Back to Journals » Cancer Management and Research » Volume 12

Cancer Cachexia: Definition, Staging, and Emerging Treatments

Authors Ni J, Zhang L

Received 11 May 2020

Accepted for publication 26 June 2020

Published 9 July 2020 Volume 2020:12 Pages 5597—5605

DOI https://doi.org/10.2147/CMAR.S261585

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 4

Editor who approved publication: Dr Sanjeev K. Srivastava



Jun Ni, Li Zhang

Department of Pulmonary and Critical Care Medicine, Peking Union Medical Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, People’s Republic of China

Correspondence: Li Zhang
Department of Pulmonary and Critical Care Medicine, Peking Union Medical Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, People’s Republic of China
Tel +86 13911339836
Fax +86 10-69158760
Email [email protected]

Abstract: Cachexia is a multifactorial disease characterized by weight loss via skeletal muscle and adipose tissue loss, an imbalance in metabolic regulation, and reduced food intake. It is caused by factors of catabolism produced by tumors in the systemic circulation as well as physiological factors such as the imbalanced inflammatory activation, proteolysis, autophagy, and lipolysis that may occur with gastric, pancreatic, esophageal, lung cancer, liver, and bowel cancer. Cancer cachexia not only negatively affects the quality of life of patients with cancer but also reduces the effectiveness of anti-cancer chemotherapy and increases its toxicity, leading to increased cancer-related mortality and expenditure of medical resources. Currently, there are no effective medical interventions to completely reverse cachexia and no approved drugs. Adequate nutritional support is the main method of cachexia treatment, while drugs that target the inhibition of catabolism, cell damage, and excessive activation of inflammation are under study. This article reviews recent advances in the diagnosis, staging, and evaluation of cancer cachexia.

Keywords: cancer cachexia, disease staging, metabolic dysfunction

Introduction

The prevalence of cachexia is as high as 87% in patients with pancreatic and gastric cancer, 61% in patients with colon, lung, and prostate cancer and non-Hodgkin lymphoma, and 40% in breast cancer, sarcoma, leukemia, and Hodgkin lymphoma.1 Overall, cachexia accounts for 20% of all cancer-related deaths and is a sign of poor prognosis. Cancer cachexia develops as a secondary disease in patients with cancer and causes progressive dysfunction, characterized by a systemic inflammatory response, negative protein-energy balance, and involuntary loss of lean body mass, with or without a decline in adipose tissue.2 Clinically, cachexia manifests as a significant reduction in adult body weight or inhibited growth in children, accompanied by changes in body composition and disturbances in the balance of the biological system. Decreased skeletal muscle mass is the most obvious symptom of cancer cachexia and is accompanied by the depletion of fat and heart muscle. This review presents the definition and staging of cancer cachexia, before presenting the latest research in treatment and understanding this syndrome further.

Definition

The term “cachexia” originates from the Greek terms “kakos” and “hexis”, meaning “poor physical state”. Cancer cachexia is a multifactorial host-phagocytic syndrome characterized by a continuous decline in skeletal muscle mass, with or without fat loss. It leads to progressive functional impairment, weakens the effects of chemotherapy, and increases mortality in patients with cancer.37 Another significant feature of this paraneoplastic syndrome is that conventional nutritional support cannot be completely reversed.8 There is great heterogeneity in its presentation and severity, so its definition and diagnostic criteria are somewhat controversial.

In 2008, Evans9 defined cachexia as a complex metabolic syndrome associated with underlying illness, characterized by loss of muscle with or without loss of fat mass (Table 1). The prominent clinical feature of cachexia is weight loss in adults (fluid retention correction) or growth failure in children (excluding endocrine disorders).9 Because cancer cachexia cannot be easily distinguished from anorexia and other causes of weight/muscle mass loss, Fearon (2012)10 proposed that cancer cachexia is a multifactorial syndrome defined by an ongoing loss of skeletal muscle mass (with or without loss of fat mass) that can be partially but not entirely reversed by conventional nutritional support (Table 1). Depletion of skeletal muscle is a key feature of cancer-associated cachexia, and its consequences include reduced antitumor efficacy,11 increased chemotherapy toxicity,1214 complications from cancer surgery,15 and mortality.12,13,15-18 The physiological characteristics of the disease are negative nitrogen balance and negative energy balance due to reduced food intake and abnormally high metabolism. The diagnostic criteria defined by Evans are applicable to all types of chronic disease-related cachexia, when taking metabolism and nutrition into account. The diagnostic criteria presented by Fearon specifically target cancer-related cachexia, emphasizing weight loss factors and reduced muscle loss. Overall, weight loss, loss of appetite, growth disorders, and decreased muscle mass are the main symptoms of cachexia.

Table 1 Definitions of Cancer Cachexia

With the trend of patients with cancer becoming obese and overweight in recent years, the European Society of Clinical Nutrition (ESPEN)19 has recommended the higher cut-off value of 22 kg/m2 for BMI in the elderly. A study conducted by Martin20 assessing cancer-related weight loss criteria also proved that the value of weight loss percentage independent of BMI is limited and proposed a risk assessment model based on BMI corrected for weight loss. To facilitate clinical practice, the SCRINIO working group21 proposed a third definition in which patients with cachexia are classified based on weight loss (<10%, pre-cachexia; ≥10%, cachexia) and whether there is at least one symptom of anorexia, fatigue, or early satiety (Table 1). Because systemic inflammation and decreased muscle content are not classified as important diagnostic factors, these definitions have stronger clinical practicality and expand the diagnosis of cancer cachexia by stressing its universality.

Staging

Cancer cachexia is divided into three consecutive clinical stages:10 pre-cachexia, cachexia, and refractory cachexia, though patients may not experience all three stages. The incidence and severity of cachexia are highly heterogeneous and depend on the type, location, and stage of the tumor. At present, there are no specific biomarkers for early stage cachexia identification. Staging is determined according to the clinical manifestations and characteristics of the patient. The refractory cachexia phase is determined by the patient’s underlying disease and overall condition; diagnosis of this stage requires a low WHO performance status score and a survival period of less than 3 months.10 The focus of treatment for refractory cachexia moves from aiming to cure and control to maintaining the patient’s quality of life. This type of grading system can provide patients with more suitable treatment options at all stages of disease development, and allows for targeted research and treatment for each stage.

Evaluation

Cancer cachexia is a multi-dimensional disease, and the condition of each patient must be fully evaluated to determine their overall status to optimize the treatment decision. Cancer cachexia assessment mainly includes nutritional status, weight/content of body weight, quality of life, and related biomarkers. Insufficient intake is a common phenomenon, screening for malnutrition in patients with advanced cancer is critical. The Patient-Generated Subjective Global Assessment (PG-SGA),22 a modified for patients with cancer, is a validated screening tool for malnutrition. The PG-SGA questionnaire has a comprehensive design, that assesses factors such as patient weight, calorie intake, functional status; and temperature, muscle status, body fat status and whether edema is present or absent. The Mini Nutritional Assessment (MNA)23 is a validated, rapid screening tool of nutritional status, that evaluates diet history, weight, mid-arm circumference and nutritional risk factors. In clinical practice, the MNA that assesses elderly malnutrition is more practical as it can be completed within 10 minutes. Other screening tools include the Malnutrition Screening Tool24 and Nutritional Risk Screening 2002,25 but only the PG-SGA is adapted to patients with cancer.26

However, nutrition screening tools cannot assess muscle mass27 or body composition. The most commonly used parameters to assess body composition in patients with cancer include anthropometric methods, bioelectrical impedance analysis (BIA), computed tomography (CT) and dual-energy X-ray absorptiometry (DXA). The anthropometric method28 is a method for evaluating the composition of the human body by measuring skinfolds, weight, height, and body area. Although this is a simple method, it has poor accuracy and cannot distinguish between lean body mass and fat tissue. BIA29 is used to estimate the percentage of body fat, fat mass, fat-free mass and total fat content based on electrical properties, and uses an equation to calculate body fluid; however, BIA is not as accurate as DXA in assessing the body composition of patients with cancer. DXA30 evaluates human body composition by predominantly scanning appendicular muscles. The radiation dose and cost are low, however, this approach does not differentiate subsets of adipose tissue into intramuscular, visceral, and subcutaneous or lean body mass into muscle, organ tissue, and tumor tissue. Therefore, lean body mass is often overestimated with this approach. In general, CT31 scans are used to assess axial skeletal muscle mass by identifying standard skeletal markers (usually the third lumbar vertebra) to assess body composition. This approach has high accuracy and specificity as the scanning can distinguish individual tissue composition; it is the gold standard for evaluating body composition. The imaging has high accuracy and specificity. Magnetic resonance imaging (MRI)32 is a high-precision method for measuring the composition of the human body. It is equivalent to CT imaging,33 and does not expose patients to ionizing radiation, but it is costly. Other methods for assessing the overall composition of the body include water density (underwater weighing) and air plethysmography (Bod Pod),34,35 but they cannot distinguish between local fat or muscle. Upper-arm grip is the preferred method of assessing muscle strength,10,36 which can indirectly reflect muscle mass and function. The quality of life is usually used to assess the psychosocial and functional characteristics of patients.37

Biomarkers of cancer cachexia are research hotspots. Lipid and protein mobilization factors produced by tumors help to activate the inflammatory cascade, which in turn stimulates the adrenal glands to release cortisol and catecholamines; this leads to changes in protein metabolism through direct cytokine activation breakdown of proteins, carbohydrates, and lipids. A variety of inflammatory markers and cytokines are potential biomarkers of cachexia, including hemoglobin, albumin, C-reactive protein (CRP), ghrelin, adiponectin, leptin, insulin-like growth factor 1 (IGF-1), interleukin 1 (IL-1), interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α). Albumin and CRP levels are currently considered to be the best indicators of cancer cachexia.38 In order to quantify cancer cachexia systematically, scoring systems have been developed, including the modified Glasgow Prognostic Score (mGPS),39,40 cancer cachexia scoring system (CASCO),41,42 and cachexia staging score (CSS).43 However, biomarkers are affected by a variety of factors such as sex, age, and underlying diseases.44 The score corresponds with either non-malignant, pre-cachexia, cachexia, or refractory cachexia to effectively predict the survival of patients with the syndrome.

Treatment

It has been proven that cancer cachexia can be separated from underlying diseases by mechanical means, as the targeted blockade of cachexia signals can prolong survival while tumors continue to grow.45,46 In order to achieve the overall goals of improving muscle mass, improving the state of the body, and increasing the tolerance of anti-tumor therapies, the treatment of cancer cachexia must follow a comprehensive, individualized, structured, and continuous treatment model. For end-stage cachexia, palliative treatment strategies are better than other therapy options.47 For certain catabolically active underlying diseases (advanced lung cancer,48 bile duct cancer,2 etc.), the first choice of treatment is to inhibit catabolic drugs. To maximize the quality of life for patients, it is important to establish a continuous treatment system based on drug therapy and supplemented with nutrition, exercise, and psychological counseling.

As the basis for the treatment of cancer cachexia, drug therapy reduces tumor-related inflammation, increases anabolic metabolism, reduces catabolism, stimulates appetite, achieves weight and muscle gain, improves physical fitness scores, and extends survival. Megestrol acetate (MA) is the only drug approved by the FDA for the treatment of AIDS-related cachexia. Its representative drug is medroxyprogesterone acetate (MPA), which increases the release of neuropeptide Y in the hypothalamus to reduce and inhibit pro-inflammatory cytokines and stimulate appetite; this leads to increased food intake, weight gain, and an improved quality of life.49,50 However, the application of MPA is limited due to its association with increased peripheral thromboembolic events, fluid retention, adrenal insufficiency, hypogonadism, hyperglycemia, and hypertension. In addition to MPA, many of the following compounds have been put into clinical and preclinical research:

  1. Cannabinoids interact with endorphin receptors, interfere with IL-1 synthesis, activate cannabinoid receptors in the leptin neural circuit, and inhibit prostaglandin synthesis. They can increase the intake of energy storage and improve the nitrogen balance in the body,51 but can also produce serious central nervous system adverse reactions such as hallucinations, dizziness, and psychosis.52
  2. Cyproheptadine, a 5-HT inhibitor, is a kind of tissue serotonin that produces mild appetite stimulation and a certain degree of sedation.53 Because the results of current clinical trials involving this drug are inconsistent, it should be used with caution in clinical applications.54
  3. Non-Steroid Anti-Inflammation Drugs (NSAIDs) include COX-2 inhibitors such as celecoxib, indomethacin, and ibuprofen. They can reduce tumor-related inflammation and TNF-α levels, increase lean weight and grip strength, and improve life treatment and mGPS score.55,56 To date, only preliminary clinical trial results have been positive,57 as these drugs are not widely used outside of clinical trials.
  4. Immunomodulatory preparations mainly include TNF-α inhibitors (infliximab), IL-6 antagonists (ALD518), IL-6R antagonists (tocilizumab), and TNF-α and IL-6 dual-target OHR/AVR118. Current clinical trials have confirmed that infliximab has no significant effect on improving body weight and physical fitness.58 Conversely, ALD518 has been shown to delay lean body mass loss in non-small cell lung patients with cancer in a Phase II clinical trial.59 Tocilizumab has been shown to significantly improve weight and nutritional status, and to reduce the inflammatory response. Many clinical trials and case reports6062 indicate that OHR/AVR118 can improve cachexia functional status, stabilize weight, and stimulate appetite.63
  5. Growth hormone releasing peptides and their receptor agonists include ghrelin, anamorelin, and macimorelin, which stimulate growth hormone secretion,64 inhibit pro-inflammatory cytokines, and inhibit NF-κB65,66 to improve weight and appetite and increase muscle mass quickly.67,68 Although its half-life is less than 30min and there is limited clinical utility,69 ghrelin may stimulate tumor growth;70 therefore further data is needed based on Phase 3 clinical trials, to understand the potential risks. Anamorelin, a novel orally active ghrelin receptor agonist, has been associated with positive results in patients with cachexia who have non-small cell lung cancer. The two double-blinded phase II trials (ROMANA 1 and ROMANA271) assessed the efficacy and safety of anamorelin (100mg/d for 12 weeks) in patients with cachexia who have advanced NSCLC. Anamorelin significantly increased total body weight. These results were obtained by assessing the least-squares mean change ± standard error in the anamorelin group vs placebo group - ROMANA 1: 2.20 ± 0.33kg vs 0.14± 0.36kg, respectively, p<0.0001; for ROMANA 2:0.95 ±0.39 kg vs −0.57 ±0.44 kg, respectively, p<0.0001. The increase in body weight was especially seen in those with a low-BMI (<20kg/m2) patients.72 The weight increase, however, failed to improve handgrip strength. ROMANA373 also confirmed that anamorelin is well tolerated and increased body weight over 24-week period in the anamorelin vs placebo group (least-squares mean change ± standard error: 3.1±0.6kg vs 0.9±0.7kg, respectively, p<0.0001). In a real world setting, anamorelin not only stimulates food intake, but it also reversed loss of muscle tissue and lean body weight.69
  6. Selective androgen receptor modulators, such as enobosarm, selectively act on the androgen receptors of skeletal muscle and bones, thereby minimizing irritation to other organs such as the prostate, skin, and liver. These modulators can increase lean body mass and improve activity tolerance.74,75 Related phase 3 clinical trials are underway (NCT 01355484, NCT 01355497).
  7. Anabolic catabolic transforming agents (ACTAs), such as spindolol, are a class of non-specific β-1 and β-2 adrenergic blockers with β2 adrenergic receptor intrinsic sympathomimetic activity. As a strong HT-1A receptor agonist, spindolol can bind to HT-1A receptors in the brain. It can increase muscle content, weight, and improve grip.76 Initial preliminary data is promising, but more research is required to indicate the efficacy and to provide a safety data disclosure.
  8. As a methylxanthine derivative, the drug pentoxifylline inhibits the characteristics of systemic inflammation and TNF-α by inhibiting phosphodiesterase; its efficacy has not been proven in cancer cachexia.77
  9. Glucocorticoids, such as the drugs prednisone acetate and dexamethasone, inhibit pro-inflammatory cytokines like TNF-α and IL-1 to increase appetite and improve nausea, but cannot eliminate muscle loss.78 Those increase in body weight was mainly due to an increase in fat mass and water retention, and was not associated with an increase in lean body mass nor skeletal muscle. Adverse reactions such as insulin resistance, adrenal insufficiency, and sleep disorders have been observed, and there is no evidence of clinical application.
  10. Melanocortin-4 receptor (MC-4R) is a member of the melanocortin receptor family, a group of G-protein-coupled receptors that secrete a peptide in the ventromedial nucleus of the hypothalamus. These substances play an important role in regulating food intake. In a mouse model, MC-4R activation has been shown to reduce foraging behavior, increase basal metabolic rate, and reduce lean body mass.79 The effectiveness of MC-4R antagonists has also been demonstrated in murine models.80 However, clinical trials have not yet begun.
  11. Other compounds: growth hormone,81 olanzapine,82 bortezomib,83 the JAK/STAT3 inhibitor ruxolitinib, and the myostatin/activin inhibitor BYM338 are all potential drugs that have not been validated in clinical trials.

These drugs have the potential to treat cancer cachexia preferentially. However, there are numerous interactions between different mediators when complex inflammatory cascades are activated, suggesting that a single target drug cannot be used as a cure. Most patients with cancer-related cachexia must choose a targeted intervention point to maximize the effect on the syndrome.

According to the ASCO Guideline,16 dietary assessment and counseling for patients with advanced cancer who suffer from insufficient intake due to various factors is the first recommendation based on robust evidence,8486 with goals of dietary structure adjustment (high energy and high protein diet), increased meal frequency, and oral nutritional supplements (β-hydroxy-β-methylbutyrate,87 eicosapentaenoic acid,88 L-carnitine,89 omega-3 fatty acids90). If the terminal patient with cancer cannot eat for a long time, artificial feeding must be carried out. The enteral route should be given priority, but parenteral nutrition can be applied when the enteral route is insufficient or not feasible.91 However, parenteral nutrition cannot improve the overall survival of patients with cancer cachexia,92,93 and as a high-calorie high-protein and nutrient-dense feeding generally does not affect body composition in these patients.94 Moreover, if the patient has difficulty digesting and absorbing nutrients, the caregivers have a tendency to repeat feeding, which can induce the patient vomiting, and cause complications such as regurgitation, aspiration, pneumonia. Often, the patient ends up feeling frustrated and to blame. In addition, proper physical exercise can preserve muscle mass and function95 while reducing systemic inflammation9698 and decreasing systemic catabolism,97,99 all of which prevent the symptoms of cancer cachexia. It can also delay muscle breakdown and insulin resistance,100 as well as provide adequate psychological support and intervention to improve quality of life.101

Research is currently underway on the correlation between cachexia treatment and cancer treatment. For example, studies have shown that cachexia tumor cell proliferation and excessive muscle-protein catabolism share a common mechanism. Specifically, dual-specific mitogen-activated protein kinase kinase-1 (MAP2K1) and MAP2K2 function downstream of RAS GTPases and RAF proto-oncogene serine/threonine protein kinase (RAF) to induce phosphorylation of MAPK1 and MAPK3, thereby conveying input promotion from growth factors for tumor cell proliferation. This signaling pathway also appears to be involved in the activation of excess muscle protein catabolism when tumor growth is present.70,102 Therefore, MAP2K inhibitors may have both anti-cachexia and anti-tumor activity. These types of interactions should be explored through further research.

The diagnosis of cachexia in patients with cancer has made significant progress over time, evolving from the label “unintentional weight loss” to “cancer cachexia” in the clinical environment. With the continuous development of this field, genomics and metabolomics may be used to assist diagnosis. Classification and reclassification provide each patient with a more individualized and refined systemic treatment strategy. However, it must be acknowledged that the overall quality of clinical trials related to cancer cachexia is not high. In recent years, whether the trials have produced clinical benefits (improved grip strength, climbing stairs) is also controversial as the end point of the study.103 Clinical trials and nutrition interventions for related new drugs are continuously being carried out and promoted, and regular efforts are being made for better treatment. The multimodal cachexia intervention, consisting of drug, nutritional supplements and adequate exercise, is feasible and safe for patients with cancer cachexia.104

Ethics Statement

No institutional approval was required to publish the work details.

Disclosure

The authors declare that they have no conflicts of interest.

References

1. Dewys WD, Begg C, Lavin PT, et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. Am J Med. 1980;69(4):491–497. doi:10.1016/S0149-2918(05)80001-3

2. Fearon KC. Cancer cachexia: developing multimodal therapy for a multidimensional problem. Eur J Cancer. 2008;44(8):1124–1132. doi:10.1016/j.ejca.2008.02.033

3. Fearon KC, Voss AC, Hustead DS; Cancer Cachexia Study G. Definition of cancer cachexia: effect of weight loss, reduced food intake, and systemic inflammation on functional status and prognosis. Am J Clin Nutr. 2006;83(6):1345–1350. doi:10.1093/ajcn/83.6.1345

4. Vaughan VC, Martin P, Lewandowski PA. Cancer cachexia: impact, mechanisms and emerging treatments. J Cachexia Sarcopenia Muscle. 2013;4(2):95–109. doi:10.1007/s13539-012-0087-1

5. Aoyagi T, Terracina KP, Raza A, Matsubara H, Takabe K. Cancer cachexia, mechanism and treatment. World J Gastrointest Oncol. 2015;7(4):17–29. doi:10.4251/wjgo.v7.i4.17

6. LeBlanc TW, Nipp RD, Rushing CN, et al. Correlation between the international consensus definition of the Cancer Anorexia-Cachexia Syndrome (CACS) and patient-centered outcomes in advanced non-small cell lung cancer. J Pain Symptom Manage. 2015;49(4):680–689. doi:10.1016/j.jpainsymman.2014.09.008

7. Anker MS, Holcomb R, Muscaritoli M, et al. Orphan disease status of cancer cachexia in the USA and in the European Union: a systematic review. J Cachexia Sarcopenia Muscle. 2019;10(1):22–34. doi:10.1002/jcsm.12402

8. Suzuki H, Asakawa A, Amitani H, Nakamura N, Inui A. Cancer cachexia–pathophysiology and management. J Gastroenterol. 2013;48(5):574–594. doi:10.1007/s00535-013-0787-0

9. Evans WJ, Morley JE, Argiles J, et al. Cachexia: a new definition. Clin Nutr. 2008;27(6):793–799. doi:10.1016/j.clnu.2008.06.013

10. Fearon K, Strasser F, Anker SD, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489–495. doi:10.1016/S1470-2045(10)70218-7

11. Senesse P, Assenat E, Schneider S, et al. Nutritional support during oncologic treatment of patients with gastrointestinal cancer: who could benefit? Cancer Treat Rev. 2008;34(6):568–575. doi:10.1016/j.ctrv.2008.03.003

12. da Rocha IMG, Marcadenti A, de Medeiros GOC, et al. Is cachexia associated with chemotherapy toxicities in gastrointestinal cancer patients? A prospective study. J Cachexia Sarcopenia Muscle. 2019;10(2):445–454. doi:10.1002/jcsm.12391

13. Baracos VE, Mazurak VC, Bhullar AS. Cancer cachexia is defined by an ongoing loss of skeletal muscle mass. Ann Palliat Med. 2019;8(1):3–12. doi:10.21037/apm.2018.12.01

14. Nicolini A, Ferrari P, Masoni MC, et al. Malnutrition, anorexia and cachexia in cancer patients: A mini-review on pathogenesis and treatment. Biomed Pharmacother. 2013;67(8):807–817. doi:10.1016/j.biopha.2013.08.005

15. Pausch T, Hartwig W, Hinz U, et al. Cachexia but not obesity worsens the postoperative outcome after pancreatoduodenectomy in pancreatic cancer. Surgery. 2012;152(3 Suppl 1):S81–S88. doi:10.1016/j.surg.2012.05.028

16. Roeland EJ, Bohlke K, Baracos VE, et al. Management of cancer cachexia: ASCO guideline. J Clin Oncol. 2020: JCO2000611. doi:10.1200/JCO.20.00611

17. 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

18. Bo Y, Yao M, Zhang L, Bekalo W, Lu W, Lu Q. Preoperative nutritional risk index to predict postoperative survival time in primary liver cancer patients. Asia Pac J Clin Nutr. 2015;24(4):591–597. doi:10.6133/apjcn.2015.24.4.26

19. Cederholm T, Bosaeus I, Barazzoni R, et al. Diagnostic criteria for malnutrition - An ESPEN consensus statement. Clin Nutr. 2015;34(3):335–340. doi:10.1016/j.clnu.2015.03.001

20. Martin L, Senesse P, Gioulbasanis I, et al. Diagnostic criteria for the classification of cancer-associated weight loss. J Clin Oncol. 2015;33(1):90–99. doi:10.1200/JCO.2014.56.1894

21. Bozzetti F, Mariani L. Defining and classifying cancer cachexia: a proposal by the SCRINIO Working Group. JPEN J Parenter Enteral Nutr. 2009;33(4):361–367. doi:10.1177/0148607108325076

22. Ottery FD. Definition of standardized nutritional assessment and interventional pathways in oncology. Nutrition. 1996;12(1 Suppl):S15–S19. doi:10.1016/0899-9007(95)00067-4

23. Vellas B, Guigoz Y, Garry PJ, et al. The mini nutritional assessment (MNA) and its use in grading the nutritional state of elderly patients. Nutrition. 1999;15(2):116–122. doi:10.1016/S0899-9007(98)00171-3

24. Ferguson M, Capra S, Bauer J, Banks M. Development of a valid and reliable malnutrition screening tool for adult acute hospital patients. Nutrition. 1999;15(6):458–464. doi:10.1016/S0899-9007(99)00084-2

25. Kondrup J, Allison SP, Elia M, et al. ESPEN guidelines for nutrition screening 2002. Clin Nutr. 2003;22(4):415–421. doi:10.1016/S0261-5614(03)00098-0

26. Du H, Liu B, Xie Y, et al. Comparison of different methods for nutrition assessment in patients with tumors. Oncol Lett. 2017;14(1):165–170. doi:10.3892/ol.2017.6154

27. Thoresen L, Frykholm G, Lydersen S, et al. Nutritional status, cachexia and survival in patients with advanced colorectal carcinoma. Different assessment criteria for nutritional status provide unequal results. Clin Nutr. 2013;32(1):65–72. doi:10.1016/j.clnu.2012.05.009

28. Harvie MN, Campbell IT, Thatcher N, Baildam A. Changes in body composition in men and women with advanced nonsmall cell lung cancer (NSCLC) undergoing chemotherapy. J Hum Nutr Diet. 2003;16(5):323–326. doi:10.1046/j.1365-277X.2003.00459.x

29. Cardoso ICR, Aredes MA, Chaves GV. Applicability of the direct parameters of bioelectrical impedance in assessing nutritional status and surgical complications of women with gynecological cancer. Eur J Clin Nutr. 2017;71(11):1278–1284. doi:10.1038/ejcn.2017.115

30. Trutschnigg B, Kilgour RD, Reinglas J, et al. Precision and reliability of strength (Jamar vs. Biodex handgrip) and body composition (dual-energy X-ray absorptiometry vs. bioimpedance analysis) measurements in advanced cancer patients. Appl Physiol Nutr Metab. 2008;33(6):1232–1239. doi:10.1139/H08-122

31. Prado CM, Birdsell LA, Baracos VE. The emerging role of computerized tomography in assessing cancer cachexia. Curr Opin Support Palliat Care. 2009;3(4):269–275. doi:10.1097/SPC.0b013e328331124a

32. Lindenberg KS, Weydt P, Muller HP, et al. Two-point magnitude MRI for rapid mapping of brown adipose tissue and its application to the R6/2 mouse model of Huntington disease. PLoS One. 2014;9(8):e105556. doi:10.1371/journal.pone.0105556

33. Bieliuniene E, Brondum Frokjaer J, Pockevicius A, et al. CT- and MRI-based assessment of body composition and pancreatic fibrosis reveals high incidence of clinically significant metabolic changes that affect the quality of life and treatment outcomes of patients with chronic pancreatitis and pancreatic cancer. Medicina (Kaunas). 2019;55(10):649.

34. Gnaedinger RH, Reineke EP, Pearson AM, Vanhuss WD, Wessel JA, Montoye HJ. Determination of body density by air displacement, helium dilution, and underwater weighing. Ann N Y Acad Sci. 1963;110:96–108. doi:10.1111/j.1749-6632.1963.tb17077.x

35. Heymsfield SB, Wang ZM. Measurement of total-body fat by underwater weighing: new insights and uses for old method. Nutrition. 1993;9(5):472–473.

36. Prado CM, Baracos VE, McCargar LJ, 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(8):2920–2926. doi:10.1158/1078-0432.CCR-08-2242

37. Wheelwright S, Darlington AS, Hopkinson JB, Fitzsimmons D, White A, Johnson CD. A systematic review of health-related quality of life instruments in patients with cancer cachexia. Support Care Cancer. 2013;21(9):2625–2636. doi:10.1007/s00520-013-1881-9

38. Takayoshi K, Uchino K, Nakano M, Ikejiri K, Baba E. Weight loss during initial chemotherapy predicts survival in patients with advanced gastric cancer. Nutr Cancer. 2017;69(3):408–415. doi:10.1080/01635581.2017.1267774

39. Demirelli B, Babacan NA, Ercelep O, et al. Modified glasgow prognostic score, prognostic nutritional index and ECOG performance score predicts survival better than sarcopenia, cachexia and some inflammatory indices in metastatic gastric cancer. Nutr Cancer. 2020: 1–9. doi:10.1080/01635581.2020.1749290

40. Silva GAD, Wiegert EVM, Calixto-Lima L, Oliveira LC. Clinical utility of the modified Glasgow Prognostic Score to classify cachexia in patients with advanced cancer in palliative care. Clin Nutr. 2020;39(5):1587–1592. doi:10.1016/j.clnu.2019.07.002

41. Argiles JM, Lopez-Soriano FJ, Toledo M, Betancourt A, Serpe R, Busquets S. The cachexia score (CASCO): a new tool for staging cachectic cancer patients. J Cachexia Sarcopenia Muscle. 2011;2(2):87–93. doi:10.1007/s13539-011-0027-5

42. Argiles JM, Betancourt A, Guardia-Olmos J, et al. Validation of the CAchexia SCOre (CASCO). staging cancer patients: the use of miniCASCO as a simplified tool. Front Physiol. 2017;8:92. doi:10.3389/fphys.2017.00092

43. Zhou T, Wang B, Liu H, et al. Development and validation of a clinically applicable score to classify cachexia stages in advanced cancer patients. J Cachexia Sarcopenia Muscle. 2018;9(2):306–314. doi:10.1002/jcsm.12275

44. Bye A, Wesseltoft-Rao N, Iversen PO, et al. Alterations in inflammatory biomarkers and energy intake in cancer cachexia: a prospective study in patients with inoperable pancreatic cancer. Med Oncol. 2016;33(6):54. doi:10.1007/s12032-016-0768-2

45. Zhou X, Wang JL, Lu J, et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell. 2010;142(4):531–543. doi:10.1016/j.cell.2010.07.011

46. Tseng YC, Kulp SK, Lai IL, et al. Preclinical Investigation Of The Novel Histone Deacetylase Inhibitor AR-42 in the treatment of cancer-induced cachexia. J Natl Cancer Inst. 2015;107(12):djv274. doi:10.1093/jnci/djv274

47. Maltoni M, Caraceni A, Brunelli C, et al. Prognostic factors in advanced cancer patients: evidence-based clinical recommendations–a study by the Steering Committee of the European Association for Palliative Care. J Clin Oncol. 2005;23(25):6240–6248. doi:10.1200/JCO.2005.06.866

48. von Haehling S, Anker SD. Cachexia as a major underestimated and unmet medical need: facts and numbers. J Cachexia Sarcopenia Muscle. 2010;1(1):1–5. doi:10.1007/s13539-010-0002-6

49. Lesniak W, Bala M, Jaeschke R, Krzakowski M. Effects of megestrol acetate in patients with cancer anorexia-cachexia syndrome–a systematic review and meta-analysis. Pol Arch Med Wewn. 2008;118(11):636–644.

50. Ruiz Garcia V, Lopez-Briz E, Carbonell Sanchis R, Gonzalvez Perales JL, Bort-Marti S. Megestrol acetate for treatment of anorexia-cachexia syndrome. Cochrane Database Syst Rev. 2013;3:CD004310.

51. Gamage TF, Lichtman AH. The endocannabinoid system: role in energy regulation. Pediatr Blood Cancer. 2012;58(1):144–148. doi:10.1002/pbc.23367

52. Tafelski S, Hauser W, Schafer M. Efficacy, tolerability, and safety of cannabinoids for chemotherapy-induced nausea and vomiting–a systematic review of systematic reviews. Schmerz. 2016;30(1):14–24. doi:10.1007/s00482-015-0092-3

53. Mantovani G, Maccio A, Massa E, Madeddu C. Managing cancer-related anorexia/cachexia. Drugs. 2001;61(4):499–514. doi:10.2165/00003495-200161040-00004

54. Couluris M, Mayer JL, Freyer DR, Sandler E, Xu P, Krischer JP. The effect of cyproheptadine hydrochloride (periactin) and megestrol acetate (megace) on weight in children with cancer/treatment-related cachexia. J Pediatr Hematol Oncol. 2008;30(11):791–797. doi:10.1097/MPH.0b013e3181864a5e

55. Lai V, George J, Richey L, et al. Results of a pilot study of the effects of celecoxib on cancer cachexia in patients with cancer of the head, neck, and gastrointestinal tract. Head Neck. 2008;30(1):67–74. doi:10.1002/hed.20662

56. Mantovani G, Maccio A, Madeddu C, et al. Phase II nonrandomized study of the efficacy and safety of COX-2 inhibitor celecoxib on patients with cancer cachexia. J Mol Med (Berl). 2010;88(1):85–92. doi:10.1007/s00109-009-0547-z

57. Maccio A, Madeddu C, Gramignano G, et al. A randomized Phase III clinical trial of a combined treatment for cachexia in patients with gynecological cancers: evaluating the impact on metabolic and inflammatory profiles and quality of life. Gynecol Oncol. 2012;124(3):417–425. doi:10.1016/j.ygyno.2011.12.435

58. Jatoi A, Ritter HL, Dueck A, 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(2):234–239. doi:10.1016/j.lungcan.2009.06.020

59. Schuster M, Rigas JR, Orlov SV, et al. ALD518, a humanized anti-IL-6 antibody, treats anemia in patients with advanced non-small cell lung cancer (NSCLC): results of a Phase II, randomized, double-blind, placebo-controlled trial. J Clin Oncol. 2010;28:15. doi:10.1200/jco.2010.28.15_suppl.7631

60. Ando K, Takahashi F, Takahashi K. Reply to A. Berti et al. J Clin Oncol. 2013;31(23):2971. doi:10.1200/JCO.2013.50.4324

61. Berti A, Boccalatte F, Sabbadini MG, Dagna L. Assessment of tocilizumab in the treatment of cancer cachexia. J Clin Oncol. 2013;31(23):2970. doi:10.1200/JCO.2012.48.4147

62. Ando K, Takahashi F, Kato M, et al. Tocilizumab, a proposed therapy for the cachexia of Interleukin6-expressing lung cancer. PLoS One. 2014;9(7):e102436. doi:10.1371/journal.pone.0102436

63. Chasen M, Hirschman SZ, Bhargava R. Phase II study of the novel peptide-nucleic acid OHR118 in the management of cancer-related anorexia/cachexia. J Am Med Dir Assoc. 2011;12(1):62–67. doi:10.1016/j.jamda.2010.02.012

64. Takaya K, Ariyasu H, Kanamoto N, et al. Ghrelin strongly stimulates growth hormone release in humans. J Clin Endocrinol Metab. 2000;85(12):4908–4911. doi:10.1210/jcem.85.12.7167

65. Li WG, Gavrila D, Liu X, et al. Ghrelin inhibits proinflammatory responses and nuclear factor-kappaB activation in human endothelial cells. Circulation. 2004;109(18):2221–2226. doi:10.1161/01.CIR.0000127956.43874.F2

66. Waseem T, Duxbury M, Ito H, Ashley SW, Robinson MK. Exogenous ghrelin modulates release of pro-inflammatory and anti-inflammatory cytokines in LPS-stimulated macrophages through distinct signaling pathways. Surgery. 2008;143(3):334–342. doi:10.1016/j.surg.2007.09.039

67. Garcia J, Boccia RV, Graham C, Kumor K, Polvino W. A phase II randomized, placebo-controlled, double-blind study of the efficacy and safety of RC-1291 (RC) for the treatment of cancer cachexia. J Clin Oncol. 2007;25(18):9133. doi:10.1200/jco.2007.25.18_suppl.9133

68. Garcia JM, Boccia RV, Graham CD, et al. Anamorelin for patients with cancer cachexia: an integrated analysis of two Phase 2, randomised, placebo-controlled, double-blind trials. Lancet Oncol. 2015;16(1):108–116. doi:10.1016/S1470-2045(14)71154-4

69. Currow DC, Maddocks M, Cella D, Muscaritoli M. Efficacy of anamorelin, a novel non-peptide ghrelin analogue, in patients with advanced non-small cell lung cancer (NSCLC) and cachexia-review and expert opinion. Int J Mol Sci. 2018;19(11):3471. doi:10.3390/ijms19113471

70. Murphy RA, Mourtzakis M, Chu QS, Baracos VE, Reiman T, Mazurak VC. Supplementation with fish oil increases first-line chemotherapy efficacy in patients with advanced nonsmall cell lung cancer. Cancer. 2011;117(16):3774–3780. doi:10.1002/cncr.25933

71. Temel JS, Abernethy AP, Currow DC, 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(4):519–531. doi:10.1016/S1470-2045(15)00558-6

72. Currow DC, Temel JS, Abernethy AP, Friend J, Giorgino R. Body weight response with anamorelin in advanced non-small cell lung cancer (NSCLC) patients with anorexia/cachexia: pooled analysis of two phase III trials. J Clin Oncol. 2017;35(15_suppl):10097. doi:10.1200/JCO.2017.35.15_suppl.10097

73. Currow D, Temel JS, Abernethy A, Milanowski J, Friend J, Fearon KC. ROMANA 3: a phase 3 safety extension study of anamorelin in advanced non-small-cell lung cancer (NSCLC) patients with cachexia. Ann Oncol. 2017;28(8):1949–1956. doi:10.1093/annonc/mdx192

74. Dalton JT, Barnette KG, Bohl CE, et al. The selective androgen receptor modulator GTx-024 (enobosarm) improves lean body mass and physical function in healthy elderly men and postmenopausal women: results of a double-blind, placebo-controlled phase II trial. J Cachexia Sarcopenia Muscle. 2011;2(3):153–161. doi:10.1007/s13539-011-0034-6

75. Dobs AS, Boccia RV, Croot CC, et al. Effects of enobosarm on muscle wasting and physical function in patients with cancer: a double-blind, randomised controlled phase 2 trial. Lancet Oncol. 2013;14(4):335–345. doi:10.1016/S1470-2045(13)70055-X

76. Stewart Coats AJ, Ho GF, Prabhash K, et al. Espindolol for the treatment and prevention of cachexia in patients with stage III/IV non-small cell lung cancer or colorectal cancer: a randomized, double-blind, placebo-controlled, international multicentre phase II study (the ACT-ONE trial). J Cachexia Sarcopenia Muscle. 2016;7(3):355–365. doi:10.1002/jcsm.12126

77. Mehrzad V, Afshar R, Akbari M. Pentoxifylline treatment in patients with cancer cachexia: A double-blind, randomized, placebo-controlled clinical trial. Adv Biomed Res. 2016;5:60. doi:10.4103/2277-9175.179182

78. Chauhan A, Sequeria A, Manderson C, Maddocks M, Wasley D, Wilcock A. Exploring autonomic nervous system dysfunction in patients with cancer cachexia: a pilot study. Auton Neurosci. 2012;166(1–2):93–95. doi:10.1016/j.autneu.2011.09.006

79. Marks DL, Ling N, Cone RD. Role of the central melanocortin system in cachexia. Cancer Res. 2001;61(4):1432–1438.

80. Dallmann R, Weyermann P, Anklin C, et al. The orally active melanocortin-4 receptor antagonist BL-6020/979: a promising candidate for the treatment of cancer cachexia. J Cachexia Sarcopenia Muscle. 2011;2(3):163–174. doi:10.1007/s13539-011-0039-1

81. Tuca A, Jimenez-Fonseca P, Gascon P. Clinical evaluation and optimal management of cancer cachexia. Crit Rev Oncol Hematol. 2013;88(3):625–636. doi:10.1016/j.critrevonc.2013.07.015

82. Naing A, Dalal S, Abdelrahim M, et al. Olanzapine for cachexia in patients with advanced cancer: an exploratory study of effects on weight and metabolic cytokines. Support Care Cancer. 2015;23(9):2649–2654. doi:10.1007/s00520-015-2625-9

83. Jatoi A, Alberts SR, Foster N, et al. Is bortezomib, a proteasome inhibitor, effective in treating cancer-associated weight loss? Preliminary results from the North Central Cancer Treatment Group. Support Care Cancer. 2005;13(6):381–386. doi:10.1007/s00520-005-0787-6

84. Baldwin C, Spiro A, Ahern R, Emery PW. Oral nutritional interventions in malnourished patients with cancer: a systematic review and meta-analysis. J Natl Cancer Inst. 2012;104(5):371–385. doi:10.1093/jnci/djr556

85. de van der Schueren MAE, Laviano A, Blanchard H, Jourdan M, Arends J, Baracos VE. Systematic review and meta-analysis of the evidence for oral nutritional intervention on nutritional and clinical outcomes during chemo(radio)therapy: current evidence and guidance for design of future trials. Ann Oncol. 2018;29(5):1141–1153. doi:10.1093/annonc/mdy114

86. Balstad TR, Solheim TS, Strasser F, Kaasa S, Bye A. Dietary treatment of weight loss in patients with advanced cancer and cachexia: a systematic literature review. Crit Rev Oncol Hematol. 2014;91(2):210–221. doi:10.1016/j.critrevonc.2014.02.005

87. Fitschen PJ, Wilson GJ, Wilson JM, Wilund KR. Efficacy of beta-hydroxy-beta-methylbutyrate supplementation in elderly and clinical populations. Nutrition. 2013;29(1):29–36. doi:10.1016/j.nut.2012.05.005

88. Sanchez-Lara K, Turcott JG, Juarez-Hernandez E, et al. Effects of an oral nutritional supplement containing eicosapentaenoic acid on nutritional and clinical outcomes in patients with advanced non-small cell lung cancer: randomised trial. Clin Nutr. 2014;33(6):1017–1023. doi:10.1016/j.clnu.2014.03.006

89. Kraft M, Kraft K, Gartner S, et al. L-Carnitine-supplementation in advanced pancreatic cancer (CARPAN)–a randomized multicentre trial. Nutr J. 2012;11:52. doi:10.1186/1475-2891-11-52

90. Ries A, Trottenberg P, Elsner F, et al. A systematic review on the role of fish oil for the treatment of cachexia in advanced cancer: an EPCRC cachexia guidelines project. Palliat Med. 2012;26(4):294–304. doi:10.1177/0269216311418709

91. Arends J, Bachmann P, Baracos V, et al. ESPEN guidelines on nutrition in cancer patients. Clin Nutr. 2017;36(1):11–48. doi:10.1016/j.clnu.2016.07.015

92. Oh SY, Jun HJ, Park SJ, et al. A randomized phase II study to assess the effectiveness of fluid therapy or intensive nutritional support on survival in patients with advanced cancer who cannot be nourished via enteral route. J Palliat Med. 2014;17(11):1266–1270. doi:10.1089/jpm.2014.0082

93. Obling SR, Wilson BV, Pfeiffer P, Kjeldsen J. Home parenteral nutrition increases fat free mass in patients with incurable gastrointestinal cancer. Results of a randomized controlled trial. Clin Nutr. 2019;38(1):182–190. doi:10.1016/j.clnu.2017.12.011

94. Lundholm K, Daneryd P, Bosaeus I, Korner U, Lindholm E. Palliative nutritional intervention in addition to cyclooxygenase and erythropoietin treatment for patients with malignant disease: effects on survival, metabolism, and function. Cancer. 2004;100(9):1967–1977. doi:10.1002/cncr.20160

95. Maddocks M, Jones LW, Wilcock A. Immunological and hormonal effects of exercise: implications for cancer cachexia. Curr Opin Support Palliat Care. 2013;7(4):376–382. doi:10.1097/SPC.0000000000000010

96. Petersen AM, Pedersen BK. The anti-inflammatory effect of exercise. J Appl Physiol. 2005;98(4):1154–1162. doi:10.1152/japplphysiol.00164.2004

97. Lira FS, Neto JC, Seelaender M. Exercise training as treatment in cancer cachexia. Appl Physiol Nutr Metab. 2014;39(6):679–686. doi:10.1139/apnm-2013-0554

98. Lira FS, Yamashita AS, Rosa JC, et al. Exercise training decreases adipose tissue inflammation in cachectic rats. Horm Metab Res. 2012;44(2):91–98. doi:10.1055/s-0031-1299694

99. Murton AJ, Greenhaff PL. Resistance exercise and the mechanisms of muscle mass regulation in humans: acute effects on muscle protein turnover and the gaps in our understanding of chronic resistance exercise training adaptation. Int J Biochem Cell Biol. 2013;45(10):2209–2214. doi:10.1016/j.biocel.2013.07.005

100. Gould DW, Lahart I, Carmichael AR, Koutedakis Y, Metsios GS. Cancer cachexia prevention via physical exercise: molecular mechanisms. J Cachexia Sarcopenia Muscle. 2013;4(2):111–124. doi:10.1007/s13539-012-0096-0

101. Reid J. Psychosocial, educational and communicative interventions for patients with cachexia and their family carers. Curr Opin Support Palliat Care. 2014;8(4):334–338. doi:10.1097/SPC.0000000000000087

102. Lainscak M, Filippatos GS, Gheorghiade M, Fonarow GC, Anker SD. Cachexia: common, deadly, with an urgent need for precise definition and new therapies. Am J Cardiol. 2008;101(11A):8E–10E. doi:10.1016/j.amjcard.2008.02.065

103. Monitto CL, Dong SM, Jen J, Sidransky D. Characterization of a human homologue of proteolysis-inducing factor and its role in cancer cachexia. Clin Cancer Res. 2004;10(17):5862–5869. doi:10.1158/1078-0432.CCR-04-0435

104. Solheim TS, Laird BJA, Balstad TR, et al. A randomized phase II feasibility trial of a multimodal intervention for the management of cachexia in lung and pancreatic cancer. J Cachexia Sarcopenia Muscle. 2017;8(5):778–788. doi:10.1002/jcsm.12201

Creative Commons License © 2020 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.