Skip to main navigation menu Skip to main content Skip to site footer

Review article: Biomedical intelligence

Vol. 142 No. 3536 (2012)

Prevention and control of surgical site infections: review of the Basel Cohort Study

  • Till Andrin Junker
  • Edin Mujagic
  • Henry Hoffmann
  • Rachel Rosenthal
  • Heidi Misteli
  • Marcel Zwahlen
  • Daniel Oertli
  • Sarah Tschudin-Sutter
  • Andreas F. Widmer
  • Walter Richard Marti
  • Walter Paul Weber
DOI
https://doi.org/10.4414/smw.2012.13616
Cite this as:
Swiss Med Wkly. 2012;142:w13616
Published
26.08.2012

Summary

Introduction: Surgical site infections (SSI) are the most common hospital-acquired infections among surgical patients, with significant impact on patient morbidity and health care costs. The Basel SSI Cohort Study was performed to evaluate risk factors and validate current preventive measures for SSI. The objective of the present article was to review the main results of this study and its implications for clinical practice and future research.

Summary of methods of the Basel SSI Cohort Study: The prospective observational cohort study included 6,283 consecutive general surgery procedures closely monitored for evidence of SSI up to 1 year after surgery. The dataset was analysed for the influence of various potential SSI risk factors, including timing of surgical antimicrobial prophylaxis (SAP), glove perforation, anaemia, transfusion and tutorial assistance, using multiple logistic regression analyses. In addition, post hoc analyses were performed to assess the economic burden of SSI, the efficiency of the clinical SSI surveillance system, and the spectrum of SSI-causing pathogens.

Review of main results of the Basel SSI Cohort Study: The overall SSI rate was 4.7% (293/6,283). While SAP was administered in most patients between 44 and 0 minutes before surgical incision, the lowest risk of SSI was recorded when the antibiotics were administered between 74 and 30 minutes before surgery. Glove perforation in the absence of SAP increased the risk of SSI (OR 2.0; CI 1.4–2.8; p <0.001). No significant association was found for anaemia, transfusion and tutorial assistance with the risk of SSI. The mean additional hospital cost in the event of SSI was CHF 19,638 (95% CI, 8,492–30,784). The surgical staff documented only 49% of in-hospital SSI; the infection control team registered the remaining 51%. Staphylococcus aureus was the most common SSI-causing pathogen (29% of all SSI with documented microbiology). No case of an antimicrobial-resistant pathogen was identified in this series.

Conclusions: The Basel SSI Cohort Study suggested that SAP should be administered between 74 and 30 minutes before surgery. Due to the observational nature of these data, corroboration is planned in a randomized controlled trial, which is supported by the Swiss National Science Foundation. Routine change of gloves or double gloving is recommended in the absence of SAP. Anaemia, transfusion and tutorial assistance do not increase the risk of SSI. The substantial economic burden of in-hospital SSI has been confirmed. SSI surveillance by the surgical staff detected only half of all in-hospital SSI, which prompted the introduction of an electronic SSI surveillance system at the University Hospital of Basel and the Cantonal Hospital of Aarau. Due to the absence of multiresistant SSI-causing pathogens, the continuous use of single-shot single-drug SAP with cefuroxime (plus metronidazole in colorectal surgery) has been validated.

Summary of methods of the Basel SSI Cohort Study

The Basel SSI cohort study was part of a quality improvement programme at Basel University Hospital. It was supported by the hospital executive board and approved by the local research ethics committee. A total of 6,283 consecutive surgeries, performed between 1 January 2000, and 31 December 2001 in the visceral, vascular, and trauma divisions were evaluated.

The outcome of interest was SSI according to CDC criteria. The surgical resident prospectively completed a paper-based surveillance form for each patient, including the type of SSI, date of diagnosis and treatment. Each form was cross-checked and signed by an attending surgeon. Follow-up after discharge was assessed by reviewing outpatient charts and by contacting family practitioners who performed the clinical checks after surgery. Where data were missing, study team physicians interviewed patients by telephone pursuant to a standardised questionnaire. An infectious disease specialist confirmed all SSI by comprehensive review of patient history, initial microbiology results and outcome >1 year after surgery.

More than eighty patient and procedure variables were recorded, including age, sex, American Society of Anaesthesiologists (ASA) score, type of procedure, surgical team members, division of surgical speciality, timing of SAP, wound class, duration of surgery, compromised asepsis, and many other known and suspected SSI risk factors. We used an electronically readable form created by Cardiff TELEForm Software (Cardiff TELEForm Desktop V 8.0, 2002, Verity Inc., Sunnyvale, CA) to export the data into an Excel file (Windows Microsoft Excel 2003, Microsoft Corporation).

The primary predictor variable of the first analysis was SAP timing. We divided time into intervals, and chose the cutoffs of 120 and 0 minutes before surgery on the basis of the findings of Classen et al. [7]. SAP was administered intravenously by the anaesthetists via single-shot infusion of 1.5 g cefuroxime in 20 ml sodium chloride solution over a few minutes in combination with metronidazole (500 mg, intravenous, 5 minutes) in colorectal patients. The exact time in minutes when the infusion of SAP ended was prospectively recorded by the anaesthesia team.

The main predictor variable of the second analysis was compromised asepsis due to glove perforation. The operating room nurse was responsible for the detection and registration, either directly when glove perforation itself was visible or indirectly when liquid was detected inside a glove. When double gloves were used, leakage of the inner glove was noted.

The main predictor variables of the third analysis were perioperative ABT and preoperative anaemia. Perioperative ABT was performed by the anesthesia team in the operating room. The number of allogeneic blood units was monitored. All blood donations were leukocyte-depleted (leukocytes <1.0 × 106/unit) by specific filtration (Optipure RZ 2000 filters, Baxter) and plasma was removed. Packed red cells were stored in saline-adenine-glucose-mannitol at 4 °C. Preoperative anaemia was defined as <120 g/l haemoglobin.

For analysis of the impact of surgical training on the incidence of SSI, surgeries were divided into two groups: tutorial assistance or autonomous surgery. Tutorial assistance was defined as surgical training of a resident by a board-certified surgeon, or operations conducted by an inexperienced general surgeon supervised by a board-certified surgeon with extensive expertise in the field.

For analysis of the economic burden of in-hospital SSI, the outcome variables were total duration of hospitalisation, duration of hospitalisation after surgery, duration of intensive care stay after surgery, duration of in-hospital use of antibiotics, patient charges and hospital costs. Since the collection of cost data was not part of the observational cohort study design, this information was derived from the hospital’s finance department. Their computerised internal cost and activity accounting database directly linked internal hospital costs with patient charges. Control patients had no SSI and were matched to case patients by age, procedure code, and National Nosocomial Infection Surveillance (NNIS) risk index.

To assess the quality of in-house SSI surveillance by surgeons during the study period, an infection control team (ICT) evaluated all general surgery patients by full chart review and by gathering additional clinical information. Data from the surgeons’ surveillance system were available for the ICT. The ICT consisted of a board certified infectious diseases specialist and a hospital epidemiologist.

Microbiological evaluation consisted of microscopic direct preparation and cultures with antibiotic resistance testing. As superficial wound swabbing is difficult to interpret, the decision to treat superficial SSI with antibiotics was made clinically rather than on the basis of the results of a wound swab.

All analyses were conducted at the Institute of Social and Preventive Medicine in Bern using Stata version 9.2 as well as version 10 (Stata Statistical Software; Stata Corp., College Station, TX). In the first step, univariable analyses were performed to assess the association between the primary predictor variable and the unadjusted likelihood of SSI occurrence. Then multivariable logistic regression models were used to examine the risk-adjusted association between the primary predictor variable and the odds of contracting SSI. The decision on what variables to include in the regression model was based on their potential role as SSI risk factors or on indications of differences in distribution. Crude and adjusted odds ratios (OR), 95% confidence intervals (CI), and values were used to express the strength of this association. All P values were two-sided and statistical significance was set at the 0.05 level.

References

  1. Mangram AJ, Horan TC, Pearson ML, Silver LC, Jarvis WR. Guideline for prevention of surgical site infection, 1999. Hospital Infection Control Practices Advisory Committee. Infect Control Hosp Epidemiol. 1999;20(4):250–78.
  2. Urban JA. Cost analysis of surgical site infections. Surg Infect. (Larchmt ) 2006;7(Suppl 1):S19–22.
  3. Weber WP, Marti WR, Zwahlen M, Misteli H, Rosenthal R, Reck S, et al. The timing of surgical antimicrobial prophylaxis. Ann Surg. 2008;247(6):918–26.
  4. Dellinger EP, Gross PA, Barrett TL, Krause PJ, Martone WJ, McGowan JE, Jr. et al. Quality standard for antimicrobial prophylaxis in surgical procedures. Infectious Diseases Society of America. Clin Infect Dis. 1994;18(3):422–7.
  5. Bratzler DW, Houck PM. Antimicrobial prophylaxis for surgery: an advisory statement from the National Surgical Infection Prevention Project. Clin Infect Dis. 2004;38(12):1706–15.
  6. Burke JF. The effective period of preventive antibiotic action in experimental incisions and dermal lesions. Surgery. 196;50:161–8.
  7. Classen DC, Evans RS, Pestotnik SL, Horn SD, Menlove RL, Burke JP. The timing of prophylactic administration of antibiotics and the risk of surgical-wound infection. N Engl J Med. 1992;326(5):281–6.
  8. Geroulanos S, Marathias K, Kriaras J, Kadas B. Cephalosporins in surgical prophylaxis. J Chemother. 2001;13 Spec No 1(1):23–6.
  9. van Kasteren ME, Kullberg BJ, de Boer AS, Mintjes-de GJ, Gyssens IC. Adherence to local hospital guidelines for surgical antimicrobial prophylaxis: a multicentre audit in Dutch hospitals. J Antimicrob Chemother. 2003;51(6):1389–96.
  10. Bratzler DW, Hunt DR. The surgical infection prevention and surgical care improvement projects: national initiatives to improve outcomes for patients having surgery. Clin Infect Dis. 2006;43(3):322–30.
  11. Misteli H, Weber WP, Reck S, Rosenthal R, Zwahlen M, Fueglistaler P, et al. Surgical glove perforation and the risk of surgical site infection. Arch Surg. 2009;144(6):553–8.
  12. Esteban JI, Gomez J, Martell M, Cabot B, Quer J, Camps J, et al. Transmission of hepatitis C virus by a cardiac surgeon. N Engl J Med. 1996;334(9):555–60.
  13. Harpaz R, Von SL, Averhoff FM, Tormey MP, Sinha SD, Kotsopoulou K, et al. Transmission of hepatitis B virus to multiple patients from a surgeon without evidence of inadequate infection control. N Engl J Med. 1996;334(9):549–54.
  14. Kiyosawa K, Sodeyama T, Tanaka E, Nakano Y, Furuta S, Nishioka K, et al. Hepatitis C in hospital employees with needlestick injuries. Ann Intern Med. 1991;115(5):367–9.
  15. Greco RJ, Wheatley M, McKenna P. Risk of blood contact through surgical gloves in aesthetic procedures. Aesthetic Plast Surg. 1993;17(2):167–8.
  16. Kojima Y, Ohashi M. Unnoticed glove perforation during thoracoscopic and open thoracic surgery. Ann Thorac Surg. 2005;80(3):1078–80.
  17. Maffulli N, Capasso G, Testa V. Glove perforation in elective orthopedic surgery. Acta Orthop Scand. 1989;60(5):565–6.
  18. Yinusa W, Li YH, Chow W, Ho WY, Leong JC. Glove punctures in orthopaedic surgery. Int Orthop. 2004;28(1):36–9.
  19. Dodds RD, Guy PJ, Peacock AM, Duffy SR, Barker SG, Thomas MH. Surgical glove perforation. Br J Surg. 1988;75(10):966–8.
  20. Gunasekera PC, Fernando RJ, de Silva KK. Glove failure: an occupational hazard of surgeons in a developing country. J R Coll Surg Edinb. 1997;42(2):95–7.
  21. Laine T, Aarnio P. How often does glove perforation occur in surgery? Comparison between single gloves and a double-gloving system. Am J Surg. 2001;181(6):564–6.
  22. Nicolai P, Aldam CH, Allen PW. Increased awareness of glove perforation in major joint replacement. A prospective, randomised study of Regent Biogel Reveal gloves. J Bone Joint Surg Br. 1997;79(3):371–3.
  23. Punyatanasakchai P, Chittacharoen A, Ayudhya NI. Randomized controlled trial of glove perforation in single- and double-gloving in episiotomy repair after vaginal delivery. J Obstet Gynaecol Res. 2004;30(5):354–7.
  24. Quebbeman EJ, Telford GL, Wadsworth K, Hubbard S, Goodman H, Gottlieb MS. Double gloving. Protecting surgeons from blood contamination in the operating room. Arch Surg. 1992;127(2):213–6.
  25. Weber WP, Zwahlen M, Reck S, Misteli H, Rosenthal R, Buser AS, et al. The association of preoperative anemia and perioperative allogeneic blood transfusion with the risk of surgical site infection. Transfusion. 2009;49(9):1964–70.
  26. Blumetti J, Luu M, Sarosi G, Hartless K, McFarlin J, Parker B, et al. Surgical site infections after colorectal surgery: do risk factors vary depending on the type of infection considered? Surgery. 2007;142(5):704–11.
  27. Ford CD, VanMoorleghem G, Menlove RL. Blood transfusions and postoperative wound infection. Surgery. 1993;113(6):603–7.
  28. Friedman ND, Bull AL, Russo PL, Leder K, Reid C, Billah B, et al. An alternative scoring system to predict risk for surgical site infection complicating coronary artery bypass graft surgery. Infect Control Hosp Epidemiol. 2007;28(10):1162–8.
  29. Morris CD, Sepkowitz K, Fonshell C, Margetson N, Eagan J, Miransky J, et al. Prospective identification of risk factors for wound infection after lower extremity oncologic surgery. Ann Surg Oncol. 2003;10(7):778–82.
  30. Murphy GJ, Reeves BC, Rogers CA, Rizvi SI, Culliford L, Angelini GD. Increased mortality, postoperative morbidity, and cost after red blood cell transfusion in patients having cardiac surgery. Circulation. 2007;116(22):2544–52.
  31. Talbot TR, D’Agata EM, Brinsko V, Lee B, Speroff T, Schaffner W. Perioperative blood transfusion is predictive of poststernotomy surgical site infection: marker for morbidity or true immunosuppressant? Clin Infect Dis. 2004;38(10):1378–82.
  32. Tang R, Chen HH, Wang YL, Changchien CR, Chen JS, Hsu KC, et al. Risk factors for surgical site infection after elective resection of the colon and rectum: a single-center prospective study of 2,809 consecutive patients. Ann Surg. 2001;234(2):181–9.
  33. Walz JM, Paterson CA, Seligowski JM, Heard SO. Surgical site infection following bowel surgery: a retrospective analysis of 1446 patients. Arch Surg. 2006;141(10):1014–8.
  34. Ali ZA, Lim E, Motalleb-Zadeh R, Ali AA, Callaghan CJ, Gerrard C, et al. Allogenic blood transfusion does not predispose to infection after cardiac surgery. Ann Thorac Surg. 2004;78(5):1542–6.
  35. Johnston P, Wynn-Jones H, Chakravarty D, Boyle A, Parker MJ. Is perioperative blood transfusion a risk factor for mortality or infection after hip fracture? J Orthop Trauma. 2006;20(10):675–9.
  36. Koval KJ, Rosenberg AD, Zuckerman JD, Aharonoff GB, Skovron ML, Bernstein RL, et al. Does blood transfusion increase the risk of infection after hip fracture? J Orthop Trauma. 1997;11(4):260–5.
  37. Bilgin YM, van de Watering LM, Eijsman L, Versteegh MI, Brand R, van Oers MH, et al. Double-blind, randomized controlled trial on the effect of leukocyte-depleted erythrocyte transfusions in cardiac valve surgery. Circulation. 2004;109(22):2755–60.
  38. Busch OR, Hop WC, Hoynck van Papendrecht MA, Marquet RL, Jeekel J. Blood transfusions and prognosis in colorectal cancer. N Engl J Med. 1993;328(19):1372–6.
  39. Heiss MM, Mempel W, Jauch KW, Delanoff C, Mayer G, Mempel M, et al. Beneficial effect of autologous blood transfusion on infectious complications after colorectal cancer surgery. Lancet. 1993;342(8883):1328–33.
  40. Houbiers JG, Brand A, van de Watering LM, Hermans J, Verwey PJ, Bijnen AB, et al. Randomised controlled trial comparing transfusion of leucocyte-depleted or buffy-coat-depleted blood in surgery for colorectal cancer. Lancet. 1994;344(8922):573–8.
  41. Jensen LS, Kissmeyer-Nielsen P, Wolff B, Qvist N. Randomised comparison of leucocyte-depleted versus buffy-coat-poor blood transfusion and complications after colorectal surgery. Lancet 1996;348(9031):841–5.
  42. Vamvakas EC, Carven JH, Hibberd PL. Blood transfusion and infection after colorectal cancer surgery. Transfusion. 1996;36(11-12):1000–8.
  43. Vamvakas EC, Carven JH. Transfusion of white-cell containing allogeneic blood components and postoperative wound infection: effect of confounding factors. Transfus Med. 1998;8(1):29–36.
  44. Dunne JR, Malone D, Tracy JK, Gannon C, Napolitano LM. Perioperative anemia: an independent risk factor for infection, mortality, and resource utilization in surgery. J Surg Res. 2002;102(2):237–44.
  45. Shander A, Knight K, Thurer R, Adamson J, Spence R. Prevalence and outcomes of anemia in surgery: a systematic review of the literature. Am J Med. 2004;116(Suppl 7A):58S–69S.
  46. Rosenthal R, Weber WP, Zwahlen M, Misteli H, Reck S, Oertli D, et al. Impact of surgical training on incidence of surgical site infection. World J Surg. 2009;33(6):1165–73.
  47. Halsted WS. The training of the surgeon. Bull Johns Hop Hosp. 15, 267–275. Ref Type: Generic
  48. Sutherland LM, Middleton PF, Anthony A, Hamdorf J, Cregan P, Scott D, et al. Surgical simulation: a systematic review. Ann Surg. 2006;243(3):291–300.
  49. Weber WP, Zwahlen M, Reck S, Feder-Mengus C, Misteli H, Rosenthal R, et al. Economic burden of surgical site infections at a European university hospital. Infect Control Hosp Epidemiol. 2008;29(7):623–9.
  50. Coello R, Charlett A, Wilson J, Ward V, Pearson A, Borriello P. Adverse impact of surgical site infections in English hospitals. J Hosp Infect. 2005;60(2):93–103.
  51. Engemann JJ, Carmeli Y, Cosgrove SE, Fowler VG, Bronstein MZ, Trivette SL, et al. Adverse clinical and economic outcomes attributable to methicillin resistance among patients with Staphylococcus aureus surgical site infection. Clin Infect Dis. 2003;36(5):592–8.
  52. Kirkland KB, Briggs JP, Trivette SL, Wilkinson WE, Sexton DJ. The impact of surgical-site infections in the 1990s: attributable mortality, excess length of hospitalization, and extra costs. Infect Control Hosp Epidemiol. 1999;20(11):725–30.
  53. Poulsen KB, Bremmelgaard A, Sorensen AI, Raahave D, Petersen JV. Estimated costs of postoperative wound infections. A case-control study of marginal hospital and social security costs. Epidemiol Infect. 1994;113(2):283–95.
  54. Vegas AA, Jodra VM, Garcia ML. Nosocomial infection in surgery wards: a controlled study of increased duration of hospital stays and direct cost of hospitalization. Eur J Epidemiol. 1993;9(5):504–10.
  55. Whitehouse JD, Friedman ND, Kirkland KB, Richardson WJ, Sexton DJ. The impact of surgical-site infections following orthopedic surgery at a community hospital and a university hospital: adverse quality of life, excess length of stay, and extra cost. Infect Control Hosp Epidemiol. 2002;23(4):183–9.
  56. Fry DE. The economic costs of surgical site infection. Surg Infect. (Larchmt ) 2002;3 (Suppl 1):S37–S43.
  57. Rosenthal R, Weber WP, Marti WR, Misteli H, Reck S, Dangel M, et al. Surveillance of surgical site infections by surgeons: biased underreporting or useful epidemiological data? J Hosp Infect. 2010 March 12.
  58. Haley RW, Morgan WM, Culver DH, White JW, Emori TG, Mosser J, et al. Update from the SENIC project. Hospital infection control: recent progress and opportunities under prospective payment. Am J Infect Control. 1985;13(3):97–108.
  59. Glenister HM, Taylor LJ, Bartlett CL, Cooke EM, Mulhall AB. Introduction of laboratory based ward liaison surveillance of hospital infection into six district general hospitals. J Hosp Infect. 1993;25(3):161–72.
  60. Glenister HM. How do we collect data for surveillance of wound infection? J Hosp Infect. 1993;24(4):283–9.
  61. Broderick A, Mori M, Nettleman MD, Streed SA, Wenzel RP. Nosocomial infections: validation of surveillance and computer modeling to identify patients at risk. Am J Epidemiol. 1990;131(4):734–42.
  62. Poulsen KB, Meyer M. Infection registration underestimates the risk of surgical wound infections. J Hosp Infect. 1996;33(3):207–15.
  63. Misteli H, Widmer AF, Rosenthal R, Oertli D, Marti WR, Weber WP. Spectrum of pathogens in surgical site infections at a Swiss university hospital. Swiss Med Wkly. 2011;140:w13146.
  64. Bratzler DW, Houck PM, Richards C, Steele L, Dellinger EP, Fry DE et al. Use of antimicrobial prophylaxis for major surgery: baseline results from the National Surgical Infection Prevention Project. Arch Surg. 2005;140(2):174–82.
  65. Steinberg JP, Braun BI, Hellinger WC, Kusek L, Bozikis MR, Bush AJ, et al. Timing of antimicrobial prophylaxis and the risk of surgical site infections: results from the Trial to Reduce Antimicrobial Prophylaxis Errors. Ann Surg. 2009;250(1):10–6.
  66. van Kasteren ME, Mannien J, Ott A, Kullberg BJ, de Boer AS, Gyssens IC. Antibiotic prophylaxis and the risk of surgical site infections following total hip arthroplasty: timely administration is the most important factor. Clin Infect Dis. 2007;44(7):921–7.
  67. Platt R, Zaleznik DF, Hopkins CC, Dellinger EP, Karchmer AW, Bryan CS, et al. Perioperative antibiotic prophylaxis for herniorrhaphy and breast surgery. N Engl J Med. 1990;322(3):153–60.
  68. Yerdel MA, Akin EB, Dolalan S, Turkcapar AG, Pehlivan M, Gecim IE, et al. Effect of single-dose prophylactic ampicillin and sulbactam on wound infection after tension-free inguinal hernia repair with polypropylene mesh: the randomized, double-blind, prospective trial. Ann Surg. 2001;233(1):26–33.
  69. Lewis RT, Weigand FM, Mamazza J, Lloyd-Smith W, Tataryn D. Should antibiotic prophylaxis be used routinely in clean surgical procedures: a tentative yes. Surgery 1995;118(4):742–6.

Most read articles by the same author(s)

1 2 3 4 > >>