v
Search
Advanced

Publications > Journals > Journal of Clinical and Translational Hepatology> Article Full Text

  • OPEN ACCESS

Repurposing SGLT2 Inhibitors for Cirrhotic Ascites: From Mechanistic Research to Clinical Exploration

  • Yuan Gao1,#,
  • Yunyi Gao2,#,
  • Dong Ji3,*  and
  • Zhongjie Hu1,* 
 Author information 

Abstract

Cirrhotic ascites develops when portal hypertension and arterial under-filling chronically activate neuro-hormonal pathways that drive renal sodium-water retention. Augmented proximal tubular sodium reabsorption, predominantly mediated by the apical sodium/hydrogen exchanger 3 (NHE3), plays a fundamental role in this process. Given the spatial coupling of NHE3 and the sodium-glucose cotransporter 2 (SGLT2), selective SGLT2 inhibition reduces NHE3 activity via functional suppression within the apical microdomain. The increased sodium chloride delivery to the macula densa augments tubuloglomerular feedback and modulates the renin–angiotensin–aldosterone system. Early clinical investigations, ranging from case reports and retrospective analyses to pilot randomized trials, indicated potential benefits in controlling ascites and reducing decompensation events. However, their limited sample size, heterogeneous endpoints, and predominantly observational design constrain the generalizability of the findings. This review concentrates on the molecular mechanisms and emerging clinical evidence supporting the therapeutic potential of SGLT2 inhibitors in the management of cirrhotic ascites.

Graphical Abstract

Keywords

SGLT2 inhibitors, Cirrhosis, Ascites, Natriuresis, Renin–angiotensin–aldosterone system, RAAS, NHE3

Introduction

In cirrhosis, portal hypertension and splanchnic arterial vasodilation chronically activate neurohumoral systems, including the renin–angiotensin–aldosterone system (RAAS), the sympathetic nervous system, and arginine vasopressin. Neurohormonal activation drives augmented tubular sodium reabsorption, and the proximal tubule acts as a dominant effector.1 Augmented proximal tubular sodium reabsorption not only promotes ascites formation but also reduces the luminal sodium gradient available to distal nephron channels, thereby diminishing the effectiveness of diuretics, a phenomenon commonly referred to as diuretic resistance.2 Clinically, the development of ascites marks a notable transition from compensated to decompensated cirrhosis. Each year, approximately 5–10% of patients with compensated cirrhosis progress to ascites.3 Once ascites develops, the five-year mortality increases to about 44%.4 Outcomes are even worse in patients with refractory ascites, in whom early referral for liver transplantation should be considered.5

Mechanistically, the sodium/hydrogen exchanger 3 (NHE3) is the principal mediator of proximal tubular sodium reabsorption.6 The spatial colocalization of NHE3 and the sodium–glucose cotransporter 2 (SGLT2) in the same apical microdomain provides a mechanistic rationale whereby pharmacologic inhibition of SGLT2 may concurrently reduce NHE3 activity, thereby attenuating excessive proximal sodium reabsorption. These insights support the development of proximal tubule–targeted therapeutic strategies.

This review integrates the current understanding of cirrhosis pathophysiology with proximal tubular mechanisms, highlights their contributions to ascites formation and diuretic resistance, and discusses the mechanistic basis and emerging clinical evidence supporting the use of SGLT2 inhibitors.

Disrupted sodium–water homeostasis: the core driver of ascites and the limitation of conventional diuretics

Under normal physiological conditions, the proximal tubule reabsorbs approximately 60% of filtered sodium, primarily via the NHE3, while the SGLT2 accounts for about 5% through 1:1 sodium–glucose coupling.7 In cirrhosis models or in states of reduced effective arterial volume, both transporters in the proximal tubule are upregulated, thereby reducing sodium delivery to the distal nephron.8 In the compensated stage, increased cardiac output partially counteracts splanchnic vasodilation, and neurohumoral activation remains mild. Once decompensation occurs, cardiac output declines, and sustained activation of the RAAS together with arginine vasopressin can markedly enhance proximal sodium–chloride reabsorption.9,10 In carbon tetrachloride (CCl4)-induced ascitic rats, proximal NHE3 expression levels and brush-border localization increase by 40–60%, exhibiting an inverse correlation with urinary sodium excretion.11 Clinically, patients with ascites demonstrate greater fractional proximal sodium reabsorption despite preserved eGFR, resulting in a diminished natriuretic response to furosemide compared with controls. After sodium chloride (NaCl) infusion, proximal reabsorption remains unchanged in decompensated cirrhosis, whereas it decreases by approximately 15% in healthy subjects.12 Thus, upregulation of proximal tubular Na+ transport underlies cirrhotic Na+ retention and contributes to the limited efficacy of standard diuretics; however, direct evidence of NHE3 alterations in cirrhotic patients remains limited.

First-line diuretic therapy for ascites typically includes spironolactone, either alone or in combination with a loop diuretic.3 Loop diuretics inhibit Na+–K+–2Cl cotransporter 2 (NKCC2) in the thick ascending limb, thereby increasing distal NaCl delivery and promoting natriuresis. However, simultaneous inhibition of NKCC2 at the macula densa (MD) reduces NaCl sensing, stimulates juxtaglomerular renin release, and further amplifies the already activated RAAS.13 The ensuing angiotensin II/aldosterone signaling upregulates sodium–chloride cotransporter (NCC) and epithelial sodium channel (ENaC), increases Na+ reabsorption, and induces the “braking phenomenon” during prolonged therapy, whereby the natriuretic response diminishes despite dose escalation.14,15

Molecular basis of SGLT2-mediated natriuresis

Heart failure and decompensated cirrhosis share a key pathophysiological feature, namely effective arterial hypovolemia. A reduction in cardiac output or systemic vascular resistance lowers arterial blood volume, leading to chronic activation of the sympathetic nervous system and the RAAS, which in turn enhances renal sodium reabsorption. The resulting fluid retention manifests as pulmonary or peripheral edema in heart failure, whereas it predominantly leads to ascites in cirrhosis.16 SGLT2 inhibitors improve cardiorenal outcomes in heart failure and are incorporated into the four foundational therapies for heart failure with reduced ejection fraction, comprising an ARNI (or ACE inhibitor/ARB), a β-blocker, a mineralocorticoid receptor antagonist, and an SGLT2 inhibitor. They have also been explored for fluid management in cirrhotic ascites.17

SGLT2 is highly expressed on the brush border of the proximal tubule and, as a Na+-dependent glucose transporter, mediates reabsorption of filtered glucose. It comprises 14 transmembrane helices arranged into a “rocking bundle” and a “stationary scaffold,” functioning in concert with its chaperone protein MAP17.18 Because Na+ reabsorption via SGLT2 is strictly stoichiometrically coupled to glucose transport, even RAAS-induced upregulation of SGLT2 increases total renal Na+ reabsorption by only 3–5%.19 Therefore, selective SGLT2 blockade alone would be expected to exert only a limited natriuretic effect.

NHE3, located on the luminal membrane of the proximal tubule, mediates Na+/H+ exchange. NHE3 and the SGLT2–MAP17 complex are organized and stabilized by the PDZ domain–containing protein PDZK1, forming a functional scaffold that coordinates sodium and glucose transport.20,21 Functional studies demonstrate that SGLT2 inhibition reduces proximal tubular sodium reabsorption in part by suppressing NHE3 activity, thereby amplifying natriuresis beyond what would be expected from direct blockade of SGLT2 alone.22 In non-diabetic rats with post-MI heart failure, in vivo microperfusion showed that the SGLT2 inhibitor empagliflozin directly inhibited proximal NHE3 activity while preserving GFR and restoring euvolemia (Fig. 1).23

Molecular mechanisms for the SGLT2 inhibitor–mediated reduction in proximal tubular sodium reabsorption.
Fig. 1  Molecular mechanisms for the SGLT2 inhibitor–mediated reduction in proximal tubular sodium reabsorption.

SGLT2, sodium glucose cotransporter 2; NHE3, sodium hydrogen exchanger 3; MAP17, membrane associated protein of 17 kDa; S1/S2, S1 and S2 segments of the proximal tubule; Na+, sodium ion; H+, hydrogen ion.

However, the precise molecular events governing this SGLT2–NHE3 interplay remain under investigation. While inhibitory phosphorylation of NHE3 was initially proposed as a primary pathway,22 recent data indicate a more complex picture. For instance, a study in healthy volunteers showed that a single dose of empagliflozin increased fractional sodium excretion without altering NHE3 phosphorylation or abundance in urinary exfoliated tubular cells.24 This suggests that the functional shift in proximal Na+ handling does not rely solely on phosphorylation. Furthermore, downstream transporter adaptation significantly shapes the phenotype during prolonged use. In normotensive rats, empagliflozin inhibited proximal NHE3 yet upregulated expression and phosphorylation of distal NCC; in hypertensive rats, NHE3 inhibition occurred without NCC upregulation.25 These findings support a network view in which distal compensation can partially offset proximal NHE3 inhibition under some backgrounds. Pair-feeding/drinking experiments showed that SGLT2 inhibition maintained a small but sustained increase in urinary sodium and water, producing a mild negative fluid balance that revealed an intrinsic natriuretic tone.26 In conclusion, current data do not support a universal increase in NHE3 phosphorylation; rather, this mechanism appears to be context-dependent and warrants further exploration, specifically within the cirrhotic environment.

Taken together, these findings indicate that inhibition of proximal tubular NHE3 is the dominant driver of SGLT2 inhibitor–induced natriuresis, and that the resulting increase in distal sodium delivery far exceeds the small amount of sodium that is directly cotransported with glucose by SGLT2 itself. In humans, a lithium clearance study demonstrated that SGLT2 inhibition diverts more than 7% of the glomerular filtrate Na+ load to the distal nephron, markedly exceeding the transporter’s theoretical 3–5% contribution.19,27 In line with these observations, clinical studies in heart failure have shown that natriuretic and glycosuric responses are not tightly positively correlated and may even be inversely related.28,29

Modulation of the SGLT2i-neurohumoral axis

By inhibiting proximal tubular sodium–glucose reabsorption, SGLT2 inhibitors increase NaCl delivery to the thick ascending limb, enhancing the ionic load sensed by the MD.30 The classical model attributes the activation of tubuloglomerular feedback (TGF) to an elevation in luminal NaCl concentration.31,32

Experimental evidence indicates that chloride, rather than sodium, is the principal trigger of the TGF response. TGF is abolished when the loop of Henle is perfused with an iso-osmotic, chloride-free solution,33 whereas replacement of luminal sodium with N-methyl-D-glutamine in the presence of chloride preserves TGF activation.34

SGLT2 inhibition also suppresses NHE3 activity, raising luminal bicarbonate concentrations in tubular fluid,35 which may lead to a disproportionate elevation in urinary Na+ relative to Cl. When the increase in distal tubular Cl is modest, the MD continues to sense a “low Cl” state, keeping TGF suppressed. Future studies should clarify how urinary chloride dynamics influence RAAS activity following SGLT2 inhibition.

Theoretically, increased chloride delivery to the MD can enhance TGF and suppress RAAS activity; however, clinical findings are more complex. Osmotic diuresis and modest natriuresis transiently reduce plasma volume, eliciting a short-lived increase in plasma renin activity, whereas aldosterone levels exhibit minimal or no change. Both parameters generally return to baseline with continued therapy.36,37 In randomized trials of SGLT2 inhibitors in heart failure, a decline in eGFR of 3–5 mL/min/1.73 m2 is mainly observed during the first weeks, while values generally return to baseline by 12 weeks and subsequently decline at a slower rate than in control subjects.38 Importantly, these heart failure–based trials have not demonstrated an increased incidence of orthostatic hypotension despite a modest 4–5 mm Hg reduction in systolic blood pressure.39–41

It is also necessary to take into account the complex effects of SGLT2 inhibition on neurohumoral regulation. For instance, in salt-loaded Dahl salt-sensitive rats, the SGLT2 inhibitor dapagliflozin blunted salt-induced hypertension and enhanced natriuresis without significantly altering circulating or intrarenal RAAS components.42 In this low-renin model, the drug primarily improved tubular sodium handling in a RAAS-independent manner. Similarly, in hypertensive BPH/2J mice, dapagliflozin lowered blood pressure in association with sympathoinhibition by reducing renal tyrosine hydroxylase and norepinephrine levels, suggesting a RAAS-independent neurohumoral modulation.43 Collectively, these studies illustrate that SGLT2 inhibitors do not universally modulate neurohumoral status via RAAS alone; rather, their effects are modulated by background renin status, salt intake, and sympathetic tone. Future studies in cirrhosis should therefore track these neurohumoral markers dynamically to determine which pathway predominates in the setting of advanced liver disease.

Effects of SGLT2 inhibitors on compensatory sodium reabsorption and sodium redistribution

Inhibition of proximal tubular sodium reabsorption by SGLT2 inhibitors is accompanied by compensatory adaptations in downstream nephron segments. In rodent models, the thiazide-sensitive NCC is consistently upregulated,25 whereas responses of the NKCC2 and the ENaC vary depending on the experimental model. In a proteomic study of 1,134 participants, SGLT2 inhibitor therapy was associated with upregulation of carbonic anhydrase isoforms and urotensin II. The former may enhance sodium–bicarbonate reabsorption in proximal and collecting tubules, while the latter can stimulate NKCC2 in the loop of Henle, providing a mechanistic explanation for the attenuation of natriuresis after several days of treatment.44 Evidence regarding ENaC is inconsistent. In diabetic rats, empagliflozin reduces α- and γ-ENaC expression,45 whereas in normotensive rats treated for 14 days, ENaC remains unchanged while NCC is upregulated, highlighting model-dependent effects.25

In addition to their natriuretic effects, SGLT2 inhibitors may influence overall body sodium distribution, including a reduction in non-osmotic tissue sodium. A randomized controlled trial (RCT) utilizing 23Na magnetic resonance imaging (MRI) demonstrated that six weeks of dapagliflozin (10 mg once daily) reduced skin sodium concentration by approximately 20 mmol/L in adult patients with type 2 diabetes, whereas placebo had no effect. Consistently, in a cohort of 74 patients with heart failure, three months of empagliflozin similarly decreased sodium content in skin and skeletal muscle. Post hoc analysis of EMPA-KIDNEY confirmed that SGLT2 inhibitors could modify body sodium storage in skin and muscle tissues without influencing overall body weight or fat mass, supporting mobilization of non-osmotic sodium from the skin–fascia compartment.46

This sodium storage site is located in the skin and subcutaneous fascia, which are rich in negatively charged sulfated glycosaminoglycans.47 High aldosterone states activate the keratinocyte- and sweat duct–mineralocorticoid receptor/ENaC pathway, further promoting sodium binding to glycosaminoglycans.48 Decompensated cirrhosis, characterized by persistent RAAS activation and elevated aldosterone levels, is therefore expected to resemble that observed in heart failure, thereby contributing to ascites formation. Loop diuretics primarily remove intravascular and tubular sodium and water and have limited capacity to mobilize tissue sodium, which may render them less effective than SGLT2 inhibitors for sodium redistribution.49,50 To date, 23Na MRI data in patients with cirrhosis and refractory ascites are lacking. Imaging-based studies evaluating the effects of SGLT2 inhibitors and loop diuretics in this population represent a notable direction for future research.

Clinical evidence and safety profile of SGLT2 inhibitors for cirrhotic ascites

Since 2020, clinical evidence with SGLT2 inhibitors in cirrhotic ascites has evolved from single-patient reports and small case series to feasibility studies and pilot RCTs. The earliest publications consisted of single-patient case reports and very small series, including three patients with refractory ascites, one patient with intractable pleuroascites, and one patient dependent on ascites-concentrating reinfusion.17,51,52 More recently, Qin et al. documented complete resolution of pleuroascites and sustained natriuresis in another refractory case treated with empagliflozin.53 Another case report described two patients with diabetes mellitus and cirrhosis-related ascites who experienced sustained reductions in ascites volume after three years of SGLT2 inhibitor therapy, without serious adverse events.54 All patients exhibited remarkable improvement in fluid overload and hyponatremia following treatment with dapagliflozin or empagliflozin.17,51 In addition, a retrospective cohort analysis suggested that exposure to SGLT2 inhibitors was associated with fewer end-stage liver disease events, hospital readmissions, and paracenteses.55

Since 2024, several pilot trials have demonstrated that empagliflozin reduced both ascites volume and the need for therapeutic paracentesis without the emergence of new safety concerns.56–59 The first nationwide real-world analysis, involving approximately 10,000 adults, found that the use of SGLT2 inhibitors was associated with 32% fewer severe hepatic outcomes, 53% fewer episodes of hepatorenal syndrome, 46% fewer paracenteses, and a 33% reduction in hospitalization rates (Table 1).17,51–63 Although these findings are promising, the majority of existing studies provide only level III–IV evidence and are constrained by small sample sizes or retrospective design. This highlights the urgent need for large, multicenter RCTs stratified by Child–Pugh class and incorporating longitudinal RAAS profiling and urinary electrolyte assessments to establish definitive efficacy and mechanistic insights (Supplementary Table 1).

Table 1

Summary of clinical studies investigating SGLT2 inhibitors in cirrhotic ascites

Study typeSample sizePopulation characteristicsIntervention & follow-upMain findingsOCEBM levelLevel of evidenceMain limitationsChild-Pugh
1Case series (2020)173Refractory ascites and T2DMSGLT2i; 46 wAscites and edema resolved4IVSmall sample size; no controlC
2Case report (2021)511PBC + Refractory hydrothorax and ascitesEmpagliflozin 10 mg/d; 4 wResolution of ascites and pleural effusion4IVSingle case; short follow-upC
3PSM cohort (2021)55846Veterans,SGLT2i vs DPP4iSGLT2i use vs non-use; 36 mMortality reduced by 67%3IIIaRetrospective; potential confounding factorsNR
4Case report (2022)521Refractory ascitesEmpagliflozin 10 mg/d;28 dNo need for paracentesis, reduced ascites4IVSingle caseC
5Open label, phase I/II (2024)5710Decompensated cirrhosisEmpagliflozin 10 mg/d; 4 wGood pharmacokinetics and safety; alleviates ascites2bIIbSmall sample size; no controlB/C
6Pilot case series (2024)5814Refractory ascitesEmpagliflozin 10 mg/d;3 m24-hour urinary sodium increased; no need for paracentesis4IVSmall sample size; single centerC
7Retrospective study (2024)61300T2DM and ascitesDapagliflozin 10 mg/d(A/B)or 5 mg(C);6 mImproved glycemia control; reduced diuretic requirement3IIIaNon-randomized; ascites grading unavailableA/B/C
8RCT (2024)5942Refractory ascitesEmpagliflozin 10 mg/d; 3 mReduced need for paracentesis2IISmall sample size; open-labelC
9Case report (2025)624Refractory ascitesDapagliflozin 10 mg/d; 6 mAscites completely resolved4IVSmall sample size; no controlC
10RCT (2025)5640Recurrent ascitesDapagliflozin 10 mg/d vs placebo;6 mImproved ascites control; increased incidence of AKI2IISmall sample size; low event rateB/C
11PSM cohort (2025)6010 660Cirrhotic ascites required diureticSGLT2i+diuretic vs diuretic;3 yHRS reduced by 53%; paracentesis reduced by 46%3IIIaBased on ICD and EMR, lack of baseline dataB/C
12Single-arm study (2025)6314Diuretic-resistant ascitesEmpagliflozin 10mg/d; 12wwell-tolerated; improved diuresis3IIISmall sample size; no controlC
13Case report (2025)531ALD + Refractory hydrothorax and ascitesEmpagliflozin 10 mg/d; 6 mResolution of ascites and pleural effusion, Diuretics discontinued, urinary sodium increased4IVSingle caseC
14Case report (2025)542Refractory ascitesLuseogliflozin 5 mg/d; 3yResolution of ascites4IVSingle caseB

Across four large-scale RCTs of empagliflozin in patients with diabetes, heart failure, and chronic kidney disease published in The New England Journal of Medicine,40,64–66 the incidence rates of urinary tract infection, hypoglycemia, and hepatotoxicity were comparable to placebo, whereas the incidence of genital mycotic infections increased modestly (Table 2).40,64–66 The low risk of hypoglycemia with SGLT2 inhibitors suggests that they primarily act by lowering the renal glucose threshold rather than exerting a direct glucose-lowering effect,67 and non-diabetic participants excrete substantially less urinary glucose than patients with diabetes.68 Mechanistically, the increased downstream glucose load to the S2/S3 segments of the proximal tubule elicits a compensatory increase in SGLT1-mediated transport.69 This renal transport reserve provides a detailed explanation for the consistently low rates of hypoglycemia across diabetic individuals and patients with heart failure or CKD.

Table 2

Four large trials of empagliflozin

EMPA-REG OUTCOME66
EMPEROR-Reduced40
EMPEROR-Preserved64
EMPA-KIDNEY65
Empagliflozin (N = 4,687, 10 mg & 25 mg)Placebo (N = 2,333)Empagliflozin (N = 1,863)Placebo (N = 1,863)Empagliflozin (N = 2,996)Placebo (N = 2,989)Empagliflozin (N = 3,304)Placebo (N = 3,305)
Serious hypoglycemia63 (1.3)36 (1.5)27 (1.4)28 (1.5)73 (2.4)78 (2.6)77 (2.3)77 (2.3)
Serious urinary tract infection82 (1.7)41 (1.8)19 (1.0)15 (0.8)57 (1.9)45 (1.5)52 (1.6)54 (1.6)
Genital infection301 (6.4)42 (1.8)31 (1.7)12 (0.6)67 (2.2)22 (0.7)NRNR
Serious genital infection301 (6.4)42 (1.8)6 (0.3)5 (0.3)8 (0.3)8 (0.3)1 (<0.1)1 (<0.1)
Serious hyperkalemiaNRNRNRNRNRNR92 (2.8)109 (3.3)
Serious acute kidney injury45 (1.0)37 (1.6)NRNRNRNR107 (3.2)135 (4.1)
Liver injuryNRNRNRNR115 (3.8)155(5.2)13 (0.4)12 (0.4)
Hypotension239 (5.1)115 (4.9)176 (9.4)163 (8.7)311 (10.4)257(8.6)NRNR

Neither randomized trials nor real-world studies have demonstrated an increased incidence of urinary tract infections, possibly because enhanced urine flow mitigates the glucosuria-associated infectious risk.70 In contrast, SGLT2 inhibitors are consistently associated with a higher incidence of genital mycotic infections. In individuals without prior genital disease, the annual risk is approximately 10.8% in women and 2.7% in men,71 rates that are similar to those observed in randomized trials. The most frequent manifestations are candidal vulvovaginitis in women and balanitis or posthitis in men, which generally resolve rapidly with standard antifungal therapy or temporary discontinuation of SGLT2 inhibitors. Given that cirrhosis predominantly affects men, the overall risk–benefit profile remains promising for the use of SGLT2 inhibitors in this population; nevertheless, genital infection should be prespecified as an important safety endpoint in future randomized trials. Importantly, existing large RCTs of SGLT2 inhibitors included few patients with decompensated cirrhosis, so complications that are particularly relevant in this population, such as the risk of spontaneous bacterial peritonitis, potential alterations in gut microbiota due to glucosuria, and the risk of hepatic encephalopathy, have been insufficiently characterized. These risks should be prioritized as key safety endpoints in future trials.

In relation to ammonia metabolism, preclinical studies have indicated that SGLT2 inhibition could enhance renal ammonium excretion through NHE3 inhibition and Rhcg upregulation.22 In contrast, the DAPASALT study reported a transient decline in fractional urea excretion, which subsequently normalized promptly,72 indicating that the overall impact of SGLT2 inhibition on ammonia handling remains unresolved.

Hyponatremia represents a frequent complication of decompensated cirrhosis, raising concern that natriuresis induced by SGLT2 inhibition could exacerbate this disturbance. Nevertheless, data from large-scale randomized trials in heart failure, supported by post hoc evidence, demonstrated that SGLT2 inhibitors do not increase the incidence of hyponatremia and may improve serum sodium concentrations,73 with similar findings also reported in cirrhotic cohorts.59 Accordingly, future randomized trials should prospectively designate serum sodium as a prespecified safety endpoint and stratify analyses according to baseline sodium concentration and Child–Pugh classification to refine the assessment of risks and benefits associated with SGLT2 inhibition in the management of cirrhotic ascites.

In hypertensive populations, SGLT2 inhibitors lower blood pressure by approximately 3–5 mmHg.74 In decompensated cirrhosis, baseline blood pressure is often low; therefore, the risk of symptomatic or orthostatic hypotension should be carefully evaluated, ideally with ambulatory or wearable blood pressure monitoring as a safety endpoint in future trials.75,76

Finally, the hemodynamic effects of SGLT2 inhibitors on the hepatic circulation warrant careful consideration. Although SGLT2 inhibitors have been shown to reduce portal pressure in animal models of cirrhosis, as well as reduce splanchnic congestion, potentially via antifibrotic mechanisms,77 direct clinical data in patients with decompensated cirrhosis are lacking. There is a theoretical concern that excessive hypovolemia could reduce hepatic perfusion in patients with already compromised effective arterial blood volume. Future trials must balance the benefits of ascites resolution against the potential risk of worsening hepatic ischemia or hepatorenal syndrome, particularly in patients with unstable hemodynamics.

Future research perspectives

Future research should aim to close mechanistic gaps while establishing a translational framework that connects molecular pathways to patient-centered outcomes. Priorities include quantifying the degree of SGLT2–NHE3 coupling and the context-dependent contribution of NHE3 phosphorylation, delineating how SGLT2 inhibition modulates the RAAS and sympathetic tone, and defining the net impact on proximal and nephron-wide sodium reabsorption. The application of 23Na MRI to assess sodium and water retention in cirrhosis, as well as dynamic changes in therapeutic response, represents a promising approach.

From the perspective of drug development, structural and genetic data support that SGLT2 functions in the proximal tubule as a complex with its accessory protein MAP17. MAP17 is required for full SGLT2 activity, and loss-of-function variants in MAP17 can cause familial renal glucosuria despite an intact SLC5A2 coding sequence, underscoring the functional importance of this microdomain. Recent cryo-electron microscopy structures of the human SGLT2–MAP17 complex bound to clinical inhibitors21 provide a framework to design microdomain-targeted or allosteric modulators that might preserve NHE3-mediated natriuresis while inducing less glucosuria. Although such “natriuretic-biased, low-glucosuria” agents remain purely hypothetical at present.

In parallel, a translational pathway is needed to connect mechanistic insights with clinical outcomes. The analytical validity and clinical utility of candidate mechanistic biomarkers should be established, including urinary chloride and urinary exosomal phosphorylated NHE3 for treatment monitoring. An initial clinical step may involve exploratory studies in patients with refractory ascites and impaired urinary sodium excretion, with fractional excretion of sodium and neurohumoral markers as primary endpoints.78 If SGLT2 inhibition exhibits sustained natriuretic efficacy in this context, it would warrant progression to multicenter RCTs with clinically relevant endpoints, including hospital readmission, paracentesis frequency, and all-cause mortality. Finally, given that the natriuretic mechanism of SGLT2 inhibition is likely to be broadly shared across agents in this class, and that the domestically developed SGLT2 inhibitor henagliflozin has already entered clinical studies in heart failure,79 future research should also consider evaluating henagliflozin for the treatment of ascites in patients with cirrhosis.

Conclusions

SGLT2 inhibitors attenuate proximal tubular sodium reabsorption through a proximal microdomain comprising SGLT2, MAP17, and NHE3. Downstream effects include modulation of the RAAS and sympathetic nervous system, potentially redistributing tissue sodium. These mechanisms directly address the low distal sodium delivery that underlies diuretic resistance in cirrhotic ascites. Early clinical observations indicate promising effects on natriuresis and reductions in ascites burden, with a safety profile consistent with experience from other trials involving SGLT2 inhibitors. Although the current evidence remains preliminary and limited, a coherent translational pathway from “mechanistic rationality” to “clinical feasibility” has been initially established, providing a solid basis for subsequent high-quality clinical validation.

Supporting information

Supplementary Table 1

Search strategy.

(DOCX)

Declarations

Acknowledgement

We thank Medjaden for its services and assistance in improving the quality of our manuscript.

Funding

This work was supported by the Beijing Hospitals Authority Clinical Medicine Development Special Funding Support (ZLRK202533).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Study conception, outline of the manuscript (YuanG), drafting of the manuscript (YuanG, YunG), figure preparation, literature search (YunG), critically revised the manuscript, final quality control (ZH, DJ). All authors have approved the final version and publication of the manuscript.

References

  1. Aithal GP, Palaniyappan N, China L, Härmälä S, Macken L, Ryan JM, et al. Guidelines on the management of ascites in cirrhosis. Gut 2021;70(1):9-29 View Article
  2. Dominguez Rieg JA, Xue J, Rieg T. Tubular effects of sodium-glucose cotransporter 2 inhibitors: intended and unintended consequences. Curr Opin Nephrol Hypertens 2020;29(5):523-530 View Article PubMed/NCBI
  3. European Association for the Study of the Liver. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. J Hepatol 2018;69(2):406-460 View Article PubMed/NCBI
  4. Biecker E. Diagnosis and therapy of ascites in liver cirrhosis. World J Gastroenterol 2011;17(10):1237-1248 View Article PubMed/NCBI
  5. Macken L, Corrigan M, Prentice W, Finlay F, McDonagh J, Rajoriya N, et al. Palliative long-term abdominal drains for the management of refractory ascites due to cirrhosis: a consensus document. Frontline Gastroenterol 2022;13(e1):e116-e125 View Article PubMed/NCBI
  6. Wilcox CS, Testani JM, Pitt B. Pathophysiology of Diuretic Resistance and Its Implications for the Management of Chronic Heart Failure. Hypertension 2020;76(4):1045-1054 View Article PubMed/NCBI
  7. Vallon V. State-of-the-Art-Review: Mechanisms of Action of SGLT2 Inhibitors and Clinical Implications. Am J Hypertens 2024;37(11):841-852 View Article PubMed/NCBI
  8. Bansal S, Lindenfeld J, Schrier RW. Sodium retention in heart failure and cirrhosis: potential role of natriuretic doses of mineralocorticoid antagonist?. Circ Heart Fail 2009;2(4):370-376 View Article PubMed/NCBI
  9. Salerno F, Guevara M, Bernardi M, Moreau R, Wong F, Angeli P, et al. Refractory ascites: pathogenesis, definition and therapy of a severe complication in patients with cirrhosis. Liver Int 2010;30(7):937-947 View Article PubMed/NCBI
  10. Arroyo V, Fernandez J. Pathophysiological basis of albumin use in cirrhosis. Ann Hepatol 2011;10(Suppl 1):S6-14 PubMed/NCBI
  11. Fernández-Llama P, Ageloff S, Fernández-Varo G, Ros J, Wang X, Garra N, et al. Sodium retention in cirrhotic rats is associated with increased renal abundance of sodium transporter proteins. Kidney Int 2005;67(2):622-630 View Article PubMed/NCBI
  12. Wood LJ, Massie D, McLean AJ, Dudley FJ. Renal sodium retention in cirrhosis: tubular site and relation to hepatic dysfunction. Hepatology 1988;8(4):831-836 View Article PubMed/NCBI
  13. Huang X, Dorhout Mees E, Vos P, Hamza S, Braam B. Everything we always wanted to know about furosemide but were afraid to ask. Am J Physiol Renal Physiol 2016;310(10):F958-F971 View Article PubMed/NCBI
  14. Casu G, Merella P. Diuretic Therapy in Heart Failure - Current Approaches. Eur Cardiol 2015;10(1):42-47 View Article PubMed/NCBI
  15. Felker GM, Ellison DH, Mullens W, Cox ZL, Testani JM. Diuretic Therapy for Patients With Heart Failure: JACC State-of-the-Art Review. J Am Coll Cardiol 2020;75(10):1178-1195 View Article PubMed/NCBI
  16. Gao Y, Liu X, Gao Y, Duan M, Hou B, Chen Y. Pharmacological Interventions for Cirrhotic Ascites: From Challenges to Emerging Therapeutic Horizons. Gut Liver 2024;18(6):934-948 View Article PubMed/NCBI
  17. Montalvo-Gordon I, Chi-Cervera LA, García-Tsao G. Sodium-Glucose Cotransporter 2 Inhibitors Ameliorate Ascites and Peripheral Edema in Patients With Cirrhosis and Diabetes. Hepatology 2020;72(5):1880-1882 View Article PubMed/NCBI
  18. Hiraizumi M, Akashi T, Murasaki K, Kishida H, Kumanomidou T, Torimoto N, et al. Transport and inhibition mechanism of the human SGLT2-MAP17 glucose transporter. Nat Struct Mol Biol 2024;31(1):159-169 View Article PubMed/NCBI
  19. Thomson SC, Vallon V. Renal Effects of Sodium-Glucose Co-Transporter Inhibitors. Am J Cardiol 2019;124(Suppl 1):S28-S35 View Article PubMed/NCBI
  20. Vergara A, Llorens-Cebrià C, Martos N, Martínez-Díaz I, Stein F, Domínguez-Báez P, et al. The membrane-associated protein 17 (MAP17) is up-regulated in response to empagliflozin on top of RAS blockade in experimental diabetic nephropathy. Clin Sci (Lond) 2023;137(1):87-104 View Article PubMed/NCBI
  21. Niu Y, Liu R, Guan C, Zhang Y, Chen Z, Hoerer S, et al. Structural basis of inhibition of the human SGLT2-MAP17 glucose transporter. Nature 2022;601(7892):280-284 View Article PubMed/NCBI
  22. Onishi A, Fu Y, Patel R, Darshi M, Crespo-Masip M, Huang W, et al. A role for tubular Na(+)/H(+) exchanger NHE3 in the natriuretic effect of the SGLT2 inhibitor empagliflozin. Am J Physiol Renal Physiol 2020;319(4):F712-F728 View Article PubMed/NCBI
  23. Borges-Júnior FA, Silva Dos Santos D, Benetti A, Polidoro JZ, Wisnivesky ACT, Crajoinas RO, et al. Empagliflozin Inhibits Proximal Tubule NHE3 Activity, Preserves GFR, and Restores Euvolemia in Nondiabetic Rats with Induced Heart Failure. J Am Soc Nephrol 2021;32(7):1616-1629 View Article PubMed/NCBI
  24. Biancalana E, Rossi C, Raggi F, Distaso M, Tricò D, Baldi S, et al. Empagliflozin and Renal Sodium-Hydrogen Exchange in Healthy Subjects. J Clin Endocrinol Metab 2023;108(8):e567-e573 View Article PubMed/NCBI
  25. Castro PC, Santos-Rios TM, Martins FL, Crajoinas RO, Caetano MV, Lessa LMA, et al. Renal upregulation of NCC counteracts empagliflozin-mediated NHE3 inhibition in normotensive but not in hypertensive male rat. Am J Physiol Cell Physiol 2024;326(6):C1573-C1589 View Article PubMed/NCBI
  26. Masuda T, Watanabe Y, Fukuda K, Watanabe M, Onishi A, Ohara K, et al. Unmasking a sustained negative effect of SGLT2 inhibition on body fluid volume in the rat. Am J Physiol Renal Physiol 2018;315(3):F653-F664 View Article PubMed/NCBI
  27. Rao VS, Ivey-Miranda JB, Cox ZL, Moreno-Villagomez J, Maulion C, Bellumkonda L, et al. Empagliflozin in Heart Failure: Regional Nephron Sodium Handling Effects. J Am Soc Nephrol 2024;35(2):189-201 View Article PubMed/NCBI
  28. Griffin M, Rao VS, Ivey-Miranda J, Fleming J, Mahoney D, Maulion C, et al. Empagliflozin in Heart Failure: Diuretic and Cardiorenal Effects. Circulation 2020;142(11):1028-1039 View Article PubMed/NCBI
  29. Boorsma EM, Beusekamp JC, Ter Maaten JM, Figarska SM, Danser AHJ, van Veldhuisen DJ, et al. Effects of empagliflozin on renal sodium and glucose handling in patients with acute heart failure. Eur J Heart Fail 2021;23(1):68-78 View Article PubMed/NCBI
  30. Gao Y, Wei L, Zhang DD, Chen Y, Hou B. SGLT2 Inhibitors: A New Dawn for Recurrent/Refractory Cirrhotic Ascites. J Clin Transl Hepatol 2021;9(6):795-797 View Article PubMed/NCBI
  31. Castrop H, Schnermann J. Isoforms of renal Na-K-2Cl cotransporter NKCC2: expression and functional significance. Am J Physiol Renal Physiol 2008;295(4):F859-F866 View Article PubMed/NCBI
  32. Burke M, Pabbidi MR, Farley J, Roman RJ. Molecular mechanisms of renal blood flow autoregulation. Curr Vasc Pharmacol 2014;12(6):845-858 View Article PubMed/NCBI
  33. Schnermann J, Ploth DW, Hermle M. Activation of tubulo-glomerular feedback by chloride transport. Pflugers Arch 1976;362(3):229-240 View Article PubMed/NCBI
  34. Bell PD, Thomas C, Williams RH, Navar LG. Filtration rate and stop-flow pressure feedback responses to nephron perfusion in the dog. Am J Physiol 1978;234(2):F154-F165 View Article PubMed/NCBI
  35. Scholtes RA, Muskiet MHA, van Baar MJB, Hesp AC, Greasley PJ, Hammarstedt A, et al. The Adaptive Renal Response for Volume Homeostasis During 2 Weeks of Dapagliflozin Treatment in People With Type 2 Diabetes and Preserved Renal Function on a Sodium-Controlled Diet. Kidney Int Rep 2022;7(5):1084-1092 View Article PubMed/NCBI
  36. Schork A, Saynisch J, Vosseler A, Jaghutriz BA, Heyne N, Peter A, et al. Effect of SGLT2 inhibitors on body composition, fluid status and renin-angiotensin-aldosterone system in type 2 diabetes: a prospective study using bioimpedance spectroscopy. Cardiovasc Diabetol 2019;18(1):46 View Article PubMed/NCBI
  37. Manosroi W, Danpanichkul P, Atthakomol P. Effect of sodium-glucose cotransporter-2 inhibitors on aldosterone and renin levels in diabetes mellitus type 2 patients: a systematic review and meta-analysis. Sci Rep 2022;12(1):19603 View Article PubMed/NCBI
  38. Packer M, Wilcox CS, Testani JM. Critical Analysis of the Effects of SGLT2 Inhibitors on Renal Tubular Sodium, Water and Chloride Homeostasis and Their Role in Influencing Heart Failure Outcomes. Circulation 2023;148(4):354-372 View Article PubMed/NCBI
  39. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N Engl J Med 2019;381(21):1995-2008 View Article PubMed/NCBI
  40. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med 2020;383(15):1413-1424 View Article PubMed/NCBI
  41. Bhm M, Anker S, Mahfoud F, Lauder L, Filippatos G, Ferreira JP, et al. Empagliflozin, irrespective of blood pressure, improves outcomes in heart failure with preserved ejection fraction: the EMPEROR-Preserved trial. Eur Heart J 2023;44(5):396-407 View Article PubMed/NCBI
  42. Kravtsova O, Bohovyk R, Levchenko V, Palygin O, Klemens CA, Rieg T, et al. SGLT2 inhibition effect on salt-induced hypertension, RAAS, and Na(+) transport in Dahl SS rats. Am J Physiol Renal Physiol 2022;322(6):F692-F707 View Article PubMed/NCBI
  43. Herat LY, Magno AL, Rudnicka C, Hricova J, Carnagarin R, Ward NC, et al. SGLT2 Inhibitor-Induced Sympathoinhibition: A Novel Mechanism for Cardiorenal Protection. JACC Basic Transl Sci 2020;5(2):169-179 View Article PubMed/NCBI
  44. Zannad F, Ferreira JP, Butler J, Filippatos G, Januzzi JL, Sumin M, et al. Effect of empagliflozin on circulating proteomics in heart failure: mechanistic insights into the EMPEROR programme. Eur Heart J 2022;43(48):4991-5002 View Article PubMed/NCBI
  45. Chung S, Kim S, Son M, Kim M, Koh ES, Shin SJ, et al. Empagliflozin Contributes to Polyuria via Regulation of Sodium Transporters and Water Channels in Diabetic Rat Kidneys. Front Physiol 2019;10:271 View Article PubMed/NCBI
  46. Mayne KJ, Staplin N, Keane DF, Wanner C, Brenner S, Cejka V, et al. Effects of Empagliflozin on Fluid Overload, Weight, and Blood Pressure in CKD. J Am Soc Nephrol 2024;35(2):202-215 View Article PubMed/NCBI
  47. Titze J, Shakibaei M, Schafflhuber M, Schulze-Tanzil G, Porst M, Schwind KH, et al. Glycosaminoglycan polymerization may enable osmotically inactive Na+ storage in the skin. Am J Physiol Heart Circ Physiol 2004;287(1):H203-H208 View Article PubMed/NCBI
  48. Farman N, Maubec E, Poeggeler B, Klatte JE, Jaisser F, Paus R. The mineralocorticoid receptor as a novel player in skin biology: beyond the renal horizon?. Exp Dermatol 2010;19(2):100-107 View Article PubMed/NCBI
  49. Hallow KM, Greasley PJ, Helmlinger G, Chu L, Heerspink HJ, Boulton DW. Evaluation of renal and cardiovascular protection mechanisms of SGLT2 inhibitors: model-based analysis of clinical data. Am J Physiol Renal Physiol 2018;315(5):F1295-F1306 View Article PubMed/NCBI
  50. Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review. Diabetologia 2018;61(10):2108-2117 View Article PubMed/NCBI
  51. Kalambokis GN, Tsiakas I, Filippas-Ntekuan S, Christaki M, Despotis G, Milionis H. Empagliflozin Eliminates Refractory Ascites and Hepatic Hydrothorax in a Patient With Primary Biliary Cirrhosis. Am J Gastroenterol 2021;116(3):618-619 View Article PubMed/NCBI
  52. Miyamoto Y, Honda A, Yokose S, Nagata M, Miyamoto J. Weaning from concentrated ascites reinfusion therapy for refractory ascites by SGLT2 inhibitor. Clin Kidney J 2022;15(4):831-833 View Article PubMed/NCBI
  53. Qin W, Gao Y, Zhao Y, Bian N, Fan W, Wang W, et al. Complete Resolution of Refractory Ascites and Pleural Effusion with Sustained Improvement in Urinary Sodium Excretion in a Cirrhotic Patient Treated with Empagliflozin. J Clin Transl Hepatol 2025;13(8):701-704 View Article PubMed/NCBI
  54. Nagayama K, Harada R, Ajima H, Orikasa S, Aoshima M, Oki Y. SGLT2 inhibitor administration to two patients with diabetes mellitus with ascites due to cirrhosis. Endocrinol Diabetes Metab Case Rep 2025;2025(3):e250015 View Article PubMed/NCBI
  55. Saffo S, Kaplan DE, Mahmud N, Serper M, John BV, Ross JS, et al. Impact of SGLT2 inhibitors in comparison with DPP4 inhibitors on ascites and death in veterans with cirrhosis on metformin. Diabetes Obes Metab 2021;23(10):2402-2408 View Article PubMed/NCBI
  56. Singh V, De A, Aggrawal R, Singh A, Charak S, Bhagat N. Safety and Efficacy of Dapagliflozin in Recurrent Ascites: A Pilot Study. Dig Dis Sci 2025;70(2):835-842 View Article PubMed/NCBI
  57. Shen I, Stojanova J, Yeo M, Olsen N, Lockart I, Wang M, et al. A potential novel treatment for cirrhosis-related ascites: Empagliflozin is safe and tolerable in advanced chronic liver disease. Br J Clin Pharmacol 2024;90(10):2529-2538 View Article PubMed/NCBI
  58. Kalambokis G, Tsiakas I, Filippas-Ntekouan S, Christaki M, Milionis H. Empagliflozin controls cirrhotic refractory ascites along with improvement of natriuresis and circulatory, cardiac, and renal function: A pilot study. Eur J Intern Med 2024;130:162-164 View Article PubMed/NCBI
  59. Bakosh MF, Ghazy RM, Ellakany WI, Kamal A. Empagliflozin as a novel therapy for cirrhotic refractory ascites: a randomized controlled study. Egypt Liver J 2024;14(1):76 View Article
  60. Abu-Hammour MN, Abdel-Razeq R, Vignarajah A, Khedraki R, Sims OT, Vigneswaramoorthy N, et al. Sodium-Glucose Cotransporter 2 Inhibitors and Serious Liver Events in Patients With Cirrhosis. JAMA Netw Open 2025;8(6):e2518470 View Article PubMed/NCBI
  61. Seif El-Din Z, Afify M, Zayed E, Elsabaawy D, Tharwa ES, Elsharawy A, et al. Dapagliflozin as an oral antihyperglycemic agent in the management of diabetes mellitus in patients with liver cirrhosis. World J Exp Med 2024;14(4):95272 View Article PubMed/NCBI
  62. Seidita A, Mandreucci F, Pistone M, Calderone S, Giuliano A, Chiavetta M, et al. Sodium-glucose cotransporter 2 inhibition in patients with liver cirrhosis and diabetes: a possible role in ascites control?. Italian J Med 2024;18:4 View Article
  63. Hu K, Goel A, Tarlow B, Cheng X, Kim S, Kim WR, et al. Empagliflozin in Diuretic-Refractory Ascites (DRAin-Em): Results of a Single-Center Feasibility Study. J Gen Intern Med 2025;40(7):1680-1682 View Article PubMed/NCBI
  64. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N Engl J Med 2021;385(16):1451-1461 View Article PubMed/NCBI
  65. Herrington WG, Staplin N, Wanner C, Green JB, Hauske SJ, Emberson JR, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med 2023;388(2):117-127 View Article PubMed/NCBI
  66. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 2015;373(22):2117-2128 View Article PubMed/NCBI
  67. DeFronzo RA, Hompesch M, Kasichayanula S, Liu X, Hong Y, Pfister M, et al. Characterization of renal glucose reabsorption in response to dapagliflozin in healthy subjects and subjects with type 2 diabetes. Diabetes Care 2013;36(10):3169-3176 View Article PubMed/NCBI
  68. Heerspink HJ, Perkins BA, Fitchett DH, Husain M, Cherney DZ. Sodium Glucose Cotransporter 2 Inhibitors in the Treatment of Diabetes Mellitus: Cardiovascular and Kidney Effects, Potential Mechanisms, and Clinical Applications. Circulation 2016;134(10):752-772 View Article
  69. Rieg T, Masuda T, Gerasimova M, Mayoux E, Platt K, Powell DR, et al. Increase in SGLT1-mediated transport explains renal glucose reabsorption during genetic and pharmacological SGLT2 inhibition in euglycemia. Am J Physiol Renal Physiol 2014;306(2):F188-F193 View Article PubMed/NCBI
  70. Kittipibul V, Cox ZL, Chesdachai S, Fiuzat M, Lindenfeld J, Mentz RJ. Genitourinary Tract Infections in Patients Taking SGLT2 Inhibitors: JACC Review Topic of the Week. J Am Coll Cardiol 2024;83(16):1568-1578 View Article PubMed/NCBI
  71. McGovern AP, Hogg M, Shields BM, Sattar NA, Holman RR, Pearson ER, et al. Risk factors for genital infections in people initiating SGLT2 inhibitors and their impact on discontinuation. BMJ Open Diabetes Res Care 2020;8(1):e001238 View Article PubMed/NCBI
  72. Scholtes RA, Muskiet MHA, van Baar MJB, Hesp AC, Greasley PJ, Karlsson C, et al. Natriuretic Effect of Two Weeks of Dapagliflozin Treatment in Patients With Type 2 Diabetes and Preserved Kidney Function During Standardized Sodium Intake: Results of the DAPASALT Trial. Diabetes Care 2021;44(2):440-447 View Article PubMed/NCBI
  73. Yeoh SE, Docherty KF, Jhund PS, Petrie MC, Inzucchi SE, Køber L, et al. Relationship of Dapagliflozin With Serum Sodium: Findings From the DAPA-HF Trial. JACC Heart Fail 2022;10(5):306-318 View Article PubMed/NCBI
  74. Baker WL, Buckley LF, Kelly MS, Bucheit JD, Parod ED, Brown R, et al. Effects of Sodium-Glucose Cotransporter 2 Inhibitors on 24-Hour Ambulatory Blood Pressure: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2017;6(5):e005686 View Article PubMed/NCBI
  75. Li S, Wang H, Ma W, Qiu L, Xia K, Zhang Y, et al. Monitoring blood pressure and cardiac function without positioning via a deep learning-assisted strain sensor array. Sci Adv 2023;9(32):eadh0615 View Article PubMed/NCBI
  76. Li Y, Li S, Song H, Shao B, Yang X, Deng N. Noninvasive blood pressure estimation with peak delay of different pulse waves. Int J Distribut Sensor Networks 2019;15(3):1550147719837877 View Article
  77. Noah AA, El-Mezayen NS, El-Ganainy SO, Darwish IE, Afify EA. Reversal of fibrosis and portal hypertension by Empagliflozin treatment of CCl(4)-induced liver fibrosis: Emphasis on gal-1/NRP-1/TGF-β and gal-1/NRP-1/VEGFR2 pathways. Eur J Pharmacol 2023;959:176066 View Article PubMed/NCBI
  78. Gao Y, Gao YY, Shi RY, Ji D, Wang Y, Xu L, et al. Effect of empagliflozin on fractional excretion of sodium in patients with cirrhosis and refractory ascites. World J Hepatol 2025;17(10):110247 View Article PubMed/NCBI
  79. Yan H, Wang W, Li Y, Qi Y, Lu R, Zhou Y, et al. Effect of henagliflozin on left ventricular mass index in dialysis patients with HFpEF (HELD-HF): protocol for a multicentre, randomised, double-blind, placebo-controlled trial. BMJ Open 2024;14(8):e087617 View Article PubMed/NCBI

About this Article

Cite this article
Gao Y, Gao Y, Ji D, Hu Z. Repurposing SGLT2 Inhibitors for Cirrhotic Ascites: From Mechanistic Research to Clinical Exploration. J Clin Transl Hepatol. Published online: Jan 22, 2026. doi: 10.14218/JCTH.2025.00465.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
September 7, 2025 December 1, 2025 December 15, 2025 January 22, 2026
DOI http://dx.doi.org/10.14218/JCTH.2025.00465
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Repurposing SGLT2 Inhibitors for Cirrhotic Ascites: From Mechanistic Research to Clinical Exploration

Yuan Gao, Yunyi Gao, Dong Ji, Zhongjie Hu
  • Reset Zoom
  • Download TIFF