Z Gastroenterol 2015; 53(05): 418-459
DOI: 10.1055/s-0034-1399337
Leitlinie
© Georg Thieme Verlag KG Stuttgart · New York

S2k-Leitlinie Gastrointestinale Infektionen und Morbus Whipple[1]

S2k-guideline gastrointestinal infectious diseases and Whipple’s disease
S. Hagel
1   Klinik für Innere Medizin IV, Zentrum für Infektionsmedizin und Krankenhaushygiene und IFB Sepsis und Sepsisfolgen, Universitätsklinikum Jena der Friedrich-Schiller-Universität Jena, Jena
,
H.-J. Epple
2   Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin
,
G. E. Feurle
3   DRK Krankenhaus Neuwied, Neuwied
,
W. V. Kern
4   Abteilung Infektiologie, Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg
,
P. Lynen Jansen
5   Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten e.V. (DGVS), Berlin
,
P. Malfertheiner
6   Klinik für Gastroenterologie, Hepatologie und Infektiologie, Otto von Guericke Universität Magdeburg, Magdeburg
,
T. Marth
7   Abteilung für Innere Medizin, Krankenhaus Maria Hilf GmbH, Daun
,
E. Meyer
8   Krankenhaushygiene, Klinikum München, München
,
M. Mielke
9   Robert Koch-Institut, Berlin
,
V. Moos
10   Medizinische Klinik I mit Schwerpunkt Gastroenterologie, Infektiologie und Rheumatologie, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Berlin
,
L. von Müller
11   Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg
,
J. Nattermann
12   Medizinische Klinik und Poliklinik I, Universitätsklinikum Bonn, Bonn
,
M. Nothacker
13   Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e.V. (AWMF), Philipps-Universität, Marburg
,
C. Pox
14   Medizinische Klinik, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum
,
E. Reisinger
15   Klinik und Poliklinik für Innere Medizin, Abteilung für Tropenmedizin und Infektionskrankheiten, Universitätsmedizin Rostock, Rostock
,
B. Salzberger
16   Klinik und Poliklinik für Innere Medizin I, Universitätsklinikum Regensburg, Regensburg
,
H. J. F. Salzer
17   I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg
,
M. Weber
18   Klinik für Innere Medizin IV (Gastroenterologie, Hepatologie und Infektiologie), Universitätsklinikum Jena der Friedrich-Schiller-Universität Jena, Jena
,
T. Weinke
19   Klinik für Gastroenterologie und Infektiologie, Klinikum Ernst von Bergmann gGmbH, Potsdam
,
S. Suerbaum
20   Institut für Medizinische Mikrobiologie und Krankenhaushygiene, Medizinische Hochschule Hannover, Hannover
,
A. W. Lohse*
17   I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg
,
A. Stallmach*#
18   Klinik für Innere Medizin IV (Gastroenterologie, Hepatologie und Infektiologie), Universitätsklinikum Jena der Friedrich-Schiller-Universität Jena, Jena
,
Weitere Mitglieder der Leitlinienkommission in alphabetischer Reihenfolge einschließlich Affiliation:
› Author Affiliations
Further Information

Publication History

20 February 2015

08 March 2015

Publication Date:
12 May 2015 (online)

Einleitung und Methodik

E-1 Hintergrund

Akute gastrointestinale Infektionen werden durch eine tbVielzahl bakterieller, viraler und parasitärer Erreger hervorgerufen. Der Krankheitsverlauf wird durch die Virulenz des jeweiligen Erregers, die Wirtsantwort, durch den Ernährungs- und Immunstatus des Wirtes sowie mögliche Begleiterkrankungen beeinflusst. Meist ist eine infektiöse Gastroenteritis selbstlimitierend, eine symptomatische Behandlung ausreichend und eine ätiologische Abklärung aus klinischen Erwägungen nicht notwendig [1] .

Akute gastrointestinale Infektionen sind häufig. Im Rahmen einer epidemiologischen Studie, bei der über 12 Monate 21 262 Erwachsene nach Symptomen einer gastrointestinalen Infektion in den vergangenen 4 Wochen telefonisch befragt wurden, wurde für Deutschland eine Inzidenz von 0,95 Episoden/Personenjahr ermittelt. Hochgerechnet bedeutet dies für Deutschland rund 65 Mio. Episoden einer akuten gastrointestinalen Erkrankung pro Jahr, von denen die überwiegende Mehrzahl infektiöser Genese ist. 78 % der Erkrankten gaben an, eine Diarrhö gehabt zu haben, 12 % der Erkrankten berichteten über Erbrechen, 10 % der Erkrankten wiesen beide Symptome auf. Die durchschnittliche Erkrankungsdauer betrug 3,7 Tage. Mehr als ein Drittel (37,8 %) der Erkrankten begaben sich in eine ambulante ärztliche Betreuung, weniger als 1 – 3 % der Patienten wurden hospitalisiert [2]. Die bei Weitem größte Gruppe der Patienten mit akuter Gastroenteritis wird somit ambulant betreut. Im Gegensatz zu diesen blanden, selbstlimitierenden Verläufen steht die Beobachtung, dass sich von 2001 – 2011 die Zahl der stationären Aufnahmen aufgrund einer akuten infektiösen Gastroenteritis von 127 867 auf 282 199 Fälle pro Jahr mehr als verdoppelt hat. Besonders stark war dieser Anstieg in der Gruppe der über 65-jährigen Patienten sowie bei Patienten mit Clostridium difficile-Infektionen (CDI) zu verzeichnen. Die Zahl der Sterbefälle aufgrund gastrointestinaler Infektionen stieg im gleichen Zeitraum um das 10fache an [3].

Entsprechend der nationalen und internationalen Literatur wurden – wie in [Tab. 1] angegeben – zentrale Bezeichnungen für den Leitlinientext definiert.

Tab. 1

Definitionen für den Leitlinientext

Definition

akute Diarrhö

≥ 3 Stuhlentleerungen sowie ein Stuhlgewicht > 250 g pro Tag bei verminderter Stuhlkonsistenz

Dysenterie

Diarrhö einhergehend mit Blut- und Schleimbeimengungen

chronische Diarrhö

Diarrhö länger als 4 Wochen anhaltend

Immundefizienz

  • angeborene Immundefizienz, z. B. im Rahmen von T- und B-Zell-Defekten

  • Patienten mit erworbener Immundefizienz, z. B. Z. n. Organ- oder Knochenmarktransplantation; Chemotherapie solider oder hämatologischer Neoplasien mit oder ohne Neutropenie; HIV-Infektion im Stadium AIDS; Immunsuppressive oder immunmodulierende Therapie bei Autoimmunopathien; Glukokortikoidtherapie über einen Zeitraum von mindestens vier Wochen mit einer Erhaltungsdosis ≥ 10 mg/d.


#

E-2 Ziele der Leitlinie

Vor dem Hintergrund der Häufigkeit, aber auch der Bandbreite vom blanden, selbstlimitierenden Verlauf, bis hin zur vital-bedrohlichen Infektion ist ein evidenzbasiertes Vorgehen in der Diagnostik und Therapie notwendig, um den Patientenbedürfnissen gerecht zu werden, aber auch um die zur Verfügung stehenden Ressourcen adäquat einzusetzen. Aufgrund dessen hat sich die Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselerkrankungen entschlossen, eine S2k-Leitlinie zu erstellen, die diesem wachsenden Problem Rechnung trägt. Die methodische Erstellung der Leitlinie folgt dem Regelwerk der AWMF und dem Leitlinienprogramm der DGVS [4]. Um der Vielfältigkeit und den Besonderheiten gastrointestinaler Infektionen bei Patienten in unterschiedlichen Situationen gerecht zu werden, wurden insgesamt 6 Arbeitsgruppen gebildet:

  • Diagnostik der ambulant erworbenen Gastroenteritis (AG 1)

  • Klinisches Bild und Therapie der ambulant erworbenen Gastroenteritis (AG 2)

  • Nosokomiale Diarrhö und Clostridium difficile (AG 3)

  • Diarrhö bei Immundefizienz (AG 4)

  • Akute Gastroenteritis bei Reiserückkehrern (AG 5)

  • Morbus Whipple (AG 6)

Obgleich ausgesprochen selten, stellt auch die Infektion mit Tropheryma whipplei eine gastrointestinale Infektion dar, die in dieser Leitlinie in einem eigenen Kapitel dargestellt wird. In dieser Arbeitsgruppe gab es zur Empfehlung 6.4 keinen Konsens; hier fand eine sehr kontroverse Diskussion statt. Da Vertreter beider Positionen aufgrund ihrer klinischen Erfahrungen und wissenschaftlichen Expertise als nationale und internationale Experten gelten, sogenannte Kompromissformulierungen zu wenig inhaltliche Aussagen enthalten und deshalb für die Praxis als nicht hilfreich erachtet wurden, entschlossen sich die Koordinatoren in Abstimmung mit dem Vertreter der Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF) (M. Nothacker), ein sog. Minderheitsvotum aufzunehmen. Ebenso wurde zur Empfehlung 1.4 eine Minderheitsmeinung der Vertreter des Robert Koch-Instituts aufgenommen, die auf eine regelhafte Diagnostik bez. Stx-Bildner bei Patienten mit blutigen Diarrhöen zum Ausschluss einer EHEC-Infektion hinwiesen.

Das Ziel der S2k-Leitlinie „Gastrointestinale Infektionen und Morbus Whipple“ ist es, den aktuellen Kenntnisstand zu klinischem Bild, Diagnostik und Therapie gastrointestinaler Infektionen bei Erwachsenen auf Basis der wissenschaftlichen Evidenz zusammenzufassen, im Expertenkonsens zu bewerten und daraus praxisrelevante Empfehlungen abzuleiten. Sie soll einen Handlungskorridor für häufige Entscheidungen liefern und der evidenzbasierten Fort- und Weiterbildung dienen, um somit eine Verbesserung der medizinischen Versorgung betroffener Personen zu erreichen. Für die Therapie und die Besonderheiten bei Kindern und Jugendlichen sei auf die AWMF-Leitlinie „Akute infektiöse Gastroenteritis“ der Gesellschaft für Pädiatrische Gastroenterologie und Ernährung (GPGE) hingewiesen (AWMF Leitlinien Register; Nr. 068/003). Für die Diagnostik und Therapie der Helicobacter pylori-Infektion wird auf die entsprechende, aktuell in Revision befindliche, AWMF-Leitlinie „Helicobacter pylori und gastroduodenale Ulkuskrankheit” der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) und auf die Mikrobiologisch-infektiologischen Qualitätsstandards (MIQ): Gastrointestinale Infektionen verwiesen [5] [6]. Da Besonderheiten hinsichtlich Diagnostik, Therapie und Verlauf bei immundefizienten Patienten beachtet werden müssen, widmet sich diesem Thema detailliert das Kapitel 4. In der Gruppe immundefizienter Patienten werden einerseits Patienten mit angeborener Immundefizienz z. B. im Rahmen von T- und B-Zell-Defekten als auch Patienten mit erworbener Immundefizienz im Rahmen einer fortgeschrittenen HIV-Erkrankung, einer Immunsuppression bei Chemotherapie oder medikamentös bedingt nach Transplantationen oder unter der Therapie einer Autoimmunerkrankung verstanden. Auf Besonderheiten bei der Therapie älterer Patienten über 65 Jahre wird, sofern die Evidenz dies zulässt, in den entsprechenden Kapiteln zu Diagnostik und Therapie gesondert hingewiesen.

Gültigkeit der Empfehlungen

Die Diagnose- und Therapieempfehlungen gelten nicht für Säuglinge und Kleinkinder; hier sei auf die entsprechenden Leitlinien der Fachgesellschaften verwiesen.

Die Gültigkeit dieser Leitlinie beträgt 5 Jahre. Eine Revision ist für 2018/2019 geplant. Ansprechpartner für die Aktualisierung ist Frau PD. Dr. med. Petra Lynen Jansen, lynen@dgvs.de.


#

1 Ergebnisse einer S2k-Konsensuskonferenz der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselerkrankungen (DGVS) gemeinsam mit der Deutschen Gesellschaft für Innere Medizin (DGIM), der Deutschen Gesellschaft für Hygiene und Mikrobiologie (DGHM), der Deutschen Gesellschaft für Infektiologie (DGI), der Deutschen Gesellschaft für Krankenhaushygiene e. V. (DGKH), der Deutschen Gesellschaft für Pathologie (DGP), der Deutschen Gesellschaft für Tropenmedizin und Internationale Gesundheit (DTG), dem Robert Koch-Institut (RKI) und der Paul-Ehrlich Gesellschaft für Chemotherapie e. V. (PEG).


* Gleichberechtigte durch die DGVS mandatierte Koordinatoren der Leitlinie


# für die Mitglieder der Leitlinienkommission „Gastrointestinale Infektion und Morbus Whipple“


 
  • Literatur

  • 1 Lankisch PG, Mahlke R, Lübbers H et al. Leitsymptom Diarrhoe. Dtsch Arztebl 2006; 103 (05) A261-A268
  • 2 Wilking H, Spitznagel H, Werber D et al. Acute gastrointestinal illness in adults in Germany: a population-based telephone survey. Epidemiol Infect 2013; 141 (11) 2365-2375
  • 3 Lynen Jansen P, Stallmach A, Lohse AW et al. Entwicklung infektiöser Durchfallserkrankungen zwischen den Jahren 2000 und 2012. Z Gastroenterol 2014; 52 (06) 549-557
  • 4 Lynen Jansen P, Preiß JC, Muche-Borowski C et al. The guidelines program of the DGVS. Z Gastroenterol 2013; 51 (07) 643-650
  • 5 Fischbach W, Malfertheiner P, Hoffmann J et al. S3-Leitlinie „Helicobacter pylori und gastroduodenale Ulkuskrankheit” der Deutschen Gesellschaft für Verdauungs- und Stoffwechselkrankheiten (DGVS). Z Gastroenterol 2009; 47 (01) 68-102
  • 6 Kist M, Mauch H, Podbielski A et al. Mikrobiologisch-infektiologische Qualitätsstandards (MiQ), MIQ 09: Gastrointestinale Infektionen. 2. Ed. München: Elsevier, Urban & Fischer; 2013
  • 7 Farthing M, Lindberg G, Dite P et al. World Gastroenterology Organisation practice guideline: Acute diarrhea. Available at: http://www.worldgastroenterology.org/assets/downloads/en/pdf/guidelines/01_acute_diarrhea.pdf
  • 8 Siegel DL, Edelstein PH, Nachamkin I. Inappropriate testing for diarrheal diseases in the hospital. JAMA 1990; 263 (07) 979-982
  • 9 Guerrant RL, Van Gilder T, Steiner TS et al. Practice guidelines for the management of infectious diarrhea. Clin Infect Dis 2001; 32 (03) 331-351
  • 10 Jansen A, Stark K, Kunkel J et al. Aetiology of community-acquired, acute gastroenteritis in hospitalised adults: a prospective cohort study. BMC Infect Dis 2008; 8: 143
  • 11 Caspary WF, Lüpke NP, Oldiges FJ et al. Diarrhoe in der ärztlichen Praxis. Eine Bestandsaufnahme. Med Wochenschr 1995; 137: 411-415
  • 12 Riaz MM, Patel MJ, Khan MS et al. Clinical characteristics and predictors of positive stool culture in adult patients with acute gastroenteritis. J Pak Med Assoc 2012; 62 (01) 20-24
  • 13 Kotloff KL, Winickoff JP, Ivanoff B et al. Global burden of Shigella infections: implications for vaccine development and implementation of control strategies. Bull World Health Organ 1999; 77 (08) 651-666
  • 14 Niyogi SK. Shigellosis. J Microbiol 2005; 43 (02) 133-143
  • 15 McAuliffe GN, Anderson TP, Stevens M et al. Systematic application of multiplex PCR enhances the detection of bacteria, parasites, and viruses in stool samples. J Infect 2013; 67 (02) 122-129
  • 16 Lennon G, Reidy N, Collins PJ et al. A comparison of the efficiency of ELISA and selected primer sets to detect Norovirus isolates in southern Ireland over a four-year period (2002-2006): variation in detection rates and evidence for continuing predominance of NoV GII.4 genotype. Arch Virol 2014; 159 (07) 1697-1705
  • 17 Robert Koch-Institut. Rotaviren-Erkrankungen: RKI-Ratgeber für Ärzte. Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Rotaviren.html
  • 18 Bielaszewska M, Mellmann A, Zhang W et al. Characterisation of the Escherichia coli strain associated with an outbreak of haemolytic uraemic syndrome in Germany, 2011: a microbiological study. Lancet Infect Dis 2011; 11 (09) 671-676
  • 19 Frank C, Werber D, Cramer JP et al. Epidemic profile of Shiga-toxin-producing Escherichia coli O104:H4 outbreak in Germany. N Engl J Med 2011; 365 (19) 1771-1780
  • 20 Robert Koch-Institut. Abschliessende Darstellung und Bewertung der epidemiologischen Erkenntnisse im EHEC O104:H4 Ausbruch. 2011 Available at: http://www.rki.de/DE/Content/InfAZ/E/EHEC/EHEC_O104/EHEC-Abschlussbericht.pdf?__blob=publicationFile
  • 21 Vallieres E, Saint-Jean M, Rallu F. Comparison of three different methods for detection of Shiga toxin-producing Escherichia coli in a tertiary pediatric care center. J Clin Microbiol 2013; 51 (02) 481-486
  • 22 Robert Koch-Institut. Erkrankungen durch Enterohämorrhagische Escherichia coli (EHEC): Ratgeber für Ärzte. Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_EHEC.html
  • 23 Bauer MP, Notermans DW, van Benthem BH et al. Clostridium difficile infection in Europe: a hospital-based survey. Lancet 2011; 377 (9759) 63-73
  • 24 D’Agostino Sr RB, Collins SH, Pencina KM et al. Risk Estimation for Recurrent Clostridium difficile Infection Based on Clinical Factors. Clin Infect Dis 2014; 58 (10) 1386-1393
  • 25 Friedman HS, Navaratnam P, Reardon G et al. A retrospective analysis of clinical characteristics, hospitalization, and functional outcomes in residents with and without Clostridium difficile infection in US long-term care facilities. Curr Med Res Opin 2014; 30 (06) 1121-1130
  • 26 Raveh D, Rabinowitz B, Breuer GS et al. Risk factors for Clostridium difficile toxin-positive nosocomial diarrhoea. Int J Antimicrob Agents 2006; 28 (03) 231-237
  • 27 Vojtilova L, Freibergerova M, Jurankova J et al. Epidemiological factors influencing the development of relapsing and severe Clostridium difficile infection. Epidemiol Mikrobiol Imunol 2014; 63 (01) 27-35
  • 28 Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother 2014; 69 (04) 881-891
  • 29 Pakyz AL, Jawahar R, Wang Q et al. Medication risk factors associated with healthcare-associated Clostridium difficile infection: a multilevel model case-control study among 64 US academic medical centres. J Antimicrob Chemother 2014; 69 (04) 1127-1131
  • 30 Kwok CS, Arthur AK, Anibueze CI et al. Risk of Clostridium difficile infection with acid suppressing drugs and antibiotics: meta-analysis. Am J Gastroenterol 2012; 107 (07) 1011-1019
  • 31 Tleyjeh IM, Bin Abdulhak AA, Riaz M et al. Association between proton pump inhibitor therapy and clostridium difficile infection: a contemporary systematic review and meta-analysis. PLoS One 2012; 7 (12) e50836
  • 32 Dulny G, Zalewska M, Mlynarczyk G. An analysis of risk factors of Clostridiumdifficile infection in patients hospitalized in the teaching hospital in 2008. Przegl Epidemiol 2013; 67 (03) 445-450 , 547–551
  • 33 Garg S, Mirza YR, Girotra M et al. Epidemiology of Clostridium difficile-associated disease (CDAD): a shift from hospital-acquired infection to long-term care facility-based infection. Dig Dis Sci 2013; 58 (12) 3407-3412
  • 34 Das R, Feuerstadt P, Brandt LJ. Glucocorticoids are associated with increased risk of short-term mortality in hospitalized patients with clostridium difficile-associated disease. Am J Gastroenterol 2010; 105 (09) 2040-2049
  • 35 Collini PJ, Bauer M, Kuijper E et al. Clostridium difficile infection in HIV-seropositive individuals and transplant recipients. J Infect 2012; 64 (02) 131-147
  • 36 West M, Pirenne J, Chavers B et al. Clostridium difficile colitis after kidney and kidney-pancreas transplantation. Clin Transplant 1999; 13 (04) 318-323
  • 37 Lübbert C, Johann C, Kekule AS et al. Immunosuppressive treatment as a risk factor for the occurrence of clostridium difficile infection (CDI). Z Gastroenterol 2013; 51 (11) 1251-1258
  • 38 Planche TD, Davies KA, Coen PG et al. Differences in outcome according to Clostridium difficile testing method: a prospective multicentre diagnostic validation study of C difficile infection. Lancet Infect Dis 2013; 13 (11) 936-945
  • 39 Debast SB, Bauer MP, Kuijper EJ et al. European Society of Clinical Microbiology and Infectious Diseases: update of the treatment guidance document for Clostridium difficile infection. Clin Microbiol Infect 2014; 20 (Suppl. 02) 1-26
  • 40 Surawicz CM, Brandt LJ, Binion DG et al. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am J Gastroenterol 2013; 108 (04) 478-498 ; quiz 499
  • 41 Finch LS, Duncan CM. Molecular test to determine toxigenic capabilities in GDH-positive, toxin-negative samples: evaluation of the Portrait toxigenic C. difficile assay. Br J Biomed Sci 2013; 70 (02) 62-66
  • 42 Jones AM, Kuijper EJ, Wilcox MH. Clostridium difficile: a European perspective. J Infect 2013; 66 (02) 115-128
  • 43 von Muller L, Halfmann A, Herrmann M. Current data and trends on the development of antibiotic resistance of Clostridium difficile. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2012; 55 (11) 1410-1417
  • 44 Wei SC, Hung CC, Chen MY et al. Endoscopy in acquired immunodeficiency syndrome patients with diarrhea and negative stool studies. Gastrointest Endosc 2000; 51 (04) 427-432
  • 45 Wahnschaffe U, Ignatius R, Loddenkemper C et al. Diagnostic value of endoscopy for the diagnosis of giardiasis and other intestinal diseases in patients with persistent diarrhea from tropical or subtropical areas. Scand J Gastroenterol 2007; 42 (03) 391-396
  • 46 Shah RJ, Fenoglio-Preiser C, Bleau BL et al. Usefulness of colonoscopy with biopsy in the evaluation of patients with chronic diarrhea. Am J Gastroenterol 2001; 96 (04) 1091-1095
  • 47 Berdichevski T, Keller N, Rahav G et al. The impact of pseudomembrane formation on the outcome of Clostridium difficile-associated disease. Infection 2013; 41 (05) 969-977
  • 48 Ben-Horin S, Margalit M, Bossuyt P et al. Prevalence and clinical impact of endoscopic pseudomembranes in patients with inflammatory bowel disease and Clostridium difficile infection. J Crohns Colitis 2010; 4 (02) 194-198
  • 49 Nomura K, Fujimoto Y, Yamashita M et al. Absence of pseudomembranes in Clostridium difficile-associated diarrhea in patients using immunosuppression agents. Scand J Gastroenterol 2009; 44 (01) 74-78
  • 50 Pavlidis P, Chedgy FJ, Tibble JA. Diagnostic accuracy and clinical application of faecal calprotectin in adult patients presenting with gastrointestinal symptoms in primary care. Scand J Gastroenterol 2013; 48 (09) 1048-1054
  • 51 Taylor WI, Schelhart D. Effect of temperature of incubation on performance of media in the detection of enteric pathogens. Appl Microbiol 1973; 25 (06) 940-944
  • 52 Valenstein P, Pfaller M, Yungbluth M. The use and abuse of routine stool microbiology: a College of American Pathologists Q-probes study of 601 institutions. Arch Pathol Lab Med 1996; 120 (02) 206-211
  • 53 Khanna S, Pardi DS, Rosenblatt JE et al. An evaluation of repeat stool testing for Clostridium difficile infection by polymerase chain reaction. J Clin Gastroenterol 2012; 46 (10) 846-849
  • 54 Deshpande A, Pasupuleti V, Patel P et al. Repeat stool testing for Clostridium difficile using enzyme immunoassay in patients with inflammatory bowel disease increases diagnostic yield. Curr Med Res Opin 2012; 28 (09) 1553-1560
  • 55 Hiatt RA, Markell EK, Ng E. How many stool examinations are necessary to detect pathogenic intestinal protozoa?. Am J Trop Med Hyg 1995; 53 (01) 36-39
  • 56 McFarland LV, Mulligan ME, Kwok RY et al. Nosocomial acquisition of Clostridium difficile infection. N Engl J Med 1989; 320 (04) 204-210
  • 57 Atia AN, Buchman AL. Oral Rehydration Solutions in Non-Cholera Diarrhea: A Review. Am J Gastroenterol 2009; 104 (10) 2596-2604
  • 58 Rao SSC, Summers RW, Rao GRS et al. Oral rehydration for viral gastroenteritis in adults: a randomized, controlled trial of 3 solutions. J Parenter Enteral Nutr 2006; 30 (05) 433-439
  • 59 Binder HJ, Brown I, Ramakrishna BS et al. Oral Rehydration Therapy in the Second Decade of the Twenty-first Century. Curr Gastroenterol Rep 2014; 16 (03) 376
  • 60 Hartling L, Bellemare S, Wiebe N et al. Oral versus intravenous rehydration for treating dehydration due to gastroenteritis in children. Cochrane Database Syst Rev 2006; (03) CD004390
  • 61 Hou FQ, Wang Y, Li J et al. Management of acute diarrhea in adults in China: a cross-sectional survey. BMC Public Health 2013; 13: 41
  • 62 Thielman NM, Guerrant RL. Clinical practice. Acute infectious diarrhea. N Engl J Med 2004; 350 (01) 38-47
  • 63 Huang DB, Awasthi M, Le BM et al. The role of diet in the treatment of travelers’ diarrhea: a pilot study. Clin Infect Dis 2004; 39 (04) 468-471
  • 64 Sanders JW, Frenck RW, Putnam SD et al. Azithromycin and loperamide are comparable to levofloxacin and loperamide for the treatment of traveler’s diarrhea in United States military personnel in Turkey. Clin Infect Dis 2007; 45 (03) 294-301
  • 65 Murphy GS, Bodhidatta L, Echeverria P et al. Ciprofloxacin and loperamide in the treatment of bacillary dysentery. Ann Intern Med 1993; 118 (08) 582-586
  • 66 Ericsson CD, DuPont HL, Okhuysen PC et al. Loperamide plus azithromycin more effectively treats travelers’ diarrhea in Mexico than azithromycin alone. J Travel Med 2007; 14 (05) 312-319
  • 67 Ericsson CD, DuPont HL, Mathewson JJ et al. Treatment of traveler’s diarrhea with sulfamethoxazole and trimethoprim and loperamide. JAMA 1990; 263 (02) 257-261
  • 68 Taylor DN, Sanchez JL, Candler W et al. Treatment of travelers’ diarrhea: ciprofloxacin plus loperamide compared with ciprofloxacin alone. A placebo-controlled, randomized trial. Ann Intern Med 1991; 114 (09) 731-734
  • 69 Dupont HL, Jiang ZD, Belkind-Gerson J et al. Treatment of travelers’ diarrhea: randomized trial comparing rifaximin, rifaximin plus loperamide, and loperamide alone. Clin Gastroenterol Hepatol 2007; 5 (04) 451-456
  • 70 Bergström T, Alestig K, Thorén K et al. Symptomatic treatment of acute infectious diarrhoea: loperamide versus placebo in a double-blind trial. J Infect 1986; 12 (01) 35-38
  • 71 Kato H, Kato H, Nakamura M et al. A case of toxic megacolon secondary to Clostridium difficile-associated diarrhea worsened after administration of an antimotility agent and molecular analysis of recovered isolates. J Gastroenterol 2007; 42 (06) 507-508
  • 72 Schneider A, Rünzi M, Peitgen K et al. Campylobacter jejuni-induced severe colitis – a rare cause of toxic megacolon. Z Gastroenterol 2000; 38 (04) 307-309
  • 73 McGregor A, Brown M, Thway K et al. Fulminant amoebic colitis following loperamide use. J Travel Med 2007; 14 (01) 61-62
  • 74 Eronen M, Putkonen H, Hallikainen T et al. Lethal gastroenteritis associated with clozapine and loperamide. Am J Psychiatry 2003; 160 (12) 2242-2243
  • 75 Walley T, Milson D. Loperamide related toxic megacolon in Clostridium difficile colitis. Postgrad Med J 1990; 66 (777) 582
  • 76 Cuadrado-Gómez LM, Arranz-Caso A, Albarrán-Hernández F et al. Toxic megacolon caused by loperamide as initial form of Crohn disease. Rev Clin Esp 1994; 194 (03) 201-202
  • 77 Petit R, Bouziane A, Calès P et al. Toxic megacolon caused by loperamide-myorelaxants combination. Gastroenterol Clin Biol 1993; 17 (01) 62-63
  • 78 Bell BP, Griffin PM, Lozano P et al. Predictors of hemolytic uremic syndrome in children during a large outbreak of Escherichia coli O157:H7 infections. Pediatrics 1997; 100 (01) E12
  • 79 Eberlin M, Mück T, Michel MC. A comprehensive review of the pharmacodynamics, pharmacokinetics, and clinical effects of the neutral endopeptidase inhibitor racecadotril. Front Pharmacol 2012; 3: 93
  • 80 Gallelli L, Colosimo M, Tolotta GA et al. Prospective randomized double-blind trial of racecadotril compared with loperamide in elderly people with gastroenteritis living in nursing homes. Eur J Clin Pharmacol 2010; 66 (02) 137-144
  • 81 Prado D. Global Adult Racecadotril Study Group. A multinational comparison of racecadotril and loperamide in the treatment of acute watery diarrhoea in adults. Scand J Gastroenterol 2002; 37 (06) 656-661
  • 82 Etienney I, Beaugerie L, Viboud C et al. Non-steroidal anti-inflammatory drugs as a risk factor for acute diarrhoea: a case crossover study. Gut 2003; 52 (02) 260-263
  • 83 Nonsteroidal anti-inflammatory drugs: add an anti-ulcer drug for patients at high risk only. Always limit the dose and duration of treatment with NSAIDs. Prescrire Int 2011; 20 (119) 216-219
  • 84 Fedorowicz Z, Jagannath VA, Carter B. Antiemetics for reducing vomiting related to acute gastroenteritis in children and adolescents. Cochrane Database Syst Rev 2011; (09) CD005506
  • 85 Passariello A, Agricole P, Malfertheiner P. A critical appraisal of probiotics (as drugs or food supplements) in gastrointestinal diseases. Curr Med Res Opin 2014; 30 (06) 1055-1064
  • 86 Allen SJ, Martinez EG, Gregorio GV et al. Probiotics for treating acute infectious diarrhoea. Cochrane Database Syst Rev 2010; (11) CD003048
  • 87 McFarland LV. Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol 2010; 16 (18) 2202-2222
  • 88 Pichler H, Diridl G, Wolf D. Ciprofloxacin in the treatment of acute bacterial diarrhea: a double blind study. Eur J Clin Microbiol 1986; 5 (02) 241-243
  • 89 Pichler HE, Diridl G, Stickler K et al. Clinical efficacy of ciprofloxacin compared with placebo in bacterial diarrhea. Am J Med 1987; 82 (04) 329-332
  • 90 Dryden MS, Gabb RJ, Wright SK. Empirical treatment of severe acute community-acquired gastroenteritis with ciprofloxacin. Clin Infect Dis 1996; 22 (06) 1019-1025
  • 91 Goodman LJ, Trenholme GM, Kaplan RL et al. Empiric antimicrobial therapy of domestically acquired acute diarrhea in urban adults. Arch Intern Med 1990; 150 (03) 541-546
  • 92 Robert Koch-Institut. Infektionsepidemiologisches Jahrbuch meldepflichtiger Krankheiten für 2013. 2014 Available at: http://www.rki.de/DE/Content/Infekt/Jahrbuch/Jahrbuch_2013.pdf?__blob=publicationFile Accessed October 27, 2014
  • 93 Robert Koch-Institut. Salmonellose (Salmonellen-Gastroenteritis): RKI-Ratgeber für Ärzte. Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Salmonellose.html
  • 94 Olsen SJ, DeBess EE, McGivern TE et al. A nosocomial outbreak of fluoroquinolone-resistant salmonella infection. N Engl J Med 2001; 344 (21) 1572-1579
  • 95 Buchwald DS, Blaser MJ. A review of human salmonellosis: II. Duration of excretion following infection with nontyphi Salmonella . Rev Infect Dis 1984; 6 (03) 345-356
  • 96 Saphra I, Winter JW. Clinical manifestations of salmonellosis in man; an evaluation of 7779 human infections identified at the New York Salmonella Center. N Engl J Med 1957; 256 (24) 1128-1134
  • 97 Helms M, Simonsen J, Molbak K. Quinolone resistance is associated with increased risk of invasive illness or death during infection with Salmonella serotype Typhimurium. J Infect Dis 2004; 190 (09) 1652-1654
  • 98 Onwuezobe IA, Oshun PO, Odigwe CC. Antimicrobials for treating symptomatic non-typhoidal Salmonella infection. Cochrane Database Syst Rev 2012; 11 CD001167
  • 99 Cohen PS, O’Brien TF, Schoenbaum SC et al. The risk of endothelial infection in adults with salmonella bacteremia. Ann Intern Med 1978; 89 (06) 931-932
  • 100 Day LJ, Qayyum QJ, Kauffman CA. Salmonella prosthetic joint septic arthritis. Clin Microbiol Infect 2002; 8 (07) 427-430
  • 101 Guerrant RL, Van Gilder T, Steiner TS et al. Practice guidelines for the management of infectious diarrhea. Clin Infect Dis 2001; 32 (03) 331-351
  • 102 Hohmann EL. Nontyphoidal salmonellosis. Clin Infect Dis 2001; 32 (02) 263-269
  • 103 Gotuzzo E, Guerra JG, Benavente L et al. Use of norfloxacin to treat chronic typhoid carriers. J Infect Dis 1988; 157 (06) 1221-1225
  • 104 Zavala TI, Quiroz C, Gutierrez MA et al. Fluoroquinolones in the treatment of typhoid fever and the carrier state. Eur J Clin Microbiol Infect Dis 1991; 10 (04) 334-341
  • 105 Gonzalez-Escobedo G, Marshall JM, Gunn JS. Chronic and acute infection of the gall bladder by Salmonella Typhi: understanding the carrier state. Nat Rev Microbiol 2011; 9 (01) 9-14
  • 106 Robert Koch-Institut. Shigellose: Ratgeber für Ärzte. Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Shigellose.html
  • 107 Kosek M, Yori PP, Olortegui MP. Shigellosis update: advancing antibiotic resistance, investment empowered vaccine development, and green bananas. Curr Opin Infect Dis 2010; 23 (05) 475-480
  • 108 Butler T. Haemolytic uraemic syndrome during shigellosis. Trans R Soc Trop Med Hyg 2012; 106 (07) 395-399
  • 109 Carter JD, Hudson AP. Reactive arthritis: clinical aspects and medical management. Rheum Dis Clin North Am 2009; 35 (01) 21-44
  • 110 Christopher PR, David KV, John SM et al. Antibiotic therapy for Shigella dysentery. Cochrane Database Syst Rev 2010; (08) CD006784
  • 111 Haltalin KC, Nelson JD, Ring R et al. Double-blind treatment study of shigellosis comparing ampicillin, sulfadiazine, and placebo. J Pediatr 1967; 70 (06) 970-981
  • 112 Hoffmann C, Sahly H, Jessen A et al. High rates of quinolone-resistant strains of Shigella sonnei in HIV-infected MSM. Infection 2013; 41 (05) 999-1003
  • 113 Gu B, Cao Y, Pan S et al. Comparison of the prevalence and changing resistance to nalidixic acid and ciprofloxacin of Shigella between Europe-America and Asia-Africa from 1998 to 2009. Int J Antimicrob Agents 2012; 40 (01) 9-17
  • 114 Khan WA, Seas C, Dhar U et al. Treatment of shigellosis: V. Comparison of azithromycin and ciprofloxacin. A double-blind, randomized, controlled trial. Ann Intern Med 1997; 126 (09) 697-703
  • 115 Bennish ML, Salam MA, Khan WA et al. Treatment of shigellosis: III. Comparison of one- or two-dose ciprofloxacin with standard 5-day therapy. A randomized, blinded trial. Ann Intern Med 1992; 117 (09) 727-734
  • 116 Bassily S, Hyams KC, el-Masry NA et al. Short-course norfloxacin and trimethoprim-sulfamethoxazole treatment of shigellosis and salmonellosis in Egypt. Am J Trop Med Hyg 1994; 51 (02) 219-223
  • 117 Robert Koch-Institut. Campylobacter: Ratgeber für Ärzte. Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Campylobacter.html
  • 118 Kapperud G, Lassen J, Ostroff SM et al. Clinical features of sporadic Campylobacter infections in Norway. Scand J Infect Dis 1992; 24 (06) 741-749
  • 119 Molina J, Casin I, Hausfater P et al. Campylobacter infections in HIV-infected patients: clinical and bacteriological features. AIDS 1995; 9 (08) 881-885
  • 120 Nielsen H, Hansen KK, Gradel KO et al. Bacteraemia as a result of Campylobacter species: a population-based study of epidemiology and clinical risk factors. Clin Microbiol Infect 2010; 16 (01) 57-61
  • 121 Pacanowski J, Lalande V, Lacombe K et al. Campylobacter bacteremia: clinical features and factors associated with fatal outcome. Clin Infect Dis 2008; 47 (06) 790-796
  • 122 Vukelic D, Trkulja V, Salkovic-Petrisic M. Single oral dose of azithromycin versus 5 days of oral erythromycin or no antibiotic in treatment of campylobacter enterocolitis in children: a prospective randomized assessor-blind study. J Pediatr Gastroenterol Nutr 2010; 50 (04) 404-410
  • 123 Lariviere LA, Gaudreau CL, Turgeon FF. Susceptibility of clinical isolates of Campylobacter jejuni to twenty-five antimicrobial agents. J Antimicrob Chemother 1986; 18 (06) 681-685
  • 124 Ostroff SM, Kapperud G, Lassen J et al. Clinical features of sporadic Yersinia enterocolitica infections in Norway. J Infect Dis 1992; 166 (04) 812-817
  • 125 Robert Koch-Institut. Yersiniose: Risikofaktoren in Deutschland. 2012 (Nr. 6). Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Archiv/2012/Ausgaben/06_12.pdf?__blob=publicationFile
  • 126 Bottone EJ. Yersinia enterocolitica: overview and epidemiologic correlates. Microbes Infect 1999; 1 (04) 323-333
  • 127 Press N, Fyfe M, Bowie W et al. Clinical and microbiological follow-up of an outbreak of Yersinia pseudotuberculosis serotype Ib. Scand J Infect Dis 2001; 33 (07) 523-526
  • 128 Frank C, Werber D, Cramer JP et al. Epidemic profile of Shiga-toxin-producing Escherichia coli O104:H4 outbreak in Germany. N Engl J Med 2011; 365 (19) 1771-1780
  • 129 Smith KE, Wilker PR, Reiter PL et al. Antibiotic treatment of Escherichia coli O157 infection and the risk of hemolytic uremic syndrome, Minnesota. Pediatr Infect Dis J 2012; 31 (01) 37-41
  • 130 Menne J, Nitschke M, Stingele R et al. Validation of treatment strategies for enterohaemorrhagic Escherichia coli O104:H4 induced haemolytic uraemic syndrome: case-control study. BMJ 2012; 345
  • 131 Geerdes-Fenge HF, Löbermann M, Nürnberg M et al. Ciprofloxacin reduces the risk of hemolytic uremic syndrome in patients with Escherichia coli O104:H4-associated diarrhea. Infection 2013; 41 (03) 669-673
  • 132 Nitschke M, Sayk F, Härtel C et al. Association between azithromycin therapy and duration of bacterial shedding among patients with Shiga toxin-producing enteroaggregative Escherichia coli O104:H4. JAMA 2012; 307 (10) 1046-1052
  • 133 Lima NL, Guerrant RL, Kaiser DL et al. A retrospective cohort study of nosocomial diarrhea as a risk factor for nosocomial infection. J Infect Dis 1990; 161 (05) 948-952
  • 134 Polage CR, Solnick JV, Cohen SH. Nosocomial diarrhea: evaluation and treatment of causes other than Clostridium difficile. J Infect Dis 2012; 55 (07) 982-989
  • 135 Magill SS, Edwards JR, Bamberg W et al. Multistate point-prevalence survey of health care-associated infections. N Engl J Med 2014; 370 (13) 1198-1208
  • 136 Meyer E, Gastmeier P, Weizel-Kage D et al. Associations between nosocomial meticillin-resistant Staphylococcus aureus and nosocomial Clostridium difficile-associated diarrhoea in 89 German hospitals. J Hosp Infect 2012; 82 (03) 181-186
  • 137 Hensgens MP, Goorhuis A, Dekkers OM et al. All-cause and disease-specific mortality in hospitalized patients with Clostridium difficile infection: a multicenter cohort study. Clin Infect Dis 2013; 56 (08) 1108-1116
  • 138 Vonberg RP, Reichardt C, Behnke M et al. Costs of nosocomial Clostridium difficile-associated diarrhoea. J Hosp Infect 2008; 70 (01) 15-20
  • 139 Robert Koch-Institut. Clostridium difficile: RKI-Ratgeber für Ärzte. Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Clostridium.html?nn=2393714
  • 140 Gorkiewicz G. Nosocomial and antibiotic-associated diarrhoea caused by organisms other than Clostridium difficile. Int J Antimicrob Agents 2009; 33 (Suppl. 01) 37-41
  • 141 Loo VG, Bourgault AM, Poirier L et al. Host and pathogen factors for Clostridium difficile infection and colonization. N Engl J Med 2011; 365 (18) 1693-1703
  • 142 Rea MC, O’Sullivan O, Shanahan F et al. Clostridium difficile carriage in elderly subjects and associated changes in the intestinal microbiota. J Clin Microbiol 2012; 50 (03) 867-875
  • 143 Howell MD, Novack V, Grgurich P et al. Iatrogenic gastric acid suppression and the risk of nosocomial Clostridium difficile infection. Arch Intern Med 2010; 170 (09) 784-790
  • 144 McFarland LV, Mulligan ME, Kwok RY et al. Nosocomial acquisition of Clostridium difficile infection. N Engl J Med 1989; 320 (04) 204-210
  • 145 Bundesamt für Verbraucherschutz und Lebensmittelsicherheit. GERMAP 2012-Bericht über den Antibiotikaverbrauch und die Verbreitung von Antibiotikaresistenzen in der Human- und Veterinärmedizin in Deutschland. Available at: http://www.bvl.bund.de/SharedDocs/Downloads/05_Tierarzneimittel/germap2012.pdf?__blob=publicationFile&v=4
  • 146 Chitnis AS, Holzbauer SM, Belflower RM et al. Epidemiology of community-associated Clostridium difficile infection, 2009 through 2011. JAMA Intern Med 2013; 173 (14) 1359-1367
  • 147 Allen SJ, Wareham K, Wang D et al. Lactobacilli and bifidobacteria in the prevention of antibiotic-associated diarrhoea and Clostridium difficile diarrhoea in older inpatients (PLACIDE): a randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2013; 382 (9900) 1249-1257
  • 148 Bauer TM, Lalvani A, Fehrenbach J et al. Derivation and validation of guidelines for stool cultures for enteropathogenic bacteria other than Clostridium difficile in hospitalized adults. JAMA 2001; 285 (03) 313-319
  • 149 Epple HJ, Zeitz M. Infectious enteritis. Internist (Berl) 2011; 52 (09) 1038, 1040-1044, 1046
  • 150 Bernard H, Hohne M, Niendorf S et al. Epidemiology of norovirus gastroenteritis in Germany 2001–2009: eight seasons of routine surveillance. Epidemiol Infect 2014; 142 (01) 63-74
  • 151 Robert Koch-Institut. Noroviren-Infektionen: RKI-Ratgeber für Ärzte. Available at: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Noroviren.html
  • 152 Cohen SH, Gerding DN, Johnson S et al. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect Control Hosp Epidemiol 2010; 31 (05) 431-455
  • 153 Debast SB, Bauer MP, Kuijper EJ et al. European Society of Clinical Microbiology and Infectious Diseases: update of the treatment guidance document for Clostridium difficile infection. Clin Microbiol Infect 2014; 20 (Suppl. 02) 1-26
  • 154 Johnson S. Recurrent Clostridium difficile infection: a review of risk factors, treatments, and outcomes. J Infect 2009; 58 (06) 403-410
  • 155 Garey KW, Sethi S, Yadav Y et al. Meta-analysis to assess risk factors for recurrent Clostridium difficile infection. J Hosp Infect 2008; 70 (04) 298-304
  • 156 McFarland LV, Surawicz CM, Rubin M et al. Recurrent Clostridium difficile disease: epidemiology and clinical characteristics. Infect Control Hosp Epidemiol 1999; 20 (01) 43-50
  • 157 Pepin J, Alary ME, Valiquette L et al. Increasing risk of relapse after treatment of Clostridium difficile colitis in Quebec, Canada. Clin Infect Dis 2005; 40 (11) 1591-1597
  • 158 Johnson S, Louie TJ, Gerding DN et al. Vancomycin, Metronidazole, or Tolevamer for Clostridium difficile Infection: Results From Two Multinational, Randomized, Controlled Trials. Clin Infect Dis 2014; 59 (03) 345-354
  • 159 Nelson RL, Kelsey P, Leeman H et al. Antibiotic treatment for Clostridium difficile-associated diarrhea in adults. Cochrane Database Syst Rev 2011; (09) CD004610
  • 160 Zar FA, Bakkanagari SR, Moorthi KM et al. A comparison of vancomycin and metronidazole for the treatment of Clostridium difficile-associated diarrhea, stratified by disease severity. Clin Infect Dis 2007; 45 (03) 302-307
  • 161 Surawicz CM, Brandt LJ, Binion DG et al. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am J Gastroenterol 2013; 108 (04) 478-498 ; quiz 499
  • 162 Gonzales M, Pepin J, Frost EH et al. Faecal pharmacokinetics of orally administered vancomycin in patients with suspected Clostridium difficile infection. BMC Infect Dis 2010; 10: 363
  • 163 Fekety R, Silva J, Kauffman C et al. Treatment of antibiotic-associated Clostridium difficile colitis with oral vancomycin: comparison of two dosage regimens. Am J Med 1989; 86 (01) 15-19
  • 164 Lam SW, Bass SN, Neuner EA et al. Effect of vancomycin dose on treatment outcomes in severe Clostridium difficile infection. Int J Antimicrob Agents 2013; 42 (06) 553-558
  • 165 Nerandzic MM, Mullane K, Miller MA et al. Reduced acquisition and overgrowth of vancomycin-resistant enterococci and Candida species in patients treated with fidaxomicin versus vancomycin for Clostridium difficile infection. Clin Infect Dis 2012; 55 (Suppl. 02) S121-S126
  • 166 Cheah AL, Peel T, Howden BP et al. Case-case-control study on factors associated with vanB vancomycin-resistant and vancomycin-susceptible enterococcal bacteraemia. BMC Infect Dis 2014; 14 (01) 353
  • 167 Sethi AK, Al-Nassir WN, Nerandzic MM et al. Skin and environmental contamination with vancomycin-resistant Enterococci in patients receiving oral metronidazole or oral vancomycin treatment for Clostridium difficile-associated disease. Infect Control Hosp Epidemiol 2009; 30 (01) 13-17
  • 168 Donskey CJ, Chowdhry TK, Hecker MT et al. Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. N Engl J Med 2000; 343 (26) 1925-1932
  • 169 Cornely OA, Miller MA, Louie TJ et al. Treatment of first recurrence of Clostridium difficile infection: fidaxomicin versus vancomycin. Clin Infect Dis 2012; 55 (Suppl. 02) S154-S161
  • 170 Louie TJ, Miller MA, Mullane KM et al. Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med 2011; 364 (05) 422-431
  • 171 Friedenberg F, Fernandez A, Kaul V et al. Intravenous metronidazole for the treatment of Clostridium difficile colitis. Dis Colon Rectum 2001; 44 (08) 1176-1180
  • 172 Apisarnthanarak A, Razavi B, Mundy LM. Adjunctive intracolonic vancomycin for severe Clostridium difficile colitis: case series and review of the literature. Clin Infect Dis 2002; 35 (06) 690-696
  • 173 Kim PK, Huh HC, Cohen HW et al. Intracolonic vancomycin for severe Clostridium difficile colitis. Surg Infect (Larchmt) 2013; 14 (06) 532-539
  • 174 Musgrave CR, Bookstaver PB, Sutton SS et al. Use of alternative or adjuvant pharmacologic treatment strategies in the prevention and treatment of Clostridium difficile infection. Int J Infect Dis 2011; 15 (07) e438-e448
  • 175 Neal MD, Alverdy JC, Hall DE et al. Diverting loop ileostomy and colonic lavage: an alternative to total abdominal colectomy for the treatment of severe, complicated Clostridium difficile associated disease. Ann Surg 2011; 254 (03) 423-427 ; discussion 427–429
  • 176 Pepin J, Routhier S, Gagnon S et al. Management and outcomes of a first recurrence of Clostridium difficile-associated disease in Quebec, Canada. Clin Infect Dis 2006; 42 (06) 758-764
  • 177 Kapoor K, Chandra M, Nag D et al. Evaluation of metronidazole toxicity: a prospective study. Int J Clin Pharmacol Res 1999; 19 (03) 83-88
  • 178 McFarland LV, Elmer GW, Surawicz CM. Breaking the cycle: treatment strategies for 163 cases of recurrent Clostridium difficile disease. Am J Gastroenterol 2002; 97 (07) 1769-1775
  • 179 Eiseman AJ, Silen B, Bascom W et al. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery 1958; 44 (05) 854-859
  • 180 Brandt LJ, Aroniadis OC, Mellow M et al. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am J Gastroenterol 2012; 107 (07) 1079-1087
  • 181 van Nood E, Vrieze A, Nieuwdorp M et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med 2013; 368 (05) 407-415
  • 182 KRINKO. Personelle und organisatorische Voraussetzungen zur Prävention nosokomialer Infektionen. 52. 2009: 951-962
  • 183 Valiquette L, Cossette B, Garant MP et al. Impact of a reduction in the use of high-risk antibiotics on the course of an epidemic of Clostridium difficile-associated disease caused by the hypervirulent NAP1/027 strain. Clin Infect Dis 2007; 45 (Suppl. 02) S112-S121
  • 184 Talpaert MJ, Gopal RG, Cooper BS et al. Impact of guidelines and enhanced antibiotic stewardship on reducing broad-spectrum antibiotic usage and its effect on incidence of Clostridium difficile infection. J Antimicrob Chemother 2011; 66 (09) 2168-2174
  • 185 Siegel JD, Rhinehart E, Jackson M et al. 2007 Guideline for Isolation Precautions: Preventing Transmission of Infectious Agents in Health Care Settings. Am J Infect Control 2007; 35 (10) 65-164
  • 186 Jabbar U, Leischner J, Kasper D et al. Effectiveness of alcohol-based hand rubs for removal of Clostridium difficile spores from hands. Infect Control Hosp Epidemiol 2010; 31 (06) 565-570
  • 187 Boyce JM, Ligi C, Kohan C et al. Lack of association between the increased incidence of Clostridium difficile-associated disease and the increasing use of alcohol-based hand rubs. Infect Control Hosp Epidemiol 2006; 27 (05) 479-483
  • 188 Kaier K, Hagist C, Frank U et al. Two time-series analyses of the impact of antibiotic consumption and alcohol-based hand disinfection on the incidences of nosocomial methicillin-resistant Staphylococcus aureus infection and Clostridium difficile infection. Infect Control Hosp Epidemiol 2009; 30 (04) 346-353
  • 189 Rupp ME, Fitzgerald T, Puumala S et al. Prospective, controlled, cross-over trial of alcohol-based hand gel in critical care units. Infect Control Hosp Epidemiol 2008; 29 (01) 8-15
  • 190 Rutala WA, Gergen MF, Weber DJ. Efficacy of different cleaning and disinfection methods against Clostridium difficile spores: importance of physical removal versus sporicidal inactivation. Infect Control Hosp Epidemiol 2012; 33 (12) 1255-1258
  • 191 Dubberke ER, Carling P, Carrico R et al. Strategies to prevent Clostridium difficile infections in acute care hospitals: 2014 Update. Infect Control Hosp Epidemiol 2014; 35 (06) 628-645
  • 192 MacCannell T, Umscheid CA, Agarwal RK et al. Guideline for the prevention and control of norovirus gastroenteritis outbreaks in healthcare settings. Infect Control Hosp Epidemiol 2011; 32 (10) 939-969
  • 193 Sattar SA, Ali M, Tetro JA. In vivo comparison of two human norovirus surrogates for testing ethanol-based handrubs: the mouse chasing the cat!. PLoS One 2011; 6 (02) e17340
  • 194 Goldenberg JZ, Ma SS, Saxton JD et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev 2013; 5 CD006095
  • 195 Kobrynski LJ, Mayer L. Diagnosis and treatment of primary immunodeficiency disease in patients with gastrointestinal symptoms. Clin Immunol 2011; 139 (03) 238-248
  • 196 Krones E, Högenauer C. Diarrhea in the immunocompromised patient. Gastroenterol Clin North Am 2012; 41 (03) 677-701
  • 197 Vaiyavatjamai P, Boitano JJ, Techasintana P et al. Immunocompromised group differences in the presentation of intestinal strongyloidiasis. Jpn J Infect Dis 2008; 61 (01) 5-8
  • 198 Fishman JA. Infections in immunocompromised hosts and organ transplant recipients: essentials. Liver Transpl 2011; 17 (Suppl. 03) 34-37
  • 199 Ghosh K, Weiss LM. Molecular diagnostic tests for microsporidia. Interdiscip Perspect Infect Dis 2009; 2009: 926521
  • 200 Kulkarni SV, Kairon R, Sane SS et al. Opportunistic parasitic infections in HIV/AIDS patients presenting with diarrhoea by the level of immunesuppression. Indian J Med Res 2009; 130 (01) 63-66
  • 201 Yakoob J, Jafri W, Beg MA et al. Blastocystis hominis and Dientamoeba fragilis in patients fulfilling irritable bowel syndrome criteria. Parasitol Res 2010; 107 (03) 679-684
  • 202 Matthes-Martin S, Feuchtinger T, Shaw PJ et al. European guidelines for diagnosis and treatment of adenovirus infection in leukemia and stem cell transplantation: summary of ECIL-4 (2011). Transpl Infect Dis 2012; 14 (06) 555-563
  • 203 Karakousis PC, Moore RD, Chaisson RE. Mycobacterium avium complex in patients with HIV infection in the era of highly active antiretroviral therapy. Lancet Infect Dis 2004; 4 (09) 557-565
  • 204 Cello JP, Day LW. Idiopathic AIDS enteropathy and treatment of gastrointestinal opportunistic pathogens. Gastroenterology 2009; 136 (06) 1952-1965
  • 205 Donoghue HD, Holton J. Intestinal tuberculosis. Curr Opin Infect Dis 2009; 22 (05) 490-496
  • 206 Assi M, McKinsey DS, Driks MR et al. Gastrointestinal histoplasmosis in the acquired immunodeficiency syndrome: report of 18 cases and literature review. Diagn Microbiol Infect Dis 2006; 55 (03) 195-201
  • 207 Fine KD, Seidel RH, Do K. The prevalence, anatomic distribution, and diagnosis of colonic causes of chronic diarrhea. Gastrointest Endosc 2000; 51 (03) 318-326
  • 208 Guarner J, Brandt ME. Histopathologic Diagnosis of Fungal Infections in the 21st Century. Clin Microbiol Rev 2011; 24 (02) 247-280
  • 209 Hommes DW, Sterringa G, van Deventer SJH et al. The pathogenicity of cytomegalovirus in inflammatory bowel disease: a systematic review and evidence-based recommendations for future research. Inflamm Bowel Dis 2004; 10 (03) 245-250
  • 210 Kojima T, Watanabe T, Hata K et al. Cytomegalovirus infection in ulcerative colitis. Scand J Gastroenterol 2006; 41 (06) 706-711
  • 211 Aapro MS, Bohlius J, Cameron DA et al. 2010 update of EORTC guidelines for the use of granulocyte-colony stimulating factor to reduce the incidence of chemotherapy-induced febrile neutropenia in adult patients with lymphoproliferative disorders and solid tumours. Eur J Cancer 2011; 47 (01) 8-32
  • 212 Torres-Madriz G, Boucher HW. Immunocompromised hosts: perspectives in the treatment and prophylaxis of cytomegalovirus disease in solid-organ transplant recipients. Clin Infect Dis 2008; 47 (05) 702-711
  • 213 Sukhrie FHA, Siebenga JJ, Beersma MFC et al. Chronic shedders as reservoir for nosocomial transmission of norovirus. J Clin Microbiol 2010; 48 (11) 4303-4305
  • 214 Sandkovsky U, Vargas L, Florescu DF. Adenovirus: current epidemiology and emerging approaches to prevention and treatment. Curr Infect Dis Rep 2014; 16 (08) 416
  • 215 Gellad ZF, Alexander BD, Liu JK et al. Severity of Clostridium difficile-associated diarrhea in solid organ transplant patients. Transpl Infect Dis 2007; 9 (04) 276-280
  • 216 Gil L, Styczynski J, Komarnicki M. Infectious complication in 314 patients after high-dose therapy and autologous hematopoietic stem cell transplantation: risk factors analysis and outcome. Infection 2007; 35 (06) 421-427
  • 217 Krishna SG, Zhao W, Apewokin SK et al. Risk factors, preemptive therapy, and antiperistaltic agents for Clostridium difficile infection in cancer patients. Transpl Infect Dis 2013; 15 (05) 493-501
  • 218 Escobedo AA, Hanevik K, Almirall P et al. Management of chronic Giardia infection. Expert Rev Anti Infect Ther 2014; 12 (09) 1143-1157
  • 219 Burchard GD, Hentschke M, Weinke T et al. Travelers’ diarrhea. Dtsch Med Wochenschr 2013; 138 (33) 1673-1683 ; quiz 1684–1686
  • 220 Steffen R. Epidemiology of traveler’s diarrhea. Clin Infect Dis 2005; 41 (Suppl. 08) 536-540
  • 221 Swaminathan A, Torresi J, Schlagenhauf P et al. A global study of pathogens and host risk factors associated with infectious gastrointestinal disease in returned international travellers. J Infect 2009; 59 (01) 19-27
  • 222 Flores J, DuPont HL, Lee SA et al. Influence of host interleukin-10 polymorphisms on development of traveler’s diarrhea due to heat-labile enterotoxin-producing Escherichia coli in travelers from the United States who are visiting Mexico. Clin Vaccine Immunol 2008; 15 (08) 1194-1198
  • 223 Mohamed JA, DuPont HL, Flores J et al. Single nucleotide polymorphisms in the promoter of the gene encoding the lipopolysaccharide receptor CD14 are associated with bacterial diarrhea in US and Canadian travelers to Mexico. Clin Infect Dis 2011; 52 (11) 1332-1341
  • 224 DuPont HL, Ericsson CD, Farthing MJG et al. Expert review of the evidence base for self-therapy of travelers’ diarrhea. J Travel Med 2009; 16 (03) 161-171
  • 225 Al-Abri SS, Beeching NJ, Nye FJ. Traveller’s diarrhoea. Lancet Infect Dis 2005; 5 (06) 349-360
  • 226 McGregor AC, Whitty CJM, Wright SG. Geographic, symptomatic and laboratory predictors of parasitic and bacterial causes of diarrhoea in travellers. Trans R Soc Trop Med Hyg 2012; 106 (09) 549-553
  • 227 von Sonnenburg F, Tornieporth N, Waiyaki P et al. Risk and aetiology of diarrhoea at various tourist destinations. Lancet 2000; 356 (9224) 133-134
  • 228 Shah N, DuPont HL, Ramsey DJ. Global etiology of travelers’ diarrhea: systematic review from 1973 to the present. Am J Trop Med Hyg 2009; 80 (04) 609-614
  • 229 Neuberger A, Saadi T, Shetern A et al. Clostridium difficile Infection in travelers – a neglected pathogen?. J Travel Med 2013; 20 (01) 37-43
  • 230 Jiang ZD, Dupont HL, Brown EL et al. Microbial etiology of travelers’ diarrhea in Mexico, Guatemala, and India: importance of enterotoxigenic Bacteroides fragilis and Arcobacter species. J Clin Microbiol 2010; 48 (04) 1417-1419
  • 231 Pandey P, Bodhidatta L, Lewis M et al. Travelers’ diarrhea in Nepal: an update on the pathogens and antibiotic resistance. J Travel Med 2011; 18 (02) 102-108
  • 232 Deutsche Gesellschaft für Tropenmedizin und Internationale Gesundheit (DTG). Leitlinie: Diagnostik und Therapie der Malaria. 2013 Available at: www.dtg.org/fileadmin/user_upload/PDF/LL-Malaria_2013.pdf
  • 233 Weinke T, Liebold I. Intestinal protozoa infections. Dtsch Med Wochenschr 2013; 138 (14) 709-711
  • 234 Taneja N, Mewara A, Kumar A et al. Cephalosporin-resistant Shigella flexneri over 9 years (2001–2009) in India. J Antimicrob Chemother 2012; 67 (06) 1347-1353
  • 235 González-Hein G, Cordero N, García P et al. Molecular analysis of fluoroquinolones and macrolides resistance in Campylobacter jejuni isolates from humans, bovine and chicken meat. Rev Chilena Infectol 2013; 30 (02) 135-139
  • 236 DuPont HL, Jiang ZD, Ericsson CD et al. Rifaximin versus ciprofloxacin for the treatment of traveler’s diarrhea: a randomized, double-blind clinical trial. Clin Infect Dis 2001; 33 (11) 1807-1815
  • 237 Riddle MS, Arnold S, Tribble DR. Effect of adjunctive loperamide in combination with antibiotics on treatment outcomes in traveler’s diarrhea: a systematic review and meta-analysis. Clin Infect Dis 2008; 47 (08) 1007-1014
  • 238 McFarland LV. Meta-analysis of probiotics for the prevention of traveler’s diarrhea. Travel Med Infect Dis 2007; 5 (02) 97-105
  • 239 Takahashi O, Noguchi Y, Omata F et al. Probiotics in the prevention of traveler’s diarrhea: meta-analysis. J Clin Gastroenterol 2007; 41 (03) 336-337
  • 240 Adachi JA, Mathewson JJ, Jiang ZD et al. Enteric pathogens in Mexican sauces of popular restaurants in Guadalajara, Mexico, and Houston, Texas. Ann Intern Med 2002; 136 (12) 884-887
  • 241 Kendall ME, Mody RK, Mahon BE et al. Emergence of salsa and guacamole as frequent vehicles of foodborne disease outbreaks in the United States, 1973–2008. Foodborne Pathog Dis 2013; 10 (04) 316-322
  • 242 Alajbegovic S, Sanders JW, Atherly DE et al. Effectiveness of rifaximin and fluoroquinolones in preventing travelers’ diarrhea (TD): a systematic review and meta-analysis. Syst Rev 2012; 1: 39
  • 243 DuPont HL, Ericsson CD, Farthing MJG et al. Expert review of the evidence base for prevention of travelers’ diarrhea. J Travel Med 2009; 16 (03) 149-160
  • 244 López-Gigosos R, Campins M, Calvo MJ et al. Effectiveness of the WC/rBS oral cholera vaccine in the prevention of traveler’s diarrhea: a prospective cohort study. Hum Vaccin Immunother 2013; 9 (03) 692-698
  • 245 Weinke T, Liebold I, Burchard GD et al. Prophylactic immunisation against traveller’s diarrhoea caused by enterotoxin-forming strains of Escherichia coli and against cholera: does it make sense and for whom?. Travel Med Infect Dis 2008; 6 (06) 362-367
  • 246 Dobbins W. Whipple`s disease. In: Springfield, IL: Charles C. Thomas
  • 247 Feurle GE, Junga NS, Marth T. Efficacy of ceftriaxone or meropenem as initial therapies in Whipple’s disease. Gastroenterology 2010; 138 (02) 478-486 ; quiz 11–12
  • 248 Feurle GE, Moos V, Bläker H et al. Intravenous ceftriaxone, followed by 12 or three months of oral treatment with trimethoprim-sulfamethoxazole in Whipple’s disease. J Infect 2013; 66 (03) 263-270
  • 249 Schneider T, Moos V, Loddenkemper C et al. Whipple’s disease: new aspects of pathogenesis and treatment. Lancet Infect Dis 2008; 8 (03) 179-190
  • 250 Marth T, Raoult D. Whipple’s disease. Lancet 2003; 361 (9353) 239-246
  • 251 Lagier JC, Lepidi H, Raoult D et al. Systemic Tropheryma whipplei: clinical presentation of 142 patients with infections diagnosed or confirmed in a reference center. Medicine (Baltimore) 2010; 89 (05) 337-345
  • 252 Geelhaar A, Moos V, Schinnerling K et al. Specific and nonspecific B-cell function in the small intestines of patients with Whipple’s disease. Infect Immun 2010; 78 (11) 4589-4592
  • 253 Fenollar F, Lepidi H, Raoult D. Whipple’s endocarditis: review of the literature and comparisons with Q fever, Bartonella infection, and blood culture-positive endocarditis. Clin Infect Dis 2001; 33 (08) 1309-1316
  • 254 Geissdörfer W, Moos V, Moter A et al. High frequency of Tropheryma whipplei in culture-negative endocarditis. J Clin Microbiol 2012; 50 (02) 216-222
  • 255 Lepidi H, Fenollar F, Dumler JS et al. Cardiac valves in patients with Whipple endocarditis: microbiological, molecular, quantitative histologic, and immunohistochemical studies of 5 patients. J Infect Dis 2004; 190 (05) 935-945
  • 256 von Herbay A. Whipple’s disease. Histologic diagnosis after the discovery of Tropheryma whippelii. Pathologe 2001; 22 (01) 82-88
  • 257 von Herbay A, Maiwald M, Ditton HJ et al. Histology of intestinal Whipple’s disease revisited. A study of 48 patients. Virchows Arch 1996; 429 (06) 335-343
  • 258 Edouard S, Fenollar F, Raoult D. The rise of Tropheryma whipplei: a 12-year retrospective study of PCR diagnoses in our reference center. J Clin Microbiol 2012; 50 (12) 3917-3920
  • 259 Fenollar F, Fournier PE, Robert C et al. Use of genome selected repeated sequences increases the sensitivity of PCR detection of Tropheryma whipplei. J Clin Microbiol 2004; 42 (01) 401-403
  • 260 Fenollar F, Raoult D. Molecular techniques in Whipple’s disease. Expert Rev Mol Diagn 2001; 1 (03) 299-309
  • 261 Moter A, Schmiedel D, Petrich A et al. Validation of an rpoB gene PCR assay for detection of Tropheryma whipplei: 10 years’ experience in a National Reference Laboratory. J Clin Microbiol 2013; 51 (11) 3858-3861
  • 262 Raoult D, Fenollar F, Rolain JM et al. Tropheryma whipplei in children with gastroenteritis. Emerging Infect Dis 2010; 16 (05) 776-782
  • 263 Keita AK, Brouqui P, Badiaga S et al. Tropheryma whipplei prevalence strongly suggests human transmission in homeless shelters. Int J Infect Dis 2013; 17 (01) e67-e68
  • 264 Fenollar F, Trani M, Davoust B et al. Prevalence of asymptomatic Tropheryma whipplei carriage among humans and nonhuman primates. J Infect Dis 2008; 197 (06) 880-887
  • 265 Panegyres PK. Diagnosis and management of Whipple’s disease of the brain. Pract Neurol 2008; 8 (05) 311-317
  • 266 Touitou V, Fenollar F, Cassoux N et al. Ocular Whipple’s disease: therapeutic strategy and long-term follow-up. Ophthalmology 2012; 119 (07) 1465-1469
  • 267 Lagier JC, Fenollar F, Lepidi H et al. Treatment of classic Whipple’s disease: from in vitro results to clinical outcome. J Antimicrob Chemother 2014; 69 (01) 219-227
  • 268 Feurle GE, Marth T. An evaluation of antimicrobial treatment for Whipple’s Disease. Tetracycline versus trimethoprim-sulfamethoxazole. Dig Dis Sci 1994; 39 (08) 1642-1648
  • 269 Fleming JL, Wiesner RH, Shorter RG. Whipple’s disease: clinical, biochemical, and histopathologic features and assessment of treatment in 29 patients. Mayo Clin Proc 1988; 63 (06) 539-551270
  • 270 Feurle GE, Moos V, Schneider T et al. The combination of chloroquine and minocycline, a therapeutic option in cerebrospinal infection of Whipple’s disease refractory to treatment with ceftriaxone, meropenem and co-trimoxazole. J Antimicrob Chemother 2012; 67 (05) 1295-1296
  • 271 Fenollar F, Rolain JM, Alric L et al. Resistance to trimethoprim/sulfamethoxazole and Tropheryma whipplei. Int J Antimicrob Agents 2009; 34 (03) 255-259
  • 272 Lagier JC, Fenollar F, Lepidi H et al. Evidence of lifetime susceptibility to Tropheryma whipplei in patients with Whipple’s disease. J Antimicrob Chemother 2011; 66 (05) 1188-1189
  • 273 Fenollar F, Lepidi H, Gérolami R et al. Whipple disease associated with giardiasis. J Infect Dis 2003; 188 (06) 828-834
  • 274 Feurle GE, Moos V, Schinnerling K et al. The immune reconstitution inflammatory syndrome in whipple disease: a cohort study. Ann Intern Med 2010; 153 (11) 710-717