487
Views
4
CrossRef citations to date
0
Altmetric
Review

An overview of anamorelin as a treatment option for cancer-associated anorexia and cachexia

&
Pages 889-895 | Received 27 Oct 2020, Accepted 06 Jan 2021, Published online: 25 Jan 2021

References

  • Fearon K, Strasser F, Anker SD, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12:489–495.
  • Cederholm T, Jensen GL, Correia MITD, et al. GLIM criteria for the diagnosis of malnutrition - A consensus report from the global clinical nutrition community. Clin Nutr. 2019;38:1–9.
  • Cole CL, Kleckner IR, Jatoi A, et al. The Role of Systemic Inflammation in Cancer-Associated Muscle Wasting and Rationale for Exercise as a Therapeutic Intervention. JCSM Clin Rep. 2018; 3.
  • Suzuki H, Asakawa A, Amitani H, et al. Cancer cachexia–pathophysiology and management. J Gastroenterol. 2013;48:574–594.
  • Fonseca GWPD, Farkas J, Dora E, et al. Cancer Cachexia and Related Metabolic Dysfunction. Int J Mol Sci. 2020;21(7):2321.
  • Peixoto da Silva S, Santos JMO. Cancer cachexia and its pathophysiology: links with sarcopenia, anorexia and asthenia. J Cachexia Sarcopenia Muscle. 2020;11:619–635.
  • Mytelka DS, Li L, Benoit K. Post-diagnosis weight loss as a prognostic factor in non-small cell lung cancer. J Cachexia Sarcopenia Muscle. 2018;9:86–92.
  • Choi MH, Yoon SB, Lee K, et al. Preoperative sarcopenia and post-operative accelerated muscle loss negatively impact survival after resection of pancreatic cancer. J Cachexia Sarcopenia Muscle. 2018;9:326–334.
  • Daly L, Dolan R, Power D, et al. The relationship between the BMI-adjusted weight loss grading system and quality of life in patients with incurable cancer. J Cachexia Sarcopenia Muscle. 2020;11:160–168.
  • Hacker UT, Hasenclever D, Linder N, et al. Prognostic role of body composition parameters in gastric/gastroesophageal junction cancer patients from the EXPAND trial. J Cachexia Sarcopenia Muscle. 2020;11:135–144.
  • Lee J, Lin JB, Wu MH, et al. Muscle radiodensity loss during cancer therapy is predictive for poor survival in advanced endometrial cancer. J Cachexia Sarcopenia Muscle. 2019;10:814–826.
  • Ali R, Baracos VE, Sawyer MB, et al. Lean body mass as an independent determinant of dose-limiting toxicity and neuropathy in patients with colon cancer treated with FOLFOX regimens. Cancer Med. 2016;5:607–616.
  • 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;69:491–497.
  • Wolf I, Sadetzki S, Kanety H, et al. Adiponectin, ghrelin, and leptin in cancer cachexia in breast and colon cancer patients. Cancer. 2006;106:966–973.
  • Mir O, Coriat R, Blanchet B, et al. Sarcopenia predicts early dose-limiting toxicities and pharmacokinetics of sorafenib in patients with hepatocellular carcinoma. PLoS One. 2012;7:e37563.
  • 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;10:22–34.
  • Ruiz-García V, López-Briz E, Carbonell-Sanchis R, et al. Megestrol acetate for cachexia-anorexia syndrome. A systematic review. J Cachexia Sarcopenia Muscle. 2018;9:444–452.
  • Dev R, Wong A, Hui D, et al. The Evolving Approach to Management of Cancer Cachexia. Oncology (Williston Park). 2017;31:23–32.
  • Yennurajalingam S, Bruera E. Role of corticosteroids for fatigue in advanced incurable cancer: is it a ‘wonder drug’ or ‘deal with the devil’. Curr Opin Support Palliat Care. 2014;8:346–351.
  • Advani SM, Advani PG, VonVille HM, et al. Pharmacological management of cachexia in adult cancer patients: a systematic review of clinical trials. BMC Cancer. 2018;18:1174.
  • 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;118:4785–4794.
  • 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;89:2832–2836.
  • 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;17:519–531.
  • Takayama K, Katakami N, Yokoyama T, et al. Anamorelin (ONO-7643) in Japanese patients with non-small cell lung cancer and cachexia: results of a randomized phase 2 trial. Support Care Cancer. 2016;24:3495–3505.
  • Katakami N, Uchino J, Yokoyama T, et al. Anamorelin (ONO-7643) for the treatment of patients with non-small cell lung cancer and cachexia: results from a randomized, double-blind, placebo-controlled, multicenter study of Japanese patients (ONO-7643-04). Cancer. 2018;124:606–616.
  • 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;21:129–137.
  • Northrup R, Kuroda K, Duus E, 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;21:2409–2415.
  • Khatib MN, Gaidhane A, Gaidhane S, et al. Ghrelin as a Promising Therapeutic Option for Cancer Cachexia. Cell Physiol Biochem. 2018;48:2172–2188.
  • Castañeda TR, Tong J, Datta R, et al. Ghrelin in the regulation of body weight and metabolism. Front Neuroendocrinol. 2010;31:44–60.
  • Guillory B, Splenser A, Garcia J. The role of ghrelin in anorexia-cachexia syndromes. Vitam Horm. 2013;92:61–106.
  • Sohn JW. Network of hypothalamic neurons that control appetite. BMB Rep. 2015;48:229–233.
  • 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;37:649–661.
  • Brown DJ, Milroy R, Preston T, et al. The relationship between an inflammation-based prognostic score (Glasgow Prognostic Score) and changes in serum biochemical variables in patients with advanced lung and gastrointestinal cancer. J Clin Pathol. 2007;60:705–708.
  • Tschöp M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature. 2000;407:908–913.
  • Fouladiun M, Körner U, Bosaeus I, et al. Body composition and time course changes in regional distribution of fat and lean tissue in unselected cancer patients on palliative care–correlations with food intake, metabolism, exercise capacity, and hormones. Cancer. 2005;103:2189–2198.
  • Shimizu Y, Nagaya N, Isobe T, et al. Increased plasma ghrelin level in lung cancer cachexia. Clin Cancer Res. 2003;9:774–778.
  • Karapanagiotou EM, Polyzos A, Dilana KD, et al. Increased serum levels of ghrelin at diagnosis mediate body weight loss in non-small cell lung cancer (NSCLC) patients. Lung Cancer. 2009;66:393–398.
  • 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;90:2920–2926.
  • Argilés JM, Stemmler B, López-Soriano FJ, et al. Inter-tissue communication in cancer cachexia. Nat Rev Endocrinol. 2018;15:9–20.
  • Lainscak M, Rosano GMC. Cancer cachexia: an orphan with a future. J Cachexia Sarcopenia Muscle. 2019;10:3–5.
  • 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.
  • Yasuda T, Masaki T, Kakuma T, et al. Centrally administered ghrelin suppresses sympathetic nerve activity in brown adipose tissue of rats. Neurosci Lett. 2003;349:75–78.
  • Lundholm K, Gunnebo L, Körner U, et al. Effects by daily long term provision of ghrelin to unselected weight-losing cancer patients: a randomized double-blind study. Cancer. 2010;116:2044–2052.
  • PorporatoPE, Filigheddu N, Reano S, et al. Acylated and unacylated ghrelin impair skeletal muscle atrophy in mice. J Clin Invest. 2013;123:611–622.
  • Filigheddu N, Gnocchi VF, Coscia M, et al. Ghrelin and des-acyl ghrelin promote differentiation and fusion of C2C12 skeletal muscle cells. Mol Biol Cell. 2007;18:986–994.
  • Sheriff S, Kadeer N, Joshi R, et al. Des-acyl ghrelin exhibits pro-anabolic and anti-catabolic effects on C2C12 myotubes exposed to cytokines and reduces burn-induced muscle proteolysis in rats. Mol Cell Endocrinol. 2012;351:286–295.
  • 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.
  • 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;149:601–611.
  • Campbell GA, Patrie JT, Gaylinn BD, et al. Oral ghrelin receptor agonist MK-0677 increases serum insulin-like growth factor 1 in hemodialysis patients: a randomized blinded study. Nephrol Dial Transplant. 2018;33:523–530.
  • Pietra C, Takeda Y, Tazawa-Ogata N, et al. Anamorelin HCl (ONO-7643), a novel ghrelin receptor agonist, for the treatment of cancer anorexia-cachexia syndrome: preclinical profile. J Cachexia Sarcopenia Muscle. 2014;5:329–337.
  • Blum RA, Mair S, Duus EM. Appetite and food intake results from phase I studies of anamorelin. J Cachexia Sarcopenia Muscle. 2019;10:1027–1035.
  • Dc Tj C, Abernethy AP, Friend J, et al. Body weight response with anamorelin in advanced non-small cell lung
  • Currow D, Temel JS, Abernethy A, et al. ROMANA 3: a phase 3 safety extension study of anamorelin in advanced non-small-cell lung cancer (NSCLC) patients with cachexia. Ann Oncol. 2017;28:1949–1956.
  • 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;16:108–116.
  • Kilgour RD, Vigano A, Trutschnigg B, et al. Handgrip strength predicts survival and is associated with markers of clinical and functional outcomes in advanced cancer patients. Support Care Cancer. 2013;21:3261–3270.
  • Crawford J. What are the criteria for response to cachexia treatment? Ann Palliat Med. 2019;8:43–49.
  • Mitchell T, Clarke L, Goldberg A, et al. Pancreatic Cancer Cachexia: the Role of Nutritional Interventions. Healthcare (Basel). 2019;7(3):89.
  • Chung HY, Park YK. Rationale, Feasibility and Acceptability of Ketogenic Diet for Cancer Treatment. J Cancer Prev. 2017;22:127–134.
  • Strasser B, Steindorf K, Wiskemann J, et al. Impact of resistance training in cancer survivors: a meta-banalysis. Med Sci Sports Exerc. 2013;45:2080–2090.
  • Cermak NM, Res PT, de Groot LC, et al. Protein supplementation augments the adaptive response of skeletal muscle to resistance-type exercise training: a meta-analysis. Am J Clin Nutr. 2012;96:1454–1464.
  • Arrieta H, Astrugue C, Regueme S, et al. Effects of a physical activity programme to prevent physical performance decline in onco-geriatric patients: a randomized multicentre trial. J Cachexia Sarcopenia Muscle. 2019;10:287–297.
  • Maddocks M, Jones LW, Wilcock A. Immunological and hormonal effects of exercise: implications for cancer cachexia. Curr Opin Support Palliat Care. 2013;7:376–382.
  • Grande AJ, Silva V, Maddocks M. Exercise for cancer cachexia in adults: executive summary of a Cochrane Collaboration systematic review. J Cachexia Sarcopenia Muscle. 2015;6:208–211.
  • Maddocks M, Murton AJ, Wilcock A. Therapeutic exercise in cancer cachexia. Crit Rev Oncog. 2012;17:285–292.
  • Hall CC, Cook J, Maddocks M, et al. Combined exercise and nutritional rehabilitation in outpatients with incurable cancer: a systematic review. Support Care Cancer. 2019;27:2371–2384.
  • Bird SR, Hawley JA. Update on the effects of physical activity on insulin sensitivity in humans. BMJ Open Sport Exerc Med. 2016;2:e000143.
  • Sargeant JA, Henson J, JA K, et al. A Review of the Effects of Glucagon-Like Peptide-1 Receptor Agonists and Sodium-Glucose Cotransporter 2 Inhibitors on Lean Body Mass in Humans. Endocrinol Metab (Seoul). 2019;34:247–262.
  • Nagai Y, Fukuda H, Kawanabe S, et al. Differing Effect of the Sodium-Glucose Cotransporter 2 Inhibitor Ipragliflozin on the Decrease of Fat Mass vs. Lean Mass in Patients With or Without Metformin Therapy. J Clin Med Res. 2019;11:297–300.
  • Aniort J, Stella A, Philipponnet C, et al. Muscle wasting in patients with end-stage renal disease or early-stage lung cancer: common mechanisms at work. J Cachexia Sarcopenia Muscle. 2019;10:323–337.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.