Article Search
검색
검색 팝업 닫기

Metrics

Help

  • 1. Aims and Scope

    Gut and Liver is an international journal of gastroenterology, focusing on the gastrointestinal tract, liver, biliary tree, pancreas, motility, and neurogastroenterology. Gut atnd Liver delivers up-to-date, authoritative papers on both clinical and research-based topics in gastroenterology. The Journal publishes original articles, case reports, brief communications, letters to the editor and invited review articles in the field of gastroenterology. The Journal is operated by internationally renowned editorial boards and designed to provide a global opportunity to promote academic developments in the field of gastroenterology and hepatology. +MORE

  • 2. Editorial Board

    Editor-in-Chief + MORE

    Editor-in-Chief
    Yong Chan Lee Professor of Medicine
    Director, Gastrointestinal Research Laboratory
    Veterans Affairs Medical Center, Univ. California San Francisco
    San Francisco, USA

    Deputy Editor

    Deputy Editor
    Jong Pil Im Seoul National University College of Medicine, Seoul, Korea
    Robert S. Bresalier University of Texas M. D. Anderson Cancer Center, Houston, USA
    Steven H. Itzkowitz Mount Sinai Medical Center, NY, USA
  • 3. Editorial Office
  • 4. Articles
  • 5. Instructions for Authors
  • 6. File Download (PDF version)
  • 7. Ethical Standards
  • 8. Peer Review

    All papers submitted to Gut and Liver are reviewed by the editorial team before being sent out for an external peer review to rule out papers that have low priority, insufficient originality, scientific flaws, or the absence of a message of importance to the readers of the Journal. A decision about these papers will usually be made within two or three weeks.
    The remaining articles are usually sent to two reviewers. It would be very helpful if you could suggest a selection of reviewers and include their contact details. We may not always use the reviewers you recommend, but suggesting reviewers will make our reviewer database much richer; in the end, everyone will benefit. We reserve the right to return manuscripts in which no reviewers are suggested.

    The final responsibility for the decision to accept or reject lies with the editors. In many cases, papers may be rejected despite favorable reviews because of editorial policy or a lack of space. The editor retains the right to determine publication priorities, the style of the paper, and to request, if necessary, that the material submitted be shortened for publication.

Search

Search

Year

to

Article Type

Online first

Split Viewer

Online first

Pharmacological Interventions for Cirrhotic Ascites: From Challenges to Emerging Therapeutic Horizons

Yuan Gao1 , Xin Liu2 , Yunyi Gao3 , Meili Duan4 , Bing Hou5 , Yu Chen1

1Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China; 2Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing, China; 3School of Basic Medicine, Qingdao University, Qingdao, China; 4Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China; 5Xenorm MedInfo Center, Beijing, China

Correspondence to: Meili Duan
ORCID https://orcid.org/0000-0003-2865-7840
E-mail dmeili@bfh.com.cn

Bing Hou
ORCID https://orcid.org/0000-0002-2882-8147
E-mail hou_bing@hotmail.com

Yu Chen
ORCID https://orcid.org/0000-0003-1906-7486
E-mail chybeyond@163.com

Yuan Gao, Xin Liu, Yunyi Gao contributed equally to this work as first authors.

Received: January 23, 2024; Revised: May 1, 2024; Accepted: May 2, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Gut Liver.

Published online August 29, 2024

Copyright © Gut and Liver.

Ascites is the most common complication in patients with decompensated cirrhosis. This condition results in a severely impaired quality of life, excessive healthcare use, recurrent hospitalizations and significant morbidity and mortality. While loop diuretics and mineralocorticoid receptor antagonists are commonly employed for symptom relief, our understanding of their impact on survival remains limited. A comprehensive understanding of the underlying pathophysiological mechanism of ascites is crucial for its optimal management. The renin-angiotensin-aldosterone system (RAAS) is increasingly believed to play a pivotal role in the formation of cirrhotic ascites, as RAAS overactivation leads to a reduction in urine sodium excretion then a decrease in the ability of the kidneys to excrete water. In this review, the authors provide an overview of the pathogenesis of cirrhotic ascites, the challenges associated with current pharmacologic treatments, and the previous attempts to modulate the RAAS, followed by a description of some emerging targeted RAAS agents with the potential to be used to treat ascites.

Keywords: Renin-angiotensin system, Liver cirrhosis, Ascites, Finerenone, Sodium-glucose transporter 2 inhibitors

The appearance of ascites is the most common sign indicating the entry of decompensated phase of cirrhosis, about 5% to 10% of patients with compensated liver cirrhosis would develop this complication each year.1 Despite its prevalence in medical settings, its management remains a great challenge for physicians. The 1-year mortality rate of patients with cirrhotic ascites is close to 40%, and only half could survive over 2 years.2 Moreover, ascites significantly impacts the quality of patient’s life, results in a substantial disease burden, and requires significant health care resources. In Western countries, the 30-day unplanned readmission rate for ascites ranges from 37% to 52%, with costs exceeding 20,000 USD in the last year of life.3,4

Blood pressure is dictated by the dynamic interplay of cardiac output, systemic vascular resistance, and blood volume. The hallmark of hemodynamic disturbances in liver cirrhosis is a precarious balance to maintain the blood pressure on a setting of splanchnic arterial vasodilation. Compensated cirrhosis is characterized by a progressive decline in systemic vascular resistance, primarily due to splanchnic arterial dilatation, which is offset by an increase in cardiac output, without appreciable changes in blood volume. However, as cirrhosis progresses to the decompensated stage, cardiac output cannot increase further, as a result, a compromised cardiac performance occurs (Fig. 1).5 Concomitantly, splanchnic arterial vasodilation continues to advance, accompanied by related changes in the molecule profile.6 For instance, portal hypertension induces overproduction of nitric oxide (NO) by endothelial cells, which, regarded as the strongest vasodilator molecule, contributes to excessive vasodilation in both splanchnic arterial vessels and systemic circulation.7 In addition, several other vasodilatory molecules have also been identified as a contributory role in the pathogenesis of portal hypertension in cirrhotic patients and experimental rodent models.8,9 Carbon monoxide, which is sourced from heme oxygenase-1, may serve as an endogenous regulator of vascular tone and exhibit anti-inflammatory and anti-apoptotic properties.10 Similarly, prostacyclin is another potent vasodilator, as well as inhibitor of platelet thrombus formation.11 Endocannabinoids are lipid-mediated signaling molecules that increase activity of vasodilatory-related pathways and modulate diverse physiological functions, including regulating inflammation status.12 Their upregulation in the setting of cirrhosis and portal hypertension suggests a potential role in the modulation of splanchnic vascular resistance and the maintenance of portal blood flow.

Figure 1.Hemodynamic manifestations of liver cirrhosis. The main hemodynamic manifestation of liver cirrhosis is a progressive decrease in arterial vessel resistance due to splanchnic arterial vasodilatation. Initially, this is compensated by an increase in cardiac output (CO). However, compromised cardiac function in decompensated stage further deteriorates effective circulating blood volume insufficiency. Finally, the components in neurohumoral system have to be activated. RAAS, renin-angiotensin-aldosterone system; ADH, antidiuretic hormone. Adapted from Arroyo V, et al. Ann Hepatol 2011;10 Suppl 1:S6-S14.5

As discussed above, pathophysiological changes in patients with cirrhosis give rise to a decrease in the effective circulating blood volume and consequent circulatory dysfunction. To maintain blood pressure despite these hemodynamic abnormalities, the renin-angiotensin-aldosterone system (RAAS) is activated, leading to a significant increase in the reabsorption of sodium and water in the kidney.6,13 The aldosterone secreted during this process can stimulate the mineralocorticoid receptor, which acts as a transcription factor to elevate the expression of inflammatory cytokines and activate genes targeting water resorption.14 This process ultimately leads to disproportionate water retention and results in dilutional hyponatremia. Additionally, relative hypovolemia reduces the afferent impulses from baroreceptors in the atria, ventricles, carotid sinus, and aortic arch that communicate with the hypothalamus through the vagus nerve. This process triggers non-osmotic secretion of antidiuretic hormone (ADH), which, by binding to V2 receptors on the basolateral membrane of the collecting duct, increases cyclic AMP production and activates protein kinase A. These facilitate the translocation of aquaporin-2 from the cytoplasm to the apical plasma membrane of the lumen side, altering water permeability.15

The above pathophysiological mechanisms collectively contribute to the most common complication in patients with decompensated cirrhosis-ascites.

Current guidelines recommend several treatment strategies for patients with decompensated cirrhosis and ascites. Non-pharmacologic options include sodium restriction and large-volume paracentesis. Mineralocorticoid receptor antagonists (MRAs) and loop diuretics form the mainstay of pharmacologic treatments for alleviating ascites.1,16 The basic principle of using diuretics to manage ascites is to promote natriuresis by blocking ion channels in renal tubular epithelial cells. This action subsequently leads to osmotic diuresis, a process that reduces the passive reabsorption of water and ions. Despite these agents have been widely used for decades, they are considered as symptom relievers without proven evidence on survival benefits.1

Loop diuretics exhibit a potent natriuretic effect by interfering with the sodium-potassium-chloride cotransporter 2 (NKCC2) located at the thick segment of the ascending limb. By increasing the sodium chloride concentration in the luminal fluid of the distal convoluted tubule, these agents have been found to remarkably increase urine production. However, this pronounced natriuretic effect is limited to an acute setting. Inhibiting NKCC2 located at the macula densa can increase renin secretion, potentially exacerbating RAAS overactivation, leading to greater reabsorption of sodium. Prolonged use of loop diuretics leads to a gradual decline in net sodium excretion, which is termed “braking phenomenon.”17 Despite guidelines allow titrating the maximum furosemide dosage up to 160 mg/day, clinicians often consider the dose-effect relationship of loop diuretic to be linear. In fact, these agents demonstrate steep dose-response curves, with minimal effects observed until a threshold dose is reached. Beyond this threshold, response rapidly peaks, a phenomenon known as the “ceiling effect.” Consequently, once the ceiling is achieved, further increases in diuretic dosages no longer enhance urinary sodium excretion.17 Yet, in patients with cirrhotic ascites, the dose-response curve shifts downward and to the right, reducing the ceiling height, as observed in heart failure cases. These suggest that increasing loop diuretic dosage is futile for urine volume improvement.

As the most commonly used neurohormonal modulator, spironolactone was developed over 60 years ago as a class of potent MRAs. It was primarily used for the clinical treatment of congestive heart failure, primary aldosteronism, essential hypertension, and other pathologies associated with aldosteronism, such as ascites secondary to decompensated cirrhosis. Spironolactone is metabolized in the liver into three active metabolites that bind to cytoplasmic mineralocorticoid receptors, functioning as aldosterone antagonists, and produce a potassium-sparing diuretic effect in the distal tubules. All three active metabolites have demonstrated long half-life (range, 13.8 to 16.5 hours).18 However, liver cirrhosis can significantly impair the metabolic process, extending the metabolites’ half-lives from 23.9 hours to 126 hours.19 In clinical practice, this slows pharmacokinetic characteristics poses a challenge for physicians in titrating the optimal dose of spironolactone. Optimal dosage adjustment can take several weeks and is often accompanied by electrolyte disturbances related to the accumulation of active constituents during the treatment.20 Due to its simultaneous blockage of progesterone receptors and androgen receptors, spironolactone also commonly causes challenging side effects such as menstrual irregularities, impotence, and gynecomastia.

Unlike spironolactone, novel nonsteroidal MRAs showed significantly higher selectivity for aldosterone receptors and greater affinity in head-to-head trials.21 Its nonsteroidal structure allows for more efficient binding to aldosterone receptors, inhibiting inflammatory and pro-fibrotic gene expression due to hyperaldosteronemia, thus slowing the pace of kidney structure remodeling and functional deterioration among diabetic patients.22 The advantages of nonsteroidal MRAs are evidenced not only by their higher selectivity and affinity for mineral receptor but also by their inactive metabolites, rapid onset of action, and significantly shorter half-life, lasting only 2 hours.23 In addition, nonsteroidal MRAs have shown a better safety profile regarding hyperkalemia, with incidence less than half those associated with spironolactone (5.3% for finerenone vs 12.7% for spironolactone) in a phase II clinical trial.24

In the FIDELIO-DKD research, the nonsteroidal MRA finerenone considerably decreased the renal composite endpoint by 18% in patients with type 2 diabetes and chronic kidney diseases. The study also indicated a potential reduction in the incidence of hospitalization due to heart failure (hazard ratio, 0.86; 95% confidence interval, 0.68 to 1.08).25 Another phase III trial investigated the compositecardiovascular events (including death from cardiovascular causes, nonfatal stroke, nonfatal myocardial infarction, or hospitalization caused by heart failure) in patients withchronic kidney disease and type 2 diabetes. Finerenone decreased the incidence of heart failure hospitalization by 29%, while only 1.2% of the finerenone group discontinued treatment due to hyperkalemia, compared to 0.4% in the placebo group.26 Importantly, neither of these trials reported any hepatotoxicity-related adverse events. Currently, there is no direct evidence supporting the use of nonsteroidal MRAs in the management of cirrhotic ascites. However, given that RAAS overactivation is a common pathogenic mechanism in cirrhotic ascites, heart failure and diabetes mellitus, further investigation into nonsteroidal MRAs is warranted.

Vaptans, also known as vasopressin receptor antagonists, disrupt the interaction between ADH and vasopressin V2 receptors, thereby facilitating the excretion of free water. Mechanistically, these medications do not affect the RAAS activity as they merely mitigate water retention without concomitant excretion of urine sodium. This action can lead to sodium retention and subsequent recurrence of water retention.27 Moreover, a rapid elevation of serum sodium concentrations could further stimulate osmotic ADH production,28 and volume depletion would further compromise hemodynamics. Given these limitations, vaptans cannot provide long-term relief from cirrhotic ascites.

Several clinical studies have reported favorable outcomes with short-term (≤2 weeks) weight reduction, coupled with the correction of hyponatremia, in patients with cirrhotic ascites.29 However, a 12-week study showed satavaptan offered no significant advantages over placebo in terms of ascites regression or large-volume paracentesis requirements.30 Interestingly, similar findings have also been observed in the treatment of heart failure. Although tolvaptan alleviated dyspnea symptoms among hospitalized patients with acute congestive heart failure,31,32 it did not offer any long-term clinical benefits.33 Notably, vaptans were associated with hepatic toxicity as an adverse effect in patients without underlying liver diseases.34 Accordingly, current guidelines do not endorse the routine use of vaptans in treating cirrhotic ascites.1,35

Liver cirrhosis and chronic heart failure share similar pathophysiological mechanism (Fig. 2).36 Generally, the reduction in effective arterial blood volume, whether due to splanchnic vasodilation in decompensated liver cirrhosis or the loss of cardiac output in heart failure, can increase RAAS activity. In addition, non-osmotic ADH release is further increased by sympathetic excitation due to relative hypovolemia,37 which in turn enhances RAAS activity through renal β-adrenergic stimulation.38 These neurohumoral system changes trigger clinical symptoms linked to water and sodium retention, such as ascites in liver cirrhosis and acute pulmonary edema in heart failure, along with a commonly observed abnormality in laboratory assay, dilutional hyponatremia. Numerous RAAS-modulating agents, which have demonstrated efficacy in the management of heart failure, have been explored for their utility in the treatment of hepatic ascites. Regrettably, unlike the favorable outcomes with angiotensin-converting enzyme inhibitors (ACEIs) and nonselective β-blockers in heart failure patients, these agents have not shown clinical benefits among decompensated cirrhosis patients with ascites.39,40

Figure 2.The common mechanism between heart failure and liver cirrhosis. The activation of the RAAS takes the pivotal place in the pathophysiology of chronic heart failure and decompensated cirrhosis. NKCC2, sodium-potassium-chloride cotransporter 2; SGLT2, sodium-glucose cotransporter-2; RAAS, renin-angiotensin-aldosterone system; ACEI, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; ARNI, angiotensin receptor neprilysin inhibitor; MRAs, mineralocorticoid receptor antagonists; SGLT2i, SGLT2 inhibitor; TIPS, transjugular intrahepatic portosystemic shunt. Adapted from Saffo S, et al. Clin Liver Dis (Hoboken) 2018;11:141-144, with permission from Wolters Kluwer Health, Inc.44

1. Angiotensin-converting enzyme inhibitors

This class of agents disrupts RAAS activity, yet does not directly impact renin levels. ACEIs block the angiotensin-converting enzyme that transforms angiotensin I to angiotensin II, thereby reducing the production of angiotensin II. A double-blind randomized controlled trial suggested that captopril substantially reduced glomerular filtration rate and urine sodium excretion in individuals with cirrhosis.41 Further research demonstrated that in patients with cirrhotic ascites, captopril dramatically reduced natriuresis, urine production, and impeded the natriuretic effect of loop diuretics.42 Pathophysiologically, ACEIs dilate the efferent arterioles and drastically diminish the glomerular filtration rate, leading to reduced urine sodium and urine volume, ultimately exacerbating the water and sodium retention. Further investigations have revealed that, contrary to expectations, ACEIs could not suppress RAAS activity among cirrhotic patients but instead increased plasma renin activity.43 Since the vasodilatory impact by ACEIs on the small arteries would be a strong stimulus to RAAS through the feedback-activated pathways and overwhelm its direct influence on RAAS. To date, attempts to manage cirrhotic ascites with ACEIs have been unsuccessful.

2. Nonselective β-blockers

Nonselective β-blockers are established as a cornerstone of pharmacotherapy for patients with compensated cirrhosis, as demonstrated by multiple trials. These agents not only reduce portal pressure but also inhibit renin expression and secretion,45,46 effectively preventing decompensation occurrence among cirrhosis patients with clinically significant portal hypertension.47 A meta-analysis showed that long-term carvedilol therapy could reduce the risk of progression to decompensated stage and improve survival in patients with cirrhosis, primarily by reducing ascites.48 Despite their success in compensated cirrhosis, β-blockers have shown very limited effects on lowering portal pressure or suppressing RAAS once the disease progresses to decompensated stage.5 In particular, for patients with advanced liver cirrhosis and compromised cardiac output function, the negative inotropic effects of nonselective β-blockers significantly worsen the hemodynamic condition, contributing to elevated mortality rates in those awaiting a liver transplant.5

We conducted a systematic review to comprehensively evaluate the impact of drugs that act directly or indirectly on the RAAS in patients with decompensated cirrhosis, summarizing our finding in Table 1.6,40-44,49-92 The search process and inclusion-exclusion criteria for literature are detailed in the Supplementary Tables 1, 2 and Supplementary Fig. 1. Our review revealed that although these drugs inhibit the overactivated RAAS, most studies observed a decrease in urinary sodium or a reduction in urine volume, a characteristic inherent to ACEI/ARB or nonselective β-blockers. This suggests that successful modulating RAAS may be challenging without simultaneous improvements in urinary sodium and urine volume.

Table 1. The Summary of ACEI/ARB or Nonselective β-Blockers in the Treatment of Liver Cirrhosis

Author (year)Study typeNo. of participantsInterventionResult
Tergast et al. (2023)49Propensity scored matching study123 vs 41RAS-inhibitorsRAS-Inhibitor is associated with lower incidences of grade III AKI.
Danielsen et al. (2023)50Cross-over study39Propranolol infusionRenal artery blood flow fell by –5%.
Nabilou et al. (2022)51Prospective study38Propranolol infusionEffect of β-blockade on cardiac index is less potent in advanced cirrhosis.
Singh et al. (2022)52Prospective study160Propranolol vs endoscopic variceal ligationPPL is associated with lower survival, poor control of ascites, and increased risk of AKI when compared with EVL.
Chen et al. (20220)53Propensity scored matching study1,788 vs 1,788Propranolol user vs not usedPropranolol was associated with reduced mortality in patients with cirrhosis and ascites.
Tapper et al. (2022)54Population survey study63,364NSBBThe risk of ascites was higher for persons taking any NSBB.
Kang et al. (2021)55Retrospective study740NSBB user vs not usedNSBB therapy was associated with longer survival in prophylactic treatment of esophageal varices.
Sasso et al. (2021)56Retrospective study2,165NSBB user vs not usedUse of NSBB for patients with cirrhosis was associated with fewer infection-related admissions.
McDowell et al. (2021)57Retrospective study152Carvedilol vs variceal band ligationcarvedilol offers a longer survival benefit than patients receiving EVL.
Kalambokis et al. (2021)58Prospective study32 vs 64Continued use propranolol vs switch to carvedilolIn patients with cirrhosis and nonrefractory ascites, as carvedilol improves renal perfusion and clinical outcomes.
Téllez et al. (2020)59Clinical trial20 vs 18NSBBNSBB impair global circulatory homeostasis and renal function in cirrhotic patients with refractory ascites.
Alvarado-Tapias et al. (2020)60Comparative study403NSBBThe short-term effect of β-blockers on cardiac output may adversely influence survival in patients with decompensated cirrhosis.
Yoo et al. (2020)61Retrospective study271EVL vs propranolol +EVLEVL alone is a more appropriate treatment option for prophylaxis of esophageal varices than propranolol combination therapy in cirrhotic ascites patients.
Ngwa et al. (2020)62Retrospective study65 vs 105NSBB user vs not usedNSBB use was associated with lower 90-day mortality.
Giannelli et al. (2020)6Retrospective study584NSBBNSBB use in cirrhotic patients with compromised cardiac performance increase the mortality before liver transplant.
Tergast et al. (2019)63Retrospective study624NSBBTreatment with NSBB was associated with a higher 28-day transplant-free survival, but no benefit in mean arterial blood pressure <65 mm Hg group.
Chen et al. (2019)64Clinical trial60 vs 61GVO + carvedilol vs GVOcarvedilol + GVO did not decrease recurrence of EGVB, no impact on survival time, but produced more adverse events.
Giannitrapani et al. (2018)65Retrospective study230NSBB user vs not usedThe use of NSBB indicated a higher risk of PVT.
Zampino et al. (2018)66Retrospective study130NSBBNSBB treatment were independent risk factors of PVT.
Pfisterer et al. (2018)67Retrospective study766NSBBNSBB do not increase efficacy of band ligation in primary prophylaxis, but they improve survival in secondary prophylaxis of variceal bleeding.
Onali et al. (2017)68Retrospective study316NSBBPatients with ascites on NSBB did not have impaired survival compared to those not receiving NSSB.
Sinha et al. (2017)69Retrospective study325CarvedilolLow dose, chronic treatment with carvedilol in patients with liver cirrhosis and ascites is not detrimental.
Kim et al. (2017)70Retrospective study2,361NSBBNSBB in patients with ascites significantly increased the risk of AKI.
Bossen et al. (2016)71Retrospective study1,198NSBBUse of NSBBs in cirrhosis patients with ascites did not increase mortality.
Mookerjee et al. (2016)72Prospective observational study349NSBBNSBBs in cirrhotic patients is safe and reduces the mortality if they develop ACLF.
Mandorfer et al. (2014)73Retrospective study607NSBBNSBBs increase the risks for hepatorenal syndrome, acute kidney injury and reduce transplant-free survival.
Sersté et al. (2011)74Clinical trial10NSBBNSBB might be associated with a high risk of paracentesis-induced circulatory dysfunction.
Sersté et al. (2010)75Observational prospective study151NSBBThe use of NSBB was associated with poor survival in patients with refractory ascites.
Therapondos et al. (2006)76Before-and-after control study10LosartanLow dose losartan did not ameliorate erect posture-induced sodium retention in post-TIPS ascites-free patients.
Cholongitas et al. (2006)77Retrospective study134PropranololPropranolol was not associated with a lower risk for SBP.
Groszmann et al. (2005)78Clinical trial213NSBBNSBB was ineffective in preventing varices in unselected patients with cirrhosis.
Abecasis et al. (2003)79Clinical trial100Nadolol + placebo vs Nadolol + spironolactoneNadolol plus spironolactone effectively reduced the incidence of both portal-hypertensive complications.
Sen et al. (2002)80Clinical trial20spironolactone, alone or with propranololSpironolactone in combination with propranolol achieved adequate reduction in HVPG in propranolol-resistant portal hypertension.
Wong et al. (2002)81Clinical trial10Losartanbeneficial natriuretic effects of low-dose losartan on erect posture-induced sodium retention.
De et al. (2002)82Clinical trial36Carvedilol vs propranololCarvedilol reduced portal pressure in both acutely and over 7 days, but not superior to propranolol.
Lee et al. (2000)83Before-and-after control study25CarvedilolSingle dose of captopril decreased glomerular filtration rate and increased plasma renin activity.
Forrest et al. (1996)84Comparative study16CarvedilolCarvedilol did not compromise renal perfusion but increase the risk of hypotension in ascitic patients.
Tsai et al. (1996)43Clinical trial50CaptoprilCaptopril did not improve sodium and water retention in cirrhotic patients with ascites.
Amarapurkar et al. (1994)85Comparative study68EnalaprilEnalapril improved creatinine clearance in patients with liver cirrhosis.
Ohnishi et al. (1994)40Before-and-after control study10EnalaprilEnalapril slightly increased daily urinary volume and sodium excretion.
Gentilini et al. (1993)41Double-blind, cross-over clinical trialNACaptoprilLow dose of captopril decreased glomerular filtration rate and sodium excretion in ascitic patients.
van Vliet et al. (1992)86Before-and-after control study8CaptoprilAmong diuretic resistance patients, half of them increases urine sodium after low-dose captopril administered.
Ibarra et al. (1992)87Before-and-after control study9Captopril7 of 9 showed enhanced natriuresis after captopril administered.
Poynard et al. (1991)88Clinical trial589NSBBPropranolol and nadolol were effective in preventing first bleeding and reducing the mortality rate.
Group (1989)89Clinical trial174PropranololPropranolol prevented first gastrointestinal bleeding in cirrhotic patients.
Daskalopoulos et al. (1987)42Before-and-after control study11CaptoprilUrinary volume was reduced and natriuretic effect of furosemide was blunted.
Pariente et al. (1985)90Before-and-after control study6CaptoprilCaptopril decreased mean arterial pressure and glomerular filtration.
Rector et al. (1984)91Before-and-after control study13PropranololPropranolol induced an anti-natriuretic effect.
Lebrec et al. (1984)92Clinical trial74PropranololPropranolol increased the survival rate in cirrhotic patients.

ACEI, angiotensin-converting enzyme inhibitor; ARB, angiotensin II receptor blocker; RAS, renin-angiotensin system; AKI, acute kidney injury; PPL, propranolol; EVL, endoscopic variceal ligation; NSBB, nonselective beta-blocker; GVO, gastroesophageal variceal obliteration; EGVB, esophagogastric variceal bleeding; PVT, portal vein thrombosis; ACLF, acute-on-chronic liver failure; TIPS, transjugular intrahepatic portosystemic shunt; SBP, spontaneous bacterial peritonitis; HVPG, hepatic venous pressure gradient.



3. Albumin: RAAS modulator rather than plasma expansion

Albumin comprises about three-quarters of the oncotic pressure and is often classed as a plasma expander in patients with cirrhosis. As a plasma expander, albumin effectively mitigates relative hypovolemia,93 and corrects hyponatremia by increasing free water clearance in cirrhosis.94 Clinical trials have demonstrated that the combination of albumin and loop diuretics is more effective than loop diuretics alone in controlling ascites,95,96 and considerably improve survival in cirrhotic patients with ascites.97,98 Moreover, unlike other artificial colloids, albumin uniquely prevents circulatory deterioration following large-volume paracentesis.99 Current guidelines therefore recommend administering 8 g of albumin after each liter of ascites removal.1 Albumin infusion could restore blood pressure, increase blood volume and improve cardiac index in patients with cirrhosis.6,100,101 The circulatory benefits of albumin in cirrhosis patients cannot be solely attributed to its volume-expanding function. If the primary function of albumin were to expand volume, its initial administration as a plasma expander should lead to a decrease in plasma sodium concentrations (5% albumin solution and 25% solution contain 130 to 160 mEq/L sodium and are considered isotonic with plasma). Stated in another way, there should be a decline in plasma sodium concentration upon albumin infusion at initial stage. However, the results from the ATTIRE study, which evaluated albumin administration in hospitalized decompensated cirrhotic patients, challenged this perception. The study's appendix indicated that serum sodium levels started to increase within 24 hours among patients in albumin group, with a significant difference in serum sodium levels between the two study groups observed within 48 hours and this difference maintained throughout the trial.102 In the ANSWER study, long-term albumin infusion was proved to bring about prolonged overall survival with the concurrent improvement of hyponatremia in patients with decompensated cirrhosis.98 Intriguingly, if albumin was regarded solely as a plasma expander, it would be difficult to rationalize why transfusion of other blood products increases portal pressure,103 whereas the incidence of or gastro-esophageal variceal bleeding was not increased in albumin group in the ANSWER study.98 Another study highlighted albumin’s dose-dependent effect on reducing cytokine plasma concentrations and alleviating cardiocirculatory dysfunction without significant increase in portal pressure.104 The evidence supports the view that albumin's role far extends beyond that of a plasma expander, further exploration of its pleiotropic effects apart from its oncotic properties is needed.

Approximately one-third of the body’s total albumin resides in the vascular bed, with the remainder distributed within the extravascular and interstitial space. Albumin constantly exchanges between the intravascular and extravascular compartments, with approximately 4% to 5% of total albumin per hour traversing the capillary endothelium to the interstitial space before, then returning to the veins via lymphatics system.92 The plasma volume expansion effect of 5% albumin infusion only lasts only 4 to 6 hours, which is significantly weaker than that provided by other artificial colloids.105 This suggests that the principle of the therapeutic benefits derived from albumin should not primarily be attributed to its volume expansion effect. Decompensated cirrhosis is characterized by systemic pro-oxidant and pro-inflammatory environments. Furthermore, gut microbiota dysbiosis and translocation lead to the release of pro-inflammatory mediators, which can increase the release of vasodilatory molecules and exacerbate the overactivation of RAAS. In patients with decompensated cirrhosis, it is not only the quantity of albumin that is insufficient; the pleiotropic non-oncotic properties of albumin, including antioxidant capacity, free radical scavenging, immune regulation, and endothelial protection function are also impaired,93 resulting in a significant deficiency in “effective” albumin. In patients with decompensated cirrhosis, the levels of interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF) are significantly elevated. It has been confirmed in other diseases that IL-6 can stimulate the secretion of renin, thereby affecting the expression of angiotensin II and potentially impacting RAAS components.106 Moreover, IL-6 can influence VEGF production, which enhances angiogenesis and alters vascular structure, thus affecting the vascular response to angiotensin II and indirectly influencing RAAS activity.107 In the Pilot-PRECIOSA and INFECIR-2 study, large-volume albumin transfusions increased body albumin level and reduced the levels of IL-6 and VEGF, thereby regulating RAAS activity through both direct and indirect pathways.104,108 In addition, albumin is capable of indirectly modulating the RAAS system by restoring left ventricular function, compensating for relative hypovolemia in patients with liver cirrhosis,100,109 a property is not fully reliant upon changes in preload,110 however, other artificial colloids do not provide similar benefits on this aspect of circulatory function.111 Additionally, albumin corrects molecule profile abnormalities by reducing endogenous NO production, which in turn restores peripheral vessel resistance,112,113 improves vascular endothelial function, counteracts endotoxin-induced inflammation and oxidative stress,114 and diminishes systemic inflammation.113 These effects ultimately improve circulatory blood volume. Therefore, it can be inferred that albumin acts as a RAAS modulator in the treatment of cirrhotic ascites.

4. Sodium-glucose cotransporter type 2 inhibitors in cirrhotic ascites treatment

In the setting of RAAS overactivation in decompensated cirrhosis, sodium reabsorption markedly increases at the proximal tubular,115 the sodium concentration in tubule fluid flow to distal nephron therefore reduced. In this case, the diuretic effect of both conventional MRAs and loop diuretics would be blunted, given that they act at the ionic channel in the distal nephron. Additionally, proximal tubular sodium reabsorption is commonly involved in long-standing or refractory ascites.115,116 In order to intervene in the process of excessive sodium reabsorption in the proximal tubules, it is imperative to introduce novel approaches.

Sodium-glucose cotransporter-2 (SGLT2) inhibitors belong to a class of oral medications for treating type 2 diabetes.117 By inhibiting SGLT2, the agents significantly reduce the reabsorption of sodium and glucose in the proximal convoluted tubule. The increased sodium concentration in tubule fluid senses by the macula densa would in turn inhibit the renin release from the juxtaglomerular cells, then the increased sodium concentration in tubular fluid is a stimulus for tubuloglomerular feedback and causing vasoconstriction of the afferent arteriole.118 In spite of slightly reduced glomerular filtration, the osmotic diuresis caused by increased sodium concentration in the tubule fluid maintains the adequate amount of urine.119 The complementarity in mechanism suggests that SGLT2 inhibitors could exert a synergistic effect with loop diuretics and mitigate the braking phenomenon (Fig. 3).120 Their characteristics on promoting urinary excretion of solutes have been recently applied to treat advanced heart failure regardless of preexistence of diabetes.121,122 Given that advanced heart failure and decompensated liver cirrhosis share common pathophysiological features, it is reasonable to infer that SGLT2 inhibitors might be a candidate for treating cirrhotic ascites as well.123

Figure 3.The individual and synergistic effect of furosemide and empagliflozin on urinary sodium excretion and tubuloglomerular feedback. When furosemide inhibits NKCC2 in the macula densa, it reduces the ability to sense sodium and chloride levels, leading to an increased release of renin. This process suppresses the tubuloglomerular feedback. SGLT2 inhibitors increase the sodium concentration in the tubular fluid, which in turn raises the sodium and chloride levels passing through the macula densa. This increase in ion concentration is detected by the osmoreceptors in the macula densa, subsequently activating the tubuloglomerular feedback. Thus, these two drugs synergistically optimize fluid management in patients with cirrhotic ascites by balancing renin release and maintaining sodium levels. RAAS, renin-angiotensin-aldosterone system; NKCC2, sodium-potassiumchloride cotransporter 2; SGLT2, sodium-glucose cotransporter-2.

Encouragingly, several case reports have observed regression of ascites after adding SGLT2 inhibitors to the treatment for patients with decompensated cirrhosis and comorbid diabetes, without appreciable adverse events.124,125 Interestingly, the correction of hyponatremia observed in these cases might also suggest potential capacity on tempering RAAS with these drugs.125-127 An interesting parallel finding in heart failure treatment, in DAPA-HF trial, dapagliflozin increased urine volume and free water clearance persistently for several weeks, the correction of hyponatremia is along with the improvement of clinical outcomes.128 A mechanism-based explanation had been proposed from the editorial comment,129 SGLT2 inhibitors probably alleviate the water and sodium retention without further stimulate the RAAS activation as well as sympathetic nervous system. This class of agents therefore has potential to become a promising complement to the current pharmacologic treatments for ascitic patients. Moreover, the clinical benefits of SGLT2 inhibitors among patients with advanced heart failure do not rely on the existence of glycemia,121,122 it merits further investigation on ascitic patients regardless of comorbid diabetes.

Before repurposing SGLT2 inhibitors for cirrhotic ascites in a clinical setting, it is imperative to evaluate whether SGLT2 expression increases in the late-stage cirrhotic model and whether the proximal tubular sodium reabsorption increase is mediated by SGLT2. Additionally, it is essential to determine whether RAAS activity regulates the upregulation of SGLT2 expression in liver cirrhosis. It is probable that the renal pathological changes in progressive cirrhosis resemble those found in heart failure models.130

Hepatic glycogen capacity is impaired in patients with decompensated liver cirrhosis, predisposing them to hypoglycemia.131 However, the impact of SGLT2 inhibitors on blood glucose levels is not dependent on the level of insulin, the magnitude of blood glucose reduction is dependent on plasma glucose level among diabetic population,132 and less likely to induce a hypoglycemic event. Data from two phase III trials suggest SGLT2 inhibitors showed a comparable incidence with placebo in regard to liver dysfunction or clinical hypoglycemia regardless of diabetes existence.133 Moreover, hepatic dysfunction does not significantly affect the pharmacokinetics of SGLT2 inhibitors.134-136 An initial dip of estimated glomerular filtration rate and mild blood pressure drop (systolic and diastolic blood pressures decreased by 4 to 6 and 1 to 2 mm Hg respectively137 observed with SGLT2 inhibitors may raise worries about hepato-renal syndrome, but in practice, such change does not irritate RAAS components,134-136,138,139 it appears unlikely that SGLT2 inhibitors would cause hepato-renal syndrome in absence of an aggravation of RAAS status.

By far, the potential value of SGLT2 inhibitors in this field has begun to be gradually recognized,140 it is also encouraging to explore this new avenue of therapeutic approach (NCT05014594, NCT05013502, NCT05430243, NCT05726032).

In conclusion, despite decades of established medication therapy for cirrhotic ascites, significant therapeutic benefits remain elusive. However, growing understanding of the pathophysiological mechanisms underlying RAAS offers promise prospect for repurposing novel MRAs and SGLT2 inhibitors in ascites treatment. These efforts may pave the way toward more effective management of the complication of decompensated liver cirrhosis.

This work is supported by the Beijing Natural Science Foundation (No. 7232081), National Key Research and Development Program of China (No. 2022YFC2304400), the Beijing Hospitals Authority’s Ascent Plan (No. DFL20221501), and the Construction Project of High-level Technology Talents in Public Health (No. Discipline leader-01-12).

  1. European Association for the Study of the Liver. EASL clinical practice guidelines for the management of patients with decompensated cirrhosis. J Hepatol 2018;69:406-460.
    Pubmed CrossRef
  2. European Association for the Study of the Liver. EASL clinical practice guidelines on the management of ascites, spontaneous bacterial peritonitis, and hepatorenal syndrome in cirrhosis. J Hepatol 2010;53:397-417.
    Pubmed CrossRef
  3. Hudson B, Round J, Georgeson B, et al. Cirrhosis with ascites in the last year of life: a nationwide analysis of factors shaping costs, health-care use, and place of death in England. Lancet Gastroenterol Hepatol 2018;3:95-103.
    Pubmed CrossRef
  4. Volk ML, Tocco RS, Bazick J, Rakoski MO, Lok AS. Hospital readmissions among patients with decompensated cirrhosis. Am J Gastroenterol 2012;107:247-252.
    Pubmed KoreaMed CrossRef
  5. Arroyo V, Fernandez J. Pathophysiological basis of albumin use in cirrhosis. Ann Hepatol 2011;10 Suppl 1:S6-S14.
    CrossRef
  6. Giannelli V, Roux O, Laouénan C, et al. Impact of cardiac function, refractory ascites and beta blockers on the outcome of patients with cirrhosis listed for liver transplantation. J Hepatol 2020;72:463-471.
    Pubmed CrossRef
  7. Iwakiri Y. Pathophysiology of portal hypertension. Clin Liver Dis 2014;18:281-291.
    Pubmed KoreaMed CrossRef
  8. Iwakiri Y. The molecules: mechanisms of arterial vasodilatation observed in the splanchnic and systemic circulation in portal hypertension. J Clin Gastroenterol 2007;41 Suppl 3:S288-S294.
    Pubmed CrossRef
  9. Iwakiri Y, Groszmann RJ. Vascular endothelial dysfunction in cirrhosis. J Hepatol 2007;46:927-934.
    Pubmed CrossRef
  10. Chodorowski Z, Sein Anand J, Nowak-Banasik L, Szydłowska M, Klimek J, Kaletha K. Carbon monoxide: a regulator of vascular tone in hypoxia?. Przegl Lek 2005;62:438-440.
  11. Stitham J, Gleim SR, Douville K, Arehart E, Hwa J. Versatility and differential roles of cysteine residues in human prostacyclin receptor structure and function. J Biol Chem 2006;281:37227-37236.
    Pubmed CrossRef
  12. Pesce M, Esposito G, Sarnelli G. Endocannabinoids in the treatment of gasytrointestinal inflammation and symptoms. Curr Opin Pharmacol 2018;43:81-86.
    Pubmed CrossRef
  13. Alukal JJ, John S, Thuluvath PJ. Hyponatremia in cirrhosis: an update. Am J Gastroenterol 2020;115:1775-1785.
    Pubmed CrossRef
  14. Barrera-Chimal J, Girerd S, Jaisser F. Mineralocorticoid receptor antagonists and kidney diseases: pathophysiological basis. Kidney Int 2019;96:302-319.
    Pubmed CrossRef
  15. John S, Thuluvath PJ. Hyponatremia in cirrhosis: pathophysiology and management. World J Gastroenterol 2015;21:3197-3205.
    Pubmed KoreaMed CrossRef
  16. Xu X, Duan Z, et al; Chinese Society of Hepatology; Chinese Medical Association. Chinese guidelines on the management of ascites and its related complications in cirrhosis. Hepatol Int 2019;13:1-21.
    Pubmed CrossRef
  17. 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;75:1178-1195.
    Pubmed CrossRef
  18. Gardiner P, Schrode K, Quinlan D, et al. Spironolactone metabolism: steady-state serum levels of the sulfur-containing metabolites. J Clin Pharmacol 1989;29:342-347.
    Pubmed CrossRef
  19. Sungaila I, Bartle WR, Walker SE, et al. Spironolactone pharmacokinetics and pharmacodynamics in patients with cirrhotic ascites. Gastroenterology 1992;102:1680-1685.
    Pubmed CrossRef
  20. de Denus S, Leclair G, Dubé MP, et al. Spironolactone metabolite concentrations in decompensated heart failure: insights from the ATHENA-HF trial. Eur J Heart Fail 2020;22:1451-1461.
    Pubmed KoreaMed CrossRef
  21. Barrera-Chimal J, Kolkhof P, Lima-Posada I, Joachim A, Rossignol P, Jaisser F. Differentiation between emerging non-steroidal and established steroidal mineralocorticoid receptor antagonists: head-to-head comparisons of pharmacological and clinical characteristics. Expert Opin Investig Drugs 2021;30:1141-1157.
    Pubmed CrossRef
  22. Bakris GL, Agarwal R, Anker SD, et al. Design and baseline characteristics of the finerenone in reducing kidney failure and disease progression in diabetic kidney disease trial. Am J Nephrol 2019;50:333-344.
    Pubmed KoreaMed CrossRef
  23. Yang P, Huang T, Xu G. The novel mineralocorticoid receptor antagonist finerenone in diabetic kidney disease: progress and challenges. Metabolism 2016;65:1342-1349.
    Pubmed CrossRef
  24. Pitt B, Kober L, Ponikowski P, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J 2013;34:2453-2463.
    Pubmed KoreaMed CrossRef
  25. Bakris GL, Agarwal R, Anker SD, et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med 2020;383:2219-2229.
    Pubmed CrossRef
  26. Pitt B, Filippatos G, Agarwal R, et al. Cardiovascular events with finerenone in kidney disease and type 2 diabetes. N Engl J Med 2021;385:2252-2263.
    Pubmed CrossRef
  27. Zeidel ML. Salt and water: not so simple. J Clin Invest 2017;127:1625-1626.
    Pubmed KoreaMed CrossRef
  28. Bankir L, Guerrot D, Bichet DG. Vaptans or voluntary increased hydration to protect the kidney: how do they compare?. Nephrol Dial Transplant 2023;38:562-574.
    Pubmed CrossRef
  29. Yan L, Xie F, Lu J, et al. The treatment of vasopressin V2-receptor antagonists in cirrhosis patients with ascites: a meta-analysis of randomized controlled trials. BMC Gastroenterol 2015;15:65.
    Pubmed KoreaMed CrossRef
  30. Wong F, Watson H, Gerbes A, et al. Satavaptan for the management of ascites in cirrhosis: efficacy and safety across the spectrum of ascites severity. Gut 2012;61:108-116.
    Pubmed CrossRef
  31. Gheorghiade M, Gattis WA, O'Connor CM, et al. Effects of tolvaptan, a vasopressin antagonist, in patients hospitalized with worsening heart failure: a randomized controlled trial. JAMA 2004;291:1963-1971.
    Pubmed CrossRef
  32. Gheorghiade M, Konstam MA, Burnett JC, et al. Short-term clinical effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for heart failure: the EVEREST Clinical Status Trials. JAMA 2007;297:1332-1343.
    Pubmed CrossRef
  33. Konstam MA, Gheorghiade M, Burnett JC, et al. Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial. JAMA 2007;297:1319-1331.
    Pubmed CrossRef
  34. Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients with autosomal dominant polycystic kidney disease. N Engl J Med 2012;367:2407-2418.
    Pubmed KoreaMed CrossRef
  35. Aithal GP, Palaniyappan N, China L, et al. Guidelines on the management of ascites in cirrhosis. Gut 2021;70:9-29.
    Pubmed KoreaMed CrossRef
  36. 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;2:370-376.
    Pubmed CrossRef
  37. Bichet DG, Van Putten VJ, Schrier RW. Potential role of increased sympathetic activity in impaired sodium and water excretion in cirrhosis. N Engl J Med 1982;307:1552-1557.
    Pubmed CrossRef
  38. Berl T, Henrich WL, Erickson AL, Schrier RW. Prostaglandins in the beta-adrenergic and baroreceptor-mediated secretion of renin. Am J Physiol 1979;236:F472-F477.
    Pubmed CrossRef
  39. Wong RJ, Robinson A, Ginzberg D, Gomes C, Liu B, Bhuket T. Assessing the safety of beta-blocker therapy in cirrhosis patients with ascites: a meta-analysis. Liver Int 2019;39:1080-1088.
    Pubmed CrossRef
  40. Ohnishi A, Murakami S, Harada M, et al. Renal and hormonal responses to repeated treatment with enalapril in non-azotemic cirrhosis with ascites. J Hepatol 1994;20:223-230.
    Pubmed CrossRef
  41. Gentilini P, Romanelli RG, La Villa G, et al. Effects of low-dose captopril on renal hemodynamics and function in patients with cirrhosis of the liver. Gastroenterology 1993;104:588-594.
    Pubmed CrossRef
  42. Daskalopoulos G, Pinzani M, Murray N, Hirschberg R, Zipser RD. Effects of captopril on renal function in patients with cirrhosis and ascites. J Hepatol 1987;4:330-336.
    Pubmed CrossRef
  43. Tsai YT, Lin HC, Lee FY, Hou MC, Wang SS, Lee SD. Effects of captopril on renal functions, renal and portal hemodynamics in patients with cirrhosis. Proc Natl Sci Counc Repub China B 1996;20:44-50.
  44. Saffo S, Taddei T. SGLT2 inhibitors and cirrhosis: a unique perspective on the comanagement of diabetes mellitus and ascites. Clin Liver Dis (Hoboken) 2018;11:141-144.
    Pubmed KoreaMed CrossRef
  45. Fung JW, Yu CM, Yip G, et al. Effect of beta blockade (carvedilol or metoprolol) on activation of the renin-angiotensin-aldosterone system and natriuretic peptides in chronic heart failure. Am J Cardiol 2003;92:406-410.
    Pubmed CrossRef
  46. Tsuyuki RT, McAlister FA, Teo KK. Beta-blockers for congestive heart failure: what is the current consensus?. Drugs Aging 2000;16:1-7.
    Pubmed CrossRef
  47. Villanueva C, Albillos A, Genescà J, et al. β blockers to prevent decompensation of cirrhosis in patients with clinically significant portal hypertension (PREDESCI): a randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2019;393:1597-1608.
    Pubmed CrossRef
  48. Villanueva C, Torres F, Sarin SK, et al. Carvedilol reduces the risk of decompensation and mortality in patients with compensated cirrhosis in a competing-risk meta-analysis. J Hepatol 2022;77:1014-1025.
    Pubmed CrossRef
  49. Tergast TL, Griemsmann M, Wedemeyer H, Cornberg M, Maasoumy B. Effects of renin-angiotensin inhibitors on renal function and the clinical course in patients with decompensated cirrhosis. Sci Rep 2023;13:17486.
    Pubmed KoreaMed CrossRef
  50. Danielsen KV, Nabilou P, Wiese SS, Hove JD, Bendtsen F, Møller S. Effect of beta-blockers on multiple haemodynamics in cirrhosis: a cross-over study by MR-imaging and hepatic vein catheterization. Liver Int 2023;43:2245-2255.
    Pubmed CrossRef
  51. Nabilou P, Danielsen KV, Kimer N, Hove JD, Bendtsen F, Møller S. Blunted cardiovascular effects of beta-blockers in patients with cirrhosis: relation to severity?. PLoS One 2022;17:e0270603.
    Pubmed KoreaMed CrossRef
  52. Singh V, Kumar P, Verma N, Vijayvergiya R, Singh A, Bhalla A. Propranolol vs. band ligation for primary prophylaxis of variceal hemorrhage in cirrhotic patients with ascites: a randomized controlled trial. Hepatol Int 2022;16:944-953.
    Pubmed CrossRef
  53. Chen YC, Li YD, Lu CM, Huang WC, Kao SS, Chen WC. Propranolol use in patients with cirrhosis and refractory ascites: a nationwide study. Saudi J Gastroenterol 2022;28:108-114.
    Pubmed KoreaMed CrossRef
  54. Tapper EB, Zhao Z, Mazumder N, Parikh ND. Incidence of, risk factors for, and outcomes after ascites in a population-based cohort of older Americans. Dig Dis Sci 2022;67:5327-5335.
    Pubmed KoreaMed CrossRef
  55. Kang SH, Lee M, Kim MY, et al. The longitudinal outcomes of applying non-selective beta-blockers in portal hypertension: real-world multicenter study. Hepatol Int 2021;15:424-436.
    Pubmed CrossRef
  56. Sasso R, Rockey DC. Non-selective beta-blocker use in cirrhotic patients is associated with a reduced likelihood of hospitalisation for infection. Aliment Pharmacol Ther 2021;53:418-425.
    Pubmed CrossRef
  57. McDowell HR, Chuah CS, Tripathi D, Stanley AJ, Forrest EH, Hayes PC. Carvedilol is associated with improved survival in patients with cirrhosis: a long-term follow-up study. Aliment Pharmacol Ther 2021;53:531-539.
    Pubmed CrossRef
  58. Kalambokis GN, Christaki M, Tsiakas I, et al. Conversion of propranolol to carvedilol improves renal perfusion and outcome in patients with cirrhosis and ascites. J Clin Gastroenterol 2021;55:721-729.
    Pubmed CrossRef
  59. Téllez L, Ibáñez-Samaniego L, Pérez Del Villar C, et al. Non-selective beta-blockers impair global circulatory homeostasis and renal function in cirrhotic patients with refractory ascites. J Hepatol 2020;73:1404-1414.
    Pubmed CrossRef
  60. Alvarado-Tapias E, Ardevol A, Garcia-Guix M, et al. Short-term hemodynamic effects of beta-blockers influence survival of patients with decompensated cirrhosis. J Hepatol 2020;73:829-841.
    Pubmed CrossRef
  61. Yoo JJ, Kim SG, Kim YS, et al. Propranolol plus endoscopic ligation for variceal bleeding in patients with significant ascites: Propensity score matching analysis. Medicine (Baltimore) 2020;99:e18913.
    Pubmed KoreaMed CrossRef
  62. Ngwa T, Orman E, Gomez EV, et al. Non-selective beta blocker use is associated with improved short-term survival in patients with cirrhosis referred for liver transplantation. BMC Gastroenterol 2020;20:4.
    Pubmed KoreaMed CrossRef
  63. Tergast TL, Kimmann M, Laser H, et al. Systemic arterial blood pressure determines the therapeutic window of non-selective beta blockers in decompensated cirrhosis. Aliment Pharmacol Ther 2019;50:696-706.
    Pubmed CrossRef
  64. Chen WC, Hsin IF, Chen PH, et al. Addition of carvedilol to gastric variceal obturation does not decrease recurrence of gastric variceal bleeding in patients with cirrhosis. Clin Gastroenterol Hepatol 2019;17:2356-2363.
    Pubmed CrossRef
  65. Giannitrapani L, Granà W, Licata A, Schiavone C, Montalto G, Soresi M. Nontumorous portal vein thrombosis in liver cirrhosis: possible role of beta-blockers. Med Princ Pract 2018;27:466-471.
    Pubmed KoreaMed CrossRef
  66. Zampino R, Lebano R, Coppola N, et al. The use of nonselective beta blockers is a risk factor for portal vein thrombosis in cirrhotic patients. Saudi J Gastroenterol 2018;24:25-29.
    Pubmed KoreaMed CrossRef
  67. Pfisterer N, Dexheimer C, Fuchs EM, et al. Betablockers do not increase efficacy of band ligation in primary prophylaxis but they improve survival in secondary prophylaxis of variceal bleeding. Aliment Pharmacol Ther 2018;47:966-979.
    Pubmed CrossRef
  68. Onali S, Kalafateli M, Majumdar A, et al. Non-selective beta-blockers are not associated with increased mortality in cirrhotic patients with ascites. Liver Int 2017;37:1334-1344.
    Pubmed CrossRef
  69. Sinha R, Lockman KA, Mallawaarachchi N, Robertson M, Plevris JN, Hayes PC. Carvedilol use is associated with improved survival in patients with liver cirrhosis and ascites. J Hepatol 2017;67:40-46.
    Pubmed CrossRef
  70. Kim SG, Larson JJ, Lee JS, Therneau TM, Kim WR. Beneficial and harmful effects of nonselective beta blockade on acute kidney injury in liver transplant candidates. Liver Transpl 2017;23:733-740.
    Pubmed KoreaMed CrossRef
  71. Bossen L, Krag A, Vilstrup H, Watson H, Jepsen P. Nonselective beta-blockers do not affect mortality in cirrhosis patients with ascites: post hoc analysis of three randomized controlled trials with 1198 patients. Hepatology 2016;63:1968-1976.
    Pubmed CrossRef
  72. Mookerjee RP, Pavesi M, Thomsen KL, et al. Treatment with non-selective beta blockers is associated with reduced severity of systemic inflammation and improved survival of patients with acute-on-chronic liver failure. J Hepatol 2016;64:574-582.
    Pubmed CrossRef
  73. Mandorfer M, Bota S, Schwabl P, et al. Nonselective beta blockers increase risk for hepatorenal syndrome and death in patients with cirrhosis and spontaneous bacterial peritonitis. Gastroenterology 2014;146:1680-1690.
    Pubmed CrossRef
  74. Sersté T, Francoz C, Durand F, et al. Beta-blockers cause paracentesis-induced circulatory dysfunction in patients with cirrhosis and refractory ascites: a cross-over study. J Hepatol 2011;55:794-799.
    Pubmed CrossRef
  75. Sersté T, Melot C, Francoz C, et al. Deleterious effects of beta-blockers on survival in patients with cirrhosis and refractory ascites. Hepatology 2010;52:1017-1022.
    Pubmed CrossRef
  76. Therapondos G, Hol L, Benjaminov F, Wong F. The effect of single oral low- dose losartan on posture-related sodium handling in post-TIPS ascites-free cirrhosis. Hepatology 2006;44:640-649.
    Pubmed CrossRef
  77. Cholongitas E, Papatheodoridis GV, Manesis EK, Burroughs AK, Archimandritis AJ. Spontaneous bacterial peritonitis in cirrhotic patients: is prophylactic propranolol therapy beneficial?. J Gastroenterol Hepatol 2006;21:581-587.
    Pubmed CrossRef
  78. Groszmann RJ, Garcia-Tsao G, Bosch J, et al. Beta-blockers to prevent gastroesophageal varices in patients with cirrhosis. N Engl J Med 2005;353:2254-2261.
    Pubmed CrossRef
  79. Abecasis R, Kravetz D, Fassio E, et al. Nadolol plus spironolactone in the prophylaxis of first variceal bleed in nonascitic cirrhotic patients: a preliminary study. Hepatology 2003;37:359-365.
    Pubmed CrossRef
  80. Sen S, De BK, Biswas PK, Biswas J, Das D, Maity AK. Hemodynamic effect of spironolactone in liver cirrhosis and propranolol-resistant portal hypertension. Indian J Gastroenterol 2002;21:145-148.
  81. Wong F, Liu P, Blendis L. The mechanism of improved sodium homeostasis of low-dose losartan in preascitic cirrhosis. Hepatology 2002;35:1449-1458.
    Pubmed CrossRef
  82. De BK, Das D, Sen S, et al. Acute and 7-day portal pressure response to carvedilol and propranolol in cirrhotics. J Gastroenterol Hepatol 2002;17:183-189.
    Pubmed CrossRef
  83. Lee JK, Hsieh JF, Tsai SC, Ho YJ, Kao CH. Effects of single dose of 50mg captopril in patients with liver cirrhosis and ascites. Hepatogastroenterology 2000;47:767-770.
    Pubmed
  84. Forrest EH, Bouchier IA, Hayes PC. Acute haemodynamic changes after oral carvedilol, a vasodilating beta-blocker, in patients with cirrhosis. J Hepatol 1996;25:909-915.
    Pubmed CrossRef
  85. Amarapurkar DN, Dhawan P, Kalro RH. Role of routine estimation of creatinine clearance in patients with liver cirrhosis. Indian J Gastroenterol 1994;13:79-82.
  86. van Vliet AA, Hackeng WH, Donker AJ, Meuwissen SG. Efficacy of low-dose captopril in addition to furosemide and spironolactone in patients with decompensated liver disease during blunted diuresis. J Hepatol 1992;15:40-47.
    Pubmed CrossRef
  87. Ibarra FR, Afione C, Garzon D, Barontini M, Santos JC, Arrizurieta E. Portal pressure, renal function and hormonal profile after acute and chronic captopril treatment in cirrhosis. Eur J Clin Pharmacol 1992;43:477-482.
    Pubmed CrossRef
  88. Poynard T, Calès P, Pasta L, et al. Beta-adrenergic-antagonist drugs in the prevention of gastrointestinal bleeding in patients with cirrhosis and esophageal varices. An analysis of data and prognostic factors in 589 patients from four randomized clinical trials. Franco-Italian Multicenter Study Group. N Engl J Med 1991;324:1532-1538.
    Pubmed CrossRef
  89. The Italian Multicenter Project for Propranolol in Prevention of Bleeding. Propranolol prevents first gastrointestinal bleeding in non-ascitic cirrhotic patients. Final report of a multicenter randomized trial. J Hepatol 1989;9:75-83.
    Pubmed CrossRef
  90. Pariente EA, Bataille C, Bercoff E, Lebrec D. Acute effects of captopril on systemic and renal hemodynamics and on renal function in cirrhotic patients with ascites. Gastroenterology 1985;88(5 Pt 1):1255-1259.
    Pubmed CrossRef
  91. Rector WG Jr, Reynolds TB. Propranolol in the treatment of cirrhotic ascites. Arch Intern Med 1984;144:1761-1763.
    Pubmed CrossRef
  92. Lebrec D, Poynard T, Bernuau J, et al. A randomized controlled study of propranolol for prevention of recurrent gastrointestinal bleeding in patients with cirrhosis: a final report. Hepatology 1984;4:355-358.
    Pubmed CrossRef
  93. Bernardi M, Angeli P, Claria J, et al. Albumin in decompensated cirrhosis: new concepts and perspectives. Gut 2020;69:1127-1138.
    Pubmed KoreaMed CrossRef
  94. McCormick PA, Mistry P, Kaye G, Burroughs AK, McIntyre N. Intravenous albumin infusion is an effective therapy for hyponatraemia in cirrhotic patients with ascites. Gut 1990;31:204-207.
    Pubmed KoreaMed CrossRef
  95. Gentilini P, Casini-Raggi V, Di Fiore G, et al. Albumin improves the response to diuretics in patients with cirrhosis and ascites: results of a randomized, controlled trial. J Hepatol 1999;30:639-645.
    Pubmed CrossRef
  96. Laffi G, Gentilini P, Romanelli RG, La Villa G. Is the use of albumin of value in the treatment of ascites in cirrhosis? The case in favour. Dig Liver Dis 2003;35:660-663.
    Pubmed CrossRef
  97. Romanelli RG, La Villa G, Barletta G, et al. Long-term albumin infusion improves survival in patients with cirrhosis and ascites: an unblinded randomized trial. World J Gastroenterol 2006;12:1403-1407.
    Pubmed KoreaMed CrossRef
  98. Caraceni P, Riggio O, Angeli P, et al. Long-term albumin administration in decompensated cirrhosis (ANSWER): an open-label randomised trial. Lancet 2018;391:2417-2429.
    Pubmed CrossRef
  99. Bernardi M, Caraceni P, Navickis RJ, Wilkes MM. Albumin infusion in patients undergoing large-volume paracentesis: a meta-analysis of randomized trials. Hepatology 2012;55:1172-1181.
    Pubmed CrossRef
  100. Shasthry SM, Kumar M, Khumuckham JS, Sarin SK. Changes in cardiac output and incidence of volume overload in cirrhotics receiving 20% albumin infusion. Liver Int 2017;37:1167-1176.
    Pubmed CrossRef
  101. Luca A, García-Pagán JC, Bosch J, et al. Beneficial effects of intravenous albumin infusion on the hemodynamic and humoral changes after total paracentesis. Hepatology 1995;22:753-758.
    Pubmed CrossRef
  102. China L, Freemantle N, Forrest E, et al. A randomized trial of albumin infusions in hospitalized patients with cirrhosis. N Engl J Med 2021;384:808-817.
    Pubmed CrossRef
  103. Elizalde JI, Moitinho E, García-Pagán JC, et al. Effects of increasing blood hemoglobin levels on systemic hemodynamics of acutely anemic cirrhotic patients. J Hepatol 1998;29:789-795.
    Pubmed CrossRef
  104. Fernández J, Clària J, Amorós A, et al. Effects of albumin treatment on systemic and portal hemodynamics and systemic inflammation in patients with decompensated cirrhosis. Gastroenterology 2019;157:149-162.
    Pubmed CrossRef
  105. Quon CY. Clinical pharmacokinetics and pharmacodynamics of colloidal plasma volume expanders. Journal of Cardiothoracic Anesthesia 1988;2:13-23.
    CrossRef
  106. Schieffer B, Schieffer E, Hilfiker-Kleiner D, et al. Expression of angiotensin II and interleukin 6 in human coronary atherosclerotic plaques: potential implications for inflammation and plaque instability. Circulation 2000;101:1372-1378.
    Pubmed CrossRef
  107. Catar R, Witowski J, Zhu N, et al. IL-6 trans-signaling links inflammation with angiogenesis in the peritoneal membrane. J Am Soc Nephrol 2017;28:1188-1199.
    Pubmed KoreaMed CrossRef
  108. Uriz J, Ginès P, Cárdenas A, et al. Terlipressin plus albumin infusion: an effective and safe therapy of hepatorenal syndrome. J Hepatol 2000;33:43-48.
    Pubmed CrossRef
  109. Fernández J, Navasa M, Garcia-Pagan JC, et al. Effect of intravenous albumin on systemic and hepatic hemodynamics and vasoactive neurohormonal systems in patients with cirrhosis and spontaneous bacterial peritonitis. J Hepatol 2004;41:384-390.
    Pubmed CrossRef
  110. Bortoluzzi A, Ceolotto G, Gola E, et al. Positive cardiac inotropic effect of albumin infusion in rodents with cirrhosis and ascites: molecular mechanisms. Hepatology 2013;57:266-276.
    Pubmed CrossRef
  111. Fernández J, Monteagudo J, Bargallo X, et al. A randomized unblinded pilot study comparing albumin versus hydroxyethyl starch in spontaneous bacterial peritonitis. Hepatology 2005;42:627-634.
    Pubmed CrossRef
  112. Walley KR, McDonald TE, Wang Y, Dai S, Russell JA. Albumin resuscitation increases cardiomyocyte contractility and decreases nitric oxide synthase II expression in rat endotoxemia. Crit Care Med 2003;31:187-194.
    Pubmed CrossRef
  113. Arroyo V, García-Martinez R, Salvatella X. Human serum albumin, systemic inflammation, and cirrhosis. J Hepatol 2014;61:396-407.
    Pubmed CrossRef
  114. Kremer H, Baron-Menguy C, Tesse A, et al. Human serum albumin improves endothelial dysfunction and survival during experimental endotoxemia: concentration-dependent properties. Crit Care Med 2011;39:1414-1422.
    Pubmed CrossRef
  115. Salerno F, Guevara M, Bernardi M, et al. Refractory ascites: pathogenesis, definition and therapy of a severe complication in patients with cirrhosis. Liver Int 2010;30:937-947.
    Pubmed CrossRef
  116. Gatta A, Angeli P, Caregaro L, Menon F, Sacerdoti D, Merkel C. A pathophysiological interpretation of unresponsiveness to spironolactone in a stepped-care approach to the diuretic treatment of ascites in nonazotemic cirrhotic patients. Hepatology 1991;14:231-236.
    Pubmed CrossRef
  117. Washburn WN, Poucher SM. Differentiating sodium-glucose co-transporter-2 inhibitors in development for the treatment of type 2 diabetes mellitus. Expert Opin Investig Drugs 2013;22:463-486.
    Pubmed CrossRef
  118. Braunwald E. Gliflozins in the management of cardiovascular disease. N Engl J Med 2022;386:2024-2034.
    Pubmed CrossRef
  119. Tanaka H, Takano K, Iijima H, et al. Factors affecting canagliflozin-induced transient urine volume increase in patients with type 2 diabetes mellitus. Adv Ther 2017;34:436-451.
    Pubmed KoreaMed CrossRef
  120. Verma A, Patel AB, Waikar SS. SGLT2 inhibitor: not a traditional diuretic for heart failure. Cell Metab 2020;32:13-14.
    Pubmed CrossRef
  121. Packer M, Anker SD, Butler J, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med 2020;383:1413-1424.
    Pubmed CrossRef
  122. McMurray JJ, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 2019;381:1995-2008.
    Pubmed CrossRef
  123. 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;9:795-797.
    Pubmed KoreaMed CrossRef
  124. Mordi NA, Mordi IR, Singh JS, McCrimmon RJ, Struthers AD, Lang CC. Renal and cardiovascular effects of SGLT2 inhibition in combination with loop diuretics in patients with type 2 diabetes and chronic heart failure: the RECEDE-CHF trial. Circulation 2020;142:1713-1724.
    Pubmed KoreaMed CrossRef
  125. 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;116:618-619.
    Pubmed CrossRef
  126. 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;15:831-833.
    Pubmed KoreaMed CrossRef
  127. 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;72:1880-1882.
    Pubmed CrossRef
  128. Yeoh SE, Docherty KF, Jhund PS, et al. Relationship of dapagliflozin with serum sodium: findings from the DAPA-HF trial. JACC Heart Fail 2022;10:306-318.
    Pubmed CrossRef
  129. Li S, Levy WC. Impact of SGLT2 inhibitors on serum sodium in heart failure with reduced ejection fraction. JACC Heart Fail 2022;10:319-320.
    Pubmed CrossRef
  130. Katsurada K, Nandi SS, Sharma NM, Patel KP. Enhanced expression and function of renal SGLT2 (sodium-glucose cotransporter 2) in heart failure: role of renal nerves. Circ Heart Fail 2021;14:e008365.
    Pubmed KoreaMed CrossRef
  131. Pfortmueller CA, Wiemann C, Funk GC, et al. Hypoglycemia is associated with increased mortality in patients with acute decompensated liver cirrhosis. J Crit Care 2014;29:316.
    Pubmed CrossRef
  132. Polidori D, Sha S, Ghosh A, Plum-Mörschel L, Heise T, Rothenberg P. Validation of a novel method for determining the renal threshold for glucose excretion in untreated and canagliflozin-treated subjects with type 2 diabetes mellitus. J Clin Endocrinol Metab 2013;98:E867-E871.
    Pubmed KoreaMed CrossRef
  133. Zannad F, Ferreira JP, Pocock SJ, et al. SGLT2 inhibitors in patients with heart failure with reduced ejection fraction: a meta-analysis of the EMPEROR-Reduced and DAPA-HF trials. Lancet 2020;396:819-829.
    Pubmed CrossRef
  134. Kasichayanula S, Liu X, Zhang W, Pfister M, LaCreta FP, Boulton DW. Influence of hepatic impairment on the pharmacokinetics and safety profile of dapagliflozin: an open-label, parallel-group, single-dose study. Clin Ther 2011;33:1798-1808.
    Pubmed CrossRef
  135. Macha S, Rose P, Mattheus M, et al. Pharmacokinetics, safety and tolerability of empagliflozin, a sodium glucose cotransporter 2 inhibitor, in patients with hepatic impairment. Diabetes Obes Metab 2014;16:118-123.
    Pubmed CrossRef
  136. Devineni D, Curtin CR, Marbury TC, et al. Effect of hepatic or renal impairment on the pharmacokinetics of canagliflozin, a sodium glucose co-transporter 2 inhibitor. Clin Ther 2015;37:610-628.
    Pubmed CrossRef
  137. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects of sodium-glucose cotransporter 2 inhibitors: JACC state-of-the-art review. J Am Coll Cardiol 2020;75:422-434.
    Pubmed CrossRef
  138. Ansary TM, Nakano D, Nishiyama A. Diuretic effects of sodium glucose cotransporter 2 inhibitors and their influence on the renin-angiotensin system. Int J Mol Sci 2019;20:629.
    Pubmed KoreaMed CrossRef
  139. Griffin M, Rao VS, Ivey-Miranda J, et al. Empagliflozin in heart failure: diuretic and cardiorenal effects. Circulation 2020;142:1028-1039.
    Pubmed KoreaMed CrossRef
  140. Miyamoto Y, Honda A, Yokose S, Nagata M, Miyamoto J. The effects of SGLT2 inhibitors on liver cirrhosis patients with refractory ascites: a literature review. J Clin Med 2023;12:2253.
    Pubmed KoreaMed CrossRef

Article

ahead

Gut and Liver

Published online August 29, 2024

Copyright © Gut and Liver.

Pharmacological Interventions for Cirrhotic Ascites: From Challenges to Emerging Therapeutic Horizons

Yuan Gao1 , Xin Liu2 , Yunyi Gao3 , Meili Duan4 , Bing Hou5 , Yu Chen1

1Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China; 2Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing, China; 3School of Basic Medicine, Qingdao University, Qingdao, China; 4Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China; 5Xenorm MedInfo Center, Beijing, China

Correspondence to:Meili Duan
ORCID https://orcid.org/0000-0003-2865-7840
E-mail dmeili@bfh.com.cn

Bing Hou
ORCID https://orcid.org/0000-0002-2882-8147
E-mail hou_bing@hotmail.com

Yu Chen
ORCID https://orcid.org/0000-0003-1906-7486
E-mail chybeyond@163.com

Yuan Gao, Xin Liu, Yunyi Gao contributed equally to this work as first authors.

Received: January 23, 2024; Revised: May 1, 2024; Accepted: May 2, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Ascites is the most common complication in patients with decompensated cirrhosis. This condition results in a severely impaired quality of life, excessive healthcare use, recurrent hospitalizations and significant morbidity and mortality. While loop diuretics and mineralocorticoid receptor antagonists are commonly employed for symptom relief, our understanding of their impact on survival remains limited. A comprehensive understanding of the underlying pathophysiological mechanism of ascites is crucial for its optimal management. The renin-angiotensin-aldosterone system (RAAS) is increasingly believed to play a pivotal role in the formation of cirrhotic ascites, as RAAS overactivation leads to a reduction in urine sodium excretion then a decrease in the ability of the kidneys to excrete water. In this review, the authors provide an overview of the pathogenesis of cirrhotic ascites, the challenges associated with current pharmacologic treatments, and the previous attempts to modulate the RAAS, followed by a description of some emerging targeted RAAS agents with the potential to be used to treat ascites.

Keywords: Renin-angiotensin system, Liver cirrhosis, Ascites, Finerenone, Sodium-glucose transporter 2 inhibitors

INTRODUCTION

The appearance of ascites is the most common sign indicating the entry of decompensated phase of cirrhosis, about 5% to 10% of patients with compensated liver cirrhosis would develop this complication each year.1 Despite its prevalence in medical settings, its management remains a great challenge for physicians. The 1-year mortality rate of patients with cirrhotic ascites is close to 40%, and only half could survive over 2 years.2 Moreover, ascites significantly impacts the quality of patient’s life, results in a substantial disease burden, and requires significant health care resources. In Western countries, the 30-day unplanned readmission rate for ascites ranges from 37% to 52%, with costs exceeding 20,000 USD in the last year of life.3,4

PATHOGENESIS

Blood pressure is dictated by the dynamic interplay of cardiac output, systemic vascular resistance, and blood volume. The hallmark of hemodynamic disturbances in liver cirrhosis is a precarious balance to maintain the blood pressure on a setting of splanchnic arterial vasodilation. Compensated cirrhosis is characterized by a progressive decline in systemic vascular resistance, primarily due to splanchnic arterial dilatation, which is offset by an increase in cardiac output, without appreciable changes in blood volume. However, as cirrhosis progresses to the decompensated stage, cardiac output cannot increase further, as a result, a compromised cardiac performance occurs (Fig. 1).5 Concomitantly, splanchnic arterial vasodilation continues to advance, accompanied by related changes in the molecule profile.6 For instance, portal hypertension induces overproduction of nitric oxide (NO) by endothelial cells, which, regarded as the strongest vasodilator molecule, contributes to excessive vasodilation in both splanchnic arterial vessels and systemic circulation.7 In addition, several other vasodilatory molecules have also been identified as a contributory role in the pathogenesis of portal hypertension in cirrhotic patients and experimental rodent models.8,9 Carbon monoxide, which is sourced from heme oxygenase-1, may serve as an endogenous regulator of vascular tone and exhibit anti-inflammatory and anti-apoptotic properties.10 Similarly, prostacyclin is another potent vasodilator, as well as inhibitor of platelet thrombus formation.11 Endocannabinoids are lipid-mediated signaling molecules that increase activity of vasodilatory-related pathways and modulate diverse physiological functions, including regulating inflammation status.12 Their upregulation in the setting of cirrhosis and portal hypertension suggests a potential role in the modulation of splanchnic vascular resistance and the maintenance of portal blood flow.

Figure 1. Hemodynamic manifestations of liver cirrhosis. The main hemodynamic manifestation of liver cirrhosis is a progressive decrease in arterial vessel resistance due to splanchnic arterial vasodilatation. Initially, this is compensated by an increase in cardiac output (CO). However, compromised cardiac function in decompensated stage further deteriorates effective circulating blood volume insufficiency. Finally, the components in neurohumoral system have to be activated. RAAS, renin-angiotensin-aldosterone system; ADH, antidiuretic hormone. Adapted from Arroyo V, et al. Ann Hepatol 2011;10 Suppl 1:S6-S14.5

As discussed above, pathophysiological changes in patients with cirrhosis give rise to a decrease in the effective circulating blood volume and consequent circulatory dysfunction. To maintain blood pressure despite these hemodynamic abnormalities, the renin-angiotensin-aldosterone system (RAAS) is activated, leading to a significant increase in the reabsorption of sodium and water in the kidney.6,13 The aldosterone secreted during this process can stimulate the mineralocorticoid receptor, which acts as a transcription factor to elevate the expression of inflammatory cytokines and activate genes targeting water resorption.14 This process ultimately leads to disproportionate water retention and results in dilutional hyponatremia. Additionally, relative hypovolemia reduces the afferent impulses from baroreceptors in the atria, ventricles, carotid sinus, and aortic arch that communicate with the hypothalamus through the vagus nerve. This process triggers non-osmotic secretion of antidiuretic hormone (ADH), which, by binding to V2 receptors on the basolateral membrane of the collecting duct, increases cyclic AMP production and activates protein kinase A. These facilitate the translocation of aquaporin-2 from the cytoplasm to the apical plasma membrane of the lumen side, altering water permeability.15

The above pathophysiological mechanisms collectively contribute to the most common complication in patients with decompensated cirrhosis-ascites.

THE DILEMMA IN THE CURRENT CHOICE FOR ASCITES MANAGEMENT

Current guidelines recommend several treatment strategies for patients with decompensated cirrhosis and ascites. Non-pharmacologic options include sodium restriction and large-volume paracentesis. Mineralocorticoid receptor antagonists (MRAs) and loop diuretics form the mainstay of pharmacologic treatments for alleviating ascites.1,16 The basic principle of using diuretics to manage ascites is to promote natriuresis by blocking ion channels in renal tubular epithelial cells. This action subsequently leads to osmotic diuresis, a process that reduces the passive reabsorption of water and ions. Despite these agents have been widely used for decades, they are considered as symptom relievers without proven evidence on survival benefits.1

LOOP DIURETICS

Loop diuretics exhibit a potent natriuretic effect by interfering with the sodium-potassium-chloride cotransporter 2 (NKCC2) located at the thick segment of the ascending limb. By increasing the sodium chloride concentration in the luminal fluid of the distal convoluted tubule, these agents have been found to remarkably increase urine production. However, this pronounced natriuretic effect is limited to an acute setting. Inhibiting NKCC2 located at the macula densa can increase renin secretion, potentially exacerbating RAAS overactivation, leading to greater reabsorption of sodium. Prolonged use of loop diuretics leads to a gradual decline in net sodium excretion, which is termed “braking phenomenon.”17 Despite guidelines allow titrating the maximum furosemide dosage up to 160 mg/day, clinicians often consider the dose-effect relationship of loop diuretic to be linear. In fact, these agents demonstrate steep dose-response curves, with minimal effects observed until a threshold dose is reached. Beyond this threshold, response rapidly peaks, a phenomenon known as the “ceiling effect.” Consequently, once the ceiling is achieved, further increases in diuretic dosages no longer enhance urinary sodium excretion.17 Yet, in patients with cirrhotic ascites, the dose-response curve shifts downward and to the right, reducing the ceiling height, as observed in heart failure cases. These suggest that increasing loop diuretic dosage is futile for urine volume improvement.

MINERALOCORTICOID RECEPTOR ANTAGONISTS

As the most commonly used neurohormonal modulator, spironolactone was developed over 60 years ago as a class of potent MRAs. It was primarily used for the clinical treatment of congestive heart failure, primary aldosteronism, essential hypertension, and other pathologies associated with aldosteronism, such as ascites secondary to decompensated cirrhosis. Spironolactone is metabolized in the liver into three active metabolites that bind to cytoplasmic mineralocorticoid receptors, functioning as aldosterone antagonists, and produce a potassium-sparing diuretic effect in the distal tubules. All three active metabolites have demonstrated long half-life (range, 13.8 to 16.5 hours).18 However, liver cirrhosis can significantly impair the metabolic process, extending the metabolites’ half-lives from 23.9 hours to 126 hours.19 In clinical practice, this slows pharmacokinetic characteristics poses a challenge for physicians in titrating the optimal dose of spironolactone. Optimal dosage adjustment can take several weeks and is often accompanied by electrolyte disturbances related to the accumulation of active constituents during the treatment.20 Due to its simultaneous blockage of progesterone receptors and androgen receptors, spironolactone also commonly causes challenging side effects such as menstrual irregularities, impotence, and gynecomastia.

Unlike spironolactone, novel nonsteroidal MRAs showed significantly higher selectivity for aldosterone receptors and greater affinity in head-to-head trials.21 Its nonsteroidal structure allows for more efficient binding to aldosterone receptors, inhibiting inflammatory and pro-fibrotic gene expression due to hyperaldosteronemia, thus slowing the pace of kidney structure remodeling and functional deterioration among diabetic patients.22 The advantages of nonsteroidal MRAs are evidenced not only by their higher selectivity and affinity for mineral receptor but also by their inactive metabolites, rapid onset of action, and significantly shorter half-life, lasting only 2 hours.23 In addition, nonsteroidal MRAs have shown a better safety profile regarding hyperkalemia, with incidence less than half those associated with spironolactone (5.3% for finerenone vs 12.7% for spironolactone) in a phase II clinical trial.24

In the FIDELIO-DKD research, the nonsteroidal MRA finerenone considerably decreased the renal composite endpoint by 18% in patients with type 2 diabetes and chronic kidney diseases. The study also indicated a potential reduction in the incidence of hospitalization due to heart failure (hazard ratio, 0.86; 95% confidence interval, 0.68 to 1.08).25 Another phase III trial investigated the compositecardiovascular events (including death from cardiovascular causes, nonfatal stroke, nonfatal myocardial infarction, or hospitalization caused by heart failure) in patients withchronic kidney disease and type 2 diabetes. Finerenone decreased the incidence of heart failure hospitalization by 29%, while only 1.2% of the finerenone group discontinued treatment due to hyperkalemia, compared to 0.4% in the placebo group.26 Importantly, neither of these trials reported any hepatotoxicity-related adverse events. Currently, there is no direct evidence supporting the use of nonsteroidal MRAs in the management of cirrhotic ascites. However, given that RAAS overactivation is a common pathogenic mechanism in cirrhotic ascites, heart failure and diabetes mellitus, further investigation into nonsteroidal MRAs is warranted.

VAPTANS

Vaptans, also known as vasopressin receptor antagonists, disrupt the interaction between ADH and vasopressin V2 receptors, thereby facilitating the excretion of free water. Mechanistically, these medications do not affect the RAAS activity as they merely mitigate water retention without concomitant excretion of urine sodium. This action can lead to sodium retention and subsequent recurrence of water retention.27 Moreover, a rapid elevation of serum sodium concentrations could further stimulate osmotic ADH production,28 and volume depletion would further compromise hemodynamics. Given these limitations, vaptans cannot provide long-term relief from cirrhotic ascites.

Several clinical studies have reported favorable outcomes with short-term (≤2 weeks) weight reduction, coupled with the correction of hyponatremia, in patients with cirrhotic ascites.29 However, a 12-week study showed satavaptan offered no significant advantages over placebo in terms of ascites regression or large-volume paracentesis requirements.30 Interestingly, similar findings have also been observed in the treatment of heart failure. Although tolvaptan alleviated dyspnea symptoms among hospitalized patients with acute congestive heart failure,31,32 it did not offer any long-term clinical benefits.33 Notably, vaptans were associated with hepatic toxicity as an adverse effect in patients without underlying liver diseases.34 Accordingly, current guidelines do not endorse the routine use of vaptans in treating cirrhotic ascites.1,35

THE EXPLORATION OF RAAS BASED INTERVENTION

Liver cirrhosis and chronic heart failure share similar pathophysiological mechanism (Fig. 2).36 Generally, the reduction in effective arterial blood volume, whether due to splanchnic vasodilation in decompensated liver cirrhosis or the loss of cardiac output in heart failure, can increase RAAS activity. In addition, non-osmotic ADH release is further increased by sympathetic excitation due to relative hypovolemia,37 which in turn enhances RAAS activity through renal β-adrenergic stimulation.38 These neurohumoral system changes trigger clinical symptoms linked to water and sodium retention, such as ascites in liver cirrhosis and acute pulmonary edema in heart failure, along with a commonly observed abnormality in laboratory assay, dilutional hyponatremia. Numerous RAAS-modulating agents, which have demonstrated efficacy in the management of heart failure, have been explored for their utility in the treatment of hepatic ascites. Regrettably, unlike the favorable outcomes with angiotensin-converting enzyme inhibitors (ACEIs) and nonselective β-blockers in heart failure patients, these agents have not shown clinical benefits among decompensated cirrhosis patients with ascites.39,40

Figure 2. The common mechanism between heart failure and liver cirrhosis. The activation of the RAAS takes the pivotal place in the pathophysiology of chronic heart failure and decompensated cirrhosis. NKCC2, sodium-potassium-chloride cotransporter 2; SGLT2, sodium-glucose cotransporter-2; RAAS, renin-angiotensin-aldosterone system; ACEI, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; ARNI, angiotensin receptor neprilysin inhibitor; MRAs, mineralocorticoid receptor antagonists; SGLT2i, SGLT2 inhibitor; TIPS, transjugular intrahepatic portosystemic shunt. Adapted from Saffo S, et al. Clin Liver Dis (Hoboken) 2018;11:141-144, with permission from Wolters Kluwer Health, Inc.44

1. Angiotensin-converting enzyme inhibitors

This class of agents disrupts RAAS activity, yet does not directly impact renin levels. ACEIs block the angiotensin-converting enzyme that transforms angiotensin I to angiotensin II, thereby reducing the production of angiotensin II. A double-blind randomized controlled trial suggested that captopril substantially reduced glomerular filtration rate and urine sodium excretion in individuals with cirrhosis.41 Further research demonstrated that in patients with cirrhotic ascites, captopril dramatically reduced natriuresis, urine production, and impeded the natriuretic effect of loop diuretics.42 Pathophysiologically, ACEIs dilate the efferent arterioles and drastically diminish the glomerular filtration rate, leading to reduced urine sodium and urine volume, ultimately exacerbating the water and sodium retention. Further investigations have revealed that, contrary to expectations, ACEIs could not suppress RAAS activity among cirrhotic patients but instead increased plasma renin activity.43 Since the vasodilatory impact by ACEIs on the small arteries would be a strong stimulus to RAAS through the feedback-activated pathways and overwhelm its direct influence on RAAS. To date, attempts to manage cirrhotic ascites with ACEIs have been unsuccessful.

2. Nonselective β-blockers

Nonselective β-blockers are established as a cornerstone of pharmacotherapy for patients with compensated cirrhosis, as demonstrated by multiple trials. These agents not only reduce portal pressure but also inhibit renin expression and secretion,45,46 effectively preventing decompensation occurrence among cirrhosis patients with clinically significant portal hypertension.47 A meta-analysis showed that long-term carvedilol therapy could reduce the risk of progression to decompensated stage and improve survival in patients with cirrhosis, primarily by reducing ascites.48 Despite their success in compensated cirrhosis, β-blockers have shown very limited effects on lowering portal pressure or suppressing RAAS once the disease progresses to decompensated stage.5 In particular, for patients with advanced liver cirrhosis and compromised cardiac output function, the negative inotropic effects of nonselective β-blockers significantly worsen the hemodynamic condition, contributing to elevated mortality rates in those awaiting a liver transplant.5

We conducted a systematic review to comprehensively evaluate the impact of drugs that act directly or indirectly on the RAAS in patients with decompensated cirrhosis, summarizing our finding in Table 1.6,40-44,49-92 The search process and inclusion-exclusion criteria for literature are detailed in the Supplementary Tables 1, 2 and Supplementary Fig. 1. Our review revealed that although these drugs inhibit the overactivated RAAS, most studies observed a decrease in urinary sodium or a reduction in urine volume, a characteristic inherent to ACEI/ARB or nonselective β-blockers. This suggests that successful modulating RAAS may be challenging without simultaneous improvements in urinary sodium and urine volume.

Table 1 . The Summary of ACEI/ARB or Nonselective β-Blockers in the Treatment of Liver Cirrhosis.

Author (year)Study typeNo. of participantsInterventionResult
Tergast et al. (2023)49Propensity scored matching study123 vs 41RAS-inhibitorsRAS-Inhibitor is associated with lower incidences of grade III AKI.
Danielsen et al. (2023)50Cross-over study39Propranolol infusionRenal artery blood flow fell by –5%.
Nabilou et al. (2022)51Prospective study38Propranolol infusionEffect of β-blockade on cardiac index is less potent in advanced cirrhosis.
Singh et al. (2022)52Prospective study160Propranolol vs endoscopic variceal ligationPPL is associated with lower survival, poor control of ascites, and increased risk of AKI when compared with EVL.
Chen et al. (20220)53Propensity scored matching study1,788 vs 1,788Propranolol user vs not usedPropranolol was associated with reduced mortality in patients with cirrhosis and ascites.
Tapper et al. (2022)54Population survey study63,364NSBBThe risk of ascites was higher for persons taking any NSBB.
Kang et al. (2021)55Retrospective study740NSBB user vs not usedNSBB therapy was associated with longer survival in prophylactic treatment of esophageal varices.
Sasso et al. (2021)56Retrospective study2,165NSBB user vs not usedUse of NSBB for patients with cirrhosis was associated with fewer infection-related admissions.
McDowell et al. (2021)57Retrospective study152Carvedilol vs variceal band ligationcarvedilol offers a longer survival benefit than patients receiving EVL.
Kalambokis et al. (2021)58Prospective study32 vs 64Continued use propranolol vs switch to carvedilolIn patients with cirrhosis and nonrefractory ascites, as carvedilol improves renal perfusion and clinical outcomes.
Téllez et al. (2020)59Clinical trial20 vs 18NSBBNSBB impair global circulatory homeostasis and renal function in cirrhotic patients with refractory ascites.
Alvarado-Tapias et al. (2020)60Comparative study403NSBBThe short-term effect of β-blockers on cardiac output may adversely influence survival in patients with decompensated cirrhosis.
Yoo et al. (2020)61Retrospective study271EVL vs propranolol +EVLEVL alone is a more appropriate treatment option for prophylaxis of esophageal varices than propranolol combination therapy in cirrhotic ascites patients.
Ngwa et al. (2020)62Retrospective study65 vs 105NSBB user vs not usedNSBB use was associated with lower 90-day mortality.
Giannelli et al. (2020)6Retrospective study584NSBBNSBB use in cirrhotic patients with compromised cardiac performance increase the mortality before liver transplant.
Tergast et al. (2019)63Retrospective study624NSBBTreatment with NSBB was associated with a higher 28-day transplant-free survival, but no benefit in mean arterial blood pressure <65 mm Hg group.
Chen et al. (2019)64Clinical trial60 vs 61GVO + carvedilol vs GVOcarvedilol + GVO did not decrease recurrence of EGVB, no impact on survival time, but produced more adverse events.
Giannitrapani et al. (2018)65Retrospective study230NSBB user vs not usedThe use of NSBB indicated a higher risk of PVT.
Zampino et al. (2018)66Retrospective study130NSBBNSBB treatment were independent risk factors of PVT.
Pfisterer et al. (2018)67Retrospective study766NSBBNSBB do not increase efficacy of band ligation in primary prophylaxis, but they improve survival in secondary prophylaxis of variceal bleeding.
Onali et al. (2017)68Retrospective study316NSBBPatients with ascites on NSBB did not have impaired survival compared to those not receiving NSSB.
Sinha et al. (2017)69Retrospective study325CarvedilolLow dose, chronic treatment with carvedilol in patients with liver cirrhosis and ascites is not detrimental.
Kim et al. (2017)70Retrospective study2,361NSBBNSBB in patients with ascites significantly increased the risk of AKI.
Bossen et al. (2016)71Retrospective study1,198NSBBUse of NSBBs in cirrhosis patients with ascites did not increase mortality.
Mookerjee et al. (2016)72Prospective observational study349NSBBNSBBs in cirrhotic patients is safe and reduces the mortality if they develop ACLF.
Mandorfer et al. (2014)73Retrospective study607NSBBNSBBs increase the risks for hepatorenal syndrome, acute kidney injury and reduce transplant-free survival.
Sersté et al. (2011)74Clinical trial10NSBBNSBB might be associated with a high risk of paracentesis-induced circulatory dysfunction.
Sersté et al. (2010)75Observational prospective study151NSBBThe use of NSBB was associated with poor survival in patients with refractory ascites.
Therapondos et al. (2006)76Before-and-after control study10LosartanLow dose losartan did not ameliorate erect posture-induced sodium retention in post-TIPS ascites-free patients.
Cholongitas et al. (2006)77Retrospective study134PropranololPropranolol was not associated with a lower risk for SBP.
Groszmann et al. (2005)78Clinical trial213NSBBNSBB was ineffective in preventing varices in unselected patients with cirrhosis.
Abecasis et al. (2003)79Clinical trial100Nadolol + placebo vs Nadolol + spironolactoneNadolol plus spironolactone effectively reduced the incidence of both portal-hypertensive complications.
Sen et al. (2002)80Clinical trial20spironolactone, alone or with propranololSpironolactone in combination with propranolol achieved adequate reduction in HVPG in propranolol-resistant portal hypertension.
Wong et al. (2002)81Clinical trial10Losartanbeneficial natriuretic effects of low-dose losartan on erect posture-induced sodium retention.
De et al. (2002)82Clinical trial36Carvedilol vs propranololCarvedilol reduced portal pressure in both acutely and over 7 days, but not superior to propranolol.
Lee et al. (2000)83Before-and-after control study25CarvedilolSingle dose of captopril decreased glomerular filtration rate and increased plasma renin activity.
Forrest et al. (1996)84Comparative study16CarvedilolCarvedilol did not compromise renal perfusion but increase the risk of hypotension in ascitic patients.
Tsai et al. (1996)43Clinical trial50CaptoprilCaptopril did not improve sodium and water retention in cirrhotic patients with ascites.
Amarapurkar et al. (1994)85Comparative study68EnalaprilEnalapril improved creatinine clearance in patients with liver cirrhosis.
Ohnishi et al. (1994)40Before-and-after control study10EnalaprilEnalapril slightly increased daily urinary volume and sodium excretion.
Gentilini et al. (1993)41Double-blind, cross-over clinical trialNACaptoprilLow dose of captopril decreased glomerular filtration rate and sodium excretion in ascitic patients.
van Vliet et al. (1992)86Before-and-after control study8CaptoprilAmong diuretic resistance patients, half of them increases urine sodium after low-dose captopril administered.
Ibarra et al. (1992)87Before-and-after control study9Captopril7 of 9 showed enhanced natriuresis after captopril administered.
Poynard et al. (1991)88Clinical trial589NSBBPropranolol and nadolol were effective in preventing first bleeding and reducing the mortality rate.
Group (1989)89Clinical trial174PropranololPropranolol prevented first gastrointestinal bleeding in cirrhotic patients.
Daskalopoulos et al. (1987)42Before-and-after control study11CaptoprilUrinary volume was reduced and natriuretic effect of furosemide was blunted.
Pariente et al. (1985)90Before-and-after control study6CaptoprilCaptopril decreased mean arterial pressure and glomerular filtration.
Rector et al. (1984)91Before-and-after control study13PropranololPropranolol induced an anti-natriuretic effect.
Lebrec et al. (1984)92Clinical trial74PropranololPropranolol increased the survival rate in cirrhotic patients.

ACEI, angiotensin-converting enzyme inhibitor; ARB, angiotensin II receptor blocker; RAS, renin-angiotensin system; AKI, acute kidney injury; PPL, propranolol; EVL, endoscopic variceal ligation; NSBB, nonselective beta-blocker; GVO, gastroesophageal variceal obliteration; EGVB, esophagogastric variceal bleeding; PVT, portal vein thrombosis; ACLF, acute-on-chronic liver failure; TIPS, transjugular intrahepatic portosystemic shunt; SBP, spontaneous bacterial peritonitis; HVPG, hepatic venous pressure gradient..



3. Albumin: RAAS modulator rather than plasma expansion

Albumin comprises about three-quarters of the oncotic pressure and is often classed as a plasma expander in patients with cirrhosis. As a plasma expander, albumin effectively mitigates relative hypovolemia,93 and corrects hyponatremia by increasing free water clearance in cirrhosis.94 Clinical trials have demonstrated that the combination of albumin and loop diuretics is more effective than loop diuretics alone in controlling ascites,95,96 and considerably improve survival in cirrhotic patients with ascites.97,98 Moreover, unlike other artificial colloids, albumin uniquely prevents circulatory deterioration following large-volume paracentesis.99 Current guidelines therefore recommend administering 8 g of albumin after each liter of ascites removal.1 Albumin infusion could restore blood pressure, increase blood volume and improve cardiac index in patients with cirrhosis.6,100,101 The circulatory benefits of albumin in cirrhosis patients cannot be solely attributed to its volume-expanding function. If the primary function of albumin were to expand volume, its initial administration as a plasma expander should lead to a decrease in plasma sodium concentrations (5% albumin solution and 25% solution contain 130 to 160 mEq/L sodium and are considered isotonic with plasma). Stated in another way, there should be a decline in plasma sodium concentration upon albumin infusion at initial stage. However, the results from the ATTIRE study, which evaluated albumin administration in hospitalized decompensated cirrhotic patients, challenged this perception. The study's appendix indicated that serum sodium levels started to increase within 24 hours among patients in albumin group, with a significant difference in serum sodium levels between the two study groups observed within 48 hours and this difference maintained throughout the trial.102 In the ANSWER study, long-term albumin infusion was proved to bring about prolonged overall survival with the concurrent improvement of hyponatremia in patients with decompensated cirrhosis.98 Intriguingly, if albumin was regarded solely as a plasma expander, it would be difficult to rationalize why transfusion of other blood products increases portal pressure,103 whereas the incidence of or gastro-esophageal variceal bleeding was not increased in albumin group in the ANSWER study.98 Another study highlighted albumin’s dose-dependent effect on reducing cytokine plasma concentrations and alleviating cardiocirculatory dysfunction without significant increase in portal pressure.104 The evidence supports the view that albumin's role far extends beyond that of a plasma expander, further exploration of its pleiotropic effects apart from its oncotic properties is needed.

Approximately one-third of the body’s total albumin resides in the vascular bed, with the remainder distributed within the extravascular and interstitial space. Albumin constantly exchanges between the intravascular and extravascular compartments, with approximately 4% to 5% of total albumin per hour traversing the capillary endothelium to the interstitial space before, then returning to the veins via lymphatics system.92 The plasma volume expansion effect of 5% albumin infusion only lasts only 4 to 6 hours, which is significantly weaker than that provided by other artificial colloids.105 This suggests that the principle of the therapeutic benefits derived from albumin should not primarily be attributed to its volume expansion effect. Decompensated cirrhosis is characterized by systemic pro-oxidant and pro-inflammatory environments. Furthermore, gut microbiota dysbiosis and translocation lead to the release of pro-inflammatory mediators, which can increase the release of vasodilatory molecules and exacerbate the overactivation of RAAS. In patients with decompensated cirrhosis, it is not only the quantity of albumin that is insufficient; the pleiotropic non-oncotic properties of albumin, including antioxidant capacity, free radical scavenging, immune regulation, and endothelial protection function are also impaired,93 resulting in a significant deficiency in “effective” albumin. In patients with decompensated cirrhosis, the levels of interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF) are significantly elevated. It has been confirmed in other diseases that IL-6 can stimulate the secretion of renin, thereby affecting the expression of angiotensin II and potentially impacting RAAS components.106 Moreover, IL-6 can influence VEGF production, which enhances angiogenesis and alters vascular structure, thus affecting the vascular response to angiotensin II and indirectly influencing RAAS activity.107 In the Pilot-PRECIOSA and INFECIR-2 study, large-volume albumin transfusions increased body albumin level and reduced the levels of IL-6 and VEGF, thereby regulating RAAS activity through both direct and indirect pathways.104,108 In addition, albumin is capable of indirectly modulating the RAAS system by restoring left ventricular function, compensating for relative hypovolemia in patients with liver cirrhosis,100,109 a property is not fully reliant upon changes in preload,110 however, other artificial colloids do not provide similar benefits on this aspect of circulatory function.111 Additionally, albumin corrects molecule profile abnormalities by reducing endogenous NO production, which in turn restores peripheral vessel resistance,112,113 improves vascular endothelial function, counteracts endotoxin-induced inflammation and oxidative stress,114 and diminishes systemic inflammation.113 These effects ultimately improve circulatory blood volume. Therefore, it can be inferred that albumin acts as a RAAS modulator in the treatment of cirrhotic ascites.

4. Sodium-glucose cotransporter type 2 inhibitors in cirrhotic ascites treatment

In the setting of RAAS overactivation in decompensated cirrhosis, sodium reabsorption markedly increases at the proximal tubular,115 the sodium concentration in tubule fluid flow to distal nephron therefore reduced. In this case, the diuretic effect of both conventional MRAs and loop diuretics would be blunted, given that they act at the ionic channel in the distal nephron. Additionally, proximal tubular sodium reabsorption is commonly involved in long-standing or refractory ascites.115,116 In order to intervene in the process of excessive sodium reabsorption in the proximal tubules, it is imperative to introduce novel approaches.

Sodium-glucose cotransporter-2 (SGLT2) inhibitors belong to a class of oral medications for treating type 2 diabetes.117 By inhibiting SGLT2, the agents significantly reduce the reabsorption of sodium and glucose in the proximal convoluted tubule. The increased sodium concentration in tubule fluid senses by the macula densa would in turn inhibit the renin release from the juxtaglomerular cells, then the increased sodium concentration in tubular fluid is a stimulus for tubuloglomerular feedback and causing vasoconstriction of the afferent arteriole.118 In spite of slightly reduced glomerular filtration, the osmotic diuresis caused by increased sodium concentration in the tubule fluid maintains the adequate amount of urine.119 The complementarity in mechanism suggests that SGLT2 inhibitors could exert a synergistic effect with loop diuretics and mitigate the braking phenomenon (Fig. 3).120 Their characteristics on promoting urinary excretion of solutes have been recently applied to treat advanced heart failure regardless of preexistence of diabetes.121,122 Given that advanced heart failure and decompensated liver cirrhosis share common pathophysiological features, it is reasonable to infer that SGLT2 inhibitors might be a candidate for treating cirrhotic ascites as well.123

Figure 3. The individual and synergistic effect of furosemide and empagliflozin on urinary sodium excretion and tubuloglomerular feedback. When furosemide inhibits NKCC2 in the macula densa, it reduces the ability to sense sodium and chloride levels, leading to an increased release of renin. This process suppresses the tubuloglomerular feedback. SGLT2 inhibitors increase the sodium concentration in the tubular fluid, which in turn raises the sodium and chloride levels passing through the macula densa. This increase in ion concentration is detected by the osmoreceptors in the macula densa, subsequently activating the tubuloglomerular feedback. Thus, these two drugs synergistically optimize fluid management in patients with cirrhotic ascites by balancing renin release and maintaining sodium levels. RAAS, renin-angiotensin-aldosterone system; NKCC2, sodium-potassiumchloride cotransporter 2; SGLT2, sodium-glucose cotransporter-2.

Encouragingly, several case reports have observed regression of ascites after adding SGLT2 inhibitors to the treatment for patients with decompensated cirrhosis and comorbid diabetes, without appreciable adverse events.124,125 Interestingly, the correction of hyponatremia observed in these cases might also suggest potential capacity on tempering RAAS with these drugs.125-127 An interesting parallel finding in heart failure treatment, in DAPA-HF trial, dapagliflozin increased urine volume and free water clearance persistently for several weeks, the correction of hyponatremia is along with the improvement of clinical outcomes.128 A mechanism-based explanation had been proposed from the editorial comment,129 SGLT2 inhibitors probably alleviate the water and sodium retention without further stimulate the RAAS activation as well as sympathetic nervous system. This class of agents therefore has potential to become a promising complement to the current pharmacologic treatments for ascitic patients. Moreover, the clinical benefits of SGLT2 inhibitors among patients with advanced heart failure do not rely on the existence of glycemia,121,122 it merits further investigation on ascitic patients regardless of comorbid diabetes.

Before repurposing SGLT2 inhibitors for cirrhotic ascites in a clinical setting, it is imperative to evaluate whether SGLT2 expression increases in the late-stage cirrhotic model and whether the proximal tubular sodium reabsorption increase is mediated by SGLT2. Additionally, it is essential to determine whether RAAS activity regulates the upregulation of SGLT2 expression in liver cirrhosis. It is probable that the renal pathological changes in progressive cirrhosis resemble those found in heart failure models.130

Hepatic glycogen capacity is impaired in patients with decompensated liver cirrhosis, predisposing them to hypoglycemia.131 However, the impact of SGLT2 inhibitors on blood glucose levels is not dependent on the level of insulin, the magnitude of blood glucose reduction is dependent on plasma glucose level among diabetic population,132 and less likely to induce a hypoglycemic event. Data from two phase III trials suggest SGLT2 inhibitors showed a comparable incidence with placebo in regard to liver dysfunction or clinical hypoglycemia regardless of diabetes existence.133 Moreover, hepatic dysfunction does not significantly affect the pharmacokinetics of SGLT2 inhibitors.134-136 An initial dip of estimated glomerular filtration rate and mild blood pressure drop (systolic and diastolic blood pressures decreased by 4 to 6 and 1 to 2 mm Hg respectively137 observed with SGLT2 inhibitors may raise worries about hepato-renal syndrome, but in practice, such change does not irritate RAAS components,134-136,138,139 it appears unlikely that SGLT2 inhibitors would cause hepato-renal syndrome in absence of an aggravation of RAAS status.

By far, the potential value of SGLT2 inhibitors in this field has begun to be gradually recognized,140 it is also encouraging to explore this new avenue of therapeutic approach (NCT05014594, NCT05013502, NCT05430243, NCT05726032).

CONCLUSION

In conclusion, despite decades of established medication therapy for cirrhotic ascites, significant therapeutic benefits remain elusive. However, growing understanding of the pathophysiological mechanisms underlying RAAS offers promise prospect for repurposing novel MRAs and SGLT2 inhibitors in ascites treatment. These efforts may pave the way toward more effective management of the complication of decompensated liver cirrhosis.

ACKNOWLEDGEMENTS

This work is supported by the Beijing Natural Science Foundation (No. 7232081), National Key Research and Development Program of China (No. 2022YFC2304400), the Beijing Hospitals Authority’s Ascent Plan (No. DFL20221501), and the Construction Project of High-level Technology Talents in Public Health (No. Discipline leader-01-12).

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

SUPPLEMENTARY MATERIALS

Supplementary materials can be accessed at https://doi.org/10.5009/gnl240038.

Fig 1.

Figure 1.Hemodynamic manifestations of liver cirrhosis. The main hemodynamic manifestation of liver cirrhosis is a progressive decrease in arterial vessel resistance due to splanchnic arterial vasodilatation. Initially, this is compensated by an increase in cardiac output (CO). However, compromised cardiac function in decompensated stage further deteriorates effective circulating blood volume insufficiency. Finally, the components in neurohumoral system have to be activated. RAAS, renin-angiotensin-aldosterone system; ADH, antidiuretic hormone. Adapted from Arroyo V, et al. Ann Hepatol 2011;10 Suppl 1:S6-S14.5
Gut and Liver 2024; :

Fig 2.

Figure 2.The common mechanism between heart failure and liver cirrhosis. The activation of the RAAS takes the pivotal place in the pathophysiology of chronic heart failure and decompensated cirrhosis. NKCC2, sodium-potassium-chloride cotransporter 2; SGLT2, sodium-glucose cotransporter-2; RAAS, renin-angiotensin-aldosterone system; ACEI, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; ARNI, angiotensin receptor neprilysin inhibitor; MRAs, mineralocorticoid receptor antagonists; SGLT2i, SGLT2 inhibitor; TIPS, transjugular intrahepatic portosystemic shunt. Adapted from Saffo S, et al. Clin Liver Dis (Hoboken) 2018;11:141-144, with permission from Wolters Kluwer Health, Inc.44
Gut and Liver 2024; :

Fig 3.

Figure 3.The individual and synergistic effect of furosemide and empagliflozin on urinary sodium excretion and tubuloglomerular feedback. When furosemide inhibits NKCC2 in the macula densa, it reduces the ability to sense sodium and chloride levels, leading to an increased release of renin. This process suppresses the tubuloglomerular feedback. SGLT2 inhibitors increase the sodium concentration in the tubular fluid, which in turn raises the sodium and chloride levels passing through the macula densa. This increase in ion concentration is detected by the osmoreceptors in the macula densa, subsequently activating the tubuloglomerular feedback. Thus, these two drugs synergistically optimize fluid management in patients with cirrhotic ascites by balancing renin release and maintaining sodium levels. RAAS, renin-angiotensin-aldosterone system; NKCC2, sodium-potassiumchloride cotransporter 2; SGLT2, sodium-glucose cotransporter-2.
Gut and Liver 2024; :

Table 1 The Summary of ACEI/ARB or Nonselective β-Blockers in the Treatment of Liver Cirrhosis

Author (year)Study typeNo. of participantsInterventionResult
Tergast et al. (2023)49Propensity scored matching study123 vs 41RAS-inhibitorsRAS-Inhibitor is associated with lower incidences of grade III AKI.
Danielsen et al. (2023)50Cross-over study39Propranolol infusionRenal artery blood flow fell by –5%.
Nabilou et al. (2022)51Prospective study38Propranolol infusionEffect of β-blockade on cardiac index is less potent in advanced cirrhosis.
Singh et al. (2022)52Prospective study160Propranolol vs endoscopic variceal ligationPPL is associated with lower survival, poor control of ascites, and increased risk of AKI when compared with EVL.
Chen et al. (20220)53Propensity scored matching study1,788 vs 1,788Propranolol user vs not usedPropranolol was associated with reduced mortality in patients with cirrhosis and ascites.
Tapper et al. (2022)54Population survey study63,364NSBBThe risk of ascites was higher for persons taking any NSBB.
Kang et al. (2021)55Retrospective study740NSBB user vs not usedNSBB therapy was associated with longer survival in prophylactic treatment of esophageal varices.
Sasso et al. (2021)56Retrospective study2,165NSBB user vs not usedUse of NSBB for patients with cirrhosis was associated with fewer infection-related admissions.
McDowell et al. (2021)57Retrospective study152Carvedilol vs variceal band ligationcarvedilol offers a longer survival benefit than patients receiving EVL.
Kalambokis et al. (2021)58Prospective study32 vs 64Continued use propranolol vs switch to carvedilolIn patients with cirrhosis and nonrefractory ascites, as carvedilol improves renal perfusion and clinical outcomes.
Téllez et al. (2020)59Clinical trial20 vs 18NSBBNSBB impair global circulatory homeostasis and renal function in cirrhotic patients with refractory ascites.
Alvarado-Tapias et al. (2020)60Comparative study403NSBBThe short-term effect of β-blockers on cardiac output may adversely influence survival in patients with decompensated cirrhosis.
Yoo et al. (2020)61Retrospective study271EVL vs propranolol +EVLEVL alone is a more appropriate treatment option for prophylaxis of esophageal varices than propranolol combination therapy in cirrhotic ascites patients.
Ngwa et al. (2020)62Retrospective study65 vs 105NSBB user vs not usedNSBB use was associated with lower 90-day mortality.
Giannelli et al. (2020)6Retrospective study584NSBBNSBB use in cirrhotic patients with compromised cardiac performance increase the mortality before liver transplant.
Tergast et al. (2019)63Retrospective study624NSBBTreatment with NSBB was associated with a higher 28-day transplant-free survival, but no benefit in mean arterial blood pressure <65 mm Hg group.
Chen et al. (2019)64Clinical trial60 vs 61GVO + carvedilol vs GVOcarvedilol + GVO did not decrease recurrence of EGVB, no impact on survival time, but produced more adverse events.
Giannitrapani et al. (2018)65Retrospective study230NSBB user vs not usedThe use of NSBB indicated a higher risk of PVT.
Zampino et al. (2018)66Retrospective study130NSBBNSBB treatment were independent risk factors of PVT.
Pfisterer et al. (2018)67Retrospective study766NSBBNSBB do not increase efficacy of band ligation in primary prophylaxis, but they improve survival in secondary prophylaxis of variceal bleeding.
Onali et al. (2017)68Retrospective study316NSBBPatients with ascites on NSBB did not have impaired survival compared to those not receiving NSSB.
Sinha et al. (2017)69Retrospective study325CarvedilolLow dose, chronic treatment with carvedilol in patients with liver cirrhosis and ascites is not detrimental.
Kim et al. (2017)70Retrospective study2,361NSBBNSBB in patients with ascites significantly increased the risk of AKI.
Bossen et al. (2016)71Retrospective study1,198NSBBUse of NSBBs in cirrhosis patients with ascites did not increase mortality.
Mookerjee et al. (2016)72Prospective observational study349NSBBNSBBs in cirrhotic patients is safe and reduces the mortality if they develop ACLF.
Mandorfer et al. (2014)73Retrospective study607NSBBNSBBs increase the risks for hepatorenal syndrome, acute kidney injury and reduce transplant-free survival.
Sersté et al. (2011)74Clinical trial10NSBBNSBB might be associated with a high risk of paracentesis-induced circulatory dysfunction.
Sersté et al. (2010)75Observational prospective study151NSBBThe use of NSBB was associated with poor survival in patients with refractory ascites.
Therapondos et al. (2006)76Before-and-after control study10LosartanLow dose losartan did not ameliorate erect posture-induced sodium retention in post-TIPS ascites-free patients.
Cholongitas et al. (2006)77Retrospective study134PropranololPropranolol was not associated with a lower risk for SBP.
Groszmann et al. (2005)78Clinical trial213NSBBNSBB was ineffective in preventing varices in unselected patients with cirrhosis.
Abecasis et al. (2003)79Clinical trial100Nadolol + placebo vs Nadolol + spironolactoneNadolol plus spironolactone effectively reduced the incidence of both portal-hypertensive complications.
Sen et al. (2002)80Clinical trial20spironolactone, alone or with propranololSpironolactone in combination with propranolol achieved adequate reduction in HVPG in propranolol-resistant portal hypertension.
Wong et al. (2002)81Clinical trial10Losartanbeneficial natriuretic effects of low-dose losartan on erect posture-induced sodium retention.
De et al. (2002)82Clinical trial36Carvedilol vs propranololCarvedilol reduced portal pressure in both acutely and over 7 days, but not superior to propranolol.
Lee et al. (2000)83Before-and-after control study25CarvedilolSingle dose of captopril decreased glomerular filtration rate and increased plasma renin activity.
Forrest et al. (1996)84Comparative study16CarvedilolCarvedilol did not compromise renal perfusion but increase the risk of hypotension in ascitic patients.
Tsai et al. (1996)43Clinical trial50CaptoprilCaptopril did not improve sodium and water retention in cirrhotic patients with ascites.
Amarapurkar et al. (1994)85Comparative study68EnalaprilEnalapril improved creatinine clearance in patients with liver cirrhosis.
Ohnishi et al. (1994)40Before-and-after control study10EnalaprilEnalapril slightly increased daily urinary volume and sodium excretion.
Gentilini et al. (1993)41Double-blind, cross-over clinical trialNACaptoprilLow dose of captopril decreased glomerular filtration rate and sodium excretion in ascitic patients.
van Vliet et al. (1992)86Before-and-after control study8CaptoprilAmong diuretic resistance patients, half of them increases urine sodium after low-dose captopril administered.
Ibarra et al. (1992)87Before-and-after control study9Captopril7 of 9 showed enhanced natriuresis after captopril administered.
Poynard et al. (1991)88Clinical trial589NSBBPropranolol and nadolol were effective in preventing first bleeding and reducing the mortality rate.
Group (1989)89Clinical trial174PropranololPropranolol prevented first gastrointestinal bleeding in cirrhotic patients.
Daskalopoulos et al. (1987)42Before-and-after control study11CaptoprilUrinary volume was reduced and natriuretic effect of furosemide was blunted.
Pariente et al. (1985)90Before-and-after control study6CaptoprilCaptopril decreased mean arterial pressure and glomerular filtration.
Rector et al. (1984)91Before-and-after control study13PropranololPropranolol induced an anti-natriuretic effect.
Lebrec et al. (1984)92Clinical trial74PropranololPropranolol increased the survival rate in cirrhotic patients.

ACEI, angiotensin-converting enzyme inhibitor; ARB, angiotensin II receptor blocker; RAS, renin-angiotensin system; AKI, acute kidney injury; PPL, propranolol; EVL, endoscopic variceal ligation; NSBB, nonselective beta-blocker; GVO, gastroesophageal variceal obliteration; EGVB, esophagogastric variceal bleeding; PVT, portal vein thrombosis; ACLF, acute-on-chronic liver failure; TIPS, transjugular intrahepatic portosystemic shunt; SBP, spontaneous bacterial peritonitis; HVPG, hepatic venous pressure gradient.


References

  1. European Association for the Study of the Liver. EASL clinical practice guidelines for the management of patients with decompensated cirrhosis. J Hepatol 2018;69:406-460.
    Pubmed CrossRef
  2. European Association for the Study of the Liver. EASL clinical practice guidelines on the management of ascites, spontaneous bacterial peritonitis, and hepatorenal syndrome in cirrhosis. J Hepatol 2010;53:397-417.
    Pubmed CrossRef
  3. Hudson B, Round J, Georgeson B, et al. Cirrhosis with ascites in the last year of life: a nationwide analysis of factors shaping costs, health-care use, and place of death in England. Lancet Gastroenterol Hepatol 2018;3:95-103.
    Pubmed CrossRef
  4. Volk ML, Tocco RS, Bazick J, Rakoski MO, Lok AS. Hospital readmissions among patients with decompensated cirrhosis. Am J Gastroenterol 2012;107:247-252.
    Pubmed KoreaMed CrossRef
  5. Arroyo V, Fernandez J. Pathophysiological basis of albumin use in cirrhosis. Ann Hepatol 2011;10 Suppl 1:S6-S14.
    CrossRef
  6. Giannelli V, Roux O, Laouénan C, et al. Impact of cardiac function, refractory ascites and beta blockers on the outcome of patients with cirrhosis listed for liver transplantation. J Hepatol 2020;72:463-471.
    Pubmed CrossRef
  7. Iwakiri Y. Pathophysiology of portal hypertension. Clin Liver Dis 2014;18:281-291.
    Pubmed KoreaMed CrossRef
  8. Iwakiri Y. The molecules: mechanisms of arterial vasodilatation observed in the splanchnic and systemic circulation in portal hypertension. J Clin Gastroenterol 2007;41 Suppl 3:S288-S294.
    Pubmed CrossRef
  9. Iwakiri Y, Groszmann RJ. Vascular endothelial dysfunction in cirrhosis. J Hepatol 2007;46:927-934.
    Pubmed CrossRef
  10. Chodorowski Z, Sein Anand J, Nowak-Banasik L, Szydłowska M, Klimek J, Kaletha K. Carbon monoxide: a regulator of vascular tone in hypoxia?. Przegl Lek 2005;62:438-440.
  11. Stitham J, Gleim SR, Douville K, Arehart E, Hwa J. Versatility and differential roles of cysteine residues in human prostacyclin receptor structure and function. J Biol Chem 2006;281:37227-37236.
    Pubmed CrossRef
  12. Pesce M, Esposito G, Sarnelli G. Endocannabinoids in the treatment of gasytrointestinal inflammation and symptoms. Curr Opin Pharmacol 2018;43:81-86.
    Pubmed CrossRef
  13. Alukal JJ, John S, Thuluvath PJ. Hyponatremia in cirrhosis: an update. Am J Gastroenterol 2020;115:1775-1785.
    Pubmed CrossRef
  14. Barrera-Chimal J, Girerd S, Jaisser F. Mineralocorticoid receptor antagonists and kidney diseases: pathophysiological basis. Kidney Int 2019;96:302-319.
    Pubmed CrossRef
  15. John S, Thuluvath PJ. Hyponatremia in cirrhosis: pathophysiology and management. World J Gastroenterol 2015;21:3197-3205.
    Pubmed KoreaMed CrossRef
  16. Xu X, Duan Z, et al; Chinese Society of Hepatology; Chinese Medical Association. Chinese guidelines on the management of ascites and its related complications in cirrhosis. Hepatol Int 2019;13:1-21.
    Pubmed CrossRef
  17. 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;75:1178-1195.
    Pubmed CrossRef
  18. Gardiner P, Schrode K, Quinlan D, et al. Spironolactone metabolism: steady-state serum levels of the sulfur-containing metabolites. J Clin Pharmacol 1989;29:342-347.
    Pubmed CrossRef
  19. Sungaila I, Bartle WR, Walker SE, et al. Spironolactone pharmacokinetics and pharmacodynamics in patients with cirrhotic ascites. Gastroenterology 1992;102:1680-1685.
    Pubmed CrossRef
  20. de Denus S, Leclair G, Dubé MP, et al. Spironolactone metabolite concentrations in decompensated heart failure: insights from the ATHENA-HF trial. Eur J Heart Fail 2020;22:1451-1461.
    Pubmed KoreaMed CrossRef
  21. Barrera-Chimal J, Kolkhof P, Lima-Posada I, Joachim A, Rossignol P, Jaisser F. Differentiation between emerging non-steroidal and established steroidal mineralocorticoid receptor antagonists: head-to-head comparisons of pharmacological and clinical characteristics. Expert Opin Investig Drugs 2021;30:1141-1157.
    Pubmed CrossRef
  22. Bakris GL, Agarwal R, Anker SD, et al. Design and baseline characteristics of the finerenone in reducing kidney failure and disease progression in diabetic kidney disease trial. Am J Nephrol 2019;50:333-344.
    Pubmed KoreaMed CrossRef
  23. Yang P, Huang T, Xu G. The novel mineralocorticoid receptor antagonist finerenone in diabetic kidney disease: progress and challenges. Metabolism 2016;65:1342-1349.
    Pubmed CrossRef
  24. Pitt B, Kober L, Ponikowski P, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J 2013;34:2453-2463.
    Pubmed KoreaMed CrossRef
  25. Bakris GL, Agarwal R, Anker SD, et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med 2020;383:2219-2229.
    Pubmed CrossRef
  26. Pitt B, Filippatos G, Agarwal R, et al. Cardiovascular events with finerenone in kidney disease and type 2 diabetes. N Engl J Med 2021;385:2252-2263.
    Pubmed CrossRef
  27. Zeidel ML. Salt and water: not so simple. J Clin Invest 2017;127:1625-1626.
    Pubmed KoreaMed CrossRef
  28. Bankir L, Guerrot D, Bichet DG. Vaptans or voluntary increased hydration to protect the kidney: how do they compare?. Nephrol Dial Transplant 2023;38:562-574.
    Pubmed CrossRef
  29. Yan L, Xie F, Lu J, et al. The treatment of vasopressin V2-receptor antagonists in cirrhosis patients with ascites: a meta-analysis of randomized controlled trials. BMC Gastroenterol 2015;15:65.
    Pubmed KoreaMed CrossRef
  30. Wong F, Watson H, Gerbes A, et al. Satavaptan for the management of ascites in cirrhosis: efficacy and safety across the spectrum of ascites severity. Gut 2012;61:108-116.
    Pubmed CrossRef
  31. Gheorghiade M, Gattis WA, O'Connor CM, et al. Effects of tolvaptan, a vasopressin antagonist, in patients hospitalized with worsening heart failure: a randomized controlled trial. JAMA 2004;291:1963-1971.
    Pubmed CrossRef
  32. Gheorghiade M, Konstam MA, Burnett JC, et al. Short-term clinical effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for heart failure: the EVEREST Clinical Status Trials. JAMA 2007;297:1332-1343.
    Pubmed CrossRef
  33. Konstam MA, Gheorghiade M, Burnett JC, et al. Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial. JAMA 2007;297:1319-1331.
    Pubmed CrossRef
  34. Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients with autosomal dominant polycystic kidney disease. N Engl J Med 2012;367:2407-2418.
    Pubmed KoreaMed CrossRef
  35. Aithal GP, Palaniyappan N, China L, et al. Guidelines on the management of ascites in cirrhosis. Gut 2021;70:9-29.
    Pubmed KoreaMed CrossRef
  36. 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;2:370-376.
    Pubmed CrossRef
  37. Bichet DG, Van Putten VJ, Schrier RW. Potential role of increased sympathetic activity in impaired sodium and water excretion in cirrhosis. N Engl J Med 1982;307:1552-1557.
    Pubmed CrossRef
  38. Berl T, Henrich WL, Erickson AL, Schrier RW. Prostaglandins in the beta-adrenergic and baroreceptor-mediated secretion of renin. Am J Physiol 1979;236:F472-F477.
    Pubmed CrossRef
  39. Wong RJ, Robinson A, Ginzberg D, Gomes C, Liu B, Bhuket T. Assessing the safety of beta-blocker therapy in cirrhosis patients with ascites: a meta-analysis. Liver Int 2019;39:1080-1088.
    Pubmed CrossRef
  40. Ohnishi A, Murakami S, Harada M, et al. Renal and hormonal responses to repeated treatment with enalapril in non-azotemic cirrhosis with ascites. J Hepatol 1994;20:223-230.
    Pubmed CrossRef
  41. Gentilini P, Romanelli RG, La Villa G, et al. Effects of low-dose captopril on renal hemodynamics and function in patients with cirrhosis of the liver. Gastroenterology 1993;104:588-594.
    Pubmed CrossRef
  42. Daskalopoulos G, Pinzani M, Murray N, Hirschberg R, Zipser RD. Effects of captopril on renal function in patients with cirrhosis and ascites. J Hepatol 1987;4:330-336.
    Pubmed CrossRef
  43. Tsai YT, Lin HC, Lee FY, Hou MC, Wang SS, Lee SD. Effects of captopril on renal functions, renal and portal hemodynamics in patients with cirrhosis. Proc Natl Sci Counc Repub China B 1996;20:44-50.
  44. Saffo S, Taddei T. SGLT2 inhibitors and cirrhosis: a unique perspective on the comanagement of diabetes mellitus and ascites. Clin Liver Dis (Hoboken) 2018;11:141-144.
    Pubmed KoreaMed CrossRef
  45. Fung JW, Yu CM, Yip G, et al. Effect of beta blockade (carvedilol or metoprolol) on activation of the renin-angiotensin-aldosterone system and natriuretic peptides in chronic heart failure. Am J Cardiol 2003;92:406-410.
    Pubmed CrossRef
  46. Tsuyuki RT, McAlister FA, Teo KK. Beta-blockers for congestive heart failure: what is the current consensus?. Drugs Aging 2000;16:1-7.
    Pubmed CrossRef
  47. Villanueva C, Albillos A, Genescà J, et al. β blockers to prevent decompensation of cirrhosis in patients with clinically significant portal hypertension (PREDESCI): a randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2019;393:1597-1608.
    Pubmed CrossRef
  48. Villanueva C, Torres F, Sarin SK, et al. Carvedilol reduces the risk of decompensation and mortality in patients with compensated cirrhosis in a competing-risk meta-analysis. J Hepatol 2022;77:1014-1025.
    Pubmed CrossRef
  49. Tergast TL, Griemsmann M, Wedemeyer H, Cornberg M, Maasoumy B. Effects of renin-angiotensin inhibitors on renal function and the clinical course in patients with decompensated cirrhosis. Sci Rep 2023;13:17486.
    Pubmed KoreaMed CrossRef
  50. Danielsen KV, Nabilou P, Wiese SS, Hove JD, Bendtsen F, Møller S. Effect of beta-blockers on multiple haemodynamics in cirrhosis: a cross-over study by MR-imaging and hepatic vein catheterization. Liver Int 2023;43:2245-2255.
    Pubmed CrossRef
  51. Nabilou P, Danielsen KV, Kimer N, Hove JD, Bendtsen F, Møller S. Blunted cardiovascular effects of beta-blockers in patients with cirrhosis: relation to severity?. PLoS One 2022;17:e0270603.
    Pubmed KoreaMed CrossRef
  52. Singh V, Kumar P, Verma N, Vijayvergiya R, Singh A, Bhalla A. Propranolol vs. band ligation for primary prophylaxis of variceal hemorrhage in cirrhotic patients with ascites: a randomized controlled trial. Hepatol Int 2022;16:944-953.
    Pubmed CrossRef
  53. Chen YC, Li YD, Lu CM, Huang WC, Kao SS, Chen WC. Propranolol use in patients with cirrhosis and refractory ascites: a nationwide study. Saudi J Gastroenterol 2022;28:108-114.
    Pubmed KoreaMed CrossRef
  54. Tapper EB, Zhao Z, Mazumder N, Parikh ND. Incidence of, risk factors for, and outcomes after ascites in a population-based cohort of older Americans. Dig Dis Sci 2022;67:5327-5335.
    Pubmed KoreaMed CrossRef
  55. Kang SH, Lee M, Kim MY, et al. The longitudinal outcomes of applying non-selective beta-blockers in portal hypertension: real-world multicenter study. Hepatol Int 2021;15:424-436.
    Pubmed CrossRef
  56. Sasso R, Rockey DC. Non-selective beta-blocker use in cirrhotic patients is associated with a reduced likelihood of hospitalisation for infection. Aliment Pharmacol Ther 2021;53:418-425.
    Pubmed CrossRef
  57. McDowell HR, Chuah CS, Tripathi D, Stanley AJ, Forrest EH, Hayes PC. Carvedilol is associated with improved survival in patients with cirrhosis: a long-term follow-up study. Aliment Pharmacol Ther 2021;53:531-539.
    Pubmed CrossRef
  58. Kalambokis GN, Christaki M, Tsiakas I, et al. Conversion of propranolol to carvedilol improves renal perfusion and outcome in patients with cirrhosis and ascites. J Clin Gastroenterol 2021;55:721-729.
    Pubmed CrossRef
  59. Téllez L, Ibáñez-Samaniego L, Pérez Del Villar C, et al. Non-selective beta-blockers impair global circulatory homeostasis and renal function in cirrhotic patients with refractory ascites. J Hepatol 2020;73:1404-1414.
    Pubmed CrossRef
  60. Alvarado-Tapias E, Ardevol A, Garcia-Guix M, et al. Short-term hemodynamic effects of beta-blockers influence survival of patients with decompensated cirrhosis. J Hepatol 2020;73:829-841.
    Pubmed CrossRef
  61. Yoo JJ, Kim SG, Kim YS, et al. Propranolol plus endoscopic ligation for variceal bleeding in patients with significant ascites: Propensity score matching analysis. Medicine (Baltimore) 2020;99:e18913.
    Pubmed KoreaMed CrossRef
  62. Ngwa T, Orman E, Gomez EV, et al. Non-selective beta blocker use is associated with improved short-term survival in patients with cirrhosis referred for liver transplantation. BMC Gastroenterol 2020;20:4.
    Pubmed KoreaMed CrossRef
  63. Tergast TL, Kimmann M, Laser H, et al. Systemic arterial blood pressure determines the therapeutic window of non-selective beta blockers in decompensated cirrhosis. Aliment Pharmacol Ther 2019;50:696-706.
    Pubmed CrossRef
  64. Chen WC, Hsin IF, Chen PH, et al. Addition of carvedilol to gastric variceal obturation does not decrease recurrence of gastric variceal bleeding in patients with cirrhosis. Clin Gastroenterol Hepatol 2019;17:2356-2363.
    Pubmed CrossRef
  65. Giannitrapani L, Granà W, Licata A, Schiavone C, Montalto G, Soresi M. Nontumorous portal vein thrombosis in liver cirrhosis: possible role of beta-blockers. Med Princ Pract 2018;27:466-471.
    Pubmed KoreaMed CrossRef
  66. Zampino R, Lebano R, Coppola N, et al. The use of nonselective beta blockers is a risk factor for portal vein thrombosis in cirrhotic patients. Saudi J Gastroenterol 2018;24:25-29.
    Pubmed KoreaMed CrossRef
  67. Pfisterer N, Dexheimer C, Fuchs EM, et al. Betablockers do not increase efficacy of band ligation in primary prophylaxis but they improve survival in secondary prophylaxis of variceal bleeding. Aliment Pharmacol Ther 2018;47:966-979.
    Pubmed CrossRef
  68. Onali S, Kalafateli M, Majumdar A, et al. Non-selective beta-blockers are not associated with increased mortality in cirrhotic patients with ascites. Liver Int 2017;37:1334-1344.
    Pubmed CrossRef
  69. Sinha R, Lockman KA, Mallawaarachchi N, Robertson M, Plevris JN, Hayes PC. Carvedilol use is associated with improved survival in patients with liver cirrhosis and ascites. J Hepatol 2017;67:40-46.
    Pubmed CrossRef
  70. Kim SG, Larson JJ, Lee JS, Therneau TM, Kim WR. Beneficial and harmful effects of nonselective beta blockade on acute kidney injury in liver transplant candidates. Liver Transpl 2017;23:733-740.
    Pubmed KoreaMed CrossRef
  71. Bossen L, Krag A, Vilstrup H, Watson H, Jepsen P. Nonselective beta-blockers do not affect mortality in cirrhosis patients with ascites: post hoc analysis of three randomized controlled trials with 1198 patients. Hepatology 2016;63:1968-1976.
    Pubmed CrossRef
  72. Mookerjee RP, Pavesi M, Thomsen KL, et al. Treatment with non-selective beta blockers is associated with reduced severity of systemic inflammation and improved survival of patients with acute-on-chronic liver failure. J Hepatol 2016;64:574-582.
    Pubmed CrossRef
  73. Mandorfer M, Bota S, Schwabl P, et al. Nonselective beta blockers increase risk for hepatorenal syndrome and death in patients with cirrhosis and spontaneous bacterial peritonitis. Gastroenterology 2014;146:1680-1690.
    Pubmed CrossRef
  74. Sersté T, Francoz C, Durand F, et al. Beta-blockers cause paracentesis-induced circulatory dysfunction in patients with cirrhosis and refractory ascites: a cross-over study. J Hepatol 2011;55:794-799.
    Pubmed CrossRef
  75. Sersté T, Melot C, Francoz C, et al. Deleterious effects of beta-blockers on survival in patients with cirrhosis and refractory ascites. Hepatology 2010;52:1017-1022.
    Pubmed CrossRef
  76. Therapondos G, Hol L, Benjaminov F, Wong F. The effect of single oral low- dose losartan on posture-related sodium handling in post-TIPS ascites-free cirrhosis. Hepatology 2006;44:640-649.
    Pubmed CrossRef
  77. Cholongitas E, Papatheodoridis GV, Manesis EK, Burroughs AK, Archimandritis AJ. Spontaneous bacterial peritonitis in cirrhotic patients: is prophylactic propranolol therapy beneficial?. J Gastroenterol Hepatol 2006;21:581-587.
    Pubmed CrossRef
  78. Groszmann RJ, Garcia-Tsao G, Bosch J, et al. Beta-blockers to prevent gastroesophageal varices in patients with cirrhosis. N Engl J Med 2005;353:2254-2261.
    Pubmed CrossRef
  79. Abecasis R, Kravetz D, Fassio E, et al. Nadolol plus spironolactone in the prophylaxis of first variceal bleed in nonascitic cirrhotic patients: a preliminary study. Hepatology 2003;37:359-365.
    Pubmed CrossRef
  80. Sen S, De BK, Biswas PK, Biswas J, Das D, Maity AK. Hemodynamic effect of spironolactone in liver cirrhosis and propranolol-resistant portal hypertension. Indian J Gastroenterol 2002;21:145-148.
  81. Wong F, Liu P, Blendis L. The mechanism of improved sodium homeostasis of low-dose losartan in preascitic cirrhosis. Hepatology 2002;35:1449-1458.
    Pubmed CrossRef
  82. De BK, Das D, Sen S, et al. Acute and 7-day portal pressure response to carvedilol and propranolol in cirrhotics. J Gastroenterol Hepatol 2002;17:183-189.
    Pubmed CrossRef
  83. Lee JK, Hsieh JF, Tsai SC, Ho YJ, Kao CH. Effects of single dose of 50mg captopril in patients with liver cirrhosis and ascites. Hepatogastroenterology 2000;47:767-770.
    Pubmed
  84. Forrest EH, Bouchier IA, Hayes PC. Acute haemodynamic changes after oral carvedilol, a vasodilating beta-blocker, in patients with cirrhosis. J Hepatol 1996;25:909-915.
    Pubmed CrossRef
  85. Amarapurkar DN, Dhawan P, Kalro RH. Role of routine estimation of creatinine clearance in patients with liver cirrhosis. Indian J Gastroenterol 1994;13:79-82.
  86. van Vliet AA, Hackeng WH, Donker AJ, Meuwissen SG. Efficacy of low-dose captopril in addition to furosemide and spironolactone in patients with decompensated liver disease during blunted diuresis. J Hepatol 1992;15:40-47.
    Pubmed CrossRef
  87. Ibarra FR, Afione C, Garzon D, Barontini M, Santos JC, Arrizurieta E. Portal pressure, renal function and hormonal profile after acute and chronic captopril treatment in cirrhosis. Eur J Clin Pharmacol 1992;43:477-482.
    Pubmed CrossRef
  88. Poynard T, Calès P, Pasta L, et al. Beta-adrenergic-antagonist drugs in the prevention of gastrointestinal bleeding in patients with cirrhosis and esophageal varices. An analysis of data and prognostic factors in 589 patients from four randomized clinical trials. Franco-Italian Multicenter Study Group. N Engl J Med 1991;324:1532-1538.
    Pubmed CrossRef
  89. The Italian Multicenter Project for Propranolol in Prevention of Bleeding. Propranolol prevents first gastrointestinal bleeding in non-ascitic cirrhotic patients. Final report of a multicenter randomized trial. J Hepatol 1989;9:75-83.
    Pubmed CrossRef
  90. Pariente EA, Bataille C, Bercoff E, Lebrec D. Acute effects of captopril on systemic and renal hemodynamics and on renal function in cirrhotic patients with ascites. Gastroenterology 1985;88(5 Pt 1):1255-1259.
    Pubmed CrossRef
  91. Rector WG Jr, Reynolds TB. Propranolol in the treatment of cirrhotic ascites. Arch Intern Med 1984;144:1761-1763.
    Pubmed CrossRef
  92. Lebrec D, Poynard T, Bernuau J, et al. A randomized controlled study of propranolol for prevention of recurrent gastrointestinal bleeding in patients with cirrhosis: a final report. Hepatology 1984;4:355-358.
    Pubmed CrossRef
  93. Bernardi M, Angeli P, Claria J, et al. Albumin in decompensated cirrhosis: new concepts and perspectives. Gut 2020;69:1127-1138.
    Pubmed KoreaMed CrossRef
  94. McCormick PA, Mistry P, Kaye G, Burroughs AK, McIntyre N. Intravenous albumin infusion is an effective therapy for hyponatraemia in cirrhotic patients with ascites. Gut 1990;31:204-207.
    Pubmed KoreaMed CrossRef
  95. Gentilini P, Casini-Raggi V, Di Fiore G, et al. Albumin improves the response to diuretics in patients with cirrhosis and ascites: results of a randomized, controlled trial. J Hepatol 1999;30:639-645.
    Pubmed CrossRef
  96. Laffi G, Gentilini P, Romanelli RG, La Villa G. Is the use of albumin of value in the treatment of ascites in cirrhosis? The case in favour. Dig Liver Dis 2003;35:660-663.
    Pubmed CrossRef
  97. Romanelli RG, La Villa G, Barletta G, et al. Long-term albumin infusion improves survival in patients with cirrhosis and ascites: an unblinded randomized trial. World J Gastroenterol 2006;12:1403-1407.
    Pubmed KoreaMed CrossRef
  98. Caraceni P, Riggio O, Angeli P, et al. Long-term albumin administration in decompensated cirrhosis (ANSWER): an open-label randomised trial. Lancet 2018;391:2417-2429.
    Pubmed CrossRef
  99. Bernardi M, Caraceni P, Navickis RJ, Wilkes MM. Albumin infusion in patients undergoing large-volume paracentesis: a meta-analysis of randomized trials. Hepatology 2012;55:1172-1181.
    Pubmed CrossRef
  100. Shasthry SM, Kumar M, Khumuckham JS, Sarin SK. Changes in cardiac output and incidence of volume overload in cirrhotics receiving 20% albumin infusion. Liver Int 2017;37:1167-1176.
    Pubmed CrossRef
  101. Luca A, García-Pagán JC, Bosch J, et al. Beneficial effects of intravenous albumin infusion on the hemodynamic and humoral changes after total paracentesis. Hepatology 1995;22:753-758.
    Pubmed CrossRef
  102. China L, Freemantle N, Forrest E, et al. A randomized trial of albumin infusions in hospitalized patients with cirrhosis. N Engl J Med 2021;384:808-817.
    Pubmed CrossRef
  103. Elizalde JI, Moitinho E, García-Pagán JC, et al. Effects of increasing blood hemoglobin levels on systemic hemodynamics of acutely anemic cirrhotic patients. J Hepatol 1998;29:789-795.
    Pubmed CrossRef
  104. Fernández J, Clària J, Amorós A, et al. Effects of albumin treatment on systemic and portal hemodynamics and systemic inflammation in patients with decompensated cirrhosis. Gastroenterology 2019;157:149-162.
    Pubmed CrossRef
  105. Quon CY. Clinical pharmacokinetics and pharmacodynamics of colloidal plasma volume expanders. Journal of Cardiothoracic Anesthesia 1988;2:13-23.
    CrossRef
  106. Schieffer B, Schieffer E, Hilfiker-Kleiner D, et al. Expression of angiotensin II and interleukin 6 in human coronary atherosclerotic plaques: potential implications for inflammation and plaque instability. Circulation 2000;101:1372-1378.
    Pubmed CrossRef
  107. Catar R, Witowski J, Zhu N, et al. IL-6 trans-signaling links inflammation with angiogenesis in the peritoneal membrane. J Am Soc Nephrol 2017;28:1188-1199.
    Pubmed KoreaMed CrossRef
  108. Uriz J, Ginès P, Cárdenas A, et al. Terlipressin plus albumin infusion: an effective and safe therapy of hepatorenal syndrome. J Hepatol 2000;33:43-48.
    Pubmed CrossRef
  109. Fernández J, Navasa M, Garcia-Pagan JC, et al. Effect of intravenous albumin on systemic and hepatic hemodynamics and vasoactive neurohormonal systems in patients with cirrhosis and spontaneous bacterial peritonitis. J Hepatol 2004;41:384-390.
    Pubmed CrossRef
  110. Bortoluzzi A, Ceolotto G, Gola E, et al. Positive cardiac inotropic effect of albumin infusion in rodents with cirrhosis and ascites: molecular mechanisms. Hepatology 2013;57:266-276.
    Pubmed CrossRef
  111. Fernández J, Monteagudo J, Bargallo X, et al. A randomized unblinded pilot study comparing albumin versus hydroxyethyl starch in spontaneous bacterial peritonitis. Hepatology 2005;42:627-634.
    Pubmed CrossRef
  112. Walley KR, McDonald TE, Wang Y, Dai S, Russell JA. Albumin resuscitation increases cardiomyocyte contractility and decreases nitric oxide synthase II expression in rat endotoxemia. Crit Care Med 2003;31:187-194.
    Pubmed CrossRef
  113. Arroyo V, García-Martinez R, Salvatella X. Human serum albumin, systemic inflammation, and cirrhosis. J Hepatol 2014;61:396-407.
    Pubmed CrossRef
  114. Kremer H, Baron-Menguy C, Tesse A, et al. Human serum albumin improves endothelial dysfunction and survival during experimental endotoxemia: concentration-dependent properties. Crit Care Med 2011;39:1414-1422.
    Pubmed CrossRef
  115. Salerno F, Guevara M, Bernardi M, et al. Refractory ascites: pathogenesis, definition and therapy of a severe complication in patients with cirrhosis. Liver Int 2010;30:937-947.
    Pubmed CrossRef
  116. Gatta A, Angeli P, Caregaro L, Menon F, Sacerdoti D, Merkel C. A pathophysiological interpretation of unresponsiveness to spironolactone in a stepped-care approach to the diuretic treatment of ascites in nonazotemic cirrhotic patients. Hepatology 1991;14:231-236.
    Pubmed CrossRef
  117. Washburn WN, Poucher SM. Differentiating sodium-glucose co-transporter-2 inhibitors in development for the treatment of type 2 diabetes mellitus. Expert Opin Investig Drugs 2013;22:463-486.
    Pubmed CrossRef
  118. Braunwald E. Gliflozins in the management of cardiovascular disease. N Engl J Med 2022;386:2024-2034.
    Pubmed CrossRef
  119. Tanaka H, Takano K, Iijima H, et al. Factors affecting canagliflozin-induced transient urine volume increase in patients with type 2 diabetes mellitus. Adv Ther 2017;34:436-451.
    Pubmed KoreaMed CrossRef
  120. Verma A, Patel AB, Waikar SS. SGLT2 inhibitor: not a traditional diuretic for heart failure. Cell Metab 2020;32:13-14.
    Pubmed CrossRef
  121. Packer M, Anker SD, Butler J, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med 2020;383:1413-1424.
    Pubmed CrossRef
  122. McMurray JJ, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 2019;381:1995-2008.
    Pubmed CrossRef
  123. 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;9:795-797.
    Pubmed KoreaMed CrossRef
  124. Mordi NA, Mordi IR, Singh JS, McCrimmon RJ, Struthers AD, Lang CC. Renal and cardiovascular effects of SGLT2 inhibition in combination with loop diuretics in patients with type 2 diabetes and chronic heart failure: the RECEDE-CHF trial. Circulation 2020;142:1713-1724.
    Pubmed KoreaMed CrossRef
  125. 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;116:618-619.
    Pubmed CrossRef
  126. 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;15:831-833.
    Pubmed KoreaMed CrossRef
  127. 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;72:1880-1882.
    Pubmed CrossRef
  128. Yeoh SE, Docherty KF, Jhund PS, et al. Relationship of dapagliflozin with serum sodium: findings from the DAPA-HF trial. JACC Heart Fail 2022;10:306-318.
    Pubmed CrossRef
  129. Li S, Levy WC. Impact of SGLT2 inhibitors on serum sodium in heart failure with reduced ejection fraction. JACC Heart Fail 2022;10:319-320.
    Pubmed CrossRef
  130. Katsurada K, Nandi SS, Sharma NM, Patel KP. Enhanced expression and function of renal SGLT2 (sodium-glucose cotransporter 2) in heart failure: role of renal nerves. Circ Heart Fail 2021;14:e008365.
    Pubmed KoreaMed CrossRef
  131. Pfortmueller CA, Wiemann C, Funk GC, et al. Hypoglycemia is associated with increased mortality in patients with acute decompensated liver cirrhosis. J Crit Care 2014;29:316.
    Pubmed CrossRef
  132. Polidori D, Sha S, Ghosh A, Plum-Mörschel L, Heise T, Rothenberg P. Validation of a novel method for determining the renal threshold for glucose excretion in untreated and canagliflozin-treated subjects with type 2 diabetes mellitus. J Clin Endocrinol Metab 2013;98:E867-E871.
    Pubmed KoreaMed CrossRef
  133. Zannad F, Ferreira JP, Pocock SJ, et al. SGLT2 inhibitors in patients with heart failure with reduced ejection fraction: a meta-analysis of the EMPEROR-Reduced and DAPA-HF trials. Lancet 2020;396:819-829.
    Pubmed CrossRef
  134. Kasichayanula S, Liu X, Zhang W, Pfister M, LaCreta FP, Boulton DW. Influence of hepatic impairment on the pharmacokinetics and safety profile of dapagliflozin: an open-label, parallel-group, single-dose study. Clin Ther 2011;33:1798-1808.
    Pubmed CrossRef
  135. Macha S, Rose P, Mattheus M, et al. Pharmacokinetics, safety and tolerability of empagliflozin, a sodium glucose cotransporter 2 inhibitor, in patients with hepatic impairment. Diabetes Obes Metab 2014;16:118-123.
    Pubmed CrossRef
  136. Devineni D, Curtin CR, Marbury TC, et al. Effect of hepatic or renal impairment on the pharmacokinetics of canagliflozin, a sodium glucose co-transporter 2 inhibitor. Clin Ther 2015;37:610-628.
    Pubmed CrossRef
  137. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects of sodium-glucose cotransporter 2 inhibitors: JACC state-of-the-art review. J Am Coll Cardiol 2020;75:422-434.
    Pubmed CrossRef
  138. Ansary TM, Nakano D, Nishiyama A. Diuretic effects of sodium glucose cotransporter 2 inhibitors and their influence on the renin-angiotensin system. Int J Mol Sci 2019;20:629.
    Pubmed KoreaMed CrossRef
  139. Griffin M, Rao VS, Ivey-Miranda J, et al. Empagliflozin in heart failure: diuretic and cardiorenal effects. Circulation 2020;142:1028-1039.
    Pubmed KoreaMed CrossRef
  140. Miyamoto Y, Honda A, Yokose S, Nagata M, Miyamoto J. The effects of SGLT2 inhibitors on liver cirrhosis patients with refractory ascites: a literature review. J Clin Med 2023;12:2253.
    Pubmed KoreaMed CrossRef
Gut and Liver

Vol.18 No.5
September, 2024

pISSN 1976-2283
eISSN 2005-1212

qrcode
qrcode

Supplementary

Share this article on :

  • line

Popular Keywords

Gut and LiverQR code Download
qr-code

Editorial Office