Open access peer-reviewed chapter - ONLINE FIRST

Non-Selective Beta-Blockers in Patients with Cirrhosis: Current Evidence and Controversy

Written By

Ting Wang, Cai’e Wang, Hongyu Li and Ran Wang

Submitted: 21 May 2024 Reviewed: 22 May 2024 Published: 17 June 2024

DOI: 10.5772/intechopen.1005683

Liver Cirrhosis and Its Complications IntechOpen
Liver Cirrhosis and Its Complications Advances in Diagnosis and Management Edited by Ran Wang

From the Edited Volume

Liver Cirrhosis and Its Complications - Advances in Diagnosis and Management [Working Title]

Ran Wang and Xingshun Qi

Chapter metrics overview

11 Chapter Downloads

View Full Metrics

Abstract

Non-selective beta-blockers (NSBBs) are the cornerstone of management of liver cirrhosis and its complications. They decrease portal vein blood flow and portal vein pressure via antagonism of β1 and β2 receptors, thereby improving the hyperdynamic circulatory status and reducing the risk of decompensated events in cirrhotic patients with portal hypertension (PH). Currently, NSBBs have been recommended for the primary and secondary prevention of variceal bleeding in patients with cirrhosis. Nevertheless, it has been noted that the clinical risk-benefit ratio appears to deteriorate for NSBBs in patients with advanced liver cirrhosis. Considering that the use of NSBBs in the clinical practice of cirrhotic patients remains controversial, this chapter aims to summarize the evidence of current recommendations and controversies regarding the use of NSBBs in patients with cirrhosis.

Keywords

  • liver cirrhosis
  • NSBBs
  • PH
  • varices
  • ascites
  • spontaneous bacterial peritonitis
  • renal dysfunction
  • portal vein thrombosis

1. Introduction

Liver cirrhosis, characterized by progressive fibrosis and liver injury, is the end stage of several liver diseases [1]. Portal hypertension (PH), a primary consequence of cirrhosis, may cause serious complications, including variceal bleeding (VB), ascites, and hepatic encephalopathy (HE) [2, 3]. Currently, hepatic venous pressure gradient (HVPG) measurement is considered the gold standard test for the diagnosis of PH in clinical practice [4]. HVPG represents the actual liver portal perfusion pressure, and its value greater than 5 mmHg indicates PH. Portal pressure increases with progressive disease, ultimately surpassing cut-off values of 10 mmHg, which marks the occurrence of clinically significant PH (CSPH). Patients with CSPH and cirrhosis exhibit splanchnic and peripheral vasodilatation, resulting in increased heart rate and cardiac output, which is characteristic of the hyperdynamic circulation state of advanced PH [5, 6].

Non-selective beta-blockers (NSBBs) are the cornerstone of management of PH [7]. NSBBs reduce peripheral blood flow via antagonism of β1 receptor, thereby reducing the heart rate and cardiac output, as well as splanchnic vasoconstriction through inhibition of β2 receptor, thereby decreasing portal venous blood flow and portal pressure [8, 9]. Accordingly, NSBBs could improve the hyperdynamic circulatory status of CSPH patients and decrease the development of decompensated events in patients with compensated cirrhosis. Additionally, NSBBs may reduce bacterial translocation and inflammatory responses through non-hemodynamic mechanisms [10]. The traditional NSBBs commonly used in clinical practice include propranolol, nadolol, and timolol. Carvedilol is a novel NSBBs with an intrinsic anti-α-adrenergic activity, which stimulates the release of nitric oxide and causes intrahepatic vasodilatation, thereby further decreasing portal pressure [11].

According to the updated consensus and guidelines, NSBBs are specifically recommended for primary or secondary prophylaxis of VB in patients with high-risk varices [12, 13, 14]. It has been recently shown that NSBBs are not always beneficial to all cirrhotic patients [15]. Tonon et al. identified that the use of NSBBs may increase the risk of acute decompensation and non-acute decompensation in compensated patients [16]. In the window hypothesis, as proposed by Krag et al., it was suggested that the efficacy and safety of NSBBs may vary based on the spectrum of cirrhosis [17]. They questioned the use of NSBBs in early cirrhosis, which is free of medium or large varices, and warned to avoid NSBBs in patients with advanced cirrhosis and refractory ascites. This chapter aims to summarize the existing evidence on the indications and controversies for NSBBs in patients with cirrhosis, in order to provide a reference for the use of NSBBs in the clinical practice.

Advertisement

2. NSBBs in cirrhosis: indications

2.1 Clinically significant portal hypertension

CSPH is defined as HVPG ≥10 mmHg, as decompensation may develop after achieving this threshold [18]. Indeed, CSPH is the strong predictor of PH-associated complications such as VB, ascites, and HE in patients with compensated cirrhosis [19]. NSBB-induced reduction of HVPG at least 10% from baseline may prevent the first episode of VB and ascites [12]. A randomized, double-blind, multicenter trial (PREDESCI) in Spain included 201 compensated cirrhotic patients with CSPH and without high-risk varices. Patients were randomized to treatment with placebo or NSBBs (propranolol or carvedilol depending on hemodynamic response). Hemodynamic response to NSBBs was determined as a decrease of HVPG of ≥10% after 20 minutes of intravenous propranolol (0.15 mg/kg). They found that primary outcomes such as ascites, VB, or HE occurred in 16% of patients in the NSBBs group vs. 27% in the placebo group (P = 0.041). The difference was due to a reduced incidence of ascites (P = 0·0297) [20]. An individual participant data meta-analysis of four randomized controlled trials (RCTs) revealed that the risk of decompensation in the carvedilol group was significantly lower than in the control group, which included endoscopic variceal ligation (EVL) and placebo among compensated patients with CSPH (P = 0.017) [21]. Kumar et al. randomly assigned 136 acute-on-chronic liver failure patients who were definitively diagnosed with CSPH and had no or small varices to carvedilol or placebo groups, which demonstrated that the use of carvedilol improved 28-day survival and reduced the risk of acute kidney injury (AKI) [22]. Thus, it appears to suggest that patients may benefit from the initiation of NSBBs once the CSPH has been diagnosed, whatever the status of the varices.

Notably, it is suggested that carvedilol is more effective in decreasing HVPG than propranolol or nadolol, including propranolol non-responders, with about 75% of patients achieving an HVPG response [23]. Besides, the Baveno VII consensus indicated that administration with NSBBs should be considered to prevent decompensation in patients with CSPH. Carvedilol is recommended as the preferred NSBBs for compensated cirrhosis compared to the traditional NSBBs (i.e., propranolol and nadolol) [12]. Overall, screening patients with CSPH in compensation cirrhosis for prompt initiation of NSBBs (preferred to carvedilol) may improve prognosis.

2.2 Primary prophylaxis

Currently, international guidelines suggest that NSBBs and EVL are similarly effective for primary prophylaxis, which are both recommended for patients with high-risk varices such as medium to large varices or small varices with red signs or Child-Pugh C [12, 13].

In a study by McDowell et al., 152 patients were included to evaluate the efficacy of carvedilol for the primary prevention of VB and survival. It showed that the median survival was significantly higher in the carvedilol group than in the EVL group (7.8 vs. 4.2 years, P = 0.03) [24]. A meta-analysis of 32 RCTs showed that monotherapy with NSBBs may reduce the risk of the first VB and mortality in cirrhotic patients with large varices, with a lower incidence of serious complications than EVL [25]. However, both these studies included patients with compensated cirrhosis and those with decompensated cirrhosis. Notably, the hemodynamic response of patients to NSBBs in cirrhosis varies across the spectrum of cirrhosis [26, 27]. In addition, the primary management goals for patients with cirrhosis also depend on the stage of cirrhosis. In patients with compensated cirrhosis, the major objective of treatment is not limited to the prevention of VB, which must focus on the prevention of decompensation events (ascites and HE). Thus, it is now preferred to choose treatments based on the stage of the disease. A competing-risk meta-analysis of 11 RCTs evaluated the efficacy of NSBBs vs. EVL on survival in patients with high-risk varices, stratifying risk based on the compensated/decompensated stage of cirrhosis. It showed that treatment with NSBBs had significantly improved survival compared with EVL in compensated cirrhotic patients, but in patients with decompensated cirrhosis, the survival was similar between the two therapies [28].

It is worth noting that the development of adverse events and the economic costs should be also considered while effective treatments are selected. Treatment with NSBBs has a lower risk of serious complications such as esophageal ulcers and esophageal perforation compared to EVL, which is more easily obtained, less depending on specialized endoscopic equipment like EVL, and has a higher economic efficiency.

2.3 Secondary prophylaxis

Nowadays, updated international guidelines and consensus recommend the use of NSBBs combined with EVL for secondary prophylaxis of VB [12, 13]. In addition, a combination therapy of NSBBs and isosorbide-mononitrate (IsMn) is also recommended as secondary prophylaxis of VB by the Japanese Society of Gastroenterology [14]. Kong et al. included 43 RCTs to compare the efficacy of medications, endoscopic options, transjugular intrahepatic portosystemic shunts (TIPS), and mainstay combination therapies in cirrhotic patients with at least one episode VB. It showed that nadolol combined with ISMN may be a preferable option to reduce mortality associated with rebleeding in patients with cirrhosis [29]. Another network meta-analysis of 26 RCTs showed that nadolol combined with EVL may be the preferred option for preventing rebleeding in cirrhotic patients who had one previous episode of VB, while NSBBs combined with ISMN may be potential alternatives to improve mortality [30]. Moreover, Jachs et al. identified that the cumulative incidences of rebleeding (P = 0.027) and liver-related death (P = 0.036) were significantly lower in patients administered with carvedilol combined with EVL versus propranolol combined with EVL [31].

Advertisement

3. NSBBs in cirrhosis: controversies

3.1 Preprimary prophylaxis

Currently, there is inadequate evidence to support the use of NSBBs for the prevention of the development or growth of varices in cirrhotic patients with no varices. Groszmann et al. randomly assigned 213 patients with HVPG ≥6 mm Hg without varices to the timolol group and the placebo group. During a median follow-up of 54.9 months, the incidence of the primary endpoint (development of gastroesophageal varices or VB) was not significantly different between the timolol and the placebo groups (39 vs. 40%, P = 0.89). Besides, severe adverse events were reported with a higher proportion of patients in the timolol group compared with the placebo group (18 vs. 6%, P = 0.006) [32]. It’s worth noting that most patients (59%) without varices had only subclinical PH (SPH) in this study. However, NSBBs exert a stronger effect for reduction of portal pressure in CSPH patients than in SPH patients who do not develop increased cardiac output and hyperdynamic circulation, which is a prerequisite for NSBBs to exert their activity [26]. This explains why NSBBs are ineffective generally in preprimary prophylaxis. Similarly, a meta-analysis of six RCTs including patients with no or small varices also showed no significant benefit of NSBBs compared with placebo for the prevention of large varices, first VB, and death [33].

In contrast, Bhardwaj et al. showed that a lower proportion of patients in the carvedilol group had a progression of small varices to large varices compared to the placebo group [34]. Di Pascoli et al. conducted a long-term observation of patients who started with NSBBs when diagnosed with small varices versus those treated with NSBBs after the confirmation of large varices by endoscopy. They also found that the small-varices group had a significantly lower rate of adverse outcomes (progression of varices or VB or death) than the large-varices group and had lower economic costs [35]. In general, the evidence is conflicting regarding the effect of NSBBs on slowing the development of small varices into large varices in cirrhotic patients. Thus, treatment should be focused on elimination or control of the cause in liver disease at this stage. Additionally, it is also important to monitor the magnitude of hemodynamic changes to determine a hyperdynamic circulatory state and prevent decompensated events in patients with no or small varices.

3.2 Ascites and refractory ascites

In 2010, a milestone study by Serste et al. revealed that treatment with propranolol was associated with poor survival in patients with refractory ascites. This study created a lively debate in the hepatology field [36]. Notably, the NSBBs group had a higher proportion of Child-Pugh class C and varices, higher bilirubin levels, and lower serum sodium in this study, which might justify these differences. Furthermore, nearly half of the patients were treated with a high-dose propranolol of 160 mg reported by Serste et al. However, a real-world multicenter study found that low-dose (≤ 80 mg/d) NSBBs treatment was associated with improved survival in primary and secondary prophylaxis of VB among advanced cirrhosis [37]. Park et al. also showed that low-dose propranolol (40–120 mg/d) decreased the risk of rebleeding of varices and mortality in patients with refractory ascites [38].

The window hypothesis, as proposed by Krag et al., suggests that the benefit windows of NSBBs may be closed once patients develop refractory ascites [17]. It may be explained that proinflammatory cytokines are increased in the splanchnic compartment among patients with refractory ascites, in whom chronic inflammation may cause decreased expression of β-adrenergic receptors and sympathetic nerve fiber loss, thereby leading to diminished effects of NSBBs [39]. However, several recent studies to investigate the efficacy of NSBBs in patients with ascites have reported opposite results. Tergast et al. enrolled 624 decompensated cirrhotic patients with ascites to evaluate the impact of NSBBs on survival. It showed that the administration of NSBBs is associated with improved 28-day transplant-free survival [40]. Leithead et al. included 322 patients with ascites who were listed for liver transplantation and identified significantly lower waiting-list mortality in NSBBs group [41]. Additionally, Onali et al. and Facciorusso et al. did not demonstrate that NSBBs affected the overall survival in cirrhotic patients with refractory ascites [42, 43].

Notably, the guideline for the management of ascites in cirrhosis by the British Society of Gastroenterology suggests that refractory ascites should not be considered a contraindication of NSBBs [44]. Patients with refractory ascites who are taking NSBBs should be closely monitored, and if they develop hypotension or acute/progressive renal dysfunction, the dosage may be reduced or discontinued appropriately. Similarly, the Baveno VII consensus proposed that NSBBs should be discontinued or decreased in patients with ascites who develop sustained hypotension (systolic blood pressure < 90 mmHg or mean arterial blood pressure < 65 mmHg) and/or hepato-renal syndrome-acute kidney injury (HRS-AKI). Once blood pressure returns to baseline and/or HRS-AKI resolves, NSBBs should be restarted [12]. In conclusion, the current evidence would not support discontinuation of NSBBs in patients with ascites or refractory ascites. NSBBs should be used with careful attention and reduced or discontinued in those, if appropriate.

3.3 NSBBs and renal dysfunction

Serste et al. included 139 patients with severe hepatitis to estimate the effect of NSBBs on the risk of development of AKI, in which the cumulative AKI incidence was significantly higher in the NSBBs group than in the non-NSBBs group (89.6 vs. 50.4%, P = 0.0001). They concluded that NSBBs were an independent risk factor for AKI in those patients [45]. Kim et al. identified that the impact of NSBBs on renal injury varied according to the severity of hepatic decompensation, with an increased risk of AKI by more than three-fold in cirrhotic patients with ascites [46]. Meanwhile, Singh et al. evaluated the effect of NSBBs on the development of AKI and 12-month transplant-free-survival in patients with ≥grade 2 ascites compared to EVL. They observed that the use of NSBBs is associated with poor survival and increased risk of AKI in those patients [47]. Finally, a meta-analysis including 14 studies, as reported by Xu et al., found that NSBBs significantly increased the risk of developing renal dysfunction [48]. The explanation for this apparent discrepancy lies in the fact that renal perfusion and its function are dependent on excessive activation of the sympathetic nervous system in patients with advanced cirrhosis, while NSBBs blunt the sympathetic overdrive, reduce cardiac output, and decrease renal perfusion pressures below critical values, thereby impairing renal function [49]. Notably, Xu et al. failed to demonstrate the association of NSBBs with an increased risk of development of renal dysfunction after covariate adjustment. Bang et al. and Scheiner et al. also found that treatment with NSBBs was associated neither with impairing of kidney function nor with increased incidence of AKI [50, 51].

However, Li et al. showed that the administration of NSBBs may reduce the occurrence of AKI and marginally alleviate chronic renal function declination [52]. Maiwall et al. observed consecutive cirrhotic patients with a complete record of baseline hemodynamics. During a median follow-up of 379 days, they found that treatment with NSBBs could prevent the frequency of AKI and the development of chronic kidney disease (CKD) [53]. It may be attributed to the fact that PH and severe vasodilation predispose cirrhotic patients to repeatedly develop AKI and progression to CKD, while NSBBs could improve hemodynamic response and reduce portal pressure, thereby decreasing the occurrence of AKI and CKD. Thus, the current evidence regarding the impact of NSBBs on renal dysfunction remains controversial. According to updated guidelines and consensus, NSBBs should be discontinued or dose-reduced in patients who develop AKI and CKD [12, 13].

3.4 NSBBs and spontaneous bacterial peritonitis

A meta-analysis included six studies to evaluate the possible role of NSBBs in spontaneous bacterial peritonitis (SBP) among cirrhotic patients with ascites, which found that NSBBs reduced the risk of SBP in the whole cohort, and the effect is still observed when compared between hemodynamic responders and non-responders [54]. It may be attributed to the non-hemodynamic mechanisms of NSBBs in reducing bacterial translocation and inflammatory response [55]. A large-scale retrospective cohort study from Denmark included 361 patients with SBP and revealed that the use of NSBBs was associated with improved survival. Additionally, in patients with severe decompensated cirrhosis, the use of NSBBs was associated with a lower risk of occurrence of SBP [50]. A prospective cohort study evaluated the possible factors associated with the prediction of recurrent SBP, which found that nonuse of prophylactic NSBBs was strongly associated with an increased rate of SBP recurrence [56].

However, Mandorfer et al. showed that treatment with NSBBs may be associated with poor transplant-free survival, increased risk of HRS and AKI, and extended periods of hospitalization in patients with SBP [57]. Notably, the NSBBs group had a higher bilirubin level, and a higher proportion of Child-Pugh class C and varices among SBP patients for subgroup analysis in this study might justify these differences. Similarly, according to the European Association for the Study of the Liver (EASL) clinical practice guideline, NSBBs should be discontinued in patients with acute complications such as VB, sepsis, SBP, or AKI. After recovery, it may be reinitiated appropriately [13]. Currently, controversy remains regarding the definitive efficacy of NSBBs in patients with SBP, but it is recommended to discontinue the use of NSBBs once the patients develop SBP. In future, large-scale prospective studies are needed to further explore the relationship between NSBBs and SBP.

3.5 NSBBs and portal vein thrombosis

Portal vein thrombosis (PVT) is mainly associated with decreased portal vein blood flow and velocity, and the portal vein blood flow <15 cm/s is considered an independent risk factor of PVT [58, 59]. The development of PVT is strongly associated with poor prognosis in cirrhotic patients [60, 61]. A significant higher risk of VB was observed in patients with PVT due to further increased portal pressure (47.33 vs. 19.63%) [62]. Currently, NSBBs are the first-line treatment for the prevention of VB, which significantly decrease the portal blood velocity and portal vein pressure [12]. However, Xu et al. indicated that a higher incidence of PVT was observed in the NSBBs group compared to the non-NSBBs group in univariate logistic regression analyses (P = 0.035), but no significant difference was found between the two groups after covariate adjustment (P = 0.194) [63]. Pan et al. demonstrated that decreased portal flow velocity and use of NSBBs were associated with the occurrence of PVT in liver cirrhosis [64]. Meanwhile, a meta-analysis included nine studies to evaluate whether NSBBs increase the risk of PVT, which found that the use of NSBBs increased a 4.62-fold risk of PVT in cirrhotic patients, but the results are limited by poor quality of included studies and significant heterogeneity across studies [65]. Therefore, there is inconclusive evidence to support the association of PVT formation with NSBBs, which requires large-scale RCTs to further confirm in the future.

Advertisement

4. Conclusion

NSBBs are the cornerstone for the management of PH in clinical practice, which has been specifically recommended for the primary and secondary prophylaxis of VB. In addition, the patient should be initiated on NSBBs promptly after the diagnosis of CSPH, regardless of the status of the varices. Considering the hemodynamic response of patients to NSBBs in cirrhosis varies based on the spectrum of cirrhosis, physicians should comprehensively assess the disease conditions of patients in clinical practice to choose the appropriate time and duration of administration with NSBBs.

Additionally, patients with ascites and refractory ascites should not be considered to be contraindications for NSBBs, which should be used under careful monitoring. Notably, if patients persistently develop hypotension and/or HRS-AKI, NSBBs should be discontinued or decreased until their blood pressure returns or HRS-AKI resolves, then restarted.

References

  1. 1. Premkumar M, Anand AC. Overview of complications in cirrhosis. Journal of Clinical and Experimental Hepatology. 2022;12(4):1150-1174
  2. 2. Iwakiri Y, Trebicka J. Portal hypertension in cirrhosis: Pathophysiological mechanisms and therapy. JHEP Reports: Innovation in Hepatology. 2021;3(4):100316
  3. 3. Mauro E, Gadano A. What's new in portal hypertension? Liver International: Official Journal of the International Association for the Study of the Liver. 2020;40(Suppl 1):122-127
  4. 4. Sauerbruch T, Schierwagen R, Trebicka J. Managing portal hypertension in patients with liver cirrhosis. F1000Research. 2 May 2018;7:F1000 Faculty Rev-533
  5. 5. Guixé-Muntet S, Quesada-Vázquez S, Gracia-Sancho J. Pathophysiology and therapeutic options for cirrhotic portal hypertension. The Lancet Gastroenterology & Hepatology. 17 Apr 2024:S2468-1253(23)00438-7
  6. 6. Shukla A, Kale A. Hyperdynamic circulation in cirrhosis: A novel gutsy central neuronal mechanism. Hepatology International. 2023;17(3):517-518
  7. 7. Rodrigues SG, Mendoza YP, Bosch J. Beta-blockers in cirrhosis: Evidence-based indications and limitations. JHEP Reports: Innovation in Hepatology. 2020;2(1):100063
  8. 8. McDonald N, Lilburn DML, Lachlan NJ, et al. Assessment of haemodynamic response to nonselective beta-blockers in portal hypertension by phase-contrast magnetic resonance angiography. BioMed Research International. 2017;2017:9281450
  9. 9. Bosch J, Iwakiri Y. The portal hypertension syndrome: Etiology, classification, relevance, and animal models. Hepatology International. 2018;12(Suppl. 1):1-10
  10. 10. Tittanegro T, China L, Forrest E, et al. Use of non-selective B-blockers is safe in hospitalised decompensated cirrhosis patients and exerts a potential anti-inflammatory effect: Data from the ATTIRE trial. EClinicalMedicine. 2023;55:101716
  11. 11. Kockerling D, Nathwani R, Forlano R, Manousou P, Mullish BH, Dhar A. Current and future pharmacological therapies for managing cirrhosis and its complications. World Journal of Gastroenterology. 2019;25(8):888-908
  12. 12. De Franchis R, Bosch J, Garcia-Tsao G, Reiberger T, Ripoll C, Baveno VIIF. Baveno VII—Renewing consensus in portal hypertension. Journal of Hepatology. 2022;76(4):959-974
  13. 13. European Association for the Study of the Liver. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. Journal of Hepatology. Aug 2018;69(2):406-460
  14. 14. Yoshiji H, Nagoshi S, Akahane T, et al. Evidence-based clinical practice guidelines for liver cirrhosis 2020. Hepatology Research. 2021;51(7):725-749
  15. 15. Cales P, Bertrais S, Boursier J, Fouchard I, Oberti F, Group S. Non-selective beta-blockers increase overall and liver mortality in alcoholic cirrhosis with MELD >/= 12 over 5 years of follow-up. Liver International: Official Journal of the International Association for the Study of the Liver. 2021;41(1):168-179
  16. 16. Tonon M, D'Ambrosio R, Calvino V, et al. A new clinical and prognostic characterization of the patterns of decompensation of cirrhosis. Journal of Hepatology. 2024;80(4):603-609
  17. 17. Krag A, Wiest R, Albillos A, Gluud LL. The window hypothesis: Haemodynamic and non-haemodynamic effects of beta-blockers improve survival of patients with cirrhosis during a window in the disease. Gut. 2012;61(7):967-969
  18. 18. Vorobioff JD, Groszmann RJ. Prevention of portal hypertension: From variceal development to clinical decompensation. Hepatology (Baltimore, Md). 2015;61(1):375-381
  19. 19. Zanetto A, Barbiero G, Battistel M, et al. Management of portal hypertension severe complications. Minerva Gastroenterol (Torino). 2021;67(1):26-37
  20. 20. 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 (London, England). 2019;393(10181):1597-1608
  21. 21. 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. Journal of Hepatology. 2022;77(4):1014-1025
  22. 22. Kumar M, Kainth S, Choudhury A, et al. Treatment with carvedilol improves survival of patients with acute-on-chronic liver failure: A randomized controlled trial. Hepatology International. 2019;13(6):800-813
  23. 23. Reiberger T, Ulbrich G, Ferlitsch A, et al. Carvedilol for primary prophylaxis of variceal bleeding in cirrhotic patients with haemodynamic non-response to propranolol. Gut. 2013;62(11):1634-1641
  24. 24. 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. Alimentary Pharmacology & Therapeutics. 2021;53(4):531-539
  25. 25. Sharma M, Singh S, Desai V, et al. Comparison of therapies for primary prevention of esophageal variceal bleeding: A systematic review and network meta-analysis. Hepatology (Baltimore, Md). 2019;69(4):1657-1675
  26. 26. Villanueva C, Albillos A, Genesca J, et al. Development of hyperdynamic circulation and response to beta-blockers in compensated cirrhosis with portal hypertension. Hepatology (Baltimore, Md). 2016;63(1):197-206
  27. 27. Bultas AC, Teshome B, Richter SK, Schafers S, Cooke E, Call WB. Use of nonselective β-blockers in patients with end-stage liver disease and select complications. The Annals of Pharmacotherapy. 2020;54(6):583-593
  28. 28. Villanueva C, Sapena V, Lo GH, et al. Improving primary prophylaxis of variceal bleeding by adapting therapy to the clinical stage of cirrhosis. A competing-risk meta-analysis of individual participant data. Alimentary Pharmacology & Therapeutics. 2024;59(3):306-321
  29. 29. Kong Y, Shi L. Comparison of the effectiveness of 11 mainstay treatments for secondary prophylaxis of variceal bleeding in patients with cirrhosis: A network meta-analysis. Experimental and Therapeutic Medicine. 2020;19(6):3479-3496
  30. 30. Shi L, Zhang X, Li J, Bai X. Favorable effects of endoscopic ligation combined with drugs on rebleeding and mortality in cirrhotic patients: A network meta-analysis. Digestive Diseases. 2018;36(2):136-149
  31. 31. Jachs M, Hartl L, Simbrunner B, et al. Carvedilol achieves higher hemodynamic response and lower rebleeding rates than propranolol in secondary prophylaxis. Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association. 2022
  32. 32. Groszmann RJ, Garcia-Tsao G, Bosch J, et al. Beta-blockers to prevent gastroesophageal varices in patients with cirrhosis. The New England Journal of Medicine. 2005;353(21):2254-2261
  33. 33. Qi XS, Bao YX, Bai M, Xu WD, Dai JN, Guo XZ. Nonselective beta-blockers in cirrhotic patients with no or small varices: A meta-analysis. World Journal of Gastroenterology. 2015;21(10):3100-3108
  34. 34. Bhardwaj A, Kedarisetty CK, Vashishtha C, et al. Carvedilol delays the progression of small oesophageal varices in patients with cirrhosis: A randomised placebo-controlled trial. Gut. 2017;66(10):1838-1843
  35. 35. Di Pascoli L, Buja A, Bolognesi M, et al. Cost-effectiveness analysis of beta-blockers vs endoscopic surveillance in patients with cirrhosis and small varices. World Journal of Gastroenterology. 2014;20(30):10464-10469
  36. 36. Serste T, Melot C, Francoz C, et al. Deleterious effects of beta-blockers on survival in patients with cirrhosis and refractory ascites. Hepatology (Baltimore, Md). 2010;52(3):1017-1022
  37. 37. Kang SH, Lee M, Kim MY, et al. The longitudinal outcomes of applying non-selective beta-blockers in portal hypertension: Real-world multicenter study. Hepatology International. 2021;15(2):424-436
  38. 38. Park JH, Jun DW, Choi J, et al. Low-dose propranolol as secondary prophylaxis for varix bleeding decreases mortality and rebleeding rate in patients with tense ascites. Journal of Clinical Medicine. 2019;8(5)
  39. 39. Almenara S, Lozano B, Gimenez P, et al. Functionality of beta-adrenergic receptors in patients with cirrhosis treated chronically with non-selective beta-blockers. Hepatology International. 2020;14(5):858-868
  40. 40. Tergast TL, Kimmann M, Laser H, et al. Systemic arterial blood pressure determines the therapeutic window of non-selective beta blockers in decompensated cirrhosis. Alimentary Pharmacology & Therapeutics. 2019;50(6):696-706
  41. 41. Leithead JA, Rajoriya N, Tehami N, et al. Non-selective β-blockers are associated with improved survival in patients with ascites listed for liver transplantation. Gut. 2015;64(7):1111-1119
  42. 42. Onali S, Kalafateli M, Majumdar A, et al. Non-selective beta-blockers are not associated with increased mortality in cirrhotic patients with ascites. Liver International: Official Journal of the International Association for the Study of the Liver. 2017;37(9):1334-1344
  43. 43. Facciorusso A, Roy S, Livadas S, et al. Nonselective beta-blockers do not affect survival in cirrhotic patients with ascites. Digestive Diseases and Sciences. 2018;63(7):1737-1746
  44. 44. Aithal GP, Palaniyappan N, China L, et al. Guidelines on the management of ascites in cirrhosis. Gut. 2021;70(1):9-29
  45. 45. Serste T, Njimi H, Degre D, et al. The use of beta-blockers is associated with the occurrence of acute kidney injury in severe alcoholic hepatitis. Liver International: Official Journal of the International Association for the Study of the Liver. 2015;35(8):1974-1982
  46. 46. 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 Transplantation. 2017;23(6):733-740
  47. 47. 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. Hepatology International. 2022;16(4):944-953
  48. 48. Xu X, Gao F, Wang T, Yang Z, Zhao Q , Qi X. Association of non-selective beta blockers with the development of renal dysfunction in liver cirrhosis: A systematic review and meta-analysis. Annals of Medicine. 2024;56(1):2305935
  49. 49. Tellez L, Albillos A. Non-selective beta-blockers in patients with ascites: The complex interplay among the liver, kidney and heart. Liver International: Official Journal of the International Association for the Study of the Liver. 2022;42(4):749-761
  50. 50. Bang UC, Benfield T, Hyldstrup L, Jensen JE, Bendtsen F. Effect of propranolol on survival in patients with decompensated cirrhosis: A nationwide study based Danish patient registers. Liver International: Official Journal of the International Association for the Study of the Liver. 2016;36(9):1304-1312
  51. 51. Scheiner B, Parada-Rodriguez D, Bucsics T, et al. Non-selective beta-blocker treatment does not impact on kidney function in cirrhotic patients with varices. Scandinavian Journal of Gastroenterology. 2017;52(9):1008-1015
  52. 52. Li TH, Liu CW, Huang CC, et al. Non-selective beta-blockers decrease infection, acute kidney injury episodes, and ameliorate sarcopenic changes in patients with cirrhosis: A propensity-score matching tertiary-center cohort study. Journal of Clinical Medicine. 2021;10(11)
  53. 53. Maiwall R, Pasupuleti SSR, Jain P, Sarin SK. Degree of portal and systemic hemodynamic alterations predict recurrent AKI and chronic kidney disease in patients with cirrhosis. Hepatology Communications. 2021;5(2):293-308
  54. 54. Senzolo M, Cholongitas E, Burra P, et al. Beta-blockers protect against spontaneous bacterial peritonitis in cirrhotic patients: A meta-analysis. Liver International: Official Journal of the International Association for the Study of the Liver. 2009;29(8):1189-1193
  55. 55. 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. Journal of Hepatology. 2016;64(3):574-582
  56. 56. Abdel-Razik A, Abdelsalam M, Gad DF, Abdelwahab A, Tawfik M, Elzehery R, et al. Recurrence of spontaneous bacterial peritonitis in cirrhosis: Novel predictors. European Journal of Gastroenterology & Hepatology. Jun 2020;32(6):718-726
  57. 57. 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(7):1680-1690 e1681
  58. 58. Senzolo M, Garcia-Tsao G, Garcia-Pagan JC. Current knowledge and management of portal vein thrombosis in cirrhosis. Journal of Hepatology. 2021;75(2):442-453
  59. 59. Harding DJ, Perera MT, Chen F, Olliff S, Tripathi D. Portal vein thrombosis in cirrhosis: Controversies and latest developments. World Journal of Gastroenterology. 2015;21(22):6769-6784
  60. 60. Qi X, Bai M, Yang Z, et al. Occlusive portal vein thrombosis as a new marker of decompensated cirrhosis. Medical Hypotheses. 2011;76(4):522-526
  61. 61. Zhang Y, Xu BY, Wang XB, et al. Prevalence and clinical significance of portal vein thrombosis in patients with cirrhosis and acute decompensation. Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association. 2020;18(11):2564-2572 e2561
  62. 62. Qi X, Su C, Ren W, et al. Association between portal vein thrombosis and risk of bleeding in liver cirrhosis: A systematic review of the literature. Clinics and Research in Hepatology and Gastroenterology. 2015;39(6):683-691
  63. 63. Xu X, Xu S, Primignani M, et al. Nonselective beta-blockers may progress the thrombosis of portal venous system in cirrhotic patients: A retrospective observational study. Advances in Therapy. 2020;37(4):1452-1463
  64. 64. Pan J, Wang L, Gao F, et al. Epidemiology of portal vein thrombosis in liver cirrhosis: A systematic review and meta-analysis. European Journal of Internal Medicine. 2022;104:21-32
  65. 65. Xu X, Guo X, De Stefano V, et al. Nonselective beta-blockers and development of portal vein thrombosis in liver cirrhosis: A systematic review and meta-analysis. Hepatology International. 2019;13(4):468-481

Written By

Ting Wang, Cai’e Wang, Hongyu Li and Ran Wang

Submitted: 21 May 2024 Reviewed: 22 May 2024 Published: 17 June 2024