua YC, Wu MH. Prognostic value of submaximal exercise data for cardiac morbidity in Fontan patients. Med Sci Sports Exerc. 2014;46:10–15. doi: 10.1249/MSS.0b013e31829f8326
381.
Butts RJ, Spencer CT, Jackson L, Heal ME, Forbus G, Hulsey TC, Atz AM. Estimating equations for cardiopulmonary exercise testing variables in Fontan patients: derivation and validation using a multicenter cross-sectional database. Pediatr Cardiol. 2015;36:393–401. doi: 10.1007/s00246-014-1020-5
382.
Cordina RL, O’Meagher S, Karmali A, Rae CL, Liess C, Kemp GJ, Puranik R, Singh N, Celermajer DS. Resistance training improves cardiac output, exercise capacity and tolerance to positive airway pressure in Fontan physiology. Int J Cardiol. 2013;168:780–788. doi: 10.1016/j.ijcard.2012.10.012
383.
Laohachai K, Winlaw D, Selvadurai H, Gnanappa GK, d'Udekem Y, Celermajer D, Ayer J. Inspiratory muscle training is associated with improved inspiratory muscle strength, resting cardiac output, and the ventilatory efficiency of exercise in patients with a Fontan circulation. J Am Heart Assoc. 2017;6:e005750. doi: 10.1161/JAHA.117.005750
384.
Duppen N, Etnel JR, Spaans L, Takken T, van den Berg-Emons RJ, Boersma E, Schokking M, Dulfer K, Utens EM, Helbing W, Hopman MT. Does exercise training improve cardiopulmonary fitness and daily physical activity in children and young adults with corrected tetralogy of Fallot or Fontan circulation? A randomized controlled trial. Am Heart J. 2015;170:606–614. doi: 10.1016/j.ahj.2015.06.018
385.
Sutherland N, Jones B, d’Udekem Y. Should we recommend exercise after the Fontan procedure? Heart Lung Circ. 2015;24:753–768. doi: 10.1016/j.hlc.2015.03.005
386.
Anderson PA, Breitbart RE, McCrindle BW, Sleeper LA, Atz AM, Hsu DT, Lu M, Margossian R, Williams RV. The Fontan patient: inconsistencies in medication therapy across seven Pediatric Heart Network centers. Pediatr Cardiol. 2010;31:1219–1228. doi: 10.1007/s00246-010-9807-5
387.
Sleeper LA, Anderson P, Hsu DT, Mahony L, McCrindle BW, Roth SJ, Saul JP, Williams RV, Geva T, Colan SD, Clark BJ; Pediatric Heart Network Investigators. Design of a large cross-sectional study to facilitate future clinical trials in children with the Fontan palliation. Am Heart J. 2006;152:427–433. doi: 10.1016/j.ahj.2006.02.009
388.
McCrindle BW, Williams RV, Mitchell PD, Hsu DT, Paridon SM, Atz AM, Li JS, Newburger JW; for the Pediatric Heart Network Investigators. Relationship of patient and medical characteristics to health status in children and adolescents after the Fontan procedure. Circulation. 2006;113:1123–1129. doi: 10.1161/CIRCULATIONAHA.105.576660
389.
McCrindle BW, Zak V, Sleeper LA, Paridon SM, Colan SD, Geva T, Mahony L, Li JS, Breitbart RE, Margossian R, Williams RV, Gersony WM, Atz AM; for the Pediatric Heart Network Investigators. Laboratory measures of exercise capacity and ventricular characteristics and function are weakly associated with functional health status after Fontan procedure. Circulation. 2010;121:34–42. doi: 10.1161/CIRCULATIONAHA.109.869396
390.
Snarr BS, Paridon SM, Rychik J, Goldberg DJ. Pulmonary vasodilator therapy in the failing Fontan circulation: rationale and efficacy. Cardiol Young. 2015;25:1489–1492. doi: 10.1017/S1047951115002309
391.
Oldenburger NJ, Mank A, Etnel J, Takkenberg JJ, Helbing WA. Drug therapy in the prevention of failure of the Fontan circulation: a systematic review. Cardiol Young. 2016;26:842–850. doi: 10.1017/S1047951115002747
392.
Hebert A, Mikkelsen UR, Thilen U, Idorn L, Jensen AS, Nagy E, Hanseus K, Sørensen KE, Søndergaard L. Bosentan improves exercise capacity in adolescents and adults after Fontan operation: the TEMPO (Treatment With Endothelin Receptor Antagonist in Fontan Patients, a Randomized, Placebo-Controlled, Double-Blind Study Measuring Peak Oxygen Consumption) study. Circulation. 2014;130:2021–2030. doi: 10.1161/CIRCULATIONAHA.113.008441
393.
Goldberg DJ, Zak V, Goldstein BH, Chen S, Hamstra MS, Radojewski EA, Maunsell E, Mital S, Menon SC, Schumacher KR, Payne RM, Stylianou M, Kaltman JR, deVries TM, Yeager JL, Paridon SM; Pediatric Heart Network Investigators. Results of a phase I/II multi-center investigation of udenafil in adolescents after Fontan palliation. Am Heart J. 2017;188:42–52. doi: 10.1016/j.ahj.2017.02.030
394.
Manlhiot C, Brandão LR, Kwok J, Kegel S, Menjak IB, Carew CL, Chan AK, Schwartz SM, Sivarajan VB, Caldarone CA, Van Arsdell GS, McCrindle BW. Thrombotic complications and thromboprophylaxis across all three stages of single ventricle heart palliation. J Pediatr. 2012;161:513–519.e3. doi: 10.1016/j.jpeds.2012.03.004
395.
Marrone C, Galasso G, Piccolo R, de Leva F, Paladini R, Piscione F, Santoro G. Antiplatelet versus anticoagulation therapy after extracardiac conduit Fontan: a systematic review and meta-analysis. Pediatr Cardiol. 2011;32:32–39. doi: 10.1007/s00246-010-9808-4
396.
Monagle P, Cochrane A, Roberts R, Manlhiot C, Weintraub R, Szechtman B, Hughes M, Andrew M, McCrindle BW; Fontan Anticoagulation Study Group. A multicenter, randomized trial comparing heparin/warfarin and acetylsalicylic acid as primary thromboprophylaxis for 2 years after the Fontan procedure in children. J Am Coll Cardiol. 2011;58:645–651. doi: 10.1016/j.jacc.2011.01.061
397.
McCrindle BW, Manlhiot C, Cochrane A, Roberts R, Hughes M, Szechtman B, Weintraub R, Andrew M, Monagle P; Fontan Anticoagulation Study Group. Factors associated with thrombotic complications after the Fontan procedure: a secondary analysis of a multicenter, randomized trial of primary thromboprophylaxis for 2 years after the Fontan procedure. J Am Coll Cardiol. 2013;61:346–353. doi: 10.1016/j.jacc.2012.08.1023
398.
Pessotti CF, Jatene MB, Jatene IB, Oliveira PM, Succi FM, Moreira Vde M, Lopes RW, Pedra SR. Comparative trial of the use of antiplatelet and oral anticoagulant in thrombosis prophylaxis in patients undergoing total cavopulmonary operation with extracardiac conduit: echocardiographic, tomographic, scintigraphic, clinical and laboratory analysis. Rev Bras Cir Cardiovasc. 2014;29:595–605. doi: 10.5935/1678-9741.20140111
399.
McCrindle BW, Li JS, Manlhiot C, Tweddell JS, Giglia TM, Massicotte MP, Monagle P, Krishnamurthy R, Mahaffey KW, Michelson AD, Verdun N, Almond CS, Newburger JW, Brandão LR, Esmon CT, Manco-Johnson MJ, Ichord R, Ortel TL, Chan AK, Portman R, Rose M, Strony J, Kaltman JR. Challenges and priorities for research: a report from the National Heart, Lung, and Blood Institute (NHLBI)/National Institutes of Health (NIH) Working Group on thrombosis in pediatric cardiology and congenital heart disease. Circulation. 2014;130:1192–1203. doi: 10.1161/CIRCULATIONAHA.113.008428
400.
Prêtre R, Häussler A, Bettex D, Genoni M. Right-sided univentricular cardiac assistance in a failing Fontan circulation. Ann Thorac Surg. 2008;86:1018–1020. doi: 10.1016/j.athoracsur.2008.03.003
401.
Poh CL, Chiletti R, Zannino D, Brizard C, Konstantinov IE, Horton S, Millar J, d’Udekem Y. Ventricular assist device support in patients with single ventricles: the Melbourne experience. Interact Cardiovasc Thorac Surg. 2017;25:310–316. doi: 10.1093/icvts/ivx066
402.
Weinstein S, Bello R, Pizarro C, Fynn-Thompson F, Kirklin J, Guleserian K, Woods R, Tjossem C, Kroslowitz R, Friedmann P, Jaquiss R. The use of the Berlin Heart EXCOR in patients with functional single ventricle. J Thorac Cardiovasc Surg. 2014;147:697–704. doi: 10.1016/j.jtcvs.2013.10.030
403.
Arnaoutakis GJ, Blitzer D, Fuller S, Eckhauser AW, Montenegro LM, Rossano JW, Gaynor JW. Mechanical circulatory support as bridge to transplantation for the failing single ventricle. Ann Thorac Surg. 2017;103:193–197. doi: 10.1016/j.athoracsur.2016.05.015
404.
Rodefeld MD. Invited commentary. Ann Thorac Surg. 2017;103:197–198. doi: 10.1016/j.athoracsur.2016.06.050
405.
Jaquiss RD, Aziz H. Is four stage management the future of univentricular hearts? Destination therapy in the young. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2016;19:50–54. doi: 10.1053/j.pcsu.2015.12.004
406.
Vanderpluym CJ, Fynn-Thompson F, Blume ED. Ventricular assist devices in children: progress with an orphan device application. Circulation. 2014;129:1530–1537. doi: 10.1161/CIRCULATIONAHA.113.005574
407.
Adachi I, Burki S, Fraser CD Current status of pediatric ventricular assist device support. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2017;20:2–8. doi: 10.1053/j.pcsu.2016.09.010
408.
Horne D, Conway J, Rebeyka IM, Buchholz H. Mechanical circulatory support in univentricular hearts: current management. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2015;18:17–24. doi: 10.1053/j.pcsu.2015.02.002
409.
Giridharan GA, Ising M, Sobieski MA, Koenig SC, Chen J, Frankel S, Rodefeld MD. Cavopulmonary assist for the failing Fontan circulation: impact of ventricular function on mechanical support strategy. ASAIO J. 2014;60:707–715. doi: 10.1097/MAT.0000000000000135
410.
Rodefeld MD, Coats B, Fisher T, Giridharan GA, Chen J, Brown JW, Frankel SH. Cavopulmonary assist for the univentricular Fontan circulation: von Kármán viscous impeller pump. J Thorac Cardiovasc Surg. 2010;140:529–536. doi: 10.1016/j.jtcvs.2010.04.037
411.
Rodefeld MD, Frankel SH, Giridharan GA. Cavopulmonary assist: (em)powering the univentricular Fontan circulation. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2011;14:45–54. doi: 10.1053/j.pcsu.2011.01.015
412.
Rossano JW, Goldberg DJ, Fuller S, Ravishankar C, Montenegro LM, Gaynor JW. Successful use of the total artificial heart in the failing Fontan circulation. Ann Thorac Surg. 2014;97:1438–1440. doi: 10.1016/j.athoracsur.2013.06.120
413.
Ross HJ, Law Y, Book WM, Broberg CS, Burchill L, Cecchin F, Chen JM, Delgado D, Dimopoulos K, Everitt MD, Gatzoulis M, Harris L, Hsu DT, Kuvin JT, Martin CM, Murphy AM, Singh G, Spray TL, Stout KK; on behalf of the American Heart Association Adults With Congenital Heart Disease Committee of the Council on Clinical Cardiology and Council on Cardiovascular Disease in the Young, the Council on Cardiovascular Radiology and Intervention, and the Council on Functional Genomics and Translational Biology. Transplantation and mechanical circulatory support in congenital heart disease: a scientific statement from the American Heart Association. Circulation. 2016;133:802–820. doi: 10.1161/CIR.0000000000000353
414.
Schumacher KR, Almond C, Singh TP, Kirk R, Spicer R, Hoffman TM, Hsu D, Naftel DC, Pruitt E, Zamberlan M, Canter CE, Gajarski RJ; on behalf of the PHTS Study Group Investigators. Predicting graft loss by 1 year in pediatric heart transplantation candidates: an analysis of the Pediatric Heart Transplant Study database. Circulation. 2015;131:890–898. doi: 10.1161/CIRCULATIONAHA.114.009120
415.
Simpson KE, Pruitt E, Kirklin JK, Naftel DC, Singh RK, Edens RE, Barnes AP, Canter CE. Fontan patient survival after pediatric heart transplantation has improved in the current era. Ann Thorac Surg. 2017;103:1315–1320. doi: 10.1016/j.athoracsur.2016.08.110
416.
Dipchand AI, Edwards LB, Kucheryavaya AY, Benden C, Dobbels F, Levvey BJ, Lund LH, Meiser B, Yusen RD, Stehlik J; International Society of Heart and Lung Transplantation. The registry of the International Society for Heart and Lung Transplantation: seventeenth official pediatric heart transplantation report–2014; focus theme: retransplantation. J Heart Lung Transplant. 2014;33:985–995. doi: 10.1016/j.healun.2014.08.002
417.
Rogers JG. Changes in United States heart allocation: a community energized to improve policy. J Thorac Cardiovasc Surg. 2016;152:1484–1486. doi: 10.1016/j.jtcvs.2016.07.077
418.
Bernstein D, Naftel D, Chin C, Addonizio LJ, Gamberg P, Blume ED, Hsu D, Canter CE, Kirklin JK, Morrow WR, Pediatric Heart Transplant Study. Outcome of listing for cardiac transplantation for failed Fontan: a multi-institutional study. Circulation. 2006;114:273–280. doi: 10.1161/CIRCULATIONAHA.105.548016
419.
Murtuza B, Hermuzi A, Crossland DS, Parry G, Lord S, Hudson M, Chaudhari MP, Haynes S, O’Sullivan JJ, Hasan A. Impact of mode of failure and end-organ dysfunction on the survival of adult Fontan patients undergoing cardiac transplantation. Eur J Cardiothorac Surg. 2017;51:135–141. doi: 10.1093/ejcts/ezw243
420.
Deal BJ, Jacobs ML. Management of the failing Fontan circulation. Heart. 2012;98:1098–1104. doi: 10.1136/heartjnl-2011-301133
421.
Soukias N, Hornung TS, Kilner PJ, Frogoudaki A, Davlouros P, Wong T, Gatzoulis MA. Determinants of atrial arrhythmia late after the atriopulmonary Fontan operation. Hellenic J Cardiol. 2004;45:384–390.
422.
Lambert E, d’Udekem Y, Cheung M, Sari CI, Inman J, Ahimastos A, Eikelis N, Pathak A, King I, Grigg L, Schlaich M, Lambert G. Sympathetic and vascular dysfunction in adult patients with Fontan circulation. Int J Cardiol. 2013;167:1333–1338. doi: 10.1016/j.ijcard.2012.04.015
423.
Ohuchi H, Miyazaki A, Negishi J, Hayama Y, Nakai M, Nishimura K, Ichikawa H, Shiraishi I, Yamada O. Hemodynamic determinants of mortality after Fontan operation. Am Heart J. 2017;189:9–18. doi: 10.1016/j.ahj.2017.03.020
424.
Mori M, Shioda K, Elder RW, Pernetz MA, Rodriguez FH, Rangosch A, Kogon BE, Book WM. Superior mesenteric arterial flow pattern is associated with major adverse events in adults with Fontan circulation. Pediatr Cardiol. 2016;37:1013–1021. doi: 10.1007/s00246-016-1382-y
425.
Ghelani SJ, Harrild DM, Gauvreau K, Geva T, Rathod RH. Comparison between echocardiography and cardiac magnetic resonance imaging in predicting transplant-free survival after the Fontan operation. Am J Cardiol. 2015;116:1132–1138. doi: 10.1016/j.amjcard.2015.07.011
426.
Khambadkone S, Li J, de Leval MR, Cullen S, Deanfield JE, Redington AN. Basal pulmonary vascular resistance and nitric oxide responsiveness late after Fontan-type operation. Circulation. 2003;107:3204–3208. doi: 10.1161/01.CIR.0000074210.49434.40
427.
Cheung YF, Penny DJ, Redington AN. Serial assessment of left ventricular diastolic function after Fontan procedure. Heart. 2000;83:420–424. doi: 10.1136/heart.83.4.420
428.
Dori Y, Keller MS, Fogel MA, Rome JJ, Whitehead KK, Harris MA, Itkin M. MRI of lymphatic abnormalities after functional single-ventricle palliation surgery. AJR Am J Roentgenol. 2014;203:426–431. doi: 10.2214/AJR.13.11797
429.
Brizard CP, Lane GK, Alex G, Cheung MM. Original surgical procedure for the treatment of protein-losing enteropathy in Fontan patients: report of two midterm successes. Circulation. 2016;134:625–627. doi: 10.1161/CIRCULATIONAHA.116.023424
430.
Hraška V. Decompression of thoracic duct: new approach for the treatment of failing Fontan. Ann Thorac Surg. 2013;96:709–711. doi: 10.1016/j.athoracsur.2013.02.046
431.
Kogon B, McConnell M, Book W. Fontan conversion with hepatic vein exclusion: a means for hepatic preservation in single ventricle heart disease. Cardiol Young. 2016;26:582–585. doi: 10.1017/S104795111500133X
432.
du Plessis K, Peters R, King I, Robertson K, Mackley J, Maree R, Stanley T, Pickford L, Rose B, Orchard M, Stewart H, d’Udekem Y. “Will she live a long happy life?” Parents’ concerns for their children with Fontan circulation. Int J Cardiol Heart Vasc. 2018;18:65–70. doi: 10.1016/j.ijcha.2018.02.008
433.
Cordina R, du Plessis K, Tran D, d’Udekem Y. Super-Fontan: is it possible? J Thorac Cardiovasc Surg. 2018;155:1192–1194. doi: 10.1016/j.jtcvs.2017.10.047
434.
Rayner M, Muscara F, Dimovski A, McCarthy MC, Yamada J, Anderson VA, Burke K, Walser R, Nicholson JM. Take a Breath: study protocol for a randomized controlled trial of an online group intervention to reduce traumatic stress in parents of children with a life threatening illness or injury. BMC Psychiatry. 2016;16:169. doi: 10.1186/s12888-016-0861-2
435.
Goldberg DJ, Zak V, Goldstein BH, McCrindle BW, Menon SC, Schumacher KR, Payne RM, Rhodes J, McHugh KE, Penny DJ, Trachtenberg F, Hamstra MS, Richmond ME, Frommelt PC, Files MD, Yeager JL, Pemberton VL, Stylianou MP, Pearson GD, Paridon SM; Pediatric Heart Network Investigators. Design and rationale of the Fontan Udenafil Exercise Longitudinal (FUEL) trial. Am Heart J. 2018;201:1–8. doi: 10.1016/j.ahj.2018.03.015
436.
Uzark K, Zak V, Shrader P, McCrindle BW, Radojewski E, Varni JW, Daniels K, Handisides J, Hill KD, Lambert LM, Margossian R, Pemberton VL, Lai WW, Atz AM; Pediatric Heart Network Investigators. Assessment of quality of life in young patients with single ventricle after the Fontan operation. J Pediatr. 2016;170:166–72.e1. doi: 10.1016/j.jpeds.2015.11.016
437.
d’Udekem Y, Cheung MM, Setyapranata S, Iyengar AJ, Kelly P, Buckland N, Grigg LE, Weintraub RG, Vance A, Brizard CP, Penny DJ. How good is a good Fontan? Quality of life and exercise capacity of Fontans without arrhythmias. Ann Thorac Surg. 2009;88:1961–1969. doi: 10.1016/j.athoracsur.2009.07.079
438.
Saliba Z, Butera G, Bonnet D, Bonhoeffer P, Villain E, Kachaner J, Sidi D, Iserin L. Quality of life and perceived health status in surviving adults with univentricular heart. Heart. 2001;86:69–73. doi: 10.1136/heart.86.1.69
Information & Authors
Information
Published In
Copyright
© 2019 American Heart Association, Inc.
Versions
You are viewing the most recent version of this article.
History
Published online: 1 July 2019
Published in print: 6 August 2019
Keywords
Subjects
Authors
Metrics & Citations
Metrics
Citations
Download Citations
If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.
- Isolated loss of vaccine immunity in the protein losings syndrome in a patient with a reverse one and a half ventricle palliation (“failing Fontan-like physiology”), Cardiology in the Young, (1-3), (2025).https://doi.org/10.1017/S1047951124036369
- The Future of the Fontan Circulation, JACC: Case Reports, 30, 1, (103031), (2025).https://doi.org/10.1016/j.jaccas.2024.103031
- The fontan circulation, Nadas' Pediatric Cardiology, (709-718), (2025).https://doi.org/10.1016/B978-1-4557-0599-3.00068-5
- Pediatric devices, Mechanical Circulatory and Respiratory Support, (373-417), (2025).https://doi.org/10.1016/B978-0-443-22148-4.00013-7
- Congestive hepatopathy, Treatment and Management of Tropical Liver Disease, (205-209), (2025).https://doi.org/10.1016/B978-0-323-87031-3.00034-2
- Mid-term results and late events after the Fontan operation: A single-center experience, Turkish Journal of Thoracic and Cardiovascular Surgery, 32, 3, (280-290), (2024).https://doi.org/10.5606/tgkdc.dergisi.2024.25793
- Brazilian Guideline for Exercise Testing in Children and Adolescents – 2024, Arquivos Brasileiros de Cardiologia, 121, 8, (2024).https://doi.org/10.36660/abc.20240525i
- Diretriz Brasileira de Ergometria em Crianças e Adolescentes – 2024, Arquivos Brasileiros de Cardiologia, 121, 8, (2024).https://doi.org/10.36660/abc.20240525
- Cardiac Rehabilitation for Fontan Circulation Patients: A Systematic Review, and Meta-Analysis, Medicina, 60, 11, (1817), (2024).https://doi.org/10.3390/medicina60111817
- The Effect of Cardiac Rehabilitation in Paediatric Fontan Circulation Patients: A Prospective Intervention Study, Medicina, 60, 10, (1566), (2024).https://doi.org/10.3390/medicina60101566
- See more
Loading...
View Options
Login options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginPurchase Options
Purchase this article to access the full text.
eLetters(0)
eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.
Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.