FBH PHD Afhandling 2014

159
Copenhagen 2014 1 FACULTY OF SCIENCE UNIVERSITY OF COPENHAGEN PhD thesis Frederik Boëtius Hertz ESBL-Producing Escherichia coli: Antibiotic Selection, Risk Factors and Population Structure. This thesis has been submitted to the PhD School of The Faculty of Science, University of Copenhagen’ Academic advisor: Anders Løbner-Olesen Academic advisor: Niels Frimodt-Møller Submitted: 29-09-2014

Transcript of FBH PHD Afhandling 2014

Copenhagen 2014

1

F A C U L T Y O F S C I E N C E UNIVERSITY OF COPENHAGEN

PhD thesis Frederik Boëtius Hertz

ESBL-Producing Escherichia coli: Antibiotic Selection, Risk

Factors and Population Structure.

This thesis has been submitted to the PhD School of The Faculty of Science, University of

Copenhagen’

Academic advisor: Anders Løbner-Olesen

Academic advisor: Niels Frimodt-Møller

Submitted: 29-09-2014

PhD Dissertation

Institutnavn: Biologisk Institut

Name of department: Functional genomics

Author: Frederik Boëtius Hertz

Title: ESBL-Producing Escherichia coli: Antibiotic Selection,

Risk Factors and Population Structure.

Academic advisor: Anders Løbner-Olesen

Academic advisor: Niels Frimodt-Møller

Submitted: 29th of September 2014

Copenhagen 2014

3

Table of Contents

Preface 5

Abbreviations 6

List of manuscripts 7

Abstract 8

Dansk resume 9

1. Introduction 10

1.1. Research objectives 11

1.2. Outline of dissertation 12

2. Background 13

2.1. Escherichia coli 13

2.1.1. Genomic Diversity of Escherichia coli 13

2.1.2. Extraintestinal Intestinal Pathogenic E.coli 14

2.2. Antibiotic Resistance in E.coli 14

2.2.1. Extended Spectrum Beta-Lactamases 15

2.2.2. Prevalence of ESBL-producing E.coli 16

2.3. Epidemiology of Resistance in E.coli 18

2.3.1. Clinical Consequence of Resistance 19

2.4. The Gastrointestinal Microflora 19

2.4.1. E.coli and Colonization 20

2.5. Reservoirs of ESBL-producing ExPEC 20

2.5.1. The Human Gut as Reservoir 21

2.5.2. Animals, Water and Food 21

2.6. Animal Experimental Models for Investigation of Colonization 22

2.7. Composition of ExPEC Susceptibility Populations 23

2.8. Risk factors for infection with ESBL-producing E.coli 25

2.8.1. Considerations on Study Design 25

2.8.2. Case-Control Studies 26

2.8.3. Double-case control study of epidemiological factors 27

2.8.4. Case-control study in low prevalence countries 28

3. Bacteriological Typing of E.coli 29

3.1. General Considerations on Typing Methods 29

3.2. Phylogrouping by PCR 29

3.3. Multi-Locus Sequence Typing 30

3.4. Multi-Locus Variable Number of Tandem Repeats Analysis 31

3.5. Serotyping 31

4. Material & Methods 33

4.1. General Protocols 33

PhD Dissertation

4.1.1. Research Ethical Approvals 33

4.1.2. CPR Registry 34

4.1.3. Statistics Denmark 34

4.2. Mouse Intestinal Colonization Model (Manuscript I) 35

4.2.1. Selection of E.coli Isolates and Preliminary Mice Experiments 35

4.3. Study Populations (Manuscript II and III) 35

4.3.1. Sample Size Determination 35

4.3.2. Strain Collection 36

4.4. Epidemiology of Collected Isolates 36

5. Results 37

5.1. Mouse intestinal colonization model (Manuscript I) 37

5.1.1. Effect of Antibiotic Selective Pressure on the Microflora 38

5.1.2. Effect of Antibiotic Selective Pressure on Prolonged Presence of 65-Ec-09 38

5.1.3. Molecular Tests of Bacteria Isolated from Mice Faeces 39

5.2. Case-control study (Manuscript II) 43

5.2.1. Urine Samples to DCM 43

5.2.2. Description of E.coli Populations 45

5.2.3. Statistical Analysis of Risk Factors and Population Data 47

5.3. Characterization of E.coli populations (Manuscript III) 49

5.3.1. Distribution of Serogroups, MLVA codes and Sequence Types 49

6. Discussion 56

6.1. Selection of CTX-M-producing E.coli in vivo 56

6.2. Investigation of Epidemiological Factors 57

6.3. Characterization of E.coli Populations 58

7. Conclusion and Perspectives 59

8. Reference 64

9. Appendix 86

Copenhagen 2014

5

Preface

The studies included in this PhD dissertation were carried out at Department of Microbiology and

Infection Control at Statens Serum Institut (SSI) and at Department of Clinical Microbiology, Hvidovre

University Hospital (DCM). The research was carried out as part of PAR, an EU FP7-Health-2009-Single

Stage Project, with support from The Danish Centre for Antibiotic Research and Development

(DanCARD).

Work has been interrupted by two periods of paternity leave, each of three and half months.

At SSI and DCM I have worked with several skilled and generous people. I am very thankful to the many

people who have kindly supported me and contributed greatly to this PhD thesis.

First I would like to extend my gratitude to my supervisors Anders Løbner-Olesen and Niels Frimodt-

Møller for their exceptional guidance and patience as well as the good-humoured atmosphere at all times.

Your support and confidence have made this work possible.

I would like to thank the medical staff and laboratory technicians at SSI and DCM for the technical

assistance and tolerance throughout the three years of research. I must also thank Lars Villiam Pallesen

and Christian Østergaard Andersen for use of department facilities and support.

Pia Jeannette Littauer, Jenny Dahl Knudsen and especially Kristian Schønning have all engaged in

excellent scientific discussions and provided appreciated guidance and aid.

Finally, I would like to thank many of my closest current and former colleagues Anne Sandberg-Schaal,

Lotte Jakobsen, Alexandra Bugay Medina, Mette Pinholt, Steen Christian Rasmussen, Karen Leth

Nielsen and Rikke Fleron Leihof for their friendship and help during this PhD. I would especially like

thank Jesper Boye Nielsen for, not only his friendship, but also his immense efforts and contribution to

my work in the laboratory and scientific deliberations.

My love and warmest gratitude go to my wife Christin for her support and encouragement.

Frederik Boëtius Hertz, August 2014

PhD Dissertation

Abbreviations

a-MLVA Abbreviated MLVA

COI Community-onset infections

CPD-R Cefpodoxime resistance

ESBL Extended-Spectrum-Beta-lactamase

ESCMID European Society for Microbiology and Infectious Diseases

ExPEC Extraintestinal pathogenic Escherichia coli

GP General Practitioner

HCAI Healthcare-associated infections

HGT Horizontal gene transfer

MALDI-TOF Matrix-Assisted Laser Desorption Ionization-Time of Flight Mass Spectrometry

MDR Multidrug resistant

MLST Multi locus sequence typing

MLVA Multi locus variable number of tandem repeat analysis

MNEC Meningitis-associated E.coli

PCR Polymerase chain reaction

PFGE Pulsed field gel-electrophoresis

SSI Statens Serum Institut

SLV Single locus variant

ST Sequence type

UTI Urinary Tract infection

UPEC Uropathogenic E.coli

VF Virulence factor

VNTR Variable number of tandem repeats

Copenhagen 2014

7

List of Manuscripts

Paper I: Hertz F.B., Løbner-Olesen A. and Frimodt-Møller N.

“Antibiotic selection of E.coli ST131 in a mouse intestinal colonization

model”. Antimicrobial Agents and Chemotherapy., 2014. Vol. 58 (10)

Manuscript II: Hertz F.B., Littauer P., Schønning K., Knudsen J.D., Løbner-Olesen A.

and Frimodt-Møller N.

“Epidemiological factors associated with ESBL- or non ESBL-

producing E.coli causing UTI in general practices.”

Manuscript III: Hertz F.B., Nielsen J.B., Littauer P., Schønning K., Knudsen J.D.,

Løbner-Olesen A., Frimodt-Møller N.

“Population structure of Drug-Susceptible, -Resistant and ESBL-

producing Escherichia coli Populations from Community-Acquired

Urinary Tract Infections as Characterized by Abbreviated MLVA,

MLST and Serogrouping in Denmark”

PhD Dissertation

Abstract

Urinary tract infection (UTI) is one the most common bacterial infections and is regularly treated in

primary health care. The most common cause of UTI is extraintestinal pathogenic Escherichia coli

(ExPEC) already present in the intestinal microflora, often as the dominating strain. Resistance in E.coli

is increasing and especially isolates producing Extended-Spectrum Beta-Lactamases (ESBL) have been

reported worldwide. Treatment of UTI is usually initiated by the general practitioners and a significant

proportion of clinical isolates are now resistant to first line antibiotics. The global dissemination of

resistant E.coli has in particular been driven by the spread of a few specific E.coli-lineages and it seems

that there is a difference between the sequence types found among resistant E.coli, ESBL-producing

E.coli and antibiotic susceptible E.coli. The overall objectives of this thesis were to investigate (i)

antibiotics involved in selection of ESBL-producing E.coli, in an experimental mouse model in vivo, (ii)

risk factors for UTI with ESBL-producing E.coli and (iii) to describe the phylogenetic composition of

E.coli populations with different resistance patterns.

We found that different antibiotics can select for the ESBL-producing E.coli, even anti-Gram-

positive antibiotics as dicloxacillin and clindamycin showed selective abilities. While dicloxacillin

has no effect on anaerobic Gram-negatives, this is the case for clindamycin. The selective abilities

of other beta-lactam antibiotics varied, with selection identified by cefotaxime, cefuroxime and

penicillin. The triple-case control study showed that exposure to antibiotics is not a good predictor

for risk of UTI with ESBL-producing E.coli. There were few differences between the case groups

when compared to the uninfected group. However, when case groups where compared to each

other, healthcare association and hospital admission proved to be independent risk factors for UTI

with ESBL-producing E.coli. When typing uropathogenic E.coli, we less frequently saw ST131

among non-ESBL than among ESBL-producing E.coli. We found that ESBL-producing E.coli,

resistant E.coli and susceptible E.coli in turn were dominated by different MLVA codes and

sequence types. Overall the susceptible E.coli was a much more diverse group, whereas resistant

and ESBL-producing E.coli were found in larger clusters, indicating that the success of resistant

lineages like O25b-ST131 is mainly due to positive selection of previously specialized UPEC with

newly gained resistance.

Copenhagen 2014

9

Abstract – Danish

Urinvejsinfektion (UVI) er en af de hyppigste bakterielle infektioner og de behandles typisk i almen

praksis. Den mest almindelige årsag til UVI er ekstra intestinale patogene Escherichia coli (ExPEC), der

forud for infektion ofte er til stede i den intestinale microflora. Resistens blandt E.coli isolater er stigende

og især isolater der producer beta-lactamaser med udvidet spektrum (ESBL) er beskrevet over hele

verden. Behandling af UVI håndteres og påbegyndes ofte af den praktiserende læge og en betydelig del af

de kliniske isolater nu er resistente over for almindeligt benyttede antibiotika. Denne globale spredning af

resistente E.coli har i høj grad været drevet af spredning af få E.coli-kloner og der er tilsyneladende en

forskel på de sekvens typer der ses blandt resistente E.coli, ESBL-producerende E.coli og fuldt følsomme

E.coli. Formålet med denne ph.d. afhandling var, at undersøge (i) hvilke almindeligt benyttede antibiotika

der er involveret i selektion af ESBL-producerende E.coli in vivo, (ii) risiko faktorer for UVI med ESBL-

producerende E.coli og endelig (iii) at beskrive den fylogenetiske sammensætning af E.coli populationer

med forskellige resistens mønstre.

Vi fandt, at forskellige antibiotika kan selektere for ESBL-producerende E.coli, selv anti-Gram-

positive antibiotika som dicloxacillin og clindamycin selekterede. Clindamycin har stor effekt på

anaerobe Gram-negative, mens dicloxacillin ingen effekt har. De selektive egenskaber af andre

beta-lactam antibiotika varierede, med positiv selektion observeret fra cefotaxim, cefuroxim og

penicillin. Case kontrol studiet viste, at antibiotikaforbrug ikke er en god variabel til at

risikostratificere patienter for infektion med ESBL-producerende E.coli. Der var kun få forskelle

mellem case grupperne når disse blev sammenlignet med den ikke inficerede gruppe. Når case

grupper blev sammenlignet med hinanden så vi dog, at kontakt med sundhedsvæsenet og hospitals

indlæggelser var risiko faktorer for UVI med ESBL-producerende E.coli. Ved den bakterielle

typning af de uropatogene E.coli, så vi at ST131 var langt mindre hyppige blandt ikke-ESBL end

blandt de ESBL-producerende E.coli. Vi fandt at ESBL-producerende E.coli, resistente E.coli og

følsomme E.coli var domineret af forskellige MLVA koder og sekvens typer. Alt i alt var de

følsomme E.coli en langt mere diverse gruppe, mens de resistente E.coli og især de ESBL-

producerende E.coli blev identificeret i større grupper, hvilket indikerer at den globale succes for

resistente linjer, som O25b-ST131, i høj grad skyldes det positive selektions tryk, der selekterer

uropatogene E.coli med erhvervet resistens.

PhD Dissertation

1. Introduction

Urinary tract infection (UTI) is one the most common bacterial infections and it is regularly treated in

primary health care by general practitioners (GPs) (1–3). The highest incidence of UTI is found in young,

healthy women, but bacterial infection of the urinary tract is seen in all ages and in both genders (3). The

most common cause of UTI are extraintestinal pathogenic Escherichia coli (ExPEC), causing more than

70% of community-acquired UTI (1,4,5). Resistance in E.coli is increasing and especially isolates

producing Extended-Spectrum Beta-Lactamases (ESBL) have, in the last decades, been reported

worldwide(3,6). Treatment of UTI is usually initiated by the GP and a significant proportion of clinical

isolates are now resistant to first line antibiotics (3,6). This poses a great challenge for the empirical

treatment of infections, caused by E.coli (3,6).

The global dissemination of resistant E.coli have in particular been driven by the spread of a few specific

E.coli-lineages, best characterized by Multi-Locus Sequence Typing (MLST) (7–10). It seems that there

is a difference between the sequence types (STs) found among resistant E.coli, ESBL-producing E.coli

and antibiotic susceptible E.coli, illustrated by recurrent findings of explicit STs in each population (7–9).

UTI is most often caused by an E.coli strain already present in the intestinal microflora, often as the

dominating strain, and as such the human intestinal tract pose a reservoir for ExPEC (11,12).

Additionally, up to 80% of recurrent UTI are caused by the index strain and 25-40% of women

experience a new case of UTI within 6 months (13,14).

Overall risk factors for UTI have been shown to be female gender, previous UTI, sexual activity and

recent exposure to antibiotics, with exposure to antibiotics also posing a major risk for UTI with resistant

bacteria (3,5,11). With the great increase in ESBL-producing E.coli, found to be the reason for higher

morbidity, prolonged hospitalization and increased health care costs, there has been an interest in the

epidemiological factors associated with infections caused by ESBL E.coli. Thus, risk factors have been

widely investigated and reports have, among other, identified age > 65 years, male sex, previous UTI, use

of antibiotics and hospitalization as common risk factors (15–18). Moreover diet, lifestyle and traveling

have been proposed as sources for colonization and infection (17–19).

Copenhagen 2014

11

1.1. Research Objectives

The overall objectives of the present dissertation were to investigate antibiotics involved in

selection of ESBL-producing E.coli, risk factors for UTI with ESBL-producing E.coli through focus

on usage of antibiotics and finally to describe E.coli populations to identify differences in the

phylogenetic compositions among resistant and susceptible E.coli.

The main objectives were therefore:

Objective I:

To evaluate the ability of nine common antibiotics, to select for a CTX-M-15-producing E.coli

isolate belonging to sequence type (ST) 131 in vivo. This was done in a mouse intestinal

colonisation model developed and tested as part of this research.

Objective II:

To investigate epidemiological factors associated with acquiring UTI with ESBL-producing E.coli,

in general practices, investigated as a case-control study involving three case groups and a single

control group.

Objective III:

To investigate if the population structure of uropathogenic E.coli populations is predominantly

composed of related isolates, regardless of resistance profile. This was done by typing of three

susceptibility groups by use of a-MLVA, MLST, serogrouping and ESBL-genotyping.

PhD Dissertation

1.2. Outline of dissertation

This thesis consists of six chapters. First, a literature review provides an overview of ExPEC,

antibiotic resistance in E.coli, possible reservoirs, previously used experimental mouse models,

known population structure of E.coli populations and risk factors for UTI with ESBL-producing

E.coli. This is followed by a brief review of typing methods used in this thesis. Secondly, a brief

method section describes methods and study designs not described in manuscript I-III, followed by

a presentation of main findings. Finally, the thesis concludes with a short discussion of main results

and an overall conclusion with future perspectives. References are presented before appendix´s are

shown and the thesis concludes with an inclusion of Manuscript I-III.

Copenhagen 2014

13

2. Background

2.1. Escherichia coli

Escherichia coli (E.coli) is a member of the family Enterobacteriaceae and is a Gram-negative,

facultative anaerobic bacterium (20). It is a highly versatile bacterial species comprised of both

harmless commensal strains and different pathogenic variants with the ability to cause either

intestinal or extraintestinal diseases (21,22). Consequently, E. coli strains are broadly classified into

three major groups of commensal E.coli, intestinal pathogenic E.coli (IPEC) and extraintestinal

pathogenic E.coli (ExPEC) (21–23). As a non-pathogenic inhabitant of the intestine of many

mammals, including humans, E.coli exists as part of the indigenous flora, often contributing to the

vital tasks performed by the intestinal microflora (20,24). Traditionally, commensal E.coli have

been described as colonizers that rarely cause infection and categorized as belonging to phylogroup

A and B1, while ExPEC isolates are mostly derived from phylogroup B2 and D (20,22,25). All four

phylogroups can, however, cause intestinal and extraintestinal infections and phylogroup B2 and D

have been found as regular colonizing strains in healthy individuals (11,20,26).

The pathogenic E.coli, IPEC and ExPEC, can each be further subcategorized into specific

pathotypes. This classification is based on clinical manifestations of disease and the pathogenic

traits such, as presence of virulence factors (VFs) (20). The most prevalent ExPEC pathotypes are

the uropathogenic E.coli (UPEC) and meningitis-associated E.coli (MNEC) (27). Often intestinal

non-pathogenic E.coli and IPEC can be distinguished by genome content and phenotypic traits, but

the discrimination between commensal E.coli and extraintestinal pathogens is not easy (21). ExPEC

strain are habitually found as part of the commensal flora of healthy individuals without causing

enteric disease (20,23). While IPEC cause diseases of the intestinal tract, ExPEC can cause a range

of diseases in almost any anatomical niche such as UTI, bacteraemia, meningitis and intra-

abdominal infections (27–29).

2.1.1. Genomic Diversity of Escherichia coli

In E.coli an extensive horizontal gene transfer (HGT) by plasmids occurs, contributing to a

noticeable genome plasticity in the species (20,21). As a result and in combination with genomic

recombination, E.coli has an amazing ability to colonize and survive in many different locations

(10,20). The genome is composed of the conserved core of genes, constituting the genetic

information for most essential cellular processes, and a flexible gene pool often consisting of

PhD Dissertation

genetic information providing the abilities to adapt to new environmental conditions (10). The

acquisition of new genomic material therefore contributes to the rapid evolution of E. coli lineages

(21). Thus, E.coli is a versatile species with a remarkable adaptive potential, alternating between

habitats and adapting rapidly to new and complex ecosystems (30,31).

2.1.2. Extra Intestinal Pathogenic Escherichia coli

Extraintestinal infections in humans have a high incidence and ExPEC is the most common Gram-

negative extraintestinal pathogen. The most frequent infection is UTI, but E.coli is also the leading

cause of bloodstream infections (9,25,32,33).

The term ExPEC was introduced by Johnson et al. in 2000 based on reports of UPEC and MNEC

isolates causing a range of extraintestinal infections (23). Several presumed virulence

genes were linked to the pathogenicity of ExPEC, enabling them to invade almost any

extraintestinal tissue (22,23). Many of these VFs are present on the chromosome, but VFs are seen

extensively on mobile elements, creating great diversity within the categories of ExPEC pathotypes

(20,24,30). Among ExPEC there is an increasing recognition of isolates resistant to antibiotics, the

most important being isolates producing plasmid-mediated β-lactamases (22).

2.2. Antibiotic resistance in E.coli

Generally, there are five main methods by which Gram-negative organism develop

resistance(34,35):

First, bacteria can carry genes coding for enzymes, such as beta-lactamases, hydrolysing and

inactivating beta-lactam antibiotics. Second, mutations can occur in the genes for binding sites for

antibiotics changing the specific target or its function. Third, alterations of the membrane porins

result in reduced permeability. Fourth, bacteria can express efflux pumps to actively transport

antibiotics out of the cell and finally, fifth, alternate metabolic pathways can by-pass paths inhibited

by antibiotics (34,35). Resistance in Gram-negative bacteria can be intrinsic, arise or be acquired

and is often composed of a combination of resistance mechanism like beta-lactamases, porin

deletions and efflux pumps (36–40). The predominant mechanism of resistance is, however, the

hydrolysis of the antibiotic by beta-lactamases (41). The ability to produce β-lactamases, including

ESBL, is frequently acquired through large plasmids holding many different genes coding

resistance against other antibiotic classes, contributing to MDR (41,42). This also means that many

different antibiotics may facilitate selection of, and colonization by, these pathogens and that the

Copenhagen 2014

15

same types of ESBLs may be recognized in clonally unrelated isolates or isolates of the same clonal

origin may encode different types of ESBLs (42). E.coli is one of the organisms most frequently

found harbouring ESBL-genes and MDR in ESBL-producing Enterobacteriaceae is rapidly

becoming a threat to the medical community (9,25,32,33,42–47).

2.2.1. Extended Spectrum Beta-Lactamases

Beta-lactams are the most widely used class of antibiotics – all characterized by having a β-lactam ring as

the chemical base and include penicillins, cephalosporins, carbapenems, monobactams and

cephamycins(48). Hydrolysis by enzymes is the most common mechanism for resistance towards this

class of antibiotics (22,49,50). Beta-lactamases belong to a very large, diverse family of enzymes capable

of hydrolysing β-lactam antibiotics by cutting open the β-lactam ring and the hydrolysing ability of β-

lactamases can be of narrow or broad spectres. Several hundred β-lactamases have been described

(http://www.lahey.org/Studies/) each with a defined spectrum (examples: penicillinases and

cephalosporinases). These enzymes are divided into molecular classes based on their amino acid

similarities (Ambler scheme A to D = Classes) or into groups by functional similarities (Bush-Jacoby-

Medeiros scheme = Groups 1-4). Ambler class A, C and D are serine- β-lactamases and class B are

metallo- β-lactamases (51). Finally, several families of β-lactamase exist, with the most common plasmid-

mediated β-lactamases being TEM and SHV, accounting for >60% of E.coli resistance to ampicillin

(41,51).

A major source for resistance in E.coli are plasmid-borne Extended-Spectrum β-Lactamases

(ESBL), which are broadly classified as enzymes capable of hydrolysing most β-lactams such as

penicillins, extended-spectrum cephalosporins and monobactams, but not cephamycins and

carbapenems. In addition, ESBLs are inhibited by β-lactamase inhibitors such as clavulanic acid,

sulbactam, and tazobactam, and are carried on large plasmids often containing various other genes,

causing resistance to other classes of antibiotics (47,50,52,53). Several ESBL families have been

described, but the majority of ESBLs belong to the four large families of SHV, TEM, CTX-M and

OXA (53,54).

The first ESBL was a SHV type detected in K.pneumoniae in 1983 in Germany and around the

same time the first TEM ESBLs were found in France and England (51). For unknown reasons,

K.pneumoniae was the bacterial species most likely to express ESBLs in the 1980s and 1990s.

These ESBLs were most frequently derivatives of common penicillinases like TEM-1 and SHV-1,

generated by point mutations creating an extended-spectrum of hydrolysis (6,42). In 1989 an ESBL

PhD Dissertation

CTX-M enzyme was described in Germany (named for cefotaxime resistance and place of

discovery, Munich) (51). CTX-M enzymes are believed to have originated from Kluyvera spp. and

are divided into five groups (CTX-M group 1, 2, 8, 9 and 25) again with several subgroups. With

the emergence of different CTX-M types the prevalence of ESBLs changed dramatically during the

early 2000s. Rapidly CTX-M types surpassed SHV and TEM as the dominating ESBL, most often

present in E.coli (22). Now, CTX-M ESBLs display a global distribution causing infections in every

part of the world (10,42,45,49).

2.2.2. Prevalence of ESBL-producing E.coli

With the emergence of CTX-M ESBLs, community-onset ESBL infections have become an important

public health issue, primarily caused by E. coli producing CTX-M type ESBLs (10,22,42). The rapid

worldwide dissemination of this particular ESBL type has been known as the “CTX-M pandemic” and the

dominance of CTX-M types ESBL are, largely, caused by dissemination of E.coli lineages, often

expressing co-resistance to other classes of antibiotics (10,41,45). It has become evident, that once a

CTX-M type enters an area, it becomes prevalent, replacing TEM and SHV as the dominating ESBL (49).

In 2007, a study based on TEST global surveillance database reported that the incidence of ESBL-

producing E.coli was highest among isolates collected in Latin America (13.5%) and Asia (12%)

followed by Europe (7.5%) and North America (2.2%) (42,55). As seen, there is a noticeable differences

in ESBL prevalence, a variation also seen in dominating CTX-M subtypes between European countries

and different parts of the world, as depicted by Hawkey and Jones in 2009 (Figure 1) (42,55). Examples

include CTX-M-1 in Italy and the Netherlands, CTX-M-2 in Argentina and Israel, CTX-M-3 enzymes in

Poland, CTX-M-9 in Spain, CTX-M-14 in China and CTX-M-15 in UK and Denmark. Nevertheless,

CTX-M-14 and -15 producing E.coli are distributed around the world and CTX-M-15 is the most

prevalent type (10,22,42,45,49,56,57).

Looking at the ESBL-prevalence in Denmark there has been a slow, but steady, increase in number of

infections caused by ESBL-producing E.coli. In clinical isolates from 1997 there were no ESBL-

producing E.coli found (58). In 2003, 0.8% of E.coli isolates were ESBL-producing and cefuroxime

resistance was found in <5% of E.coli isolates in the years 2003-2006 (59). However, a study on E.coli

isolates from 2007, reported the UPEC ESBL-prevalence to be 1.5% from general practices and 2.3% in

hospital urine, with 60% of ESBL-producing E.coli producing CTX-M-15 (60). Resistance to extended-

spectrum cephalosporins in UPEC from primary health-care, used as a marker for ESBL-production, was

found to be 4% in 2012 (61).

Copenhagen 2014

17

Figure 1. Worldwide distribution of different classes of CTX-M β-lactamases (first identified in 1989). Davies J , and Davies

D Microbiol. Mol. Biol. Rev. 2010;74:417-433

PhD Dissertation

2.3. Epidemiology of Resistance in E.coli

An important feature complicating treatment of infections caused by E.coli is the increase in

resistance to first-line antibiotics (22,61,62). Until the late 1990s ExPEC were relatively susceptible

to first-line drugs (22). Currently, resistance in Gram-negative bacteria constitutes one of the

biggest challenges to public health and the changes in antimicrobial susceptibility have the potential

to impact efficacy of antibiotics (6,45,46,63). When resistant bacteria spread to the community,

resistance creates comprehensive infection control issues, increasing morbidity for non-hospitalized

patients of all ages (63).

The estimated number of cases of uncomplicated cystitis per year, caused by E. coli alone, is 130–

175 million globally and 2-300.000 in Denmark alone (N. Frimodt-Møller, personal

communication) (64). Consequently, infections caused by E.coli, susceptible and resistant,

collectively result in considerable morbidity as well as direct and indirect financial costs seen as

increased health-care expenses, antibiotic treatment and loss of productivity (14,22,25,64).

Furthermore, UTI patients experience morbidity and impaired quality of life with an estimated 20-

40% of women having at least one UTI during their lifetime (22,64,65). It is difficult to determine

the precise incidence of UTI, but by using self-reported medical history the annual incidence in

USA was 13% among women and 3% among men (3).

Resistance in E.coli, besides β-lactam resistance, includes sulphonamides, trimethoprim and

ciprofloxacin (22). In 2008, UPEC isolates from five countries, were commonly resistant to

ampicillin (28%), sulfonamides (25%), trimethoprim (17%) and nalidixic acid (10%), with an

significant increase in resistance to nalidixic acid and trimethoprim from 2000 to 2009 (4). A total

of 60%, only, of the UPEC isolates were found to be fully susceptible (4). The antibiotic resistance

continued to increase throughout Europe, with 41% being fully susceptible in 2012, only. Especially

the current increase in resistance to extended-spectrum cephalosporins (mean = 12%) and

aminoglycosides (mean = 10%) in combination with increased resistance to at least three antibiotic

classes, are worrisome (41). The increased resistance is likewise worrying in Denmark. In 2012, the

resistance in E.coli isolated from urine (primary health care) were 40% for ampicillin with 33% for

sulphonamide and 10% were resistant to ciprofloxacin and 6% to mecillinam (61).

The continual increase in resistant E.coli has added to the enormous economic and human costs of

infections with 400.000 infections caused by MDR bacteria in Europe in 2007 (66). The economic

costs associated with these infections, counted as extra hospital costs and productivity losses

exceeds €1.5 billion in Europe and $20 billion per year in the United States (66).

Copenhagen 2014

19

2.3.1. Clinical Consequences of Resistance

There is an on-going discussion on the methods by which outcome of infections should be

investigated and uncertainty of true influence by resistant pathogens do exists (67). However,

predictors of mortality in patients with infections due to MDR Gram-negative bacteria have

previously been defined as infection severity, underlying diseases, inappropriate empiric treatment,

age and multidrug resistance (67,68). As MDR strains, including ESBL-producing strains, often are

resistant to most first line antibiotics, patients infected with these are more likely to receive

inappropriate empirical therapy why morbidity and mortality rate is generally higher (67,69–72). As

such patients with ESBL infections are more likely to suffer prolonged hospital stay and infections

are associated with higher use of broad-spectrum antibiotics (18,72–79). The increased mortality is,

however, significantly reduced if correct definitive therapy is given according to susceptibility

patterns and precise nonmedical interventions are performed (68). This, of course, makes

identification of patients at risk and carriers of resistant strains of great importance.

2.4. The Gastrointestinal Microflora

The human intestine is populated by thousands of bacterial species with the vast majority (99%)

being strict anaerobes and less than 1% being facultative aerobic and aero-tolerant (20,80). The

anaerobic Gram-negative Bacteriodetes and Gram-positive Firmicutes constitute 80–90% of the gut

bacteria (81).

The gastrointestinal microflora in the human intestine performs well recognized functions like

digestion of otherwise indigestible carbohydrates and the prevention of colonization of pathogenic

strains referred to as colonization resistance (20). The indigenous intestinal flora, therefore, acts as a

barrier against incoming pathogens and overgrowth of opportunistic microorganisms already

present in the gut. However, alterations in the microbiota can allow for colonization, with possible

subsequent infection, and antibiotic treatment is known to disturb the ecological balance of the

indigenous microflora (11,43,82). In addition, antibiotic treatment can select for resistant bacteria,

which could then disseminate in the intestine, increasing the possibility of infections (11). The level

of disturbances, caused by antibiotics, depends on route of administration, route of elimination and

spectrum of the agent (83). Resistance genes can be introduced into the gut via a travelling, non-

colonizing strain. The presence of an antibiotic, or recent exposure, will allow the resistant

microorganisms to establish themselves and proliferate (80). This overgrowth of resistant bacteria

encourages conjugation of plasmids between same and different species, increasing the resistance

PhD Dissertation

gene pool (37,43,83–88). Overgrowth and establishment of resistant strains is not seen, in nearly the

same degree, in individuals not under the influence of antibiotic (84,86,88,89). There is therefore a

likely association between last use of antibiotics and presence of resistance, but resistant E.coli have

been found in healthy individuals with no recent antibiotic exposure (84,86,88–90). Once present,

resistant pathogens may spread to other parts of the body or other individuals and the number of

colonized individuals exceeds the number of patients with identified infection (80). In clinical

settings, the gastrointestinal tracts can, as such, act as a source of infection or transmission and

previous exposure to antibiotic is a known risk factor for UTI with resistant bacteria

(37,38,43,82,83,86,87,91–93). In addition faecal carriage of ESBL-producing E.coli have been

detected in patients with ESBL infections, with risk of UTI increasing with larger quantity of the

ESBL-producing E.coli and abundance of such E.coli increasing with antibiotic exposure (42,93).

2.4.1. E.coli and Colonization

Virulence factors conventionally determining ExPEC are also found in commensal E.coli indicating

that VFs causing extraintestinal disease are also important for the intestinal colonization (10,22).

Usually the faecal flora of healthy humans is inhabited by one to five E.coli clones, with E.coli as

the dominating facultative anaerobic species and one clone habitually being dominant (11,20,89). It

has been found that E.coli belonging to classical ExPEC phylogroups B2 and D are often among the

dominating strains. Thus, there is a link between the presence of certain virulence genes,

colonization and pathogenicity with commensal E.coli often resembling ExPEC (11,20). Infections

due to ExPEC isolates, including UTI, are most often caused by E.coli already present in the

patient’s own intestinal flora (11,26,62). The human gut is now considered to be the primary

reservoir for uropathogenic E.coli (11,22,94).

2.5. Reservoirs of ESBL-producing ExPEC

Larger reservoirs have only been detected at local levels, like high rates of carriage in nursing-home

residents, with limited knowledge of true external reservoirs (52,62). Community outbreaks caused by

pathogenic E.coli have been described for ExPEC and a variety of contaminated foods have previously

been linked to outbreaks of IPEC (22,95,96). During a recent outbreak in Germany, of Shiga toxin

producing IPEC, also expressing ESBL, sprouts were identified as the likely primary source with

secondary person-to-person transmission and long-term shedding after illness (97). Transmission of E.coli

Copenhagen 2014

21

through sexual activity have been reported and infecting UPEC have been found in the faecal flora of

family members (14,22,98).

2.5.1. The Human Gut as Reservoir

Among 86 Danish army recruits, 3.5% had ESBL-producing E.coli in their faecal flora in 2011 (99). In

Sweden 2.4% of healthy individuals were colonized with ESBL-producing Enterobacteriaceae and

Jørgensen et al. found the overall ESBL carrier rate among patients in Norway, submitting faecal samples

to a hospital laboratory, to be 15.3% (100). Similar low to modest carrier rates have been seen in Japan

and Switzerland (6%), Tunisia (7%), UK (11.3%) and Saudi Arabia (13.1%), but with extremely high

carrier rates in Cairo (63.3%) and rural areas of Thailand (65.7%) and with varying findings in China (7-

50%) (56,101–104). In 2011, in the Paris area, a 10-fold increase in ESBL-carriage in healthy individuals

was detected over a 5-year period (from 0.6% to 6%) (104).

History of international travelling has been found to propose a risk for colonization of ESBL-producing

E.coli (52). Studies in the Scandinavian countries have identified considerably higher rates of carriage of

ESBL-producing Enterobacteriaceae among patients with a history of recent travelling to Africa north of

the equator, Asia and in particular the Indian subcontinent (100,101,105,106). This applied to both

hospitalized and non-hospitalized patients (100,101,105,106). These Scandinavian studies corresponded

well with a report from Canada showing that travellers were five times more likely to be carriers of ESBL

than non-travellers (107). Östholm-Balkhed et al. found that travellers >65 years had a higher risk of

ESBL acquisition than travellers aged 18-35 years and Tham et al. identified 24% of travellers as long

time-carriers (3-8 months) (101,108).

Comprehensive intestinal colonization of resistant E.coli has been seen in some nursing homes suggesting

long-term care facilities as another reservoirs for resistant bacteria (52). Rooney et al. found that 40% of

nursing home residents in Northern Ireland were colonized with ESBL-producing E.coli (109). Here

carriers of ESBL had higher levels of exposure to antibiotics, high rates of hospital admission and

frequent use of urinary catheter (109).

2.5.2. Animals, Water and Food

Finally, food, water and animal reservoirs have been suggested, with a genuine zoonotic origin of UPEC

strains previously suspected (65,110). ESBL-producing E.coli have been detected in production animals

in several studies, with the presence of known human ExPEC lineages (52,98). Pathogenic E.coli from

animals (house-hold pets and production animals) and humans do, to some extent, share a common

PhD Dissertation

genetic background based on MLST, yet with some species specific VFs (62,94,111,112). E.coli isolates

from meat have, however, been shown able to cause disease in a mouse model of ascending UTI (113).

In two Danish studies, ESBL-producing E.coli were found with increased frequency from pigs-farms with

a high consumption of extended-spectrum cephalosporin (98). Similarities between E.coli isolates and

ESBL types have additionally indicated transfer among animals, meats and humans and a voluntary ban

on use of cephalosporin in pig production, furthermore led to a decrease in ESBL-producing E.coli among

slaughter pigs in Denmark (62,98). The Netherlands have a higher level of antibiotic use in the poultry

industry, than any other European country, and the prevalence of ESBL-producing E.coli in the

gastrointestinal tract of healthy food-producing poultry were 15% in 2008 (114). Of retail chicken meat

94% of samples contained ESBL-producing E.coli isolates, of which 39% were ESBL genotypes also

found among humans (114). In Barcelona, pathogenic E.coli lineages, often associated with resistance,

were detected in sewage and river water, with high levels of clonal similarities to human isolates (110).

Lastly, ESBL-producing E. coli were detected in healthy pigs, cattle, and poultry as well as in wildlife in

China. The latter has been suspected to be due to spreading of manure containing ESBL-producing E. coli

(115).

2.6. Experimental Animal Models for Investigation of Colonization

The compositions of the intestinal microflora of laboratory mice have been examined by Krych et al. to

evaluate the similarities to the flora of the human gut (116). There were some quantitative differences, but

mouse and human faeces, to a large extent, had similar representatives of phyla and a substantial segment

of common genera. Hence, lab mice and human share the same basic bacterial species in the gut (116).

Animal models have been used extensively to study the pharmacokinetic and -dynamic parameters of

antibiotics as well as acute and chronic infections caused by bacterial pathogens with different resistance

patterns (117,118). Furthermore, since the intestinal flora of mice and men are comparable, mice are often

the mammal used to investigate the relationship between intestinal microbiome, health and disease (116).

Colonization studies of the mouse gut by bacteria have been used in a range of mouse models, with many

similarities between methods (119,120). In general, both at SSI and with foreign research groups,

colonization has been described in mice given antibiotics prior to inoculation with bacteria of interest and

conjugation between two colonizing strains have likewise been tested (37,119,121,122).

The effect of subcutaneous treatment on the indigenous intestinal microflora as well as the effect on

colonisation by a KPC-producing Klebsiella strain (KPC-Kp strain) was tested in a mouse study in 2011

by Perez et al (123). Their findings suggested that antibiotics with effect on the anaerobic intestinal flora

Copenhagen 2014

23

promoted the establishment of the KPC-Kp strain – provided the antibiotic had no effect on the strain of

interest. Hence, antibiotics with limited effect on the anaerobic flora might be less likely to promote the

colonisation of MDR Gram-negative bacteria (123). Perez et al also found that antimicrobial impact on

the total anaerobic population is proportional to the measured impact on the Bacteroides population (123).

Colonization was successfully investigated by sampling and measurement of stool, since E.coli is present

in both mucus content and intestinal content (123,124). The hypothesis gained from previous mice studies

has, however, never been confirmed or tested on more common resistant Gram-negative strains. It has

previously been shown, that a human-simulated dosing of antibiotics gives the most constant levels of

drug in mice – who have a higher elimination rate (125). Yet, a single daily subcutaneous dose can

produce similar levels of drugs in mice faeces, to those seen in humans (123). This dosing frequency of

antibiotic does, nonetheless, not precisely mimic the exposures seen in patients (123).

Mouse intestinal colonization of Gram-negative bacteria has therefore successfully been determined in

several studies, many using elimination of resident facultative bacteria prior to inoculation

(119,121,123,124,126).

2.7. Composition of ExPEC populations

With the introduction of MLST, many E.coli isolates, causing outbreaks and previously

characterized by serotypes, were re-characterized by sequence type (ST) (7). Consequently, reports

of the global spread of ExPEC belonging to a few STs followed, with reports of worldwide

occurrence of ST10, ST12, ST73, ST95, ST127, ST131 and ST393 (7). The spread of E.coli-

lineages associated with certain resistant patterns and multidrug-resistant STs were also,

retrospectively, determined.

Interestingly, the initial descriptions of ESBL-producing E.coli gave indications of a non-clonal

distribution of isolates with great diversity of MLST types and ESBL types (52,127,128). However,

reports of rapid worldwide dissemination of the CTX-M β-lactamases, and especially CTX-M-15,

were followed by descriptions of closely related groups of E.coli spreading throughout the world

and this global spread was not confined to ESBL-producing E.coli. ST69 has since 1999 appeared

worldwide as a resistant E.coli related primarily to trimethoprim-sulphamethoxazole resistance, but

not regularly as ESBL-producing strains (7,129). Conversely, ST393 emerged as a MDR ExPEC in

1986-87 in London and has since then spread to many parts of world, gaining resistance and has

now been associated with CTX-M-type ESBLs, without losing virulence (7,130). Likewise, ST131

was identified in the mid-2000s with an unknown origin and has been associated closely with

PhD Dissertation

ESBL-production and quinolone resistance (7,131,132). Today a substantial proportion of MDR

ExPEC, worldwide, belong to ST131 producing CTX-M-type ESBLs and is described as a

pandemic ST-lineage with very low incidence amongst susceptible isolates (7,52,131,132). Other

very common ExPEC lineages like ST73 and ST95 were discovered among isolates from the 1917

and 1941 and has continued as common causes of UTI, but rarely as carriers of ESBLs and rarely

multidrug resistant (52,133–135). Consequently, certain ExPEC STs seem to be associated with

resistance, ESBL and virulence genes, enabling them to cause major outbreaks at different locations

simultaneously (54,134,136). Thus, it seems that there is a distinction between resistant isolates and

fully susceptible E.coli (135,137–139). The prevalence of the different ST-lineages varies

depending on country and continent, but ST131 in particular has been reported as widely

disseminated in North America, most parts of Europe, in Asia, the Middle East and Australia

(52,140). In Norway 20% of CTX-M-producing E.coli belonged to ST131 and in Denmark, ST131

has been shown to dominate among the ESBL-producing E.coli constituting 35-45% (52,57,132). In

Canada ST131 comprised <3 % of susceptible isolates but presence of ST131 amongst non-ESBL

has never been studied in Denmark (52,57,132). As a common MDR E.coli-lineage ST131 has been

presented both as a lineages with a high virulence score and as a lineage which harboured less

virulence factors than other uropathogenic E.coli (115,135,141). Nonetheless, ST131 has proven to

have a higher metabolic potential and carry similar virulence genes regardless of location

(7,8,115,136,141–144). It might be the explicit combination of virulence and resistance features

which gives MDR ST-lineages a beneficial competitive advantage and be the possible reason for the

clonal-like global expansion (141,142,145). Thus, studies on collection of E.coli isolates has shown

that a limited set of lineages account for a large proportion of multidrug-resistant strains (7).

Sequence types can often be sub-dived by serotyping, PFGE, fimH sequencing and identification of

virotypes (133,136,146). Thus, most ST131 isolates belong to serotype O25, but a minor part of the

ST131 populations have recently been identified as O16 (132). The subdivision of STs has proven

to be a weakness of the MLST method, since most of the MLST-based lineages consist of highly

heterogeneous groups of strains (133). The spread of resistant E.coli sequence types has been

extensively investigated (7). However, only a few studies have examined non-ESBL and susceptible

E.coli (9,138,147).

Copenhagen 2014

25

2.8. Risk factors for infection with ESBL-producing E.coli

The epidemiology concerning infections due to ESBL-producing Enterobacteriaceae has for some time

been of great concern and as a result has been extensively investigated. The study designs used to

investigate these epidemiological characteristics of infections by resistant Gram-negative bacteria have

often been retrospective case-control studies (148–150).

In the early 2000s infections due to ESBL-producing E.coli were mainly investigated as infections

associated to hospital settings (15,72). As CTX-M became the dominating ESBL genotype, with a

preference for E.coli, these pathogens appeared increasingly from both healthcare settings and the

community, posing a threat to management of empirical treatment. Consequently, research focus

shifted towards E.coli and to some extent the community (15,72,74,76,149,151–155). Doi et al.

found that 36.8% of patients colonized or infected with ESBL-producing E.coli were community-

associated, of which 91.3% produced a CTX-M-type ESBL and 54.2% were caused by ST131.

Cases of UTI were composed of 67.4% community-onset infections caused by ESBL-producing

E.coli and UTI was identified as the most frequent source of bloodstream infections caused by

ESBL-producing E.coli (15,142).

2.8.1. Considerations on study design

The case–control study design is one of the most frequently used methods for identifying risk

factors related to antibiotic resistant pathogens. Yet, the case-case–control study design can more

accurately identify these risk factors than the classic single case–control study design (156,157).

When patients with resistant bacteria are compared to patients with susceptible bacteria, the design

makes it impossible to identify which risk factors are specific for resistant infections and which are

risk factors for infection in general. Control-patients with susceptible strains are not likely to be true

representative of the population for resistant cases and constitutes a risk for selection bias,

especially on the effect of antibiotics (156–161). Patients with the susceptible bacteria could,

however, represent a part of the true source population but are then not included in the control

group. Furthermore, using two different control-groups makes the results less comparable, even if

control-patients are more representative. The control-group should be representative of the source

population, among which all cases are found, and act as controls for all cases. As the source

population is the population from which all case-patients were selected, members of the control-

group with the target organism would have been classified in a case-group (156,157). When the

same control-patients are used in all analyses, comparison of the resulting risk models is easy and

PhD Dissertation

the comparison can identify risk factors specifically associated with one or the other case-group

(156–161).

Overall the case-case-control study design is best suited for investigations relating to infection

control and public health perspectives, when the focus is to detect patients who are at risk for

harbouring particular resistant organisms, and to further implement interventions to limit the spread

of these organisms (156). If the emphasis, on the other hand, is on estimating the likelihood that a

resistant pathogen is involved and hereby identify the most effective empiric antimicrobial

treatment regimen, the control-group must include patients who had clinical cultures performed

(156). Now the control-group does not represent the true source population, since all involved

patients have sought medical attention, but makes the collection of control patients easier (156,158).

The basis of the case-case-control design is the uses of separate case–control analyses within a

single study, performed as parallel t-tests representing the risk factor identification followed by

parallel multivariate logistic analysis (156,161,162).

2.8.2. Case-Control Studies

Many case-control studies of risk factors, included either admitted patients or patients from hospital

settings, and infections were retrospectively classified as either community-onset infections (COI)

or healthcare-associated infections (HCAI), by use of criteria similar to those proposed by Friedman

et al. (15,17,18,73,74,151,155,163–165). As one of the main foci of investigation, exposure to

antibiotics were looked at preceding infections in various periods as within the previous year

(18,73,155,166), 2-3 months (70,149,167–172) and 1 month prior to infection (15,173), with

hospitalization generally studied in similar intervals (17,70,73–75,77,149–

151,153,155,165,166,172,174,175).

Regularly identified risk factors were male sex, age >65 years, prior ESBL-producing E.coli

colonization, previous UTI, urinary catheterization, previous receipt of antibiotics and in particular

previous exposure to extended-spectrum cephalosporins and fluoroquinolones, nursing home

residence and hospitalization within the preceding 12 months (15,17–

19,69,70,155,165,172,173,176,177). Impact of comorbidities and underlying diagnoses´ as risk

factors varied between studies (33,69,73–75,148–153,167,169,170,178). Thus, important

epidemiological factors habitually involved antibiotic selective pressure, rather than individual

comorbidities, in the emergence of ESBL-producing isolates (153). Some investigations also

recognized patients with COI ESBL-producing E.coli who had no preceding health-care exposure

Copenhagen 2014

27

and ESBL-producing E.coli emerged as pathogens in children (<15 years) (15,18,19,155,172,176).

Finally, urinary catheterization and exposure to β-lactamases-inhibitors, cephalosporins, co-

trimoxazole and aminoglycosides were factors associated with an ESBL-infection in patients

previously colonized with an ESBL-producing strain (179).

The dominating β-lactamases identified were of the CTX-M type, primarily from CTX-M group 1,

while TEM and SHV types became less frequent (73,150,151,154,167). Major limitations related to

many case-control studies were the lack of integrated databases for use of medications, the inability

to control the use of non-prescribed antibiotics and the fact that many studies were single case-

studies, where ESBL infections were compared to non-ESBL (74–76,148,149,151).

2.8.3. Double-Case-Control Studies of Epidemiological Factors

A few studies have investigated epidemiological features associated with COI, by use of a double-

case or -control design (19,73,170). All were located at hospital settings including hospitalized

patients and only two studies included an uninfected control group (19,73,170).

In Thailand in 2007, Apisarnthanarak et al. performed a double-case control study including all

infections caused by ESBL-producing E.coli (73). They found previous ESBL colonization and

recent exposure to antibiotics, especially to 3rd generation cephalosporins and fluoroquinolones to

be independent risk factors for any COI with ESBL-producing E coli (73). Risk factors for non-

ESBL infections were stroke and diarrhoea while diabetes was found as a common risk factor for

both groups (73). Interestingly, they demonstrated an association between infections with ESBL-

producing E.coli and increased mortality, longer hospitalization and higher costs (73).

Nicolas-Chanoine et al. studied if patient’s origin and lifestyle was associated with infections with

CTX-M-producing E.coli, as a case-control-control study in Paris in 2012 (19). They compared

ESBL patients to patients with non-ESBL E.coli and uninfected individuals and found that birth

outside of Europe, chronic infections and antibiotic treatment after hospitalization, but before

inclusion, were risk factors for ESBL infections. Furthermore, functionally dependent and collective

housing before hospitalisation was associated with CTX-M-producing E.coli.

2.8.4. Case-Control Study in Low Prevalence Countries

Two research groups have studied the epidemiological factors in low prevalence countries (Søraas

et al., Norway 2013 and Rogers et al., Australia/New Zealand 2014) as single-case control studies

PhD Dissertation

including hospitalized and non-hospitalized patients (17,18). Søraas et al. looked at healthcare

contact and prescriptions of antibiotics and further included travel, diet and recreational swimming

in the analysis (18). Here they found patients with ESBL infections to be of younger age and

identified travel to the Middle East or Africa, recent use of fluoroquinolones and β-lactams (except

mecillinam), diabetes mellitus and recreational freshwater swimming as risk factors with decreased

risk associated with increasing number of fish meals per week (18). Rogers et al. looked at risk

factors for expanded-spectrum-cephalosporin-resistant E.coli and found birth on the Indian

subcontinent, previous urinary tract infection, travel, prior exposure to trimethoprim±

sulfamethoxazole and ±expanded-spectrum cephalosporins and health care exposure in the previous

6 months to be associated with ESBL infections in a low prevalent country.

The European Society for Microbiology and Infectious Diseases (ESCMID) treatment

guidelines for UTI, have changed in recent years, to meet the challenges of empirical treatment of

E.coli. As we have seen a constant development and rapid spread of antibiotic resistance,

continuous local surveillance for resistance combined with identification of patients at risk are

essential to avoid inadequate empirical therapy and adverse outcomes. In order to create

interventions to reduce the prevalence of ESBLs, we need to understand and identify the

dissemination of ESBL-producing E.coli and understand the impact of antibiotic selection pressure

(115).

Copenhagen 2014

29

3. Bacteriological Characterization of E.coli

The typing methods used in these studies are briefly introduced below.

3.1. General Considerations on Typing Methods

Bacterial typing is used to distinguish genetic variation among isolates of the same species. Typing

of bacteria has become a valuable tool for detecting and controlling outbreaks as well as identifying

spread of resistant strains (180). As for now, a variety of different typing methods exists, each with

advantages and shortcomings when it comes to discriminatory power and price. Furthermore, there

is a constant development of new methods in addition to improvements of known techniques and

such advances have to balance between creating new and maintaining existing nomenclature (181).

Maintaining elements of an existing nomenclature, that may have been well understood and

accepted, is essential for general acceptance and use (181).

E.coli can be typed with several different methods, such as phylogrouping, sequence-typing, multi

locus variable number of tandem repeat analysis (MLVA), pulsed-field gel electrophoresis (PFGE),

serotyping, random amplified polymorphic DNA (RAPD) etc., not including subdivision based on

virulence and pathogenic capacities. Such methods have proved crucial in investigation of the

epidemiology of pathogenic E.coli and are of great importance in disease control (181,182).

Additionally, each technique can often subdivide across each other’s boundaries. Therefore,

choosing a typing method requires consideration on economic costs, skills needed by the institution

and analytical properties in regards to what questions needs to be answered.

3.2. Phylogrouping by PCR

The subdivision of E.coli into the phylogroups A, B1, B2 and D by polymerase chain reaction

(PCR), has been a fast and inexpensive manner to divide E.coli into the four major phylogroups

(28,29,183). The first described PCR protocol was a triplex PCR developed by Clermont and

colleagues (28). A method which was adjusted by Gordon and colleagues and further revised by

Clermont and colleagues (29,183). The Clermont method was originally designed to assign E. coli

strains to four phylogroups, why the method clearly not will assign strains not belonging to A, B1,

B2 or D. To which extend strains are miss-assigned by this method will rest on the composition of

the collections evaluated, but an estimated 79% of isolates will be correctly assigned (29). In this

study we used the triplex PCR adjusted by Gordon et al. It is a rapid procedure based on primary

PhD Dissertation

PCR detection of three marker genes (i) chuA (ii) yjaA and (iii) TSPE4.C2, followed by potential

subsequent detection of a fourth gene ibeA. The presence or absence of genes (i)-(iii) generates a

dichotomous decision tree used to determine the phylogenetic group of an E.coli isolate (28,183).

The specific findings of chuA+, yjaA-, TSPE4.C2+ results in the need for secondary screening for

the gene ibeA, to correctly discriminate between phylogroup B2 and D. The presence of ibeA meant

assigning the isolate to group B2. Strains failing to yield any PCR products of the three primary

genes remain unassigned as non-type able (183).

E.coli has traditionally been divided into five main phylogroups (A, B1, B2, D and E). Recently,

other minor phylogroups have been described and now E.coli is divided into a total of seven

different E.coli sensu stricto phylogroups (A, B1, B2, C, D, E and F) with an eighth phylogroup

being the Escherichia cryptic clade Ι. The world of E.coli phylogroups has therefore become

increasingly large, but just 13 % of human faecal E.coli isolates belong to the newly identified

phylogroups C, E, F and clade I, why we here will focus on the four biggest phylogenetic lineages

(A, B1, B2 and D).

3.3. Multi-Locus Sequence Typing

Multi-locus sequence typing (MLST) is a typing method which have proved portable and easily

comparable between laboratories (180,181,184,185). It was introduced in 1998 and a range of

MLST schemes for different organisms has been established and made readily accessible in online

databases. In this study the Achtman scheme for E.coli characterisation was used, and this method

is briefly described. MLST of E.coli is based on the identification of allelic profiles of seven

different housekeeping genes coding for fundamental metabolic functions: adk, fumC, gyrB, icd,

mdh, purA and recA (181,186). It meets the requirement for multiple loci characterisation of

alleles that are distinguishable and easily analysed (180,181,184,185). MLST is a less strenuous

technique than whole genome sequencing, but requires PCR amplification and subsequent

sequencing of the seven genes. It holds the clear advantages of an international on-line

nomenclature (180,181). Each allele is sequenced and by accessing the online MLST database the

determined nucleotide sequence is assigned a unique number

(http://mlst.warwick.ac.uk/mlst/dbs/Ecoli). This produces high levels of discrimination between

isolates. As each allele is given a number, the combination of numbers is used in an MLST scheme

to give each isolates a unique sequence type (ST) (180,181,186). As for whole genome sequencing,

the success of MLST lies on decreased cost for nucleotide sequence determination. MLST is today

Copenhagen 2014

31

the gold standard of characterisation and can be used to describe evolutionary and phylogenetic

relationships (181).

3.4. Multi-Locus Variable Number of Tandem Repeats Analysis

The multi-locus variable number of tandem repeat analysis (MLVA) is a characterisation method

originally based on PCR amplification and size determination of eight variable number of tandem

repeats loci (VNTR), meaning that MLVA exploits naturally occurring differences in DNA regions

with tandem repeats. These regions are found throughout most bacteria as well as mammals, but

only identical cells will have the same number of repeats in each of the targeted regions.

Consequently, identical cells will generate PCR products of matching size (187,188). Each of the

VNTR can be assigned a number depending on size and one isolate will consequently be assigned

an eight-digit code when using the original MLVA method (187,188). The method relies, however,

on accurate size determination of each PCR product.

In this study we used a modified MLVA method described as an abbreviated MLVA (a-MLVA).

Here, only six of the eight original VNTR loci were used and reactions done as singleplex PCR

amplification with unlabelled primers. We used a-MLVA since this method uses loci with repeat

sizes between 12bp and 95bp, meaning that the size of fragments could easily be determined by the

use of an automated capillary electrophoresis system (QIAxcel, Qiagen) and a high-resolution

cartridge (57).

3.5. Serotyping

The serological classification of E.coli isolates, as proposed by Kauffman in 1944, is based on the O

(somatic) and the H (flagellar) surface antigen profiles (95,189,190). Since O and H antigens are

stable this method for strain characterization is considered to be very reliable (189). Serotyping has

therefore been an important technique for differentiating important pathogenic E.coli and classically

used to describe IPEC pathotypes (190). The characterization is based on bacterial agglutination

tests with antigen-antiserum. An E. coli strain which is inagglutinable by O antiserum, but

agglutinable when heated is said to have a K antigen (95). The specific combination of O and H

antigen are characterized as a serotype, while the determination of O antigens only are termed

serogroups (190). IPEC seems to belong to a limited number of O:H serotypes, which to some

extend applies to ExPEC, were virulent E.coli belong to a limited number of O serogroups (190).

More than 175 different O antigens, each defining a serogroup, and more than 50 H antigens are

PhD Dissertation

recognized (95). It is not the serological characterization which confers virulence, but rather the

separate presence of virulence genes (95). For this thesis O serogrouping was performed only, and

executed by GlycoVaxyn as part of an unpublished commercial project.

Copenhagen 2014

33

4. Material and Methods

Methods are described in each individual manuscript. This following paragraph is a short

description of materials and methods not specifically described in these (Figure 2).

Figure 2. Overview of used materials and methods

4.1. General Protocols

Work performed in the laboratory was done according to current guidelines from SSI, DCM as well

as stated by manufacturers of material and products. Furthermore, all media and products were

stored according to manufacturer’s recommendations. Guidelines were followed in agreement with

previously published papers and current literature.

4.1.1. Research Ethical Approvals

When necessary, needed approvals were obtained from Government institutions. Thus all animal

experiments were initially approved by the Danish Centre for Animal Welfare (Current permit:

2013-15-2934-00777 C3) and carried out at licensed animal facilities at SSI. Additionally,

collection of patient samples and patient data was approved by the Danish Health and Medicines

Authority as well as Statistics Denmark and The Regional Committee of Danish Data Protection

Objective 1

Strain: CTX-M-15 producing

E.coli ST131

Selective Agar

Disk disffussion, Etest, Vitek 2,

MALDI-TOF

Mouse Intestinal Colonization

Model

Detection and quantification of

bacteria

MLST

Objective 2

Three case-groups (98vs.174vs.

177).One control group(200)

Epidemiological data (Statistics

Denmark and hospital records)

Multivariate logistic regression

analysis

Disk disffussion, Etest, Vitek 2,

MALDI-TOF

Phylogrouping

ESBL Genotyping

Objective 3

Three case-groups (98 vs. 94 vs.

94).

Serogrouping

(94 vs. 49 vs. 51)

Abbreviated MLVA

MLST

Phylogrouping

ESBL

PhD Dissertation

Agency. The Regional Committee of Health Research Ethics Committee declared notification of

patients unnecessary.

4.1.2. CPR Registry

In 1968 the Danish Civil Registration System (CRS) was established. Now every person living in

Denmark is registered with a unique personal identification number called the CPR-number. The 10

digit CPR-number includes individual information on name, gender, date and place of birth, identity

of parents as well as continuously updated information on address, marriage and vital status. The

CPR is used for identification when in contact with governmental institutions, hospitals, general

practitioner and for prescribing medicine. The CRS is connected to all Danish national registers as

well as bio-banks and this information is available for research purposes (191,192).

4.1.3. Statistics Denmark

Statistics Denmark (DST) is a central authority that collects and publishes statistics on the Danish

society (http://www.dst.dk/en/). They are linked to other Danish national registers such as the

Danish National Prescription Registry (DNPR) and National Patient Registry (LPR)

(http://www.ssi.dk/Sundhedsdataogit/Registre). Both DNPR and LPR are linked to CRS. DNPR is

owned by the Danish Medicines Agency, currently maintained by SSI, and monitors all information

on dispensed prescriptions and drug user. Thus, DNPR collects CPR-data and details on prescribed

drug (ATC-code, dose, dispensing date etc.). However, DNPR does not register over-the-counter

drugs on an individual level or drugs prescribed in hospital settings. The LPR is the National Patient

Registry and monitors all hospital contacts (admissions, outpatient contacts and visits to emergency

room), the departments involved, diagnoses given and surgical procedures. Data on medicine

administered during hospital admission is not available, but data on prescriptions dispensed to

residents at long-term care institutions is. Additionally, diagnoses from general practices are not

available. Hence, DST governs all prescribed medicine, hospital contacts and diagnoses for each

individual with a CPR-number. This extensive data is available for research purposes. The

availability is granted as an encrypted, extern and time-limited access to databases and only for

publications on population- and not individual-level. The CRS linkage with nationwide individual-

level data sources renders DST as a very powerful epidemiological tool (192,193).

Copenhagen 2014

35

4.2. Mouse Intestinal Colonisation Model (Manuscript I)

The mouse intestinal colonisation model and the results gained from this have been thoroughly

described in manuscript I.

We based our experimental mouse model on similar animal models previously used at SSI

(paragraph 2.6.1.) and modified the model through pilot studies. Here follows a short description of

the preliminary studies performed to test and verify its use.

4.2.1. Selection of E.coli Isolates and Preliminary Mice Experiments

Among a collection of Danish E.coli blood isolates at SSI, collected as part of another study (194),

four ESBL-producing E.coli isolates were chosen as possible colonising pathogens. These isolates

were chosen based on their ESBL-genotype, their sequence type 131 and the fact that they were

susceptible to gentamicin (later used in selective plates). The four isolates ((A)64-Ec-09: B2,

ST131, CTX-M-15, AmpC hyper production (B) 65-Ec-09: B2, ST131, CTX-M-15 (C) 69-Ec-09:

B2, ST131, CTX-M-15 and (D) 71-Ec-09: B2, ST131, CTX-M-15) were subjected to an initial

mouse experiment, in the attempt to choose an E.coli isolate appropriate for a reproducible

experimental study of colonisation. The basics of each experiment were similar and described in

detail for the mouse intestinal colonisation model. Based on the mouse experiment, we chose our

colonising pathogen to be 65-Ec-09. Similarly, we tested if resistant E.coli would colonise the gut

without antibiotic treatment and likewise examined the effect of cefotaxime on the indigenous

E.coli population. Furthermore, for the selected strain, we repeated the initial mouse study with

cefotaxime and performed a study with cefuroxime, clindamycin and ciprofloxacin. In these studies,

the colonising ability was established as well as the appropriate interval of faeces collection.

Finally, we did one last pilot study with 65-Ec-09, implementing the faeces collection arrangement

and selection of resistant E.coli and Gram-negative anaerobes. This final pilot was performed with

the antibiotics cefotaxime, cefuroxime, ciprofloxacin, dicloxacillin and clindamycin. Selective

plates for Gram-positive bacteria were not introduced until the second run of the experimental

study.

PhD Dissertation

4.3. Study Populations (Manuscript II and III)

4.3.1. Sample Size Determination

Prior to the initiation of the case-control study we evaluated sample sizes of previous case-control

studies. We used these sample sizes to estimate the needed number of cases in our investigation in

concordance with expected monthly prevalence of community-onset ESBL UTI and an online

calculation of groups sizes, when p=0.05% and 80% power.

A statistician at Hvidovre Hospital calculated the number of isolates needed in the characterization

of susceptible populations.

4.3.2. Strain Collections

The triple-case-control study was based on the prospective collection and examination of urine

samples submitted from general practices to DCM from 1st. of October 2011 to 1st. of July 2012,

combined with the retrospective exploration of patient data. This was done as a non-intervention

study with independent patient-treatment being handled by health-care professionals. All received

samples were handled by the laboratory technicians according to general guidelines and patients

were treated accordingly. An initial susceptibility test was always performed on E.coli from urine

samples and included screening for ESBL-production by the use of cefpodoxime. E.coli with low

susceptibility for cefpodoxime was subjected to a MAST-test and phenotype confirmed.

Composition of case groups are described in manuscript II.

GlycoVaxyn AG determined serogroups for 94 of ESBL isolates, 49 resistant and 51 susceptible

isolates (Manuscript III).

For typing of the E.coli populations (Manuscript III) all 98 ESBL-producing E.coli isolates, 94

isolates from the resistant group and 94 isolates from the susceptible group were included. Due to

cost restraints, we were not able to include all isolates from each of the case groups. A statistician at

Hvidovre Hospital helped us calculate the number of isolates needed to identify the majority of STs

expected to be present in the two non-ESBL populations. Primarily, all isolates with a known

serogroup were selected. Secondly, isolates from patients with no immediate health-care association

(this was before data from DST were obtained) were chosen randomly.

Copenhagen 2014

37

4.4. Epidemiology of collected isolates from cases and controls

Patient data were collected from medical records, when necessary, or obtained from Statistics

Denmark (DST). Data on admissions, diagnoses and prescribed medications from general practices

were acquired from one year prior to, till 90 days after collection of urine sample. Since data from

Statistics Denmark were analysed by external online access only, it was not possible for us to

subsequently add data to the DST database.

PhD Dissertation

5. Results

This chapter includes several tables of results and a brief stating of the main results obtained during

this PhD. It is divided in three sections: The first section includes results found in the selection

study of the mouse intestinal colonization model. The second and third sections are results obtained

during the case-control study and the following characterization of selected E.coli isolates.

5.1. Mouse Intestinal Colonization Model (Manuscript I)

This first section shows results found during the mice experiments presented as major findings in

Table 1 and 2 and in Figure 3, 4 and 5.

Objective I

To evaluate the ability of nine common antibiotics, to select for a CTX-M-15-producing E.coli

isolate belonging to sequence type (ST) 131 in vivo.

By use of the described mouse intestinal colonisation model (manuscript I) we evaluated the

selective abilities of cefotaxime, cefuroxime, dicloxacillin, clindamycin, penicillin, ampicillin,

meropenem, ciprofloxacin and mecillinam on a fluoroquinolone resistant, CTX-M-15-producing

E.coli isolate (65-Ec-09) belonging to the lineage B2-O25b-ST131 (resistance pattern found in

Table 1).

Table 1. Antibiotic susceptibility profile for selected antibiotics for E.coli isolate 65-Ec-09.

Antibiotic MIC mg/l Interpretation

Cefotaxime >64 R

Cefuroxime >64 R

Ampicillin >32 R

Ciprofloxacin >4 R

Gentamicin <1 S

Meropenem < 0.25 S

Mecillinam 8 S

Dicloxacillin >256 R

Clindamycin >256 R

Copenhagen 2014

39

We used 8 mice per antibiotic and CFU of E.coli ST131, Bacteroides and Gram-positive aerobic

bacteria in faecal samples were studied on day 1, 2, 4 and 8. We used the Bacteroides population as

an indicator for antibiotic impact on the Gram-negative anaerobic population. Prolonged

colonization was studied for three antibiotics with added faecal CFU´s studied on day 10 and 14

(cefotaxime, dicloxacillin and clindamycin).

We collected 0.5 grams of faeces from each cage and calculated results of faecal bacteriology as

LOG CFU/0.5 grams of faeces as the average CFU count on two agar plates. The mean CFU from a

total of four cages were then used for statistical calculations (two cages for the Gram positive

aerobic flora). For the graphic depiction we used means and SDs.

5.1.1. Effect of antibiotic selective pressure on the microflora

As seen in Figure 3 we found that cefotaxime, dicloxacillin and clindamycin promoted colonization

and overgrowth of 65-Ec-09 from day 2 through 8 (P < 0.01 for dicloxacillin and clindamycin, P <

0.05 for cefotaxime). Interestingly, both benzylpenicillin and cefuroxime induced overgrowth on

day 2 and 4 (P < 0.05) with dicloxacillin and clindamycin showing the overall highest selective

abilities (P < 0.01 for dicloxacillin on day 2 and 4 and P < 0.01 for clindamycin on all days). For all

antibiotics, there was a decline in the colonization of cefuroxime resistant E.coli after ended

treatment on day 3.

As seen in Figure 4 the remaining antibiotics (ampicillin, mecillinam, meropenem and

ciprofloxacin) showed limited selective abilities and promoted overgrowth of the cefuroxime

resistant E.coli on day 2 only (P > 0.05). Antibiotics impact on the microbiota represented by

Bacteroides and Gram-positive growth are found in shown in Figure 3 and 4. It seems that none of

the antibiotics made an impact on the Gram-positive flora, inhibiting or promoting (P > 0.05).

Essentially we found clindamycin, only, suppressed the Gram-negative anaerobic flora, completely

eliminating the Bacteroides during treatment (P<0.05). As treatment stopped, CFU counts for

Bacteroides increased and returned to counts equal to before treatment.

5.1.2. Effect of antibiotic selective pressure prolonged presence of 65-Ec-09

Data for CFU of 65-Ec-09 on day 1-14 are found in Figure 5. Here we saw that the initial high CFU

count was followed by a rapid decrease for all three antibiotics. For two of them (dicloxacillin and

cefotaxime) the CFU counts were below the detection limit on day 10. For clindamycin, however,

the CFU count was assessable until day 14, which was the last day of the study. Nonetheless, we

PhD Dissertation

saw no significant differences in faecal CFU of cefuroxime resistant E.coli among the three

antibiotics in the study of prolonged colonization (P>0.05).

5.1.3. Molecular tests of bacteria isolated from mice faeces

Results of the molecular tests performed on isolates from mice faeces are found in Table 2.

To correctly identify species found in mice faeces we performed MALDI-TOF on selected isolates.

Furthermore, to properly ensure that cefuroxime resistant E.coli cultured were identical to 65-Ec-09

we tested 17 resistant E.coli isolates found in faeces during treatment and four E.coli isolates from

day 1 (prior to inoculation) for the presence of a blaCTX-M group 1. Finally, one E.coli isolate from

day 1 and five cefotaxime resistant E.coli isolates were characterized by MLST. For these tests we

included isolates from both study runs.

All 21 isolates suspected to be E.coli were confirmed as such by MALDI-TOF and all resistant

E.coli isolates contained a blaCTX-M group 1. None of the E.coli isolates collected prior to

inoculation contained such a gene. All tested resistant E.coli were characterized as ST131 while the

non-CTX-M-producing E.coli belonged to ST602. Table 2 shows E.coli isolates characterized by

MLST as well as a representative for Gram-negative and –positive isolates, which were all

identified as belonging to identical species.

Table 2. Overview of molecular testing of bacterial isolates from mice faeces.

Antibiotic Day of experiment MALDI-TOF blaCTX-M group-1

present

MLST

Dicloxacillin 4 E.coli Yes ST131

Clindamycin 4 E.coli Yes ST131

Cefotaxime 2 E.coli Yes ST131

Cefuroxime 2 E.coli Yes ST131

Clindamycin 10 E.coli Yes ST131

Dicloxacillin 1 E.coli No ST602

Dicloxacillin 2 Parabacteroides spp. - -

Clindamycin 2 Enterococcus faecalis - -

Copenhagen 2014

41

Figure 3. LOG/CFU per 0.5 grams of faeces shown for antibiotics with selective effect on 65-Ec-09. The three

cultured populations are shown. Where SD is not shown SD=0.

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

80

5

10

15

Cefotaxime resistant E.coli

Clindamycin

CefotaximeCefuroximeDicloxaillin

Penicillin

Days After Inoculation

LOG

CFU

/0.5

g

Anaerobic Gram-negative

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

80

5

10

15

Days After Inoculation

LOG

CFU

/0.5

g

Gram-pos. aerobic

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

80

5

10

15

Days After Inoculation

LOG

CFU

/0.5

g

PhD Dissertation

Figure 4. LOG/CFU per 0.5 grams of faeces shown for antibiotics with limited selective effect on 65-Ec-09. The

three cultured populations are shown. Where SD is not shown SD=0.

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

0

5

10

15AmpicillinMeropenemCiprofloxacinMecillinamControl+ 65-Ec-09

Cefotaxime resistant E.coli

Days After Inoculation

LOG

CFU

/0.5

g

Anaerobic Gram-negative

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

80

5

10

15

Days After Inoculation

LOG

CFU

/0.5

g

Gram-pos. aerobic

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

8

Day

1

Day

2

Day

4

Day

80

5

10

15

Days After Inoculation

LOG

CFU

/0.5

g

Copenhagen 2014

43

Figure 5. LOG/CFU per 0.5 grams of faeces shown for prolonged colonization of three antibiotics. Gram-positive

aerobic flora was not cultured during this study. Where SD is not shown SD=0.

Cefotaxime resistant E.coli

Day 1

Day 2

Day 4

Day 8

Day 10

Day 14

Day 1

Day 2

Day 4

Day 8

Day 10

Day 14

Day 1

Day 2

Day 4

Day 8

Day 10

Day 14

0

5

10

15 CefuroximeDicloxacillinClindamycin

Days After Inoculation

LOG

CF

U/0

.5g

Anaerobic Gram-negative

Day 1

Day 2

Day 4

Day 8

Day 10

Day 14

Day 1

Day 2

Day 4

Day 8

Day 10

Day 14

Day 1

Day 2

Day 4

Day 8

Day 10

Day 14

0

5

10

15CefuroximeDicloxacillinClindamycin

Days After Inoculation

LOG

CF

U/0

.5g

PhD Dissertation

5.2. Case Control Study (Manuscript II)

Objective II

To investigate epidemiological factors associated with acquiring UTI with ESBL-producing E.coli,

in general practices.

In the study period we collected 98 ESBL positive urine samples, 174 resistant E.coli, 177

susceptible E.coli and 200 uninfected urine samples, respectively. All associated E.coli isolates

were analysed molecularly. A few patients were, however, excluded from the analysis of

epidemiological data from DST, due to a missing date of birth. Hence, we excluded two patients

from the ESBL group, three from the resistant group and two from the susceptible group. No

patients were excluded from the uninfected group

5.2.1. Urine samples to DCM

An overview of urines submitted to DCM is shown in Table 3. We have calculated p-values by use

of Fishers exact test. These data are listed in written text when appropriate.

In the years 2011 and 2012 more than 450 general practitioners submitted 110.471 urine samples to

DCM. Of these 48.541 (44%) were positive for bacterial growth and 23.001 (47%) were positive for

growth of E.coli with 4% of E.coli isolates being resistant to cefpodoxime (CPD-R). Determination

of CPD-R is used as an indicator of possible ESBL-production.

For all 649 patient primarily included in the study, a total of 1773 urine samples were submitted to

the DCM for investigation within one year prior to sampling. More than 90% of urine samples from

the three case groups were positive for bacterial growth, with just 33% of the samples for the

uninfected control group.

Patients in the ESBL and resistant groups had submitted significantly more urine samples to DCM

compared to the uninfected group, during 365 days, as well as within 90 days, prior to sampling

(P<0.05 for all). Compared to the uninfected control group, we found that all three case groups had

significantly more urine samples positive for bacterial growth and significantly more were positive

for E.coli within 90 days before and after sampling (P<0.0001). We observed significantly more

urine samples positive for CDP-R E.coli in the ESBL group than all other groups (P<0.0001) before

and after sampling. However, the three case groups submitted significantly fewer positive urine

Copenhagen 2014

45

samples in the 90 days after the index UTI compared to the prior 90 days. Furthermore, the ESBL

group submitted significantly less CPD-R E.coli (P<0.0001). There was no change in the number of

CPD-R E.coli in the other case groups.

Table 3. An overview of urine samples submitted to DCM from 365 days prior to and within 90 days after

sampling. ×The percentage shown at positive samples are the percentages of submitted urine samples being

positive. . *P<0.05 compared to the uninfected group. ▪P<0.05 compared to the resistant group. °P>0.05

compared to the susceptible group.

Positive samples E.coli n (%) CPD Resistant E.coli n (%)

ESBL (n=98) 341*° (91.2×) 192* (56.3) 138*° (71.9)

Negative (n=200) 129 (33×) 31 (24) 3 (9.7)

One year before

index sample

Resistant (n=174) 529*° (90.6×) 339* (64.1) 12*° (3.5)

Susceptible (n=177) 388* (91.5×) 264* (68) 0

All (n=649) 1387 (78.2×) 826 (59.6) 153 (18.5)

90-0 days before

index sample

ESBL (n=98) 183* (93.4×) 126* (68.9) 105*▪° (57.4)

Negative (n=200) 77 (26.1×) 22 (28.6) 3 (3.9)

Resistant (n=174) 328* (96.2×) 229* (69.8) 8° (2.4)

Susceptible (n=177) 281* (94.9×) 215* (76.5) 0

All (n=649) 869 (77×) 592 (68.1) 116 (13.3)

0-90 days after

index sample

ESBL (n=98) 117*° (83×) 60*° (51.3) 48*▪° (80)

Negative (n=200) 39 (48.1×) 10 (25.6) 0

Resistant (n=174) 159*° (88.8×) 77*° (48.5) 4* (5.2)

Susceptible (n=177) 101* (81.5×) 44* (43.6) 1 (2.3)

All (n=649) 416 (79.2×) 191 (45.9) 53 27.7)

PhD Dissertation

5.2.2. Description of E.coli Populations

Susceptibility patterns and phylogroups

As indicated in the group criteria all isolates from the susceptible group were fully susceptible to all

tested antibiotics. Susceptibility patterns and distribution of phylogroups are shown in Figure 6 and

Table 4, respectively. We found phylogroup B2 and D to be the dominating phylogroups in all three

case groups, representing 41-58% and 22-28% respectively.

Percentage of Isolates Resistant to antibiotics

0 20 40 60 80 100

Ampicillin

Cefuroxime

Gentamycin

Aztreonam

Ciprofloxacin

*Sulfamethoxazole

Trimethroprim

*Tetracycline

Ceftazidime

Ampicillin/Clavulanic acid

Tobramycin

Nitrofurantoin

*Piperacillin/Tazobactam

Mecillinam

*Fosfomycin

Meropenem

ESBLResistant

Percentage of Isolates

Ant

ibio

tics

Figure 6. Antibiotic susceptibility pattern for E.coli isolates in the ESBL-producing and the resistant (non-ESBL)

groups.

Copenhagen 2014

47

Table 4. Distribution of phylogroups as found by use of the early Clermont et al. methodology amongst all

collected E.coli isolates. # = Number of isolates.

ESBL ISOLATES n = 98 RESISTANT ISOLATES n = 174 SUSCEPTIBLE ISOLATES n = 177

Phylogr. # Percent # Percent # Percent

A 9 9% 20 11% 19 11%

B1 6 6% 5 3% 2 1%

B2 57 58% 71 41% 91 51%

D 22 22% 49 28% 42 24%

Non-Type 4 4% 29 17% 22 12%

ESBL Genotypes

The distribution of ESBL genotypes are shown in figure 7.

Figure 7. Distribution of major ESBL Genotypes amongst the ESBL-producing E.coli isolates.

For the ESBL population 73% carried a blaCTX-M group 1 and 24% a blaCTX-M group 9. For the

remaining two isolates we were not able to detect a blaCTX-M and found blaTEM only. A total of 54%

of the isolates belonged to CTX-M-15 with CTX-M-14 and CTX-M-27 as two other large groups of

12% and 12%, respectively. Fifteen isolates also had an AmpC phenotype, but no plasmid-borne

AmpC gene was found in any of the strains.

TEM

CT-X-M Gr. 9

CTX-M Gr. 1

PhD Dissertation

5.2.3. Statistical Analysis of Risk Factors and Population Data

In the following paragraphs results of statistical analysis for risk factors are briefly listed.

Results from the initial univariate t-tests are not shown. Patient demographics and exposure to

antibiotics, defined as prescriptions one year prior to sampling, are found in Appendix 1.

Appropriate p-values from chi-square and Fisher´s exact test are found Appendix 2 and 3 in

combination with the results of the multivariate logistic regression analyses.

5.2.3.1. Multivariate Logistic Regression Models

The most important findings will be stated below.

Patient characteristics

Classifications as COI, HCAI and complicated UTI have been done according to previously stated

criteria defined in Manuscript II. Exposures to antibiotics are depicted in Appendix 1 as number of

individuals who received prescriptions and total number of prescriptions prescribed.

Patients included in the three case groups proved to be significantly older than the uninfected

controls (P<0.0001). We found, however, no significant difference between ages in the case groups

(P>0.05). Interestingly there were more men among the uninfected group than in the resistant and

the susceptible groups, but no significant difference in the number of men between the case groups.

Nonetheless, in the multivariate analysis, we found a significant difference between gender as well

as age for all case groups (P<0.005 and P<0.0001).

Epidemiological features for case groups compared to the uninfected group

The multivariate logistic regression analyses are shown in Appendix 2. We performed the

multivariate analyses for the time periods 365 days, 90 days and 30 days before and 30 days and 90

days after sampling. We have, nevertheless, decided to describe findings for 90 days and 30 days

prior to sampling and briefly included selected interesting results from the other time periods.

In the 90 days prior to sampling we found limited antibiotics and diagnoses to be independent risk

factors. For antibiotics, only use of penicillin in the backward elimination analysis was significant

for the ESBL group (P=0.015). Conversely, exposure to five and two specific antibiotics proved to

be independent risk factors for the resistant and susceptible groups, respectively (all P<0.05).

Copenhagen 2014

49

Looking at 30 days prior to sampling, exposure to antibiotics in general proved statistically

significant for all three case groups, when analysed by chi-square (P<0.0001). Yet, just mecillinam

(P<0.0001) was a risk factor for the two resistant groups with mecillinam and sulfamethoxazole

(P=0.003 and P=0.03) being independent risk factors in the susceptible group. For the ESBL group,

diagnoses “kidney infections” and “urosepsis” were risk factors (P=0.01 and P=0.03). Of note, only

for the susceptible group use of antibiotics was an independent risk factor (data not shown).

When looking, in particular, at the backward stepwise elimination procedure, only including

variables with P<0.1, use of antibiotics increased in significance for all three groups. The significant

antibiotics have been added to Appendix 2.

Among the E.coli case groups we found no significant difference in number of community- and

healthcare-associated infections, as defined by us (P>0.05, results not shown).

In the period of 90 days after sampling, use of sulfamethizole was significant in all case groups

(P=0.03, P=0.007 and P=0.001) and mecillinam in the ESBL group (P=0.006) with use of

nitrofurantoin showing significance in the two other case groups (P=0.008 and P=0.04).

Epidemiological features for the ESBL case group compared to the other case groups

These results are found in Appendix 3. When calculated by chi-square there were significantly more

HCAI patients in the ESBL group compared to the susceptible group (P=0.006).

The number of patients receiving antibiotics and total number of prescriptions made did not clearly

show that the ESBL group had a higher use of antibiotics before the index sample (Appendix 1).

The multivariate analyses in the two runs, identified the total use of antibiotics as a risk factor for

the ESBL group 90 days prior to index sample (P=0.0049 and P=0.0029). Hospital admission 30

days prior was likewise an independent risk factor for ESBL (P=0.0073 and P=0.0216).

Finally, we saw that 90 days after infection, the use of penicillin/beta-lactamase inhibitors were

significantly higher in the ESBL group compared to the resistant group (P=0.02) and the use of

penicillins with effect on Gram-negatives and specifically the use of mecillinam were higher in the

ESBL than in the susceptible group (P=0.04 and P=0.002).

PhD Dissertation

5.3. Characterization of E.coli populations (Manuscript III)

Objective III

To investigate if the population structure of uropathogenic E.coli populations is predominantly

composed of related isolates, regardless of resistance profile.

The phylogroup and ESBL genotype distributions as well as the susceptibility patterns of the E.coli

populations included here were very similar to what was found during the case control study (Table

5 and Manuscript III). Briefly, antibiotic resistance for the resistant group was found with 84%

being resistant to ampicillin and 46-68% being resistant to trimethoprim, tetracycline and

sulphonamides.

Table 5. Distribution of phylogroups amongst E.coli isolates included in the characterization study. # =

number of isolates.

ESBL n = 98 Resistant n = 94 Susceptible n = 94

Phylogroups # Percent # Percent # Percent

A 9 9% 7 7% 9 10%

B1 6 6% 3 3% 1 1%

B2 57 58% 42 45% 52 55%

D 22 22% 30 32% 22 23%

Non-Type 4 4% 12 13% 10 11%

5.3.1. Distribution of Serogroups, MLVA codes and Sequence Types

Distribution of serogroups, a-MLVA codes and Sequence Types is found in Figure 8-11 and in

detail in Appendix 4-6.

Of the ESBL-producing isolates 94 were sent for serogrouping accompanied by 49 resistant (non-

ESBL) isolates and 51 susceptible E.coli. We found a variety of serogroups amongst the selected

isolates, with 53 different serogroups. For three isolates (two in the ESBL and one in the susceptible

group) it was not possible to distinguish between two serogroups.

There were 17% of the ESBL isolates, 12% of the resistant isolates and 14% of the susceptible

isolates which were classified as either “Negative” or “Multiple” after serogrouping.

Copenhagen 2014

51

PCR amplification of the six MLVA alleles was done as previously described and this abbreviated

MLVA was used to type the three strain collections. All isolates (100%) were typable by a-MLVA

and all isolates except for one, selected for MLST, had alleles successfully sequenced. One isolate

with a unique a-MLVA code was not sent for sequence typing.

In these three populations we found a total of 83 a-MLVA-codes and 72 sequence types. Of these

13 a-MLVA´s and 13 ST´s were found in two out of three populations and six a-MLVA´s and seven

ST´s were found in all three populations. Overall 10 a-MLVA codes were classified as unique

“unknown” sequence types, here classified as “New STs”. Generally, ST131 constituted 23% of all

isolates, ST69 constituted 10% and ST73 a total of 9%. Of the found a-MLVA codes 74 identified

just one sequence type. However, there were quite a few situations where one specific ST was

subdivided by more than one a-MLVA code, but also more complex situations where several

different ST´s were assigned the same a-MLVA code.

As result:

- ST 10 were subdivided by three different a-MLVA codes

- ST38 by three codes

- ST69 by three codes

- ST73 by ten codes

- ST95 by three codes

- ST141 by three codes

- ST357 by two codes

- ST405 by two codes

- ST648 by two codes and lastly

- ST131 were subdivided by four different codes.

Likewise, we found that the a-MLVA method could not distinguish certain sequence types and in

particular eight a-MLVA-codes did not classify unique sequence types. As such

- ST58, ST101 and ST448 shared one a-MLVA code. ST58 and ST448 are double-locus

variants.

- ST998 and one of the a-MLVA codes for ST141 were identical, were ST141 and ST998 are

single-locus variants.

- It was not possible to discriminate between on isolate of ST14 and ST1193 which are also

single-locus variants.

PhD Dissertation

- ST38, ST117 and ST1177 were assigned a single a-MLVA code. Here ST38 and ST1177

are single locus variant with ST117 being unrelated.

- One a-MLVA code for ST10 also identified ST746, ST1598 and one New ST. Here ST10

and ST746 are single-locus variants while the new ST is a double-locus variant of ST10.

- A different ST10 a-MLVA code was identical to codes for ST93, ST540, ST617, ST2279

and a New ST where only ST617 belonged to ST Complex 10 and was a double-locus

variant of ST10.

- Also ST354 and a New ST had the same code.

- Finally ST88, ST410 and one New ST had a common code with ST88 and ST410 being

double locus variants and belonging to ST Complex 23.

The ESBL-producing E.coli Population

In this ESBL population we found a total of 26 serogroups, 20 a-MLVA codes and 20 different

sequence types. We found no New ST´s in this population. In the serogroups we found one large

cluster with 38% belonging to group O25 and some minor clusters with 6% being serogroup O153,

5% serogroup O16 and 4% serogroup O15 (Appendix 4).

As described above, we saw different a-MLVA codes recognized as identical ST´s and found

different ST´s with indistinguishable a-MLVA codes. Among these isolates there were seven

clusters of a-MLVA codes and five clusters of ST´s. The population was clearly dominated by a

few ST´s as 50 isolates belonged to ST131. These ST131 isolates were identified by three a-MLVA

codes, where the largest of these (153562) contained 44 isolates. Of the 44 isolates, 35 belonged to

the serogroup O25 with 2 isolates belonging to serogroup O77 and O97. The remaining isolates had

no specific serogroup identified. The two other ST131 a-MLVA codes were comprised of 5 and 1

isolates, respectively, all of which belonged to serogroup O16.

The Resistant E.coli Population

Here 19 serogroups, 36 a-MLVA codes and 30 ST´s were identified. Among the isolates there were

three larger clusters with serogroup O25, O75 and O73 totalled 16%, 14% and 10%, respectively. In

addition, there were some minor clusters with serogroup O6, O11 and O15 representing 6-8%

(Appendix 5).

Copenhagen 2014

53

The a-MLVA codes and ST´s were composed of eight MLVA clusters and eight clusters of ST´s

and the population was, like the ESBL-population, dominated by few clusters of ST´s. Hence,

ST69, ST73 and ST131 comprised the largest ST cluster with 17, 10 and 14 isolates, respectively.

The Susceptible E.coli Population

In the population of susceptible isolates we found two larger clusters of serogroups and numerous

very small collections or single-isolate serogroups. The two clusters were serogroup O2 and O6

with 18% and 10% (Appendix 6).

Among the susceptible isolates 29 serogroups, 54 a-MLVA codes and 44 ST´s were found. We saw

seven a-MLVA clusters and eight clear ST clusters. Here ST73, as identified by multiple codes and

ST95 dominated, but also a-MLVA codes for ST10/93/540 and ST141/ST998 were frequent. This

population was comprised of a more diverse and heterogeneous group of E.coli.

Percentage of identifed a-MLVA codes,STs and O-groups

MLVAMLST

Serogro

ups0

50

100

150 Susceptible

Resistant

ESBL

Typing methods

Figure 8. Shown are the percentages of a-MLVA codes, STs and O-serogroup found in each of the three

populations.

PhD Dissertation

Figure 9. Distribution of major serogroups among the three E.coli populations. We show groups with 4% or more of isolates. Groups with <4% are found among singe isolates serogroup.

O25

Single Isolate Serotypes

Multiple

O153

Negative

O16O15ESBL

O25

O75

Single Isolate Serotypes

O73

O6

O15

Multiple

O11

O2NegativeResistens

Single Isolate Serotypes

O2

O6

Multiple

O1

O4

O18

O88Negative

Susceptible

Copenhagen 2014

55

153562

Single Isolate

173050

132251

131261

173277

163562

161160

131250 224665131291

ESBL

Single Isolate

224665132251

276655

223653

131250

173277

101261

131261124645264953266655

131281152365

204665

224743253263 254665 266562

Susceptible

Single Isolate

173277

153562

276655

132251

131261

124645

132261

163562

131250254575

223653

173050121250

124643 162562 173270

Resistent

Figure 10. Distribution of major clusters of a-MLVA codes among three E.coli populations. We show codes with 2% or more of isolates. Codes with <2% are found among singe isolates.

PhD Dissertation

ST131

Single Isolate ST

ST38

ST69

ST10

ST101

ST648

ST998 ST2852

ESBL

Single Isolate ST

ST69

ST131ST73

ST10

ST1193

ST58

ST405

ST95

ST88 ST80 ST1597

Resistent

ST73

ST998

ST95

ST141ST10

ST69

ST127

ST93

ST223

ST357

ST14

ST38

ST101

ST12ST48

ST59ST540

ST4235ST131Susceptible

Figure 11. Distribution of major clusters of sequence types among three E.coli populations. We show STs with 2% or more of isolates, except for ST131 which only constituted 1% of isolates.

Copenhagen 2014

57

6. Discussion

6.1. Selection of CTX-M-producing E.coli in vivo

One of the advantages of using animal models is the relatively rapid study of large populations. In

addition, animal experiments can be repeated and are reproducible (195). Testing on animals, however,

can give an estimation of effects only and there are the obvious limitations of extrapolating from animals

to humans.

We developed a mouse model for the investigation of intestinal colonization without the need for

prior suppression of the indigenous flora by antibiotics. It was hereby possible to illustrate the

selective ability of common antibiotics with different spectra, when an ESBL-producing, virulent

E.coli lineage was introduced into the intestines. We confirmed that an antibiotic, clindamycin,

suppressing the Bacteroides population, with no activity against Enterobacteriaceae, allows for

overgrowth of an ESBL-producing E.coli in the mouse gut. Conversely, antibiotics active against

the colonizing strain will not promote overgrowth. Beta-lactam antibiotics with no inhibiting effects

on the ESBL-producing E.coli showed diverse levels of selection. Interestingly, dicloxacillin

promoted proliferation of the ESBL-producing E.coli, even with no apparent impact on the Gram-

negative anaerobic flora or other Gram-negative bacteria. On the other hand, and very surprisingly,

ciprofloxacin, with known effect on Enterobacteriaceae, had no selective abilities. Yet, treatment

with fluoroquinolones can in some instances reach faecal concentrations above MIC of even

resistant strains, a feature not measured by us (93). There are several other limitations concerning

our mouse model, besides possible problems regarding extrapolation from mice to humans. Most

importantly however, we did not investigate antibiotic impact on the total microflora of the

intestines. We could therefore not study shifts in dominating phyla or changes in the bacterial

species. Finally, it was not possible to observe alterations in the Enterobacteriaceae population

which could potentially describe features influencing colonization.

Nonetheless, we found that the indigenous microflora provides an important protection by

inhibiting colonization. Our findings strongly indicate that other parts of the flora than Gram-

negative anaerobes play an important part upholding colonization resistance. Antibiotics may

therefore disrupt the indigenous microflora, inhibiting colonization resistance and promoting

proliferation. This disturbance may be caused by a variety of antibiotics, and level of selection is

PhD Dissertation

unforeseen by spectra. Antibiotic with limited impact on Gram-negative bacteria can select for

resistant E.coli, as seen for penicillin and dicloxacillin (196).

Disruption of the anaerobic flora, or at least the Bacteroides population, seems to play a particularly

important part in preventing prolonged colonization by MDR pathogenic E.coli.

6.2. Investigations of epidemiological factors

The case-control study performed here is, to our knowledge, the first triple-case-control study of

epidemiological characteristics for UTI caused by E.coli. It has, however, been designed with

several limitations. First, the study exclusively includes non-hospitalized patients of all ages with no

knowledge on antibiotic treatment given in hospitals. Secondly, as we included patients with

previous UTI we might be collecting and investigating patients in the middle of a course of

infection, posing an apparent cause for antibiotic prescriptions. It would have been ideal, if we had

selected only cases where the E. coli infection was the first UTI with at least one year lapse to

previous UTI. The lack of data on nursing home residence as well as outpatient contacts will lead us

to underestimate the significance of healthcare exposures and make it impossible to truly identify

the impact of these locations as reservoirs for ESBL-producing E.coli.

Nevertheless, in this unique triple-case control study we found exposure to antibiotics to be

associated with UTI in general. Healthcare contact and previous UTI infection by ESBL-producing

E.coli were associated with community-onset UTI with ESBL-producing E. coli. This is not

surprising based on previous studies of epidemiological features (15,17,18,70,73,197,198).

However, the new aspects of the epidemiology identified here were the very few differences found

between the UTI case groups. There was no clear difference in exposure to antibiotics and no

significant difference in age or gender, nor was there a significant difference (P>0.05) between the

percentages of HCAI infections in the age group 18-64 and >65 (Appendix 1). There was,

conversely, an association between hospital admissions and ESBL infection when comparing the

case groups. This confirms hospital contacts as an independent risk factor for ESBL-producing

E.coli infections (15,17,18,70,73,197). The ESBL group had significantly more UTIs caused by

CPD-R E.coli, in all investigated time periods. Antibiotics found to select in the mouse intestinal

colonisation model were not identified as risk factors in this present case-control study.

Nevertheless, in Denmark cephalosporins are prescribed in hospital setting only, data we did not

Copenhagen 2014

59

investigate. Furthermore, very few patients were exposed to dicloxacillin and clindamycin which

might be the reason we did not find them as risk factors.

In conclusion, we found exposure to antibiotics to be an unreliable predictor of UTI with resistant

E.coli. We could not link specific diagnoses to the ESBL group, except kidney infections and

urosepsis. The combination of our finding with previous studies suggests that, in low prevalent

countries at least, exposure to antibiotics, comorbidity, age and gender are not necessarily decent

predictors of risk, but healthcare association, travelling and previous carriage of, and infection by,

ESBL-producing E.coli are features which needs to be explored by GPs (17–19,73,93,179).

6.3. Characterization of E.coli populations

We characterized three E.coli populations with different susceptibility patterns by use of an easily

performed and fast typing method. The abbreviated MLVA could assign 71% of isolates to

corresponding sequence types, simply comparing this novel method to a gold standard method with

known nomenclature. Data can furthermore be reproduced and compared to other laboratories. In

this study we chose to apply different typing methods on parts of our collection of isolates. This

increases the discriminatory power enabling us to identify the diversity of different UPEC

populations (190). With the availability of whole genome sequencing, at lower costs and with high

discriminatory power, new methods are at risk of being typing methods with no real impact, unless

price and speed overcomes lack of power (181). There is, however, still need for cheap analytic

tools for surveillance of resistant lineages and detection of outbreaks, a need persistently nourished

by the dissemination of MDR E.coli (11).

MLST and MLVA schemes analyse a small portion of the genome only, and these genes might be

subject to lateral transfer between strains, leading to somewhat incongruent phylogenetic trees, not

correlating with the genome content of a bacterium (20). Changes in VNTR regions will not

necessarily mean a change in ST, and vice versa. There is a higher rate of sustainable genetic

alterations within the VNTR regions, than in the housekeeping genes used for MLST. This might be

the reason for incomplete separation of all STs, but it provides a high discriminatory power and the

ability to separate closely related ST-lineages (187,188). The limitation of the abbreviated MLVA

was the inability to distinguish specific STs, which has been previously described for many of the

MLVA codes. Often the indistinguishable isolates were closely related STs, perhaps indicating a

kinship not recognized by MLST. By use of serogrouping and a-MLVA we described the STs as

PhD Dissertation

extremely heterogeneous lineages. We found, as has previously been seen, that ESBL-producing

E.coli are clustered in homogenous populations dominated by few lineages (199). We identified a

high level of diversity among susceptible E.coli isolates, a level of diversity which, to our

knowledge, has not previously been described (9,131,136,142). The successful lineage of ST131

was similar to other studies dominating this Danish ESBL-producing E.coli population.

Interestingly, a-MLVA seemed to sub-divide ST131 isolates based on O-serogroup, showing the

abilities to survey minor differences in E.coli lineages. The study is of course limited by the

relatively small number of isolates characterized and by the fact that the collection of isolates within

a very limited time period does not allow us to identify the fluctuation in dominant lineages of non-

ESBL populations.

We do, nonetheless, conclude based on serogroups, a-MLVA and MLST, that in this collection of

uropathogenic E.coli from general practices the populations were dominated by different

serogroups, MLVA-codes and ST´s and that the susceptible E.coli population was a much more

diverse group of isolates. Within a dominating ST-lineage, like ST73, we could sub-divide isolates

by a-MLVA. Conversely, the resistant E.coli populations and in particular the ESBL-producing

E.coli are disseminated as related lineages, indicating that the success of resistant lineages is mainly

due to positive selection of UPEC with gained resistance and limited loss of fitness.

Copenhagen 2014

61

7. Conclusion and Perspectives

Our knowledge on the mechanisms behind antibiotic disruption of the microbiota is limited. We

know that antibiotic therapy alters the composition of the microflora and healthy volunteers

undergoing a 5-day course of ciprofloxacin had changes in the flora not resettling for months (3,12).

Antibiotic treatment changes the microbiota and selects for antibiotic resistant pathogens increasing

the risk of dissemination of resistant clones, subsequently increasing risk of infection (11,200). Our

first objective was to evaluate the ability of common antibiotics, to select for a CTX-M-15-

producing E.coli isolate belonging to ST131 in vivo. This was successfully done in a mouse

intestinal colonisation model developed and tested as part of this research. We found that antibiotics

with variation in spectra of activity can select for MDR, virulent E.coli lineages, making new

studies on antibiotic impact on the microflora a necessity to understand selection.

The current increase in faecal carriage of ESBL-producing E.coli, seen in healthy individuals and

returning traveller’s, strongly suggests a rapid dissemination of ESBL-producing isolates in the

community (104,107,108,201). Reports of carriers with no recent hospitalization or antibiotic

treatment indicate long-term carriage proposing a further risk for person-to-person transmission

(108). To what extent ESBL-producing E. coli will spread in the community is unknown, but the

potential for dissemination of resistant lineages like ST131 has proven exceptionally high

(104,199,202). Especially the combination of virulent E.coli lineages and the CTX-M type ESBL,

often showing resistance to ciprofloxacin, shows that the dissemination could be related to the

selective pressure exerted by cephalosporins or fluoroquinolones (104). However, since different

CTX-M types differ in resistance, it might reflect the various antibiotics pressure and reservoirs

involved (104).

Our second objective was to investigate epidemiological factors associated with acquiring UTI with

ESBL-producing E.coli, in general practices. We conducted a case-control study and identified new

aspects of which risk factors are associated to COI UTI in Denmark. The results of our triple-case

control study, in concordance with previous findings, raises the question if COI ESBL infections are

in fact healthcare-associated, at least in countries of low ESBL prevalence (42). Previous antibiotic

treatment was a risk factor for UTI as has previously been seen for UTI in general and for UTI

resistant bacteria (11). We found that patients in the ESBL group were more likely to belong to this

particular group, prior to our sampling but also after sampling, than the non-ESBL case groups.

These individuals might be periodic low-level carriers or treatment can correctly eradicate ESBL-

PhD Dissertation

producing E. coli, why we found significantly less ESBL-producing E.coli during the 90 days after

sampling (93). Yet, strictly COI with ESBL-producing E.coli are being reported and true clusters of

cases in the community, and among family members, are described (42). Overestimating the risk of

ESBL-producing E.coli can lead to the use of broad-spectrum antibiotics leading to a higher

selection pressure hereby preserving existing isolates and allowing for the emergence of novel

resistant strains (179).

As our third objective we wished to investigate if the population structure of uropathogenic E.coli

populations is predominantly composed of related isolates, regardless of resistance profile. Such a

characterization is of great interest to help determine if ST-lineages producing and spreading ESBL

are successful UPEC adapting to environments with antibiotics or if plasmids, carrying ESBL, are

taken up by less prevalent UPEC gaining a new advantage.

This characterization of E.coli populations was successfully done by typing of the three

susceptibility groups by use of a-MLVA, MLST and serogrouping. The a-MLVA method was here

efficient in the characterization of resistant E.coli populations and could possibly help sub-divide

MLST-lineages (57,187,203). In non-clonal populations, there is no lineage structure why such

populations are a bigger challenge when it comes to bacterial typing, than typing of highly clonal

populations (181). Information from a single locus or few loci can be unreliable in identifying

genetic relatedness, why the use of multiple loci is essential to achieve the resolution required

(181,184). Generally it can be more efficient to examine genes displaying more diversity especially

in population investigations (181). This makes the combination of MLVA and MLST a potential

robust method for surveillance. When studying heterogenic E.coli populations by MLST only,

community ExPEC are classified as clonal groups of E. coli, regardless of drug resistance (8). This

would advocate that antibiotic resistance is not a requirement for related dissemination and suggests

that susceptible E. coli strains do disseminate clonally (8). Yet, when such E.coli populations are

described by more than one typing method, the susceptible isolates are found to be a heterogonous

group, as previously determined by fimH-typing and as determined by us in this thesis. We

conclude that the susceptible E.coli populations are not disseminated as related lineages in nearly

the same degree as ESBL-producing E.coli. Reports from other research groups, suggests that some

E.coli lineages acquire different types of mobile resistance genes, as seen with ST69 and especially

ST131, while other lineages remain fairly susceptible and rarely take up plasmids (8,204). It is

therefore very likely that antibiotic selection pressure creates this clonal-like population structure

while antibiotic free environments allows for competition and subsequent non-related population

Copenhagen 2014

63

structure (138,181). As an example, ST393 has become more frequent as its resistance profile

increases, with no change in virulence and we still see common non-ESBL and non-MDR E.coli

lineages on a global scale as we also detected a clear difference in dominating lineages (7,130). The

lineage ST69 has been found as the dominant faecal E. coli in healthy individuals. Thus, ExPEC are

well adapted to the gut of humans (11,104). As the abundance of faecal ESBL-producing E.coli is a

predictor of infection and antibiotic exposure increases ESBL-producing E.coli faecal count,

constraining use of antibiotics might maintain ESBL count at low levels, hereby minimizing the

probability of infection (93). However, ST131 must be a specialized UPEC, present in all

populations, capable of surviving in the community once present. Thus, ST131 could be one of the

UPEC lineages which are able to effectively colonize the human gut, causing UTI simply since they

dominate the microflora after antibiotic exposure (11). We speculate if resistance in E.coli is

defined by intrinsic differences in different E. coli lineages, allowing for a limited number of UPEC

ST-lineages to obtain and carry blaCTX-M.

The identified distribution of MDR E.coli indicates the existence of reservoirs for ESBL-producing

E.coli. It does, likewise, raise the question if plasmids carrying ESBL genes or not that easily transferred

between E.coli strains, why plasmids require certain E.coli lineages combined with selection pressure to

effectively spread. The likely existence of reservoirs of ESBL-producing E.coli creates a need for reliable,

cheap and fast characterization of pathogenic E.coli, to identify these reservoirs and lessen subsequent

distribution (203). Nonetheless, since not all ESBL-producing E.coli belong to known and identical

lineages, and as plasmids can be acquired by horizontal gene exchange, restraining ST131 and lineages

alike will not completely stop the spread of ESBL-producing E. coli (115). Virulence factors do not

necessarily influence abundance of faecal ESBL-producing E.coli and faecal dominance has previously

been found to be more important than virulence in the pathogenesis of UTI (93). It is therefore likely that,

even with the VFs conferring transmissibility and colonization of global lineages, other less resistant but

“classic” ExPEC as well as commensal E.coli could be adequate colonizing competitors, without

antibiotic selection pressure (115,138).

In future studies, we must identify external reservoirs of ESBL-producing E.coli, which could be nursing

homes, long-term care facilities and hospitals as previously stated, but impossible for us to determine

(205). However, a Swedish study did not find increased level of resistance in nursing homes from 2003-

12 and only very few cases of ESBL-producing E.coli with recent antibiotic treatment and hospitalisation

PhD Dissertation

during the last six months predicted higher resistance rates (206). Therefore, we must understand how

patients acquire MDR E.coli, how antibiotics affect the intestinal flora and creates a possible reservoir,

how long they carry MDR E.coli and finally discover if ESBL-producing E.coli are more likely to cause

disease than susceptible E.coli. ESBL-producing E.coli might simply be the pathogens found since they

dominate the flora. Such studies would require survey of numerous individuals of different ages and with

no history of ESBL carriages. However, carrier studies could be performed as mice colonisation

experiments with treatment given for more than three days and more than once a day. Such studies could

be prolonged to see impact on time of carriage or mice could be re-treated as CFU counts drop to detect

non-countable presence of resistant E.coli. The mouse intestinal colonisation model could furthermore be

used with different resistant UPEC or the level of conjugation between STs in the mouse gut could be

investigated. Finally, it would be of great interest to see the impact of antibiotic on the microflora,

performed as studies of the microbiome of mice, before and after antibiotic treatment. Here shifts in

domination phyla, anaerobic and aerobic and Gram-positive and –negative would be interesting to

investigate. The combination of such mice studies would help to clarify features involved in colonisation

and the dissemination of ESBL-producing E.coli.

Copenhagen 2014

65

8. References

1. Ferry S a., Holm SE, Stenlund H, Lundholm R, Monsen TJ. The natural course of uncomplicated lower urinary tract infection in women illustrated by a randomized placebo controlled study. Scand J Infect Dis. 2004 Jan;36(4):296–301.

2. Lipsky B a. Prostatitis and urinary tract infection in men: what’s new; what's true? Am J Med. 1999 Mar;106(3):327–34.

3. Foxman B. The epidemiology of urinary tract infection. Nat Rev Urol. Nature Publishing Group; 2010 Dec;7(12):653–60.

4. Kahlmeter G, Poulsen HO. Antimicrobial susceptibility of Escherichia coli from community-acquired urinary tract infections in Europe: the ECO·SENS study revisited. Int J Antimicrob Agents. Elsevier B.V.; 2012 Jan;39(1):45–51.

5. Foxman B. Urinary tract infection syndromes: occurrence, recurrence, bacteriology, risk factors, and disease burden. Infect Dis Clin North Am. Elsevier Inc; 2014 Mar;28(1):1–13.

6. Rice LB. The clinical consequences of antimicrobial resistance. Curr Opin Microbiol. 2009 Oct [cited 2011 Jul 13];12(5):476–81.

7. Riley LW. Pandemic lineages of extraintestinal pathogenic Escherichia coli. Clin Microbiol Infect. 2014 Apr 26;

8. Adams-Sapper S, Diep BA, Perdreau-Remington F, Riley LW. Clonal composition and community clustering of drug-susceptible and -resistant Escherichia coli isolates from bloodstream infections. Antimicrob Agents Chemother. 2013 Jan;57(1):490–7.

9. Banerjee R, Johnston B, Lohse C, Chattopadhyay S, Tchesnokova V, Sokurenko E V, et al. The clonal distribution and diversity of extraintestinal Escherichia coli varies according to patient characteristics. Antimicrob Agents Chemother. 2013 Sep 16;(September):1–29.

10. Pitout JDD. Extraintestinal Pathogenic Escherichia coli: A Combination of Virulence with Antibiotic Resistance. Front Microbiol. 2012 Jan;3(January):9.

11. Nielsen KL, Dynesen P, Larsen P, Frimodt-Møller N. Faecal Escherichia coli from patients with E. coli urinary tract infection and healthy controls who have never had a urinary tract infection. J Med Microbiol. 2014 Apr;63(Pt 4):582–9.

12. Foxman B, Goldberg D. Why the human microbiome project should motivate epidemiologists to learn ecology. Epidemiology. 2010;21(6):757–9.

13. Ejernæs K, Stegger M, Reisner A, Ferry S, Monsen T, Holm SE, et al. Characteristics of Escherichia coli causing persistence or relapse of urinary tract infections. 2011;2(6):528–37.

14. Drekonja DM, Johnson JR. Urinary tract infections. Prim Care. 2008 Jun;35(2):345–67, vii.

PhD Dissertation

15. Doi Y, Park YS, Rivera JI, Adams-Haduch JM, Hingwe A, Sordillo EM, et al. Community-associated extended-spectrum β-lactamase-producing Escherichia coli infection in the United States. Clin Infect Dis. 2013 Mar;56(5):641–8.

16. Banerjee R, Johnston B, Lohse C, Stephen B, Clabots C, Johnson JR, et al. Escherichia coli Sequence Type 131 Is a Dominant, Antimicrobial- Resistant Clonal Group Associated with Healthcare. 2013;34(4):361–9.

17. Rogers B a, Ingram PR, Runnegar N, Pitman MC, Freeman JT, Athan E, et al. Community-Onset Escherichia coli Infection Resistant to Expanded-Spectrum Cephalosporins in Low-Prevalence Countries. Antimicrob Agents Chemother. 2014 Apr;58(4):2126–34.

18. Søraaas A, Sundsfjord AS, Sandven I, Brunborg C, Jenum PP a, Søraas A. Risk Factors for Community-Acquired Urinary Tract Infections Caused by ESBL-Producing Enterobacteriaceae – A Case – Control Study in a Low Prevalence Country. PLoS One. PLoS One; 2013 Jan;8(7):1–7.

19. Nicolas-Chanoine M-H, Jarlier V, Robert J, Arlet G, Drieux L, Leflon-Guibout V, et al. Patient’s origin and lifestyle associated with CTX-M-producing Escherichia coli: a case-control-control study. PLoS One. 2012 Jan;7(1):e30498.

20. Dobrindt U, Hacker JH. Current Topics in Microbiology and Immunology Between Pathogenicity and Commensalism.

21. Dobrindt U. (Patho-)Genomics of Escherichia coli. Int J Med Microbiol. 2005 Oct;295(6-7):357–71.

22. Pitout JDD. Extraintestinal pathogenic Escherichia coli: an update on antimicrobial resistance, laboratory diagnosis and treatment. Expert Rev Anti Infect Ther. 2012 Oct;10(10):1165–76.

23. Russo T a, Johnson JR. Proposal for a new inclusive designation for extraintestinal pathogenic isolates of Escherichia coli: ExPEC. J Infect Dis. 2000 May;181(5):1753–4.

24. Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nat Rev Microbiol. 2004 Feb ;2(2):123–40.

25. Johnson JR, Russo T a. Extraintestinal pathogenic Escherichia coli: “The other bad E coli.”J Lab Clin Med. 2002 Mar;139(3):155–62.

26. Bailey JK, Pinyon JL, Anantham S, Hall RM. Distribution of human commensal Escherichia coli phylogenetic groups. J Clin Microbiol. 2010 Sep;48(9):3455–6.

27. Johnson JR, Russo T a. Uropathogenic Escherichia coli as agents of diverse non-urinary tract extraintestinal infections. J Infect Dis. 2002 Sep 15;186(6):859–64.

28. Clermont O, Bonacorsi S, Bingen E. Rapid and simple determination of the Escherichia coli phylogenetic group. Appl Environ Microbiol. 2000 Oct;66(10):4555–8.

Copenhagen 2014

67

29. Clermont O, Christenson JK, Denamur E, Gordon DM. The Clermont Escherichia coli phylo-typing method revisited: improvement of specificity and detection of new phylo-groups. Environ Microbiol Rep. 2013 Feb;5(1):58–65.

30. Bleibtreu A, Gros P-A, Laouénan C, Clermont O, Le Nagard H, Picard B, et al. Fitness, stress resistance, and extraintestinal virulence in Escherichia coli. Infect Immun. 2013 Aug ;81(8):2733–42.

31. Barroso-Batista J, Sousa A, Lourenço M, Bergman M-L, Sobral D, Demengeot J, et al. The first steps of adaptation of Escherichia coli to the gut are dominated by soft sweeps. PLoS Genet. 2014 Mar;10(3):e1004182.

32. Croxall G, Hale J, Weston V, Manning G, Cheetham P, Achtman M, et al. Molecular epidemiology of extraintestinal pathogenic Escherichia coli isolates from a regional cohort of elderly patients highlights the prevalence of ST131 strains with increased antimicrobial resistance in both community and hospital care settings. 2011;(August):2501–8.

33. Skippen I, Shemko M, Turton J, Kaufmann ME. Epidemiology of infections caused by Escherichia coli and Klebsiella spp .: a nested caseecontrol study from a tertiary hospital in London. Infection. 2006;

34. Ho J, Tambyah P a, Paterson DL. Multiresistant Gram-negative infections: a global perspective. Curr Opin Infect Dis. 2010 Dec;23(6):546–53.

35. Mcdermott PF, Walker RD, White DG. Antimicrobials�: Modes of Action and Mechanisms of Resistance. 2003;135–43.

36. Sommer MO a, Dantas G, Church GM. Functional characterization of the antibiotic resistance reservoir in the human microflora. Science. 2009 Aug 28;325(5944):1128–31.

37. Schjørring S, Struve C, Krogfelt K a. Transfer of antimicrobial resistance plasmids from Klebsiella pneumoniae to Escherichia coli in the mouse intestine. J Antimicrob Chemother. 2008 Nov;62(5):1086–93.

38. Alanis AJ. Resistance to antibiotics: are we in the post-antibiotic era? Arch Med Res. 2005 ;36(6):697–705.

39. Wright GD. Antibiotic resistance in the environment: a link to the clinic? Curr Opin Microbiol. Elsevier Ltd; 2010 Oct;13(5):589–94.

40. Llarrull LI, Testero S a, Fisher JF, Mobashery S. The future of the β-lactams. Curr Opin Microbiol. Elsevier Ltd; 2010 Oct;13(5):551–7.

41. ECDC. Antimicrobial resistance surveillance in Europe 2012. 2012.

42. Falagas ME, Karageorgopoulos DE. Extended-spectrum beta-lactamase-producing organisms. J Hosp Infect. Elsevier Ltd; 2009 Dec;73(4):345–54.

PhD Dissertation

43. Donskey CJ. Antibiotic regimens and intestinal colonization with antibiotic-resistant gram-negative bacilli. Clin Infect Dis . 2006 Sep 1;43 Suppl 2(Suppl 2):S62–9.

44. Oteo J, Navarro C, Cercenado E, Delgado-Iribarren A, Wilhelmi I, Orden B, et al. Spread of Escherichia coli strains with high-level cefotaxime and ceftazidime resistance between the community, long-term care facilities, and hospital institutions. J Clin Microbiol . 2006 Jul ;44(7):2359–66.

45. Bush K. Alarming β-lactamase-mediated resistance in multidrug-resistant Enterobacteriaceae. Curr Opin Microbiol . Elsevier Ltd; 2010 Oct;13(5):558–64.

46. Novais A, Comas I, Baquero F, Cantón R, Coque TM, Moya A, et al. Evolutionary trajectories of beta-lactamase CTX-M-1 cluster enzymes: predicting antibiotic resistance. PLoS Pathog . 2010 Jan;6(1):e1000735.

47. Peirano G, Pitout JDD. Molecular epidemiology of Escherichia coli producing CTX-M beta-lactamases: the worldwide emergence of clone ST131 O25:H4. Int J Antimicrob Agents . 2010 Apr;35(4):316–21.

48. Yao JD, Moellering R. Manual of Clinical Microbiology. 8th. ed. Murray PR, Baron EJ, Jorgensen J, Pfaller MA, Yolken RH, editors. 2003.

49. Cantón R, González-Alba JM, Galán JC. CTX-M Enzymes: Origin and Diffusion. Front Microbiol . 2012 Jan;3(April):110.

50. Bush K, Jacoby G a. Updated functional classification of beta-lactamases. Antimicrob Agents Chemother . 2010 Mar;54(3):969–76.

51. Paterson DL, Bonomo RA. Extended-Spectrum Beta -Lactamases�: a Clinical Update. Society. 2005;18(4):657–86.

52. Rogers B a, Sidjabat HE, Paterson DL. Escherichia coli O25b-ST131: a pandemic, multiresistant, community-associated strain. J Antimicrob Chemother. 2011 Jan;66(1):1–14.

53. Pitout JDD, Laupland KB. Enterobacteriaceae�: an emerging public-health concern. 2008;8(March):159–66.

54. Naseer U, Sundsfjord A. The CTX-M conundrum: dissemination of plasmids and Escherichia coli clones. Microb Drug Resist . 2011 Mar;17(1):83–97.

55. Hawkey PM, Jones AM. The changing epidemiology of resistance. J Antimicrob Chemother . 2009 Sep;64 Suppl 1:i3–10.

56. Tian SF, Chen BY, Chu YZ, Wang S. Prevalence of rectal carriage of extended-spectrum b -lactamase-producing Escherichia coli among elderly people in community settings in China. 2008;785:781–5.

Copenhagen 2014

69

57. Nielsen JB, Albayati A, Jørgensen RL, Hansen KH, Lundgren B, Schønning K. An abbreviated MLVA identifies Escherichia coli ST131 as the major extended-spectrum β-lactamase-producing lineage in the Copenhagen area. Eur J Clin Microbiol Infect Dis . 2013 Nov 6;32(3):431–6.

58. Kerrn MB, Klemmensen T, Espersen F. Susceptibility of Danish Escherichia coli strains isolated from urinary tract infections and bacteraemia , and distribution of sul genes conferring sulphonamide resistance. 2002;513–6.

59. Kjerulf A, Hansen DS, Sandvang D, Hansen F, Frimodt-Møller N. The prevalence of ESBL-producing E. coli and Klebsiella strains in the Copenhagen area of Denmark. APMIS . 2008 Feb;116(2):118–24.

60. Hansen DS, Schumacher H, Hansen F, Stegger M, Hertz FB, Schønning K, et al. Extended-spectrum β-lactamase (ESBL) in Danish clinical isolates of Escherichia coli and Klebsiella pneumoniae: prevalence, β-lactamase distribution, phylogroups, and co-resistance. Scand J Infect Dis . 2012 Mar;44(3):174–81.

61. DANMAP 2012. DANMAP. 2012;

62. Jakobsen L, Kurbasic A, Skjøt-rasmussen L, Ejrnæs K, Porsbo LJ, Pedersen K, et al. Escherichia coli Isolates from Broiler Chicken Meat , Broiler Chickens , Pork , and Pigs Share Phylogroups and Antimicrobial Resistance with Community-Dwelling. 2010;7(5).

63. Tenover FC. Mechanisms of antimicrobial resistance in bacteria. Am J Infect Control . 2006 Jun;34(5 Suppl 1):S3–10; discussion S64–73.

64. Russo T a, Johnson JR. Medical and economic impact of extraintestinal infections due to Escherichia coli: focus on an increasingly important endemic problem. Microbes Infect . 2003 Apr;5(5):449–56.

65. Jakobsen L, Garneau P, Bruant G, Harel J, Olsen SS, Porsbo LJ, et al. Is Escherichia coli urinary tract infection a zoonosis? Proof of direct link with production animals and meat. Eur J Clin Microbiol Infect Dis . 2012 Jun;31(6):1121–9.

66. Bush K, Courvalin P, Dantas G, Davies J, Eisenstein B, Huovinen P, et al. Tackling antibiotic resistance. Nat Rev Microbiol . Nature Publishing Group; 2011 Dec;9(12):894–6.

67. Vardakas KZ, Rafailidis PI, Konstantelias A a, Falagas ME. Predictors of mortality in patients with infections due to multi-drug resistant Gram negative bacteria: the study, the patient, the bug or the drug? J Infect . Elsevier Ltd; 2013 May;66(5):401–14.

68. Kang C-I, Wi YM, Ko KS, Chung DR, Peck KR, Lee NY, et al. Outcomes and risk factors for mortality in community-onset bacteremia caused by extended-spectrum beta-lactamase-producing Escherichia coli, with a special emphasis on antimicrobial therapy. Scand J Infect Dis . 2013 Mar 19;(November 2012):1–7.

PhD Dissertation

69. Rodríguez-Baño J, Picón E, Gijón P, Hernández JR, Cisneros JM, Peña C, et al. Risk factors and prognosis of nosocomial bloodstream infections caused by extended-spectrum-beta-lactamase-producing Escherichia coli. J Clin Microbiol . 2010 May;48(5):1726–31.

70. Hayakawa K, Gattu S, Marchaim D, Bhargava A, Palla M, Alshabani K, et al. Epidemiology and Risk Factors for Isolation of Escherichia coli Producing CTX-M-Type Extended-Spectrum β-Lactamase in a Large U.S. Medical Center. Antimicrob Agents Chemother . 2013 Aug;57(8):4010–8.

71. Vardakas KZ, Tansarli GS, Rafailidis PI, Falagas ME. Carbapenems versus alternative antibiotics for the treatment of bacteraemia due to Enterobacteriaceae producing extended-spectrum β-lactamases: a systematic review and meta-analysis. J Antimicrob Chemother . 2012 Aug 21;(August):2793–803.

72. Marchaim D, Gottesman T, Schwartz O, Korem M, Maor Y, Rahav G, et al. National multicenter study of predictors and outcomes of bacteremia upon hospital admission caused by Enterobacteriaceae producing extended-spectrum beta-lactamases. Antimicrob Agents Chemother . 2010 Dec;54(12):5099–104.

73. Apisarnthanarak A, Kiratisin P, Saifon P, Kitphati R, Dejsirilert S, Mundy LM. Clinical and molecular epidemiology of community-onset, extended-spectrum beta-lactamase-producing Escherichia coli infections in Thailand: a case-case-control study. Am J Infect Control . 2007 Nov;35(9):606–12.

74. Hsieh C, Shen Y, Hwang K. Clinical Implications , Risk Factors and Mortality Following Community-onset Bacteremia Caused by Extended-spectrum b-lactamase ( ESBL ) and non-ESBL Producing Escherichia coli. J Microbiol Immunol Infect . Taiwan Society of Microbiology; 2010;43(3):240–8.

75. Kanafani ZA, Mehio-sibai A, Araj GF, Kanaan M, Kanj SS. Epidemiology and risk factors for producing organisms�: A case control study at a tertiary care center in. Control. 2005;21–3.

76. Kang C, Song J-H, Chung DR, Peck KR, Ko KS, Yeom J, et al. Risk factors and treatment outcomes of community-onset bacteraemia caused by extended-spectrum beta -lactamase-producing Escherichia coli. Int J Antimicrob Agents . 2010 Sep;36(3):284–7.

77. Lautenbach E, Patel JB, Bilker WB, Edelstein PH, Fishman NO. Extended-spectrum beta-lactamase-producing Escherichia coli and Klebsiella pneumoniae: risk factors for infection and impact of resistance on outcomes. Clin Infect Dis . 2001 Apr 15;32(8):1162–71.

78. Melzer M, Petersen I. Mortality following bacteraemic infection caused by extended spectrum beta-lactamase (ESBL) producing E. coli compared to non-ESBL producing E. coli. J Infect . 2007 Sep [cited 2012 Feb 8];55(3):254–9.

79. Tumbarello M, Spanu T, Di Bidino R, Marchetti M, Ruggeri M, Trecarichi EM, et al. Costs of bloodstream infections caused by Escherichia coli and influence of extended-spectrum-

Copenhagen 2014

71

beta-lactamase production and inadequate initial antibiotic therapy. Antimicrob Agents Chemother . 2010 Oct;54(10):4085–91.

80. Tosh PK, McDonald LC. Infection control in the multidrug-resistant era: tending the human microbiome. Clin Infect Dis . 2012 Mar 1;54(5):707–13.

81. Dantas G, Sommer MO a, Degnan PH, Goodman AL. Experimental approaches for defining functional roles of microbes in the human gut. Annu Rev Microbiol . 2013 Sep 8;67:459–75.

82. Leatham MP, Banerjee S, Autieri SM, Mercado-Lubo R, Conway T, Cohen PS. Precolonized human commensal Escherichia coli strains serve as a barrier to E. coli O157:H7 growth in the streptomycin-treated mouse intestine. Infect Immun . 2009 Jul;77(7):2876–86.

83. Edlund C, Nord CE. Effect on the human normal microflora of oral antibiotics for treatment of urinary tract infections. J Antimicrob Chemother . 2000 Sep;46 Suppl 1:41–8; discussion 63–5.

84. Raum E, Lietzau S, von Baum H, Marre R, Brenner H. Changes in Escherichia coli resistance patterns during and after antibiotic therapy: a longitudinal study among outpatients in Germany. Clin Microbiol Infect . 2008 Jan;14(1):41–8.

85. Licht TR, Struve C, Christensen BB, Poulsen RL, Molin S, Krogfelt K a. Evidence of increased spread and establishment of plasmid RP4 in the intestine under sub-inhibitory tetracycline concentrations. FEMS Microbiol Ecol . 2003 May 1;44(2):217–23.

86. Feld L, Schjørring S, Hammer K, Licht TR, Danielsen M, Krogfelt K, et al. Selective pressure affects transfer and establishment of a Lactobacillus plantarum resistance plasmid in the gastrointestinal environment. J Antimicrob Chemother . 2008 Apr;61(4):845–52.

87. Harmoinen J, Mentula S, Heikkila M, Rest M Van Der, Rajala-schultz J, Donskey CJ, et al. Orally Administered Targeted Recombinant Beta-Lactamase Prevents Ampicillin-Induced Selective Pressure on the Gut Microbiota�: a Novel Approach to Reducing Antimicrobial Resistance. Society. 2004;48(1):75–9.

88. Faure S, Perrin-Guyomard a, Delmas JM, Chatre P, Laurentie M. Transfer of plasmid-mediated CTX-M-9 from Salmonella enterica serotype Virchow to Enterobacteriaceae in human flora-associated rats treated with cefixime. Antimicrob Agents Chemother . 2010 Jan;54(1):164–9.

89. Bailey JK, Pinyon JL, Anantham S, Hall RM. Commensal Escherichia coli of healthy humans: a reservoir for antibiotic-resistance determinants. J Med Microbiol . 2010 Nov;59(Pt 11):1331–9.

90. Kristiansson C, Grape M, Gotuzzo E, Samalvides F, Chauca J, Larsson M, et al. Socioeconomic factors and antibiotic use in relation to antimicrobial resistance in the Amazonian area of Peru. Scand J Infect Dis . 2009 Jan;41(4):303–12.

PhD Dissertation

91. Gustafsson I, Sjölund M, Torell E, Johannesson M, Engstrand L, Cars O, et al. Bacteria with increased mutation frequency and antibiotic resistance are enriched in the commensal flora of patients with high antibiotic usage. J Antimicrob Chemother . 2003 Oct;52(4):645–50.

92. Olofsson SK, Marcusson LL, Strömbäck A, Hughes D, Cars O. Dose-related selection of fluoroquinolone-resistant Escherichia coli 2007.pdf. J Antimicrob Chemother . 2007 Oct ;60(4):795–801.

93. Ruppé E, Lixandru B, Cojocaru R, Büke C, Paramythiotou E, Angebault C, et al. Relative Fecal Abundance of Extended-Spectrum-β-Lactamase-Producing Escherichia coli Strains and Their Occurrence in Urinary Tract Infections in Women. Antimicrob Agents Chemother . 2013 Sep;57(9):4512–7.

94. Kluytmans J a JW, Overdevest ITM a, Willemsen I, Kluytmans-van den Bergh MFQ, van der Zwaluw K, Heck M, et al. Extended-spectrum β-lactamase-producing Escherichia coli from retail chicken meat and humans: comparison of strains, plasmids, resistance genes, and virulence factors. Clin Infect Dis . 2013 Feb;56(4):478–87.

95. Nataro JP, Kaper JB. Diarrheagenic Escherichia coli. Clin Microbiol Rev . 1998 Jan;11(1):142–201.

96. Chapman PA. Sources of Escherichia coli 0157 and experiences over the past 15 years in Sheffield , UK. 2000;

97. 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.

98. Hammerum AM, Larsen J, Andersen VD, Lester CH, Skovgaard Skytte TS, Hansen F, et al. Characterization of extended-spectrum β-lactamase (ESBL)-producing Escherichia coli obtained from Danish pigs, pig farmers and their families from farms with high or no consumption of third- or fourth-generation cephalosporins. J Antimicrob Chemother . 2014 Jun 7;1–8.

99. Hammerum a M, Lester CH, Jakobsen L, Porsbo LJ. Faecal carriage of extended-spectrum β-lactamase-producing and AmpC β-lactamase-producing bacteria among Danish army recruits. . Clinical microbiology and infection�: the official publication of the European Society of Clinical Microbiology and Infectious Diseases. 2011. p. 566–8.

100. Jørgensen SB, Samuelsen O, Sundsfjord A, Bhatti SA, Jørgensen I, Sivapathasundaram T, et al. High prevalence of faecal carriage of ESBL-producing Enterobacteriaceae in Norwegian patients with gastroenteritis. Scand J Infect Dis . 2014 Apr 4;(1478):1–4.

101. Ostholm-Balkhed A, Tärnberg M, Nilsson M, Nilsson LE, Hanberger H, Hällgren A. Travel-associated faecal colonization with ESBL-producing Enterobacteriaceae: incidence and risk factors. J Antimicrob Chemother . 2013 Sep [cited 2014 Jul 7];68(9):2144–53.

Copenhagen 2014

73

102. Wickramasinghe NH, Xu L, Eustace A, Shabir S, Saluja T, Hawkey PM. High community faecal carriage rates of CTX-M ESBL-producing Escherichia coli in a specific population group in Birmingham, UK. J Antimicrob Chemother . 2012 Mar 8;1–6.

103. Luvsansharav U-O, Hirai I, Nakata A, Imura K, Yamauchi K, Niki M, et al. Prevalence of and risk factors associated with faecal carriage of CTX-M β-lactamase-producing Enterobacteriaceae in rural Thai communities. . The Journal of antimicrobial chemotherapy. 2012. p. 1769–74.

104. Nicolas-Chanoine M-H, Gruson C, Bialek-Davenet S, Bertrand X, Thomas-Jean F, Bert F, et al. 10-Fold increase (2006-11) in the rate of healthy subjects with extended-spectrum β-lactamase-producing Escherichia coli faecal carriage in a Parisian check-up centre. J Antimicrob Chemother . 2013 Mar;68(3):562–8.

105. Lausch KR, Fuursted K, Larsen CS, Storgaard M. Colonisation with multi-resistant Enterobacteriaceae in hospitalised Danish patients with a history of recent travel: a cross-sectional study. Travel Med Infect Dis . Elsevier Ltd; 2013;11(5):320–3.

106. Tham J, Odenholt I, Walder M, Brolund A, Ahl J, Melander E. Extended-spectrum beta-lactamase-producing Escherichia coli in patients with travellers’ diarrhoea. Scand J Infect Dis . 2010 Apr;42(4):275–80.

107. Peirano G, Laupland KB, Gregson DB, Pitout JDD. Colonization of Returning Travelers With CTX-M-Producing Escherichia coli. J Travel Med . 2011 Sep;18(5):299–303.

108. Tham J, Walder M, Melander EVA, Odenholt I. Duration of colonization with extended-spectrum beta-lactamase-producing Escherichia coli in patients with travellers’ diarrhoea. Scand J Infect Dis . 2012 Jan 31;46(December 2011):1–5.

109. Rooney PJ, O’Leary MC, Loughrey AC, McCalmont M, Smyth B, Donaghy P, et al. Nursing homes as a reservoir of extended-spectrum beta-lactamase (ESBL)-producing ciprofloxacin-resistant Escherichia coli. J Antimicrob Chemother . 2009 Sep;64(3):635–41.

110. Colomer-Lluch M, Mora A, López C, Mamani R, Dahbi G, Marzoa J, et al. Detection of quinolone-resistant Escherichia coli isolates belonging to clonal groups O25b:H4-B2-ST131 and O25b:H4-D-ST69 in raw sewage and river water in Barcelona, Spain. J Antimicrob Chemother . 2013 Apr;68(4):758–65.

111. Clermont O, Olier M, Hoede C, Diancourt L, Brisse S, Keroudean M, et al. Animal and human pathogenic Escherichia coli strains share common genetic backgrounds. Infect Genet Evol . 2011 Apr;11(3):654–62.

112. Johnson JR, Delavari P, Stell a L, Whittam TS, Carlino U, Russo T a. Molecular comparison of extraintestinal Escherichia coli isolates of the same electrophoretic lineages from humans and domestic animals. J Infect Dis . 2001 Jan 1;183(1):154–9.

113. Jakobsen L, Hammerum AM, Frimodt-Møller N. Detection of clonal group A Escherichia coli isolates from broiler chickens, broiler chicken meat, community-dwelling humans, and

PhD Dissertation

urinary tract infection (UTI) patients and their virulence in a mouse UTI model. Appl Environ Microbiol . 2010 Dec;76(24):8281–4.

114. Leverstein-van Hall M a, Dierikx CM, Cohen Stuart J, Voets GM, van den Munckhof MP, van Essen-Zandbergen a, et al. Dutch patients, retail chicken meat and poultry share the same ESBL genes, plasmids and strains. Clin Microbiol Infect . 2011 Jun;17(6):873–80.

115. Foxman B. Extended-spectrum β-lactamase-producing escherichia coli in the united states: time to rethink empirical treatment for suspected E. coli infections? Clin Infect Dis . 2013 Mar;56(5):649–51.

116. Krych L, Hansen CHF, Hansen AK, van den Berg FWJ, Nielsen DS. Quantitatively Different, yet Qualitatively Alike: A Meta-Analysis of the Mouse Core Gut Microbiome with a View towards the Human Gut Microbiome. PLoS One . 2013 Jan;8(5):e62578.

117. Knudsen JD, Frimodt-møller N. Animal Models in Bacteriology. 2001;9:1–14.

118. Jakobsen L, Cattoir V, Jensen KS, Hammerum AM, Nordmann P, Frimodt-Møller N. Impact of low-level fluoroquinolone resistance genes qnrA1, qnrB19 and qnrS1 on ciprofloxacin treatment of isogenic Escherichia coli strains in a murine urinary tract infection model. J Antimicrob Chemother . 2012 Oct;67(10):2438–44.

119. Leatham MP, Stevenson SJ, Gauger EJ, Krogfelt KA, Lins JJ, Haddock TL, et al. Mouse Intestine Selects Nonmotile flhDC Mutants of Escherichia coli MG1655 with Increased Colonizing Ability and Better Utilization of Carbon Sources. 2005;73(12):8039–49.

120. Donskey CJ, Helfand MS, Pultz NJ, Rice LB. Effect of Parenteral Fluoroquinolone Administration on Persistence of Vancomycin-Resistant Enterococcus faecium in the Mouse Gastrointestinal Tract. Society. 2004;48(1):326–8.

121. Hoyen CK, Pultz NJ, Paterson DL, Aron DC, Donskey CJ. Effect of Parenteral Antibiotic Administration on Establishment of Intestinal Colonization in Mice by Klebsiella pneumoniae Strains Producing Extended-Spectrum Beta-Lactamases. Society. 2003;47(11):3610–2.

122. Pultz MJ, Donskey CJ. Effects of imipenem-cilastatin, ertapenem, piperacillin-tazobactam, and ceftriaxone treatments on persistence of intestinal colonization by extended-spectrum-beta-lactamase-producing Klebsiella pneumoniae strains in mice. Antimicrob Agents Chemother . 2007 Aug;51(8):3044–5.

123. Perez F, Pultz MJ, Endimiani A, Bonomo R a, Donskey CJ. Effect of antibiotic treatment on establishment and elimination of intestinal colonization by KPC-producing Klebsiella pneumoniae in mice. Antimicrob Agents Chemother . 2011 Jun;55(6):2585–9.

124. Møller AK, Leatham MP, Conway T, Nuijten PJM, Haan LAM De, Krogfelt KA, et al. An Escherichia coli MG1655 Lipopolysaccharide Deep-Rough Core Mutant Grows and Survives in Mouse Cecal Mucus but Fails To Colonize the Mouse Large Intestine. 2003;71(4):2142–52.

Copenhagen 2014

75

125. Flückiger U, Segessenmann C, Gerber a U. Integration of pharmacokinetics and pharmacodynamics of imipenem in a human-adapted mouse model. Antimicrob Agents Chemother . 1991 Sep;35(9):1905–10.

126. Stiefel U, Pultz NJ, Donskey CJ. Effect of carbapenem administration on establishment of intestinal colonization by vancomycin-resistant enterococci and Klebsiella pneumoniae in mice. Antimicrob Agents Chemother . 2007 Jan;51(1):372–5.

127. Oteo J, Diestra K, Juan C, Bautista V, Novais A, Pérez-Vázquez M, et al. Extended-spectrum beta-lactamase-producing Escherichia coli in Spain belong to a large variety of multilocus sequence typing types, including ST10 complex/A, ST23 complex/A and ST131/B2. Int J Antimicrob Agents . 2009 Aug;34(2):173–6 .

128. Hernández J, Martínez-Martínez L, Cantón R, Coque T, Pascual A. Nationswide Study of Escherichia Coli and Klebsiella pneumoniae Producing Extended-Spectrum beta -Lactamases in Spain. 2005;49(5):2122–5.

129. Colpan A, Johnston B, Porter S, Clabots C, Anway R, Thao L, et al. Escherichia coli Sequence Type 131 (ST131) Sub-clone H30 as an Emergent Multidrug-Resistant Pathogen Among US Veterans. Clin Infect Dis . 2013 Aug 29;131(9):1–10.

130. Olesen B, Scheutz F, Menard M, Skov MN, Kolmos HJ, Kuskowski M a, et al. Three-decade epidemiological analysis of Escherichia coli O15:K52:H1. J Clin Microbiol . 2009 Jun;47(6):1857–62.

131. Johnson JR, Tchesnokova V, Johnston B, Clabots C, Roberts PL, Billig M, et al. Abrupt emergence of a single dominant multidrug-resistant strain of Escherichia coli. J Infect Dis . 2013 Mar 15;207(6):919–28.

132. Olesen B, Hansen DS, Nilsson F, Frimodt-Møller J, Leihof RF, Struve C, et al. Prevalence and characteristics of the epidemic multiresistant Escherichia coli ST131 clonal group among extended-spectrum beta-lactamase-producing E. coli isolates in Copenhagen, Denmark. J Clin Microbiol . 2013 Jun;51(6):1779–85.

133. Johnson JR, Tchesnokova V, Johnston B, Clabots C, Roberts PL, Billig M, et al. Abrupt emergence of a single dominant multidrug-resistant strain of Escherichia coli. J Infect Dis . 2013 Mar 15;207(6):919–28.

134. Mora A, Blanco M, López C, Mamani R, Blanco JE, Alonso MP, et al. Emergence of clonal groups O1:HNM-D-ST59, O15:H1-D-ST393, O20:H34/HNM-D-ST354, O25b:H4-B2-ST131 and ONT:H21,42-B1-ST101 among CTX-M-14-producing Escherichia coli clinical isolates in Galicia, northwest Spain. Int J Antimicrob Agents . 2011 Jan;37(1):16–21.

135. Gibreel TM, Dodgson AR, Cheesbrough J, Fox AJ, Bolton FJ, Upton M. Population structure, virulence potential and antibiotic susceptibility of uropathogenic Escherichia coli from Northwest England. J Antimicrob Chemother . 2012 Feb;67(2):346–56.

PhD Dissertation

136. Blanco J, Mora A, Mamani R, López C, Blanco M, Dahbi G, et al. National survey of Escherichia coli causing extraintestinal infections reveals the spread of drug-resistant clonal groups O25b:H4-B2-ST131, O15:H1-D-ST393 and CGA-D-ST69 with high virulence gene content in Spain. J Antimicrob Chemother . 2011 Sep;66(9):2011–21.

137. Naseer U, Haldorsen B, Tofteland S, Hegstad K, Scheutz F, Simonsen GS, et al. Molecular characterization of CTX-M-15-producing clinical isolates of Escherichia coli reveals the spread of multidrug-resistant ST131 (O25:H4) and ST964 (O102:H6) strains in Norway. APMIS . 2009 Jul;117(7):526–36.

138. Bengtsson S, Naseer U, Sundsfjord A, Kahlmeter G, Sundqvist M. Sequence types and plasmid carriage of uropathogenic Escherichia coli devoid of phenotypically detectable resistance. J Antimicrob Chemother . 2012 Jan;67(1):69–73.

139. Brisse S, Diancourt L, Laouénan C, Vigan M, Caro V, Arlet G, et al. Phylogenetic distribution of CTX-M- and non-extended-spectrum-β-lactamase-producing Escherichia coli isolates: group B2 isolates, except clone ST131, rarely produce CTX-M enzymes. J Clin Microbiol . 2012 Sep;50(9):2974–81

140. Aibinu I, Odugbemi T, Koenig W, Ghebremedhin B. Sequence type ST131 and ST10 complex (ST617) predominant among CTX-M-15-producing Escherichia coli isolates from Nigeria. Clin Microbiol Infect . 2012 Mar;18(3):E49–51.

141. Gibreel TM, Dodgson AR, Cheesbrough J, Bolton FJ, Fox AJ, Upton M. High metabolic potential may contribute to the success of ST131 uropathogenic Escherichia coli. J Clin Microbiol . 2012 Oct;50(10):3202–7.

142. Peirano G, van der Bij AK, Gregson DB, Pitout JDD. Molecular epidemiology over an 11-year period (2000 to 2010) of extended-spectrum β-lactamase-producing Escherichia coli causing bacteremia in a centralized Canadian region. Journal of clinical microbiology. 2012. p. 294–9.

143. Novais A, Pires J, Ferreira H, Costa L, Montenegro C, Vuotto C, et al. Characterization of globally spread Escherichia coli ST131 isolates (1991 to 2010). Antimicrob Agents Chemother . 2012 Jul;56(7):3973–6.

144. Kudinha T, Johnson JR, Andrew SD, Kong F, Anderson P, Gilbert GL. Distribution of phylogenetic groups, sequence type ST131, and virulence-associated traits among Escherichia coli isolates from men with pyelonephritis or cystitis and healthy controls. Clin Microbiol Infect . 2013 Apr;19(4):E173–80.

145. Johnson JR, Johnston B, Clabots C, Kuskowski M a, Castanheira M. Escherichia coli sequence type ST131 as the major cause of serious multidrug-resistant E. coli infections in the United States. Clin Infect Dis . 2010 Aug 1;51(3):286–94.

146. Blanco J, Mora A, Mamani R, López C, Blanco M, Dahbi G, et al. Four Main Virotypes among Extended-Spectrum β-Lactamase-Producing Isolates of Escherichia coli O25b:H4-

Copenhagen 2014

77

B2-ST131: Bacterial, Epidemiological, and Clinical Characteristics. J Clin Microbiol . 2013 Aug 7;(August):1–35.

147. Adams-Sapper S, Sergeevna-Selezneva J, Tartof S, Raphael E, Diep BA, Perdreau-Remington F, et al. Globally dispersed mobile drug-resistance genes in gram-negative bacterial isolates from patients with bloodstream infections in a US urban general hospital. J Med Microbiol . 2012 Jul;61(Pt 7):968–74.

148. Graffunder EM, Preston KE, Evans AM, Venezia R a. Risk factors associated with extended-spectrum b-lactamase-producing organisms at a tertiary care hospital. J Antimicrob Chemother . 2005 Jul;56(1):139–45.

149. Ben-Ami R, Rodríguez-Baño J, Arslan H, Pitout JDD, Quentin C, Calbo ES, et al. A multinational survey of risk factors for infection with extended-spectrum beta-lactamase-producing enterobacteriaceae in nonhospitalized patients. Clin Infect Dis . 2009 Sep 1;49(5):682–90.

150. Goyal A, Prasad KN, Prasad A, Gupta S, Ghoshal U, Ayyagari A. Extended spectrum β -lactamases in Escherichia coli & Klebsiella pneumoniae & associated risk factors. Indian J Med Res. 2009;(June):695–700.

151. Cassier P, Lallechère S, Aho S, Astruc K, Neuwirth C, Piroth L, et al. Cephalosporin and fluoroquinolone combinations are highly associated with CTX-M β-lactamase-producing Escherichia coli: a case-control study in a French teaching hospital. Clin Microbiol Infect . 2011 Nov;17(11):1746–51.

152. Rodríguez-Baño J, Navarro MD, Ngugro MD. Extended-spectrum beta-lactamases in ambulatory care: a clinical perspective. Clin Microbiol Infect . 2008 Jan;14 Suppl 1:104–10.

153. Wu U-I, Yang C-S, Chen W-C, Chen Y-C, Chang S-C. Risk factors for bloodstream infections due to extended-spectrum beta-lactamase-producing Escherichia coli. J Microbiol Immunol Infect . Taiwan Society of Microbiology; 2010 Aug;43(4):310–6.

154. Rodríguez-Baño J, Alcalá J, Cisneros JM, Grill F, Oliver A, Horcajada JP, et al. Escherichia coli producing SHV-type extended-spectrum beta-lactamase is a significant cause of community-acquired infection. J Antimicrob Chemother . 2009 Apr;63(4):781–4.

155. Banerjee R, Strahilevitz J, Johnson JR, Nagwekar PP, Schora DM, Shevrin I, et al. Predictors and Molecular Epidemiology of Community-Onset Extended-Spectrum β-Lactamase-Producing Escherichia coli Infection in a Midwestern Community. Infect Control Hosp Epidemiol . 2013 Sep;34(9):947–53.

156. Kaye KS, Harris AD, Samore M, Carmeli Y. The Case-Case-Control Study Design: Addressing The Limitations of Risk Factor Studies for antimicrobial resistance. 2005;26(4):346–51.

PhD Dissertation

157. Harris AD, Karchmer TB, Carmeli Y, Samore MH. Methodological principles of case-control studies that analyzed risk factors for antibiotic resistance: a systematic review. Clin Infect Dis . 2001 Apr 1;32(7):1055–61.

158. Wacholder S, Silverman DT, Mclaughlin JK, Mandel JS. Selection of Controls in Case-Control Studies. 1992;135(9):1029–41.

159. Harris AD, Samore MH, Carmeli Y. Control Group Selection Is an Important but Neglected Issue in Studies of Antibiotic Resistance. Ann Intern Med . 2000 Jul 18;133(2):158.

160. Wacholder S, Mclaughlin JK, Silverman DT, Mandel JS. Selection of controls in injury case-control studies. Ann Epidemiol . 2000 Oct 1;10(7):455.

161. Wener KM, Schechner V, Gold HS, Wright SB, Carmeli Y. Treatment with fluoroquinolones or with beta-lactam-beta-lactamase inhibitor combinations is a risk factor for isolation of extended-spectrum-beta-lactamase-producing Klebsiella species in hospitalized patients. Antimicrob Agents Chemother . 2010 May;54(5):2010–6.

162. Søraas A, Sundsfjord AS, Sandven I, Brunborg C, Jenum P. Risk Factors for Community-Acquired Urinary Tract Infections Caused by ESBL-Producing Enterobacteriaceae -A Case-Control Study in a Low Prevalence Country. PLoS One; 2013.

163. Friedman ND, Kaye KS, Stout JE, Mcgarry SA, Trivette SL, Briggs JP, et al. Health Care – Associated Bloodstream Infections in Adults�: A Reason To Change the Accepted Definition of Community-Acquired Infections. 2002;791–8.

164. Kang C-I, Wi YM, Lee MY, Ko KS, Chung DR, Peck KR, et al. Epidemiology and risk factors of community onset infections caused by extended-spectrum β-lactamase-producing Escherichia coli strains. J Clin Microbiol . 2012 Feb;50(2):312–7.

165. Briongos-Figuero LS, Gómez-Traveso T, Bachiller-Luque P, Domínguez-Gil González M, Gómez-Nieto a, Palacios-Martín T, et al. Epidemiology, risk factors and comorbidity for urinary tract infections caused by extended-spectrum beta-lactamase (ESBL)-producing enterobacteria. Int J Clin Pract . 2012 Sep;66(9):891–6.

166. Meier S, Weber R, Zbinden R, Ruef C, Hasse B. Extended-spectrum β-lactamase-producing Gram-negative pathogens in community-acquired urinary tract infections: an increasing challenge for antimicrobial therapy. Infection . 2011 Aug;39(4):333–40.

167. Azap OK, Arslan H, Serefhanoğlu K, Colakoğlu S, Erdoğan H, Timurkaynak F, et al. Risk factors for extended-spectrum beta-lactamase positivity in uropathogenic Escherichia coli isolated from community-acquired urinary tract infections. Clin Microbiol Infect . 2010 Feb;16(2):147–51.

168. Rodríguez-Baño J, Navarro MD, Romero L, Muniain M a, de Cueto M, Ríos MJ, et al. Bacteremia due to extended-spectrum beta -lactamase-producing Escherichia coli in the CTX-M era: a new clinical challenge. Clin Infect Dis . 2006 Dec 1;43(11):1407–14.

Copenhagen 2014

79

169. Quirante OF, Cerrato SG, Pardos SL. Risk factors for bloodstream infections caused by extended-spectrum β-lactamase-producing Escherichia coli and Klebsiella pneumoniae. . The Brazilian journal of infectious diseases�: an official publication of the Brazilian Society of Infectious Diseases. p. 370–6.

170. Park SH, Choi S-M, Lee D-G, Kim J, Choi J-H, Kim S-H, et al. Emergence of extended-spectrum β-lactamase-producing escherichia coli as a cause of community-onset bacteremia in South Korea: risk factors and clinical outcomes. Microb Drug Resist . 2011 Dec;17(4):537–44.

171. Rodríguez-Baño J, Picón E, Gijón P, Hernández JR, Ruíz M, Peña C, et al. Community-onset bacteremia due to extended-spectrum beta-lactamase-producing Escherichia coli: risk factors and prognosis. Clin Infect Dis . 2010 Jan 1;50(1):40–8.

172. Park K-H, Oh WS, Kim ES, Park SW, Hur J-A, Kim YK, et al. Factors associated with ciprofloxacin- and cefotaxime-resistant Escherichia coli in women with acute pyelonephritis in the emergency department. Int J Infect Dis . International Society for Infectious Diseases; 2014 Mar 18;

173. Qureshi Z a, Paterson DL, Pakstis DL, Adams-Haduch JM, Sandkovsky G, Sordillo E, et al. Risk factors and outcome of extended-spectrum β-lactamase-producing Enterobacter cloacae bloodstream infections. Int J Antimicrob Agents . Elsevier B.V.; 2011 Jan;37(1):26–32.

174. Bhavnani SM, Ambrose PG, Craig W a, Dudley MN, Jones RN. Outcomes evaluation of patients with ESBL- and non-ESBL-producing Escherichia coli and Klebsiella species as defined by CLSI reference methods: report from the SENTRY Antimicrobial Surveillance Program. Diagn Microbiol Infect Dis . 2006 Mar;54(3):231–6.

175. Han JH, Maslow J, Han X, Xie SX, Tolomeo P, Santana E, et al. Risk Factors for the Development of Gastrointestinal Colonization With Fluoroquinolone-Resistant Escherichia coli in Residents of Long-Term Care Facilities. J Infect Dis . 2013 Sep 17;209:420–5.

176. Fan N-C, Chen H-H, Chen C-L, Ou L-S, Lin T-Y, Tsai M-H, et al. Rise of community-onset urinary tract infection caused by extended-spectrum β-lactamase-producing Escherichia coli in children. J Microbiol Immunol Infect . Elsevier Taiwan LLC; 2013 Jul 5;1–7.

177. Park K-H, Oh WS, Kim ES, Park SW, Hur J-A, Kim YK, et al. Factors associated with ciprofloxacin- and cefotaxime-resistant Escherichia coli in women with acute pyelonephritis in the emergency department. Int J Infect Dis . International Society for Infectious Diseases; 2014 Mar 19;23:8–13.

178. Rodríguez-Baño J, Alcala JC, Cisneros JM, Grill F, Oliver A, Horcajada JP, et al.

Community Infections Caused by Extended-Spectrum ␤-Lactamase–Producing. Arch Intern Med. 2008;168(17):1897–902.

179. Goulenok T, Ferroni a, Bille E, Lécuyer H, Join-Lambert O, Descamps P, et al. Risk factors for developing ESBL E. coli: can clinicians predict infection in patients with prior colonization? J Hosp Infect . 2013 Aug;84(4):294–9.

PhD Dissertation

180. Urwin R, Maiden MCJ. Multi-locus sequence typing: a tool for global epidemiology. Trends Microbiol . 2003 Oct;11(10):479–87.

181. Maiden MCJ. Multilocus sequence typing of bacteria. Annu Rev Microbiol . 2006 Jan;60:561–88.

182. Nielsen KL, Godfrey P a, Stegger M, Andersen PS, Feldgarden M, Frimodt-Møller N. Selection of unique Escherichia coli clones by random amplified polymorphic DNA (RAPD): Evaluation by whole genome sequencing. J Microbiol Methods . Elsevier B.V.; 2014 Jun 6;103C:101–3.

183. Gordon DM, Clermont O, Tolley H, Denamur E. Assigning Escherichia coli strains to phylogenetic groups: multi-locus sequence typing versus the PCR triplex method. Environ Microbiol . 2008 Oct;10(10):2484–96.

184. Maiden MCJ, Bygraves JA, Feil E, Morelli G, Russel JE, Urwin R, et al. Multilocus sequence typing�: A portable approach to the identification of clones within populations of. 1998;95(March):3140–5.

185. Enright MC, Spratt BG. Multilocus sequence typing. Trends Microbiol . 1999 Dec;7(12):482–7.

186. Wirth T, Falush D, Lan R, Colles F, Mensa P, Wieler LH, et al. Sex and virulence in Escherichia coli: an evolutionary perspective. Mol Microbiol . 2006 Jun;60(5):1136–51.

187. Manges AR, Tellis P a, Vincent C, Lifeso K, Geneau G, Reid-Smith RJ, et al. Multi-locus variable number tandem repeat analysis for Escherichia coli causing extraintestinal infections. J Microbiol Methods . Elsevier B.V.; 2009 Nov;79(2):211–3.

188. Keys C, Kemper S, Keim P. Highly diverse variable number tandem repeat loci in the E. coli O157:H7 and O55:H7 genomes for high-resolution molecular typing. J Appl Microbiol . 2005 Jan;98(4):928–40.

189. Pop C, Brenner F, Fields P, Wells J, Strockbine N. Manual of Clinical Microbiology. 8th, volum. Murray PR, Baron EJ, Jorgensen JH, Pfaller MA, Yolken RH, editors. ASM Press; 2003.

190. Köhler C-D, Dobrindt U. What defines extraintestinal pathogenic Escherichia coli? Int J Med Microbiol . 2011 Dec;301(8):642–7.

191. Pedersen CB, Gøtzsche H, Møller JO, Mortensen PB. The Danish Civil Registration System. A cohort of eight million persons. Dan Med Bull . 2006 Nov;53(4):441–9.

192. Pedersen CB. The Danish Civil Registration System. Scand J Public Health . 2011 Jul;39(7 Suppl):22–5.

193. Kildemoes HW, Sørensen HT, Hallas J. The Danish National Prescription Registry. Scand J Public Health . 2011 Jul;39(7 Suppl):38–41.

Copenhagen 2014

81

194. Hansen F, Olsen SS, Heltberg O, Justesen US, Fuglsang-Damgaard D, Knudsen JD, et al. Characterization of Third-Generation Cephalosporin-Resistant Escherichia coli from Bloodstream Infections in Denmark. Microb Drug Resist . 2014 Feb 11;00(00):1–9.

195. Zak O, Sande MA, O’Reilly T. Handbook of Animal Models of Infection. Zak O, Sande MA, editors. Academic Press;

196. Frimodt-moller N, Ostri P, Pedersen IBK, Poulsen SR. Antibiotic Prophylaxis in Pulmonary Surgery - A Double-Blind Study of Penicillin Versus Placebo. 1980;195(4):444–50.

197. Ben-ami R, Arslan H, Pitout JDD, Quentin C, Calbo ES, Azap K, et al. A Multinational Survey of Risk Factors for Infection with Extended-Spectrum b -Lactamase – Producing Enterobacteriaceae in Nonhospitalized Patients. 2009;49.

198. Lautenbach E, Bilker WB, Tolomeo P, Maslow JN. Impact of diversity of colonizing strains on strategies for sampling Escherichia coli from fecal specimens. J Clin Microbiol . 2008 Sep ;46(9):3094–6.

199. Riley LW. Pandemic lineages of extraintestinal pathogenic Escherichia coli. Clin Microbiol Infect . 2014 May;20(5):380–90.

200. Sullivan a, Edlund C, Nord CE. Effect of antimicrobial agents on the ecological balance of human microflora. Lancet. Elsevier Ltd; 2001 Dec 2;1:101–14.

201. Van der Bij AK, Pitout JDD. The role of international travel in the worldwide spread of multiresistant Enterobacteriaceae. J Antimicrob Chemother . 2012 Sep;67(9):2090–100.

202. Nicolas-Chanoine M-H, Blanco J, Leflon-Guibout V, Demarty R, Alonso MP, Caniça MM, et al. Intercontinental emergence of Escherichia coli clone O25:H4-ST131 producing CTX-M-15. J Antimicrob Chemother . 2008 Feb;61(2):273–81.

203. Naseer U, Olsson-Liljequist BE, Woodford N, Dhanji H, Cantón R, Sundsfjord A, et al. Multi-locus variable number of tandem repeat analysis for rapid and accurate typing of virulent multidrug resistant Escherichia coli clones. PLoS One . 2012 Jan;7(7):e41232.

204. Bengtsson S, Naseer U, Sundsfjord A, Kahlmeter G, Sundqvist M. Sequence types and plasmid carriage of uropathogenic Escherichia coli devoid of phenotypically detectable resistance. J Antimicrob Chemother . 2012 Jan;67(1):69–73.

205. Rooney PJ, O’Leary MC, Loughrey AC, McCalmont M, Smyth B, Donaghy P, et al. Nursing homes as a reservoir of extended-spectrum beta-lactamase (ESBL)-producing ciprofloxacin-resistant Escherichia coli. J Antimicrob Chemother . 2009 Sep;64(3):635–41.

206. Sundvall P-D, Elm M, Gunnarsson R, Mölstad S, Rodhe N, Jonsson L, et al. Antimicrobial resistance in urinary pathogens among Swedish nursing home residents remains low: a cross-sectional study comparing antimicrobial resistance from 2003 to 2012. BMC Geriatr . 2014 Jan;14:30.

PhD Dissertation

9. Appendix

Appendix 1. Patient demographics. Community-onset infection (COI) and Healthcare associated infections (HCAI) have been divided into age groups and patients have been classified a complicated UTI. These classifications have been done according to previously defined criteria as stated in Manuscript II. Prescr. = Prescriptions.

Cases

ESBL n=96 Resistant n=171 Susceptible n=175 Controls n=200

Demographics n(%) n(%) n(%) n(%)

Age (Mean) 60.5 96(100) 57.95 171(100) 53,25 175(100) 42.10 200(100)

Age>18 (Mean) 65.4 88(92) 58,5 169(99) 56,07 165(94) 46.16 180(90)

Age >65 (Mean) 76.5 48(50) 79.09 85(50) 79.72 66(38) 76.49 37(19)

Male 18 (19)

17(10) 16(9)

54(27)

Female 78(81) 154(90) 159(91) 146(73)

Community-Onset-Infection (COI) 71(74)

139(81) 153(87)

163(82)

Health-Care Associated-Infection (HCAI) 25(26) 32(19) 22(13) 37(19)

COI Age < 18 years 8(100)

1(50) 10(100)

18(90)

HCAI Age < 18 years 0 1(50) 0 2(10)

COI Age 18-65 30(75) 0.5661 77(92) 0.5750 90(91) 0.7870 119(83)

HCAI Age 18-65 10(25) 0.0553 7(8) 0.0624 9(9) 0.3759 24(17)

COI Age >65 years 33(69) 61(72) 53(80)

26(70)

HCAI Age >65 years 15(31) 24(28) 0.4049 13(20) 1 11(30)

Complicated UTI 1(1) 1(0.6) 0

Complicated UTI due to Age > 65 years 44(46) 86(50) 66(38)

Appendix 1 cont. Exposure to antibiotics within one year prior to index sample.

ESBL n=96 Resistant n=171 Susceptible n=175 Controls n=200

Exposure to Antibiotics N (%) Prescr. N (%) Prescr. N (%) Prescr. N (%) Prescr.

Antibiotics in general 85 (89) 480 155 (91) 733 153 (87) 457

131 (66)

Penicillins 25 (26) 40 40 (23) 52 41 (23) 48 42 (21) 59

Trimethoprim 12 (13) 43 22 (13) 93 8 (5) 8 6 (3) 7

Sulphonamides 27 (28) 46 67 (39) 110 50 (29) 78 42 (21) 53

Metronidazole 7 (7) 11 11 (6) 13 8 (5) 8 7 (4) 8

Quinolones 21 (22) 31 22 (23) 36 14 (8) 18 18 (9) 23

Macrolides 13 (14) 29 23 (13) 31 23 (13) 25 27 (14) 43

Mecillinam 74 (77) 149 118 (69) 233 104 (59) 193 60 (30) 97

Nitrofurantoin 19 (20) 53 35 (20) 104 19 (11) 29 11 (6) 18

Tetracycline 5 (5) 8 9 (5) 21 6 (3) 9 6 (3) 9

2nd generations cephalosporins 2 (2) 3 2 (1) 6 0 0 2 (1) 3

Penicillins w. beta-lactamase inhibitors 8 (8) 11 7 (4) 8 5 (3) 5 5 (3) 11

Penicillinase stable penicillins 10 (10) 13 9 (5) 19 6 (3) 6 12 (6) 14

Penicillins with effect on Gram-negative 69 193 187 267 125 228 71 (36) 127

Copenhagen 2014

83

Appendix 2. Results of the Multivariate Logistic Regression Analysis comparing case groups with the uninfected group. We performed the multivariate analysis based on the initial student´s t-test, with all variables with P<0.1. Subsequently we did a simple manual backward step analysis including variables with P<0.1 in the multivariate analysis. The second multivariate analysis was not possible to perform in full for the ESBL group due to lack of goodness of fit. Therefore we have included results from 30 days after sampling. *P-values found by chi-square analysis. Empty squares means estimates not computed by SAS. OD = Odds Ratio.

Multivariate Logistic Regression. First run.

ESBL, n=96 Resistant, n=171 Susceptible, n=175

Variables OR 95 CI P-Value OR 95 CI P-Value OR 95 CI P-Value

Sex (Females) 3.1 1.4 6.6 0.0044 3.7 1.8 7.7 0.0004 3.5 1.807 6.9 0.0002

Age 1.02 1.01 1.04 <.0001 1.0 1.0 1.0 <.0001 1.02 1.010 1.0 <.0001

30 Days Prior

Antibiotics . . . <.0001* . . . <.0001* . . . <.0001*

Mecillinam 2.6 0.9 7.2 0.0713 4.1 1.5 11.1 0.0053 6.09 1.846 20.1 0.0030

Trimethoprim 2.01 0.19 21.1 0.5621 4.1 0.9 19.4 0.0712 1.6 0.186 12.9 0.6850

Kidney Infection Infty Infty Infty 0.0106* 1.1 0.3 4.1 0.8366 . . . 1

Urosepsis Infty 0.99 10.7 0.0334* . . . . . . . .

90 Days Prior

Mecillinam 1.8 0.98 3.3 0.0572 2.4 1.1 5.4 0.0261 2.3 0.910 5.8 0.0781

Nitrofurantoin 1.6 0.5 5.1 0.3871 3.8 1.3 11.4 0.0182 3.2 0.975 10.4 0.0552

Penicillin 2.7 0.8 8.7 0.1040 5.0 1.7 15.1 0.0041 2.6 0.848 8.2 0.0940

Sulphonamides 1.03 0.4 2.5 0.9511 2.9 1.3 6.4 0.0091 2.4 1.078 5.2 0.0318

Trimethoprim 0.99 0.2 4.4 0.9848 8.0 1.99 32.1 0.0034 1.9 0.268 13.5 0.5201

Kidney Infection 0.95 0.4 2.5 0.9187 0.5 0.2 1.6 0.2469 0.2 0.052 0.9 0.0387

90 Days After

Mecillinam 9.88 1.9 50.4 0.0059 2.99 0.9 9.6 0.0668 0.8 0.265 2.4 0.6714

Nitrofurantoin 3.2 0.9 11.4 0.0708 4.7 1.5 14.8 0.0076 3.3 1.050 10.4 0.0410

Penicillin (+ effect on Gram-neg.)

0.5 0.1 2.9 0.4727 1.3 0.4 4.2 0.6394 4.5 1.379 14.5 0.0126

Sulphonamides 6.2 1.1 33.8 0.0336 6.4 1.7 24.5 0.0070 7.8 2.284 26.9 0.0011

Trimethoprim . . . 0.0263* 9.7 1.9 50.4 0.0071 4.1 0.726 23.2 0.1100

Appendix 2 cont. Simplified manual backward stepwise elimination procedure including variables with P<0.1

Variables OD 95 CI P-Value

OD 95 CI P-Value OD 95 CI P-Value

30 Days Prior

Mecillinam 3.01 1.8 4.9 <.0001

3.9 2.5 6.4 <.0001 3.3 2.0 5.2 <.0001

Sulphonamides 0.97 0.3 2.4 0.9533

2.6 1.4 4.99 0.0041 2.1 1.1 3.99 0.0201

Trimethoprim 3.33 0.4 27.2 0.2616

4.6 1.2 17.4 0.0249 1.0 0.1 7.2 0.9894

90 Days Prior

Mecillinam 2.30 1.6 3.4 <.0001

3.1 1.8 5.4 <.0001 2.8 1.6 4.9 0.0003

Penicillin 2.78 1.2 6.3 0.0148

3.1 1.2 8.3 0.0225 2.3 0.8 6.4 0.1048

Sulphonamides 0.92 0.4 1.95 0.8234

2.0 1.1 3.9 0.0326 1.9 0.98 3.6 0.0548

30 Days After

Antibiotics 1.52 0.8 2.98 0.1549

2.3 1.4 3.8 0.0017 . . . <.0001*

PhD Dissertation

Kidney Infection 2.62 1.2 5.8 0.0171 . . . 0.0816* 0.8 0.1 6.3 0.8726

Mecillinam 11.5 3.6 36.4 <.0001 7.1 2.6 19.4 0.0001 6.9 2.8 17.0 <.0001

Nitrofurantoin 34.8 3.9 312.6 0.0015 8.0 0.9 68.0 0.0567 9.9 1.3 77.8 0.0291

Sulphonamides 8.9 0.8 98.1 0.0729 6.2 0.7 55.0 0.1036 20.7 2.7 158.7 0.0036

90 Days After

Mecillinam 2.9 0.9 9.1 0.0574 2.5 1.5 4.1 0.0006

Nitrofurantoin 3.6 1.5 8.4 0.0030 2.8 1.4 5.4 0.0954

Penicillin (+ effect on Gram-neg)

1.4 0.5 3.8 0.5569 6.5 1.96 21.5 0.0027

Sulphonamides 5.5 1.6 18.4 0.0055 7.1 2.4 21.1 0.0022

Copenhagen 2014

85

Appendix 3. . Results of multivariate logistic regression analyses comparing the ESBL group to other case groups. These tests were performed as previously described in Table 6. *P-values found by chi-square analysis. OR = Odds Ratio.

Multivariate Logistic Regression. First run.

ESBL vs. Resistant ESBL vs. Susceptible

Variables OR 95 CI P-Value OR 95 CI P-Value

COI vs. HCAI 1.5 0.8 2.8 0.1660* 2.4 1.3 4.6 0.0058*

Sex 0.7 0.3 1.7 0.4335 0.7 0.3 1.7 0.4606

Age 1.0 0.99 1.02 0.4387 1.0 0.99 1.02 0.1837

30 Days Prior

Admissions 4.95 1.5 15.9 0.0073 3.9 1.2 12.7 0.0216

Sulphonamides 0.3 0.1 0.9 0.0381 0.3 0.08 0.9 0.0299

90 Days Prior

Antibiotics 2.0 1.2 3.3 0.0049 2.5 1.4 4.7 0.0029

Insulin 1.3 0.8 2.4 0.3218 2.5 1.0 6.1 0.0447

Nitrofurantoin 0.4 0.2 0.8 0.0089 0.6 0.3 1.3 0.1840

Sulphonamides 0.3 0.1 0.6 0.0010 0.3 0.1 0.8 0.0091

Trimethoprim 0.3 0.1 0.8 0.0096 0.6 0.2 2.2 0.4663

30 Days After

Alimentary tract and metabolism 0.8 0.5 1.2 0.2260 0.6 0.4 0.96 0.0344

Antibiotics 1.1 0.6 1.9 0.7451 1.9 1.0 3.4 0.0470

Mecillinam 1.7 0.4 6.7 0.4336 4.03 0.99 16.4 0.0515

90 Days After

Genitourinary and sex hormones 0.2 0.04 0.8 0.0225 0.2 0.03 0.8 0.0245

Mecillinam 1.6 0.7 4.5 0.3806 6.9 2.04 23.5 0.0019

Penicillin (with effect on Gram-negative) 0.9 0.3 2.8 0.9081 0.2 0.075 0.9 0.0414

Penicillin in combination w. beta-lactamase inhibitors

5.3 1.2 22.2 0.0240 1.2 0.33 4.3 0.7795

Appendix 3 cont. Simplified manual backward stepwise elimination procedure including variables with P<0.1

30 Days Prior Variables OR 95 CI P-Value OR 95 CI P-Value

Trimethoprim 0.3 0.1 0.99 0.0486 1.3 0.1 11.7 0.8100

Sulphonamides 0.4 0.2 0.99 0.0486 0.3 0.1 0.7 0.0088

Quinolones 3.4 1.08 10.6 0.0365 2.08 0.6 7.3 0.2489

Antibiotics 0.98 0.6 1.5 0.9194 1.8 1.02 3.1 0.0414

Admissions 4.1 1.35 12.1 0.0124 3.4 1.1 10.5 0.0295

90 Days Prior

Antibiotics 1.2 0.97 1.4 0.1044 1.4 1.1 1.8 0.0024

Sulphonamides 0.6 0.3 0.95 0.0316 0.6 0.4 1.05 0.0723

90 Days After

Mecillinam 1.4 0.5 3.9 0.4624 4.1 1.4 11.8 0.0096 Penicillin in combination w. beta-

lactamase inhibitors 4.4 1.1 17.7 0.0362 1.767 0.8 4.0 0.1734

PhD Dissertation

Appendix 4. Distribution of sequence type (STs) in relation to assigned a-MLVA codes and serogroups among ESBL-producing E.coli. We show the serogroups found for the given a-MLVA-code. M = Multiple serogroups, N = serogroup negative. Isolates with a written ST are the isolates tested by MLST.

ST a-MLVA Serogroup Phylogroup ESBL Genotype

ESBL 14 123645 75 B2 TEM-1

ESBL 224 131271 M B1 CTX-M-15

ESBL 2852 131291 8 B1 CTX-M-15

ESBL 131291 25 B2 CTX-M-15

ESBL 120 132271 N Non-Type CTX-M-15

ESBL 131 103562 16 B2 CTX-M-14

ESBL 131 153562 25 B2 CTX-M Gr. 1

ESBL 131 153562 25 B2 CTX-M-15

ESBL 131 153562 77 B2 CTX-M-15

ESBL Unsuccessful 153562 97 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-28

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-14

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-28

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-28

ESBL 153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M 79/55

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

Copenhagen 2014

87

ESBL 153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL 153562 M B2 CTX-M-27

ESBL 153562 M B2 CTX-M-15

ESBL 153562 M B2 CTX-M-15

ESBL 153562 M B2 CTX-M-15

ESBL 153562 MISSING B2 CTX-M-28

ESBL 153562 MISSING B2 CTX-M-27

ESBL 153562 N B2 CTX-M-28

ESBL 131 163562 16 B2 CTX-M-15

ESBL 131 163562 16 B2 CTX-M-15

ESBL 163562 16 B2 CTX-M-15

ESBL 163562 16 B2 CTX-M-1

ESBL 163562 153 B2 CTX-M-15

ESBL 648 161160 1 D CTX-M-15

ESBL 648 161160 102,130 D CTX-M-15

ESBL 161160 153 D CTX-M-15

ESBL 161160 MISSING D CTX-M-15

ESBL 62 161370 MISSING D CTX-M-14

ESBL 69 173277 15 D CTX-M Gr. 1

ESBL 173277 15 D CTX-M Gr. 1

ESBL 173277 15 D CTX-M-79/55

ESBL 173277 15 D CTX-M-27

ESBL 173277 44 D TEM-1

ESBL 173277 73 D CTX-M-14

ESBL 315 173552 25 B2 CTX-M-14

ESBL 428 252365 117 B2 CTX-M-1

ESBL 354 151170 153 D CTX-M-14

ESBL 998 224665 2 B2 CTX-M-15

ESBL 998 224665 M B2 CTX-M-1

ESBL 88 132261 119 A CTX-M-1

ESBL 636 293852 21 B2 CTX-M-15

ESBL 101 131261 N B1 CTX-M-1

ESBL 448 131261 111 B1 CTX-M-15

ESBL 131261 170 NT CTX-M Gr. 9

ESBL 131261 81 B1 CTX-M-79/55

ESBL 131261 100 NT CTX-M-1

ESBL 131261 M B1 CTX-M-15

PhD Dissertation

ESBL 617 132251 12 A CTX-M-15

ESBL 132251 69 A CTX-M-28/15

ESBL 132251 N A CTX-M-15

ESBL 132251 116,162 A CTX-M-15

ESBL 132251 N A CTX-M-79/55

ESBL 132251 N A CTX-M-15

ESBL 132251 12 A CTX-M-15

ESBL 746 131251 21 A CTX-M-28

ESBL 1598 131251 9 NT CTX-M-15

ESBL 38 173050 2 D CTX-M-14

ESBL 38 173050 M D CTX-M-14

ESBL 38 173050 M D CTX-M-14

ESBL 38 173050 153 D CTX-M-14

ESBL 173050 M D CTX-M-14

ESBL 173050 M D CTX-M-14

ESBL 173050 161 D CTX-M-14

ESBL 173050 86 D CTX-M-14

ESBL 173050 153 D CTX-M-14

ESBL 173050 153 D CTX-M-14

Copenhagen 2014

89

Appendix 5. Distribution of sequence type (STs) in relation to assigned a-MLVA-codes and serogroups, for the resistant group. Not all resistant isolates were characterized by serogroup. Here we show the serogroups found for the given a-MLVA-code. M = Multiple serogroups, N = serogroup negative. Isolates with a written ST are the isolates tested by MLST.

ST a-MLVA Serogroup Phylogroup

Resistant 405 121250 M D

Resistant 121250 D

Resistant 405 131250 D

Resistant 131250 D

Resistant 14 123645 75 B2

Resistant 362 143250 D

Resistant 131 143562 B2

Resistant 131 153562 25 B2

Resistant 131 153562 B2

Resistant 153562 25 B2

Resistant 153562 25 B2

Resistant 153562 25 B2

Resistant 153562 B2

Resistant 153562 B2

Resistant 153562 B2

Resistant 153562 B2

Resistant 153562 B2

Resistant 131 163562 B2

Resistant 163562 B2

Resistant 163562 B2

Resistant 648 161150 1 D

Resistant 62 161370 D

Resistant 457 161371 11 D

Resistant 1597 162562 39 B2

Resistant 1597 162562 B2

Resistant 1193 124645 75 B2

Resistant 1193 124645 6 B2

Resistant 124645 B2

Resistant 124645 D

Resistant 124645 B2

Resistant 124645 75 B2

Resistant 393 171577 D

Resistant 69 173270 73 D

Resistant 173270 D

Resistant 69 173277 15 D

Resistant 69 173277 25 D

PhD Dissertation

Resistant 173277 11 D

Resistant 173277 11 D

Resistant 173277 15 D

Resistant 173277 15 D

Resistant 173277 25 D

Resistant 173277 68 D

Resistant 173277 73 D

Resistant 173277 73 D

Resistant 173277 73 D

Resistant 173277 73 D

Resistant 173277 M D

Resistant 173277 D

Resistant 173277 D

Resistant 95 223643 2 B2

Resistant 95 223653 2 Non-Type

Resistant 223653 Non-Type

Resistant 372 224563 18 B2

Resistant 978 226565 83 B2

Resistant 80 254575 75 B2

Resistant 80 254575 75 B2

Resistant 88 132261 8 NT

Resistant 132261 NT

Resistant 127 264953 6 B2

Resistant 117 173050 M D

Resistant 1177 173050 D

Resistant 135 225661 B2

Resistant 141 124665 B2

Resistant 73 176655 Non-Type

Resistant 73 266655 6 B2

Resistant 73 276655 6 B2

Resistant 73 276655 25 B2

Resistant 73 276655 B2

Resistant 276655 M B2

Resistant 276655 B2

Resistant 276655 B2

Resistant 276655 B2

Resistant 276655 B2

Resistant 10 124643 75 B2

Resistant 124643 75 B2

Resistant 10 132251 A

Resistant 132251 3 A

Copenhagen 2014

91

Resistant 132251 A

Resistant 132251 A

Resistant 132251 A

Resistant 132251 N A

Resistant 2279 132251 15 A

Resistant 38 173051 25 D

Resistant 58 131261 19 B1

Resistant 131261 B1

Resistant 131261 NT

Resistant 131261 N NT

Resistant 131261 69 B1

Resistant 131261 NT

Resistant New ST 123552 B2

Resistant New ST 131251 NT

Resistant New ST 132261 NT

Resistant New ST 162438 NT

Resistant MISSING 266561 NT

PhD Dissertation

Appendix 6. Distribution of sequence type (STs) in relation to assigned a-MLVA-codes and serogroups for the susceptible group. Not all susceptible isolates were characterized by serogroup. Here we show the serogroups found for the given a-MLVA-code. M = Multiple serogroups, N = serogroup negative. Isolates with a written ST are the isolates tested by MLST.

ST a-MLVA Serogroup Phylogroup

Susceptible 223 101261 B2

Susceptible 101261 Non-Type

Susceptible 101261 B2

Susceptible 69 103277 B1

Susceptible 69 173277 D

Susceptible 173277 D

Susceptible 173277 D

Susceptible New ST 112871 D

Susceptible 1161 124675 B2

Susceptible 405 131250 2 D

Susceptible 48 131281 Non-Type

Susceptible 131281 119 Non-Type

Susceptible 357 150365 B2

Susceptible 357 152365 73 B2

Susceptible 152365 M B2

Susceptible 538 152363 13 B2

Susceptible 131 153562 B2

Susceptible 62 161370 D

Susceptible 501 163777 D

Susceptible New ST 171250 D

Susceptible 95 203653 B2

Susceptible 95 223653 1 B2

Susceptible 223653 1 B2

Susceptible 223653 M B2

Susceptible 223653 B2

Susceptible 223653 Non-Type

Susceptible 73 176655 18 B2

Susceptible 73 203655 2 D

Susceptible 73 206653 B2

Susceptible 73 206655 B2

Susceptible 73 226655 B2

Susceptible 73 256653 D

Susceptible 73 266655 2 B2

Susceptible 266655 B2

Susceptible 73 276555 25 B2

Susceptible 73 276655 6 B2

Copenhagen 2014

93

Susceptible 73 276655 22 B2

Susceptible 73 276655 M B2

Susceptible 276655 2 B2

Susceptible 276655 B2

Susceptible 73 276665 120 B2

Susceptible 162 211251 95 A

Susceptible 681 224452 8 D

Susceptible 1858 224645 6 B2

Susceptible 4235 224743 88 B2

Susceptible 224743 88 B2

Susceptible 3672 226962 B2

Susceptible New ST 232251 105 A

Susceptible 59 241370 D

Susceptible 59 251380 D

Susceptible 80 254575 B2

Susceptible 127 264953 B2

Susceptible 264953 6 B2

Susceptible 127 274953 6 B2

Susceptible 12 266562 4 B2

Susceptible 12 266562 B2

Susceptible New ST 272275 133 B2

Susceptible 1444 273673 4 B2

Susceptible 582 274242 B2

Susceptible 1331 274655 6 D

Susceptible 589 287572 B2

Susceptible 714 473050 D

Susceptible 3846 254665 B2

Susceptible 420 254665 82 B2

Susceptible 10 131251 109 A

Susceptible 10 131251 M A

Susceptible 131251 107,117 A

Susceptible 10 132251 81 A

Manuscript I to III

  Published Ahead of Print 4 August 2014. 10.1128/AAC.03021-14.

2014, 58(10):6139. DOI:Antimicrob. Agents Chemother. Frimodt-MøllerFrederik Boetius Hertz, Anders Løbner-Olesen and Niels Colonization ModelSequence Type 131 in a Mouse Intestinal Antibiotic Selection of Escherichia coli

http://aac.asm.org/content/58/10/6139Updated information and services can be found at:

These include:

REFERENCEShttp://aac.asm.org/content/58/10/6139#ref-list-1at:

This article cites 31 articles, 16 of which can be accessed free

CONTENT ALERTS more»articles cite this article),

Receive: RSS Feeds, eTOCs, free email alerts (when new

http://journals.asm.org/site/misc/reprints.xhtmlInformation about commercial reprint orders: http://journals.asm.org/site/subscriptions/To subscribe to to another ASM Journal go to:

on Septem

ber 27, 2014 by guesthttp://aac.asm

.org/D

ownloaded from

on S

eptember 27, 2014 by guest

http://aac.asm.org/

Dow

nloaded from

Antibiotic Selection of Escherichia coli Sequence Type 131 in a MouseIntestinal Colonization Model

Frederik Boetius Hertz,a,b,c Anders Løbner-Olesen,b Niels Frimodt-Møllera

Department of Clinical Microbiology, Hvidovre University Hospital, Hvidovre, Denmarka; Department of Biology, University of Copenhagen, Copenhagen, Denmarkb;Statens Serum Institut, Copenhagen, Denmarkc

The ability of different antibiotics to select for extended-spectrum �-lactamase (ESBL)-producing Escherichia coli remains atopic of discussion. In a mouse intestinal colonization model, we evaluated the selective abilities of nine common antimicrobials(cefotaxime, cefuroxime, dicloxacillin, clindamycin, penicillin, ampicillin, meropenem, ciprofloxacin, and amdinocillin) againsta CTX-M-15-producing E. coli sequence type 131 (ST131) isolate with a fluoroquinolone resistance phenotype. Mice (8 pergroup) were orogastrically administered 0.25 ml saline with 108 CFU/ml E. coli ST131. On that same day, antibiotic treatmentwas initiated and given subcutaneously once a day for three consecutive days. CFU of E. coli ST131, Bacteroides, and Gram-posi-tive aerobic bacteria in fecal samples were studied, with intervals, until day 8. Bacteroides was used as an indicator organism forimpact on the Gram-negative anaerobic population. For three antibiotics, prolonged colonization was investigated with addi-tional fecal CFU counts determined on days 10 and 14 (cefotaxime, dicloxacillin, and clindamycin). Three antibiotics (cefo-taxime, dicloxacillin, and clindamycin) promoted overgrowth of E. coli ST131 (P < 0.05). Of these, only clindamycin suppressedBacteroides, while the remaining two antibiotics had no negative impact on Bacteroides or Gram-positive organisms. Only clin-damycin treatment resulted in prolonged colonization. The remaining six antibiotics, including ciprofloxacin, did not promoteovergrowth of E. coli ST131 (P > 0.95), nor did they suppress Bacteroides or Gram-positive organisms. The results showed thatantimicrobials both with and without an impact on Gram-negative anaerobes can select for ESBL-producing E. coli, indicatingthat not only Gram-negative anaerobes have a role in upholding colonization resistance. Other, so-far-unknown bacterial popu-lations must be of importance for preventing colonization by incoming E. coli.

Escherichia coli is a versatile and ubiquitous species that is regu-larly represented in the commensal flora of the gut. The species

includes nonpathogenic, intestinal pathogenic, and extraintesti-nal pathogenic E. coli (ExPEC), all of which can be variably presentin the human gut. ExPEC can cause a wide range of infections,from uncomplicated cystitis to life-threatening sepsis (1–3). Ma-jor sources for resistance in E. coli are plasmid-borne extended-spectrum �-lactamases (ESBL), which are enzymes capable of hy-drolyzing, and thus conferring resistance toward, most �-lactamantibiotics, except for the cephamycins and carbapenems. In ad-dition, ESBLs are inhibited by �-lactamase inhibitors, such as cla-vulanic acid, sulbactam, and tazobactam (1, 4). Plasmids carryingESBL genes often carry various other genes that cause resistance toother classes of antibiotics (e.g., aminoglycosides) (1, 4, 5). One ofthe most common types of ESBLs identified in the world is CTX-M-15, and the spread of its areas of endemicity seems to be asso-ciated with a few E. coli sequence types (ST), such as ST131. ST131is most frequently linked to quinolone resistance and CTX-M-15,but it also harbors specific virulence genes coding for factors suchas adhesins (fimH, papEF), toxins (sat), and capsules (kpsM II),contributing to its ability to colonize the human gut (2, 4–7). Thepossible human colonization is of great concern, since ST131 iso-lates often are resistant to several antibiotics and are known tocause urosepsis to a higher degree than non-ST131 isolates. Inclinical settings, multiresistance, including production of ESBL,delays appropriate treatment, leading to extended hospital stays aswell as increased mortality and morbidity (8–10). Finally, urinarytract infections are primarily caused by E. coli present in the pa-tient’s own microbiota, making knowledge on colonization byresistant E. coli of great importance (1, 4, 11).

Several case-control studies have identified recent antibiotic

exposure, especially to cephalosporins and fluoroquinolones, andhospitalization as significant risk factors for acquiring an infectionwith an ESBL-producing Enterobacteriaceae (within 30 days) (10,12–14). A systematic study of the selective ability of all antibioticclasses has been lacking. In this study, we wished to evaluate theability of nine common antimicrobials, including antibiotics usedfor Gram-positive infections, to select for a CTX-M-15-producingE. coli ST131 isolate in a mouse intestinal colonization model.

(This work was presented as a poster at the 52nd InterscienceConference on Antimicrobial Agents and Chemotherapy, SanFrancisco, CA, 9 to 12 September 2012.)

MATERIALS AND METHODSStrain. For the colonizing pathogen, we used a clinical blood isolate of E.coli that belongs to the lineage B2-O25b-ST131. This isolate (65-Ec-09)carries some of the virulence factors previously seen in ST131 isolatesfound throughout Denmark in 2009, and it is similarly resistant towardmany commonly used antibiotics (MICs when resistant: cefotaxime, �32�g/ml; cefuroxime, �256 �g/ml; ceftazidime, 32 �g/ml; ampicillin, �256�g/ml; aztreonam, 32 �g/ml; amoxicillin-clavulanate, �256 �g/ml; cip-rofloxacin, �32 �g/ml; cloxacillin, �256 �g/ml; clindamycin, �256 �g/ml. MICs when susceptible are as follows: amdinocillin, 2 �g/ml; trim-ethoprim-sulfamethoxazole, �0.125 �g/ml; gentamicin, �0.5 �g/ml;

Received 8 April 2014 Returned for modification 24 May 2014Accepted 31 July 2014

Published ahead of print 4 August 2014

Address correspondence to Frederik Boetius Hertz, [email protected].

Copyright © 2014, American Society for Microbiology. All Rights Reserved.

doi:10.1128/AAC.03021-14

October 2014 Volume 58 Number 10 Antimicrobial Agents and Chemotherapy p. 6139 – 6144 aac.asm.org 6139

on Septem

ber 27, 2014 by guesthttp://aac.asm

.org/D

ownloaded from

piperacillin-tazobactam, 2 �g/ml; meropenem, �0.064 �g/ml; nitrofu-rantoin, 13 millimeters in zone diameter [mm]; trimethoprim, 27 mm)(15). Pheno- and genotypic characterizations were performed by usingthe MAST-test, PCR, and DNA sequence analysis according to methodsused at the clinical laboratory of Department of Clinical Microbiology,Hvidovre Hospital, Denmark (HVH) or at Statens Serum Institut (SSI) aspreviously described (15). Sequence type verification was performed viafull multilocus sequence typing (MLST, using the Achtmann scheme[http://mlst.warwick.ac.uk/mlst/dbs/Ecoli]) by SSI, who also found thatthe isolate harbored the virulence factors kpsM II and iutA (15–17).

Media. To test bacterial growth in collected fecal samples from mice,we used selective agar plates, all from SSI Diagnostica, Hilleroed, Den-mark. For the CTX-M-15-producing E. coli population, ID Flexicult agarcontaining cefotaxime at 32 mg/liter and vancomycin at 6 mg/liter wereused. The Gram-positive aerobic population was selected on 5% bloodagar plates containing gentamicin at 5 mg/liter, and the Gram-negativeanaerobic population was selected on anaerobic plates containing genta-micin at 32 mg/liter and vancomycin at 16 mg/liter. Culturing of anaero-bic species were performed under anaerobic conditions in GasPak EZcontainers and an anaerobic atmosphere created by using AnaeroGen(Oxoid) (18).

MIC determinations. Antibiotic susceptibility testing was performedas Etests when possible and as disc diffusion tests where no Etest wasavailable (the MIC for dicloxacillin was found by using an Etest for clox-acillin, since neither Etest nor disks for dicloxacillin were available). Thediffusion test methodology has been described elsewhere (15), and a sim-ilar Etest methodology was used according to guidelines from the Depart-ment of Clinical Microbiology, Hvidovre Hospital. All results were inter-preted according to current recommendations from the EuropeanCommittee on Antimicrobial Susceptibility Testing (http://www.eucast.org/clinical_breakpoints/).

Antibiotics used for treatment. Commonly used antibiotics werechosen for this study. These included different �-lactam antibiotics, cip-rofloxacin, and clindamycin. All mouse dosages were calculated based onhuman doses (in mg per kg of body weight) from pharmacokinetic (PK)studies performed at SSI or from previously published mouse studies(Table 1) (19–26). Doses were chosen to mimic the serum antibiotic con-centrations achieved in humans on standard doses. All antibiotics wereadministered subcutaneously once each day for 3 consecutive days. Con-centrations of antibiotics in mouse feces had been measured in a previ-ously published study for a �-lactam antibiotic, an expanded-spectrumcephalosporin, a carbapenem, clindamycin, and ciprofloxacin (19). Thedoses used in this study were similar or higher.

Mouse intestinal colonization model. (i) Mice. The animal experi-ment was approved by the Danish Centre for Animal Welfare and carried

out at Statens Serum Institut in Copenhagen, Denmark. In all studies, 7-to 10-week-old female albino, outbred NMRI mice (Harlan, the Nether-lands) weighing 26 to 30 g were used. The mice used in each study were allfrom the same litter and were brought simultaneously to the stable andhoused in pairs of two per cage. At the end of the study, all mice weresacrificed to ensure that no mice were kept alive with no immediate pur-pose. Animals were housed, treated, and sacrificed according to currentguidelines.

(ii) The mouse model. The mouse intestinal colonization model wasan experimental model where all mice were kept in pairs of two per cage.Two cages constituted one group and each group received one antibiotic.Thus, each antibiotic was given to a total of four mice in two differentcages, and a total of 20 to 22 cages were included in the study. Treatmentwas given subcutaneously in the neck once a day for three consecutivedays (day 1 to day 3). Inoculation of mice with the bacterial strain wasdone through a stainless steel orogastric feeding tube on day 1 prior toinitiation of treatment. The intestinal flora was unaltered prior to thestudy, and no mice were anesthetized during the study. The experimentwas conducted from day 1 to day 8, and cages were changed daily. At theend of day 8 all mice were sacrificed.

(iii) Experimental study. We executed our full experimental study byusing the described mouse intestinal colonization model. The study wasconducted from day 1 to day 8 with feces collected prior to inoculation onday 1 and on days 2, 4, and 8. On day 1, mice were inoculated once with0.25 ml of saline containing 108 CFU/ml of 65-Ec-09. Thus, each mousewas given an inoculum of 2.5 � 107 CFU.

The mice were left for 3 h before the first doses of antibiotics wereadministered subcutaneously. Each treatment group, consisting of fourmice housed in two different cages, received cefotaxime, cefuroxime, am-picillin, dicloxacillin, amdinocillin, meropenem, clindamycin, cipro-floxacin, or benzylpenicillin (Table 1). A control group received 65-Ec-09but no antibiotic treatment. The complete selection study of E. coli ST131CTX-M-15 (65-Ec-09) in the mouse intestinal colonization model wasperformed twice, with the exception of CFU counts of the Gram-positiveaerobic flora, which were only studied in the second run. The controlgroup receiving 65-Ec-09 only was included in both runs. Furthermore, agroup of mice that received treatment with cefotaxime without receivingthe ESBL-producing strain was included in the first run only.

(iv) Prolonged presence of resistant pathogen after completed anti-biotic treatment. Additionally, the prolonged presence of the E. coliST131 after completed antibiotic treatment was studied in the first run forthree antibiotics (cefotaxime, dicloxacillin, and clindamycin). These threeantibiotics were chosen based on their selective abilities found in thestudy. Treatment stopped, for all groups, on day 3. Cages were changed

TABLE 1 Antibiotics used as treatmentsa

Antibiotic

Humanb Mouse

Dose (g, i.v.) Cmax (�g/ml)Needed dose(mg/kg) Cmax (�g/ml)

Dose given(mg/mouse/day)

Cefuroxime 1.5 65 120 50–60 4Cefotaxime 1 40 60 100 2Ampicillin 1 40 50 75 1.5Dicloxacillin 1 30–40 60 90 2Benzylpenicillin 2 mill. IEb 60 70 60 2Amdinocillin 0.4 30–40 60 30 2Meropenem 0.5 26 50 50 1.5Clindamycin 1.8 6 36 8 1.4Ciprofloxacin 0.4 4 15 2 0.5a All doses were administered subcutaneously once a day, and the needed doses were calculated based on the expected average weight of the mice (weights given by provider), asdescribed in previously published studies (18–25). All mouse dosages were calculated based on human doses from PK studies performed at SSI or from previously published mousestudies (19–26). i.v., intravenous.b Benzylpenicillin (1.2 grams � 2 millions units � 2 mill. IE) was administered intravenously.

Boetius Hertz et al.

6140 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 27, 2014 by guesthttp://aac.asm

.org/D

ownloaded from

daily from days 1 to 14, and feces samples were collected on days 10 and14. On day 14, the study was terminated and mice were killed.

Detection and quantification of bacteria in feces. On specified days,0.5 g of feces was collected from each cage. Feces were dissolved in 5 ml ofsaline and further diluted 10-fold in saline for a total of 6 times. Dilutionswere plated on the different selective agar plates, and the log CFU per 0.5g of stool for 65-Ec-09, the Gram-negative anaerobic flora, and the Gram-positive aerobic flora were calculated for each cage and two or three col-onies from each day frozen. Each CFU count was performed on two agarplates and calculated as the average CFU count for the two plates. Toensure no presence of cefotaxime-resistant E. coli prior to inoculation,dilutions of feces from day 1, from each cage, were spotted on selectiveplates and this showed no growth of resistant E. coli. The lower detectionlimit was 10 CFU per 0.5 g of feces.

Molecular tests. To ensure that the E. coli found in feces was identicalto the isolate given through inoculation, we tested a total of 17 cefotaxime-resistant E. coli isolates found in feces during treatment and 4 E. coliisolates from day 1 for the presence of a CTX-M group 1 gene. Samplesfrom both experimental runs were included (see below). Three to fourisolates per frozen sample were used for DNA purification. As these testswere performed on isolates from both study runs, they were not per-formed until both runs had been completed. Thus, isolates tested wereisolated from frozen samples. We were therefore likely to isolate the dom-inating E. coli strain and thereby determine if 65-Ec-09 had become thedominating E. coli of the microbiota (11, 27).

All of these 21 isolates were identified as E. coli by matrix-assisted laserdesorption–time of flight analysis as described elsewhere (28). All resis-tant E. coli isolates contained a CTX-M group 1 gene, whereas none of theisolates from day 1 contained a CTX-M group 1 gene.

Isolates from the following groups were tested: cefotaxime (day 2 fromboth study runs and day 1 from the first run), cefuroxime (day 2 fromboth study runs and day 1 from the first run), ampicillin (day 2 from bothstudy runs), dicloxacillin (days 1, 2, 4, and 8 from the first run and days 2and 8 from the second run), clindamycin (days 1, 2, 4, and 10 from the firstone and days 2 and 8 from the second run), and benzylpenicillin (day 2from second run).

Additionally, five of the cefotaxime-resistant E. coli isolates and oneisolate from day 1 were characterized by MLST (cefotaxime, cefuroxime[day 2], dicloxacillin [days 1 and 4], and clindamycin [days 4 and 10]).The five resistant E. coli were identified as ST131 and the one isolate fromday 1 belonged to ST602.

Statistical analysis. Data were analyzed with the use of SAS software,version 9.3 (SAS Institute). When just one group receiving one antibioticwas compared to the control group, this was done via a one-way analysisof variance (ANOVA). When multiple CFU counts were compared, it wasdone as a multiple variant analysis, adjusted for multiple comparisonswith the Bonferroni correction. The conservative Bonferroni correction

was used to avoid multiple comparisons. A P value of �0.05 was consid-ered significant.

RESULTS

Results of fecal bacteriology were calculated as the log CFU/0.5 gof feces, and the mean CFU of four cages (two cages for Gram-positive aerobic flora) were used for statistical calculations. Forgraphic depictions, means and standard deviations (SD) wereused. Data for the selective abilities of the different antibiotics on65-Ec-09 are shown in Table 2 and Fig. 1 and 2 for E. coli, Gram-negative anaerobic flora, and Gram-positive flora, respectively.

Effect of antibiotic treatment on establishment of resistantpathogens and on the indigenous microflora. Data for the effectson colonization of 65-Ec-09 by the different antibiotics are shownin Fig. 1. Cefotaxime, dicloxacillin, and clindamycin promotedthe colonization and overgrowth of 65-Ec-09 from day 2 throughday 8 (P � 0.01 for dicloxacillin and clindamycin; P � 0.05 forcefotaxime). Benzylpenicillin and cefuroxime showed less over-growth but selective abilities on days 2 and 4 (P � 0.05). Dicloxa-cillin and clindamycin showed the highest selective abilities (P �0.01 for dicloxacillin on days 2 and 4 and P � 0.01 for clindamycinon all days). After treatment was completed on day 3, there was adecline in the colonization of 65-Ec-09 from day 4 to 8. In com-parison, neither ampicillin, amdinocillin, meropenem, nor cipro-floxacin promoted overgrowth of 65-Ec-09 beyond day 2 (P �0.05).

Data for the impact on the original microbiota as Gram-nega-tive anaerobic flora, represented by Bacteroides, and Gram-posi-tive flora, respectively, are shown in Fig. 2. None of the antibioticsused had an inhibiting or promoting impact on the Gram-positiveflora (P � 0.05). Only clindamycin had an impact on the Gram-negative anaerobic flora, as it completely eliminated the Gram-negative anaerobic flora during treatment (P � 0.05). After treat-ment, the CFU counts for the Gram-negative anaerobic floraincreased to counts equal to those before treatment.

Prolonged presence of 65-Ec-09 after completed antibiotictreatment. Data for CFU of 65-Ec-09 on all sampling days arefound in Fig. 1. After the initial increase, the CFU decreased for allthree groups of antibiotics. For dicloxacillin and cefotaxime, theCFU counts dropped below the detection limit on day 10. Forclindamycin, the CFU was measurable until day 14, the last day ofthe study. There were no significant differences in fecal CFU of65-Ec-09 over time among the three antibiotics (P � 0.05).

TABLE 2 Results of the CFU counts for E. coli ST131

Antibiotic

Log CFU � SD (P value) ona:

Day 2 Day 4 Day 8 Day 10 Day 14

Cefotaxime 7.75 � 4.33 (�0.01) 5.25 � 6.54 (�0.01) 3.61 � 2.5 (0.011) 1.5 � 1.5 (0.98) 0.5* � 3.55 (1)Clindamycin 8.5 � 1 (�0.01) 7.75 � 0.87 (�0.01) 5.75 � 0.87 (�0.01) 2.5 � 3.55 (0.44) 1* � 1.41 (1)Dicloxacillin 8.5 � 1 (�0.01) 9 � 5.35 (�0.01) 3 � 3.74 (�0.01) 0 (1) 0 (1)Cefuroxime 7.5 � 3 (�0.01) 4.25 � 5.89 (0.015) 1.75 � 4 (0.07)Penicillin 6.25 � 3.57 (0.02) 3.75 � 6.76 (0.03) 0 (1)Amdinocillin 2.25 � 2.96 (0.3) 1.25 � 5.55 (0.5) 0 (1)Meropenem 3.5 � 2.96 (1) 0 (1) 0 (1)Ciprofloxacin 2.25 � 2.6 (0.3) 0 (1) 0 (1)Ampicillin 3.25 � 4.33 (0.8) 2.5 � 6.33 (0.14) 0.75* � 2.6 (0.4)Control 3.5 � 1.73 0 0a P values for days 2 to 8 were determined via an ANOVA, while P values from days 10 and 14 were found by a multiple variant analysis with the Bonferroni correction.*, below thelower detection limit.

Antibiotic Selection of E. coli ST131

October 2014 Volume 58 Number 10 aac.asm.org 6141

on Septem

ber 27, 2014 by guesthttp://aac.asm

.org/D

ownloaded from

DISCUSSION

To our knowledge, this is the first study to experimentally evaluatein vivo selection in the gut of a CTX-M-15-producing E. coli ST131isolate by a range of commonly used antibiotics, including antibi-otics with no activity against Gram-negative bacteria. With themouse intestinal colonization model, we were able to illustrate theselective abilities of different antibiotics on the intestinal florawhen a virulent isolate of CTX-M-15-producing E. coli ST131 wasintroduced. We found several interesting aspects of selection.First, we confirmed that antibiotics with activity against the colo-nizing strain did not promote proliferation, since neither amdi-nocillin nor meropenem resulted in colonization. Second, ourfindings confirmed that an antibiotic, clindamycin, that elimi-nates the Gram-negative anaerobic flora with no in vitro activityagainst Enterobacteriaceae promotes proliferation of ESBL-pro-ducing E. coli. It has been shown that antimicrobial impact on thetotal anaerobic population is proportional to the impact on theBacteroides population (19). Third, of the �-lactams with no in-hibiting effects on the ESBL-producing E. coli, cefotaxime showedthe highest level of selection. Cefuroxime and benzylpenicillin did,however, show higher selection propensities than ampicillin, inagreement with what has previously been seen for high doses ofpenicillin (29). Furthermore, we discovered that dicloxacillin,with no obvious influence on either the Gram-negative anaerobicflora or other Gram-negative bacteria, promoted colonizationwith the ESBL-producing E. coli, while ciprofloxacin, with a lim-ited in vitro effect on the Gram-negative anaerobic bacteria but aneffect on Enterobacteriaceae, showed no abilities to select. Theseresults could indicate that not only Gram-negative anaerobes havea key responsibility upholding colonization resistance. It appearsthat, as for penicillin (29), an antibiotic with limited impact onGram-negative bacteria can select for a resistant E. coli isolate,suggesting that other bacterial populations, not measured in ourstudy, such as certain anaerobic Gram positives, are of importancefor preventing colonization from an incoming E. coli strain.

Finally, we found that the antibiotic with the highest impact on

the anaerobic population, here represented by the Bacteroidespopulation, seems to give room for prolonged colonization, i.e.,clindamycin showed high to medium levels of 65-Ec-09 until day14, compared to dicloxacillin and cefotaxime. This points to thepossibility that even if unknown Gram-positive populations, an-aerobic or aerobic, display colonization resistance, the Bacteroidespopulation seems to play a role in preventing prolonged coloni-zation.

Our study suggests that selection of E. coli ST131 is not derivedalone by the antibiotic impact on the major population, such asanaerobes, as seen with dicloxacillin versus ampicillin and cipro-floxacin. This finding could potentially alter the perception ofwhich antibiotics drive the spread of ESBL-producing E. coli, in-cluding ST131, even if clindamycin was the antibiotic that showedthe highest level of selection and longest duration of colonization.

The model has some limitations, since it does not include clin-ical aspects of selection, such as treatment with multiple antimi-crobials or long-term treatment, nor does it take into accountreexposure to resistant pathogens or retreatment after exposure.Also, we did not investigate the antibiotics’ effects on the totalbacterial population of the intestines, including shifts in the dom-inating phyla. Such an investigation of changes in species andchanges in the total Enterobacteriaceae population could poten-tially have described, in detail, factors influencing colonization.Future studies of the microbiota should be designed to fully de-scribe antibiotic impacts on the different phyla. The lack of selec-tive ability seen from ciprofloxacin on the CTX-M-15-producingE. coli ST131 isolate was not expected and was surprising. We havenot tested the mice used in this study for the presence of cipro-floxacin-resistant strains, and such a presence could explain thelack of selection seen here. Furthermore, extrapolation of resultsfrom the model is limited by the physiological differences betweenmice and humans, even though studies have shown that subcuta-neous treatment once a day in mice gives fecal antibiotic concen-trations equivalent to those of humans (19). Finally, we studiedonly the fecal—and not the mucosal—microflora of mice. Yet, it

FIG 1 The mean and SD of the log CFU/0.5 g of feces for 65-Ec-09 from day 1 to day 14. The arrow indicates treatment from day 1 to day 3.

Boetius Hertz et al.

6142 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 27, 2014 by guesthttp://aac.asm

.org/D

ownloaded from

has been postulated that, first, the mouse fecal flora is a mixture ofmucosal and luminal flora and, second, the intestinal microfloraof laboratory mice is comparable to the intestinal flora in humans(30, 31). We have no obvious explanation for the selective abilityof dicloxacillin, since this drug had no impact on the Gram-neg-ative anaerobic or the aerobic Gram-positive flora. More detailedevaluation of the mouse and human intestinal microbiomes mayprovide the reason for the change in intestinal flora imposed bydicloxacillin. A study of antibiotics’ general impacts on the micro-flora of mice and men would further illustrate the elements in-volved in selection.

In summary, our study confirms that antibiotics with an im-pact on Gram-negative anaerobes support overgrowth and colo-nization of a CTX-M-15 producing E. coli ST131 isolate. None-theless, our study shows that selection could be driven byantibiotics with limited effect on anaerobes and no effect on com-peting Gram negatives, but not by other antimicrobials with abroader spectrum of activity. The results indicate a need for inves-tigation of selective mechanisms of different drugs, to fully de-velop rules and guidelines for stewardship of antibiotics.

ACKNOWLEDGMENTS

This study was performed with financial support from PAR, an EU FP7-Health-2009-Single-Stage project. Additionally, the work was supportedby The Danish Council for Strategic Research (DanCARD project 09-

067075/DSF), Roskilde University, the Aase & Ejnar Danielsens Founda-tion, and the SSAC Foundation.

Furthermore, the work was performed in cooperation between StatensSerum Institut, Hvidovre University Hospital, Roskilde University, andCopenhagen University. We are grateful to and thank Anette M. Ham-merum and Frank Hansen from SSI, who provided strain 65-Ec-09.

REFERENCES1. Rogers BA, Sidjabat HE, Paterson DL. 2011. Escherichia coli O25b-

ST131: a pandemic, multiresistant, community-associated strain. J. Anti-microb. Chemother. 66:1–14. http://dx.doi.org/10.1093/jac/dkq415.

2. Pitout JDD. 2012. Extraintestinal pathogenic Escherichia coli: a combi-nation of virulence with antibiotic resistance. Front. Microbiol. 3:9. http://dx.doi.org/10.3389/fmicb.2012.00009.

3. Kaper JB, Nataro JP, Mobley HL. 2004. Pathogenic Escherichia coli. Nat.Rev. Microbiol. 2:123–140. http://dx.doi.org/10.1038/nrmicro818.

4. Pitout JDD, Laupland KB. 2008. Extended-spectrum beta-lactamase-producing Enterobacteriaceae: an emerging public-health concern. Lan-cet 8:159 –166.

5. Peirano G, Pitout JDD. 2010. Molecular epidemiology of Escherichia coliproducing CTX-M beta-lactamases: the worldwide emergence of cloneST131 O25:H4. Int. J. Antimicrob. Agents 35:316 –321. http://dx.doi.org/10.1016/j.ijantimicag.2009.11.003.

6. Johnson JR, Urban C, Weissman SJ, Jorgensen JH, Lewis JS, Hansen G,Edelstein PH, Robicsek A, Cleary T, Adachi J, Paterson D, Quinn J,Hanson ND, Johnston BD, Clabots C, Kuskowski MA. 2012. Molecularepidemiological analysis of Escherichia coli sequence type ST131 (O25:H4) and blaCTX-M-15 among extended-spectrum �-lactamase-producing

FIG 2 The mean and SD of the log CFU/0.5 g of feces for Gram-negative anaerobic flora and Gram-positive aerobic flora, shown at day 1 and day 4 to comparethe impact of 3 days of treatment. The SD was 0 for bars where the SD is not shown.

Antibiotic Selection of E. coli ST131

October 2014 Volume 58 Number 10 aac.asm.org 6143

on Septem

ber 27, 2014 by guesthttp://aac.asm

.org/D

ownloaded from

E. coli from the United States, 2000 to 2009. Antimicrob. Agents Che-mother. 56:2364 –2370. http://dx.doi.org/10.1128/AAC.05824-11.

7. Dahbi G, Mora A, López C, Alonso MP, Mamani R, Marzoa J, Coira A,García-Garrote F, Pita JM, Velasco D, Herrera A, Viso S, Blanco JJE,Blanco M. 2013. Emergence of new variants of ST131 clonal group amongextraintestinal pathogenic Escherichia coli producing extended-spectrum�-lactamases. Int. J. Antimicrob. Agents 42:347–351. http://dx.doi.org/10.1016/j.ijantimicag.2013.06.017.

8. Tumbarello M, Spanu T, Di Bidino R, Marchetti M, Ruggeri M,Trecarichi EM, De Pascale G, Proli EM, Cauda R, Cicchetti A, Fadda G.2010. Costs of bloodstream infections caused by Escherichia coli and in-fluence of extended-spectrum beta-lactamase production and inadequateinitial antibiotic therapy. Antimicrob. Agents Chemother. 54:4085– 4091.http://dx.doi.org/10.1128/AAC.00143-10.

9. Pitout JDD. 2010. Infections with extended-spectrum beta-lactamase-producing Enterobacteriaceae: changing epidemiology and drug treat-ment choices. Drugs 70:313–333. http://dx.doi.org/10.2165/11533040-000000000-00000.

10. Rodríguez-Baño J, Alcala JC, Cisneros JM, Grill F, Oliver A, HorcajadaJP, Tortola T, Mirelis B, Navarro G, Cuenca M, Peña C, Llanos AC,Canton R, Pascual A. 2008. Community infections caused by extended-spectrum beta-lactamase-producing. Arch. Intern. Med. 168:1897–1902.http://dx.doi.org/10.1001/archinte.168.17.1897.

11. Nielsen KL, Dynesen P, Larsen P, Frimodt-Møller N. 2014. FaecalEscherichia coli from patients with E. coli urinary tract infection andhealthy controls who have never had a urinary tract infection. J. Med.Microbiol. 63:582–589. http://dx.doi.org/10.1099/jmm.0.068783-0.

12. Rodríguez-Baño J, Navarro MD, Romero L, Muniain MA, de Cueto M,Rios MJ, Hernandez JR, Pascual A. 2006. Bacteremia due to extended-spectrum beta-lactamases producing Escherichia coli in the CTX-M era: anew clinical challenge. Clin. Infect. Dis 43:1407–1414. http://dx.doi.org/10.1086/508877.

13. Lautenbach E, Patel JB, Bilker WB, Edelstein PH, Fishman NO. 2001.Extended-spectrum beta-lactamase-producing Escherichia coli and Kleb-siella pneumoniae: risk factors for infection and impact of resistance onoutcomes. Clin. Infect. Dis. 32:1162–1171. http://dx.doi.org/10.1086/319757.

14. Bellíssimo-Rodrigues, F, Carolina A, Gomes F, Dinis A, Passos C,Achcar JA, Castro S, Martinez R, Gomes ACF, Passas ADC, PerdonáGDSC. 2006. Clinical outcome and risk factors related to extended-spectrum beta-lactamase-producing Klebsiella spp. infection among hos-pitalized patients. Mem. Inst. Oswaldo Cruz 101:415– 421. http://dx.doi.org/10.1590/S0074-02762006000600012.

15. Hansen F, Olsen SS, Heltberg O, Justesen US, Fuglsang-Damgaard D,Knudsen JD, Hammerum AM. 2014. Characterization of third-generation cephalosporin-resistant Escherichia coli from bloodstream in-fections in Denmark. Microb. Drug Resist. 20:316 –3240. http://dx.doi.org/10.1089/mdr.2013.0157.

16. Clermont O, Dhanji H, Upton M, Gibreel T, Fox A, Boyd D, MulveyMR, Nordmann P, Ruppé E, Sarthou JL, Frank T, Vimont S, Arlet G,Branger C, Woodford N, Denamur E. 2009. Rapid detection of theO25b-ST131 clone of Escherichia coli encompassing the CTX-M-15-producing strains. J. Antimicrob. Chemother. 64:274 –277. http://dx.doi.org/10.1093/jac/dkp194.

17. Nielsen JB, Albayati A, Jørgensen RL, Hansen KH, Lundgren B, Schøn-ning K. 2013. An abbreviated MLVA identifies Escherichia coli ST131 asthe major extended-spectrum �-lactamase-producing lineage in the Co-penhagen area. Eur. J. Clin. Microbiol. Infect. Dis. 32:431– 436. http://dx.doi.org/10.1007/s10096-012-1764-x.

18. Imhof A. 1996. Continuous monitoring of oxygen concentrations in sev-eral systems for cultivation of anaerobic bacteria. J. Clin. Microbiol. 34:1646 –1648.

19. Perez F, Pultz MJ, Endimiani A, Bonomo RA, Donskey CJ. 2011. Effectof antibiotic treatment on establishment and elimination of intestinal col-onization by KPC-producing Klebsiella pneumoniae in mice. Antimicrob.Agents Chemother. 55:2585–2589. http://dx.doi.org/10.1128/AAC.00891-10.

20. Erlendsdottir H, Knudsen JD, Odenholt I, Cars O, Espersen F, Fri-modt-møller N, Fuursted K, Kristinsson KG, Gudmundsson S. 2001.Penicillin pharmacodynamics in four experimental pneumococcal infec-tion models. Antimicrob. Agents Chemother. 45:1078 –1085. http://dx.doi.org/10.1128/AAC.45.4.1078-1085.2001.

21. Knudsen JD, Fuursted K, Frimodt-Møller N, Espersen F. 1997. Com-parison of the effect of cefepime with four cephalosporins against pneu-mococci with various susceptibilities to penicillin, in vitro and in themouse peritonitis model. J. Antimicrob. Chemother. 40:679 – 686. http://dx.doi.org/10.1093/jac/40.5.679.

22. Jakobsen L, Cattoir V, Jensen KS, Hammerum AM, Nordmann P, Fri-modt-Møller N. 2012. Impact of low-level fluoroquinolone resistance genesqnrA1, qnrB19 and qnrS1 on ciprofloxacin treatment of isogenic Escherichiacoli strains in a murine urinary tract infection model. J. Antimicrob. Che-mother. 67:2438–2444. http://dx.doi.org/10.1093/jac/dks224.

23. Sandberg A, Jensen KS, Baudoux P, Van Bambeke F, Tulkens PM,Frimodt-Møller N. 2010. Intra- and extracellular activities of dicloxacillinagainst Staphylococcus aureus in vivo and in vitro. Antimicrob. AgentsChemother. 54:2391–2400. http://dx.doi.org/10.1128/AAC.01400-09.

24. Kerrn MB, Frimodt-Møller N, Espersen F. 2003. Effects of sulfamethi-zole and amdinocillin against Escherichia coli strains (with various sus-ceptibilities) in an ascending urinary tract infection mouse model. Anti-microb. Agents Chemother. 47:1002–1009. http://dx.doi.org/10.1128/AAC.47.3.1002-1009.2003.

25. Asahi Y, Miyazaki S, Yamaguchi K. 1995. In vitro and in vivo antibac-terial activities of BO-2727, a new carbapenem. Antimicrob. Agents Che-mother. 39:1030 –1037. http://dx.doi.org/10.1128/AAC.39.5.1030.

26. Frimodt-Møller N, Thomsen V. 1987. The pneumococcus and the mouseprotection test: correlation of in vitro and in vivo activity for beta-lactamantibiotics, vancomycin, erythromycin and gentamicin. Acta Pathol. Mi-crobiol. Immunol. Scand. 95:159 –165.

27. Achá SJ, Kühn I, Mbazima G, Colque-Navarro P, Möllby R. 2005.Changes of viability and composition of the Escherichia coli flora in faecalsamples during long time storage. J. Microbiol. Methods 63:229 –238.http://dx.doi.org/10.1016/j.mimet.2005.04.024.

28. Haigh J, Degun A, Eydmann M, Millar M, Wilks M. 2011. Improvedperformance of bacterium and yeast identification by a commercial ma-trix-assisted laser desorption ionization-time of flight mass spectrometrysystem in the clinical microbiology laboratory. J. Clin. Microbiol. 49:3441.http://dx.doi.org/10.1128/JCM.00576-11.

29. Frimodt-moller N, Ostri P, Pedersen IBK, Poulsen SR. 1980. Antibioticprophylaxis in pulmonary surgery: a double-blind study of penicillin ver-sus placebo. Ann. Surg. 195:444 – 450.

30. Wilson KH, Brown RS, Andersen GL, Tsang J, Sartor B. 2006. Com-parison of fecal biota from specific pathogen free and feral mice. Anaerobe12:249 –253. http://dx.doi.org/10.1016/j.anaerobe.2006.09.002.

31. Krych L, Hansen CHF, Hansen AK, van den Berg FWJ, Nielsen DS.2013. Quantitatively different, yet qualitatively alike: a meta-analysis ofthe mouse core gut microbiome with a view towards the human gut mi-crobiome. PLoS One 8:e62578. http://dx.doi.org/10.1371/journal.pone.0062578.

Boetius Hertz et al.

6144 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 27, 2014 by guesthttp://aac.asm

.org/D

ownloaded from

“Epidemiological factors associated with ESBL- or non ESBL-producing E.coli causing UTI in

general practices.”

Keywords: UTI; Escherichia coli; Risk Factors; Community-acquired; ESBL.

Running title: Risk factors for E.coli UTI in general practices

(1,2) Frederik Boëtius Hertz, (1) Kristian Schønning, (1) Steen Christian Rasmussen (1), Pia

Littauer, (1) Jenny Dahl Knudsen, (2) Anders Løbner-Olesen and (1) Niels Frimodt-Møller

1) Dept. of Clinical Microbiology, Hvidovre University Hospital, Hvidovre, Denmark.

2) Dept. of Biology, University of Copenhagen, Denmark.

Corresponding author:

Frederik B. Hertz, MD

Hvidovre University Hospital

Dept. of Clinical Microbiology

Copenhagen

004538626971

[email protected]

Abstract

Background: The frequency of community-onset urinary tract infections (UTI), caused by ESBL-

producing E.coli is increasing. This demands general practitioners to rapidly identify patients at

risk. Epidemiological differences between UTIs caused by ESBL-producing, resistant (non-ESBL)

and susceptible E.coli have never been thoroughly investigated.

Methods: This was a triple-case-control study with three case groups and one group of uninfected

controls, all from general practice. We performed prospective collection of urine samples at Dept.

of Clinical Microbiology at Hvidovre Hospital, Denmark, among samples from general practices

from October 2011 to July 2012 followed by a retrospective analysis of epidemiological

characteristics. All included cases had a urine sample positive for E.coli > 1000 CFU/ml.

Results: We included 98 cases with UTI caused by ESBL-producing E.coli, 174 patients with

antibiotic-resistant (non-ESBL) E.coli and 177 patients with susceptible E.coli. These were

individually compared to a group of 200 individuals with negative urine samples. Case groups had a

significantly higher use of antibiotics than the control group within 30 days prior to infection

(P<0.0001). Prior to infection the ESBL group had significantly more hospital admissions, higher

use of antibiotics and more urinary tract infections with cefpodoxime resistant E.coli, than the other

case groups (P<0.05).

Conclusion: We found previous exposure to antibiotics to be a risk factor for UTI with E.coli in

general. Antibiotics did not prove to be indisputable predictors of ESBL infection. Healthcare

contact and prior UTI with ESBL-producing E.coli within 90 days of infection were independent

risk factors for community onset UTI by ESBL-producing E.coli.

Introduction

Urinary tract infection (UTI) is one of the most common bacterial infections in hospital settings and in the

community and Escherichia coli is the most common etiology (1,2). E.coli is additionally becoming

increasingly resistant, in part due to the presence of plasmid-borne Extended-Spectrum Beta-Lactamases

(ESBL) (2–4). Plasmids with genes coding for co-resistance to beta-lactams, fluoroquinolones,

aminoglycosides and sulphonamides are now a common observation, leaving the isolates multi-drug

resistant (3,5). Community-acquired UTI is the infection most often caused by ESBL-producing E.coli

and community-onset infections (COI) represent a major clinical challenge (5–8). Furthermore, resistant

E.coli are able to colonize the gut of humans for long periods of time, and current studies report increased

carriage of ESBL-producing E.coli among all age groups (9–12). Numerous studies have consequently

investigated epidemiological characteristics of patients infected with ESBL-producing E.coli, reporting

frequently identified risk factors such as age > 65, male sex, recent use of antibiotics (in particular

cephalosporins and fluoroquinolones), recent hospitalization and the presence of underlying disease

(3,5,6,13–15). Previous colonization with ESBL-producing E.coli, the presence of an urinary catheter and

previous UTI were likewise independent risk factors (14–16). Finally, recent publications have identified

travel to high-prevalence countries and lifestyle to be involved in colonization and infection (10,13,15).

Antibiotic resistance and subsequent inappropriate empirical therapy are associated with increased

mortality, a higher use of broad-spectrum antibiotics as well as increased healthcare costs (6,8,14). The

described risk factors apply to both healthcare and community associated infections, but ESBL -

producing E.coli among patients with no recent healthcare exposure are emerging (5). Uncomplicated

infections with a community-onset are often diagnosed and treated empirically at general practices

without preceding susceptibility tests. There is therefore an essential need to rapidly identify patients at

risk to avoid the continuous selection of resistant strains and inappropriate antibiotic therapy (5,17). The

association between use of antibiotics and infections caused by ESBL-producing E.coli from primary care

has been sparsely investigated in low-ESBL prevalence countries. A triple-case-control study focusing on

E.coli has never been performed, and very few case-control studies have included an uninfected group

(15,18,19). In the 2012 Danish population, 4% and 6% of E.coli from urine samples from general practice

and hospitals were resistant to extended-spectrum cephalosporins, respectively. In Denmark, we have

comprehensive nationwide databases on hospital contacts and prescription of medicine from general

practices. Thus, Denmark is an appropriate site for investigation of epidemiological factors associated

exclusively with non-hospitalized patients with E.coli UTI. Our purpose with this triple-case-control

study was therefore to evaluate the risk factors, antibiotics in particular, for contracting an UTI with an

ESBL-producing E.coli compared to less resistant and fully susceptible E.coli.

Material and methods

Research ethical approvals

This study was approved by the Danish Health and Medicines Authority, Statistics Denmark (DST),

The Regional Committee of Danish Data Protection Agency and patient notification was declared

not necessary by the Regional Committee of Health Research Ethics Committee (HVH-2012-017,

J.nr. 3-3013-230/1/KWH and H-4-2012-088).

Study design and population

Three parallel retrospective case-control analyses were carried out as a triple-case-control study at

Department of Clinical Microbiology at Hvidovre University Hospital in Copenhagen, Denmark

(DCM). This is an urban area with approximately 950.000 inhabitants and DCM serves more than

450 general practitioners. Three different case groups and one control group were included. All

three case groups were individually compared to the control group. All groups consisted of a unique

collection since a patient could only be included in one group and only once. The study period was

from the 1st of October 2011 to the 30th of June 2012. Patients, including controls, were eligible for

inclusion if a urine sample had been submitted in the given time period, disregarding age, gender

and previous ESBL-positive samples. Exclusion criteria were no Danish social security (CPR)

number meaning no available records on prescriptions or hospital contacts. No extended matching

of controls was performed.

Collection of subjects was conducted at the laboratory at DCM and samples were collected only

among urine specimens from general practices. Case groups were all patients with a urine sample

positive for E.coli > 1000 CFU/ml and the control group consisted of patients, randomly collected,

with a urine sample without bacterial growth (referred to as uninfected) during the study period.

The uninfected group was therefore represented by patient data only, with no existing

microbiological analysis. Case group 1 was the group with ESBL-producing E.coli (referred to as

ESBL), case group 2 consisted of patients in which the E.coli was resistant to at least one of the

tested antimicrobials (ampicillin, cefuroxime, aztreonam, ampicillin/clavulanic acid,

piperacillin/tazobactam, mecillinam, ceftazidime, meropenem, ciprofloxacin, sulfamethoxazole,

trimethoprim, tetracycline, gentamicin, tobramycin, nitrofurantoin, fosfomycin,) but without an

ESBL phenotype (referred to as resistant) and case group 3 comprised patients with UTI caus

Patient variables and definitions

The following variables were assessed from Statistics Denmark (DST) and DCM records one year

prior to until 90 days after index sampling: Age, sex, hospital admission, prescriptions of

medication, reported use of urinary catheter and number of urine samples submitted to DCM for

investigation. Urine samples were not included in the multiple statistical analyses, since data from

DST are available by extern access only, with no possible merging of DCM and DST data.

Retrospectively, UTI was defined as either complicated or uncomplicated by previously established

definitions: An uncomplicated UTI occurs in patients with no structural or functional abnormalities,

are not pregnant or who have not been submitted to instrumented procedures (as catheterization)

(2). Furthermore, UTI was categorized as either true community onset infections (COI) or

healthcare associated (HCAI), by modified Friedman criteria (23): HCAI was defined from a

sample obtained from a patient with at least one of the two criteria: (і) Patient admitted to a hospital

>2 days within 3 months prior to urine sampling or (іі) patient who initiated hospital associated

outpatient clinic contact within 30 days prior to index sample (obtained from DST for this

classification). Prescription of medicine was not available from hospital settings and data included

community-associated prescriptions only. On the contrary, diagnoses were just obtainable from

hospital settings.

Microbiological analysis

All microbiological analyses were performed at DCM.

Susceptibility patterns

Bacterial susceptibility testing was performed by either Antibiotic Disc Diffusion Test

Methodology or by The Vitek 2 automated system (Cards AST-N209 and AST-N122) (24,25). Disc

Diffusion was performed using the Disk Diffusion Test Methodology (Version 1.0, December 18,

2009) as described in the European Committee on Antimicrobial Susceptibility Testing (EUCAST))

(26). Vitek 2 testing was performed according to guidelines of DCM and in accordance with

direction from the manufacturer (bioMérieux) and as described elsewhere (24,25). For Vitek 2,

isolates found resistant to mecillinam were tested by disk diffusion or Etest to confirm resistance.

An ATCC 25922 E.coli was used for quality control for all methods and results interpreted

according to recommendations from EUCAST (www.eucast.org/clinical_breakpoints/). All strain

found as I (intermediately resistant) and R were reported as resistant and strains found as S were

reported as susceptible.

ESBL-producing strains were phenotypically identified by the MAST-test, performed as a

combined-disk method, using disk containing Cefpodoxime ±ESBL and/or AmpC inhibitors

(MAST®, Merseyside, UK). A difference in zone of inhibition >5mm between disks with and

without inhibitor combinations was considered a positive phenotypic result as previously described

(26,27).

Molecular tests

Phylogroup testing

All isolates were classified according to phylogroups A, B1, B2, D or Non-typeable, respectively,

by singleplex PCR amplification of three genes as previously described and isolates were tested for

the presence of ibeA when necessary (28,29). The method was chosen albeit a recently published

paper indicates that some isolates are possibly assigned incorrect phylogroups (30).

ESBL genotype identification

As previously described, identification of ESBL genotype, on ESBL-producing isolates, was done

by screening for the presence of blaCTX-M by PCR with subsequent identification of specific CTX-M

gene by sequencing (23,31).

Statistical analysis

The triple-case-control study was analysed using a conservative two step evaluation. We performed

an initial univariate t-test with a no-hypothesis strategy. We left out variables with no immediate

relevance for the study and included all medications prescribed, admissions to hospitals and hospital

diagnoses. We divided data into time intervals as 90 and 30 days prior to urine sampling. Variables

with P<0.1, as found by the univariate analysis, were subsequently included in the multivariate

logistic-regression models to identify individual risk factors. Variables were included in the time

period where P<0.1 and similar variables were tested for all three case groups. A second run was

performed as a simplified manual backward stepwise elimination procedure including variables

with P<0.1 in the multivariate tests only. Finally, the multivariate analyses were repeated separately

comparing the ESBL case group to the other case groups. Outcome was set as the ESBL group.

Odds ratios were estimated with the reservations needed for the study design and are therefore not

shown (33). Hosmer and Lemeshow Goodness of Fit test was evaluated for all multivariate

analyses. When appropriate, chi-square or Fisher´s exact tests were used. All analyses were

performed using SAS version 9.2 and 9.3 (SAS Institute, Cary, NC) and P<0.05 was considered

significant. Results from the initial univariate t-tests are not shown, but appropriate p-values from

chi-square and Fisher´s exact test are presented in written text and appropriate tables.

We did not include COI and HCAI in the multivariate statistical analysis. We simply assessed the

number of patients in each group rather than to compare HCAI with COI (34).

Results

Patient characteristics and multivariate analysis

In the study period we included 98 patients to the ESBL case group, 174 patients to the resistant

groups, 177 to the susceptible group and 200 patients to the uninfected group. All collected E.coli

isolates were included in the molecular analysis but a few patients were excluded during analysis of

data from DST, due to a missing date of birth. We excluded two patients from the ESBL group,

three from the resistant group and two from the susceptible group. No patients were excluded from

the uninfected group. Relevant data on patient characteristics and use of antibiotics are shown in

Table 1. Results of the multivariate analysis are found in Table 2 and 3. We found that patients in

the three case groups were significantly older than among the uninfected controls (P<0.0001).

Conversely, there was no significant difference in age between the case groups (P>0.05).

Furthermore, there were more men among the uninfected group than in the resistant and the

susceptible groups, but no significant difference among the case groups. When assessed as part of

the multivariate analysis, there was a significant difference between both gender and age for all case

groups compared to the uninfected group (P<0.005 and P<0.0001).

Risk factors for case groups compared to the uninfected group

Results of the multivariate analyses are found in Table 2. Data were analysed during two periods,

i.e. 90 and 30 days prior to sampling. During 90 days prior to sampling, few antibiotics and

diagnoses were found as independent risk factors. In the ESBL group only use of mecillinam in the

backward elimination analysis was found to be significant (P=0.0426). The use of five and two

specific antibiotics were independent risk factors for the resistant and susceptible groups,

respectively (all P<0.05). In the period of 30 days prior to index sample, prescription of antibiotics

in general was significant in all case groups, when analysed by chi-square (P<0.0001), but only

mecillinam (P<0.0001), mecillinam and trimethoprim (P<0.0001 and P=0.0249), mecillinam and

sulphamethizole (P<0.0001 and P=0.0205) were independent risk factors in the two resistant groups

and the susceptible group, respectively. The diagnoses “kidney infections” and “urosepsis” were

risk factors for the ESBL group (P=0.0106 and P=0.0334). There was no significant difference in

number of community- and healthcare-associated infections, (P>0.05, data not shown).

Risk factors for the ESBL case group compared to the other case groups

These results are illustrated in Table 3. There were significantly more HCAI patients in the ESBL

group than in the susceptible group (P=0.0058) as calculated by the chi-square test. Looking at

number of patients receiving antibiotics and prescriptions made, it was not evident that the ESBL

group had a higher use of antibiotics 90 days before index sample (Table 1). Analysis of the time

periods and the multivariate analyses performed in two runs, showed that use of antibiotics in

general was a risk factor for the ESBL group 90 days prior to index sample (P=0.0049 and

P=0.0029) and hospital admission 30 days prior was an independent risk factor for ESBL

(P=0.0073 and P=0.0216). The use of trimethoprim and sulphamethizole, in percentages, were

higher or equivalent in the resistant and the susceptible groups compared to the ESBL group (Table

1). There was a significant difference in use of trimethoprim (P<0.0298) and sulphamethizole

(P<0.0381) between the ESBL and the resistant groups and in use of sulphamethizole between the

ESBL group and the susceptible group (P=0.0299). Only with the manual backward selection

model, did use of quinolones prove significantly higher in the ESBL group 30 days prior to

sampling, compared to the resistant group (P=0.0365) and total use of antibiotics comparing the

ESBL and susceptible groups (P=0.04).

Urine Samples to Dept. of Clinical Microbiology

The sum of positive urines sample submitted from general practice to DCM as related to the

different patient case groups, as shown in Table 4, were analysed by Fishers exact test. P-values are

listed in the text where appropriate.

Overall, significantly more urine samples were submitted from patients in the ESBL and resistant

groups than from patients in the uninfected group during one year prior as well as during 90 days

before the index sample (P<0.05 for all). All case groups had significantly more positive urine

samples and significantly more samples positive for E.coli than the control group, 90 days before

and after index sampling (P<0.0001 for all). The ESBL group had significantly more urine samples

positive for CDP-R E.coli than the other three groups (P<0.0001) before and after the index sample.

Compared to the period prior to the index sample, all three case groups had significantly fewer

positive urine samples during the 90 days after the index sample and the ESBL group had

significantly less urine samples positive for cefpodoxime resistant E.coli (P<0.0001) with no

significant changes in the other case groups.

Antibiotic susceptibility, phylogroups and ESBL genotypes

The ESBL-producing E.coli isolates displayed increased resistance to most of the tested antibiotics

except towards mecillinam (2%), nitrofurantoin (7%), fosfomycin (3%) and piperacillin/tazobactam

(4%). Regarding prescriptions 90 days prior to index sample (Table 1), mecillinam was given to

55% of patients, nitrofurantoin to 14% and sulphamethizole to 19%, respectively.

E.coli from the resistant group showed lower rates of resistance, than the ESBL group, towards

most antibiotics (1%-15%) except towards trimethoprim (46%), sulphamethizole (68%),

tetracycline (47%), ampicillin (84%) and to some extent towards ciprofloxacin (22%), with low

resistance to towards mecillinam (3%), nitrofurantoin (5%), fosfomycin (3%) and

piperacillin/tazobactam (3%). In this group 58% received mecillinam and 13% nitrofurantoin,

respectively with 27% receiving sulphamethizole.

Distributions of phylogroups were very similar among the three case groups with phylogroup B2

dominating and group D being the second largest group: Phylogroup A , 7-10%; phylogroup B1, 1-

6%; phylogroup B2 , 45-58%; phylogroup D ,22-32%; and non-typeable, 4-13%, respectively.

All 98 ESBL-producing E.coli were investigated for the presence of an ESBL gene. A total of 98%

carried a blaCTX-M type ESBL and 2% did not have an ESBL gene identified. blaCTX-M group 1

represented 73% of all isolates with 54% of all isolates carrying CTX-M-15. Furthermore, 24%

belonged to blaCTX-M group 9 with two clusters of CTX-M-14 (12%) and CTX-M-27 (12%).

Discussion

To our knowledge, this is the first triple-case-control study of epidemiological characteristics for

UTI caused by E.coli. It is moreover the first study to exclusively include non-hospitalized patients

of all ages in a country with low ESBL prevalence. As reported from previous, mainly hospital

based, investigations, we found healthcare contact and previous UTI with ESBL-producing E.coli to

be associated with community-onset UTI with ESBL-producing E. coli (3,5,6,13–15,35). However,

we have also exposed some new and interesting aspects of the epidemiology of UTI caused by

E.coli. First of all, surprisingly few differences were found between the UTI case groups, when

these were compared to an uninfected control group and here no clear difference was found

regarding exposure to antibiotics as risk factors for ESBL-producing E.coli. We did find our case

groups to be of older age than the control group, but there was no significant difference in age or

gender among the case groups, even though the ESBL group did have a higher average age and a

higher number of men, respectively. In Norway, another low prevalence country, the ESBL

population was found to be younger but they also found travel to be a risk factor, indicating that

other features than healthcare contact has an influence for infections with ESBL-producing

Enterobacteriaceae in younger individuals (8,36). In our study, we found no significant difference

(P>0.05) between the percentages of HCAI ESBL infections in the age group 18-64 and >65,

respectively (Table 1). Travel history was not studied.

Regarding the prescriptions of antibiotics, the case groups had a significantly higher use before

index sampling than in the uninfected control group. Interestingly, use of mecillinam posed a risk

factor for UTI for all case groups within 90 days of infection, which is in contradiction to what has

been seen in Norway where mecillinam was found not to be associated with community-onset

ESBL infections (8). The Norwegian study did however not include an uninfected control group

and we did not find mecillinam to be associated with ESBL infection when compared to the other

case groups. Mecillinam is widely used as a first-line drug for treatment of UTI in Denmark and

considered to be an effective agent, also against most ESBL-producing E.coli, with an inhibiting

effect on E.coli in the intestines (37,38). It was previously seen that combination drugs of beta-

lactams/beta-lactamase-inhibitors were a risk factor for ESBL infection, but it remains unclear why

antibiotics with an effect on most resistant E.coli, including ESBL, seems to select for these

uropathogens (16). The healthcare aspect of hospital admissions as a risk factor became evident

when comparing the case groups. Here, we found the ESBL group to have significantly more

healthcare associated infection compared to the group of susceptible E.coli, and significantly more

admissions than both of the other case groups. This confirms hospital contact as an independent risk

factor for ESBL-producing E.coli infections as has been establish previously (3,5,6,13–15). Total

use of antibiotics was significantly higher in the ESBL group. However, exposure to antibiotics was

not unmistakeably higher among patients from the ESBL group, with varied antibiotics being used

in the groups (Table 1 and 3).

We found that the ESBL group had significantly more urine samples being positive for growth of

CPD-R E.coli. This was the case one year prior, 90 days prior and also 90 days after index

sampling, clearly showing that previous UTI plays an important role when assessing patients at risk

(14–16).

We chose to study epidemiological characteristics of ESBL-producing E.coli as a triple-case-control

study in accordance with what was previously suggested for investigations of risk factors (19,33).

When designing such a study, it is of great importance that the design includes an uninfected

control group, especially to reduce the risk of overestimating the use of antibiotics (19,33).

Furthermore, the control group should represent the background population from which all

individuals are included (19,33). As we chose a control group among individuals with a negative

urine culture, there is a clear risk of selection bias, as this group seeks medical attention and might

not represent the true background population. Our triple-case-control design is therefore not suited

for the investigation of possible infection control. However, the design can address the issue of

recommendations for empiric treatment when susceptibility patterns of the suspected pathogen are

unknown (19). Other limitations of our study are the lack of data on nursing home residence and

outpatient contacts, which will possibly lead us to underestimate the significance of healthcare

exposures and make it difficult to identify possible reservoirs for ESBL-producing E.coli. It would

also have been informative to include diagnoses from general practices as well as medications from

hospitals to give us the full overview of illnesses and treatments. Since we only included patients

who had urine samples submitted to the laboratory, we have not been able to include patients

treated empirically with no sampling or patients treated by general practitioners performing in-

house susceptibility testing. Finally, as we have had no direct contact to the included individuals,

we have not been able to investigate issues like travel and lifestyle. Furthermore, as all case groups

have more positive urine samples submitted to DCM, with no matching of patients based on UTI-

free periods, there is a risk that we are actually studying three case-groups of patients with recurrent

UTI, and this would pose an obvious reason for antibiotic prescriptions. However, our data on

hospital diagnoses argues against this bias.

Nonetheless, ESBL-producing E.coli are becoming increasingly frequent as community-acquired

infections and underestimating the risk of ESBL-producing E.coli may lead physicians to treat

patients with inappropriate empirical regimens. This may lead to selection of ESBL-positive

isolates, treatment failure and risk of severe morbidity. The selection pressure can be enforced by

several different antibiotics. Overestimating the risk of ESBL-producing E.coli can lead to the use

of broad-spectrum antibiotics leading to a higher selection pressure and the emergence of other

resistant strains (16). Therefore, to fight the spread of ESBL-producing E.coli, the physicians need

to rapidly identify patients infected with resistant E.coli and treat them accordingly. The use of

drugs with potentially good effect on ESBL should lower the risk of selection of ESBL-producing

E.coli. Nevertheless, our findings and previous studies indicate that this is not enough to avoid an

increase in ESBL prevalence, even if urine samples with CPD-R E.coli did significantly decrease

after treatment. We found mecillinam to be an independent risk factor for all case groups (16). This

could be as a result of selection of present ESBL-producing E. coli due to inefficient gut

concentrations, thereby maintaining ESBL in the faeces, or due to constant re-exposure to resistant

pathogens from an unknown reservoir (9). This reservoir could be nursing homes and long-term

care facilities, as previously stated (39) and which is not possible for us to determine. The reservoir

could also be hospital settings suggested by fact that the number of HCAI and hospital admissions

seemed to be strictly linked to ESBL infection – at least within 90 days of UTI. We did not find

admissions within one year prior to infection to be a risk factor, which corresponds to a recent

publication describing six months as the duration that healthcare exposure increases the risk of a

ESBL infection (15). We speculate if the hospital use of antibiotics with broad spectra select for

ESBL-producing E.coli in healthcare settings while use of oral antibiotics in the community select

for resistant, non-ESBL-producing E.coli.

In conclusion, we found similar risk factors for ESBL-producing E.coli infections in non-

hospitalized patients as previously proposed. However, in our study exposure to antibiotics proved

to be a problematic variable to use as guidance for general practitioners when assessing patients. All

in all, patients at risk of UTI caused by E.coli are likely to have had a UTI within 90 days and to

have received antibiotics. Nonetheless, only previous UTI by ESBL-producing E.coli and

healthcare exposure including hospital admission within 90 days are true risk factors for UTI caused

by ESBL-producing E.coli.

Acknowledgements

Work has been performed as cooperation between HVH, Roskilde University and University of

Copenhagen with laboratory facilities provided by HVH. This work was, in parts, presented at ECCMID

in Berlin 2013 and at 53rd Interscience Conference on Antimicrobial Agents and Chemotherapy, Denver,

CO, 9th-13th September 2013.

Funding

This work was supported by the EU-FP funded project PAR7, DanCARD (Danish National

Strategic Research Foundation, project 09-067075/DSF), Roskilde University, University of

Copenhagen and the SSAC Foundation (SLS-327421 and SLS-251761).

Transparency declarations

None to declare.

1. Kahlmeter G, Poulsen HO. Antimicrobial susceptibility of Escherichia coli from community-

acquired urinary tract infections in Europe: the ECO·SENS study revisited. Int J Antimicrob

Agents. Elsevier B.V.; 2012 Jan [cited 2012 Nov 27];39(1):45–51.

2. Foxman B. The epidemiology of urinary tract infection. Nat Rev Urol [Internet]. Nature

Publishing Group; 2010 Dec;7(12):653–60.

3. Doi Y, Park YS, Rivera JI et al. Community-associated extended-spectrum β-lactamase-

producing Escherichia coli infection in the United States. Clin Infect Dis. 2013 Mar.

4. Livermore DM. Fourteen years in resistance. Int J Antimicrob Agents. Elsevier B.V.; 2012

Apr [cited 2014 Mar 29];39(4):283–94.

5. Ben-ami R, Arslan H, Pitout JDD et al. A Multinational Survey of Risk Factors for Infection

with Extended-Spectrum b -Lactamase – Producing Enterobacteriaceae in Nonhospitalized

Patients. 2009;49.

6. Hayakawa K, Gattu S, Marchaim D et al. Epidemiology and Risk Factors for Isolation of

Escherichia coli Producing CTX-M-Type Extended-Spectrum β-Lactamase in a Large U.S.

Medical Center. Antimicrob Agents Chemother. 2013 Aug;57(8):4010–8.

7. Peirano G, van der Bij AK, Gregson DB et al. Molecular epidemiology over an 11-year

period (2000 to 2010) of extended-spectrum β-lactamase-producing Escherichia coli causing

bacteremia in a centralized Canadian region. Journal of clinical microbiology. 2012. p. 294–

9.

8. Søraas A, Sundsfjord AS, Sandven I et al. Risk Factors for Community-Acquired Urinary

Tract Infections Caused by ESBL-Producing Enterobacteriaceae -A Case-Control Study in a

Low Prevalence Country. PLoS One; 2013.

9. Ruppé E, Lixandru B, Cojocaru R et al. Relative Fecal Abundance of Extended-Spectrum-β-

Lactamase-Producing Escherichia coli Strains and Their Occurrence in Urinary Tract

Infections in Women. Antimicrob Agents Chemother. 2013 Sep;57(9):4512–7.

10. Jørgensen SB, Samuelsen O, Sundsfjord A et al. High prevalence of faecal carriage of ESBL-

producing Enterobacteriaceae in Norwegian patients with gastroenteritis. Scand J Infect Dis.

2014 Apr 4;(1478):462–5.

11. Fernández-Reyes M, Vicente D, Gomariz M et al. High Rate of Fecal Carriage of Extended-

Spectrum-β-Lactamase-Producing Escherichia coli in Healthy Children in Gipuzkoa,

Northern Spain. Antimicrob Agents Chemother. 2014 Mar;58(3):1822–4.

12. Dobrindt U, Hacker JH. Current Topics in Microbiology and Immunology Between

Pathogenicity and Commensalism.

13. Søraas A, Sundsfjord A, Sandven I et al. Risk factors for community-acquired urinary tract

infections caused by ESBL-producing enterobacteriaceae--a case-control study in a low

prevalence country. PLoS One. 2013 Jan;8(7):e69581.

14. Apisarnthanarak A, Kiratisin P, Saifon P et al. Clinical and molecular epidemiology of

community-onset, extended-spectrum beta-lactamase-producing Escherichia coli infections

in Thailand: a case-case-control study. Am J Infect Control. 2007 Nov;35(9):606–12.

15. Rogers BA, Ingram PR, Runnegar N et al. Community-Onset Escherichia coli Infection

Resistant to Expanded-Spectrum Cephalosporins in Low-Prevalence Countries. Antimicrob

Agents Chemother. 2014 Apr;58(4):2126–34.

16. Goulenok T, Ferroni A, Bille E et al. Risk factors for developing ESBL E. coli: can clinicians

predict infection in patients with prior colonization? J Hosp Infect. 2013 Aug;84(4):294–9.

17. Foxman B. Extended-spectrum β-lactamase-producing escherichia coli in the united states:

time to rethink empirical treatment for suspected E. coli infections? Clin Infect Dis. 2013

Mar;56(5):649–51.

18. Søraaas A, Sundsfjord A, Sandven I et al. Risk Factors for Community-Acquired Urinary

Tract Infections Caused by ESBL-Producing Enterobacteriaceae – A Case – Control Study in

a Low Prevalence Country. 2013;8(7):1–7.

19. Kaye KS, Harris AD, Samore M et al. The Case-Case-Control Study Design: Addressing The

Limitations of Risk Factor Studies for antimicrobial resistance. 2005;26(4):346–51.

20. Lausch KR, Fuursted K, Larsen CS et al. Colonisation with multi-resistant

Enterobacteriaceae in hospitalised Danish patients with a history of recent travel: A cross-

sectional study. Travel Med Infect Dis. Elsevier Ltd; 2013 Jun 27;4–7.

21. Hansen DS, Schumacher H, Hansen F et al., Stegger M, Hertz FB, Schønning K, et al.

Extended-spectrum β-lactamase (ESBL) in Danish clinical isolates of Escherichia coli and

Klebsiella pneumoniae: prevalence, β-lactamase distribution, phylogroups, and co-resistance.

Scand J Infect Dis. 2012 Mar;44(3):174–81.

22. DANMAP 2012. DANMAP. 2012;

23. Nielsen JB, Albayati A, Jørgensen RL et al. An abbreviated MLVA identifies Escherichia

coli ST131 as the major extended-spectrum β-lactamase-producing lineage in the

Copenhagen area. Eur J Clin Microbiol Infect Dis. 2013 Nov 6;32(3):431–6.

24. Livermore DM, Struelens M, Amorim J et al. et al. Multicentre evaluation of the VITEK 2

Advanced Expert System for interpretive reading of antimicrobial resistance tests. J

Antimicrob Chemother. 2002 Feb;49(2):289–300.

25. Nyberg SD, Meurman O, Jalava J et al. Evaluation of detection of extended-spectrum beta-

lactamases among Escherichia coli and Klebsiella spp. isolates by VITEK 2 AST-N029

compared to the agar dilution and disk diffusion methods. Scand J Infect Dis. 2008

Jan;40(5):355–62.

26. Hansen F, Olsen SS, Heltberg O et al. Characterization of Third-Generation Cephalosporin-

Resistant Escherichia coli from Bloodstream Infections in Denmark. Microb Drug Resist.

2014 Feb 11;00(00):1–9.

27. Hsieh C, Shen Y, Hwang K. Clinical Implications , Risk Factors and Mortality Following

Community-onset Bacteremia Caused by Extended-spectrum b-lactamase ( ESBL ) and non-

ESBL Producing Escherichia coli. J Microbiol Immunol Infect. Taiwan Society of

Microbiology; 2010;43(3):240–8.

28. Clermont O, Bonacorsi S, Bingen E. Rapid and simple determination of the Escherichia coli

phylogenetic group. Appl Environ Microbiol. 2000 Oct;66(10):4555–8.

29. Gordon DM, Clermont O, Tolley H et al. Assigning Escherichia coli strains to phylogenetic

groups: multi-locus sequence typing versus the PCR triplex method. Environ Microbiol.

2008 Oct;10(10):2484–96.

30. Clermont O, Christenson JK, Denamur E et al. The Clermont Escherichia coli phylo-typing

method revisited: improvement of specificity and detection of new phylo-groups. Environ

Microbiol Rep. 2013 Feb;5(1):58–65.

31. Jørgensen RL, Nielsen JB, Friis-Møller A et al. Prevalence and molecular characterization of

clinical isolates of Escherichia coli expressing an AmpC phenotype. J Antimicrob

Chemother. 2010 Mar;65(3):460–4.

32. Pérez-pérez FJ, Hanson ND. Detection of Plasmid-Mediated AmpC Beta -Lactamase Genes

in Clinical Isolates by Using Multiplex PCR. 2002;40(6):2153–62.

33. Harris AD, Karchmer TB, Carmeli Y et al. Methodological principles of case-control studies

that analyzed risk factors for antibiotic resistance: a systematic review. Clin Infect Dis. 2001

Apr 1;32(7):1055–61.

34. Marchaim D, Gottesman T, Schwartz O et al. National multicenter study of predictors and

outcomes of bacteremia upon hospital admission caused by Enterobacteriaceae producing

extended-spectrum beta-lactamases. Antimicrob Agents Chemother. 2010 Dec;54(12):5099–

104.

35. Lautenbach E, Bilker WB, Tolomeo P et al. Impact of diversity of colonizing strains on

strategies for sampling Escherichia coli from fecal specimens. J Clin Microbiol. 2008

Sep;46(9):3094–6.

36. Rogers B a, Sidjabat HE, Paterson DL. Escherichia coli O25b-ST131: a pandemic,

multiresistant, community-associated strain. J Antimicrob Chemother. 2011 Jan;66(1):1–14.

37. Jansåker F, Frimodt-Møller N, Sjögren I et al. Clinical and bacteriological effects of

pivmecillinam for ESBL-producing Escherichia coli or Klebsiella pneumoniae in urinary

tract infections. J Antimicrob Chemother. 2013 Oct 9;(October 2013):769–72.

38. Sullivan a, Edlund C, Svenungsson B et al. Effect of perorally administered pivmecillinam

on the normal oropharyngeal, intestinal and skin microflora. J Chemother. 2001

Jun;13(3):299–308.

39. Rooney PJ, O’Leary MC, Loughrey AC et al. Nursing homes as a reservoir of extended-

spectrum beta-lactamase (ESBL)-producing ciprofloxacin-resistant Escherichia coli. J

Antimicrob Chemother. 2009 Sep;64(3):635–41.

Table 1. Patient characteristics. Demographics of individuals included in the study and a list

of exposure to antibiotics shown for 90 days prior to index sample. The list is given as number

of patients receiving antibiotics and the total number of prescriptions. N* = Number of

patients. ▪Prescr. = Number of prescriptions.

Case groups

ESBL n=96 Resistant n=171 Susceptible n=175 Controls n=200

Demographics N* (%) N* (%) N* (%) N* (%)

Age (Mean), years 60.45 96(100) 57.95 171(100) 53,25 175(100) 42.10 200(100)

Age>18 (Mean), years 65.37 88(92) 58,5 169(99) 56,07 165(94) 46.16 180(90)

Age >65 (Mean), years 76.49 48(50) 79.09 85(50) 79.72 66(38) 76.49 37(19)

Male 18 (19) 17(10) 16(9) 54(27)

Female 78(81) 154(90) 159(91) 146(73)

Community-Onset-Infection (COI) 71(74) 139(81) 153(87) 163(82)

Health-Care-Associated-Infection (HCAI) 25(26) 32(19) 22(13) 37(19)

COI < 18 8(100) 1(50) 10(100) 18(90)

HCAI < 18 0 1(50) 0 2(10)

COI Age 18-64 30(75) 77(92) 90(91) 119(83)

HCAI Age 18-64 10(25) 7(8) 9(9) 24(17)

COI Age >65 33(69) 61(72) 53(80) 26(70)

HCAI Age >65 15(31) 24(28) 13(20) 11(30)

Complicated urinary tract infection 1(1) 1(0.6) 0

Prescriptions of antibiotics ESBL n=96 Resistant n=171 Susceptible n=175 Controls n=200

90 Days Prior N* (%) ▪Prescr. N*

(%)

▪Prescr. N* (%) ▪Prescr. N* (%) ▪Prescr.

Total antibiotics 78 (81) 199 148

(87)

329 137 (78) 231 92 (46) 164

Nitrofurantoin 13 (14) 21 22

(13)

38 16 (9) 18 8 (4) 8

Fluoroquinolones 11 (11) 13 7 (4) 9 6 (3) 7 12 (6) 13

Trimethoprim 4 (4) 8 20

(12)

31 4 (2) 4 4 (2) 4

Sulphonamides 18 (19) 22 47

(27)

58 39 (22) 47 31 (26) 31

Macrolides 6 (6) 10 5 (3) 6 5 (3) 6 6 (3) 8

Penicillins 13 (14) 15 19

(11)

21 13 (7) 14 9 (5) 10

Mecillinam 53 (55) 77 99

(58)

137 90 (51) 118 49 (25) 62

Penicillinase stable penicillins 3 (3) 5 2 (1) 3 1 (0.5) 1 4 (2) 5

Penicillins with effect on Gram-negative

(minus mecillinam)

56 (58) 86 105

(61)

153 98 (56) 130 53 (27) 75

Penicillins/beta-lactamase inhibitors 4 (4) 5 3 (2) 3 1 (0.5) 1 3 (2) 4

Table 2. Results of the Multivariate Logistic Regression analysis comparing case groups with the

uninfected group. First we performed the multivariate analysis based on results from the initial

student’s t-test followed by the manual backward stepwise elimination analysis. Finally, results of

statistical analyses on diagnoses are shown. All variables with a P<0.1 were included in both methods.

Empty squares mean estimates not computed by the statistic tool SAS. Only variables with P<0.05 are

shown. *P-values found by chi-square analyses. **P-values found by manual backward stepwise

elimination analysis.

ESBL, n=96

Resistant, n=171 Susceptible, n=175

Variables P-Value P-Value P-Value

30 Days Prior

Sex(Female) 0.0044 0.0004 0.0002

Age <.0001 <.0001 <.0001

Total antibiotics <.0001* <.0001* <.0001*

Mecillinam** <.0001** <.0001** <.0001**

Sulphamethizole 0.7379 0.0654 0.0205

Trimethoprim** 0.2616 0.0249** 0.9894

Kidney infection 0.0106* 0.9874 0.9939

Urosepsis 0.0334* . .

90 Days Prior

Mecillinam** <.0426** <.0001** 0.0001**

Nitrofurantoin 0.3871 0.0112 0.07

Penicillin** 0.1258 0.0216** 0.1048

Sulphonamides 0.9511 0.0091 0.0077

Trimethoprim 0.9848 0.0034 0.9873

Underlying diseases

90 Days Prior

Endocrine and Metabolic Diseases

0.0001 0.0005 0.5901

Diseases of the Circulatory System

<0.0001 <0.0001 0.1253

Diseases of the Respiratory System

<0.0001 0.0710 0.1919

Diseases of the Circulatory System

<0.0001 <0.0001 0.1253

Diseases of the Urinary/Reproductive

Organs <0.0001 0.0020 0.0034

Table 3. Results of the Multivariate Logistic Regression Analysis comparing the ESBL case

group with the two other case groups. These tests were performed as previously described for

case groups compared to the uninfected control group. Here we, in turn, compared the ESBL

group to each of the two other case groups. *P-values found by chi-square analyses. **P-

values found by manual backward stepwise elimination analysis.

ESBL vs. Resistant

ESBL vs. Susceptible

Variables P-Value P-Value

HCAI vs. COI 0.1660* 0.0058*

Sex (Female) 0.4335 0.4606

Age 0.4387 0.1837

30 Days Prior to index sample

Admissions 0.0073 0.0216

Trimethoprim 0.0298 0.9998

Sulphamethizole 0.0381 0.0299

Fluoroquinolones 0.0365** 0.25**

Total antibiotics 0.9194** 0.04**

90 Days Prior to index sample

Total antibiotics 0.0049 0.0029

Insulin 0.3218 0.0447

Nitrofurantoin 0.0089 0.1840

Sulphamethizole 0.0010 0.0091

Trimethoprim 0.0096 0.4663

Table 4. The ratio between patients and numbers of urine samples submitted to Dept. of

Clinical Microbiology from general practices from 365 days prior to and within 90 days after

sampling. The number of urine samples submitted within one year prior to sampling until 90

days after has been evaluated and counted for each individual patient. All isolates found in

urine are submitted to susceptibility testing towards cefpodoxime (CPD). Resistance to CPD is

used as an indication of ESBL production and leads to MAST-testing, results of which we do

not have. We use CPD-R as a possibility of ESBL-production, with the limitations it includes.

We show the ratio between total number of urine samples, number of positive samples, sum of

samples positive for E.coli and finally number of E.coli resistant to CPD divided to patients

included in each group. *P<0.05 compared to the uninfected group. ▪P<0.05 compared to the

resistant group. °P>0.05 compared to the susceptible group.

Case groups No. of urine samples

No. of positive samples (%)

No. of samples with E.coli (%)

No. of CPD Resistant E.coli (%)

ESBL (n=98) 3.8*° 3.5*° (91.2) 2* (56.3) 1.4*▪° (71.9)

Negative (n=200)

2 0.6 (33) 0.155 (24) 0.015 (9.7)

One year before index sample

Resistant (n=174)

3.4*° 3*° (90.6) 1.9* (64.1) 0.07*° (3.5)

Susceptible (n=177)

2.4 2.2* (91.5) 1.5* (68) 0

90-0 days before index sample

ESBL (n=98) 2* 1.9* (93.4) 1.3* (68.9) 1.1*▪° (57.4)

Negative (n=200)

1.5 0.4 (26.1) 0.11 (28.6) 0.02 (3.9)

Resistant (n=174)

2* 1.9* (96.2) 1.3* (69.8) 0.05° (2.4)

Susceptible (n=177)

1.7 1.6* (94.9) 1.2* (76.5) 0

0-90 days after index sample

ESBL (n=98) 1.4*° 1.2*° (83) 0.6*° (51.3) 0.5*▪° (80)

Negative (n=200)

0.4 0.2 (48.1) 0.05 (25.6) 0

Resistant (n=174)

1*° 0.9*° (88.8) 0.4*° (48.5) 0.02* (5.2)

Susceptible (n=177)

0.7* 0.6* (81.5) 0.3* (43.6) 0.01 (2.3)

1

“Population structure of Drug-Susceptible, -Resistant and ESBL-producing Escherichia coli

Populations from Community-Acquired Urinary Tract I nfections as Characterized by

Abbreviated MLVA, MLST and Serogrouping in Denmark”

Keywords: Escherichia coli; lineages; urinary tract infection; community-acquired; ESBL.

Running title: Characterization of E. coli according to susceptibility

(1,2) Frederik Boëtius Hertz, (1) Jesper Boye Nielsen, (1) Kristian Schønning (1) Pia Littauer, (1) Jenny Dahl

Knudsen, (2) Anders Løbner-Olesen and (3) Niels Frimodt-Møller

1) Dept. of Clinical Microbiology, Hvidovre Hospital, Denmark

2) Dept. of Biology, University of Copenhagen, Denmark

3) Dept. of Clinical Microbiology, Rigshospitalet, Denmark

Corresponding author:

Frederik B. Hertz, MD

Hvidovre University Hospital

Dept. of Clinical Microbiology

Copenhagen

004538626971

[email protected]

2

Abstract

Background: Escherichia coli is the most common cause of urinary tract infection in all age

groups. The pathogenic isolates are becoming increasingly resistant to antibiotics; with a worldwide

dissemination of resistant sequence types (ST). We characterized three different uropathogenic

E.coli (UPEC) populations, from non-hospitalized patients, by use of three typing methods, to

describe the genetic kinship between resistant and susceptible isolates.

Methods: We used MLST, a-MLVA, O-serogrouping and ESBL-genotyping. Urine samples

submitted for testing, by general practitioners, were identified at Dept. of Clinical Microbiology at

Hvidovre Hospital, Denmark, from Oct. 2011 to July 2012.

Results: We included 94 fully susceptible, 94 resistant (non-ESBL) and 98 ESBL-producing E.coli

isolates. The ESBL population was dominated by serogroup O25 constituting 39% of isolates. In

the ESBL population 51% belonged to ST131, 9% to ST38 and 6% to ST69. In the resistant group

ST69, ST73 and ST131 were the largest clusters with 18%, 11% and 15%, respectively. The

susceptible population had significantly more STs and a-MLVA codes than the other groups and

ST73 and ST95 were found as the only clusters with 16% and 6%, respectively. Ninety-eight per

cent of the ESBL-producing E.coli isolates were CTX-M-producers, 54% produced CTX-M-15.

Conclusion: Sequence type 131 dominated the population of community-associated uropathogenic

ESBL-producing E.coli, but was less frequent among non-ESBL-producing E.coli. The fully

susceptible E.coli population was a much more diverse group than the resistant and ESBL-

producing E.coli populations. Over findings suggest that dominant ESBL-producing lineages are

derived from UPEC lineages already established in the general population.

3

Introduction

Escherichia coli is the most common Gram-negative extraintestinal pathogen and the primary cause

of urinary tract infection (UTI) in all age groups (1,2). It is a highly heterogonous species with

certain lineages becoming increasingly resistant to antibiotics (2–5). Thus, resistant E.coli isolates

are emerging worldwide, including E.coli producing extended-spectrum beta-lactamase (ESBL),

now primarily producing the CTX-M type ESBLs (2,6). This dissemination of ESBL-producing

E.coli in hospital settings as well as in the community has been reported as spread of extraintestinal

pathogenic E.coli (ExPEC) belonging to a limited number of lineages or sequence types (STs) (2,6).

However, ExPEC lineages, like ST73 and ST95 continue to be common causes of primarily urinary

tract infection (UTI), but are rarely multidrug resistant and seldom associated with ESBL

production (7–10). There is a need to describe the prevalent lineages among non-ESBL-producing

E.coli to recognize if ESBL-producing E.coli lineages are identified in all populations or are

relatively newly emerged ExPEC (3,11). Different typing methods have previously sub-typed many

of the frequent E.coli ST-lineages and an abbreviated-MLVA (a-MLVA) method has been

evaluated and proved efficient in the characterization of ESBL-producing E.coli and the method is

congruent with multi-locus sequence typing (MLST) (12). However, only a few studies have

specifically studied the heterogeneity among susceptible, resistant and ESBL-producing E.coli

using several typing methods (2–4,6).

In the present study we characterize three different E.coli populations, all from non-hospitalized

patients, by use of O-serogrouping, a-MLVA and subsequent MLST of a-MLVA codes found.

MLST was performed to link a-MLVA codes to an international known nomenclature and we chose

urine samples from general practices to describe prevalence in an environment with reduced

selection pressure, compared to hospital settings. Hereby, we describe differences and kinship

among E.coli populations to assess if the spread of resistant E.coli are caused by successful

4

uropathogenic E.coli (UPEC) or other less successful UPEC with newly gained resistance (13).

Differences in dominating E.coli lineages among the populations would indicate that resistance and

ESBL-production generates selective advantages in certain bacterial populations and environments.

Materials and Methods

Strain collections

The Department of Clinical Microbiology at Hvidovre Hospital, Denmark (DCM) provides services

to more than 450 general practitioners in an area of approximately 950.000 inhabitants. From the 1st

of October 2011 until the 30th of June 2012 we collected 449 E.coli isolates from urine samples,

submitted from general practices, as part of an on-going case-control study. Hence, from non-

hospitalized patients, one unique strain was collected per patient and three susceptibility groups

were included; (i) ESBL-producing E.coli, (ii) E.coli resistant to at least one tested antibiotic (non-

ESBL) and (iii) fully susceptible E.coli. O-serogrouping was performed by GlycoVaxyn on the 94

ESBL-positive isolates as well as on 49 resistant and 51 susceptible isolates available at time of

serogrouping. From the complete case-control collections of isolates, we randomly chose isolates

for characterization by MLVA and MLST. Not all isolates or patients had been thoroughly

identified at the time of typing: First we included all isolates with a serogroup from GlycoVaxyn

and we included the remaining four ESBL-producing E.coli isolates. Secondly, we identified

resistant and susceptible isolates from patients with feasible community-onset infections (COI) and

randomly chose 45 and 43 isolates, respectively. Thus, in this study we included all 98 ESBL-

producing E.coli isolates, 94 isolates resistant (non-ESBL) isolates and 94 fully susceptible E.coli.

5

Strain identification and susceptibility testing

The UPEC isolates were all identified at species level by MALDI-TOF MS (Bruker, Germany) and

antimicrobial susceptibility testing was performed by Disk Diffusion Test Methodology as

described in the European Committee on Antimicrobial Susceptibility Testing (EUCAST, Version

1.0, December 18, 2009) or by The Vitek 2 automated system by use of cards AST-N209 and AST-

N122 (bioMérieux, France). Methods were performed according to guidelines of DCM in

agreement with direction from manufacturers and as described elsewhere (14–17). The ATCC

25922 E.coli was used for quality control and susceptibility interpreted as recommended by

EUCAST (www.eucast.org/clinical_breakpoints/). Isolates denoted as I (intermediately resistant)

were reported as resistant. ESBL phenotypes were identified by a double-disk diffusion method,

here performed as combined-disk diffusion using a AmpC + ESBL detection set (MAST®,

Merseyside, UK) as previously described (16).

Molecular characterization

Phylogrouping

The major phylogroups (group A, B1, B2 and D) were determined using singleplex PCR

amplification of three genes (chuA, yjaA and TSPE4.C2) (18,19). Strains with no detected PCR

products were classified as non-typeable (NT) (19,20).

ESBL-genotyping

For all ESBL-producing E.coli, phenotypically recognized by the double-disk diffusion MAST®

test, the ESBL genotypes were identified by PCR and subsequent sequencing. Isolates were

screened for the presence of blaCTX-M genes by multiplex PCR assay, detecting alleles encoding

6

the five CTX-M groups 1, 2, 8, 9 and 25 (12,21). Positive samples were re-amplified and products

sequenced to identify the exact genotypes (12,21).

MLVA and MLST

All included isolates were typed using a-MLVA, with PCR amplification of six variable number of

tandem repeats (VNTR) loci, as described by Nielsen et al. (12). In short, PCR were done as

singleplex PCR with unlabelled primers. Subsequent size determination were done using an

automated capillary electrophoresis system (QIAxcel, Qiagen) and a high-resolution cartridge (12).

Each of the VNTR loci was manually binned depending on size and assigned a number. As result

each isolate were given a six-digit a-MLVA code. For isolates without a measured band size, for a

VNTR locus, PCR amplification was performed twice for the given locus. The identified codes

were primarily translated to STs by an in-house library, but to further evaluate the a-MLVA and

confirm our findings, at least one isolate from each identified a-MLVA code were verified by

MLST, with the exception of one a-MLVA code. In groups with several isolates having identical a-

MLVA codes, we included isolates with extreme band sizes within bins, to effectively cohere

clusters. In the resistant and susceptible groups we furthermore included some isolates with

different O-serogroups. In all, MLST was performed on 31 isolates from the ESBL population, on

45 isolates from the resistant population and on 52 isolates from the susceptible population. MLST

was done using the Achtmann MLST scheme with PCR and sequencing of seven housekeeping

genes followed by assignment of an allelic number from the MLST-database

(http://mlst.warwick.ac.uk/mlst/dbs/Ecoli) (12,22).

7

Statistics

For statistical analysis categorical variables were compared by a Chi-square or Fisher exact test

when appropriate and p<0.05 was considered significant.

Results

For each of the three typing methods used in this investigation, we defined a cluster as three or

more isolates identified as the same O-serogroup, ST or a-MLVA code.

Characterization of the E.coli populations

The distributions of serogroups, a-MLVA codes and STs in the three populations are found in

Figure 1.

O-Serogroup distribution

We identified several serogroups among the selected 194 isolates; 24 different serogroups among

the 94 ESBL-producing E.coli, 17 among the 49 resistant E.coli and 27 among the 51 susceptible

E.coli isolates. Interestingly, we found dissimilar patterns of dominating serogroups in the three

populations. The five largest clusters in the ESBL population constituted 57% of all isolates.

Furthermore, 39% belonged to one large cluster of serogroup O25, constituting the largest cluster

found in any of the populations. The five major clusters in the resistant population amounted to

25% of isolates and in the susceptible population the two largest clusters constituted 7% of isolates

only. In addition, several serogroups with one or two isolates were identified in each group.

Characterization by MLVA and MLST

Results for the a-MLVA are presented in Table 1, 2 and 3.

8

PCR amplification of the six MLVA alleles was successfully performed on all isolates. A total of 83

unique a-MLVA codes were identified among the 286 isolates. Seventy four codes were associated

to one unique sequence type. There were, however, several occasions where isolates belonging to

one ST were subdivided into more than one a-MLVA codes. As examples, ST69 was subdivided by

three different a-MLVA codes, ST73 subdivided by ten different codes and ST131 was subdivided

by four different codes (Table 1-3).

Results from the MLST are likewise presented in Table 1-3. We found 72 different sequence types

in the 141 selected isolates from the three populations. We discovered ten STs, not previously

identified, here labelled “New ST”. Complex situations existed where the a-MLVA method did not

distinguish between two or more STs. Thus for example, ST38, ST117 and ST1177 were assigned

same a-MLVA code. ST38 and ST1177 are single locus variant with ST117 being unrelated.

A single a-MLVA code was likewise assigned to ST10, ST746, ST1598 and one New ST (Table 1-

3). Here ST10 and ST746 are single-locus variants with the new ST being a double-locus variant of

ST10. For a total of eight a-MLVA codes it was not possible to discriminate between isolates of

different STs. Phenotypic relations were tested by use of eBURST, on online tool displaying the

relationships between closely-related isolates (http://eburst.mlst.net/v3), which showed that the

majority of STs in each population were unrelated, with only a few isolates being single locus

variants (data not shown).

In the ESBL population we found 20 a-MLVA codes and 20 different STs. We found no unknown

ST´s in this population, but had one isolate unsuccessfully typed. This isolate belonged to a a-

MLVA cluster identified by MLST performed on other isolates. The population was dominated by a

few lineages with seven a-MLVA clusters and five clear ST-clusters. Fifty isolates belonged to

ST131 with code “153562” containing 44 isolates. Of the 44 isolates 35 belonged to the serogroup

O25 and phylogroup B2. Of the isolates identified as ST131 by the other a-MLVA codes, in this

9

population, five belonged to serogroup O16 and one to O153. Overall, the ESBL population had

significantly more isolates belonging to ST131 than the two other populations (p<0.0001 and p

=0.0001). ST73 and ST95 were not found among the ESBL-producing isolates.

In the resistant E.coli population, 36 a-MLVA codes and 30 ST´s were identified with a single a-

MLVA code for which MLST was not performed. These codes and STs were grouped in eight a-

MLVA- and eight ST-clusters. Here ST69, ST73 and ST131 amounted to the largest ST clusters

with 18%, 11% and 15%, respectively.

In the susceptible E.coli population 54 a-MLVA codes and 44 ST´s were found with seven a-

MLVA- and ten ST clusters. The susceptible population had significantly more STs and a-MLVA

codes compared to the ESBL population (p =0.0097 and p =0.0005). ST73 and ST95 dominated

with 16% and 6%, respectively, and ST73´s were sub-divided by ten a-MLVA codes.

Overall, ST131 constituted 23% of all isolates, ST69 constituted 10% and ST73 a total of 9%.

During the time period of sampling four per cent of E.coli isolates, from urine samples from general

practices at Hvidovre Hospital, were resistant to cefpodoxime, used as an indicator for ESBL-

production.

Antibiotic resistance patterns

Results of antibiotic susceptibility testing are found in Figure 2 and 3. Generally the ESBL

population showed high levels of resistance with 100% of the isolates showing resistance towards

three or more of the tested antibiotics. In the resistant population 66% of the isolates were resistant

to >3 of the antibiotics. All isolates were susceptible to meropenem and most to mecillinam (97-

99%), fosfomycin (96-97%), nitrofurantoin (93-96), and piperacillin/tazobactam (92-95%). Table 2

shows that resistant, non-ESBL ST131 have a broader spectrum of resistance than resistant (non-

ESBL) ST73 and ST69.

10

Phylogrouping of E.coli isolates

The distribution of phylogroup A, B1, B2, D and non-typeables (NT) were similar among the three

populations of uropathogenic E.coli. Phylogroup A was found in 7-9%, phylogroup B1 in 1-6%,

phylogroup B2 in 45-58%, phylogroup D in 22-32% and NT´s were found in 4-11% of isolates in

the three populations, respectively.

ESBL-genotyping

We found that 98% of the ESBL-producing E.coli isolates were positive for the presence of a

blaCTX-M. blaCTX-M group 1 dominated with 73% of all isolates carrying this type. Furthermore, 54%

of all isolates produced CTX-M-15. A total of 24% of isolates belonged to CTX-M group 9 with

two large clusters of CTX-M-14 (12%) and CTX-M-27 (12%).

Discussion

This study was carried out to describe the diversity among susceptible, resistant (non-ESBL) and

ESBL-producing E.coli using three typing methods (13). The objective was to clarify if E.coli

isolates from these dissimilar populations are genetically related. This would determine if ESBL are

carried by current successful UPEC gaining resistance or spread by less efficient UPEC effective in

the presence of antibiotic. Overall we found ST131, ST73 and ST69 to be the dominating lineages

among all isolates, in accordance with previous studies (23). Of these, non-ESBL-producing ST131

seemed to have a broader spectrum of resistance than ST73 and non-ESBL-producing ST69, as

described by Horner et al. (Figure 3) (23). However, based on serogroups, a-MLVA and MLST, in

this collection of uropathogenic E.coli from general practices, we found that ESBL-producing

E.coli, resistant E.coli and susceptible E.coli in turn were dominated by different serogroups, a-

MLVA codes and STs. In general, the susceptible E.coli population was a much more diverse group

11

of isolates with more STs and fewer and smaller clusters. Here the biggest ST-lineage, ST73, was

extensively sub-divided by a-MLVA indicating high diversity among these isolates.

The resistant E.coli and ESBL-producing E.coli were found in larger clusters. Especially the ESBL-

population was a more homogenous population with fewest a-MLVA codes and STs. The STs

found differed to some extent from those of the susceptible population with an overlap of some a-

MLVA codes and STs among the three populations. The prevalence of STs were, however,

different. We found that ST131 completely dominated this ESBL-producing E.coli population with

the combination of serogroup O25, phylogroup B2 and ST131 being by far the largest cluster, as

previously seen in the Copenhagen area (12,24). A single ST131 isolate was found among the

susceptible isolates and 14 detected in the resistant population and still ST131 was able to

effectively dominate the ESBL-producing UPEC. The presence of ESBL might not cause higher

levels of virulence or fitness in environments with limited use of antibiotics, to which the ESBL-

producing E.coli are resistant. Thus, ESBL-producing STs does not necessarily hold an advantage

in the community in Denmark, where use of extended spectrum cephalosporins, ciprofloxacin and

aminoglycosides are limited, but can spread in settings were these groups of antibiotics are

intensely used. Nonetheless, ST131 seems to be a specialized UPEC adapted as an efficient resistant

and ESBL-producing lineage, sustainable in the community once present, at least for some time. Of

the resistant ST73 just four out of ten were resistant >3 antibiotics, an absence of multidrug

resistance also seen in previous studies (2,6–10). Furthermore, our result indicates that resistance to

one or few antibiotics creates some positive selection in the community or among patients from

general practices. ST131 could be one of the UPEC lineages, among these resistant and ESBL-

producing isolates, with the highest ability to colonize the human gut, creating a high prevalence in

these populations. Urinary tract infections are often caused by the dominating ExPEC already

present in the gut of the patient and ST131, and other less successful ESBL-producing UPEC-

12

lineages, might be found to cause UTI simply since they dominate the microflora during and after

antibiotic exposure (25). Less prevalent ESBL-producing ST-lineages could be less effective

UPEC, allowing for the few specialized UPEC actually carrying ESBL, like ST131, to fiercely

dominate the population. We speculate if resistance in different E.coli populations are somewhat

defined by intrinsic differences in distinct E. coli lineages, making a limited number of UPEC ST-

lineages capable of obtaining and spreading blaCTX-M. Some lineages successfully acquire and

maintain different types of mobile resistance genes like ST69 and especially ST131, while other

E.coli lineages remains fairly susceptible and rarely take up plasmids, as seen with ST73 and ST95

(3,4). However, productions of ESBLs are found among specific UPEC present, to some, extent in

all populations. Antibiotic selection therefore creates a less varied population structure of related

isolates while antibiotic free environments allows for competition and diverse non-related

population structure (26). This is seen as difference in diversity of O-serogroups, a-MLVA codes

and STs in the three populations. The UPEC we found in our three populations have been found in

other population-studies of E.coli isolates causing bacteremia, strongly indicating that E.coli

isolates capable one invasive disease can cause severe invasive infections, independent of

susceptibility (23).

The a-MLVA and MLST had very high typeability and showed somewhat similar discriminatory

power dividing the populations into 83 a-MLVA codes and 72 STs, respectively. Though, MLVA

seemed slightly more discriminatory; subdividing several STs. There is a higher rate of sustainable

genetic variations within the VNTR regions used in MLVA, than in the housekeeping genes used

for MLST. However, a change in a VNTR region does not necessarily lead to a change in ST, and

vice versa. This poses a challenge in identifying isolates with new VNTR-sequences, here seen as

limitations in incomplete separation of all STs, but it also provides high discriminatory power in the

ability to separate closely related ST-lineages and thereby describe E.coli populations in detail

13

(27,28). Some of the undistinguishable STs was found to share a-MLVA codes by Nielsen et al,

such as ST101 and ST448 (12). The missing identification of variation in STs are, however,

unlikely to cause bias in our overall conclusion as the prevalence and identification of the major

lineages in the three populations are unique. The a-MLVA typing method was successful in the

characterization and sub-division of some large cluster of ST-lineages (Table 1-3). Thus, ST69 was

the largest cluster in the resistant population and found in all populations with three different a-

MLVA codes. ST73 and ST95 amounted for larger clusters in both the resistant and susceptible

populations and here the a-MLVA method could describe the STs as heterogeneous lineages, by

classifying each lineage by different a-MLVA codes. Finally, the lineage of ST131 was sub-divided

by a-MLVA. All isolates identified as ST131 belonged to phylogroup B2, but we detected a large

ST131 cluster with one a-MLVA code primarily belonging to O25, all of which were resistant to

ciprofloxacin. A minor ST131 cluster, with another a-MLVA code belonging to O16, with no

isolates showing resistance to ciprofloxacin, was likewise detected. This corresponds well with

reports that most ST131 isolates belong to serogroup O25 and a minor part being identified as O16

(24,29). Furthermore, sub-division of other ST-lineages have successfully been done by fimH

sequencing and more discriminatory typing method like PFGE and our results are in line with the

reports identifying susceptible ST-lineages as more heterogeneous populations (2,6,29,30). This is

in clear contradiction to what has previously been stated, in a study based on sequence typing only,

where all E.coli isolates were found to be of clonal distribution, disregarding resistance (3).

The study is limited by the relatively small number of isolates characterized and the limited time

period of collection, which does not allow us to identify any fluctuation in dominant lineages of

non-ESBL populations. Furthermore, the ESBL-population is a more selected group, allowing us to

detect less successful STs with limited impact as UPEC. As all isolates are from general practices, it

14

would have been beneficial to precisely know to what extent UTIs were devoid of healthcare

association.

Nonetheless, our results strongly suggest that the observed dissemination of ESBL-producing

E.coli are due to the spread of certain UPEC lineages already present in the general UPEC

population (3). Less prevalent ESBL-producing E.coli might be non-specialized UPEC, why they

are not found in the non-ESBL populations. In contrast, lineages like ST69 and ST131 are UPEC

present in all populations and there able to dominate in the ESBL-population. It seems that the level

of virulence could be of limited epidemiological importance for the spread of resistant E.coli, since

this most likely occurs due to selection of previously specialized UPEC with limited fitness loss due

to ESBL-production (31).

Acknowledgements

Work has been performed as cooperation between HVH, Roskilde University and University of Copenhagen with

laboratory facilities provided by HVH. This work was, in parts, presented at ECCMID in Barcelona 2014.

Funding

This work was supported by the EU-FP funded project PAR7, DanCARD (Danish National Strategic Research

Foundation, project 09-067075/DSF), Roskilde University, University of Copenhagen and the SSAC

Foundation (SLS-327421 and SLS-251761).

Transparency declarations

None to declare.

15

1. Croxall G, Hale J, Weston V, Manning G, Cheetham P, Achtman M, et al. Molecular

epidemiology of extraintestinal pathogenic Escherichia coli isolates from a regional cohort of

elderly patients highlights the prevalence of ST131 strains with increased antimicrobial

resistance in both community and hospital care settings. 2011;(August):2501–8.

2. Banerjee R, Johnston B, Lohse C, Chattopadhyay S, Tchesnokova V, Sokurenko E V, et al.

The clonal distribution and diversity of extraintestinal Escherichia coli varies according to

patient characteristics. Antimicrob Agents Chemother. 2013 Sep 16;(September):1–29.

3. Adams-Sapper S, Diep BA, Perdreau-Remington F, Riley LW. Clonal composition and

community clustering of drug-susceptible and -resistant Escherichia coli isolates from

bloodstream infections. Antimicrob Agents Chemother. 2013 Jan;57(1):490–7.

4. Bengtsson S, Naseer U, Sundsfjord A, Kahlmeter G, Sundqvist M. Sequence types and

plasmid carriage of uropathogenic Escherichia coli devoid of phenotypically detectable

resistance. J Antimicrob Chemother. 2012 Jan;67(1):69–73.

5. Nicolas-Chanoine M-H, Blanco J, Leflon-Guibout V, Demarty R, Alonso MP, Caniça MM,

et al. Intercontinental emergence of Escherichia coli clone O25:H4-ST131 producing CTX-

M-15. J Antimicrob Chemother. 2008 Feb;61(2):273–81.

6. Blanco J, Mora A, Mamani R, López C, Blanco M, Dahbi G, et al. National survey of

Escherichia coli causing extraintestinal infections reveals the spread of drug-resistant clonal

groups O25b:H4-B2-ST131, O15:H1-D-ST393 and CGA-D-ST69 with high virulence gene

content in Spain. J Antimicrob Chemother. 2011 Sep;66(9):2011–21.

7. Johnson JR, Tchesnokova V, Johnston B, Clabots C, Roberts PL, Billig M, et al. Abrupt

emergence of a single dominant multidrug-resistant strain of Escherichia coli. J Infect Dis.

2013 Mar 15;207(6):919–28.

16

8. Mora A, Blanco M, López C, Mamani R, Blanco JE, Alonso MP, et al. Emergence of clonal

groups O1:HNM-D-ST59, O15:H1-D-ST393, O20:H34/HNM-D-ST354, O25b:H4-B2-

ST131 and ONT:H21,42-B1-ST101 among CTX-M-14-producing Escherichia coli clinical

isolates in Galicia, northwest Spain. Int J Antimicrob Agents. 2011 Jan;37(1):16–21.

9. Rogers B a, Sidjabat HE, Paterson DL. Escherichia coli O25b-ST131: a pandemic,

multiresistant, community-associated strain. J Antimicrob Chemother. 2011 Jan;66(1):1–14.

10. Gibreel TM, Dodgson AR, Cheesbrough J, Fox AJ, Bolton FJ, Upton M. Population

structure, virulence potential and antibiotic susceptibility of uropathogenic Escherichia coli

from Northwest England. J Antimicrob Chemother. 2012 Feb;67(2):346–56.

11. Foxman B. Extended-spectrum β-lactamase-producing escherichia coli in the united states:

time to rethink empirical treatment for suspected E. coli infections? Clin Infect Dis. 2013

Mar;56(5):649–51.

12. Nielsen JB, Albayati A, Jørgensen RL, Hansen KH, Lundgren B, Schønning K. An

abbreviated MLVA identifies Escherichia coli ST131 as the major extended-spectrum β-

lactamase-producing lineage in the Copenhagen area. Eur J Clin Microbiol Infect Dis. 2013

Nov 6;32(3):431–6.

13. Köhler C-D, Dobrindt U. What defines extraintestinal pathogenic Escherichia coli? Int J Med

Microbiol. 2011 Dec;301(8):642–7.

14. Livermore DM, Struelens M, Amorim J, Baquero F, Bille J, Canton R, et al. Multicentre

evaluation of the VITEK 2 Advanced Expert System for interpretive reading of antimicrobial

resistance tests. J Antimicrob Chemother. 2002 Feb;49(2):289–300.

15. Nyberg SD, Meurman O, Jalava J, Rantakokko-Jalava K. Evaluation of detection of

extended-spectrum beta-lactamases among Escherichia coli and Klebsiella spp. isolates by

17

VITEK 2 AST-N029 compared to the agar dilution and disk diffusion methods. Scand J

Infect Dis. 2008 Jan;40(5):355–62.

16. Hansen F, Olsen SS, Heltberg O, Justesen US, Fuglsang-Damgaard D, Knudsen JD, et al.

Characterization of Third-Generation Cephalosporin-Resistant Escherichia coli from

Bloodstream Infections in Denmark. Microb Drug Resist. 2014 Feb 11;00(00):1–9.

17. Haigh J, Degun A, Eydmann M, Millar M, Wilks M. Improved performance of bacterium

and yeast identification by a commercial matrix-assisted laser desorption ionization-time of

flight mass spectrometry system in the clinical microbiology laboratory. J Clin Microbiol.

2011 Sep;49(9):3441.

18. Clermont O, Bonacorsi S, Bingen E. Rapid and simple determination of the Escherichia coli

phylogenetic group. Appl Environ Microbiol. 2000 Oct;66(10):4555–8.

19. Gordon DM, Clermont O, Tolley H, Denamur E. Assigning Escherichia coli strains to

phylogenetic groups: multi-locus sequence typing versus the PCR triplex method. Environ

Microbiol. 2008 Oct;10(10):2484–96.

20. Clermont O, Christenson JK, Denamur E, Gordon DM. The Clermont Escherichia coli

phylo-typing method revisited: improvement of specificity and detection of new phylo-

groups. Environ Microbiol Rep. 2013 Feb;5(1):58–65.

21. Jørgensen RL, Nielsen JB, Friis-Møller A, Fjeldsøe-Nielsen H, Schønning K. Prevalence and

molecular characterization of clinical isolates of Escherichia coli expressing an AmpC

phenotype. J Antimicrob Chemother. 2010 Mar;65(3):460–4.

22. Wirth T, Falush D, Lan R, Colles F, Mensa P, Wieler LH, et al. Sex and virulence in

Escherichia coli: an evolutionary perspective. Mol Microbiol. 2006 Jun;60(5):1136–51.

18

23. Horner C, Fawley W, Morris K, Parnell P, Denton M, Wilcox M. Escherichia coli

bacteraemia: 2 years of prospective regional surveillance (2010-12). J Antimicrob

Chemother. 2014 Jan;69(1):91–100.

24. Olesen B, Hansen DS, Nilsson F, Frimodt-Møller J, Leihof RF, Struve C, et al. Prevalence

and Characteristics of the Epidemic Multiresistant Escherichia coli ST131 Clonal Group

among Extended-Spectrum Beta-Lactamase-Producing E. coli Isolates in Copenhagen,

Denmark. J Clin Microbiol.2013 Jun;51(6):1779–85.

25. Nielsen KL, Dynesen P, Larsen P, Frimodt-Møller N. Faecal Escherichia coli from patients

with E. coli urinary tract infection and healthy controls who have never had a urinary tract

infection. J Med Microbiol. 2014 Apr;63(Pt 4):582–9.

26. Maiden MCJ. Multilocus sequence typing of bacteria. Annu Rev Microbiol. 2006

Jan;60:561–88.

27. Keys C, Kemper S, Keim P. Highly diverse variable number tandem repeat loci in the E. coli

O157:H7 and O55:H7 genomes for high-resolution molecular typing. J Appl Microbiol. 2005

Jan;98(4):928–40.

28. Manges AR, Tellis P a, Vincent C, Lifeso K, Geneau G, Reid-Smith RJ, et al. Multi-locus

variable number tandem repeat analysis for Escherichia coli causing extraintestinal

infections. J Microbiol Methods. Elsevier B.V.; 2009 Nov;79(2):211–3.

29. Peirano G, van der Bij AK, Gregson DB, Pitout JDD. Molecular epidemiology over an 11-

year period (2000 to 2010) of extended-spectrum β-lactamase-producing Escherichia coli

causing bacteremia in a centralized Canadian region. Journal of clinical microbiology. 2012.

p. 294–9.

19

30. Johnson JR, Tchesnokova V, Johnston B, Clabots C, Roberts PL, Billig M, et al. Abrupt

emergence of a single dominant multidrug-resistant strain of Escherichia coli. J Infect Dis.

2013 Mar 15;207(6):919–28.

31. Bengtsson S, Naseer U, Sundsfjord A, Kahlmeter G, Sundqvist M. Sequence types and

plasmid carriage of uropathogenic Escherichia coli devoid of phenotypically detectable

resistance. J Antimicrob Chemother. 2012 Jan;67(1):69–73.

20

Figure 1. Here we show the percentages of found a-MLVA codes, STs and O-serogroups found in each of the three populations.

Percentage of identifed a-MLVA codes,STs and O-groups

MLVA

MLST

Serogro

ups0

20

40

60

80

100

120

140

160 Susceptible Resistant ESBL

Typing methods

21

Figure 2.

Percentage of Isolates Resistant to antibiotics

Ampicillin

Cefuroxime

Aztreonam

Ciprofloxacin

*Sulfamethoxazole

Trimethroprim

*Tetracycline

Ampicillin/Clavulanic acid

Gentamicin

*Piperacillin/Tazobactam

Nitrofurantoin

Mecillinam

*Fosfomycin

Meropenem ESBLResistant

22

Figure 3. Percentages of isolates resistant to chosen antibiotics used for oral administration. We show resistance patterns for three common UPEC ST lineages, here all isolates are non-ESBL-producers but present in either the susceptible or resistant population. N: ST131 = 17, ST73 = 25, ST69 = 20.

Resistance patterns for non-ESBL UPEC lineages

0 20 40 60 80 100 120 140 160 180 200

Ampicillin

Ciprofloxacin

*Sulfamethoxazole

Trimethroprim

Nitrofurantoin

Mecillinam

*Fosfomycin

Resistance to >3 antibiotics

Fully susceptible

ST131ST73ST69

23

Table 1. Distribution of sequence types (STs) in relation to assigned a-MLVA codes and serogroups among ESBL-producing E.coli. We show the serogroups found for the given a-MLVA code. M = Multiple serogroups, N = serogroup negative. Isolates with a written ST are the isolates tested by MLST.

ST a-MLVA Serogroup Phylogroup ESBL Genotype

ESBL 14 123645 75 B2 TEM-1

ESBL 38 173050 2 D CTX-M-14

ESBL 38 173050 M D CTX-M-14

ESBL 38 173050 M D CTX-M-14

ESBL 38 173050 153 D CTX-M-14

ESBL 173050 M D CTX-M-14

ESBL

173050 M D CTX-M-14

ESBL 173050 161 D CTX-M-14

ESBL

173050 86 D CTX-M-14

ESBL 173050 153 D CTX-M-14

ESBL 173050 153 D CTX-M-14

ESBL 62 161370 MISSING D CTX-M-14

ESBL 69 173277 15 D CTX-M Gr. 1

ESBL

173277 15 D CTX-M Gr. 1

ESBL 173277 15 D CTX-M-79/55

ESBL

173277 15 D CTX-M-27

ESBL 173277 44 D TEM-1

ESBL 173277 73 D CTX-M-14

ESBL 88 132261 119 A CTX-M-1

ESBL 120 132271 N Non-Type CTX-M-15

ESBL 131 103562 16 B2 CTX-M-14

ESBL 131 153562 25 B2 CTX-M Gr. 1

ESBL 131 153562 25 B2 CTX-M-15

ESBL 131 153562 77 B2 CTX-M-15

ESBL Unsuccessful 153562 97 B2 CTX-M-27

24

ESBL 153562 25 B2 CTX-M-27

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-28

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-14

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-28

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-27

ESBL

153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-28

ESBL 153562 25 B2 CTX-M-27

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M 79/55

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-27

ESBL 153562 25 B2 CTX-M-15

25

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 25 B2 CTX-M-15

ESBL 153562 25 B2 CTX-M-15

ESBL

153562 M B2 CTX-M-27

ESBL 153562 M B2 CTX-M-15

ESBL

153562 M B2 CTX-M-15

ESBL 153562 M B2 CTX-M-15

ESBL

153562 MISSING B2 CTX-M-28

ESBL 153562 MISSING B2 CTX-M-27

ESBL 153562 N B2 CTX-M-28

ESBL 131 163562 16 B2 CTX-M-15

ESBL 131 163562 16 B2 CTX-M-15

ESBL 163562 16 B2 CTX-M-15

ESBL

163562 16 B2 CTX-M-1

ESBL 163562 153 B2 CTX-M-15

ESBL 224 131271 M B1 CTX-M-15

ESBL 315 173552 25 B2 CTX-M-14

ESBL 354 151170 153 D CTX-M-14

ESBL 428 252365 117 B2 CTX-M-1

ESBL 617 132251 12 A CTX-M-15

ESBL 132251 69 A CTX-M-28/15

ESBL

132251 N A CTX-M-15

ESBL 132251 116,162 A CTX-M-15

ESBL

132251 N A CTX-M-79/55

ESBL 132251 N A CTX-M-15

ESBL 132251 12 A CTX-M-15

ESBL 636 293852 21 B2 CTX-M-15

ESBL 648 161160 1 D CTX-M-15

26

ESBL 648 161160 102,13 D CTX-M-15

ESBL

161160 153 D CTX-M-15

ESBL 161160 MISSING D CTX-M-15

ESBL 998 224665 2 B2 CTX-M-15

ESBL 998 224665 M B2 CTX-M-1

ESBL 2852 131291 8 B1 CTX-M-15

ESBL 131291 25 B2 CTX-M-15

ESBL 101 131261 N B1 CTX-M-1

ESBL 448 131261 111 B1 CTX-M-15

ESBL

131261 170 NT CTX-M Gr. 9

ESBL 131261 81 B1 CTX-M-79/55

ESBL

131261 100 NT CTX-M-1

ESBL 131261 M B1 CTX-M-15

ESBL 746 131251 21 A CTX-M-28

ESBL 1598 131251 9 NT CTX-M-15

Table 2. Distribution of sequence types (STs) in relation to assigned a-MLVA codes and serogroups, for the resistant group. Not all resistant isolates were characterized by serogroup. Here we show the serogroups found for the given a-MLVA code. M = Multiple serogroups, N = serogroup negative. Isolates with a written ST are the isolates tested by MLST.

ST a-MLVA Serogroup Phylogroup

Resistant 14 123645 75 B2

Resistant 38 173051 25 D

Resistant 58 131261 19 B1

Resistant

131261

B1

Resistant

131261

NT

Resistant

131261 N NT

27

Resistant

131261 69 B1

Resistant

131261

NT

Resistant 62 161370

D

Resistant 69 173270 73 D

Resistant

173270

D

Resistant 69 173277 15 D

Resistant 69 173277 25 D

Resistant

173277 11 D

Resistant

173277 11 D

Resistant

173277 15 D

Resistant

173277 15 D

Resistant

173277 25 D

Resistant

173277 68 D

Resistant

173277 73 D

Resistant

173277 73 D

Resistant

173277 73 D

Resistant

173277 73 D

Resistant

173277 M D

Resistant

173277

D

Resistant

173277

D

Resistant 73 176655

Non-Type

Resistant 73 266655 6 B2

Resistant 73 276655 6 B2

Resistant 73 276655 25 B2

Resistant 73 276655

B2

Resistant

276655 M B2

Resistant

276655

B2

Resistant

276655

B2

Resistant

276655

B2

28

Resistant

276655

B2

Resistant 80 254575 75 B2

Resistant 80 254575 75 B2

Resistant 88 132261 8 NT

Resistant 132261 NT

Resistant New ST 132261

NT

Resistant 95 223643 2 B2

Resistant 95 223653 2 Non-Type

Resistant

223653

Non-Type

Resistant 127 264953 6 B2

Resistant 131 143562

B2

Resistant 131 153562 25 B2

Resistant 131 153562

B2

Resistant

153562 25 B2

Resistant

153562 25 B2

Resistant

153562 25 B2

Resistant

153562

B2

Resistant

153562

B2

Resistant

153562

B2

Resistant

153562

B2

Resistant

153562

B2

Resistant 131 163562

B2

Resistant

163562

B2

Resistant

163562

B2

Resistant 135 225661

B2

Resistant 141 124665

B2

Resistant 362 143250

D

Resistant 372 224563 18 B2

29

Resistant 393 171577

D

Resistant 405 121250 M D

Resistant

121250

D

Resistant 405 131250

D

Resistant

131250

D

Resistant 457 161371 11 D

Resistant 648 161150 1 D

Resistant 978 226565 83 B2

Resistant 1193 124645 75 B2

Resistant 1193 124645 6 B2

Resistant 124645 B2

Resistant 124645 D

Resistant

124645

B2

Resistant

124645 75 B2

Resistant 1597 162562 39 B2

Resistant 1597 162562

B2

Resistant New ST 123552

B2

Resistant New ST 131251

NT

Resistant New ST 162438

NT

Resistant MISSING 266561

NT

Resistant 10 124643 75 B2

Resistant 124643 75 B2

Resistant 10 132251 A

Resistant 132251 3 A

Resistant 132251 A

Resistant

132251

A

Resistant

132251

A

Resistant

132251 N A

30

Resistant 2279 132251 15 A

Resistant 117 173050 M D

Resistant 1177 173050 D

Table 3. Distribution of sequence types (STs) in relation to assigned a-MLVA codes and serogroups for the susceptible group. Not all susceptible isolates were characterized by serogroup. Here we show the serogroups found for the given a-MLVA code. M = Multiple serogroups, N = serogroup negative. Isolates with a written ST are the isolates tested by MLST.

ST a-MLVA Serogroup Phylogroup

Susceptible 12 266562 4 B2

Susceptible 12 266562 B2

Susceptible 14 124645 18 B2

Susceptible 124645 M B2

Susceptible 38 253263 114 D

Susceptible 253263 N D

Susceptible 48 131281 Non-Type

Susceptible 131281 119 Non-Type

Susceptible 59 241370 D

Susceptible 59 251380 D

Susceptible 62 161370 D

Susceptible 69 103277 B1

Susceptible 69 173277 D

Susceptible 173277 D

Susceptible 173277 D

Susceptible 73 176655 18 B2

Susceptible 73 203655 2 D

Susceptible 73 206653 B2

Susceptible 73 206655 B2

Susceptible 73 226655 B2

Susceptible 73 256653 D

Susceptible 73 266655 2 B2

Susceptible 266655 B2

Susceptible 73 276555 25 B2

Susceptible 73 276655 6 B2

Susceptible 73 276655 22 B2

31

Susceptible 73 276655 M B2

Susceptible 276655 2 B2

Susceptible 276655 B2

Susceptible 73 276665 120 B2

Susceptible 80* 254575 B2

Susceptible 95 203653 B2

Susceptible 95 223653 1 B2

Susceptible 223653 1 B2

Susceptible 223653 M B2

Susceptible 223653 B2

Susceptible 223653 Non-Type

Susceptible 101 131261 174 Non-Type

Susceptible 131261 Non-Type

Susceptible 127 264953 B2

Susceptible 264953 6 B2

Susceptible 127 274953 6 B2

Susceptible 131 153562 B2

Susceptible 141 204665 B2

Susceptible 204665 D

Susceptible 162 211251 95 A

Susceptible 223 101261 B2

Susceptible 101261 Non-Type

Susceptible 101261 B2

Susceptible 357 150365 B2

Susceptible 357 152365 73 B2

Susceptible 152365 M B2

Susceptible 405 131250 2 D

Susceptible 410 132261 A

Susceptible 501 163777 D

Susceptible 538 152363 13 B2

Susceptible 582 274242 B2

Susceptible 589 287572 B2

Susceptible 681 224452 8 D

Susceptible 714 473050 D

Susceptible 1161 124675 B2

Susceptible 1331 274655 6 D

Susceptible 1444 273673 4 B2

Susceptible 1858 224645 6 B2

Susceptible 3672 226962 B2

Susceptible 4235 224743 88 B2

Susceptible 224743 88 B2

32

Susceptible New ST 1 151170 D

Susceptible New ST 5 112871 D

Susceptible New ST 6 272275 133 B2

Susceptible New ST 8 232251 105 A

Susceptible New ST 9 171250 D

Susceptible 10 131251 109 A

Susceptible 10 131251 M A

Susceptible 131251 107,117 A

Susceptible 10 132251 81 A

Susceptible 93 132251 7 Non-Type

Susceptible 132251 162 A

Susceptible 132251 N A

Susceptible 540 132251 15 Non-Type

Susceptible 540 132251 Non-Type

Susceptible New ST 3 132251 11 Non-Type

Susceptible 141 224665 2 D

Susceptible 141 224665 2 B2

Susceptible 141 224665 B2

Susceptible 998 224665 2 B2

Susceptible 998 224665 2 D

Susceptible 224665 B2

Susceptible 224665 2 B2

Susceptible 224665 138 D

Susceptible 224665 161 B2

Susceptible 224665 B2

Susceptible 420 254665 82 B2

Susceptible 3846 254665 B2