Study to Determine the Maximum Tolerated Dose, Safety and Tolerability of a Single Dose of Lanreotide Prolonged Release Formulation (PRF) in Subjects With Acromegaly

April 24, 2019 updated by: Ipsen

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 Either Octreotide LAR or Lanreotide Autogel

The objectives of the protocol is to determine the maximum tolerated dose and to investigate the pharmacokinetics of a single dose of lanreotide PRF in subjects with acromegaly.

Study Overview

Status

Completed

Conditions

Intervention / Treatment

Study Type

Interventional

Enrollment (Actual)

28

Phase

  • Phase 2
  • Phase 1

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

      • Edegem, Belgium
        • Antwerp University Hospital
      • Liège, Belgium
        • Domaine Universitaire Sart Tilman
      • Brno, Czechia
        • Fakultni nemocnice u sv. Anny v Brne
      • Hradec Králové, Czechia
        • Fakultní nemocnice Hradec Králové (University Hospital Hradec)
      • Dijon, France, 21079
        • CHU Le Bocage
      • Le Kremlin-Bicêtre, France, 94275
        • Hôpital de Bicêtre (AP-HP)
      • Marseille, France, 13385
        • CHU de la Timone
      • Pessac, France, 33604
        • Hopital Haut Leveque
      • Berlin, Germany, 10117
        • University Medicine Berlin
      • Hamburg, Germany, 20246
        • University Medical Center Hamburg-Eppendorf
      • Genova, Italy, 16139
        • IRCCS AOU San Martino-IST, University of Genova
      • Padova, Italy, 35128
        • Azienda Ospedaliera Padova
      • Palermo, Italy, 90127
        • Policlinico of Palermo
      • Pisa, Italy, 56124
        • Ospedale Cisanello
      • Siena, Italy, 53100
        • Azienda Ospedaliera Universitaria Senese
      • Torino, Italy, 10126
        • AO Città della Salute e della Scienza di Torino
      • Kaunas, Lithuania
        • Lithuanian University of Health Sciences (LUHS) Kauno klinikos
      • Vilnius, Lithuania
        • Vilnius University Hospital Santariskiu Klinikos
      • Rotterdam, Netherlands, 3000 ca
        • Erasmus University Medical Centre Rotterdam
      • Bialystok, Poland, 15-276
        • Uniwersytecki Szpital Kliniczny w Białymstoku
      • Poznan, Poland, 60-355
        • Szpital Kliniczny im. H. Święcickiego UM w Poznaniu
      • Warszawa, Poland, 01-809
        • Szpital Bielanski im. ks. Jerzego Popieluszki SPZOZ
      • Wroclaw, Poland, 50-367
        • Szpital Kliniczny nr 1
      • Bucharest, Romania, 11863
        • National Institute of Endocrinology
      • Kazan, Russian Federation, 420012
        • Kazan state Medical Academy
      • Kemerovo, Russian Federation, 650066
        • Kemerovo Regional Clinical Hospital
      • Moscow, Russian Federation, 117036
        • Endocrinological Research Center Ministry of Health Russian Federation
      • Nizhniy Novgorod, Russian Federation, 603126
        • Healthcare Institution
      • Saint-Petersburg, Russian Federation, 191015
        • Federal State Budgetary Military
      • Saint-Petersburg, Russian Federation, 191015
        • North-Western State Medical University
      • Barcelona, Spain, 08035
        • Hospital Universitario Vall d' Hebrón
      • Madrid, Spain, 28034
        • Hospital Ramón y Cajal
      • Sevilla, Spain, 41013
        • Hospital Universitario Virgen del Rocio
      • London, United Kingdom, EC1M 6BQ
        • Queen Mary, University of London
      • Manchester, United Kingdom, M20 4BX
        • Christie NHS Foundation Trust
      • Oxford, United Kingdom, OX3 7LE
        • Churchill Hospital

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years to 75 years (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria:

  • Documented diagnosis of acromegaly.
  • Provided written informed consent prior to any study related procedures.
  • Between 18 and 75 years of age inclusive.
  • Female of non-childbearing potential or male. Non-childbearing potential is defined as being postmenopausal for at least 1 year, or women with documented infertility (natural or acquired).
  • Male subjects must agree that, if their partner is at risk of becoming pregnant, they will use a medically accepted, effective method of contraception (i.e. condom) for the duration of the study (maximum of 7.5 months).
  • Treatment with a stable dose of either octreotide LAR or lanreotide Autogel for at least 3 months immediately prior to study entry, with confirmation of disease control during this treatment period (documentation of age adjusted IGF 1 <1.3 x upper limit of normal (ULN), based on local laboratory results, during screening period).
  • If the subject is receiving treatment for hypertension, the dose has been stable for at least 1 month prior to study entry.
  • Subjects must be willing and able to comply with study restrictions and to remain at the clinic for the required duration during the study period and willing to return to the clinic for the follow up evaluation as specified in the protocol.

Exclusion Criteria:

  • Has undergone radiotherapy within 2 years prior to study entry.
  • Has been treated with a dopamine agonist and/or GH receptor antagonist or has undergone pituitary surgery within 3 months prior to study entry.
  • Is anticipated to require pituitary surgery or radiotherapy during the study.
  • Has clinically significant hepatic abnormalities and/or alanine aminotransferase (ALT) and/or aspartate aminotransferase (AST) ≥3 x ULN and/or alkaline phosphatase (AP) ≥2.5 x ULN and/or total bilirubin ≥1.5 x ULN and/or gamma-glutamyl transpeptidase (GGT) ≥2.5 x ULN during the Screening period (central laboratory results) or a history of these findings when on somatostatin analogue (SSTa) treatment.
  • Has clinically significant pancreatic abnormalities and/or amylase and/or lipase ≥1.5 x ULN during the Screening period (central laboratory results).
  • Has any significant renal abnormalities and/or creatinine ≥1.5 x ULN during the screening period (central laboratory results).
  • Has uncontrolled diabetes (glycosylated haemoglobin (HbA1c) ≥9%, centrally assessed during the Screening period), or has diabetes treated with insulin for less than 6 months prior to study entry.
  • Has any known uncontrolled cardiovascular disease or had any of the following within 6 months of Screening: ventricular or atrial dysrhythmia

    ≥grade 2, bradycardia ≥grade 2, electrocardiogram (ECG) QT interval corrected (QTc) prolonged ≥grade 2, myocardial infarction, severe/unstable angina, symptomatic congestive heart failure, cerebrovascular accident or transient ischemic attack, pulmonary embolism, hypertension not adequately controlled by current medications.

  • Use of any hormone replacement therapy (HRT) with oestrogens.
  • Has symptomatic gallstones/ sludge at the Screening Visit echography (local assessment) OR is asymptomatic but has echography showing clear evidence of impending inflammation such as localised mucosal thickening suggesting the subject is at high risk of developing acute disease. Subjects with asymptomatic gallstones/ sludge and otherwise normal echography may be entered at the discretion of the investigator.
  • Has abnormal findings during the Screening period, any other medical condition(s) or laboratory findings that, in the opinion of the investigator, might jeopardise the subject's safety.
  • Has been treated with any other investigational medicinal product (IMP) prior to the first study visit without undergoing a washout period of seven times the elimination half-life of the investigational compound.
  • Has a known hypersensitivity to any of the test materials or related compounds.
  • Is likely to require treatment during the study with drugs that are not permitted by the study protocol.
  • Has a history of, or known current, problems with alcohol or drug abuse.
  • Has any mental condition rendering him/her unable to understand the nature, scope and possible consequences of the study, and/or evidence of an uncooperative attitude.

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: N/A
  • Interventional Model: Single Group Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: lanreotide PRF
One single dose of lanreotide PRF (via subcutaneous injection) either 180mg or 270mg or 360mg.
Other Names:
  • Lanreotide acetate

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Determination of the Maximum Tolerated Dose (MTD) by Number of Subjects With DLTs.
Time Frame: From Day 1 up to Week 25.
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.
From Day 1 up to Week 25.
PK Analysis of Lanreotide: Maximum Observed Serum Concentration (Cmax).
Time Frame: From Baseline (pre-dose) up to Week 25.

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.

From Baseline (pre-dose) up to Week 25.
PK Analysis of Lanreotide: Time to Reach Maximum Serum Concentration (Tmax).
Time Frame: From Baseline (pre-dose) up to Week 25.

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.

From Baseline (pre-dose) up to Week 25.
PK Analysis of Lanreotide: Apparent Terminal Elimination Half-life (t1/2).
Time Frame: From Baseline (pre-dose) up to Week 25.

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.

From Baseline (pre-dose) up to Week 25.
PK Analysis of Lanreotide: Area Under the Serum Concentration-time Curve From Time 0 to 85 Days (AUC0-85).
Time Frame: From Baseline (pre-dose) up to Day 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.

From Baseline (pre-dose) up to Day 85
PK Analysis of Lanreotide: Area Under the Serum Concentration-time Curve Extrapolated to Infinity (AUC0-∞).
Time Frame: From Baseline (pre-dose) up to Week 25.

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.

From Baseline (pre-dose) up to Week 25.

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Overall Summary of Number of Subjects With AEs.
Time Frame: From Day -42 up to Week 25.
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.
From Day -42 up to Week 25.
PK Analysis of Glycofurol Excipients: Cmax.
Time Frame: From Baseline (pre-dose) up to Day 5.
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.
From Baseline (pre-dose) up to Day 5.
PK Analysis of Glycofurol Excipients: Tmax.
Time Frame: From Baseline (pre-dose) up to Day 5.

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.

From Baseline (pre-dose) up to Day 5.
PK Analysis of Glycofurol Excipients: AUC0-∞ and Area Under the Serum Concentration Time Curve From Time 0 to Last Quantifiable Timepoint (AUC0-t).
Time Frame: From Baseline (pre-dose) up to Day 5.

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.

From Baseline (pre-dose) up to Day 5.
PD Analysis: Mean Change From Baseline in Insulin-like Growth Factor 1 (IGF-1).
Time Frame: From Baseline (pre-dose) up to Week 25.
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.
From Baseline (pre-dose) up to Week 25.
PD Analysis: Mean Change From Baseline in Growth Hormone (GH).
Time Frame: From Baseline (pre-dose) up to Week 13.
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.
From Baseline (pre-dose) up to Week 13.
PD Analysis: Mean Change From Baseline in Free Triiodothyroxine (FT3) and Free Thyroxine (FT4).
Time Frame: From Baseline (pre-dose) up to Week 25.
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.
From Baseline (pre-dose) up to Week 25.
PD Analysis: Mean Change From Baseline in Thyroid Stimulating Hormone (TSH).
Time Frame: From Baseline (pre-dose) up to Week 25.
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.
From Baseline (pre-dose) up to Week 25.
PD Analysis: Mean Change From Baseline in Prolactin.
Time Frame: From Baseline (pre-dose) up to Week 25.
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.
From Baseline (pre-dose) up to Week 25.

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Sponsor

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start

March 1, 2015

Primary Completion (Actual)

November 28, 2017

Study Completion (Actual)

November 28, 2017

Study Registration Dates

First Submitted

March 18, 2015

First Submitted That Met QC Criteria

March 23, 2015

First Posted (Estimate)

March 24, 2015

Study Record Updates

Last Update Posted (Actual)

April 25, 2019

Last Update Submitted That Met QC Criteria

April 24, 2019

Last Verified

April 1, 2019

More Information

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Acromegaly

Clinical Trials on Lanreotide PRF

3
Subscribe