Diagnosis and Management of Hemochromatosis


AASLD Practice Guidelines
Diagnosis and Management of Hemochromatosis
ANTHONY S. TAVILL
PREAMBLE BACKGROUND
Practice guidelines, intended for use by physicians, suggest Hereditary hemochromatosis (HH) is the most common,
preferable approaches to the diagnostic, therapeutic, and pre- identified, genetic disorder in the Caucasian population. Al-
though its geographic distribution is worldwide, it is concen-
ventative aspects of care. These guidelines are intended to be
trated in individuals of northern European origin, particularly
flexible, in contrast with  standards of care, which are inflex-
of Nordic or Celtic ancestry, in whom it occurs with a preva-
ible policies to be followed in almost every case.1 They are
lence close to 1 per 200 of the population.4-6 The pathophys-
developed in a manner consistent with the American Gastro-
iologic predisposition to increased and inappropriate absorp-
enterological Associations Policy Statement on Development
tion of dietary iron may lead to the progressive development
and Use of Practice Guidelines.2
of life-threatening complications of cirrhosis, hepatocellular
Specific recommendations are based on relevant published
cancer, diabetes, and heart disease. The gene defect described
information. In an attempt to standardize recommendations,
in 19967 isaGtoAmissense mutation (C282Y) leading to the
the Practice Guidelines Committee of the American Associa-
substitution of tyrosine for cysteine at the 282 amino acid
tion for the Study of Liver Diseases modified the categories of
position of the protein product of the newly discovered HFE
the Infectious Diseases Society of America s Quality Stan-
gene located on the short arm of chromosome 6 (6p). Another
dards.3 These categories are reported with each recommenda-
mutation (H63D) in which aspartic acid is substituted for
tion, using the letters A through E to determine the strength of
histidine at position 63 has also been associated as a cofactor
recommendation (Table 1) and Roman numerals I through IV
in some cases of hemochromatosis. The homozygous state in
to determine quality of evidence upon which recommenda-
which both alleles of chromosome 6 possess the C282Y mu-
tions are based (Table 2).
tation or the compound heterozygous state with C282Y on
These guidelines provide data-supported peer-reviewed
one chromosome and H63D on the other, are the predomi-
recommendations for the care of patients with hemochroma-
nant genetic abnormalities associated with phenotypic HH. In
tosis. They are based on the following: (1) a formal review and
most studies to date, C282Y/C282Y homozygosity has been
analysis of the recent published literature on hemochromato-
found in more than 90% of patients with hemochromatosis,
sis (Medline Search from 1990-2000); (2) the American Col-
while compound heterozygosity (C282Y/H63D) accounts for
lege of Physicians Manual for Assessing Health Practices and
3% to 5% of such cases in published series. Possession of the
Designing Practice Guidelines; (3) several published guide- C282Y mutation on both alleles of the chromosome pair has a
lines, including the American Association for the Study of
high positive predictive accuracy for phenotypic HH. In the
Liver Diseases Policy Statement on Development and Use of Prac- only large population study published to date in which pen-
tice Guidelines and the American Gastroenterological Associa- etrance of the HFE gene mutation has been studied compre-
hensively, all C282Y homozygotes had elevated transferrin
tion s Policy Statement on Guidelines2; and (4) the experience
of the author in the clinical care of patients with hemochro- saturation (100% positive predictive accuracy). However, full
expression as defined by progressive tissue iron overload oc-
matosis.
curred in only 58% of these homozygotes.6
Although the vast majority of familial cases of hemochro-
matosis in the Anglo-Celtic population are associated with the
described pathogenic mutations of the HFE gene, it is highly
Abbreviations: HH, hereditary hemochromatosis; HIC, hepatic iron concentration;
probable that genes other than HFE play a role in familial iron
HII, hepatic iron index; HCC, hepatocellular carcinoma; TIBC, total iron-binding capac-
ity; UIBC, unsaturated iron-binding capacity. overload in other populations. In particular, there are well
From the MetroHealth Medical Center, and Case Western Reserve University, Cleve-
documented families in Italy with iron overload comparable
land, OH.
with HFE-related hemochromatosis,8-10 in whom neither the
Received February 2, 2001; accepted March 16, 2001.
C282Y nor H63D mutation existed, and in whom the genetic
This Guideline has been commissioned and approved by the American Association for
abnormality could not be located to chromosome 6p.
the Study of Liver Diseases and has received the endorsement of the American College of
Gastroenterology and the American Gastroenterological Association.
The clinical condition of hereditary hemochromatosis
Address correspondence to: Anthony S. Tavill, M.D., Gastroenterology Division,
evolves in a series of stages beginning with clinically insignif-
MetroHealth Medical Center, 2500 MetroHealth Dr., Cleveland, OH 44109. E-mail:
icant iron accumulation (0-20 years of age, 0-5 g parenchymal
ast2@po.cwru.edu; fax: 216-778-4873.
iron storage). This evolves to a stage of iron overload without
Individual copies of these guidelines can be obtained from the AASLD website at
www.aasld.org. For multiple reprints (100 copies or more) contact W.B. Saunders Com- disease (approximately 20-40 years of age, 10-20 g parenchy-
pany, The Curtis Center, Independence Square West, Philadelphia, PA 19106-3399, and
mal iron storage), which if left untreated, may progress to a
obtain permission from Anthony S. Tavill.
stage of iron overload with organ damage (usually more than
Copyright © 2001 by the American Association for the Study of Liver Diseases.
40 years of age and 20 g parenchymal iron storage).11,12
0270-9139/01/3305-0038$35.00/0
doi:10.1053/jhep.2001.24783 Ideally, any strategy for diagnosis should identify cases before
1321
1322 TAVILL HEPATOLOGY May 2001
TABLE 1. Categories Reflecting the Evidence to Support the Use of a TABLE 3. Management Objectives for HH
Guideline Recommendation
Management Objectives
Category Definition
Early diagnosis to prevent organ damage and dysfunction due to tissue iron
A Survival benefit toxicity
B Improved diagnosis Screening and early detection of asymptomatic HH cases to reduce
C Improvement in quality of life mortality
D Relevant pathophysiologic parameters improved Recognition and diagnosis of symptomatic cases of HH, to minimize
E Impacts cost of health care progression and complications of the disease
Adequate treatment of HH to promote rapid, safe, and effective removal of
Adapted and modified from Gross et al.3
iron
Vigilant follow-up and maintenance treatment of all cases of HH
the third stage of disease has developed so that therapy to
remove iron can prevent progression to irreversible tissue
The degree of iron overload has a direct impact on life
damage. Fortunately, biochemical serum testing with indirect
expectancy of the individual with HH. The major causes of
iron markers is capable of identifying most cases of iron over-
death are decompensated cirrhosis, hepatocellular carcinoma
load well before tissue damage has become irreversible.
(HCC), diabetes mellitus, and cardiomyopathy.15 These oc-
Therefore, these guidelines will emphasize the fundamental
curred with a frequency 10- to 119-fold higher than expected
objective of detection of HH before organ damage has oc-
in an age- and sex-matched population without HH. Survival
curred (Table 3).
was normal in HH patients in whom treatment was initiated
Current clinical practice in diagnosis and management of
before the development of cirrhosis or diabetes, confirming
HH has evolved from experience in screening healthy blood
the importance of early diagnosis and treatment.
donors or selected populations and managing patients and
DIAGNOSIS OF HEREDITARY HEMOCHROMATOSIS
their discovered relatives with phenotypic HH.6,12-14 Early in-
stitution of phlebotomy has proven to be a highly effective
Target Populations
therapy for HH, which prevents morbidity and promotes nor-
Target populations are shown in Table 4. The diagnosis of
mal longevity.15 As a result, randomized, controlled trials of
hemochromatosis is based on documentation of increased
other therapies or observation have been regarded as unethi-
iron stores, namely increased hepatic iron concentrations as-
cal and have not been done.
sociated with elevated serum ferritin levels. HH can be further
The development of liver injury in those with HH is related
defined genotypically by the familial occurrence of iron over-
to the progressive accumulation of hepatic iron.15-18 Hepatic
load associated with C282Y homozygosity or C282Y/H63D
iron concentration increases with age in most homozygotes.
compound heterozygosity.20 As serologic iron markers have
In HH patients over the age of 40 years, hepatic iron concen-
become more widely available over the last several years, the
tration is likely to exceed 10,000 g/g dry weight and liver
biopsy results are more likely to show fibrosis or cirrhosis.16-18 majority of patients with HH are now identified while still
asymptomatic and without evidence of hepatic fibrosis or cir-
It was the observation that the hepatic iron concentration
rhosis.15 Diagnostic screening strategies should target high-
(HIC) increased with age that led to the concept of hepatic
risk groups such as those with suspicious organ involvement,
iron index (HIC in micromoles per gram dry weight divided
a familial history of HH, and those with chance detection of
by age in years). A hepatic iron index (HII) in excess of 1.9
biochemical or radiologic abnormalities suggestive of the pos-
mol/g per year of life was found to effectively distinguish
sibility of iron overload.
homozygous hemochromatosis from heterozygotes and pa-
tients with alcohol-induced liver disease. However, it is now
clear that the rate of iron accumulation is variable and excep-
TABLE 4. Target Populations for Screening for HH
tions may occur in between 8% and 50% of individuals with
Target Populations for Hemochromatosis Evaluation
HH.6,12,14,19 Therefore, while an HII less than 1.9 does not
entirely exclude HH, a value greater than this certainly docu- Symptomatic patients
Unexplained manifestations of liver disease or a presumably known
ments significant iron overload in the C282Y homozygote and
cause of liver disease with abnormality of one or more indirect serum
in individuals with certain forms of secondary iron overload.
iron markers
Type 2 diabetes mellitus, particularly with hepatomegaly, elevated liver
enzymes, atypical cardiac disease or early-onset sexual dysfunction
TABLE 2. Quality of Evidence on Which Recommendation Is Based Early-onset atypical arthropathy, cardiac disease, and male sexual
dysfunction
Grade Definition
Asymptomatic patients
I Evidence from multiple well-designed randomized controlled Priority groups
trials each involving a number of participants to be of First-degree relatives of a confirmed case of hemochromatosis
sufficient statistical power Individuals with abnormal serum iron markers discovered during
II Evidence from at least one large well-designed clinical trial routine testing
with or without randomization, from cohort or case-control Individuals with unexplained elevation of liver enzymes or the
analytic studies, or well-designed meta-analysis serendipitous finding of asymptomatic hepatomegaly or radiologic
III Evidence based on clinical experience, descriptive studies, or detection of enhanced computed tomography attenuation of the
reports of expert committees liver
IV Not rated General population
See Fig. 1.
Adapted and modified from Gross et al.3
HEPATOLOGY Vol. 33, No. 5, 2001 TAVILL 1323
derived by dividing the serum iron by the total iron binding
capacity. When the fasting value exceeds 50% for women and
60% for men, TS has a sensitivity of 0.92, a specificity of 0.93,
and a positive predictive value of 86% for the diagnosis of
HH.27-29 Overnight fasting avoids circadian or postprandial
variations and eliminates 80% of false-positive TS results.27
Lowering the cutoff TS value to 45% increases sensitivity, but
reduces specificity and positive predictive value. In a recent
study, values exceeding 45% correctly identified 97.9% of ho-
mozygotes with no false positives among the normal popula-
tion.30 However, this cutoff did include 22.2% of the hetero-
zygote population, a group recognized to occasionally have
phenotypic markers of iron overload. In another report, this
cutoff was 100% sensitive for the detection of C282Y homozy-
gotes; however, only 44% of those with TS more than 45%
were genetic homozygotes.6 Thus, lowering the threshold for
TS to 45% will also identify other groups with relatively minor
degrees of secondary iron overload (e.g., alcohol-induced
liver disease, steatohepatitis, chronic hepatitis C, previous
FIG. 1. Proposed algorithm for management of HH.
surgical portacaval shunt, etc.) and these cases will require
further evaluation by the clinician.
In most clinical laboratories TS was customarily measured
Evidence is accumulating to support the cost effectiveness of
by determining two serum iron measurements; the first a mea-
serologic strategies for screening the general population for iron
surement of the subjects fasting serum iron, the second a
overload.13,21-25 Most of these reports have only assessed the use-
repeat measurement after adding exogenous iron to saturate
fulness of standard serologic tests such as serum iron, transferrin
the serum transferrin followed by removal of the nontrans-
saturation, or serum ferritin; only one study has included the re-
ferrin-bound iron. The latter determines the total iron binding
cently discovered HFE gene mutation.25 This latter study com-
capacity (TIBC). The ratio serum iron to TIBC gives the trans-
pared screening of blood donors by phenotypic or genotypic meth-
ferrin saturation (TS in percent). Although the serum iron is
ods. It was concluded that the most cost-effective strategy for
an automated test, the TIBC is not, making the traditional
identifying cases in the general population was phenotypic screen-
ing (standard iron markers) with genotypic confirmation of ho- method for deriving TS relatively expensive. Alternatively, it
has been proposed that costs could be reduced by using un-
mozygosity in those with indirect markers of iron overload
saturated iron binding capacity (UIBC).31 Values for UIBC
($2,700 per case). This strategy had a high predictive value for the
less than 28 mol/L are indicative of iron overload. In fact,
detection of homozygotes with iron overload and remained cost
many laboratories now determine TIBC by summing serum
effective even when it was assumed that as few as 20% of cases
would ever develop life-threatening complications of the dis- iron and UIBC (both automated methods). TS is then ex-
ease.25,26 In contrast, genotypic screening (by mutation analysis) pressed as the ratio of serum iron to the calculated TIBC:
of the general population would be prohibitively expensive Fe/(Fe UIBC). This allows the clinician to judge the signif-
($110,000 to detect one case) and the strategy would have speci- icance of a raised TS, by noting those that might be spuriously
ficity limitations in the light of accumulating evidence for the in- elevated by a low TIBC (low serum transferrin concentration).
complete penetrance of the gene mutations.6 These limitations It is recommended that TS be calculated in this way to reduce
may become less important as newer and less expensive tech- costs, particularly for large-scale screening.
niques of mutation analysis are developed. At this time we are
Other indirect markers of iron stores such as serum iron or
supportive of a low-cost phenotypic approach for screening the
ferritin lack specificity when used alone. The serum iron has
general population.
positive and negative predictive values for HH of 61% and
Data on sensitivity, specificity, and predictive value of phe-
87%, respectively, compared with 74% and 93% for TS.29 Se-
notypic screening tests have been provided by studies both in
rum ferritin is also nonspecific particularly in the face of in-
asymptomatic populations (e.g., healthy blood donors and
flammatory conditions, chronic hepatitis C, alcohol-induced
large-scale screening of a healthy population) and in families
liver disease, and neoplastic diseases. However, a serum fer-
of detected homozygotes.6,27-29 More recent studies are avail-
ritin level in combination with TS has a negative predictive
able for sensitivity and specificity of genotyping studies.6,12,20
value of 97% and exceeds the accuracy of any of the indirect
The following diagnostic algorithm proceeds in 3 steps, begin-
tests used in isolation.29 In confirmed HH, a level of serum
ning with phenotypic evaluation followed by genotyping of those
ferritin 1,000 ng/mL is an accurate predictor of the degree of
with elevated iron markers (Fig. 1). The proposed algorithm is
hepatic fibrosis (cirrhosis).32
constructed to detect iron overload caused by HH with a high
Recommendation 1. Initial screening of individuals with sus-
degree of accuracy, while providing a pathway for those cases of
pected iron overload and those over the age of 20 years who
iron overload unassociated with the HFE mutation.
are first-degree relatives of known cases of HH should be done
by measurement of transferrin saturation after an overnight
ALGORITHM STEP 1
fast. Simultaneous serum ferritin determination increases the
Indirect Serologic Markers of Iron Stores
predictive accuracy for diagnosis of iron overload. TS is also
The initial approach to diagnosis of HH is by indirect sero- the test of choice for screening the general adult population
logic markers of iron stores. Transferrin saturation (TS) is for iron overload states (Fig. 1) (rating: II A, B, C, D, and E).
1324 TAVILL HEPATOLOGY May 2001
ALGORITHM STEP 2
or C282Y heterozygotes with elevated TS, particularly those
who have had abnormal liver enzyme levels or clinical evi-
Genotypic Testing: Mutation Analysis
dence of liver disease (rating: II A, B, C, D, and E).
Fasting transferrin saturation less than 45% and a normal
serum ferritin would require no further evaluation. Elevation
ALGORITHM STEP 3
of TS and serum ferritin would require genotypic testing as
Liver Biopsy for HIC
indicated in step 2 of the diagnostic algorithm in Fig. 1. The
presence of the HFE mutations C282Y and H63D can now be
Liver biopsy is useful to document the presence of cirrhosis
detected by polymerase chain reaction using whole blood
(if not evident from radiologic studies) to rule out significant
samples.7 Individuals with serum indicators of iron overload
iron overload when iron markers are equivocal, or to investi-
who are homozygous for the C282Y mutation require phle-
gate other possible causes of liver disease. Histopathology and
botomy therapy. Those who are unlikely to have significant
staging of fibrosis is best determined with hematoxylin-eosin
hepatic injury may be offered therapeutic phlebotomy with-
and Masson trichrome staining, respectively. The liver is the
out the necessity for a liver biopsy. This includes individuals
most easily accessible tissue for accurately assessing iron
less than 40 years of age who have no clinical evidence of liver
stores. The degree and cellular distribution of iron stores is
disease (raised alanine transaminase, hepatomegaly, etc.) and
best assessed using a Perls Prussian blue stain. Before 1985,
whose serum ferritin is less than 1,000 ng/mL. Higher values
the extent of iron deposition was judged exclusively by this
of serum ferritin are associated with an increased likelihood of
method and, in fact, a qualitative assessment of iron stores was
significant hepatic fibrosis or cirrhosis.20,32 On the other
derived based on stainable iron.34 Two qualitative scales have
hand, liver biopsy should be offered to document the degree
been proposed.35,36 The most commonly used of these, the
of fibrosis in all homozygotes who are over the age of 40 years
Ludwig-Batts system, estimates the proportion of hepatocytes
or those who have an elevated serum alanine transaminase
that stain for iron, recognizing the progressive nature of iron
level, have clinical evidence of liver disease, or have a serum
accretion through the hepatic acinus from Rappaport zone 1
ferritin greater than 1,000 ng/mL. Since these are likely to be
(periportal) to zone 3 (pericentral).36 Although grade 4 iron
individuals over the age of 40 years, discretion is appropriate
deposition (panacinar) usually indicates HH range quantita-
in recommending liver biopsy on the basis of age alone. In the
tive iron levels, grades 2 and 3 correlate poorly with quanti-
absence of the above indicators of cirrhosis, other risk factors
tative iron content. For this reason the quantitative, biochem-
(e.g., alcohol abuse, or coexisting clinical features of HH, such
ical HIC has become the preferred method for evaluating the
as diabetes, impotence, etc.) may play a role in making this
hepatic iron stores. Quantitative iron determinations from
recommendation. Liver biopsy and hepatic iron evaluation
fresh frozen and formalin-fixed, paraffin-embedded samples
are also recommended in compound heterozygotes (C282Y/
are comparable.36,37 Accordingly, a biopsy core at least 2.5 to
H63D), C282Y heterozygotes, or non-HFE mutated individu-
3.0 cm in total length should be obtained. A 0.5- to 1.0-cm
als who have indirect markers of iron overload, particularly if
piece of the tissue core should be removed and placed in a dry
they also have abnormal liver enzymes or clinical evidence of
tube or in 10% formalin (not in saline, which may leach out
liver disease. Although these individuals account for a small
iron). The remainder of the fixed tissue is processed for rou-
proportion of phenotypic hemochromatosis, they have a low
tine histopathologic evaluation and a Perls Prussian blue
likelihood of significant iron overload, and elevated iron tests
stain. If tissue was not separated and saved before fixation and
are often due to other causes of liver disease.33
embedding, the remaining tissue can be removed from the
Although recognizing that the penetrance of the C282Y
paraffin block and sent for quantitative iron.
mutation is variable, the option is provided in step 1 of the
The normal HIC is less than 1,800 g/g dry weight (equiv-
diagnostic algorithm to proceed to gene mutation analysis
alent to 32 mol/g). It is now clear from several studies that
regardless of the TS or serum ferritin in first-degree relatives
most patients with homozygous HH steadily and inexorably
of a known HH individual. In the case of the children of an HH
accumulate iron at least through early adult and middle life,
patient, mutation analysis in the spouse allows for assessment
unless they have had blood loss or have been blood donors, in
of the genotypic status of the children.14 If the spouse pos-
contrast to patients with secondary iron overload caused by
sesses no C282Y mutation, the offspring can only be heterozy-
other chronic liver diseases. The concept of the HII as a mea-
gous. An HH patient with a spouse who is a heterozygote for
sure of the iron accretion rate was developed to distinguish
C282Y has a 50% chance of having homozygous offspring.
HH from these other potentially confounding clinical situa-
Because organ damage is virtually unknown in HH before
tions, particularly alcohol-induced liver disease.16-18 A rate in
adult life, evaluation of first-degree relatives can be postponed
excess of 1.9 mol/g/y is strong evidence for homozygous
until about 20 years of age.
hemochromatosis. However, it has recently been shown that
Recommendation 2. Genotyping to detect HFE mutations
up to 15% of genotypic homozygotes for HH do not meet the
should be performed for all individuals who have abnormal
previously defined rate of at least 1.9 mol/g/y. Thus, an ele-
iron studies and on those who are first-degree relatives of
vated HII is no longer considered essential for diagnosis.20
identified homozygotes as detailed in step 1 of the diagnostic
algorithm. In the absence of indicators suggestive of signifi- Yet, even these individuals with partial expression of homozy-
gous HH have HIC at least 3 times the upper limit of normal if
cant liver disease, C282Y homozygotes under the age of 40
they are more than 20 years old. Finally, it should be empha-
years may be treated by therapeutic phlebotomy without the
sized that secondary iron overload due to dyserythropoietic or
need for liver biopsy. Liver biopsy is recommended in all
homozygotes with clinical evidence of liver disease, serum hemolytic anemia may have HIC comparable with that seen in
ferritin greater than 1,000 ng/mL, and particularly in those HH, particularly in those who require repeated blood transfu-
greater than 40 years of age with other risk factors for liver sions. These causes should be easily distinguishable by other
disease. Liver biopsy should also be considered in compound clinical criteria.
HEPATOLOGY Vol. 33, No. 5, 2001 TAVILL 1325
TABLE 5. Treatment of Iron Overload
Although the rate of hepatic iron accumulation (HII) has
lost some of its importance in the diagnosis of HH, it is nev-
Treatment of Hemochromatosis
ertheless the correlation between HIC and age that determines
Hereditary hemochromatosis
the age at which fibrosis will develop. Sallie et al.18 found no
One phlebotomy (removal of 500 mL of blood) weekly or biweekly
patient who developed hepatic fibrosis before the HIC levels
Check hematocrit prior to each phlebotomy; allow hematocrit to fall by
exceeded 14,000 g/g dry weight. Hepatic fibrosis was not
no more than 20% of prior level
present in any patient less than 40 years of age in the series
Check serum ferritin level every 10-12 phlebotomies
reported by Bacon et al.20 and Guyader et al.32 and occurred
Stop frequent phlebotomy when serum ferritin falls below 50 ng/mL
only at a younger age or lower levels of HIC in individuals who Continue phlebotomy at intervals to keep serum ferritin to between 25
also abuse alcohol.11 The latter is the basis for recommenda- and 50 ng/mL
Avoid vitamin C supplements
tion 2 (Fig. 1) regarding the lack of need for liver biopsy in
Secondary iron overload due to dyserythropoiesis
some patients. Indeed, Bacon et al. retrospectively applied this
Deferoxamine (Desferal) at a dose of 20-40 mg/kg body weight per day
algorithm to 66 patients who were C282Y homozygotes and
Consider follow-up liver biopsy to ascertain adequacy of iron removal
found that 19 of the 66 would not have required the liver
Avoid vitamin C supplements
biopsy, which otherwise would have been necessary to deter-
mine HIC.20
The value of liver biopsy is not limited to determination of
HIC. Documentation of extensive bridging fibrosis or cirrho- and powerful argument for preventive therapy prior to the
development of cirrhosis.38
sis by liver biopsy has a profound impact on the prognosis in
The mainstay of treatment for HH remains phlebotomy
HH patients. Serum aminotransferase levels may be helpful in
(Table 5). One unit of blood (equal to about 250 mg of iron,
identifying chronic liver disease but lack negative predictive
depending on the hematocrit) should be removed once or
accuracy since half of cirrhotic HH patients have normal ala-
twice per week as tolerated. In HH patients who may have
nine transaminase or aspartate transaminase values.20 Sur-
total body iron stores greater than 30 g, this phlebotomy reg-
vival in noncirrhotic HH patients is similar to the normal
imen may take up to 2 to 3 years to adequately reduce iron
control population, while those with cirrhosis have signifi-
stores to the desired end point just short of iron deficiency.
cantly increased mortality. Cirrhosis or its complications, par-
Each venesection should be preceded by measurement of the
ticularly hepatocellular cancer, account for three quarters of
hematocrit. The hematocrit should have returned to within 10
HH-related deaths.15 Thus, close surveillance for HCC has
points of or no lower than 20% below its starting value. Trans-
been proposed for cirrhotic individuals, although there are
ferrin saturation usually remains elevated until iron stores are
currently no data to guide the optimal method or interval for
depleted. Serum ferritin may initially fluctuate, but eventually
such screening in HH. Further studies are needed.
begin to fall progressively with iron mobilization. Serum fer-
Recommendation 3. Liver biopsy is helpful in suspected HH
ritin should only be done after every 10 to 12 phlebotomies in
when documentation of HIC and the stage of fibrosis is nec-
the initial stages of treatment. It can be confidently assumed
essary (see recommendation 2) or to rule out other causes of
that excess iron stores have been mobilized when the serum
liver disease. In addition to routine histologic assessment,
ferritin falls below 50 ng/mL. As the target figure of 50 ng/mL
qualitative hepatic iron determination should be performed
is approached, it may be repeated more frequently to preempt
by Perls staining. If this suggests increased iron stores, this
the development of overt iron deficiency. Levels less than 25
should be confirmed by a quantitative iron measurement in
ng/mL indicate iron deficiency and require a temporary hold
stored tissue (rating: II A, B, C, D, and E).
on further phlebotomies. Iron deficiency anemia should be
avoided. At the point at which the above-mentioned criteria
TREATMENT OF HEMOCHROMATOSIS
indicate incipient iron deficiency, frequent phlebotomy can
Hereditary Hemochromatosis
be stopped and a maintenance schedule started. The fre-
There is overwhelming evidence that institution of phlebot- quency of maintenance phlebotomies varies among individu-
omy therapy before cirrhosis and/or diabetes develop will sig- als, as might be expected given the variable rate of iron accu-
nificantly reduce the morbidity and mortality of HH.15 There- mulation in HH. Certain persons (either male or female)
fore, early identification (step 1 in algorithm, Fig. 1) and require phlebotomy every month, whereas others who pre-
preemptive treatment of those at risk is required. This in- sumably reaccumulate iron at a slower rate may need only 3 to
cludes treatment of asymptomatic individuals with homozy- 4 units of blood removed per year. Currently, in the United
gous HH and markers of iron overload, as well as others with States, blood acquired by therapeutic phlebotomy cannot be
evidence of potentially toxic levels of hepatic iron. In symp- used for blood donation, a ruling that is under scrutiny.
tomatic patients treatment is also advocated to mitigate as Therefore, phlebotomy remains a therapeutic procedure with
much of the organ damage as possible. Certain clinical fea- a coding recognized by the Health Care Finance Administra-
tures may be ameliorated by phlebotomy (malaise, fatigue, tion and third-party insurers.
skin pigmentation, insulin requirements in diabetes, abdom- Cardiac dysrhythmias and cardiomyopathy are the most
inal pain), whereas other features are either less responsive to common causes of sudden death in iron overload states. Since
iron removal or do not respond at all (arthropathy, hypogo- the risk of these complications may increase during rapid
nadism, cirrhosis). The life-threatening complications of cir- mobilization of iron, certain additional precautions and ther-
rhosis, particularly HCC, continue to be a threat to survival apy may be required. Pharmacologic doses of vitamin C accel-
even after adequate phlebotomy. HCC accounts for about 30% erate mobilization of iron to a level that may saturate circulat-
of all deaths in HH, whereas other complications of cirrhosis ing transferrin, which results potentially in an increase in
account for an additional 20%.11,15 The observation that HCC pro-oxidant and free-radical activity. Therefore, supplemental
is exceedingly rare in noncirrhotic HH provides an additional vitamin C should be avoided by patients undergoing phlebot-
1326 TAVILL HEPATOLOGY May 2001
TABLE 6. Iron Overload States
omy. Patients receiving iron chelators should not exceed 200
mg of vitamin C intake daily.39
Classification
Cirrhosis does not reverse with iron removal and the devel-
Hereditary hemochromatosis
opment of decompensated liver disease is an indication to
HH: HFE related
consider orthotopic liver transplantation. However, survival
C282Y homozygosity
of transplanted HH patients is lower than in those trans-
C282Y/H63D compound heterozygosity
planted for other causes of liver disease.40,41 Most posttrans-
Other mutations of HFE
plantation deaths in HH patients occur in the perioperative
HH: non-HFE related; other gene mutations
period from cardiac or infection-related complications.
Juvenile hemochromatosis
Whether these problems are related to inadequate removal of Autosomal dominant hemochromatosis (Solomon Islands)
Secondary iron overload
excess iron stores before orthotopic liver transplantation is
Iron-loading anemias transfusion
not known. However, this is certainly the case in some pa-
Thalassemia major
tients in whom HH is not or cannot be diagnosed before or-
Sideroblastic anemia
thotopic liver transplantation. Persistent tissue iron toxicity
Chronic hemolytic anemias
may lead to posttransplantation end-organ problems in these
Dietary iron overload
cases despite removal of the bulk of parenchymal iron stores
Chronic liver diseases
with the explanted liver.
Hepatitis C and B
Early studies reported that HCC accounted for 30% of all
Alcohol-induced liver disease
deaths in HH patients and that the risk did not decrease after
Porphyria cutanea tarda
adequate iron removal by phlebotomy.41 However, these data Fatty liver disease
Miscellaneous causes of iron overload
were obtained before institution of screening programs that
African iron overload
promote earlier identification and treatment of cases. None-
Neonatal iron overload
theless, close life-long observation of HH patients with signif-
Aceruloplasminemia
icant fibrosis or cirrhosis would seem prudent. However,
Congenital atransferrinemia
there are currently no data on the optimal method or fre-
quency for such surveillance for the development of HCC, nor
are there data supporting the cost effectiveness of such a prac-
tice in HH. Until there is information specific to HH on this to reduce hepatic iron concentrations below 15,000 g/g dry
life-threatening complication, the recommendation for sur- weight significantly reduces the risk of clinical disease.43
veillance of HCC is based on analogy to other causes of cir- However, the aim of chelation therapy should be more ambi-
rhosis, particularly chronic viral hepatitis. tious and should attempt to achieve and maintain near normal
Recommendation 4. All patients with HH who have evidence hepatic iron concentrations. The application of deferoxamine
of iron overload should be strongly encouraged to undergo therapy is limited by cost (particularly in developing coun-
regular phlebotomies until iron stores are depleted. These tries), the need for a parenteral route of therapy, discomfort
should be continued for life, gauging the frequency of main- and inconvenience (a challenging prospect in young pa-
tenance therapy on the serum ferritin level. Vitamin C sup- tients), and neurotoxicity. In addition, a variety of opportu-
plements should be avoided. HH patients with cirrhosis nistic bacterial infections have been described after prolonged
should undergo regular screening for HCC (rating: II A, B, C, chelation therapy.44
D, and E). Monitoring iron reduction in patients with secondary iron
overload is a challenge. In contrast to HH, where serum fer-
Secondary Iron Overload
ritin reliably reflects iron burden during therapy, ferritin lev-
Although these guidelines have concentrated on the man- els are misleading in secondary overload cases. In many pa-
agement of HH it is pertinent to review the treatment of non- tients, it may be necessary to repeat the liver biopsy to assess
hereditary forms of secondary iron overload. The causes of the progress of therapy and ensure adequate chelation.45 Su-
secondary iron overload are listed in Table 6. perquantum magnetic susceptibility determinations are capa-
Phlebotomy is useful only in certain forms of secondary ble of measuring hepatic iron concentrations over a wide
iron overload (Table 5). It has been used in African iron over- range, but it is expensive and available only at two centers
load and porphyria cutanea tarda with reduction in morbidity worldwide. The recent advent of high Tesla magnetic reso-
and mortality. No systematic, randomized controlled studies nance imaging instruments showed some initial promise as a
have been done in secondary iron overload states associated noninvasive way to estimate hepatic iron levels, but it has
with chronic liver diseases to evaluate long-term outcomes of proven to be inaccurate when there is marked iron overload or
iron removal therapy. In secondary iron overload associated hepatic fibrosis/cirrhosis.46 In selected patients with thalasse-
with ineffective erythropoiesis, iron chelation therapy with mia major, allogeneic bone marrow transplantation offers the
parenteral deferoxamine is the treatment of choice. Multiple possibility of permanent cure of the hematologic disorder.47
studies have documented the efficacy of deferoxamine in pre- However, the preexisting iron overload persists after the
venting the complications of iron overload in -thalassemia.42 transplantation and such patients may be recommended for
Deferoxamine mesylate (Desferal; Novartis Pharmaceuticals phlebotomy with the expectation that the bone marrow is
Corporation, East Hanover, NJ) is the only approved iron capable of enhanced erythropoiesis. As in patients treated by
chelation agent that is widely available. Usually it is adminis- chelation, the liver iron concentration provides an accurate,
tered subcutaneously, using an implanted minipump, by con- quantitative means for monitoring iron balance.48
tinuous infusion over a 24-hour cycle at a dose of 20 to 40 Recommendation 5. Treatment of secondary iron overload
mg/kg/d. A total dose of about 2 g per 24 hours usually should be tailored to the underlying cause. Phlebotomy using
achieves maximum urinary iron excretion. Chelation therapy a regimen similar to HH (see recommendation 4) may be
HEPATOLOGY Vol. 33, No. 5, 2001 TAVILL 1327
6. Olynyk JK, Cullen DJ, Aquila S, Rossi E, Summerville L, Powell LW. A
tolerated in some forms of secondary iron overload without
population based study of the clinical expression of the hemochromato-
preexisting anemia. Parenteral chelation therapy with defer-
sis gene. N Engl J Med 1999;341:718-724.
oxamine is currently the treatment of choice in patients with
7. Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, Ruddy DA, Basava A,
chronic dyserythropoietic syndromes or chronic hemolytic
Dormishian F, et al. A novel MHC class 1-like gene is mutated in patients
anemia. Monitoring of the efficacy of therapy during chelation
with hereditary haemochromatosis. Nat Genet 1996;13:399-408.
may require repeat liver biopsies to confirm adequate reduc- 8. Carella M, D Ambrosio L, Totaro A, Grifa A, Valentino MA, Piperno A,
Girelli D, et al. Mutation analysis of the HLA-H gene in Italian hemochro-
tion of HIC (rating: II A, B, C, D, and E).
matosis patients. Am J Hum Genet 1997;60:828-832.
9. Cameschella C, Fargion S, Sampietro M, Roetto A, Bosio S, Garozzo G,
SUMMARY
Arosio C, et al. Inherited HFE-unrelated hemochromatosis in Italian fam-
ilies. HEPATOLOGY 1999;29:1563-1564.
HH is one of the few genetic disorders in which phenotypic
10. Pietrangelo A, Montosi G, Totaro A, Garuti C, Conte D, Cassanelli S,
manifestations (organ damage) are delayed to adult life. How-
Fraquelli M, et al. Hereditary hemochromatosis in adults without patho-
ever, sensitive and specific phenotypic and genotypic testing
genic mutations in the hemochromatosis gene. N Engl J Med 1999;341:
now allows diagnosis of HH while it is still a disorder of iron
725-732.
11. Adams PC, Deugnier Y, Moirand R, Brissot P. The relationship between
metabolism and before it results in end-organ damage.49 The
iron overload, clinical symptoms, and age in 410 patients with genetic
practical clinical guidelines provided here offer a cost-effec-
hemochromatosis. HEPATOLOGY 1997;25:162-166.
tive, preemptive diagnostic approach whereby early identifi-
12. Crawford DHG, Jazwinska EC, Cullen LM, Powell LW. Expression of
cation of individuals at risk and initiation of life-saving phle-
HLA-linked hemochromatosis in subjects homozygous for the C282Y
botomy therapy in the presymptomatic stage of the disorder
mutation. Gastroenterology 1998;114:1003-1008.
are facilitated. 13. Adams PC, Gregor JC, Kertesz AE, Valberg LS. Screening blood donors
for hereditary hemochromatosis. Decision analysis model based on a 30
year database. Gastroenterology 1995;109:177-188.
ADDENDUM
14. Adams PC, Chakrabarti S. Genotypic/phenotypic correlations in genetic
Since the development of these practice guidelines was ini- hemochromatosis: evolution of diagnostic criteria. Gastroenterology
1998;114:319-323.
tiated by the AASLD, an International Consensus Conference
15. Niederau C, Fischer R, Purschel A, Stremmel W, Haussinger D, Stroh-
on Haemochromatosis was conducted by the European Asso-
meyer G. Long-term survival in patients with hereditary hemochroma-
ciation for the Study of Liver (EASL), cosponsored by the
tosis. Gastroenterology 1996;110:1107-1119.
WHO, NIH, and CDC, and was recently published as a con-
16. Bassett ML, Halliday JW, Powell LW. Value of hepatic iron measurement
ference report.50 Although many of the conclusions and rec- in early hemochromatosis and determination of the critical iron level
associated with fibrosis. HEPATOLOGY 1986;6:24-29.
ommendations of this conference accord with the AASLD
17. Summers KM, Halliday JW, Powell LW. Identification of homozygous
Practice Guidelines on hemochromatosis, others remain ten-
hemochromatosis subjects by measurement of hepatic iron index. HEPA-
tative for the international group, particularly those relating
TOLOGY 1990;12:20-25.
to screening for iron overload. However, we share with the
18. Sallie RW, Reed WD, Shilkin KB. Confirmation of the efficacy of hepatic
international group an awareness of the need for information tissue iron index in differentiating genetic hemochromatosis from alco-
holic liver disease complicated by alcoholic haemosiderosis. Gut 1991;
derived from well-designed screening programs that incorpo-
32:207-210.
rate careful follow-up of identified HH patients and matched
19. Kowdley KV, Trainer TD, Saltzman JR, Pedrosa M, Krawitt EL, Knox TA,
controls.
Susskind K, et al. Utility of hepatic iron index in American patients with
hereditary hemochromatosis: a multicenter study. Gastroenterology
Acknowledgment: This guideline was produced in collab-
1997;113:1270-1277.
oration with the Practice Guideline Committee of the Ameri-
20. Bacon BR, Olynyk JK, Brunt EM, Britton RS, Wolff RK. HFE genotype in
can Association for the Study of Liver Diseases. This commit- patients with hemochromatosis and other liver diseases. Ann Int Med
1999;130:953-962.
tee in concert with additional external consultants, supplied
21. Balan V, Baldus W, Fairbanks V, Michels V, Burritt M, Klee G. Screening
extensive peer review of the document. Members of the Prac-
for hemochromatosis: a cost-effectiveness study based on 12,258 pa-
tice Guidelines Committee included Henry C. Bodenheimer,
tients. Gastroenterology 1994;107:453-459.
Jr. (Chair), Thomas D. Boyer (Governing Board Liaison),
22. Phatak PD, Guzman G, Woll JE, Robeson A, Phelps CE. Cost-effective-
David E. Bernstein, Gary L. Davis, Stuart C. Gordon, F. Blaine
ness of screening for hereditary hemochromatosis. Arch Int Med 1994;
154:769-776.
Hollinger, Donald M. Jensen, Jacob Korula, Jan M. Novak,
23. Baer DM, Simons JL, Staples RL, Rumore GJ, Morton CJ. Hemochroma-
Melissa Palmer, Eve A. Roberts, Thomas Shaw-Stiffel, and
tosis screening in asymptomatic ambulatory men 30 years of age and
James R. Spivey.
older. Am J Med 1995;98:464-468.
24. Bassett ML, Leggett BA, Halliday JW, Webb S, Powell LW. Analysis of the
REFERENCES
cost of population screening for hemochromatosis using biochemical
and genetic markers. J Hepatol 1997;27:517-524.
1. Eddy DM. A Manual for Assessing Health Practices and Designing Prac-
25. Adams PC, Valberg LS. Screening blood donors for hereditary hemochro-
tice Guidelines. Philadelphia: American College of Physicians 1996:1-
matosis: Decision analysis model comparing genotyping to phenotyping.
126.
Am J Gastroenterol 1999;94:1593-1600.
2. Position and policy statement. American Gastroenterological Association
26. Tavill AS. Screening for hemochromatosis: phenotyping or genotyping or
policy statement on the use of medical practice guidelines by managed
both? Am J Gastroenterol 1999;94:1430-1433.
care organizations and insurance carriers. Gastroenterology 1995;108:
27. Edwards CQ, Griffen LM, Goldgar D, Drummond C, Skolnick MH, Kush-
925-926.
ner JP. Prevalence of hemochromatosis among 11,065 presumably
3. Gross PA, Barrett TL, Dellinger EP, Krause PJ, Martone WJ, McGowan JE,
healthy blood donors. N Engl J Med 1988;318:1355-1362.
Sweet RL, et al. Infectious Diseases Society of America quality standards
28. Borwein S, Ghent CN, Valberg LS. Diagnostic efficacy of screening tests
for infectious diseases: purpose of quality standards for infectious dis-
for hereditary hemochromatosis. Cen Med Assoc 1984;131:895-901.
eases. Clin Infect Dis 1994;18:421.
29. Bassett ML, Halliday JW, Ferris RA, Powell LW. Diagnosis of hemochro-
4. Merryweather-Clarke AT, Pointon JJ, Shearman JD, Robson KJ. Global
matosis in young subjects: Predictive accuracy of biochemical screening
prevalence of putative haemochromatosis mutations. J Med Genet 1997;
34:275-278. tests. Gastroenterology 1984;87:628-633.
5. Lucotte G. Celtic origin of the C282Y mutation of hemochromatosis. 30. McLaren CE, McLachlan GJ, Halliday JW, Webb SI, Leggett BA, Jazwin-
Blood Cells Mol Dis 1998;24:433-438. ska EC, Crawford DH, et al. Distribution of transferrin saturation in an
1328 TAVILL HEPATOLOGY May 2001
Australian population: relevance to the early diagnosis of hemochroma- 40. Farrell FJ, Nguyen KM, Woodley S, Imperial JC, Garcia-Kennedy R, Man
tosis. Gastroenterology 1998;114:543-549. K, Esquivel CO, et al. Outcome of liver transplantation in patients with
31. Adams PC, Kertesz AE, McLaren CE, Barr R, Bamford A, Chakrabarti S. hemochromatosis. HEPATOLOGY 1994;20:404-410.
Population screening for hemochromatosis: a comparison of unbound 41. Kowdley KV, Hassanein T, Kaur S, Farrell FJ, Van Thiel DH, Keefe EB,
iron binding capacity transferrin saturation, and C282Y genotyping in Sorrell MF, et al. Primary liver cancer and survival in hereditary hemo-
5,211 voluntary blood donors. HEPATOLOGY 2000;31:1160-1164. chromatosis patients undergoing orthotopic liver transplantation. Liver
32. Guyader D, Jacquelinet C, Moirand R, Turlin B, Mendler MH, Chaperon Transplant Surg 1995;1:237-241.
J, David V, et al. Non-invasive prediction of fibrosis in C282Y homozy- 42. Oliveri NF. The -thalassemias. N Engl J Med 1999;341:99-109.
gous hemochromatosis. Gastroenterology 1998;115:929-936. 43. Brittenham GM, Griffith PM, Nienhuis AW, McLaren CE, Young NS,
33. Bacon BR, Powell LW, Adams PC, Kresina T, Hoofnagle JH. Molecular Tucker EE, Allen CJ, et al. Efficacy of deferoxamine in preventing com-
medicine and hemochromatosis: at the crossroads. Gastroenterology plications of iron overload in patients with thalassemia major. N Engl
1999;116:193-207. J Med 1994;331:567-573.
34. Deugnier YM, Turlin B, Powell LW, Summers KM, Moirand R, Fletcher 44. Porter JB. A risk-benefit assessment of iron chelation therapy. Drug Saf
L, Loreal O, et al. Differentiation between heterozygotes and homozy- 1997;17:407-421.
gotes in genetic hemochromatosis by means of a histological hepatic iron 45. Brittenham GM, Cohen AR, McLaren CE, Martin MB, Griffith PM, Nien-
index: a study of 192 cases. HEPATOLOGY 1993;17:30-34. huis AW, Young NS, et al. Hepatic iron stores and plasma ferritin con-
35. Scheuer PJ, Williams R, Muir AR. Hepatic pathology in relatives of pa- centration in patients with sickle cell anemia and thalassemia major. Am J
tients with hemochromatosis. J Pathol 1962;84:53-64. Hematol 1993;42:81-85.
36. Ludwig J, Batts K, Moyer T, Baldus W, Fairbanks V. Liver biopsy diag- 46. Angelucci E, Giovagnoni A, Valeri, Paci E, Ripalti M, Muretto P, McLaren
nosis of homozygous hemochromatosis: a diagnostic algorithm. Mayo C, et al. Limitations of magnetic resonance imaging in measurement of
Clin Proc 1993;68:263-267. hepatic iron. Blood 1997;90:4736-4742.
37. Olynyk JK, O Neill R, Britton RS, Bacon BR. Determination of hepatic 47. Angelucci E, Muretto P, Lucarelli G, Ripalti M, Barconciani D, Erer B,
iron concentration in fresh and paraffin-embedded tissue: diagnostic im- Galimberti M, et al. Phlebotomy to reduce iron overload in patients cured
plications. Gastroenterology 1994;106:674-677. of thalassemia by some marrow transplantation. Blood 1997;90:994-998.
38. Deugnier YM, Gruyader D, Crantock L, Lopez J-M, Turlin B, Yaouang J, 48. Angelucci E, Brittenham GM, McLaren CE, Ripalti M, Baronciani D,
Jouanolle H, et al. Primary liver cancer in genetic hemochromatosis: a Giardini C, Galimberti M, et al. Hepatic iron concentration and total
clinical pathological and pathogenic study of 54 cases. Gastroenterology body iron stores in thalassemia major. N Engl J Med 2000;343:327-331.
1993;104:228-234. 49. Tavill AS. Clinical implications of the hemochromatosis gene. N Engl
39. Cao A, Gabutti V, Galanello R, et al. Management Protocol for the Treat- J Med 1999;341:755-757.
ment of Thalassaemia Patients. Nicosia, Cyprus: Thalassaemia Interna- 50. EASL International Consensus Conference on Haemochromatosis.
tional Federation, 1997. J Hepatol 2000;33:485-504.


Wyszukiwarka