Page last updated: 2024-12-11

lanreotide

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

Cross-References

ID SourceID
PubMed CID6918011
CHEMBL ID1201185
MeSH IDM0167728

Synonyms (47)

Synonym
lanreotide acetate
ipstyl
dc 13-116
lanreotide [inn:ban]
lanreotidum [inn-latin]
lanreotida [inn-spanish]
l-threoninamide, 3-(2-naphthalenyl)-d-alanyl-l-cysteinyl-l-tyrosyl-d-tryptophyl-l-lysyl-l-valyl-l-cysteinyl-, cyclic (2-7)-disulfide
autogel
bim 23014
nal-cyclo(cys-tyr-trp-lys-val-cys)-thr-nh2
somatulin
l-threoninamide, 3-(1-naphthalenyl)-d-alanyl-l-cysteinyl-l-tyrosyl-d-tryptophyl-l-lysyl-l-valyl-l-cysteinyl-, cyclic (2-7)-disulfide
CHEMBL1201185
(4r,7s,10s,13r,16s,19r)-10-(4-aminobutyl)-n-[(2s,3r)-1-amino-3-hydroxy-1-oxobutan-2-yl]-19-[[(2r)-2-amino-3-naphthalen-2-ylpropanoyl]amino]-16-[(4-hydroxyphenyl)methyl]-13-(1h-indol-3-ylmethyl)-6,9,12,15,18-pentaoxo-7-propan-2-yl-1,2-dithia-5,8,11,14,17-p
naphthalenyl-cyclo(cysteinyl-tyrosyl-tryptophyl-lysyl-valyl-cysteinyl)threoninamide
naphthyl-cyclo(cys-tyr-trp-lys-val-cys)thr-nh2
nal-cyclo(cys-tyr-trp-lys-val-cys)thr-nh2
118992-92-0
2-naphthylalanyl-cyclo(cysteinyl-tyrosyl-tryptophyl-lysyl-valyl-cysteinyl)-threoninamide
bdbm82470
3-(2-naphtyl)-d-ala-l-cys(1)-l-tyr-d-trp-l-lys-l-val-l-cys(1)-l-thr-nh2
somatulin-autogel
unii-0g3de8943y
lanreotide autogel
lanreotida
0g3de8943y ,
lanreotidum
AKOS030573447
l-threoninamide, 3-(2-naphthalenyl)-d-alanyl-l-cysteinyl-l-tyrosyl-d-tryptophyl-l-lysyl-l-valyl-l-cysteinyl-, cyclic (2->7)-disulfide
l-threoninamide, 3-(2-naphthalenyl)-d-alanyl-l-cysteinyl-l-tyrosyl-d-tryptophyl-l-lysyl-l-valyl-l-cysteinyl-, cyclic (2->7)-disulphide
3-(2-naphthyl)-d-alanyl-l-cysteinyl-l-tyrosyl-d-tryptophyl-l-lysyl-l-valyl-l-cysteinyl-l-threoninamide, cyclic (2->7)-disulfide
3-(2-naphthyl)-d-alanyl-l-cysteinyl-l-tyrosyl-d-tryptophyl-l-lysyl-l-valyl-l-cysteinyl-l-threoninamide, cyclic (2->7)-disulphide
DTXSID60897514 ,
(4r,7s,10s,13r,16s,19r)-13-((1h-indol-3-yl)methyl)-19-((r)-2-amino-3-(naphthalen-2-yl)propanamido)-n-((2s,3r)-1-amino-3-hydroxy-1-oxobutan-2-yl)-10-(4-aminobutyl)-16-(4-hydroxybenzyl)-7-isopropyl-6,9,12,15,18-pentaoxo-1,2-dithia-5,8,11,14,17-pentaazacyclo
Q1707877
l-threoninamide, 3-(2-naphthalenyl)-d-alanyl-l-cysteinyl-l-tyrosyl-d-tryptophyl-l-lysyl-l-valyl-l-cysteinyl-, cyclic (2-->7)-disulfide
108736-35-2 (free base)
C71317
CS-0103335
HY-P1959
dtxcid601326869
lanreotidum (inn-latin)
dc 13116
dermopeptin
h01cb03
lanreotide (mart.)
lanreotida (inn-spanish)
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Protein Targets (5)

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Somatostatin receptor type 1Homo sapiens (human)Ki0.50120.00030.41232.3920AID203390
Somatostatin receptor type 2Homo sapiens (human)Ki0.00050.00000.40914.7200AID203409
Somatostatin receptor type 4Homo sapiens (human)Ki1.00000.00060.27333.2000AID203570
Somatostatin receptor type 3Homo sapiens (human)Ki0.06310.00010.03200.1290AID203551
Somatostatin receptor type 5Homo sapiens (human)Ki0.00400.00010.44794.0700AID203709
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (23)

Processvia Protein(s)Taxonomy
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)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (6)

Processvia Protein(s)Taxonomy
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)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (6)

Processvia Protein(s)Taxonomy
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)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (42)

Assay IDTitleYearJournalArticle
AID1079938Chronic liver disease either proven histopathologically, or through a chonic elevation of serum amino-transferase activity after 6 months. Value is number of references indexed. [column 'CHRON' in source]
AID625280Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholecystitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID625283Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for elevated liver function tests2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID625285Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic necrosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID625287Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatomegaly2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1079939Cirrhosis, proven histopathologically. Value is number of references indexed. [column 'CIRRH' in source]
AID1079946Presence of at least one case with successful reintroduction. [column 'REINT' in source]
AID588213Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in non-rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1079944Benign tumor, proven histopathologically. Value is number of references indexed. [column 'T.BEN' in source]
AID625281Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholelithiasis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1079940Granulomatous liver disease, proven histopathologically. Value is number of references indexed. [column 'GRAN' in source]
AID625289Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver disease2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID625279Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for bilirubinemia2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID625284Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic failure2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1079947Comments (NB not yet translated). [column 'COMMENTAIRES' in source]
AID1079937Severe hepatitis, defined as possibly life-threatening liver failure or through clinical observations. Value is number of references indexed. [column 'MASS' in source]
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.
AID1079932Highest frequency of moderate liver toxicity observed during clinical trials, expressed as a percentage. [column '% BIOL' in source]
AID625291Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver function tests abnormal2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
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.
AID625290Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver fatty2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1079945Animal toxicity known. [column 'TOXIC' in source]
AID588211Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in humans2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1079941Liver damage due to vascular disease: peliosis hepatitis, hepatic veno-occlusive disease, Budd-Chiari syndrome. Value is number of references indexed. [column 'VASC' in source]
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.
AID1079943Malignant tumor, proven histopathologically. Value is number of references indexed. [column 'T.MAL' in source]
AID1079933Acute liver toxicity defined via clinical observations and clear clinical-chemistry results: serum ALT or AST activity > 6 N or serum alkaline phosphatases activity > 1.7 N. This category includes cytolytic, choleostatic and mixed liver toxicity. Value is
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.
AID1079936Choleostatic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is < 2 (see ACUTE). Value is number of references indexed. [column 'CHOLE' in source]
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.
AID1079949Proposed mechanism(s) of liver damage. [column 'MEC' in source]
AID625282Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cirrhosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID588212Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1079935Cytolytic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is > 5 (see ACUTE). Value is number of references indexed. [column 'CYTOL' in source]
AID1079931Moderate liver toxicity, defined via clinical-chemistry results: ALT or AST serum activity 6 times the normal upper limit (N) or alkaline phosphatase serum activity of 1.7 N. Value is number of references indexed. [column 'BIOL' in source]
AID625292Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) combined score2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID195175In vitro GH release inhibiting potency on anterior pitutaries from adult male rats, relative to somatostatin1995Journal of medicinal chemistry, May-26, Volume: 38, Issue:11
Three-dimensional quantitative structure-activity relationships of somatostatin analogues. 1. Comparative molecular field analysis of growth hormone release-inhibiting potencies.
AID1079934Highest frequency of acute liver toxicity observed during clinical trials, expressed as a percentage. [column '% AIGUE' in source]
AID1079948Times to onset, minimal and maximal, observed in the indexed observations. [column 'DELAI' in source]
AID625288Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for jaundice2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID625286Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1079942Steatosis, proven histopathologically. Value is number of references indexed. [column 'STEAT' in source]
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (4)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's1 (25.00)18.2507
2000's1 (25.00)29.6817
2010's2 (50.00)24.3611
2020's0 (0.00)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 75.54

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be very strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index75.54 (24.57)
Research Supply Index1.79 (2.92)
Research Growth Index4.55 (4.65)
Search Engine Demand Index124.38 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (75.54)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials0 (0.00%)5.53%
Reviews0 (0.00%)6.00%
Case Studies0 (0.00%)4.05%
Observational0 (0.00%)0.25%
Other5 (100.00%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (81)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Long-Acting Lanreotide as a Volume Reducing Treatment of Polycystic Livers [NCT00565097]Phase 2/Phase 338 participants (Anticipated)Interventional2007-10-31Completed
Continuing Somatostatin Analogues Upon Progression in Neuroendocrine Tumour pAtients - The SAUNA Trial [NCT05701241]Phase 4270 participants (Anticipated)Interventional2023-06-28Recruiting
The Longitudinal Approach to Acromegaly: A Pattern of Treatment and Comparative Effectiveness Research [NCT03158090]1,965 participants (Anticipated)Observational2017-12-15Recruiting
Traitement Des Carcinomes à Cellules de Merkel inopérables et/ou métastatiques Par Analogue de la Somatostatine - Etude Nationale Multicentrique Mono-bras de Phase II. [NCT02351128]Phase 235 participants (Actual)Interventional2015-04-30Completed
A Prospective, Randomized Trial of Preoperative Lanreotide Treatment in Acromegalic Patients With Macroadenomas [NCT00993356]Phase 296 participants (Anticipated)Interventional2004-01-31Recruiting
Comparison of Combination Low-Dose SRL + Daily Pegvisomant Therapy, Low-Dose SRL + Weekly Pegvisomant Therapy, and High-Dose SRL + Weekly Pegvisomant Therapy [NCT01538966]76 participants (Actual)Interventional2012-03-29Terminated(stopped due to Review of the 12-month data showed patients who deviated from protocol with full IRB monitoring and reporting could not be optimally analyzed. Reopening the study for new enrollment is not feasible and the study was terminated with IRB approval.)
An Open-label, Phase II Clinical Study to Evaluate the Efficacy and Safety of Lanreotide Autogel 90mg Every 4 Weeks in the Treatment of Symptomatic Polycystic Liver Disease, Including a Dose Escalation at Month 6 to Lanreotide Autogel 120mg for Non Respon [NCT01315795]Phase 2/Phase 359 participants (Actual)Interventional2011-03-31Completed
The Effect of Lanreotide on Volume of Polycystic Liver and Kidney in Autosomal Dominant Polycystic Kidney Disease [NCT01354405]43 participants (Actual)Observational2011-05-31Completed
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
Influence of Somatuline Autogel 120mg on Post-operative Drainage After Total Mesorectum Excision for Rectumcarcinoma [NCT01372007]Phase 324 participants (Actual)Interventional2011-07-31Completed
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.)
A Multicenter, Open, Prospective, Observational Study to Investigate the Effect of Lanreotide Autogel 120 mg on Control of GH and IGF-I Excess and Tumor Shrinkage in Newly Diagnosed Patients With Acromegaly [NCT00627796]Phase 427 participants (Actual)Interventional2003-01-31Completed
Evaluation of Lanreotide Efficacity in High Output Stoma: a Multicentric Randomized Study [NCT02354768]Phase 319 participants (Actual)Interventional2015-10-22Terminated
A Phase II Trial of Lanreotide for the Prevention of Postoperative Pancreatic Fistula [NCT03174353]Phase 2114 participants (Actual)Interventional2018-02-20Completed
Open-Label Extension of the LOCKCYST Trial, LOCKCYST: Long Acting Lnareotide as as Volume Reducing Treatment of Polycystic Livers [NCT00771888]Phase 2/Phase 354 participants (Anticipated)Interventional2008-04-30Active, not recruiting
Phase II, Multicentre, Open Label Study to Evaluate the Efficacy of the Combination of Lanreotide Autogel 120mg and Temozolomide in Patients With Progressive Gastro-entero-pancreatic Neuroendocrine Tumours (GEP-NET) G1/G2 - A Pilot-Study [NCT02231762]Phase 257 participants (Actual)Interventional2014-10-31Completed
Phase III, Randomised, Double-blind, Stratified Comparative, Placebo Controlled, Parallel Group, Multi-centre Study to Assess the Effect of Deep Subcutaneous Injections of Lanreotide Autogel 120mg Administered Every 28 Days on Tumour Progression Free Surv [NCT00353496]Phase 3264 participants (Actual)Interventional2006-06-30Completed
Resolving Bile Reflux by Lanreotide in Patients With Roux-en-Y Gastrojejunostomy [NCT02054637]Phase 210 participants (Anticipated)Interventional2014-04-30Not yet recruiting
Exploratory Phase II Study of LAnreotide in Metastatic Pheochromocytoma/PARAganglioma (LAMPARA) [NCT03946527]Phase 240 participants (Anticipated)Interventional2019-06-17Recruiting
Randomised, Phase III Multicenter, Open Study of Lanreotide in Non Metastatic Castration-resistant Prostate Cancer Patients Presenting Elevated Chromogranin A Levels [NCT01313273]Phase 33 participants (Actual)Interventional2011-06-30Terminated(stopped due to Poor enrolment)
A Phase III Multicentre Randomised Double-blind Comparative Study of Efficacy and Safety of Lanreotide 30 mg Versus Placebo for Treatment of Patients With Digestive Fistulae [NCT00729313]Phase 3111 participants (Actual)Interventional2000-04-30Completed
A Phase IV, International, Open-label, Randomised, Cross-over Study to Assess Patient Preference and Health Economy in Patients With Neuroendocrine Tumours, Treated With Lanreotide Autogel Given as Self Administration. [NCT00681187]Phase 426 participants (Actual)Interventional2008-06-30Completed
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
Lanreotide In Polycystic Kidney Disease Study [NCT02127437]Phase 3159 participants (Actual)Interventional2014-09-19Completed
Prospective Study to Assess the Efficacy and Safety of Lanreotide 120 mg as a Palliative Treatment of Clinical Symptoms Associated With Malignant Intestinal Obstruction in Inoperable Patients [NCT01076803]Phase 240 participants (Actual)Interventional2009-10-31Completed
Phase 1b/3 Global, Randomized, Controlled, Open-label Trial Comparing Treatment With RYZ101 to Standard of Care Therapy in Subjects With Inoperable, Advanced, SSTR+, Well-differentiated GEP-NETs That Have Progressed Following Prior 177Lu-SSA Therapy [NCT05477576]Phase 3288 participants (Anticipated)Interventional2022-03-24Recruiting
A Phase II-III, Multicentre, Prospective, Exploratory, Open Label Study to Assess the Efficacy and Safety of Lanreotide Autogel 120mg in the Symptomatic Treatment of Patients With Refractory Diarrhea [NCT00891371]Phase 2/Phase 342 participants (Actual)Interventional2009-07-31Completed
A Phase 1b Study in Patients With Acromegaly or Functioning Gastroenteropancreatic Neuroendocrine Tumors (GEP-NETs) to Characterize the Pharmacokinetics, Pharmacodynamics, Safety and Tolerability of Debio 4126, a 12-week Prolonged-release Octreotide Formu [NCT05364944]Phase 130 participants (Anticipated)Interventional2022-05-18Active, not recruiting
A Phase II Study of Lanreotide in Patients With Metastatic Gastrointestinal Neuroendocrine Tumors Undergoing Liver-directed Radioembolization With Yttrium-90 Microspheres (SIR-Spheres®) [NCT02859064]Phase 26 participants (Actual)Interventional2017-07-28Terminated(stopped due to Closed due to slow accrual)
Impact of Somatostatin Analogs vs. Surgery on Glucose Metabolism in Acromegaly: Results of a 5 Years Observational, Open, Prospective Study [NCT00703079]100 participants (Actual)Observational1997-01-31Completed
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
Randomized Phase III Clinical Trial of Lanreotide Combined With Telotristat Ethyl or Placebo for the First-line Treatment in Patients With Advanced Well Differentiated Small Intestinal Neuroendocrine Tumours (siNET) With Highly-functioning Carcinoid Syndr [NCT04065165]Phase 30 participants (Actual)Interventional2020-04-30Withdrawn(stopped due to Company decision)
A Multi Center Open Label Study to Assess the Ability of Subjects With Acromegaly or Their Partners to Administer Somatuline Autogel [NCT00447499]Phase 359 participants (Actual)Interventional2007-04-30Completed
Phase IIIb, Multicentre, Open-label, Single-arm, Study to Assess the Efficacy and Safety of Lanreotide Autogel 120 mg Administered Every 28 Days as Primary Medical Treatment in Acromegalic Patients With Macroadenoma [NCT00690898]Phase 3108 participants (Actual)Interventional2008-05-31Completed
A Phase I/II Study Using Cabozantinib and Lanreotide as Treatment for Advanced Gastroenteropancreatic Neuroendocrine Tumors That Failed Molecular Targeted Therapies or Chemotherapy (SCALET) [NCT05048901]Phase 1/Phase 249 participants (Anticipated)Interventional2021-09-17Recruiting
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
Phase II, Multicentre, Randomized, Double-blind Study in Acromegalic Patients Evaluating the Efficacy and Safety of a Single Deep Subcutaneous Administration of Lanreotide Autogel (60, 90 or 120mg) Versus Placebo Followed by a Single Blind Fixed Dose Phas [NCT00234572]Phase 2/Phase 3108 participants (Actual)Interventional2000-05-31Completed
Treatment With Lanreotide Autogel (Somatostatin Analogue) in Patients With Congenital Hyperinsulinism of Infancy Already Treated With Somatostatin Analog by Pump [NCT01070758]Phase 410 participants (Anticipated)Interventional2010-02-28Recruiting
Phase III, Multicentre, Open Study to Assess the Efficacy and Safety Profiles of the Co-administration of Lanreotide Autogel 120 mg (Administered Via Deep Subcutaneous Injections Every 28 Days) and Pegvisomant 40 to 120 mg Per Week (Administered Via Subcu [NCT00383708]Phase 3125 participants (Actual)Interventional2006-10-31Completed
A Phase IV, Open-label, Single Group Study to Evaluate the Dosing, Efficacy and Safety of Lanreotide Autogel® in Patients With Acromegaly Previously Treated With Octreotide LAR [NCT00216398]Phase 48 participants (Actual)Interventional2004-06-30Completed
Non-interventional, Observational, Long Term Study to Evaluate Common Therapeutic Algorithms and Possible Predictive Parameters for Somatuline Autogel (ATG) Treatment in Patients With Acromegaly. [NCT02020499]5 participants (Actual)Observational2014-08-31Terminated(stopped due to Poor enrolment)
A Phase III, Multicenter, Randomized, Parallel-group Study to Assess the Efficacy and Safety of Double-blind Pasireotide LAR 40 mg and Pasireotide LAR 60 mg Versus Open-label Octreotide LAR or Lanreotide ATG in Patients With Inadequately Controlled Acrome [NCT01137682]Phase 3198 participants (Actual)Interventional2010-07-19Completed
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
Safety of Lanreotide 120 mg ATG in Combination With Metformin in Patients With Progressive Advanced Well-differentiated Gastro-intestinal (GI) or Lung Carcinoids: A Pilot, One-arm, Open-label, Prospective Study: the MetNET-2 Trial [NCT02823691]Early Phase 120 participants (Actual)Interventional2016-04-30Active, not recruiting
Treatment of the Dumping Syndrome With Lanreotide Autogel® [NCT00543179]Phase 40 participants Interventional2007-10-31Recruiting
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
A Pilot Study to Evaluate Safety and Effectiveness of Lanreotide in the Treatment of Patients With Small Bowel Motility Disorders (SBMD): a Prospective, Non-randomized, Single-center Study of 20 Participants [NCT03012594]Phase 212 participants (Actual)Interventional2017-05-11Completed
A Phase IV, Multicentre, Open Label, Controlled Study to Assess the Ability of Patients With Acromegaly, or Their Partners, to Administer Somatuline Autogel. [NCT00149188]Phase 430 participants (Actual)Interventional2004-02-29Completed
A EUROPEAN, MULTICENTRE, PHASE II/III RANDOMISED DOUBLE-BLIND, PLACEBO CONTROLLED STUDY EVALUATING LANREOTIDE AS MAINTENANCE THERAPY IN PATIENTS WITH NON-RESECTABLE DUODENO-PANCREATIC NEUROENDOCRINE TUMOURS AFTER FIRST-LINE TREATMENT [NCT02288377]Phase 2/Phase 353 participants (Actual)Interventional2015-01-31Terminated(stopped due to Because of slow recruitment)
Prospective Observational Study to Assess the Effectiveness and Safety of Lanreotide Autogel® in Patients With Locally Advanced or Metastatic Gastroenteropancreatic Neuroendocrine Tumors (GEP-NETs) in Asia Region [NCT04696042]95 participants (Anticipated)Observational [Patient Registry]2021-01-01Recruiting
Phase III, Multicentre, Open Clinical Study on the Efficacy and Tolerability of a New Slow-release Formulation of Lanreotide (Autogel 120 mg) in Patients With Active Acromegaly [NCT00499993]Phase 363 participants (Actual)Interventional2001-01-31Completed
Circulating Tumor Cells and Tumor DNA in HCC and NET - Patient-specific Biomarkers for Clinical Decision Support and Tailored Relapse Diagnostics [NCT02973204]167 participants (Actual)Observational [Patient Registry]2016-11-30Completed
Phase III, Multicentre, Randomised, Double-blind, Comparative Study to Assess the Efficacy and Safety of Lanreotide 30 mg Versus Placebo as a Palliative Treatment of Clinical Symptoms Associated With Intestinal Obstruction Due to Peritoneal Carcinomatosis [NCT00216372]Phase 380 participants (Actual)Interventional2003-09-30Completed
A Phase III, Multicentre, Open Label, Comparative, Dose-interval Titration Study Evaluating the Efficacy and Safety of Six Repeated Deep Subcutaneous Administrations of Lanreotide Autogel 120mg, in Acromegalic Patients Previously Treated With Octreotide L [NCT00444873]Phase 338 participants (Actual)Interventional2005-01-31Completed
Treatment of Advanced Hepatocellular Carcinoma With Depot Somatostatin Analogues: a Pilot Prospective Study Based on Somatostatin Receptors Tumors Expression [NCT00495846]Phase 2/Phase 325 participants (Actual)Interventional2007-04-30Completed
A Pilot Study to Evaluate the Correlation Between Plasma Lanreotide Levels and GH/IGF-1 Dynamics and Clinical Improvement in Patients With Acromegaly Treated With Lanreotide Autogel (Somatuline ATG) [NCT00517491]Phase 20 participants (Actual)Interventional2008-05-31Withdrawn(stopped due to Administrative reasons)
Phase III, Multicentre, Open Study to Assess the Efficacy and Safety of Lanreotide Autogel (60, 90 or 120 mg) in Acromegalic Patients Previously Treated or Not by Somatostatin Analogues. [NCT00210457]Phase 363 participants (Actual)Interventional2000-09-30Completed
Comparable Effects of Lanreotide Autogel and Octreotide LAR on GH, IGF-I Levels and Patient Satisfaction - A Twelve Month Randomized Cross-Over Study in Patients With Acromegaly [NCT00145405]Phase 412 participants Interventional2002-09-30Completed
A Phase II Trial Aiming to Assess the Safety and Activity of the Combination of Cabozantinib Plus Lanreotide in Gastroenteropancreatic (GEP) and Thoracic Neuroendocrine Tumor (NET): The LOLA Trial [NCT04427787]Phase 269 participants (Anticipated)Interventional2020-06-20Recruiting
PILOT STUDY OF LANREOTIDE IN METASTATIC OR RECURRENT GRADE I-II HINDGUT NET [NCT03083210]Phase 428 participants (Anticipated)Interventional2017-07-05Recruiting
A Phase II, Single Centre, Randomized, Double-blind, Parallel and Placebo Controlled, Pilot Study to Evaluate the Efficacy and Safety of Somatuline Autogel 60mg in Patients With Active Thyroid-associated Ophthalmopathy of Moderate Intensity. [NCT00288522]Phase 25 participants (Actual)Interventional2006-01-31Terminated
Phase II, Open, Single Group, Multicentre Study to Evaluate the Efficacy and Safety of Lanreotide Autogel Administered Every 4 Weeks by Deep Subcutaneous Injection in the Tumour's Growth Stabilization of Patients With Progressive Neuroendocrine Tumours Wh [NCT00326469]Phase 230 participants (Actual)Interventional2006-05-31Completed
A Randomized, Multi-center, Open-label, Active-controlled Phase 3 Trial to Assess the Efficacy and Safety of Octreotide Subcutaneous Depot (CAM2029) Versus Octreotide LAR or Lanreotide ATG in Patients With GEP-NET [NCT05050942]Phase 3300 participants (Anticipated)Interventional2021-10-22Recruiting
A RANDOMIZED PHASE II DOUBLE-BLIND TRIAL OF SUNITINIB VERSUS PLACEBO IN COMBINATION WITH LANREOTIDE IN PATIENTS WITH PROGRESSIVE ADVANCED/METASTATIC MIDGUT CARCINOID TUMORS [NCT01731925]Phase 244 participants (Actual)Interventional2013-01-07Active, not recruiting
A Prospective, International, Multi-centric, Open-label Study to Assess the Efficacy of an Extended Injection Interval Schedule of Lanreotide Autogel 120 mg in Acromegalic Subjects Who Are Biochemically Controlled on the Long Term Treatment With Octreotid [NCT00701363]Phase 4124 participants (Actual)Interventional2008-10-31Completed
Non-interventional, Observational Study to Evaluate Common Therapeutic Algorithms and Possible Predictive Parameters for Somatuline Autogel® (ATG) Treatment in Subjects With Either Acromegaly or Neuroendocrine Tumours (NET) [NCT01840449]156 participants (Actual)Observational2012-06-30Completed
A Phase II, Multicentre, Randomized Controlled Study Evaluating The Quality Of Life In Patients With Inoperable Malignant Bowel Obstruction Treated With Lanreotide Autogel 120 mg in Combination With Standard Care vs. Standard Care Alone (QOL IN IMBO STUDY [NCT02365584]Phase 243 participants (Actual)Interventional2015-01-31Terminated(stopped due to The study was terminated early due to insufficient recruitment.)
An International, Multicentric, Prospective, Open Label Study to Assess the Efficacy and Safety of Lanreotide Autogel 120 MG Associated to Standard of Care in the Treatment of Clinical Symptoms Associated With Inoperable Malignant Intestinal Obstruction [NCT02275338]Phase 252 participants (Actual)Interventional2014-11-19Completed
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
A Phase 3, Single-arm, Open-label, Multicentre Study to Assess the Efficacy and Safety of Deep Subcutaneous Injections of Lanreotide Autogel® 120 mg Administered Every 28 Days in Chinese Participants With Unresectable, Locally Advanced or Metastatic Grade [NCT04852679]Phase 343 participants (Actual)Interventional2021-05-24Completed
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
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 Ib/II Study of Pembrolizumab With Lanreotide Depot for Gastroenteropancreatic Neuroendocrine Tumors [NCT03043664]Phase 1/Phase 222 participants (Actual)Interventional2017-07-01Completed
A Pilot Study To Evaluate Patient Experience With the Somatostatin Analogs Octreotide Long Acting Release and Lanreotide During the Treatment of Advanced, Nonfunctional, Well Differentiated Neuroendocrine Tumors [NCT03289741]Phase 453 participants (Actual)Interventional2017-09-19Completed
Efficacy and Safety of Lanreotide Autogel® 120 mg Administered Every 14 Days in Well Differentiated, Metastatic or Locally Advanced, Unresectable Pancreatic or Midgut Neuroendocrine Tumours Having Progressed Radiologically While Previously Treated With La [NCT02651987]Phase 299 participants (Actual)Interventional2015-12-15Completed
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
A Pilot Study of Pre- and Post-operative Somatuline Depot Therapy in Acromegalic Patients Treated by Endonasal Endoscopic Surgery: Impact on Early Remission Rates and Perioperative Morbidity [NCT01861717]Phase 44 participants (Actual)Interventional2013-05-31Terminated(stopped due to Didn't enroll enough subjects)
SOMATULINE Autogel 90 mg IN DUMPING SYNDROME [NCT01923649]Phase 224 participants (Actual)Interventional2008-04-30Completed
Multicenter Evaluation of the Effect of Upfront Radiosurgery on Residual Growth Hormone-secreting Pituitary Adenoma From an Endocrinological Point of View (MERGE Study): a Randomized, Phase 3 Trial [NCT03439709]Phase 390 participants (Anticipated)Interventional2018-03-01Not yet recruiting
A Phase IV, Multicentre, Open Label, Single Group Exploratory Study to Assess the Clinical Value of Enumeration of Circulating Tumour Cells (CTCs) to Predict Clinical Symptomatic Response and Progression Free Survival in Patients Receiving Deep Subcutaneo [NCT02075606]Phase 450 participants (Actual)Interventional2014-05-31Completed
Efficacy and Safety of Lanreotide ATG 120 mg in Combination With Temozolomide in Subjects With Progressive Well Differentiated Thoracic Neuroendocrine Tumors. A Phase II, Multicentre, Single Arm, Open-label Trial. [NCT02698410]Phase 240 participants (Actual)Interventional2016-07-31Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00353496 (6) [back to overview]Change in the Global Health Status Quality of Life Assessment
NCT00353496 (6) [back to overview]Percentage of Patients With a Greater Than or Equal to 50% Decrease in Plasma Chromogranin A (CgA) Levels
NCT00353496 (6) [back to overview]Percentage of Patients Still Alive Based on Available Overall Survival Data
NCT00353496 (6) [back to overview]Pharmacokinetic Profile of Lanreotide
NCT00353496 (6) [back to overview]Percentage of Patients Alive & Without Disease Progression
NCT00353496 (6) [back to overview]Progression-Free Survival (PFS)
NCT00383708 (30) [back to overview]Percentage of Subjects With Serum GH Levels Lesser or Equal to 2.5 ng/mL During the Study
NCT00383708 (30) [back to overview]Percentage of Subjects With Acromegaly With a Normalised (Age and Sex Adjusted) IGF-1 Level at the End of the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Acid Labile Subunit Levels From Baseline During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Mean PR Interval, QRS Interval, QT Interval, RR Interval and QTcF During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Mean Weight From Baseline During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Mean Supine Systolic and Diastolic Blood Pressure (BP) During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Mean Supine Heart Rate During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Mean Pituitary Tumour Size During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Mean Glycosylated Haemoglobin (HbA1C) During the Co-administration Period; Assessed in Non Diabetic and Diabetic Subjects
NCT00383708 (30) [back to overview]Change From Baseline in Prolactin Levels During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Mean Fasting Insulin Concentration During the Co-administration Period; Assessed in Non Diabetic Subjects
NCT00383708 (30) [back to overview]Change From Baseline in Mean Fasting Insulin / Glucose Ratio During the Co-administration Period; Assessed in Non Diabetic Subjects
NCT00383708 (30) [back to overview]Change From Baseline in Prothrombin Time (Expressed as a Percentage of Normal) During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Serum GH Binding Protein Levels During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Serum GH Levels During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Serum IGF-1 Levels (Expressed as Z-scores) During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Total Bilirubin During the Co-administration Period
NCT00383708 (30) [back to overview]Correlation Between the Changes in ACROQoL Assessments With the Corresponding Changes in Z-score of IGF-1 Levels Over the Run-in Period and Co-administration Period
NCT00383708 (30) [back to overview]Number of Subjects With Putative Antibodies to Lanreotide and to Pegvisomant During the Co-administration Period
NCT00383708 (30) [back to overview]Number of Subjects With Shift in Presence/Absence of Lithiasis and/or Sludge During Co-administration Period
NCT00383708 (30) [back to overview]Percentage of Subjects With a Normalised (Age and Sex Adjusted) IGF-1 Level at Any Time During the Co-administration Period
NCT00383708 (30) [back to overview]Percentage of Subjects With Acromegaly With a Normalised (Age and Sex Adjusted) IGF-1 Level at the End of the Co-administration Period; Summarised by Diabetic Status at Baseline
NCT00383708 (30) [back to overview]Percentage of Subjects With Acromegaly With a Normalised (Age and Sex Adjusted) IGF-1 Level at the End of the Co-administration Period; Summarised by Previous Treatment and by Final Dose of Pegvisomant
NCT00383708 (30) [back to overview]Percentage of Subjects With Normalised (Age and Sex Adjusted) IGF-1 at Each Assessment
NCT00383708 (30) [back to overview]Change From Baseline in Mean Fasting Glucose Concentration During the Co-administration Period; Assessed in Non Diabetic Subjects
NCT00383708 (30) [back to overview]Change From Baseline in Mean Blood Glucose Maximum Concentration (Cmax) From Oral Glucose Tolerance Test (OGTT) During the Co-administration Period; Assessed in Non Diabetic Subjects
NCT00383708 (30) [back to overview]Change From Baseline in Liver Function Test Parameters During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Electrocardiogram (ECG) Mean Heart Rate During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Acromegaly Symptoms During the Co-administration Period
NCT00383708 (30) [back to overview]Change From Baseline in Acromegaly Quality of Life (ACROQoL) Assessments During the Co-administration Period
NCT00447499 (7) [back to overview]Total Symptom Questionnaire Score at Week 24/Termination
NCT00447499 (7) [back to overview]Change of GH Concentration Levels From Basaeline to Week 24 in Switch Patients
NCT00447499 (7) [back to overview]Percentage of Switch Subjects That Have Glucose Suppressed GH Levels ≤ 2.5 ng/ml at the End of the Study, Week 24/Termination.
NCT00447499 (7) [back to overview]Percentage of Switch Subjects Who Find Self-administration of Somatuline Autogel Convenient as Assessed by the Subject Convenience Questionnaire Score.
NCT00447499 (7) [back to overview]The Percentage of Subjects or Their Partners That Are Competent to Self-administer Somatuline Autogel at the End of the Study, (Week 24/Early Termination), as Assessed by the Competence Questionnaire Score.
NCT00447499 (7) [back to overview]Total Health Care Professional Convenience Questionnaire Score at Week 24/Termination
NCT00447499 (7) [back to overview]Percentage of Switch Subjects That Have IGF-1 Levels Within the Normal Range for Age and Gender at the End of the Study
NCT00681187 (10) [back to overview]5-hydroxyindoleacetic Acid (5-HIAA) Levels
NCT00681187 (10) [back to overview]Chromogranin A Levels
NCT00681187 (10) [back to overview]Days Sick Leave
NCT00681187 (10) [back to overview]Healthcare Professionals With Positive Response to Specified Questions on Self or Partner Administration Method
NCT00681187 (10) [back to overview]Number of Patients Stating at Least One Injection Negatively Interfered With Psychological Wellbeing
NCT00681187 (10) [back to overview]Subject Preference for Self or Partner Administration
NCT00681187 (10) [back to overview]Total Number of Visits to HCP Due to Carcinoid Symptoms
NCT00681187 (10) [back to overview]Perceived Symptom Control Evaluation in Respect to Episodes of Diarrhoea
NCT00681187 (10) [back to overview]Perceived Symptom Control Evaluation in Respect to Episodes of Flushing
NCT00681187 (10) [back to overview]Number of Patients Stating at Least One Injection Interfered With Daily Activities
NCT00690898 (11) [back to overview]Number of Patients With at Least a 20% Reduction in Tumour Volume From Baseline Volume (Visit 1) to Week 12 (Visit 3) and Week 24 (Visit 4).
NCT00690898 (11) [back to overview]Percent Variation From Baseline to Visit 3, 4 and 5 (Week 12, 24, and 48) of IGF-1 Levels
NCT00690898 (11) [back to overview]Percent Variation From Baseline to Visit 3, 4 and 5 (Week 12, 24, and 48) of Serum GH Levels.
NCT00690898 (11) [back to overview]Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Arthralgia) From Baseline
NCT00690898 (11) [back to overview]Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Fatigue) From Baseline
NCT00690898 (11) [back to overview]Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Headache) From Baseline
NCT00690898 (11) [back to overview]Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Soft Tissue Swelling) From Baseline
NCT00690898 (11) [back to overview]Percentage of Patients With Relevant Reduction in Pituitary Tumour Volume (as Measured by MRI) From Baseline Volume (Visit 1) to Week 48 (After 12 Injections at Visit 5)
NCT00690898 (11) [back to overview]Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Excessive Perspiration) From Baseline
NCT00690898 (11) [back to overview]Change From Baseline to Visit 3, 4 and 5 (Week 12, 24, and 48) of Prolactin Levels
NCT00690898 (11) [back to overview]Changes in the Global Acromegaly Quality of Life Assessment (AcroQoL) From Baseline
NCT00701363 (15) [back to overview]Mean Baseline IGF-1 Levels (Expressed as % of ULN) in All Groups (A, B and C) Versus Mean Baseline IGF-1 Levels (Expressed as % of ULN) in Subjects With Uncontrolled IGF-1 Levels at Week 24
NCT00701363 (15) [back to overview]Serum Growth Hormone (GH) Levels
NCT00701363 (15) [back to overview]Percentage of Subjects Who Extend Their Injection Interval to Eight Weeks During Phase 2 of the Study, Whilst Maintaining Normalised IGF-1 Levels
NCT00701363 (15) [back to overview]Percentage of Subjects Having Maintained Their Injection Interval Schedule of Six Weeks or Increased Their Injection Interval to Eight Weeks Whilst Keeping Their Normalised Insulin Growth Factor (IGF-1) Levels (Age and Sex Adjusted)
NCT00701363 (15) [back to overview]Percentage of Subjects Having Maintained an Injection Interval of Six Weeks or Increasing Their Injection Interval to Eight Weeks
NCT00701363 (15) [back to overview]Mean Changes From Baseline in Quality of Life Scores (SF-36)
NCT00701363 (15) [back to overview]Mean Changes From Baseline in Quality of Life Scores (AcroQoL)
NCT00701363 (15) [back to overview]Subject Treatment Schedule Preference
NCT00701363 (15) [back to overview]Symptoms of Acromegaly (Headache, Excessive Perspiration, Fatigue, Soft Tissue Swelling and Arthralgia)
NCT00701363 (15) [back to overview]Mean Change From Baseline in IGF-1 Values [Expressed as % of Upper Limit of Normal (ULN)], Overall and by Injection Interval
NCT00701363 (15) [back to overview]Treatment Group (A, B or C) Mean Baseline IGF-1 Levels (Expressed as % of ULN) in Subjects Who Maintained Normalised IGF-1 Values at Week 48. Comparisons Will be Made as Follows: A Versus B, A Versus C, A Versus (B+C) and B Versus C
NCT00701363 (15) [back to overview]Correlation Between the Changes From Baseline in Quality of Life (AcroQoL) With the Corresponding Changes in IGF-1 Level (Expressed as % of ULN) at Each Visit
NCT00701363 (15) [back to overview]Percentage of Subjects With GH Level Less Than or Equal to 2.5 ng/mL
NCT00701363 (15) [back to overview]Percentage of Subjects With Normalised IGF-1 Levels (Age and Sex Adjusted)
NCT00701363 (15) [back to overview]Percentage of Subjects With Normalized IGF-1 Levels (Age and Sex Adjusted), Without Any Worsening of the AcroQoL Change Score Between Inclusion and Week 48
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 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.
NCT00774930 (10) [back to overview]Average Frequency of Flushing Events (Per Day) Based on Subject Diary Records.
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]"
NCT00842348 (2) [back to overview]Progression Free Survival (PFS): Kaplan-Meier Estimate
NCT00842348 (2) [back to overview]Adverse Events
NCT00891371 (6) [back to overview]Percentage of Patients Having Minimum Reduction of At Least 50% or Normalization of the Mean Number of Stools
NCT00891371 (6) [back to overview]Percentage of Patients Having Minimum Reduction of 50% or Normalization (≤3 Stools/24hours) in the Mean Number of Stools (Mean of Last 7 Days)
NCT00891371 (6) [back to overview]Percent Change in Mean Number of Stools Compared to Baseline
NCT00891371 (6) [back to overview]Change in QOL-Quality of Life {Assess Using Short Form (SF-36) and Irritable Bowel Syndrome (IBS)-QOL} Compared to Baseline
NCT00891371 (6) [back to overview]Change in Median Score of Stool Consistency (Bristol Stool Form Scale) Compared to Baseline
NCT00891371 (6) [back to overview]Change From Baseline in Relative Frequency of Normalization (≤3 Stools) in Subjects
NCT01137682 (16) [back to overview]Change From Baseline in Standardized IGF-1 Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for Extension Visits (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Percentage of Patients With Mean GH < 1.0 μg/L and Normalization of IGF-1, Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Percentage of Patients With Mean GH < 2.5 μg/L and Normalization of IGF-1, Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Percentage of Patients With Mean GH <1.0 μg/L Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Summary of Pasireotide Trough Concentrations in Acromegaly Patients Following Monthly i.m. Injections of Pasireotide LAR by Incident Dose From Start of Extension Phase up to Week 196 of the Extension Phase (PK Set)
NCT01137682 (16) [back to overview]Summary of Pasireotide Trough Concentrations in Acromegaly Patients Following Monthly i.m. Injections of Pasireotide LAR by Incident Dose From Start of Extension Phase up to Week 196 of the Extension Phase (PK Set)
NCT01137682 (16) [back to overview]Percentage of Participants With Normalization of Sex- and Age-adjusted IGF-1treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set).
NCT01137682 (16) [back to overview]Change From Baseline in Mean GH Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for Extension Visits (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Change From Baseline in Mean GH Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for CORE Visits (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Percentage of Patients With Mean GH < 2.5 μg/L Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Change From Baseline in Standardized IGF-1 Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for CORE Visits (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Change From Baseline in AcroQoL Total Scores for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for CORE Visits(Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Time to First Response (Weeks) by Treatment for Patients Achieving a Reduction of Mean GH Level to < 2.5 µg/L and Normalization of IGF-1 and Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly
NCT01137682 (16) [back to overview]Percentage of Participants With a Reduction of Mean GH Levels to < 2.5 µg/L and Normalization of Sex- and Age-adjusted IGF-1.
NCT01137682 (16) [back to overview]Duration of the First Response for Patients Achieving a Reduction of Mean GH Level to < 2.5 μg/L and Normalization of IGF-1 and Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)
NCT01137682 (16) [back to overview]Change From Baseline in AcroQoL Total Scores for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for Extension Visits (Extension Full Analysis Set)
NCT01538966 (1) [back to overview]Cost Effectiveness
NCT02075606 (9) [back to overview]Percentage of Subjects Alive and Progression Free at One Year
NCT02075606 (9) [back to overview]Assessment of Clinical Symptomatic Response
NCT02075606 (9) [back to overview]Mean Change From Baseline in Number of Episodes of Diarrhoea and Flushing
NCT02075606 (9) [back to overview]Mean Change From Baseline in Number of Episodes of Diarrhoea and Flushing
NCT02075606 (9) [back to overview]Mode Symptom Severity of Episodes of Flushing
NCT02075606 (9) [back to overview]Mode Symptom Severity of Episodes of Flushing
NCT02075606 (9) [back to overview]Percentage of Subjects With Time Point Responses According to Response Evaluation Criteria in Solid Tumours (RECIST) Assessments at Weeks 25 and 53
NCT02075606 (9) [back to overview]QoL Questionnaire: EORTC QLQ-G.I.NET21
NCT02075606 (9) [back to overview]Quality of Life (QoL) Questionnaire: European Organisation for Research and Treatment of Cancer (EORTC) QoL Questionnaire (QLQ)-C30
NCT02231762 (18) [back to overview]The Number of Subjects With a Symptomatic Response After 12 Months - Maintenance Phase
NCT02231762 (18) [back to overview]The Number of Subjects With a Symptomatic Response After 6 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using 5-Hydroxy-Indol-Amino-Acid (HIAA) Levels After 6 Months
NCT02231762 (18) [back to overview]DCR After 12 Months
NCT02231762 (18) [back to overview]Disease Control Rate (DCR) After 6 Months
NCT02231762 (18) [back to overview]Duration of Response (DoR) Within 12 Months
NCT02231762 (18) [back to overview]Progression-Free Survival (PFS) Within 12 Months
NCT02231762 (18) [back to overview]Time To Response (TtR) Within 12 Months
NCT02231762 (18) [back to overview]DCR by O6-methylguanine-DNA Methyl-transferase (MGMT) Expression and Methylation and Somatostatin Receptor (SSTR) Expression After 6 Months
NCT02231762 (18) [back to overview]EORTC QoL Questionnaire QLQ-C30: Mean Change From Baseline at 12 Months
NCT02231762 (18) [back to overview]European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life (QoL) Core 30 Questionnaire (QLQ-C30): Mean Change From Baseline at 6 Months
NCT02231762 (18) [back to overview]Pharmacokinetic (PK) Results: Lanreotide ATG 120 mg Serum Concentrations Within 12 Months
NCT02231762 (18) [back to overview]QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months
NCT02231762 (18) [back to overview]QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months
NCT02231762 (18) [back to overview]Quality of Life Gastrointestinal Neuroendocrine Tumour 21 Questionnaire (QLQ-GI.NET21): Mean Change From Baseline at 6 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using 5-HIAA Levels After 12 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using CgA Levels After 12 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using Chromogranin-A (CgA) Levels After 6 Months
NCT02275338 (9) [back to overview]Median Time Between First Lanreotide Autogel® Injection and Clinical Response in Phase 1
NCT02275338 (9) [back to overview]Percentage of Responders Before or at Day 7
NCT02275338 (9) [back to overview]Median Change From Baseline in Quality of Life as Assessed by ESAS in Phase 2
NCT02275338 (9) [back to overview]Median Change From Baseline in Number of Daily Episodes of Nausea in Phase 1
NCT02275338 (9) [back to overview]Median Change From Baseline in General Activity as Assessed by the Karnofsky Performance Status (KPS) Scale in Phase 1
NCT02275338 (9) [back to overview]Median Change From Baseline in Abdominal Pain Scores Assessed Using Visual Analogue Scale (VAS) in Phase 1
NCT02275338 (9) [back to overview]Median Change From Baseline in Quality of Life as Assessed by Edmonton Symptom Assessment System (ESAS) in Phase 1
NCT02275338 (9) [back to overview]Percentage of Responders Before or at Phase 2 Timepoints
NCT02275338 (9) [back to overview]Percentage of Responders in Phase 1
NCT02288377 (3) [back to overview]Progression-Free Survival
NCT02288377 (3) [back to overview]Overall Survival
NCT02288377 (3) [back to overview]Proportion of Patients Alive and Progression-free at 6 Months
NCT02365584 (10) [back to overview]Mean Change From Baseline in Number of Daily Vomiting Episodes; ITT Population
NCT02365584 (10) [back to overview]Mean Change From Baseline in Performing General Activity (Karnofsky Performance Status [KPS]); ITT Population
NCT02365584 (10) [back to overview]Mean Change From Baseline in Single ESAS Items Symptom Scores; ITT Population
NCT02365584 (10) [back to overview]Mean Daily NGT Secretion Volume, in Patients With a NGT
NCT02365584 (10) [back to overview]Mean Change From Baseline in ESAS Total Score; FAS
NCT02365584 (10) [back to overview]Number of Patients Experiencing ≤ 2 Vomiting Episodes/Day During at Least 3 Consecutive Days, in Patients Without NGT
NCT02365584 (10) [back to overview]Assessment of Passage of Stools; ITT Population
NCT02365584 (10) [back to overview]Least Squares (LS) Mean Area Under Curve (AUC) of Edmonton Symptom Assessment System (ESAS) Total Scores Collected for the First 7 Days; Full Analysis Set (FAS)
NCT02365584 (10) [back to overview]Mean Change From Baseline in Daily Intensity of Abdominal Pain Score (Visual Analogue Scale [VAS]); ITT Population
NCT02365584 (10) [back to overview]Mean Change From Baseline in ESAS Total Score; ITT Population
NCT02396953 (17) [back to overview]PK Analysis of Glycofurol Excipients: Cmax.
NCT02396953 (17) [back to overview]PK Analysis of Glycofurol Excipients: Tmax.
NCT02396953 (17) [back to overview]Overall Summary of Number of Subjects With AEs.
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]Determination of the Maximum Tolerated Dose (MTD) by Number of Subjects With DLTs.
NCT02396953 (17) [back to overview]PK Analysis of Lanreotide: Apparent Terminal Elimination Half-life (t1/2).
NCT02396953 (17) [back to overview]PK Analysis of Lanreotide: Area Under the Serum Concentration-time Curve Extrapolated to Infinity (AUC0-∞).
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 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]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 Insulin-like Growth Factor 1 (IGF-1).
NCT02396953 (17) [back to overview]PD Analysis: Mean Change From Baseline in Prolactin.
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]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 GH ≤2.5 Micrograms Per Litre (mcg/L) at the EOST/EW Visit
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 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]Percentage of Subjects With Normal Age-adjusted IGF-1 Levels 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 at Least One Symptom of Acromegaly at Week 13 and at the EOST/EW Visit Compared to Baseline
NCT02651987 (19) [back to overview]Mean Change From Baseline in PanNet Specific Tumour Biomarkers: Pancreatic Polypeptide, Gastrin
NCT02651987 (19) [back to overview]Mean Change From Baseline in QoL Questionnaire Gastrointestinal Neuroendocrine Tumour 21 (QLQ-GI.NET21; 2006)
NCT02651987 (19) [back to overview]Objective Response Rate (ORR)
NCT02651987 (19) [back to overview]Objective Response Rate (ORR)
NCT02651987 (19) [back to overview]Percentage of Subjects Alive and Progression Free
NCT02651987 (19) [back to overview]Percentage of Subjects Alive and Progression Free
NCT02651987 (19) [back to overview]Factors Associated With PFS
NCT02651987 (19) [back to overview]Mean Change From Baseline in EQ-5D-5L v1.0 Questionnaire (Descriptive System)
NCT02651987 (19) [back to overview]Mean Change From Baseline in EQ-5D-5L v1.0 Questionnaire (VAS)
NCT02651987 (19) [back to overview]Mean Change From Baseline in PanNet Specific Tumour Biomarkers: Glucagon
NCT02651987 (19) [back to overview]Mean Change From Baseline in QoL Measured Using EORTC, QLQ-C30 v3.0 (Global Health Status Sub-score)
NCT02651987 (19) [back to overview]Median Duration of Stable Disease
NCT02651987 (19) [back to overview]Median Progression Free Survival (PFS)
NCT02651987 (19) [back to overview]Median Time to Progression
NCT02651987 (19) [back to overview]Overall Survival
NCT02651987 (19) [back to overview]Best Overall Response Rate
NCT02651987 (19) [back to overview]Disease Control Rate (DCR)
NCT02651987 (19) [back to overview]Mean Change From Baseline in Nonspecific Tumour Biomarkers
NCT02651987 (19) [back to overview]Mean Change From Baseline in Number of Stools and Flushing Episodes
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 Change From Baseline in the Biomarker Chromogranin A (CgA) in the Double-Blind Phase and Open-Label Treatment Phase
NCT02683941 (12) [back to overview]Time to Treatment Failure (TTF) in the Double-Blind Phase
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]Percentage of Subjects Who Experienced QoL Deterioration
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]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]Median PFS Time in the Double-Blind Phase, Assessed by Central Review
NCT02683941 (12) [back to overview]Objective Response Rate (ORR) in the Double-Blind Phase
NCT02683941 (12) [back to overview]Median PFS Time in the Double-Blind Phase, Assessed by Local Review
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
NCT02698410 (15) [back to overview]Coefficient of Agreement Between Central and Local Assessment of Tumor Radiological Response at Month 9
NCT02698410 (15) [back to overview]DCR Assessed Centrally at Month 9
NCT02698410 (15) [back to overview]Best Overall Response (BOR) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]DCR Assessed Locally and Centrally at Month 12
NCT02698410 (15) [back to overview]DCR Assessed Locally and Centrally at Month 9 by Carcinoid Type
NCT02698410 (15) [back to overview]Disease Control Rate (DCR) Assessed Locally at Month 9
NCT02698410 (15) [back to overview]Influence of Biomarkers Expression on Locally and Centrally Assessed DCR at Months 9 and 12
NCT02698410 (15) [back to overview]Influence of Biomarkers Expression on Locally and Centrally Assessed PFS
NCT02698410 (15) [back to overview]Median Duration of Response (DOR) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]Median Progression Free Survival (PFS) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]Median Time to Progression (TTP) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]Median Time to Response (TTR) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]Neuron-Specific Enolase (NSE) and CgA Biomarker Levels
NCT02698410 (15) [back to overview]Objective Response Rate (ORR) Assessed Locally and Centrally at Months 9 and 12
NCT02698410 (15) [back to overview]Percentage of Biochemical Responders According to Chromogranin A (CgA) Plasma Levels
NCT02859064 (5) [back to overview]Number of Participants With Treatment-emergent Adverse Events as a Measure of Safety and Tolerability
NCT02859064 (5) [back to overview]Disease Control Rate
NCT02859064 (5) [back to overview]Overall Response Rate
NCT02859064 (5) [back to overview]Overall Survival
NCT02859064 (5) [back to overview]Progression Free Survival
NCT03012594 (2) [back to overview]Effect of Lanreotide on Gastrointestinal Motility as Measured by Smart Pill
NCT03012594 (2) [back to overview]"Improvement in Symptoms as Accessed by Patient Assessment of Upper GastroIntestinal Symptom Severity Index"
NCT03043664 (5) [back to overview]Number of Participants Experiencing Treatment-related AEs Leading to Drug Discontinuations During the First 12 Weeks of Treatment
NCT03043664 (5) [back to overview]Objective Response Rate (ORR) as Measured by Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1)
NCT03043664 (5) [back to overview]ORR as Measured by Immune-related Response Criteria (irRC)
NCT03043664 (5) [back to overview]Months of Progression-free Survival (PFS)
NCT03043664 (5) [back to overview]Months of Overall Survival (OS)
NCT03174353 (3) [back to overview]Overall Postoperative Morbidity
NCT03174353 (3) [back to overview]Combined Clinically Significant Postoperative Pancreatic Fistula and Intra-abdominal Abscess
NCT03174353 (3) [back to overview]Biochemical Leak (Grade A Postoperative Pancreatic Fistula)

Change in the Global Health Status Quality of Life Assessment

Transformed scores from European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire responses (QLQ)-C30. Questionnaire response scores range from 0 to 100. Higher scores indicate best possible Quality of Life. (NCT00353496)
Timeframe: Week 12 to Week 96 (last visit)

Interventionscore on a scale (Least Squares Mean)
Lanreotide (Autogel Formulation)-5.2
Placebo-4.9

[back to top]

Percentage of Patients With a Greater Than or Equal to 50% Decrease in Plasma Chromogranin A (CgA) Levels

(NCT00353496)
Timeframe: Week 12 to Week 96 (last visit)

Interventionpercentage of participants (Number)
Lanreotide (Autogel Formulation)42.2
Placebo4.7

[back to top]

Percentage of Patients Still Alive Based on Available Overall Survival Data

Overall survival defined as the time from randomisation to death due to any cause. Subjects were followed for overall survival beyond study completion/withdrawal via annual telephone contact until the last subject completed the study. (NCT00353496)
Timeframe: Randomisation to death or last visit, up to 321 weeks

Interventionpercentage of participants (Number)
Lanreotide (Autogel Formulation)84
Placebo77

[back to top]

Pharmacokinetic Profile of Lanreotide

Pharmacokinetic Profile of Lanreotide assessed by mean serum concentration at specified timepoints (NCT00353496)
Timeframe: Week 4, 12, 24, 36, 48, 72, 96

Interventionng/mL (Mean)
At Week 4 predose (n=81)At Week 12 predose (n=87)At Week 24 predose (n=74)At Week 36 predose (n=67)At Week 48 predose (n=62)At Week 72 predose (n=52)At Week 96 predose (n=48)
Lanreotide (Autogel Formulation)2.55.06.16.26.66.86.6

[back to top]

Percentage of Patients Alive & Without Disease Progression

Percentage of patients still ongoing (or completing at Week 96) without centrally assessed disease progression or death at Weeks 48 and 96. (NCT00353496)
Timeframe: Week 48 & 96

,
InterventionPercentage of participants (Number)
Week 48Week 96
Lanreotide (Autogel Formulation)6653
Placebo4925

[back to top]

Progression-Free Survival (PFS)

Time from randomization to first documentation of disease progression, or death. Disease progression centrally assessed using Response Evaluation Criteria in Solid Tumours (RECIST) v1.0 (NCT00353496)
Timeframe: From randomisation up to the last tumour assessment (scheduled at 96 weeks). Radiological scans were performed every 12 weeks during the first year and every 24 weeks during the second year

InterventionWeeks (Median)
Lanreotide (Autogel Formulation)NA
Placebo72

[back to top]

Percentage of Subjects With Serum GH Levels Lesser or Equal to 2.5 ng/mL During the Study

Serum samples were assessed for GH levels at the same timepoints as the IGF-1 sample at V1 and V2 and during the OGTT (non diabetic subjects only) at V3 and V11 (or at the early withdrawal visit in case of premature discontinuation). Five samples were taken over 2 hours in order to assess GH nadir and confirm the subject's eligibility before entering the co-administration period. For diabetic subjects, the OGTT was replaced by a measurement of GH level on a blood sample taken at the same time as IGF-1 at V3 and V11 (or at the early withdrawal visit). The percentage of subjects with serum GH levels ≤ 2.5 ng/mL at Baseline, V11 and LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpercentage of subjects (Number)
At V3At V11At LVA
Lanreotide Autogel + Pegvisomant Co-administration38.625.022.8

[back to top]

Percentage of Subjects With Acromegaly With a Normalised (Age and Sex Adjusted) IGF-1 Level at the End of the Co-administration Period

Serum IGF-1 is a well known and validated marker of acromegaly. IGF-1 assessments were performed at Visit (V) 1, V2, V3, V5, V7, V9 and V11 (or in case of premature study discontinuation, at the early withdrawal visit) and were based on a single serum sample taken in fasting conditions, prior to investigational medicinal product (IMP) administration. Percentage of subjects with a normalised (age and sex adjusted) IGF-1 level at the end of the co-administration period are presented. The last observation carried forward (LOCF) was used to replace missing IGF-1 values. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpercentage of subjects (Number)
Lanreotide Autogel + Pegvisomant Co-administration57.9

[back to top]

Change From Baseline in Acid Labile Subunit Levels From Baseline During the Co-administration Period

Serum samples were assessed for acid labile subunit levels at V1, V2, V3 and V11 (or at the early withdrawal visit in case of premature discontinuation). The change in mean acid labile subunit levels from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmilli IU per millilitre (mIU/mL) (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-561-541

[back to top]

Change From Baseline in Mean PR Interval, QRS Interval, QT Interval, RR Interval and QTcF During the Co-administration Period

Twelve-lead ECG recordings were performed at V2, V3 and V11. Sinus rhythm, heart rate, PR interval, RR interval, QRS interval and QT interval were measured and QTcF was calculated. The change in mean ECG parameter for PR interval, QRS interval, QT interval, RR interval and QTcF from Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmilliseconds (ms) (Mean)
PR Interval: Change at V11PR Interval: Change at LVAQRS Interval: Change at V11QRS Interval: Change at LVAQT Interval: Change at V11QT Interval: Change at LVARR Interval: Change at V11RR Interval: Change at LVAQTcF: Change at V11QTcF: Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration1.11.30.10.34.02.434.727.6-1.8-2.2

[back to top]

Change From Baseline in Mean Weight From Baseline During the Co-administration Period

Weight was recorded at V2, V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). The change in mean weight from Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionkilograms (kg) (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-0.3-0.3

[back to top]

Change From Baseline in Mean Supine Systolic and Diastolic Blood Pressure (BP) During the Co-administration Period

Blood pressure (supine after resting for 3 minutes) was recorded at V1, V2, V3, V5, V7, V9 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). The change in mean BP (systolic and diastolic) from Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmillimetres mercury (mmHg) (Mean)
Supine Systolic BP: Change at V11Supine Systolic BP: Change at LVASupine Diastolic BP: Change at V11Supine Diastolic BP: Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-0.40.2-0.1-0.3

[back to top]

Change From Baseline in Mean Supine Heart Rate During the Co-administration Period

Heart rate (supine after resting for 3 minutes) was recorded at V1, V2, V3, V5, V7, V9 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). The change in mean heart rate from Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionbeats per minute (bpm) (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-3.1-2.5

[back to top]

Change From Baseline in Mean Pituitary Tumour Size During the Co-administration Period

Pituitary tumour size was assessed by Magnetic Resonance Imaging at V2, V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). The two longest diameters of the pituitary tumour were to be measured. The change in mean pituitary tumour size from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmm^3 (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration2.46.3

[back to top]

Change From Baseline in Mean Glycosylated Haemoglobin (HbA1C) During the Co-administration Period; Assessed in Non Diabetic and Diabetic Subjects

Glycosylated haemoglobin (HbA1C) was measured at V2, V3 and V11 (or in case of premature discontinuation, at the early withdrawal visit). The change in mean HbA1C in diabetic and non diabetic subjects from Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpercentage (Mean)
Diabetic Subjects: Change at V11Diabetic Subjects: Change at LVANon Diabetic Subjects: Change at V11Non Diabetic Subjects: Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-0.05-0.050.050.05

[back to top]

Change From Baseline in Prolactin Levels During the Co-administration Period

Serum samples were assessed for prolactin levels at V1, V2, V3 and V11 (or at the early withdrawal visit in case of premature discontinuation). The change in mean prolactin levels from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmicrograms per litre (mcg/L) (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-0.1-0.4

[back to top]

Change From Baseline in Mean Fasting Insulin Concentration During the Co-administration Period; Assessed in Non Diabetic Subjects

Glucose tolerance was only evaluated in non diabetic subjects. Glucose tolerance was assessed based on measurement of fasting blood glucose and insulin levels taken at V2 and OGTT performed at V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). A dose of 75 g oral glucose was given to subjects and blood samples then taken at 30, 60, 90 and 120 minutes after oral glucose to measure glucose, insulin and GH levels, and to evaluate the GH nadir level. The OGTT was performed after the assessment of IGF-1 and all safety laboratory tests, but before IMPs administration at V3. Glucose, insulin and GH levels were assessed before OGTT in fasting conditions and at the same time as IGF-1 assessment. The change in mean fasting insulin concentration from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpicomoles per litre (pmol/L) (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-12.7-11.7

[back to top]

Change From Baseline in Mean Fasting Insulin / Glucose Ratio During the Co-administration Period; Assessed in Non Diabetic Subjects

Glucose tolerance was only evaluated in non diabetic subjects. Glucose tolerance was assessed based on measurement of fasting blood glucose and insulin levels taken at V2 and OGTT performed at V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). A dose of 75 g oral glucose was given to subjects and blood samples then taken at 30, 60, 90 and 120 minutes after oral glucose to measure glucose, insulin and GH levels, and to evaluate the GH nadir level. The OGTT was performed after the assessment of IGF-1 and all safety laboratory tests, but before IMPs administration at V3. Glucose, insulin and GH levels were assessed before OGTT in fasting conditions and at the same time as IGF-1 assessment. The change in mean fasting insulin / glucose ratio from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionratio (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-2.61-2.33

[back to top]

Change From Baseline in Prothrombin Time (Expressed as a Percentage of Normal) During the Co-administration Period

Blood samples for clinical laboratory tests were taken at V1, V2, V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). During the co-administration period, hepatic toxicity was assessed at each visit (V5 to V10) by measuring ALT, AST, alkaline phosphatase, GGT, prothrombin time and total bilirubin. Prothrombin time was expressed as a percentage of the time taken for a control blood sample to clot (designated as 100%) and the mean change from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpercentage of time (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-3.7-3.6

[back to top]

Change From Baseline in Serum GH Binding Protein Levels During the Co-administration Period

Serum samples were assessed for GH binding protein levels at V1, V2, V3 and V11 (or at the early withdrawal visit in case of premature discontinuation). The change in mean serum GH binding protein from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpmol/L (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration841811

[back to top]

Change From Baseline in Serum GH Levels During the Co-administration Period

Serum samples were assessed for GH levels at the same timepoints as the IGF-1 sample at V1 and V2 and during the OGTT (non diabetic subjects only) at V3 and V11 (or at the early withdrawal visit in case of premature discontinuation). Five samples were taken over 2 hours in order to assess GH nadir and confirm the subject's eligibility before entering the co-administration period. For diabetic subjects, the OGTT was replaced by a measurement of GH level on a blood sample taken at the same time as IGF-1 at V3 and V11 (or at the early withdrawal visit). The change in mean serum GH levels from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionnanograms per millilitre (ng/mL) (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration6.66.6

[back to top]

Change From Baseline in Serum IGF-1 Levels (Expressed as Z-scores) During the Co-administration Period

The change in serum IGF-1 levels, expressed as z-scores calculated using the age and sex specific mean and standard deviation [SD] values from Baseline to V11 and to LVA are presented. A z-score between +/- 2 indicates a normal IGF-1 concentration. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionz-score (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-4.50-4.25

[back to top]

Change From Baseline in Total Bilirubin During the Co-administration Period

Blood samples for clinical laboratory tests were taken at V1, V2, V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). During the co-administration period, hepatic toxicity was assessed at each visit (V5 to V10) by measuring ALT, AST, alkaline phosphatase, GGT, prothrombin time and total bilirubin. The change in mean total bilirubin from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmicromoles per litre (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-1.3-1.2

[back to top]

Correlation Between the Changes in ACROQoL Assessments With the Corresponding Changes in Z-score of IGF-1 Levels Over the Run-in Period and Co-administration Period

The correlation between the changes in ACROQoL (expressed as standardised scores and undertaken for global score, physical and psychological dimension scores and appearance and personal relationships sub-dimension scores) over the run-in period (V3 minus V2) and co-administration period (V11 and LVA minus V3) with the corresponding changes in z-score for the IGF-1 level is presented. A decrease in IGF-1 z-score represents an improvement and an increase in ACROQoL score represents an improvement. Spearman's rank correlation (r) values are presented for change from V2 to V3 (Baseline) and from Baseline to V11/LVA for each of the specified ACROQoL categories. Corr = Correlation; Dim = Dimension; Relnship = Relationship. (NCT00383708)
Timeframe: At V2 (Day 1; Run-in), V3 (Week 12; Baseline) and V11 (Week 44)

Interventioncorrelation value (Number)
Corr: Change V2 to V3 for Global ScoreCorr: Change V3 to V11/LVA for Global ScoreCorr: Change V2 to V3 for Physical DimCorr: Change V3 to V11/LVA for Physical DimCorr: Change V2 to V3 for Psychological DimCorr: Change V3 to V11/LVA for Psychological DimCorr: Change V2 to V3 for AppearanceCorr: Change V3 to V11/LVA for AppearanceCorr: Change V2 to V3 for Personal RelnshipCorr: Change V3 to V11/LVA; Personal Relnship
Lanreotide Autogel + Pegvisomant Co-administration-0.160.09-0.170.14-0.10.08-0.120.04-0.010.02

[back to top]

Number of Subjects With Putative Antibodies to Lanreotide and to Pegvisomant During the Co-administration Period

Presence of putative antibodies to lanreotide and antibodies to pegvisomant were assessed prior to IMP administration at V2, V4 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). The number of subjects with putative antibodies to lanreotide and to pegvisomant during the co-administration period (Baseline up to V11) is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

InterventionNumber of subjects (Number)
Antibodies for LanreotideAntibodies for Pegvisomant
Lanreotide Autogel + Pegvisomant Co-administration46

[back to top]

Number of Subjects With Shift in Presence/Absence of Lithiasis and/or Sludge During Co-administration Period

A gallbladder ultrasound was performed at V2, V3, and V11 (or in case of premature study discontinuation, at the early withdrawal visit). Presence of lithiasis and sludge was recorded. Number of subjects who developed or resolved lithiasis and developed or resolved sludge, comparing Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionparticipants (Number)
Developed Sludge: V11 vs V3Resolved Sludge: V11 vs V3Developed Lithiasis: V11 vs V3Resolved Lithiasis: V11 vs V3Developed Sludge: LVA vs V3Resolved Sludge: LVA vs V3Developed Lithiasis: LVA vs V3Resolved Lithiasis: LVA vs V3
Lanreotide Autogel + Pegvisomant Co-administration35113521

[back to top]

Percentage of Subjects With a Normalised (Age and Sex Adjusted) IGF-1 Level at Any Time During the Co-administration Period

Serum IGF-1 is a well known and validated marker of acromegaly. IGF-1 assessments were performed at V1, V2, V3, V5, V7, V9 and V11 (or in case of premature study discontinuation, at the early withdrawal visit) and were based on a single serum sample taken in fasting conditions, prior to IMP administration. Percentage of subjects with a normalised (age and sex adjusted) IGF-1 level at least once during the co-administration period, summarised by 'while taking the final dose during co-administration' and 'at any time during co-administration' are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpercentage of subjects (Number)
While Taking Final Dose During Co-administrationAt Any Time During Co-administration
Lanreotide Autogel + Pegvisomant Co-administration66.778.9

[back to top]

Percentage of Subjects With Acromegaly With a Normalised (Age and Sex Adjusted) IGF-1 Level at the End of the Co-administration Period; Summarised by Diabetic Status at Baseline

Serum IGF-1 is a well known and validated marker of acromegaly. IGF-1 assessments were performed at V1, V2, V3, V5, V7, V9 and V11 (or in case of premature study discontinuation, at the early withdrawal visit) and were based on a single serum sample taken in fasting conditions, prior to IMP administration. Percentage of subjects with a normalised (age and sex adjusted) IGF-1 level at the end of the co-administration period, summarised by diabetic status are presented. The denominator used to calculate percentages was the number of subjects in each subgroup (diabetic and non diabetic). The LOCF approach was used to replace missing IGF-1 values. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionpercentage of subjects (Number)
Diabetic SubjectsNon Diabetic Subjects
Lanreotide Autogel + Pegvisomant Co-administration47.463.2

[back to top] [back to top]

Percentage of Subjects With Normalised (Age and Sex Adjusted) IGF-1 at Each Assessment

Serum IGF-1 is a well known and validated marker of acromegaly. IGF-1 assessments were performed at V1, V2, V3, V5, V7, V9 and V11 (or in case of premature study discontinuation, at the early withdrawal visit) and were based on a single serum sample taken in fasting conditions, prior to IMP administration. The percentage of subjects with a normalised (age and sex adjusted) IGF-1 level is presented. The denominator used to calculate the percentages was the number of ITT population subjects with an assessment at the visit. In addition to the data for each individual visit, the last value available (LVA) data is also presented. None of the ITT population subjects had serum IGF-1 normalised at V3, consistent with the criterion to continue in the study and be treated in the co-administration period. (NCT00383708)
Timeframe: V1 (Screening) up to V11 (Week 44)

Interventionpercentage of subjects (Number)
V1 (Screening)V2 (Day 1)V5 (Week 20)V7 (Week 28)V9 (Week 36)V11 (Week 44)LVA
Lanreotide Autogel + Pegvisomant Co-administration17.524.556.448.157.761.557.9

[back to top]

Change From Baseline in Mean Fasting Glucose Concentration During the Co-administration Period; Assessed in Non Diabetic Subjects

Glucose tolerance was only evaluated in non diabetic subjects. Glucose tolerance was assessed based on measurement of fasting blood glucose and insulin levels taken at V2 and OGTT performed at V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). A dose of 75 g oral glucose was given to subjects and blood samples then taken at 30, 60, 90 and 120 minutes after oral glucose to measure glucose, insulin and GH levels, and to evaluate the GH nadir level. The OGTT was performed after the assessment of IGF-1 and all safety laboratory tests, but before IMPs administration at V3. Glucose, insulin and GH levels were assessed before OGTT in fasting conditions and at the same time as IGF-1 assessment. The change in mean fasting glucose concentration from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmmol/L (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-0.05-0.09

[back to top]

Change From Baseline in Mean Blood Glucose Maximum Concentration (Cmax) From Oral Glucose Tolerance Test (OGTT) During the Co-administration Period; Assessed in Non Diabetic Subjects

Glucose tolerance was only evaluated in non diabetic subjects. Glucose tolerance was assessed based on measurement of fasting blood glucose and insulin levels taken at V2 and OGTT performed at V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). A dose of 75 g oral glucose was given to subjects and blood samples then taken at 30, 60, 90 and 120 minutes after oral glucose to measure glucose, insulin and GH levels, and to evaluate the GH nadir level. The OGTT was performed after the assessment of IGF-1 and all safety laboratory tests, but before IMPs administration at V3. Glucose, insulin and GH levels were assessed before OGTT in fasting conditions and at the same time as IGF-1 assessment. The change in mean blood glucose Cmax (as determined from OGTT) from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionmillimoles per litre (mmol/L) (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration0.530.61

[back to top]

Change From Baseline in Liver Function Test Parameters During the Co-administration Period

Blood samples for clinical laboratory tests were taken at V1, V2, V3 and V11 (or in case of premature study discontinuation, at the early withdrawal visit). During the co-administration period, hepatic toxicity was assessed at each visit (V5 to V10) by measuring alanine amino transferase (ALT), aspartate amino transferase (AST), alkaline phosphatase, gamma glutamyl transferase (GGT), prothrombin time and total bilirubin. The change in mean ALT, AST, GGT and alkaline phosphatase from Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

InterventionIU/L (Mean)
AST: Change at V11AST: Change at LVAALT: Change at V11ALT: Change at LVAGGT: Change at V11GGT: Change at LVAAlkaline Phosphatase: Change at V11Alkaline Phosphatase: Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration3.34.54.17.8-1.01.5-1.7-0.8

[back to top]

Change From Baseline in Electrocardiogram (ECG) Mean Heart Rate During the Co-administration Period

Twelve-lead ECG recordings were performed at V2, V3 and V11. Sinus rhythm, heart rate, PR interval, RR interval, QRS interval and QT interval were measured and heart rate corrected QT interval using the Fridericia method (QTcF) was calculated. The change in ECG mean heart rate from Baseline to V11 and to LVA is presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionbpm (Mean)
Change at V11Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-2.8-2.2

[back to top]

Change From Baseline in Acromegaly Symptoms During the Co-administration Period

Acromegaly symptoms, including arthralgia, excessive perspiration, fatigue, headache and soft tissue swelling were assessed with scores ranging from 0 (no symptoms) to 8 (severe, incapacitating symptoms). Symptoms were assessed by the subject in paper format before any other procedure planned during the visit. The change in acromegaly symptoms from Baseline to V11 and to LVA are presented. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionunits on a scale (Mean)
Arthralgia: Change at V11Arthralgia: Change at LVAExcessive Perspiration: Change at V11Excessive Perspiration: Change at LVAFatigue: Change at V11Fatigue: Change at LVAHeadache: Change at V11Headache: Change at LVASoft Tissue Swelling: Change at V11Soft Tissue Swelling: Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration-0.7-0.6-0.4-0.4-0.2-0.2-0.4-0.3-0.6-0.6

[back to top]

Change From Baseline in Acromegaly Quality of Life (ACROQoL) Assessments During the Co-administration Period

The ACROQoL is a health-related quality of life (QoL) questionnaire for patients with acromegaly consisting of 22 items measured on a 5-point Likert-type scale that assesses frequency of occurrence (always to never) or degree of agreement (completely agree to completely disagree) with the statements. The ACROQoL consists of questions that evaluate physical (8 items) and psychological aspects related to appearance and personal relations (7 items each). Answers are transformed to a percentage value, where 100 is the maximal (best) and 0 the minimum (worse) score depicting self-perceived quality QoL. An increase in ACROQoL score is associated with an improved QoL. The change in ACROQoL global score, physical and psychological dimension scores and appearance and personal relationships sub-dimension scores from Baseline to V11 and to LVA are presented. Relnship = Relationship; Dim = Dimension. (NCT00383708)
Timeframe: V3 (Week 12; Baseline) up to V11 (Week 44)

Interventionunits on a scale (Mean)
Global Score: Change at V11Global Score: Change at LVAPhysical Dimensions Score: Change at V11Physical Dimensions Score: Change at LVAPsychological Dimensions Score: Change at V11Psychological Dimensions Score: Change at LVAAppearance Sub-Dimension Score: Change at V11Appearance Sub-Dimension Score: Change at LVAPersonal Relnship Sub-Dim Score: Change at V11Personal Relnship Sub-Dim Score: Change at LVA
Lanreotide Autogel + Pegvisomant Co-administration2.42.24.23.51.31.43.83.7-0.1-0.9

[back to top]

Total Symptom Questionnaire Score at Week 24/Termination

Acromegaly symptoms are sweating, snoring, joint pain, headache and fatigue. Each symptom was scored as -2 = 'always', -1 = 'most of the time', 0 = 'sometimes', 1 = 'rarely and 2 = 'never'. The total score was used to evaluate symptom control in each patient at Week 0 and Week 24/Termination. The total worst score is -10 and best score is 10. (NCT00447499)
Timeframe: 24 Weeks

InterventionOn a Scale from -10 to 10 (Mean)
Somatuline Autogel (Lanreotide Acetate) Injection0.9

[back to top]

Change of GH Concentration Levels From Basaeline to Week 24 in Switch Patients

Blood samples taken before and 60 and 120 min after glucose load from fasting patient. (NCT00447499)
Timeframe: 24 Weeks

Interventionng/mL (Mean)
Somatuline Autogel (Lanreotide Acetate) Injection-0.3

[back to top]

Percentage of Switch Subjects That Have Glucose Suppressed GH Levels ≤ 2.5 ng/ml at the End of the Study, Week 24/Termination.

Blood samples taken before and 60 and 120 min after glucose load from fasting patient. (NCT00447499)
Timeframe: 24 Weeks

InterventionPercent of Participants (Number)
Somatuline Autogel (Lanreotide Acetate)89

[back to top]

Percentage of Switch Subjects Who Find Self-administration of Somatuline Autogel Convenient as Assessed by the Subject Convenience Questionnaire Score.

Experienced Convenience of Somatuline® Autogel® Injections was assessed by the subject as: Very convenient; somewhat convenient; neither convenient nor inconvenient; Neither convenient nor inconvenient; Somewhat inconvenient; very inconvenient. (NCT00447499)
Timeframe: 24 weeks

InterventionPercent of Participants (Number)
Somatuline Autogel (Lanreotide Acetate)/Switch Patient91

[back to top]

The Percentage of Subjects or Their Partners That Are Competent to Self-administer Somatuline Autogel at the End of the Study, (Week 24/Early Termination), as Assessed by the Competence Questionnaire Score.

The primary efficacy endpoint was the percentage of patients (Switch and other) or their partners who were competent to self-administer lanreotide at the end of the study (Week 24/Early Termination), as assessed by the Assessment of Competence Questionnaire (0 = 'No' and 1 = 'Yes'). (NCT00447499)
Timeframe: 24 weeks

InterventionPercentage of Participants (Number)
Somatuline Autogel (Lanreotide Acetate) Injection98

[back to top]

Total Health Care Professional Convenience Questionnaire Score at Week 24/Termination

Healthcare professional convenience questionnaires are: Confident the Subject Properly Administering the Injection; Subject Complained About Pain When Administering the Injection; Subject Appreciated the Option of Self-Injection at Home. Each Healthcare professional convenience questionnaire was scored -2, -1, 0, 1 and 2; from most negative to most positive response. A total score across all questions was calculated and was used to evaluate the convenience. The worst total score is -6 and best total score is 6. (NCT00447499)
Timeframe: 24 weeks

InterventionOn a Scale from -6 to 6 (Mean)
Somatuline Autogel (Lanreotide Acetate) Injection4.6

[back to top]

Percentage of Switch Subjects That Have IGF-1 Levels Within the Normal Range for Age and Gender at the End of the Study

Blood sample was collected while subject is in a fasting state or non-fasting state for measuring the level of IGF-1. (NCT00447499)
Timeframe: 24 weeks

InterventionPercent of Participants (Number)
Somatuline Autogel (Lanreotide Acetate)/Switch Patient94

[back to top]

5-hydroxyindoleacetic Acid (5-HIAA) Levels

"Biochemical control was assessed by analysing 5-HIAA levels at each site visit, which was judged as necessary by the investigator at each site.~'Before self or partner administration' was assessed at baseline for group 1 and at week 12 for group 2.~'After self or partner administration' was assessed at week 16 to 20 for group 1 and at week 12 for group 2.~'Before HCP administration' was assessed at week 16 to 20 for group 1 and at baseline for group 2.~'After HCP administration' was assessed at week 30 to 34 for group 1 and week 12 for group 2." (NCT00681187)
Timeframe: Group 1 - Baseline, week 16 to 20 and 30 to 34. Group 2 - Baseline, week 12 and 30 to 34.

Interventionnmol/l (Mean)
Before Self or Partner Administration220.17
After Self or Partner Administration219.17
Before HCP Administration217.08
After HCP Administration219.58

[back to top]

Chromogranin A Levels

"Biochemical control was assessed by analysing chromogranin A levels at each site visit, which was mandatory for all subjects.~'Before self or partner administration' was assessed at baseline for group 1 and at week 12 for group 2.~'After self or partner administration' was assessed at week 16 to 20 for group 1 and at week 12 for group 2.~'Before HCP administration' was assessed at week 16 to 20 for group 1 and at baseline for group 2.~'After HCP administration' was assessed at week 30 to 34 for group 1 and week 12 for group 2." (NCT00681187)
Timeframe: Group 1 - Baseline, week 16 to 20 and 30 to 34. Group 2 - Baseline, week 12 and 30 to 34.

Interventionnmol/l (Mean)
Before Self or Partner Administration37.48
After Self or Partner Administration47.85
Before HCP Administration42.05
After HCP Administration37.42

[back to top]

Days Sick Leave

Health care and patient costs associated with the treatment of carcinoid symptoms in subjects treated with lanreotide Autogel were assessed through recording loss of production for subject through total number of days sick leave of the employed patients (n=6). (NCT00681187)
Timeframe: Group 1 - between week 8 to 20 (self or partner administration), between week 20 to 32 (HCP administration). Group 2 - between week 20 to 32 (self or partner administration), between week 0 to week 12 (HCP administration)

Interventiondays (Number)
Self or Partner Administration23
HCP Administration0

[back to top]

Healthcare Professionals With Positive Response to Specified Questions on Self or Partner Administration Method

"Assessed by the number of HCP with a positive response 'yes' to two questions:~Based on your experience during this trial, did you feel confident in the safety of your patients?~Based on your experience during this trial, would you recommend suitable patients to try self or partner administration?" (NCT00681187)
Timeframe: Between week 30 to 34

Interventionparticipants (Number)
Overall Study Group9

[back to top]

Number of Patients Stating at Least One Injection Negatively Interfered With Psychological Wellbeing

The subject was asked: 'Does the treatment administration used today negatively interfere with your psychological wellbeing?' (NCT00681187)
Timeframe: Between baseline to week 32, after each injection (8-9 injections)

Interventionparticipants (Number)
Training Period4
Self or Partner Administration2
HCP Administration1

[back to top]

Subject Preference for Self or Partner Administration

A global question was asked: 'If you could choose, which administration method would you like to use on a regular basis?' A) Healthcare professional provided injection B) Self/ partner administered injection (NCT00681187)
Timeframe: Between week 30 to 34

Interventionparticipants (Number)
Group 1 Self or Partner First Then HCP Administration10
Group 2 HCP Then Self or Partner Administration12

[back to top]

Total Number of Visits to HCP Due to Carcinoid Symptoms

Health care and patient costs associated with the treatment of carcinoid symptoms in subjects treated with lanreotide Autogel were assessed by recording the total number of visits made by participants (n=12) to HCP due to carcinoid symptoms. (NCT00681187)
Timeframe: Group 1 - between week 8 to 20 (self or partner administration), between week 20 to 32 (HCP administration). Group 2 - between week 20 to 32 (self or partner administration), between week 0 to week 12 (HCP administration)

Interventionvisits (Number)
Self or Partner Administration17
HCP Administration25

[back to top]

Perceived Symptom Control Evaluation in Respect to Episodes of Diarrhoea

Participants were asked how they perceived the symptoms in respect to episodes of diarrhoea since the last injection. Participants included in the study were previously treated with lanreotide autogel and therefore the assessment at baseline was made in comparison to previous injection outside of the study protocol. (NCT00681187)
Timeframe: Group 1 - baseline, week 16 to 20 (self or partner administration) and week 30 to 34 (HCP administration). Group 2 - baseline, week 12 to 16 (HCP administration) and week 30 to 34 (self or partner administration).

,,
Interventionparticipants (Number)
WorsenedSimilarImprovedMissing
Baseline12220
HCP Administration02410
Self or Partner Administration41731

[back to top]

Perceived Symptom Control Evaluation in Respect to Episodes of Flushing

Participants were asked how they perceived the symptoms in respect to episodes of flushing since the last injection. Participants included in the study were previously treated with lanreotide Autogel and therefore the assessment at baseline was made in comparison to their previous injection outside of the study protocol. (NCT00681187)
Timeframe: Group 1 - baseline, week 16 to 20 (self or partner administration) and week 30 to 34 (HCP administration). Group 2 - baseline, week 12 (HCP administration) and week 30 (self or partner administration).

,,
Interventionparticipants (Number)
WorsenedSimilarImprovedMissing
Baseline02410
HCP Administration12040
Self or Partner Administration12121

[back to top]

Number of Patients Stating at Least One Injection Interfered With Daily Activities

The subject was asked: 'Does the treatment administration used today interfere with your daily activities?' (NCT00681187)
Timeframe: Between baseline to week 32, after each injection (8-9 injections)

InterventionParticipants (Number)
Training Period6
Self or Partner Administration2
HCP Administration6

[back to top]

Number of Patients With at Least a 20% Reduction in Tumour Volume From Baseline Volume (Visit 1) to Week 12 (Visit 3) and Week 24 (Visit 4).

(NCT00690898)
Timeframe: Baseline (week 1) to week 12 and week 24

Interventionparticipants (Number)
Greater than or equal to 20% at week 12 (n=85)Less than 20% at week 12 (n=85)Greater than or equal to 20% at week 24 (n=80)Less than 20% at week 24 (n=80)
Lanreotide Autogel 120 mg46394535

[back to top]

Percent Variation From Baseline to Visit 3, 4 and 5 (Week 12, 24, and 48) of IGF-1 Levels

(NCT00690898)
Timeframe: Week 12, 24, and 48

Interventionpercentage change (Mean)
Week 12 (n=85)Week 24 (n=78)Week 48 (n=62)
Lanreotide Autogel 120 mg-43.8-47.4-56.7

[back to top]

Percent Variation From Baseline to Visit 3, 4 and 5 (Week 12, 24, and 48) of Serum GH Levels.

(NCT00690898)
Timeframe: Week 12, 24, and 48

Interventionpercentage change (Mean)
Week 12 (n=85)Week 24 (n=78)Week 48 (n=63)
Lanreotide Autogel 120 mg-62.1-64.6-70.9

[back to top]

Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Arthralgia) From Baseline

The status of clinical signs of acromegaly assessed by an acromegaly symptoms questionnaire (paper form) completed by the patient at each study visit. The scoring for each clinical sign of acromegaly on the questionnaire is from 0 (no symptom) to 8 (severe, incapacitating symptom). The variation (or no variation) in scores indicate whether the clinical sign of acromegaly had improved, worsened or was unchanged. (NCT00690898)
Timeframe: Week 12, 24 and 48

Interventionpercentage of subjects (Number)
Baseline to week 12 - Worsened (n=88)Baseline to week 12 - Unchanged (n=88)Baseline to week 12 - Improved (n=88)Baseline to week 24 - Worsened (n=81)Baseline to week 24 - Unchanged (n=81)Baseline to week 24 - Improved (n=81)Baseline to week 48 - Worsened (n=62)Baseline to week 48 - Unchanged (n=62)Baseline to week 48 - Improved (n=62)
Lanreotide Autogel 120 mg9.126.164.811.124.764.217.722.659.7

[back to top]

Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Fatigue) From Baseline

The status of clinical signs of acromegaly assessed by an acromegaly symptoms questionnaire (paper form) completed by the patient at each study visit. The scoring for each clinical sign of acromegaly on the questionnaire is from 0 (no symptom) to 8 (severe, incapacitating symptom). The variation (or no variation) in scores indicate whether the clinical sign of acromegaly had improved, worsened or was unchanged. (NCT00690898)
Timeframe: Week 12, 24 and 48

Interventionpercentage of subjects (Number)
Baseline to week 12 - Worsened (n=88)Baseline to week 12 - Unchanged (n=88)Baseline to week 12 - Improved (n=88)Baseline to week 24 - Worsened (n=81)Baseline to week 24 - Unchanged (n=81)Baseline to week 24 - Improved (n=81)Baseline to week 48 - Worsened (n=62)Baseline to week 48 - Unchanged (n=62)Baseline to week 48 - Improved (n=62)
Lanreotide Autogel 120 mg13.625.061.416.017.366.714.529.056.5

[back to top]

Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Headache) From Baseline

The status of clinical signs of acromegaly assessed by an acromegaly symptoms questionnaire (paper form) completed by the patient at each study visit. The scoring for each clinical sign of acromegaly on the questionnaire is from 0 (no symptom) to 8 (severe, incapacitating symptom). The variation (or no variation) in scores indicate whether the clinical sign of acromegaly had improved, worsened or was unchanged. (NCT00690898)
Timeframe: Week 12, 24 and 48

Interventionpercentage of subjects (Number)
Baseline to week 12 - Worsened (n=88)Baseline to week 12 - Unchanged (n=88)Baseline to week 12 - Improved (n=88)Baseline to week 24 - Worsened (n=81)Baseline to week 24 - Unchanged (n=81)Baseline to week 24 - Improved (n=81)Baseline to week 48 - Worsened (n=62)Baseline to week 48 - Unchanged (n=62)Baseline to week 48 - Improved (n=62)
Lanreotide Autogel 120 mg13.646.639.811.142.046.914.546.838.7

[back to top]

Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Soft Tissue Swelling) From Baseline

The status of clinical signs of acromegaly assessed by an acromegaly symptoms questionnaire (paper form) completed by the patient at each study visit. The scoring for each clinical sign of acromegaly on the questionnaire is from 0 (no symptom) to 8 (severe, incapacitating symptom). The variation (or no variation) in scores indicate whether the clinical sign of acromegaly had improved, worsened or was unchanged. (NCT00690898)
Timeframe: Week 12, 24 and 48

Interventionpercentage of subjects (Number)
Baseline to week 12 - Worsened (n=88)Baseline to week 12 - Unchanged (n=88)Baseline to week 12 - Improved (n=88)Baseline to week 24 - Worsened (n=81)Baseline to week 24 - Unchanged (n=81)Baseline to week 24 - Improved (n=81)Baseline to week 48 - Worsened (n=62)Baseline to week 48 - Unchanged (n=62)Baseline to week 48 - Improved (n=62)
Lanreotide Autogel 120 mg10.226.163.611.122.266.714.519.466.1

[back to top]

Percentage of Patients With Relevant Reduction in Pituitary Tumour Volume (as Measured by MRI) From Baseline Volume (Visit 1) to Week 48 (After 12 Injections at Visit 5)

A blinded, centrally assessed evaluation of all MRIs was performed. A 20% reduction from the volume at Visit 1 was considered to be clinically relevant. (NCT00690898)
Timeframe: Week 1 and Week 48

Interventionpercentage of subjects (Number)
Greater than or equal to 20%Less than 20%
Lanreotide Autogel 120 mg62.937.1

[back to top]

Percentage of Patients With Improved, Unchanged or Worsened Clinical Signs of Acromegaly (Excessive Perspiration) From Baseline

The status of clinical signs of acromegaly assessed by an acromegaly symptoms questionnaire (paper form) completed by the patient at each study visit. The scoring for each clinical sign of acromegaly on the questionnaire is from 0 (no symptom) to 8 (severe, incapacitating symptom). The variation (or no variation) in scores indicate whether the clinical sign of acromegaly had improved, worsened or was unchanged. (NCT00690898)
Timeframe: Week 12, 24 and 48

Interventionpercentage of subjects (Number)
Baseline to week 12 - Worsened (n=88)Baseline to week 12 - Unchanged (n=88)Baseline to week 12 - Improved (n=88)Baseline to week 24 - Worsened (n=81)Baseline to week 24 - Unchanged (n=81)Baseline to week 24 - Improved (n=81)Baseline to week 48 - Worsened (n=62)Baseline to week 48 - Unchanged (n=62)Baseline to week 48 - Improved (n=62)
Lanreotide Autogel 120 mg11.425.063.614.821.064.29.724.266.1

[back to top]

Change From Baseline to Visit 3, 4 and 5 (Week 12, 24, and 48) of Prolactin Levels

(NCT00690898)
Timeframe: Week 12, 24 and 48

Interventionmcg/L (Mean)
Prolactin levels at baseline (n=21)Change From Baseline to Week 12 (n=20)Change From Baseline to Week 24 (n=20)Change From Baseline to Week 48 (n=12)
Lanreotide Autogel 120 mg48.0-18.0-18.6-17.3

[back to top]

Changes in the Global Acromegaly Quality of Life Assessment (AcroQoL) From Baseline

Acromegaly Quality of Life Assessment (AcroQoL) questionnaire response scores range from 0 to 100. Higher scores indicate best possible Quality of Life. (NCT00690898)
Timeframe: Week 12, 24 and 48

Interventionunits on a scale (Mean)
Baseline to week 12 (n=82)Baseline to week 24 (n=75)Baseline to week 48 (n=59)
Lanreotide Autogel 120 mg7.88.09.5

[back to top]

Mean Baseline IGF-1 Levels (Expressed as % of ULN) in All Groups (A, B and C) Versus Mean Baseline IGF-1 Levels (Expressed as % of ULN) in Subjects With Uncontrolled IGF-1 Levels at Week 24

(NCT00701363)
Timeframe: Baseline (visit 1)

InterventionPercentage of ULN (Mean)
Uncontrolled IGF-1 levels at Week 24 (n=14)Normalized IGF-1 levels at Week 24 (A+B+C) (n=108)Difference in Mean Baseline
Overall Study95.9267.4928.43

[back to top]

Serum Growth Hormone (GH) Levels

(NCT00701363)
Timeframe: At Baseline, week 24 and week 48

,,,,
Interventionng/mL (Mean)
ITT - Baseline (n=15, 13, 70, 26, and 124)ITT - Week 24 (n=3, 13, 70, 26, and 112)ITT - Week 48 (n=0, 13, 68, 26, and 107)MITT - Baseline (n=0, 13, 70, 26, and 109)MITT - Week 24 (n=0, 13, 70, 26, and 109)MITT - Week 48 (n=0, 13, 68, 26, and 107)
Overall Study0.970.990.920.960.880.92
Phase 1 Only: Lanreotide Autogel 120 mg1.044.89NANANANA
Phase 2 (Group A): Lanreotide Autogel 120 mg Every 4 Weeks0.920.900.690.920.900.69
Phase 2 (Group B): Lanreotide Autogel 120 mg Every 6 Weeks0.920.950.990.920.950.99
Phase 2 (Group C): Lanreotide Autogel 120 mg Every 8 Weeks1.080.670.831.080.670.83

[back to top]

Percentage of Subjects Who Extend Their Injection Interval to Eight Weeks During Phase 2 of the Study, Whilst Maintaining Normalised IGF-1 Levels

The criterion for a subject is satisfied if he extended his injection interval to eight weeks during Phase 2 of the study, whilst maintaining normalised IGF-1 levels at Week 48. (NCT00701363)
Timeframe: At week 48

InterventionPercentage of subjects (Number)
Intention-to-Treat (n=124)Modified Intention-to-Treat (n=109)
Overall Study20.222.9

[back to top]

Percentage of Subjects Having Maintained Their Injection Interval Schedule of Six Weeks or Increased Their Injection Interval to Eight Weeks Whilst Keeping Their Normalised Insulin Growth Factor (IGF-1) Levels (Age and Sex Adjusted)

A subject was responder if he maintained his injection interval schedule of 6 weeks or increased his injection interval to eight weeks whilst keeping his normalised IGF-1 level (age and sex adjusted) at the end of the study (Week 48) (NCT00701363)
Timeframe: At week 48 (End of Study)

InterventionPercentage of subjects (Number)
Intention-to-Treat (n=124)Modified Intention-to-Treat (n=109)
Overall Study75.886.2

[back to top]

Percentage of Subjects Having Maintained an Injection Interval of Six Weeks or Increasing Their Injection Interval to Eight Weeks

The criterion for a subject is satisfied if he maintained an injection interval of six weeks or increasing his injection interval to eight weeks during Phase 2 of the study. (NCT00701363)
Timeframe: During phase 2 of the study (up to week 48)

InterventionPercentage of subjects (Number)
Intention-to-Treat (n=124)Modified Intention-to-Treat (n=109)
Overall Study78.288.1

[back to top]

Mean Changes From Baseline in Quality of Life Scores (SF-36)

Short Form-36 questionnaire (SF-36) score comprises eight components: Physical function, role-physical, bodily pain, general health, vitality, social functioning, role-emotional and mental health on a scale of 100, where a score of 100 corresponds to the best possible QoL and 0 to the worst. (NCT00701363)
Timeframe: At weeks 24 and 48

,,,,
InterventionUnits on a scale (Mean)
At week 24 (n=3, 13, 70, 26, and 112)At week 48 (n=0, 13, 68, 26, and 107)
Overall Study1.030.06
Phase 1 Only: Lanreotide Autogel 120 mg-0.66NA
Phase 2 (Group A): Lanreotide Autogel 120 mg Every 4 Weeks4.914.96
Phase 2 (Group B): Lanreotide Autogel 120 mg Every 6 Weeks1.79-0.61
Phase 2 (Group C): Lanreotide Autogel 120 mg Every 8 Weeks-2.75-0.62

[back to top]

Mean Changes From Baseline in Quality of Life Scores (AcroQoL)

AcroQoL score groups 22 components: Eight physical, Seven psychological appearance and Seven psychological personal relations, adjusted to a scale of 100, where a score of 100 corresponds to the best possible QoL and 0 to the worst. (NCT00701363)
Timeframe: At weeks 24 and 48

,,,,
InterventionUnits on a scale (Mean)
At week 24 (n=1, 9, 55, 21, and 86)At week 48 (n=0, 9, 53, 21, and 83)
Overall Study-0.35-1.08
Phase 1 Only: Lanreotide Autogel 120 mg21.59NA
Phase 2 (Group A): Lanreotide Autogel 120 mg Every 4 Weeks-2.151.31
Phase 2 (Group B): Lanreotide Autogel 120 mg Every 6 Weeks-0.27-1.32
Phase 2 (Group C): Lanreotide Autogel 120 mg Every 8 Weeks-0.84-1.52

[back to top]

Subject Treatment Schedule Preference

"At week 24, the preference assessed between Octreotide Long Acting Repeatable intramuscular injection (Oct-LAR IM) every 4 weeks and Lanreotide Autogel 120 mg subcutaneous injection (SC) every 6 weeks.~At week 48, the preference is assessed between Oct-LAR IM every 4 weeks and Lanreotide Autogel 120 mg SC either injected every 4, 6 or 8 weeks (as injected during Phase II of the study)." (NCT00701363)
Timeframe: At weeks 24 and 48

InterventionPercentage of subjects (Number)
W24:Oct-LAR IM inj evy 4 wks Phs 1 only (n=15)W24:Lan Autogel inj evy 6 wks Phs 1 only (n=15)W24:Oct-LAR IM inj evy 4 wks Phs 1/Grp A(n=13)W24:Lan Autogel inj evy 6 wks Phs 1/Grp A(n=13)W24:Oct-LAR IM inj evy 4 wks Phs 1/Grp B(n=70)W24:Lan Autogel inj evy 6 wks Phs 1/Grp B(n=70)W24:Non precised Phs 1/Grp B (n=70)W24:Oct-LAR IM inj evy 4 wks Phs 1/Grp C(n=26)W24:Lan Autogel inj every 6 wks Phs 1/Grp C(n=26)W 24:Oct-LAR IM inj evy 4 wks-Overall study(n=124)W24:Lan Autogel inj evy 6 wks-Overall study(n=124)W 24: Non precised - Overall study (n=124)W48:Oct-LAR IM inj evy 4 wks Phs 1/Grp A(n=13)W48:Lan Autogel inj evy 6 wks Phs 1/Grp A(n=13)W48:Non precised Phs 1/Grp A (n=13)W48:Oct-LAR IM inj evy 4 wks Phs 1/Grp B(n=70)W48:Lan Autogel inj evy 4 wks Phs 1/Grp B(n=70)W48:Lan Autogel inj evy 6 wks Phs 1/Grp B(n=70)W48:Non precised Phs 1/Grp B (n=70)W48:Oct-LAR IM inj evy 4 wks Phs 1/Grp C(n=26)W48:Lan Autogel inj evy 8 wks Phs 1/Grp C(n=26)W48:Oct-LAR IM inj evy 4 wks-Overall study(n=124)W48:Lan Autogel inj evy 4 wks-Overall study(n=124)W48:Lan Autogel inj evy 6 wks-Overall study(n=124)W48:Lan Autogel inj evy 8 wks-Overall study(n=124)W48:Non precised-Overall study (n=124)
Overall Study20807.792.310.385.34.43.896.28.988.42.715.476.97.714.71.577.95.97.792.313.110.349.522.44.7

[back to top]

Symptoms of Acromegaly (Headache, Excessive Perspiration, Fatigue, Soft Tissue Swelling and Arthralgia)

Acromegaly symptoms were assessed by the patients using the Patient Assessed Acromegaly Symptom Questionnaire (PASQ) scale ranging from 0 (No symptoms) to 8 (Severe, incapacitating symptoms). (NCT00701363)
Timeframe: At baseline, week 24 and week 48

,,,
InterventionUnits on a scale (Mean)
Baseline - HeadacheBaseline - Excessive perspirationBaseline - FatigueBaseline - Soft tissue swellingBaseline - ArthralgiaWeek 24 - HeadacheWeek 24 - Excessive perspirationWeek 24 - FatigueWeek 24 - Soft tissue swellingWeek 24 - ArthralgiaWeek 48 - HeadacheWeek 48 - Excessive perspirationWeek 48 - FatigueWeek 48 - Soft tissue swellingWeek 48 - Arthralgia
Phase 1 Only: Lanreotide Autogel 120 mg2.732.803.401.132.732.672.002.331.332.00NANANANANA
Phase 2 (Group A): Lanreotide Autogel 120 mg Every 4 Weeks2.151.924.232.773.461.382.083.852.463.621.772.313.772.853.46
Phase 2 (Group B): Lanreotide Autogel 120 mg Every 6 Weeks1.802.413.271.813.131.932.253.361.713.092.322.503.162.103.60
Phase 2 (Group C): Lanreotide Autogel 120 mg Every 8 Weeks2.042.083.311.424.422.042.644.081.763.682.622.503.921.624.04

[back to top]

Mean Change From Baseline in IGF-1 Values [Expressed as % of Upper Limit of Normal (ULN)], Overall and by Injection Interval

IGF-1 change from Baseline to Week 48 = Mean IGF-1 level at Week 48 - Mean IGF-1 level at Baseline (NCT00701363)
Timeframe: Baseline (visit 1) and week 48

InterventionPercentage of ULN (Mean)
Phase 2 (Group A): Lanreotide Autogel 120 mg Every 4 Weeks-1.70
Phase 2 (Group B): Lanreotide Autogel 120 mg Every 6 Weeks5.74
Phase 2 (Group C): Lanreotide Autogel 120 mg Every 8 Weeks-4.33

[back to top]

Treatment Group (A, B or C) Mean Baseline IGF-1 Levels (Expressed as % of ULN) in Subjects Who Maintained Normalised IGF-1 Values at Week 48. Comparisons Will be Made as Follows: A Versus B, A Versus C, A Versus (B+C) and B Versus C

(NCT00701363)
Timeframe: Baseline (visit 1)

InterventionPercentage of ULN (Mean)
Phase 2 (Group A): Lanreotide Autogel 120 mg Every 4 Weeks98.69
Phase 2 (Group B): Lanreotide Autogel 120 mg Every 6 Weeks67.75
Phase 2 (Group C): Lanreotide Autogel 120 mg Every 8 Weeks51.30

[back to top]

Correlation Between the Changes From Baseline in Quality of Life (AcroQoL) With the Corresponding Changes in IGF-1 Level (Expressed as % of ULN) at Each Visit

"AcroQoL change from Baseline to Week 24 (48) = AcroQoL at Week 24 (48) - AcroQoL at Baseline.~IGF-1 change from Baseline to Week 24 (48) = IGF-1 at Week 24 (48) - IGF-1 at Baseline.~Correlation presented is a Spearman correlation (non parametric)." (NCT00701363)
Timeframe: At weeks 24 and 48

InterventionCorrelation coefficient (Number)
Week 24: Phase I / Group A (n=8)Week 24: Phase I / Group B (n=55)Week 24: Phase I / Group C (n=21)Week 24: Overall study (n=85)Week 48: Phase I / Group A (n=8)Week 48: Phase I / Group B (n=53)Week 48: Phase I / Group C (n=21)Week 48: Overall study (n=82)
Overall Study0.43-0.02-0.41-0.010.69-0.06-0.09-0.01

[back to top]

Percentage of Subjects With GH Level Less Than or Equal to 2.5 ng/mL

(NCT00701363)
Timeframe: At weeks 24 and 48

InterventionPercentage of subjects (Number)
ITT: Week 24 - Phase I only (n=15)ITT: Week 24 - Phase I / Group A (n=13)ITT: Week 24 - Phase I / Group B (n=70)ITT: Week 24 - Phase I / Group C (n=26)ITT: Week 24 - Overall study (n=124)ITT: Week 48 - Phase I / Group A (n=13)ITT: Week 48 - Phase I / Group B (n=70)ITT: Week 48 - Phase I / Group C (n=26)ITT: Week 48 - Overall study (n=124)MITT: Week 24 - Phase I / Group A (n=13)MITT: Week 24 - Phase I / Group B (n=70)MITT: Week 24 - Phase I / Group C (n=26)MITT: Week 24 - Overall study (n=109)MITT: Week 48 - Phase I / Group A (n=13)MITT: Week 48 - Phase I / Group B (n=70)MITT: Week 48 - Phase I / Group C (n=26)MITT: Week 48 - Overall study (n=109)
Overall Study66.7100.091.4100.093.8100.092.696.294.4100.091.4100.094.5100.092.696.294.4

[back to top]

Percentage of Subjects With Normalised IGF-1 Levels (Age and Sex Adjusted)

The criterion for a subject is satisfied if he has a normalised IGF-1 level (age and sex adjusted) at week 24. (NCT00701363)
Timeframe: At week 24

InterventionPercentage of subjects (Number)
Intention-to-Treat (n=124)Modified Intention-to-Treat (n=109)
Overall Study88.7100

[back to top]

Percentage of Subjects With Normalized IGF-1 Levels (Age and Sex Adjusted), Without Any Worsening of the AcroQoL Change Score Between Inclusion and Week 48

The criterion for a subject is satisfied if he had a IGF-1 level (age and sex adjusted) without any worsening of the AcroQoL change score between Inclusion and Week 48. (NCT00701363)
Timeframe: At week 48 (End of Study)

InterventionPercentage of subjects (Number)
Phase I / Group A (n=9)Phase I / Group B (n=55)Phase I / Group C (n=21)Overall study (n=85)
Overall Study44.447.238.144.6

[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 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]

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]

"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]

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]

Percentage of Patients Having Minimum Reduction of At Least 50% or Normalization of the Mean Number of Stools

(NCT00891371)
Timeframe: Day 56

InterventionPercentage of participants (Number)
Reduction of at least 50% or normalization - YesReduction of at least 50% or normalization - No
Lanreotide Autogel 120 mg54.345.7

[back to top]

Percentage of Patients Having Minimum Reduction of 50% or Normalization (≤3 Stools/24hours) in the Mean Number of Stools (Mean of Last 7 Days)

(NCT00891371)
Timeframe: Day 28

InterventionPercentage of patients (Number)
Reduction of at least 50% or normalization - YesReduction of at least 50% or normalization - No
Lanreotide Autogel 120 mg44.455.6

[back to top]

Percent Change in Mean Number of Stools Compared to Baseline

(NCT00891371)
Timeframe: Baseline (Day 1), Day 28 and Day 56

InterventionPercent change (Mean)
Change from Baseline to Day 28Change from Baseline to Day 56
Lanreotide Autogel 120 mg-25.9-30.7

[back to top]

Change in QOL-Quality of Life {Assess Using Short Form (SF-36) and Irritable Bowel Syndrome (IBS)-QOL} Compared to Baseline

"SF36 QOL includes 1 multi-item scale measuring each of 8 health concepts. These scores are summed to produce raw scale scores for each health concept which are transformed to a 0-100 scale. The lower the score the more disability. The higher the score the less disability. There is in addition a single-item measure of Health Transition~IBS-QOL is a self-report QOL measure specific to IBS that can be used to assess impact of IBS and its treatment. This consists of 34 items,each with a 5 point response scale.Individual responses to 34 items are summed and averaged for a total score and transformed to a 0-100 scale with higher scores indicating better IBS specific QOL" (NCT00891371)
Timeframe: Baseline (Day 1), Day 21, Day 28, Day 49 and Day 56

Interventionunits on a scale (Mean)
SF36 QOL: Physical Functioning (D21), n=33SF36 QOL: Physical Functioning (D28), n=34SF36 QOL: Physical Functioning (D49), n=26SF36 QOL: Physical Functioning (D56), n=35SF36 QOL: Role Physical (D21), n=33SF36 QOL: Role Physical (D28), n=34SF36 QOL: Role Physical (D49), n=26SF36 QOL: Role Physical (D56), n=35SF36 QOL: Role Emotional (D21), n=33SF36 QOL: Role Emotional (D28), n=34SF36 QOL: Role Emotional (D49), n=25SF36 QOL: Role Emotional (D56), n=35SF36 QOL: Vitality (D21), n=32SF36 QOL: Vitality (D28), n=33SF36 QOL: Vitality (D49), n=24SF36 QOL: Vitality (D56), n=34SF36 QOL: Mental Health (D21), n=32SF36 QOL: Mental Health (D28), n=33SF36 QOL: Mental Health (D49), n=24SF36 QOL: Mental Health (D56), n=34SF36 QOL: Social Functioning (D21), n=31SF36 QOL: Social Functioning (D28), n=33SF36 QOL: Social Functioning (D49), n=24SF36 QOL: Social Functioning (D56), n=33SF36 QOL: Bodily Pain (D21), n=33SF36 QOL: Bodily Pain (D28), n=34SF36 QOL: Bodily Pain (D49), n=26SF36 QOL: Bodily Pain (D56), n=35SF36 QOL: General Health (D21), n=33SF36 QOL: General Health (D28), n=33SF36 QOL: General Health (D49), n=26SF36 QOL: General Health (D56), n=34IBS QOL: Total Score (D21), n=27IBS QOL: Total Score (D28), n=26IBS QOL: Total Score (D49), n=22IBS QOL: Total Score (D56), n=29
Lanreotide Autogel 120 mg-1.32.42.91.14.512.96.311.8-0.33.47.05.03.15.92.17.25.88.38.310.19.316.719.314.0-4.68.38.85.3-2.82.84.13.79.913.916.816.3

[back to top]

Change in Median Score of Stool Consistency (Bristol Stool Form Scale) Compared to Baseline

Each patient scored his/her stool on the Bristol Stool Form Scale: Type 1 - Separate hard lumps, like nuts (hard to pass); Type 2 - Sausage-shaped but lumpy; Type 3 - Like a sausage but with cracks on its surface; Type 4 - Like a sausage or snake, smooth and soft; Type 5 - Soft blobs with clear-cut edges (passed easily); Type 6 - Fluffy pieces with ragged edges, a mushy stool; Type 7 - Water no solid pieces, Entirely liquid (NCT00891371)
Timeframe: Baseline (day 1), day 28 and day 56

Interventionunits on a scale (Median)
Change from Baseline to Day 28Change from Baseline to Day 56
Lanreotide Autogel 120 mg0.0-0.5

[back to top]

Change From Baseline in Relative Frequency of Normalization (≤3 Stools) in Subjects

Normalization of stool frequency in subjects with refractory diarrhoea at Day 28 and Day 56 (mean of last 7 days) compared to Baseline. (NCT00891371)
Timeframe: Baseline (Day 1), Day 28 and Day 56

InterventionPercentage of days per Week (Mean)
Change from Baseline to Day 28Change from Baseline to Day 56
Lanreotide Autogel 120 mg27.533.0

[back to top]

Change From Baseline in Standardized IGF-1 Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for Extension Visits (Extension Full Analysis Set)

Change from CORE baseline at each scheduled assessment was performed for patients randomized to pasireotide arms. Change from extension baseline at each scheduled assessment was performed for patients randomized to active control arm. Standardized IGF-1 = IGF-1 value / ULN, where ULN is the upper limit of the normal range (NCT01137682)
Timeframe: CORE and extension baseline up to approximately 268 weeks

,,
Interventionmcg/L (Mean)
Week 16 extensionWeek 28 extensionWeek 40 extensionWeek 52 extensionWeek 64 extensionWeek 76 extensionWeek 88 extensionWeek 100 extensionWeek 112 extensionWeek 124 extensionWeek 136 extensionWeek 148 extensionWeek 160 extensionWeek 172 extensionWeek 184 extensionWeek 196 extensionWeek 208 extensionWeek 220 extensionWeek 232 extensionWeek 244 extensionWeek 256 extensionWeek 268 extension
Cross Over to Pasireotide Extension-0.9-0.9-1.1-1.1-1.3-1.3-1.3-1.3-1.4-1.3-1.4-1.3-1.4-1.3-1.4-1.3-1.2-1.4-1.7-1.5-1.8-1.7
Pasireotide LAR 40 mg Extension-0.8-0.9-1.1-1.1-1.3-1.3-1.3-1.4-1.5-1.4-1.4-1.4-1.4-1.5-1.3-1.4-1.4-1.4-1.4-1.3-1.3-1.4
Pasireotide LAR 60 mg Extension-1.4-1.3-1.4-1.3-1.4-1.5-1.6-1.5-1.5-1.5-1.7-1.5-1.6-1.6-1.5-1.5-1.6-1.6-1.5-1.9-1.4-1.7

[back to top]

Percentage of Patients With Mean GH < 1.0 μg/L and Normalization of IGF-1, Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)

The percentage of patients achieving mean growth hormone (GH) levels < 1.0 μg/L and normalization of sex and age-adjusted IGF-1 was calculated with two sided 95% confidence interval. All GH assessments were based on a 5-point mean growth hormone (GH) assessed from a 2-hour profile. Scheduled time points for blood sampling were pre-dose at 0, 30, 60, 90 and 120 minutes. Total insulin-like growth factor (IGF-1) levels were assessed with one pre-dose sample at the same visits as GH. Concomitant medication known to affect GH or IGF-1 levels were allowed in patients who were not biochemically controlled after at least one year treatment with pasireotide LAR monotherapy: dopamine agonists and growth hormone receptor antagonists (Extension full analysis set (NCT01137682)
Timeframe: Extension baseline up to approximately week 268

,,
InterventionParticipants (Number)
Week 16Week 28Week 40Week 52Week 64Week 76Week 88Week 100Week 112Week 124Week 136Week 148Week 160Week 172Week 184Week 196Week 208Week 220Week 232Week 244Week 256Week 268
Cross Over to Pasireotide Extension6.58.14.84.89.76.54.88.19.78.111.38.19.78.13.28.16.56.54.86.53.21.6
Pasireotide LAR 40 mg Extension10.58.810.58.88.810.57.012.314.07.03.510.58.814.07.08.88.810.510.58.88.81.8
Pasireotide LAR 60 mg Extension16.714.89.313.013.013.020.414.820.414.818.514.814.813.013.013.09.37.43.73.73.73.7

[back to top]

Percentage of Patients With Mean GH < 2.5 μg/L and Normalization of IGF-1, Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)

The percentage of patients achieving mean growth hormone (GH) levels < 2.5 μg/L and normalization of sex and age-adjusted IGF-1 was calculated with two sided 95% confidence interval. All GH assessments were based on a 5-point mean growth hormone (GH) assessed from a 2-hour profile. Scheduled time points for blood sampling were pre-dose at 0, 30, 60, 90 and 120 minutes. Total insulin-like growth factor (IGF-1) levels were assessed with one pre-dose sample at the same visits as GH. Concomitant medication known to affect GH or IGF-1 levels were allowed in patients who were not biochemically controlled after at least one year treatment with pasireotide LAR monotherapy: dopamine agonists and growth hormone receptor antagonists (extension full analysis set) (NCT01137682)
Timeframe: Extension baseline up to approximately week 268

,,
Interventionpercentage of participants (Number)
Week 16Week 28Week 40Week 52Week 64Week 76Week 88Week 100Week 112Week 124Week 136Week 148Week 160Week 172Week 184Week 196Week 208Week 220Week 232Week 244Week 256Week 268
Cross Over to Pasireotide Extension19.419.417.721.025.827.425.832.325.825.829.029.030.621.017.724.219.414.514.511.33.21.6
Pasireotide LAR 40 mg Extension19.317.521.121.122.821.124.624.624.621.115.821.119.322.817.515.817.521.114.014.010.55.3
Pasireotide LAR 60 mg Extension25.925.927.829.620.429.631.524.125.924.124.120.420.420.420.420.418.511.114.87.47.45.6

[back to top]

Percentage of Patients With Mean GH <1.0 μg/L Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)

The percentage of patients achieving mean growth hormone (GH) levels < 1.0 μg/L was calculated with two sided 95% confidence interval. All GH assessments were based on a 5-point mean growth hormone (GH) assessed from a 2-hour profile. Scheduled time points for blood sampling were pre-dose at 0, 30, 60, 90 and 120 minutes. Concomitant medication known to affect GH levels were allowed in patients who were not biochemically controlled after at least one year treatment with pasireotide LAR monotherapy: dopamine agonists and growth hormone receptor antagonists (Extension full analysis set) (NCT01137682)
Timeframe: Extension baseline up to approximately week 268

,,
Interventionpercentage of participants (Number)
Week 16Week 28Week 40Week 52Week 64Week 76Week 88Week 100Week 112Week 124Week 136Week 148Week 160Week 172Week 184Week 196Week 208Week 220Week 232Week 244Week 256Week 268
Cross Over to Pasireotide Extension8.19.74.89.711.311.311.317.716.114.514.514.514.514.56.512.99.79.78.18.14.81.6
Pasireotide LAR 40 mg Extension14.014.014.012.315.812.315.817.517.512.38.814.017.517.517.515.817.515.815.812.312.33.5
Pasireotide LAR 60 mg Extension27.822.213.022.218.522.222.220.425.916.725.916.718.514.818.513.013.011.13.75.63.73.7

[back to top]

Summary of Pasireotide Trough Concentrations in Acromegaly Patients Following Monthly i.m. Injections of Pasireotide LAR by Incident Dose From Start of Extension Phase up to Week 196 of the Extension Phase (PK Set)

"PK samples were collected for those patients treated with pasireotide LAR in the core study and who continued on pasireotide LAR in the extension phase. PK samples were collected before the injection of pasireotide LAR only at weeks 112 and 196. PK samples were also collected at weeks 48 and 132 only for all patients treated with octreotide LAR 30 mg or lanreotide ATG 120 mg in the core study who started treatment with pasireotide LAR in the extension study.~Blood samples (2.5 mL each sample) were collected to yield 1-mL plasma for analysis of pasireotide LAR oncentration." (NCT01137682)
Timeframe: Extension baseline up to approximately 196 weeks

InterventionmL (Mean)
Week 112Week 196
Pasireotide LAR 60 mg14.0614.96

[back to top]

Summary of Pasireotide Trough Concentrations in Acromegaly Patients Following Monthly i.m. Injections of Pasireotide LAR by Incident Dose From Start of Extension Phase up to Week 196 of the Extension Phase (PK Set)

"PK samples were collected for those patients treated with pasireotide LAR in the core study and who continued on pasireotide LAR in the extension phase. PK samples were collected before the injection of pasireotide LAR only at weeks 112 and 196. PK samples were also collected at weeks 48 and 132 only for all patients treated with octreotide LAR 30 mg or lanreotide ATG 120 mg in the core study who started treatment with pasireotide LAR in the extension study.~Blood samples (2.5 mL each sample) were collected to yield 1-mL plasma for analysis of pasireotide LAR oncentration." (NCT01137682)
Timeframe: Extension baseline up to approximately 196 weeks

InterventionmL (Mean)
Week 48Week 112Week 132Week 196
Pasireotide LAR 40 mg5.708.669.2810.32

[back to top]

Percentage of Participants With Normalization of Sex- and Age-adjusted IGF-1treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set).

The percentage of patients achieving normalization of sex and age-adjusted IGF-1 was calculated with two sided 95% confidence interval. Total insulin-like growth factor (IGF-1) levels were assessed with one pre-dose sample at the same visits as GH. Concomitant medication known to affect IGF-1 levels were allowed in patients who were not biochemically controlled after at least one year treatment with pasireotide LAR monotherapy: dopamine agonists and growth hormone receptor antagonists (Extension full analysis set) (NCT01137682)
Timeframe: Extension baseline up to approximately week 268

,,
Interventionpercentage of participants (Number)
Week 16Week 28Week 40Week 52Week 64Week 76Week 88Week 100Week 112Week 124Week 136Week 148Week 160Week 172Week 184Week 196Week 208Week 220Week 232Week 244Week 256Week 268
Cross Over to Pasireotide Extension25.822.624.225.829.029.025.832.330.629.037.133.933.922.622.627.421.019.417.714.56.51.6
Pasireotide LAR 40 mg Extension33.329.836.828.133.326.328.131.631.624.621.126.324.626.319.324.622.822.814.014.012.35.3
Pasireotide LAR 60 mg Extension29.033.337.033.327.835.235.229.627.831.525.922.225.922.222.222.222.211.114.89.37.45.6

[back to top]

Change From Baseline in Mean GH Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for Extension Visits (Extension Full Analysis Set)

Change from CORE baseline at each scheduled assessment was performed for patients randomized to pasireotide arms. Change from extension baseline at each scheduled assessment was performed for patients randomized to active control arm. (NCT01137682)
Timeframe: CORE and extension baseline up to approximately 268 weeks

,,
Interventionmcg/L (Mean)
Week 16 - extensionWeek 28 - extensionWeek 40 - extensionWeek 52 - extensionWeek 64 - extensionWeek 76 - extensionWeek 88 - extensionWeek 100 - extensionWeek 112 - extensionWeek 124 - extensionWeek 136 - extensionWeek 148 - extensionWeek 160 - extensionWeek 172 - extensionWeek 184 - extensionWeek 196 - extensionWeek 208 - extension (n=22,20,23)Week 220 - extensionWeek 232 - extensionWeek 244 - extensionWeek 256 - extensionWeek 268 - extension
Cross Over to Pasireotide Extension-8.4-3.0-11.2-2.5-15.7-3.5-3.6-3.8-3.9-4.0-4.0-3.8-3.2-3.0-3.3-3.4-3.5-3.8-4.2-4.2-5.0-3.7
Pasireotide LAR 40 mg Extension-7.9-9.0-9.7-10.7-11.3-5.5-5.7-5.6-5.8-5.5-6.0-6.3-5.5-6.3-6.3-7.1-7.0-7.1-5.7-6.0-6.4-3.6
Pasireotide LAR 60 mg Extension-6.9-4.5-5.8-7.1-7.9-7.2-6.2-5.3-4.4-6.0-5.36.1-4.9-5.9-5.8-6.5-6.2-7.1-6.8-2.4-4.9-2.5

[back to top]

Change From Baseline in Mean GH Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for CORE Visits (Extension Full Analysis Set)

(NCT01137682)
Timeframe: CORE baseline up to approximately 24 weeks

,
Interventionmcg/L (Mean)
Week 12 - COREWeek 24 - CORE
Pasireotide LAR 40 mg-0.8-0.6
Pasireotide LAR 60 mg-6.4-7.2

[back to top]

Percentage of Patients With Mean GH < 2.5 μg/L Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)

The percentage of patients achieving mean growth hormone (GH) levels < 2.5 μg/L was calculated with two sided 95% confidence interval. All GH assessments were based on a 5-point mean growth hormone (GH) assessed from a 2-hour profile. Scheduled time points for blood sampling were pre-dose at 0, 30, 60, 90 and 120 minutes. Concomitant medication known to affect GH levels were allowed in patients who were not biochemically controlled after at least one year treatment with pasireotide LAR monotherapy: dopamine agonists and growth hormone receptor antagonists (Extension full analysis set) (NCT01137682)
Timeframe: Extension baseline up to approximately week 268

,,
Interventionpercentage of participants (Number)
Week 16Week 28Week 40Week 52Week 64Week 76Week 88Week 100Week 112Week 124Week 136Week 148Week 160Week 172Week 184Week 196Week 208Week 220Week 232Week 244Week 256Week 268
Control Arm (Octreotide or Lanreotide) Extension33.943.540.341.941.945.245.246.840.341.943.543.540.338.733.935.525.822.621.012.96.53.2
Pasireotide LAR 40 mg Extension38.640.438.638.640.433.340.440.436.840.436.840.438.640.433.329.831.629.824.619.315.87.0
Pasireotide LAR 60 mg Extension55.644.442.646.337.044.442.640.744.431.535.233.333.337.031.529.624.118.518.511.19.35.6

[back to top]

Change From Baseline in Standardized IGF-1 Values for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for CORE Visits (Extension Full Analysis Set)

Standardized IGF-1 = IGF-1 value / ULN, where ULN is the upper limit of the normal range (NCT01137682)
Timeframe: CORE baseline up to approximately 24 weeks

,
Interventionmcg/L (Mean)
CORE Week 12CORE Week 24
Pasireotide LAR 40 mg0.7-0.7
Pasireotide LAR 60 mg-1.1-1.1

[back to top]

Change From Baseline in AcroQoL Total Scores for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for CORE Visits(Extension Full Analysis Set)

Acromegaly Quality of Life questionnaire (AcroQoL) is a validated disease specific questionnaire. It contains 22 items divided into two scales: physical aspects (8 items) and psychological aspects (14 items) which is divided in two sub-scales: physical appearance and personal relationships of the patient (seven items each). The total score and sub-scores were calculated using the following formula: ((X -Y) / 4Y) x 100, X=sum of the scores for individual items (between 1 and 5 for each item), Y=number of individual items included in above sum (i.e. 22 for the total score, 8 for the physical sub-score, 14 for the psychological sub-score, 7 for the sub-score 'appearance' and 'personal relations'). The scoring of the questionnaire was performed as specified by the instrument developers. Total scores range from 0 to 100. Higher scores represent better quality of life. If more than 25% of items are not completed, results were considered invalid. (NCT01137682)
Timeframe: CORE baseline up to approximately 24 weeks

,
Interventionscores on a scale (Mean)
Week 4 COREWeek 8 COREWeek 12 COREWeek 16 COREWeek 20 COREWeek24 CORE
Pasireotide LAR 40 mg3.52.43.03.33.53.0
Pasireotide LAR 60 mg2.32.51.96.64.05.4

[back to top]

Time to First Response (Weeks) by Treatment for Patients Achieving a Reduction of Mean GH Level to < 2.5 µg/L and Normalization of IGF-1 and Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly

Time to first response is defined as the time from the date of first dose to the date of first occurrence of a reduction of mean GH < 2.5 µg/L and the normalization of IGF-1. The weeks correspond to time taken to achieve first mean GH < 2.5 µg/L and the normalization of IGF-1. (NCT01137682)
Timeframe: CORE baseline up to approximately 268 weeks

Interventionweeks (Median)
Pasireotide LAR 40 mg Extension112.3
Pasireotide LAR 60 mg Extension65.3
Cross Over to Pasireotide Extension95.1

[back to top]

Percentage of Participants With a Reduction of Mean GH Levels to < 2.5 µg/L and Normalization of Sex- and Age-adjusted IGF-1.

The primary objective of this study was to compare the percentage of patients achieving biochemical control (defined as mean GH levels <2.5 µg/L and normalization of sex- and age- adjusted IGF-1) at 24 weeks with pasireotide LAR 40 mg and pasireotide LAR 60 mg separately versus continued treatment with octreotide LAR 30 mg or lanreotide autogel (ATG) 120 mg. The primary efficacy variable is the proportion of patients with a reduction of mean GH levels to < 2.5 µg/L and normalization of sex- and age-adjusted IGF-1 at 24 weeks. (NCT01137682)
Timeframe: At 24 weeks

Interventionpercentage of participants (Number)
Pasireotide LAR 40 mg15.4
Pasireotide LAR 60 mg20.0
Control Arm (Octreotide or Lanreotide)0

[back to top]

Duration of the First Response for Patients Achieving a Reduction of Mean GH Level to < 2.5 μg/L and Normalization of IGF-1 and Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly (Extension Full Analysis Set)

n is the number of patients achieving response criteria. The weeks correspond to duration of first response (in weeks) for patients achieving biomedical control. Median and 95% CI are derived from Kaplan-Meier curves. Kaplan-Meier estimates [95% CI] at each time point are estimates of probability of response. (NCT01137682)
Timeframe: CORE baseline up to approximately 268 weeks

Interventionweeks (Median)
Pasireotide LAR 40 mg Extension29.1
Pasireotide LAR 60 mg Extension26.9
Cross Over to Pasireotide Extension24.9

[back to top]

Change From Baseline in AcroQoL Total Scores for Patients Treated With Pasireotide LAR Alone or With Concomitant Medications Used to Treat Acromegaly for Extension Visits (Extension Full Analysis Set)

Acromegaly Quality of Life questionnaire (AcroQoL) is a validated disease specific questionnaire. It contains 22 items divided into two scales: physical aspects (8 items) and psychological aspects (14 items) which is divided in two sub-scales: physical appearance and personal relationships of the patient (seven items each). The total score and sub-scores were calculated using the following formula: ((X -Y) / 4Y) x 100, X=sum of the scores for individual items (between 1 and 5 for each item), Y=number of individual items included in above sum (i.e. 22 for the total score, 8 for the physical sub-score, 14 for the psychological sub-score, 7 for the sub-score 'appearance' and 'personal relations'). The scoring of the questionnaire was performed as specified by the instrument developers. Total scores range from 0 to 100. Higher scores represent better quality of life. If more than 25% of items are not completed, results were considered invalid. (NCT01137682)
Timeframe: CORE Baseline and extension baseline up to approximately 268 weeks

,,
Interventionscores on a scale (Mean)
Week 16 extensionWeek 28 extensionWeek 40 extensionWeek 52 extensionWeek 64 extensionWeek 76 extensionWeek 88 extensionWeek 100 extensionWeek 112 extensionWeek 124 extensionWeek 136 extensionWeek 148 extensionWeek 160 extensionWeek 172 extensionWeek 184 extensionWeek 196 extensionWeek 208 extensionWeek 220 extensionWeek 232 extensionWeek 244 extensionWeek 256 extensionWeek 268 extension
Cross Over to Pasireotide Extension0.83.33.24.25.47.76.45.94.12.06.55.51.52.11.63.84.57.37.03.00.3-10.6
Pasireotide LAR 40 mg Extension4.25.63.26.15.87.74.64.34.65.87.57.67.04.85.86.11.24.82.14.56.10.6
Pasireotide LAR 60 mg Extension2.94.82.55.74.22.55.63.96.52.05.46.85.15.75.86.06.25.8-0.26.3-3.0-4.9

[back to top]

Cost Effectiveness

We evaluated cost-effectiveness of combination low-dose SRL and weekly or daily pegvisomant compared to combination high-dose SRL and weekly pegvisomant by assessing cost of therapy per month in each treatment arm among patients who achieved normal IGF-1 levels. (NCT01538966)
Timeframe: 24 weeks

InterventionUS dollars/month (Mean)
High Dose SRL + Weekly Pegvisomant14,261.33
Low Dose SRL + Daily Pegvisomant22,542.86
Low Dose SRL + Weekly Pegvisomant9,836.52

[back to top]

Percentage of Subjects Alive and Progression Free at One Year

"Subjects underwent CT or MRI scans at baseline and Week 53. Progression was assessed by investigators using RECIST v1.1. The best overall response to study treatment is the highest time point response achieved by the subject and was assessed as a complete response, partial response, stable disease, progressive disease or non evaluable. For analysis of PFS, event dates were assigned to the first time that progressive disease was noted or the date of death. In case of progressive disease followed by death, the first event was considered in the analysis. Censoring dates were defined in subjects with no progressive disease or death before end of study.~At one year (end of study), the mean percentage of subjects who were alive and progression free, as calculated using the Kaplan-Meier method, is reported by CTC presence and overall." (NCT02075606)
Timeframe: From baseline up to Week 53.

Interventionpercentage of subjects (Mean)
CTC Presence at Baseline69.00
No CTC Presence at Baseline67.75
Lanreotide Autogel66.43

[back to top]

Assessment of Clinical Symptomatic Response

"This endpoint was assessed using 2 efficacy variables:~CTCs, enumerated at baseline and Weeks 5, 17, 25, 53~Clinical symptomatic response, assessed by the use of symptom reporting~Subjects recorded 24-hour symptom frequency and severity for 7 days prior to first treatment (baseline), throughout the study, and up to 28 days following final drug administration. Symptoms were recorded by answering predetermined questions on the interactive voice response system (IVRS).~Subjects were considered to have a clinical symptomatic response between baseline and last study visit if any 1 of the following criteria were fulfilled: the average number of episodes of diarrhoea decreased by at least 50%, the average number of episodes of flushing decreased by at least 50%, the mode severity of flushing decreased by at least 1 level. Clinical symptomatic response was assessed as a qualitative variable (Yes/No) and reported according to CTC presence at baseline and overall." (NCT02075606)
Timeframe: From baseline up to Week 53.

,,
InterventionPercentage of Subjects (Number)
Clinical Symptomatic Response = Yes(2)Clinical Symptomatic Response = No(3)
CTC Presence at Baseline77.822.2
Lanreotide Autogel87.512.5
No CTC Presence at Baseline95.54.5

[back to top]

Mean Change From Baseline in Number of Episodes of Diarrhoea and Flushing

"The effect of lanreotide Autogel on the symptoms of diarrhoea and flushing in subjects was assessed through subject reporting of symptoms every 24-hours for the 7 days prior to treatment (baseline), for the first 16 weeks and on days 11 to 17 after each subsequent injection interval. After the final study drug injection at Week 49, subjects provided 24-hour symptom frequency on days 11 to 28 (up to Week 53). Symptom frequency was recorded by answering predetermined questions on the IVRS.~Mean change from baseline in frequency (number of episodes) of diarrhoea and flushing are described at Visit 2 (average number of episodes in Week 1) and at Visit 14 (average number of episodes over days 11 to 17 after Week 49 injection and over days 11 to 28 after Week 49 injection) by CTC presence at baseline and overall. A negative change indicates an improvement in symptoms from baseline." (NCT02075606)
Timeframe: From baseline up to Week 53.

Interventionnumber of episodes (Mean)
Diarrhoea: Visit 2 (daily)Flushing: Visit 2 (daily)
Missing CTC Status at Baseline0.380.00

[back to top]

Mean Change From Baseline in Number of Episodes of Diarrhoea and Flushing

"The effect of lanreotide Autogel on the symptoms of diarrhoea and flushing in subjects was assessed through subject reporting of symptoms every 24-hours for the 7 days prior to treatment (baseline), for the first 16 weeks and on days 11 to 17 after each subsequent injection interval. After the final study drug injection at Week 49, subjects provided 24-hour symptom frequency on days 11 to 28 (up to Week 53). Symptom frequency was recorded by answering predetermined questions on the IVRS.~Mean change from baseline in frequency (number of episodes) of diarrhoea and flushing are described at Visit 2 (average number of episodes in Week 1) and at Visit 14 (average number of episodes over days 11 to 17 after Week 49 injection and over days 11 to 28 after Week 49 injection) by CTC presence at baseline and overall. A negative change indicates an improvement in symptoms from baseline." (NCT02075606)
Timeframe: From baseline up to Week 53.

,,
Interventionnumber of episodes (Mean)
Diarrhoea: Visit 2 (daily)Diarrhoea: Visit 14 (days 11-17)Diarrhoea: Visit 14 (days 11-28)Flushing: Visit 2 (daily)Flushing: Visit 14 (days 11-17)Flushing: Visit 14 (days 11-28)
CTC Presence at Baseline-0.66-1.91-2.15-1.76-3.37-3.49
Lanreotide Autogel-0.42-1.18-1.30-1.43-2.88-2.79
No CTC Presence at Baseline-0.27-0.64-0.63-1.25-2.51-2.23

[back to top]

Mode Symptom Severity of Episodes of Flushing

The effect of lanreotide Autogel on the mode severity of flushing was assessed through subject reporting of symptoms every 24-hours for the 7 days prior to treatment (baseline), for the first 16 weeks and on days 11 to 17 after each subsequent injection interval until Week 49. After the final study drug injection at Week 49, subjects provided 24-hour symptom severity on days 11 to 28 (up to Week 53). Symptom severity was recorded by answering predetermined questions on the IVRS using a three-point system (mild, moderate or severe). The mode (most frequent) intensity of flushing are reported at baseline and at Visit 14 (average number of episodes over days 11 to 17 after Week 49 injection and over days 11 to 28 after Week 49 injection). Percentages of subjects in each severity category are based on the number of subjects in the analysis set with available responses. Data is presented according to CTC presence at baseline and overall. (NCT02075606)
Timeframe: From baseline up to Week 53.

,,
Interventionpercentage of subjects (Number)
No flushing: BaselineMild: BaselineModerate: BaselineSevere: BaselineNo flushing: Visit 14 (days 11-17)Mild: Visit 14 (days 11-17)Moderate: Visit 14 (days 11-17)Severe: Visit 14 (days 11-17)No flushing: Visit 14 (days 11-28)Mild: Visit 14 (days 11-28)Moderate: Visit 14 (days 11-28)Severe: Visit 14 (days 11-28)
CTC Presence at Baseline22.727.345.54.537.543.818.80.013.360.026.70.0
Lanreotide Autogel14.038.046.02.032.445.921.60.023.555.917.62.9
No CTC Presence at Baseline0.050.050.00.028.647.623.80.031.652.610.55.3

[back to top]

Mode Symptom Severity of Episodes of Flushing

The effect of lanreotide Autogel on the mode severity of flushing was assessed through subject reporting of symptoms every 24-hours for the 7 days prior to treatment (baseline), for the first 16 weeks and on days 11 to 17 after each subsequent injection interval until Week 49. After the final study drug injection at Week 49, subjects provided 24-hour symptom severity on days 11 to 28 (up to Week 53). Symptom severity was recorded by answering predetermined questions on the IVRS using a three-point system (mild, moderate or severe). The mode (most frequent) intensity of flushing are reported at baseline and at Visit 14 (average number of episodes over days 11 to 17 after Week 49 injection and over days 11 to 28 after Week 49 injection). Percentages of subjects in each severity category are based on the number of subjects in the analysis set with available responses. Data is presented according to CTC presence at baseline and overall. (NCT02075606)
Timeframe: From baseline up to Week 53.

Interventionpercentage of subjects (Number)
No flushing: BaselineMild: BaselineModerate: BaselineSevere: Baseline
Missing CTC Status at Baseline1000.00.00.0

[back to top]

Percentage of Subjects With Time Point Responses According to Response Evaluation Criteria in Solid Tumours (RECIST) Assessments at Weeks 25 and 53

"Subjects underwent Computed Tomography (CT) or Magnetic Resonance Imaging (MRI) scans at baseline, Visit 8 (Week 25) and Visit 15 (Week 53). Progression was assessed by investigators using RECIST v1.1, and classified as a complete response, partial response, stable disease, progressive disease or non evaluable.~The time point responses at Week 25 and Week 53 were analysed by CTC presence at baseline and overall. The percentage of subjects within each response category are presented. Percentages are based on the number of subjects in the concerned population with available responses." (NCT02075606)
Timeframe: Week 25 and Week 53.

,,
Interventionpercentage of subjects (Number)
Week 25: Complete ResponseWeek 25: Partial ResponseWeek 25: Stable DiseaseWeek 25: Progressive DiseaseWeek 25: Non evaluableWeek 53: Complete ResponseWeek 53: Partial ResponseWeek 53: Stable DiseaseWeek 53: Progressive DiseaseWeek 53: Non evaluable
CTC Presence at Baseline0.018.272.79.10.00.06.766.726.70.0
Lanreotide Autogel0.013.073.913.00.00.05.464.929.70.0
No CTC Presence at Baseline0.08.375.016.70.00.04.563.631.80.0

[back to top]

QoL Questionnaire: EORTC QLQ-G.I.NET21

The effect of lanreotide Autogel treatment on QoL was assessed using the EORTC QLQ-G.I.NET21 at baseline, Weeks 13 (Visit 5), 25 (Visit 8) and 53 (Visit 15/end of study). The QLQ-G.I.NET21 questionnaire contained 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Each individual subscore was transformed to range from 0 to 100. Higher scores indicate worse symptoms or more problems. The mean change from baseline at each time point is reported for each of the category subscores. (NCT02075606)
Timeframe: From baseline up to Week 53.

InterventionUnits on a scale (Mean)
Endocrine symptoms: Visit 5Endocrine symptoms: Visit 8Endocrine symptoms: End of studyGastrointestinal symptoms: Visit 5Gastrointestinal symptoms: Visit 8Gastrointestinal symptoms: End of studyTreatment symptoms: Visit 5Treatment symptoms: Visit 8Treatment symptoms: End of studySocial function: Visit 5Social function: Visit 8Social function: End of studyDisease related worries: Visit 5Disease related worries: Visit 8Disease related worries: End of studyMuscle/Bone pain: Visit 5Muscle/Bone pain: Visit 8Muscle/Bone pain: End of studySexual function: Visit 5Sexual function: Visit 8Sexual function: End of studyInformation/communication function: Visit 5Information/communication function: Visit 8Information/communication function: End of studyBody image: Visit 5Body image: Visit 8Body image: End of study
Lanreotide Autogel-15.4-17.0-16.02.11.21.01.27.612.0-11.3-6.5-4.5-14.1-15.9-12.2-7.9-6.5-11.8-5.6-8.9-13.3-8.5-3.7-5.60.91.9-1.0

[back to top]

Quality of Life (QoL) Questionnaire: European Organisation for Research and Treatment of Cancer (EORTC) QoL Questionnaire (QLQ)-C30

"The effect of lanreotide Autogel treatment on QoL was assessed using the EORTC QLQ-C30 at baseline, Weeks 13 (Visit 5), 25 (Visit 8) and 53 (Visit 15/end of study). The 30 item scale is divided into 9 multi item scales (including 5 functional scales, 1 global health status/QoL scale and 3 general symptom scales) and 6 single items. Possible answers to the first 28 items (all items except the 2 concerning global quality of life) go from 1 (Not at all) to 4 (Very much). The answers for the 2 last questions (Q29- 30) go from 1 (Very poor) to 7 (Excellent). All of the scales and single-item measures range in score from 0 to 100. For multi-item scales, the raw score will be calculated by the addition of item responses divided by the number of items. Higher scores for global health and functional domains indicate a better QoL, while higher symptom scores indicate worse symptoms.~The mean change from baseline at each time point is reported for each of the category subscores." (NCT02075606)
Timeframe: From baseline up to Week 53.

InterventionUnits on a scale (Mean)
Physical functioning: Visit 5Physical functioning: Visit 8Physical functioning: End of studyRole functioning: Visit 5Role functioning: Visit 8Role functioning: End of studyEmotional functioning: Visit 5Emotional functioning: Visit 8Emotional functioning: End of studyCognitive functioning: Visit 5Cognitive functioning: Visit 8Cognitive functioning: End of studySocial functioning: Visit 5Social functioning: Visit 8Social functioning: End of studyGlobal QoL: Visit 5Global QoL: Visit 8Global QoL: End of studyFatigue: Visit 5Fatigue: Visit 8Fatigue: End of studyNausea and vomiting: Visit 5Nausea and vomiting: Visit 8Nausea and vomiting: End of studyPain: Visit 5Pain: Visit 8Pain: End of studyDyspnoea: Visit 5Dyspnoea: Visit 8Dyspnoea: End of studyInsomnia: Visit 5Insomnia: Visit 8Insomnia: End of studyAppetite loss: Visit 5Appetite loss: Visit 8Appetite loss: End of studyConstipation: Visit 5Constipation: Visit 8Constipation: End of studyDiarrhoea: Visit 5Diarrhoea: Visit 8Diarrhoea: End of studyFinancial difficulties: Visit 5Financial difficulties: Visit 8Financial difficulties: End of study
Lanreotide Autogel1.22.01.91.33.2-1.46.14.31.1-0.91.5-2.410.04.53.912.57.44.3-4.4-6.3-4.2-4.2-1.4-0.9-7.9-2.31.8-3.51.0-4.8-6.3-5.7-2.9-0.9-6.5-0.01.9-1.01.9-18.5-12.7-10.8-6.7-4.0-1.0

[back to top]

The Number of Subjects With a Symptomatic Response After 12 Months - Maintenance Phase

"Symptomatic response was evaluated as absolute change from baseline in the number of episodes of the lead symptoms (i.e. diarrhoea and flushing) using the mean of the last 3 days before the visit, at each visit, as compared to baseline.~Symptomatic responses were categorised as: Reduction, Increase or Stability of occurrences of diarrhoea / Reduction, Increase or Stability of occurrences of flushing.~The number of subjects in each response category at week 48 (end of study) is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Diarrhoea - ReductionDiarrhoea - IncreaseDiarrhoea - StabilityDiarrhoea - MissingFlushing - ReductionFlushing - IncreaseFlushing - StabilityFlushing - Missing
Maintenance Phase - Functioning NET, Lanreotide41332333

[back to top]

The Number of Subjects With a Symptomatic Response After 6 Months

"Symptomatic response was evaluated as absolute change from baseline in the number of episodes of the lead symptoms (i.e. diarrhoea and flushing) using the mean of the last 3 days before the visit, at each visit, as compared to baseline.~Symptomatic responses were categorised as: Reduction, Increase or Stability of occurrences of diarrhoea / Reduction, Increase or Stability of occurrences of flushing.~The number of subjects in each response category at week 24 (end of the combination phase) is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Diarrhoea - ReductionDiarrhoea - IncreaseDiarrhoea - StabilityDiarrhoea - MissingFlushing - ReductionFlushing - IncreaseFlushing - StabilityFlushing - Missing
Combination Phase - Functioning NET42564436

[back to top]

The Number of Subjects With a Biochemical Response Using 5-Hydroxy-Indol-Amino-Acid (HIAA) Levels After 6 Months

"Urine samples for 5-HIAA urinary tumour marker analysis were taken at at baseline, weeks 12, 24and early withdrawal.~Biochemical response based on 5-HIAA levels was categorised as: Response (5-HIAA reduction compared to baseline) or Progression (5-HIAA increase compared to baseline).~The number of subjects in each response category at each time point in the combination phase is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Week 12 - ProgressionWeek 12 - ResponseWeek 12 - Not evaluableWeek 12 - MissingWeek 24 - ProgressionWeek 24 - ResponseWeek 24 - Not evaluableWeek 24 - MissingEarly Withdrawal - ProgressionEarly Withdrawal - ResponseEarly Withdrawal - Not EvaluableEarly Withdrawal - Missing
Combination Phase - Functioning NET461663130107

[back to top]

DCR After 12 Months

"All tumour assessments were performed using the RECIST criteria (1.1). CT-scan or MRI could be used for as method of tumour measurement and the same method of tumour measurement was used throughout the study for each subject. CT scans/MRI were performed at screening or baseline visit then at baseline, weeks 12, 24, 36, 48 (end of study) and at study withdrawal or at anytime during the study in the case of any clinical or biological signs of tumour progression.~The DCR was defined as the proportion of subjects with a response of CR, PR or SD after 6 months combination treatment followed by either 6 months of lanreotide ATG 120 mg maintenance treatment or no treatment. The DCR was described in the ITT population along with its 95% CI and was compared to 45% with an exact binomial proportion test. The LOCF method was used to replace missing assessments at the end of the maintenance phase." (NCT02231762)
Timeframe: 12 months

Interventionpercentage of subjects (Number)
Maintenance Phase - Functioning NET, Lanreotide54.5
Maintenance Phase - Non-functioning NET, Lanreotide71.4
Maintenance Phase - Non-functioning NET, No Treatment41.7

[back to top]

Disease Control Rate (DCR) After 6 Months

"All tumour assessments were performed using the Response Evaluation Criteria In Solid Tumours (RECIST) criteria (1.1). Computer Tomography (CT-scan) or Magnetic Resonance Imaging (MRI) could be used for as method of tumour measurement and the same method of tumour measurement was used throughout the study for each subject. CT scans/MRI were performed at screening or baseline visit then at weeks 12, 24 and at early withdrawal or at anytime during the study in the case of any clinical or biological signs of tumour progression.~The DCR was defined as the proportion of subjects with a response of CR, PR or SD after 6 months of combination treatment and was described in the ITT population along with its 95% Confidence Interval (CI) and was compared to 45% with an exact binomial proportion test. The Last Observation Carried Forward (LOCF) method was used to replace missing assessments at the end of the combination phase." (NCT02231762)
Timeframe: 6 months

Interventionpercentage of subjects (Number)
Combination Phase73.5

[back to top]

Duration of Response (DoR) Within 12 Months

"The DoR is an estimation of the time from first documented objective response (CR or PR) to the first date of progressive disease (PD) or death due to disease progression for subjects who experienced an objective response within the first 12 months of treatment (combination and maintenance phases).~The Kaplan-Meier method was used to estimate the median DoR and its 95% CI for subjects in the ITT population who had an objective response." (NCT02231762)
Timeframe: 12 months

Interventionmonths (Median)
Intention-to-treat (ITT) PopulationNA

[back to top]

Progression-Free Survival (PFS) Within 12 Months

"PFS was defined as the time from the date of treatment start to the date of the first documented disease progression or death due to any cause within the first 12 months of treatment. If a subject had not progressed or died after 12 months of treatment or when any further anti-neoplastic therapy was received, PFS was censored at the time of the last tumour assessment before the analysis cut-off date or the anti-neoplastic therapy date.~A Kaplan-Meier estimate of the PFS was calculated to determine the number of subjects at risk. Median PFS time (50% of subjects who would not progress or die) of the ITT population is presented along with 95 % CI." (NCT02231762)
Timeframe: 12 months

Interventionmonths (Median)
Intention-to-treat (ITT) Population11.1

[back to top]

Time To Response (TtR) Within 12 Months

"TtR was defined as the time from the date of treatment start to the date of the first documented objective response (CR or PR) within the first 12 months of treatment (combination and maintenance phases). A Kaplan Meier estimate of the TtR survival function was constructed.~The Kaplan-Meier method was used to estimate the median TtR and its 95% CI for subjects in the ITT population (50% of subjects were expected to have a CR or PR at this time)." (NCT02231762)
Timeframe: 12 months

Interventionmonths (Median)
Intention-to-treat (ITT) PopulationNA

[back to top]

DCR by O6-methylguanine-DNA Methyl-transferase (MGMT) Expression and Methylation and Somatostatin Receptor (SSTR) Expression After 6 Months

"In all subjects whose tumour tissue was available, MGMT expression/methylation and SSTR expression was analysed. After 6 months, the DCR (SD+PR+CR) by MGMT methylation and expression and by SSTR 2a and SSTR 5 expression was evaluated.~DCR in response to MGMT methylation and expression results are presented. SSTR 2a and SSTR 5 expression is categorised as: No Receptors, Cytoplasmatic Expression (CE), Focal Expression (FE), Complete Circumferent Membrane Expression (CCME).~The DCR was defined as the proportion of subjects with a response of CR, PR or SD after 6 months of combination treatment within each methylation/expression category. The DCR was described in the ITT population along with its 95% CI and was compared to 45% with an exact binomial proportion test." (NCT02231762)
Timeframe: 6 months

Interventionpercentage of subjects (Number)
MGMT MethylationMGMT No methylationMGMT ExpressionMGMT No expressionSSTR 2a FESSTR 2a CCMESSTR 5 - No ReceptorsSSTR 5 CESSTR 5 FE
Combination Phase100.084.690.970.086.772.775.0100.081.8

[back to top]

EORTC QoL Questionnaire QLQ-C30: Mean Change From Baseline at 12 Months

Subjects were instructed to complete QLQ-C30 questionnaire at baseline, weeks 12, 24, 36, 48 (end of study) or at early withdrawal. The first 28 questions used a 4-point scale (1=not at all, 2=a little, 3=quite a bit, 4=very much) for evaluating 5 functional scales (physical, role, emotional, cognitive, social), 3 symptom scales (fatigue, nausea/vomiting, pain) & 6 other single items. The last 2 questions represented subject's assessment of overall health & quality of life, coded on a 7-point scale (1=very poor to 7=excellent). The mean change from baseline at week 48 (end of study) is presented for global health status (scoring of questions 29 & 30) and 5 functional scales, 3 symptom scales and other single items (scoring of questions 1 to 28). Each individual subscore was transformed to range from 0 to 100. A higher score represents a higher level response. Thus, a better QoL/a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 12 months

,,
Interventionunits on a scale (Mean)
Global health statusPhysical functioningRole functioningEmotional functioningCognitive functioningSocial functioningFatigueNausea and vomitingPainDyspnoeaInsomniaAppetite lossConstipationDiarrhoeaFinancial difficulties
Maintenance Phase - Functioning NET, Lanreotide-11.78.03.315.03.313.3-22.2-10.0-13.3-8.3-13.30.020.0-40.06.7
Maintenance Phase - Non-functioning NET, Lanreotide-3.1-12.5-2.1-6.2-2.1-22.9-9.74.24.24.2-8.30.0-4.28.34.2
Maintenance Phase - Non-functioning NET, No Treatment-7.1-11.4-7.1-6.02.4-4.84.82.49.59.516.714.3-4.80.09.5

[back to top]

European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life (QoL) Core 30 Questionnaire (QLQ-C30): Mean Change From Baseline at 6 Months

Subjects were instructed to complete the QLQ-C30 questionnaire at baseline, weeks 12, 24 or at early withdrawal. The first 28 questions used a 4-point scale (1=not at all, 2=a little, 3=quite a bit, 4=very much) for evaluating 5 functional scales (physical, role, emotional, cognitive, social), 3 symptom scales (fatigue, nausea/vomiting, pain) & 6 other single items. The last 2 questions represented subject's assessment of overall health & quality of life, coded on a 7-point scale (1=very poor to 7=excellent). The mean change from baseline at week 24 (end of the combination phase) is presented for global health status (scoring of questions 29 & 30) and 5 functional scales, 3 symptom scales and other single items (scoring of questions 1 to 28). Each individual subscore was transformed to range from 0 to 100. A higher score represents a higher level response. Thus, a better QoL/a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 6 months

Interventionunits on a scale (Mean)
Global health statusPhysical functioningRole functioningEmotional functioningCognitive functioningSocial functioningFatigueNausea and vomitingPainDyspnoeaInsomniaAppetite lossConstipationDiarrhoeaFinancial difficulties
Combination Phase-4.9-9.6-8.3-4.7-5.9-11.86.96.9-1.012.70.02.06.9-3.92.9

[back to top]

Pharmacokinetic (PK) Results: Lanreotide ATG 120 mg Serum Concentrations Within 12 Months

"Lanreotide ATG levels were measured in a subset of subjects to evaluate if temozolomide co-treatment had an impact on lanreotide serum concentration over a 12 month period.~Blood samples were collected for the determination of lanreotide ATG in serum at baseline, weeks 4, 12, 24 and 48 (end of study).~The concentrations of lanreotide ATG in serum were determined by a validated radioimmunoassay analysis method with a lower limit of quantitation of 0.08 nanograms [ng]/mL).~Serum concentrations of lanreotide ATG at each of the time points in the combination and maintenance phase are presented. Only subjects with data available for analysis are presented." (NCT02231762)
Timeframe: Baseline (week 1) and weeks 4, 12, 24 and 48

Interventionng/mL (Mean)
BaselineWeek 4Week 12Week 24Week 48
PK Subset0.442.455.065.833.68

[back to top]

QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months

Subjects were instructed to complete the QLQ-GI.NET21 questionnaire at baseline, weeks 12, 24, 36, 48 (end of study) or at early withdrawal. It contained 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Answers were converted into grading scale, with values between 0 and 100. Each individual subscore was transformed to range from 0 to 100. The mean change from baseline at week 48 (end of study) is presented with a higher score representing a higher level response. Thus, a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 12 months

,
Interventionunits on a scale (Mean)
Endocrine symptomsG.I. symptomsTreatment related symptomsSocial functionDisease related worriesMuscle/bone pain symptomsBody imageWeight gainInformation/communication functionSexual function
Maintenance Phase - Functioning NET, Lanreotide-13.3-4.00.0-6.7-6.7-6.70.0-20.00.00.0
Maintenance Phase - Non-functioning NET, Lanreotide-5.60.0-11.19.71.44.24.29.50.00.0

[back to top]

QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months

Subjects were instructed to complete the QLQ-GI.NET21 questionnaire at baseline, weeks 12, 24, 36, 48 (end of study) or at early withdrawal. It contained 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Answers were converted into grading scale, with values between 0 and 100. Each individual subscore was transformed to range from 0 to 100. The mean change from baseline at week 48 (end of study) is presented with a higher score representing a higher level response. Thus, a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 12 months

Interventionunits on a scale (Mean)
Endocrine symptomsG.I. symptomsSocial functionDisease related worriesMuscle/bone pain symptomsBody imageWeight gainInformation/communication functionSexual function
Maintenance Phase - Non-functioning NET, No Treatment1.66.7-6.3-3.24.84.8-9.5-4.80.0

[back to top]

Quality of Life Gastrointestinal Neuroendocrine Tumour 21 Questionnaire (QLQ-GI.NET21): Mean Change From Baseline at 6 Months

Subjects were instructed to complete the QLQ-GI.NET21 questionnaire at baseline, weeks 12, 24 or at early withdrawal. It contained 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Each individual subscore was transformed to range from 0 to 100. The mean change from baseline at week 24 (end of combination phase) is presented with a higher score representing a higher level response. Thus, a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 6 months

InterventionUnits on a scale (Mean)
Endocrine symptomsGastrointestinal (G.I.) symptomsTreatment related symptomsSocial functionDisease related worriesMuscle/bone pain symptomsBody imageWeight gainInformation/communication functionSexual function
Combination Phase-1.05.73.62.31.61.00.0-11.8-9.4-4.8

[back to top]

The Number of Subjects With a Biochemical Response Using 5-HIAA Levels After 12 Months

"Urine samples for 5-HIAA urinary tumour marker analysis were taken at baseline, weeks 12, 24, 36, 48 (end of study) and early withdrawal.~Biochemical response based on 5-HIAA levels was categorised as: Response (5-HIAA reduction compared to baseline) or Progression (5-HIAA increase compared to baseline).~The number of subjects in each response category at each time point in the maintenance phase is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Week 24 - ProgressionWeek 24 - ResponseWeek 24 - Not evaluableWeek 24 - MissingWeek 36 - ProgressionWeek 36 - ResponseWeek 36 - Not evaluableWeek 36 - MissingWeek 48 - ProgressionWeek 48 - ResponseWeek 48 - Not evaluableWeek 48 - MissingEarly Withdrawal - ProgressionEarly Withdrawal - ResponseEarly Withdrawal - Not evaluableEarly Withdrawal - Missing
Maintenance Phase - Functioning NET, Lanreotide6311330342020003

[back to top]

The Number of Subjects With a Biochemical Response Using CgA Levels After 12 Months

"Blood samples for CgA blood tumour marker analysis were taken at baseline, weeks 12, 24, 36, 48 (end of study) and at early withdrawal. The biochemical response after 12 months combination and maintenance treatment was estimated for subjects with abnormal CgA levels at baseline. Abnormal CgA levels were defined as above the upper limit of normal range (≥100 mcg/L).~Biochemical response based on CgA levels was categorised as: PR (decrease of CgA ≥50 % compared to the baseline CgA), SD (decrease < 50% or an increase ≤ 25% compared to the baseline CgA) or PD (defined as an increase ≥ 25%, compared to the baseline CgA).~The number of subjects in each response category at each time point in the maintenance phase is presented. Analysis was only carried out on subjects in the ITT population who had abnormal CgA at baseline." (NCT02231762)
Timeframe: 12 months

,,
InterventionParticipants (Count of Participants)
Week 24 - PDWeek 24 - SDWeek 24 - PRWeek 24 - MissingWeek 36 - PDWeek 36 - SDWeek 36 - PRWeek 36 - MissingWeek 48 - PDWeek 48 - SDWeek 48 - PRWeek 48 - MissingEarly Withdrawal - PDEarly Withdrawal - SDEarly Withdrawal - PREarly Withdrawal - Missing
Maintenance Phase - Functioning NET, Lanreotide2300120010101002
Maintenance Phase - Non-functioning NET, Lanreotide1431141112202010
Maintenance Phase - Non-functioning NET, No Treatment2232324023101010

[back to top]

The Number of Subjects With a Biochemical Response Using Chromogranin-A (CgA) Levels After 6 Months

"Blood samples for CgA blood tumour marker analysis were taken at baseline, weeks 12, 24 and at early withdrawal. The biochemical response after 6 months combination treatment was estimated for subjects with abnormal CgA levels at baseline. Abnormal CgA levels were defined as above the upper limit of normal range (≥100 micrograms/litre [mcg/L]).~Biochemical response based on CgA levels was categorised as: PR (decrease of CgA ≥ 50%, compared to the baseline CgA), SD (decrease < 50 % or an increase ≤25%, compared to the baseline CgA) or PD (defined as an increase ≥25 %, compared to the baseline CgA).~The number of subjects in each response category at each time point in the combination phase is presented. Analysis was only carried out on subjects in the ITT population who had abnormal CgA at baseline." (NCT02231762)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Week 12- PDWeek 12 - SDWeek 12- PRWeek 12 - MissingWeek 24 - PDWeek 24 - SDWeek 24 - PRWeek 24 - MissingEarly Withdrawal - PDEarly Withdrawal - SDEarly Withdrawal - PREarly Withdrawal - Missing
Combination Phase815101597012114

[back to top]

Median Time Between First Lanreotide Autogel® Injection and Clinical Response in Phase 1

The time for clinical response in Phase 1 (up to Day 28) was defined as the time from inclusion (Day 0) to the date of clinical response. A response was defined as occurrence of ≤ 2 vomiting episodes/day for at least 3 consecutive days at any timepoint between Day 0 and Day 28 (for patients without NGT use at baseline) or the removal of NGT for at least 3 consecutive days at any timepoint between Day 0 and Day 28 without vomiting recurrence (for patients with NGT use at baseline). The Kaplan-Meier estimate of median time to clinical response are presented. (NCT02275338)
Timeframe: From Day 0 to Day 28

Interventiondays (Median)
Lanreotide Autogel® 120 mg - All Subjects9.00

[back to top]

Percentage of Responders Before or at Day 7

The primary endpoint assessed the percentage of responding subjects before or at Day 7. A responder was defined as a subject experiencing ≤2 vomiting episodes/day during at least 3 consecutive days at any timepoint between Day 0 and Day 7 (for subjects without NGT at baseline), or as a subject in whom the NGT had been removed during at least 3 consecutive days at any timepoint between Day 0 and Day 7 without vomiting recurrence (for subjects with NGT at baseline), as recorded on diary cards which were completed every day. (NCT02275338)
Timeframe: From Day 0 to Day 7

Interventionpercentage of responders (Number)
Without NGT at BaselineWith NGT at BaselineAll Subjects
Lanreotide Autogel® 120 mg88.225.746.2

[back to top]

Median Change From Baseline in Quality of Life as Assessed by ESAS in Phase 2

Quality of Life was assessed by both subject and investigator based on the ESAS. The ESAS scale evaluates 9 symptoms common in cancer subjects: pain, tiredness, nausea, depression, anxiety, drowsiness, appetite, wellbeing and shortness of breath. The severity at the time of assessment of each symptom is rated from 0 to 10 on a numerical scale, 0 = symptom is absent and 10 = worst possible severity. Each symptom rating was interpreted independently and a total symptom distress score was calculated for both subject and investigator assessed scores as the sum of the 9 items. Median change from baseline (Day 0) in total symptom distress score, at each of the Phase 2 timepoints is presented and a positive change indicates a worsening condition. (NCT02275338)
Timeframe: Days 0, 35, 42 and 56

Interventionunits on a scale (Median)
Day 35 - assessed by subjectDay 35 - assessed by investigatorDay 42 - assessed by subjectDay 42 - assessed by investigatorDay 56 - assessed by subjectDay 56 - assessed by investigator
Lanreotide Autogel® 120 mg - All Subjects-4.0-12.0-10.5-13.5-8.0-9.0

[back to top]

Median Change From Baseline in Number of Daily Episodes of Nausea in Phase 1

"The mean number of daily episodes of nausea were calculated as the sum of episodes of nausea reported the last 3 days before the corresponding visit, divided by 3.~The median change from baseline (Day 0) at each of the Phase 1 timepoints is presented and positive change indicates a worsening condition." (NCT02275338)
Timeframe: Days 0, 7, 14 and 28

InterventionDaily episodes of nausea (Median)
At Day 7At Day 14At Day 28
Lanreotide Autogel® 120 mg - All Subjects-0.17-1.50-1.50

[back to top]

Median Change From Baseline in General Activity as Assessed by the Karnofsky Performance Status (KPS) Scale in Phase 1

The KPS scale was used to quantify subject's general well-being and activities of daily life. Subjects were classified based on their functional impairment and KPS scores range from 0 (death) to 100 (no evidence of disease). KPS scores are classified as 0-40 = unable to care for self; requires equivalent of institutional or hospital care; disease may be progressing rapidly; 50-70 = unable to work; able to live at home and care for most personal needs; varying amount of assistance needed; 80-100 = able to carry on normal activity and to work; no special care needed. Median change from baseline (Day 0) at each of the Phase 1 timepoints is presented and a negative change indicates a worsening condition. (NCT02275338)
Timeframe: Days 0, 7, 14 and 28

Interventionunits on a scale (Median)
At Day 7At Day 14At Day 28
Lanreotide Autogel® 120 mg - All Subjects0.00.010.0

[back to top]

Median Change From Baseline in Abdominal Pain Scores Assessed Using Visual Analogue Scale (VAS) in Phase 1

Abdominal pain was assessed using the VAS numeric pain distress scale. The VAS is a 100-millimetre (10-centimetre) scoring scale on which subjects marked on their perceived level of pain. Score range on VAS is from 0 to 100 where 0 = no pain and 100 = unbearable pain. Median change from baseline (Day 0) at each of the Phase 1 timepoints is presented and a positive change indicates a worsening condition. (NCT02275338)
Timeframe: Days 0, 7, 14 and 28

Interventionunits on a scale (Median)
At Day 7At Day 14At Day 28
Lanreotide Autogel® 120 mg - All Subjects-3.0-1.00.0

[back to top]

Median Change From Baseline in Quality of Life as Assessed by Edmonton Symptom Assessment System (ESAS) in Phase 1

Quality of Life was assessed by both subject and investigator based on the ESAS. The ESAS scale evaluates 9 symptoms common in cancer subjects: pain, tiredness, nausea, depression, anxiety, drowsiness, appetite, wellbeing and shortness of breath. The severity at the time of assessment of each symptom is rated from 0 to 10 on a numerical scale; 0 = symptom is absent and 10 = worst possible severity. Each symptom rating was interpreted independently and a total symptom distress score was calculated for both subject and investigator assessed scores as the sum of the 9 items. Median change from baseline (Day 0) in total symptom distress score, at each of the Phase 1 timepoints is presented and a positive change indicates a worsening condition. (NCT02275338)
Timeframe: Days 0, 7, 14 and 28

Interventionunits on a scale (Median)
Day 7 - assessed by subjectDay 7 - assessed by investigatorDay 14 - assessed by subjectDay 14 - assessed by investigatorDay 28 - assessed by subjectDay 28 - assessed by investigator
Lanreotide Autogel® 120 mg - All Subjects-3.0-4.5-2.0-7.5-5.5-5.0

[back to top]

Percentage of Responders Before or at Phase 2 Timepoints

This endpoint assessed the overall percentage of subjects continuing from Phase 1 and confirmed as a responder at the end of Phase 1, showing a continued response at Days 35, 42 and 56. A responder was defined as a subject experiencing ≤2 vomiting episodes/day during at least 3 consecutive days at any timepoint between Day 0 and Days 35, 42, 56 (for subjects without NGT at baseline), or as a subject in whom the NGT had been removed during at least 3 consecutive days at any timepoint between Day 0 and Days 35, 42, 56 without vomiting recurrence (for subjects with NGT at baseline), as recorded on diary cards which were completed every day. (NCT02275338)
Timeframe: From Day 0 to Day 56

Interventionpercentage of responders (Number)
By Day 35: Without NGT at BaselineBy Day 35: With NGT at BaselineBy Day 35: All SubjectsBy Day 42: Without NGT at BaselineBy Day 42: With NGT at BaselineBy Day 42: All SubjectsBy Day 56: Without NGT at BaselineBy Day 56: With NGT at BaselineBy Day 56: All Subjects (n=21)
Lanreotide Autogel® 120 mg - All Subjects100100100100100100100100100

[back to top]

Percentage of Responders in Phase 1

This endpoint assessed the overall percentage of responding subjects at the Phase 1 timepoints of Days 14 and 28. A responder was defined as a subject experiencing ≤ 2 vomiting episodes/day during at least 3 consecutive days at any timepoint between Day 0 and Days 14 or 28 (for subjects without NGT at baseline) or as a subject in whom the NGT has been removed, during at least 3 consecutive days without vomiting recurrence, at any timepoint between Day 0 and Days 14 and 28 (for subjects with NGT at baseline), as recorded on diary cards which were completed every day. (NCT02275338)
Timeframe: From Day 0 to Day 28

Interventionpercentage of responders (Number)
By Day 14: Without NGT at BaselineBy Day 14: With NGT at BaselineBy Day 14: All SubjectsBy Day 28: Without NGT at BaselineBy Day 28: With NGT at BaselineBy Day 28: All Subjects
Lanreotide Autogel® 120 mg - All Subjects88.245.759.688.254.365.4

[back to top]

Progression-Free Survival

"The progression-free survival is the time from inclusion to the first radiological progression or death (all causes). For patients alive without progression date of last news will be considered.~Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as a 20% increase in the sum of the longest diameter of target lesions compared the little sum of diameters observed durin the study (NADIR), or a measurable increase in a nontarget lesion, or the appearance of new lesions" (NCT02288377)
Timeframe: up to 2 years

InterventionMonths (Median)
Placebo7.6
Lanreotide19.4

[back to top]

Overall Survival

Overall survival considered all deaths, and time was calculated from randomisation to death. (NCT02288377)
Timeframe: 2 years after the end of the treatment

InterventionPercentage of patients alive (Number)
Placebo86.1
Lanreotide95.0

[back to top]

Proportion of Patients Alive and Progression-free at 6 Months

The primary endpoint for this phase II study was the proportion of pts alive and progression-free at 6 months after randomisation, evaluated according to the results of the imaging assessment done by the investigator in line with RECIST 1.1 criteria. (NCT02288377)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Placebo13
Lanreotide19

[back to top]

Mean Change From Baseline in Number of Daily Vomiting Episodes; ITT Population

Vomiting episodes were recorded on the Patient Diary daily until the end of study (Day 28), by the patient or filled in by the nurse/caregiver in case of patient's physical inability. Mean change from baseline in number of daily vomiting episodes is presented for the ITT population. (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation) and Days 7, 14 and 28.

,
InterventionEpisodes (daily) (Mean)
Change at Day 7Change at Day 14Change at Day 28
Standard Care5.710.616.5
Standard Care + Lanreotide Autogel6.010.414.1

[back to top]

Mean Change From Baseline in Performing General Activity (Karnofsky Performance Status [KPS]); ITT Population

"The KPS allows patients to be classified as to their functional impairment and was used to assess general activity. KPS scores range from 0 (dead) to 100 (normal/no disease) and are classified as 0-40 = unable to care for self; 50-70 = unable to work; 80-100 = able to work. The lower the KPS score, the worse the survival for most serious illnesses. Scores were recorded on the patient's medical file at each study visit (Days 1, 7, 14 and 28).~Mean change from baseline of KPS score at Days 7, 14 and 28 is presented for the ITT population (all randomised patients); a negative change indicates a worsening condition." (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation) and Days 7, 14 and 28.

,
InterventionScores on a scale (Mean)
Change at Day 7Change at Day 14Change at Day 28
Standard Care-2.8-5.6-5.0
Standard Care + Lanreotide Autogel-3.7-5.4-7.1

[back to top]

Mean Change From Baseline in Single ESAS Items Symptom Scores; ITT Population

"Quality of Life was assessed using ESAS, evaluating 9 common symptoms in cancer patients: pain, activity, nausea, depression, anxiety, drowsiness, appetite, well-being and shortness of breath. Symptom severity is rated 0-10 on a numerical scale (0=symptom absent; 10=worst severity). Low scores indicate good quality of life; high scores indicate strong discomfort. Questionnaire assessments by the patient or by nurse/caregiver in case of patient's physical inability.~Mean change from baseline of each individual ESAS item score at Days 7, 14 and 28 is presented; a positive change indicates a worsening condition." (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation) and Days 7, 14 and 28.

,
InterventionScores on a scale (Mean)
Change at Day 7: PainChange at Day 14: PainChange at Day 28: PainChange at Day 7: ActivityChange at Day 14: ActivityChange at Day 28: ActivityChange at Day 7: NauseaChange at Day 14: NauseaChange at Day 28: NauseaChange at Day 7: DepressionChange at Day 14 DepressionChange at Day 28: DepressionChange at Day 7: AnxietyChange at Day 14: AnxietyChange at Day 28: AnxietyChange at Day 7: DrowsinessChange at Day 14: DrowsinessChange at Day 28: DrowsinessChange at Day 7: AppetiteChange at Day 14: AppetiteChange at Day 28: AppetiteChange at Day 7: Well-beingChange at Day 14: Well-beingChange at Day 28: Well-beingChange at Day 7: Shortness of breathChange at Day 14: Shortness of breathChange at Day 28: Shortness of breath
Standard Care-1.8-1.3-3.0-0.3-0.3-1.40.50.1-0.6-1.8-1.9-2.60.00.4-1.1-0.7-0.1-1.9-0.20.90.4-0.80.40.4-0.10.6-0.4
Standard Care + Lanreotide Autogel0.1-1.9-1.30.7-0.7-0.30.90.50.0-1.1-2.4-3.7-0.2-2.7-1.50.4-0.6-1.50.50.4-2.0-0.4-0.5-2.30.70.7-0.2

[back to top]

Mean Daily NGT Secretion Volume, in Patients With a NGT

NGT presence and related secretion volume were recorded on the Patient Diary daily until the end of study (Day 28), by the patient or filled in by the nurse/caregiver in case of patient's physical inability. Mean daily secretion volumes, in patients with NGT, is presented. (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation) and Days 7, 14 and 28.

,
Interventionmillilitres (Mean)
BaselineDay 7Day 14Day 28
Standard Care200.04875.012675.027501.0
Standard Care + Lanreotide Autogel700.01025.07090.818301.4

[back to top]

Mean Change From Baseline in ESAS Total Score; FAS

"Quality of Life was assessed using ESAS, evaluating 9 common symptoms in cancer patients: pain, activity, nausea, depression, anxiety, drowsiness, appetite, well-being and shortness of breath. Symptom severity is rated 0-10 on a numerical scale (0=symptom absent; 10=worst severity). ESAS total score is sum of the 9 items (min score=0, max score=90). Low scores indicate good quality of life; high scores indicate strong discomfort. Questionnaire assessments by the patient or by nurse/caregiver in case of patient's physical inability.~Secondary endpoints were analysed using the ITT population but to permit following the FAS which was used for primary endpoint analysis, ESAS total score results are reported for both the ITT and the FAS. Mean change from baseline of ESAS total score at Days 7, 14 and 28 is presented here for the FAS; a positive change indicates a worsening condition." (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation) and Days 7, 14 and 28.

,
InterventionScores on a scale (Mean)
Change at Day 7Change at Day 14Change at Day 28
Standard Care-5.1-1.3-6.8
Standard Care + Lanreotide Autogel1.9-7.3-10.3

[back to top]

Number of Patients Experiencing ≤ 2 Vomiting Episodes/Day During at Least 3 Consecutive Days, in Patients Without NGT

Vomiting episodes and NGT presence were recorded on the Patient Diary daily until the end of study (Day 28), by the patient or filled in by the nurse/caregiver in case of patient's physical inability. Number of patients experiencing ≤ 2 vomiting episodes/day during at least 3 consecutive days, in patients without NGT, is presented. (NCT02365584)
Timeframe: From Baseline (Day 1, before randomisation) to Days 7, 14 and 28.

,
InterventionParticipants (Count of Participants)
From Day 1 to Day 7From Day 1 to Day 14From Day 1 to Day 28
Standard Care544
Standard Care + Lanreotide Autogel444

[back to top]

Assessment of Passage of Stools; ITT Population

Passage of stools assessments (Yes/No) were recorded on the Patient Diary daily until the end of study (Day 28), by the patient or filled in by the nurse/caregiver in case of patient's physical inability. (NCT02365584)
Timeframe: From Baseline (Day 1, before randomisation) to Day 28.

InterventionParticipants (Count of Participants)
Day 172161255Day 172161256Day 272161255Day 272161256Day 372161255Day 372161256Day 472161255Day 472161256Day 572161256Day 572161255Day 672161255Day 672161256Day 772161255Day 772161256Day 872161255Day 872161256Day 972161255Day 972161256Day 1072161255Day 1072161256Day 1172161255Day 1172161256Day 1272161255Day 1272161256Day 1372161255Day 1372161256Day 1472161256Day 1472161255Day 1572161256Day 1572161255Day 1672161256Day 1672161255Day 1772161256Day 1772161255Day 1872161255Day 1872161256Day 1972161255Day 1972161256Day 2072161255Day 2072161256Day 2172161256Day 2172161255Day 2272161255Day 2272161256Day 2372161255Day 2372161256Day 2472161256Day 2472161255Day 2572161256Day 2572161255Day 2672161256Day 2672161255Day 2772161255Day 2772161256Day 2872161255Day 2872161256
YesNo
Standard Care19
Standard Care + Lanreotide Autogel20
Standard Care6
Standard Care + Lanreotide Autogel18
Standard Care + Lanreotide Autogel2
Standard Care15
Standard Care16
Standard Care + Lanreotide Autogel16
Standard Care + Lanreotide Autogel3
Standard Care + Lanreotide Autogel17
Standard Care + Lanreotide Autogel5
Standard Care + Lanreotide Autogel14
Standard Care13
Standard Care + Lanreotide Autogel15
Standard Care5
Standard Care10
Standard Care4
Standard Care11
Standard Care + Lanreotide Autogel11
Standard Care12
Standard Care + Lanreotide Autogel13
Standard Care + Lanreotide Autogel1
Standard Care + Lanreotide Autogel10
Standard Care + Lanreotide Autogel9
Standard Care8
Standard Care + Lanreotide Autogel8
Standard Care + Lanreotide Autogel7
Standard Care2
Standard Care9
Standard Care3
Standard Care7
Standard Care + Lanreotide Autogel6
Standard Care1
Standard Care + Lanreotide Autogel4

[back to top]

Least Squares (LS) Mean Area Under Curve (AUC) of Edmonton Symptom Assessment System (ESAS) Total Scores Collected for the First 7 Days; Full Analysis Set (FAS)

"Quality of Life was assessed using ESAS, evaluating 9 common symptoms in cancer patients: pain, activity, nausea, depression, anxiety, drowsiness, appetite, well-being and shortness of breath. Symptom severity is rated 0-10 on a numerical scale (0=symptom absent; 10=worst severity). ESAS total score is sum of the 9 items (min score=0, max score=90). Low scores indicate good quality of life; high scores indicate strong discomfort. Questionnaire assessments by the patient or by nurse/caregiver in case of patient's physical inability. AUC is area under the line which joins the points defined by plotting ESAS total score on vertical axis and time values on horizontal axis, computed using trapezoidal rule.~Primary endpoint was analysed using the FAS. LS mean AUC of ESAS total scores during first 7 days is presented." (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation), Days 2, 3, 4, 5, 6 and 7.

InterventionScores on a scale x time (day) (Least Squares Mean)
Standard Care179
Standard Care + Lanreotide Autogel190

[back to top]

Mean Change From Baseline in Daily Intensity of Abdominal Pain Score (Visual Analogue Scale [VAS]); ITT Population

"Abdominal pain was assessed using the VAS numeric pain distress scale which is a 100-millimetre (10-centimetre) scoring scale on which patients mark their perceived level of pain. Scores range from 0 to 100 where 0=no pain and 100=unbearable pain. Higher scores indicate a worse outcome. Scores were recorded on the Patient Diary daily until the end of study (Day 28), by the patient or filled in by the nurse/caregiver in case of patient's physical inability.~Mean change from baseline of VAS for abdominal pain at Days 7, 14 and 28 is presented for the ITT population; a positive change indicates a worsening condition." (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation) and Days 7, 14 and 28.

,
InterventionScores on a scale (Mean)
Change at Day 7Change at Day 14Change at Day 28
Standard Care-22.0-15.6-32.0
Standard Care + Lanreotide Autogel-9.6-27.7-17.0

[back to top]

Mean Change From Baseline in ESAS Total Score; ITT Population

"Quality of Life was assessed using ESAS, evaluating 9 common symptoms in cancer patients: pain, activity, nausea, depression, anxiety, drowsiness, appetite, well-being and shortness of breath. Symptom severity is rated 0-10 on a numerical scale (0=symptom absent; 10=worst severity). ESAS total score is sum of the 9 items (min score=0, max score=90). Low scores indicate good quality of life; high scores indicate strong discomfort. Questionnaire assessments by the patient or by nurse/caregiver in case of patient's physical inability.~Secondary endpoints were analysed using the ITT population but to permit following the FAS which was used for primary endpoint analysis, ESAS total score results are reported for both the ITT and the FAS. Mean change from baseline of ESAS total score at Days 7, 14 and 28 is presented here for the ITT population; a positive change indicates a worsening condition." (NCT02365584)
Timeframe: Baseline (Day 1, before randomisation) and Days 7, 14 and 28.

,
InterventionScores on a scale (Mean)
Change at Day 7Change at Day 14Change at Day 28
Standard Care-5.1-1.3-6.8
Standard Care + Lanreotide Autogel1.5-7.3-10.3

[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 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]

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]

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]

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: 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]

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]

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 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)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 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]

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]

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]

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 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]

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 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]

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]

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 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]

Mean Change From Baseline in PanNet Specific Tumour Biomarkers: Pancreatic Polypeptide, Gastrin

PanNET specific tumour peptide biomarkers were evaluated in pancreas subjects at baseline. Only the tumour biomarkers that were above normal range at baseline were evaluated every 12 weeks thereafter and at the end of study visit. The mean change from baseline values in picomole/liter (pmol/L) are presented for the end of study visit. (NCT02651987)
Timeframe: Baseline (Day 1) and end of study (approximately 64 weeks)

Interventionpmol/L (Mean)
Pancreatic PolypeptideGastrin
PanNET Cohort82.7-9.8

[back to top]

Mean Change From Baseline in QoL Questionnaire Gastrointestinal Neuroendocrine Tumour 21 (QLQ-GI.NET21; 2006)

Subjects were asked to complete the EORTC QLQ-GI.NET21 module which comprised 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Answers were converted into grading scale, with values between 0 and 100. Each individual sub-score was transformed to range from 0 to 100. The mean change from baseline at the end of study/early withdrawal visit is presented with a higher score representing more or worse problems. (NCT02651987)
Timeframe: Baseline (Day 1) and end of study (approximately 64 weeks for panNET cohort and 108 weeks for midgut NET (and overall) cohort)

,,
Interventionscore on a scale (Mean)
Endocrine SymptomsGastrointestinal SymptomsTreatment Related SymptomsSocial FunctionDisease Related WorriesMuscle/Bone PainSexual FunctionInformation/Communication FunctionBody Image
Midgut NET Cohort-5.09-2.78-3.47-9.49-0.930.00-2.78-2.90-7.58
Overall-2.96-3.111.08-5.430.99-0.760.002.27-3.97
PanNET Cohort-0.53-3.495.93-0.793.17-1.672.387.940.00

[back to top]

Objective Response Rate (ORR)

The ORR was defined as the percentage of subjects who achieve either complete response (CR) or partial response (PR) according to RECIST v1.0 criteria. ORR was evaluated every 12 weeks and results are presented for each cohort. (NCT02651987)
Timeframe: Weeks 12, 24, 36, 48, 60 (for both cohorts) and Weeks 72, 84, and 96 (for midgut cohort)

Interventionpercentage of subjects (Number)
Week 12Week 24Week 36Week 48Week 60
PanNET Cohort0.00.00.00.00.0

[back to top]

Objective Response Rate (ORR)

The ORR was defined as the percentage of subjects who achieve either complete response (CR) or partial response (PR) according to RECIST v1.0 criteria. ORR was evaluated every 12 weeks and results are presented for each cohort. (NCT02651987)
Timeframe: Weeks 12, 24, 36, 48, 60 (for both cohorts) and Weeks 72, 84, and 96 (for midgut cohort)

Interventionpercentage of subjects (Number)
Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96
Midgut NET Cohort0.00.00.00.02.03.92.02.0

[back to top]

Percentage of Subjects Alive and Progression Free

The percentage of subjects alive and progression-free was assessed throughout the study up to Week 60 for the panNET cohort and Week 96 for the midgut cohort. Disease progression was assessed by tumour response evaluation according to RECIST v1.0, every 12 weeks measured by independent central review using the same imaging technique (CT scan or MRI) for each subject throughout the study. The percentage of subjects alive and progression free was estimated using the Kaplan Meier method for each cohort. (NCT02651987)
Timeframe: Weeks 12, 24, 36, 48, 60 (for both cohorts) and Weeks 72, 84 and 96 (for midgut NET cohort)

Interventionpercentage of subjects (Number)
Week 12Week 24Week 36Week 48Week 60
PanNET Cohort93.364.437.828.520.7

[back to top]

Percentage of Subjects Alive and Progression Free

The percentage of subjects alive and progression-free was assessed throughout the study up to Week 60 for the panNET cohort and Week 96 for the midgut cohort. Disease progression was assessed by tumour response evaluation according to RECIST v1.0, every 12 weeks measured by independent central review using the same imaging technique (CT scan or MRI) for each subject throughout the study. The percentage of subjects alive and progression free was estimated using the Kaplan Meier method for each cohort. (NCT02651987)
Timeframe: Weeks 12, 24, 36, 48, 60 (for both cohorts) and Weeks 72, 84 and 96 (for midgut NET cohort)

Interventionpercentage of subjects (Number)
Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96
Midgut NET Cohort91.865.359.238.336.129.827.525.2

[back to top]

Factors Associated With PFS

"A univariate cox proportional hazards model was used to assess whether the following factors were associated with PFS:~Hepatic tumour load: >25% versus reference ≤25%~Tumour Grade: Grade 2 versus reference Grade 1,~Previous surgery of the primary tumour: No versus reference Yes,~Proliferation index Ki67: ≥10% versus reference <10%~Duration of treatment with lanreotide Autogel® 120 mg every 28 days by category: ≥median value versus reference NCT02651987)
Timeframe: Screening/Baseline (Day 1)

,
InterventionHazard Ratio (Number)
Hepatic tumour load: >25% Vs ≤25%Tumour Grade: 2 Vs 1Previous surgery: No Vs YesKi67: ≥10% Vs <10%Duration of treatment with lanreotide Autogel® 120 mg every 28 days: ≥median Vs Age: ≥65 years Vs <65 yearsTime from diagnosis: ≥3 years Vs <3 yearsTime between CT scans: ≥12 months Vs <12 monthsSymptoms: No Vs Yes
Midgut NET Cohort1.540.902.142.260.761.150.940.721.32
PanNET Cohort0.960.681.043.600.681.550.490.472.55

[back to top]

Mean Change From Baseline in EQ-5D-5L v1.0 Questionnaire (Descriptive System)

"Subjects were instructed to complete the EQ-5D-5L descriptive system at baseline and every 12 weeks throughout the study.~The EQ-5D-5L descriptive system comprised the following 5 dimensions: mobility, self-care, usual activities, pain/discomfort and anxiety/depression. Each dimension had 5 levels: no problems, slight problems, moderate problems, severe problems, extreme problems. The EQ-5D-5L health states, defined by the EQ-5D-5L descriptive system, was converted into a single index value with scores ranging from 0 (no problems) to 1 (extreme problems). The mean change from baseline at the end of study/early withdrawal visit is presented with a positive change from baseline in the index values indicating a worsening of symptoms." (NCT02651987)
Timeframe: Baseline (Day 1) and end of study (approximately 64 weeks for panNET cohort and 108 weeks for midgut NET (and overall) cohort)

InterventionIndex value (Mean)
PanNET Cohort-0.04
Midgut NET Cohort0.00
Overall-0.02

[back to top]

Mean Change From Baseline in EQ-5D-5L v1.0 Questionnaire (VAS)

Subjects were instructed to complete the EQ-5D-5L VAS at baseline and every 12 weeks throughout the study. The EQ-5D-5L VAS recorded the subject's self-rated health on a vertical VAS which is numbered from 0 (worst health state) to 100 (best health state). The mean change from baseline at the end of study/early withdrawal visit is presented with a positive change in the VAS indicating an improvement in symptoms. (NCT02651987)
Timeframe: Baseline (Day 1) and end of study (approximately 64 weeks for panNET cohort and 108 weeks for midgut NET (and overall) cohort)

Interventionscore on a scale (Mean)
PanNET Cohort-1.90
Midgut NET Cohort-1.76
Overall-1.83

[back to top]

Mean Change From Baseline in PanNet Specific Tumour Biomarkers: Glucagon

PanNET specific tumour peptide biomarkers were evaluated in pancreas subjects at baseline. Only the tumour biomarkers that were above normal range at baseline were evaluated every 12 weeks thereafter and at the end of study visit. The mean change from baseline values in nanograms (ng)/L are presented for the end of study visit. (NCT02651987)
Timeframe: Baseline (Day 1) and end of study (approximately 64 weeks)

Interventionng/L (Mean)
PanNET Cohort5.5

[back to top]

Mean Change From Baseline in QoL Measured Using EORTC, QLQ-C30 v3.0 (Global Health Status Sub-score)

"Subjects were instructed to complete the 30 questions in the EORTC-QLQ-C30 v3.0 questionnaire at baseline and every 12 weeks throughout the study.~The global health status sub-score was assessed using the last 2 questions which represented subject's assessment of overall health & QoL. Each question was coded on a 7-point scale (1=very poor to 7=excellent). The sub-score was transformed to range from 0-100, with a high score for global health status representing a high QoL. The mean change from baseline in the transformed global health status are presented for the end of study/early withdrawal visit, with a positive change indicating an improvement in QoL." (NCT02651987)
Timeframe: Baseline (Day 1) and end of study (approximately 64 weeks for panNET cohort and 108 weeks for midgut NET (and overall) cohort)

Interventionscore on a scale (Mean)
PanNET Cohort-0.38
Midgut NET Cohort-1.33
Overall-0.89

[back to top]

Median Duration of Stable Disease

Median duration of SD was the time from first injection of lanreotide Autogel® 120 mg every 14 days until the first occurrence of PD by central assessment. Disease progression was assessed by tumour response evaluation according to RECIST v1.0, every 12 weeks, measured using the same imaging technique (CT scan or MRI) for each subject throughout the study. Median duration of stable disease was estimated using the Kaplan Meier method for each cohort. (NCT02651987)
Timeframe: From Day 1 up to Week 60 for the panNET cohort and Week 103 for the midgut NET cohort

Interventionmonths (Median)
PanNET Cohort8.3
Midgut NET Cohort13.8

[back to top]

Median Progression Free Survival (PFS)

PFS was defined as the time from first injection of lanreotide Autogel® 120 mg every 14 days to progression or death. Disease progression was assessed by tumour response evaluation according to RECIST v1.0, every 12 weeks, measured by independent central review using the same imaging technique (computed tomography [CT] scan or magnetic resonance imaging [MRI]) for each subject throughout the study. The median PFS time was estimated using the Kaplan Meier method for each cohort. (NCT02651987)
Timeframe: From Day 1 up to Week 60 for the panNET cohort and Week 103 for the midgut NET cohort

Interventionmonths (Median)
PanNET Cohort5.6
Midgut NET Cohort8.3

[back to top]

Median Time to Progression

Time to Progression was defined as time from first injection of lanreotide Autogel® 120 mg every 14 days to progression. Disease progression was assessed by tumour response evaluation according to RECIST v1.0, every 12 weeks, measured by independent central review using the same imaging technique (CT scan or MRI) for each subject throughout the study. Median time to progression was estimated using the Kaplan Meier method for each cohort. (NCT02651987)
Timeframe: From Day 1 up to Week 60 for the panNET cohort and Week 103 for the midgut NET cohort

Interventionmonths (Median)
PanNET Cohort5.6
Midgut NET Cohort8.7

[back to top]

Overall Survival

Overall survival was defined as the time in months from the first injection of lanreotide Autogel® 120 mg every 14 days to death due to any cause. Median overall survival was estimated using the Kaplan Meier method for each cohort. (NCT02651987)
Timeframe: From Day 1 up to Week 60 for the panNET cohort and Week 103 for the midgut NET cohort

Interventionmonths (Median)
PanNET CohortNA
Midgut NET CohortNA

[back to top]

Best Overall Response Rate

Best overall response was defined as the best response recorded from the initiation of treatment until disease progression, according to RECIST v1.0 evaluation. The percentage of subjects in each response category and those who were non-evaluable (i.e. with no tumour assessment after the start of study treatment) throughout the study are presented for each cohort. (NCT02651987)
Timeframe: From Day 1 up to Week 60 for the panNET cohort and Week 103 for the midgut NET cohort

,
Interventionpercentage of subjects (Number)
CRPRSDPDNot evaluable
Midgut NET Cohort0.03.968.623.52.0
PanNET Cohort0.00.066.731.30.0

[back to top]

Disease Control Rate (DCR)

The DCR was defined as the percentage of subjects who achieved CR plus PR plus Stable Disease (SD), evaluated according to RECIST v1.0 criteria. The DCR at Weeks 24 and 48 is presented for each cohort. (NCT02651987)
Timeframe: Weeks 24 and 48

,
Interventionpercentage of subjects (Number)
Week 24Week 48
Midgut NET Cohort58.833.3
PanNET Cohort43.822.9

[back to top]

Mean Change From Baseline in Nonspecific Tumour Biomarkers

Nonspecific tumour peptide biomarkers (chromogranin A [CgA], neuron specific enolase [NSE] and plasma/urinary 5-hydroxyindoleacetic acid [5-HIAA]) were evaluated in both pancreas and midgut subjects at baseline and Week 12 and every 12 weeks thereafter. At all scheduled visits, except baseline, plasma/urinary 5-HIAA was only performed in subjects with symptoms of carcinoid syndrome (diarrhoea and/or flushing) or if urinary 5-HIAA was elevated (above upper limit of normal [ULN]) at baseline. Mean change from baseline values were normalised by the ULN (xULN) and are presented for each cohort. (NCT02651987)
Timeframe: Baseline (Day 1) and end of study (approximately 64 weeks for panNET cohort and 108 weeks for midgut NET cohort)

,
InterventionxULN (Mean)
CgANSEPlasma 5-HIAA
Midgut NET Cohort0.370-0.493.90
PanNET Cohort0.2050.03-0.42

[back to top]

Mean Change From Baseline in Number of Stools and Flushing Episodes

Symptom control was measured by the total number of stools (diarrhoea) and flushing episodes during the 7 days prior to the visit, reported orally by the subject to the investigator. The mean change from baseline in number of stools and flushing episodes reported at each visit is presented for each cohort. (NCT02651987)
Timeframe: Baseline (Day 1), Weeks 8,12, 48 and end of study (approximately 64 weeks for panNET cohort and 108 weeks for midgut NET cohort)

,
Interventionepisodes (Mean)
Stools - Week 8Stools - Week 12Stools - Week 48Stools - End of StudyFlushing - Week 8Flushing - Week 12Flushing - Week 48Flushing - End of Study
Midgut NET Cohort-1.00.73.4-1.2-3.31.5-1.5-0.5
PanNET Cohort1.0-1.2-1.00.50.7-1.0-1.00.0

[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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

Coefficient of Agreement Between Central and Local Assessment of Tumor Radiological Response at Month 9

Differences between central radiology review and local investigator review were assessed according to the DCR status and the number of agreements and disagreements between the evaluators (central versus local) along with the p-values from the kappa test. A kappa statistic was used to evaluate the concordance between the central and the local review. (NCT02698410)
Timeframe: Month 9

Interventionkappa coefficient (Number)
Lanreotide ATG Plus Temozolomide0.71

[back to top]

DCR Assessed Centrally at Month 9

The DCR was defined as SD, PR or CR according to RECIST criteria v1.1. A second set of the original computed tomography (CT) scan images were used for a centralized RECIST v1.1 assessment by an independent radiologist. (NCT02698410)
Timeframe: Up to Month 9

Interventionpercentage of participants (Number)
Lanreotide ATG Plus Temozolomide28.2

[back to top]

Best Overall Response (BOR) Assessed Locally and Centrally

The BOR was defined as the highest OR achieved by the participant from the time of first treatment until disease progression/recurrence or the end of study according to RECIST criteria v1.1 and was classified as: CR > PR > Non-CR/Non-progressive disease (NCR/NPD) > SD > PD > ND > not evaluable (NE). The BOR was assessed locally by the investigator and centrally by an independent radiologist. Percentages are based on the number of participants in the ITT/Safety population with non-missing observations. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionpercentage of participants (Number)
Local assessment: CRLocal assessment: PRLocal assessment: SDLocal assessment: NCR/NPDLocal assessment: PDLocal assessment: NELocal assessment: Not applicableCentral assessment: CRCentral assessment: PRCentral assessment: SDCentral assessment: NCR/NPDCentral assessment: PDCentral assessment: NECentral assessment: Not applicable
Lanreotide ATG Plus Temozolomide07.771.8020.500013.265.82.615.802.6

[back to top]

DCR Assessed Locally and Centrally at Month 12

The DCR was defined as SD, PR or CR according to RECIST criteria v1.1. The DCR was assessed locally by the investigator and centrally by an independent radiologist. (NCT02698410)
Timeframe: Month 12

Interventionpercentage of participants (Number)
Local assessmentCentral assessment
Lanreotide ATG Plus Temozolomide17.515.4

[back to top]

DCR Assessed Locally and Centrally at Month 9 by Carcinoid Type

The influence of type of carcinoid [typical, atypical and undetermined carcinoid neuroendocrine tumors (NET)] on the local and central DCR at 9 months was analysed. Typical carcinoids were defined with absent foci of necrosis and mitotic count < 2 mitoses/2 millimeters^2 (mm^2); atypical carcinoids were defined with presence of foci of necrosis and/or 2 mitoses/2 mm^2 <= mitotic count <= 10 mitoses/2 mm^2; and carcinoid NET were defined as confirmed carcinoid without foci of necrosis and/or mitotic count reported. The DCR was assessed locally by the investigator and centrally by an independent radiologist. (NCT02698410)
Timeframe: Up to Month 9

Interventionpercentage of participants (Number)
Local assessment: Typical carcinoidLocal assessment: Atypical carcinoidLocal assessment: Carcinoid NETCentral assessment: Typical carcinoidCentral assessment: Atypical carcinoidCentral assessment: Carcinoid NET
Lanreotide ATG Plus Temozolomide12.547.627.3035.036.4

[back to top]

Disease Control Rate (DCR) Assessed Locally at Month 9

Responders were participants who showed disease control according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria v 1.1 assessed locally by the investigator. The DCR was defined as complete response (CR), partial response (PR) or stable disease (SD) according to RECIST criteria v1.1. A sensitivity analysis-1 of local DCR was performed excluding participants withdrawn before 9 months with reason other than progressive disease (PD) or missing assessment and considering participants with PD prior or at 9 months as failures. In addition, a sensitivity analysis-2 was performed in order to consider assessments done between 7.5 and 10.5 months as 9 months assessments when 9-month assessment was missing using same methodology, i.e. considering PD prior or at 9 months and participants withdrawn with other or missing reasons as failures. (NCT02698410)
Timeframe: Up to Month 9; for sensitivity analysis-2, up to 10.5 months

Interventionpercentage of participants (Number)
DCR at Month 9Sensitivity analysis-1Sensitivity analysis-2
Lanreotide ATG Plus Temozolomide35.045.245.0

[back to top]

Influence of Biomarkers Expression on Locally and Centrally Assessed DCR at Months 9 and 12

The following biomarkers were investigated: Immunohistochemistry assay SSTR2 including HER-2 score (0, 1+, 2+, 3+ and +ve versus -ve); H-score (from 0 to 300 as quantitative variable and +ve versus -ve); IRS score (from 0 to 12 and reference for odds ratio was 0-1). Ki67 index (from 0% to 100% and reference for odds ratio was 4%-25%). MGMT expression including percentage of +ve nuclei stained and methylated sites and H-score (from 0 to 300 as quantitative variable). For all these categories, higher score indicates a higher expression of biomarkers. Carcinoid type (typical or NET) was also investigated. Prognostic value of biomarkers expression at screening on DCR were assessed locally and centrally using univariate logistic regression models. (NCT02698410)
Timeframe: Screening period, Months 9 and 12

Interventionodds ratio (Number)
Month 9: Local: SSTR2: HER-2 score; +ve (2+, 3+)Month 9: Local: SSTR2: H-score; +ve (>= 50)Month 9: Local: SSTR2: IRS score; 2-3Month 9: Local: SSTR2: IRS score; 4-8Month 9: Local: SSTR2: IRS score; 9-12Month 9: Local: Ki67: <4Month 9: Local: Ki67: >=25Month 9:Local: MGMT: +ve nuclei stained; +ve(>=5%)Month 9: Local: MGMT: Methylated sites; +ve (>10%)Month 9: Local: MGMT: H-scoreMonth 9: Local: Carcinoid type: TypicalMonth 9: Local: Carcinoid type: Carcinoid NETMonth 9: Central: SSTR2: HER-2 score; +ve (2+, 3+)Month 9: Central: SSTR2: H-score; +ve (>= 50)Month 9: Central: SSTR2: IRS score; 2-3Month 9: Central: SSTR2: IRS score; 4-8Month 9: Central: SSTR2: IRS score; 9-12Month 9: Central: Ki67: <4Month 9: Central: Ki67: >=25Month 9:Central:MGMT:+ve nuclei stained; +ve(>=5%)Month 9:Central: MGMT: Methylated sites; +ve(>10%)Month 9: Central: MGMT: H-scoreMonth 9: Central: Carcinoid type: TypicalMonth 9: Central: Carcinoid type: Carcinoid NETMonth 12: Local: SSTR2: HER-2 score; +ve (2+, 3+)Month 12: Local: SSTR2: H-score; +ve (>= 50)Month 12: Local: SSTR2: IRS score; 2-3Month 12: Local: SSTR2: IRS score; 4-8Month 12: Local: SSTR2: IRS score; 9-12Month 12: Local: Ki67: <4Month 12: Local: Ki67: >=25Month 12:Local:MGMT: +ve nuclei stained; +ve(>=5%)Month 12: Local: MGMT: Methylated sites; +ve(>10%)Month 12: Local: MGMT: H-scoreMonth 12: Local: Carcinoid type: TypicalMonth 12: Local: Carcinoid type: Carcinoid NETMonth 12: Central: SSTR2: HER-2 score; +ve(2+, 3+)Month 12: Central: SSTR2: H-score; +ve (>= 50)Month 12: Central: SSTR2: IRS score; 2-3Month 12: Central: SSTR2: IRS score; 4-8Month 12: Central: SSTR2: IRS score; 9-12Month 12: Central: Ki67: <4Month 12: Central: Ki67: >=25Month 12:Central:MGMT:+ve nuclei stained;+ve(>=5%)Month 12:Central: MGMT: Methylated sites;+ve(>10%)Month 12: Central: MGMT: H-scoreMonth 12: Central: Carcinoid type: TypicalMonth 12: Central: Carcinoid type: Carcinoid NET
Lanreotide ATG Plus Temozolomide0.900.78NA0.6712.00NA1.67NA3.250.990.160.411.402.253.000.6712.00NANA0.505.001.00NA1.063.004.40NA0.7010.50NANA0.202.130.990.460.322.083.00NANA10.50NANA0.143.000.990.570.40

[back to top]

Influence of Biomarkers Expression on Locally and Centrally Assessed PFS

The following biomarkers were investigated: Immunohistochemistry assay human somatostatin receptors 2 (SSTR2) including human epidermal growth factor receptor 2 (HER-2) score [0, 1+, 2+, 3+ and positive (+ve) versus negative (-ve)]; hormone receptor score (H-score) (from 0 to 300 as quantitative variable and +ve versus -ve); immunoreactive score (IRS) score (from 0 to 12 and reference for hazard ratio was 0-1). Ki67 index (from 0% to 100% and reference for hazard ratio was 4%-25%). O^6-methylguanine-DNA methyltransferase (MGMT) expression including percentage of +ve nuclei stained and methylated sites and H-score (from 0 to 300 as quantitative variable). For all these categories, higher score indicates a higher expression of biomarkers. Carcinoid type (typical or NET) was also investigated. Prognostic value of biomarkers expression at screening on PFS were assessed locally and centrally using univariate cox proportional hazard models. (NCT02698410)
Timeframe: From Screening period (-4 weeks) up to Week 52

Interventionhazard ratio (Number)
Local: SSTR2: HER-2 score; +ve (2+, 3+)Local: SSTR2: H-score; +ve (>= 50)Local: SSTR2: IRS score; 2-3Local: SSTR2: IRS score; 4-8Local: SSTR2: IRS score; 9-12Local: Ki67: <4Local: Ki67: >=25Local: MGMT: +ve nuclei stained; +ve (>=5%)Local: MGMT: Methylated sites; +ve (>10%)Local: MGMT: H-scoreLocal: Carcinoid type: TypicalLocal: Carcinoid type: Carcinoid NETCentral: SSTR2: HER-2 score; +ve (2+, 3+)Central: SSTR2: H-score; +ve (>= 50)Central: SSTR2: IRS score; 2-3Central: SSTR2: IRS score; 4-8Central: SSTR2: IRS score; 9-12Central: Ki67: <4Central: Ki67: >=25Central: MGMT: +ve nuclei stained; +ve (>=5%)Central: MGMT: Methylated sites; +ve (>10%)Central: MGMT: H-scoreCentral: Carcinoid type: TypicalCentral: Carcinoid type: Carcinoid NET
Lanreotide ATG Plus Temozolomide0.710.660.310.900.121.081.682.060.411.001.051.590.500.360.780.880.100.002.752.110.731.000.990.61

[back to top]

Median Duration of Response (DOR) Assessed Locally and Centrally

The DOR was defined as the time from onset of the first objective tumor response (PR or CR) to objective tumor progression (PD) according to RECIST criteria v 1.1. The DOR was assessed locally by the investigator and centrally by an independent radiologist. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus TemozolomideNANA

[back to top]

Median Progression Free Survival (PFS) Assessed Locally and Centrally

The PFS was defined as the time from the first treatment administration to disease progression according to RECIST criteria v 1.1 or death from any cause. The PFS was assessed locally by the investigator and centrally by an independent radiologist. The distribution of PFS times was estimated using the Kaplan-Meier method. The PFS of participants who were lost to follow-up and those who had not progressed at end of study were censored at the date of the last disease assessment. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus Temozolomide37.137.1

[back to top]

Median Time to Progression (TTP) Assessed Locally and Centrally

The TTP was defined as the time from first treatment administration to the first objective tumor progression (PD) according to RECIST criteria v 1.1. The TTP was assessed locally by the investigator and centrally by an independent radiologist. The distribution of TTP times was estimated using the Kaplan-Meier method. The TTP of participants who were lost to follow-up, and those who had not progressed at end of study were censored at the date of the last disease assessment. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus Temozolomide37.137.1

[back to top]

Median Time to Response (TTR) Assessed Locally and Centrally

The TTR was defined as the time from first treatment administration to the first objective tumor response (PR or CR according to RECIST criteria v 1.1). The TTR was assessed locally by the investigator and centrally by an independent radiologist. The distribution of TTR was estimated using the Kaplan-Meier method. The TTR of participants who were lost to follow-up or died prior to any objective tumor response were censored at the date of the last disease assessment. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus TemozolomideNANA

[back to top]

Neuron-Specific Enolase (NSE) and CgA Biomarker Levels

The NSE and CgA levels were classified according to ULN as follows: < 1 ULN, 1-2 ULN and > 2 ULN. Baseline was defined as value at Day 1. Percentages are based on the number of participants in the ITT/Safety population who attended the visit with non-missing observations. (NCT02698410)
Timeframe: Baseline and Weeks 4, 12, 24, 36 and 52

Interventionpercentage of participants (Number)
CgA: Baseline: <1 ULNCgA: Baseline: 1-2 ULNCgA: Baseline: >2 ULNCgA: Week 4: <1 ULNCgA: Week 4: 1-2 ULNCgA: Week 4: >2 ULNCgA: Week 12: <1 ULNCgA: Week 12: 1-2 ULNCgA: Week 12: >2 ULNCgA: Week 24: <1 ULNCgA: Week 24: 1-2 ULNCgA: Week 24: >2 ULNCgA: Week 36: <1 ULNCgA: Week 36: 1-2 ULNCgA: Week 36: >2 ULNCgA: Week 52: <1 ULNCgA: Week 52: 1-2 ULNCgA: Week 52: >2 ULNNSE: Baseline: <1 ULNNSE: Baseline: 1-2 ULNNSE: Baseline: >2 ULNNSE: Week 4: <1 ULNNSE: Week 4: 1-2 ULNNSE: Week 4: >2 ULNNSE: Week 12: <1 ULNNSE: Week 12: 1-2 ULNNSE: Week 12: >2 ULNNSE: Week 24: <1 ULNNSE: Week 24: 1-2 ULNNSE: Week 24: >2 ULNNSE: Week 36: <1 ULNNSE: Week 36: 1-2 ULNNSE: Week 36: >2 ULNNSE: Week 52: <1 ULNNSE: Week 52: 1-2 ULNNSE: Week 52: >2 ULN
Lanreotide ATG Plus Temozolomide40.015.045.034.322.942.946.417.935.730.025.045.031.312.556.327.39.163.665.025.010.062.925.711.478.617.93.680.010.010.087.56.36.363.618.218.2

[back to top]

Objective Response Rate (ORR) Assessed Locally and Centrally at Months 9 and 12

The ORR was defined as the percentage of participants with CR or PR according to RECIST criteria v1.1. The ORR was assessed locally by the investigator and centrally by an independent radiologist. The ORR was based on the participants with PD prior to 9 and 12 months with respectively PD at 9 and 12 months, and participants withdrawn before the assessment for reason other than PD or missing as failures. (NCT02698410)
Timeframe: Months 9 and 12

Interventionpercentage of participants (Number)
Local assessment: Month 9Local assessment: Month 12Central assessment: Month 9Central assessment: Month 12
Lanreotide ATG Plus Temozolomide2.52.55.12.6

[back to top]

Percentage of Biochemical Responders According to Chromogranin A (CgA) Plasma Levels

Participants with baseline CgA plasma levels greater than the upper limit of normal (ULN) were assessed for a biochemical response. A biochemical responder was defined as a participant who had a decrease of CgA >= 50% compared to baseline, while biochemical SD was defined as a decrease < 50% or an increase <= 25% compared to baseline. Biochemical non-responders had an increase >25% compared to baseline. Baseline was defined as value at Day 1. (NCT02698410)
Timeframe: Baseline (Day 1) and Week 4, 12, 24, 36 and 52

Interventionpercentage of participants (Number)
Week 4: RespondersWeek 4: SDWeek 4: Non-respondersWeek 12: RespondersWeek 12: SDWeek 12: Non-respondersWeek 24: RespondersWeek 24: SDWeek 24: Non-respondersWeek 36: RespondersWeek 36: SDWeek 36: Non-respondersWeek 52: RespondersWeek 52: SDWeek 52: Non-responders
Lanreotide ATG Plus Temozolomide27.359.113.637.537.525.023.130.846.236.427.336.412.550.037.5

[back to top]

Number of Participants With Treatment-emergent Adverse Events as a Measure of Safety and Tolerability

Treatment-emergent Adverse Events were assessed according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) v4.03. (NCT02859064)
Timeframe: From the day of the first dose to 30 days after the last dose of study medication, up to 52 months

InterventionParticipants (Count of Participants)
Lanreotide/Y-90 Microspheres6

[back to top]

Disease Control Rate

Percentage of patients with CR, PR or stable disease (SD) according to RECIST v1.1. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1),: Complete Response (CR): Disappearance of all target and non-target lesions; Partial Response (PR): >=30% decrease in the sum of the diameters of target lesions; Stable Disease: Meeting neither criteria for PR or Progression (PD= greater than 20% increase in the sum of diameters of target lesions, or an unequivocal increase in a non-target lesion, or the appearance of new lesions). Disease Control Rate (DCR) = CR + PR + SD. (NCT02859064)
Timeframe: At 12 weeks post-treatment with SIR-Spheres then every 8 weeks, up to 52 months.

Interventionpercentage of participants (Number)
Lanreotide/Y-90 Microspheres100

[back to top]

Overall Response Rate

Percentage of patients with confirmed complete or partial response (CR or PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1),: Complete Response (CR): Disappearance of all target and non-target lesions; Partial Response (PR): >=30% decrease in the sum of the diameters of target lesions; Overall Response (OR) = CR + PR. (NCT02859064)
Timeframe: At 12 weeks post-treatment with SIR-Spheres then every 8 weeks, up to 52 months

Interventionpercentage of participants (Number)
Lanreotide/Y-90 Microspheres16.7

[back to top]

Overall Survival

The time from Day 1 of study drug administration until death from any cause. (NCT02859064)
Timeframe: At 12 weeks post-treatment with SIR-Spheres then every 8 weeks, up to 52 months

Interventionmonths (Median)
Lanreotide/Y-90 Microspheres11.5

[back to top]

Progression Free Survival

The time from Day 1 of study drug administration to disease progression as defined by RECIST v1.1, or death on study. Per RECIST V1.1 criteria, progression is defined as a greater than 20% increase in the sum of diameters of target lesions, or an unequivocal increase in a non-target lesion, or the appearance of new lesions. (NCT02859064)
Timeframe: At 12 weeks post-treatment with SIR-Spheres then every 8 weeks, up to 52 months

Interventionmonths (Median)
Lanreotide/Y-90 Microspheres11

[back to top]

Effect of Lanreotide on Gastrointestinal Motility as Measured by Smart Pill

If the small bowel transit time, as measured by wireless capsule endoscopy, is decreased to < 6hrs, then patient would be considered a responder and that lanreotide is efficacious. (NCT03012594)
Timeframe: 3 months

Interventionminutes (Mean)
Gastric emptying timeSmall bowel transit timeColonic transit timeSmall bowel and Colonic transit timeWhole gut transit time
Lanreotide371.52392476751595530

[back to top]

"Improvement in Symptoms as Accessed by Patient Assessment of Upper GastroIntestinal Symptom Severity Index"

"Improvement in symptoms assessed by improvement in Patient Assessment of Gastrointestinal Disorders Symptom Severity Index(PAGI-SYM) scores. If the PAGI-Sym scores were decreased by at least 0.7 points at 3 months when compared to baseline/pre treatment, then it will be considered that Lanreotide has significantly improved the symptom severity. Higher values represent worse symptoms.~The participant rated each of the measured gastrointestinal symptom severity as described 0=No symptom, 1=Very Mild Symptom, 2= Mild Symptoms, 3= Moderate symptom, 4=Severe symptom, 5= Very Severe symptom.~PAGI-SYM is a brief (20-items with 6 sub scales) symptom severity questionnaire that captures information on common upper gastrointestinal symptoms which include including Heartburn/regurgitation, Nausea/vomiting, Fullness/early satiety, bloating, Upper abdominal pain, and Lower abdominal pain. The presented data is an average of each sub scale." (NCT03012594)
Timeframe: 3 months

Interventionscore on a scale (Mean)
Heartburn/regurgitationNausea/vomitingFullness/early satietyBloatingUpper abdominal painLower abdominal pain
Lanreotide1.481.002.362.432.142.00

[back to top] [back to top]

Objective Response Rate (ORR) as Measured by Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1)

ORR is calculated as the number of people with a complete response (CR) or partial response (PR), divided by the total number of people treated. Complete response is defined as disappearance of all target lesions. Partial response is defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. A lower limit of the true ORR will be estimated by the 90% exact lower confidence bound (LCB) for the binomial proportion. A 90% LCB of < 0.1 will be considered not to be of clinical value. If the 90% LCB is ≥ 0.1, the regimen will be considered efficacious. (NCT03043664)
Timeframe: Approximately every 12 weeks until study completion (up to 2 years)

Interventionproportion of participants (Number)
Somatuline Depot and Keytruda0.045

[back to top] [back to top]

Months of Progression-free Survival (PFS)

Months from treatment start date until the date of first documented radiographic progression or date of death from any cause (whichever is first); assessed up to 48 weeks after the last subject has finished study drug regimen. Progression is defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered progression. (NCT03043664)
Timeframe: Up to 3 years

Interventionmonths (Median)
Somatuline Depot and Keytruda5.04

[back to top]

Months of Overall Survival (OS)

Months from treatment start date until the date of death from any cause; assessed up to 48 weeks after the last subject has finished the study drug regimen (NCT03043664)
Timeframe: Up to 59 months

Interventionmonths (Median)
Somatuline Depot and Keytruda28.6

[back to top]

Overall Postoperative Morbidity

Overall morbidity was defined based on the National Surgical Quality Improvement Program (NSQIP) templates. This measure counts the number of participants who had a complication during the 60-day study period. (NCT03174353)
Timeframe: Measured throughout 60 days

InterventionParticipants (Count of Participants)
Lanreotide Arm26

[back to top]

Combined Clinically Significant Postoperative Pancreatic Fistula and Intra-abdominal Abscess

Reduction in the incidence of intraabdominal abscess or development of clinically significant (grade B or C) pancreatic fistula within 60 postoperative days. The grading of the primary outcome measure is based on a method published by the International Study Group of Pancreatic Surgery (ISGPS) in 2005 and then revised in 2016. As this study was developed prior to the revision, the 2005 version was used. This system for grading pancreatic fistula is based on progressively increasing severity of the complication from grade A to C. Grade Grade A fistula are characterized by elevated amylase detected within fluid coming from drain(s) placed at the time of surgery. Grade B fistula require change in patient management such as need for drain placement or institution of antimicrobial therapy, while grade C fistula are associated with severe clinical signs or symptoms such as sepsis. Please refer to page 12 of the study protocol for further details. (NCT03174353)
Timeframe: Measured throughout 60 days

InterventionParticipants (Count of Participants)
Lanreotide Arm8

[back to top]

Biochemical Leak (Grade A Postoperative Pancreatic Fistula)

Grade A pancreatic fistula will be defined based on principles noted in evaluation of Primary endpoint. Grade A fistula are characterized by elevated amylase detected within fluid coming from drain(s) placed at the time of surgery, but do not cause deviation from standard post-operative recovery. (NCT03174353)
Timeframe: Measured throughout 60 days

InterventionParticipants (Count of Participants)
Lanreotide Arm12

[back to top]