SciELO - Scientific Electronic Library Online

 
vol.35 número2Hipersensibilidade a Meios de Contraste Derivados do Gadolínio: Caracterização Retrospectiva de uma Coorte PortuguesaSegurança na Neuroimagem em Idade Pediátrica índice de autoresíndice de assuntosPesquisa de artigos
Home Pagelista alfabética de periódicos  

Serviços Personalizados

Journal

Artigo

Indicadores

Links relacionados

  • Não possue artigos similaresSimilares em SciELO

Compartilhar


Acta Radiológica Portuguesa

versão impressa ISSN 2183-1351

Acta Radiol Port vol.35 no.2 Lisboa ago. 2023  Epub 31-Ago-2023

https://doi.org/10.25748/arp.29823 

Artigos Originais

Sarcopenia’s Prognostic Impact in Gastric Cancer Patients Undergoing Surgery

O Impacto Prognóstico da Sarcopenia em Doentes com Neoplasia Gástrica Submetidos a Cirurgia

1Department of Radiology, Hospital de Braga, Braga, Portugal

2Faculty of Medicine, Universidade of Minho, Braga, Portugal

3Department of General Surgery, Hospital de Braga, Braga, Portugal


Abstract

Introduction:

Surgical resection is the first-line treatment for patients with resectable gastric cancer. The impact of preoperative sarcopenia on the post-operative outcomes of these patients remains unclear. For this reason, we studied the association between sarcopenia and its prognostic value for gastric cancer patients undergoing surgery.

Methods:

A retrospective, observational, longitudinal, and descriptive analysis was conducted. A sample of 147 patients, who underwent gastrectomy at the Hospital of Braga between January 2008 and December 2016, was considered. Sarcopenia was diagnosed using ImageJ® software by measuring total abdominal area through axial images of preoperative computed tomography scans. The Prado et al. cut-off for sarcopenia was used. Statistical analysis was performed using SPSS®-26.0 version. The Overall Survival (OS) of these patients was assessed using Kaplan-Meier method. Multivariate Cox Regression was performed to evaluate independent risk factors for OS.

Results:

Thirty-two (21.8%) of the evaluated patients had sarcopenia. Sarcopenia was significantly more frequently diagnosed in males (p=0.040); yet no significant relation was established with patients' age (p=0.114). Sarcopenia influenced the presence of postoperative complications (p=0.01), length of hospital stay (p<0.001) and OS (p=0.027). However, it did not prove to be an independent prognostic factor for worse OS, unlike age and TNM stage which were found to be independent risk factors for OS.

Conclusion:

Our results demonstrate that sarcopenia influences incidence and severity of postoperative complications, length of hospital stay and OS of patients with gastric cancer who underwent surgery.

Keywords: Gastric cancer; Overall survival; Prognosis; Sarcopenia; Surgery.

Resumo

Introdução:

O tratamento cirúrgico é a primeira linha para doentes com neoplasia gástrica ressecável. O impacto da presença de sarcopenia pré-operatoriamente nos resultados pós-cirúrgicos destes doentes permanece por esclarecer. Neste contexto, avaliou-se a relação entre a presença de sarcopenia e o prognóstico de doentes submetidos a cirurgia por cancro gástrico.

Métodos:

Realizou-se um estudo retrospetivo, observacional, descritivo, analítico e longitudinal. Foi selecionada uma amostra de 147 doentes submetidos a gastrectomia no Hospital de Braga entre Janeiro/2008 e Dezembro/2016. O diagnóstico de sarcopenia foi estabelecido com o software ImageJ®, pela medição da área abdominal total através de imagens axiais de tomografias computorizadas pré-operatórias. Foi utilizado o cut-off de Prado et al. A análise estatística dos dados foi feita com recurso ao software SPSS® 26.0. A sobrevida global destes doentes foi avaliada pelo método Kaplan-Meier. A regressão de Cox Multivariada foi utilizada para encontrar fatores de risco para a sobrevida global.

Resultados:

Trinta e dois (21.8%) dos doentes avaliados apresentavam sarcopenia, que foi mais frequentemente diagnosticada no sexo masculino (p=0.040), no entanto, não foi estabelecida relação com a idade (p=0.114). Mostrou influenciar a ocorrência (p=0.01) de complicações pós-operatórias, duração do internamento (p<0.001) e sobrevida global (p=0.027). Contudo, não provou ser fator prognóstico independente para pior sobrevida global, ao contrário da idade e estadiamento TNM, que mostraram ser fatores de risco independentes para a sobrevida global.

Conclusão:

Neste estudo, a sarcopenia mostrou influenciar a incidência e severidade das complicações pós-operatórias, a duração do internamento e a sobrevida global dos doentes submetidos a cirurgia por cancro Gástrico.

Palavras-Chave: Cancro gástrico; Cirurgia; Prognóstico; Sarcopenia; Sobrevida global.

Introduction

According to GLOBOCAN 2020, gastric cancer (GC) is the fifth most frequently diagnosed neoplastic condition worldwide, being the fourth most lethal,1 with a higher prevalence in the male gender.2 Its incidence exceeded one million new cases along 2020, with an estimated absolute mortality of 769.000 deaths,1 which is thought to be related to its delayed diagnosis.

One of the major prognostic factors for GC concerns disease’s stage at the time of diagnosis/surgical intervention, as deeper neoplastic invasion of the gastric wall is associated with worse outcomes.3 Initial GC staging requires both thoracic and abdominopelvic computed tomography (CT) scan and, in patients with suspected distant metastatic involvement, staging laparoscopy with concomitant cytological analysis of the peritoneal fluid is appropriate.4,5

Surgical resection is the first-line treatment for resectable GC. Nevertheless, in patients with locally advanced disease, it has been shown that perioperative treatment improves survival rates amongst patients with resectable GC when compared to surgery alone.6

In 2010, The European Working Group on Sarcopenia in Older People (EWGSOP) defined sarcopenia as a progressive syndrome characterized by general loss of skeletal muscle mass and overall strength, which may lead to adverse outcomes such as physical disability, worse quality of life and, ultimately, death.7 The EWGSOP states that both reduced muscle mass as well as decreased muscle function (strength or performance), are enough to diagnose sarcopenia.7

In 2018, the EWGSOP group assembled again (EWGSOP2) setting out a new definition for sarcopenia, modifying its diagnosis criteria. In this revised definition, sarcopenia is no longer solely connected with older individuals, and is now known to begin earlier in life.8 Reduced muscular strength is the primary criterion for sarcopenia, apart from being the most accurate indicator of muscle function nowadays. Sarcopenia is diagnosed if, besides reduced muscle strength, there is also decreased muscle quantity or quality.8

Currently, there is a wide array of techniques available for the diagnosis of sarcopenia, ranging from anthropometric and bioimpedance analyses to imaging methods.

With regards to fat volume and lean body mass calculation, bioimpedance analysis (BIA) constitutes a useful tool, as it provides an estimate of muscle mass based on whole-body electrical conductivity.8

On the other hand, anthropometric measurements (AM) are a valuable tool for the initial assessment of sarcopenia in children and young adults, specially in primary care settings where alternative techniques are often unavailable. However, AM value in the elderly is uncertain as progressive changes in fat deposits and skin flexibility provoked by aging, contributes to inaccuracies in body composition estimation, rendering it as an inaccurate method for older and obese patients.9

Finally, both CT, Magnetic Resonance Imaging (MRI) and Dual-energy X-ray Absorptiometry (DXA) are imaging methods that have been used to measure muscular mass.

Photon transmission at two different energy levels is the basis of DXA, resulting in differential attenuation of bone, fat and lean tissue. Predicated in this assumption, various attenuations visualized in tissues, at different X-ray energies, enable several measurements such as lean body mass (LBM), fat mass (FM) and bone mineral mass (BMM).10 The results are later extrapolated to the whole body.11

Both MRI and CT are regarded as gold-standards for non-invasive muscle quantity/mass evaluation.8 Even though the theoretical basis for MRI and CT measurements is the same, MRI has the major advantage of allowing body composition analysis without radiation exposure.11 In these imaging methods, the third lumbar vertebra (L3) landmark is often used in cross-sectional body composition analysis, and it is found to correlate with whole-body tissue measurements.12,13Both the psoas, paraspinal muscles (erector spinae, quadratus lumborum) and abdominal wall muscles (external and internal obliques, transversus abdominus, rectus abdominis) are visualized at this vertebral level, turning it into an excellent reference point for skeletal muscle measurement.11

Alternatively, visceral fat quantification has been suggested as an alternative diagnostic tool for sarcopenia, comprising measurements at several anatomic levels as it is known that sex, age and body level influence the amount of body fat. Nevertheless, several studies have failed to establish meaningful clinical associations between visceral fat measurements at different lumbar/mid waist levels and patient outcomes.11

In recent studies, sarcopenia was associated with worse long-term and short-term prognosis in patients with GC who underwent surgery.14,15Yet, discordant results regarding this topic have been reported, hence it is of major importance to pursue sarcopenia analysis in order to enable a better understanding of its impact on patients, ultimately allowing the establishment of newer strategies to reduce its incidence.

Methods

Population:

A retrospective, observational, longitudinal and descriptive analysis was conducted, comprising all adults with GC who underwent surgery in Hospital of Braga between 2008 and 2016, resulting in a sample of 147 patients. Patients who died within 30 days following surgery and patients with alternative cancers diagnosed in the following 5 years were excluded from this study. Patients without preoperative abdominal CT imaging were also excluded.

The collected data included gender, age, year of surgery, tumour-nodes-metastasis (TNM) staging, length of hospital stay, postoperative complications and overall survival (OS). Body’s composition encompassed total abdominal muscle area (TAMA) at L3 level and patients’ height (these two measures were accessed only to enable calculation of skeletal muscle index (SMI)).

SMI calculation and Image analysis:

Following clinical and laboratorial data collection, access was granted to the patients' thoracoabdominal-pelvic CT scans in order to get cross-sectional images of the inferior aspect of L3. These images were gathered and saved using DICOM (Digital Imaging and Communications in Medicine) format.

The “National Institutes of Health ImageJ®”16 software was used to calculate the TAMA, following image saving.

For every patient, images were firstly adjusted to their original grey colour (using the [-250, -250] Hounsfield Units (HU) threshold) as it allowed better visualization of anatomical structures16. Then, the outer muscle perimeter was manually drawn and the limits switched to [-29, +150] HU, the threshold for skeletal muscle, followed by outer muscle area determination.16,17An identical process was performed to determine the inner muscle area, as it is illustrated in figure 1.

In order to calculate the TAMA, the inner muscle area was subtracted from the outer muscle area and divided by 100, to obtain results in square centimetres. Later, patients’ SMI calculation was performed by dividing the TAMA by the square of their heights.16

For the 45 patients with unknown height (with no information in their electronic medical record), the SMI was determined using the mean height of each gender in the Portuguese population (women:1.63m; men:1.72m), reported in worldwide research from 2016.18

The sarcopenia cut-off used in this study was the Prado et al. cut-off, which defines sarcopenia as a SMI ≤38.5cm2/m2 for women and ≤52.4cm2/m2 for men.19

Figure 1: A) Drawing of the outer muscle perimeter at L3 level with the [-250, -250 HU] threshold (left) and with the [-25, +129 HU] threshold (right). B) Drawing of the inner muscle perimeter at L3 level with the [-250, -250 HU] threshold (left) and with the [-25, +129 HU] threshold (right). 

Statistical analysis:

Categorical variables are presented as absolute frequencies (n) and percentages (%). Quantitative variables were submitted to normality tests evaluation. Normally distributed variables are characterized by mean (M) and standard deviation (SD), while non-normally distributed data are presented as median (Mdn) and interquartile range (IQR).

Statistical tests used include:

  1. - Pearson's Chi-Square tests (χ2) for the categorical variables,20

  2. - Independent Samples T Tests for the normally distributed data,21

  3. - Mann Whitney U Test for the non-normally distributed variables.

To report survival analysis, we used the Kaplan-Meier method and the Long Rank test to look at OS - defined as the period of time between surgery and date of death.

Univariate and Multivariate Cox Regression were used to evaluate if explored variables were significantly independent risk factors for OS. Results are reported as Hazard Ratio (HR) and Confidence Intervals (CI).

Statistical significance was considered when confidence interval was 95% and p ≤0.05.

The statistical analysis was performed using IBM® Statistical Package for the Social Sciences (SPSS®) - version 26.0.

Results

Study population description:

As indicated above, a sample of 147 patients was selected for this study, of whom 60 (40.8%) were women and 87 (59.2%) were men, with a mean age of 66.03 years (SD=11.51). When we looked for sarcopenia in this study population, 32 patients (21.8%) were found to be sarcopenic. The average length of patients’ hospital stay was 10 days (IQR=4), ranging from 2 to 60 days.

When analysing tumours TNM staging, it was found that 4 (2.7%) tumours were classified as in situ, while 77 (52.4%) patients had stage I cancer, 36 (24.5%) had stage II and 30 (20.4%) were diagnosed with stage III cancer. Postoperative complications occurred in 52 (35.4%) of the 147 sampled individuals.

Regarding SMI, in women, a mean of 47.2 cm2/m2 was obtained with a SD of 1.13, whereas in men, a mean of 57.7 cm2/m2 with a SD of 0.86 was registered. In the entire sample, SMI was found to range between 24.97 cm2/m2 and 84.20 cm2/m2 (Table 1).

Table 1: Characterization of sample’s body composition. 

Association between sarcopenia, patient’s gender and postoperative complications

In the sarcopenic patient’s subgroup, eight (25%) out of 32 were women, while the remaining 24 (75%) were men, and a statistically significant association between gender and sarcopenia’s incidence was found (χ2(1) = 4.24; p=0.040; Φ =-0170) (Table 2). Furthermore, the occurrence of any postoperative complication during hospitalization (χ2(1)=10.3; p=0.001; Φ=0.265) also showed to be significantly different between sarcopenic and non-sarcopenic patients (Table 2). In contrast, there was no statistically significant difference between the two groups regarding age (t(145)= -1.59; p=0.114), either in sarcopenic (68.9±2.15) or non-sarcopenic subgroups (65.2±1.05) (Table 2). Figure 2 shows the difference on computed tomograms between a sarcopenic patient and a non-sarcopenic patient.

Table 2: Statistical analysis of the association between the presence of sarcopenia with the patient’s gender and the occurrence of any postoperative complications. 

Figure 2: A and B) Non-sarcopenic male patient. C and D) Sarcopenic male patient. 

Association between sarcopenia and length of hospital stay

Considering the duration of hospital stay in both groups, we observed that the median in the sarcopenia group (Mdn=15.5; IQR =12) was 5.50 days longer than the non-sarcopenia group (Mdn=10; IQR=2), representing a statistically significant difference between the groups (U=651; p<0.001; r=-0.465) (Table 3).

Table 3: Statistical analysis of the association between sarcopenia and length of hospital stay. 

Survival Analysis:

With regards to survival analysis, it was found that the OS of sarcopenic patients was lower than OS of non-sarcopenic ones, specifically with a mean of 74.1 months for the first group (Std = 8.15; CI [58.1; 90.1]) and 91.9 months for the second one (Std = 4.75; CI [82.5; 101]) (Table 4). Moreover, this difference in total OS time between groups was found to be statistically significant (χ 2 (1) = 4.88 p=0.027).

Table 4: Kaplan Meier analysis comparing the sarcopenic and non-sarcopenic patients’ overall survival. 

Several variables were associated with worse OS, including age (p<0.001), length of hospitalization (p=0.048), TNM stage (p<0.001) and sarcopenia (p=0.03). Of these, only age (p<0.001) and TNM stage (p<0.001) proved to be independent risk factors for worse OS, as length of hospitalization and sarcopenia are related to patient’s OS however they can’t be considered as independent risk factors. This means that each of these predictors effect was correlated when introduced into multivariate analysis. Statistically significant differences were not detected regarding gender (Table 5).

Table 5: Uni and Multivariate Cox regression for overall survival. 

Discussion

The emphasis on sarcopenia and its effect on oncological patients has led to further research on the subject over the last few years.

Considering sarcopenia prevalence, our study documented a higher percentage of sarcopenic patients (21.8%) than Wang et al. (12.5%), Chen et al. (12.8%) and Zheng et al. (14.9%).22,23,24In contrast, our percentage of sarcopenic patients was lower than the ones reported by Tegels et al. (57.7%), Shi et al. (24.4%) and Zhuang et al. (41.5%).25,26,27This disparity is probably explained by the varying sarcopenia cut-offs applied in different analyses. Tegels et al., for example, employed the Martin et al. cut-off, which includes Body Mass Index (BMI) and sex-specific cut-off values.25,28This cut-off proved to be ideal for predicting survival in patients with advanced GC, but it couldn’t be applied to our study since we didn’t have access to the patients BMI.28 Therefore, we used the Prado et al. cut-off, which has been used in previous studies to assess the prognostic value of CT-based sarcopenia in solid tumours,28 in addition to being based on western population’s characteristics.27

In terms of clinical data, only the patients' gender was found to have a significant association with sarcopenia, implying that it is more common in men. This finding is consistent with several previous analyses.24.26,29,30

Looking at postoperative complications as well as length of hospital stay, previous studies revealed significant differences between sarcopenic and non-sarcopenic patients, which correlates with our results.22,23,26,31

In terms of survival, we observed that non-sarcopenic individuals had an average survival time 17.75 months longer than sarcopenic patients, which was found to be statistically significant (p=0.027), and in line with findings reported by Kudou et al., Zhuang et al., Kuwada et al., Tamandl et al..14,27,32,33When using univariate and multivariate analyses, we realized that, although sarcopenia had a significant impact (p=0.03) on outcomes, it cannot be considered as an independent risk factor for poorer OS (p=0.413). This particular finding differs from what was observed by Lee et al. and Kuwada et al. who noticed that sarcopenia could be held as an independent risk factor for worse OS, in addition to having significant impact on outcome.30,32 While Black et al. found no evidence of an association between sarcopenia and lower OS.34

The discrepancies detected when analysing different variables in various studies, highlight the need for further research in order to obtain more conclusive results, allowing implementation and adjustment to clinical practice of new approaches for sarcopenia prevention in oncological patients.

Our study had some limitations, as we did not have full access/availability to patients’ information, namely patients' heights, so in specific cases an average measure was used instead, which may have influenced sarcopenia diagnosis in these patients. Additionally, perimeters obtained from CT-scan for calculation of SMI were manually drawn, which allows for possible operator-dependent measurement errors. Finally, we also point out that this is a retrospective study, which precludes the use of other parameters included in sarcopenia definition, low muscle strength and low physical performance, which could have aided us in establishing more accurate diagnoses.

Conclusion

In conclusion, we would like to emphasize that sarcopenia, in patients with GC who underwent surgery, has an impact on postoperative complications, length of hospital stay and OS, albeit it cannot be considered an independent risk factor for poorer OS.

With this in mind, we believe that some interventions, such as personalized physical exercise and nutritional monitoring, should be considered while managing these patients in order to prevent sarcopenia development.

Since all patients with GC undergo preoperative staging CT, possibly sarcopenia assessment can become a common practice in this population, as several studies, including our own, have demonstrated its impact on patient’s outcomes. Bearing that in mind, we propose the creation of an artificial intelligence software to automatically measure sarcopenia, reducing potential operator-dependent errors and facilitating its use in clinical practice.

References

1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2021;71:209-49. [ Links ]

2. Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Przeglad Gastroenterologiczny. 2019; 14:26-38. [ Links ]

3. Nakamura K, Ueyama T, Yao T, Xuan ZX, Ambe K, Adachi Y, Yakeishi Y, Matsukuma A, Enjoji M. Pathology and prognosis of gastric carcinoma. Findings in 10,000 patients who underwent primary gastrectomy. Cancer. 1992;70:1030-7. [ Links ]

4. Edge SB, Compton CC. The american joint committee on cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Annals of Surgical Oncology. 2010;17:1471-4. [ Links ]

5. Nath J, Moorthy K, Taniere P, Hallissey M, Alderson D. Peritoneal lavage cytology in patients with oesophagogastric adenocarcinoma. The British Journal of Surgery. 2008;95:721-6. [ Links ]

6. Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ, Smith DB, Langley RE, Verma M, Weeden S, Chua YJ, & MAGIC Trial Participants. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. The New England Journal of Medicine. 2006;355:11-20. [ Links ]

7. Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, Martin FC, Michel JP, Rolland Y, Schneider SM, Topinková E, Vandewoude M, Zamboni M, & European Working Group on Sarcopenia in Older People. Sarcopenia: european consensus on definition and diagnosis: report of the european working group on sarcopenia in older people. Age and Ageing. 2010:39:412-23. [ Links ]

8. Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, Cooper C, Landi F, Rolland Y, Sayer AA, Schneider SM, Sieber CC, Topinkova E, Vandewoude M, Visser M, Zamboni M, & Writing Group for the European Working Group on Sarcopenia in Older People 2 (EWGSOP2), and the Extended Group for EWGSOP2. Sarcopenia: revised European consensus on definition and diagnosis. Age and ageing. 2019;48:16-31. [ Links ]

9. Marzetti E, Calvani R, Tosato M, Cesari M, Di Bari M, Cherubini A, Collamati A, D'Angelo E, Pahor M, Bernabei R, Landi F, & SPRINTT Consortium. Sarcopenia: an overview. Aging Clinical and Experimental Research. 2017;29:11-7. [ Links ]

10. Albanese CV, Diessel E, Genant HK. Clinical applications of body composition measurements using DXA. Journal of Clinical Densitometry. 2003;6:75-85. https://doi.org/10.1385/jcd:6:2:75. [ Links ]

11. Yip C, Dinkel C, Mahajan A, Siddique M, Cook GJ, Goh V. Imaging body composition in cancer patients: visceral obesity, sarcopenia and sarcopenic obesity may impact on clinical outcome. Insights into Imaging. 2015;6:489-97. [ Links ]

12. Mitsiopoulos N, Baumgartner RN, Heymsfield SB, Lyons W, Gallagher D, Ross R. Cadaver validation of skeletal muscle measurement by magnetic resonance imaging and computerized tomography. Journal of Applied Physiology. 1998;85:115-22. [ Links ]

13. Shen W, Punyanitya M, Wang Z, Gallagher D, St-Onge MP, Albu J, Heymsfield SB, Heshka S. Total body skeletal muscle and adipose tissue volumes: estimation from a single abdominal cross-sectional image. Journal of Applied Physiology. 2004;97:2333-8. [ Links ]

14. Kudou K, Saeki H, Nakashima Y, Edahiro K, Korehisa S, Taniguchi D, Tsutsumi R, Nishimura S, Nakaji Y, Akiyama S, Tajiri H, Nakanishi R, Kurashige J, Sugiyama M, Oki E, Maehara Y. Prognostic significance of sarcopenia in patients with esophagogastric junction cancer or upper gastric cancer. Annals of Surgical Oncology . 2017;24:1804-10. [ Links ]

15. Su H, Ruan J, Chen T, Lin E, Shi L. 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. 2019;19:82. [ Links ]

16. Gomez-Perez SL, Haus JM, Sheean P, Patel B, Mar W, Chaudhry V, McKeever L, Braunschweig C. Measuring abdominal circumference and skeletal muscle from a single cross-sectional computed tomography image: a step-by-step guide for clinicians using national institutes of health image. JPEN- Journal of Parenteral and Enteral Nutrition. 2016;40:308-18. [ Links ]

17. Mourtzakis M, Prado CM, Lieffers JR, Reiman T, McCargar LJ, Baracos VE. A practical and precise approach to quantification of body composition in cancer patients using computed tomography images acquired during routine care. Applied Physiology, Nutrition, and Metabolism. 2008;33:997-1006. [ Links ]

18. NCD Risk Factor Collaboration (NCD-RisC). A century of trends in adult human height. eLife, 2016;5:e13410. [ Links ]

19. Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, Baracos VE. Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. The Lancet - Oncology. 2008;9:629-35. [ Links ]

20. Chan YH. Biostatistics 103: qualitative data - tests of independence. Singapore Medical Journal. 2003;44:498-503. [ Links ]

21. Chan YH. Biostatistics 102: quantitative data--parametric & non-parametric tests. Singapore Medical Journal. 2003;44:391-6. [ Links ]

22. Wang SL, Zhuang CL, Huang DD, Pang WY, Lou N, Chen FF, Zhou CJ, Shen X, Yu Z. Sarcopenia adversely impacts postoperative clinical outcomes following gastrectomy in patients with gastric cancer: a prospective study. Annals of Surgical Oncology . 2016;23:556-64. [ Links ]

23. Chen XY, Li B, Ma BW, Zhang XZ, Chen WZ, Lu LS, Shen X, Zhuang CL, Yu Z. Sarcopenia is an effective prognostic indicator of postoperative outcomes in laparoscopic-assisted gastrectomy. European Journal of Surgical Oncology. 2019;45:1092-8. [ Links ]

24. Zheng ZF, Lu J, Zheng CH, Li P, Xie JW, Wang JB, Lin JX, Chen QY, Lin M, Huang CM. A novel prognostic scoring system based on preoperative sarcopenia predicts the long-term outcome for patients after R0 resection for gastric cancer: experiences of a high-volume center. Annals of Surgical Oncology . 2017;24:1795-803. [ Links ]

25. Tegels JJ, van Vugt JL, Reisinger KW, Hulsewé KW, Hoofwijk AG, Derikx JP, Stoot JH. Sarcopenia is highly prevalent in patients undergoing surgery for gastric cancer but not associated with worse outcomes. Journal of Surgical Oncology. 2015;112:403-7. [ Links ]

26. Shi B, Liu S, Chen J, Liu J, Luo Y, Long L, Lan Q, Zhang Y. Sarcopenia is associated with perioperative outcomes in gastric cancer patients undergoing gastrectomy. Annals of Nutrition & Metabolism. 2019;75:213-22. [ Links ]

27. Zhuang CL, Huang DD, Pang WY, Zhou CJ, Wang SL, Lou N, Ma LL, Yu Z, Shen X. Sarcopenia is an independent predictor of severe postoperative complications and long-term survival after radical gastrectomy for gastric cancer: analysis from a large-scale cohort. Medicine. 2016;95:e3164. [ Links ]

28. Nishigori T, Tsunoda S, Obama K, Hisamori S, Hashimoto K, Itatani Y, Okada K, Sakai Y. Optimal cutoff values of skeletal muscle index to define sarcopenia for prediction of survival in patients with advanced gastric cancer. Annals of Surgical Oncology . 2018;25:3596-603. [ Links ]

29. Fukuda Y, Yamamoto K, Hirao M, Nishikawa K, Nagatsuma Y, Nakayama T, Tanikawa S, Maeda S, Uemura M, Miyake M, Hama N, Miyamoto A, Ikeda M, Nakamori S, Sekimoto M, Fujitani K, Tsujinaka T. Sarcopenia is associated with severe postoperative complications in elderly gastric cancer patients undergoing gastrectomy. Gastric Cancer . 2016;19:986-93. [ Links ]

30. Lee JS, Kim YS, Kim EY, Jin W. Prognostic significance of CT-determined sarcopenia in patients with advanced gastric cancer. PloS One. 2018;13:e0202700. [ Links ]

31. Huang DD, Zhou CJ, Wang SL, Mao ST, Zhou XY, Lou N, Zhang Z, Yu Z, Shen X, Zhuang CL. Impact of different sarcopenia stages on the postoperative outcomes after radical gastrectomy for gastric cancer. Surgery. 2017;161:680-93. [ Links ]

32. Kuwada K, Kuroda S, Kikuchi S, Yoshida R, Nishizaki M, Kagawa S, Fujiwara T. Sarcopenia and comorbidity in gastric cancer surgery as a useful combined factor to predict eventual death from other causes. Annals of Surgical Oncology . 2018;25:1160-6. [ Links ]

33. Tamandl D, Paireder M, Asari R, Baltzer PA, Schoppmann SF, & Ba-Ssalamah A. Markers of sarcopenia quantified by computed tomography predict adverse long-term outcome in patients with resected oesophageal or gastro-oesophageal junction cancer. European Radiology. 2016;26:1359-67. [ Links ]

34. Black D, Mackay C, Ramsay G, Hamoodi Z, Nanthakumaran S, Park K, Loudon MA, Richards CH. Prognostic value of computed tomography: measured parameters of body composition in primary operable gastrointestinal cancers. Annals of Surgical Oncology . 2017;24:2241-51. [ Links ]

Ethical Disclosures

Financing Support: This work has not received any contribution, grant or scholarship.

Received: February 21, 2023; Accepted: June 14, 2023

Address Daniela Barros, Serviço de Radiologia, Hospital de Braga, Sete Fontes - São Victor, 4710-243 Braga, Portugal, e-mail: danielabarros93@gmail.com

Conflicts of interest: The authors have no conflicts of interest to declare.

Confidentiality of data: The authors declare that they have followed the protocols of their work center on the publication of data from patients.

Protection of human and animal subjects: The authors declare that the procedures followed were in accordance with the regulations of the relevant clinical research ethics committee and with those of the Code of Ethics of the World Medical Association (Declaration of Helsinki).

© Author(s) (or their employer(s)) and ARP 2023. Re-use permitted under CC BY-NC. No commercial re-use.

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License