Scolaris Content Display Scolaris Content Display

Antibioticoterapia de corta duración versus ciclo prolongado para la neumonía adquirida en el hospital en adultos graves

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Capellier 2012 {published data only}

Capellier G, Mockly H, Charpentier C, Annane D, Blasco G, Desmettre T, et al. Early‐onset ventilator‐associated pneumonia in adults randomized clinical trial: comparison of 8 versus 15 days of antibiotic treatment. PloS One 2012;7(8):e41290.

Chastre 2003a {published data only}

Chastre J, Wolff M, Fagon JY, Chevret S, Thomas F, Wermert D, et al. Comparison of 8 vs 15 days of antibiotic therapy for ventilator‐associated pneumonia in adults: a randomized trial. JAMA 2003;290(19):2588‐98.

Fekih Hassen 2009 {published data only}

Fekih Hassen M, Ayed S, Ben Sik Ali H, Gharbi R, Marghli S, Elatrous S. Duration of antibiotic therapy for ventilator‐associated pneumonia: comparison of 7 and 10 days. A pilot study. Annales Françaises d'Anesthésie et de Réanimation 2009;28(1):16‐23.

Kollef 2012 {published data only}

Kollef M, Chastre J, Restrepo M, Michiels B, Kaniga K, Cirillo I, et al. A randomized trial of 7‐day doripenem versus 10‐day imipenem‐cilastatin for ventilator‐associated pneumonia. Critical Care 2012;16:R218.

Medina 2007 {published and unpublished data}

Medina JC, Perez Protto SE, Paciel D, Pontet J, Saldun P, Berro M. Antibiotic treatment for the ventilator‐associated pneumonia: 8 vs. 12 days randomized trial preliminary data. Annual Interscience Conference on Antimicrobial Agents and Chemotherapy, Chicago, IL. 2007.

Singh 2000 {published data only}

Singh N, Rogers P, Atwood CW, Wagener MM, Yu VL. Short‐course empiric antibiotic therapy for patients with pulmonary infiltrates in the intensive care unit. A proposed solution for indiscriminate antibiotic prescription. American Journal of Respiratory and Critical Care Medicine 2000;162:505‐11.

Referencias de los estudios excluidos de esta revisión

Bouadma 2010 {published data only}

Bouadma L, Luyt CE, Tubach F, Cracco C, Alvarez A, Schwebel C, et al. Use of procalcitonin to reduce patients' exposure to antibiotics in intensive care units (PRORATA trial): a multicentre randomised controlled trial. Lancet 2010;375(9713):463‐74.

Cai 2001 {published data only}

Cai S, Zhang J, Qian G. Impact of quantitative and qualitative pathogen culture on the outcome of ventilator‐associated pneumonia. Zhonghua Jie He He Hu Xi Za Zhi 2001;24(8):494‐7.

CCCTG 2006 {published data only}

Canadian Critical Care Trials Group. A randomized trial of diagnostic techniques for ventilator‐associated pneumonia. New England Journal of Medicine 2006;355(25):2619‐30.

Chastre 2003b {published data only}

Chastre J, Wolff M, Fagon JY, Chevret S. Comparison of two durations of antibiotic therapy to treat ventilator‐associated pneumonia (VAP). American Thoracic Society Meeting, Seattle, Washington. 2003:A015.

Fagon 2000 {published data only}

Fagon JY, Chastre J, Wolff M, Gervais C, Parer‐Aubas S, Stéphan F, et al. Invasive and noninvasive strategies for management of suspected ventilator‐associated pneumonia. A randomized trial. Annals of Internal Medicine 2000;132(8):621‐30.

Hochreiter 2009 {published data only (unpublished sought but not used)}

Hochreiter M,  Köhler T,  Schweiger AM,  Keck FS,  Bein B,  von Spiegel T,  et al. Procalcitonin to guide duration of antibiotic therapy in intensive care patients: a randomized prospective controlled trial. Critical Care 2009;13(3):R83.

Ibrahim 2001 {published data only}

Ibrahim EH,  Ward S,  Sherman G,  Schaiff R,  Fraser VJ,  Kollef MH. Experience with a clinical guideline for the treatment of ventilator‐associated pneumonia. Critical Care Medicine 2001;29(6):1109‐15.

Kim 2009 {published data only}

Kim JW, Chung J, Jang HJ, Hong SB, Lim CM, Koh Y. De‐escalation of antibiotics in critically ill patients with hospital‐acquired pneumonia. Respirology. 2009; Vol. 14:A129.

Kim 2012 {published data only}

Kim JW, Chung J, Choi SH, Jang HJ, Hong SB, Lim CM, et al. Early use of imipenem/cilastatin and vancomycin followed by de‐escalation versus conventional antimicrobials without de‐escalation for patients with hospital‐acquired pneumonia in a medical ICU: a randomized clinical trial. Critical Care 2012;16(1):R28.

Kollef 2005 {published data only}

Kollef MH,  Kollef KE. Antibiotic utilization and outcomes for patients with clinically suspected ventilator‐associated pneumonia and negative quantitative BAL culture results. Chest 2005;128:2706‐13.

Labelle 2012 {unpublished data only}

Labelle AJ, Schoenberg N, Skrupky L, Kollef M. Five versus seven day antibiotic course for the treatment of pneumonia in the intensive care unit. American Journal of Respiratory and Critical Care Medicine. 2012; Vol. 185:A6078.

Maldonado‐Ortiz 2004 {unpublished data only}

Maldonado‐Ortiz EA, Niederman MS, Nacul F, Rodríguez A, Ceballos S, Osorio J. How long is enough to treat ventilator associated pneumonia? A randomized pilot study: the Latinoamerican experience. Chest 2004;126(Suppl 4):717.

Micek 2004 {published data only}

Micek ST,  Ward S,  Fraser VJ,  Kollef MH. A randomized controlled trial of an antibiotic discontinuation policy for clinically suspected ventilator‐associated pneumonia. Chest 2004;125(5):1791‐9.

Nobre 2008 {published data only}

Nobre V, Harbarth S, Graf JD, Rohner P, Pugin J. Use of procalcitonin to shorten antibiotic treatment duration in septic patients: a randomized trial. American Journal of Respiratory and Critical Care Medicine 2008;177(5):498‐505.

Peery 2001 {published data only}

Peery C, Chendrasekhar A, Moorman DW, Timberlake GA. Ventilator‐associated pneumonia therapy: protected specimen brushing versus tracheal aspirate data. Journal of Applied Research 2001;1(2):1.

Pontet 2007 {published and unpublished data}

Pontet J, Paciel D, Olivera W, Bentancourt S, Cancela M, Gervas J. Procalcitonin (PCT) guided antibiotic treatment in ventilator associated pneumonia (VAP). Multi‐centre, clinical prospective, randomized‐controlled study. American Thoracic Society International Conference, San Francisco, California, USA. 2007:A76.

Rello 2004 {published data only}

Rello J,  Vidaur L,  Sandiumenge A,  Rodríguez A,  Gualis B,  Boque C,  et al. De‐escalation therapy in ventilator‐associated pneumonia. Critical Care Medicine 2004;32(11):2183‐90.

Sanchez‐Nieto 1998 {published data only}

Sanchez‐Nieto JM, Torres A, Garcia‐Cordoba F, El‐Ebiary M, Carrillo A, Ruiz J, et al. Impact of invasive and noninvasive quantitative culture sampling on outcome of ventilator‐associated pneumonia: a pilot study. American Journal of Respiratory and Critical Care Medicine 1998;157(2):371‐6.

Schroeder 2007 {published data only (unpublished sought but not used)}

Schroeder S,  Hochreiter M,  Koehler T,  Schweiger AM,  Bein B,  Keck FS,  et al. Procalcitonin (PCT)‐guided algorithm reduces length of antibiotic treatment in surgical intensive care patients with severe sepsis: results of a prospective randomized study. Langenbeck's Archives of Surgery 2009;394:221‐6.

Singh 1998 {published data only}

Singh N. Short‐course empiric antibiotic therapy for suspected nosocomial pneumonia: a proposed solution for indiscriminate antibiotic prescription for pulmonary infiltrates in the ICU. Annual Meeting of the Infectious Disease Society of America, Denver, Colorado. 1998.

Sole 2000 {published data only}

Sole VJ, Fernandez JA, Benitez AB, Cardenosa Cendrero JA, Rodriguez de CF. Impact of quantitative invasive diagnostic techniques in the management and outcome of mechanically ventilated patients with suspected pneumonia. Critical Care Medicine 2000;28(8):2737‐41.

Stolz 2009a {published and unpublished data}

Stolz D,  Smyrnios N,  Eggimann P,  Pargger H,  Thakkar N,  Siegemund M,  et al. Procalcitonin for reduced antibiotic exposure in ventilator‐associated pneumonia: a randomised study. European Respiratory Journal 2009;34(6):1364‐75.

Stolz 2009b {published data only}

Stolz D, Smyrnios N, Eggimann P, Pargger H, Thakkar N, Siegemund M, et al. Procalcitonin for antibiotic de‐escalation in ventilator associated pneumonia ‐ a randomized study. European Respiratory Society Annual Congress, Vienna. 2009:2798.

Svoboda 2007 {published data only}

Svoboda P,  Kantorová I,  Scheer P,  Radvanova J,  Radvan M. Can procalcitonin help us in timing of re‐intervention in septic patients after multiple trauma or major surgery?. Hepatogastroenterology 2007;54:359‐63.

Wolff 2003 {published data only}

Wolff M, Chastre J, Fagon JY, Chevret S. Comparison of two durations of antibiotic therapy for ventilator‐associated pneumonia (VAP) caused by non‐fermenting gram negative bacilli (NF‐GNB). American Thoracic Society Meeting Seattle, Washington. 2003:A860.

NCT00410527 {published data only}

Effectiveness of short‐course versus standard antibiotic therapy in ICU patients. http://clinicaltrials.gov/ct2/show/study/NCT00410527 2006 (accessed 5 June, 2015).

NCT01554657 {published data only}

Five versus seven day antibiotic course for the treatment of pneumonia in the intensive care unit. https://clinicaltrials.gov/show/NCT01554657 2012 (accessed 5 June, 2015).

NCT01994980 {published data only}

Duration of antibiotic treatment for early VAP. http://clinicaltrials.gov/show/NCT01994980 2013 (accessed 5 June, 2015).

Agrafiotis 2010

Agrafiotis M, Siempos II, Falagas ME. Frequency, prevention, outcome and treatment of ventilator‐associated tracheobronchitis: systematic review and meta‐analysis. Respiratory Medicine 2010;104:325‐36.

ATS/IDSA 2005

American Thoracic Society, Infectious Diseases Society of America. Guidelines for the management of adults with hospital‐acquired, ventilator‐associated, and healthcare‐associated pneumonia. American Journal of Respiratory and Critical Care Medicine 2005;171(4):388‐416.

Bekaert 2011

Bekaert M, Timsit JF, Vansteelandt S, Depuydt P, Vésin A, Garrouste‐Orgeas M, et al. Attributable mortality of ventilator‐associated pneumonia: a reappraisal using causal analysis. American Journal of Respiratory and Critical Care Medicine 2011;184(10):133‐9.

Bignardi 1998

Bignardi GE. Risk factors for Clostridium difficile infection. Journal of Hospital Infection 1998;40(1):1‐15.

Blot 2013

Blot S, Koulenti D, Dimopoulos G, Martin C, Komnos A, Krueger WA, et al. Prevalence, risk factors, and mortality for ventilator‐associated pneumonia in middle‐aged, old, and very old critically ill patients. Critical Care Medicine 2014;42:601‐9.

Boyer 2015

Boyer AF, Schoenberg N, Babcock H, McMullen KM, Micek ST, Kollef MH. A prospective evaluation of ventilator‐associated conditions and infection‐related ventilator‐associated conditions. Chest 2015;147:68‐81.

BSAC 2008

Masterton RG,  Galloway A,  French G,  Street M,  Armstrong J,  Brown E,  et al. Guidelines for the management of hospital‐acquired pneumonia in the UK: report of the working party on hospital‐acquired pneumonia of the British Society for Antimicrobial Chemotherapy. Journal of Antimicrobial Chemotherapy 2008;62(1):5‐34.

Burgmann 2010

Burgmann H, Hiesmayer J, Savey A, Bauer P, Metnitz B, Metnitz P. Impact of nosocomial infections on clinical outcome and resource consumption in critically ill patients. Intensive Care Medicine 2010;36:1597‐601.

Chant 2012

Chant C, Leung A, Friedrich JO. Optimal dosing of antibiotics in critically ill patients by using continuous/extended infusions: a systematic review and meta‐analysis. Critical Care 2012;17:R279.

Chastre 2002

Chastre J, Fagon JY. Ventilator‐associated pneumonia. American Journal of Respiratory and Critical Care Medicine 2002;165:867‐903.

Combes 2007

Combes A, Luyt CE, Fagon JY, Wolff M, Trouillet JL, Chastre J. Early predictors for infection recurrence and death in patients with ventilator‐associated pneumonia. Critical Care Medicine 2007;35(1):146‐54.

Conway Morris 2011

Morris AC, Kefala K, Simpson AJ, Wilkinson TS, Everingham K, Kerslake D, et al. Evaluation of the effect of diagnostic methodology on the reported incidence of ventilator‐associated pneumonia. Thorax 2011;64:516‐22.

Dallas 2011

Dallas J, Skrupky L, Abebe N, Boyle W, Kollef M. Ventilator‐associated tracheobronchitis in a mixed surgical and medical ICU population. Chest 2011;139:513‐8.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Dennesen 2001

Dennesen PJ, van der Ven AJ, Kessels AG, Ramsay G, Bonten MJ. Resolution of infectious parameters after antimicrobial therapy in patients with ventilator‐associated pneumonia. American Journal of Respiratory and Critical Care Medicine 2001;163(6):1371‐5.

Dimopoulos 2013

Dimopoulos G, Poulakou G, Pneumatikos IA, Armaganidis A, Kollef MH, Matthaiou DK. Short‐ versus long‐duration antibiotic regimens for ventilator‐associated pneumonia: a systematic review and meta‐analysis. Chest 2013;144:1759‐67.

Dubberke 2014

Dubberke E, Carling P, Carrico R, Donskey C, Loo V, McDonald L, et al. Strategies to prevent Clostridium difficile infections in acute care hospitals: 2014 Update. Infection Control and Hospital Epidemiology 2014;35:628‐45.

Dugan 2003

Dugan HA, MacLaren R, Jung R. Duration of antimicrobial therapy for nosocomial pneumonia: possible strategies for minimizing antimicrobial use in intensive care units. Journal of Clinical Pharmacy and Therapeutics 2003;28(2):123‐9.

El Solh 2007

El Solh AA, Choi G, Schultz MJ, Pineda LA, Mankowski C. Clinical and hemostatic responses to treatment in ventilator‐associated pneumonia: role of bacterial pathogens. Critical Care Medicine 2007;35(2):490‐6.

Esperatti 2010

Esperatti M, Ferrer M, Theessen A, Liapikou A, Valencia M, Saucedo L, et al. Nosocomial pneumonia in the intensive care unit acquired by mechanically ventilated versus nonventilated patients. American Journal of Respiratory and Critical Care Medicine 2010;182:1533‐9.

FDA 2014

US Food, Drug Administration. FDA approves label changes for antibacterial Doribax (doripenem) describing increased risk of death for ventilator patients with pneumonia. FDA Drug Safety Communication2014:http://www.fda.gov/Drugs/DrugSafety/ucm387971.htm (accessed 4 May 2015).

Gastmeier 2009

Gastmeier P, Sohr D, Geffers C, Rüden H, Vonberg R, Welte T. Early‐ and late‐onset pneumonia: is this still a useful classification?. Antimicrobial Agents and Chemotherapy 2009;53:2714‐8.

Grammatikos 2008

Grammatikos AP, Siempos II, Michalopoulos A, Falagas ME. Optimal duration of the antimicrobial treatment of ventilator‐acquired pneumonia. Expert Review of Anti‐infective Therapy 2008;6(6):861‐6.

Hayashi 2013

Hayashi Y, Morisawa K, Klompas M, Jones M, Bandeshe H, Boots R, et al. Toward improved surveillance: the impact of ventilator‐associated complications on length of stay and antibiotic use in patients in intensive care units. Clinical Infectious Disease 2013;56:471‐7.

HELICS 2004

Hospital in Europe Link for Infection Control through Surveillance (HELICS). Surveillance of Nosocomial infections in intensive care units, Protocol Version 6.1. http://ecdc.europa.eu/en/activities/surveillance/HAI/Documents/0409_IPSE_ICU_protocol.pdf 2004 (accessed 9 January 2015).

Higgins 2002

Higgins JP, Thompson SG. Quantifying heterogeneity in a meta‐analysis. Statistics in Medicine 2002;21:1539‐58.

Horan 2008

Horan TC, Andrus M, Dudeck MA. CDC/NHSN surveillance definition of health care‐associated infection and criteria for specific types of infections in the acute care setting. American Journal of Infection Control 2008;36:309‐32.

Jensen 2011

Jensen JU, Hein L, Lundgren B, Bestle MH, Mohr TT, Andersen MH, et al. Procalcitonin‐guided interventions against infections to increase early appropriate antibiotics and improve survival in the intensive care unit: a randomized trial. Critical Care Medicine 2011;39:2048‐58.

Kipnis 2014

Kipnis E, Nseir S. Ventilator‐associated events (VAEs): VAE Victis? (Woe to the conquered?)*. Critical Care Medicine 2014;42(8):1949‐50.

Klein Klouwenberg 2014

Klein Klouwenberg P, van Mourik M, Ong D, Horn J, Schultz M, Cremer O, et al. Electronic implementation of a novel surveillance paradigm for ventilator‐associated events. Feasibility and validation. American Journal of Respiratory and Critical Care Medicine 2014;189:947‐55.

Kohlenberg 2010

Kohlenberg A, Schwab F, Behnke M, Geffers C, Gastmeier P. Pneumonia associated with invasive and noninvasive ventilation: an analysis of the German nosocomial infection surveillance system database. Intensive Care Medicine 2010;36(6):971‐8.

Kollef 1995

Kollef MH, Silver P, Murphy DM, Trovillion E. The effect of late‐onset ventilator‐associated pneumonia in determining patient mortality. Chest 1995;108(6):1655‐62.

Kollef 2012b

Kollef MH, Micek ST. Antimicrobial stewardship programs: mandatory for all ICUs. Critical Care 2012;16(6):179.

Kopterides 2010

Kopterides P, Siempos II, Tsangaris I, Tsantes A, Armaganidis A. Procalcitonin‐guided algorithms of antibiotic therapy in the intensive care unit: a systematic review and meta‐analysis of randomized controlled trials. Critical Care Medicine 2010;38(11):2229‐41.

Koulenti 2009

Koulenti D, Lisboa T, Brun‐Buisson C, Krueger W, Macor A, Sole‐Violan J, et al. Spectrum of practice in the diagnosis of nosocomial pneumonia in patients requiring mechanical ventilation in European intensive care units. Critical Care Medicine 2009;37:2360‐8.

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6. Searching for studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Luna 2003

Luna CM,  Blanzaco D,  Niederman MS,  Matarucco W,  Baredes NC,  Desmery P,  et al. Resolution of ventilator‐associated pneumonia: prospective evaluation of the clinical pulmonary infection score as an early clinical predictor of outcome. Critical Care Medicine 2003;31(3):676‐82.

Magill 2013

Magill S, Klompas M, Balk R, Burns SM, Deutschman C, Diekema D, et al. Developing a new, national approach to surveillance for ventilator‐associated events. Critical Care Medicine 2013;41:2467‐75.

Martin‐Loeches 2015

Martin‐Loeches I, Torres A, Rinaudo M, Terraneo S, de Rosa F, Ramirez P, et al. Resistance patterns and outcomes in intensive care unit (ICU)‐acquired pneumonia. Validation of European Centre for Disease Prevention and Control (ECDC) and the Centers for Disease Control and Prevention (CDC) classification of multidrug resistant organisms. Journal of Infection 2015;70:213‐22.

Melsen 2011

Melsen WG, Rovers MM, Koeman M, Bonten MJ. Estimating the attributable mortality of ventilator‐associated pneumonia from randomized prevention studies. Critical Care Medicine 2011;39(12):2736‐42.

Melsen 2013

Melsen WG, Rovers MM, Groenwold RH, Bergmans DC, Camus C, Bauer TT, et al. Attributable mortality of ventilator‐associated pneumonia: a meta‐analysis of individual patient data from randomised prevention studies. Lancet Infectious Diseases 2013;13(8):665‐71.

Mueller 2007

Mueller EW,  Croce MA,  Boucher BA,  Hanes SD,  Wood GC,  Swanson JM,  et al. Repeat bronchoalveolar lavage to guide antibiotic duration for ventilator‐associated pneumonia. Journal of Trauma 2007;63(6):1329‐37.

Muscedere 2013

Muscedere J, Sinuff T, Heyland D, Dodek P, Keenan S, Wood G, et al. The clinical impact and preventability of ventilator‐associated conditions in critically ill patients who are mechanically ventilated. Chest 2013;144:1453‐60.

Niederman 2012

Niederman MS, Chastre J, Corkery K, Fink JB, Luyt CE, García MS. BAY41‐6551 achieves bactericidal tracheal aspirate amikacin concentrations in mechanically ventilated patients with Gram‐negative pneumonia. Intensive Care Medicine 2012;38:263‐71.

Rello 2002

Rello J, Ollendorf DA, Oster G, Vera‐Llonch M, Bellm L, Redman R, et al. Epidemiology and outcomes of ventilator‐associated pneumonia in a large US database. Chest 2002;122:2115‐21.

Restrepo 2013

Restrepo M, Peterson J, Fernandez J, Qin Z, Fisher A, Nicholson S. Comparison of the bacterial etiology of early‐onset and late‐onset ventilator‐associated pneumonia in subjects enrolled in 2 large clinical studies. Respiratory Care 2013;58:1220‐5.

RevMan 2014 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Richards 2000

Richards MJ, Edwards JR, Culver DH, Gaynes RP. Nosocomial infections in combined medical‐surgical intensive care units in the United States. Infection Control and Hospital Epidemiology 2000;21(8):510‐5.

Rotstein 2008

Rotstein C, Evans G, Born A, Grossman R, Light RB, Magder S, et al. Clinical practice guidelines for hospital‐acquired pneumonia and ventilator‐associated pneumonia in adults. Canadian Journal of Infectious Diseases and Medical Microbiology 2008;19(1):19‐53.

Safdar 2005

Safdar N, Dezfulian C, Collard HR, Saint S. Clinical and economic consequences of ventilator‐associated pneumonia: a systematic review. Critical Care Medicine 2005;33:2184‐93.

Schuetz 2012a

Schuetz P, Briel M, Christ‐Crain M, Stolz D, Bouadma L, Wolff M, et al. Procalcitonin to guide initiation and duration of antibiotic treatment in acute respiratory infections: an individual patient data meta‐analysis. Clinical Infectious Diseases 2012;55:651‐62.

Schuetz 2012b

Schuetz P, Müller B, Christ‐Crain M, Stolz D, Tamm M, Bouadma L, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database of Systematic Reviews 2012, Issue 9. [DOI: 10.1002/14651858.CD007498.pub2]

Sharpe 2015

Sharpe JP, Magnotti LJ, Weinberg JA, Swanson JM, Schroeppel TJ, Clement LP, et al. Adherence to an established diagnostic threshold for ventilator‐associated pneumonia contributes to low false‐negative rates in trauma patients. Journal of Trauma and Acute Care Surgery 2015;78(3):468‐73.

Sterne 2011

Sterne JAC, Egger M, Moher D (editors). Chapter 10: Addressing reporting biases. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Stoeppel 2014

Stoeppel CM, Eriksson EA, Hawkins K, Eastman A, Wolf S, Minei J, et al. Applicability of the National Healthcare Safety Network's surveillance definition of ventilator‐associated events in the surgical intensive care unit: a 1‐year review. Journal of Trauma and Acute Care Surgery 2014;77:934‐7.

Szakmany 2013

Szakmany T, Harrison W, Roberts H, Scholey G, Pugh R. Surveillance for ventilator‐associated pneumonia (VAP) in Wales: is HELICS the best way?. Critical Care Medicine 2013;41(Suppl 12):46.

Vincent 1995

Vincent JL,  Bihari DJ,  Suter PM,  Bruining HA,  White J,  Nicolas‐Chanoin MH,  et al. The prevalence of nosocomial infection in intensive care units in Europe. Results of the European Prevalence of Infection in Intensive Care (EPIC) Study. EPIC International Advisory Committee. JAMA 1995;274(8):639‐44.

Vincent 2009

Vincent JL,  Rello J,  Marshall J,  Silva E,  Anzueto A,  Martin CD,  et al. International study of the prevalence and outcomes of infection in intensive care units. JAMA 2009;302(21):2323‐9.

Visscher 2008

Visscher S, Schurink CA, Melsen WG, Lucas PJ, Bonten MJ. Effects of systemic antibiotic therapy on bacterial persistence in the respiratory tract of mechanically ventilated patients. Intensive Care Medicine 2008;34(4):692‐9.

Warren 2003

Warren DK, Shukla SJ, Olsen MA, Kollef MH, Hollenbeak CS, et al. Outcome and attributable cost of ventilator‐associated pneumonia among intensive care unit patients in a suburban medical center. Critical Care Medicine 2003;31:1312‐7.

Wunderink 2000

Wunderink R. Radiologic diagnosis of ventilator‐associated pneumonia. Chest 117;117(Suppl):188‐90.

Zhuo 2008

Zhuo H, Yang K, Lynch SV, Dotson RH, Glidden DV, Singh G, et al. Increased mortality of ventilated patients with endotracheal Pseudomonas aeruginosa without clinical signs of infection. Critical Care Medicine 2008;36(9):2495‐503.

Referencias de otras versiones publicadas de esta revisión

Pugh 2009

Pugh R, Grant C, Cooke RPD, Dempsey G. Short‐course versus prolonged‐course antibiotic therapy for hospital‐acquired pneumonia in critically ill adults. Cochrane Database of Systematic Reviews 2009, Issue 1. [DOI: 10.1002/14651858.CD007577]

Pugh 2011

Pugh R, Grant C, Cooke RPD, Dempsey G. Short‐course versus prolonged‐course antibiotic therapy for hospital‐acquired pneumonia in critically ill adults. Cochrane Database of Systematic Reviews 2011, Issue 10. [DOI: 10.1002/14651858.CD007577.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Capellier 2012

Methods

Multi‐centre study, based in France. Unblinded, randomised controlled trial comparing fixed durations (8‐day versus 15‐day) of antibiotic therapy for early‐onset VAP conducted between 1998 and 2002

Participants

225 adult patients in 13 ICUs (medical, surgical, trauma; 70% male; mean age 49; mean SAPS II score 39; no episodes due to NF‐GNB; no episodes due to MRSA). VAP diagnosis required 2 of the following: temperature of 38.3 °C or higher, white cell count > 10 x 109/L, excessive purulent secretions, and meeting radiological criteria. VAP diagnosis was confirmed with BAL culture of ≥ 104 cfu/ml. Early onset VAP was defined as pneumonia developing more than 24 hours but less than 8 days after onset of mechanical ventilation. Mean interval between hospital admission and intubation was 1.6 days, and between intubation and BAL (i.e. onset of VAP) 3.5 days. Bacterial pathogen identified on blood culture in 8% cases
Exclusions: prior receipt of antibiotics for suspected pneumonia, pregnancy, immunocompromise, pulmonary abscess

Interventions

116 patients were allocated to receive an 8‐day course of beta‐lactam (from choice of 3) plus 5 days of aminoglycoside antibiotic (from choice of 3); 109 patients were allocated to receive a 15‐day course of beta‐lactam plus 5 days of aminoglycoside

Outcomes

The trial was designed to show equivalence in terms of clinical cure rate

Primary outcome:

  • Clinical cure rate at day 21, a composite measure defined by absence of: death, septic shock (due to respiratory infection), need to modify antibiotic treatment, relapse (new infectious event caused by same organism as identified on original BAL, with clinical and radiological criteria as referred to above)

Secondary outcomes (with the exception of 90‐day mortality, all evaluated at day 21):

  • Secondary nosocomial infection

  • Number of days of antibiotic treatment

  • Duration of mechanical ventilation (data reported as duration of intubation)

  • Duration of ICU stay

  • Mortality (21‐day and 90‐day)

Notes

Appropriateness of initial antibiotic therapy was not reported. Mean duration of first‐line treatment was 7.8 days (SD 1.6) in the short course and 13.1 (SD 3.6) in the prolonged course group

The interval between the end of data collection and publication was 10 years

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomization is allocated in each centre by a dedicated randomisation table."

Allocation concealment (selection bias)

Unclear risk

"The assigned treatment arm is communicated by fax at the latest at D5 by the main investigator centre."

Comment: unclear to what extent participants/investigators at main centre could have access to/have foreseen allocation according to randomisation table

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Non‐blinded

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Non‐blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Data regarding exclusions not reported. No loss to 21‐day follow‐up. However, loss to 90‐day follow‐up of 14 patients (12%) in short‐course, and of 13 (12%) in prolonged‐course group. Explanation not provided and significance not explored

Selective reporting (reporting bias)

Unclear risk

Antibiotic‐free days unreported

Other bias

Unclear risk

Follow‐up at 21‐days is relatively early compared with other studies, for assessment of, e.g. duration of ICU stay. Data were collected between 1998 and 2002. Though recently published, results will reflect critical care medicine as practised a decade earlier

Chastre 2003a

Methods

The PNEUMA study, a multi‐centre study based in France. Unblinded, randomised controlled trial comparing fixed durations (8‐day versus 15‐day) of antibiotic therapy for VAP. Randomisation occurred 3 days after BAL results confirmed VAP

Participants

401 adult patients. 51 French ICUs. 72% male; mean age 61; episodes due to NF‐GNB 32.5%, MRSA 11.2%; mean SAPS II score 45; mean SOFA score 7.4 at admission. VAP was diagnosed according to the following criteria: new and persistent radiographic infiltrate, plus 1 of: purulent tracheal secretions, temperature of 38.4 °C or higher, or leukocyte count > 10,000/µL; and positive quantitative culture of 104 cfu/mL from BAL or 103 cfu/mL from PSB. Duration of mechanical ventilation prior to VAP: 13.6 days. No significant differences between groups at baseline, with the exception of significantly higher proportion of men (76.6%) in 8‐day regimen versus men in 15‐day regimen (67.6%; P value = 0.046). All patients received appropriate initial antibiotics. Exclusions included early onset pneumonia (within 5 days of commencing mechanical ventilation) in patients who had received no antimicrobial therapy in the 15 days prior to diagnosis of pneumonia, and immunocompromised state, characterised by: neutropenia, AIDS, long‐term corticosteroids or other immunosuppressant therapy

Interventions

197 patients received a fixed 8‐day course of antibiotics (chosen by treating physician); 204 patients received a 15‐day course

Outcomes

The trial was designed to demonstrate the non‐inferiority of short‐course therapy in terms of all‐cause mortality and pulmonary infection recurrence, and superiority in terms of 28‐day antibiotic‐free days. It was powered to detect a 10% difference in death and pulmonary infection recurrence, and a 20% difference in 28‐day antibiotic‐free days

The primary outcome measures were:

  • 28‐day death from any cause

  • 28‐day microbiologically confirmed pulmonary infection recurrence

  • 28‐day antibiotic‐free days

The following additional measures were reported:

  • Death from any cause (60‐day)

  • Mechanical ventilation‐free days (28‐day)

  • Number of organ failure‐free days (28‐day)

  • Evolution of parameters comprising the SOFA score and ODIN score (Day 1 to 28)

  • Clinical features relevant to pulmonary infection (fever, leukocyte count, PaO2/ FiO2 ratio, radiologic score; day 1 to 28)

  • Duration of ICU stay

  • Rate of unfavourable outcome (death, recurrence, prescription of new antibiotic therapy)

  • In‐hospital mortality

  • Percentage of emerging multi‐resistant bacteria during ITU in bronchoscopic samples collected to investigate possible recurrence

Notes

Repeat bronchoscopy was performed on the basis of fever, purulent secretions, new or progressive pulmonary infiltrates, or deterioration in respiratory or haemodynamic parameters. It was not performed routinely, e.g. on completion of 8‐day course of therapy, and consequently data regarding persistent colonisation with NF‐GNB are not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation was performed...and stratified... according to a computer‐generated random‐number table."

Allocation concealment (selection bias)

Low risk

"Randomisation performed centrally, using an interactive voice system... randomisation was not communicated to the investigators until day 8... On that day, investigators had to telephone the randomisation centre to receive the treatment assignment by fax."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"...Patients, medical and nursing staffs, and pharmacists remained blinded until [day 8]." However, importantly, no attempt was made to blind from day 8, i.e. the point from which allocation might make a significant difference

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"...Patients, medical and nursing staffs, and pharmacists remained blinded until [day 8]." Again, no attempt was made to blind from day 8, i.e. the point from which allocation might make a significant difference

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Following randomisation, 0 patients were lost to follow‐up. 1 patient was excluded from analysis following withdrawal of consent

Selective reporting (reporting bias)

Unclear risk

Study protocol not available for examination. However, outcome measures are those expected and appropriately presented within the report

Other bias

Low risk

There was a significantly higher proportion of men in the 8‐day group (76.6%) compared with the 15‐day group (67.6%; P value = 0.046). However, there were not significant differences in illness severity scores, prior duration of mechanical ventilation, prior antibiotic administration, micro‐organisms responsible for VAP, and antibiotic regimes to treat VAP

Data regarding proportions of patients who contrary to protocol did not receive a full 8‐day or 15‐day course of antibiotics are not provided. However, absolute antibiotic‐treatment days and 28‐day antibiotic‐free day data indicate significantly less antibiotic exposure as a consequence of allocation to short or prolonged‐course therapy

Fekih Hassen 2009

Methods

Single‐centre study based in Tunisia. RCT comparing fixed durations (7‐day versus 10‐day) of antibiotic therapy for VAP

Participants

Medical ICU. 30 adult patients (63% male; mean age 63 years; episodes due to NF‐GNB 72%; SAPS II 42.4). VAP (onset more than 48 hours after mechanical ventilation in ICU) diagnosis was suspected on the basis of: new and persistent radiographic infiltrate, purulent secretions, fever or deteriorating gas exchange or white cell count and confirmed on quantitative analysis of culture of endotracheal aspirate (> 104 cfu/ml) or protected distal respiratory specimens (>103 cfu/ml). Mean onset of VAP after institution of mechanical ventilation: 10 days. Initial antibiotics were appropriate in 94% of cases. No significant differences in baseline characteristics between the 2 groups

Exclusions include: second episode of pneumonia during single hospitalisation, terminal illness, failure to isolate bacterial growth

30 patients randomised from 39 patients with clinical features of VAP: 9 not enrolled because of terminal illness or failure to isolate bacteria

Interventions

14 patients randomised to receive 7‐day course of antibiotics; 16 patients to receive 16‐day course. Choice of antibiotic: on microbiology advice, taking into account whether early (up to including 5 days after commencing mechanical ventilation) or late‐onset VAP, and whether risk factors for multi‐resistant bacteria present, and modified according to culture/sensitivity results

94% patients received appropriate initial antibiotic therapy

Outcomes

Primary outcome measures were:

  • Mortality (14‐day and 28‐day)

  • 28‐day antibiotic‐free days

Power calculation is not reported

The following additional outcome measures were presented:

  • Microbiological resolution (timescale not specified)

  • CPIS resolution (days 1, 10, 14)

  • Mechanical ventilation‐free days (28‐day)

  • Recurrence: relapse or super‐infection (timescale not specified)

  • Duration of ICU stay

Notes

Data regarding protocol violations and patients lost to follow‐up not available. Unable to make contact with trial authors to request supplementary information

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Use of random number table

Allocation concealment (selection bias)

Unclear risk

Inadequately reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Inadequately reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Inadequately reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Inadequately reported

Selective reporting (reporting bias)

Unclear risk

Inadequately reported

Other bias

Low risk

No evidence of other source of bias

Kollef 2012

Methods

International multi‐centre study (56 sites in 19 countries, stratified according to region as Central and Southern America, Eastern Europe and Asia, and Western Europe, North America and Australia) conducted between April 2008 and June 2011. Double‐blinded randomised controlled trial comparing a fixed 7‐day course of doripenem, with a fixed 10‐day course of imipenem‐cilastatin

Participants

274 patients (APACHE II score > 8 and < 35, baseline CPIS > 6, prior hospitalisation or management in chronic care facility for total 5 days or more within previous 90 days) with pneumonia acquired at least 48 hours after onset of mechanical ventilation. Of these, 227 (65% male, mean age 78 years, 57% APACHE II > 15, mean SOFA score 5.8) were included in the ITT analysis. Baseline characteristics of intervention groups overall appear balanced, but statistical significance of any differences not explored. VAP diagnosed on the basis of new or worsening radiographic infiltrates and at least one of: temperature > 39 °C or < 35 °C, or an increase of temperature of > 1 °C, or white blood cell count > 10 x 109/L. Among the microbiological intention‐to‐treat group (MITT; at least 1 gram‐negative pathogen identified on BAL or mini‐BAL at density of ≥ 104 cfu/mL with an imipenem MIC < 8 μg/mL), a gram‐negative pathogen was identified in 89% cases. Within this MITT group a higher proportion of the patients receiving doripenem had either Pseudomonas aeruginosa (22% versus 11%) or Acinetobacter spp. (19% versus 11%) identified. No data presented for duration of mechanical ventilation prior to onset of VAP
Exclusions: pregnancy, administration of antibiotics for > 24 hours before enrolment, baseline presence of only MRSA or Stenotrophomonas maltophilia, ARDS or other condition which would hinder interpretation of VAP (including pulmonary contusion, pleural effusion, lung cancer, decompensated COPD, congestive cardiac failure, aspiration pneumonitis) and beta‐lactam sensitivity

Interventions

Among the ITT group, 115 patients received a 7‐day course of doripenem 1 g administered as 4‐hour infusion every 8 hours, and 112 patients a 10‐day course of imipenem‐cilastatin 1 g as a 1‐hour infusion every 8 hours. Among the subgroup meeting the MITT criteria as above, 79 received doripenem and 88 imipenem‐cilastatin

Outcomes

The study was designed to demonstrate non‐inferiority for the primary outcome measure, clinical cure (according to radiological and clinical features) at end of treatment (day 10)

The following additional outcomes were reported for the MITT group:

  • CPIS up to day 11

  • All‐cause 28‐day mortality

Notes

Study was closed prematurely because of inferior efficacy and higher mortality in 1 treatment arm

Median duration of study drug therapy 9.7 days in each group, and for active drug 7.0 days in doripenem group and 10.0 days in imipenem group

5 sites (3 in Guatemala, 1 in Germany, 1 in United States) deemed to be non‐compliant with good clinical practices, and for this reason data regarding 41 patients were excluded from analysis. Note additional data published elsewhere regarding ITT patients not included in MITT analysis (Dimopoulos 2013). However, given the intention described in the original report to restrict data analysis to MITT patients, only data from these patients are included in this review

The US Food and Drug Administration agency issued a warning regarding use of doripenem for ventilator‐associated pneumonia in 2014 (FDA 2014)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Treatment was randomised with use of a central interactive phone system."

Comment: this implies that the randomisation process is computer‐generated, but is unclear from the text

Allocation concealment (selection bias)

Low risk

"Treatment was randomised with use of a central interactive phone system."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blinded. All patients received a 7‐day infusion (whether placebo or active drug) and 10‐day infusion (whether active drug or placebo)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Double‐blinded", implying outcome assessment probably blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Outcome data are complete for the MITT subgroup of interest

Selective reporting (reporting bias)

Unclear risk

Duration of mechanical ventilation before episode of VAP not presented, which is unexpected given the intention to compare interventions in patients with late‐onset VAP

In terms of outcome data, duration of mechanical ventilation and of ICU and hospital length of stay are unreported, but possibly such outcome data would have been difficult to interpret given variation in processes at such a large number of centres

Other bias

High risk

Unfortunately, there are multiple interventions. This study is comparing not just a short course of therapy with a prolonged course, but a short course of one antibiotic administered over a long infusion time (which may be more effective against resistant bacteria), with a long course of another antibiotic administered over a short infusion time

There is a higher proportion of Pseudomonas aeruginosa and Acinetobacter spp. in the doripenem group

The timing of EOT assessment (at day 10) may have favoured patients in the 10‐day therapy group rather than the 7‐day therapy group, not having allowed for relapse to have occurred in the prolonged‐therapy group

Medina 2007

Methods

2‐centre study, based in Uruguay, conducted May 2003 to December 2006. Randomised controlled study comparing fixed short (8‐day) and long (12‐day) courses of antibiotic therapy for VAP

Participants

77 patients (medical, surgical and neurosurgical; mean age 59 years, 53% male, 63.6% episodes due to NF‐GNB, 9.1% episodes due to MRSA; median APACHE II score 21, MODS score 5, SOFA score 6) with VAP. VAP was diagnosed on the basis of: new and persistent radiographic infiltrates, 2 of temperature ≥ 38.5 ºC or < 36 ºC, leukocytes ≥ 12,000/ mm3 or ≤ 4 x 103/mm3, and BAL culture ≥ 104 cfu/ml, or positive ETA plus CPIS > 6, or micro‐organism in ≥ 2 blood cultures with identical sensitivity to tracheal secretions and in the absence of other possible infection, or positive culture of pleural fluid. Mean time after commencing mechanical ventilation before onset of VAP: 9.3 days. 68% patients had received a prior course of antibiotics. In 97% cases, initial antibiotic therapy for VAP was appropriate. There were no significant differences in baseline characteristics between treatment groups
Exclusion criteria: failure to meet clinical and microbiological criteria, decision not to institute antibiotic therapy, duration of therapy < 6 days, death before 6th day of treatment, onset of VAP within 48 hours of admission from other centre

Interventions

77 patients randomised to 8‐day treatment or 12‐day course on day 8 of antibiotic therapy. Antibiotic choice was that of the attending physician; in 75/77 (97%) cases, initial antibiotic therapy was appropriate. The most commonly used antibiotics were: cefoperazone‐sulbactam, carbapenem and other third‐generation cephalosporins. In 51% cases, antibiotic combinations were used

Outcomes

The study was designed to demonstrate the non‐inferiority of short‐course therapy in terms of clinical resolution and non‐resolution ("therapeutic failure") of VAP

The following other outcomes were also reported:

  • ITU mortality

  • VAP‐related mortality

  • Recurrence of VAP

  • Duration of mechanical ventilation

Notes

No patients were lost to follow‐up. No patient received a shorter duration of antibiotic therapy than allocated; data are incomplete for patients who had antibiotics continued beyond the allocated duration

At the time our searches were performed, this study had not been published in a peer‐reviewed journal

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Block randomisation using random number table

Allocation concealment (selection bias)

Low risk

Sealed, numbered envelopes opened sequentially

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Non‐blinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Non‐blinded study

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No patients lost to follow‐up

Selective reporting (reporting bias)

Unclear risk

Unclear. Data incomplete for patients who had antibiotics continued beyond the allocated duration

Other bias

Low risk

Study appears to be free of other sources of bias

Singh 2000

Methods

Single‐centre study from the USA. Randomised controlled trial comparing strategy of 3 days' ciprofloxacin monotherapy versus standard therapy (duration and antibiotic choice at the discretion of physician) for patients with pulmonary infiltrates but low‐probability pneumonia.

Participants

Study entry criteria: new‐onset pulmonary infiltrate associated with possible nosocomial pneumonia. Modified Clinical Pulmonary Infection Score (CPIS) < 7 on day 1 (suggesting low probability pneumonia)

81 adult medical and surgical ICU patients; 47 (58%) receiving mechanical ventilation; mean age 66.7 years; no data on sex. APACHE III score 41.8; mean CPIS 4.9. Prior mean duration of ICU stay 8.8 days and duration of mechanical ventilation 7.6 days. Chronic obstructive airways disease present in 27% cases. 5% patients were transplant recipients. With the exception of abnormal respiration (92% experimental group versus 71% standard‐therapy group, P value = 0.016), there were no significant differences in baseline characteristics between the 2 groups, though mean duration of mechanical ventilation prior to enrolment was 10 days in the short‐course group and 5 days in the standard‐therapy group. Exclusions: HIV, chemotherapy‐induced neutropenia, concurrent antibiotic administration (other than surgical prophylaxis), flouroquinolone allergy

Interventions

Randomised at day 1 of episode of possible pneumonia. Intervention group (N = 39): 3 days' ciprofloxacin monotherapy. At day 3 if CPIS < 7, antibiotics would be discontinued; if CPIS > 6, therapy would be continued, with choice of agent and duration of therapy at the discretion of treating physician, and incorporating microbiology results

Standard therapy (N = 42): choice and duration of antibiotic therapy at choice of treating physician

Outcomes

The following outcomes were reported:

  • Mortality (day 3, 14, 30)

  • Duration of ITU stay

  • Emergence of antimicrobial resistance or superinfection (day 28)

  • Duration of antibiotic therapy

  • Antimicrobial therapy cost

Power calculation was conducted according to "worse outcome" and development of antimicrobial resistance

Notes

Pathogens associated with possible HAP were incompletely presented

Outcome data for patients who were and were not mechanically ventilated are not reported separately

For patients allocated to the short‐course therapy, 0 patients with CPIS < 7 at day 3 had antibiotics continued beyond 72 hours

A significant decrease in duration of therapy in the "standard therapy" group was observed with time (P value = 0.0001), thought to be a result of the unblinded nature of the study. The study was terminated by institutional review board as it was deemed "unethical to continue study"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation process not described

Allocation concealment (selection bias)

Unclear risk

Randomisation/concealment not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Non‐blinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Non‐blinded study

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing outcome data

Selective reporting (reporting bias)

High risk

Microbiological data are partially reported. The components of the composite measure of "emergence of antimicrobial resistance or superinfection" are not reported

Other bias

High risk

Stopped early by institutional review board. Results for patients managed in "experimental" group appear to have influenced management of patients subsequently allocated to "standard therapy group"

APACHE II score: Acute Physiology and Chronic Health Evaluation II score
ARDS: acute respiratory distress syndrome
BAL: broncho‐alveolar lavage
cfu/ml: colony‐forming units per millilitre
COPD: chronic obstructive pulmonary disease
CPIS: clinical pulmonary infection score
DNR order: do not resuscitate order
EOT: end‐of‐treatment
ETA: endo‐tracheal aspirate
FiO2: fraction of inspired oxygen (in a gas mixture)
HAP: hospital‐acquired pneumonia
ICU: intensive care unit
ITT: intention‐to‐treat
ITU: intensive therapy unit
MITT: microbiological intention‐to‐treat
MODS: multiple organ dysfunction score
MRSA: methicillin‐resistant Staphylococcus aureus
NF‐GNB: non‐fermenting Gram‐negative bacilli
ODIN: organ dysfunction and/or infection score
PaO2: partial pressure of oxygen
PSB: protected specimen brush
RCT: randomised controlled trial
SAPS II: Simplified Acute Physiology Score II
SD: standard deviation
SOFA score: Sequential Organ Failure Assessment score
VAP: ventilator‐associated pneumonia

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Bouadma 2010

The PRORATA randomised controlled study investigating the utility of a procalcitonin‐guided (PCT‐guided) algorithm for antibiotic administration for intensive care patients (34% with hospital‐acquired pneumonia, of whom approximately 2/3 were mechanically ventilated). Mean duration of antibiotic therapy was significantly lower in the PCT group among patients with VAP. However, the study was excluded because interventions did not include a fixed duration of therapy

Cai 2001

A randomised controlled study investigating diagnostic strategies (quantitative versus qualitative culture of respiratory tract specimens) on outcome from VAP

CCCTG 2006

This 2‐by‐2 factorial randomised controlled study aiming to compare: i. invasive and non‐invasive diagnostic strategies for diagnosis of VAP, and ii. initial empiric treatment with meropenem or meropenem plus ciprofloxacin for suspected VAP. There was no significant difference in number of antibiotic‐free days between groups of patients allocated to invasive or non‐invasive strategies

Chastre 2003b

Data subsequently published in full (Chastre 2003a)

Fagon 2000

This was a randomised study designed to compare strategies for diagnosis and selection of initial antimicrobial therapy of VAP, not duration of therapy. For groups of patients randomised to both invasive and non‐invasive diagnostic strategies, recommended duration of therapy in the presence of positive respiratory culture (i.e. confirmed pneumonia) was 14 days

Hochreiter 2009

This was a randomised controlled study comparing a strategy using serial PCT measurement to guide discontinuation of antibiotic therapy for treatment of sepsis in 110 surgical intensive care patients with standard therapy. Of these patients, 43 had "pneumonia"; it is unclear what proportion had nosocomial or community‐acquired pneumonia, and what proportion of patients was receiving mechanical ventilation at time of diagnosis. Duration of antibiotic therapy was significantly shorter in the intervention (PCT‐guided) group; however, the study was excluded because interventions did not include a fixed duration of therapy

Ibrahim 2001

Non‐randomised before‐and‐after study investigating a clinical guideline regarding initial treatment and subsequent discontinuation of antibiotic therapy

Kim 2009

Data subsequently published in full (Kim 2012)

Kim 2012

This was a single‐centre RCT investigating effectiveness of an antibiotic de‐escalation protocol for intensive care patients with HAP. 109 medical ICU patients aged 18 years and over with HAP (defined according to ACCP clinical and radiographic criteria) were randomised to the de‐escalation (DE) protocol (N = 54) or non de‐escalation (NDE) protocol (N = 55). There was no significant difference in duration of antibiotic therapy between intervention groups, and the study was excluded on the basis that fixed courses of therapy were not used

Kollef 2005

Prospective observational study investigating application of previously described clinical guideline (Ibrahim 2001) for patients with suspected VAP, but negative quantitative BAL results

Labelle 2012

This randomised controlled study attempted to evaluate the feasibility (and outcomes) with a treatment goal of 5 or 7 days for pneumonia in intensive care. It was excluded on consideration of a conference proceedings abstract because fixed courses of therapy appear not to have been utilised and duration of therapy was not significantly different between groups

Maldonado‐Ortiz 2004

This multi‐centre randomised controlled study enrolled 65 patients in a study intended to evaluate a strategy of early discontinuation of empirical antibiotic therapy: it has been published in abstract form only. 31 patients were allocated to early (< 8 days) discontinuation of empirical therapy versus 34 patients to late discontinuation (> 9 days). Patient characteristics and diagnostic criteria were not described. Outcome data presented in the abstract were inadequate for the study's inclusion in this review, and contact with a trial author did not yield any further information. Furthermore, a highly significant risk of bias was identified: antibiotic discontinuation at day 8 was higher (70.6%) among patients allocated to late discontinuation than among patients allocated to early discontinuation (67.7%)

Micek 2004

This RCT sought to evaluate the effectiveness of an antibiotic discontinuation policy for suspected VAP. Mean duration of antibiotic therapy was lower in the intervention group (6 days) compared with the standard therapy group (8 days). However, the study was excluded because interventions did not include a fixed duration of therapy

Nobre 2008

This was a randomised controlled study comparing a strategy using serial measurements of procalcitonin (PCT) to guide cessation of antibiotic therapy with standard therapy in critically ill patients with sepsis or septic shock. 79 patients were randomised, of whom a total of 47 patients had sepsis of pulmonary origin. A high proportion of patients (68%) had community‐acquired sepsis. A significant proportion of patients in the PCT group (19%) had the protocol overridden to receive a longer course of antibiotics than advised by the algorithm. On intention‐to‐treat analysis, the difference in antibiotic days between control and intervention groups for all patients was not significant. The study was excluded because interventions did not include a fixed duration of therapy

Peery 2001

This was a randomised study comparing diagnostic strategies for suspected VAP. It was not designed to investigate duration of therapy; there were no protocols to guide duration of antibiotic therapy

Pontet 2007

This RCT compared use of a PCT‐guided algorithm with standard therapy in antibiotic treatment of VAP; the study was excluded because interventions did not include a fixed duration of therapy

Rello 2004

This was an observational rather than a randomised controlled study. It investigated outcomes following introduction of a "De‐escalation" strategy which incorporated the initial administration of broad spectrum antibiotics and subsequent simplification of antibiotic treatment with culture results: 1. changing to monotherapy in absence ofPseudomonas sp; 2. shortening therapy to < 5 days if culture negative and > 48 hours of defervescence; 3. changing from a broad to narrow spectrum agent on basis of culture results

Sanchez‐Nieto 1998

A randomised clinical trial comparing the effects of an invasive quantitative diagnostic strategy versus a non‐invasive strategy on management of and outcome from suspected VAP

Schroeder 2007

This was a RCT comparing a procalcitonin‐guided antibiotic discontinuation strategy with standard treatment for surgical intensive care patients with severe sepsis. The study was excluded because interventions did not include a fixed duration of therapy

Singh 1998

Data subsequently published in full (Singh 2000)

Sole 2000

A RCT to evaluate invasive versus non‐invasive diagnostic methods on outcome from VAP

Stolz 2009a

The ProVAP study was a randomised controlled trial comparing a PCT‐guided algorithm with standard therapy for ICU patients with VAP. Duration of antibiotic therapy was lower in the PCT‐guided group (median 15 days versus 10 days in standard therapy group). The study was excluded because interventions did not include a fixed duration of therapy

Stolz 2009b

Data subsequently published in full (Stolz 2009a)

Svoboda 2007

RCT evaluating PCT‐guided strategies in the management of septic illness after multiple trauma or major surgery. The study was excluded because interventions did not include a fixed duration of therapy

Wolff 2003

Data subsequently published in full (Chastre 2003a)

BAL: broncho‐alveolar lavage
CPIS: clinical pulmonary infection score
HAP: hospital‐acquired pneumonia
ICU: intensive care unit
ITU: intensive therapy unit
PCT: procalcitonin
RCT: randomised controlled trial
VAP: ventilator‐associated pneumonia

Characteristics of ongoing studies [ordered by study ID]

NCT00410527

Trial name or title

'Effectiveness of short‐course versus standard antibiotic therapy in ICU patients'

Methods

Randomised controlled trial

Participants

ICU patients (not necessarily mechanically ventilated) aged 18 years and older with pulmonary infiltrates on chest radiography, but low probability of pneumonia according to CPIS of 6 or less

Interventions

3‐day course of meropenem versus standard course (8 days or more) of antibiotic therapy

Outcomes

Identification of resistant organisms in respiratory specimen; duration of antibiotic therapy; duration of ICU and hospital length of stay

Starting date

August 2006

Contact information

Not available

Notes

https://clinicaltrials.gov/ct2/show/study/NCT00410527

NCT01554657

Trial name or title

'Five versus seven day antibiotic course for the treatment of pneumonia in the Intensive Care Unit'

Methods

Randomised controlled trial

Participants

Medical and surgical patients aged 18 years and over with pneumonia

Interventions

5 days of antibiotic therapy versus 7 days of antibiotic therapy

Outcomes

Length of antibiotic therapy, 28‐day mortality, 28‐day hospital length of stay, 28‐day development of C. difficile‐related diarrhoea, identification of MDR bacteria from lower respiratory tract specimen

Starting date

January 2011

Contact information

Not available

Notes

https://clinicaltrials.gov/ct2/show/study/NCT01554657

NCT01994980

Trial name or title

'Duration of antibiotic treatment for early VAP (Date) trial'

Methods

Randomised controlled trial

Participants

Surgical patients aged 18 years and older with VAP, defined according to CPIS and BAL threshold culture

Interventions

4 days of antibiotic therapy versus 8 days of antibiotic therapy

Outcomes

Clinical outcomes (CPIS); microbiological outcome (BAL culture); biomarker response (procalcitonin)

Starting date

December 2013

Contact information

Maria Rodil, Denver Medical Health Centre, Denver, Colorado, United States 80204

[email protected]

Notes

https://clinicaltrials.gov/ct2/show/NCT01994980

BAL: broncho‐alveolar lavage
CPIS: Clinical Pulmonary Infection Score
CRP: C‐reactive protein
ICU: intensive care unit
MDR: multi‐drug resistant pathogen
PCT: procalcitonin
SIRS: systemic inflammatory response syndrome
VAP: ventilator‐associated pneumonia

Data and analyses

Open in table viewer
Comparison 1. Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 28‐day mortality Show forest plot

3

598

Odds Ratio (M‐H, Random, 95% CI)

1.18 [0.77, 1.80]

Analysis 1.1

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 1 28‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 1 28‐day mortality.

1.1 NF‐GNB

2

179

Odds Ratio (M‐H, Random, 95% CI)

0.95 [0.39, 2.27]

1.2 MRSA

1

42

Odds Ratio (M‐H, Random, 95% CI)

1.28 [0.32, 5.09]

1.3 Non NF‐GNB/MRSA

1

232

Odds Ratio (M‐H, Random, 95% CI)

1.65 [0.73, 3.73]

1.4 Unspecified organism

2

145

Odds Ratio (M‐H, Random, 95% CI)

1.22 [0.53, 2.78]

2 Recurrence of pneumonia Show forest plot

4

733

Odds Ratio (M‐H, Random, 95% CI)

1.41 [0.94, 2.12]

Analysis 1.2

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 2 Recurrence of pneumonia.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 2 Recurrence of pneumonia.

2.1 NF‐GNB

2

176

Odds Ratio (M‐H, Random, 95% CI)

2.18 [1.14, 4.16]

2.2 MRSA

2

49

Odds Ratio (M‐H, Random, 95% CI)

1.56 [0.12, 19.61]

2.3 Non NF‐GNB/MRSA

3

478

Odds Ratio (M‐H, Random, 95% CI)

1.17 [0.64, 2.14]

2.4 Unspecified organism

1

30

Odds Ratio (M‐H, Random, 95% CI)

1.17 [0.14, 9.59]

3 28‐day antibiotic‐free days Show forest plot

2

431

Mean Difference (IV, Random, 95% CI)

4.02 [2.26, 5.78]

Analysis 1.3

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 3 28‐day antibiotic‐free days.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 3 28‐day antibiotic‐free days.

3.1 NF‐GNB

1

127

Mean Difference (IV, Random, 95% CI)

4.5 [2.25, 6.75]

3.2 MRSA

1

42

Mean Difference (IV, Random, 95% CI)

8.0 [4.14, 11.86]

3.3 Non NF‐GNB/ MRSA

1

232

Mean Difference (IV, Random, 95% CI)

3.70 [2.09, 5.31]

3.4 Unspecified organism

1

30

Mean Difference (IV, Random, 95% CI)

2.3 [1.03, 3.57]

4 ITU mortality Show forest plot

2

107

Odds Ratio (M‐H, Random, 95% CI)

0.85 [0.37, 1.91]

Analysis 1.4

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 4 ITU mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 4 ITU mortality.

5 In‐hospital mortality Show forest plot

1

401

Odds Ratio (M‐H, Random, 95% CI)

1.09 [0.71, 1.68]

Analysis 1.5

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 5 In‐hospital mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 5 In‐hospital mortality.

5.1 NF‐GNB

1

127

Odds Ratio (M‐H, Random, 95% CI)

0.75 [0.36, 1.53]

5.2 MRSA

1

42

Odds Ratio (M‐H, Random, 95% CI)

1.47 [0.43, 4.95]

5.3 Non NF‐GNB/MRSA

1

232

Odds Ratio (M‐H, Random, 95% CI)

1.32 [0.72, 2.42]

6 21‐day mortality Show forest plot

1

225

Odds Ratio (M‐H, Random, 95% CI)

1.05 [0.41, 2.69]

Analysis 1.6

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 6 21‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 6 21‐day mortality.

7 60‐day mortality Show forest plot

1

401

Odds Ratio (M‐H, Fixed, 95% CI)

0.86 [0.55, 1.35]

Analysis 1.7

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 7 60‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 7 60‐day mortality.

7.1 NF‐GNB

1

127

Odds Ratio (M‐H, Fixed, 95% CI)

0.74 [0.35, 1.54]

7.2 MRSA

1

42

Odds Ratio (M‐H, Fixed, 95% CI)

1.0 [0.29, 3.47]

7.3 Non NF‐GNB/ MRSA

1

232

Odds Ratio (M‐H, Fixed, 95% CI)

0.93 [0.49, 1.76]

8 90‐day mortality Show forest plot

1

198

Odds Ratio (M‐H, Random, 95% CI)

0.99 [0.49, 1.99]

Analysis 1.8

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 8 90‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 8 90‐day mortality.

9 Clinical resolution Show forest plot

3

472

Odds Ratio (M‐H, Random, 95% CI)

0.75 [0.49, 1.15]

Analysis 1.9

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 9 Clinical resolution.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 9 Clinical resolution.

9.1 NF‐GNB

1

55

Odds Ratio (M‐H, Random, 95% CI)

0.48 [0.16, 1.48]

9.2 Unspecified organism

3

417

Odds Ratio (M‐H, Random, 95% CI)

0.81 [0.51, 1.28]

10 Relapse of pneumonia Show forest plot

2

626

Odds Ratio (M‐H, Random, 95% CI)

1.70 [0.97, 2.97]

Analysis 1.10

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 10 Relapse of pneumonia.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 10 Relapse of pneumonia.

10.1 NF‐GNB

1

127

Odds Ratio (M‐H, Random, 95% CI)

2.08 [0.92, 4.70]

10.2 MRSA

1

42

Odds Ratio (M‐H, Random, 95% CI)

0.71 [0.14, 3.64]

10.3 Non NF‐GNB/MRSA

1

232

Odds Ratio (M‐H, Random, 95% CI)

1.41 [0.51, 3.92]

10.4 Unspecified organism

1

225

Odds Ratio (M‐H, Random, 95% CI)

2.92 [0.58, 14.78]

11 Subsequent infection due to resistant organism Show forest plot

1

110

Odds Ratio (M‐H, Fixed, 95% CI)

0.44 [0.21, 0.95]

Analysis 1.11

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 11 Subsequent infection due to resistant organism.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 11 Subsequent infection due to resistant organism.

12 Duration of ICU stay Show forest plot

3

656

Mean Difference (IV, Random, 95% CI)

0.15 [1.00, 1.29]

Analysis 1.12

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 12 Duration of ICU stay.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 12 Duration of ICU stay.

12.1 NF‐GNB

1

127

Mean Difference (IV, Random, 95% CI)

0.90 [‐5.40, 7.20]

12.2 MRSA

1

42

Mean Difference (IV, Random, 95% CI)

2.90 [‐8.39, 14.19]

12.3 Non NF‐GNB/MRSA

2

457

Mean Difference (IV, Random, 95% CI)

0.45 [‐1.02, 1.92]

12.4 Unspecified organism

1

30

Mean Difference (IV, Random, 95% CI)

‐1.60 [‐4.61, 1.41]

13 Duration of hospital stay Show forest plot

1

30

Mean Difference (IV, Fixed, 95% CI)

‐1.0 [‐4.11, 2.11]

Analysis 1.13

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 13 Duration of hospital stay.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 13 Duration of hospital stay.

14 Duration of mechanical ventilation Show forest plot

3

332

Mean Difference (IV, Random, 95% CI)

0.02 [‐0.51, 0.54]

Analysis 1.14

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 14 Duration of mechanical ventilation.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 14 Duration of mechanical ventilation.

Open in table viewer
Comparison 2. Discontinuation of antibiotics according to Clinical Pulmonary Infection Score

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 30‐day mortality Show forest plot

1

81

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.10, 1.03]

Analysis 2.1

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 1 30‐day mortality.

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 1 30‐day mortality.

2 Episodes of superinfection or antimicrobial resistance Show forest plot

1

81

Odds Ratio (M‐H, Fixed, 95% CI)

0.29 [0.09, 0.92]

Analysis 2.2

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 2 Episodes of superinfection or antimicrobial resistance.

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 2 Episodes of superinfection or antimicrobial resistance.

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 1

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 1 28‐day mortality.
Figuras y tablas -
Analysis 1.1

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 1 28‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 2 Recurrence of pneumonia.
Figuras y tablas -
Analysis 1.2

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 2 Recurrence of pneumonia.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 3 28‐day antibiotic‐free days.
Figuras y tablas -
Analysis 1.3

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 3 28‐day antibiotic‐free days.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 4 ITU mortality.
Figuras y tablas -
Analysis 1.4

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 4 ITU mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 5 In‐hospital mortality.
Figuras y tablas -
Analysis 1.5

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 5 In‐hospital mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 6 21‐day mortality.
Figuras y tablas -
Analysis 1.6

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 6 21‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 7 60‐day mortality.
Figuras y tablas -
Analysis 1.7

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 7 60‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 8 90‐day mortality.
Figuras y tablas -
Analysis 1.8

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 8 90‐day mortality.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 9 Clinical resolution.
Figuras y tablas -
Analysis 1.9

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 9 Clinical resolution.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 10 Relapse of pneumonia.
Figuras y tablas -
Analysis 1.10

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 10 Relapse of pneumonia.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 11 Subsequent infection due to resistant organism.
Figuras y tablas -
Analysis 1.11

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 11 Subsequent infection due to resistant organism.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 12 Duration of ICU stay.
Figuras y tablas -
Analysis 1.12

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 12 Duration of ICU stay.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 13 Duration of hospital stay.
Figuras y tablas -
Analysis 1.13

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 13 Duration of hospital stay.

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 14 Duration of mechanical ventilation.
Figuras y tablas -
Analysis 1.14

Comparison 1 Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP, Outcome 14 Duration of mechanical ventilation.

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 1 30‐day mortality.
Figuras y tablas -
Analysis 2.1

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 1 30‐day mortality.

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 2 Episodes of superinfection or antimicrobial resistance.
Figuras y tablas -
Analysis 2.2

Comparison 2 Discontinuation of antibiotics according to Clinical Pulmonary Infection Score, Outcome 2 Episodes of superinfection or antimicrobial resistance.

Summary of findings for the main comparison. Should short‐course antibiotic therapy versus prolonged‐course antibiotic therapy be used in critically ill patients with hospital‐acquired pneumonia?

Should short‐course antibiotic therapy versus prolonged‐course antibiotic therapy be used in critically ill patients with hospital‐acquired pneumonia?

Patient or population: hospital‐acquired pneumonia
Settings: intensive care
Intervention: short‐course antibiotic therapy
Comparison: prolonged‐course antibiotic therapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Prolonged‐course antibiotic therapy

Short‐course antibiotic therapy

Mortality
Follow‐up: 28 days

175 per 1000

201 per 1000
(141 to 277)

OR 1.18
(0.77 to 1.8)

598
(3 studies)

⊕⊕⊕⊝
moderate1

Mortality NF‐GNB
Follow‐up: 28 days

265 per 1000

255 per 1000
(123 to 450)

OR 0.95
(0.39 to 2.27)

179
(2 studies)

⊕⊕⊝⊝
low1,2

Mortality MRSA
Follow‐up: 28 days

238 per 1000

286 per 1000
(91 to 614)

OR 1.28
(0.32 to 5.09)

42
(1 study)

⊕⊕⊕⊝
moderate1

Recurrence of pneumonia
Clinical and/or microbiological criteria

180 per 1000

237 per 1000
(171 to 318)

OR 1.41
(0.94 to 2.12)

733
(19 studies)

⊕⊕⊝⊝
low1,3

Recurrence of pneumonia NF‐GNB
Clinical and/or microbiological criteria

247 per 1000

417 per 1000
(272 to 577)

OR 2.18
(1.14 to 4.16)

176
(2 studies)

⊕⊕⊕⊝
moderate1

Recurrence of pneumonia MRSA
Clinical and/or microbiological criteria

370 per 1000

479 per 1000
(66 to 920)

OR 1.56
(0.12 to 19.61)

49
(2 studies)

⊕⊕⊕⊝
moderate1

28‐day antibiotic‐free days
Follow‐up: 28 days

The mean 28‐day antibiotic free days in the intervention groups was
4.02 higher
(2.26 to 5.78 higher)

431
(2 studies)

⊕⊕⊝⊝
low1,4

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; MRSA: methicillin‐resistant Staphylococcus aureus; NF‐GNB: non‐fermenting Gram‐negative bacilli; OR: odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Low total number of events.
2Kollef et al. Multiple interventions (short versus prolonged duration, doripenem versus imipenem, extended versus standard infusion), protocol violations at several centres and premature cessation of study.
3Differences in duration of mechanical ventilation prior to episode of pneumonia and differences in bacterial aetiology.
4Presumed differences in administration of antibiotics (outside of context of study) between studies.

Figuras y tablas -
Summary of findings for the main comparison. Should short‐course antibiotic therapy versus prolonged‐course antibiotic therapy be used in critically ill patients with hospital‐acquired pneumonia?
Comparison 1. Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 28‐day mortality Show forest plot

3

598

Odds Ratio (M‐H, Random, 95% CI)

1.18 [0.77, 1.80]

1.1 NF‐GNB

2

179

Odds Ratio (M‐H, Random, 95% CI)

0.95 [0.39, 2.27]

1.2 MRSA

1

42

Odds Ratio (M‐H, Random, 95% CI)

1.28 [0.32, 5.09]

1.3 Non NF‐GNB/MRSA

1

232

Odds Ratio (M‐H, Random, 95% CI)

1.65 [0.73, 3.73]

1.4 Unspecified organism

2

145

Odds Ratio (M‐H, Random, 95% CI)

1.22 [0.53, 2.78]

2 Recurrence of pneumonia Show forest plot

4

733

Odds Ratio (M‐H, Random, 95% CI)

1.41 [0.94, 2.12]

2.1 NF‐GNB

2

176

Odds Ratio (M‐H, Random, 95% CI)

2.18 [1.14, 4.16]

2.2 MRSA

2

49

Odds Ratio (M‐H, Random, 95% CI)

1.56 [0.12, 19.61]

2.3 Non NF‐GNB/MRSA

3

478

Odds Ratio (M‐H, Random, 95% CI)

1.17 [0.64, 2.14]

2.4 Unspecified organism

1

30

Odds Ratio (M‐H, Random, 95% CI)

1.17 [0.14, 9.59]

3 28‐day antibiotic‐free days Show forest plot

2

431

Mean Difference (IV, Random, 95% CI)

4.02 [2.26, 5.78]

3.1 NF‐GNB

1

127

Mean Difference (IV, Random, 95% CI)

4.5 [2.25, 6.75]

3.2 MRSA

1

42

Mean Difference (IV, Random, 95% CI)

8.0 [4.14, 11.86]

3.3 Non NF‐GNB/ MRSA

1

232

Mean Difference (IV, Random, 95% CI)

3.70 [2.09, 5.31]

3.4 Unspecified organism

1

30

Mean Difference (IV, Random, 95% CI)

2.3 [1.03, 3.57]

4 ITU mortality Show forest plot

2

107

Odds Ratio (M‐H, Random, 95% CI)

0.85 [0.37, 1.91]

5 In‐hospital mortality Show forest plot

1

401

Odds Ratio (M‐H, Random, 95% CI)

1.09 [0.71, 1.68]

5.1 NF‐GNB

1

127

Odds Ratio (M‐H, Random, 95% CI)

0.75 [0.36, 1.53]

5.2 MRSA

1

42

Odds Ratio (M‐H, Random, 95% CI)

1.47 [0.43, 4.95]

5.3 Non NF‐GNB/MRSA

1

232

Odds Ratio (M‐H, Random, 95% CI)

1.32 [0.72, 2.42]

6 21‐day mortality Show forest plot

1

225

Odds Ratio (M‐H, Random, 95% CI)

1.05 [0.41, 2.69]

7 60‐day mortality Show forest plot

1

401

Odds Ratio (M‐H, Fixed, 95% CI)

0.86 [0.55, 1.35]

7.1 NF‐GNB

1

127

Odds Ratio (M‐H, Fixed, 95% CI)

0.74 [0.35, 1.54]

7.2 MRSA

1

42

Odds Ratio (M‐H, Fixed, 95% CI)

1.0 [0.29, 3.47]

7.3 Non NF‐GNB/ MRSA

1

232

Odds Ratio (M‐H, Fixed, 95% CI)

0.93 [0.49, 1.76]

8 90‐day mortality Show forest plot

1

198

Odds Ratio (M‐H, Random, 95% CI)

0.99 [0.49, 1.99]

9 Clinical resolution Show forest plot

3

472

Odds Ratio (M‐H, Random, 95% CI)

0.75 [0.49, 1.15]

9.1 NF‐GNB

1

55

Odds Ratio (M‐H, Random, 95% CI)

0.48 [0.16, 1.48]

9.2 Unspecified organism

3

417

Odds Ratio (M‐H, Random, 95% CI)

0.81 [0.51, 1.28]

10 Relapse of pneumonia Show forest plot

2

626

Odds Ratio (M‐H, Random, 95% CI)

1.70 [0.97, 2.97]

10.1 NF‐GNB

1

127

Odds Ratio (M‐H, Random, 95% CI)

2.08 [0.92, 4.70]

10.2 MRSA

1

42

Odds Ratio (M‐H, Random, 95% CI)

0.71 [0.14, 3.64]

10.3 Non NF‐GNB/MRSA

1

232

Odds Ratio (M‐H, Random, 95% CI)

1.41 [0.51, 3.92]

10.4 Unspecified organism

1

225

Odds Ratio (M‐H, Random, 95% CI)

2.92 [0.58, 14.78]

11 Subsequent infection due to resistant organism Show forest plot

1

110

Odds Ratio (M‐H, Fixed, 95% CI)

0.44 [0.21, 0.95]

12 Duration of ICU stay Show forest plot

3

656

Mean Difference (IV, Random, 95% CI)

0.15 [1.00, 1.29]

12.1 NF‐GNB

1

127

Mean Difference (IV, Random, 95% CI)

0.90 [‐5.40, 7.20]

12.2 MRSA

1

42

Mean Difference (IV, Random, 95% CI)

2.90 [‐8.39, 14.19]

12.3 Non NF‐GNB/MRSA

2

457

Mean Difference (IV, Random, 95% CI)

0.45 [‐1.02, 1.92]

12.4 Unspecified organism

1

30

Mean Difference (IV, Random, 95% CI)

‐1.60 [‐4.61, 1.41]

13 Duration of hospital stay Show forest plot

1

30

Mean Difference (IV, Fixed, 95% CI)

‐1.0 [‐4.11, 2.11]

14 Duration of mechanical ventilation Show forest plot

3

332

Mean Difference (IV, Random, 95% CI)

0.02 [‐0.51, 0.54]

Figuras y tablas -
Comparison 1. Short (fixed)‐course antibiotic therapy versus prolonged‐course antibiotic therapy for HAP
Comparison 2. Discontinuation of antibiotics according to Clinical Pulmonary Infection Score

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 30‐day mortality Show forest plot

1

81

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.10, 1.03]

2 Episodes of superinfection or antimicrobial resistance Show forest plot

1

81

Odds Ratio (M‐H, Fixed, 95% CI)

0.29 [0.09, 0.92]

Figuras y tablas -
Comparison 2. Discontinuation of antibiotics according to Clinical Pulmonary Infection Score