Korean J Gastroenterol.  2019 Aug;74(2):87-94. 10.4166/kjg.2019.74.2.87.

Nutritional Support for Patients with Pancreatic Cancer

Affiliations
  • 1Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea. minky1973@knu.ac.kr
  • 2Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Korea.

Abstract

Pancreatic cancer is the ninth common malignancy in South Korea. It has a dismal prognosis with a 5-year overall survival rate of less than 10%, and pancreatic cancer is associated with cancer cachexia, which is defined as the loss of muscle mass that is not reversible by conventional nutritional support. Cachexia is noted in over 85% of all pancreatic cancer patients and it is strongly related with the disease's mortality. Nearly 30% of pancreatic cancer deaths are due to cachexia rather than being due to the tumor burden. Therefore, it is crucial to discover the mechanisms behind the development of muscle wasting in pancreatic cancer patients and find novel therapeutics for targeting cachexia. This review deals with the current understanding about the development of cachexia and nutritional support in those patients suffering with pancreatic cancer.

Keyword

Nutritional support; Pancreatic neoplasms; Cachexia

MeSH Terms

Cachexia
Humans
Korea
Mortality
Nutritional Support*
Pancreatic Neoplasms*
Prognosis
Survival Rate
Tumor Burden

Figure

  • Fig. 1 Proinflammatory cytokines are drivers of cancer-induced cachexia. Proinflammatory cytokines IL-6 and TNF-α activate downstream signaling of NF-kB and this results in overstimulation of NF-kB and degradation of muscle proteins through the production of catabolic cytokines. Cytokines can also activate the Jak2/STAT3 signaling pathways that can lead to degradation of skeletal muscle protein. TNF-α, tumor necrosis factor α; MuRF1, muscle RING finger-containing protein 1; IL, interleukin.


Reference

1. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013; 369:1691–1703.
2. Koutsounas I, Giaginis C, Patsouris E, Theocharis S. Current evidence for histone deacetylase inhibitors in pancreatic cancer. World J Gastroenterol. 2013; 19:813–828.
Article
3. Kneuertz PJ, Cunningham SC, Cameron JL, et al. Palliative surgical management of patients with unresectable pancreatic adenocarcinoma: trends and lessons learned from a large, single institution experience. J Gastrointest Surg. 2011; 15:1917–1927.
Article
4. Wigmore SJ, Plester CE, Richardson RA, Fearon KC. Changes in nutritional status associated with unresectable pancreatic cancer. Br J Cancer. 1997; 75:106–109.
Article
5. Gärtner S, Krüger J, Aghdassi AA, et al. Nutrition in pancreatic cancer: a review. Gastrointest Tumors. 2016; 2:195–202.
Article
6. Fearon KC, Baracos VE. Cachexia in pancreatic cancer: new treatment options and measures of success. HPB (Oxford). 2010; 12:323–324.
Article
7. Kyle UG, Pirlich M, Lochs H, Schuetz T, Pichard C. Increased length of hospital stay in underweight and overweight patients at hospital admission: a controlled population study. Clin Nutr. 2005; 24:133–142.
Article
8. Bachmann J, Heiligensetzer M, Krakowski-Roosen H, Büchler MW, Friess H, Martignoni ME. Cachexia worsens prognosis in patients with resectable pancreatic cancer. J Gastrointest Surg. 2008; 12:1193–1201.
Article
9. Evans WJ, Morley JE, Argilés J, et al. Cachexia: a new definition. Clin Nutr. 2008; 27:793–799.
Article
10. 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.
Article
11. Felix K, Fakelman F, Hartmann D, et al. Identification of serum proteins involved in pancreatic cancer cachexia. Life Sci. 2011; 88:218–225.
Article
12. Fearon K, Strasser F, Anker SD, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011; 12:489–495.
Article
13. Skipworth RJ, Stewart GD, Dejong CH, Preston T, Fearon KC. Pathophysiology of cancer cachexia: much more than host-tumour interaction? Clin Nutr. 2007; 26:667–676.
Article
14. Stewart GD, Skipworth RJ, Fearon KC. Cancer cachexia and fatigue. Clin Med (Lond). 2006; 6:140–143.
Article
15. Metter EJ, Conwit R, Tobin J, Fozard JL. Age-associated loss of power and strength in the upper extremities in women and men. J Gerontol A Biol Sci Med Sci. 1997; 52:B267–B276.
Article
16. Fearon KC, Glass DJ, Guttridge DC. Cancer cachexia: mediators, signaling, and metabolic pathways. Cell Metab. 2012; 16:153–166.
Article
17. Mace TA, Ameen Z, Collins A, et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 2013; 73:3007–3018.
Article
18. Argilés JM, Busquets S, Stemmler B, López-Soriano FJ. Cachexia and sarcopenia: mechanisms and potential targets for intervention. Curr Opin Pharmacol. 2015; 22:100–106.
Article
19. 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.
Article
20. Denley SM, Jamieson NB, McCall P, et al. Activation of the IL-6R/Jak/stat pathway is associated with a poor outcome in resected pancreatic ductal adenocarcinoma. J Gastrointest Surg. 2013; 17:887–898.
Article
21. Mitsunaga S, Ikeda M, Shimizu S, et al. Serum levels of IL-6 and IL-1β can predict the efficacy of gemcitabine in patients with advanced pancreatic cancer. Br J Cancer. 2013; 108:2063–2069.
Article
22. Pop VV, Seicean A, Lupan I, Samasca G, Burz CC. IL-6 roles - molecular pathway and clinical implication in pancreatic cancer - a systemic review. Immunol Lett. 2017; 181:45–50.
Article
23. Farren MR, Mace TA, Geyer S, et al. Systemic immune activity predicts overall survival in treatment-naïve patients with metastatic pancreatic cancer. Clin Cancer Res. 2016; 22:2565–2574.
Article
24. Zhou W, Jiang ZW, Tian J, Jiang J, Li N, Li JS. Role of NF-kappaB and cytokine in experimental cancer cachexia. World J Gastroenterol. 2003; 9:1567–1570.
25. Bonetto A, Aydogdu T, Jin X, et al. JAK/STAT3 pathway inhibition blocks skeletal muscle wasting downstream of IL-6 and in experimental cancer cachexia. Am J Physiol Endocrinol Metab. 2012; 303:E410–E421.
Article
26. Bonetto A, Aydogdu T, Kunzevitzky N, et al. STAT3 activation in skeletal muscle links muscle wasting and the acute phase response in cancer cachexia. PLoS One. 2011; 6:e22538.
Article
27. Guttridge DC, Mayo MW, Madrid LV, Wang CY, Baldwin AS Jr. NF-kappaB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia. Science. 2000; 289:2363–2366.
28. Zhang L, Tang H, Kou Y, et al. MG132-mediated inhibition of the ubiquitin-proteasome pathway ameliorates cancer cachexia. J Cancer Res Clin Oncol. 2013; 139:1105–1115.
Article
29. Melstrom LG, Melstrom KA Jr, Ding XZ, Adrian TE. Mechanisms of skeletal muscle degradation and its therapy in cancer cachexia. Histol Histopathol. 2007; 22:805–814.
30. Fogelman DR, Morris J, Xiao L, et al. A predictive model of inflammatory markers and patient-reported symptoms for cachexia in newly diagnosed pancreatic cancer patients. Support Care Cancer. 2017; 25:1809–1817.
Article
31. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997; 387:83–90.
32. Chen YG, Wang Q, Lin SL, Chang CD, Chuang J, Ying SY. Activin signaling and its role in regulation of cell proliferation, apoptosis, and carcinogenesis. Exp Biol Med (Maywood). 2006; 231:534–544.
Article
33. Sartori R, Milan G, Patron M, et al. Smad2 and 3 transcription factors control muscle mass in adulthood. Am J Physiol Cell Physiol. 2009; 296:C1248–C1257.
Article
34. Loumaye A, de Barsy M, Nachit M, et al. Role of activin A and myostatin in human cancer cachexia. J Clin Endocrinol Metab. 2015; 100:2030–2038.
Article
35. 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.
Article
36. Togashi Y, Kogita A, Sakamoto H, et al. Activin signal promotes cancer progression and is involved in cachexia in a subset of pancreatic cancer. Cancer Lett. 2015; 356(2 Pt B):819–827.
Article
37. Greco SH, Tomkötter L, Vahle AK, et al. TGF-β blockade reduces mortality and metabolic changes in a validated murine model of pancreatic cancer cachexia. PLoS One. 2015; 10:e0132786.
Article
38. Miyamoto Y, Hanna DL, Zhang W, Baba H, Lenz HJ. Molecular pathways: cachexia signaling-a targeted approach to cancer treatment. Clin Cancer Res. 2016; 22:3999–4004.
Article
39. Calnan DR, Brunet A. The FoxO code. Oncogene. 2008; 27:2276–2288.
Article
40. Schmitt TL, Martignoni ME, Bachmann J, et al. Activity of the Akt-dependent anabolic and catabolic pathways in muscle and liver samples in cancer-related cachexia. J Mol Med (Berl). 2007; 85:647–654.
Article
41. Avan A, Avan A, Le Large TY, et al. AKT1 and SELP polymorphisms predict the risk of developing cachexia in pancreatic cancer patients. PLoS One. 2014; 9:e108057.
Article
42. Sacheck JM, Ohtsuka A, McLary SC, Goldberg AL. IGF-I stimulates muscle growth by suppressing protein breakdown and expression of atrophy-related ubiquitin ligases, atrogin-1 and MuRF1. Am J Physiol Endocrinol Metab. 2004; 287:E591–E601.
Article
43. Abbott CR, Monteiro M, Small CJ, et al. The inhibitory effects of peripheral administration of peptide YY(3-36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res. 2005; 1044:127–131.
Article
44. Kim SY, Park SM, Lee ST. Apolipoprotein C-II is a novel substrate for matrix metalloproteinases. Biochem Biophys Res Commun. 2006; 339:47–54.
Article
45. Russell ST, Zimmerman TP, Domin BA, Tisdale MJ. Induction of lipolysis in vitro and loss of body fat in vivo by zinc-alpha2-glycoprotein. Biochim Biophys Acta. 2004; 1636:59–68.
46. Ronga I, Gallucci F, Riccardi F, Uomo G. Anorexia-cachexia syndrome in pancreatic cancer: recent advances and new pharmacological approach. Adv Med Sci. 2014; 59:1–6.
Article
47. Uomo G, Gallucci F, Rabitti PG. Anorexia-cachexia syndrome in pancreatic cancer: recent development in research and management. JOP. 2006; 7:157–162.
48. Ramos EJ, Suzuki S, Marks D, Inui A, Asakawa A, Meguid MM. Cancer anorexia-cachexia syndrome: cytokines and neuropeptides. Curr Opin Clin Nutr Metab Care. 2004; 7:427–434.
Article
49. Mueller TC, Burmeister MA, Bachmann J, Martignoni ME. Cachexia and pancreatic cancer: are there treatment options? World J Gastroenterol. 2014; 20:9361–9373.
50. Sikkens EC, Cahen DL, de Wit J, Looman CW, van Eijck C, Bruno MJ. Prospective assessment of the influence of pancreatic cancer resection on exocrine pancreatic function. Br J Surg. 2014; 101:109–113.
Article
51. Hackert T, Schütte K, Malfertheiner P. The pancreas: causes for malabsorption. Viszeralmedizin. 2014; 30:190–197.
Article
52. Kanda M, Fujii T, Kodera Y, Nagai S, Takeda S, Nakao A. Nutritional predictors of postoperative outcome in pancreatic cancer. Br J Surg. 2011; 98:268–274.
Article
53. Loh KW, Vriens MR, Gerritsen A, et al. Unintentional weight loss is the most important indicator of malnutrition among surgical cancer patients. Neth J Med. 2012; 70:365–369.
54. La Torre M, Ziparo V, Nigri G, Cavallini M, Balducci G, Ramacciato G. Malnutrition and pancreatic surgery: prevalence and outcomes. J Surg Oncol. 2013; 107:702–708.
Article
55. Billingsley KG, Hur K, Henderson WG, Daley J, Khuri SF, Bell RH Jr. Outcome after pancreaticoduodenectomy for periampullary cancer: an analysis from the veterans affairs national surgical quality improvement program. J Gastrointest Surg. 2003; 7:484–491.
Article
56. Gibbs J, Cull W, Henderson W, Daley J, Hur K, Khuri SF. Preoperative serum albumin level as a predictor of operative mortality and morbidity: results from the national VA surgical risk study. Arch Surg. 1999; 134:36–42.
57. Augustin T, Burstein MD, Schneider EB, et al. Frailty predicts risk of life-threatening complications and mortality after pancreatic resections. Surgery. 2016; 160:987–996.
Article
58. Imaoka H, Mizuno N, Hara K, et al. Evaluation of modified glasgow prognostic score for pancreatic cancer: a retrospective cohort study. Pancreas. 2016; 45:211–217.
59. Lucas DJ, Schexneider KI, Weiss M, et al. Trends and risk factors for transfusion in hepatopancreatobiliary surgery. J Gastrointest Surg. 2014; 18:719–728.
Article
60. Sikkens EC, Cahen DL, de Wit J, Looman CW, van Eijck C, Bruno MJ. A prospective assessment of the natural course of the exocrine pancreatic function in patients with a pancreatic head tumor. J Clin Gastroenterol. 2014; 48:e43–e46.
Article
61. Lindkvist B, Phillips ME, Domínguez-Muñoz JE. Clinical, anthropometric and laboratory nutritional markers of pancreatic exocrine insufficiency: prevalence and diagnostic use. Pancreatology. 2015; 15:589–597.
Article
62. Roeyen G, Jansen M, Chapelle T, et al. Diabetes mellitus and pre-diabetes are frequently undiagnosed and underreported in patients referred for pancreatic surgery. A prospective observational study. Pancreatology. 2016; 16:671–676.
Article
63. Illés D, Terzin V, Holzinger G, et al. New-onset type 2 diabetes mellitus--a high-risk group suitable for the screening of pancreatic cancer? Pancreatology. 2016; 16:266–271.
64. Gupta S, Vittinghoff E, Bertenthal D, et al. New-onset diabetes and pancreatic cancer. Clin Gastroenterol Hepatol. 2006; 4:1366–1372.
Article
65. Pannala R, Leirness JB, Bamlet WR, Basu A, Petersen GM, Chari ST. Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology. 2008; 134:981–987.
Article
66. Giovannucci E, Michaud D. The role of obesity and related metabolic disturbances in cancers of the colon, prostate, and pancreas. Gastroenterology. 2007; 132:2208–2225.
Article
67. Hart PA, Baichoo E, Bi Y, Hinton A, Kudva YC, Chari ST. Pancreatic polypeptide response to a mixed meal is blunted in pancreatic head cancer associated with diabetes mellitus. Pancreatology. 2015; 15:162–166.
Article
68. Cooperman AM, Chivati J, Chamberlain RS. Nutritional and metabolic aspects of pancreatic cancer. Curr Opin Clin Nutr Metab Care. 2000; 3:17–21.
Article
69. Klek S, Sierzega M, Szybinski P, et al. The immunomodulating enteral nutrition in malnourished surgical patients - a prospective, randomized, double-blind clinical trial. Clin Nutr. 2011; 30:282–288.
Article
70. Gianotti L, Braga M, Gentilini O, Balzano G, Zerbi A, Di Carlo V. Artificial nutrition after pancreaticoduodenectomy. Pancreas. 2000; 21:344–351.
Article
71. Di Carlo V, Gianotti L, Balzano G, Zerbi A, Braga M. Complications of pancreatic surgery and the role of perioperative nutrition. Dig Surg. 1999; 16:320–326.
Article
72. Karagianni VT, Papalois AE, Triantafillidis JK. Nutritional status and nutritional support before and after pancreatectomy for pancreatic cancer and chronic pancreatitis. Indian J Surg Oncol. 2012; 3:348–359.
Article
73. Park JS, Chung HK, Hwang HK, Kim JK, Yoon DS. Postoperative nutritional effects of early enteral feeding compared with total parental nutrition in pancreaticoduodectomy patients: a prosepective, randomized study. J Korean Med Sci. 2012; 27:261–267.
Article
74. Liu C, Du Z, Lou C, et al. Enteral nutrition is superior to total parenteral nutrition for pancreatic cancer patients who underwent pancreaticoduodenectomy. Asia Pac J Clin Nutr. 2011; 20:154–160.
75. Park JW, Jang JY, Kim EJ, et al. Effects of pancreatectomy on nutritional state, pancreatic function and quality of life. Br J Surg. 2013; 100:1064–1070.
Article
76. Nussbaum DP, Zani S, Penne K, et al. Feeding jejunostomy tube placement in patients undergoing pancreaticoduodenectomy: an ongoing dilemma. J Gastrointest Surg. 2014; 18:1752–1759.
Article
77. Richter E, Denecke A, Klapdor S, Klapdor R. Parenteral nutrition support for patients with pancreatic cancer--improvement of the nutritional status and the therapeutic outcome. Anticancer Res. 2012; 32:2111–2118.
78. Pelzer U, Arnold D, Gövercin M, et al. Parenteral nutrition support for patients with pancreatic cancer. Results of a phase II study. BMC Cancer. 2010; 10:86.
Article
79. Vashi PG, Dahlk S, Popiel B, Lammersfeld CA, Ireton-Jones C, Gupta D. A longitudinal study investigating quality of life and nutritional outcomes in advanced cancer patients receiving home parenteral nutrition. BMC Cancer. 2014; 14:593.
Article
80. Davidson W, Ash S, Capra S, Bauer J. Cancer Cachexia Study Group. Weight stabilisation is associated with improved survival duration and quality of life in unresectable pancreatic cancer. Clin Nutr. 2004; 23:239–247.
Article
81. Wigmore SJ, Barber MD, Ross JA, Tisdale MJ, Fearon KC. Effect of oral eicosapentaenoic acid on weight loss in patients with pancreatic cancer. Nutr Cancer. 2000; 36:177–184.
Article
82. Fearon KC, Von Meyenfeldt MF, Moses AG, et al. Effect of a protein and energy dense N-3 fatty acid enriched oral supplement on loss of weight and lean tissue in cancer cachexia: a randomised double blind trial. Gut. 2003; 52:1479–1486.
Article
83. Heller AR, Rössel T, Gottschlich B, et al. Omega-3 fatty acids improve liver and pancreas function in postoperative cancer patients. Int J Cancer. 2004; 111:611–616.
Article
84. Arshad A, Isherwood J, Mann C, et al. Intravenous ω-3 fatty acids plus gemcitabine. JPEN J Parenter Enteral Nutr. 2017; 41:398–403.
Article
85. Kraft M, Kraft K, Gärtner S, et al. L-carnitine-supplementation in advanced pancreatic cancer (CARPAN)--a randomized multi-centre trial. Nutr J. 2012; 11:52.
Article
86. Corish CA, Kennedy NP. Protein-energy undernutrition in hospital in-patients. Br J Nutr. 2000; 83:575–591.
Article
87. Barker LA, Gout BS, Crowe TC. Hospital malnutrition: prevalence, identification and impact on patients and the healthcare system. Int J Environ Res Public Health. 2011; 8:514–527.
Article
Full Text Links
  • KJG
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr