Lanreotide

Lanreotide
Shipped with Ice Packs
In Stock

Description

Pharmacological Profile

Molecular structure: C₅₄H₆₉N₁₁O₁₀S₂ (molecular weight: 1,096.33 g/mol) .
Mechanism: Binds somatostatin receptors (SSTR2 > SSTR5) with 78% protein binding, inhibiting growth hormone (GH), insulin, glucagon, and serotonin secretion .

Pharmacokinetic ParameterValue
Bioavailability80% (subcutaneous)
Half-life22 days (extended release)
Clearance23.1 L/h
ExcretionPrimarily biliary

Self-assembling nanotube formation (24.4 nm diameter) enables sustained drug release .

Approved Indications:

  1. Acromegaly: Reduces GH levels <2.5 µg/L in 63% of patients at 12 months .

  2. GEP-NETs:

    • Prolongs progression-free survival (PFS) to 32.8 months vs. 18 months with placebo .

    • Controls carcinoid syndrome symptoms (diarrhea/flushing) with 65% reduction in rescue octreotide use .

Off-Label Uses:

  • Thyroid-stimulating hormone-secreting pituitary adenomas .

CLARINET Program (Phase III Core + OLE Studies) :

ParameterLanreotidePlacebo
Median PFS (Pancreatic NETs)30.8 months14.0 months*
24-month PFS Rate65.1%33.0%
Time to Symptom ProgressionNot reached6.1 months

*After crossover to lanreotide in OLE .

Real-World Outcomes (n=69) :

  • Median treatment duration: 21.6 months

  • First-line usage: 87% of patients

  • Dose escalation (120 mg/21 days): 10% of cases

Dosage and Administration

Standard Regimen:

  • Somatuline Autogel: 120 mg subcutaneous every 28 days

  • Sustained Release: 90 mg IM every 14 days

Dose Adjustments:

  • Acromegaly: Titrate based on GH/IGF-1 levels

  • Tumor progression: Escalate to 120 mg every 21 days

Emerging Research Directions

  1. Combination Therapies:

    • Paired with everolimus in PRIME trial (NCT04032327) for pancreatic NETs .

  2. High-Dose Protocols:

    • CLARINET FORTE showed 120 mg/14 days improved disease control in 44% of progressing patients .

  3. Diagnostic Synergy:

    • Enhanced SSTR-PET imaging correlation (92% receptor positivity) .

Product Specs

Introduction
Lanreotide is a potent synthetic analog of somatostatin, exhibiting a longer duration of action. Its binding affinity to peripheral somatostatin receptors makes it effective in regulating hormone secretion, particularly growth hormone. This targeted action finds clinical significance in managing conditions like acromegaly. Additionally, Lanreotide has shown efficacy in normalizing thyroid function in some patients with TSH-secreting adenomas, though without significantly impacting tumor size. Its inhibitory effects extend to intestinal exocrine secretions, making it a versatile therapeutic agent.
Description
Lanreotide is a synthetically produced octapeptide, structurally similar to the naturally occurring hormone somatostatin.
Physical Appearance
White, sterile, lyophilized powder.
Formulation
Each vial contains 1 mg of lyophilized Lanreotide, formulated with 5 mg of mannitol and 0.04 mg of Tween-80 for optimal reconstitution and stability.
Solubility
To reconstitute, dissolve the lyophilized Lanreotide in sterile 18 MΩ-cm water to a concentration of at least 100 µg/ml. This solution can be further diluted with other aqueous solutions as needed.
Stability
While lyophilized Lanreotide remains stable at room temperature for up to 3 weeks, it is recommended to store it desiccated below -18°C. After reconstitution, Lanreotide should be stored at 4°C for a maximum of 2-7 days. For long-term storage, adding a carrier protein like 0.1% HSA or BSA is recommended. Avoid repeated freeze-thaw cycles to maintain product integrity.
Purity
The purity of Lanreotide is determined using rigorous analytical techniques including RP-HPLC and SDS-PAGE, ensuring a purity exceeding 99.0%.

Q&A

What is Lanreotide and how does it exert its therapeutic effects in neuroendocrine tumors?

Lanreotide is a synthetic long-acting somatostatin analog that functions as a somatostatin receptor agonist. It binds primarily to somatostatin receptor subtypes 2 and 5 expressed on neuroendocrine tumor cells, inhibiting cell proliferation and hormone secretion .

The drug works by decreasing specific natural substances produced by the body, including growth hormone and various peptide hormones secreted by neuroendocrine tumors. This dual action mechanism provides both anti-proliferative effects and symptom control, particularly in functional NETs producing hormonal syndromes .

In clinical studies, Lanreotide has demonstrated significant anti-proliferative properties in grade 1 and 2 NETs originating from various tumor sites, making it a valuable first-line treatment option . Its mechanism supports both tumor control and symptomatic management in appropriate patient populations.

What are the standard dosing regimens for Lanreotide in research settings?

The standard dosing regimen for Lanreotide in research protocols is 120 mg administered subcutaneously every 28 days, which has been consistently used across clinical trials and real-world studies . This dosing schedule establishes the foundation for most research investigations.

Most clinical protocols begin with this standard dose, as evidenced by data showing 96% of patients receive this regimen initially . The formulation used is typically Lanreotide autogel/depot, which provides sustained release over the dosing interval .

CharacteristicFrequency (n=69)
Starting dose: 120 mg66 (96%)
Standard administration scheduleEvery 28 days
Dose escalation to 120 mg every 21 days7 (10%)
Primary indications for dose escalation:
- Tumor progression5 (7%)
- Symptom control1 (1%)
- Undocumented reason1 (1%)

Table 1: Lanreotide dosing characteristics from real-world clinical usage data

Alternative dosing strategies, particularly dose escalation to 120 mg every 21 days, occur in approximately 10% of patients, primarily in response to disease progression (72% of escalation cases) or inadequate symptom control (14% of escalation cases) . The CLARINET FORTE trial has validated this escalation approach as well-tolerated in selected patients .

What methodological approaches are most appropriate for evaluating efficacy in Lanreotide clinical studies?

Evaluating Lanreotide efficacy requires a multi-faceted approach that accounts for both the cytostatic nature of treatment and potential hormonal control. Recommended methodological approaches include:

Tumor Response Assessment:

  • RECIST 1.1 criteria as the primary standardized evaluation method

  • Clinical benefit rate (CBR), defined as complete response, partial response, or stable disease over a specified timeframe (typically 24 weeks)

  • Progressive disease documentation using consistent imaging protocols

Progression Metrics:

  • Progression-free survival (PFS) as a primary endpoint, with clear definitions of progression and censoring rules

  • Time on treatment analysis, with median duration calculations and proper handling of patients continuing treatment post-study

Dynamic Growth Assessment:

  • Tumor growth rate (TGR) calculations using sequential imaging, comparing pre-treatment growth rates with on-treatment rates

  • Measurement of changes in sum of target lesion diameters as a continuous variable

In a Japanese study of Lanreotide, this multi-modal assessment demonstrated that while the partial response rate was modest (7.1%), disease stabilization was achieved in 71.4% of patients . More significantly, the mean tumor growth rate decreased from 25.3% (±35.7%) per month pre-treatment to 6.4% (±9.6%) per month during treatment, illustrating Lanreotide's cytostatic effect .

How should researchers approach studying the effects of Lanreotide on hormone-related symptoms?

Studying Lanreotide's effect on hormone-related symptoms requires systematic assessment methods that capture both subjective symptomatic improvement and objective biochemical changes. Effective methodological approaches include:

Symptom Quantification:

  • Implementation of validated symptom scales specific to the relevant hormonal syndrome

  • Daily patient diaries documenting frequency and severity of symptoms such as diarrhea and flushing

  • Quality of life instruments with modules specific to neuroendocrine symptoms

Biochemical Monitoring:

  • Serial measurement of specific biomarkers relevant to the tumor type

  • Standardized collection protocols with attention to timing relative to Lanreotide administration

  • Correlation analyses between biochemical markers and symptomatic improvement

Response Definitions:

  • Clear pre-specified criteria for symptom response (e.g., >50% reduction in diarrhea episodes)

  • Integrated response assessments combining both biochemical and symptomatic parameters

  • Documentation of concurrent medications that might influence symptom control

Studies have demonstrated that Lanreotide is effective for both tumor control and symptom management, with approximately 49% of patients receiving the medication for this dual purpose and 4% for symptom control alone . The PRELUDE study specifically examined Lanreotide's effectiveness in managing diarrhea and flushing in the context of carcinoid syndrome .

What statistical considerations are essential when designing Lanreotide studies for rare tumor types?

Neuroendocrine tumors are relatively rare, presenting statistical challenges that require specialized approaches:

Sample Size Considerations:

  • Power calculations based on realistic effect sizes derived from prior studies

  • Consideration of adaptive designs that can accommodate smaller patient populations

  • Clear justification of assumptions used in statistical planning

Endpoint Selection:

  • Prioritization of endpoints that can be robustly assessed with limited sample sizes

  • Use of surrogate endpoints with established correlation to clinical outcomes

  • Composite endpoints that capture multiple aspects of treatment benefit

Analytical Strategies:

  • Bayesian methods that can incorporate prior information to enhance precision

  • Planned subgroup analyses with appropriate stratification factors

  • Multiple sensitivity analyses to evaluate robustness of findings

Data Monitoring:

  • Interim analyses with clear stopping rules to protect against uninformative study completion

  • Comprehensive strategies for handling missing data, which is particularly problematic in small studies

  • Central review of imaging to minimize variability

In the Japanese phase II trial of Lanreotide, these considerations were implemented with a sample size of 32 patients, which allowed meaningful assessment of the clinical benefit rate (primary endpoint, 64.3% at 24 weeks) despite the limited population .

How can researchers effectively design studies to evaluate Lanreotide in combination with Peptide Receptor Radionuclide Therapy (PRRT)?

The combination of Lanreotide with PRRT represents an advanced research area requiring specialized methodological approaches:

Study Design Elements:

  • Clear patient selection criteria focusing on progressive metastatic or locally advanced grade 1 or 2 GEP- or lung-NETs

  • Strategic timing of combination therapy initiation based on disease trajectory

  • Detailed protocols for administration of both modalities, including interval between treatments

Efficacy Assessment Framework:

  • Primary endpoint of progression-free survival with detailed imaging protocols

  • Secondary endpoints including objective response rate, disease control rate, and symptom improvement

  • Assessment timepoints at baseline, end of last PRRT cycle, and long-term follow-up

Safety Monitoring Requirements:

  • Comprehensive evaluation of additive or synergistic toxicities

  • Special attention to renal function, hematological parameters, and hepatic function

  • Grading of adverse events using standardized criteria (CTCAE)

Specialized Analytics:

  • Comparison of pre-treatment and on-treatment tumor growth rates

  • Exploratory analyses to identify predictive biomarkers for combination response

  • Assessment of somatostatin receptor expression before and during treatment

The PRELUDE study demonstrated this approach, evaluating Lanreotide combined with 177Lu-DOTATOC or 177Lu-DOTATATE in progressive NETs, with carefully specified assessment timepoints and endpoints .

What are the methodological challenges in studying tumor growth rate (TGR) as an efficacy endpoint for Lanreotide?

Tumor growth rate represents an advanced metric that can detect subtle treatment effects in slow-growing NETs, but presents several methodological challenges:

Standardization Requirements:

  • Consistent imaging protocols at precisely defined intervals

  • Minimum of three timepoints (pre-treatment, early treatment, and later follow-up)

  • Standardized measurement techniques across all imaging timepoints

Calculation Methodology:

  • Clear mathematical formula for TGR determination

  • Accounting for variable intervals between scans

  • Handling of lesions that become unmeasurable during treatment

Interpretation Framework:

  • Establishment of clinically significant thresholds for TGR changes

  • Correlation with traditional RECIST outcomes for contextual understanding

  • Analysis by tumor subtype to account for heterogeneity in natural growth patterns

Statistical Considerations:

  • Handling of missing data points that may compromise TGR calculations

  • Methods for determining optimal pre-treatment TGR cutoffs that predict treatment response

  • Appropriate statistical tests for comparing pre- and on-treatment TGRs

Research has demonstrated that mean tumor growth rate can decrease dramatically during Lanreotide treatment (from 25.3%/month pre-treatment to 6.4%/month during treatment) , providing a sensitive measure of drug effect that may not be captured by categorical RECIST assessment.

What research approaches can elucidate predictive biomarkers for Lanreotide response?

Identifying predictive biomarkers for treatment response remains a significant research challenge requiring methodological rigor:

Biospecimen Collection Protocol:

  • Standardized collection of tissue and blood samples at defined timepoints

  • Processing methods that preserve protein and nucleic acid integrity

  • Centralized biobanking with quality control measures

Biomarker Candidate Selection:

  • Evaluation of somatostatin receptor density and subtype distribution

  • Assessment of tumor proliferation markers (Ki-67, mitotic index)

  • Exploration of circulating biomarkers including CgA, NSE, and specific hormones

Analytical Approaches:

  • Integration of genomic, transcriptomic, and proteomic data

  • Correlation analyses between biomarker levels and clinical outcomes

  • Machine learning algorithms to identify complex biomarker signatures

Validation Requirements:

  • Internal validation using training and testing cohorts

  • External validation in independent patient populations

  • Analytical validation of biomarker assays for reproducibility

While specific predictive biomarkers for Lanreotide response are still being investigated, research suggests that pre-treatment tumor growth rate may serve as a predictive factor, with post-hoc analyses determining optimal TGR cutoffs that best predict objective response rates during treatment .

What methodological approaches are recommended for studying the continuation of Lanreotide beyond radiological progression?

The question of continuing Lanreotide beyond radiological progression is clinically relevant and requires specific research methodologies:

Protocol Design Elements:

  • Clear definition of progression that would trigger decision-making

  • Pre-specified criteria for post-progression continuation

  • Systematic documentation of rationale for treatment continuation decisions

Assessment Framework:

  • Continued regular imaging to track post-progression growth kinetics

  • Monitoring of symptom control in functional tumors

  • Quality of life assessments to capture patient-perceived benefit

Analytical Considerations:

  • Time-to-subsequent-progression as a potential endpoint

  • Growth rate comparisons pre-progression versus post-progression on continued therapy

  • Subgroup analyses based on progression pattern (new lesions versus growth of existing lesions)

Real-world data has shown that Lanreotide is continued beyond progression in approximately 16% of patients . Among these, functional disease was present in 9% of cases, while other reasons for continuation included frailty, patient preference, contraindications to alternative therapies, and the favorable safety profile of Lanreotide .

What are the key considerations for designing long-term safety studies of Lanreotide?

Long-term safety assessment requires methodological approaches that address the challenges of extended observation periods:

Study Duration Planning:

  • Extension studies with sufficient duration to detect late-emerging adverse events

  • Median exposure times in successful studies ranging from approximately 53 to 151 weeks

  • Clear protocols for continued safety monitoring after treatment discontinuation

Comprehensive Safety Assessment:

  • Systematic collection of adverse events using standardized terminology (MedDRA)

  • Laboratory monitoring with attention to gallbladder/biliary function, glucose metabolism, and thyroid function

  • Specific monitoring for events that may emerge only after prolonged exposure

Special Population Considerations:

  • Tailored monitoring for patients with renal or hepatic impairment

  • Age-specific safety assessments for elderly populations

  • Interaction monitoring for patients on multiple concomitant medications

Analysis and Reporting:

  • Time-to-event analyses for adverse events of special interest

  • Cumulative incidence calculations with extended time horizons

  • Comparison of early versus late adverse event profiles

Long-term safety data from a Japanese extension study revealed that new adverse drug reactions can emerge beyond 48 weeks of treatment, including upper abdominal pain and urticaria . The study also identified serious biliary events (bile duct stones) as potential concerns requiring monitoring during extended treatment .

How should researchers evaluate Lanreotide dose escalation strategies in progressive disease?

Evaluating dose escalation strategies requires methodological rigor to determine efficacy, safety, and appropriate patient selection:

Research Protocol Elements:

  • Clear criteria triggering consideration of dose escalation

  • Standardized escalation schedule (typically 120 mg every 21 days)

  • Pre-specified assessments before and after dose modification

Efficacy Assessment:

  • Re-establishment of baseline measurements before escalation

  • Consistent post-escalation evaluation timepoints

  • Comparison of pre- and post-escalation tumor growth kinetics

Safety Monitoring:

  • Dose-dependent adverse event assessment

  • Pharmacokinetic sampling at standard and escalated dosing

  • Quality of life measures to capture patient tolerance

Analysis Strategy:

  • Identification of patient characteristics associated with escalation benefit

  • Time-to-progression calculations from escalation date

  • Cost-effectiveness evaluations given increased drug utilization

Real-world data indicates that dose escalation to 120 mg every 21 days occurs in approximately 10% of patients, primarily for tumor progression (7%) or symptom control (1%) . The CLARINET FORTE trial has provided evidence supporting this dose escalation strategy as well-tolerated , though optimal patient selection criteria continue to be refined.

What are the recommended approaches for standardizing imaging protocols in Lanreotide trials?

Imaging standardization is critical for reliable efficacy assessment in Lanreotide studies:

Modality Selection and Protocol Specification:

  • Contrast-enhanced CT or MRI as primary assessment tools

  • Standardized slice thickness, contrast timing, and acquisition parameters

  • Functional imaging (somatostatin receptor imaging) as complementary assessment

Timing Considerations:

  • Baseline imaging within 4 weeks of treatment initiation

  • Regular follow-up at consistent intervals (typically every 12-24 weeks)

  • Additional imaging upon clinical suspicion of progression

Measurement Technique Standardization:

  • RECIST 1.1 as the primary measurement framework

  • Consistent selection of target and non-target lesions

  • Additional volumetric assessments for selected studies

Quality Control Measures:

  • Central independent radiological review to minimize reader variability

  • Blinded assessment when comparing treatment strategies

  • Reader training and qualification requirements

In research settings, these standardized approaches have enabled reliable assessment of tumor response, with one study showing a mean reduction of 5.5% in the sum of diameters of target lesions during Lanreotide treatment .

What methodological approaches are optimal for studying the pharmacokinetic-pharmacodynamic relationship of Lanreotide?

Understanding the relationship between Lanreotide pharmacokinetics and its clinical effects requires specialized research approaches:

PK Sampling Strategy:

  • Pre-dose trough levels immediately before next injection

  • Early post-injection sampling (2-8 hours) to capture peak concentration

  • Limited sampling strategy across the dosing interval

  • Steady-state assessment after 4-5 injections

PD Assessment Correlation:

  • Simultaneous measurement of relevant biomarkers

  • Temporal correlation between drug levels and symptom control

  • Integration with imaging timepoints for tumor response correlation

Analytical Methods:

  • Population PK/PD modeling to characterize variability

  • Exposure-response analyses for efficacy and safety endpoints

  • Evaluation of covariates influencing PK/PD relationships

Special Considerations:

  • Assessment of dose proportionality between standard and escalated regimens

  • Evaluation of accumulation with repeated dosing

  • Investigation of potential immunogenicity affecting drug levels

These approaches allow researchers to investigate questions such as whether trough Lanreotide concentrations correlate with tumor response or symptom control, and whether dose escalation provides proportional increases in drug exposure.

How can researchers effectively capture patient-reported outcomes in Lanreotide clinical trials?

Patient-reported outcomes (PROs) provide essential data on symptomatic benefit and quality of life during Lanreotide treatment:

Instrument Selection:

  • NET-specific validated questionnaires (e.g., EORTC QLQ-GINET21)

  • General cancer instruments (e.g., EORTC QLQ-C30)

  • Symptom-specific scales for functional NETs (diarrhea, flushing assessment)

Implementation Framework:

  • Electronic collection methods to enhance compliance and data quality

  • Consistent assessment schedule (baseline, regular on-treatment, end-of-treatment)

  • Clear protocols for handling missing data

Domain Coverage Requirements:

  • Physical symptoms (diarrhea, flushing, pain, fatigue)

  • Emotional functioning (anxiety, depression)

  • Social functioning and role performance

  • Treatment-related burden and side effects

Analysis Approaches:

  • Mixed effects models for longitudinal data

  • Responder analyses using minimal clinically important differences

  • Time-to-deterioration analyses for key symptomatic endpoints

Studies have shown that Lanreotide treatment impacts both tumor control and symptom management, with approximately 49% of patients receiving the medication for this dual purpose . Systematic PRO assessment allows quantification of this symptomatic benefit from the patient perspective.

Product Science Overview

Development and Approval

Lanreotide was developed by Ipsen Pharmaceuticals and is marketed under the brand name Somatuline . It was first approved for use in the United States by the Food and Drug Administration (FDA) on August 30, 2007 . The drug is also approved in several other countries, including the United Kingdom, Australia, and Canada .

Mechanism of Action

Lanreotide works by mimicking the action of somatostatin, thereby inhibiting the secretion of growth hormone and other hormones . This inhibition helps manage the symptoms of acromegaly and neuroendocrine tumors. The drug is available in a long-acting formulation, which allows for less frequent dosing compared to other treatments .

Medical Uses

Lanreotide is indicated for:

  • The long-term treatment of patients with acromegaly who have had an inadequate response to, or cannot be treated with, surgery and/or radiotherapy .
  • The treatment of adult patients with unresectable, well- or moderately-differentiated, locally advanced or metastatic gastroenteropancreatic neuroendocrine tumors to improve progression-free survival .
  • The treatment of adults with carcinoid syndrome to reduce the frequency of short-acting somatostatin analog rescue therapy .
Pharmacokinetics

Lanreotide has a bioavailability of approximately 80% and a protein binding rate of 78% . It is metabolized in the gastrointestinal tract and has an elimination half-life of 2 hours for the immediate-release formulation and 5 days for the sustained-release formulation . The drug is primarily excreted through the bile duct .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.