Page last updated: 2024-09-21

chenodeoxycholic acid

Description

Chenodeoxycholic Acid: A bile acid, usually conjugated with either glycine or taurine. It acts as a detergent to solubilize fats for intestinal absorption and is reabsorbed by the small intestine. It is used as cholagogue, a choleretic laxative, and to prevent or dissolve gallstones. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

chenodeoxycholic acid : A dihydroxy-5beta-cholanic acid that is (5beta)-cholan-24-oic acid substituted by hydroxy groups at positions 3 and 7 respectively. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

chenodeoxycholate : Conjugate base of chenodeoxycholic acid; major species at pH 7.3. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

FloraRankFlora DefinitionFamilyFamily Definition
GlycinegenusA non-essential amino acid. It is found primarily in gelatin and silk fibroin and used therapeutically as a nutrient. It is also a fast inhibitory neurotransmitter.[MeSH]FabaceaeThe large family of plants characterized by pods. Some are edible and some cause LATHYRISM or FAVISM and other forms of poisoning. Other species yield useful materials like gums from ACACIA and various LECTINS like PHYTOHEMAGGLUTININS from PHASEOLUS. Many of them harbor NITROGEN FIXATION bacteria on their roots. Many but not all species of beans belong to this family.[MeSH]

Cross-References

ID SourceID
PubMed CID10133
CHEMBL ID240597
CHEBI ID16755
SCHEMBL ID25055
MeSH IDM0004029

Synonyms (193)

Synonym
BIDD:PXR0056
smr000857194
MLS002154253
HY-76847
AB00513822-06
leadiant (formerly chenodeoxycholic acid sigma-tau)
gtpl608
CHEBI:16755 ,
anthropodesoxycholic acid
cdca
chenix
3alpha,7alpha-dihydroxy-5beta-cholan-24-oic acid
7alpha-hydroxylithocholic acid
chenorm
xenbilox
chenofalk
(3alpha,5beta,7alpha,8xi)-3,7-dihydroxycholan-24-oic acid
5-beta-cholan-24-oic acid, 3-alpha,7-alpha-dihydroxy-
henohol
3-alpha,7-alpha-dihydroxy-5-beta-cholan-24-oic acid
chendeoxycholic acid
(3beta,7beta,8xi,9xi,14xi,17alpha)-3,7-dihydroxycholan-24-oic acid
cholan-24-oic acid, 3,7-dihydroxy-, (3alpha,5beta,7alpha)-
acido chenodeoxicholico [inn-spanish]
nsc 657949
chenodiol [usan]
chenodesoxycholsaeure [german]
cholan-24-oic acid, 3,7-dihydroxy-, (3-alpha,5-beta,7-alpha)-
chenique acid
einecs 207-481-8
acide chenodeoxycholique [inn-french]
7-alpha-hydroxylithocholic acid
ccris 2195
acidum chenodeoxycholicum [inn-latin]
3-alpha,7-alpha-dihydroxycholanic acid
3-alpha,7-alpha-dihydroxycholansaeure [german]
nsc-657949
D00163
chenix (tn)
chenodeoxycholic acid (jp17/inn)
chenodiol (usan)
PRESTWICK2_000285
cas-474-25-9
NCGC00016387-02
SMP1_000064
PRESTWICK3_000285
BPBIO1_000210
BSPBIO_000190
AB00513822
bdbm21674
(4r)-4-[(1s,2s,5r,7s,9r,10r,11s,14r,15r)-5,9-dihydroxy-2,15-dimethyltetracyclo[8.7.0.0^{2,7}.0^{11,15}]heptadecan-14-yl]pentanoic acid
chenodeoxycholic acid
474-25-9
C02528
chenodiol
3alpha,7alpha-dihydroxy-5beta-cholanic acid
chenodeoxycholate
anthropodeoxycholic acid
(4r)-4-[(3r,5s,7r,8r,9s,10s,13r,14s,17r)-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1h-cyclopenta[a]phenanthren-17-yl]pentanoic acid
cholan-24-oic acid, 3,7-dihydroxy-, (3-.alpha., 5-.beta., 7-.alpha.)-
chenic acid
gallodesoxycholic acid
chenodesoxycholic acid
anthropododesoxycholic acid
chendol
chenodeoxycholic acid, >=97%
jn3 ,
LMST04010032
SPECTRUM5_002009
chenodeoxycholic acid, free acid
PRESTWICK0_000285
PRESTWICK1_000285
SPBIO_002409
HSCI1_000210
NCGC00142400-03
acid, chenique
acid, chenic
acid, gallodesoxycholic
acid, chenodeoxycholic
C-2900
0DBBBC66-0CFA-4DB9-97F4-5B1492756A02
cholanorm
nsc-757798
chenocol
fluibil
CHEMBL240597
chenossil
DB06777
chenocholic acid
HMS1568J12
NCGC00142400-04
HMS2095J12
ec 207-481-8
acide chenodeoxycholique
unii-0gei24lg0j
chenodeoxycholic acid [inn]
3-alpha,7-alpha-dihydroxycholansaeure
chenodesoxycholsaeure
acidum chenodeoxycholicum
0gei24lg0j ,
acido chenodeoxicholico
(4r)-4-[(1r,3as,3br,4r,5as,7r,9as,9bs,11ar)-4,7-dihydroxy-9a,11a-dimethyl-hexadecahydro-1h-cyclopenta[a]phenanthren-1-yl]pentanoic acid
EN300-75328
NCGC00258045-01
tox21_200491
A827222
3,7-dihydroxy-5-cholanicacid
5beta-cholanic acid-3alpha,7alpha-diol
cholan-24-oic acid, 3,7-dihydroxy-, (3-alpha,5-beta,7-alpha)- (9ci)
BMSE000908
chenodeoxycholic acid (jp16/inn)
dtxcid40260
tox21_110412
dtxsid2020260 ,
HMS2234I22
(4r)-4-((1s,2s,7s,11s,5r,9r,10r,14r,15r)-5,9-dihydroxy-2,15-dimethyltetracyclo [8.7.0.0<2,7>.0<11,15>]heptadec-14-yl)pentanoic acid
chenodal
chenodiol [orange book]
ursodeoxycholic acid impurity a [ep impurity]
cholan-24-oic acid, 3,7-dihydroxy-, (3.alpha.,5.beta.,7.alpha.)
chenodeoxycholic acid [who-dd]
chenodeoxycholic acid [jan]
chenodeoxycholic acid [ep impurity]
chenodiol [vandf]
chenodeoxycholic acid [mart.]
chenodiol [mi]
chenodeoxycholic acid [ep monograph]
CS-0834
CCG-220285
SCHEMBL25055
3.alpha.,7.alpha.-dihydroxy-5.beta.-cholanoic acid
chenocedon
cholan-24-oic acid, 3,7-dihydroxy-, (3.alpha.,5.beta.,7.alpha.)-
3,7-dihydroxycholan-24-oic acid, (3.alpha.,5.beta.,7.alpha.)- #
3.alpha.,7.alpha.-dihydroxy-5.beta.-cholan-24-oic acid
chendal
7.alpha.-hydroxylithocholic acid
3.alpha.,7.alpha.-dihydroxy-5.beta.-cholanic acid
hekbilin
chenodex
kebilis
5.beta.-cholan-24-oic acid, 3.alpha.,7.alpha.-dihydroxy-
3.alpha.,7.alpha.-dihydroxycholanic acid
3.alpha.,7.alpha.-dihydroxycholansaeure
dihydroxy-3.alpha.,7.alpha.(5.beta.)cholanic acid
3alpha, 7alpha,-dihydroxy-5beta-cholanic acid
3alpha, 7alpha-dihydroxy-5beta-cholanoic acid
AKOS024280614
cholan-24-oic acid, 3,7-dihydroxy-, (3a,5b,7a)-
(r)-4-((3r,5s,7r,8r,9s,10s,13r,14s,17r)-3,7-dihydroxy-10,13-dimethylhexadecahydro-1h-cyclopenta[a]phenanthren-17-yl)pentanoic acid
AB00513822_07
mfcd00064142
chenodeoxycholic acid, european pharmacopoeia (ep) reference standard
chenodeoxycholic acid, 500 mug/ml in methanol, certified reference material
(3alpha,5beta,7alpha)-3,7-dihydroxycholan-24-oic acid
(4r)-4-[(1s,2s,5r,7s,9r,10r,11s,14r,15r)-5,9-dihydroxy-2,15-dimethyltetracyclo[8.7.0.0?,?.0??,??]heptadecan-14-yl]pentanoic acid
HMS3712J12
(3a,5b,7a)-3,7-dihydroxy-cholan-24-oic acid
3a,7a-dihydroxy-5b-cholanic acid
3a,7a-dihydroxy-5b-cholan-24-oic acid
3a,7a-dihydroxy-5b-cholanate
(+)-chenodeoxycholate
(+)-chenodeoxycholic acid
3a,7a-dihydroxy-5b,14a,17b-cholanic acid
7a-hydroxy-desoxycholsaeure
(3a,5b,7a)-3,7-dihydroxy-cholan-24-oate
3a,7a-dihydroxy-5b-cholan-24-oate
3a,7a-dihydroxy-5b,14a,17b-cholanate
'(3alpha,5alpha,7beta,8alpha,17alpha)-3,7-dihydroxycholan-24-oic acid'
(3alpha,5alpha,7beta,8alpha,17alpha)-3,7-dihydroxycholan-24-oic acid
S1843
24404-86-2
AS-13636
Q419028
BRD-K18135438-001-16-7
chenodeoxycholic-acid
(4r)-4-[(3r,5s,7r,8r,9s,10s,13r,14s,17r)-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1h-cyclopenta[a]phenanthren-17-yl]pentanoicacid
NCGC00142400-08
acido chenodesossicolico
acidum chenodeoxycholicum (inn-latin)
acide chenodeoxycholique (inn-french)
a05aa01
3alpha,7alpha-dihydroxycholanic acid
chenodeoxycholic acid (ep monograph)
acido chenodeoxicholico (inn-spanish)
acido quenodeoxicolico
acide chenodesoxycholique
chanodasoxycholique (acide)
chenodeoxycholic acid (ep impurity)
chenodeoxycholic acid (mart.)
ursodeoxycholic acid impurity a (ep impurity)
cholan-24-oic acid, 3,7-dihydroxy-, (3alpha,5beta,7alpha)
Z1172235745

Research Excerpts

Overview

ExcerptReference
"Chenodeoxycholic acid (CDCA) is a natural germination inhibitor for C. "( Abel-Santos, E; Firestine, SM; Phan, J; Sharma, SK; Simon, MP; Yip, C, 2021)
"Chenodeoxycholic acid (CDCA) is a liver-formed detergent and plays an important role in the control of cholesterol homeostasis. "( Barbier, O; Bélanger, J; Caron, P; Grepper, S; Inaba, TT; Kaeding, J; Monté, D; Trottier, J; Verreault, M, 2006)
"Chenodeoxycholic acid (CDCA) is an effective treatment for dissolving gallstones but experimental studies have suggested that it might be hepatotoxic. "( Giampieri, MP; Jezequel, AM; Koch, MM; Lorenzini, I; Orlandi, F, 1980)
"Chenodeoxycholic acid (CDCA) is a potent suppressor of the cholesterol 7alpha-hydroxylase, the rate-determining enzyme in bile acid formation."( Axelson, M; Einarsson, C; Hillebrant, CG, 2001)
"Chenodeoxycholic acid is an important drug for the treatment of cholesterol cholelithiasis in man. "( Chen, T; Dyrszka, H; Mosbach, EH; Salen, G, 1975)

Effects

ExcerptReference
"Chenodeoxycholic acid has a broad influence on lipid metabolism, including reducing plasma clearance of LDL. "( Angelin, B; Eriksson, M; Ghosh Laskar, M; Rudling, M, 2017)
"Chenodeoxycholic acid has a broad influence on lipid metabolism, including reducing plasma clearance of LDL. "( Angelin, B; Eriksson, M; Ghosh Laskar, M; Rudling, M, 2017)
"Chenodeoxycholic acid has been administered to 50 patients with cholelithiasis and 8 with choledocholithiasis for a period of 15 months at doses varying between 2- and 15 mg/kg/day. "( Aldini, R; Barbara, L; Festi, D; Mazzella, G; Roda, A; Roda, E; Sama, C, 1976)

Treatment

ExcerptReference
"Chenodeoxycholic acid treatment increased plasma LDL cholesterol by ~10% as a result of reduced clearance of plasma LDL-apolipoprotein (apo)B; LDL production was somewhat reduced. "( Angelin, B; Eriksson, M; Ghosh Laskar, M; Rudling, M, 2017)
"Chenodeoxycholic acid (CDCA) treatment stabilized chimeric GFP-LDLR 3'-UTR mRNA and accompanied mitogen-activated protein kinase (MAPK) activation."( Inoue, J; Sato, R; Shimizu, M; Yashiro, T; Yokoi, Y, 2011)
"Chenodeoxycholic acid treatment was effective in improving liver function and decreasing unusual bile acids such as 7α-hydroxy- and 7α,12α-dihydroxy-3-oxo-4-cholen-24-oic acids in serum and urine."( Hasegawa, T; Hayashi, S; Homma, K; Hoshina, T; Ihara, K; Kimura, A; Kurosawa, T; Matsuishi, T; Mizuochi, T; Morimura, T; Nittono, H; Ohno, Y; Ohtake, A; Seki, Y; Takahashi, T; Takei, H, 2013)
"Chenodeoxycholic acid (CDCA) treatment specifically decreased both PCSK9 mRNA and protein contents."( Cariou, B; Caron, S; Costet, P; Kourimate, S; Krempf, M; Langhi, C; Le May, C; Staels, B, 2008)
"Chenodeoxycholic acid treatment resulted in a significant lowering of the serum triglycerides in both groups of patients."( Camarri, E; Marcolongo, R; Marini, G; Zaccherotti, L, 1978)
"Chenodeoxycholic acid treatment for periods ranging from 6 months to 3 years and 4 months lowered serum lathosterol (57.7% reduction) and campesterol (57.8%) levels in parallel with cholestanol (70.8%) level, but the sitosterol level (19.7%) decreased less."( Fujiyama, J; Kasama, T; Kuriyama, M; Osame, M, 1991)
"Chenodeoxycholic acid treatment for two separate periods of 10 and 6 months each increased nerve conduction velocity."( Donaghy, M; King, RH; McKeran, RO; Schwartz, MS; Thomas, PK, 1990)
"Chenodeoxycholic acid treatment lowered biliary cholesterol saturation in obese gallstone patients by reducing biliary cholesterol secretion."( Dowling, RH; Maton, PN; Murphy, GM; Reuben, A, 1985)
"Treatment with chenodeoxycholic acid (CDCA; chenodiol) is the current standard of care."( Salen, G; Steiner, RD, 2017)
"Treatment with chenodeoxycholic acid (CDCA) may slow the progression of the disease and reverse some symptoms in a proportion of patients."( Bartkiewicz, J; Biskup, S; Catarino, CB; Gallenmüller, C; Hörtnagel, K; Klopstock, T; Küpper, C; Seelos, K; Vollmar, C, 2018)
"Treatment with chenodeoxycholic acid normalizes cholestanol concentrations and abrogates progression of the disease."( Aaref, S; Berginer, VM; Falik-Zaccai, TC; Gross, B; Kfir, N; Morad, K; Morkos, S, 2009)
"Treatment with chenodeoxycholic acid resulted in a 40% reduction of HMG CoA reductase activity."( Ahlberg, J; Angelin, B; Einarsson, K, 1981)
"Treatment with chenodeoxycholic acid was tried on two occasions."( Casteels, M; Clayton, PT; Lawson, AM; Mieli-Vergani, G, 1995)
"Treatment with chenodeoxycholic acid (CDCA) 750 mg/day orally improved neurological and biochemical abnormalities."( Berginer, J; Berginer, VM; Korczyn, AD; Tadmor, R, 1994)
"Oral treatment with chenodeoxycholic acid causes dissolution of cholesterol gallstones in man. "( Hofmann, AF; LaRusso, NF; Northfield, TC; Thistle, JL, 1975)
"Treatment with chenodeoxycholic acid led to clinical and biochemical improvement in one patient."( Clayton, PT; Horslen, SP; Lawson, AM; Malone, M, 1992)
"Treatment with chenodeoxycholic acid resulted in marked clinical improvement and normalized liver function tests."( Baginski, ES; Clayton, PT; Egestad, B; Ichimiya, H; Nazer, H; Sjövall, J, 1991)
"Treatment with chenodeoxycholic acid reduced cerebrospinal fluid cholesterol by 34 percent and cholestanol threefold."( Berginer, V; Horak, E; Horak, I; Salen, G; Shefer, S; Shore, V; Tint, GS, 1987)
"Treatment with chenodeoxycholic acid, which suppresses abnormal bile acid synthesis in these patients, reduced plasma bile alcohol concentrations dramatically."( Batta, AK; Batta, M; Salen, G; Shefer, S; Tint, GS, 1987)
"Treatment with chenodeoxycholic acid, 15 mg per kg body weight per day for 3-4 weeks before surgery, decreased the mean level to 20 +/- 7 ng/ml (P greater than 0.05)."( Akerlund, JE; Angelin, B; Björkhem, I; Einarsson, K; Ewerth, S; Reihnér, E, 1987)
"Treatment with chenodeoxycholic acid (0.5-1.5 g/day by mouth) did not change serum cholesterol levels but did significantly reduce serum triglyceride concentrations from a pretreatment level of 118 (+/- S.E."( Bell, GD; Dowling, RH; Lewis, B; Petrie, A, 1973)

Roles (2)

RoleDescription
human metaboliteAny mammalian metabolite produced during a metabolic reaction in humans (Homo sapiens).
mouse metaboliteAny mammalian metabolite produced during a metabolic reaction in a mouse (Mus musculus).
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (3)

ClassDescription
bile acidAny member of a group of hydroxy-5beta-cholanic acids occuring in bile, where they are present as the sodium salts of their amides with glycine or taurine. In mammals bile acids almost invariably have 5beta-configuration.
dihydroxy-5beta-cholanic acidA hydroxy-5beta-cholanic acid carrying two hydroxy groups at unspecified positions.
C24-steroidA steroid compound with a structure based on a 24-carbon (cholane) skeleton.
[compound class information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Pathways (9)

chenodeoxycholic acid is involved in 9 pathway(s), involving a total of 157 unique proteins and 924 unique compounds

PathwayProteinsCompounds
Bile Acid Biosynthesis1761
Congenital Bile Acid Synthesis Defect Type II1761
Congenital Bile Acid Synthesis Defect Type III1761
Familial Hypercholanemia (FHCA)1761
Zellweger Syndrome1761
Cerebrotendinous Xanthomatosis (CTX)1761
27-Hydroxylase Deficiency1761
Biochemical pathways: part I0466
Oxysterols derived from cholesterol3831

Protein Targets (37)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
LuciferasePhotinus pyralis (common eastern firefly)Potency79.90070.007215.758889.3584AID1224835
RAR-related orphan receptor gammaMus musculus (house mouse)Potency57.98300.006038.004119,952.5996AID1159521; AID1159523
SMAD family member 2Homo sapiens (human)Potency43.77470.173734.304761.8120AID1346859
SMAD family member 3Homo sapiens (human)Potency43.77470.173734.304761.8120AID1346859
TDP1 proteinHomo sapiens (human)Potency15.68120.000811.382244.6684AID686978; AID686979
AR proteinHomo sapiens (human)Potency30.98610.000221.22318,912.5098AID743035; AID743040; AID743042; AID743054; AID743063
caspase 7, apoptosis-related cysteine proteaseHomo sapiens (human)Potency68.79890.013326.981070.7614AID1346978
nuclear receptor subfamily 1, group I, member 3Homo sapiens (human)Potency56.87160.001022.650876.6163AID1224838; AID1224839
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency33.78580.01237.983543.2770AID1645841
glucocorticoid receptor [Homo sapiens]Homo sapiens (human)Potency61.83340.000214.376460.0339AID720692
retinoic acid nuclear receptor alpha variant 1Homo sapiens (human)Potency19.39020.003041.611522,387.1992AID1159552; AID1159555
retinoid X nuclear receptor alphaHomo sapiens (human)Potency19.55340.000817.505159.3239AID1159527; AID1159531
estrogen-related nuclear receptor alphaHomo sapiens (human)Potency49.96900.001530.607315,848.9004AID1224841; AID1224842; AID1224848; AID1224849; AID1259401
farnesoid X nuclear receptorHomo sapiens (human)Potency24.15290.375827.485161.6524AID743220; AID743239
estrogen nuclear receptor alphaHomo sapiens (human)Potency36.57870.000229.305416,493.5996AID743069; AID743075; AID743078
peroxisome proliferator-activated receptor deltaHomo sapiens (human)Potency27.62000.001024.504861.6448AID743212; AID743215
peroxisome proliferator activated receptor gammaHomo sapiens (human)Potency20.74140.001019.414170.9645AID743094; AID743140; AID743191
vitamin D (1,25- dihydroxyvitamin D3) receptorHomo sapiens (human)Potency41.39440.023723.228263.5986AID743222; AID743223
caspase-3Homo sapiens (human)Potency68.79890.013326.981070.7614AID1346978
v-jun sarcoma virus 17 oncogene homolog (avian)Homo sapiens (human)Potency58.37450.057821.109761.2679AID1159528
Caspase-7Cricetulus griseus (Chinese hamster)Potency68.79890.006723.496068.5896AID1346980
chromobox protein homolog 1Homo sapiens (human)Potency100.00000.006026.168889.1251AID540317
caspase-3Cricetulus griseus (Chinese hamster)Potency68.79890.006723.496068.5896AID1346980
nuclear factor erythroid 2-related factor 2 isoform 1Homo sapiens (human)Potency51.67740.000627.21521,122.0200AID743202; AID743219
Cellular tumor antigen p53Homo sapiens (human)Potency70.83720.002319.595674.0614AID651631; AID720552
Guanine nucleotide-binding protein GHomo sapiens (human)Potency31.62281.995325.532750.1187AID624288
TAR DNA-binding protein 43Homo sapiens (human)Potency22.38721.778316.208135.4813AID652104
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Bile salt export pumpHomo sapiens (human)IC50 (µMol)10.60000.11007.190310.0000AID1443986
Vitamin D3 receptorHomo sapiens (human)IC50 (µMol)100.00000.00000.43746.4300AID1277414; AID1277417
Ileal sodium/bile acid cotransporterHomo sapiens (human)Ki3.30003.30006.400010.0000AID681332
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Alpha-2A adrenergic receptorHomo sapiens (human)EC50 (µMol)6.71000.00080.37336.7100AID297450
Vitamin D3 receptorHomo sapiens (human)EC50 (µMol)150.00000.00000.14232.1400AID1277413; AID1277415
Alpha-2B adrenergic receptorHomo sapiens (human)EC50 (µMol)6.71000.00051.36446.7100AID297450
Alpha-2C adrenergic receptorHomo sapiens (human)EC50 (µMol)6.71000.00050.55416.7100AID297450
Retinoic acid receptor RXR-alphaMus musculus (house mouse)EC50 (µMol)16.80000.04001.71805.0000AID629094
Bile acid receptorMus musculus (house mouse)EC50 (µMol)16.80000.15202.04315.0000AID629094; AID635573
G-protein coupled bile acid receptor 1Homo sapiens (human)EC50 (µMol)15.48200.02372.52598.9000AID1059690; AID1193333; AID1569580; AID1569582; AID1686017; AID1798108; AID297450; AID324923; AID592917; AID673744
Bile acid receptorHomo sapiens (human)EC50 (µMol)13.38060.00401.419110.0000AID1166181; AID1232938; AID1305701; AID1313848; AID1349338; AID1351287; AID1413747; AID1446797; AID1546885; AID1576216; AID1633315; AID1686015; AID1798109; AID1798115; AID1799080; AID240134; AID240176; AID240314; AID265705; AID267784; AID294203; AID297451; AID324925; AID388376; AID441572; AID592913; AID629055; AID662692; AID673746; AID72525; AID72526; AID72531
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (281)

Processvia Protein(s)Taxonomy
fatty acid metabolic processBile salt export pumpHomo sapiens (human)
bile acid biosynthetic processBile salt export pumpHomo sapiens (human)
xenobiotic metabolic processBile salt export pumpHomo sapiens (human)
xenobiotic transmembrane transportBile salt export pumpHomo sapiens (human)
response to oxidative stressBile salt export pumpHomo sapiens (human)
bile acid metabolic processBile salt export pumpHomo sapiens (human)
response to organic cyclic compoundBile salt export pumpHomo sapiens (human)
bile acid and bile salt transportBile salt export pumpHomo sapiens (human)
canalicular bile acid transportBile salt export pumpHomo sapiens (human)
protein ubiquitinationBile salt export pumpHomo sapiens (human)
regulation of fatty acid beta-oxidationBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transportBile salt export pumpHomo sapiens (human)
bile acid signaling pathwayBile salt export pumpHomo sapiens (human)
cholesterol homeostasisBile salt export pumpHomo sapiens (human)
response to estrogenBile salt export pumpHomo sapiens (human)
response to ethanolBile salt export pumpHomo sapiens (human)
xenobiotic export from cellBile salt export pumpHomo sapiens (human)
lipid homeostasisBile salt export pumpHomo sapiens (human)
phospholipid homeostasisBile salt export pumpHomo sapiens (human)
positive regulation of bile acid secretionBile salt export pumpHomo sapiens (human)
regulation of bile acid metabolic processBile salt export pumpHomo sapiens (human)
transmembrane transportBile salt export pumpHomo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycle G2/M phase transitionCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
ER overload responseCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
mitophagyCellular tumor antigen p53Homo sapiens (human)
in utero embryonic developmentCellular tumor antigen p53Homo sapiens (human)
somitogenesisCellular tumor antigen p53Homo sapiens (human)
release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
hematopoietic progenitor cell differentiationCellular tumor antigen p53Homo sapiens (human)
T cell proliferation involved in immune responseCellular tumor antigen p53Homo sapiens (human)
B cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
T cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
response to ischemiaCellular tumor antigen p53Homo sapiens (human)
nucleotide-excision repairCellular tumor antigen p53Homo sapiens (human)
double-strand break repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
protein import into nucleusCellular tumor antigen p53Homo sapiens (human)
autophagyCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrestCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 class mediatorCellular tumor antigen p53Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
Ras protein signal transductionCellular tumor antigen p53Homo sapiens (human)
gastrulationCellular tumor antigen p53Homo sapiens (human)
neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
protein localizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA replicationCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
determination of adult lifespanCellular tumor antigen p53Homo sapiens (human)
mRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
rRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
response to salt stressCellular tumor antigen p53Homo sapiens (human)
response to inorganic substanceCellular tumor antigen p53Homo sapiens (human)
response to X-rayCellular tumor antigen p53Homo sapiens (human)
response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
positive regulation of gene expressionCellular tumor antigen p53Homo sapiens (human)
cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
viral processCellular tumor antigen p53Homo sapiens (human)
glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
cerebellum developmentCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell growthCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
mitotic G1 DNA damage checkpoint signalingCellular tumor antigen p53Homo sapiens (human)
negative regulation of telomere maintenance via telomeraseCellular tumor antigen p53Homo sapiens (human)
T cell differentiation in thymusCellular tumor antigen p53Homo sapiens (human)
tumor necrosis factor-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
regulation of tissue remodelingCellular tumor antigen p53Homo sapiens (human)
cellular response to UVCellular tumor antigen p53Homo sapiens (human)
multicellular organism growthCellular tumor antigen p53Homo sapiens (human)
positive regulation of mitochondrial membrane permeabilityCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
entrainment of circadian clock by photoperiodCellular tumor antigen p53Homo sapiens (human)
mitochondrial DNA repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
transcription initiation-coupled chromatin remodelingCellular tumor antigen p53Homo sapiens (human)
negative regulation of proteolysisCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of RNA polymerase II transcription preinitiation complex assemblyCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
response to antibioticCellular tumor antigen p53Homo sapiens (human)
fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
circadian behaviorCellular tumor antigen p53Homo sapiens (human)
bone marrow developmentCellular tumor antigen p53Homo sapiens (human)
embryonic organ developmentCellular tumor antigen p53Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationCellular tumor antigen p53Homo sapiens (human)
protein stabilizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of helicase activityCellular tumor antigen p53Homo sapiens (human)
protein tetramerizationCellular tumor antigen p53Homo sapiens (human)
chromosome organizationCellular tumor antigen p53Homo sapiens (human)
neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
hematopoietic stem cell differentiationCellular tumor antigen p53Homo sapiens (human)
negative regulation of glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
type II interferon-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
cardiac septum morphogenesisCellular tumor antigen p53Homo sapiens (human)
positive regulation of programmed necrotic cell deathCellular tumor antigen p53Homo sapiens (human)
protein-containing complex assemblyCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressCellular tumor antigen p53Homo sapiens (human)
thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
necroptotic processCellular tumor antigen p53Homo sapiens (human)
cellular response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
cellular response to xenobiotic stimulusCellular tumor antigen p53Homo sapiens (human)
cellular response to ionizing radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to UV-CCellular tumor antigen p53Homo sapiens (human)
stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
cellular response to actinomycin DCellular tumor antigen p53Homo sapiens (human)
positive regulation of release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
cellular senescenceCellular tumor antigen p53Homo sapiens (human)
replicative senescenceCellular tumor antigen p53Homo sapiens (human)
oxidative stress-induced premature senescenceCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
oligodendrocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of execution phase of apoptosisCellular tumor antigen p53Homo sapiens (human)
negative regulation of mitophagyCellular tumor antigen p53Homo sapiens (human)
regulation of mitochondrial membrane permeability involved in apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of G1 to G0 transitionCellular tumor antigen p53Homo sapiens (human)
negative regulation of miRNA processingCellular tumor antigen p53Homo sapiens (human)
negative regulation of glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
negative regulation of pentose-phosphate shuntCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
regulation of fibroblast apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
positive regulation of cellular senescenceCellular tumor antigen p53Homo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
positive regulation of cytokine productionAlpha-2A adrenergic receptorHomo sapiens (human)
DNA replicationAlpha-2A adrenergic receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
adenylate cyclase-activating G protein-coupled receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
Ras protein signal transductionAlpha-2A adrenergic receptorHomo sapiens (human)
Rho protein signal transductionAlpha-2A adrenergic receptorHomo sapiens (human)
female pregnancyAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of cell population proliferationAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of norepinephrine secretionAlpha-2A adrenergic receptorHomo sapiens (human)
regulation of vasoconstrictionAlpha-2A adrenergic receptorHomo sapiens (human)
actin cytoskeleton organizationAlpha-2A adrenergic receptorHomo sapiens (human)
platelet activationAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of cell migrationAlpha-2A adrenergic receptorHomo sapiens (human)
activation of protein kinase activityAlpha-2A adrenergic receptorHomo sapiens (human)
activation of protein kinase B activityAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of epinephrine secretionAlpha-2A adrenergic receptorHomo sapiens (human)
cellular response to hormone stimulusAlpha-2A adrenergic receptorHomo sapiens (human)
receptor transactivationAlpha-2A adrenergic receptorHomo sapiens (human)
vasodilationAlpha-2A adrenergic receptorHomo sapiens (human)
glucose homeostasisAlpha-2A adrenergic receptorHomo sapiens (human)
fear responseAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of potassium ion transportAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of MAP kinase activityAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of MAPK cascadeAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of epidermal growth factor receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of calcium ion-dependent exocytosisAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of insulin secretionAlpha-2A adrenergic receptorHomo sapiens (human)
intestinal absorptionAlpha-2A adrenergic receptorHomo sapiens (human)
thermoceptionAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of lipid catabolic processAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of membrane protein ectodomain proteolysisAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of calcium ion transportAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of insulin secretion involved in cellular response to glucose stimulusAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of uterine smooth muscle contractionAlpha-2A adrenergic receptorHomo sapiens (human)
adrenergic receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
adenylate cyclase-inhibiting adrenergic receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
phospholipase C-activating adrenergic receptor signaling pathwayAlpha-2A adrenergic receptorHomo sapiens (human)
positive regulation of wound healingAlpha-2A adrenergic receptorHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of calcium ion transmembrane transporter activityAlpha-2A adrenergic receptorHomo sapiens (human)
negative regulation of DNA-templated transcriptionVitamin D3 receptorHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIVitamin D3 receptorHomo sapiens (human)
cell morphogenesisVitamin D3 receptorHomo sapiens (human)
skeletal system developmentVitamin D3 receptorHomo sapiens (human)
calcium ion transportVitamin D3 receptorHomo sapiens (human)
intracellular calcium ion homeostasisVitamin D3 receptorHomo sapiens (human)
lactationVitamin D3 receptorHomo sapiens (human)
negative regulation of cell population proliferationVitamin D3 receptorHomo sapiens (human)
positive regulation of gene expressionVitamin D3 receptorHomo sapiens (human)
negative regulation of keratinocyte proliferationVitamin D3 receptorHomo sapiens (human)
positive regulation of vitamin D 24-hydroxylase activityVitamin D3 receptorHomo sapiens (human)
positive regulation of bone mineralizationVitamin D3 receptorHomo sapiens (human)
phosphate ion transmembrane transportVitamin D3 receptorHomo sapiens (human)
bile acid signaling pathwayVitamin D3 receptorHomo sapiens (human)
mRNA transcription by RNA polymerase IIVitamin D3 receptorHomo sapiens (human)
positive regulation of keratinocyte differentiationVitamin D3 receptorHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIVitamin D3 receptorHomo sapiens (human)
decidualizationVitamin D3 receptorHomo sapiens (human)
intestinal absorptionVitamin D3 receptorHomo sapiens (human)
apoptotic process involved in mammary gland involutionVitamin D3 receptorHomo sapiens (human)
positive regulation of apoptotic process involved in mammary gland involutionVitamin D3 receptorHomo sapiens (human)
regulation of calcidiol 1-monooxygenase activityVitamin D3 receptorHomo sapiens (human)
mammary gland branching involved in pregnancyVitamin D3 receptorHomo sapiens (human)
vitamin D receptor signaling pathwayVitamin D3 receptorHomo sapiens (human)
positive regulation of vitamin D receptor signaling pathwayVitamin D3 receptorHomo sapiens (human)
response to bile acidVitamin D3 receptorHomo sapiens (human)
multicellular organism developmentVitamin D3 receptorHomo sapiens (human)
cell differentiationVitamin D3 receptorHomo sapiens (human)
MAPK cascadeAlpha-2B adrenergic receptorHomo sapiens (human)
angiogenesisAlpha-2B adrenergic receptorHomo sapiens (human)
regulation of vascular associated smooth muscle contractionAlpha-2B adrenergic receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayAlpha-2B adrenergic receptorHomo sapiens (human)
cell-cell signalingAlpha-2B adrenergic receptorHomo sapiens (human)
female pregnancyAlpha-2B adrenergic receptorHomo sapiens (human)
negative regulation of norepinephrine secretionAlpha-2B adrenergic receptorHomo sapiens (human)
platelet activationAlpha-2B adrenergic receptorHomo sapiens (human)
activation of protein kinase B activityAlpha-2B adrenergic receptorHomo sapiens (human)
negative regulation of epinephrine secretionAlpha-2B adrenergic receptorHomo sapiens (human)
receptor transactivationAlpha-2B adrenergic receptorHomo sapiens (human)
positive regulation of MAPK cascadeAlpha-2B adrenergic receptorHomo sapiens (human)
positive regulation of neuron differentiationAlpha-2B adrenergic receptorHomo sapiens (human)
positive regulation of blood pressureAlpha-2B adrenergic receptorHomo sapiens (human)
positive regulation of uterine smooth muscle contractionAlpha-2B adrenergic receptorHomo sapiens (human)
adrenergic receptor signaling pathwayAlpha-2B adrenergic receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayAlpha-2B adrenergic receptorHomo sapiens (human)
regulation of smooth muscle contractionAlpha-2C adrenergic receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayAlpha-2C adrenergic receptorHomo sapiens (human)
cell-cell signalingAlpha-2C adrenergic receptorHomo sapiens (human)
negative regulation of norepinephrine secretionAlpha-2C adrenergic receptorHomo sapiens (human)
regulation of vasoconstrictionAlpha-2C adrenergic receptorHomo sapiens (human)
platelet activationAlpha-2C adrenergic receptorHomo sapiens (human)
activation of protein kinase B activityAlpha-2C adrenergic receptorHomo sapiens (human)
negative regulation of epinephrine secretionAlpha-2C adrenergic receptorHomo sapiens (human)
receptor transactivationAlpha-2C adrenergic receptorHomo sapiens (human)
positive regulation of MAPK cascadeAlpha-2C adrenergic receptorHomo sapiens (human)
positive regulation of neuron differentiationAlpha-2C adrenergic receptorHomo sapiens (human)
adrenergic receptor signaling pathwayAlpha-2C adrenergic receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayAlpha-2C adrenergic receptorHomo sapiens (human)
negative regulation of insulin secretionAlpha-2C adrenergic receptorHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusGuanine nucleotide-binding protein GHomo sapiens (human)
renal water homeostasisGuanine nucleotide-binding protein GHomo sapiens (human)
G protein-coupled receptor signaling pathwayGuanine nucleotide-binding protein GHomo sapiens (human)
regulation of insulin secretionGuanine nucleotide-binding protein GHomo sapiens (human)
cellular response to glucagon stimulusGuanine nucleotide-binding protein GHomo sapiens (human)
sodium ion transportIleal sodium/bile acid cotransporterHomo sapiens (human)
response to bacteriumIleal sodium/bile acid cotransporterHomo sapiens (human)
bile acid and bile salt transportIleal sodium/bile acid cotransporterHomo sapiens (human)
transmembrane transportIleal sodium/bile acid cotransporterHomo sapiens (human)
negative regulation of protein phosphorylationTAR DNA-binding protein 43Homo sapiens (human)
mRNA processingTAR DNA-binding protein 43Homo sapiens (human)
RNA splicingTAR DNA-binding protein 43Homo sapiens (human)
negative regulation of gene expressionTAR DNA-binding protein 43Homo sapiens (human)
regulation of protein stabilityTAR DNA-binding protein 43Homo sapiens (human)
positive regulation of insulin secretionTAR DNA-binding protein 43Homo sapiens (human)
response to endoplasmic reticulum stressTAR DNA-binding protein 43Homo sapiens (human)
positive regulation of protein import into nucleusTAR DNA-binding protein 43Homo sapiens (human)
regulation of circadian rhythmTAR DNA-binding protein 43Homo sapiens (human)
regulation of apoptotic processTAR DNA-binding protein 43Homo sapiens (human)
negative regulation by host of viral transcriptionTAR DNA-binding protein 43Homo sapiens (human)
rhythmic processTAR DNA-binding protein 43Homo sapiens (human)
regulation of cell cycleTAR DNA-binding protein 43Homo sapiens (human)
3'-UTR-mediated mRNA destabilizationTAR DNA-binding protein 43Homo sapiens (human)
3'-UTR-mediated mRNA stabilizationTAR DNA-binding protein 43Homo sapiens (human)
nuclear inner membrane organizationTAR DNA-binding protein 43Homo sapiens (human)
amyloid fibril formationTAR DNA-binding protein 43Homo sapiens (human)
regulation of gene expressionTAR DNA-binding protein 43Homo sapiens (human)
cell surface bile acid receptor signaling pathwayG-protein coupled bile acid receptor 1Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeG-protein coupled bile acid receptor 1Homo sapiens (human)
cellular response to bile acidG-protein coupled bile acid receptor 1Homo sapiens (human)
positive regulation of cholangiocyte proliferationG-protein coupled bile acid receptor 1Homo sapiens (human)
regulation of bicellular tight junction assemblyG-protein coupled bile acid receptor 1Homo sapiens (human)
G protein-coupled receptor signaling pathwayG-protein coupled bile acid receptor 1Homo sapiens (human)
negative regulation of very-low-density lipoprotein particle remodelingBile acid receptorHomo sapiens (human)
positive regulation of DNA-templated transcriptionBile acid receptorHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIBile acid receptorHomo sapiens (human)
nitrogen catabolite activation of transcription from RNA polymerase II promoterBile acid receptorHomo sapiens (human)
intracellular glucose homeostasisBile acid receptorHomo sapiens (human)
regulation of transcription by RNA polymerase IIBile acid receptorHomo sapiens (human)
transcription by RNA polymerase IIBile acid receptorHomo sapiens (human)
inflammatory responseBile acid receptorHomo sapiens (human)
cell-cell junction assemblyBile acid receptorHomo sapiens (human)
Notch signaling pathwayBile acid receptorHomo sapiens (human)
bile acid metabolic processBile acid receptorHomo sapiens (human)
negative regulation of tumor necrosis factor-mediated signaling pathwayBile acid receptorHomo sapiens (human)
regulation of low-density lipoprotein particle clearanceBile acid receptorHomo sapiens (human)
intracellular receptor signaling pathwayBile acid receptorHomo sapiens (human)
negative regulation of type II interferon productionBile acid receptorHomo sapiens (human)
negative regulation of interleukin-1 productionBile acid receptorHomo sapiens (human)
negative regulation of interleukin-2 productionBile acid receptorHomo sapiens (human)
negative regulation of interleukin-6 productionBile acid receptorHomo sapiens (human)
negative regulation of tumor necrosis factor productionBile acid receptorHomo sapiens (human)
positive regulation of interleukin-17 productionBile acid receptorHomo sapiens (human)
toll-like receptor 9 signaling pathwayBile acid receptorHomo sapiens (human)
regulation of urea metabolic processBile acid receptorHomo sapiens (human)
intracellular triglyceride homeostasisBile acid receptorHomo sapiens (human)
positive regulation of insulin secretion involved in cellular response to glucose stimulusBile acid receptorHomo sapiens (human)
bile acid signaling pathwayBile acid receptorHomo sapiens (human)
intracellular bile acid receptor signaling pathwayBile acid receptorHomo sapiens (human)
cholesterol homeostasisBile acid receptorHomo sapiens (human)
defense response to bacteriumBile acid receptorHomo sapiens (human)
negative regulation of apoptotic processBile acid receptorHomo sapiens (human)
negative regulation of canonical NF-kappaB signal transductionBile acid receptorHomo sapiens (human)
innate immune responseBile acid receptorHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIBile acid receptorHomo sapiens (human)
positive regulation of insulin receptor signaling pathwayBile acid receptorHomo sapiens (human)
fatty acid homeostasisBile acid receptorHomo sapiens (human)
regulation of insulin secretion involved in cellular response to glucose stimulusBile acid receptorHomo sapiens (human)
regulation of bile acid biosynthetic processBile acid receptorHomo sapiens (human)
cellular response to lipopolysaccharideBile acid receptorHomo sapiens (human)
cellular response to fatty acidBile acid receptorHomo sapiens (human)
cellular response to organonitrogen compoundBile acid receptorHomo sapiens (human)
negative regulation of monocyte chemotactic protein-1 productionBile acid receptorHomo sapiens (human)
regulation of cholesterol metabolic processBile acid receptorHomo sapiens (human)
cellular response to bile acidBile acid receptorHomo sapiens (human)
positive regulation of adipose tissue developmentBile acid receptorHomo sapiens (human)
positive regulation of phosphatidic acid biosynthetic processBile acid receptorHomo sapiens (human)
positive regulation of glutamate metabolic processBile acid receptorHomo sapiens (human)
positive regulation of ammonia assimilation cycleBile acid receptorHomo sapiens (human)
cell differentiationBile acid receptorHomo sapiens (human)
negative regulation of inflammatory responseBile acid receptorHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (74)

Processvia Protein(s)Taxonomy
protein bindingBile salt export pumpHomo sapiens (human)
ATP bindingBile salt export pumpHomo sapiens (human)
ABC-type xenobiotic transporter activityBile salt export pumpHomo sapiens (human)
bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
canalicular bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transporter activityBile salt export pumpHomo sapiens (human)
ABC-type bile acid transporter activityBile salt export pumpHomo sapiens (human)
ATP hydrolysis activityBile salt export pumpHomo sapiens (human)
transcription cis-regulatory region bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
core promoter sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
TFIID-class transcription factor complex bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
protease bindingCellular tumor antigen p53Homo sapiens (human)
p53 bindingCellular tumor antigen p53Homo sapiens (human)
DNA bindingCellular tumor antigen p53Homo sapiens (human)
chromatin bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activityCellular tumor antigen p53Homo sapiens (human)
mRNA 3'-UTR bindingCellular tumor antigen p53Homo sapiens (human)
copper ion bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingCellular tumor antigen p53Homo sapiens (human)
zinc ion bindingCellular tumor antigen p53Homo sapiens (human)
enzyme bindingCellular tumor antigen p53Homo sapiens (human)
receptor tyrosine kinase bindingCellular tumor antigen p53Homo sapiens (human)
ubiquitin protein ligase bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase regulator activityCellular tumor antigen p53Homo sapiens (human)
ATP-dependent DNA/DNA annealing activityCellular tumor antigen p53Homo sapiens (human)
identical protein bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase bindingCellular tumor antigen p53Homo sapiens (human)
protein heterodimerization activityCellular tumor antigen p53Homo sapiens (human)
protein-folding chaperone bindingCellular tumor antigen p53Homo sapiens (human)
protein phosphatase 2A bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingCellular tumor antigen p53Homo sapiens (human)
14-3-3 protein bindingCellular tumor antigen p53Homo sapiens (human)
MDM2/MDM4 family protein bindingCellular tumor antigen p53Homo sapiens (human)
disordered domain specific bindingCellular tumor antigen p53Homo sapiens (human)
general transcription initiation factor bindingCellular tumor antigen p53Homo sapiens (human)
molecular function activator activityCellular tumor antigen p53Homo sapiens (human)
promoter-specific chromatin bindingCellular tumor antigen p53Homo sapiens (human)
alpha2-adrenergic receptor activityAlpha-2A adrenergic receptorHomo sapiens (human)
protein bindingAlpha-2A adrenergic receptorHomo sapiens (human)
protein kinase bindingAlpha-2A adrenergic receptorHomo sapiens (human)
alpha-1B adrenergic receptor bindingAlpha-2A adrenergic receptorHomo sapiens (human)
alpha-2C adrenergic receptor bindingAlpha-2A adrenergic receptorHomo sapiens (human)
thioesterase bindingAlpha-2A adrenergic receptorHomo sapiens (human)
heterotrimeric G-protein bindingAlpha-2A adrenergic receptorHomo sapiens (human)
protein homodimerization activityAlpha-2A adrenergic receptorHomo sapiens (human)
protein heterodimerization activityAlpha-2A adrenergic receptorHomo sapiens (human)
epinephrine bindingAlpha-2A adrenergic receptorHomo sapiens (human)
norepinephrine bindingAlpha-2A adrenergic receptorHomo sapiens (human)
guanyl-nucleotide exchange factor activityAlpha-2A adrenergic receptorHomo sapiens (human)
DNA-binding transcription factor activityVitamin D3 receptorHomo sapiens (human)
vitamin D response element bindingVitamin D3 receptorHomo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificVitamin D3 receptorHomo sapiens (human)
DNA bindingVitamin D3 receptorHomo sapiens (human)
nuclear receptor activityVitamin D3 receptorHomo sapiens (human)
protein bindingVitamin D3 receptorHomo sapiens (human)
zinc ion bindingVitamin D3 receptorHomo sapiens (human)
bile acid nuclear receptor activityVitamin D3 receptorHomo sapiens (human)
nuclear retinoid X receptor bindingVitamin D3 receptorHomo sapiens (human)
calcitriol bindingVitamin D3 receptorHomo sapiens (human)
lithocholic acid bindingVitamin D3 receptorHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingVitamin D3 receptorHomo sapiens (human)
alpha2-adrenergic receptor activityAlpha-2B adrenergic receptorHomo sapiens (human)
protein bindingAlpha-2B adrenergic receptorHomo sapiens (human)
epinephrine bindingAlpha-2B adrenergic receptorHomo sapiens (human)
alpha2-adrenergic receptor activityAlpha-2C adrenergic receptorHomo sapiens (human)
protein bindingAlpha-2C adrenergic receptorHomo sapiens (human)
alpha-2A adrenergic receptor bindingAlpha-2C adrenergic receptorHomo sapiens (human)
protein homodimerization activityAlpha-2C adrenergic receptorHomo sapiens (human)
protein heterodimerization activityAlpha-2C adrenergic receptorHomo sapiens (human)
epinephrine bindingAlpha-2C adrenergic receptorHomo sapiens (human)
guanyl-nucleotide exchange factor activityAlpha-2C adrenergic receptorHomo sapiens (human)
G protein activityGuanine nucleotide-binding protein GHomo sapiens (human)
adenylate cyclase activator activityGuanine nucleotide-binding protein GHomo sapiens (human)
protein bindingIleal sodium/bile acid cotransporterHomo sapiens (human)
bile acid:sodium symporter activityIleal sodium/bile acid cotransporterHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingTAR DNA-binding protein 43Homo sapiens (human)
DNA bindingTAR DNA-binding protein 43Homo sapiens (human)
double-stranded DNA bindingTAR DNA-binding protein 43Homo sapiens (human)
RNA bindingTAR DNA-binding protein 43Homo sapiens (human)
mRNA 3'-UTR bindingTAR DNA-binding protein 43Homo sapiens (human)
protein bindingTAR DNA-binding protein 43Homo sapiens (human)
lipid bindingTAR DNA-binding protein 43Homo sapiens (human)
identical protein bindingTAR DNA-binding protein 43Homo sapiens (human)
pre-mRNA intronic bindingTAR DNA-binding protein 43Homo sapiens (human)
molecular condensate scaffold activityTAR DNA-binding protein 43Homo sapiens (human)
protein bindingG-protein coupled bile acid receptor 1Homo sapiens (human)
bile acid receptor activityG-protein coupled bile acid receptor 1Homo sapiens (human)
G protein-coupled bile acid receptor activityG-protein coupled bile acid receptor 1Homo sapiens (human)
RNA polymerase II transcription regulatory region sequence-specific DNA bindingBile acid receptorHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingBile acid receptorHomo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificBile acid receptorHomo sapiens (human)
transcription coregulator bindingBile acid receptorHomo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificBile acid receptorHomo sapiens (human)
DNA-binding transcription factor activityBile acid receptorHomo sapiens (human)
nuclear receptor activityBile acid receptorHomo sapiens (human)
protein bindingBile acid receptorHomo sapiens (human)
zinc ion bindingBile acid receptorHomo sapiens (human)
nuclear receptor bindingBile acid receptorHomo sapiens (human)
bile acid bindingBile acid receptorHomo sapiens (human)
bile acid receptor activityBile acid receptorHomo sapiens (human)
sequence-specific DNA bindingBile acid receptorHomo sapiens (human)
nuclear retinoid X receptor bindingBile acid receptorHomo sapiens (human)
chenodeoxycholic acid bindingBile acid receptorHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (48)

Processvia Protein(s)Taxonomy
basolateral plasma membraneBile salt export pumpHomo sapiens (human)
Golgi membraneBile salt export pumpHomo sapiens (human)
endosomeBile salt export pumpHomo sapiens (human)
plasma membraneBile salt export pumpHomo sapiens (human)
cell surfaceBile salt export pumpHomo sapiens (human)
apical plasma membraneBile salt export pumpHomo sapiens (human)
intercellular canaliculusBile salt export pumpHomo sapiens (human)
intracellular canaliculusBile salt export pumpHomo sapiens (human)
recycling endosomeBile salt export pumpHomo sapiens (human)
recycling endosome membraneBile salt export pumpHomo sapiens (human)
extracellular exosomeBile salt export pumpHomo sapiens (human)
membraneBile salt export pumpHomo sapiens (human)
nuclear bodyCellular tumor antigen p53Homo sapiens (human)
nucleusCellular tumor antigen p53Homo sapiens (human)
nucleoplasmCellular tumor antigen p53Homo sapiens (human)
replication forkCellular tumor antigen p53Homo sapiens (human)
nucleolusCellular tumor antigen p53Homo sapiens (human)
cytoplasmCellular tumor antigen p53Homo sapiens (human)
mitochondrionCellular tumor antigen p53Homo sapiens (human)
mitochondrial matrixCellular tumor antigen p53Homo sapiens (human)
endoplasmic reticulumCellular tumor antigen p53Homo sapiens (human)
centrosomeCellular tumor antigen p53Homo sapiens (human)
cytosolCellular tumor antigen p53Homo sapiens (human)
nuclear matrixCellular tumor antigen p53Homo sapiens (human)
PML bodyCellular tumor antigen p53Homo sapiens (human)
transcription repressor complexCellular tumor antigen p53Homo sapiens (human)
site of double-strand breakCellular tumor antigen p53Homo sapiens (human)
germ cell nucleusCellular tumor antigen p53Homo sapiens (human)
chromatinCellular tumor antigen p53Homo sapiens (human)
transcription regulator complexCellular tumor antigen p53Homo sapiens (human)
protein-containing complexCellular tumor antigen p53Homo sapiens (human)
cytoplasmAlpha-2A adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-2A adrenergic receptorHomo sapiens (human)
basolateral plasma membraneAlpha-2A adrenergic receptorHomo sapiens (human)
neuronal cell bodyAlpha-2A adrenergic receptorHomo sapiens (human)
axon terminusAlpha-2A adrenergic receptorHomo sapiens (human)
presynaptic active zone membraneAlpha-2A adrenergic receptorHomo sapiens (human)
dopaminergic synapseAlpha-2A adrenergic receptorHomo sapiens (human)
postsynaptic density membraneAlpha-2A adrenergic receptorHomo sapiens (human)
glutamatergic synapseAlpha-2A adrenergic receptorHomo sapiens (human)
GABA-ergic synapseAlpha-2A adrenergic receptorHomo sapiens (human)
receptor complexAlpha-2A adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-2A adrenergic receptorHomo sapiens (human)
nucleusVitamin D3 receptorHomo sapiens (human)
nucleusVitamin D3 receptorHomo sapiens (human)
nucleoplasmVitamin D3 receptorHomo sapiens (human)
cytosolVitamin D3 receptorHomo sapiens (human)
RNA polymerase II transcription regulator complexVitamin D3 receptorHomo sapiens (human)
chromatinVitamin D3 receptorHomo sapiens (human)
receptor complexVitamin D3 receptorHomo sapiens (human)
cytosolAlpha-2B adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-2B adrenergic receptorHomo sapiens (human)
cell surfaceAlpha-2B adrenergic receptorHomo sapiens (human)
intracellular membrane-bounded organelleAlpha-2B adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-2B adrenergic receptorHomo sapiens (human)
cytoplasmAlpha-2C adrenergic receptorHomo sapiens (human)
endosomeAlpha-2C adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-2C adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-2C adrenergic receptorHomo sapiens (human)
nucleoplasmRetinoic acid receptor RXR-alphaMus musculus (house mouse)
plasma membraneGuanine nucleotide-binding protein GHomo sapiens (human)
plasma membraneIleal sodium/bile acid cotransporterHomo sapiens (human)
microvillusIleal sodium/bile acid cotransporterHomo sapiens (human)
apical plasma membraneIleal sodium/bile acid cotransporterHomo sapiens (human)
intracellular non-membrane-bounded organelleTAR DNA-binding protein 43Homo sapiens (human)
nucleusTAR DNA-binding protein 43Homo sapiens (human)
nucleoplasmTAR DNA-binding protein 43Homo sapiens (human)
perichromatin fibrilsTAR DNA-binding protein 43Homo sapiens (human)
mitochondrionTAR DNA-binding protein 43Homo sapiens (human)
cytoplasmic stress granuleTAR DNA-binding protein 43Homo sapiens (human)
nuclear speckTAR DNA-binding protein 43Homo sapiens (human)
interchromatin granuleTAR DNA-binding protein 43Homo sapiens (human)
nucleoplasmTAR DNA-binding protein 43Homo sapiens (human)
chromatinTAR DNA-binding protein 43Homo sapiens (human)
cytoplasmG-protein coupled bile acid receptor 1Homo sapiens (human)
plasma membraneG-protein coupled bile acid receptor 1Homo sapiens (human)
receptor complexG-protein coupled bile acid receptor 1Homo sapiens (human)
plasma membraneG-protein coupled bile acid receptor 1Homo sapiens (human)
nucleoplasmBile acid receptorHomo sapiens (human)
chromatinBile acid receptorHomo sapiens (human)
euchromatinBile acid receptorHomo sapiens (human)
receptor complexBile acid receptorHomo sapiens (human)
RNA polymerase II transcription regulator complexBile acid receptorHomo sapiens (human)
nucleusBile acid receptorHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (384)

Assay IDTitleYearJournalArticle
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1345827Human FPR1 (Formylpeptide receptors)2000Inflammation research : official journal of the European Histamine Research Society ... [et al.], Dec, Volume: 49, Issue:12
Characterization of chenodeoxycholic acid as an endogenous antagonist of the G-coupled formyl peptide receptors.
AID1346437Human GPBA receptor (Bile acid receptor)2002Biochemical and biophysical research communications, Nov-15, Volume: 298, Issue:5
Identification of membrane-type receptor for bile acids (M-BAR).
AID1346766Human Farnesoid X receptor (1H. Liver X receptor-like receptors)1999Science (New York, N.Y.), May-21, Volume: 284, Issue:5418
Bile acids: natural ligands for an orphan nuclear receptor.
AID1346766Human Farnesoid X receptor (1H. Liver X receptor-like receptors)1999Molecular cell, May, Volume: 3, Issue:5
Endogenous bile acids are ligands for the nuclear receptor FXR/BAR.
AID1347411qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Mechanism Interrogation Plate v5.0 (MIPE) Libary2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID504812Inverse Agonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID504810Antagonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID592922Induction of FXR-mediated Ostbeta mRNA expression in human HepG2 cells at 20 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID1686056Drug metabolism in bile of fistula Wistar-Han rat model assessed as maximum biliary secretion of the administered compound including the secreted taurine conjugates at 60 umol/kg, iv by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID673747Agonist activity at GST-tagged FXR-LBD using biotinylated-SRC-1 peptide as substrate preincubated with compound for 30 mins measured after 4 hrs relative to 10 uM CDCA2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID240134Activation of human farnesoid X receptor; range is 10-302005Journal of medicinal chemistry, Aug-25, Volume: 48, Issue:17
Farnesoid X receptor: from structure to potential clinical applications.
AID588219FDA HLAED, gamma-glutamyl transferase (GGT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID1443986Inhibition of recombinant human BSEP expressed in baculovirus infected sf9 cell membrane vesicles assessed as reduction in ATP or AMP-dependent [3H]-taurocholic acid uptake in to vesicles preincubated for 5 mins followed by ATP/AMP addition measured after2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID1187915Agonist activity at GAL4 DNA binding domain tagged FXR ligand binding domain (unknown origin) expressed in human HepG2 cells co-expressing human human SHP gene promoter assessed as transactivation of SHP promoter driven luciferase activity at 10 uM relati2014Bioorganic & medicinal chemistry letters, Sep-01, Volume: 24, Issue:17
Cryptochinones from Cryptocarya chinensis act as farnesoid X receptor agonists.
AID1233566Cytotoxicity against human WI38 cells assessed as inhibition of cell growth at 20 ug/ml by MTT assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID240176Binding affinity for human Farnesoid X receptor in FRET assay2004Journal of medicinal chemistry, Aug-26, Volume: 47, Issue:18
Bile acid derivatives as ligands of the farnesoid X receptor. Synthesis, evaluation, and structure-activity relationship of a series of body and side chain modified analogues of chenodeoxycholic acid.
AID1079939Cirrhosis, proven histopathologically. Value is number of references indexed. [column 'CIRRH' in source]
AID1412048Induction of angiogenesis in HUVEC assessed as increase in tube formation at 12.5 uM after overnight incubation by inverted microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1079940Granulomatous liver disease, proven histopathologically. Value is number of references indexed. [column 'GRAN' in source]
AID409882Cytotoxicity against human GBM cells at 50 uM after 48 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID588218FDA HLAED, lactate dehydrogenase (LDH) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID680256TP_TRANSPORTER: inhibition of Taurocholate uptake (Taurochorate: 20 uM, CDCA: 200 uM) in Xenopus laevis oocytes1994Hepatology (Baltimore, Md.), Aug, Volume: 20, Issue:2
Functional characterization of the basolateral rat liver organic anion transporting polypeptide.
AID1059688Selectivity ratio of EC50 for TGR5 Y89A mutant (unknown origin) to EC50 for wild type TGR5 (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1852830Anticancer activity against human Huh-7 cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID294203Agonistic activity at FXR in HEK293 cells by GAL4 transactivation activity2007Bioorganic & medicinal chemistry, Apr-01, Volume: 15, Issue:7
Design, synthesis, and evaluation of non-steroidal farnesoid X receptor (FXR) antagonist.
AID441586Metabolic stability in bile fistula rat model assessed as conjugated drug level recovered in bile at 1 umol/min/kg, iv over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID1079932Highest frequency of moderate liver toxicity observed during clinical trials, expressed as a percentage. [column '% BIOL' in source]
AID1166186Agonist activity at FXR in human HepG2 cells assessed as induction of SHP mrNA expression at 50 uM by quantitative PCR method2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Extending the structure-activity relationship of anthranilic acid derivatives as farnesoid X receptor modulators: development of a highly potent partial farnesoid X receptor agonist.
AID1852826Anticancer activity against human HCT-116 cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID346025Binding affinity to beta cyclodextrin2009Bioorganic & medicinal chemistry, Jan-15, Volume: 17, Issue:2
Convenient QSAR model for predicting the complexation of structurally diverse compounds with beta-cyclodextrins.
AID422552Binding affinity to human pancreatic recombinant 1B PLA2 expressed in Escherichia coli at 100 uM by saturation transfer difference NMR spectroscopy2009Journal of natural products, Jan, Volume: 72, Issue:1
Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia.
AID490124Lipophilicity, log P of the compound2010European journal of medicinal chemistry, Jul, Volume: 45, Issue:7
Synthesis of bile acid derivatives and in vitro cytotoxic activity with pro-apoptotic process on multiple myeloma (KMS-11), glioblastoma multiforme (GBM), and colonic carcinoma (HCT-116) human cell lines.
AID1161666Agonist activity at FXR (unknown origin) expressed in human HepG2 cells assessed as stimulation of FXR response element IR1-mediated receptor transactivation at 1 uM by luciferase reporter gene assay2014Journal of medicinal chemistry, Sep-25, Volume: 57, Issue:18
Modification on ursodeoxycholic acid (UDCA) scaffold. discovery of bile acid derivatives as selective agonists of cell-surface G-protein coupled bile acid receptor 1 (GP-BAR1).
AID1059683Agonist activity at TGR5 E169A mutant (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1079944Benign tumor, proven histopathologically. Value is number of references indexed. [column 'T.BEN' in source]
AID1609608Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as increase in SULT2A1 protein expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1233570Antiplasmodial activity against Plasmodium falciparum 3D7 assessed as inhibition of parasite viability at 20 ug/ml by parasite lactate dehydrogenase assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID303502Activation of BSEP gene expression in human HepG2 cells at 25 uM after 18 hrs by RT-PCR2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1398006Inhibition of lipid accumulation in mouse 3T3L1 cells at 10 uM by oil red o staining-based assay2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Lipid accumulation inhibitory activities of novel isoxazole-based chenodeoxycholic acids: Design, synthesis and preliminary mechanism study.
AID1852831Anticancer activity against human SK-HEP1 cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID1569582Agonist activity at TGR5 in human NCI-H716 cells assessed as increase in cAMP accumulation after 60 mins by HTR-FRET assay2019Journal of medicinal chemistry, 07-25, Volume: 62, Issue:14
7-Methylation of Chenodeoxycholic Acid Derivatives Yields a Substantial Increase in TGR5 Receptor Potency.
AID588216FDA HLAED, serum glutamic oxaloacetic transaminase (SGOT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID1277415Agonist activity at VP16 tagged-VDR-LBD (unknown origin) expressed in HEK293T cells assessed as SRC1 coactivator peptide recruitment after 16 hrs by luciferase reporter gene based two hybrid assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Synthesis and evaluation of vitamin D receptor-mediated activities of cholesterol and vitamin D metabolites.
AID1609542Upregulation of FXR mRNA expression in human LO2 cells at 80 uM after 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1187925Agonist activity at GAL4 DNA binding domain tagged FXR ligand binding domain (unknown origin) expressed in human HepG2 cells co-expressing human PLTP gene promoter assessed as transactivation of PLTP promoter driven luciferase activity at 10 uM relative t2014Bioorganic & medicinal chemistry letters, Sep-01, Volume: 24, Issue:17
Cryptochinones from Cryptocarya chinensis act as farnesoid X receptor agonists.
AID1059679Agonist activity at TGR5 S270A mutant (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1351287Agonist activity at GST-tagged FXR-LBD (unknown origin) assessed as biotin-labeled SRC-1 recruitment after 30 mins by Alpha Screen assay2018European journal of medicinal chemistry, Jan-20, Volume: 144Synthesis, physicochemical properties, and biological activity of bile acids 3-glucuronides: Novel insights into bile acid signalling and detoxification.
AID1079946Presence of at least one case with successful reintroduction. [column 'REINT' in source]
AID1412071Pro-angiogenic activity in HUVEC assessed as decrease in VE-cadherin expression at 12.5 uM after overnight incubation by FITC-conjugated phalloidin staining based confocal microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1412068Increase in MMP9 level in HUVEC at 12.5 uM after 24 hrs by ELISA2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID592920Induction of FXR-mediated BSEP mRNA expression in human HepG2 cells at 20 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID1059692Selectivity ratio of EC50 for TGR5 N93A mutant (unknown origin) to EC50 for wild type TGR5 (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID297449Agonist activity at human TGR5 expressed in CHO cells after 5 hrs by CRE-driven luciferase reporter gene assay relative to lithocholic acid2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.
AID1138172Agonist activity at FXR in human HepG2 cells assessed as upregulation of BESP mRNA expression at 10 uM after 18 hrs by RT-PCR analysis2014Journal of medicinal chemistry, Feb-13, Volume: 57, Issue:3
Design, synthesis, and biological evaluation of potent dual agonists of nuclear and membrane bile acid receptors.
AID592918Induction of FXR-mediated SHP mRNA expression in human HepG2 cells at 20 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID409880Cytotoxicity against human HCT116 cells at 50 uM after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID588211Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in humans2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID588210Human drug-induced liver injury (DILI) modelling dataset from Ekins et al2010Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 38, Issue:12
A predictive ligand-based Bayesian model for human drug-induced liver injury.
AID1576216Agonist activity at GST-tagged FXR-LBD (unknown origin) assessed as biotinylated SRC-1 recruitment after 30 mins by Alpha Screen assay2019MedChemComm, Aug-01, Volume: 10, Issue:8
Dissecting the allosteric FXR modulation: a chemical biology approach using guggulsterone as a chemical tool.
AID1609588Upregulation of FXR protein expression in human LO2 cells at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1852833Anticancer activity against human MGC-803 cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID515398Cytotoxicity against human HET-1A cells assessed as cell viability after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Sep-15, Volume: 18, Issue:18
Bile acid toxicity structure-activity relationships: correlations between cell viability and lipophilicity in a panel of new and known bile acids using an oesophageal cell line (HET-1A).
AID1232938Agonist activity at human full length FXR expressed in HeLa cells cotransfected with pSG5-human RXR after 24 hrs by Dual-Glo luciferase reporter gene assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Fragmentation of GW4064 led to a highly potent partial farnesoid X receptor agonist with improved drug-like properties.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1305701Agonist activity at farnesoid x receptor(unknown origin)2016Bioorganic & medicinal chemistry, Aug-01, Volume: 24, Issue:15
Novel approaches to map small molecule-target interactions.
AID1166210Agonist activity at FXR in human HepG2 cells assessed as induction of IBABP mrNA expression at 50 uM by quantitative PCR method2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Extending the structure-activity relationship of anthranilic acid derivatives as farnesoid X receptor modulators: development of a highly potent partial farnesoid X receptor agonist.
AID714968Agonist activity at human FXR expressed in human HepG2 cells assessed as transactivation of SHP gene by measuring relative luciferase activity by transient transfection reporter assay relative to control2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Sesquiterpenoids from Atractylodes macrocephala act as farnesoid X receptor and progesterone receptor modulators.
AID1079937Severe hepatitis, defined as possibly life-threatening liver failure or through clinical observations. Value is number of references indexed. [column 'MASS' in source]
AID679680TP_TRANSPORTER: inhibition of Taurocholate uptake (Taurochorate: 10 uM, CDCA: 100 uM) in Xenopus laevis oocytes1994The Journal of clinical investigation, Mar, Volume: 93, Issue:3
Molecular cloning, chromosomal localization, and functional characterization of a human liver Na+/bile acid cotransporter.
AID1633315Transactivation of human FXR (unknown origin) expressed in HepG2 cells co-expressing pSG5RXR/pGL4.70 after 24 hrs post transfection by luciferase reporter gene assay2019ACS medicinal chemistry letters, Apr-11, Volume: 10, Issue:4
Novel Isoxazole Derivatives with Potent FXR Agonistic Activity Prevent Acetaminophen-Induced Liver Injury.
AID1232958Partial agonist activity at FXR in human HepG2 cells assessed as induction of BSEP gene expression at 50 uM after 24 hrs by qRT-PCR analysis (Rvb = 100%)2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Fragmentation of GW4064 led to a highly potent partial farnesoid X receptor agonist with improved drug-like properties.
AID1079943Malignant tumor, proven histopathologically. Value is number of references indexed. [column 'T.MAL' in source]
AID1412064Increase in VEGFR1 m-RNA expression in HUVEC at 12.5 uM after 12 to 24 hrs by RT-qPCR analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1079934Highest frequency of acute liver toxicity observed during clinical trials, expressed as a percentage. [column '% AIGUE' in source]
AID303512Activation of TGR5 expressed in HEK293 cells assessed as elevation of cAMP production at 10 uM relative to control2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID588217FDA HLAED, serum glutamic pyruvic transaminase (SGPT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID1609604Increase in FXR protein expression in guggulsterone-treated human LO2 cells at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1609545Agonist activity at FXR in human LO2 cells assessed as increase in UGT2A4 mRNA expression at 80 uM after 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1413747Transactivation of full-length FXR (unknown origin) expressed in HEK293 cells after 18 hrs by dual-glo luciferase reporter gene assay2018MedChemComm, Oct-01, Volume: 9, Issue:10
Discovery of new FXR agonists based on 6-ECDCA binding properties by virtual screening and molecular docking.
AID1609543Agonist activity at FXR in human LO2 cells assessed as increase in SHP1 mRNA expression at 80 uM after 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID662691Enhancement of human BSEP E297G mutant trafficking to cell membrane in MDCK2 cells co-expressing NTCP assessed as accumulation of [3H]taurocholate at 10 uM preincubated for 24 hrs prior incubation with [3H]taurocholate measured after 2 hrs by liquid scint2012Bioorganic & medicinal chemistry letters, Jun-15, Volume: 22, Issue:12
E297G mutated bile salt export pump (BSEP) function enhancers derived from GW4064: structural development study and separation from farnesoid X receptor-agonistic activity.
AID681660TP_TRANSPORTER: inhibition of Taurocholate uptake (Taurochorate: 5 uM, CDCA: 50 uM) in membrane vesicles isolated from Bsep-expressing Balb-3T3 cells2000Gene, Jan-04, Volume: 241, Issue:1
Molecular cloning and characterization of the murine bile salt export pump.
AID422554Agonist activity at human FXR transfected in human HuH7 cells co-transfected with human BSEP promoter reporter plasmid at 10 uM after 30 hrs by dual-luciferase reporter assay relative to control2009Journal of natural products, Jan, Volume: 72, Issue:1
Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia.
AID1233568Antitrypanosomal activity against Trypanosoma brucei brucei Lister 427 bloodstream forms assessed as inhibition of parasite growth at 20 ug/ml by microtiter plate based assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID1451548Transactivation of FXR in human HepG2 cells assessed as FGF19 mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID303493Activation of human PXR expressed in HEK293 cells at 50 uM by GAL4 activation assay relative to control2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID657096Restoration of human BSEP E297G mutant trafficking to cell membrane in MDCK2 cells co-expressing NTCP assessed as accumulation of [3H]taurocholate at 100 uM preincubated for 24 hrs prior incubation with [3H]taurocholate measured after 2 hrs by liquid scin2012Bioorganic & medicinal chemistry, May-01, Volume: 20, Issue:9
Discovery and structural development of small molecules that enhance transport activity of bile salt export pump mutant associated with progressive familial intrahepatic cholestasis type 2.
AID714963Agonist activity at human FXR expressed in human HepG2 cells assessed as down-regulation of CYP7A1 gene transactivation at 10 uM by measuring relative luciferase activity by transient transfection reporter assay relative to control2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Sesquiterpenoids from Atractylodes macrocephala act as farnesoid X receptor and progesterone receptor modulators.
AID54340Binding constant towards cyclodextrin conjugated peptide (with pyrene) at 25 degrees Centigrade2003Bioorganic & medicinal chemistry letters, Dec-15, Volume: 13, Issue:24
Fluorescence resonance energy transfer in a novel cyclodextrin-peptide conjugate for detecting steroid molecules.
AID1233552Cytotoxicity against human WI38 cells by MTT assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID1079935Cytolytic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is > 5 (see ACUTE). Value is number of references indexed. [column 'CYTOL' in source]
AID672335Agonist activity at FXR LBD expressed in human HepG2 cells co-expressing GAL4 DBD and pG5-luc assessed as SHP promoter-driven luciferase activity at 10 uM after 48 hrs by transient transfection reporter assay relative to control2012Bioorganic & medicinal chemistry letters, Jul-15, Volume: 22, Issue:14
Triterpenes from Alisma orientalis act as farnesoid X receptor agonists.
AID1138170Agonist activity at FXR in human HepG2 cells assessed as upregulation of SHP mRNA expression at 10 uM after 18 hrs by RT-PCR analysis2014Journal of medicinal chemistry, Feb-13, Volume: 57, Issue:3
Design, synthesis, and biological evaluation of potent dual agonists of nuclear and membrane bile acid receptors.
AID265705Agonist activity at human recombinant FXR by transactivation of TK-MH100x4-LUC reporter gene in HEK293 cells2006Bioorganic & medicinal chemistry letters, Jun-15, Volume: 16, Issue:12
Diphenylmethane skeleton as a multi-template for nuclear receptor ligands: preparation of FXR and PPAR ligands.
AID592919Induction of FXR-mediated SHP mRNA expression in human HepG2 cells at 1 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID1327990Antiproliferative activity against human HT-29 cells after 48 hrs by SRB assay2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel 3,4-seco bile acid diamides as selective anticancer proliferation and migration agents.
AID1609590Agonist activity at FXR in human LO2 cells assessed as increase in BSEP protein expression at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1686017Agonist activity at TGR5 in human NCI-H716 cells assessed as stimulation of intracellular cAMP accumulation incubated for 60 mins by HTR-FRET assay2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID629056Agonist activity at full length mouse FXR/RXRalpha expressed in human HEK293 cells assessed as induction of transcriptional activity at 10 ug/mL after 18 hrs by dual luciferase reporter gene assay relative to control2011Bioorganic & medicinal chemistry, Nov-15, Volume: 19, Issue:22
Pharmacophore-based discovery of FXR-agonists. Part II: identification of bioactive triterpenes from Ganoderma lucidum.
AID441576Solubility in 0.1M HCl water solution2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID587932Antagonist activity at FXR expressed in human HepG2 cells assessed as inhibition of CDCA-induced transactivation of FXR at 50 uM after 18 hrs by luminometric analysis2011Journal of medicinal chemistry, Mar-10, Volume: 54, Issue:5
Discovery of sulfated sterols from marine invertebrates as a new class of marine natural antagonists of farnesoid-X-receptor.
AID303511Activation of IBABP gene expression in human Caco-2 cells at 25 uM after 6 days by RT-PCR2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1234671Antiparasitic activity against Trypanosoma cruzi strain CL Brener epimastigotes after 12 days by ELISA reader2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Enzymatic synthesis of bile acid derivatives and biological evaluation against Trypanosoma cruzi.
AID592913Agonist activity at human GST-fused FXR LBD assessed as coactivator interaction with receptor ligand binding domain by Alphascreen assay2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID1165599Agonist activity at human FXR ligand binding domain assessed as induction of biotinylated SRC1 peptide recruitment at 4 uM by coactivator recruitment assay2014Journal of medicinal chemistry, Oct-23, Volume: 57, Issue:20
Exploitation of cholane scaffold for the discovery of potent and selective farnesoid X receptor (FXR) and G-protein coupled bile acid receptor 1 (GP-BAR1) ligands.
AID1059687Selectivity ratio of EC50 for TGR5 N76A mutant (unknown origin) to EC50 for wild type TGR5 (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1187912Agonist activity at GAL4 DNA binding domain tagged FXR ligand binding domain (unknown origin) expressed in human HepG2 cells assessed as receptor transactivation measured by relative luciferase activity at 10 uM by luciferase reporter gene based mammalian2014Bioorganic & medicinal chemistry letters, Sep-01, Volume: 24, Issue:17
Cryptochinones from Cryptocarya chinensis act as farnesoid X receptor agonists.
AID1232961Partial agonist activity at FXR in human HepG2 cells assessed as induction of SHP gene expression at 50 uM after 24 hrs by qRT-PCR analysis (Rvb = 100%)2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Fragmentation of GW4064 led to a highly potent partial farnesoid X receptor agonist with improved drug-like properties.
AID680230TP_TRANSPORTER: uptake of Chenodeoxycholate at 20 uM in Oatp1-expressing HeLa cells2003American journal of physiology. Gastrointestinal and liver physiology, Nov, Volume: 285, Issue:5
Substrate specificities of rat oatp1 and ntcp: implications for hepatic organic anion uptake.
AID680037TP_TRANSPORTER: inhibition of BSP uptake (BSP: 10 uM, CDCA: 200 uM) in Xenopus laevis oocytes1995Gastroenterology, Oct, Volume: 109, Issue:4
Molecular and functional characterization of an organic anion transporting polypeptide cloned from human liver.
AID1124818Activation of FXR (unknown origin) at 10 uM2014Bioorganic & medicinal chemistry, Apr-01, Volume: 22, Issue:7
Some non-conventional biomolecular targets for diamidines. A short survey.
AID1166192Agonist activity at FXR in human HepG2 cells assessed as induction of CYP7A1 mrNA expression at 50 uM by quantitative PCR method2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Extending the structure-activity relationship of anthranilic acid derivatives as farnesoid X receptor modulators: development of a highly potent partial farnesoid X receptor agonist.
AID1686061Drug metabolism in bile of fistula Wistar-Han rat model assessed as area under the curve of the biliary concentration corrected for the bile volume at 60 umol/kg, ID measured at 4 hrs by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID441572Agonist activity at FXR expressed in COS1 cells by cell-based bioluminescence assay2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID303505Activation of SHP gene expression in human HepG2 cells at 25 uM after 18 hrs by RT-PCR2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1609605Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as increase in SHP1 protein expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID404304Effect on human MRP2-mediated estradiol-17-beta-glucuronide transport in Sf9 cells inverted membrane vesicles relative to control2008Journal of medicinal chemistry, Jun-12, Volume: 51, Issue:11
Prediction and identification of drug interactions with the human ATP-binding cassette transporter multidrug-resistance associated protein 2 (MRP2; ABCC2).
AID1451551Transactivation of FXR in human HepG2 cells assessed as PDK4 mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID1059690Agonist activity at wild type TGR5 (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID409878Cytotoxicity against human HCT116 cells after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID680638TP_TRANSPORTER: inhibition of BSP uptake (BSP: 2 uM, CDCA: 100 uM) in Xenopus laevis oocytes1994Hepatology (Baltimore, Md.), Aug, Volume: 20, Issue:2
Functional characterization of the basolateral rat liver organic anion transporting polypeptide.
AID1063186Induction of necrosis in human HT1080 cells up to 500 uM after 24 hrs by annexin V-FITC/propidium iodide staining-based flow cytometric analysis2014Bioorganic & medicinal chemistry, Jan-01, Volume: 22, Issue:1
New highly toxic bile acids derived from deoxycholic acid, chenodeoxycholic acid and lithocholic acid.
AID1609593Agonist activity at FXR in human LO2 cells assessed as decrease in CYP7A1 protein expression at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1193333Agonist activity at human GPBAR1 expressed in HEK293 cells assessed as increase in intracellular cAMP level after 30 mins by cAMP-Glo assay2015Bioorganic & medicinal chemistry, Apr-01, Volume: 23, Issue:7
Stereoselective synthesis, biological evaluation, and modeling of novel bile acid-derived G-protein coupled bile acid receptor 1 (GP-BAR1, TGR5) agonists.
AID659712Agonist activity at human farnesoid X receptor expressed in human HepG2 cells at 5 uM after 24 hrs by dual-luciferase assay relative to control2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Nelumal A, the active principle from Ligularia nelumbifolia, is a novel farnesoid X receptor agonist.
AID441583Cholerectic effect in bile fistula rat model assessed as maximum bile secretion rate at 1 umol/min/kg administered intraduodenally over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID1351286Binding affinity to bovine serum albumin at 10 uM after 24 hrs by equilibrium dialysis method2018European journal of medicinal chemistry, Jan-20, Volume: 144Synthesis, physicochemical properties, and biological activity of bile acids 3-glucuronides: Novel insights into bile acid signalling and detoxification.
AID1079941Liver damage due to vascular disease: peliosis hepatitis, hepatic veno-occlusive disease, Budd-Chiari syndrome. Value is number of references indexed. [column 'VASC' in source]
AID1852829Anticancer activity against human SW480 cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID672332Agonist activity at FXR LBD expressed in human HepG2 cells co-expressing GAL4 DBD and pG5-luc assessed as luciferase activity at 10 uM after 48 hrs by transient transfection reporter assay relative to control2012Bioorganic & medicinal chemistry letters, Jul-15, Volume: 22, Issue:14
Triterpenes from Alisma orientalis act as farnesoid X receptor agonists.
AID72526Effective concentration against Farnesoid X receptor (FXR)2002Journal of medicinal chemistry, Aug-15, Volume: 45, Issue:17
6alpha-ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity.
AID303496Activity at human VP16-FXR assessed as activation of SRC1 interaction domain 2 recruitment at 50 uM by mammalian two-hybrid assay2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID388375Agonist activity at FXR assessed as activation by cell based luciferase transactivation assay at 10 uM2008Bioorganic & medicinal chemistry letters, Oct-15, Volume: 18, Issue:20
Pyrazolidine-3,5-dione derivatives as potent non-steroidal agonists of farnesoid X receptor: virtual screening, synthesis, and biological evaluation.
AID1233559Antiplasmodial activity against Plasmodium falciparum 3D7 assessed as reduction in parasite viability by parasite lactate dehydrogenase assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID1451493Transactivation of FXR in human HepG2 cells assessed as BSEP mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID6737491-Octanol/water partition coefficient, log P of the compound by conventional shake-flask method2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID1412050Antiangiogenic activity in HUVEC assessed as inhibition of endothelial tube formation at 25 uM after overnight incubation by inverted microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1059684Agonist activity at TGR5 N93A mutant (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1686033Critical micelle concentration in 0.15 M NaCl water solution2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID422555Antagonist activity at human FXR transfected in human HuH7 cells co-transfected with human BSEP promoter reporter plasmid at 10 uM after 30 hrs by dual-luciferase reporter assay relative to chenodeoxycholate2009Journal of natural products, Jan, Volume: 72, Issue:1
Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia.
AID1412053Pro-angiogenic activity in Tg(fli:eGFP) zebrafish embryo assessed as increase in branching of sub-intestinal veins at 12.5 uM after 72 hrs post fertilization by fluorescence microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID588214FDA HLAED, liver enzyme composite activity2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID294206Inhibition of FXR in HEK293 cells in presence of CDCA at 100 uM by GAL4 transactivation activity2007Bioorganic & medicinal chemistry, Apr-01, Volume: 15, Issue:7
Design, synthesis, and evaluation of non-steroidal farnesoid X receptor (FXR) antagonist.
AID441581Cholerectic effect in bile fistula rat model assessed as maximum bile secretion rate at 1 umol/min/kg, iv over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID1451530Transactivation of FXR in human HepG2 cells assessed as SHP mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID1166204Agonist activity at FXR in human HepG2 cells assessed as induction of BSEP mrNA expression at 50 uM by quantitative PCR method2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Extending the structure-activity relationship of anthranilic acid derivatives as farnesoid X receptor modulators: development of a highly potent partial farnesoid X receptor agonist.
AID1059678Agonist activity at TGR5 N76A mutant (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID673750Aqueous solubility of the compound in 0.1 M HCl water at pH 3 by HPLC-ES-MS/MS2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID1609607Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as increase in MRP2 protein expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID659715Agonist activity at human farnesoid X receptor expressed in human HepG2 cells at 50 uM after 24 hrs by dual-luciferase assay relative to control2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Nelumal A, the active principle from Ligularia nelumbifolia, is a novel farnesoid X receptor agonist.
AID441580Metabolic stability in bile fistula rat model assessed as conjugated drug level recovered in bile at 1 umol/min/kg administered intraduodenally over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID1609591Agonist activity at FXR in human LO2 cells assessed as increase in MRP2 protein expression at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1609592Agonist activity at FXR in human LO2 cells assessed as increase in SULT2A protein expression at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID324925Agonist activity at human FXR expressed in COS1 cells by luciferase assay2008Journal of medicinal chemistry, Mar-27, Volume: 51, Issue:6
Novel potent and selective bile acid derivatives as TGR5 agonists: biological screening, structure-activity relationships, and molecular modeling studies.
AID624618Specific activity of expressed human recombinant UGT2B42000Annual review of pharmacology and toxicology, , Volume: 40Human UDP-glucuronosyltransferases: metabolism, expression, and disease.
AID635574Agonist activity at FXR in human HepG2 cells assessed as decrease in CYP7A1 mRNA expression at 50 uM after 6 hrs by quantitative PCR analysis2011Bioorganic & medicinal chemistry, Dec-01, Volume: 19, Issue:23
Pharmacophore-based discovery of FXR agonists. Part I: Model development and experimental validation.
AID625295Drug Induced Liver Injury Prediction System (DILIps) validation dataset; compound DILI positive/negative as observed in Pfizer data2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1686052Drug metabolism in bile of fistula Wistar-Han rat model assessed as maximum bile secretion at 60 umol/kg, iv by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID588215FDA HLAED, alkaline phosphatase increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID673745Agonist activity at TGR5 expressed in NCI-H716 cells assessed as cAMP level after 60 mins by FRET analysis relative to 10 uM LCA2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID1686034Surface tension value in 0.15 M NaCl water solution2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID1451541Transactivation of FXR in human HepG2 cells assessed as SREBP1c mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID673751Binding affinity to albumin by equilibrium dialysis2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID409876Cytotoxicity against human GBM cells at 50 uM after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID1609594Agonist activity at FXR in human LO2 cells assessed as decrease in CYP8B1 protein expression at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1079931Moderate liver toxicity, defined via clinical-chemistry results: ALT or AST serum activity 6 times the normal upper limit (N) or alkaline phosphatase serum activity of 1.7 N. Value is number of references indexed. [column 'BIOL' in source]
AID72525Binding affinity for Farnesoid X Receptor (FXR)2003Bioorganic & medicinal chemistry letters, Jun-02, Volume: 13, Issue:11
Binding mode of 6ECDCA, a potent bile acid agonist of the farnesoid X receptor (FXR).
AID54341Sensitivity value towards cyclodextrin conjugated peptide (with pyrene) at 25 degrees Centigrade2003Bioorganic & medicinal chemistry letters, Dec-15, Volume: 13, Issue:24
Fluorescence resonance energy transfer in a novel cyclodextrin-peptide conjugate for detecting steroid molecules.
AID1852832Anticancer activity against human A549 cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID679790TP_TRANSPORTER: inhibition of Daunomycin uptake in canalicular membrane vesicles1994Hepatology (Baltimore, Md.), Jul, Volume: 20, Issue:1 Pt 1
Bile acid inhibition of P-glycoprotein-mediated transport in multidrug-resistant cells and rat liver canalicular membrane vesicles.
AID672341Agonist activity at FXR LBD expressed in human HepG2 cells co-expressing GAL4 DBD and pG5-luc assessed as PLTP promoter-driven luciferase activity at 10 uM after 48 hrs by transient transfection reporter assay relative to control2012Bioorganic & medicinal chemistry letters, Jul-15, Volume: 22, Issue:14
Triterpenes from Alisma orientalis act as farnesoid X receptor agonists.
AID1233565Cytotoxicity against human WI38 cells assessed as inhibition of cell growth at 100 ug/ml by MTT assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID515399Cytotoxicity against human HuH7 cells assessed as cell viability at 500 uM after 24 hrs by MTT assay relative to control2010Bioorganic & medicinal chemistry, Sep-15, Volume: 18, Issue:18
Bile acid toxicity structure-activity relationships: correlations between cell viability and lipophilicity in a panel of new and known bile acids using an oesophageal cell line (HET-1A).
AID303498Activation of human FXR/RXRalpha heterodimer at 50 uM by FXRE TK-luciferase reporter gene assay relative to control2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1277420Cytotoxicity against HEK293T cells assessed as reduction in cell viability after 18 hrs by CellTiter-Glo luminescence assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Synthesis and evaluation of vitamin D receptor-mediated activities of cholesterol and vitamin D metabolites.
AID679473TP_TRANSPORTER: Western in vitro, primary human hepatocyte2001The Journal of biological chemistry, Oct-19, Volume: 276, Issue:42
Disrupted bile acid homeostasis reveals an unexpected interaction among nuclear hormone receptors, transporters, and cytochrome P450.
AID388376Agonist activity at FXR assessed as activation by cell based luciferase transactivation assay2008Bioorganic & medicinal chemistry letters, Oct-15, Volume: 18, Issue:20
Pyrazolidine-3,5-dione derivatives as potent non-steroidal agonists of farnesoid X receptor: virtual screening, synthesis, and biological evaluation.
AID1161667Agonist activity at human GPBAR1 expressed in HEK293T cells assessed as stimulation of cAMP response element-mediated receptor transactivation at 10 uM by luciferase reporter gene assay2014Journal of medicinal chemistry, Sep-25, Volume: 57, Issue:18
Modification on ursodeoxycholic acid (UDCA) scaffold. discovery of bile acid derivatives as selective agonists of cell-surface G-protein coupled bile acid receptor 1 (GP-BAR1).
AID441578Binding affinity to albumin2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID1313848Agonist activity at FXR (unknown origin)2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Novel FXR (farnesoid X receptor) modulators: Potential therapies for cholesterol gallstone disease.
AID1063178Induction of necrosis in human Caco2 cells up to 500 uM after 24 hrs by annexin V-FITC/propidium iodide staining-based flow cytometric analysis2014Bioorganic & medicinal chemistry, Jan-01, Volume: 22, Issue:1
New highly toxic bile acids derived from deoxycholic acid, chenodeoxycholic acid and lithocholic acid.
AID629094Agonist activity at full length mouse FXR/RXRalpha expressed in human HEK293 cells assessed as induction of transcriptional activity after 18 hrs by dual luciferase reporter gene assay2011Bioorganic & medicinal chemistry, Nov-15, Volume: 19, Issue:22
Pharmacophore-based discovery of FXR-agonists. Part II: identification of bioactive triterpenes from Ganoderma lucidum.
AID1609611Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as reduction in NTCP protein expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1609589Agonist activity at FXR in human LO2 cells assessed as increase in SHP1 protein expression at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1412075Increase in wound healing in HUVEC at 12.5 uM after 18 to 24 hrs by phase contrast microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1847971Invivo antagonist activity against FXR in mouse assessed as reduction in Fxr level at 50 uM2022Journal of medicinal chemistry, 10-13, Volume: 65, Issue:19
Discovery of Betulinic Acid Derivatives as Potent Intestinal Farnesoid X Receptor Antagonists to Ameliorate Nonalcoholic Steatohepatitis.
AID592923Induction of FXR-mediated Ostbeta mRNA expression in human HepG2 cells at 1 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID1412072Pro-angiogenic activity in HUVEC assessed as decrease in cell-to-cell contact at 12.5 uM after overnight incubation by FITC-conjugated phalloidin staining based confocal microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1779666Agonist activity at human BLT2 overexpressed in CHO-K1 cells assessed as accumulation of inositol monophosphate measured after 90 mins by HTRF assay2021ACS medicinal chemistry letters, Aug-12, Volume: 12, Issue:8
Discovery of Irbesartan Derivatives as BLT2 Agonists by Virtual Screening.
AID1412049Induction of angiogenesis in HUVEC assessed as increase in number of nodes at 12.5 uM after overnight incubation by inverted microscopic analysis relative to control2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1609587Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as increase in UGT2A4 mRNA expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1686055Drug metabolism in bile of fistula Wistar-Han rat model assessed as maximum biliary secretion at 60 umol/kg, ID by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID240314Effective concentration for recruitment of SRC-1 LxxLL-containing peptide to human Farnesoid X receptor2005Journal of medicinal chemistry, Aug-25, Volume: 48, Issue:17
Farnesoid X receptor: from structure to potential clinical applications.
AID681332TP_TRANSPORTER: inhibition of Taurocholate uptake in ASBT-expressing COS cells1998The American journal of physiology, 01, Volume: 274, Issue:1
Expression and transport properties of the human ileal and renal sodium-dependent bile acid transporter.
AID72531Ligand dependent recruitment of SRC1(676-700) peptide to human Farnesoid X-activated receptor by fluorescence resonance energy transfer assay2000Journal of medicinal chemistry, Aug-10, Volume: 43, Issue:16
Identification of a chemical tool for the orphan nuclear receptor FXR.
AID1233569Antiplasmodial activity against Plasmodium falciparum 3D7 assessed as inhibition of parasite viability at 100 ug/ml by parasite lactate dehydrogenase assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID673744Agonist activity at TGR5 expressed in NCI-H716 cells assessed as cAMP level after 60 mins by FRET analysis2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID1412060Pro-angiogenic activity in Tg(fli:eGFP) zebrafish embryo assessed as sub-intestinal vein baskets extending towards ventral side at 12.5 uM after 72 hrs post fertilization by fluorescence microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1232963Partial agonist activity at FXR in human HepG2 cells assessed as induction of OST-alpha gene expression at 50 uM after 24 hrs by qRT-PCR analysis (Rvb = 100%)2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Fragmentation of GW4064 led to a highly potent partial farnesoid X receptor agonist with improved drug-like properties.
AID1451554Transactivation of FXR in human HepG2 cells assessed as FABP1 mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID588209Literature-mined public compounds from Greene et al multi-species hepatotoxicity modelling dataset2010Chemical research in toxicology, Jul-19, Volume: 23, Issue:7
Developing structure-activity relationships for the prediction of hepatotoxicity.
AID592925Induction of FXR-mediated down-regulation of Cyp7A1 mRNA expression in human HepG2 cells at 20 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID1852825Anticancer activity against human HT-29 cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID1686054Drug metabolism in bile of fistula Wistar-Han rat model assessed as maximum biliary secretion at 60 umol/kg, iv by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID1079947Comments (NB not yet translated). [column 'COMMENTAIRES' in source]
AID1443991Induction of mitochondrial dysfunction in Sprague-Dawley rat liver mitochondria assessed as inhibition of mitochondrial respiration per mg mitochondrial protein measured for 20 mins by A65N-1 oxygen probe based fluorescence assay2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID1412065Increase in VEGFR2 m-RNA expression in HUVEC at 12.5 uM after 12 to 24 hrs by RT-qPCR analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID303494Antagonist activity at human FXR expressed in HEK293 cells assessed as inhibition of GW-4064-induced response at 100 uM by GAL4 activation assay2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1079936Choleostatic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is < 2 (see ACUTE). Value is number of references indexed. [column 'CHOLE' in source]
AID592921Induction of FXR-mediated BSEP mRNA expression in human HepG2 cells at 1 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID592917Agonist activity at human TGR5 receptor expressed in NCI-H716 cells assessed as intracellular cAMP level by TR-FRET assay2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID587939Activation of FXR expressed in human HepG2 cells assessed as dissociation of NCoR from OSTalpha promoter at 50 uM after 18 hrs by chromatin immunoprecipitation2011Journal of medicinal chemistry, Mar-10, Volume: 54, Issue:5
Discovery of sulfated sterols from marine invertebrates as a new class of marine natural antagonists of farnesoid-X-receptor.
AID1166198Agonist activity at FXR in human HepG2 cells assessed as induction of OSTalpha mrNA expression at 50 uM by quantitative PCR method2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Extending the structure-activity relationship of anthranilic acid derivatives as farnesoid X receptor modulators: development of a highly potent partial farnesoid X receptor agonist.
AID1443992Total Cmax in human administered as single dose2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID673748Critical micelle concentration of the compound in 0.15 M NaCl water solution2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID1138165Induction of cAMP production in GP-BAR1-deficient HEK293T cells at 10 uM by cAMP responsive element containing luciferase reporter gene assay2014Journal of medicinal chemistry, Feb-13, Volume: 57, Issue:3
Design, synthesis, and biological evaluation of potent dual agonists of nuclear and membrane bile acid receptors.
AID1327991Antiproliferative activity against human ES2 cells after 48 hrs by SRB assay2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel 3,4-seco bile acid diamides as selective anticancer proliferation and migration agents.
AID422543Reduction in human pancreatic recombinant 1B PLA2 activity expressed in Escherichia coli assessed as hydrolysis of 1,2-dimyristoyl-sn-3-glycerophosphocholine vesicles at 0.03 mol fraction relative to cholate2009Journal of natural products, Jan, Volume: 72, Issue:1
Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia.
AID1079938Chronic liver disease either proven histopathologically, or through a chonic elevation of serum amino-transferase activity after 6 months. Value is number of references indexed. [column 'CHRON' in source]
AID1686015Agonist activity at glutathione transferase-tagged human FXR-LBD using biotinylated Src-1 peptide incubated for 30 mins by recruitment coactivator assay2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID297453Ratio of EC50 for TGR5 expressed in CHO cells to EC50 for FXR expressed in COS1 cells2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.
AID977599Inhibition of sodium fluorescein uptake in OATP1B1-transfected CHO cells at an equimolar substrate-inhibitor concentration of 10 uM2013Molecular pharmacology, Jun, Volume: 83, Issue:6
Structure-based identification of OATP1B1/3 inhibitors.
AID1079933Acute liver toxicity defined via clinical observations and clear clinical-chemistry results: serum ALT or AST activity > 6 N or serum alkaline phosphatases activity > 1.7 N. This category includes cytolytic, choleostatic and mixed liver toxicity. Value is
AID1349338Agonist activity at human FXR assessed as recruitment of SRC1 peptide by TR-FRET assay2017ACS medicinal chemistry letters, Dec-14, Volume: 8, Issue:12
Synthesis and Biological Evaluation of a Series of Bile Acid Derivatives as FXR Agonists for Treatment of NASH.
AID1059685Selectivity ratio of EC50 for TGR5 S270A mutant (unknown origin) to EC50 for wild type TGR5 (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1686060Drug metabolism in bile of fistula Wistar-Han rat model assessed as area under the curve of the biliary concentration corrected for the bile volume at 60 umol/kg, iv measured at 4 hrs by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID303504Activation of SHP gene expression in human Huh7 cells at 20 uM after 18 hrs by RT-PCR2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1059689Selectivity ratio of EC50 for TGR5 E169A mutant (unknown origin) to EC50 for wild type TGR5 (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1412054Pro-angiogenic activity in Tg(fli:eGFP) zebrafish embryo assessed as increase in sprouting and branching of sub-intestinal vessels (after 72 hrs post fertilization by fluorescence microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1609595Agonist activity at FXR in human LO2 cells assessed as decrease in NTCP protein expression at 80 uM after 24 hrs by Western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID409890Cytotoxicity against human HCT116 cells at 25 uM after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID1161670Antagonist activity against FXR (unknown origin) expressed in human HepG2 cells assessed as inhibition of CDCA-induced stimulation of FXR response element IR1-mediated receptor transactivation at 50 uM by luciferase reporter gene assay2014Journal of medicinal chemistry, Sep-25, Volume: 57, Issue:18
Modification on ursodeoxycholic acid (UDCA) scaffold. discovery of bile acid derivatives as selective agonists of cell-surface G-protein coupled bile acid receptor 1 (GP-BAR1).
AID1569579Agonist activity at recombinant human TGR5 expressed in CHO cells assessed as increase in cAMP accumulation after 30 mins by TR-FRET assay relative to control2019Journal of medicinal chemistry, 07-25, Volume: 62, Issue:14
7-Methylation of Chenodeoxycholic Acid Derivatives Yields a Substantial Increase in TGR5 Receptor Potency.
AID702678Agonist activity at GST tagged human recombinant FXRalphaLBD expressed in Escherichia coli BL21 cells assessed as SRC-1 co-activator association after 30 mins by HTRF assay2012Journal of medicinal chemistry, Aug-23, Volume: 55, Issue:16
Discovery and optimization of 1,3,4-trisubstituted-pyrazolone derivatives as novel, potent, and nonsteroidal farnesoid X receptor (FXR) selective antagonists.
AID1412074Induction of angiogenesis in HUVEC assessed as increase in tube length at 12.5 uM after overnight incubation by inverted microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID297450Agonist activity at human TGR5 expressed in CHO cells after 5 hrs by CRE-driven luciferase reporter gene assay2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.
AID1166181Agonist activity at human FXR expressed in human HeLa cells assessed as receptor activation by BSEP promoter-driven firefly luciferase reporter gene assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Extending the structure-activity relationship of anthranilic acid derivatives as farnesoid X receptor modulators: development of a highly potent partial farnesoid X receptor agonist.
AID409891Cytotoxicity against human KMS11 cells at 25 uM after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID672338Agonist activity at FXR LBD expressed in human HepG2 cells co-expressing GAL4 DBD and pG5-luc assessed as transrepression of rat CYP7A1 promoter-driven luciferase activity at 10 uM after 48 hrs by transient transfection reporter assay relative to control2012Bioorganic & medicinal chemistry letters, Jul-15, Volume: 22, Issue:14
Triterpenes from Alisma orientalis act as farnesoid X receptor agonists.
AID1442220Binding affinity to FXR-LBD (unknown origin) assessed as melting temperature at 200 mM by CD spectroscopic method (Rvb = 46 +/- 0.1 degC)2017European journal of medicinal chemistry, Mar-31, Volume: 129Synthesis and biological evaluations of chalcones, flavones and chromenes as farnesoid x receptor (FXR) antagonists.
AID72530Farnesoid X-activated receptor activity compared to 50 uM chenodeoxycholic acid (CDCA)2000Journal of medicinal chemistry, Aug-10, Volume: 43, Issue:16
Identification of a chemical tool for the orphan nuclear receptor FXR.
AID441585Metabolic stability in bile fistula rat model assessed as intact drug level recovered in bile at 1 umol/min/kg, iv over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID1232965Partial agonist activity at FXR in human HT-29 cells assessed as induction of IBABP gene expression at 50 uM after 24 hrs by qRT-PCR analysis (Rvb = 100%)2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Fragmentation of GW4064 led to a highly potent partial farnesoid X receptor agonist with improved drug-like properties.
AID660288Induction of apoptosis in human multinucleated osteoclasts obtained from PBMC assessed as formation of apoptotic nuclear bodies at 10'-5 M after 7 days by Hoechst 33258 staining2012European journal of medicinal chemistry, Jun, Volume: 52Synthesis, characterization and biological activity of hydroxyl-bisphosphonic analogs of bile acids.
AID1546885Agonist activity at FXR (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
The Race to Bash NASH: Emerging Targets and Drug Development in a Complex Liver Disease.
AID1412052Pro-angiogenic activity in Tg(fli:eGFP) zebrafish embryo assessed as increase in sprouting of sub-intestinal veins at 12.5 uM after 72 hrs post fertilization by fluorescence microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID409883Cytotoxicity against human HCT116 cells at 50 uM after 48 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID1446797Agonist activity at FXR (unknown origin) assessed as recruitment of SRC1 peptide to FXR by FRET assay2017Journal of medicinal chemistry, 07-13, Volume: 60, Issue:13
Opportunities and Challenges for Fatty Acid Mimetics in Drug Discovery.
AID680004TP_TRANSPORTER: inhibition of Taurocholate uptake (Taurochorate: 10 uM, CDCA: 200 uM) in Xenopus laevis oocytes1995Gastroenterology, Oct, Volume: 109, Issue:4
Molecular and functional characterization of an organic anion transporting polypeptide cloned from human liver.
AID16860351-octanol-water partition coefficient, logP of compound as ionized species2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID1686058Drug metabolism in bile of fistula Wistar-Han rat model assessed as taurine conjugate formation at 60 umol/kg, iv by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID1277417Antagonist activity against VP16 tagged-VDR-LBD (unknown origin) expressed in HEK293T cells assessed as inhibition of 1,25-dihydroxyvitamin D3-induced SRC1 coactivator peptide recruitment after 16 hrs by luciferase reporter gene based two hybrid assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Synthesis and evaluation of vitamin D receptor-mediated activities of cholesterol and vitamin D metabolites.
AID1686036Binding affinity to human serum albumin2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID1079949Proposed mechanism(s) of liver damage. [column 'MEC' in source]
AID269847Agonist activity at FXR in HepG2 cells by luciferase activity relative to DMSO at 10 uM2006Bioorganic & medicinal chemistry letters, Aug-15, Volume: 16, Issue:16
Identification of a lead pharmacophore for the development of potent nuclear receptor modulators as anticancer and X syndrome disease therapeutic agents.
AID1351284Octanol-phosphate buffer distribution coefficient, log P of the compound at pH 7.4 at 1 uM after 24 hrs by shake flask method2018European journal of medicinal chemistry, Jan-20, Volume: 144Synthesis, physicochemical properties, and biological activity of bile acids 3-glucuronides: Novel insights into bile acid signalling and detoxification.
AID1852828Anticancer activity against human LoVo cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID303492Activation of human PXR expressed in HEK293 cells at 20 uM by GAL4 activation assay relative to control2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1412040Toxicity in zebrafish embryo assessed as reduction in embryo survival up to 50 uM2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1443906Antibacterial activity against Clostridium difficile NAP1 assessed as concentration required for complete inhibition of TCA-induced spore germination measured after 20 mins under anaerobic condition2017Journal of medicinal chemistry, 04-27, Volume: 60, Issue:8
Synthesis and Biological Evaluation of Bile Acid Analogues Inhibitory to Clostridium difficile Spore Germination.
AID1059686Selectivity ratio of EC50 for TGR5 Y89F mutant (unknown origin) to EC50 for wild type TGR5 (unknown origin)2013ACS medicinal chemistry letters, Dec-12, Volume: 4, Issue:12
Probing the Binding Site of Bile Acids in TGR5.
AID1327989Antiproliferative activity against human PC3M cells after 48 hrs by SRB assay2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel 3,4-seco bile acid diamides as selective anticancer proliferation and migration agents.
AID1633314Agonist activity at GST-tagged FXR-LBD (unknown origin) assessed as biotin-labeled SRC-1 recruitment at 5 uM after 30 mins by Alpha Screen assay relative to 6-ECDCA2019ACS medicinal chemistry letters, Apr-11, Volume: 10, Issue:4
Novel Isoxazole Derivatives with Potent FXR Agonistic Activity Prevent Acetaminophen-Induced Liver Injury.
AID1079948Times to onset, minimal and maximal, observed in the indexed observations. [column 'DELAI' in source]
AID1451534Transactivation of FXR in human HepG2 cells assessed as CYP7A1 mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID1609609Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as reduction in CYP7A1 protein expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID490122Cytotoxicity against human GBM cells after 24 hrs by neutral red uptake assay2010European journal of medicinal chemistry, Jul, Volume: 45, Issue:7
Synthesis of bile acid derivatives and in vitro cytotoxic activity with pro-apoptotic process on multiple myeloma (KMS-11), glioblastoma multiforme (GBM), and colonic carcinoma (HCT-116) human cell lines.
AID659713Agonist activity at human farnesoid X receptor expressed in human HepG2 cells at 10 uM after 24 hrs by dual-luciferase assay relative to control2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Nelumal A, the active principle from Ligularia nelumbifolia, is a novel farnesoid X receptor agonist.
AID662693Agonist activity at human recombinant FXR expressed in HEK293 cells coexpressing CMX-GAL4N by luciferase reporter gene assay relative to GW40642012Bioorganic & medicinal chemistry letters, Jun-15, Volume: 22, Issue:12
E297G mutated bile salt export pump (BSEP) function enhancers derived from GW4064: structural development study and separation from farnesoid X receptor-agonistic activity.
AID1569580Agonist activity at recombinant human TGR5 expressed in CHO cells assessed as increase in cAMP accumulation after 30 mins by TR-FRET assay2019Journal of medicinal chemistry, 07-25, Volume: 62, Issue:14
7-Methylation of Chenodeoxycholic Acid Derivatives Yields a Substantial Increase in TGR5 Receptor Potency.
AID592924Induction of FXR-mediated down-regulation of Cyp7A1 mRNA expression in human HepG2 cells at 1 uM by RT-PCR2011Bioorganic & medicinal chemistry, Apr-15, Volume: 19, Issue:8
Extending SAR of bile acids as FXR ligands: discovery of 23-N-(carbocinnamyloxy)-3α,7α-dihydroxy-6α-ethyl-24-nor-5β-cholan-23-amine.
AID1569578Cytotoxicity against CHO cells expressing human TGR5 at 30 to 100 uM2019Journal of medicinal chemistry, 07-25, Volume: 62, Issue:14
7-Methylation of Chenodeoxycholic Acid Derivatives Yields a Substantial Increase in TGR5 Receptor Potency.
AID1063187Cytotoxicity against human HT1080 cells assessed as cell viability after 24 hrs by MTT assay2014Bioorganic & medicinal chemistry, Jan-01, Volume: 22, Issue:1
New highly toxic bile acids derived from deoxycholic acid, chenodeoxycholic acid and lithocholic acid.
AID409881Cytotoxicity against human KMS11 cells at 50 uM after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID1458345Lipophilicity, logD of compound at pH 7.4 by LC-MS analysis2017Journal of medicinal chemistry, 08-24, Volume: 60, Issue:16
Nonacidic Farnesoid X Receptor Modulators.
AID297452Agonist activity at human FXR expressed in COS1 cells after 5 hrs by CRE-driven luciferase reporter gene assay relative to 6ECDCA2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.
AID297451Agonist activity at human FXR expressed in COS1 cells after 5 hrs by CRE-driven luciferase reporter gene assay2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.
AID1351283Solubility of the compound in pH 1 HCl solution at 300 uM after 24 hrs by HPLC-ESI-MS analysis2018European journal of medicinal chemistry, Jan-20, Volume: 144Synthesis, physicochemical properties, and biological activity of bile acids 3-glucuronides: Novel insights into bile acid signalling and detoxification.
AID659710Agonist activity at human farnesoid X receptor expressed in human HepG2 cells at 0.1 uM after 24 hrs by dual-luciferase assay relative to control2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Nelumal A, the active principle from Ligularia nelumbifolia, is a novel farnesoid X receptor agonist.
AID490121Cytotoxicity against human KMS11 cells after 24 hrs by neutral red uptake assay2010European journal of medicinal chemistry, Jul, Volume: 45, Issue:7
Synthesis of bile acid derivatives and in vitro cytotoxic activity with pro-apoptotic process on multiple myeloma (KMS-11), glioblastoma multiforme (GBM), and colonic carcinoma (HCT-116) human cell lines.
AID267784Binding affinity to FXR assessed as ligand-dependent SRC1 recruitment by FRET based co-activator assay2006Journal of medicinal chemistry, Jul-13, Volume: 49, Issue:14
Back door modulation of the farnesoid X receptor: design, synthesis, and biological evaluation of a series of side chain modified chenodeoxycholic acid derivatives.
AID1686032Solubility in water measured for protonated species2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID1079942Steatosis, proven histopathologically. Value is number of references indexed. [column 'STEAT' in source]
AID1412070Pro-angiogenic activity in HUVEC assessed as stress fiber formation at 12.5 uM after overnight incubation by FITC-conjugated phalloidin staining based confocal microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID324926Agonist activity against human FXR expressed in COS1 cells by luciferase assay relative to 6-enantiomeric chenodeoxy cholic acid2008Journal of medicinal chemistry, Mar-27, Volume: 51, Issue:6
Novel potent and selective bile acid derivatives as TGR5 agonists: biological screening, structure-activity relationships, and molecular modeling studies.
AID1609606Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as increase in BSEP protein expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID587931Agonist activity at FXR expressed in human HepG2 cells assessed as beta-galactosidase activity at 10 uM after 18 hrs by luminometric analysis2011Journal of medicinal chemistry, Mar-10, Volume: 54, Issue:5
Discovery of sulfated sterols from marine invertebrates as a new class of marine natural antagonists of farnesoid-X-receptor.
AID682019TP_TRANSPORTER: inhibition of Taurocholate uptake (Taurocholate: 0.72 uM, CDCA: 72 uM) in Ntcp-expressing COS-7 cells2002Bioscience, biotechnology, and biochemistry, May, Volume: 66, Issue:5
Characterization of cloned mouse Na+/taurocholate cotransporting polypeptide by transient expression in COS-7 cells.
AID1412059Pro-angiogenic activity in Tg(fli:eGFP) zebrafish embryo assessed as arcades formation at 12.5 uM after 72 hrs post fertilization by fluorescence microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID588213Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in non-rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID588212Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1079945Animal toxicity known. [column 'TOXIC' in source]
AID1719697Agonist activity at FXR in human HepG2 cells assessed as increase in BSEP mRNA expression at 30 uM incubated for 8 hrs by qRT-PCR analysis2021ACS medicinal chemistry letters, Feb-11, Volume: 12, Issue:2
A New FXR Ligand Chemotype with Agonist/Antagonist Switch.
AID1686057Drug metabolism in bile of fistula Wistar-Han rat model assessed as maximum biliary secretion of the administered compound including the secreted taurine conjugates at 60 umol/kg, ID by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID629057Agonist activity at FXR in human HepG2 cells assessed as inhibition of CYP7A1 mRNA expression at 50 uM for 6 hrs by RT-PCR relative to control2011Bioorganic & medicinal chemistry, Nov-15, Volume: 19, Issue:22
Pharmacophore-based discovery of FXR-agonists. Part II: identification of bioactive triterpenes from Ganoderma lucidum.
AID681352TP_TRANSPORTER: transepithelial transport (basal to apical) in Ntcp/Bsep double transfected MDCK cell2005American journal of physiology. Gastrointestinal and liver physiology, Jan, Volume: 288, Issue:1
Vectorial transport of bile salts across MDCK cells expressing both rat Na+-taurocholate cotransporting polypeptide and rat bile salt export pump.
AID303508Decrease in CYP7A1 gene expression in human HepG2 cells at 25 uM after 18 hrs by RT-PCR2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID303500Activation of BSEP gene expression in human Huh7 cells at 20 uM after 18 hrs by RT-PCR2007Journal of medicinal chemistry, Nov-29, Volume: 50, Issue:24
Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids.
AID1686053Drug metabolism in bile of fistula Wistar-Han rat model assessed as maximum bile secretion at 60 umol/kg, ID by HPLC-ES-MS/MS analysis2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID1277414Antagonist activity against VDR-LBD (unknown origin) expressed in Escherichia coli assessed as inhibition of VDR agonist LG190178-induced SRC2-3 coactivator peptide recruitment after 30 mins by fluorescence polarization assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Synthesis and evaluation of vitamin D receptor-mediated activities of cholesterol and vitamin D metabolites.
AID441584Cholerectic effect in bile fistula rat model assessed as maximum bile acids secretion rate per kg body weight at 1 umol/min/kg administered intraduodenally over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID409877Cytotoxicity against human GBM cells after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID324927Selectivity index, ratio of EC50 for human TGR5 to EC50 for human FXR2008Journal of medicinal chemistry, Mar-27, Volume: 51, Issue:6
Novel potent and selective bile acid derivatives as TGR5 agonists: biological screening, structure-activity relationships, and molecular modeling studies.
AID1233567Antitrypanosomal activity against Trypanosoma brucei brucei Lister 427 bloodstream forms assessed as inhibition of parasite growth at 100 ug/ml by microtiter plate based assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID1412073Cytotoxicity against HUVEC assessed as reduction in cell viability at 6.25 to 100 uM after 24 hrs by MTT assay2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1852827Anticancer activity against human RKO cells assessed as inhibition of cell proliferation by MTT assay2022RSC medicinal chemistry, Nov-16, Volume: 13, Issue:11
Design and synthesis of bile acid derivatives and their activity against colon cancer.
AID1609585Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as increase in SHP1 mRNA expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1063189Cytotoxicity against human Caco2 cells assessed as cell viability after 24 hrs by MTT assay2014Bioorganic & medicinal chemistry, Jan-01, Volume: 22, Issue:1
New highly toxic bile acids derived from deoxycholic acid, chenodeoxycholic acid and lithocholic acid.
AID1165586Agonist activity at FXR (unknown origin) expressed in human HepG2 cells assessed as receptor transactivation at 10 uM incubated for 16 hrs by FXR response element driven HSP27-TK-luciferase reporter gene assay2014Journal of medicinal chemistry, Oct-23, Volume: 57, Issue:20
Exploitation of cholane scaffold for the discovery of potent and selective farnesoid X receptor (FXR) and G-protein coupled bile acid receptor 1 (GP-BAR1) ligands.
AID1412069Decrease in MMP9 level in HUVEC at 12.5 uM after 24 hrs by ELISA2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID645565Agonist activity at FXR in human HepG2 cells at 10 uM after 48 hrs by luciferase reporter gene based transactivation assay2012Journal of medicinal chemistry, Jan-12, Volume: 55, Issue:1
Conicasterol E, a small heterodimer partner sparing farnesoid X receptor modulator endowed with a pregnane X receptor agonistic activity, from the marine sponge Theonella swinhoei.
AID324923Agonist activity at human TGR5 expressed in CHO cells by luciferase assay2008Journal of medicinal chemistry, Mar-27, Volume: 51, Issue:6
Novel potent and selective bile acid derivatives as TGR5 agonists: biological screening, structure-activity relationships, and molecular modeling studies.
AID409889Cytotoxicity against human GBM cells at 25 uM after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID1166182Agonist activity at human FXR expressed in human HeLa cells assessed as receptor activation by BSEP promoter-driven firefly luciferase reporter gene assay relative to 3 uM GW40642014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Extending the structure-activity relationship of anthranilic acid derivatives as farnesoid X receptor modulators: development of a highly potent partial farnesoid X receptor agonist.
AID1451544Transactivation of FXR in human HepG2 cells assessed as FAS mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID1609544Agonist activity at FXR in human LO2 cells assessed as increase in BSEP mRNA expression at 80 uM after 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID422556Enhancement of bile salt export pump promoter transactivation2009Journal of natural products, Jan, Volume: 72, Issue:1
Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia.
AID673746Agonist activity at GST-tagged FXR-LBD using biotinylated-SRC-1 peptide as substrate preincubated with compound for 30 mins measured after 4 hrs2012ACS medicinal chemistry letters, Apr-12, Volume: 3, Issue:4
Avicholic Acid: A Lead Compound from Birds on the Route to Potent TGR5 Modulators.
AID1811597Inhibition of Taurocholate-induced spore germination in Clostridioides difficile UK1 at 100 mM by spectrophotometric method2021Bioorganic & medicinal chemistry, 12-15, Volume: 52Studies on the Importance of the 7α-, and 12α- hydroxyl groups of N-Aryl-3α,7α,12α-trihydroxy-5β-cholan-24-amides on their Antigermination Activity Against a Hypervirulent Strain of Clostridioides (Clostridium) difficile.
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID629055Agonist activity at FXR2011Bioorganic & medicinal chemistry, Nov-15, Volume: 19, Issue:22
Pharmacophore-based discovery of FXR-agonists. Part II: identification of bioactive triterpenes from Ganoderma lucidum.
AID1351282Solubility of the compound in pH 7.4 PBS buffer at 300 uM after 24 hrs by HPLC-ESI-MS analysis2018European journal of medicinal chemistry, Jan-20, Volume: 144Synthesis, physicochemical properties, and biological activity of bile acids 3-glucuronides: Novel insights into bile acid signalling and detoxification.
AID659711Agonist activity at human farnesoid X receptor expressed in human HepG2 cells at 1 uM after 24 hrs by dual-luciferase assay relative to control2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Nelumal A, the active principle from Ligularia nelumbifolia, is a novel farnesoid X receptor agonist.
AID72527Relative recruitment of SRC1 peptide to Farnesoid X receptor (FXR) where CDCA=100%2002Journal of medicinal chemistry, Aug-15, Volume: 45, Issue:17
6alpha-ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity.
AID441582Cholerectic effect in bile fistula rat model assessed as maximum bile acids secretion rate per kg body weight at 1 umol/min/kg, iv over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID422544Reduction in human pancreatic recombinant 1B PLA2 activity expressed in Escherichia coli assessed as hydrolysis of 1,2-dimyristoyl-sn-3-glycerophosphocholine vesicles at 0.007 mol fraction relative to cholate2009Journal of natural products, Jan, Volume: 72, Issue:1
Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia.
AID1412043Cytotoxicity against HUVEC assessed as reduction in cell death up to 12.5 uM after 24 hrs by MTT assay2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1451537Transactivation of FXR in human HepG2 cells assessed as PPARalpha mRNA expression at 50 uM after 8 to 16 hrs by SYBR green based qRT-PCR analysis relative to DMSO control2017Journal of medicinal chemistry, 09-28, Volume: 60, Issue:18
A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis.
AID1351285Octanol-HCl distribution coefficient, log P of the compound at pH 1 at 1 uM after 24 hrs by shake flask method2018European journal of medicinal chemistry, Jan-20, Volume: 144Synthesis, physicochemical properties, and biological activity of bile acids 3-glucuronides: Novel insights into bile acid signalling and detoxification.
AID409884Cytotoxicity against human KMS11 cells at 50 uM after 48 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID1277413Agonist activity at VDR-LBD (unknown origin) expressed in Escherichia coli assessed as SRC2-3 coactivator peptide recruitment after 30 mins by fluorescence polarization assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Synthesis and evaluation of vitamin D receptor-mediated activities of cholesterol and vitamin D metabolites.
AID1719696Agonist activity at FXR in human HepG2 cells assessed as increase in SHP mRNA expression at 30 uM incubated for 8 hrs by qRT-PCR analysis2021ACS medicinal chemistry letters, Feb-11, Volume: 12, Issue:2
A New FXR Ligand Chemotype with Agonist/Antagonist Switch.
AID409879Cytotoxicity against human KMS11 cells after 24 hrs by neutral red uptake assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
New lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative and pro-apoptotic effects on human cancer cell lines.
AID441577Critical micellar concentration in 0.15 M NaCl water solution2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID1232939Agonist activity at human full length FXR expressed in HeLa cells cotransfected with pSG5-human RXR after 24 hrs by Dual-Glo luciferase reporter gene assay relative to 3 uM GW40642015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Fragmentation of GW4064 led to a highly potent partial farnesoid X receptor agonist with improved drug-like properties.
AID515400Cytotoxicity against human HET-1A cells assessed as cell viability at 500 uM after 24 hrs by MTT assay relative to control2010Bioorganic & medicinal chemistry, Sep-15, Volume: 18, Issue:18
Bile acid toxicity structure-activity relationships: correlations between cell viability and lipophilicity in a panel of new and known bile acids using an oesophageal cell line (HET-1A).
AID1609610Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as reduction in CYP8B1 protein expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by western blot analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1412076Pro-angiogenic activity in Tg(fli:eGFP) zebrafish embryo at 6.25 uM after 72 hrs post fertilization by fluorescence microscopic analysis2017MedChemComm, Dec-01, Volume: 8, Issue:12
Deciphering the role of hydrophobic and hydrophilic bile acids in angiogenesis using
AID1686018Agonist activity at TGR5 in human NCI-H716 cells assessed as stimulation of intracellular cAMP accumulation incubated for 60 mins by HTR-FRET assay relative to 10 uM LCA2016Journal of medicinal chemistry, Oct-13, Volume: 59, Issue:19
Discovery of 3α,7α,11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid (TC-100), a Novel Bile Acid as Potent and Highly Selective FXR Agonist for Enterohepatic Disorders.
AID977602Inhibition of sodium fluorescein uptake in OATP1B3-transfected CHO cells at an equimolar substrate-inhibitor concentration of 10 uM2013Molecular pharmacology, Jun, Volume: 83, Issue:6
Structure-based identification of OATP1B1/3 inhibitors.
AID1187920Agonist activity at GAL4 DNA binding domain tagged FXR ligand binding domain (unknown origin) expressed in human HepG2 cells co-expressing rat CYP7A1 gene promoter assessed as transrepression of CYP7A1 promoter driven luciferase activity at 10 uM2014Bioorganic & medicinal chemistry letters, Sep-01, Volume: 24, Issue:17
Cryptochinones from Cryptocarya chinensis act as farnesoid X receptor agonists.
AID635573Agonist activity at mouse FXR expressed in HEK293 cells co-expressing mouse RXRalpha and ECRE-luc by luciferase reporter gene assay2011Bioorganic & medicinal chemistry, Dec-01, Volume: 19, Issue:23
Pharmacophore-based discovery of FXR agonists. Part I: Model development and experimental validation.
AID324924Agonist activity at human TGR5 expressed in CHO cells by luciferase assay relative to lithocholic acid2008Journal of medicinal chemistry, Mar-27, Volume: 51, Issue:6
Novel potent and selective bile acid derivatives as TGR5 agonists: biological screening, structure-activity relationships, and molecular modeling studies.
AID662692Agonist activity at human recombinant FXR expressed in HEK293 cells coexpressing CMX-GAL4N by luciferase reporter gene assay2012Bioorganic & medicinal chemistry letters, Jun-15, Volume: 22, Issue:12
E297G mutated bile salt export pump (BSEP) function enhancers derived from GW4064: structural development study and separation from farnesoid X receptor-agonistic activity.
AID441579Metabolic stability in bile fistula rat model assessed as intact drug level recovered in bile at 1 umol/min/kg administered intraduodenally over 1 hr2009Journal of medicinal chemistry, Dec-24, Volume: 52, Issue:24
Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.
AID490123Cytotoxicity against human HCT116 cells after 24 hrs by neutral red uptake assay2010European journal of medicinal chemistry, Jul, Volume: 45, Issue:7
Synthesis of bile acid derivatives and in vitro cytotoxic activity with pro-apoptotic process on multiple myeloma (KMS-11), glioblastoma multiforme (GBM), and colonic carcinoma (HCT-116) human cell lines.
AID1847972Invivo antagonist activity against FXR in mouse assessed as reduction in Fgf15 level at 50 uM2022Journal of medicinal chemistry, 10-13, Volume: 65, Issue:19
Discovery of Betulinic Acid Derivatives as Potent Intestinal Farnesoid X Receptor Antagonists to Ameliorate Nonalcoholic Steatohepatitis.
AID679670TP_TRANSPORTER: Northern blot from HepG2 cells2002The Journal of biological chemistry, Jan-25, Volume: 277, Issue:4
Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor.
AID1609586Agonist activity at FXR in guggulsterone-treated human LO2 cells assessed as increase in BSEP mRNA expression at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID1234672Antiparasitic activity against Trypanosoma cruzi strain CL Brener epimastigotes at 300 uM after 12 days by ELISA reader2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Enzymatic synthesis of bile acid derivatives and biological evaluation against Trypanosoma cruzi.
AID1138171Agonist activity at FXR in human HepG2 cells assessed as upregulation of OST-alpha mRNA expression at 10 uM after 18 hrs by RT-PCR analysis2014Journal of medicinal chemistry, Feb-13, Volume: 57, Issue:3
Design, synthesis, and biological evaluation of potent dual agonists of nuclear and membrane bile acid receptors.
AID659714Agonist activity at human farnesoid X receptor expressed in human HepG2 cells at 25 uM after 24 hrs by dual-luciferase assay relative to control2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Nelumal A, the active principle from Ligularia nelumbifolia, is a novel farnesoid X receptor agonist.
AID1443995Hepatotoxicity in human assessed as drug-induced liver injury2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID422553Binding affinity to pig PLA2 at 100 uM by saturation transfer difference NMR spectroscopy2009Journal of natural products, Jan, Volume: 72, Issue:1
Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia.
AID657097Restoration of human BSEP E297G mutant trafficking to cell membrane in MDCK2 cells co-expressing NTCP assessed as accumulation of [3H]taurocholate at 10 uM preincubated for 24 hrs prior incubation with [3H]taurocholate measured after 2 hrs by liquid scint2012Bioorganic & medicinal chemistry, May-01, Volume: 20, Issue:9
Discovery and structural development of small molecules that enhance transport activity of bile salt export pump mutant associated with progressive familial intrahepatic cholestasis type 2.
AID659716Agonist activity at human farnesoid X receptor expressed in human HepG2 cells at 100 uM after 24 hrs by dual-luciferase assay relative to control2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Nelumal A, the active principle from Ligularia nelumbifolia, is a novel farnesoid X receptor agonist.
AID660287Inhibition of osteoclastogenesis in human osteoclasts obtained from PBMC assessed as formation of TRAPC positive multinucleated cells at 10'-5 M after 7 days2012European journal of medicinal chemistry, Jun, Volume: 52Synthesis, characterization and biological activity of hydroxyl-bisphosphonic analogs of bile acids.
AID682147TP_TRANSPORTER: inhibition of Taurocholate uptake (Taurocholate: 5 uM, CDCA: 100 uM) in Ntcp-expressing COS-7 cells1994The American journal of physiology, Mar, Volume: 266, Issue:3 Pt 1
Expression and characterization of a functional rat liver Na+ bile acid cotransport system in COS-7 cells.
AID1609584Increase in FXR mRNA expression in guggulsterone-treated human LO2 cells at 80 uM treated with FXR antagonist, guggulsterone for 24 hrs followed by incubation with compound for 24 hrs by RT-qPCR analysis2019European journal of medicinal chemistry, Nov-15, Volume: 182Highly potent non-steroidal FXR agonists protostane-type triterpenoids: Structure-activity relationship and mechanism.
AID679712TP_TRANSPORTER: inhibition of Taurocholate uptake (Taurocholate: 0.72 uM, CDCA: a 100-fold excess) in isolated mouse ileocytes1999Journal of biochemistry, Apr, Volume: 125, Issue:4
Characterization, cDNA cloning, and functional expression of mouse ileal sodium-dependent bile acid transporter.
AID243397Efficacy for human Farnesoid X receptor in FRET assay2004Journal of medicinal chemistry, Aug-26, Volume: 47, Issue:18
Bile acid derivatives as ligands of the farnesoid X receptor. Synthesis, evaluation, and structure-activity relationship of a series of body and side chain modified analogues of chenodeoxycholic acid.
AID1233553Antitrypanosomal activity against Trypanosoma brucei brucei Lister 427 bloodstream forms by microtiter plate based assay2015European journal of medicinal chemistry, Jul-15, Volume: 100Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
AID540299A screen for compounds that inhibit the MenB enzyme of Mycobacterium tuberculosis2010Bioorganic & medicinal chemistry letters, Nov-01, Volume: 20, Issue:21
Synthesis and SAR studies of 1,4-benzoxazine MenB inhibitors: novel antibacterial agents against Mycobacterium tuberculosis.
AID588519A screen for compounds that inhibit viral RNA polymerase binding and polymerization activities2011Antiviral research, Sep, Volume: 91, Issue:3
High-throughput screening identification of poliovirus RNA-dependent RNA polymerase inhibitors.
AID1799080HTRF Coactivator Recruitment Assay from Article 10.1073/pnas.0710981105: \\Identification of a potent synthetic FXR agonist with an unexpected mode of binding and activation.\\2008Proceedings of the National Academy of Sciences of the United States of America, Apr-08, Volume: 105, Issue:14
Identification of a potent synthetic FXR agonist with an unexpected mode of binding and activation.
AID1798108TGR5 Activity Assay from Article 10.1021/jm070633p: \\Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.\\2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.
AID1798109FXR Activity Assay from Article 10.1021/jm070633p: \\Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.\\2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5.
AID1798115Cotransfection Assay from Article 10.1016/j.bmc.2007.01.046: \\Design, synthesis, and evaluation of non-steroidal farnesoid X receptor (FXR) antagonist.\\2007Bioorganic & medicinal chemistry, Apr-01, Volume: 15, Issue:7
Design, synthesis, and evaluation of non-steroidal farnesoid X receptor (FXR) antagonist.
AID1159607Screen for inhibitors of RMI FANCM (MM2) intereaction2016Journal of biomolecular screening, Jul, Volume: 21, Issue:6
A High-Throughput Screening Strategy to Identify Protein-Protein Interaction Inhibitors That Block the Fanconi Anemia DNA Repair Pathway.
AID1159550Human Phosphogluconate dehydrogenase (6PGD) Inhibitor Screening2015Nature cell biology, Nov, Volume: 17, Issue:11
6-Phosphogluconate dehydrogenase links oxidative PPP, lipogenesis and tumour growth by inhibiting LKB1-AMPK signalling.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (3,200)

TimeframeStudies, This Drug (%)All Drugs %
pre-19901604 (50.13)18.7374
1990's460 (14.38)18.2507
2000's345 (10.78)29.6817
2010's582 (18.19)24.3611
2020's209 (6.53)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials227 (6.79%)5.53%
Reviews304 (9.09%)6.00%
Case Studies151 (4.52%)4.05%
Observational7 (0.21%)0.25%
Other2,654 (79.39%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (34)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Study of the Effects of Obeticholic Acid on Farnesoid X Receptor Expression in Jejunum and on Gut Microbiota in Morbidly Obese Patients and Healthy Volunteers[NCT02532335]Phase 140 participants (Anticipated)Interventional2015-08-31Recruiting
Effect of Obeticholic Acid (INT-747, Intercept) on the Hepatobiliary Transport of Bile Acids in Patients With PBC Examined by 11C-cholyl-sarcosine PET/CT[NCT03253276]Early Phase 18 participants (Actual)Interventional2016-05-19Completed
A Phase 3 Study to Evaluate the Effects of Chenodeoxycholic Acid in Adult and Pediatric Patients With Cerebrotendinous Xanthomatosis[NCT04270682]Phase 318 participants (Actual)Interventional2020-01-31Completed
Obeticholic Acid Treatment in Patients With Bile Acid Diarrhoea: an Open-label, Pilot Study of Mechanisms, Safety and Symptom Response.[NCT01585025]Phase 235 participants (Actual)Interventional2012-04-30Completed
Therapeutic Metabolic Intervention in Patients With Spastic Paraplegia SPG5[NCT02314208]Phase 212 participants (Actual)Interventional2015-01-31Completed
Changes in Bile Acid Homeostasis and Stool Habits After Cholecystectomy[NCT03168555]Phase 423 participants (Actual)Interventional2017-06-22Completed
A Phase 2a, Double-Blind, Randomized, Active Controlled, Parallel Group Study Evaluating the Efficacy, Safety, and Tolerability of Bezafibrate Administered in Combination With Obeticholic Acid in Subjects With Primary Biliary Cholangitis[NCT05239468]Phase 260 participants (Anticipated)Interventional2022-03-21Active, not recruiting
Retrospective Cohort Study to Investigate the Safety and Efficacy of Chenodeoxycholic Acid (CDCA) in Patients Affected by Cerebrotendinous Xanthomatosis (CTX)[NCT05499026]28 participants (Actual)Observational2014-12-09Completed
Phase 2 Study of Obeticholic Acid for Lipodystrophy Patients[NCT02430077]Phase 210 participants (Actual)Interventional2016-06-30Completed
A Multicenter, Open-Label, Single- and Multiple-Dose, Dose-Finding Study, With an Optional Open-Label Extension to Assess the Safety, Tolerability, and Pharmacokinetics of Obeticholic Acid in Pediatric Subjects With Biliary Atresia[NCT05321524]Phase 27 participants (Actual)Interventional2015-07-01Terminated(stopped due to After extensive efforts to improve recruitment, it is deemed not feasible to enroll the requisite number of subjects to generate data needed to meet the study objectives. EMA Paediatric Committee agreed with the Sponsor to terminate this study.)
Randomized, Two-way, Two-period, Single Oral Dose, Open-label, Crossover, Bioequivalence Study to Compare Chenodeoxycholic Acid Capsules (250mg Chenodeoxycholic Acid) [Dose: 1 x 02 Capsules] Versus Chenodeoxycholic Acid Leadiant 250 mg Hard Capsules (250m[NCT06180057]Phase 124 participants (Actual)Interventional2022-06-12Completed
A Phase 3, Double-Blind, Randomized, Placebo-Controlled, Multicenter Study to Evaluate the Efficacy and Safety of Obeticholic Acid in Subjects With Compensated Cirrhosis Due to Nonalcoholic Steatohepatitis[NCT03439254]Phase 3919 participants (Actual)Interventional2017-08-30Completed
An Open-Label, Single-Dose Trial to Assess the Effects of Hepatic Impairment on the Pharmacokinetics of Obeticholic Acid (OCA)[NCT01904539]Phase 132 participants (Actual)Interventional2013-06-30Completed
An Open Label Trial to Assess the Effects of Food on the Pharmacokinetic Parameters of Obeticholic Acid (OCA)[NCT01914562]Phase 132 participants (Actual)Interventional2013-08-31Completed
An Open Label, Randomized, Single Dose and Multiple Dose Trial to Assess the Pharmacokinetics of Obeticholic Acid (OCA)[NCT01933503]Phase 124 participants (Actual)Interventional2013-10-31Completed
The Farnesoid X Receptor (FXR) Ligand Obeticholic Acid in Nonalcoholic Steatohepatitis (NASH) Treatment (FLINT) Trial[NCT01265498]Phase 2283 participants (Actual)Interventional2011-03-31Completed
A Study of INT-747 (6-ECDCA) Monotherapy in Patients With Primary Biliary Cirrhosis[NCT00570765]Phase 260 participants (Actual)Interventional2008-01-17Completed
Effects of Chenodeoxycholic Acid and Colesevelam on GLP-1 Secretion, During a Meal, After Roux-en-Y Gastric Bypass[NCT02876484]Phase 412 participants (Actual)Interventional2016-06-30Completed
A Phase 3, Double-Blind, Placebo-Controlled Trial and Long-Term Safety Extension of Obeticholic Acid in Patients With Primary Biliary Cirrhosis[NCT01473524]Phase 3217 participants (Actual)Interventional2012-01-31Completed
Effect of Bile Acids and Bile Acid Sequstrants on GLP-1 Secretion After Roux-en-Y Gastric Bypass[NCT02952963]Phase 48 participants (Actual)Interventional2016-10-31Completed
Effects of Activation of the Farnesoid X Receptor (FXR) on Hepatic Lipid and Glucose Metabolism in Patients With the Metabolic Syndrome and Familial Forms of Hypertriglyceridemia[NCT00465751]Early Phase 130 participants (Actual)Interventional2004-10-31Completed
Biologic Significance of Cholestanol in Man[NCT00018694]0 participants (Actual)Interventional1999-10-31Withdrawn
Effect of Chenodeoxycholic Acid on Gastrointestinal Transit and Colonic Functions in Health and Constipation-predominant Irritable Bowel Syndrome (IBS-C)[NCT00912301]Phase 236 participants (Actual)Interventional2009-04-30Completed
Effects of Obeticholic Acid on Hepatic Fatty Acid/Triglyceride Metabolism and Hepatobiliary Detoxification/Elimination in Morbidly Obese and Gallstone Patients[NCT01625026]Phase 240 participants (Actual)Interventional2013-09-30Completed
Effect of Bile Acids in the Gut on GLP-1 Secretion in Healthy Subjects and Patients With Type 2 Diabetes[NCT01666223]20 participants (Actual)Interventional2012-11-30Completed
Study Evaluating the Effectiveness of Obeticholic Acid on Hepatic Real-World Outcomes in Patients With Primary Biliary Cholangitis[NCT05293938]0 participants (Actual)Observational2022-03-28Withdrawn(stopped due to A fundamental protocol deviation on data capture left the database no longer fit for purpose.)
Effects of Diet and Medication in Patients With Cerebrotendinous Xanthomatosis (CTX)[NCT00004346]Phase 25 participants Interventional1996-01-31Recruiting
[NCT00004442]25 participants (Anticipated)InterventionalTerminated
A Phase 2 Clinical Trial Investigating the Effects of Obeticholic Acid on Lipoprotein Metabolism in Subjects With Primary Biliary Cirrhosis[NCT01865812]Phase 227 participants (Actual)Interventional2013-12-03Completed
Effects of Ursodeoxycholic Acid and Chenodeoxycholic Acid on GLP-1 Secretion After Roux-en-Y Gastric Bypass[NCT02340247]Phase 411 participants (Actual)Interventional2014-11-30Completed
Effect of Intraduodenal Perfusion of Bile Acids on the Secretion of Gastrointestinal Satiation Peptides in Healthy Male Volunteers[NCT01674946]Phase 112 participants (Actual)Interventional2011-09-30Completed
Validation of Stimulated ∆FGF19 for Diagnosing Bile Acid Diarrhoea[NCT03059537]Phase 471 participants (Actual)Interventional2017-03-13Completed
Replicate Studies Evaluating the Effectiveness of Obeticholic Acid on Hepatic Real-World Outcomes in Patients With Primary Biliary Cholangitis[NCT05292872]4,937 participants (Actual)Observational2022-03-28Completed
Impact of Metformin and Polysorbate 80 on Drug Absorption and Disposition[NCT04640571]Phase 418 participants (Actual)Interventional2021-04-01Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

Change in Stool Index

"Change in index calculated on a weekly basis, between week 2 (baseline) and week 4 (week 2 of treatment).~Index calculated as ([weekly stool frequency x mean Bristol Stool Form Scale score] = Loperamide use [weekly mg x 3]).~Individual scores ranged from 25 to 1095, with higher scores being worst." (NCT01585025)
Timeframe: Week 2, Week 4

Interventionindex score (Median)
Primary BAD-37.5
Secondary BAD-32.5
Idiopathic Diarrhoea Controls-5.5

Changes in Fasting 7α-hydroxy-4-cholesten-3-one

Change in fasting 7α-hydroxy-4-cholesten-3-one before and after 15 day administration of OCA. (NCT01585025)
Timeframe: Day 0, Day 15

Interventionmicrogram/L (Median)
Primary BAD-11.5
Secondary BAD-27.5
Idiopathic Diarrhoea Controls-6.5

Changes in Fasting FGF19

The primary outcome measure is the change over 2 weeks in fasting serum fibroblast growth factor (FGF19) in 3 groups of patients: primary bile acid diarrhoea, secondary bile acid diarrhoea, and a control population of patients with chronic diarrhoea but with normal bile acid retention. (NCT01585025)
Timeframe: Day 0, Day 15

Interventionpercentage increase of baseline (Median)
Primary BAD71
Secondary BAD25
Idiopathic Diarrhoea Controls130

Changes in Mean Stool Form

Change in mean stool form reported per week between week 2 (baseline) and week 4 (week 2 of treatment) using the Bristol Stool Form Scale (range of scores 1 to 7). High scores are a worse outcome (7=liquid stools). (NCT01585025)
Timeframe: Week 2, Week 4

InterventionScore on Bristol scale (Median)
Primary BAD-0.71
Secondary BAD-0.38
Idiopathic Diarrhoea Controls-0.46

Changes in Non-fasting Response of FGF19 to OCA

Change in dynamic response of FGF19 in 6 hours following OCA administration; at start and end of 15 day OCA test period. (NCT01585025)
Timeframe: Day 0, Day 15

Interventionpercentage change (Median)
Primary BAD11.6
Secondary BAD14.6
Idiopathic Diarrhoea Controls32.4

Changes in Serum Total Bile Acids.

Dynamic changes of total bile acids over 6 hour period following OCA administration before and after 15 day OCA period. (NCT01585025)
Timeframe: Day 0, Day 15

Interventionmicromol/L (Median)
Primary BAD-18.1
Secondary BAD-2.0
Idiopathic Diarrhoea Controls-15.5

Changes in Stool Frequency

Change in total number of stool episodes reported per week between week 2 (baseline) and week 4 (week 2 of treatment) (NCT01585025)
Timeframe: Week 2, Week 4

Interventionstools per week (Median)
Primary BAD-4.5
Secondary BAD-2.5
Idiopathic Diarrhoea Controls3.0

Change in C4 Total Area Under the Curve From Baseline Before Versus After Cholecystectomy

Change in total AUC for C4 between visit 1 and visit 2 (NCT03168555)
Timeframe: before cholecystectomy and 3-5 months after cholecystectomy

Interventionng/mL x minutes (Mean)
Visit 1774
Visit 21040

Change in Chenodeoxycholic Acid (CDCA) Absorption to Plasma From Baseline Before Versus After Cholecystectomy

Change from baseline to after cholecystectomy in median unconjugated CDCA plus meal stimulated absorption of unconjugated CDCA to measured in plasma (total area under the CDCA curve with measurement at fasting ie. t=0 minutes and subsequently at 60, 90, 120, and finally at 150minutes. (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

InterventionµM * minutes (Median)
Visit 11554
Visit 21847

Change in Fasting 7-alpha-hydroxy-cholestenone (C4) From Baseline Before Versus After Cholecystectomy

Change from baseline to after cholecystectomy in fasting C4 (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

Interventionng/mL (Mean)
Visit 16.0
Visit 27.5

Change in Fasting FGF19 From Baseline Before Versus After Cholecystectomy

change in fasting FGF19 before versus after cholecystectomy (NCT03168555)
Timeframe: before and 3-5 months after cholecystectomy

Interventionpg per mL (Mean)
Visit 1102
Visit 292

Change in Lipid Status From Baseline Before Versus After Cholecystectomy

Change from baseline to after cholecystectomy in plasma triglycerides (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

Interventionmmol/L (Median)
Visit 11.4
Visit 21.5

Change in Patient Reported Diarrhea Symptoms Correlated With Change in C4 From Baseline Before Versus After Cholecystectomy

Correlation between patient reported frequency of diarrhea (gastrointestinal quality of life index item 31) and fasting C4 before versus after cholecystectomy (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

InterventionSpearman correlation coefficient (Number)
Visit 1-.23
Visit 2-0.52

Change in Patient Reported Diarrhea Symptoms Correlated With Change in FGF19 From Baseline Before Versus After Cholecystectomy

Correlation between patient reported frequency of diarrhea (gastrointestinal quality of life index item 31) and fasting FGF19 before versus after cholecystectomy (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

InterventionSpearman correlation coefficient (Number)
Visit 10.08
Visit 2-0.07

Change in Stimulated Fibroblast Growth Factor 19 (FGF19) From Baseline Before Versus Post Cholecystectomy

Change from baseline to after cholecystectomy in median chenodeoxycholic acid (CDCA) plus meal stimulated FGF19 (delta 0 min to 150 min after stimulation) (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

Interventionpg per mL (Median)
Visit 181
Visit 2186

Change in Stool Consistency (Bristol Stool Type) From Baseline Before Versus After Cholecystectomy

Change from baseline to after cholecystectomy in mean Bristol type per stool as of a seven-day diary baseline versus after cholecystectomy. The Bristol scale divides stool into seven categories from 1 (hard lumps) to 7 (completely watery stool). The diary shows pictograms with short text descriptions. (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

InterventionBristol stool type (Median)
Visit 14.2
Visit 24.0

Change in Stool Frequency From Baseline Before Versus After Cholecystectomy

Change from baseline to after cholecystectomy in number of stools as a mean of a seven-day diary baseline versus after cholecystectomy. That is the diary results for each study participant is tallied using mean values. The tallying of these diary results is done using medians. Therefore the unit used is 'mean stools per day' and this is reported with medians (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

Interventionmean stools per day (Median)
Visit 11.6
Visit 21.6

Change in Stool Pattern Correlated to FGF19

Spearman correlation between change from baseline to after cholecystectomy in FGF19 and in mean number of stools (NCT03168555)
Timeframe: baseline and 3 - 5 months after cholecystectomy

InterventionSpearman correlation coefficient (Number)
Intervention-0.21

Change in Serum Levels of Alanine Aminotransferase

Change in serum levels of Alanine Aminotransferase from baseline to month 4 is assessed (NCT02430077)
Timeframe: Baseline, Month 4

InterventionU/L (Median)
Baseline (Month 0)16.3
Obeticholic Acid (Month 4)15.8
Placebo (Month 0)19.0
Placebo (Month 4)18.5

Change in Serum Levels of Aspartate Aminotransferases

Change in serum levels of Aspartate Aminotransferases from baseline to month 4 is assessed (NCT02430077)
Timeframe: Baseline, Month4

InterventionU/L (Median)
Baseline (Month 0)15.5
Obeticholic Acid (Month 4)16.8
Placebo (Month 0)18.7
Placebo (Month 4)16.5

Change in Serum Levels of Gamma-Glutamyl Transpeptidase

Change in serum levels of Gamma-Glutamyl Transpeptidase from baseline to month 4 is assessed (NCT02430077)
Timeframe: Baseline, Month 4

InterventionU/L (Median)
Baseline (Month 0)27.5
Obeticholic Acid (Month 4)17
Placebo (Month 0)21.6
Placebo (Month 4)21.2

Change in Serum Triglyceride Levels

Change in the serum levels of Triglycerides from baseline to month 4 is being assessed. (NCT02430077)
Timeframe: Baseline, Month 4

Interventionmg/dL (Median)
Baseline (Month 0)199.2
Obeticholic Acid (Month 4)182.3
Placebo (Month 0)169.3
Placebo (Month 4)161.3

Change in the Liver Triglycerides (TG).

The primary end-point variable was the change in the liver TG content assessed using proton-density fat fraction mapping by Magnetic Resonance Imaging (MRI). (NCT02430077)
Timeframe: Baseline, 4 months

Interventionpercentage of liver triglycerides (Median)
Baseline (Month 0)13.2
Obeticholic Acid (Month 4)6.4
Placebo (Month 0)12.3
Placebo (Month 4)10.6

DB Phase: Change From Baseline to Month 18 in LSM

Non-invasive radiological methods to assess liver stiffness were conducted at selected study sites where the respective devices were available. These assessments were taken by vibration controlled TE method using FibroScan®. Participant was included as a random effect and an unstructured covariance matrix was used assuming convergence could be attained. The principal comparison was at Month 18. Baseline was defined as the last value collected prior to the first administration of the IP. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03439254)
Timeframe: Baseline and up to Month 18

InterventionKilopascal (kPa) (Median)
DB: Placebo-0.50
DB: OCA 10 Milligrams (mg)-3.10
DB: OCA 10 mg Titrated to OCA 25 mg-2.90

DB Phase: ELF at Baseline

ELF was non-invasive panel of circulating fibrosis markers calculated from serum biomarkers. The markers of fibrosis comprised HA, TIMP1 and PIIINP. Each of these markers was measured by an immunoassay and an ELF score was generated, from which a level of fibrosis severity could be determined. The ELF test was a composite score: < 7.7: no to mild fibrosis; ≥ 7.7 - < 9.8: Moderate fibrosis; ≥ 9.8 - < 11.3: Severe fibrosis; ≥ 11.3: Cirrhosis.; higher ELF scores were associated with worsening liver fibrosis. Baseline was defined as the last value collected prior to the first administration of the IP. (NCT03439254)
Timeframe: Baseline (Day 1)

InterventionScores on a scale (Mean)
DB: Placebo10.50
DB: OCA 10 Milligrams (mg)10.60
DB: OCA 10 mg Titrated to OCA 25 mg10.61

DB Phase: FIB-4 at Baseline

FIB-4 was a noninvasive assessment of liver disease assessed by a combination of age, ALT and platelet results. FIB-4 was the ratio of age in years and aminotransferase to platelet count. It was a non-invasive hepatic fibrosis index score combining standard biochemical values, platelets, ALT, AST and age that was calculated using formula: FIB-4 = (Age [years] x AST [U/L]) / (platelets [10^9/L] x (square root of ALT [U/L])). A FIB-4 index of <1.45 indicated no or moderate fibrosis and an index of > 3.25 indicated extensive fibrosis/cirrhosis. Higher ratio indicated worse condition. Baseline was defined as the last value collected prior to the first administration of the IP. (NCT03439254)
Timeframe: Baseline (Day 1)

InterventionRatio (Mean)
DB: Placebo2.279
DB: OCA 10 Milligrams (mg)2.475
DB: OCA 10 mg Titrated to OCA 25 mg2.405

DB Phase: Number of Participants Who Were Responders and Showed Improvement in Fibrosis by at Least 1 Stage Without Worsening of Nonalcoholic Steatohepatitis (NASH)

Fibrosis stage was evaluated by NASH Clinical Research Network(CRN)Fibrosis Staging System with stages:0=no fibrosis;1=perisinusoidal/periportal;1A=mild,zone 3,perisinusoidal;1B=moderate,zone 3,perisinusoidal;1C=portal/periportal;2=perisinusoidal and portal/periportal;3=bridging fibrosis;4=cirrhosis.No worsening of steatohepatitis was defined as no worsening of lobular inflammation or hepatocellular ballooning grade as per scoring in relevant nonalcoholic fatty liver disease activity score (NAS) categories.NAS is semiquantitative scoring system based on unweighted sum of:steatosis (0=<5% to 3=>66%),lobular inflammation(0=no foci to 3=>4 foci/200x),hepatocellular ballooning(0=none to 2=many cells/prominent ballooning)scores.Total scale range:0-12;0:no features of fatty liver disease and 12:highest degree of fatty liver disease.Higher scores:worse symptoms.Responders:did not discontinue treatment due to Adverse event(AE) or did not die and had evaluable post-Baseline biopsy assessment (NCT03439254)
Timeframe: Up to 18 months

InterventionParticipants (Count of Participants)
DB: Placebo31
DB: OCA 10 Milligrams (mg)33
DB: OCA 10 mg Titrated to OCA 25 mg37

OLE Phase: Change From Baseline to Month 12 in Liver Stiffness Measurement (LSM)

Non-invasive radiological methods to assess liver stiffness were conducted at selected study sites where the respective devices were available. These assessments were taken by vibration controlled transient elastography (TE) method using FibroScan®. Participant was included as a random effect and an unstructured covariance matrix was used assuming convergence could be attained. Baseline was defined as the last value collected prior to the first administration of the investigational product (IP). Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03439254)
Timeframe: Baseline and up to Month 12

InterventionKilopascal (kPa) (Median)
OLE: OCA 10 mg (DB Placebo)-2.10
OLE: OCA 10 mg (DB OCA 10 mg)-2.90
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)-3.45

OLE Phase: Enhanced Liver Fibrosis (ELF) at Baseline

ELF was non-invasive panel of circulating fibrosis markers calculated from serum biomarkers. The markers of fibrosis comprised hyaluronic acid (HA), tissue inhibitor of metalloproteinase (TIMP1) and procollagen III N-terminal peptide (PIIINP). Each of these markers was measured by an immunoassay and an ELF score was generated, from which a level of fibrosis severity could be determined. The ELF test was a composite score: < 7.7: no to mild fibrosis; ≥ 7.7 - < 9.8: Moderate fibrosis; ≥ 9.8 - < 11.3: Severe fibrosis; ≥ 11.3: Cirrhosis.; higher ELF scores were associated with worsening liver fibrosis. Baseline was defined as the last value collected prior to the first administration of the IP. (NCT03439254)
Timeframe: Baseline (Day 1)

InterventionScores on a scale (Mean)
OLE: OCA 10 mg (DB Placebo)10.46
OLE: OCA 10 mg (DB OCA 10 mg)10.50
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)10.48

OLE Phase: Fibrosis-4 (FIB-4) at Baseline

FIB-4 was a noninvasive assessment of liver disease assessed by a combination of age, alanine aminotransferase (ALT) and platelet results. FIB-4 was the ratio of age in years and aminotransferase to platelet count. It was a non-invasive hepatic fibrosis index score combining standard biochemical values, platelets, ALT, Aspartate aminotransferase (AST) and age that was calculated using formula: FIB-4 = (Age [years] x AST [Units per Liter {U/L}]) / (platelets [10^9/L] x (square root of ALT [U/L])). A FIB-4 index of <1.45 indicated no or moderate fibrosis and an index of > 3.25 indicated extensive fibrosis/cirrhosis. Higher ratio indicated worse condition. Baseline was defined as the last value collected prior to the first administration of the IP. (NCT03439254)
Timeframe: Baseline (Day 1)

InterventionRatio (Mean)
OLE: OCA 10 mg (DB Placebo)2.179
OLE: OCA 10 mg (DB OCA 10 mg)2.267
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)2.266

OLE Phase: Number of Participants Reporting All-cause Mortality

All-cause mortality is defined as death due to any cause. Number of participants reporting all-cause mortality is presented (NCT03439254)
Timeframe: Up to Month 12

InterventionParticipants (Count of Participants)
OLE: OCA 10 mg (DB Placebo)1
OLE: OCA 10 mg (DB OCA 10 mg)0
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)1

OLE Phase: Number of Participants With Adjudicated Liver Related Clinical Outcomes: Model for End-Stage Liver Disease (MELD) Score ≥15

MELD was a scoring system for assessing the severity of chronic liver disease and to assess prognosis and suitability for liver transplantation. It uses the participant's values for total bilirubin, serum creatinine, and the international normalized ratio for prothrombin time to predict survival. MELD score ranges from 6 (less ill) to 40 (gravely ill) with scores and mortality probability being: Score 40=71.3% mortality; Scores 30-39=52.6% mortality; Scores 20-29=19.6% mortality; Scores10-19=6.0% mortality; Score 9 or less=1.9% mortality. Higher scores indicated greater disease severity. Adjudication was performed under the review of HSAC of all available data for each identified participant to determine liver injury status. Number of participants with adjudicated liver related clinical outcomes for MELD score ≥15 is presented. (NCT03439254)
Timeframe: Up to 12 months

InterventionParticipants (Count of Participants)
OLE: OCA 10 mg (DB Placebo)0
OLE: OCA 10 mg (DB OCA 10 mg)0
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)1

OLE Phase: Number of Participants With Adjudicated Liver Related Clinical Outcomes: Worsening of Child-Pugh Score

The Child-Pugh classification was a scoring system used for the classification of the severity of cirrhosis. It included three continuous variables (bilirubin, albumin, and international normalized ratio) and two discrete variables (ascites and encephalopathy). Each variable was scored 1-3 with 3 indicating most severe derangement. The determination of Child-Pugh score ranged from 5 to 15. The higher the score, the sicker the participant. Adjudication was performed under the review of HSAC of all available data for each identified participant to determine liver injury status. Number of participants with adjudicated liver related clinical outcomes for worsening of Child-Pugh score is presented. (NCT03439254)
Timeframe: Up to 12 months

InterventionParticipants (Count of Participants)
OLE: OCA 10 mg (DB Placebo)0
OLE: OCA 10 mg (DB OCA 10 mg)1
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)2

OLE Phase: Number of Participants With Adjudicated Liver Related Clinical Outcomes: Ascites, Hepatocellular Carcinoma (HCC) and Non-liver Related Death

Adjudication was performed under the review of Hepatic Safety Adjudication Committee (HSAC) of all available data for each identified participant to determine liver injury status. Number of participants with adjudicated liver related clinical outcomes for the following is presented: Ascites (secondary to cirrhosis and requiring medical intervention), Hepatocellular carcinoma (HCC) and non-liver related death. (NCT03439254)
Timeframe: Up to 12 months

,,
InterventionParticipants (Count of Participants)
Ascites (secondary to cirrhosis and requiring medical intervention)HCCNon-liver related death
OLE: OCA 10 mg (DB OCA 10 mg)010
OLE: OCA 10 mg (DB Placebo)131
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)211

OLE Phase: Number of Participants With Non-serious Adverse Events (AEs) and Serious Adverse Events (SAEs)

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE is any untoward medical occurrence that, at any dose results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent disability/incapacity, is a congenital anomaly/birth defect or any other situation according to medical or scientific judgment. (NCT03439254)
Timeframe: Up to 12 months

,,
InterventionParticipants (Count of Participants)
AEsSAEs
OLE: OCA 10 mg (DB OCA 10 mg)21350
OLE: OCA 10 mg (DB Placebo)19926
OLE: Titrated to OCA 25 mg (DB OCA 10 mg Titrated to OCA 25 mg)19748

Change in Alanine Aminotransferase

(NCT01265498)
Timeframe: baseline to 72 weeks

InterventionU/L (Mean)
Obeticholic Acid-38
Placebo-18

Change in Albumin

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventiongl/L (Mean)
Obeticholic Acid-0.2
Placebo0.3

Change in Alkaline Phosphatase

(NCT01265498)
Timeframe: baseline to 72 weeks

InterventionU/L (Mean)
Obeticholic Acid12
Placebo-6

Change in Asparate Aminotransferase

(NCT01265498)
Timeframe: baseline to 72 weeks

InterventionU/L (Mean)
Obeticholic Acid-27
Placebo-10

Change in Bicarbonate

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid-0.7
Placebo-0.1

Change in Body-mass Index

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionkg/m² (Mean)
Obeticholic Acid-0.7
Placebo0.1

Change in Calcium

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid0.01
Placebo-0.01

Change in Creatinine

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionμmol/L (Mean)
Obeticholic Acid1.5
Placebo-1.1

Change in Diastolic Blood Pressure

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmm Hg (Mean)
Obeticholic Acid0
Placebo0

Change in Fasting Serum Glucose

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid0.4
Placebo0.2

Change in Glycated Haemoglobin A1c

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/mol (Mean)
Obeticholic Acid0.5
Placebo0.4

Change in Haematocrit

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionproportion of 1.0 (Mean)
Obeticholic Acid0.00
Placebo0.00

Change in Haemoglobin

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventiong/L (Mean)
Obeticholic Acid0.6
Placebo0.3

Change in HDL Cholesterol

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid-0.02
Placebo0.03

Change in HOMA-IR

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionglucose[mmol/L]× insulin[pmol/L] / 22.5 (Mean)
Obeticholic Acid15
Placebo4

Change in Insulin

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionpmol/L (Mean)
Obeticholic Acid29
Placebo10

Change in International Normalised Ratio

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionratio (Mean)
Obeticholic Acid-0.03
Placebo0.00

Change in LDL Cholesterol

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid0.22
Placebo-0.22

Change in Mean Corpuscular Volume

(NCT01265498)
Timeframe: baseline to 72 weeks

InterventionfL (Mean)
Obeticholic Acid-0.8
Placebo0.3

Change in Phosphate

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid0.01
Placebo0.02

Change in Platelet Count

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionplatelets *10^9 per L (Mean)
Obeticholic Acid12
Placebo-4

Change in Prothrombin Time

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventions (Mean)
Obeticholic Acid-0.1
Placebo0.0

Change in SF-36 Quality of Life Mental Component Summary

Short Form (36) Health Survey The SF-36 evaluates health-related quality of life in 8 domains consisting of two components: physical and mental. The score for each domain ranges from 0 to 100. Norm based scoring (based on the general US population) is used with a mean of 50 and standard deviation of 10. Higher values represent a better outcome. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid0
Placebo1

Change in SF-36 Quality of Life Physical Component Summary

Short Form (36) Health Survey The SF-36 evaluates health-related quality of life in 8 domains consisting of two components: physical and mental. The score for each domain ranges from 0 to 100. Norm based scoring (based on the general US population) is used with a mean of 50 and standard deviation of 10. Higher values represent a better outcome. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid0
Placebo-1

Change in Systolic Blood Pressure

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmm Hg (Mean)
Obeticholic Acid-4
Placebo-1

Change in Total Bilirubin

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionμmol/L (Mean)
Obeticholic Acid-1.0
Placebo0.6

Change in Total Cholesterol

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid0.16
Placebo-0.19

Change in Total Protein

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventiongl/L (Mean)
Obeticholic Acid0.2
Placebo-0.5

Change in Triglycerides

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionmmol/L (Mean)
Obeticholic Acid-0.22
Placebo-0.08

Change in Uric Acid

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionμmol/L (Mean)
Obeticholic Acid2
Placebo-11

Change in Waist Circumference

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventioncm (Mean)
Obeticholic Acid-1.5
Placebo-0.6

Change in Waist-to-hip Ratio

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionratio (Mean)
Obeticholic Acid0.00
Placebo0.00

Change in Weight

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionkg (Mean)
Obeticholic Acid-2.3
Placebo0.0

Change in White Blood Cell Count

(NCT01265498)
Timeframe: baseline to 72 weeks

Interventionwhite blood cells *10^9 per L (Mean)
Obeticholic Acid0.0
Placebo0.0

Change in γ-glutamyl Transpeptidase

(NCT01265498)
Timeframe: baseline to 72 weeks

InterventionU/L (Mean)
Obeticholic Acid-37
Placebo-6

Fibrosis: Change in Score

Change in fibrosis score. Fibrosis was assessed on a scale of 0-4, with higher scores showing more severe fibrosis. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid-0.2
Placebo0.1

Fibrosis: Patient With Improvement

Patients with improvement in fibrosis score. Fibrosis was assessed on a scale of 0-4, with higher scores showing more severe fibrosis. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionparticipants (Number)
Obeticholic Acid36
Placebo19

Hepatic Histological Improvement in Nonalcoholic Fatty Liver Disease (NAFLD) Activity Score (NAS)

"Centrally scored histological improvement in nonalcoholic fatty liver disease (NAFLD) from baseline to the end of 72 weeks of treatment, where improvement is defined as:~No worsening in fibrosis; and~A decrease in NAFLD Activity Score (NAS) of at least 2 points" (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionparticipants (Number)
Obeticholic Acid50
Placebo23

Hepatocellular Ballooning: Change in Score

Change in hepatocellular ballooning score. Hepatocellular ballooning was assessed on a scale of 0-2, with higher scores showing more severe ballooning. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid-0.5
Placebo-0.2

Hepatocellular Ballooning: Patients With Improvement

Patients with improvement in hepatocellular ballooning score. Hepatocellular ballooning was assessed on a scale of 0-2, with higher scores showing more severe ballooning. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionparticipants (Number)
Obeticholic Acid47
Placebo30

Lobular Inflammation: Change in Score

Change in lobular inflammation score. Lobular inflammation was assessed on a scale of 0-3, with higher scores showing more severe lobular inflammation. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid-0.5
Placebo-0.2

Lobular Inflammation: Patients With Improvement

Patients with improvement in lobular inflammation score. Lobular inflammation was assessed on a scale of 0-3, with higher scores showing more severe lobular inflammation. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionparticipants (Number)
Obeticholic Acid54
Placebo34

Portal Inflammation: Change in Score

Change in portal inflammation score. Portal inflammation was assessed on a scale of 0-3, with higher scores showing more severe portal inflammation. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid0.2
Placebo0.2

Portal Inflammation: Patients With Improvement

Patients with improvement in portal inflammation score. Portal inflammation was assessed on a scale of 0-2, with higher scores showing more severe portal inflammation. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionparticipants (Number)
Obeticholic Acid12
Placebo13

Resolution of NASH Diagnosis

Resolution of definite nonalcoholic steatohepatitis. Resolution defined as either not NAFLD, or NAFLD but not non-alcoholic steatohepatitis on week 72 biopsy (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionparticipants (Number)
Obeticholic Acid22
Placebo13

Steatosis: Change in Score

Change in steatosis score. Steatosis was assessed on a scale of 0-3, with higher scores showing more severe steatosis. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid-0.8
Placebo-0.4

Steatosis: Patients With Improvement

Patients with improvement in steatosis score. Steatosis was assessed on a scale of 0-3, with higher scores showing more severe steatosis. (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionparticipants (Number)
Obeticholic Acid62
Placebo37

Total NAFLD Activity Score: Change in Score

NAFLD activity score was assessed on a scale of 0-8, with higher scores showing more severe disease (the components of this measure are steatosis [assessed on a scale of 0-3], lobular inflammation [assessed on a scale of 0-3], and hepatocellular ballooning [assessed on a scale of 0-2]). (NCT01265498)
Timeframe: baseline to 72 weeks

Interventionunits on a scale (Mean)
Obeticholic Acid-1.7
Placebo-0.7

DB Phase: Mean Percent Change In Alanine Transaminase (ALT) From Baseline To Day 85

As a marker of hepatocellular injury and liver function, the percent change in ALT from baseline to Day 85 is reported. (NCT00570765)
Timeframe: Baseline, Day 85

InterventionPercent change (Mean)
DB OCA 10 mg-37
DB OCA 50 mg-35
DB OCA Placebo-4

DB Phase: Mean Percent Change In Conjugated Bilirubin From Baseline To Day 85

As a marker of hepatocellular injury and liver function, the percent change in conjugated bilirubin from baseline to Day 85 is reported. (NCT00570765)
Timeframe: Baseline, Day 85

InterventionPercent Change (Mean)
DB OCA 10 mg0.7
DB OCA 50 mg-1.7
DB OCA Placebo30.3

DB Phase: Mean Percent Change In Gamma-glutamyl Transferase (GGT) From Baseline To Day 85

As a marker of hepatocellular injury and liver function, the percent change in GGT from baseline to Day 85 is reported. (NCT00570765)
Timeframe: Baseline, Day 85

InterventionPercent change (Mean)
DB OCA 10 mg-73
DB OCA 50 mg-65
DB OCA Placebo-3

DB Phase: Mean Percent Change In Serum Alkaline Phosphatase (ALP) From Baseline To Day 85

The percent change in serum ALP from baseline to Day 85 is reported. The baseline value used was the mean of the pretreatment Screening and Day 0 evaluations. (NCT00570765)
Timeframe: Baseline, Day 85

InterventionPercent change (Mean)
DB OCA 10 mg-44.5
DB OCA 50 mg-37.6
DB OCA Placebo0.4

LTSE: Mean Percent Change In ALT From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in ALT from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Mean)
LTSE OCA Total-39.6

LTSE: Mean Percent Change In Conjugated Bilirubin From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in conjugated bilirubin from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Mean)
LTSE OCA Total57.8

LTSE: Mean Percent Change In GGT From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in GGT from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Mean)
LTSE OCA Total-55.6

LTSE: Mean Percent Change In Total Bilirubin From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in total bilirubin from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Mean)
LTSE OCA Total2.2

LTSE: Median Percent Change In ALT From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in ALT from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Median)
LTSE OCA Total-52.2

LTSE: Median Percent Change In Conjugated Bilirubin From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in conjugated bilirubin from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Median)
LTSE OCA Total33.3

LTSE: Median Percent Change In GGT From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in GGT from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Median)
LTSE OCA Total-71.1

LTSE: Median Percent Change In Total Bilirubin From Baseline To Last Available Visit

As a marker of hepatocellular injury and liver function, the percent change in total bilirubin from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Last Available Visit (up to 96 months)

InterventionPercent Change (Median)
LTSE OCA Total5.2

LTSE Phase: Mean Percent Change In Serum ALP From Baseline To Month 24, Month 48, Month 72, And Last Available Visit

The percent change in serum ALP from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Month 24, Month 48, Month 72, Last Available Visit (up to 96 months)

InterventionPercent Change (Mean)
Month 24Month 48Month 72Last Available Visit
LTSE OCA Total-38.8-39.3-31.7-30.4

LTSE Phase: Median Percent Change In Serum ALP From Baseline To Month 24, Month 48, Month 72, And Last Available Visit

The percent change in serum ALP from baseline to the last available visit is reported. The DB baseline value was used as the baseline. (NCT00570765)
Timeframe: Baseline (DB), Month 24, Month 48, Month 72, Last Available Visit (up to 96 months)

InterventionPercent Change (Median)
Month 24Month 48Month 72Last Available Visit
LTSE OCA Total-43.1-44.4-33.4-31.8

DB Phase: Alanine Aminotransferase (ALT) Absolute Change From Baseline To Month 12

Blood samples were evaluated for ALT levels. ALT absolute change from baseline (ALT at Month 12 - ALT at Baseline) is presented. (NCT01473524)
Timeframe: Baseline, DB Month 12

InterventionU/L (Least Squares Mean)
DB OCA 5-10 mg-21.26
DB OCA 10 mg-25.31
DB Placebo-4.95

DB Phase: ALP Absolute Change From Baseline To Month 12

Blood samples were evaluated for ALP levels. ALP Absolute Change From Baseline (ALP at Month 12 - ALP at Baseline) is presented. (NCT01473524)
Timeframe: Baseline, DB Month 12

InterventionU/L (Least Squares Mean)
DB OCA 5-10 mg-112.51
DB OCA 10 mg-129.90
DB Placebo-14.42

DB Phase: Aspartate Aminotransferase (AST) Absolute Change From Baseline To Month 12

Blood samples were evaluated for AST levels. AST absolute change from baseline (AST at Month 12 - AST at Baseline) is presented. (NCT01473524)
Timeframe: Baseline, DB Month 12

InterventionU/L (Least Squares Mean)
DB OCA 5-10 mg-13.03
DB OCA 10 mg-15.00
DB Placebo1.04

DB Phase: Composite Endpoint Alkaline Phosphatase (ALP) And Total Bilirubin, 10 mg OCA Versus Placebo

Percentage of participants at Month 12 with ALP < 1.67 x upper limit of normal (ULN) and total bilirubin ≤ ULN and ALP decrease of ≥ 15% from baseline. (NCT01473524)
Timeframe: DB Month 12

Interventionpercentage of participants (Number)
DB OCA 10 mg47
DB Placebo10

DB Phase: Composite Endpoint ALP And Total Bilirubin, 10 mg Versus Placebo

Percentage of participants at Month 6 with ALP < 1.67x ULN and total bilirubin ≤ ULN and ALP decrease of ≥ 15% from baseline. (NCT01473524)
Timeframe: DB Month 6

Interventionpercentage of participants (Number)
Double-blind OCA 10 mg51
Double-blind Placebo7

DB Phase: Composite Endpoint ALP And Total Bilirubin, 5-10 mg Versus Placebo

Percentage of participants at Month 12 with ALP < 1.67x ULN and total bilirubin ≤ ULN and ALP decrease of ≥ 15% from baseline. (NCT01473524)
Timeframe: DB Month 12

Interventionpercentage of participants (Number)
DB OCA 5-10 mg46
DB Placebo10

DB Phase: Composite Endpoint ALP And Total Bilirubin, 5-10 mg Versus Placebo

Percentage of participants at Month 6 with ALP < 1.67x ULN and total bilirubin ≤ ULN and ALP decrease of ≥ 15% from baseline. (NCT01473524)
Timeframe: DB Month 6

Interventionpercentage of participants (Number)
DB OCA 5-10 mg34
DB Placebo7

DB Phase: Direct Bilirubin Absolute Change From Baseline To Month 12

Blood samples were evaluated for bilirubin levels. Direct bilirubin absolute change from baseline (direct bilirubin at Month 12 - direct bilirubin at Baseline) is presented. (NCT01473524)
Timeframe: Baseline, DB Month 12

Interventionumol/L (Least Squares Mean)
DB OCA 5-10 mg-0.13
DB OCA 10 mg-0.49
DB Placebo1.89

DB Phase: Gamma-glutamyltransferase (GGT) Absolute Change From Baseline To Month 12

Blood samples were evaluated for GGT levels. GGT absolute change from baseline (GGT at Month 12 - GGT at Baseline) is presented. (NCT01473524)
Timeframe: Baseline, DB Month 12

InterventionU/L (Least Squares Mean)
DB OCA 5-10 mg-140.83
DB OCA 10 mg-176.66
DB Placebo6.70

DB Phase: Total Bilirubin Absolute Change From Baseline To Month 12

Blood samples were evaluated for bilirubin levels. Total bilirubin absolute change from baseline (total bilirubin at Month 12 - total bilirubin at Baseline) is presented. (NCT01473524)
Timeframe: Baseline, DB Month 12

Interventionumol/L (Least Squares Mean)
DB OCA 5-10 mg-0.33
DB OCA 10 mg-0.90
DB Placebo1.98

LTSE Phase: ALP Change From DB Baseline

Blood samples were evaluated for ALP levels. ALP Change From Baseline (ALP at LTSE Months 12, 24, 36, 48, and 60 - ALP at Baseline) is presented. DB baseline is the mean of all available evaluations prior to DB treatment. (NCT01473524)
Timeframe: DB Baseline, LTSE Months 12, 24, 36, 48, and 60

,,,
InterventionU/L (Mean)
Month 12Month 24Month 36Month 48Month 60
LTSE OCA (DB OCA 10 mg)-104.39-102.52-84.65-101.50-117.49
LTSE OCA (DB OCA 5-10 mg)-106.63-120.86-100.98-118.23-118.99
LTSE OCA (DB Placebo)-104.36-100.99-112.73-115.51-119.52
Overall LTSE OCA-105.13-108.34-98.96-111.49-118.74

LTSE Phase: ALP Levels

Blood samples were evaluated for ALP levels. (NCT01473524)
Timeframe: LTSE Day 0 and LTSE Months 12, 24, 36, 48, and 60

,,,
InterventionU/L (Mean)
LTSE Day 0LTSE Month 12LTSE Month 24LTSE Month 36LTSE Month 48LTSE Month 60
LTSE OCA (DB OCA 10 mg)191.24198.68194.57214.66192.00191.37
LTSE OCA (DB OCA 5-10 mg)218.69209.49195.14204.52189.75200.90
LTSE OCA (DB Placebo)317.79226.28215.99205.37198.70209.38
Overall LTSE OCA243.75211.47201.47208.27193.38200.94

LTSE Phase: Composite Endpoint ALP And Total Bilirubin

Percentage of participants at Months 24, 36, 48, and 60 with ALP < 1.67x ULN and total bilirubin ≤ ULN and ALP decrease of ≥ 15% from baseline. DB Month 12 is the baseline for the LTSE phase. (NCT01473524)
Timeframe: Baseline (DB Month 12), LTSE Months 24, 36, 48, and 60

,,,
Interventionpercentage of participants (Number)
Baseline (Double-blind Month 12)LTSE Month 12LTSE Month 24LTSE Month 36LTSE Month 48LTSE Month 60
LTSE OCA (DB OCA 10 mg)565861515552
LTSE OCA (DB OCA 5-10 mg)515560485248
LTSE OCA (DB Placebo)94154496050
Overall LTSE OCA385158495650

Ascending Colon Emptying (AC t_1/2)

(NCT00912301)
Timeframe: after 4 days' treatment

Interventionhours (Mean)
NaCDC 500 mg9.5
NaCDC 1000 mg8.2
Placebo15.8

Colonic Filling at 6 Hours

Percent of the radio-labeled meal that reached the colon at 6 hours, indirectly reflecting small bowel transit time. (NCT00912301)
Timeframe: after 4 days' treatment

Interventionpercentage of the radio-labeled meal (Mean)
NaCDC 500 mg52.6
NaCDC 1000 mg54.8
Placebo50.6

Colonic Geometric Center at 24 Hours (GC24)

The scintigraphic method is used to measure colonic transit. An isotope is adsorbed on activated charcoal particles and delivered to the colon in a delayed release capsule. Anterior and posterior gamma images are taken hourly. The geometric center (GC) is the weighted average of counts in the different colonic regions. The scale ranges from 1 to 5; a high GC implies faster colonic transit, a GC of 1 implies all isotope is in the ascending colon, and a GC of 5 implies all isotope is in the stool. (NCT00912301)
Timeframe: after 4 days of treatment

Interventionunits on a scale (Mean)
NaCDC 500 mg3.1
NaCDC 1000 mg3.5
Placebo2.2

Colonic Transit at 48 Hours (GC48)

The scintigraphic method is used to measure colonic transit. An isotope is adsorbed on activated charcoal particles and delivered to the colon in a delayed release capsule. Anterior and posterior gamma images are taken hourly. The geometric center (GC) is the weighted average of counts in the different colonic regions. The scale ranges from 1 to 5; a high GC implies faster colonic transit, a GC of 1 implies all isotope is in the ascending colon, and a GC of 5 implies all isotope is in the stool. (NCT00912301)
Timeframe: after 4 days of treatment

Interventionunits on a scale (Mean)
NaCDC 500 mg4.1
NaCDC 1000 mg4.3
Placebo3.8

Stool Consistency

"The subjects rated their stool consistency using the Bristol Stool Scale. The Bristol Stool Scale is a medical aid designed to classify the form of human feces into seven categories or types. Types 1 and 2 indicate constipation with 3 and 4 being the ideal stools especially the latter, as they are the easiest to defecate, and 5-7 tending towards diarrhea." (NCT00912301)
Timeframe: after 4 days' treatment

Interventionunits on a scale (Mean)
NaCDC 500 mg4.4
NaCDC 1000 mg4.4
Placebo2.9

Absolute Change From Baseline In HDL Cholesterol Concentration

(NCT01865812)
Timeframe: Baseline, Month 24/EOT

Interventionmmol/L (Least Squares Mean)
Long-term Safety Extension Phase0.5

Absolute Change From Baseline In HDL Particle Number

(NCT01865812)
Timeframe: Baseline, Month 24/EOT

Interventionumol/L (Least Squares Mean)
Long-term Safety Extension Phase-0.09

Absolute Change From Baseline In HDL Particle Number

(NCT01865812)
Timeframe: Baseline, Week 8

Interventionumol/L (Least Squares Mean)
Primary Treatment Phase-0.06

Absolute Change From Baseline In HDL Particle Size

(NCT01865812)
Timeframe: Baseline, Month 24/EOT

Interventionnm (Least Squares Mean)
Long-term Safety Extension Phase0.04

Absolute Change From Baseline In HDL Particle Size

(NCT01865812)
Timeframe: Baseline, Week 8

Interventionnm (Least Squares Mean)
Primary Treatment Phase-0.44

Absolute Change From Baseline In High-density Lipoprotein (HDL) Cholesterol Concentration

(NCT01865812)
Timeframe: Baseline, Week 8

Interventionmmol/L (Least Squares Mean)
Primary Treatment Phase-0.38

Median Change From Week 8 In HDL Cholesterol Concentration At Week 12

(NCT01865812)
Timeframe: Week 8, Week 12

Interventionmmol/L (Median)
Primary Treatment Phase0.3108

Median Change From Week 8 In HDL Particle Number At Week 12

(NCT01865812)
Timeframe: Week 8, Week 12

Interventionumol/L (Median)
Primary Treatment Phase1.40

Median Change From Week 8 In HDL Particle Size At Week 12

(NCT01865812)
Timeframe: Week 8, Week 12

Interventionnm (Median)
Primary Treatment Phase0.30

Area Under The Concentration-time Curve From Hour 0 To Last Sampling Time (Hour 6) (AUC0-6) For OCA And Conjugates

Results are reported in hour*nanograms per milliliter (h*ng/mL). (NCT01865812)
Timeframe: Week 8

Interventionh*ng/mL (Mean)
OCAGlyco-OCATauro-OCATotal-OCA
Primary Treatment Phase1897026981360

Maximum Plasma Concentration (Cmax) Of OCA And Conjugates

Results are reported in nanograms per milliliter (ng/mL). (NCT01865812)
Timeframe: Week 8

Interventionng/mL (Mean)
OCAGlycine Conjugate (Glyco)-OCATaurine Conjugate (Tauro)-OCATotal-OCA
Primary Treatment Phase107212219409

Median Change From Baseline In Alanine Aminotransferase

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

InterventionU/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-9.80-9.80-11.80-5.95

Median Change From Baseline In Albumin

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventiong/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-0.60-0.201.300.05

Median Change From Baseline In Alkaline Phosphatase

Results are reported in units/Liter (U/L). (NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

InterventionU/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-43.40-31.50-31.906.40

Median Change From Baseline In Amino-terminal Propeptide Of Type III Procollagen

Results are reported in micrograms/Liter (ug/L). (NCT01865812)
Timeframe: Baseline, Month 12, Month 24/EOT

Interventionug/L (Median)
Month 12Month 24/EOT
Long-term Safety Extension Phase0.6702.095

Median Change From Baseline In ApoA1/ApoB Ratio

(NCT01865812)
Timeframe: Baseline, Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

InterventionRatio (Median)
Week 12
Primary Treatment Phase-0.0503

Median Change From Baseline In ApoA1/ApoB Ratio

(NCT01865812)
Timeframe: Baseline, Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

InterventionRatio (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-0.2174-0.1659-0.2860-0.1172-0.291

Median Change From Baseline In Apolipoprotein A1 (ApoA1)

Results are reported in grams per liter (g/L). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventiong/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase-0.1000-0.06000.0400

Median Change From Baseline In Apolipoprotein A1 (ApoA1)

Results are reported in grams per liter (g/L). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventiong/L (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-0.1400-0.0900-0.10000.0100-0.045

Median Change From Baseline In Apolipoprotein B (ApoB)

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionunits on a scale (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase0.09500.07000.0500

Median Change From Baseline In Apolipoprotein B (ApoB)

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionunits on a scale (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase0.04000.04000.06000.09000.060

Median Change From Baseline In Apolipoprotein E

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmg/dL (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase-0.85-0.650.50

Median Change From Baseline In Apolipoprotein E

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmg/dL (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-0.30-0.30-0.100.000.00

Median Change From Baseline In Aspartate Aminotransferase

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

InterventionU/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-5.30-3.60-6.00-4.50

Median Change From Baseline In C-reactive Protein

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionnmol/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase0.00000.000011.4288

Median Change From Baseline In C-reactive Protein

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionnmol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase0.000.000.000.00

Median Change From Baseline In Cholesteryl Ester Transfer Protein

Results are reported in picomole/milliliter/minute (pmol/mL/min). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionpmol/mL/min (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase1.950.304.70

Median Change From Baseline In Cholesteryl Ester Transfer Protein

Results are reported in picomole/milliliter/minute (pmol/mL/min). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionpmol/mL/min (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase1.20-0.600.500.40

Median Change From Baseline In Enhanced Liver Fibrosis (ELF) Score

"Change in ELF was calculated as ELF score at the end of the study minus ELF score prior to the intervention (at baseline). A decrease in the ELF score was considered good as it reflected a decrease in liver fibrosis, and an increase in ELF score was considered bad as it reflected an increase in liver fibrosis.~Change in ELF scores ranged from -0.56 (good) to + 0.68 (bad)." (NCT01865812)
Timeframe: Baseline, Month 12, Month 24/EOT

Interventionscore on a scale (Median)
Month 12Week 24/EOT
Long-term Safety Extension Phase0.0000.150

Median Change From Baseline In Fibroblast Growth Factor-19

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionpg/mL (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase81.8800112.546016.8400

Median Change From Baseline In Fibroblast Growth Factor-19

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionpg/mL (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase81.39029.22055.230-0.740

Median Change From Baseline In Gamma-glutamyl Transferase

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

InterventionU/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-59.40-41.60-40.80-30.05

Median Change From Baseline In Glycoprotein A

Results are reported in picograms/milliliter (pg/mL). (NCT01865812)
Timeframe: Baseline, Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionpg/mL (Median)
Week 12
Primary Treatment Phase12.0

Median Change From Baseline In Glycoprotein A

Results are reported in picograms/milliliter (pg/mL). (NCT01865812)
Timeframe: Baseline, Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionpg/mL (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-27.0-13.0-26.010.0

Median Change From Baseline In HDL Cholesterol Concentration At Weeks 4, 8, and 12

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8, Week 12

Interventionmmol/L (Median)
Week 4Week 8Week 12
Primary Treatment Phase-0.2072-0.31080.0518

Median Change From Baseline In HDL Particle Number At Weeks 4, 8, and 12

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8, Week 12

Interventionumol/L (Median)
Week 4Week 8Week 12
Primary Treatment Phase0.550.601.60

Median Change From Baseline In HDL Particle Size At Weeks 4, 8, and 12

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8, Week 12

Interventionnm (Median)
Week 4Week 8Week 12
Primary Treatment Phase-0.30-0.300.00

Median Change From Baseline In Hepatic Stiffness

Results are reported in kilopascal (kPa). (NCT01865812)
Timeframe: Baseline, Month 12, Month 24/EOT

InterventionkPa (Median)
Month 12Month 24/EOT
Long-term Safety Extension Phase-1.15-1.70

Median Change From Baseline In Hyaluronic Acid

(NCT01865812)
Timeframe: Baseline, Month 12, Month 24/EOT

Interventionng/mL (Median)
Month 12Month 24/EOT
Long-term Safety Extension Phase-5.700-1.805

Median Change From Baseline In LDL Particle Size

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnm (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase-0.30-0.100.00

Median Change From Baseline In LDL Particle Size

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnm (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-0.10-0.20-0.10-0.10-0.10

Median Change From Baseline In Lecithin-cholesterol Acyltransferase Activity

Results are reported in nanomoles/milliliter/hour (nmol/mL/h). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionnmol/mL/h (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase-20.5-13.515.5

Median Change From Baseline In Lecithin-cholesterol Acyltransferase Activity

Results are reported in nanomoles/milliliter/hour (nmol/mL/h). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT

Interventionnmol/mL/h (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-47.046.0-19.0-56.0

Median Change From Baseline In Lipoprotein-a

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionumol/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase0.00000.00000.0000

Median Change From Baseline In Lipoprotein-a

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase0.00000.00000.00000.00000.000

Median Change From Baseline In Low-density Lipoprotein (LDL) Cholesterol (Direct)

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmmol/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase0.270.310.18

Median Change From Baseline In Low-density Lipoprotein (LDL) Cholesterol (Direct)

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmmol/L (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase0.31080.44030.59570.31080.518

Median Change From Baseline In Macrophage Cholesterol Efflux

Results are reported as a percentage of cholesterol. (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Month 6, Month 12, Month 18, Month 24/EOT

Interventionpercentage of cholesterol (Median)
Week 4Week 8
Primary Treatment Phase-0.800-0.705

Median Change From Baseline In Macrophage Cholesterol Efflux

Results are reported as a percentage of cholesterol. (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Month 6, Month 12, Month 18, Month 24/EOT

Interventionpercentage of cholesterol (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-1.745-1.940-2.450-0.770

Median Change From Baseline In Prebeta-1 HDL Concentration

Results are reported in microgram/milliliter (ug/mL). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Month 6, Month 12, Month 18, Month 24/EOT

Interventionug/mL (Median)
Week 4Week 8
Primary Treatment Phase1.557.55

Median Change From Baseline In Prebeta-1 HDL Concentration

Results are reported in microgram/milliliter (ug/mL). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Month 6, Month 12, Month 18, Month 24/EOT

Interventionug/mL (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-6.90-10.051.94-1.35

Median Change From Baseline In Prothrombin International Normalized Ratio

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionratio (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase0.00000.00000.00000.0000

Median Change From Baseline In Prothrombin Time

Results are reported in seconds (sec). (NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionsec (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase0.000-0.0500.4000.200

Median Change From Baseline In Tissue Inhibitor Of Metalloproteinases 1

(NCT01865812)
Timeframe: Baseline, Month 12, Month 24/EOT

Interventionug/L (Median)
Month 12Month 24/EOT
Long-term Safety Extension Phase9.6002.000

Median Change From Baseline In Total And Unconjugated (Direct) Bilirubin

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase0.00000.00000.00000.0000

Median Change From Baseline In Total Bile Acids

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-1.56-1.14-4.61-4.91

Median Change From Baseline In Total Chenodeoxycholic Acid

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-0.20-0.35-1.28-1.17

Median Change From Baseline In Total Cholesterol

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmmol/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase-0.0518-0.28490.2331

Median Change From Baseline In Total Cholesterol

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmmol/L (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-0.18130.07770.2849-0.07770.285

Median Change From Baseline In Total Cholic Acid

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-0.39-0.48-0.49-0.39

Median Change From Baseline In Total Deoxycholic Acid

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-0.59-0.56-0.51-0.59

Median Change From Baseline In Total Endogenous Bile Acid

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-0.83-0.77-2.19-2.02

Median Change From Baseline In Total LDL Particles

Results are reported in nanomoles per liter (nmol/L). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnmol/L (Median)
Month 6 (Small)Month 12 (Small)Month 18 (Small)Month 24/EOT (Small)Last Dose (Small)Month 6 (Large)Month 12 (Large)Month 18 (Large)Month 24/EOT (Large)Last Dose (Large)Month 6 (Intermediate-density Lipoprotein [IDL])Month 12 (IDL)Month 18 (IDL)Month 24/EOT (IDL)Last Dose (IDL)
Long-term Safety Extension Phase148.089.018.034.022.09.0-53.0-18.0-122.0-8.0-12.0-57.0-43.078.011.0

Median Change From Baseline In Total LDL Particles

Results are reported in nanomoles per liter (nmol/L). (NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnmol/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase108.0128.0159.0

Median Change From Baseline In Total Lithocholic Acid

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase0.000.000.000.00

Median Change From Baseline In Total Triglycerides

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmmol/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase-0.02260.05650.1130

Median Change From Baseline In Total Triglycerides

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmmol/L (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-0.1469-0.0113-0.05650.000-0.068

Median Change From Baseline In Total UDCA

(NCT01865812)
Timeframe: Baseline, Month 6, Month 12, Month 18, Month 24/EOT

Interventionumol/L (Median)
Month 6Month 12Month 18Month 24/EOT
Long-term Safety Extension Phase-0.34-0.47-2.89-1.68

Median Change From Baseline In Very Low-density Lipoprotein (VLDL) Cholesterol

Results are reported in milligrams per deciliter (mg/dL). (NCT01865812)
Timeframe: Baseline, Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmg/dL (Median)
Week 12
Primary Treatment Phase5.0

Median Change From Baseline In Very Low-density Lipoprotein (VLDL) Cholesterol

Results are reported in milligrams per deciliter (mg/dL). (NCT01865812)
Timeframe: Baseline, Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionmg/dL (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-21.0-7.0-14.0-1.0-11.5

Median Change From Baseline In VLDL Particle Size

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnm (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase42.5543.4045.10

Median Change From Baseline In VLDL Particle Size

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnm (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-4.80-3.50-4.900.50-4.10

Median Change From Baseline In VLDL Particles

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnmol/L (Median)
Week 4Week 8/EOTWeek 12
Primary Treatment Phase-5.552.70-0.90

Median Change From Baseline In VLDL Particles

(NCT01865812)
Timeframe: Baseline, Week 4, Week 8/End of Treatment (EOT), Week 12, Month 6, Month 12, Month 18, Month 24/EOT, Last Dose

Interventionnmol/L (Median)
Month 6Month 12Month 18Month 24/EOTLast Dose
Long-term Safety Extension Phase-4.70-6.20-6.80-0.30-6.80

Participants With Lipoprotein X

Lipoprotein samples were assessed using nuclear magnetic resonance spectroscopy for the presence/absence of Lipoprotein X. Lipoprotein X sometimes appears with advanced cholestasis and can confound assessment of other lipoprotein concentrations, particularly LDL. (NCT01865812)
Timeframe: Week 12 and Last Dose

,
InterventionParticipants (Count of Participants)
Week 12Last Dose
Long-term Safety Extension Phase00
Primary Treatment Phase10

Time To Reach Cmax (Tmax) For OCA And Conjugates

Results are reported in hours (h). (NCT01865812)
Timeframe: Week 8

Interventionh (Median)
OCAGlyco-OCATauro-OCATotal-OCA
Primary Treatment Phase1.005.005.985.00

Research Highlights

Safety/Toxicity (46)

ArticleYear
Safety and tolerability of obeticholic acid in chronic liver disease: a pooled analysis of 1878 individuals.
Hepatology communications, 03-01, Volume: 7, Issue: 3
2023
Predictors of serious adverse events and non-response in cirrhotic patients with primary biliary cholangitis treated with obeticholic acid.
Liver international : official journal of the International Association for the Study of the Liver, Volume: 42, Issue: 11
2022
Efficacy and safety of obeticholic acid in liver disease-A systematic review and meta-analysis.
Clinics and research in hepatology and gastroenterology, Volume: 45, Issue: 3
2021
Effectiveness and safety of obeticholic acid in a Southern European multicentre cohort of patients with primary biliary cholangitis and suboptimal response to ursodeoxycholic acid.
Alimentary pharmacology & therapeutics, Volume: 53, Issue: 4
2021
The safety and effectiveness of chenodeoxycholic acid treatment in patients with cerebrotendinous xanthomatosis: two retrospective cohort studies.
Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, Volume: 41, Issue: 4
2020
REGENERATE: Design of a pivotal, randomised, phase 3 study evaluating the safety and efficacy of obeticholic acid in patients with fibrosis due to nonalcoholic steatohepatitis.
Contemporary clinical trials, Volume: 84
2019
Long-term efficacy and safety of obeticholic acid for patients with primary biliary cholangitis: 3-year results of an international open-label extension study.
The lancet. Gastroenterology & hepatology, Volume: 4, Issue: 6
2019
Are Clinicians Ready for Safe Use of Stratified Therapy in Primary Biliary Cholangitis (PBC)? A Study of Educational Awareness.
Digestive diseases and sciences, Volume: 63, Issue: 10
2018
Review article: therapeutic bile acids and the risks for hepatotoxicity.
Alimentary pharmacology & therapeutics, Volume: 47, Issue: 12
2018
Identification of Bile Acids Responsible for Inhibiting the Bile Salt Export Pump, Leading to Bile Acid Accumulation and Cell Toxicity in Rat Hepatocytes.
Journal of pharmaceutical sciences, Volume: 106, Issue: 9
2017
Activation of the Farnesoid X-receptor in breast cancer cell lines results in cytotoxicity but not increased migration potential.
Cancer letters, Jan-28, Volume: 370, Issue: 2
2016
Toxicity and intracellular accumulation of bile acids in sandwich-cultured rat hepatocytes: role of glycine conjugates.
Toxicology in vitro : an international journal published in association with BIBRA, Volume: 28, Issue: 2
2014
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
PLoS computational biology, Volume: 7, Issue: 12
2011
FXR activation improves myocardial fatty acid metabolism in a rodent model of obesity-driven cardiotoxicity.
Nutrition, metabolism, and cardiovascular diseases : NMCD, Volume: 23, Issue: 2
2013
Bile acid toxicity structure-activity relationships: correlations between cell viability and lipophilicity in a panel of new and known bile acids using an oesophageal cell line (HET-1A).
Bioorganic & medicinal chemistry, Sep-15, Volume: 18, Issue: 18
2010
Developing structure-activity relationships for the prediction of hepatotoxicity.
Chemical research in toxicology, Jul-19, Volume: 23, Issue: 7
2010
Plasma bile acid concentrations in patients with human immunodeficiency virus infection receiving protease inhibitor therapy: possible implications for hepatotoxicity.
Pharmacotherapy, Volume: 30, Issue: 1
2010
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
Current drug discovery technologies, Volume: 1, Issue: 4
2004
Role of mitochondrial dysfunction in combined bile acid-induced cytotoxicity: the switch between apoptosis and necrosis.
Toxicological sciences : an official journal of the Society of Toxicology, Volume: 79, Issue: 1
2004
Bile acid composition in snake bile juice and toxicity of snake bile acids to rats.
Comparative biochemistry and physiology. Toxicology & pharmacology : CBP, Volume: 136, Issue: 3
2003
Effect of chenodeoxycholic acid on the toxicity of 5alpha-cyprinol sulfate in rats.
Toxicology, Sep-30, Volume: 179, Issue: 1-2
2002
The role of ursodeoxycholic acid in bile acid-mediated kidney fragment toxicity.
Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie, Volume: 51, Issue: 1
1999
Extrahepatic deposition and cytotoxicity of lithocholic acid: studies in two hamster models of hepatic failure and in cultured human fibroblasts.
Hepatology (Baltimore, Md.), Volume: 27, Issue: 2
1998
Cytotoxicity of bile salts against biliary epithelium: a study in isolated bile ductule fragments and isolated perfused rat liver.
Hepatology (Baltimore, Md.), Volume: 26, Issue: 1
1997
Toxicity order of cholanic acids using an immobilised cell biosensor.
Journal of pharmaceutical and biomedical analysis, Volume: 14, Issue: 8-10
1996
Toxicity of bile acids to colon cancer cell lines.
Cancer letters, Jul-16, Volume: 70, Issue: 3
1993
[In vitro analysis for cellular toxicity of polychlorinated biphenyls (PCBs) and 2,3,4,7,8-pentacholorodibenzofuran (PCDF) on PLC/PRF/5 cell proliferation (II)--The effects of ursodeoxycholic acid and chenodeoxycholic acid on cell toxicity].
Fukuoka igaku zasshi = Hukuoka acta medica, Volume: 84, Issue: 5
1993
Ursodeoxycholate protects oxidative mitochondrial metabolism from bile acid toxicity: dose-response study in isolated rat liver mitochondria.
Hepatology (Baltimore, Md.), Volume: 20, Issue: 6
1994
Toxicity of bile acids on the electron transport chain of isolated rat liver mitochondria.
Hepatology (Baltimore, Md.), Volume: 19, Issue: 2
1994
Hepatotoxicity of bile acids in rabbits: ursodeoxycholic acid is less toxic than chenodeoxycholic acid.
Laboratory investigation; a journal of technical methods and pathology, Volume: 46, Issue: 4
1982
Relationship between serum and biliary bile acids as an indicator of chenodeoxycholic and ursodeoxycholic acid-induced hepatotoxicity in the rhesus monkey.
Digestive diseases and sciences, Volume: 27, Issue: 5
1982
Chenodiol (chenodeoxycholic acid) for dissolution of gallstones: the National Cooperative Gallstone Study. A controlled trial of efficacy and safety.
Annals of internal medicine, Volume: 95, Issue: 3
1981
Prevention of toxicity to isolated hepatocytes by liposome entrapment of chenodeoxycholic acid.
Toxicology letters, Volume: 21, Issue: 1
1984
Investigations on the toxicity of bile salt solutions, Capmul 8210 and a bile salt-EDTA solution for common bile duct perfusion in dogs.
Digestion, Volume: 30, Issue: 1
1984
Comparative efficacy and side effects of ursodeoxycholic and chenodeoxycholic acids in dissolving gallstones. A double-blind controlled study.
Gastroenterology, Volume: 85, Issue: 6
1983
Gallstone dissolution treatment with a combination of chenodeoxycholic and ursodeoxycholic acids. Studies of safety, efficacy and effects on bile lithogenicity, bile acid pool, and serum lipids.
Digestive diseases and sciences, Volume: 31, Issue: 10
1986
Bile acid-induced liver toxicity: relation to the hydrophobic-hydrophilic balance of bile acids.
Medical hypotheses, Volume: 19, Issue: 1
1986
Tauroursodeoxycholate prevents taurolithocholate-induced cholestasis and toxicity in rat liver.
Journal of hepatology, Volume: 10, Issue: 3
1990
Efficacy and safety of ursodeoxycholic acid for dissolution of gallstone fragments: comparison with the combination of ursodeoxycholic acid and chenodeoxycholic acid.
Hepatology (Baltimore, Md.), Volume: 14, Issue: 6
1991
The lack of relationship between hepatotoxicity and lithocholic-acid sulfation in biliary bile acids during chenodiol therapy in the National Cooperative Gallstone Study.
Hepatology (Baltimore, Md.), Volume: 14, Issue: 3
1991
Toxicity of chenodeoxycholic acid in the rhesus monkey.
Gastroenterology, Volume: 69, Issue: 2
1975
Toxicity of chenodeoxycholic acid in the nonhuman primate.
Surgery, Volume: 77, Issue: 6
1975
Impaired lithocholate sulfation in the rhesus monkey: a possible mechanism for chenodeoxycholate toxicity.
Gastroenterology, Volume: 70, Issue: 6
1976
Lack of toxicity of chenodeoxycholic acid in the squirrel monkey.
Gastroenterology, Volume: 73, Issue: 2
1977
Studies on plasma cytotoxicity and liver regeneration in fulminant hepatic failure.
Ciba Foundation symposium, Issue: 55
1977
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Long-term Use (9)

ArticleYear
Differential effects of FXR or TGR5 activation in cholangiocarcinoma progression.
Biochimica et biophysica acta. Molecular basis of disease, Volume: 1864, Issue: 4 Pt B
2018
Emerging and future therapies for nonalcoholic steatohepatitis in adults.
Expert opinion on pharmacotherapy, Volume: 17, Issue: 14
2016
Polyneuropathy in cerebrotendinous xanthomatosis and response to treatment with chenodeoxycholic acid.
Journal of neurology, Volume: 260, Issue: 1
2013
High-dose ursodeoxycholic acid is associated with the development of colorectal neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis.
The American journal of gastroenterology, Volume: 106, Issue: 9
2011
15 alpha-hydroxylation of a bile acid analogue, sodium 3 alpha,7 alpha-dihydroxy-25,26-bishomo-5 beta-cholane-26-sulfonate in the hamster.
Journal of lipid research, Volume: 37, Issue: 6
1996
Current management of symptomatic gallstones.
Postgraduate medicine, Volume: 93, Issue: 1
1993
Serum bile acids in relation to disease activity and intake of dietary fibers in juvenile ulcerative colitis.
Digestion, Volume: 50, Issue: 3-4
1991
Hepatic toxicity in the rhesus monkey treated with chenodeoxycholic acid for 6 months: biochemical and ultrastructural studies.
Gastroenterology, Volume: 70, Issue: 1
1976
Chronic administration of chenodeoxycholic acid increases cholesterol saturation in bile in the dog.
European journal of clinical investigation, Volume: 9, Issue: 1
1979
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Pharmacokinetics (23)

ArticleYear
Differential effects of metformin-mediated BSEP repression on pravastatin and bile acid pharmacokinetics in humans: A randomized controlled trial.
Clinical and translational science, Volume: 15, Issue: 10
2022
A Sensitive HPLC-MS/MS Method for Determination of Obeticholic Acid in Human Plasma: Application to a Pharmacokinetic Study in Healthy Volunteers.
Journal of chromatographic science, Jul-12, Volume: 60, Issue: 6
2022
A novel LC-MS/MS method for simultaneous estimation of obeticholic acid, glyco-obeticholic acid, tauro-obeticholic acid in human plasma and its application to a pharmacokinetic study.
Journal of separation science, Volume: 44, Issue: 7
2021
Improvements in the Oral Absorption and Anticancer Efficacy of an Oxaliplatin-Loaded Solid Formulation: Pharmacokinetic Properties in Rats and Nonhuman Primates and the Effects of Oral Metronomic Dosing on Colorectal Cancer.
International journal of nanomedicine, Volume: 15
2020
An ultra-performance liquid chromatography-tandem mass spectrometry method for the determination of obeticholic acid in rat plasma and its application in preclinical pharmacokinetic studies.
Journal of chromatography. B, Analytical technologies in the biomedical and life sciences, Jul-15, Volume: 1121
2019
Assessment of Pharmacokinetic Interactions Between Obeticholic Acid and Caffeine, Midazolam, Warfarin, Dextromethorphan, Omeprazole, Rosuvastatin, and Digoxin in Phase 1 Studies in Healthy Subjects.
Advances in therapy, Volume: 34, Issue: 9
2017
The Associations between Circulating Bile Acids and the Muscle Volume in Patients with Non-alcoholic Fatty Liver Disease (NAFLD).
Internal medicine (Tokyo, Japan), Volume: 56, Issue: 7
2017
Treatment with the natural FXR agonist chenodeoxycholic acid reduces clearance of plasma LDL whilst decreasing circulating PCSK9, lipoprotein(a) and apolipoprotein C-III.
Journal of internal medicine, Volume: 281, Issue: 6
2017
Farnesoid X Receptor Activation Promotes Hepatic Amino Acid Catabolism and Ammonium Clearance in Mice.
Gastroenterology, Volume: 152, Issue: 6
2017
Effect of 12-oxochenodeoxycholate on the pharmacokinetics and pharmacodynamics of morphine 6-glucuronide in Wistar rats.
The Journal of pharmacy and pharmacology, Volume: 65, Issue: 4
2013
Pharmacodynamic and clinical endpoints for functional colonic disorders: statistical considerations.
Digestive diseases and sciences, Volume: 58, Issue: 2
2013
Chenodeoxycholate in females with irritable bowel syndrome-constipation: a pharmacodynamic and pharmacogenetic analysis.
Gastroenterology, Volume: 139, Issue: 5
2010
Clinical pharmacokinetics of therapeutic bile acids.
Clinical pharmacokinetics, Volume: 30, Issue: 5
1996
[Effect of cisapride on clearance of gallstone fragments after extracorporeal shockwave lithotripsy of gallstones].
Wiener klinische Wochenschrift, Volume: 106, Issue: 4
1994
Caffeine clearance in subjects with constitutional unconjugated hyperbilirubinemia.
The Italian journal of gastroenterology, Volume: 27, Issue: 3
1995
Gall bladder volume and serum bile acids in cholestasis of pregnancy.
British journal of obstetrics and gynaecology, Volume: 89, Issue: 1
1982
Simulation of the metabolism and enterohepatic circulation of endogenous deoxycholic acid in humans using a physiologic pharmacokinetic model for bile acid metabolism.
Gastroenterology, Volume: 93, Issue: 4
1987
Simulation of the metabolism and enterohepatic circulation of endogenous chenodeoxycholic acid in man using a physiological pharmacokinetic model.
European journal of clinical investigation, Volume: 16, Issue: 5
1986
Increased gallbladder residual volume in nonresponders to extracorporeal shock wave lithotripsy of gallbladder stones.
The American journal of gastroenterology, Volume: 87, Issue: 10
1992
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Bioavailability (29)

ArticleYear
Development of an orally delivered GLP-1 receptor agonist through peptide engineering and drug delivery to treat chronic disease.
Scientific reports, 11-18, Volume: 11, Issue: 1
2021
Improvements in the Oral Absorption and Anticancer Efficacy of an Oxaliplatin-Loaded Solid Formulation: Pharmacokinetic Properties in Rats and Nonhuman Primates and the Effects of Oral Metronomic Dosing on Colorectal Cancer.
International journal of nanomedicine, Volume: 15
2020
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Molecular pharmacology, Volume: 96, Issue: 5
2019
Ceftriaxone Absorption Enhancement for Noninvasive Administration as an Alternative to Injectable Solutions.
Antimicrobial agents and chemotherapy, Volume: 62, Issue: 12
2018
Enhanced oral absorption of pemetrexed by ion-pairing complex formation with deoxycholic acid derivative and multiple nanoemulsion formulations: preparation, characterization, and in vivo oral bioavailability and anticancer effect.
International journal of nanomedicine, Volume: 13
2018
Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities.
European journal of medicinal chemistry, Jul-15, Volume: 100
2015
Cyclodextrins improve oral absorption of a novel factor Xa inhibitor by interfering with interaction between the drug and bile acids in rats.
The Journal of pharmacy and pharmacology, Volume: 65, Issue: 11
2013
Probiotics decreased the bioavailability of the bile acid analog, monoketocholic acid, when coadministered with gliclazide, in healthy but not diabetic rats.
European journal of drug metabolism and pharmacokinetics, Volume: 37, Issue: 2
2012
Structural requirements of the ASBT by 3D-QSAR analysis using aminopyridine conjugates of chenodeoxycholic acid.
Bioconjugate chemistry, Nov-17, Volume: 21, Issue: 11
2010
Increasing ursodeoxycholic acid in the enterohepatic circulation of pigs through the administration of living bacteria.
The British journal of nutrition, Volume: 93, Issue: 4
2005
Increased acyclovir oral bioavailability via a bile acid conjugate.
Molecular pharmaceutics, Jan-12, Volume: 1, Issue: 1
2004
Gum arabic (GA) modifies paracellular water and electrolyte transport in the small intestine.
Digestive diseases and sciences, Volume: 48, Issue: 4
2003
Single or multiple dose ursodeoxycholic acid for cholestatic liver disease: biliary enrichment and biochemical response.
Journal of hepatology, Volume: 25, Issue: 6
1996
Permeation enhancement of octreotide by specific bile salts in rats and human subjects: in vitro, in vivo correlations.
British journal of pharmacology, Volume: 117, Issue: 1
1996
Clinical pharmacokinetics of therapeutic bile acids.
Clinical pharmacokinetics, Volume: 30, Issue: 5
1996
Intestinal absorption of bile acids in the rabbit: different transport rates in jejunum and ileum.
Gastroenterology, Volume: 110, Issue: 2
1996
Formation, absorption, and biotransformation of delta 6-lithocholenic acid in humans.
The American journal of physiology, Volume: 264, Issue: 1 Pt 1
1993
Intestinal solubilization, absorption, pharmacokinetics and bioavailability of chenodeoxycholic acid.
European journal of clinical investigation, Volume: 10, Issue: 4
1980
Absorption of 7-ketolithocholic acid in rat jejunum, ileum and colon.
Zeitschrift fur Gastroenterologie, Volume: 23, Issue: 12
1985
Absorption of urso- and chenodeoxycholic acid and their taurine and glycine conjugates in rat jejunum, ileum, and colon.
Digestion, Volume: 32, Issue: 1
1985
Colonic absorption of sulfated and nonsulfated bile acids in rat.
Digestion, Volume: 33, Issue: 1
1986
Intestinal absorption of ursodeoxycholic acid in patients with extrahepatic biliary obstruction and bile drainage.
Gastroenterology, Volume: 102, Issue: 3
1992
Effect of varying doses of chenodeoxycholic acid on bile lipid and biliary bile acid composition in gallstone patients: a dose-response study.
The American journal of digestive diseases, Volume: 22, Issue: 1
1977
Changes in biliary lipid and biliary bile acid composition in patients after administration of ursodeoxycholic acid.
Journal of lipid research, Volume: 19, Issue: 6
1978
Colonic absorption of unconjugated bile acids: perfusion studies in man.
Digestive diseases and sciences, Volume: 24, Issue: 7
1979
Intestinal oxalate absorption. I. Absorption in vitro.
Research in experimental medicine. Zeitschrift fur die gesamte experimentelle Medizin einschliesslich experimenteller Chirurgie, Aug-16, Volume: 171, Issue: 1
1977
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Dosage (64)

ArticleYear
Primary biliary cholangitis.
Lancet (London, England), 12-12, Volume: 396, Issue: 10266
2020
Improvements in the Oral Absorption and Anticancer Efficacy of an Oxaliplatin-Loaded Solid Formulation: Pharmacokinetic Properties in Rats and Nonhuman Primates and the Effects of Oral Metronomic Dosing on Colorectal Cancer.
International journal of nanomedicine, Volume: 15
2020
Therapeutic effects of the selective farnesoid X receptor agonist obeticholic acid in a monocrotaline-induced pulmonary hypertension rat model.
Journal of endocrinological investigation, Volume: 42, Issue: 8
2019
FXR-Dependent Modulation of the Human Small Intestinal Microbiome by the Bile Acid Derivative Obeticholic Acid.
Gastroenterology, Volume: 155, Issue: 6
2018
Obeticholic acid for severe bile acid diarrhea with intestinal failure: A case report and review of the literature.
World journal of gastroenterology, Jun-07, Volume: 24, Issue: 21
2018
Clarification of Obeticholic Acid Dosing: Response to "Occurrence of Jaundice Following Simultaneous Ursodeoxycholic Acid Cessation and Obeticholic Acid Initiation" by Quigley et al.
Digestive diseases and sciences, Volume: 63, Issue: 7
2018
A randomized trial of obeticholic acid monotherapy in patients with primary biliary cholangitis.
Hepatology (Baltimore, Md.), Volume: 67, Issue: 5
2018
Simultaneous Determination of Ursodeoxycholic Acid and Chenodeoxycholic Acid in Pharmaceutical Dosage Form by HPLC-UV Detection.
Journal of AOAC International, Jan-01, Volume: 100, Issue: 1
2017
Therapy of Primary Biliary Cirrhosis: Novel Approaches for Patients with Suboptimal Response to Ursodeoxycholic Acid.
Digestive diseases (Basel, Switzerland), Volume: 33 Suppl 2
2015
Hepatotoxicity due to chenodeoxycholic acid supplementation in an infant with cerebrotendinous xanthomatosis: implications for treatment.
European journal of pediatrics, Volume: 175, Issue: 1
2016
Nonalcoholic steatohepatitis as a novel player in metabolic syndrome-induced erectile dysfunction: an experimental study in the rabbit.
Molecular and cellular endocrinology, Mar-25, Volume: 384, Issue: 1-2
2014
Bile acids increase alveolar epithelial permeability via mitogen-activated protein kinase, cytosolic phospholipase A2 , cyclooxygenase-2, prostaglandin E2 and junctional proteins.
Respirology (Carlton, Vic.), Volume: 18, Issue: 5
2013
Chenodeoxycholic acid reduces intestinal permeability in newly weaned piglets.
Journal of animal science, Volume: 90 Suppl 4
2012
The UDCA dosage deficit: a fate shared with CDCA.
European journal of gastroenterology & hepatology, Volume: 14, Issue: 3
2002
Enzyme immunoassay for conjugated 7alpha-hydroxy-3-oxo-4-cholenoic acid in human urine.
Journal of immunoassay & immunochemistry, Volume: 22, Issue: 1
2001
Ursodeoxycholic acid administration in patients with cholestasis of pregnancy: effects on primary bile acids in babies and mothers.
Hepatology (Baltimore, Md.), Volume: 33, Issue: 3
2001
Comparison of the effects of bile acids on cell viability and DNA synthesis by rat hepatocytes in primary culture.
Biochimica et biophysica acta, Feb-21, Volume: 1500, Issue: 2
2000
The sensitivity of the lactulose/rhamnose gut permeability test.
European journal of clinical investigation, Volume: 29, Issue: 2
1999
Clinical pharmacokinetics of therapeutic bile acids.
Clinical pharmacokinetics, Volume: 30, Issue: 5
1996
Intestinal absorption of bile acids in the rabbit: different transport rates in jejunum and ileum.
Gastroenterology, Volume: 110, Issue: 2
1996
Protective action of putrescine against rat liver injury.
Scandinavian journal of gastroenterology, Volume: 29, Issue: 2
1994
Ursodeoxycholate protects oxidative mitochondrial metabolism from bile acid toxicity: dose-response study in isolated rat liver mitochondria.
Hepatology (Baltimore, Md.), Volume: 20, Issue: 6
1994
The effect of chenodeoxycholic acid on the development of aberrant crypt foci in the rat colon.
Cancer letters, Jan-30, Volume: 76, Issue: 2-3
1994
Dose-related effects of chenodeoxycholic acid in the rabbit colon.
Digestive diseases and sciences, Volume: 25, Issue: 6
1980
Effect of chenodiol on the small intestine. Unimpaired structure and function during therapy for gallstone dissolution.
JAMA, Dec-04, Volume: 246, Issue: 22
1981
[Internist therapy of cholelithiasis].
Fortschritte der Medizin, May-28, Volume: 99, Issue: 20
1981
Clinical studies on cheno- and ursodeoxycholic acid treatment for gallstone dissolution.
Hepato-gastroenterology, Volume: 27, Issue: 1
1980
Changes of bile acid and lipid composition in blood and bile in the clinical course after the initiation of cheno- and ursodeoxycholic acid therapy in patients with cholesterol gallstones.
The Tokai journal of experimental and clinical medicine, Volume: 7, Issue: 6
1982
Retrospective comparison of 'Cheno' and 'Urso' in the medical treatment of gallstones.
Gut, Volume: 23, Issue: 5
1982
Differing effects of ursodeoxycholic or chenodeoxycholic acid on biliary cholesterol saturation and bile acid metabolism in man. A dose-response study.
Digestive diseases and sciences, Volume: 27, Issue: 2
1982
Medical therapy for gallstones. 2. Initiation and monitoring.
Postgraduate medicine, Volume: 71, Issue: 5
1982
[Treatment of cholesterol gallstones with ursodesoxycholic acid (author's transl)].
La Nouvelle presse medicale, Feb-20, Volume: 11, Issue: 8
1982
Chenodeoxycholic acid: a review of its pharmacological properties and therapeutic use.
Drugs, Volume: 21, Issue: 2
1981
Effect of chenodeoxycholic acid on liver structure and function in man: a stereological and biochemical study.
Digestion, Volume: 20, Issue: 1
1980
Chenodeoxycholic acid administration monitored by serum bile acid profiles: a dose-response study.
Annals of clinical biochemistry, Volume: 20 (Pt 6)
1983
Management of gallstones. The new option of bile acid therapy.
Postgraduate medicine, Volume: 74, Issue: 5
1983
Comparison of a monitored dose with a standard dose of chenodeoxycholic acid for gallstone dissolution.
Digestion, Volume: 30, Issue: 4
1984
Ursodeoxycholic acid treatment of gallstones.
Seminars in liver disease, Volume: 3, Issue: 2
1983
Drug and treatment efficacy of chenodeoxycholic acid in 97 patients with cholelithiasis and increased surgical risk.
Digestive diseases and sciences, Volume: 28, Issue: 6
1983
Effect of oral chenodeoxycholic acid on bile acid kinetics and biliary lipid composition in women with cholelithiasis.
The Journal of clinical investigation, Volume: 52, Issue: 11
1973
Metabolism of lithocholic and chenodeoxycholic acids in the squirrel monkey.
Gastroenterology, Volume: 89, Issue: 3
1985
Mass spectrometry identification of biliary bile acids in bile from patients with gallstones before and during treatment with chenodeoxycholic acid. An ancillary study of the National Cooperative Gallstone Study.
The Journal of laboratory and clinical medicine, Volume: 105, Issue: 4
1985
Changes in electrical potential difference of rectal mucosa and of gallbladder bile constituents of dogs fed chenodeoxycholic acid.
Canadian journal of physiology and pharmacology, Volume: 64, Issue: 8
1986
Determination of chenodiol bioequivalence using an immobilized multi-enzyme bioluminescence technique.
Journal of pharmaceutical sciences, Volume: 75, Issue: 3
1986
Chenodeoxycholic acid therapy in erythrohepatic protoporphyria.
Journal of hepatology, Volume: 3, Issue: 3
1986
Medical management of cholesterol gallstones.
Drug intelligence & clinical pharmacy, Volume: 20, Issue: 2
1986
Effect of 17alpha-ethinylestradiol on biliary excretion of bile acids.
Biochimica et biophysica acta, Jul-14, Volume: 399, Issue: 1
1975
[Conjugation of chenodeoxycholic acid and cholic acid during passage through liver].
Schweizerische medizinische Wochenschrift, Mar-29, Volume: 105, Issue: 13
1975
Metabolism of chenodeoxycholic acid in hamsters.
Lipids, Volume: 11, Issue: 12
1976
[Summary of work session 3: Drug-induced dissolution of gallstones].
Zeitschrift fur Gastroenterologie, Volume: 14 Suppl
1976
[Morphologic investigations on the toxicity of orally applied CDCA in the liver, gastro intestinal tract, kidney and adrenal gland of the rat (author's transl)].
Research in experimental medicine. Zeitschrift fur die gesamte experimentelle Medizin einschliesslich experimenteller Chirurgie, Aug-16, Volume: 171, Issue: 1
1977
Effects of anionic surfactants on hamster small intestinal membrane structure and function: relationship to surface activity.
Gastroenterology, Volume: 73, Issue: 3
1977
Effect of varying doses of chenodeoxycholic acid on bile lipid and biliary bile acid composition in gallstone patients: a dose-response study.
The American journal of digestive diseases, Volume: 22, Issue: 1
1977
[Morphologic, animal experimental study on the question of sex specific liver lesions due to chenodesoxycholic acid].
Zeitschrift fur Gastroenterologie, Volume: 15, Issue: 1
1977
Changes in biliary lipid and biliary bile acid composition in patients after administration of ursodeoxycholic acid.
Journal of lipid research, Volume: 19, Issue: 6
1978
Chenotherapy for gallstone dissolution. II. Induced changes in bile composition and gallstone response.
JAMA, Mar-20, Volume: 239, Issue: 12
1978
[The influence of chenodeoxycholic acid and ursodeoxycholic acid on the hepatic structure of the rat (author's transl)].
Zeitschrift fur Gastroenterologie, Volume: 17, Issue: 4
1979
Influence of chenodeoxycholic acid on serum triglycerides in patients with primary hypertriglyceridemia.
International journal of clinical pharmacology and biopharmacy, Volume: 16, Issue: 11
1978
Chenotherapy for gallstone dissolution. I. Efficacy and safety.
JAMA, Mar-13, Volume: 239, Issue: 11
1978
Comparison of fixed doses of chenodeoxycholic acid for gallstone dissolution.
Lancet (London, England), May-27, Volume: 1, Issue: 8074
1978
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Interactions (8)

ArticleYear
CYP1A2 Downregulation by Obeticholic Acid: Usefulness as a Positive Control for the In Vitro Evaluation of Drug-Drug Interactions.
Journal of pharmaceutical sciences, Volume: 108, Issue: 12
2019
Rapid analysis of the Tanreqing injection by near-infrared spectroscopy combined with least squares support vector machine and Gaussian process modeling techniques.
Spectrochimica acta. Part A, Molecular and biomolecular spectroscopy, Jul-05, Volume: 218
2019
Investigation of Glycochenodeoxycholate Sulfate and Chenodeoxycholate Glucuronide as Surrogate Endogenous Probes for Drug Interaction Studies of OATP1B1 and OATP1B3 in Healthy Japanese Volunteers.
Pharmaceutical research, Volume: 34, Issue: 8
2017
Comparison on Response and Dissolution Rates Between Ursodeoxycholic Acid Alone or in Combination With Chenodeoxycholic Acid for Gallstone Dissolution According to Stone Density on CT Scan: Strobe Compliant Observation Study.
Medicine, Volume: 94, Issue: 50
2015
Probiotics decreased the bioavailability of the bile acid analog, monoketocholic acid, when coadministered with gliclazide, in healthy but not diabetic rats.
European journal of drug metabolism and pharmacokinetics, Volume: 37, Issue: 2
2012
Efficacy on anaplastic thyroid carcinoma of valproic acid alone or in combination with doxorubicin, a synthetic chenodeoxycholic acid derivative, or lactacystin.
International journal of oncology, Volume: 34, Issue: 5
2009
Prediction and identification of drug interactions with the human ATP-binding cassette transporter multidrug-resistance associated protein 2 (MRP2; ABCC2).
Journal of medicinal chemistry, Jun-12, Volume: 51, Issue: 11
2008
[Effect of proteolytic enzymes and bile salts combined with EDTA 4Na on the dissolution of calcium bilirubinate gallstones].
Nihon Geka Gakkai zasshi, Volume: 88, Issue: 1
1987
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]