What are the new molecules for SGLT1 inhibitors?

11 March 2025
Introduction to SGLT1 Inhibitors
Sodium–glucose cotransporter 1 (SGLT1) is a key member of the SGLT family that plays a fundamental role in the absorption of dietary glucose, galactose, and other hexoses primarily in the small intestine, and in part in the kidney’s late proximal tubules. SGLT1’s action enables the efficient uptake of these sugars from the intestinal lumen into epithelial cells, helping to maintain systemic glucose homeostasis. Its physiological role extends to other tissues, where it may be involved in fluid balance and cellular energy supply. The inhibition of SGLT1, therefore, represents a promising therapeutic strategy to reduce postprandial blood glucose spikes, manage hyperglycemia, and ultimately mitigate the risk factors associated with diabetes and metabolic syndrome. This therapeutic approach offers an opportunity to modulate the absorption process at its origin, thereby presenting a complementary or alternative mechanism to other antidiabetic medications that often target insulin secretion or sensitivity.

Role of SGLT1 in Physiology
SGLT1 is predominantly localized in the intestinal brush border membranes and is responsible for mediating the active transport of glucose and galactose against their concentration gradient using the sodium gradient as a driving force. In the gastrointestinal tract, this transporter represents the first line of entry for dietary carbohydrates, which are subsequently metabolized or utilized for energy production in peripheral tissues. Beyond the small intestine, SGLT1 is expressed in renal tubules—albeit at a lesser extent compared to SGLT2—and in other tissues including the lung, heart, and pancreas, suggesting additional roles related to energy metabolism and cellular homeostasis. This diversity in expression patterns underscores its potential involvement not only in glucose uptake but also in other metabolic and physiologic processes.

Importance of SGLT1 Inhibition
Inhibiting SGLT1 may directly reduce the intestinal uptake of dietary glucose, thereby blunting postprandial hyperglycemia—a critical factor in the pathogenesis of type 2 diabetes. Moreover, by limiting glucose absorption, SGLT1 inhibitors could be beneficial in reducing the overall glycemic load which, in turn, might help lessen the progression of insulin resistance and associated metabolic derangements. Selectively targeting SGLT1 may also circumvent some of the adverse effects associated with SGLT2 inhibitors, such as genitourinary infections, as intestinal SGLT1 inhibition can be localized to the gut with minimal systemic exposure. This approach offers an attractive complementary mechanism to other therapeutic regimens, expanding the scope of intervention for metabolic diseases while potentially mitigating side effects linked to off-target tissue accumulation.

New Molecules for SGLT1 Inhibition
The focus on novel molecules for SGLT1 inhibition has accelerated in recent years, driven by advances in computational modeling, high‐throughput screening, and structure–activity relationship studies. Researchers have identified numerous candidate compounds with promising SGLT1 inhibitory activity, offering new avenues for therapeutic interventions. These new molecules generally fall into two broad categories: those discovered via rational design and in silico methodologies and those derived from natural product libraries or modifications of existing glycoside structures.

Recent Discoveries and Developments
Among the novel chemical entities reported in the literature and patents, several molecules have emerged as strong candidates for SGLT1 inhibition. For instance, a compound described as a 4-isopropylphenylglucitole was disclosed in a patent, noted for its capacity to inhibit SGLT1 activity without significant in vivo accumulation. This molecule demonstrated a significant effect on suppressing postprandial hyperglycemia and glucose absorption in the small intestine, thereby offering a potential pathway to prevent or treat diabetes and metabolic disorders.

Another breakthrough came with the presentation of a novel SGLT1 inhibitor referred to simply as a “compound of Formula I” or its hydrate, which has been detailed in patent literature. This invention, attributed to one of the leading companies in the field, underscores the innovative approaches being taken to design molecules that not only achieve the desired inhibitory effect but also possess favorable pharmacokinetic and safety profiles. The molecule’s design is centered around strategic modifications at specific sites on the scaffold to enhance selectivity and potency toward SGLT1.

Additionally, computational studies and virtual screening efforts have further broadened the chemical space available for SGLT1 inhibition. One study integrated homology modeling with ligand-based pharmacophore modeling and molecular docking techniques, resulting in the identification of 16 new compounds with structural features reminiscent of documented SGLT1 inhibitors. Out of these, two compounds, referred to as compound 81 and compound 91, displayed particularly enhanced stability and favorable binding free energies, suggesting their potential as robust candidates for further development. These findings highlight the role of advanced in silico methodologies in rapidly narrowing down large databases to a handful of promising molecules that merit further experimental validation.

Proteochemometric modeling approaches have also contributed to these discoveries. By combining compound-based and protein-based descriptors into random forest models, researchers have been able to predict and subsequently validate SGLT1 inhibitory activity with considerable success. Out of 77 compounds tested in one such study, 30 were confirmed to possess inhibitory activity in vitro, yielding a hit rate of 39% and showcasing the effectiveness of these modern AI-driven screening tools. This has opened up possibilities for the synthesis of entirely new chemical series that were previously unexplored in the context of SGLT1 inhibition.

Moreover, the natural product space has not been left untouched. Some studies have identified novel natural and synthetic inhibitors that fall outside the typical glycoside chemical space. For example, a study reported the discovery of several molecules—including (+)-pteryxin, (+)-ε-viniferin, quinidine, cloperastine, bepridil, trihexyphenidyl, and bupivacaine—that exhibit SGLT1 inhibitory activity. Although these compounds were initially evaluated for dual SGLT1/2 inhibition, their differential potencies and selectivity suggest that further chemical refinement could eventually lead to highly selective SGLT1 inhibitors.

Structural biology has provided another leap forward in this area. The cryo-electron microscopy (cryo-EM) structure of the human SGLT1-MAP17 complex in the presence of LX2761—a high-affinity inhibitor—revealed the precise binding mode of the inhibitor, which wedges into the substrate-binding site and locks the transporter in an outward-open conformation. This discovery not only delineates the molecular determinants necessary for potent inhibition but also serves as a blueprint for designing next-generation SGLT1 inhibitors that leverage similar binding interactions. LX2761’s selective action in the intestinal lumen offers the promise of minimizing systemic side effects while effectively reducing postprandial glucose uptake.

In addition to LX2761, mizagliflozin has emerged as another selective SGLT1 inhibitor. Mizagliflozin has been investigated in preclinical studies, notably in a mouse model of small vessel disease, where it showed improvements in vascular cognitive impairment by specifically targeting SGLT1 without influencing SGLT2 activity. The molecule’s selective profile and its effectiveness in reversing certain cognitive deficits associated with altered cerebral blood flow hint at broader therapeutic applications beyond glycemic control.

The timeline of these discoveries reflects a rapid progression from early computational predictions to preclinical models and, potentially soon, to clinical evaluations. From patents disclosing early inhibitor scaffolds around 2010–2014, to more recent drug candidates leveraging state-of-the-art structural and computational methodologies, the landscape of SGLT1 inhibitors is evolving quickly. These molecules are not only structurally diverse but are also being optimized for better selectivity, lower accumulation in the body, and minimal off-target effects—a critical attribute given the widespread expression of SGLT1 in various tissues.

Mechanisms of Action
The mechanisms by which new SGLT1 inhibitor molecules exert their effects have been elucidated through various structural, computational, and experimental studies. Central to their design is the principle of competitive inhibition at the substrate-binding site of SGLT1. Many of these inhibitors are structured to mimic, at least partially, the natural substrate (glucose or galactose) in order to engage key interactions within the transporter’s active site. For instance, LX2761 binds deep into the substrate-binding pocket, thereby preventing glucose occupancy and locking the protein in an outward-open conformation, which essentially halts the transport cycle.

Other molecules, such as the 4-isopropylphenylglucitole compound, appear to act through similar competitive means. They achieve high binding affinities by forming multiple hydrogen bonds and hydrophobic interactions within the active site, ensuring that their occupancy effectively displaces the natural substrate. The selectivity toward SGLT1 over SGLT2 is achieved by designing these molecules to interact with residues that are unique or more accessible in the structure of SGLT1. This level of molecular precision not only underscores the success of structure-activity relationship (SAR) studies but also guides further modifications in future generations of inhibitors.

Computational approaches have further revealed quantitative insights into the binding free energies of these novel molecules, correlating predicted stability with inhibitory potency. For example, compounds identified from virtual screening pipelines, including compounds 81 and 91, were demonstrated to have favorable free energy calculations, suggesting robust binding. These free energy evaluations are critical, as they offer a direct measure of the strength and durability of the inhibitor–transporter complex. Proteochemometric models have helped in recognizing key structural features—such as specific aromatic or alkyl side chains—that optimize interactions with the SGLT1 binding site.

Furthermore, many of these inhibitors are designed to operate locally in the gastrointestinal tract. This localization is achieved by incorporating polar groups or labile bonds that limit systemic absorption, thereby confining the inhibitory effect to the gut, where controlling the uptake of dietary glucose is most needed. Such an approach contrasts with systemic SGLT2 inhibitors that act in the kidney. By restricting the inhibitor to the intestinal lumen, it is possible to minimize adverse systemic effects while directly targeting the physiological role of SGLT1 in glucose absorption.

In summary, the mechanisms of action for these new molecules are twofold: first, they competitively inhibit the binding of glucose at the transporter’s substrate-binding site; second, they stabilize the protein in a conformation that is non-permissive for further glucose transport. These dual actions account for their potent ability to reduce postprandial glucose levels and offer a template for future chemical refinement.

Clinical Applications and Benefits
The clinical application of new SGLT1 inhibitors is of significant interest because of their potential to address unmet needs in the management of diabetes and metabolic disorders. By directly reducing dietary glucose absorption, these inhibitors can complement existing therapies and provide an innovative mechanism to control hyperglycemia, particularly postprandial spikes, which are critical in the progression of type 2 diabetes.

Therapeutic Uses of SGLT1 Inhibitors
The primary therapeutic application of SGLT1 inhibitors lies in their ability to moderate postprandial blood glucose levels. Unlike SGLT2 inhibitors, which promote urinary glucose excretion by blocking renal reabsorption, SGLT1 inhibitors act in the gut to attenuate the absorption of dietary sugars. This is particularly beneficial for patients who struggle with postprandial hyperglycemia—a condition that contributes to chronic glycemic variability and increases the risk of vascular complications. For instance, the 4-isopropylphenylglucitole compound has been designed to specifically reduce glucose uptake in the small intestine, thereby lowering the overall glycemic load following meals.

In addition, early clinical investigations and preclinical models suggest that SGLT1 inhibitors may have a role in preventing the onset of diabetes. By limiting the surge of glucose after ingestion, these drugs can reduce the overall insulin demand on pancreatic β-cells, potentially preserving pancreatic function over the long term. Some patents and experimental reports highlight that compounds such as the molecule of Formula I and LX2761 could be especially useful in these scenarios because they exhibit favorable pharmacological profiles that limit systemic side effects while maximizing local intestinal effects.

Moreover, SGLT1 inhibitors are being explored not only for glycemic control but also for their potential benefits in weight management. Since inhibition of SGLT1 leads to reduced caloric intake via decreased carbohydrate absorption, patients might experience modest weight loss. This could serve as an ancillary benefit for individuals with type 2 diabetes, where weight reduction is often an important component of comprehensive management strategies.

Potential Benefits in Treating Diseases
Beyond the well-recognized benefits in glycemic control, SGLT1 inhibitors may offer a broader spectrum of therapeutic advantages. In addition to improving postprandial glycemia, the local action of these inhibitors in the gastrointestinal tract potentially reduces the burden of glucose-induced oxidative stress and inflammation in the gut. This, in turn, may contribute to the prevention of gastrointestinal disorders that are sometimes associated with diabetes.

Furthermore, by reducing the flux of glucose into the circulation after meals, SGLT1 inhibitors may indirectly protect against microvascular and macrovascular complications. Chronic hyperglycemia is a known risk factor for diabetic cardiomyopathy, nephropathy, and retinopathy; hence, effective modulation of glucose absorption might slow the progression of these complications. Animal model studies with molecules like mizagliflozin have shown reversal of certain deleterious biological markers, including improvements in cell survival and reductions in proinflammatory cytokine gene expressions in the brain and possibly in other tissues.

Another potential clinical application lies in the treatment of metabolic syndrome. Since SGLT1 inhibitors reduce the absorption of sugars, they can help manage insulin resistance—a central component of metabolic syndrome—and consequent cardiovascular risk. Moreover, some of these molecules are expected to have a dual inhibitory effect when combined with SGLT2 inhibition, paving the way for new therapeutic paradigms that simultaneously target both intestinal glucose uptake and renal reabsorption. Such dual SGLT1/2 inhibitors are already in various stages of clinical development, thus expanding the therapeutic toolbox available for personalized treatment of diabetes and its complications.

In summary, these new SGLT1 inhibitors, with their novel molecular scaffolds and advanced mechanisms of targeting, hold the potential to substantially alter clinical practice in diabetes management by offering an additional pathway to control blood sugar—and by doing so with a focus on minimizing systemic side effects.

Challenges and Future Directions
Despite the promising data and the exciting potential of these new molecules, several practical challenges and areas for future research remain. Addressing these challenges through continued innovation and rigorous clinical testing will be crucial for the successful translation of these compounds into routine clinical use.

Current Challenges in Development
One of the primary challenges in the development of SGLT1 inhibitors is achieving high selectivity for SGLT1 over SGLT2. Given that both transporters share ancestral structural similarities and overlapping substrate specificities, designing molecules that exclusively target SGLT1 requires precise knowledge of the subtle differences in their binding sites. Although recent cryo-EM studies—such as the one detailing LX2761’s binding mechanism—offer valuable insights, further refinement is necessary to balance potency with selectivity.

Additionally, optimizing the pharmacokinetic profiles of these molecules is essential to ensure that they act predominantly within the intestinal lumen while minimizing systemic absorption. This is particularly important because systemic exposure to SGLT1 inhibitors could lead to unintended off-target effects, given the expression of SGLT1 in tissues outside the gut, such as the heart and lungs. Molecules like the 4-isopropylphenylglucitole compound have been designed with low bioaccumulation in mind, but their long-term safety profiles require thorough investigation in clinical trials.

Another challenge is the dosage optimization and formulation development. Achieving the right concentration in the gut to inhibit SGLT1 without affecting SGLT2 or causing gastrointestinal disturbances is critical. Clinical development must address these issues with robust trial designs and careful monitoring of adverse effects, such as potential alterations in the gut microbiome or unexpected impacts on nutrient absorption.

Furthermore, while preclinical models and in vitro studies provide encouraging data on the efficacy of these molecules, there remains a gap between laboratory findings and clinical outcomes. Many of the novel compounds, including those identified through virtual screening and proteochemometric modeling (e.g., compounds 81 and 91), have not yet progressed to large-scale clinical evaluations. Bridging this translational gap is one of the pressing challenges for upcoming research efforts.

Finally, as many of these compounds represent first-in-class agents, regulatory pathways and guidelines specifically tailored for SGLT1 inhibitors are still evolving. Regulatory bodies may require additional data regarding long-term safety and efficacy, particularly because these drugs may be used in combination with other therapies for complex metabolic diseases.

Future Research and Development Trends
Looking ahead, future research will likely leverage advancements in structural biology, computational chemistry, and high-throughput screening to further optimize SGLT1 inhibitors. Researchers are expected to expand efforts in the following directions:

1. Refinement of Structure–Activity Relationships (SARs):
Future studies will focus on systematic modifications to the chemical scaffolds of late-generation SGLT1 inhibitors. By understanding the critical pharmacophores and specific molecular interactions that underpin the binding of inhibitors such as LX2761, medicinal chemists can design analogs with improved potency, selectivity, and minimal systemic absorption. This refinement process will also be informed by emerging structural insights from cryo-EM studies that detail the transporter’s conformational states.

2. Integration of Computational and AI-Driven Approaches:
The use of in silico models and machine learning tools, such as those employed in proteochemometric modeling, will continue to expedite the identification of promising candidate molecules. By screening vast chemical libraries and predicting binding affinities with high accuracy, future research can rapidly optimize hit compounds into lead candidates. This trend is already evidenced by studies that validated SGLT1 inhibitors with a hit rate exceeding 35% from virtual screenings.

3. Development of Dual or Combination Inhibitors:
Recognizing that SGLT1 and SGLT2 together orchestrate glucose homeostasis in the gut and kidney respectively, there is significant interest in developing dual inhibitors that can modulate both pathways synergistically. Although the focus of the current discussion is on selective SGLT1 inhibitors, future medicinal efforts might explore combination strategies where dual inhibition offers enhanced glycemic control with a balanced side-effect profile. Early-stage compounds from patents and clinical studies are already paving the way for such advancements.

4. Targeting Localized Drug Delivery:
Innovations in drug formulation and delivery systems will likely focus on ensuring that SGLT1 inhibitors exert their effects predominantly in the gastrointestinal tract. This may involve technologies such as enteric-coated formulations, pro-drugs activated within the gut, or localized delivery systems that minimize systemic exposure. Such strategies are critical for maximizing therapeutic benefits while reducing adverse effects from off-target interactions.

5. Expansion into Non-Glycemic Applications:
Beyond diabetes management, future research might explore the role of SGLT1 inhibitors in other conditions where altered glucose transport plays a role, such as metabolic syndrome, certain cardiovascular disorders, and even gastrointestinal diseases. The emerging data suggesting benefits in vascular cognitive impairment with mizagliflozin hints at a broader therapeutic potential that warrants exploration in diverse clinical settings.

6. Rigorous Clinical Evaluation and Post-Marketing Surveillance:
As more promising candidates move from preclinical studies to clinical trials, it will be essential to design robust, large-scale randomized controlled trials that can assess long-term outcomes, safety profiles, and comparative effectiveness. Post-marketing surveillance will also play a pivotal role in monitoring rare adverse events and ensuring that the benefits observed in controlled settings translate into everyday clinical practice.

7. Personalized Therapeutic Approaches:
Given the heterogeneity of type 2 diabetes and related metabolic disorders, future developments might also incorporate personalized medicine strategies. Genetic profiling and biomarker discovery could help identify patient subgroups more likely to benefit from SGLT1 inhibition, thereby optimizing treatment outcomes and reducing the risk of adverse effects. In this regard, the development of predictive models based on proteochemometrics might not only assist in drug discovery but also in patient stratification.

Conclusion
In conclusion, the emergence of new molecules for SGLT1 inhibition represents a significant advance in the pharmacotherapy of metabolic diseases, particularly type 2 diabetes mellitus. The new wave of SGLT1 inhibitors—including the 4-isopropylphenylglucitole compound, the novel compound of Formula I, LX2761, mizagliflozin, and computationally predicted molecules such as compounds 81 and 91—exemplifies the diverse approaches that researchers are employing in this field. These molecules have been designed using a blend of rational chemical design, high-throughput computational methods, and innovative proteochemometric modeling to yield compounds capable of selectively inhibiting intestinal glucose uptake. Their mechanisms of action are rooted in competitive inhibition at the substrate-binding site of SGLT1, with some agents, such as LX2761, uniquely locking the transporter in an outward-open conformation.

Clinically, these molecules offer several therapeutic advantages. They can reduce postprandial hyperglycemia by limiting dietary glucose uptake, potentially delay the onset of diabetes, and contribute to weight management and the prevention of vascular complications. Their localized activity in the gut may also minimize systemic side effects that are sometimes observed with systemic SGLT2 inhibitors, setting the stage for a complementary role in the treatment of metabolic syndrome and cardiovascular dysregulation.

Despite these promising developments, challenges remain. Selectivity, optimization of pharmacokinetics, formulation design, translation from in vitro models to clinical efficacy, and regulatory hurdles are all areas that require focused attention in future research. Advancements in structural biology, artificial intelligence, and personalized medicine approaches are expected to drive the next generation of SGLT1 inhibitors. Future trends include the development of dual inhibitors that target both SGLT1 and SGLT2, localized drug delivery systems, and more comprehensive clinical trials to address long-term safety and efficacy.

Overall, the current landscape underscores a dynamic and evolving field where innovative molecules are reshaping the treatment paradigm for diabetes and metabolic disorders. By employing a general‐specific‐general approach, we have seen that while the general role of SGLT1 inhibition is well established as central to controlling postprandial glucose absorption, specific molecular breakthroughs continue to expand our therapeutic options. This dual focus on enhancing our mechanistic understanding through structural insights and translating that knowledge into clinically viable drugs holds great promise for the future of metabolic disease management.

To summarize, the new molecules for SGLT1 inhibition are characterized by their innovative design, high selectivity, and advanced mechanisms of action that directly target the glucose absorption process in the gut. Their development, driven by a combination of traditional medicinal chemistry and cutting-edge computational techniques, marks a significant step forward in the quest for improved therapies for type 2 diabetes and related metabolic disorders. Continued research, both in preclinical models and clinical settings, will be essential to overcome current challenges and ensure that these novel agents achieve their full potential in delivering meaningful clinical benefits.

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.