Biliary Atresia – 10 Interesting Facts
- Biliary atresia is the most common cause of obstructive jaundice in the first 3 months after birth; it is also the most common cause of end-stage liver disease and liver transplant in children worldwide2
- It is imperative to differentiate benign causes of jaundice from those that require emergent intervention
- Early identification of BA allows for earlier intervention and improved outcomes
- Dark urine and acholic stools are distinctive of cholestasis
- Elevated direct bilirubin level (greater than 1 mg/dL the first 2 weeks after birth) is diagnostic of neonatal cholestasis
- Obtain abdominal ultrasonography to assess for underlying anatomic cause of cholestatic jaundice when direct hyperbilirubinemia is confirmed
- Intraoperative cholangiogram is the gold standard diagnostic study for BA
- First line treatment for BA is the KPE, which allows bile drainage into the small intestine, thereby decreasing liver injury
- Curative treatment for BA is liver transplant
- Patients who undergo transplant require lifelong immunosuppression
Alarm Signs and Symptoms
- Acholic stools are distinctive of cholestasis and necessitate prompt evaluation to diagnose etiology
- Ascites consistent with portal hypertension requires salt and fluid restriction and diuresis
- Variceal bleeding, a sign of decompensated liver cirrhosis, requires fluid resuscitation and transfusion. In addition, coagulopathy may require fresh frozen plasma and platelet transfusion
- Fever, irritability, and abdominal pain can be consistent with cholangitis and require further workup and broad-spectrum antibiotics
Introduction
- BA (biliary atresia) is a neonatal progressive obliterative fibroinflammatory disease of the intra- and extrahepatic biliary tree1
- Cholestasis is defined as reduced bile formation or excretion
- Neonatal cholestasis is defined as direct or conjugated bilirubin level above 95% the reference range for laboratory value within the first few days of life or greater than 1 mg/dL 2 weeks after birth
- Cholestasis can be caused by various etiologies affecting the formation or excretion of bile at the level of the hepatocyte (hepatocellular cholestasis) or at the level of the biliary tree (obstructive cholestasis)
- Cholestasis is always pathologic and characterized by elevation in serum conjugated bilirubin level and bile acids
Background Information
- Neonatal cholestasis, although uncommon (1 per 2500 term infants), can be present at birth and is due to impaired bile flow
- BA
- Most common cause of obstructive jaundice in the first 3 months after birth
- Most common cause of end-stage liver disease and liver transplant in children worldwide2
- Surgical excision of the obliterated extrahepatic biliary tree and reestablishment of bile flow by KPE (Kasai portoenterostomy) are essential for overall native liver survival, with earlier intervention associated with better outcomes34
- If untreated, BA is uniformly fatal
- Only 20% to 30% of children reach adulthood with their native liver56 despite the surgical intervention due to the progressive fibroinflammatory nature of the disease
Epidemiology
- Prevalence is 5 to 10 per 100,000 births in the United States7
- Incidence varies around the globe, which is possibly related to genetics, environmental exposure, viruses, and toxins8910
- Highest incidence in French Polynesia (1-3 per 10,000), followed by Taiwan (1 per 8000), and then Europe and North America (approximately 0.5 per 10,000)311
- More common in females, preterm infants, and Asian and Black people712
- Seasonal variation has been reported in some studies, with a small increased incidence from August to October13 and December to March;14 the seasonality suggests the possibility of viral-induced etiology8
- Isolated BA is more common in infants born to mothers with pregestational diabetes (PR [prevalence ratio], 4.94)7
Etiology
Figure 1. Multifactorial etiology for BA.BA, biliary atresia; BASM, biliary atresia splenic malformation syndrome.
- Onset of disease has been controversial; however, evidence now supports a “fetal disease”715
- Several causative factors, both acquired and genetic, have been reported (Figure 1)
- Viral etiology
- Clustering of cases during certain seasons can support a viral etiology; however, observing the presence of viruses in babies with BA did not have reproducible outcomes16
- Largest body of evidence in support of viral infection pertains to cytomegalovirus15171819
- Other viruses implicated include reovirus and group C rotavirus2021
- Toxic etiology
- Clustering of BA cases in lambs in Australia in 1964, 1988, and 2007 during times of drought, now thought to be related to isoflavonoid toxin isolated from Dysphania plant22
- Use of the isoflavonoid, biliatresone, induces cholangiopathy in zebrafish, mice, and human organoids23242526
- Genetic etiology
- More common in some children with BASM subtype (biliary atresia splenic malformation syndrome)
- Exome sequencing has identified several genes associated with BA
- JAG1 variants found in BA27
- CFC1 mutations are 2 times more frequent in children with BASM than in general population28
- ZIC3 variants, PKD1L1 gene, and ciliary genes are found in some children with BASM2930
- Genome-wide association studies revealed BA susceptibility loci in children with isolated BA
- Chromosome 10q24 (downstream of 2 genes XPNPEP1, involved in metabolism of inflammatory mediators, and ADD3, which encodes the protein adducin 3)3132
- Chromosome 2p16.1 signals intron EFEMP1 (EGF-containing fibulin-like extracellular matrix protein 1)33
- Chromosome 14q21.3-3’ enhancer region for ARF6 (ADP ribosylation factor 6) (mediates epidermal growth factor receptor] signaling and regulates actin cytoskeletal modeling)34
- MAN1A2 regulates ciliogenesis; its expression is reduced in the liver of patients with BA35
- Copy number variants are also found in some patients with BA36
- Chromosome 2q37: heterozygous deletion found in approximately 10% of patients with BA and only in 0.08% of controls
- Deletion at 2q37.3 resulted in deletion of 1 copy of GPC1, a gene that encodes glypican 1, which is a heparan sulfate proteoglycan that regulates hedgehog signaling of inflammation
- More common in some children with BASM subtype (biliary atresia splenic malformation syndrome)
- Immunologic etiology
- Immune dysregulation has been implicated in various studies
- Livers of some infants with BA show activation of genes involved in lymphocyte differentiation, with overexpression of osteopontin and interferon-γ and subsequent dysregulated T-helper 1 immunity37
- Single-nucleotide polymorphisms at CD14 gene, which plays a role in the recognition of bacterial endotoxin, are associated with BA and idiopathic neonatal cholestasis38
- Viral etiology
Risk Factors
- Maternal pregestational diabetes
- Associated with increased risk (adjusted PR, 4.94; 95% CI, 2.32-10.54)739
- Premature birth
- An infant born between 32 and 37 weeks of gestation (adjusted PR, 1.64; 95% confidence interval)
- An infant born before 32 weeks of gestation (adjusted PR, 3.85; 95% CI, 2.38-6.22) and before 37 weeks of gestation7404142
- Small for gestational age increases risk for cholestasis by 2 to 3 times4344
- Female sex
- Reported in some but not all studies (adjusted PR, 1.68; 95% CI, 1.33-2.12)7
- Maternal race
- Non-Hispanic Black patients have a higher risk when compared with non-Hispanic White patients (adjusted PR, 1.66; 95% CI, 1.11-2.49)714
Diagnosis
Approach to Diagnosis
Figure 2. Diagnostic evaluation of neonatal cholestasis.BA, biliary atresia; AR, autosomal recessive; DB, direct bilirubin; CBD, common bile duct ; CSF, cerebrospinal fluid; NBS, newborn screening; MMP7, matrix metalloproteinase 7; PT, prothrombin time; PTT, partial thromboplastin time; SOD, septo-optic dysplasia; UA, urinalysis; US, ultrasonography; UTI, urinary tract infection.
- Jaundice in the neonatal period and early infancy may be benign or serious, requiring emergent management
- Presentation for potentially serious causes of neonatal jaundice may include:
- Jaundice that develops in the first 24 hours after birth or persists for more than 14 to 21 days
- Ill appearance or concerns for infection (eg, fever, hypothermia)
- Cholestatic jaundice is always pathologic; dark urine and acholic stools are characteristic findings associated with cholestatic jaundice
- Presentation for potentially serious causes of neonatal jaundice may include:
- Fractionated bilirubin is the key initial laboratory test to determine the type (direct or indirect) and the degree of hyperbilirubinemia
- With initial diagnostic evaluation, rule out more common diagnoses that may present with neonatal cholestasis, for example, sepsis or α1-antitrypsin deficiency
- When cholestatic jaundice is a clinical concern (or direct hyperbilirubinemia is found on fractionated bilirubin measurement), obtain the following laboratory tests: CBC, PT (prothrombin time) and PTT (partial thromboplastin time), liver profile, and α1-antitrypsin. If concerns for infection (eg, ill appearance, fever, hypothermia), obtain blood, urine, and cerebrospinal fluid cultures
- Tailor further laboratory evaluation to differential diagnosis guided by clinical presentation and based on results of initial screening blood tests
- First imaging test to obtain during workup of direct hyperbilirubinemia is fasting abdominal ultrasonography with a focus on liver and biliary system to differentiate causes of obstructive jaundice
- If after ultrasonography, clinical concern for BA remains, then obtain serum MMP7 (matrix metalloproteinase 7) level
- Gold standard imaging test to confirm BA is intraoperative cholangiogram45
- With improvements in genetic testing and newer studies such as the MMP7, liver biopsy is not always necessary. It can be helpful in instances of diagnostic uncertainty
- Secondary evaluation for commonly associated comorbid conditions may be warranted with studies, including chest radiograph and echocardiogram
- Figure 2 outlines diagnostic approach of neonatal cholestasis
Staging or Classification
- Isolated BA
- Without major malformations
- Most common, accounting for approximately 84% of infants with BA46
- Cytomegalovirus variant
- Variable incidence based on geography and methodologic detection
- 10% to 20% in European series
- Up to 50% in some series from China154748
- Variable incidence based on geography and methodologic detection
- BA with laterality defects
- Also known as BASM 10%1246
- Incidence of BASM is much lower in Asian countries compared with that in North America and Europe49
- Malformations may include:
- Splenic malformations
- Most common anomaly (70%)
- Asplenia, polysplenia, right-sided spleen, or double spleen
- Cardiovascular anomalies
- Dextrocardia, mesocardia, total or partial anomalous pulmonary venous return, absent or interrupted inferior vena cava, anomalous or bilateral superior vena cava, and/or preduodenal portal vein
- Gastrointestinal anomalies
- Abdominal heterotaxy, midline or transverse liver, right-sided stomach, intestinal malrotation, and anomalous or annular pancreas
- History of gestational diabetes may be more prevalent in this group of infants7
- Splenic malformations
- BA without laterality but with at least 1 major malformation46
- Accounts for 6%46
- Malformations can include:
- Cardiovascular anomalies (71%)
- Aortic arch abnormalities, aortic coarctation, atrial septal defects, patent ductus arteriosus, patent foramen ovale, pulmonary artery stenosis, pulmonary valvular stenosis, tetralogy of Fallot, transposition of the great vessels, and ventricular septal defect
- Gastrointestinal anomalies (24%)
- Duodenal or jejunal atresia, esophageal atresia, and imperforate anus
- Genitourinary anomalies (47%)
- Most commonly cystic kidney and hydronephrosis
- Cardiovascular anomalies (71%)
- Cystic BA50
- Onset prenatal
- Accounts for 5% to 10% of some large series
- Can be seen antenatally on maternal ultrasonography
- Has better prognosis, likely due to earlier presentation and intervention48
Workup
History
- Jaundice in newborns is a common symptom. Most often the etiology is benign—either physiologic jaundice or breastmilk jaundice. History consistent with physiologic or breastmilk jaundice includes:
- Breastfed infants
- Onset a few days after birth
- ABO or Rh (Rhesus) incompatibility can suggest hemolytic etiology to jaundice
- Jaundice beyond 14 to 21 days increases concern for a pathologic etiology and warrants further evaluation51
- Jaundice within the first 24 hours of birth suggests a pathologic etiology
- Dark urine and acholic stools are distinctive of cholestasis
- Pale stool color may be noted when using SCC (stool color card) screening tool
- Perinatal factors may contribute to increased risk of neonatal cholestasis
- Premature birth
- Small for gestational age increases risk of cholestasis by 2- to 3-fold4344
- Neonatal hypoxia44
- History of neonatal infection
- Sepsis
- Urinary tract infection
- TORCH (toxoplasmosis, other agents, rubella, cytomegalovirus, herpes simplex)
- Symptoms and historical elements that may suggest a specific etiology for neonatal cholestasis
- Abnormal newborn screening test result may be reported
- May aid in the identification of an etiology for cholestasis (eg, cystic fibrosis, galactosemia, hypothyroidism)
- Parenteral nutrition can suggest cholestasis secondary to parenteral nutrition–associated cholestasis parenteral nutrition–associated cholestasis
- History of septo-optic dysplasia can suggest hypopituitarism52
- Hearing loss may suggest TORCH infection or PFIC (progressive familial intrahepatic cholestasis)
- Vomiting suggests the following:
- Metabolic disease, such as galactosemia
- Bowel obstruction
- Delayed passage of meconium suggests cystic fibrosis
- History of abdominal surgery or necrotizing enterocolitis
- Abnormal newborn screening test result may be reported
- Elements of prenatal history that increase risk of neonatal cholestasis are as follows:
- Cholestasis of pregnancy (suggests maternal heterozygosity for PFIC gene mutations)
- Acute fatty liver of pregnancy (suggests long-chain 3-hydroxyacyl-coenzyme A dehydrogenase deficiency)
- Maternal infections (TORCH)
- Prenatal abnormal imaging results can often identify the etiology of cholestasis
- Growth restriction
- Choledochal cyst
- Bowel anomalies
- Cystic BA
- BASM
- Family history that may increase risk for neonatal cholestasis
- Previous miscarriages or fetal loss suggests GALD (gestational alloimmune liver disease)
- Neonatal cholestasis in parent or siblings can suggest a hereditary etiology
- PFIC
- Cystic fibrosis
- α1-Antitrypsin deficiency
- Alagille syndrome (OMIM #118450)
Physical Examination
- General appearance
- Ill appearance suggests pathologic etiology of jaundice, such as infection, or genetic or metabolic liver disease, such as galactosemia
- Poor growth can be consistent with cystic fibrosis or metabolic or genetic liver disease
- Low tone suggests genetic or metabolic liver disease
- Dysmorphic features suggest Alagille syndrome
- Eyes
- Scleral icterus is consistent with elevated levels of bilirubin
- Cataracts suggest TORCH infections, or genetic or metabolic liver disease
- Inability to fix and follow beyond 3 months of age suggests septo-optic dysplasia
- Cardiac examination
- Heart murmurs in the context of neonatal cholestasis can be consistent with Alagille syndrome, BASM, or congenital heart disease
- Abdominal examination
- Ascites suggests liver failure as seen with GALD
- Hepatomegaly consistent with BA
- Splenomegaly is a later finding in BA
- Umbilical hernia can be seen with hypothyroidism
- Stool inspection
- Integral part of the physical examination in any infant who has jaundice
- Acholic stools are consistent with obstruction of biliary tree
- Acholic stools are present in two-thirds of infants with BA at day of life 3053
- Petechiae and bleeding
- Petechiae suggests possible TORCH infection
- Bleeding can be consistent with vitamin K deficiency which can also be seen in severe liver failure such as GALD and tyrosinemia
Laboratory Tests
- Liver profile
- Total bilirubin
- If level is elevated in the context of a normal direct bilirubin level, it indicates an elevated indirect bilirubin level
- Elevated indirect bilirubin level consistent with ABO or RH incompatibility
- Direct bilirubin
- Surrogate marker of cholestasis in most cases5455
- Indicated in any patient with jaundice beyond 14 days of age
- Levels greater than 1 mg/dL 2 weeks after birth is diagnostic of neonatal cholestasis
- Values exceeding 95% the reference range in the first few days of life is also diagnostic of neonatal cholestasis
- Level uniformly elevated in patients with BA in the first 24 to 48 hours after birth55
- GGT (γ-glutamyltransferase)
- Higher in the neonatal period compared with older children56
- GGT value is usually high in cholestasis
- Generally higher in BA, Alagille syndrome, and α1-antitrypsin deficiency than in other etiologies of neonatal cholestasis57
- Normal GGT level at presentation predicts poor outcome in BA58
- Low or normal GGT level in the context of cholestasis
- Suggests metabolic or inherited disorders, such as PFIC or bile acid synthesis defects59
- AST/ALT levels60
- Elevations suggest liver injury
- Isolated AST elevation
- In instances when AST level is elevated, but total bilirubin, direct bilirubin, and ALT levels are not significantly elevated, hematologic or muscular sources are most likely
- Total bilirubin
- CBC
- Elevated WBC count suggests infection
- Low hemoglobin and RBC count suggest hemolysis
- Thrombocytopenia suggests infection or splenic sequestration
- PT or INR
- May be prolonged with impaired liver synthetic function
- PTT
- Prolongation is usually seen in hematologic disorders but may be prolonged with severe advanced liver disease
- Newborn screening test results
- May help identify genetic causes of liver disease (eg, cystic fibrosis, galactosemia, hypothyroidism)
- TSH and free T4 (thyroxine)
- Both hypothyroidism and hyperthyroidism can cause neonatal cholestasis
- Elevated TSH level with low levels of free T4, consistent with hypothyroidism
- Low TSH level with elevated levels of free T4, consistent with hyperthyroidism
- α1-Antitrypsin phenotype60
- α1-Antitrypsin deficiency can present similarly to BA
- Normal allelic variant PiMM
- PiZZ and PiSZ associated with liver disease
- Patients with BA with non-M allele are listed for transplant earlier; these patients have lower survival with native liver at 24 months61
- MMP7
- Indicated in neonates with cholestasis and elevated GGT level
- A protease expressed in normal cholangiocytes
- Responsible for tissue remodeling
- Associated with liver fibrosis
- Second-tier testing in neonates with high GGT cholestasis
- Levels vary with age, fibrotic disorders, and with congenital heart disease62
- Levels also vary significantly among different enzyme-linked immunosorbent assay kits63 but uniformly elevated in BA
- In a prospective study of 135 infants with cholestasis compared with healthy infants, serum MMP7 values were higher in infants with BA
- Diagnostic sensitivity and specificity were 98.67% and 95.00%, respectively, with a negative predictive value of 98.28% for BA (ROC AUC [area under the receiver operating characteristic curve] 0.99 using a cutoff of 52.85 ng/mL)63
- In another prospective study of 288 patients with neonatal obstructive jaundice, MMP7 demonstrated good accuracy in diagnosing BA (ROC AOC of 0.9829 at a cutoff value of 10.37 ng/mL), with sensitivity, specificity, positive predictive value, and negative predictive values of 95.19%, 93.07%, 97.27%, and 91.43%, respectively64
- In a prospective study of 135 infants with cholestasis compared with healthy infants, serum MMP7 values were higher in infants with BA
- With increasing evidence of its accuracy and specificity, experts note its increasing use in clinical practice
- Turnaround times can take several days
Imaging Studies
- Fasting abdominal ultrasonography
- Initial imaging study to evaluate cholestasis in neonates
- Fasting should be carried out with caution in infants with a higher suspicion for metabolic liver disease
- Identify cystic lesions
- Choledochal cyst
- Cyst in liver hilum as seen with cystic BA
- Visible obstructing lesions of the biliary tree
- Signs with a high specificity for BA65
- Triangular cord sign (obliterated fibrous ductal remnant)
- Gallbladder abnormalities
- Variable sensitivity depending on the operator or institution
- Can identify vascular or splenic abnormalities as seen with BASM
- Evaluate for signs of advanced liver disease, such as splenomegaly, nodular liver, and varices
- Many, but not all, infants with BA have a small or undetectable gallbladder6066
- A normal study does not exclude BA65
- HBS (hepatobiliary scintigraphy)
- HBS uses technetium-99m–labeled iminodiacetic acid derivatives as a radiotracer
- HBS can exclude BA by demonstrating transit of the radiotracer into the bowel
- No need for NPO status for infants to exclude BA
- Administration of phenobarbital for 5 days before the study may enhance biliary excretion of the isotope; its use is center dependent
- Acquire images dynamically up until 1 hour; if no excretion, delayed static imaging should be acquired at 4 hours and at 24 hours postinjection
- Nonsecretion is not specific to BA and can also be seen with other etiologies of cholestatis67
- An appropriately performed study is sensitive with rare false-negative results;68 for example, excretion of radiotracer into the intestine is very helpful to exclude BA; absence of excretion does not in aid in diagnosis
- Magnetic resonance cholecystopancreatography
- Findings consistent with BA67
- Nonvisualization of extrahepatic biliary tree
- Nonvisualization of gallbladder
- Periportal thickening at porta hepatis on T2-weighted images
- Use is center dependent
- Gentle swaddling or sedation is a possible option to avoid movement artifact
- Meta-analysis of 7 studies demonstrated sensitivity of 89% and specificity of 64.7%69
- Findings consistent with BA67
- PTCC (percutaneous transhepatic cholecysto-cholangiography)
- Limited role, at present, because few centers in the United States are using this study70
- Limited expertise
- Invasive
- Alternative to intraoperative cholangiogram
- PTCC is an ultrasonography-guided technique performed under general anesthesia
- Nonionic contrast medium is injected into the gallbladder via liver parenchyma
- Can only be performed when gallbladder lumen is present67
- Might miss an atretic common hepatic duct71
- Is usually done instead of intraoperative cholangiogram, which is considered the gold standard
- Limited role, at present, because few centers in the United States are using this study70
- ERCP (endoscopic retrograde cholangiopancreatography)
- Limited use due to the need of an experienced endoscopist and infant-specific endoscope
- Requires general anesthesia
- ERCP sensitivity ranges between 86% and 100%, with the specificity 79% to 94% (age range: 19-175 days); range of sensitivity and specificity due to interoperator variability7273
Diagnostic Procedures
Figure 3. Histology of BA. Hematoxylin and eosin stain of a liver biopsy of a 3-month-old girl show a proliferation of bile ductules. Bile plugs are present. – BA, biliary atresia.From Lane ER et al. Disorders of the liver. In: Gleason CA et al, eds. Avery’s Diseases of the Newborn. 10th ed. Elsevier; 2018:1098-1112.e2, Figure 77.3.
- Liver biopsy
- Continues to play an important role in the diagnostic workup of infants with BA; however, timing of liver biopsy might evolve with the addition of new diagnostic tests, such as MMP7, and improvements in genetic testing
- Ultrasonography-guided percutaneous core liver biopsy is a safe option in children with a low complication rate of 1.7%74
- Liver biopsy interpretation by an experienced pathologist can provide the correct diagnosis in 90% to 95% of cases in patients with cholestasis7576
- Pathologists are able to diagnose obstruction in 79% to 98% of the cases, with a positive predictive value of 90.7%
- Classic histologic features of biliary obstruction (Figure 3)
- Bile duct proliferation
- Bile plugs
- Portal or perilobular fibrosis
- Edema
- Preservation of the basic hepatic lobular architecture
- Parenteral nutrition–associated liver disease and α1-antitrypsin deficiency have findings similar to those of BA
- α1-Antitrypsin phenotype and history of total parenteral nutrition infusion to differentiate
- Early liver biopsies may not have the classic obstructive signs77
- In many centers, liver biopsy (needle core or surgical wedge biopsy) is obtained at the time of PTCC or intraoperative cholangiogram to exclude BA
- Degree of liver fibrosis can correlate with outcomes
- IOC (intraoperative cholangiogram)
- IOC and histologic examination of the duct remnant are considered the gold standard to diagnose BA60
- A catheter is placed into the cystic duct and dye is injected to visualize patency of the bile ducts proximally into the liver and distally into the duodenum
- If IOC is consistent with an atretic bile duct, then the surgeon proceeds to KPE at the same time
Differential Diagnosis
Table 1. Differential Diagnosis: Neonatal cholestasis.
Condition | Description | Differentiated by |
---|---|---|
Bile duct obstruction | ||
BA | • Neonatal progressive obliterative fibroinflammatory disease of the intra- and extrahepatic biliary tree | • Intraoperative cholangiogram that shows obstruction of biliary tree |
Alagille syndrome | • Autosomal dominant genetic disorder with abnormal bile duct development often associated with cardiac anomalies, triangular facies, or butterfly-shaped vertebra | • Liver biopsy demonstrating few bile ducts and associated cardiac, bony, or facial abnormalities • Genetic testing |
Choledochal cyst | • Congenital cyst in the bile duct | • Isolated abnormality and remainder of biliary tree is normal |
Neonatal sclerosing cholangitis | • Fibrosis of biliary ducts | • Patent biliary tree • Gene sequencing: CLDN1 and DCDC2 can help in diagnosis |
Genetic disorders | ||
Cystic fibrosis | • Genetic disorder of chloride channel that causes thickened secretions affecting the lungs and digestive tracts | • Abnormal newborn screening test result • Abnormal sweat chloride test result |
α1-Antitrypsin deficiency | • Inadequate production of α1-antitrypsin protein | • Low levels of α1-antitrypsin • Confirmed with α1-antitrypsin phenotype or genotype |
Metabolic disorders | ||
Galactosemia | • Inability to break down galactose, which can lead to seizures, vomiting, increased risk of infection, or developmental delay | • Generally identified on newborn screening test results with consistent laboratory findings |
Fatty acid oxidation defects | • Inability to break down fats and can present with weakness, hypotonia, poor growth, or vomiting | • Patients often present with hypoglycemia and identified on newborn screening test results |
Glycogen storage disease | • Inability to use or produce glycogen that can result in hypoglycemia and hepatomegaly | • Tissue biopsy and genetic testing |
Mitochondrial disorders78 | • Abnormalities of the mitochondria with a wide array of symptoms but often with hypotonia, seizures, or developmental delay | • Consistent laboratory studies, including abnormalities in serum or urine organic acid, amino acid, pyruvate, acylcarnitine, and lactate levels, with muscle biopsy for confirmation78 |
Endocrine disorders | ||
Hypothyroidism | • Often presents with lethargy, constipation, feeding difficulty | • Can be identified by newborn screening tests and confirmed with low levels of free T4 |
Panhypopituitarism | • Deficiency of all hormones produced by pituitary gland | • Confirmed by newborn screening tests, blood tests, and brain imaging |
Toxins | ||
Parenteral nutrition–associated cholestasis | • Neonates requiring prolonged parenteral nutrition can develop cholestasis with increasing bilirubin level | • Normal hepatobiliary scintigraphy, cholangiogram, and abdominal ultrasonography |
Infection or immune mediated | ||
TORCH | • Ill-appearing infants | • Serologies |
GALD | • Ill-appearing infants, fetal growth retardation, jaundice, or acute liver failure soon after birth | • MRI of the abdomen • Tissue biopsy demonstrating iron deposits |
Caption: CMV, cytomegalovirus; GALD, gestational alloimmune liver disease; T4, thyroxine; TORCH, toxoplasmosis, other agents, rubella, cytomegalovirus, herpes simplex.
Treatment
Approach to Treatment
- Treatment of BA is primarily surgical and aimed at reestablishing bile flow by HPE (hepatoportoenterostomy)
- Infants who present late can have evidence of cirrhosis at the time of referral and may require primary liver transplant rather than HPE
- In addition to surgery, treatment modalities include:
- Nutritional optimization to meet caloric demands and attain appropriate growth parameters
- Medications aimed at improving bile flow and decrease risk of infection
Initial Treatment Procedures
Kasai Portoenterostomy
Figure 4. Anatomy of KPE. – KPE, Kasai portoenterostomy.From Rogers JL. McCance and Huether’s Pathophysiology: The Biologic Basis for Disease in Adults and Children. 9th ed. Elsevier;2024:chap 42, Figure 42.9.
- HPE procedure that allows for drainage of bile into the small intestine, thereby decreasing liver injury79 (Figure 4)
- Developed by Dr Morio Kasai in the 1950s
- Excision of entire remnant of extrahepatic biliary tree
- A limb of the jejunum is anastomosed to the exposed remaining ductules or the hilum of the liver surface
- Distal duodenum is anastomosed to the jejunal limb, creating the Roux-en-Y configuration
- Most infants with BA undergo KPE
- KPE is not a curative procedure due to the intrahepatic biliary duct involvement of BA
- Previous studies have indicated longer operating times with worse outcomes when the procedure was done laparoscopically
- With technological improvements, procedures done laparoscopically can have equivalent outcomes compared with those done open, often with decreased operative bleeding and shortened duration of time when infants are able to tolerate feeds8081
- Early KPE, ideally before 30 days of life, is associated with improved native liver survival11
Nondrug and Supportive Care
- Nutritional supplementation
- Essential in infants with BA, especially in those with nondraining KPE
- Malnutrition can be due to multifactorial causes82
- Poor caloric intake
- Intolerance to enteral feeding
- Fat malabsorption secondary to insufficient intraluminal bile salts
- Increased caloric demand
- Caloric needs in infants with BA are approximately 130% to 150% of the recommended energy intake for healthy infants and children
- Protein needs are also increased 3 to 4 g/kg/day in infants and 2 to 3 g/kg/day in children83
- Expressed breast milk can be fortified with medium-chain triglyceride oil, which is beneficial because it does not require bile acids for absorption
- Use high medium-chain triglyceride content formulas, which can be concentrated to provide 24 to 27 kcal/oz or more to achieve growth83
- Supplemental feeding by a nasogastric tube is frequently needed in infants with cholestasis and in children with nondraining KPE due to inability to meet their caloric needs
- Gastrostomy tubes are usually avoided, because many patients with BA develop portal hypertension early in their disease course
- Parenteral nutrition can be considered in patients with malnutrition or with complications of chronic liver disease and awaiting liver transplant84
Drug Therapy
- FSV (fat-soluble vitamin) supplementation
- Indicated in all infants with BA
- FSV deficiencies are very common in patients with BA with persistent cholestasis at all time points85
- Supplementation with high-dose FSVs, such as DEKAs Plus, is typically preferred with extra supplementation of specific FSV deficiencies86
- DEKAs Plus liquid dosing: 1 mL PO daily87
- Ursodeoxycholic acid88
- A hydrophilic bile acid
- Although generally recognized as standard of care, it has not been rigorously studied8990
- Increases hydrophilic bile acid pool
- Stabilizes plasma membranes
- Prevents apoptosis
- Acts as an immunomodulator
- Dose: 10-20 mg/kg/day PO divided twice a day
- Prophylactic antibiotics
- Most infants will have at least 1 episode of cholangitis before age 2 years,9192 with the incidence of cholangitis post-KPE between 40% and 90%9394
- Use of IV antibiotics immediately after KPE is standard of care
- Transition to oral prophylactic antibiotics for a minimum of 6 to 12 months after KPE9192 to prevent cholangitis, which may affect long- and short-term outcomes9192
- Prophylactic antibiotic regimens can include:959697
- Trimethoprim-sulfamethoxazole, which is most commonly prescribed
- Neomycin
- Oral penicillins, such as amoxicillin or amoxicillin-clavulanate
- Oral cephalosporins
- Glucocorticoids
- Not clinically indicated
- Use did not improve outcomes
- In a randomized placebo-controlled trial of glucocorticoid treatment in 140 infants with BA given for 13 weeks did not improve bile drainage at 6 and 24 months post-HPE, nor did it improve survival with native liver at age 2 years98
- More adverse events related to impaired growth across all parameters (length, weight, and head circumference) for at least 6 months post-HPE99
- Despite evidence to the contrary, various centers continue to use high-dose glucocorticoid regimens, some demonstrating improved outcome
- Larger cohorts are needed to validate these results
- Immunoglobulin therapy
- Has not been demonstrated to improve outcomes
- In a multicenter, prospective, open-labeled, phase I or IIA trial of IVIg, with 1 g/kg/dose of IVIg infused at 3 to 5, 30, and 60 days post-HPE, there was no trend to lower bilirubin levels or improve 360-day survival in 29 infants with the native liver as compared with historical controls100
Liver Transplant
- Most patients with BA require liver transplant in childhood, with only 20% to 30% entering adulthood with their native liver56
- Indications84
- Nondraining KPE
- Refractory growth failure
- Decompensated cirrhosis manifests as follows:
- Ascites
- Recurrent variceal bleeding
- Hepatopulmonary syndrome: pulmonary vascular dilation and hypoxemia
- Recurrent cholangitis
- Metabolic bone disease
- Hepatocellular carcinoma
- 1- and 5-Year patient survival after liver transplant for patients with BA is excellent at 95% to 97%, respectively101
- Long-term survival after liver transplant in general is excellent, with a projected survival of 80% at 30 years after liver transplant102
- Liver transplant is curative
- After liver transplant, patients require lifelong immunosuppression
Admission Criteria
- At initial presentation, extensive evaluation may necessitate admission
- Other indications for admission include:
- Symptoms of cholangitis, for example, fever, abdominal pain, or vomiting
- Poor growth despite outpatient interventions
- Signs and symptoms of liver failure, for example, confusion or altered mental status, or coagulopathy
- Bleeding varices (because of portal hypertension)
Follow-Up
Monitoring
- FSV (fat-soluble vitamin) levels
- Monitor FSV levels routinely starting at the first month after HPE or at the establishment of diagnosis of BA if late referral
- Low levels of specific vitamins may require individual vitamin supplementation in addition to DEKAs
- If FSV levels are in the target range, then frequency of monitoring can be decreased from every 3 months to 6 months86
- INR
- Indicator of vitamin K deficiency, in addition to liver function
- Every 2 to 4 weeks in infants with cholestasis
- Posttransplant103
- Immediately after transplant, follow-up visits are more frequent, for example, weekly
- Over time, follow-up visits may be less frequent, for example, every few months but at least annually
- Monitor growth. Measure height and weight at each follow-up visit
- Screen for hearing loss at 1 year after transplant
- Monitor immunosuppression levels at each visit
- Serial measurements of liver enzyme and bilirubin levels to assess graft health
Complications
Cholangitis
- Ascending cholangitis is inflammation and infection of bile ducts and is the most common complication in patients after HPE, with incidence as high as 50% to 90% (more common in draining HPE)9394
- Can recur with highest incidence in the first 2 years after HPE
- Appropriate prophylactic antimicrobial treatment is used to decrease the incidence of ascending cholangitis after KPE
- Gram-negative pathogens, such as Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa, are the most frequently reported isolated bacteria
- Of the gram-positive pathogens, Enterococcus species and Staphylococcus aureus are more common
- Candidal fungal infection is less commonly reported92
- Cholangitis can present with the following:
- Fever without a clear source of infection, the most common symptom
- Abdominal pain
- Acholic stools
- Irritability
- Treatment with broad-spectrum IV antibiotics
- Piperacillin-tazobactam
- Ceftriaxone plus metronidazole
- Ampicillin-sulbactam
- In patients with penicillin allergy, quinolones, such as ciprofloxacin, or aminoglycosides with metronidazole can be used
- Recurrent cholangitis may increase risk for progressive liver disease104
- Presence of bile lakes increases risk for cholangitis105106
Portal Hypertension, Variceal Bleeding, and Ascites
- Most children with BA have chronic liver disease and portal hypertension107
- In children with BA, suspect portal hypertension if108
- Thrombocytopenia (platelet count less than 150,000/mm3)
- Splenomegaly (2 cm below costal margin)
- Most common complications of decompensated cirrhosis107
- Ascites
- Variceal bleeding108
- Incidence in children with BA at 5 years is around 10%
Prognosis
- Early identification of cholestatic jaundice improves outcome
- Native liver survival and overall survival rates are inversely correlated with age at KPE and reestablishment of bile flow3109110
- Infants with poor biliary drainage after HPE develop end-stage liver disease and progress to liver transplant within the first 2 to 3 years of life
- Some countries have adopted centralization of surgical procedures at designated centers where expertise and higher caseloads have yielded better long-term outcomes111112
- After KPE, bilirubin levels at 3 months of age in infants with BA correlate with transplant-free survival at 2 years113
- 86% in the patient group with total bilirubin level less than 2 mg/dL
- 20% in the patient group with total bilirubin level more than 2 mg/dL. These patients also are more likely to experience the following:113
- Diminished weight gain
- Ascites
- Hypoalbuminemia
- Coagulopathy
- Death
- Almost all patients (more than 98%) with BA living with their native liver for 5 or more years after KPE have chronic liver disease107
- 20-Year transplant-free survival rate is around approximately 30%.114 All these patients have chronic liver disease and portal hypertension
Referral
- Refer any infant with cholestasis to a pediatric gastroenterologist or hepatologist for appropriate workup
- Refer any infant with BA to a liver transplant center for evaluation and transplant listing if they have persistent cholestasis, growth failure, and/or decompensated cirrhosis
Screening and Prevention
Screening
- SCC programs
- SCC programs give caregivers after birth of their baby a card with color photographs of normal and pale stools in infants. Caregivers are instructed to call a central phone number if they notice pale stools. They are also instructed to return the stool card after 1 month by mail or during health visit
- National screening for BA using SCC program was first implemented in Taiwan, which has one of the highest incidences of BA
- Sensitivity, specificity, and positive predictive value for detecting BA by 60 days of age were 89.7%, 99.9%, and 28.6%, respectively
- Screening program led to more KPs before 60 days of life9
- SCC has also been implemented successfully in other countries, such as Japan, Germany, Switzerland, and Brazil with some variations115116
- Although not widely adopted, an app for stool color using a smartphone’s camera and internal color recognition software has been developed and may be a valuable tool in earlier identification of cholestasis117
References
1.Sundaram SS et al. Biliary atresia: Indications and timing of liver transplantation and optimization of pretransplant care. Liver Transpl. 2017;23(1):96-109.
View In Article|Cross Reference
2.Mysore KR et al. Biliary atresia as a disease starting in utero: implications for treatment, diagnosis, and pathogenesis. J Pediatr Gastroenterol Nutr. 2019;69(4):396-403.
View In Article|Cross Reference
3.Schreiber RA et al. Biliary atresia: the Canadian experience. J Pediatr. 2007;151(6):659-665.e1.
View In Article|Cross Reference
4.Serinet MO et al. Management of patients with biliary atresia in France: results of a decentralized policy 1986-2002. Hepatology. 2006;44(1):75-84.
View In Article|Cross Reference
5.Parolini F et al. Biliary atresia: 20-40-year follow-up with native liver in an Italian centre. J Pediatr Surg. 2019;54(7):1440-1444.
View In Article|Cross Reference
6.Jain V et al. Prognostic markers at adolescence in patients requiring liver transplantation for biliary atresia in adulthood. J Hepatol. 2019;71(1):71-77.
View In Article|Cross Reference
7.Cavallo L et al. The epidemiology of biliary atresia: exploring the role of developmental factors on birth prevalence. J Pediatr. 2022;246:89-94.e2.
View In Article|Cross Reference
8.Jimenez-Rivera C et al. International incidence and outcomes of biliary atresia. J Pediatr Gastroenterol Nutr. 2013;56(4):344-354.
View In Article|Cross Reference
9.Chen SM et al. Screening for biliary atresia by infant stool color card in Taiwan. Pediatrics. 2006;117(4):1147-1154.
View In Article|Cross Reference
10.Vic P et al. Biliary atresia in French Polynesia. Retrospective study of 10 years. Article in French. Arch Pediatr. 1994;1(7):646-651.
View In Article|Cross Reference
11.Hoshino E et al. Age at surgery and native liver survival in biliary atresia: a systematic review and meta-analysis. Eur J Pediatr. 2023;182(6):2693-2704.
View In Article|Cross Reference
12.Davenport M et al. The biliary atresia splenic malformation syndrome: a 28-year single-center retrospective study. J Pediatr. 2006;149(3):393-400.
View In Article|Cross Reference
13.Strickland AD et al. Studies in the etiology of extrahepatic biliary atresia: time-space clustering. J Pediatr. 1982;100(5):749-753.
View In Article|Cross Reference
14.Yoon PW et al. Epidemiology of biliary atresia: a population-based study. Pediatrics. 1997;99(3):376-382.
View In Article|Cross Reference
15.Bezerra JA et al. Biliary atresia: clinical and research challenges for the twenty-first century. Hepatology. September 2018;68(3):1163-1173.
View In Article|Cross Reference
16.Saito T et al. Evidence for viral infection as a causative factor of human biliary atresia. J Pediatr Surg. 2015;50:1398-1404.
View In Article|Cross Reference
17.Zani A. et al. Cytomegalovirus-associated biliary atresia: an aetiological and prognostic subgroup. J Pediatr Surg. 2015;50(10):1739-1745.
View In Article|Cross Reference
18.Xu Y et al. The perinatal infection of cytomegalovirus is an important etiology for biliary atresia in China. Clin Pediatr (Phila). 2012;51:109-113.
View In Article|Cross Reference
19.Brindley SM et al. Cytomegalovirus-specific T-cell reactivity in biliary atresia at the time of diagnosis is associated with deficits in regulatory T cells. Hepatology. 2012;55:1130-1138.
View In Article|Cross Reference
20.Shivakumar P et al. Obstruction of extrahepatic bile ducts by lymphocytes is regulated by IFN-gamma in experimental biliary atresia. J Clin Invest. 2004;114(3):322-329.
View In Article|Cross Reference
21.Bates MD et al. Biliary atresia: pathogenesis and treatment. Semin Liver Dis. 1998;18(3):281-293.
View In Article|Cross Reference
22.Lorent K et al. Identification of a plant isoflavonoid that causes biliary atresia. Sci Transl Med. 2015;7(286):286ra67.
View In Article|Cross Reference
23.Koo KA et al. Biliatresone, a reactive natural toxin from Dysphania glomulifera and D. littoralis: discovery of the toxic moiety 1,2-diaryl-2-propenone. Chem Res Toxicol. 2015;8:1519-1521.
View In Article|Cross Reference
24.Yang Y et al. The synthetic toxin biliatresone causes biliary atresia in mice. Lab Invest. 2020;100:1425-1435.
View In Article|Cross Reference
25.Schmidt HC et al. Biliatresone induces cholangiopathy in C57BL/6J neonates. Sci Rep. June 29, 2023;13(1):10574.
View In Article|Cross Reference
26.Hai-Bing Y et al . Environmental toxin biliatresone-induced biliary atresia-like abnormal cilia and bile duct cell development of human liver organoids. Toxins. March 11, 2024;16(3):144.
View In Article|Cross Reference
27.Kohsaka T et al. The significance of human jagged 1 mutations detected in severe cases of extrahepatic biliary atresia. Hepatology. 2002;36(4 pt 1):904-912.
View In Article|Cross Reference
28.Davit-Spraul A et al. CFC1 gene involvement in biliary atresia with polysplenia syndrome. J Pediatr Gastroenterol Nutr. 2008;46(1):111-112.
View In Article|Cross Reference
29.Berauer JP et al. Identification of polycystic kidney disease 1 like 1 gene variants in children with biliary atresia splenic malformation syndrome. Hepatology. 2019;70(3):899-910.
View In Article|Cross Reference
30.Hellen DJ et al. Liver-restricted deletion of the biliary atresia candidate gene Pkd1l1 causes bile duct dysmorphogenesis and ciliopathy. Hepatology. 2023;77(4):1274-1286.
View In Article|Cross Reference
31.Garcia-Barcelo MM et al. Genome-wide association study identifies a susceptibility locus for biliary atresia on 10q24.2. Hum Mol Genet. 2010;19(14):2917-2925.
View In Article|Cross Reference
32.Tsai EA et al. Replication of a GWAS signal in a Caucasian population implicates ADD3 in susceptibility to biliary atresia. Hum Genet. 2014;133(2):235-243.
View In Article|Cross Reference
33.Chen Y et al. A genome-wide association study identifies a susceptibility locus for biliary atresia on 2p16.1 within the gene EFEMP1. PLoS Genet. 2018;14(8):e1007532.
View In Article|Cross Reference
34.Ningappa M et al. The role of ARF6 in biliary atresia. PLoS One. 2015;10(9):e0138381.
View In Article|Cross Reference
35.So J et al. Biliary-atresia-associated mannosidase-1-alpha-2 gene regulates biliary and ciliary morphogenesis and laterality. Front Physiol. 2020;11:538701.
View In Article|Cross Reference
36.Cui S et al. Evidence from human and zebrafish that GPC1 is a biliary atresia susceptibility gene. Gastroenterology. 2013;144(5):1107-1115.e3.
View In Article|Cross Reference
37.Bezerra JA et al. Genetic induction of proinflammatory immunity in children with biliary atresia. Lancet. 2002;360(9346):1653-1659.
View In Article|Cross Reference
38.Shih HH et al. Promoter polymorphism of the CD14 endotoxin receptor gene is associated with biliary atresia and idiopathic neonatal cholestasis. Pediatrics. 2005;116(2):437-441.
View In Article|Cross Reference
39.Tinker S et al. Specific birth defects in pregnancies of women with diabetes: National Birth Defects Prevention Study, 1997–2011. Am J Obstet Gynecol. February 1, 2020;222:176.e1-176.e11.
View In Article|Cross Reference
40.Chiu CY et al. Biliary atresia in preterm infants in Taiwan: a nationwide survey. J Pediatr. 2013;163(1):100-103.e1.
View In Article|Cross Reference
41.Fischler B et al. A population-based study on the incidence and possible pre-and perinatal etiologic risk factors of biliary atresia. J Pediatr. August 1, 2002;141:217-222.
View In Article|Cross Reference
42.van Wessel D et al. Preterm infants with biliary atresia: a nationwide cohort analysis from the Netherlands. J Pediatr Gastroenterol Nutr. October 1, 2017;65:370-374.
View In Article|Cross Reference
43.Lee SM et al. Parenteral nutrition associated cholestasis is earlier, more prolonged and severe in small for gestational age compared with appropriate for gestational age very low birth weight infants. Yonsei Med J. 2013;54(4):839-844.
View In Article|Cross Reference
44.Teng J et al. Cholestasis after very preterm birth was associated with adverse neonatal outcomes but no significant long-term liver disease: a population-based study. Acta Paediatr. January 2021;110(1):141-148.
View In Article|Cross Reference
45.Alkan M et al. Laparoscopy-assisted percutaneous cholangiography in biliary atresia diagnosis: comparison with open technique. Gastroenterol Res Pract. 2016;2016:5637072.
View In Article|Cross Reference
46.Schwarz KB et al. Extrahepatic anomalies in infants with biliary atresia: results of a large prospective North American multicenter study. Hepatology. 2013;58(5):1724-1731.
View In Article|Cross Reference
47.Xu Y et al. The perinatal infection of cytomegalovirus is an important etiology for biliary atresia in China. Clin Pediatr. 2012;51:109-113.
View In Article|Cross Reference
48.Lakshminarayan B et al. Biliary atresia: a comprehensive review. J Autoimmun. 2016;73:1-9.
View In Article|Cross Reference
49.Yang MC et al. Implication of early-onset biliary atresia and extrahepatic congenital anomalies. Pediatr Int. 2010;52(4):569-572.
View In Article|Cross Reference
50.Caponcelli E et al. Cystic biliary atresia: an etiologic and prognostic subgroup. J Pediatr Surg. 2008;43(9):1619-1624.
View In Article|Cross Reference
51.Gilmour SM. Prolonged neonatal jaundice: when to worry and what to do. Paediatr Child Health. December 2004;9(10):700-704.
View In Article|Cross Reference
52.Vitiello L et al. Pediatric liver diseases and ocular changes: what hepatologists and ophthalmologists should know and share with each other. Dig Liver Dis. January 2020;52(1):1-8.
View In Article|Cross Reference
53.Gu YH et al. Stool color card screening for early detection of biliary atresia and long-term native liver survival: a 19-year cohort study in Japan. J Pediatr. 2015;166(4):897-902.e1.
View In Article|Cross Reference
54.Harpavat S et al. Patients with biliary atresia have elevated direct/conjugated bilirubin levels shortly after birth. Pediatrics. 2011;128(6):e1428-e1433.
View In Article|Cross Reference
55.Harpavat S et al. Diagnostic yield of newborn screening for biliary atresia using direct or conjugated bilirubin measurements. JAMA. 2020;323(12):1141-1150.
View In Article|Cross Reference
56.Hirfanoglu IM et al. Analysis of serum gamma-glutamyl transferase levels in neonatal intensive care unit patients. J Pediatr Gastroenterol Nutr. 2014;58(1):99-101.
View In Article|Cross Reference
57.Chen X et al. Value of gamma-glutamyl transpeptidase for diagnosis of biliary atresia by correlation with age. J Pediatr Gastroenterol Nutr. 2016;63(3):370-373.
View In Article|Cross Reference
58.Shankar S et al. Normal gamma glutamyl transferase levels at presentation predict poor outcome in biliary atresia. J Pediatr Gastroenterol Nutr. 2020;70(3):350-355.
View In Article|Cross Reference
59.Chen HL et al. Panel-based next-generation sequencing for the diagnosis of cholestatic genetic liver diseases: clinical utility and challenges. J Pediatr. 2019;205:153-159.e6.
View In Article|Cross Reference
60.Fawaz R et al. Guideline for the evaluation of cholestatic jaundice in infants: joint recommendations of the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition and the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition. J Pediatr Gastroenterol Nutr. 2017;64(1):154-168.
View In Article|Cross Reference
61.Campbell KM et al. High prevalence of alpha-1-antitrypsin heterozygosity in children with chronic liver disease. J Pediatr Gastroenterol Nutr. 2007;44(1):99-103.
View In Article|Cross Reference
62.Wu JF et al. Transient elastography is useful in diagnosing biliary atresia and predicting prognosis after hepatoportoenterostomy. Hepatology. 2018;68(2):616-624.
View In Article|Cross Reference
63.Yang L et al. Diagnostic accuracy of serum matrix metalloproteinase-7 for biliary atresia. Hepatology. 2018;68(6):2069-2077.
View In Article|Cross Reference
64.Jiang J et al. Serum MMP-7 in the diagnosis of biliary atresia. Pediatrics. 2019;144(5):e20190902.
View In Article|Cross Reference
65.Napolitano M et al. Practical approach to imaging diagnosis of biliary atresia, part 1: prenatal ultrasound and magnetic resonance imaging, and postnatal ultrasound. Pediatr Radiol. 2021;51(2):314-331.
View In Article|Cross Reference
66.Farrant P et al. Mieli-Vergani, improved diagnosis of extraheptic biliary atresia by high frequency ultrasound of the gall bladder. Br J Radiol. 2001;74(886):952-954.
View In Article|Cross Reference
67.Napolitano M et al. Practical approach for the diagnosis of biliary atresia on imaging, part 2: magnetic resonance cholecystopancreatography, hepatobiliary scintigraphy, percutaneous cholecysto-cholangiography, endoscopic retrograde cholangiopancreatography, percutaneous liver biopsy, risk scores and decisional flowchart. Pediatr Radiol. 2021;51(8):1545-1554.
View In Article|Cross Reference
68.Kianifar HR et al. Accuracy of hepatobiliary scintigraphy for differentiation of neonatal hepatitis from biliary atresia: systematic review and meta-analysis of the literature. Pediatr Radiol. 2013;43(8):905-919.
View In Article|Cross Reference
69.He JP et al. Comparison of different noninvasive diagnostic methods for biliary atresia: a meta-analysis. World J Pediatr. 2016;12(1):35-43.
View In Article|Cross Reference
70.Parra D et al. Findings in percutaneous trans-hepatic cholecysto-cholangiography in neonates and infants presenting with conjugated hyperbilirubinemia: emphasis on differential diagnosis and cholangiographic patterns. BMC Pediatr. January 14, 2023;23(1):22.
View In Article|Cross Reference
71.Jensen MK et al. HIDA, percutaneous transhepatic cholecysto-cholangiography and liver biopsy in infants with persistent jaundice: can a combination of PTCC and liver biopsy reduce unnecessary laparotomy? Pediatr Radiol. 2012;42(1):32-39.
View In Article|Cross Reference
72.Negm AA et al. The role of endoscopic retrograde cholangiopancreatography in the diagnosis of biliary atresia: 14 years’ experience. Eur J Pediatr Surg. 2018;28(3):261-267.
View In Article|Cross Reference
73.Shanmugam NP et al. Selective use of endoscopic retrograde cholangiopancreatography in the diagnosis of biliary atresia in infants younger than 100 days. J Pediatr Gastroenterol Nutr. 2009;49(4):435-441.
View In Article|Cross Reference
74.Govender P et al. Sonography-guided percutaneous liver biopsies in children. AJR Am J Roentgenol. 2013;201(3):645-650.
View In Article|Cross Reference
75.Russo P et al. Design and validation of the biliary atresia research consortium histologic assessment system for cholestasis in infancy. Clin Gastroenterol Hepatol. 2011;9(4):357-362.e2.
View In Article|Cross Reference
76.Morotti RA et al. Pediatric cholestatic disorders: approach to pathologic diagnosis. Surg Pathol Clin. 2013;6(2):205-225.
View In Article|Cross Reference
77.Azar G et al. Atypical morphologic presentation of biliary atresia and value of serial liver biopsies. J Pediatr Gastroenterol Nutr. 2002;34(2):212-215.
View In Article|Cross Reference
78.Koenig MK. Presentation and diagnosis of mitochondrial disorders in children. Pediatr Neurol. May 2008;38(5):305-313.
View In Article|Cross Reference
79.Davenport M et al. Surgery for biliary atresia–is there a European consensus? Eur J Pediatr Surg, 2007;17(3):180-183.
View In Article|Cross Reference
80.Hussain MH et al. Outcomes of laparoscopic Kasai portoenterostomy for biliary atresia: a systematic review. J Pediatr Surg. 2017;52(2):264-267.
View In Article|Cross Reference
81.Okur MH et al. Laparoscopic versus open portoenterostomy for treatment of biliary atresia: a meta-analysis. Pediatr Surg Int. 2023;39(1):148.
View In Article|Cross Reference
82.Boster JM et al. Malnutrition in biliary atresia: assessment, management, and outcomes. Liver Transpl. 2022;28(3):483-492.
View In Article|Cross Reference
83.Novy MA et al. Nutritional considerations and management of the child with liver disease. Nutrition. 1997;13(3):177-184.
View In Article|Cross Reference
84.Squires RH et al. Evaluation of the pediatric patient for liver transplantation: 2014 practice guideline by the American Association for the Study of Liver Diseases, American Society of Transplantation and the North American Society for Pediatric Gastroenterology, Hepatology and Nutrition. Hepatology. 2014;60(1):362-398.
View In Article|Cross Reference
85.Shneider BL et al. Efficacy of fat-soluble vitamin supplementation in infants with biliary atresia. Pediatrics. 2012;130(3):e607-e614.
View In Article|Cross Reference
86.Kamath BM et al. Fat soluble vitamin assessment and supplementation in cholestasis. Clin Liver Dis. 2022;26(3):537-553.
View In Article|Cross Reference
87.DEKAs plus liquid dietary supplement–multivitamin and mineral dietary supplement with delivery technology: orange-peach-mango flavored. Callion Pharma; 2024. Accessed August 2, 2024.
View In Article|Cross Reference
88.Trauner M et al. Graziadei, review article: mechanisms of action and therapeutic applications of ursodeoxycholic acid in chronic liver diseases. Aliment Pharmacol Ther. 1999;13(8):979-996.
View In Article|Cross Reference
89.Meyers RL et al. High-dose steroids, ursodeoxycholic acid, and chronic intravenous antibiotics improve bile flow after Kasai procedure in infants with biliary atresia. J Pediatr Surg. 2003;38(3):406-411.
View In Article|Cross Reference
90.Stringer MD et al. Kasai portoenterostomy: 12-year experience with a novel adjuvant therapy regimen. J Pediatr Surg. 2007;42(8):1324-1328.
View In Article|Cross Reference
91.Cheng K et al. Cholangitis in patients with biliary atresia receiving hepatoportoenterostomy: a national database study. J Pediatr Gastroenterol Nutr. 2020;71(4):452-458.
View In Article|Cross Reference
92.Baek SH et al. The epidemiology and etiology of cholangitis after Kasai portoenterostomy in patients with biliary atresia. J Pediatr Gastroenterol Nutr. 2020;70(2):171-177.
View In Article|Cross Reference
93.Rothenberg SS et al. Cholangitis after the Kasai operation for biliary atresia. J Pediatr Surg. 1989;24(8):729-732.
View In Article|Cross Reference
94.Davenport M et al. Biliary atresia: the King’s College Hospital experience (1974-1995). J Pediatr Surg. 1997;32(3):479-485.
View In Article|Cross Reference
95.Bu LN et al. Prophylactic oral antibiotics in prevention of recurrent cholangitis after the Kasai portoenterostomy. J Pediatr Surg. 2003;38(4):590-593.
View In Article|Cross Reference
96.de Vries W et al. Biliary atresia in the Netherlands: outcome of patients diagnosed between 1987 and 2008. J Pediatr. 2012;160(4):638-644.e2.
View In Article|Cross Reference
97.Calinescu AM et al. Cholangitis definition and treatment after Kasai hepatoportoenterostomy for biliary atresia: a Delphi process and international expert panel. J Clin Med. January 19,2022;11(3):494.
View In Article|Cross Reference
98.Bezerra JA et al. Use of corticosteroids after hepatoportoenterostomy for bile drainage in infants with biliary atresia: the START randomized clinical trial. JAMA. 2014;311(17):1750-1759.
View In Article|Cross Reference
99.Alonso EM et al. Impact of steroid therapy on early growth in infants with biliary atresia: the multicenter steroids in biliary atresia randomized trial. J Pediatr. 2018;202:179-185.e4.
View In Article|Cross Reference
100.Mack CL et al. A phase I/IIa trial of intravenous immunoglobulin following portoenterostomy in biliary atresia. J Pediatr Gastroenterol Nutr. 2019;68(4):495-501.
View In Article|Cross Reference
101.Arnon R et al. Liver transplantation for biliary atresia: is there a difference in outcome for infants? J Pediatr Gastroenterol Nutr. 2016;62(2):220-225.
View In Article|Cross Reference
102.Bowring MG et al. Projected 20- and 30-year outcomes for pediatric liver transplant recipients in the United States. J Pediatr Gastroenterol Nutr. 2020;70(3):356-363.
View In Article|Cross Reference
103.Kelly DA et al. Long-term medical management of the pediatric patient after liver transplantation: 2013 practice guideline by the American Association for the Study of Liver Diseases and the American Society of Transplantation. Liver Transpl. August 2013;19(8):798-825.
View In Article|Cross Reference
104.Luo Y et al. Current concept about postoperative cholangitis in biliary atresia. World J Pediatr. 2008;4(1):14-19.
View In Article|Cross Reference
105.Jiang J et al. Intrahepatic cystic lesions in children with biliary atresia after Kasai procedure. J Pediatr Surg. 2019;54(12):2565-2569.
View In Article|Cross Reference
106.Ginstrom DA et al. Biliary atresia-associated cholangitis: the central role and effective management of bile lakes. J Pediatr Gastroenterol Nutr. 2019;68(4):488-494.
View In Article|Cross Reference
107.Ng VL et al. Medical status of 219 children with biliary atresia surviving long-term with their native livers: results from a North American multicenter consortium. J Pediatr. 2014;165(3):539-546.e2.
View In Article|Cross Reference
108.Bas LM et al . Risk of variceal hemorrhage and pretransplant mortality in children with biliary atresia. Hepatology. September 2022;76(3):712-726.
View In Article|Cross Reference
109.Carvalho E et al. Biliary atresia: the Brazilian experience. J Pediatr (Rio J). 2010;86(6):473-479.
View In Article|Cross Reference
110.Serinet MO et al. Impact of age at Kasai operation on its results in late childhood and adolescence: a rational basis for biliary atresia screening. Pediatrics. 2009;123(5):1280-126.
View In Article|Cross Reference
111.McKiernan PJ et al. British paediatric surveillance unit study of biliary atresia: outcome at 13 years. J Pediatr Gastroenterol Nutr. 2009;48(1):78-81.
View In Article|Cross Reference
112.McKiernan PJ et al. The frequency and outcome of biliary atresia in the UK and Ireland. Lancet. 2000;355(9197):25-29.
View In Article|Cross Reference
113.Shneider BL et al. Total serum bilirubin within 3 months of hepatoportoenterostomy predicts short-term outcomes in biliary atresia. J Pediatr. 2016;170:211-217.e1-e2.
View In Article|Cross Reference
114.de Vries W et al. Twenty-year transplant-free survival rate among patients with biliary atresia. Clin Gastroenterol Hepatol. 2011;9(12):1086-1091.
View In Article|Cross Reference
115.Madadi-Sanjani O. et al. Home-based color card screening for biliary atresia: the first steps for implementation of a nationwide newborn screening in Germany. Pediatr Surg Int. 2019;35(11):1217-1222.
View In Article|Cross Reference
116.Borgeat M et al. Newborn biliary atresia screening with the stool colour card: a questionnaire survey of parents. BMJ Paediatr Open. 2018;2(1):e000269.
View In Article|Cross Reference
117.Franciscovich A et al. PoopMD, a mobile health application, accurately identifies infant acholic stools. PLoS One. 2015;10(7):e0132270.
View In Article|Cross Reference