Hostname: page-component-8448b6f56d-tj2md Total loading time: 0 Render date: 2024-04-19T11:47:41.196Z Has data issue: false hasContentIssue false

A window beneath the skin: how computed tomography assessment of body composition can assist in the identification of hidden wasting conditions in oncology that profoundly impact outcomes

Published online by Cambridge University Press:  10 May 2018

L. E. Daly
Affiliation:
School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland APC Microbiome Institute, University College Cork, Cork, Ireland
C. M. Prado
Affiliation:
Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
A. M. Ryan*
Affiliation:
School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland Cork Cancer Research Centre, University College Cork, Cork, Ireland
*
*Corresponding author: Dr Aoife Ryan, email a.ryan@ucc.ie
Rights & Permissions [Opens in a new window]

Abstract

Advancements in image-based technologies and body composition research over the past decade has led to increased understanding of the importance of muscle abnormalities, such as low muscle mass (sarcopenia), and more recently low muscle attenuation (MA), as important prognostic indicators of unfavourable outcomes in patients with cancer. Muscle abnormalities can be highly prevalent in patients with cancer (ranging between 10 and 90 %), depending on the cohort under investigation and diagnostic criteria used. Importantly, both low muscle mass and low MA have been associated with poorer tolerance to chemotherapy, increased risk of post-operative infectious and non-infectious complications, increased length of hospital stay and poorer survival in patients with cancer. Studies have shown that systemic antineoplastic treatment can exacerbate losses in muscle mass and MA, with reported loss of skeletal muscle between 3 and 5 % per 100 d, which are increased exponentially with progressive disease and proximity to death. At present, no effective medical intervention to improve muscle mass and MA exists. Most research to date has focused on treating muscle depletion as part of the cachexia syndrome using nutritional, exercise and pharmacological interventions; however, these single-agent therapies have not provided promising results. Rehabilitation care to modify body composition, either increasing muscle mass and/or MA should be conducted, and its respective impact on oncology outcomes explored. Although the optimal timing and treatment strategy for preventing or delaying the development of muscle abnormalities are yet to be determined, multimodal interventions initiated early in the disease trajectory appear to hold the most promise.

Type
Conference on ‘What governs what we eat?’
Copyright
Copyright © The Authors 2018 

Over the past decade, there has been a growing interest in the measurement of body composition in patients with cancer. This has largely been in response to advancements in image-based technologies, including gold standard computed tomography (CT) that allow the precise quantification of both muscle and adipose tissue. This research has led to increased understanding of the importance of abnormal body composition phenotypes, such as low muscle mass (sarcopenia), and more recently low muscle attenuation (MA) as important prognostic indicators of unfavourable outcomes in patients with cancer(Reference Martin, Birdsell and Macdonald1Reference Daly, Power and O'Reilly4).

Cancer is a disease associated with ageing. As a result, the aetiology of muscle loss in patients with cancer can be two-fold. Firstly, resulting from the age-related decline in muscle mass (sarcopenia), and secondly due to the metabolic changes induced by malignancy and cancer cachexia. Muscle loss related to sarcopenia begins relatively early in life, as muscle mass begins to decline from the age of 40 at a rate of 6 % per decade(Reference Janssen and Ross5) and accelerates to a rate of 25–40 % per decade above age 70 years(Reference Goodpaster, Park and Harris6). The precise definition of sarcopenia remains controversial and a topic of much debate today; however, a generally accepted criterion in geriatric populations is a level of muscle mass greater than two standard deviations below that of a healthy young reference population(Reference Baumgartner, Koehler and Gallagher7). Cancer cachexia is a multifactorial syndrome that is characterised by the loss of muscle with or without the loss of fat mass leading to progressive functional impairment(Reference Fearon, Strasser and Anker8). It is driven by a variable combination of reduced food intake and abnormal metabolism(Reference Fearon, Strasser and Anker8). Systemic inflammation is commonly described as part of the pathogenesis of cancer cachexia, and may suppress appetite, increase body's metabolic needs and energy expenditure, and accelerate muscle protein catabolism in patients(Reference Fearon, Strasser and Anker8Reference Donohoe, Ryan and Reynolds10). Cancer cachexia represents a spectrum of conditions and can range in severity and clinical presentation from pre-cachexia, identified by early clinical and metabolic signs (e.g. anorexia and impaired glucose tolerance) to refractory cachexia, where extensive muscle and fat depletion is evident, and patients are often immunocompromised(Reference Fearon, Strasser and Anker8). Recognition of these stages of cachexia is important as these stages have different implications in the anabolic therapy response.

Reduced skeletal MA is a relatively newly characterised and distinctive abnormality in patients with cancer(Reference Aubrey, Esfandiari and Baracos11). It represents a ‘qualitative’ measure of skeletal muscle, as low radiation attenuation is reflective of intramuscular adipose tissue infiltration, and therefore, poor ‘quality’ skeletal muscle(Reference Aubrey, Esfandiari and Baracos11). It has been speculated that low MA precedes the development of sarcopenia, as the increase in lipid content occurs before a decline in muscle mass(Reference Chu, Lieffers and Ghosh12, Reference Hayashi, Ando and Gyawali13). Importantly, in recent years, low MA is emerging as an important prognostic indicator in patients with cancer, and in some cases, a better prognostic indicator compared with muscle mass alone(Reference Chu, Lieffers and Ghosh12Reference Sjøblom, Grønberg and Wentzel-Larsen18).

Low muscle mass and low MA can occur at any given BMI (kg/m2)(Reference Martin, Birdsell and Macdonald1, Reference Prado, Lieffers and McCargar19), and now with 40–60 % of cancer patients presenting with overweight and obesity(Reference Ryan, Power and Daly20), identification of these conditions is becoming increasingly difficult. Patients may therefore not appear malnourished as muscle depletion is often hidden behind mantles of adipose tissue and can often go undiagnosed and untreated. Body composition assessment is therefore crucial within this patient group.

This review will focus on the diagnostic criteria, prevalence and clinical consequences associated with CT-defined muscle abnormalities (low muscle mass and low MA) in relation to chemotherapy tolerance, post-operative outcomes and survival in oncology patients. We will examine the changes in body composition that occur in patients undergoing active anti-cancer treatment, and finally, we will briefly examine the evidence behind current treatments aimed at restoring muscle mass and MA, with a particular emphasis on nutritional interventions, physical activity, pharmacological agents and multimodal treatments.

Measurement of skeletal muscle in oncology

The three most commonly used methods to measure body composition in patients with cancer are bioelectrical impedance analysis, dual-energy x-ray absorptiometry and CT. Each technique has its advantages and disadvantages, and methods differ in terms of cost, reliability, validity, availability and training required(Reference Prado and Heymsfield21). CT images are considered a gold standard method for body composition assessment, and allow the precise quantification of tissue area, volume and attenuation. Unlike bioelectrical impedance analysis or dual-energy x-ray absorptiometry, CT can measure body composition at a tissue-organ level, particularly total and regional adipose and skeletal muscle tissues. The use of CT for body composition assessment in non-cancer populations is limited by the high radiation dose, high cost and lack of availability. However, in oncology, CT scans are obtained routinely during diagnostic and surveillance purposes, and therefore represent a unique and exploitable opportunity to assess body composition within this patient group.

Measuring muscle mass by CT is usually done by measuring total skeletal muscle area (SMA) at the third lumber vertebra (L3). Using a commercially available image analysis software (e.g. Sliceomatic (TomoVision), OsiriX (Pixmeo), Image J (National Institutes of Health), among others), muscle and adipose tissues can be evaluated based on Hounsfield unit (HU) thresholds. Muscles in this area include the psoas, paraspinal muscles (erector spinea, quadratus lumborum) and the abdominal wall muscles (transversus abdominus, external and internal oblique, rectus abdominus). Measurements are commonly taken at the third lumber vertebra, as the SMA obtained at this level is a good correlate for whole body muscle in healthy individuals (r 0·92)(Reference Shen, Punyanitya and Wang22). From this skeletal muscle index (SMI; total SMA (cm2)/height (m2)) can be calculated and patients are often compared on this basis. Mean MA is typically derived by averaging HU of the SMA at the third lumber vertebra. Although SMI and MA are continuous variables and could be modelled as such to predict survival/outcome, many clinicians find the interpretation of continuous prognostic covariates difficult, and prefer categorical or binary covariates based on a threshold/cut point to stratify patients into distinct risk groups when making treatment decisions.

Defining low muscle mass (sarcopenia) in patients with cancer

Many oncological studies define sarcopenia based solely on low muscle mass, and studies often lack information on muscle strength or physical function. Therefore, the criteria to define sarcopenia in this group differ from those proposed in geriatric populations(Reference Cruz-Jentoft, Baeyens and Bauer23, Reference Fielding, Vellas and Evans24). In the oncology setting, consensus-based cut points to define low muscle mass or sarcopenia are lacking, and a variety have been devised. Studies(Reference Prado, Lieffers and Bowthorpe25Reference Tamandl, Paireder and Asari28) have defined sarcopenia based on cut points developed by Baumgartner et al. (Reference Baumgartner, Koehler and Gallagher7) in elderly individuals (2-sd below a healthy reference population), by converting the original dual-energy x-ray absorptiometry cut points (<7·26 kg/m2 for men and <5·45 kg/m2 for women) to corresponding CT cut points using published regression equations(Reference Mourtzakis, Prado and Lieffers29). These cut points are SMI <55·4 cm2/m2 for men and <38·9 cm2/m2 for women(Reference Mourtzakis, Prado and Lieffers29), and are those used in the cancer cachexia consensus definition(Reference Fearon, Strasser and Anker8). To date, no healthy population reference values for muscle mass obtained from CT exist.

Studies have employed a statistical technique known as optimal stratification, which is used to identify threshold values associated with elevated risk of poor outcome (e.g. mortality) to define sarcopenia. Using this technique, Prado et al. (Reference Prado, Lieffers and McCargar19) identified cut points for SMI that best predicted survival in a cohort of 250 obese (BMI >30 kg/m2) cancer patients. These cut points are <52·4 cm2/m2 for men and <38·5 cm2/m2 for women(Reference Prado, Lieffers and McCargar19), and have been widely applied in the literature. In 2013, Martin et al. (Reference Martin, Birdsell and Macdonald1) identified both sex and BMI-specific cut points for SMI that best predicted survival (men: <43 cm2/m2 if BMI ≤24·9 kg/m2 and <53 cm2/m2 if BMI ≥25 kg/m2; women: <41 cm2/m2)(Reference Martin, Birdsell and Macdonald1) in a large cohort of 1473 patients with lung and gastrointestinal (GI) cancer, which are more applicable to non-obese cohorts. More recently, in 2017, Caan et al. (Reference Caan, Meyerhardt and Kroenke30) identified sex, BMI and cancer-specific cut points for survival in a very large cohort of early-stage colorectal cancer patients (n 3262; men: <52·3 cm2/m2 if BMI <30 kg/m2 and <54·3 cm2/m2 if BMI ≥30 kg/m2; women: <38·6 cm2/m2 if BMI <30 kg/m2 and <46·6 cm2/m2 if BMI ≥30 kg/m2)(Reference Caan, Meyerhardt and Kroenke30).

In addition to sex and BMI, ethnicity should be considered when applying cut points to a specific cohort. Fujiwara et al. (Reference Fujiwara, Nakagawa and Kudo31) defined CT-derived cut points for a large (n 1257) homogenous cohort of Japanese patients with mixed stage (I–IV) hepatocellular carcinoma that best predicted survival (<36·2 cm2/m2 for men and <29·0 cm2/m2 for women), providing a reference population for these individuals. Notably, these cut points are lower(Reference Fujiwara, Nakagawa and Kudo31, Reference Iritani, Imai and Takai32) compared with those derived from large Caucasian populations(Reference Martin, Birdsell and Macdonald1, Reference van Vugt, Braam and van Oudheusden3, Reference Coelen, Wiggers and Nio34).

Using optimal stratification methodology, several other studies have reported cut points for SMI associated with mortality in a number of cancer cohorts (see Table 1), these range 36–55·8 cm2/m2 for men and 29–46·6 cm2/m2 for women. Several factors influence patient's muscularity (ethnicity, age, sex, physical activity and magnitude of adiposity)(Reference Heymsfield, Gonzalez and Lu35); hence, published cut points may not be applicable to all cancer populations. For example, cut points published by Prado et al. (Reference Prado, Lieffers and McCargar19) were devised in a cohort of obese patients with cancer, and may have limited relevance when applied to populations with varying prevalence of obesity, but have subsequently been used in studies comprised of predominantly non-obese individuals(Reference Dalal, Hui and Bidaut36Reference Elliott, Doyle and Murphy40).

Table 1. Cut points for skeletal muscle index (SMI) at the third lumber vertebra (L3) associated with mortality in patients with cancer

GI, gastrointestinal.

* Obese individuals only (BMI >30 kg/m2).

Other studies have defined sarcopenia based on more data-orientated approaches, dichotomizing SMI based on predetermined percentiles, such as quartiles(Reference Miyamoto, Baba and Sakamoto41), tertiles(Reference van Dijk, Bakens and Coolsen17, Reference Harada, Ida and Baba42) or based on the median(Reference Rutten, van Dijk and Kruitwagen43, Reference Boer, de Graaff and Brusse-Keizer44). Studies focused solely on one muscle group, the psoas muscle, generating a psoas muscle index, often considered patients within the lowest quartile of psoas muscle index to be sarcopenic(Reference Amini, Spolverato and Gupta45Reference Peng, Hyder and Firoozmand47). However, no studies have related psoas muscle area to whole body measures, and it has only been weakly correlated with total lumbar muscle area(Reference Rutten, Ubachs and Kruitwagen48). Therefore, it has been argued that the use of the psoas muscle as a sentinel muscle for the diagnosis of sarcopenia is flawed(Reference Baracos49). Other investigations have included additional measures of physical function (hand grip strength and/or gait speed) in conjunction with skeletal muscle from CT scan analysis when defining sarcopenia in patients with cancer(Reference Huang, Zhou and Wang50, Reference Fukuda, Yamamoto and Hirao51). The rationale for including the measures of both muscle mass and function is that muscle strength does not depend solely on muscle mass, and the relationship between muscle strength and mass is not linear(Reference Goodpaster, Park and Harris6). Hence, several expert groups have proposed the use of both muscle mass and function to define sarcopenia in older adults(Reference Cruz-Jentoft, Baeyens and Bauer23, Reference Fielding, Vellas and Evans24, Reference Chen, Liu and Woo52).

Prevalence of low muscle mass (sarcopenia) in patients with cancer

As discussed previously, comparison among studies reporting the prevalence of sarcopenia is often difficult because of inconsistent methodology used to evaluate body composition (dual-energy x-ray absorptiometry, bioelectrical impedance analysis, CT), and diagnostic criteria/cut points used to define sarcopenia. Table 2 summarises the prevalence of sarcopenia among patients with cancer using the same methodology (CT defined SMI) but using varying published cut points. The prevalence of sarcopenia is highly variable across primary cancer sites, and has been shown to range from 11 to 90 %(Reference Fujiwara, Nakagawa and Kudo31, Reference Auclin, Bourillon and De Maio53). Of note is the heterogeneity in the prevalence of sarcopenia among cohorts of the same primary cancer site and stage, e.g. 19–71 % in advanced colorectal cancer(Reference van Vledder, Levolger and Ayez54, Reference Barret, Antoun and Dalban55), 33–90 % in metastatic kidney cancer(Reference Auclin, Bourillon and De Maio53Reference Cushen, Power and Teo56) and 21–89 % in advanced pancreatic cancer(Reference Choi, Oh and Kim57, Reference Wesseltoft-Rao, Hjermstad and Ikdahl58). This large variability could be attributed to the varying cut points used to define sarcopenia; limitations related to sample size; or patient characteristics such as age, sex, ethnicity, BMI and concurrent co-morbidities, thus limiting the ability to draw conclusions as to the true prevalence according to the cancer site. Sarcopenia appears to be the most prevalent in patients with any stage of pancreatic or lung cancer, while in other cancer types (e.g. gastric and breast), it appears to be more frequent in advanced stage disease compared with earlier loco regional disease. Sarcopenia can be present at any given BMI, and the prevalence of sarcopenic obesity (sarcopenia and obesity (BMI ≥30 kg/m2)) has been shown to vary between 1 and 29 % in studies including individuals from all BMI categories, and between 15 and 36 % in studies including obese individuals only(Reference Carneiro, Mazurak and Prado59).

Table 2. Prevalence of sarcopenia according to cancer site, country and definition used

NSC, non-small cell; SC, small cell; GI, gastrointestinal; NR, not recorded; GIST, gastrointestinal stromal tumour.

* Prevalence of sarcopenia in both sexes combined.

Sarcopenia definitions stratified as follows:

1: Skeletal muscle index (SMI) <52·4 cm2/m2 for males; <38·5 cm2/m2 for females(Reference Prado, Lieffers and McCargar19).

2: SMI <55·4 cm2/m2 for males; <38·9 cm2/m2 for females (Baumgartner sarcopenia cut points for elderly healthy subjects(Reference Baumgartner, Koehler and Gallagher7) converted to CT cut points using regression equations(Reference Mourtzakis, Prado and Lieffers29).

3: SMI <43 cm2/m2 BMI ≤24·9 kg/m2 and <53 cm2/m2 for BMI >25 kg/m2 for males; <41 cm2/m2 for females(Reference Martin, Birdsell and Macdonald1).

4: SMI <43·75 cm2/m2 for males; <41·1 cm2/m2 for females(Reference van Vledder, Levolger and Ayez54).

5: SMI <55·8 cm2/m2 for males; <38·9 cm2/m2 for females(Reference Lanic, Kraut-Tauzia and Modzelewski33).

6: SMI <49·5 cm2/m2 and for women <42·1 cm2/m2 (lowest quartile)(Reference Miyamoto, Baba and Sakamoto41).

7: SMI <42·2 cm2/m2 for males; <33·9 cm2/m2 for females(Reference Choi, Oh and Kim57).

8: SMI <36·2 cm2/m2 for males; <29·0 cm2/m2 for females(Reference Fujiwara, Nakagawa and Kudo31).

9: SMI ≤36·0 cm2/m2 for males; ≤29·0 cm2/m2 for females(Reference Iritani, Imai and Takai32).

10: SMI <41·5 cm2/m2 (median) for females(Reference Rutten, van Dijk and Kruitwagen43).

11: SMI <46·8 cm2/m2 for males; <39·1 cm2/m2 for females(Reference Coelen, Wiggers and Nio34).

12: SMI <52·3 cm2/m2 for men and <38·6 cm2/m2 for men with a BMI <30 kg/m2 and <54·3 cm2/m2 for men and <46·6 cm2/m2 for women with a BMI ≥30 kg/m2(Reference Caan, Meyerhardt and Kroenke30).

Low muscle attenuation in patients with cancer

Similar to the measures of SMI, no widely agreed upon cut points are available for defining low MA. Martin et al. (Reference Martin, Birdsell and Macdonald1) provided the first set of cut points for low MA (using optimal stratification) that related to poor survival in a large cohort of lung and GI cancer patients (n 1473; <41 HU for BMI <25 kg/m2 and <33 for BMI ≥25 kg/m2)(Reference Martin, Birdsell and Macdonald1). Since then, cancer-specific cut points for low MA that relate optimally to survival have been reported for lung(Reference Sjøblom, Grønberg and Wentzel-Larsen18, Reference Bowden, Williams and Simms60), ovarian(Reference Kumar, Moynagh and Multinu61, Reference Aust, Knogler and Pils62), periampullary(Reference Van Rijssen, van Huijgevoort and Coelen16), pancreatic(Reference Okumura, Kaido and Hamaguchi63), gastro-oesophageal(Reference Tamandl, Paireder and Asari28) and large B-cell lymphoma(Reference Chu, Lieffers and Ghosh15). These range from <28·0 to 44·1 HU in men(Reference Van Rijssen, van Huijgevoort and Coelen16, Reference Sjøblom, Grønberg and Wentzel-Larsen18, Reference Bowden, Williams and Simms60) and <23·8 to 40·5 HU in women(Reference Van Rijssen, van Huijgevoort and Coelen16, Reference Sjøblom, Grønberg and Wentzel-Larsen18, Reference Bowden, Williams and Simms60Reference Aust, Knogler and Pils62). Other studies have defined low MA based on the sample sex-specific median(Reference Boer, de Graaff and Brusse-Keizer44, Reference Antoun, Lanoy and Iacovelli64), tertile(Reference van Dijk, Bakens and Coolsen17) or quartile(Reference Akahori, Sho and Kinoshita65).

The prevalence of low MA among patients with cancer varies greatly, and is dependent on the cohort under investigation and cut point used, but has been shown to range from 10 to 86 %(Reference Sjøblom, Grønberg and Wentzel-Larsen18, Reference Cushen, Power and Murphy66). Using the cut points devised by Martin et al. (Reference Martin, Birdsell and Macdonald1), which have been applied most widely in the literature, the prevalence of low MA has been reported to be between 46 and 53 % in two large cohorts of patients with cancer (mixed tumour sites and stages (I–IV))(Reference Martin, Birdsell and Macdonald1, Reference Ní Bhuachalla, Daly and Power67). In the setting of advanced disease, the prevalence of low MA has been reported to be 33 % in melanoma(Reference Daly, Power and O'Reilly4), 55 % in pancreatic(Reference Rollins, Tewari and Ackner68), 59 % in gastric(Reference Hayashi, Ando and Gyawali13), 60 % in breast(Reference Rier, Jager and Sleijfer14) and 86 % in prostate cancer(Reference Cushen, Power and Murphy66), while in patients with operable colorectal cancer, low MA is present in 58–78 % of patients(Reference McSorley, Black and Horgan69, Reference Malietzis, Currie and Athanasiou70).

Impact of muscle abnormalities on clinical outcomes

Notwithstanding the controversies in determining and defining low muscle mass and low MA in oncology, it has been well established and reported over the past decade that these muscle abnormalities are unequivocally associated with negative clinical outcomes in patients with cancer.

Muscle abnormalities and tolerance to chemotherapy

Chemotherapy can often be associated with severe toxicity (grades III–IV) that can result in dose delays, dose reductions and treatment termination, referred to as dose-limiting toxicities (DLT). DLT may lead to hospitalisations and can be life threatening. Exploratory studies provided the initial evidence of an association between low muscularity and increased incidence of severe toxicity/DLT to chemotherapy, and subsequent work has confirmed these observations in multiple cancer sites and treatments (Fig. 1). In advanced disease, low muscularity has been associated with poorer tolerance to chemotherapy in patients with breast(Reference Prado, Baracos and McCargar71, Reference Shachar, Deal and Weinberg72), renal(Reference Cushen, Power and Teo56, Reference Antoun, Baracos and Birdsell73, Reference Huillard, Mir and Peyromaure74), liver(Reference Mir, Coriat and Blanchet75), lung(Reference Sjøblom, Grønberg and Benth76), colorectal(Reference Barret, Antoun and Dalban55, Reference Chemama, Bayar and Lanoy77), thyroid(Reference Massicotte, Borget and Broutin78) and melanoma skin cancer(Reference Daly, Power and O'Reilly4, Reference Valentine, François and Nora79). Sarcopenia, in patients with peritoneal metastasis from colorectal cancer, was associated with significantly more chemotherapy toxicities (57 v. 26 %, P = 0·004) and particularly neutropenia (36 v. 17 %, P = 0·04) in a cohort of 97 patients receiving hyperthermic intraperitoneal chemotherapy.

Fig. 1. Sarcopenic patients experience more dose-limiting toxicities (DLT) to flurouracil (5-FU) in colon cancer(Reference Prado, Baracos and McCargar80); capecitabine in breast cancer(Reference Prado, Baracos and McCargar71); sorafenib in renal cell carcinoma(Reference Antoun, Baracos and Birdsell73); 5-flurouracil, epirubicin, cyclophosphamide (FEC) in breast cancer(Reference Prado, Lima and Baracos90); sorafenib in hepatocellular carcinoma(Reference Mir, Coriat and Blanchet75); sunitunib in renal cell carcinoma(Reference Huillard, Mir and Peyromaure74); vantetanib in medullary thyroid cancer(Reference Massicotte, Borget and Broutin78); fluropyramidine in colorectal cancer(Reference Barret, Antoun and Dalban55); phase I drugs in mixed cancer types(Reference Cousin, Hollebecque and Koscielny177); imatinib in gastrointestinal stromal tumours (grade I–II toxicity)(Reference Moryoussef, Dhooge and Volet176); epirubicin, cisplatin, capecitabine (ECX) and cisplatin, 5-flurouracil (CF) in oesophagogastric cancer(Reference Tan, Brammer and Randhawa84); taxane-based chemotherapy (placitaxel, docetaxel, nab-paclitaxel) in breast cancer(Reference Shachar, Deal and Weinberg72); hyperthermic intraperitoneal chemotherapy (oxaliplatin and irrinotecan) in colorectal cancer(Reference Chemama, Bayar and Lanoy77), neoadjuvant chemotherapy (NACT) (mixed types) in gastric cancer(Reference Palmela, Velho and Agostinho81); ipilimumab in metastatic melanoma(Reference Daly, Power and O'Reilly4).

Even in early-stage disease (stages I–III), sarcopenia is associated with poorer tolerance to chemotherapy(Reference Cespedes Feliciano, Lee and Prado2, Reference Prado, Baracos and McCargar80Reference Tan, Brammer and Randhawa84). Interestingly, in patients with early-stage breast cancer (n 151) and receiving anthracycline- and taxane-based chemotherapy, with every five-unit decrease in SMI, the risk of any grade III–IV toxicity increased by 27 % (relative risk 1·27 (95 % CI 1·09, 1·49), P = 0·002)(Reference Shachar, Deal and Weinberg82). In a large study of patients with non-metastatic colon cancer (n 533) treated with adjuvant FOLOX treatment, patients with low muscle mass (lowest sex-specific tertile) were twice as likely to experience dose reductions (OR 2·28, P = 0·01), dose delays (OR 2·24, P = 0·002) and early discontinuation of treatment (OR 2·34, P = 0·03)(Reference Cespedes Feliciano, Lee and Prado2). Although the majority of studies have consistently demonstrated an association between low muscle mass and increased incidence of severe toxicity/DLT (twenty-six of thirty-two studies; see Supplementary material for summary of each study), fewer small studies found contradictory findings(Reference Yip, Goh and Davies85Reference Blauwhoff-Buskermolen, Versteeg and de van der Schueren88).

Emerging data suggest low MA to be also associated with poorer tolerance to antineoplastic agents(Reference Daly, Power and O'Reilly4, Reference Cushen, Power and Murphy66, Reference Shachar, Deal and Weinberg82). In metastatic melanoma patients (n 84), we have previously shown that patients with low MA more frequently experienced high-grade toxicities (75 v. 31 %, P = 0·001) and immune-related toxicities to ipilimumab (54 v. 23 %, P = 0·017) compared with those without low MA. More importantly, these patients were more susceptible to experience a DLT (37·5 v. 10·4 %, P = 0·011)(Reference Daly, Power and O'Reilly4). In patients with metastatic prostate cancer treated with docetaxel (n 63), a combination of both low muscle mass and low MA was associated with an increased risk of DLT (59 v. 29 %, P = 0·021)(Reference Cushen, Power and Murphy66). Similarly, in early-stage disease, Shachar et al. (Reference Shachar, Deal and Weinberg82) demonstrated that in breast cancer (n 151), the risk of hospitalisations due to chemotherapy toxicity increased 19 % with every five-unit decrease in MA (relative risk 1·19 (95 % CI 1·00, 1·43), P = 0·05).

Increased toxicity in patients with sarcopenia may be attributed to alterations in the distribution, metabolism and clearance of systemic chemotherapy drugs(Reference Prado, Baracos and McCargar80). The practice of administration of cytotoxic chemotherapy based on body surface area, and targeted therapy as a flat dose, ignores several sources of inter-individual variation. Using body surface area as the only method to individualise chemotherapy drug dose is insufficient to avoid severe toxicity, but the continued use mainly relies on the lack of other more precise methods for dose individualisation(Reference Felici, Verweij and Sparreboom89). It is recently acknowledged that variability in body composition (lean mass, fat mass and total body water) of cancer patients may be a source of disparities in the metabolism of cytotoxic agents resulting in increased toxicity(Reference Antoun, Baracos and Birdsell73, Reference Prado, Baracos and McCargar80, Reference Prado, Lima and Baracos90). The rationale is that body weight comprises two major compartments, lean mass and fat mass, which may be the two major sites of distribution of hydrophilic and lipophilic drugs, respectively(Reference Prado, Baracos and McCargar80, Reference Prado, Baracos and Xiao91). Therefore, changes in body composition may lead to changes in the volume of distribution and adversely impact the effectiveness and tolerance of cancer therapies.

Administration of hydrophilic chemotherapy drugs that are mainly distributed to the lean mass compartment in sarcopenic patients would result in a disproportionally small volume of the drug distribution in relation to their body weight or body surface area(Reference Prado, Baracos and McCargar71, Reference Prado, Baracos and McCargar80). This has been hypothesised to lead to considerable variation in milligram of chemotherapeutic agent per kg lean mass, and a higher dose per kg lean mass is associated with more frequent severe toxic side effects(Reference Sjøblom, Grønberg and Benth76, Reference Prado, Baracos and McCargar80, Reference Ali, Baracos and Sawyer92, Reference Sjøblom, Benth and Grønberg93). In patients with advanced non-small-cell lung cancer (n 424), when the dose of non-platinum drugs was expressed as mg/kg lean mass, a 3-fold range was observed, and the dose of non-platinum chemotherapeutic agent per kg lean mass was a significant predictor of haematologic toxicity. Every 1 % increase in drug dose per kg lean mass above the mean was associated with a 3 % increased risk of grade III–IV haematologic toxicity, while patients with doses >20 % above the mean were at almost double the risk of experiencing a grade III–IV haematologic toxicity(Reference Sjøblom, Benth and Grønberg93). Pharmacokinetic data have supported this hypothesis, with sarcopenic patients experiencing higher plasma concentrations of antineoplastic drugs, and experiencing more toxicity(Reference Mir, Coriat and Blanchet75, Reference Massicotte, Borget and Broutin78). The same hypothesis holds true for toxicity to lipophilic drugs. In a study of ovarian cancer patients receiving doxorubicin and trabectedin, the risk of DLT decreased with increased fat mass:lean mass ratio. As lipophilic drugs are mainly distributed in the adipose tissue, leaner individuals would present with reduced volume of distribution for the drug and consequently increase their risk for DLT(Reference Prado, Baracos and Xiao91).

In addition to the pharmacokinetic hypothesis to explain the increased toxicity in sarcopenic patients, other mechanisms have been suggested. Low levels of lean mass and altered concentration of plasma proteins (e.g. albumin) may affect the distribution of highly protein-bound drugs, and may explain the increased toxicity of the vandetanib(Reference Massicotte, Borget and Broutin78), sorafenib(Reference Mir, Coriat and Blanchet75) and epirubicin(Reference Prado, Lima and Baracos90). Systemic inflammation is known to inhibit hepatic enzymes and may contribute to higher drug exposure, and subsequently excess toxicity in patients with sarcopenia(Reference Mir, Coriat and Blanchet75, Reference Massicotte, Borget and Broutin78). In addition, sarcopenic patients are generally more susceptible to acute medical events and perhaps chemotherapy toxicity may be an additional generalised intolerance(Reference Antoun, Borget and Lanoy94). Whether altered chemotherapy dosing based on lean mass in patients receiving treatment is effective in preventing toxicity is currently being investigated (ClinicalTrials.gov identifier: NCT01624051).

Sarcopenia and survival

The impact of sarcopenia on survival in oncology was first identified in a cohort of 250 obese lung and GI cancer patients by Prado et al. in 2008(Reference Prado, Lieffers and McCargar19). Within this study, sarcopenic obese patients had a lower median overall survival compared with their non-sarcopenic counterparts (11·3 v. 21·6 months, P < 0·001), and sarcopenic obesity independently predicted survival when adjusted for known prognostic covariates (hazard ratio (HR) 4·2 (95 % CI 2·4, 7·2), P < 0·0001)(Reference Prado, Lieffers and McCargar19). Since then, sarcopenia has repeatedly been shown to be independently prognostic of reduced survival in a number of cancer sites and stages including pancreatic(Reference Tan, Birdsell and Martin95), kidney(Reference Sharma, Zargar-Shoshtari and Caracciolo96, Reference Fukushima, Nakanishi and Kataoka97), liver(Reference Meza-Junco, Montano-Loza and Baracos98Reference Levolger, van Vledder and Muslem100), lung(Reference Kim, Kim and Park101), oesophageal(Reference Tamandl, Paireder and Asari28), colorectal(Reference Miyamoto, Baba and Sakamoto41, Reference van Vledder, Levolger and Ayez54, Reference Malietzis, Currie and Athanasiou70), urothelial(Reference Psutka, Carrasco and Schmit102, Reference Fukushima, Yokoyama and Nakanishi103) and lung and GI cancers(Reference Martin, Birdsell and Macdonald1). Its important prognostic ability was reported in a study of 196 patients following resection for colorectal cancer liver metastases, whereby sarcopenic patients had a median survival of 24 months, which was in stark contrast to 60 months in those without sarcopenia (P < 0·001)(Reference van Vledder, Levolger and Ayez54). In patients with hepatocellular carcinoma (n 116), similar results were obtained with a median survival of 16 months in patients with sarcopenia compared with 28 months in those without sarcopenia (P = 0·003)(Reference Meza-Junco, Montano-Loza and Baracos98). More recently, a large cohort of stage I–III colorectal cancer patients (n 3262) showed that sarcopenic patients had a 27 % higher risk of overall mortality than their non-sarcopenic counterparts(Reference Caan, Meyerhardt and Kroenke30).

The impact of sarcopenia on survival has been extensively reviewed previously(Reference Shachar, Williams and Muss104Reference Gibson, Burden and Strauss106) and of note is a recent systematic review and meta-analysis(Reference Shachar, Williams and Muss104), which examined the prognostic value of sarcopenia in 7843 patients with solid tumours from a total of thirty-eight studies. Sarcopenia was found to be associated with poor overall survival (HR 1·44 (95 % CI 1·32, 1·56), P < 0·001), the effect of which remained in various tumour types and across disease stages. Additionally, sarcopenia was associated with cancer-specific survival (HR 1·93 (95 % CI 1·38, 2·70), P < 0·001), as well as disease-free survival (HR 1·16 (95 % CI 1·00, 1·30), P = 0·014). However, it is noteworthy to mention the limitations of this study. The authors included several studies in the review that contained duplicated data, and this is prohibited in a meta-analysis as it may lead to overestimation of an effect(Reference Tramèr, Reynolds and Moore107). In addition, the studies were largely heterogeneous in terms of the tumours considered, stage of disease, treatments and cut points used to define sarcopenia, which ranged from 29 to 42·1 cm2/m2 in women and 36 to 55·4 cm2/m2 in men(Reference Shachar, Williams and Muss104).

In spite of the abundance of evidence supporting the role of sarcopenia as an important and independent prognostic factor in cancer, this finding has not always been consistently reported and several studies have reported no impact on survival(Reference Van Rijssen, van Huijgevoort and Coelen16Reference Sjøblom, Grønberg and Wentzel-Larsen18, Reference Stene, Helbostad and Amundsen37, Reference Rollins, Tewari and Ackner68, Reference Blauwhoff-Buskermolen, Versteeg and de van der Schueren88). It is possible that the impact of sarcopenia may vary depending on cancer diagnosis, treatment and overall prognosis. For example, in colorectal cancer patients, sarcopenia was shown to be predictive of survival in patients undergoing curative resection(Reference Miyamoto, Baba and Sakamoto41), but not in patients with unresectable disease receiving chemotherapy(Reference Miyamoto, Baba and Sakamoto108). Alternatively, this may be a consequence of the choice of cut points to define sarcopenia in many of these studies. As discussed earlier, cut points obtained in different studies are highly dependent on the methodology and the population from which they were devised, and when used in dissimilar populations, they may represent a suboptimal approach in identifying the true prevalence of low muscle mass and its relationship to survival within these cohorts.

Muscle quality v. quantity

Low MA, often referred to as myosteatosis, is emerging as an important prognostic indicator in patients with cancer and in some cases more superior to predicting survival compared with muscle mass alone(Reference Chu, Lieffers and Ghosh12Reference Sjøblom, Grønberg and Wentzel-Larsen18). The first report by Sabel et al. (Reference Sabel, Lee and Cai109) in melanoma patients showed poorer disease-free survival (P = 0·04) and distant disease-free survival (P = 0·0002) in patients with the lowest tertile of psoas MA. Antoun et al. (Reference Antoun, Lanoy and Iacovelli64) corroborated these findings in a cohort of metastatic renal cell carcinoma patients, and reported that the median survival of patients with a MA below the median (<38 HU in men; <36 HU in women) was half that of those with MA above the median (29 v. 14 months, P = 0·001). In a large cohort of 1473 patients with lung and GI cancer, Martin et al. (Reference Martin, Birdsell and Macdonald1) reported that patients with a MA below the identified cut point (<41 HU for BMI <25 kg/m2 and <33 HU for BMI ≥25 kg/m2) were at a 36 % increased risk of mortality (HR 1·36 (95 % CI 1·19, 1·55), P < 0·001). Recently, low MA has been shown to be prognostic of reduced survival in a variety of different tumours, including lung(Reference Sjøblom, Grønberg and Wentzel-Larsen18, Reference Bowden, Williams and Simms60), kidney(Reference Antoun, Lanoy and Iacovelli64), breast(Reference Rier, Jager and Sleijfer14), gastric(Reference Hayashi, Ando and Gyawali13), pancreatic(Reference van Dijk, Bakens and Coolsen17, Reference Akahori, Sho and Kinoshita65), periampullary(Reference Van Rijssen, van Huijgevoort and Coelen16), cholangiocarcinoma(Reference Rollins, Tewari and Ackner68), lymphoma(Reference Chu, Lieffers and Ghosh12, Reference Chu, Lieffers and Ghosh15) and melanoma(Reference Sabel, Lee and Cai109). In a cohort of 734 patients with advanced lung cancer, Sjøblom et al. (Reference Sjøblom, Grønberg and Wentzel-Larsen18) reported that even minor changes in MA are associated with mortality risk. An incremental increase of 1 HU was associated with a 2 % reduction in the risk of death (HR 0·98 (95 % CI 0·97, 0·99), P < 0·001). Accordingly, the mortality risk reduction of a 5 HU and 10 HU increase in MA were 8 % and 16 %, respectively(Reference Sjøblom, Grønberg and Wentzel-Larsen18).

Sarcopenia, low muscle attenuation and post-operative outcomes

Sarcopenia has also been identified as a predictor of post-operative infections, complications, readmissions, longer length of hospital stay and higher health care costs(Reference van Vugt, Braam and van Oudheusden3, Reference Levolger, van Vledder and Muslem100, Reference Lieffers, Bathe and Fassbender110Reference Sharma, Zargar-Shoshtari and Caracciolo114). Elliott et al. (Reference Elliott, Doyle and Murphy40) demonstrated that sarcopenia, in a cohort of 252 patients with oesophageal cancer undergoing resection, was independently associated with increased risk of major postoperative complications (grade ≥IIIb; OR 5·30 (95 % CI 1·94, 14·45), P = 0·001) and pulmonary complications (OR 2·17 (95 % CI 1·12, 4·23), P = 0·023). Similarly, in patients with colorectal cancer undergoing cytoreductive surgery for peritoneal carcinomatosis (n 206), sarcopenia was associated with severe post-operative complications, and sarcopenic patients underwent significantly more reoperations (26 v. 12 %, P = 0·012)(Reference van Vugt, Braam and van Oudheusden3). Consequently, length of hospital stay has shown to be significantly longer in patients with sarcopenia(Reference Elliott, Doyle and Murphy40, Reference Joglekar, Asghar and Mott46, Reference Sharma, Zargar-Shoshtari and Caracciolo96, Reference Lieffers, Bathe and Fassbender110, Reference Peng, van Vledder and Tsai111, Reference van Vugt, Buettner and Levolger115). Following colorectal cancer surgery (n 234), the length of hospital stay for sarcopenic patients was 15·9 d compared with 12·3 d in patients without sarcopenia (P = 0·038)(Reference Lieffers, Bathe and Fassbender110). Ultimately, adverse post-operative outcomes in patients with sarcopenia lead to increased health care costs. In a Western-Europe healthcare system, sarcopenia was independently associated with increased hospital costs of €4061 per patient (P = 0·015) in a study of 452 patients who underwent cancer surgery of the alimentary tract(Reference van Vugt, Buettner and Levolger115).

Akin to sarcopenia, low MA is emerging as a predictor of poor post-operative outcomes(Reference Van Rijssen, van Huijgevoort and Coelen16, Reference Boer, de Graaff and Brusse-Keizer44, Reference Sabel, Lee and Cai109, Reference Margadant, Bruns and Sloothaak116). In patients with colon cancer undergoing open resection (n 91), low MA was an independent risk factor for one or more complications (P < 0·001)(Reference Boer, de Graaff and Brusse-Keizer44). Van Rijssen et al. (Reference Van Rijssen, van Huijgevoort and Coelen16) reported similar findings, whereby patients with low MA experienced significantly more major post-operative complications (58 v. 37 %, P = 0·005) following resection for periampullary cancer (n 166) compared with those without low MA. Examining MA as a continuous variable, Sabel et al. (Reference Sabel, Lee and Cai109) reported that a 10 HU decrease in psoas MA was associated with an 8·1 % increase in complication rate in patients with stage III melanoma (n 101). In line with this, low MA was associated with prolonged length of hospital stay following colorectal cancer surgery in a large cohort of 805 patients (7 v. 6 d (P = 0·034))(Reference Malietzis, Currie and Athanasiou70), and to a greater extent following pancreatic cancer resection, whereby patients with low MA (post-neoadjuvant chemoradiotherapy) had a mean length of stay of 42 d compared with 23 d in those without low MA (P = 0·001)(Reference Akahori, Sho and Kinoshita65).

Muscle loss during chemotherapy

The precision associated with CT analysis of body composition (1–1·5 %) has allowed recent investigations to focus on the nature and magnitude of changes in body composition during the disease trajectory in patients with cancer. Studies have consistently shown that losses in muscle mass and MA are exacerbated by antineoplastic treatment(Reference Palmela, Velho and Agostinho81, Reference Blauwhoff-Buskermolen, Versteeg and de van der Schueren88, Reference Prado, Sawyer and Ghosh117Reference Reisinger, Bosmans and Uittenbogaart119). In oesophagogastric cancer patients, neoadjuvant chemotherapy treatment which is typically delivered over a 6–8-week period can decrease SMA by 9·59 cm2 (P < 0·0001), which is the equivalent to a loss of 2·9 kg of lean mass(Reference Awad, Tan and Cui118). These results have been corroborated in a study of 252 oesophageal cancer patients, whereby the prevalence of sarcopenia almost doubled (15·9–30·8 %) during a course of neoadjuvant chemotherapy(Reference Elliott, Doyle and Murphy40).

In patients with cancer, muscle is lost at a very high rate of 3–5 % per 100 d during systemic chemotherapy(Reference Daly, Power and O'Reilly4, Reference Dalal, Hui and Bidaut36, Reference Rutten, van Dijk and Kruitwagen43, Reference Tan, Birdsell and Martin95), and losses are exponentially increased with progressive disease and proximity to death(Reference Prado, Sawyer and Ghosh117, Reference Lieffers, Mourtzakis and Hall120). In a recent study of metastatic colorectal cancer patients (n 63), on average patients lost muscle at a rate of 6·1 % during 3 months of chemotherapy(Reference Blauwhoff-Buskermolen, Versteeg and de van der Schueren88); it is noteworthy that the rate of decline is 24-fold more rapid than that observed in healthy ageing adults who tend to lose muscle at a rate of 1–1·4 % per year(Reference Goodpaster, Park and Harris6, Reference Frontera, Hughes and Fielding121).

Alterations in muscle mass may be a consequence of some cancer-directed therapies. Sorafenib, a multi-kinase inhibitor, has been shown to provoke muscle wasting in metastatic renal cell carcinoma patients, through the downstream suppression of PI3K, AKTm and mTOR, key mediators in activating muscle protein synthesis by amino acids and other stimuli(Reference Antoun, Birdsell and Sawyer38). Abiraterone(Reference Pezaro, Mukherji and Tunariu122) and MK-0646 (anti-insulin-like growth factor 1 receptor)(Reference Fogelman, Holmes and Mohammed123) have also been reported to stimulate muscle loss through maximal androgen suppression and inhibition of anti-insulin-like growth factor 1 receptor, respectively. In contrast, two chemotherapy agents (vandetanib and selumetinib) have resulted in significant muscle gain in patients with advanced cancer(Reference Massicotte, Borget and Broutin78, Reference Prado, Bekaii-Saab and Doyle124).

Loss of muscle during systemic anti-cancer treatment is associated with increased mortality in patients with pancreatic(Reference Dalal, Hui and Bidaut36, Reference Cooper, Slack and Fogelman125), lung(Reference Stene, Helbostad and Amundsen37, Reference Nattenmüller, Wochner and Muley126), colorectal(Reference Blauwhoff-Buskermolen, Versteeg and de van der Schueren88, Reference Miyamoto, Baba and Sakamoto108), ovarian(Reference Rutten, van Dijk and Kruitwagen43), melanoma(Reference Daly, Power and O'Reilly4) and foregut cancer(Reference Daly, Ní Bhuachalla and Power127) (see Table 3 for a summary of available studies reporting that muscle loss during treatment is prognostic of reduced survival). Loss of muscle >2 %/100 d was independently associated with reduced survival in ovarian cancer patients receiving neoadjuvant chemotherapy (n 123; HR 1·77 (95 % CI 1·02, 3·09), P = 0·043)(Reference Rutten, van Dijk and Kruitwagen43), and we have previously reported that in patients with advanced cancers of the foregut, those with a skeletal muscle loss >6 %/100 d are at more than double the risk of mortality (HR 2·66 (95 % CI 1·42, 4·97), P = 0·002)(Reference Daly, Ní Bhuachalla and Power127). Importantly, skeletal muscle depletion during chemotherapy has also been associated with reductions in physical function in elderly patients with advanced non-small-cell lung cancer (Reference Naito, Okayama and Aoyama128).

Table 3. Summary of studies reporting muscle loss during treatment as prognostic of reduced survival

IQR, interquartile range; NSC, non-small cell; SM, skeletal muscle; SMA, skeletal muscle area; HR, hazard ratio; SMI, skeletal muscle index; se, standard error; OS, overall survival.

Strategies to improve muscle mass and attenuation

Currently, there is no consensus treatment for attenuating or reversing the muscle wasting caused by sarcopenia and/or cachexia in patients with cancer. Research to date has focused on strategies to treat sarcopenia in the context of cancer cachexia using nutritional, exercise and pharmacological interventions. However, these single-agent therapies have not provided promising results, and perhaps multifaceted treatment strategies may be more effective in treating cachexia. Ideally, interventions for cachexia should be initiated in the early stages of weight loss (i.e. pre-cachexia); however, patients are often referred for cachexia interventions late in their disease trajectory (i.e. refractory cachexia). At this stage, patients are in a catabolic state and respond poorly to anti-cancer treatment(Reference Fearon, Strasser and Anker8). Experts have speculated that the failure of treatments to show benefit at clinical evaluation may not have been because of the drugs ability to ameliorate or treat cancer cachexia, but because they were introduced to patients too late(Reference Martin and Sawyer129).

Rehabilitation care to modify body composition, either increasing muscle mass and/or MA should be conducted, and its respective impact on oncology outcomes explored. Previous investigations have shown that intramuscular adipose tissue was responsive to exercise in non-cancer individuals(Reference Marcus, Addison and Kidde130), and that cancer patients have anabolic potential(Reference Prado, Sawyer and Ghosh117). Current evidence in evaluating the efficacy of prehabilitation on postoperative outcomes among cancer patients is still limited(Reference Bruns, Argillander and Van Den Heuvel131, Reference Santa Mina, Clarke and Ritvo132). Future clinical trials are warranted to test whether the window of time between cancer diagnosis and initiation of treatment is an opportunity to commence interventions to increase muscle mass and MA.

Nutritional interventions

A meta-analysis of oral nutritional interventions in malnourished patients with cancer identified thirteen randomised controlled trials and included 1414 patients. The analysis, conducted by Baldwin et al. (Reference Baldwin, Spiro and Ahern133), concluded that oral nutritional interventions had no significant effect on body weight, energy intake or mortality compared with standard care. Given the wide range of pathophysiological alterations that occur in cancer, complex and individualised targeted strategies incorporating modulations of the metabolic components of cachexia (e.g. inflammation) may be required to allow nutrition interventions to be more effective(Reference Arends, Baracos and Bertz134). To date, n-3 fatty acids, such as EPA have received a lot of attention for the treatment of cancer cachexia due to their potent anti-inflammatory properties. In cancer, plasma n-3 fatty acid levels appear to be depleted in patients with sarcopenia and may contribute to accelerate muscle mass(Reference Murphy, Mourtzakis and Chu135). The results of several small clinical trials suggested that using EPA supplements or oral nutritional supplements containing EPA in patients with advanced cancer improved appetite, energy intake, body weight, lean mass and/or physical activity(Reference Silva, Trindade and Fabre136Reference Ryan, Reynolds and Healy139). The n-3 fatty acid supplementation has also been shown to be effective in reducing intramuscular adipose tissue while maintaining muscle mass (compared with standard care) in a small cohort of patients with non-small-cell lung cancer(Reference Murphy, Mourtzakis and Chu138), supporting what is observed in preclinical experimental animal models(Reference Almasud, Giles and Miklavcic140). However, the clinical evidence supporting n-3 fatty acids remain inconclusive. Three systematic reviews published in 2007, 2009 and 2012 concluded that there was insufficient evidence to support a recommendation for n-3 fatty acids to treat muscle depletion and cancer cachexia(Reference Ries, Trottenberg and Elsner141Reference Arends, Bachmann and Baracos144). Due to the inconsistencies in the reported effects of n-3 fatty acids(Reference Fearon, Barber and Moses145, Reference Jatoi, Rowland and Loprinzi146), the ESPEN oncology nutrition guidelines issue only a weak recommendation for their use to stabilise or improve appetite, food intake, lean mass and body weight(Reference Arends, Bachmann and Baracos144).

Achieving adequate protein intake is an essential component to achieving muscle anabolism; however, no optimal amino acid and protein requirements have been established to prevent or treat sarcopenia in patients with cancer. Current recommendations suggest intakes of 1–1·5 g/kg/d in patients with cancer(Reference Arends, Bachmann and Baracos144); however, many patients fail to reach these requirements(Reference Prado, Lieffers and Bergsten147). Moreover, recent research has suggested that protein requirements should be based on the measures of lean mass as opposed to body weight(Reference Geisler, Prado and Müller148, Reference Prado, Cushen and Orsso149).

Physical activity

A recent meta-analysis(Reference Padilha, Marinello and Galvão150) has shown resistance exercise to be effective in increasing muscle strength and mass in cancer patients undergoing neoadjuvant and adjuvant therapy, with mean increases in lower limb strength of 26·2 kg (P = 0·00001) and lean mass by 0·8 kg (P < 0·00001)(Reference Padilha, Marinello and Galvão150). In a study of early-stage breast cancer patients receiving adjuvant chemotherapy, resistance exercise resulted in the reversal of sarcopenia and dynapenia in 43 and 59 % of patients, respectively(Reference Adams, Segal and McKenzie151). To date, most studies have been conducted in early-stage breast and prostate cancer and evidence of the effect of physical activity on muscle mass and strength in patients with advanced disease and cancer cachexia is lacking(Reference Stene, Helbostad and Balstad152, Reference Grande, Silva and Maddocks153). A recent Cochrane systemic review(Reference Grande, Silva and Maddocks153), which aimed to examine the impact of exercise on lean mass in patients with cancer cachexia, showed disappointing results. The authors screened more than 3000 individual articles, but found no trials that met the inclusion criteria for analysis(Reference Grande, Silva and Maddocks153). Initial findings indicate that exercise is safe and well tolerated in patients with advanced disease(Reference Heywood, McCarthy and Skinner154) and improves physical performance and several domains of quality of life(Reference Salakari, Surakka and Nurminen155), but further work is needed. Evidence of interventions aimed at improving MA in patients with cancer are scarce; however, evidence in older adults suggests that exercise may represent a counter measure to improve MA, as studies have shown that muscle fatty infiltration is amenable to change after 12 weeks of thrice-weekly resistance training(Reference Marcus, Addison and Kidde130).

Recently, a few studies have demonstrated that exercise interventions are capable of improving cancer outcomes. Cho et al. (Reference Cho, Yoshikawa and Oba156) reported that preoperative exercise significantly improved operative risk factors and decreased the frequency of serious postoperative complications in gastric cancer patients. In breast cancer, a supervised moderate- to high-intensity exercise programme during adjuvant chemotherapy was associated with a beneficial effect on chemotherapy completion rates with a significantly smaller proportion of patients requiring dose adjustments compared with usual care (12 v. 34 %)(Reference van Waart, Stuiver and van Harten157). The impact of exercise prehabilitation on cancer outcome warrants further investigation.

Pharmacological agents

To date, no pharmacotherapies have been approved to specifically treat the cancer cachexia syndrome. Non-steroidal anti-inflammatory drugs have been suggested as a potential treatment for cancer cachexia, with the aim of reducing systemic inflammation. In a recent systematic review of thirteen studies, eleven showed either improvement or stabilisation in weight or lean mass in patients with cancer(Reference Solheim, Fearon and Blum158); however, the evidence is still insufficient to recommend non-steroidal anti-inflammatory drugs to treat cancer cachexia outside of clinical trials. Promising results have been obtained for anamorelin (a ghrelin receptor agonist); phase III trials (ROMANA 1 and ROMANA 2) have shown that anamorelin significantly increases lean mass and improves anorexia/cachexia symptoms compared with placebo over a 12-week intervention period. However, muscle function (measured by hand grip strength) failed to improve during the intervention(Reference Temel, Abernethy and Currow159). Food and drug administration regulators generally require improvements in lean mass and functional outcomes as co-primary endpoints for the approval of new medications to treat cancer cachexia. As a result, anamorelin has not received the food and drug administration's approval to date. Phase III trials have recently been completed for enobosarm, a selective androgen receptor modulator (NCT01355484), and MABp1, an anti Il-1 α monoclonal antibody (NCT01767857), the results of which are yet to be published.

Multimodal interventions

Results so far suggest that single-agent therapies may be insufficient to counteract cancer cachexia and that early multimodal interventions may be necessary to combat its multifactorial and complex pathogenesis. A multimodal approach including the use of individualised nutritional interventions to promote energy balance and ensure optimal protein intake, decreasing inflammation and hyper metabolic stress with the aid of anti-inflammatory agents (non-steroidal anti-inflammatory drugs s and EPA) and increasing physical activity to stabilise/increase muscle mass, strength and physical performance has been recommended(Reference Arends, Baracos and Bertz134). One such intervention currently under investigation is the MENAC trial (Multimodal Exercise/Nutrition/Anti-inflammatory treatment for Cachexia), whereby phase II feasibility studies have yielded encouraging results and suggest multimodal interventions are feasible and safe in patients with cancer(Reference Solheim, Laird and Balstad160). The phase III trial (NCT02330926) is currently being conducted across a number of international sites.

Conclusion

In summary, the evidence reviewed here shows that muscle abnormalities are highly prevalent in adult patients with cancer, across cancer sites and stages, and overweight and obesity do not preclude their presence. However, the heterogeneity in the diagnostic criteria limits the ability to accurately compare the reported prevalence rates between different cohorts and study findings overall. Efforts are required to standardise muscle measurements from CT images, as well as the diagnostic criteria for sarcopenia and low MA in patients with cancer. Nonetheless, the clinical importance of muscle abnormalities in these patients is evident, given their associations with negative clinical outcomes, such as poorer tolerance to chemotherapy, increased risk of post-operative complications and infections, increased length of hospital stays and poor prognosis.

In an age of increasingly personalised medicine, CT scans, obtained as part of routine patient care, provide valuable individualised information on muscle mass and MA with well-established prognostic implications. Incorporating routine assessment of muscle abnormalities from CT images into clinical practice has the potential to play a major role in stratifying patients at risk of poorer clinical outcomes, who may benefit from targeted interventions. At present, no effective medical intervention to improve muscle mass and MA exists; however, in recent years, substantial progress has been made, with the results of several clinical trials awaited in the near future. The optimal timing and treatment strategy for preventing or delaying the development of muscle abnormalities are yet to be determined, but multimodal interventions appear to hold the most promise.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0029665118000046

Acknowledgements

The authors would like to thank the Irish section of the Nutrition Society for inviting the present review paper as part of the postgraduate review competition. The authors would also like to acknowledge the support of the Health Research Board Clinical Research Facility, Cork.

Financial Support

This publication has emanated from research conducted with the financial support of Science Foundation Ireland (SFI) under grant number SFI/12/RC/2273.

Conflict of interest

None.

Authorship

L. E. D. reviewed the literature and wrote the manuscript. A. M. R. and C. M. P. participated in writing and revising the manuscript. All authors approved the final manuscript.

Footnotes

Presented by Louise Daly at The Nutrition Society Summer Meeting, Queens University Belfast, Belfast, Ireland, on 21–23 June 2017

References

1.Martin, L, Birdsell, L, Macdonald, N et al. (2013) Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol 31, 15391547.Google Scholar
2.Cespedes Feliciano, EM, Lee, VS, Prado, CM et al. (2017) Muscle mass at the time of diagnosis of nonmetastatic colon cancer and early discontinuation of chemotherapy, delays, and dose reductions on adjuvant FOLFOX: The C-SCANS study. Cancer 123, 48684877.CrossRefGoogle ScholarPubMed
3.van Vugt, JL, Braam, HJ, van Oudheusden, TR et al. (2015) Skeletal muscle depletion is associated with severe postoperative complications in patients undergoing cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol 22, 36253631.CrossRefGoogle ScholarPubMed
4.Daly, LE, Power, DG, O'Reilly, Á et al. (2017) The impact of body composition parameters on ipilimumab toxicity and survival in patients with metastatic melanoma. Br J Cancer 116, 310317.Google Scholar
5.Janssen, I & Ross, R (2005) Linking age-related changes in skeletal muscle mass and composition with metabolism and disease. J Nutr Health Aging 9, 408419.Google ScholarPubMed
6.Goodpaster, BH, Park, SW, Harris, TB et al. (2006) The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study. J Gerontol A Biol Sci Med Sci 61, 10591064.Google Scholar
7.Baumgartner, RN, Koehler, KM, Gallagher, D et al. (1998) Epidemiology of sarcopenia among the elderly in New Mexico. Am J Epidemiol 147, 755763.CrossRefGoogle ScholarPubMed
8.Fearon, K, Strasser, F, Anker, SD et al. (2011) Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 12, 489495.Google Scholar
9.Argilés, JM, Busquets, S, Stemmler, B et al. (2014) Cancer cachexia: understanding the molecular basis. Nat Rev Cancer 14, 754762.CrossRefGoogle ScholarPubMed
10.Donohoe, CL, Ryan, AM & Reynolds, JV (2011) Cancer cachexia: mechanisms and clinical implications. Gastroenterol Res Pract 2011, 601434.Google Scholar
11.Aubrey, J, Esfandiari, N, Baracos, VE et al. (2014) Measurement of skeletal muscle radiation attenuation and basis of its biological variation. Acta Physiol (Oxf) 210, 489497.Google Scholar
12.Chu, MP, Lieffers, J, Ghosh, S et al. (2015) Skeletal muscle radio-density is an independent predictor of response and outcomes in follicular lymphoma treated with chemoimmunotherapy. PLoS ONE 10, e0127589.CrossRefGoogle ScholarPubMed
13.Hayashi, N, Ando, Y, Gyawali, B et al. (2016) Low skeletal muscle density is associated with poor survival in patients who receive chemotherapy for metastatic gastric cancer. Oncol Rep 35, 17271731.CrossRefGoogle ScholarPubMed
14.Rier, HN, Jager, A, Sleijfer, S et al. (2017) Low muscle attenuation is a prognostic factor for survival in metastatic breast cancer patients treated with first line palliative chemotherapy. Breast 31, 915.Google Scholar
15.Chu, MP, Lieffers, J, Ghosh, S et al. (2017) Skeletal muscle density is an independent predictor of diffuse large B-cell lymphoma outcomes treated with rituximab-based chemoimmunotherapy. J Cachexia Sarcopenia Muscle 8, 298304.CrossRefGoogle ScholarPubMed
16.Van Rijssen, LB, van Huijgevoort, NC, Coelen, RJ et al. (2017) Skeletal muscle quality is associated with worse survival after pancreatoduodenectomy for periampullary, nonpancreatic cancer. Ann Surg Oncol 24, 272280.Google Scholar
17.van Dijk, DP, Bakens, MJ, Coolsen, MM et al. (2017) Low skeletal muscle radiation attenuation and visceral adiposity are associated with overall survival and surgical site infections in patients with pancreatic cancer. J Cachexia Sarcopenia Muscle 8, 317326.Google Scholar
18.Sjøblom, B, Grønberg, BH, Wentzel-Larsen, T et al. (2016) Skeletal muscle radiodensity is prognostic for survival in patients with advanced non-small cell lung cancer. Clin Nutr 35, 13861393.CrossRefGoogle ScholarPubMed
19.Prado, CM, Lieffers, JR, McCargar, LJ et al. (2008) Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol 9, 629635.CrossRefGoogle ScholarPubMed
20.Ryan, AM, Power, DG, Daly, L et al. (2016) Cancer-associated malnutrition, cachexia and sarcopenia: the skeleton in the hospital closet 40 years later. Proc Nutr Soc 75, 199211.CrossRefGoogle ScholarPubMed
21.Prado, CM & Heymsfield, SB (2014) Lean tissue imaging: a new era for nutritional assessment and intervention. JPEN J Parenter Enteral Nutr 38, 940953.Google Scholar
22.Shen, W, Punyanitya, M, Wang, Z et al. (2004) Total body skeletal muscle and adipose tissue volumes: estimation from a single abdominal cross-sectional image. J Appl Physiol (1985) 97, 23332338.CrossRefGoogle ScholarPubMed
23.Cruz-Jentoft, AJ, Baeyens, JP, Bauer, JM et al. (2010) Sarcopenia: European consensus on definition and diagnosis: report of the European working group on sarcopenia in older people. Age Ageing 39, 412423.CrossRefGoogle ScholarPubMed
24.Fielding, RA, Vellas, B, Evans, WJ et al. (2011) Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences. International working group on sarcopenia. J Am Med Dir Assoc 12, 249256.CrossRefGoogle ScholarPubMed
25.Prado, CM, Lieffers, JR, Bowthorpe, L et al. (2013) Sarcopenia and physical function in overweight patients with advanced cancer. Can J Diet Pract Res 74, 6974.CrossRefGoogle ScholarPubMed
26.Prado, CM, Baracos, VE, Reiman, T et al. (2010) Overweight and obese patients with solid tumors may have sarcopenia, poor prognosis and early features of cachexia. International Journal of Body Composition Research 8, 715.Google Scholar
27.Baracos, VE, Reiman, T, Mourtzakis, M et al. (2010) Body composition in patients with non-small cell lung cancer: a contemporary view of cancer cachexia with the use of computed tomography image analysis. Am J Clin Nutr 91, 1133S1137S.Google Scholar
28.Tamandl, D, Paireder, M, Asari, R et al. (2016) Markers of sarcopenia quantified by computed tomography predict adverse long-term outcome in patients with resected oesophageal or gastro-oesophageal junction cancer. Eur Radiol 26, 13591367.Google Scholar
29.Mourtzakis, M, Prado, CM, Lieffers, JR et al. (2008) A practical and precise approach to quantification of body composition in cancer patients using computed tomography images acquired during routine care. Appl Physiol Nutr Metab 33, 9971006.Google Scholar
30.Caan, BJ, Meyerhardt, JA, Kroenke, CH et al. (2017) Explaining the obesity paradox: the association between body composition and colorectal cancer survival (C-SCANS study). Cancer Epidemiol Biomarkers Prev 26, 10081015.CrossRefGoogle ScholarPubMed
31.Fujiwara, N, Nakagawa, H, Kudo, Y et al. (2015) Sarcopenia, intramuscular fat deposition, and visceral adiposity independently predict the outcomes of hepatocellular carcinoma. J Hepatol 63, 131140.Google Scholar
32.Iritani, S, Imai, K, Takai, K et al. (2015) Skeletal muscle depletion is an independent prognostic factor for hepatocellular carcinoma. J Gastroenterol 50, 323332.Google Scholar
33.Lanic, H, Kraut-Tauzia, J, Modzelewski, R et al. (2014) Sarcopenia is an independent prognostic factor in elderly patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Leuk Lymphoma 55, 817823.CrossRefGoogle ScholarPubMed
34.Coelen, RJ, Wiggers, JK, Nio, CY et al. (2015) Preoperative computed tomography assessment of skeletal muscle mass is valuable in predicting outcomes following hepatectomy for perihilar cholangiocarcinoma. HPB (Oxford) 17, 520528.CrossRefGoogle ScholarPubMed
35.Heymsfield, SB, Gonzalez, MC, Lu, J et al. (2015) Skeletal muscle mass and quality: evolution of modern measurement concepts in the context of sarcopenia. Proc Nutr Soc 74, 355366.Google Scholar
36.Dalal, S, Hui, D, Bidaut, L et al. (2012) Relationships among body mass index, longitudinal body composition alterations, and survival in patients with locally advanced pancreatic cancer receiving chemoradiation: a pilot study. J Pain Symptom Manage 44, 181191.Google Scholar
37.Stene, GB, Helbostad, JL, Amundsen, T et al. (2014) Changes in skeletal muscle mass during palliative chemotherapy in patients with advanced lung cancer. Acta Oncol 54, 340348.Google Scholar
38.Antoun, S, Birdsell, L, Sawyer, MB et al. (2010) Association of skeletal muscle wasting with treatment with sorafenib in patients with advanced renal cell carcinoma: results from a placebo-controlled study. J Clin Oncol 28, 10541060.CrossRefGoogle ScholarPubMed
39.Veasey Rodrigues, H, Baracos, VE, Wheler, JJ et al. (2013) Body composition and survival in the early clinical trials setting. Eur J Cancer 49, 30683075.CrossRefGoogle ScholarPubMed
40.Elliott, JA, Doyle, SL, Murphy, CF et al. (2017) Sarcopenia: prevalence, and impact on operative and oncologic outcomes in the multimodal management of locally advanced esophageal cancer. Ann Surg 266, 822830.Google Scholar
41.Miyamoto, Y, Baba, Y, Sakamoto, Y et al. (2015) Sarcopenia is a negative prognostic factor after curative resection of colorectal cancer. Ann Surg Oncol 22, 26632668.Google Scholar
42.Harada, K, Ida, S, Baba, Y et al. (2016) Prognostic and clinical impact of sarcopenia in esophageal squamous cell carcinoma. Dis Esophagus 29, 627633.Google Scholar
43.Rutten, IJ, van Dijk, DP, Kruitwagen, RF et al. (2016) Loss of skeletal muscle during neoadjuvant chemotherapy is related to decreased survival in ovarian cancer patients. J Cachexia Sarcopenia Muscle 7, 458466.CrossRefGoogle ScholarPubMed
44.Boer, BC, de Graaff, F, Brusse-Keizer, M et al. (2016) Skeletal muscle mass and quality as risk factors for postoperative outcome after open colon resection for cancer. Int J Colorectal Dis 31, 11171124.Google Scholar
45.Amini, N, Spolverato, G, Gupta, R et al. (2015) Impact total psoas volume on short- and long-term outcomes in patients undergoing curative resection for pancreatic adenocarcinoma: a new tool to assess sarcopenia. J Gastrointest Surg 19, 15931602.Google Scholar
46.Joglekar, S, Asghar, A, Mott, SL et al. (2015) Sarcopenia is an independent predictor of complications following pancreatectomy for adenocarcinoma. J Surg Oncol 111, 771775.Google Scholar
47.Peng, P, Hyder, O, Firoozmand, A et al. (2012) Impact of sarcopenia on outcomes following resection of pancreatic adenocarcinoma. J Gastrointest Surg 16, 14781486.Google Scholar
48.Rutten, IJG, Ubachs, J, Kruitwagen, RFPM et al. (2017) Psoas muscle area is not representative of total skeletal muscle area in the assessment of sarcopenia in ovarian cancer. J Cachexia Sarcopenia Muscle 8, 630638.CrossRefGoogle Scholar
49.Baracos, VE (2017) Psoas as a sentinel muscle for sarcopenia: a flawed premise. J Cachexia Sarcopenia Muscle 8, 527528.CrossRefGoogle ScholarPubMed
50.Huang, DD, Zhou, CJ, Wang, SL et al. (2017) Impact of different sarcopenia stages on the postoperative outcomes after radical gastrectomy for gastric cancer. Surgery 161, 680693.Google Scholar
51.Fukuda, Y, Yamamoto, K, Hirao, M et al. (2016) Sarcopenia is associated with severe postoperative complications in elderly gastric cancer patients undergoing gastrectomy. Gastric Cancer 19, 986993.Google Scholar
52.Chen, LK, Liu, LK, Woo, J et al. (2014) Sarcopenia in Asia: consensus report of the Asian working group for sarcopenia. J Am Med Dir Assoc 15, 95101.Google Scholar
53.Auclin, E, Bourillon, C, De Maio, E et al. (2017) Prediction of everolimus toxicity and prognostic value of skeletal muscle index in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 15, 350355.CrossRefGoogle ScholarPubMed
54.van Vledder, MG, Levolger, S, Ayez, N et al. (2012) Body composition and outcome in patients undergoing resection of colorectal liver metastases. Br J Surg 99, 550557.CrossRefGoogle ScholarPubMed
55.Barret, M, Antoun, S, Dalban, C et al. (2014) Sarcopenia is linked to treatment toxicity in patients with metastatic colorectal cancer. Nutr Cancer 66, 583589.Google Scholar
56.Cushen, SJ, Power, DG, Teo, MY et al. (2017) Body composition by computed tomography as a predictor of toxicity in patients with renal cell carcinoma treated with sunitinib. Am J Clin Oncol 40, 4752.Google Scholar
57.Choi, Y, Oh, DY, Kim, TY et al. (2015) Skeletal muscle depletion predicts the prognosis of patients with advanced pancreatic cancer undergoing palliative chemotherapy, independent of body mass index. PLoS ONE 10, e0139749.CrossRefGoogle ScholarPubMed
58.Wesseltoft-Rao, N, Hjermstad, MJ, Ikdahl, T et al. (2015) Comparing two classifications of cancer cachexia and their association with survival in patients with unresected pancreatic cancer. Nutr Cancer 67, 472480.CrossRefGoogle ScholarPubMed
59.Carneiro, IP, Mazurak, VC & Prado, CM (2016) Clinical implications of sarcopenic obesity in cancer. Curr Oncol Rep 18, 62.Google Scholar
60.Bowden, JCS, Williams, LJ, Simms, A et al. (2017) Prediction of 90 day and overall survival after chemoradiotherapy for lung cancer: role of performance status and body composition. Clin Oncol (R Coll Radiol) 29, 576584.Google Scholar
61.Kumar, A, Moynagh, MR, Multinu, F et al. (2016) Muscle composition measured by CT scan is a measurable predictor of overall survival in advanced ovarian cancer. Gynecol Oncol 142, 311316.CrossRefGoogle ScholarPubMed
62.Aust, S, Knogler, T, Pils, D et al. (2015) Skeletal muscle depletion and markers for cancer cachexia are strong prognostic factors in epithelial ovarian cancer. PLoS ONE 10, e0140403.Google Scholar
63.Okumura, S, Kaido, T, Hamaguchi, Y et al. (2017) Visceral adiposity and sarcopenic visceral obesity are associated with poor prognosis after resection of pancreatic cancer. Ann Surg Oncol 24, 37323740.CrossRefGoogle ScholarPubMed
64.Antoun, S, Lanoy, E, Iacovelli, R et al. (2013) Skeletal muscle density predicts prognosis in patients with metastatic renal cell carcinoma treated with targeted therapies. Cancer 119, 33773384.Google Scholar
65.Akahori, T, Sho, M, Kinoshita, S et al. (2015) Prognostic significance of muscle attenuation in pancreatic cancer patients treated with neoadjuvant chemoradiotherapy. World J Surg 39, 29752982.CrossRefGoogle ScholarPubMed
66.Cushen, SJ, Power, DG, Murphy, KP et al. (2016) Impact of body composition parameters on clinical outcomes in patients with metastatic castrate-resistant prostate cancer treated with docetaxel. Clin Nutr 13, e39e45.Google Scholar
67.Ní Bhuachalla, É, Daly, LE, Power, DG et al. (2017) Computed tomography diagnosed cachexia and sarcopenia in 725 oncology patients: is nutritional screening capturing hidden malnutrition? J Cachexia Sarcopenia Muscle [Epublication ahead of print version].Google ScholarPubMed
68.Rollins, KE, Tewari, N, Ackner, A et al. (2015) The impact of sarcopenia and myosteatosis on outcomes of unresectable pancreatic cancer or distal cholangiocarcinoma. Clin Nutr 35, 11031109.Google Scholar
69.McSorley, ST, Black, DH, Horgan, PG et al. (2017) The relationship between tumour stage, systemic inflammation, body composition and survival in patients with colorectal cancer. Clin Nutr [Epublication ahead of print version].Google ScholarPubMed
70.Malietzis, G, Currie, AC, Athanasiou, T et al. (2016) Influence of body composition profile on outcomes following colorectal cancer surgery. Br J Surg 103, 572580.Google Scholar
71.Prado, CM, Baracos, VE, McCargar, LJ et al. (2009) Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment. Clin Cancer Res 15, 29202926.Google Scholar
72.Shachar, SS, Deal, AM, Weinberg, M et al. (2017) Skeletal muscle measures as predictors of toxicity, hospitalization, and survival in patients with metastatic breast cancer receiving taxane-based chemotherapy. Clin Cancer Res 23, 658665.Google Scholar
73.Antoun, S, Baracos, VE, Birdsell, L et al. (2010) Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma. Ann Oncol 21, 15941598.Google Scholar
74.Huillard, O, Mir, O, Peyromaure, M et al. (2013) Sarcopenia and body mass index predict sunitinib-induced early dose-limiting toxicities in renal cancer patients. Br J Cancer 108, 10341041.Google Scholar
75.Mir, O, Coriat, R, Blanchet, B et al. (2012) Sarcopenia predicts early dose-limiting toxicities and pharmacokinetics of sorafenib in patients with hepatocellular carcinoma. PLoS ONE 7, e37563.Google Scholar
76.Sjøblom, B, Grønberg, BH, Benth, J et al. (2015) Low muscle mass is associated with chemotherapy-induced haematological toxicity in advanced non-small cell lung cancer. Lung Cancer 90, 8591.CrossRefGoogle ScholarPubMed
77.Chemama, S, Bayar, MA, Lanoy, E et al. (2016) Sarcopenia is associated with chemotherapy toxicity in patients undergoing cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis from colorectal cancer. Ann Surg Oncol 23, 38913898.CrossRefGoogle ScholarPubMed
78.Massicotte, MH, Borget, I, Broutin, S et al. (2013) Body composition variation and impact of low skeletal muscle mass in patients with advanced medullary thyroid carcinoma treated with vandetanib: results from a placebo-controlled study. J Clin Endocrinol Metab 98, 24012408.CrossRefGoogle ScholarPubMed
79.Valentine, H, François, G, Nora, K et al. (2017) Sarcopenic overweight is associated with early acute limiting toxicity of anti-PD1 checkpoint inhibitors in melanoma patients. Invest New Drugs 35, 436441.Google Scholar
80.Prado, CM, Baracos, VE, McCargar, LJ et al. (2007) Body composition as an independent determinant of 5-fluorouracil-based chemotherapy toxicity. Clin Cancer Res 13, 32643268.Google Scholar
81.Palmela, C, Velho, S, Agostinho, L et al. (2017) Body composition as a prognostic factor of neoadjuvant chemotherapy toxicity and outcome in patients with locally advanced gastric cancer. J Gastric Cancer 17, 7487.CrossRefGoogle ScholarPubMed
82.Shachar, SS, Deal, AM, Weinberg, M et al. (2017) Body composition as a predictor of toxicity in patients receiving anthracycline and taxane-based chemotherapy for early-stage breast cancer. Clin Cancer Res 23, 35373543.Google Scholar
83.Anandavadivelan, P, Brismar, TB, Nilsson, M et al. (2016) Sarcopenic obesity: a probable risk factor for dose limiting toxicity during neo-adjuvant chemotherapy in oesophageal cancer patients. Clin Nutr 35, 724730.CrossRefGoogle ScholarPubMed
84.Tan, BH, Brammer, K, Randhawa, N et al. (2015) Sarcopenia is associated with toxicity in patients undergoing neo-adjuvant chemotherapy for oesophago-gastric cancer. Eur J Surg Oncol 41, 333338.Google Scholar
85.Yip, C, Goh, V, Davies, A et al. (2014) Assessment of sarcopenia and changes in body composition after neoadjuvant chemotherapy and associations with clinical outcomes in oesophageal cancer. Eur Radiol 24, 9981005.CrossRefGoogle ScholarPubMed
86.Parsons, HA, Tsimberidou, AM, Pontikos, M et al. (2012) Evaluation of the clinical relevance of body composition parameters in patients with cancer metastatic to the liver treated with hepatic arterial infusion chemotherapy. Nutr Cancer 64, 206217.Google Scholar
87.Veasey-Rodrigues, H, Parsons, HA, Janku, F et al. (2013) A pilot study of temsirolimus and body composition. J Cachexia Sarcopenia Muscle 4, 259265.Google Scholar
88.Blauwhoff-Buskermolen, S, Versteeg, KS, de van der Schueren, MA et al. (2016) Loss of muscle mass during chemotherapy is predictive for poor survival of patients with metastatic colorectal cancer. J Clin Oncol 34, 13391344.Google Scholar
89.Felici, A, Verweij, J & Sparreboom, A (2002) Dosing strategies for anticancer drugs: the good, the bad and body-surface area. Eur J Cancer 38, 16771684.Google Scholar
90.Prado, CM, Lima, IS, Baracos, VE et al. (2011) An exploratory study of body composition as a determinant of epirubicin pharmacokinetics and toxicity. Cancer Chemother Pharmacol 67, 93101.Google Scholar
91.Prado, CM, Baracos, VE, Xiao, J et al. (2014) The association between body composition and toxicities from the combination of doxil and trabectedin in patients with advanced relapsed ovarian cancer. Appl Physiol Nutr Metab 39, 693698.Google Scholar
92.Ali, R, Baracos, VE, Sawyer, MB et al. (2016) Lean body mass as an independent determinant of dose-limiting toxicity and neuropathy in patients with colon cancer treated with FOLFOX regimens. Cancer Med 5, 607616.Google Scholar
93.Sjøblom, B, Benth, J, Grønberg, BH et al. (2017) Drug dose per kilogram lean body mass predicts hematologic toxicity from carboplatin-doublet chemotherapy in advanced non-small-cell lung cancer. Clin Lung Cancer 18, e129e136.Google Scholar
94.Antoun, S, Borget, I & Lanoy, E (2013) Impact of sarcopenia on the prognosis and treatment toxicities in patients diagnosed with cancer. Curr Opin Support Palliat Care 7, 383389.Google Scholar
95.Tan, BH, Birdsell, LA, Martin, L et al. (2009) Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin Cancer Res 15, 69736979.Google Scholar
96.Sharma, P, Zargar-Shoshtari, K, Caracciolo, JT et al. (2015) Sarcopenia as a predictor of overall survival after cytoreductive nephrectomy for metastatic renal cell carcinoma. Urol Oncol 33, 339.e317–323.Google Scholar
97.Fukushima, H, Nakanishi, Y, Kataoka, M et al. (2016) Prognostic significance of sarcopenia in patients with metastatic renal cell carcinoma. J Urol 195, 2632.Google Scholar
98.Meza-Junco, J, Montano-Loza, AJ, Baracos, VE et al. (2013) Sarcopenia as a prognostic index of nutritional status in concurrent cirrhosis and hepatocellular carcinoma. J Clin Gastroenterol 47, 861870.Google Scholar
99.Voron, T, Tselikas, L, Pietrasz, D et al. (2015) Sarcopenia impacts on short- and long-term results of hepatectomy for hepatocellular carcinoma. Ann Surg 261, 11731183.Google Scholar
100.Levolger, S, van Vledder, MG, Muslem, R et al. (2015) Sarcopenia impairs survival in patients with potentially curable hepatocellular carcinoma. J Surg Oncol 112, 208213.Google Scholar
101.Kim, EY, Kim, YS, Park, I et al. (2015) Prognostic significance of CT-determined sarcopenia in patients with small-cell lung cancer. J Thorac Oncol 10, 17951799.Google Scholar
102.Psutka, SP, Carrasco, A, Schmit, GD et al. (2014) Sarcopenia in patients with bladder cancer undergoing radical cystectomy: impact on cancer-specific and all-cause mortality. Cancer 120, 29102918.Google Scholar
103.Fukushima, H, Yokoyama, M, Nakanishi, Y et al. (2015) Sarcopenia as a prognostic biomarker of advanced urothelial carcinoma. PLoS ONE 10, e0115895.Google Scholar
104.Shachar, SS, Williams, GR, Muss, HB et al. (2016) Prognostic value of sarcopenia in adults with solid tumours: a meta-analysis and systematic review. Eur J Cancer 57, 5867.Google Scholar
105.Kazemi-Bajestani, SM, Mazurak, VC & Baracos, V (2016) Computed tomography-defined muscle and fat wasting are associated with cancer clinical outcomes. Semin Cell Dev Biol 54, 210.Google Scholar
106.Gibson, DJ, Burden, ST, Strauss, BJ et al. (2015) The role of computed tomography in evaluating body composition and the influence of reduced muscle mass on clinical outcome in abdominal malignancy: a systematic review. Eur J Clin Nutr 69, 10791086.Google Scholar
107.Tramèr, MR, Reynolds, DJ, Moore, RA et al. (1997) Impact of covert duplicate publication on meta-analysis: a case study. BMJ 315, 635640.Google Scholar
108.Miyamoto, Y, Baba, Y, Sakamoto, Y et al. (2015) Negative impact of skeletal muscle loss after systemic chemotherapy in patients with unresectable colorectal cancer. PLoS ONE 10, e0129742.Google Scholar
109.Sabel, MS, Lee, J, Cai, S et al. (2011) Sarcopenia as a prognostic factor among patients with stage III melanoma. Ann Surg Oncol 18, 35793585.Google Scholar
110.Lieffers, JR, Bathe, OF, Fassbender, K et al. (2012) Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. Br J Cancer 107, 931936.Google Scholar
111.Peng, PD, van Vledder, MG, Tsai, S et al. (2011) Sarcopenia negatively impacts short-term outcomes in patients undergoing hepatic resection for colorectal liver metastasis. HPB (Oxford) 13, 439446.Google Scholar
112.Lou, N, Chi, CH, Chen, XD et al. (2017) Sarcopenia in overweight and obese patients is a predictive factor for postoperative complication in gastric cancer: a prospective study. Eur J Surg Oncol 43, 188195.Google Scholar
113.Ida, S, Watanabe, M, Yoshida, N et al. (2015) Sarcopenia is a predictor of postoperative respiratory complications in patients with esophageal cancer. Ann Surg Oncol 22, 44324437.Google Scholar
114.Sharma, P, Zargar-Shoshtari, K, Caracciolo, JT et al. (2015) Sarcopenia as a predictor of complications in penile cancer patients undergoing inguinal lymph node dissection. World J Urol 33, 15851592.Google Scholar
115.van Vugt, JLA, Buettner, S, Levolger, S et al. (2017) Low skeletal muscle mass is associated with increased hospital expenditure in patients undergoing cancer surgery of the alimentary tract. PLoS ONE 12, e0186547.Google Scholar
116.Margadant, CC, Bruns, ER, Sloothaak, DA et al. (2016) Lower muscle density is associated with major postoperative complications in older patients after surgery for colorectal cancer. Eur J Surg Oncol 42, 16541659.Google Scholar
117.Prado, CM, Sawyer, MB, Ghosh, S et al. (2013) Central tenet of cancer cachexia therapy: do patients with advanced cancer have exploitable anabolic potential? Am J Clin Nutr 98, 10121019.Google Scholar
118.Awad, S, Tan, BH, Cui, H et al. (2012) Marked changes in body composition following neoadjuvant chemotherapy for oesophagogastric cancer. Clin Nutr 31, 7477.Google Scholar
119.Reisinger, KW, Bosmans, JW, Uittenbogaart, M et al. (2015) Loss of skeletal muscle mass during neoadjuvant chemoradiotherapy predicts postoperative mortality in esophageal cancer surgery. Ann Surg Oncol 22, 44454452.Google Scholar
120.Lieffers, JR, Mourtzakis, M, Hall, KD et al. (2009) A viscerally driven cachexia syndrome in patients with advanced colorectal cancer: contributions of organ and tumor mass to whole-body energy demands. Am J Clin Nutr 89, 11731179.Google Scholar
121.Frontera, WR, Hughes, VA, Fielding, RA et al. (2000) Aging of skeletal muscle: a 12-yr longitudinal study. J Appl Physiol (1985) 88, 13211326.CrossRefGoogle ScholarPubMed
122.Pezaro, C, Mukherji, D, Tunariu, N et al. (2013) Sarcopenia and change in body composition following maximal androgen suppression with abiraterone in men with castration-resistant prostate cancer. Br J Cancer 109, 325331.Google Scholar
123.Fogelman, DR, Holmes, H, Mohammed, K et al. (2014) Does IGFR1 inhibition result in increased muscle mass loss in patients undergoing treatment for pancreatic cancer? J Cachexia Sarcopenia Muscle 5, 307313.Google Scholar
124.Prado, CM, Bekaii-Saab, T, Doyle, LA et al. (2012) Skeletal muscle anabolism is a side effect of therapy with the MEK inhibitor: selumetinib in patients with cholangiocarcinoma. Br J Cancer 106, 15831586.Google Scholar
125.Cooper, AB, Slack, R, Fogelman, D et al. (2015) Characterization of anthropometric changes that occur during neoadjuvant therapy for potentially resectable pancreatic cancer. Ann Surg Oncol 22, 24162423.Google Scholar
126.Nattenmüller, J, Wochner, R, Muley, T et al. (2017) Prognostic impact of CT-quantified muscle and fat distribution before and after first-line-chemotherapy in lung cancer patients. PLoS ONE 12, e0169136.Google Scholar
127.Daly, LE, Ní Bhuachalla, É, Power, DG et al. (2017) Loss of skeletal muscle during systemic chemotherapy is prognostic of poor survival in patients with foregut cancer. J Cachexia Sarcopenia Muscle [Epublication ahead of print version].Google Scholar
128.Naito, T, Okayama, T, Aoyama, T et al. (2017) Skeletal muscle depletion during chemotherapy has a large impact on physical function in elderly Japanese patients with advanced non-small-cell lung cancer. BMC Cancer 17, 571.Google Scholar
129.Martin, L & Sawyer, MB (2015) Cancer cachexia: emerging pre-clinical evidence and the pathway forward to clinical trials. J Natl Cancer Inst 107, Issue 12, djv322.Google Scholar
130.Marcus, RL, Addison, O, Kidde, JP et al. (2010) Skeletal muscle fat infiltration: impact of age, inactivity, and exercise. J Nutr Health Aging 14, 362366.Google Scholar
131.Bruns, ERJ, Argillander, TE, Van Den Heuvel, B et al. (2017) Oral nutrition as a form of pre-operative enhancement in patients undergoing surgery for colorectal cancer: a systematic review. Surg Infect (Larchmt) 19, 110.Google Scholar
132.Santa Mina, D, Clarke, H, Ritvo, P et al. (2014) Effect of total-body prehabilitation on postoperative outcomes: a systematic review and meta-analysis. Physiotherapy 100, 196207.CrossRefGoogle ScholarPubMed
133.Baldwin, C, Spiro, A, Ahern, R et al. (2012) Oral nutritional interventions in malnourished patients with cancer: a systematic review and meta-analysis. J Natl Cancer Inst 104, 371385.Google Scholar
134.Arends, J, Baracos, V, Bertz, H et al. (2017) ESPEN expert group recommendations for action against cancer-related malnutrition. Clin Nutr 36, 11871196.Google Scholar
135.Murphy, RA, Mourtzakis, M, Chu, QS et al. (2010) Skeletal muscle depletion is associated with reduced plasma (n-3) fatty acids in non-small cell lung cancer patients. J Nutr 140, 16021606.Google Scholar
136.Silva, JeA, Trindade, EB, Fabre, ME et al. (2012) Fish oil supplement alters markers of inflammatory and nutritional status in colorectal cancer patients. Nutr Cancer 64, 267273.Google Scholar
137.van der Meij, BS, Langius, JA, Spreeuwenberg, MD et al. (2012) Oral nutritional supplements containing n-3 polyunsaturated fatty acids affect quality of life and functional status in lung cancer patients during multimodality treatment: an RCT. Eur J Clin Nutr 66, 399404.Google Scholar
138.Murphy, RA, Mourtzakis, M, Chu, QS et al. (2011) Nutritional intervention with fish oil provides a benefit over standard of care for weight and skeletal muscle mass in patients with nonsmall cell lung cancer receiving chemotherapy. Cancer 117, 17751782.Google Scholar
139.Ryan, AM, Reynolds, JV, Healy, L et al. (2009) Enteral nutrition enriched with eicosapentaenoic acid (EPA) preserves lean body mass following esophageal cancer surgery: results of a double-blinded randomized controlled trial. Ann Surg 249, 355363.Google Scholar
140.Almasud, AA, Giles, KH, Miklavcic, JJ et al. (2017) Fish oil mitigates myosteatosis and improves chemotherapy efficacy in a preclinical model of colon cancer. PLoS ONE 12, e0183576.Google Scholar
141.Ries, A, Trottenberg, P, Elsner, F et al. (2012) A systematic review on the role of fish oil for the treatment of cachexia in advanced cancer: an EPCRC cachexia guidelines project. Palliat Med 26, 294304.Google Scholar
142.Dewey, A, Baughan, C, Dean, T et al. (2007) Eicosapentaenoic acid (EPA, an omega-3 fatty acid from fish oils) for the treatment of cancer cachexia. Cochrane Database Syst Rev, CD004597.Google Scholar
143.Mazzotta, P & Jeney, CM (2009) Anorexia-cachexia syndrome: a systematic review of the role of dietary polyunsaturated Fatty acids in the management of symptoms, survival, and quality of life. J Pain Symptom Manage 37, 10691077.Google Scholar
144.Arends, J, Bachmann, P, Baracos, V et al. (2017) ESPEN guidelines on nutrition in cancer patients. Clin Nutr 36, 1148.Google Scholar
145.Fearon, KC, Barber, MD, Moses, AG et al. (2006) Double-blind, placebo-controlled, randomized study of eicosapentaenoic acid diester in patients with cancer cachexia. J Clin Oncol 24, 34013407.Google Scholar
146.Jatoi, A, Rowland, K, Loprinzi, CL et al. (2004) An eicosapentaenoic acid supplement versus megestrol acetate versus both for patients with cancer-associated wasting: a North Central Cancer Treatment Group and National Cancer Institute of Canada collaborative effort. J Clin Oncol 22, 24692476.Google Scholar
147.Prado, CM, Lieffers, JR, Bergsten, G et al. (2012) Dietary patterns of patients with advanced lung or colorectal cancer. Can J Diet Pract Res 73, e298e303.Google Scholar
148.Geisler, C, Prado, CM & Müller, MJ (2016) Inadequacy of body weight-based recommendations for individual protein intake-lessons from body composition analysis. Nutrients 9, 23.Google Scholar
149.Prado, CM, Cushen, SJ, Orsso, CE et al. (2016) Sarcopenia and cachexia in the era of obesity: clinical and nutritional impact. Proc Nutr Soc 75, 188198.Google Scholar
150.Padilha, CS, Marinello, PC, Galvão, DA et al. (2017) Evaluation of resistance training to improve muscular strength and body composition in cancer patients undergoing neoadjuvant and adjuvant therapy: a meta-analysis. J Cancer Surviv 11, 339349.Google Scholar
151.Adams, SC, Segal, RJ, McKenzie, DC et al. (2016) Impact of resistance and aerobic exercise on sarcopenia and dynapenia in breast cancer patients receiving adjuvant chemotherapy: a multicenter randomized controlled trial. Breast Cancer Res Treat 158, 497507.Google Scholar
152.Stene, GB, Helbostad, JL, Balstad, TR et al. (2013) Effect of physical exercise on muscle mass and strength in cancer patients during treatment – a systematic review. Crit Rev Oncol Hematol 88, 573593.Google Scholar
153.Grande, AJ, Silva, V & Maddocks, M (2015) Exercise for cancer cachexia in adults: executive summary of a Cochrane Collaboration systematic review. J Cachexia Sarcopenia Muscle 6, 208211.Google Scholar
154.Heywood, R, McCarthy, AL & Skinner, TL (2017) Safety and feasibility of exercise interventions in patients with advanced cancer: a systematic review. Support Care Cancer [Epublication ahead of print version].Google Scholar
155.Salakari, MR, Surakka, T, Nurminen, R et al. (2015) Effects of rehabilitation among patients with advances cancer: a systematic review. Acta Oncol 54, 618628.Google Scholar
156.Cho, H, Yoshikawa, T, Oba, MS et al. (2014) Matched pair analysis to examine the effects of a planned preoperative exercise program in early gastric cancer patients with metabolic syndrome to reduce operative risk: the Adjuvant Exercise for General Elective Surgery (AEGES) study group. Ann Surg Oncol 21, 20442050.Google Scholar
157.van Waart, H, Stuiver, MM, van Harten, WH et al. (2015) Effect of low-intensity physical activity and moderate- to high-intensity physical exercise during adjuvant chemotherapy on physical fitness, fatigue, and chemotherapy completion rates: results of the paces randomized clinical trial. J Clin Oncol 33, 19181927.Google Scholar
158.Solheim, TS, Fearon, KC, Blum, D et al. (2013) Non-steroidal anti-inflammatory treatment in cancer cachexia: a systematic literature review. Acta Oncol 52, 617.Google Scholar
159.Temel, JS, Abernethy, AP, Currow, DC et al. (2016) Anamorelin in patients with non-small-cell lung cancer and cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double-blind, phase 3 trials. Lancet Oncol 17, 519531.Google Scholar
160.Solheim, TS, Laird, BJA, Balstad, TR et al. (2017) A randomized phase II feasibility trial of a multimodal intervention for the management of cachexia in lung and pancreatic cancer. J Cachexia Sarcopenia Muscle 8, 778788.CrossRefGoogle ScholarPubMed
161.Kimura, M, Naito, T, Kenmotsu, H et al. (2015) Prognostic impact of cancer cachexia in patients with advanced non-small cell lung cancer. Support Care Cancer 23, 16991708.Google Scholar
162.Thoresen, L, Frykholm, G, Lydersen, S et al. (2012) The association of nutritional assessment criteria with health-related quality of life in patients with advanced colorectal carcinoma. Eur J Cancer Care (Engl) 21, 505516.Google Scholar
163.Thoresen, L, Frykholm, G, Lydersen, S et al. (2013) Nutritional status, cachexia and survival in patients with advanced colorectal carcinoma. Different assessment criteria for nutritional status provide unequal results. Clin Nutr 32, 6572.Google Scholar
164.Reisinger, KW, van Vugt, JL, Tegels, JJ et al. (2015) Functional compromise reflected by sarcopenia, frailty, and nutritional depletion predicts adverse postoperative outcome after colorectal cancer surgery. Ann Surg 261, 345352.Google Scholar
165.Malietzis, G, Johns, N, Al-Hassi, HO et al. (2016) Low muscularity and myosteatosis is related to the host systemic inflammatory response in patients undergoing surgery for colorectal cancer. Ann Surg 263, 320325.Google Scholar
166.Grotenhuis, B, Shapiro, J, van Adrichem, S et al. (2016) Sarcopenia/muscle mass is not a prognostic factor for short- and long-term outcome after esophagectomy for cancer. World J Surg 40, 26982704.Google Scholar
167.Nishigori, T, Okabe, H, Tanaka, E et al. (2016) Sarcopenia as a predictor of pulmonary complications after esophagectomy for thoracic esophageal cancer. J Surg Oncol 113, 678684.Google Scholar
168.Tegels, JJ, van Vugt, JL, Reisinger, KW et al. (2015) Sarcopenia is highly prevalent in patients undergoing surgery for gastric cancer but not associated with worse outcomes. J Surg Oncol 112, 403407.Google Scholar
169.Nishigori, T, Tsunoda, S, Okabe, H et al. (2016) Impact of sarcopenic obesity on surgical site infection after laparoscopic total gastrectomy. Ann Surg Oncol 23, 524531.Google Scholar
170.Kudou, K, Saeki, H, Nakashima, Y et al. (2017) Prognostic significance of sarcopenia in patients with esophagogastric junction cancer or upper gastric cancer. Ann Surg Oncol 24, 18041810.Google Scholar
171.Arrieta, O, De la Torre-Vallejo, M, López-Macías, D et al. (2015) Nutritional status, body surface, and low lean body mass/body mass index are related to dose reduction and severe gastrointestinal toxicity induced by afatinib in patients with non-small cell lung cancer. Oncologist 20, 967974.Google Scholar
172.Di Sebastiano, KM, Yang, L, Zbuk, K et al. (2013) Accelerated muscle and adipose tissue loss may predict survival in pancreatic cancer patients: the relationship with diabetes and anaemia. Br J Nutr 109, 302312.Google Scholar
173.Harimoto, N, Shirabe, K, Yamashita, YI et al. (2013) Sarcopenia as a predictor of prognosis in patients following hepatectomy for hepatocellular carcinoma. Br J Surg 100, 15231530.Google Scholar
174.Del Fabbro, E, Parsons, H, Warneke, CL et al. (2012) The relationship between body composition and response to neoadjuvant chemotherapy in women with operable breast cancer. Oncologist 17, 12401245.Google Scholar
175.Camus, V, Lanic, H, Kraut, J et al. (2014) Prognostic impact of fat tissue loss and cachexia assessed by computed tomography scan in elderly patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Eur J Haematol 93, 918.Google Scholar
176.Moryoussef, F, Dhooge, M, Volet, J et al. (2015) Reversible sarcopenia in patients with gastrointestinal stromal tumor treated with imatinib. J Cachexia Sarcopenia Muscle 6, 343350.Google Scholar
177.Cousin, S, Hollebecque, A, Koscielny, S et al. (2014) Low skeletal muscle is associated with toxicity in patients included in phase I trials. Invest New Drugs 32, 382387.Google Scholar
Figure 0

Table 1. Cut points for skeletal muscle index (SMI) at the third lumber vertebra (L3) associated with mortality in patients with cancer

Figure 1

Table 2. Prevalence of sarcopenia according to cancer site, country and definition used

Figure 2

Fig. 1. Sarcopenic patients experience more dose-limiting toxicities (DLT) to flurouracil (5-FU) in colon cancer(80); capecitabine in breast cancer(71); sorafenib in renal cell carcinoma(73); 5-flurouracil, epirubicin, cyclophosphamide (FEC) in breast cancer(90); sorafenib in hepatocellular carcinoma(75); sunitunib in renal cell carcinoma(74); vantetanib in medullary thyroid cancer(78); fluropyramidine in colorectal cancer(55); phase I drugs in mixed cancer types(177); imatinib in gastrointestinal stromal tumours (grade I–II toxicity)(176); epirubicin, cisplatin, capecitabine (ECX) and cisplatin, 5-flurouracil (CF) in oesophagogastric cancer(84); taxane-based chemotherapy (placitaxel, docetaxel, nab-paclitaxel) in breast cancer(72); hyperthermic intraperitoneal chemotherapy (oxaliplatin and irrinotecan) in colorectal cancer(77), neoadjuvant chemotherapy (NACT) (mixed types) in gastric cancer(81); ipilimumab in metastatic melanoma(4).

Figure 3

Table 3. Summary of studies reporting muscle loss during treatment as prognostic of reduced survival

Supplementary material: File

Daly et al. supplementary material

Table S1

Download Daly et al. supplementary material(File)
File 23.4 KB