What GLP-2R agonists are in clinical trials currently?

11 March 2025
Introduction to GLP-2R and its Agonists

Definition and Function of GLP-2R
The glucagon‐like peptide-2 receptor (GLP-2R) is a member of the class B G protein–coupled receptor family predominantly expressed in the gastrointestinal tract, particularly on intestinal enteroendocrine cells and within the subepithelial myofibroblast network. GLP-2, the natural ligand for this receptor, is secreted from enteroendocrine L-cells in response to nutrient ingestion. Once released, it binds to the GLP-2R to regulate several key physiological processes including intestinal growth (trophic effects), maintenance of mucosal integrity, enhancement of nutrient absorption, and modulation of intestinal permeability. The receptor mediates actions through coupling with intracellular signaling pathways that raise cyclic adenosine monophosphate (cAMP) levels and activate protein kinase A (PKA), thus orchestrating cellular responses that support intestinal adaptation and repair. This signaling cascade is essential for preserving gut barrier function and promoting mucosal proliferation—a function that becomes critical in states where the intestinal surface area is compromised.

Therapeutic Potential of GLP-2R Agonists
Given the central role of GLP-2R in regulating gut integrity and absorptive capacity, GLP-2R agonists have attracted significant attention as promising therapeutic agents. They hold potential for treating a variety of gastrointestinal disorders that are characterized by mucosal atrophy or deficits in nutrient absorption. In particular, conditions such as short bowel syndrome (SBS)—where patients suffer from a dramatically reduced intestinal surface area—and inflammatory bowel disease (IBD), which is associated with chronic inflammation and impaired barrier function, may benefit from therapies that enhance mucosal regrowth and repair. Moreover, by improving the overall absorptive function of the gut, these agents could lead to a better nutritional status and quality of life in affected patients. Consequently, the concept of agonizing the GLP-2 receptor to stimulate intestinal healing forms the basis of ongoing clinical efforts and research into next‐generation therapies aimed at a broader array of intestinal disorders.

Current GLP-2R Agonists in Clinical Trials

List of GLP-2R Agonists
Based on the available references from the Synapse source, one key report clearly states that “A GLP-2R agonist is being evaluated in clinical trials for the treatment of inflammatory bowel disease and short bowel syndrome.” Although the provided list does not include an extensive array of names under the GLP-2 category (as is more common with GLP-1 receptor agonists), the mention indicates that at least one candidate molecule is actively undergoing clinical investigation in these indications. In addition to this clinical trial mention, industry patents such as the one described in reference detail the development of novel peptide GLP-2 agonists. This patent highlights efforts to create GLP-2 analogues with increased protease resistance, improved pharmacokinetic properties (e.g., prolonged half-life via modifications such as lipidation or amino acid substitutions), and enhanced bioavailability in comparison to native GLP-2. While the patent does not always disclose the commercial name or phase of study explicitly, its content strongly implicates that a series of investigational peptide candidates—potentially including modifications of previously known agents like teduglutide—are being optimized for clinical development.

Teduglutide, the first GLP-2 analogue approved for short bowel syndrome, has established proof-of-concept for this entire therapeutic class; however, the current clinical trials are more focused on newer analogues that extend its beneficial attributes while reducing drawbacks such as frequent dosing or susceptibility to rapid degradation. In the field there are also candidates like glepaglutide—an agent described in other scientific literature though not explicitly named in the provided Synapse references—as well as several novel, structurally modified GLP-2 analogues mentioned in patents and earlier references. They are primarily being developed to address the limitations of existing therapies and to target diseases beyond SBS, such as various forms of inflammatory bowel disease (IBD).

Phases and Status of Clinical Trials
The reference from Synapse specifies that a GLP-2R agonist is currently in clinical trials but does not provide the precise phase, details of the dosing regimen, or quantitative endpoints. Available information implies that these clinical investigations are likely in early to mid-phase stages (Phase I/II) focusing on safety, tolerability, pharmacokinetics (PK), and preliminary measures of efficacy in patient populations with impaired intestinal function, such as those with short bowel syndrome (SBS) or IBD. The typical endpoints in these trials may include assessment of gut mucosal growth, nutrient absorption markers, intestinal barrier function, patient quality of life indices, and adverse effects related to gastrointestinal motility.

The clinical trial pipeline for GLP-2R agonists is designed to gather data that will eventually support longer-term efficacy outcomes and clinical benefit, particularly in relation to improved intestinal adaptation and reduced reliance on parenteral support in SBS patients. Given that a significant amount of accession is still ongoing in the patent domain, it suggests that many next-generation GLP-2 analogues have not yet transitioned fully from preclinical studies into advanced clinical testing, and that the current clinical trials are exploring a select number of promising candidates. As noted, while teduglutide remains the established therapy approved for SBS, the clinical trials for newer agents are seeking to overcome its dosing frequency and potential side effects by optimizing molecular structure for increased duration of action and reduced immunogenicity.

Mechanisms of Action and Benefits

Mechanisms of GLP-2R Agonists
GLP-2R agonists work by mimicking the endogenous hormone GLP-2. Upon binding to the GLP-2 receptor in the gastrointestinal tract, these agents stimulate several intracellular signaling pathways primarily mediated through cAMP and associated effectors such as protein kinase A (PKA). This cascade leads to a series of cellular events including intestinal epithelial proliferation, decreased apoptosis in enterocytes, and increased crypt cell survival—critical processes that stimulate the growth and maintenance of the gut mucosa.

Beyond promoting mucosal growth, the activation of GLP-2R also results in enhanced intestinal blood flow, which can aid in nutrient delivery and the repair of damaged mucosal tissues. Some studies suggest that this receptor-mediated activation can also decrease intestinal permeability and promote anti-inflammatory effects by modulating cytokine profiles within the gut. These molecular effects cumulatively contribute to improved gut barrier integrity and nutrient absorption. The molecular modifications applied to newer agonists—as described in patents—further enhance these mechanisms by ensuring sustained receptor activation with less frequent dosing. This protraction strategy allows for a once-weekly or similarly infrequent dosing schedule, potentially increasing patient compliance and overall clinical efficacy.

Clinical Benefits Observed
Clinical benefits of GLP-2R agonists have been most prominently observed in conditions characterized by severe intestinal insufficiency, such as SBS and IBD. In SBS patients, stimulation of mucosal growth leads to expansion of the absorptive surface area. This translates into increased nutrient and fluid absorption, reduced dependence on parenteral nutrition, and subsequently, improvements in quality of life and reduction in complications associated with parenteral support. Enhanced mucosal structure not only contributes to overall gut health but also stabilizes gastrointestinal function, thereby reducing symptoms such as diarrhea and malabsorption.

For patients with inflammatory bowel conditions, the protective effect on the intestinal barrier can result in decreased intestinal inflammation and enhanced mucosal healing. This therapeutic effect is particularly promising given the challenges associated with chronic inflammation and the risk of complications like strictures and fistulas in IBD. The early clinical trial designs of new GLP-2 analogues often include secondary endpoints that monitor biomarkers related to cytokine production and intestinal permeability changes, further validating the fundamental mechanisms of action and their potential benefits. Although detailed quantitative data from these trials are still emerging, the general consensus from early-phase studies suggests that these agents can confer clinically meaningful improvements that extend beyond mere trophic effects, offering protection against intestinal injury and aiding in functional restoration.

Challenges and Future Prospects

Current Challenges in Development
Despite the promising therapeutic potential of GLP-2R agonists, several challenges complicate their clinical development. One of the primary challenges is the inherent short half‐life of native GLP-2 peptides due to rapid degradation by dipeptidyl peptidase-4 (DPP-4) and other proteolytic enzymes within the gastrointestinal tract. This necessitates frequent dosing or the development of modified analogues with increased protease resistance. Patent addresses this challenge by introducing chemical modifications—such as specific amino acid substitutions and lipidation strategies—that significantly prolong the half-life and improve the overall bioavailability of these agonists. However, these modifications must be carefully optimized to preserve receptor binding affinity and minimize immunogenicity.

Another significant challenge is balancing the efficacy and safety profile. While robust stimulation of intestinal growth is desirable, excessive proliferation could potentially increase the risk of neoplasia or other long-term adverse effects. Thus, clinical trials must meticulously monitor safety endpoints along with efficacy measures in order to establish an acceptable therapeutic window. Additionally, the local gastrointestinal side effects, such as nausea or abdominal discomfort—which are also observed with GLP-1 receptor agonists—must be addressed through careful titration and dosing regimens.

From a regulatory perspective, the demonstration of clear clinical benefit—such as a reduction in parenteral nutrition requirements in SBS patients or measurable improvement in mucosal healing in IBD patients—remains paramount. Because these conditions are complex and multifactorial, robust trial designs with sufficient patient numbers and well-defined endpoints are needed to convince regulatory agencies of the benefit-risk profile. Finally, the relative novelty of GLP-2R agonist therapy means that long-term safety data are limited. Questions surrounding chronic use, potential immunogenicity of modified peptides, and unexpected adverse effects complicate the clinical development pathway.

Future Research Directions and Potential
Looking into the future, several avenues exist for further developing GLP-2R agonists. First, ongoing research continues to focus on optimizing the chemical structure of these candidates to maximize receptor agonism while minimizing degradation. Strategies such as novel lipidation techniques, cyclization of the peptide backbone, and incorporation of non-natural amino acids are being explored to create super-stable analogues that allow for infrequent dosing such as weekly or even monthly administration. Such improvements would greatly enhance patient adherence and overall treatment outcomes. The patent is a prime example of these innovative approaches in design that are poised to transition into clinical implementation.

Second, future clinical trials will likely broaden the range of indications for GLP-2R agonists beyond SBS and IBD. While these two conditions represent the current primary targets, emerging research suggests that improved gut barrier function and anti-inflammatory effects might benefit patients with other gastrointestinal disorders, including celiac disease and even systemic disorders where gut health plays a central role. Moreover, by integrating biomarkers of intestinal integrity, such as changes in citrulline levels, inflammatory cytokines, and functional imaging of intestinal morphology, clinical studies can more precisely measure therapeutic impact and tailor patient selection.

Third, combination therapies could represent the next frontier for GLP-2R agonists. There is potential for synergies when combining GLP-2R agonists with other regenerative or anti-inflammatory agents. For instance, pairing a GLP-2R agonist with a GLP-1R agonist may exploit complementary mechanisms—enhancing both mucosal growth and metabolic regulation, which could be particularly beneficial for patients with metabolic disorders complicated by gastrointestinal dysfunction. Preclinical studies and early clinical experiments focusing on combination therapies might pave the way for multipronged approaches that address both the structural and functional deficiencies in compromised intestinal tracts.

Additionally, advances in the fields of imaging and non-invasive biomarker assessment are set to revolutionize how the efficacy of GLP-2R agonists is measured. Modern imaging techniques, such as high-resolution endoscopy coupled with advanced computational analysis, could allow clinicians to quantify mucosal changes more effectively over time. In parallel, the development of sensitive blood-based or stool-based biomarkers that reflect intestinal epithelial turnover and barrier function will help in monitoring treatment response with greater accuracy. These innovations will contribute to a more precise evaluation of the pharmacodynamics of GLP-2R agonists in clinical trials and encourage further development in this promising field.

Furthermore, a key area of future research is to understand the interplay between the gut microbiota and GLP-2R agonist therapy. The gut microbiome plays an essential role in modulating intestinal immunity and barrier function, and emerging evidence suggests that GLP-2R signaling might influence the composition of the gut microbial community. Future studies could investigate whether GLP-2R agonist-induced mucosal growth is accompanied by beneficial shifts in the microbiota, which in turn could amplify clinical benefits. Integrating microbiome analysis into large-scale clinical trials may uncover additional mechanisms of action and open new therapeutic windows.

Lastly, personalized medicine is another exciting prospect for the future of GLP-2R agonist therapy. Genetic variations in GLP-2 receptor expression or function may explain differences in therapeutic responses among patients. With advances in pharmacogenomics, it could become possible to stratify patients based on their likelihood of response to GLP-2R agonists, thus tailoring treatment plans more precisely. This precision approach would enhance the efficacy of therapy while minimizing adverse events, thereby optimizing the overall benefit-risk balance for individual patients.

Conclusion
In summary, the role of GLP-2 receptor agonists in clinical trials is rapidly evolving. On a general level, these agents are designed to mimic the actions of the native GLP-2 hormone, promoting intestinal growth, enhancing mucosal regeneration, and improving nutrient absorption by stimulating specific intracellular signaling cascades. From a specific perspective, current clinical trials—mentioned in Synapse reference—focus on evaluating at least one GLP-2R agonist in conditions such as inflammatory bowel disease and short bowel syndrome, while patent literature shows that there is significant effort toward developing novel GLP-2 analogues with improved pharmacokinetic properties. These efforts aim not only to overcome the limitations of existing agents like teduglutide but also to extend the therapeutic potential of GLP-2R agonists into new clinical domains.

From a broad perspective, further advancements in the chemical modification and molecular design of these agents are expected to lead to improved stability and longer dosing intervals, enhancing patient compliance and clinical outcomes. Mechanistically, GLP-2R agonists work through the upregulation of cAMP signaling cascades, resulting in mucosal proliferation, enhanced blood flow, and improved barrier function, all of which contribute to better intestinal adaptation and reduced morbidity in gastrointestinal diseases. However, challenges remain in ensuring the long-term safety of these agents, particularly given the potential risks associated with excessive mucosal proliferation and the need for a balanced therapeutic index.

Looking forward, future research directions include optimizing the molecular structure of GLP-2 analogues, integrating combination treatment approaches, leveraging novel imaging and biomarker technologies, and exploring the regulatory relationships with the gut microbiome. Advances in pharmacogenomics and personalized medicine may also enable more tailored and effective therapies for patients with gastrointestinal insufficiency. While definitive clinical results are still emerging, the promising early-phase data and robust patent activity underscore the potential of GLP-2R agonists to redefine the treatment landscape for conditions such as short bowel syndrome and inflammatory bowel disease.

In conclusion, as clinical trials continue and additional data become available, it is anticipated that new GLP-2R agonists will offer a transformative approach in managing patients with compromised gut function by not only enhancing mucosal repair and nutrient absorption but also by improving overall gastrointestinal health. This progress holds the promise of reducing dependence on parenteral nutrition in SBS patients and mitigating inflammatory processes in IBD, ultimately leading to improved patient outcomes and quality of life. Future studies will be critical in confirming these benefits and in addressing the challenges inherent in peptide drug development, paving the way for these agents to become a cornerstone in the treatment of complex gastrointestinal disorders.

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.