gms | German Medical Science

GMS Infectious Diseases

Paul-Ehrlich-Gesellschaft für Infektionstherapie e.V. (PEG) (PEG)

ISSN 2195-8831

Antibiotic therapy in Shiga toxin producing Escherichia coli infection and colonization

Review Article

  • corresponding author Johannes K.-M. Knobloch - University Hospital of Schleswig-Holstein, Campus Lübeck, Department of Medical Microbiology and Hygiene, Lübeck, Germany
  • Susanne Hauswaldt - University Hospital of Schleswig-Holstein, Campus Lübeck, Department of Medical Microbiology and Hygiene, Lübeck, Germany
  • Werner Solbach - University Hospital of Schleswig-Holstein, Campus Lübeck, Department of Medical Microbiology and Hygiene, Lübeck, Germany
  • Martin Nitschke - University Hospital of Schleswig-Holstein, Campus Lübeck, Department of Internal Medicine I, Lübeck, Germany
  • Friedhelm Sayk - University Hospital of Schleswig-Holstein, Campus Lübeck, Department of Internal Medicine I, Lübeck, Germany

GMS Infect Dis 2013;1:Doc04

doi: 10.3205/id000004, urn:nbn:de:0183-id0000043

Veröffentlicht: 8. Juli 2013

© 2013 Knobloch et al.
Dieser Artikel ist ein Open Access-Artikel und steht unter den Creative Commons Lizenzbedingungen (http://creativecommons.org/licenses/by-nc-nd/3.0/deed.de). Er darf vervielfältigt, verbreitet und öffentlich zugänglich gemacht werden, vorausgesetzt dass Autor und Quelle genannt werden.


Abstract

The post diarrheal hemolytic uremic syndrome (HUS) is a major complication of enteric infections with Shiga toxin producing E. coli (STEC). According to the present recommendations, antibiotic therapy of acute bloody diarrhea caused by STEC is generally discouraged. These recommendations are based on historically conflicting results describing the potential induction of HUS by antibiotic treatment during the early phase of infection with enterohemorrhagic E. coli O157 whereas no guidelines are available for the use of antibiotics in cases of already fully established HUS or in asymptomatic long term STEC carriers. In 2011, a large outbreak of hemorrhagic colitis complicated by HUS occurred in northern Germany caused by a STEC strain of serotype O104:H4 harbouring both a phage encoding Stx 2 as well as a plasmid mediated enteroaggregative phenotype. The majority of infections were observed in adults, complicated by the highest number of HUS cases ever encountered. Due to different newly introduced therapeutic strategies (e.g. complement blockade) antibiotic therapy was used in many patients once HUS was established. The outbreak therefore provided important new insights for the understanding of antibiotic therapy of STEC associated HUS in adults and for decolonization of long term STEC carriers. This review highlights new aspects concerning use of antibiotics in STEC infection and colonization.

Keywords: Shiga toxin producing E. coli, enterohemorrhagic E. coli, haemolytic uremic syndrome, O104:H4 outbreak, STEC decolonization


Introduction

The hemolytic uremic syndrome (HUS) was first described by Gasser et al. in 1955 with the clinical hallmarks of acquired hemolytic anaemia, acute renal failure, hemorrhagic diathesis and cerebral symptoms [1]. Nowadays, HUS is classified as typical or post-diarrheal (D+) HUS versus atypical HUS not preceded by infectious diarrhea (D-) [2]. Development of D+ HUS is based on diarrheagenic bacteria expressing Shiga toxins (Stx) including Shigella dysenteria type 1 and Escherichia coli (STEC). STEC infections associated with D+ HUS were first described as Vero toxin producing E. coli (VTEC) about 30 years ago [3], [4]. STEC strains can express Stx 1 and/or 2, also known as Vero toxins or Verocytotoxins, which are encoded by phages [4], [5]. The toxin is believed to be responsible for the vascular damage (hemorrhagic colitis) and for systemic effects as seen in HUS. For both Stx 1 and Stx 2 several allelic variants are described, of which Stx 2 and 2c are more frequently associated with HUS development than others [6], [7]. The presence of the eae gene which is characteristic for enteropathogenic E. coli, grants adherence to the intestinal mucosa and defines the “classical” enterohemorrhagic E. coli (EHEC) subset within the STEC family. STEC strains lacking eae have traditionally been regarded as less virulent, but were also documented as causative agents of STEC disease including the recent German outbreak [8], [9]. E. coli can be serotyped by their O an H antigens. In the vast majority of STEC-related D+ HUS the serotype O157:H7 was reported [10]. However, in some parts of the world non-O157 serotypes like O26 and O111 caused up to half of all D+ HUS cases [11], [12]. Ruminant animals (especially cattle) are considered reservoirs of STEC. Transmission usually occurs via contaminated food or water.

STEC are commonly viewed as rare pathogens that cause severe disease predominantly in children. Before 2011, about 1,000 infections per year and less than 100 cases of HUS were registered in Germany in a nationwide surveillance [13]. According to the guidelines for STEC infections, antibiotic therapy of acute bloody diarrhea is generally discouraged due to its assumed potential to induce or promote D+ HUS in infections caused by enterohemorrhagic E. coli O157. In 2011, a large outbreak occurred in northern Germany caused by a STEC strain of serotype O104:H4 with the highest number of HUS cases ever encountered. A major issue discussed in the therapy of STEC infections deals with the question of whether or not to discourage the use of antibiotics. This review will discuss conflicting data on antibiotic therapy in STEC infection from both in vitro and in vivo studies. New concepts, especially concerning decolonization of patients in the post-diarrheal phase, will be highlighted.


Interaction of antibiotics and STEC in vitro

It is widely accepted that Stx production is boosted in vitro by subinhibitory concentrations of specific antibiotics with a possible impact on HUS pathogenesis. Since the early nineties, this topic has been investigated extensively. Results are partially conflicting and difficult to compare due to a variety of STEC strains investigated. Additionally, various antibiotics in inhibitory and subinhibitory concentrations as well as multiple methods for detection of phage induction and/or modification of Stx expression and the release of active toxin were used [14], [15], [16], [17], [18].

Due to these discrepancies in study design, solid data is mainly available for EHEC O157 and for two classes of antibiotics, the fluoroquinolones and trimethoprim-sulfamethoxazol (TMP/SMZ). Both substances have repeatedly been shown to induce Stx production in vitro, especially when applied in subinhibitory concentrations [15], [17], [19], [20]. This is highly plausible, as both antibiotics, targeting DNA synthesis, induce the bacterial SOS stress response to DNA damage which is linked to an increase in phage production and toxin release [17], [18].

For other antibiotics like makrolides, fosfomycin, clindamycin, cephalosporins and carbapenems results have been conflicting, showing either an increase or a decrease in Stx production or even no change at all [17], [20], [21]. Apart from variations in study design, this might be attributed to a strain dependent response to antibiotics [15], [16], [22]. Pedersen et al. showed that makrolides (azithromycin, telithromycin) at minimum inhibitory concentrations (MIC) increased Stx release from Stx-1 producing strains but decreased toxin release in STEC harboring Stx-2 variants with the exception of serotype O157 [16]. However, Stx induction from pure cultures may differ from Stx production in the complex intestinal environment [17].

For the recent outbreak strain STEC O104:H4, Bielaszewska et al. [23] confirmed that azithromycin did not induce Stx expression in vitro. Comparing subinhibitory concentrations of various antibiotics on the induction of Stx production of STEC O104:H4 they found that ciprofloxacin increased, while meropenem, rifaximin, tigecycline and azithromycin did not affect Stx production [23]. In the outbreak situation, early evaluation of these interactions was helpful to precisely determine the risk of antibiotic treatment.


Antibiotic therapy in acute STEC diarrhea and haemolytic uremic syndrome

Looking back on previous STEC outbreaks or sporadic infections, the impact of antibiotic treatment on the course of disease yielded inconsistent results. The vast majority of these reports exclusively dealt with EHEC O157 infections [24], but limited evidence is present in EHEC/STEC infections caused by other serotypes. The following section of this review comprises data discrediting the use of antibiotics, showing neither positive nor negative effects as well as potential benefits at least in the analyses of subgroups.

In 1990, a high rate of HUS was observed in patients receiving TMP/SMZ or sulfasalazine in a case control study of O157 infections [25]. All of these patients had received antibiotic treatment during the first 72 h of diarrheal illness. Consistently, in a prospective cohort study including 71 children aged less than 10 years, an increased risk for HUS was confirmed for TMP/SMZ administration during the first three days of diarrheal illness. Here β-lactams were identified as a second class of antibiotics increasing the likelihood of progression to HUS [26]. In a recent large multicentre trial analysing risk factors for the development of HUS in children infected by EHEC O157:H7 [27], antibiotic exposure during the first 7 days after onset of diarrhea was associated with increased risk of HUS development (OR 3.62; 95% CI, 1.23–10.6; p=0.02) in the overall analysis. Subgroup analysis of particular antibiotic substances revealed a significantly increased risk only for TMP-SMZ and metronidazol, whereas no significant differences were observed for β-lactams and azithromycin [27].

In a large case control study of O157 infections between 1996 and 2002 antibiotic treatment was not associated with HUS development in general [28]. However, subgroup analysis again revealed an increased risk for HUS if bactericidal antibiotic therapy was administered in the early phase of disease. The case control study presented by Slutsker et al. did not find an association between antibiotic treatment and progression to HUS [29]. Subgroup analysis in this study revealed that patients <13 years old who developed HUS were more likely to have received any antimicrobial agent within the first three days after onset [29]. In a retrospective analysis of a large O157 outbreak taking place in Scotland in 1996 [30], the administration of ciprofloxacin in the early stage of EHEC O157 hemorrhagic colitis was associated with a trend towards higher incidence of HUS, without reaching statistical significance. Administration of antibiotics during the four weeks preceding the onset of O157 disease, however, significantly increased the risk of developing HUS. This might be explained by residual subinhibitory intestinal concentrations after the end of antibiotic treatment. Alternatively, alterations of the post-antibiotic gut flora might predispose to D+ HUS. This coincides with reports from 1987 showing that antibiotic treatment prior to infection with E. coli O157 was associated with the risk of secondary transmission during an outbreak in a nursing home [31]. During this outbreak antibiotic treatment was also associated with an increased case fatality rate. However, this finding was interpreted cautiously by the authors, due to selection bias.

In summary, the afore mentioned studies may lead to the conclusion, that the first few days of acute hemorrhagic colitis might constitute a vulnerable period for an increased risk of HUS induction due to antibiotic treatment. Moreover, pre-diarrheal antibiotic exposure might increase the risk of HUS.

These findings are, however, in some contrast to a retrospective analysis of 278 children infected with EHEC O157 during an outbreak in Washington State in 1993. Here, no significant difference (OR 1.3; 95% CI, 0.6–2.6; p=0.56) in HUS development was observed between those children receiving antibiotics (16%, n=50) and those patients where antibiotics were withheld (12.8%, n=278), respectively [32]. In a subanalysis the administration of TMP-SMZ was accompanied with a slightly increased rate of HUS development (19.4%) still not attaining statistical significance (OR 1.5; 95% CI, 0.7–3.3; p=0.32). Moreover, TMP-SMZ treatment had no significant effect on the duration of EHEC shedding. Interestingly, in contrast to reports of TMP/SMZ as a risk factor for HUS, a prospective trial with TMP/SMZ in 47 children during O157:H7 enteritis [33] reported a lower incidence of HUS in the antibiotic group (9.1%) compared to untreated children (16.0%) without however statistical significance (p=0.67). In the analysis of 238 hospitalized patients with confirmed EHEC O157 infections in an endemic situation in New York State between 1998 and 1999 no significant association between antibiotic therapy and HUS development was observed [34].

Only few studies described a potential benefit for patients treated with antibiotics if given at the very early stage of disease. In 1999, Ikeda et al. reported a reduced risk of HUS development in patients receiving fosfomycin within the first two days of bloody diarrhea compared to patients not treated with fosfomycin at this very early stage [35]. However, the control group mainly consisted of patients treated with fosfomycin at a later time or with other antibiotics, but did not include a sufficient number of patients lacking any antibiotic therapy. In contrast to this study, Shiomi et al. reported reduced HUS development by early administration of oral fluoroquinolones compared to intravenously administered fosfomycin or oral fosfomycin in combination with intravenous cefotaxime [36]. Cimolai et al. reported a lower incidence of HUS in those patients treated with antibiotics, which however, could not be confirmed in the subsequent multivariate analysis [37]. In this study, the classes of antibiotics administered were not stated in detail.

In conclusion, some reports were able to demonstrate an increased risk for progression to HUS related to antibiotic exposure during diarrheal illness caused by EHEC O157. However, most studies reported neither beneficial nor adverse effects of antibiotic treatment in general and only few studies reported possible beneficial outcomes in specific subgroups of patients.

Based on these inconsistent reports in previous literature, the use of antibiotics was strongly discouraged during the 2011 German outbreak unless secondary complications urged for antibiotic treatment. Clinicians were confronted with a high number of adult patients with HUS-related severe acute kidney injury (high levels of blood urea nitrogen and serum creatinine), hemolysis and neurological complications [38], [39]. Standardized guidelines for causative treatment or randomized clinical trials approving any therapeutic concept to be beneficial beyond best supportive therapy were missing [40]. Therefore, therapeutic strategies were proposed ad hoc [41] based on theoretical considerations and preceding observations, but without any proof for the effectiveness of such “best guess” concepts. Moreover, these ad hoc strategies were continuously adjusted according to new observations made during the outbreak. Therefore, different medical centres used varying therapeutic regimens [42].

Despite the in vitro induction of Stx expression by quinolones and betalactams, pre-emptive therapy of STEC-HUS patients with a combination therapy of meropenem and ciprofloxacin in one medical centre in Northern Germany resulted in a statistically significant reduction of death, seizures and STEC shedding [42]. Results of previous studies analysing the influence of antimicrobial treatment on the clinical outcome of patients suffering from already fully established HUS were inconsistent. Martin et al. observed in a retrospective study patients with typical (n=101) and atypical (n=16) HUS that antimicrobial treatment before progression to HUS was associated with a mild clinical course. Only 3.0% of patients with severe disease received antibiotics compared with 22.6% of treated patients (p=0.01) displaying mild disease [43]. In a large prospective surveillance study in 395 patients suffering from D+ HUS, no differences were observed in the clinical outcome for patients (n=71) who had received antibiotics (β-lactams, metronidazol or ciprofloxacin) prior to admission to the hospital [44].


Antibiotic therapy in long term colonized carriers

By the end of May 2011 rapid clinical improvement under therapy with the anti-C5a antibody eculizumab was reported in three children suffering from STEC-HUS [45]. From this point on, patients of the German outbreak were therefore treated with eculizumab off-label. As eculizumab disrupts the complement cascade and thereby increases the risk for meningococcal meningitis [46], antibiotic meningitis prophylaxis was mandatory in non-vaccinated patients receiving this antibody-based therapy. For this purpose azithromycin was recommended in the ad hoc guidelines due to its documented in vitro inability to induce Stx production [41]. At the university hospital of Lübeck STEC-shedding was closely monitored. In patients not treated with antibiotics, individuals who developed HUS were compared with individuals who had mild signs of infection (non-HUS; Table 1 [Tab. 1]). The HUS and non-HUS groups were observed for similar periods. No significant differences in age or sex distributions were observed. The mean time of confirmed carriage was similar between the 2 groups (p=.98). This data indicates that the course of infection has no significant influence on STEC shedding.

All patients who were treated with eculizumab and had received azithromycin as meningitis prophylaxis were rapidly decolonized from STEC O104:H4, while untreated patients displayed significantly longer STEC shedding [47]. In detail, among azithromycin-treated HUS patients, long-term STEC carriage (>28 days) was observed in 1 of 22 patients (4.5%; 95% CI, 0%–13.3%), compared with 35 of 43 patients with or without HUS (81.4%; 95% CI, 69.8%–93.0%) who were not treated with this antibiotic (p<0.001). All 22 patients receiving azithromycin had at least 3 STEC-negative stool specimens after the completion of their antibiotic meningitis prophylaxis, and no recurrence of STEC was observed in these patients. The shortening of STEC shedding by azithromycin treatment was also confirmed by a larger multicenter study [48].

In contrast to the most prevalent STEC strains, the O104:H4 outbreak strain had an enteroaggregative phenotype, which might mediate the high rate of long term carriage. Azithromycin is an approved therapy in diarrheal disease caused by enteroaggregative E. coli [49]. Therefore, as proof of principle, a three day course of oral azithromycin (500 mg/d) was offered at our hospital to long term carriers (>28 d) of STEC O104:H4 who had initially not been treated with antibiotics, but, though now asymptomatic, were restricted in their social or working life (e.g. ban from work). After the 3-day course all 15 long term carriers treated with azithromycin for STEC decolonization had consistently negative stool specimens without any deterioration of renal function or development of other HUS related symptoms [47]. Therefore, successful decolonization treatment was extended to more than 40 persons without any adverse effects (unpublished data) up to the present time. Such a decolonization regimen, however, must always be weighed cautiously against the risk of other potential, pathogen-independent adverse drug side effects. Moreover, it has to be taken into account, that all promising results concerning the use of antibiotics for the treatment of STEC during the German STEC O104:H4 outbreak were retrieved either from patients already suffering from HUS, or from clinically recovered, now asymptomatic long-term carriers with a shedding time of at least 28 days. Therefore, at present, no definite conclusions can be drawn for the use of antibiotics in acute STEC-related hemorrhagic diarrhea. Future research has to further elucidate the risk or benefit of specified antibiotic treatment in the prevention or induction of HUS in this and other STEC strains. To date, antibiotics should be handled cautiously in patients with acute bloody diarrhea caused by STEC until their benefit might be approved in controlled trials.


Future therapeutic strategies and needs for research

During the northern German outbreak of STEC O104:H4 in 2011, new aspects regarding the antibiotic therapy in STEC infections and HUS were investigated retrospectively raising new options for treatment in STEC disease and carriage. However, there are still many questions which must be answered in the future. From a clinical point of view, the previous dogma that antibiotics are absolutely contraindicated in STEC disease needs to be revised. In our opinion, the point of time during the course of STEC disease should be a key landmark for the decision of therapeutic interventions with antibiotics. The contraindication of antibiotic use during early STEC disease (diarrheal phase) should still be strictly followed as the interaction of antibiotics with the expression of the Shiga toxin is strain specific and each substance class might be able to increase the risk of severe disease. The development of diagnostic assays enabling rapid quantitative Shiga toxin detection in differential growth conditions should be developed to enforce a risk assessment for individual strains during the early phase of STEC diagnostics. In patients with already established HUS a strict contraindication of antibiotics in all STEC caused HUS cannot be perpetuated for all STEC strains. At least for STEC O104:H4 it was demonstrated that use of azithromycin did not worsen the outcome [47] and a combination antibiotic therapy including substances known to induce Shiga toxin expression might be even beneficial in patients with established HUS [42]. Further studies on the effect of antibiotic therapy in established D+HUS are necessary to evaluate the observations made during the outbreak for other individual STEC serotypes. In the case of long term STEC shedding the safe use of azithromycin as decolonization therapy was demonstrated for 15 patients carrying the O104:H4 outbreak strain [47]. Several additional O104:H4 carriers could be eradicated by azithromycin treatment with high efficiency (unpublished data), though the authors have little experience in decolonization of STEC strains other than STEC O104:H4. However, in individual cases azithromycin treatment resulted in sustainable eradication of non-O104:H4 STEC long term carriers (unpublished data). Therefore, we agree with Mody and Griffin [50], that azithromycin eradication therapy could be offered to long term carriers after detailed discussion of the possible risks of treatment in a case to case decision if patients are strongly affected in their social or economic living.


Notes

Competing interests

The authors declare that they have no competing interests.


References

1.
Gasser C, Gautier E, Steck A, Siebenmann RE, Oechslin R. Hämolytisch-urämische Syndrome: bilaterale Nierenrindennekrosen bei akuten erworbenen hämolytischen Anämien [Hemolytic-uremic syndrome: bilateral necrosis of the renal cortex in acute acquired hemolytic anemia]. Schweiz Med Wochenschr. 1955 Sep;85(38-39):905-9.
2.
Elliott EJ, Robins-Browne RM. Hemolytic uremic syndrome. Curr Probl Pediatr Adolesc Health Care. 2005 Sep;35(8):310-30. DOI: 10.1016/j.cppeds.2005.06.002 Externer Link
3.
Karmali MA, Petric M, Lim C, Fleming PC, Steele BT. Escherichia coli cytotoxin, haemolytic-uraemic syndrome, and haemorrhagic colitis. Lancet. 1983 Dec;2(8362):1299-1300. DOI: 10.1016/S0140-6736(83)91167-4  Externer Link
4.
Karmali MA, Steele BT, Petric M, Lim C. Sporadic cases of haemolytic-uraemic syndrome associated with faecal cytotoxin and cytotoxin-producing Escherichia coli in stools. Lancet. 1983 Mar;1(8325):619-20. DOI: 10.1016/S0140-6736(83)91795-6  Externer Link
5.
Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nat Rev Microbiol. 2004 Feb;2(2):123-40. DOI: 10.1038/nrmicro818 Externer Link
6.
Persson S, Olsen KE, Ethelberg S, Scheutz F. Subtyping method for Escherichia coli shiga toxin (verocytotoxin) 2 variants and correlations to clinical manifestations. J Clin Microbiol. 2007 Jun;45(6):2020-4. DOI: 10.1128/JCM.02591-06 Externer Link
7.
Scheutz F, Teel LD, Beutin L, Piérard D, Buvens G, Karch H, Mellmann A, Caprioli A, Tozzoli R, Morabito S, Strockbine NA, Melton-Celsa AR, Sanchez M, Persson S, O'Brien AD. Multicenter evaluation of a sequence-based protocol for subtyping Shiga toxins and standardizing Stx nomenclature. J Clin Microbiol. 2012 Sep;50(9):2951-63. DOI: 10.1128/JCM.00860-12 Externer Link
8.
Karmali MA, Mascarenhas M, Shen S, Ziebell K, Johnson S, Reid-Smith R, Isaac-Renton J, Clark C, Rahn K, Kaper JB. Association of genomic O island 122 of Escherichia coli EDL 933 with verocytotoxin-producing Escherichia coli seropathotypes that are linked to epidemic and/or serious disease. J Clin Microbiol. 2003 Nov;41(11):4930-40. DOI: 10.1128/JCM.41.11.4930-4940.2003  Externer Link
9.
Beutin L, Zimmermann S, Gleier K. Human infections with Shiga toxin-producing Escherichia coli other than serogroup O157 in Germany. Emerging Infect Dis. 1998 Oct-Dec;4(4):635-9. DOI: 10.3201/eid0404.980415 Externer Link
10.
Tarr PI, Gordon CA, Chandler WL. Shiga-toxin-producing Escherichia coli and haemolytic uraemic syndrome. Lancet. 2005 Mar 19-25;365(9464):1073-86. DOI: 10.1016/S0140-6736(05)71144-2 Externer Link
11.
Johnson KE, Thorpe CM, Sears CL. The emerging clinical importance of non-O157 Shiga toxin-producing Escherichia coli. Clin Infect Dis. 2006 Dec;43(12):1587-95. DOI: 10.1086/509573 Externer Link
12.
Gerber A, Karch H, Allerberger F, Verweyen HM, Zimmerhackl LB. Clinical course and the role of shiga toxin-producing Escherichia coli infection in the hemolytic-uremic syndrome in pediatric patients, 1997-2000, in Germany and Austria: a prospective study. J Infect Dis. 2002 Aug;186(4):493-500. DOI: 10.1086/341940 Externer Link
13.
Robert Koch Institut. EHEC-Erkrankungen. In: Robert Koch Institut (RKI), ed. Infektionsepidemiologisches Jahrbuch meldepflichtiger Krankheiten für 2010. Berlin: RKI; 2011. p. 80-4.
14.
Walterspiel JN, Ashkenazi S, Morrow AL, Cleary TG. Effect of subinhibitory concentrations of antibiotics on extracellular Shiga-like toxin I. Infection. 1992 Jan-Feb;20(1):25-9. DOI: 10.1007/BF01704889  Externer Link
15.
Grif K, Dierich MP, Karch H, Allerberger F. Strain-specific differences in the amount of Shiga toxin released from enterohemorrhagic Escherichia coli O157 following exposure to subinhibitory concentrations of antimicrobial agents. Eur J Clin Microbiol Infect Dis. 1998 Nov;17(11):761-6. DOI: 10.1007/s100960050181  Externer Link
16.
Pedersen MG, Hansen C, Riise E, Persson S, Olsen KE. Subtype-specific suppression of Shiga toxin 2 released from Escherichia coli upon exposure to protein synthesis inhibitors. J Clin Microbiol. 2008 Sep;46(9):2987-91. DOI: 10.1128/JCM.00871-08 Externer Link
17.
McGannon CM, Fuller CA, Weiss AA. Different classes of antibiotics differentially influence shiga toxin production. Antimicrob Agents Chemother. 2010 Sep;54(9):3790-8. DOI: 10.1128/AAC.01783-09 Externer Link
18.
Kimmitt PT, Harwood CR, Barer MR. Toxin gene expression by shiga toxin-producing Escherichia coli: the role of antibiotics and the bacterial SOS response. Emerging Infect Dis. 2000 Sep-Oct;6(5):458-65. DOI: 10.3201/eid0605.000503 Externer Link
19.
Kimmitt PT, Harwood CR, Barer MR. Induction of type 2 Shiga toxin synthesis in Escherichia coli O157 by 4-quinolones. Lancet. 1999 May;353(9164):1588-9. DOI: 10.1016/S0140-6736(99)00621-2  Externer Link
20.
Yoh M, Frimpong EK, Voravuthikunchai SP, Honda T. Effect of subinhibitory concentrations of antimicrobial agents (quinolones and macrolide) on the production of verotoxin by enterohemorrhagic Escherichia coli O157:H7. Can J Microbiol. 1999 Sep;45(9):732-9.
21.
Murakami J, Kishi K, Hirai K, Hiramatsu K, Yamasaki T, Nasu M. Macrolides and clindamycin suppress the release of Shiga-like toxins from Escherichia coli O157:H7 in vitro. Int J Antimicrob Agents. 2000 Jul;15(2):103-9. DOI: 10.1016/S0924-8579(00)00126-6  Externer Link
22.
Corogeanu D, Willmes R, Wolke M, Plum G, Utermöhlen O, Krönke M. Therapeutic concentrations of antibiotics inhibit Shiga toxin release from enterohemorrhagic E. coli O104:H4 from the 2011 German outbreak. BMC Microbiol. 2012;12:160. DOI: 10.1186/1471-2180-12-160 Externer Link
23.
Bielaszewska M, Idelevich EA, Zhang W, Bauwens A, Schaumburg F, Mellmann A, Peters G, Karch H. Effects of antibiotics on Shiga toxin 2 production and bacteriophage induction by epidemic Escherichia coli O104:H4 strain. Antimicrob Agents Chemother. 2012 Jun;56(6):3277-82. DOI: 10.1128/AAC.06315-11 Externer Link
24.
Safdar N, Said A, Gangnon RE, Maki DG. Risk of hemolytic uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 enteritis: a meta-analysis. JAMA. 2002 Aug;288(8):996-1001. DOI: 10.1001/jama.288.8.996  Externer Link
25.
Pavia AT, Nichols CR, Green DP, Tauxe RV, Mottice S, Greene KD, Wells JG, Siegler RL, Brewer ED, Hannon D. Hemolytic-uremic syndrome during an outbreak of Escherichia coli O157:H7 infections in institutions for mentally retarded persons: clinical and epidemiologic observations. J Pediatr. 1990 Apr;116(4):544-51. DOI: 10.1016/S0022-3476(05)81600-2  Externer Link
26.
Wong CS, Jelacic S, Habeeb RL, Watkins SL, Tarr PI. The risk of the hemolytic-uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 infections. N Engl J Med. 2000 Jun;342(26):1930-6. DOI: 10.1056/NEJM200006293422601 Externer Link
27.
Wong CS, Mooney JC, Brandt JR, Staples AO, Jelacic S, Boster DR, Watkins SL, Tarr PI. Risk factors for the hemolytic uremic syndrome in children infected with Escherichia coli O157:H7: a multivariable analysis. Clin Infect Dis. 2012 Jul;55(1):33-41. DOI: 10.1093/cid/cis299 Externer Link
28.
Smith KE, Wilker PR, Reiter PL, Hedican EB, Bender JB, Hedberg CW. Antibiotic treatment of Escherichia coli O157 infection and the risk of hemolytic uremic syndrome, Minnesota. Pediatr Infect Dis J. 2012 Jan;31(1):37-41. DOI: 10.1097/INF.0b013e31823096a8 Externer Link
29.
Slutsker L, Ries AA, Maloney K, Wells JG, Greene KD, Griffin PM. A nationwide case-control study of Escherichia coli O157:H7 infection in the United States. J Infect Dis. 1998 Apr;177(4):962-6. DOI: 10.1086/515258  Externer Link
30.
Dundas S, Todd WT, Stewart AI, Murdoch PS, Chaudhuri AK, Hutchinson SJ. The central Scotland Escherichia coli O157:H7 outbreak: risk factors for the hemolytic uremic syndrome and death among hospitalized patients. Clin Infect Dis. 2001 Oct;33(7):923-31. DOI: 10.1086/322598 DOI: 10.1086/322598  Externer Link
31.
Carter AO, Borczyk AA, Carlson JA, Harvey B, Hockin JC, Karmali MA, Krishnan C, Korn DA, Lior H. A severe outbreak of Escherichia coli O157:H7--associated hemorrhagic colitis in a nursing home. N Engl J Med. 1987 Dec;317(24):1496-500. DOI: 10.1056/NEJM198712103172403 Externer Link
32.
Bell BP, Griffin PM, Lozano P, Christie DL, Kobayashi JM, Tarr PI. Predictors of hemolytic uremic syndrome in children during a large outbreak of Escherichia coli O157:H7 infections. Pediatrics. 1997 Jul;100(1):E12. DOI: 10.1542/peds.100.1.e12  Externer Link
33.
Proulx F, Turgeon JP, Delage G, Lafleur L, Chicoine L. Randomized, controlled trial of antibiotic therapy for Escherichia coli O157:H7 enteritis. J Pediatr. 1992 Aug;121(2):299-303. DOI: 10.1016/S0022-3476(05)81209-0  Externer Link
34.
Tserenpuntsag B, Chang HG, Smith PF, Morse DL. Hemolytic uremic syndrome risk and Escherichia coli O157:H7. Emerging Infect Dis. 2005 Dec;11(12):1955-7. DOI: 10.3201/eid1112.050607 Externer Link
35.
Ikeda K, Ida O, Kimoto K, Takatorige T, Nakanishi N, Tatara K. Effect of early fosfomycin treatment on prevention of hemolytic uremic syndrome accompanying Escherichia coli O157:H7 infection. Clin Nephrol. 1999 Dec;52(6):357-62.
36.
Shiomi M, Togawa M, Fujita K, Murata R. Effect of early oral fluoroquinolones in hemorrhagic colitis due to Escherichia coli O157:H7. Pediatr Int. 1999 Apr;41(2):228-32. DOI: 10.1046/j.1442-200X.1999.4121038.x  Externer Link
37.
Cimolai N, Basalyga S, Mah DG, Morrison BJ, Carter JE. A continuing assessment of risk factors for the development of Escherichia coli O157:H7-associated hemolytic uremic syndrome. Clin Nephrol. 1994 Aug;42(2):85-9.
38.
Trachtman H, Austin C, Lewinski M, Stahl RA. Renal and neurological involvement in typical Shiga toxin-associated HUS. Nat Rev Nephrol. 2012 Nov;8(11):658-69. DOI: 10.1038/nrneph.2012.196 Externer Link
39.
Magnus T, Röther J, Simova O, Meier-Cillien M, Repenthin J, Möller F, Gbadamosi J, Panzer U, Wengenroth M, Hagel C, Kluge S, Stahl RK, Wegscheider K, Urban P, Eckert B, Glatzel M, Fiehler J, Gerloff C. The neurological syndrome in adults during the 2011 northern German E. coli serotype O104:H4 outbreak. Brain. 2012 Jun;135(Pt 6):1850-9. DOI: 10.1093/brain/aws090 Externer Link
40.
Michael M, Elliott EJ, Ridley GF, Hodson EM, Craig JC. Interventions for haemolytic uraemic syndrome and thrombotic thrombocytopenic purpura. Cochrane Database Syst Rev. 2009;(1):CD003595. DOI: 10.1002/14651858.CD003595.pub2 Externer Link
41.
German Society of Nephrology. Advice of the German Society of Nephrology on the use of Eculizumab during the 2011 EHEC HUS outbreak, 04.06.2011 [Pamphlet]. Available from: http://www.dgfn.eu/aktuell/ehec-informationen/fuer-das-fachpublikum/advice-on-the-use-of-ecilizumab.html Externer Link
42.
Menne J, Nitschke M, Stingele R, Abu-Tair M, Beneke J, Bramstedt J, et al. Validation of treatment strategies for enterohaemorrhagic Escherichia coli O104:H4 induced haemolytic uraemic syndrome: case-control study. BMJ. 2012;345:e4565. DOI: 10.1136/bmj.e4565 Externer Link
43.
Martin DL, MacDonald KL, White KE, Soler JT, Osterholm MT. The epidemiology and clinical aspects of the hemolytic uremic syndrome in Minnesota. N Engl J Med. 1990 Oct;323(17):1161-7. DOI: 10.1056/NEJM199010253231703 Externer Link
44.
Lynn RM, O'Brien SJ, Taylor CM, Adak GK, Chart H, Cheasty T, Coia JE, Gillespie IA, Locking ME, Reilly WJ, Smith HR, Waters A, Willshaw GA. Childhood hemolytic uremic syndrome, United Kingdom and Ireland. Emerging Infect Dis. 2005 Apr;11(4):590-6. DOI: 10.3201/eid1104.040833 Externer Link
45.
Lapeyraque AL, Malina M, Fremeaux-Bacchi V, Boppel T, Kirschfink M, Oualha M, Proulx F, Clermont MJ, Le Deist F, Niaudet P, Schaefer F. Eculizumab in severe Shiga-toxin-associated HUS. N Engl J Med. 2011 Jun;364(26):2561-3. DOI: 10.1056/NEJMc1100859 Externer Link
46.
Parker C. Eculizumab for paroxysmal nocturnal haemoglobinuria. Lancet. 2009 Feb;373(9665):759-67. DOI: 10.1016/S0140-6736(09)60001-5 Externer Link
47.
Nitschke M, Sayk F, Härtel C, Roseland RT, Hauswaldt S, Steinhoff J, et al. Association between azithromycin therapy and duration of bacterial shedding among patients with Shiga toxin-producing enteroaggregative Escherichia coli O104:H4. JAMA. 2012 Mar;307(10):1046-52. DOI: 10.1001/jama.2012.264 Externer Link
48.
Vonberg RP, Höhle M, Aepfelbacher M, Bange FC, Belmar Campos C, Claussen K, et al. Duration of fecal shedding of Shiga toxin-producing Escherichia coli O104:H4 in patients infected during the 2011 outbreak in Germany: a multicenter study. Clin Infect Dis. 2013 Apr;56(8):1132-40. DOI: 10.1093/cid/cis1218 Externer Link
49.
de la Cabada Bauche J, Dupont HL. New Developments in Traveler's Diarrhea. Gastroenterol Hepatol (N Y). 2011 Feb;7(2):88-95.
50.
Mody RK, Griffin PM. Fecal shedding of Shiga toxin-producing Escherichia coli: what should be done to prevent secondary cases? Clin Infect Dis. 2013 Apr;56(8):1141-4. DOI: 10.1093/cid/cis1222 Externer Link