What are the therapeutic candidates targeting LEPR?

11 March 2025
Introduction to LEPR
The leptin receptor (LEPR) is a critical mediator of the hormone leptin, playing a central role in the regulation of energy homeostasis, food intake, and various neuroendocrine functions. LEPR is broadly expressed in the central nervous system—particularly within the hypothalamus—and in peripheral organs, where it contributes to a variety of physiological responses. Studies have shown that its activation not only helps regulate appetite and body weight but is also important for metabolic processes, reproduction, bone homeostasis, and immune modulation, making it a key target for disease intervention.

Role of LEPR in Physiology
LEPR functions as the primary receptor for leptin, an adipocyte‐derived hormone whose serum levels correlate with adipose stores. Upon binding of leptin to its full‐length receptor isoform (OB-Rb), downstream signaling cascades—including the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway, the phosphatidylinositol 3-kinase (PI3K) pathway, and the mitogen-activated protein kinase (MAPK) pathway—are activated. These signaling mechanisms help regulate feeding behavior, energy expenditure, and glucose homeostasis. The receptor is also implicated in neuroprotective mechanisms and modulation of inflammatory responses, underscoring its pleiotropic role in physiological regulation.

Importance of LEPR in Disease
Disruptions or alterations in LEPR signaling have been linked to several pathological conditions. In obesity, high circulating levels of leptin coexist with a reduced sensitivity to its effects—a phenomenon known as leptin resistance—which contributes to metabolic dysregulation. In lipodystrophy, reduced or dysfunctional leptin signaling leads to severe metabolic complications that can be life-threatening. Moreover, abnormalities in LEPR function have been implicated in reproductive disorders, as well as in tumor progression where leptin signaling contributes to a proangiogenic and anti-apoptotic tumor microenvironment. The complexity of LEPR signaling, including its involvement in multiple intracellular cascades, makes it an attractive target for therapeutic intervention in a wide range of diseases such as obesity, metabolic syndrome, and certain cancers.

Therapeutic Candidates Targeting LEPR
Over the years, various therapeutic candidates have been developed to modulate LEPR signaling. These candidates can be broadly classified into those that enhance LEPR activity (agonists) and those that inhibit its downstream effects (antagonists). The therapeutic candidates include current approved therapies as well as a host of investigational drugs at different stages of preclinical and clinical development.

Current Approved Therapies
In the realm of approved therapies, recombinant leptin products such as metreleptin (marketed as Myalepta) represent an established approach to modulating leptin receptor activity. Metreleptin is designed to mimic the endogenous hormone leptin, binding to LEPR and thereby ameliorating conditions characterized by leptin deficiency, such as certain forms of lipodystrophy. This therapy has received regulatory approval in various regions including the European Union, Japan, and by the FDA and is administered subcutaneously. The effectiveness of metreleptin in patients with congenital or acquired lipodystrophy supports the concept that restoring leptin signaling can substantially improve metabolic and endocrine disturbances.

Although metreleptin does not directly act on LEPR in the sense of a small-molecule modulator, its use is predicated on its ability to engage the receptor and trigger downstream signaling pathways that are essential for energy balance, appetite regulation, and metabolic control. Its clinical performance in these approved indications demonstrates the therapeutic potential of targeting the leptin–LEPR axis.

Investigational Drugs
There is significant ongoing research into novel therapeutic candidates that target LEPR more directly. These investigational drugs include both LEPR agonists and antagonists that operate via diverse mechanisms:

1.  Preclinical LEPR Antagonists:
Recent drug development efforts have led to the discovery of small-molecule and biologic candidates that act as LEPR antagonists. For example, ARV-1802 and ARV-1803, developed by organizations such as Arrevus, Inc. and Temple University Graduate School, are two investigational candidates that have been characterized as LEPR antagonists in the context of neoplasms and other diseases. These drugs have been shown to block the interaction between leptin and its receptor, thereby inhibiting the downstream signaling pathways that contribute to tumor survival and progression. Although still in preclinical phases, these candidates offer promise for the treatment of cancers where aberrant leptin signaling plays a role.

2.  LEPR Modulators via RNA Interference:
Another innovative approach has been the development of RNA interference (RNAi) therapeutic candidates. An example is CCT-417 from Canary Cure Therapeutics Inc., which is based on siRNA technology; it modulates the activity of a receptor complex involving CB1 and LEPR. By downregulating the expression of these proteins, CCT-417 aims to restore or alter leptin signaling, particularly in diseases of the nervous system or metabolic disorders. This method offers a targeted strategy to fine-tune receptor expression without entirely abolishing the receptor’s physiological functions.

3.  Antibody-Based Therapeutics:
The design of monoclonal antibodies against LEPR represents another frontier in targeting this receptor. Several patents and research publications describe antibody-based strategies for either agonism or antagonism. Antibodies that act as leptin receptor agonists have been explored in order to treat conditions such as hypoleptinemia and metabolic dysfunction. Conversely, anti-LEPR antibodies that block receptor signaling are being investigated for therapeutic applications in cancer by inhibiting the pro-survival and pro-angiogenic effects of leptin in the tumor microenvironment. The targeted nature of these therapies allows for high specificity and the potential to avoid some of the off-target effects associated with small-molecule drugs.

4.  Novel Peptide-Based Agents:
Design, synthesis, and evaluation of radiolabeled and modified peptides that interact with LEPR are also under investigation. These peptides may serve both diagnostic and therapeutic purposes, particularly in assessing receptor expression in neoplastic tissues and as direct therapeutic agents when modified to function as receptor antagonists. Peptide-based agents offer advantages such as high tissue penetration and potentially lower immunogenicity compared to full-length antibodies.

5.  Investigational Leptin Receptor Agonist Antibodies for Rare Diseases:
In some early-phase clinical studies, investigational leptin receptor agonist antibodies (sometimes referred to as novel LEPR agonist antibodies) have been tested in patients with congenital lipodystrophy. Case studies presented at scientific meetings such as ENDO 2024 have shown that switching from metreleptin therapy to a novel LEPR agonist (mibavademab) has resulted in significant improvements in metabolic parameters such as triglyceride levels and overall clinical outcomes. Although these studies are at an early phase, the ability of these antibodies to overcome the loss of efficacy associated with metreleptin resistance or secondary failure is an important advancement in the field.

The investigational candidates represent a diverse array of approaches, spanning small molecules, RNAi agents, peptide conjugates, and monoclonal antibodies. Each approach targets LEPR directly, either by mimicking leptin’s agonistic activity, antagonizing the receptor to block pathological signaling, or modulating receptor expression, and is designed to overcome limitations inherent to endogenous leptin therapy and leptin resistance.

Mechanisms of Action
A clear understanding of the molecular and physiological mechanisms by which therapeutic candidates interact with LEPR is essential, as these mechanisms ultimately determine the therapeutic efficacy and safety profile.

How Therapeutic Candidates Interact with LEPR
Therapeutic candidates targeting LEPR engage the receptor through different modes of interaction:

• Agonists such as metreleptin mimic the natural hormone leptin by binding to the extracellular domain of the receptor (specifically the full-length OB-Rb isoform). Once bound, these agonists induce receptor dimerization and initiate downstream signaling cascades. This activation typically results in the phosphorylation of JAK2, leading to STAT3 activation, and subsequent transcription of target genes that control appetite and energy expenditure.

• Antagonists, including ARV-1802 and ARV-1803, bind competitively to the leptin binding site on LEPR. By occupying the receptor without triggering the conformational changes necessary for signaling, these agents block leptin from engaging the receptor. This inhibition is particularly relevant in pathologic states such as certain cancers, where blocking leptin signaling can inhibit tumor growth and angiogenesis.

• Antibody-based therapeutics have been engineered either to mimic leptin (agonist antibodies) or to block leptin binding (antagonist antibodies). In designing these agents, researchers typically focus on maintaining high affinity for specific receptor epitopes while ensuring that they can either trigger or inhibit the natural receptor conformation changes necessary for signal transduction.

• RNAi-based modulators like CCT-417 reduce the expression of LEPR (or associated receptor complexes) through targeted gene silencing. This approach can fine-tune the receptor’s abundance and, as a result, modulate the downstream signaling cascade without entirely eliminating the receptor function. This strategy might be ideal in cases where only partial attenuation of leptin signaling is desirable, such as in metabolic disorders or neurodegenerative conditions.

Biological Pathways Involved
Upon activation by its ligand (or therapeutic substitute), LEPR triggers a cascade of intracellular events. The primary pathways implicated include:

• The JAK/STAT Pathway: After binding of leptin or an agonist, LEPR activates JAK2, which phosphorylates specific tyrosine residues on the receptor. These phosphorylated sites then serve as docking platforms for STAT proteins, particularly STAT3. Activated STAT3 dimerizes and translocates to the nucleus to modulate gene transcription involved in appetite regulation and metabolic control.

• The PI3K Pathway: LEPR activation also engages the PI3K pathway, which contributes to the regulation of glucose metabolism and cell survival. This pathway can interact with the insulin signaling cascade, thus playing a synergistic role in maintaining metabolic homeostasis.

• The MAPK Pathway: Activation of the MAPK pathway leads to the regulation of cell proliferation and differentiation. In pathological conditions such as cancer, aberrant activation of this pathway by leptin has been associated with increased cell proliferation and angiogenesis.

• Additional Pathways: Other signaling cascades, including those mediated by mTOR and AMPK, are also influenced by LEPR activation. These pathways collectively modulate energy balance, autophagy, and inflammatory responses, each of which is crucial in both normal physiology and disease states.

Clinical Development and Trials
Clinical evaluation of therapeutic candidates targeting LEPR spans from extensive preclinical studies in cellular and animal models to various phases of human clinical trials. A robust preclinical dataset is essential to establish both efficacy and safety before moving into early human studies.

Preclinical Studies
Preclinical investigations of LEPR-targeting candidates have focused on demonstrating the mechanism of receptor interaction, modulation of downstream signaling, and therapeutic efficacy in animal models. For instance:

• ARV-1802 and ARV-1803 have been investigated in preclinical models of neoplasms. These compounds have been shown to modulate leptin signaling pathways in cell-based assays and animal models, providing evidence for their potential to inhibit tumor growth and angiogenesis through the blockade of LEPR signaling.

• RNAi therapies such as CCT-417 have undergone gene silencing studies in cellular models and early animal studies. These studies assess not only the reduction of LEPR expression but also the downstream effects on metabolic and neural endpoints, proving the feasibility of this novel approach.

• Antibody-based candidates have been evaluated in animal models to determine their pharmacokinetics, biodistribution, and effect on metabolic endpoints. Investigational LEPR agonist antibodies have shown promising results in preclinical models of congenital lipodystrophy, where correction of metabolic abnormalities has been observed after administration.

In these preclinical models, a variety of endpoints are assessed, including receptor occupancy, activation of downstream pathways (measuring phosphorylated STAT3 levels, for example), metabolic biomarkers, tumor growth inhibition, and comprehensive safety evaluations. The use of relevant biomarkers and gene expression profiling in these early studies is critical for guiding further clinical development.

Clinical Trial Phases and Results
In the clinical arena, metreleptin stands as the only currently approved agent that indirectly influences LEPR by mimicking endogenous leptin. It has undergone rigorous evaluation in Phase 2 and Phase 3 clinical trials, demonstrating significant improvements in metabolic and endocrine parameters in patients with lipodystrophy.

For investigational agents, early-phase clinical trials are underway or being planned:

• Investigational LEPR agonist antibodies have been tested in small-scale, early-phase clinical studies. For instance, case studies reported at ENDO 2024 have highlighted the potential of novel LEPR agonist antibodies to improve metabolic outcomes in patients with congenital lipodystrophy who have previously experienced inadequate responses to metreleptin therapy. Although the number of patients in these evaluations is small, the observed improvements in biochemical markers such as triglyceride concentrations underscore the promising potential of this therapeutic approach.

• Investigational LEPR antagonists, such as ARV-1802 and ARV-1803, remain at the preclinical stage. Their advancement into clinical testing will depend on further demonstration of their efficacy in animal models and optimization of their pharmacokinetic and safety profiles.

• RNAi-based approaches targeting LEPR, exemplified by CCT-417, have shown strong preclinical promise but are still awaiting transition into Phase 1 trials. The challenge for these agents lies in ensuring targeted delivery, efficient gene silencing in vivo, and mitigation of off-target effects.

Across these studies, investigators are rigorously evaluating parameters such as receptor binding affinity, modulation of critical signaling cascades (evidenced by changes in phosphorylated STAT3 and other biomarkers), improvement in clinical endpoints (such as weight reduction, improved glycemic control, or tumor response), and safety profiles. The heterogeneous nature of diseases linked to LEPR dysfunction means that clinical trial designs often use enrichment strategies based on biomarker discovery, thereby ensuring that patients most likely to benefit are enrolled. The results from early-phase studies are encouraging; however, larger randomized controlled trials will be necessary to confirm the therapeutic potential and delineate the precise role of these agents in the clinical armamentarium.

Challenges and Future Directions
Despite the promising advances in therapeutic candidates targeting LEPR, several challenges must be overcome to fully realize their clinical potential. The complexity of LEPR signaling and the heterogeneity of diseases associated with leptin resistance present unique hurdles for drug development.

Current Challenges in Targeting LEPR
One major challenge is the inherent complexity of LEPR biology. Leptin receptor signaling pathways are highly redundant and cross-talk with multiple intracellular cascades—including JAK/STAT, PI3K, and MAPK—which can lead to compensatory mechanisms that blunt therapeutic efficacy. Additionally, leptin resistance, a hallmark of obesity, means that simply increasing leptin or stimulating its receptor may not be sufficient to achieve long-lasting therapeutic benefits. This situation is further complicated by the presence of multiple receptor isoforms (full-length versus short forms) that have different and sometimes opposing functions within various tissues.

For antagonists, the risk of disrupting physiologic leptin signaling in healthy tissues poses a significant safety concern. LEPR antagonists must be designed to selectively block pathological signaling without causing adverse effects such as increased susceptibility to infections, metabolic disturbances, or negative impacts on neuroendocrine function.

Further, the development of biological agents, such as monoclonal antibodies, faces challenges related to immunogenicity, stability, and cost of production. Ensuring that these agents maintain sufficient specificity and affinity for LEPR, while avoiding cross-reactivity with other cytokine receptors, is a non-trivial aspect of drug design.

RNA interference approaches, while promising, must overcome the challenges of efficient in vivo delivery, potential off-target gene silencing, and safe long-term modulation of gene expression.

From a clinical trial perspective, the selection of appropriate patient populations and biomarkers is critical. Given the variability in leptin sensitivity and the heterogeneity of conditions like obesity, lipodystrophy, and certain cancers, robust biomarker-driven stratification will be required to identify those patients who are most likely to benefit from LEPR-targeting therapies.

Future Research Directions and Opportunities
Looking ahead, several research directions hold promise in overcoming current challenges and advancing the therapeutic targeting of LEPR:

• Optimization of Drug Design:
Advanced computational modeling, molecular dynamics simulations, and structure–activity relationship (QSAR) studies are essential tools for refining the design of LEPR-targeting agents. Improving binding affinity and selectivity through iterative design and utilizing machine learning algorithms to predict off-target effects may accelerate drug discovery efforts.

• Development of Combination Therapies:
Given the multiple signaling pathways interconnected with LEPR, one promising strategy is the development of combination therapies that target both LEPR and downstream effectors. For example, combining a LEPR agonist with a modulator of the PI3K or mTOR pathway may prove beneficial in overcoming leptin resistance and enhancing metabolic outcomes. Similarly, in oncology, combining LEPR antagonists with other targeted agents could lead to improved tumor control by simultaneously inhibiting several pro-survival signals in cancer cells.

• Personalized Medicine Approaches:
Future research should focus on the identification and validation of biomarkers that predict response to LEPR-targeting therapies. Integrating genomic, proteomic, and metabolomic data can facilitate the development of predictive models that guide patient selection and dosage optimization. Stratification of patients based on specific genetic or molecular signatures related to leptin signaling could greatly enhance clinical trial outcomes.

• Innovative Delivery Platforms for RNAi Therapies:
For RNAi-based therapeutic candidates such as CCT-417, developing advanced delivery platforms—including nanoparticle-based systems or conjugation with targeting ligands—will be crucial for efficient and selective delivery to tissues of interest. Improved delivery methods could minimize off-target effects and maximize gene knockdown in target cells.

• Enhanced Antibody Engineering:**
Continued advancements in antibody engineering, including the development of bispecific antibodies and antibody-drug conjugates (ADCs), offer exciting opportunities for targeting LEPR. These engineered antibodies can be tailored to either stimulate or block receptor signaling as needed and may have improved pharmacokinetic profiles and reduced immunogenicity compared to traditional monoclonal antibodies.

• Long-Term Efficacy and Safety Studies:
As new LEPR targeting agents progress from preclinical stages to clinical trials, it will be essential to conduct longitudinal studies to assess long-term efficacy and potential adverse effects. In diseases like obesity and cancer, where treatment may be chronic, understanding the long-term impact of modulating LEPR signaling is crucial. These studies should incorporate comprehensive biomarker analyses to monitor the physiological and metabolic effects over time.

• Exploration of New Disease Indications:
While much of the current focus is on metabolic diseases, lipodystrophy, and cancer, emerging evidence suggests that LEPR plays a role in other conditions—including neurodegenerative diseases and inflammatory disorders. Investigating the potential benefits of LEPR-targeting therapies in these new indications could open up additional therapeutic avenues and expand the clinical utility of these agents.

• Translational Research and Public–Private Partnerships:
Advancing LEPR-targeted therapies will greatly benefit from close collaboration between academia, biotechnology companies, and regulatory agencies. These partnerships can help streamline the translational process from bench to bedside, ensuring that promising candidates move efficiently through preclinical testing and clinical evaluation. The integration of robust real-world evidence and post-marketing surveillance data will further refine treatment protocols and enhance our understanding of these therapies in diverse patient populations.

Conclusion
In summary, targeting the leptin receptor (LEPR) offers a multifaceted therapeutic opportunity across a range of diseases—from metabolic disorders and lipodystrophy to certain cancers. Current approved therapies, such as metreleptin, serve as proof-of-concept for the benefits of restoring or modulating leptin signaling. At the same time, a variety of investigational approaches—including small-molecule antagonists (ARV-1802, ARV-1803), RNAi-based agents (CCT-417), and both agonist and antagonist monoclonal antibodies—are being developed to more directly and precisely regulate LEPR activity.

The mechanism of action of these therapeutic candidates is intimately linked to their effects on key intracellular pathways such as JAK/STAT, PI3K, MAPK, and mTOR, which govern energy homeostasis and cellular proliferation. Preclinical studies have generated significant insights into receptor occupancy, downstream signaling modulation, and efficacy in animal models, and early-phase clinical trials—especially those involving LEPR agonist antibodies—show promising results in improving metabolic parameters in difficult-to-treat populations.

Nevertheless, challenges persist due to the complexity of leptin receptor regulation, the phenomenon of leptin resistance, and the need for precise patient stratification using biomarkers. Future research will likely focus on optimizing drug design via advanced computational methods, exploring combination therapies, and deploying personalized medicine strategies. Innovative delivery systems for RNAi and next-generation antibody engineering will further enhance the specificity and safety profiles of these therapies, while robust long-term studies will be essential to understanding their sustained effects.

In conclusion, therapeutic candidates targeting LEPR represent a dynamic and evolving field of research. Their development is underpinned by a deepening understanding of LEPR signaling pathways and an increasing ability to modulate these pathways with high precision. With ongoing investigations and coordinated translational efforts, these agents have the potential to broaden our therapeutic arsenal against complex conditions like obesity, lipodystrophy, and cancer, ultimately improving patient outcomes and advancing precision medicine in these challenging disease areas.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.