Effect of L-carnitine on quality of life in covert hepatic encephalopathy: a randomized, double-blind, placebo-controlled study

Article information

Korean J Intern Med. 2022;37(4):757-767
Publication date (electronic) : 2022 May 06
doi : https://doi.org/10.3904/kjim.2021.338
1Department of Internal Medicine, Inje University Sanggye Paik Hospital, Seoul, Korea
2Hanyang University Seoul Hospital, Hanyang University College of Medicine, Seoul, Korea
3Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, Korea
4Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
5St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
6National Medical Center, Seoul, Korea
7Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
8Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Korea
9Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
10Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
11Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
12Hallym University Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
13Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
14Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
15Konyang University College of Medicine, Daejeon, Korea
Correspondence to: Dae Won Jun, M.D., Department of Internal Medicine, Hanyang University College of Medicine, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea, Tel: +82-2-2290-8338, Fax: +82-2-972-0068, E-mail: noshin@hanyang.ac.kr
*

These authors contributed equally to this work.

Received 2021 July 22; Revised 2021 September 22; Accepted 2021 October 05.

Abstract

Background/Aims

L-carnitine is potentially beneficial in patients with hepatic encephalopathy (HE). We aimed to evaluate the impact of L-carnitine on the quality of life and liver function in patients with liver cirrhosis and covert HE.

Methods

We conducted an investigator-initiated, prospective, multi-center, double-blind, randomized phase III trial in patients with covert HE. A total of 150 patients were randomized 1:1 to L-carnitine (2 g/day) or placebo for 24 weeks. Changes in quality of life and liver function were assessed at 6 months. The model for end-stage liver disease (MELD), the 36-Item Short Form Survey (SF-36), the psychometric hepatic encephalopathy score (PHES), and the Stroop Test were evaluated in all patients.

Results

The total SF-36 score significantly improved in the L-carnitine group after 24 weeks (difference: median, 2; interquartile range, 0 to 11; p < 0.001); however, these values were comparable between the two groups. Furthermore, there was a significant ordinal improvement in PHES scores among patients with minimal HE who were in the L-carnitine group (p = 0.007). Changes in the total carnitine level also positively correlated with improvements in the Stroop test in the L-carnitine group (color test, r = 0.3; word test, r = 0.4; inhibition test, r = 0.5; inhibition/switching test, r = 0.3; all p < 0.05). Nevertheless, the MELD scores at week 24 did not differ between the groups.

Conclusions

Twenty-four weeks of L-carnitine supplementation was safe but ineffective in improving quality of life and liver function.

Graphical abstract

INTRODUCTION

Patients with covert hepatic encephalopathy (HE), which includes West Haven grade I HE to minimal HE, have decreased cognitive functions, including attention, space-time, coordination, exercise speed, and reaction time [1,2]. Covert HE decreases the ability to work and increases the risk of job loss [3]. Additionally, it increases the risk of falls, fractures, and death [4]. Therefore, the early diagnosis of covert HE is important for timely intervention and prognosis in patients with liver cirrhosis. Lactulose and rifaximin have been shown to improve cognitive function and quality of life in covert HE [5]. Other treatment options are under investigation, including carnitine, probiotics, and L-ornithine-L-aspartate.

L-carnitine plays an important role in the transportation of long-chain fatty acids into the mitochondria [6] and activates the tricarboxylic cycle, hence inducing ureagenesis, and thereby reducing ammonia concentrations [7]. Several reports have suggested that carnitine has additional effects on chronic liver disease [810]. To date, there have been six clinical studies on carnitine use in patients with HE [1116]. However, these studies have all been conducted by the same group. Moreover, their definition of covert HE and assessment of cognitive dysfunction were unclear. Furthermore, whether carnitine can improve quality of life or can have additional benefits in patients with covert HE has not yet been elucidated. We hence need more data based on double-blind studies with a larger cohort to draw a better conclusion on these issues.

Therefore, we performed a randomized, double-blind, placebo-controlled study, wherein we administered L-carnitine to patients with liver cirrhosis and covert HE and evaluated its effects on quality of life and liver function.

METHODS

Study design

This multicenter, double-blind, investigator-initiated, and phase III clinical trial (HELIOS study; registered at the Clinical Research Information Service; KCT0002029, https://cris.nih.go.kr/) was conducted at 13 academic centers in the Republic of Korea. Patients with covert HE were randomized 1:1 to receive L-carnitine or placebo for 6 months (Fig. 1).

Figure 1

Flow chart of patient selection.

Randomization

Participants who met the inclusion criteria were registered in the trial via electronic data capture (a web-based registration system) and an interactive web-based response system (a web-based randomization system). A random allocation sequence was generated using a computer. Randomization was performed with the blocks stratified by the center and severity of liver disease (Child-Pugh A vs. B or C). Participant allocation was concealed from both the investigator and the participant.

Inclusion criteria

Thirteen academic centers in the Republic of Korea participated in this study. The inclusion criteria were as follows: (1) patients with liver cirrhosis aged 19 to 65 years and (2) diagnosed with covert HE. Liver cirrhosis was diagnosed through either liver biopsy or clinical findings, meeting one or more of the following criteria: (1) presence of surface nodularity of the liver and one or more signs of portal hypertension (ascites, splenomegaly, or portosystemic shunt) on abdominal imaging; (2) esophageal or gastric varices on endoscopy; (3) thrombocytopenia (platelet count ≤ 100,000/mm3); (4) hypoalbuminemia (albumin ≤ 3.5 g/dL); and (5) prolongation of prothrombin time (PT) (international normalized ratio ≥ 1.3). Minimal HE was diagnosed when the psychometric hepatic encephalopathy score (PHES) was < −4. Covert HE was defined as either grade 1 in the West Haven classification or grade 0 in the same classification while meeting the minimal HE definition described above.

Exclusion criteria

Patients were excluded if they met any of the following criteria: (1) previously diagnosed with overt HE; (2) prescribed and treated with medications affecting the natural course of HE, such as neomycin, lactulose, lactitol, branched-chain amino acids, probiotics, and rifaximin in the 2 weeks before enrollment; (3) had been or is currently being treated for a neurological or psychiatric disease; (4) is being treated with viable hepatocellular carcinoma or other malignancies; (5) had developed an infection or gastrointestinal bleeding within 6 weeks before enrollment; (6) has a serum alanine aminotransferase level > 400 IU/L; (7) has a serum creatinine level > 1.5 mg/dL; or (8) has alcohol dependence or a history of alcohol abuse.

Intervention

At the baseline visit (Visit 1), each patient was provided with 2 g (three tablets of 333 mg to be taken twice a day) of either L-carnitine or placebo drugs orally, to be repeatedly taken for 24 weeks.

Study endpoints

The primary endpoint was to assess the effect of L-carnitine on the improvement of quality of life compared to that of the placebo. This assessment was performed using the 36-Item Short Form Survey (SF-36) questionnaire at 24 weeks. The secondary endpoints were to assess the additional effects of L-carnitine administration for 24 weeks on the improvement of the model for end-stage liver disease (MELD), serum ammonia, and normalization of covert HE compared to the placebo.

Study follow-up and measurements

After the baseline visit, the patients were followed up every 3 months. The West Haven criteria, SF-36, PHES, and Stroop test were performed on all patients [17]. The PHES consisted of five tests: the digit symbol test, number connection test (NCT)-A, NCT-B, serial dotting, and line tracing. Assessments were based on normative Korean data [18]. Minimal HE was diagnosed when PHES scores were < −4. The Stroop test was developed to screen minimal/covert HE in both easier and more objective ways by using a web-based smartphone method. The diagnostic performance of minimal HE was excellent when using the Stroop test (area under the receiver operating characteristic curve, 0.75; 95% confidence interval, 0.67 to 0.84; p < 0.001) [17].

Sample size and rationale

The sample size was based on a previous study assessing the effects of acetyl-L-carnitine administration on the improvement of SF-36 in patients with minimal HE [16]. The difference in SF-36 score changes from the score measured at the baseline to that at 90 days between the acetyl-L-carnitine group and placebo group was 4.85, and the standard deviation was presumed to be 10 based on these results. We maintained a statistical power of 80% with a 2.5% one-sided significance level. In addition, we allowed a rate of withdrawal or loss to follow-up of up to 10%. The final sample size was estimated to be 150 participants.

Handling of dropouts or missing data

For the efficacy endpoints, the last observation carried forward method was used for missing data imputation. The SF-36 is expected to improve over time, hence it is more conservative to replace the missing data using this method. However, the available data set underwent safety analysis without imputation of missing data.

Statistical analysis

All analyses were performed using the full analysis set according to the intention-to-treat principle, which included all patients who received at least one dose of the investigational product. Demographic and baseline characteristics were summarized by the intervention group and were tested statistically to identify any imbalance between the groups. For continuous variables, group comparisons of efficacy endpoints were tested using an independent two-sample t test or Wilcoxon rank-sum test, whereas within-group comparisons were tested using the paired t test or Wilcoxon signed test. These tests were also performed depending on the results of the normality test. The generalized estimating equations logit model was used for categorical variables to test the time effect within a group and the interaction effect of time × group.

Ethics approval and informed consent

This study was approved by the Institutional Review Boards of all participating centers (HYUH 2015-10-003, SGPAIK 2015-11-010) and was performed in accordance with relevant guidelines and regulations. The study was conducted in accordance with the ethical principles of the Declaration of Helsinki. Informed consent was obtained from all the participants enrolled in this study.

RESULTS

Basic characteristics

A total of 238 patients with liver cirrhosis were screened between June 2015 and December 2017. Among them, 150 patients with covert HE evaluated using PHES underwent randomization. Among them, 72 and 70 patients were assigned to the placebo group and the L-carnitine group, respectively (Fig. 1, full analysis dataset). Among them, 21 patients in the placebo group and 12 in the L-carnitine group were dropped out because of consent withdrawal (16 patients in the placebo group and eight in the L-carnitine group), loss to follow-up (two patients in the placebo group), discontinuation due to adverse events (two patients in the placebo group and three in the L-carnitine group), and discontinuation at the investigator’s discretion (one patient from each group). Fifty-one and 58 patients completed the study at week 24 in each group. The most common etiology for liver cirrhosis was chronic hepatitis B (88.7%), and the proportion of patients with chronic hepatitis B receiving antiviral treatment did not differ between the groups (Table 1). The median MELD score was 8.0 points. The compliance of the patients in the placebo and L-carnitine groups was 90.5% ± 11.5% and 85.1% ± 17.2%, respectively, which was not significantly different.

Baseline characteristics of patients

Effects on quality of life

There was a significant improvement in SF-36 values within the L-carnitine group after 24 weeks (p < 0.001), but the median SF-36 values of both groups were comparable at week 24 (Table 2). Both physical and mental component scores were improved in the L-carnitine group, but the mental component score was improved in the placebo group as well, resulting in no difference between both groups at 24 weeks.

The comparison of quality of life assessed by SF-36 between the groups and within the group during 24 weeks

Effects on liver chemistry and MELD score

Aspartate aminotransferase levels were decreased in the L-carnitine group after 24 weeks of treatment compared to that at the baseline (p = 0.014) (Supplementary Table 1). PT also improved in the L-carnitine group, whereas total bilirubin concentration worsened in the placebo group after 24 weeks. The MELD scores were increased in the placebo group (p = 0.047) and were significantly decreased in the L-carnitine group (p = 0.008). Hence, the gaps in the change of MELD scores were significantly different between the two groups (p = 0.003). However, the MELD scores of both groups at week 24 were comparable (Fig. 2). Furthermore, the serum ammonia levels did not exhibit significant differences between or within each group after 24 weeks of treatment.

Figure 2

Comparison of the model for end-stage liver disease (MELD) scores and their components between the two groups. (A) MELD score, (B) prothrombin time-international normalized ratio (PT-INR), (C) total bilirubin, (D) creatinine. NS, not significant.

Effects on cognitive function

The changes in PHES scores and the normalization rates of minimal and covert HE at week 24 were compared between the groups (Supplementary Table 2). Both groups exhibited improved PHES scores, with no difference in values between groups at week 24. However, the ordinal association of the change in PHES scores during sequential visits (e.g., baseline, week 12, and week 24) among patients with minimal HE diagnosed at the baseline was significantly improved within the L-carnitine group (p = 0.007) (Fig. 3). The inhibition test, which is one of the subtests in the Stroop test, also showed significant improvement within the L-carnitine group after 24 weeks (2.3 ± 1.1 at the baseline vs. 2.7 ± 1.1 at 24 weeks, p < 0.001), but not in the placebo group (Fig. 4A). Additionally, this difference between the baseline level and the level at week 24 was significantly higher in the L-carnitine group (0.0 ± 0.8 in the placebo group vs. 0.3 ± 0.7 in the L-carnitine group, p = 0.038). However, the rate-correct scores of each subtest in the Stroop test were comparable for both groups at 24 weeks.

Figure 3

Comparison of the improvement in psychometric hepatic encephalopathy scores between the two patient groups among those with minimal hepatic encephalopathy diagnosed at the baseline.

Figure 4

Stroop test. (A) Comparison of the Stroop inhibition subtest between the two groups. (B) Correlation of serum carnitine level and Stroop results among the patients in the L-carnitine group. RCS, rate-correct score; NS, not significant; Δ, difference.

Correlation of serum carnitine level and cognitive function/quality of life

Serum levels of total carnitine and free carnitine were elevated in the L-carnitine group and were significantly different between the two groups at week 24 (reference values: 42–81 μmol/L [males] and 31–67 μmol/L [females] for total carnitine; 35–67 μmol/L [males] and 25–55 μmol/L [females] for free carnitine; 3.8–19 μmol/L for acyl carnitine). However, serum acylcarnitine levels did not significantly increase in the L-carnitine group. Furthermore, the change in total carnitine level between the baseline and at week 24 was positively correlated with the improvement of rate-correct scores of all four components in the Stroop test (color test, r = 0.3; word test, r = 0.4; inhibition test, r = 0.5; switching test, r = 0.3; all p values < 0.05) (Fig. 4B). However, the change in total carnitine level between the baseline and at week 24 was not significantly correlated with improvement in quality of life as assessed by the SF-36.

Safety analysis

Adverse events were observed in 25 out of 72 patients in the placebo group and 29 out of 70 patients in the L-carnitine group (34.7% vs. 41.4%, p = 0.490). In addition, the rate of serious adverse events did not differ between the two groups (8% in the placebo group vs. 11% in the L-carnitine group). However, there were no serious adverse events related to the study drugs in either group. Overall, 21 patients out of the full analysis data set stopped the study drugs either transiently or permanently. There were 5.6% (four out of 72 patients) and 7.1% (five out of 70 patients) in the placebo and L-carnitine groups, respectively, who permanently discontinued the medication due to adverse events, and these numbers did not differ between the groups. Events leading to death were also not observed in either group.

Per-protocol analysis of the endpoints

Sixty-five patients were included in the per-protocol analysis (32 patients in the placebo group and 33 in the L-carnitine group) (Fig. 1). Nineteen patients in the placebo group and 25 patients in the L-carnitine group were excluded from the per-protocol analysis due to one or more reasons such as inadequate compliance to the medications (< 80%), violation of the visiting windows, or not meeting the inclusion/exclusion criteria. The total SF-36 scores, especially the physical component scores, PHES scores, and the rate-correct scores of the inhibition test were improved within the L-carnitine group after 24 weeks of treatment (Supplementary Table 3). However, the SF-36 scores, PHES scores, normalization rate of minimal HE, and rate-correct scores on the Stroop test at week 24 did not differ between the two groups.

DISCUSSION

This is the first prospective, randomized, double-blind, placebo-controlled, multicenter clinical trial that evaluated the effects of carnitine on quality of life and liver function. The definition and diagnostic criteria of minimal HE were unclear in two previous clinical trials conducted in patients with minimal HE [15,16]. The patients included in those studies had liver cirrhosis but had no overt HE compared to patients with liver cirrhosis and only minimal HE. Moreover, their assessment of minimal HE had not been based on data normative to their population [16].

Our study has several strengths compared to previous ones. First, this study was based on the largest number of participants with the longest period of treatment (up to 24 weeks). Most previous studies have evaluated the effects of carnitine for only 4 to 12 weeks [19]. Secondly, this is the first study to evaluate the improvement of cognitive function by performing an assessment using the Stroop test as well as the PHES, which were based on normative data [17,18]. Stroop results and MELD scores are more objective than the PHES values in the evaluation of liver function among patients with covert HE. The difference in the median rate-correct scores of the Stroop inhibition test after 24 weeks was significantly higher in the L-carnitine group than in the placebo group. Additionally, liver function improved within the L-carnitine group based on MELD scores. However, the median rate-correct scores of the Stoop inhibition test and the MELD scores did not significantly differ between the placebo and L-carnitine groups at week 24. Therefore, we are cautious in drawing a conclusion regarding the beneficial effects of L-carnitine on the quality of life of patients with covert HE, which was the primary endpoint of this study.

Our study had some limitations. First, a quarter of the patients dropped before the completion of the study period. Missing data were handled using the last observation carried forward method in the analysis of the endpoints, except for the safety outcome. Therefore, the effects of L-carnitine may have been underestimated. Additionally, the possibility of a type II error should be considered in the interpretation of our findings, since the limited number of patients in each arm could have led to the limited effect of L-carnitine on the quality of life. Nevertheless, we arrived at similar results when the per-protocol analysis was applied, despite half of the patients in each group not being included in the analyses. Additionally, the change in total carnitine levels after 24 weeks showed a positive correlation with the improvement in the Stroop test. Traditionally, serum carnitine levels are reportedly maintained at normal concentrations among patients with liver cirrhosis [20,21]. The disruption of mitochondrial permeability and the subsequent lowering of L-carnitine levels incorporated into the mitochondria in patients with cirrhosis reportedly lead to intrinsic insufficiency at the mitochondrial level, even with normal serum levels [22]. Therefore, we could assume that patients with liver cirrhosis and covert HE may benefit from L-carnitine supplementation even with normal serum concentrations. Second, the appropriate dose of L-carnitine required for the improvement of cognitive function and quality of life could not be determined in our study. Nevertheless, the little increase in serum acylcarnitine levels following 24 weeks of L-carnitine treatment suggests that 2 g of L-carnitine may not be sufficient to discriminate its positive effects. In previous studies, L-carnitine was administered at doses up to 4 to 6 g per day [23]. If we had administered higher doses of L-carnitine, the effects on cognitive function and quality of life could have been more remarkable than our present findings. Third, as 89% of our participants had chronic hepatitis B, we could not exclude the effect of antiviral treatment on the marginal improvement in the results of the inhibition test or the MELD scores in the L-carnitine group. However, the proportion of patients who were already on stable antiviral treatment did not differ between the groups. Therefore, the possible beneficial effects of antiviral treatment were minimal in this study. Fourth, following the improved results of PHES in both groups without significant change in serum ammonia levels, we could not exclude the possibility of learning effects that patients could have achieved during the repeated examinations throughout the study period.

In conclusion, 24 weeks of L-carnitine supplementation was safe but was not effective in improving the quality of life among patients with covert HE. However, their MELD scores were stabilized with L-carnitine supplementation, and the difference in total carnitine concentration exhibited a positive correlation with improvements in cognitive function among these patients.

KEY MESSAGE

1. Twenty-four weeks of L-carnitine supplementation (2 g/day) was ineffective in the improvement of quality of life in liver cirrhosis patients with covert hepatic encephalopathy (CHE).

2. Twenty-four weeks of L-carnitine supplementation (2 g/day) was safe in liver cirrhosis patients with CHE.

3. Further studies with higher doses of L-carnitine supplementation would be needed to confirm the possible beneficial effects on the improvement of covert hepatic encephalopathy and liver function.

Supplementary Information

Notes

Conflict of interest

This study was financially supported by the Ildong Pharmaceutical Company. The funding source had no role in the study design, implementation, data collection, analysis, and interpretation, or in the preparation, review, and approval of the manuscript.

References

1. Flud CR, Duarte-Rojo A. Prognostic implications of minimal/covert hepatic encephalopathy: large-scale validation cohort studies. J Clin Exp Hepatol 2019;9:112–116.
2. Vilstrup H, Amodio P, Bajaj J, et al. Hepatic encephalopathy in chronic liver disease: 2014 practice guideline by the American Association for the Study of Liver Diseases and the European Association for the Study of the Liver. Hepatology 2014;60:715–735.
3. Roman E, Cordoba J, Torrens M, et al. Minimal hepatic encephalopathy is associated with falls. Am J Gastroenterol 2011;106:476–482.
4. Patidar KR, Thacker LR, Wade JB, et al. Covert hepatic encephalopathy is independently associated with poor survival and increased risk of hospitalization. Am J Gastroenterol 2014;109:1757–1763.
5. Bajaj JS, Pinkerton SD, Sanyal AJ, Heuman DM. Diagnosis and treatment of minimal hepatic encephalopathy to prevent motor vehicle accidents: a cost-effectiveness analysis. Hepatology 2012;55:1164–1171.
6. Jun DW, Cho WK, Jun JH, et al. Prevention of free fatty acid-induced hepatic lipotoxicity by carnitine via reversal of mitochondrial dysfunction. Liver Int 2011;31:1315–1324.
7. Morris AJ, Carey EM. Postnastal changes in the concentration of carnitine and acylcarnitines in the rat brain. Brain Res 1983;284:381–384.
8. Hassan A, Tsuda Y, Asai A, et al. Effects of oral L-carnitine on liver functions after transarterial chemoembolization in intermediate-stage HCC patients. Mediators Inflamm 2015;2015:608216.
9. Bae JC, Lee WY, Yoon KH, et al. Improvement of nonalcoholic fatty liver disease with carnitine-orotate complex in type 2 diabetes (CORONA): a randomized controlled trial. Diabetes Care 2015;38:1245–1252.
10. Jun DW, Kim BI, Cho YK, et al. Efficacy and safety of entecavir plus carnitine complex (GODEX®) compared to entecavir monotherapy in patient with ALT elevated chronic hepatitis B: randomized, multicenter open-label trials: the GOAL study. Clin Mol Hepatol 2013;19:165–172.
11. Malaguarnera M, Vacante M, Motta M, et al. Acetyl-L-carnitine improves cognitive functions in severe hepatic encephalopathy: a randomized and controlled clinical trial. Metab Brain Dis 2011;26:281–289.
12. Malaguarnera M, Vacante M, Giordano M, et al. Oral acetyl-L-carnitine therapy reduces fatigue in overt hepatic encephalopathy: a randomized, double-blind, placebo-controlled study. Am J Clin Nutr 2011;93:799–808.
13. Malaguarnera M, Risino C, Cammalleri L, et al. Branched chain amino acids supplemented with L-acetylcarnitine versus BCAA treatment in hepatic coma: a randomized and controlled double blind study. Eur J Gastroenterol Hepatol 2009;21:762–770.
14. Malaguarnera M, Pistone G, Astuto M, et al. Effects of L-acetylcarnitine on cirrhotic patients with hepatic coma: randomized double-blind, placebo-controlled trial. Dig Dis Sci 2006;51:2242–2247.
15. Malaguarnera M, Gargante MP, Cristaldi E, et al. Acetyl-L-carnitine treatment in minimal hepatic encephalopathy. Dig Dis Sci 2008;53:3018–3025.
16. Malaguarnera M, Bella R, Vacante M, et al. Acetyl-L-carnitine reduces depression and improves quality of life in patients with minimal hepatic encephalopathy. Scand J Gastroenterol 2011;46:750–759.
17. Yoon EL, Jun DW, Jeong JY, et al. Validation of the Korean Stroop Test in diagnosis of minimal hepatic encephalopathy. Sci Rep 2019;9:8027.
18. Seo YS, Yim SY, Jung JY, et al. Psychometric hepatic encephalopathy score for the detection of minimal hepatic encephalopathy in Korean patients with liver cirrhosis. J Gastroenterol Hepatol 2012;27:1695–1704.
19. Askarpour M, Djafarian K, Ghaedi E, Sadeghi O, Sheikhi A, Shab-Bidar S. Effect of L-carnitine supplementation on liver enzymes: a systematic review and meta-analysis of randomized controlled trials. Arch Med Res 2020;51:82–94.
20. Fuller RK, Hoppel CL. Elevated plasma carnitine in hepatic cirrhosis. Hepatology 1983;3:554–558.
21. D’Arienzo A, Mattera D, Ambrogio G, Duranti L, Zeuli L, Mazzacca G. Hypercarnitinemia in cirrhosis. Hepatology 1985;5:343.
22. Saito M, Hirano H, Yano Y, Momose K, Yoshida M, Azuma T. Serum level of taurine would be associated with the amelioration of minimal hepatic encephalopathy in cirrhotic patients. Hepatol Res 2016;46:215–224.
23. Abbasnezhad A, Choghakhori R, Kashkooli S, Alipour M, Asbaghi O, Mohammadi R. Effect of L-carnitine on liver enzymes and biochemical factors in hepatic encephalopathy: a systematic review and meta-analysis. J Gastroenterol Hepatol 2019;34:2062–2070.

Article information Continued

Figure 1

Flow chart of patient selection.

Figure 2

Comparison of the model for end-stage liver disease (MELD) scores and their components between the two groups. (A) MELD score, (B) prothrombin time-international normalized ratio (PT-INR), (C) total bilirubin, (D) creatinine. NS, not significant.

Figure 3

Comparison of the improvement in psychometric hepatic encephalopathy scores between the two patient groups among those with minimal hepatic encephalopathy diagnosed at the baseline.

Figure 4

Stroop test. (A) Comparison of the Stroop inhibition subtest between the two groups. (B) Correlation of serum carnitine level and Stroop results among the patients in the L-carnitine group. RCS, rate-correct score; NS, not significant; Δ, difference.

Table 1

Baseline characteristics of patients

Characteristic Total (n = 142) Placebo (n = 72) L-carnitine (n = 70) p valuea
Age, yr 54 (49.0 to 59.0) 55 (49.0 to 59.0) 54 (49.0 to 59.0) 0.325
Male ex 92 (64.8) 46 (63.9) 46 (65.7) 0.325
Body mass index 25.3 (23.0 to 27.2) 25.0 (22.2 to 26.5) 25.6 (23.0 to 27.5) 0.273
Current alcohol drinker 38 (26.8) 16 (22.2) 22 (31.4) 0.470
Etiologies of liver cirrhosis 0.698
 Alcohol 14 (9.9) 9 (12.5) 5 (7.1)
 Hepatitis B 126 (88.7) 62 (86.1) 64 (91.4)
  On antiviral treatment 117 (82.3) 58 (80.6) 59 (84.3)
 Others 2 (1.4) 1 (1.4) 1 (1.4)
Child-Pugh Class 0.824
 A 117 (82.4) 59 (81.9) 58 (82.9)
 B 24 (16.9) 13 (18.1) 11 (15.7)
 C 1 (0.7) 0 1 (1.4)
MELD 8 (7 to 10) 8 (7 to 10) 8 (7 to 10) 0.868
Platelets, × 1,000/mm3 121 (79 to 157) 118 (67 to 162) 125 (83 to 153) 0.450
Prothrombin time, INR 1.1 (1.1 to 1.2) 1.1 (1.1 to 1.2) 1.1 (1.1 to 1.2) 0.648
Albumin, g/dL 4.3 (3.8 to 4.5) 4.2 (3.8 to 4.5) 4.3 (3.9 to 4.6) 0.550
Total bilirubin, mg/dL 0.9 (0.7 to 1.4) 0.9 (0.7 to 1.4) 0.9 (0.7 to 1.3) 0.989
Cholesterol, mg/dL 155 (126 to 179) 152 (122 to 175) 156 (127 to 183) 0.350
ALT, U/L 26 (19 to 39) 24 (16 to 38) 26 (20 to 41) 0.304
Glucose, mg/dL 107 (97 to 123) 105 (96 to 120) 109 (100 to 129) 0.133
Creatinine, mg/dL 0.8 (0.7 to 1.0) 0.8 (0.7 to 1.0) 0.8 (0.7 to 1.0) 0.799
Ascites 0.668
 No 120 (84.5) 63 (87.5) 57 (81.4)
 Controlled 20 (14.1) 8 (11.1) 12 (17.1)
 Uncontrolled 2 (1.4) 1 (1.4) 1 (1.4)
SF-36 71.9 (59.4 to 81.6) 72.4 (60.1 to 80.8) 71.7 (59.1 to 81.7) 0.998
West Haven Criteria 0.852
 MHE 103 (72.5) 53 (73.6) 50 (71.4)
 Grade 1 39 (27.5) 19 (26.4) 20 (28.6)
PHES (Z-score)
 NCT-A −6.0 (−1.5 to 0.1) −0.7 (−1.6 to 0) −0.6 (−1.5 to 0.1) 0.509
 NCT-B −0.9 (−2.3 to 0.4) −0.9 (−2.1 to 0.2) −1.0 (−2.4 to 0.7) 0.890
 Digit symbol test −0.6 (−1.2 to 0.2) −0.6 (−1.2 to 0.1) −0.5 (−1.1 to 0.3) 0.320
 Serial dotting test 0.9 (0.3 to 1.4) 0.9 (0.3 to 1.4) 0.9 (0.3 to 1.5) 0.797
 Line tracing test (time) −0.8 (−1.6 to 0.3) −0.6 (−1.6 to 0.2) −0.8 (−1.6 to 0.3) 0.842
 Line tracing test (error) −0.7 (−3 to 0.5) −0.7 (−3 to 0.6) −0.6 (−2.7 to 0.3) 0.912
 Total score −3.0 (−5.0 to −1.0) −3.0 (−5.0 to −1.0) −3.0 (−5.0 to 0.0) 0.980
Stroop test
 Color to RCS 2.9 (2.1 to 3.4) 2.8 (2.1 to 3.3) 3 (2.1 to 3.6) 0.265
 Word to RCS 3.6 (3.1 to 4.0) 3.5 (3.1 to 4.0) 3.6 (3.1 to 4.0) 0.408
 Inhibition to RCS 2.3 (1.5 to 3.3) 2.5 (1.5 to 3.3) 2.2 (1.6 to 3.1) 0.782
 Switching to RCS 1.6 (1.1 to 2.3) 1.6 (1.1 to 2.3) 1.6 (1.2 to 2.4) 0.550
 Korean Stroop score 1 (0.0 to 2.0) 1 (0.0 to 2.0) 1 (0.0 to 2.0) 0.351

Values are presented as median (interquartile range), or number (%).

MELD, model for end-stage liver disease; INR, international normalized ratio; ALT, alanine aminotransferase; SF-36, 36-Item Short Form Survey; MHE, minimal hepatic encephalopathy; PHES, psychometric hepatic encephalopathy score; NCT, number connection test; RCS, rate correct score.

a

Comparison between the groups.

Table 2

The comparison of quality of life assessed by SF-36 between the groups and within the group during 24 weeks

Variable Baseline Week 24 Change of values (24 weeks–baseline)



Value p valuea Value p valuea Within group difference p valueb p valuec
Physical functioning 0.757 0.519 0.330

 Placebo 85 (68–95) 85 (68–93) 0 (0 to 5) 0.416

 L-carnitine 80 (70–95) 85 (70–95) 0 (0 to 10) 0.135

Role-physical 0.651 0.193 0.306

 Placebo 75 (56–94) 84 (63–100) 0 (0 to 13) 0.153

 L-carnitine 81 (63–100) 91 (75–100) 0 (0 to 19) < 0.001

Bodily pain 0.863 0.106 0.152

 Placebo 90 (70–100) 89 (63–100) 0 (−5 to 10) 0.617

 L-carnitine 90 (67.5–100) 90 (78–100) 0 (0 to 10) 0.112

General health 0.310 0.677 0.398

 Placebo 48 (35–60) 50 (40–60) 0 (−5 to 10) 0.398

 L-carnitine 50 (30–55) 50 (35–65) 0 (−5 to 10) 0.038

Vitality 0.739 0.944 0.773

 Placebo 50 (31–72) 50 (41–72) 0 (0 to 16) 0.006

 L-carnitine 50 (38–69) 56 (44–69) 0 (0 to 13) 0.022

Social functioning 0.878 0.513 0.245

 Placebo 88 (75–100) 88 (75–100) 0 (0 to 13) 0.285

 L-carnitine 88 (75–100) 100 (75–100) 0 (0 to 13) 0.058

Role-emotional 0.593 0.262 0.629

 Placebo 83 (67–100) 92 (67–100) 0 (−8 to 17) 0.169

 L-carnitine 92 (67–100) 92 (75–100) 0 (0 to 8) 0.148

Mental health 0.577 0.796 0.752

 Placebo 68 (55–80) 70 (58–80) 0 (−5 to 10) 0.213

 L-carnitine 70 (55–80) 70 (60–85) 0 (−10 to 10) 0.518

Physical component 0.789 0.216 0.107

 Placebo 73 (63–82) 74 (61–84) 0 (−4 to 5) 0.400

 L-carnitine 72 (58–83) 79 (66–86) 1 (−1 to 13) 0.001

Mental component score 0.723 0.735 0.546

 Placebo 72 (58–80) 74 (60–84) 0 (−2 to 9) 0.042

 L-carnitine 72 (59–81) 77 (64–86) 1.7 (−1 to 11) 0.010

Total score 0.998 0.342 0.163

 Placebo 72 (60–81) 74 (60–82) 0 (−2 to 7) 0.059

 L-carnitine 72 (59–82) 78 (63–86) 2 (0 to 11) < 0.001

Values are presented as median (interquartile range).

SF-36, 36-Item Short Form Survey.

a

Comparison between the groups.

b

Comparison between within group differences from two groups.

c

Comparison within the groups.