Small Molecule
C25H29ClO6
1800115-22-3
Janagliflozin (DrugBank ID: DB16209) is a novel, orally administered, selective small-molecule inhibitor of the sodium-glucose cotransporter 2 (SGLT2). Developed by Sihuan Pharmaceutical's subsidiary, Huisheng Biopharmaceutical, it is designated as a national Class 1 innovative drug in China.[1] In January 2024, Janagliflozin, under the trade name Huiyoujing®, received drug registration approval from the National Medical Products Administration (NMPA) of China for the treatment of Type 2 Diabetes Mellitus (T2DM). This approval covers its use as both monotherapy and in combination with metformin.[1] The drug remains investigational in other global regions.[2]
Consistent with its therapeutic class, Janagliflozin exerts its antihyperglycemic effect through an insulin-independent mechanism. It selectively inhibits SGLT2 in the proximal renal tubules, which reduces the reabsorption of glucose from the glomerular filtrate, thereby promoting urinary glucose excretion (glucosuria) and lowering plasma glucose concentrations.[5] This mechanism confers a low intrinsic risk of hypoglycemia.
Pivotal Phase 3 clinical trials conducted in Chinese patients with T2DM have demonstrated robust efficacy. As a monotherapy in drug-naive patients, Janagliflozin at 25 mg and 50 mg daily doses achieved statistically significant, placebo-adjusted glycated hemoglobin (HbA1c) reductions of -0.80% and -0.88%, respectively.[7] When used as an add-on therapy to metformin in patients with inadequate glycemic control, both the 25 mg and 50 mg doses provided an identical placebo-adjusted
HbA1c reduction of -0.58%.[8] This glycemic efficacy is accompanied by clinically significant pleiotropic benefits, including reductions in body weight and systolic blood pressure, which are characteristic of the SGLT2 inhibitor class.[7]
The drug exhibits a favorable pharmacokinetic profile that supports a once-daily dosing regimen. It is characterized by rapid absorption, with a median time to maximum concentration (Tmax) of 2-6 hours, and a long terminal half-life (t1/2) of approximately 21-30 hours at steady state.[10] A key feature of its profile is that drug exposure is only minimally affected by renal impairment, suggesting pharmacokinetic stability in this important patient subgroup.[10]
In its clinical program, Janagliflozin was generally well-tolerated. No instances of severe hypoglycemia or diabetic ketoacidosis were reported, and the incidence of class-characteristic adverse events, such as urinary tract and genital mycotic infections, was reported to be low.[7]
Strategically, Janagliflozin's development was notably accelerated through the application of a Model-Informed Drug Development (MIDD) strategy, which successfully supported a waiver for a dedicated Phase 2 study.[14] Its approval marks it as the second domestically developed SGLT2 inhibitor in China, positioning Sihuan Pharmaceutical to compete in a rapidly expanding therapeutic market.[1]
Type 2 Diabetes Mellitus (T2DM) represents a significant and escalating global health crisis, with a particularly high prevalence and growing incidence in China. The management of T2DM has historically relied on therapeutic agents that primarily modulate insulin secretion or sensitivity. While effective, many older antidiabetic agents are associated with limitations such as the risk of hypoglycemia (e.g., sulfonylureas, insulin), weight gain (e.g., sulfonylureas, thiazolidinediones, insulin), and a progressive loss of efficacy over time due to the natural history of beta-cell decline. These limitations have driven the search for novel therapeutic approaches that can provide durable glycemic control with an improved safety profile and offer benefits beyond simple glucose lowering, particularly in the realms of cardiovascular and renal protection.
Janagliflozin emerged from this therapeutic landscape as a product of Sihuan Pharmaceutical Holdings Group, a major Chinese pharmaceutical manufacturer, through its non-wholly owned subsidiary, Huisheng Biopharmaceutical.[2] Positioned as a "national Class 1 innovative drug," Janagliflozin's development and approval signify a strategic pivot within China's pharmaceutical industry from a focus on generic manufacturing towards domestic innovation to address critical public health challenges.[1] As the second self-developed SGLT2 inhibitor to be approved and marketed in China, it represents a significant milestone for Huisheng Biopharmaceutical and a competitive entry into the rapidly growing domestic market for this drug class, which saw a 46% increase in sales in sample hospitals in 2022.[1] This development reflects a deliberate industrial strategy to meet the demands of a large patient population with a domestically originated therapeutic, thereby reducing reliance on foreign-developed pharmaceuticals and capturing a lucrative market.
Under normal physiological conditions, the kidneys play a crucial role in glucose homeostasis. Approximately 180 grams of glucose are filtered daily by the glomeruli and subsequently reabsorbed back into circulation via specialized transporters in the proximal convoluted tubules. This reabsorption process is mediated primarily by two sodium-glucose cotransporters: SGLT1 and SGLT2. SGLT2, a high-capacity, low-affinity transporter located in the early (S1) segment of the proximal tubule, is responsible for reabsorbing approximately 90% of the filtered glucose.[6] The remaining 10% is reabsorbed by SGLT1, a low-capacity, high-affinity transporter located further down the tubule. In patients with T2DM, the expression and activity of SGLT2 are pathologically upregulated, which exacerbates hyperglycemia by increasing the capacity for renal glucose reabsorption.[17]
The SGLT2 inhibitor class of drugs, including Janagliflozin, was developed to specifically target this renal mechanism. By competitively and selectively inhibiting the SGLT2 protein, these agents reduce the reabsorption of glucose from the glomerular filtrate.[5] This action effectively lowers the renal threshold for glucose (RTG), the plasma glucose concentration above which glucose begins to appear in the urine. The result is a significant increase in urinary glucose excretion (glucosuria), leading to a net loss of glucose and calories from the body.[17] A key feature of this mechanism is that it is independent of insulin secretion and beta-cell function, which confers two major advantages: a very low intrinsic risk of hypoglycemia when used as monotherapy, and continued efficacy across different stages of T2DM, even in patients with diminished beta-cell reserve.[20]
The therapeutic benefits of SGLT2 inhibition extend far beyond glycemic control. The induced glucosuria creates an osmotic diuresis, leading to a modest reduction in intravascular volume and blood pressure.[5] The associated caloric loss contributes to weight reduction. These hemodynamic and metabolic effects are believed to underpin the profound and now well-established cardiovascular and renal protective benefits of the class. Landmark clinical trials for other SGLT2 inhibitors have demonstrated significant reductions in major adverse cardiovascular events (MACE), hospitalizations for heart failure (HHF), and the progression of chronic kidney disease (CKD).[10] These findings have fundamentally transformed the role of SGLT2 inhibitors, elevating them from purely antihyperglycemic agents to foundational therapies in the management of heart failure and CKD, irrespective of a patient's diabetic status.
A comprehensive understanding of Janagliflozin begins with its precise chemical identity and physicochemical properties, which dictate its pharmacological behavior and suitability as an oral therapeutic agent.
Janagliflozin is classified as a small molecule drug.[25] Its structure is a C-glucoside derivative, a common feature of the gliflozin class, which provides stability against enzymatic degradation compared to O-glucosides. The molecule's precise stereochemistry and atomic connectivity are defined by its International Union of Pure and Applied Chemistry (IUPAC) name: (2S,3R,4R,5S,6R)-2-hexanyl]oxy]phenyl]methyl]-4-chlorophenyl]-6-(hydroxymethyl)oxane-3,4,5-triol.[2] This complex structure consists of a central glucose moiety linked via a carbon-carbon bond to a substituted aromatic system, which includes a bicyclo[3.1.0]hexane group. Its molecular formula is
C25H29ClO6, corresponding to a molar mass of 460.95 g·mol⁻¹.[2]
The physicochemical profile of Janagliflozin is consistent with that of a modern, orally bioavailable drug. Analysis of its structure reveals properties conducive to absorption and distribution. It has 6 hydrogen bond acceptors and 4 hydrogen bond donors, with a topological polar surface area (TPSA) of 99.38 A˚2.[25] The calculated partition coefficient (XLogP of 1.82) indicates a balance between hydrophilicity and lipophilicity necessary for membrane permeation and solubility.[27] These parameters contribute to its favorable "druglikeness" profile. The molecule adheres to Lipinski's Rule of Five, with zero violations reported, as well as the Ghose Filter and MDDR-like Rule, further supporting its suitability as an oral medication.[25]
The chemical synthesis of Janagliflozin has been described as a multi-step process designed for efficiency and scalability, which are critical for commercial manufacturing. The route involves the coupling of two advanced intermediates under cryogenic conditions using n-butyllithium (nBuLi).[3] Key transformations in the synthesis include a Friedel–Crafts acylation to form a ketone intermediate, followed by a reduction step using triethylsilane and boron trifluoride–diethyl etherate.[3] The final step is a hydrolysis of an acetylated precursor using lithium hydroxide monohydrate in a mixed solvent system, which proceeds with a high yield of 91%. The overall synthetic route is notably described as "protection-group-free," which simplifies the process, reduces the number of steps, and improves overall efficiency, making it well-suited for large-scale production.[3]
Property | Value | Source(s) |
---|---|---|
DrugBank ID | DB16209 | 2 |
Type | Small Molecule | 25 |
IUPAC Name | (2S,3R,4R,5S,6R)-2-hexanyl]oxy]phenyl]methyl]-4-chlorophenyl]-6-(hydroxymethyl)oxane-3,4,5-triol | 2 |
CAS Number | 1800115-22-3 | 2 |
Chemical Formula | C25H29ClO6 | 2 |
Molar Mass | 460.95 g·mol⁻¹ | 2 |
InChIKey | WDBIPGHUEJEKTC-VWQPKTIXSA-N | 2 |
Canonical SMILES | C1=C(C=CC(=C1)OC2C[C@H]3C[C@H]3C2)CC4=CC(=CC=C4Cl)[C@H]5C@@HO | |
Hydrogen Bond Acceptors | 6 | |
Hydrogen Bond Donors | 4 | |
Rotatable Bonds | 6 | |
Topological Polar Surface Area | 99.38 A˚2 | |
XLogP | 1.82 | |
Lipinski's Rule of Five | Yes (0 violations) |
The pharmacological profile of Janagliflozin defines its therapeutic effects and clinical utility. This profile is characterized by its specific mechanism of action, its quantifiable effects on physiological biomarkers (pharmacodynamics), and its absorption, distribution, metabolism, and excretion (pharmacokinetics) within the human body.
Janagliflozin's primary molecular target is the sodium-glucose cotransporter 2 (SGLT2), encoded by the SLC5A2 gene. As a highly selective inhibitor of this transporter, Janagliflozin functions by competitively binding to SGLT2 proteins located on the apical membrane of epithelial cells in the S1 segment of the proximal renal tubule. The drug's chemical structure includes a glucose-mimicking moiety, which facilitates this binding and blocks the transporter's normal function of reabsorbing glucose from the glomerular filtrate back into the bloodstream.
By inhibiting SGLT2, Janagliflozin effectively prevents the reabsorption of approximately 90% of the glucose that is filtered by the kidneys. This leads to a substantial increase in urinary glucose excretion (glucosuria) and a lowering of the renal threshold for glucose (RTG), the plasma glucose level at which glucosuria begins. The resulting therapeutic effects include a reduction in plasma glucose levels, a modest osmotic diuretic effect due to the increased solute load in the urine, and a net caloric loss, which contributes to observed reductions in body weight and systolic blood pressure. This entire mechanism is independent of pancreatic beta-cell function and insulin levels, which is a hallmark of the SGLT2 inhibitor class.
The pharmacodynamic (PD) effects of Janagliflozin have been well-characterized in clinical studies, demonstrating a clear and quantifiable impact on glucose handling.
Urinary Glucose Excretion (UGE): The primary PD effect of Janagliflozin is a dose-dependent increase in 24-hour UGE. In single-ascending dose studies in healthy Chinese subjects, the mean 24-hour UGE ranged from approximately 18 g to 85 g across doses from 10 mg to 450 mg. A critical finding is the saturation of this effect at higher doses. At steady state, mean 24-hour UGE in healthy subjects was approximately 40 g, 43 g, and 50 g for the 25 mg, 50 mg, and 100 mg doses, respectively, indicating a plateauing of the effect above the 25 mg dose. This observation was mirrored in T2DM patients, where multiple doses of 25 mg and 50 mg Janagliflozin produced nearly identical increases in 24-hour UGE from baseline (92.35 g and 94.17 g, respectively). These effects were comparable to those of dapagliflozin 10 mg (87.61 g). This PD plateau provides a strong mechanistic basis for the clinical finding that the 50 mg dose offers little to no additional glycemic benefit over the 25 mg dose in certain settings.
Glycemic and Metabolic Parameters: As a direct consequence of increased UGE, Janagliflozin treatment leads to significant reductions in both fasting plasma glucose (FPG) and 2-hour postprandial glucose (PPG).
Renal Function: Consistent with the SGLT2 inhibitor class, Janagliflozin can cause a transient, short-term reduction in the glomerular filtration rate (GFR) upon initiation. This effect is considered hemodynamic in nature, related to tubuloglomerular feedback, and typically stabilizes over time or reverses upon discontinuation of the drug. Importantly, Janagliflozin has been shown to maintain a significant pharmacologic effect, as measured by UGE, even in patients with moderate renal impairment (e.g., eGFR between 30 and 44 mL/min/1.73 m²), indicating that the drug remains active at the renal tubule despite reduced overall filtration.
The pharmacokinetic (PK) profile of Janagliflozin is well-suited for a once-daily oral antidiabetic agent.
Absorption, Distribution, Metabolism, and Excretion (ADME):
Pharmacokinetics in Special Populations:
Parameter | Healthy Subjects | T2DM Patients | Notes / Source(s) |
---|---|---|---|
Tmax (median) | 2.0–4.5 hours | 2.0–6.0 hours | Rapid oral absorption |
t1/2 at steady state | ~26–30 hours | ~21–23 hours | Supports once-daily dosing |
Dose Proportionality | Roughly dose-dependent increase in plasma exposure | Dose-proportional increase in plasma exposure | Consistent behavior across dose ranges |
Accumulation Factor | Not specified | < 2 | No significant accumulation with multiple doses |
Effect of Renal Impairment on AUC | N/A | ~11% increase in moderate RI vs. normal renal function | PK profile is stable in renal impairment |
Effect of Liver Cirrhosis on AUC (predicted) | N/A | Predicted to increase with severity of cirrhosis | PBPK modeling suggests caution in liver disease |
The clinical development of Janagliflozin was strategically designed to establish its efficacy and safety profile efficiently, primarily focusing on the Chinese population with T2DM. This program culminated in two pivotal Phase 3 trials that supported its regulatory approval in China.
The core of Janagliflozin's clinical evidence comes from two key Phase 3 studies: one evaluating its use as monotherapy in drug-naive patients (NCT03811548) and another assessing it as an add-on therapy to metformin (NCT03851432). Both trials were multicenter, randomized, double-blind, and placebo-controlled, with a 24-week primary efficacy period followed by a 28-week extension period, providing data over a total of 52 weeks.
A defining feature of Janagliflozin's development was the sophisticated application of a Model-Informed Drug Development (MIDD) strategy. This approach involved constructing mechanistic pharmacokinetic/pharmacodynamic (PK/PD) models using preclinical and Phase 1 data. These models were then integrated with data from a model-based meta-analysis (MBMA) of the broader SGLT2 inhibitor class to predict the drug's long-term efficacy on HbA1c in T2DM patients. The strength and reliability of these predictions were sufficient to convince regulators to grant a waiver for a traditional, large-scale Phase 2 dose-ranging study. This strategic use of computational modeling significantly accelerated the drug's development timeline, allowing it to proceed directly to Phase 3 trials after early-phase studies were completed. This represents a modern, efficient approach to drug development, reducing both time and cost to market.
This Phase 3 trial enrolled 432 drug-naive Chinese patients with T2DM who had inadequate glycemic control with diet and exercise alone. Patients were randomized to receive once-daily Janagliflozin 25 mg, Janagliflozin 50 mg, or placebo.
Primary Endpoint (Change in HbA1c at 24 weeks): The trial met its primary endpoint with high statistical significance.
Secondary Endpoints: Janagliflozin also demonstrated significant improvements across multiple secondary endpoints at 24 weeks.
This trial evaluated Janagliflozin in 421 Chinese patients with T2DM who were inadequately controlled on a stable dose of metformin monotherapy. Patients were randomized to receive once-daily Janagliflozin 25 mg, Janagliflozin 50 mg, or placebo, in addition to their ongoing metformin therapy.
Primary Endpoint (Change in HbA1c at 24 weeks): The results were again highly statistically significant.
The identical efficacy of the 25 mg and 50 mg doses in this add-on setting is a critical clinical finding. It suggests that in patients already receiving metformin, the additional glycemic benefit from SGLT2 inhibition is maximized at the 25 mg dose. This observation is mechanistically supported by the pharmacodynamic data showing a plateau in urinary glucose excretion at doses above 25 mg. This implies that for glycemic control in this combination, the 25 mg dose is the most efficient, and routine use of the 50 mg dose may be clinically redundant.
Secondary Endpoints: Similar to the monotherapy trial, Janagliflozin provided broad metabolic benefits when added to metformin.
The safety and tolerability of a new therapeutic agent are paramount considerations. The clinical development program for Janagliflozin provides a solid foundation for its safety profile, which is further contextualized by the extensive data available for the SGLT2 inhibitor class.
Across its pivotal Phase 3 trials, Janagliflozin was demonstrated to be generally well-tolerated.
Overall Adverse Event (AE) Incidence: In the 24-week monotherapy trial, the overall incidence of AEs was slightly higher in the active treatment groups compared to placebo but did not suggest a significant safety concern. The rates were 67.8% for the 25 mg group, 71.5% for the 50 mg group, and 60.7% for the placebo group.
Key Safety Endpoints:
While Janagliflozin's own trial data are favorable, prescribers must consider the established safety profile of the SGLT2 inhibitor class as a whole.
Based on the data for Janagliflozin and the established profile of the SGLT2 inhibitor class, the following contraindications and precautions can be inferred:
Adverse Event | Monotherapy Trial (24 Weeks) | Notes |
---|---|---|
Any Adverse Event (%) | Placebo: 60.7% Jana 25 mg: 67.8% Jana 50 mg: 71.5% | Overall incidence was comparable between active and placebo groups |
Severe Hypoglycemia (n) | 0 in all groups | Consistent with insulin-independent mechanism |
Diabetic Ketoacidosis (n) | 0 in all groups | No cases observed in the pivotal trial program |
Urinary Tract Infections | Incidence reported as "low" | A positive safety signal, but requires quantification from full study reports |
Genital Mycotic Infections | Incidence reported as "low" | A potential point of differentiation if confirmed in real-world use |
The appropriate use of Janagliflozin requires an understanding of its recommended dosing, administration, and application in specific patient populations, particularly those with common comorbidities like renal or hepatic disease.
Based on the successful outcomes of its Phase 3 clinical trials, Janagliflozin is administered orally as a once-daily tablet. The doses approved for use in China are 25 mg and 50 mg.
The clinical evidence provides clear guidance for dose selection. In the add-on to metformin trial, the 25 mg and 50 mg doses demonstrated identical efficacy in reducing HbA1c. This, combined with the pharmacodynamic data showing a plateau in urinary glucose excretion above the 25 mg dose, strongly suggests that
25 mg once daily is the optimal and most efficient therapeutic dose for the majority of patients requiring glycemic control. The 50 mg dose did show a slight numerical advantage in the monotherapy trial, but its lack of additional benefit in the more common combination-therapy setting makes the 25 mg dose the logical starting and standard maintenance dose.
The use of any antidiabetic medication must be carefully considered in populations with altered physiology or limited clinical data.
Renal Impairment: Janagliflozin exhibits a particularly favorable profile for use in patients with chronic kidney disease (CKD), a common comorbidity in T2DM.
Hepatic Impairment: There are no direct clinical trial data for Janagliflozin in patients with hepatic impairment. However, physiologically based pharmacokinetic (PBPK) modeling predicts that drug exposure increases with the severity of liver cirrhosis. This suggests that a careful risk-benefit assessment is necessary for patients with liver disease. For comparator SGLT2 inhibitors, prescribing information generally indicates no dose adjustment is needed for mild-to-moderate hepatic impairment, but use is not recommended in patients with severe impairment. A similar approach would be prudent for Janagliflozin.
Geriatric Population: Specific studies of Janagliflozin in the elderly have not been published. However, extensive data for the SGLT2 inhibitor class show that these drugs are effective and generally safe in older adults. The primary concern in this population is an increased risk of volume depletion-related adverse events, such as dehydration, postural dizziness, and hypotension, due to age-related declines in renal function and fluid homeostasis. Therefore, while Janagliflozin can likely be used effectively in the elderly, careful monitoring of volume status and renal function is warranted.
Pediatric Population: There are no data on the use of Janagliflozin in pediatric patients. Recently, other SGLT2 inhibitors like empagliflozin and dapagliflozin have received regulatory approval from the FDA for use in children aged 10 years and older with T2DM. These approvals were based on dedicated pediatric trials. However, these studies also noted a higher risk of hypoglycemia in the pediatric population compared to adults. The use of Janagliflozin in children is not established and would require dedicated clinical trials.
Pregnancy and Lactation: No clinical data are available for Janagliflozin in pregnant or lactating women. The SGLT2 inhibitor class is not recommended for use during pregnancy, particularly during the second and third trimesters. Animal studies have shown adverse effects on renal development during periods corresponding to these trimesters in humans. Similarly, because the drugs are excreted into animal milk and human kidney maturation continues for the first two years of life, there is a potential risk to the developing infant kidney. Therefore, breastfeeding is not recommended while taking SGLT2 inhibitors.
To fully appreciate Janagliflozin's clinical value and market potential, it must be evaluated within the context of the highly competitive SGLT2 inhibitor landscape, which is dominated by globally established agents such as dapagliflozin, empagliflozin, and canagliflozin.
A key study in Janagliflozin's development program was a head-to-head trial in Chinese patients with T2DM that directly compared Janagliflozin (25 mg and 50 mg) with dapagliflozin 10 mg. The results of this study are central to its initial positioning. The trial concluded that Janagliflozin exhibited favorable pharmacokinetic, pharmacodynamic, and tolerability profiles that were
comparable to those of dapagliflozin 10 mg. In terms of the primary pharmacodynamic endpoint, both the 25 mg and 50 mg doses of Janagliflozin produced a slightly higher mean 24-hour urinary glucose excretion (UGE) compared to dapagliflozin 10 mg (approximately 92-94 g vs. 88 g, respectively). This direct comparison establishes Janagliflozin as a non-inferior, and potentially slightly more potent in terms of glucosuria, alternative to a leading SGLT2 inhibitor, at least within the studied population.
While direct comparative data with empagliflozin and canagliflozin are not available, an indirect comparison can be made based on their respective clinical trial data.
Janagliflozin's current market strategy appears to be a "fast-follower" approach, focused on establishing a strong presence within the domestic Chinese market. This strategy leverages several key advantages: its status as a domestically developed innovative drug, a clinical profile that is comparable to established multinational brands, and a development pathway that was significantly accelerated by the use of MIDD. This allows it to compete effectively against existing, and likely more expensive, foreign-developed drugs for the primary indication of glycemic control in T2DM.
The company's ownership of international patents in the United States, Europe, Japan, and other regions indicates a long-term ambition for global expansion. However, its future outside of China is entirely contingent on its ability to generate the type of evidence that now defines the SGLT2 inhibitor class. To be competitive on a global scale, Janagliflozin will need to undergo its own large-scale CVOT. Without data demonstrating, at a minimum, cardiovascular non-inferiority, and ideally, superiority on hard cardiorenal endpoints, it will be unable to compete for the lucrative and clinically vital indications in heart failure and chronic kidney disease, which are now the primary drivers of SGLT2 inhibitor prescribing in many parts of the world.
Janagliflozin is a potent, selective, and orally active SGLT2 inhibitor that has demonstrated robust glycemic and metabolic efficacy in patients with Type 2 Diabetes Mellitus. Clinical data from its Phase 3 program, conducted within the Chinese population, establish its non-inferiority to established agents in its class, such as dapagliflozin, for the primary endpoint of glucose lowering. Its favorable pharmacokinetic profile, characterized by a long half-life suitable for once-daily dosing and notable stability in patients with moderate renal impairment, is a significant clinical advantage. Furthermore, its development was distinguished by the innovative and successful application of a Model-Informed Drug Development strategy, which accelerated its path to market. The safety profile observed in clinical trials appears favorable, with no reports of severe hypoglycemia or diabetic ketoacidosis and a reportedly low incidence of common class-related adverse events.
The approval of Janagliflozin in China represents a major achievement for Sihuan Pharmaceutical and its subsidiary Huisheng Biopharmaceutical. It validates their capacity for innovative drug development and positions them to compete effectively in the large and growing domestic diabetes market. The current strategy appears focused on leveraging its status as a domestically developed alternative to established multinational products for T2DM management.
While Janagliflozin is a clinically effective and safe treatment for T2DM, its long-term success and global potential are contingent on further strategic development. The following recommendations are proposed:
In its current state, Janagliflozin is a valuable addition to the therapeutic armamentarium for T2DM in China. It possesses the fundamental pharmacological characteristics of a drug with significant global potential. However, realizing this potential will require a substantial commitment to generating the high-level, long-term outcomes data that have become the hallmark of the SGLT2 inhibitor class.
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.