2614 PART 10 Disorders of the Gastrointestinal System
■ IMMUNOPATHOGENESIS
The weight of evidence suggests that the progressive liver injury in
patients with autoimmune hepatitis is the result of a cell-mediated
immunologic attack directed against liver cells in the setting of a loss
of, or failed, immunologic tolerance for self-liver antigens. In all likelihood, predisposition to autoimmunity is inherited, whereas the liver
specificity of this injury is triggered by environmental (e.g., chemical,
drug [e.g., minocycline], or viral) factors. For example, patients have
been described in whom apparently self-limited cases of acute hepatitis
A, B, or C led to autoimmune hepatitis, presumably because of genetic
susceptibility or predisposition. Evidence to support an autoimmune
pathogenesis in this type of hepatitis includes the following: (1) in the
liver, the histopathologic lesions are composed predominantly of cytotoxic T cells and plasma cells; (2) circulating autoantibodies (nuclear,
smooth muscle, thyroid, etc.; see below), rheumatoid factor, and hyperglobulinemia are common; (3) other autoimmune disorders—such as
autoimmune thyroiditis, rheumatoid arthritis, autoimmune hemolytic
anemia, ulcerative colitis, membranoproliferative glomerulonephritis, juvenile diabetes mellitus, vitiligo, celiac disease, and Sjögren’s
syndrome—occur with increased frequency in patients who have autoimmune hepatitis and in their relatives; (4) histocompatibility haplotypes associated with autoimmune diseases, such as HLA-B1, B8, DR3,
and DR4 as well as extended haplotype DRB1*
0301 and DRB1*
0401
alleles, are common in patients with autoimmune hepatitis; and (5) this
type of chronic hepatitis is responsive to glucocorticoid/immunosuppressive therapy, effective in a variety of autoimmune disorders.
Cellular immune mechanisms appear to be important in the pathogenesis of autoimmune hepatitis. In vitro studies have suggested that
in patients with this disorder, CD4+ T lymphocytes are capable of
becoming sensitized to hepatocyte membrane proteins and of destroying liver cells. Molecular mimicry by cross-reacting antigens that contain epitopes similar to liver antigens is postulated to activate these T
cells, which infiltrate, and result in injury to, the liver. Abnormalities
of immunoregulatory control over cytotoxic lymphocytes (impaired
regulatory CD4+CD25+ T-cell influences) may play a role as well.
Studies of genetic predisposition to autoimmune hepatitis demonstrate
that certain haplotypes are associated with the disorder, as enumerated above, as are polymorphisms in cytotoxic T lymphocyte antigens
(CTLA-4) and tumor necrosis factor α (TNFA*
2). The precise triggering factors, genetic influences, and cytotoxic and immunoregulatory
mechanisms involved in this type of liver injury remain incompletely
defined.
Intriguing clues into the pathogenesis of autoimmune hepatitis come
from the observation that circulating autoantibodies are prevalent in
patients with this disorder. Among the autoantibodies described in
these patients are antibodies to nuclei (so-called antinuclear antibodies
[ANAs], primarily in a homogeneous pattern) and smooth muscle
(so-called anti-smooth-muscle antibodies, directed at actin, vimentin,
and skeletin), antibodies to F-actin, anti-LKM (see below), antibodies
to “soluble liver antigen” (directed against a uracil-guanine-adenine
transfer RNA suppressor protein), antibodies to α-actinin, and antibodies to the liver-specific asialoglycoprotein receptor (or “hepatic
lectin”) and other hepatocyte membrane proteins. Although some of
these provide helpful diagnostic markers, their involvement in the
pathogenesis of autoimmune hepatitis has not been established.
Humoral immune mechanisms have been shown to play a role in the
extrahepatic manifestations of autoimmune and idiopathic hepatitis.
Arthralgias, arthritis, cutaneous vasculitis, and glomerulonephritis
occurring in patients with autoimmune hepatitis appear to be mediated
by the deposition of circulating immune complexes in affected tissue
vessels, followed by complement activation, inflammation, and tissue
injury. While specific viral antigen-antibody complexes can be identified in acute and chronic viral hepatitis, the nature of the immune
complexes in autoimmune hepatitis has not been defined.
■ CLINICAL FEATURES
Many of the clinical features of autoimmune hepatitis are similar to
those described for chronic viral hepatitis. The onset of disease may
be insidious or abrupt; the disease may present initially like, and be
confused with, acute viral hepatitis; a history of recurrent bouts of what
had been labeled acute hepatitis is not uncommon. In approximately a
quarter of patients, the diagnosis is made in the absence of symptoms,
based on abnormal liver laboratory tests. A subset of patients with
autoimmune hepatitis has distinct features. Such patients are predominantly young to middle-aged women with marked hyperglobulinemia
and high titer circulating ANAs. This is the group with positive lupus
erythematosus (LE) preparations (initially labeled lupoid hepatitis)
in whom other autoimmune features are common. Fatigue, malaise,
anorexia, amenorrhea, acne, arthralgias, and jaundice are common.
Occasionally, arthritis, maculopapular eruptions (including cutaneous
vasculitis), erythema nodosum, colitis, pleuritis, pericarditis, anemia, azotemia, and sicca syndrome (keratoconjunctivitis, xerostomia)
occur. In some patients, complications of cirrhosis, such as ascites and
edema (associated with portal hypertension and hypoalbuminemia),
encephalopathy, hypersplenism, coagulopathy, or variceal bleeding
may bring the patient to initial medical attention.
The course of autoimmune hepatitis may be variable. In patients
with mild disease or limited histologic lesions (e.g., piecemeal necrosis [inflammation and erosion of the limiting place of periportal
hepatocytes] without bridging), progression to cirrhosis is limited,
but, even in this subset, clinical monitoring is important to identify
progression; up to half left untreated can progress to cirrhosis over
the course of 15 years. In North America, cirrhosis at presentation
is more common in African Americans than in whites. In those with
severe symptomatic autoimmune hepatitis (aminotransferase levels
>10 times normal, marked hyperglobulinemia, “aggressive” histologic
lesions—bridging necrosis or multilobular collapse, cirrhosis), the
6-month mortality without therapy may be as high as 40%. Such severe
disease accounts for only 20% of cases; the natural history of milder
disease is variable, often accentuated by spontaneous remissions and
exacerbations. In a 10-year (2006–2016) national Dutch study, mortality in patients with autoimmune hepatitis was higher than that of the
general population only in patients with cirrhosis; for patients without
cirrhosis, survival was comparable to that of the general population.
Especially poor prognostic signs include the presence histologically of
multilobular collapse at the time of initial presentation and failure of
serum bilirubin to improve after 2 weeks of therapy. Death may result
from hepatic failure, hepatic coma, other complications of cirrhosis
(e.g., variceal hemorrhage), and intercurrent infection. In patients with
established cirrhosis, HCC may be a late complication (Chap. 82) but
occurs less frequently than in cirrhosis associated with viral hepatitis.
Laboratory features of autoimmune hepatitis are similar to those
seen in chronic viral hepatitis. Liver biochemical tests are invariably
abnormal but may not correlate with the clinical severity or histopathologic features in individual cases. Many patients with autoimmune hepatitis have normal serum bilirubin, alkaline phosphatase,
and globulin levels with only minimal aminotransferase elevations.
Serum AST and ALT levels are increased and fluctuate in the range
of 100−1000 units. In severe cases, the serum bilirubin level is moderately elevated (51−171 μmol/L [3−10 mg/dL]). Hypoalbuminemia
occurs in patients with very active or advanced disease. Serum alkaline
phosphatase levels may be moderately elevated or near normal. In
a small proportion of patients, marked elevations of alkaline phosphatase activity occur; in such patients, clinical and laboratory features
overlap with those of primary biliary cholangitis (Chap. 344). The
prothrombin time is often prolonged, particularly late in the disease or
during active phases.
Polyclonal hypergammaglobulinemia (>2.5 g/dL) is common in
autoimmune hepatitis, as is the presence of rheumatoid factor. As noted
above, circulating autoantibodies are also prevalent, most characteristically ANAs in a homogeneous staining pattern. Smooth-muscle antibodies are less specific, seen just as frequently in chronic viral hepatitis.
Because of the high levels of globulins achieved in the circulation of
some patients with autoimmune hepatitis, occasionally the globulins
may bind nonspecifically in solid-phase binding immunoassays for
viral antibodies. This has been recognized most commonly in tests for
antibodies to HCV, as noted above. In fact, studies of autoantibodies
in autoimmune hepatitis have led to the recognition of new categories
2615Chronic Hepatitis CHAPTER 341
of autoimmune hepatitis. Type I autoimmune hepatitis is the classic
syndrome prevalent in North America and northern Europe occurring
in young women, associated with marked hyperglobulinemia, lupoid
features, circulating ANAs, and HLA-DR3 or HLA-DR4 (especially
B8-DRB1*
03). Also associated with type I autoimmune hepatitis are
autoantibodies against actin and atypical perinuclear antineutrophilic
cytoplasmic antibodies (pANCA). Included in the spectrum of type I
autoimmune hepatitis is a subset of patients who lack ANA and antiLKM1 but who have circulating antibodies to soluble liver antigen.
Most of these patients are women and have clinical features similar
to, or perhaps more severe than, those of other patients with type I
autoimmune hepatitis.
Type II autoimmune hepatitis, often seen in children, more common
in Mediterranean populations, and linked to HLA-DRB1 and HLADQB1 haplotypes, is associated not with ANA but with anti-LKM.
Actually, anti-LKM represent a heterogeneous group of antibodies.
In type II autoimmune hepatitis, the antibody is anti-LKM1, directed
against cytochrome P450 2D6. This is the same anti-LKM seen in some
patients with chronic hepatitis C. Anti-LKM2 is seen in drug-induced
hepatitis, and anti-LKM3 (directed against uridine diphosphate glucuronyltransferases) is seen in patients with chronic hepatitis D. Another
autoantibody observed in type II autoimmune hepatitis is directed
against liver cytosol formiminotransferase cyclodeaminase (anti-liver
cytosol 1).
Liver biopsy abnormalities are similar to those described for chronic
viral hepatitis. Expanding portal tracts and extending beyond the plate
of periportal hepatocytes into the parenchyma (designated interface
hepatitis or piecemeal necrosis) is a mononuclear cell infiltrate that,
in autoimmune hepatitis, may include the presence of plasma cells.
Necroinflammatory activity characterizes the lobular parenchyma,
and evidence of hepatocellular regeneration is reflected by “rosette”
formation, the occurrence of thickened liver cell plates, and regenerative “pseudolobules.” Septal fibrosis, bridging fibrosis, and cirrhosis are
frequent. In patients with early autoimmune hepatitis presenting as an
acute-hepatitis-like illness, lobular and centrilobular (as opposed to the
more common periportal) necrosis has been reported. Bile duct injury
and granulomas are uncommon; however, a subgroup of patients
with autoimmune hepatitis has histologic, biochemical, and serologic
features overlapping those of primary biliary cholangitis (Chap. 344).
■ DIAGNOSTIC CRITERIA
An international group has suggested a set of criteria for establishing a diagnosis of autoimmune hepatitis. Exclusion of liver disease
caused by genetic disorders, viral hepatitis, drug hepatotoxicity, and
alcohol is linked with such inclusive diagnostic criteria as hyperglobulinemia, autoantibodies, and characteristic histologic features. This
international group has also suggested a comprehensive diagnostic
scoring system that, rarely required for typical cases, may be helpful
when typical features are not present. Factors that weigh in favor of
the diagnosis include female gender; predominant aminotransferase
elevation; presence and level of globulin elevation; presence of nuclear,
smooth-muscle, LKM1, and other autoantibodies; concurrent other
autoimmune diseases; characteristic histologic features (interface
hepatitis, plasma cells, rosettes); HLA-DR3 or DR4 markers; and
response to treatment (see below). A more simplified, more specific
scoring system relies on four variables: autoantibodies, serum IgG
level, typical or compatible histologic features, and absence of viral
hepatitis markers. Weighing against the diagnosis are predominant
alkaline phosphatase elevation, mitochondrial antibodies, markers of
viral hepatitis, history of hepatotoxic drugs or excessive alcohol, histologic evidence of bile duct injury, or such atypical histologic features as
fatty infiltration, iron overload, and viral inclusions.
■ DIFFERENTIAL DIAGNOSIS
Early during the course of chronic hepatitis, autoimmune hepatitis may
resemble typical acute viral hepatitis (Chap. 339). Without histologic
assessment, severe chronic hepatitis cannot be readily distinguished
based on clinical or biochemical criteria from mild chronic hepatitis.
In adolescence, Wilson’s disease (Chaps. 344 and 415) may present
with features of chronic hepatitis long before neurologic manifestations
become apparent and before the formation of Kayser-Fleischer rings
(copper deposition in Descemet’s membrane in the periphery of the
cornea). In this age group, serum ceruloplasmin and serum and urinary copper determinations plus measurement of liver copper levels
establish the correct diagnosis. Postnecrotic or cryptogenic cirrhosis
and primary biliary cholangitis (Chap. 344) share clinical features
with autoimmune hepatitis, and both alcoholic hepatitis (Chap. 342)
and nonalcoholic steatohepatitis (Chap. 343) may present with many
features common to autoimmune hepatitis; historic, biochemical, serologic, and histologic assessments are usually sufficient to allow these
entities to be distinguished from autoimmune hepatitis. Of course,
the distinction between autoimmune and chronic viral hepatitis is
not always straightforward, especially when viral antibodies occur
in patients with autoimmune disease or when autoantibodies occur
in patients with viral disease. Furthermore, the presence of extrahepatic features such as arthritis, cutaneous vasculitis, or pleuritis—not
to mention the presence of circulating autoantibodies—may cause
confusion with rheumatologic disorders such as rheumatoid arthritis
and systemic LE. The existence of clinical and biochemical features of
progressive necroinflammatory liver disease distinguishes chronic hepatitis from these other disorders, which are not associated with severe
liver disease. Rarely, hepatic venous outflow obstruction (Budd-Chiari
syndrome) may present with features suggestive of autoimmune hepatitis, but painful hepatomegaly, ascites, and vascular imaging provide
distinguishing diagnostic clues. Other diagnostic considerations would
include celiac disease and ischemic liver disease, which would be
readily distinguishable by clinical and laboratory features from autoimmune hepatitis.
In patients treated with immune checkpoint inhibitors for malignancy, the liver may be one of the autoimmune targets of therapy;
the syndrome resembles autoimmune hepatitis in clinical features
and response to glucocorticoid-based treatment. Finally, occasionally,
features of autoimmune hepatitis overlap with features of autoimmune
biliary disorders such as primary biliary cholangitis, primary sclerosing
cholangitis (Chaps. 344 and 346), or, even more rarely, mitochondrial
antibody-negative autoimmune cholangitis. Such overlap syndromes
are difficult to categorize, and often response to therapy may be the
distinguishing factor that establishes the diagnosis.
TREATMENT
Autoimmune Hepatitis
The mainstay of management in autoimmune hepatitis is glucocorticoid therapy. Several controlled clinical trials have documented
that such therapy leads to symptomatic, clinical, biochemical, and
histologic improvement as well as increased survival. A therapeutic
response can be expected in up to 80% of patients. Unfortunately,
therapy has not been shown in clinical trials to prevent ultimate
progression to cirrhosis; however, instances of reversal of fibrosis
and cirrhosis have been reported in patients responding to treatment, and rapid treatment responses within 1 year do translate into
a reduction in progression to cirrhosis. Although some advocate
the use of prednisolone (the hepatic metabolite of prednisone),
prednisone is just as effective and is favored by most authorities.
Therapy may be initiated at 20 mg/d, but a popular regimen in
the United States relies on an initiation dose of 60 mg/d. This high
dose is tapered successively over the course of a month down to
a maintenance level of 20 mg/d. An alternative, but equally effective, more appealing approach is to begin with half the prednisone
dose (30 mg/d) along with azathioprine (50 mg/d). With azathioprine maintained at 50 mg/d, the prednisone dose is tapered over
the course of a month down to a maintenance level of 10 mg/d.
The advantage of the combination approach is a reduction, over the
span of an 18-month course of therapy, in serious, life-threatening
complications of steroid therapy (e.g., cushingoid features, hypertension, diabetes, osteoporosis) from 66% down to <20%. Genetic
analysis for thiopurine S-methyltransferase allelic variants does not
2616 PART 10 Disorders of the Gastrointestinal System
correlate with azathioprine-associated cytopenias or efficacy and
is not assessed routinely in patients with autoimmune hepatitis.
In combination regimens, 6-mercaptopurine may be substituted
for its prodrug azathioprine, but this is rarely required. Azathioprine alone, however, is not effective in achieving remission, nor
is alternate-day glucocorticoid therapy. Limited experience with
budesonide in noncirrhotic patients suggests that this steroid side
effect−sparing drug may be effective; however, the few randomized
controlled trials of budesonide have not consistently shown efficacy. Although therapy has been shown to be effective for severe
autoimmune hepatitis (AST ≥10× the upper limit of normal or
≥5× the upper limit of normal in conjunction with serum globulin
greater than or equal to twice normal; bridging necrosis or multilobular necrosis on liver biopsy; presence of symptoms), therapy is
not indicated for mild forms of chronic hepatitis, and the efficacy
of therapy in mild or asymptomatic autoimmune hepatitis has not
been established.
Improvement of fatigue, anorexia, malaise, and jaundice tends
to occur within days to several weeks; biochemical improvement
occurs over the course of several weeks to months, with a fall in
serum bilirubin and globulin levels and an increase in serum albumin. Serum aminotransferase levels usually drop promptly, but
improvements in AST and ALT alone do not appear to be reliable
markers of recovery in individual patients; histologic improvement,
characterized by a decrease in mononuclear infiltration and in
hepatocellular necrosis, may be delayed for 6−24 months. Still, if
interpreted cautiously, aminotransferase levels are valuable indicators of relative disease activity, and, although recommended,
many authorities do not advocate for serial liver biopsies to assess
therapeutic success or to guide decisions to alter or stop therapy.
Rapidity of response is more common in older patients (≥69 years)
and those with HLA DBR1*
04; although rapid responders may
progress less slowly to cirrhosis and liver transplantation, they are
no less likely than slower responders to relapse after therapy. Therapy should continue for at least 12−18 months. After tapering and
cessation of therapy, the likelihood of relapse is at least 50%, even if
posttreatment histology has improved to show mild chronic hepatitis, and most patients require therapy at maintenance doses indefinitely. Continuing azathioprine alone (2 mg/kg body weight daily)
after cessation of prednisone therapy has been shown to reduce
the frequency of relapse. Long-term maintenance with low-dose
prednisone (≤10 mg daily) has also been shown to keep autoimmune hepatitis in check without the theoretical risk of azathioprine
marrow suppression and, in young women of child-bearing age,
teratogenicity; however, maintenance azathioprine is more effective
in preserving remission.
In medically refractory cases, an attempt should be made to
intensify treatment with high-dose glucocorticoid monotherapy
(60 mg daily) or combination glucocorticoid (30 mg daily) plus
high-dose azathioprine (150 mg daily) therapy. After a month,
doses of prednisone can be reduced by 10 mg a month, and doses
of azathioprine can be reduced by 50 mg a month toward ultimate,
conventional maintenance doses. Patients refractory to this regimen
may be treated with cyclosporine, tacrolimus, or mycophenolate
mofetil. Similarly, in exploratory studies, infusions of monoclonal antibodies directed at tumor necrosis factor (infliximab) and
against the B-lymphocyte antigen CD20 (rituximab) have been
reported to be of clinical benefit (improved aminotransferase levels,
immunoglobulin G levels, histologic inflammatory activity) as rescue therapy for refractory autoimmune hepatitis. To date, however,
only limited, often anecdotal, data in small numbers of patients
support these alternative approaches. If medical therapy fails, or
when chronic hepatitis progresses to cirrhosis and is associated
with life-threatening complications of liver decompensation, liver
transplantation is the only recourse (Chap. 345); in patients with
severe autoimmune hepatitis, failure of the bilirubin to improve
after 2 weeks of therapy should prompt early consideration of the
patient for liver transplantation. Recurrence of autoimmune hepatitis in the new liver occurs rarely in most experiences but in as many
as 35−40% of cases in others; nonetheless, 5-year patient and graft
survival exceed 80%.
Like all patients with chronic liver disease, patients with autoimmune hepatitis should be vaccinated against hepatitis A and
B, ideally before immunosuppressive therapy is begun, if practical. Patients with autoimmune hepatitis and cirrhosis should be
screened for HCC with ultrasound at 6-month intervals and for
gastroesophageal varices with upper gastrointestinal endoscopy at
intervals of 1–3 years, based on severity of liver disease.
■ FURTHER READING
AASLD/IDSA HCV Guidance Panel: Hepatitis C guidance 2019
update: American Association for the Study of Liver Diseases–
Infectious Diseases Society of America recommendations for testing,
managing, and treating hepatitis C virus infection. Hepatology
71:686, 2020. Updated regularly and available at http://www.hcvguidelines.org. Accessed April 20, 2020.
Bersoff-Matcha SJ et al: Hepatitis B virus reactivation associated
with direct-acting antiviral therapy for chronic hepatitis C virus: A
review of cases reported to the U.S. Food and Drug Administration
Adverse Event Reporting System. Ann Intern Med 166:792, 2017.
Bourliere M et al: Sofosbuvir, velpatasvir, and voxilaprevir for previously treated HCV infection. N Engl J Med 376:2134, 2017.
Buti M et al: Tenofovir alafenamide versus tenofovir disproxil fumarate for the treatment of HBeAg-negative chronic hepatitis B virus
infection: A randomized, double-blind, phase 3 non-inferiority trial.
Lance Gastroenterol Hepatol 1:196, 2017.
Butt AA et al: Direct-acting antiviral therapy for HCV infection is
associated with a reduced risk of cardiovascular disease events. Gastroenterology 156:987, 2019.
Carbone M, Neuberger JM: Autoimmune liver disease, autoimmunity and liver transplantation. J Hepatol 60:210, 2014.
Carrat F et al: Clinical outcomes in patients with chronic hepatitis C
after direct-acting antiviral treatments: A prospective cohort study.
Lancet 393:1453, 2019.
Chan HLY et al: Tenofovir alafenamide versus tenofovir disproxil
fumarate for the treatment of HBeAg-positive chronic hepatitis B
virus infection: A randomized, double-blind, phase 3 non-inferiority
trial. Lancet Gastroenterol Hepatol 1:185, 2017.
European Association for the Study of the Liver: EASL 2017
clinical practice guidelines on the management of hepatitis B virus
infection. J Hepatol 67:370, 2017.
European Association for the Study of the Liver: EASL recommendations on treatment of hepatitis C 2018. J Hepatol 69:461, 2018.
European Association for the Study of the Liver: EASL recommendations on treatment of hepatitis C: Final update of the series. J
Hepatol 73:1170, 2020.
Forns X et al: Glecaprevir plus pibrentasvir for chronic hepatitis C
virus genotype 1, 2, 4, 5, or 6 infection in adults with compensated
cirrhosis (EXPEDITION-1): A single-arm, open-label, multicentre
phase 3 trial. Lancet Infect Dis 17:1062, 2017.
Jacobson IM et al: American Gastroenterological Association Institute
clinical practice update-expert review: Care of patients who have
achieved a sustained virologic response after antiviral therapy for
chronic hepatitis C infection. Gastroenterology 152:1578, 2017.
Kwo PY et al: Glecaprevir and pibrentasvir yield high response rates in
patients with HCV genotype 1-6 without cirrhosis. J Hepatol 67:263,
2017.
Liem KS et al: Limited sustained response after stopping nucleos(t)ide
analogues in patients with chronic hepatitis B: Results from a randomized controlled trial (Toronto STOP study). Gut 68:2206, 2019.
Lok ASG et al: Antiviral therapy for chronic hepatitis B viral infection
in adults: A systematic review and meta-analysis. Hepatology 63:284,
2016.
Loomba R, Liang TJ: Hepatitis B reactivation associated with immune
suppressive and biological modifier therapies: Current concepts,
management strategies, and future directions. Gastroenterology
152:1297, 2017.
2617Alcohol-Associated Liver Disease CHAPTER 342
Mcglynn EA et al: Assessing the safety of direct-acting antiviral agents
for hepatitis C. JAMA Open Network 2(6):e194765, 2019.
Papatheodoridis GV et al: Eight-year survival in chronic hepatitis
B patients under long-term entecavir or tenofovir is similar to the
general population. J Hepatol 68:1129, 2018.
Papatheodoridis GV et al: DARING-B: Discontinuation of effective
entecavir or tenofovir disoporxil fumarate long-term therapy before
HBsAg loss in non-cirrhotic HBeAg-negative chronic hepatitis B.
Antivir Ther 23:677, 2018.
Pawlotsky J-M et al: From non-A, non-B hepatitis to hepatitis C virus
cure. J Hepatol 62:S87, 2015.
Perrillo RP et al: American Gastroenterological Association Institute
technical review on prevention and treatment of hepatitis B virus
reactivation during immunosuppressive drug therapy. Gastroenterology 148:221, 2015.
Reddy KJ et al: American Gastroenterological Association Institute
guideline on the prevention and treatment of hepatitis B virus reactivation during immunosuppressive drug therapy. Gastroenterology
148:215, 2015.
Rossi C et al: Sustained virologic response from interferon-based
hepatitis C regimens is associated with reduced risk of extrahepatic
manifestations. J Hepatol 71:1116, 2019.
Singal AG et al: AGA clinical practice update on interaction between
oral direct-acting antivirals for chronic hepatitis C infection and
hepatocellular carcinoma: Expert review. Gastroenterology 156:2149,
2019.
Singh S et al: Magnitude and kinetics of decrease in liver stiffness after
antiviral therapy in patients with chronic hepatitis C: A systematic
review and meta-analysis. Clin Gastroenterol Hepatol 16:27, 2018.
Spearman CW et al: Hepatitis C. Lancet 394:1451, 2019.
Tang LS et al: Chronic hepatitis B infection: A review. JAMA 319:1802,
2018.
Terrault N et al: Update on prevention, diagnosis, and treatment of
chronic hepatitis B: AASLD 2018 hepatitis B guidance. Hepatology
67:1560, 2018.
Van Den Brand FF et al: Increased mortality among patients with
vs without cirrhosis and autoimmune hepatitis. Clin Gastroenterol
Hepatol 17:940, 2019.
Yurdaydin C et al: Treating chronic hepatitis delta: The need for surrogate markers of treatment efficacy. J Hepatol 70:1008, 2019.
342 Alcohol-Associated
Liver Disease
Bernd Schnabl
Alcohol-associated liver diseases (ALD) comprise a spectrum of diseases associated with chronic alcohol consumption ranging from
alcohol-associated fatty liver disease and steatohepatitis to more
advanced liver disease including fibrosis and cirrhosis. Acute alcoholic
hepatitis is an acute-on-chronic form of ALD that is associated with
liver failure and high mortality.
■ EPIDEMIOLOGY
Approximately 5.8% of adults in the United States have an alcohol use
disorder, defined as >2 drinks per day in women and >3 drinks per day
in men, or partake in binge drinking, defined as 4 drinks for women
and 5 drinks for men in ~2 h (1 drink equals ~14 g of ethanol, which
is 1 beer, 4 oz of wine, or 1 oz of 80% spirits). Prevalence of ALD correlates with the amount of alcohol consumption in different regions.
Prevalence of alcohol-associated fatty liver disease is 4.7% of the general
population in the United States, and 1.5% has stage 2 or greater fibrosis.
Liver cirrhosis is the eleventh leading cause of mortality worldwide,
causing 1.16 million deaths annually; 48% of cases of cirrhosis can be
attributed to alcohol. Among patients with alcohol use disorder, 18%
had fibrosis, 26% had cirrhosis, and 7% had acute alcoholic hepatitis
without underlying cirrhosis. In the European population, the annual
incidence rate for acute alcoholic hepatitis is between 24 and 27 per
million persons in women and between 46 and 65 per million persons
in men.
■ PATHOGENESIS
Alcohol in the form of ethanol is rapidly absorbed in the upper gastrointestinal tract and predominantly metabolized in the liver. Ethanol
reaches the liver through the portal vein, and the majority of ethanol
is oxidized via alcohol dehydrogenase 1 (ADH1) into acetaldehyde
in hepatocytes. Chronic alcohol consumption induces the expression
of a second ethanol-metabolizing enzyme, cytochrome P450 family
2 subfamily E member 1 (CYP2E1), which also converts ethanol into
acetaldehyde. In addition to the direct cellular toxic effects of acetaldehyde, metabolism of ethanol into acetaldehyde causes the generation of reactive oxygen species (ROS), resulting in further injury of
hepatocytes via lipid peroxidation and DNA damage. Acetaldehyde is
then oxidized into acetate via acetaldehyde dehydrogenase (ALDH).
Inherited deficiency of ALDH2 is common in Asian countries and
leads to acetaldehyde accumulation after alcohol consumption. These
individuals develop nausea and cutaneous flushing. Several mechanisms contribute to the development of hepatic steatosis related to
alcohol consumption. Acetate is converted into acetyl-coenzyme A
(CoA), which contributes to fatty acid and triglyceride synthesis.
Alcohol, in part through epigenetic changes, increases the expression
of genes involved in lipogenesis, while genes involved in fatty acid
transport and oxidation are suppressed. Alcohol also increases the ratio
of reduced nicotinamide adenine dinucleotide (NAD)/oxidized NAD
(NADH/NAD+) in hepatocytes, which further reduces mitochondrial
β-oxidation. Alcohol can increase fatty acid mobilization in adipose
tissue and the intestine, which will lead to hepatic accumulation of fatty
acids and increased hepatic steatosis. Overall, the net effect of these
processes contributes to fat accumulation in the liver.
■ RISK FACTORS FOR PROGRESSION OF ALD
Daily alcohol consumption or heavy drinking results in hepatic steatosis, but only 10–20% of such individuals will develop progressive liver
disease and cirrhosis. Therefore, other cofactors such as behavioral,
environmental, and genetic factors play important roles in progression
of ALD (Table 342-1). There is a dose-dependent increase, with regard
to the amount of alcohol consumed, in the likelihood of developing
liver cirrhosis. Women develop ALD at a lower daily alcohol intake.
Cigarette smoking is an independent risk factor for alcohol-associated
cirrhosis. The drinking pattern, in particular binge drinking and
excessive alcohol drinking outside meals, increases the risk of developing progressive ALD. Obesity and other chronic liver diseases such
as viral hepatitis, hemochromatosis, and nonalcoholic steatohepatitis
(NASH), are frequent cofactors contributing to progression of ALD.
Twin studies demonstrated a genetic predisposition to alcohol-associated liver cirrhosis that is independent from the genetic predisposition
to alcohol use disorder. Gene polymorphisms conferring increased
risk of alcohol-associated liver cirrhosis have been found in three
genes, patatin-like phospholipase domain-containing 3 (PNPLA3),
TABLE 342-1 Factors for Progression of Alcohol-Associated Liver
Disease
• Alcohol dose (>1 drink per day for women, >2 drinks per day for men)
• Drinking pattern (drinking without meal, binge drinking)
• Genetic factors, especially PNPLA3 polymorphism
• Female gender
• Smoking
• Increased body mass index and chronic liver diseases
• Intestinal microbiota
2618 PART 10 Disorders of the Gastrointestinal System
membrane bound O-acyltransferase domain-containing 7 (MBOAT7),
and transmembrane 6 superfamily member 2 (TM6SF2), although
the molecular mechanism is not well understood. A subset of patients
with alcohol use disorder develop changes in the gut microbiome and
increased intestinal permeability resulting in activation of hepatic
inflammation, hepatocyte death, and activation of fibrotic pathways.
Ongoing fibrosis due to continued alcohol consumption can result in
the development of cirrhosis with portal hypertension (Chap. 344).
■ CLINICAL FEATURES
The development of alcohol-associated steatosis, steatohepatitis, and
cirrhosis is most often clinically silent. Symptoms arise once the patient
with alcohol-associated liver cirrhosis decompensates or develops
alcoholic hepatitis (Table 342-2). Patients with alcoholic hepatitis have
been drinking heavily for typically >5 years and until at least 8 weeks
before onset of symptoms. They present with rapid onset of jaundice
(serum bilirubin >3 mg/dL), often accompanied by fever, malaise,
tender hepatomegaly, and clinical signs of hepatic decompensation,
such as ascites, bacterial infection, variceal bleeding, and hepatic
encephalopathy. Infections occur in 12–26% of patients with severe
alcoholic hepatitis at the time of admission. Alcoholic hepatitis is often
accompanied by systemic inflammatory response syndrome (SIRS) and
acute kidney injury (AKI) secondary to hepatorenal syndrome.
■ LABORATORY FINDINGS
Patients with simple hepatic steatosis can present with normal liver
function tests. Steatohepatitis is characterized by elevated levels of
aspartate aminotransferase (AST) and γ-glutamyl transferase (GGT).
Characteristic laboratory parameters for ALD include a ratio of AST
to alanine aminotransferase (ALT) of >1, and serum AST is rarely
>300 IU/L. Serum bilirubin and international normalized ratio (INR)
are typically normal. Elevated bilirubin and INR and low serum albumin and platelet count are common laboratory finings in patients with
cirrhosis. Patients with alcoholic hepatitis have AST and ALT elevations that do not exceed 400 IU/L, with AST/ALT ratio of >1.5 and
serum bilirubin >3 mg/dL.
■ DIAGNOSIS
The Alcohol Use Disorders Inventory Test (AUDIT) is a validated tool
for identifying patients with alcohol use disorder (Chap. 453). Diagnosis of ALD requires exclusion of other liver diseases in heavy drinkers.
Alcohol-associated steatosis can be diagnosed by simple ultrasound,
magnetic resonance imaging (MRI), or computed tomography (CT).
Noninvasive quantification of hepatic fat can be achieved with the
ultrasound technique of controlled attenuation parameter (CAP) or
with magnetic resonance proton density fat fraction (MR-PDFF). Liver
biopsy is rarely indicated for diagnosing alcohol-associated hepatic
steatosis or steatohepatitis. Liver biopsy typically shows hepatocytes
TABLE 342-2 Symptoms and Signs Associated with Alcohol-Associated
Cirrhosis and Alcoholic Hepatitis
• Tiredness
• Malnutrition and sarcopenia
• Abdomen: abdominal discomfort, hepatomegaly, splenomegaly, caput
medusae, ascites with weight gain, abdominal pain, and shortness of breath
• Skin: spider angioma, palmar erythema, jaundice, ecchymoses
• Eyes: icteric sclerae
• Hands: Dupuytren contracture
• Face: rhinophyma
• Reproductive system: gynecomastia, gonadal atrophy, loss of libido,
amenorrhea
• Neurologic:
• Peripheral neuropathy
• Alcohol withdrawal: tachycardia, agitation, tremor, seizures, delirium
• Hepatic encephalopathy: asterixis (flapping tremor), forgetfulness, inversion
of sleep/wake pattern, altered consciousness, confusion, lethargy, coma
• Wernicke-Korsakoff syndrome
with large lipid droplets (macrovesicular steatosis) around pericentral
veins (zone 3). Morphologic features of alcohol-associated steatohepatitis include hepatocyte injury and ballooning with Mallory-Denk
bodies, necrosis, and lobular inflammation with mononuclear and
neutrophilic granulocytes.
Progression of alcohol-associated steatohepatitis to fibrosis can be
diagnosed using liver stiffness measurement by techniques such as
transient elastography (e.g., FibroScan). Liver stiffness <6 kPa indicates
normal liver, whereas cutoffs for each stage of alcohol-associated liver
fibrosis have been validated (>8 kPa indicates ≥F3 advanced fibrosis;
>12.5 kPa indicates F4 cirrhosis). Histology shows initially perivenular fibrosis with subsequent extension of collagen fibers into hepatic
lobules, described as septal fibrosis. Patients with cirrhosis show liver
nodularity on imaging with ultrasound, MRI, or CT scan. Radiologic
signs of portal hypertension include ascites, splenomegaly, and portalsystemic collateral vessels. Prognosis and risk of mortality are assessed
using Child-Pugh-Turcotte (CPT) or Model for End-Stage Liver Disease (MELD; or sodium-MELD) scores (Chap. 344).
In patients presenting with features suggestive of alcoholic hepatitis,
imaging is obtained to exclude biliary obstruction and hepatocellular
carcinoma (HCC). In addition, other causes of liver disease such as
viral hepatitis, Wilson’s disease, and severe autoimmune liver disease
should be ruled out. Histology shows macrovesicular steatosis, hepatocyte ballooning with Mallory-Denk bodies, megamitochondria,
neutrophil infiltration, bilirubinostasis, and chicken wire fibrosis. The
majority of patients with alcoholic hepatitis have underlying cirrhosis
(80%) (Chap. 344), and 10–20% of patients with a clinical diagnosis of
alcoholic hepatitis will have other liver diseases on biopsy. Therefore, in
the presence of potential confounding factors, including possible ischemic hepatitis (in the setting of, e.g., hypotension, massive gastrointestinal bleeding, recent cocaine use, septic shock), drug-induced liver
injury (DILI), autoimmune liver disease, uncertain alcohol use assessment, or atypical laboratory tests (AST <50 IU/L or >400 IU/L, AST/ALT
ratio <1.5), a transjugular liver biopsy is recommended to confirm the
diagnosis of alcoholic hepatitis. Infections need to be assessed routinely
with chest x-ray and blood, urine, and ascites cultures in patients presenting with alcoholic hepatitis.
TREATMENT
Alcohol-Associated Liver Disease (Fig. 342-1)
To date, the most effective therapy to reduce the progression of and
reverse ALD is prolonged alcohol abstinence. In particular, alcoholassociated hepatic steatosis and steatohepatitis are reversible with
cessation of alcohol consumption. Thus, treatment of the underlying alcohol use disorder is an integral part for therapy of ALD. There
are currently no approved drugs for treatment of alcohol-associated
steatosis and steatohepatitis with or without fibrosis.
Patients with alcohol-associated cirrhosis and ongoing alcohol
consumption are at risk for decompensation and development
of hepatic encephalopathy, ascites, variceal bleeding, hepatorenal
syndrome, and HCC (Chap. 344). Patients with cirrhosis should
undergo an upper gastrointestinal endoscopy to screen for varices.
HCC screening is recommended using ultrasonography every 6
months in patients with cirrhosis. Management of complications of
cirrhosis such as variceal bleeding, ascites, hepatic encephalopathy,
and HCC does not differ from patients with cirrhosis due to a different etiology (Chap. 344). Liver transplantation for patients with
alcohol-associated decompensated cirrhosis or HCC is a definitive
therapy and is currently the leading indication for liver transplantation in the United States. Liver transplantation evaluation should
be taken into consideration for patients with end-stage liver disease
(Chap. 345).
In patients diagnosed with alcoholic hepatitis, short-term mortality can be predicted using the Maddrey discriminant function
(MDF; calculated as 4.6 × [the prolongation of the prothrombin
time above control {seconds}] + serum bilirubin [mg/dL]), MELD
score (Chap. 344), or age-bilirubin-INR-creatinine (ABIC) score.
2619 Nonalcoholic Fatty Liver Diseases and Nonalcoholic Steatohepatitis CHAPTER 343
Patients with MDF <32 or MELD ≤20 are defined as having moderate alcoholic hepatitis. Currently, patients with moderate alcoholic
hepatitis are treated under a multidisciplinary team including an
alcohol use disorder specialist, dietitian for nutritional supplementation for patients with markedly reduced intake, and hepatologist
for managing liver disease complications. Enteral nutrition with a
goal of >21 kcal/kg and supplementation of micronutrients (in particular zinc) and vitamin supplementation (in particular vitamin B1
)
are recommended for patients with alcoholic hepatitis. Intravenous
albumin is preferred for volume expansion. MDF ≥32 or MELD
>20 identifies patients with severe alcoholic hepatitis and high
short-term mortality who will have a survival benefit with glucocorticoid treatment. Contraindications for glucocorticoid treatment
include uncontrolled infections or sepsis, AKI and hepatorenal syndrome, uncontrolled upper gastrointestinal bleeding, concomitant
diseases (including viral hepatitis, HCC, pancreatitis, DILI, active
tuberculosis, and HIV), multiorgan failure, and shock. Glucocorticoids can be used once infection, sepsis, and gastrointestinal bleeding are adequately controlled. Glucocorticoid use reduces the risk
of death in patients with severe alcoholic hepatitis within 28 days
of treatment but not in the following 6 months. Oral prednisolone,
40 mg/d for a total duration of 4 weeks, is preferred. For patients
unable to take oral medications, methylprednisolone, 32 mg/d IV,
is used. The combination of glucocorticoids with N-acetylcysteine
infusion might add short-term survival benefit at 1 month. Failure
of improvement of Lille score (≥0.45) after 7 days of glucocorticoid
treatment will determine patients with severe alcoholic hepatitis
who will unlikely benefit from continued treatment with glucocorticoids. Glucocorticoids should be stopped in nonresponders, and
early liver transplantation should be considered. Although shortterm prognosis is dependent on liver disease severity at the time
of presentation, long-term prognosis (>1 year) largely depends on
Alcoholic Hepatitis (AH)
Clinical diagnosis with
laboratory findings
Confounding
diagnostic
factors
TJ liver biopsy
7 days
Lille score <0.45
Continue prednisolone
for 28 days total
Stop prednisolone
Lille score ≥0.45
Severe AH
MDF ≥32
or MELD >20
Moderate AH
MDF <32
or MELD ≤20
- Alcohol
abstinence
- Nutritional
support
- If eligible, early liver transplantation (LT)
- If not eligible for LT: Supportive/palliative care
Contraindications
for corticosteroids
Oral prednisolone 40 mg/d
(unable to take oral medications:
methylprednisolone 32 mg/d IV)
FIGURE 342-1 Treatment algorithm for alcoholic hepatitis. In patients with a
clinical diagnosis of alcoholic hepatitis, confounding factors (see text) need to
be ruled out, if necessary, by transjugular (TJ) liver biopsy. Patients with severe
alcoholic hepatitis (AH), defined as Maddrey discriminant function (MDF) ≥32 or
Model for End-Stage Liver Disease (MELD) score >20, without contraindications
for glucocorticoids (see text) are candidates for such treatment. Nonresponders or
patients with contraindications for treatment should be considered for early liver
transplantation (LT) or supportive or palliative care, as clinically appropriate.
alcohol abstinence and underlying cirrhosis. Patients with severe
alcoholic hepatitis that is nonresponsive to medical therapy have
high 30-day mortality and are therefore unable to fulfill a minimum
of 6 months of alcohol abstinence, which is required in many centers for liver transplantation evaluation. Early liver transplantation
can be successfully performed in highly selected patients with an
excellent psychosocial profile (Chap. 345). If a nonresponder is
ineligible for early liver transplantation, supportive or palliative
care should be considered for patients with multiple-organ failure.
■ FURTHER READING
Crabb DW et al: Standard definitions and common data elements for
clinical trials in patients with alcoholic hepatitis: Recommendation
from the NIAAA Alcoholic Hepatitis Consortia. Gastroenterology
150:785, 2016.
Crabb DW et al: Diagnosis and treatment of alcohol-associated liver
diseases: 2019 practice guidance from the American Association for
the Study of Liver Diseases. Hepatology 71:306, 2020.
Louvet A et al: Corticosteroids reduce risk of death within 28 days for
patients with severe alcoholic hepatitis, compared with pentoxifylline
or placebo-a meta-analysis of individual data from controlled trials.
Gastroenterology 155:458, 2018.
Seitz HK et al: Alcoholic liver disease. Nat Rev Dis Primers 4:16, 2018.
Singal AK et al: ACG clinical guideline: Alcoholic liver disease. Am J
Gastroenterol 113:175, 2018.
343 Nonalcoholic Fatty Liver
Diseases and Nonalcoholic
Steatohepatitis
Manal F. Abdelmalek, Anna Mae Diehl
■ INCIDENCE, PREVALENCE,
AND NATURAL HISTORY
Nonalcoholic fatty liver disease (NAFLD) is the most common cause
of chronic liver disease in the United States, as well as worldwide. The
global prevalence of NAFLD is estimated to be as high as one billion.
In the United States, NAFLD is estimated to effect between 80 and
100 million individuals. NAFLD is strongly associated with insulin
resistance, overweight/obesity, and metabolic syndrome. However, it
can also occur in lean individuals and is particularly common in those
with a paucity of adipose depots (i.e., lipodystrophy). Ethnic/racial
factors also appear to influence liver fat accumulation; the documented
prevalence of NAFLD is lowest in African Americans (~25%), highest in Americans of Hispanic ancestry (~50%), and intermediate in
American whites (~33%).
NAFLD encompasses a spectrum of liver pathology with different
clinical prognoses (Fig. 343-1). The simple accumulation of triglyceride within hepatocytes (hepatic steatosis) is on the most clinically
benign extreme of the spectrum. On the opposite, most clinically
ominous extreme, are cirrhosis (Chap. 344) and primary liver cancer (Chap. 82). The risk of developing cirrhosis is extremely low in
individuals with isolated steatosis (nonalcoholic fatty liver [NAFL])
but increases as steatosis becomes complicated by liver-cell injury and
death and the accumulation of inflammatory cells (i.e., nonalcoholic
steatohepatitis [NASH]). At least a quarter of adults with NAFLD are
presumed to have NASH. NASH itself is also a heterogeneous condition; it can improve to steatosis or normal histology, remain relatively
stable for years, or cause progressive accumulation of fibrous scar that
eventuates in cirrhosis (stage 4 fibrosis). Advanced hepatic fibrosis is
the primary predictor of eventual liver-related morbidity and mortality
2620 PART 10 Disorders of the Gastrointestinal System
in NAFLD. Once NAFLD-related cirrhosis develops, the annual incidence of primary liver cancer can be as high as 1–2% per year.
Abdominal imaging is not able to determine which individuals
with NAFLD have associated liver-cell death and inflammation (i.e.,
NASH), and specific blood tests to diagnose NASH are not yet available. However, population-based studies that have used elevated serum
alanine aminotransferase (ALT) as a marker of liver injury indicate
that ~6–8% of American adults have serum ALT elevations that cannot
be explained by excessive alcohol consumption, other known causes
of fatty liver disease (Table 343-1), viral hepatitis, or drug-induced or
congenital liver diseases. Because the prevalence of such “cryptogenic”
Dynamic process
Healthy liver
A B
Steatosis (NAFL) Steatohepatitis (NASH) Cirrhosis
C D
FIGURE 343-1 Histopathologic spectrum of nonalcoholic fatty liver disease (NAFLD). NAFLD encompasses a dynamic spectrum of liver pathology. A. Healthy liver.
B. Simple steatosis (nonalcoholic fatty liver [NAFL]); arrow shows fatty hepatocyte. C. Nonalcoholic steatohepatitis (NASH); ballooned hepatocyte (arrow) near central vein
with adjacent blue-stained pericellular fibrosis (arrowheads). D. Cirrhosis with blue-stained bridging fibrosis surrounding micronodules of liver parenchyma.
ALT elevations increases with body mass index, it is presumed that
they are due to NASH. Hence, at any given point in time, NASH is
present in ~25% of individuals who have NAFLD (i.e., ~6–8% of the
general U.S. adult population has NASH). Smaller cross-sectional
studies in which liver biopsies have been performed on NASH patients
at tertiary referral centers consistently demonstrate advanced fibrosis
or cirrhosis in ~25% of those cohorts. By extrapolation, therefore,
TABLE 343-1 Alternative Causes of Hepatic Steatosis
• Alcoholic liver disease
• Hepatitis C (particularly genotype 3)
• Inborn errors of metabolism
• Abetalipoproteinemia
• Cholesterol ester storage disease
• Galactosemia
• Glycogen storage disease
• Hereditary fructose intolerance
• Homocystinuria
• Systemic carnitine deficiency
• Tyrosinemia
• Weber-Christian syndrome
• Wilson’s disease
• Wolman’s disease
• Medications (see Table 343-2)
• Miscellaneous
• Industrial exposure to petrochemical
• Inflammatory bowel disease
• Lipodystrophy
• Bacterial overgrowth
• Starvation
• Parenteral nutrition
• Surgical procedures
• Bilopancreatic diversion
• Extensive small-bowel resection
• Gastric bypass
• Jejunoileal bypass
• Reye’s syndrome
• Acute fatty liver of pregnancy
• HELLP syndrome (hemolytic anemia, elevated liver enzymes, low platelet
count)
TABLE 343-2 Medications Associated with Hepatic Steatosis
• Cytotoxic and cytostatic drugs
• 5-Fluorouraci
• l-Asparaginase
• Azacitidine
• Azaserine
• Bleomycin
• Methotrexate
• Puromycin
• Tetracycline
• Doxycycline
• Metals
• Antimony
• Barium salts
• Chromates
• Phosphorus
• Rare earths of low atomic number
• Thallium compounds
• Uranium compounds
• Other drugs and toxins
• Amiodarone
• 4,4’-Diethylaminoethoxyhexesterol
• Ethionine
• Ethyl bromide
• Estrogens
• Glucocorticoids
• Highly active antiretroviral therapy
• Hydralazine
• Hypoglycin
• Orotate
• Perhexiline maleate
• Safrole
• Tamoxifen
• Valproic acid
• Acetylsalicylic acid intoxication
• Apo-B inhibitors: Mipomersen and lomitapide
2621 Nonalcoholic Fatty Liver Diseases and Nonalcoholic Steatohepatitis CHAPTER 343
cirrhosis develops in ~6% of individuals with NAFLD (i.e., in ~1.5–2%
of the general U.S. population). The risk for advanced liver fibrosis is
highest in individuals with NASH who are aged >45–50 years and overweight/obese or afflicted with type 2 diabetes. Having a first-degree
relative with cryptogenic hepatitis or cirrhosis also increases the risk
for developing cirrhosis.
Heritable factors clearly impact susceptibility to hepatic steatosis,
NASH, liver fibrosis, and liver cancer. Genetic variants on or near
TM6SF2 or MBOAT7 (genes involved in lipid homeostasis) and
palatin-like phospholipase domain-containing 3 gene (PNPLA3, a gene
that encodes an enzyme involved in intracellular trafficking of lipids)
may increase the heritability of NAFLD. A recent meta-analysis showed
that PNPLA3 exerts a strong influence not only on hepatic fat accumulation but also on the severity of NASH and liver fibrosis. Indeed,
recent twin studies suggest that inheritance accounts for about half the
risk for developing cirrhosis. Epigenetic factors (i.e., heritable traits
that do not result from direct changes in DNA) may also influence
NAFLD pathogenesis and/or progression based on evidence that intrauterine exposures influence susceptibility to obesity and the metabolic
syndrome in adolescence. Studies of families with adult-onset obesity
have identified genome-wide epigenetic alterations that dysregulate
metabolic pathways controlling adiposity, insulin sensitivity, and tissue
generation or regeneration. Whether such epigenetic mechanisms
influence susceptibility to NASH and cirrhosis is being investigated.
NAFLD is currently the leading indication for liver transplantation
in the United States. Similar to cirrhosis caused by other liver diseases,
cirrhosis caused by NAFLD increases the risk for primary liver cancer.
Both hepatocellular carcinoma and intrahepatic cholangiocarcinoma
(ICC) have also been reported to occur in NAFLD patients without
cirrhosis, suggesting that NAFLD per se may be a premalignant condition. NAFLD, NASH, and NAFLD-related cirrhosis are not limited to
adults. All have been well documented in children. As in adults, obesity
and insulin resistance are the main risk factors for pediatric NAFLD.
Thus, the rising incidence and prevalence of childhood obesity suggests that NAFLD will be a major contributor to society’s burden of
liver disease in the future.
■ PATHOGENESIS
The mechanisms underlying the pathogenesis and progression of
NAFLD are not entirely clear. The best-understood mechanisms
pertain to hepatic steatosis. This is proven to result when hepatocyte
mechanisms for triglyceride synthesis (e.g., lipid uptake and de novo
lipogenesis) overwhelm mechanisms for triglyceride disposal (e.g.,
degradative metabolism and lipoprotein export), leading to accumulation of fat (i.e., triglyceride) within hepatocytes. Obesity stimulates hepatocyte triglyceride accumulation by altering the intestinal
microbiota to enhance both energy harvest from dietary sources and
intestinal permeability. Reduced intestinal barrier function increases
hepatic exposure to gut-derived products, which stimulate liver cells
to generate inflammatory mediators that inhibit insulin actions. Obese
adipose depots also produce excessive soluble factors (adipokines) that
inhibit tissue insulin sensitivity. Insulin resistance promotes hyperglycemia, which drives the pancreas to produce more insulin to maintain
glucose homeostasis. However, hyperinsulinemia also promotes lipid
uptake, fat synthesis, and fat storage. The net result is hepatic triglyceride accumulation (i.e., steatosis).
Triglyceride per se is not hepatotoxic. However, its precursors (e.g.,
fatty acids and diacylglycerols) and metabolic by-products (e.g., reactive oxygen species) may damage hepatocytes, leading to hepatocyte
lipotoxicity. Lipotoxicity also triggers the generation of other factors
(e.g., inflammatory cytokines, hormonal mediators) that deregulate
systems that normally maintain hepatocyte viability. The net result is
increased hepatocyte death. Dying hepatocytes, in turn, release various factors that trigger wound healing responses that aim to replace
(regenerate) lost hepatocytes. Such repair involves transient expansion
of other cell types, such as myofibroblasts and progenitor cells, that
make and degrade matrix, remodel the vasculature, and generate
replacement hepatocytes, as well as the recruitment of immune cells
that release factors that modulate liver injury and repair. NASH is
the morphologic manifestation of lipotoxicity and resultant wound
healing responses. Because the severity and duration of lipotoxic liver
injury dictate the intensity and duration of repair, the histologic features and outcomes of NASH are variable. Cirrhosis and liver cancer
are potential outcomes of chronic NASH. Cirrhosis results from futile
repair, i.e., progressive accumulation of wound healing cells, fibrous
matrix, and abnormal vasculature (scarring), rather than efficient
reconstruction/regeneration of healthy hepatic parenchyma. Primary
liver cancers develop when malignantly transformed liver cells escape
mechanisms that normally control regenerative growth. The mechanisms responsible for futile repair (cirrhosis) and liver carcinogenesis
are not well understood. Because normal liver regeneration is a very
complex process, there are multiple opportunities for deregulation
and, thus, pathogenic heterogeneity. To date, this heterogeneity has
confounded development of both diagnostic tests and treatments for
defective/deregulated liver repair (i.e., cirrhosis and cancer). Hence,
current strategies focus on circumventing misrepair by preventing and/
or reducing lipotoxic liver injury.
■ DIAGNOSIS
Diagnosing NAFLD requires demonstration of increased liver fat in
the absence of hazardous levels of alcohol consumption. Thresholds
for potentially dangerous alcohol ingestion have been set at more than
one drink per day in women and two drinks per day in men based on
epidemiologic evidence that the prevalence of serum aminotransferase
elevations increases when alcohol consumption habitually exceeds
these levels. In those studies, one drink was defined as having 10 g
of ethanol and, thus, is equivalent to one can of beer, 4 oz of wine,
or 1.5 oz (one shot) of distilled spirits. Other causes of liver fat accumulation (particularly exposure to certain drugs; Table 343-2) and
liver injury (e.g., viral hepatitis, autoimmune liver disease, iron or copper
overload, α1
antitrypsin deficiency) must also be excluded. Thus, establishing the diagnosis of NAFLD does not require invasive testing: it can be
accomplished by history and physical examination, liver imaging (ultrasound is an acceptable first-line test; computed tomography [CT] or magnetic resonance imaging [MRI] enhances sensitivity for liver fat detection
but adds expense), and blood tests to exclude other liver diseases.
It is important to emphasize that, in individuals with NAFLD, the
liver may not be enlarged and serum aminotransferases and liver
function tests (e.g., bilirubin, albumin, prothrombin time) may be
completely normal. Because there is yet no one specific blood test for
NAFLD, confidence in the diagnosis of NAFLD is increased by identification of NAFLD risk factors. The latter include increased body mass
index, insulin resistance/type 2 diabetes mellitus, and other parameters
indicative of the metabolic syndrome (e.g., systemic hypertension,
dyslipidemia, hyperuricemia/gout, cardiovascular disease; Chap. 408)
in the patient or family members. Individuals who have, or have had,
pituitary or hypothalamic neoplasms and women with polycystic ovary
syndrome are also at increased risk for NAFLD. Hypothyroidism and
obstructive sleep apnea may also increase NAFLD, presumably by promoting obesity and/or exacerbating the metabolic syndrome.
Establishing the severity of NAFLD-related liver injury and related
scarring (i.e., staging NAFLD) is more difficult than simply diagnosing
NAFLD. Staging is critically important, however, because it is necessary
to define prognosis and thereby determine treatment recommendations. The goal of staging is to distinguish patients with NASH from
those with simple steatosis and to identify which of the NASH patients
have advanced fibrosis. The 10-year probability of developing liverrelated morbidity or mortality in steatosis is negligible, and hence, this
subgroup of NAFLD patients tends to be managed conservatively (see
below). In contrast, more intensive follow-up and therapy are justified
in NASH patients, and the subgroup with advanced fibrosis merits the
most intensive scrutiny and intervention because their 10-year risk of
liver-related morbidity and mortality is clearly increased.
Staging approaches can be separated into noninvasive testing
(i.e., blood testing, physical examination, and imaging) and invasive
approaches (i.e., liver biopsy). Blood test evidence of hepatic dysfunction (e.g., hyperbilirubinemia, hypoalbuminemia, prothrombin time
prolongation) or portal hypertension (e.g., thrombocytopenia) and
2622 PART 10 Disorders of the Gastrointestinal System
stigmata of portal hypertension on physical examination (e.g., spider
angiomata, palmar erythema, splenomegaly, ascites, clubbing, encephalopathy) suggest a diagnosis of advanced NAFLD. Liver biopsy has
been the gold standard for establishing the severity of liver injury and
fibrosis because it is both more sensitive and more specific than these
other tests for establishing NAFLD severity. Further, although invasive,
liver biopsy is seldom complicated by serious adverse sequelae such as
significant bleeding, pain, or inadvertent puncture of other organs and
thus is relatively safe. However, biopsy suffers from potential sampling
error unless tissue cores of 2 cm or longer are acquired. Also, examination of tissue at a single point in time is not reliable for determining
whether the pathologic processes are progressing or regressing. The
risk of serial liver biopsies within short time intervals is generally
deemed as unacceptable outside of research studies. These limitations of liver biopsy have stimulated efforts to develop noninvasive
approaches to stage NAFLD.
As is true for many other types of chronic liver disease, in NAFLD,
the levels of serum aminotransferases (aspartate aminotransferase
[AST] and ALT) do not reliably reflect the severity of liver cell injury,
extent of liver-cell death, or related liver inflammation and fibrosis.
Thus, they are imperfect for determining which individuals with
NAFLD have NASH. This has prompted efforts to identify superior
markers of NASH and, particularly, liver fibrosis, because fibrosis stage
predicts eventual liver outcomes and mortality in NASH. Algorithms
that combine various laboratory tests (e.g., Enhanced Liver Fibrosis
[ELF] score, BARD score, AST to Platelet Ratio Index [APRI] score,
NAFLD fibrosis score, and Fibrosis-4 [FIB-4] score) are somewhat
helpful in separating NASH patients with advanced versus mild liver
fibrosis. The NAFLD fibrosis score (NFS) and FIB-4 score, two of
the most commonly employed noninvasive tests to assess severity of
hepatic fibrosis, can be calculated from a few readily available clinical variables (age, body mass index, glucose, platelet count, albumin,
AST, ALT) using published formulas that are readily accessed via an
online calculator. Both scores are helpful for gauging the severity of
NASH and liver fibrosis. Combining these tests with new imaging
approaches that permit noninvasive quantification of liver fat (e.g.,
MRI using proton density fat fraction [MRI-PDFF]) and liver stiffness,
a surrogate marker of liver fibrosis (e.g., magnetic resonance elastography [MRE], and transient elastography [FibroScan]), improves their
predictive power (Chap. 337). Transient elastography in particular
has become widely available and is relatively inexpensive. It is most
useful for excluding advanced liver fibrosis as cirrhosis is extremely
unlikely when the liver stiffness score is low. However, higher stiffness
scores must be interpreted with caution since several factors (obesity,
nonfasting state, hepatic inflammation, iron overload, and/or hepatic
congestion) decrease the specificity of the test. Increasingly, these new
serologic and imaging tools are being used serially or in combination
to monitor fibrosis progression and regression in NAFLD patients. As
a result, liver biopsy staging is becoming restricted to patients who
cannot be stratified reliably using these noninvasive assessments. Indeterminant or discordant results of noninvasive testing should prompt
referral to a liver specialist and consideration of liver biopsy.
■ CLINICAL FEATURES OF NAFLD
Most subjects with NAFLD are asymptomatic. The diagnosis is often
made when abnormal liver aminotransferases or features of fatty liver
are noted during an evaluation performed for other reasons. NAFLD
may also be diagnosed during the workup of vague right upper
quadrant abdominal pain, hepatomegaly, or an abnormal-appearing
liver at time of abdominal surgery. Obesity is present in 50–90% of subjects. Most patients with NAFLD also have other features of the metabolic syndrome (Chap. 408). Some have subtle stigmata of chronic liver
disease, such as spider angiomata, palmer erythema, or splenomegaly.
In a small minority of patients with advanced NAFLD, complications
of end-stage liver disease (e.g., jaundice, features of portal hypertension
such as ascites or variceal hemorrhage) may be the initial findings.
The association of NAFLD with obesity, diabetes, hypertriglyceridemia, hypertension, and cardiovascular disease is well known. Other
associations include chronic fatigue, mood alterations, obstructive
sleep apnea, thyroid dysfunction, polycystic ovary syndrome, and
chronic pain syndrome. NAFLD is an independent risk factor for metabolic syndrome (Chap. 408). Longitudinal studies suggest that patients
with NASH are at two- to threefold increased risk for the development
of metabolic syndrome. Similarly, studies have shown that patients
with NASH have a higher risk for the development of hypertension and
diabetes mellitus. The presence of NAFLD is also independently associated with endothelial dysfunction, increased carotid intimal thickness,
and the number of plaques in carotid and coronary arteries. Such data
indicate that NAFLD has many deleterious effects on health in general.
■ TREATMENT OF NAFLD
Treatment of NAFLD can be divided into three components: (1) specific
therapy of NAFLD-related liver disease; (2) treatment of NAFLDassociated comorbidities; and (3) treatment of the complications of
advanced NAFLD. The subsequent discussion focuses on specific
therapies for NAFLD, with some mention of their impact on major
NAFLD comorbidities (insulin resistance/diabetes, obesity, and dyslipidemia). Treatment of the complications of advanced NAFLD involves
management of the complications of cirrhosis and portal hypertension, including primary liver cancers. Approaches to accomplish these
objectives are similar to those used in other chronic liver diseases and
are covered elsewhere in the textbook (Chaps. 344 and 82).
At present, there are no U.S. Food and Drug Administration (FDA)–
approved therapies for the treatment of NAFLD. Thus, the current
approach to NAFLD management focuses on treatment to improve
the risk factors for NASH (i.e., obesity, insulin resistance, metabolic
syndrome, dyslipidemia). Based on our understanding of the natural history of NAFLD, only patients with NASH or hepatic fibrosis
are considered currently for targeted pharmacologic therapies. This
approach may change as our understanding of disease pathophysiology
improves and potential targets of therapy evolve.
Diet and Exercise Lifestyle changes and dietary modifications
that result in weight loss and/or improve insulin sensitivity are the primary treatments for NAFLD. Many studies indicate that loss of 3–5%
of body weight improves steatosis and that greater weight loss (i.e.,
≥7–10%) improves steatohepatitis and hepatic fibrosis. The benefits
of modifying dietary macronutrient contents (e.g., low-carbohydrate
vs low-fat diets, saturated vs unsaturated fat diets) generally parallel
changes in calorie consumption, suggesting that diet modifications
are mainly beneficial because they reduce energy intake and improve
obesity. However, a Mediterranean-type diet has been reported to
improve NASH and liver fibrosis independently of weight loss. Excluding foods and beverages high in added fructose and increasing coffee
consumption are also recommended because high-fructose diets have
been shown to exacerbate hepatic steatosis, steatohepatitis, and fibrosis, and consuming two or more cups of coffee per day is associated
with reduced risk of liver fibrosis. Changes in diet composition particularly merit consideration in lean individuals with NAFLD, although
available data are insufficient to determine if this improves their liver
histology. Modifying lifestyle to increase physical activity (i.e., energy
expenditure) complements dietary caloric restriction and, thus, expedites weight loss. Exercise also improves muscle insulin sensitivity,
which improves the metabolic syndrome independent of weight loss.
Both aerobic exercise and resistance training effectively reduce liver
fat. At least 30 min of moderate-intensity aerobic exercise or resistance
training five times per week is recommended. The choice of training
should be tailored to patients’ preferences and functional capacity to
enable long-term maintenance. Any activity is better than remaining
sedentary. Unfortunately, most NAFLD patients cannot sustain longterm compliance with diet and lifestyle modifications and, thus, fail
to maintain a healthier weight. Although pharmacologic therapies to
facilitate weight loss, such as orlistat, topiramate, phentermine, and
GLP-1 receptor agonists, are available, their role in the treatment of
NAFLD remains experimental.
Pharmacologic Therapies Several drug therapies have been
tried in both research and clinical settings. There are currently no
FDA-approved drugs for the treatment of NAFLD. Hence, at present,
2623 Nonalcoholic Fatty Liver Diseases and Nonalcoholic Steatohepatitis CHAPTER 343
NAFLD patients without NASH or fibrosis should receive only counseling for healthy diet and physical activity. Consideration of additional
specific pharmacotherapy for liver disease is restricted to NAFLD
patients with more serious liver damage (i.e., NASH or liver fibrosis).
A number of large clinical trials designed to identify effective and safe
treatments for these conditions are in progress. Because NAFLD is
strongly associated with the metabolic syndrome and type 2 diabetes
(Chaps. 403 and 404), the efficacy of various insulin-sensitizing agents
has been examined. Metformin, an agent that mainly improves hepatic
insulin sensitivity, has been evaluated in several small, open-label
studies in adults and a recent larger, prospectively randomized trial
in children (dubbed the TONIC study). Although several of the adult
NASH studies suggested improvements in aminotransferases and, less
consistently, liver histology, metformin did not improve liver histology
in the TONIC study of children with NASH. Thus, it is not currently
recommended as a treatment for NASH. Thiazolidinediones (pioglitazone and rosiglitazone), drugs known to improve systemic insulin resistance, have been studied in adults with NASH. Both agents reduced
aminotransferases and improved some of the histologic features of
NASH in small, uncontrolled studies. A large, randomized, placebocontrolled clinical trial sponsored by the National Institutes of Health,
the PIVENS Study (Pioglitazone vs Vitamin E vs Placebo for the
Treatment of 247 Nondiabetic Adults with NASH), demonstrated
that resolution of histologic NASH occurred more often in subjects
treated with pioglitazone (30 mg/d) than with placebo for 18 months
(47 vs 21%, p = .001). However, many subjects in the pioglitazone group
gained weight, and liver fibrosis did not improve. Five-year follow-up of
subjects who were treated with rosiglitazone for up to 2 years demonstrated that extending treatment and follow-up duration did not further
improve NASH or liver fibrosis, and rosiglitazone has been associated
with increased long-term risk for cardiovascular mortality. Pioglitazone
may be safer than rosiglitazone, however, because in a recent large
meta-analysis it was associated with reduced overall morality, myocardial infarction, and stroke. Caution is still warranted, however, because
long-term use of thiazolidinediones has been associated with weight
gain, increased risk for bladder cancer, and bone fractures in women.
Incretin mimetics, drugs that act on the pancreas to optimize insulin
and glucagon release, have improved liver enzyme elevations. A small
pilot trial of daily injections of liraglutide and phase 2 studies of semaglutide demonstrated remission of NASH without worsening liver fibrosis. Agents that improve hyperglycemia by blocking renal reabsorption
of glucose, sodium glucose cotransporter (SGLT2) inhibitors, have been
observed to improve serum liver enzymes in diabetic patients with
and are also under formal evaluation as treatments for NASH. Both
incretin mimetics and SGLT2 inhibitors can be used in NASH patients
with type 2 diabetes or obesity (conditions for which the drugs have
an FDA-registered indication for use); however, they are not currently
approved specifically for the treatment of NASH.
Antioxidants have also been evaluated for the treatment of NAFLD
because oxidant stress is thought to contribute to the pathogenesis of
NASH. Vitamin E, an inexpensive yet potent antioxidant, has been
examined in several small pediatric and adult studies with varying
results. In all of those studies, vitamin E was well tolerated, and most
studies showed modest improvements in aminotransferase levels,
radiographic features of hepatic steatosis, and/or histologic features of
NASH. Vitamin E (800 IU/d) was compared to placebo in the PIVENS
and TONIC studies. In PIVENS, vitamin E was the only agent that
achieved the predetermined primary endpoint (i.e., improvement
in steatohepatitis without worsening of fibrosis). This endpoint was
met in 43% of patients in the vitamin E group (p = .001 vs placebo),
34% in the pioglitazone group (p = .04 vs placebo), and 19% in the
placebo group. Vitamin E also improved NASH histology in pediatric
patients with NASH (TONIC trial). However, recent population-based
studies suggest that chronic vitamin E therapy may increase the risk
for cardiovascular mortality, hemorrhagic stroke, and prostate cancer.
Thus, vitamin E should only be considered as a first-line pharmacotherapy for nondiabetic, noncirrhotic NASH patients who are at low
risk for cardiovascular disease or prostate cancer. Further studies are
needed before firm recommendations can be made regarding the
risk-to-benefit ratio and long-term therapeutic efficacy of vitamin
E in NASH. Ursodeoxycholic acid (a bile acid that improves certain
cholestatic liver diseases) and betaine (a metabolite of choline that
raises S-adenosylmethionine [SAM] levels and decreases cellular
oxidative damage) offer no histologic benefit over placebo in patients
with NASH. Experimental evidence to support the use of omega-3 fatty
acids in NAFLD exists; however, a recent large, multicenter, placebocontrolled study failed to demonstrate a histologic benefit.
Many other pharmacotherapies that target dysregulated energy
homeostasis, lipotoxicity, cell death, and liver inflammation, processes
that are critically involved in the pathogenesis and/or progression of
NASH and liver fibrosis, are currently in clinical trials (e.g., probiotics,
farnesoid X receptor agonists, fibroblast growth factor agonists, antiapoptotic agents, anticytokine agents, dipeptidyl IV antagonists, PPAR
modulators, thyroid hormone receptor β-selective agonists, stearyl-CoA
desaturase-1 inhibitors, DGAT inhibitors, acyl-CoA carboxylase inhibitors, and direct modulators of liver fibrosis). Sufficient data do not yet
exist to justify their use as NASH treatments in clinical practice. Given
that liver disease outcomes in NASH patients are highly heterogeneous,
optimal treatment of NASH may need to be individualized by tailoring
therapy based on clinical or histologic phenotypes of NASH and/or
genetic susceptibility for disease progression.
Statins are an important class of agents to treat dyslipidemia and
decrease cardiovascular risk. There is no evidence to suggest that
statins cause liver failure in patients with any chronic liver disease,
including NAFLD. The incidence of liver enzyme elevations in NAFLD
patients taking statins is also no different than that of healthy controls
or patients with other chronic liver diseases. Moreover, several studies
have suggested that statins may improve aminotransferases and histology in patients with NASH. Yet there is continued reluctance to
use statins in patients with NAFLD. The lack of evidence that statins
harm the liver in NAFLD patients, combined with the increase risk for
cardiovascular morbidity and mortality in NAFLD patients, justifies
the use of statins to treat dyslipidemia in patients with NAFLD/NASH.
Bariatric Surgery Although interest in bariatric surgery as a
treatment for NAFLD exists, a recently published Cochrane review
concluded that lack of randomized clinical trials or adequate clinical
studies prevents definitive assessment of benefits and harms of bariatric surgery as a treatment for NASH. Most studies of bariatric surgery
have shown that bariatric surgery is generally safe in individuals with
well-compensated chronic liver disease and improves hepatic steatosis
and necroinflammation (i.e., features of NAFLD/NASH); however,
effects on hepatic fibrosis have been variable. Concern lingers because
some of the largest prospective studies suggest that hepatic fibrosis
might progress after bariatric surgery. Thus, the Cochrane review
deemed it premature to recommend bariatric surgery as a primary
treatment for NASH. This opinion was challenged by a recently study
that demonstrated that fibrosis stage had improved by 5 years after
surgery in about half the patients in one large bariatric surgery cohort.
However, most of those individuals had relatively mild fibrosis initially,
and thus, it is unclear if similar outcomes would occur in individuals
with more advanced liver disease. Indeed, there is general agreement that patients with NAFLD-related cirrhosis and, particularly,
those with portal hypertension should be excluded as candidates for
bariatric surgery. However, given growing evidence for the benefits of
bariatric surgery on metabolic syndrome complications in individuals
with refractory obesity, it is not contraindicated in otherwise eligible
patients with NAFLD or NASH.
Liver Transplantation Patients with NAFLD in whom end-stage
liver disease develops should be evaluated for liver transplantation
(Chap. 345). The outcomes of liver transplantation in well-selected
patients with NAFLD are generally good, but comorbid medical conditions associated with NAFLD, such as diabetes mellitus, obesity, and
cardiovascular disease, often limit transplant candidacy. NAFLD may
recur after liver transplantation. The risk factors for recurrent or de
novo NAFLD after liver transplantation are multifactorial and include
hypertriglyceridemia, obesity, diabetes mellitus, and immunosuppressive therapies, particularly glucocorticoids.
2624 PART 10 Disorders of the Gastrointestinal System
■ GLOBAL HEALTH CONSIDERATIONS
Obesity is an accelerating global disease. The worldwide prevalence
of obesity has more than doubled since 1980, and there are now >1
billion overweight adults, of whom at least 300 million are obese. In
the wake of the obesity epidemic follow numerous comorbidities,
including NAFLD. NAFLD is the most common liver disease identified
in Western countries and the fastest rising form of chronic liver disease
worldwide. The economic burden directly attributable to NAFLD is
already enormous (estimated direct medical costs of ~$103 billion/year
in the United States and €35 billion/year in the Europe-4 countries:
Germany, France, Italy, and United Kingdom) and predicted to increase
tenfold by the year 2025. Present understanding of NAFLD’s natural
history is based mainly on studies in whites who became overweight/
obese and developed the metabolic syndrome in adulthood. The
impact of the global childhood obesity epidemic on NAFLD pathogenesis/progression is unknown. Emerging evidence demonstrates that
advanced NAFLD, including cirrhosis and primary liver cancer, can
occur in children, prompting concerns that childhood-onset NAFLD
might follow a more aggressive course than typical adult-acquired
NAFLD. Some of the most populated parts of the world are in the
midst of industrial revolutions, and certain environmental pollutants
seem to exacerbate NAFLD. Some studies also suggest that the risk for
NASH and NAFLD-related cirrhosis may be higher in certain ethnic
groups such as Asians, Hispanics, and Native Americans, and lower in
others such as African Americans, compared with whites. Although
all of these variables confound efforts to predict the net impact of
this obesity-related liver disease on global health, it seems likely that
NAFLD will remain a major cause of chronic liver disease worldwide
for the foreseeable future.
■ FURTHER READING
Chalasani N et al: The diagnosis and management of nonalcoholic
fatty liver disease: Practice guidance from the American Association
for the Study of Liver Diseases. Hepatology 67:328, 2018.
Diehl AM, Day CSC: Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis. N Engl J Med 377:2063, 2017.
European Association for the Study of the Liver (EASL) et al:
EASL-EASD-EASO Clinical Practice Guidelines for the management
of non-alcoholic fatty liver disease. J Hepatol 64:1388, 2016.
Vos MB et al: NASPGHAN clinical practice guideline for the diagnosis
and treatment of nonalcoholic fatty liver disease in children: Recommendations from the Expert Committee on NAFLD (ECON) and the
North American Society of Pediatric Gastroenterology, Hepatology
and Nutrition (NASPGHAN). J Pediatr Gastroenterol Nutr 64:319,
2017.
Cirrhosis is a condition that is defined histopathologically and has a
variety of clinical manifestations and complications, some of which
can be life-threatening. In the past, it has been thought that cirrhosis
was never reversible; however, it has become apparent that when the
underlying insult that has caused the cirrhosis has been removed, there
can be reversal of fibrosis. This is most apparent with the successful
treatment of chronic hepatitis C; however, reversal of fibrosis is also
seen in patients with hemochromatosis who have been successfully
treated and in patients with alcohol associate liver disease who have
discontinued alcohol use.
Regardless of the cause of cirrhosis, the pathologic features consist
of the development of fibrosis to the point that there is architectural
344 Cirrhosis and Its
Complications
Alex S. Befeler, Bruce R. Bacon
TABLE 344-1 Causes of Cirrhosis
Alcohol Cardiac cirrhosis
Chronic viral hepatitis Inherited metabolic liver disease
Hepatitis B Hemochromatosis
Hepatitis C Wilson’s disease
Autoimmune hepatitis α1
Antitrypsin deficiency
Nonalcoholic steatohepatitis Cystic fibrosis
Biliary cirrhosis Cryptogenic cirrhosis
Primary biliary cholangitis
Primary sclerosing cholangitis
Autoimmune cholangiopathy
distortion with the formation of regenerative nodules. This results in a
decrease in hepatocellular mass, and thus function, and an alteration of
blood flow. The induction of fibrosis occurs with activation of hepatic
stellate cells, resulting in the formation of increased amounts of collagen and other components of the extracellular matrix.
Clinical features of cirrhosis are the result of pathologic changes
and mirror the severity of the liver disease. Most hepatic pathologists
provide an assessment of grading and staging when evaluating liver
biopsy samples. These grading and staging schemes vary between
disease states and have been developed for most conditions, including
chronic viral hepatitis, nonalcoholic fatty liver disease, and primary
biliary cholangitis. Advanced fibrosis usually includes bridging fibrosis
with nodularity designated as stage 3 and cirrhosis designated as stage 4.
Patients who have cirrhosis have varying degrees of liver function, and
clinicians need to differentiate between those who have stable, compensated cirrhosis and those who have decompensated cirrhosis. Patients
who have developed ascites, hepatic encephalopathy, or variceal bleeding are classified as decompensated. They should be considered for
liver transplantation, particularly if the decompensations are poorly
controlled. Many of the complications of cirrhosis will require specific
therapy. Portal hypertension is a significant complicating feature of
decompensated cirrhosis and is responsible for the development of
ascites and bleeding from esophagogastric varices, two complications
that signify decompensated cirrhosis. Loss of hepatocellular function
results in jaundice, coagulation disorders, and hypoalbuminemia and
contributes to the causes of portosystemic encephalopathy. The complications of cirrhosis are basically the same regardless of the etiology.
Nonetheless, it is useful to classify patients by the cause of their liver
disease (Table 344-1); patients can be divided into broad groups,
including those with alcohol-associated cirrhosis, cirrhosis due to
chronic viral hepatitis, biliary cirrhosis, nonalcoholic fatty liver disease,
and other, less common causes, such as cardiac cirrhosis, cryptogenic
cirrhosis, and other miscellaneous causes.
ALCOHOL-ASSOCIATED CIRRHOSIS
Excessive chronic alcohol use can cause several different types of
chronic liver disease, including alcohol-associated fatty liver, alcoholic hepatitis, and alcohol-associated cirrhosis. Furthermore, use of
excessive alcohol can contribute to liver damage in patients with other
liver diseases, such as hepatitis C, hemochromatosis, and fatty liver
disease related to obesity. Chronic alcohol use can produce fibrosis in
the absence of accompanying inflammation and/or necrosis. Fibrosis
can be centrilobular, pericellular, or periportal. When fibrosis reaches
a certain degree, there is disruption of the normal liver architecture
and replacement of liver cells by regenerative nodules. In alcoholassociated cirrhosis, the nodules are usually <3 mm in diameter; this
form of cirrhosis is referred to as micronodular. With cessation of alcohol use, larger nodules may form, resulting in a mixed micronodular
and macronodular cirrhosis.
Pathogenesis Alcohol is the most commonly used drug in the
United States, and >70% of adults drink alcohol each year. Twenty
percent have had a binge within the past month, and >7% of adults
regularly consume more than four or five drinks five or more times
a month. Unfortunately, >14 million adults in the United States meet
2625Cirrhosis and Its Complications CHAPTER 344
the diagnostic criteria for alcohol use disorder. In the United States,
chronic liver disease is the tenth most common cause of death in
adults, and alcohol-associated cirrhosis accounts for ~48% of deaths
due to cirrhosis.
Ethanol is mainly absorbed by the small intestine and, to a lesser
degree, through the stomach. Gastric alcohol dehydrogenase (ADH)
initiates alcohol metabolism. Three enzyme systems account for
metabolism of alcohol in the liver. These include cytosolic ADH, the
microsomal ethanol oxidizing system (MEOS) utilizing the inducible
cytochrome P450 CYP2E1, and peroxisomal catalase. Normally the
majority of ethanol oxidation occurs via ADH to form acetaldehyde,
which is a highly reactive molecule that may have multiple effects. The
MEOS pathway in chronic alcohol use causes induction of CYP2E1,
which leads to generation of reactive oxygen species and produces
more acetaldehyde. Ultimately, acetaldehyde is metabolized to acetate
by aldehyde dehydrogenase (ALDH). Intake of ethanol increases intracellular accumulation of triglycerides by increasing fatty acid uptake
and by reducing fatty acid oxidation and lipoprotein secretion. Protein
synthesis, glycosylation, and secretion are impaired. Oxidative damage
to hepatocyte membranes occurs due to the formation of reactive oxygen species; acetaldehyde is a highly reactive molecule that combines
with proteins and nucleic acids to form acetaldehyde adducts. These
adducts may interfere with specific enzyme activities, including microtubular formation and hepatic protein trafficking. With acetaldehydemediated hepatocyte damage, certain reactive oxygen species can result
in Kupffer cell activation. As a result, profibrogenic cytokines are produced that initiate and perpetuate stellate cell activation, with the resultant production of excess collagen and extracellular matrix. Connective
tissue appears in both periportal and pericentral zones and eventually
connects portal triads with central veins forming regenerative nodules.
Hepatocyte loss occurs, and with increased collagen production and
deposition, together with continuing hepatocyte destruction, the liver
contracts and shrinks in size. This process generally takes from years
to decades to occur and requires repeated insults.
Clinical Features The diagnosis of alcohol associate liver disease
requires an accurate history regarding both amount and duration of
alcohol consumption. Patients with alcohol associate liver disease can
present with nonspecific symptoms such as vague right upper quadrant
abdominal pain, fever, nausea and vomiting, diarrhea, anorexia, and
malaise. Alternatively, they may present with more specific complications
of chronic liver disease, including ascites, edema, upper gastrointestinal
(GI) hemorrhage, jaundice, or encephalopathy. Many cases present incidentally at the time of autopsy or elective surgery. The abrupt onset of
any of these complications may be the first event prompting the patient to
seek medical attention. Other patients may be identified in the course of
an evaluation of routine laboratory studies that are found to be abnormal.
On physical examination, the liver and spleen may be enlarged, with the
liver edge being firm and nodular. Other frequent findings include scleral
icterus, palmar erythema (Fig. 344-1), spider angiomas (Fig. 344-2),
parotid gland enlargement, digital clubbing, muscle wasting, edema, and
ascites. Men may have decreased body hair and gynecomastia as well
as testicular atrophy, which may be a consequence of hormonal abnormalities or a direct toxic effect of alcohol on the testes. In women with
advanced alcohol-associated cirrhosis, menstrual irregularities usually
occur including amenorrhea. These changes are often reversible following
cessation of alcohol ingestion.
Laboratory tests may be completely normal in patients with early
compensated alcohol-associated cirrhosis. Alternatively, in advanced
liver disease, many abnormalities usually are present. Patients may be
anemic from chronic GI blood loss, nutritional deficiencies, or hypersplenism or as a direct suppressive effect of alcohol on the bone marrow.
A unique form of hemolytic anemia (with spur cells and acanthocytes)
called Zieve’s syndrome can occur in patients with severe alcoholic hepatitis. Platelet counts are often reduced early in the disease, reflective of
portal hypertension with hypersplenism. Serum total bilirubin can be
normal or elevated with advanced disease. Prothrombin times are often
prolonged and usually do not respond to administration of parenteral
vitamin K. Serum sodium levels are usually normal unless patients have
ascites and then can be depressed, largely due to ingestion of excess free
water. Serum alanine and aspartate aminotransferases (ALT, AST) are
typically elevated, particularly in patients who continue to drink, with
AST levels being higher than ALT levels, usually by a 2:1 ratio.
Diagnosis Patients who have any of the above-mentioned clinical
features, physical examination findings, or laboratory studies should be
considered to have alcohol associate liver disease. The diagnosis, however, requires accurate knowledge that the patient is continuing to use
or has recently stopped alcohol. Furthermore, other forms of chronic
liver disease (e.g., chronic viral hepatitis or metabolic or autoimmune
liver diseases) must be considered or ruled out, or if present, an estimate of relative causality along with the alcohol use should be determined. Liver biopsy can be helpful to confirm a diagnosis but generally
is not performed unless there is a suspicion of an alternative diagnosis.
In patients who have had complications of cirrhosis and who
continue to drink, there is a <50% 5-year survival. In contrast, in
patients who are able to remain abstinent, the prognosis is significantly
improved, particularly when they have resolution of liver complications; however, some individuals who remain abstinent do not improve
and liver transplantation is a viable option.
TREATMENT
Alcohol-Associated Cirrhosis and Alcoholic Hepatitis
Abstinence is the cornerstone of therapy for patients with alcohol
associate liver disease. In addition, patients require good nutrition
and long-term medical supervision to manage underlying complications that may develop. Complications such as the development of
FIGURE 344-1 Palmar erythema. This figure shows palmar erythema in a patient with
alcohol-associated cirrhosis. The erythema is peripheral over the palm with central pallor.
FIGURE 344-2 Spider angioma. This figure shows a spider angioma in a patient with
hepatitis C cirrhosis. With release of central compression, the arteriole fills from the
center and spreads out peripherally.
2626 PART 10 Disorders of the Gastrointestinal System
ascites and edema, variceal hemorrhage, or portosystemic encephalopathy all require specific management and treatment. Liver transplantation can be an effective long-term treatment in those who
have been deemed a low enough risk for alcohol relapse and do not
respond to other treatments.
Glucocorticoids are occasionally used in patients with severe
alcoholic hepatitis in the absence of infection. Short-term survival
has been shown to be improved in certain studies and meta-analysis,
although 6-month survival is more dependent on abstinence. Treatment is restricted to patients with a discriminant function (DF)
value of >32. The DF is calculated as the serum total bilirubin plus
the difference in the patient’s prothrombin time compared to upper
limit of control (in seconds) multiplied by 4.6. Failure to improve
total bilirubin after 7 days predicts treatment failure, and glucocorticoids can be stopped; otherwise, they are continued for 28 days.
There is modest evidence that intravenous N-acetylcysteine plus
glucocorticoids may have survival benefit in alcoholic hepatitis if
the DF is >32. Other therapies including oral pentoxifylline, parenterally administered inhibitors of tumor necrosis factor (TNF) α
such as infliximab or etanercept, anabolic steroids, propylthiouracil,
antioxidants, colchicine, and penicillamine have not shown clearcut benefits and are not recommended. A variety of nutritional
therapies have been tried, both parenteral and enteral feedings;
however, there is no clear evidence of improved survival. There
is evidence that persons who consume >21.5 kcal/kg body weight
per day have better survival, so achieving better caloric intake is
recommended. Finally, in highly selected patients with good social
support structure who fail other treatments for alcoholic hepatitis,
early liver transplant can be an effective treatment.
The cornerstone to treatment is cessation of alcohol use. Recent
experience with medications that reduce craving for alcohol, such
as acamprosate calcium and baclofen, have been favorable. Patients
may take other necessary medications even in the presence of cirrhosis. Acetaminophen use is often discouraged in patients with
liver disease; however, if no more than 2 g of acetaminophen per
day are consumed, there generally are no problems unless there is
active alcohol use.
■ CIRRHOSIS DUE TO CHRONIC VIRAL
HEPATITIS B OR C
Of patients exposed to the hepatitis C virus (HCV), ~80% develop
chronic hepatitis C, and of those, ~20–30% will develop cirrhosis over
20–30 years. Many of these patients have had concomitant alcohol
use, and the true incidence of cirrhosis due to hepatitis C alone is
unknown. It is expected that an even higher percentage will go on to
develop cirrhosis over longer periods of time. In the United States,
~5–6 million people have been exposed to HCV, and ~4–5 million are
chronically viremic. Worldwide, ~170 million individuals have hepatitis C, with some areas of the world (e.g., Egypt) having up to 15%
of the population infected. HCV is a noncytopathic virus, and liver
damage is probably immune-mediated. Progression of liver disease
due to chronic hepatitis C is characterized by portal-based fibrosis with
bridging fibrosis and nodularity developing, ultimately culminating in
the development of cirrhosis. In cirrhosis due to chronic hepatitis C,
the liver is small and shrunken with characteristic features of a mixed
micro- and macronodular cirrhosis seen on liver biopsy. In addition
to the increased fibrosis that is seen in cirrhosis due to hepatitis C, an
inflammatory infiltrate is found in portal areas with interface hepatitis
and occasionally some lobular hepatocellular injury and inflammation.
In patients with HCV genotype 3, steatosis is often present.
Similar findings are seen in patients with cirrhosis due to chronic
hepatitis B. Of adult patients exposed to hepatitis B, ~5% develop
chronic hepatitis B, and ~20% of those patients will go on to develop
cirrhosis. Special stains for hepatitis B core (HBc) and hepatitis B
surface (HBs) antigen will be positive, and ground-glass hepatocytes
signifying HBs antigen (HBsAg) may be present. In the United States,
there are ~2 million carriers of hepatitis B, whereas in other parts
of the world where hepatitis B virus (HBV) is endemic (i.e., Asia,
Southeast Asia, sub-Saharan Africa), up to 15% of the population
may be infected, having acquired the infection vertically at the time of
birth. Thus, >300–400 million individuals are thought to have hepatitis
B worldwide. Approximately 25% of these individuals may ultimately
develop cirrhosis.
Clinical Features and Diagnosis Patients with cirrhosis due to
either chronic hepatitis C or B can present with the usual symptoms
and signs of chronic liver disease. Fatigue, malaise, vague right upper
quadrant pain, and laboratory abnormalities are frequent presenting
features. Diagnosis requires a thorough laboratory evaluation, including quantitative HCV RNA testing and analysis for HCV genotype, or
hepatitis B serologies to include HBsAg, anti-HBs, HBeAg (hepatitis B
e antigen), anti-HBe, and quantitative HBV DNA levels.
TREATMENT
Cirrhosis due to Chronic Viral Hepatitis B or C
Management of complications of cirrhosis revolves around specific
therapy for treatment of whatever complications occur (e.g., esophageal variceal hemorrhage, development of ascites and edema, or
encephalopathy). In patients with chronic hepatitis B, numerous
studies have shown beneficial effects of antiviral therapy, which is
effective at viral suppression, as evidenced by reducing aminotransferase levels and HBV DNA levels and improving histology by
reducing inflammation and fibrosis. Several clinical trials and case
series have demonstrated that patients with decompensated liver
disease can become compensated with the use of antiviral therapy
directed against hepatitis B. Currently available therapy includes
lamivudine, adefovir, telbivudine, entecavir, and tenofovir, with the
latter two being preferred because of reduced risk of viral resistance.
Interferon α can also be used for treating hepatitis B, but it should
not be used in cirrhotics (see Chap. 341).
Treatment of patients with cirrhosis due to hepatitis C used to be
more difficult because the side effects of pegylated interferon and
ribavirin therapy were difficult to manage. Over the past several
years, interferon-based regimens have been replaced by directacting antiviral protocols that are highly successful (>95% cure
rate), well tolerated, and usually of short duration (8–12 weeks), but
costly. These medications have truly revolutionized the treatment of
hepatitis C (see Chap. 341).
CIRRHOSIS FROM AUTOIMMUNE
HEPATITIS AND NONALCOHOLIC FATTY
LIVER DISEASE
Other causes of posthepatitic cirrhosis include autoimmune hepatitis
(AIH) and cirrhosis due to nonalcoholic steatohepatitis. Many patients
with AIH present with cirrhosis that is already established. Typically,
these patients will not benefit from immunosuppressive therapy with
glucocorticoids or azathioprine because the AIH is “burned out.” In
this situation, liver biopsy does not show a significant inflammatory
infiltrate. Diagnosis in this setting requires positive autoimmune
markers such as antinuclear antibody (ANA) or anti-smooth-muscle
antibody (ASMA). When patients with AIH present with cirrhosis and
active inflammation accompanied by elevated liver enzymes, there can
be considerable benefit from the use of immunosuppressive therapy.
Patients with nonalcoholic steatohepatitis are increasingly being
found to have progressed to cirrhosis. With the epidemic of obesity
that continues in Western countries, more and more patients are
identified with nonalcoholic fatty liver disease (Chap. 343). Of these,
a significant subset has nonalcoholic steatohepatitis and can progress
to increased fibrosis and cirrhosis. Over the past several years, it has
been increasingly recognized that many patients who were thought to
have cryptogenic cirrhosis in fact have nonalcoholic steatohepatitis.
As their cirrhosis progresses, they become catabolic and then lose the
telltale signs of steatosis seen on biopsy. Management of complications
of cirrhosis due to either AIH or nonalcoholic steatohepatitis is similar
to that for other forms of cirrhosis.
No comments:
Post a Comment
اكتب تعليق حول الموضوع