Organ Transplantation


Liver Transplantation

The first successful pediatric liver transplant was performed by Tom Starzl and colleagues in 1967, but the history of liver transplantation actually began in 1955 with Stuart Welch in Albany and Jack Cannon at UCLA. Welch was the first to describe auxiliary liver transplantation in the dog and Cannon was the first to attempt orthotopic liver transplantation (OLT) in dogs. Unfortunately, none of the dogs survived the operation. Francis Moore and Tom Starzl continued research with the dog model. From 1958 to 1959, they each successfully transplanted the liver, but all of the dogs died within 4 to 20 days from rejection. These deaths highlighted the barriers that prevented the first OLT application in humans.

In the early stages of animal experimentation, the main barriers to success involved surgical technique, organ preservation, and immunosuppression. The livers were initially preserved with chilled electrolyte solutions such as lactated Ringer's and normal saline; preservation time was only 5 to 6 hours. In 1987, the University of Wisconsin developed a solution that increased the preservation of livers to 18 to 24 hours. The third barrier, immunosuppression, was the most significant and likely explained most of the canine deaths. Medawar reported the role of the immune system in organ rejection. Since that time several unsuccessful attempts to deliberately weaken the immune system and control rejection had failed. It was not until an animal model demonstrated that the combination of azathioprine and prednisone were synergistic and ameliorated rejection. This combination was first used in human kidney transplants and then expanded to liver transplantation.

The first human liver transplantation was performed in 1963 in a 3-year-old boy with biliary atresia. This attempt ended in failure secondary to fatal intraoperative hemorrhage from venous collaterals. Six more attempts at three different institutions (Denver, Boston, and Paris) produced the same result. Attempts to control the intraoperative hemorrhage with coagulation factor replacement and ε-aminocaproic acid resulted in clots and fatal pulmonary emboli in the veno-venous bypass system. Inadequate immunosuppression played a significant role in these fatalities as well. At that time a self-imposed moratorium was established. In 1967, antilymphocyte globulin was introduced, providing lymphoid depletion and supplemented azathioprine and prednisone to provide better immunosuppression, allowing Starzl to successfully transplant a liver in a 1-year-old with hepatoblastoma who survived for 13 months.

Despite the initial success of the first pediatric liver transplant, the 1-year survival rate in subsequent transplant patients remained no greater than 50%. With the introduction of cyclosporine in 1979, the 1-year patient survival increased to 70%. In 1989, tacrolimus replaced cyclosporine and the 1-year patient survival further increased to approximately 80%.

Epidemiology and Demographics

There has been a steady increase in the number of liver transplants performed yearly in the United States (from 1713 per year in 1988 to 7127 per year in 2015). The vast majority of this increase is attributed to adult transplants. The total number of pediatric liver transplants in 1990 was 513; this increased to 589 in 2000 and has essentially remained unchanged over the past 15 years with 580 in 2015. The increase from 1990 to 2015 is only a 13% increase compared with the 200% increase in adults (2177 in 1990 to 6547 in 2015) (United Network for Organ Sharing [UNOS] Scientific Registry, 2016; https://www.unos.org/data/ ).

Indications for liver transplantation in children include the presence of an underlying primary liver pathology with acute or chronic liver failure caused by cholestatic liver disease, acute hepatic failure, metabolic disorders, cirrhosis, tumors, and other derangements (e.g., Budd-Chiari syndrome) ( Table 31.1 ). The most common cause for liver transplantation in children is cholestatic liver disease secondary to biliary atresia. This is especially true in patients younger than 1 year of age, in whom it accounts for ≥50% of liver transplants. Biliary atresia continues to be the most common overall cause for liver transplantation and the most common cholestatic cause, but cholestatic liver disease secondary to total parenteral nutrition (TPN) has become more prominent over the past 10 years, accounting for just over 4% of all pediatric liver transplants. After cholestatic liver disease, acute hepatic failure and metabolic disorders are the next most common causes for pediatric liver transplantation. Historically, the most common metabolic disorders in decreasing frequency were α 1 -antitrypsin deficiency, tyrosinemia, Wilson disease, oxalosis, and glycogen storage diseases. The most common disorders have changed. Maple syrup urine disease (MSUD) is now the most common, with cystic fibrosis the second most common metabolic indication for pediatric liver transplantation (UNOS/Organ Procurement and Transplantation Network [OPTN]; https://www.unos.org/data/ ).

TABLE 31.1
Primary Diagnosis of Liver Disease in Pediatric Patients: 1988–2015
Primary Diagnosis
Total 13,340
Cholestatic 52.0%
Biliary atresia
TPN-induced cholestasis
Alagille syndrome
Primary sclerosing cholangitis
Secondary biliary cirrhosis
Familiar cholestasis (Byler's, others)
Biliary hypoplasia
Primary biliary cirrhosis
Neonatal cholestatic disease
Other causes of cholestasis
Acute hepatic necrosis 13.3%
Neonatal hepatitis
Drug-induced
Hepatitis A
Hepatitis B
Hepatitis C
Unknown
Other causes of hepatic necrosis
Metabolic Disorder 12.9%
α 1 -Antitrypsin
Cystic fibrosis
Wilson disease
Tyrosinemia
Oxalosis
Glycogen storage disease
Maple syrup urine disease
Hemochromatosis
Other metabolic disorders
Cirrhosis 8.0%
Idiopathic
Autoimmune
Hepatitis C
Chronic active hepatitis
Hepatitis B
Drug/toxin
Hepatitis A
Combined exposure (alcohol, hepatitis A, B, C)
Alcoholic
Other causes of cirrhosis
Hepatic tumors 4.8%
Hepatoblastoma
Hepatocellular carcinoma
Hemangioendothelioma
Benign tumor
Other tumors
Other 9%
Congenital hepatic fibrosis
Budd-Chiari syndrome
Graft versus host secondary to nonliver toxicity
Trauma
Other miscellaneous diagnoses

The cause for acute or fulminant hepatic failure is not known in most pediatric patients. A viral cause (A, B, or C) may account for almost half of the acute hepatic failures in infants and children. Acetaminophen is the most common overall cause of drug- or toxin-induced liver failure.

There are few absolute contraindications to pediatric liver transplantation. Children with neoplastic processes such as hepatocellular carcinoma and infections with human immunodeficiency virus (HIV) have received transplants. However, patients with acute infections from bacterial or fungal agents, metastatic neoplasm, or disease processes that are considered an immediate threat to life (severe cardiopulmonary disease, sepsis/septic shock) generally do not undergo transplantation.

Allocation of the available livers to the appropriate recipients has been a challenge. Initially liver transplant candidates were prioritized based on geographic location and medical condition defined by the Child-Turcotte-Pugh (CTP) score. Patients were ranked as status 1, 2a, 2b, or 3. Status 1 patients received the highest priority and were defined by the presence of acute liver failure of less than 6 weeks or a failed liver transplant within 1 week. Status 2a, 2b, and 3 were defined by their CTP score and time on the wait-list. Efforts by the UNOS/OPTN Liver Disease Severity Scale (LDSS) committee to identify predictors of mortality in children with chronic liver disease resulted in the implementation of the M odel for E nd-Stage L iver D isease (MELD) and the P ediatric E nd-Stage L iver D isease (PELD) severity score in 2002. The PELD score incorporates variables for age, growth failure, serum albumin, bilirubin, and international normalized ratio (INR). PELD formerly applied to children younger than 18 years old; this was changed in 2005 to include children 12 years of age or younger. The MELD score is now used for children 13 years of age or older. Serum creatinine was incorporated into the MELD score because it predicts mortality for adult patients awaiting liver transplantation. Although serum creatinine may predict survival after liver transplantation, it does not predict mortality in the child awaiting liver transplantation.

The allocation of deceased liver donors has changed with the new MELD/PELD policy. Prior to this policy, organs from donors younger than 18 years were distributed only to pediatric recipients. With the new policy, the donor graft is first allocated to a status 1 pediatric recipient in the local region. If none is available, it is offered to the next status 1 adult in the region. If no status 1 adult is available, the liver is made available to pediatric patients with a greater than 50% risk of mortality. Adults with mortality risk greater than 50% are next, and then all pediatric patients are offered the liver over all other adult candidates. If there are no appropriate pediatric recipients in the region, the donor organ is offered to the national pool. The introduction of the MELD/PELD score has decreased the wait time for transplantation. Analysis of pre- and post-MELD/PELD data indicate that the median time to transplant, defined as the number of days for half of the new registrants to receive organs, decreased from 981 days in 2002 to 361 days in 2007.

Survival for deceased donor transplants is an age-dependent variable. Infants younger than 1 year had the lowest 3-month and 1-year survival at 88% and 83%, respectively, compared with older children. However, if the infant recipient survives the first year, the survival rate increases. In fact, the 5-year survival (84%) is the greatest for infants younger than 1 year. The 10-year survival is 77% for infants younger than 1 year, 79% for children 1 to 5 years of age, and 81% for children 6 to 11 years of age.

There also appears to be a difference in recipient survival between living donor and deceased donor grafts. Children receiving a living donor organ had 10-year survival rates greater than 90%, whereas recipients of deceased donor organ had a 10-year survival rate less than 90%. In addition to survival, other outcomes still need to be evaluated in children (e.g., growth and cognitive function).

Pathophysiology of Liver Disease

The liver is the only organ that can regenerate itself when damaged. The stigmata and multiorgan involvement from end-stage liver disease occurs because of loss of hepatocytes and the resulting fibrosis. The hepatic injury and loss of hepatocytes leads to decreased synthetic function. This cellular dysfunction results in coagulopathy, hypocholesterolemia, hypoalbuminemia, and encephalopathy. Attempts at regeneration result in fibrosis and destruction of the portal triad with increased resistance to blood flow through the liver. Portal hypertension is the final consequence of this increased resistance. Much of the characteristic features of liver disease occur because of portal hypertension, specifically varices (esophageal, bowel), hemorrhoids, ascites, spontaneous bacterial peritonitis, splenomegaly with thrombocytopenia, and hepatic encephalopathy.

Cardiac Considerations

Cardiac disturbances occur because of altered physiology, congenital heart defects, and toxic medication adverse effects. A hyperdynamic circulation with a compensatory increase in cardiac output (CO) secondary to vasodilation characterizes the altered cardiac physiology from liver disease. Vasodilation is central to the hyperdynamic circulation that accompanies portal hypertension. It likely is the result of the presence of vasoactive mediators. These mediators or gut-derived “humoral factors” (nitric oxide [NO], tumor necrosis factor [TNF]-α, endocannabinoids) enter the systemic circulation through portosystemic collaterals and bypass the hepatic detoxification that usually occurs. Shunting also occurs in the skin and lungs. Mixed venous saturation is increased in patients with liver disease. Poor oxygen extraction and increased CO likely explain the increased mixed venous saturation. The arterial-venous oxygen difference is reduced because of decreased oxygen consumption and hypoxia from arterial-venous shunting.

Cardiomyopathy associated with portal hypertension is well described in adults but is not well characterized in children with liver disease. However, children with liver disease can have a cardiomyopathy for other reasons. Inborn errors of metabolism and other syndromes are associated with cardiomyopathies and cardiac anomalies. Some of the inborn errors include Wilson disease, oxalosis, glycogen storage disease type III, tyrosinemia, and Gaucher disease. Tacrolimus and cyclosporine A have also been associated with hypertrophic cardiomyopathy in animal studies and in pediatric liver transplant recipients. Other studies have demonstrated that the cardiac function is generally well preserved in pediatric liver transplant patients who are receiving tacrolimus, but there may be evidence of subtle cardiovascular changes that predispose a small percentage of patients to develop hypertrophic cardiomyopathy. Alagille disease is commonly associated with congenital heart disease (CHD) such as pulmonary stenosis, coarctation, tetralogy of Fallot, and atrial and ventricular septal defects. Diastolic dysfunction has been associated with increased mortality after transplantation.

QT prolongation of the electrocardiogram (ECG; see Chapter 16 ) has been described in adult patients with alcoholic liver disease and may be associated with sudden cardiac death. A decrease in K + currents in cardiomyocytes in rats with cirrhosis may provide a possible mechanism for the QT prolongation. Children with liver failure have also been shown to have an increase in QTc interval. A QTc greater than 450 msec has been reported in 18% of children with liver disease. These findings may increase the risk of ventricular arrhythmias; however, there are conflicting data regarding resolution after liver transplantation. Nonselective β-blockade has also been shown to reduce the QT prolongation, but it is unclear if this reduces the risk of arrhythmias or improves survival. Although previous data suggested prolonged QT did not predict survival, more recently the presence of prolonged QT was associated with a greater PELD score and portal hypertension. Pediatric patients with chronic liver disease and prolonged QT may be at increased risk of mortality while waiting for a transplant.

Pulmonary Considerations

Pulmonary hallmarks of liver disease are hypoxia and pulmonary hypertension. Hypoxia can occur for several reasons including hepatopulmonary syndrome (HPS) and V̇/Q̇ mismatch from atelectasis owing to ascites, hepatosplenomegaly, or pleural effusions. HPS is characterized by hypoxia from intrapulmonary arteriovenous shunting (caused by increased angiogenesis) and intrapulmonary vascular dilatation. The diagnosis is made by demonstrating either arterial hypoxia (Pa o 2 <70 mm Hg) or an increased alveolar-arterial gradient greater than 20 mm Hg in the setting of pulmonary vascular dilatation. Intrapulmonary vascular dilatation can best be demonstrated using echocardiography or a lung perfusion scan with macroaggregated albumin. HPS occurs in at least 15% to 20% of adults with cirrhosis. HPS has been reported in infants 6 months of age and is described in 0.5% to 20% of all children with liver disease. It appears to be more prevalent in children with biliary atresia and polysplenia syndrome. When adjusted for the severity of hepatic disease, HPS does not affect mortality. A normal pulse oximetry value in children with cirrhosis does not necessarily rule out HPS. Children with normal pulse oximetry may exhibit other criteria for the diagnosis (intrapulmonary vasodilatation and alveolar-arterial gradient greater than 15 mm Hg) and may be at increased risk for morbidity and mortality.

Treatment for hypoxia is long-term supplemental oxygen. Definitive treatment may occur with liver transplantation. One case series described seven children with HPS who had successful transplants; all seven children recovered from their HPS postoperatively. The average time required to correct hypoxia after transplantation was 24 weeks.

Portopulmonary hypertension (PPH) is defined by the World Health Organization (WHO) as pulmonary artery hypertension (pressure >25 mm Hg) in the setting of normal pulmonary capillary wedge pressure and portal hypertension. The incidence of PPH is 0.2% to 0.7% in adults with cirrhosis but increases to 3% to 9% in adults presenting for liver transplantation. The number is not known in children and is limited to case reports and case series. Evidence from case series and autopsy data suggest that the incidence of PPH is 0.5% to 5% in children with portal hypertension. Signs and symptoms on presentation were new heart murmurs, dyspnea, and syncope. Echocardiography can successfully identify pulmonary hypertension in children and adult patients with PPH. The severity of PPH predicts mortality. Adult patients with mild PPH had no increase in mortality during liver transplantation. However, those patients who underwent OLT with moderate PPH (pulmonary artery pressure [PAP] = 35–45 mm Hg) had a 50% mortality rate and those with severe PPH (PAP >50 mm Hg) had a 100% mortality rate.

There are no definitive guidelines for the management of children with PPH. Early identification is essential, and all children who present for liver transplantation should be evaluated for the presence of PPH with echocardiography. If present, cardiac catheterization needs to be performed to confirm the diagnosis, measure PAPs, and assess the response to NO and epoprostenol. Children who respond to medical management may be candidates for liver transplantation. Otherwise, severe PPH is generally a contraindication for liver transplantation because of the increased risk of mortality.

Neurologic Considerations

Hepatic encephalopathy (HE) is a neurologic complication of liver disease classified as either acute (seen in fulminant hepatic failure) or chronic (seen in chronic cirrhosis or chronic portal hypertension). Minimal encephalopathy, diagnosed with neuropsychological testing, may be present in as many as 50% of children with chronic liver disease. The pathophysiology is not entirely known but cerebral edema is a feature of both acute and chronic HE. The cerebral edema is more severe in acute HE and can result in increased intracranial pressure (ICP). Ammonia is repeatedly implicated in the pathogenesis of HE and may participate in the process by causing astrocyte to swell, resulting in low-grade cerebral edema. The two major sources of ammonia in humans are catabolism of endogenous protein and gastrointestinal absorption of exogenous protein. Bacterial breakdown of nitrogen-containing products in the gut results in ammonia formation, which is then absorbed in the portal circulation. Factors that can increase blood ammonia concentrations can exacerbate the signs and symptoms of HE. These typically include increased catabolism from infection or increased gut absorption from high-protein diets, constipation, and gastrointestinal bleeding. Other factors that have been implicated in the exacerbation of HE include benzodiazepines, hyponatremia, and inflammatory cytokines, which may all share a final common pathway to increase cerebral edema.

Management of HE should begin with assessing the child's ability to manage his or her airway. Children with grade 3 and 4 HE may require tracheal intubation to protect the airway to ensure adequate oxygenation and ventilation. Otherwise, management typically focuses on reducing gastrointestinal production and absorption of ammonia. Lactulose is often prescribed to create an osmotic diarrhea and to acidify the lumen of the gut to trap ammonia and minimize absorption. Antibiotics such as neomycin and metronidazole have been used to kill the gastrointestinal bacteria involved in metabolizing nitrogen products to ammonia. Other medications include sodium benzoate, which combines in the liver with ammoniagenic amino acids, such as glycine, to facilitate their excretion. Ornithine aspartate may also provide a substrate to the liver for enhancing the urea cycle and glutamine synthesis and to reduce ammonia concentrations. Flumazenil has been postulated to reduce the symptoms of HE by inhibiting endogenous benzodiazepines and γ-aminobutyric acid. However, this benefit was not demonstrated in children who received 0.01 mg/kg flumazenil for HE in the setting of fulminant hepatic failure.

Patients with fulminant hepatic failure can have increased ICP, which is the major cause of mortality and may be a contraindication for liver transplantation. Intracranial hypertension occurs in 38% to 81% of patients with fulminant hepatic failure. ICP is often monitored in patients with fulminant hepatic failure with grade 3 to 4 HE. However, there is a risk of intracranial hemorrhage secondary to coagulopathy. This risk can be reduced by replacing clotting factors and platelets and by placing an epidural rather than a subdural ICP monitor. Management strategies for patients with increased ICP should focus on maintaining a cerebral perfusion pressure greater than 60 mm Hg and an ICP less than 20 mm Hg. Often this includes tracheal intubation and ventilation. Patients should be positioned with their heads midline and slightly elevated to 30 degrees to facilitate venous drainage. Ventilation should focus on achieving a Pa co 2 of 30 to 35 mm Hg with minimal positive end-expiratory pressure (PEEP). Medical management to reduce ICP includes administering thiopental or propofol to minimize stimulation and to reduce ICP. Mannitol can be administered if ICP remains increased. Hypothermia has also been described in a small trial with 14 patients with fulminant hepatic failure; maintaining core body temperature at 32°C to 33°C reduced ICP, but the impact on outcome is less certain. Orthotopic liver transplantation is the definitive treatment for patients with acute or chronic HE.

Hematologic Considerations

Anemia is common and occurs because of a combination of gastrointestinal bleeding, poor nutritional state, and decreased erythropoietin production from renal failure. Portal hypertension can result in splenomegaly, which causes platelet sequestration and thrombocytopenia. All the coagulation factors (except factor VIII) are synthesized in the liver. As synthetic function declines, coagulation factor production diminishes. The reduction in bile salt also decreases the absorption of fat-soluble vitamins (A, D, E, K) and contributes to the deficiency of factors II, VII, IX, and X. The result is an elevated prothrombin time (PT) and partial thromboplastin time (PTT). Patients with acute or fulminant hepatic failure can present with a hematologic profile similar to disseminated intravascular coagulation (DIC).

Renal Manifestations

Renal failure is common in patients with acute and chronic liver disease and its cause is multifactorial. Renal failure can be classified as prerenal azotemia, acute tubular necrosis (ATN), or hepatorenal syndrome. Prerenal azotemia from hypovolemia occurs secondary to diuretic therapy, gastrointestinal bleeding, splanchnic pooling, and sepsis. ATN occurs because of decreased central blood volume secondary to central splanchnic pooling and decreased prostaglandin synthesis. The hepatorenal syndrome is characterized by renal failure in the setting of liver failure and portal hypertension. The incidence in adults with chronic liver disease is approximately 10% to 15%. The lower incidence in children (5%) possibly reflects the lack of criteria for the diagnosis of hepatorenal syndrome in children. It occurs secondary to intense renal vasoconstriction from activation of the renin-angiotensin, arginine vasopressin, and sympathetic nervous systems. This activation is a homeostatic response to the profound splanchnic vasodilation that occurs in patients with portal hypertension. Hepatorenal syndrome appears similar to prerenal azotemia (increased serum creatinine, decreased urine Na (U Na <10 mM, fractional excretion of sodium [FE Na ] <1%]) but it is differentiated by its lack of response to a fluid challenge (see Chapter 28 ). Hepatorenal syndrome is classified into two types. The rate of progression of renal failure distinguishes the two types. Type 1, which has a worse prognosis, is characterized by a rapid progression of renal failure with a 100% increase in serum creatinine in less than 2 weeks. It usually occurs in acute liver failure. Type 2 progresses over weeks to months and usually occurs in children with chronic liver disease. Regardless of the type, prognosis is poor in patients with hepatorenal syndrome with a mortality rate of 80% to 95%. The definitive treatment for hepatorenal syndrome is liver transplantation because the renal failure is reversible if the liver is replaced.

The primary goal in the management of patients with liver disease and renal failure is to exclude treatable and reversible causes of renal failure such as nephrotoxins (e.g., nonsteroidal antiinflammatory drugs), hypovolemia (e.g., diuretics, gastrointestinal bleeding), and sepsis (e.g., spontaneous bacterial peritonitis [SBP]). All nephrotoxins should be stopped and patients should be given a fluid challenge, ideally with a colloid solution. If sepsis is suspected, extensive cultures should be obtained and nonnephrotoxic antibiotics should be started.

Pretransplant renal function predicts mortality in adults undergoing transjugular intrahepatic shunt and liver transplantation. This underscores the importance or renal function and explains why serum creatinine is used in the MELD score. Preexisting renal failure is also a major determinant of survival after liver transplantation in adults. Efforts to improve renal function pretransplant may improve posttransplantation outcome. It is not clear whether serum creatinine is a predictor of mortality in children with liver disease. Type 1 hepatorenal syndrome can be managed by treating reversible causes such as providing antibiotic treatment for spontaneous bacterial peritonitis before transplantation. Critically ill children may require continuous renal replacement therapy (continuous venovenous hemofiltration, continuous venovenous hemodiafiltration) and vasopressors as a bridge to transplantation.

Metabolic Considerations

Metabolic derangements include glucose, ammonia, electrolyte, and acid-base disturbances. Electrolyte abnormalities include hyponatremia, hypokalemia and hyperkalemia, hypocalcemia, and hypomagnesemia. Hypoglycemia may occur in patients with fulminant hepatic failure or abrupt discontinuation of TPN, but hyperglycemia is more common in the intraoperative and postoperative period.

Preoperative Evaluation

The preoperative evaluation begins with a history and physical examination to identify the primary cause of liver failure and to identify liver and non–liver-related alterations in physiology that may affect the anesthetic and surgical plan. A complete review of systems identifies most of the perioperative concerns ( Table 31.2 ).

TABLE 31.2
Preoperative Evaluation of Liver Transplant Candidates
History and Physical Examination
Cause for liver failure
Identifiable syndrome or metabolic disorder
Past medical history: non–liver-related medical problems (e.g., asthma)
Past surgical history: portoenterostomy (Kasai), previous anesthetic concerns
Medications: diuretics, lactulose
Allergies
Family history of anesthesia-related problems
NPO history
Cardiovascular
Echocardiography: to identify cardiomyopathy, pulmonary HTN, congenital cardiac defects, and intrapulmonary vasodilation
Electrocardiogram: to identify arrhythmias and QT prolongation
Pulmonary
Oxygen saturation (possible arterial blood gas): to assess hypoxia, A-a gradient (HPS)
Chest x-ray: to identify pleural effusions and central line position
Hematology
Complete blood cell count: to assess anemia, leukocytosis/leukopenia (sepsis)
Prothrombin time and partial thromboplastin times
Platelet count
Thromboelastography
Renal
Blood urea nitrogen
Creatinine
Bicarbonate: to assess degree of metabolic acidosis
Neurologic
Assessment of increased intracranial pressure in acute/fulminant hepatic failure
Hepatic encephalopathy: ammonia level
Electrolytes
Na + and K + : hyponatremia and hypokalemia secondary to diuretics
Calcium
Albumin
Magnesium
Glucose
A-a gradient, alveolar-arterial gradient; HPS, hepatopulmonary syndrome; HTN, hypertension; NPO, nothing by mouth.

The primary cardiovascular concerns include acquired cardiomyopathies from liver disease and inborn errors of metabolism, congenital cardiac defects, and QT prolongation. Aside from a cardiovascular physical examination, the preoperative cardiac evaluation should include an echocardiogram and 12-lead ECG.

The pulmonary manifestations of concern include hypoxia and PPH. Oxygen saturation on room air and with oxygen identifies hypoxic patients and their response to oxygen. Children with significant intrapulmonary shunts from HPS will not increase their oxygen saturation significantly. HPS can be diagnosed by demonstrating intrapulmonary vascular dilatation on echocardiography with macroaggregated albumin.

PPH can usually be identified on echocardiography (if a tricuspid regurgitation jet is present). Patients suspected of having PPH should undergo cardiac catheterization to define the severity of pulmonary hypertension and to assess the response to pulmonary vasodilators (NO, epoprostenol). Children with severe pulmonary hypertension (PAP >50 mm Hg) are at increased risk of perioperative mortality and liver transplantation may be contraindicated.

Anemia and thrombocytopenia are common in children with liver disease and a complete blood cell count should be obtained. In addition, because of the decreased synthetic function of the liver and because of decreased vitamin K absorption, concentrations of clotting factors II, VII, IX, and X may be decreased. A PT, PTT, and platelet count should also be obtained before starting surgery.

Renal failure is common and predicts decreased survival during the posttransplant period in adults. Samples for blood urea nitrogen (BUN) and serum creatinine (SeCr) testing should be obtained.

Children should be evaluated for evidence of altered mental status, particularly those with acute hepatic failure. Increased ICP is common and it is a common cause of mortality in those with fulminant hepatic failure. Altered mental status may also be caused by HE. An ammonia concentration should be obtained as part of their evaluation. Children with advanced HE (grade 3 and 4) may require orotracheal intubation to protect their airway and mechanical ventilation to control Pa co 2 .

Baseline laboratory values should be obtained for liver function tests, sodium, potassium, calcium, glucose, and albumin. Hyponatremia and hypokalemia are common with diuretic therapy. Patients may have already received citrated containing blood products and may be hypocalcemic secondary to the chelation of calcium. Hypoglycemia occurs secondary to depleted glycogen stores in the failed liver and/or removal of long-term TPN.

A key portion of the preoperative evaluation is the preparation of the patient (child or adolescent) and the family for the anticipated risks, benefits, and clinical course. Specifically, a critically ill child will likely remain intubated and mechanically ventilated in the immediate postoperative period and may have significant facial and extremity edema. Infants and children with less severe disease or disease that does not result in portal hypertension (e.g., MSUD) may be extubated at the end of surgery. Similarly, informing the family about the potential number and location of the intravascular catheters and their associated risks can be helpful in preparing them to see their child after surgery. Informed consent should also include a discussion about the use of blood products and risks associated with prolonged positioning (peripheral nerve injury, occipital alopecia).

Intraoperative Care

Appropriate intraoperative care of the pediatric liver transplant patient requires an understanding of the surgical and anesthetic issues. Several factors affect the patient's physiology, including the underlying pathophysiology of liver disease, surgery, and response to anesthetic drugs. The surgical approach for OLT in children is similar to that for adults. The major difference is the smaller size of the recipient. The obstacles imposed by the size of the patient include a smaller blood volume, more challenging vascular access, size restriction of donor graft, veno-venous bypass (VVBP), and surgical complications such as hepatic artery thrombosis.

Anesthetic management begins with a thorough preoperative evaluation. Children older than 1 year will likely be anxious during the preoperative period. They can be premedicated with an anxiolytic like midazolam, which may be administered intravenously, orally, nasally, or rectally. Patients with HE should not receive premedication with midazolam.

Most children are regarded as having a full stomach because of delayed gastric emptying from ascites, gastrointestinal bleeding, HE, and the nonelective nature of most transplants. The exception may be those presenting for an “elective” transplant without any stigmata of portal hypertension (e.g., Crigler-Najjar syndrome or MSUD). Patients considered to have a full stomach should receive a rapid sequence induction (RSI). Induction agents should be tailored to meet the needs of the patient, but etomidate (0.2–0.3 mg/kg), propofol (2–4 mg/kg), or ketamine (2 mg/kg) are suitable options. Appropriate muscle relaxants for RSI include succinylcholine and high-dose rocuronium. The trachea should be secured with a tracheal tube. Some children may require greater inspiratory pressures to achieve adequate ventilation in the intraoperative and postoperative period compared with unaffected children because of atelectasis from pleural effusions and ascites, surgical retractors placed on the abdominal and chest wall, and a tight abdominal closure; a cuffed tracheal tube is appropriate for these children. PEEP should be used in all patients. PEEP (10 cm H 2 O) has no negative effects on liver function in donors.

Children who are not at risk for aspiration may have an inhalation induction with sevoflurane and nitrous oxide. The use of nitrous oxide is not recommended after induction of anesthesia because it may distend the bowel and expand gas emboli. Anesthesia is typically maintained with an inhalational agent, an opioid, and a neuromuscular blocking drug. Isoflurane and sevoflurane are commonly used because they are readily available, undergo minimal hepatic metabolism, and have minimal adverse effects on the liver. Desflurane also undergoes minimal hepatic metabolism and appears to be quite safe, although there are three cases reports of hepatotoxicity after desflurane exposure. Sevoflurane provided more stable hemodynamics than desflurane in one study. Propofol (with or without remifentanil, total intravenous anesthesia [TIVA], see Chapter 8 ) is also an option to maintain anesthesia during liver transplantation. It is relatively short-acting and even though the primary metabolic pathway is hepatic, there appears to be extrahepatic metabolism in the lung, kidney, and intestine. Neuromuscular blockade can be maintained with a variety of agents. Rocuronium, vecuronium, pancuronium, atracurium, and cisatracurium have all been described. Pancuronium may have the added advantage of increased heart rate, long duration, and reduced cost. The disadvantage of pancuronium, rocuronium, and vecuronium is their partial hepatic metabolism (see Chapter 7 ), but this can be overcome with appropriate monitoring and dose adjustments. Dose requirements of continuous infusions of rocuronium, vecuronium, and pancuronium are reduced during the anhepatic phase of liver transplantation but return to initial infusion rates after reperfusion. There is no change in the dose requirements of atracurium during the anhepatic phase. Atracurium or cisatracurium may be ideal in patients with combined hepatic and renal insufficiency because they do not rely on hepatic or renal function for elimination.

The liver metabolizes all opioids, with the exception of remifentanil, which is metabolized by plasma and tissue esterases. The metabolic pathway for most opioids is oxidation, although morphine undergoes glucuronidation. There is evidence that the elimination half-life and clearance of alfentanil and fentanyl are not dramatically altered in patients with cholestatic and cirrhotic liver disease. Fentanyl, sufentanil, alfentanil, and morphine have all been described and used in children undergoing liver transplantation. Fentanyl is commonly selected and is usually administered as a bolus during induction (2–10 µg/kg) and maintained as an infusion throughout the anesthetic and the immediate postoperative period (2–5 µg/kg per hour).

Vascular access is important for resuscitation and monitoring. At least two peripheral IV catheters should be placed along with a central venous line (CVL) for administration of drug infusions, vasopressors as indicted, and assessment of volume resuscitation. The CVL can also be used to monitor trends in central venous pressure (CPV) and measure superior vena cava oxygen saturation (a surrogate marker for Sv o 2 ). Larger patients can tolerate rapid infusion catheters. Blood loss can be significant during liver transplantation with estimates between 0.5 and 25 blood volumes (mean = 3.95 blood volumes). Fluid warmers and infusion devices (Level 1 Fast Flow Fluid Warmer, Smiths Medical, Rockland, MA; Belmont Rapid Infuser, Belmont Instrument Corporation, Billerica, MA) need to be available to facilitate volume resuscitation if massive hemorrhage occurs (see Chapters 12 and 52 ). The early version of the Level 1 Fast Flow Fluid Warmer was associated with massive air emboli but the newer models are equipped with air detectors. Nevertheless, all air needs to be removed from the infusion bags before starting the device. The Belmont Rapid Infuser is not routinely used in infants or small children (see Chapter 52 ). The choice of resuscitation fluids should be limited to 0.9% normal saline solution and PlasmaLyte. Lactated Ringer's solution is not recommended because the lactate will remain unmetabolized during the anhepatic stage. Many patients with liver disease are hypoalbuminemic, so the use of 5% albumin is appropriate. However, 5% albumin is hypertonic owing to a large sodium concentration and care must be taken when administering this to children with hyponatremia because it may correct the hyponatremia too rapidly and cause adverse cerebral pressure changes.

Standard monitoring should include ECG, pulse oximetry (upper and lower extremities), noninvasive blood pressure, invasive arterial blood pressure, CVP, and temperature. Other high-technology monitoring commonly used in adult liver transplantation includes transesophageal echocardiography (TEE), continuous cardiac output (CCO) catheter, bispectral index (BIS), VVBP, and more than one arterial catheter. There are limitations to the use of these monitors in children because of patient size; TEE, CCO, BIS, and VVBP are used in 0%, 7.7%, 15.4%, and 7.7% of U.S. pediatric transplant centers, respectively. The recent development of continuous noninvasive or minimally invasive CO monitors such as the Cardiotronic ICON (Osypka Medical, La Jolla, CA), which simply requires four ECG pads to assess changes in bioimpedance (approved by the U.S. Food and Drug Administration [FDA] for use in neonates) or small esophageal Doppler monitors (Deltex Medical, Chichester, West Sussex, England) (FDA-approved for use in children weighing 3 kg or more) may prove to be of great value in the future (see Chapter 52 ).

Hematologic and electrolyte changes are common during liver transplantation, and measurements of arterial blood gases, sodium, potassium, calcium, magnesium, hemoglobin platelets, and coagulation parameters (PT, PTT, fibrinogen and d -dimers, thromboelastography [TEG]) need to be performed frequently throughout the procedure. Most centers use either portable devices or an operating room laboratory to obtain these data. Assessment of coagulation variables can be obtained with TEG) ( E-Fig. 31.1 ). Point-of-care testing with TEG may reduce transfusion requirements in patients having liver transplants. However, only 28% of U.S. pediatric transplant centers used TEG.

E-FIGURE 31.1, A, The reaction time r as measured by thromboelastography (TEG) denotes the time to onset of the start of coagulation and should be 6 to 8 minutes. It represents the rate of thromboplastin formation. Prolongation of this portion of TEG usually represents factor deficiency and is treated with the administration of fresh frozen plasma (FFP). The coagulation time ( r + k ) is the period between the start of the TEG recording and the time to the generation of an amplitude of 20 mm. It is a measurement of the speed of solid clot formation. The clot formation rate is measured by the α angle and normally is greater than 50 degrees. Abnormalities of the α angle represent platelet function, fibrinogen, and the intrinsic pathway. The α-angle abnormalities are usually corrected by cryoprecipitate administration. The maximum amplitude ( MA ) is most indicative of platelet function and normally measures between 50 and 70 mm. B, Abnormalities of coagulation noted before surgery. 1: there is prolongation of the r time and a diminution of the α angle, as well as the MA. 2: improved coagulation during stage 1 is noted, as there is administration of FFP and platelets as the surgery progresses. Note the improved MA and r time. There is also an improved α angle. 3: TEG representative of the continued coagulopathy seen during stage 2 of the surgery. Note the progressive diminution of the MA and the progressive tapering of the MA, which suggest possible fibrinolysis. 4: a classic representation of fibrinolysis seen in stages 1 and 2. 5: fibrinolysis on graft reperfusion. Note that, in this case, the clot formed takes somewhat more time before dissolution occurs. 6: Amicar-treated TEG, demonstrating dramatic improvement of the fibrinolytic stage. Note the improved MA and r time, and α angle. 7: the end TEG showing a normal coagulation profile, which is representative of normal liver function from the graft, as well as continued administration of coagulation factors.

Acid-base disturbances commonly occur during liver transplantation. Children with renal disease may have a preexisting metabolic acidosis from increased bicarbonate elimination. A metabolic acidosis is typically present during the dissection and anhepatic phase of surgery but it is usually most pronounced immediately following reperfusion. Lactic acid and citrate (from blood products) are not metabolized during the anhepatic phase and contribute to the acidosis. Cross-clamping of the inferior vena cava [IVC] and aorta alters blood flow to gut and lower extremity tissue beds and may also contribute to the development of a lactic acidosis. Once the liver graft begins to function, a metabolic alkalosis can develop as the lactate and citrate are metabolized.

During the dissection and anhepatic phase there are several causes of hyperglycemia. Serum glucose will increase if there is an exogenous source or if there is altered glucose metabolism. Exogenous sources of glucose include glucose from blood products, dextrose-containing IV fluids, and damaged hepatocytes from the liver graft. Typically glucose concentrations will increase immediately after reperfusion. Glucose uptake is altered from the administration of methylprednisolone because of steroid-induced insulin resistance. Hepatic denervation likely results in alterations in insulin and glucose clearance during the postoperative period and may explain the frequent occurrence of impaired glucose tolerance and diabetes in liver transplant recipients.

Positioning is critical to prevent soft tissue and peripheral nerve injuries. All extremities should be padded and all cables and wires need to be wrapped and protected from the skin. The head should be rotated and repositioned periodically to prevent the development of a pressure sore and alopecia. To minimize the risk of peripheral neuropathy, the upper extremities should not be abducted more than 90 degrees and the wrists should not be hyperextended for the arterial catheter.

Surgical Technique

The surgical approach can be divided into four stages: hepatectomy, anhepatic, reperfusion, and biliary reconstruction.

Hepatectomy (Stage 1)

The initial description of OLT is referred to as the “classic” technique. In the classic approach, the liver is dissected to its vascular supply and the suprahepatic and infrahepatic vena cava (VC) are clamped along with the portal vein and hepatic artery. The liver is removed en bloc ( Fig. 31.1 ). The disadvantage of this approach is the VC cross-clamp and the associated reduction in preload. The piggyback technique was described 1989 and is the preferred approach for pediatric transplants because there is more flexibility with the organ size and it requires only partial clamping of the VC. The liver is dissected away from the IVC, the short hepatic veins, portal vein, and left, right, and middle hepatic vein. The infrahepatic VC of the donor is oversewn and the suprahepatic VC is anastomosed to the native hepatic veins ( Figs. 31.2 and 31.3 ). This requires only partial clamping of the IVC. A portocaval shunt can be established for patients who do not tolerate clamping of the portal vein (see Fig. 31.3 ). Typically, these are patients who have not developed collateral flow secondary to portal hypertension (e.g., MSUD).

FIGURE 31.1, The classic approach for orthotopic liver transplantation. The suture lines are visible at the suprahepatic and infrahepatic anastomoses.

FIGURE 31.2, The piggyback technique preserves the inferior vena cava (IVC). This is the view of the liver graft after the recipient's hepatic confluence is anastomosed to the donor's IVC (the infrahepatic IVC of the donor is ligated).

FIGURE 31.3, The native liver has been removed. There is a clamp across the right, middle, and left hepatic veins. The black arrow shows the portacaval shunt. The white arrow shows the hepatic artery. The dashed white arrows show the short hepatic veins.

Several physiologic considerations that take place during the hepatic dissection affect the anesthetic management. Hypotension is common and can occur as the result of changes to the cardiovascular, hematologic, and metabolic systems. The most common cause of hypotension includes hypovolemia secondary to hemorrhage and third-space volume losses. Resuscitation with citrated blood products can result in hypocalcemia and surgical manipulation can cause mechanical compression of the IVC or right ventricle. Bleeding occurs from fragile collaterals, adhesions from prior surgery (e.g., Kasai procedure), and coagulopathy. Blood conservation with a reduction in allogeneic blood exposure may have a significant benefit on perioperative morbidity including infections, intensive care unit (ICU) stay, and hospital stay. One technique to reduce blood loss is the maintenance of low CVP (decrease of 30% from baseline) as has been described in adults. The reported benefit of a low CVP is less bleeding with subsequent decrease in allogeneic blood requirements and decreased morbidity. Some studies have also suggested an overall reduction in morbidity and 1-year mortality. The technique is controversial and the potential risks include end-organ injury, such as renal or graft failure. This technique has not been described in the pediatric liver transplant population.

Hematologic abnormalities include anemia, thrombocytopenia, coagulation factor deficiency, and low fibrinogen. A progressive coagulopathy can develop during this stage. Metabolic derangements including hyperkalemia, hypocalcemia, hypomagnesemia, and acidosis occur from volume resuscitation with blood products.

Anhepatic Stage (Stage 2)

The anhepatic phase begins once the hepatic veins, hepatic artery, and portal vein are cross clamped. The dissection of the recipient's liver is completed and the organ is removed. This phase ends when the hepatic and portal vein cross clamps are removed and the graft is reperfused.

Cardiovascular changes that occur during this stage can result in hypotension. This occurs from the inferior vena cava cross-clamp, which decreases the preload. Hemodynamically, CO, CVP, and PAP decrease while systemic vascular resistance increases. Preload should be gently augmented to maintain mean arterial pressure (MAP) with the minimum filling pressures possible. Hypervolemia may cause hepatic congestion during reperfusion. Inotropic agents such as dopamine or epinephrine may be needed to maintain mean arterial blood pressure. A portocaval shunt may mollify some of these hemodynamic changes by preserving preload from the portal vein. (If VVBP is planned, it is initiated during this period.) Unmetabolized citrate causes hypocalcemia and hypomagnesemia since it chelates both cations. Acidosis occurs from the unmetabolized citrate, lactate, and other acids. Bicarbonate may be administered if there is a significant metabolic acidosis, although there is no evidence that this improves outcomes. In fact, a mild metabolic acidosis during reperfusion may not be detrimental since it will be later offset by the metabolism of citrate, which produces bicarbonate.

Once the liver is on the surgical field warm, ischemia time begins. The time to reperfusion will be brief and steps need to be in place for reperfusion. The potassium should be low-normal and the calcium should be high-normal, hemoglobin should be maintained between 9 and 10 g/dL. If the potassium is greater than 5 mEq/L, steps need to be taken to reduce it. This includes increasing the serum pH with hyperventilation and the administration of sodium bicarbonate (1–3 mEq/kg). Glucose and insulin can be administered to acutely reduce the potassium concentration as well (see Chapters 7 and 27 ). Potassium-wasting diuretics such as furosemide (0.5–1 mg/kg) can also be used. Administering fresh blood or washed red blood cells will minimize the increase in potassium during transfusion therapy. β 2 -Agonists may decrease potassium and can be used. Calcium and epinephrine should be immediately available for reperfusion.

Reperfusion (Stage 3)

The liver graft can be reperfused once the hepatic and portal vein anastomosis are complete. Before reestablishing hepatic blood flow, the graft is flushed to remove the preservation solution to minimize the reperfusion syndrome.

Many changes can occur acutely during the reperfusion period secondary to cardiovascular, hematologic, and metabolic derangements. Reperfusion syndrome is characterized by a decrease in MAP of greater than 30%. Factors participating in this event include myocardial dysfunction, arrhythmias, and bleeding. The myocardial dysfunction is attributed to the release of NO and TNF-α. Cardiovascular collapse can occur and patients may require epinephrine to correct the hemodynamic effects of reperfusion. Hyperkalemia is a common event immediately following reperfusion and may cause ventricular arrhythmias. Hyperkalemia should be treated with calcium chloride (10–30 mg/kg) initially to stabilize the cardiac membrane and then insulin and dextrose, hyperventilation, furosemide, β 2 -agonists, and sodium bicarbonate to decrease the serum potassium concentration (see previous section). The increased potassium content of the preservation solution is the cause of hyperkalemia. The University of Wisconsin solution, a very commonly used preservation solution, contains large amounts of potassium (120 mmol/L). Histidine-tryptophan-ketoglutarate solution was introduced in 1980 as a cardioplegic solution and contains significantly less potassium (10 mmol/L) than University of Wisconsin solution. A recent study comparing the two solutions found equal 1-month and 1-year graft survival with histidine-tryptophan-ketoglutarate and University of Wisconsin solution. The viscosity is reduced with histidine-tryptophan-ketoglutarate solution and may introduce itself more easily into the vascular spaces in the donor liver. Although hyperkalemia is the hallmark electrolyte disturbance in the immediate reperfusion period, hypokalemia is more common in children throughout the intraoperative period and may require correction.

Fibrinolysis can occur after reperfusion and in one study it occurred in 60% of children and 80% of adults. This occurs from increased tissue plasminogen activator activity and decreased synthesis of fibrinolysis inhibitors. Heparin effect occurs from endogenous heparinoids from the graft and residual heparin from the preservation and the release of tissue plasminogen activator from endothelial cells of the revascularized graft. Antifibrinolytic medications blunt this process, but there is concern that there may be an association with antifibrinolytics and intraoperative thrombotic events (hepatic artery and portal vein thrombosis) in pediatric patients receiving liver transplants. Children can have a mixed coagulation picture after transplantation and may be hypercoagulable because of a decrease in protein C and antithrombin III. This may result in hepatic artery thrombosis. Tranexamic acid, ε-aminocaproic acid, and other methods to reduce transfusion needs in adult liver transplants have been advocated, but there are no pediatric transplant data to support or refute the use of tranexamic acid or ε-aminocaproic acid and the adult series have small sample sizes.

Biliary and Hepatic Artery Reconstruction (Stage 4)

The final step is reestablishing hepatic artery blood flow and reconstructing the biliary system. The hepatic artery may require an anastomosis via a conduit to the infrarenal aorta in infants. This requires temporary cross-clamping of the aorta. Biliary reconstruction is established by either directly connecting the graft and the recipient's common bile ducts or by connecting the common duct of the graft to a Roux-en- Y limb of the recipient's jejunum.

During biliary reconstruction, metabolic and hematologic alterations are addressed. As the liver graft begins to function the citrate administered during the previous three phases is metabolized and the patient can develop a metabolic alkalosis . One of the hemodynamic goals includes maintaining a normal CVP. If the CVP is increased (>8–10 mm Hg), there is concern that the liver graft can become congested and not function normally. The risk of hepatic artery thrombosis ranges from 0% to 25% and is greater in infants and children. This risk may be increased if the PT and PTT are corrected to normal values. Also, the viscosity from a greater hematocrit may increase the risk of hepatic artery or portal vein thrombosis. The hematocrit does not need to be corrected to normal values; maintaining the hematocrit at 8 to 9 g/dL is safe and reasonable. Surgical techniques to reduce the risk of hepatic artery thrombosis include anticoagulation with heparin, dextran, aspirin, and alprostadil.

Split Liver Techniques and Living Donor Liver Transplants

Advances in surgical technique, tissue preservation, and immunosuppression have improved the survival in patients undergoing liver transplantation. The result is more patients waiting for liver transplantation without an increase in available organs. Children are at a disadvantage because of the size limitations. Two techniques have attempted to address these issues. In 1984, Bismuth and Houssin split an adult liver and transplanted it into a child. The reduced liver technique does not increase the number of available grafts and efforts were made to perform split liver techniques to make two grafts from one adult donor. The initial results were poor, with an increase in complications and mortality. The technique has evolved and today the graft is split while still in vivo (in the heart-beating donor) compared with ex vivo (splitting performed after the graft is removed from the donor). This decreases cold ischemia time and facilitates hemostasis of the liver edge. The result is improved patient and graft survival. Patient survival has increased from 60% to 70% in the 1990s to 80% to 90% in 2003. In one series, 218 split liver technique grafts were transplanted between 1995 and 2002; overall patient survival at 1 year was 81.7% and overall graft survival was 75.8%. Surgical complications that caused a return to the operating room were bleeding (9.2%), bowel perforation (8.3%), and biliary problems (7.5%). Hepatic artery complications occurred in 6.7%.

Living donor liver transplantation was first described in 1989. The result has been a reduction in mortality among children awaiting liver transplantation. The benefit of a living donor (especially if related) is improved posttransplant results because of better graft quality, shorter ischemic times, and better immune compatibility. One- and five-year patient survival rates were 94% and 92%, respectively. The left hepatic segment is removed for pediatric recipients, whereas the right hepatic lobe is removed for adult recipients. The regenerative capacity of the liver allows the donor to regenerate the liver without hepatic insufficiency. Despite the success of this technique for the recipients, there is considerable risk to the donor. Complications include exposure to blood products, short- and long-term peripheral nerve injuries, biliary leakage, abdominal wall defects, pleural effusions, pneumonia, pulmonary emboli, and death.

Outcomes

The Studies of the Pediatric Liver Transplantation (SPLIT) registry was initiated in 1995 and consists of 38 centers in the United States and Canada. These centers contributed 85% of the pediatric liver transplants in 2002. Transplants performed more recently had improved survival rates. In the past, age younger than 1 year was considered an increased risk factor for mortality, but over the past 20 years there is little difference between patients younger than 2 years and those older than 2 years. There is also no significant difference between male and female gender.

Review of the MELD/PELD data indicates that survival also depends on the preoperative MELD/PELD score. Patients stratified to status 1 had a lower 1-year survival rate compared with other transplant recipients (76% vs. 87%). Adults with greater MELD scores (scores >35) demonstrated decreased 1-year patient and graft survival. Pediatric patients with greater PELD scores showed a trend toward decreased 1-year patient and graft survival but the association was not statistically significant. The overall 1-year survival rate remained excellent at >85%. Cognitive outcomes appear to be reduced in pediatric recipients of liver transplants; long-term cognitive and academic deficits persist with verbal comprehension, working memory, mathematical computation, and executive deficits. Factors that seemed to predict cognitive deficits included operative complications and intraoperative transfusion volume. Conversely, liver transplantation improved global functioning in some children.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here