You are on page 1of 23

Current Clinical Pharmacology, 2007, 2, 155-177 155

Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid in Cholestatic


Liver Diseases
Davide Festi1,*, Marco Montagnani1, Francesco Azzaroli1, Francesca Lodato1, Giuseppe Mazzella1,
Aldo Roda2, Anna Rita Di Biase3, Enrico Roda1, Patrizia Simoni1 and Antonio Colecchia1

1
Departments of Internal Medicine and Gastroenterology and 2of Pharmaceutical Sciences, University of Bologna and
3
Department of Pediatrics, University of Modena, Italy

Abstract: Ursodeoxycholic acid (UDCA), previously used for cholesterol gallstone dissolution, is currently considered
the first choice therapy for many forms of cholestatic syndromes. Many mechanisms and sites of action have been pro-
posed for UDCA, but definitive data are still missing regarding the key points of its efficacy and optimal dosage in order
to achieve a sustained clinical effect. Among the suggested mechanisms of action of UDCA, changes in bile acid pool
composition, hepatocyte membrane protection, immunomodulatory effects and bicarbonate-rich hypercholeresis have
been extensively studied. However, recent evidence indicate that UDCA is a potent intracellular signalling agent that
counterbalances impaired biliary secretion, inhibits hepatocyte apoptosis and protects injured cholangiocytes against toxic
effects of bile acids. It is clear that the relative contribution of these mechanisms to the anticholestatic action of UDCA
depends on the type and stage of the liver injury. Available clinical evidence suggest that UDCA treatment has to be initi-
ated as early as possible and that higher doses could be more efficacious in inducing and maintaining clinical remission of
cholestatic diseases. The future availability of UDCA derivatives will possibly enhance the chances to effectively treat
chronic cholestatic diseases.

Key Words: Bile secretion, apoptosis, liver cirrhosis, nuclear receptors, intestinal absorption, transport systems, ursodeoxy-
cholic acid, cholestatic liver diseases.

INTRODUCTION enterohepatic circulation of bile acids and the main physiol-


ogic concepts of the bile acid transport system.
Ursodeoxycholic acid (UDCA; 3,7-dihydroxy-5-
cholanoic acid) is a dihydroxy bile acid, which is normally Bile Acid Transport Systems and the Modulatory Role of
present in human bile, although at low concentrations (about Ursodeoxycholic Acid
1-3% of total bile acids) [1]. It represents the major bile acid
Bile acids enable the intestinal absorption of lipids and
of the bile of black bears [2] and it has been used for centu-
fat-soluble vitamins [8]. Moreover, new findings on the in-
ries in traditional Chinese medicine as a remedy for liver
teraction between bile acids and nuclear receptors have re-
diseases. In modern medicine, UDCA has been employed as
newed interest in the role of bile acids as regulators of lipid
a litholytic agent in place of chenodeoxycholic acid since
1975, when Makino et al. [3] documented its ability to re- homeostasis [9]. Bile acids are secreted by hepatocytes into
the biliary system and successively reach the intestinal lu-
duce biliary cholesterol supersaturation, the main biochemi-
men, where they are absorbed and recirculated back to the
cal defect in cholesterol gallstone disease. Since that pre-
liver via the portal blood. These physiological events realize
liminary report, several clinical studies have identified UDCA
the entero-hepatic circulation of bile acids [10], which is
as the best therapeutic agent in the medical treatment of cho-
maintained mainly through active transport systems located
lesterol gallstones [4]. In 1985, Leuschner et al. [5] first re-
ported the potential use of UDCA as a hepatoprotective in the terminal ileum and in the liver [11]. Bile acids trans-
port kinetics in the liver [12] and in the intestine [13, 14]
agent in chronic active hepatitis and, subsequently, Poupon
have been characterized in vivo using animal models and in
et al. [6] suggested UDCA administration in chronic cho-
vitro using membrane vesicles.
lestatic liver diseases. Today, UDCA is used for the treat-
ment of different chronic cholestatic liver diseases, such as Bile acids are taken up via the basolateral sodium-
primary biliary cirrhosis (PBC), primary sclerosing cholangi- taurocholate cotransporter (NTCP) and organic anion trans-
tis (PSC), intrahepatic cholestasis of pregnancy (ICP) and port proteins (OATPs) at the basolateral membrane of the
pediatric cholestasis [7]. hepatocyte [12]. The hepatocyte intracellular transport of
bile acids has not yet been completely elucidated (Fig. 1).
The aim of the present article is to review the available
Following uptake across the basolateral membrane, bile ac-
evidence regarding the therapeutic role of UDCA in chronic
ids bind to the cytosolic protein 3-hydroxysteroid dehydro-
cholestatic liver diseases, after a short introduction on the
genase in rodents and to the hepatic bile acid binding protein
(HBAB) in the human liver. Vesicular trancytosis is possibly
*Address correspondence to this author at the Department of Internal Medi- involved in the transport of bile acids across hepatocytes, as
cine and Gastroenterology, Policlinico S. Orsola-Malpighi, Via Massarenti is suggested by its strong association with organelles, but
9, 40138 Bologna, Italy; Tel/Fax: +39-051-6364123; this mechanism is probably restricted to UDCA and litho-
E-mail: davide.festi@unibo.it
cholic acid [15]. Canalicular secretion is the rate limiting

1574-8847/07 $50.00+.00 ©2007 Bentham Science Publishers Ltd.


156 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

Fig. (1). Hepatobiliary transport systems.


The figure represents the major hepatobiliary bile acids (BA) transport systems. Na+/taurocholate cotransporter (NTCP) and organic anion
transporting proteins (OATPs) represent the basis for import into hepatocytes of BA which are then secreted into bile by the bile salt export
pump (BSEP). The Multidrug Resistance Protein 2 (MRP2) transports bilirubin, reduced glutathione (GSH), sulphated BA and various or-
ganic anions (OA) into bile while phospholipids (Pl) are secreted by multidrug resistance 3 protein (MDR3). The multidrug resistance-related
proteins 3 and 4 (MRP 3 & 4) are basolateral efflux transporters for BA and OA. Familial Intrahepatic cholestasis 1 protein (FIC1) is an
aminophospholipid (amino-Pl) flippase maintaining the canalicular membrane physiological inner/outer asymmetry. In cholangiocytes, the
apical sodium-bile acid transporter (ASBT) is located at the apical membrane, where it allows bile acid active uptake. In the basolateral
membrane of cholangiocytes, Ost / and MRP3 enable bile acid extrusion into the pericanalicular plexus, from which bile acids recirculate
back to hepatocytes (chole-hepatic shunt). In the apical membrane of cholangiocytes, the cystic fibrosis transmembrane conductance regula-
tor (CFTR) is possibly responsible for chloride secretion and maintainance of fluid balance across epithelium.

step in the blood-to-bile transport of bile acids. A specific patic cholestasis and in benign recurrent intrahepatic cho-
canalicular membrane bile salt export pump (BSEP) has lestasis [17]. FIC1 is an aminophospholipid flippase with
been cloned and characterized [16-18]. BSEP is an ABC- regulatory function in the secretory processes, derived from
type transmembrane protein, responsible for ATP-dependent its role in maintaining the canalicular membrane asymmetry
transport of monovalent bile acids into bile, which in turn between the inner and the outer layer [24, 25].
drives the bile acid-dependent bile flow. In the mouse model,
The terminal ileum is the intestinal site where the apical
UDCA can both prevent and reduce the Bsep down-
sodium-bile acid cotransporter (ASBT) [13, 14] is located,
regulation caused by DCA, explaining one of the mecha-
accounting for the active absorption of glycine and taurine-
nisms responsible for UDCA’s protective role in the treat-
conjugated bile acids [14]. The correct functioning of ASBT
ment of cholestatic liver disease [19]. The multidrug resis-
has been demonstrated to be necessary for adequate bile acid
tence protein 2 (MRP2) which works as bilirubin conjugate
export pump, is also a divalent bile acid transporter [20] reabsorption [26, 27], allowing more than 90% of these
molecules to be recirculated back to the liver via the portal
which allows the biliary excretion of bile acids with two
blood [14]. In addition to the ileal active transport, ionic and
negative charges, such as sulfated tauro- or glycolithocholic
non-ionic passive diffusion of bile acids occurs across the
acid. MRP2 also mediates the canalicular excretion of re-
small intestinal and colonic epithelia [14], accounting for the
duced glutathione (GSH) [21] and organic anions, such as
absorption of unconjugated and, to a lesser extent, glycine-
divalent amphipathic conjugates with glutathione, glucuro-
nate, and sulfate [22, 23]. Another transporter involved in conjugated dihydroxy bile acids [28, 29]. Furthermore, in
vitro experiments using brush border membrane vesicles
bile acid secretion is familial intrahepatic cholestasis 1 (FIC1),
indicate the presence of a facilitated transport system for
a P-type ATPase mutated, in progressive familial intrahe-
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 157

unconjugated bile acids in the jejunum [13, 30]. Studies us- MECHANISMS OF ACTION OF URSODEOXY-
ing intestinal brush border membrane vesicles showed that CHOLIC ACID IN CHOLESTASIS
UDCA is capable of reducing both the putative jejunal facili-
Since the first report of UDCA efficacy in liver diseases
tated and the ileal active bile acid uptake [13].
[5], different mechanisms of action have been suggested [49]
Additional transport systems are involved in bile acid to explain its therapeutic effects: changes in bile acid pool
transport through the enterocyte [28]: the cytosolic ileal bile composition, hepatocyte membrane protective effects, im-
acid binding protein (I-BABP) [31, 32] and an heteromeric munomodulatory effects and bicarbonate-rich hypercholere-
organic solute transporter (OST-OST) which has been sis. Furthermore, recent evidence indicate that UDCA is a
recently characterized as the ileal basolateral bile acid and potent intracellular signalling agent which induces stimula-
sterol carrier [33, 34]. tion of impaired biliary secretion, inhibits hepatocyte apop-
tosis and may protect injured cholangiocytes against the
In addition to hepatocytes and ileocytes, other cells pro- toxic effects of bile acids [50] (Fig. 2). Although it is not
vided with specialized bile acid transport systems are located clear which of these mechanisms has a primary role in the
along the biliary tree and in the kidney. Large and medium therapeutic action in cholestasis, it is likely that the effects
sized rat cholangiocytes express both Asbt (identical to the mainly depend on the specific cholestatic liver disease and
ileal transporter) and a truncated form of the same protein (t- the stage of the disease itself [51].
Asbt) [35, 36]. ASBT is located at the apical membrane of
cholangiocytes where it accounts for the active sodium cou- In fact, cholestatic diseases can differ with respect to the
pled bile acid uptake. Furthermore, nonionic diffusion may primary site of bile secretory impairment: in patients with
also contribute to bile acid apical uptake in cholangiocytes PBC, PSC and liver disease associated with cystic fibrosis
[8]. A sodium-independent transport system mediates the biliary secretion from both hepatocytes and cholangiocytes
basolateral efflux of bile acids from cholangiocytes into the may be impaired, while in patients with ICP and drug-
peribiliary plexus [37] and MRP3 has been identified as the induced and sepsis-associated cholestasis, the predominant
basolateral efflux pump [38-41]. In rodents, t-Asbt [36] and defect is related to bile secretion from the hepatocytes [52].
Oatp3 [12] have been proposed as basolateral bile acid efflux Furthermore, the stage of the disease may influence UDCA
pumps. More recently, Ost /, which are selectively local- therapeutic action. In the early stage of PBC, when the dam-
ized in the basolateral plasma membrane of the epithelial age is mainly related to a toxic effect of bile acids, the pri-
cells with bile acid and sterol absorption capacity, have been mary therapeutic mechanism is towards the protection of
identified as an important basolateral transport system in injured cholangiocytes and subsequently the excretory func-
cholangiocytes [33]. The cholangiocyte transport systems are tion. In the later stage of the disease, stimulation of biliary
supposed to realize a chole-hepatic shunt [42] which should secretion is crucial for preventing retention of hydrophobic
allow a constant bile acid recycling to the hepatocytes, and bile acids and other toxic substances in the hepatocytes.
could explain the hypercholeretic effect of certain bile acids When bile acids are retained within the hepatocyte, the inhi-
[42, 43]. This system is also supposed to be important in the bition of bile acid-induced hepatocyte apoptosis represents
adaptation to chronic extrahepatic cholestasis [44, 45]. More- another important protective mechanism [51] (Fig. 2).
over, bile acids can activate intracellular signalling pathways Effect of Ursodeoxycholic Acid on Transport Proteins
and modulate cholangiocyte secretion, proliferation and sur- and Metabolic Pathways
vival [35]. Since bile acid-dependent biliary flow is the main
physiologic choleretic mechanism, the chole-hepatic shunt In the course of cholestasis, bile acids, bilirubin and other
could help biliary secretion during the inter-prandial period, cholephils accumulate in the liver and blood, while their
when the intestinal uptake cannot provide a sufficient bile concentration in the intestinal lumen is reduced [7]. In turn,
acid supply through the portal circulation. Similarly to cho- adaptive protective mechanisms determine profound changes
langiocytes, the gallbladder epithelial cells also express in the transporter and enzyme expression [7, 24, 41, 53].
ASBT, MRP2 and OATP-A, which are located in the apical Adaptive changes also occur during UDCA administration in
membrane and mediate sodium-dependent and –independent rodents [54, 55], but an additional role of UDCA on these
uptake [35]. MRP3 has been identified as a basolateral bile adaptive mechanisms in the course of cholestasis has not yet
acid transporter in gallbladder epithelial cells [39, 40]. been proven. In the hepatocyte, UDCA induces transcrip-
tional and post-transcriptional changes, leading to enhanced
In addition to the hepatic, intestinal and cholangiocyte expression and function of canalicular and basolateral trans-
transport, bile acid uptake systems are located in the apical porters. In mice, the expression of genes for the canalicular
and basolateral membranes of the proximal renal tubular transporters Bsep (monovalent bile acids transporter) and
cells. ASBT is expressed in the proximal renal tubular cells, Mrp2 (divalent bile acids and anionic conjugate transporter)
where it is located in the apical membrane and is responsible are enhanced by UDCA administration [19, 54]. The expres-
for the uptake of bile acids from glomerular filtrate, mini- sion of genes for basolateral bile acid efflux pumps, Mrp3
mizing the urinary loss under normal conditions [34, 46]. and Mrp4, is also enhanced by UDCA in mice [55]. Post-
MRP2 is also located in the apical membrane, where it me- transcriptional mechanisms, such as vesicular exocytosis and
diates the excretion of organic anions and sulphated/glucu- targeting of the transporters to the canalicular membrane, are
ronidated bile acids under normal conditions and during cho- induced by UDCA in rodents, through mechanisms involv-
lestasis, respectively [47, 48]. Basolateral output from the ing calcium-dependent PKC- and MAP-kinases [51, 56,
proximal renal tubular cells is provided by Mrp3 and Ost/ 57]. In patients with PBC, UDCA improves the excretory
[33]. function [58] and the expression of MRP2 has been shown to
158 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

Fig. (2). Cholestatic liver diseases: pathogenetic mechanisms and mechanisms of action of ursodeoxycholic acid. BSEP function can be in-
hibited by estrogens. BSEP function and targeting to the canalicular membrane can be inhibited by cyclosporin (CyA).
UDCA= ursodeoxycholic acid; BSEP= Bile Salt Export Pump; PFIC= progressive familial intrahepatic cholestasis; PBC= primary biliary
cirrhosis; PSC= primary sclerosing cholangitis.

be increased during treatment with UDCA [59]. In PBC pa- Among these nuclear receptors, FXR is considered the
tients, UDCA treatment also upregulates the chloride-bicar- primary intracellular bile acid sensor [67]. In the hepatocyte,
bonate anion exchanger AE2, which is present in the apical bile acids activate FXR which in turn, after induction of SHP
membrane of both hepatocytes and cholangiocytes, improv- gene transcription, represses bile acid synthesis. Moreover,
ing the hepatobiliary bicarbonate secretion [60-62]. FXR induces different target genes encoding bile acids and
phospholipid canalicular transport proteins, while repressing
In primary human hepatocytes, UDCA stimulates CYP3A4 the hepatocyte basolateral sodium-bile acid cotransporter
expression [63, 64], which may increase hydroxylation and a (NTCP). In the ileal enterocyte, FXR represses ASBT ex-
more efficient renal excretion of bile acids. On the other pression and induces the transcription of I-BABP, OST/,
hand, reduced expression of CYP7A1 has been shown after and the fibroblast growth factor 19 (FGF 19), the enterocyte
UDCA treatment in primary human hepatocytes [65, 66], derived factor that mediates repression of the hepatic bile
and this could reduce the synthesis and accumulation of acid synthesis.
more hydrophobic, potentially toxic bile acids.
Recently, it has been demonstrated that UDCA is able to
The effect of UDCA on the expression of bile acid trans- activate the glucocorticoid receptor [68] which, in turn,
porters also operates in different cell types. For example, bile transactivates the ASBT gene (SLC10A2) [69]. In the liver,
acid efflux pumps are upregulated by UDCA both in the the NTCP gene (SLC10A1) is also activated by the gluco-
proximal renal tubule (Mrp2 and Mrp4) and in the enterocyte corticoid receptor and by the PPAR- coactivator-1, and
(Mrp2 and Mrp3) [55], protecting them from the intracellular suppressed by bile acids via a SHP- dependent mechanism
and systemic accumulation of more hydrophobic and toxic [70].
bile acids. Similarly, the apical and basolateral efflux pumps
(Mrp2 and Mrp3, respectively) are upregulated by UDCA in Protection of Cholangiocytes Against Toxic Effects of
the murine enterocytes [55]. Hydrophobic Bile Acids
The biliary tree is the anatomic compartment in which
Interaction of Ursodeoxycholic Acid with Nuclear Recep- bile acids reach the highest concentration (millimolar range).
tors Therefore, the protective effect of UDCA from the detergent
The growing knowledge about nuclear receptor activity damage caused by the more hydrophobic bile acids is maxi-
in bile acid homeostasis has opened a new field of interest mal for cholangiocyte and hepatocyte apical membranes. In
for both physiology and pharmacology research. The nuclear chronic cholestatic liver diseases, UDCA accumulates in bile
receptors involved in bile acid metabolism and transport, during the treatment, enhancing the hydrophilic/hydrophobic
with a possible pivotal involvement in the adaptive responses ratio in bile acid pool composition [7, 71]. This change in the
to cholestasis, are: farnesoid X receptor (FXR), short het- hydrophilic/hydrophobic balance of bile acid pool is consid-
erodimer partner (SHP), pregnane X receptor (PXR), vitamin ered an important factor for the protection of the biliary epi-
D receptor (VDR), constitutive androstane receptor (CAR), thelium from the detergent effect of more hydrophobic bile
and peroxisome proliferator-activated receptor-alpha (PPAR- acids [7, 71]. Moreover, UDCA treatment increases the bil-
) [11]. iary secretion of phospholipids [7, 71], which in turn con-
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 159

tribute to the formation of mixed micelles, protecting the ids and phospholipids [1]. Unconjugated UDCA is absorbed
cholangiocytes from the detergent effect of bile acids. In by passive diffusion throughout the intestinal tract and the
vitro studies demonstrate that UDCA protects cholesterol rate and amount absorbed is related to the effective solubili-
rich membranes from the detergent damage induced by the zation of the drug in the intestinal content, which is en-
more hydrophobic bile acids, and this mechanism seems to hanced when it is administered with a meal (high pH) and
be dependent primarily on the altered structure and composi- may be decreased in patients with cholestasis owing to a
tion of the simple and mixed micelles [72, 73]. reduced biliary and pancreatic secretion, which result in a
relatively acidic pH [94]. After intestinal absorption, UDCA
Ursodeoxycholic Acid and Apoptosis is taken up from the portal blood by the hepatocytes in their
Among the various properties assigned to UDCA, the sinusoidal domain via specific bile acid transporters. The
inhibition of apoptosis is one of the most recent. This sug- first-pass clearance is up to 60-70% and a relatively low
gestion comes from the observation that UDCA was protec- amount spill in the systemic circulation. In hepatocytes,
tive against cholestasis and the toxicity induced by hydro- UDCA is conjugated mainly with the aminoacid glycine and,
phobic bile acids, such as deoxycholic acid (DCA) and che- to a lesser extent, with taurine and it is secreted in bile only
nodeoxycholic acid (CDCA) [73-75]. as conjugated by BSEP, the canalicular bile acid transport
protein [1, 12]. Unconjugated UDCA is also secreted by the
Hydrophobic bile acids are able to induce apoptosis [76] hepatocytes in bile, where it is actively reabsorbed by cho-
by a ligand independent activation of the Fas death receptor langiocytes. The chole-hepatic shunt of UDCA is linked to
[77, 78]. Fas activation then triggers intracellular signal increased biliary bicarbonate secretion [42].
transduction pathways, among which is the activation of
caspase 8 and Bid [77] leading to Bax translocation to the In the intestinal lumen, glycoconjugated - UDCA is reab-
mitochondria, with disruption of the mitochondrial mem- sorbed by both active and passive transport mechanisms,
brane permeability [79]. The subsequent mitochondrial swel- while tauroconjugated -UDCA is absorbed in the terminal
ling determines cytochrome-C release, followed by the acti- ileum by active transport mechanism [1, 12]. Like other en-
vation of apoptotic protease activating factor 1 (APAF-1) dogenous bile acids, UDCA conjugates undergo an efficient
and the downstream caspases (9, 3, 6 and 7), finally leading enterohepatic circulation [1]. The unabsorbed fraction of
to apoptotic cell death [80]. UDCA and of UDCA conjugates enter the colon, where,
after bacterial deamidation and dehydroxilation of conju-
In vitro studies have shown that UDCA is capable of gates, they are mostly converted into lithocholic acid (LCA)
protecting hepatocyte from apoptosis induced by a variety of by intestinal bacteria and eliminated in feces [1]. Only small
different stimuli, including hydrophobic bile acids [81, 82]. amounts of LCA are reabsorbed by the colonic mucosa, sul-
UDCA diminishes Fas-ligand induced apoptosis preventing phated in the liver and secreted into bile [1].
cytochrome-C release from the mitochondria, probably by
modulating the mitochondrial membrane permeability transi- During chronic administration, UDCA becomes the pre-
tion and the production of reactive oxygen species [81-86]. dominant bile acid and this enrichment in the bile acid pool
Another line of evidence suggests that UDCA in its taurine seems to be dependent on the dose administered [95, 96].
conjugated form protects against hydrophobic bile salt in-
Bioavailability of Ursodeoxycholic Acid
duced apoptosis through the activation of survival pathways
(PI3K, p38, ERK MAPK) even though this latter possibility In the last 20 years, UDCA has been widely used for cho-
is still uncertain [87, 88]. Together with the in vitro experi- lesterol gallstone dissolution [94] and more recently for cho-
ments are the in vivo observations that UDCA administration lestatic liver disease treatment [4, 97]. However, clinical
prevents the increase of apoptotic hepatocytes in rats fed studies have demonstrated that, at a daily dose of 10-15
DCA [82] and reduces DNA fragmentation and bcl-2 expres- mg/kg, UDCA could have a limited therapeutic efficacy due
sion in biliary epithelial cells of patients with PBC [89]. to its poor intestinal absorption and to a relatively high pH
However, the real relevance of apoptosis in cholestasis is not (up to 8.4) necessary to go in solution as a ionized salt [96].
well defined [90, 91]. Experimental studies conducted on humans have demon-
strated that the orally administered drug is poorly absorbed,
In virtue of its anti-apoptotic properties, UDCA has also with almost 50% being eliminated in the stools [98]. Intesti-
been proposed as an additional treatment for viral hepatitis
nal absorption is further impaired in patients affected by cho-
[92]. This concept might be supported by an improved re-
lestatic liver disease, due to the relatively low pH of their
sponse rate to interferon-alfa in non responder patients with
intestinal content caused by a decreased biliary secretion [94,
hepatitis C virus (HCV)-related chronic hepatitis [93] even
99]. This defect could be overcome by the use of higher
though in vitro studies using co-cultures of Fas-Ligand ex-
doses of UDCA. Studies have shown that in PBC patients
pressing fibroblasts and primary mouse hepatocytes have high doses (up to 30 mg/kg day) produce higher biliary en-
shown a modest efficacy of UDCA in protecting from Fas
richment in UDCA [95, 100, 101]. In turn, these data suggest
induced apoptosis [86].
that the increase of the conventionally used therapeutic doses
PHARMACOKINETICS AND PHARMACODYNAM- may be of benefit in treating cholestatic liver diseases. Alter-
ICS OF URSODEOXYCHOLIC ACID natively, drug formulations could be optimized to increase
UDCA intestinal absorption.
UDCA is administered orally in its protonated unconju-
gated form and is solubilized at a pH greater than 8 in the It has been recently shown that UDCA’s effectiveness in
proximal jejunum in mixed micelles of endogenous bile ac- the treatment of cystic fibrosis-related cholestatic liver dis-
160 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

ease, PBC and PSC increases at a raised dose (i.e. 30-50 following the fate of the solids. Therefore, since UDCA was
mg/kg) [62, 100-102]. At this dose, biliary UDCA accumula- released when the pH was high because of a sustained biliary
tion reaches 55-70% of the total bile acid pool, which is sig- secretion, its solubilization was greatly facilitated, thus im-
nificantly higher than the amount (30-40%) reached after proving bioavailability.
administration of the conventional daily dose of 10-15 mg/kg
[95]. These data underline the importance of UDCA pharma- Various formulations have also been developed which
ceutical preparations and indicate that, since its intestinal modulate the kinetics of UDCA release [114]. The first de-
absorption is incomplete, either the conventionally used layed release preparation was developed and reported in
therapeutic dose needs to be increased or the drug formula- 1983 by Sama et al. [115]: a gelatin capsule containing three
tion needs to be further optimized. small tablets, each with a different Eudragit coating which
provided therapeutic UDCA levels over a long period to-
Few accurate studies are available regarding UDCA gether with good bioavailability. Nevertheless, few data are
pharmacokinetics and bioavailability in humans. In the first available concerning the rationale of the drug design and the
(and still the most comprehensive) study, Parquet et al. [103] increased efficacy of such formulations [116]. In order to
demonstrated that, when administered to fasting subjects, allow selection of the most efficient formulation, especially
UDCA is poorly absorbed because of its low solubility in in cost-benefit terms, it is important to provide data about its
gastro-intestinal contents (21-50% of the ingested doses relative intestinal absorption, release kinetics, and serum
were recovered in the solid form). In fact, the profile of levels. Therefore, a comparative study of pharmacokinetics
plasma concentrations paralleled the amount of soluble and bioavailability of the most representative formulations is
UDCA present in the intestinal lumen; as a consequence, the important. Recently a comparative study has been reported
bioavailability of the drug varies with its progressive solubi- [117]. The results obtained indicate that the pharmacokinet-
lization along the gastrointestinal tract. To overcome these ics of commercially available oral UDCA formulations can
problems, the authors [103] proposed multiple daily admini- be highly variable. The effect of the intestinal pH is a limit-
strations to improve absorption. ing factor regarding UDCA solubility, since the molecule
UDCA’s solubility in water (8 mol/L) is much lower requires a relatively alkaline pH for the formation of the
than that of other physiological dihydroxy bile acids, such as soluble salt. An elevated intestinal pH is driven by the extent
CDCA or DCA. UDCA has a relatively high critical micellar of biliary and pancreatic secretions, which are influenced by
concentration when compared with other dihydroxy bile ac- the meal hormones and could be impaired in cholestatic liver
ids such as CDCA and according to its pKa of 5, which is diseases. In addition gastric emptying, gallbladder contrac-
similar to that of endogenous bile acids, it produces a rela- tion, biliary secretion, and the overall motility of the gastro-
tively high critical micellizzation pH (CMpH) of 8.4, which intestinal tract associated with food intake play a major role
is the pH required to the drug to be dissolved in a micellar in determining the serum concentration-time curve.
solution. The CMpH of the endogenous dihydroxy bile acid In conclusion, the new enteric coated formulations above
is much lower, i.e. 7. UDCA therefore requires an alkaline discussed generally enhance the intestinal absorption of the
pH to form a micellar solution [104-106], and this accounts drug from 50% to 90-95% of the administered dose.
for its slow dissolution rate and the loss of the molecule in
the solid form. Metabolism
A recently comprehensive serum level study involving When administered orally, the absorbed UDCA is effi-
four UDCA formulations indicated the importance of formu- ciently metabolized in the liver. Like other endogenous bile
lation optimization for efficient intestinal absorption and acids UDCA is mainly conjugated in C-24 position with gly-
high bioavailability [107]. cine, (glycoursodeoxycholic acid, GUDCA) and to a less
extent with taurine (tauroursodeoxycholic acid, TUDCA).
To facilitate UDCA solubilization, Roda et al. [106] de-
Only in this form UDCA is secreted and accumulated in bile.
veloped an enteric coated formulation of UDCA sodium salt
The only forms of UDCA in a steady-state condition are
which released the active ingredient only at a pH higher than
5.5; gastroprotection prevented protonation by the gastric these forms, with a glycine /taurine ratio of 5-10 /1.
juice and allowed UDCA sodium salt release only in the The unabsorbed fraction of UDCA and of UDCA conju-
duodenum, where it quickly dissolved, leading to enhanced gates enter the colon, where, after bacterial deamidation of
absorption with respect to conventional formulations [108], the conjugated form, are mostly dehydroxylated and con-
from 50% to almost 90% of the administered dose. Other verted into LCA, and to a less extent, in a 7-oxo derivative
improved formulations were based on the formation of inclu- by intestinal bacteria and excreted in faeces [1]. Only small
sion complexes of UDCA with cyclodextrins or polyethylene amounts of LCA are reabsorbed by the colonic mucosa, sul-
glycol (PEG), which enhanced the solubility of the proto- phated in the liver and secreted into bile [1].
nated form [109-111]. In addition, the effect of different ex-
cipients on UDCA wet ability and also the effect of solid- During chronic administration, UDCA becomes the pre-
state structure (i.e. crystalline, amorph, micronised) were dominant bile acid and it is present mainly as GUDCA and
investigated in vitro [112]. A duodenal release formulation this enrichment in the bile acid pool seems to be dependent
[113] has been developed by modifying the gastric delivery on the dose administered: usually from 30-50% of total bile
time of a gastro-resistant tablet sinking in the stomach. Be- acids at a dose of 10 mg/Kg/day to 50-70 % at higher doses
cause of its sinking capacity, the tablet was released in the of 20-30 mg/Kg/day [95].
duodenum only at the end of the gastric phase of digestion,
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 161

URSODEOXYCHOLIC ACID IN CHRONIC CHO- the basis of the autoimmune nature of the disease, a number
LESTATIC LIVER DISEASES of different combinations with immune regulatory drugs
have been used. Combination with classical steroids (predni-
Primary Biliary Cirrhosis
sone and prednisolone) seemed to add something to mono-
UDCA is the first choice treatment for PBC at a dose of therapy with UDCA with regard to improvement in liver
13-15 mg/Kg/day [118]. The first report of UDCA efficacy function tests and liver histology [146, 147]. However, side
dates back to 1987 when Poupon et al. [6] reported a signifi- effects related to steroid treatment [148, 149] have limited its
cant improvement in biochemical parameters [gamma- long term use among clinicians. More appealing is the asso-
glutamyltranspeptidase (GT), alkaline phosphatases (ALP), ciation of UDCA with budesonide, a glucocorticoid with a
and alanine aminotransferase (ALT) and bilirubin] in PBC very high first pass metabolism [150], which has been shown
patients treated with the drug over a period of two years (Ta- to possess fewer side effects than conventional glucocorti-
ble 1). These results were later confirmed by a double blind coids in patients with inflammatory bowel disease [151]. In
controlled trial [119] which opened the way to many trials fact, budesonide has been shown to improve liver histology
investigating the efficacy of UDCA in PBC. In fact, a series and biochemical parameters of liver function at the cost of
of trials followed which confirmed the efficacy of UDCA in lower steroid related side effects [152, 153]. More recently, a
improving biochemical cholestasis in PBC patients [58, 120- three year randomized trial with 6 mg/day of bude-sonide has
130]. Furthermore, studies evaluating the effect of UDCA on confirmed previous results of efficacy in terms of biochemis-
histology have reported that UDCA administration delays try and liver histology with a mild systemic glucocorticoid
the histologic progression of the disease [125, 126, 131-133]. effect evident after two years [154]. Similarly to UDCA,
The combined analysis of three large trials have also sug- budesonide has also been shown to be effective in the early
gested that UDCA treatment of up to four years prolongs stages of PBC, while an unfavourable pharmacokinetic pro-
transplantation free survival [134]. However, meta-analyses, file and the development of significant side effects discour-
each of which involved more than a thousand patients, found age its use in stage IV PBC patients [155].
no difference between UDCA and the placebo with respect
Bezafibrate may represent another option for combina-
to transplantation free survival or death [135, 136]. These
tion therapy. This association was proposed after the obser-
meta-analyses have been criticized by experts in the field
vation that ALP, gamma-GT and IgM were decreased by its
because they involved many studies of short duration and administration in patients with hypercholesterolemia [156].
used low doses of UDCA [137]. A wide range of UDCA
The mechanisms at the basis of its efficacy in PBC patients
dosages have been used in PBC and few studies have inves-
are not well known. Laboratory data for a series of Japanese
tigated doses higher than those recommended [95, 100, 118,
studies with a limited number of patients [156-161], includ-
138-140]. Despite some differences among the studies,
ing a small prospective randomized crossover trial [158],
higher doses of UDCA, up to 30 mg/Kg/day, have proven to
have shown that the combination of 600 mg/day of UDCA
be well tolerated and to induce greater improvements in bio- with bezafibrate is more effective than low dose UDCA
chemical data of the patients. However, the short duration of
monotherapy. However, whether the combination has any
those studies does not allow any evaluation on long term
added benefit over standard dose UDCA in PBC is still un-
benefits from higher doses in incomplete responders PBC
known.
patients, particularly those without an advanced disease. The
recommended dosage will remain 13-15 mg/Kg/day for all Colchicine was proposed in the past as an adjunct to
patients until longer randomized controlled trials show a UDCA in view of its immunomodulatory and anti-fibrotic
significant benefit from higher doses of UDCA in partial properties [162, 163]. However, it does not seem to be supe-
responders. rior to a placebo according to a recent Cochrane systematic
review of randomized clinical trials [164], and the longest
Another important aspect influencing the efficacy of
double blind placebo-controlled trial of the combination
UDCA is the stage of the disease. Patients in stage III-IV of
therapy in PBC patients has failed to find any added benefit
the disease are likely to have limited or no benefit from
over UDCA monotherapy [165].
UDCA administration. A long term prospective randomized
trial showed no demonstrable effect on the long term out- Methotrexate added to UDCA, as well, has no effect on
come or survival of PBC patients treated with UDCA stan- the course of PBC, as shown by the more recent randomized
dard dose [141]. This result was certainly due in part to 65% controlled trials [166, 167].
of the stage III-IV patients. In fact, recently, three long term
A randomized, placebo controlled trial has reported the effi-
studies have shown that UDCA administration improves
cacy on both biochemistry and liver histology of a triple
liver biochemistry and survival free of liver transplantation
(OLT) as well as delays histological progression in early combination of azathioprine and prednisolone with UDCA
after a 1 year of treatment in PBC patients [168]. To the
stage patients [142-144]. Furthermore, two of these studies
best of our knowledge, no further studies on triple therapy in
[142, 144] show that patients in the early stages with a com-
PBC patients have been published, even though results were
plete response to UDCA achieve a survival comparable to
promising.
that of a control population, in line with previous suggestions
by Poupon [145]. There is no doubt that UDCA is now the Intrahepatic Cholestasis of Pregnancy
first choice treatment for PBC at the accepted dosage of 13-
15 mg/Kg/day [118]; however, there is still a minority of ICP is a disease typical of the third trimester of preg-
patients who will not respond to standard treatment and on nancy. It is benign for the mother, but potentially harmful for
the foetus and usually presents with pruritus associated with
162 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

Table 1. Studies on UDCA for the Treatment of Primary Biliary Cirrhosis

First Author Treatment Schedule Type of Study Lenghth of the Study Results

UDCA monotherapy

Poupon [6] UDCA 13-15 mg/kg Prospective observational 2 years Improved biochemistry
b.w./day

Leuschner [119] UDCA 10 mg/Kg b.w./day Double blind controlled trial 6 months Improved biochemistry

Poupon [58] UDCA 13-15 mg/kg Multicenter, double blind placebo 2 years Improved biochemistry, Mayo
b.w./day controlled Risk Score, antimitochondrial-
antibody titer and histology
except for fibrosis

Lindor [121] UDCA 13-15 mg/Kg Randomized placebo controlled 2 years Delayed progression of disease
b.w./day

Poupon [130] UDCA 13-15 mg/kg Multicenter, double blind placebo 2 + 2 years Reduced progression and need
b.w./day controlled + open label extension for liver transplantation

Heathcote [122] UDCA 14 mg/kg b.w./day Multicenter double blind placebo 2 years Improvement of biochemical
controlled parameters

Combes [124] UDCA 10-12 mg/Kg Randomized, double blind pla- 2 years Histology favourably affected
b.w./day cebo controlled by ursodiol in stage I and II
patients

Van Hoogstraten [100] UDCA 10 mg/Kg b.w./day Multicenter Randomized con- 6 months Significant Improvement in
vs 20 mg/Kg b.w./day in trolled open liver biochemistry with higher
non responders to 10 dosages
mg/Kg b.w./day High dose well tolerated with
greater biliary enrichment

Angulo [126] UDCA 13-15 mg/kg Cohort study 5-9 years UDCA delays progression to
b.w./day cirrhosis

Degott [132] UDCA 13-15 mg/kg Double blind placebo controlled 2 – 4 years UDCA delays progression of
b.w./day liver fibrosis

Combes [131] UDCA 10-12 mg/Kg Randomized double blind placebo 2 years No significant differences in
b.w./day controlled florid duct lesions between the
placebo and the treated arm

Poupon [145] UDCA 13-15 mg/Kg/day Cohort study 6 years Longer transplantation free
survival in patients treated with
UDCA

Van Hoogstraten [128] 10 mg/Kg b.w./day Cohort study 2 - 5 years Improved liver biochemistry
Bilirubin the best predictor for
prognosis
Five year treatment failure of
27% and transplantation free
survival of 79%

Angulo [138] UDCA 5-7 mg/Kg b.w./day Randomized open 1 year No significant differences be-
vs 13-15 mg/Kg b.w./day tween standard and high dose
vs 23-25 mg/Kg/day for biliary enrichment and liver
biochemistry
No side effects or toxicity for
any dose

Verma [140] UDCA 0, 300, 600, 900 Observational Crossover each dose for 8 weeks Higher doses better than the low
and 1200 mg/Kg b.w./day with a 4 week washout ones with no significant differ-
between doses ences between the 900 and 1200
doses
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 163

(Table 1. Contd….)

First Author Treatment Schedule Type of Study Lenghth of the Study Results

Pares [120] UDCA 14-16 mg/Kg/day Multicenter double blind placebo 3.4 years Improved biochemical parame-
controlled ters and portal inflammation;
UDCA prevented histological
stage progression; no signifi-
cant effect on death or time to
liver transplantation

Corpechot [125] UDCA 13-15 mg/Kg Randomized double blind placebo 2 + 2 years UDCA delays progression of
b.w./day controlled + open label extension liver fibrosis

Angulo [139] UDCA 28-32 mg/Kg Observational 1 year No significant improvement


b.w./day in incomplete from baseline
responders to 13-15 High dose well tolerated
mh/Kg/dat

Roda [95] UDCA 15 mg/Kg b.w./day Crossover observational 6 months Improved liver tests with higher
vs 30 mg/Kg/day dosages in early stage PBC

Jorgensen [127] UDCA 13-15 mg/Kg Retrospective up to 12 years UDCA of most benefit when
b.w./day instituted in early stage disease

Papatheodoridis [141] UDCA 12-15 mg/kg Prospective randomized con- 12 years No significant effect on long
b.w./day trolled term outcome and survival

Siegel [129] UDCA 13-15 mg/Kg Randomized controlled 4 years Significant weight gain in
b.w./day vs placebo treated patients in the first year
of treament

Combes [123] UDCA 10-12 mg/Kg Open label extension of previous 2 + 2 years No significant differences in
b.w./day randomized double blind placebo time to liver transplantation or
controlled survival

Pares [144] UDCA 15 mg/kg b.w./day Cohort study 7.5 years Survival of good responders at 1
year comparable to control
population, suboptimal survival
in non responders

ter Borg [142] UDCA 13.3 mg/Kg Cohort study 10 years Survival of responders compa-
b.w./day rable to matched general popu-
lation

UDCA + Colchicine

Vuoristo [162] UDCA 12-15 mg/kg Multicenter double blind placebo 2 years UDCA superior to colchicine
b.w./day or Colchicine 1 controlled
mg/day or placebo

Poupon [163] UDCA 13-15 mg/kg Multicenter double blind placebo 2 years No significant differences
b.w./day + Colchicine 1 controlled
mg/day, 5 days per week

Battezzati [165] UDCA 8.8 mg/kg b.w./day Double blind placebo controlled 10 years No added benefit of colchicine
+ Colchicine 1 mg/day to UDCA

UDCA + Methotrexate

Kaplan [167] MTX 15 mg per week or Randomized controlled 10 years No improvement in survival
colchicine 0.6 mg twice compared to UDCA alone
daily + UDCA 12-15
mg/kg b.w./day

Combes [166] UDCA 15 mg/kg b.w./day Multicenter, double blind placebo 7.6 years No added effect of methotrexate
for at least 6 months + controlled
MTX 15 mg/m2 body sur-
face area (maximum dose
20 mg) once a week
164 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

(Table 1. Contd….)

First Author Treatment Schedule Type of Study Lenghth of the Study Results

UDCA + Steroids (Budesonide, Prednisone, Prednisolone)

Angulo [152] UDCA 13-15 mg/kg Observational on suboptimal 1 year Transitory improvement in
b.w./day + Budesonide 9 responders to UDCA biochemistry, increase in Mayo
mg/day Risk Score, steroid related side
effects

Leuschner [153] UDCA 10-15 mg/kg Prospective double blind 2 years Improved biochemistry and
b.w./day + Budesonide 9 histology
mg/day vs UDCA 10-15
mg/kg b.w./day + placebo

Rautiainen [154] UDCA 15 mg/kg b.w./day Randomized controlled 3 years Combination treatment im-
+ Budesonide 6 mg/day vs proved liver histology
UDCA 15 mg/kg b.w./day UDCA alone effective mainly
on biochemistry

Wolfhagen [147] UDCA 10 mg/Kg b.w./day Retrospective 1 year Combined treatment improved
+ prednisone 10 mg/day symptoms and biochemistry to a
larger extent than either treat-
ment alone

Leuschner [146] UDCA 10 mg/Kg b.w./day Randomized placebo controlled 9 months Improved biochemistry and
+ prednisolone 10 mg/day histology with the combination
compared to monotherapy

UDCA + Bezafibrate

Nakai [156] bezafibrate 400 mg/day + Randomized open label 1 year Improved biochemistry with
UDCA 600 mg/day vs combination treatment
UDCA 600 mg/day

Kurihara [160] bezafibrate 400 mg/day vs Randomized open label 1 year Improved biochemistry with
UDCA 600 mg/day both treatments

Ohmoto [161] bezafibrate 400 mg/day Observational 1 year Improved biochemistry with
added to UDCA 600 combination treatment
mg/day

Kanda [159] bezafibrate 400 mg/day + Open label controlled 1 year Improved biochemistry with
UDCA 600 mg/day vs combination treatment
UDCA 600 mg/day

Ikatura [158] Bezafibrate 400 mg/day ± Prospective randomized crossover 6 months + 6 months Improved biochemistry with the
UDCA 600 mg/day combination

Akbar [157] bezafibrate 400 mg/day ± Open label 1 year Improvement of liver function
UDCA dose not stated in tests
the paper Reduced nitrite production by
dendritic cells

UDCA + Prednisone + Azathioprine

Wolfhagen [168] UDCA 10 mg/Kg/day + Randomized placebo controlled 1 year Improved biochemistry and
Prednisone 30 tapered to 10 histology
mg/day + Azathioprine 50
mg/day

elevation of transaminases and serum bile salts [169]. The pruritus called for the development of an effective treatment.
presence of a high percentage of preterm deliveries as well The first report of UDCA efficacy in ICP was published in
as adverse effects of the disease on the foetus (i.e. stillbirths, 1991 showing an improvement in serum bile salts and pruri-
intrapartum foetal distress) and a sometimes severe maternal tus with the treatment [170]. This preliminary observation
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 165

was later confirmed by other groups [171-176] and by two oxia and uterine contractions [191]. Thus the accumulation
randomized, double blind controlled trials which confirmed of bile acids in the foetal compartment (amniotic fluid, cord
that UDCA, both at low and standard dose, is effective in blood) during ICP [179] may have an important role in the
attenuating ICP pruritus and improving maternal transa- pathophysiology of the disease and adverse foetal effects.
minases and serum bile salts (Table 2) [177, 178]. Further- Functional evidence for bile acid transport through the hu-
more, deliveries occurred closer to term in treated mothers man placenta has been described [192, 193]. Placental bile
compared to controls receiving placebo [177]. This impor- acid transport is impaired by cholestasis and restored by
tant observation has been confirmed by our group [179] and UDCA [194, 195] and is probably mediated by MRP iso-
by a retrospective evaluation of UDCA on a twelve year ex- forms [196]. In line with data from experimental animals
perience by a Chilean group [180]. [197] we have recently shown that UDCA administration
results in an up-regulation of Mrp2 human placenta [198].
The efficacy of UDCA in the treatment of ICP has also
These data, coupled with evidence that UDCA administra-
been compared to other proposed treatments such as cho-
tion reduces bile acid accumulation in the foetal compart-
lestyramine, dexamethasone and S-Adenosyl-L-Methionine
ment in ICP [179], suggest that the positive effect of UDCA
(SAMe) [181-184]. Cholestyramine is a bile acid binding might come not only from improved maternal biliary secre-
resin that may be effective in reducing pruritus in patients
tion but also from improved extrusion of bile acids from the
with ICP [185]; however a major drawback is given by its
foetal compartment, which might include a positive effect on
capacity of binding vitamin K possibly leading to coagulo-
foetal liver function (secretion into meconium). These hy-
pathies. In fact, severe fetal intracranial hemorrhage has been
potheses await further demonstrations.
described with its use [186]. This observation, coupled to the
evidence that UDCA is significantly superior to cholestyra- Primary Sclerosing Cholangitis
mine both on pruritus and biochemical parameters in patients
PSC is a chronic cholestatic liver disease causing in-
with ICP, should discourage further use of the resin in this
flammation and obliterative fibrosis of intra and/or extrahe-
condition. Dexamethasone was first proposed in an observa-
patic bile ducts. It is usually progressive, leading to bile sta-
tional study on 10 patients on the basis of its theoretical ca-
sis, fibrosis, cirrhosis and end-stage liver disease with an
pability of reducing the fetoplacental estrogen production
estimated median survival time from diagnosis to death of 12
[187]. A recent randomized controlled trial has shown that
dexamethasone is ineffective in ICP, while UDCA improved years [199-201]. The target cell involved in disease onset
and progression is the cholangiocyte which interacts with
biochemical parameters irrespective of disease severity [182].
other cells (immunocytes, macrophages, stellate cells) in
SAMe has positive effects on bile secretion and increases the
response to injury; the reactive cholangiocyte seems to play
bioavailability of sulphates for the conjugation of bile acids.
a crucial role in liver damage and repair [200]. The patho-
A recent randomized prospective comparative study of UDCA,
genesis of PSC is unknown; a genetic predisposition is
SAMe and a combination of the two drugs has shown that
UDCA and the combination treatment led to a significant shown by the increased risk of disease development in sib-
lings [202] and the association with specific alleles of the
improvement in biochemical parameters compared to SAMe
major histocompatibility complex (MHC) [203] but it seems
monotherapy [183]. The authors observed a faster decrease
that the inheritance of PSC is complex. Immune dysregula-
of serum concentrations of bile acids and transaminases
tion seems to play an important role [204-206] in disease
compared to UDCA monotherapy, suggesting a possible
development. The prevalence of PSC is 8.5 cases per 100.000
synergistic effect [183]. However, this data, combined to
previous evidence from a randomized double blind placebo people in Western Countries, with 70-80% of patients with
concomitant inflammatory bowel disease (IBD), mostly ul-
controlled trial showing that SAMe monotherapy does not
cerative colitis (UC) [207]. The risk of developing cholan-
improve ICP [188], does not support the use of SAMe in
giocarcinoma is increased in patients with PSC [208, 209]
routine clinical practice.
and it seems that the risk of colorectal cancer is higher in
No published study until now has reported any adverse those with PSC and UC than in those with UC alone [210,
effect of UDCA on the foetus; on the contrary its administra- 211].
tion has resulted in deliveries closer to term and a higher
UDCA has been used in PSC since the early 1990s (Ta-
infant weight as compared to those born from controls, sug-
ble 3); initial small studies demonstrated biochemical and in
gesting that treatment may be beneficial for the foetus [177,
some cases histological improvement in patients treated with
180]. Furthemore, the observation that UDCA reduces pri-
low doses of UDCA (10 to 15 mg/Kg/day) [212-214] with
mary bile acids levels in colostrum and its presence in breast
no relevant side effects. Later, Lindor et al. [215] conducted
milk is less than 0.01% of the dose administered to the
mother supports the safety of its use [189]. a larger study on 105 patients with a median follow-up of 2.2
years; this study showed good results with regard to liver
The mechanisms underlying UDCA efficacy in ICP are function tests but no improvement in symptoms or in histol-
still unclear, even though some hypotheses can be made. It is ogy was demonstrated. A meta-analysis of six randomised
likely that the efficacy regarding pruritus results from im- controlled trials of UDCA for the treatment of PSC failed to
provement of cholestasis achieved by the increased biliary show significant differences to the placebo in terms of death,
secretion [140], while the more fascinating aspect is how treatment failures, liver histology or cholangiographic dete-
UDCA may be beneficial in eliminating adverse foetal ef- rioration [216]. However the authors state that the studies
fects. We know that elevated bile acids may induce vasocon- had a low methodological quality, with small samples and a
striction of the umbilical artery [190], leading to foetal an- short follow-up.
166 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

Table 2. Studies on UDCA for the Treatment of Intrahepatic Cholestasis of Pregnancy

First Author Treatment Type of Study Results

UDCA monotherapy

Palma [176] 1 g/day Observational open Improved biochemistry


No adverse effects on the foetus

Floreani [171] 450 mg/day Observational open Improved pruritus and biochemistry

Diaferia [177] 600 mg/day vs placebo Randomized double blind pla- Improved pruritus and biochemistry
cebo controlled Lower rate of prematurity

Palma [178] 1g/day Randomized double blind pla- Improved pruritus and biochemistry
cebo controlled Lower rate of prematurity
Higher birthweight in infants from treated mothers
No adverse effects on the foetus

Nicastri [184] UDCA 600 mg/day vs SAMe Randomised placebo controlled Improved biochemistry and parameters in all groups
800 mg/day vs UDCA + SAMe Combination more effective on bile salts
800 mg/day vs placebo

Brites [189] UDCA 14 mg/Kg b.w./day vs Observational UDCA decreases endogenous bile acids in colostrum
no treatment

Brites [173] UDCA 14 mg/Kg b.w./day vs Observational open label Improved biochemistry
no treatment

Serrano [194] UDCA 1g/day vs no treatment Observational open label UDCA improves pruritus and biochemical parameters
Improved bile acid transport through the placenta

Berkane [172] 10 mg/Kg/day Observational open Improved pruritus and biochemistry

Mazzella [179] UDCA 1.5-2 g/day vs observa- Observational Improved pruritus and biochemistry
tion Reduced accumulation of bile acids in the foetal
compartment
No adverse effects on the foetus

Laifer [174] UDCA 600-1200 mg/day Retrospective Improved pruritus and biochemistry

Zapata [180] UDCA 15 mg/Kg/day vs his- Observational retrospective on Improved pruritus and biochemistry
torical controls 12 years Lower rate of prematurity
Higher birthweight in treated group
No adverse effects on the foetus

Kondrackiene [181] UDCA 8-10 mg/Kg/day vs Randomized open label UDCA is safe and more effective than cholestyramine
cholestyramine 8 g/day Improved pruritus and biochemistry
Lower rate of prematurity

Glantz [182] UDCA 1 g/day vs dexametha- Randomized double blind pla- UDCA improved pruritus and biochemistry
sone 12 mg/day followed vs cebo controlled Dexamethasone had no effect on pruritus and ALT and
placebo was less effective than UDCA in reducing bile acids and
bilirubin
No adverse effects on the foetus

Binder [183] SAMe 500 mg x 2/day i.v. for Prospective randomized open All treatments improved pruritus
14 days and then per os vs label Combined and UDCA monotherapy improved serum
UDCA 250 mg x 3/day vs concentrations of bile acids and transaminases compared
combination of both drugs at to SAMe monotherapy
the same dosage No adverse effects on the foetus

Liu [175] UDCA 900 mg/day vs Randomized open label Improved biochemistry
10%glucos+Vitamin C+Inosine Lower rate of prematurity, foetal asphixya and meconium
staining in amniotic fluid with UDCA
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 167

Table 3. Studies on UDCA for the Treatment of Primary Sclerosing Cholangitis

Author N Drug Treatment Type of Study Treatment Period Results

UDCA 10 mg/kg body Improved biochemistry, fa-


O’Brien [212] 12 Open-label, pilot 2.5 years
weight/day tigue and pruritus

Prospective, randomized,
UDCA 13-15 mg/kg body Improved biochemistry and
Beuers [213] 14 double-blind, placebo- 1 year
weight/day histology
controlled

Double-blind, placebo-
Stiehl [214] 20 UDCA 750 mg/day 3 months Improved biochemistry
controlled

UDCA 13-15 mg/kg body Randomized, double-blind,


Lindor [215] 105 2 years Improved biochemistry
weight/day placebo-controlled

UDCA 25-30 mg/kg body Improvement of biochemistry


Harnois [101] 30 Open-label, pilot 1 year
weight/day and Mayo Risk Score

Improved biochemistry, fibro-


UDCA 20 mk/kg body Randomized, double-blind,
Mitchell [102] 26 2 year sis and cholangiographic ap-
weight/day placebo-controlled
pearances

No differences in biochemis-
UDCA 17-23 mg/kg body Randomized, placebo-
Olsson [217] 219 5 years try, survival, symptoms, qual-
weight/day controlled
ity of life

Trials based on high-dose UDCA schedules were con- provement, but there is no conclusive evidence that it may
ducted in the early 2000’s. The hypothesis was that larger lead to an improvement in histology and survival, although a
doses would be needed to provide a sufficient enrichment of trend toward a statistically significant benefit is present.
bile acids in cholestasis [95]. Two small studies comparing Combined endoscopic and UDCA treatment is necessary
high-dose UDCA (20-30 mg/Kg/day) vs placebo or standard when a major bile duct stricture is present. When PSC is
doses (13-15 mg/Kg/day) suggested a better efficacy of these associated with UC, UDCA may have beneficial effects in
dose regimens, with improvement in liver enzymes, cholan- reducing the risk of colorectal cancer development.
giographic features, liver fibrosis and survival [101, 102].
PEDIATRIC AND INHERITED INTRAHEPATIC
The treatment was well tolerated in both studies. Again, a
larger prospective placebo-controlled study [217], conducted CHOLESTASIS
on 219 patients, failed to show a significant difference in Pediatric cholestatic liver diseases represent one of the
terms of survival or cholangiocarcinoma prevention, although major causes of death for children and although the true
there was a tendency to an improved survival in the treat- prevalence is unknown, it is estimated that roughly 15000
ment group and the study was insufficient to give statisti- children /yr are hospitalised for liver disease in the USA,
cally significant results. about 1 each 2500 live birth, the major disorders being rep-
When a major bile duct stricture is present, UDCA alone resented by biliary atresia, metabolic diseases and intrahe-
is not efficacious; therefore, a combined endoscopic and patic cholestasis [223].
medical treatment in these cases is required. In an 8 years Intrahepatic cholestasis is a heterogeneous spectrum of
follow-up study of patients treated with UDCA in combina- cholestatic diseases characterized by specific syndromes
tion with endoscopic dilatation of strictures, survival was with different clinical features and clinical outcomes ranging
significantly higher in treated patients when compared to the from benign forms, known as benign recurrent intrahepatic
calculated survival without treatment [218]. cholestasis (BRIC), to more severe diseases, indicated as
Some studies suggested that UDCA may be helpful in PFIC (progressive familial intrahepatic cholestasis), requir-
preventing colorectal cancer in patients with associated IBD ing OLT. The pathogenetic basis of these disorders is still
[219, 220]. This hypothesis was then confirmed by clinical partially unknown and only future genetic and molecular
studies: in a cross sectional study, patients treated with insight can permit us to better define and classify them with
UDCA showed a decreased prevalence of colonic dysplasia the aim of carring out more specific and efficacious thera-
(OD, 0.18, 95% CI, 0.05 to 0.61; p= 0.005) [221]; in a pro- peutic strategies. However, over the past 30 yrs significant
spective study of 52 patients with PSC and UC patients progress has been made in differentiating the various syn-
treated with UDCA had a relative risk of 0.26 for dysplasia dromes from the idiopathic neonatal hepatitis spectrum. Spe-
or cancer (95% CI 0.06-0.92; p= 0.034) when compared to cifically, substantial progress has been made in subcategoris-
patients treated with a placebo [222]. ing the different forms of the disease. In fact, the report of a
single topic conference, sponsored by the American Associa-
In conclusion, UDCA has been extensively used in the
treatment of PSC with good results in terms of liver test im- tion for the Study of the Liver has recently been published
168 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

[224]. The new proposed classification subdivides pediatric the best timing for OLT since in some patients the transplant
intrahepatic cholestasis into: a) disorders of membrane is performed because of a poor quality of life rather than end
transport and secretion which include disorders of canalicu- stage liver disease. Furthermore, in some patients, after OLT
lar secretion (PFIC type 2, BRIC2, PFIC type 3 and cystic peculiar severe complications can occur, such as severe non-
fibrosis) and complex or multiorgan disorders (PFIC type 1, alcoholic steatohepatitis [231] and intractable diarrhoea [232,
Bylers disease, BRIC type 1); b) disorders of bile acid syn- 233], with an overall morbidity and mortality of about 10-15
thesis and conjugation; c) disorders of embryogenesis % at one year. The most important medical treatment is rep-
(Alagille syndrome, Caroli’s disease etc); d) unclassified resented by the use of UDCA together with standard nutri-
(idiopatic neonatal hepatitis). tional support. The mechanism of action is not completely
clear and it could depend on the degree of the gene defect
On the basis of the evolving knowledge in terms of clas-
penetrance and the type of PFIC. In fact, in patients with
sification, morphological and clinical findings and the main
PFIC type 3 with partial defect of MDR3 and a residual
molecular pathogenetic mechanisms of bile formation, new
phospholipid concentration in bile, UDCA administration
therapeutic approaches will be investigated. In particular,
could be an effective treatment. From a clinical point of view
agonists and antagonists of nuclear hormone receptors are some studies have demonstrated that chronic UDCA admini-
particularly promising drugs [225, 226].
stration can improve biochemical parameters and alleviate
Up to now, UDCA is the only medical treatment for most pruritus [234, 235]. The most important and convincing
pediatric cholestatic syndromes and, in some of these, it rep- study [236] demonstrated in 39 children (26 with normal
resents the bridge therapy for OLT. GT and 13 with high GT) treated with 20-30 mg /Kg/day
of UDCA for 2-4 yrs a significant decrease of ALT and GT
Familial Intrahepatic Cholestasis levels and an improved nutritional state, with corresponding
Rare inherited forms of cholestasis are the consequence improvement of hepatosplenomegaly and pruritus. Moreover
of mutations in the canalicular transport systems [25] and it has been recently shown [230] that UDCA efficacy is re-
can produce mild to severe hepatic injury, leading to cirrho- lated to early initiation of the therapy, low penetrance of the
sis and liver failure. Mutations of three genes encoding for genetic defect and less severe pre-existing liver damage
three different proteins located at the hepatocyte canalicular Liver Disease in Cystic Fibrosis
membrane are responsible for the development of these cho-
lestatic syndromes. PFIC type 1 (also known as Bylers dis- CF is a multi-organ autosomal recessive disorder, affect-
ease) is caused by mutations in the ATP8B1 gene (ATP8B1), ing roughly 1/ 2500 newborn worldwide, caused by a muta-
resulting in dysfunction of an aminophospholipid flippase tion in the gene encoding the cystic fibrosis transmembrane
(FIC-I) whose activity is essential for bile acid secretion. A conductance regulator (CFTR), which plays a key role in
mild cholestatic form presenting as BRIC1, is associated maintaining fluid balance across the epithelium [237]. In CF,
with ATP8B1 mutations too. the CFTR mutations are associated with an epithelial secre-
tory defect, with reduced luminal hydration and progressive
PFIC type 2 is the consequence of mutations in the luminal obstruction in different organs (sweat gland, pan-
ABCB11 gene (ABCB11), which encodes for the canalicular creas, liver, lung).
bile acid transporter (BSEP). Mutations of ABCB11 are as-
sociated both with progressive (PFIC type 2) and benign Hepatobiliary involvement ranges from 18 to 37% [238,
(BRIC2) cholestatic syndromes. Unlike BRIC1, BRIC2 is 239] and represents the third cause of death in CF, account-
often associated with gallstones [227]. ing for about 2.3% of overall mortality [240]. In the hepato-
biliary system, CFTR is expressed only in the cholangiocytes
PFIC type 3 is the consequence of mutations of the and gallbladder epithelium, but not in the hepatocytes [241].
ABCB4 gene (ABCB4), which resuls in a dysfunctional Thus the primary event into the development of biliary cir-
phosphatidylcholine flippase and leads to defective biliary rhosis is the bile duct cell injury [242] due to a long-standing
phospholipid secretion. In these patients, bile acid secretion impairment in the ductular bile flow. For these reasons the
is maintained. The consequence of maintained bile acid se- hepatobiliary disease in CF is considered the first inherent
cretion, in the absence of a sufficient concentration of biliary liver disease resulting from an alterated secretory function of
phospholipids, is a damage to the apical membrane of the the biliary epithelium; accumulation of potentially toxic
cholangiocytes, with consequently high GT activity, which compounds, such as hydrophobic bile acids, and eventually
distinguishes this form from PFIC/BRIC type 1 and type 2. common bile duct compression by pancreatic fibrosis or sec-
ABCB4 mutations can be associated with either progressive ondary sclerosing cholangitis may represent additional fac-
or milder phenotypes [25]. Moreover, ABCB4 mutations tors [243].
have been found to be associated with anti-mitochondrial
antibodies negative PBC and with intrahepatic cholesterol The spectrum of hepatic manifestations ranges from as-
gallstones [228, 229]. ymptomatic elevation of the liver function tests (10-35 %), to
neonatal cholestasis (2%), hepatic steatosis/ steatohepatitis
The best strategy for the treatment of PFIC has not been (20-60%), focal biliary cirrhosis (11-70%) and multilobular
completely established and probably depends on the type of biliary cirrhosis (5-15 %); focal biliary cirrhosis being con-
PFIC, the severity of the disease at diagnosis, and mainly on sidered the specific hepatic lesion of CF which can result in
the penetrance of gene defects. OLT is considered to be a multilobular cirrhosis and liver failure [244]. In recent years
definitive therapeutic strategy, with a survival rate of about the improved life expectancy in patients with CF due to im-
92 % at five years [230]. However, it is difficult to establish proved management of lung and pancreatic manifestations of
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 169

the disease prompted to focus on the increasing importance mainly on pruritus, although results are conflicting [234,
of the hepatobiliary disorder. At present, UDCA is the only 260, 261].
approved drug for hepatobiliary disease in CF. It has been
Biliary Atresia
suggested that UDCA can replays the hydrophobic bile acids
and stimulates Cl- secretion through a Ca++ dependent Cl- Biliary atresia is the most common neonatal cholestatic
channel [245] or by a mechanism of ATP secretion different disorder, with a prevalence ranging from 1/8000 in Far East
from the CFTR [246]. countries (Asian populations) to 1/18000 in European popu-
lations (European countries), with female predominance
Early uncontrolled studies [247-249] and two RCTs [250,
251] showed an improvement in the biochemical and the [262]. The disease is characterized by progressive inflamma-
tory cholangiopathy evolving toward sclerosing fibrosis,
nutritional status of the patients treated with UDCA at a dose
involving early the extrahepatic biliary tree, and eventually
of 15 mg/kg/day. An improvement on liver histology was
later the intrahepatic biliary tree. For the two different forms
also documented [61]. Evidence suggests that higher UDCA
of the disease, i.e the fetal (20%) and the post-natal (80%),
doses (20-30 mg/Kg/day) are more efficacious than standard
genetical, environmental (viral infection?), immunological
doses (5-15 mg/Kg/day) [248, 252].
and embryological factors have been hypothesized [263].
Although UDCA is the only drug accepted for the treat-
The treatment of choice of biliary atresia in children
ment of hepatobiliary disease in CF, its effectiveness on the
younger than 3 months is the portoenterostomy (Kasai’s op-
natural history of liver disease in CF has yet to be defined
eration) that allows an efficacious bile flow, with an im-
[253]. In particular, its effect on the complications of liver
provement of serum bilirubin levels, jaundice and liver en-
disease (variceal hemorrage, portal hypertension), on the
zymes. However, most of the patients will progress to cir-
need for OLT or on overall survival are unknown. Thus it is
necessary to design adequate RCTs to justify the long- term rhosis [263]. After Kasai’s operation, the survival rate at 20
yrs is less than 20% with native liver [264]; thus Kasai’s
use of UDCA. Additional points to be elucidated are repre-
operation should be considered a bridge treatment to OLT. In
sented by the preventive effect of UDCA in either high risk
fact OLT is the definite treatment, with survival rate of about
subjects or asymptomatic patients for hepatobiliary disease.
84 % at 10 years [265].
Alagille Syndrome
UDCA has been employed after Kasai’s operation, in
Alagille syndrome (AGS) is an autosomal dominant dis- order to delay progression to cirrhosis, although to date no
order, clinically defined by the presence of at least three of randomized clinical trial has been performed. Preliminary
the following major features: chronic cholestasis, congenital reports [266] and a more recent study [267] demonstrated an
heart disease, peculiar faces, butterfly- like vertebrae and improvement of biochemical parameters as well as an in-
posterior embryotoxon [254]. AGS occurs in about 1/10000 crease of body weight. However, neither an increase of the
live births and is caused by mutations in the Jag1 gene, patients’ survival time nor a reduction of the need for OLT
which encodes a protein belonging to a family of ligands of were observed on the caution. On the contrary a combined
the Notch receptor [255, 256]. Mutation of Jag1 were identi- therapy with UDCA together with steroids and antibiotics
fied in about 94% of the subjects with AGS [257]. Notch 2 has been suggested to be able to reduce the need of early
mutations can also be present in patients without Jag1 muta- OLT [268].
tions, confirming that AGS is a heterogeneous disorder
URSODEOXYCHOLIC ACID IN DIFFERENT CLINI-
[258].
CAL SETTINGS
Jag1 is involved in cell differentation and regulation of
biliary epithelium development [255, 256] and its mutation Besides therapeutic use in chronic cholestatic syndromes
and in gallstone disease, UDCA use has been proposed for
leads to bile duct paucity with an increased portal tract to
prevention of colon carcinoma in PSC patients [198,199],
bile duct ratio. Hepatic manifestation in ASG are present in
reduction of liver complications in the graft versus host dis-
the majority of children, mainly in the first three months of
ease [269, 270], decrease of rejection after liver and heart
life, with a wide clinical spectrum ranging from mild cho-
transplantation [271, 272], and treatment of bile reflux gas-
lestasis to jaundice, pruritus and to progressive liver failure.
Jaundice is present as a result of conjugated bilirubin associ- tritis [273, 274]. Among the above mentioned clinical set-
tings, the most promising seems to be UDCA use in preven-
ated with high serum levels of bile acids, tryglicerides and
tion of colon carcinoma, which starts from observations in
cholesterol, and with typical xanthomas. The management of
the in vitro and animal models showing that UDCA exerts
liver disease in AGS is more difficult than in other cho-
antineoplastic effects on colonic mucosa [275-277]. Besides
lestatic pediatric diseases because of the presence of serious
the proposed UDCA prevention on colon carcinoma in PSC
extrahepatic manifestations, conditioning the survival. OLT
represents the treatment of choice in end stage liver disease, patients with concomitant ulcerative colitis [221, 222], a
reduced recurrence of colorectal adenoma has been demon-
in severe hypercholesterolemia and in diffuse xanthomas,
strated in PBC patients treated with UDCA, as compared to
with a survival rate of about 80% at 5 years [259]. This rela-
controls [278]. Finally, the results of a recent phase-III study
tively low survival rate is due to the multisystemic disease
in patients who had undergone removal of a colorectal ade-
involvement.
noma demonstrate that UDCA treatment is associated with a
UDCA in AGS, unlike other cholestatic syndromes, has statistically significant reduction in recurrence of adenomas
no efficacy on the biochemical and histological parameters, with high-grade dysplasia [279], supporting the possible role
or on survival, but it can be utilized as a symptomatic drug, of this bile acid in the chemoprevention strategies.
170 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

DEVELOPMENT OF UDCA-DERIVATIVES FOR GSH = Reduced glutathione


CLINICAL USE IN HEPATOBILIARY DISEASES
FIC1 = Familial intrahepatic cholestasis 1
Although UDCA is an effective agent in different cho-
ASBT = Apical sodium-bile acid cotransporter
lestatic liver diseases, its physico-chemical characteristics
and biotransformation routes may limit the potential thera- I-BABP = Ileal bile acid binding protein
peutic efficacy. For this reason, efforts have been performed
to identify UDCA derivatives with improved efficacy. OST-OST = Heteromeric organic solute transporter -

The C23 (C24-nor) homolog of UDCA, nor-ursodeoxy- CYP = Cytochrome p-450


cholic acid (nor-UDCA) has shown peculiar biotransforma- FXR = Farnesoid X receptor
tion and physiological properties in humans [280]. In par-
ticular, in human liver nor-UDCA is glucuronidated rather PXR = Pregnane X receptor
than amidated with glycline or taurine, with considerable VDR = Vitamin D receptor
renal elimination of the C23 glucuronide, which is its domi-
nant metabolite in humans [280]. Moreover, nor-UDCA oral CAR = Constitutive androstane receptor
administration in two volunteers induced a bicarbonate-rich PPAR- = Peroxisome proliferators-activated recep-
hypercholeresis, with lower biliary secretion of phospholip- tor-alpha
ids and cholesterol [280]. Therefore, the hypercholeretic
properties of nor-UDCA suggest its potential benefit in cho- SHP = Small heterodimer partner
lestatic liver diseases.
DCA = Deoxycholic acid
Finally, it has recently been shown that nor-UDCA is CDCA = Chenodeoxycholic acid
superior to UDCA in the treatment of a mice model of scle-
rosing cholangitis [281]. APAF-1 = Apoptotic protease activating factor 1
CONCLUSIONS HCV = Hepatitis C virus
UDCA use in the treatment of chronic cholestatic dis- LCA = Lithocholic acid
eases is consolidated and growing evidence has been pro-
CMpH = Critical micellization pH
duced in the last years on its beneficial role not only in the
biochemical improvement but also in the long term remis- PEG = Polyethylenglycol
sion of cholestatic syndromes. The optimal safety profile and
a better knowledge of the optimal dose for inducing and GT = Gamma-glutamyltranspeptidase
maintaining disease remission in chronic cholestatic diseases ALP = Alkaline phosphatases
prompted to the common use of high doses UDCA in the
clinical practice. Further studies with well defined end points ALT = Alanine aminotransferase
(e.g. death or liver transplantation) are needed in order to OLT = Ortotopic liver transplantation
better clarify the long term efficacy of UDCA in PSC, while
the long term efficacy on major end points in PBC has been SAMe = S-Adenosyl-L-Methionine
clearly demonstrated. Pediatric cholestasis is also an impor- MHC = Major histocompatibility complex
tant field in which UDCA has achieved important therapeu-
tic results. Continuous advances in the fields of physiology IBD = Inflammatory bowel disease
and molecular biology have better clarified the mechanisms
UC = Ulcerative colitis
of different cholestatic syndromes and will possibly help in
the development of new UDCA derivatives with higher PFIC = Progressive familial intrahepatic cholestasis
therapeutic effectiveness.
BRIC = Benign recurrent intrahepatic cholestasis
ABBREVIATIONS
AMA = Antimitochondrial antibodies
UDCA = Ursodeoxycholic acid
CFTR = Cystic fibrosis transmembrane conductance
PBC = Primary biliary cirrhosis regulator
PSC = Primary sclerosing cholangitis AGS = Alagille syndrome
ICP = Intrahepatic cholestasis of pregnancy Used abbreviations: According to the conventional use of
abbreviations, upper case will be used to indicate human
NTCP = Na+/taurocholate cotransporter
proteins, while lower case (with upper case only for the first
OATPs = Organic anion transport proteins letter) will indicate proteins in rodents and non human spe-
cies in general.
HBAB = Hepatic bile acid binding protein
REFERENCES
BSEP = Bile salt export pump
[1] Hofmann AF. Pharmacology of ursodeoxycholic acid, an enterohe-
MRP2/3 = Multidrug resistance protein 2/3 patic drug. Scand J Gastroenterol Suppl 1994; 204: 1-15.
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 171

[2] Hagey LR, Crombie DL, Espinosa E, et al. Ursodeoxycholic acid [26] Wong MH, Oelkers P, Dawson PA. Identification of a mutation in
in the Ursidae: biliary bile acids of bears, pandas, and related car- the ileal sodium-dependent bile acid transporter gene that abolishes
nivores. J Lipid Res 1993; 34: 1911-1917. transport activity. J Biol Chem 1995; 270: 27228-27234.
[3] Makino J, Shinizaki K, Nakagawa K, Yoshino K. Dissolution of [27] Oelkers P, Kirby LC, Heubi JE, Dawson PA. Primary bile acid
cholesterol gallstones by long-term administration of ursodeoxy- malabsorption caused by mutations in the ileal sodium-dependent
cholic acid. Jpn J Gastroenterol 1975; 72: 690. bile acid transporter gene (SLC10A2). J Clin Invest 1997; 99:
[4] Roda E, Bazzoli F, Labate AM, et al. Ursodeoxycholic acid vs. 1880-1887.
chenodeoxycholic acid as cholesterol gallstone-dissolving agents: a [28] Montagnani M, Aldini R, Roda A, Roda E. New insights in the
comparative randomized study. Hepatology 1982; 2: 804-810. physiology and molecular basis of the intestinal bile acid absorp-
[5] Leuschner U, Leuschner M, Sieratzki J, Kurtz W, Hubner K. Gall- tion. Ital J Gastroenterol Hepatol 1998; 30: 435-440.
stone dissolution with ursodeoxycholic acid in patients with [29] Aldini R, Roda A, Lenzi PL, et al. Bile acid active and passive ileal
chronic active hepatitis and two years follow-up. A pilot study. Dig transport in the rabbit: effect of luminal stirring. Eur J Clin Invest
Dis Sci 1985; 30: 642-649. 1992; 22: 744-750.
[6] Poupon R, Chretien Y, Poupon RE, et al. Is ursodeoxycholic acid [30] Kramer W, Girbig F, Gutjahr U, et al. Intestinal bile acid absorp-
an effective treatment for primary biliary cirrhosis? Lancet 1987; 1: tion. Na(+)-dependent bile acid transport activity in rabbit small in-
834-836. testine correlates with the coexpression of an integral 93-kDa and a
[7] Paumgartner G. Medical treatment of cholestatic liver diseases: peripheral 14-kDa bile acid-binding membrane protein along the
From pathobiology to pharmacological targets. World J Gastroen- duodenum-ileum axis. J Biol Chem 1993; 268: 18035-18046.
terol 2006; 12: 4445-4451. [31] Gong YZ, Everett ET, Schwartz DA, Norris JS, Wilson FA. Mo-
[8] Hofmann AF. The continuing importance of bile acids in liver and lecular cloning, tissue distribution, and expression of a 14-kDa bile
intestinal disease. Arch Intern Med 1999; 159: 2647-2658. acid-binding protein from rat ileal cytosol. Proc Natl Acad Sci U S
[9] Chiang JY. Regulation of bile acid synthesis: pathways, nuclear A 1994; 91: 4741-4745.
receptors, and mechanisms. J Hepatol 2004; 40: 539-551. [32] Oelkers P, Dawson PA. Cloning and chromosomal localization of
[10] Hofmann AF. The enterohepatic circulation of bile acids in man. the human ileal lipid-binding protein. Biochim Biophys Acta 1995;
Adv Intern Med 1976; 21: 501-534. 1257: 199-202.
[11] Trauner M, Wagner M, Fickert P, Zollner G. Molecular regulation [33] Ballatori N, Christian WV, Lee JY, et al. OSTalpha-OSTbeta: a
of hepatobiliary transport systems: clinical implications for under- major basolateral bile acid and steroid transporter in human intesti-
standing and treating cholestasis. J Clin Gastroenterol 2005; 39: nal, renal, and biliary epithelia. Hepatology 2005; 42: 1270-1279.
S111-124. [34] Dawson PA, Hubbert M, Haywood J, et al. The heteromeric or-
[12] Kullak-Ublick GA, Stieger B, Hagenbuch B, Meier PJ. Hepatic ganic solute transporter alpha-beta, Ostalpha-Ostbeta, is an ileal ba-
transport of bile salts. Semin Liver Dis 2000; 20: 273-292. solateral bile acid transporter. J Biol Chem 2005; 280: 6960-6968.
[13] Aldini R, Montagnani M, Roda A, et al. Intestinal absorption of [35] Xia X, Francis H, Glaser S, Alpini G, LeSage G. Bile acid interac-
bile acids in the rabbit: different transport rates in jejunum and il- tions with cholangiocytes. World J Gastroenterol 2006; 12: 3553-
eum. Gastroenterology 1996; 110: 459-468. 3563.
[14] Wilson FA. Intestinal transport of bile acids. Am J Physiol 1981; [36] Lazaridis KN, Tietz P, Wu T, et al. Alternative splicing of the rat
241: G83-92. sodium/bile acid transporter changes its cellular localization and
[15] Wilton JC, Matthews GM, Burgoyne RD, et al. Fluorescent chol- transport properties. Proc Natl Acad Sci U S A 2000; 97: 11092-
eretic and cholestatic bile salts take different paths across the hepa- 11097.
tocyte: transcytosis of glycolithocholate leads to an extensive redis- [37] Benedetti A, Di Sario A, Marucci L, et al. Carrier-mediated trans-
tribution of annexin II. J Cell Biol 1994; 127: 401-410. port of conjugated bile acids across the basolateral membrane of
[16] Gerloff T, Stieger B, Hagenbuch B, et al. The sister of P- biliary epithelial cells. Am J Physiol 1997; 272: G1416-1424.
glycoprotein represents the canalicular bile salt export pump of [38] Donner MG, Keppler D. Up-regulation of basolateral multidrug
mammalian liver. J Biol Chem 1998; 273: 10046-10050. resistance protein 3 (Mrp3) in cholestatic rat liver. Hepatology
[17] Strautnieks SS, Bull LN, Knisely AS, et al. A gene encoding a 2001; 34: 351-359.
liver-specific ABC transporter is mutated in progressive familial in- [39] Jacquemin E, De Vree JM, Cresteil D, et al. The wide spectrum of
trahepatic cholestasis. Nat Genet 1998; 20: 233-238. multidrug resistance 3 deficiency: from neonatal cholestasis to cir-
[18] Noe J, Stieger B, Meier PJ. Functional expression of the canalicular rhosis of adulthood. Gastroenterology 2001; 120: 1448-1458.
bile salt export pump of human liver. Gastroenterology 2002; 123: [40] Rost D, Konig J, Weiss G, et al. Expression and localization of the
1659-1666. multidrug resistance proteins MRP2 and MRP3 in human gallblad-
[19] Paolini M, Pozzetti L, Montagnani M, et al. Ursodeoxycholic acid der epithelia. Gastroenterology 2001; 121: 1203-1208.
(UDCA) prevents DCA effects on male mouse liver via up- [41] Soroka CJ, Lee JM, Azzaroli F, Boyer JL. Cellular localization and
regulation of CYP [correction of CXP]and preservation of BSEP up-regulation of multidrug resistance-associated protein 3 in hepa-
activities. Hepatology 2002; 36: 305-314. tocytes and cholangiocytes during obstructive cholestasis in rat
[20] Taniguchi K, Wada M, Kohno K, et al. A human canalicular mul- liver. Hepatology 2001; 33: 783-791.
tispecific organic anion transporter (cMOAT) gene is overex- [42] Hofmann AF. Current concepts of biliary secretion. Dig Dis Sci
pressed in cisplatin-resistant human cancer cell lines with de- 1989; 34: 16S-20S.
creased drug accumulation. Cancer Res 1996; 56: 4124-4129. [43] Kanai S, Kitani K, Sato Y. The nature of choleresis induced by
[21] Paulusma CC, van Geer MA, Evers R, et al. Canalicular multis- deoxycholate and its conjugates in the rabbit. Jpn J Physiol 1989;
pecific organic anion transporter/multidrug resistance protein 2 39: 907-918.
mediates low-affinity transport of reduced glutathione. Biochem J [44] Buscher HP, Miltenberger C, MacNelly S, Gerok W. The his-
1999; 338 (Pt 2): 393-401. toautoradiographic localization of taurocholate in rat liver after bile
[22] Cui Y, Konig J, Buchholz JK, et al. Drug resistance and ATP- duct ligation. Evidence for ongoing secretion and reabsorption
dependent conjugate transport mediated by the apical multidrug re- processes. J Hepatol 1989; 8: 181-191.
sistance protein, MRP2, permanently expressed in human and ca- [45] Alpini G, McGill JM, Larusso NF. The pathobiology of biliary
nine cells. Mol Pharmacol 1999; 55: 929-937. epithelia. Hepatology 2002; 35: 1256-1268.
[23] Konig J, Nies AT, Cui Y, Leier I, Keppler D. Conjugate export [46] Christie DM, Dawson PA, Thevananther S, Shneider BL. Com-
pumps of the multidrug resistance protein (MRP) family: localiza- parative analysis of the ontogeny of a sodium-dependent bile acid
tion, substrate specificity, and MRP2-mediated drug resistance. transporter in rat kidney and ileum. Am J Physiol 1996; 271: G377-
Biochim Biophys Acta 1999; 1461: 377-394. 385.
[24] Trauner M, Boyer JL. Bile salt transporters: molecular characteri- [47] Schaub TP, Kartenbeck J, Konig J, et al. Expression of the conju-
zation, function, and regulation. Physiol Rev 2003; 83: 633-671. gate export pump encoded by the mrp2 gene in the apical mem-
[25] Oude Elferink RP, Paulusma CC, Groen AK. Hepatocanalicular brane of kidney proximal tubules. J Am Soc Nephrol 1997; 8:
transport defects: pathophysiologic mechanisms of rare diseases. 1213-1221.
Gastroenterology 2006; 130: 908-925. [48] Schaub TP, Kartenbeck J, Konig J, et al. Expression of the MRP2
gene-encoded conjugate export pump in human kidney proximal
172 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

tubules and in renal cell carcinoma. J Am Soc Nephrol 1999; 10: cholestasis and effect of ursodeoxycholic acid treatment. J Hepatol
1159-1169. 1995; 23: 283-289.
[49] Beuers U, Boyer JL, Paumgartner G. Ursodeoxycholic acid in [72] Heuman DM, Bajaj RS, Lin Q. Adsorption of mixtures of bile salt
cholestasis: potential mechanisms of action and therapeutic appli- taurine conjugates to lecithin-cholesterol membranes: implications
cations. Hepatology 1998; 28: 1449-1453. for bile salt toxicity and cytoprotection. J Lipid Res 1996; 37: 562-
[50] Pusl T, Beuers U. Ursodeoxycholic acid treatment of vanishing bile 573.
duct syndromes. World J Gastroenterol 2006; 12: 3487-3495. [73] Heuman DM, Mills AS, McCall J, et al. Conjugates of ursodeoxy-
[51] Paumgartner G, Beuers U. Mechanisms of action and therapeutic cholate protect against cholestasis and hepatocellular necrosis
efficacy of ursodeoxycholic acid in cholestatic liver disease. Clin caused by more hydrophobic bile salts. In vivo studies in the rat.
Liver Dis 2004; 8: 67-81, vi. Gastroenterology 1991; 100: 203-211.
[52] Beuers U. Drug insight: Mechanisms and sites of action of ursode- [74] Heuman DM, Bajaj R. Ursodeoxycholate conjugates protect
oxycholic acid in cholestasis. Nat Clin Pract Gastroenterol Hepatol against disruption of cholesterol-rich membranes by bile salts. Gas-
2006; 3: 318-328. troenterology 1994; 106: 1333-1341.
[53] Lee J, Azzaroli F, Wang L, et al. Adaptive regulation of bile salt [75] Benz C, Angermuller S, Tox U, et al. Effect of tauroursodeoxy-
transporters in kidney and liver in obstructive cholestasis in the rat. cholic acid on bile-acid-induced apoptosis and cytolysis in rat he-
Gastroenterology 2001; 121: 1473-1484. patocytes. J Hepatol 1998; 28: 99-106.
[54] Trauner M, Meier PJ, Boyer JL. Molecular pathogenesis of cho- [76] Patel T, Bronk SF, Gores GJ. Increases of intracellular magnesium
lestasis. N Engl J Med 1998; 339: 1217-1227. promote glycodeoxycholate-induced apoptosis in rat hepatocytes. J
[55] Zollner G, Fickert P, Fuchsbichler A, et al. Role of nuclear bile Clin Invest 1994; 94: 2183-2192.
acid receptor, FXR, in adaptive ABC transporter regulation by [77] Faubion WA, Guicciardi ME, Miyoshi H, et al. Toxic bile salts
cholic and ursodeoxycholic acid in mouse liver, kidney and intes- induce rodent hepatocyte apoptosis via direct activation of Fas. J
tine. J Hepatol 2003; 39: 480-488. Clin Invest 1999; 103: 137-145.
[56] Paumgartner G, Beuers U. Ursodeoxycholic acid in cholestatic [78] Sodeman T, Bronk SF, Roberts PJ, Miyoshi H, Gores GJ. Bile salts
liver disease: mechanisms of action and therapeutic use revisited. mediate hepatocyte apoptosis by increasing cell surface trafficking
Hepatology 2002; 36: 525-531. of Fas. Am J Physiol Gastrointest Liver Physiol 2000; 278: G992-
[57] Trauner M, Graziadei IW. Review article: mechanisms of action 999.
and therapeutic applications of ursodeoxycholic acid in chronic [79] Hsu YT, Wolter KG, Youle RJ. Cytosol-to-membrane redistribu-
liver diseases. Aliment Pharmacol Ther 1999; 13: 979-996. tion of Bax and Bcl-X(L) during apoptosis. Proc Natl Acad Sci U S
[58] Poupon RE, Balkau B, Eschwege E, Poupon R. A multicenter, A 1997; 94: 3668-3672.
controlled trial of ursodiol for the treatment of primary biliary cir- [80] Higuchi H, Gores GJ. Bile acid regulation of hepatic physiology:
rhosis. UDCA-PBC Study Group. N Engl J Med 1991; 324: 1548- IV. Bile acids and death receptors. Am J Physiol Gastrointest Liver
1554. Physiol 2003; 284: G734-738.
[59] Poupon RE, Chretien Y, Poupon R, Paumgartner G. Serum bile [81] Rodrigues CM, Fan G, Ma X, Kren BT, Steer CJ. A novel role for
acids in primary biliary cirrhosis: effect of ursodeoxycholic acid ursodeoxycholic acid in inhibiting apoptosis by modulating mito-
therapy. Hepatology 1993; 17: 599-604. chondrial membrane perturbation. J Clin Invest 1998; 101: 2790-
[60] Medina JF, Martinez A, Vazquez JJ, Prieto J. Decreased anion 2799.
exchanger 2 immunoreactivity in the liver of patients with primary [82] Rodrigues CM, Fan G, Wong PY, Kren BT, Steer CJ. Ursodeoxy-
biliary cirrhosis. Hepatology 1997; 25: 12-17. cholic acid may inhibit deoxycholic acid-induced apoptosis by
[61] Prieto J, Qian C, Garcia N, Diez J, Medina JF. Abnormal expres- modulating mitochondrial transmembrane potential and reactive
sion of anion exchanger genes in primary biliary cirrhosis. Gastro- oxygen species production. Mol Med 1998; 4: 165-178.
enterology 1993; 105: 572-578. [83] Rodrigues CM, Ma X, Linehan-Stieers C, et al. Ursodeoxycholic
[62] Prieto J, Garcia N, Marti-Climent JM, et al. Assessment of biliary acid prevents cytochrome c release in apoptosis by inhibiting mito-
bicarbonate secretion in humans by positron emission tomography. chondrial membrane depolarization and channel formation. Cell
Gastroenterology 1999; 117: 167-172. Death Differ 1999; 6: 842-854.
[63] Schuetz EG, Strom S, Yasuda K, et al. Disrupted bile acid homeo- [84] Botla R, Spivey JR, Aguilar H, Bronk SF, Gores GJ. Ursodeoxy-
stasis reveals an unexpected interaction among nuclear hormone cholate (UDCA) inhibits the mitochondrial membrane permeability
receptors, transporters, and cytochrome P450. J Biol Chem 2001; transition induced by glycochenodeoxycholate: a mechanism of
276: 39411-39418. UDCA cytoprotection. J Pharmacol Exp Ther 1995; 272: 930-938.
[64] Bodin K, Bretillon L, Aden Y, et al. Antiepileptic drugs increase [85] Benz C, Angermuller S, Otto G, et al. Effect of tauroursodeoxy-
plasma levels of 4beta-hydroxycholesterol in humans: evidence for cholic acid on bile acid-induced apoptosis in primary human hepa-
involvement of cytochrome p450 3A4. J Biol Chem 2001; 276: tocytes. Eur J Clin Invest 2000; 30: 203-209.
38685-38689. [86] Azzaroli F, Mehal W, Soroka CJ, et al. Ursodeoxycholic acid di-
[65] Lew JL, Zhao A, Yu J, et al. The farnesoid X receptor controls minishes Fas-ligand-induced apoptosis in mouse hepatocytes. He-
gene expression in a ligand- and promoter-selective fashion. J Biol patology 2002; 36: 49-54.
Chem 2004; 279: 8856-8861. [87] Schoemaker MH, Conde de la Rosa L, Buist-Homan M, et al.
[66] Ellis E, Axelson M, Abrahamsson A, et al. Feedback regulation of Tauroursodeoxycholic acid protects rat hepatocytes from bile acid-
bile acid synthesis in primary human hepatocytes: evidence that induced apoptosis via activation of survival pathways. Hepatology
CDCA is the strongest inhibitor. Hepatology 2003; 38: 930-938. 2004; 39: 1563-1573.
[67] Lee FY, Lee H, Hubbert ML, Edwards PA, Zhang Y. FXR, a mul- [88] Graf D, Kurz AK, Fischer R, Reinehr R, Haussinger D. Taurolitho-
tipurpose nuclear receptor. Trends Biochem Sci 2006; 31: 572-580. cholic acid-3 sulfate induces CD95 trafficking and apoptosis in a c-
[68] Miura T, Ouchida R, Yoshikawa N, et al. Functional modulation of Jun N-terminal kinase-dependent manner. Gastroenterology 2002;
the glucocorticoid receptor and suppression of NF-kappaB- 122: 1411-1427.
dependent transcription by ursodeoxycholic acid. J Biol Chem [89] Koga H, Sakisaka S, Ohishi M, Sata M, Tanikawa K. Nuclear DNA
2001; 276: 47371-47378. fragmentation and expression of Bcl-2 in primary biliary cirrhosis.
[69] Jung D, Fantin AC, Scheurer U, Fried M, Kullak-Ublick GA. Hu- Hepatology 1997; 25: 1077-1084.
man ileal bile acid transporter gene ASBT (SLC10A2) is transacti- [90] Fickert P, Trauner M, Fuchsbichler A, et al. Oncosis represents the
vated by the glucocorticoid receptor. Gut 2004; 53: 78-84. main type of cell death in mouse models of cholestasis. J Hepatol
[70] Eloranta JJ, Jung D, Kullak-Ublick GA. The human Na+- 2005; 42: 378-385.
taurocholate cotransporting polypeptide gene is activated by gluco- [91] Guicciardi ME, Gores GJ. Cholestatic hepatocellular injury: what
corticoid receptor and peroxisome proliferator-activated receptor- do we know and how should we proceed. J Hepatol 2005; 42: 297-
gamma coactivator-1alpha, and suppressed by bile acids via a small 300.
heterodimer partner-dependent mechanism. Mol Endocrinol 2006; [92] Kountouras J, Zavos C, Chatzopoulos D. Apoptosis in hepatitis C. J
20: 65-79. Viral Hepat 2003; 10: 335-342.
[71] Stiehl A, Rudolph G, Sauer P, Theilmann L. Biliary secretion of [93] Fabbri C, Marchetto S, Pezzoli A, et al. Efficacy of ursodeoxy-
bile acids and lipids in primary sclerosing cholangitis. Influence of cholic acid in association with alpha-interferon for chronic hepatitis
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 173

C in alpha-interferon non-responder patients. Eur J Gastroenterol [117] Simoni P, Sabatini L, Baraldini M, et al. Pharmacokinetics and
Hepatol 2000; 12: 511-515. bioavailability of four modified-release ursodeoxycholic acid
[94] Sauer P, Benz C, Rudolph G, et al. Influence of cholestasis on preparations for once-a-day administration. Int J Clin Pharmacol
absorption of ursodeoxycholic acid. Dig Dis Sci 1999; 44: 817-822. Res 2002; 22: 37-45.
[95] Roda E, Azzaroli F, Nigro G, et al. Improved liver tests and greater [118] Heathcote EJ. Management of primary biliary cirrhosis. The
biliary enrichment with high dose ursodeoxycholic acid in early American Association for the Study of Liver Diseases practice
stage primary biliary cirrhosis. Dig Liver Dis 2002; 34: 523-527. guidelines. Hepatology 2000; 31: 1005-1013.
[96] Roda E, Aldini R, Bazzoli F, et al. Pathophysiology and pharma- [119] Leuschner U, Fischer H, Kurtz W, et al. Ursodeoxycholic acid in
cotherapy of cholelithiasis. Pharmacol Ther 1992; 53: 167-185. primary biliary cirrhosis: results of a controlled double-blind trial.
[97] Hofmann AF, Popper H. Ursodeoxycholic acid for primary biliary Gastroenterology 1989; 97: 1268-1274.
cirrhosis. Lancet 1987; 2: 398-399. [120] Pares A, Caballeria L, Rodes J, et al. Long-term effects of ursode-
[98] Stiehl A, Raedsch R, Rudolph G. Acute effects of ursodeoxycholic oxycholic acid in primary biliary cirrhosis: results of a double-blind
and chenodeoxycholic acid on the small intestinal absorption of controlled multicentric trial. UDCA-Cooperative Group from the
bile acids. Gastroenterology 1990; 98: 424-428. Spanish Association for the Study of the Liver. J Hepatol 2000; 32:
[99] Walker S, Rudolph G, Raedsch R, Stiehl A. Intestinal absorption of 561-566.
ursodeoxycholic acid in patients with extrahepatic biliary obstruc- [121] Lindor KD, Dickson ER, Baldus WP, et al. Ursodeoxycholic acid
tion and bile drainage. Gastroenterology 1992; 102: 810-815. in the treatment of primary biliary cirrhosis. Gastroenterology
[100] Van Hoogstraten HJ, De Smet MB, Renooij W, et al. A random- 1994; 106: 1284-1290.
ized trial in primary biliary cirrhosis comparing ursodeoxycholic [122] Heathcote EJ, Cauch-Dudek K, Walker V, et al. The Canadian
acid in daily doses of either 10 mg/kg or 20 mg/kg. Dutch Multi- Multicenter Double-blind Randomized Controlled Trial of ursode-
centre PBC Study Group. Aliment Pharmacol Ther 1998; 12: 965- oxycholic acid in primary biliary cirrhosis. Hepatology 1994; 19:
971. 1149-1156.
[101] Harnois DM, Angulo P, Jorgensen RA, Larusso NF, Lindor KD. [123] Combes B, Luketic VA, Peters MG, et al. Prolonged follow-up of
High-dose ursodeoxycholic acid as a therapy for patients with pri- patients in the U.S. multicenter trial of ursodeoxycholic acid for
mary sclerosing cholangitis. Am J Gastroenterol 2001; 96: 1558- primary biliary cirrhosis. Am J Gastroenterol 2004; 99: 264-268.
1562. [124] Combes B, Carithers RL, Jr., Maddrey WC, et al. A randomized,
[102] Mitchell SA, Bansi DS, Hunt N, et al. A preliminary trial of high- double-blind, placebo-controlled trial of ursodeoxycholic acid in
dose ursodeoxycholic acid in primary sclerosing cholangitis. Gas- primary biliary cirrhosis. Hepatology 1995; 22: 759-766.
troenterology 2001; 121: 900-907. [125] Corpechot C, Carrat F, Bonnand AM, Poupon RE, Poupon R. The
[103] Parquet M, Metman EH, Raizman A, et al. Bioavailability, gastro- effect of ursodeoxycholic acid therapy on liver fibrosis progression
intestinal transit, solubilization and faecal excretion of ursodeoxy- in primary biliary cirrhosis. Hepatology 2000; 32: 1196-1199.
cholic acid in man. Eur J Clin Invest 1985; 15: 171-178. [126] Angulo P, Batts KP, Therneau TM, et al. Long-term ursodeoxy-
[104] Roda A, Fini A. Effect of nuclear hydroxy substituents on aqueous cholic acid delays histological progression in primary biliary cir-
solubility and acidic strength of bile acids. Hepatology 1984; 4: rhosis. Hepatology 1999; 29: 644-647.
72S-76S. [127] Jorgensen R, Angulo P, Dickson ER, Lindor KD. Results of long-
[105] Igimi H, Carey MC. pH-Solubility relations of chenodeoxycholic term ursodiol treatment for patients with primary biliary cirrhosis.
and ursodeoxycholic acids: physical-chemical basis for dissimilar Am J Gastroenterol 2002; 97: 2647-2650.
solution and membrane phenomena. J Lipid Res 1980; 21: 72-90. [128] van Hoogstraten HJ, Hansen BE, van Buuren HR, et al. Prognostic
[106] Roda A, Hofmann AF, Mysels KJ. The influence of bile salt struc- factors and long-term effects of ursodeoxycholic acid on liver bio-
ture on self-association in aqueous solutions. J Biol Chem 1983; chemical parameters in patients with primary biliary cirrhosis.
258: 6362-6370. Dutch Multi-Centre PBC Study Group. J Hepatol 1999; 31: 256-
[107] Williams CN, Al-Knawy B, Blanchard W. Bioavailability of four 262.
ursodeoxycholic acid preparations. Aliment Pharmacol Ther 2000; [129] Siegel JL, Jorgensen R, Angulo P, Lindor KD. Treatment with
14: 1133-1139. ursodeoxycholic acid is associated with weight gain in patients
[108] Roda A, Roda E, Marchi E, et al. Improved intestinal absorption of with primary biliary cirrhosis. J Clin Gastroenterol 2003; 37: 183-
an enteric-coated sodium ursodeoxycholate formulation. Pharm 185.
Res 1994; 11: 642-647. [130] Poupon RE, Poupon R, Balkau B. Ursodiol for the long-term
[109] Panini R, Vandelli MA, Forni F, Pradelli JM, Salvioli G. Improve- treatment of primary biliary cirrhosis. The UDCA-PBC Study
ment of ursodeoxycholic acid bioavailability by 2-hydroxypropyl- Group. N Engl J Med 1994; 330: 1342-1347.
beta-cyclodextrin complexation in healthy volunteers. Pharmacol [131] Combes B, Markin RS, Wheeler DE, et al. The effect of ursode-
Res 1995; 31: 205-209. oxycholic acid on the florid duct lesion of primary biliary cirrhosis.
[110] Ventura P, Panini R, Montosi G, et al. Ursodeoxycholic acid com- Hepatology 1999; 30: 602-605.
plexation with 2-hydroxypropyl-beta-cyclodextrin increases ur- [132] Degott C, Zafrani ES, Callard P, et al. Histopathological study of
sodeoxycholic acid biliary excretion after single oral administration primary biliary cirrhosis and the effect of ursodeoxycholic acid
in rats. Pharmacology 2001; 62: 107-112. treatment on histology progression. Hepatology 1999; 29: 1007-
[111] Vandelli M, Salvioli G, Mucci A, Panini R, Malmusi L, Forni F. 2- 1012.
hydroxypropyl--cyclodextrin complexation with ursodeoxycholic [133] Poupon RE, Lindor KD, Pares A, et al. Combined analysis of the
acid. Int J Pharm 1995; 188: 77-83. effect of treatment with ursodeoxycholic acid on histologic pro-
[112] Higginbottom S, Mallinson CB, Burns SJ, Attwood D, Barnwell gression in primary biliary cirrhosis. J Hepatol 2003; 39: 12-16.
SG. Ursodeoxycholic acid: effects of formulation on in vitro disso- [134] Poupon RE, Lindor KD, Cauch-Dudek K, et al. Combined analysis
lution. Int J Pharm 1994; 109: 173. of randomized controlled trials of ursodeoxycholic acid in primary
[113] Simoni P, Cerre C, Cipolla A, et al. Bioavailability study of a new, biliary cirrhosis. Gastroenterology 1997; 113: 884-890.
sinking, enteric-coated ursodeoxycholic acid formulation. Pharma- [135] Goulis J, Leandro G, Burroughs AK. Randomised controlled trials
col Res 1995; 31: 115-119. of ursodeoxycholic-acid therapy for primary biliary cirrhosis: a
[114] Carli F, Brambilla A, Di Rella M, Frattini C, Brandt A. [Character- meta-analysis. Lancet 1999; 354: 1053-1060.
istics of a new controlled release formulation of ursodesoxycholic [136] Gluud C, Christensen E. Ursodeoxycholic acid for primary biliary
acid]. Boll Chim Farm 1996; 135: 12-14. cirrhosis. Cochrane Database Syst Rev 2002: CD000551.
[115] Sama C, Morselli Labate AM, Chiodarelli C, Roda A, Briganti M, [137] Kaplan MM, Gershwin ME. Primary biliary cirrhosis. N Engl J
Roda E. Bioavailability of a delayed-release preparation of ursode- Med 2005; 353: 1261-1273.
oxycholic acid in man. Curr Therap Res 1983; 33: 558. [138] Angulo P, Dickson ER, Therneau TM, et al. Comparison of three
[116] Scalia S, Scagliarini R, Pazzi P. Evaluation of ursodeoxycholic acid doses of ursodeoxycholic acid in the treatment of primary biliary
bioavailability from immediate- and sustained-release preparations us- cirrhosis: a randomized trial. J Hepatol 1999; 30: 830-835.
ing gas chromatography-mass spectrometry and high-performance liq- [139] Angulo P, Jorgensen RA, Lindor KD. Incomplete response to ur-
uid chromatography. Arzneimittelforschung 2000; 50: 129-134. sodeoxycholic acid in primary biliary cirrhosis: is a double dosage
worthwhile? Am J Gastroenterol 2001; 96: 3152-3157.
174 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

[140] Verma A, Jazrawi RP, Ahmed HA, et al. Optimum dose of ursode- [161] Ohmoto K, Mitsui Y, Yamamoto S. Effect of bezafibrate in pri-
oxycholic acid in primary biliary cirrhosis. Eur J Gastroenterol He- mary biliary cirrhosis: a pilot study. Liver 2001; 21: 223-224.
patol 1999; 11: 1069-1076. [162] Vuoristo M, Farkkila M, Karvonen AL, et al. A placebo-controlled
[141] Papatheodoridis GV, Hadziyannis ES, Deutsch M, Hadziyannis SJ. trial of primary biliary cirrhosis treatment with colchicine and ur-
Ursodeoxycholic acid for primary biliary cirrhosis: final results of a sodeoxycholic acid. Gastroenterology 1995; 108: 1470-1478.
12-year, prospective, randomized, controlled trial. Am J Gastroen- [163] Poupon RE, Huet PM, Poupon R, et al. A randomized trial compar-
terol 2002; 97: 2063-2070. ing colchicine and ursodeoxycholic acid combination to ursode-
[142] ter Borg PC, Schalm SW, Hansen BE, van Buuren HR. Prognosis oxycholic acid in primary biliary cirrhosis. UDCA-PBC Study
of ursodeoxycholic Acid-treated patients with primary biliary cir- Group. Hepatology 1996; 24: 1098-1103.
rhosis. Results of a 10-yr cohort study involving 297 patients. Am J [164] Gong Y, Gluud C. Colchicine for primary biliary cirrhosis: a Coch-
Gastroenterol 2006; 101: 2044-2050. rane Hepato-Biliary Group systematic review of randomized clini-
[143] Shi J, Wu C, Lin Y, et al. Long-term effects of mid-dose ursode- cal trials. Am J Gastroenterol 2005; 100: 1876-1885.
oxycholic acid in primary biliary cirrhosis: a meta-analysis of ran- [165] Battezzati PM, Zuin M, Crosignani A, et al. Ten-year combination
domized controlled trials. Am J Gastroenterol 2006; 101: 1529- treatment with colchicine and ursodeoxycholic acid for primary bil-
1538. iary cirrhosis: a double-blind, placebo-controlled trial on sympto-
[144] Pares A, Caballeria L, Rodes J. Excellent long-term survival in matic patients. Aliment Pharmacol Ther 2001; 15: 1427-1434.
patients with primary biliary cirrhosis and biochemical response to [166] Combes B, Emerson SS, Flye NL, et al. Methotrexate (MTX) plus
ursodeoxycholic acid. Gastroenterology 2006; 130: 715-720. ursodeoxycholic acid (UDCA) in the treatment of primary biliary
[145] Poupon RE, Bonnand AM, Chretien Y, Poupon R. Ten-year sur- cirrhosis. Hepatology 2005; 42: 1184-1193.
vival in ursodeoxycholic acid-treated patients with primary biliary [167] Kaplan MM, Cheng S, Price LL, Bonis PA. A randomized con-
cirrhosis. The UDCA-PBC Study Group. Hepatology 1999; 29: trolled trial of colchicine plus ursodiol versus methotrexate plus ur-
1668-1671. sodiol in primary biliary cirrhosis: ten-year results. Hepatology
[146] Leuschner M, Guldutuna S, You T, et al. Ursodeoxycholic acid and 2004; 39: 915-923.
prednisolone versus ursodeoxycholic acid and placebo in the treat- [168] Wolfhagen FH, van Hoogstraten HJ, van Buuren HR, et al. Triple
ment of early stages of primary biliary cirrhosis. J Hepatol 1996; therapy with ursodeoxycholic acid, prednisone and azathioprine in
25: 49-57. primary biliary cirrhosis: a 1-year randomized, placebo-controlled
[147] Wolfhagen FH, van Buuren HR, Schalm SW. Combined treatment study. J Hepatol 1998; 29: 736-742.
with ursodeoxycholic acid and prednisone in primary biliary cir- [169] Davidson KM. Intrahepatic cholestasis of pregnancy. Semin Peri-
rhosis. Neth J Med 1994; 44: 84-90. natol 1998; 22: 104-111.
[148] Mitchison HC, Bassendine MF, Malcolm AJ, et al. A pilot, double- [170] Mazzella G, Rizzo N, Salzetta A, et al. Management of intrahepatic
blind, controlled 1-year trial of prednisolone treatment in primary cholestasis in pregnancy. Lancet 1991; 338: 1594-1595.
biliary cirrhosis: hepatic improvement but greater bone loss. Hepa- [171] Floreani A, Paternoster D, Grella V, et al. Ursodeoxycholic acid in
tology 1989; 10: 420-429. intrahepatic cholestasis of pregnancy. Br J Obstet Gynaecol 1994;
[149] Mazzella G, Fusaroli P, Pezzoli A, et al. Methylprednisolone ad- 101: 64-65.
ministration in primary biliary cirrhosis increases cholic acid turn- [172] Berkane N, Cocheton JJ, Brehier D, et al. Ursodeoxycholic acid in
over, synthesis, and deoxycholate concentration in bile. Dig Dis Sci intrahepatic cholestasis of pregnancy. A retrospective study of 19
1999; 44: 2478-2483. cases. Acta Obstet Gynecol Scand 2000; 79: 941-946.
[150] Ryrfeldt A, Andersson P, Edsbacker S, et al. Pharmacokinetics and [173] Brites D, Rodrigues CM, Oliveira N, Cardoso M, Graca LM. Cor-
metabolism of budesonide, a selective glucocorticoid. Eur J Respir rection of maternal serum bile acid profile during ursodeoxycholic
Dis Suppl 1982; 122: 86-95. acid therapy in cholestasis of pregnancy. J Hepatol 1998; 28: 91-
[151] Greenberg GR, Feagan BG, Martin F, et al. Oral budesonide for 98.
active Crohn's disease. Canadian Inflammatory Bowel Disease [174] Laifer SA, Stiller RJ, Siddiqui DS, Dunston-Boone G, Whetham
Study Group. N Engl J Med 1994; 331: 836-841. JC. Ursodeoxycholic acid for the treatment of intrahepatic cho-
[152] Angulo P, Jorgensen RA, Keach JC, et al. Oral budesonide in the lestasis of pregnancy. J Matern Fetal Med 2001; 10: 131-135.
treatment of patients with primary biliary cirrhosis with a subopti- [175] Liu Y, Qiao F, Liu H, Liu D. Ursodeoxycholic acid in the treatment
mal response to ursodeoxycholic acid. Hepatology 2000; 31: 318- of intraheptic cholestasis of pregnancy. J Huazhong Univ Sci
323. Technolog Med Sci 2006; 26: 350-352.
[153] Leuschner M, Maier KP, Schlichting J, et al. Oral budesonide and [176] Palma J, Reyes H, Ribalta J, et al. Effects of ursodeoxycholic acid
ursodeoxycholic acid for treatment of primary biliary cirrhosis: re- in patients with intrahepatic cholestasis of pregnancy. Hepatology
sults of a prospective double-blind trial. Gastroenterology 1999; 1992; 15: 1043-1047.
117: 918-925. [177] Diaferia A, Nicastri PL, Tartagni M, et al. Ursodeoxycholic acid
[154] Rautiainen H, Karkkainen P, Karvonen AL, et al. Budesonide therapy in pregnant women with cholestasis. Int J Gynaecol Obstet
combined with UDCA to improve liver histology in primary biliary 1996; 52: 133-140.
cirrhosis: a three-year randomized trial. Hepatology 2005; 41: 747- [178] Palma J, Reyes H, Ribalta J, et al. Ursodeoxycholic acid in the
752. treatment of cholestasis of pregnancy: a randomized, double-blind
[155] Hempfling W, Grunhage F, Dilger K, et al. Pharmacokinetics and study controlled with placebo. J Hepatol 1997; 27: 1022-1028.
pharmacodynamic action of budesonide in early- and late-stage [179] Mazzella G, Rizzo N, Azzaroli F, et al. Ursodeoxycholic acid ad-
primary biliary cirrhosis. Hepatology 2003; 38: 196-202. ministration in patients with cholestasis of pregnancy: effects on
[156] Nakai S, Masaki T, Kurokohchi K, Deguchi A, Nishioka M. Com- primary bile acids in babies and mothers. Hepatology 2001; 33:
bination therapy of bezafibrate and ursodeoxycholic acid in pri- 504-508.
mary biliary cirrhosis: a preliminary study. Am J Gastroenterol [180] Zapata R, Sandoval L, Palma J, et al. Ursodeoxycholic acid in the
2000; 95: 326-327. treatment of intrahepatic cholestasis of pregnancy. A 12-year expe-
[157] Akbar SM, Furukawa S, Nakanishi S, et al. Therapeutic efficacy of rience. Liver Int 2005; 25: 548-554.
decreased nitrite production by bezafibrate in patients with primary [181] Kondrackiene J, Beuers U, Kupcinskas L. Efficacy and safety of
biliary cirrhosis. J Gastroenterol 2005; 40: 157-163. ursodeoxycholic acid versus cholestyramine in intrahepatic cho-
[158] Itakura J, Izumi N, Nishimura Y, et al. Prospective randomized lestasis of pregnancy. Gastroenterology 2005; 129: 894-901.
crossover trial of combination therapy with bezafibrate and UDCA [182] Glantz A, Marschall HU, Lammert F, Mattsson LA. Intrahepatic
for primary biliary cirrhosis. Hepatol Res 2004; 29: 216-222. cholestasis of pregnancy: a randomized controlled trial comparing
[159] Kanda T, Yokosuka O, Imazeki F, Saisho H. Bezafibrate treatment: dexamethasone and ursodeoxycholic acid. Hepatology 2005; 42:
a new medical approach for PBC patients? J Gastroenterol 2003; 1399-1405.
38: 573-578. [183] Binder T, Salaj P, Zima T, Vitek L. Randomized prospective com-
[160] Kurihara T, Niimi A, Maeda A, Shigemoto M, Yamashita K. parative study of ursodeoxycholic acid and S-adenosyl-L-
Bezafibrate in the treatment of primary biliary cirrhosis: compari- methionine in the treatment of intrahepatic cholestasis of preg-
son with ursodeoxycholic acid. Am J Gastroenterol 2000; 95: 2990- nancy. J Perinat Med 2006; 34: 383-391.
2992.
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 175

[184] Nicastri PL, Diaferia A, Tartagni M, Loizzi P, Fanelli M. A ran- ulcerative colitis: prevalence, titre, and IgG subclass. Gut 1996; 38:
domised placebo-controlled trial of ursodeoxycholic acid and S- 384-389.
adenosylmethionine in the treatment of intrahepatic cholestasis of [207] Boberg KM, Aadland E, Jahnsen J, et al. Incidence and prevalence
pregnancy. Br J Obstet Gynaecol 1998; 105: 1205-1207. of primary biliary cirrhosis, primary sclerosing cholangitis, and
[185] Heikkinen J, Maentausta O, Ylostalo P, Janne O. Serum bile acid autoimmune hepatitis in a Norwegian population. Scand J Gastro-
levels in intrahepatic cholestasis of pregnancy during treatment enterol 1998; 33: 99-103.
with phenobarbital or cholestyramine. Eur J Obstet Gynecol Re- [208] Bergquist A, Ekbom A, Olsson R, et al. Hepatic and extrahepatic
prod Biol 1982; 14: 153-162. malignancies in primary sclerosing cholangitis. J Hepatol 2002; 36:
[186] Sadler LC, Lane M, North R. Severe fetal intracranial haemorrhage 321-327.
during treatment with cholestyramine for intrahepatic cholestasis of [209] Burak K, Angulo P, Pasha TM, et al. Incidence and risk factors for
pregnancy. Br J Obstet Gynaecol 1995; 102: 169-170. cholangiocarcinoma in primary sclerosing cholangitis. Am J Gas-
[187] Hirvioja ML, Tuimala R, Vuori J. The treatment of intrahepatic troenterol 2004; 99: 523-526.
cholestasis of pregnancy by dexamethasone. Br J Obstet Gynaecol [210] Broome U, Lofberg R, Veress B, Eriksson LS. Primary sclerosing
1992; 99: 109-111. cholangitis and ulcerative colitis: evidence for increased neoplastic
[188] Ribalta J, Reyes H, Gonzalez MC, et al. S-adenosyl-L-methionine potential. Hepatology 1995; 22: 1404-1408.
in the treatment of patients with intrahepatic cholestasis of preg- [211] Marchesa P, Lashner BA, Lavery IC, et al. The risk of cancer and
nancy: a randomized, double-blind, placebo-controlled study with dysplasia among ulcerative colitis patients with primary sclerosing
negative results. Hepatology 1991; 13: 1084-1089. cholangitis. Am J Gastroenterol 1997; 92: 1285-1288.
[189] Brites D, Rodrigues CM. Elevated levels of bile acids in colostrum [212] O'Brien CB, Senior JR, Arora-Mirchandani R, Batta AK, Salen G.
of patients with cholestasis of pregnancy are decreased following Ursodeoxycholic acid for the treatment of primary sclerosing cho-
ursodeoxycholic acid therapy. J Hepatol 1998; 29: 743-751. langitis: a 30-month pilot study. Hepatology 1991; 14: 838-847.
[190] Zimber A, Zusman I, Bentor R, Pinus H. Effects of lithocholic acid [213] Beuers U, Spengler U, Kruis W, et al. Ursodeoxycholic acid for
exposure throughout pregnancy on late prenatal and early postnatal treatment of primary sclerosing cholangitis: a placebo-controlled
development in rats. Teratology 1991; 43: 355-361. trial. Hepatology 1992; 16: 707-714.
[191] Altshuler G, Arizawa M, Molnar-Nadasdy G. Meconium-induced [214] Stiehl A, Walker S, Stiehl L, et al. Effect of ursodeoxycholic acid
umbilical cord vascular necrosis and ulceration: a potential link be- on liver and bile duct disease in primary sclerosing cholangitis. A
tween the placenta and poor pregnancy outcome. Obstet Gynecol 3-year pilot study with a placebo-controlled study period. J Hepatol
1992; 79: 760-766. 1994; 20: 57-64.
[192] Marin JJ, Bravo P, el-Mir MY, Serrano MA. ATP-dependent bile [215] Lindor KD. Ursodiol for primary sclerosing cholangitis. Mayo
acid transport across microvillous membrane of human term tro- Primary Sclerosing Cholangitis-Ursodeoxycholic Acid Study
phoblast. Am J Physiol 1995; 268: G685-694. Group. N Engl J Med 1997; 336: 691-695.
[193] Marin JJ, Serrano MA, el-Mir MY, Eleno N, Boyd CA. Bile acid [216] Chen W, Gluud C. Bile acids for primary sclerosing cholangitis.
transport by basal membrane vesicles of human term placental tro- Cochrane Database Syst Rev 2003: CD003626.
phoblast. Gastroenterology 1990; 99: 1431-1438. [217] Olsson R, Boberg KM, de Muckadell OS, et al. High-dose ursodeoxy-
[194] Serrano MA, Brites D, Larena MG, et al. Beneficial effect of ur- cholic acid in primary sclerosing cholangitis: a 5-year multicenter,
sodeoxycholic acid on alterations induced by cholestasis of preg- randomized, controlled study. Gastroenterology 2005; 129: 1464-
nancy in bile acid transport across the human placenta. J Hepatol 1472.
1998; 28: 829-839. [218] Stiehl A, Rudolph G, Kloters-Plachky P, Sauer P, Walker S. De-
[195] Macias RI, Pascual MJ, Bravo A, et al. Effect of maternal cholesta- velopment of dominant bile duct stenoses in patients with primary
sis on bile acid transfer across the rat placenta-maternal liver tan- sclerosing cholangitis treated with ursodeoxycholic acid: outcome
dem. Hepatology 2000; 31: 975-983. after endoscopic treatment. J Hepatol 2002; 36: 151-156.
[196] St-Pierre MV, Serrano MA, Macias RI, et al. Expression of mem- [219] Earnest DL, Holubec H, Wali RK, et al. Chemoprevention of
bers of the multidrug resistance protein family in human term pla- azoxymethane-induced colonic carcinogenesis by supplemental
centa. Am J Physiol Regul Integr Comp Physiol 2000; 279: R1495- dietary ursodeoxycholic acid. Cancer Res 1994; 54: 5071-5074.
1503. [220] Ikegami T, Matsuzaki Y, Shoda J, et al. The chemopreventive role
[197] Serrano MA, Macias RI, Vallejo M, et al. Effect of ursodeoxy- of ursodeoxycholic acid in azoxymethane-treated rats: suppressive
cholic acid on the impairment induced by maternal cholestasis in effects on enhanced group II phospholipase A2 expression in colo-
the rat placenta-maternal liver tandem excretory pathway. J Phar- nic tissue. Cancer Lett 1998; 134: 129-139.
macol Exp Ther 2003; 305: 515-524. [221] Tung BY, Emond MJ, Haggitt RC, et al. Ursodiol use is associated
[198] Azzaroli F, Feletti V, Mazzeo C, et al. MRP2 is upregulated by with lower prevalence of colonic neoplasia in patients with ulcera-
UDCA in cholestasis of pregnancy. J Hepatol 2004; 40: 4. tive colitis and primary sclerosing cholangitis. Ann Intern Med
[199] Broome U, Olsson R, Loof L, et al. Natural history and prognostic 2001; 134: 89-95.
factors in 305 Swedish patients with primary sclerosing cholangitis. [222] Pardi DS, Loftus EV, Jr., Kremers WK, Keach J, Lindor KD. Ur-
Gut 1996; 38: 610-615. sodeoxycholic acid as a chemopreventive agent in patients with ul-
[200] LaRusso NF, Shneider BL, Black D, et al. Primary sclerosing cho- cerative colitis and primary sclerosing cholangitis. Gastroenterol-
langitis: summary of a workshop. Hepatology 2006; 44: 746-764. ogy 2003; 124: 889-893.
[201] Boberg KM, Rocca G, Egeland T, et al. Time-dependent Cox re- [223] Arya G, Balistreri WF. Pediatric liver disease in the United States:
gression model is superior in prediction of prognosis in primary epidemiology and impact. J Gastroenterol Hepatol 2002; 17: 521-
sclerosing cholangitis. Hepatology 2002; 35: 652-657. 525.
[202] Bergquist A, Lindberg G, Saarinen S, Broome U. Increased preva- [224] Balistreri WF, Bezerra JA, Jansen P, et al. Intrahepatic cholestasis:
lence of primary sclerosing cholangitis among first-degree rela- summary of an American Association for the Study of Liver Dis-
tives. J Hepatol 2005; 42: 252-256. eases single-topic conference. Hepatology 2005; 42: 222-235.
[203] Donaldson PT, Norris S. Evaluation of the role of MHC class II [225] Fiorucci S, Clerici C, Antonelli E, et al. Protective effects of 6-
alleles, haplotypes and selected amino acid sequences in primary ethyl chenodeoxycholic acid, a farnesoid X receptor ligand, in es-
sclerosing cholangitis. Autoimmunity 2002; 35: 555-564. trogen-induced cholestasis. J Pharmacol Exp Ther 2005; 313: 604-
[204] Grant AJ, Lalor PF, Hubscher SG, Briskin M, Adams DH. MAd- 612.
CAM-1 expressed in chronic inflammatory liver disease supports [226] Wagner M, Halilbasic E, Marschall HU, et al. CAR and PXR ago-
mucosal lymphocyte adhesion to hepatic endothelium (MAdCAM- nists stimulate hepatic bile acid and bilirubin detoxification and
1 in chronic inflammatory liver disease). Hepatology 2001; 33: elimination pathways in mice. Hepatology 2005; 42: 420-430.
1065-1072. [227] van Mil SW, van der Woerd WL, van der Brugge G, et al. Benign
[205] Cullen S, Chapman R. Primary sclerosing cholangitis. Autoimmun recurrent intrahepatic cholestasis type 2 is caused by mutations in
Rev 2003; 2: 305-312. ABCB11. Gastroenterology 2004; 127: 379-384.
[206] Bansi DS, Fleming KA, Chapman RW. Importance of antineutro- [228] Lucena JF, Herrero JI, Quiroga J, et al. A multidrug resistance 3
phil cytoplasmic antibodies in primary sclerosing cholangitis and gene mutation causing cholelithiasis, cholestasis of pregnancy, and
176 Current Clinical Pharmacology, 2007, Vol. 2, No. 2 Festi et al.

adulthood biliary cirrhosis. Gastroenterology 2003; 124: 1037- cretion and liver morphology in cystic fibrosis-associated liver dis-
1042. ease. Hepatology 1998; 27: 166-174.
[229] Rosmorduc O, Hermelin B, Boelle PY, et al. ABCB4 gene muta- [252] van de Meeberg PC, Houwen RH, Sinaasappel M, et al. Low-dose
tion-associated cholelithiasis in adults. Gastroenterology 2003; versus high-dose ursodeoxycholic acid in cystic fibrosis-related
125: 452-459. cholestatic liver disease. Results of a randomized study with 1-year
[230] Wanty C, Joomye R, Van Hoorebeek N, et al. Fifteen years single follow-up. Scand J Gastroenterol 1997; 32: 369-373.
center experience in the management of progressive familial intra- [253] Cheng K, Ashby D, Smyth R. Ursodeoxycholic acid for cystic
hepatic cholestasis of infancy. Acta Gastroenterol Belg 2004; 67: fibrosis-related liver disease. Cochrane Database Syst Rev 2000:
313-319. CD000222.
[231] Shneider BL. Progressive intrahepatic cholestasis: mechanisms, [254] Alagille D, Estrada A, Hadchouel M, et al. Syndromic paucity of
diagnosis and therapy. Pediatr Transplant 2004; 8: 609-612. interlobular bile ducts (Alagille syndrome or arteriohepatic dyspla-
[232] Egawa H, Yorifuji T, Sumazaki R, et al. Intractable diarrhea after sia): review of 80 cases. J Pediatr 1987; 110: 195-200.
liver transplantation for Byler's disease: successful treatment with [255] Li L, Krantz ID, Deng Y, et al. Alagille syndrome is caused by
bile adsorptive resin. Liver Transpl 2002; 8: 714-716. mutations in human Jagged1, which encodes a ligand for Notch1.
[233] Lykavieris P, Cresteil D, Fabre M, Hadchouel M, Bernard O, Nat Genet 1997; 16: 243-251.
Jachemin E. Catch-up growth failure, refractory diarrhea and ap- [256] Oda T, Elkahloun AG, Pike BL, et al. Mutations in the human
pearance of liver statosis in PFIC 1 patients after liver transplanta- Jagged1 gene are responsible for Alagille syndrome. Nat Genet
tion of biliary diversion. J Pediatr Gastroenterol Nutr 2002; 34: 1997; 16: 235-242.
434A. [257] Warthen DM, Moore EC, Kamath BM, et al. Jagged1 (JAG1)
[234] Balistrieri W, Setchell KDR, Ryckman FC. Bile acid therapy in mutations in Alagille syndrome: increasing the mutation detection
pediatric liver disease. In: Bile acids and hepatobiliary system- rate. Hum Mutat 2006; 27: 436-443.
from basic science to clinical practice. Paumgartner G, Stiehl, A, [258] McDaniell R, Warthen DM, Sanchez-Lara PA, et al. NOTCH2
Gerok, W (Editor) Kluwer academic Publishers; 1993; pp. 271- mutations cause Alagille syndrome, a heterogeneous disorder of the
283. notch signaling pathway. Am J Hum Genet 2006; 79: 169-173.
[235] Dinler G, Kocak N, Ozen H, Yuce A, Gurakan F. Ursodeoxycholic [259] Maldini G, Torri E, Lucianetti A, et al. Orthotopic liver transplan-
acid treatment in children with Byler disease. Pediatr Int 1999; 41: tation for alagille syndrome. Transplant Proc 2005; 37: 1174-1176.
662-665. [260] Kardoff R, Melter M, Rodeck B, Brodehl J. Long term ursode-
[236] Jacquemin E, Hermans D, Myara A, et al. Ursodeoxycholic acid oxicholic acid treatment of cholestatic liver disease in childhood-
therapy in pediatric patients with progressive familial intrahepatic clinical and biochemical effects. Klin Paediatr 1996; 208: 118-122.
cholestasis. Hepatology 1997; 25: 519-523. [261] Paradis K, El Arab N, Yousef I, et al. Use of ursodeoxicholic acid in
[237] Rowe SM, Miller S, Sorscher EJ. Cystic fibrosis. N Engl J Med children with cholestatic liver disease. Hepatology 1993; 18: 301A.
2005; 352: 1992-2001. [262] Balistreri WF, Grand R, Hoofnagle JH, et al. Biliary atresia: cur-
[238] Lindblad A, Glaumann H, Strandvik B. Natural history of liver rent concepts and research directions. Summary of a symposium.
disease in cystic fibrosis. Hepatology 1999; 30: 1151-1158. Hepatology 1996; 23: 1682-1692.
[239] Colombo C, Battezzati PM, Crosignani A, et al. Liver disease in [263] Sokol RJ, Dahl R, Devereaux MW, et al. Human hepatic mito-
cystic fibrosis: A prospective study on incidence, risk factors, and chondria generate reactive oxygen species and undergo the perme-
outcome. Hepatology 2002; 36: 1374-1382. ability transition in response to hydrophobic bile acids. J Pediatr
[240] FitzSimmons S. Cystic fibrosis Foundation, Patient Registry 1996 Gastroenterol Nutr 2005; 41: 235-243.
In: Annual data report Bethesda Maryland; 1997. [264] Lykavieris P, Chardot C, Sokhn M, et al. Outcome in adulthood of
[241] Kinnman N, Lindblad A, Housset C, et al. Expression of cystic biliary atresia: a study of 63 patients who survived for over 20
fibrosis transmembrane conductance regulator in liver tissue from years with their native liver. Hepatology 2005; 41: 366-371.
patients with cystic fibrosis. Hepatology 2000; 32: 334-340. [265] Barshes NR, Lee TC, Balkrishnan R, et al. Orthotopic liver trans-
[242] Lindblad A, Hultcrantz R, Strandvik B. Bile-duct destruction and plantation for biliary atresia: the U.S. experience. Liver Transpl
collagen deposition: a prominent ultrastructural feature of the liver 2005; 11: 1193-1200.
in cystic fibrosis. Hepatology 1992; 16: 372-381. [266] Nittono H, Tokita A, Hayashi M, et al. Ursodeoxycholic acid ther-
[243] Debray D, Lykavieris P, Gauthier F, et al. Outcome of cystic fibro- apy in the treatment of biliary atresia. Biomed Pharmacother 1989;
sis-associated liver cirrhosis: management of portal hypertension. J 43: 37-41.
Hepatol 1999; 31: 77-83. [267] Balistreri WF. Bile acid therapy in pediatric hepatobiliary disease:
[244] Colombo C, Russo MC, Zazzeron L, Romano G. Liver disease in the role of ursodeoxycholic acid. J Pediatr Gastroenterol Nutr 1997;
cystic fibrosis. J Pediatr Gastroenterol Nutr 2006; 43 (Suppl 1) : S49- 24: 573-589.
55. [268] Meyers RL, Book LS, O'Gorman MA, et al. High-dose steroids,
[245] Chinet T, Fouassier L, Dray-Charier N, et al. Regulation of elec- ursodeoxycholic acid, and chronic intravenous antibiotics improve
trogenic anion secretion in normal and cystic fibrosis gallbladder bile flow after Kasai procedure in infants with biliary atresia. J Pe-
mucosa. Hepatology 1999; 29: 5-13. diatr Surg 2003; 38: 406-411.
[246] Nathanson MH, Burgstahler AD, Masyuk A, Larusso NF. Stimula- [269] Arat M, Idilman R, Soydan EA, et al. Ursodeoxycholic acid treat-
tion of ATP secretion in the liver by therapeutic bile acids. Bio- ment in isolated chronic graft-vs.-host disease of the liver. Clin
chem J 2001; 358: 1-5. Transplant 2005; 19: 798-803.
[247] Colombo C, Castellani MR, Balistreri WF, et al. Scintigraphic docu- [270] Chiba T, Yokosuka O, Kanda T, et al. Hepatic graft-versus-host
mentation of an improvement in hepatobiliary excretory function after disease resembling acute hepatitis: additional treatment with ur-
treatment with ursodeoxycholic acid in patients with cystic fibrosis and sodeoxycholic acid. Liver 2002; 22: 514-517.
associated liver disease. Hepatology 1992; 15: 677-684. [271] Bahrle S, Szabo G, Stiehl A, et al. Adjuvant treatment with ursode-
[248] Colombo C, Crosignani A, Assaisso M, et al. Ursodeoxycholic acid oxycholic acid may reduce the incidence of acute cardiac allograft
therapy in cystic fibrosis-associated liver disease: a dose-response rejection. J Heart Lung Transplant 1998; 17: 592-598.
study. Hepatology 1992; 16: 924-930. [272] Barnes D, Talenti D, Cammell G, et al. A randomized clinical trial
[249] Cotting J, Lentze MJ, Reichen J. Effects of ursodeoxycholic acid of ursodeoxycholic acid as adjuvant treatment to prevent liver
treatment on nutrition and liver function in patients with cystic fi- transplant rejection. Hepatology 1997; 26: 853-857.
brosis and longstanding cholestasis. Gut 1990; 31: 918-921. [273] Ozkaya M, Erten A, Sahin I, et al. The effect of ursodeoxycholic
[250] Colombo C, Battezzati PM, Podda M, Bettinardi N, Giunta A. acid treatment on epidermal growth factor in patients with bile re-
Ursodeoxycholic acid for liver disease associated with cystic fibro- flux gastritis. Turk J Gastroenterol 2002; 13: 198-202.
sis: a double-blind multicenter trial. The Italian Group for the [274] Stefaniwsky AB, Tint GS, Speck J, Shefer S, Salen G. Ursodeoxy-
Study of Ursodeoxycholic Acid in Cystic Fibrosis. Hepatology cholic acid treatment of bile reflux gastritis. Gastroenterology
1996; 23: 1484-1490. 1985; 89: 1000-1004.
[251] Lindblad A, Glaumann H, Strandvik B. A two-year prospective [275] Liu F, Cheng Y, Wu J, Tauschel HD, Duan RD. Ursodeoxycholic
study of the effect of ursodeoxycholic acid on urinary bile acid ex- acid differentially affects three types of sphingomyelinase in hu-
man colon cancer Caco 2 cells. Cancer Lett 2006; 235: 141-146.
Clinical Efficacy and Effectiveness of Ursodeoxycholic Acid Current Clinical Pharmacology, 2007, Vol. 2, No. 2 177

[276] Cheng Y, Tauschel HD, Nilsson A, Duan RD. Ursodeoxycholic acid [279] Alberts DS, Martinez ME, Hess LM, et al. Phase III trial of ur-
increases the activities of alkaline sphingomyelinase and caspase-3 in sodeoxycholic acid to prevent colorectal adenoma recurrence. J
the rat colon. Scand J Gastroenterol 1999; 34: 915-920. Natl Cancer Inst 2005; 97: 846-853.
[277] Duan RD, Cheng Y, Tauschel HD, Nilsson A. Effects of ursode- [280] Hofmann AF, Zakko SF, Lira M, et al. Novel biotransformation
oxycholate and other bile salts on levels of rat intestinal alkaline and physiological properties of norursodeoxycholic acid in humans.
sphingomyelinase: a potential implication in tumorigenesis. Dig Hepatology 2005; 42: 1391-1398.
Dis Sci 1998; 43: 26-32. [281] Fickert P, Wagner M, Marschall HU, et al. 24-norUrsodeoxycholic
[278] Serfaty L, De Leusse A, Rosmorduc O, et al. Ursodeoxycholic acid acid is superior to ursodeoxycholic acid in the treatment of scleros-
therapy and the risk of colorectal adenoma in patients with primary bil- ing cholangitis in Mdr2 (Abcb4) knockout mice. Gastroenterology
iary cirrhosis: an observational study. Hepatology 2003; 38: 203-209. 2006; 130: 465-481.

Received: 05 January, 2007 Revised: 23 January, 2007 Accepted: 22 February, 2007

You might also like