10.1111@apt.15571

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Received: 22 April 2019 | First decision: 20 May 2019 | Accepted: 13 October 2019

DOI: 10.1111/apt.15571

Review article: malnutrition/sarcopenia and frailty in patients


with cirrhosis

Chalermrat Bunchorntavakul1 | K. Rajender Reddy2

1
Division of Gastroenterology
and Hepatology, Department of Summary
Medicine, Rajavithi Hospital, College of Background: Malnutrition/sarcopenia and frailty are common in patients with cir‐
Medicine, Rangsit University, Bangkok,
Thailand rhosis and are associated with poor outcomes.
2
Division of Gastroenterology Aim: To provide an overview of data on the importance, assessment and manage‐
and Hepatology, Department of
ment of malnutrition/sarcopenia and frailty in cirrhosis.
Medicine, University of Pennsylvania,
Philadelphia, PA, USA Methods: A literature search was conducted in PubMed and other sources, using
the search terms “sarcopenia,” “muscle,” “malnutrition,” “cirrhosis,” “liver” and “frailty”
Correspondence
K. Rajender Reddy, Division of from inception to April 2019, to identify the relevant studies and international
Gastroenterology and Hepatology,
guidelines.
Department of Medicine, University of
Pennsylvania, 2 Dulles, 3400 Spruce Street, Results: The prevalence of malnutrition/sarcopenia in cirrhosis is 23%‐60%. Frailty
Philadelphia, PA 19104, USA.
generally overlaps with malnutrition/sarcopenia in cirrhosis, leading to increased
Email: reddyr@pennmedicine.upenn.edu
morbidity and mortality. Rapid nutritional screening assessment should be per‐
formed in all patients with cirrhosis, and more specific tests for sarcopenia should
be performed in those at high risk. The pathogenesis of malnutrition/sarcopenia in
cirrhosis is complex/multifactorial and not just reduction in protein/calorie intake.
Hyperammonemia appears to be the main driver of sarcopenia in cirrhosis through
several molecular signalling pathways. Nutritional management in malnourished pa‐
tients with cirrhosis should be undertaken by a multidisciplinary team to achieve
adequate protein/calorie intake. While the role of branched‐chained amino acids
remains somewhat contentious in achieving a global benefit of decreasing mortality‐
and liver‐related events, they, and vitamin supplements, are recommended for those
with advanced liver disease. Novel strategies to reverse sarcopenia such as hormone
supplementation, long‐term ammonia‐lowering agents and myostatin antagonists,
are currently under investigation.
Conclusions: Malnutrition/sarcopenia and frailty are unique, inter‐related and multi‐
dimensional problems in cirrhosis which require special attention, prompt assessment
and appropriate management as they significantly impact morbidity and mortality.

The Handling Editor for this article was Dr Colin Howden, and this uncommissioned
review was accepted for publication after full peer‐review.

Aliment Pharmacol Ther. 2019;00:1–14. wileyonlinelibrary.com/journal/apt


© 2019 John Wiley & Sons Ltd | 1
2 | BUNCHORNTAVAKUL and REDDY

1 | I NTRO D U C TI O N 2 | A S S E S S M E NT A N D D I AG N OS I S O F
M A LN U TR ITI O N/ SA RCO PE N I A A N D
Malnutrition (or undernutrition) is a general term used to describe a FR A I LT Y I N C I R R H OS I S
nutrition‐related disorder resulting from lack of intake or uptake of nu‐
trition that leads to altered body composition (decreased fat‐free mass) The assessment of malnutrition/sarcopenia appears to be more
1
and body cell mass. Sarcopenia is defined by generalised reduction in complicated among patients with cirrhosis as most conventional
muscle mass and function due to ageing (primary sarcopenia), or acute methods may have some limitations.1,5,7-9 Estimation of nutri‐
or chronic illness (secondary sarcopenia), including cirrhosis. Frailty is tional status and muscle mass by anthropometry, such as body
a multi‐dimensional construct that represents the end‐manifestation mass index (BMI) and mid‐arm muscle circumference (MAMC),
of derangements of multiple physiologic systems leading to decreased may be influenced by features such as ascites and peripheral
physiologic reserve and increased vulnerability to health stressors.2 fluid retention. Several laboratory tests for evaluating nutritional
These three conditions are increasingly common among patients with status in general patients, such as serum albumin and retinol‐
cirrhosis, particularly in those with decompensated liver disease, and in binding protein, are produced by the liver and are decreased in
fact relatively overlap (but not identical), and all contributing to physi‐ patients with advanced cirrhosis, with or without the presence
cal deconditioning and increased mortality.1 The main components of of malnutrition. Subjective global assessment (SGA), either gen‐
frailty are sarcopenia and physical impairment, which result in a cumu‐ eral SGA or liver‐specific SGA, such as the Royal Free Hospital
lative decline in physiologic reserve.3 It has clearly been shown that in Global Assessment, can be affected by altered laboratory tests
those with cirrhosis and with malnutrition/sarcopenia/frailty there is and physical deconditioning from advanced liver disease.1,7,10
reduced quality of life and survival, increased rates of cirrhotic com‐ Bioelectrical impedance analysis (BIA) also depends on stable
plications and infections and have worse outcomes following surgery hydration status, which may be altered in those with cirrhosis.1
and liver transplantation (LT).1,3-5 The pathogenesis of these conditions Thus, the measurement of thigh muscle thickness by ultrasound
is complex and multifactorial and is more than just simple reduction of has also been proposed.11 Despite some limitations, these con‐
5
protein and calorie intake. Hyperammonemia appears to be the main ventional methods are inexpensive, widely available and practical
driver of sarcopenia in cirrhosis through several molecular signalling and therefore should be used as initial nutritional risk assessment
pathways.5,6 Prompt diagnosis and correction of malnutrition/sarcope‐ (screening methods) for all patients with cirrhosis, while more ac‐
nia are very important, and have shown to improve the poor outcomes curate tests, such as dual‐energy x‐ray absorptiometry (DEXA)
to variable extents.1,4 Nutritional counselling and management in mal‐ and computed tomographic (CT) scan to measure muscle area at
nourished patients with cirrhosis should be performed by a multidisci‐ L3, can be used for the definite diagnosis of malnutrition/sarco‐
plinary team to be able to achieve adequate caloric/protein intake and penia in cirrhosis.1,5,7-11 (Table 1 and Figure 1) In addition, frailty
to avoid hypomobility. Branched‐chained amino acids (BCAA) and vi‐ has recently emerged as a strong predictor of outcomes in pa‐
tamin supplementations are recommended for patients with advanced tients with cirrhosis, independent of liver function, and has made
liver disease.1,4 Other and novel strategies to reverse sarcopenia are its way into decision‐making within LT. 2 Therefore, the American
6
currently under investigation. Society of Transplantation has recommended risk stratification

TA B L E 1 Screening and diagnosis tools for sarcopenia in cirrhosis

Anthropometry BIA US DEXA CT/MRI

Measures Overall body size Prediction equations Thickness of muscle Whole‐body and X‐sectional area and
(BMI, MAC), to calculate lean and (thigh muscle regional integrity of specific
predicted muscle fat mass (by passing thickness) and sub‐ fat, lean, bone mineral muscle and adipose
(MAMC), predicted an electrical current cutaneous adipose content tissue groups (L3‐SMI,
visceral adipose tis‐ through the body) tissue, echogenicity and BMD muscle attenuation)
sue (WC, W:H, TSF) for tissue integrity
Simplicity +++ ++ + + +
Cost − + + ++ ++/+++
Validity + + +/++ ++ +++
Clinical ++ + + + +
application
Research −/+ + + ++/+++ +++
application

Abbreviations: BIA, bioelectrical impedance analysis; BMD, bone mass density; BMI, body mass index; CT, computed tomographic; DEXA, dual‐en‐
ergy x‐ray absorptiometry; MAC, mid‐arm circumference; MAMC, mid‐arm muscle circumference; MRI, magnetic resonance imaging; SMI, skeletal
muscle index; TSF, triceps skin fold; WC, waist circumference; W:H, waist/hip ratio; US, ultrasound.
BUNCHORNTAVAKUL and REDDY | 3

F I G U R E 1 Nutritional screening and Cirrhosis and advanced chronic liver disease


assessment in patients with cirrhosis
[adapted from EASL Clinical Practice Child C Child A or B
Guidelines on nutrition in chronic liver Assess fluid retention
disease. J Hepatol 2019;70(1):172‐193]; Estimate dry weight if needed
with permission. Abbreviations: BIA, BMI
<18.5 18.5-29.9 ≥30
bioelectrical impedance analysis; Nutritional assessment
BMI, body mass index; CT, computed Underweight Obesity and LSM
Screen for malnutrition
tomographic; DEXA, dual‐energy x‐ by screening tools* +
ray absorptiometry; LSM, lifestyle Consider assessing
modification; MAMC, mid‐arm muscle sarcopenia
High risk Medium risk Low risk
circumference; RFH, Royal Free Hospital;
SPPB, short physical performance battery;
Assess sarcopenia: Detailed nutritional
SGA, subjective global assessment • Consider CT to measure assessment (by dietician): Follow-up:
muscle area at L3 • SGA Re-assess at least
• Consider DEXA or BIA if • RFH-GA every 1 y
no fluid retention • Reported dietary intake

Sarcopenia Malnutrition No malnutrition


Treat:
• Nutritional supplementation
• Appropriate follow-up (every1-3 mo in the 1st y) *e.g. MAMC,HGS,SPPB,SGA

for LT candidates by using standardised simple frailty assessment cirrhosis and such a feature has been correlated with adverse clini‐
tools such as the Liver Frailty Index, Clinical Frailty Scale (CFS), cal outcomes.6,22
Six‐minute walk test, Karnofsky Performance Status scale and
Short Physical Performance Battery (SPPB). 2,12-16 Frailty tools
have been best studied in the outpatient setting (or in the pa‐ 3 | PR E VA LE N C E O F M A LN U TR ITI O N/
tients’ with steady state). 2 SA RCO PE N I A A N D FR A I LT Y I N C I R R H OS I S
The diagnosis of sarcopenia virtually requires direct quantifica‐
tion of skeletal muscle mass by cross‐sectional imaging.1 CT scan Malnutrition and sarcopenia are common in patients with cirrhosis
analysis at L3 level, reported as skeletal muscle index (L3‐SMI), is and appear to be correlated strongly with the severity of cirrhosis.
a specific method to quantify muscle mass that has been shown In a prospective study of 1053 patients with cirrhosis in Italy, the
to be of higher accuracy in the diagnosis of sarcopenia in cirrhosis prevalence of protein‐calorie malnutrition evaluated by anthropom‐
than anthropometry or DEXA scan.1,5,7,17 Notably, it is currently the etry was 23%, 44% and 57% in patients with cirrhosis Child‐Pugh
most commonly utilised method in clinical studies investigating sar‐ (CP)‐A, CP‐B and CP‐C respectively. 23 Both adipose tissue and mus‐
copenia in cirrhosis. CT scan is commonly used for hepatocellular cle tissue can be depleted; female patients more frequently develop
carcinoma (HCC) surveillance in patients with cirrhosis and as such a depletion in fat deposits while males more rapidly lose muscle
may serve a dual purpose of calculating L3‐SMI, although special tissue.1 In a Canadian study of 142 patients awaiting LT, the preva‐
software may be required. All measures require normal values that lence of sarcopenia diagnosed by L3‐SMI was 51%. 24 Sarcopenia
are based on age, ethnicity and gender (lower predictive validity in was more prevalent in males vs females (54% vs 21%, P < .001) and
women).1,7,18 The mean muscle mass of Asians is approximately 15% increased with the CP class (10%, 34% and 54% for CP‐A, CP‐B
lower than that of Western population after height adjustments, and CP‐C, respectively, P = .007). 24 Interestingly, the prevalence
and ethnicity‐specific diagnostic criteria for sarcopenia may be nec‐ of sarcopenia increased with the advancing stages of liver disease
essary.19 Based on data from the US, the L3‐SMI cut‐off values of in males but not in females. There has been limited data regarding
less than 50 cm2/m2 in men and 39 cm2/m2 in women have been the prevalence of sarcopenia in patients with cirrhosis in Asia. In a
suggested for the diagnosis of sarcopenia in patients with cirrho‐ Japanese study of 807 patients with chronic liver disease, sarcope‐
sis as they correlated best with waitlist mortality.1,20 The Japanese nia was encountered in 4%, 5% and 17% of patients with chronic
Guideline, however, has suggested using cut‐off values of 42 cm2/ hepatitis, cirrhosis CP‐A and cirrhosis CP‐B/C respectively. 25
2 2 2 21
m for men and 38 cm /m for women to define sarcopenia. Apart Similar to sarcopenia, frailty is common in patients with cirrho‐
from quantity, another consideration is the quality of skeletal mus‐ sis, particularly in those with decompensated liver disease; however,
cle which can also be assessed indirectly by CT scan. CT attenuation the prevalence of frailty is somewhat more difficult to report pre‐
of the muscles in patients with cirrhosis appears to be lower (in‐ cisely since there are several assessment tools, cut‐offs, and some‐
dicative of fatty infiltration or myosteatosis) than in those without times being reported as a continuous index.3 In outpatient setting,
4 | BUNCHORNTAVAKUL and REDDY

the prevalence of frailty was 18% (defined by CFS > 4) among 300 beta blockers use may further contribute to malnutrition in cirrho‐
16
patients with cirrhosis from various aetiologies (28% had ascites). sis7,36 (Figure 2). Alteration of macronutrient metabolism is a corner‐
while the prevalence of frailty was up to 43% (defined by Fried stone mechanism contributing to malnutrition in cirrhosis.7 Protein
Frailty Index ≥ 3) among 542 patients with cirrhosis referred for LT and amino acids metabolism is abnormal due to increased protein
(52% had ascites). 26 catabolism, decreased protein synthesis, increased ureagenesis and
Obesity is frequently observed in cirrhosis (20%‐40%), regard‐ decreased serum BCAA/aromatic amino acids (AAA) ratio (Fischer's
less of the aetiology of liver disease, and patients with cirrhosis may ratio).8,36-38 Notably, BCAA are reduced due to increased utilisation
develop simultaneous loss of skeletal muscle and gain of adipose as the energy source and for ammonia disposal in the skeletal muscle,
tissue, culminating in the condition of sarcopenic obesity. 27,28 In an while AAA are increased due to decreased metabolism from liver in‐
analysis of 678 patients with cirrhosis (most were CP‐B), sarcopenia, sufficiency and portosystemic shunting, and increased release from
sarcopenic obesity and myosteatosis was present in 43%, 20% and protein degradation in the liver and muscle.37 The low Fischer's ratio
28
52% respectively. has been shown to be associated with the pathogenesis of compli‐
Aetiology of cirrhosis appears to have less impact on nutrition cations related to cirrhosis, such as hepatic encephalopathy (HE).38
status, although some studies have reported that patients with al‐ Carbohydrate metabolism is abnormal and includes increased gluco‐
coholic cirrhosis (particularly with active drinking) have higher prev‐ neogenesis, increased insulin resistance and glucose tolerance, and
alence of malnutrition/sarcopenia when compared to non‐alcoholic impaired hepatic glycogen synthesis.8,36 Taken together, cirrhosis is
8,29,30
aetiologies. Apart from macronutrients, micronutrient and a state of accelerated starvation in which these effects are evident
vitamin deficiencies are frequently seen in patients with cirrhosis. after a short overnight fast (>6 hours) and which may resemble the
Patients with cholestasis are at higher risk of having fat‐soluble rate of fat and protein catabolism of healthy subjects undergoing
vitamin deficiencies, particularly vitamin A and D.31 Vitamin D de‐ 2‐3 days of starvation.39 Abnormal lipid metabolism includes in‐
ficiency is almost universal in patients with advanced cirrhosis, par‐ creased lipolysis, oxidation of nonesterified fatty acids and ketogen‐
32,33
ticularly from hepatitis C, cholestasis and alcohol. Deficiencies esis.8,36 In cirrhotic patients, the resting energy expenditure (REE)
of thiamine, folate, magnesium and zinc are commonly observed in is typically increased (~1.3‐fold higher than predicted REE) and the
patients with advanced cirrhosis, especially from alcohol.34,35 total energy expenditure varies between 28 and 37.5 kcal/kg.BW/
day.1,4,6,40
The pathogenesis of sarcopenia in cirrhosis is even more complex
4 | PATH O G E N E S I S O F M A LN U TR ITI O N and is more than just reduced oral intake and negative macronutri‐
A N D SA RCO PE N I A I N C I R R H OS I S ents metabolism. Skeletal muscle mass is maintained by a balance
between protein synthesis, protein breakdown and regenerative
The pathogenesis of malnutrition is cirrhosis is complex and multifac‐ capacity regulated by muscle satellite cell function which involves
torial that includes reduced nutrient intake, altered protein/energy several metabolic‐tracer kinetics and molecular signalling pathways
metabolism, maldigestion and malabsorption. In addition, frequent (Figure 3).5,6 The two major skeletal muscle proteolytic pathways
fasting state and external factors, such as alcohol, infections, and are the ubiquitin‐proteasome pathway (UPP) and the autophagy

Reduced nutrient intake Altered protein/energy metabolism


• Anorexia, nausea/vomiting • Hypermetabolic state
• Altered taste • Altered protein and amino
• Impaired gastric expansion acids metabolism
(from ascites) • Altered CBH metabolism
• Impaired conscious state • Altered fat metabolism
• Protein/salt restriction

External factors
Malnutrition
• Infections
• SIRS
Maldigestion and
• Beta-blockers
malabsorption
• Alcohol
• Portosystematic shunting
• Bile salt and pancreatic Fasting status
enzymes deficiency
• GI bleeding F I G U R E 2 Factors contributing to
• Enteropathy malnutrition in cirrhosis. Abbreviations:
• Altered mental status
CBH, carbohydrates; GI, gastrointestinal;
• Bacterial overgrowth • Before and after SIRS, systemic inflammatory response
medical procedures
syndrome
BUNCHORNTAVAKUL and REDDY | 5

F I G U R E 3 Pathogenesis and potential Liver transplant


therapeutic options for sarcopenia in
Potential Portal HT Ammonia
cirrhosis. Abbreviations: ATB, antibiotics; treatments
BCAA, branched‐chain amino acids;
Lower
mTOR, mammalian target of rapamycin TIPS ammonia Nutrition support

Mitochodrial Nutrient
Myostatin
dysfunction intake

M Follistatin
Endotoxin ito Starvation
p Myostatin antagonists
ag rote response
Systemic en ct mTOR
ts ive
inflammation AT mTOR
B
An , pro agonists
tio bio
xid ti BCAA
Testosterone an cs
ts
Growth hormone
BCAA supplement
Hormone supplement Anaplerotic agents
Vitamin D supplement
Exe
Decreased muscle rcise Physical
Vitamin D inactivity
mass and contractile
function

system. 5 The UPP is the major muscle breakdown pathway, and in‐ Follistatin, a negative regulator of myostatin and other muscle
volves the tagging of protein by conjugation with ubiquitin and then growth inhibitors of TGF‐β family, has also been proposed to me‐
subsequent degradation by the 26S proteasome, and both physical diate liver‐muscle axis.46,54 It is secreted by the hepatocyte and is
inactivity and systemic inflammation (common in cirrhosis) activate regulated by the glucagon‐to‐insulin ratio.46,54 Baseline plasma lev‐
5
the UPP. Several potential mediators of the liver‐muscle axis have els of follistatin are similar or slightly elevated in patients with cir‐
been proposed including hyperammonemia,41-44 endotoxemia,45 de‐ rhosis and chronic inflammatory conditions as compared to healthy
46 47,48
creased follistatin, testosterone and growth hormone. In addi‐ controls55-57; however, the capacity to acutely secrete follistatin is
tion, amino acid perturbations, specifically reduction in the BCAA, impaired in patients with cirrhosis.46 The cause of altered follistatin
l‐leucine and consequent impaired global protein synthesis has also release in cirrhosis in unknown, but may be related to several factors
been reported to contribute to sarcopenia in cirrhosis. 1,37,49 such as decreased hepatic synthetic function, abnormal glucose me‐
Both hepatocellular dysfunction and portosystemic shunting tabolism, and glucagon and insulin resistance.46
contribute to impaired ureagenesis and hyperammonemia in cir‐
rhosis. Apart from the liver, skeletal muscle is the main source for
4.1 | Post‐transplant sarcopenia
ammonia disposal, which is increasingly prominent when advanced
cirrhosis evolves. In patients with cirrhosis, decreased muscle mass Unlike other complications of cirrhosis that are generally improved
compromises the place to detoxify ammonia and vice versa in that (or even reversed) after LT, the course of sarcopenia following LT is
hyperammonemia has shown to be the main driver of sarcopenia, variable with either improvement, remaining unchanged or continu‐
mainly through the myostatin‐mediated pathway. 5,6 Myostatin is ing to decline.58-63 Further reduction in skeletal muscle mass follow‐
a potent autocrine growth inhibitor (member of the transforming ing LT is most intense during the 1st year and may reverse thereafter,
growth factor beta (TGF‐β) family) produced by myocytes that in‐ while muscle functions generally improve early after LT and before
hibits skeletal muscle growth and reduces muscle mass through the improvement in muscle mass.60,61,63,64 Increased weight and BMI
41,50
impaired mammalian target of rapamycin (mTOR) signalling. are commonly observed following LT (median weight gain is 4‐6 kg at
Hyperammonemia upregulates myostatin expression by inducing 1 year and 8‐10 kg at 3 year); however, much of this weight gain is an
transcriptional regulation of myostatin via NF‐κB‐mediated path‐ increase in fat mass and sarcopenic obesity can be increasingly seen
way.41,50 In addition, hyperammonemia also mediates muscle au‐ in this population.58-61
42 51
tophagy and impair muscle contractility. Notably, BCAA have The underlying pathogenesis and mediators of post‐LT sarco‐
several potential benefits in maintaining liver‐muscle axis in cirrhosis penia are unclear. Several immunosuppressive agents, especially
including the stimulation of protein synthesis, inhibition of proteoly‐ corticosteroids and mTOR inhibitors, have negative impact on skel‐
sis and autophagy, and improving insulin resistance and β‐cell func‐ etal muscle and appear to be the key drivers of post‐LT sarcope‐
tion.37,49,52,53 In the presence of hyperammonemia, serum BCAA nia (Table 2).65-70 Although hyperammonemia resolves after LT, the
levels are further decreased as BCAA are important source of gluta‐ reversibility of hyperammonemia‐induced signalling responses and
mate, which are increasingly used to detoxify ammonia by glutamine impaired protein synthesis following LT are not known.6 A prospec‐
37,49,52
synthetase in the skeletal muscle and also in the brain. tive study in 53 LT recipients (100% received calcineurin inhibitors,
6 | BUNCHORNTAVAKUL and REDDY

TA B L E 2 Effects of
Clinically relevant
immunosuppressive agents on skeletal
Agents Effects of skeletal muscle strength of effect
muscle
Corticosteroids65 Induce muscle protein catabolism +++
Non‐specific type II fiber atrophy
(steroid‐induced myopathy)
Calcineurin inhibitors66,67 Inhibit muscle growth and hypertrophy ++
(intracellular calcineurin activation
regulates genes involved in skeletal
muscle growth and remodeling)
Increase myostatin expression
Prevent a switch in the type of muscle fiber
from slow to fast fibers
mTOR inhibitors68,69 Block protein synthesis responses and ++
accelerate autophagy
MMF70 No/minimal effects –/+
A case of reversible MMF‐induced myopa‐
thy has been reported

Abbreviations: MMF, mycophenolate mofetil; mTOR, mammalian target of rapamycin.

15% received mTOR inhibitors and 21% received corticosteroids) re‐ itself (Figure 4).6,7,71-76 Muscle is crucial for mobility, metabolic regu‐
vealed that the skeletal muscle expression of myostatin was higher lation of glucose and lipids, heart and respiratory function, immune
and that of UPP components lower in LT recipients than in con‐ function, and cytokine activity.7 Thus, there is a strong interplay be‐
62
trols. This may suggest that epigenetic changes in the regulatory tween malnutrition, sarcopenia, physical deconditioning and frailty
molecules/mediators can result in long‐term or persistent sarcope‐ in cirrhosis. A number of cross‐sectional and longitudinal studies
nia even after LT.6 using different diagnostic methods have reported that survival rates
are lower in cirrhotic patients with sarcopenia.6,7,71-74 The impact of
sarcopenia on prognosis in cirrhosis is beyond the degree of portal
5 | CO N S EQ U E N C E S O F M A LN U TR ITI O N hypertension and MELD score,73,74 and in fact, the modification of
SA RCO PE N I A A N D FR A I LT Y I N C I R R H OS I S MELD to include sarcopenia (Sarcopenia‐MELD) has shown to im‐
prove the prediction of mortality in patients with cirrhosis, primarily
The presence of malnutrition and/or sarcopenia in cirrhosis is associ‐ in patients with low MELD (score < 15).77 Interestingly, in Asian pop‐
ated with worsened clinical outcomes in various (almost all) aspects, ulation‐based studies, sarcopenia in Asian populations had more im‐
including survival, in addition to the effects from the liver disease pact on mortality related to sarcopenia than in Western populations

Encephalopathy* Bacterial infection Physical activity

Quality of life* Overall survival* Peri/post-operative


complications

Advanced Malnutrition
liver disease
Frailty

In LT
Sarcopenia
In HCC
Waitlist mortality*
Overall survival*
Peri-LT complications*
Treatment-related
complications* Post-LT survival
F I G U R E 4 Consequences of
malnutrition/sarcopenia/frailty in
Metabolic complications
* Supported by strong evidence cirrhosis. Abbreviation: LT, liver
(sarcopenic obesity)
transplantation
BUNCHORNTAVAKUL and REDDY | 7

TA B L E 3 Potential therapeutic strategies for malnourished/sarcopenia patients with cirrhosis

Intervention Administration Mechanism Potential outcomes

High energy and protein Ensure total calorie intake • Improves nitrogen balance • Improves muscle mass
diet1,4 of ≥ 35 kcal/kg/d and • Decreases skeletal muscle proteolysis • Improves quality of life
protein intake ≥ 1.5 g/kg/d • Improves liver functions
(if oral intake is inad‐ • Reduces liver‐related
equate, a period of EN is complications
recommended) • Improves survival
Late evening snack88-90 ≥50 g of complex carbohy‐ • Decreases lipid oxidation and improves • Improves muscle mass,
drate snack/meal with pro‐ nitrogen balance • Improves quality of life
tein, preferably high BCAA • Decreases skeletal muscle proteolysis dur‐ • Improves liver functions
ing overnight fasting
BCAA supplementation BCAA 0.25 g/kg/d • Activates muscle protein synthesis through • Improves muscle mass
53,91-93
supplementation mTOR signalling pathway • Improves quality of life
• Decreases muscle autophagy • Improves HE
• Helps detoxifying ammonia • Improves liver functions
• Improves event‐free survival
Exercise 102-107,130 A combination of aerobic • Activates IGF‐1 → increased mTOR and • Improves functional capacity
and resistance exercise in downregulation of muscle breakdown via and fatigue,
moderate intensity (under the UPP • Increases muscle mass and
supervision) • Increased phosphatidic acid in the muscle strength
that directly stimulates mTOR signalling • Decreases body fat
• Increased expression of follistatin mRNA in • Decreases HVPG
the liver and plasma follistatin
Testosterone Testosterone IM or gel for • Activates androgen receptors in muscle • Increases muscle and bone
supplement5,48 patients with low serum which inhibits myostatin, activates muscle mass
testosterone protein synthesis via the mTOR pathway • Increases haemoglobin
and downregulates the UPP • Reduces fat mass
• Reduces HbA1c
Zinc supplement114,115 150‐200 mg of elemental • Improves symptoms of zinc deficiency, for • Improves taste sensation
zinc/d example, altered taste, loss of appetite, and • May improve liver functions
encephalopathy and mental status
Vitamin D Vitamin D 600‐1000 IU/d • Prevents negative effects of vitamin D • Reduces bone loss
supplement116,117 (preferably with calcium deficiency on musculoskeletal and other • May increase BMD
1000‐1500 mg/d); correct systems • May improve musculoskeletal
deficiency if in vitamin D • Vitamin D deficiency is associated with functions (eg increase muscle
levels <20 ng/mL, to reach increased mortality in cirrhosis strength and reduced fall)
>30 ng/mL
Long‐term ammonia‐ Rifaximin, lactulose, LOLA, • Downregulates myostatin‐mediated • Theoretically may increase
lowering strategies5,43 anaplerotic agents pathway muscle mass (no human
• Decreases muscle autophagy studies)
• Improves muscle contractility • Improves HE
Reversal of portal TIPS placement • Reduces HVPG • Reduces ascites
hypertension121,122 • May reduce metabolic rate • May increase muscle mass
• May improve nutrient absorption and • Can precipitate HE and ACLF
systemic inflammation
Antagonising my‐ Myostatin antagonists, fol‐ • Downregulates myostatin‐mediated • Improves muscle mass and
ostatin‐mediated listatin, mTOR agonists pathway functions in elderly (no studies
pathway6,125-127 • Enhances mTOR signalling in cirrhosis)

Abbreviations: ACLF, acute‐on‐chronic liver failure; BCAA, branched‐chain amino acids; BMD, bone mass density; EN, enteral nutrition; HE, hepatic
encephalopathy; HVPG, hepatic venous portal gradient; IGF, insulin‐like growth factor; LOLA, L‐ornithine‐L‐aspartate; mTOR, mammalian target of
rapamycin; TIPS, transjugular intrahepatic portosystemic shunt; UPP, ubiquitin‐proteasome pathway.

(HR 2.45 vs HR 1.45 respectively).71 The cause(s) of higher mortal‐ malnutrition/sarcopenia due to reduced muscle mass and contrac‐
ity is, however, not as evident though both increased risk of infec‐ tile function.79,80 Sarcopenia may also impair diaphragmatic work
6,72
tion and HE may be contributing factors. HE is more frequent due to reduced muscle mass and this event may facilitate pulmo‐
in sarcopenic than nonsarcopenic cirrhotic patients.78 Physical ac‐ nary complications especially in the context of surgery.69 In pa‐
tivity and quality of life are likely to be impaired in patients with tients with HCC, sarcopenia is an independent prognostic factor of
8 | BUNCHORNTAVAKUL and REDDY

decreasing survival, and increasing treatment‐related complications


6 | M A N AG E M E NT O F M A LN U TR ITI O N
and mortality. 22,81 In addition, sarcopenic obesity and myosteatosis
A N D SA RCO PE N I A I N C I R R H OS I S
are independently associated with a higher long‐term mortality in
cirrhosis. 28
6.1 | Nutritional support and BCAA
In the setting of LT, malnutrition, sarcopenia and frailty ad‐
supplementation
versely impact outcomes in patients on the waitlist, in the peri‐
LT and post‐LT period.1,2,6,12,13 The presence of sarcopenia is Nutritional counselling should be provided for malnourished cir‐
predictive of waitlist mortality in addition to MELD score. 6,24 rhotic patients, when possible, by a multidisciplinary team, and thus
Furthermore, the presence of frailty is associated with waitlist helping patients to achieve adequate caloric and protein intake.1,4
mortality independent of MELD and CP scores. 2,12,13,82 Of note, General nutritional recommendations and potential therapeutic
frailty appears to be a more useful indicator for functional ca‐ strategies for malnutrition/sarcopenia in patients with cirrhosis are
pacity or quality of life in LT candidates, than the presence of summarised in Figure 5 and Table 3. It should be emphasised that the
sarcopenia.14 During peri‐LT period, malnourished/sarcopenic optimal daily energy intake should not be lower than 35 kcal/kg/d (in
patients commonly have prolonged the length of intensive care non‐obese individuals) and protein intake should not be lower than
unit and hospital stay, longer time of mechanical ventilation 1.2‐1.5 g/kg/d (amount of protein > 1.5 g/kg/d should be ingested
and higher rate of infections compared with those who are well to replenish malnourished and/or sarcopenic state).1,4 The daily in‐
7,12,62,63,73,83
nourished. take should be divided into frequent small meals and/or snack (5‐6
Following LT, pre‐LT sarcopenia is associated with decreased times/d) and, of importance, late evening oral nutritional supplemen‐
medium‐term survival. 61,62,84,85 The severity of pre‐LT frailty has tation containing ≥ 50 g of complex carbohydrates must be included,
been shown to be an independent predictor of patient and graft as it has been shown to be more effective on substrate utilisa‐
survival. 86,87 LT recipients who have low performance status be‐ tion and nitrogen retention than daytime calorie supplementation
fore LT and do not show an improvement in performance status alone.1,4,88-90 Several studies have demonstrated that late evening
between 3 and 12 months after liver transplant have very poor snack or meal is an effective intervention to prevent and to reverse
survival. 86 Notably, sarcopenic obesity has been increasingly re‐ sarcopenia in cirrhosis, resulting in an improvement in quality of life
ported in LT recipients and may contribute to the development of and also liver functions. 88-90 Notably, when prescribing a low sodium
metabolic consequences of insulin resistance, diabetes mellitus, (<2‐3 g/d) diet, the increased risk of even lower food intake (due to
hyperlipidemia and possibly recurrent or de novo fatty liver, which unpalatability) should be balanced against its moderate advantage in
may eventually increase cardiovascular morbidity in the long the treatment of ascites.4
6,61,85
term. It should be noted that the degree of frailty and sar‐ The decreased levels of BCAA in the blood plasma (also
copenia should not be used as the sole criterion for delisting a pa‐ Fischer's ratio) is the hallmark of cirrhosis and which plays a role
tient for LT, but rather should be considered one of many criteria in pathogenesis of HE, sarcopenia and impaired liver regenera‐
when evaluating transplant candidacy and suitability. 2 Currently, tion. 37,38,52 Therefore, BCAA are a preferred source of amino acids
whether or not patients with low MELD and sarcopenia should in cirrhotic patients and BCAA supplementation seems theoreti‐
be prioritised for LT (MELD exception) is debatable at this stage. cally rationale. 37,52 (Figure 6) Among the BCAAs (leucine, isoleucine

Recommended total energy intake: 35-40 kcal/kg/d

Frequent meals and snacks (5-6 times/d)


Protein >1.2-1.5 g/kg/d
• Encourage vegetable proteins
• BCAA (0.25 g/kg/d) and leucine
enriched AA supplementation for
decompensated cirrhosis
CBH 50-70%
• Consider BCAA if encephalopathy
• Late-evening snack/meal
(>50 g complex CBH)
• Low glycemic index and high-fiber
CBH sources are preferred
• Reduce simple sugars, esp. fructose F I G U R E 5 General nutritional
• Restrict CBH if NAFLD and/or DM
recommendations for malnourished
Fat 10-20% patients with cirrhosis. Abbreviations: AA,
• High unsaturated fat
Sodium restriction <2-3 g/d, amino acids; BCAA, branched‐chain amino
is preferred acids; CBH, carbohydrates; DM, diabetes
if ascites and/or edema
• Include adequate mellitus; NAFLD, non‐alcoholic fatty liver
essential fatty acids Water restriction, if Na <130
disease
BUNCHORNTAVAKUL and REDDY | 9

F I G U R E 6 Possible targets and Prevention and Prevention and Repair and


outcomes of BCAA supplementation in treatment of treatment of regeneration of
the management of malnourished patients sarcopenia encephalopathy injured liver
with cirrhosis. Abbreviations: AAA,
aromatic amino acids; BCAA, branched‐ • Activates mTOR • Facilitates ammonia • Energy/protein source
chain amino acids; HCC, hepatocellular signaling pathway – detoxification (through • Stimulates HGF
carcinoma; HGF, hepatocyte growth stimulates protein glutamine synthetase) production by hepatic
factor; mTOR, mammalian target of synthesis (improves in skeletal muscle and stellate cells
muscle mass) brain • Enhances production
rapamycin
• Inhibits proteolysis • Normalizes Fischer’s of glutamine –
Improves insulin ratio – decrease brain stimulates liver

resistance and β-cell influx of AAA regeneration
function • Improves cerebral • Suppresses HCC
blood flow carcinogenesis

Muscle mass Encephalopathy* Liver functions


Muscle strength Quality of life* Liver biomarkers
Serum albumin Hospital stay
Liver events The grey boxes represent
*Supported by strong evidence
Survival potential modes of action
(there is low-moderate evidence
based on experimental evidence
for the other outcomes)

and valine), leucine is particularly crucial in increasing muscle pro‐ more adequate than low‐protein diets, and without negative im‐
tein synthesis. 38,44,53 Several clinical trials, including three RCTs pact on HE recovery.96
(n = 174,91 n = 64692 and n = 11693) and one large prospective study In malnourished patients with cirrhosis who are unable to
94
(n = 267), have demonstrated the positive effects of BCAA sup‐ achieve adequate dietary intake (even with oral supplements), a pe‐
plementation in malnourished cirrhotic patients, such as improve‐ riod of enteral nutrition (EN) is recommended.1 Oesophageal varices
ment in nutritional status, liver functions, HE, quality of life and are not an absolute contraindication for placing a nasogastric tube.4
37,38,52
event‐free survival. There were few unique observations Some, however, recommend withholding EN for 48‐72 h after acute
among these BCAA trials that included: (a) the BCAA dose ranged variceal bleeding because EN may increase splanchnic blood flow,
from 12 to 30 g/d; (b) beneficial effects were predominantly seen which in turn may increase portal pressure and the risk of re‐bleed‐
in those with advanced liver disease and/or previous HE and (c) ad‐ ing.1,97 Percutaneous endoscopic gastrostomy placement is associ‐
herence to BCAA supplementation was moderate (75%‐90%) even ated with a higher risk of complications and should only be used in
in the context of RCT which may be partly explained by its unpalat‐ exceptional cases.4 Parenteral nutrition (PN) should be used in cir‐
ability for some patients. Accordingly, the European Guidelines on rhotic patients in whom oral and/or EN are ineffective or not fea‐
nutrition in liver disease have recommended long‐term oral BCAA sible.1,4 A meta‐analysis of nutrition therapy in cirrhosis (13 RCT; 9
supplement (0.25 g/kg/d) in patients with advanced cirrhosis in EN, 4 PN) revealed significant beneficial effects on clinical outcomes
order to improve event‐free survival or quality of life, particularly including mortality, overt HE and infections, although all included
when adequate nitrogen intake is not achieved by oral diet (grade trials were classed as having a high risk of bias.98 In general, stan‐
of recommendation B by ESPEN and evidence and Grade II‐1, C1 dard nutrition regimens can be utilised since specialised regimens (eg
by EASL).1,4 BCAA‐enriched, immune‐enhancing diets) have not been shown to
In patients with chronic HE, the optimal daily protein and en‐ improve morbidity or mortality in intervention trials.1 Meta‐analyses
ergy intake should not be lower than the general recommenda‐ of BCAA‐enriched EN formula and PN solution showed an improve‐
tions for patients with cirrhosis. Diets rich in vegetables and dairy ment in mental state, but no definite benefit in survival and other
protein are encouraged as they have higher Fischer's ratio and outcomes.99,100
possibly prebiotic properties (eg decreased intestinal transit time, Nutritional management of obese cirrhotic patients, particu‐
reduced intraluminal pH and increased faecal ammonia excre‐ larly with sarcopenia, is even more complicated and often requires
tion).1,95 BCAA supplementation should be considered to improve a specialised multidisciplinary team.1,27,101 In general, a nutritional
neuropsychiatric performance and to reach the recommended ni‐ and lifestyle program to achieve progressive weight loss (>5%‐10%)
trogen intake, particularly in those who are intolerant to dietary should be implemented.1 A tailored, moderately hypocaloric
1
proteins. It should be emphasised that protein intake should not (−500‐800 kcal/d) diet, including adequate protein intake (>1.5 g/
be restricted in patients with episodic HE admitted to hospital as kg/d) can be adopted to achieve weight loss without compromising
it increases protein catabolism rate.4 A small RCT in this context protein stores in obese cirrhotic patients.1,101 Furthermore, obese
showed that the normal content of protein intake is metabolically cirrhotic patients can be given EN and/or PN with a target energy
10 | BUNCHORNTAVAKUL and REDDY

of 25 kcal/kg/d and an increased target protein intake of 2.0‐2.5 g/ including altered taste and smell, poor appetite, hair loss, skin lesions,
4
kg/d. poor wound healing/liver regeneration, altered mental status or al‐
tered immune function.34,114 Zinc supplementation (150‐200 mg/d
taken with a meal to decrease the potential side effect of nausea) has
6.2 | Exercise
shown to at least improve taste sensation in cirrhosis.114,115 Likewise,
Physical inactivity and frailty are highly prevalent, independent pre‐ the data on vitamin D supplementation in improving outcomes are
dictors of morbidity and mortality in patients with cirrhosis.102,103 not robust but the data regarding poor outcomes in cirrhosis with
Consistent with the broader chronic disease literature, the experi‐ vitamin D deficiency are compelling.116,117 Research experience in
mental and clinical evidence for a benefit of exercise in cirrhosis is the elderly suggests that vitamin D supplementation may have ben‐
promising. Several small clinical trials have demonstrated significant efits on musculoskeletal system (eg increased bone density, mus‐
improvements in muscle health (mass, strength, functional capacity), cle strength and physical activity)118,119; however, whether these
quality of life, fatigue, cardiopulmonary fitness and reductions in findings can be extrapolated for those with cirrhosis is unknown.
the hepatic venous pressure gradient, without adverse events.102-107 Vitamin D level should be checked in all malnourished patients with
Unfortunately, most patients (63%‐92%) included in these trials were cirrhosis and supplemental vitamin D orally in those with vitamin D
cirrhosis CP‐A, while patients at the lower end of physical decondi‐ levels < 20 ng/mL, to reach serum 25(OH) vitamin D > 30 ng/mL is
tioning (CP‐B/C) have not been evaluated systematically so far.102,103 recommended.1
Patients with cirrhosis, whenever possible, can be encouraged to
avoid hypomobility and to progressively increase physical activity
6.4 | Other strategies
to prevent and/or ameliorate sarcopenia.1 It is reasonable to recom‐
mend 30‐to‐60‐minute sessions of supervised moderated intensity Long‐term ammonia‐lowering strategies may result in increased
exercise combining both aerobic and resistance training to achieve muscle mass and strength; however, apart from indirectly by BCAA
≥150 min/wk for cirrhotic patients.102,103 Aerobic training would supplementation, the data are derived from preclinical studies and
be particularly relevant to improve overall fitness, while resistance require further validation in humans.6,43 A 4‐week ammonia‐lower‐
training would help reverse sarcopenia, improve strength and bal‐ ing therapy by L‐ornithine‐L‐aspartate and rifaximin has shown to
ance, and bone mineral density.102,103 Whether the use of weights improve skeletal muscle mass and functions in hyperammonemic
is appropriate for patients with varices is unclear, and thus weight‐ portacaval anastomosis rats.43 Potential novel strategies to lower
bearing exercises or repetitions with small weights (eg 0.5‐1 kg) may muscle ammonia include the use of cell permeable esters of a alpha‐
be advised.103 ketoglutarate that can provide a direct anaplerotic influx with the
removal of ammonia as glutamine.6 In addition, isoleucine and va‐
line may help detoxifying ammonia as anaplerotic substrates but the
6.3 | Hormones and micronutrient replacement
molecular and functional responses to these interventions have not
Testosterone and growth hormone have been evaluated in chronic been evaluated in clinical studies to lower muscle ammonia or re‐
liver disease to improve nutritional status and, potentially, muscle verse sarcopenia.6 Similarly, long‐term ammonia lowering by rifaxi‐
mass in cirrhosis but have not been consistently beneficial.6,48,108-110 min plus lactulose has shown to improve HE and survival in patients
It is possible that the effects of testosterone may be blunted by with cirrhosis and overt HE,120 but its impact on skeletal muscle has
increased aromatase activity in patients with cirrhosis which then not been directly investigated.
enhances the conversion of testosterone to estradiol.6 Thus, admin‐ Reducing portal pressure by transjugular intrahepatic portosys‐
istration of an aromatase inhibitor has shown to improve nutritional temic shunt (TIPS) placement may have beneficial effects on muscle
and endocrine abnormalities in rats with portocaval anastomosis.111 mass.121 A small uncontrolled trial demonstrated a significant in‐
A RCT (n = 101) has demonstrated that testosterone therapy for crease in 72% of patients following successful TIPS placement.122
1 year in men with cirrhosis and low serum testosterone safely in‐ Furthermore, the improvement of sarcopenia after TIPS was accom‐
creases muscle mass, bone mass and haemoglobin, and reduces fat panied by lower mortality (43.5%) compared with patients in whom
mass.48 Nevertheless, despite controversies, cautions should be ex‐ sarcopenia had not improved (9.8% vs 43.5%, P = .007).122 It is to be
ercised when using long‐term testosterone replacement therapy due recognised that apart from reduce portal pressure, many other fac‐
to the possibility of increased risk of cardiovascular events, poly‐ tors such as reduced ascites, reduced metabolic rate and improved
cythemia, cholestasis, prostate cancer and HCC.1,112,113 nutrition could have contributed to the beneficial effects.5,121 Of
Specific evidence about the beneficial effect, especially on mus‐ note, caution is needed when performing TIPS in malnourished pa‐
cle mass, of micronutrients and vitamin supplementation in cirrhotic tients with cirrhosis as sarcopenia is a risk factor for the develop‐
patients is not available. However, confirmed clinically suspected ment of HE and acute‐on‐chronic liver failure after TIPS.123,124
deficiency should be treated based on accepted general recom‐ Novel approaches, such as myostatin antagonists,125-127 folli‐
1
mendations. Zinc and vitamin D deficiency are being increasingly statin,128 direct mTORC1 activators,53,129 antioxidants6 and mito‐
recognised to contribute to worse clinical outcomes in patients with chondrial protective agents,6 theoretically have potential benefits
cirrhosis. Zinc deficiency may manifest several ways in cirrhosis, on liver‐muscle axis, but have not been adequately evaluated in
BUNCHORNTAVAKUL and REDDY | 11

clinical studies. In sarcopenic rats with portocaval anastomosis, the 10. Morgan MY, Madden AM, Soulsby CT, Morris RW. Derivation and
inhibitory effect of myostatin is abolished by injections of follista‐ validation of a new global method for assessing nutritional status
in patients with cirrhosis. Hepatology. 2006;44:823‐835.
tin, resulting in increased skeletal muscle mass and muscle synthetic
11. Tandon P, Low G, Mourtzakis M, et al. A model to identify sar‐
rate.128 Notably, small RCTs (Phase II) evaluating myostatin antibody copenia in patients with cirrhosis. Clin Gastroenterol Hepatol.
have demonstrated positive effects on muscle mass in older and 2016;14:1473‐1480; e1473.
weak fallers126 and patients undergoing elective total hip arthro‐ 12. Wang CW, Feng S, Covinsky KE, et al. A comparison of muscle
function, mass, and quality in liver transplant candidates: results
plasty.127 Further clinical studies are required before these interven‐
from the functional assessment in liver transplantation study.
tions can be translated to clinical practice, especially for those with Transplantation. 2016;100:1692‐1698.
cirrhosis. 13. 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;66:564‐574.
AC K N OW L E D G E M E N T 14. Yadav A, Chang Y‐H, Carpenter S, et al. Relationship between sar‐
copenia, six‐minute walk distance and health‐related quality of life
Declaration of personal interests: None. in liver transplant candidates. Clin Transplant. 2015;29:134‐141.
15. Carey EJ, Steidley DE, Aqel BA, et al. Six‐minute walk distance
predicts mortality in liver transplant candidates. Liver Transplant.
AU T H O R S H I P 2010;16:1373‐1378.
16. Tandon P, Tangri N, Thomas L, et al. A rapid bedside screen to
Guarantor of the article: K. R. Reddy. predict unplanned hospitalization and death in outpatients with
Author contributions: C. Bunchorntavakul: conceptualised, cirrhosis: a prospective evaluation of the clinical frailty scale. Am J
searched and reviewed literature, created the tables and drafted the Gastroenterol. 2016;111:1759‐1767.
17. Giusto M, Lattanzi B, Albanese C, et al. Sarcopenia in liver cirrho‐
manuscript. KR Reddy: conceptualised and critically reviewed the
sis: the role of computed tomography scan for the assessment of
paper. muscle mass compared with dual‐energy X‐ray absorptiometry
Both authors approved the final version of the manuscript. and anthropometry. Eur J Gastro Hepatol. 2015;27:328‐334.
18. Peng S, Plank LD, McCall JL, Gillanders LK, McIlroy K, Gane EJ.
Body composition, muscle function, and energy expenditure in pa‐
ORCID tients with liver cirrhosis: a comprehensive study. Am J Clin Nutr.
2007;85:1257‐1266.
Chalermrat Bunchorntavakul https://orcid. 19. Chen L‐K, Liu L‐K, Woo J, et al. Sarcopenia in Asia: consensus re‐
org/0000-0002-8842-032X port of the Asian Working Group for Sarcopenia. J Am Med Dir
Assoc. 2014;15:95‐101.
K. Rajender Reddy https://orcid.org/0000-0002-4898-7778 20. Carey EJ, Lai JC, Wang CW, et al. A multicenter study to define sar‐
copenia in patients with end‐stage liver disease. Liver Transplant.
2017;23:625‐633.
REFERENCES 21. Nishikawa H, Shiraki M, Hiramatsu A, Moriya K, Hino K, Nishiguchi
S. Japan Society of Hepatology guidelines for sarcopenia in liver
1. Merli M, Berzigotti A, Zelber‐Sagi S, et al. Practice guidelines on disease (1st edition): Recommendation from the working group
nutrition in chronic liver disease. J Hepatol. 2019;70:172‐193. for creation of sarcopenia assessment criteria. Hepatol Res.
2. Lai JC, Sonnenday CJ, Tapper EB, et al. Frailty in liver transplan‐ 2016;46:951‐963.
tation: an expert opinion statement from the American Society of 22. Fujiwara N, Nakagawa H, Kudo Y, et al. Sarcopenia, intramuscular
Transplantation Liver and Intestinal Community of Practice. Am J fat deposition, and visceral adiposity independently predict the
Transplant. 2019;19:896‐1906. outcomes of hepatocellular carcinoma. J Hepatol. 2015;63:131‐140.
3. Ebadi M, Montano‐Loza AJ. Sarcopenia and frailty in the progno‐ 23. Merli M, Riggio O, Dally L. Does malnutrition affect survival in cir‐
sis of patients on the liver transplant waiting list. Liver Transplant. rhosis? PINC (Policentrica Italiana Nutrizione Cirrosi). Hepatology.
2019;25:7‐9. 1996;23:1041‐1046.
4. Plauth M, Bernal W, Dasarathy S, et al. ESPEN guideline on clinical 24. Tandon P, Ney M, Irwin I, et al. Severe muscle depletion in patients
nutrition in liver disease. Clin Nutr. 2019;38:485‐521. on the liver transplant wait list: its prevalence and independent
5. Sinclair M, Gow PJ, Grossmann M, Angus PW. Review article: sar‐ prognostic value. Liver Transplant. 2012;18:1209‐1216.
copenia in cirrhosis–aetiology, implications and potential thera‐ 25. Hiraoka A, Michitaka K, Ueki H, et al. Sarcopenia and two types of
peutic interventions. Aliment Pharmacol Ther. 2016;43:765‐777. presarcopenia in Japanese patients with chronic liver disease. Eur
6. Dasarathy S, Merli M. Sarcopenia from mechanism to diag‐ J Gastro Hepatol. 2016;28:940‐947.
nosis and treatment in liver disease. J Hepatol. 2016;65:1232‐ 26. Cron DC, Friedman JF, Winder GS, et al. Depression and frailty in
1244. patients with end‐stage liver disease referred for transplant evalu‐
7. Tandon P, Raman M, Mourtzakis M, Merli M. A practical approach ation. Am J Transplant. 2016;16:1805‐1811.
to nutritional screening and assessment in cirrhosis. Hepatology. 27. Berzigotti A, Garcia‐Tsao G, Bosch J, et al. Obesity is an indepen‐
2017;65:1044‐1057. dent risk factor for clinical decompensation in patients with cirrho‐
8. Zhao VM, Ziegler TR. Nutrition support in end‐stage liver disease. sis. Hepatology. 2011;54:555‐561.
Crit Care Nurs Clin North Am. 2010;22:369‐380. 28. Montano‐Loza AJ, Angulo P, Meza‐Junco J, et al. Sarcopenic obe‐
9. Santos LAA, Lima TB, Ietsugu MDV, Nunes HRC, Qi X, Romeiro sity and myosteatosis are associated with higher mortality in pa‐
FG. Anthropometric measures associated with sarcopenia in tients with cirrhosis. J Cachexia Sarcopenia Muscle. 2016;7:126‐135.
outpatients with liver cirrhosis. Nutr Diet. 2019. https​://doi. 29. Tai ML, Goh KL, Mohd‐Taib SH, Rampal S, Mahadeva S.
org/10.1111/1747-0080.12523​. [Epub ahead of print.] Anthropometric, biochemical and clinical assessment of
12 | BUNCHORNTAVAKUL and REDDY

malnutrition in Malaysian patients with advanced cirrhosis. Nutr J. a potential contributor to skeletal muscle wasting. Anest Analg.
2010;9:27. 2010;111:707‐709.
30. Roongpisuthipong C, Sobhonslidsuk A, Nantiruj K, 51. McDaniel J, Davuluri G, Hill EA, et al. Hyperammonemia results in
Songchitsomboon S. Nutritional assessment in various stages of reduced muscle function independent of muscle mass. Am J Physiol
liver cirrhosis. Nutrition. 2001;17:761‐765. Gastrointest Liver Physiol. 2016;310:G163‐170.
31. Phillips JR, Angulo P, Petterson T, Lindor KD. Fat‐soluble vitamin 52. Holecek M. Branched‐chain amino acid supplementation in treat‐
levels in patients with primary biliary cirrhosis. Am J Gastroenterol. ment of liver cirrhosis: Updated views on how to attenuate their
2001;96:2745‐2750. harmful effects on cataplerosis and ammonia formation. Nutrition.
32. Konstantakis C, Tselekouni P, Kalafateli M, Triantos C. Vitamin 2017;41:80‐85.
D deficiency in patients with liver cirrhosis. Ann Gastroenterol. 53. Tsien C, Davuluri G, Singh D, et al. Metabolic and molecular re‐
2016;29:297‐306. sponses to leucine‐enriched branched chain amino acid supple‐
33. Lim LY, Chalasani N. Vitamin d deficiency in patients with chronic mentation in the skeletal muscle of alcoholic cirrhosis. Hepatology.
liver disease and cirrhosis. Curr Gastroenterol Rep. 2012;14:67‐73. 2015;61:2018‐2029.
34. Mohammad MK, Zhou Z, Cave M, Barve A, McClain CJ. Zinc and 54. Hansen JS, Rutti S, Arous C, et al. Circulating follistatin is liver‐
liver disease. Nutr Clin Pract. 2012;27:8‐20. derived and regulated by the glucagon‐to‐insulin ratio. J Clin
35. Katayama K, Kawaguchi T, Shiraishi K, et al. The prevalence and Endocrinol Metab. 2016;101:550‐560.
implication of zinc deficiency in patients with chronic liver disease. 55. Sakamoto Y, Shintani Y, Harada K, Abe M, Shitsukawa K, Saito
J Clin Med Res. 2018;10:437‐444. S. Determination of free follistatin levels in sera of normal
36. Cheung K, Lee SS, Raman M. Prevalence and mechanisms of malnu‐ subjects and patients with various diseases. Eur J Endocrinol.
trition in patients with advanced liver disease, and nutrition man‐ 1996;135:345‐351.
agement strategies. Clin Gastroenterol Hepatol. 2012;10:117‐125. 56. Yuen MF, Norris S, Evans LW, Langley PG, Hughes RD. Transforming
37. Holecek M. Three targets of branched‐chain amino acid sup‐ growth factor‐beta 1, activin and follistatin in patients with hepa‐
plementation in the treatment of liver disease. Nutrition. tocellular carcinoma and patients with alcoholic cirrhosis. Scand J
2010;26:482‐490. Gastroenterol. 2002;37:233‐238.
38. Ooi PH, Gilmour SM, Yap J, Mager DR. Effects of branched chain 57. Tomoda T, Nouso K, Miyahara K, et al. Prognotic impact of serum
amino acid supplementation on patient care outcomes in adults follistatin in patients with hepatocellular carcinoma. J Gastroenterol
and children with liver cirrhosis: A systematic review. Clin Nutr Hepatol. 2013;28:1391‐1396.
ESPEN. 2018;28:41‐51. 58. Pravisani R, Soyama A, Isola M, et al. Chronological changes in
39. Owen OE, Reichle FA, Mozzoli MA, et al. Hepatic, gut, and renal skeletal muscle mass following living‐donor liver transplantation:
substrate flux rates in patients with hepatic cirrhosis. J Clin Investig. An analysis of the predictive factors for long‐term post‐transplant
1981;68:240‐252. low muscularity. Clin Transplant. 2019;33:e13495.
40. Greco AV, Mingrone G, Benedetti G, Capristo E, Tataranni PA, 59. Schütz T, Hudjetz H, Roske A‐E, et al. Weight gain in long‐term
Gasbarrini G. Daily energy and substrate metabolism in patients survivors of kidney or liver transplantation–another paradigm of
with cirrhosis. Hepatology. 1998;27:346‐350. sarcopenic obesity? Nutrition. 2012;28:378‐383.
41. Qiu J, Thapaliya S, Runkana A, et al. Hyperammonemia in cir‐ 60. Bergerson JT, Lee J‐G, Furlan A, et al. Liver transplantation arrests
rhosis induces transcriptional regulation of myostatin by an and reverses muscle wasting. Clin Transplant. 2015;29:216‐221.
NF‐kappaB‐mediated mechanism. Proc Natl Acad Sci USA. 61. Kallwitz ER. Sarcopenia and liver transplant: the relevance of too
2013;110:18162‐18167. little muscle mass. World J Gastroenterol. 2015;21:10982‐10993.
42. Qiu J, Tsien C, Thapalaya S, et al. Hyperammonemia‐mediated au‐ 62. Tsien C, Garber A, Narayanan A, et al. Post‐liver transplantation
tophagy in skeletal muscle contributes to sarcopenia of cirrhosis. sarcopenia in cirrhosis: a prospective evaluation. J Gastroenterol
Am J Physiol Endocrinol Metab. 2012;303:E983‐993. Hepatol. 2014;29:1250‐1257.
43. Kumar A, Davuluri G, Silva RNE, et al. Ammonia lowering reverses 63. Bhanji RA, Takahashi N, Moynagh MR, et al. The evolution and
sarcopenia of cirrhosis by restoring skeletal muscle proteostasis. impact of sarcopenia pre‐ and post‐liver transplantation. Aliment
Hepatology. 2017;65:2045‐2058. Pharmacol Ther. 2019;49:807‐813.
44. Davuluri G, Krokowski D, Guan B‐J, et al. Metabolic adaptation of 64. Kaido T, Tamai Y, Hamaguchi Y, et al. Effects of pretransplant sar‐
skeletal muscle to hyperammonemia drives the beneficial effects copenia and sequential changes in sarcopenic parameters after
of l‐leucine in cirrhosis. J Hepatol. 2016;65:929‐937. living donor liver transplantation. Nutrition. 2017;33:195‐198.
45. Kovarik M, Muthny T, Sispera L, Holecek M. The dose‐dependent 65. Gupta A, Gupta Y. Glucocorticoid‐induced myopathy:
effects of endotoxin on protein metabolism in two types of rat Pathophysiology, diagnosis, and treatment. Indian J Endocrinol
skeletal muscle. J Physiol Biochem. 2012;68:385‐395. Metab. 2013;17:913‐916.
46. Rinnov AR, Plomgaard P, Pedersen BK, Gluud LL. Impaired 66. Michel RN, Chin ER, Chakkalakal JV, Eibl JK, Jasmin BJ. Ca2+/calm‐
follistatin secretion in cirrhosis. J Clin Endocrinol Metab. odulin‐based signalling in the regulation of the muscle fibre pheno‐
2016;101:3395‐3400. type and its therapeutic potential via modulation of utrophin A and
47. Mowat NA, Edwards CR, Fisher R, McNeilly AS, Green JR, Dawson myostatin expression. Appl Physiol Nutr Metab. 2007;32:921‐929.
AM. Hypothalamic‐pituitary‐gonadal function in men with cirrho‐ 67. Semsarian C, Wu M‐J, Ju Y‐K, et al. Skeletal muscle hypertrophy
sis of the liver. Gut. 1976;17:345‐350. is mediated by a Ca2+‐dependent calcineurin signalling pathway.
48. Sinclair M, Grossmann M, Hoermann R, Angus PW, Gow PJ. Nature. 1999;400:576‐581.
Testosterone therapy increases muscle mass in men with cirrho‐ 68. Bodine SC, Stitt TN, Gonzalez M, et al. Akt/mTOR pathway is a
sis and low testosterone: a randomised controlled trial. J Hepatol. crucial regulator of skeletal muscle hypertrophy and can prevent
2016;65:906‐913. muscle atrophy in vivo. Nat Cell Biol. 2001;3:1014‐1019.
49. Kawaguchi T, Izumi N, Charlton MR, Sata M. Branched‐chain 69. Dasarathy S. Consilience in sarcopenia of cirrhosis. J Cachexia
amino acids as pharmacological nutrients in chronic liver disease. Sarcopenia Muscle. 2012;3:225‐237.
Hepatology. 2011;54:1063‐1070. 70. Galindo M, Cabello A, Joven B, et al. Mycophenolate mofetil in‐
50. Garcia PS, Cabbabe A, Kambadur R, Nicholas G, Csete M. Brief‐ duced myopathy in a patient with lupus nephritis. J Rheumatol.
reports: elevated myostatin levels in patients with liver disease: 2005;32:188‐190.
BUNCHORNTAVAKUL and REDDY | 13

71. Kim G, Kang SH, Kim MY, Baik SK. Prognostic value of sarcopenia a meta‐analysis of randomized controlled trials. J Gastroenterol
in patients with liver cirrhosis: A systematic review and meta‐anal‐ Hepatol. 2019;34:1143‐1152.
ysis. PLoS ONE. 2017;12:e0186990. 91. Marchesini G, Bianchi G, Merli M, et al. Nutritional supplementa‐
72. van Vugt JL, Levolger S, de Bruin RW, van Rosmalen J, Metselaar tion with branched‐chain amino acids in advanced cirrhosis: a dou‐
HJ, IJzermans JNM. Systematic review and meta‐analysis of the ble‐blind, randomized trial. Gastroenterology. 2003;124:1792‐1801.
impact of computed tomography‐assessed skeletal muscle mass 92. Muto Y, Sato S, Watanabe A, et al. Effects of oral branched‐chain
on outcome in patients awaiting or undergoing liver transplanta‐ amino acid granules on event‐free survival in patients with liver
tion. Am J Transplant. 2016;16:2277‐2292. cirrhosis. Clin Gastroenterol Hepatol. 2005;3:705‐713.
73. Kalafateli M, Mantzoukis K, Choi Yau Y, et al. Malnutrition and 93. Les I, Doval E, García‐Martínez R, et al. Effects of branched‐chain
sarcopenia predict post‐liver transplantation outcomes inde‐ amino acids supplementation in patients with cirrhosis and a pre‐
pendently of the model for end‐stage liver disease score. J vious episode of hepatic encephalopathy: a randomized study. Am
Cachexia Sarcopenia Muscle. 2017;8:113‐121. J Gastroenterol. 2011;106:1081‐1088.
74. Kang SH, Jeong WK, Baik SK, Cha SH, Kim MY. Impact of sarco‐ 94. Kawaguchi T, Shiraishi K, Ito T, et al. Branched‐chain amino acids
penia on prognostic value of cirrhosis: going beyond the hepatic prevent hepatocarcinogenesis and prolong survival of patients
venous pressure gradient and MELD score. J Cachexia Sarcopenia with cirrhosis. Clin Gastroenterol Hepatol. 2014;12:1012‐1018;
Muscle. 2018;9:860‐870. e1011.
75. Bunchorntavakul C, Supanun R, Atsawarungruangkit A. Nutritional 95. Amodio P, Bemeur C, Butterworth R, et al. The nutritional man‐
status and its impact on clinical outcomes for patients admitted to agement of hepatic encephalopathy in patients with cirrhosis:
hospital with cirrhosis. J Med Assoc Thai. 2016;99(Suppl 2):S47‐55. International Society for Hepatic Encephalopathy and Nitrogen
76. Bhanji RA, Carey EJ, Watt KD. Review article: maximising quality Metabolism Consensus. Hepatology. 2013;58:325‐336.
of life while aspiring for quantity of life in end‐stage liver disease. 96. Córdoba J, López‐Hellı́n J, Planas M, et al. Normal protein diet for
Aliment Pharmacol Ther. 2017;46:16‐25. episodic hepatic encephalopathy: results of a randomized study. J
77. Montano‐Loza AJ, Duarte‐Rojo A, Meza‐Junco J, et al. Inclusion Hepatol. 2004;41:38‐43.
of sarcopenia within MELD (MELD‐sarcopenia) and the prediction 97. de Ledinghen V, Beau P, Mannant PR, et al. Early feeding or enteral
of mortality in patients with cirrhosis. Clin Transl Gastroenterol. nutrition in patients with cirrhosis after bleeding from esophageal
2015;6:e102. varices? A randomized controlled study. Dig Dis Sci. 1997;42:536‐541.
78. Merli M, Giusto M, Lucidi C, et al. Muscle depletion increases the 98. Fialla AD, Israelsen M, Hamberg O, Krag A, Gluud LL. Nutritional
risk of overt and minimal hepatic encephalopathy: results of a pro‐ therapy in cirrhosis or alcoholic hepatitis: a systematic review and
spective study. Metab Brain Dis. 2013;28:281‐284. meta‐analysis. Liver Int. 2015;35:2072‐2078.
79. Ohashi K, Ishikawa T, Hoshi A, et al. Relationship between sar‐ 99. Naylor CD, O'Rourke K, Detsky AS, Baker JP. Parenteral nutrition
copenia and both physical activity and lifestyle in patients with with branched‐chain amino acids in hepatic encephalopathy. A
chronic liver disease. J Clin Med Res. 2018;10:920‐927. meta‐analysis. Gastroenterology. 1989;97:1033‐1042.
80. Hayashi F, Matsumoto Y, Momoki C, et al. Physical inactivity and 100. Gluud LL, Dam G, Les I, et al. Branched‐chain amino acids for
insufficient dietary intake are associated with the frequency of people with hepatic encephalopathy. Cochrane Database Syst Rev.
sarcopenia in patients with compensated viral liver cirrhosis. 2017;5:Cd001939.
Hepatol Res. 2013;43:1264‐1275. 101. Everhart JE, Lok AS, Kim H, et al. Weight‐related effects on dis‐
81. Levolger S, van Vledder MG, Muslem R, et al. Sarcopenia impairs ease progression in the hepatitis C antiviral long‐term treatment
survival in patients with potentially curable hepatocellular carci‐ against cirrhosis trial. Gastroenterology. 2009;137:549‐557.
noma. J Surg Oncol. 2015;112:208‐213. 102. Tandon P, Ismond KP, Riess K, et al. Exercise in cirrhosis:
82. Lai JC, Rahimi RS, Verna EC, et al. Frailty associated with waitlist translating evidence and experience to practice. J Hepatol.
mortality independent of ascites and hepatic encephalopathy in a 2018;69:1164‐1177.
multicenter study. Gastroenterology. 2019;156:1675‐1682. 103. Duarte‐Rojo A, Ruiz‐Margain A, Montano‐Loza AJ, Macias‐
83. Merli M, Giusto M, Gentili F, et al. Nutritional status: its influence Rodriguez RU, Ferrando A, Kim WR. Exercise and physical activ‐
on the outcome of patients undergoing liver transplantation. Liver ity for patients with end‐stage liver disease: Improving functional
Int. 2010;30:208‐214. status and sarcopenia while on the transplant waiting list. Liver
84. Hamaguchi Y, Kaido T, Okumura S, et al. Impact of quality as well Transplant. 2018;24:122‐139.
as quantity of skeletal muscle on outcomes after liver transplanta‐ 104. Berzigotti A, Albillos A, Villanueva C, et al. Effects of an inten‐
tion. Liver Transplant. 2014;20:1413‐1419. sive lifestyle intervention program on portal hypertension in pa‐
85. Kamo N, Kaido T, Hamaguchi Y, et al. Impact of sarcopenic obesity tients with cirrhosis and obesity: the SportDiet study. Hepatology.
on outcomes in patients undergoing living donor liver transplanta‐ 2017;65:1293‐1305.
tion. Clin Nutr. 2019;38:2202‐2209. 105. Román E, García‐Galcerán C, Torrades T, et al. Effects of an exer‐
86. Thuluvath PJ, Thuluvath AJ, Savva Y. Karnofsky performance sta‐ cise programme on functional capacity, body composition and risk
tus before and after liver transplantation predicts graft and patient of falls in patients with cirrhosis: a randomized clinical trial. PLoS
survival. J Hepatol. 2018;69:818‐825. ONE. 2016;11:e0151652.
87. Dolgin NH, Martins PN, Movahedi B, Lapane KL, Anderson FA, 106. Zenith L, Meena N, Ramadi A, et al. Eight weeks of exercise train‐
Bozorgzadeh A. Functional status predicts postoperative mortal‐ ing increases aerobic capacity and muscle mass and reduces
ity after liver transplantation. Clin Transplant. 2016;30:1403‐1410. fatigue in patients with cirrhosis. Clin Gastroenterol Hepatol.
88. Tsien CD, McCullough AJ, Dasarathy S. Late evening snack: exploit‐ 2014;12:1920‐1926.e1922.
ing a period of anabolic opportunity in cirrhosis. J Gastroenterol 107. Kruger C, McNeely ML, Bailey RJ, et al. Home exercise training
Hepatol. 2012;27:430‐441. improves exercise capacity in cirrhosis patients: role of exercise
89. Plank LD, Gane EJ, Peng S, et al. Nocturnal nutritional supplemen‐ adherence. Sci Rep. 2018;8(1):99.
tation improves total body protein status of patients with liver cir‐ 108. Matsumoto R, Fukuoka H, Iguchi G, et al. Long‐term effects of
rhosis: a randomized 12‐month trial. Hepatology. 2008;48:557‐566. growth hormone replacement therapy on liver function in adult
90. Chen CJ, Wang LC, Kuo HT, Fang YC, Lee HF. Late‐evening patients with growth hormone deficiency. Growth Hormone IGF
snacking improves liver functions in patients with liver cirrhosis: Res. 2014;24:174‐179.
14 | BUNCHORNTAVAKUL and REDDY

109. Rambaldi A, Gluud C. Anabolic‐androgenic steroids for alcoholic 122. Tsien C, Shah SN, McCullough AJ, Dasarathy S. Reversal of sarco‐
liver disease. Cochrane Database Syst Rev. 2006;4:Cd003045. penia predicts survival after a transjugular intrahepatic portosys‐
110. Yurci A, Yucesoy M, Unluhizarci K, et al. Effects of testosterone gel temic stent. Eur J Gastro Hepatol. 2013;25:85‐93.
treatment in hypogonadal men with liver cirrhosis. Clin Res Hepatol 123. Nardelli S, Lattanzi B, Torrisi S, et al. Sarcopenia is risk factor for
Gastroenterol. 2011;35:845‐854. development of hepatic encephalopathy after transjugular intra‐
111. Dasarathy S, Mullen KD, Dodig M, Donofrio B, McCullough AJ. hepatic portosystemic shunt placement. Clin Gastroenterol Hepatol.
Inhibition of aromatase improves nutritional status following por‐ 2017;15:934‐936.
tacaval anastomosis in male rats. J Hepatol. 2006;45:214‐220. 124. Praktiknjo M, Clees C, Pigliacelli A, et al. Sarcopenia is associated
112. Grech A, Breck J, Heidelbaugh J. Adverse effects of testosterone with development of acute‐on‐chronic liver failure in decompen‐
replacement therapy: an update on the evidence and controversy. sated liver cirrhosis receiving transjugular intrahepatic portosys‐
Ther Adv Drug Saf. 2014;5:190‐200. temic shunt. Clin Transl Gastroenterol. 2019;10(4):e00025.
113. Johnson FLeonard, Lerner KennethG, Siegel M, et al. Association 125. Han HQ, Zhou X, Mitch WE, Goldberg AL. Myostatin/activin path‐
of androgenic‐anabolic steroid therapy with development of hepa‐ way antagonism: molecular basis and therapeutic potential. Int J
tocellular carcinoma. Lancet. 1972;2:1273‐1276. Biochem Cell Biol. 2013;45:2333‐2347.
114. Grungreiff K, Reinhold D, Wedemeyer H. The role of zinc in liver 126. Becker C, Lord SR, Studenski SA, et al. Myostatin antibody
cirrhosis. Ann Hepatol. 2016;15:7‐16. (LY2495655) in older weak fallers: a proof‐of‐concept, randomised,
115. Weismann K, Christensen E, Dreyer V. Zinc supplementation in phase 2 trial. Lancet Diabetes Endocrinol. 2015;3:948‐957.
alcoholic cirrhosis. A double‐blind clinical trial. Acta Med Scand. 127. Woodhouse L, Gandhi R, Warden SJ, et al. A phase 2 randomized
1979;205:361‐366. study investigating the efficacy and safety of myostatin antibody
116. Bjelakovic G, Nikolova D, Bjelakovic M, Gluud C. Vitamin D supple‐ LY2495655 versus placebo in patients undergoing elective total
mentation for chronic liver diseases in adults. Cochrane Database hip arthroplasty. J Frailty Aging. 2016;5:62‐70.
Syst Rev. 2017;11:Cd011564. 128. Dasarathy S, McCullough AJ, Muc S, et al. Sarcopenia associated
117. Yang F, Ren H, Gao Y, Zhu Y, Huang W. The value of severe vitamin with portosystemic shunting is reversed by follistatin. J Hepatol.
D deficiency in predicting the mortality risk of patients with liver cir‐ 2011;54:915‐921.
rhosis: a meta‐analysis. Clin Res Hepatol Gastroenterol. 2019. https​:// 129. Carroll B, Korolchuk VI, Sarkar S. Amino acids and autophagy:
doi.org/10.1016/j.clinre.2019.03.001. [Epub ahead of print.] cross‐talk and co‐operation to control cellular homeostasis. Amino
118. Bischoff‐Ferrari HA, Dawson‐Hughes B, Staehelin HB, et al. Acids. 2015;47:2065‐2088.
Fall prevention with supplemental and active forms of vita‐ 130. Hansen J, Brandt C, Nielsen AR, et al. Exercise induces a marked
min D: a meta‐analysis of randomised controlled trials. BMJ. increase in plasma follistatin: evidence that follistatin is a contrac‐
2009;339:b3692. tion‐induced hepatokine. Endocrinology. 2011;152:164‐171.
119. Wintermeyer E, Ihle C, Ehnert S, et al. Crucial Role of Vitamin D in
the Musculoskeletal System. Nutrients. 2016;8(6):319.
120. Sharma BC, Sharma P, Lunia MK, Srivastava S, Goyal R, Sarin SK.
A randomized, double‐blind, controlled trial comparing rifaximin How to cite this article: Bunchorntavakul C, Reddy KR.
plus lactulose with lactulose alone in treatment of overt hepatic Review article: malnutrition/sarcopenia and frailty in patients
encephalopathy. Am J Gastroenterol. 2013;108:1458‐1463. with cirrhosis. Aliment Pharmacol Ther. 2019;00:1–14. https​://
121. Dasarathy J, Alkhouri N, Dasarathy S. Changes in body composi‐
doi.org/10.1111/apt.15571​
tion after transjugular intrahepatic portosystemic stent in cirrho‐
sis: a critical review of literature. Liver Int. 2011;31:1250‐1258.

You might also like