검색
검색 팝업 닫기

Advanced search

Article

Split Viewer

Review Article

J Hosp Palliat Care 2025; 28(1): 10-17

Published online March 1, 2025 https://doi.org/10.14475/jhpc.2025.28.1.10

Copyright © Journal of Hospice and Palliative Care.

Sarcopenia in Terminally Ill Patients with Cancer: Clinical Implications, Diagnostic Challenges, and Management Strategies

Se-Il Go , Myoung Hee Kang , Hoon-Gu Kim

Division of Hematology-Oncology, Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea

Correspondence to:Hoon-Gu Kim
ORCID: https://orcid.org/0009-0003-4521-9620
E-mail: sharp-khg@hanmail.net
1These authors contributed equally to this work.

Received: February 9, 2025; Revised: February 14, 2025; Accepted: February 18, 2025

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Sarcopenia, characterized by progressive loss of skeletal muscle mass and strength, is a prevalent but often overlooked condition in patients with cancer who are terminally ill. It contributes to functional decline, increased symptom burden, and reduced quality of life, yet remains underrecognized in palliative care. Diagnosing sarcopenia in this population is challenging because conventional imaging techniques are often impractical. Instead, alternative assessments, such as the Strength, Assistance with walking, Rise from a chair, Climb stairs, and Falls questionnaire (SARC-F), anthropometric measurements, and bioelectrical impedance analysis offer feasible options. Management should focus on symptom relief, functional preservation, and patient comfort, rather than on muscle mass restoration. Nutritional support must be tailored to prognosis, with aggressive interventions generally avoided during end-of-life care. Although exercise may help to maintain mobility and alleviate symptoms, its feasibility is often limited. Pharmacological interventions, including appetite stimulants and anti-cachexia agents, remain largely investigational, with insufficient evidence for routine use in palliative care. Future research should refine sarcopenia assessment methods and develop patient-centered interventions that align with palliative care principles, emphasizing quality of life and individualized needs.

Keywords: Sarcopenia, Palliative care, Cachexia, Terminally ill

Sarcopenia is a condition characterized by progressive and generalized loss of skeletal muscle mass and strength, leading to physical disability, poor quality of life (QoL), and increased mortality [1]. In patients with cancer, sarcopenia is caused by various factors, including the cancer itself, treatment-related adverse events, and reduced physical activity [2,3]. Sarcopenia in patients with cancer is associated with shorter survival, increased treatment-related toxicity, higher postoperative complications, reduced physical function, and decreased QoL [4-6].

The prevalence of sarcopenia in patients with cancer has been widely reported, with rates ranging from 11% to 74%, depending on the cancer type and stage [7]. In terminally ill patients with cancer, sarcopenia is nearly universal due to progressive catabolism and systemic inflammation [8]. Despite the well-established prognostic significance of sarcopenia in advanced cancer, evidence addressing sarcopenia, specifically in terminally ill patients, remains limited. In this review, we aim to investigate the clinical implications of sarcopenia in terminally ill patients with cancer, focusing on its impact on prognosis, functional decline, symptom burden, and QoL. Additionally, we discuss the diagnostic challenges and potential management of sarcopenia in this population.

Sarcopenia has been consistently identified as a strong prognostic factor in patients with cancer, with a greater impact in terminally ill patients. A prospective study investigating nutritional indices in terminally ill patients found that poor nutritional status was significantly associated with worse survival. Specifically, low triceps skinfold thickness was associated with a 2.9-fold increase in mortality risk [9]. Similarly, a prospective cohort study of 727 patients with terminal cancer showed that muscle power, assessed using the Medical Research Council scale, was a predictor of imminent death within a week of admission to hospice care. In the most accurate prediction model, lower mean muscle power was associated with an increased risk of imminent death within a week [10]. Another prospective observational study of 628 patients with advanced cancer in a palliative care outpatient unit reported that the phase angle measured by bioelectrical impedance analysis (BIA) was a key indicator for detecting sarcopenia and a significant predictor of shorter survival (hazard ratio, 2.3) [11]. A prospective observational study of 334 patients with advanced cancer in a palliative care unit assessed the relationship among sarcopenia, nutritional status, and overall survival using mid-upper arm muscle area (MUAMA), calf circumference (CC), and appendicular skeletal muscle mass. In this population, sarcopenia assessed using MUAMA (43 vs. 67 days) and CC (44 vs. 77 days) was significantly associated with shorter survival [12]. Additionally, a retrospective cohort study of 304 patients with cancer receiving palliative care evaluated the prognostic value of the Strength, Assistance with walking, Rise from a chair, Climb stairs, and Falls (SARC-F), a self-report questionnaire assessing muscle strength and physical function. The study found that patients with SARC-F scores ≥4 had significantly worse overall survival (40 vs. 121 days) [13]. Collectively, these studies have suggested the significant prognostic value of sarcopenia in terminally ill patients with cancer, as evaluated through various clinical, functional, and nutritional parameters.

Sarcopenia significantly contributes to the functional decline and deterioration of QoL in terminally ill patients with cancer. Progressive muscle wasting leads to reduced physical strength, impaired mobility, and increased risk of falls, ultimately resulting in greater dependence on caregivers. In this population, the impact of sarcopenia extends beyond physical limitations and affects psychological well-being, social interactions, and overall comfort during end-of-life care.

Several studies have demonstrated an association between sarcopenia and worsening physical function. A cross-sectional study of 135 patients with advanced cancer receiving home palliative care found that 77% had sarcopenia (SARC-F ≥4) and 68% were malnourished. Sarcopenia was found to be potentially associated with malnutrition [14]. A retrospective cohort study of 304 patients receiving palliative care found that those with higher SARC-F scores (≥4) experienced significantly greater difficulty in performing daily activities. Compared with patients with lower SARC-F scores (<4), those with SARC-F scores ≥4 were far more likely to have impaired strength (100% vs. 43.6%), require walking assistance (98.3% vs. 3.7%), struggle with rising from a chair (90.5% vs. 3.2%), and have difficulty climbing stairs (98.3% vs. 34.0%) [13]. In addition to impairing mobility and increasing dependence, sarcopenia exacerbates overall functional decline in terminally ill patients with cancer. A prospective study involving 1,660 patients with incurable cancer at the Palliative Care Unit of the Brazilian National Cancer Institute demonstrated that lower muscle mass and strength, assessed using MUAMA and handgrip strength (HGS), were strongly associated with worse performance status. Patients in the sarcopenia group had a median Karnofsky Performance Status of 40% compared with 60% in those with better muscle mass phenotypes. Notably, Patient-Generated Subjective Global Assessment Short Form (PG-SGA SF) scores, which reflect nutritional deterioration and symptom burden, also differed significantly according to sarcopenia severity and sex. Among men, those in the sarcopenia group had a median PG-SGA SF score of 15, compared with a score of 10 in those with better muscle mass phenotypes. Among women, the difference was even more pronounced, with PG-SGA SF scores of 21 in the sarcopenia group compared with a score of 13 in those with better muscle mass phenotypes [15].

Sarcopenia not only affects physical function but also significantly worsens QoL and symptom burden in patients receiving palliative care. A cross-sectional study of 770 patients with incurable cancer demonstrated that a worsening muscle mass phenotype, as assessed using the MUAMA and HGS, was strongly associated with declines in multiple QoL domains. Compared with patients without sarcopenia, those classified as having sarcopenia had significantly worse scores in physical function (9.1 vs. 32.2), emotional well-being (57.4 vs. 61.1), fatigue (59.9 vs. 44.7), and global health (50.2 vs. 57.5). Additionally, patients with sarcopenia were more likely to experience insomnia (odds ratio [OR] 3.11), fatigue (OR 1.83), and loss of appetite (OR 1.40), further exacerbating symptom burden and reducing overall well-being [16].

Collectively, sarcopenia progression leads to greater physical disability, worsening fatigue, increased nutritional deficits, and a higher prevalence of distressing symptoms. The interplay among muscle loss, malnutrition, and symptom burden further accelerates the decline in functional independence and overall well-being.

Sarcopenia is typically diagnosed using imaging-based methods, body composition analysis, and functional assessment in patients with cancer undergoing surgery or chemotherapy [17]. Computed tomography (CT)-based muscle mass assessment is often used, as routine CT for cancer staging and response evaluation provides an opportunity to assess skeletal muscle depletion, particularly at the L3 level, or in some cases, using the pectoralis muscle index [18-21]. Other methods, such as dual-energy X-ray absorptiometry and BIA, allow whole-body muscle mass estimation [22,23]. In contrast, functional tests, such as the HGS and 6-minute walk test, assess muscle strength and physical performance [24,25].

In terminally ill patients with cancer, sarcopenia assessment is often challenging owing to less frequent routine imaging and a shift in focus from tumor response evaluation to symptom management. Systemic inflammation, cancer-associated cachexia, and prolonged bedridden status further limit the feasibility of advanced imaging and functional testing. Instead, diagnosis in this population requires noninvasive, bedside-friendly approaches prioritizing functional and nutritional status over detailed body composition analysis. The primary goal is not early detection for intervention but symptom monitoring and maintaining QoL.

The SARC-F questionnaire is a widely used screening tool for sarcopenia that evaluates strength, walking ability, ability to rise from a chair, climbing stairs, and history of falls. It is quick, noninvasive, and requires no equipment, making it ideal for palliative care settings [26]. Anthropometric measurements provide simple and accessible estimates of muscle mass and function. HGS, assessed using a dynamometer, serves as a surrogate marker for muscle strength and overall functional capacity and has been associated with frailty, functional decline, and decreased QoL in terminally ill patients with cancer [15,16]. MUAMA, calculated using the mid-upper arm circumference and triceps skinfold thickness, is an indicator of muscle wasting and nutritional status [12,15,16]. Similarly, the CC serves as an estimate of appendicular muscle mass and is correlated with frailty and physical function in older adults receiving palliative care [12,27]. BIA is a noninvasive and rapid method for evaluating skeletal muscle mass and hydration status [28]. Owing to its portability and ease of use, BIA can be performed at the bedside, making it a feasible alternative for sarcopenia assessment in patients who are bedridden or for whom imaging-based evaluations are inappropriate. However, the interpretation may be affected by fluid imbalances and systemic inflammation, which are common in this population [29]. Although each method has limitations, a combined approach may provide the most practical assessment in palliative care.

In terminally ill patients, given their progressive catabolism and limited life expectancy, aggressive interventions provide little benefit and may increase discomfort [30]. Instead, care should prioritize nutritional support, maintenance of mobility, and symptom control tailored to the patient’s prognosis and goals. It is also important to consider that most available evidence does not clearly distinguish among anorexia, cachexia, and sarcopenia, making it essential to interpret management strategies with caution. Although these conditions share overlapping features, their underlying mechanisms and clinical implications differ, necessitating individualized approaches to palliative care.

1. Nutritional support

According to the American Society of Clinical Oncology guidelines, nutritional interventions in palliative care should prioritize maintaining energy intake, minimizing symptom burden, and preserving QoL [31]. According to the European Society for Clinical Nutrition and Metabolism clinical guidelines, indications for nutritional support in patients with advanced cancer should be guided by their estimated life expectancy and clinical situation. For patients with an expected survival time of several months or more, oral nutritional support and parenteral nutrition (PN) may be considered in select cases. For patients with a life expectancy of several weeks, nutritional interventions should be noninvasive, focusing primarily on symptom relief and psychosocial support. For patients with an expected survival of <7 days, total PN is generally avoided because it does not improve outcomes and may cause metabolic distress, potentially increasing discomfort. Instead, care should focus on oral comfort measures, hydration as needed, and minimizing interventions that may compromise QoL [32]. Some nutritional supplements, such as a combination therapy of β-hydroxy-β-methylbutyrate, arginine, and glutamine, have shown potential benefits in increasing lean body mass in patients with cancer [33]. However, most studies have focused on patients receiving disease-directed treatment with limited evidence supporting its use in terminally ill patients [34].

2. Exercise and physical activity

Exercise interventions in palliative care settings have been explored as potential strategies to improve physical function, fatigue, and QoL in patients with advanced cancer. A systematic review and meta-analysis of 14 studies, including 1,034 adults receiving palliative care, found that structured exercise interventions significantly improved exercise capacity, reduced pain and fatigue, and enhanced overall QoL [35]. Similarly, a study assessing the feasibility of a structured two-week exercise program in a palliative care unit found that, despite recruitment challenges, exercise was well tolerated and led to short-term improvements in physical function and QoL [36]. However, various clinical factors, including disease progression, physical function, and patient preferences, may pose hurdles to exercise in palliative care. A systematic review that assessed different exercise interventions, including aerobic, resistance, and combined training reported mixed results in patients with advanced cancer receiving palliative care. While resistance-based exercises improved at least one functional outcome in most studies, overall adherence was highly variable [37]. Despite these limitations, exercise remains a promising intervention that aligns with palliative care goals by enhancing mobility and reducing symptom burden. Feasibility depends on individualized adaptation, patient preferences, and practical considerations such as home-based or remotely supervised regimens. Given the heterogeneity of available studies, further research is needed to establish evidence-based exercise recommendations tailored for terminally ill patients receiving palliative care.

3. Pharmacological interventions

While nutritional support and physical activity remain central to sarcopenia management, pharmacological interventions have been explored to stimulate appetite, reduce inflammation, and mitigate muscle loss in patients with cancer. However, strong evidence supporting their routine use in terminally ill patients or palliative care settings remains scarce. Most studies on appetite stimulants and anti-cachexia therapies have been conducted on patients receiving disease-directed treatment, with limited data on their efficacy and safety in palliative care. Megestrol acetate, a progestin-derived appetite stimulant, has been shown to increase appetite and body weight in patients with cancer; however, its impact on muscle preservation and physical function is unclear [38]. A recent study reported that megestrol acetate showed a potential survival benefit in patients with metastatic gastric cancer who did not receive palliative chemotherapy. However, in multivariate analysis, the impact on survival was not statistically significant in this population. Moreover, the risks of thromboembolism, adrenal insufficiency, and fluid retention limit its use in palliative care, where symptom control and patient comfort are paramount [39]. Some studies have suggested that corticosteroids can temporarily improve appetite and energy levels, offering short-term relief from severe anorexia and fatigue [40]. However, prolonged use accelerates muscle wasting and exacerbates sarcopenia rather than mitigating it [41,42]. Adverse effects, including hyperglycemia, immunosuppression, and osteoporosis, often outweigh the potential benefits [43]. Given these risks, corticosteroids should be cautiously prescribed, prioritizing short-term symptom relief over long-term sarcopenia management.

Inflammation plays a central role in cachexia and sarcopenia, with elevated levels of proinflammatory cytokines contributing to muscle catabolism [44]. Several anti-inflammatory agents have been investigated in patients with cancer, primarily in the context of cancer cachexia rather than in sarcopenia. A systematic review assessed the efficacy and safety of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) in patients with cancer cachexia. The review included five randomized controlled trials focusing on indomethacin, ibuprofen, and celecoxib. The findings indicated that there is inadequate evidence to recommend any NSAID for cancer cachexia, whereas celecoxib is generally safe for use [45]. Beyond NSAIDs, other anti-inflammatory agents have been assessed. Thalidomide, an immunomodulatory drug with anti-tumor necrosis factor-α properties, has been investigated in cancer cachexia. A small randomized controlled trial found that improvements in symptoms and cytokine levels did not differ significantly between the thalidomide and placebo groups [46]. A systematic review further highlighted the lack of strong evidence supporting thalidomide for cancer cachexia, with high heterogeneity across studies and poor tolerance in some patients [47]. Similarly, investigational cytokine-targeting therapies have yielded inconsistent results. Although tocilizumab, an interleukin-6 receptor antagonist, has shown potential benefits in reducing inflammatory markers and improving body weight in patients with lung cancer, its effectiveness against cancer cachexia and sarcopenia remains inconclusive and requires further validation [48]. Other investigational anti-inflammatory agents such as infliximab and etanercept have failed to demonstrate efficacy in treating cancer, cachexia, and sarcopenia [49,50]. However, the roles of these anti-inflammatory agents in palliative care settings remain largely unknown. Considering the frailty of this population, the potential harm caused by anti-inflammatory therapies, including immunosuppression, thromboembolic risk, and other toxicities, should be carefully weighed against their uncertain benefits.

Recent advances in cancer cachexia and sarcopenia research have led to the development of novel pharmacological agents. Anamorelin, a selective ghrelin receptor agonist, has been shown in ROMANA 1 and 2 trials to increase appetite, body weight, and lean body mass in patients with advanced cancer [51]. A recent study suggested that ponsegromab, a humanized monoclonal antibody targeting Growth Differentiation Factor 15 (GDF15), was associated with improved weight, appetite, and physical activity in patients with cancer cachexia [52]. Despite promising early results, the role of ghrelin receptor agonist and anti-GDF15 therapy in palliative care has not yet been established. Further studies are required to determine whether these therapies can provide meaningful benefits beyond weight gain in patients receiving palliative care.

Sarcopenia is a prevalent but underexplored condition in terminally ill patients with cancer that contributes to functional decline and reduced QoL. Management should prioritize symptom relief, functional preservation, and patient comfort. Although nutritional and exercise interventions present challenges, they can improve mobility and well-being through individualized approaches. Current pharmacological options are limited; however, ongoing research on emerging therapies may provide future treatment options. A multidisciplinary approach integrating palliative care, nutritional support, and rehabilitation may optimize symptom control and enhance QoL in this vulnerable population. Future research should refine assessment methods and develop patient-centered, noninvasive strategies that align with palliative care principles.

Study conception and design: SG, HK. Data acquisition: SG, MHK, HK. Data analysis and interpretation: SG, MHK, HK. Manuscript drafting: SG, MHK, HK. Critical manuscript revision: SG, MHK, HK. Final manuscript approval: SG, MHK, HK.

  1. Muscaritoli M, Anker SD, Argiles J, Aversa Z, Bauer JM, Biolo G, et al. Consensus definition of sarcopenia, cachexia and pre-cachexia: joint document elaborated by Special Interest Groups (SIG) "cachexia-anorexia in chronic wasting diseases" and "nutrition in geriatrics". Clin Nutr 2010;29:154-9.
    Pubmed CrossRef
  2. Oflazoglu U, Alacacioglu A, Varol U, Kucukzeybek Y, Salman T, Taskaynatan H, et al. Chemotherapy-induced sarcopenia in newly diagnosed cancer patients: Izmir Oncology Group (IZOG) study. Support Care Cancer 2020;28:2899-910.
    Pubmed CrossRef
  3. Albini A, La Vecchia C, Magnoni F, Garrone O, Morelli D, Janssens JP, et al. Physical activity and exercise health benefits: cancer prevention, interception, and survival. Eur J Cancer Prev 2025;34:24-39.
    Pubmed CrossRef
  4. Williams GR, Dunne RF, Giri S, Shachar SS, Caan BJ. Sarcopenia in the older adult with cancer. J Clin Oncol 2021;39:2068-78.
    Pubmed KoreaMed CrossRef
  5. Luo L, Shen X, Fang S, Wan T, Liu P, Li P, et al. Sarcopenia as a risk factor of progression-free survival in patients with metastases: a systematic review and meta-analysis. BMC Cancer 2023;23:127.
    Pubmed KoreaMed CrossRef
  6. Go SI, Choi BH, Park MJ, Park S, Kang MH, Kim HG, et al. Prognostic impact of pretreatment skeletal muscle index and CONUT score in diffuse large B-cell Lymphoma. BMC Cancer 2023;23:1071.
    Pubmed KoreaMed CrossRef
  7. Shachar SS, Williams GR, Muss HB, Nishijima TF. Prognostic value of sarcopenia in adults with solid tumours: A meta-analysis and systematic review. Eur J Cancer 2016;57:58-67.
    Pubmed CrossRef
  8. Xia Z, Cholewa J, Zhao Y, Shang HY, Yang YQ, Araujo Pessoa K, et al. Targeting inflammation and downstream protein metabolism in sarcopenia: A brief up-dated description of concurrent exercise and leucine-based multimodal intervention. Front Physiol 2017;8:434.
    Pubmed KoreaMed CrossRef
  9. Ho SY, Guo HR, Chen HH, Peng CJ. Nutritional predictors of survival in terminally ill cancer patients. J Formos Med Assoc 2003;102:544-50.
  10. Chiang JK, Cheng YH, Koo M, Kao YH, Chen CY. A computer-assisted model for predicting probability of dying within 7 days of hospice admission in patients with terminal cancer. Jpn J Clin Oncol 2010;40:449-55.
    Pubmed KoreaMed CrossRef
  11. Perez Camargo DA, Allende Perez SR, Verastegui Aviles E, Rivera Franco MM, Meneses Garcia A, Herrera Gomez A, et al. Assessment and impact of phase angle and sarcopenia in palliative cancer patients. Nutr Cancer 2017;69:1227-33.
    Pubmed CrossRef
  12. da Silva JR, Wiegert EVM, Oliveira L, Calixto-Lima L; Jr. Different methods for diagnosis of sarcopenia and its association with nutritional status and survival in patients with advanced cancer in palliative care. Nutrition 2019;60:48-52.
    Pubmed CrossRef
  13. Mori N, Maeda K, Fukami Y, Matsuyama R, Nonogaki T, Kato R, et al. High SARC-F score predicts poor survival of patients with cancer receiving palliative care. Support Care Cancer 2022;30:4065-72.
    Pubmed CrossRef
  14. Mercadante S, Bellavia GM, Fusco F, Adamoli L, Scibilia C, Lo Cascio A, et al. Malnutrition is associated with fatigue and anxiety in advanced cancer patients admitted to home palliative care. Am J Hosp Palliat Care :10499091241278924. Epup 2024 Aug 27.
    Pubmed CrossRef
  15. Calixto-Lima L, Souza-Silva R, Oliveira LC, Chaves GV, Wiegert EVM. Development and validation of a grading system for assessing muscle mass phenotype using mid-upper arm muscle area and handgrip strength in patients with incurable cancer. Nutr Clin Pract 2022;37:1385-99.
    Pubmed CrossRef
  16. Wiegert EVM, Oliveira LC, Rosa KSDC, Calixto-Lima L. Association of the muscle mass phenotype as assessed by a grading system with the quality of life of patients with incurable cancer in palliative care. Clin Nutr ESPEN 2024;62:216-23.
    Pubmed CrossRef
  17. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 2011;12:489-95.
    Pubmed CrossRef
  18. Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, et al. Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol 2008;9:629-35.
    Pubmed CrossRef
  19. Go SI, Kim HG, Kang MH, Park S, Lee GW. Prognostic model based on the geriatric nutritional risk index and sarcopenia in patients with diffuse large B-cell lymphoma. BMC Cancer 2020;20:439.
    Pubmed KoreaMed CrossRef
  20. Park SE, Choi JH, Park JY, Kim BJ, Kim JG, Kim JW, et al. Loss of skeletal muscle mass during palliative chemotherapy is a poor prognostic factor in patients with advanced gastric cancer. Sci Rep 2020;10:17683.
    Pubmed KoreaMed CrossRef
  21. Kim YS, Kim EY, Kang SM, Ahn HK, Kim HS. Single cross-sectional area of pectoralis muscle by computed tomography - correlation with bioelectrical impedance based skeletal muscle mass in healthy subjects. Clin Physiol Funct Imaging 2017;37:507-11.
    Pubmed CrossRef
  22. Kim EY, Kim K, Kim YS, Ahn HK, Jeong YM, Kim JH, et al. Prevalence of and factors associated with sarcopenia in Korean cancer survivors: based on data obtained by the Korea National Health and Nutrition Examination Survey (KNHANES) 2008-2011. Nutr Cancer 2017;69:394-401.
    Pubmed CrossRef
  23. Amano K, Bruera E, Hui D. Diagnostic and prognostic utility of phase angle in patients with cancer. Rev Endocr Metab Disord 2023;24:479-89.
    Pubmed CrossRef
  24. Sakaguchi T, Maeda K, Takeuchi T, Ishida Y, Kato R, Ueshima J, et al. Low handgrip strength as a marker of severity in the diagnostic criteria for cancer cachexia. Clin Nutr ESPEN 2024;64:435-40.
    Pubmed CrossRef
  25. Katakami N, Uchino J, Yokoyama T, Naito T, Kondo M, Yamada K, et al. Anamorelin (ONO-7643) for the treatment of patients with non-small cell lung cancer and cachexia: results from a randomized, double-blind, placebo-controlled, multicenter study of Japanese patients (ONO-7643-04). Cancer 2018;124:606-16.
    Pubmed KoreaMed CrossRef
  26. Malmstrom TK, Morley JE. SARC-F: a simple questionnaire to rapidly diagnose sarcopenia. J Am Med Dir Assoc 2013;14:531-2.
    Pubmed CrossRef
  27. Sousa IM, Bielemann RM, Gonzalez MC, da Rocha IMG, Barbalho ER, de Carvalho ALM, et al. Low calf circumference is an independent predictor of mortality in cancer patients: a prospective cohort study. Nutrition 2020;79-80:110816. Epub 2020 Mar 19.
    Pubmed CrossRef
  28. Branco MG, Mateus C, Capelas ML, Pimenta N, Santos T, Makitie A, et al. Bioelectrical impedance analysis (BIA) for the assessment of body composition in oncology: a scoping review. Nutrients 2023;15:4792.
    Pubmed KoreaMed CrossRef
  29. Lin SQ, Xie HL, Ge YZ, Ruan GT, Zhang Q, Song MM, et al. Association between systemic inflammation and water composition and survival in colorectal cancer. Front Oncol 2022;12:896160. eCollection 2022.
    Pubmed KoreaMed CrossRef
  30. Koroukian SM, Douglas SL, Vu L, Fein HL, Gairola R, Warner DF, et al. Incidence of aggressive end-of-life care among older adults with metastatic cancer living in nursing homes and community settings. JAMA Netw Open 2023;6:e230394.
    Pubmed KoreaMed CrossRef
  31. Roeland EJ, Bohlke K, Baracos VE, Bruera E, Del Fabbro E, Dixon S, et al. Management of cancer cachexia: ASCO guideline. J Clin Oncol 2020;38:2438-53.
    Pubmed CrossRef
  32. Muscaritoli M, Arends J, Bachmann P, Baracos V, Barthelemy N, Bertz H, et al. ESPEN practical guideline: Clinical nutrition in cancer. Clin Nutr 2021;40:2898-913.
    Pubmed CrossRef
  33. Berk L, James J, Schwartz A, Hug E, Mahadevan A, Samuels M, et al. A randomized, double-blind, placebo-controlled trial of a beta-hydroxyl beta-methyl butyrate, glutamine, and arginine mixture for the treatment of cancer cachexia (RTOG 0122). Support Care Cancer 2008;16:1179-88.
    Pubmed CrossRef
  34. Cuhls H, Marinova M, Kaasa S, Stieber C, Conrad R, et al; Mochamat. A systematic review on the role of vitamins, minerals, proteins, and other supplements for the treatment of cachexia in cancer: a European Palliative Care Research Centre cachexia project. J Cachexia Sarcopenia Muscle 2017;8:25-39.
    Pubmed KoreaMed CrossRef
  35. Tanriverdi A, Ozcan Kahraman B, Ergin G, Karadibak D, Savci S. Effect of exercise interventions in adults with cancer receiving palliative care: a systematic review and meta-analysis. Support Care Cancer 2023;31:205.
    Pubmed CrossRef
  36. Schwonke I, Freitag N, Aschendorf P, Wucharz K, Thieme J, Appelmann I, et al. Feasibility of a physical exercise intervention for patients on a palliative care unit: a critical analysis. BMC Palliat Care 2024;23:58.
    Pubmed KoreaMed CrossRef
  37. Rogers-Shepp I, Bhattacharya S, Mennillo HA, Kumar R, Hsieh B, Anandarajah G. Exercise interventions for advanced cancer palliative care patients: A systematic literature review and descriptive evidence synthesis of randomized controlled trials. Palliat Med 2023;37:677-91.
    Pubmed CrossRef
  38. Lim YL, Teoh SE, Yaow CYL, Lin DJ, Masuda Y, Han MX, et al. A systematic review and meta-analysis of the clinical use of megestrol acetate for cancer-related anorexia/cachexia. J Clin Med 2022;11:3756.
    Pubmed KoreaMed CrossRef
  39. Lee CM, Kang JH, Go SI. Clinical implication of megestrol acetate in metastatic gastric cancer: a big data analysis from Health Insurance Review and Assessment (HIRA) database. Support Care Cancer 2024;32:249.
    Pubmed CrossRef
  40. Miller S, McNutt L, McCann MA, McCorry N. Use of corticosteroids for anorexia in palliative medicine: a systematic review. J Palliat Med 2014;17:482-5.
    Pubmed CrossRef
  41. Salehian B, Kejriwal K. Glucocorticoid-induced muscle atrophy: mechanisms and therapeutic strategies. Endocr Pract 1999;5:277-81.
    Pubmed CrossRef
  42. Kuzuya M. Drug-related sarcopenia as a secondary sarcopenia. Geriatr Gerontol Int 2024;24:195-203.
    Pubmed KoreaMed CrossRef
  43. Liu D, Ahmet A, Ward L, Krishnamoorthy P, Mandelcorn ED, Leigh R, et al. A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol 2013;9:30.
    Pubmed KoreaMed CrossRef
  44. Setiawan T, Sari IN, Wijaya YT, Julianto NM, Muhammad JA, Lee H, et al. Cancer cachexia: molecular mechanisms and treatment strategies. J Hematol Oncol 2023;16:54.
    Pubmed KoreaMed CrossRef
  45. Bowers M, Cucchiaro B, Reid J, Slee A. Non-steroidal anti-inflammatory drugs for treatment of cancer cachexia: a systematic review. J Cachexia Sarcopenia Muscle 2023;14:2473-97.
    Pubmed KoreaMed CrossRef
  46. Yennurajalingam S, Willey JS, Palmer JL, Allo J, Del Fabbro E, Cohen EN, et al. The role of thalidomide and placebo for the treatment of cancer-related anorexia-cachexia symptoms: results of a double-blind placebo-controlled randomized study. J Palliat Med 2012;15:1059-64.
    Pubmed KoreaMed CrossRef
  47. Reid J, Mills M, Cantwell M, Cardwell CR, Murray LJ, Donnelly M. Thalidomide for managing cancer cachexia. Cochrane Database Syst Rev 2012;2012:CD008664.
    Pubmed KoreaMed CrossRef
  48. Du Y, Liu XY, Pan RL, Zhang XT, Si XY, Chen MJ, et al. Tocilizumab for advanced non-small-cell lung cancer with concomitant cachexia: an observational study. J Cachexia Sarcopenia Muscle 2024;15:2815-25.
    Pubmed KoreaMed CrossRef
  49. Jatoi A, Ritter HL, Dueck A, Nguyen PL, Nikcevich DA, Luyun RF, et al. A placebo-controlled, double-blind trial of infliximab for cancer-associated weight loss in elderly and/or poor performance non-small cell lung cancer patients (N01C9). Lung Cancer 2010;68:234-9.
    Pubmed KoreaMed CrossRef
  50. Jatoi A, Dakhil SR, Nguyen PL, Sloan JA, Kugler JW, Rowland KM Jr, et al. A placebo-controlled double blind trial of etanercept for the cancer anorexia/weight loss syndrome: results from N00C1 from the North Central Cancer Treatment Group. Cancer 2007;110:1396-403.
    Pubmed CrossRef
  51. Temel JS, Abernethy AP, Currow DC, Friend J, Duus EM, Yan Y, et al. Anamorelin in patients with non-small-cell lung cancer and cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double-blind, phase 3 trials. Lancet Oncol 2016;17:519-31.
    Pubmed CrossRef
  52. Groarke JD, Crawford J, Collins SM, Lubaczewski S, Roeland EJ, Naito T, et al. Ponsegromab for the treatment of cancer cachexia. N Engl J Med 2024;391:2291-303.
    Pubmed CrossRef

Article

Review Article

J Hosp Palliat Care 2025; 28(1): 10-17

Published online March 1, 2025 https://doi.org/10.14475/jhpc.2025.28.1.10

Copyright © Journal of Hospice and Palliative Care.

Sarcopenia in Terminally Ill Patients with Cancer: Clinical Implications, Diagnostic Challenges, and Management Strategies

Se-Il Go , Myoung Hee Kang , Hoon-Gu Kim

Division of Hematology-Oncology, Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea

Correspondence to:Hoon-Gu Kim
ORCID: https://orcid.org/0009-0003-4521-9620
E-mail: sharp-khg@hanmail.net
1These authors contributed equally to this work.

Received: February 9, 2025; Revised: February 14, 2025; Accepted: February 18, 2025

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Sarcopenia, characterized by progressive loss of skeletal muscle mass and strength, is a prevalent but often overlooked condition in patients with cancer who are terminally ill. It contributes to functional decline, increased symptom burden, and reduced quality of life, yet remains underrecognized in palliative care. Diagnosing sarcopenia in this population is challenging because conventional imaging techniques are often impractical. Instead, alternative assessments, such as the Strength, Assistance with walking, Rise from a chair, Climb stairs, and Falls questionnaire (SARC-F), anthropometric measurements, and bioelectrical impedance analysis offer feasible options. Management should focus on symptom relief, functional preservation, and patient comfort, rather than on muscle mass restoration. Nutritional support must be tailored to prognosis, with aggressive interventions generally avoided during end-of-life care. Although exercise may help to maintain mobility and alleviate symptoms, its feasibility is often limited. Pharmacological interventions, including appetite stimulants and anti-cachexia agents, remain largely investigational, with insufficient evidence for routine use in palliative care. Future research should refine sarcopenia assessment methods and develop patient-centered interventions that align with palliative care principles, emphasizing quality of life and individualized needs.

Keywords: Sarcopenia, Palliative care, Cachexia, Terminally ill

INTRODUCTION

Sarcopenia is a condition characterized by progressive and generalized loss of skeletal muscle mass and strength, leading to physical disability, poor quality of life (QoL), and increased mortality [1]. In patients with cancer, sarcopenia is caused by various factors, including the cancer itself, treatment-related adverse events, and reduced physical activity [2,3]. Sarcopenia in patients with cancer is associated with shorter survival, increased treatment-related toxicity, higher postoperative complications, reduced physical function, and decreased QoL [4-6].

The prevalence of sarcopenia in patients with cancer has been widely reported, with rates ranging from 11% to 74%, depending on the cancer type and stage [7]. In terminally ill patients with cancer, sarcopenia is nearly universal due to progressive catabolism and systemic inflammation [8]. Despite the well-established prognostic significance of sarcopenia in advanced cancer, evidence addressing sarcopenia, specifically in terminally ill patients, remains limited. In this review, we aim to investigate the clinical implications of sarcopenia in terminally ill patients with cancer, focusing on its impact on prognosis, functional decline, symptom burden, and QoL. Additionally, we discuss the diagnostic challenges and potential management of sarcopenia in this population.

SARCOPENIA AS A PROGNOSTIC INDICATOR IN TERMINALLY ILL PATIENTS

Sarcopenia has been consistently identified as a strong prognostic factor in patients with cancer, with a greater impact in terminally ill patients. A prospective study investigating nutritional indices in terminally ill patients found that poor nutritional status was significantly associated with worse survival. Specifically, low triceps skinfold thickness was associated with a 2.9-fold increase in mortality risk [9]. Similarly, a prospective cohort study of 727 patients with terminal cancer showed that muscle power, assessed using the Medical Research Council scale, was a predictor of imminent death within a week of admission to hospice care. In the most accurate prediction model, lower mean muscle power was associated with an increased risk of imminent death within a week [10]. Another prospective observational study of 628 patients with advanced cancer in a palliative care outpatient unit reported that the phase angle measured by bioelectrical impedance analysis (BIA) was a key indicator for detecting sarcopenia and a significant predictor of shorter survival (hazard ratio, 2.3) [11]. A prospective observational study of 334 patients with advanced cancer in a palliative care unit assessed the relationship among sarcopenia, nutritional status, and overall survival using mid-upper arm muscle area (MUAMA), calf circumference (CC), and appendicular skeletal muscle mass. In this population, sarcopenia assessed using MUAMA (43 vs. 67 days) and CC (44 vs. 77 days) was significantly associated with shorter survival [12]. Additionally, a retrospective cohort study of 304 patients with cancer receiving palliative care evaluated the prognostic value of the Strength, Assistance with walking, Rise from a chair, Climb stairs, and Falls (SARC-F), a self-report questionnaire assessing muscle strength and physical function. The study found that patients with SARC-F scores ≥4 had significantly worse overall survival (40 vs. 121 days) [13]. Collectively, these studies have suggested the significant prognostic value of sarcopenia in terminally ill patients with cancer, as evaluated through various clinical, functional, and nutritional parameters.

IMPACT OF SARCOPENIA ON FUNCTIONAL DECLINE, SYMPTOM BURDEN, AND QUALITY OF LIFE

Sarcopenia significantly contributes to the functional decline and deterioration of QoL in terminally ill patients with cancer. Progressive muscle wasting leads to reduced physical strength, impaired mobility, and increased risk of falls, ultimately resulting in greater dependence on caregivers. In this population, the impact of sarcopenia extends beyond physical limitations and affects psychological well-being, social interactions, and overall comfort during end-of-life care.

Several studies have demonstrated an association between sarcopenia and worsening physical function. A cross-sectional study of 135 patients with advanced cancer receiving home palliative care found that 77% had sarcopenia (SARC-F ≥4) and 68% were malnourished. Sarcopenia was found to be potentially associated with malnutrition [14]. A retrospective cohort study of 304 patients receiving palliative care found that those with higher SARC-F scores (≥4) experienced significantly greater difficulty in performing daily activities. Compared with patients with lower SARC-F scores (<4), those with SARC-F scores ≥4 were far more likely to have impaired strength (100% vs. 43.6%), require walking assistance (98.3% vs. 3.7%), struggle with rising from a chair (90.5% vs. 3.2%), and have difficulty climbing stairs (98.3% vs. 34.0%) [13]. In addition to impairing mobility and increasing dependence, sarcopenia exacerbates overall functional decline in terminally ill patients with cancer. A prospective study involving 1,660 patients with incurable cancer at the Palliative Care Unit of the Brazilian National Cancer Institute demonstrated that lower muscle mass and strength, assessed using MUAMA and handgrip strength (HGS), were strongly associated with worse performance status. Patients in the sarcopenia group had a median Karnofsky Performance Status of 40% compared with 60% in those with better muscle mass phenotypes. Notably, Patient-Generated Subjective Global Assessment Short Form (PG-SGA SF) scores, which reflect nutritional deterioration and symptom burden, also differed significantly according to sarcopenia severity and sex. Among men, those in the sarcopenia group had a median PG-SGA SF score of 15, compared with a score of 10 in those with better muscle mass phenotypes. Among women, the difference was even more pronounced, with PG-SGA SF scores of 21 in the sarcopenia group compared with a score of 13 in those with better muscle mass phenotypes [15].

Sarcopenia not only affects physical function but also significantly worsens QoL and symptom burden in patients receiving palliative care. A cross-sectional study of 770 patients with incurable cancer demonstrated that a worsening muscle mass phenotype, as assessed using the MUAMA and HGS, was strongly associated with declines in multiple QoL domains. Compared with patients without sarcopenia, those classified as having sarcopenia had significantly worse scores in physical function (9.1 vs. 32.2), emotional well-being (57.4 vs. 61.1), fatigue (59.9 vs. 44.7), and global health (50.2 vs. 57.5). Additionally, patients with sarcopenia were more likely to experience insomnia (odds ratio [OR] 3.11), fatigue (OR 1.83), and loss of appetite (OR 1.40), further exacerbating symptom burden and reducing overall well-being [16].

Collectively, sarcopenia progression leads to greater physical disability, worsening fatigue, increased nutritional deficits, and a higher prevalence of distressing symptoms. The interplay among muscle loss, malnutrition, and symptom burden further accelerates the decline in functional independence and overall well-being.

CHALLENGES IN DIAGNOSING SARCOPENIA IN TERMINALLY ILL PATIENTS WITH CANCER

Sarcopenia is typically diagnosed using imaging-based methods, body composition analysis, and functional assessment in patients with cancer undergoing surgery or chemotherapy [17]. Computed tomography (CT)-based muscle mass assessment is often used, as routine CT for cancer staging and response evaluation provides an opportunity to assess skeletal muscle depletion, particularly at the L3 level, or in some cases, using the pectoralis muscle index [18-21]. Other methods, such as dual-energy X-ray absorptiometry and BIA, allow whole-body muscle mass estimation [22,23]. In contrast, functional tests, such as the HGS and 6-minute walk test, assess muscle strength and physical performance [24,25].

In terminally ill patients with cancer, sarcopenia assessment is often challenging owing to less frequent routine imaging and a shift in focus from tumor response evaluation to symptom management. Systemic inflammation, cancer-associated cachexia, and prolonged bedridden status further limit the feasibility of advanced imaging and functional testing. Instead, diagnosis in this population requires noninvasive, bedside-friendly approaches prioritizing functional and nutritional status over detailed body composition analysis. The primary goal is not early detection for intervention but symptom monitoring and maintaining QoL.

The SARC-F questionnaire is a widely used screening tool for sarcopenia that evaluates strength, walking ability, ability to rise from a chair, climbing stairs, and history of falls. It is quick, noninvasive, and requires no equipment, making it ideal for palliative care settings [26]. Anthropometric measurements provide simple and accessible estimates of muscle mass and function. HGS, assessed using a dynamometer, serves as a surrogate marker for muscle strength and overall functional capacity and has been associated with frailty, functional decline, and decreased QoL in terminally ill patients with cancer [15,16]. MUAMA, calculated using the mid-upper arm circumference and triceps skinfold thickness, is an indicator of muscle wasting and nutritional status [12,15,16]. Similarly, the CC serves as an estimate of appendicular muscle mass and is correlated with frailty and physical function in older adults receiving palliative care [12,27]. BIA is a noninvasive and rapid method for evaluating skeletal muscle mass and hydration status [28]. Owing to its portability and ease of use, BIA can be performed at the bedside, making it a feasible alternative for sarcopenia assessment in patients who are bedridden or for whom imaging-based evaluations are inappropriate. However, the interpretation may be affected by fluid imbalances and systemic inflammation, which are common in this population [29]. Although each method has limitations, a combined approach may provide the most practical assessment in palliative care.

STRATEGIES FOR MANAGING SARCOPENIA IN PALLIATIVE CARE

In terminally ill patients, given their progressive catabolism and limited life expectancy, aggressive interventions provide little benefit and may increase discomfort [30]. Instead, care should prioritize nutritional support, maintenance of mobility, and symptom control tailored to the patient’s prognosis and goals. It is also important to consider that most available evidence does not clearly distinguish among anorexia, cachexia, and sarcopenia, making it essential to interpret management strategies with caution. Although these conditions share overlapping features, their underlying mechanisms and clinical implications differ, necessitating individualized approaches to palliative care.

1. Nutritional support

According to the American Society of Clinical Oncology guidelines, nutritional interventions in palliative care should prioritize maintaining energy intake, minimizing symptom burden, and preserving QoL [31]. According to the European Society for Clinical Nutrition and Metabolism clinical guidelines, indications for nutritional support in patients with advanced cancer should be guided by their estimated life expectancy and clinical situation. For patients with an expected survival time of several months or more, oral nutritional support and parenteral nutrition (PN) may be considered in select cases. For patients with a life expectancy of several weeks, nutritional interventions should be noninvasive, focusing primarily on symptom relief and psychosocial support. For patients with an expected survival of <7 days, total PN is generally avoided because it does not improve outcomes and may cause metabolic distress, potentially increasing discomfort. Instead, care should focus on oral comfort measures, hydration as needed, and minimizing interventions that may compromise QoL [32]. Some nutritional supplements, such as a combination therapy of β-hydroxy-β-methylbutyrate, arginine, and glutamine, have shown potential benefits in increasing lean body mass in patients with cancer [33]. However, most studies have focused on patients receiving disease-directed treatment with limited evidence supporting its use in terminally ill patients [34].

2. Exercise and physical activity

Exercise interventions in palliative care settings have been explored as potential strategies to improve physical function, fatigue, and QoL in patients with advanced cancer. A systematic review and meta-analysis of 14 studies, including 1,034 adults receiving palliative care, found that structured exercise interventions significantly improved exercise capacity, reduced pain and fatigue, and enhanced overall QoL [35]. Similarly, a study assessing the feasibility of a structured two-week exercise program in a palliative care unit found that, despite recruitment challenges, exercise was well tolerated and led to short-term improvements in physical function and QoL [36]. However, various clinical factors, including disease progression, physical function, and patient preferences, may pose hurdles to exercise in palliative care. A systematic review that assessed different exercise interventions, including aerobic, resistance, and combined training reported mixed results in patients with advanced cancer receiving palliative care. While resistance-based exercises improved at least one functional outcome in most studies, overall adherence was highly variable [37]. Despite these limitations, exercise remains a promising intervention that aligns with palliative care goals by enhancing mobility and reducing symptom burden. Feasibility depends on individualized adaptation, patient preferences, and practical considerations such as home-based or remotely supervised regimens. Given the heterogeneity of available studies, further research is needed to establish evidence-based exercise recommendations tailored for terminally ill patients receiving palliative care.

3. Pharmacological interventions

While nutritional support and physical activity remain central to sarcopenia management, pharmacological interventions have been explored to stimulate appetite, reduce inflammation, and mitigate muscle loss in patients with cancer. However, strong evidence supporting their routine use in terminally ill patients or palliative care settings remains scarce. Most studies on appetite stimulants and anti-cachexia therapies have been conducted on patients receiving disease-directed treatment, with limited data on their efficacy and safety in palliative care. Megestrol acetate, a progestin-derived appetite stimulant, has been shown to increase appetite and body weight in patients with cancer; however, its impact on muscle preservation and physical function is unclear [38]. A recent study reported that megestrol acetate showed a potential survival benefit in patients with metastatic gastric cancer who did not receive palliative chemotherapy. However, in multivariate analysis, the impact on survival was not statistically significant in this population. Moreover, the risks of thromboembolism, adrenal insufficiency, and fluid retention limit its use in palliative care, where symptom control and patient comfort are paramount [39]. Some studies have suggested that corticosteroids can temporarily improve appetite and energy levels, offering short-term relief from severe anorexia and fatigue [40]. However, prolonged use accelerates muscle wasting and exacerbates sarcopenia rather than mitigating it [41,42]. Adverse effects, including hyperglycemia, immunosuppression, and osteoporosis, often outweigh the potential benefits [43]. Given these risks, corticosteroids should be cautiously prescribed, prioritizing short-term symptom relief over long-term sarcopenia management.

Inflammation plays a central role in cachexia and sarcopenia, with elevated levels of proinflammatory cytokines contributing to muscle catabolism [44]. Several anti-inflammatory agents have been investigated in patients with cancer, primarily in the context of cancer cachexia rather than in sarcopenia. A systematic review assessed the efficacy and safety of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) in patients with cancer cachexia. The review included five randomized controlled trials focusing on indomethacin, ibuprofen, and celecoxib. The findings indicated that there is inadequate evidence to recommend any NSAID for cancer cachexia, whereas celecoxib is generally safe for use [45]. Beyond NSAIDs, other anti-inflammatory agents have been assessed. Thalidomide, an immunomodulatory drug with anti-tumor necrosis factor-α properties, has been investigated in cancer cachexia. A small randomized controlled trial found that improvements in symptoms and cytokine levels did not differ significantly between the thalidomide and placebo groups [46]. A systematic review further highlighted the lack of strong evidence supporting thalidomide for cancer cachexia, with high heterogeneity across studies and poor tolerance in some patients [47]. Similarly, investigational cytokine-targeting therapies have yielded inconsistent results. Although tocilizumab, an interleukin-6 receptor antagonist, has shown potential benefits in reducing inflammatory markers and improving body weight in patients with lung cancer, its effectiveness against cancer cachexia and sarcopenia remains inconclusive and requires further validation [48]. Other investigational anti-inflammatory agents such as infliximab and etanercept have failed to demonstrate efficacy in treating cancer, cachexia, and sarcopenia [49,50]. However, the roles of these anti-inflammatory agents in palliative care settings remain largely unknown. Considering the frailty of this population, the potential harm caused by anti-inflammatory therapies, including immunosuppression, thromboembolic risk, and other toxicities, should be carefully weighed against their uncertain benefits.

Recent advances in cancer cachexia and sarcopenia research have led to the development of novel pharmacological agents. Anamorelin, a selective ghrelin receptor agonist, has been shown in ROMANA 1 and 2 trials to increase appetite, body weight, and lean body mass in patients with advanced cancer [51]. A recent study suggested that ponsegromab, a humanized monoclonal antibody targeting Growth Differentiation Factor 15 (GDF15), was associated with improved weight, appetite, and physical activity in patients with cancer cachexia [52]. Despite promising early results, the role of ghrelin receptor agonist and anti-GDF15 therapy in palliative care has not yet been established. Further studies are required to determine whether these therapies can provide meaningful benefits beyond weight gain in patients receiving palliative care.

CONCLUSION

Sarcopenia is a prevalent but underexplored condition in terminally ill patients with cancer that contributes to functional decline and reduced QoL. Management should prioritize symptom relief, functional preservation, and patient comfort. Although nutritional and exercise interventions present challenges, they can improve mobility and well-being through individualized approaches. Current pharmacological options are limited; however, ongoing research on emerging therapies may provide future treatment options. A multidisciplinary approach integrating palliative care, nutritional support, and rehabilitation may optimize symptom control and enhance QoL in this vulnerable population. Future research should refine assessment methods and develop patient-centered, noninvasive strategies that align with palliative care principles.

SUPPLEMENTARY MATERIALS

Supplementary materials can be found via https://doi.org/10.14475/jhpc.2025.28.1.10.

CONFLICT OF INTEREST

No potential conflict of interest relevant to this article was reported.

AUTHOR’S CONTRIBUTIONS

Study conception and design: SG, HK. Data acquisition: SG, MHK, HK. Data analysis and interpretation: SG, MHK, HK. Manuscript drafting: SG, MHK, HK. Critical manuscript revision: SG, MHK, HK. Final manuscript approval: SG, MHK, HK.

There is no Figure.

There is no Table.

References

  1. Muscaritoli M, Anker SD, Argiles J, Aversa Z, Bauer JM, Biolo G, et al. Consensus definition of sarcopenia, cachexia and pre-cachexia: joint document elaborated by Special Interest Groups (SIG) "cachexia-anorexia in chronic wasting diseases" and "nutrition in geriatrics". Clin Nutr 2010;29:154-9.
    Pubmed CrossRef
  2. Oflazoglu U, Alacacioglu A, Varol U, Kucukzeybek Y, Salman T, Taskaynatan H, et al. Chemotherapy-induced sarcopenia in newly diagnosed cancer patients: Izmir Oncology Group (IZOG) study. Support Care Cancer 2020;28:2899-910.
    Pubmed CrossRef
  3. Albini A, La Vecchia C, Magnoni F, Garrone O, Morelli D, Janssens JP, et al. Physical activity and exercise health benefits: cancer prevention, interception, and survival. Eur J Cancer Prev 2025;34:24-39.
    Pubmed CrossRef
  4. Williams GR, Dunne RF, Giri S, Shachar SS, Caan BJ. Sarcopenia in the older adult with cancer. J Clin Oncol 2021;39:2068-78.
    Pubmed KoreaMed CrossRef
  5. Luo L, Shen X, Fang S, Wan T, Liu P, Li P, et al. Sarcopenia as a risk factor of progression-free survival in patients with metastases: a systematic review and meta-analysis. BMC Cancer 2023;23:127.
    Pubmed KoreaMed CrossRef
  6. Go SI, Choi BH, Park MJ, Park S, Kang MH, Kim HG, et al. Prognostic impact of pretreatment skeletal muscle index and CONUT score in diffuse large B-cell Lymphoma. BMC Cancer 2023;23:1071.
    Pubmed KoreaMed CrossRef
  7. Shachar SS, Williams GR, Muss HB, Nishijima TF. Prognostic value of sarcopenia in adults with solid tumours: A meta-analysis and systematic review. Eur J Cancer 2016;57:58-67.
    Pubmed CrossRef
  8. Xia Z, Cholewa J, Zhao Y, Shang HY, Yang YQ, Araujo Pessoa K, et al. Targeting inflammation and downstream protein metabolism in sarcopenia: A brief up-dated description of concurrent exercise and leucine-based multimodal intervention. Front Physiol 2017;8:434.
    Pubmed KoreaMed CrossRef
  9. Ho SY, Guo HR, Chen HH, Peng CJ. Nutritional predictors of survival in terminally ill cancer patients. J Formos Med Assoc 2003;102:544-50.
  10. Chiang JK, Cheng YH, Koo M, Kao YH, Chen CY. A computer-assisted model for predicting probability of dying within 7 days of hospice admission in patients with terminal cancer. Jpn J Clin Oncol 2010;40:449-55.
    Pubmed KoreaMed CrossRef
  11. Perez Camargo DA, Allende Perez SR, Verastegui Aviles E, Rivera Franco MM, Meneses Garcia A, Herrera Gomez A, et al. Assessment and impact of phase angle and sarcopenia in palliative cancer patients. Nutr Cancer 2017;69:1227-33.
    Pubmed CrossRef
  12. da Silva JR, Wiegert EVM, Oliveira L, Calixto-Lima L; Jr. Different methods for diagnosis of sarcopenia and its association with nutritional status and survival in patients with advanced cancer in palliative care. Nutrition 2019;60:48-52.
    Pubmed CrossRef
  13. Mori N, Maeda K, Fukami Y, Matsuyama R, Nonogaki T, Kato R, et al. High SARC-F score predicts poor survival of patients with cancer receiving palliative care. Support Care Cancer 2022;30:4065-72.
    Pubmed CrossRef
  14. Mercadante S, Bellavia GM, Fusco F, Adamoli L, Scibilia C, Lo Cascio A, et al. Malnutrition is associated with fatigue and anxiety in advanced cancer patients admitted to home palliative care. Am J Hosp Palliat Care :10499091241278924. Epup 2024 Aug 27.
    Pubmed CrossRef
  15. Calixto-Lima L, Souza-Silva R, Oliveira LC, Chaves GV, Wiegert EVM. Development and validation of a grading system for assessing muscle mass phenotype using mid-upper arm muscle area and handgrip strength in patients with incurable cancer. Nutr Clin Pract 2022;37:1385-99.
    Pubmed CrossRef
  16. Wiegert EVM, Oliveira LC, Rosa KSDC, Calixto-Lima L. Association of the muscle mass phenotype as assessed by a grading system with the quality of life of patients with incurable cancer in palliative care. Clin Nutr ESPEN 2024;62:216-23.
    Pubmed CrossRef
  17. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 2011;12:489-95.
    Pubmed CrossRef
  18. Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, et al. Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol 2008;9:629-35.
    Pubmed CrossRef
  19. Go SI, Kim HG, Kang MH, Park S, Lee GW. Prognostic model based on the geriatric nutritional risk index and sarcopenia in patients with diffuse large B-cell lymphoma. BMC Cancer 2020;20:439.
    Pubmed KoreaMed CrossRef
  20. Park SE, Choi JH, Park JY, Kim BJ, Kim JG, Kim JW, et al. Loss of skeletal muscle mass during palliative chemotherapy is a poor prognostic factor in patients with advanced gastric cancer. Sci Rep 2020;10:17683.
    Pubmed KoreaMed CrossRef
  21. Kim YS, Kim EY, Kang SM, Ahn HK, Kim HS. Single cross-sectional area of pectoralis muscle by computed tomography - correlation with bioelectrical impedance based skeletal muscle mass in healthy subjects. Clin Physiol Funct Imaging 2017;37:507-11.
    Pubmed CrossRef
  22. Kim EY, Kim K, Kim YS, Ahn HK, Jeong YM, Kim JH, et al. Prevalence of and factors associated with sarcopenia in Korean cancer survivors: based on data obtained by the Korea National Health and Nutrition Examination Survey (KNHANES) 2008-2011. Nutr Cancer 2017;69:394-401.
    Pubmed CrossRef
  23. Amano K, Bruera E, Hui D. Diagnostic and prognostic utility of phase angle in patients with cancer. Rev Endocr Metab Disord 2023;24:479-89.
    Pubmed CrossRef
  24. Sakaguchi T, Maeda K, Takeuchi T, Ishida Y, Kato R, Ueshima J, et al. Low handgrip strength as a marker of severity in the diagnostic criteria for cancer cachexia. Clin Nutr ESPEN 2024;64:435-40.
    Pubmed CrossRef
  25. Katakami N, Uchino J, Yokoyama T, Naito T, Kondo M, Yamada K, et al. Anamorelin (ONO-7643) for the treatment of patients with non-small cell lung cancer and cachexia: results from a randomized, double-blind, placebo-controlled, multicenter study of Japanese patients (ONO-7643-04). Cancer 2018;124:606-16.
    Pubmed KoreaMed CrossRef
  26. Malmstrom TK, Morley JE. SARC-F: a simple questionnaire to rapidly diagnose sarcopenia. J Am Med Dir Assoc 2013;14:531-2.
    Pubmed CrossRef
  27. Sousa IM, Bielemann RM, Gonzalez MC, da Rocha IMG, Barbalho ER, de Carvalho ALM, et al. Low calf circumference is an independent predictor of mortality in cancer patients: a prospective cohort study. Nutrition 2020;79-80:110816. Epub 2020 Mar 19.
    Pubmed CrossRef
  28. Branco MG, Mateus C, Capelas ML, Pimenta N, Santos T, Makitie A, et al. Bioelectrical impedance analysis (BIA) for the assessment of body composition in oncology: a scoping review. Nutrients 2023;15:4792.
    Pubmed KoreaMed CrossRef
  29. Lin SQ, Xie HL, Ge YZ, Ruan GT, Zhang Q, Song MM, et al. Association between systemic inflammation and water composition and survival in colorectal cancer. Front Oncol 2022;12:896160. eCollection 2022.
    Pubmed KoreaMed CrossRef
  30. Koroukian SM, Douglas SL, Vu L, Fein HL, Gairola R, Warner DF, et al. Incidence of aggressive end-of-life care among older adults with metastatic cancer living in nursing homes and community settings. JAMA Netw Open 2023;6:e230394.
    Pubmed KoreaMed CrossRef
  31. Roeland EJ, Bohlke K, Baracos VE, Bruera E, Del Fabbro E, Dixon S, et al. Management of cancer cachexia: ASCO guideline. J Clin Oncol 2020;38:2438-53.
    Pubmed CrossRef
  32. Muscaritoli M, Arends J, Bachmann P, Baracos V, Barthelemy N, Bertz H, et al. ESPEN practical guideline: Clinical nutrition in cancer. Clin Nutr 2021;40:2898-913.
    Pubmed CrossRef
  33. Berk L, James J, Schwartz A, Hug E, Mahadevan A, Samuels M, et al. A randomized, double-blind, placebo-controlled trial of a beta-hydroxyl beta-methyl butyrate, glutamine, and arginine mixture for the treatment of cancer cachexia (RTOG 0122). Support Care Cancer 2008;16:1179-88.
    Pubmed CrossRef
  34. Cuhls H, Marinova M, Kaasa S, Stieber C, Conrad R, et al; Mochamat. A systematic review on the role of vitamins, minerals, proteins, and other supplements for the treatment of cachexia in cancer: a European Palliative Care Research Centre cachexia project. J Cachexia Sarcopenia Muscle 2017;8:25-39.
    Pubmed KoreaMed CrossRef
  35. Tanriverdi A, Ozcan Kahraman B, Ergin G, Karadibak D, Savci S. Effect of exercise interventions in adults with cancer receiving palliative care: a systematic review and meta-analysis. Support Care Cancer 2023;31:205.
    Pubmed CrossRef
  36. Schwonke I, Freitag N, Aschendorf P, Wucharz K, Thieme J, Appelmann I, et al. Feasibility of a physical exercise intervention for patients on a palliative care unit: a critical analysis. BMC Palliat Care 2024;23:58.
    Pubmed KoreaMed CrossRef
  37. Rogers-Shepp I, Bhattacharya S, Mennillo HA, Kumar R, Hsieh B, Anandarajah G. Exercise interventions for advanced cancer palliative care patients: A systematic literature review and descriptive evidence synthesis of randomized controlled trials. Palliat Med 2023;37:677-91.
    Pubmed CrossRef
  38. Lim YL, Teoh SE, Yaow CYL, Lin DJ, Masuda Y, Han MX, et al. A systematic review and meta-analysis of the clinical use of megestrol acetate for cancer-related anorexia/cachexia. J Clin Med 2022;11:3756.
    Pubmed KoreaMed CrossRef
  39. Lee CM, Kang JH, Go SI. Clinical implication of megestrol acetate in metastatic gastric cancer: a big data analysis from Health Insurance Review and Assessment (HIRA) database. Support Care Cancer 2024;32:249.
    Pubmed CrossRef
  40. Miller S, McNutt L, McCann MA, McCorry N. Use of corticosteroids for anorexia in palliative medicine: a systematic review. J Palliat Med 2014;17:482-5.
    Pubmed CrossRef
  41. Salehian B, Kejriwal K. Glucocorticoid-induced muscle atrophy: mechanisms and therapeutic strategies. Endocr Pract 1999;5:277-81.
    Pubmed CrossRef
  42. Kuzuya M. Drug-related sarcopenia as a secondary sarcopenia. Geriatr Gerontol Int 2024;24:195-203.
    Pubmed KoreaMed CrossRef
  43. Liu D, Ahmet A, Ward L, Krishnamoorthy P, Mandelcorn ED, Leigh R, et al. A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol 2013;9:30.
    Pubmed KoreaMed CrossRef
  44. Setiawan T, Sari IN, Wijaya YT, Julianto NM, Muhammad JA, Lee H, et al. Cancer cachexia: molecular mechanisms and treatment strategies. J Hematol Oncol 2023;16:54.
    Pubmed KoreaMed CrossRef
  45. Bowers M, Cucchiaro B, Reid J, Slee A. Non-steroidal anti-inflammatory drugs for treatment of cancer cachexia: a systematic review. J Cachexia Sarcopenia Muscle 2023;14:2473-97.
    Pubmed KoreaMed CrossRef
  46. Yennurajalingam S, Willey JS, Palmer JL, Allo J, Del Fabbro E, Cohen EN, et al. The role of thalidomide and placebo for the treatment of cancer-related anorexia-cachexia symptoms: results of a double-blind placebo-controlled randomized study. J Palliat Med 2012;15:1059-64.
    Pubmed KoreaMed CrossRef
  47. Reid J, Mills M, Cantwell M, Cardwell CR, Murray LJ, Donnelly M. Thalidomide for managing cancer cachexia. Cochrane Database Syst Rev 2012;2012:CD008664.
    Pubmed KoreaMed CrossRef
  48. Du Y, Liu XY, Pan RL, Zhang XT, Si XY, Chen MJ, et al. Tocilizumab for advanced non-small-cell lung cancer with concomitant cachexia: an observational study. J Cachexia Sarcopenia Muscle 2024;15:2815-25.
    Pubmed KoreaMed CrossRef
  49. Jatoi A, Ritter HL, Dueck A, Nguyen PL, Nikcevich DA, Luyun RF, et al. A placebo-controlled, double-blind trial of infliximab for cancer-associated weight loss in elderly and/or poor performance non-small cell lung cancer patients (N01C9). Lung Cancer 2010;68:234-9.
    Pubmed KoreaMed CrossRef
  50. Jatoi A, Dakhil SR, Nguyen PL, Sloan JA, Kugler JW, Rowland KM Jr, et al. A placebo-controlled double blind trial of etanercept for the cancer anorexia/weight loss syndrome: results from N00C1 from the North Central Cancer Treatment Group. Cancer 2007;110:1396-403.
    Pubmed CrossRef
  51. Temel JS, Abernethy AP, Currow DC, Friend J, Duus EM, Yan Y, et al. Anamorelin in patients with non-small-cell lung cancer and cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double-blind, phase 3 trials. Lancet Oncol 2016;17:519-31.
    Pubmed CrossRef
  52. Groarke JD, Crawford J, Collins SM, Lubaczewski S, Roeland EJ, Naito T, et al. Ponsegromab for the treatment of cancer cachexia. N Engl J Med 2024;391:2291-303.
    Pubmed CrossRef

Supplementary

Stats or Metrics

Share this article on :

  • line