Radiol Oncol 2024; 58(1): 1-8. doi: 10.2478/raon-2024-0011 1 review Sarcopenic obesity in cancer Mihaela Jurdana 1 , Maja Cemazar 1,2 1 Faculty of Health Sciences, University of Primorska, Izola, Slovenia 2 Institute of Oncology Ljubljana, Ljubljana, Slovenia Radiol Oncol 2024; 58(1): 1-8. Received 2 November 2023 Accepted 4 December 2023 Correspondence to: Mihaela Jurdana, Ph.D., Faculty of Health Sciences, University of Primorska, Polje 42, Izola, Slovenia. E-mail: mihaela.jurdana@fvz.upr.si Disclosure: No potential conflicts of interest were disclosed. This is an open access article distributed under the terms of the CC-BY license (https://creativecommons.org/licenses/by/4.0/). Background. Sarcopenic obesity is a relatively new term. It is a clinical condition characterized by sarcopenia (loss of muscle mass and function) and obesity (increase in fat mass) that mainly affects older adults. As the incidence of sar- copenia and obesity increases worldwide, sarcopenic obesity is becoming a greater problem also in cancer patients. In fact, sarcopenic obesity is associated with poorer treatment outcomes, longer hospital stays, physical disability, and shorter survival in several cancers. Oxidative stress, lipotoxicity, and systemic inflammation, as well as altered expres- sion of skeletal muscle anti-inflammatory myokines in sarcopenic obesity, are also associated with carcinogenesis. Conclusions. Reported prevalence of sarcopenic obesity in cancer varies because of heterogeneity in definitions and variability in diagnostic criteria used to estimate the prevalence of sarcopenia and obesity. Therefore, the aim of this review is to describe the definitions, prevalence, and diagnostic criteria as well as the mechanisms that cancer has in common with sarcopenic obesity. Key words: sarcopenia; obesity; cancer; inflammation Introduction Sarcopenic obesity is a clinical condition character- ised by the coexistence of obesity, excess fat mass (FM) and sarcopenia (decrease in skeletal muscle mass and function). 1,2 Baumgartner was the first to propose the term sarcopenic obesity, which is con- sidered a unique clinical condition distinct from obesity and sarcopenia alone. 1 The prevalence of sarcopenic obesity is expected to become a pub- lic health problem as the prevalence of sarcopenic obesity in adults is rapidly increasing worldwide. The clinical consequences of sarcopenic obesity are considerably more severe than those of sarcope- nia or obesity alone. In comparison to both, it can lead to physical disability, morbidity, and even mortality. 3 In addition, cancer is another serious global health problem with increasing incidence and mortality worldwide. 4 Interestingly, sarcopen- ic obesity is more common in older adult cancer patients and patients with other chronic diseases, but its prevalence is also increased in younger patients with obesity and chronic diseases such as cancer and is associated with worse treatment outcomes. 5 Sarcopenic obesity and cancer, as well as other chronic diseases, share some key pathoge- netic mechanism such as inflammation, oxidative stress, and insulin resistance, which are considered key factors. 6-8 Among these, insulin resistance is considered a central condition in both, cancer and sarcopenic obesity. 8 Several factors in sarcopenic obesity may lead to progressive loss of muscle mass and altered fat metabolism, which influence each other in a recip- rocal pattern. Sarcopenia is known to be a common problem in cancer patients, especially if they are suffering cachexia and is associated with physical disability, surgical complications, increased risk of severe toxicity during cancer treatment, pro- longed hospitalisation, and shortened survival. 6 Sarcopenia can occur at any stage of cancer and in any body mass index (BMI) category and is of- Radiol Oncol 2024; 58(1): 1-8. Jurdana M et al. / Sarcopenic obesity and cancer 2 ten associated with obesity. In addition, obesity, particularly abdominal obesity, can independently lead to loss of muscle mass and function due to the negative effects of oxidative stress, inflamma- tion, and insulin resistance, all of which negatively impact muscle mass. 7 Recently, there has been an increasing interest in utilizing body composition phenotype as an additional indicator of cancer prognosis. Because the burden of sarcopenia and obesity are concurrent, they can be expected to have a combined impact on health outcomes in several clinical settings, including cancer. 6 As the prevalence of sarcopenic obesity with poor prognosis is rapidly increasing in cancer patients, in this review we discuss definitions, prevalence, mechanisms and management strate- gies for sarcopenic obese patients in oncology. This review aims to provide clinician’s with additional evidence helping them to make rational decisions that will improve patients’ outcome. Identification of sarcopenic obesity and diagnostic criteria Sarcopenic obesity has been identified using vari- ous definitions and diagnostic criteria. Screening for sarcopenic obesity is based on the simultane- ous presence of increased BMI or waist circumfer- ence with ethnicity-specific cut-offs and surrogate parameters of sarcopenia (clinical symptoms, vali- dated questionnaires -SARC-F in the elderly). In addition, altered body composition is required to make a definitive diagnosis. 9,10 Many studies have investigated the prevalence of sarcopenic obesity in different cohorts of can- cer patients, including oropharyngeal 11 , lung 12,13 , gastrointestinal 14,15-24 , liver and pancreatic 25,26 , uri- nary 27-29 , breast 30 , melanoma 31 and lymphoma. 32 On the other hand, the lack of uniform diagnos- tic criteria for sarcopenic obesity hampers the identification of patients and the assessment of associated outcomes and, consequently, nega- tively affects the development of prevention and treatment strategies for sarcopenic obesity. The prevalence of sarcopenic obesity in studies that in- clude cancer patients varies from 1-29% in studies of patients in all BMI categories and from 15% to 36% in studies of overweight/obese patients. 6 The lowest prevalence of sarcopenic obesity is found in early disease stages and the highest in locally advanced or metastatic disease. 6 Heterogeneity in diagnostic criteria between studies, as well as the metabolic impact of different cancer types, other patient characteristics such as ethnicity, and concurrent comorbidities, limit interpretation of results. In addition, The European Society for Clinical Nutrition and Metabolism (ESPEN) and the European Association for the Study of Obesity (EASO) confirmed heterogeneity in definition and diagnostic approaches for sarcopenic obesity, due to different definition of obesity and sarcopenia, differences in methodologies to assess body com- position and function, as well as in the applied references values for variables used. 9 Regardless of this variability, most studies report that sarcopenic obesity is an important problem in cancer patients and a negative prognostic factor. Commonly used technics in oncology setting Various techniques and body composition parame- ters with different cut off as well as muscle function parameters have been used to identify sarcopenic obesity. Body composition is traditionally meas- ured using Dual Energy X-ray Absorptiometry (DEXA) or Bioelectrical Impedance Analysis (BIA) but is known to have some limitations. 9 Currently, computed tomography (CT) and magnetic reso- nance imaging (MRI) are considered the gold standard methods. Both CT and MRI are not rou- tinely used to assess sarcopenia and obesity due to high costs and radiation concerns for CT. 33,34 CT should be used when possible (e.g., in patients undergoing a scan for diagnostic reasons in oncol- ogy). 9 In cancer patients, the CT scan provides the highest available precision in determining body composition parameters by measuring the cross- sectional area of total skeletal muscle at the third lumbar vertebra (L3), which correlates strongly with total body skeletal muscle mass. 35 The DEXA method, which is considered inex- pensive, is the most accurate method for measur- ing appendicular muscle mass, and exposes the patient to minimal radiation, but is not widely available. The BIA method is an alternative, inex- pensive, and readily available method, but results are easily confounded by various factors, espe- cially fluid status. 34 All these techniques provide anatomic information about the patient but not functional information, because loss of muscle function is also required to diagnose sarcopenia. 36 Therefore, the diagnostic process must include a direct assessment of altered skeletal muscle func- tion parameters along with altered body compo- sition. Thereafter, individuals with a positive di- Radiol Oncol 2024; 58(1): 1-8. Jurdana M et al. / Sarcopenic obesity and cancer 3 agnosis should be classified into two stages: Stage I, when no clinical complications are present, and Stage II, with clinical complications such as chron- ic disease (e.g. cancer), dietary events (weight loss, decreased food intake), immobility, falls, and other complaints associated with altered body compo- sition and muscle function. 9 All procedures are summarized in Table 1. Biological pathways leading to sarcopenic obesity The biological pathway leading to sarcopenic obesity includes changes in body composition re- lated to ageing, hormonal changes, the interplay between metabolism and inflammation, environ- mental factors such as poor nutrition and lack of exercise and chronic diseases. 37-40 This results in a decrease in oxidative capacities, mitochondrial number, atrophy of fast type II muscle fibres and neurodegeneration, decrease in protein synthesis and increase muscle protein degradation. 41 Metabolic disfunctions Both, skeletal muscle mass and adipose tissue interplay with several cancers at metabolic lev- els. 42-45 Few studies have investigated the path- way between sarcopenia, obesity and cancer. The mechanisms involved in pathogenesis of metabolic imbalances associated with obesity are in part com- mon with pathway modulating cancer related sar- copenia. 5 Inflammation promoted by cancer and/or inadequate intake of essential nutrients could con- tribute to the presence of fatigue and decrease in physical activity and mobility of cancer patients. 6 Physical inactivity can lead to skeletal muscle loss by reduced protein anabolic pathway and activation of proteolytic pathway. 5,6 In addition, inadequate food intake can also impair muscle anabolic pathway due to low omega -3 (n-3) fatty acid intake. Namely, it was demonstrated that low level of n-3 fatty acids is associated with loss of muscle mass and skeletal muscle fat infiltration or myosteatosys. 46 In addition, atrophy of fast type II muscle fibre and switch to slow type I muscle fibre increase the lipid deposition into the mus- cle. 6 Importantly, myosteatosis is associated with metabolic muscle disfunction and muscle function loss and is widespread in cancer associated mal- nourished patients. 47,48 A vicious cycle between myocytes and adipocytes is responsible for sarco- penic obesity. Specifically, adipose tissue inflam- mation and dysfunction leads to overproduction of fatty acids which in combination with low oxi- dation capacity of skeletal muscle stimulate the formation of intramyocellular lipid (IMCL). 49 This process blocks the translocation of glucose trans- porter type 4 (GLUT4) to the surface of the muscle fiber and therefore hampers uptake of glucose by skeletal muscle resulting in a decrease of glucose utilization and increase of fatty acid oxidation in mitochondria, which leads to impaired insulin sensitivity of skeletal muscle, inhibition of mito- chondrial respiration, increase in reactive oxygen species formation (ROS), myocyte toxicity, inflam- mation and finally sarcopenia. 5,6 Various forms of fat accumulation in skeletal muscle have been associated with insulin resistance, mitochondrial dysfunction and decreased muscle contracting force. 50 Based on these mechanisms, myosteato- sis is a potentially important factor in sarcopenic obesity in the cancer setting, which could contrib- ute to further muscle dysfunction in sarcopenic obese cancer patients and is an independent pre- dictor of reduced survival in cancer patients. 47, 51 Additionally, ectopic fat deposition surrounding muscle accelerate proteolysis in muscle tissue lead- ing to further muscle loss and worsen outcome. 47 Another consequence of insulin resistance is the reduction in the uptake of amino acids by mus- cle cells, altering the balance of protein synthesis/ degradation in favour of proteolysis. It is proposed TABLE 1. Selected criteria to identify sarcopenic obesity. Altered skeletal muscle function parameters considering muscle strength and physical performance and altered body composition parameters should be present to assess sarcopenic obesity SCREENING DIAGNOSIS STAGES High BMI and WC (based on ethnic cut-points) Altered skeletal muscle strength (HGS, chair stand test) STAGE 1: Without complications Surrogate markers of sarcopenia: (clinical symptoms or validated questionaries’ e.g. SARC-F) Altered body composition (increased FM, decrease MM) STAGE 2: One or more complications attributable to sarcopenic obesity BMI = body mass index; FM = fat mass; HGS = hand grip strength; MM = muscle mass; SARC-F = strength, assistance in walking, rise from a chair, climb stairs, and falls; WC = waist circumference; Radiol Oncol 2024; 58(1): 1-8. Jurdana M et al. / Sarcopenic obesity and cancer 4 that the amino acids released by muscle proteoly- sis in sarcopenia contribute to the supply of tumor growth. 8 Hormonal imbalances Furthermore, cancer is associated with an altera- tion in hormones that severely affects skeletal mus- cle and fat metabolism. 6 The body composition of cancer patients is associated with insulin, insulin resistance, and the hormone ghrelin, the levels of which are modulated in obese patients and affect skeletal muscle metabolism. In addition, increased levels of stress hormones and a decrease in andro- gens and estrogens affect the anabolic and catabolic conditions for muscle protein metabolism and lead to alterations in the production and metabolism of anabolic hormones (growth hormone, insulin-like growth factor (IGF-1)) that may result in a sarco- penic obese phenotype. 52,53 Increasing insulin re- sistance is associated with an increase in intramyo- cellular fat mass and loss of muscle function. 54 Cytokine imbalances The systemic inflammatory condition in sar- copenic obesity as well as the altered expression of myokines and adipokines are also involved in carcinogenesis. Skeletal muscle and adipose tissue are considered endocrine organs due to release an- ti-inflammatory myokines and pro -inflammatory cytokines, respectively. 55 Myokines are proteins re- leased by muscle cells in response to contractions. They play autocrine, paracrine, and endocrine roles in many exercise-induced adaptations (e.g., muscle hypertrophy and cancer protection). 55,56 The re- lease of pro- and anti-inflammatory cytokines by adipose and muscle tissue has a strong influence on skeletal muscle and adipose tissue metabolism and is involved in various cancer related changes in body composition. 5 Dysregulation of pro- and anti-inflammatory cytokines is responsible for muscle intracellular adipose tissue. They contrib- ute to the secretion of myostatin a negative regu- lator of muscle mass secreted by skeletal muscle cells, tumor necrosis factor –α (TNF- α), proin- flammatory interleukin -6 (IL-6), interleukin -1β (IL-1β), mononuclear chemoattractant protein-1 (MCP-1), and downregulate the secretion of anti- inflammatory adiponectin, leading to lipotoxic- ity and insulin resistance. 8,37 ,57-59 Inflammatory cytokines directly affect skeletal muscle and accel- erate muscle proteins degradation and apoptosis and induces muscle tissue reduction and fat tissue accumulation. 5,6,8 Levels of IL -6 and TNF- α are further increased by the hormone leptin, reduc- ing the anabolic pathway of IGF-1 and enhancing lipotoxicity. The cytokine-like hormone leptin is a classic adipokine that is secreted by adipocytes, its blood concentration correlates with triglyceride accumulation in adipocytes. 60 In addition to this, the impaired secretion of skeletal muscle anti-in- flammatory myokines such as interleukin -15 (IL- 15), irisin, muscle derived IL-6 accelerate muscle atrophy and disfunction. 8,56,59,61 All these cytokines are known to be involved in several cancer induced alteration of body compo- sition. 62 Altered fat metabolism promotes inflam- mation, having an important role in cancer and non-cancer muscle wasting 63 creating a vicious cycle for sarcopenic obesity, leading to morbidity and mortality. Sarcopenic obesity and carcinogenesis, there- fore, are mediated by mechanisms, such as insu- lin resistance, adiposity, proteolysis, myosteatosis, inflammation, oxidative stress, imbalance of adi- pokines and myokines (Figure 1). Therapeutic strategies Patients with cancer and sarcopenic obesity have several specific adverse clinical outcomes, includ- ing higher risk of dose-limiting toxicity, surgical complications, prolonged hospitalisation, physi- cal disability, and shorter survival. This was dem- onstrated for several cancers. 6 Chemotherapy has been shown to alter body composition by reducing lean mass, thus favouring the development of sar- copenic obesity. 64 Low muscle mass and strength is common in any cancer stage and is clearly consid- ered an independent predictor for cancer progres- sion, making it a preferred target in the treatment of sarcopenic obesity. On the other hand, obesity is not univocally associated with worse outcome in cancer patients regardless of the cancer type. 65 Although obesity is considered an important risk factor for the development of various cancers, the presence of moderate obesity paradoxically ap- pears to be a protective factor that may improve response to treatment and consequently survival in patients but there are conflicting and controversial results. 66 Several therapeutic strategies, exercise and nu- tritional exists to counteract sarcopenic obesity. Prado et al. have shown that poor nutritional sta- tus in cancer is primarily manifested by a severe loss of muscle mass, which can occur at any stage Radiol Oncol 2024; 58(1): 1-8. Jurdana M et al. / Sarcopenic obesity and cancer 5 (from curative to palliative treatment) and often co-exists with obesity. They have addressed the role of diet in preventing and reversing sarcopenia in cancer patients, which may also apply to sar- copenic obesity. 67 Nutritional strategies comprise the importance of adequate intake of macro- and micronutrients, including high-quality proteins, branched-chain amino acids (leucine), β-hydroxy- β-methylbutyrate, glutamine, carnitine, creatine, fish oil/eicosapentanoic acid (EPA and DHA), vi- tamins/minerals (e.g., vitamin D), and multimodal approaches (diet, exercise, and medications) to counteract low muscle mass in cancer. Moreover, physical activity could be an impor- tant and effective treatment strategy to reverse sarcopenia by promoting insulin sensitivity, re- ducing oxidative stress and inflammation, and stimulating mitochondrial biosynthesis. Both re- sistance training and aerobic exercise have been shown to improve muscle mass and physical per- formance. 68,69 We note that standard treatment recommendations derived from studies in the el- derly and certain diseases may not be applicable to cancer patients due to fatigue or pain. However, many intervention studies of physical activity in cancer indicate some benefit of exercise for mus- cle strength and endurance. 70 In addition, cancer obese individuals, who exercise more and are not insulin resistant or hypertensive might then have a lower mortality risk. 8 Another effective treatment for sarcopenic obe- sity in older adults and cancer patients is skeletal muscle electrostimulation. It causes contraction of muscle fibres via neuromuscular activation and can induce a change in body composition. 71 Whole-body vibration therapy has been shown to significantly increase muscle strength and func- tion in older adults. 72 Further clinical studies are needed to verify its efficacy in clinical practise. Conclusions The prevalence of sarcopenic obesity is considered a novel factor of great clinical relevance in cancer patients, leading to postoperative complications, worse functional status, and shorter survival pos- sibly mediated by interactions among patho- SARCOPENIC OBESITY Aging Obesity Physical in ac�vity Muscle mass and strenght An �-in flammatory myokines GLUT 4 IGF IMCL Fat mass, visceral fat In flammatory cytokines Lep �n Adiponec �n Cancer In flammatory cytokines Inadequate food intake Fa �gue Physical ac�vity and mobility Myokine de ficiency Proteolysis Insuline resistance In flamma �o n Oxida �ve stress Lipotoxicity Myosteatosis SKELETAL MUSCLE ATROPHY AND DISFUNCTION MORTALITY .. FIGURE 1. Main pathophysiological mechanisms in cancer patients with sarcopenic obesity. Body composition (low skeletal muscle mass and strength, increase in fat mass), inflammation, insulin resistance, myosteatosis, myokine dysregulation, and oxidative stress in sarcopenic obese cancer patients significantly induce muscle proteolysis, leading to muscle wasting and dysfunction and premature mortality. Radiol Oncol 2024; 58(1): 1-8. Jurdana M et al. / Sarcopenic obesity and cancer 6 physiological mechanisms (inflammation, insulin resistance, dysregulation of myokines and pro- inflammatory cytokines). Specific prevention and treatment strategies are needed to address sarco- penic obesity in cancer patients. One of the major challenges in prevention strategies is to maintain skeletal muscle mass and function and reduce fat mass, because the combination of decreasing skel- etal muscle mass and increasing fat mass leads to physical limitations that worsen the prognosis for chronic disease, including cancer. Exercise and proper nutrition are two key components in the prevention and treatment of sarcopenic obesity, but effective interventions should be explored for cancer patients. 3,4,6,56,73 The key question is how to maintain muscle anabolism in an energy deficit situation to avoid a high percentage of weight loss in the form of lean mass in this muscle loss prone population. The underestimated prevalence of sarcopenic obesity in cancer are the consequence of the lack of standard methods and definitions used in previ- ous studies. Therefore, further studies need to fo- cus on screening sarcopenic obesity in cancer pa- tients, and additional studies are needed to clarify the pathogenesis of sarcopenic obesity, with em- phasis on identifying new markers. In conclusion, the development of standard- ised diagnostic criteria is urgently needed. Up to now the identification of sarcopenia by measuring muscle mass and strength as a physical function has been done only in few studies, while in all oth- ers muscle mass was used as the only criteria. ESPEN and EASO have launched an initiative to reach expert consensus on diagnostic procedures that include an assessment of skeletal muscle func- tion followed by an assessment of body composi- tion. Individuals with sarcopenic obesity should be classified into stages 1 and 2 based on clinical complications associated with body composition or muscle dysfunction. The proposed definition should be implemented in routine clinical practice. In addition, validation and prospective follow-up studies and secondary analyses of existing co- horts are proposed and encouraged by ESPEN and EASO. 9 References 1. Baumgartner RN. Body composition in healthy aging. Ann N Y Acad Sci 2000; 904: 437-48. doi: 10.1111/j.1749-6632.2000.tb06498.x 2. Barazzoni R, Bischoff SC, Boirie Y, Busetto L, Cederholm T, Dicker D, et al. Sarcopenic obesity: Time to meet the challenge. Clin Nutr 2018; 37(6 Pt A): 1787-93. doi: 10.1016/j.clnu.2018.04.018 3. Gao Q, Mei F, Shang Y , Hu K, Chen F, Zhao L, et al. Global prevalence of sar- copenic obesity in older adults: a systematic review and meta-analysis. Clin Nutr 2021; 40: 4633-41. doi: 10.1016/j.clnu.2021.06.009 4. Gao Q, Hu K, Gao J, Shang Y, Mei F, Zhao L, et al. Prevalence and prog- nostic value of sarcopenic obesity in patients with cancer: a system- atic review and meta-analysis. Nutrition 2022; 101: 111704. doi: 10.1016/j. nut.2022.111704 5. Gortan Cappellari G, Brasacchio C, Laudisio D, Lubrano C, Pivari F, Barrea L, et al. Sarcopenic obesity: what about in the cancer setting? Nutrition 2022; 98: 111624. doi: 10.1016/j.nut.2022.111624 6. Carneiro IP , Mazurak VC, Prado CM. Clinical implications of sarcopenic obe- sity in cancer . Curr Oncol Rep 2016; 18: 62. doi: 10.1007/s11912-016-0546-5 7. Atkins JL, Wannamathee SG. Sarcopenic obesity in ageing: cardiovascular outcomes and mortality. Br J Nutr 2020; 124: 1102-13. doi: 10.1017/ S0007114520002172 8. Silveira EA, da Silva Filho RR, Spexoto MCB, Haghighatdoost F , Sarrafzadegan N, de Oliveira C. The role of sarcopenic obesity in cancer and cardiovascular disease: a synthesis of the evidence on pathophysiological aspects and clini- cal implications. Int J Mol Sci 2021; 22: 4339. doi: 10.3390/ijms22094339 9. Donini LM, Busetto L, Bischoff SC, Cederholm T, Ballesteros-Pomar MD, Batsis JA, et al. Definition and diagnostic criteria for sarcopenic obesity: ESPEN and EASO consensus statement. Obes Facts 2022; 15: 321-35. doi: 10.1159/000521241 10. Batsis JA, Villareal DT . Sarcopenic obesity in older adults: aetiology, epidemi- ology and treatment strategies. Nat Rev Endocrinol 2018; 14: 513-37. doi: 10.1038/s41574-018-0062-9 11. Chargi N, Bril SI, Swartz JE, Wegner I, Willems SM, de Bree R. Skeletal muscle mass is an imaging biomarker for decreased survival in patients with oro- pharyngeal squamous cell carcinoma. Oral Oncol 2020; 101: 104519. doi: 10.1016/j.oraloncology.2019.104519 12. Kiss N, Beraldo J, Everitt S. Early skeletal muscle loss in non-small cell lung cancer patients receiving chemoradiation and relationship to survival. Support Care Cancer 2019; 27: 2657-64. doi: 10.1007/s00520-018-4563-9 13. Recio-Boiles A, Galeas JN, Goldwasser B, Sanchez K, Man LMW , Gentzler RD, et al. 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 2018; 26: 2353-59. doi: 10.1007/s00520-018-4051-2 14. Lodewick TM, van Nijnatten TJ, van Dam RM, van Mierlo K, Dello SA, Neumann UP, et al. Are sarcopenia, obesity and sarcopenic obesity predic- tive of outcome in patients with colorectal liver metastases? HPB (Oxford) 2015; 17: 438-46. doi: 10.1111/hpb.12373 15. Grotenhuis BA, Shapiro J, van Adrichem S, de Vries M, Koek M, Wijnhoven BP, et al. Sarcopenia/muscle mass is not a prognostic factor for short- and long-term outcome after esophagectomy for cancer. World J Surg 2016; 40: 2698-704. doi: 10.1007/s00268-016-3603-1 16. Kim YM, Kim JH, Baik SJ, Chun J, Youn YH, Park H. Sarcopenia and sarcopenic obesity as novel risk factors for gastric carcinogenesis: a health checkup cohort study. Front Oncol 2019; 9: 1249. doi: 10.3389/fonc.2019.01249 17. Lou N, Chi CH, Chen XD, Zhou CJ, Wang SL, Zhuang CL, et al. Sarcopenia in overweight and obese patients is a predictive factor for postoperative complication in gastric cancer: a prospective study. Eur J Surg Oncol 2017; 43: 188-95. doi: 10.1016/j.ejso.2016.09.006 18. Nishigori T, Tsunoda S, Okabe H, Tanaka E, Hisamori S, Hosogi H, et al. Impact of sarcopenic obesity on surgical site infection after laparoscopic total gastrectomy. Ann Surg Oncol 2016; 23(Suppl 4): 524-31. doi: 10.1245/ s10434-016-5385-y 19. Palmela C, Velho S, Agostinho L, Branco F, Santos M, Santos MP , et al. Body composition as a prognostic factor of neoadjuvant chemotherapy toxic- ity and outcome in patients with locally advanced gastric cancer. J Gastric Cancer 2017; 17: 74-87. doi: 10.5230/jgc.2017.17.e8 20. Sugawara K, Yamashita H, Okumura Y , Yagi K, Yoshimura S, Kawasaki K, et al. Relationships among body composition, muscle strength, and sarcopenia in esophageal squamous cell carcinoma patients. Support Care Cancer 2020; 28: 2797-803. doi: 10.1007/s00520-019-05110-7 21. Zhang WT , Lin J, Chen WS, Huang YS, Wu RS, Chen XD, et al. Sarcopenic obe- sity is associated with severe postoperative complications in gastric cancer patients undergoing gastrectomy: a prospective study. J Gastrointest Surg 2018; 22: 1861-69. doi: 10.1007/s11605-018-3835-5 Radiol Oncol 2024; 58(1): 1-8. Jurdana M et al. / Sarcopenic obesity and cancer 7 22. Giani A, Famularo S, Riva L, Tamini N, Ippolito D, Nespoli L, et al. Association between specific presurgical anthropometric indexes and morbidity in pa- tients undergoing rectal cancer resection. Nutrition. 2020; 75-76: 110779. doi: 10.1016/j.nut.2020.110779 23. Han JS, Ryu H, Park IJ, Kim KW , Shin Y , Kim SO, et al. Association of body com- position with long-term survival in non-metastatic rectal cancer patients. Cancer Res Treat 2020; 52: 563-72. doi: 10.4143/crt.2019.249 24. Malietzis G, Currie AC, Athanasiou T, Johns N, Anyamene N, Glynne-Jones R, et al. Influence of body composition profile on outcomes following colo- rectal cancer surgery. Br J Surg 2016; 103: 572-80. doi: 10.1002/bjs.10075 25. Gruber ES, Jomrich G, Tamandl D, Gnant M, Schindl M, Sahora K. Correction: Sarcopenia and sarcopenic obesity are independent adverse prognostic fac- tors in resecTable pancreatic ductal adenocarcinoma. PLoS One 2020; 15: e0244896. doi: 10.1371/journal.pone.0244896. Erratum for: PLoS One 2019 6; 14: e0215915. doi: 10.1371/journal.pone.0215915 26. Kobayashi A, Kaido T, Hamaguchi Y , Okumura S, Shirai H, Yao S, et al. Impact of sarcopenic obesity on outcomes in patients undergoing hepatectomy for hepatocellular carcinoma. Ann Surg 2019; 269: 924-31. doi: 10.1097/ SLA.0000000000002555 27. Kimura Y, Yamada M, Ohji S, Ishiyama D, Nishio N, Otobe Y, et al. Presence of sarcopenic obesity and evaluation of the associated muscle quality in Japanese older men with prostate cancer undergoing androgen deprivation therapy. J Geriatr Oncol 2019; 10: 835-8. doi: 10.1016/j.jgo.2019.03.017 28. Kocher NJ, Jafri S, Balabhadra S, Lehman E, Gardner J, Vijay K, et al. Is sarcopenia and sarcopenic obesity associated with clinical and pathological outcomes in patients undergoing radical nephroureterectomy? Urol Oncol 2018; 36: 156. e17-156.e22. doi: 10.1016/j.urolonc.2017.12.004 29. Cushen SJ, Power DG, Murphy KP, McDermott R, Griffin BT, Lim M, et al. Impact of body composition parameters on clinical outcomes in patients with metastatic castrate-resistant prostate cancer treated with docetaxel. Clin Nutr ESPEN 2016; 13: e39-45. doi: 10.1016/j.clnesp.2016.04.001 30. Rier HN, Jager A, Sleijfer S, van Rosmalen J, Kock MCJM, Levin MD. Low muscle attenuation is a prognostic factor for survival in metastatic breast cancer patients treated with first line palliative chemotherapy. Breast 2017; 31: c9-15. doi: 10.1016/j.breast.2016.10.014 31. Heidelberger V, Goldwasser F, Kramkimel N, Jouinot A, Huillard O, Boudou- Rouquette P , et al. Sarcopenic overweight is associated with early acute lim- iting toxicity of anti-PD1 checkpoint inhibitors in melanoma patients. Invest New Drugs 2017; 35: 436-41. doi: 10.1007/s10637-017-0464-x. Erratum in: Invest New Drugs 2017; 35: 537. doi: 10.1007/s10637-017-0475-7 32. Jabbour J, Manana B, Zahreddine A, Saade C, Charafeddine M, Bazarbachi A, et al. Sarcopenic obesity derived from PET/CT predicts mortality in lym- phoma patients undergoing hematopoietic stem cell transplantation. Curr Res Transl Med 2019; 67: 93-9. doi: 10.1016/j.retram.2018.12.001 33. Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, et al. 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 Ageing 2019; 48: 601. doi: 10.1093/ageing/afz046. Erratum for: Age Ageing 2019; 48: 16-31. doi: 10.1093/ageing/afy169 34. Mei KL, Batsis JA, Mills JB, Holubar SD. Sarcopenia and sarcopenic obesity: do they predict inferior oncologic outcomes after gastrointestinal cancer surgery? Perioper Med 2016; 5: 30. doi: 10.1186/s13741-016-0052-1 35. Prado CM, Heymsfield SB. Lean tissue imaging: a new era for nutritional as- sessment and intervention. JPEN J Parenter Enteral Nutr 2014; 38: 940-53. doi: 10.1177/0148607114550189. Erratum in: JPEN J Parenter Enteral Nutr 2016; 40: 742. doi: 10.1177/0148607116647919 36. Donini LM, Busetto L, Bauer JM, Bischoff S, Boirie Y, Cederholm T, et al. Critical appraisal of definitions and diagnostic criteria for sarcopenic obesity based on a systematic review. Clin Nutr 2020; 39: 2368-88. doi: 10.1016/j. clnu.2019.11.024 37. Ji T , Li Y , Ma L. Sarcopenic obesity: an emerging public health problem. Aging Dis 2022; 13: 379-88. doi: 10.14336/AD.2021.1006 38. Batsis JA, Villareal DT . Sarcopenic obesity in older adults: aetiology, epidemi- ology and treatment strategies. Nat Rev Endocrinol 2018; 14: 513-37. doi: 10.1038/s41574-018-0062-9 39. Lynch GM, Murphy CH, Castro EM, Roche HM. Inflammation and me- tabolism: the role of adiposity in sarcopenic obesity. Proc Nutr Soc 2020; 16: 1-13. doi: 10.1017/S0029665120007119 40. Guo A, Li K, Xiao Q. Sarcopenic obesity: myokines as potential diagnostic biomarkers and therapeutic targets? Exp Gerontol 2020; 139: 111022. doi: 10.1016/j.exger.2020.111022 41. Murton AJ, Marimuthu K, Mallinson JE, Selby AL, Smith K, Rennie MJ, et al. Obesity appears to be associated with altered muscle protein synthetic and breakdown responses to increased nutrient delivery in older men, but not reduced muscle mass or contractile function. Diabetes 2015; 64: 3160-71. doi: 10.2337/db15-0021 42. Abbass T, Dolan RD, Laird BJ, McMillan DC. The relationship between imaging-based body composition analysis and the systemic inflammatory response in patients with cancer: A systematic review. Cancers 2019; 11: 1304. doi: 10.3390/cancers11091304 43. Fouladiun M, Körner U, Bosaeus I, Daneryd P, Hyltander A, Lundholm KG. Body composition and time course changes in regional distribution of fat and lean tissue in unselected cancer patients on palliative care − cor - relations with food intake, metabolism, exercise capacity, and hormones. Cancer 2005; 103: 2189-98. doi: 10.1002/cncr.21013 44. Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. Nat Rev Dis Primers 2018; 4: 17105. doi: 10.1038/nrdp.2017.105 45. Baracos VE, Arribas L. Sarcopenic obesity: hidden muscle wasting and its impact for survival and complications of cancer therapy. Ann Oncol 2018; 29(Suppl 2): ii1-9. doi: 10.1093/annonc/mdx810 46. Murphy RA, Mourtzakis M, Chu QS, Reiman T, Mazurak VC. Skeletal muscle depletion is associated with reduced plasma (n-3) fatty acids in non-small cell lung cancer patients. J Nutr 2010;140: 1602-6. doi: 10.3945/ jn.110.123521 47. Martin L, Gioulbasanis I, Senesse P, Baracos VE. Cancer-associated malnu- trition and CT-defined sarcopenia and myosteatosis are endemic in over- weight and obese patients. JPEN J Parenter Enteral Nutr 2020; 44: 227-38. doi: 10.1002/jpen.1597 48. Correa-de-Araujo R, Addison O, Miljkovic I, Goodpaster BH, Bergman BC, Clark RV, et al. Myosteatosis in the context of skeletal muscle function defi- cit: an interdisciplinary workshop at the National Institute on Aging. Front Physiol 2020; 11: 963. doi: 10.3389/fphys.2020.00963 49. Gemmink A, Goodpaster BH, Schrauwen P , Hesselink MKC. Intramyocellular lipid droplets and insulin sensitivity, the human perspective. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862(10 Pt B): 1242-9. doi: 10.1016/j.bbalip.2017.07.010 50. Maltais A, Alméras N, Lemieux I, Tremblay A, Bergeron J, Poirier P, et al. Trunk muscle quality assessed by computed tomography: association with adiposity indices and glucose tolerance in men. Metabolism 2018; 85: 205- 12. doi: 10.1016/j.metabol.2018.04.003 51. Rollins KE, Tewari N, Ackner A, Awwad A, Madhusudan S, Macdonald IA, et al. The impact of sarcopenia and myosteatosis on outcomes of unresecT- able pancreatic cancer or distal cholangiocarcinoma. Clin Nutr 2016; 35: 1103-9. doi: 10.1016/j.clnu.2015.08.005 52. Knapp ML, al-Sheibani S, Riches PG, Hanham IW, Phillips RH. Hormonal fac- tors associated with weight loss in patients with advanced breast cancer . Ann Clin Biochem 1991; 28 (Pt 5): 480-6. doi: 10.1177/000456329102800510 53. Crown AL, Cottle K, Lightman SL, Falk S, Mohamed-Ali V, Armstrong L, et al. What is the role of the insulin-like growth factor system in the pathophysiol- ogy of cancer cachexia, and how is it regulated? Clin Endocrinol 2002; 56: 723-33. doi: 10.1046/j.1365-2265.2002.01540.x 54. Yoshikawa T, Noguchi Y , Doi C, Makino T, Nomura K. Insulin resistance in pa- tients with cancer: relationships with tumor site, tumor stage, body-weight loss, acute-phase response, and energy expenditure. Nutrition 2001; 17: 590-3. doi: 10.1016/s0899-9007(01)00561-5 55. Hoffmann C, Weigert C. Skeletal muscle as an endocrine organ: the role of myokines in exercise adaptations. Cold Spring Harb Perspect Med 2017; 7: a029793. doi: 10.1101/cshperspect.a029793 56. Jurdana M. Physical activity and cancer risk. Actual knowledge and pos- sible biological mechanisms. Radiol Oncol 2021; 55: 7-17. doi: 10.2478/ raon-2020-0063 57. Rivas DA, McDonald DJ, Rice NP, Haran PH, Dolnikowski GG, Fielding RA. Diminished anabolic signaling response to insulin induced by intramuscular lipid accumulation is associated with inflammation in aging but not obesity. Am J Physiol Regul Integr Comp Physiol 2016; 310: R561-9. doi: 10.1152/ ajpregu.00198.2015 Radiol Oncol 2024; 58(1): 1-8. Jurdana M et al. / Sarcopenic obesity and cancer 8 58. Yamauchi T , Kamon J, Minokoshi Y , Ito Y , Waki H, Uchida S, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP- activated protein kinase. Nat Med 2002; 8: 1288-95. doi: 10.1038/nm788 59. Hong SH, Choi KM. Sarcopenic obesity, insulin resistance, and their implica- tions in cardiovascular and metabolic consequences. Int J Mol Sci 2020; 21: 494. doi: 10.3390/ijms21020494 60. Hamrick MW . Role of the cytokine-like hormone leptin in muscle-bone cross- talk with aging. J Bone Metab 2017; 24: 1-8. doi: 10.11005/jbm.2017.24.1.1 61. Kurdiova T , Balaz M, Vician M, Maderova D, Vlcek M, Valkovic L, et al. Effects of obesity, diabetes and exercise on Fndc5 gene expression and irisin release in human skeletal muscle and adipose tissue: in vivo and in vitro studies. J Physiol 2014; 592: 1091-107. doi: 10.1113/jphysiol.2013.264655 62. Johns N, Greig C, Fearon KC. Is tissue cross-talk important in cancer cachex- ia? Crit Rev Oncog 2012; 17: 263-76. doi: 10.1615/critrevoncog.v17.i3.40 63. Argilés JM, Busquets S, Stemmler B, López-Soriano FJ. Cancer cachexia: understanding the molecular basis. Nat Rev Cancer 2014; 14: 754-62. doi: 10.1038/nrc3829 64. Prado CM, Baracos VE, McCargar LJ, Reiman T, Mourtzakis M, Tonkin K, et al. Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment. Clin Cancer Res 2009; 15: 2920-6. doi: 10.1158/1078-0432. CCR-08-2242 65. Petrelli F, Cortellini A, Indini A, Tomasello G, Ghidini M, Nigro O, et al. Association of obesity with survival outcomes in patients with cancer: a systematic review and meta-analysis. JAMA Netw Open 2021; 4: e213520 doi: 10.1001/jamanetworkopen.2021.3520 66. Gallo M, Adinolfi V, Barucca V, Prinzi N, Renzelli V, Barrea L, et al. Expected and paradoxical effects of obesity on cancer treatment response. Rev Endocr Metab Disord 2021; 22: 681-702. doi: 10.1007/s11154-020-09597-y 67. Prado CM, Purcell SA, Laviano A. Nutrition interventions to treat low mus- cle mass in cancer. J Cachexia Sarcopenia Muscle 2020; 11: 366-80. doi: 10.1002/jcsm.12525 68. Argilés JM, Busquets S, López-Soriano FJ, Costelli P, Penna F. Are there any benefits of exercise training in cancer cachexia? J Cachexia Sarcopenia Muscle 2012; 3: 73-6. doi: 10.1007/s13539-012-0067-5 69. Stene GB, Helbostad JL, Balstad TR, Riphagen II, Kaasa S, Oldervoll LM. Effect of physical exercise on muscle mass and strength in cancer patients during treatment--a systematic review. Crit Rev Oncol Hematol 2013; 88: 573-93. doi: 10.1016/j.critrevonc.2013.07.001 70. Galvão DA, Newton RU. Review of exercise intervention studies in cancer patients. J Clin Oncol 2005; 23: 899-909. doi: 10.1200/JCO.2005.06.085 71. Poggiogalle E, Parrinello E, Barazzoni R, Busetto L, Donini LM. Therapeutic strategies for sarcopenic obesity: a systematic review. Curr Opin Clin Nutr Metab Care 2021; 24: 33-41. doi: 10.1097/MCO.0000000000000714 72. Verschueren SM, Bogaerts A, Delecluse C, Claessens AL, Haentjens P, Vanderschueren D, et al. The effects of whole-body vibration training and vitamin D supplementation on muscle strength, muscle mass, and bone density in institutionalized elderly women: a 6-month randomized, con- trolled trial. J Bone Miner Res 2011; 26: 42-9. doi: 10.1002/jbmr 73. Gyergyek, A, Rotovnik Kozjek N, Klen, J. Monitoring the effect of periopera- tive nutritional care on body composition and functional status in patients with carcinoma of gastrointestinal and hepatobiliary system and pancreas. Radiol Oncol 2023; 57: 371-9. doi: 10.2478/raon-2023-0028