186018-45-1
Congenital leptin deficiency
Metreleptin is a synthetic, recombinant analog of the human hormone leptin, engineered as a targeted replacement therapy for rare and severe metabolic disorders stemming from leptin deficiency.[1] Marketed under the brand names Myalept® and Myalepta®, it represents a significant therapeutic advance for patients with lipodystrophy syndromes, a group of diseases characterized by a pathological loss of adipose tissue and subsequent metabolic derangements.[3] Its primary approved indication is as an adjunct to diet for treating the complications of leptin deficiency in patients with congenital or acquired generalized lipodystrophy (GL).[1]
The drug functions as a direct agonist of the human leptin receptor (ObR), mimicking the action of the absent endogenous hormone to restore critical metabolic signaling pathways.[6] Clinical evidence has robustly demonstrated its efficacy in improving glycemic control, reducing severe hypertriglyceridemia, and mitigating hepatic steatosis in patients with GL.[6] However, the therapeutic landscape is complex. Regulatory approvals differ globally, with agencies in the European Union and Canada extending the indication to include certain patients with partial lipodystrophy (PL), a distinction not made by the U.S. Food and Drug Administration (FDA).[3]
The use of metreleptin is governed by significant safety considerations, most notably a U.S. Boxed Warning regarding the risks of developing neutralizing anti-drug antibodies and a potential association with T-cell lymphoma.[6] The high potential for immunogenicity can lead to a loss of efficacy and an increased risk of severe infections. To manage these risks, metreleptin is available in the U.S. exclusively through a restricted Risk Evaluation and Mitigation Strategy (REMS) program, which mandates prescriber certification and ensures appropriate patient selection.[12] This report provides a comprehensive analysis of metreleptin, detailing its molecular characteristics, pharmacology, clinical efficacy, global regulatory status, and safety profile.
Metreleptin is the internationally recognized generic name (International Nonproprietary Name, INN) for this biotech therapeutic.[6] It is commercially distributed under the brand names Myalept® and Myalepta®.[3] Chemically, it is identified by several synonyms that reflect its structure and origin, including N-Methionylleptin and recombinant methionyl-human leptin (r-metHuLeptin).[3] In Japan, it may be referred to as Mettreleptin (genetical recombination).[3] The compound is uniquely identified across scientific and regulatory databases by its Chemical Abstracts Service (CAS) Number, 186018-45-1, and its DrugBank Accession Number, DB09046.[3] Its classification in the Anatomical Therapeutic Chemical (ATC) system is A16AA07.[3]
Metreleptin is a protein-based therapeutic classified as a leptin analog.[1] It is a recombinant, non-glycosylated polypeptide chain consisting of 147 amino acids, produced through biotechnological processes using an
E. coli expression system.[14] Its structure is nearly identical to that of native human leptin, with one critical distinction: the addition of a methionine residue at the N-terminus of the protein chain.[1] This modification is a direct result of its recombinant synthesis in bacteria. The full amino acid sequence is as follows: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTLSKMDQTLA VYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPWASGLETLDSLGGVLEASGY STEVVALSRLQGSLQDMLWQLDLSPGC.[6] The protein's tertiary structure is stabilized by a single intramolecular disulfide bond formed between the cysteine residues at positions 97 and 147.[19]
The empirical molecular formula for metreleptin is C714H1167N191O221S6.[6] It has an average molecular weight of approximately 16.15 kDa.[6] This single N-terminal methionine, while seemingly a minor alteration, is a crucial feature. Given that metreleptin is highly immunogenic in clinical use, with anti-drug antibodies developing in the majority of patients, this structural difference is the most probable epitope responsible for triggering the immune response.[11] The immune system can recognize this non-native N-terminus, leading to the formation of antibodies that can bind to the drug and, in some cases, neutralize its biological activity. This direct link between the manufacturing-derived structure and the primary safety concern underscores the challenges inherent in developing recombinant protein therapeutics.
Metreleptin is supplied for clinical use as a sterile, white, solid, lyophilized (freeze-dried) cake in a single-use vial containing 11.3 mg of the active drug.[23] It is formulated for subcutaneous injection and must be reconstituted with a suitable diluent prior to administration.[3] When reconstituted according to the manufacturer's instructions with 2.2 mL of diluent, the resulting solution has a final concentration of 5 mg/mL.[23]
Table 1: Metreleptin Drug Identifiers and Properties
Property | Value | Source(s) |
---|---|---|
Generic Name | Metreleptin | 6 |
Brand Names | Myalept®, Myalepta® | 3 |
DrugBank ID | DB09046 | 3 |
CAS Number | 186018-45-1 | 3 |
ATC Code | A16AA07 | 3 |
Type | Biotech, Leptin Analog | 1 |
Molecular Formula | C714H1167N191O221S6 | 6 |
Average Molecular Weight | 16155.44 Da | 6 |
Amino Acid Length | 147 amino acids (+ N-terminal Met) | 14 |
To understand the mechanism of metreleptin, it is essential to first understand the role of its endogenous counterpart, leptin. Leptin is a pleiotropic hormone secreted predominantly by adipocytes (fat cells) that acts as a critical regulator of energy homeostasis.[7] It signals to the central nervous system, particularly the hypothalamus, conveying information about the body's energy stores.[19]
Lipodystrophy syndromes are a heterogeneous group of rare disorders characterized by a generalized or partial loss of adipose tissue.[2] This loss of functional fat tissue leads to a profound deficiency of circulating leptin.[2] The absence of this key hormonal signal creates a state of perceived starvation in the brain, regardless of actual caloric intake. This drives a powerful, persistent sensation of hunger (hyperphagia), which in turn exacerbates the metabolic abnormalities.[6] Furthermore, without sufficient adipose tissue to store lipids, triglycerides are deposited ectopically in non-adipose tissues such as the liver, skeletal muscle, and pancreas. This ectopic fat deposition is the primary driver of the severe metabolic complications seen in these patients, including extreme insulin resistance, difficult-to-control diabetes mellitus, severe hypertriglyceridemia, and non-alcoholic fatty liver disease (NAFLD), which can progress to steatohepatitis (NASH) and cirrhosis.[2]
Metreleptin is designed as a direct replacement for the missing endogenous hormone. It functions as a pharmacological agonist of the human leptin receptor (ObR), a protein also known as LEPR or CD295.[6] By binding to and activating the ObR, metreleptin effectively mimics the physiological effects of native leptin, thereby restoring the deficient signaling pathway.[6] The ObR is a member of the Class I cytokine receptor family, and its activation by metreleptin, especially within the hypothalamus, is the foundational step of the drug's therapeutic action.[18]
Upon binding of metreleptin to the extracellular domain of the ObR, the receptor undergoes a conformational change that triggers the activation of an intracellular signaling cascade. The primary pathway utilized by the leptin receptor is the Janus kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway.[11] This activation leads to the phosphorylation and nuclear translocation of STAT proteins, which then act as transcription factors to modulate the expression of genes involved in appetite, energy expenditure, and metabolism.
The pharmacodynamic effects of metreleptin are a direct consequence of restoring leptin signaling. Centrally, activation of ObR in the hypothalamus re-establishes the body's sense of energy sufficiency, leading to a reduction in hyperphagia and an increase in satiety.[6] This reduction in caloric intake helps to alleviate the metabolic burden on the body.
Peripherally and centrally, the restored leptin signaling promotes profound improvements in systemic metabolism. Clinical studies have consistently shown that metreleptin therapy increases insulin sensitivity and enhances lipid metabolism.[1] These effects manifest as clinically meaningful reductions in glycated hemoglobin (HbA1c), fasting plasma glucose, and severely elevated fasting triglyceride levels.[3] The conceptualization of metreleptin as a true hormone replacement therapy, analogous to insulin for type 1 diabetes, explains its powerful and multi-faceted effects.[3] It addresses the root hormonal deficit rather than merely managing downstream symptoms. This model also clarifies why metreleptin is contraindicated and ineffective in patients with general obesity, who typically exhibit high leptin levels and a state of leptin resistance, not deficiency.[12]
Metreleptin is administered via subcutaneous injection, from which it is absorbed into the systemic circulation.[3] In patients with lipodystrophy, the median time to reach peak serum concentration (Tmax) is approximately 4 hours, with a range of 2 to 8 hours.[11] Pharmacokinetic studies in healthy subjects demonstrated that the peak concentration (Cmax) and total exposure (Area Under the Curve, AUC) are roughly proportional to the dose administered over a range of 0.01 mg/kg to 0.3 mg/kg.[29]
Following intravenous administration in healthy subjects, metreleptin exhibits a volume of distribution (Vd) that is approximately 4 to 5 times the plasma volume.[6] This suggests that the drug distributes beyond the vascular compartment into extravascular tissues. Formal studies on the extent of plasma protein binding have not been conducted.[29]
As a polypeptide therapeutic, metreleptin is not expected to undergo metabolism by the cytochrome P450 enzyme system. No formal metabolism studies have been performed in humans.[1] Nonclinical data indicate that systemic metabolism and degradation do not play a significant role in its elimination.[11] It is presumed to be catabolized into smaller peptides and constituent amino acids through general proteolytic pathways, similar to other endogenous proteins.
The primary route of elimination for metreleptin is renal clearance.[1] The drug is likely filtered by the glomeruli and subsequently taken up and degraded by the renal tubules, a common clearance mechanism for small proteins.[29] In healthy subjects who have not developed antibodies, the elimination half-life is between 3.8 and 4.7 hours.[11] This relatively short half-life suggests that without the influence of antibodies, the drug would not accumulate with a once-daily dosing schedule.[29]
The pharmacokinetic profile of metreleptin is profoundly altered by its high immunogenicity. The development of anti-metreleptin antibodies, which occurs in a majority of treated patients, creates a complex feedback loop that directly impacts the drug's disposition.[11] The clearance of metreleptin is significantly delayed in the presence of these antibodies.[1] This is because the formation of large drug-antibody complexes reduces the efficiency of renal filtration and clearance, leading to a prolonged terminal half-life and drug accumulation over time.[21] This phenomenon transforms the drug's otherwise predictable pharmacokinetic behavior into a highly variable and patient-specific profile. Consequently, monitoring for the development of antibodies is not merely a safety assessment but a critical component for interpreting a patient's clinical response and understanding the drug's effective exposure over time.
The approval of metreleptin for generalized lipodystrophy was primarily based on the results of a long-term, open-label, single-arm study conducted at the U.S. National Institutes of Health (NIH).[9] The study enrolled 48 patients with either congenital generalized lipodystrophy (CGL, n=32) or acquired generalized lipodystrophy (AGL, n=16), all of whom had baseline metabolic complications such as diabetes mellitus, hypertriglyceridemia, or hyperinsulinemia.[9] The patient population had extremely low baseline leptin levels, confirming a state of absolute leptin deficiency.[3]
After 12 months of metreleptin therapy, patients demonstrated substantial and clinically meaningful improvements in key metabolic parameters:
In contrast to the robust and consistent effects seen in GL, the efficacy of metreleptin in patients with partial lipodystrophy is more modest and variable.[3] This is attributed to the underlying pathophysiology; PL patients often have a "relative" rather than an absolute leptin deficiency, with baseline leptin levels that can be low, normal, or even elevated.[3]
Clinical studies have shown that while some patients with PL do benefit, the response is not universal.[3] One study reported a mean change in HbA1c of -0.6% and less consistent triglyceride reductions.[30] The benefit appears to be greater in patients who have more severe metabolic abnormalities at baseline.[31] This divergence in efficacy between GL and PL is a critical factor that has led to different regulatory decisions in the U.S. and Europe. The FDA concluded that the evidence was insufficient to establish effectiveness in PL, leading to an indication restricted to GL.[24] In contrast, the European Medicines Agency (EMA) determined that the benefit, although more modest, was sufficient to warrant approval for PL patients who have failed standard therapies, addressing a high unmet medical need.[8] This highlights that patient selection is paramount, and the therapeutic benefit is most pronounced in those with the most severe leptin deficiency.
The benefits of metreleptin appear to be durable. The pivotal NIH study followed patients for a median duration of 2.7 years, with some patients treated for over a decade, demonstrating sustained metabolic control.[9] Other reports from the NIH cohort confirm that improvements in HbA1c, triglycerides, and liver enzymes were maintained for up to 3 years of treatment.[31]
Table 2: Summary of Pivotal Clinical Trial Efficacy Data in Generalized Lipodystrophy (12-Month Follow-up)
Parameter | Baseline Value (Mean) | Mean Change from Baseline at 12 Months | Source(s) |
---|---|---|---|
HbA1c | 8.5% | -2.2% (95% CI: -2.7% to -1.6%) | 30 |
Fasting Triglycerides | 899 mg/dL | -32.1% (95% CI: -51.0% to -13.2%) | 30 |
Liver Volume | 3853 mL | -33.8% (p <0.001) | 9 |
Given its ability to reduce ectopic fat accumulation in the liver, metreleptin has been a subject of investigation for treating NASH, particularly when associated with lipodystrophy.[3] A completed Phase 2 clinical trial (NCT01679197) specifically evaluated its efficacy in this context.[32] Studies have shown that metreleptin therapy can lead to improvements in both hepatic steatosis (fat accumulation) and hepatic injury scores.[3] Despite these promising findings, regulatory bodies have not approved metreleptin for the standalone treatment of liver disease, including NASH, and this is noted as a limitation of use on its official labels.[12]
An emerging and compelling area of research involves the off-label use of metreleptin for anorexia nervosa. This application is based on the hypothesis that many of the severe psychological and behavioral symptoms of AN are driven by the profound hypoleptinemia that results from extreme fat mass loss.[3] In this context, leptin is viewed not just as a satiety hormone, but as a crucial neuro-hormonal signal of energy sufficiency. Its absence is thought to trigger a powerful, evolutionarily conserved "starvation response" that includes not only hunger but also heightened anxiety, obsessive thinking about food, and a compulsive drive for physical activity.
Case reports and small case series of off-label metreleptin use in patients with AN and atypical AN have documented rapid and significant improvements in psychological symptoms.[3] In one detailed case study of a patient with atypical AN, an 11-day course of metreleptin led to a dramatic reduction in depressive symptoms (Beck Depression Inventory-II score fell from 29 to 12), decreased weight phobia, and cessation of purging behaviors.[33] These changes occurred without a significant change in body weight, suggesting a direct neuro-psychiatric effect of restoring the leptin signal to the brain. This research reframes leptin's role and suggests metreleptin could be a novel neuro-regulatory agent for treating the core psychological aspects of eating disorders.
Metreleptin's path to market began in Japan, which granted the first global approval in March 2013 for the treatment of lipodystrophy.[15] This was followed by the U.S. Food and Drug Administration (FDA), which approved the drug as an orphan product on February 24, 2014.[5] The European Medicines Agency (EMA) granted marketing authorization on July 30, 2018.[1] Most recently, Health Canada approved the drug in January 2024.[3]
A key feature of metreleptin's regulatory history is the significant difference in the scope of its approved indications between the U.S. and other major jurisdictions.
Table 3: Comparison of Global Regulatory Indications (FDA vs. EMA)
Lipodystrophy Type | U.S. Food and Drug Administration (FDA) | European Medicines Agency (EMA) |
---|---|---|
Generalized Lipodystrophy (GL) | Approved: For congenital or acquired GL as replacement therapy for complications of leptin deficiency. | Approved: For confirmed congenital or acquired GL in adults and children ≥2 years of age. |
Partial Lipodystrophy (PL) | Not Approved: Safety and effectiveness have not been established. | Approved: For confirmed familial or acquired PL in adults and children ≥12 years of age for whom standard treatments have failed. |
In the United States, the significant safety risks associated with metreleptin prompted the FDA to mandate a Risk Evaluation and Mitigation Strategy (REMS) program.[5] The MYALEPT REMS PROGRAM is a restricted distribution program designed to mitigate the risks of neutralizing antibodies and lymphoma by ensuring appropriate patient selection.[13] Key requirements of the program include:
Reflecting the rarity of lipodystrophy syndromes, metreleptin has been granted orphan drug designation by both the FDA and the EMA.[1] This status provides regulatory and financial incentives to encourage the development of treatments for rare diseases that might otherwise lack commercial viability.
The FDA prescribing information for metreleptin carries a Boxed Warning, the agency's most stringent warning, highlighting two critical risks.[6]
The most frequently observed adverse reactions in clinical trials reflect the drug's potent metabolic effects and its route of administration.
Table 4: Common and Serious Adverse Reactions by Frequency
System Organ Class | Frequency Category | Adverse Reaction |
---|---|---|
Endocrine & Metabolic | Very Common (≥10%) | Hypoglycemia, Weight Loss |
Nervous System | Very Common (≥10%) | Headache |
Gastrointestinal | Very Common (≥10%) | Abdominal Pain |
Common (1-10%) | Nausea, Diarrhea, Pancreatitis | |
Immunologic | Common (1-10%) | Antibody Development (Neutralizing) |
General/Administration Site | Common (1-10%) | Fatigue, Pyrexia, Injection Site Reactions |
Musculoskeletal | Common (1-10%) | Arthralgia |
Hematologic | Common (1-10%) | Anemia |
Oncologic | Frequency Not Reported | T-Cell Lymphoma |
The dosing strategy for metreleptin is tailored based on patient body weight and sex to account for differences in body composition and leptin physiology.
Dose adjustments should be guided by clinical response, including metabolic parameters (HbA1c, triglycerides) and tolerability, with particular attention to avoiding excessive weight loss, especially in pediatric patients.[24]
Table 5: Recommended Dosing Regimens for Metreleptin
Patient Population | Starting Daily Dose | Dose Adjustments | Maximum Daily Dose |
---|---|---|---|
Weight ≤ 40 kg (males & females) | 0.06 mg/kg | Increments of 0.02 mg/kg | 0.13 mg/kg |
Males > 40 kg | 2.5 mg | Increments of 1.25 mg to 2.5 mg | 10 mg |
Females > 40 kg | 5 mg | Increments of 1.25 mg to 2.5 mg | 10 mg |
Metreleptin is supplied as a lyophilized powder and requires careful reconstitution prior to use. The choice of diluent is a critical safety step.
Metreleptin stands as a landmark therapeutic achievement, exemplifying the potential of targeted, mechanism-based treatments for rare diseases. As a true hormone replacement therapy, it addresses the fundamental pathophysiological deficit in lipodystrophy—the absence of leptin—rather than merely palliating its downstream metabolic consequences. Its profound efficacy in reversing severe insulin resistance and hypertriglyceridemia in patients with generalized lipodystrophy has transformed the clinical management and prognosis for this devastating condition.
However, the clinical journey with metreleptin is complex and highlights several critical, unresolved questions. The divergence in efficacy between generalized and partial lipodystrophy underscores the need for more sophisticated patient selection strategies. Future research must move beyond simple baseline leptin measurements to identify more robust biomarkers that can predict which subset of PL patients will derive a clinically meaningful benefit, thereby refining its use and justifying its high cost. Furthermore, the high rate of immunogenicity remains a central challenge. Long-term strategies for monitoring and managing the development of neutralizing antibodies are needed. Investigating whether immune tolerance protocols could mitigate this risk represents an important frontier for improving the drug's long-term safety and durability of response. Finally, while the association with T-cell lymphoma appears to be confounded by the natural history of acquired lipodystrophy, prospective, long-term registry data are essential to definitively quantify any attributable risk from the drug itself.
Perhaps the most exciting future perspective for metreleptin lies beyond lipodystrophy. The preliminary but compelling findings from its off-label use in anorexia nervosa suggest a potential paradigm shift in our understanding of leptin's function. These observations challenge the simplistic view of leptin as only a satiety hormone, repositioning it as a critical neuro-hormonal signal of energy sufficiency. Its absence appears to trigger a cascade of adverse psychological and behavioral adaptations characteristic of a starvation state. The rapid alleviation of these symptoms with metreleptin therapy, independent of weight change, strongly supports this hypothesis and opens a novel therapeutic avenue for treating the core neuropsychiatric components of eating disorders. Rigorously designed randomized controlled trials in this population are not only warranted but are an urgent priority.
In conclusion, metreleptin serves as a powerful case study in modern drug development. It demonstrates how a deep understanding of pathophysiology can lead to a life-altering therapy for a rare disease, while simultaneously presenting complex challenges in safety, patient selection, and risk management that require ongoing vigilance and research.
Published at: September 6, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.