touchONCOLOGY touchONCOLOGY
Supportive Cancer Care
Read Time: 7 mins

The Potential of Ghrelin in Cancer Anorexia–Cachexia

Copy Link
Published Online: Jan 13th 2013 European Oncology & Haematology, 2013;9(2):77–83 DOI: https://doi.org/10.17925/EOH.2013.09.2.77
Authors: Jose M Garcia, Aminah Jatoi, Egidio Del Fabbro
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

The cancer anorexia–cachexia syndrome (CACS) is often seen in patients with incurable malignancies and is associated with increased mortality, decreased efficacy of anticancer treatments and poor quality of life. However, there are currently no effective therapies for CACS. Since CACS is a multifactorial syndrome with a complex pathophysiology, therapeutic interventions for CACS might potentially include anabolic agents as well as anti-inflammatory effects. Ghrelin affects numerous key pathways in the regulation of body weight and body composition through increased appetite and growth hormone (GH) secretion. Preliminary clinical data indicates that the administration of ghrelin and ghrelin receptor agonist such as anamorelin have beneficial effects on appetite and body weight in patients with CACS. Anamorelin is currently in phase III clinical trials.

Keywords

Anamorelin, cancer anorexia–cachexia syndrome, ghrelin

Article:

Cachexia is a complex metabolic disorder that results in progressive weight loss, muscle and adipose tissue wasting and inflammation and is frequently observed in patients with incurable malignancies.1 Weight loss in cancer patients results from reduction in adipose tissue and skeletal mass. Anorexia is defined as the loss of desire to eat. Although anorexia frequently accompanies cachexia, there does not appear to be a cause–effect relationship between the two. Anorexia occurs in almost half of patients with cancer,2,3 and can be present even in patients not receiving chemotherapy.4 Although our understanding of cachexia has increased over the last decade, lack of consensus on its definition, diagnostic criteria and classification have impaired the development of therapeutic interventions. In 2011, an international consensus panel defined cancer anorexia–cachexia syndrome (CACS) as a multifactorial group of signs and symptoms defined by ongoing loss of skeletal muscle mass (with or without loss of fat mass) and by the fact that these changes are not able to be fully reversed by conventional nutritional support. The result of this detrimental process is functional impairment and early demise.5

The incidence of CACS depends on the tumour type and ranges from 16 % to over 50 % of patients.6,7 The degree of severity also varies, with loss of more than 10 % body weight in 15 % of patients.8 The highest incidence occurs in patients with solid tumours, in particular pancreatic and gastric cancers, where weight loss is seen in over 80 % of patients. The lowest incidence is seen in patients with non- Hodgkin lymphoma, breast cancer, acute non-lymphocytic leukaemia and sarcomas.9 More than 50 % of patients with cancer die with CACS being present.7 It should be noted, however, that the percentage above are heavily influenced by the scope of practice or clinical setting from which they have been derived, and, in this review, we point out that CACS is by definition observed in patients with advanced, incurable malignancies.

CACS is associated with poor QoL,1,10–13 poor physical function,10,12 decreased response to therapy,14,15 decreased tolerance to therapy1,5,16 and poor prognosis.9,10,17,18 A meta-analysis of 30 randomised controlled trials from the European Organisation for Research and Treatment of Cancer (EORTC) found a significant correlation between poor appetite and poor prognosis.19 The predictive value of CACS is independent of disease stage and performance status.

Diagnostic criteria for CACS were recently established by a consensus group and are summarised in Table 1. The same group concluded that cachexia progresses through three stages. Precachexia involves a weight loss ≤5 %, anorexia and metabolic change. Cachexia comprises a weight loss >5 % or body mass index (BMI) <20 and weight loss >2 % or sarcopenia and weight loss >2 %. In refractory CACS, reversal of weight loss is no longer possible since the cancer is both procatabolic and is unresponsive to treatment. Refractory CACS is characterised by a low performance score (World Health Organization [WHO] 3 or 4) and an expected survival of less than 3 months.5 However, it must be stressed that CACS is a continuum and not all patients traverse the entire spectrum. Importantly, these criteria have not been validated clinically, but they provide a meaningful scaffold that may lead to future research. Recently published retrospective data found that, before the publication of these criteria, CACS was under-diagnosed. While 49 % of the sample cohort met at least one of the weight criteria for CACS, only 5 % were prescribed medication for the condition.20 There are currently no standard effective treatments for CACS,10 although it affects most metastatic cancer patients at some point during their disease course. This article aims to review the clinical features and pathogenesis of CACS, the role of ghrelin and its potential as a therapeutic strategy in CACS.

Pathophysiology and Clinical Features of Cancer Anorexia–Cachexia Syndrome
Clinical features of CACS include weight loss in adults or growth failure in children (excluding endocrine disorders), anorexia, inflammation, insulin resistance and increased muscle protein breakdown.21 There is often reduced food intake and systemic inflammation. It is a multi-organ syndrome that involves the liver, heart and fat, but its most important target is skeletal muscle, since this represents over 40 % of total body weight.22 The weight loss associated with CACS is a result of altered metabolism with decreased energy intake (reduced appetite, early satiety, changes in taste/smell) and possibly also changes in energy needs.5,23 Other symptoms associated with CACS include anaemia, hypogonadism, immunodepression, resistance to antineoplastic agents and increased treatment-related toxicities.5,24

The pathophysiological changes responsible for these symptoms are summarised in Figure 1 and can be attributed to tumour-derived factors, cytokines and neuroendocrine changes. Other factors that contribute to anorexia include altered taste perception, side effects of therapy and psychological factors, including depression.23 CACS is associated with an increase in muscle protein degradation,26 largely mediated by the ubiquitin–proteasome pathway26 and a decrease in protein synthesis.27 There is an increase in lipolysis, leading to a loss of adipose tissue.28,29 Furthermore, interaction between host cells and cancer cells causes the release of pro-inflammatory cytokines, including tumour necrosis factor-a (TNF-a) and interleukin-1b (IL-1b), IL-6 and IL-8. These stimulate proteolytic pathways, resulting in muscle atrophy and the breakdown of adipose tissue.30 They also increase basal energy expenditure31 and induce anorexia.32

The pathogenesis of cancer anorexia is caused by the inability of the hypothalamus to respond appropriately to peripheral signals, resulting in an imbalance between orexigenic (i.e. appetite stimulating) signals, such as neuropeptide Y (NPY) and anorexigenic signals, such as proopiomelanocortin (POMC). NPY stimulates parasympathetic activity and decreases resting energy expenditure, while POMC increases sympathetic activity and increases resting energy expenditure.8 Cytokines, including IL-1 and TNF-a, appear to mediate this effect.33,34

The Importance of Treating Cancer Anorexia– Cachexia Syndrome
Since CACS is associated with a poor prognosis, it is important to try to treat the condition, with the aim of improving symptoms and possibly also improving survival. Treatment goals in CACS include improvements in appetite, lean body mass, resting energy expenditure, quality of life (QoL), performance status and reduction of the levels of pro-inflammatory cytokines.1,12 The European Palliative Care Research Collaboration (EPCRC) has developed evidence-based recommendations for the classification and treatment of CACS in advanced cancer patients. As a minimal goal, body weight should be maintained and further loss prevented.36 Early diagnosis is central to managing disease progression and preventing unnecessary deterioration in QoL.5

Treatment options for CACS are limited and there are no standard effective treatments for this indication. Patients do not appear to benefit from nutritional supplementation alone; patients on complete parenteral nutrition still undergo weight loss, emphasising the metabolic role in CACS.8,36,37 Corticosteroids and progestational agents, such as megestrol acetate, are the most widely used treatment options but only partially alleviate CACS.38–41 Corticosteroids may be beneficial for stimulation of appetite in patients with refractory CACS. Recent evidence has also suggested a role for insulin resistance in CACS;42 insulin treatment has been found to potentially play a palliative role in CACS.43

To effectively treat CACS, it is important to target both tumour and host factors. Current and emerging therapeutic options for CACS are summarised in Table 2. Drugs with a strong rationale that have not demonstrated consistent and convincing efficacy in clinical trials to date include eicosapentaenoic acid,44 cannabinoids,45 bortezomib46 and anti-cytokine therapies including thalidomide47 and anti-TNF-alpha MoAb (infliximab).46 A recent clinical trial failed to show any benefit for melatonin.48 However, following advances in understanding of the pathological processed underlying CACS, several targeted therapies are in clinical development. These include anti-IL-6 antibodies, cytokine antagonists, myostatin inhibitors, selective androgen receptor modulators and ghrelin receptor agonists.46,49 Of these, the largest body of clinical data to date describes the efficacy and safety of ghrelin.

The Role of Ghrelin in Cancer Anorexia– Cachexia Syndrome
Ghrelin is a gastric hormone secreted in response to fasting. It stimulates appetite and increases food intake and is the endogenous ligand for the ghrelin receptor (GRLN receptor, also known as growth hormone [GH] secretagogue receptor-1a).50,51 Circulating ghrelin exists in two forms: acylated ghrelin (the biologically active form that binds and activates the GRLN receptor and des-acyl ghrelin (a form that lacks biological activity at the GRLN receptor although it has recently been suggested that it has biological activity by binding a yet unidentified receptor).52,53 Ghrelin plays a major role in a number of physiological processes including stimulation of GH secretion and regulation of energy homeostasis by a GH-independent mechanism.54,55 Increased GH secretions stimulates the production of insulin-like growth factor-1 (IGF-1). Together, GH and IGF-1 promote anabolism and increase muscle strength.

Ghrelin has multiple roles in the regulation of energy balance (see Figure 2). Although its mechanism of action is not fully understood, its action appears to involve a central hypothalamic mechanism, as well as metabolic and anti-inflammatory effects. It stimulates food intake by blocking anorexigenic mediators and stimulating the production of orexigenic substances.56,57 It also activates the mesolimbic dopamine system in the hypothalamus, affecting feeding behaviour.58 Another important role of ghrelin involves its anti-inflammatory actions.34,59 Ghrelin suppresses the production of proinflammatory cytokines (IL-1b, IL-6 and TNF-a) and stimulates the production of anti-inflammatory cytokines (IL-10).60,61 Ghrelin also inhibits the activation of nuclear factor-kB (NFKB), a transcription factor that stimulates the production of numerous proinflammatory cytokines and may be involved in protein degradation.62 In terms of metabolic effects, ghrelin promotes adiposity through the activation of lipogenic pathways in the central nervous system.34,63 Ghrelin activates white adipocytes, while inactivating brown adipocytes, resulting in decreased energy expenditure.64 It also promotes lipogenesis and decreases lipolysis and lipid oxidation in white adipose tissue in an animal model of cisplatin-induced cachexia.65 Other physiological actions of ghrelin include stimulating gastric emptying,66 increasing cardiac output and decreasing blood pressure.67 Ghrelin also stimulates the release of endogenous nitric oxide, which may partly mediate its anti-inflammatory and orexigenic actions.68,69 The combination of these actions suggests that ghrelin may have therapeutic benefits in CACS.

Data between the association between ghrelin levels and CACS are contradictory, partly due to heterogeneity of the study populations. Although one study reported reduced plasma ghrelin concentrations in cancer patients,70 total ghrelin levels in CACS patients with gastric, colon, breast and lung cancer were significantly higher than levels in patients without CACS.71–73 Furthermore, CACS has been associated with an increase in acylated ghrelin and ratio of acylated to total ghrelin levels.74 The same study suggested that CACS could be a state of ghrelin resistance. It is also possible that ghrelin levels increase to compensate for the increased metabolic rate and energy that is often observed in patients with CACS.75 Hence, it has been suggested that administration of exogenous ghrelin or ghrelin receptor agonists may have therapeutic value for muscle wasting in CACS.22,34

Clinical Use of Ghrelin in the Treatment of Cancer Anorexia–Cachexia Syndrome
Preliminary studies in healthy humans have shown that ghrelin stimulates the release of GH,76 enhances appetite and results in increased food intake.77 No serious adverse events (SAEs) were reported in these studies. In a small randomised, placebo-controlled crossover trial, ghrelin demonstrated a stimulatory effect on food intake in CACS, with an increase in patients’ meal appreciation score.78 No AEs were observed.

In a randomised, placebo-controlled, double-blind, double-crossover study of patients with CACS, nutritional intake or eating-related symptoms did not differ significantly between the ghrelin- and placebo-exposed groups, but an increase in patients’ meal appreciation, as measured by a visual analogue scale score, was observed in the ghrelin group.79 In a study of patients with solid gastrointestinal tumours, patients were randomised to either high-dose (13 mg/kg daily) or low-dose (0.7 mg/kg daily) ghrelin for 8 weeks. Appetite scores were increased significantly in the high-dose group and this group manifested a relative maintenance of whole body fat.80

In a phase II study, ghrelin therapy resulted in increased food intake, significantly better global health status scores, reduced nausea and vomiting and decreased appetite loss compared with placebo. In addition, ghrelin was associated with fewer AEs during chemotherapy and reduced duration of hospital stay.81 In another phase II study in patients with gastric cancer, short-term administration of synthetic ghrelin was safe, lessened postoperative body weight loss and improved appetite and food intake after total gastrectomy.82

Ghrelin has a very short half-life (<30 minutes) and requires parenteral administration. Studies are therefore investigating the use of ghrelin receptor agonists that have longer half-life and good bioavailability.34

The Clinical use of Ghrelin Receptor Agonists
Anamorelin (Helsinn) is a novel, orally active, non-peptidic ghrelin receptor agonist that demonstrated efficacy and safety in preclinical trials,83 and in healthy volunteers increased body weight and GH secretion with good tolerability.84,85

Clinical trial data on the safety and efficacy of anamorelin are summarised in Table 3. A multicentre, double blind, placebo-controlled, crossover study evaluated the effects of anamorelin in 16 patients with different cancers and CACS. Anamorelin significantly increased body weight, GH, IGF-1, insulin-like growth factor-binding protein 3 (IGFBP-3) and patient-reported symptoms, including appetite compared with placebo (see Figure 3).86 Mild treatment-related AEs were reported in 25 % of patients. In a phase II study of anamorelin, 81 patients were treated for 12 weeks. At 8 weeks, there was an increase in lean body mass and total body mass, as well as a significant increase in handgrip strength. There was no significant difference in QoL and AEs between treatment and placebo arms.87 A recent phase II study investigated the efficacy and safety of anamorelin in patients with non-small cell lung cancer (NSCLC). Patients (n=226) were randomised to anamorelin 100 mg or 50 mg once daily or placebo for 12 weeks. The 100 mg anamorelin group gained an average of 0.14 kg in body weight from baseline compared with mean losses of 0.3 kg and 1.32 kg for the 50 mg and placebo groups, respectively. The mean treatment difference between 100 mg anamorelin and placebo was 1.47 kg (p=0.0005). A similar percentage of patients reported at least one treatment-emergent AE in the placebo (93.5 %), 50 mg (93.4 %) and 100 mg (94.5 %) treatment arms, and the majority were unrelated to the study medication.88

Three trials: Safety and Efficacy of Anamorelin HCl in Patients With Non-Small Cell Lung Cancer–Cachexia (ROMANA 1 and ROMANA 2), and an extension study (ROMANA 3) are currently ongoing. These are phase III trials in patients with unresectable stage III/IV NSCLC and CACS. In these trials, CACS is defined as involuntary weight loss of ≥5 % body weight within 6 months prior to screening or a screening BMI <20 kg/m2. Patients will receive once-daily oral doses of anamorelin or placebo for 12 weeks. The primary endpoints of ROMANA 1 and 2 are lean body mass and muscle strength, as measured by handgrip strength. Secondary endpoints include body weight, QoL and overall survival. The primary endpoints of ROMANA 3 are concerned with safety and tolerability.89

Concern has been expressed that ghrelin may increase growth factors, such as GH and IGF-1, with a resulting stimulation of tumour growth. Furthermore, ghrelin itself may have a mitogenic potential.59 However, a recent study found that neither anamorelin nor ghrelin promoted tumour growth in an animal model, despite increased levels of GH and a trend of increased IGF-1.83

An oral ghrelin receptor agonist, macimorelin, is currently under investigation. An ongoing randomised clinical trial will explore whether macimorelin is effective and well tolerated in patients with CACS.90 Other agents in development include capromorelin.91,92 A naturally occurring splice variant of ghrelin, Dln-101, has also received approval to start phase I clinical trials, based on positive data from preclinical trials.93 Further trials are needed to establish the role of these agents in CACS.

Summary and Concluding Remarks
Since CACS is a multifactorial syndrome with a complex pathophysiology, therapeutic interventions for CACS might include anabolic effects as well as anti-inflammatory effects or, in essence, targeting more than one therapeutic pathway. A growing body of evidence indicates that ghrelin increases appetite and body weight and preserves lean body mass by decreasing protein degradation. In addition, ghrelin has numerous anti-inflammatory and metabolic effects. There is a need to generate more clinical trial data to support the role of ghrelin receptor agonists in the treatment of CACS. Results to date indicate that anamorelin is effective at stimulating appetite and increasing body weight, as well as being safe and well tolerated – results of large phase III clinical trials are eagerly awaited.

Article Information:
Disclosure

Jose M Garcia receives research support and is a consultant for Aeterna Zentaris, Inc. and Helsinn Therapeutics, Inc. Aminah Jatoi has received research funding from Novartis, XBiotech, Aveo and Enterahealth. She has served as a consultant to Helsinn. Egidio Del Fabbro has been a consultant for and on the advisory board for Helsinn Therapeutics.

Correspondence

Jose M Garcia, Assistant Professor, Michael E DeBakey Veterans Affairs Medical Center, Division of Endocrinology, Diabetes and Metabolism, Departments of Medicine and Molecular and Cell Biology, Huffington Center on Aging, Baylor College of Medicine, 2002

Support

The publication of this article was supported by Helsinn Therapeutics. The views and opinions expressed are those of the authors and not necessarily those of Helsinn Therapeutics.

Acknowledgements

This material is also based upon work supported with resources and the use of facilities at the Houston Veterans Affairs. The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the US government. Editorial assistance was provided by Katrina Mountfort at Touch Medical Media.

Received

2013-09-02T00:00:00

References

  1. Fearon K, Arends J, Baracos V, Understanding the mechanisms and treatment options in cancer cachexia, Nat Rev Clin Oncol, 2013;10:90–99.
  2. Fernandez Lopez MT, Saenz Fernandez CA, de Sas Prada MT, et al., [Malnutrition in patients with cancer; four years experience], Nutr Hosp, 2013;28:372–81.
  3. Sanchez-Lara K, Ugalde-Morales E, Motola-Kuba D, et al., Gastrointestinal symptoms and weight loss in cancer patients receiving chemotherapy, Br J Nutr, 2013;109:894–7.
  4. Tranmer JE, Heyland D, Dudgeon D, et al., Measuring the symptom experience of seriously ill cancer and noncancer hospitalized patients near the end of life with the memorial symptom assessment scale, J Pain Symptom Manage, 2003;25:420–29.
  5. Fearon K, Strasser F, Anker SD, et al., Definition and classification of cancer cachexia: an international consensus, Lancet Oncol, 2011;12:489–95.
  6. Bachmann J, Heiligensetzer M, Krakowski-Roosen H, et al., Cachexia worsens prognosis in patients with resectable pancreatic cancer, J Gastrointest Surg, 2008;12:1193–201.
  7. von Haehling S, Anker SD, Cachexia as a major underestimated and unmet medical need: facts and numbers, J Cachexia Sarcopenia Muscle, 2010;1:1–5.
  8. Tisdale MJ, Mechanisms of cancer cachexia, Physiol Rev, 2009;89:381–410.
  9. 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;69:491–7.
  10. Maccio A, Madeddu C, Mantovani G, Current pharmacotherapy options for cancer anorexia and cachexia, Expert Opin Pharmacother, 2012;13:2453–72.
  11. Ravasco P, Monteiro-Grillo I, Vidal PM, et al., Cancer: disease and nutrition are key determinants of patients’ quality of life, Support Care Cancer, 2004;12:246–52.
  12. Donohoe CL, Ryan AM, Reynolds JV, Cancer cachexia: mechanisms and clinical implications, Gastroenterol Res Pract, 2011;2011:601434.
  13. Wheelwright S, Darlington AS, Hopkinson JB, et al., A systematic review of health-related quality of life instruments in patients with cancer cachexia, Support Care Cancer, 2013;21:2625–36.
  14. Andreyev HJ, Norman AR, Oates J, et al., Why do patients with weight loss have a worse outcome when undergoing chemotherapy for gastrointestinal malignancies?, Eur J Cancer, 1998;34:503–9.
  15. Ross PJ, Ashley S, Norton A, et al., Do patients with weight loss have a worse outcome when undergoing chemotherapy for lung cancers?, Br J Cancer, 2004;90:1905–11.
  16. Kumar NB, Kazi A, Smith T, et al., Cancer cachexia: traditional therapies and novel molecular mechanism-based approaches to treatment, Curr Treat Options Oncol, 2010;11:107–17.
  17. 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;23:6240–8.
  18. Trajkovic-Vidakovic M, de Graeff A, Voest EE, et al., Symptoms tell it all: a systematic review of the value of symptom assessment to predict survival in advanced cancer patients, Crit Rev Oncol Hematol, 2012;84:130–48.
  19. Quinten C, Coens C, Mauer M, et al., Baseline quality of life as a prognostic indicator of survival: a meta-analysis of individual patient data from EORTC clinical trials, Lancet Oncol, 2009;10:865–71.
  20. Benner A, Hirsch B, Abernethy A Cancer anorexia-cachexia syndrome (CACS) is under-recognized among patients with metastatic non-small cell lung cancer (MNSCLC), Support Care in Cancer, 2013;21(Suppl. 1):Abstract MASCC-0808
  21. Evans WJ, Morley JE, Argiles J, et al., Cachexia: a new definition, Clin Nutr, 2008;27:793–9.
  22. Argiles JM, Stemmler B, The potential of ghrelin in the treatment of cancer cachexia, Expert Opin Biol Ther, 2013;13:67–76.
  23. Laviano A, Inui A, Marks DL, et al., Neural control of the anorexia-cachexia syndrome, Am J Physiol Endocrinol Metab, 2008;295:E1000–1008.
  24. Burney BO, Hayes TG, Smiechowska J, et al., Low testosterone levels and increased inflammatory markers in patients with cancer and relationship with cachexia, J Clin Endocrinol Metab, 2012;97:E700–709.
  25. Lundholm K, Bennegard K, Eden E, et al., Efflux of 3-methylhistidine from the leg in cancer patients who experience weight loss, Cancer Res, 1982;42:4807–11.
  26. Lecker SH, Solomon V, Mitch WE, et al., Muscle protein breakdown and the critical role of the ubiquitinproteasome pathway in normal and disease states, J Nutr, 1999;129:227S–37S.
  27. Warren RS, Jeevanandam M, Brennan MF, Protein synthesis in the tumor-influenced hepatocyte, Surgery, 1985;98:275–82.
  28. Bing C, Brown M, King P, et al., Increased gene expression of brown fat uncoupling protein (UCP)1 and skeletal muscle UCP2 and UCP3 in MAC16-induced cancer cachexia, Cancer Res, 2000;60:2405–10.
  29. Bing C, Trayhurn P, New insights into adipose tissue atrophy in cancer cachexia, Proc Nutr Soc, 2009;68:385–92.
  30. Langhans W, Peripheral mechanisms involved with catabolism, Curr Opin Clin Nutr Metab Care, 2002;5:419–26.
  31. Van der Poll T, Romijn JA, Endert E, et al., Tumor necrosis factor mimics the metabolic response to acute infection in healthy humans, Am J Physiol, 1991;261:E457–65.
  32. . Sonti G, Ilyin SE, Plata-Salaman CR, Anorexia induced by cytokine interactions at pathophysiological concentrations, Am J Physiol, 1996;270:R1394–402.
  33. Laviano A, Meguid MM, Rossi-Fanelli F, Cancer anorexia: clinical implications, pathogenesis, and therapeutic strategies, Lancet Oncol, 2003;4:686–94.
  34. Guillory B, Splenser A, Garcia J, The role of ghrelin in anorexiacachexia syndromes, Vitam Horm, 2013;92:61–106.
  35. Radbruch L, Elsner, F, Trottenberg, P, et al, Clinical practice guidelines on cancer cachexia in advanced cancer patients. Aachen, Department of Palliative Medicinen/European Palliative Care Research Collaborative. Avaialable at: epcrc_ cachexia_guideline_web-1.pdf (accessed 30 July 2013).
  36. Ovesen L, Allingstrup L, Hannibal J, et al., Effect of dietary counseling on food intake, body weight, response rate, survival, and quality of life in cancer patients undergoing chemotherapy: a prospective, randomized study, J Clin Oncol, 1993;11:2043–9.
  37. Bozzetti F, Gavazzi C, Mariani L, et al., Artificial nutrition in cancer patients: which route, what composition?, World J Surg, 1999;23:577–83.
  38. Loprinzi CL, Kugler JW, Sloan JA, et al., Randomized comparison of megestrol acetate versus dexamethasone versus fluoxymesterone for the treatment of cancer anorexia/ cachexia, J Clin Oncol, 1999;17:3299–306.
  39. Loprinzi CL, Bernath AM, Schaid DJ, et al., Phase III evaluation of 4 doses of megestrol acetate as therapy for patients with cancer anorexia and/or cachexia, Oncology, 1994;51(Suppl. 1):2–7.
  40. Willox JC, Corr J, Shaw J, et al., Prednisolone as an appetite stimulant in patients with cancer, Br Med J (Clin Res Ed), 1984;288:27.
  41. Pascual Lopez A, Roque i Figuls M, Urrutia Cuchi G, et al., Systematic review of megestrol acetate in the treatment of anorexia-cachexia syndrome, J Pain Symptom Manage, 2004;27:360–69.
  42. Honors MA, Kinzig KP, The role of insulin resistance in the development of muscle wasting during cancer cachexia, J Cachexia Sarcopenia Muscle, 2012;3:5–11.
  43. Lundholm K, Korner U, Gunnebo L, et al., Insulin treatment in cancer cachexia: effects on survival, metabolism, and physical functioning, Clin Cancer Res, 2007;13:2699–706.
  44. Murphy RA, Yeung E, Mazurak VC, et al., Influence of eicosapentaenoic acid supplementation on lean body mass in cancer cachexia, Br J Cancer, 2011;105:1469–73.
  45. Gorter RW, [Experiences with dronabinol (deltatetrahydrocannabinol) in oncological patients with anorexiacachexia syndrome. Illustration of clinical problems and therapy based on 2 case reports], Schmerz, 2004;18(Suppl. 2):S31–3.
  46. Tazi E, Errihani H, Treatment of cachexia in oncology, Indian J Palliat Care, 2010;16:129–37.
  47. Yennurajalingam S, Willey JS, Palmer JL, 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.
  48. Del Fabbro E, Dev R, Hui D, et al., Effects of melatonin on appetite and other symptoms in patients with advanced cancer and cachexia: a double-blind placebo-controlled trial, J Clin Oncol, 2013;31:1271–6.
  49. Ebner N, Werner CG, Doehner W, et al., Recent developments in the treatment of cachexia: highlights from the 6th Cachexia Conference, J Cachexia Sarcopenia Muscle, 2012;3:45–50.
  50. Cummings DE, Purnell JQ, Frayo RS, et al., A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans, Diabetes, 2001;50:1714–19.
  51. Inui A, Ghrelin: an orexigenic and somatotrophic signal from the stomach, Nat Rev Neurosci, 2001;2:551–60.
  52. Castaneda TR, Tong J, Datta R, et al., Ghrelin in the regulation of body weight and metabolism, Front Neuroendocrinol, 2010;31:44–60.
  53. Porporato PE, Filigheddu N, Reano S, et al., Acylated and unacylated ghrelin impair skeletal muscle atrophy in mice, J Clin Invest, 2013;123:611–22.
  54. Kojima M, Hosoda H, Date Y, et al., Ghrelin is a growthhormone- releasing acylated peptide from stomach, Nature, 1999;402:656–60.
  55. Takaya K, Ariyasu H, Kanamoto N, et al., Ghrelin strongly stimulates growth hormone release in humans, J Clin Endocrinol Metab, 2000;85:4908–11.
  56. van der Lely AJ, Tschop M, Heiman ML, et al., Biological, physiological, pathophysiological, and pharmacological aspects of ghrelin, Endocr Rev, 2004;25:426–57.
  57. Kojima M, Kangawa K, Ghrelin: structure and function, Physiol Rev, 2005;85:495–522.
  58. Jerlhag E, Systemic administration of ghrelin induces conditioned place preference and stimulates accumbal dopamine, Addict Biol, 2008;13:358–63.
  59. Akamizu T, Kangawa K, Ghrelin for cachexia, J Cachexia Sarcopenia Muscle, 2010;1:169–76.
  60. Waseem T, Duxbury M, Ito H, et al., Exogenous ghrelin modulates release of pro-inflammatory and anti-inflammatory cytokines in LPS-stimulated macrophages through distinct signaling pathways, Surgery, 2008;143:334–42.
  61. Dixit VD, Schaffer EM, Pyle RS, et al., Ghrelin inhibits leptin- and activation-induced proinflammatory cytokine expression by human monocytes and T cells, J Clin Invest, 2004;114:57–66.
  62. Li WG, Gavrila D, Liu X, et al., Ghrelin inhibits proinflammatory responses and nuclear factor-kappaB activation in human endothelial cells, Circulation, 2004;109:2221–6.
  63. Cowley MA, Smith RG, Diano S, et al., The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis, Neuron, 2003;37:649–61.
  64. Mano-Otagiri A, Iwasaki-Sekino A, Nemoto T, et al., Genetic suppression of ghrelin receptors activates brown adipocyte function and decreases fat storage in rats, Regul Pept, 2010;160:81–90.
  65. Garcia JM, Scherer T, Chen JA, et al., Inhibition of Cisplatininduced lipid catabolism and weight loss by ghrelin in male mice, Endocrinology, 2013;154:3118–29.
  66. Peeters TL, Central and peripheral mechanisms by which ghrelin regulates gut motility, J Physiol Pharmacol, 2003;54(Suppl. 4):95–103.
  67. Nagaya N, Uematsu M, Kojima M, et al., Chronic administration of ghrelin improves left ventricular dysfunction and attenuates development of cardiac cachexia in rats with heart failure, Circulation, 2001;104:1430–35.
  68. Sibilia V, Rindi G, Pagani F, et al., Ghrelin protects against ethanol-induced gastric ulcers in rats: studies on the mechanisms of action, Endocrinology, 2003;144:353–9.
  69. Konturek PC, Brzozowski T, Engel M, et al., Ghrelin ameliorates colonic inflammation. Role of nitric oxide and sensory nerves, J Physiol Pharmacol, 2009;60:41–7.
  70. Legakis I, Stathopoulos J, Matzouridis T, et al., Decreased plasma ghrelin levels in patients with advanced cancer and weight loss in comparison to healthy individuals, Anticancer Res, 2009;29:3949–52.
  71. Shimizu Y, Nagaya N, Isobe T, et al., Increased plasma ghrelin level in lung cancer cachexia, Clin Cancer Res, 2003;9:774–8.
  72. Wolf I, Sadetzki S, Kanety H, et al., Adiponectin, ghrelin, and leptin in cancer cachexia in breast and colon cancer patients, Cancer, 2006;106:966–73.
  73. Kerem M, Ferahkose Z, Yilmaz UT, et al., Adipokines and ghrelin in gastric cancer cachexia, World J Gastroenterol, 2008;14:3633–41.
  74. Garcia JM, Garcia-Touza M, Hijazi RA, et al., Active ghrelin levels and active to total ghrelin ratio in cancer-induced cachexia, J Clin Endocrinol Metab, 2005;90:2920–26.
  75. Nagaya N, Uematsu M, Kojima M, et al., Elevated circulating level of ghrelin in cachexia associated with chronic heart failure: relationships between ghrelin and anabolic/catabolic factors, Circulation, 2001;104:2034–8.
  76. Akamizu T, Takaya K, Irako T, et al., Pharmacokinetics, safety, and endocrine and appetite effects of ghrelin administration in young healthy subjects, Eur J Endocrinol, 2004;150:447–55.
  77. Wren AM, Seal LJ, Cohen MA, et al., Ghrelin enhances appetite and increases food intake in humans, J Clin Endocrinol Metab, 2001;86:5992.
  78. Neary NM, Small CJ, Wren AM, et al., Ghrelin increases energy intake in cancer patients with impaired appetite: acute, randomized, placebo-controlled trial, J Clin Endocrinol Metab, 2004;89:2832–6.
  79. Strasser F, Lutz TA, Maeder MT, et al., Safety, tolerability and pharmacokinetics of intravenous ghrelin for cancerrelated anorexia/cachexia: a randomised, placebo-controlled, double-blind, double-crossover study, Br J Cancer, 2008;98:300–8.
  80. Lundholm K, Gunnebo, L, Korner U, et al., Effects by daily long term provision of ghrelin to unselected weight-losing cancer patients: a randomized double-blind study, Cancer, 2011;116:2044–52.
  81. Hiura Y, Takiguchi S, Yamamoto K, et al., Effects of ghrelin administration during chemotherapy with advanced esophageal cancer patients: a prospective, randomized, placebo-controlled phase 2 study, Cancer, 2012;118:4785–94.
  82. Adachi S, Takiguchi S, Okada K, et al., Effects of ghrelin administration after total gastrectomy: a prospective, randomized, placebo-controlled phase II study, Gastroenterology, 2010;138:1312–20.
  83. Northrup R, Kuroda K, Duus EM, et al., Effect of ghrelin and anamorelin (ONO-7643), a selective ghrelin receptor agonist, on tumor growth in a lung cancer mouse xenograft model, Support Care Cancer, 2013;21:2409–15
  84. Garcia JM, Polvino WJ, Pharmacodynamic hormonal effects of anamorelin, a novel oral ghrelin mimetic and growth hormone secretagogue in healthy volunteers, Growth Horm IGF Res, 2009;19:267–73.
  85. Garcia JM, Polvino WJ, Effect on body weight and safety of RC-1291, a novel, orally available ghrelin mimetic and growth hormone secretagogue: results of a phase I, randomized, placebo-controlled, multiple-dose study in healthy volunteers, Oncologist, 2007;12:594–600.
  86. Garcia JM, Friend J, Allen S, Therapeutic potential of anamorelin, a novel, oral ghrelin mimetic, in patients with cancer-related cachexia: a multicenter, randomized, double-blind, crossover, pilot study, Support Care Cancer, 2013;21:129–37.
  87. Garcia JB, V; Graham, C et al., 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(S):9133.
  88. Temel JB, S; Jain, M, et al., Efficacy and Safety of Anamorelin HCl in NSCLC Patients: Results from a Randomized, Double-Blind, Placebo-controlled, Multicenter Phase II Study, Presented at the European Cancer Congress, 27 September–1 Oct 2013, Amsterdam, Netherlands; Abstract no. 1308.
  89. Abernethy A, Temel, Jl, Currow D, et al., Phase III clinical trials with anamorelin HCl, a novel oral treatment for NSCLC cachexia, 2013, Clin Oncol, (Suppl. 31):abstract no TPS9649.
  90. Ali SA, Garcia JM, Randomized clinical trial of the novel oral ghrelin mimetic macimorelin in the treatment of cancer cachexia: study design and preliminary results., Endocr Rev, 2013;34(03_MeetingAbstracts): MON-327.
  91. White HK, Petrie CD, Landschulz W, et al., Effects of an oral growth hormone secretagogue in older adults, J Clin Endocrinol Metab, 2009;94:1198–206.
  92. Nass R, Pezzoli SS, Oliveri MC, et al., Effects of an oral ghrelin mimetic on body composition and clinical outcomes in healthy older adults: a randomized trial, Ann Intern Med, 2008;149:601–11.
  93. Mintz L, Dln101 a naturally occuring ghrelin splice variant for the treatment of cachexia, J Cachexia Sarcopenia Muscle, 2011;2:209–61.
  94. Fearon KC, Barber MD, Moses AG, et al., Double-blind, placebo-controlled, randomized study of eicosapentaenoic acid diester in patients with cancer cachexia, J Clin Oncol, 2006;24:3401–7.
  95. Cannabis In Cachexia Study G, Strasser F, Luftner D, et al., Comparison of orally administered cannabis extract and delta-9-tetrahydrocannabinol in treating patients with cancer-related anorexia-cachexia syndrome: a multicenter, phase III, randomized, double-blind, placebo-controlled clinical trial from the Cannabis-In-Cachexia-Study-Group, J Clin Oncol, 2006;24:3394–400.
  96. Gordon JN, Trebble TM, Ellis RD, et al., Thalidomide in the treatment of cancer cachexia: a randomised placebo controlled trial, Gut, 2005;54:540–45.
  97. 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;30:67–74.
  98. Braiteh FD S, Khuwaja A, et al., David H, Bruera E, Kurzrock R. Phase I pilot study of the safety and tolerability of olanzapine (OZA) for the treatment of cachexia in patients with advanced cancer, J Clin Oncol, 2008;26:196–203.
  99. Lesser GJ, Case D, Ottery, F, et al., ASCO Meeting. A phase III randomized study comparing the effects of oxandrolone (Ox) and megestrol acetate (Meg) on lean body mass (LBM), weight (wt) and quality of life (QOL) in patients with solid tumors and weight loss receiving chemotherapy, Proc Am Soc Clin Onc, 2008;26:505s.
  100. Eley HL, Russell ST, Tisdale MJ, Effect of branched-chain amino acids on muscle atrophy in cancer cachexia, Biochem J, 2007;407:113–20.
  101. Foster AC, Chen C, Melanocortin-4 receptor antagonists as potential therapeutics in the treatment of cachexia, Curr Top Med Chem, 2007;7:1131–6.
  102. Kenley RA, Denissenko MF, Mullin RJ, et al., Formoterol fumarate and roxithromycin effects on muscle mass in an animal model of cancer cachexia, Oncol Rep, 2008;19:1113–21.
  103. Zhang L, Rajan V, Lin E, et al., Pharmacological inhibition of myostatin suppresses systemic inflammation and muscle atrophy in mice with chronic kidney disease, FASEB J, 2011;25:1653–63.
  104. Murakami MM T, Aoki C, et al, Blocking interleukin-6 signal ameliorates inflammatory manifestations and laboratories of cachexia in a patient with malignant mesothelioma: a case study, J Cachexia Sarcopenia Muscle, 2011;2:209–61.
  105. Dalton JB 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;2:153–61.
  106. Thum T, Springer J, Breakthrough in cachexia treatment through a novel selective androgen receptor modulator?!, J Cachexia Sarcopenia Muscle, 2011;2:121–3.
  107. Dodson SH ML, Johnston MA, et al., GTx-024, a selective androgen receptor modulator (SARM), improves physical function in non-small cell lung cancer (NSCLC) patients with muscle wasting, J Cachexia Sarcopenia Muscle, 2011;2:209–61.
  108. 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;14:335–45.
  109. Crawford JD JT, Hancock ML, et al., Results from two Phase 3 randomized trials of enobosarm, selective androgen receptor modulator (SARM), for the prevention and treatment of muscle wasting in NSCLC, Eur J Cancer, 2013;49 (Suppl. 3):LBA21.
  110. Paspaliaris VL B, DeAndrea R, et al., Phase I/II study of IP-1510 a novel interleukin-1 receptor antagonist in the management of cancer-related cachexia, J Cachexia Sarcopenia Muscle, 2011;2:209–61.

Further Resources

Share this Article
Related Content In Supportive Cancer Care
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72