Scolaris Content Display Scolaris Content Display

Вмешательства при некротизирующих инфекциях мягких тканей у взрослых

Contraer todo Desplegar todo

Referencias

Bulger 2014 {published data only}

Bulger EM, Maier RV, Sperry J, Joshi M, Henry S, Moore FA, et al. A novel drug for treatment of necrotizing soft‐tissue infections: A randomized clinical trial. JAMA Surgery 2014;149(6):528‐36. [CENTRAL: CN‐00995333; PUBMED: 24740134]CENTRAL

Madsen 2017 {published data only}

Madsen MB, Hjortrup PB, Hansen MB, Lange T, Norrby‐Teglund A, Hyldegaard O, et al. Immunoglobulin G for patients with necrotising soft tissue infection (INSTINCT): a randomised, blinded, placebo‐controlled trial. Intensive Care Medicine 2017;43(11):1585‐93. [CENTRAL: CN‐01365445; DOI: 10.1007/s00134‐017‐4786‐0; PUBMED: 28421246]CENTRAL

Vick‐Fragoso 2009 {published data only}

Vick‐Fragoso RG, Hernández‐Oliva J, Cruz‐Alcázar CF, Amábile‐Cuevas P, Arvis P, Reimnitz JR, et al. Efficacy and safety of sequential intravenous/oral moxifloxacin vs intravenous/oral amoxicillin/clavulanate for complicated skin and skin structure infections. Infection 2009;37(5):407‐17. [CENTRAL: CN‐00752976; PUBMED: 19768381]CENTRAL

Abbar 2014 {published data only}

Abbar M, Janane A, Dakkak Y, Ghadouane M, Ameur A. Combination of hyperbaric oxygen therapy and surgical debridement in management of fournier's gangrene. 34th Congress of the Societe Internationale d'Urologie, SIU 2014 Glasgow United Kingdom. Urology 2014;84(4 Suppl 1):S236‐7. [EMBASE: 71654403]CENTRAL

Danino 2010 {published data only}

Danino AM, Guberman S, Mondie JM, Jebrane A, Servant JM. Calcium polyuronate dressing supplemented with zinc and manganese (trionic) in necrotizing dermohypodermitis of the extremities: a randomized multicentre study. Annals of Burns & Fire Disasters 2010;23(2):95‐101. [PUBMED: 21991205]CENTRAL

Fass 1989 {published data only}

Fass RJ, Plouffe JF, Russell JA. Intravenous/oral ciprofloxacin versus ceftazidime in the treatment of serious infections. American Journal of Medicine 1989;87(5A):164S‐8S. [PUBMED: 2589361]CENTRAL

Huang 2006 {published data only}

Huang WS, Hsieh SC, Hsieh CS, Schoung JY, Huang T. Use of vacuum‐assisted wound closure to manage limb wounds in patients suffering from acute necrotizing fasciitis. Asian Journal of Surgery 2006;29(3):135‐9. [PUBMED: 16877210]CENTRAL

Linner 2014 {published data only}

Linner A, Darenberg J, Sjolin J, Henriques‐Normark B, Norrby‐Teglund A. Clinical efficacy of polyspecific intravenous immunoglobulin therapy in patients with streptococcal toxic shock syndrome: A comparative observational study. Clinical Infectious Diseases 2014;59(6):851‐7. [PUBMED: 24928291]CENTRAL

Matejec 2013 {published data only}

Matejec R, Kayser F, Schmal F, Uhle F, Bodeker RH, Maxeiner H, et al. Effects of corticotropin‐releasing hormone on proopiomelanocortin derivatives and monocytic HLA‐DR expression in patients with septic shock. Peptides 2013;47:133‐41. [PUBMED: 23891702]CENTRAL

Morelli 2008 {published data only}

Morelli A, Lange M, Ertmer C, Dunser M, Rehberg S, Bachetoni A, et al. Short‐term effects of phenylephrine on systemic and regional hemodynamics in patients with septic shock: a crossover pilot study. Shock 2008;29(4):446‐51. [PUBMED: 17885646]CENTRAL

Norrby‐Teglund 2009 {published data only}

Norrby‐Teglund A. Managing group A streptococcal infection: the role of IVIg. 19th European Congress of Clinical Microbiology and Infectious Diseases, Helsinki Finland. Clinical Microbiology and Infection 2009;15:S50. [EMBASE: 70070333]CENTRAL

Solomkin 1986 {published data only}

Solomkin JS, Cocchetto DM. Ceftazidime versus tobramycin plus ticarcillin in the treatment of soft‐tissue infections. Clinical Therapeutics 1986;9(1):123‐34. [PUBMED: 3545476]CENTRAL

Souyri 2008 {published data only}

Souyri C, Olivier P, Grolleau S, Lapeyre‐Mestre M, French Network of Pharmacovigilance Centres. Severe necrotizing soft‐tissue infections and nonsteroidal anti‐inflammatory drugs. Clinical & Experimental Dermatology 2008;33(3):249‐55. [PUBMED: 18261144]CENTRAL

Subrahmanyam 2004 {published data only}

Subrahmanyam M, Ugane SP. Honey dressing beneficial in treatment of Fournier's gangrene. Indian Journal of Surgery 2004;66(2):75‐7. [CENTRAL: CN‐00645330]CENTRAL

Wong 2004 {published data only}

Wong CH, Khin LW, Heng KS, Tan KC, Low CO. The LRINEC (Laboratory Risk Indicator for Necrotizing Fasciitis) score: a tool for distinguishing necrotizing fasciitis from other soft tissue infections. Critical Care Medicine 2004;32(7):1535‐41. [PUBMED: 15241098]CENTRAL

Xu 2015 {published data only}

Xu X‐P, Cai X‐H, Zhang L, Li X‐H, Liu Z‐H, Pan Y, et al. Clinical efficacy of vacuum sealing drainage in treatment of fournier's gangrene. World Chinese Journal of Digestology 2015;23(2):348‐52. CENTRAL

Darenberg 2003 {published data only}

Darenberg J, Ihendyane N, Sjolin J, Aufwerber E, Haidl S, Follin P, et al. Intravenous immunoglobulin G therapy in streptococcal toxic shock syndrome: a European randomized, double‐blind, placebo‐controlled trial. Clinical Infectious Diseases 2003;37(3):333‐40. [PUBMED: 12884156]CENTRAL

Tally 1986 {published data only}

Tally FP, Kellum JM, Ho JL, O'Donnell TF, Barza M, Gorbach SL. Randomized prospective study comparing moxalactam and cefoxitin with or without tobramycin for the treatment of serious surgical infections. Antimicrobial Agents and Chemotherapy 1986;29(2):244‐9. [PUBMED: 3717931]CENTRAL

NCT02469857 {unpublished data only}

NCT02469857. Phase III efficacy and safety study of AB103 in the treatment of patients with necrotizing soft tissue infections (ACCUTE) [Phase III, randomized, double‐blind, placebo‐controlled, parallel group, study of AB103 as compared to placebo in patients with necrotizing soft tissue infections. ACCUTE (AB103 clinical composite endpoint study in necrotizing soft tissue infections)]. clinicaltrials.gov/ct2/show/NCT02469857 (First received: 6 June 2015). CENTRAL

Adams 1985

Adams EM, Gudmundsson S, Yocum DE, Haselby RC, Craig WA, Sundstrom WR. Streptococcal myositis. Archives of Internal Medicine 1985;145(6):1020‐3. [MEDLINE: 3890787]

Anaya 2005

Anaya DA, McMahon K, Nathens AB, Sullivan SR, Foy H, Bulger E. Predictors of mortality and limb loss in necrotizing soft tissue infections. Archives of Surgery 2005;140(2):151‐7; discussion 158. [MEDLINE: 15723996]

Anaya 2007

Anaya DA, Dellinger EP. Necrotizing soft‐tissue infection: diagnosis and management. Clinical Infectious Diseases 2007;44(5):705‐10. [MEDLINE: 17278065]

Anglemyer 2014

Anglemyer A, Horvath HT, Bero L. Healthcare outcomes assessed with observational study designs compared with those assessed in randomized trials. Cochrane Database of Systematic Reviews 2014;4:MR000034.

Audureau 2017

Audureau E, Hua C, de Prost N, Hemery F, Decousser JW, Bosc R, et al. Mortality of necrotising fasciitis: relative influence of individual and hospital‐level factors, a nationwide multilevel study, France, 2007‐2012. British Journal of Dermatology 2017;177(6):1575‐82. [DOI: 10.1111/bjd.15615; PUBMED: 28452064]

Bilton 1998

Bilton BD, Zibari GB, McMillan RW, Aultman DF, Dunn G, McDonald JC. Aggressive surgical management of necrotizing fasciitis serves to decrease mortality: a retrospective study. American S 1998;64(5):397‐400; discussion 400‐1. [PUBMED: 9585771]

Blencowe 2015

Blencowe NS, Brown JM, Cook JA, Metcalfe C, Morton DG, Nicholl J, et al. Interventions in randomised controlled trials in surgery: issues to consider during trial design. Trials 2015;16:392. [PUBMED: 26337522]

Boutron 2008

Boutron I, Moher D, Altman DG, Schulz KF, Ravaud P, CONSORT Group. Extending the CONSORT statement to randomized trials of nonpharmacologic treatment: explanation and elaboration. Annals of Internal Medicine 2008;148(4):295‐309. [PUBMED: 18283207]

Boyer 2009

Boyer A, Vargas F, Coste F, Saubusse E, Castaing Y, Gbikpi‐Benissan G, et al. Influence of surgical treatment timing on mortality from necrotizing soft tissue infections requiring intensive care management. Intensive Care Medicine 2009;35(5):847‐53. [PUBMED: 19099288]

Bryant 2006

Bryant AE, Bayer CR, Huntington JD, Stevens DL. Group A streptococcal myonecrosis: increased vimentin expression after skeletal‐muscle injury mediates the binding of Streptococcus pyogenes. Journal of Infectious Diseases 2006;193(12):1685‐92. [PUBMED: 16703512]

Carapetis 2014

Carapetis JR, Jacoby P, Carville K, Ang SJ, Curtis N, Andrews R. Effectiveness of clindamycin and intravenous immunoglobulin, and risk of disease in contacts, in invasive group a streptococcal infections. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America 2014;59(3):358‐65. [PUBMED: 24785239]

CDC 2012

Centers for Disease Control and Prevention. Active Bacterial Core Surveillance Report, Emerging Infections Program Network, Group A Streptococcus, 2010. www.cdc.gov/abcs/reports‐findings/survreports/gas10.html (accessed 29 April 2015).

Chelsom 1994

Chelsom J, Halstensen A, Haga T, Hoiby EA. Necrotising fasciitis due to group A streptococci in western Norway: incidence and clinical features. Lancet 1994;344(8930):1111‐5. [PUBMED: 7934492]

Childers 2002

Childers BJ, Potyondy LD, Nachreiner R, Rogers FR, Childers ER, Oberg KC, et al. Necrotizing fasciitis: a fourteen‐year retrospective study of 163 consecutive patients. American Surgeon 2002;68(2):109‐16. [MEDLINE: 11842952]

Chosidow 2001

Chosidow O. Subacute forms of necrotizing fasciitis and necrotizing cellulitis: diagnosis criteria and surgical decision‐making. Annales de Dermatologie et de Vénérologie 2001;128(3 Pt 2):390‐3. [MEDLINE: 11319370]

Colledge 2009

Colledge A, Hunter B, Bunkall LD, Holmes EB. Impairment rating ambiguity in the United States: the Utah impairment guides for calculating workers' compensation Impairments. Journal of Korean Medical Science 2009;24(Suppl 2):S232‐41. [MEDLINE: 19503678]

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. The Cochrane Collaboration, 2011.

Delaney 2008

Delaney A, Angus DC, Bellomo R, Cameron P, Cooper DJ, Finfer S, et al. Bench‐to‐bedside review: the evaluation of complex interventions in critical care. Critical Care 2008;12(2):210. [PUBMED: 18439321]

Dellinger 1981

Dellinger EP. Severe necrotizing soft‐tissue infections. Multiple disease entities requiring a common approach. JAMA 1981;246(15):1717‐21. [MEDLINE: 7277653]

DerSimonian 1986

DerSimonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7(3):177‐88. [MEDLINE: 3802833]

Elliott 1996

Elliott DC, Kufera JA, Myers RA. Necrotizing soft tissue infections. Risk factors for mortality and strategies for management. Annals of Surgery 1996;224(5):672‐83. [MEDLINE: 8916882]

Elliott 2000

Elliott D, Kufera JA, Myers RA. The microbiology of necrotizing soft tissue infections. American Journal of Surgery 2000;179(5):361‐6. [MEDLINE: 10930480]

Ellis Simonsen 2006

Ellis Simonsen SM, van Orman ER, Hatch BE, Jones SS, Gren LH, Hegmann KT, et al. Cellulitis incidence in a defined population. Epidemiology & Infection 2006;134(2):293‐9. [MEDLINE: 16490133]

Fournier 1883

Fournier AJ. Devastating gangrene of the penis [Gangrene Foudroyante de la verge]. Seminars in Medicine 1883;3:345.

Goh 2014

Goh T, Goh LG, Ang CH, Wong CH. Early diagnosis of necrotizing fasciitis. British Journal of Surgery 2014;101(1):e119‐25. [MEDLINE: 24338771]

GRADEpro [Computer program]

GRADE Working Group, McMaster University. GRADEpro GDT. Version accessed 11 October 2017. Hamilton (ON): GRADE Working Group, McMaster University, 2014.

Guidance FDA 2012

Food, Drug Administration. Guidances for industry and investigators on safety reporting requirements for investigational new drug applications and bioavailability/bioequivalence studies, and a small entity compliance guide. www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM332846.pdf (accessed 11 October 2017).

Guyatt 2011

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction‐GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383‐94. [PUBMED: 21195583 ]

Hasham 2005

Hasham S, Matteucci P, Stanley PR, Hart NB. Necrotising fasciitis. BMJ 2005;330(7495):830‐3. [MEDLINE: 15817551]

Higgins 2011

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.

Hsiao 2008

Hsiao CT, Weng HH, Yuan YD, Chen CT, Chen IC. Predictors of mortality in patients with necrotizing fasciitis. American Journal of Emergency Medicine 2008;26(2):170‐5. [MEDLINE: 18272096]

Ioannidis 2007

Ioannidis JP, Trikalinos TA. The appropriateness of asymmetry tests for publication bias in meta‐analyses: a large survey. CMAJ : Canadian Medical Association Journal 2007;176(8):1091‐6. [MEDLINE: 17420491]

Johansson 2010

Johansson L, Thulin P, Low DE, Norrby‐Teglund A. Getting under the skin: the immunopathogenesis of Streptococcus pyogenes deep tissue infections. Clinical Infectious Diseases 2010;51(1):58‐65. [MEDLINE: 20491545]

Kadri 2017

Kadri SS, Swihart BJ, Bonne SL, Hohmann SF, Hennessy LV, Louras P, et al. Impact of intravenous immunoglobulin on survival in necrotizing fasciitis with vasopressor‐dependent shock: a propensity score‐matched analysis from 130 US hospitals. Clinical Infectious Diseases 2017;64(7):877‐85. [PUBMED: 28034881]

Kaul 1997

Kaul R, McGeer A, Low DE, Green K, Schwartz B. Population‐based surveillance for group A streptococcal necrotizing fasciitis: Clinical features, prognostic indicators, and microbiologic analysis of seventy‐seven cases. Ontario Group A Streptococcal Study. American Journal of Medicine 1997;103(1):18‐24. [MEDLINE: 9236481]

Kobayashi 2011

Kobayashi L, Konstantinidis A, Shackelford S, Chan LS, Talving P, Inaba K, et al. Necrotizing soft tissue infections: delayed surgical treatment is associated with increased number of surgical debridements and morbidity. Journal of Trauma‐Injury Infection & Critical Care 2011;71(5):1400‐5. [MEDLINE: 21768906]

Korhonen 2000

Korhonen K. Hyperbaric oxygen therapy in acute necrotizing infections with a special reference to the effects on tissue gas tensions. Annales Chirurgiae et Gynaecologiae 2000;89(214):7‐36. [MEDLINE: 11288993]

Lamagni 2008

Lamagni TL, Darenberg J, Luca‐Harari B, Siljander T, Efstratiou A, Henriques‐Normark B, et al. Epidemiology of severe Streptococcus pyogenes disease in Europe. Journal of Clinical Microbiology 2008;46(7):2359‐67. [MEDLINE: 18463210]

Lamb 2015

Lamb LE, Sriskandan S, Tan LK. Bromine, bear‐claw scratch fasciotomies, and the Eagle effect: management of group A streptococcal necrotising fasciitis and its association with trauma. Lancet Infectious Diseases 2015;15(1):109‐21. [PUBMED: 25541175]

Le Cleach 2014

Le Cleach L, Trinquart L, Do G, Maruani A, Lebrun‐Vignes B, Ravaud P, et al. Oral antiviral therapy for prevention of genital herpes outbreaks in immunocompetent and nonpregnant patients. Cochrane Database of Systematic Reviews 2014, Issue 8. [DOI: 10.1002/14651858.CD009036.pub2]

Lepoutre 2011

Lepoutre A, Doloy A, Bidet P, Leblond A, Perrocheau A, Bingen E, et al. Epidemiology of invasive Streptococcus pyogenes infections in France in 2007. Journal of Clinical Microbiology 2011;49(12):4094‐100. [MEDLINE: 21976764]

Levett 2015

Levett D, Bennett MH, Millar I. Adjunctive hyperbaric oxygen for necrotizing fasciitis. Cochrane Database of Systematic Reviews 2015, Issue 1. [DOI: 10.1002/14651858.CD007937.pub2]

May 2009

May AK, Stafford RE, Bulger EM, Heffernan D, Guillamondegui O, Bochicchio G, et al. Treatment of complicated skin and soft tissue infections. Surgical Infections 2009;10(5):467‐99. [MEDLINE: 19860574]

McClaine 2010

McClaine RJ, Husted TL, Hebbeler‐Clark RS, Solomkin JS. Meta‐analysis of trials evaluating parenteral antimicrobial therapy for skin and soft tissue infections. Clinical Infectious Diseases 2010;50(8):1120‐6. [PUBMED: 20210644]

McHenry 1995

McHenry CR, Piotrowski JJ, Petrinic D, Malangoni MA. Determinants of mortality for necrotizing soft‐tissue infections. Annals of Surgery 1995;221(5):558‐63; discussion 563‐5. [MEDLINE: 7748037]

Norrby‐Teglund 1998

Norrby‐Teglund A, Stevens DL. Novel therapies in streptococcal toxic shock syndrome: attenuation of virulence factor expression and modulation of the host response. Current Opinion in Infectious Diseases 1998;11(3):285‐91. [MEDLINE: 17033393]

Norrby‐Teglund 2005

Norrby‐Teglund A, Muller MP, Mcgeer A, Gan BS, Guru V, Bohnen J, et al. Successful management of severe group A streptococcal soft tissue infections using an aggressive medical regimen including intravenous poly specific immunoglobulin together with a conservative surgical approach. Scandinavian Journal of Infectious Diseases 2005;37(3):166‐72. [MEDLINE: 15849047]

Nuwayhid 2007

Nuwayhid ZB, Aronoff DM, Mulla ZD. Blunt trauma as a risk factor for group A streptococcal necrotizing fasciitis. Annals of Epidemiology 2007;17(11):878‐81. [MEDLINE: 17697787]

O'Grady 2007

O'Grady KA, Kelpie L, Andrews RM, Curtis N, Nolan TM, Selvaraj G, et al. The epidemiology of invasive group A streptococcal disease in Victoria, Australia. Medical Journal of Australia 2007;186(11):565‐9. [MEDLINE: 17547544]

Park 2009

Park KH, Jung SI, Jung YS, Shin JH, Hwang JH. Marine bacteria as a leading cause of necrotizing fasciitis in coastal areas of South Korea. American Journal of Tropical Medicine & Hygiene 2009;80(4):646‐50. [MEDLINE: 19346393]

Pham 2009

Pham TN, Moore ML, Costa BA, Cuschieri J, Klein MB. Assessment of functional limitation after necrotizing soft tissue infection. Journal of Burn Care & Research 2009;30(2):301‐6. [MEDLINE: 19165118]

Phan 2010

Phan HH, Cocanour CS. Necrotizing soft tissue infections in the intensive care unit. Critical Care Medicine 2010;38(9 Suppl):S460‐8. [MEDLINE: 20724879]

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.

Rondinelli 2009

Rondinelli RD. Changes for the new AMA Guides to impairment ratings, 6th Edition: implications and applications for physician disability evaluations. PM & R : the Journal of Injury, Function, and Rehabilitation 2009;1(7):643‐56. [MEDLINE: 19627958]

Sarani 2009

Sarani B, Strong M, Pascual J, Schwab CW. Necrotizing fasciitis: current concepts and review of the literature. Journal of the American College of Surgeons 2009;208(2):279‐88. [MEDLINE: 19228540]

Sartelli 2014

Sartelli M, Malangoni MA, May AK, Viale P, Kao LS, Catena F, et al. World Society of Emergency Surgery (WSES) guidelines for management of skin and soft tissue infections. World Journal of Emergency Surgery 2014;9(1):57. [MEDLINE: 25422671]

SFD‐SPILF 2000

The French Society of Dermatology (SFD) and The French Society of infectious Disease (SPILF). Management of erysipelas and necrotizing fasciitis (long text) [Erysipèle et fasciite Nécrosante : Prise en charge]. Annales de Dermatologie et de Vénérologie 2000;127(12):1118‐37. [MEDLINE: 11173699]

Stevens 1989

Stevens DL, Tanner MH, Winship J, Swarts R, Ries KM, Schlievert PM, et al. Severe group A streptococcal infections associated with a toxic shock‐like syndrome and scarlet fever toxin A. New England Journal of Medicine 1989;321(1):1‐7. [MEDLINE: 2659990]

Stevens 2005

Stevens DL, Bisno AL, Chambers HF, Everett ED, Dellinger P, Goldstein EJ, et al. Practice guidelines for the diagnosis and management of skin and soft‐tissue infections. Clinical Infectious Diseases 2005;41(10):1373‐406. [PUBMED: 16231249]

Stevens 2014

Stevens DL, Bisno AL, Chambers HF, Dellinger EP, Goldstein EJ, Gorbach SL, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clinical Infectious Diseases 2014;59(2):e10‐52. [PUBMED: 24973422]

Wilkinson 2004

Wilkinson D, Doolette D. Hyperbaric oxygen treatment and survival from necrotizing soft tissue infection. Archives of Surgery 2004;139(12):1339‐45. [MEDLINE: 15611459]

Wilson 1952

Wilson B. Necrotizing fasciitis. American Surgeon 1952;18(4):416‐31. [MEDLINE: 14915014]

Wong 2003

Wong CH, Chang HC, Pasupathy S, Khin LW, Tan JL, Low CO. Necrotizing fasciitis: clinical presentation, microbiology, and determinants of mortality. Journal of Bone & Joint Surgery ‐ American volume 2003;85‐A(8):1454‐60. [MEDLINE: 12925624]

Hua 2015

Hua C, Bosc R, Sbidian E, De Prost N, Jabre P, Chosidow O, et al. Interventions for necrotizing soft tissue infections in adults. Cochrane Database of Systematic Reviews 2015, Issue 4. [DOI: 10.1002/14651858.CD011680]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Bulger 2014

Methods

Prospective, Phase IIA, randomised, parallel‐group, placebo‐controlled, double‐blinded study

Multicentric (6 centres) in the USA

Period of Inclusion: December 2011 to August 2012

Participants

Inclusion criteria

  • A clinical diagnosis of NSTI due to bacterial infection (e.g. necrotizing fasciitis, group A Streptococcus toxic shock, Fournier gangrene, clostridial gangrene or myonecrosis, and synergistic necrotizing cellulitis)

  • A decision to perform urgent surgical exploration and debridement

Exclusion criteria

  • Age less than 18 years

  • Weight greater than 150 kg

  • Diabetes mellitus with below‐ankle infection

  • Overt peripheral vascular disease in the involved area refractory

  • Refractory haemodynamic instability

  • Coagulopathy or hypoxia

  • Burn wounds

  • Cardiac arrest within the past 30 days

  • Expected survival less than 30 days because of underlying medical condition

  • Human immunodeficiency virus infection (CD4 count < 200 cells/mm3 or < 14% of lymphocytes)

  • Pregnancy or lactation

  • Prior curative tissue debridement

  • Immunosuppression

Baseline characteristics

N = 43, mean age of 50.7 years and 65% of men

Interventions

Each participant was treated with standardised empiric antibiotic therapy, and underwent surgical debridement of necrotic tissue

Intervention 1 (n = 17)

AB103, one dose intravenous (IV), 0.5 mg/kg, < 6 hours after diagnosis (infusion before, during or after the surgery)

Intervention 2 (n = 15)

AB103, one dose IV 0.25 mg/kg, < 6 hours after diagnosis (infusion before, during or after the surgery)

Intervention 3 (n = 11)

Placebo IV, < 6 hours after diagnosis (infusion before during or after the surgery)

Outcomes

Primary outcome

  • Change in the SOFA score from baseline within 28 days. The SOFA scores were calculated on days 0 (before drug administration), D1, D2, D4, D28 in modified ITT population

Secondary outcomes

  • Intensive care unit‐free, vasopressor‐free and ventilator‐free days within the first 28 days in modified ITT population

  • Number of debridements through day 7 in modified ITT population

  • Hospital length of stay in modified ITT population

  • Plasma and tissue cytokine levels at 0 to 72 hours in modified ITT population

  • Adverse events *including death* in ITT population

Notes

Financial support and the drug for this trial was provided by the manufacturer Atox Bio Ltd.

Authors declared conflict of interest for Bayer HealthCare, BayerSchering Pharma, Novartis, GlaxoSmith Kline and Wyeth.

* outcomes reported that were of interest in the review

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “Patients were randomised by a computer‐generated system to receive placebo or a low or high dose of AB103 in a 10:15:15 ratio.”

Comment: computer‐generated random sequence is adequate

Allocation concealment (selection bias)

Unclear risk

Comment: there was no description of the method used to guarantee allocation concealment

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote “double blinded”

Comment: placebo‐controlled with similar adverse‐effect profile reported. However, concerning the risk of performance bias, 2/43 (4.6%) patients randomised had received co‐treatment IVIG and HBO and the group of these two patients was not indicated. Authors did not specify the method of blinding of physicians and participants but they assess that all investigators and care providers remained masked throughout the study.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote “double blinded" "investigators remained blinded throughout the study"

Comment: there was no clear description of the process used to guarantee blinding of the assessor but it was a placebo‐controlled study with similar adverse‐effect profile reported and assessment of outcome was objective: death. Then we considered there was a low risk of detection bias for investigator‐reported outcomes for the same reasons.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote “Because drug administration may have started before definitive surgical diagnosis of NSTI, a modified ITT population was defined as patients in the ITT analysis who were properly randomised, treated (received AB103 or placebo), and assessable (definitive surgical diagnosis of NSTI).”

Comment: three patients were excluded from the ITT population; 1/11 (9%) in the placebo group who did not meet the clinical diagnosis of NSTI and 2/15 (13.3%) in the high‐dose group.

Selective reporting (reporting bias)

Low risk

Comment: in accordance with the protocol available in clinical trials.gov (NCT01417780)

Other bias

Low risk

No other bias identified

Madsen 2017

Methods

Randomised, blinded, parallel‐group, placebo‐controlled trial

Monocentric at Copenhagen University Hospital, where the management of patients with NSTI in Denmark is centralised

Period of inclusion: 7 April 2014 to 1 March 2016

Participants

Inclusion criteria

  • 18 years of age or older

  • Patients with confirmed NSTI at surgical exploration who were admitted to, or planned to be admitted to, the ICU

Exclusion criteria

  • Patients who had received more than one dose of IVIG before randomisation

  • Hypersensitivity to IVIG

  • Known hyperprolinaemia

  • Women who were pregnant or breast‐feeding

  • Diagnosis of NSTI for more than 48 hours

Baseline characteristics

N = 100, Mean age of 60 years and 60% of men

Interventions

Each patient was treated in accordance with the clinical protocols in place at Copenhagen University Hospital with standardised empiric antibiotic therapy (meropenem, clindamycin and ciprofloxacin), repeated surgical revisions, three sessions of hyperbaric oxygenation; sepsis, and supportive intensive care.

Intervention 1 (N = 50)

IVIG (Privigen, CSL Behring, Bern,Switzerland), 25 g/day for three consecutive days. The first dose of trial medicine was given immediately after arrival to the ICU or in the operating room before admission to ICU.

Intervention 2 (N = 50)

Equivalent amount of intravenous 0.9% saline for three consecutive days (placebo)

Outcomes

Primary outcome

  • Patient‐reported physical function as the physical component summary (PCS) score of the Medical Outcomes Study 36‐item short form health survey version 2 (SF‐36) at day 180 after randomisation*

Secondary outcomes

  • Mortality at 28*, 90 and180 days

  • Time to resolution of shock defined as maintenance of a systolic blood pressure of at least 90 mmHg without vasopressor agents for 24 hours

  • Severe bleeding defined as clinical bleeding and use of 3 units of red blood cells (RBC) within 24 hours of the episode in the ICU after randomisation

  • Bleeding in the ICU after randomisation

  • Total volumes of blood products used in the ICU after randomisation

  • Sepsis‐related organ failure assessment (SOFA) scores at days 1–7 after randomisation

  • Use of renal replacement therapy (RRT)

  • Ventilation and vasopressor in the ICU after randomisation

  • Serious adverse reactions observed in the ICU after randomisation*

  • Days alive off life support in the 90 days after randomisation

  • Days alive and out of hospital in the 180‐day follow‐up period

  • Amputation (yes/no) within the 180 days

Notes

* outcomes reported that were of interest in the review

Authors declared conflict of interest for CSL Behring.

Financial support from Fresenius Kabi, Germany, and Ferring Pharmaceuticals, Denmark, was declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomised 1:1 to IVIG or placebo. Two allocation lists with variable block sizes of 2, 4 and 6 were computer generated."

Comment: computer‐generated random sequence is adequate

Allocation concealment (selection bias)

Low risk

Quote: "Two allocation lists with variable block sizes of 2, 4 and 6 were computer generated to form two separate boxes that contained sequentially numbered, opaque and sealed envelopes" "Patients were randomised by dedicated personnel who drew the next envelope from the box according to the site of NSTI. The randomisation note was handed to an ICU nurse not otherwise involved in the care of the patient who placed both IVIG and 0.9% saline in a black, opaque plastic bag, inserted an orange‐coloured infusion set into the allocated intervention (IVIG or saline) and sealed the bag with a plastic strip (more details are given in the Electronic Supplementary Material (ESM)."

Comment: the method used to guarantee allocation concealment seems to be adequate

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The randomisation note was handed to an ICU nurse not otherwise involved in the care of the patient who placed both IVIG and 0.9% saline in a black, opaque plastic bag, inserted an orange‐coloured infusion set into the allocated intervention (IVIG or saline) and sealed the bag with a plastic strip."

"Patients, clinical staff caring for the patients, research staff, the statistician and the authors when writing the first draft for the abstract (supplementary results in the ESM) were all blinded to the intervention."

Comment: the method used to guarantee blinding of participants and personnel was adequate and well described

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Patients, clinical staff caring for the patients, research staff, the statistician and the authors when writing the first draft for the abstract (supplementary results in the ESM) were all blinded to the intervention" "The statistician (TL) did the analyses while still blinded to the intervention according to the statistical analysis plan."

Comment: the method used to guarantee blinding of outcome assessment was well described and primary outcome is a patient‐reported outcome.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "129 patients were screened, of whom 100 were enrolled; 50 patients were assigned to the IVIG group and 50 patients to the placebo group" "Of the 100 patients randomised, 87 were included in the intention‐to‐treat analysis."

Comment: 100 patients were randomised but the SF‐36 could not be obtained from 13 patients, thus analysis of primary outcome was based on 87patients. For the primary outcome, there were no data available for 38% of patients in each group with 25% of death and 13% of missing data.

Selective reporting (reporting bias)

Low risk

Comment: in accordance with the protocol available in clinical trials.gov NCT02111161

Other bias

High risk

Comment: There was baseline imbalance: number of patients who had received IVIG before randomisation was higher in placebo group 20 (40%) than in IVIG group 8 (16%) and number of patients with acute kidney injury was higher in IVIG group 5 (10%) than in placebo group 1 (2%). This is a placebo‐controlled trial, however nearly half of patients in placebo group had received one dose of IVIG before randomisation.

Vick‐Fragoso 2009

Methods

Prospective, non‐inferiority randomised controlled, open‐label, parallel‐group trial

Multicentric, 74 centres, worldwide

Period of inclusion: April 2001 to April 2002

Participants

Inclusion criteria

  • Patients ≥ 18 years

  • Patients with a complicated skin and soft tissue infection (cSSSI) at one site only (cSSSI diagnoses were prospectively defined: diabetic foot infection, necrotizing fasciitis, post‐surgical wound infection, complicated cellulitis, complicated erysipelas, major abscess of the skin, infection of traumatic lesion, and infected Ischaemic ulcer)

  • If it was anticipated that participants required systemic antimicrobial therapy

  • If they had a sample culture taken within 24 hours prior to being included

  • Patients had to had one of the following signs and symptoms: fever ≥ 38.0 C axillary or ≥ 38.5 C orally; leukocytosis (absolute white blood cell (WBC) count > 10,000 cells/mL) with neutrophilia (> 80% neutrophils), tachycardia (> 90 beats per minute), increased respiratory rate (> 20 breaths per minute), or elevated C‐reactive protein (CRP) – plus two or more of the following signs and symptoms within 24 hours preceding enrolment: local pain or tenderness, anaesthesia or hypoaesthesia of the affected area, swelling of the presumed affected area, purulent, serosanguinous, ‘dishwater’ or foul‐smelling discharge, gas formation detected under the skin, and changes in the appearance of the involved area, such as discolouration of skin, presence of black necrotic areas, red‐brown or haemorrhagic bullae, or skin colour changes from red‐blue to patches of blue‐grey.

Exclusion criteria

  • Patients with a diagnoses of uncomplicated mild‐to‐moderate SSSIs and of secondary infected burns, atopic dermatitis, or eczema

  • Pregnant or nursing patients

  • Patients with severe life‐threatening diseases with a life expectancy < 2 months

  • End‐stage liver cirrhosis

  • Severe renal impairment requiring dialysis,

  • Hypersensitivity to fluoroquinolones, and beta‐lactams

  • A congenital or sporadic syndromes of QTc prolongation or patients taking concomitant medication reported to increase the QTc

  • Patients with hypersensitivity to fluoroquinolones, and beta‐lactams

  • History of tendinopathy with quinolones

  • Severe renal impairment requiring dialysis

  • Septic shock

  • Patients receiving chronic immunosuppressant treatment

  • Patients with neutropenia (neutrophil absolute count < 1000 cells/mL) or at AIDS stage 1 or 2 (CD4+ absolute count < 200 cells/µL)

  • HIV‐seropositive individuals receiving highly‐activated antiretroviral treatment

  • SSSI secondary to prosthetic materials

  • 18% of the skin and soft tissue affected

  • Suspected underlying osteomyelitis not related to diabetic foot infection

  • Requirement for systemic concomitant antibacterial agents

  • fFilure to respond to previous antibacterial treatment only if previous treatment contained a fluoroquinolone, amoxicillin or a beta‐lactam/beta‐lactamase inhibitor combination

  • Patients who had received systemic antibacterial treatment (orally or parenteral) for > 24 hours within the 24 hours immediately prior to enrolment in the study

Baseline characteristics ITT population

Subgroup of NSTI (n = 54): 57.4% of male; mean age 52.2 years (provided upon a request to the authors)

Interventions

Intervention 1 (Per protocol (PP), n = 315); abscess n = 98; necrotizing fasciitis, n = 22; surgical wound infection n = 9; diabetic foot n = 49; complicated erysipelas n = 101; infected traumatic lesion n = 21; infected ischaemic ulcers n = 6; complicated cellulitis n = 9.

moxifloxacin 400 mg per day intravenous (IV), for at least 3 days, followed by moxifloxacin 400 mg orally, once daily, for 7–21 days

Intervention 2 (PP, n = 317): abscess n = 93, necrotizing fasciitis, n = 13, surgical wound infection n = 13, diabetic foot n = 63, complicated erysipelas n = 95, infected traumatic lesion n = 19, infected ischaemic ulcers n = 4, complicated cellulitis n = 17

amoxicillin‐clavulanate, IV 1000 mg/200 mg three times daily for at least 3 days, followed by amoxicillin‐clavulanate 500 mg/125 mg orally, three times daily, for 7–21 days

Outcomes

Primary outcome

  • Clinical response (CR) at test of cure (TOC) for the PP population

CR at the TOC, days 14‐28, visit was defined as: cure (total resolution or marked improvement of all cSSSI signs and symptoms; no additional or alternative antimicrobial treatment necessary). Evaluations (both the visual description of the lesion and the assessment of clinical outcome) were performed by investigators.

Secondary outcomes

  • Clinical response at TOC days 14‐28 for the intention to treat (ITT) population

  • Clinical response at TOC days14‐28 by indication in the PP and ITT population

  • Bacteriological success (eradication/presumed eradication) at TOC days 14‐28 for the PP/microbiologically evaluable population

  • Adverse events in ITT population*. Assessments for safety were based on physical examination and routine laboratory tests throughout the study

Notes

This study was sponsored by Bayer HealthCare AG.

Authors declared conflict of interest for Atox Bio Ltd and Biomedical Statistical Consulting, Wynnewood, Pennsylvania.

* outcomes reported that were of interest in the review

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: “randomised into two groups in a 1:1 ratio”

Comment: the method used to generate the allocation sequence was not reported

Allocation concealment (selection bias)

Unclear risk

Quote: "Patients were randomised into two groups in a 1:1 ratio to receive either moxifloxacin or the control regimen."

Comment: there was no mention of how allocation concealment was guaranteed

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: “open label”

Comment: both participants and study personnel were aware of the assigned treatment

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: “open‐label”, “Clinical outcomes were assessed bimodally as cure or failure, such that patients with only limited improvement in clinical status and partial resolution of symptoms would conservatively be considered as failures."

Comment: assessment was performed by investigators who were not blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: number of randomly assigned participants: 804

Number of analysed participants for the main outcome per protocol: 632

Patients withdrawn: 172

The main reasons for withdrawal after randomisation (moxifloxacin vs amoxicillin‐clavulanate) were adverse events (6.2% vs 3.8%), insufficient therapeutic effect (4.2% vs 5.3%), patient lost to follow‐up (3.9% vs 5.3%), and protocol violation (3.4% vs 2.8%) (P > 0.1 in all cases). There was a high rate of withdrawal mainly due to serious adverse events and insufficient therapeutic effect.

Selective reporting (reporting bias)

Unclear risk

Comment: no protocol was available, however, the outcomes predefined in the methods were well reported in the results section.

Other bias

High risk

Comment: there was baseline imbalance; number of patients in the NSTI subgroup was two‐fold higher in the moxifloxacin group (36/54) treatment than in the amoxicillin‐clavulanate group (18/54) treatment

AIDS: Acquired Immune Deficiency Syndrome; cSSSI: complicated skin and skin structure infections; cSSTI: complicated skin and soft tissue infection;CR: clinical response; ESM: electronic supplementary material; HBO: hyperbaric oxygen therapy;ICU: intensive care unit; IVIG: intravenous immunoglobulin; ITT: intention‐to‐treat;IV: intravenous; NSTI: necrotizing soft tissue infections; PCS: physical component summary; PP: per protocol; RBC: red blood cells; RRT: renal replacement therapy; SOFA: Sequential Organ Failure Assessment score; SF‐36: the Short Form (36) Health Survey; TOC: test of cure; WBC: white blood cells.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Abbar 2014

The study was not a randomised controlled trial.

Danino 2010

The study compared wound care dressings after surgery and not an intervention to treat NSTI.

Fass 1989

The study included patients with skin and soft tissue infection with no clear definition; we were unable to contact the main author (deceased).

Huang 2006

The study compared wound care dressings after surgery and not an intervention to treat NSTI.

Linner 2014

The study was not a randomised controlled trial.

Matejec 2013

The study's cross‐over design was not relevant for assessment of outcomes.

Morelli 2008

The study's cross‐over design was not relevant for assessment of outcomes.

Norrby‐Teglund 2009

The study was not a randomised controlled trial.

Solomkin 1986

The definition of necrotizing soft tissue infection was inaccurate and included diabetic foot infection and Ischaemic gangrene.

Souyri 2008

The study was not a randomised controlled trial.

Subrahmanyam 2004

The study compared wound care dressings after surgery and not an intervention to treat NSTI.

Wong 2004

The study was not a randomised controlled trial.

Xu 2015

The study compared wound care dressings after surgery.

NSTI: necrotizing soft tissue infection

Characteristics of studies awaiting assessment [ordered by study ID]

Darenberg 2003

Methods

This was a randomised controlled trial

Participants

Participants with a streptococcal toxic shock syndrome with or without NSTI

Interventions

Intravenous immunoglobulin G therapy versus placebo

Outcomes

Mortality at 28 days

Time to resolution of shock

Time to no further progression of the tissue infection

Survival on day 180

Notes

Outcomes in the subgroup of patient with NSTI are not available. Email to the study authors without answer

Tally 1986

Methods

This was a randomised controlled trial

Participants

Participants with serious surgical infections, including intraabdominal, pelvic, biliary tract, and necrotizing soft‐tissue infections suspected of containing B.fragilis

Interventions

Moxalactam versus Mefoxitin, with or without tTobramycin

Outcomes

Cure rates

Notes

Outcomes in the subgroup of patient with NSTI are not available. Email to the study authors without answer

NSTI: necrotizing soft tissue infection

Characteristics of ongoing studies [ordered by study ID]

NCT02469857

Trial name or title

Phase III efficacy and safety study of AB103 in the treatment of patients with necrotizing soft tissue infections (ACCUTE)

Methods

This was a randomised, double‐blind controlled trial

Participants

Participants with a necrotizing soft tissue infection

Interventions

AB103 0.5 mg/kg vs placebo

Outcomes

Clinical composite success end point (Time frame: 28 days)

Safety measures: adverse events

Safety measures: clinical safety laboratory

Safety measures: secondary infections

Recovery from acute kidney injury

Time to resolution of SOFA score to ≤ 1

Critical care and hospital stay parameters: ICU days

Critical care and hospital stay parameters: ventilator days

Critical care and hospital stay parameters: hospital length of stay

Starting date

September 2015

Contact information

Eileen M Bulger, MD Harborview Injury Prevention and Research Center

Notes

ICU: intensive care unit; SOFA: Sequential Organ Failure Assessment score

Data and analyses

Open in table viewer
Comparison 1. Moxifloxacin versus amoxicillin‐clavulanate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality within 30 days Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.39, 23.07]

Analysis 1.1

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 1 Mortality within 30 days.

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 1 Mortality within 30 days.

2 Proportion of patients who experienced serious adverse events Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.30, 1.31]

Analysis 1.2

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 2 Proportion of patients who experienced serious adverse events.

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 2 Proportion of patients who experienced serious adverse events.

Open in table viewer
Comparison 2. AB 103 versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality within 30 days Show forest plot

1

43

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.05, 2.16]

Analysis 2.1

Comparison 2 AB 103 versus placebo, Outcome 1 Mortality within 30 days.

Comparison 2 AB 103 versus placebo, Outcome 1 Mortality within 30 days.

2 Proportion of patients who experienced serious adverse events Show forest plot

1

43

Risk Ratio (M‐H, Random, 95% CI)

1.49 [0.52, 4.27]

Analysis 2.2

Comparison 2 AB 103 versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.

Comparison 2 AB 103 versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.

Open in table viewer
Comparison 3. IGIV versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality within 30 days Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.42, 3.23]

Analysis 3.1

Comparison 3 IGIV versus placebo, Outcome 1 Mortality within 30 days.

Comparison 3 IGIV versus placebo, Outcome 1 Mortality within 30 days.

2 Proportion of patients who experienced serious adverse events Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.73 [0.32, 1.65]

Analysis 3.2

Comparison 3 IGIV versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.

Comparison 3 IGIV versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.

Flow diagram.
Figuras y tablas -
Figure 1

Flow diagram.

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

'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 3

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

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 1 Mortality within 30 days.
Figuras y tablas -
Analysis 1.1

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 1 Mortality within 30 days.

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 2 Proportion of patients who experienced serious adverse events.
Figuras y tablas -
Analysis 1.2

Comparison 1 Moxifloxacin versus amoxicillin‐clavulanate, Outcome 2 Proportion of patients who experienced serious adverse events.

Comparison 2 AB 103 versus placebo, Outcome 1 Mortality within 30 days.
Figuras y tablas -
Analysis 2.1

Comparison 2 AB 103 versus placebo, Outcome 1 Mortality within 30 days.

Comparison 2 AB 103 versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.
Figuras y tablas -
Analysis 2.2

Comparison 2 AB 103 versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.

Comparison 3 IGIV versus placebo, Outcome 1 Mortality within 30 days.
Figuras y tablas -
Analysis 3.1

Comparison 3 IGIV versus placebo, Outcome 1 Mortality within 30 days.

Comparison 3 IGIV versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.
Figuras y tablas -
Analysis 3.2

Comparison 3 IGIV versus placebo, Outcome 2 Proportion of patients who experienced serious adverse events.

Summary of findings for the main comparison. Moxifloxacin compared to amoxicillin‐clavulanate for NSTI

Moxifloxacin compared to amoxicillin‐clavulanate for NSTI

Patient or population: NSTI
Setting: hospital
Intervention: moxifloxacin
Comparison: amoxicillin‐clavulanate

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality/certainty of the evidence
(GRADE)

Comments

Risk with Amoxicillin‐clavulanate

Risk with Moxifloxacin

Mortality
follow‐up: 30 days

Study population

RR 3.00
(0.39 to 23.07)

54
(1 RCT)

⊕⊝⊝⊝
Very lowa

Data from a larger trial including several types of soft tissue infections; total number of included patients N = 804

6 per 100

17 per 100
(2 to 100)

Serious adverse events (SAE)
follow‐up: 28 days

Study population

RR 0.63
(0.30 to 1.31)

54
(1 RCT)

⊕⊝⊝⊝
Very lowa

Description of nature of serious adverse events was not available

44 per 100

28 per 100
(13 to 58)

Survival time

54
(1 RCT)

⊕⊝⊝⊝
Very lowa

The median time of death after start of antibiotic treatment was shorter in the moxifloxacin group than in the amoxicillin‐clavulanate group (10.5 days versus 42 days) (not possible to calculate hazard ratio with the data provided)

Assessment of long‐term morbidity

Not reported

*The risk in the intervention group (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). Assumed risk for mortality was based on data of the literature (Audureau 2017; May 2009). For serious adverse effects it was based on the results of the trial.

CI: Confidence interval; RR: Risk ratio

GRADE Working Group grades of evidence
High quality/certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality/certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality/certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality/certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

aDowngraded by five levels to very low certainty of evidence. We downgraded two levels because of high risk of bias regarding blinding (open label trial) and high risk for attrition bias because of a high rate of withdrawal (20%). We downgraded one level for serious imprecision because of small sample size (and CI of RR included 1, where reported). We downgraded a further two levels because no clear criteria for clinical diagnosis of necrotizing fasciitis were provided and because antibiotic used as comparator is not relevant (indirectness)

Figuras y tablas -
Summary of findings for the main comparison. Moxifloxacin compared to amoxicillin‐clavulanate for NSTI
Summary of findings 2. AB103 compared to placebo for NSTI

AB103 compared to Placebo for NSTI

Patient or population: NSTI
Setting: hospital
Intervention: AB103
Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality/certainty of the evidence
(GRADE)

Comments

Risk with Placebo

Risk with AB103

Mortality
follow‐up: 30 days

Study population

RR 0.34
(0.05 to 2.16)

43
(1 RCT)

⊕⊝⊝⊝
Very lowa

23 per 100*

6 per 100
(1 to 39)

Serious adverse events (SAE)
follow‐up: 28 days

Study population

RR 1.49
(0.52 to 4.27)

43
(1 RCT)

⊕⊝⊝⊝
Very lowa

There were no data about the nature of serious adverse events reported

27 per 100

41 per 100
(14 to 100)

Survival time

Not reported

Assessment of long‐term morbidity

Not reported

*The risk in the intervention group (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). Assumed risk for mortality was based on data of the literature (Audureau 2017; May 2009). For serious adverse effects it was based on the results of the trial.

CI: Confidence interval; RR: Risk ratio

GRADE Working Group grades of evidence
High quality/certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality/certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality/certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality/certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

aDowngraded by three levels: one level for high risk of attrition bias, one level for no clear clinical definition of criteria for necrotizing fasciitis diagnosis at inclusion (indirectness), and one level for serious imprecision because of small sample size and CI included no difference

Figuras y tablas -
Summary of findings 2. AB103 compared to placebo for NSTI
Summary of findings 3. Intravenous immunoglobulin compared to placebo for NSTI

Intravenous immunoglobulin compared to placebo for NSTI

Patient or population: NSTI
Setting: intensive care unit
Intervention: intravenous immunoglobulin
Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality/Certainty of the evidence
(GRADE)

Comments

Risk with placebo

Risk with Intravenous immunoglobulin

Mortality
follow‐up: 30 days

Study population

RR 1.17
(0.42 to 3.23)

100
(1 RCT)

⊕⊕⊝⊝
Lowa

0 per 100

0 per 100
(0 to 0)

Moderate

23 per* 100

21 per 100
(8 to 58)

Serious adverse events (SAE)
follow‐up: unclear

Study population

RR 0.73
(0.32 to 1.65)

100
(1 RCT)

⊕⊕⊝⊝
Lowa

Serious adverse reactions included acute kidney injury, allergic reactions, aseptic meningitis syndrome, haemolytic anaemia, thrombi, and transmissible agents

22 per 100

16 per 100
(7 to 36)

Survival time

100
(1 RCT)

⊕⊕⊝⊝
Lowa

The median time of death was shorter in the IVIG group than in the placebo group (25 days versus 49 days) (not possible to calculate hazard ratio with the data provided)

Assessment of long‐term morbidity

Not reported

*The risk in the intervention group (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). Assumed risk for mortality was based on data of the literature (Audureau 2017; May 2009). For serious adverse effects it was based on the results of the trial.
CI: Confidence interval; RR: Risk ratio

GRADE Working Group grades of evidence
High quality/certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality/certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality/certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality/certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

aDowngraded by two levels: one level for high risk of attrition bias (38% lost of follow‐up); other bias: imbalance at baseline for one dose 25 IVIG received before randomisation (40% in placebo group vs 16% IVIG group). One level for indirectness as a minority of patients have an infection linked to bacteria producing toxins

Figuras y tablas -
Summary of findings 3. Intravenous immunoglobulin compared to placebo for NSTI
Table 1. Glossary of terms used

Term used

Explanation

Adjuvant treatment

Treatment that is given in addition to the primary or initial therapy to improve its effectiveness

Empiric antimicrobial therapy

Antimicrobial therapy given before the specific bacteria causing an infection is known

Aseptic meningitis

Serious inflammation of the linings of the brain not caused by pyogenic bacteria

Empiric antibiotic therapy

Antibiotics that acts against a wide range of bacteria

Bullae

Blisters on the skin usually more than 5 mm in diameters

Cirrhosis

Advanced liver disease

Crepitus

Clinical signs characterised by a peculiar sound under the skin

Debridement

Surgery excision of necrotic tissues (medical removal of dead, damaged, or infected tissue)

Endotoxin

A toxin contained in bacteria that is released only when the bacteria are broken down

Exotoxin

A toxin that is secreted by bacteria into the surrounding medium

Fascia

A fibrous connective tissue that surrounds muscle and other soft tissue. Fasciae are classified according to their distinct layers and their anatomical location: superficial fascia and deep (muscle) fascia

Fulminant inflammatory response

Systemic inflammatory response

Gram‐negative bacteria

Class of bacteria gram‐negative staining

Haemolytic anaemia

Decrease in the total amount of red blood cells due to the abnormal breakdown of red blood cells

Hyperbaric oxygen therapy

Medical use of oxygen at a level higher than atmospheric pressure. This helps fight bacteria and infection

Hypoxia

Insufficient levels of oxygen in blood or tissue

Intravenous immunoglobulin (IVIG)

Administration of antibodies through the veins

Motricity

Strength in upper and lower extremities after disease

Morbidity

Disability or degree that the health condition affects the patient

Mortality

Death rate

MRSA

Methicillin‐resistant Staphylococcus aureus

Myonecrosis

The destruction or death of muscle tissue

Necrosis

Death of body tissue

Obliterating endarteritis

Severe proliferating endarteritis (inflammation of the inner lining of an artery) that results in an occlusion of the lumen (the space inside a tubular structure) of the smaller vessels

Person‐years

Unit of measurement used to estimate rate of a disease during a defined period of observation

Polymicrobial

Polymicrobial infection is caused by several species of micro‐organisms

Subcutaneous tissue

Layer of tissue below the epidermis and the dermis of the skin. It is also called the hypodermis

Synergistic combination

Additive effects of bacterial agents

Synergistic gangrenes

Necrotizing soft tissue infection caused by a mix of bacteria (usually a mix of anaerobic and aerobic micro‐organisms)

Systemic

Affecting the entire body

Third‐generation quinolones

The quinolones are a family of synthetic broad‐spectrum antibiotic drugs

Thrombi

A blood clot inside a blood vessel

Transmissible agents

Infectious pathogens that can be transmitted

Vasopressors

Any medication that induces vasoconstriction of blood vessels to raise reduced blood pressure

Vimentin

A protein, the expression of which is increased after skeletal muscle injury

Figuras y tablas -
Table 1. Glossary of terms used
Table 2. Incidence of necrotizing fasciitis

Incidence of necrotizing fasciitis

Authors

Period of study

Country

Pathology

Incidence

Kaul R et al (Kaul 1997)

1991

Canada

GAS NF

0.085 per 100,000 p‐y

1995

0.4 per 100,000 p‐y

Ellis Simonsen et al (Ellis Simonsen 2006)

January 1997 to December 2002

United States

NF

0.04 per 1000 p‐y

O'Grady et al (O'Grady 2007)

March 2002 to August 2004

Australia

IGAS

2.7 per 100,000 p‐y (10.9% of NF)

Lamagni et al (Lamagni 2008)

January 2003 to December 2004

Europe

IGAS

2.37 per 100,000 p‐y (8% of NF)

Lepoutre et al (Lepoutre 2011)

November 2006 to November 2007

France

IGAS

3.1 per 100,000 p‐y (18% of NF)

GAS NF:group A streptococcal necrotizing fasciitis; IGAS: invasive group A streptococcal disease; NF: necrotizing fasciitis; p‐y: person‐years

Figuras y tablas -
Table 2. Incidence of necrotizing fasciitis
Table 3. Details of contacting authors

Study

Contact

Requested information

Contacted

Reply (last check 23 April 2017)

Darenberg 2003 (awaiting classification study)

Dr Norrby‐Teglund

Outcomes in the specific subgroup of patients with NSTI:

‐Mortality at day 30,

‐Proportion of patients with serious adverse events

‐Survival time

‐Patients with alteration of 25% of Functional Impairment Scale (%)

July 28, 2015

September 07, 2015

No response

Tally 1986 (awaiting classification study)

Dr Kellum

Outcomes in the specific subgroup of patients with NSTI:

‐Mortality at day 30,

‐Proportion of patients with serious adverse events

‐Survival time

‐Patients with alteration of 25% of Functional Impairment Scale (%)

July 24, 2015

September 07, 2015

No response

Vick‐Fragoso 2009 (included study)

Dr Bogner, Dr Petri

Outcomes in the specific subgroup of patients with NSTI:

‐Mortality at day 30,

‐Proportion of patients with serious adverse events

‐Survival time

‐Patients with alteration of 25% of Functional Impairment Scale (%)

September 07, 2015

Additional data to the publication provided for mortality, proportion of patients with serious adverse events and survival time.

Outcome data for assessment of long term morbidity not provide

Bulger 2014 (included study)

Dr Bulger

Outcomes:

‐Survival time

‐Patients with alteration of 25% of Functional Impairment Scale (%)

September 07, 2015

September 09, 2015

Outcome data not provided

Figuras y tablas -
Table 3. Details of contacting authors
Comparison 1. Moxifloxacin versus amoxicillin‐clavulanate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality within 30 days Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.39, 23.07]

2 Proportion of patients who experienced serious adverse events Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.30, 1.31]

Figuras y tablas -
Comparison 1. Moxifloxacin versus amoxicillin‐clavulanate
Comparison 2. AB 103 versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality within 30 days Show forest plot

1

43

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.05, 2.16]

2 Proportion of patients who experienced serious adverse events Show forest plot

1

43

Risk Ratio (M‐H, Random, 95% CI)

1.49 [0.52, 4.27]

Figuras y tablas -
Comparison 2. AB 103 versus placebo
Comparison 3. IGIV versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality within 30 days Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.42, 3.23]

2 Proportion of patients who experienced serious adverse events Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.73 [0.32, 1.65]

Figuras y tablas -
Comparison 3. IGIV versus placebo