Cancer Cachexia in Pancreatic Cancer Patients: Recent Advances and New Therapeutic Approach

  • Sang Hoon Lee (Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine) ;
  • Moon Jae Chung (Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine)
  • Received : 2015.12.16
  • Accepted : 2015.12.21
  • Published : 2015.12.31

Abstract

About 80% of all pancreatic cancer patients suffer from a wasting syndrome defined as the cancer cachexia characterized by abnormally low weight, weakness, and loss of skeletal muscle mass, which directly impacts physical activity, quality of life and overall survival. Over the past decades, we have gained new insights into the underlying mechanism of cachexia associated with pancreatic cancer. The aim of this review was to explore recent findings about cancer cachexia pathophysiology and describe the current pharmacologic approach. Pancreatic cancer cachexia is a multifactorial syndrome mediated by mechanical factors, inflammatory cytokines, neuropeptides, hormones and tumor-derived factors. The treatment of cancer cachexia remains controversial but is currently an active area of research. Several new targeted drugs are under investigation, and we hope to open a new prospect in the management of cancer cachexia in the future.

Keywords

References

  1. Fearon KC, Voss AC, Hustead DS, et al. Definition of cancer cachexia: effect of weight loss, reduced food intake, and systemic inflammation on functional status and prognosis. Am J Clin Nutr 2006;83:1345-1350.
  2. Inui A. Cancer anorexia-cachexia syndrome: current issues in research and management. CA Cancer J Clin 2002;52:72-91.
  3. Wigmore SJ, Plester CE, Richardson RA, et al. Changes in nutritional status associated with unresectable pancreatic cancer. Br J Cancer 1997;75:106-109.
  4. 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.
  5. Mueller TC, Burmeister MA, Bachmann J, et al. Cachexia and pancreatic cancer: are there treatment options? World J Gastroenterol 2014;20:9361-9373.
  6. Fearon KC. Cancer cachexia: developing multimodal therapy for a multidimensional problem. Eur J Cancer 2008;44:1124-1132.
  7. Ozola Zalite I, Zykus R, Francisco Gonzalez M, et al. Influence of cachexia and sarcopenia on survival in pancreatic ductal adenocarcinoma: a systematic review. Pancreatology 2015;15:19-24.
  8. Pausch T, Hartwig W, Hinz U, et al. Cachexia but not obesity worsens the postoperative outcome after pancreatoduodenectomy in pancreatic cancer. Surgery 2012;152:S81-S88.
  9. Fearon K, Strasser F, Anker SD, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 2011;12:489-495.
  10. Pannala R, Leibson CL, Rabe KG, et al. Temporal association of changes in fasting blood glucose and body mass index with diagnosis of pancreatic cancer. Am J Gastroenterol 2009;104:2318-2325.
  11. Chari ST, Leibson CL, Rabe KG, et al. Pancreatic cancer-associated diabetes mellitus: prevalence and temporal association with diagnosis of cancer. Gastroenterology 2008;134:95-101.
  12. Sah RP, Nagpal SJ, Mukhopadhyay D, et al. New insights into pancreatic cancer-induced paraneoplastic diabetes. Nat Rev Gastroenterol Hepatol 2013;10:423-433.
  13. Tisdale MJ. Cachexia in cancer patients. Nat Rev Cancer 2002;2:862-871.
  14. Imoto A, Mitsunaga S, Inagaki M, et al. Neural invasion induces cachexia via astrocytic activation of neural route in pancreatic cancer. Int J Cancer 2012;131:2795-2807.
  15. Mitsunaga S, Kinoshita T, Hasebe T, et al. Low serum level of cholinesterase at recurrence of pancreatic cancer is a poor prognostic factor and relates to systemic disorder and nerve plexus invasion. Pancreas 2008;36:241-248.
  16. Deutsch J, Kolhouse JF. Assessment of gastrointestinal function and response to megesterol acetate in subjects with gastrointestinal cancers and weight loss. Support Care Cancer 2004;12:503-510.
  17. Laviano A, Meguid MM, Rossi-Fanelli F. Cancer anorexia: clinical implications, pathogenesis, and therapeutic strategies. Lancet Oncol 2003;4:686-694.
  18. Fearon K, Arends J, Baracos V. Understanding the mechanisms and treatment options in cancer cachexia. Nat Rev Clin Oncol 2013;10:90-99.
  19. Suzuki H, Asakawa A, Amitani H, et al. Cancer cachexia pathophysiology and translational aspect of herbal medicine. Jpn J Clin Oncol 2013;43:695-705.
  20. Tuca A, Jimenez-Fonseca P, Gascon P. Clinical evaluation and optimal management of cancer cachexia. Crit Rev Oncol Hematol 2013;88:625-636.
  21. Wisse BE, Frayo RS, Schwartz MW, et al. Reversal of cancer anorexia by blockade of central melanocortin receptors in rats. Endocrinology 2001;142:3292-3301.
  22. Marks D, Cone RD. The role of the melanocortin-3 receptor in cachexia. Ann N Y Acad Sci 2003;994:258-266.
  23. Marks DL, Butler AA, Turner R, et al. Differential role of melanocortin receptor subtypes in cachexia. Endocrinology 2003;144:1513-1523.
  24. Scarlett JM, Jobst EE, Enriori PJ, et al. Regulation of central melanocortin signaling by interleukin-1 beta. Endocrinology 2007;148:4217-4225.
  25. Inui A. Cancer anorexia-cachexia syndrome: are neuropeptides the key? Cancer Res 1999;59:4493-4501.
  26. Shintani F, Kanba S, Nakaki T, et al. Interleukin-1 beta augments release of norepinephrine, dopamine, and serotonin in the rat anterior hypothalamus. J Neurosci 1993;13:3574-3581.
  27. Heisler LK, Cowley MA, Tecott LH, et al. Activation of central melanocortin pathways by fenfluramine. Science 2002;297:609-611.
  28. Llovera M, Carbo N, Lopez-Soriano J, et al. Different cytokines modulate ubiquitin gene expression in rat skeletal muscle. Cancer Lett 1998;133:83-87.
  29. Li YP, Reid MB. NF-kappaB mediates the protein loss induced by TNF-alpha in differentiated skeletal muscle myotubes. Am J Physiol Regul Integr Comp Physiol 2000;279:1165-1170.
  30. Guttridge DC, Mayo MW, Madrid LV, et al. NF-kappaB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia. Science 2000;289:2363-2366.
  31. Zhang HH, Halbleib M, Ahmad F, et al. Tumor necrosis factor-alpha stimulates lipolysis in differentiated human adipocytes through activation of extracellular signal-related kinase and elevation of intracellular cAMP. Diabetes 2002;51:2929-2935.
  32. Ryden M, Arvidsson E, Blomqvist L, et al. Targets for TNF-alpha-induced lipolysis in human adipocytes. Biochem Biophys Res Commun 2004;318:168-175.
  33. Cawthorn WP, Heyd F, Hegyi K, et al. Tumour necrosis factor-alpha inhibits adipogenesis via a beta-catenin/TCF4(TCF7L2)-dependent pathway. Cell Death Differ 2007;14:1361-1373.
  34. Hammarstedt A, Isakson P, Gustafson B, et al. Wnt-signaling is maintained and adipogenesis inhibited by TNFalpha but not MCP-1 and resistin. Biochem Biophys Res Commun 2007;357:700-706.
  35. Moses AG, Maingay J, Sangster K, et al. Pro-inflammatory cytokine release by peripheral blood mononuclear cells from patients with advanced pancreatic cancer: relationship to acute phase response and survival. Oncol Rep 2009;21:1091-1095.
  36. Martignoni ME, Dimitriu C, Bachmann J, et al. Liver macrophages contribute to pancreatic cancer-related cachexia. Oncol Rep 2009;21:363-369.
  37. Martignoni ME, Kunze P, Hildebrandt W, et al. Role of mononuclear cells and inflammatory cytokines in pancreatic cancer-related cachexia. Clin Cancer Res 2005;11:5802-5808.
  38. Todorov PT, McDevitt TM, Meyer DJ, et al. Purification and characterization of a tumor lipid-mobilizing factor. Cancer Res 1998;58:2353-2358.
  39. Hirai K, Hussey HJ, Barber MD, et al. Biological evaluation of a lipid-mobilizing factor isolated from the urine of cancer patients. Cancer Res 1998;58:2359-2365.
  40. Russell ST, Hirai K, Tisdale MJ. Role of beta3-adrenergic receptors in the action of a tumour lipid mobilizing factor. Br J Cancer 2002;86:424-428.
  41. Tan CR, Yaffee PM, Jamil LH, et al. Pancreatic cancer cachexia: a review of mechanisms and therapeutics. Front Physiol 2014;5:88.
  42. Felix K, Fakelman F, Hartmann D, et al. Identification of serum proteins involved in pancreatic cancer cachexia. Life Sci 2011;88:218-225.
  43. Todorov P, Cariuk P, McDevitt T, et al. Characterization of a cancer cachectic factor. Nature 1996;379:739-742.
  44. Wigmore SJ, Todorov PT, Barber MD, et al. Characteristics of patients with pancreatic cancer expressing a novel cancer cachectic factor. Br J Surg 2000;87:53-58.
  45. Cariuk P, Lorite MJ, Todorov PT, et al. Induction of cachexia in mice by a product isolated from the urine of cachectic cancer patients. Br J Cancer 1997;76:606-613.
  46. Belizario JE, Lorite MJ, Tisdale MJ. Cleavage of caspases-1, -3, -6, -8 and -9 substrates by proteases in skeletal muscles from mice undergoing cancer cachexia. Br J Cancer 2001;84:1135-1140.
  47. Whitehouse AS, Tisdale MJ. Increased expression of the ubiquitin-proteasome pathway in murine myotubes by proteolysis-inducing factor (PIF) is associated with activation of the transcription factor NF-kappaB. Br J Cancer 2003;89:1116-1122.
  48. Wyke SM, Tisdale MJ. NF-kappaB mediates proteolysis-inducing factor induced protein degradation and expression of the ubiquitin-proteasome system in skeletal muscle. Br J Cancer 2005;92:711-721.
  49. Eley HL, Tisdale MJ. Skeletal muscle atrophy, a link between depression of protein synthesis and increase in degradation. J Biol Chem 2007;282:7087-7097.
  50. Donohoe CL, Ryan AM, Reynolds JV. Cancer cachexia: mechanisms and clinical implications. Gastroenterol Res Pract 2011;2011:601434.
  51. Bozzetti F, Group SW. Screening the nutritional status in oncology: a preliminary report on 1,000 outpatients. Support Care Cancer 2009;17:279-284.
  52. Evans WJ, Morley JE, Argiles J, et al. Cachexia: a new definition. Clin Nutr 2008;27:793-799.
  53. 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.
  54. McCarthy HD, Crowder RE, Dryden S, et al. Megestrol acetate stimulates food and water intake in the rat: effects on regional hypothalamic neuropeptide Y concentrations. Eur J Pharmacol 1994;265:99-102.
  55. Mantovani G, Maccio A, Lai P, et al. Cytokine activity in cancer-related anorexia/cachexia: role of megestrol acetate and medroxyprogesterone acetate. Semin Oncol 1998;25:45-52.
  56. Bruera E, Macmillan K, Kuehn N, et al. A controlled trial of megestrol acetate on appetite, caloric intake, nutritional status, and other symptoms in patients with advanced cancer. Cancer 1990;66:1279-1282.
  57. Loprinzi CL, Ellison NM, Schaid DJ, et al. Controlled trial of megestrol acetate for the treatment of cancer anorexia and cachexia. J Natl Cancer Inst 1990;82:1127-1132.
  58. Loprinzi CL, Michalak JC, Schaid DJ, et al. Phase III evaluation of four doses of megestrol acetate as therapy for patients with cancer anorexia and/or cachexia. J Clin Oncol 1993;11:762-767.
  59. Westman G, Bergman B, Albertsson M, et al. Megestrol acetate in advanced, progressive, hormone-insensitive cancer. Effects on the quality of life: a placebo-controlled, randomised, multicentre trial. Eur J Cancer 1999;35:586-595.
  60. Pascual Lopez A, Roquei Figuls M, Urrutia Cuchi G, et al. Systematic review of megestrol acetate in the treatment of anorexia-cachexia syndrome. J Pain Symptom Manage 2004;27:360-369.
  61. Lesniak W, Bala M, Jaeschke R, et al. Effects of megestrol acetate in patients with cancer anorexia-cachexia syndrome--a systematic review and meta-analysis. Pol Arch Med Wewn 2008;118:636-644.
  62. 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.
  63. Bruera E, Roca E, Cedaro L, et al. Action of oral methylprednisolone in terminal cancer patients: a prospective randomized double-blind study. Cancer Treat Rep 1985;69:751-754.
  64. Plata-Salaman CR. Dexamethasone inhibits food intake suppression induced by low doses of interleukin-1 beta administered intracerebroventricularly. Brain Res Bull 1991;27:737-738.
  65. 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;17:3299-3306.
  66. Nelson K, Walsh D, Deeter P, et al. A phase II study of delta9-tetrahydrocannabinol for appetite stimulation in cancer-associated anorexia. J Palliat Care 1994;10:14-18.
  67. Jatoi A, Windschitl HE, Loprinzi CL, et al. Dronabinol versus megestrol acetate versus combination therapy for cancer-associated anorexia: a North Central Cancer Treatment Group study. J Clin Oncol 2002;20:567-573.
  68. Preston T, Fearon KC, McMillan DC, et al. Effect of ibuprofen on the acute-phase response and protein metabolism in patients with cancer and weight loss. Br J Surg 1995;82:229-234.
  69. Wigmore SJ, Falconer JS, Plester CE, et al. Ibuprofen reduces energy expenditure and acute-phase protein production compared with placebo in pancreatic cancer patients. Br J Cancer 1995;72:185-8.
  70. Lundholm K, Gelin J, Hyltander A, et al. Anti-inflammatory treatment may prolong survival in undernourished patients with metastatic solid tumors. Cancer Res 1994;54:5602-5606.
  71. McMillan DC, O'Gorman P, Fearon KC, et al. A pilot study of megestrol acetate and ibuprofen in the treatment of cachexia in gastrointestinal cancer patients. Br J Cancer 1997;76:788-790.
  72. McMillan DC, Wigmore SJ, Fearon KC, et al. A prospective randomized study of megestrol acetate and ibuprofen in gastrointestinal cancer patients with weight loss. Br J Cancer 1999;79:495-500.
  73. Sampaio EP, Sarno EN, Galilly R, et al. Thalidomide selectively inhibits tumor necrosis factor alpha production by stimulated human monocytes. J Exp Med 1991;173:699-703.
  74. Gordon JN, Trebble TM, Ellis RD, et al. Thalidomide in the treatment of cancer cachexia: a randomised placebo controlled trial. Gut 2005;54:540-545.
  75. Couluris M, Mayer JL, Freyer DR, et al. The effect of cyproheptadine hydrochloride (periactin) and megestrol acetate (megace) on weight in children with cancer/treatment-related cachexia. J Pediatr Hematol Oncol 2008;30:791-797.
  76. Kardinal CG, Loprinzi CL, Schaid DJ, et al. A controlled trial of cyproheptadine in cancer patients with anorexia and/or cachexia. Cancer 1990;65:2657-2662.
  77. Meydani SN, Lichtenstein AH, Cornwall S, et al. Immunologic effects of national cholesterol education panel step-2 diets with and without fish-derived N-3 fatty acid enrichment. J Clin Invest 1993;92:105-113.
  78. Wigmore SJ, Fearon KC, Maingay JP, et al. Down-regulation of the acute-phase response in patients with pancreatic cancer cachexia receiving oral eicosapentaenoic acid is mediated via suppression of interleukin-6. Clin Sci (Lond) 1997;92:215-221.
  79. Tisdale MJ, Beck SA. Inhibition of tumour-induced lipolysis in vitro and cachexia and tumour growth in vivo by eicosapentaenoic acid. Biochem Pharmacol 1991;41:103-107.
  80. Tisdale MJ. Inhibition of lipolysis and muscle protein degradation by EPA in cancer cachexia. Nutrition 1996;12:S31-S33.
  81. Hussey HJ, Tisdale MJ. Effect of a cachectic factor on carbohydrate metabolism and attenuation by eicosapentaenoic acid. Br J Cancer 1999;80:1231-1235.
  82. Bruera E, Strasser F, Palmer JL, et al. Effect of fish oil on appetite and other symptoms in patients with advanced cancer and anorexia/cachexia: a double-blind, placebo-controlled study. J Clin Oncol 2003;21:129-134.
  83. 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;24:3401-3407.
  84. Persson C, Glimelius B, Ronnelid J, et al. Impact of fish oil and melatonin on cachexia in patients with advanced gastrointestinal cancer: a randomized pilot study. Nutrition 2005; 21:170-178.
  85. Dewey A, Baughan C, Dean T, et al. Eicosapentaenoic acid (EPA, an omega-3 fatty acid from fish oils) for the treatment of cancer cachexia. Cochrane Database Syst Rev 2007: CD 004597.
  86. Mazzotta P, Jeney CM. Anorexia-cachexia syndrome: a systematic review of the role of dietary polyunsaturated Fatty acids in the management of symptoms, survival, and quality of life. J Pain Symptom Manage 2009;37:1069-1077.
  87. Mantovani G, Maccio A, Madeddu C, et al. Randomized phase III clinical trial of five different arms of treatment in 332 patients with cancer cachexia. Oncologist 2010;15:200-211.
  88. 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;12:62-67.
  89. Rigas JR, Schuster M, Orlov SV, et al. Efect of ALD518, a humanized anti-IL-6 antibody, on lean body mass loss and symptoms in patients with advanced non-small cell lung cancer (NSCLC): Results of a phase II randomized double-blind safety and efficacy trial.J Clin Oncol 2010;28s.Abstracts 7622.
  90. 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;28s.Abstracts 7631.
  91. Kojima M, Hosoda H, Date Y, et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999;402:656-660.
  92. Gonzalez PV, Cragnolini AB, Schioth HB, et al. Interleukin-1 beta-induced anorexia is reversed by ghrelin. Peptides 2006;27:3220-3225.
  93. 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;143:334-342.
  94. 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.
  95. 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.
  96. 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.
  97. Garcia JM, Friend J, Allen S. Therapeutic potential of anamorelin, a novel, oral ghrelin mimetic, in patients with cancerrelated cachexia: a multicenter, randomized, double-blind, crossover, pilot study. Support Care Cancer 2013;21:129-137.
  98. Broglio F, Boutignon F, Benso A, et al. EP1572: a novel peptidomimetic GH secretagogue with potent and selective GH-releasing activity in man. J Endocrinol Invest 2002;25:RC26-28.
  99. Stewart Coats AJ, Srinivasan V, Surendran J, et al. The ACTONE trial, a multicentre, randomised, double-blind, placebo-controlled, dose-finding study of the anabolic/catabolic transforming agent, MT-102 in subjects with cachexia related to stage III and IV non-small cell lung cancer and colorectal cancer: study design. J Cachexia Sarcopenia Muscle 2011;2:201-207.
  100. Ma JD, Heavey SF, Revta C, et al. Novel investigational biologics for the treatment of cancer cachexia. Expert Opin Biol Ther 2014;14:1113-1120.
  101. Zhou X, Wang JL, Lu J, et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell 2010;142:531-543.
  102. Bachmann J, Ketterer K, Marsch C, et al. Pancreatic cancer related cachexia: influence on metabolism and correlation to weight loss and pulmonary function. BMC Cancer 2009;9:255.