Article Data

  • Views 583
  • Dowloads 120

Reviews

Open Access

Effects of anesthesia and other perioperative factors on immune function: a narrative review

  • Yining Chen1,†
  • Junwei Kang1,†
  • Yulang Wang2
  • Xiongxiong Pan1,*,

1Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China

2Department of Anesthesiology, the Affiliated Hospital of Xuzhou Medical University, 221002 Xuzhou, Jiangsu, China

DOI: 10.22514/sv.2024.105 Vol.20,Issue 9,September 2024 pp.1-15

Submitted: 01 January 2024 Accepted: 08 May 2024

Published: 08 September 2024

*Corresponding Author(s): Xiongxiong Pan E-mail: xiongxiong@njmu.edu.cn

† These authors contributed equally.

Abstract

This study investigates the impact of perioperative factors on patients’ immune functions including physiological condition, surgical strategy, anesthesia methodology, intraoperative usage of anesthetics, blood transfusion, intraoperative body temperature, and inspired oxygen concentration. Results demonstrate that the individual perioperative factors have varying degree of impact, while their combined effect may cause changes in patients’ immune function. The study outcomes suggest that patient’s immune function must be assessed prior to the surgery based on individual characteristics. The surgical and anesthetic methods are accordingly selected. During the surgery, prudent usage and monitoring of factors like anesthesia techniques, anesthetics, inhaled oxygen concentration, blood transfusion, and patient’s body temperature are vital for minimizing the immune system disruptions. These findings practically guide regarding the optimization of perioperative care strategies and mitigation of immunosuppression risks.


Keywords

Immune function; Perioperative factors; Anesthesia


Cite and Share

Yining Chen,Junwei Kang,Yulang Wang,Xiongxiong Pan. Effects of anesthesia and other perioperative factors on immune function: a narrative review. Signa Vitae. 2024. 20(9);1-15.

References

[1] Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, et al. An aged immune system drives senescence and ageing of solid organs. Nature. 2021; 594: 100–105.

[2] De Maeyer RPH, Chambers ES. The impact of ageing on monocytes and macrophages. Immunology Letters. 2021; 230: 1–10.

[3] Nauseef WM. How human neutrophils kill and degrade microbes: an integrated view. Immunological Reviews. 2007; 219: 88–102.

[4] Van Avondt K, Strecker J, Tulotta C, Minnerup J, Schulz C, Soehnlein O. Neutrophils in aging and aging-related pathologies. Immunological Reviews. 2023; 314: 357–375.

[5] Fulop T, Larbi A, Douziech N, Fortin C, Guérard K, Lesur O, et al. Signal transduction and functional changes in neutrophils with aging. Aging Cell. 2004; 3: 217–226.

[6] Tortorella C, Simone O, Piazzolla G, Stella I, Antonaci S. Age-related impairment of GM-CSF-induced signalling in neutrophils: role of SHP-1 and SOCS proteins. Ageing Research Reviews. 2007; 6: 81–93.

[7] Pilkington SM, Bulfone-Paus S, Griffiths CEM, Watson REB. Inflammaging and the skin. Journal of Investigative Dermatology. 2021; 141: 1087–1095.

[8] Cui Q, Li W, Wang D, Wang S, Liu A, Zhang G, et al. Immune signature and phagocytosis of circulating DC subsets in healthy adults during aging. International Immunopharmacology. 2024; 130: 111715.

[9] Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. The Journal of Immunology. 2010; 184: 2518–2527.

[10] Agrawal A, Agrawal S, Cao J, Su H, Osann K, Gupta S. Altered innate immune functioning of dendritic cells in elderly humans: a role of phosphoinositide 3-kinase-signaling pathway. The Journal of Immunology. 2007; 178: 6912–6922.

[11] Brauning A, Rae M, Zhu G, Fulton E, Admasu TD, Stolzing A, et al. Aging of the immune system: focus on natural killer cells phenotype and functions. Cells. 2022; 11: 1017.

[12] Beli E, Clinthorne JF, Duriancik DM, Hwang I, Kim S, Gardner EM. Natural killer cell function is altered during the primary response of aged mice to influenza infection. Mechanisms of Ageing and Development. 2011; 132: 503–510.

[13] Borrego F, Alonso MC, Galiani MD, Carracedo J, Ramirez R, Ostos B, et al. NK phenotypic markers and IL2 response in NK cells from elderly people. Experimental Gerontology. 1999; 34: 253–265.

[14] Castelo-Branco C, Soveral I. The immune system and aging: a review. Gynecological Endocrinology. 2014; 30: 16–22.

[15] Hirokawa K, Utsuyama M, Kasai M, Kurashima C. Aging and immunity. Acta Pathologica Japonica. 1992; 42: 537–548.

[16] Mahbub S, Brubaker AL, Kovacs EJ. Aging of the innate immune system: an update. Current Immunology Reviews. 2011; 7: 104–115.

[17] Pawelec G, Barnett Y, Forsey R, Frasca D, Globerson A, McLeod J, et al. T cells and aging, January 2002 update. Frontiers in Bioscience. 2002; 7: d1056–d11183.

[18] Effros RB, Cai Z, Linton PJ. CD8 T cells and aging. Critical Reviews in Immunology. 2003; 23: 45–64.

[19] Sadighi Akha AA, Miller RA. Signal transduction in the aging immune system. Current Opinion in Immunology. 2005; 17: 486–491.

[20] Chong Y, Ikematsu H, Yamaji K, Nishimura M, Nabeshima S, Kashiwagi S, et al. CD27+ (memory) B cell decrease and apoptosis-resistant CD27− (naive) B cell increase in aged humans: implications for age-related peripheral B cell developmental disturbances. International Immunology. 2005; 17: 383–390.

[21] Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng NY, et al. Limited efficacy of inactivated influenza vaccine in elderly individuals is associated with decreased production of vaccine-specific antibodies. Journal of Clinical Investigation. 2011; 121: 3109–3119.

[22] Ruiz-Cortes K, Villageliu DN, Samuelson DR. Innate lymphocytes: role in alcohol-induced immune dysfunction. Frontiers in Immunology. 2022; 13: 934617.

[23] Malherbe DC, Messaoudi I. Transcriptional and epigenetic regulation of monocyte and macrophage dysfunction by chronic alcohol consumption. Frontiers in Immunology. 2022; 13: 911951.

[24] Cao L, Wu D, Qin L, Tan D, Fan Q, Jia X, et al. Single-cell RNA transcriptome profiling of liver cells of short-term alcoholic liver injury in mice. International Journal of Molecular Sciences. 2023; 24: 4344.

[25] Malacco NLSO, Souza JAM, Martins FRB, Rachid MA, Simplicio JA, Tirapelli CR, et al. Chronic ethanol consumption compromises neutrophil function in acute pulmonary Aspergillus fumigatus infection. eLife. 2020; 9: e58855.

[26] Gacouin A, Roussel M, Le Priol J, Azzaoui I, Uhel F, Fest T, et al. Acute alcohol exposure has an independent impact on C-reactive protein levels, neutrophil CD64 expression, and subsets of circulating white blood cells differentiated by flow cytometry in nontrauma patients. Shock. 2014; 42: 192–198.

[27] Rehman S, Chandel N, Salhan D, Rai P, Sharma B, Singh T, et al. Ethanol and vitamin D receptor in T cell apoptosis. Journal of Neuroimmune Pharmacology. 2013; 8: 251–261.

[28] Wang H, Zhou H, Mahler S, Chervenak R, Wolcott M. Alcohol affects the late differentiation of progenitor B cells. Alcohol and Alcoholism. 2011; 46: 26–32.

[29] Verma S, Alexander CM, Carlson MJ, Tygrett LT, Waldschmidt TJ. B-cell studies in chronic ethanol mice. Methods in Molecular Biology. 2008; 447: 295–323.

[30] Pohl K, Moodley P, Dhanda AD. Alcohol’s impact on the gut and liver. Nutrients. 2021; 13: 3170.

[31] Watzl B, Watson RR. Role of alcohol abuse in nutritional immunosuppression. Journal of Nutrition. 1992; 122: 733–737.

[32] Shi X, DeLucia AL, Bao J, Zhang P. Alcohol abuse and disorder of granulopoiesis. Pharmacology & Therapeutics. 2019; 198: 206–219.

[33] Dukić M, Radonjić T, Jovanović I, Zdravković M, Todorović Z, Kraišnik N, et al. Alcohol, inflammation, and microbiota in alcoholic liver disease. International Journal of Molecular Sciences. 2023; 24: 3735.

[34] Georgakouli K, Manthou E, Fatouros IG, Deli CK, Koutedakis Y, Theodorakis Y, et al. HPA-axis activity and nutritional status correlation in individuals with alcohol use disorder. Nutrients. 2022; 14: 4978.

[35] Antar SA, Ashour NA, Sharaky M, Khattab M, Ashour NA, Zaid RT, et al. Diabetes mellitus: classification, mediators, and complications; a gate to identify potential targets for the development of new effective treatments. Biomedicine & Pharmacotherapy. 2023; 168: 115734.

[36] Fitas AL, Martins C, Borrego LM, Lopes L, Jörns A, Lenzen S, et al. Immune cell and cytokine patterns in children with type 1 diabetes mellitus undergoing a remission phase: a longitudinal study. Pediatric Diabetes. 2018; 19: 963–971.

[37] Sun Y, Zhou L, Chen W, Zhang L, Zeng H, Sun Y, et al. Immune metabolism: a bridge of dendritic cells function. International Reviews of Immunology. 2022; 41: 313–325.

[38] Kennedy DM, Skillen AW, Self CH. Glycation of monoclonal antibodies impairs their ability to bind antigen. Clinical and Experimental Immunology. 1994; 98: 245–251.

[39] Richard C, Wadowski M, Goruk S, Cameron L, Sharma AM, Field CJ. Individuals with obesity and type 2 diabetes have additional immune dysfunction compared with obese individuals who are metabolically healthy. BMJ Open Diabetes Research & Care. 2017; 5: e000379.

[40] Martinez PJ, Mathews C, Actor JK, Hwang SA, Brown EL, De Santiago HK, et al. Impaired CD4+ and T-helper 17 cell memory response to streptococcus pneumoniae is associated with elevated glucose and percent glycated hemoglobin A1c in Mexican Americans with type 2 diabetes mellitus. Translational Research. 2014; 163: 53–63.

[41] Moura J, Rodrigues J, Gonçalves M, Amaral C, Lima M, Carvalho E. Impaired T-cell differentiation in diabetic foot ulceration. Cellular & Molecular Immunology. 2017; 14: 758–769.

[42] Bettini M, Bettini ML. Function, failure, and the future potential of Tregs in type 1 diabetes. Diabetes. 2021; 70: 1211–1219.

[43] Galway U, Chahar P, Schmidt MT, Araujo-Duran JA, Shivakumar J, Turan A, et al. Perioperative challenges in management of diabetic patients undergoing non-cardiac surgery. World Journal of Diabetes. 2021; 12: 1255–1266.

[44] van Niekerk G, Christowitz C, Conradie D, Engelbrecht AM. Insulin as an immunomodulatory hormone. Cytokine & Growth Factor Reviews. 2020; 52: 34–44.

[45] Agus MS, Steil GM, Wypij D, Costello JM, Laussen PC, Langer M, et al. Tight glycemic control versus standard care after pediatric cardiac surgery. The New England Journal of Medicine. 2012; 367: 1208–1219.

[46] Vellanki P, Cardona S, Galindo RJ, Urrutia MA, Pasquel FJ, Davis GM, et al. Efficacy and safety of intensive versus nonintensive supplemental insulin with a basal-bolus insulin regimen in hospitalized patients with type 2 diabetes: a randomized clinical study. Diabetes Care. 2022; 45: 2217–2223.

[47] Yao M, Hao Y, Wang T, Xie M, Li H, Feng J, et al. A review of stress-induced hyperglycaemia in the context of acute ischaemic stroke: definition, underlying mechanisms, and the status of insulin therapy. Frontiers in Neurology. 2023; 14: 1149671.

[48] Cao SG, Ren JA, Shen B, Chen D, Zhou YB, Li JS. Intensive versus conventional insulin therapy in type 2 diabetes patients undergoing D2 gastrectomy for gastric cancer: a randomized controlled trial. World Journal of Surgery. 2011; 35: 85–92.

[49] Bhamidipati CM, LaPar DJ, Stukenborg GJ, Morrison CC, Kern JA, Kron IL, et al. Superiority of moderate control of hyperglycemia to tight control in patients undergoing coronary artery bypass grafting. The Journal of Thoracic and Cardiovascular Surgery. 2011; 141: 543–551.

[50] Jasinski-Bergner S, Radetzki AL, Jahn J, Wohlrab D, Kielstein H. Impact of the body mass index on perioperative immunological disturbances in patients with hip and knee arthroplasty. Journal of Orthopaedic Surgery and Research. 2017; 12: 58.

[51] Vrieling F, Stienstra R. Obesity and dysregulated innate immune responses: impact of micronutrient deficiencies. Trends in Immunology. 2023; 44: 217–230.

[52] SantaCruz-Calvo S, Bharath L, Pugh G, SantaCruz-Calvo L, Lenin RR, Lutshumba J, et al. Adaptive immune cells shape obesity-associated type 2 diabetes mellitus and less prominent comorbidities. Nature Reviews Endocrinology. 2022; 18: 23–42.

[53] Schleh MW, Caslin HL, Garcia JN, Mashayekhi M, Srivastava G, Bradley AB, et al. Metaflammation in obesity and its therapeutic targeting. Science Translational Medicine. 2023; 15: eadf9382.

[54] Hata M, Andriessen EMMA, Hata M, Diaz-Marin R, Fournier F, Crespo-Garcia S, et al. Past history of obesity triggers persistent epigenetic changes in innate immunity and exacerbates neuroinflammation. Science. 2023; 379: 45–62.

[55] Green WD, Beck MA. Obesity altered T cell metabolism and the response to infection. Current Opinion in Immunology. 2017; 46: 1–7.

[56] Oleinika K, Slisere B, Catalán D, Rosser EC. B cell contribution to immunometabolic dysfunction and impaired immune responses in obesity. Clinical and Experimental Immunology. 2022; 210: 263–272.

[57] Touch S, Clément K, André S. T cell populations and functions are altered in human obesity and type 2 diabetes. Current Diabetes Reports. 2017; 17: 81.

[58] Frasca D, Diaz A, Romero M, Blomberg BB. Ageing and obesity similarly impair antibody responses. Clinical and Experimental Immunology. 2017; 187: 64–70.

[59] Huang CJ, Zourdos MC, Jo E, Ormsbee MJ. Influence of physical activity and nutrition on obesity-related immune function. The Scientific World Journal. 2013; 2013: 752071.

[60] Vega-Robledo GB, Rico-Rosillo MG. Adipose tissue: immune function and alterations caused by obesity. Revista alergia Mexico. 2019; 66: 340–353. (In Spanish)

[61] Ait Eldjoudi D, Cordero Barreal A, Gonzalez-Rodríguez M, Ruiz-Fernández C, Farrag Y, Farrag M, et al. Leptin in osteoarthritis and rheumatoid arthritis: player or bystander? International Journal of Molecular Sciences. 2022; 23: 2859.

[62] Nguyen MT, Favelyukis S, Nguyen AK, Reichart D, Scott PA, Jenn A, et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. Journal of Biological Chemistry. 2007; 282: 35279–35292.

[63] Gunst MA, Minei JP. Transfusion of blood products and nosocomial infection in surgical patients. Current Opinion in Critical Care. 2007; 13: 428–432.

[64] Gao Y, Jin H, Tan H, Cai X, Sun Y. Erythrocyte-derived extracellular vesicles aggravate inflammation by promoting the proinflammatory macrophage phenotype through TLR4-MyD88-NF-kB-MAPK pathway. Journal of Leukocyte Biology. 2022; 112: 693–706.

[65] Strumper-Groves D. Perioperative blood transfusion and outcome. Current Opinion in Anesthesiology. 2006; 19: 198–206.

[66] Guo JR, Xu F, Jin XJ, Shen HC, Liu Y, Zhang YW, et al. Impact of allogenic and autologous transfusion on immune function in patients with tumors. Asian Pacific Journal of Cancer Prevention. 2014; 15: 467–474.

[67] Han ZZ, Li M, Zhang Y. The effects of allogeneic and autologous blood transfusion on immune function in patients receiving total hip replacement. American Journal of Translational Research. 2023; 15: 4709–4717.

[68] Gong Y, Tang Y, Xue Y, Chen L. Impact of intraoperative allogenic and autologous transfusion on immune function and prognosis in patients with hepatocellular carcinoma. Medicine. 2020; 99: e22568.

[69] Lan X, Chen Y, Bi Q, Xu W, Huang J. Effects of storage duration of suspended red blood cells before intraoperative infusion on coagulation indexes, routine blood examination and immune function in patients with gastrointestinal tumors. Pakistan Journal of Medical Sciences. 2023; 39: 182–187.

[70] Muszynski JA, Frazier E, Nofziger R, Nateri J, Hanson-Huber L, Steele L, et al; Pediatric Critical Care Blood Research Network (Blood Net) subgroup of the Pediatric Acute Lung Injury and Sepsis Investigators (PALISI). Red blood cell transfusion and immune function in critically ill children: a prospective observational study. Transfusion. 2015; 55: 766–774.

[71] Aguilar-Nascimento JE, Zampieri-Filho JP, Bordin JO. Implications of perioperative allogeneic red blood cell transfusion on the immune-inflammatory response. Hematology, Transfusion and Cell Therapy. 2021; 43: 58–64.

[72] Qiu L, Wang DR, Zhang XY, Gao S, Li XX, Sun GP, et al. Impact of perioperative blood transfusion on immune function and prognosis in colorectal cancer patients. Transfusion and Apheresis Science. 2016; 54: 235–241.

[73] Shi J, Gao B, Yang Y, Yang L, Li X. Influence of perioperative leukodeplated red blood cell transfusion on immune function of patients with bladder cancer. Chinese Journal of Cellular and Molecular Immunology. 2018; 34: 632–636. (In Chinese)

[74] Abe T, Amano H, Hanada K, Minami T, Yonehara S, Hattori M, et al. Perioperative red blood cell transfusion is associated with poor long-term survival in pancreatic adenocarcinoma. Anticancer Research. 2017; 37: 5863–5870.

[75] De Oliveira GS Jr, Schink JC, Buoy C, Ahmad S, Fitzgerald PC, McCarthy RJ. The association between allogeneic perioperative blood transfusion on tumour recurrence and survival in patients with advanced ovarian cancer. Transfusion Medicine. 2012; 22: 97–103.

[76] Sessler DI. Temperature monitoring and perioperative thermoregulation. Anesthesiology. 2008; 109: 318–338.

[77] Yang C, Cheng Y, Liu S, Huang S, Yu X. Effect of preoperative oral carbohydrate loading on body temperature during combined spinal-epidural anesthesia for elective cesarean delivery. Anesthesia & Analgesia. 2021; 133: 731–738.

[78] Kaneko S, Hara K, Sato S, Nakashima T, Kawazoe Y, Taguchi M, et al. Association between preoperative toe perfusion index and maternal core temperature decrease during cesarean delivery under spinal anesthesia: a prospective cohort study. BMC Anesthesiology. 2021; 21: 250.

[79] Chen J, Li H, Xu P, Tang Y, Su S, Liu G, et al. Hypothermia-mediated apoptosis and inflammation contribute to antioxidant and immune adaption in freshwater drum, aplodinotus grunniens. Antioxidants. 2022; 11: 1657.

[80] Qadan M, Gardner SA, Vitale DS, Lominadze D, Joshua IG, Polk HC Jr. Hypothermia and surgery: immunologic mechanisms for current practice. Annals of Surgery. 2009; 250: 134–140.

[81] Marek-Trzonkowska N, Piekarska K, Filipowicz N, Piotrowski A, Gucwa M, Vogt K, et al. Mild hypothermia provides Treg stability. Scientific Reports. 2017; 7: 11915.

[82] Oldman AH, Martin DS, Feelisch M, Grocott MPW, Cumpstey AF. Effects of perioperative oxygen concentration on oxidative stress in adult surgical patients: a systematic review. British Journal of Anaesthesia. 2021; 126: 622–632.

[83] Mattishent K, Thavarajah M, Sinha A, Peel A, Egger M, Solomkin J, et al. Safety of 80% vs 30–35% fraction of inspired oxygen in patients undergoing surgery: a systematic review and meta-analysis. British Journal of Anaesthesia. 2019; 122: 311–324.

[84] Allegranzi B, Zayed B, Bischoff P, Kubilay NZ, de Jonge S, de Vries F, et al. New WHO recommendations on intraoperative and postoperative measures for surgical site infection prevention: an evidence-based global perspective. The Lancet Infectious Diseases. 2016; 16: e288–e303.

[85] Schietroma M, Colozzi S, Pessia B, Carlei F, Di Furia M, Amicucci G. Laparoscopic Nissen fundoplication: the effects of high-concentration supplemental perioperative oxygen on the inflammatory and immune response: a randomised controlled trial. Journal of Minimal Access Surgery. 2018; 14: 221–229.

[86] Schietroma M, Colozzi S, Pessia B, Carlei F, Amicucci G. The effects of high-concentration oxygen on inflammatory markers in laparoscopic cholecystectomy: a randomized controlled trial. Surgical Laparoscopy Endoscopy & Percutaneous Techniques. 2017; 27: 83–89.

[87] Huang C, Huang R, Jiang T, Huang K, Cao J, Qiu Z. Laparoscopic and open resection for colorectal cancer: an evaluation of cellular immunity. BMC Gastroenterology. 2010; 10: 127.

[88] Ypsilantis P, Lambropoulou M, Anagnostopoulos K, Kiroplastis K, Tepelopoulos G, Bangeas P, et al. Gut-barrier disruption after laparoscopic versus open major liver resection in the rat. Surgery. 2022; 171: 973–979.

[89] Martínez-Martínez AB, Arbonés-Mainar JM. Colorectal cancer: immune response in laparoscopic versus open colorectal surgery. Cirugia y Cirujanos. 2022; 90: 295–302.

[90] Yang X, Cheng Y, Cheng N, Gong J, Bai L, Zhao L, et al. Gases for establishing pneumoperitoneum during laparoscopic abdominal surgery. Cochrane Database of Systematic Reviews. 2022; 3: CD009569.

[91] Neuhaus SJ, Watson DI. Pneumoperitoneum and peritoneal surface changes: a review. Surgical Endoscopy. 2004; 18: 1316–1322.

[92] Kusano T, Etoh T, Inomata M, Shiraishi N, Kitano S. CO2 pneumoperitoneum increases secretory IgA levels in the gut compared with laparotomy in an experimental animal model. Surgical Endoscopy. 2014; 28: 1879–1885.

[93] Albers KI, Polat F, Helder L, Panhuizen IF, Snoeck MMJ, Polle SBW, et al. Quality of recovery and innate immune homeostasis in patients undergoing low-pressure versus standard-pressure pneumoperitoneum during laparoscopic colorectal surgery (RECOVER): a randomized controlled trial. Annals of Surgery. 2022; 276: e664–e673.

[94] Schietroma M, Pessia B, Stifini D, Lancione L, Carlei F, Cecilia EM, et al. Effects of low and standard intra-abdominal pressure on systemic inflammation and immune response in laparoscopic adrenalectomy: a prospective randomised study. Journal of Minimal Access Surgery. 2016; 12: 109–117.

[95] Konstantis G, Tsaousi G, Kitsikidou E, Zacharoulis D, Pourzitaki C. The immunomodulatory effect of various anaesthetic practices in patients undergoing gastric or colon cancer surgery: a systematic review and meta-analysis of randomized clinical trials. Journal of Clinical Medicine. 2023; 12: 6027.

[96] Imani Rastabi H, Khosravi M, Avizeh R, Moslemi M. Evaluation of the effect of lidocaine epidural injection on immunological indices in dogs under total intravenous anesthesia submitted to ovariohysterectomy. PLOS ONE. 2021; 16: e0253731.

[97] Wang L, Liang S, Chen H, Xu Y, Wang Y. The effects of epidural anaesthesia and analgesia on T lymphocytes differentiation markers and cytokines in patients after gastric cancer resection. BMC Anesthesiology. 2019; 19: 102.

[98] Cong X, Huang Z, Zhang L, Sun M, Chang E, Zhang W, et al. Effect of different anaesthesia methods on perioperative cellular immune function and long-term outcome in patients undergoing radical resection of esophageal cancer: a prospective cohort study. American Journal of Translational Research. 2021; 13: 11427–11438.

[99] Hu C, Zhang S, Chen Q, Wang R. Effects of different anesthetic and analgesic methods on cellular immune function and stress hormone levels in patients undergoing esophageal cancer surgery. Journal of Healthcare Engineering. 2022; 2022: 4752609.

[100] Ezhevskaia AA, Prusakova ZhB, Maksimova LP, Sholkina MN, Balmusova EA, Ovechkin AM. Effects of epidural anesthesia on stress-induced immune supression during major corrective spine surgery. Anesteziol Reanimatol. 2014; 59: 4–9. (In Russian)

[101] Kurosawa S, Kato M. Anesthetics, immune cells, and immune responses. Journal of Anesthesia. 2008; 22: 263–277.

[102] Lattermann R, Schricker T, Wachter U, Georgieff M, Goertz A. Understanding the mechanisms by which isoflurane modifies the hyperglycemic response to surgery. Anesthesia & Analgesia. 2001; 93: 121–127.

[103] Buddeberg BS, Seeberger MD. Anesthesia and oncology: friend or foe? Frontiers in Oncology. 2022; 12: 802210.

[104] Vrbanović Mijatović V, Gatin L, Tonković D, Bandić Pavlović D, Smuđ Orehovec S, Miklić Bublić M, et al. The effect of regional vs. general anesthesia on the immune response in breast cancer surgery: a narrative review of the literature. Acta Clinica Croatica. 2022; 61: 115–120.

[105] Eisenstein TK. The role of opioid receptors in immune system function. Frontiers in Immunology. 2019; 10: 2904.

[106] Lin M, Deng K, Li Y, Wan J. Morphine enhances LPS-induced macrophage apoptosis through a PPARgamma-dependent mechanism. Experimental and Therapeutic Medicine. 2021; 22: 714.

[107] Luan G, Pan F, Bu L, Wu K, Wang A, Xu X. Butorphanol promotes macrophage phenotypic transition to inhibit inflammatory lung injury via kappa receptors. Frontiers in Immunology. 2021; 12: 692286.

[108] Lambert DG. Opioids and opioid receptors; understanding pharmacological mechanisms as a key to therapeutic advances and mitigation of the misuse crisis. BJA Open. 2023; 6: 100141.

[109] Sun Q, Li Z, Wang Z, Wang Q, Qin F, Pan H, et al. Immunosuppression by opioids: mechanisms of action on innate and adaptive immunity. Biochemical Pharmacology. 2023; 209: 115417.

[110] Gao M, Sun J, Jin W, Qian Y. Morphine, but not ketamine, decreases the ratio of Th1/Th2 in CD4-positive cells through T-bet and GATA3. Inflammation. 2012; 35: 1069–1077.

[111] Franchi S, Moretti S, Castelli M, Lattuada D, Scavullo C, Panerai AE, et al. Mu opioid receptor activation modulates Toll like receptor 4 in murine macrophages. Brain, Behavior, and Immunity. 2012; 26: 480–488.

[112] Jalodia R, Kolli U, Braniff RG, Tao J, Abu YF, Chupikova I, et al. Morphine mediated neutrophil infiltration in intestinal tissue play essential role in histological damage and microbial dysbiosis. Gut Microbes. 2022; 14: 2143225.

[113] Plein LM, Rittner HL. Opioids and the immune system—friend or foe. British Journal of Pharmacology. 2018; 175: 2717–2725.

[114] Li Z, Sun Q, Liu Q, Mu X, Wang H, Zhang H, et al. Compound 511 ameliorates MRSA-induced lung injury by attenuating morphine-induced immunosuppression in mice via PI3K/AKT/mTOR pathway. Phytomedicine. 2023; 108: 154475.

[115] Martucci C, Panerai AE, Sacerdote P. Chronic fentanyl or buprenorphine infusion in the mouse: similar analgesic profile but different effects on immune responses. Pain. 2004; 110: 385–392.

[116] Shavit Y, Ben-Eliyahu S, Zeidel A, Beilin B. Effects of fentanyl on natural killer cell activity and on resistance to tumor metastasis in rats. Dose and timing study. Neuroimmunomodulation. 2004; 11: 255–260.

[117] Gong L, Qin Q, Zhou L, Ouyang W, Li Y, Wu Y, et al. Effects of fentanyl anesthesia and sufentanil anesthesia on regulatory T cells frequencies. International Journal of Clinical and Experimental Pathology. 2014; 7: 7708–7716.

[118] Qi Y, Yao X, Zhang B, Du X. Comparison of recovery effect for sufentanil and remifentanil anesthesia with TCI in laparoscopic radical resection during colorectal cancer. Oncology Letters. 2016; 11: 3361–3365.

[119] Tsuboi I, Tanaka H, Nakao M, Shichijo S, Itoh K. Nonsteroidal anti-inflammatory drugs differentially regulate cytokine production in human lymphocytes: up-regulation of TNF, IFN-g and IL-2, in contrast to down-regulation of IL-6 production. Cytokine. 1995; 7: 372–379.

[120] Zhang Y, Du Z, Zhou Q, Wang Y, Li J. Remifentanil attenuates lipopolysaccharide-induced acute lung injury by downregulating the NF-kB signaling pathway. Inflammation. 2014; 37: 1654–1660.

[121] Cronin AJ, Aucutt-Walter NM, Budinetz T, Bonafide CP, DiVittore NA, Gordin V, et al. Low-dose remifentanil infusion does not impair natural killer cell function in healthy volunteers. British Journal of Anaesthesia. 2003; 91: 805–809.

[122] Boland JW, Foulds GA, Ahmedzai SH, Pockley AG. A preliminary evaluation of the effects of opioids on innate and adaptive human in vitro immune function. BMJ Supportive & Palliative Care. 2014; 4: 357–367.

[123] Filipczak-Bryniarska I, Nazimek K, Nowak B, Kozlowski M, Wąsik M, Bryniarski K. In contrast to morphine, buprenorphine enhances macrophage-induced humoral immunity and, as oxycodone, slightly suppresses the effector phase of cell-mediated immune response in mice. International Immunopharmacology. 2018; 54: 344–353.

[124] Saeed I, La Caze A, Hollmann MW, Shaw PN, Parat MO. New insights on tramadol and immunomodulation. Current Oncology Reports. 2021; 23: 123.

[125] Bradley A, Boland JW. Effects of opioids on immune and endocrine function in patients with cancer pain. Current Treatment Options in Oncology. 2023; 24: 867–879.

[126] Hou LW, Ding HL, Li MQ, Jin S, Wang XS, Ji LJ. Effect of tramadol on perioperative immune function in patients undergoing gastric cancer surgeries. Anesthesia Essays and Researches. 2013; 7: 54–57.

[127] Lin Y, Miao Z, Wu Y, Ge FF, Wen QP. Effect of low dose naloxone on the immune system function of a patient undergoing video-assisted thoracoscopic resection of lung cancer with sufentanil controlled analgesia—a randomized controlled trial. BMC Anesthesiology. 2019; 19: 236.

[128] 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. International Immunopharmacology. 2020; 83: 106388.

[129] Nelson CJ, Carrigan KA, Lysle DT. Naltrexone administration attenuates surgery-induced immune alterations in rats. Journal of Surgical Research. 2000; 94: 172–177.

[130] Toleska M, Dimitrovski A, Dimitrovska NT. Comparation among opioid-based, low opioid and opioid free anesthesia in colorectal oncologic surgery. Pril (Makedon Akad Nauk Umet Odd Med Nauki). 2023; 44: 117–126.

[131] Qu N, Meng Y, Handley MK, Wang C, Shan F. Preclinical and clinical studies into the bioactivity of low-dose naltrexone (LDN) for oncotherapy. International Immunopharmacology. 2021; 96: 107714.

[132] Abrahams D, Ibrahim-Hashim A, Ackerman RS, Brown JS, Whelan CJ, Garfinkel MB, et al. Immunomodulatory and pro-oncologic effects of ketamine and isoflurane anesthetics in a murine model. PLOS ONE. 2023; 18: e0292492.

[133] Inada T, Yamanouchi Y, Jomura S, Sakamoto S, Takahashi M, Kambara T, et al. Effect of propofol and isoflurane anaesthesia on the immune response to surgery. Anaesthesia. 2004; 59: 954–959.

[134] Loop T, Dovi-Akue D, Frick M, Roesslein M, Egger L, Humar M, et al. Volatile anesthetics induce caspase-dependent, mitochondria-mediated apoptosis in human T lymphocytes in vitro. Anesthesiology. 2005; 102: 1147–1157.

[135] Woo JH, Baik HJ, Kim CH, Chung RK, Kim DY, Lee GY, et al. Effect of propofol and desflurane on immune cell populations in breast cancer patients: a randomized trial. Journal of Korean Medical Science. 2015; 30: 1503–1508.

[136] Yang H, Liang G, Hawkins BJ, Madesh M, Pierwola A, Wei H. Inhalational anesthetics induce cell damage by disruption of intracellular calcium homeostasis with different potencies. Anesthesiology. 2008; 109: 243–250.

[137] Odeh D, Oršolić N, Adrović E, Gaćina L, Perić P, Odeh S, et al. Effects of volatile anaesthetics and iron dextran on chronic inflammation and antioxidant defense system in rats. Antioxidants. 2022; 11: 708.

[138] Yuki K, Hou L, Shibamura-Fujiogi M, Koutsogiannaki S, Soriano SG. Mechanistic consideration of the effect of perioperative volatile anesthetics on phagocytes. Clinical Immunology. 2021; 222: 108635.

[139] Zha H, Matsunami E, Blazon-Brown N, Koutsogiannaki S, Hou L, Bu W, et al. Volatile anesthetics affect macrophage phagocytosis. PLOS ONE. 2019; 14: e0216163.

[140] Schneemilch CE, Ittenson A, Ansorge S, Hachenberg T, Bank U. Effect of 2 anesthetic techniques on the postoperative proinflammatory and anti-inflammatory cytokine response and cellular immune function to minor surgery. Journal of Clinical Anesthesia. 2005; 17: 517–527.

[141] Tylman M, Sarbinowski R, Bengtson JP, Kvarnström A, Bengtsson A. Inflammatory response in patients undergoing colorectal cancer surgery: the effect of two different anesthetic techniques. Minerva Anestesiologica. 2011; 77: 275–282.

[142] Kitamura T, Sato K, Kawamura G, Yamada Y. The involvement of adenosine triphosphate-sensitive potassium channels in the different effects of sevoflurane and propofol on glucose metabolism in fed rats. Anesthesia & Analgesia. 2012; 114: 110–116.

[143] Kusunoki M, Hayashi M, Shoji T, Uba T, Tanaka H, Sumi C, et al. Propofol inhibits stromatoxin-1-sensitive voltage-dependent K+ channels in pancreatic beta-cells and enhances insulin secretion. PeerJ. 2019; 7: e8157.

[144] Suzuki K, Sato Y, Kai S, Nishi K, Adachi T, Matsuo Y, et al. Volatile anesthetics suppress glucose-stimulated insulin secretion in MIN6 cells by inhibiting glucose-induced activation of hypoxia-inducible factor 1. PeerJ. 2015; 3: e1498.

[145] Chen MS, Yang KS, Lin WC, Fang CL, Chen HF, Sheu SM. Lipofundin mediates major inhibition of intravenous propofol on phorbol myristate acetate and Escherichia coli-induced neutrophil extracellular traps. Molecular Biology Reports. 2022; 49: 6517–6529.

[146] Yi S, Tao X, Wang Y, Cao Q, Zhou Z, Wang S. Effects of propofol on macrophage activation and function in diseases. Frontiers in Pharmacology. 2022; 13: 964771.

[147] Chang MC, Chen YL, Chiang YC, Cheng YJ, Jen YW, Chen CA, et al. Anti-CD40 antibody and toll-like receptor 3 ligand restore dendritic cell-mediated anti-tumor immunity suppressed by morphine. American Journal of Cancer Research. 2016; 6: 157–172.

[148] Tazawa K, Koutsogiannaki S, Chamberlain M, Yuki K. The effect of different anesthetics on tumor cytotoxicity by natural killer cells. Toxicology Letters. 2017; 266: 23–31.

[149] Zhou M, Liu W, Peng J, Wang Y. Impact of propofol epidural anesthesia on immune function and inflammatory factors in patients undergoing gastric cancer surgery. American Journal of Translational Research. 2021; 13: 3064–3073.

[150] Kushida A, Inada T, Shingu K. Enhancement of antitumor immunity after propofol treatment in mice. Immunopharmacology and Immunotoxicology. 2007; 29: 477–486.

[151] Yu H, Chen L, Yue CJ, Xu H, Cheng J, Cornett EM, et al. Effects of propofol and sevoflurane on T-cell immune function and Th cell differentiation in children with SMPP undergoing fibreoptic bronchoscopy. Annals of Medicine. 2002; 54: 2574–2580.

[152] Li LC, Tian Y, Xiao J, Yang Y, Wu JN, Chen Y, et al. Dexmedetomidine promotes inflammation resolving through TGF-b1 secreted by F4/80+Ly6G+ macrophage. International Immunopharmacology. 2021; 95: 107480.

[153] Jin Z, Mendu SK, Birnir B. GABA is an effective immunomodulatory molecule. Amino Acids. 2013; 45: 87–94.

[154] Wheeler DW, Thompson AJ, Corletto F, Reckless J, Loke JC, Lapaque N, et al. Anaesthetic impairment of immune function is mediated via GABA(A) receptors. PLOS ONE. 2011; 6: e17152.

[155] Yuki K, Soriano SG, Shimaoka M. Sedative drug modulates T-cell and lymphocyte function-associated antigen-1 function. Anesthesia & Analgesia. 2011; 112: 830–838.

[156] Chen RM, Wu CH, Chang HC, Wu GJ, Lin YL, Sheu JR, et al. Propofol suppresses macrophage functions and modulates mitochondrial membrane potential and cellular adenosine triphosphate synthesis. Anesthesiology. 2003; 98: 1178–1185.

[157] Sun L, Ma W, Gao W, Xing Y, Chen L, Xia Z, et al. Propofol directly induces caspase-1-dependent macrophage pyroptosis through the NLRP3-ASC inflammasome. Cell Death & Disease. 2019; 10: 542.

[158] Senoner T, Velik-Salchner C, Luckner G, Tauber H. Anesthesia-induced oxidative stress: are there differences between intravenous and inhaled anesthetics? Oxidative Medicine and Cellular Longevity. 2021; 2021: 8782387.

[159] Cai Q, Liu G, Huang L, Guan Y, Wei H, Dou Z, et al. The role of dexmedetomidine in tumor-progressive factors in the perioperative period and cancer recurrence: a narrative review. Drug Design, Development and Therapy. 2022; 16: 2161–2175.

[160] Liu W, Yu W, Weng Y, Wang Y, Sheng M. Dexmedetomidine ameliorates the inflammatory immune response in rats with acute kidney damage. Experimental and Therapeutic Medicine. 2017; 14: 3602–3608.

[161] Li R, Lai IK, Pan JZ, Zhang P, Maze M. Dexmedetomidine exerts an anti-inflammatory effect via a2 adrenoceptors to prevent lipopolysaccharide-induced cognitive decline in mice. Anesthesiology. 2020; 133: 393–407.

[162] Umamaheswaran S, Dasari SK, Yang P, Lutgendorf SK, Sood AK. Stress, inflammation, and eicosanoids: an emerging perspective. Cancer and Metastasis Reviews. 2018; 37: 203–211.

[163] Sharify A, Mahmoudi M, Izad MH, Hosseini MJ, Sharify M. Effect of acute pain on splenic NK cell activity, lymphocyte proliferation and cytokine production activities. Immunopharmacology and Immunotoxicology. 2007; 29: 465–476.

[164] Jang Y, Yeom MY, Kang ES, Kang JW, Song HK. The antinociceptive effect of dexmedetomidine modulates spleen cell immunity in mice. International Journal of Medical Sciences. 2014; 11: 226–233.

[165] Cho JS, Seon K, Kim MY, Kim SW, Yoo YC. Effects of perioperative dexmedetomidine on immunomodulation in uterine cancer surgery: a randomized, controlled trial. Frontiers in Oncology. 2021; 11: 749003.

[166] Kamohara H, Kamohara T, Hikasa Y. A randomized clinical trial on effects of alfaxalone combined with medetomidine and midazolam in preventing stress-related neurohormonal and metabolic responses of isoflurane-anesthetized cats undergoing surgery. American Journal of Veterinary Research. 2022; 83: 1–10.

[167] Kang J, Zheng Z, Li X, Huang T, Rong D, Liu X, et al. Midazolam exhibits antitumour and enhances the efficiency of Anti-PD-1 immunotherapy in hepatocellular carcinoma. Cancer Cell International. 2022; 22: 312.

[168] Park HJ, Piao L, Seo EH, Lee SH, Kim SH. The effect of repetitive exposure to intravenous anesthetic agents on the immunity in mice. International Journal of Medical Sciences. 2020; 17: 428–436.

[169] Ohta N, Ohashi Y, Takayama C, Mashimo T, Fujino Y. Midazolam suppresses maturation of murine dendritic cells and priming of lipopolysaccharide-induced T helper 1-type immune response. Anesthesiology. 2011; 114: 355–362.

[170] Li J, Tan H, Zhou X, Zhang C, Jin H, Tian Y, et al. The protection of midazolam against immune mediated liver injury induced by lipopolysaccharide and galactosamine in mice. Frontiers in Pharmacology. 2018; 9: 1528.

[171] Horiguchi Y, Ohta N, Yamamoto S, Koide M, Fujino Y. Midazolam suppresses the lipopolysaccharide-stimulated immune responses of human macrophages via translocator protein signaling. International Immunopharmacology. 2019; 66: 373–382.

[172] Kim SN, Son SC, Lee SM, Kim CS, Yoo DG, Lee SK, et al. Midazolam inhibits proinflammatory mediators in the lipopolysaccharide-activated macrophage. Anesthesiology. 2006; 105: 105–110.

[173] Bosch DJ, Nieuwenhuijs-Moeke GJ, van Meurs M, Abdulahad WH, Struys MMRF. Immune modulatory effects of nonsteroidal anti-inflammatory drugs in the perioperative period and their consequence on postoperative outcome. Anesthesiology. 2022; 136: 843–860.

[174] Tao X, Zhang R, Du R, Yu T, Yang H, Li J, et al. EP3 enhances adhesion and cytotoxicity of NK cells toward hepatic stellate cells in a murine liver fibrosis model. Journal of Experimental Medicine. 2022; 219: e20212414.

[175] Wu Q, Liu Z, Gao Z, Luo Y, Li F, Yang C, et al. KLF5 inhibition potentiates anti-PD1 efficacy by enhancing CD8+ T-cell-dependent antitumor immunity. Theranostics. 2023; 13: 1381–1400.

[176] Pelly VS, Moeini A, Roelofsen LM, Bonavita E, Bell CR, Hutton C, et al. Anti-inflammatory drugs remodel the tumor immune environment to enhance immune checkpoint blockade efficacy. Cancer Discovery. 2021; 11: 2602–2619.

[177] Rao CV. Anti-inflammatory drugs decrease the PD-L1 expression and increase the CD8+ T-cell infiltration. Cancer Prevention Research. 2022; 15: 209–211.

[178] Wei J, Zhang J, Wang D, Cen B, Lang JD, DuBois RN. The COX-2-PGE2 pathway promotes tumor evasion in colorectal adenomas. Cancer Prevention Research. 2022; 15: 285–296.

[179] Abdel Shaheed C, Beardsley J, Day RO, McLachlan AJ. Immunomodulatory effects of pharmaceutical opioids and antipyretic analgesics: mechanisms and relevance to infection. British Journal of Clinical Pharmacology. 2022; 88: 3114–3131.

[180] Wang RD, Zhu JY, Zhu Y, Ge YS, Xu GL, Jia WD. Perioperative analgesia with parecoxib sodium improves postoperative pain and immune function in patients undergoing hepatectomy for hepatocellular carcinoma. Journal of Evaluation in Clinical Practice. 2020; 26: 992–1000.

[181] Ma W, Wang K, Du J, Luan J, Lou G. Multi-dose parecoxib provides an immunoprotective effect by balancing T helper 1 (Th1), Th2, Th17 and regulatory T cytokines following laparoscopy in patients with cervical cancer. Molecular Medicine Reports. 2015; 11: 2999–3008.

[182] Li Y, Zhou L, Li X, Chen G, Duan K, Ding B, et al. Parecoxib suppresses the increase of neutrophil-to-lymphocyte ratio after the modified radical mastectomy. Journal of Central South University. 2017; 42: 1048–1052. (In Chinese)

[183] Lv X, Li X, Guo K, Li T, Yang Y, Lu W, et al. Effects of systemic lidocaine on postoperative recovery quality and immune function in patients undergoing laparoscopic radical gastrectomy. Drug Design, Development and Therapy. 2021; 15: 1861–1872.

[184] Lee IW, Schraag S. The use of intravenous lidocaine in perioperative medicine: anaesthetic, analgesic and immune-modulatory aspects. Journal of Clinical Medicine. 2022; 11: 3543.

[185] Cata JP, Ramirez MF, Velasquez JF, Di AI, Popat KU, Gottumukkala V, et al. Lidocaine stimulates the function of natural killer cells in different experimental settings. Anticancer Research. 2017; 37: 4727–4732.

[186] Zheng B, Yang H, Zhang J, Wang X, Sun H, Hu F, et al. Lidocaine alleviates sepsis-induced acute lung injury in mice by suppressing tissue factor and matrix metalloproteinase-2/9. Oxidative Medicine and Cellular Longevity. 2021; 2021: 3827501.

[187] Zhang H, Qu M, Guo K, Wang Y, Gu J, Wu H, et al. Intraoperative lidocaine infusion in patients undergoing pancreatectomy for pancreatic cancer: a mechanistic, multicentre randomised clinical trial. British Journal of Anaesthesia. 2022; 129: 244–253.

[188] Yin Q, Sun L, Cai X, Lou F, Sun Y, Wang B, et al. Lidocaine ameliorates psoriasis by obstructing pathogenic CGRP signaling‒mediated sensory neuron‒dendritic cell communication. Journal of Investigative Dermatology. 2022; 142: 2173–2183.e6.

[189] Su K, Li XT, Hong FX, Jin M, Xue FS. Lidocaine pretreatment attenuates inflammatory response and protects against sepsis-induced acute lung injury via inhibiting potassium efflux-dependent NLRP3 activation. Inflammation Research. 2023; 72: 2221–2235.

[190] Wei Q, Xia M, Zhang Q, Wang Z. Effect of intravenous lidocaine infusion on perioperative cellular immunity and the quality of postoperative recovery in breast cancer patients: a randomized controlled trial. Gland Surgery. 2022; 11: 599–610.

[191] 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 Design, Development and Therapy. 2021; 15: 3379–3390.


Abstracted / indexed in

Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,200 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.

Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.

Chemical Abstracts Service Source Index The CAS Source Index (CASSI) Search Tool is an online resource that can quickly identify or confirm journal titles and abbreviations for publications indexed by CAS since 1907, including serial and non-serial scientific and technical publications.

Index Copernicus The Index Copernicus International (ICI) Journals database’s is an international indexation database of scientific journals. It covered international scientific journals which divided into general information, contents of individual issues, detailed bibliography (references) sections for every publication, as well as full texts of publications in the form of attached files (optional). For now, there are more than 58,000 scientific journals registered at ICI.

Geneva Foundation for Medical Education and Research The Geneva Foundation for Medical Education and Research (GFMER) is a non-profit organization established in 2002 and it works in close collaboration with the World Health Organization (WHO). The overall objectives of the Foundation are to promote and develop health education and research programs.

Scopus: CiteScore 1.3 (2023) Scopus is Elsevier's abstract and citation database launched in 2004. Scopus covers nearly 36,377 titles (22,794 active titles and 13,583 Inactive titles) from approximately 11,678 publishers, of which 34,346 are peer-reviewed journals in top-level subject fields: life sciences, social sciences, physical sciences and health sciences.

Embase Embase (often styled EMBASE for Excerpta Medica dataBASE), produced by Elsevier, is a biomedical and pharmacological database of published literature designed to support information managers and pharmacovigilance in complying with the regulatory requirements of a licensed drug.

Submission Turnaround Time

Conferences

Top