Page last updated: 2024-12-09

somatostatin

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth

Cross-References

ID SourceID
PubMed CID16129706
CHEMBL ID1823872
CHEBI ID64628
SCHEMBL ID21361053
MeSH IDM0020152

Synonyms (47)

Synonym
somatostatin-1
einecs 254-186-5
somatostatine [inn-french]
ccris 3629
somatostatinum [inn-latin]
somatostatina [inn-spanish]
somatostatin, cyclic
NCGC00167137-01
cyclic somatostatin
cb6417646
cas_38916-34-6
15-28-somatostatin-28
bdbm81767
chebi:64628 ,
CHEMBL1823872
l-alanyl-n-[(4r,7s,10s,13s,16s,19s,22s,25s,28s,31s,34s,37r)-19,34-bis(4-aminobutyl)-31-(2-amino-2-oxoethyl)-13,25,28-tribenzyl-4-carboxy-10,16-bis[(1r)-1-hydroxyethyl]-7-(hydroxymethyl)-22-(1h-indol-3-ylmethyl)-6,9,12,15,18,21,24,27,30,33,36-undecaoxo-1,2
ala-gly-cyclo-[cys-lys-asn-phe-phe-trp-lys-thr-phe-thr-ser-cys]
l-alanylglycyl-l-cysteinyl-l-lysyl-l-asparaginyl-l-phenylalanyl-l-phenylalanyl-l-tryptophyl-l-lysyl-l-threonyl-l-phenylalanyl-l-threonyl-l-seryl-l-cysteine cyclic (3-14) disulfide
AKOS015994634
somatostatinum
somatostatine
somatostatin (sheep)
6e20216q0l ,
unii-6e20216q0l
somatostatina
synthetic somatostatin-14
somatostatin (pigeon)
sr 9357
somatostatin [inn:ban]
somatostatin (human)
somatostatin (rat)
BRD-K14681867-015-01-6
HS-2027
NHXLMOGPVYXJNR-ATOGVRKGSA-N
DB09099
somatostatin-14 (h-l-ala-l-gly-l-cys(1)-l-lys-l-asn-l-phe-l-phe-l-trp-l-lys-l-thr-l-phe-l-thr-l-ser-l-cys(1)-oh)
SCHEMBL21361053
Q22075835
somatostatin [mart.]
somatostatin [inn]
somatostatin [ep monograph]
ala-gly-cys-lys-asn-phe-phe-trp-lys-thr-phe-thr-ser-cys 3,14-disulfide
somatostatin [mi]
somatostatin [who-dd]
somatostatin-14srif-14
C74981
AC-8931

Research Excerpts

Bioavailability

ExcerptReferenceRelevance
", ip) indicate that the liver is the primary site of biotransformation of the compound, suggesting that both 22a and its metabolite(s) are active, compensating probably low bioavailability of the parent molecule."( Design, physico-chemical properties and biological evaluation of some new N-[(phenoxy)alkyl]- and N-{2-[2-(phenoxy)ethoxy]ethyl}aminoalkanols as anticonvulsant agents.
Bednarski, M; Gunia-Krzyżak, A; Marona, H; Nitek, W; Pękala, E; Powroźnik, B; Słoczyńska, K; Walczak, M; Waszkielewicz, AM; Żesławska, E, 2016
)
0.43

Dosage Studied

ExcerptRelevanceReference
"8 nM (where K(B) is the concentration of antagonist that shifts the agonist dose-response 2-fold)."( Identification of potent non-peptide somatostatin antagonists with sst(3) selectivity.
Bigg, D; Contour-Galcéra, MO; Lannoy, J; Moinet, C; Plas, P; Poitout, L; Pommier, J; Roubert, P; Thurieau, C, 2001
)
0.31
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Drug Classes (2)

ClassDescription
heterodetic cyclic peptideA heterodetic cyclic peptide is a peptide consisting only of amino-acid residues, but in which the linkages forming the ring are not solely peptide bonds; one or more is an isopeptide, disulfide, ester, or other bond.
peptide hormoneAny peptide with hormonal activity in animals, whether endocrine, neuroendocrine, or paracrine.
[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 (1)

PathwayProteinsCompounds
GPCRs, class A rhodopsin-like03

Protein Targets (19)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
histone acetyltransferase KAT2A isoform 1Homo sapiens (human)Potency39.81070.251215.843239.8107AID504327
[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)
Somatostatin receptor type 5Mus musculus (house mouse)IC50 (µMol)0.00010.00010.00010.0001AID1285665
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)133.00000.63154.45319.3000AID1473740
Multidrug resistance-associated protein 4Homo sapiens (human)IC50 (µMol)133.00000.20005.677410.0000AID1473741
Bile salt export pumpHomo sapiens (human)IC50 (µMol)133.00000.11007.190310.0000AID1473738
Protein-lysine 6-oxidaseHomo sapiens (human)IC50 (µMol)0.01000.01001.19705.0000AID242522
Somatostatin receptor type 1Homo sapiens (human)IC50 (µMol)0.00120.00010.00130.0022AID1071309; AID242514; AID254725; AID616817; AID752208
Somatostatin receptor type 1Homo sapiens (human)Ki0.00070.00030.41232.3920AID1061947; AID203262; AID203386; AID203390; AID227836; AID254314
Somatostatin receptor type 1Mus musculus (house mouse)IC50 (µMol)0.00010.00010.00010.0001AID1285665
Somatostatin receptor type 2Homo sapiens (human)IC50 (µMol)0.00070.00040.00240.0140AID1071308; AID242516; AID254725; AID254874; AID616818
Somatostatin receptor type 2Homo sapiens (human)Ki0.00010.00000.40914.7200AID1061946; AID203087; AID203400; AID203404; AID203409; AID227837; AID238338; AID254315
Somatostatin receptor type 2Mus musculus (house mouse)IC50 (µMol)0.00020.00010.00020.0003AID1285665; AID203221
Somatostatin receptor type 3Mus musculus (house mouse)IC50 (µMol)0.00010.00010.00010.0001AID1285665; AID203230
Somatostatin receptor type 5Rattus norvegicus (Norway rat)IC50 (µMol)0.00090.00090.00090.0009AID203253
Somatostatin receptor type 4Homo sapiens (human)IC50 (µMol)0.00120.00010.00210.0042AID1071306; AID1533639; AID203233; AID242520; AID254725; AID616820
Somatostatin receptor type 4Homo sapiens (human)Ki0.00120.00060.27333.2000AID1061944; AID203237; AID203563; AID203565; AID203570; AID227839; AID254317
Somatostatin receptor type 3Homo sapiens (human)IC50 (µMol)0.00200.00040.11100.8800AID1071307; AID242518; AID254725; AID254877; AID616819
Somatostatin receptor type 3Homo sapiens (human)Ki0.00050.00010.03200.1290AID1061945; AID203420; AID203547; AID203551; AID227838; AID254316
Delta-type opioid receptorRattus norvegicus (Norway rat)IC50 (µMol)2.00000.00030.38877.0000AID149513
Mu-type opioid receptorRattus norvegicus (Norway rat)IC50 (µMol)0.00600.00010.887410.0000AID151430
Somatostatin receptor type 5Homo sapiens (human)IC50 (µMol)0.00300.00020.04720.3930AID1071305; AID1533640; AID242522; AID254725; AID254878; AID616821
Somatostatin receptor type 5Homo sapiens (human)Ki0.01150.00010.44794.0700AID1061943; AID203251; AID203581; AID203584; AID203709; AID227840; AID238285; AID238339; AID254318
Somatostatin receptor type 4 Mus musculus (house mouse)IC50 (µMol)0.00010.00010.00010.0001AID1285665
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)133.00002.41006.343310.0000AID1473739
[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)
Somatostatin receptor type 2Homo sapiens (human)EC50 (µMol)0.00010.00000.00080.0023AID246214
Somatostatin receptor type 3Homo sapiens (human)EC50 (µMol)0.00080.00080.00080.0008AID616823
Somatostatin receptor type 5Homo sapiens (human)EC50 (µMol)0.00010.00010.00010.0002AID203243; AID246204
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (93)

Processvia Protein(s)Taxonomy
xenobiotic metabolic processATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
bile acid and bile salt transportATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transportATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
leukotriene transportATP-binding cassette sub-family C member 3Homo sapiens (human)
monoatomic anion transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transport across blood-brain barrierATP-binding cassette sub-family C member 3Homo sapiens (human)
prostaglandin secretionMultidrug resistance-associated protein 4Homo sapiens (human)
cilium assemblyMultidrug resistance-associated protein 4Homo sapiens (human)
platelet degranulationMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic metabolic processMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
bile acid and bile salt transportMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transportMultidrug resistance-associated protein 4Homo sapiens (human)
urate transportMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
cAMP transportMultidrug resistance-associated protein 4Homo sapiens (human)
leukotriene transportMultidrug resistance-associated protein 4Homo sapiens (human)
monoatomic anion transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
export across plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
transport across blood-brain barrierMultidrug resistance-associated protein 4Homo sapiens (human)
guanine nucleotide transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
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)
osteoblast differentiationProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of protein phosphorylationProtein-lysine 6-oxidaseHomo sapiens (human)
heart developmentProtein-lysine 6-oxidaseHomo sapiens (human)
response to xenobiotic stimulusProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of gene expressionProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of striated muscle tissue developmentProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of transforming growth factor beta receptor signaling pathwayProtein-lysine 6-oxidaseHomo sapiens (human)
peptidyl-lysine oxidationProtein-lysine 6-oxidaseHomo sapiens (human)
bone mineralizationProtein-lysine 6-oxidaseHomo sapiens (human)
lung developmentProtein-lysine 6-oxidaseHomo sapiens (human)
platelet-derived growth factor receptor-beta signaling pathwayProtein-lysine 6-oxidaseHomo sapiens (human)
ascending aorta developmentProtein-lysine 6-oxidaseHomo sapiens (human)
descending aorta developmentProtein-lysine 6-oxidaseHomo sapiens (human)
protein modification processProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of apoptotic processProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of megakaryocyte differentiationProtein-lysine 6-oxidaseHomo sapiens (human)
muscle cell cellular homeostasisProtein-lysine 6-oxidaseHomo sapiens (human)
elastic fiber assemblyProtein-lysine 6-oxidaseHomo sapiens (human)
blood vessel morphogenesisProtein-lysine 6-oxidaseHomo sapiens (human)
response to steroid hormoneProtein-lysine 6-oxidaseHomo sapiens (human)
negative regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionProtein-lysine 6-oxidaseHomo sapiens (human)
muscle cell developmentProtein-lysine 6-oxidaseHomo sapiens (human)
cell chemotaxisProtein-lysine 6-oxidaseHomo sapiens (human)
connective tissue developmentProtein-lysine 6-oxidaseHomo sapiens (human)
DNA biosynthetic processProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of bone developmentProtein-lysine 6-oxidaseHomo sapiens (human)
cellular response to chemokineProtein-lysine 6-oxidaseHomo sapiens (human)
regulation of platelet-derived growth factor receptor-beta signaling pathwayProtein-lysine 6-oxidaseHomo sapiens (human)
collagen fibril organizationProtein-lysine 6-oxidaseHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerSomatostatin receptor type 1Homo sapiens (human)
glutamate receptor signaling pathwaySomatostatin receptor type 1Homo sapiens (human)
spermatogenesisSomatostatin receptor type 1Homo sapiens (human)
negative regulation of cell population proliferationSomatostatin receptor type 1Homo sapiens (human)
cerebellum developmentSomatostatin receptor type 1Homo sapiens (human)
forebrain developmentSomatostatin receptor type 1Homo sapiens (human)
somatostatin signaling pathwaySomatostatin receptor type 1Homo sapiens (human)
response to starvationSomatostatin receptor type 1Homo sapiens (human)
cellular response to leukemia inhibitory factorSomatostatin receptor type 1Homo sapiens (human)
cellular response to estradiol stimulusSomatostatin receptor type 1Homo sapiens (human)
neuropeptide signaling pathwaySomatostatin receptor type 1Homo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerSomatostatin receptor type 2Homo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwaySomatostatin receptor type 2Homo sapiens (human)
spermatogenesisSomatostatin receptor type 2Homo sapiens (human)
negative regulation of cell population proliferationSomatostatin receptor type 2Homo sapiens (human)
cerebellum developmentSomatostatin receptor type 2Homo sapiens (human)
peristalsisSomatostatin receptor type 2Homo sapiens (human)
forebrain developmentSomatostatin receptor type 2Homo sapiens (human)
somatostatin signaling pathwaySomatostatin receptor type 2Homo sapiens (human)
response to starvationSomatostatin receptor type 2Homo sapiens (human)
cellular response to glucocorticoid stimulusSomatostatin receptor type 2Homo sapiens (human)
cellular response to estradiol stimulusSomatostatin receptor type 2Homo sapiens (human)
neuropeptide signaling pathwaySomatostatin receptor type 2Homo sapiens (human)
G protein-coupled receptor signaling pathwaySomatostatin receptor type 4Homo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerSomatostatin receptor type 4Homo sapiens (human)
negative regulation of cell population proliferationSomatostatin receptor type 4Homo sapiens (human)
cell migrationSomatostatin receptor type 4Homo sapiens (human)
forebrain developmentSomatostatin receptor type 4Homo sapiens (human)
somatostatin signaling pathwaySomatostatin receptor type 4Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeSomatostatin receptor type 4Homo sapiens (human)
positive regulation of arachidonic acid secretionSomatostatin receptor type 4Homo sapiens (human)
negative regulation of adenylate cyclase-activating G protein-coupled receptor signaling pathwaySomatostatin receptor type 4Homo sapiens (human)
neuropeptide signaling pathwaySomatostatin receptor type 4Homo sapiens (human)
cellular response to glucocorticoid stimulusSomatostatin receptor type 4Homo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerSomatostatin receptor type 3Homo sapiens (human)
cell-cell signalingSomatostatin receptor type 3Homo sapiens (human)
spermatogenesisSomatostatin receptor type 3Homo sapiens (human)
negative regulation of cell population proliferationSomatostatin receptor type 3Homo sapiens (human)
hormone-mediated apoptotic signaling pathwaySomatostatin receptor type 3Homo sapiens (human)
cerebellum developmentSomatostatin receptor type 3Homo sapiens (human)
forebrain developmentSomatostatin receptor type 3Homo sapiens (human)
somatostatin signaling pathwaySomatostatin receptor type 3Homo sapiens (human)
response to starvationSomatostatin receptor type 3Homo sapiens (human)
cellular response to glucocorticoid stimulusSomatostatin receptor type 3Homo sapiens (human)
cellular response to estradiol stimulusSomatostatin receptor type 3Homo sapiens (human)
neuropeptide signaling pathwaySomatostatin receptor type 3Homo sapiens (human)
G protein-coupled receptor signaling pathwaySomatostatin receptor type 5Homo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerSomatostatin receptor type 5Homo sapiens (human)
negative regulation of cell population proliferationSomatostatin receptor type 5Homo sapiens (human)
positive regulation of cytokinesisSomatostatin receptor type 5Homo sapiens (human)
somatostatin signaling pathwaySomatostatin receptor type 5Homo sapiens (human)
cellular response to glucocorticoid stimulusSomatostatin receptor type 5Homo sapiens (human)
neuropeptide signaling pathwaySomatostatin receptor type 5Homo sapiens (human)
regulation of insulin secretionSomatostatin receptor type 5Homo sapiens (human)
xenobiotic metabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of gene expressionCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bile acid and bile salt transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
heme catabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic export from cellCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transepithelial transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
leukotriene transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
monoatomic anion transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (34)

Processvia Protein(s)Taxonomy
ATP bindingATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type bile acid transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATP hydrolysis activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
icosanoid transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
guanine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ATP bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type xenobiotic transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
urate transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
purine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type bile acid transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
efflux transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
15-hydroxyprostaglandin dehydrogenase (NAD+) activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATP hydrolysis activityMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
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)
protein-lysine 6-oxidase activityProtein-lysine 6-oxidaseHomo sapiens (human)
copper ion bindingProtein-lysine 6-oxidaseHomo sapiens (human)
protein bindingProtein-lysine 6-oxidaseHomo sapiens (human)
collagen bindingProtein-lysine 6-oxidaseHomo sapiens (human)
small molecule bindingProtein-lysine 6-oxidaseHomo sapiens (human)
molecular adaptor activityProtein-lysine 6-oxidaseHomo sapiens (human)
somatostatin receptor activitySomatostatin receptor type 1Homo sapiens (human)
protein bindingSomatostatin receptor type 1Homo sapiens (human)
neuropeptide bindingSomatostatin receptor type 1Homo sapiens (human)
protein bindingSomatostatin receptor type 2Homo sapiens (human)
PDZ domain bindingSomatostatin receptor type 2Homo sapiens (human)
somatostatin receptor activitySomatostatin receptor type 2Homo sapiens (human)
neuropeptide bindingSomatostatin receptor type 2Homo sapiens (human)
protein bindingSomatostatin receptor type 4Homo sapiens (human)
somatostatin receptor activitySomatostatin receptor type 4Homo sapiens (human)
neuropeptide bindingSomatostatin receptor type 4Homo sapiens (human)
somatostatin receptor activitySomatostatin receptor type 3Homo sapiens (human)
signaling receptor bindingSomatostatin receptor type 3Homo sapiens (human)
protein bindingSomatostatin receptor type 3Homo sapiens (human)
neuropeptide bindingSomatostatin receptor type 3Homo sapiens (human)
G protein-coupled receptor activitySomatostatin receptor type 3Homo sapiens (human)
protein bindingSomatostatin receptor type 5Homo sapiens (human)
neuropeptide bindingSomatostatin receptor type 5Homo sapiens (human)
somatostatin receptor activitySomatostatin receptor type 5Homo sapiens (human)
protein bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
organic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type xenobiotic transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP hydrolysis activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (26)

Processvia Protein(s)Taxonomy
plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basal plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basolateral plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
nucleolusMultidrug resistance-associated protein 4Homo sapiens (human)
Golgi apparatusMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
platelet dense granule membraneMultidrug resistance-associated protein 4Homo sapiens (human)
external side of apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
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)
extracellular regionProtein-lysine 6-oxidaseHomo sapiens (human)
extracellular spaceProtein-lysine 6-oxidaseHomo sapiens (human)
collagen trimerProtein-lysine 6-oxidaseHomo sapiens (human)
extracellular spaceProtein-lysine 6-oxidaseHomo sapiens (human)
collagen-containing extracellular matrixProtein-lysine 6-oxidaseHomo sapiens (human)
plasma membraneSomatostatin receptor type 1Homo sapiens (human)
plasma membraneSomatostatin receptor type 1Homo sapiens (human)
neuron projectionSomatostatin receptor type 1Homo sapiens (human)
cytosolSomatostatin receptor type 2Homo sapiens (human)
plasma membraneSomatostatin receptor type 2Homo sapiens (human)
neuron projectionSomatostatin receptor type 2Homo sapiens (human)
plasma membraneSomatostatin receptor type 2Homo sapiens (human)
plasma membraneSomatostatin receptor type 4Homo sapiens (human)
neuron projectionSomatostatin receptor type 4Homo sapiens (human)
plasma membraneSomatostatin receptor type 4Homo sapiens (human)
plasma membraneSomatostatin receptor type 3Homo sapiens (human)
ciliumSomatostatin receptor type 3Homo sapiens (human)
ciliary membraneSomatostatin receptor type 3Homo sapiens (human)
non-motile ciliumSomatostatin receptor type 3Homo sapiens (human)
neuron projectionSomatostatin receptor type 3Homo sapiens (human)
plasma membraneSomatostatin receptor type 3Homo sapiens (human)
plasma membraneSomatostatin receptor type 5Homo sapiens (human)
plasma membraneSomatostatin receptor type 5Homo sapiens (human)
neuron projectionSomatostatin receptor type 5Homo sapiens (human)
plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell surfaceCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
intercellular canaliculusCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (105)

Assay IDTitleYearJournalArticle
AID1071307Displacement of [125I]-[LTT]-SS28 from human SST3 receptor expressed in Chinese hamster CCL-39 cells after 2 hrs by autoradiographic analysis2014European journal of medicinal chemistry, Feb-12, Volume: 73[DOTA]Somatostatin-14 analogs and their (111)In-radioligands: effects of decreasing ring-size on sst1-5 profile, stability and tumor targeting.
AID254314Binding affinity towards somatostatin receptor type 12005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID203551Binding affinity towards human Somatostatin receptor type 3 (sst3) using Tyr11-[125I]-SRIF as radioligand was determined in CHO cells2003Journal of medicinal chemistry, Jun-05, Volume: 46, Issue:12
A novel somatostatin mimic with broad somatotropin release inhibitory factor receptor binding and superior therapeutic potential.
AID203253Tested for inhibition of radioligand binding to cloned somatostatin receptor rSSTR51997Journal of medicinal chemistry, Jul-04, Volume: 40, Issue:14
Lanthionine-somatostatin analogs: synthesis, characterization, biological activity, and enzymatic stability studies.
AID751845Binding affinity to somatostatin receptor (unknown origin) by radioligand displacement assay2013European journal of medicinal chemistry, May, Volume: 63Synthesis and structure-activity relationship studies in serotonin 5-HT(1A) receptor agonists based on fused pyrrolidone scaffolds.
AID203237In vitro binding affinity at somatostatin receptor 4 in transfected BHK cells using [125 I]Tyr11-SRIF-14 as radioligand1998Journal of medicinal chemistry, Nov-19, Volume: 41, Issue:24
Nonpeptide somatostatin agonists with sst4 selectivity: synthesis and structure-activity relationships of thioureas.
AID1882822Stability of the compound in simulated intestinal fluid at pH 6.8 assessed as half life measured up to 24 hrs by RP-HPLC-UV analysis2022Journal of medicinal chemistry, 04-28, Volume: 65, Issue:8
On the Utility of Chemical Strategies to Improve Peptide Gut Stability.
AID203386Inhibition of [125 I -Tyr]SRIF-14 binding to membranes isolated from CHO-K1 cells expressing cloned human SRIF receptor (sst-1) subtype2001Journal of medicinal chemistry, Aug-30, Volume: 44, Issue:18
Identification of potent non-peptide somatostatin antagonists with sst(3) selectivity.
AID238285Binding affinity against human sst5 receptor at a dose of 10 uM2005Bioorganic & medicinal chemistry letters, Aug-01, Volume: 15, Issue:15
3-Thio-1,2,4-triazoles, novel somatostatin sst2/sst5 agonists.
AID1071309Displacement of [125I]-[LTT]-SS28 from human SST1 receptor expressed in CHO cells after 2 hrs by autoradiographic analysis2014European journal of medicinal chemistry, Feb-12, Volume: 73[DOTA]Somatostatin-14 analogs and their (111)In-radioligands: effects of decreasing ring-size on sst1-5 profile, stability and tumor targeting.
AID203565Inhibition of [125 I -Tyr]SRIF-14 binding to membranes isolated from CHO-K1 cells expressing cloned human SRIF receptor (sst-4) subtype2001Journal of medicinal chemistry, Aug-30, Volume: 44, Issue:18
Identification of potent non-peptide somatostatin antagonists with sst(3) selectivity.
AID346461Displacement of radiolabeled somatostatin-14 from somatostatin receptor2008Journal of medicinal chemistry, Nov-27, Volume: 51, Issue:22
cis-4-(Piperazin-1-yl)-5,6,7a,8,9,10,11,11a-octahydrobenzofuro[2,3-h]quinazolin-2-amine (A-987306), a new histamine H4R antagonist that blocks pain responses against carrageenan-induced hyperalgesia.
AID203230Tested for inhibition of radioligand binding to cloned somatostatin receptor mSSTR31997Journal of medicinal chemistry, Jul-04, Volume: 40, Issue:14
Lanthionine-somatostatin analogs: synthesis, characterization, biological activity, and enzymatic stability studies.
AID254318Binding affinity towards somatostatin receptor type 52005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID680110TP_TRANSPORTER: uptake in membrane vesicles prepared from human MDR1 transfected KB3-1 cells2004FEBS letters, Sep-10, Volume: 574, Issue:1-3
Transport of somatostatin and substance P by human P-glycoprotein.
AID1473740Inhibition of human MRP3 overexpressed in Sf9 insect cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 10 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1071306Displacement of [125I]-[LTT]-SS28 from human SST4 receptor expressed in Chinese hamster CCL-39 cells after 2 hrs by autoradiographic analysis2014European journal of medicinal chemistry, Feb-12, Volume: 73[DOTA]Somatostatin-14 analogs and their (111)In-radioligands: effects of decreasing ring-size on sst1-5 profile, stability and tumor targeting.
AID252848Relative potency of compound to inhibit growth hormone secretion in rat anterior pituitary cells2005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
AID254317Binding affinity towards somatostatin receptor type 42005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID1381539Agonist activity at SSTR4 (unknown origin) expressed in HEK293 cells assessed as inhibition of forskolin-induced CRE promoter activity at 10 to 1000 nM by luciferase reporter gene assay2018Bioorganic & medicinal chemistry, 07-30, Volume: 26, Issue:13
New somatostatin-drug conjugates for effective targeting pancreatic cancer.
AID616821Displacement of 125I-[LTT]-SRIF-28 from human sst5 receptor in HEK293 cells after 2 hrs by autoradiography2011Journal of medicinal chemistry, Sep-08, Volume: 54, Issue:17
N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function.
AID203251Inhibitory constant on human somatostatin receptor 52001Bioorganic & medicinal chemistry letters, Mar-12, Volume: 11, Issue:5
Synthesis of substituted imidazopyrazines as ligands for the human somatostatin receptor subtype 5.
AID203390Binding affinity towards human Somatostatin receptor type 1 (sst1) using Tyr11-[125I]-SRIF as radioligand was determined in CHO cells2003Journal of medicinal chemistry, Jun-05, Volume: 46, Issue:12
A novel somatostatin mimic with broad somatotropin release inhibitory factor receptor binding and superior therapeutic potential.
AID1061943Displacement of [125I]-somatostatin from human SSTR5 expressed in CHO-K1 cells2014Bioorganic & medicinal chemistry letters, Jan-01, Volume: 24, Issue:1
A tetradecapeptide somatostatin dicarba-analog: Synthesis, structural impact and biological activity.
AID1533640Inhibition of human SSTR5 by radioligand binding assay2019European journal of medicinal chemistry, Feb-01, Volume: 163Ligand design for somatostatin receptor isoforms 4 and 5.
AID254725Binding affinity for SRIF (somatostatin) receptor2005Journal of medicinal chemistry, Sep-08, Volume: 48, Issue:18
Joys of molecules. 2. Endeavors in chemical biology and medicinal chemistry.
AID254316Binding affinity towards somatostatin receptor type 32005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID1473741Inhibition of human MRP4 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID203584Inhibition of [125 I -Tyr]SRIF-14 binding to membranes isolated from CHO-K1 cells expressing cloned human SRIF receptor (sst-5) subtype2001Journal of medicinal chemistry, Aug-30, Volume: 44, Issue:18
Identification of potent non-peptide somatostatin antagonists with sst(3) selectivity.
AID203547Inhibition of [125 I -Tyr]SRIF-14 binding to membranes isolated from CHO-K1 cells expressing cloned human SRIF receptor (sst-3) subtype2001Journal of medicinal chemistry, Aug-30, Volume: 44, Issue:18
Identification of potent non-peptide somatostatin antagonists with sst(3) selectivity.
AID203243Effective concentration on human somatostatin receptor 52001Bioorganic & medicinal chemistry letters, Mar-12, Volume: 11, Issue:5
Synthesis of substituted imidazopyrazines as ligands for the human somatostatin receptor subtype 5.
AID1285665Displacement of [125I]Tyr11-somatostatin-14 from somatostatin receptor in mouse AtT20 cells2016Bioorganic & medicinal chemistry, Apr-15, Volume: 24, Issue:8
Design, physico-chemical properties and biological evaluation of some new N-[(phenoxy)alkyl]- and N-{2-[2-(phenoxy)ethoxy]ethyl}aminoalkanols as anticonvulsant agents.
AID1381536Agonist activity at SSTR1 (unknown origin) expressed in HEK293 cells assessed as inhibition of forskolin-induced CRE promoter activity at 10 to 1000 nM by luciferase reporter gene assay2018Bioorganic & medicinal chemistry, 07-30, Volume: 26, Issue:13
New somatostatin-drug conjugates for effective targeting pancreatic cancer.
AID255815Inhibitory concentration on PRL release from rat pituitary cells2005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID680111TP_TRANSPORTER: uptake in membrane vesicles prepared from K562/ADM cells2004FEBS letters, Sep-10, Volume: 574, Issue:1-3
Transport of somatostatin and substance P by human P-glycoprotein.
AID752208Binding affinity to Somatostatin receptor type 1 (unknown origin) by radioligand displacement assay2013Bioorganic & medicinal chemistry, May-15, Volume: 21, Issue:10
Synthesis and biological evaluation of 2-(5-methyl-4-phenyl-2-oxopyrrolidin-1-yl)-acetamide stereoisomers as novel positive allosteric modulators of sigma-1 receptor.
AID242518Inhibition of 125I-[Leu8,D-Trp22,Tyr25]SRIF-28 binding to human somatostatin receptor type 32005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
AID203408Binding assay at human somatostatin receptor 2 (hsst2) expressed in CCL-39 cells using LTT-CST-14-(Tyr10) as radioligand.2001Journal of medicinal chemistry, Jul-19, Volume: 44, Issue:15
Peptide folding induces high and selective affinity of a linear and small beta-peptide to the human somatostatin receptor 4.
AID230500Ratio of receptor binding inhibitory concentrations against somatostatin receptors and mu opioid receptors from rat brain IC50 (vs [3H]-CGP-23,996) / IC50(vs [3H]naloxone)1986Journal of medicinal chemistry, Nov, Volume: 29, Issue:11
Design and synthesis of conformationally constrained somatostatin analogues with high potency and specificity for mu opioid receptors.
AID203221Tested for inhibition of radioligand binding to cloned somatostatin receptor mSSTR2b1997Journal of medicinal chemistry, Jul-04, Volume: 40, Issue:14
Lanthionine-somatostatin analogs: synthesis, characterization, biological activity, and enzymatic stability studies.
AID1533639Inhibition of human SSTR4 by radioligand binding assay2019European journal of medicinal chemistry, Feb-01, Volume: 163Ligand design for somatostatin receptor isoforms 4 and 5.
AID1061947Displacement of [125I]-somatostatin from human SSTR1 expressed in CHO-K1 cells2014Bioorganic & medicinal chemistry letters, Jan-01, Volume: 24, Issue:1
A tetradecapeptide somatostatin dicarba-analog: Synthesis, structural impact and biological activity.
AID203581Binding affinity towards Somatostatin receptor type 5 (hsst5)2002Journal of medicinal chemistry, Dec-19, Volume: 45, Issue:26
2002 Alfred Burger Award Address in Medicinal Chemistry. Natural products and design: interrelated approaches in drug discovery.
AID203570Binding affinity towards human Somatostatin receptor type 4 (sst4) using Tyr11-[125I]-SRIF as radioligand was determined in CHO cells2003Journal of medicinal chemistry, Jun-05, Volume: 46, Issue:12
A novel somatostatin mimic with broad somatotropin release inhibitory factor receptor binding and superior therapeutic potential.
AID1473739Inhibition of human MRP2 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1061946Displacement of [125I]-somatostatin from human SSTR2 expressed in CHO-K1 cells2014Bioorganic & medicinal chemistry letters, Jan-01, Volume: 24, Issue:1
A tetradecapeptide somatostatin dicarba-analog: Synthesis, structural impact and biological activity.
AID203087In vitro binding affinity at somatostatin 2 receptor in transfected HEK 293 cell using [125 I]Tyr11-SRIF-14 as radioligand1998Journal of medicinal chemistry, Nov-19, Volume: 41, Issue:24
Nonpeptide somatostatin agonists with sst4 selectivity: synthesis and structure-activity relationships of thioureas.
AID242516Inhibition of 125I-[Leu8,D-Trp22,Tyr25]SRIF-28 binding to human somatostatin receptor type 22005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
AID1882821Stability of the compound in simulated gastric fluid at pH 1.2 assessed as half life measured up to 24 hrs by RP-HPLC-UV analysis2022Journal of medicinal chemistry, 04-28, Volume: 65, Issue:8
On the Utility of Chemical Strategies to Improve Peptide Gut Stability.
AID203404Inhibition of [125 I -Tyr]SRIF-14 binding to membranes isolated from CHO-K1 cells expressing cloned human SRIF receptor (sst-2) subtype2001Journal of medicinal chemistry, Aug-30, Volume: 44, Issue:18
Identification of potent non-peptide somatostatin antagonists with sst(3) selectivity.
AID203262Binding affinity towards Somatostatin receptor type 1 (hsst1)2002Journal of medicinal chemistry, Dec-19, Volume: 45, Issue:26
2002 Alfred Burger Award Address in Medicinal Chemistry. Natural products and design: interrelated approaches in drug discovery.
AID1150982Retardation factor of the compound using 2.5% pyridine, 5% 1-butanol and 90% water at pH 7.4 buffer relative to lysine and alanine by ninhydrin spray1976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Tyrosylated analogues of somatostatin.
AID203569Binding assay at human somatostatin receptor 4 (hsst4) expressed in CCL-39 cells using LTT-CST-14-(Tyr10) as radioligand.2001Journal of medicinal chemistry, Jul-19, Volume: 44, Issue:15
Peptide folding induces high and selective affinity of a linear and small beta-peptide to the human somatostatin receptor 4.
AID616820Displacement of 125I-[LTT]-SRIF-28 from human sst4 receptor in CCL39 cells after 2 hrs by autoradiography2011Journal of medicinal chemistry, Sep-08, Volume: 54, Issue:17
N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function.
AID238338Inhibitory constant against human sst2 receptor at a dose of 10 uM2005Bioorganic & medicinal chemistry letters, Aug-01, Volume: 15, Issue:15
3-Thio-1,2,4-triazoles, novel somatostatin sst2/sst5 agonists.
AID203400Binding affinity towards Somatostatin receptor type 2 (hsst2)2002Journal of medicinal chemistry, Dec-19, Volume: 45, Issue:26
2002 Alfred Burger Award Address in Medicinal Chemistry. Natural products and design: interrelated approaches in drug discovery.
AID1473738Inhibition of human BSEP overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-taurocholate in presence of ATP measured after 15 to 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1061942Half life in plasma (unknown origin)2014Bioorganic & medicinal chemistry letters, Jan-01, Volume: 24, Issue:1
A tetradecapeptide somatostatin dicarba-analog: Synthesis, structural impact and biological activity.
AID246214Inhibition of forskolin-induced cAMP accumulation in CHO-K1 cells expressing human sst2 receptor2005Bioorganic & medicinal chemistry letters, Aug-01, Volume: 15, Issue:15
3-Thio-1,2,4-triazoles, novel somatostatin sst2/sst5 agonists.
AID616819Displacement of 125I-[LTT]-SRIF-28 from human sst3 receptor in CCL39 cells after 2 hrs by autoradiography2011Journal of medicinal chemistry, Sep-08, Volume: 54, Issue:17
N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function.
AID242522Inhibition of 125I-[Leu8,D-Trp22,Tyr25]SRIF-28 binding to human somatostatin receptor type 52005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
AID256253Percent inhibition on PRL release from rat pituitary cells2005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID1381540Agonist activity at SSTR5 (unknown origin) expressed in HEK293 cells assessed as inhibition of forskolin-induced CRE promoter activity at 10 to 1000 nM by luciferase reporter gene assay2018Bioorganic & medicinal chemistry, 07-30, Volume: 26, Issue:13
New somatostatin-drug conjugates for effective targeting pancreatic cancer.
AID203709Binding affinity towards human Somatostatin receptor type 5 (sst5) using Tyr11-[125I]-SRIF as radioligand was determined in COS cell2003Journal of medicinal chemistry, Jun-05, Volume: 46, Issue:12
A novel somatostatin mimic with broad somatotropin release inhibitory factor receptor binding and superior therapeutic potential.
AID242520Inhibition of 125I-[Leu8,D-Trp22,Tyr25]SRIF-28 binding to human somatostatin receptor type 42005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
AID255814Inhibitory concentration on GH release from rat pituitary cells2005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID227840Inhibition of human sst5 receptor expressed in CHO cells2001Bioorganic & medicinal chemistry letters, Apr-23, Volume: 11, Issue:8
Novel non-peptide ligands for the somatostatin sst3 receptor.
AID1381538Agonist activity at SSTR3 (unknown origin) expressed in HEK293 cells assessed as inhibition of forskolin-induced CRE promoter activity at 10 to 1000 nM by luciferase reporter gene assay2018Bioorganic & medicinal chemistry, 07-30, Volume: 26, Issue:13
New somatostatin-drug conjugates for effective targeting pancreatic cancer.
AID616818Displacement of 125I-[LTT]-SRIF-28 from human sst2 receptor in CCL39 cells after 2 hrs by autoradiography2011Journal of medicinal chemistry, Sep-08, Volume: 54, Issue:17
N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function.
AID227837Inhibition of human sst2 receptor expressed in CHO cells2001Bioorganic & medicinal chemistry letters, Apr-23, Volume: 11, Issue:8
Novel non-peptide ligands for the somatostatin sst3 receptor.
AID254874Inhibitory concentration against human somatostatin receptor type 2 in CC531 cells2005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID227836Inhibition of human sst1 receptor expressed in CHO cells2001Bioorganic & medicinal chemistry letters, Apr-23, Volume: 11, Issue:8
Novel non-peptide ligands for the somatostatin sst3 receptor.
AID203708Binding assay at human somatostatin receptor 5 (hsst5) expressed in CCL-39 cells using LTT-CST-14-(Tyr10) as radioligand.2001Journal of medicinal chemistry, Jul-19, Volume: 44, Issue:15
Peptide folding induces high and selective affinity of a linear and small beta-peptide to the human somatostatin receptor 4.
AID616823Agonist activity at human sst3 receptor expressed in CCL39 cells after 5 hrs by luciferase reporter gene assay2011Journal of medicinal chemistry, Sep-08, Volume: 54, Issue:17
N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function.
AID1071305Displacement of [125I]-[LTT]-SS28 from human SST5 receptor expressed in HEK293 cells after 2 hrs by autoradiographic analysis2014European journal of medicinal chemistry, Feb-12, Volume: 73[DOTA]Somatostatin-14 analogs and their (111)In-radioligands: effects of decreasing ring-size on sst1-5 profile, stability and tumor targeting.
AID1071308Displacement of [125I]-[LTT]-SS28 from human SST2 receptor expressed in Chinese hamster CCL-39 cells after 2 hrs by autoradiographic analysis2014European journal of medicinal chemistry, Feb-12, Volume: 73[DOTA]Somatostatin-14 analogs and their (111)In-radioligands: effects of decreasing ring-size on sst1-5 profile, stability and tumor targeting.
AID1336365Displacement of [125I]Tyr11-somatostatin-14 from Sst receptor in mouse AtT-20 cells measured after 60 mins by scintillation counting method2017Bioorganic & medicinal chemistry, 01-15, Volume: 25, Issue:2
Structure-anticonvulsant activity studies in the group of (E)-N-cinnamoyl aminoalkanols derivatives monosubstituted in phenyl ring with 4-Cl, 4-CH
AID203233Tested for inhibition of radioligand binding to cloned somatostatin receptor hSSTR41997Journal of medicinal chemistry, Jul-04, Volume: 40, Issue:14
Lanthionine-somatostatin analogs: synthesis, characterization, biological activity, and enzymatic stability studies.
AID151430Inhibition of binding of [3H]-naloxone to Opioid receptor mu 1 of rat brain membranes1986Journal of medicinal chemistry, Nov, Volume: 29, Issue:11
Design and synthesis of conformationally constrained somatostatin analogues with high potency and specificity for mu opioid receptors.
AID254315Binding affinity towards somatostatin receptor type 22005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID616817Displacement of 125I-[LTT]-SRIF-28 from human sst1 receptor in CHO cells after 2 hrs by autoradiography2011Journal of medicinal chemistry, Sep-08, Volume: 54, Issue:17
N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function.
AID149513Inhibition of [3H]-DPDPE binding to Opioid receptor delta 1 of rat brain membranes1986Journal of medicinal chemistry, Nov, Volume: 29, Issue:11
Design and synthesis of conformationally constrained somatostatin analogues with high potency and specificity for mu opioid receptors.
AID242514Inhibition of 125I-[Leu8,D-Trp22,Tyr25]SRIF-28 binding to human somatostatin receptor type 12005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
AID616822Induction of sst3 receptor internalization in human HEK cells after 30 mins by immunofluorescence microscopy2011Journal of medicinal chemistry, Sep-08, Volume: 54, Issue:17
N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function.
AID227839Inhibition of human sst4 receptor expressed in CHO cells2001Bioorganic & medicinal chemistry letters, Apr-23, Volume: 11, Issue:8
Novel non-peptide ligands for the somatostatin sst3 receptor.
AID231929Ratio calculated as IC50 of SSTR2b to that of rSSTR5.1997Journal of medicinal chemistry, Jul-04, Volume: 40, Issue:14
Lanthionine-somatostatin analogs: synthesis, characterization, biological activity, and enzymatic stability studies.
AID203550Binding assay at human somatostatin receptor 3 (hsst3) expressed in CCL-39 cells using LTT-CST-14-(Tyr10) as radioligand.2001Journal of medicinal chemistry, Jul-19, Volume: 44, Issue:15
Peptide folding induces high and selective affinity of a linear and small beta-peptide to the human somatostatin receptor 4.
AID1061945Displacement of [125I]-somatostatin from human SSTR3 expressed in CHO-K1 cells2014Bioorganic & medicinal chemistry letters, Jan-01, Volume: 24, Issue:1
A tetradecapeptide somatostatin dicarba-analog: Synthesis, structural impact and biological activity.
AID254878Inhibitory concentration against human somatostatin receptor type 5 in CHO-K1 cells2005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID256252Percent inhibition on GH release from rat pituitary cells2005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID195035Tested for inhibition of growth hormone (GH)release from primary cultures of male rat anterior pituitary cells1997Journal of medicinal chemistry, Jul-04, Volume: 40, Issue:14
Lanthionine-somatostatin analogs: synthesis, characterization, biological activity, and enzymatic stability studies.
AID1061944Displacement of [125I]-somatostatin from human SSTR4 expressed in CHO-K1 cells2014Bioorganic & medicinal chemistry letters, Jan-01, Volume: 24, Issue:1
A tetradecapeptide somatostatin dicarba-analog: Synthesis, structural impact and biological activity.
AID203409Binding affinity towards human Somatostatin receptor type 2 (sst2) using Tyr11-[125I]-SRIF as radioligand was determined in CHO cells2003Journal of medicinal chemistry, Jun-05, Volume: 46, Issue:12
A novel somatostatin mimic with broad somatotropin release inhibitory factor receptor binding and superior therapeutic potential.
AID187295In vitro relative potency for the inhibition of growth hormone release2003Journal of medicinal chemistry, Dec-18, Volume: 46, Issue:26
Design and synthesis of novel bioactive peptides and peptidomimetics.
AID340268Inhibition of somatostatin receptor2008Journal of medicinal chemistry, Jul-24, Volume: 51, Issue:14
Identification of a potent, selective, and orally active leukotriene a4 hydrolase inhibitor with anti-inflammatory activity.
AID252846Relative potency of compound to inhibit insulin secretion in rat anterior pituitary cells2005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
AID203389Binding assay at human somatostatin receptor 1 (hsst1) expressed in CCL-39 cells using LTT-CST-14-(Tyr10) as radioligand.2001Journal of medicinal chemistry, Jul-19, Volume: 44, Issue:15
Peptide folding induces high and selective affinity of a linear and small beta-peptide to the human somatostatin receptor 4.
AID238339Inhibitory constant against human sst5 receptor at a dose of 10 uM2005Bioorganic & medicinal chemistry letters, Aug-01, Volume: 15, Issue:15
3-Thio-1,2,4-triazoles, novel somatostatin sst2/sst5 agonists.
AID203420Binding affinity towards Somatostatin receptor type 3 (hsst3)2002Journal of medicinal chemistry, Dec-19, Volume: 45, Issue:26
2002 Alfred Burger Award Address in Medicinal Chemistry. Natural products and design: interrelated approaches in drug discovery.
AID1381537Agonist activity at SSTR2 (unknown origin) expressed in HEK293 cells assessed as inhibition of forskolin-induced CRE promoter activity at 10 to 1000 nM by luciferase reporter gene assay2018Bioorganic & medicinal chemistry, 07-30, Volume: 26, Issue:13
New somatostatin-drug conjugates for effective targeting pancreatic cancer.
AID254877Inhibitory concentration against human somatostatin receptor type 3 in CHO-K1 cells2005Journal of medicinal chemistry, Oct-20, Volume: 48, Issue:21
Discovery of iodinated somatostatin analogues selective for hsst2 and hsst5 with excellent inhibition of growth hormone and prolactin release from rat pituitary cells.
AID246204Inhibition of forskolin-induced cAMP accumulation in CHO-K1 cells expressing human sst5 receptor2005Bioorganic & medicinal chemistry letters, Aug-01, Volume: 15, Issue:15
3-Thio-1,2,4-triazoles, novel somatostatin sst2/sst5 agonists.
AID227838Inhibition of human sst3 receptor expressed in CHO cells2001Bioorganic & medicinal chemistry letters, Apr-23, Volume: 11, Issue:8
Novel non-peptide ligands for the somatostatin sst3 receptor.
AID203563Binding affinity towards Somatostatin receptor type 4 (hsst4)2002Journal of medicinal chemistry, Dec-19, Volume: 45, Issue:26
2002 Alfred Burger Award Address in Medicinal Chemistry. Natural products and design: interrelated approaches in drug discovery.
AID252847Relative potency of compound to inhibit glucagon secretion in rat anterior pituitary cells2005Journal of medicinal chemistry, Jan-27, Volume: 48, Issue:2
Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (29)

TimeframeStudies, This Drug (%)All Drugs %
pre-19902 (6.90)18.7374
1990's2 (6.90)18.2507
2000's14 (48.28)29.6817
2010's10 (34.48)24.3611
2020's1 (3.45)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 (%)
Trials0 (0.00%)5.53%
Reviews0 (0.00%)6.00%
Case Studies1 (3.45%)4.05%
Observational0 (0.00%)0.25%
Other28 (96.55%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (78)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Rediscovering Biomarkers for the Diagnosis and Early Treatment Response in NEN. REBORN Study [NCT04464122]60 participants (Anticipated)Observational2020-09-14Recruiting
A Randomized, Controlled Trial of Ligation Plus Vasoconstrictor vs.Ligation Plus Proton Pump Inhibitor in the Control of Acute Esophageal Variceal Bleeding [NCT01112852]Phase 4118 participants (Actual)Interventional2006-12-31Completed
Phase II Study of the IGF-1 Inhibitor Pasireotide Lar in Combination With the m-TOR Inhibitor Everolimus in Patients With Relapsed/Refractory Multiple Myeloma [NCT01234974]Phase 20 participants (Actual)Interventional2010-12-31Withdrawn(stopped due to Study never undertaken)
Continuing Somatostatin Analogues Upon Progression in Neuroendocrine Tumour pAtients - The SAUNA Trial [NCT05701241]Phase 4270 participants (Anticipated)Interventional2023-06-28Recruiting
Terlipressin Versus Somatostatin/Octreotide on Effect of Renal Function in Cirrhotic Patients With Acute Gastrointestinal Hemorrhage (TORCH): A Retrospective Multicenter Observational Study [NCT03846180]1,682 participants (Actual)Observational2019-03-01Completed
A Randomized, Multicenter, Phase II Study to Investigate Efficacy and Safety of ITF2984 in Acromegalic Patients [NCT02111044]Phase 248 participants (Actual)Interventional2014-04-30Completed
Evaluation of Postoperative Ascites After Somatostatin Infusion Following Hepatectomy for Hepatocellular Carcinoma: Multicenter Randomized Double-blind Placebo Controlled Trial (SOMAPROTECT01) [NCT02799212]Phase 3183 participants (Anticipated)Interventional2018-01-10Recruiting
Open Label Extension Study of Lanreotide Autogel 120 mg in Patients With Non-functioning Entero-pancreatic Endocrine Tumour [NCT00842348]Phase 389 participants (Actual)Interventional2009-02-28Completed
A Single-center, Randomized, Double Blind, Placebo-controlled Study to Evaluate the Efficacy and Safety of Somatostatin Used as Inflow Modulator in Liver Transplantation. [NCT01290172]Phase 133 participants (Actual)Interventional2010-12-31Completed
Effect of Liver Glycogen Content on Hypoglycemic Counterregulation [NCT03241706]Phase 140 participants (Anticipated)Interventional2018-08-02Recruiting
Comparative Effectiveness Research: Effects of Terlipressin When Usual Somatostatin or Octreotide Dose Fails to Treat the Patients With Acute Variceal Bleeding [NCT02311608]1,320 participants (Anticipated)Observational2014-02-28Recruiting
A Multi-center, Randomized, Controlled Study: Somatostatin Prevent Post-operation Pancreatic Fistula (POPF) in Intermediate Risk Patients After Pancreaticoduodenectomy [NCT03349424]Phase 4205 participants (Actual)Interventional2018-06-22Completed
Prospective, Multi-centers, Investigator Sponsored, Randomized Controlled Trial Towards Identifying the Effect of Somatostatin Treatment in Early Postoperative Simple Small Bowel Obstruction [NCT01082627]Phase 444 participants (Anticipated)Interventional2009-06-30Completed
A Double Blind Randomized Placebo-Controlled Trial of Somatostatin in Association With Endoscopic Variceal Ligation (EVL) in Control of Acute Variceal Bleeding [NCT01267669]61 participants (Actual)Interventional2005-11-30Completed
Reduction by Pasireotide of the Effluent Volume in High-output Enterostomy in Patients Refractory to Usual Medical Treatment : Phase II Multicentric Randomized Double Bland Placebo Controlled Study [NCT02713776]Phase 257 participants (Actual)Interventional2016-12-13Completed
An Open-label Phase I Study Evaluating the Safety and Tolerability of Pasireotide LAR in Combination With Everolimus in Advanced Metastatic NETs - The COOPERATE-1 Study [NCT01263353]Phase 136 participants (Actual)Interventional2010-11-30Completed
Measurement of Glucose Metabolism in Humans Using Magnetic Resonance at 4 Tesla. Substudy: Hypoglycemia Unawareness [NCT00793741]19 participants (Actual)Observational2007-12-31Completed
Evaluation of the Efficacity of Lanreotide LP 90 mg to Minimized the Lymphorrhea Post Axillary Lymphadenectomy in Breast Cancer [NCT00630695]Phase 3148 participants (Actual)Interventional2008-03-31Completed
A Phase 3, Prospective, Randomized, Double-blind, Multi-center Study of the Efficacy and Safety of Lanreotide Autogel®/Depot 120 mg Plus BSC vs. Placebo Plus BSC for Tumour Control in Subjects With Well Differentiated, Metastatic and/or Unresectable, Typi [NCT02683941]Phase 377 participants (Actual)Interventional2017-03-06Terminated(stopped due to National Comprehensive Cancer Network & European Neuroendocrine Tumor Society guidelines (2015/2016) led to prescription of somatostatin analogues (SSAs) in this setting, thereby limiting recruitment.)
The Effects of Endoscopy Combined With Partial Splenic Artery Embolization in the Treatment of Cirrhosis With Esophageal and Gastric Varices Complicated With Hyperplenism or Splenomegaly:A Randomized Controlled Study. [NCT05055713]368 participants (Anticipated)Interventional2020-09-25Enrolling by invitation
Phase II Study of [90Y-DOTA]-TOC and [177Lu-DOTA]-TOC in Metastasized Neuroendocrine Tumors [NCT00978211]Phase 21,499 participants (Actual)Interventional1997-09-30Completed
Partial Splenic Embolization Combined With Endoscopic Therapies and NSBB Decreases the Variceal Rebleeding Rate in Cirrhosis Patients With Hypersplenism: a Multicenter Randomized Controlled Trial [NCT02778425]108 participants (Actual)Interventional2016-02-29Completed
The Effect of Somatostatin in the Regulation of Velocity and Blood Flow of the Hepatic Circulation in Patients Undergoing Liver Resection [NCT04010669]Phase 340 participants (Anticipated)Interventional2019-08-01Not yet recruiting
Co-treatment With Pegvisomant and a Somatostatin Analogue (SA) in SA-responsive Acromegalic Patients: Impact on Insulin Sensitivity, Glucose Tolerance, and Pharmacoeconomics [NCT00652379]18 participants (Actual)Interventional2008-06-30Completed
Somatostatin as Inflow Modulator in Adult-to-adult Living Donor Liver Transplantation: a Randomized, Double-blind, Placebo-controlled Trial [NCT04107428]Phase 456 participants (Anticipated)Interventional2019-12-31Not yet recruiting
A Prospective, Multi-center, Investigator Sponsored, Randomized Controlled Trial-- The Prophylactic Effect of Stilamin on Post-ERCP Pancreatitis [NCT01431781]908 participants (Anticipated)Interventional2011-08-31Recruiting
The Metabolic Consequences of Gastrointestinal Surgery [NCT02836353]81 participants (Actual)Interventional2017-01-31Terminated(stopped due to Reached scientific goals, further recruitment would not further advance the findings of study.)
A Double Blind, Randomized Placebo Controlled Clinical Trial Investigating the Efficacy and Safety of Somatuline Depot (Lanreotide) Injection in the Treatment of Carcinoid Syndrome [NCT00774930]Phase 3115 participants (Actual)Interventional2009-05-31Completed
Peripheral Metabolic Effects of Intra Arterial Somatostatin Infusion in Healthy Controls [NCT01062529]8 participants (Actual)Interventional2009-10-31Completed
Prospective, Randomized, Double-Blind, Placebo-Controlled Multicenter Phase III-Study to Evaluate the Efficacy of Octreotide in Patients With Inoperable Hepatocellular Carcinoma [NCT00386984]Phase 3108 participants Interventional1999-10-31Completed
A Multicenter, Randomized, Double-Blind, Placebo Controlled Trial To Study The Efficacy And The Safety Of The Early Administration Of Somatostatin (Ucb) 12 Mg/24 H IV Infusion And Two IV Boluses Of 250 Mcg During 72 Hours In The Treatment Of Subjects With [NCT00152399]Phase 2370 participants Interventional2000-09-30Completed
A Single Center, Open Study to Evaluate Changes in Serum Electrolytes at Different Glucose Concentrations and Their Impact on a Non-invasive Glucose Monitoring Method [NCT01058577]Phase 120 participants (Actual)Interventional2001-12-31Completed
A Phase III Study of New Chemotherapy Regimen in the Treatment of Advanced Gallbladder Carcinoma [NCT01053390]Phase 3216 participants (Actual)Interventional2009-10-31Completed
Randomized, Double-blind Study to Evaluate the Efficacy of Administration With Intravenous Bolus Followed by a Continuous Infusion of Somatostatin in the Prevention of Post-ERCP Pancreatitis [NCT01060826]Phase 3510 participants (Anticipated)Interventional2009-05-31Recruiting
Application of Somatostatin for Advanced Gastric Cancer After D2 Lymph Node Dissection -a Prospective Randomized Controlled Study [NCT01914692]Phase 42 participants (Actual)Interventional2013-01-31Completed
Escalade or Deseacalade Antibiotic Use in Severe Acute Pancreatitis [NCT01992198]Phase 460 participants (Anticipated)Interventional2012-07-31Recruiting
RCT for the Effect of Early Administration of Vasoactive Substances When Combined With Endoscopic Treatment in Acute Gastro-esophageal Variceal Bleeds: Comparisons Among Terlipressin, Somatostatin, and Octreotide [NCT00966355]Phase 41,034 participants (Actual)Interventional2006-09-30Completed
Impact of Endogenous Estrogen on Somatostatin Inhibition and Growth Hormone Rebound in Older Women [NCT02026973]Phase 162 participants (Actual)Interventional2014-03-31Completed
The Effect of Somatostatin for Treatment of Post Hepatectomy Liver Failure (PHLF) [NCT02882347]Phase 320 participants (Anticipated)Interventional2015-07-31Recruiting
Fatigue and Quality of Life in Patients With Neuroendocrine Neoplasia [NCT04557137]100 participants (Anticipated)Observational2020-09-01Recruiting
The Effect of Acute Application of Pegvisomant Alone and in Combination With Octreotide on Endogenous GH Levels During a 6 Hour Test in Patients With Acromegaly on Constant Pegvisomant Treatment [NCT00595140]Phase 410 participants (Actual)Interventional2008-01-31Completed
Non-functioning Pancreatic Neuroendocrine Tumors (NF-pNETs) in Multiple Endocrine Neoplasia Type 1 (MEN1) Treated With Somatostatin Analogs (SA) Versus NO Treatment - a Prospective, Randomized, Controlled Multicenter Study [NCT02705651]Phase 3180 participants (Anticipated)Interventional2019-12-31Not yet recruiting
Short-course Somatostatin Versus Terlipressin Infusion in Combination With Endoscopic Variceal Ligation for the Prevention of Early Esophageal Variceal Rebleeding [NCT02757703]Phase 4150 participants (Actual)Interventional2010-05-31Completed
Treatment With Sandostatin in Patients With Castrate Resistance Prostate Cancer Showing Uptake of 68Ga-DOTATET [NCT02631616]Phase 130 participants (Anticipated)Interventional2016-02-29Not yet recruiting
An Open-Label Extension Trial of IONIS GHR-LRx, an Antisense Inhibitor of the Growth Hormone Receptor Administered Monthly Subcutaneously to Patients With Acromegaly Being Treated With Long-Acting Somatostatin Receptor Ligands (SRL) [NCT03967249]Phase 239 participants (Actual)Interventional2019-07-25Completed
Investigator Sponsored Study of Effects of Somatostatin on Post-endoscopic Portal Hemodynamic in Cirrhotic Patients With Esophageal Gastric Varices [NCT01426087]126 participants (Anticipated)Interventional2011-07-31Recruiting
A Phase III, Prospective, Multicenter, Randomized, Open, Parallel Group Comparison of Lanreotide Autogel® (90 and 120 mg) Administered by Deep Subcutaneous Injection Every Four Weeks, With Sandostatin LAR Depot (20 and 30 mg) Administered by Intramuscular [NCT00092287]Phase 34 participants (Actual)Interventional2004-07-31Terminated
Incidence of Medical and Nutritional Complications After Bariatric Surgery, Especially Focusing on Assessment and Treatment of Severe Hypoglycemia [NCT01865760]33 participants (Actual)Interventional2013-06-30Completed
A Within Group, Randomised, Phase I, Repeated Doses, Placebo and Octreotide Controlled Study in Healthy Volunteers to Investigate the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics After Incremental Doses of ITF2984 [NCT01871844]Phase 146 participants (Actual)Interventional2012-01-31Completed
The Effect of Lifestyle-induced Hepatic Steatosis on Glucagon-stimulated Amino Acid Turnover [NCT04859322]20 participants (Actual)Interventional2021-02-08Completed
Measurement of Glucose/Glycogen Metabolism in Humans Using Magnetic Resonance at 4 or 7 Tesla [NCT00786825]Phase 210 participants (Actual)Interventional2004-12-31Completed
Effect of a Long-acting Somatostatin on Disease Progression in Nephropathy Due to Autosomal Dominant Polycystic Kidney Disease: a Long-term Three Year Follow up Study [NCT00309283]Phase 378 participants (Actual)Interventional2006-04-30Completed
Effect of Elevated Plasma-Free-Fatty-Acids on Renal Hemodynamic Parameters [NCT00431665]9 participants Interventional1999-07-31Completed
The Protection of Telipression on Developing of Acute Kidney Injury in Cirrhotic Patients With Upper-gastroentestinal Bleeding [NCT04358016]Phase 454 participants (Actual)Interventional2018-01-01Completed
Effect of Portal Vein Thrombosis on the Prognosis of Liver Cirrhosis: A Single-center, Prospective, Cohort Study [NCT02335580]475 participants (Actual)Observational2014-12-31Completed
Phase II Study of Vatalanib and Octreotide in Patients With Progressive Low-Grade Neuroendocrine Tumors [NCT00227773]Phase 20 participants (Actual)InterventionalWithdrawn
A 3-Arm Randomized Phase II Trial Evaluating Single Agent and Combined Efficacy of Pasireotide and Everolimus in Adult Patients With Radioiodine-Refractory Differentiated and Medullary Thyroid Cancer [NCT01270321]Phase 242 participants (Actual)Interventional2010-11-30Completed
Randomized Control Trial Comparing Somatostatin Analogues With Perioperative Antibiotics Versus Prolonged Antibiotics [NCT04329039]Phase 2/Phase 3400 participants (Anticipated)Interventional2019-07-16Enrolling by invitation
Neurodegeneration as Early Event in Pathogenesis of Diabetic Retinopathy:Multicentric, Prospective, Ph. II-III,Random.Controlled Trial to Assess Efficacy of Neuroprotective Drugs Administered Topically to Prevent/Arrest Diabetic Retinopathy [NCT01726075]Phase 2/Phase 3450 participants (Actual)Interventional2013-02-28Completed
A Phase I, Exploratory, Intra-patient Dose Escalation Study to Investigate the Preliminary Safety, Pharmacokinetics, and Anti-tumor Activity of Pasireotide (SOM230) s.c.Followed by Pasireotide LAR in Patients With Metastaticmelanoma or Metastatic Merkel C [NCT01652547]Phase 110 participants (Actual)Interventional2012-11-30Completed
A Double-Blind, Randomized, Placebo-Controlled, 2-Way Cross-Over Study to Evaluate the Effect of Single Dose Canagliflozin on Kinetics of C-Peptide in Healthy Subjects [NCT01665638]Phase 110 participants (Actual)Interventional2012-09-30Completed
Randomized Phase 3 Trial Evaluating the Efficacy of Locoregional Treatment With Transarterial Embolization (TAE) for Liver Metastases, in Combination With Pharmacotherapy, in Patients With Neuroendocrine Tumor and Inoperable Liver Metastasis. [NCT01755182]Phase 31 participants (Actual)Interventional2013-07-31Terminated(stopped due to difficulty enrollment, no patients received treatment)
A Phase II Study of Ramucirumab With Somatostatin Analog Therapy in Patients With Advanced, Progressive Carcinoid Tumors [NCT02795858]Phase 244 participants (Actual)Interventional2016-06-14Active, not recruiting
IMMUNeOCT Study: Octreotide LAR in the Induction of Immunologic Response in Patient With Neuroendocrine Tumors: an Interventional Pharmacological Study [NCT04129255]Phase 234 participants (Actual)Interventional2017-06-28Completed
The DIPAK 1 Study: A Randomised, Controlled Clinical Trial Assessing the Efficacy of Lanreotide to Halt Disease Progression in ADPKD [NCT01616927]Phase 3300 participants (Anticipated)Interventional2012-06-30Active, not recruiting
Comparator Study of 68Ga-DOTATOC PET/CT With Octreoscan + High-resolution, Contrast-enhanced CT for Diagnosis and Staging in Neuroendocrine Tumors and Other Somatostatin Receptor Positive Tumors [NCT01869725]Phase 268 participants (Actual)Interventional2013-04-01Completed
A Phase III, Prospective, Randomised, Open Label Study to Compare the Efficacy and Safety of Lanreotide Autogel® 60, 90 or 120 mg With Lanreotide 40 mg PR in Subjects With Active Acromegaly [NCT02493517]Phase 3128 participants (Actual)Interventional2014-10-31Completed
Phase IIa, Open Label, Dose Ascending Study to Determine the Maximum Tolerated Dose, Safety and Tolerability, Pharmacokinetics and Pharmacodynamics of a Single Dose of Lanreotide PRF in Subjects With Acromegaly Previously Treated and Controlled With Eithe [NCT02396953]Phase 1/Phase 228 participants (Actual)Interventional2015-03-31Completed
Phase II Single Arm Study of Everolimus and Pasireotide (SOM230) in Patients With Advanced or Metastatic Hepatocellular Carcinoma (HCC) [NCT01488487]Phase 224 participants (Actual)Interventional2011-12-31Completed
An Open Label, Multi-center Pasireotide Roll-over Protocol for Patients Who Have Completed a Previous Novartis-sponsored Pasireotide Study and Are Judged by the Investigator to Benefit From Continued Pasireotide Treatment [NCT01794793]Phase 4413 participants (Actual)Interventional2013-06-10Active, not recruiting
A Two-part Phase I Study in Male Healthy Volunteers to Investigate the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics After Repeated Incremental Doses of ITF2984. Part A is Double-Blind, Randomized, Subcutaneous Administration. Part B is Open [NCT01897844]Phase 136 participants (Actual)Interventional2013-05-31Completed
Effect of Fasting Free Fatty Acids and Fasting Glucose on 1st and 2nd Phase Insulin Secretion [NCT03998709]Phase 40 participants (Actual)Interventional2020-02-01Withdrawn(stopped due to results from other study suggest no good reason to proceed)
Prevention of Postoperative Pancreatic Fistula by SOMATOSTATIN Compared to OCTREOTIDE: Prospective, Randomized, Controlled Study [NCT03000946]Phase 3655 participants (Actual)Interventional2017-05-15Completed
In Vivo Assessment of the Tricarboxylic Acid Cycle Flux in the Muscle and Splanchnic Bed of Humans: A Pilot Study [NCT02748369]Phase 117 participants (Actual)Interventional2016-07-31Completed
Multicenter 3-arm Trial to Evaluate the Efficacy and Safety of Pasireotide LAR or Everolimus Alone or in Combination in Patients With Well Differentiated Neuroendocrine Carcinoma of the Lung and Thymus - LUNA Trial [NCT01563354]Phase 2124 participants (Actual)Interventional2013-08-16Completed
Phase III, Open-Label, Multicenter International Study to Evaluate the Efficacy and Safety of an Octreotide Implant vs. Sandostatin LAR Depot in Patients With Acromegaly [NCT00765323]Phase 3169 participants (Actual)Interventional2008-09-30Terminated
A Multi-center, Randomized, Open-label, Phase IV Study to Investigate the Management of Pasireotide-induced Hyperglycemia With Incretin Based Therapy or Insulin in Adult Patients With Cushing's Disease or Acromegaly [NCT02060383]Phase 4249 participants (Actual)Interventional2014-05-23Completed
Direct Measurement of Proinsulin Clearance in Humans [NCT03998293]Phase 40 participants (Actual)Interventional2022-11-30Withdrawn(stopped due to Newly available data suggests that measuring proinsulin clearance would not enhance predictive value of serum proinsulin)
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00774930 (10) [back to overview]Average Frequency of Flushing Events (Per Day) Based on Subject Diary Records.
NCT00774930 (10) [back to overview]Percentage of Days of Use of Other Rescue Medication
NCT00774930 (10) [back to overview]Percentage of Days With Subcutaneous Octreotide as Rescue Medication
NCT00774930 (10) [back to overview]Proportion of Subjects Who Rolled Over Into the IOL Phase Before Completing the DB Phase of the Study
NCT00774930 (10) [back to overview]"Changes From Baseline in Gastrointestinal (G.I). Symptoms Subscore (Based on Items Q34, Q35, Q36, Q37 and Q38 of EORTC G.I. Neuroendocrine Tumour [NET] 21]"
NCT00774930 (10) [back to overview]"Changes From Baseline in Global Health Status/Quality of Life (QoL) Score (Based on Items 29 and 30 of the European Organisation for the Research and Treatment of Cancer Quality of Life Questionnaire [EORTC-QLQ] C30)"
NCT00774930 (10) [back to overview]"Changes From Baseline in QoL in Endocrine Symptoms Subscore (Assessed Based on Items Q31, Q32 and Q33 Using EORTC QLQ-G.I.NET-21 Questionnaires)"
NCT00774930 (10) [back to overview]Absolute Changes From Baseline in Biochemical Markers (Plasma Chromogranin A [CgA])
NCT00774930 (10) [back to overview]Absolute Changes From Baseline in Biochemical Markers (Urinary 5-hydroxyindoleacetic Acid [5-HIAA])
NCT00774930 (10) [back to overview]Average Frequency of Diarrhoea Events (Per Day) Based on Subject Diary Records.
NCT00786825 (1) [back to overview]Brain Glycogen Turnover Rate
NCT00842348 (2) [back to overview]Progression Free Survival (PFS): Kaplan-Meier Estimate
NCT00842348 (2) [back to overview]Adverse Events
NCT00966355 (2) [back to overview]5-day Treatment Failure (Failure to Control Bleeding, Rebleeding, or Death)
NCT00966355 (2) [back to overview]Active Bleeding During the First Endoscopic Exam, Needing Blood Transfusion for 5 Days, Experiencing Adverse Effects
NCT01270321 (2) [back to overview]Number of Participants With Progression-free Survival
NCT01270321 (2) [back to overview]Number of Participants With Response Per Responsive Evaluation Criteria in Solid Tumors Criteria (RECIST v1.0)
NCT01488487 (4) [back to overview]Number of Individuals Experiencing Toxicity
NCT01488487 (4) [back to overview]Time to Progression (TTP)
NCT01488487 (4) [back to overview]Overall Survival (OS)
NCT01488487 (4) [back to overview]Objective Response Rate (ORR)
NCT01563354 (12) [back to overview]Biochemical Response Rate (BRR) for 5HIAA Levels
NCT01563354 (12) [back to overview]Biochemical Response Rate (BRR) for Chromogranin A (CgA) Levels
NCT01563354 (12) [back to overview]Kaplan-Meier Estimates of Progression-free Survival (PFS)
NCT01563354 (12) [back to overview]Kaplan-Meier Event-free Probability Estimate Based on CgA Levels
NCT01563354 (12) [back to overview]Percentage of Participants Progression-free at 9 Months Based on Response Evaluation Criteria In Solid Tumors v1.1 (RECIST v1.1)
NCT01563354 (12) [back to overview]Summary of Duration of Response (Months)
NCT01563354 (12) [back to overview]Summary of Time to Response (Months)
NCT01563354 (12) [back to overview]Kaplan-Meier Event-free Probability Estimate for Biochemical Progression-free Survival Based on CgA Levels
NCT01563354 (12) [back to overview]Duration of Biochemical Response (DBR), by Treatment (Full Analysis Set)
NCT01563354 (12) [back to overview]Summary of Biochemical Progression-free Survival Based on CgA Levels by Treatment
NCT01563354 (12) [back to overview]Summary of Progression-free Survival (PFS) Based on RECIST v1.1
NCT01563354 (12) [back to overview]12-month Disease Control Rate (DCR) and Objective Response Rate (ORR)
NCT01869725 (3) [back to overview]Compare Sensitivity of 68Ga-DOTATOC PET/CT With Octreoscan
NCT01869725 (3) [back to overview]Compare Specificity of 68Ga-DOTATOC PET/CT With Octreoscan
NCT01869725 (3) [back to overview]Comparison of Conventional Imaging and Gallium Ga 68-edotreotide PET Using Concordance in Tumor Detection With Pathology
NCT02060383 (7) [back to overview]Change in HbA1c From Randomization (R) Over Time Per Randomized Arm
NCT02060383 (7) [back to overview]Change in FPG (Fasting Plasma Glucose) From Randomization Until End of Core Phase
NCT02060383 (7) [back to overview]Absolute Change in HbA1c From Baseline to End of Core Phase
NCT02060383 (7) [back to overview]Change in HbA1c From Randomization to Approximately 16 Weeks
NCT02060383 (7) [back to overview]Absolute Change in FPG From Baseline to End of Core Phase
NCT02060383 (7) [back to overview]Percentage of Participants With ≤ 0.3% HbA1c Increase to End of Core Phase
NCT02060383 (7) [back to overview]Percentage of Participants in the Incretin-based Arm Who Required Anti-diabetic Rescue Therapy With Insulin
NCT02396953 (17) [back to overview]PK Analysis of Lanreotide: Area Under the Serum Concentration-time Curve From Time 0 to 85 Days (AUC0-85).
NCT02396953 (17) [back to overview]PK Analysis of Lanreotide: Maximum Observed Serum Concentration (Cmax).
NCT02396953 (17) [back to overview]PK Analysis of Lanreotide: Time to Reach Maximum Serum Concentration (Tmax).
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Free Triiodothyroxine (FT3) and Free Thyroxine (FT4).
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Insulin-like Growth Factor 1 (IGF-1).
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Growth Hormone (GH).
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Insulin-like Growth Factor 1 (IGF-1).
NCT02396953 (17) [back to overview]Overall Summary of Number of Subjects With AEs.
NCT02396953 (17) [back to overview]PK Analysis of Glycofurol Excipients: Tmax.
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Insulin-like Growth Factor 1 (IGF-1).
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Prolactin.
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Thyroid Stimulating Hormone (TSH).
NCT02396953 (17) [back to overview]PK Analysis of Glycofurol Excipients: AUC0-∞ and Area Under the Serum Concentration Time Curve From Time 0 to Last Quantifiable Timepoint (AUC0-t).
NCT02396953 (17) [back to overview]PK Analysis of Glycofurol Excipients: Cmax.
NCT02396953 (17) [back to overview]PK Analysis of Lanreotide: Apparent Terminal Elimination Half-life (t1/2).
NCT02396953 (17) [back to overview]Determination of the Maximum Tolerated Dose (MTD) by Number of Subjects With DLTs.
NCT02396953 (17) [back to overview]PK Analysis of Lanreotide: Area Under the Serum Concentration-time Curve Extrapolated to Infinity (AUC0-∞).
NCT02493517 (9) [back to overview]Percentage of Subjects With at Least 20% Reduction in Tumour Volume at EOST/EW Visit Compared to Baseline
NCT02493517 (9) [back to overview]Percentage of Subjects With GH ≤2.5 Micrograms Per Litre (mcg/L) at the EOST/EW Visit
NCT02493517 (9) [back to overview]The Percentage of Subjects With GH ≤1 mcg/L at the EOST/EW Visit
NCT02493517 (9) [back to overview]Percentage of Subjects With Normal Age-adjusted IGF-1 Levels at the EOST/EW Visit
NCT02493517 (9) [back to overview]Standardised Mean Change From Baseline in Age-adjusted IGF-1 Levels at the EOST/EW Visit
NCT02493517 (9) [back to overview]Percentage of Subjects With Normal Age-adjusted IGF-1 Levels and Who Have GH Levels >1 mcg/L and ≤2.5 mcg/L at the EOST/EW Visit
NCT02493517 (9) [back to overview]Median Percentage Change From Baseline in Tumour Volume at the EOST/EW Visit
NCT02493517 (9) [back to overview]Mean Change From Baseline in GH Values at the EOST/EW Visit
NCT02493517 (9) [back to overview]Percentage of Subjects With at Least One Symptom of Acromegaly at Week 13 and at the EOST/EW Visit Compared to Baseline
NCT02683941 (12) [back to overview]Median PFS Time in the Double-Blind Phase, Assessed by Central Review
NCT02683941 (12) [back to overview]Median PFS Time in the Double-Blind Phase, Assessed by Local Review
NCT02683941 (12) [back to overview]Median Progression-Free Survival (PFS) Time in Subjects Randomised to Lanreotide in the Double-Blind Phase or Open-Label Treatment Phase, Assessed by Central Review
NCT02683941 (12) [back to overview]Percentage of Subjects Who Experienced QoL Deterioration
NCT02683941 (12) [back to overview]Percentage of Subjects With a Decrease of CgA ≥30% From Baseline at Week 8 in the Double-Blind Phase and Open-Label Treatment Phase
NCT02683941 (12) [back to overview]Mean Change From Baseline in the Biomarker Chromogranin A (CgA) in the Double-Blind Phase and Open-Label Treatment Phase
NCT02683941 (12) [back to overview]Mean Changes From Baseline in Urinary 5-hydroxyindoleacetic Acid (5-HIAA) Levels in the Double-Blind Phase and Open-Label Treatment Phase
NCT02683941 (12) [back to overview]Mean Changes From Baseline in Urinary 5-hydroxyindoleacetic Acid (5-HIAA) Levels in the Double-Blind Phase and Open-Label Treatment Phase
NCT02683941 (12) [back to overview]Mean Changes From Baseline in Quality of Life (QoL), as Assessed by the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (EORTC QLQ-C30) Global Health Status/QoL Score
NCT02683941 (12) [back to overview]Time to Treatment Failure (TTF) in the Double-Blind Phase
NCT02683941 (12) [back to overview]Mean Change From Baseline in the Biomarker Chromogranin A (CgA) in the Double-Blind Phase and Open-Label Treatment Phase
NCT02683941 (12) [back to overview]Objective Response Rate (ORR) in the Double-Blind Phase

Average Frequency of Flushing Events (Per Day) Based on Subject Diary Records.

(NCT00774930)
Timeframe: 16-week DB phase

InterventionNumber of events per day (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg0.92
Placebo1.75

[back to top]

Percentage of Days of Use of Other Rescue Medication

Usage of other concomitant rescue medications for diarrhoea and/or flushing events, measured as the percentage of days that the medications were used as rescue medication based on subject IVRS/IWRS diary records. Subjects were required to record the use and dose of s.c. octreotide, if any, as well as the use of other concomitant rescue medications (e.g. loperamide 2 mg tabs, and/or tincture of opium). (NCT00774930)
Timeframe: 16-week DB phase

InterventionPercentage of days (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg8.86
Placebo6.25

[back to top]

Percentage of Days With Subcutaneous Octreotide as Rescue Medication

Use of s.c. octreotide required to control symptoms associated with carcinoid syndrome, measured as the percentage of days that s.c. octreotide was used as rescue medication, based on subject Interactive Voice Response System (IVRS) or Interactive Web Response System (IWRS) diary records. (NCT00774930)
Timeframe: 16-week DB phase

InterventionPercentage of days (Least Squares Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg33.72
Placebo48.49

[back to top]

Proportion of Subjects Who Rolled Over Into the IOL Phase Before Completing the DB Phase of the Study

Subjects who rolled over early were those who received less than four DB injections before receiving the first IOL injection. (NCT00774930)
Timeframe: 16-week DB phase

InterventionPercentage of subjects (Number)
Lanreotide Autogel (Somatuline Depot) 120 mg18.6
Placebo21.4

[back to top]

"Changes From Baseline in Gastrointestinal (G.I). Symptoms Subscore (Based on Items Q34, Q35, Q36, Q37 and Q38 of EORTC G.I. Neuroendocrine Tumour [NET] 21]"

"Baseline is defined as the last non-missing observation obtained prior to the initiation of study treatment.~The QLQ-G.I.NET21 questionnaire contains 21 single items (Q31 to Q51) which are supplemental items to the EORTC QLQ-C30 questionnaire. Q31 to Q51 range from 1 to 4 with 1 being the most favourable answer and 4 the worst case (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much). Based on these items the scores were generated. All of the scores range in score from 0 to 100. A high score for a symptom scale represents a high level of symptomatology. The principle for scoring the scale is: Estimate the average of the items (I1, I2, ..., In) that contribute to the scale; this is the raw score. RS = (I1 + I2 +…+ In)/n.~For symptom scales: Score = {(RS - 1)/range} x 100, where range is the difference between the maximum possible value of RS and the minimum possible value of RS." (NCT00774930)
Timeframe: Baseline and Week 12 of DB phase

InterventionUnits on a scale (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg-4.06
Placebo0.10

[back to top]

"Changes From Baseline in Global Health Status/Quality of Life (QoL) Score (Based on Items 29 and 30 of the European Organisation for the Research and Treatment of Cancer Quality of Life Questionnaire [EORTC-QLQ] C30)"

"Baseline is defined as the last non-missing observation obtained prior to the initiation of study treatment.~Q29 and Q30 range from 1 (Very poor) to 7 (Excellent) with 1 being worst case and 7 the most favourable answer. Scores were derived according to the rules contained within the EORTC scoring manual. All of the scores range in score from 0 to 100. A high score for global health status/QoL represents high QoL. The principle for scoring the scale is: Estimate the average of the items (I1, I2, ..., In) that contribute to the scale; this is the raw score. Raw score = RS = (I1 + I2 +…+ In)/n.~For global health status/QoL: Score = {(RS - 1)/range} x 100, where range is the difference between the maximum possible value of RS and the minimum possible value of RS." (NCT00774930)
Timeframe: Baseline and Week 12 of DB phase

InterventionUnits on a scale (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg4.17
Placebo-1.72

[back to top]

"Changes From Baseline in QoL in Endocrine Symptoms Subscore (Assessed Based on Items Q31, Q32 and Q33 Using EORTC QLQ-G.I.NET-21 Questionnaires)"

"Baseline is defined as the last non-missing observation obtained prior to the initiation of study treatment.~The QLQ-G.I.NET21 questionnaire contains 21 single items (Q31 to Q51) which are supplemental items to the EORTC QLQ-C30 questionnaire. Q31 to Q51 range from 1 to 4 with 1 being the most favourable answer and 4 the worst case (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much). Based on these items the scores were generated. All of the scores range in score from 0 to 100. A high score for a symptom scale represents a high level of symptomatology. The principle for scoring the scale is: Estimate the average of the items (I1, I2, ..., In) that contribute to the scale; this is the raw score. RS = (I1 + I2 +…+ In)/n.~For symptom scales: Score = {(RS - 1)/range} x 100, where range is the difference between the maximum possible value of RS and the minimum possible value of RS." (NCT00774930)
Timeframe: Baseline and Week 12 of DB phase

InterventionUnits on a scale (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg-6.83
Placebo-2.69

[back to top]

Absolute Changes From Baseline in Biochemical Markers (Plasma Chromogranin A [CgA])

Baseline is defined as the last non-missing observation obtained prior to the initiation of study treatment. (NCT00774930)
Timeframe: Baseline and Week 12 of DB phase

Interventionμg/L (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg1125.8
Placebo801.5

[back to top]

Absolute Changes From Baseline in Biochemical Markers (Urinary 5-hydroxyindoleacetic Acid [5-HIAA])

Baseline is defined as the last non-missing observation obtained prior to the initiation of study treatment. (NCT00774930)
Timeframe: Baseline and Week 12 of DB phase

Interventionμmol/day (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg-201.4
Placebo36.3

[back to top]

Average Frequency of Diarrhoea Events (Per Day) Based on Subject Diary Records.

(NCT00774930)
Timeframe: 16-week DB phase

InterventionNumber of events per day (Mean)
Lanreotide Autogel (Somatuline Depot) 120 mg1.56
Placebo1.35

[back to top]

Brain Glycogen Turnover Rate

(NCT00786825)
Timeframe: 1 month

Interventionmg/kg/min (Mean)
Somatostatin0.31
Healthy Controls0.28

[back to top]

Progression Free Survival (PFS): Kaplan-Meier Estimate

"The time from randomisation in Study 726 to the first occurrence of either disease progression (measured using Response Evaluation Criteria In Solid Tumours [RECIST] criteria) or death in Study 726 or in Study 729, or equivalently, the Progression Free Survival (PFS) time.~Tumour assessments for the placebo group after switching to open label lanreotide Autogel were excluded for the purpose of this analysis. Estimation of the median was based on the Kaplan-Meier method." (NCT00842348)
Timeframe: Throughout the study (every 24 weeks and at completion/withdrawal visit)

Interventionweeks (Median)
Lanreotide Autogel - Randomised Treatment in Study 726154.14
Placebo - Randomised Treatment in Study 72672.00

[back to top]

Adverse Events

"Adverse events (AEs) that were ongoing from Study 726 at the time of entry into Study 729 were transcribed into the case report form (CRF) for Study 729 with a start date corresponding to the original report of this AE in Study 726. All new AEs that started after the last visit in Study 726 (i.e. irrespective of whether the AE had onset before or after giving informed consent for Study 729) were recorded Study 729.~An AE was considered as a treatment emergent adverse event (TEAE) for Study 729 if:~It was not present prior to receiving the first dose of study treatment in Study 729; or,~It was present prior to receiving the first dose of study treatment in Study 729 but the intensity increased after the first dose of study treatment in Study 729.~Adverse event data are presented in the AE section." (NCT00842348)
Timeframe: Throughout the study until the completion/early discontinuation visit.

Interventionparticipants with any TEAEs (Number)
Lanreotide Autogel40
Placebo46
Total86

[back to top]

5-day Treatment Failure (Failure to Control Bleeding, Rebleeding, or Death)

(NCT00966355)
Timeframe: 5 days after enrollment

Interventionparticipants (Number)
Octreotide218
Somatostatin216
Terlipressin225

[back to top]

Active Bleeding During the First Endoscopic Exam, Needing Blood Transfusion for 5 Days, Experiencing Adverse Effects

at least one of the three criteria (NCT00966355)
Timeframe: 5 days after enrollment

Interventionparticipants (Number)
Octreotide113
Somatostatin115
Terlipressin114

[back to top]

Number of Participants With Progression-free Survival

"Time from starting treatment until disease progression as defined using Response Evaluation CriteriaIn Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions, or death." (NCT01270321)
Timeframe: Through study completion, an average of 1 year

InterventionParticipants (Count of Participants)
Arm A (Everolimus Alone)17
Arm B (Pasireotide Alone)9
Arm C (Everolimus + Pasireotide)11

[back to top]

Number of Participants With Response Per Responsive Evaluation Criteria in Solid Tumors Criteria (RECIST v1.0)

"Number of participants with response per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR." (NCT01270321)
Timeframe: Through study completion, an average of 1 year

InterventionParticipants (Count of Participants)
Arm A (Everolimus Alone)17
Arm B (Pasireotide Alone)9
Arm C (Everolimus + Pasireotide)11

[back to top]

Number of Individuals Experiencing Toxicity

Safety determinations are based on the rate of drug-related adverse events (AEs) reported based upon the toxicity as measured by the NCI Common Terminology Criteria for Adverse Events version 4.0 (CTCAE v4.0). (NCT01488487)
Timeframe: 3.5 years

Interventionparticipants (Number)
Dose Limiting Toxicities (DLT)Any adverse event
Everolimus + Pasireotide124

[back to top]

Time to Progression (TTP)

Time to progression is defined as the time from study enrollment until radiological progression in a previously embolized lobe, development of new lesions in an untreated lobe, or evidence of extrahepatic progression (based on modified Hepatocellular Carcinoma (HCC) Response Evaluation Criteria In Solid Tumors (RECIST) criteria). Patients will be followed until death. Patients that die of causes unrelated to the study drug without evidence of progression will be censored. (NCT01488487)
Timeframe: 3.5 years

Interventionmonths (Median)
Everolimus + Pasireotide3.5

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from study enrollment until death. (NCT01488487)
Timeframe: 3.5 years

Interventionmonths (Median)
Everolimus + Pasireotide6.7

[back to top]

Objective Response Rate (ORR)

"ORR is the rate of complete responses (CRs) + partial responses (PRs) as determined by RECIST (v1.1) and modified HCC RECIST criteria. Responses defined as follows:~CR: disappearance of all clinical/radiological evidence of tumor including any intratumoral arterial enhancement in all target lesions.~Partial Response (PR): at least a 30% decrease in the sum of diameters of viable (contrast enhancement in the arterial phase) target lesions, referencing the baseline sum.~Stable Disease (SD): any cases that do not qualify for either partial response or progressive disease.~Progressive Disease (PD): an increase of at least 20% in the sum of the diameters of viable target lesions, referencing the nadir sum, and/or the appearance of one or more new lesions. A new hepatic nodule signals PD when the longest diameter is at least 10 mm and the nodule shows the typical vascular pattern of HCC on dynamic imaging or if at least 1-cm interval growth is seen in subsequent scans." (NCT01488487)
Timeframe: 3.5 years

Interventionpercentage of participants (Number)
Radiographic response (CR or PR)Stable disease
Everolimus + Pasireotide045.5

[back to top]

Biochemical Response Rate (BRR) for 5HIAA Levels

The percentages are the biochemical response rates i.e. percentage of patients showing normalization i.e. return to within normal ranges, or a decrease of >= 50% from baseline of 5HIAA concentrations. (NCT01563354)
Timeframe: Baseline up Week 52

,,
Interventionpercentage of participants (Number)
Week 12Week 24Week 36Week 48Week 52
Everolimus11.111.111.100
Pasireotide LAR20.05.05.05.05.0
Pasireotide LAR and Everolimus Combination10.020.05.05.010.0

[back to top]

Biochemical Response Rate (BRR) for Chromogranin A (CgA) Levels

Percentage of patients showing normalization or a decrease of ≥ 30% of serum CgA concentrations compared to baseline. (NCT01563354)
Timeframe: Baseline up to Week 52

,,
Interventionpercentage of participants (Number)
Week 12Week 24Week 36Week 48Week 52
Everolimus7.47.43.700
Pasireotide LAR20.68.88.88.85.9
Pasireotide LAR and Everolimus Combination17.120.011.411.45.7

[back to top]

Kaplan-Meier Estimates of Progression-free Survival (PFS)

Percent (%) event-free probability estimate is the estimated probability that a patient will remain event-free up to the specified time point. Percent event-free probability estimates are obtained from the Kaplan-Meier survival estimates. Events are time from first study drug administration to objective tumor progression or death from any cause according to RECIST v1.1. (NCT01563354)
Timeframe: Baseline, every 3 months up to 69 months

,,
Interventionevent free probability estimates (Number)
3 months6 months9 months12 months15 months18 months21 months24 months27 months30 months33 months36 months39 months42 months45 months48 months51 months54 months57 months60 months63 months66 months69 months
Everolimus91.263.556.950.246.838.629.419.619.69.89.89.89.89.89.89.8NANANANANANANA
Pasireotide LAR83.668.249.639.932.621.814.514.514.510.910.910.910.910.910.910.910.910.910.910.910.9NANA
Pasireotide LAR and Everolimus Combination88.685.579.255.551.242.738.028.528.519.019.014.214.214.214.214.214.214.27.17.17.17.17.1

[back to top]

Kaplan-Meier Event-free Probability Estimate Based on CgA Levels

Kaplan Meier estimates are for Duration of biochemical response (DBR) outcome measure. Events are biochemical progressions i.e. an increase of CgA levels >= 25% compared to baseline or deaths due to any cause. Percent (%) Event-free probability estimate is the estimated probability that a patient will remain event-free up to the specified time point. (NCT01563354)
Timeframe: Baseline, every 3 months up to Month 18

,,
Interventionevent free probability estimates (Number)
3 months6 months9 months12 months15 months18 months
Everolimus37.5NANANANANA
Pasireotide LAR75.056.356.356.337.537.5
Pasireotide LAR and Everolimus Combination77.877.844.444.444.444.4

[back to top]

Percentage of Participants Progression-free at 9 Months Based on Response Evaluation Criteria In Solid Tumors v1.1 (RECIST v1.1)

"Patients with Complete Response (CR), Partial Response (PR), or Stable Disease (SD) at Month 9 were to be considered as progression-free based on RECIST v1.1. Patients with missing tumor assessment, or with overall lesion response unknown at Month 9 were considered as non progression-free, unless any of the following assessments at Week 48 or Week 52 indicate CR, PR, or SD, in which case the patient was to be considered as progression-free at Month 9. Patients discontinuing the study for any reason prior to the 9 month assessment were to be considered as non progression-free." (NCT01563354)
Timeframe: Baseline up to 9 months

,,
Interventionpercentage of participants (Number)
Complete responsePartial responseStable diseaseProgression-free (PF) at Month 9
Everolimus02.431.033.3
Pasireotide LAR02.434.139.0
Pasireotide LAR and Everolimus Combination02.448.858.5

[back to top]

Summary of Duration of Response (Months)

Date of first objective tumor response to date of tumor progression or death due to any cause. (NCT01563354)
Timeframe: Every 3 months up to Year 1

,,
Interventionmonths (Number)
25th percentileMedian75th percentile
EverolimusNANANA
Pasireotide LARNANANA
Pasireotide LAR and Everolimus CombinationNANANA

[back to top]

Summary of Time to Response (Months)

Time from start of treatment to the first observed objective tumor response (partial response or complete response) observed according to RECIST v1.1. (NCT01563354)
Timeframe: Every 3 months up to Year 1

,,
Interventionmonths (Number)
25th percentileMedian75th percentile
EverolimusNANANA
Pasireotide LARNANANA
Pasireotide LAR and Everolimus CombinationNANANA

[back to top]

Kaplan-Meier Event-free Probability Estimate for Biochemical Progression-free Survival Based on CgA Levels

Percent (%) Event-free probability estimate is the estimated probability that a patient will remain event-free up to the specified time point. Percent event-free probability estimates are obtained from the Kaplan-Meier survival estimates. Events are biochemical progressions, i.e., an increase of CgA levels >= 25% compared to baseline or deaths due to any cause. (NCT01563354)
Timeframe: Baseline, every 3 months up to Month 24

,,
Interventionevent free probability estimates (Number)
3 months6 months9 months12 months15 months18 months21 months24 months
Everolimus35.417.711.07.4NANANANA
Pasireotide LAR43.129.518.518.518.513.813.8NA
Pasireotide LAR and Everolimus Combination77.144.529.726.418.118.118.118.1

[back to top]

Duration of Biochemical Response (DBR), by Treatment (Full Analysis Set)

Time from the first documentation of biochemical response to the first documentation of biochemical progression or to death due to any cause, whichever occurred first. Biochemical progression is defined as an increase of serum CgA levels ≥ 25% compared to baseline. (NCT01563354)
Timeframe: Baseline up to Month 18

Interventionmonths (Median)
Pasireotide LAR14.75
Everolimus2.00
Pasireotide LAR and Everolimus Combination8.38

[back to top]

Summary of Biochemical Progression-free Survival Based on CgA Levels by Treatment

Time from the first documentation of biochemical response to the first documentation of biochemical progression or to death due to any cause, whichever occurred first. Biochemical progression is defined as an increase of serum CgA levels ≥ 25% compared to baseline. (NCT01563354)
Timeframe: Baseline up Month 24

Interventionmonths (Median)
Pasireotide LAR2.89
Everolimus2.86
Pasireotide LAR and Everolimus Combination5.62

[back to top]

Summary of Progression-free Survival (PFS) Based on RECIST v1.1

Time from first study drug administration to objective tumor progression or death from any cause according to RECIST v1.1 (NCT01563354)
Timeframe: Baseline, every 3 months up to 69 months

Interventionmonths (Median)
Pasireotide LAR8.51
Everolimus12.48
Pasireotide LAR and Everolimus Combination16.53

[back to top]

12-month Disease Control Rate (DCR) and Objective Response Rate (ORR)

Objective response rate (ORR) was defined as the percentage of patients showing a best overall response (BOR) of CR or PR during the core study according to RECIST v1.1 criteria. The best overall response is interpreted as the best response recorded from the start of the treatment until disease progression/recurrence, death from any cause or until the patient withdraws consent, whichever is earliest. DCR was was defined as the percentage of participants with a best overall response of complete response, partial response or stable disease during 12 months of treatment according to RECIST v1.1. (NCT01563354)
Timeframe: Baseline up to Month 12

,,
Interventionpercentage of participants (Number)
Objective response (CR+PR)Disease control rate (CR+PR+SD)Complete response (CR)Partial response (PR)Stable diseaseProgressive diseaseUnknownNot assessedDiscontinued before month 12
Everolimus2.473.802.471.44.84.816.764.3
Pasireotide LAR2.480.502.478.014.62.42.468.3
Pasireotide LAR and Everolimus Combination4.978.004.973.27.3014.663.4

[back to top]

Compare Sensitivity of 68Ga-DOTATOC PET/CT With Octreoscan

Compare sensitivity of 68Ga-DOTATOC PET/CT with Octreoscan + high-resolution, contrast-enhanced CT for diagnosis and staging in neuroendocrine tumors and other somatostatin receptor positive tumors (NCT01869725)
Timeframe: Up to 6 months

Interventionpercentage of sensitivity (Number)
Sensitivity of Ga-68-DOTATOCSensitivity of OctreoScanSensitivity of Conventional Imaging (CI)
Diagnostic (Gallium Ga 68-edotreotide PET/CT)96.5579.3182.24

[back to top]

Compare Specificity of 68Ga-DOTATOC PET/CT With Octreoscan

Compare specificity of 68Ga-DOTATOC PET/CT with Octreoscan + high-resolution, contrast-enhanced CT for diagnosis and staging in neuroendocrine tumors and other somatostatin receptor positive tumors (NCT01869725)
Timeframe: 6 months

Interventionpercentage of specificity (Number)
Specificity of 68Ga-DOTATOCSpecificity of OctreoscanSpecificity of Conventional Imaging (CI)
Diagnostic (Gallium Ga 68-edotreotide PET/CT)10010047.82

[back to top]

Comparison of Conventional Imaging and Gallium Ga 68-edotreotide PET Using Concordance in Tumor Detection With Pathology

Tumor lesions detected on 68Ga-DOTATOC PET/CT compared with tumor lesions detected on Octreoscan SPECT imaging plus high-resolution, contrast-enhanced CT. (NCT01869725)
Timeframe: Up to 6 months between the timing of the Octreoscan SPECT/CT plus high-resolution, contrast-enhanced CT and the time of the 68Ga-DOTATOC PET/CT (either imaging type may occur first)

InterventionParticipants (Count of Participants)
Sensitivity and Specificity for Ga-68-DOTATOC as Compared to Pathology72023597Sensitivity and Specificity for OctreoScan as Compared to Pathology72023597
Total positiveFalse positionFalse negativeTotal negative
Diagnostic (Gallium Ga 68-edotreotide PET/CT)28
Diagnostic (Gallium Ga 68-edotreotide PET/CT)1
Diagnostic (Gallium Ga 68-edotreotide PET/CT)23
Diagnostic (Gallium Ga 68-edotreotide PET/CT)0
Diagnostic (Gallium Ga 68-edotreotide PET/CT)6
Diagnostic (Gallium Ga 68-edotreotide PET/CT)5

[back to top]

Change in HbA1c From Randomization (R) Over Time Per Randomized Arm

Absolute change in HbA1c overtime from randomization (i.e. start of randomized antidiabetic treatment) to end of core phase per randomized arm (NCT02060383)
Timeframe: Randomization (R), Week (W) 4 post R, W 8 post R, W 16 post R, end of Core phase (up to week 16 post R)

,
InterventionHbA1c percentage (Mean)
RandomizationChange at RW4 D29Change at RW8 D57Change at RW12 D85Change at RW16 D113End of Core Phase
Incretin Based Therapy (Randomized Group)7.10.50.30.20.00.0
Insulin (Randomized Group)7.10.50.50.40.30.3

[back to top]

Change in FPG (Fasting Plasma Glucose) From Randomization Until End of Core Phase

Absolute change in fasting glucose overtime from randomization (i.e. start of randomized antidiabetic treatment) to end of core phase per randomized arm (NCT02060383)
Timeframe: Randomization, R(randomization) Week 2, R-Week 4, R-Week 6, R-Week 8, R-Week 10, R-Week 12, R-Week 14, R-Week 16, end of Core phase

,
Interventionmg/dL (Mean)
RandomizationChange at RW2 D15Change at RW4 D29Change at RW6 D43Change at RW8 D57Change at RW10 D71Change at RW12 D85Change at RW14 D99Change at RW16 D113End of Core Phase
Incretin Based Therapy (Randomized Group)172.24.6-15.0-17.7-25.7-28.8-33.4-35.1-38.8-40.1
Insulin (Randomized Group)167.9-31.1-28.3-37.5-38.3-36.9-41.1-35.6-33.4-36.0

[back to top]

Absolute Change in HbA1c From Baseline to End of Core Phase

Absolute change in HbA1c from baseline to end of core phase in the incretin based therapy arm and the insulin arm (NCT02060383)
Timeframe: Baseline, up to 32 weeks (end of Core phase)

,,,,
InterventionHbA1c percentage (Mean)
Baseline: All PatientsChange at EOP: All PatientsBaseline: Cushing'sChange at EOP: Cushing'sBaseline: AcromegalyChange at EOP: Acromegaly
Baseline Insulin (BL) (Non-randomized Group)7.71.36.91.48.01.2
Incretin Based Therapy (Randomized Group)6.30.86.61.36.10.6
Insulin (Randomized Group)6.31.16.51.76.30.8
No OAD (Non-randomized Group)5.40.45.50.55.40.4
Oral Antidiabetic Drugs (OAD) (Non-randomized Group)5.70.85.90.95.60.7

[back to top]

Change in HbA1c From Randomization to Approximately 16 Weeks

Absolute change in HbA1c from randomization to end of core phase (16 weeks) in incretin based therapy arm and insulin arm, and mean difference of change in HbA1c between the two treatment groups based on an ANOVA model using treatment (Incretin, Insulin) and the two randomization stratification factors (Disease: Cushing's disease vs Acromegaly; Baseline glycemic status: HbA1c <7% vs HbA1c ≥ 7%) as fixed effects. For Participants who discontinued the study or required rescue treatment before the time of assessing the primary endpoint, the last HbA1c assessment collected 8 weeks (56 days) after randomization (and prior to or on the date of start of rescue treatment) was carried forward. If the participant discontinued the study or used rescue treatment within 8 weeks after randomization, it was considered missing. (NCT02060383)
Timeframe: Randomization, 16 weeks

,
InterventionHba1c percentage (Mean)
All PatientsCushing's DiseaseAcromegaly
Incretin Based Therapy (Randomized Group)-0.120.33-0.25
Insulin (Randomized Group)0.260.450.19

[back to top]

Absolute Change in FPG From Baseline to End of Core Phase

Absolute change in FPG from baseline to end of core phase in the incretin based therapy arm and the insulin arm. (NCT02060383)
Timeframe: Baseline, Up to 32 weeks (end of Core Phase)

,,,,
Interventionmg/dL (Mean)
Baseline: All PatientsChange at EOP: All PatientsBaseline: Cushing'sChange at EOP: Cushing'sBaseline: AcromegalyChange at EOP: Acromegaly
Baseline Insulin (BL) (Non-randomized Group)157.79.8147.221.3162.54.6
Incretin Based Therapy (Randomized Group)111.122.2117.913.4107.926.5
Insulin (Randomized Group)111.822.5106.336.4114.216.7
No OAD (Non-randomized Group)92.216.385.511.793.417.0
Oral Antidiabetic Drugs (OAD) (Non-randomized Group)97.222.993.315.898.825.8

[back to top]

Percentage of Participants With ≤ 0.3% HbA1c Increase to End of Core Phase

Percentage of participants with ≤ 0.3% HbA1c increase in the incretin based therapy arm and the insulin arm. (NCT02060383)
Timeframe: Randomization, up to 16 weeks

InterventionPercentage of participants (Number)
Incretin Based Therapy (Randomized Group)73.7
Insulin (Randomized Group)65.1

[back to top]

Percentage of Participants in the Incretin-based Arm Who Required Anti-diabetic Rescue Therapy With Insulin

The percentage of participants who received anti-diabetic rescue therapy in incretin based therapy is summarized. (NCT02060383)
Timeframe: Randomization to up to 16 weeks

InterventionPercentage of participants (Number)
Incretin Based Therapy (Randomized Group)31.6

[back to top]

PK Analysis of Lanreotide: Area Under the Serum Concentration-time Curve From Time 0 to 85 Days (AUC0-85).

"Blood samples for determination of lanreotide serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8,12 and 24 hours post-dose, on Days 3 and 5 and at Weeks 2, 3, 5, 9 and 13 after lanreotide PRF administration. Sample were also collected during follow-up at Weeks 17, 21 and 25 (or EW).~Mean serum lanreotide AUC0-85 values were determined using non-compartmental analysis." (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Day 85

Interventionng*day/mL (Mean)
Lanreotide PK Set: 180 mg Lanreotide PRF161
Lanreotide PK Set: 270 mg Lanreotide PRF179
Lanreotide PK Set: 360 mg Lanreotide PRF265

[back to top]

PK Analysis of Lanreotide: Maximum Observed Serum Concentration (Cmax).

"Blood samples for determination of lanreotide serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8,12 and 24 hours post-dose, on Days 3 and 5 and at Weeks 2, 3, 5, 9 and 13 after lanreotide PRF administration. Samples were also collected during follow-up at Weeks 17, 21 and 25 (or EW).~Mean serum lanreotide Cmax values were determined using non-compartmental analysis." (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

Interventionnanograms/millilitre (ng/mL) (Mean)
Lanreotide PK Set: 180 mg Lanreotide PRF19.0
Lanreotide PK Set: 270 mg Lanreotide PRF14.0
Lanreotide PK Set: 360 mg Lanreotide PRF20.5

[back to top]

PK Analysis of Lanreotide: Time to Reach Maximum Serum Concentration (Tmax).

"Blood samples for determination of lanreotide serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8,12 and 24 hours post-dose, on Days 3 and 5 and at Weeks 2, 3, 5, 9 and 13 after lanreotide PRF administration. Samples were also collected during follow-up at Weeks 17, 21 and 25 (or EW).~Median serum lanreotide Tmax values were determined using non-compartmental analysis." (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

Interventiondays (Median)
Lanreotide PK Set: 180 mg Lanreotide PRF0.250
Lanreotide PK Set: 270 mg Lanreotide PRF0.253
Lanreotide PK Set: 360 mg Lanreotide PRF0.250

[back to top]

PD Analysis: Mean Change From Baseline in Free Triiodothyroxine (FT3) and Free Thyroxine (FT4).

Blood samples were collected for the determination of FT3 and FT4 in serum at Baseline (pre-dose) and at Weeks 2, 5, 13 and 25 (or EW). Summary data for serum concentrations of FT3 and FT4 were calculated and the mean change from Baseline at each time point is presented. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

,,
Interventionpicomole/litre (L) (Mean)
FT3: Week 2FT3: Week 5FT3: Week 13FT3: Week 25 (EOS/EW)FT4: Week 2FT4: Week 5FT4: Week 13FT4: Week 25 (EOS/EW)
180 mg Lanreotide PRF-0.278-0.1320.1000.2680.501.181.690.81
270 mg Lanreotide PRF-0.164-0.1700.1910.295-0.020.331.801.06
360 mg Lanreotide PRF-0.3740.1700.4380.396-0.68-0.67-0.460.38

[back to top]

PD Analysis: Mean Change From Baseline in Insulin-like Growth Factor 1 (IGF-1).

Blood samples were collected for the determination of IGF-1 in serum at Baseline (pre-dose), 6 hours post-dose and at Weeks 5, 9 and 13. Samples were also collected during follow-up at Weeks 17, 21 and 25 (or EW). Serum concentrations of IGF-1 were calculated using the Immulite 2000 Platform for all subjects in the safety population. The production of the reagent kits was stopped by the vendor during the study. The old reagent kits were used for Cohorts 1 and 2 until their expiry date and then the kits were switched to a new reagent and used for remaining subjects in Cohorts 2 and 3. Summary data for serum concentrations of IGF-1 were obtained using both methods (old and new reagent) and the mean change from Baseline at each time point is presented. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

Interventionng/mL (Mean)
Old Reagent: 6 hours post-doseOld Reagent: Week 5Old Reagent: Week 9Old Reagent: Week 13Old Reagent: Week 17Old Reagent: Week 21Old Reagent: Week 25 (EOS/EW)New Reagent: 6 hours post-doseNew Reagent: Week 5New Reagent: Week 9New Reagent: Week 13New Reagent: Week 17New Reagent: Week 21New Reagent: Week 25 (EOS/EW)
270 mg Lanreotide PRF-7.127.971.4111.196.2135.5136.46.03.013.030.020.565.045.5

[back to top]

PD Analysis: Mean Change From Baseline in Growth Hormone (GH).

GH cycle assessments were performed by taking 5 samples in the morning (with a sample taken every 30 minutes for 2 hours) at Baseline (pre-dose), Week 5 and Week 13. Summary data for the mean of the 5 samplings of the GH cycle were generated and the mean change from Baseline at each time point is presented. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 13.

,,
Interventionng/mL (Mean)
Week 5Week 13
180 mg Lanreotide PRF0.2680.667
270 mg Lanreotide PRF0.3670.684
360 mg Lanreotide PRF-0.2280.003

[back to top]

PD Analysis: Mean Change From Baseline in Insulin-like Growth Factor 1 (IGF-1).

Blood samples were collected for the determination of IGF-1 in serum at Baseline (pre-dose), 6 hours post-dose and at Weeks 5, 9 and 13. Samples were also collected during follow-up at Weeks 17, 21 and 25 (or EW). Serum concentrations of IGF-1 were calculated using the Immulite 2000 Platform for all subjects in the safety population. The production of the reagent kits was stopped by the vendor during the study. The old reagent kits were used for Cohorts 1 and 2 until their expiry date and then the kits were switched to a new reagent and used for remaining subjects in Cohorts 2 and 3. Summary data for serum concentrations of IGF-1 were obtained using both methods (old and new reagent) and the mean change from Baseline at each time point is presented. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

Interventionng/mL (Mean)
Old Reagent: 6 hours post-doseOld Reagent: Week 5Old Reagent: Week 9Old Reagent: Week 13Old Reagent: Week 17Old Reagent: Week 21Old Reagent: Week 25 (EOS/EW)
180 mg Lanreotide PRF-1.82.472.353.448.372.486.3

[back to top]

Overall Summary of Number of Subjects With AEs.

AEs reported by the investigators using the National Cancer Institute-Common Toxicity Criteria (NCI CTCAE) classification (Version 4.03) and incidence of all reported treatment emergent AEs (TEAEs) and serious AEs (SAEs) are presented by dose cohort. AEs were assigned to a NCI CTCAE Grade from 1 through 5 as follows: Grade 1: Mild; Grade 2: Moderate; Grade 3: Severe or medically significant but not immediately life threatening or requiring hospitalisation; Grade 4: Life-threatening consequences; Grade 5: Death related to AE. TEAEs were defined as any AE that occurs during the active phase of the study (between the start of the 3 month treatment period and 3 months after the end of study treatment). The worst intensity of TEAES at each grade are reported for all and for related TEAES. In the event of multiple occurrences of the same AEs being reported by the same subject, the maximum intensity and the most serious causality were reported. (NCT02396953)
Timeframe: From Day -42 up to Week 25.

,,
Interventionparticipants (Number)
TEAEsRelated TEAEsTEAEs leading to study drug withdrawalSAEsSerious TEAEsSerious related TEAEsAEs leading to deathWorst TEAE: Grade 1Worst TEAE: Grade 2Worst TEAE: Grade 3Worst TEAE: Grade 4Worst TEAE: Grade 5Worst related TEAE: Grade 1Worst related TEAE: Grade 2Worst related TEAE: Grade 3Worst related TEAE: Grade 4Worst related TEAE: Grade 5
180 mg Lanreotide PRF62011001320002000
270 mg Lanreotide PRF73000003400012000
360 mg Lanreotide PRF74011005110031000

[back to top]

PK Analysis of Glycofurol Excipients: Tmax.

"Blood samples for determination of the excipients (N1-glycofurol and N2-glycofurol) serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8, 12 and 24 hours post-dose and on Days 3 and 5.~Median serum N1-glycofurol and N2-glycofurol Tmax values were determined using non-compartmental analysis." (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Day 5.

,
Interventionhours (Median)
N1-glycofurolN2-glycofurol
Glycofurol PK Set: 180 mg Lanreotide PRF2.002.00
Glycofurol PK Set: 270 mg Lanreotide PRF2.002.00

[back to top]

PD Analysis: Mean Change From Baseline in Insulin-like Growth Factor 1 (IGF-1).

Blood samples were collected for the determination of IGF-1 in serum at Baseline (pre-dose), 6 hours post-dose and at Weeks 5, 9 and 13. Samples were also collected during follow-up at Weeks 17, 21 and 25 (or EW). Serum concentrations of IGF-1 were calculated using the Immulite 2000 Platform for all subjects in the safety population. The production of the reagent kits was stopped by the vendor during the study. The old reagent kits were used for Cohorts 1 and 2 until their expiry date and then the kits were switched to a new reagent and used for remaining subjects in Cohorts 2 and 3. Summary data for serum concentrations of IGF-1 were obtained using both methods (old and new reagent) and the mean change from Baseline at each time point is presented. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

Interventionng/mL (Mean)
Old Reagent: 6 hours post-doseOld Reagent: Week 5New Reagent: 6 hours post-doseNew Reagent: Week 5New Reagent: Week 9New Reagent: Week 13New Reagent: Week 17New Reagent: Week 21New Reagent: Week 25 (EOS/EW)
360 mg Lanreotide PRF-46.5-32.0-11.61.325.247.140.956.964.1

[back to top]

PD Analysis: Mean Change From Baseline in Prolactin.

Blood samples were collected for the determination of prolactin in serum at Baseline (pre-dose) and at Weeks 2, 5, 13 and 25 (or EW). Summary data for serum concentration of prolactin were calculated and the mean change from Baseline at each time point is presented. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

,,
InterventionmU/L (Mean)
Week 2Week 5Week 13Week 25 (EOS/EW)
180 mg Lanreotide PRF-33.2-10.4-1.919.3
270 mg Lanreotide PRF37.616.333.958.3
360 mg Lanreotide PRF1.00.3-6.4-3.1

[back to top]

PD Analysis: Mean Change From Baseline in Thyroid Stimulating Hormone (TSH).

Blood samples were collected for the determination of TSH in serum at Baseline (pre-dose) and at Weeks 2, 5, 13 and 25 (or EW). Summary data for serum concentrations of TSH were calculated and the mean change from Baseline at each time point is presented. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

,,
InterventionmIU/L (Mean)
Week 2Week 5Week 13Week 25 (EOS/EW)
180 mg Lanreotide PRF-0.222-0.156-0.404-0.480
270 mg Lanreotide PRF0.1190.062-0.0600.073
360 mg Lanreotide PRF0.0470.0510.6010.235

[back to top]

PK Analysis of Glycofurol Excipients: AUC0-∞ and Area Under the Serum Concentration Time Curve From Time 0 to Last Quantifiable Timepoint (AUC0-t).

"Blood samples for determination of the excipients (N1-glycofurol and N2-glycofurol) serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8, 12 and 24 hours post-dose and on Days 3 and 5.~Mean serum N1-glycofurol and N2-glycofurol AUC0-∞ and AUC0-t values were determined using non-compartmental analysis. Only AUC0-∞ values fulfilling the accuracy determination rules were analysed." (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Day 5.

,
Interventionng*h/mL (Mean)
AUC0-∞: N1-glycofurolAUC0-∞: N2-glycofurolAUC0-t: N1-glycofurolAUC0-t: N2-glycofurol
Glycofurol PK Set: 180 mg Lanreotide PRF77910087831007
Glycofurol PK Set: 270 mg Lanreotide PRF1049135910821360

[back to top]

PK Analysis of Glycofurol Excipients: Cmax.

Blood samples for determination of the excipients (N1-glycofurol and N2-glycofurol) serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8, 12 and 24 hours post-dose and on Days 3 and 5. Mean serum N1-glycofurol and N2-glycofurol Cmax values were determined using non-compartmental analysis. (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Day 5.

,
Interventionng/mL (Mean)
N1-glycofurolN2-glycofurol
Glycofurol PK Set: 180 mg Lanreotide PRF75.479.7
Glycofurol PK Set: 270 mg Lanreotide PRF61.762.1

[back to top]

PK Analysis of Lanreotide: Apparent Terminal Elimination Half-life (t1/2).

"Blood samples for determination of lanreotide serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8,12 and 24 hours post-dose, on Days 3 and 5 and at Weeks 2, 3, 5, 9 and 13 after lanreotide PRF administration. Samples were also collected during follow-up at Weeks 17, 21 and 25 (or EW).~Mean serum lanreotide t1/2 values were determined using non-compartmental analysis. Only values fulfilling the determination rules for t1/2 were analysed." (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

Interventiondays (Mean)
Lanreotide PK Set: 180 mg Lanreotide PRF54.2
Lanreotide PK Set: 270 mg Lanreotide PRF61.7
Lanreotide PK Set: 360 mg Lanreotide PRF63.1

[back to top]

Determination of the Maximum Tolerated Dose (MTD) by Number of Subjects With DLTs.

The MTD was defined based on the DLTs observed in each cohort. A DLT was defined as an adverse event (AE) (excluding anorexia and fatigue) or an abnormal laboratory value occurring within the first week (up to Week 2) following lanreotide PRF administration and during the entire study duration, assessed as unrelated to acromegaly, intercurrent illness or concomitant medications and which met any of the pre-established toxicity criteria. If no DLTs were reported then no MTD could be defined. (NCT02396953)
Timeframe: From Day 1 up to Week 25.

Interventionparticipants (Number)
180 mg Lanreotide PRF0
270 mg Lanreotide PRF0
360 mg Lanreotide PRF0

[back to top]

PK Analysis of Lanreotide: Area Under the Serum Concentration-time Curve Extrapolated to Infinity (AUC0-∞).

"Blood samples for determination of lanreotide serum concentrations were collected at Baseline (pre-dose), at 1, 2, 4, 6, 8,12 and 24 hours post-dose, on Days 3 and 5 and at Weeks 2, 3, 5, 9 and 13 after lanreotide PRF administration. Samples were also collected during follow-up at Weeks 17, 21 and 25 (or EW).~Mean serum lanreotide AUC0-∞ values were determined using non-compartmental analysis. Only values fulfilling the accuracy determination rules for AUC0-∞ were analysed." (NCT02396953)
Timeframe: From Baseline (pre-dose) up to Week 25.

Interventionng*day/mL (Mean)
Lanreotide PK Set: 180 mg Lanreotide PRFNA
Lanreotide PK Set: 270 mg Lanreotide PRFNA
Lanreotide PK Set: 360 mg Lanreotide PRFNA

[back to top]

Percentage of Subjects With at Least 20% Reduction in Tumour Volume at EOST/EW Visit Compared to Baseline

"The percentage of subjects with at least a 20% reduction in the solid component of the tumour volume at the EOST/EW Visit compared to baseline is presented for the subgroup of subjects who had solid tumours at baseline.~The tumour volume was measured by Magnetic Resonance Imaging (MRI) at Screening and at the EOST/EW Visit, and then assessed by two independent blinded readers." (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

InterventionPercentage of Subjects (Number)
Lanreotide Autogel45.5
Lanreotide PR50.9

[back to top]

Percentage of Subjects With GH ≤2.5 Micrograms Per Litre (mcg/L) at the EOST/EW Visit

"The percentage of subjects with GH ≤2.5 mcg/L at the EOST/EW Visit is presented for subjects treated with lanreotide Autogel and lanreotide PR.~At baseline, all subjects had GH levels >2.5 mcg/L as per protocol entry criteria." (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

InterventionPercentage of Subjects (Number)
Lanreotide Autogel29.7
Lanreotide PR31.3

[back to top]

The Percentage of Subjects With GH ≤1 mcg/L at the EOST/EW Visit

"The percentage of subjects with GH ≤1 mcg/L at the EOST/EW Visit is presented for subjects treated with lanreotide Autogel and lanreotide PR.~At baseline, all subjects had GH levels >2.5 mcg/L as per protocol entry criteria." (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

InterventionPercentage of Subjects (Number)
Lanreotide Autogel10.9
Lanreotide PR9.4

[back to top]

Percentage of Subjects With Normal Age-adjusted IGF-1 Levels at the EOST/EW Visit

"The percentage of subjects with normal age-adjusted IGF-1 levels at the EOST/EW Visit is presented for subjects treated with Lanreotide Autogel and Lanreotide PR.~At baseline, all subjects had abnormal IGF-1 levels as per protocol entry criteria." (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

InterventionPercentage of Subjects (Number)
Lanreotide Autogel15.6
Lanreotide PR9.4

[back to top]

Standardised Mean Change From Baseline in Age-adjusted IGF-1 Levels at the EOST/EW Visit

"The standardised mean change from Baseline in age-adjusted log-transformed IGF-1 standard deviation score (SDS) at EOST/EW is presented for subjects treated with both lanreotide Autogel and lanreotide PR. Back-transformed results are presented in addition to the results without back-transformation.~For each subject the IGF-1 SDS value was calculated based on the z-score derivation: IGF-1 SDS = (IGF-1 - mean)/ standard deviation (SD), with mean and SD derived from the upper limit of normal (ULN) and lower limit of normal (LLN) margins for each age category. ULN = Mean + 2 SD; LLN = Mean - 2 SD.~The SDS indicates the number of standard deviations away from the mean. A SDS of 0 is equal to the mean with negative numbers indicating values lower than the mean and positive values higher. A negative change in the SDS indicates a decrease in the mean age-adjusted IGF-1 values." (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

,
InterventionSDS (Least Squares Mean)
Without back-transformationWith back transformation
Lanreotide Autogel-0.840.43
Lanreotide PR-0.520.59

[back to top]

Percentage of Subjects With Normal Age-adjusted IGF-1 Levels and Who Have GH Levels >1 mcg/L and ≤2.5 mcg/L at the EOST/EW Visit

"The percentage of subjects with normal age-adjusted IGF-1 levels and who have GH levels >1 mcg/L but ≤2.5 mcg/L at the EOST/EW Visit is presented for subjects treated with lanreotide Autogel and lanreotide PR. The calculation of percentages was based on the overall ITT population.~At baseline, all subjects had abnormal IGF-1 levels and GH levels >2.5 mcg/L as per protocol entry criteria." (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

InterventionPercentage of Subjects (Number)
Lanreotide Autogel7.8
Lanreotide PR3.1

[back to top]

Median Percentage Change From Baseline in Tumour Volume at the EOST/EW Visit

"The median percentage change in the solid component of the tumour volume from baseline to the EOST/EW Visit is presented.~The tumour volume was measured by MRI at Screening and at the EOST/EW Visit, and then assessed by two independent blinded readers." (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

InterventionPercentage change in tumour volume (Median)
Lanreotide Autogel-17.870
Lanreotide PR-20.120

[back to top]

Mean Change From Baseline in GH Values at the EOST/EW Visit

The mean change from baseline in GH values at the EOST/EW Visit is presented for subjects treated with lanreotide Autogel and lanreotide PR. (NCT02493517)
Timeframe: Baseline to EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

Interventionmcg/L (Mean)
Lanreotide Autogel-9.548
Lanreotide PR-13.182

[back to top]

Percentage of Subjects With at Least One Symptom of Acromegaly at Week 13 and at the EOST/EW Visit Compared to Baseline

The percentage of subjects with at least one symptom of acromegaly at Week 13 and at the EOST/EW Visit compared with baseline is presented for subjects treated with lanreotide Autogel and lanreotide PR. The symptoms of acromegaly monitored included: headache, excessive perspiration, fatigue, soft tissue swelling and arthralgia. (NCT02493517)
Timeframe: Baseline, Week 13 Visit and EOST/EW Visit (up to Week 33 for the lanreotide Autogel group and up to Week 32 for the lanreotide PR group).

,
InterventionPercentage of Subjects (Number)
Week 13 VisitEOST/EW Visit
Lanreotide Autogel70.375.0
Lanreotide PR59.459.4

[back to top]

Median PFS Time in the Double-Blind Phase, Assessed by Central Review

PFS was assessed by central review using RECIST v1.1 criteria every 12 weeks, defined as the time from randomisation to disease progression or death from any causes during the double-blind phase. The distribution of PFS times were estimated using the Kaplan-Meier product limit method. (NCT02683941)
Timeframe: Up to a maximum of 15 months

Interventionmonths (Median)
Double-Blind Phase: Lanreotide16.6
Double-Blind Phase: Placebo13.6

[back to top]

Median PFS Time in the Double-Blind Phase, Assessed by Local Review

PFS was assessed by local review using RECIST v1.1 criteria every 12 weeks, defined as the time from randomisation to disease progression or death from any causes during the double-blind phase. The distribution of PFS times were estimated using the Kaplan-Meier product limit method. (NCT02683941)
Timeframe: Up to a maximum of 15 months

Interventionmonths (Median)
Double-Blind Phase: Lanreotide14.1
Double-Blind Phase: Placebo13.6

[back to top]

Median Progression-Free Survival (PFS) Time in Subjects Randomised to Lanreotide in the Double-Blind Phase or Open-Label Treatment Phase, Assessed by Central Review

PFS for subjects randomised in the lanreotide group, assessed by central review using Response Evaluation Criteria In Solid Tumours Version 1.1 (RECIST v1.1) criteria every 12 weeks, defined as the time from randomisation to disease progression or death from any causes during either the double-blind phase, or the open-label treatment phase. The distribution of PFS times were estimated using the Kaplan-Meier product limit method. (NCT02683941)
Timeframe: Up to a maximum of 33 months

Interventionmonths (Median)
Overall Study: Lanreotide16.6

[back to top]

Percentage of Subjects Who Experienced QoL Deterioration

QoL deterioration was defined by a decrease from baseline in EORTC QLQ-C30 Global Health Status/QoL Score of at least 10 points. The EORTC QLQ-C30 (V3.0) consisted of 30 questions. The final 2 questions were related to global health status/QoL, with responses requested on a 7-point scale from 1 ('Very poor') to 7 ('Excellent'). The global health status/QoL scale ranges in score from 0 to 100. A high score for the global health status/QoL scale represents a high QoL, thus, an increase in score represents an increase in QoL. 95% Clopper-Pearson confidence intervals were estimated using the exact method for binomial distributions. Baseline was defined as the last non-missing measurement collected prior to the first dose of study treatment (lanreotide). (NCT02683941)
Timeframe: Baseline and post-treatment in the double-blind phase (a maximum of 15 months); Baseline and post-treatment in the open-label treatment phase (a maximum of 33 months)

Interventionpercentage of subjects (Number)
Double-Blind Phase: Lanreotide32.0
Double-Blind Phase: Placebo66.7
Open-Label Treatment Phase: All Subjects23.5

[back to top]

Percentage of Subjects With a Decrease of CgA ≥30% From Baseline at Week 8 in the Double-Blind Phase and Open-Label Treatment Phase

Measured in subjects with an elevated CgA at baseline (≥2 x ULN). Blood samples were collected to determine plasma CgA. Baseline was defined as the last non-missing measurement collected prior to the first dose of study treatment (lanreotide). (NCT02683941)
Timeframe: Baseline and Week 8 in the double-blind phase; Baseline and Week 8 in the open-label treatment phase

Interventionpercentage of subjects (Number)
Double-Blind Phase: Lanreotide63.3
Double-Blind Phase: Placebo7.7
Open-Label Treatment Phase: All Subjects73.7

[back to top]

Mean Change From Baseline in the Biomarker Chromogranin A (CgA) in the Double-Blind Phase and Open-Label Treatment Phase

Blood samples were collected to determine plasma CgA. Baseline was defined as the last non-missing measurement collected prior to the first dose of study treatment (lanreotide). The x of upper limit of normal (ULN) was calculated as raw value/ULN. (NCT02683941)
Timeframe: Baseline, Weeks 8, 12, 24, and 48, and post-treatment in the double-blind phase (a maximum of 15 months); Baseline, Weeks 12, 24, and 48, and post-treatment in the the open-label treatment phase (a maximum of 33 months)

Interventionx of ULN (Mean)
Week 12Week 24Week 48Post-treatment
Open-Label Treatment Phase: All Subjects-28.27-1.42-1.66-31.59

[back to top]

Mean Changes From Baseline in Urinary 5-hydroxyindoleacetic Acid (5-HIAA) Levels in the Double-Blind Phase and Open-Label Treatment Phase

Measured in subjects with an elevated 5-HIAA at baseline (≥2 x ULN). The assessment of urinary 5-HIAA required subjects to collect their urine for the 24 hour period prior to the study visit. Baseline was defined as the last non-missing measurement collected prior to the first dose of study treatment (lanreotide). The x of ULN was calculated as raw value/ULN. (NCT02683941)
Timeframe: Baseline, Weeks 8, 12, 24, and 48, and post-treatment in the double-blind phase (a maximum of 15 months); Baseline, Weeks 12, 24, and 48, and post-treatment in the the open-label treatment phase (a maximum of 33 months)

Interventionx of ULN (Mean)
Week 12Week 24Week 48Post-treatment
Open-Label Treatment Phase: All Subjects-0.47-4.84-2.470.60

[back to top]

Mean Changes From Baseline in Urinary 5-hydroxyindoleacetic Acid (5-HIAA) Levels in the Double-Blind Phase and Open-Label Treatment Phase

Measured in subjects with an elevated 5-HIAA at baseline (≥2 x ULN). The assessment of urinary 5-HIAA required subjects to collect their urine for the 24 hour period prior to the study visit. Baseline was defined as the last non-missing measurement collected prior to the first dose of study treatment (lanreotide). The x of ULN was calculated as raw value/ULN. (NCT02683941)
Timeframe: Baseline, Weeks 8, 12, 24, and 48, and post-treatment in the double-blind phase (a maximum of 15 months); Baseline, Weeks 12, 24, and 48, and post-treatment in the the open-label treatment phase (a maximum of 33 months)

,
Interventionx of ULN (Mean)
Week 8Week 12Week 24Week 48Post-treatment
Double-Blind Phase: Lanreotide-2.900.210.15-3.00-1.27
Double-Blind Phase: Placebo2.785.38-1.401.13-5.13

[back to top]

Mean Changes From Baseline in Quality of Life (QoL), as Assessed by the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (EORTC QLQ-C30) Global Health Status/QoL Score

The EORTC QLQ-C30 (V3.0) consisted of 30 questions. The final 2 questions were related to global health status/QoL, with responses requested on a 7-point scale from 1 ('Very poor') to 7 ('Excellent'). The global health status/QoL scale ranges in score from 0 to 100. A high score for the global health status/QoL scale represents a high QoL, thus, an increase in score represents an increase in QoL. 95% Clopper-Pearson confidence intervals were estimated using the exact method for binomial distributions. Baseline was defined as the last non-missing measurement collected prior to the first dose of study treatment (lanreotide). (NCT02683941)
Timeframe: Baseline and post-treatment in the double-blind phase (a maximum of 15 months); Baseline and post-treatment in the open-label treatment phase (a maximum of 33 months)

Interventionscore on a scale (Mean)
Double-Blind Phase: Lanreotide-2.7
Double-Blind Phase: Placebo-19.4
Open-Label Treatment Phase: All Subjects0.0

[back to top]

Time to Treatment Failure (TTF) in the Double-Blind Phase

TTF was defined as the time from randomisation to disease progression using RECIST v1.1, death, consent withdrawn, an adverse event, protocol deviations, lost to follow-up, the appearance of carcinoid syndrome or other hormone related syndrome necessitating the initiation of SSAs (rescue octreotide and/or long-acting release SSA), or initiation of anticancer treatment in the double-blind phase. The distribution of TTF times were estimated using the Kaplan-Meier product limit method. (NCT02683941)
Timeframe: Up to a maximum of 15 months

Interventionmonths (Median)
Double-Blind Phase: Lanreotide13.3
Double-Blind Phase: Placebo9.8

[back to top]

Mean Change From Baseline in the Biomarker Chromogranin A (CgA) in the Double-Blind Phase and Open-Label Treatment Phase

Blood samples were collected to determine plasma CgA. Baseline was defined as the last non-missing measurement collected prior to the first dose of study treatment (lanreotide). The x of upper limit of normal (ULN) was calculated as raw value/ULN. (NCT02683941)
Timeframe: Baseline, Weeks 8, 12, 24, and 48, and post-treatment in the double-blind phase (a maximum of 15 months); Baseline, Weeks 12, 24, and 48, and post-treatment in the the open-label treatment phase (a maximum of 33 months)

,
Interventionx of ULN (Mean)
Week 8Week 12Week 24Week 48Post-treatment
Double-Blind Phase: Lanreotide-213.63-214.49-3.42-4.0712.22
Double-Blind Phase: Placebo0.095.7360.411.24160.11

[back to top]

Objective Response Rate (ORR) in the Double-Blind Phase

ORR was assessed by central review and local review using RECIST v1.1 criteria every 12 weeks, defined as the percentage of subjects who achieved a best overall response of complete response or partial response in the double-blind phase. (NCT02683941)
Timeframe: Up to a maximum of 15 months

,
InterventionPercentage of subjects (Number)
Central reviewLocal review
Double-Blind Phase: Lanreotide14.006.00
Double-Blind Phase: Placebo0.004.00

[back to top]