Radiol Oncol 2023; 57(2): 168-177. doi: 10.2478/raon-2023-0025 168 research article Radiological assessment of skeletal muscle index and myosteatosis and their impact postoperative outcomes after liver transplantation Miha Petric 1,2 , Taja Jordan 3 , Popuri Karteek 4 , Sabina Licen 5 , Blaz Trotovsek 1,2 , Ales Tomazic 1,2 1 Department of Abdominal Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia 2 Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia 3 Institute of Radiology, University Medical Centre Ljubljana, Ljubljana, Slovenia 4 Department of Computer Science, Memorial University of Newfoundland, St. John’s, NL, Canada 5 Faculty of Health Sciences, University of Primorska, Izola, Slovenia Radiol Oncol 2023; 57(2): 168-177. Received 12 March 2023 Accepted 16 May 2023 Correspondence to: Miha Petrič; Department of Abdominal Surgery, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1000 Ljubljana, Slovenia. E mail: miha.petric@kclj.si Disclosure: No potential conflicts of interest were disclosed. This is an open access article distributed under the terms of the CC-BY license (https://creativecommons.org/licenses/by/4.0/). Background. Liver transplantation offers curative treatment to patients with acute and chronic end-stage liver disease. The impact of nutritional status on postoperative outcomes after liver transplantation remains poorly under- stood. The present study investigated the predictive value of radiologically assessed skeletal muscle index (SMI) and myosteatosis (MI) on postoperative outcomes. Patients and methods. Data of 138 adult patients who underwent their first orthotopic liver transplantation were retrospectively analysed. SMI and MI in computer tomography (CT) scan at the third lumbar vertebra level were cal- culated. Results were analyzed for the length of hospitalisation and postoperative outcomes. Results. In 63% of male and 28.9% of female recipients, low SMI was found. High MI was found in 45(32.6%) patients. Male patients with high SMI had longer intensive care unit (ICU) stay (P < 0.025). Low SMI had no influence on ICU stay in female patients (P = 0.544), length of hospitalisation (male, P > 0.05; female, P = 0.843), postoperative complication rates (males, P = 0.883; females, P = 0.113), infection rate (males, P = 0.293, females, P = 0.285) and graft rejection (males, P = 0.875; females, P = 0.135). The presence of MI did not influence ICU stay (P = 0.161), hospitalization (P = 0.771), postoperative complication rates (P = 0.467), infection rate (P = 0.173) or graft rejection rate (P = 0.173). Conclusions. In our study, changes in body composition of liver transplant recipients observed with SMI and MI had no impact on postoperative course after liver transplantation. CT body composition analysis of recipients and uni- formly accepted cut-off points are crucial to producing reliable data in the future. Key words: muscle mass; liver transplantation; myosteatosis; skeletal muscle index; GLIM score Introduction Since 1963 when the first liver transplantation (LT) was performed by Starzl 1 , it has become a standard treatment modality for patients with acute liver failure and chronic liver disease. 2 Ninety percent 5-year survival rate and better quality of life are the two most important outcomes of LT. 2 Most liv- er transplant centres use the Model for End-stage Liver Disease (MELD) score for organ allocation. 3 Patients with MELD score 15 or more, patients with poor quality of life due to chronic liver disease Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 169 symptoms (diuretic-intractable ascites, variceal bleeding, pruritus, cachexia), and patients with acute liver failure are those who benefit most from LT. 3 MELD score can underestimate the severity of liver disease in specific groups of patients (acute on chronic liver disease, presence of sarcopenia, chronic kidney disease, etc.). 3 Several modifica- tions of the MELD score have been introduced, but none offers a more reliable and accurate scoring system. Albumin-bilirubin (ALBI) score is mainly used as an objective method to assess liver func- tion and predict postoperative complications, par- ticularly after hepatectomy in patients with hepa- tocellular carcinoma (HCC). 4 Its role in determin- ing post-LT outcomes is not yet determined. Two main objective parameters of nutritional status are sarcopenia and myosteatosis (MI). The European Working Group on Sarcopenia defines sarcopenia as the presence of low muscle mass (under the 5 th percentile) and low muscle function (strength or performance) in patients with advanced age, can- cer, or other diseases. 5,6 Myosteatosis is defined as the abnormal fatty transformation of skeletal muscle. It negatively affects muscle strength and is common in advanced age 7,8 , diabetes 7,8 , obesity 9,10 , chronic 9 , and malignant diseases. 11,12 Overview of the literature shows large number of different methods used for body composition assessment in patients with liver cirrhosis. 13 There is still no con- sensus on the best tools for each body component in patients with liver cirrhosis. Most frequently used are computed tomography (CT), bioimped- ance analysis (BIA), dual-energy X-ray absorp- tiometry (DXA) and anthropometry. 13 Some of them BIA, DXA and body mass index (BMI)) are not applicable in patients with end-stage liver dis- ease due to frequent water retention. 14,15 In the last decade, computed tomography with automated or semiautomated body composition analysis at the third lumbar vertebra has emerged as an objec- tive method of defining the nutritional status of patients with chronic liver disease. 15,16 Nutritional assessment with CT is not affected by water reten- tion or presence of ascites. Skeletal muscle volume and myosteatosis can be measured from CT images obtained as a part of routine pre-transplant evalu- ation. 14 Skeletal muscle mass index (SMI) is calcu- lated as muscle mass area divided by the square of the height. The Global Leadership Initiative on Malnutrition (GLIM) 16 score was introduced as a potential nutritional assessment tool in recent years. It was shown to have good predictive value as a risk assessment tool for postoperative morbid- ity and mortality in patients after colorectal sur- gery. 17 However, its role as a predictive factor in LT is not yet established. This study aimed to investigate feasibility of radiological assessment of a nutritional status of a patient and the predictive value of SMI and MI on postoperative complications, length of hospi- talization, liver graft rejection, and mortality. We further compared the predictive value of SMI and MI with MELD, ALBI, and GLIM scores. Patients and methods We retrospectively analyzed 138 adult patients who had first orthotropic LT from brain dead donors be- tween 1.1.2012 and 1.1.2020 in our institution. We excluded patients who had re-LT procedure and those whose abdominal CT scan could not be ob- tained from data base or received reduced size and liver graft from donation after cardiac death (DCD) donor. From the medical database, we collected re- cipient age, gender, body mass index, underlying liver disease, presence of ascites, hepatocellular carcinoma (HCC), and laboratory parameters (se- rum levels of sodium, creatinine, albumin, pro- tein, bilirubin, and International normalized ratio (INR)). We calculated MELD and ALBI scores from laboratory parameters. Among GLIM criteria, we used chronic liver failure and end-stage liver dis- ease as etiologic and SMI or MI as phenotypic cri- teria. Length of intensive care unit (ICU) stay, hos- pitalization, postoperative complications accord- ing to Clavien-Dindo classification 18 , infections, and 90-day mortality were collected from the da- tabase of liver recipients and analyzed. Diagnosis of liver rejections was confirmed with laboratory tests and histological examination of all liver graft specimens obtained by ultrasound-guided biopsy. Acute rejection was defined with 6 points or more according to the liver allograft fibrosis score. 19 The Slovenian National Medical Ethics Committee approved our study design (approval number 0120–230/2018–10) and waived the need to obtain informed consent from participants. CT-body composition analysis Abdominal CT scans were obtained from the hos- pital database system. In case of multiple CT scans, we used last CT scan before LT procedure. A single slice of each patient at the level of the 3 rd lumbar vertebrae was selected for automatic segmentation. CT scans were analysed using the “Automated Body Composition Analyzer using Computed to- Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 170 mography image Segmentation” (ABACS) 20 soft- ware, which uses predefined Hounsfield units (HU) values to recognize different tissues. ABACS uses HU values from −29 to +150 HU to assess and calculate the total cross-sectional area for mus- cular tissue (SMA – skeletal muscle area). The L3 skeletal muscles included the psoas muscle, the lumbar muscles, the erector spinae, the transver- sus abdominis muscle, the internal and external oblique muscles, and the rectus abdominis. SMI was calculated using the following formula: SMI = SMA (cm 2 ) / height2 (m 2 ) 9,21 and patients were di- vided into a group with lower SMI (men < 52.4 cm 2 / m 2 , women < 38 cm 2 /m 2 ) 9,21 and another with nor- mal SMI. MI was determined by the medium value of HU in a skeletal muscle area. We used recently defined threshold parameters for MI in a patient with a chronic liver disease (< 33 HU in patients with a BMI ≥ 25 kg/m 2 and < 41 HU in those with a BMI < 25). 22-24 Based on previous literature findings and conclusions no adjustment for sex was made for MI. 23,24 Statistical analysis Data were analysed using SPSS for macOS, 26 th edi- tion. Descriptive statistics such as frequencies, per- centages, mean/median, and standard deviations were used for description and summary. Because the data were not normally distributed, patient characteristics were compared between groups us- ing the Kruskal-Wallis test and the Mann-Whitney U test. In addition, Spearman rank-order correla- tion and multiple linear regression were used to determine the relationship between variables and predict patient outcomes based on their character- istics and condition, as well as the chi-square in- dependence test. A P-value ≤ 0.05 was considered statistically significant. Results Patients characteristics Between 1.1.2012 and 1.1.2020, 138 patients (100 men and 38 women) met the criteria for inclusion in the study. The median age was 57.5 years (22 to 69 years). Table 1 provides general data on the pop- ulation laboratory and clinical variables. Sixty-one (61, 44.2%) patients had LT due to in- herited or metabolic liver disease, 60 patients had alcoholic liver disease (43.5%), and 17 patients had virus-related liver disease (12.3%). At the time of LT, 80 (58%) patients had ascites, and 27 patients had HCC (19.4%). Incidence of low SMI and MI In our study group, 63% of male and 28.9% of fe- male patients had low SMI. MI was present in 45 (32.6%) patients. We found no statistical signifi- cance between the aetiology of underlying liver disease and SMI (P = 0.214). The aetiology of liver disease had a statistically significant influence on the incidence of MI. Patients with alcoholic ae- tiology had more fatty infiltrated muscles than patients with other liver disease aetiologies (P = 0.008) (Table 2). A Spearman’s rank-order correlation was run to determine the relationship between the MI and BMI. There was a moderate, negative correlation between MI and BMI, which was statistically sig- nificant (rs = -0.597, p < 0.000). TABLE 1. Laboratory and clinical data of patients Variable Min Max IQR SD 95% CI Lower Upper BMI 15 38 6 4.775 25.13 26.74 Waiting time for liver transplantation (days) 1 691 166 151.311 108.44 159.38 Sodium 121 147 6 4.701 136.09 137.6 8 Creatinine 41 696 46 67.0 82 85.91 108.49 Albumin 10 62 10 7.674 31.28 33.86 Protein 23 89 12 11.494 65.80 69.74 Bilirubin 2 687 56 107.535 51.83 88.04 INR 1 4 1 .502 1.44 1.61 MELD score 7 46 9 6.700 14.30 16.56 ALBI score -4 0 1.13 0.802 -1.87 -1.60 ALBI score = albumin-bilirubin score; BMI = body mass index; INR = international normalized ratio; IQR = Interquartile range; MELD score = model for end-stage liver disease score TABLE 2. Aetiology of liver disease and incidence of myosteatosis Variable Myosteatosis no yes Liver failure Alcohol-related 32 28 Virus-related 14 3 Other 47 14 χ2 9.717 Degrees of freedom (Df) 2 p 0.008 Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 171 Eighty patients (57 .9%) had decompensated liver cirrhosis with the presence of ascites at the time of CT scan. The Mann-Whitney U test indicates that ascites significantly impacted patients’ SMI. The presence of ascites correlated with low SMI (P < 0.05, Figure 1) and had no impact on the incidence of MI (P = 0.244). Presence of HCC in patients with liver disease (27 patients, 19.6%) had no influence on SMI (P = 0.546) or MI (P = 0.174). Influence of low SMI on hospitalization The Mann-Whitney U test indicates that male pa- tients with normal or high SMI (> 52.4 cm 2 /m 2) had a longer ICU length of stay (P < 0.025). Linear re- gression was calculated to predict ICU length of stay based on SMI. A significant regression equa- tion was found (F (1.96) = 6.823, P = 0.010) (Figure 2), with R 2 of 0.066. SMI was found to significantly predict length of ICU stay (β = 0.258; 95% CI 0.026, 0.189; P = 0.010). The predicted length of stay equals -1.117+0.107 (SMI) days when SMI is measured in cm 2 /m 2 . The ICU length of stay increased by 0.107 days for each cm 2 /m 2 of SMI in the male population. A Spearman’s rank-order correlation showed no difference in hospitalization time between males with low or normal SMI. Mann-Whitney U test in- dicates statistically significant differences between low or normal SMI (P < 0.05) in more extended hos- pitalization in male patients with normal SMI. In addition, the linear regression analysis was per- formed to predict the length of hospital stay based on SMI in male patients; however, no statistically significant regression equation was found (β = -0.031; 95% CI -0.656, 0.480; P > 0.05). In the female population, we found no statistically significant influence of SMI on ICU length of stay (β = -0.123; 95% CI -0.741, 0.463; P = 0.544) and hospitalization time (β = 0.013; 95% CI 0.075, 0.941; P = 0.843). Influence of MI on hospitalisation The Mann-Whitney U test indicates that there were no differences among the groups (MI and no- MI) regarding the ICU length of stay (P = 0.161) or hospitalization (P = 0.771). Influence of SMI and MI on postoperative complications Postoperative complications of stage 2 or more by Clavien-Dindo classification 19 were present in 79 patients (57.2%). Infection occurred in 59 patients (42.8%), most commonly as intra-abdominal in- fection (50.8%), respiratory tract infection (23.7%) and urosepsis (6.8%). 5 (3.6%) patients developed critical illness myopathy. Surgical intervention was needed in 38% of patients with postoperative complications. There was no statistically significant differ- ence in the frequency of postoperative compli- cations between males (P = 0.883) and females (P = 0.113) with low or normal SMI. The postopera- tive infection rate was similar in males (P = 0.293) and females (P = 0.285) with low or normal SMI. MI did not show significant influence on postop- erative complications (P = 0.839) and infection rate (P = 0.703). FIGURE 1. Correlation between ascites and skeletal muscle index. FIGURE 2. Scatter diagram showing a positive correlation between the intensive care unit length of stay (days) and skeletal muscle index. Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 172 Influence of SMI and MI on liver graft rejection Rejection was diagnosed in 22 patients (16.1%), 20 in male and 2 in female patients. We found no sta- tistically significant influence of SMI (males, P = 0.875; females, P = 0.135) and MI (P = 0.449) on liver graft rejection. Influence of MELD and ALBI score on postoperative outcomes The median MELD score was 14 (7 – 46). A Spearman’s rank-order correlation was run to de- termine the relationship between the MELD score and ICU length of stay (days). There was a weak, positive correlation between the MELD score and ICU length of stay, which was statistically signifi- cant (r s = 0.261, P < 0.002). There was no significant relationship between the MELD score and compli- cation rate or length of hospitalization (P > 0.05). There is a week, positive correlation between the ALBI score and ICU length of stay and hospi- talisation time, which was statistically significant (r s = 0.279, P < 0.001; r s = 0.197; P = 0.022). There is no correlation between the ALBI score and postopera- tive complications. GLIM score GLIM score using skeletal muscle index as a phenotypic factor We analyzed differences between patients who meets criteria for positive GLIM score (acute or chronic liver disease as etiological factor and skel- etal muscle index as a phenotypic factor) and those with negative GLIM score regarding ICU length of stay, postoperative complications, rate of infec- tions, graft rejections and mortality. Based on the chi-square independence test, no association was found between the groups in post- operative complications (χ2(1) = 0.600; P = 0.438), rate of infections (χ2(1) = 0.918; P = 0.338), graft re- jection (χ2(1) = 1.205; P = 0.272), and mortality (χ2(1) = 0.232; P = 0.630). In addition, there were no statis- tically significant differences in ICU length of stay between groups (χ2(14) = 15.125; P = 0.370), based on the Mann-Whitney test. GLIM score using myosteatosis as a phenotypic factor We analyzed differences between patient who meets criteria for positive GLIM score (acute or chronic liver disease as etiological factor and my- osteatosis as a phenotypic factor) and those with negative GLIM score regarding ICU length of stay, postoperative complications, rate of infections, graft rejections and mortality. Based on the chi-square independence test, there was no association between the groups in terms of postoperative complications (χ2(1) = 1.378; P = 0.242), infection rate (χ2(1) = 2.921; P = 0.089), graft rejection (χ2(1) = 0.873; P = 0.352), and mor- tality (χ2(1) = 0.010; P = 0.922). In addition, there were no statistically significant differences in ICU length of stay between groups (χ2(14) = 16.271; P = 0.297), based on the Mann-Whitney test. Multiple linear regression analysis Multiple linear regression was calculated to pre- dict ICU length of stay on SMI and infection. A sig- nificant regression equation was found (F (2. 95) = 11.192, P < 0.000), with R 2 of 0.191. SMI was found to significantly predict length of ICU stay (β = 0.292; 95% CI 0.045, 0.199; P = 0.002). The length of ICU stays increased by 0.122 days per cm 2 /m 2 SMI in male patients with infection. Mortality In our study group, 5 (3.6%) patients died in the first 90 days after liver transplantation. Two pa- tients died immediately after the procedure due to irreversible haemorrhagic shock with dissemi- nated intravascular thrombosis and abdominal organ ischemia. The cause of death in three pa- tients was sepsis with multiorgan failure in one patient (day 66), and severe postoperative bleed- ing, intraabdominal infection with liver abscess, and multiorgan failure in two patients (34 and 53 days). One year survival rate was 95%. Due to the small number of patients, statistical analysis was not performed. Discussion Malnutrition with skeletal muscle mass loss is a frequent complication in patients with chronic or end-stage liver disease. 25 The aetiology of malnu- trition is multifactorial. 23 Insufficient calorie intake (early satiety, loss of appetite, alcohol consump- tion, diet restriction), metabolic abnormalities, cat- abolic state of metabolism, and malabsorption are the main contributors to muscle loss. 25,26 It affects from 30 to 70% 27,28 of patients with end-stage liver disease and up to 80% 27 of patients with alcohol-re- Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 173 lated liver disease. Myosteatosis affects more than half of patients with chronic liver disease. 9 In our patient population, we used selected parameters for low SMI (men < 52.4 cm 2 /m 2 , women < 38 cm 2 / m 2 ) 9,21 and MI (< 33 HU in patients with a BMI ≥ 25 kg/m 2 and < 41 HU in those with a BMI < 25). 22-24 These cut-off values were determined specifically for patients on LT waiting list are the most widely used in literature. 22-24 Low SMI was present in 63% of males and 28.9% of females. More than one- third of the patients (32.6%) had fatty infiltration of the skeletal muscle. Patients with alcoholic aeti- ology had more fat-infiltrated muscles than other aetiologies for end-stage liver disease (P < 0.008). Alterations in the metabolism of fatty acids, eating habits, and alcohol consumption probably result in fatty infiltration of skeletal muscle fibres. However, due to the small numbers of patients (3 patients) with non-alcoholic steatohepatitis (NASH) or non- alcoholic fatty liver disease (NAFLD) in our study group, we cannot properly assess the relationship between the aetiology of underlying liver disease and the degree of MI. Several studies have shown that sarcopenia and myosteatosis are associated with a higher in- cidence of postoperative complications, infection rate, duration of hospitalization, and mortality in a wide range of gastrointestinal cancers, including tumours of the hepatopancreatobiliary system. 29-32 Lack of a strictly defined patient population, dif- ferent methods of measurement of muscle mass and function, and the absence of uniformly ac- cepted cut-off points for patients with chronic or end-stage liver disease are the leading causes for wide ranges of sarcopenia incidence reported in the literature. 14,21,33 Therefore, the selected cut-off points for patients with cancer are used for evalua- tion, which may affect the quality of results. Sarcopenia is associated with poorer outcomes in patients with chronic liver disease on the wait- list or after LT. 34,35 Patients with end-stage liver disease and sarcopenia have shorter survival than non-sarcopenic patients (22 ± 3 vs. 95 ± 22-month, P < 0.001). 9,36 The main cause of death in patients with end-stage liver disease is sepsis. 37 Incidence of sep- sis as a cause of death is even higher in presence of sarcopenia (36% vs. 16%, P < 0.001) or myosteato- sis (32% vs. 19%, P = 0.020). 9 Lower muscle mass is also associated with a higher incidence of hepatic encephalopathy in patients with end-stage liver disease. 38,39 On the other hand, a group from USA 40 showed that sarcopenic patients had a higher ten- dency for pulmonary complications than the non- sarcopenic group (38% vs. 18%, P = 0.100); however, there was no significant difference in morbidity and mortality. CT imaging is considered as the gold standard for body composition assessment. SMI calculated on the level of third lumbar vertebra is superior since it correlates best with the actual quantity of the muscles in the body. 41 It is not affected by as- cites and is part of preoperative evaluation in pa- tients with hepatocellular carcinoma or the com- plications of portal hypertension. The value of SMI as a predicting factor for postoperative morbidity and mortality is yet to be determined. However, it has been shown in several reports that it has a negative impact on postoperative outcomes. Lower values of SMI are associated with more significant postoperative mortality, higher infection risk, and postoperative complications, more extended in- tensive care unit stay, ventilator dependency, and higher waitlist mortality rates in patients with end- stage liver disease. 33,42,43 SMI as a predicting factor for higher mortality rate in patients with end-stage liver disease was superior to other nutritional as- sessment indicators, such as BMI, upper arm mus- cle circumference, and triceps skinfold thickness. 44 However, some other reports showed contrasting findings. Using SMI for prespecified definitions of sarcopenia had no impact on mortality or delisting from the transplant waitlist between patients with and without sarcopenia. 45 Length of hospitaliza- tion following LT, days of hospitalization during the first year post-LT, survival at one year, or over- all survival was not different between sarcopenic and nonsarcopenic patients. 46 Low SMI alone was not associated with graft and patient survival (P = 0.273 and P = 0.278) after LT. 47 These conflicting results are probably due to heterogeneity of used specific cut-off points (sex, age, race) and lack of strictly defined parameters representative unique- ly for patients with chronic liver disease. We found a negative correlation between BMI and SMI, probably due to high-volume ascites in the patient population This confirms findings that BMI is not suitable for body assessment in patients with end- stage liver disease. In our study, the presence of as- cites was associated with significantly lower SMI (P < 0.05) in patients with end-stage liver disease but did not influence MI. There is a weak, positive correlation between SMI and ICU length of stay in male patients, which was statistically significant (rs = 0.226, P < 0.025). We speculate that the main reason is a more pronounced systemic inflamma- tory reaction to surgical stress in patients with preserved muscle mass. It is known that muscle mass is mandatory for the normal function of the Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 174 immune system. 48 Experience from Japan 49 shows a connection between low SMI and decreased incidence of graft rejection in living-donor liver transplantation. Analysis of our results shows no statistically significant difference in postopera- tive complications, rate of postoperative infection, and liver graft rejection rate between males and females with low or normal SMI (P > 0.05). BIA is commonly used technique in body composition analysis in every day clinical practice. 50 Its key pa- rameters are resistance, reactance and phase angle (PA). PA is found to be associated with outcomes in different diseases and has been found to be use- ful for monitoring fluid changes and response to interventions. 50 The main limitation is the com- plexity of the determinants that requires its ad- justment to the individual phenotypic diagnosis of each patient. Results can be affected by altered water and electrolyte balance, fluid retention and diuretic therapy. 50 DXA allows for the quantifica- tion of three body compartments (bone mass, fat mass, and bone fat-free mass (or lean mass)) based upon the differential tissue attenuation of X-ray photons. 51 However, it can be affected by presence of ascites. 52 Even though DXA can be modified to exclude influence of ascites or tissue oedema the correlation between the lean mass index and SMI was weaker (γ = 0.29, p = 0.035) and falsely high in patients with ascites before liver transplantation. 53 Main disadvantages of DXA compared to CT is in- ability to assess muscle mass quality (myosteato- sis). 51 Myosteatosis is a more clearly defined factor, and its influence is more uniformly established in literature. 54 However, there are many different cut off points (HU < 25 to HU < 39) reported in litera- ture in patients with malignant disease. 55 Majority of reports uses a Martin cut-off point (< 33 HU in patients with a BMI ≥ 25 kg/m 2 and < 41 HU in those with a BMI < 25) 56 for determining presence of myosteatosis. Martin cut-off point is also recent- ly defined threshold parameters for MI in a patient with a chronic liver disease. 22-24 It has been shown that severely ill patients with myosteatosis have a lower survival rate than those without fatty infil- tration in muscles. 57 Myosteatosis negatively im- pacts the survival of patients with end-stage liver disease (28 ± 5 vs. 95 ± 22-month, P < 0.001) 9,36 and is associated with longer hospitalization and higher morbidity. 43 Patients with myosteatosis showed a higher mortality rate, most commonly due to res- piratory and septic complication. 47 Myosteatosis had no influence on ICU length of stay (P = 0.161), hospitalisation (P = 0.771), postoperative complica- tions (P = 0.839), infection rate (P = 0.703) and graft rejection (P= 0.449) in our patient population. The nutritional status on the waitlist for LT as a possible risk factor is overlooked with the MELD score. 58 Another important limiting factor of the MELD score is using serum creatinine levels for score calculation. 3 Serum creatinine level may vary significantly and is influenced by chronic kidney disease, ascites, paracentesis, and the in- fluence of gender and liver disease on skeletal muscle mass. 59 Variation of serum creatinine levels may affect the MELD score and underestimate the severity of liver disease. 58 Thus, several modifica- tions of the MELD score were developed to incor- porate the nutritional parameters. Body composi- tion MELD (BC-MELD) 60 has a better predictive value for waiting list mortality than MELD score. MELD-sarcopenia score showed a positive predic- tive value in patients with a lower score (< 15) on the postoperative course; however, it was not use- ful in patients with MELD above 15. 61,62 Combining SMI in MELD in multivariate analysis (AUROC= 0.812) is significantly better than MELD alone (AUROC = 0.787) for predicting 5-year mortality (P < 0.001). 44 We found a weak, positive correlation between the MELD score and ICU length of stay (rs = 0.261, P < 0.002). ALBI score showed promis- ing results in more accurate prediction of liver dis- ease severity and mortality on waitlist compared to the Child-Pugh score; however, its predictive value was inferior to the MELD score. 63 Patients with pre-transplant ALBI grade 3 liver disease had increased mortality after LT. 64 We found a weak, positive correlation between the ALBI score and ICU length of stay and hospitalization time, which was statistically significant (rs = 0.279, P < 0.001; rs = 0.197; P = 0.022). We found no correlation between the ALBI score and postoperative complications. The GLIM score 16 was established as a potential assessment tool for evaluating patient nutritional status in recent years. Our results showed no dif- ference between the patients who met GLIM cri- teria and those who did not, regarding ICU stay, length of hospitalization, postoperative complica- tions, infection, and mortality. We believe that nutritional status in a patient with end-stage liver disease is an essential aspect of pre-transplant workup. However, its precise role is not yet determined. LT is a complex surgi- cal procedure influenced by numerous factors. In the early phase of LT, donor characteristics and comorbidities, quality of liver graft retrieval, liver graft quality, cold preservation duration, ischemic damage to the liver graft during static cold storage, Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 175 recipient medical conditions and comorbidities, surgical procedure, and early postoperative ther- apy are by our opinion the most essential factors. However, muscle mass status, muscle function and myosteatosis are crucial factors in a period of rehabilitation. 65 Patient with cachexia or with high-risk (< 18.5 kg/ m 2 , Child-Pough C) 66 to lose muscle mass should be screened and involved in intensive but person- alized nutritional support therapy. Nutritional re- quirements in end stage liver disease are 35 kcal/ kg per day in non-obese patients (BMI 30 kg/m 2 ) and 1,2g/kg per day intake of proteins. 66 It is safe to calculate requirements based on the dry weight of a patients. 66 In presence of hepatic encephalopa- thy (HE), animal protein should be substituted with vegetable protein origin. Randomized control study 67 showed that substitution of animal protein with vegetable protein for a period of six months (30–35 kcal/kg/d,1.0–1.5 g/kg/d protein) improved neuropsychiatric performance in patients with minimal HE and decrease their risk of develop- ing overt HE compared to no intervention. It is unreal to expect to gain muscle mass in cirrhotic patients, but muscle mass preservation should be focus of such nutritional interventions. There are several strategies to prevent muscle mass loss in patients with end stage liver disease. First strategy is nutritional supplementations. Patients should have frequent small meals to avoid prolong fast- ing period (> 6 h). 66,68,69 Enteral supplements with side branched amino acids should be adminis- tered. 66,68,69 Snacks rich in carbohydrates should be taken as a late-night snack. 66,68,69 Second, physi- cal activity in a form of resistance and endurance exercise, is probably appropriate and beneficial. 70 Micronutrition, especially administration of fat- soluble vitamins 71 and ammonia lowering thera- py 72 is also important. Role of immunonutrition in patient with end stage liver disease is not yet estab- lished. 66,73 Post-transplant screening is advised in all patients after liver transplantation. 66,73 There are several drawbacks to our study. The two most important are the retrospective nature of data collection and the lack of functional assess- ment of recipients’ muscles. Another factor that may influence results is the relatively small num- ber of cases that may affect some results or may not produce statistical significance due to the com- plexity of the treatment and numerous factors that determine its outcome. In conclusion radiological assessment of a pa- tient’s nutritional status at the third lumbar ver- tebra represents an objective and reproducible method. It should become a standard screening tool in patients with acute or chronic end-stage liver disease. Due to complexity of liver transplant procedure, liver graft and liver recipients’ factors, it is difficult to established impact of a skeletal muscle index and myosteatosis on postoperative outcomes. However, nutritional interventions and physical activity should be part of the clinical pathway in patients with end-stage liver disease waiting for liver transplantation. Prospective ran- domized controlled studies are not possible due to ethical considerations; hence, standardization and uniformity in definitions, methods, and cut- off points are crucial to producing reliable data in the future. Acknowledgments Authors would like to acknowledge all partici- pants in liver transplant programme in our insti- tution for their dedicated work. References 1. Starzl TE, Marchioro TL, Vonkaulla KN, Hermann G, Brittain RS, Waddell WR. Homotransplantation of the liver in humans. Surg Gynecol Obstet 1963; 117: 659-76. PMID: 14100514 2. Song AT , Avelino-Silva VI, Pecora RA, Pugliese V, D’ Albuquerque LA, Abdala E. Liver transplantation: fifty years of experience. World J Gastroenterol 2014; 20: 5363-74. doi: 10.3748/wjg.v20.i18.5363 3. Polyak A, Kuo A, Sundaram V. Evolution of liver transplant organ allocation policy: current limitations and future directions. World J Hepatol 2021; 13: 830-9. doi: 10.4254/with.v13.i8.830 4. Deng M, Ng SWY, Cheung ST, Chong CCN. Clinical application of Albumin- Bilirubin (ALBI) scores: the current status. Surgeon 2020; 18: 178-86. doi: 10.1016/j.surge.2019.09.002 5. Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing 2019; 48: 16-31. doi: 10.1093/ageing/afy169 6. Reginster JY, Beaudart C, Al-Daghri N, Avouac B, Bauer J, Bere N, et al. Update on the ESCEO recommendation for the conduct of clinical trials for drugs aiming at the treatment of sarcopenia in older adults. Aging Clin Exp Res 2021; 33: 3-17. doi: 10.1007/s40520-020-01663-4 7. Delmonico MJ, Harris TB, Visser M, Park SW, Conroy MB, Velasquez-Mieyer P, et al. Health, aging, and body. Longitudinal study of muscle strength, quality, and adipose tissue infiltration. Am J Clin Nutr 2009; 90: 1579-85. doi: 10.3945/ajcn.2009.28047 8. Miljkovic I, Kuipers AL, Cvejkus R, Bunker CH, Patrick AL, Gordon CL, el al. Myosteatosis increases with aging and is associated with incident diabetes in African ancestry men. Obesity 2016; 24: 476-82. doi: 10.1002/oby.21328 9. Montano-Loza AJ, Angulo P, Meza-Junco J, Prado CM, Sawyer MB, Beaumont C, et al. Sarcopenic obesity and myosteatosis are associated with higher mortality in patients with cirrhosis. J Cachexia Sarcopenia Muscle 2015; 7: 126-35. doi: 10.1002/jcsm.12039 10. Goodpaster BH, Theriault R, Watkins SC, Kelley DE. Intramuscular lipid con- tent is increased in obesity and decreased by weight loss. Metabolism 2000; 49: 467-72. doi: 10.1016/s0026-0495(00)80010-4 Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 176 11. Lee CM, Kang J. Prognostic impact of myosteatosis in patients with colorec- tal cancer: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 2020; 11: 1270-82. doi: 10.1002/jcsm.12575 12. Srpcic M, Jordan T , Popuri K, Sok M. Sarcopenia and myosteatosis at presen- tation adversely affect survival after esophagectomy for esophageal cancer. Radiol Oncol 2020; 54: 237-46. doi: 10.2478/raon-2020-0016 13. Espinosa-Flores AJ, Guzman-Ortiz E, Melendez-Mier G, Ternovoy SK, Bueno- Hernandez N, Roldan-Valadez E. A scoping review of the methods used in patients with liver cirrhosis to assess body composition and their nutritional findings. [Ahead of print]. Eur J Clin Nutr 2023. doi:10.1038/s41430-023- 01287-7 14. Lee CM, Kang BK, Kim M. Radiologic definition of sarcopenia in chronic liver disease. Life (Basel) 2021; 11: 86. doi: 10.3390/life11020086 15. Kjønigsen LJ, Harneshaug M, Fløtten AM, Karterud LK, Petterson K, Skjolde G, et al. Reproducibility of semiautomated body composition segmentation of abdominal computed tomography: a multiobserver study. Eur Radiol Exp 2019; 3: 42. doi: 10.1186/s41747-019-0122-5 16. Jensen GL, Cederholm T, Correia MITD, Gonzalez MC, Fukushima R, Higashiguchi T, et al. GLIM criteria for the diagnosis of malnutrition: a con- sensus report from the global clinical nutrition community. JPEN J Parenter Enteral Nutr 2019; 43: 32-40. doi: 10.1002/jpen.1440 17. Song HN, Wang WB, Luo X, Huang DD, Ruan XJ, Xing CG, et al. Effect of GLIM-defined malnutrition on postoperative clinical outcomes in patients with colorectal cancer. Jpn J Clin Oncol 2022; 52: 466-74. doi: 10.1093/ jjco/hyab215. 18. Dindo D, Demartines N, Clavien PA. Classification of surgical complica- tions: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg 2004; 240: 205-13. doi: 10.1097/01. sla.0000133083.54934.ae 19. Venturi C, Sempoux C, Bueno J, Ferreres Pinas JC, Bourdeaux C, Abarca- Quinones J, et al. Novel histologic scoring system for long-term allograft fibrosis after liver transplantation in children. Am J Transplant 2012; 12: 2986-96. doi: 10.1111/j.1600-6143.2012.04210.x 20. Popuri K, Cobzas D, Esfandiari N, Baracos V, Jagersand M. Body composition assessment in axial CT images using FEM-based automatic segmentation of skeletal muscle. IEEE Trans Med Imaging 2016; 35: 512-20. doi: 10.1109/ TMI.2015.2479252 21. van Vugt JL, Levolger S, de Bruin RW, van Rosmalen J, Metselaar HJ, IJzermans JN. Systematic review and meta-analysis of the impact of com- puted tomography-assessed skeletal muscle mass on outcome in patients awaiting or undergoing liver transplantation. Am J Transplant 2016; 16: 2277-92. doi: 10.1111/ajt.13732 22. Ebadi M, Tsien C, Bhanji RA, Dunichand-Hoedl AR, Rider E, Motamedrad M, et al. Myosteatosis in cirrhosis: a review of diagnosis, pathophysiological mechanisms and potential interventions. Cells 2022; 11: 1216. doi: 10.3390/ cells11071216 23. Eslamparast T , Montano-Loza AJ, Raman M, Tandon P . Sarcopenic obesity in cirrhosis − the confluence of 2 prognostic titans. Liver Int 2018; 38: 1706-17. doi: 10.1111/liv.13876 24. Czigany Z, Kramp W, Bednarsch J, van der Kroft G, Boecker J, Strnad P , et al. Myosteatosis to predict inferior perioperative outcome in patients undergo- ing orthotopic liver transplantation. Am J Transplant 2020; 20: 493-503. doi: 10.1111/ajt.15577 25. Dasarathy S, Merli M. Sarcopenia from mechanism to diagnosis and treatment in liver disease. J Hepatol 2016; 65: 1232-44. doi: 10.1016/j. jhep.2016.07.040 26. Ebadi M, Bhanji RA, Mazurak VC, Montano-Loza AJ. Sarcopenia in cirrhosis: from pathogenesis to interventions. J Gastroenterol 2019; 54: 845-59. doi:10.1007/s00535-019-01605-6 27. Montano-Loza AJ. Skeletal muscle abnormalities and outcomes after liver transplantation. Liver Transpl 2014; 20: 1293-95. doi: 10.1002/lt.23995 28. Kim HY , Jang JW. Sarcopenia in the prognosis of cirrhosis: going beyond the MELD score. World J Gastroenterol 2015; 21: 7637-47. doi: 10.3748/wjg. v21.i25.7637 29. Levolger S, van Vugt JLA, de Bruin RWF , IJzermans JNM. Systematic review of sarcopenia in patients operated on for gastrointestinal and hepatopancrea- tobiliary malignancies. Br J Surg 2015, 102: 1448-58. doi: 10.1002/bjs.9893 30. Cao Q, Xiong Y , Zhong Z, Ye Q. Computed tomography-assessed sarcopenia indexes predict major complications following surgery for hepatopancrea- tobiliary malignancy: a meta-analysis. Ann Nutr Metab 2019; 74: 24-34. doi: 10.1159/000494887 31. Kaibori M, Ishizaki M, Iida H, Matsui K, Sakaguchi T, Inoue K, et al. Effect of intramuscular adipose tissue content on prognosis in patients undergoing hepatocellular carcinoma resection. J Gastrointest Surg 2015; 19: 1315-23. doi: 10.1007/s11605-015-2838-8 32. Ooi PH, Hager A, Mazurak VC, Dajani K, Bhargava R, Gilmour SM, et al. Sarcopenia in chronic liver disease: impact on outcomes. Liver Transpl 2019; 25: 1422-38. doi: 10.1002/lt.25591 33. Weerink LBM, van der Hoorn A, van Leeuwen BL, de Bock GH. Low skeletal muscle mass and postoperative morbidity in surgical oncology: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 2020; 11: 636-49. doi: 10.1002/jcsm.12529 34. Hsu CS, Kao JH. Sarcopenia and chronic liver diseases. Expert Rev Gastroenterol Hepatol 2018; 12: 1229-44. doi: 10.1080/17474124.2018.1534586 35. Pinto Dos Santos D, Kloeckner R, Koch S, Hoppe-Lotichius M, Zöller D, T oenges G, et al. Sarcopenia as prognostic factor for survival after orthotopic liver transplantation. Eur J Gastroenterol Hepatol 2020; 32: 626-34. doi: 10.1097/MEG.0000000000001552 36. Montano-Loza AJ, Meza-Junco J, Prado CM, Lieffers JR, Baracos VE, Bain VG, et al. Muscle wasting is associated with mortality in patients with cir- rhosis. Clin Gastroenterol Hepatol 2012; 10: 166-73, 173.e1. doi: 10.1016/j. cgh.2011.08.028 37. Lucidi C, Lattanzi B, Di Gregorio V, Incicco S, D’ Ambrosio D, Venditti M, et al. A low muscle mass increases mortality in compensated cirrhotic patients with sepsis. Liver Int 2018; 38: 851-7. doi: 10.1111/liv.13691 38. Chang KV, Chen JD, Wu WT, Huang KC, Lin HY , Han DS. Is sarcopenia associ- ated with hepatic encephalopathy in liver cirrhosis? A systematic review and meta-analysis. J Formos Med Assoc 2019; 118: 833-42. doi: 10.1016/j. jfma.2018.09.011 39. Lattanzi B, D’ Ambrosio D, Merli M. Hepatic encephalopathy and sarcopenia: two faces of the same metabolic alteration. J Clin Exp Hepatol 2019; 9: 125- 13. doi: 10.1016/j.jceh.2018.04.007 40. Dhaliwal A, Larson D, Hiat M, Muinov LM, Harrison WL, Sayles H, et al. Impact of sarcopenia on mortality in patients undergoing liver re-trans- plantation. World J Hepatol 2020; 12: 807-15. doi: 10.4254/wjh.v12.i10.807 41. Lee CM, Kang BK, Kim M. Radiologic definition of sarcopenia in chronic liver disease. Life 2021; 11: 86. doi: 10.3390/life11020086 42. Meeks AC, Madill J. Sarcopenia in liver transplantation: a review. Clin Nutr ESPEN 2017; 22: 76-80. doi: 10.1016/j.clnesp.2017.08.005 43. van Vugt JL, Levolger S, de Bruin RW, van Rosmalen J, Metselaar HJ, IJzermans JN. Systematic review and meta-analysis of the impact of com- puted tomography-assessed skeletal muscle mass on outcome in patients awaiting or undergoing liver transplantation. Am J Transplant 2016; 16: 2277-92. doi: 10.1111/ajt.13732 44. Yao J, Zhou X, Yuan L, Niu LY, Zhang A, Shi H, et al. Prognostic value of the third lumbar skeletal muscle mass index in patients with liver cirrhosis and ascites. Clin Nutr 2020; 39: 1908-13. doi: 10.1016/j.clnu.2019.08.006 45. Kappus MR, Wegermann K, Bozdogan E, Patel YA, Janas G, Shropshire E, et al. Use of skeletal muscle index as a predictor of wait-list mortality in patients with end-stage liver disease. Liver Transpl 2020; 26: 1090-9. doi: 10.1002/lt.25802 46. Aby ES, Lee E, Saggi SS, Viramontes MR, Grotts JF, Agopian VG, et al. Pretransplant sarcopenia in patients with NASH cirrhosis does not impact rehospitalization or mortality. J Clin Gastroenterol 2019; 53: 680-5. doi: 10.1097/MCG.0000000000001109 47. Czigany Z, Kramp W, Lurje I, Miller H, Bednarsch J, Lang SA, et al. The role of recipient myosteatosis in graft and patient survival after deceased donor liver transplantation. J Cachexia Sarcopenia Muscle 2021; 12: 358-67. doi: 10.1002/jcsm.12669 48. Nelke C, Dziewas R, Minnerup J, Meuth SG, Ruck T . Skeletal muscle as poten- tial central link between sarcopenia and immune senescence. EBioMedicine 2019; 49: 381-8. doi: 10.1016/j.ebiom.2019.10.034 Radiol Oncol 2023; 57(2): 168-177. Petric M et al. / Radiological assessment of skeletal muscle index and myosteatosis 177 49. Wakabayashi T, Shinoda M, Obara H, Kitago M, Yagi H, Abe Y, et al. Decreased incidence of acute cellular rejection in low-muscle-mass re- cipients after living-donor liver transplantation. Transplant Proc 2018; 50: 3626-34. doi: 10.1016/j.transproceed.2018.06.028 50. Bellido D, García-García C, Talluri A, Lukaski HC, García-Almeida JM. Future lines of research on phase angle: strengths and limitations. Rev Endocr Metab Disord 2023; 24: 563-83. doi: 10.1007/s11154-023-09803-7 51. Lee CM, Kang BK, Kim M. Radiologic definition of sarcopenia in chronic liver disease. Life 2021; 11: 86. doi: 10.3390/life11020086 52. Kallwitz ER. Sarcopenia and liver transplant: the relevance of too little muscle mass. World J Gastroenterol 2015; 21: 10982-93. doi: 10.3748/wjg. v21.i39.10982 53. Lindqvist C, Brismar TB, Majeed A, Wahlin S. Assessment of muscle mass depletion in chronic liver disease: dual-energy x-ray absorptiometry com- pared with computed tomography. Nutrition 2019; 61: 93-8. doi: 10.1016/j. nut.2018.10.031 54. Shenvi SD, Taber DJ, Hardie AD, Botstein JO, McGillicuddy JW . Assessment of magnetic resonance imaging derived fat fraction as a sensitive and reliable predictor of myosteatosis in liver transplant recipients. HPB (Oxford) 2020; 22: 102-8. doi: 10.1016/j.hpb.2019.06.006 55. Ahn H, Kim DW, Ko Y, Ha J, Shin YB, Lee J, et al. Updated systematic review and meta-analysis on diagnostic issues and the prognostic impact of my- osteatosis: a new paradigm beyond sarcopenia. Ageing Res Rev 2021; 70: 101398. doi: 10.1016/j.arr.2021.101398 56. Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, et al. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol 2013; 31:1539-47. doi: 10.1200/JCO.2012.45.2722 57. Loosen SH, Schulze-Hagen M, Püngel T, Bündgens L, Wirtz T, Kather JN, et al. Skeletal muscle composition predicts outcome in critically Ill patients. Crit Care Explor 2020; 2: e0171. doi: 10.1097/CCE.0000000000000171 58. Kim HY , Jang JW. Sarcopenia in the prognosis of cirrhosis: going beyond the MELD score. World J Gastroenterol 2015; 21: 7637-47. doi: 10.3748/wjg. v21.i25.7637 59. Francoz C, Prié D, Abdelrazek W, Moreau R, Mandot A, Belghiti J, et al. Inaccuracies of creatinine and creatinine-based equations in candidates for liver transplantation with low creatinine: impact on the model for end-stage liver disease score. Liver Transpl 2010; 16: 1169-77. doi: 10.1002/lt.22128 60. Hamaguchi Y, Kaido T, Okumura S, Kobayashi A, Shirai H, Yao S, et al. Including body composition in MELD scores improves mortality prediction among patients awaiting liver transplantation. Clin Nutr 2020; 39: 1885-92. doi: 10.1016/j.clnu.2019.08.012 61. Montano-Loza AJ, Duarte-Rojo A, Meza-Junco J, Baracos VE, Sawyer MB, Pang JX, et al. Inclusion of sarcopenia within MELD (MELD-Sarcopenia) and the prediction of mortality in patients with cirrhosis. Clin Transl Gastroenterol 2015; 6: e102. doi: 10.1038/ctg.2015.31 62. van Vugt JLA, Alferink LJM, Buettner S, Gaspersz MP , Bot D, Darwish Murad S, et al. A model including sarcopenia surpasses the MELD score in predict- ing waiting list mortality in cirrhotic liver transplant candidates: a competing risk analysis in a national cohort. J Hepatol 2018; 68: 707-14. doi: 10.1016/j. jhep.2017.11.030 63. Chedid MF, Picon RV, Chedid AD. ALBI and PALBI: novel scores for outcome prediction of cirrhotic outpatients awaiting liver transplantation. Ann Hepatol 2018; 17: 906-7. doi: 10.5604/01.3001.0012.7190 64. Bernardi N, Chedid MF, Grezzana-Filho TJM, Chedid AD, Pinto MA, Leipnitz I, et al. Pre-transplant ALBI grade 3 is associated with increased mortality after liver transplantation. Dig Dis Sci 2019; 64: 1695-704. doi: 10.1007/ s10620-019-5456-6 65. Miarka M, Gibiński K, Janik MK, Główczyńska R, Zając K, Pacho R, et al. Sarcopenia-the impact on physical capacity of liver transplant patients. Life 2021; 11: 740. doi: 10.3390/life11080740. 66. European Association for the Study of the Liver. EASL Clinical Practice Guidelines on nutrition in chronic liver disease. J Hepatol 2019; 70: 172-93. doi: 10.1016/j.jhep.2018.06.024 67. Maharshi S, Sharma BC, Sachdeva S, Srivastava S, Sharma P. Efficacy of nutritional therapy for patients with cirrhosis and minimal hepatic encepha- lopathy in a randomized trial. Clin Gastroenterol Hepatol 2016; 14: 454-60. e453. doi: 10.1016/j.cgh.2015.09.028 68. Ebadi M, Bhanji RA, Mazurak VC, Montano-Loza AJ. Sarcopenia in cirrhosis: from pathogenesis to interventions. J Gastroenterol 2019; 54: 845-59. doi: 10.1007/s00535-019-01605-6 69. Dasarathy S, Merli M. Sarcopenia from mechanism to diagnosis and treatment in liver disease. J Hepatol 2016; 65: 1232-44. doi: 10.1016/j. jhep.2016.07.040 70. Berzigotti A, Albillos A, Villanueva C, Genesca J, Ardevol A, Augustin S, et al. Effects of an intensive lifestyle intervention program on portal hypertension in patients with cirrhosis and obesity: the SportDiet study. Hepatology 2017; 65: 1293-305. doi: 10.1002/hep.28992 71. Stokes CS, Volmer DA, Grunhage F, Lammert F. Vitamin D in chronic liver disease. Liver Int 2013; 33: 338-52. doi: 10.1111/liv.12106 72. Miarka M, Gibiński K, Janik MK, Główczyńska R, Zając K, Pacho R, et al. Sarcopenia-the Impact on physical capacity of liver transplant patients. Life 2021; 11: 740. doi: 10.3390/life11080740 73. Lei Q, Wang X, Zheng H, Bi J, Tan S, Li N. Peri-operative immunonutrition in patients undergoing liver transplantation: a meta-analysis of randomized controlled trials. Asia Pac J Clin Nutr 2015; 24: 583-90. doi: 10.6133/ap- jcn.2015.24.4.20.