J Gastric Cancer.  2019 Sep;19(3):254-277. 10.5230/jgc.2019.19.e29.

Biomarkers for Evaluating the Inflammation Status in Patients with Cancer

Affiliations
  • 1Department of Surgery, Yonsei University College of Medicine, Seoul, Korea. cairus@yuhs.ac
  • 2Department of General Surgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
  • 3Department of Biostatistics and Medical Informatics, Institute of Medical Science, Karadeniz Technical University, Trabzon, Turkey.
  • 4Open NBI Convergence Technology Research Laboratory, Severance Hospital, Yonsei University Health System, Seoul, Korea.
  • 5Gastric Cancer Center, Yonsei Cancer Hospital; Seoul, Korea.

Abstract

Inflammation can be a causative factor for carcinogenesis or can result from a consequence of cancer progression. Moreover, cancer therapeutic interventions can also induce an inflammatory response. Various inflammatory parameters are used to assess the inflammatory status during cancer treatment. It is important to select the most optimal biomarker among these parameters. Additionally, suitable biomarkers must be examined if there are no known parameters. We briefly reviewed the published literature for the use of inflammatory parameters in the treatment of patients with cancer. Most studies on inflammation evaluated the correlation between host characteristics, effect of interventions, and clinical outcomes. Additionally, the levels of C-reactive protein, albumin, lymphocytes, and platelets were the most commonly used laboratory parameters, either independently or in combination with other laboratory parameters and clinical characteristics. Furthermore, the immune parameters are classically examined using flow cytometry, immunohistochemical staining, and enzyme-linked immunosorbent assay techniques. However, gene expression profiling can aid in assessing the overall peri-interventional immune status. The checklists of guidelines, such as STAndards for Reporting of Diagnostic accuracy and REporting recommendations for tumor MARKer prognostic studies should be considered when designing studies to investigate the inflammatory parameters. Finally, the data should be interpreted after adjusting for clinically important variables, such as age and cancer stage.

Keyword

Inflammation; Cancer; Biomarkers; Immune system; Outcome assessment

MeSH Terms

Biomarkers*
C-Reactive Protein
Carcinogenesis
Checklist
Enzyme-Linked Immunosorbent Assay
Flow Cytometry
Gene Expression Profiling
Humans
Immune System
Inflammation*
Lymphocytes
Biomarkers
C-Reactive Protein

Figure

  • Fig. 1 Relationship among parameters. Parameters with continuous values (Green boxes) and with categorical values (Orange boxes). Detail of values are presented in Appendix 1. BMI = body mass index; MUST = malnutrition universal screening tool; NRS = nutritional risk screening; NRI = nutritional risk index; AST = aspartate transaminase; CRP = C-reactive protein; GPS = Glasgow prognostic score; CAR = C-reactive protein-to-albumin ratio; CONUT = controlling nutritional status; PI = prognostic index; PNI = prognostic nutritional index; WBC = white blood cell; APRI = aspartate aminotransferase/platelet count ratio index; NLR = neutrophil-to-lymphocyte ratio; NMLR = neutrophil-monocyte-to-lymphocyte ratio; LMR = lymphocyte-to-monocyte ratio; SII = systemic immune-inflammation index; GLR = granulocyte-to-lymphocyte ratio; PLR = platelet-to-lymphocyte ratio; COP-NLR = combination of platelet count and neutrophil-to-lymphocyte ratio.

  • Fig. 2 Relationship of albumin level with age according to stage. Each point represents age and albumin level of a patient. Linear regression of each stage group shows that mean albumin level at same age decrease as the disease progresses. Dataset from Lee et al. [115].

  • Fig. 3 Associations of interventions with response patterns INF = interferon; IL = interleukin; TNF = tumor necrosis factor; NK = natural killer; HLA-DR = human leukocyte antigen-DR isotype; ERAS = enhanced recovery after surgery; Th = T helper; DC = dendritic cell; CIK = cytokine-induced killer; GM-CSF = granulocyte-macrophage colony-stimulating factor.

  • Fig. 4 Unsupervised clustering of inferred immune fraction using leukocyte signature matrix in the cancer genome atlas stomach adenocarcinoma (TCGA-STAD) transcriptomic dataset (n=450). Integrative transcriptomic analysis enables the identification of distinct immune landscapes associated clinical phenotypes using the fraction of infiltrated immune subsets in each sample as well as the evaluation of oncogenic pathway specific to each sample.


Reference

1. Hensler T, Hecker H, Heeg K, Heidecke CD, Bartels H, Barthlen W, et al. Distinct mechanisms of immunosuppression as a consequence of major surgery. Infect Immun. 1997; 65:2283–2291.
Article
2. Zeltsman M, Mayor M, Jones DR, Adusumilli PS. Surgical immune interventions for solid malignancies. Am J Surg. 2016; 212:682–690.e5.
Article
3. Guner A, Kim SY, Yu JE, Min IK, Roh YH, Roh C, et al. Parameters for predicting surgical outcomes for gastric cancer patients: simple is better than complex. Ann Surg Oncol. 2018; 25:3239–3247.
Article
4. Pennell LM, Galligan CL, Fish EN. Sex affects immunity. J Autoimmun. 2012; 38:J282–J291.
Article
5. Roved J, Westerdahl H, Hasselquist D. Sex differences in immune responses: Hormonal effects, antagonistic selection, and evolutionary consequences. Horm Behav. 2017; 88:95–105.
Article
6. Ono S, Tsujimoto H, Hiraki S, Takahata R, Kinoshita M, Mochizuki H. Sex differences in cytokine production and surface antigen expression of peripheral blood mononuclear cells after surgery. Am J Surg. 2005; 190:439–444.
Article
7. Kosuga T, Ichikawa D, Okamoto K, Komatsu S, Konishi H, Takeshita H, et al. Impact of age on early surgical outcomes of laparoscopy-assisted gastrectomy with suprapancreatic nodal dissection for clinical stage I gastric cancer. Anticancer Res. 2015; 35:2191–2198.
8. Kudoh A, Katagai H, Takazawa T, Matsuki A. Plasma proinflammatory cytokine response to surgical stress in elderly patients. Cytokine. 2001; 15:270–273.
Article
9. Milner JJ, Beck MA. The impact of obesity on the immune response to infection. Proc Nutr Soc. 2012; 71:298–306.
Article
10. Kolb R, Sutterwala FS, Zhang W. Obesity and cancer: inflammation bridges the two. Curr Opin Pharmacol. 2016; 29:77–89.
Article
11. Gunderson CC, Java J, Moore KN, Walker JL. The impact of obesity on surgical staging, complications, and survival with uterine cancer: a Gynecologic Oncology Group LAP2 ancillary data study. Gynecol Oncol. 2014; 133:23–27.
12. Khoury W, Lavery IC, Kiran RP. Effects of chronic immunosuppression on long-term oncologic outcomes for colorectal cancer patients undergoing surgery. Ann Surg. 2011; 253:323–327.
Article
13. Moulin CM, Marguti I, Peron JP, Halpern A, Rizzo LV. Bariatric surgery reverses natural killer (NK) cell activity and NK-related cytokine synthesis impairment induced by morbid obesity. Obes Surg. 2011; 21:112–118.
Article
14. Kotani N, Hashimoto H, Sessler DI, Yoshida H, Kimura N, Okawa H, et al. Smoking decreases alveolar macrophage function during anesthesia and surgery. Anesthesiology. 2000; 92:1268–1277.
Article
15. Na YM, Kim MY, Kim YK, Ha YR, Yoon DS. Exercise therapy effect on natural killer cell cytotoxic activity in stomach cancer patients after curative surgery. Arch Phys Med Rehabil. 2000; 81:777–779.
Article
16. Ukkonen M, Karlsson S, Laukkarinen J, Rantanen T, Paajanen H. Finnsepsis Study Group. Severe sepsis in elderly patients undergoing gastrointestinal surgery-a prospective multicenter follow-up study of Finnish Intensive Care Units. J Gastrointest Surg. 2016; 20:1028–1033.
Article
17. Solaini L, Atmaja BT, Arumugam P, Hutchins RR, Abraham AT, Bhattacharya S, et al. The role of perioperative inflammatory-based prognostic systems in patients with colorectal liver metastases undergoing surgery. A cohort study. Int J Surg. 2016; 36:8–12.
Article
18. Kudoh A, Katagai H, Takazawa T. Plasma inflammatory cytokine response to surgical trauma in chronic depressed patients. Cytokine. 2001; 13:104–108.
Article
19. Sutter PM, Spagnoli GC, Marx A, Gürke L, Troeger H, Fricker R, et al. Increased surface expression of CD18 and CD11b in leukocytes after tourniquet ischemia during elective hand surgery. World J Surg. 1997; 21:179–184.
Article
20. Weighardt H, Heidecke CD, Emmanuilidis K, Maier S, Bartels H, Siewert JR, et al. Sepsis after major visceral surgery is associated with sustained and interferon-gamma-resistant defects of monocyte cytokine production. Surgery. 2000; 127:309–315.
21. Sammour T, Kahokehr A, Chan S, Booth RJ, Hill AG. The humoral response after laparoscopic versus open colorectal surgery: a meta-analysis. J Surg Res. 2010; 164:28–37.
Article
22. Evans C, Galustian C, Kumar D, Hagger R, Melville DM, Bodman-Smith M, et al. Impact of surgery on immunologic function: comparison between minimally invasive techniques and conventional laparotomy for surgical resection of colorectal tumors. Am J Surg. 2009; 197:238–245.
Article
23. Zawadzki M, Krzystek-Korpacka M, Gamian A, Witkiewicz W. Comparison of inflammatory responses following robotic and open colorectal surgery: a prospective study. Int J Colorectal Dis. 2017; 32:399–407.
Article
24. Pilka R, Marek R, Adam T, Kudela M, Ondrová D, Neubert D, et al. Systemic inflammatory response after open, laparoscopic and robotic surgery in endometrial cancer patients. Anticancer Res. 2016; 36:2909–2922.
25. Schneider C, von Aulock S, Zedler S, Schinkel C, Hartung T, Faist E. Perioperative recombinant human granulocyte colony-stimulating factor (Filgrastim) treatment prevents immunoinflammatory dysfunction associated with major surgery. Ann Surg. 2004; 239:75–81.
Article
26. Crozier JE, Leitch EF, McKee RF, Anderson JH, Horgan PG, McMillan DC. Relationship between emergency presentation, systemic inflammatory response, and cancer-specific survival in patients undergoing potentially curative surgery for colon cancer. Am J Surg. 2009; 197:544–549.
Article
27. Kimura A, Ono S, Hiraki S, Takahata R, Tsujimoto H, Miyazaki H, et al. The postoperative serum interleukin-15 concentration correlates with organ dysfunction and the prognosis of septic patients following emergency gastrointestinal surgery. J Surg Res. 2012; 175:e83–e88.
Article
28. Aosasa S, Ono S, Mochizuki H, Tsujimoto H, Osada S, Takayama E, et al. Activation of monocytes and endothelial cells depends on the severity of surgical stress. World J Surg. 2000; 24:10–16.
Article
29. Flohé S, Lendemans S, Schade FU, Kreuzfelder E, Waydhas C. Influence of surgical intervention in the immune response of severely injured patients. Intensive Care Med. 2004; 30:96–102.
Article
30. Gu CY, Zhang J, Qian YN, Tang QF. Effects of epidural anesthesia and postoperative epidural analgesia on immune function in esophageal carcinoma patients undergoing thoracic surgery. Mol Clin Oncol. 2015; 3:190–196.
Article
31. Chen WK, Ren L, Wei Y, Zhu DX, Miao CH, Xu JM. General anesthesia combined with epidural anesthesia ameliorates the effect of fast-track surgery by mitigating immunosuppression and facilitating intestinal functional recovery in colon cancer patients. Int J Colorectal Dis. 2015; 30:475–481.
Article
32. Sacerdote P, Bianchi M, Gaspani L, Manfredi B, Maucione A, Terno G, et al. The effects of tramadol and morphine on immune responses and pain after surgery in cancer patients. Anesth Analg. 2000; 90:1411–1414.
Article
33. Martínez AB, Longás J, Ramírez JM. A model for lymphocyte activation in open versus laparoscopic surgery in colorectal cancer patients in enhanced recovery after surgery (ERAS) protocols. Int J Colorectal Dis. 2017; 32:913–916.
Article
34. Veenhof AA, Vlug MS, van der Pas MH, Sietses C, van der Peet DL, de Lange-de Klerk ES, et al. Surgical stress response and postoperative immune function after laparoscopy or open surgery with fast track or standard perioperative care: a randomized trial. Ann Surg. 2012; 255:216–221.
35. Yang D, He W, Zhang S, Chen H, Zhang C, He Y. Fast-track surgery improves postoperative clinical recovery and immunity after elective surgery for colorectal carcinoma: randomized controlled clinical trial. World J Surg. 2012; 36:1874–1880.
Article
36. Heiss MM, Fasol-Merten K, Allgayer H, Ströhlein MA, Tarabichi A, Wallner S, et al. Influence of autologous blood transfusion on natural killer and lymphokine-activated killer cell activities in cancer surgery. Vox Sang. 1997; 73:237–245.
Article
37. Theodoraki K, Markatou M, Rizos D, Fassoulaki A. The impact of two different transfusion strategies on patient immune response during major abdominal surgery: a preliminary report. J Immunol Res. 2014; 2014:945829.
Article
38. Fukuda T, Seto Y, Yamada K, Hiki N, Fukunaga T, Oyama S, et al. Can immune-enhancing nutrients reduce postoperative complications in patients undergoing esophageal surgery? Dis Esophagus. 2008; 21:708–711.
Article
39. Giger U, Büchler M, Farhadi J, Berger D, Hüsler J, Schneider H, et al. Preoperative immunonutrition suppresses perioperative inflammatory response in patients with major abdominal surgery-a randomized controlled pilot study. Ann Surg Oncol. 2007; 14:2798–2806.
Article
40. Arumugam S, Lau CS, Chamberlain RS. Probiotics and Synbiotics Decrease Postoperative Sepsis in Elective Gastrointestinal Surgical Patients: a Meta-Analysis. J Gastrointest Surg. 2016; 20:1123–1131.
Article
41. Liu Z, Li C, Huang M, Tong C, Zhang X, Wang L, et al. Positive regulatory effects of perioperative probiotic treatment on postoperative liver complications after colorectal liver metastases surgery: a double-center and double-blind randomized clinical trial. BMC Gastroenterol. 2015; 15:34.
Article
42. Braga M, Gianotti L, Vignali A, Di Carlo V. Immunonutrition in gastric cancer surgical patients. Nutrition. 1998; 14:831–835.
Article
43. Furukawa K, Tashiro T, Yamamori H, Takagi K, Morishima Y, Sugiura T, et al. Effects of soybean oil emulsion and eicosapentaenoic acid on stress response and immune function after a severely stressful operation. Ann Surg. 1999; 229:255–261.
Article
44. Wang WP, Yan XL, Ni YF, Guo K, Ke CK, Cheng QS, et al. Effects of lipid emulsions in parenteral nutrition of esophageal cancer surgical patients receiving enteral nutrition: a comparative analysis. Nutrients. 2013; 6:111–123.
45. Schmidt SC, Hamann S, Langrehr JM, Höflich C, Mittler J, Jacob D, et al. Preoperative high-dose steroid administration attenuates the surgical stress response following liver resection: results of a prospective randomized study. J Hepatobiliary Pancreat Surg. 2007; 14:484–492.
Article
46. Pratschke S, von Dossow-Hanfstingl V, Dietz J, Schneider CP, Tufman A, Albertsmeier M, et al. Dehydroepiandrosterone modulates T-cell response after major abdominal surgery. J Surg Res. 2014; 189:117–125.
Article
47. Li H, Wang C, Yu J, Cao S, Wei F, Zhang W, et al. Dendritic cell-activated cytokine-induced killer cells enhance the anti-tumor effect of chemotherapy on non-small cell lung cancer in patients after surgery. Cytotherapy. 2009; 11:1076–1083.
Article
48. Brivio F, Lissoni P, Gilardi R, Ferrante R, Vigore L, Curzi L, et al. Abrogation of surgery-induced decline in circulating dendritic cells by subcutaneous preoperative administration of IL-2 in operable cancer patients. J Biol Regul Homeost Agents. 2000; 14:200–203.
49. Nishiyama J, Matsuda M, Ando S, Hirasawa M, Suzuki T, Makuuchi H. The effects of the early administration of sivelestat sodium, a selective neutrophil elastase inhibitor, on the postoperative course after radical surgery for esophageal cancer. Surg Today. 2012; 42:659–665.
Article
50. Ono S, Aosasa S, Mochizuki H. Effects of a protease inhibitor on reduction of surgical stress in esophagectomy. Am J Surg. 1999; 177:78–82.
Article
51. Kerr SF, Klonizakis M, Glynne-Jones R. Suppression of the postoperative neutrophil leucocytosis following neoadjuvant chemoradiotherapy for rectal cancer and implications for surgical morbidity. Colorectal Dis. 2010; 12:549–554.
Article
52. Tanis E, Julié C, Emile JF, Mauer M, Nordlinger B, Aust D, et al. Prognostic impact of immune response in resectable colorectal liver metastases treated by surgery alone or surgery with perioperative FOLFOX in the randomised EORTC study 40983. Eur J Cancer. 2015; 51:2708–2717.
Article
53. Westerterp M, Boermeester MA, Omloo JM, Hulshof MC, Vervenne WL, Lutter R, et al. Differential responses of cellular immunity in patients undergoing neoadjuvant therapy followed by surgery for carcinoma of the oesophagus. Cancer Immunol Immunother. 2008; 57:1837–1847.
Article
54. Wang D, Yang XL, Chai XQ, Shu SH, Zhang XL, Xie YH, et al. A short-term increase of the postoperative naturally circulating dendritic cells subsets in flurbiprofen-treated patients with esophageal carcinoma undergoing thoracic surgery. Oncotarget. 2016; 7:18705–18712.
Article
55. Liakopoulos OJ, Dörge H, Schmitto JD, Nagorsnik U, Grabedünkel J, Schoendube FA. Effects of preoperative statin therapy on cytokines after cardiac surgery. Thorac Cardiovasc Surg. 2006; 54:250–254.
Article
56. Amar D, Zhang H, Park B, Heerdt PM, Fleisher M, Thaler HT. Inflammation and outcome after general thoracic surgery. Eur J Cardiothorac Surg. 2007; 32:431–434.
Article
57. De Lorenzo A, Pittella F, Rocha A. Increased preoperative C-reactive protein levels are associated with inhospital death after coronary artery bypass surgery. Inflammation. 2012; 35:1179–1183.
Article
58. Ishizuka M, Kubota K, Kita J, Shimoda M, Kato M, Sawada T. Usefulness of a modified inflammation-based prognostic system for predicting postoperative mortality of patients undergoing surgery for primary hepatocellular carcinoma. J Surg Oncol. 2011; 103:801–806.
Article
59. Josse JM, Cleghorn MC, Ramji KM, Jiang H, Elnahas A, Jackson TD, et al. The neutrophil-to-lymphocyte ratio predicts major perioperative complications in patients undergoing colorectal surgery. Colorectal Dis. 2016; 18:O236–O242.
Article
60. Mokart D, Capo C, Blache JL, Delpero JR, Houvenaeghel G, Martin C, et al. Early postoperative compensatory anti-inflammatory response syndrome is associated with septic complications after major surgical trauma in patients with cancer. Br J Surg. 2002; 89:1450–1456.
Article
61. Rettig TC, Verwijmeren L, Dijkstra IM, Boerma D, van de Garde EM, Noordzij PG. Postoperative interleukin-6 level and early detection of complications after elective major abdominal surgery. Ann Surg. 2016; 263:1207–1212.
Article
62. Saeed K, Dale AP, Leung E, Cusack T, Mohamed F, Lockyer G, et al. Procalcitonin levels predict infectious complications and response to treatment in patients undergoing cytoreductive surgery for peritoneal malignancy. Eur J Surg Oncol. 2016; 42:234–243.
Article
63. Weighardt H, Heidecke CD, Westerholt A, Emmanuilidis K, Maier S, Veit M, et al. Impaired monocyte IL-12 production before surgery as a predictive factor for the lethal outcome of postoperative sepsis. Ann Surg. 2002; 235:560–567.
Article
64. Anitei MG, Zeitoun G, Mlecnik B, Marliot F, Haicheur N, Todosi AM, et al. Prognostic and predictive values of the immunoscore in patients with rectal cancer. Clin Cancer Res. 2014; 20:1891–1899.
Article
65. Matsutani S, Shibutani M, Maeda K, Nagahara H, Fukuoka T, Nakao S, et al. Significance of tumor-infiltrating lymphocytes before and after neoadjuvant therapy for rectal cancer. Cancer Sci. 2018; 109:966–979.
Article
66. Ishizuka M, Nagata H, Takagi K, Iwasaki Y, Kubota K. Systemic inflammatory response predicts perioperative central venous catheter-related bloodstream infection in patients Undergoing colorectal cancer surgery with administration of parenteral nutrition. Anticancer Res. 2012; 32:4045–4050.
67. Strohmeyer JC, Blume C, Meisel C, Doecke WD, Hummel M, Hoeflich C, et al. Standardized immune monitoring for the prediction of infections after cardiopulmonary bypass surgery in risk patients. Cytometry B Clin Cytom. 2003; 53:54–62.
Article
68. Song B, Zhen S, Meng F. T cell inflammation profile after surgical resection may predict tumor recurrence in HBV-related hepatocellular carcinoma. Int Immunopharmacol. 2016; 41:35–41.
Article
69. Chan AW, Chan SL, Wong GL, Wong VW, Chong CC, Lai PB, et al. Prognostic nutritional index (PNI) predicts tumor recurrence of very early/early stage hepatocellular carcinoma after surgical resection. Ann Surg Oncol. 2015; 22:4138–4148.
Article
70. Lindenmann J, Fink-Neuboeck N, Avian A, Pichler M, Habitzruther M, Maier A, et al. Preoperative Glasgow prognostic score as additional independent prognostic parameter for patients with esophageal cancer after curative esophagectomy. Eur J Surg Oncol. 2017; 43:445–453.
Article
71. Toiyama Y, Inoue Y, Saigusa S, Kawamura M, Kawamoto A, Okugawa Y, et al. C-reactive protein as predictor of recurrence in patients with rectal cancer undergoing chemoradiotherapy followed by surgery. Anticancer Res. 2013; 33:5065–5074.
72. Wei IH, Harmon CM, Arcerito M, Cheng DF, Minter RM, Simeone DM. Tumor-associated macrophages are a useful biomarker to predict recurrence after surgical resection of nonfunctional pancreatic neuroendocrine tumors. Ann Surg. 2014; 260:1088–1094.
Article
73. Galizia G, Lieto E, Auricchio A, Cardella F, Mabilia A, Podzemny V, et al. Naples prognostic score, based on nutritional and inflammatory status, is an independent predictor of long-term outcome in patients undergoing surgery for colorectal cancer. Dis Colon Rectum. 2017; 60:1273–1284.
Article
74. He YF, Luo HQ, Wang W, Chen J, Yao YW, Yan Y, et al. Preoperative NLR and PLR in the middle or lower ESCC patients with radical operation. Eur J Cancer Care (Engl). 2017; 26:e12445.
Article
75. Ishizuka M, Nagata H, Takagi K, Iwasaki Y, Shibuya N, Kubota K. Clinical significance of the C-reactive protein to albumin ratio for survival after surgery for colorectal cancer. Ann Surg Oncol. 2016; 23:900–907.
Article
76. Ishizuka M, Oyama Y, Abe A, Kubota K. Combination of platelet count and neutrophil to lymphocyte ratio is a useful predictor of postoperative survival in patients undergoing surgery for gastric cancer. J Surg Oncol. 2014; 110:935–941.
Article
77. Malietzis G, Giacometti M, Askari A, Nachiappan S, Kennedy RH, Faiz OD, et al. A preoperative neutrophil to lymphocyte ratio of 3 predicts disease-free survival after curative elective colorectal cancer surgery. Ann Surg. 2014; 260:287–292.
Article
78. Mimatsu K, Fukino N, Ogasawara Y, Saino Y, Oida T. Utility of inflammatory marker- and nutritional status-based prognostic factors for predicting the prognosis of stage IV gastric cancer patients undergoing non-curative surgery. Anticancer Res. 2017; 37:4215–4222.
Article
79. Watt DG, Martin JC, Park JH, Horgan PG, McMillan DC. Neutrophil count is the most important prognostic component of the differential white cell count in patients undergoing elective surgery for colorectal cancer. Am J Surg. 2015; 210:24–30.
Article
80. Yamamoto T, Yanagimoto H, Satoi S, Toyokawa H, Yamao J, Kim S, et al. Circulating myeloid dendritic cells as prognostic factors in patients with pancreatic cancer who have undergone surgical resection. J Surg Res. 2012; 173:299–308.
Article
81. Zikos TA, Donnenberg AD, Landreneau RJ, Luketich JD, Donnenberg VS. Lung T-cell subset composition at the time of surgical resection is a prognostic indicator in non-small cell lung cancer. Cancer Immunol Immunother. 2011; 60:819–827.
Article
82. Jin Z, Li R, Liu J, Lin J. Long-term prognosis after cancer surgery with inhalational anesthesia and total intravenous anesthesia: a systematic review and meta-analysis. Int J Physiol Pathophysiol Pharmacol. 2019; 11:83–94.
83. Weng M, Chen W, Hou W, Li L, Ding M, Miao C. The effect of neuraxial anesthesia on cancer recurrence and survival after cancer surgery: an updated meta-analysis. Oncotarget. 2016; 7:15262–15273.
Article
84. Giger-Pabst U, Lange J, Maurer C, Bucher C, Schreiber V, Schlumpf R, et al. Short-term preoperative supplementation of an immunoenriched diet does not improve clinical outcome in well-nourished patients undergoing abdominal cancer surgery. Nutrition. 2013; 29:724–729.
Article
85. Klek S, Sierzega M, Szybinski P, Szczepanek K, Scislo L, Walewska E, et al. The immunomodulating enteral nutrition in malnourished surgical patients - a prospective, randomized, double-blind clinical trial. Clin Nutr. 2011; 30:282–288.
Article
86. Chen L, Sun L, Lang Y, Wu J, Yao L, Ning J, et al. Fast-track surgery improves postoperative clinical recovery and cellular and humoral immunity after esophagectomy for esophageal cancer. BMC Cancer. 2016; 16:449.
Article
87. Liu HT, Huang YC, Cheng SB, Huang YT, Lin PT. Effects of coenzyme Q10 supplementation on antioxidant capacity and inflammation in hepatocellular carcinoma patients after surgery: a randomized, placebo-controlled trial. Nutr J. 2016; 15:85.
Article
88. Parolari A, Camera M, Alamanni F, Naliato M, Polvani GL, Agrifoglio M, et al. Systemic inflammation after on-pump and off-pump coronary bypass surgery: a one-month follow-up. Ann Thorac Surg. 2007; 84:823–828.
Article
89. Papadima A, Boutsikou M, Lagoudianakis EE, Kataki A, Konstadoulakis M, Georgiou L, et al. Lymphocyte apoptosis after major abdominal surgery is not influenced by anesthetic technique: a comparative study of general anesthesia versus combined general and epidural analgesia. J Clin Anesth. 2009; 21:414–421.
Article
90. Reisinger KW, Derikx JP, van Vugt JL, Von Meyenfeldt MF, Hulsewé KW, Olde Damink SW, et al. Sarcopenia is associated with an increased inflammatory response to surgery in colorectal cancer. Clin Nutr. 2016; 35:924–927.
Article
91. Buttenschoen K, Schneider ME, Utz K, Kornmann M, Beger HG, Carli Buttenschoen D. Effect of major abdominal surgery on endotoxin release and expression of Toll-like receptors 2/4. Langenbecks Arch Surg. 2009; 394:293–302.
Article
92. Brix-Christensen V, Tønnesen E, Sørensen IJ, Bilfinger TV, Sanchez RG, Stefano GB. Effects of anaesthesia based on high versus low doses of opioids on the cytokine and acute-phase protein responses in patients undergoing cardiac surgery. Acta Anaesthesiol Scand. 1998; 42:63–70.
Article
93. Greco F, Hoda MR, Wagner S, Reichelt O, Inferrera A, Fischer K, et al. Adipocytokine: a new family of inflammatory and immunologic markers of invasiveness in major urologic surgery. Eur Urol. 2010; 58:781–787.
Article
94. Iwasaki A, Shirakusa T, Maekawa T, Enatsu S, Maekawa S. Clinical evaluation of systemic inflammatory response syndrome (SIRS) in advanced lung cancer (T3 and T4) with surgical resection. Eur J Cardiothorac Surg. 2005; 27:14–18.
Article
95. Bruce DM, Smith M, Walker CB, Heys SD, Binnie NR, Gough DB, et al. Minimal access surgery for cholelithiasis induces an attenuated acute phase response. Am J Surg. 1999; 178:232–234.
Article
96. Mokart D, Merlin M, Sannini A, Brun JP, Delpero JR, Houvenaeghel G, et al. Procalcitonin, interleukin 6 and systemic inflammatory response syndrome (SIRS): early markers of postoperative sepsis after major surgery. Br J Anaesth. 2005; 94:767–773.
Article
97. Karth GD, Buberl A, Nikfardjam M, Meyer B, Wollenek G, Grimm M, et al. Role of amiodarone on the systemic inflammatory response induced by cardiac surgery: proinflammatory actions. Can J Anaesth. 2007; 54:262–268.
Article
98. Ni XC, Yi Y, Fu YP, He HW, Cai XY, Wang JX, et al. Prognostic value of the modified Glasgow prognostic score in patients undergoing radical surgery for hepatocellular carcinoma. Medicine (Baltimore). 2015; 94:e1486.
Article
99. Ji F, Liang Y, Fu SJ, Guo ZY, Shu M, Shen SL, et al. A novel and accurate predictor of survival for patients with hepatocellular carcinoma after surgical resection: the neutrophil to lymphocyte ratio (NLR) combined with the aspartate aminotransferase/platelet count ratio index (APRI). BMC Cancer. 2016; 16:137.
Article
100. Dias Rodrigues V, Barroso de Pinho N, Abdelhay E, Viola JP, Correia MI, Brum Martucci R. Nutrition and immune-modulatory intervention in surgical patients with gastric cancer. Nutr Clin Pract. 2017; 32:122–129.
Article
101. Sun KY, Xu JB, Chen SL, Yuan YJ, Wu H, Peng JJ, et al. Novel immunological and nutritional-based prognostic index for gastric cancer. World J Gastroenterol. 2015; 21:5961–5971.
Article
102. Xie X, Luo KJ, Hu Y, Wang JY, Chen J. Prognostic value of preoperative platelet-lymphocyte and neutrophil-lymphocyte ratio in patients undergoing surgery for esophageal squamous cell cancer. Dis Esophagus. 2016; 29:79–85.
Article
103. Liao R, Jiang N, Tang ZW, Li W, Huang P, Luo SQ, et al. Systemic and intratumoral balances between monocytes/macrophages and lymphocytes predict prognosis in hepatocellular carcinoma patients after surgery. Oncotarget. 2016; 7:30951–30961.
Article
104. Hyun MH, Lee CH, Kwon YJ, Cho SI, Jang YJ, Kim DH, et al. Robot versus laparoscopic gastrectomy for cancer by an experienced surgeon: comparisons of surgery, complications, and surgical stress. Ann Surg Oncol. 2013; 20:1258–1265.
Article
105. Hsu JT, Wang CC, Le PH, Chen TH, Kuo CJ, Lin CJ, et al. Lymphocyte-to-monocyte ratios predict gastric cancer surgical outcomes. J Surg Res. 2016; 202:284–290.
Article
106. Pang S, Zhou Z, Yu X, Wei S, Chen Q, Nie S, et al. The predictive value of integrated inflammation scores in the survival of patients with resected hepatocellular carcinoma: a retrospective cohort study. Int J Surg. 2017; 42:170–177.
Article
107. Jain S, Gautam V, Naseem S. Acute-phase proteins: As diagnostic tool. J Pharm Bioallied Sci. 2011; 3:118–127.
Article
108. Warschkow R, Tarantino I, Ukegjini K, Beutner U, Müller SA, Schmied BM, et al. Diagnostic study and meta-analysis of C-reactive protein as a predictor of postoperative inflammatory complications after gastroesophageal cancer surgery. Langenbecks Arch Surg. 2012; 397:727–736.
Article
109. Warschkow R, Ukegjini K, Tarantino I, Steffen T, Müller SA, Schmied BM, et al. Diagnostic study and meta-analysis of C-reactive protein as a predictor of postoperative inflammatory complications after pancreatic surgery. J Hepatobiliary Pancreat Sci. 2012; 19:492–500.
Article
110. Kim S, McClave SA, Martindale RG, Miller KR, Hurt RT. Hypoalbuminemia and Clinical Outcomes: What is the Mechanism behind the Relationship? Am Surg. 2017; 83:1220–1227.
Article
111. Iwaku A, Kinoshita A, Onoda H, Fushiya N, Nishino H, Matsushima M, et al. The Glasgow prognostic score accurately predicts survival in patients with biliary tract cancer not indicated for surgical resection. Med Oncol. 2014; 31:787.
Article
112. Dupré A, Malik HZ. Inflammation and cancer: what a surgical oncologist should know. Eur J Surg Oncol. 2018; 44:566–570.
113. Roxburgh CS, Salmond JM, Horgan PG, Oien KA, McMillan DC. The relationship between the local and systemic inflammatory responses and survival in patients undergoing curative surgery for colon and rectal cancers. J Gastrointest Surg. 2009; 13:2011–2018.
Article
114. Guner A, Kim HI. ASO Author Reflections: Parameters for Predicting Surgical Outcomes for Gastric Cancer Patients: Simple Is Better Than Complex. Ann Surg Oncol. 2018; 25:699–700.
Article
115. Lee JY, Kim HI, Kim YN, Hong JH, Alshomimi S, An JY, et al. Clinical significance of the prognostic nutritional index for predicting short- and long-term surgical outcomes after gastrectomy: a retrospective analysis of 7781 gastric cancer patients. Medicine (Baltimore). 2016; 95:e3539.
116. Kohira S, Oka N, Inoue N, Itatani K, Hanayama N, Kitamura T, et al. Effect of the neutrophil elastase inhibitor sivelestat on perioperative inflammatory response after pediatric heart surgery with cardiopulmonary bypass: a prospective randomized study. Artif Organs. 2013; 37:1027–1033.
Article
117. Ng CS, Lee TW, Wan S, Wan IY, Sihoe AD, Arifi AA, et al. Thoracotomy is associated with significantly more profound suppression in lymphocytes and natural killer cells than video-assisted thoracic surgery following major lung resections for cancer. J Invest Surg. 2005; 18:81–88.
118. Volk T, Schenk M, Voigt K, Tohtz S, Putzier M, Kox WJ. Postoperative epidural anesthesia preserves lymphocyte, but not monocyte, immune function after major spine surgery. Anesth Analg. 2004; 98:1086–1092.
Article
119. Ammendola M, Sacco R, Zuccalà V, Luposella M, Patruno R, Gadaleta P, et al. Mast cells density positive to tryptase correlate with microvascular density in both primary gastric cancer tissue and loco-regional lymph node metastases from patients that have undergone radical surgery. Int J Mol Sci. 2016; 17:1905.
Article
120. Decker D, Tolba R, Springer W, Lauschke H, Hirner A, von Ruecker A. Abdominal surgical interventions: local and systemic consequences for the immune system--a prospective study on elective gastrointestinal surgery. J Surg Res. 2005; 126:12–18.
121. Leal-Noval SR, Muñoz-Gómez M, Arellano V, Adsuar A, Jiménez-Sánchez M, Corcia Y, et al. Influence of red blood cell transfusion on CD4+ T-helper cells immune response in patients undergoing cardiac surgery. J Surg Res. 2010; 164:43–49.
122. Matsuda A, Furukawa K, Takasaki H, Suzuki H, Kan H, Tsuruta H, et al. Preoperative oral immune-enhancing nutritional supplementation corrects Th1/Th2 imbalance in patients undergoing elective surgery for colorectal cancer. Dis Colon Rectum. 2006; 49:507–516.
Article
123. Kirman I, Belizon A, Balik E, Feingold D, Arnell T, Horst P, et al. Perioperative sargramostim (recombinant human GM-CSF) induces an increase in the level of soluble VEGFR1 in colon cancer patients undergoing minimally invasive surgery. Eur J Surg Oncol. 2007; 33:1169–1176.
Article
124. Crucitti A, Corbi M, Tomaiuolo PM, Fanali C, Mazzari A, Lucchetti D, et al. Laparoscopic surgery for colorectal cancer is not associated with an increase in the circulating levels of several inflammation-related factors. Cancer Biol Ther. 2015; 16:671–677.
Article
125. Spittler A, Sautner T, Gornikiewicz A, Manhart N, Oehler R, Bergmann M, et al. Postoperative glycyl-glutamine infusion reduces immunosuppression: partial prevention of the surgery induced decrease in HLA-DR expression on monocytes. Clin Nutr. 2001; 20:37–42.
Article
126. Welters ID, Feurer MK, Preiss V, Müller M, Scholz S, Kwapisz M, et al. Continuous S-(+)-ketamine administration during elective coronary artery bypass graft surgery attenuates pro-inflammatory cytokine response during and after cardiopulmonary bypass. Br J Anaesth. 2011; 106:172–179.
127. Ikuta S, Miki C, Hatada T, Inoue Y, Araki T, Tanaka K, et al. Allogenic blood transfusion is an independent risk factor for infective complications after less invasive gastrointestinal surgery. Am J Surg. 2003; 185:188–193.
Article
128. Leijte GP, Custers H, Gerretsen J, Heijne A, Roth J, Vogl T, et al. Increased plasma levels of danger-associated molecular patterns are associated with immune suppression and postoperative infections in patients undergoing cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Front Immunol. 2018; 9:663.
Article
129. Schilling T, Kozian A, Senturk M, Huth C, Reinhold A, Hedenstierna G, et al. Effects of volatile and intravenous anesthesia on the alveolar and systemic inflammatory response in thoracic surgical patients. Anesthesiology. 2011; 115:65–74.
Article
130. Zak-Nejmark T, Jankowska R, Passowicz-Muszynska E, Malolepszy J, Marciniak M, Jonkisz A, et al. Skin reactivity to histamine and expression of histamine receptors mRNA in lymphocytes of healthy subjects and non-small-cell lung cancer patients before and after surgery. Lung Cancer. 2004; 45:31–38.
Article
131. Leaver HA, Craig SR, Yap PL, Walker WS. Lymphocyte responses following open and minimally invasive thoracic surgery. Eur J Clin Invest. 2000; 30:230–238.
Article
132. Qadan M, Battista C, Gardner SA, Anderson G, Akca O, Polk HC Jr. Oxygen and surgical site infection: a study of underlying immunologic mechanisms. Anesthesiology. 2010; 113:369–377.
133. Kim HI, Kim H, Cho HW, Kim SY, Song KJ, Hyung WJ, et al. The ratio of intra-tumoral regulatory T cells (Foxp3+)/helper T cells (CD4+) is a prognostic factor and associated with recurrence pattern in gastric cardia cancer. J Surg Oncol. 2011; 104:728–733.
Article
134. Lee JY, Son T, Cheong JH, Hyung WJ, Noh SH, Kim CB, et al. Association between chemotherapy-response assays and subsets of tumor-infiltrating lymphocytes in gastric cancer: a pilot study. J Gastric Cancer. 2015; 15:223–230.
Article
135. Lee S, Choi S, Kim SY, Yun MJ, Kim HI. Potential utility of FDG PET-CT as a non-invasive tool for monitoring local immune responses. J Gastric Cancer. 2017; 17:384–393.
Article
136. Zhuang Y, Peng LS, Zhao YL, Shi Y, Mao XH, Chen W, et al. CD8+ T cells that produce interleukin-17 regulate myeloid-derived suppressor cells and are associated with survival time of patients with gastric cancer. Gastroenterology. 2012; 143:951–962.e8.
137. Gérard A, Beemiller P, Friedman RS, Jacobelli J, Krummel MF. Evolving immune circuits are generated by flexible, motile, and sequential immunological synapses. Immunol Rev. 2013; 251:80–96.
Article
138. Xie J, Tato CM, Davis MM. How the immune system talks to itself: the varied role of synapses. Immunol Rev. 2013; 251:65–79.
Article
139. Annunziato F, Romagnani C, Romagnani S. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol. 2015; 135:626–635.
Article
140. Romagnani S. T-cell subsets (Th1 versus Th2). Ann Allergy Asthma Immunol. 2000; 85:9–18.
Article
141. Moraga I, Spangler J, Mendoza JL, Garcia KC. Multifarious determinants of cytokine receptor signaling specificity. Adv Immunol. 2014; 121:1–39.
Article
142. Miller MC, Mayo KH. Chemokines from a structural perspective. Int J Mol Sci. 2017; 18:18.
Article
143. Lin E, Calvano SE, Lowry SF. Inflammatory cytokines and cell response in surgery. Surgery. 2000; 127:117–126.
Article
144. Cavaillon JM, Singer M. Inflammation: From Molecular and Cellular Mechanisms to the Clinic. Weinheim: Wiley-VCH;2017.
145. Datta J, Berk E, Xu S, Fitzpatrick E, Rosemblit C, Lowenfeld L, et al. Anti-HER2 CD4+ T-helper type 1 response is a novel immune correlate to pathologic response following neoadjuvant therapy in HER2-positive breast cancer. Breast Cancer Res. 2015; 17:71.
Article
146. Kvarnström A, Swartling T, Kurlberg G, Bengtson JP, Bengtsson A. Pro-inflammatory cytokine release in rectal surgery: comparison between laparoscopic and open surgical techniques. Arch Immunol Ther Exp (Warsz). 2013; 61:407–411.
Article
147. Wu FP, Sietses C, von Blomberg BM, van Leeuwen PA, Meijer S, Cuesta MA. Systemic and peritoneal inflammatory response after laparoscopic or conventional colon resection in cancer patients: a prospective, randomized trial. Dis Colon Rectum. 2003; 46:147–155.
148. Leung KL, Lai PB, Ho RL, Meng WC, Yiu RY, Lee JF, et al. Systemic cytokine response after laparoscopic-assisted resection of rectosigmoid carcinoma: a prospective randomized trial. Ann Surg. 2000; 231:506–511.
149. Ahlers O, Nachtigall I, Lenze J, Goldmann A, Schulte E, Höhne C, et al. Intraoperative thoracic epidural anaesthesia attenuates stress-induced immunosuppression in patients undergoing major abdominal surgery. Br J Anaesth. 2008; 101:781–787.
150. Sun HZ, Song YL, Wang XY. Effects of different anesthetic methods on cellular immune and neuroendocrine functions in patients with hepatocellular carcinoma before and after surgery. J Clin Lab Anal. 2016; 30:1175–1182.
Article
151. Yeh CN, Lee HL, Liu YY, Chiang KC, Hwang TL, Jan YY, et al. The role of parenteral glutamine supplement for surgical patient perioperatively: result of a single center, prospective and controlled study. Langenbecks Arch Surg. 2008; 393:849–855.
Article
152. Chen DW, Wei Fei Z, Zhang YC, Ou JM, Xu J. Role of enteral immunonutrition in patients with gastric carcinoma undergoing major surgery. Asian J Surg. 2005; 28:121–124.
Article
153. Watt SK, Hasselbalch HC, Skov V, Kjær L, Thomassen M, Kruse TA, et al. Whole blood gene expression profiling in patients undergoing colon cancer surgery identifies differential expression of genes involved in immune surveillance, inflammation and carcinogenesis. Surg Oncol. 2018; 27:208–215.
Article
154. Allen CJ, Griswold AJ, Schulman CI, Sleeman D, Levi JU, Livingstone AS, et al. Global gene expression change induced by major thoracoabdominal surgery. Ann Surg. 2017; 266:981–987.
Article
155. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014; 513:202–209.
156. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015; 12:453–457.
Article
157. Altman DG, McShane LM, Sauerbrei W, Taube SE. Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK): explanation and elaboration. PLoS Med. 2012; 9:e1001216.
Article
158. Cohen JF, Korevaar DA, Altman DG, Bruns DE, Gatsonis CA, Hooft L, et al. STARD 2015 guidelines for reporting diagnostic accuracy studies: explanation and elaboration. BMJ Open. 2016; 6:e012799.
Article
159. Welsch T, Müller SA, Ulrich A, Kischlat A, Hinz U, Kienle P, et al. C-reactive protein as early predictor for infectious postoperative complications in rectal surgery. Int J Colorectal Dis. 2007; 22:1499–1507.
Article
160. Lee K, Hwang H, Nam KT. Immune response and the tumor microenvironment: how they communicate to regulate gastric cancer. Gut Liver. 2014; 8:131–139.
Article
161. Kim CH, Park SM, Kim JJ. The impact of preoperative low body mass index on postoperative complications and long-term survival outcomes in gastric cancer patients. J Gastric Cancer. 2018; 18:274–286.
Article
162. Seo HS, Jung YJ, Kim JH, Park CH, Kim IH, Lee HH. Long-term nutritional outcomes of near-total gastrectomy in gastric cancer treatment: a comparison with total gastrectomy using propensity score matching analysis. J Gastric Cancer. 2018; 18:189–199.
Article
163. Lee HJ, Hyung WJ, Yang HK, Han SU, Park YK, An JY, et al. Short-term outcomes of a multicenter randomized controlled trial comparing laparoscopic distal gastrectomy with D2 lymphadenectomy to open distal gastrectomy for locally advanced gastric cancer (KLASS-02-RCT). Ann Surg. 2019.
Article
164. Kim HH, Han SU, Kim MC, Kim W, Lee HJ, Ryu SW, et al. Effect of laparoscopic distal gastrectomy vs open distal gastrectomy on long-term survival among patients with stage I gastric cancer: The KLASS-01 Randomized Clinical Trial. JAMA Oncol. 2019; 5:506–513.
165. Desiderio J, Stewart CL, Sun V, Melstrom L, Warner S, Lee B, et al. Enhanced recovery after surgery for gastric cancer patients improves clinical outcomes at a US Cancer Center. J Gastric Cancer. 2018; 18:230–241.
Article
166. Mortensen K, Nilsson M, Slim K, Schäfer M, Mariette C, Braga M, et al. Consensus guidelines for enhanced recovery after gastrectomy: Enhanced Recovery After Surgery (ERAS®) Society recommendations. Br J Surg. 2014; 101:1209–1229.
Full Text Links
  • JGC
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