Anesth Pain Med.  2024 Apr;19(2):94-108. 10.17085/apm.24041.

Anesthesia and cancer recurrence: a narrative review

Affiliations
  • 1Department of~, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea

Abstract

Cancer is a leading cause of death worldwide. With the increasingly aging population, the number of emerging cancer cases is expected to increase markedly in the foreseeable future. Surgical resection with adjuvant therapy is the best available option for the potential cure of many solid tumors; thus, approximately 80% of patients with cancer undergo at least one surgical procedure during their disease. Agents used in general anesthesia can modulate cytokine release, transcription factors, and/or oncogenes. This can affect host immunity and the capability of cancer cells to survive and migrate, not only during surgery but for up to several weeks after surgery. However, it remains unknown whether exposure to anesthetic agents affects cancer recurrence or metastasis. This review explores the current literature to explain whether and how the choice of anesthetic and perioperative medication affect cancer surgery outcomes.

Keyword

Anesthesia; Cancer recurrence; Opioids; Propofol; Volatile anesthetics

Reference

1. Luo J, Shi Y, Wang X, Zhang R, Chen S, Yu W, et al. A 20-year research trend analysis of the influence of anesthesia on tumor prognosis using bibliometric methods. Front Oncol. 2021; 11:683232.
Article
2. Sekandarzad MW, Van Zundert AAJ, Lirk PB, Doornebal CW, Hollmann MW. Perioperative anesthesia care and tumor progression. Anesth Analg. 2017; 124:1697–708.
Article
3. Forget P, Aguirre JA, Bencic I, Borgeat A, Cama A, Condron C, et al. How anesthetic, analgesic and other non-surgical techniques during cancer surgery might affect postoperative oncologic outcomes: A summary of current state of evidence. Cancers (Basel). 2019; 11:592.
Article
4. Montejano J, Jevtovic-Todorovic V. Anesthesia and cancer, friend or foe? A narrative review. Front Oncol. 2021; 11:803266.
Article
5. Vona G, Sabile A, Louha M, Sitruk V, Romana S, Schütze K, et al. Isolation by size of epithelial tumor cells: a new method for the immunomorphological and molecular characterization of circulating tumor cells. Am J Pathol. 2000; 156:57–63.
6. Horowitz M, Neeman E, Sharon E, Ben-Eliyahu S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat Rev Clin Oncol. 2015; 12:213–26.
Article
7. Hiller JG, Perry NJ, Poulogiannis G, Riedel B, Sloan EK. Perioperative events influence cancer recurrence risk after surgery. Nat Rev Clin Oncol. 2018; 15:205–18.
Article
8. Eschwège P, Dumas F, Blanchet P, Le Maire V, Benoit G, Jardin A, et al. Haematogenous dissemination of prostatic epithelial cells during radical prostatectomy. Lancet. 1995; 346:1528–30.
Article
9. Foss OP, Brennhovd IO, Messelt OT, Efskind J, Liverud K. Invasion of tumor cells into the bloodstream caused by palpation or biopsy of the tumor. Surgery. 1966; 59:691–5.
10. Denis MG, Lipart C, Leborgne J, LeHur PA, Galmiche JP, Denis M, et al. Detection of disseminated tumor cells in peripheral blood of colorectal cancer patients. Int J Cancer. 1997; 74:540–4.
Article
11. L Holmgren L, O'Reilly MS, Folkman J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med. 1995; 1:149–53.
Article
12. Shakhar G, Ben-Eliyahu S. Potential prophylactic measures against postoperative immunosuppression: could they reduce recurrence rates in oncological patients? Ann Surg Oncol. 2003; 10:972–92.
Article
13. Smyth MJ, Godfrey DI, Trapani JA. A fresh look at tumor immunosurveillance and immunotherapy. Nat Immunol. 2001; 2:293–9.
Article
14. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002; 420:860–7.
Article
15. Chen P, Luo X, Dai G, Jiang Y, Luo Y, Peng S. Dexmedetomidine promotes the progression of hepatocellular carcinoma through hepatic stellate cell activation. Exp Mol Med. 2020; 52:106274.
Article
16. Deng F, Ouyang M, Wang X, Yao X, Chen Y, Tao T, et al. Differential role of intravenous anesthetics in colorectal cancer progression: implications for clinical application. Oncotarget. 2016; 7:77087–95.
Article
17. Nguyen J, Luk K, Vang D, Soto W, Vincent L, Robiner S, et al. Morphine stimulates cancer progression and mast cell activation and impairs survival in transgenic mice with breast cancer. Br J Anaesth. 2014; 113 Suppl 1(Suppl 1):i4–13.
Article
18. Su X, Fan Y, Yang L, Huang J, Qiao F, Fang Y, et al. Dexmedetomidine expands monocytic myeloid-derived suppressor cells and promotes tumour metastasis after lung cancer surgery. J Transl Med. 2018; 16:347.
Article
19. Szpunar MJ, Burke KA, Dawes RP, Brown EB, Madden KS. The antidepressant desipramine and alpha2-adrenergic receptor activation promote breast tumor progression in association with altered collagen structure. Cancer Prev Res (Phila). 2013; 6:1262–72.
20. Tian H, Hou L, Xiong Y, Cheng Q, Huang J. Effect of dexmedetomidine-mediated insulin-like growth factor 2 (IGF2) signal pathway on immune function and invasion and migration of cancer cells in rats with ovarian cancer. Med Sci Monit. 2019; 25:4655–64.
Article
21. Zhang P, He H, Bai Y, Liu W, Huang L. Dexmedetomidine suppresses the progression of esophageal cancer via miR-143-3p/epidermal growth factor receptor pathway substrate 8 axis. Anticancer Drugs. 2020; 31:693–701.
Article
22. Kim R. Anesthetic technique and cancer recurrence in oncologic surgery: unraveling the puzzle. Cancer Metastasis Rev. 2017; 36:159–77.
Article
23. Wall T, Sherwin A, Ma D, Buggy DJ. Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: a narrative review. Br J Anaesth. 2019; 123:135–50.
Article
24. Bar-Yosef S, Melamed R, Page GG, Shakhar G, Shakhar K, Ben-Eliyahu S. Attenuation of the tumor-promoting effect of surgery by spinal blockade in rats. Anesthesiology. 2001; 94:1066–73.
Article
25. 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; 90:1411–4.
Article
26. Yeager MP, Colacchio TA, Yu CT, Hildebrandt L, Howell AL, Weiss J, et al. Morphine inhibits spontaneous and cytokine-enhanced natural killer cell cytotoxicity in volunteers. Anesthesiology. 1995; 83:500–8.
Article
27. Schlagenhauff B, Ellwanger U, Breuninger H, Stroebel W, Rassner G, Garbe C. Prognostic impact of the type of anaesthesia used during the excision of primary cutaneous melanoma. Melanoma Res. 2000; 10:165–9.
Article
28. Brand JM, Kirchner H, Poppe C, Schmucker P. The effects of general anesthesia on human peripheral immune cell distribution and cytokine production. Clin Immunol Immunopathol. 1997; 83:190–4.
Article
29. Markovic SN, Knight PR, Murasko DM. Inhibition of interferon stimulation of natural killer cell activity in mice anesthetized with halothane or isoflurane. Anesthesiology. 1993; 78:700–6.
Article
30. Shapiro J, Jersky J, Katzav S, Feldman M, Segal S. Anesthetic drugs accelerate the progression of postoperative metastases of mouse tumors. J Clin Invest. 1981; 68:678–85.
Article
31. Melamed R, Bar-Yosef S, Shakhar G, Shakhar K, Ben-Eliyahu S. Suppression of natural killer cell activity and promotion of tumor metastasis by ketamine, thiopental, and halothane, but not by propofol: mediating mechanisms and prophylactic measures. Anesth Analg. 2003; 97:1331–9.
Article
32. Looney M, Doran P, Buggy DJ. Effect of anesthetic technique on serum vascular endothelial growth factor C and transforming growth factor beta in women undergoing anesthesia and surgery for breast cancer. Anesthesiology. 2010; 113:1118–25.
33. Tavare AN, Perry NJS, Benzonana LL, Takata M, Ma D. Cancer recurrence after surgery: direct and indirect effects of anesthetic agents. Int J Cancer. 2012; 130:1237–50.
Article
34. Buckley A, McQuaid S, Johnson P, Buggy DJ. Effect of anaesthetic technique on the natural killer cell anti-tumour activity of serum from women undergoing breast cancer surgery: a pilot study. Br J Anaesth. 2014; 113 Suppl 1:i56–62.
Article
35. Jaura AI, Flood G, Gallagher HC, Buggy DJ. Differential effects of serum from patients administered distinct anaesthetic techniques on apoptosis in breast cancer cells in vitro: a pilot study. Br J Anaesth. 2014; 113 Suppl 1:i63–7.
Article
36. Cho JS, Lee MH, Kim SI, Park S, Park HS, Oh E. The effects of perioperative anesthesia and analgesia on immune function in patients undergoing breast cancer resection: A prospective randomized study. Int J Med Sci. 2017; 14:970–6.
Article
37. Iwasaki M, Zhao H, Jaffer T, Unwith S, Benzonana L, Lian Q. Volatile anaesthetics enhance the metastasis related cellular signalling including CXCR2 of ovarian cancer cells. Oncotarget. 2016; 7:26042–56.
Article
38. Markovic-Bozic J, Karpe B, Potocnik I, Jerin A, Vranic A, Novak-Jankovic V. Effect of propofol and sevoflurane on the inflammatory response of patients undergoing craniotomy. BMC Anesthesiol. 2016; 16:18.
Article
39. Hiller JG, Perry NJ, Poulogiannis G, Riedel B, Sloan EK. Perioperative events influence cancer recurrence risk after surgery. Nat Rev Clin Oncol. 2018; 15:205–18.
Article
40. Lai HC, Lee MS, Lin C, Lin KT, Huang YH, Wong CS. Propofol-based total intravenous anaesthesia is associated with better survival than desflurane anaesthesia in hepatectomy for hepatocellular carcinoma: a retrospective cohort study. Br J Anaesth. 2019; 123:151–60.
Article
41. Wigmore TJ, Mohammed K, Jhanji S. Long-term survival for patients undergoing volatile versus IV anesthesia for cancer surgery: A retrospective analysis. Anesthesiology. 2016; 124:69–79.
Article
42. Lee JH, Kang SH, Kim Y, Kim HA, Kim BS. Effects of propofol-based total intravenous anesthesia on recurrence and overall survival in patients after modified radical mastectomy: a retrospective study. Korean J Anesthesiol. 2016; 69:126–32.
Article
43. Zheng X, Wang Y, Dong L, Zhao S, Wang L, Chen H, et al. Effects of propofol-based total intravenous anesthesia on gastric cancer: a retrospective study. Onco Targets Ther. 2018; 11:1141–8.
Article
44. Jun IJ, Jo JY, Kim JI, Chin JH, Kim WJ, Kim HR, et al. Impact of anesthetic agents on overall and recurrence-free survival in patients undergoing esophageal cancer surgery: A retrospective observational study. Sci Rep. 2017; 7:14020.
Article
45. Yoo S, Lee HB, Han W, Noh DY, Park SK, Kim WH, et al. Total intravenous anesthesia versus inhalation anesthesia for breast cancer surgery: A retrospective cohort study. Anesthesiology. 2019; 130:31–40.
Article
46. Hasselager RP, Hallas J, Gögenur I. Inhalation or total intravenous anaesthesia and recurrence after colorectal cancer surgery: a propensity score matched Danish registry-based study. Br J Anaesth. 2021; 126:921–30.
Article
47. Enlund M, Berglund A, Enlund A, Bergkvist L. Volatile versus propofol general anesthesia and long-term survival after breast cancer surgery: A national registry retrospective cohort study. Anesthesiology. 2022; 137:315–26.
48. Yoon S, Jung SY, Kim MS, Yoon D, Cho Y, Jeon Y. Impact of propofol-based total intravenous anesthesia versus inhalation anesthesia on long-term survival after cancer surgery in a nationwide cohort. Ann Surg. 2023; 278:1024–31.
Article
49. Makito K, Matsui H, Fushimi K, Yasunaga H. Volatile versus total intravenous anesthesia for cancer prognosis in patients having digestive cancer surgery. Anesthesiology. 2020; 133:764–73.
50. Chang CY, Wu MY, Chien YJ, Su IM, Wang SC, Kao MC. Anesthesia and long-term oncological outcomes: A systematic review and meta-analysis. Anesth Analg. 2021; 132:623–34.
Article
51. Hovaguimian F, Braun J, Schläpfer M, Puhan MA, Beck-Schimmer B. Anesthesia and circulating tumor cells: Reply. Anesthesiology. 2021; 134:507–8.
Article
52. Enlund M, Berglund A, Enlund A, Lundberg J, Wärnberg F, Wang DX, et al. Impact of general anaesthesia on breast cancer survival: a 5-year follow up of a pragmatic, randomised, controlled trial, the CAN-study, comparing propofol and sevoflurane. EClinicalMedicine. 2023; 60:102037.
Article
53. Sessler DI, Pei L, Huang Y, Fleischmann E, Marhofer P, Kurz A, et al. Recurrence of breast cancer after regional or general anaesthesia: a randomised controlled trial. Lancet. 2019; 394:1807–15.
Article
54. Tsuchiya Y, Sawada S, Yoshioka I, Ohashi Y, Matsuo M, Harimaya Y, et al. Increased surgical stress promotes tumor metastasis. Surgery. 2003; 133:547–55.
Article
55. Ganesh K, Massague J. Targeting metastatic cancer. Nat Med. 2021; 27:34–44.
Article
56. Suhail Y, Cain MP, Vanaja K, Kurywchak PA, Levchenko A, Kalluri R, et al. Systems biology of cancer metastasis. Cell Syst. 2019; 9:109–27.
Article
57. Alazawi W, Pirmadjid N, Lahiri R, Bhattacharya S. Inflammatory and immune responses to surgery and their clinical impact. Ann Surg. 2016; 264:73–80.
Article
58. Angka L, Khan ST, Kilgour MK, Xu R, Kennedy MA, Auer RC. Dysfunctional natural killer cells in the aftermath of cancer surgery. Int J Mol Sci. 2017; 18:1787.
Article
59. Perez-Sayans M, Pérez-Sayáns M, Somoza-Martín JM, Barros-Angueira F, Diz PG, Rey JMG, García-García A. Beta-adrenergic receptors in cancer: therapeutic implications. Oncol Res. 2010; 19:45–54.
60. Wu WKK, Sung JJY, Lee CW, Yu J, Cho CH. Cyclooxygenase-2 in tumorigenesis of gastrointestinal cancers: an update on the molecular mechanisms. Cancer Lett. 2010; 295:7–16.
Article
61. Huang H, Benzonana LL, Zhao H, Watts HR, Perry NJS, Bevan C, et al. Prostate cancer cell malignancy via modulation of HIF-1alpha pathway with isoflurane and propofol alone and in combination. Br J Cancer. 2014; 111:1338–49.
Article
62. Zhu L, Zhang Y, Zhang Z, Ding X, Gong C, Qian Y. Activation of PI3K/Akt/HIF-1alpha signaling is involved in lung protection of dexmedetomidine in patients undergoing video-assisted thoracoscopic surgery: A pilot study. Drug Des Devel Ther. 2020; 14:5155–66.
63. Chen HY, Li GH, Tan GC, Liang H, Lai XH, Huang Q, et al. Dexmedetomidine enhances hypoxia-induced cancer cell progression. Exp Ther Med. 2019; 18:4820–8.
Article
64. Cata JP, Singh V, Lee BM, Villarreal J, Mehran JR, Yu J, et al. Intraoperative use of dexmedetomidine is associated with decreased overall survival after lung cancer surgery. J Anaesthesiol Clin Pharmacol. 2017; 33:317–23.
Article
65. Darby IA, Hewitson TD. Hypoxia in tissue repair and fibrosis. Cell Tissue Res. 2016; 365:553–62.
Article
66. Schito L, Semenza GL. Hypoxia-inducible factors: master regulators of cancer progression. Trends Cancer. 2016; 2:758–70.
Article
67. Karnezis T, Shayan R, Caesar C, Roufail S, Harris NC, Ardipradja K, et al. VEGF-D promotes tumor metastasis by regulating prostaglandins produced by the collecting lymphatic endothelium. Cancer Cell. 2012; 21:181–95.
Article
68. Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008; 14:818–29.
Article
69. Stollings LM, Jia LJ, Tang P, Dou H, Lu B, Xu Y. Immune modulation by volatile anesthetics. Anesthesiology. 2016; 125:399–411.
Article
70. Benzonana LL, Perry NJS, Watts HR, Yang B, Perry IA, Coombes C, et al. Isoflurane, a commonly used volatile anesthetic, enhances renal cancer growth and malignant potential via the hypoxia-inducible factor cellular signaling pathway in vitro. Anesthesiology. 2013; 119:593–605.
Article
71. Desmond F, McCormack J, Mulligan N, Stokes M, Buggy DJ. Effect of anaesthetic technique on immune cell infiltration in breast cancer: a follow-up pilot analysis of a prospective, randomised, investigator-masked study. Anticancer Res. 2015; 35:1311–9.
72. Ecimovic P, Blaithnaid McHugh B, Murray D, Doran P, Buggy DJ. Effects of sevoflurane on breast cancer cell function in vitro. Anticancer Res. 2013; 33:4255–60.
73. Inada T, Kubo K, Shingu K. Promotion of interferon-gamma production by natural killer cells via suppression of murine peritoneal macrophage prostaglandin E2 production using intravenous anesthetic propofol. Int Immunopharmacol. 2010; 10:1200–8.
Article
74. Luo X, Zhao H, Hennah L, Ning J, Liu J, Tu H, et al. Impact of isoflurane on malignant capability of ovarian cancer in vitro. Br J Anaesth. 2015; 114:831–9.
Article
75. Deng X, Vipani M, Liang G, Gouda D, Wang B, Wei H. Sevoflurane modulates breast cancer cell survival via modulation of intracellular calcium homeostasis. BMC Anesthesiol. 2020; 20:253.
Article
76. Liu Y, Sun J, Wu T, Lu X, Du Y, Duan H, et al. Effects of serum from breast cancer surgery patients receiving perioperative dexmedetomidine on breast cancer cell malignancy: A prospective randomized controlled trial. Cancer Med. 2019; 8:7603–12.
Article
77. Ciechanowicz S, Zhao H, Chen Q, Cui J, Mi E, Mi E, et al. Differential effects of sevoflurane on the metastatic potential and chemosensitivity of non-small-cell lung adenocarcinoma and renal cell carcinoma in vitro. Br J Anaesth. 2018; 120:368–75.
78. Hooijmans CR, Geessink FJ, Ritskes-Hoitinga M, Scheffer GJ. A systematic review of the modifying effect of anaesthetic drugs on metastasis in animal models for cancer. PLoS One. 2016; 11:e0156152.
Article
79. Jiang S, Liu Y, Huang L, Zhang F, Kang R. Effects of propofol on cancer development and chemotherapy: Potential mechanisms. Eur J Pharmacol. 2018; 831:46–51.
Article
80. Yang C, Gao J, Yan N, Wu B, Ren Y, Li H, et al. Propofol inhibits the growth and survival of gastric cancer cells in vitro through the upregulation of ING3. Oncol Rep. 2017; 37:587–93.
Article
81. Yang N, Liang Y, Yang P, Ji F. Propofol suppresses LPS-induced nuclear accumulation of HIF-1α and tumor aggressiveness in non-small cell lung cancer. Oncol Rep. 2017; 37:2611–9.
Article
82. Yu B, Gao W, Zhou H, Miao X, Chang Y, Wang L, et al. Propofol induces apoptosis of breast cancer cells by downregulation of miR-24 signal pathway. Cancer Biomark. 2018; 21:513–9.
Article
83. Du Q, Liu J, Zhang X, Zhang X, Zhu H, Wei M, et al. Propofol inhibits proliferation, migration, and invasion but promotes apoptosis by regulation of Sox4 in endometrial cancer cells. Braz J Med Biol Res. 2018; 51:e6803.
Article
84. Zhou CL, Li JJ, Ji P. Propofol suppresses esophageal squamous cell carcinoma cell migration and invasion by down-regulation of sex-determining region Y-box 4 (SOX4). Med Sci Monit. 2017; 23:419–27.
Article
85. Chen J, Ju HL, Yuan XY, Wang TJ, Lai BQ. SOX4 is a potential prognostic factor in human cancers: a systematic review and meta-analysis. Clin Transl Oncol. 2016; 18:65–72.
Article
86. Freeman J, Crowley PD, Foley AG, Gallagher HC, Iwasaki M, Ma D, et al. Effect of perioperative lidocaine, propofol and steroids on pulmonary metastasis in a murine model of breast cancer surgery. Cancers (Basel). 2019; 11:613.
Article
87. Ferrell JK, Cattano D, Brown RE, Patel CB, Karni RJ. The effects of anesthesia on the morphoproteomic expression of head and neck squamous cell carcinoma: a pilot study. Transl Res. 2015; 166:674–82.
Article
88. Xu YJ, Li SY, Cheng Q, Chen WK, Wang SL, Ren Y, et al. Effects of anaesthesia on proliferation, invasion and apoptosis of LoVo colon cancer cells in vitro. Anaesthesia. 2016; 71:147–54.
Article
89. Selby LV, Fernandez-Bustamante A, Ejaz A, Gleisner A, Pawlik TM, Douin DJ. Association between anesthesia delivered during tumor resection and cancer survival: a systematic review of a mixed picture with constant themes. J Gastrointest Surg. 2021; 25:2129–41.
Article
90. Enlund M, Berglund A, Andreasson K, Cicek C, Enlund A, Bergkvist J. The choice of anaesthetic--sevoflurane or propofol--and outcome from cancer surgery: a retrospective analysis. Ups J Med Sci. 2014; 119:251–61.
Article
91. Oh TK, Kim K, Jheon S, Lee J, Do SH, Hwang JW. Long-term oncologic outcomes for patients undergoing volatile versus intravenous anesthesia for non-small cell lung cancer surgery: A retrospective propensity matching analysis. Cancer Control. 2018; 25:1073274818775360.
92. Oh TK, Kim HH, Jeon YT. Retrospective analysis of 1-year mortality after gastric cancer surgery: Total intravenous anesthesia versus volatile anesthesia. Acta Anaesthesiol Scand. 2019; 63:1169–77.
Article
93. Enlund M, Berglund A, Ahlstrand R, Walldén J, Lundberg J, Wärnberg F, et al. Survival after primary breast cancer surgery following propofol or sevoflurane general anesthesia-A retrospective, multicenter, database analysis of 6305 Swedish patients. Acta Anaesthesiol Scand. 2020; 64:1048–54.
Article
94. Wu ZF, Lee MS, Wong CS, Lu CH, Huang YS, Lin KT, et al. Propofol-based total intravenous anesthesia is associated with better survival than desflurane anesthesia in colon cancer surgery. Anesthesiology. 2018; 129:932–41.
Article
95. Yap A, Lopez-Olivo MA, Dubowitz J, Hiller J, Riedel B; Global Onco-Anesthesia Research Collaboration Group. Anesthetic technique and cancer outcomes: a meta-analysis of total intravenous versus volatile anesthesia. Can J Anaesth. 2019; 66:546–61.
96. Kim MH, Kim DW, Kim JH, Lee KY, Park S, Yoo YC. Does the type of anesthesia really affect the recurrence-free survival after breast cancer surgery? Oncotarget. 2017; 8:90477–87.
Article
97. Miao L, Lv X, Huang C, Li P, Sun Y, Jiang H. Long-term oncological outcomes after oral cancer surgery using propofol-based total intravenous anesthesia versus sevoflurane-based inhalation anesthesia: A retrospective cohort study. PLoS One. 2022; 17:e0268473.
Article
98. Liu J, Yang L, Guo X, Jin G, Wang Q, Lv D, et al. Sevoflurane suppresses proliferation by upregulating microRNA-203 in breast cancer cells. Mol Med Rep. 2018; 18:455–60.
Article
99. Wang L, Wang T, Gu JQ, Su HB. Volatile anesthetic sevoflurane suppresses lung cancer cells and miRNA interference in lung cancer cells. Onco Targets Ther. 2018; 11:5689–93.
Article
100. Yan T, Zhang GH, Wang BN, Sun L, Zheng H. Effects of propofol/remifentanil-based total intravenous anesthesia versus sevoflurane-based inhalational anesthesia on the release of VEGF-C and TGF-beta and prognosis after breast cancer surgery: a prospective, randomized and controlled study. BMC Anesthesiol. 2018; 18:131.
101. Oh CS, Lee J, Yoon TG, Seo EH, Park HJ, Piao L, et al. Effect of equipotent doses of propofol versus sevoflurane anesthesia on regulatory T cells after breast cancer surgery. Anesthesiology. 2018; 129:921–31.
102. Oh CS, Park HJ, Piao L, Sohn KM, Koh SE, Hwang DY, et al. Expression profiles of immune cells after propofol or sevoflurane anesthesia for colorectal cancer surgery: A prospective double-blind randomized trial. Anesthesiology. 2022; 136:448–58.
Article
103. O'Bryan LJ, Atkins KJ, Lipszyc A, Scott DA, Silbert BS, Evered LA. Inflammatory biomarker levels after propofol or sevoflurane anesthesia: A meta-analysis. Anesth Analg. 2022; 134:69–81.
104. Enlund M, Enlund A, Berglund A, Bergkvist L. Rationale and design of the CAN study: an RCT of survival after propofol- or sevoflurane-based anesthesia for cancer surgery. Curr Pharm Des. 2019; 25:3028–33.
Article
105. Dubowitz JA, Cata JP, De Silva AP, Braat S, Shan D, Yee K, et al. Volatile anaesthesia and peri-operative outcomes related to cancer: a feasibility and pilot study for a large randomised control trial. Anaesthesia. 2021; 76:1198–206.
Article
106. Beilin B, Martin FC, Shavit Y, Gale RP, Liebeskind JC. Suppression of natural killer cell activity by high-dose narcotic anesthesia in rats. Brain Behav Immun. 1989; 3:129–37.
Article
107. Shavit Y, Terman GW, Lewis JW, Zane CJ, Gale RP, Liebeskind JC. Effects of footshock stress and morphine on natural killer lymphocytes in rats: studies of tolerance and cross-tolerance. Brain Res. 1986; 372:382–5.
Article
108. Boland JW, Pockley AG. Influence of opioids on immune function in patients with cancer pain: from bench to bedside. Br J Pharmacol. 2018; 175:2726–36.
Article
109. Cui JH, Jiang WW, Liao YJ, Wang QH, Min Xu, Li Y. Effects of oxycodone on immune function in patients undergoing radical resection of rectal cancer under general anesthesia. Medicine (Baltimore). 2017; 96:e7519.
Article
110. Ni Eochagain A, Burns D, Riedel B, Sessler DI, Buggy DJ. The effect of anaesthetic technique during primary breast cancer surgery on neutrophil-lymphocyte ratio, platelet-lymphocyte ratio and return to intended oncological therapy. Anaesthesia. 2018; 73:603–11.
Article
111. Wu Q, Chen X, Wang J, Sun P, Weng M, Chen W, et al. Nalmefene attenuates malignant potential in colorectal cancer cell via inhibition of opioid receptor. Acta Biochim Biophys Sin (Shanghai). 2018; 50:156–63.
Article
112. Sen Y, Xiyang H, Yu H. Effect of thoracic paraspinal block-propofol intravenous general anesthesia on VEGF and TGF-beta in patients receiving radical resection of lung cancer. Medicine (Baltimore). 2019; 98:e18088.
113. Ma M, Wang X, Liu N, Shan F, Feng Y. Low-dose naltrexone inhibits colorectal cancer progression and promotes apoptosis by increasing M1-type macrophages and activating the Bax/Bcl-2/caspase-3/PARP pathway. Int Immunopharmacol. 2020; 83:106388.
Article
114. Saurer TB, Ijames SG, Carrigan KA, Lysle DT. Neuroimmune mechanisms of opioid-mediated conditioned immunomodulation. Brain Behav Immun. 2008; 22:89–97.
Article
115. Kraus J. Regulation of mu-opioid receptors by cytokines. Front Biosci (Schol Ed). 2009; 1:164–70.
Article
116. Wigmore T, Farquhar-Smith P. Opioids and cancer: friend or foe? Curr Opin Support Palliat Care. 2016; 10:109–18.
117. Singleton PA, Mirzapoiazova T, Hasina R, Salgia R, Moss J. Increased mu-opioid receptor expression in metastatic lung cancer. Br J Anaesth. 2014; 113 Suppl 1(Suppl 1):i103–8.
118. Zhang H, Sun M, Zhou D, Gorur A, Sun Z, Zeng W, et al. Increased mu-opioid receptor expression is associated with reduced disease-free and overall survival in laryngeal squamous cell carcinoma. Br J Anaesth. 2020; 125:722–9.
Article
119. Zylla D, Gourley BL, Vang D, Jackson S, Boatman S, Lindgren B, et al. Opioid requirement, opioid receptor expression, and clinical outcomes in patients with advanced prostate cancer. Cancer. 2013; 119:4103–10.
Article
120. Bortsov AV, Millikan RC, Belfer I, Boortz-Marx RL, Arora H, McLean SA. μ-Opioid receptor gene A118G polymorphism predicts survival in patients with breast cancer. Anesthesiology. 2012; 116:896–902.
Article
121. Janku F, Johnson LK, Karp DD, Atkins JT, Singleton PA, Moss J. Treatment with methylnaltrexone is associated with increased survival in patients with advanced cancer. Ann Oncol. 2016; 27:2032–8.
Article
122. Mathew B, Lennon FE, Siegler J, Mirzapoiazova T, Mambetsariev N, Sammani S, et al. The novel role of the mu opioid receptor in lung cancer progression: a laboratory investigation. Anesth Analg. 2011; 112:558–67.
123. Bimonte S, Barbieri A, Cascella M, Rea D, Palma G, Vecchio VD, et al. The effects of naloxone on human breast cancer progression: in vitro and in vivo studies on MDA.MB231 cells. Onco Targets Ther. 2018; 11:185–91.
Article
124. Connolly C, Buggy DJ. Opioids and tumour metastasis: does the choice of the anesthetic-analgesic technique influence outcome after cancer surgery? Curr Opin Anaesthesiol. 2016; 29:468–74.
125. Bugada D, Lorini LF, Lavand'homme P. Opioid free anesthesia: evidence for short and long-term outcome. Minerva Anestesiol. 2021; 87:230–7.
Article
126. Chen J, Luo F, Lei M, Chen Z. A study on cellular immune function of patients treated with radical resection of pulmonary carcinoma with two different methods of anesthesia and analgesia. J BUON. 2017; 22:1416–21.
127. Koodie L, Yuan H, Pumper JA, Yu H, Charboneau R, Ramkrishnan S, et al. Morphine inhibits migration of tumor-infiltrating leukocytes and suppresses angiogenesis associated with tumor growth in mice. Am J Pathol. 2014; 184:1073–84.
Article
128. Doornebal CW, Vrijland K, Hau CS, Coffelt SB, Ciampricotti M, Jonkers J, et al. Morphine does not facilitate breast cancer progression in two preclinical mouse models for human invasive lobular and HER2+ breast cancer. Pain. 2015; 156:1424–32.
Article
129. Afsharimani B, Baran J, Watanabe S, Lindner D, Cabot PJ, Parat MO. Morphine and breast tumor metastasis: the role of matrix-degrading enzymes. Clin Exp Metastasis. 2014; 31:149–58.
Article
130. Friesen C, Hormann I, Roscher M, Fichtner I, Alt A, Hilger R, et al. Opioid receptor activation triggering downregulation of cAMP improves effectiveness of anti-cancer drugs in treatment of glioblastoma. Cell Cycle. 2014; 13:1560–70.
Article
131. Friesen C, Roscher M, Hormann I, Fichtner I, Alt A, Hilger RA, et al. Cell death sensitization of leukemia cells by opioid receptor activation. Oncotarget. 2013; 4:677–90.
Article
132. Kim JY, Ahn HJ, Jin Kim K, Kim J, Lee SH, Chae HB. Morphine suppresses lung cancer cell proliferation through the interaction with opioid growth factor receptor: An in vitro and human lung tissue study. Anesth Analg. 2016; 123:1429–36.
133. Silagy AW, Hannum ML, Mano R, Attalla K, Scarpa JR, DiNatale RG, et al. Impact of intraoperative opioid and adjunct analgesic use on renal cell carcinoma recurrence: role for onco-anaesthesia. Br J Anaesth. 2020; 125:e402–4.
Article
134. Cata JP, Keerty V, Keerty D, Feng L, Norman PH, Gottumukkala V, et al. A retrospective analysis of the effect of intraoperative opioid dose on cancer recurrence after non-small cell lung cancer resection. Cancer Med. 2014; 3:900–8.
Article
135. Oh TK, Jeon JH, Lee JM, Kim MS, Kim JH, Cho H, et al. Investigation of opioid use and long-term oncologic outcomes for non-small cell lung cancer patients treated with surgery. PLoS One. 2017; 12:e0181672.
Article
136. Tai YH, Wu HL, Chang WK, Tsou MY, Chen HH, Chang KY. Intraoperative fentanyl consumption does not impact cancer recurrence or overall survival after curative colorectal cancer resection. Sci Rep. 2017; 7:10816.
Article
137. Montagna G, Gupta HV, Hannum M, Tan KS, Lee J, Scarpa JR, et al. Intraoperative opioids are associated with improved recurrence-free survival in triple-negative breast cancer. Br J Anaesth. 2021; 126:367–76.
Article
138. Diaz-Cambronero O, Mazzinari G, Cata JP. Perioperative opioids and colorectal cancer recurrence: a systematic review of the literature. Pain Manag. 2018; 8:353–61.
Article
139. Du YT, Li YW, Zhao BJ, Guo XY, Feng Y, Zuo MZ, et al. Long-term survival after combined epidural-general anesthesia or general anesthesia alone: follow-up of a randomized trial. Anesthesiology. 2021; 135:233–45.
Article
140. Xu ZZ, Li HJ, Li MH, Huang SM, Li X, Liu QH, et al. Epidural anesthesia-analgesia and recurrence-free survival after lung cancer surgery: a randomized trial. Anesthesiology. 2021; 135:419–32.
Article
141. Yuval JB, Lee J, Wu F, Thompson HM, Verheij FS, Gupta HV, et al. Intraoperative opioids are associated with decreased recurrence rates in colon adenocarcinoma: a retrospective observational cohort study. Br J Anaesth. 2022; 129:172–81.
Article
142. Franchi S, Moschetti G, Amodeo G, Sacerdote P. Do all opioid drugs share the same immunomodulatory properties? A review from animal and human studies. Front Immunol. 2019; 10:2914.
Article
143. Cata JP, Sessler DI. Lost in translation: Failure of preclinical studies to accurately predict the effect of regional analgesia on cancer recurrence. Anesthesiology. 2024; 140:361–74.
Article
144. Fleischmann E, Marschalek C, Schlemitz K, Dalton JE, Gruenberger T, Herbst F, et al. Nitrous oxide may not increase the risk of cancer recurrence after colorectal surgery: a follow-up of a randomized controlled trial. BMC Anesthesiol. 2009; 9:1.
Article
145. Forget P, Collet V, Lavand'homme P, Kock MD. Does analgesia and condition influence immunity after surgery? Effects of fentanyl, ketamine and clonidine on natural killer activity at different ages. Eur J Anaesthesiol. 2010; 27:233–40.
Article
146. Nishina K, Akamatsu H, Mikawa K, Shiga M, Maekawa N, Obara H, et al. The inhibitory effects of thiopental, midazolam, and ketamine on human neutrophil functions. Anesth Analg. 1998; 86:159–65.
Article
147. He H, Chen J, Xie WP, Cao S, Hu HY, Yang LQ, et al. Ketamine used as an acesodyne in human breast cancer therapy causes an undesirable side effect, upregulating anti-apoptosis protein Bcl-2 expression. Genet Mol Res. 2013; 12:1907–15.
Article
148. Zhang J, Liu G, Zhang F, Fang H, Zhang D, Liu S, et al. Analysis of postoperative cognitive dysfunction and influencing factors of dexmedetomidine anesthesia in elderly patients with colorectal cancer. Oncol Lett. 2019; 18:3058–64.
Article
149. Liu M, Yi Y, Zhao M. Effect of dexmedetomidine anesthesia on perioperative levels of TNF-alpha and IL-6 in patients with ovarian cancer. Oncol Lett. 2019; 17:5517–22.
150. Huyan T, Hu X, Peng H, Zhu Z, Li Q, Zhang W. Perioperative dexmedetomidine reduces delirium in elderly patients after lung cancer surgery. Psychiatr Danub. 2019; 31:95–101.
Article
151. Kim JA, Ahn HJ, Yang M, Lee SH, Jeong H, Seong BG. Intraoperative use of dexmedetomidine for the prevention of emergence agitation and postoperative delirium in thoracic surgery: a randomized-controlled trial. Can J Anaesth. 2019; 66:371–9.
Article
152. Lavon H, Matzner P, Benbenishty A, Sorski L, Rossene E, Haldar R, et al. Dexmedetomidine promotes metastasis in rodent models of breast, lung, and colon cancers. Br J Anaesth. 2018; 120:188–96.
Article
153. Yi XL, Wang JT, Chu CQ, Li YX, Yin JH, Liu SL. Cardiocerebral protective effects of dexmedetomidine as anesthetic in colorectal cancer surgery. Eur Rev Med Pharmacol Sci. 2018; 22:3570–6.
154. Xia M, Ji NN, Duan ML, Tong JH, Xu JG, Zhang YM, et al. Dexmedetomidine regulate the malignancy of breast cancer cells by activating alpha2-adrenoceptor/ERK signaling pathway. Eur Rev Med Pharmacol Sci. 2016; 20:3500–6.
155. Forget P, Berlière M, Poncelet A, Kock MD. Effect of clonidine on oncological outcomes after breast and lung cancer surgery. Br J Anaesth. 2018; 121:103–4.
Article
156. Wall T, Sherwin A, Ma D, Buggy DJ. Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: a narrative review. Br J Anaesth. 2019; 123:135–50.
Article
157. Weibel S, Jelting Y, Pace NL, Helf A, Eberhart LH, Hahnenkamp K, et al. Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery in adults. Cochrane Database Syst Rev. 2018; 6:CD009642.
Article
158. Masic D, Liang E, Long C, Sterk EJ, Barbas B, Rech MA. Intravenous lidocaine for acute pain: a systematic review. Pharmacotherapy. 2018; 38:1250–9.
Article
159. Khan JS, Hodgson N, Choi S, Reid S, Paul JE, Hong NJL, et al. Perioperative pregabalin and intraoperative lidocaine infusion to reduce persistent neuropathic pain after breast cancer surgery: A multicenter, factorial, randomized, controlled pilot trial. J Pain. 2019; 20:980–93.
Article
160. Lee JT, Sanderson CR, Xuan W, Agar M. Lidocaine for cancer pain in adults: a systematic review and meta-analysis. J Palliat Med. 2019; 22:326–34.
Article
161. Hermanns H, Hollmann MW, Stevens MF, Lirk P, Brandenburger T, Piegeler T, et al. Molecular mechanisms of action of systemic lidocaine in acute and chronic pain: a narrative review. Br J Anaesth. 2019; 123:335–49.
Article
162. Galoș EV, Tat TF, Popa R, Efrimescu CI, Finnerty D, Buggy DJ, et al. Neutrophil extracellular trapping and angiogenesis biomarkers after intravenous or inhalation anaesthesia with or without intravenous lidocaine for breast cancer surgery: a prospective, randomised trial. Br J Anaesth. 2020; 125:712–21.
Article
163. Van Haren F, Van den Heuvel S, Radema S, Van Erp N, Van den Bersselaar L, Vissers K, et al. Intravenous lidocaine affects oxaliplatin pharmacokinetics in simultaneous infusion. J Oncol Pharm Pract. 2020; 26:1850–6.
Article
164. Chamaraux-Tran TN, Piegeler T. The amide local anesthetic lidocaine in cancer surgery-potential antimetastatic effects and preservation of immune cell function? A narrative review. Front Med (Lausanne). 2017; 4:235.
Article
165. Soto G, Gonzalez MN, Calero F. Intravenous lidocaine infusion. Rev Esp Anestesiol Reanim (Engl Ed). 2018; 65:269–74.
Article
166. Piegeler T, Schläpfer M, Dull RO, Schwartz DE, Borgeat A, Minshall RD, et al. Clinically relevant concentrations of lidocaine and ropivacaine inhibit TNFalpha-induced invasion of lung adenocarcinoma cells in vitro by blocking the activation of Akt and focal adhesion kinase. Br J Anaesth. 2015; 115:784–91.
167. Chamaraux-Tran TN, Mathelin C, Aprahamian M, Joshi GP, Tomasetto C, Diemunsch P, et al. Antitumor effects of lidocaine on human breast cancer cells: an in vitro and in vivo experimental trial. Anticancer Res. 2018; 38:95–105.
168. Johnson MZ, Crowley PD, Foley AG, Xue C, Connolly C, Gallagher HC, et al. Effect of perioperative lidocaine on metastasis after sevoflurane or ketamine-xylazine anaesthesia for breast tumour resection in a murine model. Br J Anaesth. 2018; 121:76–85.
Article
169. Wall TP, Crowley PD, Buggy DJ. The effect of lidocaine and bosutinib on 4T1 murine breast cancer cell behaviour in vitro. Anticancer Res. 2021; 41:2835–40.
Article
170. Wall TP, Crowley PD, Sherwin A, Foley AG, Buggy DJ. Effects of lidocaine and Src inhibition on metastasis in a murine model of breast cancer surgery. Cancers (Basel). 2019; 11:1414.
Article
171. Chong PH, Yeo ZZ. Parenteral lidocaine for complex cancer pain in the home or inpatient hospice setting: a review and synthesis of the evidence. J Palliat Med. 2021; 24:1154–60.
Article
172. Zhang H, Yang L, Zhu X, Zhu M, Sun Z, Cata JP, et al. Association between intraoperative intravenous lidocaine infusion and survival in patients undergoing pancreatectomy for pancreatic cancer: a retrospective study. Br J Anaesth. 2020; 125:141–8.
Article
173. Ji W, Zhang X, Sun G, Wang X, Liu J, Bian J, et al. Effect of perioperative intravenous lidocaine on postoperative outcomes in patients undergoing resection of colorectal cancer: a protocol for systematic review and meta-analysis. BMJ Open. 2021; 11:e048803.
Article
174. Hou YH, Shi WC, Cai S, Liu H, Zheng Z, Qi FW, et al. Effect of intravenous lidocaine on serum interleukin-17 after video-assisted thoracic surgery for non-small-cell lung cancer: a randomized, double-blind, placebo-controlled trial. Drug Des Devel Ther. 2021; 15:3379–90.
Article
175. Cazenave L, Faucher M, Tourret M, Marques M, Tezier M, Mokart D. Intravenous lidocaine and cancer outcomes after radical cystectomy. Eur J Anaesthesiol. 2022; 39:396–9.
Article
176. Wall TP, Buggy DJ. Perioperative intravenous lidocaine and metastatic cancer recurrence - a narrative review. Front Oncol. 2021; 11:688896.
Article
177. Gündisch s, Boeckeler E, Behrends U, Amtmann E, Ehrhardt H, Jeremias I. Glucocorticoids augment survival and proliferation of tumor cells. Anticancer Res. 2012; 32:4251–61.
178. De Oliveira GS Jr, McCarthy R, Turan A, Schink JC, Fitzgerald PC, Sessler DI. Is dexamethasone associated with recurrence of ovarian cancer? Anticancer Res. 2014; 118:1213–8.
Article
179. Merk BA, Havrilesky LJ, Ehrisman JA, Broadwater G, Habib AS. Impact of postoperative nausea and vomiting prophylaxis with dexamethasone on the risk of recurrence of endometrial cancer. Curr Med Res Opin. 2016; 32:453–8.
Article
180. Huang WW, Zhu WZ, Mu DL, Ji XQ, Nie XL, Li XY, et al. Perioperative management may improve long-term survival in patients after lung cancer surgery: a retrospective cohort study. Anesth Analg. 2018; 126:1666–74.
Article
181. Yu HC, Luo YX, Peng H, Kang L, Huang MJ, Wang JP. Avoiding perioperative dexamethasone may improve the outcome of patients with rectal cancer. Eur J Surg Oncol. 2015; 41:667–73.
Article
182. Forget P, Bentin C, Machiels JP, Berliere M, Coulie PG, Kock MD. Intraoperative use of ketorolac or diclofenac is associated with improved disease-free survival and overall survival in conservative breast cancer surgery. Br J Anaesth. 2014; 113 Suppl 1:i82–7.
Article
183. Forget P, Vandenhende J, Berliere M, Machiels JP, Nussbaum B, Legrand C, et al. Do intraoperative analgesics influence breast cancer recurrence after mastectomy? A retrospective analysis. Anesth Analg. 2010; 110:1630–5.
Article
184. Yeh CC, Lin JT, Jeng LB, Ho HJ, Yang HR, Wu MS, et al. Nonsteroidal anti-inflammatory drugs are associated with reduced risk of early hepatocellular carcinoma recurrence after curative liver resection: a nationwide cohort study. Ann Surg. 2015; 261:521–6.
185. Shaashua L, Shabat-Simon M, Haldar R, Matzner P, Zmora O, Shabtai M, et al. Perioperative COX-2 and β-adrenergic blockade improves metastatic biomarkers in breast cancer patients in a phase-II randomized trial. Clin Cancer Res. 2017; 23:4651–61.
Article
Full Text Links
  • APM
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