Influence of immunological nutrition on treatment of patients with oncological profile

Authors

  • M. O. Katrichenko National Cancer Institute of Ministry of Health of Ukraine, Kyiv
  • I. I. Lisnyi National Cancer Institute of Ministry of Health of Ukraine, Kyiv

DOI:

https://doi.org/10.31636/prmd.v1i1.3

Keywords:

tumor, nutrition, nutrients, T-cells, NK-cells, treatment

Abstract

In the reviewed article, we consider epidemiological and laboratory data that confirm the protective effects of biologically active nutrients in our diet for various diseases. Along with various factors such as alcohol, smoking, nutrition plays a vital role in influencing the patient’s immune response by transforming cells or by preventing, or acceleration of malignancy. Many data suggest that immunoactive nutrients control inflammatory and precancerous reactions in immune cells. Immunoprophylaxis is usually associated with modulation of the immune response when inflamed, thereby improving clinical outcomes. Different nutrients, including glutamine, arginine, vitamins, minerals and long-chain fatty acids, are important components of immunological nutrition. Clinical studies associated with these substances show different results with minimal effect. However, some studies have shown that these nutrients may have immunomodulatory effects that can reduce the risk of developing cancer. Pre-clinical studies claim that most of these nutrients have a positive effect in the complex treatment of cancer patients. In this article, we will consider the effect of the above nutrients on the immune system in patients of oncologic profile.

Recent evidences suggest that immunological nutrition plays an important role in the development of cancer and its progression. Data from animal studies have clearly shown that the use of immunomodulatory nutrients isolated from food, by launching a cascade of immunological reactions, can detect and eliminate the tumor. Although the technology has evolved to such an extent that we can study each individual cytokine or function of the immune cell, it is difficult to demonstrate the powerful role of the immune system in preventing or treating cancer due to the complexity of the tumor cell or heterogeneity in different patients' populations. However, the study sheds light on interactions in immune responses and cancer development, prevention and therapeutic strategies that involve modulation through biologically active agents.

Downloads

Download data is not yet available.

References

Lisnyy II, Kolesnik EA, Mazanko YV, Katrichenko MO, Kuchin YI. The role of parenteral and enteral clinical nutrition in cancer patients. Clinical oncology [Internet]. 2015;1(17):16–23. Available from: https://www.clinicaloncology.com.ua/article/13185/rol-parenteralnogo-i-enteralnogo-klinicheskogo-pitaniya-u-bolnyx-onkologicheskogo-profilya#en

Percival SS. Nutrition and Immunity. Nutrition Today [Internet]. Ovid Technologies (Wolters Kluwer Health); 2011 Jan;46(1):12–7. Available from: https://doi.org/10.1097/nt.0b013e3182076fc8

Liu RH. Dietary Bioactive Compounds and Their Health Implications. Journal of Food Science [Internet]. Wiley; 2013 Jun;78(s1):A18–A25. Available from: https://doi.org/10.1111/1750-3841.12101

Manhart N, Stehle P. Nutritive amino acids-effective modulators of the immune response. Forum Nutr. 2003;56:151-4.

McMurray DN. Cell-mediated immunity in nutritional deficiency. Prog Food Nutr Sci. 1984;8(3-4):193-228.

Gajewski TF, Schreiber H, Fu Y-X. Innate and adaptive immune cells in the tumor microenvironment. Nature Immunology [Internet]. Springer Science and Business Media LLC; 2013 Sep 18;14(10):1014–22. Available from: https://doi.org/10.1038/ni.2703

Cheng PN-M, Lam T-L, Lam W-M, Tsui S-M, Cheng AW-M, Lo W-H, et al. Pegylated Recombinant Human Arginase (rhArg-peg5,000mw) Inhibits the In vitro and In vivo Proliferation of Human Hepatocellular Carcinoma through Arginine Depletion. Cancer Research [Internet]. American Association for Cancer Research (AACR); 2007 Jan 1;67(1):309–17. Available from: https://doi.org/10.1158/0008-5472.can-06-1945

Ensor CM, Holtsberg FW, Bomalaski JS, Clark MA. Pegylated arginine deiminase (ADI-SS PEG20,000 mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo. Cancer Res. 2002 Oct 1;62(19):5443-50.

Kim RH, Coates JM, Bowles TL, McNerney GP, Sutcliffe J, Jung JU, et al. Arginine Deiminase as a Novel Therapy for Prostate Cancer Induces Autophagy and Caspase-Independent Apoptosis. Cancer Research [Internet]. American Association for Cancer Research (AACR); 2009 Jan 15;69(2):700–8. Available from: https://doi.org/10.1158/0008-5472.can-08-3157

Yoon C-Y, Shim Y-J, Kim E-H, Lee J-H, Won N-H, Kim J-H, et al. Renal cell carcinoma does not express argininosuccinate synthetase and is highly sensitive to arginine deprivationviaarginine deiminase. International Journal of Cancer [Internet]. Wiley; 2006 Nov 9;120(4):897–905. Available from: https://doi.org/10.1002/ijc.22322

Dillon BJ, Prieto VG, Curley SA, Ensor CM, Holtsberg FW, Bomalaski JS, et al. Incidence and distribution of argininosuccinate synthetase deficiency in human cancers. Cancer [Internet]. Wiley; 2004;100(4):826–33. Available from: https://doi.org/10.1002/cncr.20057

Ochoa AC, Zea AH, Hernandez C, Rodriguez PC. Arginase, Prostaglandins, and Myeloid-Derived Suppressor Cells in Renal Cell Carcinoma. Clinical Cancer Research [Internet]. American Association for Cancer Research (AACR); 2007 Jan 15;13(2):721s–726s. Available from: https://doi.org/10.1158/1078-0432.ccr-06-2197

Obermajer N, Wong JL, Edwards RP, Chen K, Scott M, Khader S, et al. Induction and stability of human Th17 cells require endogenous NOS2 and cGMP-dependent NO signaling. The Journal of Experimental Medicine [Internet]. Rockefeller University Press; 2013 Jun 24;210(7):1433–45. Available from: https://doi.org/10.1084/jem.20121277

Janakiram NB, Rao CV. iNOS-selective inhibitors for cancer prevention: promise and progress. Future Medicinal Chemistry [Internet]. Future Science Ltd; 2012 Nov;4(17):2193–204. Available from: https://doi.org/10.4155/fmc.12.168

Lamas B, Vergnaud-Gauduchon J, Goncalves-Mendes N, Perche O, Rossary A, Vasson M-P, et al. Altered functions of natural killer cells in response to L-Arginine availability. Cellular Immunology [Internet]. Elsevier BV; 2012 Dec;280(2):182–90. Available from: https://doi.org/10.1016/j.cellimm.2012.11.018

Norian LA, Rodriguez PC, O’Mara LA, Zabaleta J, Ochoa AC, Cella M, et al. Tumor-Infiltrating Regulatory Dendritic Cells Inhibit CD8+ T Cell Function via L-Arginine Metabolism. Cancer Research [Internet]. American Association for Cancer Research (AACR); 2009 Mar 24;69(7):3086–94. Available from: https://doi.org/10.1158/0008-5472.can-08-2826

Oberlies J, Watzl C, Giese T, Luckner C, Kropf P, Müller I, et al. Regulation of NK Cell Function by Human Granulocyte Arginase. The Journal of Immunology [Internet]. The American Association of Immunologists; 2009 Apr 20;182(9):5259–67. Available from: https://doi.org/10.4049/jimmunol.0803523

Rodriguez PC, Quiceno DG, Ochoa AC. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood [Internet]. American Society of Hematology; 2007 Feb 15;109(4):1568–73. Available from: https://doi.org/10.1182/blood-2006-06-031856

Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, Perera RM, Ferrone CR, Mullarky E, Shyh-Chang N, Kang YA. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013 Apr;496(7443):101.

Wang J-B, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, et al. Targeting Mitochondrial Glutaminase Activity Inhibits Oncogenic Transformation. Cancer Cell [Internet]. Elsevier BV; 2010 Sep;18(3):207–19. Available from: https://doi.org/10.1016/j.ccr.2010.08.009

Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, et al. Bidirectional Transport of Amino Acids Regulates mTOR and Autophagy. Cell [Internet]. Elsevier BV; 2009 Feb;136(3):521–34. Available from: https://doi.org/10.1016/j.cell.2008.11.044

Klimberg VS. Is Glutamine Effective in Enhancing Host Immune Response to Tumors? The Journal of Nutrition [Internet]. Oxford University Press (OUP); 2005 Dec 1;135(12):2920S–2920S. Available from: https://doi.org/10.1093/jn/135.12.2920s

Viora M, Quaranta MG, Straface E, Vari’ R, Masella R, Malorni W. Redox imbalance and immune functions: opposite effects of oxidized low-density lipoproteins and N-acetylcysteine. Immunology [Internet]. Wiley; 2001 Dec;104(4):431–8. Available from: https://doi.org/10.1046/j.1365-2567.2001.01334.x

Klysz D, Tai X, Robert PA, Craveiro M, Cretenet G, Oburoglu L, et al. Glutamine-dependent α-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Science Signaling [Internet]. American Association for the Advancement of Science (AAAS); 2015 Sep 29;8(396):ra97–ra97. Available from: https://doi.org/10.1126/scisignal.aab2610

Quan Z-F. Effect of glutamine on change in early postoperative intestinal permeability and its relation to systemic inflammatory response. World Journal of Gastroenterology [Internet]. Baishideng Publishing Group Inc.; 2004;10(13):1992. Available from: https://doi.org/10.3748/wjg.v10.i13.1992

Krishna Rao R. Role of Glutamine in Protection of Intestinal Epithelial Tight Junctions. Journal of Epithelial Biology and Pharmacology [Internet]. Bentham Science Publishers Ltd.; 2012 Jan 16;5(1):47–54. Available from: https://doi.org/10.2174/1875044301205010047

Cetinbas F, Yelken B, Gulbas Z. Role of glutamine administration on cellular immunity after total parenteral nutrition enriched with glutamine in patients with systemic inflammatory response syndrome. Journal of Critical Care [Internet]. Elsevier BV; 2010 Dec;25(4):661.e1–661.e6. Available from: https://doi.org/10.1016/j.jcrc.2010.03.011

OʼRiordain MG, Fearon KCH, Ross JA, Rogers P, Falconer JS, Bartolo DCC, et al. Glutamine-Supplemented Total Parenteral Nutrition Enhances T-Lymphocyte Response in Surgical Patients Undergoing Colorectal Resection. Annals of Surgery [Internet]. Ovid Technologies (Wolters Kluwer Health); 1994 Aug;220(2):212–21. Available from: https://doi.org/10.1097/00000658-199408000-00014

Hopkins FG, Cole SW. A contribution to the chemistry of proteids. The Journal of Physiology [Internet]. Wiley; 1901 Dec 23;27(4-5):418–28. Available from: https://doi.org/10.1113/jphysiol.1901.sp000880

Sainio E-L, Pulkki K, Young SN. L-Tryptophan: Biochemical, nutritional and pharmacological aspects. Amino Acids [Internet]. Springer Science and Business Media LLC; 1996;10(1):21–47. Available from: https://doi.org/10.1007/bf00806091

Fallarino F, Grohmann U, You S, McGrath BC, Cavener DR, Vacca C, et al. Tryptophan catabolism generates autoimmune-preventive regulatory T cells. Transplant Immunology [Internet]. Elsevier BV; 2006 Dec;17(1):58–60. Available from: https://doi.org/10.1016/j.trim.2006.09.017

Szczepanik M. Melatonin and its influence on immune system. J Physiol Pharmacol. 2007 Dec;58 Suppl 6:115-24.

Metz R, Smith C, DuHadaway JB, Chandler P, Baban B, Merlo LMF, et al. IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant function in inflammation. International Immunology [Internet]. Oxford University Press (OUP); 2014 Jan 13;26(7):357–67. Available from: https://doi.org/10.1093/intimm/dxt073

Manlapat AK, Kahler DJ, Chandler PR, Munn DH, Mellor AL. Cell-autonomous control of interferon type I expression by indoleamine 2,3-dioxygenase in regulatory CD19+ dendritic cells. European Journal of Immunology [Internet]. Wiley; 2007 Apr;37(4):1064–71. Available from: https://doi.org/10.1002/eji.200636690

Prendergast GC. Immune escape as a fundamental trait of cancer: focus on IDO. Oncogene [Internet]. Springer Nature; 2008 Mar 3;27(28):3889–900. Available from: https://doi.org/10.1038/onc.2008.35

Peng Y-P, Zhang J-J, Liang W, Tu M, Lu Z-P, Wei J-S, et al. Elevation of MMP-9 and IDO induced by pancreatic cancer cells mediates natural killer cell dysfunction. BMC Cancer [Internet]. Springer Nature; 2014 Oct 2;14(1). Available from: https://doi.org/10.1186/1471-2407-14-738

Ferdinande L, Decaestecker C, Verset L, Mathieu A, Moles Lopez X, Negulescu A-M, et al. Clinicopathological significance of indoleamine 2,3-dioxygenase 1 expression in colorectal cancer. British Journal of Cancer [Internet]. Springer Nature; 2011 Nov 22;106(1):141–7. Available from: https://doi.org/10.1038/bjc.2011.513

Brandacher G. Prognostic Value of Indoleamine 2,3-Dioxygenase Expression in Colorectal Cancer: Effect on Tumor-Infiltrating T Cells. Clinical Cancer Research [Internet]. American Association for Cancer Research (AACR); 2006 Feb 15;12(4):1144–51. Available from: https://doi.org/10.1158/1078-0432.ccr-05-1966

Calviello G, Di Nicuolo F, Gragnoli S, Piccioni E, Serini S, Maggiano N, et al. n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE 2 induced ERK-1 and -2 and HIF-1α induction pathway. Carcinogenesis [Internet]. Oxford University Press (OUP); 2004 Dec;25(12):2303–10. Available from: https://doi.org/10.1093/carcin/bgh265

Clarke RG, Lund EK, Latham P, Pinder AC, Johnson IT. Effect of eicosapentaenoic acid on the proliferation and incidence of apoptosis in the colorectal cell line HT29. Lipids [Internet]. Wiley; 1999 Dec;34(12):1287–95. Available from: https://doi.org/10.1007/s11745-999-0480-7

Courtney ED, Matthews S, Finlayson C, Di Pierro D, Belluzzi A, Roda E, et al. Eicosapentaenoic acid (EPA) reduces crypt cell proliferation and increases apoptosis in normal colonic mucosa in subjects with a history of colorectal adenomas. International Journal of Colorectal Disease [Internet]. Springer Science and Business Media LLC; 2007 Jan 10;22(7):765–76. Available from: https://doi.org/10.1007/s00384-006-0240-4

Latham P, Lund EK, Johnson IT. Dietary n-3 PUFA increases the apoptotic response to 1,2-dimethylhydrazine, reduces mitosis and suppresses the induction of carcinogenesis in the rat colon. Carcinogenesis [Internet]. Oxford University Press (OUP); 1999 Apr;20(4):645–50. Available from: https://doi.org/10.1093/carcin/20.4.645

Kantor ED, Lampe JW, Peters U, Vaughan TL, White E. Long-Chain Omega-3 Polyunsaturated Fatty Acid Intake and Risk of Colorectal Cancer. Nutrition and Cancer [Internet]. Informa UK Limited; 2013 Sep 20;66(4):716–27. Available from: https://doi.org/10.1080/01635581.2013.804101

Calder PC. n−3 Polyunsaturated fatty acids and inflammation: From molecular biology to the clinic. Lipids [Internet]. Wiley; 2003 Apr;38(4):343–52. Available from: https://doi.org/10.1007/s11745-003-1068-y

Influence of immunological nutrition on treatment of patients with oncological profile

Published

2018-05-16

How to Cite

1.
Influence of immunological nutrition on treatment of patients with oncological profile. prmd [Internet]. 2018 May 16 [cited 2024 Mar. 28];1(1):19-24. Available from: https://perioperative.org.ua/index.php/prtmdc/article/view/5