Sarcopenia in cirrhosis: a clinical practice review
Review Article | Palliative Medicine and Palliative Care for Serious or Advanced Diseases

Sarcopenia in cirrhosis: a clinical practice review

Ussama Ghumman1, Brian Lee1, Dakota Bigham1, Eugenia Tsai2,3

1Department of Internal Medicine, UT Health San Antonio, San Antonio, TX, USA; 2Department of Hepatology, Texas Liver Institute, San Antonio, TX, USA; 3Division of Gastroenterology, UT Health San Antonio, San Antonio, TX, USA

Contributions: (I) Conception and design: E Tsai, U Ghumman; (II) Administrative support: E Tsai; (III) Provision of study materials or patients: U Ghumman, E Tsai; (IV) Collection and assembly of data: U Ghumman, E Tsai; (V) Data analysis and interpretation: U Ghumman, E Tsai, Brian Lee; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Eugenia Tsai, MD. Department of Hepatology, Texas Liver Institute, 607 Camden Street, San Antonio, TX 78215, USA; Division of Gastroenterology, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA. Email: tsaie@uthscsa.edu.

Abstract: Cirrhosis, a leading cause of death in the United States, is a result of chronic liver injury leading to progressive liver fibrosis. Initially asymptomatic, cirrhosis progresses to decompensated forms characterized by jaundice, ascites, gastroesophageal variceal bleeding, and hepatic encephalopathy (HE). Malnutrition, frailty, and sarcopenia are prevalent comorbidities in cirrhosis patients and are often used interchangeably in the clinical setting. Malnutrition is a condition marked by imbalanced nutrient intake and is closely related to the development of frailty and sarcopenia. Frailty is characterized by the decline in physiological reserve and function, while sarcopenia is the generalized loss of skeletal mass. Both are insidious complications of cirrhosis and also significantly influence morbidity, mortality, and transplant outcomes. Major contributing factors include decreased oral intake, poor nutrient uptake and deranged metabolism. Accurate assessment of frailty and sarcopenia in patients with cirrhosis is essential for predicting clinical outcomes. Interventions targeting frailty and sarcopenia could significantly improve patient prognosis. Nutritional interventions and physical activity promote muscle protein synthesis, increase muscle mass and mitigate sarcopenia. A cirrhosis-specific treatment includes ammonia-lowering agents to improve cognitive function and ultimately oral intake, and increase muscle mass. Emerging therapies, such as including L-ornithine L-aspartate, leucine-enriched branch-chained amino acids, hold promise in modulating skeletal muscle. This review explores the definitions, clinical manifestations, and consequences of malnutrition, frailty, and sarcopenia in cirrhosis, emphasizing the importance of early assessment and intervention.

Keywords: Sarcopenia; cirrhosis; frailty; malnutrition; muscle


Submitted Dec 19, 2024. Accepted for publication Apr 18, 2025. Published online May 27, 2025.

doi: 10.21037/apm-24-173


Introduction

Cirrhosis is the end stage of any chronic liver disease and globally accounts for two million deaths annually (1). The disease process is the result of chronic liver injury leading to progressive scarring, or fibrosis. Etiologies include chronic alcohol use, metabolic dysfunction associated steatotic liver disease (MASLD)—formerly known as non-alcoholic fatty liver disease (NAFLD), hepatitis B or C virus infection, genetic conditions and autoimmune diseases. Liver function is initially preserved; however, if left untreated, ongoing fibrosis leads to disarray in hepatic architecture and nodule formation with gradual impairment of hepatic function (2).

Compensated cirrhosis is primarily asymptomatic, without significant biochemical or anatomic complications. Decompensated cirrhosis is characterized by the deterioration in liver function with the development of jaundice, esophageal variceal bleeding, hepatic encephalopathy (HE) and ascites (3). The Child-Pugh scoring system is a tool initially designed to predict mortality and categorize the severity of portal hypertension. The five components of the Child-Pugh score include subjective parameters along with objective laboratory data indicating liver function: (I) degree of HE, (II) severity of ascites, (III) bilirubin, (IV) albumin and (V) prothrombin time, which is often substituted by international normalized ratio (INR) (4). Cirrhosis is one of the most significant predisposing factors in developing malnutrition, frailty, and sarcopenia (5). These conditions directly correlate to morbidity, mortality, and overall poorer outcomes in patients with end-stage liver disease (ESLD) and liver transplant recipients (6). In this review article, we highlight the current understanding of malnutrition, frailty, and sarcopenia with a focus on assessments and interventions in the setting of cirrhosis.


Malnutrition

In the clinical setting, the terms malnutrition, frailty and sarcopenia are often used interchangeably. However, frailty and sarcopenia are distinct clinical manifestations of malnutrition. A corollary to this ideology is in patients with metabolic syndrome and diabetes who develop physical manifestations of insulin resistance, such as acanthosis nigricans.

Malnutrition is a clinical syndrome related to the imbalance (deficiency or excess) of nutrients and impaired nutrient utilization that is associated with measurable adverse effects on function and clinical outcome (7,8). The reported prevalence of malnutrition in patients with cirrhosis ranges from 50% to 90% and increases with the severity of disease (9-11). Malnutrition is one of the most impactful independent prognostic factors in cirrhosis outcomes, hospital length of stay and mortality (9). In a cross-sectional study evaluating nutritional status, major complications such as uncontrolled ascites, HE, spontaneous bacterial peritonitis (SBP), and hepatorenal syndrome (HRS) developed in 65.5% of malnourished patients vs. 11.8% of well-nourished ones (P<0.05) (12). In a prospective study of 212 hospitalized patients with cirrhosis who were followed clinically for two years or until death, the prevalence of severe malnutrition in those with Child-Pugh class A, B and C were 12.5% (P<0.001), 35.2% (P<0.01) and 45.5%, respectively (13). Malnutrition is highly prevalent amongst patients on the waiting list for liver transplantation (14). By not meeting nutritional requirements, the patients are in a vicious cycle of progressive cirrhosis and deranged nutritional status.

The etiology for malnutrition is multifactorial, but reduced oral intake plays a significant role. Illness, dietary restrictions, fasting periods, hospitalization and symptoms of cirrhosis, including anorexia, nausea, and vomiting all have an impact on malnutrition. Decompensations further exacerbate poor daily food intake. In those with ascites, compression of the stomach leads to loss of appetite and early satiety. In those with HE, preparing meals or consuming food is often not feasible. Nutritional intake in patients with cirrhosis is characterized by low overall energy, protein intake and inadequate food composition (15). Limited access to food due to socioeconomic factors and selection of nutritious foods due to low health literacy also play a role in inadequate macro- and micronutrient intake (16).

Chronic liver disease and inflammation affect impaired macro- and micronutrients. In alcohol-associated liver disease, common deficiencies include folate, thiamine, zinc, selenium, vitamin D and vitamin E (17). Fat-soluble vitamin (A, D and E) deficiencies are commonly seen in cholestatic liver diseases due to the disruption of enterohepatic circulation of bile salts leading to maldigestion and malabsorption (18). Vitamin D directly results in decreased muscle contractility leading to frailty and sarcopenia (19). In a study of 33 patients with chronic liver disease who were treated with oral vitamin D3 at a dose of 2,000 IU once a daily for 12 months compared to control, the prevalence of sarcopenia decreased from 80.0% to 33.3% (P=0.025) demonstrating vitamin D restores skeletal muscle volume and strength (20). MASLD pathophysiology is linked to metabolic risk factors including elevated fasting plasma glucose, hypertriglyceridemia and obesity, which increases risk for sarcopenic obesity (SO).

The liver plays a key role in nutritional metabolism. The development of cirrhosis leads to an increased catabolic state. Glucose uptake in both hepatic and skeletal tissue results in impaired glucose homeostasis (21). Hepatic glucose metabolism is thus altered. Net hepatic glycogenolysis is 3.5 fold lower (P=0.01) and gluconeogenesis is markedly higher (P=0.03) in patients with cirrhosis when compared with no cirrhosis (22). Imbalances in synthesis and storage result in a state of accelerated starvation and increased protein breakdown. The inappropriate dependency on gluconeogenesis diverts amino acids from skeletal muscle exacerbating sarcopenia (23,24).

Altered protein metabolism leading to reduction of plasma free branched-chain amino acids (BCAA) affects extrahepatic ammonia detoxification and results in accelerated muscle breakdown (25). Approximately 33% of patients with cirrhosis develop a hypercatabolic state in which proteolysis outpaces protein synthesis (26). Resting energy expenditure (REE) is also increased in patients with cirrhosis. Hypermetabolism, the state of increased energy expenditure, is prevalent in patients with cirrhosis requiring higher caloric intake compared to the general population (26).

Protein homeostasis is further disturbed in cirrhosis due to factors such as hyperammonemia. Ammonia is derived from the breakdown of proteins and under normal physiologic conditions, the liver plays a central role in ammonia detoxification. Cirrhosis and portosystemic shunting result in inadequate ammonia detoxification and hyperammonemia results in ammonia toxicity, most notably neurotoxicity (24). Hyperammonemia can lead to increased mitochondrial free radical production, causing abnormal oxidative stress and autophagy of skeletal muscle, leading to loss of muscle mass/sarcopenia (27,28). Increased ammonia levels crossing the blood-brain barrier can result in HE, a common neuropsychiatric complication of cirrhosis marked by confusion, psychiatric abnormalities, memory loss and in severe situations, coma. Skeletal muscle plays a role in extrahepatic ammonia detoxification thus decreased muscle mass reciprocally causes hyperammonemia perpetuating a negatively symbiotic relationship (27).

Gastrointestinal dysfunction in cirrhosis can be either anatomical in the setting of tense ascites negatively impacting meal-induced accommodation of the stomach or functional as cirrhosis has been shown to prolong gastric emptying and small bowel transit (29-31). Dysbiosis, characterized by an imbalance in the gut microbiota, leads to an increased intestinal permeability and bacterial translocation (32). This process is driven by the disruption of the intestinal barrier, which allows bacteria and their products to enter the systemic circulation. Systemic inflammation is thus further promoted, decreasing micronutrient production by the gut microbiota and affecting muscle mass and metabolism (33,34). Additionally, portosystemic shunting, pancreatic enzyme deficiency, bacterial overgrowth, and altered intestinal flora contribute to malabsorption. Dysbiosis in cirrhosis is also associated with small intestinal bacterial overgrowth (SIBO), which can impair nutrient absorption and digestion, contributing to malnutrition (35).

Hormonal and cytokine abnormalities also contribute to malnutrition. Cytokines, specifically tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6), along with “hepatokines”, which are liver-specific cytokines, perpetuate the inflammatory process, hepatocyte necrosis and progressive fibrosis (24,36). The pathophysiology of malnutrition in cirrhosis involves several interrelated mechanisms, including that ultimately lead to a catastrophic result (Figure 1).

Figure 1 Contributing factors in cirrhosis leading to frailty and sarcopenia.

Frailty and sarcopenia

Frailty is the clinical state of increased vulnerability to health stressors in the setting of decreased physiologic reserve that leads to adverse health outcomes (7,37,38). Phenotypically, frailty is represented by impaired muscle contractile function. The prevalence of frailty in cirrhosis is 17–43% in the ambulatory setting and 38% in the inpatient setting with concomitant HE (5,39). Physical frailty is the primary manifestation of impaired muscle contraction; this has multiple implications, including disability, decreased physical function, and decreased functional performance (40). This is especially evident in cirrhosis patients as it impairs their overall physical function/performance and compounds on their baseline nutritional and muscular pathophysiology. Progressive immobility, often in the setting of worsening volume overload, episodic or uncontrolled HE and periods of hospitalization for acute decompensations contribute to frailty in patients with cirrhosis. A bidirectional relationship whereby factors that occur with decompensation and acute on chronic liver failure, such as anorexia, ascites and inflammatory cytokine release result in physical inactivity and reduced caloric intake.

Sarcopenia was initially used to describe the loss of muscle in the elderly population; however, it is now acknowledged in an array of chronic diseases, including cirrhosis (1). Classically defined as the age-related, progressive decline in skeletal muscle mass and function, sarcopenia is seen in 40–70% of cirrhosis patients, with severity and prevalence increasing in worsening liver disease (13). Sarcopenia is intrinsic to the aging process but is accelerated by factors such as inflammation, chronic illness, inactivity, and malnutrition. In a prospective study of 1,200 patients, it was determined that age >60 negatively impacted the skeletal muscle index (SMI) (P<0.001) (6). Sarcopenia appears to be more common in men than women with cirrhosis and is associated with an approximately 2-fold higher risk of death (41,42). In a meta-analysis of 8,945 patients with cirrhosis, sarcopenia emerged as a significant risk factor for mortality [hazard ratio (HR) 2.13, 95% confidence interval (CI): 1.86–2.44, P<0.001] (43).

SO is the co-existence of excess adiposity, or obesity [body mass index (BMI) >30 kg/m2], and decreased skeletal muscle mass and function. Prevalence of SO has been reported as high as 42% in patients with cirrhosis (44). Obesity can independently lead to sarcopenia due to the negative impact of adipose tissue-dependent metabolic derangements including oxidative stress, inflammation and insulin resistance (45). Obesity can also mask muscle wasting in patients with cirrhosis thus sarcopenia can go underrecognized in the absence of measures of physical function. The landscape of chronic liver disease has shifted dramatically in recent years with a notable increase in the incidence of MASLD and patients with liver disease are presenting with obesity. Obesity and insulin resistance are closely associated factors with metabolic syndrome, so SO is often seen in patients with cirrhosis due to MASLD. Like sarcopenia, SO is associated with high morbidity and mortality.


Assessment of frailty and sarcopenia

Early detection to allow for immediate intervention is imperative to improve the overall prognosis in patients with disease-related frailty and sarcopenia due to liver disease. Clinicians often utilize subjective frailty measurements in clinical decision-making. The Eyeball test, an assessment in which the physician makes a determination based on patient history and exam, the clinician’s knowledge of comorbidities and intuition can be an accurate method to rule out frailty. The Eyeball test was compared against the Fried test in 300 patients with cardiovascular disease and identified 41% (compared to 36%) of patients as frail with 86% sensitivity and 82% specificity, area under the curve (AUC) 0.82 for diagnosing frailty (46). Subjective assessments including frailty surveys and self-reported physical activity levels inherently lack objectivity, consistency, reproducibility, and meaningful serial variability.

Single objective metrics of frailty commonly used in the clinical setting include assessment of activities of daily living (ADL), and the 6-minute walk test (6MWT). ADL was evaluated in an observational study of 734 inpatients with decompensated cirrhosis which found that the odds ratio (OR) for ADL score of less than 12 of 15 on mortality was 1.83 (95% CI: 1.05–3.20) (47). In a study of 250 patients with chronic liver disease, of which 98 subjects had cirrhosis, 6MWT was found to be an independent predictor of survival (P=0.024) while Child-Pugh score was found to negatively correlate with the 6 minute walk distance (6MWD) (R=−0.328, P<0.01) (48). Patient participation is required in these metrics thus feasibility may be severely limited in acutely ill patients or those with significant decompensations.

The Fried Frailty Index (FFI) is a 5-point score that encompasses reported subjective measures—exhaustion, unintentional weight loss, and low physical activity, and objective measures—walk speed, and hand grip strength (49). Patients are scored between 0 (not frail) and 5 (most frail). FFI ≥3 is consistent with decreased independent ADL and increased risk of falls (50). In a single-center study of 267 liver transplant waitlist patients, frailty defined by FFI was more significantly associated with removal from the waitlist or waitlist mortality, compared with patients who were not frail (22% vs. 10%, P=0.03) (6). Every 1 unit increase in FFI corresponds to a 50% increase in liver transplant waitlist mortality (HR 1.50, P=0.01) (5). While FFI provides a good overview of frailty, the index does not incorporate important factors such as age and medical comorbidities or cirrhosis-specific factors such as malnutrition, liver failure and HE.

The Clinical Frailty Scale (CFS) is a 1-minute bedside clinical assessment that incorporates comorbidities, level of daily physical activity, dependence for ADLs and life expectancy less than 6 months (51). The index is divided into nine categories ranging from “severely frail” to “very fit” with CFS of 4 indicating the patient is pre-frail. Frailty, CFS >4, is associated with hospitalization or death (OR 3.6, P=0.008) in a study of patients with cirrhosis (51). In a validation study of 299 outpatients/inpatients, for every 1 unit increase in CFS score, there was an independent association with mortality in the outpatient cohort (HR 1.534, P=0.007) while a CFS >4 was found to be an independent predictor of 28-day mortality (OR 4.627, P=0.045) (52). CFS limitations include providing only a snapshot of frailty and inability to identify specific areas of frailty.

The Liver Frailty Index (LFI) combines three objective performance-based tests—hand grip strength, time to complete five chair stands (seconds), and time holding three balance positions (feet side by side, tandem, and semi tandem)—to provide a score that categorizes patients with cirrhosis in the ambulatory setting as robust, pre-frail, or frail (index <3.2; 3.2–4.5, and >4.5, respectively) (53). LFI is well-studied in patients with cirrhosis and a multicenter study of 1,044 patients frailty, ascites, and HE were significantly associated with waitlist mortality sub-hazard ratio (sHR) 2.38 (95% CI: 1.77–3.20, P<0.001), 1.52 (95% CI: 1.14–2.05, P=0.004), and 1.84 (95% CI: 1.38–2.45, P<0.001), respectively (39). In adjusted multivariable models only frailty by LFI was significantly associated with waitlist mortality (sHR 1.82, 95% CI: 1.31–2.52, P<0.001) (39). LFI is inexpensive, easy to administer, quick to complete and most importantly, liver-specific. In a study of 529 patients awaiting liver transplantation, addition of LFI to clinician assessment, compared to clinical assessment alone, significantly improved predicting risk of mortality (0.74 vs. 0.68, P=0.02) (54). A summary of the three validated assessments to evaluate frailty in patients with advanced liver disease is provided in Table 1. Patients with cirrhosis should be assessed for frailty with a standardized tool at baseline and longitudinally.

Table 1

Comparison of frailty indices utilized in the ambulatory setting

Test Criteria Metrics Strengths Limitations
FFI (49) 5-point score based on: Robust: 0 criteria Criteria are easy to measure and based on objective physical performance The most time consuming of the three indices (10 minutes)
• Hand grip strength Prefrail: 1–2 criteria Does not account for comorbidities, age, malnutrition, hepatic encephalopathy
• Exhaustion Frail: ≥3 criteria Does not track progress with prehabilitation/interventions
• Unintentional weight loss
• Low physical activity
• Walk speed
CFS (51) Scale of 1–9 based on clinical judgment accounting for physical health, mental health, and functional status 1: very fit Quick to perform Does not identify specific areas of frailty
2: fit Simplicity of use/no need for complex testing Strictly uses subjective information
3: managing well Offers a comprehensive assessment Does not track progress with prehabilitation/interventions
4: very mild frailty
5: mild frailty
6: moderate frailty
7: severe frailty
8: very severe frailty
9: terminally ill
LFI (53) Composite metric based on: Robust: <3.2 Emphasizes functional status Not validated in hospitalized or acutely ill patients
• Hand grip strength Prefrail: 3.2–4.5 Easy to administer in clinical settings Focus is on functionality but does not account for cognitive decline
• Time to do five chair stands Frail: >4.5
• Time holding three balance positions

CFS, Clinical Frailty Scale; FFI, Fried Frailty Index; LFI, Liver Frailty Index.

There is significant heterogeneity in the evaluation of sarcopenia in patients with cirrhosis. Several indirect and direct modalities exist for muscle quantification including anthropometry, ultrasound, bioelectrical impedance analysis (BIA), dual-energy X-ray absorptiometry (DEXA), computed tomography (CT) and magnetic resonance imaging (MRI). Traditional anthropometric measures such as weight and BMI have fallen out of favor due to their inability to correctly distinguish between fat and muscle. Mid-arm muscle circumference (MAMC, in cm) is a safe and inexpensive method that estimates the skeletal muscle mass and can avoid confounding from fluid overload. Proposed MAMC cutoff points for severe and moderate depletion of muscle mass were ≤21.5 cm and ≤24.2 cm; ≤20.9 cm and ≤22.9 cm for women and men, respectively (55).

BIA estimates body fat and muscle mass through a weak electrical current that flows through the body (56). Muscle mass loss is diagnosed by BIA <7.0 kg/m2 for men and <5.7 kg/m2 for women (57). BIA for the measurement of body composition is a quick, noninvasive and reliable study (58). However, fluid retention, specifically ascites in patients with cirrhosis, affects the reliability of lean body mass assessment (59). BIA devices are not readily available in clinical practice and are also not well-validated in patients with cirrhosis.

Of the quantitative measures, ultrasound technology is the simplest, cheapest tool that can be employed to assess muscle mass. A transducer produces ultrasound waves that provide a noninvasive image of the targeted muscle (or muscle group). Quadriceps muscle thickness and quality showed concordance with CT for identifying patients with sarcopenia. Ultrasound measurement of thigh muscle thickness in combination with BMI is able to identify men and women with sarcopenia, AUC 0.78 and 0.89, respectively, when compared with CT imaging (60). Ultrasound measurement of the psoas muscle was significantly related to hospitalization and mortality (HR 0.881, 95% CI: 0.836–0.929, P<0.0001, and HR 0.930, 95% CI: 0.876–0.987, P=0.017, respectively) (61). Although readily available and non-invasive, ultrasound for sarcopenia assessment is not well validated in decompensated cirrhosis patients.

DEXA is a compartmentalized 3-dimensional whole-body scan with an emitting X-ray source that allows the measurement of lean mass (LM), fat mass (FM), and bone mineral content (BMC) (62). Muscle mass loss is diagnosed by DEXA at <7.0 kg/m2 for males and <5.4 kg/m2 for females (57). Appendicular LM assessed by DEXA in men with cirrhosis is strongly associated with mortality [HR 0.78 (0.62; 0.98), P=0.03] (63). However, accuracy may be restricted. In a study of 1,587 community-dwelling older adults evaluating sarcopenia via appendicular skeletal muscle mass by BIA and DEXA, BIA overestimated muscle mass compared with DEXA (2.89±0.38 vs. 2.97±0.45 kg/m2, P<0.005) (64). One major limitation of DEXA is the overestimation of muscle mass in patients with extracellular fluid accumulation due to its inability to differentiate between water and bone-free LM (65).

Cross-sectional imaging is by far the most widely used and current gold standard for detecting sarcopenia in cirrhosis. The L3 SMI is a well-validated tool that requires specialized software and expertise to diagnose muscle wasting. The area of skeletal muscle (measured in cm2/m2) at the lumbar vertebra, L3, is quantified by cross-sectional imaging utilizing body segmentation analysis and further stratified based on sex-specific cutoffs SMI ≤52–55 cm2/m2 for males and SMI ≤39–41 cm2/m2 for females (66,67). In a study of 149 patients with cirrhosis, the degree of cirrhosis categorized by Child-Pugh class A, B and C, was associated with changes in skeletal muscle area per year, –1.3%, –3.5% and –6.1%, respectively (68). CT is straightforward, replicable and minimally affected by fluid retention. A single-center observational cohort study of 145 men with cirrhosis demonstrated prevalence of sarcopenia differed between CT and DEXA, 70.3% and 38.7%, respectively (69). While CT imaging is the preferred modality, cost and radiation exposure limit its practical use for the sole purpose of detecting sarcopenia (70). Table 1 compares the utility, strengths and limitations of the three major modalities used in assessing sarcopenia.


Outcomes

Frailty and sarcopenia are associated with increased risk of adverse outcomes including falls, fractures, disability, hospitalizations and mortality. In a study of 734 hospitalized patients with cirrhosis, disability defined by need for some assistance with ≥3 ADLs, was associated with a nearly 2-fold increased 90-day mortality (OR 1.83; 95% CI: 1.05–3.20) (47). In the ambulatory setting, physical frailty (LFI ≥4.5) was associated with a 1.9× increased risk of waitlist mortality when compared to non-frail patients (sHR 1.82; 95% CI: 1.31–2.52) (39). Changes in frailty over time also informs mortality risk. In a study of 1,093 patients with cirrhosis, a 0.1 unit increase in LFI every 3 months was associated with a 2-fold increased risk of waitlist death (HR 2.04; 95% CI: 1.35–3.09) (54). Outpatient assessments including clinical frailty scale, Fried frailty phenotype, frailty index, and short gait speed also correlate with both incidence and length of hospitalization (50,71,72). Two metrics of physical frailty/performance, the Karnofsky performance status (KPS) and ADL, have data from hospitalized cohorts. Loss of ADL was associated with adjusted risk of 90-day mortality after discharge (HR 1.83, 95% CI: 1.05–3.20) and for every 10-points of improvement in KPS score there was a 30% reduction in odds of 30 days death on discharge (95% CI: 20–40%) (47,73).

Sarcopenia and cirrhosis are bidirectional. Sarcopenia increases the incidence of HE (OR 2.42, P=0.001) and is significantly associated with mortality (HR 2.15; 95% CI: 1.52–3.05, P<0.001) (74). A prospective study of 911 patients with cirrhosis demonstrated that overall survival was significantly lower in cirrhosis patients with sarcopenia compared with those without sarcopenia [risk ratio (RR) 2.643, 95% CI: 1.646–4.244, P<0.001] (6). Median survival was shorter in patients with cirrhosis and sarcopenia when compared to non-sarcopenic patients (20±3 vs. 95±24 months, P<0.001) (75).

Patients with cirrhosis and sarcopenia were found to have increased cumulative incidence of ascites (RR 1.827, 95% CI: 1.259–2.651, P=0.002), SBP (RR 3.331, 95% CI: 1.404–7.903, P=0.006), HE (RR 1.962, 95% CI: 1.070–3.600, P=0.029) and gastrointestinal varices (RR 2.138, 95% CI: 1.319–3.466, P=0.002) (6). Aside from worsening complications of cirrhosis, sarcopenia was found to be an independent predictor of mortality (HR 2.36, 95% CI: 1.23–4.53) (72). Median survival time for cirrhosis patients with sarcopenia compared with no sarcopenia was 19±6 vs. 34±11 months (P=0.005) (41). In a study with a median follow-up period of 46.2 months (range, 3.4–87.6 months), cumulative mortality was significantly higher in patients with changes in skeletal muscle area per year <−2.4% vs. ≥−2.4% (P<0.001) (76).

Sarcopenia is a powerful predictor of waitlist mortality. More severe sarcopenia is associated with higher rates of delisting on the waitlist and waitlist mortality in 396 patients with cirrhosis awaiting liver transplantation (45.6 cm2/m2, 95% CI: 38.5–50.8 cm2/m2 compared with 48.5 cm2/m2, 95% CI: 42.4–54.9 cm2/m2, respectively, P<0.001) (77). In liver transplant recipients, sarcopenic patients had longer hospital stays when compared with nonsarcopenic patients (40±4 vs. 25±3 d; P=0.005) (78). Sarcopenia is a predictor of pre- and post-liver transplant mortality (79,80).


Interventions

Nutrition

Effective management of nutrition in patients with cirrhosis is a vital aspect of treatment. Interventions that address these nutritional deficits and counter the high metabolic demands of cirrhosis are essential. Determining a patient’s ideal caloric intake can be difficult. Indirect calorimetry is a method used to measure energy expenditure by assessing respiratory gas exchange thereby estimating metabolic rate and substrate utilization (81). However, assessment requires specialized equipment and thus is not widely available. A second option is estimating caloric need via weight-based equations using estimated dry weight. In non-obese patients at least 35 kcal/kg daily and in obese patients a moderately hypocaloric (500–800 kcal/d deficit) diet is clinically applicable (82).

Protein intake of 1.2–1.5 g/(kg·d) is recommended for cirrhosis patients, with up to 2.0 g/(kg·d) allowance for critically ill patients (83,84). BCAA supplementation at 0.25 g/(kg·d) may be an add-on therapy if protein goals are not met. There is insufficient data for the use of BCAAs in critically ill patients (83). A multicenter, randomized controlled trial compared 12 g/d of BCAA over two years vs. a diet comprised of 1–1.4 g/kg protein daily and a minimal caloric threshold. The primary endpoints included death (regardless of cause), esophageal variceal bleed, hepatocellular carcinoma or progression of cirrhosis. All of these were decreased in the BCAA group (HR 0.67, 95% CI: 0.49–0.93; P=0.015) vs. the diet group (85).

The pattern of food intake is also important in patients with cirrhosis. A “nibbling” or “grazing” pattern characterized by a large number of small meals including a good breakfast and a late evening meal, improved energy metabolism and resulted in shorter episodes of catabolism, compared to a “gorging” pattern characterized by a small number of large meals (86). Timing of intake is of importance in cirrhosis with the goal of avoiding extended fasting intervals. An early breakfast with small meals every 3–4 waking hours followed by a late evening/nighttime snack is the optimal strategy (87). In a cohort of 103 patients with cirrhosis who received either daytime or nighttime supplementary nutrition there was an associated increase in total body protein in the nighttime group at 3 months (0.38±0.10 kg, P=0.0004), 6 months (0.48±0.13 kg, P=0.0007) and 12 months (0.53±0.17 kg, P=0.003) compared to baseline. No significant improvement was seen in the daytime group (87).

All cirrhosis patients also require dietary counseling and educational resources. The mortality benefit of inpatient nutritional assessment has been contentious due to the inconclusive data gathered thus far. However, this may be skewed due to the limited duration of intervention in hospitalized patients with cirrhosis (88). A recent study tracking inpatient nutrition consults and education in hospitalized patients with cirrhosis prospectively over 90 days demonstrated a 10% (39.4% vs. 28.4%; P=0.04) absolute reduction in hospital re-admission and increased outpatient nutrition follow-up (89).

Physical activity & exercise

Structured exercise improves muscle function and overall health outcomes. Resistance and endurance exercises help promote muscle protein synthesis by activating the mammalian target of rapamycin (mTOR) signaling pathways, which are essential for muscle growth and repair, reducing muscle degradation, enhancing mitochondrial function, and improving muscle perfusion. This effectively counteracts the progression of sarcopenia (72,90). The upregulation of the mTOR signaling pathway can be seen on immunofluorescence and in a prospective study of 17 patients (91,92). Protein synthesis after exercise increased by 52% in males and 47% in females (P<0.05). The phosphorylation of mTOR also increased in both groups (P<0.05) (92). Aerobic exercise, such as walking, is recommended 4–7 days a week for a total of 150 minutes. Resistance exercise, such as functional or progressive weight/band training is recommended 2–3 days a week with flexibility and balance exercises at similar intervals (93).

Hormonal therapy

Hormone supplementation with products such as testosterone has shown potential benefits in restoring muscle mass. However, these interventions are accompanied with various potential adverse effects, most significantly increased cardiovascular risk (94). Testosterone has anabolic activity which leads to increased overall skeletal muscle mass, plasma albumin level but lacks finite long-term utility data (24). In a 12-month, double-blind, placebo-controlled trial of intramuscular testosterone undecanoate administered to 101 men with established cirrhosis and low serum testosterone, primary outcome of appendicular LM was significantly higher in testosterone-treated men [mean adjusted difference (MAD) +1.69 kg, P=0.021] and secondary outcomes of total LM and femoral neck mineral density were higher in testosterone-treated men (94). Strict nutrition management, guided exercise programs, along with select pharmacological aids offer a multifaceted approach to sarcopenia management.

Interventions specific to cirrhosis

Cirrhotic patients who develop sarcopenia require strict targeted management of complications such as ascites, HE, and portal hypertension to improve patient outcomes and mitigate disease progression (91). Ascites, the accumulation of fluid in the abdominal cavity, is a consequence of cirrhosis and often exacerbates sarcopenia through decreased protein intake secondary to diminished appetite and the compressive effect of intra-abdominal fluid. Effective initial management includes sodium restriction with concomitant diuretic use to assist with hypervolemia (95). Diuretics must be used cautiously, particularly loop diuretics, as they can negatively impact muscle mass at higher doses and increase mortality (68). However, spironolactone and tolvaptan have both been shown to decrease progression of sarcopenia and can be beneficial adjuncts to loop diuretics (96,97).

Patients with HE benefit significantly from lactulose and rifaximin as these medications improve cognitive function and slow sarcopenic decline (93). Lactulose is a non-absorbable disaccharide that acidifies the gut upon its breakdown leading to increased conversion of NH3 to NH4. This increases the ammonium ions trapped in the gut for excretion on bowel movement, thereby decreasing total ammonia in the plasma. Rifaximin decreases the ammonia-producing bacteria population in the gut, although the mechanism of action is disputed. Fecal microbial transplantation (FMT) has been proposed as a method for restoring healthy microbial function, but there is insufficient evidence on its efficacy (24).

In patients with advanced portal hypertension, a transjugular intrahepatic portosystemic shunt (TIPS) procedure is a shunt that reduces pressure in the portal vein (98). TIPS can significantly improve symptoms of cirrhosis such as protein-wasting ascites and indirectly support muscle health (99). TIPS placement also addresses gastrointestinal edema associated with cirrhosis thus enhancing uptake of protein and fats leading to improved muscle synthesis (100). In advanced cirrhosis, liver transplantation may be the only definitive treatment, showing substantial improvement in muscle mass as it restores liver function thereby enhancing nutrient metabolism and decreasing muscle breakdown (99). For these transplant candidates, pre- and post-operative nutritional management, routine exercise therapy, and consistent ammonia-lowering interventions are crucial to maximize outcomes and long-term recovery (101).

Emerging interventions

The landscape of therapeutic targets for skeletal muscle modulation continues to evolve. Rifaximin and L-ornithine L-aspartate have been shown to reduce ammonia levels resulting in increased muscle mass (102). In an experimental model, rats given L-ornithine L-aspartate and rifaximin over 4 weeks showed an improvement in muscle fiber size (P<0.001) (103). BCAAs activate mTOR and supplementation improves muscle mass in cirrhotic patients with sarcopenia. In a study of 32 patients with cirrhosis and sarcopenia, 12 weeks of BCAA supplementation demonstrated improvement in muscle mass in the BCAA group when compared to placebo (83.3% vs. 46.7%; P=0.056) (104). Mitochondrial-targeted antioxidants are another category requiring further research as they may improve protein synthesis along with muscle mass (103).

Future research directions for sarcopenia in cirrhosis include exploring novel therapeutic approaches, however this necessitates further clarifying the mechanisms of muscle wasting in cirrhosis along with refining diagnostic tools.


Palliative care in cirrhosis and sarcopenia

As cirrhosis progresses, patients experience more decompensations due to increasing portal pressures. Such complications include the development and/or worsening of ascites, HE, gastroesophageal variceal bleeding, jaundice, renal damage, pulmonary complications, and hepatocellular carcinoma. More insidious and under-recognized are sarcopenia and frailty both of which confer high rates of morbidity and mortality. While liver transplantation may be curative for a majority of these decompensations, barriers including organ availability, financial or insurance-related limitations, and active substance use may preclude patients from timely transplantation. For patients affected by these barriers, palliative medicine can offer tremendous aid in the form of supportive physical and psychosocial care.

In patients with cirrhosis, managing symptoms such as pain, fatigue, and anorexia, factors that exacerbate malnutrition and sarcopenia, is essential for improving quality of life. Palliative care teams may provide a multidisciplinary approach to nutritional support, involving dietitians and other specialists to ensure adequate protein and calorie intake.

Palliative care plays a crucial role in the management of cirrhosis symptoms. Addressing symptom management helps improve quality of life. Despite its benefits, palliative care is often underutilized and introduced late in the disease course. Early integration of palliative care can lead to improved management of symptoms thus optimizing frailty and sarcopenia, reduced hospitalizations, and enhanced patient and caregiver satisfaction.


Conclusions

Malnutrition is a severe complication of chronic liver disease and cirrhosis. Frailty and sarcopenia often present simultaneously in patients with cirrhosis. This perpetuates further decompensations which adversely affect quality of life, overall mortality and transplant candidacy. In patients with cirrhosis, early recognition of frailty and sarcopenia, understanding the interplay between the two processes and providing timely interventions is imperative. While we have a robust understanding of the symptoms and complications of these disease processes, many questions remain. Clarification of optimal diagnostic tools, effective interventions, longitudinal evaluation and management are still topics requiring further investigation.


Acknowledgments

None.


Footnote

Peer Review File: Available at https://apm.amegroups.com/article/view/10.21037/apm-24-173/rc

Funding: None.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://apm.amegroups.com/article/view/10.21037/apm-24-173/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Asrani SK, Devarbhavi H, Eaton J, et al. Burden of liver diseases in the world. J Hepatol 2019;70:151-71. [Crossref] [PubMed]
  2. Sharma B, John S. Hepatic Cirrhosis. In: StatPearls [Internet]. Treasure Island (FL): StatPearls; 2024 [cited 2024 Nov 12]. Available online: http://www.ncbi.nlm.nih.gov/books/NBK482419/
  3. Mansour D, McPherson S. Management of decompensated cirrhosis. Clin Med (Lond) 2018;18:s60-5. [Crossref] [PubMed]
  4. Tsoris A, Marlar CA. Use of the Child Pugh Score in Liver Disease. In: StatPearls [Internet]. Treasure Island (FL): StatPearls; 2024 [cited 2024 Dec 14]. Available online: http://www.ncbi.nlm.nih.gov/books/NBK542308/
  5. Lai JC, Feng S, Terrault NA, et al. Frailty predicts waitlist mortality in liver transplant candidates. Am J Transplant 2014;14:1870-9. [Crossref] [PubMed]
  6. Zeng X, Shi ZW, Yu JJ, et al. Sarcopenia as a prognostic predictor of liver cirrhosis: a multicentre study in China. J Cachexia Sarcopenia Muscle 2021;12:1948-58. [Crossref] [PubMed]
  7. Lai JC, Tandon P, Bernal W, et al. Malnutrition, Frailty, and Sarcopenia in Patients With Cirrhosis: 2021 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2021;74:1611-44. [Crossref] [PubMed]
  8. Pashayee-Khamene F, Saber-Firoozi M, Hatami B, et al. Food groups intake of cirrhotic patients, comparison with the nutritional status and disease stage. Gastroenterol Hepatol Bed Bench 2019;12:226-32.
  9. Merli M, Giusto M, Gentili F, et al. Nutritional status: its influence on the outcome of patients undergoing liver transplantation. Liver Int 2010;30:208-14. [Crossref] [PubMed]
  10. Kalafateli M, Mantzoukis K, Choi Yau Y, et al. Malnutrition and sarcopenia predict post‐liver transplantation outcomes independently of the Model for End-stage Liver Disease score. J Cachexia Sarcopenia Muscle 2017;8:113-21. [Crossref] [PubMed]
  11. Luong R, Kim M, Lee A, et al. Assessing nutritional status in a cohort of liver cirrhosis outpatients: A prospective cross-sectional study. Nutr Health 2020;26:19-25. [Crossref] [PubMed]
  12. Alvares-da-Silva MR, Reverbel da Silveira T. Comparison between handgrip strength, subjective global assessment, and prognostic nutritional index in assessing malnutrition and predicting clinical outcome in cirrhotic outpatients. Nutrition 2005;21:113-7. [Crossref] [PubMed]
  13. Alberino F, Gatta A, Amodio P, et al. Nutrition and survival in patients with liver cirrhosis. Nutrition 2001;17:445-50. [Crossref] [PubMed]
  14. Goh SK, Chapman B, Romero S, et al. Malnutrition is Associated with Poor Outcomes after Liver Transplantation: A Promising Target for Pre-Transplantation Nutritional Intervention. Transplantation 2018;102:S870.
  15. Ferreira LG, Ferreira Martins AI, Cunha CE, et al. Negative energy balance secondary to inadequate dietary intake of patients on the waiting list for liver transplantation. Nutrition 2013;29:1252-8. [Crossref] [PubMed]
  16. Volk ML, Fisher N, Fontana RJ. Patient knowledge about disease self-management in cirrhosis. Am J Gastroenterol 2013;108:302-5. [Crossref] [PubMed]
  17. Wu J, Meng QH. Current understanding of the metabolism of micronutrients in chronic alcoholic liver disease. World J Gastroenterol 2020;26:4567-78. [Crossref] [PubMed]
  18. Jorgensen RA, Lindor KD, Sartin JS, et al. Serum lipid and fat-soluble vitamin levels in primary sclerosing cholangitis. J Clin Gastroenterol 1995;20:215-9. [Crossref] [PubMed]
  19. Holick MF. Vitamin D deficiency. N Engl J Med 2007;357:266-81. [Crossref] [PubMed]
  20. Okubo T, Atsukawa M, Tsubota A, et al. Effect of Vitamin D Supplementation on Skeletal Muscle Volume and Strength in Patients with Decompensated Liver Cirrhosis Undergoing Branched Chain Amino Acids Supplementation: A Prospective, Randomized, Controlled Pilot Trial. Nutrients 2021;13:1874. [Crossref] [PubMed]
  21. Imano E, Kanda T, Nakatani Y, et al. Impaired splanchnic and peripheral glucose uptake in liver cirrhosis. J Hepatol 1999;31:469-73. [Crossref] [PubMed]
  22. Petersen KF, Krssak M, Navarro V, et al. Contributions of net hepatic glycogenolysis and gluconeogenesis to glucose production in cirrhosis. Am J Physiol 1999;276:E529-35. [Crossref] [PubMed]
  23. Ebadi M, Bhanji RA, Mazurak VC, et al. Sarcopenia in cirrhosis: from pathogenesis to interventions. J Gastroenterol 2019;54:845-59. [Crossref] [PubMed]
  24. Henin G, Lanthier N, Dahlqvist G. Pathophysiological changes of the liver-muscle axis in end-stage liver disease: what is the right target? Acta Gastroenterol Belg 2022;85:611-24. [Crossref] [PubMed]
  25. Yamato M, Muto Y, Yoshida T, et al. Clearance rate of plasma branched-chain amino acids correlates significantly with blood ammonia level in patients with liver cirrhosis. Int Hepatol Commun 1995;3:91-6.
  26. Santangeli E, Abbati C, Chen R, et al. Pathophysiological-Based Nutritional Interventions in Cirrhotic Patients with Sarcopenic Obesity: A State-of-the-Art Narrative Review. Nutrients 2024;16:427. [Crossref] [PubMed]
  27. Davuluri G, Allawy A, Thapaliya S, et al. Hyperammonaemia-induced skeletal muscle mitochondrial dysfunction results in cataplerosis and oxidative stress. J Physiol 2016;594:7341-60. [Crossref] [PubMed]
  28. Qiu J, Tsien C, Thapalaya S, et al. Hyperammonemia-mediated autophagy in skeletal muscle contributes to sarcopenia of cirrhosis. Am J Physiol Endocrinol Metab 2012;303:E983-93. [Crossref] [PubMed]
  29. Aqel BA, Scolapio JS, Dickson RC, et al. Contribution of ascites to impaired gastric function and nutritional intake in patients with cirrhosis and ascites. Clin Gastroenterol Hepatol 2005;3:1095-100. [Crossref] [PubMed]
  30. Kalaitzakis E. Gastrointestinal dysfunction in liver cirrhosis. World J Gastroenterol 2014;20:14686-95. [Crossref] [PubMed]
  31. Aprile LR, Meneghelli UG, Martinelli AL, et al. Gastric motility in patients with presinusoidal portal hypertension. Am J Gastroenterol 2002;97:3038-44. [Crossref] [PubMed]
  32. Pinzone MR, Celesia BM, Di Rosa M, et al. Microbial translocation in chronic liver diseases. Int J Microbiol 2012;2012:694629. [Crossref] [PubMed]
  33. O'Toole PW, Jeffery IB. Gut microbiota and aging. Science 2015;350:1214-5. [Crossref] [PubMed]
  34. Walsh ME, Bhattacharya A, Sataranatarajan K, et al. The histone deacetylase inhibitor butyrate improves metabolism and reduces muscle atrophy during aging. Aging Cell 2015;14:957-70. [Crossref] [PubMed]
  35. Ghosh G, Jesudian AB. Small Intestinal Bacterial Overgrowth in Patients With Cirrhosis. J Clin Exp Hepatol 2019;9:257-67. [Crossref] [PubMed]
  36. Goral V, Atayan Y, Kaplan A. The relation between pathogenesis of liver cirrhosis, hepatic encephalopathy and serum cytokine levels: what is the role of tumor necrosis factor α? Hepatogastroenterology 2011;58:943-8.
  37. Williams FR, Milliken D, Lai JC, et al. Assessment of the Frail Patient with End-Stage Liver Disease: A Practical Overview of Sarcopenia, Physical Function, and Disability. Hepatol Commun 2021;5:923-37. [Crossref] [PubMed]
  38. Morley JE, Vellas B, van Kan GA, et al. Frailty consensus: a call to action. J Am Med Dir Assoc 2013;14:392-7. [Crossref] [PubMed]
  39. Lai JC, Rahimi RS, Verna EC, et al. Frailty Associated With Waitlist Mortality Independent of Ascites and Hepatic Encephalopathy in a Multicenter Study. Gastroenterology 2019;156:1675-82. [Crossref] [PubMed]
  40. Elsheikh M, El Sabagh A, Mohamed IB, et al. Frailty in end-stage liver disease: Understanding pathophysiology, tools for assessment, and strategies for management. World J Gastroenterol 2023;29:6028-48. [Crossref] [PubMed]
  41. Montano-Loza AJ, Meza-Junco J, Prado CM, et al. Muscle wasting is associated with mortality in patients with cirrhosis. Clin Gastroenterol Hepatol 2012;10:166-73, 173.e1.
  42. Tantai X, Liu Y, Yeo YH, et al. Effect of sarcopenia on survival in patients with cirrhosis: A meta-analysis. J Hepatol 2022;76:588-99. [Crossref] [PubMed]
  43. Cui Y, Zhang M, Guo J, et al. Correlation between sarcopenia and cirrhosis: a meta-analysis. Front Nutr 2023;10:1342100. [Crossref] [PubMed]
  44. Carias S, Castellanos AL, Vilchez V, et al. Nonalcoholic steatohepatitis is strongly associated with sarcopenic obesity in patients with cirrhosis undergoing liver transplant evaluation. J Gastroenterol Hepatol 2016;31:628-33. [Crossref] [PubMed]
  45. Hong SH, Choi KM. Sarcopenic Obesity, Insulin Resistance, and Their Implications in Cardiovascular and Metabolic Consequences. Int J Mol Sci 2020;21:494. [Crossref] [PubMed]
  46. Arow Z, Gabarin M, Abu-Hosein H, et al. Eyeball Test for the Assessment of Frailty in Elderly Patients With Cardiovascular Disease: A Prospective Study. Am J Cardiol 2023;204:9-13. [Crossref] [PubMed]
  47. Tapper EB, Finkelstein D, Mittleman MA, et al. Standard assessments of frailty are validated predictors of mortality in hospitalized patients with cirrhosis. Hepatology 2015;62:584-90. [Crossref] [PubMed]
  48. Alameri HF, Sanai FM, Al Dukhayil M, et al. Six Minute Walk Test to assess functional capacity in chronic liver disease patients. World J Gastroenterol 2007;13:3996-4001. [Crossref] [PubMed]
  49. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001;56:M146-56. [Crossref] [PubMed]
  50. Lai JC, Dodge JL, Sen S, et al. Functional decline in patients with cirrhosis awaiting liver transplantation: Results from the functional assessment in liver transplantation (FrAILT) study. Hepatology 2016;63:574-80. [Crossref] [PubMed]
  51. Tandon P, Tangri N, Thomas L, et al. A Rapid Bedside Screen to Predict Unplanned Hospitalization and Death in Outpatients With Cirrhosis: A Prospective Evaluation of the Clinical Frailty Scale. Am J Gastroenterol 2016;111:1759-67. [Crossref] [PubMed]
  52. Kremer WM, Nagel M, Reuter M, et al. Validation of the Clinical Frailty Scale for the Prediction of Mortality in Patients With Liver Cirrhosis. Clin Transl Gastroenterol 2020;11:e00211. [Crossref] [PubMed]
  53. Lai JC, Covinsky KE, Dodge JL, et al. Development of a novel frailty index to predict mortality in patients with end-stage liver disease. Hepatology 2017;66:564-74. [Crossref] [PubMed]
  54. Lai JC, Covinsky KE, McCulloch CE, et al. The Liver Frailty Index Improves Mortality Prediction of the Subjective Clinician Assessment in Patients With Cirrhosis. Am J Gastroenterol 2018;113:235-42. [Crossref] [PubMed]
  55. Saueressig C, Alves BC, Luft VC, et al. Mid-arm muscle circumference cutoff points in patients with cirrhosis: Low muscle mass related to malnutrition predicts mortality. Nutrition 2024;125:112471. [Crossref] [PubMed]
  56. Kyle UG, Bosaeus I, de Lorenzo AD, et al. Bioelectrical impedance analysis—part I: review of principles and methods. Clin Nutr 2004;23:1226-43. [Crossref] [PubMed]
  57. Chen H, Yang C, Yan S, et al. Sarcopenia in cirrhosis: From pathophysiology to interventional therapy. Exp Gerontol 2024;196:112571. [Crossref] [PubMed]
  58. Gonzalez MC, Barbosa-Silva TG, Heymsfield SB. Bioelectrical impedance analysis in the assessment of sarcopenia. Curr Opin Clin Nutr Metab Care 2018;21:366-74. [Crossref] [PubMed]
  59. Pirlich M, Schütz T, Spachos T, et al. Bioelectrical impedance analysis is a useful bedside technique to assess malnutrition in cirrhotic patients with and without ascites. Hepatology 2000;32:1208-15. [Crossref] [PubMed]
  60. Tandon P, Low G, Mourtzakis M, et al. A Model to Identify Sarcopenia in Patients With Cirrhosis. Clin Gastroenterol Hepatol 2016;14:1473-1480.e3. [Crossref] [PubMed]
  61. Hari A, Berzigotti A, Štabuc B, et al. Muscle psoas indices measured by ultrasound in cirrhosis - Preliminary evaluation of sarcopenia assessment and prediction of liver decompensation and mortality. Dig Liver Dis 2019;51:1502-7. [Crossref] [PubMed]
  62. Messina C, Albano D, Gitto S, et al. Body composition with dual energy X-ray absorptiometry: from basics to new tools. Quant Imaging Med Surg 2020;10:1687-98. [Crossref] [PubMed]
  63. Sinclair M, Hoermann R, Peterson A, et al. Use of Dual X-ray Absorptiometry in men with advanced cirrhosis to predict sarcopenia-associated mortality risk. Liver Int 2019;39:1089-97. [Crossref] [PubMed]
  64. Cheng KY, Chow SK, Hung VW, et al. Diagnosis of sarcopenia by evaluating skeletal muscle mass by adjusted bioimpedance analysis validated with dual-energy X-ray absorptiometry. J Cachexia Sarcopenia Muscle 2021;12:2163-73. [Crossref] [PubMed]
  65. Hey P, Hoermann R, Gow P, et al. Reduced upper limb lean mass on dual energy X-ray absorptiometry predicts adverse outcomes in male liver transplant recipients. World J Transplant 2022;12:120-30. [Crossref] [PubMed]
  66. Kong M, Geng N, Zhou Y, et al. Defining reference values for low skeletal muscle index at the L3 vertebra level based on computed tomography in healthy adults: A multicentre study. Clin Nutr 2022;41:396-404. [Crossref] [PubMed]
  67. Amini B, Boyle SP, Boutin RD, et al. Approaches to Assessment of Muscle Mass and Myosteatosis on Computed Tomography: A Systematic Review. J Gerontol A Biol Sci Med Sci 2019;74:1671-8. [Crossref] [PubMed]
  68. Hanai T, Shiraki M, Ohnishi S, et al. Rapid skeletal muscle wasting predicts worse survival in patients with liver cirrhosis. Hepatol Res 2016;46:743-51. [Crossref] [PubMed]
  69. Sinclair M, Chapman B, Hoermann R, et al. Handgrip Strength Adds More Prognostic Value to the Model for End‐Stage Liver Disease Score Than Imaging‐Based Measures of Muscle Mass in Men with Cirrhosis. Liver Transpl 2019;25:1480-7. [Crossref] [PubMed]
  70. Carey EJ, Lai JC, Sonnenday C, et al. A North American Expert Opinion Statement on Sarcopenia in Liver Transplantation. Hepatology 2019;70:1816-29. [Crossref] [PubMed]
  71. Dunn MA, Josbeno DA, Tevar AD, et al. Frailty as Tested by Gait Speed is an Independent Risk Factor for Cirrhosis Complications that Require Hospitalization. Am J Gastroenterol 2016;111:1768-75. [Crossref] [PubMed]
  72. Tandon P, Ney M, Irwin I, et al. Severe muscle depletion in patients on the liver transplant wait list: its prevalence and independent prognostic value. Liver Transpl 2012;18:1209-16. [Crossref] [PubMed]
  73. Tandon P, Reddy KR, O'Leary JG, et al. A Karnofsky performance status-based score predicts death after hospital discharge in patients with cirrhosis. Hepatology 2017;65:217-24. [Crossref] [PubMed]
  74. Bhanji RA, Moctezuma-Velazquez C, Duarte-Rojo A, et al. Myosteatosis and sarcopenia are associated with hepatic encephalopathy in patients with cirrhosis. Hepatol Int 2018;12:377-86. [Crossref] [PubMed]
  75. Montano-Loza AJ, Duarte-Rojo A, Meza-Junco J, et al. Inclusion of Sarcopenia Within MELD (MELD-Sarcopenia) and the Prediction of Mortality in Patients With Cirrhosis. Clin Transl Gastroenterol 2015;6:e102. [Crossref] [PubMed]
  76. Jeong JY, Lim S, Sohn JH, et al. Presence of Sarcopenia and Its Rate of Change Are Independently Associated with Long-term Mortality in Patients with Liver Cirrhosis. J Korean Med Sci 2018;33:e299. [Crossref] [PubMed]
  77. Carey EJ, Lai JC, Wang CW, et al. A multicenter study to define sarcopenia in patients with end-stage liver disease. Liver Transpl 2017;23:625-33. [Crossref] [PubMed]
  78. Montano-Loza AJ, Meza-Junco J, Baracos VE, et al. Severe muscle depletion predicts postoperative length of stay but is not associated with survival after liver transplantation. Liver Transpl 2014;20:640-8. [Crossref] [PubMed]
  79. Kaido T, Ogawa K, Fujimoto Y, et al. Impact of sarcopenia on survival in patients undergoing living donor liver transplantation. Am J Transplant 2013;13:1549-56. [Crossref] [PubMed]
  80. Englesbe MJ, Patel SP, He K, et al. Sarcopenia and mortality after liver transplantation. J Am Coll Surg 2010;211:271-8. [Crossref] [PubMed]
  81. Delsoglio M, Achamrah N, Berger MM, et al. Indirect Calorimetry in Clinical Practice. J Clin Med 2019;8:1387. [Crossref] [PubMed]
  82. Córdoba J, López-Hellín J, Planas M, et al. Normal protein diet for episodic hepatic encephalopathy: results of a randomized study. J Hepatol 2004;41:38-43. [Crossref] [PubMed]
  83. Plauth M. Nutritional Intervention in Chronic Liver Failure. Visc Med 2019;35:292-8. [Crossref] [PubMed]
  84. EASL Clinical Practice Guidelines on nutrition in chronic liver disease. J Hepatol 2019;70:172-93. [Crossref] [PubMed]
  85. Muto Y, Sato S, Watanabe A, et al. Effects of oral branched-chain amino acid granules on event-free survival in patients with liver cirrhosis. Clin Gastroenterol Hepatol 2005;3:705-13. [Crossref] [PubMed]
  86. Georgiou A, Yannakoulia M, Papatheodoridis GV, et al. Assessment of dietary habits and the adequacy of dietary intake of patients with cirrhosis-the KIRRHOS study. Clin Nutr 2021;40:3992-8. [Crossref] [PubMed]
  87. Plank LD, Gane EJ, Peng S, et al. Nocturnal nutritional supplementation improves total body protein status of patients with liver cirrhosis: A randomized 12-month trial. Hepatology 2008;48:557-66. [Crossref] [PubMed]
  88. Krasnoff JB, Vintro AQ, Ascher NL, et al. A randomized trial of exercise and dietary counseling after liver transplantation. Am J Transplant 2006;6:1896-905. [Crossref] [PubMed]
  89. Reuter B, Shaw J, Hanson J, et al. Nutritional Assessment in Inpatients With Cirrhosis Can Be Improved After Training and Is Associated With Lower Readmissions. Liver Transpl 2019;25:1790-9. [Crossref] [PubMed]
  90. Aamann L, Dam G, Borre M, et al. Resistance Training Increases Muscle Strength and Muscle Size in Patients With Liver Cirrhosis. Clin Gastroenterol Hepatol 2020;18:1179-1187.e6. [Crossref] [PubMed]
  91. Song Z, Moore DR, Hodson N, et al. Resistance exercise initiates mechanistic target of rapamycin (mTOR) translocation and protein complex co-localisation in human skeletal muscle. Sci Rep 2017;7:5028. [Crossref] [PubMed]
  92. Dreyer HC, Fujita S, Glynn EL, et al. Resistance exercise increases leg muscle protein synthesis and mTOR signalling independent of sex. Acta Physiol (Oxf) 2010;199:71-81. [Crossref] [PubMed]
  93. Tandon P, Zanetto A, Piano S, et al. Liver transplantation in the patient with physical frailty. J Hepatol 2023;78:1105-17. [Crossref] [PubMed]
  94. Sinclair M, Grossmann M, Hoermann R, et al. Testosterone therapy increases muscle mass in men with cirrhosis and low testosterone: A randomised controlled trial. J Hepatol 2016;65:906-13. [Crossref] [PubMed]
  95. Traub J, Reiss L, Aliwa B, et al. Malnutrition in Patients with Liver Cirrhosis. Nutrients 2021;13:540. [Crossref] [PubMed]
  96. Burton LA, McMurdo ME, Struthers AD. Mineralocorticoid antagonism: a novel way to treat sarcopenia and physical impairment in older people? Clin Endocrinol (Oxf) 2011;75:725-9. [Crossref] [PubMed]
  97. Namba M, Hiramatsu A, Aikata H, et al. Management of refractory ascites attenuates muscle mass reduction and improves survival in patients with decompensated cirrhosis. J Gastroenterol 2020;55:217-26. [Crossref] [PubMed]
  98. Rajesh S, George T, Philips CA, et al. Transjugular intrahepatic portosystemic shunt in cirrhosis: An exhaustive critical update. World J Gastroenterol 2020;26:5561-96. [Crossref] [PubMed]
  99. Hsu CS, Kao JH. Sarcopenia and chronic liver diseases. Expert Rev Gastroenterol Hepatol 2018;12:1229-44. [Crossref] [PubMed]
  100. Liu J, Ma J, Yang C, et al. Sarcopenia in Patients with Cirrhosis after Transjugular Intrahepatic Portosystemic Shunt Placement. Radiology 2022;303:711-9. [Crossref] [PubMed]
  101. Anand AC. Nutrition and Muscle in Cirrhosis. J Clin Exp Hepatol 2017;7:340-57. [Crossref] [PubMed]
  102. Di Cola S, Nardelli S, Ridola L, et al. Ammonia and the Muscle: An Emerging Point of View on Hepatic Encephalopathy. J Clin Med 2022;11:611. [Crossref] [PubMed]
  103. Kumar A, Davuluri G, Welch N, et al. Oxidative stress mediates ethanol-induced skeletal muscle mitochondrial dysfunction and dysregulated protein synthesis and autophagy. Free Radic Biol Med 2019;145:284-99. [Crossref] [PubMed]
  104. Hernández-Conde M, Llop E, Gómez-Pimpollo L, et al. Adding Branched-Chain Amino Acids to an Enhanced Standard-of-Care Treatment Improves Muscle Mass of Cirrhotic Patients With Sarcopenia: A Placebo-Controlled Trial. Am J Gastroenterol 2021;116:2241-9. [Crossref] [PubMed]
Cite this article as: Ghumman U, Lee B, Bigham D, Tsai E. Sarcopenia in cirrhosis: a clinical practice review. Ann Palliat Med 2025;14(3):255-268. doi: 10.21037/apm-24-173

Download Citation