Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-19T05:46:25.470Z Has data issue: false hasContentIssue false

The use of alternate vertebral levels to L3 in computed tomography scans for skeletal muscle mass evaluation and sarcopenia assessment in patients with cancer: a systematic review

Published online by Cambridge University Press:  29 April 2021

Belinda Vangelov*
Affiliation:
Department of Radiation Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital and Community Health Services, Sydney, NSW, Australia Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
Judy Bauer
Affiliation:
School of Human Movement and Nutrition Sciences, Faculty of Health and Behavioural Sciences, University of Queensland, Brisbane, QLD, Australia
Damian Kotevski
Affiliation:
Department of Radiation Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital and Community Health Services, Sydney, NSW, Australia
Robert I. Smee
Affiliation:
Department of Radiation Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital and Community Health Services, Sydney, NSW, Australia Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia Department of Radiation Oncology, Tamworth Base Hospital, Tamworth, NSW, Australia
*
*Corresponding author: Belinda Vangelov, email belinda.vangelov@health.nsw.gov.au
Rights & Permissions [Opens in a new window]

Abstract

Body composition measurement using diagnostic computed tomography (CT) scans has emerged as a method to assess sarcopenia (low muscle mass) in oncology patients. Assessment of skeletal muscle mass (SMM) using the cross-sectional area of a single vertebral slice (at lumbar L3) in a CT scan is correlated with whole-body skeletal muscle volume. This method is used to assess CT-defined sarcopenia in patients with cancer, with low SMM effecting outcomes. However, as diagnostic scans are based on tumour location, not all include L3. We evaluated the evidence for the use of alternate vertebral CT slices for SMM evaluation when L3 is not available. Five electronic databases were searched from January 1996 to April 2020 for studies using CT scan vertebral slices above L3 for SM measurement in adults with cancer (solid tumours). Validation with whole-body SMM, rationale for the chosen slice and sarcopenia cut-off values were investigated. Thirty-two studies were included, all retrospective and cross-sectional in design. Cervical, thoracic and lumbar slices were used (from C3 to L1), with no validation of whole-body SMM using CT scans. Alternate slices were used in lung, and head and neck cancer patients. Sarcopenia cut-off values were reported in 75 % of studies, with differing methods, with or without sex-specific values, and a lack of consensus. Current evidence is inadequate to provide definitive recommendations for alternate vertebral slice use for SMM evaluation in cancer patients. Variation in sarcopenia cut-offs warrants more robust investigation, in order for risk stratification to be applied to all patients with cancer.

Type
Full Papers
Copyright
© The Author(s), 2021. Published by Cambridge University Press on behalf of The Nutrition Society

Malnutrition and involuntary weight loss are common in cancer patients and can often be attributed to the metabolic burden of the tumour itself with associated cachexia, and/or the toxicities of the treatment modalities used(Reference Von Meyenfeldt, Fredrix and Haagh1Reference Baracos, Martin and Korc3). Recently, skeletal muscle mass (SMM) depletion has gained particular interest, with the use of various body composition methods to determine actual loss of lean tissue rather than total body weight alone(Reference Prado and Heymsfield4). This is particularly relevant, as depletion of muscle mass can occur independently of adipose tissue, and overweight or obese patients may have low skeletal muscle reserves(Reference Prado, Lieffers and McCargar5). In patients with cancer, skeletal muscle and fat are not necessarily gained or lost in equal proportions and although patients may gain fat, muscle mass can be lost simultaneously(Reference Fearon, Strasser and Anker2,Reference Mourtzakis, Prado and Lieffers6) , and as a result, these patients can be overlooked for potential nutritional risk(Reference Barazzoni, Bischoff and Boirie7).

The progressive and general depletion of SMM and related functional decline are key components of sarcopenia(Reference Cruz-Jentoft and Sayer8). Full consensus on the definition of sarcopenia in terms of included variables and cut-off values has yet to be reached; however, diagnosis requires a combination of muscle mass measurement, muscle strength and physical performance indicators(Reference Cruz-Jentoft, Bahat and Bauer9). Sarcopenia has been associated with subsequent adverse outcomes, and although originally mostly investigated in the ageing population, with an increased risk of falls, frailty, functional decline and mortality(Reference Cruz-Jentoft, Bahat and Bauer9), in patients with cancer, low SMM and low muscle attenuation have been shown to be poor prognostic indicators(Reference Martin, Birdsell and MacDonald10,Reference Daly, Prado and Ryan11) . Where sarcopenia has been assessed in cancer research, it is generally defined as low muscle mass, with physical function assessment rarely included(Reference Cruz-Jentoft and Sayer8). The term ‘skeletal muscle index’ (SMI) is also used to define sarcopenia as a normalised measure of muscle (cm2/m2).

The negative impact of sarcopenia on outcomes, particularly survival, in patients with cancer, has been investigated across various malignancies such as head and neck, colorectal, upper gastrointestinal and lung(Reference Prado, Lieffers and McCargar5,Reference Martin, Birdsell and MacDonald10,Reference Shachar, Williams and Muss12Reference Prado, Birdsell and Baracos18) . As a result, there is increasing evidence that sarcopenia should be included in risk stratification tools to identify patients at high risk of potential nutritional issues and complications of treatment.

Measurement of body composition using diagnostic computed tomography (CT) scans has emerged as an effective method to determine levels of muscle depletion, in oncology patients, as images are readily available for analysis and do not require additional cost, radiation exposure or burden to the patient. The accuracy and reliability of a CT scan at the tissue-organ level has led to this being accepted as the ‘gold-standard’ imaging method in body composition analysis(Reference Prado and Heymsfield4,Reference Mourtzakis, Prado and Lieffers6,Reference Prado, Birdsell and Baracos18) . In this context, low muscle mass, without functional assessment, that is associated with an increased risk of mortality, has been described as ‘CT-defined sarcopenia’(Reference Martin, Gioulbasanis and Senesse19,Reference Martin, Hopkins and Malietzis20) . For the purposes of this review, the term sarcopenia will refer to CT-defined sarcopenia.

The cross-sectional area (CSA) of a single vertebral slice in CT imaging at the lumbar vertebral level of L3 has been shown to correlate well with total body skeletal muscle volume in healthy subjects(Reference Shen, Punyanitya and Wang21). The CSA of muscle at the level of L3 can be used to diagnose sarcopenia; however, cut-off values have been previously defined by numerous researchers based on mortality outcomes, often with varied values for sex, BMI, specific populations, and in varying tumour groups(Reference Prado, Lieffers and McCargar5,Reference Mourtzakis, Prado and Lieffers6,Reference Martin, Birdsell and MacDonald10,Reference van Vledder, Levolger and Ayez22,Reference Su, Ruan and Chen23) , making direct comparisons difficult in patients with cancer without standardisation.

One of the issues with the use of CT scans in muscle mass evaluation for many tumour types is that diagnostic, rather than whole-body scans, is frequently used to evaluate a specific anatomical area such as: the head and neck, chest or abdomen and pelvis. As a result, those who have not had an abdominal CT are unable to be assessed using validated measures. This has led researchers to use alternative landmarks.

The selection of an alternate slice to L3 should consider whole-body musculature, specifically whether muscle groups are more susceptible to wasting than others, and minimum mass required for actual function. Alternate CT scan slices have been investigated in non-cancer or healthy subjects, with vertebral levels ranging from T4 to L5, with varying results(Reference Shen, Punyanitya and Wang21,Reference Derstine, Holcombe and Ross24Reference Rozenberg, Mathur and Herridge32) . Dertstine et al., in a study of 735 healthy subjects, compared vertebral levels from T10 to T5, and the authors concluded that the further away from L3 that SMM is measured, the less reflective it is of whole-body SMM(Reference Derstine, Holcombe and Ross24). Although validation of alternate slices has been conducted in healthy subjects, this may not be applicable to patients with solid tumours, due to potential alterations in body composition caused by altered metabolic and energy balance(Reference Demark-Wahnefried, Peterson and Winer33Reference Silver, Dietrich and Murphy35).

Single muscle groups at a particular vertebral level have also been used, such as the psoas or pectoralis muscle, rather than the CSA of the entire slice, again with varying definitions of sarcopenia cut-offs (if any), and subsequent varied results(Reference Kinsey, San José Estépar and van der Velden36Reference Veld, Vossen and De Amorim Bernstein47). Baracos raises significant concerns with using one sentinel muscle for sarcopenia measurement, including that all muscles have specific functions, and wasting can occur differently in certain muscle groups, bringing into question whether one muscle can be representative of whole-body muscle status(Reference Baracos48). This approach has also yet to be validated by any expert group. Adiposity at a particular vertebral level should ideally also be considered to enable the impact of sarcopenia and obesity to be explored; however, for the purposes of this review, assessment of SMM is the focus.

With much being currently explored in this area, and the importance of identifying sarcopenia in cancer patients, the aim of this systematic review was to investigate the use of alternate vertebral levels above the lumbar L3 in the evaluation of skeletal muscle in this population. The objective of this narrative synthesis is to determine if there is a feasible alternative that can be used when L3 is not available.

Methodology

Search strategy

This systematic review followed criteria outlined by the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines(Reference Liberati, Altman and Tetzlaff49) and has been registered with PROSPERO (the international prospective register of systematic reviews) with Registration number CRD42019137317 in October 2019.

All relevant studies were identified through extensive searches of electronic databases and reference lists of eligible papers. The search included the databases: Ovid MEDLINE, Embase, CINAHL (Cumulative Index to Nursing and Allied Health Literature), Web of Science and Scopus (papers published from January 1996 to 30 April 2020). The medical subject headings (MeSH) used in each database were: (sarcopenia, ‘skeletal muscle’, ‘muscle wasting’, ‘muscle atrophy’, ‘body composition’, ‘muscle mass’, cachexia and ‘muscle weakness’) combined (using Boolean operator AND) with (CT, ‘computed tomography’) and (cancer, carcinoma, neoplasms) and (lumbar, thoracic, cervical). The systematic selection process is illustrated in Preferred Reporting Items for Systematic Reviews and Meta-Analysis format in Fig. 1.

Fig. 1. PRISMA flow diagram of literature search and article selection.

Eligibility criteria

Studies using adult human subjects (≥18 years) with a confirmed cancer diagnosis (solid tumours only) were included. All studies that used alternate vertebral landmark slices to the lumbar L3 in CT scans in the evaluation of SMM and all papers that investigated the CSA of skeletal muscle at any slice above the level of L3 were eligible. Those that included validation or comparison of the alternate slice with L3 were also included. Studies selected for review included only those that had measured skeletal muscle using the CSA of an entire vertebral slice and not isolated muscle groups. Other exclusions were conference abstracts, case studies, opinion papers, non-English papers and reviews.

Study appraisal and synthesis

Eligibility assessment was performed by the primary investigator, independently and unblinded. Electronic database search results were transferred into EndNote X9 (Thompson Reuters), where duplicates were then removed. The abstracts and titles of the remaining papers were assessed using the eligibility criteria, excluding those that did not obviously meet this. The full texts of publications that met the criteria were then reviewed once again to ensure compliance. The quality of the methodology used in each paper and validity of presented results were then assessed using the Academy of Nutrition and Dietetics Quality Criteria Checklist, where studies were given a positive (+), neutral (Ø) or negative (–) quality rating based on the appraisal tool(50). Two independent reviewers completed the checklist individually and blinded, and findings were compiled and any discordance was discussed until consensus was reached.

Data extraction

Data relating to the type of cancer, specific vertebral slice and/or slices used, the rationale for selecting a particular slice, whether association with L3 was investigated and the sarcopenia cut-off points used (if included) are displayed in Table 1.

Table 1. Characteristics of thirty-two selected studies using alternate vertebral computed tomography (CT) scan slices for skeletal muscle mass (SMM) estimation

Quality rating: + positive, Ø neutral.

C, cervical; T, thoracic; L, lumbar; HNC, head and neck cancer; SMM, skeletal muscle mass; CSMA, cross-sectional muscle area; SMA, skeletal muscle area; SMI, skeletal muscle index; CSA, cross-sectional area; CDLT, chemotherapy dose-limiting toxicity; CT, computed tomography; SAT, subcutaneous adipose tissue; SM, skeletal muscle; pts, patients; Ca, cancer; N/A, not applicable; PNS, paranasal sinuses; SMD, skeletal muscle radiodensity; DFS, disease-free survival; PFS, progression-free survival; OS, overall survival; DSS, disease-specific survival; CRF, chronic renal failure; ROC, receiver operating characteristic.

Results

There were 4576 studies identified through the database searching process. After screening titles and abstracts, fifty-three studies were scanned in full. Studies that did not meet the criteria for inclusion were then removed; reference lists of the remaining papers were checked, resulting in thirty-two included in the systematic review (Fig. 1).

Study characteristics

All thirty-two papers were retrospective, cross-sectional studies, with the largest proportion originating from The Netherlands (13/32, 41 %). The number of patient scans used in each study varied, with the majority 24/32 (75 %) having sample sizes <250, with only three studies having much larger cohorts of >800(Reference Pai, Chuang and Chuang51Reference Dabiri, Popuri and Cespedes Feliciano53). The majority (52 %) of papers focused on muscle mass assessment in patients with head and neck cancer (HNC) (seventeen studies(Reference Pai, Chuang and Chuang51,Reference Popuri, Cobzas and Esfandiari52,Reference Bril, Wendrich and Swartz54Reference Huiskamp, Chargi and Devriese68) ), with ten (32 %) investigating lung cancer(Reference Goncalves, Taylor and Halpenny69Reference Madariaga, Troschel and Best78) and the remaining papers focusing on a mixed tumour cohort of head and neck, lung, breast and colorectal(Reference Dabiri, Popuri and Cespedes Feliciano53,Reference Blauwhoff-Buskermolen, Langius and Becker79Reference Neefjes, van den Hurk and Blauwhoff-Buskermolen82) . More detailed characteristics of the studies are shown in Table 1.

Study quality and bias assessment found that there were no studies of negative quality and eighteen was rated as being neutral. As all studies were retrospective in nature and involved the use of CT scans from patients who had already had a scan for diagnostic purposes, patients were not ‘recruited’ into studies and received no intervention other than standard care. The risk of bias therefore was associated with the limiting of selected tumour groups, and as there was no randomisation and small populations, results may not be representative of larger sample cohorts. Due to the nature of CT scans, scans were excluded from all studies if they were not clear for analysis and this may have led to some bias in the assessment of the remaining scans. Potential biases were addressed in most papers; however, three failed to discuss risk of bias or limitations at all in the presented results(Reference Jung, Roh and Kim63,Reference Kim, Kim and Park70,Reference Recio-Boiles, Galeas and Goldwasser71) .

Analysis of computed tomography scans

The Slice-O-Matic (Tomovision, Canada) medical imaging software was used in the majority of studies (15/32, 47 %), with the remainder using either in-house software or different software packages. Fifteen papers used a cervical vertebral slice (C3)(Reference Bril, Wendrich and Swartz54Reference Wendrich, Swartz and Bril57,Reference Bril, Pezier and Tijink59Reference Ansari, Chargi and van Gemert66,Reference Huiskamp, Chargi and Devriese68,Reference Muresan, Juan and Artero80) with most using the study by Swartz et al. as the reference for the use of C3, and applying the prediction formula described to the CSA at C3 to predict CSA at L3(Reference Swartz, Pothen and Wegner56). Seven of these studies were conducted at the same institution in the Netherlands where the use of C3 in muscle mass estimation was first investigated(Reference Bril, Wendrich and Swartz54Reference Wendrich, Swartz and Bril57,Reference Bril, Pezier and Tijink59,Reference Ansari, Chargi and van Gemert66Reference Huiskamp, Chargi and Devriese68) . The use of thoracic vertebral landmarks varied and included T2, T4, T5, T8, T10 and T12, and these levels were used in the studies investigating lung, head and neck and colorectal cancer(Reference Pai, Chuang and Chuang51Reference Dabiri, Popuri and Cespedes Feliciano53,Reference Sealy, Dechaphunkul and van der Schans58,Reference Goncalves, Taylor and Halpenny69,Reference Fintelmann, Troschel and Mario74Reference Madariaga, Troschel and Best78,Reference van der Kroft, van Dijk and Rensen81,Reference Neefjes, van den Hurk and Blauwhoff-Buskermolen82) . The remaining studies used the higher lumbar vertebral slice of L1(Reference Goncalves, Taylor and Halpenny69Reference Sun, Anraku and Karasaki73). The most common reason given for using an alternative slice to L3 was that an abdominal scan was not available diagnostically for many tumour sites, and an alternative needed to be used for sarcopenia analysis. However, most gave no rationale as to why a specific slice level was chosen (either cervical or thoracic), or any previously published validation evidence to support its use.

Two Canadian studies investigated auto-segmentation modelling using a head and neck and lung cancer data set (1004 patients) of CT scans(Reference Popuri, Cobzas and Esfandiari52,Reference Dabiri, Popuri and Cespedes Feliciano53) . Both presented models for thoracic auto-segmentation at T4 and at L3 following comparison with manual segmentation of the same slices. Direct comparison of SMM at the thoracic level with that at L3 was not conducted. Both studies suggest ‘good’ performance from their models for auto-segmentation of muscle from CT images suitable for analysis of large-scale cancer databases. Although these two studies were quite clinically different to those measuring outcomes, they met the selection criteria for this review. Rationale for the use of the thoracic slice T4 as well as L3 was that both had been ‘widely used for body composition analysis’(Reference Popuri, Cobzas and Esfandiari52).

Sarcopenia analysis and cut-off values

Sarcopenia was not the focus of all the studies in the review, and specific sarcopenia cut-off measurements were not provided in these instances. These studies investigated the association between L3 and another slice of choice as an alternative, however, did not determine actual sarcopenia incidence in the populations used(Reference Bril, Wendrich and Swartz54,Reference Swartz, Pothen and Wegner56,Reference Wieland, Stewart and Skipworth76) . Where sarcopenia cut-off values were used, there was inconsistency in the values provided. Five studies referenced the sarcopenia cut-off value determined by Wendrich et al. (Reference Wendrich, Swartz and Bril57) of SMI < 43·2 cm2/m2 which was found to be predictive of chemotherapy dose-limiting toxicity, however provided no sex-specific values(Reference Chargi, Bril and Emmelot-Vonk55,Reference Bril, Pezier and Tijink59,Reference Zwart61,Reference Ansari, Chargi and van Gemert66,Reference Chargi, Bril and Swartz67) . Others used the sex-specific cut-offs applied for CSA at L3 by Martin et al. (Reference Martin, Birdsell and MacDonald10) which also provides cut-off values for obese males(Reference Ganju, Morse and Hoover60,Reference Muresan, Juan and Artero80,Reference Neefjes, van den Hurk and Blauwhoff-Buskermolen82) . Twelve studies provided their own cut-off values based on mortality or morbidity prediction, or using receiver operating characteristic curves in their population analysis, with or without sex-specific cut-offs, at the alternate vertebral slice levels(Reference Pai, Chuang and Chuang51,Reference Sealy, Dechaphunkul and van der Schans58,Reference Karsten, Al-Mamgani and Bril62,Reference Jung, Roh and Kim63,Reference Lin, Lin and Kang65,Reference Huiskamp, Chargi and Devriese68,Reference Kim, Kim and Park70,Reference Sun, Anraku and Karasaki73Reference Troschel, Kuklinski and Knoll75,Reference Madariaga, Troschel and Best78,Reference Blauwhoff-Buskermolen, Langius and Becker79) . Most papers provided cut-offs in comparable SMI format, others looked at volume or mass values only. All cut-off values are shown in Table 1, and those studies that provided sex-specific cut-off values are represented in Fig. 2 to graphically demonstrate the variation in cut-offs used.

Fig. 2. Sarcopenia cut-offs using alternate vertebral slices in (a) females and (b) males. Legend: T2; T4; L1; L3

Alternate vertebral slice results

All studies presented positive results for the use of the particular vertebral slice chosen as an alternative to L3. Few, however, performed direct comparative evaluation to CSA at L3(Reference Pai, Chuang and Chuang51,Reference Jung, Roh and Kim63,Reference Kim, Kim and Park70,Reference Sanders, Degens and Dingemans72,Reference Gronberg, Sjoblom and Wentzel-Larsen77,Reference van der Kroft, van Dijk and Rensen81) , and none compared the alternate slice of choice with whole-body SMM. Swartz et al. (Reference Swartz, Pothen and Wegner56) were the first to demonstrate a correlation between CSA at L3 and C3 in fifty-two patients with HNC (r = 0·891, P < 0·001) and developed a prediction equation for the estimation of skeletal muscle at L3 from C3 values (using the sum of the paravertebral and sternocleidomastoid muscles). This method has been applied in twelve of the studies in this review(Reference Chargi, Bril and Emmelot-Vonk55,Reference Wendrich, Swartz and Bril57,Reference Bril, Pezier and Tijink59Reference Karsten, Al-Mamgani and Bril62,Reference Hua, Liao and Liu64Reference Huiskamp, Chargi and Devriese68,Reference Muresan, Juan and Artero80) . Jung et al. created differing models for survival prediction using CSA at C3 and provide low SMM cut-offs in mass measures, not SMI(Reference Jung, Roh and Kim63). No studies were found that used other cervical vertebral levels.

In the studies that investigated thoracic SMM, Pai et al. (Reference Pai, Chuang and Chuang51) found a positive correlation between CSA at T2 and L3 (r = 0·63, P < 0·001) in 173 HNC patients with full-body scans (20 % of the total study cohort). Van de Kroft et al. compared CSA at T4 with L3 and showed significant correlations in patients with liver metastases from colorectal cancer and present their own sarcopenia cut-offs for L3 and T4 (r 2 = 0·60, P < 0·001)(Reference van der Kroft, van Dijk and Rensen81). Gronberg et al. also compared CSA at L3 with T4 and only found a moderate agreement when 401 scans in patients with lung cancer were investigated at both levels, in males and females (r 2 = 0·51 and 0·28)(Reference Gronberg, Sjoblom and Wentzel-Larsen77). Another study used CSA at T4 in patients with lung cancer, and results were analysed separately with authors, suggesting that they be ‘interpreted with caution’ as validation of measurements at the T4 level was currently being undertaken(Reference Neefjes, van den Hurk and Blauwhoff-Buskermolen82). Sealy et al. also looked at SMI at T4, however in HNC patients, and used a method of pooling the SMI values at L3 and T4 using standard deviations from the mean and presented their own set of sex-specific cut-off values(Reference Sealy, Dechaphunkul and van der Schans58).

The vertebral level L1 was used by two groups with L3 comparisons. Sanders et al. demonstrated a ‘stronger’ correlation with muscle CSA at L1 to L3 when compared with pectoralis muscle alone in patients with lung cancer (r = 0·90, P < 0·001)(Reference Sanders, Degens and Dingemans72). Kim et al. also compared muscle area at L1 and the pectoralis muscle to that of the area at L3, and they too demonstrated greater correlation between L1 and L3 than pectoralis muscle alone and L3 (r = 0·851 v. r = 0·447, P < 0·001)(Reference Kim, Kim and Park70). This finding was then used to generate estimated sex-specific cut-offs for sarcopenia at the level of L1.

All but one study used baseline CT scans for analysis and did not investigate muscle changes over time in alternate slices. Wieland et al. compared scans at two time intervals to assess muscle changes at T4; however, sarcopenia was not a focus of this particular paper, and muscle loss or gain was for the purposes of unrelated investigation(Reference Wieland, Stewart and Skipworth76).

Discussion

This review demonstrates that there is limited work in the area of alternate CT slices for the diagnosis of sarcopenia in patients with cancer and, what does exist, varies widely not only in the choice of slice but also in the appropriate methods of comparison, and the sarcopenia cut-off values used. Overall, the evidence is not sufficiently robust to offer a single suitable alternative to L3 for the assessment of CT-defined sarcopenia in this population.

The use of the CSA at the L3 level has been widely accepted as the site best associated with total body SMM. Since the pioneering work of Shen et al. in 2004, who compared the muscle CSA of lumbar slices in CT scans with whole-body MRI in healthy adults, total body SMM was best correlated with skeletal muscle area in a single abdominal slice about 5 cm above L4–L5 (around the L3 mark). Whole-body SMM was measured using the CSA of forty axial slices and the estimated volume measurements in between each slice(Reference Shen, Punyanitya and Wang21). Mourtzakis et al. also demonstrated whole-body correlation with L3 in patients with locally advanced or metastatic lung or colorectal cancer, when L3 CSA was compared with whole-body dual-energy X-ray absorptiometry measures(Reference Mourtzakis, Prado and Lieffers6). No study in this review attempted to determine the association between CSA at an alternate vertebral slice, with actual total body SMM.

Full-body PET-CT scans are not routinely used in many centres, mainly due to access, costs involved or Oncologist preference for patient diagnostic requirements. A common issue in each study was a lack of rationale for the particular alternate slice chosen by the research team. Random selection of a vertebral level without consideration of the musculature in that area, and their functions, calls into question the validity and applicability of an alternate level for body composition analysis.

The highest alternate vertebral level in this review used was C3. Several papers investigating SMM in HNC have used the method introduced by Swartz et al. where muscle measurement at C3 is used to predict CSA at L3(Reference Swartz, Pothen and Wegner56). This initial study, however, only investigated a small population of HNC patients (n 52) and compared these with a control group of ‘trauma’ patients with no specific reason for the selection of C3 as the cervical level of choice. The agreement between muscle values at C3 and L3 was measured using both correlation (r = 0·785, P < 0·001) and Bland–Altman methodology(Reference Bland and Altman83). ‘Reasonable’ agreement and a prediction rule were then established following multivariate analysis and involved the inclusion of a patient’s age, sex and weight. Due to the very small cohort in this study, there is serious concern regarding imprecision, as well as results not validated against full-body SMM measurements. Muscles in the C3 area include the sternocleidomastoid and paravertebral muscles, and it is unclear if these muscles high in the neck region are susceptible to severe wasting, and even suitable to be associated with whole-body SMM. These muscles are very different to those found at the L3 level, and whether these can even be interconverted is questionable. A further limitation of the use of C3 in the HNC population is tumour invasion in this area, and although the authors addressed this with the suggestion of doubling one side of the sternocleidomastoid muscles, this is not feasible in patients with bilateral neck nodal involvement. Also missing at this level is adequate adipose tissue (specifically visceral), deeming adiposity analysis impossible. Twelve studies have subsequently used the Swartz equation method in patients with HNC with varying sample sizes and results(Reference Hua, Deng and Long43,Reference Chargi, Bril and Emmelot-Vonk55,Reference Wendrich, Swartz and Bril57,Reference Bril, Pezier and Tijink59Reference Karsten, Al-Mamgani and Bril62,Reference Lin, Lin and Kang65,Reference Ansari, Chargi and van Gemert66,Reference Huiskamp, Chargi and Devriese68,Reference Muresan, Juan and Artero80,Reference Chargi, Ansari and Huiskamp84) .

Thoracic slices ranged from T2 to T10 with no clear indication of which particular slice is the most suitable, and cut-off values for sarcopenia varied with each vertebral level. The use of a thoracic slice makes clinical sense, especially in lung cancer patients, as most will have a chest CT scan at time of diagnosis. The muscles located in the thoracic region are responsible for respiration, which is itself important for function and recovery; therefore, analysis of muscle wasting in this area could be useful. Certain muscles in the L3 region extend to the thoracic level, such as the erector spinae; however, other muscles at the level of T4, for example, include the pectoralis muscle, and these have functions associated with arm and shoulder movements. This may impact on muscle volume in this area specifically in relation to manual activities, potentially more applicable to men, and these functional differences need to be considered when selecting a thoracic slice(Reference Gronberg, Sjoblom and Wentzel-Larsen77). There was only one study that used T2. This study compared SMM at T2 with L3 in 173 HNC patients with r = 0·63 (P < 0·001)(Reference Pai, Chuang and Chuang51). The r 2 value is low at 0·40, and no information is provided with regard to adjustments for co-variables. This slice was chosen ‘along an alignment of both humeral heads and the secondary thoracic vertebra (T2)’; however, no mention is made of the potential issue of skewing of the shoulders and muscles in this area, and the difficulty of identifying a slice with identical muscle visible on both sides. Important also to note is that this study was conducted in a Taiwanese population, and ethnicity should also be taken into consideration. Due to the uncertainty of results presented in this study, the use of T2 may not be suitable, however, warrants further investigation. The Patient-Generated Subjective Global Assessment tool used in the nutritional assessment of cancer patients incorporates the physical examination of the clavicle and scapula regions for signs of muscle wasting(Reference Jager-Wittenaar85); therefore, the T2 area may have some validity for future use in sarcopenia analysis. Four studies investigated lower thoracic slices from T5 to 12, yet all provided skeletal muscle measurements in m2 or as volume in m3, not as SMI, making comparisons with other studies difficult(Reference Goncalves, Taylor and Halpenny69,Reference Fintelmann, Troschel and Mario74,Reference Troschel, Kuklinski and Knoll75,Reference Madariaga, Troschel and Best78) .

The lumbar L1 slice was used by five studies(Reference Goncalves, Taylor and Halpenny69Reference Sun, Anraku and Karasaki73) with two assessing the relationship between SMI at L3 and L1, both demonstrating reasonable correlation. The muscles found at L1 are similar to those at L3, so this is likely a suitable alternative which, however, may be too low down in a CT scan for many other cancer sites to be applicable for wider use.

Sarcopenia incidence or diagnosis was the focus of 24/32 (75 %) of the papers in this review, and there was heterogeneity in the SMI values used as the cut-off for diagnosis of sarcopenia. Sex-specific cut-offs associated with mortality have been published by Prado et al. and classify sarcopenia as <52·4 cm2/m2 for men and <38·5 cm2/m2 for women at L3, in obese patients with solid respiratory or gastrointestinal tumours(Reference Prado, Liefers and McCargar86). Martin et al. have established sarcopenia cut-off values at L3 for mortality as: <53 cm2/m2 in obese males, <43 cm2/m2 in non-obese males and <41 cm2/m2 in females regardless of obesity status, in 1473 patients with lung or gastrointestinal cancers(Reference Martin, Birdsell and MacDonald10). The studies in this review have used varied methods for low SMI assessment based on morbidity and mortality outcomes, and this makes comparability difficult, not only with specific vertebral levels but also on a tumour site level. Of concern is the lack of sex-specific threshold values for low SMM. Males generally have a higher proportion of SMM than females, and this is especially so in the upper body(Reference Janssen, Heymsfield and Wang87,Reference Baracos and Kazemi-Bajestani88) . Therefore, alternate levels in the thoracic region need to account for this potential difference in muscularity. Van de Kroft et al. (Reference van der Kroft, van Dijk and Rensen81) provided sex-specific cut-off values for both L3 and T4 with quite varied results to those given by Sealy et al. at the same levels(Reference Sealy, Dechaphunkul and van der Schans58), especially at L3, in colorectal and HNC patients, respectively. In the papers that used C3, only three(Reference Hua, Deng and Long43,Reference Ganju, Morse and Hoover60,Reference Muresan, Juan and Artero80) gave sex-specific values based on previously defined cut-off values(Reference Martin, Birdsell and MacDonald10). In the study by Wendrich et al., sex was not shown to be a significant contributing variable when estimating risk of chemotherapy dose-limiting toxicities and low SMM in a HNC population(Reference Wendrich, Swartz and Bril57). The concern with this is that four other papers have used this outcome-specific sarcopenia cut-off value to apply to unrelated outcomes such as overall survival and post-operative complications(Reference Chargi, Bril and Emmelot-Vonk55,Reference Bril, Pezier and Tijink59,Reference Zwart61,Reference Ansari, Chargi and van Gemert66) . Separate thresholds should be applied for males and females in any cohort when investigating sarcopenia.

Similarly, there is a difference in SMM with ethnicity. Patients of Asian origin have been shown to have reduced SMM when compared with Europeans(Reference Wells89), and sarcopenia seems to be less prevalent in not only the Asian population but also in African-Americans when compared with Caucasians(Reference Hilmi, Jouinot and Burns90). This is an important factor to consider, as sarcopenia cut-off values may not be optimal across differing populations and should only be used where applicable. The majority of papers in this review were European in origin; however, all but one study that included an Asian population used their own established cut-off values. Hua et al. used cut-off values by Martin et al. (Reference Martin, Birdsell and MacDonald10), which includes BMI ranges that are not necessarily suitable for this Chinese population(Reference Hua, Deng and Long43).

Cut-off values for sarcopenia diagnosis are yet to be established for all patients with cancer and have not been validated in vertebral landmarks other than L3, yet the papers in this review have established their own values without direct comparison with full-body SMM. Halpenny et al. compared six vertebral and regional measurements of SMM, including T10 and L3, in whole-body PET-CT images in 148 patients with advanced melanoma and found that skeletal muscle at T10 was strongly associated with whole body (r 2 = 0·78); however, this paper was ineligible for inclusion in this review as it did not meet eligibility criteria for patient age (range 15–91 years)(Reference Halpenny, Goncalves and Schwitzer91). Including younger subjects under the age of 18 years is problematic as there may be changes in muscle mass with growth. T10 is also too low to be included in a head and neck scan, and this level may only have applicability to patients who have had a chest CT. Association of muscle mass to vertebral slices other than L3 has yet to be validated against full-body measurements using CT scans in adult cancer patients.

The lack of consensus and varying results not only for slices used but also tumour sites makes comparison challenging. In addition, the lack of validation methods investigating the relationship of muscle CSA at other vertebral slices to whole-body SMM, specifically in cancer, is a concern and brings into question their application in sarcopenia diagnosis. The muscle CSA at the level of L3 is a surrogate of whole-body SMM in healthy adults(Reference Shen, Punyanitya and Wang21), and determining the association of muscle CSA at a different vertebral level by comparing with measures at L3 is a further surrogate of a surrogate value, increasing the possible degree of error. The myriad of results, many with poor correlations to L3, a lack of adequate consideration of musculature at alternate vertebral levels, and potentially unsuitable cut-offs for sarcopenia assessment, give cause for concern with the research that has been done in this area and should be interpreted with caution.

With emerging evidence of worse outcomes for patients with cancer who are sarcopenic, it is important that all patients are assessed where possible and that a consistent and clinically relevant evidence base be established. Having an accurate and validated alternative to the L3 slice will enable this crucial assessment in not only patients who have had an abdominal CT scan but also all patients with cancer. This review highlights the requisite need for more robust methods of validation in order for suitable alternate slices to be used for sarcopenia assessment and should ideally aim to include comparison of whole-body SMM measurements from the CT scans of patients with cancer.

Conclusion

This systematic review indicates that the current level of evidence is inadequate to provide definitive recommendations for the use of alternate vertebral slices to L3 in CT scans of cancer patients, for the evaluation of SMM and the diagnosis of CT-defined sarcopenia.

Acknowledgements

Support was provided for this work by an Australian Government Research Training Program Scholarship.

All authors contributed to the conceptualizatiuon and methodology. Belinda Vangelov: Investigation, Validation, Formal Analysis, Writing-Original draft preparation, Writing - Review and Editing, Visualisation, Project Administration. Damian Kotevski: Validation, Formal Analysis, Writing - Review and Editing. Judy Bauer: Supervision, Writing - Review and Editing. Robert Smee: Supervision, Writing - Review and Editing.

B. V., J. B., D. K. and R. S. declare they have no conflict of interest.

References

Von Meyenfeldt, MF, Fredrix, EWHM, Haagh, WAJJM, et al. (1988) The aetiology and management of weight loss and malnutrition in cancer patients. Baillière’s Clin Gastroenterol 2, 869885.CrossRefGoogle ScholarPubMed
Fearon, K, Strasser, F, Anker, SD, et al. (2011) Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 12, 489495.CrossRefGoogle Scholar
Baracos, VE, Martin, L, Korc, M, et al. (2018) Cancer-associated cachexia. Nat Rev Dis Primers 4, 17105.CrossRefGoogle ScholarPubMed
Prado, CMM & Heymsfield, SB (2014) Lean tissue imaging: a new era for nutritional assessment and intervention. J Parenter Enteral Nutr 38, 940953.CrossRefGoogle ScholarPubMed
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
Mourtzakis, M, Prado, CMM, 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 Metabol 33, 9971006.CrossRefGoogle ScholarPubMed
Barazzoni, R, Bischoff, SC, Boirie, Y, et al. (2018) Sarcopenic obesity: time to meet the challenge. Clin Nutr 37, 17871793.CrossRefGoogle ScholarPubMed
Cruz-Jentoft, AJ & Sayer, AA (2019) Sarcopenia. Lancet 393, 26362646.CrossRefGoogle ScholarPubMed
Cruz-Jentoft, AJ, Bahat, G, Bauer, J, et al. (2018) Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing 48, 1631.CrossRefGoogle Scholar
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.CrossRefGoogle ScholarPubMed
Daly, LE, Prado, CM & Ryan, AM (2018) 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. Proc Nutr Soc 77, 135151.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Tan, BHL, 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.CrossRefGoogle ScholarPubMed
Miller, BS, Ignatoski, KM, Daignault, S, et al. (2012) Worsening central sarcopenia and increasing intra-abdominal fat correlate with decreased survival in patients with adrenocortical carcinoma. World J Surg 36, 15091516.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Collins, J, Noble, S, Chester, J, et al. (2014) The assessment and impact of sarcopenia in lung cancer: a systematic review. Br Med J 4, 118.Google Scholar
Mintziras, I, Miligkos, M, Wächter, S, et al. (2018) Sarcopenia and sarcopenic obesity are significantly associated with poorer overall survival in patients with pancreatic cancer: systematic review and meta-analysis. Int J Surg 59, 1926.CrossRefGoogle ScholarPubMed
Prado, CMM, Birdsell, LA & Baracos, VE (2009) The emerging role of computerized tomography in assessing cancer cachexia. Curr Opin Support Palliat Care 3, 269275.CrossRefGoogle ScholarPubMed
Martin, L, Gioulbasanis, I, Senesse, P, et al. (2020) Cancer-associated malnutrition and CT-defined sarcopenia and myosteatosis are endemic in overweight and obese patients. J Parenter Enteral Nutr 44, 227238.CrossRefGoogle ScholarPubMed
Martin, L, Hopkins, J, Malietzis, G, et al. (2018) Assessment of computed tomography (CT)-defined muscle and adipose tissue features in relation to short-term outcomes after elective surgery for colorectal cancer: a multicenter approach. Ann Surg Oncol 25, 26692680.CrossRefGoogle ScholarPubMed
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 97, 23332338.CrossRefGoogle ScholarPubMed
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
Su, H, Ruan, J, Chen, T, et al. (2019) CT-assessed sarcopenia is a predictive factor for both long-term and short-term outcomes in gastrointestinal oncology patients: a systematic review and meta-analysis. Cancer Imaging 19, 82.CrossRefGoogle ScholarPubMed
Derstine, BA, Holcombe, SA, Ross, BE, et al. (2018) Skeletal muscle cutoff values for sarcopenia diagnosis using T10 to L5 measurements in a healthy US population. Sci Rep 8, 18.CrossRefGoogle Scholar
Mishra, A, Bigam, KD, Extermann, M, et al. (2020) Sarcopenia and low muscle radiodensity associate with impaired FEV1 in allogeneic haematopoietic stem cell transplant recipients. J Cachexia Sarcopenia Muscle 11, 15701579.CrossRefGoogle ScholarPubMed
Mathur, S, Rodrigues, N, Mendes, P, et al. (2017) Computed tomography–derived thoracic muscle size as an indicator of sarcopenia in people with advanced lung disease. Cardiopulm Phys Ther J 28, 99105.CrossRefGoogle Scholar
Moon, SW, Choi, JS, Lee, SH, et al. (2019) Thoracic skeletal muscle quantification: low muscle mass is related with worse prognosis in idiopathic pulmonary fibrosis patients. Respir Res 20, 3539.CrossRefGoogle ScholarPubMed
Tanimura, K, Sato, S, Fuseya, Y, et al. (2016) Quantitative assessment of erector spinae muscles in patients with chronic obstructive pulmonary disease: novel chest computed tomography-derived index for prognosis. Ann Am Thorac Soc 13, 334341.CrossRefGoogle ScholarPubMed
Cho, YH, Do, KH, Chae, EJ, et al. (2019) Association of chest CT-based quantitative measures of muscle and fat with post-lung transplant survival and morbidity: a single institutional retrospective cohort study in Korean population. Korean J Radiol 20, 522.CrossRefGoogle ScholarPubMed
Panthofer, AM, Olson, SL, Harris, DG, et al. (2019) Derivation and validation of thoracic sarcopenia assessment in patients undergoing thoracic endovascular aortic repair. J Vasc Surg 69, 13791386.CrossRefGoogle ScholarPubMed
Nemec, U, Heidinger, B, Sokas, C, et al. (2017) Diagnosing sarcopenia on thoracic computed tomography: quantitative assessment of skeletal muscle mass in patients undergoing transcatheter aortic valve replacement. Acad Radiol 24, 11541161.CrossRefGoogle ScholarPubMed
Rozenberg, D, Mathur, S, Herridge, M, et al. (2017) Thoracic muscle cross-sectional area is associated with hospital length of stay post lung transplantation: a retrospective cohort study. Transpl Int 30, 713724.CrossRefGoogle ScholarPubMed
Demark-Wahnefried, W, Peterson, BL, Winer, EP, et al. (2001) Changes in weight, body composition, and factors influencing energy balance among premenopausal breast cancer patients receiving adjuvant chemotherapy. J Clin Oncol 19, 23812389.CrossRefGoogle ScholarPubMed
Fouladiun, M, Körner, U, Bosaeus, I, et al. (2005) Body composition and time course changes in regional distribution of fat and lean tissue in unselected cancer patients on palliative care—Correlations with food intake, metabolism, exercise capacity, and hormones. Cancer 103, 21892198.CrossRefGoogle ScholarPubMed
Silver, HJ, Dietrich, MS & Murphy, BA (2007) Changes in body mass, energy balance, physical function, and inflammatory state in patients with locally advanced head and neck cancer treated with concurrent chemoradiation after low-dose induction chemotherapy. Head Neck 29, 893900.CrossRefGoogle Scholar
Kinsey, CM, San José Estépar, R, van der Velden, J, et al. (2017) Lower pectoralis muscle area is associated with a worse overall survival in non–small cell lung cancer. Cancer Epidemiol Biomarkers Prev 26, 3843.CrossRefGoogle ScholarPubMed
Go, S, Park, M, Song, H, et al. (2016) Sarcopenia and inflammation are independent predictors of survival in male patients newly diagnosed with small cell lung cancer. Support Care Cancer 24, 20752084.CrossRefGoogle ScholarPubMed
Takamori, S, Toyokawa, G, Okamoto, T, et al. (2018) Clinical impact and risk factors for skeletal muscle loss after complete resection of early non-small cell lung cancer. Ann Surg Oncol 25, 12291236.CrossRefGoogle ScholarPubMed
Miller, J, Harris, K, Roche, C, et al. (2018) Sarcopenia is a predictor of outcomes after lobectomy. J Thorac Dis 10, 432440.CrossRefGoogle ScholarPubMed
Singal, AG, Zhang, P, Waljee, AK, et al. (2016) Body composition features predict overall survival in patients with hepatocellular carcinoma. Clin Transl Gastroenterol 7, e172e.CrossRefGoogle ScholarPubMed
Kim, JS, Kim, WY, Park, HK, et al. (2017) Simple age specific cutoff value for sarcopenia evaluated by computed tomography. Ann Nutr Metab 71, 157163.CrossRefGoogle ScholarPubMed
Takamori, S, Tagawa, T, Toyokawa, G, et al. (2020) Prognostic impact of postoperative skeletal muscle decrease in non-small cell lung cancer. Ann Thorac Surg 109, 914920.CrossRefGoogle ScholarPubMed
Hua, X, Deng, JP, Long, ZQ, et al. (2020) Prognostic significance of the skeletal muscle index and an inflammation biomarker in patients with breast cancer who underwent postoperative adjuvant radiotherapy. Curr Prob Cancer 44, 100513.CrossRefGoogle Scholar
Wallace, JDMD, Calvo, RYP, Lewis, PRDO, et al. (2017) Sarcopenia as a predictor of mortality in elderly blunt trauma patients: comparing the masseter to the psoas using computed tomography. J Trauma Acute Care Surg 82, 6572.CrossRefGoogle Scholar
Teigen, LMRD, John, RMD, Kuchnia, AJPRD, et al. (2017) Preoperative pectoralis muscle quantity and attenuation by computed tomography are novel and powerful predictors of mortality after left ventricular assist device implantation. Circ Heart Fail 10, e004069.CrossRefGoogle ScholarPubMed
Komatsu, R, Okazaki, T, Ebihara, S, et al. (2018) Aspiration pneumonia induces muscle atrophy in the respiratory, skeletal, and swallowing systems. J Cachexia Sarcopenia Muscle 9, 643653.CrossRefGoogle ScholarPubMed
Veld, J, Vossen, JA, De Amorim Bernstein, K, et al. (2016) Adipose tissue and muscle attenuation as novel biomarkers predicting mortality in patients with extremity sarcomas. Eur Radiol 26, 46494655.CrossRefGoogle ScholarPubMed
Baracos, VE (2017) Psoas as a sentinel muscle for sarcopenia: a flawed premise. J Cachexia Sarcopenia Muscle 8, 527528.CrossRefGoogle ScholarPubMed
Liberati, A, Altman, DG, Tetzlaff, J, et al. (2009) The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. Br Med J 339.CrossRefGoogle Scholar
Academy of Nutrition and Dietetics (2016) Evidence analysis manual: Steps in the Academy evidence analysis process. Chicago, IL. https://www.andeal.org/evidence-analysis-manual (accessed June 2019).Google Scholar
Pai, PC, Chuang, CC, Chuang, WC, et al. (2018) Pretreatment subcutaneous adipose tissue predicts the outcomes of patients with head and neck cancer receiving definitive radiation and chemoradiation in Taiwan. Cancer Med 7, 16301641.CrossRefGoogle ScholarPubMed
Popuri, K, Cobzas, D, Esfandiari, N, et al. (2016) Body composition assessment in axial CT images using FEM-based automatic segmentation of skeletal muscle. IEE Transact Med Imaging 35, 512520.CrossRefGoogle ScholarPubMed
Dabiri, S, Popuri, K, Cespedes Feliciano, EM, et al. (2019) Muscle segmentation in axial computed tomography (CT) images at the lumbar (L3) and thoracic (T4) levels for body composition analysis. Comput Med Imaging Graph 75, 4755.CrossRefGoogle ScholarPubMed
Bril, SI, Wendrich, AW, Swartz, JE, et al. (2019) Interobserver agreement of skeletal muscle mass measurement on head and neck CT imaging at the level of the third cervical vertebra. Eur Arch Otorhinolaryngol 276, 11751182.CrossRefGoogle ScholarPubMed
Chargi, N, Bril, SI, Emmelot-Vonk, MH, et al. (2019) Sarcopenia is a prognostic factor for overall survival in elderly patients with head-and-neck cancer. Eur Arch Otorhinolaryngol 276, 14751486.CrossRefGoogle ScholarPubMed
Swartz, JE, Pothen, AJ, Wegner, I, et al. (2016) Feasibility of using head and neck CT imaging to assess skeletal muscle mass in head and neck cancer patients. Oral Oncol 62, 2833.CrossRefGoogle ScholarPubMed
Wendrich, AW, Swartz, JE, Bril, SI, et al. (2017) Low skeletal muscle mass is a predictive factor for chemotherapy dose-limiting toxicity in patients with locally advanced head and neck cancer. Oral Oncol 71, 2633.CrossRefGoogle ScholarPubMed
Sealy, MJ, Dechaphunkul, T, van der Schans, CP, et al. (2020) Low muscle mass is associated with early termination of chemotherapy related to toxicity in patients with head and neck cancer. Clin Nutr 39, 501519.CrossRefGoogle ScholarPubMed
Bril, SI, Pezier, TF, Tijink, BM, et al. (2019) Preoperative low skeletal muscle mass as a risk factor for pharyngocutaneous fistula and decreased overall survival in patients undergoing total laryngectomy. Head Neck 41, 17451755.CrossRefGoogle ScholarPubMed
Ganju, R, Morse, R, Hoover, A, et al. (2019) The impact of sarcopenia on tolerance of radiation and outcome in patients with head and neck cancer receiving chemoradiation. Radiother Oncol 137, 117124.CrossRefGoogle ScholarPubMed
Zwart, AT (2019) Anouk van der H, Peter MAvO, Roel JHMS, de Bock GH, Halmos GB. CT-measured skeletal muscle mass used to assess frailty in patients with head and neck cancer. J Cachexia Sarcopenia Muscle 10, 10601069.CrossRefGoogle Scholar
Karsten, RT, Al-Mamgani, A, Bril, SI, et al. (2019) Sarcopenia, a strong determinant for prolonged feeding tube dependency after chemoradiotherapy for head and neck cancer. Head Neck 41, 40004008.CrossRefGoogle Scholar
Jung, AR, Roh, J-L, Kim, JS, et al. (2019) Efficacy of head and neck computed tomography for skeletal muscle mass estimation in patients with head and neck cancer. Oral Oncol 95, 9599.CrossRefGoogle ScholarPubMed
Hua, X, Liao, JF, Liu, S, et al. (2020) Low skeletal muscle mass impairs quality of life in nasopharyngeal carcinoma patients treated with concurrent chemoradiotherapy. Front Nutr 6, 195.CrossRefGoogle ScholarPubMed
Lin, SC, Lin, YS, Kang, BH, et al. (2020) Sarcopenia results in poor survival rates in oral cavity cancer patients. Clin Otolaryngol 45, 327333.CrossRefGoogle ScholarPubMed
Ansari, E, Chargi, N, van Gemert, JTM, et al. (2020) Low skeletal muscle mass is a strong predictive factor for surgical complications and a prognostic factor in oral cancer patients undergoing mandibular reconstruction with a free fibula flap. Oral Oncol 101, 104530.CrossRefGoogle Scholar
Chargi, N, Bril, SI, Swartz, JE, et al. (2020) Skeletal muscle mass is an imaging biomarker for decreased survival in patients with oropharyngeal squamous cell carcinoma. Oral Oncol 101, 104519.CrossRefGoogle ScholarPubMed
Huiskamp, LFJ, Chargi, N, Devriese, LA, et al. (2020) The predictive and prognostic value of low skeletal muscle mass for dose-limiting toxicity and survival in head and neck cancer patients receiving concomitant cetuximab and radiotherapy. Eur Arch Otorhinolaryngol 277, 28472858.CrossRefGoogle ScholarPubMed
Goncalves, MD, Taylor, S, Halpenny, DF, et al. (2018) Imaging skeletal muscle volume, density, and FDG uptake before and after induction therapy for non-small cell lung cancer. Clin Radiol 73, 505e1.CrossRefGoogle ScholarPubMed
Kim, EY, Kim, YS, Park, I, et al. (2016) Evaluation of sarcopenia in small-cell lung cancer patients by routine chest CT. Support Care Cancer 24, 47214726.CrossRefGoogle ScholarPubMed
Recio-Boiles, A, Galeas, JN, Goldwasser, B, et al. (2018) Enhancing evaluation of sarcopenia in patients with non-small cell lung cancer (NSCLC) by assessing skeletal muscle index (SMI) at the first lumbar (L1) level on routine chest computed tomography (CT). Support Care Cancer 26, 23532359.CrossRefGoogle Scholar
Sanders, KJC, Degens, JHRJ, Dingemans, AMC, et al. (2019) Cross-sectional and longitudinal assessment of muscle from regular chest computed tomography scans: L1 and pectoralis muscle compared to L3 as reference in non-small cell lung cancer. Int J Chron Obstruct Pulmon Dis 14, 781789.CrossRefGoogle ScholarPubMed
Sun, C, Anraku, M, Karasaki, T, et al. (2019) Low truncal muscle area on chest computed tomography: a poor prognostic factor for the cure of early-stage non-small-cell lung cancer. E J Cardiothorac Surg 55, 414420.CrossRefGoogle Scholar
Fintelmann, FJ, Troschel, FM, Mario, J, et al. (2018) Thoracic skeletal muscle is associated with adverse outcomes after lobectomy for lung cancer. Ann Thorac Surg 105, 15071515.CrossRefGoogle ScholarPubMed
Troschel, FM, Kuklinski, MW, Knoll, SJ, et al. (2019) Preoperative thoracic muscle area on computed tomography predicts long-term survival following pneumonectomy for lung cancer. Interact Cardiovasc Thorac Surg 28, 542549.CrossRefGoogle ScholarPubMed
Wieland, BM, Stewart, GD, Skipworth, RJE, et al. (2007) Is there a human homologue to the murine proteolysis-inducing factor? Clin Cancer Res 13, 49844992.CrossRefGoogle Scholar
Gronberg, BH, Sjoblom, B, Wentzel-Larsen, T, et al. (2018) A comparison of CT based measures of skeletal muscle mass and density from the Th4 and L3 levels in patients with advanced non-small-cell lung cancer. Eur J Clin Nutr 73, 10691076.CrossRefGoogle ScholarPubMed
Madariaga, MLL, Troschel, FM, Best, TD, et al. (2020) Low thoracic skeletal muscle area predicts morbidity after pneumonectomy for lung cancer. Ann Thorac Surg 109, 907913.CrossRefGoogle ScholarPubMed
Blauwhoff-Buskermolen, S, Langius, JAE, Becker, A, et al. (2017) The influence of different muscle mass measurements on the diagnosis of cancer cachexia. J Cachexia Sarcopenia Muscle 8, 615622.CrossRefGoogle Scholar
Muresan, BT, Juan, CS, Artero, A, et al. (2019) Diagnosis of pre-sarcopenia from a single selectional crosscut at c3 region, using ct scans before radiotherapy. Nutr Hosp 36, 11011108.Google ScholarPubMed
van der Kroft, G, van Dijk, DPJ, Rensen, SS, et al. (2019) Low thoracic muscle radiation attenuation is associated with postoperative pneumonia following partial hepatectomy for colorectal metastasis. HPB (In Press).CrossRefGoogle Scholar
Neefjes, ECW, van den Hurk, RM, Blauwhoff-Buskermolen, S, et al. (2017) Muscle mass as a target to reduce fatigue in patients with advanced cancer. J Cachexia Sarcopenia Muscle 8, 623629.CrossRefGoogle ScholarPubMed
Bland, JM & Altman, DG (2010) Statistical methods for assessing agreement between two methods of clinical measurement. Int J Nurs Studies 47, 931936.CrossRefGoogle Scholar
Chargi, N, Ansari, E, Huiskamp, LFJ, et al. (2019) Agreement between skeletal muscle mass measurements using computed tomography imaging and magnetic resonance imaging in head and neck cancer patients. Oral Oncol 99, 104341.CrossRefGoogle ScholarPubMed
Jager-Wittenaar, Hab (2017) Ottery FDac. Assessing nutritional status in cancer: role of the Patient-Generated Subjective Global Assessment. Curr Opin Clin Nutr Metab Care 20, 322329.CrossRefGoogle Scholar
Prado, CMM, Liefers, 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
Janssen, I, Heymsfield, SB, Wang, Z, et al. (2000) Skeletal muscle mass and distribution in 468 men and women aged 18–88 year. J Appl Physiol 89, 8188.CrossRefGoogle Scholar
Baracos, V & Kazemi-Bajestani, SMR (2013) Clinical outcomes related to muscle mass in humans with cancer and catabolic illnesses. Int J Biochem Cell Biol 45, 23022308.CrossRefGoogle ScholarPubMed
Wells, JCK (2012) Ethnic variability in adiposity, thrifty phenotypes and cardiometabolic risk: addressing the full range of ethnicity, including those of mixed ethnicity. Obes Rev 13, 1429.CrossRefGoogle ScholarPubMed
Hilmi, M, Jouinot, A, Burns, R, et al. (2019) Body composition and sarcopenia: the next-generation of personalized oncology and pharmacology? Pharmacol Ther 196, 135159.CrossRefGoogle ScholarPubMed
Halpenny, DF, Goncalves, M, Schwitzer, E, et al. (2018) Computed tomography-derived assessments of regional muscle volume: validating their use as predictors of whole body muscle volume in cancer patients. Br J Radiol 91, 20180451.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. PRISMA flow diagram of literature search and article selection.

Figure 1

Table 1. Characteristics of thirty-two selected studies using alternate vertebral computed tomography (CT) scan slices for skeletal muscle mass (SMM) estimation

Figure 2

Fig. 2. Sarcopenia cut-offs using alternate vertebral slices in (a) females and (b) males. Legend: T2; T4; L1; L3