575
views
0
recommends
+1 Recommend
1 collections
    2
    shares

      CVIA now indexed by SCOPUS from February 2024. CVIA received its first Journal Impact Factor (0.5) in the 2023 Journal Citation Reports Release. 

      Interested in becoming a CVIA published author?

      • Platinum Open Access with no APCs. 
      • Fast peer review/Fast publication online after article acceptance.

      Submissions should be made electronically at: https://mc04.manuscriptcentral.com/cvia-journal.

      Please refer to the Author Guidelines at https://cvia-journal.org/instructions-to-authors/ before submission.

       

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The Fontan Circulation: Contemporary Review of Ongoing Challenges and Management Strategies

      review-article
      , MD 1 , , MD 1 , , MD, PhD 2 ,
      Cardiovascular Innovations and Applications
      Compuscript
      Fontan, single ventricle
      Bookmark

            Abstract

            Since its original description, the Fontan operation has been widely used for the palliation of children with single ventricle physiology and has resulted in an increasing number of these patients surviving to adulthood. The Fontan operation is a unique approach to create a circulation in series without two distinct pumping chambers. Although the Fontan operation increased the survival rates of patients with single ventricle physiology, it carries an inevitable risk of long-term morbidities that impacts the outcomes and quality of life in these patients. In this review, we discuss the challenges resulting from the unique pathophysiology of Fontan circulation and propose management strategies.

            Main article text

            Introduction

            The Fontan operation, first described by Fontan and Baudet [1] in 1971, was introduced as a palliative procedure for patients with tricuspid atresia. It later evolved to become the palliative surgery for other forms of congenital heart disease (CHD) with single ventricle physiology in which biventricular repair is not feasible. Since the original description, there have been multiple modifications of the surgical technique [2, 3]. The procedure creates nonpulsatile pulmonary flow via direct connection of the venae cavae to the pulmonary arteries and results in an immediate elevation in central venous pressure. The unique physiological consequences of the Fontan circulation not only impact the cardiovascular system but also impose challenges at a multisystem level. In this review, we provide a summary of the Fontan surgeries and resultant physiology, discuss long-term complications, and provide a contemporary review of the management strategies.

            Evolution of the Fontan Operation

            The Fontan procedure attempts to circumvent the need for a subpulmonary ventricle by redirecting systemic venous return from the superior vena cava and inferior vena cava directly to the pulmonary arteries. The initial concept, when Fontan palliation was first described, was to use the right atrium as a pumping chamber. The surgical technique included a classic Glenn anastomosis with a valved homograft between the right atrial appendage and the main pulmonary artery, trying to use the right atrium as a pump to the pulmonary arteries, which led to the classic Fontan procedure, or the atriopulmonary connection (APC; see Figure 1A). This procedure leads to substantial adverse hemodynamic consequences, including massive right atrial dilatation and resultant complex atrial arrhythmias and thromboembolic events.

            Figure 1

            Variations of the Fontan Operation.

            (A) Modified classic right atrium to pulmonary artery Fontan operation. (B) Lateral tunnel Fontan oparation. (C) Extracardiac conduit (with extracardiac pacemaker leads). (From Khairy et al. [4]).

            Subsequently, multiple modifications to the surgical technique have been proposed to improve the flow dynamics of the Fontan circuit. In 1987, de Leval et al. [2] described the creation of an intra-atrial channel to direct the inferior vena cava flow to the pulmonary artery called the intracardiac lateral tunnel (ILT, Figure 1B). Soon thereafter, in 1990, Marcelletti et al. [3] described the use of extracardiac conduit directing the inferior vena cava flow to the pulmonary arteries called the extracardiac conduit modification (Figure 1C). In the current era the latter two modifications are the standard surgical techniques used in most surgical centers. Another modification of the Fontan procedure was the creation of a fenestration to permit a controlled right-to-left atrial shunt so as to maintain cardiac output in high-risk patients, particularly during phasic elevation of pulmonary vascular resistance (PVR) [5]. This technique has been used to reduce the complications of low postoperative cardiac output, pleural and pericardial effusions, ascites, ventricular dysfunction, diminished exercise performance, and protein-losing enteropathy (PLE) and to improve postoperative survival [6 ,7]. In the long term, patients are at increased risk of systemic embolization and systemic desaturation. Careful assessment should be performed before fenestration closure is undertaken [6, 8, 9]. The practice of creating a fenestration initially differs at different centers.

            Hemodynamic Principles of the Fontan Circulation and Its Consequences

            The hemodynamic principle of the Fontan operation is to separate the systemic circulation from the pulmonary circulation by connecting the systemic venous circulation directly to the pulmonary circulation. In the absence of a pumping chamber to the pulmonary circulation, the pulmonary flow relies on the differential pressure gradient between the central systemic veins and left atrial pressure, along with low PVR [10]. The systemic ventricle acts as a suction pump during diastole, adding to the forward driving force, emphasizing the importance of ventricular diastolic function in the Fontan circulation [11]. Spontaneous respiration also plays a role through the negative intrathoracic pressure generated during inspiration; therefore positive pressure ventilation is usually poorly tolerated in Fontan patients.

            The main hemodynamic sequelae of the Fontan operation are systemic venous hypertension and chronic low cardiac output. The underlying cause of low cardiac output is multifactorial, and it can occur despite normal single ventricular dysfunction. The causes include (1) elevation in systemic venous pressures, (2) passive pulmonary blood flow reliant on low pulmonary vascular resistance, which in the absence of a subpulmonary ventricle limits the preload reserve and the ability of the single ventricle to augment cardiac output in response to higher metabolic demands, and (3) the inability to augment heart rate in many patients because of underlying sinus node dysfunction [10, 12].

            This chronic state of low cardiac output leads to hepatic venous hypertension, elevation of pressures in the thoracic duct with a potential risk of prolonged chylous effusion in the early post-Fontan period, and development of plastic bronchitis and PLE at long-term follow-up [13, 14].

            Cardiac Performance During Exercise

            Exercise performance in patients with a Fontan circulation is decreased because of inadequate augmentation of cardiac output. This is due to a variety of factors, including blunted heart rate response from sinus node dysfunction, lack of a subpulmonary pumping chamber, abnormal pulmonary vasculature adaptation to exercise, and inadequate preload reserve.

            Cardiopulmonary exercise testing provides valuable information as a predictor for various adverse events, with peak oxygen uptake (Vo2max) reported as an independent predictor for death and morbidity [1518]. Ohuchi et al. [17] reported a cutoff value of 21 mL/kg/min for Vo2max (corresponding to 49% of the predicted value for age) as a strong predictor for mortality. Recent studies have suggested that serial cardiopulmonary exercise testing could add significant value in routine evaluation of exercise tolerance in Fontan patients as well as in risk stratification for clinical outcomes [19].

            Long-term Complications and Management Strategies

            With advances in surgical, catheter, and medical therapies, the survival rates of patients undergoing the Fontan operation have increased dramatically over time. As many patients are now surviving to adulthood, there is growing interest in studying the long-term functional status and prevalence of late morbidities specific to the Fontan circulation. In a systemic review by Alsaied et al. [20] of 6700 Fontan patients with an average follow-up of 8 years, the most common cardiac cause of long-term death was heart failure, including Fontan failure (22% of late deaths), followed by arrhythmia/sudden death (16%), and the most common noncardiac causes were respiratory failure (15%) and renal diseases (12%). The rate of freedom from death or transplant has been reported to be 90% at 10 years, 83% at 20 years, and 70% at 25 years [21], and the 30-year survival rate has been reported as 43% [22]. Some reported predictors of adverse outcomes include impaired preoperative ventricular function, pulmonary artery pressure greater than 15 mmHg, atrioventricular valve (AVV) regurgitation before Fontan completion, AVV replacement at the time of the Fontan procedure, heterotaxy syndrome, pacemaker implantation, including in patients in nonsinus rhythm before Fontan palliation, older age at the time of the operation (>7 years), hypoplastic left-sided heart syndrome, and APC Fontan procedure [2227].

            Cardiovascular-Related Complications

            Arrhythmia/Sudden Death

            Development of early and late arrhythmias remains a challenging and extremely common complication after the Fontan operation. The overall incidence of postoperative arrhythmias has been reported to be 48% at an average follow-up of 15.3±9.3 years, while the 20- and 30-year rates of freedom from new-onset arrhythmia are 42% and 24%, respectively [22]. The incidence is much higher in APC Fontan patients, with atrial arrhythmias occurring in to 50–60% of these patients [28, 29]. The newer modifications of ILT and extracardiac conduit Fontan modifications were introduced with the hope to reduce the arrhythmia burden by improving the flow dynamics in the Fontan circuit, and on long-term follow-up the incidence was found to be lower with these newer approaches as compared with the APC Fontan procedure [30, 31]. However, there is likely a similar arrhythmia burden when these newer modifications are compared with each other [30]. Reported risk factors for increased arrhythmia in Fontan patients include severe branch pulmonary artery stenosis, systemic ventricular end-diastolic pressure greater than 12 mmHg preoperatively, AVV replacement during the Fontan operation, and early postoperative arrhythmia [32].

            Intra-atrial reentrant tachycardia is the most common type of supraventricular tachycardia, accounting for 75% of supraventricular tachycardia in these patients [33]. Focal atrial tachycardia can also occur in some patients, and atrial fibrillation incidence is increasing as the population ages [34, 35]. Patients with single ventricle physiology may present with symptoms of decompensated heart failure, especially those with elevated Fontan pressures and poor ventricular function. Sinus node dysfunction is also commonly seen in patients following the Fontan operation as injury to the sinus node can frequently occur during the surgery [30]. Epicardial pacemaker therapy will often be needed, although there is potential for a transvenous atrial lead in APC or ILT Fontan patients. Ventricular arrhythmias account for 5% of the arrhythmias that occur in Fontan patients, and can be seen in patients with poor ventricular function, likely accounting for the incidence of sudden cardiac death [34]. Risk factors for sudden cardiac death include AVV replacement at the time of the Fontan operation and Fontan pressures greater than 20 mmHg [32].

            A thorough evaluation should be performed in Fontan patients with arrhythmias, especially in patients with frequent atrial arrhythmia or a severer presentation. This may include evaluation of ventricular function, assessment for anatomical connections by cross-sectional imaging, and hemodynamic measurements through cardiac catheterization, with intervention as needed. Cardioversion can be performed in cases of symptomatic rapid conduction, but radiofrequency ablation and long-term medical therapy is often needed in addition [25, 27]. Radiofrequency ablation can be helpful in reducing the occurrence of arrhythmia, but because of new reentrant pathways emerging, the recurrence rate is 50% within 4–5 years [34]. Although rate control can be an option, most patients are dependent on atrioventricular synchrony to maintain cardiac output and can feel unwell without it. Anticoagulation needs to be initiated in patients with atrial arrhythmias, and careful assessment for thrombosis should be performed. Fontan conversion surgery can be considered in carefully selected cases to treat refractory intra-atrial reentrant tachycardia [36].

            Ventricular Dysfunction

            Loading conditions on the ventricle throughout the various stages of Fontan palliation can cause structural remodeling (such as dilatation and hypertrophy) and cardiac dysfunction [37]. There are limited data supporting the use of medical therapy for ventricular dysfunction in single ventricle patients. Angiotensin-converting enzyme inhibitors and beta-blockers have been shown to have some benefit for systolic dysfunction of the single ventricle, but data are limited [38]. Carvedilol has been shown to improve New York Heart Association class and increase ejection fraction in Fontan patients, but use of beta-blockers may be limited by the degree of sinus node dysfunction [39]. Diuretic therapy with both loop diuretics and aldosterone antagonists can be used to treat fluid overload; additionally, aldosterone antagonists may have some positive effect on the pulmonary vascular bed [40].

            Maintaining atrioventricular synchrony can be of symptomatic benefit in patients with a single ventricle. Cardiac resynchronization therapy in patients with a single ventricle (multisite pacing) can be considered to improve synchronous ventricular activation and has shown some improvement in cardiac function after surgery [41]; however, long-term data are limited, and usually the technical limitations imposed by the need for thoracotomy for epicardial lead placement makes it a less attractive modality.

            Elevated Pulmonary Vascular Resistance

            PVR increases with the process of aging, but there are additional factors that contribute to abnormal pulmonary vasculature in the patient following the Fontan operation. A few anatomical subtypes result in pulmonary overcirculation and elevated PVR from a young age due to unobstructed pulmonary blood flow or palliative shunts. These patients continue to have changes in the vascular bed, with decreased capillary recruitment and medial hypertrophy even after the Fontan operation [42], resulting in altered nitric oxide synthesis and an increase in endothelin receptor expression [43, 44]. Elevated PVR has been associated with Fontan failure and is predictive of death in the population of patients with PLE [37, 38].

            Modifying the PVR in Fontan patients to increase cardiac output and exercise capacity has been an attractive hypothesis for many investigators. Mixed results with pulmonary vasodilator medical therapy have been reported [4548]. Van De Bruaene et al. [45] demonstrated that sildenafil increased cardiac index during exercise, with a significant decrease in PVR. Targeting the endothelin 1 system with an endothelin receptor antagonist such as bosentan has been shown to increase Vo2max and improve functional status in the short term [33, 34], although the benefits were not seen after 6 months of therapy [48]. Some studies have shown the benefit of spironolactone in combination with endothelin A receptor antagonists to increase exercise capacity in patients with pulmonary arterial hypertension [40]. While the effects have been attributed to improved cardiac function and its diuretic properties, spironolactone has also been shown to improve endothelial cell function and reduce inflammation [49]. Further multicenter randomized controlled trials are needed to explore the true benefit of pulmonary vasodilator therapy in Fontan patients.

            Cyanosis

            Pulmonary arteriovenous malformations (AVMs), persistent fenestrations, or the presence of venovenous collaterals (systemic veins–pulmonary veins) can result in persistent cyanosis in the Fontan patient [50]. Careful hemodynamic evaluation, including measurement of the Fontan pressures and the degree of shunting across the fenestration, can help identify patients with favorable hemodynamics for closure [51]. Pulmonary AVMs are seen commonly in patients who have an isolated Glenn shunt with the lack of inclusion of hepatic venous flow into the Fontan circuit or if there is a mechanical obstruction diverting the hepatic venous flow from the pulmonary circulation [52, 53]. Surgical revision to include hepatic vein flow into the Fontan baffle can be performed to treat pulmonary AVMs. Venovenous collaterals can be coiled, but Fontan pressures and patency should be carefully assessed as part of the evaluation [54].

            Mechanical or Structural Lesions

            Careful assessment for obstruction of the Fontan pathway, pulmonary arteries, and pulmonary veins should be performed by noninvasive imaging such as magnetic resonance imaging (MRI) or cardiac computed tomography. Stenosis or narrowing can potentially be treated in the cardiac catheterization laboratory or operating room. In some cases, the pulmonary veins can be mechanically compressed by the massively enlarged right atrium in an APC Fontan patient [55]. Certain anatomical subtypes might also have components of outflow tract obstruction, such as subaortic obstruction, coarctation of the aorta, or stenosis at the sites of neoaortic anastomosis such as in the Norwood operation. Alleviating these obstructions can increase cardiac output. Patients with severe valve regurgitation may need valve replacement if they are symptomatic and showing signs of low cardiac output [56].

            Extracardiac Organ System Involvement

            Fontan-Associated Liver Disease

            The liver is one of the main organs affected by the altered hemodynamics of the Fontan circulation. Recent studies have reported approximately 21% of Fontan patients develop cirrhosis [57, 58]. Probably the strongest predictor of liver dysfunction is the time since the Fontan operation. Pundi et al. [58] demonstrated that the 10-, 20-, and 30-year rates of freedom from cirrhosis are 99%, 94%, and 57%, respectively.

            The cause of Fontan-associated liver disease is likely multifold, with chronic venous congestion/elevated central venous pressures, hypoxia, and decreased cardiac output all playing a role in development [59, 60]. Other potential mechanisms include pre-Fontan hypoxemia [61], perioperative liver injury [62], and altered hepatic blood flow.

            However, it is not evident how long these changes have to exist for significant hepatic dysfunction to occur. Additionally, the diminished portal venous flow in the setting of systemic venous hypertension increases the hepatic arterial flow, leading to the formation of hypervascular nodules [63]. Usually these nodules are benign and reported as focal nodular hyperplasia, but they often raise a concern for hepatocellular malignancy, which has been observed in Fontan patients [64, 65].

            The most common histological feature of the liver in Fontan patients is the presence of hepatic fibrosis, which can be either portal or sinusoidal, which is different from other causes of cardiac cirrhosis, where the predominant feature is sinusoidal fibrosis [66, 67]. Results from imaging modalities have not corresponded consistently with biopsy results, but ultrasonography, computed tomography, or MRI should be considered for baseline screening and evaluation in these patients [68]. Elastography (ultrasonography or MRI) can quantitate the degree of fibrosis noninvasively, but limited data exist in the Fontan population. The imaging modalities could also identify markers for portal hypertension, including splenomegaly and esophageal varices, collateral vessels, including intrahepatic and extrahepatic venovenous collaterals, and portopulmonary venous collaterals.

            Regular screening for liver dysfunction could include checking the hepatic panel (alanine aminotransferase, aspartate aminotransferase, γ-glutamyltransferase, prothrombin time, activated partial thromboplastin time, albumin, total protein) and complete blood count (especially platelet count) every year. Usually hepatic function as measured by routine blood parameters is well preserved in these patients [58, 6971], although mild elevation of indirect bilirubin and alkaline phosphatase levels in addition to decreased platelet levels might be seen in some. All patients should undergo evaluation for hepatitis B and hepatitis C, especially those operated on before routine screening, and it is reasonable to check alpha fetoprotein levels during initial screening in all patients. Vaccination against hepatitis A and hepatitis B should be considered in all Fontan patients. Screening guidelines are being established, but in general, imaging should be performed as a screening tool with frequency determined by the cardiac and hepatology teams. In cases with suspected more advanced liver disease, liver biopsy may be indicated and can be performed through the transjugular or percutaneous approach, simultaneously with cardiac catheterization. Fontan patients should be counseled to avoid hepatic toxins, including alcohol, and to adhere to available preventive measures, including hepatitis vaccination. There are no established screening or treatment protocols, although use of therapies to maximize cardiac output, decrease pulmonary pressures and resistance, and optimize oxygen delivery is a strategy that may delay the progression of disease.

            Thromboembolism

            Patients after the Fontan operation have been reported to have numerous clotting abnormalities, including deficiencies of protein C, protein S, and antithrombin III, in addition to increased platelet reactivity [64, 65]. The combination of decreased cardiac output, a nonpulsatile low-flow state to the pulmonary circulation, atrial arrhythmias, and the presence of prosthetic material places these patients at high risk of thrombus formation. The rate of thromboembolism has been reported to be as high as 20% in patients after Fontan completion; for those with extracardiac conduit, it is estimated to be 7.1% at 10 years after Fontan completion [66, 67, 72]. Among patients with underlying atrial arrhythmia, the prevalence of thromboembolic complications is 18% and 55% at 5 and 10 years, respectively [73]. This can result in pulmonary emboli, acute and chronic; systemic emboli through patent fenestration or baffle leaks; and systemic thrombi in the atria, pulmonary artery stump, or rudimentary ventricle. Thromboemboli in the pulmonary vasculature can produce acute hemodynamic instability and chronically result in elevated Fontan pressures and increased PVR. Systemic sources of thrombi place the patient at risk of stroke and end-organ system damage. Cerebrovascular events have been reported in 4.1–5.5% of patients; most are embolic in nature [74, 75].

            Prophylactic treatment guidelines recommend aspirin, heparin, or vitamin K antagonists, although the efficacy of each has not been well established. In patients with atrial arrhythmia after the Fontan operation, anticoagulation with vitamin K antagonists with or without antiplatelet therapy reduces the risk of thromboembolic complications by 2.5-fold as compared with those not receiving anticoagulation, and is associated with lower risk of death and hospitalization, while the rates of bleeding are similar (at 7%) [73]. There is a global ongoing prospective registry (NOTE registry) evaluating the safety of initiation of treatment with novel oral anticoagulants and transition from vitamin K antagonists to novel oral anticoagulants; promising results were shown during the first month [76].

            Pulmonary/Plastic Bronchitis

            Plastic bronchitis is a debilitating late complication after the Fontan procedure. It is characterized by the formation of casts in the tracheobronchial tree. The exact cause is unknown; it is speculated that it could be related to hypersecretion of mucus in the presence of chronically elevated systemic venous pressure, or lymph leak into the airway [77, 78]. It is a strong predictor of poor outcome as the onset of plastic bronchitis within 1 year of Fontan surgery is associated with a five-fold increase in mortality rate [77]. The risk factors for developing this complication include prolonged chest tube drainage after Fontan surgery, history of chylothorax associated with staged palliation, and history of ascites. Various management strategies have been proposed, including systemic steroids, dornase alfa, inhaled tissue plasminogen activator, and casts removed via bronchoscopy, all of which have only marginal benefit [79, 80]. More recently, embolization of pulmonary lymphatics has been shown to be an effective treatment for patients with plastic bronchitis and abnormal pulmonary lymphatic flow [81]. This is a promising new treatment for a subset of these patients where lymphatic contribution is a basis for the development of plastic bronchitis. Heart transplant for severe plastic bronchitis refractory to medical treatment has a high mortality rate; however, patients who survive the early postoperative period have a resolution of the plastic bronchitis.

            Fontan patients are at a risk of other pulmonary complications, also seen in other CHD or non-CHD populations. Airway disease such as ciliary dysfunction has a high prevalence in patients with heterotaxy syndrome. These patients can present with impaired airway clearance, and may require prolonged intubation after the procedure, which can be harmful for a Fontan patient. Any single-ventricle patient with heterotaxy should be evaluated for symptoms of ciliary dysfunction [69]. Pulmonary venous desaturation in the absence of venovenous collaterals should prompt a pulmonary evaluation for lung parenchymal disease.

            Obstructive sleep apnea, very commonly seen in the general population, can also raise pulmonary pressures, increase PVR in Fontan patients, and increase the risk of refractory atrial arrhythmias [82]. While continuous positive airway pressure may seem harmful for the Fontan physiology, the rise in pressure during periodic obstruction causes greater alterations in Fontan filling and rise in Fontan pressure. Untreated sleep apnea can alter endothelial function, further compounding the vascular changes. Screening for sleep apnea in Fontan patients should be performed when clinically indicated [83]. Finally, restrictive lung disease from prior surgery, scoliosis, and obesity is very common in the adult CHD population and places these patients at higher risk of elevated PVR and impaired filling of the Fontan pathway. Cardiopulmonary rehabilitation may be needed for respiratory muscle training to help relieve symptoms and increase forced vital capacity [84].

            Chronic Venous Insufficiency

            Chronic venous insufficiency (CVI) is associated with chronic pain, decreased physical function, social isolation, and depression. It is usually graded according to a scale describing clinical, etiological, anatomical, and pathophysiological (CEAP) classifications [85, 86]. The prevalence of CVI is twice as high in Fontan patients compared with the general population [87], with 22% of Fontan patients having severe venous insufficiency (CEAP grade ≥4). The cause of CVI in Fontan patients is due to underlying systemic venous hypertension on distention and engorgement of the veins with induction of inflammation in the vascular wall and venous valves, leading to loss of function of the venous valves and reflux [88, 89].

            Renal Failure

            Renal failure has been demonstrated as one of the predictors of late death in Fontan patients, with a hazard ratio of 2.5 [22]. There are limited data on the prevalence and exact pathology of renal dysfunction in Fontan patients. Anne et al. [90] reported the presence of microalbuminuria in a small cohort of Fontan patients, defined as albumin/creatinine ratio greater than 20 μg/mg. Microalbuminuria is usually an early indicator of glomerulopathy, and in diabetic patients corresponds to increase in cardiovascular morbidity and mortality [91]. The renal injury in Fontan patients is likely related to the elevated central venous pressure with its effect in increasing the efferent arteriolar pressure. Renal hypoperfusion, due to decrease cardiac output, could also be a contributing factor through decreasing the afferent arteriolar pressure, analogous to the pathophysiology encountered in Fontan-associated liver disease. Other potential mechanisms include the effect of cardiopulmonary bypass injury [92]. The use of angiotensin-converting enzyme inhibitors may be beneficial in preserving the renal function in these patients. Further studies are required to explore the prevalence, pathophysiology, and management of renal dysfunction in Fontan patients.

            Protein-Losing Enteropathy

            PLE is a rare complication that occurs in cardiac diseases where there are chronically elevated systemic venous pressures, such as constrictive pericarditis, long-standing congestive heart failure, and after the Fontan operation [93, 94]. The exact pathophysiology is unknown, although it is hypothesized that elevated systemic venous pressures in the superior vena cava lead to disturbed thoracic duct drainage, with increase in the lymphatic system pressures, resulting in intestinal protein loss. Moreover, inflammatory response caused by chronic lymphatic congestion and chronic protein leakage from the lymphatics could contribute. PLE occurs in 5–15% of patients after the modified Fontan operation, and has been historically a difficult complication to treat [95, 96]. It is characterized by the enteric loss of proteins such as albumin, immunoglobulins, and clotting factors, leading to the clinical findings of peripheral edema, ascites, diarrhea, weight loss, and malabsorption. Diagnosis is obtained through measurement of serum total protein and serum albumin level. Gastrointestinal protein loss can be assessed by measurement of α1-antitrypsin level in the stool [97]. PLE is usually a marker of poor prognosis, with 50% mortality at 5 years after initial diagnosis, although more recent reports have shown an increased survival rate likely related to advances in management strategies [95, 98, 99]. Patients with Fontan pressures greater than 15 mmHg at the time of diagnosis of PLE had lower 5- and 10-year survival rates (83% and 63%, respectively) compared with patients with Fontan pressures lower than 15 mmHg (95% and 86%, respectively). Additional factors seen with increased mortality are lower cardiac index, decreased ventricular function, elevated PVR, lower mixed venous saturation, New York Heart Association class greater than II, and higher serum creatinine level.

            There is no reliable therapy for PLE, and management strategies are primarily individualized. A thorough evaluation should be performed on any Fontan patient with PLE or failing physiology. A suggested evaluation is detailed in Table 1. A treatment algorithm for PLE in Fontan patients has been outlined, and primarily involves the combination of intestinal directed therapies and cardiac and extracardiac directed therapies [99]. The proposed mechanisms of benefit from dietary and medical therapies are outlined in Table 2 [101103]. Surgical and interventional therapies include creation of fenestration to decrease the Fontan pressures but at the expense of accepting a degree of cyanosis [103]. Patients should be considered for anticoagulation after fenestration because they are at higher risk of embolic phenomena. Data on the impact of heart transplant on PLE are limited; however, resolution of PLE symptoms after transplant has been documented, with similar morbidity and mortality after transplant as compared with patients without PLE [99, 104, 105]. More recently, lymphatic embolization has been shown to be effective in increasing albumin levels and reducing symptoms [106].

            Table 1

            Evaluation of Adults with Fontan Palliation.

            Evaluation typeGoals of evaluation
            Detailed historyDetails of operations and reoperations as a child
            Postoperative complications
            Onset of symptoms
            Arrhythmia symptoms
            Sleep apnea symptoms
            GI bleeding history Presence of ascites, edema, cyanosis
            Thyroid dysfunction
            Liver dysfunction
            LaboratoryComplete blood count
            Basic metabolic profile with BUN/Cr ratio
            Serum albumin and total protein levels
            Hepatic function testing
            Hepatitis panel, alpha fetoprotein level
            Thyroid function testing Serum pregnancy testing
            Urine analysis for protein
            Stool α1-antitrypsin level
            Referrals
             Gastroenterology/hepatologyEvaluation for hepatic dysfunction
            Rule out other causes of PLE
             Obstetrics/gynecologyContraceptive counseling
            Pregnancy risk assessment
            Noninvasive
             EchocardiographyVentricular function
            Valve regurgitation/stenosis
            Suggestion of Fontan obstruction
            Presence of fenestration
            Aortic coarctation or pulmonary artery stenosis
             Electrophysiology assessment (ECG, 24-h Holter monitoring)Sinus node dysfunction
            Heart blocks
            Atrial or ventricular arrhythmias
            Pacing
             Cardiopulmonary exercise testingExercise tolerance
            Chronotropic competence
            Arrhythmias
            Cardiac respiratory/skeletal muscle limitations
             Cardiac CT/cardiac MRIObstruction in Fontan pathway
            Cardiac index
            Presence of collateral vessels
            Ventricular volumes and function
            Degree of valve regurgitation
            Aortopathy
            Invasive
             Cardiac catheterizationObstruction in Fontan pathway
            Pressure in Fontan circuit
            Pulmonary vascular resistance
            Cardiac index
            Mixed venous saturation
            Ventricular end-diastolic pressures
            Presence of collateral vessels

            Modified from John [100].

            BUN, blood urea nitrogen; Cr, creatinine; CT, computed tomography; ECG, electrocardiography; GI, gastrointestinal; MRI, magnetic resonance imaging; PLE, protein-losing enteropathy.

            Table 2

            Proposed Mechanism of Benefit of Medical Therapies for Protein-Losing Enteropathy (PLE).

            TherapyMechanismMonitoring
            High-protein (>2 g/kg/day), low-fat (<25% of calories from fat) diet with medium-chain triglyceridesMedium-chain triglyceride supplements are absorbed directly into the bloodstream, bypassing the damaged lymphatic system in patients with PLE
            Subcutaneous unfractionated heparinCreates mechanical barrier by decreasing permeability of the basal membrane to large molecules such as albumin, may decrease inflammation and potential microthrombi to the mesenteric arteriesAssess bleeding risk
            BudesonideAn enteric-specific steroid, targets inflammationLiver function (laboratory tests and imaging) – contraindicated if abnormal
            OctreotideDecreases lymphatic flow; used as an adjunct and not as initial therapyIncreased gallstone formation
            SpironolactoneDiuretic properties, improves endothelial function, and reduces inflammationElectrolytes, renal function
            Pulmonary vasodilatorsReduce the pulmonary vascular resistance and promote forward flow through the Fontan circulation, thus improve cardiac outputLiver function tests, strict contraception
            Loop diureticsDiuretic propertiesElectrolytes, renal function

            Fontan Failure

            Fontan failure is a complex clinical syndrome that includes some physiological phenotypes in addition to the traditional acquired heart failure definitions of “heart failure with preserved ejection fraction” and “heart failure with reduced ejection fraction” [107]. As discussed earlier, even in the presence of preserved systemic ventricle function, patients after the Fontan operation are in a chronic state of low cardiac output. The long-term consequences of this circulation affect many organs systems, as described in detail earlier. However, the organ systems in individual patients are affected differently, and to variable extents. These multiorgan complications contribute to the diverse phenotypes of Fontan failure. After anatomical issues contributing to Fontan failure have been ruled out, four phenotypes of Fontan failure have been described on the basis of the underlying hemodynamics [107]. The characteristics of Fontan failure may change over time, and therapies should be directed on the basis of individual phenotypes. Clinical response to treatment usually differs depending on the type of phenotype the patient is experiencing.

            Consideration for Transplantation

            While survival has continued to improve in the Fontan population, heart transplant is still reserved for patients after all other medical and surgical therapies have failed. The survival rate has increased overall, although it is still significantly lower in than the general population without CHD [108, 109]. Unfortunately, death while on the waiting list or delisting because of worsening clinical status is disproportionately common for adult CHD patients [110]. An additional challenge is that many Fontan patients also have high panel reactive antibody levels because of previous blood transfusions and procedures that markedly increase the risk of rejection.

            As part of the transplant workup, it is important to perform a multiorgan system evaluation to assess the patient for liver, kidney, and pulmonary dysfunction. In some cases, the heart transplant needs to be performed in combination with transplant for another failing organ system. While elective cardiac operations using cardiopulmonary bypass are contraindicated in patients with moderate to severe cirrhosis (Child-Pugh class B and C), an earlier stage of liver injury tends to be reversible after heart transplant, and in itself is not considered a contraindication for heart transplant [111]. Combined heart and liver transplant should thus be considered in patients with irreversible liver disease. However, it is still unknown what criteria should be used to determine when the hepatic changes will be reversible after heart transplant.

            Despite all these challenges, the post–heart transplant survival rate for Fontan patients at 1, 5, and 10 years in an Australian and New Zealand Fontan registry [112] was 91%, 78%, and 71%, respectively. Compared with other patients with CHD (n=87), Fontan patients had similar in-hospital outcomes and long-term survival. Similarly, a Mayo study showed overall 1-, 5-, 10-, and 15-year posttransplant survival rates of 80%, 72%, 69%, and 55%, respectively (in 44 of 1138 Fontan patients, 27% had PLE), suggesting that heart transplant remains a reasonable option for patients with a failed Fontan circulation [113].

            The use of ventricular assist devices has gained increasing attention over the past few years. Devices have been used both as a subpulmonary pump to drive blood forward through the pulmonary circuit and also in the standard systemic ventricular position to pull blood through the Fontan circuit by lowering the left atrial pressure [114, 115]. Placement of the device may be dependent on the underlying hemodynamics: systemic ventricular failure versus elevation of PVR. Further investigation of ideal device strategies and prototypes is needed as ventricular assist device therapy has the potential to not only serve as a bridge to transplant but as eventual destination therapy for this complex cohort of patients.

            Pregnancy in Fontan Patients

            With the increase in survival among Fontan patients, more women are reaching the reproductive age group and expressing interest in having children; therefore there has been growing interest in developing risk scores that could predict maternal and neonatal outcomes during pregnancy and at long-term follow-up. Although the existing risk scoring models (CAPREG, ZAHARA, WHO) are helpful, they are not specific and do not take into account the physiological and hemodynamic considerations of the Fontan patient. Preconception evaluation of the Fontan patient should take into account the ventricular function, degree of AVV regurgitation, functional capacity, arrhythmia burden, and any other potential comorbidities, including liver disease, renal disease, and history of thromboembolic events. Care during pregnancy should be provided by a multidisciplinary team including an adult CHD specialist, a high-risk obstetrician, and anesthesia specialists with expertise and experience in managing such complex patients. Although increasing data have been accumulating on the outcomes of pregnant Fontan women, the evidence in this area is still limited to a very small number of patients in the studies. In a multicenter observational study by Gouton et al. [116], the incidence of cardiovascular events during pregnancy was 10% (6 of the 59 pregnancies), with atrial arrhythmia being the most common (three patients), followed by thromboembolic events (two patients). Fetal loss was seen in 34% patients (27% were miscarriages), and among women who completed pregnancy, prematurity (69%) was the most common neonatal complication. There was no significant worsening of clinical status after a median follow-up of 24 months.

            Anticoagulation of the Fontan patient during pregnancy is clearly indicated in the presence of atrial thrombus, atrial arrhythmia, or a history of thromboembolic events; however, there is no evidence for the benefit of routine use of prophylactic anticoagulation (recommendation class IIa, level of evidence C) [117].

            Conclusions

            With recent advances in surgical techniques and medical management, long-term survival after the Fontan operation has significantly improved. However, there are numerous long-term complications, including Fontan failure, arrhythmia, premature cardiac death, liver disease, PLE, and need for reoperation. As more patients survive to adulthood, innovative management strategies to maintain quality of life and extend life expectancy are needed.

            Conflict of Interest

            The authors declare that they have no conflicts of interest.

            References

            1. FontanF, BaudetE. Surgical repair of tricuspid atresia. Thorax 1971;26(3):2408.

            2. de LevalMR, KilnerPJ, GewilligM, BullC. Total cavopulmonary connection: a logical alternative to atriopulmonary connection for complex Fontan operations. J Thorac Cardiovasc Surg 1988;96(5): 68295.

            3. MarcellettiC, CornoA, GiannicoS, MarinoB. Inferior vena cava-pulmonary artery extracardiac conduit. A new form of right heart bypass. J Thorac Cardiovasc Surg 1990;100(2):22832.

            4. KhairyP, PoirierN, MercierL. Univentricular heart. Circulation 2007;115:80012.

            5. BridgesND, MayerJE, LockJE, JonasRA, HanleyFL, KeaneJF, et al. Effect of baffle fenestration on outcome of the modified Fontan operation. Circulation 1992;86(6):17629.

            6. BridgesND, LockJE, CastanedaAR. Baffle fenestration with subsequent transcatheter closure. Modification of the Fontan operation for patients at increased risk. Circulation 1990;82(5):16819.

            7. OnoM, BoethigD, GoerlerH, LangeM, Westhoff-BleckM, BreymannT. Clinical outcome of patients 20 years after Fontan operation – effect of fenestration on late morbidity. Eur J Cardiothorac Surg 2006;30:9239.

            8. MonagleP, KarlTR. Thromboembolic problems after the Fontan operation. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu 2002;5(1):3647.

            9. KaulitzR, ZiemerG, PaulT, PeusterM, BertramH, HausdorfG. Fontan-type procedures: residual lesions and late interventions. Ann Thorac Surg 2002;74(3):77885.

            10. SenzakiH, MasutaniS, IshidoH, TaketazuM, KobayashiT, SasakiN, et al. Cardiac rest and reserve function in patients with Fontan circulation. J Am Coll Cardiol 2006;47(12):252835.

            11. FrommeltPC, SniderAR, MelionesJN, VermilionRP. Doppler assessment of pulmonary artery flow patterns and ventricular function after the Fontan operation. Am J Cardiol 1991;68(11):12115.

            12. PiranS, VeldtmanG, SiuS, WebbGD, LiuPP. Heart failure and ventricular dysfunction in patients with single or systemic right ventricles. Circulation 2002;105(10):118994.

            13. Akbari AsbaghP, Navabi ShiraziMA, SoleimaniA, RazzazM, Akbari AsbaghN, RayatzadehH, et al. Incidence and etiology of chylothorax after congenital heart surgery in children. J Tehran Heart Cent 2014;9(2):5963.

            14. SalmanS, DrinkwaterDC, ChristianKG. Plastic bronchitis: is thoracic duct ligation a real surgical option? Ann Thorac Surg 2006;81(6):22813.

            15. DillerG-P, DimopoulosK, OkonkoD, LiW, Babu-NarayanSV, BrobergCS, et al. Exercise intolerance in adult congenital heart disease: comparative severity, correlates, and prognostic implication. Circulation 2005;112(6):82835.

            16. DillerGP, GiardiniA, DimopoulosK, GargiuloG, MüllerJ, DerrickG, et al. 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(24):307383.

            17. OhuchiH, NegishiJ, NoritakeK, HayamaY, SakaguchiH, MiyazakiA, et al. 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(2):10516.

            18. CunninghamJW, NathanAS, RhodesJ, ShaferK, LandzbergMJ, OpotowskyAR. Decline in peak oxygen consumption over time predicts death or transplantation in adults with a Fontan circulation. Am Heart J 2017;189:18492.

            19. EgbeAC, DriscollDJ, KhanAR, SaidSS, AkintoyeE, BerganzaFM, et al. Cardiopulmonary exercise test in adults with prior Fontan operation: the prognostic value of serial testing. Int J Cardiol 2017;235:610.

            20. AlsaiedT, BokmaJP, EngelME, KuijpersJM, HankeSP, ZuhlkeL, et al. Factors associated with long-term mortality after Fontan procedures: a systematic review. Heart 2017;103(2):10410.

            21. KhairyP, FernandesSM, MayerJE, TriedmanJK, WalshEP, LockJE, et al. Long-term survival, modes of death, and predictors of mortality in patients with Fontan surgery. Circulation 2008;117(1):8592.

            22. PundiKN, JohnsonJN, DearaniJA, PundiKN, LiZ, HinckCA, et al. Forty Year follow-up after the Fontan operation: long-term outcomes of 1,052 patients. J Am Coll Cardiol 2015;66(15):170010.

            23. HoseinRBM, ClarkeAJB, McGuirkSP, GriselliM, StumperO, De GiovanniJV, et al. Factors influencing early and late outcome following the Fontan procedure in the current era. The “two commandments” Eur J Cardiothorac Surg 2007;31(3):34453.

            24. D’UdekemY, XuMY, GalatiJC, LuS, IyengarAJ, KonstantinovIE, et al. Predictors of survival after single-ventricle palliation: the impact of right ventricular dominance. J Am Coll Cardiol 2012;59(13):117885.

            25. NakanoT, KadoH, TatewakiH, HinokiyamaK, OdaS, UshinohamaH, et al. Results of extracardiac conduit total cavopulmonary connection in 500 patients. Eur J Cardiorhorac Surg 2015;48(6):82532.

            26. IyengarAJ, WinlawDS, GalatiJC, WheatonGR, GentlesTL, GriggLE, et al. 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(3):46573.

            27. FontanF, KirklinJW, FernandezG, CostaF, NaftelDC, TrittoF, et al. Outcome after a “perfect” Fontan operation. Circulation 1990;81(5):152036.

            28. CollinsKK. The spectrum of long-term electrophysiologic abnormalities in patients with univentricular hearts. Congenit Heart Dis 2009;4(5):3107.

            29. GiannakoulasG, DimopoulosK, YukselS, InuzukaR, Pijuan-DomenechA, HussainW, et al. Atrial tachyarrhythmias late after Fontan operation are related to increase in mortality and hospitalization. Int J Cardiol 2012;157(2):2216.

            30. LeeJR, KwakJ, KimKC, MinSK, KimWH, KimYJ, et al. Comparison of lateral tunnel and extracardiac conduit Fontan procedure. Interact Cardiovasc Thorac Surg 2007;6(3):32830.

            31. StammC, FriehsI, MayerJE, ZurakowskiD, TriedmanJK, MoranAM, et al. Long-term results of the lateral tunnel Fontan operation. J Thorac Cardiovasc Surg 2001;121(1):2841.

            32. PundiKN, PundiKN, JohnsonJN, DearaniJA, LiZ, DriscollDJ, et al. Sudden cardiac death and late arrhythmias after the Fontan operation. Congenit Heart Dis 2017;12(1):1723.

            33. MondesertB, MarcotteF, MongeonFP, DoreA, MercierLA, IbrahimR, et al. Fontan circulation: success or failure? Can J Cardiol 2013;29(7):81120.

            34. DealBJ. Late arrhythmias following Fontan surgery. World J Pediatr Congenit Heart Surg 2012;3(2):194200.

            35. StephensonEA, LuM, BerulCI, EtheridgeSP, IdrissSF, MargossianR, et al. Arrhythmias in a contemporary Fontan cohort: prevalence and clinical associations in a multicenter cross-sectional study. J Am Coll Cardiol 2010;56(11):8906.

            36. DealBJ, CostelloJM, WebsterG, TsaoS, BackerCL, MavroudisC. Intermediate-term outcome of 140 consecutive Fontan conversions with arrhythmia operations. Ann Thorac Surg 2016;101(2):71724.

            37. GewilligM, BrownSC. The Fontan circulation after 45 years: update in physiology. Heart 2016;102(14):10816.

            38. ShaddyRE, BoucekMM, HsuDT, BoucekRJ, CanterCE, MahonyL, et al. Carvedilol for children and adolescents with heart failure: a randomized controlled trial. J Am Med Assoc 2007;298(10):11719.

            39. IshibashiN, ParkIS, TakahashiY, NishiyamaM, MurakamiY, MoriK, et al. Effectiveness of carvedilol for congestive heart failure that developed long after modified Fontan operation. Pediatr Cardiol 2006;27(4):4735.

            40. MaronBA, OpotowskyAR, LandzbergMJ, LoscalzoJ, WaxmanAB, LeopoldJA. Plasma aldosterone levels are elevated in patients with pulmonary arterial hypertension in the absence of left ventricular heart failure: a pilot study. Eur J Heart Fail 2013;15(3):27783.

            41. SojakV, MazicU, CesenM, SchraderJ, DanojevicN. Cardiac resynchronization therapy for the failing Fontan patient. Ann Thorac Surg 2008;85(6):21368.

            42. PressonRG, BaumgartnerWA, PetersonAJ, GlennyRW, WagnerWW. Pulmonary capillaries are recruited during pulsatile flow. J Appl Physiol 2002;92(3):118390.

            43. BusseR, FlemingI. Pulsatile stretch and shear stress: physical stimuli determining the production of endothelium-derived relaxing factors. J Vasc Res 1998;35(2):7384.

            44. IshidaH, KogakiS, IchimoriH, NaritaJ, NawaN, UenoT, et al. Overexpression of endothelin-1 and endothelin receptors in the pulmonary arteries of failed Fontan patients. Int J Cardiol 2012;159(1):349.

            45. Van De BruaeneA, La GercheA, ClaessenG, De MeesterP, DevroeS, GillijnsH, et al. Sildenafil improves exercise hemodynamics in Fontan patients. Circ Cardiovasc Imaging 2014;7(2):26573.

            46. HebertA, MikkelsenUR, ThilenU, IdornL, JensenAS, NagyE, et al. 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(23):202130.

            47. DerkG, HouserL, MinerP, WilliamsR, MoriartyJ, FinnP, et al. Efficacy of endothelin blockade in adults with Fontan physiology. Congenit Heart Dis 2015;10(1):E116.

            48. SchuuringMJ, VisJC, Van DijkAPJ, van MelleJP, VliegenHW, PieperPG, et al. Impact of bosentan on exercise capacity in adults after the Fontan procedure: a randomized controlled trial. Eur J Heart Fail 2013;15(6):6908.

            49. ElinoffJM, RameJE, ForfiaPR, HallMK, SunJ, GharibAM, et al. A pilot study of the effect of spironolactone therapy on exercise capacity and endothelial dysfunction in pulmonary arterial hypertension: study protocol for a randomized controlled trial. Trials 2013;14:91.

            50. VyasH, DriscollDJ, CabalkaAK, CettaF, HaglerDJ. Results of transcatheter Fontan fenestration to treat protein losing enteropathy. Catheter Cardiovasc Interv 2007;69(4):5849.

            51. GoffDA, BlumeED, GauvreauK, MayerJE, LockJE, JenkinsKJ. Clinical outcome of fenestrated Fontan patients after closure: the first 10 years. Circulation 2000;102(17):20949.

            52. KavaranaM, JonesJ, StroudRE, BradleyS, IkonomidisJS, MukherjeeR. Pulmonary arteriovenous malformations after the superior cavopulmonary shunt: mechanisms and clinical implications. Expert Rev Cardiovasc Ther 2015;12(6):70313.

            53. MasuraJ, BorodacovaL, TittelP, BerdenP, PodnarT. Percutaneous management of cyanosis in Fontan patients using Amplatzer occluders. Catheter Cardiovasc Interv 2008;71(6):8439.

            54. MageeAG, McCrindleBW, MawsonJ, BensonLN, WilliamsWG, FreedomRM. Systemic venous collateral development after the bidirectional cavopulmonary anastomosis: orevalence and predictors. J Am Coll Cardiol 1998;32(2):5028.

            55. O’DonnellCP, LockJE, PowellAJ, PerrySB. Compression of pulmonary veins between the left atrium and the descending aorta. Am J Cardiol 2003;91(2):24851.

            56. MenonSC, DearaniJA, CettaF. Long-term outcome after atrioventricular valve surgery following modified Fontan operation. Cardiol Young 2011;21(1):838.

            57. BaekJS, BaeEJ, KoJS, KimGB, KwonBS, LeeSY, et al. Late hepatic complications after Fontan operation; non-invasive markers of hepatic fibrosis and risk factors. Heart 2010;96(21):17505.

            58. PundiK, PundiKN, KamathPS, CettaF, LiZ, PoteruchaJT, et al. Liver disease in patients after the Fontan operation. Am J Cardiol 2016;117(3):45660.

            59. MoriM, AguirreAJ, ElderRW, KashkouliA, FarrisAB, FordRM, et al. Beyond a broken heart: circulatory dysfunction in the failing Fontan. Pediatr Cardiol 2014;13(1):56979.

            60. RychikJ. Forty years of the Fontan operation: a failed strategy. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu 2010;13(1):96100.

            61. GhoshML, EmeryJL. Hypoxia and asymmetrical fibrosis of the liver in children. Gut 1973;14(3):20912.

            62. MatsudaH, CovinoE, HiroseH, NakanoS, KishimotoH, MiyamotoY, et al. Acute liver dysfunction after modified Fontan operation for complex cardiac lesions. Analysis of the contributing factors and its relation to the early prognosis. J Thorac Cardiovasc Surg 1988;96:21926.

            63. BryantT, AhmadZ, Millward-SadlerH, BurneyK, StedmanB, KendallT, et al. Arterialised hepatic nodules in the Fontan circulation: hepatico-cardiac interactions. Int J Cardiol 2011;151(3):26872.

            64. GhaferiAA, HutchinsGM. 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(6):134852.

            65. WallihanDB, PodbereskyDJ. Hepatic pathology after Fontan palliation: spectrum of imaging findings. Pediatr Radiol 2013;43(3):3308.

            66. JohnsonJA, CettaF, GrahamRP, SmyrkTC, DriscollDJ, PhillipsSD, et al. Identifying predictors of hepatic disease in patients after the Fontan operation: a postmortem analysis. J Thorac Cardiovasc Surg 2013;146(1):1405.

            67. SchwartzMC, SullivanL, CohenMS, RussoP, JohnAS, GuoR, et al. Hepatic pathology may develop before the Fontan operation in children with functional single ventricle: an autopsy study. J Thorac Cardiovasc Surg 2012;143:9049.

            68. KendallTJ, StedmanB, HackingN, HawM, VettukattillJJ, SalmonAP, et al. Hepatic fibrosis and cirrhosis in the Fontan circulation: a detailed morphological study. J Clin Pathol 2008;61(4):5048.

            69. van NieuwenhuizenRC, PetersM, LubbersLJ, TripMD, TijssenJG, MulderBJ. Abnormalities in liver function and coagulation profile following the Fontan procedure. Heart 1999;82(1):406.

            70. KaulitzR, LuhmerI, BergmannF, RodeckB, HausdorfG. Sequelae after modified Fontan operation: postoperative haemodynamic data and organ function. Heart 1997;78(2): 1549.

            71. GuhaIN, BokhandiS, AhmadZ, SheronN, CopeR, MarshallC, et al. Structural and functional uncoupling of liver performance in the Fontan circulation. Int J Cardiol 2013;164(1):7781.

            72. ElderRW, McCabeNM, HebsonC, VeledarE, RomeroR, FordRM, et al. Features of portal hypertension are associated with major adverse events in Fontan patients: the VAST study. Int J Cardiol 2013;168(4):37649.

            73. EgbeAC, ConnollyHM, McLeodCJ, AmmashNM, NiazT, YogeswaranV, et al. Thrombotic and embolic complications associated with atrial arrhythmia after Fontan operation: role of prophylactic therapy. J Am Coll Cardiol 2016;68(12):13129.

            74. HoffmannA, ChockalingamP, BalintOH, DadashevA, DimopoulosK, EngelR, et al. Cerebrovascular accidents in adult patients with congenital heart disease. Heart 2010;96(15):12236.

            75. ChunDS, SchambergerMS, FlaspohlerT, TurrentineMW, BrownJW, FarrellAG, et al. Incidence, outcome, and risk factors for stroke after the Fontan procedure. Am J Cardiol 2004;93(1):1179.

            76. YangH, BoumaBJ, MulderBJM; Non vitamin K antagonist Oral anticoagulants for ThromboEmbolic prevention in adult congenital heart disease (NOTE) investigators. Is initiating NOACs for atrial arrhythmias safe in adults with congenital heart disease? Cardiovasc Drugs Ther 2017;31(4):4137.

            77. SchumacherKR, SinghTP, KueblerJ, AprileK, O’BrienM, BlumeED. Risk factors and outcome of Fontan-associated plastic bronchitis: a case-control study. J Am Heart Assoc 2014;3(2):17.

            78. LanguepinJ, ScheinmannP, MahutB, Le BourgeoisM, JaubertF, BrunelleF, et al. Bronchial casts in children with cardiopathies: the role of pulmonary lymphatic abnormalities. Pediatr Pulmonol 1999;28(5):32936.

            79. LubckeNL, NussbaumVM, SchrothM. Use of aerosolized tissue plasminogen activator in the treatment of plastic bronchitis. Ann Pharmacother 2013;47(3):e13.

            80. GibbE, BlountR, LewisN, NielsonD, ChurchG, JonesK, et al. Management of plastic bronchitis with topical tissue-type plasminogen activator. Pediatrics 2012;130(2):e44650.

            81. ItkinMG, McCormackFX, DoriY. Diagnosis and treatment of lymphatic plastic bronchitis in adults using advanced lymphatic imaging and percutaneous embolization. Ann Am Thorac Soc 2016;13(10): 168996.

            82. WatsonNF, StoutK. Management of obstructive sleep apnea in patients with congenital heart disease and Fontan procedures. Sleep Med 2007;8(5):5378.

            83. WatsonNF, BushnellT, JonesTK, StoutK. A novel method for the evaluation and treatment of obstructive sleep apnea in four adults with complex congenital heart disease and Fontan repairs. Sleep Breath 2009;13(4):4214.

            84. GindeS, BartzPJ, HillGD, DanduranMJ, BillerJ, SowinskiJ, et al. Restrictive lung disease is an independent predictor of exercise intolerance in the adult with congenital heart disease. Congenit Heart Dis 2013;8(3):24654.

            85. EklöfB, RutherfordRB, BerganJJ, CarpentierPH, GloviczkiP, KistnerRL, et al. Revised CEAP classification for chronic venous disorders: consensus statement. Phlebologie 2005;34(4):2205.

            86. PorterJM, MonetaGL, International Consensus Committee on Chronic Venous Disease. Reporting standards in venous disease: an update. J Vasc Surg 1995;21(4):63545.

            87. ValenteAM, BhattAB, CookS, EaringMG, GersonyDR, AboulhosnJ, et al. The CALF (Congenital Heart Disease in Adults Lower Extremity Systemic Venous Health in Fontan Patients) study. J Am Coll Cardiol 2010;56(2):14450.

            88. TakaseS, PascarellaL, BerganJJ, Schmid-SchönbeinGW. Hypertension-induced venous valve remodeling. J Vasc Surg 2004;39(6): 132934.

            89. TakaseS, PascarellaL, LerondL, BerganJJ, Schmid-SchönbeinGW. Venous hypertension, inflammation and valve remodeling. Eur J Vasc Endovasc Surg 2004;28(5):48493.

            90. AnneP, DuW, MattooTK, ZilbermanMV. Nephropathy in patients after Fontan palliation. Int J Cardiol 2009;132(2):2447.

            91. DinneenSF, GersteinHC. The association of microalbuminuria and mortality in non-insulin-dependent diabetes mellitus. A systematic overview of the literature. Arch Intern Med 1997;157(13):14138.

            92. RigdenSP, BarrattTM, DillonMJ, De LevalM, StarkJ. Acute renal failure complicating cardiopulmonary bypass surgery. Arch Dis Child 1982;57(6):42530.

            93. NelsonDL, BlaeseRM, StroberW, BruceRM, WaldmannTA. Constrictive pericarditis, intestinal lymphangiectasia, and reversible immunologic deficiency. J Pediatr 1975;86(4):54854.

            94. MeijersBKI, SchallaS, EerensF, Van SuylenRJ, BroersB, CheriexEM, et al. Protein-losing enteropathy in association with constrictive pericarditis. Int J Cardiovasc Imaging 2006;22(3–4):38992.

            95. MertensL, HaglerDJ, SauerU, SomervilleJ, GewilligM. Protein-losing enteropathy after the Fontan operation: an international multicenter study. J Thorac Cardiovasc Surg 1998;115(5):106373.

            96. CrupiG, LocatelliG, TiraboschiR. Protein-losing enteropathy after Fontan operation for tricuspid atresia (imperforate tricuspid valve). Thorac Cardiovasc Surg 1980;28(5):35463.

            97. ThorneSA, HooperJ, KempM, SomervilleJ. Gastro-intestinal protein loss in late survivors of Fontan surgery and other congenital heart disease. Eur Heart J 1998;19(3):51420.

            98. DriscollDJ, OffordKP, FeldtRH, SchaffHV, PugaFJ, DanielsonGK. Five- to fifteen-year follow-up after Fontan operation. Circulation 1992;85(2):46996.

            99. JohnAS, JohnsonJA, KhanM, DriscollDJ, WarnesCA, CettaF. Clinical outcomes and improved survival in patients with protein-losing enteropathy after the Fontan operation. J Am Coll Cardiol 2014;64(1):5462.

            100. JohnAS. Fontan repair of single ventricle physiology: consequences of a unique physiology and possible treatment options. Cardiol Clin 2015;33(4):55969.

            101. JohnAS, DriscollDJ, WarnesCA, PhillipsSD, CettaF. The use of oral budesonide in adolescents and adults with protein-losing enteropathy after the Fontan operation. Ann Thorac Surg 2011;92(4):14516.

            102. JohnAS, PhillipsSD, DriscollDJ, WarnesCA, CettaF. The use of octreotide to successfully treat protein-losing enteropathy following the Fontan operation. Congenit Heart Dis 2011;6:6536.

            103. MertensL, DumoulinM, GewilligM. Effect of percutaneous fenestration of the atrial septum on protein-losing enteropathy after the Fontan operation. Heart 1994;72(6):5912.

            104. GambaA, MerloM, FiocchiR, TerziA, MammanaC, SebastianiR, et al. Heart transplantation in patients with previous Fontan operations. J Thorac Cardiovasc Surg 2004;127(2):55562.

            105. SchumacherKR, GossettJ, GuleserianK, NaftelDC, PruittE, DoddD, et al. Fontan associated protein losing enteropathy and heart transplant: a pediatric heart transplant study analysis. J Heart Lung Transplant 2015;34(9):116976.

            106. ItkinM, PiccoliDA, NadolskiG, RychikJ, DeWittA, PintoE, et al. Protein-losing enteropathy in patients with congenital heart disease. J Am Coll Cardiol 2017;69(24):292937.

            107. BookWM, GerardinJ, SarafA, Marie ValenteA, RodriguezF. Clinical phenotypes of Fontan failure: implications for management. Congenit Heart Dis 2016;11:296308.

            108. BernsteinD, NaftelD, ChinC, AddonizioLJ, GambergP, BlumeED, et al. Outcome of listing for cardiac transplantation for failed Fontan: a multi-institutional study. Circulation 2006;114(4):27380.

            109. KanterKR, MahleWT, VincentRN, BergAM, KogonBE, KirshbomPM. Heart transplantation in children with a Fontan procedure. Ann Thorac Surg 2011;91(3):82330.

            110. AlshawabkehLI, HuN, CarterKD, OpotowskyAR, Light-McGroaryK, CavanaughJE, et al. Wait-list outcomes for adults with congenital heart disease listed for heart transplantation in the U.S. J Am Coll Cardiol 2016;68(9):90817.

            111. DichtlW, VogelW, DunstKM, GranderW, AlberHF, FrickM, et al. Cardiac hepatopathy before and after heart transplantation. Transpl Int 2005;18(6):697702.

            112. ShiWY, YongMS, McGiffinDC, JainP, RuygrokPN, MarascoSF, et al. Heart transplantation in Fontan patients across Australia and New Zealand. Heart 2016;102(14):11206.

            113. PundiKN, PundiK, DriscollDJ, DearaniJA, LiZ, DahlSH, et al. Heart transplantation after Fontan: results from a surgical Fontan cohort. Pediatr Transplant 2016;20(8):108792.

            114. DerkG, LaksH, BiniwaleR, PatelS, De LaCruzK, MazorE, et al. Novel techniques of mechanical circulatory support for the right heart and Fontan circulation. Int J Cardiol 2014;176(3):82832.

            115. SinhaP, DeutschN, RatnayakaK, LedermanR, HeD, NuszkowskiM, et al. Effect of mechanical assistance of the systemic ventricle in single ventricle circulation with cavopulmonary connection. J Thorac Cardiovasc Surg 2014;147(4):12715.

            116. GoutonM, NizardJ, PatelM, SassolasF, JimenezM, RadojevicJ, et al. Maternal and fetal outcomes of pregnancy with Fontan circulation: a multicentric observational study. Int J Cardiol 2015;187(1):849.

            117. European Society of Gynecology (ESG), Association for European Paediatric Cardiology (AEPC), German Society for Gender Medicine (DGesGM), Regitz-ZagrosekV, Blomstrom LundqvistC, BorghiC, CifkovaR, FerreiraR, et al. ESC guidelines on the management of cardiovascular diseases during pregnancy: the Task Force on the Management of Cardiovascular Diseases during Pregnancy of the European Society of Cardiology (ESC). Eur Heart J 2011;32(24):314797.

            Author and article information

            Journal
            CVIA
            Cardiovascular Innovations and Applications
            CVIA
            Compuscript (Ireland )
            2009-8782
            2009-8618
            May 2018
            May 2018
            : 3
            : 1
            : 107-122
            Affiliations
            [1] 1Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
            [2] 2Division of Cardiology, Department of Pediatrics, Children’s National Health System, George Washington University, 111 Michigan Avenue Northwest, WW 3rd Floor, Washington, DC 20010, USA
            Author notes
            Correspondence: Anitha John, MD, PhD, Associate Professor of Pediatrics, George Washington University, Director, Adult Congenital Heart Program, Children’s National Health System, Washington, DC 20008, USA, Tel.: +1-202-4762728, Fax: +1-202-476-5700, E-mail: anjohn@ 123456childrensnational.org
            Article
            cvia20170041
            10.15212/CVIA.2017.0041
            ed3b3fe5-6e2f-4ae5-97ec-4af516b0be8e
            Copyright © 2018 Cardiovascular Innovations and Applications

            This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 Unported License (CC BY-NC 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. See https://creativecommons.org/licenses/by-nc/4.0/.

            History
            : 29 December 2017
            : 10 January 2018
            Categories
            Reviews

            General medicine,Medicine,Geriatric medicine,Transplantation,Cardiovascular Medicine,Anesthesiology & Pain management
            Fontan,single ventricle

            Comments

            Comment on this article