Next Article in Journal
Tumor Microenvironment
Previous Article in Journal
Possible Interaction between Dabigatran and Ranolazine in Patients with Renal Failure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Role of Non-Selective β-Blockers in Compensated Cirrhotic Patients without Major Complications

1
Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83330, Taiwan
2
Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83330, Taiwan
3
Department of Traditional Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
4
Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
5
School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
6
Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83330, Taiwan
7
School of Pharmacy, Kaohsiung Medical University, Kaohsiung 80700, Taiwan
8
Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83330, Taiwan
*
Author to whom correspondence should be addressed.
Medicina 2020, 56(1), 14; https://doi.org/10.3390/medicina56010014
Submission received: 21 November 2019 / Revised: 22 December 2019 / Accepted: 22 December 2019 / Published: 30 December 2019

Abstract

:
Background and Objectives: Non-selective β-blockers (NSBB) could prevent decompensation and hepatocellular carcinoma (HCC) in cirrhotic patients with clinically significant portal hypertension (CSPH), but remained uncertain for compensated cirrhotic patients without major complications. We aimed to compare the clinical outcomes between propranolol users and non-users of a CC group without major complications. Material and Methods: We conducted this population-based cohort study by using the Taiwanese Longitudinal Health Insurance Database 2000. Propranolol users (classified as cumulative defined daily dose (cDDD)) and non-PPL users were matched with a 1:1 propensity score in both cohorts. Results: This study comprised 6896 propranolol users and 6896 non-propranolol users. There was no significant impact on the development of spontaneous bacterial peritonitis between the two groups (aHR: 1.24, 95% confidence interval (CI): 0.88~1.75; p = 0.2111). Male gender, aged condition, and non-liver related diseases (peripheral vascular disease, cerebrovascular disease, dementia, pulmonary disease, and renal disease) were the independent risk factors of mortality. PPL users had significantly lower incidence of HCC development than non-users (aHR: 0.81, p = 0.0580; aHR: 0.80, p = 0.1588; and aHR: 0.49, p < 0.0001 in the groups of 1–28, 29–90, and >90 cDDD, respectively). Conclusion: The current study suggested that high cumulative doses of propranolol could decrease the risk of hepatocellular carcinoma among compensated cirrhotic patients without major complications. Further large-scale prospective studies are still required to confirm the findings in this study. Results: It remained uncertain whether non-selective β-blockers (NSBB) could prevent decompensation and hepatocellular carcinoma (HCC) in compensatory cirrhotic patients without major complications. This study aimed to compare the clinical outcomes between propranolol users and non-users of the CC group without major complications.

1. Introduction

Portal hypertension (PHT) is the driving force of clinical progression in patients with liver cirrhosis. Non-selective β-blockers (NSBB), available as propranolol in Taiwan, can effectively reduce PHT by the mechanism of reducing the splanchnic blood flow and lowering the cardiac output [1]. Current practice guidelines recommend the use of NSBB as a primary and secondary prophylaxis strategy for cirrhotic patients with presence of esophageal varices (EV) [2,3]. Villanueva C, et al. [4,5] reported that cirrhotic patients with the development of clinically significant portal hypertension (CSPH) had a greater hepatic vein pressure gradient (HVPG) reduction after NSBB treatment than those without CSPH. NSBB could prevent decompensation in cirrhotic patients with CSPH. Apart from this, many studies focused on the use of NSBB in a decompensated group. Some studies reported that NSBB was not associated with increased mortality among decompensated cirrhotic patients with ascites [6,7,8,9], whereas Kalambokis GN, et al. [10] found that an increased mortality was observed in Child-Pugh C cirrhotic patients with ascites if using NSBB for more than six months. Moreover, NSBB could reduce cancer risk [11,12], including hepatocellular carcinoma (HCC) [13,14]. The issue about the use of NSBB on the prognosis in compensated cirrhotic patients without major complications has seldom been reported.
Therefore, we conducted a large population-based cohort study in a national health care setting in an attempt to clarify the clinical impacts of NSBB on cirrhotic patients without major complications.

2. Methods

2.1. Compliance with Ethical Requirements

The study protocol was approved by the Institutional Review Board and the Ethics Committee of Chang Gung Memorial Hospital at Taoyan in Taiwan (permitted number 201800318B0C503 on 1st April 2019). The Ethics Committee waived the requirement for informed consent for this study, and the data were analyzed anonymously.

2.2. Data Sources

The present study analyzed data extracted from the Longitudinal Health Insurance Database 2000 (LHID 2000) of one million individuals (approximately 5% of the entire Taiwan population) who were randomly sampled from the year 2000 Registry for Beneficences of 23.75 million individuals involved in Taiwan’s National Health Insurance (NHI) program [15]. Taiwan’s National Health Insurance program was initiated in 1995, and covers over 99% of Taiwan’s 23 million individuals. LHID 2000 contains the demographic information, diagnostics, medical treatments, prescriptions, and total costs from 1 January 1997 to 31 December 2013.

2.3. Study Cohort, and Inclusion and Exclusion Criteria

Figure 1 shows a schematic flowchart of the study design. The cohort of patients with liver cirrhosis was identified using ICD-9 CM (International Classification of Diseases, Ninth Revision, Clinical Modification, codes: 571.2, 571.5, or 571.6, based on ≥1 claim of inpatients or ≥2 claims of outpatients in one year and apart ≥28 days) between 1997 and 2013. Patients with ≥18 years old were enrolled in the study. The etiology of cirrhosis was collected, namely: chronic hepatitis B virus (ICD-9 CM codes: 070.2, 070.22, 070.23, 070.3, 070.32, 070.33, and V02.61), chronic hepatitis C virus (ICD-9 CM codes: 070.51, 070.54, and V02.62), and alcohol-related disease (ICD-9 CM codes: 291, 303.0, 303.9, 305.0, 571.0, 571.2, and 571.3). In the present study, we focused on only cirrhotic patients without major complications, and with a later stage of chronic liver disease with the development of portal hypertension. Those who had a diagnosis of spontaneous bacterial peritonitis (SBP; ICD-9-CM code: 567), variceal bleeding (ICD-9-CM codes: 456.8, 456.0, and 456.20), ascites (ICD-9-CM code: 7895, or with frequent abdominal tapping, 54.91, based on ≥3 claims of inpatients in one year or ≥1 claims of admission), jaundice (ICD-9-CM code: 7824), hepato-renal syndrome (ICD-9-CM code: 5724), hepatic coma (ICD-9-CM code: 5722), and other sequelae of chronic liver disease (ICD-9-CM code: 572.8) were defined as having a decompensated status and were excluded from the analyses. Accordingly, patients with cirrhosis without any of the above conditions were considered to have liver cirrhosis without major complications. Those who were <18 years old, had a history of Human Immunodeficiency Virus (HIV) co-infection (ICD-9-CM codes: 042, 044, and V08), hepatocellular carcinoma (ICD-9-CM code: 155), malignancy other than HCC (ICD-9-CM codes: 140–208, with major illness certificate), loss of medical record for one year, or prior use of PPL before enrollment were also excluded. After propensity score matching, 6896 patients with propranolol exposure and 6896 patients without exposure were analyzed.

2.4. Definition of PPL Exposure

We defined the exposure of propranolol (ATC code: C07AA05) from the diagnosis of liver cirrhosis to the occurrence of outcomes or to the end of follow-up. To further recognize the possible influence of the dose effect, we adopted the concept of quantifying a prescribed dose of medication, anticipating the average prescription dose per day in adult population, which was known as a cumulative defined daily dose (cDDD) [16]. We classified the propranolol dose into four sets in each group (0, 28, 29–89, and >90 cDDDs). Patients were considered as not taking any propranolol if the cDDD was zero. Moreover, some concomitant drugs with potential confounding effects, including selective β-blockers (ATC code: C07AB), diuretics (ATC code: C03), thiazides (ATC code: C03A-C) potassium-sparing drugs (ATC code: C03D), and diuretics and potassium-sparing agents in combination (ATC code: C03E) were identified from the index date to the event of interest or to the end of follow-up.

2.5. Study Outcomes

The primary outcome was the development of hepatocellular carcinoma. All of the patients were followed from the index date to the event of interest, or to the end of the three-year follow-up. The secondary outcome was spontaneous bacterial peritonitis, all-cause mortality, liver transplantation (ICD9 code: V42.7), or any diagnosis indicative of decompensation, which is defined as above.

2.6. Confounder Assessment

The patients’ underlying comorbid conditions were identified within one year prior to the index date. The burden of comorbid illness was assessed based on the Deyo modification of the Charlson comorbidity index (CCI), which has been shown to be a well-validated measure of comorbidity, adjusting for disease burden in the administrative data [17]. To better understand the comorbid illness on the confounding impact on the development of HCC, the burden of comorbid illness was further separately discussed.
Other potential risk factors, including the liver disease treatment (statins (atorvastatin ATC codes: C10AA05, C10BX08, C10BX03, C10BA05, C10BX12, C10BX06, and C10BX11; fluvastatin ATC code: C10AA04; pitavastatin ATC code: C10AA08; rosuvastatin ATC codes: C10AA07, C10BX05, 10BX09, C10BA06, C10BX10, and C10BX07; simvastatin ATC codes: C10AA01, C10BX01, C10BA02, C10BA04, C10BX04, and A10BH51) [18,19], lipid-lowering agents (clofibrate ATC code: C10AB01; bezafibrate ATC code: C10AB02; gemfibrozil ATC code: C10AB04; fenofibrate ATC code: C10AB05; nicotinic acid ATC code: C04AC01; and acipimox ATC code: C10AD06), angiotensin converting-enzyme inhibitor (captopril ATC code: C09AA01; lisinopril ATC code: C09AA03; perindopril ATC code: C09AA04; ramipril ATC code: C09AA05; quinapril ATC code: C09AA06; benazepril ATC code: C09AA07; cilazapril ATC code: C09AA08; and fosinopril ATC code: C09AA09), aspirin (ATC code: B01AC06), metformin (ATC code: A10BA02), diuretics (furosemide ATC code: C03CA01, C02LA01; spironolactone ATC code: C03DA01), and selective β-blockers (ATC code: C07AB)), were assessed in the study. Only patients with drug use for at least 28 days within one year prior to the index hospitalization, or use for at least 28 days during the study follow-up were analyzed.

2.7. Statistical Analysis

We performed a matched case control study by using propensity score adjustment with immortal time, sex, age group, CCI, HTN, and prior medication (interferon, metformin, aspirin, angiotensin converting-enzyme inhibitor, lipid lowering drugs, statins, diuretics, and selective beta blockers). Continuous data were presented as means ± standard deviation (SD), and categorical data were presented as frequencies and percentages. Pearson’s chi-square or Fisher’s exact two-tailed tests were used for the analysis of categorical data, while continuous variables were analyzed using the t-test, where appropriate.
To assess the impact of the competing risk of death during the index hospitalization on the outcome prediction, the standard and cause-specific approach of the Cox proportional hazard model was employed to estimate the relative hazard ratio of an outcome event. Adjustments of patient demographics, clinical conditions, and medication usage were made in the regression model. The Kaplan–Meier method with the log-rank test was employed to compare the cumulative incidence between comparison groups. Two-tailed p-values of <0.05 were considered statistically significant. All of the statistical analyses were conducted using SAS version 9.4 (SAS Institute’s Inc., Cary, NC, USA, 2013).

3. Results

3.1. Patient Characteristics

The baseline characteristics of the parameters before matching are presented in Supplementary Table S1. Table 1 shows the demographic data for the two groups after propensity score adjustment, which included the etiologies of liver cirrhosis, such as hepatitis B virus infection, hepatitis C virus infection, and alcoholism.

3.2. Outcomes of Spontaneous Bacterial Peritonitis and Assessments of Risk Factors

As shown in Table 2, patients with propranolol exposure had significantly higher rates of spontaneous bacterial peritonitis (1.42% vs. 0.75%, p = 0.0002) during the three-year follow-up period (log-rank p = 0.0177, shown in Figure 2) and for ascites (4.32% vs. 0.64%, p < 0.0001). No significant difference was observed with respect to hepatorenal syndrome (0.26% vs. 0.15%, p = 0.1302).
As shown in Table 3, male gender (aHR: 1.90, 95% CI: 1.30~5.76, p = 0.0008), aged ≥65 (aHR: 2.02, 95% CI: 1.08~3.75, p = 0.0270), and concomitant diuretic usage (aHR: 8.56, 95% CI: 5.93~12.37, p < 0.0001) were independent risk factors for the development of SBP on the multivariate analysis. On the contrary, the concomitant use of selective β-blockers was a protective factor from SBP (aHR: 0.35, 95% CI: 0.19~0.67, p = 0.0014). Notably, there was no significant impact on the development of SBP about the exposure of propranolol or not (aHR: 1.24, 95% CI: 0.88~1.75; p = 0.2111).

3.3. All-Cause Mortality

During the three-year follow-up period, cirrhotic patients without major complications, who had PPL exposure for more than 90 days, had a significantly lower rate of all-cause mortality than those without exposure. Notably, those who had exposure between 1~28 days and 29~90 days had a higher all-cause mortality rate (Figure 3).
As shown in Table 4, on the multivariate analysis, male gender (aHR: 1.59, 95% CI: 1.38~1.83, p < 0.0001), an aged condition (35–49 aHR: 1.73, 95% CI: 1.34~2.21; 50–64 aHR: 2.63, 95% CI: 2.04~3.40; ≥65 aHR: 5.07, 95% CI: 3.89~6.61, p < 0.0001), peripheral vascular disease (aHR: 3.15, 95% CI: 1.67~5.93, p = 0.0004), cerebrovascular disease (aHR: 1.56, 95% CI: 1.20~2.01, p = 0.0008), dementia (aHR: 1.86, 95% CI: 1.04~3.32, p = 0.0350), pulmonary disease (aHR: 1.25, 95% CI: 1.03~1.52, p = 0.0218), peptic ulcer (aHR: 1.29, 95% CI: 1.11~1.50, p = 0.0009), renal disease (aHR: 1.87, 95% CI: 1.42~2.48, p < 0.0001), baseline (aHR: 1.88, 95% CI: 1.45~2.44, p < 0.0001), and concomitant diuretics (aHR: 2.69, 95% CI: 2.33~3.11, p < 0.0001) were independent risk factors of all-cause mortality. Baseline Angiotensin-converting enzyme inhibitor ACEI (aHR: 0.71, 95% CI: 0.52~0.97, p = 0.0307) and concomitant selective beta blockers (aHR: 0.44, 95% CI: 0.35~0.56, p < 0.0001) were protective factors. As for propranolol, a trend from a harmful effect if cDDD was <90 days (1~28 cDDD, aHR: 1.33, 95% CI: 1.15~1.53, p = 0.0001; 29~90 cDDD, aHR: 1.12, 95% CI: 0.91~1.40, p = 0.2888), to a protective effect if cDDD was more than 90 days (aHR: 0.79, 95% CI: 0.64~0.98, p = 0.0340) was observed.

3.4. Outcomes of Hepatocellular Carcinoma and Assessments of Risk Factors

In the multivariate analysis, the independent risk factors of HCC development were gender (aHR: 1.47, 95% CI: 1.20~1.79, p = 0.0002), aging (35–49 aHR: 3.33, 95% CI: 2.02~5.49; 50–64 aHR: 7.67, 95% CI: 4.68~12.55; ≥65 aHR: 10.72, 95% CI: 6.43~17.88, p < 0.0001), and concomitant use of diuretics (aHR: 5.65, 95% CI: 4.61~6.92, p < 0.0001). The concomitant use of selective beta blockers was a protective factor (aHR: 0.54, 95% CI: 0.39~0.74, p = 0.0001).
As for propranolol, the cirrhotic patients without major complications had an increased protective effect free from the development of HCC when they had an increased cDDD of propranolol exposure (Figure 4 and Table 5). The protective effect was even more significant among those with propranolol exposure for more than 90 days when compared with those without propranolol exposure (aHR: 0.49, 95% CI: 0.36~0.67, p < 0.0001).

4. Discussions

Clinically significant portal hypertension was defined as a hepatic venous pressure gradient (HVPG) of ≥10 mmHg. Compensated cirrhotic patients with CSPH may present with the appearance of varices and a risk of decompensation [20,21]. Owning to the development of hyperdynamic circulation in cirrhotic patients with CSPH, NSBB could effectively reduce HVPG, and hence improve the clinical outcomes, including bleeding and mortality [22]. On the contrary, compensated cirrhotic patients without CSPH were not suggested the use of NSBB because of the lack of development of hyperdynamic circulation. This current study focused on patients with a diagnosis of cirrhosis, and excluded any diagnosis of decompensation, which meant that we enrolled cirrhotic patients without CSPH clinically. We found that patients with PPL exposure had significantly higher rates of decompensation events than those without PPL (Table 2). It did not mean that PPL caused worse outcomes among cirrhotic patients without CSPH. As shown in Table 1, patients with propranolol exposure still had more comorbidity than those without exposure, even after propensity score matching adjustment. The bottom line was that, in real world practice, the clinical physicians prescribed propranolol to patients with a more serious clinical condition, including a higher trend toward CSPH and decompensations events. The use of NSBB was not helpful in preventing further progress of decompensation among cirrhotic patients without CSPH (Supplementary Figure S1 and Supplementary Table S2).
Most cirrhotic patients suffered from death after the development of decompensation. Many studies focused on the discussion of the safety concerns of NSBB use among decompensated cirrhotic patients, especially with presence of ascites, but they have not been conclusive to date [6,7,8,9,10]. The NSBB might not affect the mortality, but should be used cautiously, especially among enrolled patients with a history of spontaneous bacterial peritonitis [23]. On the contrary, non-liver-related diseases were predominant causes of mortality among compensated cirrhotic patients [24,25]. Similarly, the current study showed that host factors (male gender and aged condition) and comorbidity (peripheral vascular disease, cerebrovascular disease, dementia, pulmonary disease, and renal disease) were the independent risk factors of mortality. Moreover, a protective effect from mortality was observed among patients with NSBB exposure for more than 90 days cDDD (aHR: 0.79, 95% CI: 0.64~0.98, p = 0.0340). Brito-Azevedo A, et al. reported that improving endothelial function was detected among compensated cirrhotic patients receiving PPL compared with those without propranolol use (propranolol users, n = 6, 567 ± 377% vs. non-propranolol users, n = 14, 490 ± 188%; p = 0.01) [26]. Moreover, propranolol could reduce inflammation by decreasing intestinal permeability, bacterial translocation, and serum levels of IL-6 [27,28], and therefore might explain the dose-dependent effect of PPL on the impact of mortality.
Ripoll C, et al. reported that cirrhotic patients with CSPH had a higher annual incidence of HCC than those without CSPH (2.1% vs. 0.35%) [29]. Furthermore, propranolol had anti-cancer effects and could block the β-2 adrenergic receptor (ADRB2), whose expression was upregulated in HCC [30]. A meta-analysis by Thiele M, et al. reported that NSBB may prevent HCC in patients with cirrhosis [14]. However, those trials enrolled in this meta-analysis were not targeted for the survey of NSBB and HCC. Herrera l, et al. performed a retrospective cohort study including 73 patients treated with NSBB and 100 patients without NSBB use, and found that a lower cumulative incidence of HCC during five or ten years of follow-up was observed among propranolol users. It was reported that increasing the protective effect from the development of HCC was observed among patients with propranolol users (1~28 cDDD aHR: 0.81→29~90 cDDD aHR: 0.80→>90 cDDD aHR: 0.49, as shown in Table 5) compared with those without exposure. The strength of this study was that it was a large sample size population-based study. To our knowledge, it is the first study to demonstrate the quantification of PPL exposure to the association of HCC among cirrhotic patients without CSPH.
Increased vascular resistance was the predominant mechanism of PHT in the early phase of cirrhosis. Carvedilol could decrease the vascular resistance by its intrinsic vasodilator activity, and was reported to be more effective in reducing PHT than propranolol [31,32]. Current practice guidelines recommend carvedilol as the choice of primary prevention for cirrhotic patients with esophageal varices [2]. In this study, we found that patients with a concomitant use of selective BB had beneficial effects for the prevention of SBP, better survival rates, and lower rates of HCC development. For patients with intolerances or non-responders to NSBB, carvedilol might be a promising therapy for compensated cirrhotic patients without CSPH.
This study has several limitations. First, to select compensated cirrhotic patients without CSPH, we performed rigorous exclusions of the diagnosis supportive of decompensation clinically. We could only define the study cohort as a majority of patients with compensated cirrhosis without major complications, but not as compensated patients without CSPH. Non-invasive exams highly suggestive of CSPH, including liver stiffness (≥20–25 kPa by transient elastography) [33] or the detection of porto-systemic shunting or varices [3,33] should be collected in future studies. Second, we could not accurately classify the patients clinically because of the lack of hemodynamics and laboratory data. Propensity score matching adjustment was performed for the correction of potential confounding factors. Third, the actual dose of PPL and the duration were not available from the NHIRD. In this current study, a positive dose-response effect of PPL on the prevention of HCC by using the concept of cDDD was observed. Based on this important finding, a prospective study focused on the dose and the duration of PPL use among compensated cirrhotic patients without CSPH should be further clarified.
In conclusion, the current study suggested that high cumulative doses of propranolol could decreased the risk of hepatocellular carcinoma among compensated cirrhotic patients without major complications. Further large-scale prospective studies are still required to confirm the finding in this study.

Supplementary Materials

The following are available online at https://www.mdpi.com/1648-9144/56/1/14/s1. Table S1: Patient’s characteristics before propensity score matching. Table S2: Factors associated with decompensation. Figure S1: Cumulative incidence of decompensation between groups.

Author Contributions

Conception and design, C.-N.H., S.-K.C., and C.-K.W.; manuscript writing, W.-S.Y. and C.-K.W.; collection and assembly of data: C.-K.W., S.-C.Y., C.-M.L., Y.-C.L., W.-S.Y., W.-C.T., C.-H.L., Y.-H.Y., C.-N.H., T.-H.T., and S.-K.C.; data analysis and interpretation, C.-N.H., S.-K.C., and C.-K.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Kaohsiung Chang Gung Memorial Hospital (CFRPG8H0081) at Kaohsiung, and Chia-Yi Chang Gung Memorial Hospital (CLRPG6G0081) at Chia-Yi in Taiwan.

Acknowledgments

The authors appreciate Yi-Hsuan Tsai in the Division of Gastroenterology, Kaohsiung Chang Gung Memorial Hospital, for assistance with the programming and analyses. The authors would like to thank the Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chia-Yi Branch.

Conflicts of Interest

The authors declared no conflicts of interest.

References

  1. Lebrec, D.; Corbic, M.; Nouel, O.; Benhamou, J.P. PPL—A medical treatment for portal hypertension? Lancet 1980, 2, 180–182. [Google Scholar] [CrossRef]
  2. Garcia-Tsao, G.; Abraldes, J.G.; Berzigotti, A.; Bosch, J. Portal hypertensive bleeding in cirrhosis: Risk stratification, diagnosis, and management: 2016 practice guidance by the American Association for the study of liver diseases. Hepatology 2017, 65, 310–335. [Google Scholar] [CrossRef] [Green Version]
  3. de Franchis, R.; Baveno, V.I. Faculty. Expanding consensus in portal hypertension: Report of the Baveno VI Consensus Workshop: Stratifying risk and individualizing care for portal hypertension. J. Hepatol. 2015, 63, 743–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Villanueva, C.; Albillos, A.; Genescà, J.; Abraldes, J.G.; Calleja, J.L.; Aracil, C.; Bañares, R.; Morillas, R.; Poca, M.; Peñas, B.; et al. Development of hyperdynamic circulation and response to β-blockers in compensated cirrhosis with portal hypertension. Hepatology 2016, 63, 197–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Villanueva, C.; Albillos, A.; Genescà, J.; Garcia-Pagan, J.C.; Calleja, J.L.; Aracil, C.; Bañares, R.; Morillas, R.M.; Poca, M.; Peñas, B.; 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. [Google Scholar] [CrossRef]
  6. Chirapongsathorn, S.; Valentin, N.; Alahdab, F.; Krittanawong, C.; Erwin, P.J.; Murad, M.H.; Kamath, P.S. β-Blockers and Survival in Patients with Cirrhosis and Ascites: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2016, 14, 1096–1104. [Google Scholar] [CrossRef] [Green Version]
  7. Bang, U.C.; Benfield, T.; Hyldstrup, L.; Jensen, J.E.; Bendtsen, F. Effect of PPL on survival in patients with decompensated cirrhosis: A nationwide study based Danish patient registers. Liver Int. 2016, 36, 1304–1312. [Google Scholar] [CrossRef]
  8. Onali, S.; Kalafateli, M.; Majumdar, A.; Westbrook, R.; O’Beirne, J.; Leandro, G.; Patch, D.; Tsochatzis, E.A. Non-selective beta-blockers are not associated with increased mortality in cirrhotic patients with ascites. Liver Int. 2017, 37, 1334–1344. [Google Scholar] [CrossRef] [Green Version]
  9. Bossen, L.; Krag, A.; Vilstrup, H.; Watson, H.; Jepsen, P. Nonselective β-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. [Google Scholar] [CrossRef]
  10. Kalambokis, G.N.; Christodoulou, D.; Baltayiannis, G.; Christou, L. PPL use beyond 6 months increases mortality in patients with Child-Pugh C cirrhosis and ascites. Hepatology 2016, 64, 1806–1808. [Google Scholar] [CrossRef]
  11. Chang, P.Y.; Huang, W.Y.; Lin, C.L.; Huang, T.C.; Wu, Y.Y.; Chen, J.H.; Kao, C.H. PPL Reduces Cancer Risk: A Population-Based Cohort Study. Medicine 2015, 94, 1097. [Google Scholar] [CrossRef]
  12. Monami, M.; Filippi, L.; Ungar, A.; Sgrilli, F.; Antenore, A.; Dicembrini, I.; Bagnoli, P.; Marchionni, N.; Rotella, C.M.; Mannucci, E. Further data on beta-blockers and cancer risk: Observational study and meta-analysis of randomized clinical trials. Curr. Med. Res. Opin. 2013, 29, 369–378. [Google Scholar] [CrossRef]
  13. Herrera, I.; Pascual, S.; Zapater, P.; Carnicer, F.; Bellot, P.; María Palazón, J. The use of β-blockers is associated with a lower risk of developing hepatocellular carcinoma in patients with cirrhosis. Eur. J. Gastroenterol. Hepatol. 2016, 28, 1194–1197. [Google Scholar] [CrossRef]
  14. Thiele, M.; Albillos, A.; Abazi, R.; Wiest, R.; Gluud, L.L.; Krag, A. Non-selective beta-blockers may reduce risk of hepatocellular carcinoma: A meta-analysis of randomized trials. Liver Int. Off. J. Int. Assoc. Study Liver 2015, 35, 2009–2016. [Google Scholar] [CrossRef]
  15. Longitudinal Health Insurance Database 2000 (LHID 2000), National Health Insurance Research Database. Taiwan at. Available online: https://nhird.nhri.org.tw/en/Data_Subsets.html (accessed on 1 May 2019).
  16. WHO Collaborating Center for Drugs Statistics Methodology: ATC/DDD Index 2013. Available online: http://www.whocc.no/atc_ddd_index/ (accessed on 1 May 2019).
  17. Deyo, R.A.; Cherkin, D.C.; Ciol, M.A. Adapting a clinical comorbidity index for use with ICD-9-CM administrative databases. J. Clin. Epidemiol. 1992, 45, 613–619. [Google Scholar] [CrossRef]
  18. Abraldes, J.G.; Villanueva, C.; Aracil, C.; Turnes, J.; Hernandez-Guerra, M.; Genesca, J.; García-Pagán, J.C.; Torres, F.; Calleja, J.L.; Albillos, A. Addition of Simvastatin to Standard Therapy for the Prevention of Variceal Rebleeding Does Not Reduce Rebleeding but Increases Survival in Patients with Cirrhosis. Gastroenterology 2016, 150, 1160–1170. [Google Scholar] [CrossRef] [Green Version]
  19. Kim, R.G.; Loomba, R.; Prokop, L.J.; Singh, S. Statin Use and Risk of Cirrhosis and Related Complications in Patients with Chronic Liver Diseases: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2017, 15, 1521–1530. [Google Scholar] [CrossRef] [Green Version]
  20. Garcia-Tsao, G.; Friedman, S.; Iredale, J.; Pinzani, M. Now there are many (stages) where before there was one: In search of pathophysiological classification of cirrhosis. Hepatology 2010, 51, 1445–1449. [Google Scholar] [CrossRef] [Green Version]
  21. Ripoll, C.; Groszmann, R.; Garcia-Tsao, G.; Grace, N.; Burroughs, A.; Planas, R.; Escorsell, A.; García-Pagán, J.C.; Makuch, R.; Patch, D. Hepatic venous pressure gradient predicts clinical decompensation in patients with compensated cirrhosis. Gastroenterology 2007, 133, 481–488. [Google Scholar] [CrossRef]
  22. Poynard, T.; Cale’s, P.; Pasta, L.; Ideo, G.; Pascal, J.P.; Pagliaro, L.; Lebrec, D. Franco—Italian Multicenter Study Group. Betaadrenergic-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. N. Engl. J. Med. 1991, 324, 1532–1538. [Google Scholar]
  23. Mandorfer, M.; Bota, S.; Schwabl, P.; Bucsics, T.; Pfisterer, N.; Kruzik, M.; Hagmann, M.; Blacky, A.; Ferlitsch, A.; Sieghart, W.; et al. Nonselective β blockers increase risk for hepatorenal syndrome and death in patients with cirrhosis and spontaneous bacterial peritonitis. Gastroenterology 2014, 146, 1680–9160. [Google Scholar] [CrossRef] [Green Version]
  24. Groszmann, R.J.; Garcia-Tsao, G.; Bosch, J.; Grace, N.D.; Burroughs, A.K.; Planas, R.; Escorsell, A.; Garcia-Pagan, J.C.; Patch, D.; Matloff, D.S.; et al. Beta-blockers to prevent gastroesophageal varices in patients with cirrhosis. N. Engl. J. Med. 2005, 353, 2254–2261. [Google Scholar] [CrossRef] [Green Version]
  25. Abraldes, J.G.; Iwakiri, Y.; Loureiro-Silva, M.; Haq, O.; Sessa, W.C.; Groszmann, R.J. Mild increases in portal pressure up-regulate VEGF and eNOS in the intestinal microcirculation leading to hyperdynamic state. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 290, 980–987. [Google Scholar] [CrossRef] [Green Version]
  26. Brito-Azevedo, A.; de Mello Perez, R.; Coelho, H.S.M.; Fernandes, E.D.S.M.; Castiglione, R.C.; Villela-Nogueira, C.A.; Bouskela, E. PPL improves endothelial dysfunction in advanced cirrhosis: The ‘endothelial exhaustion’ hypothesis. Gut 2016, 65, 1391–1392. [Google Scholar] [CrossRef]
  27. Reiberger, T.; Ferlitsch, A.; Payer, B.A.; Mandorfer, M.; Heinisch, B.B.; Hayden, H.; Lammert, F.; Trauner, M.; Peck-Radosavljevic, M.; Vogelsang, H.; et al. Non-selective betablocker therapy decreases intestinal permeability and serum levels of LBP and IL-6 in patients with cirrhosis. J. Hepatol. 2013, 58, 911–921. [Google Scholar] [CrossRef]
  28. Brito-Azevedo, A.; de Mello Perez, R.; Coelho, H.S.M.; Fernandes, E.D.S.M.; Castiglione, R.C.; Villela-Nogueira, C.A.; Bouskela, E. The anti-inflammatory role of PPL in cirrhosis: Preventing the inflammatory exhaustion? J. Hepatol. 2017, 66, 240–241. [Google Scholar] [CrossRef] [Green Version]
  29. Ripoll, C.; Groszmann, R.J.; Garcia-Tsao, G.; Bosch, J.; Grace, N.; Burroughs, A.; Planas, R.; Escorsell, A.; Garcia-Pagan, J.C.; Makuch, R.; et al. Hepatic venous pressure gradient predicts development of hepatocellular carcinoma independently of severy of cirrhosis. J. Hepatol. 2009, 50, 923–928. [Google Scholar] [CrossRef] [Green Version]
  30. Kassahun, W.T.; Guenl, B.; Ungemach, F.R.; Jonas, S.; Abraham, G. Expression and functional coupling of liver beta2—Adrenoceptors in the human hepatocellular carcinoma. Pharmacology 2012, 89, 313–320. [Google Scholar] [CrossRef]
  31. Sinagra, E.; Perricone, G.; D’Amico, M.; Tinè, F.; D’Amico, G. Systematic review with meta-analysis: The haemodynamic effects of carvedilol compared with propranolol for portal hypertension in cirrhosis. Aliment. Pharmacol. Ther. 2014, 39, 557–568. [Google Scholar] [CrossRef]
  32. Gupta, V.; Rawat, R.; Shalimar Saraya, A. Carvedilol versus propranolol effect on hepatic venous pressure gradient at 1 month in patients with index variceal bleed: RCT. Hepatol. Int. 2017, 11, 181–187. [Google Scholar] [CrossRef]
  33. Abraldes, J.G.; Bureau, C.; Stefanescu, H.; Augustin, S.; Ney, M.; Blasco, H.; Procopet, B.; Bosch, J.; Genesca, J.; Berzigotti, A. Noninvasive tools and risk of clinically significant portal hypertension and varices in compensated cirrhosis: The “Anticipate” study. Hepatology 2016, 64, 2173–2184. [Google Scholar] [CrossRef]
Figure 1. Schematic flowchart of the study design.
Figure 1. Schematic flowchart of the study design.
Medicina 56 00014 g001
Figure 2. Cumulative incidence of spontaneous bacterial peritonitis between the groups.
Figure 2. Cumulative incidence of spontaneous bacterial peritonitis between the groups.
Medicina 56 00014 g002
Figure 3. Cumulative incidence of all-cause mortality between the groups.
Figure 3. Cumulative incidence of all-cause mortality between the groups.
Medicina 56 00014 g003
Figure 4. Cumulative incidence of hepatocellular carcinoma between the groups.
Figure 4. Cumulative incidence of hepatocellular carcinoma between the groups.
Medicina 56 00014 g004
Table 1. Patient’s characteristics after propensity score matching adjustment.
Table 1. Patient’s characteristics after propensity score matching adjustment.
VariableExposureNon-Exposure
Nn(%)n(%)SMDp-Value
Total13,7926896(50.00)6896(50.00)
Sex0.8196
Female52372625(38.07)2612(37.88)0.00
Male85554271(61.93)4284(62.12)0.00
Age group0.010.7577
18–3432541609(23.33)1645(23.85)
35–4954632722(39.47)2741(39.75)
50–6434611755(25.45)1706(24.74)
65+1614810(11.75)804(11.66)
Covariate
Acute myocardial infarction129(0.13)3(0.04)0.030.0831
Congestive heart failure11167(0.97)44(0.64)0.040.0284
Peripheral vascular disease3218(0.26)14(0.20)0.010.4790
Cerebral vascular accident369184(2.67)185(2.68)0.000.9579
Dementia3720(0.29)17(0.25)0.010.6214
Pulmonary disease876449(6.51)427(6.19)0.010.4424
Connective tissue disorder9952(0.75)47(0.68)0.010.6140
Peptic ulcer21941113(16.14)1081(15.68)0.010.4563
Liver cirrhosis34471964(28.48)1483(21.51)<0.0001
Hepatitis B Virus78853605(52.28)4280(62.06)<0.0001
Hepatitis C Virus30791653(23.97)1426(20.68)<0.0001
Alcohol35442159(31.31)1385(20.08)<0.0001
Diabetes920485(7.03)435(6.31)0.030.0879
Diabetes complications189115(1.67)74(1.07)0.050.0027
Paraplegia3618(0.26)18(0.26)0.001.0000
Renal disease250139(2.02)111(1.61)0.030.0739
Severe liver disease32(0.03)1(0.01)0.010.5637
Hypertension19691032(14.97)937(13.59)0.040.0208
Prior medications
Interferon-based therapy
interferon4323(0.33)20(0.29)0.010.6468
Metformin605328(4.76)277(4.02)0.040.0340
Aspirin520286(4.15)234(3.39)0.040.0201
Angiotensin converting-enzyme inhibitor376200(2.90)176(2.55)0.020.2095
Captopril13670(1.02)66(0.96)0.7303
Lisinopril6335(0.51)28(0.41)0.3767
Perindopril5125(0.36)26(0.38)0.8884
Ramipril4223(0.33)19(0.28)0.5365
Quinapril2613(0.19)13(0.19)1.0000
Benazepril32(0.03)1(0.01)0.5637
Cilazapril178(0.12)9(0.13)0.8082
Fosinopril6236(0.52)26(0.38)0.2031
Lipid lowering drugs222129(1.87)93(1.35)0.040.0149
Clofibrate0.0149
Bezafibrate3013(0.19)17(0.25)0.4647
Gemfibrozil12978(1.13)51(0.74)0.0169
Fenofibrate7444(0.64)30(0.44)0.1027
Nicotinic acid42(0.03)2(0.03)1.0000
Acipimox76(0.09)1(0.01)0.0587
Statins8148(0.70)33(0.48)0.030.0946
Atorvastatin0.0946
Fluvastatin4726(0.38)21(0.30)0.4650
Pitavastatin0.4650
Rosuvastatin3019(0.28)11(0.16)0.1437
Simvastatin75(0.07)2(0.03)0.2567
Diuretics272145(2.10)127(1.84)0.020.2703
Furosemide233125(1.81)108(1.57)0.2613
Spironolactone7536(0.52)39(0.57)0.7283
Selective beta blockers577293(4.25)284(4.12)0.010.7019
Table 2. Outcomes between the two groups. HCC—hepatocellular carcinoma.
Table 2. Outcomes between the two groups. HCC—hepatocellular carcinoma.
VariableNExposureNon-Exposurep-Value
n(%)n(%)
Total13,7926896(50.00)6896(50.00)
Spontaneous bacterial peritonitis15098(1.42)52(0.75)0.0002
Decompensation546396(5.74)150(2.18)<0.0001
Hepatorenal syndrome2818(0.26)10(0.15)0.1302
Other sequelae of chronic liver disease4129(0.42)12(0.17)0.0078
Ascites342298(4.32)44(0.64)<0.0001
Jaundice7554(0.78)21(0.30)0.0001
Hepatic coma294191(2.77)103(1.49)<0.0001
Variceal bleeding260197(2.86)63(0.91)<0.0001
All-cause mortality966577(8.37)389(5.64)<0.0001
Liver transplantation216(0.09)15(0.22)0.0494
HCC462232(3.36)230(3.34)0.9246
Table 3. Factors associated with spontaneous bacterial peritonitis. CCI—Charlson comorbidity index.
Table 3. Factors associated with spontaneous bacterial peritonitis. CCI—Charlson comorbidity index.
VariableAdjusted HR95% CIp-Value
Propranolol Exposure vs. Non-Exposure1.24(0.88)(1.75)0.2111
Sex
 Male vs. female1.90(1.30)(2.76)0.0008
Age Group
 18–34
 35–491.15(0.66)(2.00)0.6204
 50–641.34(0.74)(2.40)0.3307
 65+2.02(1.08)(3.75)0.0270
Covariate
CCI
 Congestive heart failure1.98(0.76)(5.11)0.1603
 Peripheral vascular disease2.98(0.41)(21.83)0.2819
 Cerebral vascular accident0.53(0.18)(1.61)0.2657
 Pulmonary disease0.92(0.52)(1.63)0.7716
 Peptic ulcer1.27(0.86)(1.88)0.2279
 Liver cirrhosis1.02(0.69)(1.49)0.9260
 Diabetes1.17(0.61)(2.24)0.6318
 Diabetes complications0.57(0.17)(1.90)0.3612
 Paraplegia4.72(0.55)(40.53)0.1570
 Renal disease1.63(0.70)(3.80)0.2605
 Hypertension0.71(0.43)(1.18)0.1845
Baseline Medications
 Metformin1.41(0.67)(2.95)0.3676
 Aspirin0.45(0.17)(1.15)0.0949
 Angiotensin converting-enzyme inhibitor0.82(0.34)(2.01)0.6670
 Lipid lowering drugs1.10(0.35)(3.52)0.8688
 Diuretics1.83(0.96)(3.47)0.0652
 Selective beta blockers0.83(0.34)(2.04)0.6904
Concomitant Medications
 Selective beta blockers0.35(0.19)(0.67)0.0014
 Diuretics8.56(5.93)(12.37)<0.0001
Table 4. Factors associated with all-cause mortality.
Table 4. Factors associated with all-cause mortality.
VariableAdjusted HR95% CIp-Value
Propranolol Exposure
1~28 Days vs. non-exposure1.33(1.15)(1.53)0.0001
29~90 Days vs. non-exposure1.12(0.91)(1.40)0.2888
>90 Days vs. non-exposure0.79(0.64)(0.98)0.0340
Sex
 Male vs. female1.59(1.38)(1.83)<0.0001
Age Group
 18–34
 35–491.73(1.34)(2.21)<0.0001
 50–642.63(2.04)(3.40)<0.0001
 65+5.07(3.89)(6.61)<0.0001
Covariate
CCI
 Acute myocardial infarction2.27(0.89)(5.82)0.0870
 Congestive heart failure1.37(0.95)(1.97)0.0914
 Peripheral vascular disease3.15(1.67)(5.93)0.0004
 Cerebral vascular accident1.56(1.20)(2.01)0.0008
 Dementia1.86(1.04)(3.32)0.0350
 Pulmonary disease1.25(1.03)(1.52)0.0218
 Connective tissue disorder0.49(0.16)(1.51)0.2133
 Peptic ulcer1.29(1.11)(1.50)0.0009
 Liver cirrhosis0.78(0.67)(0.92)0.0025
 Diabetes1.17(0.92)(1.50)0.2098
 Diabetes complications1.21(0.85)(1.72)0.2872
 Paraplegia0.75(0.31)(1.79)0.5116
 Renal disease1.87(1.42)(2.48)<0.0001
 Severe liver disease1.61(0.22)(11.63)0.6392
 Hypertension1.06(0.89)(1.27)0.5056
Baseline Medications
 Metformin1.16(0.87)(1.55)0.3157
 Aspirin0.84(0.65)(1.09)0.1964
 Angiotensin converting-enzyme inhibitor0.71(0.52)(0.97)0.0307
 Lipid lowering drugs0.80(0.48)(1.32)0.3820
 Statins0.91(0.40)(2.04)0.8147
 Diuretics1.88(1.45)(2.44)<0.0001
 Selective beta blockers0.94(0.70)(1.26)0.6631
Concomitant Medications
 Selective beta blockers0.44(0.35)(0.56)<0.0001
 Diuretics2.69(2.33)(3.11)<0.0001
Table 5. Factors associated with hepatocellular carcinoma.
Table 5. Factors associated with hepatocellular carcinoma.
VariableAdjusted HR95% CIp-Value
Propranolol Exposure
1~28 Days vs. non-exposure0.81(0.65)(1.01)0.0580
29~90 Days vs. non-exposure0.80(0.58)(1.09)0.1588
>90 Days vs. non-exposure0.49(0.36)(0.67)<0.0001
Sex
 Male vs. female1.47(1.20)(1.79)0.0002
Age Group
 18–34
 35–493.33(2.02)(5.49)<0.0001
 50–647.67(4.68)(12.55)<0.0001
 65+10.72(6.43)(17.88)<0.0001
Covariate
CCI
 1Acute myocardial infarction1.82(0.25)(13.18)0.5552
 2Congestive heart failure0.98(0.45)(2.10)0.9489
 3Peripheral vascular disease0.74(0.10)(5.26)0.7597
 4Cerebral vascular accident0.78(0.46)(1.33)0.3695
 6Pulmonary disease0.79(0.57)(1.10)0.1584
 7Connective tissue disorder0.64(0.16)(2.58)0.5294
 8Peptic ulcer1.03(0.82)(1.30)0.7978
 9Liver cirrhosis1.46(1.19)(1.78)0.0002
 10Diabetes0.75(0.51)(1.10)0.1362
 11Diabetes complications1.10(0.62)(1.96)0.7375
 12Renal disease0.90(0.47)(1.71)0.7458
 13Hypertension0.49(0.37)(0.66)<0.0001
Baseline Medications
 Metformin2.08(1.40)(3.08)0.0003
 Aspirin0.71(0.46)(1.11)0.1355
 Angiotensin converting-enzyme inhibitor1.23(0.80)(1.90)0.3513
 Lipid lowering drugs0.50(0.20)(1.21)0.1248
 Statins1.30(0.41)(4.09)0.6527
 Diuretics0.74(0.44)(1.24)0.2547
 Selective beta blockers1.21(0.78)(1.87)0.3858
Concomitant Medications
 Selective beta blockers0.54(0.39)(0.74)0.0001
 Diuretics5.65(4.61)(6.92)<0.0001

Share and Cite

MDPI and ACS Style

Yeh, W.-S.; Yang, S.-C.; Liang, C.-M.; Li, Y.-C.; Tai, W.-C.; Lee, C.-H.; Yang, Y.-H.; Hsu, C.-N.; Tsai, T.-H.; Chuah, S.-K.; et al. The Role of Non-Selective β-Blockers in Compensated Cirrhotic Patients without Major Complications. Medicina 2020, 56, 14. https://doi.org/10.3390/medicina56010014

AMA Style

Yeh W-S, Yang S-C, Liang C-M, Li Y-C, Tai W-C, Lee C-H, Yang Y-H, Hsu C-N, Tsai T-H, Chuah S-K, et al. The Role of Non-Selective β-Blockers in Compensated Cirrhotic Patients without Major Complications. Medicina. 2020; 56(1):14. https://doi.org/10.3390/medicina56010014

Chicago/Turabian Style

Yeh, Wen-Shuo, Shih-Cheng Yang, Chih-Ming Liang, Yu-Chi Li, Wei-Chen Tai, Chen-Hsiang Lee, Yao-Hsu Yang, Chien-Ning Hsu, Tzu-Hsien Tsai, Seng-Kee Chuah, and et al. 2020. "The Role of Non-Selective β-Blockers in Compensated Cirrhotic Patients without Major Complications" Medicina 56, no. 1: 14. https://doi.org/10.3390/medicina56010014

Article Metrics

Back to TopTop