Liraglutide

Liraglutide
Shipped with Ice Packs
In Stock

Description

Chemical Structure and Mechanism of Action

Liraglutide is a synthetic analog of human GLP-1-(7-37), modified by attaching a C16 palmitic acid via a glutamic acid spacer to lysine at position 26. This structural alteration enhances binding to serum albumin, prolonging its half-life to 13 hours (vs. <2 minutes for endogenous GLP-1) .

Key Mechanisms:

  • Insulin Secretion: Stimulates glucose-dependent insulin release from pancreatic β-cells .

  • Glucagon Suppression: Reduces hepatic glucose production by inhibiting glucagon secretion .

  • Gastric Retardation: Delays gastric emptying, reducing postprandial glucose spikes .

  • Satiety Regulation: Acts on central GLP-1 receptors to decrease appetite and food intake .

Clinical Applications

Liraglutide is administered subcutaneously once daily and marketed under distinct brands for different indications:

BrandPrimary UseDosePopulation
VictozaT2D management, cardiovascular risk reduction1.8 mgAdults and children ≥10 years
SaxendaChronic weight management3.0 mgAdults and adolescents ≥12 years
GenericT2D management1.8 mgAdults

Type 2 Diabetes

Liraglutide improves glycemic control, reduces cardiovascular risk, and promotes modest weight loss. Key trial findings:

  • LEAD Trials:

    • LEAD-6: Superior HbA1c reduction vs. exenatide (1.8 mg liraglutide vs. 10 µg exenatide twice daily): −1.1% vs. −0.8% .

    • LEAD-1–5: Consistent efficacy when combined with metformin, sulfonylureas, or insulin, with minimal hypoglycemia risk .

  • Cardiovascular Benefits: Reduces major adverse cardiovascular events (MACE) in high-risk populations by ~13% .

Obesity

Liraglutide 3.0 mg (Saxenda) induces clinically significant weight loss:

  • SCALE Trials:

    • SCALE Obesity: 56-week trial showed 6.2% additional weight loss vs. placebo after a 6% run-in loss .

    • SCALE Diabetes: 56-week trial in T2D patients achieved 4.7% weight loss .

  • Real-World Data:

    • Turkish cohort: 19 kg mean weight loss over 24 weeks .

    • Meta-analyses confirm ≥5% weight loss in 50–81% of participants vs. placebo .

Comparative Efficacy

Liraglutide’s weight-loss efficacy varies by population and dosage:

ComparatorWeight Loss (vs. Liraglutide 3.0 mg)Source
Placebo6–8 kg higher
Exenatide1.8–2.5 kg higher
Semaglutide2–3 kg lower

Real-World Effectiveness

StudyPopulationWeight LossDuration
Turkish CohortObese adults19 kg24 weeks
Spanish CohortT2D patients7.4 kg12 months
Canadian CohortNon-diabetic7.28 kg6 months

Data suggest real-world outcomes may exceed trial results due to improved adherence or comorbidity management .

Limitations and Future Directions

  • Safety Concerns: Long-term thyroid cancer risks remain uncertain .

  • Access Barriers: High cost and injection route limit widespread adoption .

  • Research Gaps: Optimal dosing in pediatric populations and synergies with other anti-obesity agents require study .

Product Specs

Introduction
Liraglutide, a long-acting GLP-1 receptor agonist, shares a 97% amino acid sequence homology with human GLP-1. A key structural difference is the substitution of arginine for lysine at position 34, and the addition of a fatty acid side chain. These modifications increase Liraglutide's resistance to degradation by the enzyme DPP-4, leading to a longer half-life compared to native GLP-1. Liraglutide's mechanism of action involves binding to GLP-1 receptors, which stimulates insulin secretion in a glucose-dependent manner. Additionally, it suppresses glucagon secretion and slows gastric emptying.
Description
Liraglutide peptide is a single-chain, non-glycosylated polypeptide comprised of 31 amino acids. It possesses a molecular mass of 3751 Daltons and a molecular formula of C172H265N43O51.
Physical Appearance
Sterile, white lyophilized powder.
Formulation
The lyophilized protein is provided at a concentration of 1 mg/ml and contains no additives.
Solubility
To reconstitute lyophilized Liraglutide, it is recommended to dissolve the powder in sterile 18 MΩ-cm H2O to a concentration of at least 100 µg/ml. This solution can then be further diluted into other aqueous solutions as needed. It's important to note that Liraglutide is also soluble in a 1% acetic acid solution.
Stability
Lyophilized Liraglutide should be stored between 2-8°C and should not be frozen. After reconstitution, the solution should be stored at 4°C and is stable for up to 7 days. For long-term storage, the reconstituted Liraglutide should be stored below -18°C. Avoid repeated freeze-thaw cycles.
Purity
The purity of this Liraglutide product is greater than 97% as determined by reverse-phase high-performance liquid chromatography (RP-HPLC).
Amino Acid Sequence

H-His-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys(γ-Glu-palmitoyl)-Glu-Phe-Ile-Ala-Trp-Leu-Val-Arg-Gly-Arg-Gly-OH

Q&A

How does liraglutide's molecular structure compare to native GLP-1, and what are the pharmacokinetic implications?

Liraglutide was specifically engineered to overcome the limited therapeutic potential of native GLP-1, which has a half-life of less than 2 minutes due to rapid degradation by dipeptidyl peptidase-IV (DPP-IV) and neutral endopeptidases (NEP). The molecular modification includes substituting arginine for lysine at position 34 in the GLP-1 peptide and adding a palmitic acid chain with a glutamic acid spacer on the lysine residue at position 26. These structural changes result in significant pharmacokinetic improvements, creating a compound with 98% protein binding (due to the fatty acid chain) and an extended half-life of approximately 13 hours in both healthy individuals and those with type 2 diabetes. Following subcutaneous injection, peak absorption occurs at 11 hours with an absolute bioavailability of 55%, enabling once-daily administration rather than the continuous infusion that would be required with native GLP-1 .

What experimental protocols are typically employed to assess liraglutide efficacy in clinical research?

Clinical investigations of liraglutide typically follow a structured experimental design that includes:

  • Initial screening and run-in periods: Studies often incorporate a 2-week single-blind placebo run-in period to establish baseline measurements and exclude placebo responders.

  • Dose titration phase: A 4-week dose titration period is common, with doses typically starting at 0.6 mg/day and increasing by 0.6 mg weekly until reaching the target dose (often 1.2-3.0 mg/day).

  • Maintenance phase: Following titration, a constant dose period (typically 16 weeks or longer) allows for assessment of sustained efficacy.

  • Lifestyle intervention standardization: Most protocols prescribe standardized lifestyle interventions across all treatment arms, including specified caloric deficits (commonly 500 kcal/day based on estimated 24-hour energy expenditure) and physical activity guidelines.

  • Comprehensive endpoint assessment: Primary efficacy endpoints typically include weight change from baseline, glycemic parameters (HbA1c, FPG, PPG), and cardiometabolic risk factors .

This methodical approach enables researchers to isolate drug effects from lifestyle intervention effects and assess both short-term and sustained responses.

How were the phase III clinical trials for liraglutide structured, and what were the key methodological strengths?

The liraglutide phase III clinical development program, primarily encompassed by the Liraglutide Effect and Action in Diabetes (LEAD) trials, was strategically designed to comprehensively evaluate efficacy and safety across the treatment continuum of T2D. These multinational, randomized controlled trials incorporated several methodological strengths:

  • Diverse patient populations: The trials included a large number and diverse range of patients across the diabetes treatment spectrum, enhancing generalizability.

  • Active comparator design: Rather than relying solely on placebo comparisons, the studies included active comparisons against common antidiabetic agents including metformin, rosiglitazone, glimepiride, insulin glargine, exenatide, and sitagliptin, enabling direct assessment of clinical differences between therapeutic options.

  • Long-term follow-up: Extension studies (up to 2 years) were conducted to assess durability of effect and long-term safety profiles.

  • Comprehensive endpoint assessment: Beyond glycemic control, the trials assessed multiple clinically relevant outcomes including weight changes, cardiometabolic risk factors, and quality of life measures .

This methodologically robust approach provided clinically meaningful evidence regarding liraglutide's place in the therapeutic landscape and helped inform subsequent clinical practice guidelines.

What inclusion/exclusion criteria are critical when designing liraglutide studies?

Based on established protocols, researchers should consider the following carefully calibrated inclusion/exclusion criteria when designing liraglutide studies:

Key inclusion criteria:

  • Adults (typically 18-75 years) with confirmed T2DM and/or obesity (BMI thresholds commonly ≥30 kg/m² or ≥27 kg/m² with weight-related comorbidities)

  • For diabetes studies: Specific HbA1c ranges (e.g., 7.0-10.0%)

  • For weight management studies: Often stratified by presence/absence of prediabetes

Critical exclusion criteria:

  • Type 1 diabetes

  • Recent significant weight change (±5% within previous 3 months)

  • Prior use of GLP-1 receptor agonists

  • History of pancreatitis (idiopathic, acute, or chronic)

  • Severe psychiatric illness, including major depressive disorder

  • Clinically significant cardiovascular disease

  • Use of medications known to affect weight

  • History of bariatric surgery

  • Significant organ dysfunction or malignancy

Careful application of these criteria helps isolate the effects of liraglutide from confounding factors and ensures participant safety while maintaining clinical relevance.

How do researchers define and measure "response" to liraglutide treatment?

The definition of "response" to liraglutide varies based on the primary indication being studied, but established methodological approaches include:

In type 2 diabetes research:

  • Primary response measures typically include reductions in HbA1c (with thresholds of ≥0.5% or ≥1.0% reduction, or achievement of target HbA1c <7.0%)

  • Secondary response metrics include improvements in fasting and postprandial glucose levels, HOMA-IR (insulin resistance), and HOMA-B (beta-cell function)

In weight management research:

  • Responders are often defined as achieving ≥5% reduction in baseline body weight

  • Alternative definitions include ≥10% weight reduction or achievement of predefined cardiometabolic risk reduction targets

Composite endpoints may also be used, requiring both glycemic improvement and weight reduction to classify a patient as a responder. In the study cited, researchers classified responders as patients achieving HbA1c reduction ≥1% and weight reduction ≥5% from baseline after 6 months of treatment .

What predictive factors have been identified for liraglutide treatment response?

Recent research has identified several baseline characteristics that may predict favorable response to liraglutide treatment. A retrospective study of 206 patients with T2DM and metabolic syndrome found significant differences between responders and non-responders:

ParameterRespondersNon-respondersStatistical significance
Baseline HbA1cHigherLowerp < 0.05
Baseline PPGHigherLowerp < 0.05
Baseline body weightHigherLowerp < 0.05
Baseline BMIHigherLowerp < 0.05

Interestingly, by 6 months of treatment, the differences in body weight and BMI between responders and non-responders had disappeared, while HbA1c and PPG levels were significantly lower in responders compared to non-responders. These findings suggest that patients with poorer baseline glycemic control and higher initial body weight may experience more pronounced benefits from liraglutide therapy .

The identification of such predictive factors allows for more personalized application of liraglutide therapy and enables researchers to stratify populations in future clinical trials.

How do side effects impact treatment adherence and efficacy outcomes in liraglutide research?

Contrary to conventional expectations, research indicates that liraglutide's side effects do not necessarily compromise treatment efficacy. A retrospective study examining this relationship found that among patients experiencing side effects who continued treatment (MAIN_SE(+) group), weight reduction was actually greater (-7.9 ± 0.9%) compared to those without side effects who continued treatment (MAIN_SE(-) group, -5.9 ± 0.6%) at the 3-month timepoint .

These findings highlight the importance of methodologically distinguishing between efficacy in adherent populations versus effectiveness in real-world settings. Researchers should implement strategies to enhance adherence, such as graduated dose escalation protocols, proactive side effect management, and frequent follow-up during initial treatment phases.

What methodological approaches are being used to develop oral formulations of liraglutide?

Current injectable formulations of liraglutide face limitations in patient acceptance and adherence, driving research into oral delivery systems. A notable methodological approach involves the DehydraTECH processing technology, which has shown promising results in preclinical models:

  • Animal model validation: In a 12-week rodent study with ad libitum feeding, orally administered DehydraTECH-processed liraglutide demonstrated a 5.88% weight reduction and 11.54% blood sugar reduction compared to baseline. Notably, this outperformed the oral semaglutide control (Rybelsus®), which resulted in a 5.65% increase in body weight and only 0.41% blood sugar reduction .

  • Transition to human studies: Based on these findings, researchers have designed a human pilot study (designated GLP-1-H25-5) with 8-10 healthy volunteers to assess safety and pharmacokinetic performance of the oral DehydraTECH-liraglutide formulation. The study will compare the oral formulation against Saxenda® injectable liraglutide .

  • Staged research approach: The research process follows a staged approach, using positive animal study data to inform human study design, with initial pilot studies in healthy volunteers potentially leading to Phase I registered trials if results are promising .

This methodical research progression from animal models to human studies represents a standard approach for developing novel drug delivery systems, with careful attention to comparative efficacy against existing formulations.

What are the key challenges in long-term efficacy assessment of liraglutide?

Long-term efficacy assessment of liraglutide presents several methodological challenges that researchers must address:

  • Treatment persistence: Real-world studies show that continuance rates with liraglutide are not high, with data indicating only about 40.8% of patients remain on treatment at 6 months. This creates challenges in assessing true long-term efficacy versus effectiveness .

  • Weight regain dynamics: Evidence suggests that weight may gradually increase after the initial 3-month treatment period, even in those continuing therapy. Designing studies to capture these temporal dynamics requires extended observation periods and appropriate analytical approaches to account for non-linear response patterns .

  • Differentiating pharmacological from behavioral effects: As treatment continues, disentangling the direct pharmacological effects from behavioral adaptations becomes increasingly complex. Methodological approaches might include mechanistic sub-studies examining appetite hormones, energy expenditure, and food preference changes over time.

  • Adaptive dosing considerations: Longer-term studies must address whether dose adjustments over time influence efficacy, particularly as patients experience physiological adaptations to treatment.

Future research should incorporate adaptive trial designs, frequent assessment timepoints beyond the typical 6-month endpoint, and comprehensive mechanistic assessments to better characterize the durability of liraglutide's effects.

How might combination therapies with liraglutide be systematically evaluated?

As the field advances, systematic evaluation of liraglutide combination therapies requires rigorous methodological approaches:

  • Mechanistic rationale assessment: Combinations should be evaluated based on complementary mechanisms of action. For example, combining liraglutide with agents targeting different pathways in glucose regulation or appetite control might provide synergistic effects.

  • Factorial study designs: The gold standard approach employs 2×2 factorial designs that allow assessment of individual agents alone and in combination, enabling determination of additive versus synergistic effects. This design requires larger sample sizes but provides the most comprehensive data on interaction effects.

  • Sequential add-on protocols: Alternative designs include sequential add-on protocols where liraglutide is added to stable background therapy or vice versa, with appropriate wash-in periods to establish baseline effects of the initial therapy.

  • Outcome prioritization: Studies should prespecify whether glycemic control, weight reduction, or composite cardiometabolic outcomes serve as the primary endpoints, as different combinations may differentially affect these parameters.

  • Pharmacokinetic/pharmacodynamic interaction assessment: Early-phase studies should assess potential PK/PD interactions between liraglutide and combination agents to inform appropriate dosing in larger efficacy trials.

The LEAD clinical trial program provides a model for such systematic evaluation, having examined liraglutide in combination with various antidiabetic agents across the treatment continuum .

How should researchers interpret discrepancies between clinical trial results and real-world effectiveness of liraglutide?

The translation of liraglutide research findings into clinical practice requires careful consideration of several methodological factors:

  • Selection bias effects: Clinical trials typically enroll motivated participants meeting strict eligibility criteria, potentially overestimating real-world effectiveness. Research comparing trial populations with typical clinical populations can help quantify this effect.

  • Adherence differences: Real-world studies show substantially lower persistence rates (approximately 40.8% at 6 months) compared to clinical trials. Understanding the drivers of non-adherence beyond side effects is critical, as data indicate that 40.1% of patients discontinue liraglutide despite not reporting adverse events .

  • Support infrastructure disparities: Clinical trials provide frequent monitoring, counseling, and structured lifestyle interventions that may not be replicated in routine care. Pragmatic trial designs that more closely mirror real-world conditions can provide more applicable efficacy estimates.

  • Effectiveness-efficacy gap quantification: Systematic efforts to quantify the gap between efficacy in trials and effectiveness in practice can help establish realistic expectations and identify implementation strategies to narrow this gap.

These considerations highlight the importance of complementing traditional randomized controlled trials with pragmatic studies, observational research, and implementation science approaches to develop a comprehensive understanding of liraglutide's role in clinical practice.

What methodological advances are needed to better individualize liraglutide therapy?

Advancing personalized approaches to liraglutide therapy requires several methodological innovations:

  • Biomarker development: Identification and validation of predictive biomarkers beyond baseline clinical characteristics could enhance patient selection. Research suggests that baseline HbA1c, PPG, body weight, and BMI may help predict response, but more precise molecular or genetic markers are needed .

  • Machine learning approaches: Application of advanced analytics to large datasets combining clinical, genetic, metabolomic, and microbiomic data could identify complex patterns predicting response that are not apparent with traditional statistical methods.

  • N-of-1 trial designs: For selected patients, structured N-of-1 trials with crossover between liraglutide and alternative therapies could help determine individual response patterns and optimize treatment selection.

  • Response-adaptive randomization: Future trials could employ response-adaptive designs that allow for treatment adjustments based on early response indicators, potentially identifying factors associated with rapid versus delayed therapeutic effects.

Product Science Overview

Mechanism of Action

Liraglutide works by increasing the amount of insulin produced by the pancreas when blood sugar levels are high. It also reduces the amount of glucagon, a hormone that increases blood sugar levels, produced by the pancreas . Additionally, liraglutide slows down the emptying of the stomach, which helps to reduce appetite and food intake .

Clinical Uses
  1. Type 2 Diabetes Mellitus: Liraglutide is used as a second-line therapy for type 2 diabetes, particularly when first-line treatments like metformin are not sufficient . It helps to control blood sugar levels and has been shown to reduce the risk of cardiovascular complications associated with diabetes .

  2. Chronic Obesity: Under the brand name Saxenda, liraglutide is used for weight management in individuals with obesity. It helps to reduce appetite and increase feelings of fullness, leading to reduced food intake and weight loss .

Administration

Liraglutide is administered via subcutaneous injection, typically once daily . The dosage may vary depending on the specific condition being treated and the patient’s response to the medication.

Side Effects

Common side effects of liraglutide include nausea, vomiting, diarrhea, and constipation. Some patients may also experience headaches, dizziness, and reactions at the injection site . Serious side effects, although rare, can include pancreatitis, kidney problems, and allergic reactions .

Industrial Production

Liraglutide is synthesized using recombinant DNA technology. The production process involves the insertion of the gene encoding the GLP-1 analog into a host organism, typically Escherichia coli or Saccharomyces cerevisiae. The host organism then produces the GLP-1 analog, which is subsequently purified and formulated into the final product .

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.