Download as pdf or txt
Download as pdf or txt
You are on page 1of 131

Proceedings of

The Lancet Conference on Healthcare-Associated Infections


London, UK, 11—12 December 2008
Guest Editor: Professor Kevin G. Kerr
The Official Journal of the Hospital Infection Society

Editor Editorial Coordinator Hospital Infection Society


Office
S. J. Dancer (Glasgow) N. Atherton
162 Kings Cross Road,
London WC1X 9DH
Tel: 020 7713 0273
Assistant Editors Editorial Board Editorial Advisers
Chief Executive
S. P. Barrett (London) P. Astagneau (France) G. Ayliffe (UK) Dr T. R. Logan
J. Bates (Worthing) T. S. J. Elliott (Birmingham) F. Daschner (Germany) President
A. Berrington (Sunderland) A. P. Fraise (Birmingham) G. L. French (London) Dr G. L. Ridgway
162 King’s Cross Road,
J. Child (Worthing) C. M. Fry (London) O. B. Jepsen (Denmark) London WC1X 9DH
R. Cooke (Liverpool) G. Gopal Rao (London) H. Kobayashi (Japan)
Chairman
A. Guyot (Guildford) K. Gould (Newcastle) S. Mehtar (South Africa) Dr A. P. Fraise
P. N. Hoffman (London) J.-J. Haxhe (Belgium) J. W. Pearman (Australia) Department of Clinical
Microbiology,
H. Humphreys (Dublin) A. Holmes (London) G. Reybrouck (Belgium) University Hospital Birmingham
T. Inkster (Glasgow) P. P. Mortimer (London) H.-G. Sonntag (Germany) NHS Foundation Trust,
Queen Elizabeth Medical
K. Jacobson (Bristol) R. N. Olmsted (USA) A. Voss (Netherlands) Centre,
P. Jumaa (Birmingham) J. Philpott-Howard (London) Edgbaston,
Birmingham B15 2TJ
K. G. Kerr (Harrowgate) G. L. Ridgway (London)
G. McDonnell (Basingstoke) M. Rotter (Austria) Secretary
Ms C. Fry
A. J. Mifsud (London) E. T. M. Smyth (Belfast) Department of Health,
S. W. B. Newsom (Cambridge) Wellington House,
133–155 Waterloo Road,
B. Oppenheim (Birmingham) London SE1 8UG
S. Schelenz (Norwich)
Scientific Secretary
P. Shears (Wirral) Dr T. C. Boswell
R. C. Spencer (Bristol) Department of Microbiology,
Queens Campus,
J. Stephenson (Surrey) Nottingham University Hospitals
J. Stockley (Worcester) NHS Trust, Derby Road,
Nottingham NG7 2UH
E. L. Teare (Chelmsford)
A. M. Walker (Pentraeth) Treasurer
Dr W. A. Telfer Brunton
A. P. R. Wilson (London) Department of Clinical
Microbiology,
Royal Cornwall Hospitals NHS
Reviews Trust,
I. M. Gould (Aberdeen) Penventinnie Lane, Treliske,
Truro TR1 3LQ

Correspondence
G. D. Corcoran (Cork)
VOLUME 73 ISSUE 4 DECEMBER 2009

Contents
Introduction
Healthcare-associated infection: moving behind headlines to clinical
solutions
D. Pittet 293
The Hospital Infection Society

Reviews
Historical and changing epidemiology of healthcare-associated infections
A. Pearson 296
The authors, editors, owners and
publishers do not accept any
Role of hand hygiene in healthcare-associated infection prevention
responsibility for any loss or damage
arising from actions or decisions B. Allegranzi and D. Pittet 305
arising from information contained
in this publication; ultimate respon- Preventing surgical site infection. Where now?
sibility for the treatment of patients
H. Humphreys 316
lies with the medical practitioner.
The opinions expressed are those of
the authors and the inclusion in this Intravascular catheter infections
publication of material relating to J. Edgeworth 323
a particular product, method or
technique does not amount to
an endorsement of its value or Noroviruses in healthcare settings: a challenging problem
quality, or of the claims made by its M. Koopmans 331
manufacturers.

Abstracted/Indexed by: Pseudomonas aeruginosa: a formidable and ever-present adversary


Current Contents, ASCA, Science K.G. Kerr and A.M. Snelling 338
Citation Index, Infomed, Index
Medicus, Medline, EMBASE/Excerpta
Extended-spectrum ␤-lactamase-producing organisms
Medica, Abstracts of Hygiene,
Communicable Disease and Tropical M.E. Falagas and D.E. Karageorgopoulos 345
Diseases Bulletin and Cumulative
Index to Nursing and Allied Health Acinetobacter: an old friend, but a new enemy
Literature.
K.J. Towner 355

Community-associated meticillin-resistant Staphylococcus aureus as a


cause of hospital-acquired infections
R.L. Skov and K.S. Jensen 364

Screening and isolation for infection control


E. Tacconelli 371

The role of environmental cleaning in the control of hospital-acquired


infection
S.J. Dancer 378

Controversies in infection: infection control or antibiotic stewardship to


control healthcare-acquired infection?
I.M. Gould 386

Where does infection control fit into a hospital management structure?


E.T. Brannigan, E. Murray and A. Holmes 392
Hand hygiene and infection in hospitals: what do the public know; what
should the public know?
M. Fletcher 397

What are the drivers of the UK media coverage of meticillin-resistant


Staphylococcus aureus, the inter-relationships and relative influences?
T. Boyce, E. Murray and A. Holmes 400

Are national targets the right way to improve infection control practice?
M. Millar 408

Responsibility for managing healthcare-associated infections: where does


the buck stop?
B.I. Duerden 414

Letter to the Editor


Review of Lancet conference on healthcare-associated infections
M. Meda 418
Journal of Hospital Infection (2009) 73, 293e295
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

INTRODUCTION

Healthcare-associated infection: moving behind


headlines to clinical solutions
D. Pittet*

Infection Control Programme, University of Geneva Hospitals and Faculty of Medicine, 4 Rue Gabrielle-
Perret Gentil, 1211 Geneva 14, Switzerland

Available online 12 October 2009

Introduction pressure on the MRSA reservoir deserves con-


trolled interventions. Importantly, MRSA control
Healthcare-associated infection (HCAI) is universal is feasible and many examples are available at
and complicates patient care both in developed hospital, regional, or even national levels.
and developing countries. Worldwide, it is esti- VRE spread continues to represent a paradox
mated that as many as 1.4 million patients acquire on both sides of the Atlantic Ocean. VRE is
infections each day in hospitals alone.1 Recognis- a coloniser of the gut like Gram-negatives, of
ing that HCAI prevention is of paramount impor- the skin like MRSA, and of the environment like
tance on the current patient safety agenda, The C. difficile. Thus, VRE control mandates a com-
Lancet organised an international conference on plete, multimodal infection prevention strategy,
HCAI in London in December 2008. but is clearly feasible. P. aeruginosa remains
a key pathogen in hospitals and causes multiple
What did we learn? infections. However, the jury is still out on the
role of tap-to-patient and patient-to-tap cross-
Our old friendsdmeticillin-resistant Staphylococ- transmission as the reservoir/source. Should
cus aureus (MRSA), vancomycin-resistant P. aeruginosa be considered now as an occupa-
enterococci (VRE), Pseudomonas aeruginosa, tional hazard? Acinetobacter spp. are acquiring
Acinetobacter spp., and Clostridium difficiled pan-resistance to antimicrobials and causing
continue to challenge healthcare settings world- outbreaks, particularly among specific patient
wide. An MRSA pandemic is ongoing and strains populations. In particular, their capacity for
are capable of acquiring new virulence factors long-term survival in the environment compli-
in addition to multiple resistance. Rapid diagnosis cates control. Should we propose a zero toler-
seems useful, but only in conjunction with ance for P. aeruginosa and Acinetobacter spp.?
preventive actions. The screening of target popu- Extended-spectrum b-lactamase-producing en-
lations deserves further investigation, whereas terobacteriaceae (ESBLs) are now becoming
universal screening appears to be neither effec- universal with multiple, complex resistance
tive nor cost-effective. The role of antibiotic mechanisms that differ worldwide, and studies
are needed to better delineate their epidemiol-
ogy and control. The emergence of a new C. dif-
* Tel.: þ41 22 372 9828; fax: þ41 22 372 3987. ficile strain over the past years has highlighted
E-mail address: didier.pittet@hcuge.ch major failures in patient safety. Large
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.08.004
294 D. Pittet

differences in the disease epidemiology and con- emphasis on hand-touch sites, it is not the sole
trol are observed among hospitals and countries, responsibility of the infection control team.
but control is feasibledeven at country level. Cleanliness is everyone’s business, from
Our old friends are going public: community housekeeping to executive staff and board members.
pathogens are now entering the hospital. Examples There is a clear link between antimicrobial use
include ESBLs and community-acquired MRSA. and resistance at all levels; ward, hospital, re-
Norovirus outbreaks originating in the community gional, country, and even continental. As shown
are affecting institutionalised patient populations, recently through successful nationwide campaigns,
sometimes with devastating consequences. And the impact of antibiotic control is easier to
finally, C. difficile is now responsible for severe demonstrate in the community. Infection control
community infections among young adults and teams in hospitals have traditionally worked as
non-immunocompromised populations in contrast fire-fighters and now need to move on to more
to its more traditional role previously restricted prevention-oriented stewardship. When well-
to healthcare settings. organised and effective, antimicrobial stewardship
is largely cost-effective.
Are there clinical solutions? Infection control must be integrated into the
hospital management structure at all levels, in-
Targeted HCAI prevention is clearly effective. cluding the decision-making process. It should use
Prevent intravascular line infection? e ‘Yes, we a system-based approach to ensure sustainability,
can!’ How? By using multimodal intervention strat- be viewed as a corporate priority, and be based on
egies, sometimes also called ‘bundles’. Prevent corporate accountability and institutional leader-
surgical site infection? e ‘Yes, we possibly can.’ ship. Performance management tools related to
Again, by decreasing several risk factors, in infection control metrics are available and HCAI
particular, by the optimal and timely use of rates can be used as a proxy indicator for differ-
perioperative antibiotic prophylaxis. Preventing ences in system management. The best healthcare
ventilator-associated pneumonia remains difficult model needs to deliver effective, reliable and
and complex, but is certainly possible using mul- resilient infection prevention.
timodal interventions whose components still need
to be defined. In particular, there is a pressing Should we go public?
need to revisit the content of the currently
proposed bundle strategies. HCAI is a topic of high public and media interest.
The critical role of setting targets for practice However, this increased openness and transpar-
improvement needs to be reviewed, especially at ency could also result in a rising concern about the
national level. First, healthcare systems should infection risk. As a consequence of the ‘public
decide what and how to measure. Other ques- right to know’, public reporting and performance
tions that follow are: what are the best in- benchmarking is increasingly well-established in
terventions to implement? What are the targets several countries. But does it make a difference to
for improvement? Targets should be set locally the HCAI risk in hospitals and other healthcare
considering feasibility and cost-effectiveness. settings?
Risk taking is an integral part of healthcare, Martin Fletcher reviews the evidence on
similar to leadership. In the UK, mandatory publication reporting and proposes actions for
surveillance, public reporting, and the setting of boards and senior leaders to improve the
performance targets have clearly provided effec- institutional safety culture.3 He suggests hand
tive leverage. hygiene promotion as an exemplar model for in-
creasing patient safety at institutional, regional,
Towards integrated solutions or even national level, and the necessity to
allow patients to participate in this continuous
Hand hygiene remains the key measure for HCAI and complex challenge. It follows, therefore,
prevention. As illustrated by the successful imple- that communication at all levels must be opti-
mentation of the ‘‘Clean Care is Safer Care’’ multi- mised in the field of infection prevention; it
modal strategy proposed by the World Health must consider the public right to know,
Organization First Global Patient Safety Challenge allow patients and families to participate,
and launched in 2005, promotion is feasible and work in synergy with the media in multidis-
and effective worldwide (http://www.who.int/ ciplinary forums that include patients, health-
gpsc/en).2 Although environmental control must care workers, scientists, journalists and
improve in most healthcare settings with special politicians.
Healthcare-associated infection 295

Infection control is moving behind the headlines References


to guarantee safer healthcare by applying effec-
tive clinical solutions and by conducting research 1. Lynch P, Pittet D, Borg MA, Mehtar S. Infection control in
where needed. Infection control is no longer countries with limited resources. J Hosp Infect 2007;
optional. For the sake of every patient in our 65(Suppl. 2):148e150.
2. Pittet D, Donaldson L. Clean Care is Safer Care: a worldwide
care, the healthcare community across the globe priority. Lancet 2006;366:1246e1247.
must make every effort to ensure that it is fully 3. Fletcher M. Hand hygiene and infection in hospitals: what do
integrated into each institution’s management the public know; what should the public know? J Hosp Infect
structure. 2009;73: 397e399.
Journal of Hospital Infection (2009) 73, 296e304
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Historical and changing epidemiology of


healthcare-associated infections
A. Pearson a,b,*
a
Department of HCAI & AMR, Centre for Infections, London, UK
b
Department of Advanced Computational Biology, University of Maryland, College Park, Maryland, USA

Available online 3 November 2009

KEYWORDS Summary This review compares the historical perspectives on health-


Clostridium difficile; care-associated infections (HCAIs) with the current changing epidemio-
Healthcare-associated logical picture as it relates to these infections. Evidence in support of
infection;
these changes is given using trends in mortality for Clostridium difficile
Medical history;
MRSA;
and meticillin-resistant Staphylococcus aureus bacteraemia in England as
Surveillance examples. The impact of current intervention programmes which target
these, and other HCAIs, is also considered and knowledge gaps and options
for changes in public health strategy required to achieve further reductions
in HCAIs in the National Health Service in England are identified.
ª 2009 Published by Elsevier Ltd on behalf of The Hospital Infection Society.

Historical perspectives strictly attributable to him. Subsequently the pro-


tocol has been adopted universally by medical
In the fourth century BC, Hippocrates, the father of schools and accreditation bodies including the
western medicine, is generally attributed as General Medical Council in the UK.1
having introduced the need to ‘use treatments By the twenty-first century AD the Institute for
for the benefit of the ill in accordance with my Health Improvement in its ‘5 Million Lives’ cam-
ability and my judgment, but from what is to their paign had enhanced the definition of medical
harm and injustice I will keep them.’ This is one of harm as:
a series of statements that were subsequently Unintended physical injury resulting from or contrib-
referred to as the Hippocratic Oath. The oath uted to by medical care, including the absence of
was an ancient protocol in medicine at the time indicated medical treatment, that requires additional
of Hippocrates although it may not have been monitoring, treatment or hospitalisation, or that
results in death. Such injury is considered harm
whether or not it is considered preventable, whether
* Corresponding address: Department of HCAI & AMR, Centre
for Infections, 61 Colindale Avenue, London NW9 5EQ, UK. or not it resulted from a medical error or it occurred
E-mail address: dandrewpearson@googlemail.com within a hospital.2

0195-6701/$ - see front matter ª 2009 Published by Elsevier Ltd on behalf of The Hospital Infection Society.
doi:10.1016/j.jhin.2009.08.016
Historical and changing epidemiology of healthcare-associated infections 297

Through the following centuries the Egyptians aureus (MRSA). Figures 1 and 2 respectively show
and Europeans clearly enunciated their interest in the UK trends for increasing numbers of death
medical oversight and culpable negligence. In certificates reporting as a cause of death or men-
Egypt, Moses ben Maimon practised and taught tioning C. difficile (1993e2007) and MRSA (1999e
medicine insisting on cleanliness as the physician’s 2007). These trends are subject to ascertainment
best friend: ‘Never forget to wash your hands after bias as clinicians may change their threshold for
having touched a sick person’ and ‘I dismount from notifying deaths due to HCAI.
my animal, wash my hands, go forth to my
patients.’
In Europe by the nineteenth century several
physicians developed further the realisation of the National surveillance and evidence for
importance of hand washing. Dr Ignaz Semmelweis, the changing epidemiology of C. difficile
the Hungarian-born physician, generally regarded infection
as the father of infection control practice, was
responsible in 1847 for the maternity service of the Case surveillance based on laboratory reporting is
Allgemeine Krankenhaus teaching hospital in accepted as the most reliable routinely available
Vienna. There he observed that women delivered information by which to measure changes in
by physicians and medical students had a much incidence. In England there are two surveillance
higher rate of post-delivery mortality (13e18%) than systems for measuring S. aureus (including MRSA)
women delivered by midwives or trainee midwives and C. difficile infection: voluntary laboratory
(2%). Earlier, less well-evidenced, statements are reporting (LabBase electronic reporting system)
attributed to Oliver Wendell Holmes: and the Department of Health-initiated mandatory
MRSA bacteraemia and C. difficile surveillance
Let the men who mould opinions here look to it; if
programmes. The increase in laboratory-confirmed
there is any interested oversight, any culpable neg-
cases of C. difficile infection (CDI) reported to the
ligence and the facts shall reach the public ear, the
voluntary national laboratory reporting system
pestilence-carrier of the lying-in chamber must look
between 1990 and 2007 is shown in Figure 3. These
to God for pardon, for man will never forgive him.
English National Health Service (NHS)-based lab-
The work of Florence Nightingale in October oratories receive specimens from both hospital
1854 and April 1855 made a meticulous analysis of and community patients across the whole NHS.
the mortality data and identified poor sanitation as Ascertainment during this time varied from around
the underlying cause as well as creating new 45% in 1990 to 70% in 2007. Notwithstanding this
statistical diagrams that persuaded government increased reporting there was a steady increase
to reform health. This development of a modern- from 1990 to 2001, after which there was an accel-
day analytical approach to patient outcome was erating rate of increase recognised in retrospect as
the evidence base for her Notes on hospitals. This related to the emergence of newly recognised hy-
had a profound impact on the design and manage- per-toxin-producing strains of C. difficile. Figure 3
ment of hospitals as it tackled problems of over- also indicates the timelines for the introduction of
crowding in civil hospitals, poor ventilation and mandatory surveillance of laboratory-confirmed
lack of cleanliness and reported deaths from pre- cases of CDI, mandatory strain surveillance and
ventable diseases, deaths from wounds and deaths mandatory enhanced surveillance to monitor a per-
from all other causes. formance target to reduce CDI. In addition the UK
Treasury’s Public Service Delivery Agreement, pub-
lished in 2007, introduced a target for a 30% reduc-
HCAI in the twenty-first century tion in the number of cases of CDI reported by
English NHS hospitals in 2010e11 compared with
The contribution of deaths from HCAI to the total an agreed baseline in 2007e8.
lives lost per year has been estimated by Amalberti The impact of this mandatory surveillance pro-
who compared healthcare with ultrasafe activities gramme and the setting of a 30% reduction target
such as flying with scheduled airlines, the nuclear is seen in Figure 4. Data from the voluntary surveil-
power industry, road traffic deaths, and with lance shows ascertainment for patients 65 years
dangerous activities such as bungee jumping and as 77e85% of the mandatory scheme for each quar-
mountain climbing.2 ter between April 2007 and March 2008. So manda-
The risk from healthcare is further exemplified tory surveillance, public reporting and the setting
by reference to the published mortality reports for of a performance target was highly effective at in-
C. difficile and meticillin-resistant Staphylococcus creasing ascertainment. This intervention might
298 A. Pearson

9000
Underlying cause Mentions
8000

7000

6000
No. of deaths

5000

4000

3000

2000

1000

0
1999 2000 2001 2002 2003 2004 2005 2006 2007
Year

Figure 1 Number of deaths reported with C. difficile as underlying cause or mentioned 1993e2007. Data from the
UK Office of National Statistics.

itself be expected to influence physicians’ behav- guide physicians and clinical teams in the preven-
iour. During 2006 and 2007 the Department of tion of CDI.3 The overall impact of introducing
Health (DoH) launched the Saving Lives campaign mandatory surveillance, the Saving Lives campaign
with a High Impact Intervention care bundle to and the follow-up of NHS trusts against the

1800

Underlying cause Mentions


1600

1400

1200
No. of deaths

1000

800

600

400

200

0
1993 1995 1997 1999 2001 2003 2005 2007
Year

Figure 2 Number of deaths reported with MRSA as underlying cause or mentioned 1999e2007. Data from the UK
Office of National Statistics.
Historical and changing epidemiology of healthcare-associated infections 299

45 000

40 000

35 000

30 000
C. difficile counts

25 000

20 000

15 000

10 000

5000

0
90

91

92

93

94

95

96

97

98

99

00

01

02

03

04

05

06

07

08
19

19

19

19

19

19

19

19

19

19

20

20

20

20

20

20

20

20

20
Year

Figure 3 Voluntary NHS laboratory C. difficile surveillance. Timeline for introduction of mandatory surveillance:
2004, mandatory surveillance introduced; 2005, mandatory strain sampling introduced; 2007, mandatory enhanced
surveillance introduced. Source: Health Protection Agency LabBase Voluntary Reports; Department of Health Manda-
tory Surveillance Reports.

performance target by a DoH-based improvement National surveillance and evidence for


team has had a remarkable effect within the first the reduction in MRSA bacteraemia
full year of the programme. Figure 5 compares
the pattern of changes in rates of CDI per 1000 The laboratory-based voluntary system showed
bed-days. Marked variation across the English a steep rise in the reported number of MRSA bac-
NHS underlines both the variability and opportu- teraemia cases between 1990 and 2001 (Figure 6).
nity for further health gain that exists across the In 2001, the Chief Medical Officer noted that ‘the
NHS. prevalence of methicillin-resistant S. aureus has
The enhanced surveillance programme has also increased markedly in the last decade, primarily
begun to provide an insight as to the incidence of associated with hospital acquired infection.’4 Man-
CDI in patients aged <65 years as well as in datory reporting of MRSA and meticillin-suscep-
the general population diagnosed with CDI in tible S. aureus (MSSA) bacteraemia was
non-acute care settings. Measurement and the introduced for all NHS acute trusts in England in
interpretation of enhanced surveillance data April 2001. Enhanced mandatory surveillance was
from the mandatory scheme is complex but initial initiated in 2005 which allows cases to be entered
analysis indicates that an average of 29% of CDI in real time and collects more detailed information
cases would fit a definition of community-acquired regarding the incidence and risks of infection.
infection based on three categories of specimen: The voluntary reporting system continues to
on-presentation cases, cases developing less than operate alongside the mandatory surveillance
2 days after admission, and non-acute specimens. scheme. Voluntary surveillance will underestimate
Aggregate analysis may hide important local vari- the true number of MRSA bacteraemia cases (data
ation. This is the case with CDI in the community. not shown). Ascertainment is affected by the
The proportion of community CDI cases in local number of NHS trusts contributing voluntary data
NHS trusts varies markedly from 0 to >55% in and the completeness of these data, both of which
a small number of localities. These findings are are likely to have varied over time. In total, 30 946
newly recognised and under further investigation reports of MRSA bacteraemia were made via
by both epidemiologists and local microbiologists. LabBase between 2002 and 2008. The introduction
Early indications are that, as in the past, a pro- of mandatory reporting to the web increased this
portion of CDI cases do not have either exposure to number to 44 344. Mandatory reporting had the
antibiotics or healthcare as risk factors for their greatest impact in the first year when reporting
infection. increased by 49% from 4879 to 7274 cases.
300

18 000

16 000

14 000

12 000

10 000

8000

C. difficile counts
6000

4000

2000

Jul-Sep
Jul-Sep
Jul-Sep
Jul-Sep
Jul-Sep
Jul-Sep
Jul-Sep
Jul-Sep

Apr-Jun
Apr-Jun
Apr-Jun
Apr-Jun
Apr-Jun
Apr-Jun
Apr-Jun
Apr-Jun
Apr-Jun

Jan-Mar
Jan-Mar
Jan-Mar
Jan-Mar
Jan-Mar
Jan-Mar
Jan-Mar
Jan-Mar
Jan-Mar

Oct-Dec
Oct-Dec
Oct-Dec
Oct-Dec
Oct-Dec
Oct-Dec
Oct-Dec
Oct-Dec

2000 2001 2002 2003 2004 2005 2006 2007 2008


Quarter

Figure 4 Comparison of Mandatory and Voluntary NHS laboratory C. difficile surveillance. Solid black line: mandatory 65 years; solid grey line: voluntary 65
years; dashed line: mandatory 2e64 years; dotted line: voluntary 2e64 years. Source: Health Protection Agency LabBase Voluntary Surveillance Reports.
A. Pearson
3.50

3.00

2.50

2.00

1.50

C. difficile rate per 1000 bed-days


1.00

0.50
Historical and changing epidemiology of healthcare-associated infections

0
2004 - 2007 2004 - 2007 2004 - 2007 2004 - 2007 2004 - 2007 2004 - 2007 2004 - 2007 2004 - 2007 2004 - 2007
and nds n ast est ast st s ber
ndo hE hE We nd
E ngl i dla Lo r t r t hW u t u t h i dla Hum
f st M No No So So st M &
st o Ea We re
Ea k shi
r
Yo
HPA Region
301

Figure 5 Regional C. difficile rate per 1000 bed-days. HPA, Health Protection Agency. Source: Department of Health Mandatory MRSA bacteraemia surveillance.
302 A. Pearson

16 000

14 000

12 000
MRSA
S. aureus bacteraemia counts

10 000 MSSA

No susceptibility
8000

6000

4000

2000

0
1990 1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006
Year

Figure 6 Staphylococcus aureus bacteraemia voluntary laboratory reporting 1990e2006: meticillin sensitivity
reported by English NHS acute trusts.

Comparison of the reported numbers to 2008 and the Health Protection Agency has provided the
showed that mandatory reporting has increased NHS with high quality information for the public,
ascertainment by between approximately 900 and patients and clinical teams so that the risks
2300 cases per year to 39%. associated with the performance of certain pro-
The impact of introducing mandatory surveil- cedures are transparent.
lance, the Saving Lives campaign and the use of Worldwide data on measurement of HCAI is
improvement teams by the DoH along with a na- remarkable in its inadequacy. An editorial from
tional ‘cleanyourhands’ campaign has effected the leading US senators compared measurement in
required national reduction of 50% by June 2008 healthcare and baseball and cited that:
(Figure 7). Again the introduction of mandatory
Remarkably, a doctor today can get more data on the
enhanced surveillance of MRSA bacteraemia cases
starting third baseman on his baseball team than on the
gave a new insight to the epidemiology as it dem-
effectiveness of life-and-death medical procedures.
onstrated that only 65% of cases were acquired
Studies have shown that most health care is not based
during the current hospital admission whereas
on clinical studies of what works best. Instead, most
34% were reported from patients who were recent
care is based on informed opinion, personal observa-
admissions from the community or non-acute care
tion or tradition .
facilities. Voluntary submitted risk factor data on
3495/12 483 (28%) cases reported to the mandatory
To deliver better health care, we should learn from the
surveillance system between May 2006 and Decem-
successful teams that have adopted baseball’s new
ber 2008 indicated key information on the sources
evidence-based methods. The best way to start im-
of MRSA bacteraemia and highlighted the impor-
proving quality and lowering costs is to study the stats .
tance of vascular lines, skin and soft tissue infec-
tions as well as other localised infections and In the past decade, baseball has experienced a data-
urinary tract infections as potentially preventable driven information revolution. Numbers-crunchers now
sources of infection. This is invaluable information routinely use statistics to put better teams on the field
on which to base local strategies to further reduce for less money. Our overpriced, underperforming
MRSA bacteraemia. health care system needs a similar revolution .
So, with the introduction of mandatory surveil-
lance, HCAI has been made a visible and unambig- To deliver better health care, we should learn from
uous indicator of quality and safety of patient care the successful teams that have adopted baseball’s
Historical and changing epidemiology of healthcare-associated infections 303

2000

1500
MRSA bacteraemias reported

1000

500

0
Apr-Jun
Jul-Sep
Oct-Dec

Apr-Jun
Jul-Sep
Oct-Dec

Apr-Jun
Jul-Sep
Oct-Dec

Apr-Jun
Jul-Sep
Oct-Dec

Apr-Jun
Jul-Sep
Oct-Dec

Apr-Jun
Jul-Sep
Oct-Dec

Apr-Jun
Jul-Sep
Oct-Dec

Apr-Jun
Jan-Mar

Jan-Mar

Jan-Mar

Jan-Mar

Jan-Mar

Jan-Mar

Jan-Mar
2001 2002 2003 2004 2005 2006 2007 2008
Quarter

Figure 7 Reduction of mandatory MRSA bacteraemia surveillance 2005e2008. Sources: NHS Mandatory and Volun-
tary MRSA bacteraemia surveillance.

new evidence-based methods. The best way to start tracking will assist infection control teams in pre-
improving quality and lowering costs is to study the venting as well as controlling clusters of infection
stats.5 in hospitals at the local level but rapid strain char-
acterisation will be invaluable in recognising
The introduction into the English NHS of a web-
changes not only in local but also in regional and
enabled reporting and user access networked
national epidemiology. Similarly, real-time elec-
mandatory surveillance system has addressed this
tronic prescribing systems will allow enhanced an-
deficiency in measurement for a limited number
tibiotic audit and better analysis of antibiotic mis-
of infections currently targeted by mandatory
prescribing which, in turn, will help in efforts to
surveillance and performance management.
control CDI and antimicrobial resistance.
Selective use of improved evidence-based
Knowledge gaps performance management interventions such as
reduction targets, ‘never events’ and ‘zero toler-
Encouraging though these trends are, current gaps ance’ will also be beneficial. Finally development
in our knowledge relating to healthcare-associated and refinement of tools which permit assessment
infection need to be addressed if further reduc- of new interventions to identify how they can be
tions in these infections are to be achieved. These deployed in a cost-effective manner remains a key
include introduction of robust surveillance systems priority.
to monitor infections other than those related to
MRSA bacteraemia and C. difficile. This should
include systematic population-based epidemiolog-
ical surveillance. Technologies which permit real- Conflict of interest statement
time strain characterisation and real-time patient None declared.
304 A. Pearson

Funding sources 2. Amalberti R. The paradoxes of almost totally safe transpor-


None. tation systems. Saf Sci 2001;37:109e126.
3. Department of Health. Saving Lives: a delivery programme
to reduce healthcare associated infection including MRSA.
London: DoH; 2006.
References 4. Department of Health. CMO’s update 30: surveillance of
healthcare associated infections. London: DoH; 2001.
1. General Medical Council. Good medical practice: the duties 5. Beane W, Gingrich N, Kerry J. How to take American health
of a doctor registered with the General Medical Council. care from worst to first. New York Times Online, 24 October
London: GMC; 2006. 2008.
Journal of Hospital Infection (2009) 73, 305e315
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Role of hand hygiene in healthcare-associated


infection prevention
B. Allegranzi a,*, D. Pittet a,b
a
World Alliance for Patient Safety, World Health Organization, Geneva, Switzerland
b
Infection Control Programme, University of Geneva Hospitals and Faculty of Medicine,
Geneva, Switzerland

Available online 31 August 2009

KEYWORDS Summary Healthcare workers’ hands are the most common vehicle for
Alcohol-based hand the transmission of healthcare-associated pathogens from patient to
rub; Hand hygiene; patient and within the healthcare environment. Hand hygiene is the leading
Healthcare-associated
measure for preventing the spread of antimicrobial resistance
infection;
Intervention; Patient
and reducing healthcare-associated infections (HCAIs), but healthcare
safety; Promotion; worker compliance with optimal practices remains low in most settings.
World Health This paper reviews factors influencing hand hygiene compliance, the
Organization impact of hand hygiene promotion on healthcare-associated pathogen
cross-transmission and infection rates, and challenging issues related to
the universal adoption of alcohol-based hand rub as a critical system
change for successful promotion. Available evidence highlights the fact
that multimodal intervention strategies lead to improved hand hygiene
and a reduction in HCAI. However, further research is needed to evaluate
the relative efficacy of each strategy component and to identify the most
successful interventions, particularly in settings with limited resources.
The main objective of the First Global Patient Safety Challenge, launched
by the World Health Organization (WHO), is to achieve an improvement in
hand hygiene practices worldwide with the ultimate goal of promoting
a strong patient safety culture. We also report considerations and
solutions resulting from the implementation of the multimodal strategy
proposed in the WHO Guidelines on Hand Hygiene in Health Care.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

* Corresponding author. Address: First Global Patient Safety Challenge, World Alliance for Patient Safety, IER/PSP, Room L319,
L Building, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland. Tel.: þ41 22 791 2689; fax: þ41 22 791 1388.
E-mail address: allegranzib@who.int

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.04.019
306 B. Allegranzi, D. Pittet

Introduction Factors influencing hand hygiene


compliance
Numerous studies document the pivotal role of
healthcare workers’ (HCWs) hands in the propaga- It has been known for many years that HCWs
tion of micro-organisms within the healthcare encounter difficulties in complying with hand
environment and ultimately to patients.1 As hygiene indications at different levels.4 Insuffi-
recently described, patient-to-patient transmis- cient or very low compliance rates have been
sion of pathogens via HCWs’ hands involves five reported from both developed and developing
sequential steps.2 Patients’ skin can be colonised countries.1,4 Reasons which explain suboptimal
by transient pathogens that are subsequently practices are multiple and may vary according to
shed onto surfaces in the immediate patient the setting and the resources available. For exam-
surroundings, thus leading to environmental ple, the lack of appropriate infrastructure and
contamination.2 As a consequence, HCWs contam- equipment to enable hand hygiene performance,
inate their hands by touching the environment or the cultural background, and even religious beliefs
patients’ skin during routine care activities, can play an important role in hindering good prac-
sometimes even despite glove use.2 It has been tices.13e15 The most frequently observed factors
shown that organisms are capable of surviving determining poor hand hygiene compliance are:
on HCWs’ hands for at least several minutes fol- (i) belonging to a certain professional category
lowing contamination.2 Thus, if hand hygiene (i.e. doctor, nursing assistant, physiotherapist,
practices are suboptimal, microbial colonisation technician); (ii) working in specific care areas
is more easily established and/or direct transmis- (i.e. intensive care, surgery, anaesthesiology,
sion to patients or a fomite in direct contact with emergency medicine); (iii) understaffing and over-
the patient may occur.2 crowding; and (iv) wearing gowns and/or gloves.1
Based on this evidence and the demonstration Unfortunately, hand hygiene indications at higher
of its effectiveness, optimal hand hygiene behaviour risk of being neglected are the ones that prevent
is considered the cornerstone of healthcare- pathogen transmission to the patient (i.e. before
associated infection (HCAI) prevention.2e4 Fur- patient contact and clean/aseptic procedures).1
thermore, not only is it a key element of standard This is also in concordance with the fact that
and isolation precautions, but its importance is care activities with a higher risk of cross-transmis-
emphasised also in the most modern ‘bundle’ sion lead to a higher risk of poor compliance.1
approaches for the prevention of specific site in- Individual factors such as social cognitive de-
fections such as catheter-related bloodstream terminants may provide additional insight into
infection (CRBSI), catheter-related urinary tract hand hygiene behaviour.3,16e18 Many factors play
infection (CRUTI), surgical site infection (SSI), a role in eventually determining either a hand
and ventilator-associated pneumonia (VAP).5e9 hygiene action or lack of compliance: perception
Together with other specific prevention and knowledge of the transmission risk and of the im-
measures, environmental cleaning is another pact of HCAI; social pressure; HCWs’ conviction of
essential measure to prevent the spread of their self-efficacy; the evaluation of perceived ben-
some pathogens, particularly Clostridium diffi- efits against the existing barriers; the intention to
cile, vancomycin-resistant enterococci (VRE), perform the hand hygiene action. For instance, in-
norovirus, Acinetobacter spp. and meticillin- tention to wash hands did not predict observed hand-
resistant Staphylococcus aureus (MRSA), and washing behaviour in one study, whereas it did in
should not be neglected.10e12 another.19,20 Hence, hand hygiene behaviour ap-
Over the past few years, scientific evidence to pears not to be homogeneous and can be classified
support the role of hand hygiene in the improve- into at least two types of practice.21 Inherent hand
ment of patient safety has increased consider- hygiene practice, which drives most community
ably, but some key controversial issues still and HCW hand hygiene actions, occurs when hands
challenge care practitioners and researchers. are visibly soiled, sticky or gritty. On the other
This review summarises the key themes on hand, elective hand hygiene practice represents
the role of hand hygiene in preventing HCAI. those opportunities for hand cleansing not encom-
Interpretations and solutions based on the passed in the inherent category. Among HCWs, this
evidence and experience available through the component of hand hygiene behaviour is similar to
work of the First Global Patient Safety Challenge many common social interactions, such as shaking
of the WHO World Alliance for Patient Safety are hands. During healthcare, it would include touching
suggested. a patient (e.g. taking a pulse or blood pressure) or
Hand hygiene and HCAI prevention 307

having contact with an inanimate object in the pa- reliability cannot be assessed.44 In a prospective,
tient’s surroundings. As they recall a common social controlled, cross-over trial, Rupp and colleagues ob-
behaviour, these contacts do not necessarily trigger served no substantial change in device-associated
an intrinsic need to cleanse hands, although they infection rates and infections due to multidrug-
do involve the risk of cross-transmission. According resistant pathogens, despite a significant and sus-
to behavioural theories, this is the component of tained improvement in hand hygiene adherence.41
hand hygiene most likely to be omitted by busy Nevertheless, although the study was well designed,
HCWs and it has been repeatedly confirmed by field it was criticised for lack of screening for cross-trans-
observations. mission, lack of statistical power, and use of an
alcohol-based hand rub that failed to meet the EN
Impact of hand hygiene promotion on 1500 standards for antimicrobial efficacy.46e48
HCAI In many countries, the evidence from studies on
hand hygiene effectiveness has been convincing
Given the complexity of hand hygiene behaviour enough to motivate governments to invest re-
and the influence of numerous external factors, sources in hand hygiene national and subnational
promotion of good practices is complex and its campaigns.49 However, this evidence mainly
potential for success depends on the delicate reflects findings from interventions implemented
balance between evaluation of benefits and exis- in healthcare settings in developed countries.
tent barriers. Demonstration of the effectiveness of Further research is needed to evaluate the relative
recommendations and strategies to improve hand efficacy of each key element of multimodal strat-
hygiene on the ultimate outcome, i.e. the HCAI egies, to assess their implementation feasibility
rate, is crucial in both motivating HCWs’ behav- in settings with limited resources, and to gather
ioural change and securing an investment in this information on successful solutions allowing adap-
preventive measure by policy-makers and health- tation. Among its main objectives, the First Global
care managers. However, research in this field Patient Safety Challenge, launched by the WHO
represents a very challenging activity since meth- World Alliance for Patient Safety, intends to
odological and ethical concerns make it difficult to make available implementation tools for field use
conduct randomised controlled trials with appro- and to assess their validation and adoption in
priate sample sizes that could establish the relative countries at different income levels.49
importance of hand hygiene in the prevention of Another controversial issue is how significant
HCAI. In addition, HCAI surveillance is a very should be the hand hygiene compliance increase
resource- and time-consuming activity requiring following the intervention in order to be considered
rigorous and standardised methods, and therefore satisfactory. No data are available yet to answer
is seldom available on a regular and reliable basis. this question. Among all the above-mentioned
Nevertheless, there is convincing evidence that studies, increased compliance rates at follow-up
improved hand hygiene can reduce infection rates. did not exceed 81% (Table I). One study with a fol-
More than 20 hospital-based studies of the impact of low-up of eight years showed a sustained compli-
hand hygiene on the risk of HCAI have been pub- ance increase of up to a maximum of 66% and
lished between 1977 and 2008 (Table I).22e45 Of succeeded in parallel to maintain the achieved
these, some were conducted hospital-wide and reduction in HCAI rates of <10%.29,30 To achieve
report long-term follow-up to demonstrate sustain- 100% compliance is not strictly necessary to deter-
ability.29,30,38,42 Despite study limitations, almost mine improvement of patient safety at the bedside.
all reports showed a temporal association between On the other hand, the goal of sustained 100% com-
improved hand hygiene practices and reduced pliance appears unlikely to be achieved because of
infection and cross-transmission rates. Most investi- the complex range of factors influencing HCWs’ be-
gations were conducted in adult or neonatal inten- haviour related to hand hygiene performance.
sive care units (ICUs) and the large majority Thus, there is a need for careful consideration be-
introduced the use of alcohol-based hand rubs in fore setting a goal of zero tolerance to hand hygiene
association with other promotional components in non-compliance to avoid failure and frustration.
a multimodal implementation strategy (Table I).
Three studies failed to show HCAI reduction follow-
ing hand hygiene promotion.24,41,44 In one study, Challenging issues related to the
the intervention did not succeed in significantly in- adoption of alcohol-based hand rubs
creasing hand hygiene compliance.24 In another,
the methods and definitions used to detect HCAI The adoption of alcohol-based hand rubs is con-
were not described and therefore the data sidered the gold standard for hand hygiene in most
Table I Most relevant studies assessing the impact of hand hygiene promotion on HCAI (1977e2008)
308

Year Hospital Intervention Impact on hand Impact on HCAI Duration of Reference


setting hygiene compliance follow-up
1977 Adult ICU Promotion of hand washing NA Significant reduction (P < 0.001) in 2 years 22
with a chlorhexidine hand the percentage of patients
cleanser colonised/infected by Klebsiella
spp.
1989 Adult ICU Education on hand Compliance increase from 14% to Significant reduction (P ¼ 0.02) in 6 years 23
washing, hand hygiene 73% (before patient contact) and HCAI rates (from 33% to 12% and
observation, performance from 28% to 81% (after patient from 33% to 10%, respectively,
feedback contact) after two intervention periods 4
years apart)
1990 Adult ICU Hand-washing promotion Compliance increase from 22% to No impact on HCAI rates 11 months 24
29.9%
1992 Adult ICUs Prospective multiple cross- NA Significant reduction (P < 0.02) in 8 months 25
over trial on hand hygiene HCAI rates using hand washing
with either chlorhexidine with chlorhexidine soap
soap or 60% isopropyl
alcohol with optional hand
washing with plain soap
1994 NICU Introduction of hand NA Elimination of MRSA, when 9 months 26
washing with triclosan 1% combined with multiple other
(w/v) infection control measures.
Significant reduction (P < 0.02) in
nosocomial bacteraemia (from
2.6% to 1.1%) using triclosan
compared with chlorhexidine for
hand washing
1995 Newborn Introduction of HCWs’ NA Control of MRSA outbreak 3.5 years 27
nursery hand washing and
neonates’ bathing with
triclosan 0.3% (w/v)
2000 MICU/NICU Organisational climate NA Significant (85%) relative 8 months 28
intervention reduction (P ¼ 0.02) in VRE rate in
the intervention hospital;
statistically not significant (44%)
relative reduction in control
hospital; no significant change in
MRSA
B. Allegranzi, D. Pittet
2000 Hospital-wide Alcohol-based hand rub Significant increase in Significant reduction (P ¼ 0.04 and 8 years 29,30
introduction, hand compliance from 48% to 66% P < 0.001) in the annual overall
hygiene observation, HCAI prevalence (42%) and MRSA
training, performance cross-transmission rates (87%).
feedback, posters Active surveillance cultures and
contact precautions implemented
during same period. A follow-up
study showed continuous
increase in hand rub use,
stable HCAI rates and cost
savings.
2003 Orthopaedic Alcohol-based hand rub NA 36% decrease (P value, NA) in 10 months 31
surgical unit introduction, posters, HCAI (mainly urinary tract
Hand hygiene and HCAI prevention

feedback on HCAI rates, infection and SSI) rates (from 8.2%


patient education and to 5.3%)
involvement
2004 Hospital-wide Alcohol-based hand rub No significant increase in Significant reduction (P ¼ 0.03) in 1 year 32
introduction, hand compliance before and after hospital-acquired MRSA cases
hygiene observation, patient contact (from 1.9% to 0.9%)
posters, performance
feedback, informal
discussions
2004 Adult Hand hygiene electronic Compliance increase from 19.1% Reduction in HCAI rates (not 2.5 months 33
intermediate monitoring at exit from to 27.3% by electronic statistically significant, P value,
care unit patient rooms, direct monitoring NA)
observation and voice
prompts
2004 NICU Alcohol-based hand rub Compliance increase from 40% to Reduction (P ¼ 0.14) in HCAI rates 6 months 34
introduction, hand 53% (before patient contact) and (from 11.3 to 6.2 per 1000
hygiene observation, from 39% to 59% (after patient patient-days)
training, hand-hygiene contact)
protocols, posters
2004 NICU Education, written Compliance increase from 43% to Significant reduction (P ¼ 0.003) 2 years 35
instructions, hand hygiene 80% in HCAI rates (from 15.1 to 10.7
observation, posters, per 1000 patient-days), in
performance feedback, particular for respiratory
financial incentives infections
(continued on next page)
309
Table I (continued)
310

Year Hospital Intervention Impact on hand Impact on HCAI Duration of Reference


setting hygiene compliance follow-up
2005 Hospital-wide Alcohol-based hand rub Compliance increase from Significant reduction (P ¼ 0.01) in 4 years 36
introduction, hand 62% to 81% hospital-associated rotavirus
hygiene observation, infections
training, posters
2005 Adult ICUs Hand-washing Compliance increase from Significant reduction (P < 0.001) 21 months 37
observation, training, 23.1% to 64.5% in HCAI rates (from 47.5 to 27.9
guideline dissemination, per 1000 patient-days)
posters, performance
feedback
2005 Hospital-wide Alcohol-based hand rub Compliance increase from Significant reduction (57%, 36 months 38
introduction, hand 21% to 42% P ¼ 0.01) in MRSA bacteraemia
hygiene observation,
training, posters,
promotional gadgets
2007 Neurosurgery Alcohol-based hand rub NA Reduction (54%, P ¼ 0.09) in 2 years 39
introduction, training, overall incidence of SSI.
posters Significant reduction (100%,
P ¼ 0.007) in superficial SSI rates
2007 Neonatal unit Posters, focus groups, Compliance increase from Reduction (P value, NA) in overall 27 months 40
hand hygiene observation, 42% to 55% HCAI rates (from 11 to 8.2
HCWs’ perception infections per 1000 patient-days)
assessment, feedback on and 60% decrease (P value NA) in
performance, perception risk of HCAI in very low birth
and HCAI rates weight neonates (from 15.5 to 8.8
episodes per 1000 patient-days)
2008 ICU Prospective, controlled, Compliance increase from No impact on device-associated 2 years 41
cross-over trial in two 38e37% to 68e69% infection and infections
units with education, due to multidrug-resistant
posters and alcohol- pathogens
based hand rub
introduction
B. Allegranzi, D. Pittet
2008 (1) Six pilot Alcohol-based hand rub (1) Compliance increase from (1) Significant reduction (1) 2 years 42
hospitals introduction, hand 21% to 48% (P ¼ 0.035) in MRSA bacteraemia
hygiene observation, (from 0.05 to 0.02 per 100 patient
training, posters, discharges per month) and of
promotional gadgets clinical MRSA isolates (P ¼ 0.003)
(2) All public (2) Compliance increase from (2) Reduction in MRSA (2) 1 year
hospitals in 20% to 53% bacteraemia (from 0.03 to 0.01
Victoria per 100 patient discharges per
(Australia) month, P ¼ 0.09) and of clinical
MRSA isolates (P ¼ 0.043)
2008 Urology Unit Alcohol-based hand rub Compliance increase from 0% Significant reduction (P < 0.001) 6 months 43
introduction, hand (estimation) to 28.2% in HCAI rates from 13.1% to 2.1%
Hand hygiene and HCAI prevention

hygiene observation,
training, posters, patient
education
2008 NICU Alcohol-based hand rub NA Significant reduction (P ¼ 0.009) 18 months 44
introduction, training, in HCAI incidence (4.1 vs 1.2 per
posters 1000 patient-days)
2008 NICU Alcohol-based hand rub Compliance increase from 6.3% No impact on HCAI rates (9.7 vs 7 months 45
introduction, hand to 81.2% 13.5 per 1000 patient-days) (P-
hygiene observation, value NA)
training, posters,
performance feedback,
focus groups
HCAI, healthcare-associated infection; ICU, intensive care unit; NICU, neonatal intensive care unit; MICU, medical intensive care unit; VRE, vancomycin-resistant enterococcus; MRSA,
meticillin-resistant Staphylococcus aureus; SSI, surgical site infection; NA, not available.
311
312 B. Allegranzi, D. Pittet

clinical situations. This recommendation, pro- Controversial issues related to the


moted by the CDC and WHO and embraced by use of alcohol-based hand rubs and
many national hand hygiene guidelines, is based on
Clostridium difficile spread
the evidence of better microbiological efficacy,
less time required to achieve the desired effect,
Following the widespread use of alcohol-based
point of patient care accessibility and a better skin
hand rubs as the gold standard for hand hygiene in
tolerance profile.1,29,50e56
healthcare, concern has been raised about their
The WHO Guidelines on Hand Hygiene in Health
lack of efficacy against spore-forming pathogens.
Care have been conceived to catalyse hand
Indeed, apart from iodophors, albeit at a concen-
hygiene improvement in any setting regardless
tration remarkably higher than the one used in
of the resources available and the cultural back-
antiseptics, no hand hygiene agent (including
ground.1,49,57 Since there is a strong emphasis in
alcohols, chlorhexidine, hexachlorophene,
the Guidelines and in their implementation tools
chloroxylenol, and triclosan) is reliably sporicidal
on the availability of alcohol-based hand rubs as
against Clostridium or Bacillus spp.1,58 Mechani-
a key factor for hand hygiene improvement, the
cal friction while washing hands with soap and
issue of the procurement and cost of these
water may help physically remove spores from
products, especially in developing countries, chal-
the surface of contaminated hands.59e61 As a con-
lenges the recommendation feasibility. Indeed,
sequence, contact precautions are highly recom-
global sales of commercially produced, alcohol-
mended during C. difficile-associated outbreaks,
based hand rubs in 2007 were as high as US $3
in particular, glove use and hand washing with
billion, corresponding to 295 million L in volume,
a non-antimicrobial or antimicrobial soap and
with an overall 16.3% increase compared with
water following glove removal after caring for
2003 (WHO, unpublished data), mostly observed
patients with diarrhoea.5
in Europe and North America (27% and 23% in-
The widespread use of alcohol-based hand rubs
crease, respectively). Looking at procurement
in healthcare settings has been blamed repeatedly
opportunities, these products are available only
for the increase in C. difficile-associated disease
in South Africa in the African continent and in
rates, although this has not been demonstrated
China, India, and Japan in the AsiaePacific region
by any study to date.62,63 On the contrary, the
(WHO unpublished data). The most important is-
observed increase in C. difficile-associated disease
sue curbing the purchasing power in these regions
began in the USA long before the wide use of alco-
is the high cost of these products. Market prices
hol-based hand rubs.64,65 Furthermore, one large
vary from US $2.50 to 8.40 per 100 mL dispenser
outbreak with the epidemic strain REA-group B1
and are clearly unaffordable for many developing
(equivalent to ribotype 027) was managed success-
countries. The WHO multimodal hand hygiene im-
fully by introducing alcohol-based hand rub for all
provement strategy offers a possible solution to
patients other than those with C. difficile-
this obstacle: the local production of either of
associated disease.66 In addition, several studies
two WHO-recommended hand rub formulations.1
recently demonstrated a lack of association be-
The implementation toolkit accompanying the
tween the consumption of alcohol-based hand
WHO Guidelines on Hand Hygiene in Health Care
rubs and the incidence of clinical isolates of
includes a Guide to Local Production to manufac-
C. difficile.67e69 In conclusion, discouraging the
ture alcohol-based hand rubs in hospital pharma-
widespread use of alcohol-based hand rubs for
cies or other facilities for local use.1 Two
the care of patients other than those with C. diffi-
formulations are proposed: one based on ethanol
cile-associated disease will only jeopardise overall
80% v/v, and one based on isopropyl alcohol 75%
patient safety in the long term.
v/v; both include hydrogen peroxide 0.125% v/v
and glycerol 1.45% v/v. Local production has
been carried out in many healthcare settings Discussion
worldwide and was carefully monitored and eval-
uated by WHO in several sites (WHO unpublished From the available evidence it appears that multi-
data). No major procurement, production, and modal interventions are the most suitable strategy
storage obstacles were encountered and long- to determine behavioural change leading to im-
term stability at tropical temperatures was shown proved hand hygiene compliance and reduction in
(up to 19 months). The final products complied HCAI rates. Introduction of alcohol-based hand rubs
with quality control standards and had good skin and continuous educational programmes are key
tolerability at very low cost (less than US $0.50 factors to overcome infrastructure barriers and to
per 100 mL). build solid knowledge improvement. Support by
Hand hygiene and HCAI prevention 313

healthcare administrators and commitment by Guideline for prevention of surgical site infection, 1999.
national and local governments are essential to Infect Control Hosp Epidemiol 1999;20:247e278.
10. Rampling A, Wiseman S, Davis L, et al. Evidence that hospi-
make hand hygiene an institutional and national tal hygiene is important in the control of methicillin-
priority for patient safety and to ensure long-term resistant Staphylococcus aureus. J Hosp Infect 2001;49:
sustainability of promotional programmes. Higher 109e116.
priority should also be given to hand hygiene as a 11. Dancer SJ. Importance of the environment in meticillin-
research topic, through good-quality, randomised, resistant Staphylococcus aureus acquisition: the case for
hospital cleaning. Lancet Infect Dis 2008;8:101e113.
controlled trials to determine definitively its impact 12. Goodman ER, Platt R, Bass R, Onderdonk AB, Yokoe DS,
on HCAI and the relative effectiveness of the Huang SS. Impact of an environmental cleaning intervention
different components of multimodal strategies. on the presence of methicillin-resistant Staphylococcus
aureus and vancomycin-resistant enterococci on surfaces
in intensive care unit rooms. Infect Control Hosp Epidemiol
2008;29:593e599.
Acknowledgements 13. Ahmed QA, Memish ZA, Allegranzi B, Pittet D. Muslim
health-care workers and alcohol-based handrubs. Lancet
We wish to thank all members of the Infection 2006;367:1025e1027.
Control Programme, University of Geneva Hospi- 14. Duerink DO, Farida H, Nagelkerke NJD, et al. Preventing
nosocomial infections: improving compliance with standard
tals and members of the WHO First Global Patient precautions in an Indonesian teaching hospital. J Hosp
Safety Challenge ‘Clean Care is Safer Care’ core Infect 2006;64:36e43.
group (lead, D. Pittet): J. Boyce, B. Cookson, N. 15. Allegranzi B, Memish ZA, Donaldson L, Pittet D. Religion and
Damani, D. Goldmann, L. Grayson, E. Larson, G. culture: potential undercurrents influencing hand hygiene
Mehta, Z. Memish, H. Richet, M. Rotter, S. Sattar, promotion in healthcare. Am J Infect Control 2009;37:28e34.
16. Whitby M, PessoaSilva CL, McLaws ML, et al. Behavioural
H. Sax, W.H. Seto, A. Voss, A. Widmer. considerations for hand hygiene practices: the basic build-
ing blocks. J Hosp Infect 2007;65:1e8.
Conflict of interest statement 17. Seto WH. Staff compliance with infection control practices:
WHO takes no responsibility for the information application of behavioural sciences. J Hosp Infect 1995;
provided or the views expressed in this paper. 30(Suppl.):107e115.
18. Pittet D. The Lowbury lecture: behaviour in infection
control. J Hosp Infect 2004;58:1e13.
Funding sources 19. O’Boyle CA, Henly SJ, Larson E. Understanding adherence
None. to hand hygiene recommendations: the theory of planned
behavior. Am J Infect Control 2001;29:352e360.
20. Jenner EA, Watson PWB, Miller L, Jones F, Scott GM. Ex-
plaining hand hygiene practice: an extended application
References of the theory of planned behaviour. Psychol Health Med
2002;7:311e326.
1. WHO guidelines for hand hygiene in health care (Advanced 21. Whitby M, McLaws M-L, Ross RW. Why healthcare workers
draft). Geneva: World Health Organization; 2006. don’t wash their hands: a behavioral explanation. Infect
2. Pittet D, Allegranzi B, Sax H, et al. Evidence-based model Control Hosp Epidemiol 2006;27:484e492.
for hand transmission during patient care and the role of 22. Casewell M, Phillips I. Hands as route of transmission for
improved practices. Lancet Infect Dis 2006;6:641e652. Klebsiella species. Br Med J 1977;2:1315e1317.
3. Kretzer EK, Larson EL. Behavioral interventions to improve 23. Conly JM, Hill S, Ross J, et al. Handwashing practices in an
infection control practices. Am J Infect Control 1998;26: intensive care unit: the effects of an educational program
245e253. and its relationship to infection rates. Am J Infect Control
4. Pittet D, Boyce J. Hand hygiene during patient care: pursuing 1989;17:330e339.
the Semmelweis legacy. Lancet Infect Dis 2001; April:9e20. 24. Simmons B, Bryand J, Neiman K, Spencer L, Arheart K. The
5. Siegel JD, Rhinehart E, Jackson M, Chiarello L. Guideline for role of handwashing in prevention of endemic intensive
isolation precautions: preventing transmission of infectious care unit infections. Infect Control Hosp Epidemiol 1990;
agents in health care settings. Am J Infect Control 2007; 11:589e594.
35(Suppl. 2):S65eS164. 25. Doebbeling BN, Stanley GL, Sheetz CT, et al. Comparative
6. Eggimann P, Harbarth S, Constantin MN, Touveneau S, efficacy of alternative hand-washing agents in reducing nos-
Chevrolet JC, Pittet D. Impact of a prevention strategy ocomial infections in intensive care units. N Engl J Med
targeted at vascular-access care on incidence of infections 1992;327:88e93.
acquired in intensive care. Lancet 2000;355:1864e1868. 26. Webster J, Faoagali JL, Cartwright D. Elimination of meth-
7. O’Grady NP, Alexander M, Dellinger EP, et al. Guidelines for icillin-resistant Staphylococcus aureus from a neonatal
the prevention of intravascular catheter-related infections. intensive care unit after hand washing with triclosan.
MMWR Recomm Rep 2002;51:1e29. J Paediatr Child Health 1994;30:59e64.
8. Pronovost P, Needham D, Berenholtz S, Sinopoli D, Chu H, 27. Zafar AB, Butler RC, Reese DJ, Gaydos LA, Mennonna PA.
Cosgrove S. An intervention to decrease catheter-related Use of 0.3% triclosan (Bacti-Stat) to eradicate an outbreak
bloodstream infections in the ICU. N Engl J Med 2006;335: of methicillin-resistant Staphylococcus aureus in a neonatal
2725e2732. nursery. Am J Infect Control 1995;23:200e208.
9. Mangram AL, Horan TC, Pearson ML, Silver LC, Jarvis WR, 28. Larson EL, Early E, Cloonan P, Sugrue S, Parides M. An orga-
Hospital Infection Control Practices Advisory Committee. nizational climate intervention associated with increased
314 B. Allegranzi, D. Pittet

handwashing and decreased nosocomial infections. Behav 46. Mermel LA, Boyce JM, Voss A, Allegranzi B, Pittet D. Trial of
Med 2000;26:14e22. alcohol-based hand gel in critical care units. Infect Control
29. Pittet D, Hugonnet S, Harbarth S, et al. Effectiveness of Hosp Epidemiol 2008;29:577e579; author reply 580e582.
a hospital-wide programme to improve compliance with 47. McGuckin M, Waterman R. ‘‘Cannot detect a change’’ is not
hand hygiene. Lancet 2000;356:1307e1312. the same as ‘‘there is not a change’’. Infect Control Hosp
30. Pittet D, Sax H, Hugonnet S, Harbarth S. Cost implications Epidemiol 2008;29:576e577; author reply 580e582.
of successful hand hygiene promotion. Infect Control Hosp 48. Widmer AF, Rotter M. Effectiveness of alcohol-based hand
Epidemiol 2004;25:264e266. hygiene gels in reducing nosocomial infection rates. Infect
31. Hilburn J, Hammond BS, Fendler EJ, Groziak PA. Use of Control Hosp Epidemiol 2008;29:576; author reply 580e582.
alcohol hand sanitizer as an infection control strategy in an 49. Allegranzi B, Pittet D. Healthcare-associated infection in
acute care facility. Am J Infect Control 2003;31:109e116. developing countries: simple solutions to meet complex
32. MacDonald A, Dinah F, MacKenzie D, Wilson A. Performance challenges. Infect Control Hosp Epidemiol 2007;28:
feedback of hand hygiene, using alcohol gel as the skin 1323e1327.
decontaminant, reduces the number of inpatients newly 50. Boyce JM, Pittet D. Guideline for hand hygiene in
affected by MRSA and antibiotic costs. J Hosp Infect 2004; health-care settings. Recommendations of the Healthcare
56:56e63. Infection Control Practices Advisory Committee and the
33. Swoboda SM, Earsing K, Strauss K, Lane S, Lipsett PA. Elec- HICPAC/SHEA/APIC/IDSA Hand Hygiene Task Force. MMWR
tronic monitoring and voice prompts improve hand hygiene Recomm Rep 2002;51:1e45.
and decrease nosocomial infections in an intermediate care 51. Larson EL, Eke PI, Laughon BE. Efficacy of alcohol-based
unit. Crit Care Med 2004;32:358e363. hand rinses under frequent-use conditions. Antimicrob
34. Lam BC, Lee J, Lau YL. Hand hygiene practices in a neonatal Agents Chemother 1986;30:542e544.
intensive care unit: a multimodal intervention and impact 52. Larson EL, Aiello AE, Bastyr J, et al. Assessment of two hand
on nosocomial infection. Pediatrics 2004;114:e565ee571. hygiene regimens for intensive care unit personnel. Crit
35. Won SP, Chou HC, Hsieh WS, et al. Handwashing program for Care Med 2001;29:944e951.
the prevention of nosocomial infections in a neonatal intensive 53. Picheansathian W. A systematic review on the effectiveness
care unit. Infect Control Hosp Epidemiol 2004;25:742e746. of alcohol-based solutions for hand hygiene. Int J Nurs Pract
36. Zerr DM, Allpress AL, Heath J, et al. Decreasing hospital- 2004;10:3e9.
associated rotavirus infection: a multidisciplinary hand 54. Widmer AF. Replace hand washing with use of a waterless
hygiene campaign in a children’s hospital. Pediatr Infect alcohol hand rub? Clin Infect Dis 2000;31:136e143.
Dis J 2005;24:397e403. 55. Boyce JM. Scientific basis for handwashing with alcohol and
37. Rosenthal VD, Guzman S, Safdar N. Reduction in nosocomial other waterless antiseptic agents. In: Rutala WA, editor.
infection with improved hand hygiene in intensive care Disinfection, sterilization and antisepsis: principles and
units of a tertiary care hospital in Argentina. Am J Infect practices in healthcare facilities. Washington, DC: Associa-
Control 2005;33:392e397. tion for Professionals in Infection Control and Epidemiology;
38. Johnson PD, Martin R, Burrell LJ, et al. Efficacy of an alco- 2001. p. 140e151.
hol/chlorhexidine hand hygiene program in a hospital with 56. Graham M. Frequency and duration of handwashing in an in-
high rates of nosocomial methicillin-resistant Staphylococ- tensive care unit. Am J Infect Control 1990;18:77e81.
cus aureus (MRSA) infection. Med J Aust 2005;183:509e514. 57. Pittet D, Allegranzi B, Storr J, et al. Infection control as
39. Thi Anh Thu L, Dibley MJ, Nho VV, Archibald L, Jarvis WR, a major WHO priority for developing countries. J Hosp
Sohn AH. Reduction in surgical site infections in neurosurgi- Infect 2008;68:285e292.
cal patients associated with a bedside hand hygiene pro- 58. Rotter ML. Hand washing and hand disinfection. In:
gram in Vietnam. Infect Control Hosp Epidemiol 2007;28: Mayhall G, editor. Hospital epidemiology and infection
583e588. control. Baltimore: Williams & Wilkins; 1996. p. 1052e1068.
40. Pessoa-Silva CL, Hugonnet S, Pfister R, et al. Reduction of 59. McFarland LV, Mulligan ME, Kwok RY, Stamm WE. Nosoco-
health care associated infection risk in neonates by success- mial acquisition of Clostridium difficile infection. N Engl J
ful hand hygiene promotion. Pediatrics 2007;120: Med 1989;320:204e210.
e382ee390. 60. Bettin K, Clabots C, Mathie P, Willard K, Gerding DN. Effec-
41. Rupp ME, Fitzgerald T, Puumala S, et al. Prospective, con- tiveness of liquid soap vs chlorhexidine gluconate for the re-
trolled, cross-over trial of alcohol-based hand gel in critical moval of Clostridium difficile from bare hands and gloved
care units. Infect Control Hosp Epidemiol 2008;29:8e15. hands. Infect Control Hosp Epidemiol 1994;15:697e702.
42. Grayson ML, Jarvie LJ, Martin R, et al. Significant reductions 61. Hubner NO, Kampf G, Löffler H, Kramer A. Effect of a 1 min
in methicillin-resistant Staphylococcus aureus bacteraemia hand wash on the bactericidal efficacy of consecutive surgi-
and clinical isolates associated with a multisite, hand hy- cal hand disinfection with standard alcohols and on skin
giene culture-change program and subsequent successful hydration. Int J Hyg Environ Health 2006;209:285e291.
statewide roll-out. Med J Aust 2008;188:633e640. 62. Clabots CR, Gerding SJ, Olson MM, Peterson LR, Gerding DN.
43. Nguyen KV, Nguyen PT, Jones SL. Effectiveness of an alco- Detection of asymptomatic Clostridium difficile carriage by
hol-based hand hygiene programme in reducing nosocomial an alcohol shock procedure. J Clin Microbiol 1989;27:
infections in the urology ward of Binh Dan Hospital, Viet- 2386e2387.
nam. Trop Med Int Health 2008;13:1297e1302. 63. Wullt M, Odenholt I, Walder M. Activity of three disinfec-
44. Capretti MG, Sandri F, Tridapalli E, Galletti S, Petracci E, tants and acidified nitrite against Clostridium difficile
Faldella G. Impact of a standardized hand hygiene program spores. Infect Control Hosp Epidemiol 2003;24:765e768.
on the incidence of nosocomial infection in very low birth 64. McDonald LC, Owings M, Jernigan DB. Clostridium difficile
weight infants. Am J Infect Control 2008;36:430e435. infection in patients discharged from US short-stay hospi-
45. Picheansathian W, Pearson A, Suchaxaya P. The effective- tals, 1996e2003. Emerg Infect Dis 2006;12:409e415.
ness of a promotion programme on hand hygiene compli- 65. Archibald LK, Banerjee SN, Jarvis WR. Secular trends in hos-
ance and nosocomial infections in a neonatal intensive pital-acquired Clostridium difficile disease in the United
care unit. Int J Nurs Pract 2008;14:315e321. States, 1987e2001. J Infect Dis 2004;189:1585e1589.
Hand hygiene and HCAI prevention 315

66. Muto CA, Pokrwka M, Shutt K, et al. A large outbreak of 68. Vernaz N, Sax H, Pittet D, Bonnabry P, Schrenzel J,
Clostridium difficile-associated disease with an unexpected Harbarth S. Temporal effects of antibiotic use and hand
proportion of deaths and colectomies at a teaching hospital rub consumption on the incidence of MRSA and Clostridium
following increased fluoroquinolone use. Infect Control difficile. J Antimicrob Chemother 2008;62:601e617.
Hosp Epidemiol 2005;26:273e280. 69. Kaier K, Hagist C, Frank U, Conrad A, Meyer E. Two time-
67. Boyce JM, Ligi C, Kohan C, Dumigan D, Havill NL. Lack of series analyses of the impact of antibiotic consumption
association between the increased incidence of Clostridium and alcohol-based hand disinfection on the incidences of
difficile-associated disease and the increasing use of alco- nosocomial methicillin-resistant Staphylococcus aureus in-
hol-based hand rubs. Infect Control Hosp Epidemiol 2006; fection and Clostridium difficile infection. Infect Control
27:479e483. Hosp Epidemiol 2008;29:593e599.
Journal of Hospital Infection (2009) 73, 316e322
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Preventing surgical site infection. Where now?


H. Humphreys*

Department of Clinical Microbiology, RCSI Education and Research Centre, Smurfit Building, Beaumont
Hospital, PO Box 9063, Dublin 9, Ireland

Available online 22 August 2009

KEYWORDS Summary Surgical site infection (SSI) is increasingly recognised as a mea-


Healthcare bundles; sure of the quality of patient care by surgeons, infection control practi-
Laparoscopic surgery; tioners, health planners and the public. There is increasing pressure to
Meticillin-resistant
compare SSI rates between surgeons, institutions and countries. For this
Staphylococcus
aureus;
to be meaningful, data must be standardised and must include post-
Performance discharge surveillance (PDS) as many superficial SSIs do not present to
indicators; the original institution. Further work is required to determine the best
Post-discharge method of conducting PDS. In 2008 two important documents on SSI were
surveillance; published from the Society for Healthcare Epidemiology of America/The
Surgical site infection Infectious Disease Society of America and the National Institute for Health
and Clincal Excellence, UK. Both emphasise key aspects during the preop-
erative, operative and postoperative phases of patient care. In addition to
effective interventions known to be important for some time, e.g. not
shaving the surgical site until the day of the procedure, there is increasing
emphasis on physiological parameters, e.g. blood glucose concentrations,
oxygen tensions and body temperature. Laparoscopic procedures are in-
creasingly associated with reduced SSI rates, and the screening and
decontamination of meticillin-resistant Staphylococcus aureus carriers is
effective for certain surgical procedures but has to be balanced by cost
and the risk of mupirocin resistance. Finally, there is a need to convert
theory into practice by the rigorous application of SSI healthcare bundles.
Recent studies suggest that, with a multidisciplinary approach, simple
measures can be effective in reducing SSI rates.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

Introduction

* Tel.: þ353 1 8093708/3710; fax: þ353 1 8093709. Recent years have seen an increasing emphasis
E-mail address: hhumphreys@rcsi.ie on the importance of surgical site infection (SSI)
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.03.028
Preventing surgical site infection 317

from the perspective of the surgeon, infection SSI rates are increasingly seen as an example of
prevention and control practitioners, health ad- a performance indicator of the quality of health-
ministrators, managers and patients. While some care. Changes are taking place in some countries
healthcare-associated infections (HCAIs) such as leading to the public reporting of HCAI, including
bloodstream infections are more likely to be SSI rates.7,8 Although there are some logistical
associated with life-threatening illness or even difficulties in ensuring that rates are truly
death, SSI remains among the commonest infec- comparable, it is likely that the future will see
tions occurring in acute hospitals. patients having access to the SSI rate of their lo-
In a survey of 75 694 patients in England, Wales, cal hospital before admission for elective surgery.
Northern Ireland and the Republic of Ireland, the In North America, this aspect of what could be
overall prevalence of HCAI was 7.59%, with SSI described as ‘healthcare consumerism’ has led
accounting for 14.5% of these infections.1,2 The to major initiatives involving healthcare profes-
surveillance of some or all SSIs is conducted in sionals, patient groups and others, to try to signif-
many hospitals as part of routine surveillance and icantly improve the quality of care by focusing on
as a measure of patient safety. For a period of those aspects that are amenable to prevention,
15 years from 1982 to 1997, 59 335 surgical sites thus aiming to reduce infection rates and produce
were surveyed in a 600-bed hospital with an over- better patient care.9 In this review, some aspects
all SSI rate of 4.5%, and for clean surgery 2.4%, of surveillance and recent issues in SSI prevention
with little change in rates during that period.3 are discussed.
The significance of SSI and its prevention to
surgeons carrying out procedures has increased as
they realise that this is a measure of their effec- Measuring the SSI rate
tiveness. Many surgeons rightly consider that the
most critical factor in the prevention of SSI, The details of techniques used for the surveillance
although difficult to scientifically quantify, is the of SSI are beyond the scope of this review,
sound judgement and proper technique of the nevertheless it is essential that standardised def-
surgeon and his/her team, as well as the general initions are used, e.g. classifying infections into
health of the patient.4 It is important to ensure that superficial, incisional, deep incisional and organ/
the actual surgical procedure is successful, e.g. space, and that allowance is made for risk factors,
complete removal of a tumour, and that preventible especially when infection rates are likely to be
adverse consequences arising from not adequately compared between one surgeon or one institution
addressing infection prevention are addressed. and another.10,11 This must be done accurately and
Quality iniatives taken by surgeons in collabora- allow for changes in healthcare such as those re-
tion with others are bearing fruit in highlighting sulting in shorter acute hospital stay, leading to
the issues and possible interventions. As part of many infections being diagnosed either in the out-
the American College of Surgeons Standard patients or in the community. This emphasises the
National Surgical Quality Improvement Programme importance of post-discharge surveillance (PDS).
(NSQIP), a survey of 117 private sector hosptials Although there are a number of options for PDS,
was undertaken to identify low and high outliers in existing research does not appear to have identi-
terms of SSI prevention.5 High outliers (increased fied a valid, reliable, practical and universally
infection rates) were assoicated with higher num- agreed method for PDS.12
bers of trainees, longer operations, and low hae- Whitby and colleagues in Queensland, Australia
moglobin levels in operated patients; low outliers attempted to improve the reliability of PDS
had a policy of minimising operating-room traffic.5 through patient self-determination and a pro-
Apart from the obvious morbidity and some- gramme of educating patients on SSI.13 The posi-
times mortality that follows SSI, it is increasingly tive predictive value for a correct diagnosis was
recognised that there is a significant cost to the 65.2% for the educated group compared with
healthcare system such as prolonged hospital stay 83.3% for the non-educated patient group, and
as well as the cost to society in disability payments the authors concluded that pre-discharge educa-
while out of work and loss of income tax revenue. tion resulted in patients overdiagnosing SSI.13
An example of the healthcare costs is described in PDS is particularly important for superficial
a caseecontrol study of SSI following coronary incisional SSI, which may not be serious enough
artery bypass surgery in Australia between 1996 to warrant re-admission to hospital. For example,
and 1998 in which 125 patients developed SSI; the in a survey in Finland of orthopaedic SSI, 86% of
mean excess healthcare cost attributable to SSI SSIs detected after hospital discharge were super-
was $AU12,419 per case.6 ficial.14 Data derived from 12 885 operations in
318 H. Humphreys

Scotland yielded a statistically higher SSI rate from both documents are outlined in Table I. Some
when PDS was performed (6.34% compared with are discussed below in terms of the evidence and
2.61%) and the authors concluded that a proced- what should be incorporated into current practice.
ure-specific approach was required with direct
observation of patients.15 In The Netherlands, a na- Hand hygiene and the surgical scrub
tional nosocomial surveillance network has been
collecting data on HCAI for some years. SSI rates Hand hygiene is regarded as one of the key
were determined according to different PDS components in any infection prevention strategy.
methods for surgical procedures performed in For many years, the traditional surgical scrub
62 hospitals.16 Recommended PDS methods, e.g. where the surgeon ensures that hands, nails and
direct observation by the surgeon, were compared parts of the forearm are lathered and scrubbed has
with passive surveillance, e.g. patients re-present- been standard practice. However, surgeons them-
ing to the hospital. Recommended compared with selves accept that their practice, both in the
passive surveillance methods detected a higher SSI operating theatre and outside, has often been
rate, 3.7% compared with 3.1%.16 suboptimal; 90% compliance is not sufficient.18 Re-
It is universally acknowledged that PDS is cent evidence suggests that the traditional ritual
essential in accurately determining the SSI rate, of scrubbing can be replaced by a less lengthy
but there is a need for consensus as to how best and less ritualistic procedure. In a study involving
this can be achieved while taking into account six surgical services and 4387 consecutive patients
limited resources. who underwent clean and clean-contaminated sur-
gery earlier this decade, there was no difference
Preventing SSI in the SSI rate, whether the surgeon used the
hand-rubbing protocol with 75% aqueous alcohol
Approaches to the prevention and control of SSI or a hand-scrubbing protocol with povidone-iodine
have evolved over many years and traditionally or chlorhexidine.19 Furthermore, the alcohol solu-
have been classified into those interventions tion was better tolerated and associated with
before surgery, during surgery and after surgery. improved compliance.
However, prevention must be underpinned by
a knowledge and understanding of the microbial Physiological parameters
pathogenesis, and the importance of surveillance.
Much of the advice from the Hospital Infection Tissue hypoxia leads to necrosis and is often
Control Practices Advisory Committee (HICPAC) followed by infection. High blood glucose levels,
from North America on SSI prevention is derived such as occur in patients with diabetes mellitus for
from practice and custom, rather than rigorous example, are also associated with increased risks
scientific studies such as randomised controlled of infection. It is logical to assume that normalising
trials, but practice, custom and even ritual, these will assist in the prevention of SSI, and
continue to influence approaches to prevention.17 recent studies have supported this. Gottrup has
In 2008 two important documents were pub- emphasised that the decisive period for the de-
lished that synthesised and presented clearly many velopment of SSI is during surgery and the first few
of the important issues pertaining to the pre- hours thereafter; consequently, the physiological
vention of SSI. The Society for Healthcare Epi- status of the patient at that time is crucial.20 A
demiology of America and the Infectious Diseases higher fraction of inspired oxygen (80% compared
Society of America produced recommendations on with 30%) was associated with a 39% lower risk
HCAI including SSI, addressing surveillance of SSI in a randomised controlled trial of 300
methods, interventions to actively prevent SSI patients.21 It is believed that the mechanism of
and approaches to monitoring the implementation action is by more effective neutrophil killing of
of those strategies.11 In the UK, the National Insti- potential pathogens.22
tute for Health and Clinical Excellence published In a case-controlled study of 260 patients un-
a document on the prevention and treatment of dergoing mastectomy, variables associated with
SSI which emerged after an extensive review of SSI included high blood glucose levels.23 Similarly,
the literature and a wide consultation exercise.10 in an assessment of risk factors for SSI after surgery
Both documents repesent an impressive body of for hepato-biliary pancreatic cancer, poor blood
literature on SSI but combine this with practical glucose levels were associated with an odds ratio
and feasible advice about prevention; both em- of 6.6 for the development of SSI.24 Physiological
phasise the importance of education and the iden- homeostasis is therefore important in maintaing
tification of risk factors for SSI. Some of the issues body defences, and inspired oxygen, controlled
Preventing surgical site infection 319

Table I Major headings and some factors in preventing surgical site infection from recent UK and North American
guidelines
NICEa SHEA/IDSAb
Preoperative phase Surgical Care Improvement Project
Patient showering and hair removal Proper hair removal
Patient and staff theatre wear Controlling blood glucose
Movement to and from theatre area Maintain normothermia
Nasal decontamination (do not use mupirocin routinely) Infrastructure
Mechanical bowel preparation (not routine) Trained personnel
Patient and staff jewellery Education
Antibiotic prophylaxis (which patients, Computer-assisted decision support and automated
when and number of doses) reminders
Intraoperative phase Antimicrobial prophylaxis
Hand decontamination Measure and provide feedback on process measures,
Incise drapes e.g. hair removal
Gowns and gloves Accountability
Antiseptic skin preparation and diathermy Chief executive responsible for support
Patient homeostasis (oxygenation, normothermia, etc.) Senior management ensures adequate
Wound irrigation and dressings personnel and perform job responsibilities
Antiseptics before closure Healthcare workers responsible for their practices
Postoperative phase Non-routine approaches
Dressings Vancomycin not routine for antimicrobial prophylaxis
Postoperative cleansing of surgical site Don’t delay surgery for parenteral nutrition
Topical agents (not indicated) Unresolved issues
Antibiotic treatment and debridement for SSI Preoperative bathing with chlorhexidine
Specialist wound care services Positive screening for, and decolonisation of, MRSA
Supplemental oxygenation for colorectal
procedures
Maintaining normothermia after colorectal surgery
MRSA, meticillin-resistant Staphylococcus aureus; SSI, surgical site infection.
a
Adapted from reference 10.
b
Society for Healthcare Epidemiology of America/Infectious Diseases Society of America (adapted from reference 11).

blood glucose concentrations and normothermia, procedures such as those involving the breast.
among other things, are important. However, prophylaxis has been recommended for
patients undergoing breast cancer surgery if they
Surgical practice have had previous chemotherapy, or if the surgery
involves reconstruction.26 Therefore surgeons
Appropriate use of antimicrobial prophylaxis, i.e. need to liaise with others, e.g. anaesthetists, mi-
during induction to ensure adequate tissue levels crobiologists, infectious disease physicians and
at the time of the first incision, is associated with others, to ensure that the correct agent is given
reduced SSI rates, particularly with surgical pro- at the right time and for the appropriate duration.
cedures at high risk of infection, such as those One of the major advances in surgical practice in
involving the gastrointestinal tract. However, it recent decades has been the development of
has taken some time before best practice has been laparoscopic or minimally invasive surgery. This
incorporated into routine protocols by surgeons offers the potential for reducing infection as the
and others. Recently, a statement for urological incision site is much smaller, but it is unclear
surgery incorporated important principles of ap- whether all other practices and the environment
propriate surgical prophylaxis and offered sensible setting associated with open surgery should be
options in terms of the choice of antibiotics for replicated. In a survey of 150 hospitals in the UK
a variety of urological procedures.25 Indications for and Ireland, 3.9e5.6% of minimally invasive surgical
prophylaxis are likely to increase with the com- procedures associated with urology, e.g. trans-
plexity of surgical procedures performed, and urethral prosthetic surgery, uretheric stent inser-
with the increasing longevity of the patient popu- tion, etc. took place in a non-ventilated room or in
lation. For example, antibiotic prophylaxis is not a treatment room with some form of ventilation,
usually recommended for elective clean surgical but outside a conventionally ventilated theatre.27
320 H. Humphreys

Meta-analysis of laparoscopic compared with Alternatively, there may have been a failure of
open repair of a perforated peptic ulcer suggests one intervention, i.e. rapid MRSA detection, to
that the risk of postoperative SSI is reduced when impact on what is a multifactorial process.
carried out laparoscopically, and similar findings
have been associated with colon surgery.28e30 Most Converting theory to practice
of these procedures currently take place in a full
operating theatre, but it may be that some proced- Despite the scientific literature, and the publi-
ures may not require the conventional ventilated cation of recommended best practice by many
theatre with appropriate air changes, air pressures professional bodies, compliance with preventive
and filters. Surgical factors, e.g. the necessity to measures is often suboptimal. A survey of almost
convert from a laparascopic to an open procedure 590 surgeons in Canada revealed that 63% were
if the procedure is complicated, may demand that not in compliance with recommended guidelines
all laparscopic procedures are carried out in oper- on preoperative bathing, hair removal, anti-
ating theatres, yet the benefits for patients from microbial prophylaxis or intraoperative skin
laparoscopic surgery e irrespective of where it preparation.35 The absence of full compliance
takes place e in terms of reduced morbidity are indicates that infection prevention and its impor-
increasingly clear. tance is not yet embedded in all routine surgical
practice. However, studies increasingly show the
Surgical site infection and Staphylococcus potential impact of collective efforts to minimise
aureus SSI.
Enhancing compliance with best practice re-
Staphylococcus aureus is the commonest cause of quires a multidisciplinary approach and ownership
SSI and increasingly meticillin-resistant S. aureus from all concerned. In a survey of antimicrobial
(MRSA) accounts for a greater proportion of infec- prophylaxis, each surgical service was internally
tions in many hospitals throughout the world. Rec- monitored for 30 consecutive surgical procedures
ommended measures to prevent MRSA will also with considerable educational and communication
assist in the reduction of SSI and these include efforts to improve practice.36 Compliance on en-
screening, decontamination and glycopeptide suring that prophylactic antibiotics were started
prophylaxis.31 within 60 min of surgical incision was increased
The role of pre-emptive mupirocin prophylaxis from 72.25% in the first six-month period to
to the anterior nares to reduce MRSA carriage and 83.78% during the conduct of the survey.36 None-
its impact on postoperative infection has been theless, the authors were disappointed that they
studied in detail in recent years. This has to be did not achieve their objective of 90% compliance.
balanced by the cost of mupirocin, the risk of Fifty-six hospitals participated in a project to
resistance e particularly with repeated courses e implement a quality improvement approach to
and the impact on postoperative SSI. However, two decrease SSI that involved appropriate prophy-
recent systematic reviews and meta-analyses lactic antibiotic practice, preventing hypergly-
strongly suggest that prophylactic intranasal mu- caemia, maintaining normothermia, optimising
pirocin significantly reduces the rate of postoper- oxygen tension and avoiding the shaving of the
ative infections including that caused by MRSA and surgical site.37 Improvements in process measure-
meticillin-susceptible S. aureus (MSSA).32,33 None- ments ranged from 3% to 27% and, during the first
theless, there is a reluctance to use mupirocin in three months of the programme, SSI rates fell from
all patients but to identify those patients most at 2.28% to 1.65%.37 Improving compliance with best
risk, i.e. those carrying S. aureus or those under- practice is also possible locally.38
going certain types of surgery such as orthopaedic The introduction of healthcare bundles, incor-
and cardiovascular surgery, where the con- porating validated methods and measures to pre-
sequences of postoperative infection are great. vent infection, which are rigorously policed and
The rapid detection of S. aureus might poten- audited, is the way forward to decreasing SSI and
tially assist in achieving this, as only patients other HCAIs. Such bundles can be combined with
carrying S. aureus would receive mupirocin pro- other measures to ensure the quality and safety of
phylaxis. However, a prospective interventional patient care. Eight hospitals in eight cities around
cohort study on twelve surgical ward patients using the world implemented a surgical safety list that
PCR to detect MRSA did not lead to a reduction reduced mortality and inpatient complications.39
overall in HCAI;34 the failure of this study to There is likely to be increasing pressure in the
demonstrate a benefit may be partly related to years ahead from patients/consumers and health-
the local setting, where MRSA was not endemic. care management/funding agencies to comply
Preventing surgical site infection 321

with best practice and to demonstrate improve- duration and blood transfusion e results of the first Ameri-
ments in outcomes. can College of SurgeonseNational Surgical Quality Improv-
ment Program Best Practices Initiative. J Am Coll Surg
2008;207:810e820.
6. Jenney AWJ, Harrington GA, Russo PL, Spelman DW. Cost
Discussion of surgical site infections following coronary artery bypass
surgery. Aust N Z J Surg 2001;71:662e664.
SSIs are one of the commonest HCAIs and are 7. Humphreys H, Cunney R. Performance indicators and the
increasingly recognised as a measure of the quality public reporting of healthcare-associated infection rates.
Clin Microbiol Infect 2008;14:892e894.
of healthcare. When benchmarking one surgeon 8. McKibben L, Horan T, Tokars JI, et al. Guidance on public
with another or between institutions, it is impor- reporting of healthcare-associated infections: recommen-
tant to ensure that data collected are accurate, dations of the Healthcare Infection Control Practices Advi-
adjusted for risk, and incorporate PDS. There is sory Committee. Am J Infect Control 2005;33:217e226.
now a wealth of scientific literature and consensus 9. Bratzler DW, Hunt DR. The surgical infection prevention and
surgical care improvement projects: national initiatives to
recommendations regarding those measures that improve outcomes for patients having surgery. Clin Infect
are important before, during and after surgery. Dis 2006;43:322e330.
When these are implemented in the form of 10. National Institute for Health and Clinical Excellence. Surgi-
healthcare bundles and involve multidisciplinary cal site infection, prevention and treatment of surgical site
input, significant improvements can be achieved. infection. National Collaborating Centre for Women’s and
Children’s Health; 2008.
Such improvements will be expected by patients 11. Anderson DJ, Kaye KS, Classen D, et al. Strategies to pre-
and members of the public as part of the in- vent surgical site infections in acute care hospitals. Infect
creasing consumerisation of healthcare. The years Control Hosp Epidemiol 2008;29(Suppl. 1):S51eS61.
ahead will see better surveillance of SSI, particu- 12. Petherick ES, Dalton JE, Moore PJ, Cullum N. Methods for
larly PDS. New products or devices used in patient identifying surgical wound infection after discharge from
hospital: a systematic review. BMC Infect Dis 2006;6:170.
care, e.g. application of shock wave therapy to the 13. Whitby M, McLaws M-L, Doidge S, Collopy B. Post-discharge
surgical site, are likely to be widely evaluated and surgical site surveillance: does patient education improve
some may be incorporated into routine practice.40 reliability of diagnosis? J Hosp Infect 2007;66:237e242.
Ultimately, while there has been some progress to 14. Huotari K, Lyytikäinen O. Impact of postdischarge surveil-
date, more can be done to prevent SSI by the sys- lance on the rate of surgical site infection after orthopaedic
surgery. Infect Control Hosp Epidemiol 2006;27:
tematic application of best practice before, during 1324e1329.
and after all surgical procedures. 15. Reilly J, Allardice G, Bruce J, Hill R, McCoubrey J. Proced-
ure-specific surgical site infection rates and postdischarge
surveillance in Scotland. Infect Control Hosp Epidemiol
2006;27:1318e1323.
16. Manniën J, Wille JC, Snoeren RLLM, van den Hof S. Impact
Conflict of interest statement/funding sources of postdischarge surveillance on surgical site infection rates
The author was not in receipt of financial for several surgical procedures: results from the nosocomial
support during the drafting of this manuscript. surveillance network in The Netherlands. Infect Control
However, he has received support in the last Hosp Epidemiol 2006;27:809e816.
17. Mangram AJ, Horan TC, Pearson ML, Silver LC, Jarvis WR.
two years for research from Steris Corporation,
Guideline for prevention of surgical site infection, 1999.
3M, Cepheid and Inova8 Science. He has also Infect Control Hosp Epidemiol 1999;20:247e280.
received fees for lectures or consultancy work 18. Gawande A. On washing hands. N Engl J Med 2004;350:
from 3M, Pfizer and Astellas. 1283e1286.
19. Parienti JJ, Thibon P, Heller R, et al. Hand-rubbing with
an aqueous alcoholic solution vs traditional surgical
hand-scrubbing and 30-day surgical site infection rates. A
References randomised equivalence study. J Am Med Assoc 2002;288:
722e727.
1. Smyth ETM, McIllvenny G, Enstone JE, et al. Four country 20. Gottrup F. Prevention of surgical-wound infections. N Engl J
healthcare associated infection prevalence survey 2006: Med 2000;342:202e204.
overview of the results. J Hosp Infect 2008;69:230e248. 21. Belda FJ, Aguilera L, Garcı́a de la Asunción J, et al. Supple-
2. Humphreys H, Newcombe RG, Enstone J, et al. Four country mental perioperative oxygen and the risk of surgical wound
healthcare associated infection prevalence survey 2006: infection. A randomised controlled trial. J Am Med Assoc
risk factor analysis. J Hosp Infect 2008;69:249e257. 2005;294:2035e2042.
3. Creamer E, Cunney RJ, Humpreys H, Smyth EG. Sixteen 22. Alan DB, Maguire JJ, Mahdavian M, et al. Wound hypoxia
year’s surveillance of surgical sites in an Irish acute-care and acidosis limit neutrophil bacteria killing mechanisms.
hospital. Infect Control Hosp Epidemiol 2002;23:36e40. Arch Surg 1997;132:991e996.
4. Nichols RL. Preventing surgical site infections: a surgeon’s 23. Vilar-Compte D, de Iturbe I, Martı́n-Onraet A, et al. Hyper-
perspective. Emerg Infect Dis 2001;7:220e224. glycemia as a risk factor for surgical site infections in pa-
5. Campbell DA, Henderson WG, Englesbe MJ, et al. Surgical tients undergoing mastectomy. Am J Infect Control 2008;
site infection prevention: the importance of operative 36:192e198.
322 H. Humphreys

24. Ambiru S, Kato A, Kimura F, et al. Poor postoperative blood systematic review of the literature and meta-analysis. In-
glucose control increases surgical site infections after sur- fect Control Hosp Epidemiol 2005;26:916e922.
gery for hepato-biliary-pancreatic cancer: a prospective 33. van Rijen M, Bonten M, Wenzel RP, Kluytmans JAJW. Intra-
study in a high volume institute in Japan. J Hosp Infect nasal mupirocin for reduction of Staphylococcus aureus
2008;68:230e233. infections in surgical patients with nasal carriage:
25. Wolf JS, Bennett CJ, Dmochowski R, Hollenbeck BK, a systematic review. J Antimicrob Chemother 2008;61:
Pearle MS, Schaeffer AJ. Best practice policy statement 254e261.
on urologic surgery antimicrobial prophylaxis. J Urol 2008; 34. Harbath S, Fankhauser C, Schrenzel J, et al. Universal
179:1379e1390. screening for methicllin-resistant Staphylococcal aureus
26. Nicolas P, Yazdanpanah Y, Marie-Pierre C, et al. Prevention at hospital admission and nosocomial infection in surgical
of surgical site infection after breast cancer surgery by tar- patients. J Am Med Assoc 2008;299:1149e1157.
geted prophylaxis antibiotic in patients at high risk of surgi- 35. Davis PJB, Spady D, deGara C, Forgie SED. Practices and at-
cal site infection. J Surg Oncol 2007;96:124e129. titudes of surgeons toward the prevention of surgical site
27. Smyth ETM, Humphreys H, Stacey A, Taylor EW, Hoffman P, infections: a provincial survey in Alberta, Canada. Infect
Bannister G. Survey of operating theatre ventilation facili- Control Hosp Epidemiol 2008;29:1164e1166.
ties for minimally invasive surgery in Great Britain and 36. McCahill LE, Ahern JW, Gruppi LA, et al. Enhancing compli-
Northern Ireland: current practice and considerations for ance with medicare guidelines for surgical infection preven-
the future. J Hosp Infect 2005;61:112e122. tion. Arch Surg 2007;142:355e361.
28. Lunevicius R, Morkevicius M. Systematic review comparing 37. Dellinger EP, Hausmann SM, Bratzler DW, et al. Hospitals
laparoscopic and open repair for perforated peptic ulcer. collaborate to decrease surgical site infections. Am J Surg
Br J Surg 2005;92:1195e1207. 2005;190:9e15.
29. Romy S, Eisenbring M-C, Bettschart V, Petignat C, 38. Trussell J, Gerkin R, Coates B, et al. Impact of a patient
Francioli P, Troillet N. Laparoscope use and surgical site in- care pathway protocol on surgical site infection rates in
fections in digestive surgery. Ann Surg 2008;247:627e632. cardiothoracic surgery patients. Am J Surg 2008;196:
30. Poon JT, Law W-L, Wong IW, et al. Impact of laparoscopic 883e889.
colorectal resection on surgical site infection. Ann Surg 39. Haynes AB, Weiser TG, Berry WR, et al. A surgical safety
2009;249:77e81. checklist to reduce morbidity and mortality in a global pop-
31. Rao GG, Osman M, Johnson L, Ramsey D, Jones S, Fidler H. ulation. N Engl J Med 2009;360:491e499.
Prevention of percutaneous endoscopic gastrostomy site 40. Dumfarth J, Zimpfer D, Vo }gele-Kadletz M, et al. Prophylac-
infections caused by methicillin-resistant Staphylococcus tic low-energy shock wave therapy improves wound healing
aureus. J Hosp Infect 2004;58:81e83. after vein harvesting for coronary artery bypass graft sur-
32. Kallen AJ, Wilson CT, Larson RJ. Perioperative intranasal gery: a prospective, randomised trial. Ann Thorac Surg
mupirocin for the prevention of surgical-site infections: 2008;86:1901e1913.
Journal of Hospital Infection (2009) 73, 323e330
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Intravascular catheter infections


J. Edgeworth*

Directorate of Infection, Guy’s and St Thomas’ Hospital, London SE1 7EH, UK

Available online 22 August 2009

KEYWORDS Summary Formerly an under-appreciated iatrogenic infection, catheter-


Catheter-related related bloodstream infections (CRBSIs) are now the focus of considerable
bloodstream infection; preventive strategies. Although robust clinical definitions remain elusive
Costebenefit analysis;
due to the difficulty in identifying the focus of infection in hospitalised
Infection control
patients, surveillance definitions are proving useful to monitor and com-
pare institutional rates of CRBSI and to target infection control resources.
New catheter-sparing diagnostic techniques have been developed, that are
probably most applicable to assessment of infection in stable ambulatory
patients with single long-term tunnelled catheters rather than acutely un-
well hospitalised patients. There is an impressive body of evidence that can
be used to support implementation, surveillance and audit of basic infec-
tion control practices that should help institutions reduce CRBSI rates. The
introduction of preventive antimicrobial strategies at the catheter site has
been recommended by international guidelines, yet there remains
justifiable concern about long-term selection of resistant organisms. This
has not been adequately addressed in current studies. Economic analyses
require data on the clinical effect of CRBSIs to adequately assess the
benefit; such data are scarce, owing to the difficulty in assessing the con-
tribution from comorbidities, with consequential conflicting results. Over-
all, institutions can justifiably first assess the benefit of a sustained
programme of re-enforcing basic infection control practice on CRBSI before
assessing whether the introduction of additional preventive antimicrobial
strategies are likely to have any benefit.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

Introduction

Intravascular catheters are indispensable for


modern healthcare.1 They include short-term pe-
* Tel.: þ44 207188 3107; fax: þ44 7261 9816. ripheral venous and arterial catheters, short-term
E-mail address: Jonathan.Edgeworth@gstt.nhs.uk central venous catheters (CVCs), long-term tunnelled
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.05.008
324 J. Edgeworth

and cuffed CVCs and long-term peripherally in- term catheters, and along the body and tip of
serted CVCs. Venous catheters are predominantly the catheter within the intravascular compart-
used for administration of fluids, drugs, total paren- ment.3,13 Significant exit-site and tunnel infections
teral nutrition and blood products and for renal re- are commonly due to pyogenic organisms, particu-
placement therapy. Arterial catheters are larly S. aureus, resulting in a combination of a hot,
predominantly used for haemodynamic monitoring red catheter exit site, cellulitis along the tract and
and collection of blood samples in the critical care pus formation. The visual signs usually prompt
setting. The main complication of catheter use is early removal of the catheter, although laboratory
the development of an infection which can be confirmation of infection is only made in a small
either localised, within the bloodstream or distal.2,3 proportion of cases, with mechanical and chemical
Such infections can have life-threatening conse- irritation being alternative causes of local inflam-
quences, particularly distal infections at sites such mation. Catheters lacking any overt signs of local
as heart valves and bones, which are most infection are also frequently removed from
frequently due to Staphylococcus aureus.4e6 There patients suspected of having a healthcare-associ-
are a number of new approaches to diagnosis and ated infection, and between 10% and 25% are
prevention that have been considered in recent found to grow significant numbers of organisms
comprehensive meta-analyses and reviews.1,7e10 without an attendant bloodstream infection.3,10
This article provides an overview of the potential This has been referred to as either catheter coloni-
benefit of these new strategies in the context of sation or infection. All organisms commonly
current infection prevention and control practice. cultured from hospitalised patients can also be
isolated from catheter tips including skin staphylo-
cocci, Candida spp., enterococci and Gram-negative
Epidemiology bacteria. The extent to which these organisms
cultured from catheter tips contribute to a sys-
It is estimated that about 250 000 CVCs are used in temic inflammatory response in the absence of
the UK each year, and in the USA nearly 300 million a documented CRBSI remains unclear. It is an
catheters are used yearly of which about 3 million area warranting further investigation.
are CVCs.7,8,11 Numbers of other catheters used, There are two main pathways leading to
particularly peripheral venous catheters on gen- CRBSI.3,14,15 The first is contact between skin sur-
eral wards, are not known. Most epidemiological face organisms either at the time of insertion or
studies on vascular catheters report on the use of thereafter, leading to migration of organisms
CVCs in intensive care units (ICUs), where there down, and colonisation of, the external catheter
are comparatively high rates of both catheter use surface. It is thought to be the dominant mech-
and infection within a limited, easily assessable, anism associated with short-term catheters.15
geographical setting. It is important to remember, The second involves transfer of organisms to the
however, that insertion of any vascular catheter catheter hubs from patient skin or healthcare
in any clinical setting can result in a catheter workers’ hands, usually leading to colonisation of
infection. Most studies and surveillance schemes the internal catheter surface, and is more common
focus on catheter-related bloodstream infections in long-term catheters.14,16 In both cases organisms
(CRBSIs) as a measure of catheter infection and should be present on the catheter tip and in the
use different denominators to compare rates blood, although for the hub infections organisms
including number of patients, patient days, cathet- may be confined to the internal catheter surface.
ers and catheter days. A recent systematic review Organisms colonise catheters through two main
of 200 prospective studies estimated that mechanisms: adherence to the catheter surface,
incidence rates, expressed as CRBSIs/1000 catheter- facilitated by host proteins such as fibronectin and
days, range from 0.5 for peripheral venous fibrinogen adsorbed on to the catheter surface,
catheters, to 1.6 for long-term CVCs, 1.7 for ar- and formation of biofilm on the catheter surface,
terial catheters, 2.1 for peripherally inserted which protects organisms in a vegetative state
CVCs, and 2.7 for short-term CVCs.12 where they resist both the innate immune system
and antibiotics.3,13,17,18 S. aureus and coagulase-
negative staphylococci (CoNS) both express
Clinical descriptions, microbiology and surface adhesins that permit binding to host extra-
pathogenesis cellular matrix proteins deposited on catheters
and CoNS, in particular, can produce factors
Local catheter infections occur at the exit site, involved in formation of a biofilm.19e22 However,
along the subcutaneous tract of tunnelled long- no single factor or group of related molecules has
Intravascular catheter infections 325

been shown to be essential for catheter colonisa- of risk, it requires the most resources, particularly
tion. Although staphylococci are the predominant if applied across a whole hospital.12
organisms isolated from catheters, other hospital
bacteria can be isolated from catheters suggesting
Improving diagnosis of CRBSI for clinical
other mechanisms of colonisation. Indeed, the and economic benefit
dominance of staphylococci may be at least in part
explained by the opportunity they have for contact A common approach to diagnosis of CRBSI in
with catheters due to their residence on skin. a patient with suspected healthcare-associated
infection with a single short-term peripheral cath-
eter or CVC is to take a blood culture and remove
Definitions of bloodstream infections the catheter for semiquantitative analysis.29 The
catheter tip is ‘rolled’ across an agar plate and
Robust definitions of bloodstream infections are CRBSI is diagnosed if the same organism is isolated
required for clinical diagnosis and management, from blood culture and the tip culture, and the
surveillance and investigational studies. Definitions quantity of organisms isolated from the tip is >15
used depend upon these varied indications and also colonies. This approach is simple and cheap, and
any mandated requirements and local resources. pooled data from 19 studies including short- and
Healthcare-associated BSI have been divided into long-term catheters demonstrate a sensitivity
primary (unknown), secondary to a specified focus and specificity of 85% and 82%, respectively.9
(such as skin and soft tissue, or the respiratory, Alternatively a fixed length of catheter can be
urinary and gastrointestinal tracts) and catheter- immersed in broth, sonicated and vortexed to
related events. Clinical definitions of CRBSI gener- release organisms into suspension that can then be
ally require culture of an organism from blood plated out and counted to give a quantitative mea-
supported by one of a number of diagnostic tests sure of catheter load. A cut-off of >103 organisms
(to be discussed later) implicating the catheter, and has been used. This technique has the advantage
no apparent alternative focus for the infection.23 of removing organisms from the internal catheter
The difficulty with such clinical definitions is the surface. Pooled data from 14 studies indicate a sensi-
extent to which alternative foci are sought and tivity and specificity of 83% and 87%, respectively.9
then deemed to be a ‘plausible’ alternative focus. Diagnoses requiring catheter removal are prob-
Identification of foci for infection causing sepsis in lematic in settings such as the ICU where patients
ICU is notoriously difficult, and even when strict cri- have multiple short-term central venous and ar-
teria are used the identification retains a degree of terial catheters, any or none of which may be the
subjectivity in patients who are acutely unwell for cause of an infection. Replacement of all cathet-
other reasons. A review of five representative ers is time-consuming, expensive and not without
large ICU clinical studies shows a wide variation in clinical risk including pneumothorax and haemor-
the proportion of hospital-acquired bloodstream rhage, particularly in patients with a bleeding
infection assigned to these three categories: tendency. Similarly, in ambulatory patients who
primary (22e61%), secondary (16e60%), CRBSI usually have a single long-term tunnelled catheter,
(19e62%).24e28 Although this may represent hetero- immediate catheter removal often necessitates
geneity in clinical practice and patient populations hospital admission with surgical and or radiological
it probably also reflects differences in local inter- intervention and unnecessary replacement may
pretation, which complicates communication of compromise precious vascular access. In both
studies in this field. Given the location of colonised settings, excluding a catheter as the infected
catheters in the bloodstream, available evidence source while still in situ can preserve the catheter
would equally support definitions where alternative life with potential clinical and economic benefit. A
foci for a bloodstream infection were considered number of catheter-sparing diagnostic approaches
only after catheters had been excluded. have been developed, of which simultaneous,
Surveillance definitions remove much of the quantitative culture and differential time to
subjectivity surrounding clinical definitions. The positivity have shown the most promise.
CDC definition of a catheter-associated infection is
a positive blood culture in a patient in whom Simultaneous quantitative blood cultures
a central line has been in use <48 h previously.23
Surveillance definitions are expressed as per 1000 Equal volumes of blood, usually 10 mL, are drawn
bed-days, per 1000 catheters or per 1000 cath- simultaneously through catheter(s) and a peripheral
eter-days. Although per 1000 catheter-days is vein. Blood is lysed and centrifuged and the blood is
recognised to be the most informative measure plated out on agar. Although studies have reported
326 J. Edgeworth

different cut-offs for a positive diagnosis, the Infec- on how to prevent CRBSI, supported by an impres-
tious Diseases Society of America has recommended sive body of evidence.23,30 Some guidelines
a five-fold greater colony count from a catheter consider all catheters whereas most focus on
culture to indicate it as the source of CRBSI. A long-term CVCs which are associated with the
meta-analysis of diagnostic techniques has shown greatest infection burden.12 The challenge for
this to be the most accurate test with a sensitivity healthcare institutions is to integrate these com-
and specificity for short-term catheters of 75% and prehensive guidelines into different clinical set-
97%, respectively, and for long-term catheters of tings within available resources, supported by
93% and 100%, respectively.9 This increases the a surveillance system to monitor adherence and
number of blood cultures processed by a laboratory, improvements.
however, and requires considerable additional work. Core components of a strategy for preventing
CRBSI include: hand hygiene; maximum sterile
Differential time to positivity barrier precautions for CVC insertions; 2% chlor-
hexidine for skin antisepsis; use of sterile semi-
Blood cultures are usually monitored using a real- permeable dressings to facilitate daily inspection;
time continuous detection system which can re- documentation of exit site appearance; avoidance
cord the time when organism growth is first of the femoral site for insertion for CVCs; removal
detected. Studies have shown that, when blood of the catheter as soon as it is no longer required;
cultures drawn through a catheter flag positive routine removal of peripheral venous catheters
more than 2 h before a peripheral blood culture, after 3 days; routine replacement of tubings and
the catheter can be implicated as the focus with administration sets; and use of a tunnelled dual
a sensitivity and specificity of 89% and 87%, respec- lumen catheter if placement is anticipated to be
tively, for short-term catheters and 90% and 72%, more than 21 days.23,30
respectively, for long-term catheters.9 There are There is a considerable body of data to support
concerns that this method particularly may be the majority of these interventions. One recent
compromised by antibiotics received through the comprehensive review stated that, ‘This is a well
catheter at or around the same time. researched area and few realistic research needs
Institutions considering catheter-sparing tech- were identified in developing these guidelines.’30
niques should assess the impact on the laboratory Nonetheless, some aspects of care still require fur-
before implementation. Where a single blood ther investigation. For example, it has not been
culture may have been taken from a patient with determined whether 2% chlorhexidine in 70% alco-
a suspected hospital-acquired infection, this may hol, the current preferred solution for skin anti-
increase 3e5-fold in patients that have many sepsis, is superior to 5% povidone iodine in 70%
catheters in situ such as those on ICU. Laboratory alcohol, which has itself been demonstrated as
blood culture machines have a limited capacity. superior to 10% povidone iodine solution. It is
Furthermore, the simultaneous collection of blood also not clear exactly how frequently dressings
through catheters can result in isolation of organ- and tubings should be changed.
isms such as meticillin-resistant S. aureus (MRSA), An important focus recently has been to in-
without confirmation of being a true CRBSI from tegrate these recommendations into a care pack-
a peripherally obtained culture; in the UK this is age that can be implemented across an institution.
nonetheless recorded as an institutional MRSA There have been a number of reports in the
bacteraemia for surveillance purposes and could literature, mostly in ICUs, documenting consider-
potentially divert infection control resources and able success using a combination of these inter-
lead to unnecessary clinical interventions. There- ventions.31,32 One notable study combined
fore, if catheter-sparing diagnostic techniques improved hand hygiene, full barrier precautions
are being considered it seems most appropriate for CVC insertion, chlorhexidine for skin antisepsis,
to introduce them in a setting such as renal dialysis avoidance of the femoral site and removal of un-
or oncology where single long-term catheters are necessary lines.32 It reported a reduction in CRBSIs
used and where catheter salvage probably has from 7.7 per 1000 CVC-days to 2.3 per 1000 CVC-
the greatest clinical benefit. days, which remained at low rates for 18 months
after the intervention period. Concerns have
Basic and enhanced strategies for been raised about the before-and-after study
preventing CRBSI design, compliance, potential for reversion to
mean, lack of data on relative importance of
Comprehensive national and international guide- individual components, definitions used and the
lines include a large number of recommendations Hawthorne effect of being involved in the study.
Intravascular catheter infections 327

Nonetheless, it is consistent with previous studies chloride, so as to prevent internal colonisation,


and has contributed to a body of data supporting thought to be a more common mechanism after
the UK national intervention programme, ‘Saving the first couple of weeks of insertion. Catheters
Lives’, which includes care bundles consisting of impregnated with either minocycline and rifampi-
the core components described above to target cin or miconazole and rifampicin have also been
peripheral catheters, long- and short-term CVCs.33 developed.
It is recognised that the success of individual There have been six systematic reviews of the
institutional programmes will be dependent upon clinical effectiveness of antimicrobial CVCs on re-
introducing a comprehensive education and ducing CRBSI, and a recent comprehensive Health
training programme supported by pragmatic easy- Technology Assessment, in which earlier reviews
to-introduce audit, surveillance and feedback, were assessed and compared.8 The odds ratios (ORs)
clear lines of accountability from ‘board to floor’ of reducing CRBSIs for first-generation, second-
and clinical engagement and leadership. generation and antibiotic-coated CVCs all indicated
an effect compared with standard catheters,
although for first-generation catheters there was
Antimicrobial strategies and a wider, non-significant confidence interval (CI)
antimicrobial lock (OR: 0.67, 95% CI: 0.43e1.06; 0.43, 0.26e0.70;
0.26, 0.15e0.46 respectively).8 It has been noted
A number of enhanced strategies have also been that when studies including first-generation cath-
assessed that have as a common mechanism the eters with short median insertion times (6 days;
provision of sustained antimicrobials or antiseptics range: 5.2e7.5) are assessed, a significant reduc-
at the catheter site.1,7,10 They include chlorhexi- tion in CRBSI is observed (0.48, 0.25e0.91).35
dine-impregnated sponges and dressings, cathe- Indeed, when all antimicrobial catheters are
ters impregnated with antimicrobial agents, and included, a significant reduction is only seen for
antimicrobial locks using antibiotics (including studies in which average catheter insertion was
vancomycin, gentamicin, cephalosporins, minocy- 5e12 days and not 13e20 days (0.4, 0.27e0.58 and
cline and ciprofloxacin) or taurolidine. 0.69, 0.42e1.14 respectively) although the
trend is in the same direction.8 Only one study
with a catheter insertion of >20 days has been
Chlorhexidine-impregnated sponges reported, demonstrating a significant benefit of
the antimicrobial catheter.36 Overall, the number
These are placed over the CVC insertion site and of antimicrobial catheters required to prevent one
covered with a transparent dressing. In theory they CRBSI ranges from 13 to 221 depending upon the
should prevent growth and migration of organisms rate in the control group.
along the external catheter surface but would
have no effect on hub colonisation and migration Antimicrobial locks
on the internal surface. A number of randomised
studies in adult and paediatric groups have shown This strategy involves infusion of 2e3 mL of an
variable reductions in CRBSI with use of these antibiotic-containing solution, usually with an anti-
sponges.1 A meta-analysis of five randomised coagulant into a CVC at the end of a period of use.
studies showed a significant reduction in catheter Since patients in critical care usually have
tip colonisation and a non-significant trend to- frequently accessed multiple short-term catheters,
wards a reduction in CRBSI.34 this strategy has predominantly been assessed with
long-term catheters in haemodialysis and oncology
Antimicrobial CVCs patients. A recent review identified three studies
assessing vancomycin lock with heparin compared
Impregnation of CVCs with antimicrobial agents is with heparin alone in a neonatal ICU, paediatric
potentially an effective way of ensuring an appro- and adult cancer patients.10 Although each setting
priate concentration of antimicrobial along the is quite different, all three studies demonstrated
whole length of the catheter to prevent colonisa- a significant reduction in CRBSI (0.37 vs 1.72 per
tion.7 First-generation antimicrobial catheters 1000 catheter-days; 2.3 vs 17.8 per 1000 catheter-
have either chlorhexidine and silver sulphadiazine days; 0 per 60 vs 4/57 CVCs; vancomycin vs controls
(CSS), or silver alone impregnated on the external in each case).37e39 A few studies in haemodialysis
catheter surface. Second-generation catheters are patients have assessed use of gentamicin and one
treated on both the external surface and internal study using either a cephalosporin or neomycin
lumen with either CSS, silver or benzalkonium have all shown a similar reduction.10 Three small
328 J. Edgeworth

studies have assessed the use of a 1.38% taurolidine antimicrobial catheters of £10 (range: £2.50e
plus 3.8% citrate lock compared with heparin in £25). All but one of the 81 potential scenarios
haemodialysis patients. Two reported a significant tested in their model using multivariate sensitivity
reduction in either CRBSI (90 day bacteraemia analysis demonstrated an economic benefit of an-
free survival rate 94 vs 47%; P ¼ 0.001) or an in- timicrobial catheters. Antimicrobial catheters
crease in sepsis free catheter survival (0 vs 4 epi- were shown not to be cost-effective only at the
sodes; P ¼ 0.047), but one larger study reported lowest rate of CRBSI in control population, lowest
no significant effect on CRBSI (0.85 vs 0.81 per relative risk reduction, highest cost different for
1000 catheter days).40e42 Further studies are the antimicrobial catheters, and lowest cost of
warranted. a CRBSI e a scenario the authors consider to be
very unlikely in the real world.
Many ICU studies have attempted to estimate
Clinical and economic evaluation of the the excess length of stay (LOS) due to CRBSI as
benefit of enhanced antimicrobial a proxy for additional costs; these have reported
strategies between 10 and 20 days.8 None has been per-
formed in the UK. Most of these studies have
The EPIC (Evidence-based Practice in Infection used a caseecontrol design which is fraught with
Control) guidelines have recommended antimicro- difficulties in both selecting controls and adjusting
bial CVC for adult patients at risk of CRBSI who for potential confounding variables. Traditional
require central venous access for 1e3 weeks.30 statistical approaches cannot overcome the prob-
The Healthcare Infection Control Practices lem that whereas a CRBSI can increase LOS, the
Advisory Committee (HICPAC) of the Centers for reverse may be equally true. Alternative statistical
Disease Control and Prevention has recommended approaches are being used that overcome these
the use of antimicrobial CVCs in adults whose cath- inherent confounders, such as longitudinal model-
eter is expected to remain in situ for >5 days and ling, where effects of variables on LOS can be
where the CRBSI rate is above the goal set by the assessed daily and so a temporal relationship
institution despite implementation of a compre- between healthcare-acquired infection and in-
hensive strategy to reduce rates.23 There are creased length of stay can be made. One study
frequent methodological concerns in reported using such an approach reported a non-significant
studies, including the diagnostic criteria used, increase in LOS for CRBSI of 2.6 days.46
inadequate blinding and in many cases poor ran- An important clinical consideration for use of
domisation, so it is understandable that different enhanced interventions to prevent CRBSI is the
views have emerged on implementation of these potential to save lives. The attributable mortality
recommendations.7,10 It is useful to review the of CRBSI in caseecontrol studies, mostly recruiting
clinical and economic evaluation of the benefit of patients from ICUs, has been reported to vary from
preventing CRBSIs that have underpinned these 0 to 35%.26,28,47e51 Studies have used a wide range
recommendations. of matching criteria to select controls based on
There have been three formal cost-effective- admission parameters [specialty, diagnosis, Acute
ness studies of antimicrobial catheters performed Physiological Assessment and Chronic Health Eval-
in high risk or ICU settings.43e45 All included eco- uation (APACHE) II, Simplified Acute Physiology
nomic models informed by a combination of Score (SAPS), Logistic Organ Dysfunction (LOD)
patient-level data from randomised studies and score] and on-unit parameters (3 day recalibrated
outcome data from other sources. Models were outcome score). Adjustments have also been made
tested using multivariate sensitivity analysis across for confounders occurring on the unit prior to the
a range of clinical and economic assumptions. All CRBSI that impact on mortality using a day 3 or
concluded that antimicrobial catheters were day 4 LOD score. Studies that employed these so-
cost-effective. None was performed in the UK. An phisticated approaches have generally not been
estimate has been made of the cost-effectiveness able to show significant effects of CRBSI on mortal-
of using antimicrobial catheters to prevent CRBSIs ity when all organisms are included.26,47,49,51 One
in the UK using a simple economic model.8 The large study performed a subgroup analysis and
model used parameter estimates derived from reported no attributable mortality even when
published studies with an incidence of CRBSI at CoNS were excluded, either for S. aureus or for
3% (range: 2e5%), relative risk reduction with anti- Gram-negative CRBSI alone.47
microbial catheters of 54% (range: 38e66%), cost At an institutional level a number of additional
of CRBSI of £9,148 (range: £2,500e£71,000) and questions may well be raised before introducing
a price differential between normal and antimicrobial catheters. For example, it is
Intravascular catheter infections 329

probable that meta-analyses of randomised stud- 8. Hockenhull JC, Dwan K, Boland A, et al. The clinical effec-
ies have been able to show a significant benefit tiveness and cost-effectiveness of central venous catheters
treated with anti-infective agents in preventing blood-
only because more than half of CRBSIs are CoNS; stream infections: a systematic review and economic eval-
although a significant clinical burden in a neonatal uation. Health Technol Assess 2008;12:iiieiv. xiexii, 1e154.
unit and perhaps ambulatory patients with long- 9. Safdar N, Fine JP, Maki DG. Meta-analysis: methods for diag-
term catheters, CoNS are less significant in adult nosing intravascular device-related bloodstream infection.
critical care. There is also concern about resis- Ann Intern Med 2005;142:451e466.
10. Timsit JF. Diagnosis and prevention of catheter-related
tance to both antiseptics and antibiotics; qacA/B infections. Curr Opin Crit Care 2007;13:563e571.
and other efflux pumps conferring in vitro resis- 11. Crnich CJ, Maki DG. The role of intravascular devices in
tance to cationic biocides including chlorhexidine sepsis. Curr Infect Dis Rep 2001;3:496e506.
are carried by both CoNS and increasingly in 12. Maki DG, Kluger DM, Crnich CJ. The risk of bloodstream
some strains of MRSA.52 Resistance to rifampicin infection in adults with different intravascular devices:
a systematic review of 200 published prospective studies.
has been noted in CoNS at the catheter site in Mayo Clin Proc 2006;81:1159e1171.
one ICU study.53 Two studies have also noted an in- 13. Wenzel R, editor. Prevention and control of nosocomial
creased risk of catheter colonisation with candida infections. Baltimore: Lippincott Williams & Wilkins; 2003.
[10/187 (5.2%) vs 2/180 (1.1%); relative risk: 4.9; 14. Linares J, Sitges-Serra A, Garau J, Perez JL, Martin R.
95% CI: 1.07e22.2].54,55 It may be tempting to Pathogenesis of catheter sepsis: a prospective study with
quantitative and semiquantitative cultures of catheter
wait for a larger, more comprehensive study to hub and segments. J Clin Microbiol 1985;21:357e360.
be performed, but in an era of reduced incidence 15. Maki DG, Stolz SM, Wheeler S, Mermel LA. Prevention of
of CRBSI in institutions that have re-enforced basic central venous catheter-related bloodstream infection by
infection control measures e arguably the setting use of an antiseptic-impregnated catheter. A randomized,
where such studies should be conducted e it is controlled trial. Ann Intern Med 1997;127:257e266.
16. Raad I, Costerton W, Sabharwal U, Sacilowski M, Anaissie E,
estimated that 10 000 patients would be required Bodey GP. Ultrastructural analysis of indwelling vascular
in each study arm to address the methodological catheters: a quantitative relationship between luminal col-
concerns of previous investigations, and so such onization and duration of placement. J Infect Dis 1993;168:
a study is unlikely to take place.8 400e407.
17. Bisognano C, Vaudaux P, Rohner P, Lew DP, Hooper DC.
Induction of fibronectin-binding proteins and increased
Conflict of interest statement adhesion of quinolone-resistant Staphylococcus aureus
None declared. by subinhibitory levels of ciprofloxacin. Antimicrob
Agents Chemother 2000;44:1428e1437.
18. Stewart PS, Rayner J, Roe F, Rees WM. Biofilm penetration
Funding sources and disinfection efficacy of alkaline hypochlorite and chloro-
None. sulfamates. J Appl Microbiol 2001;91:525e532.
19. Francois P, Schrenzel J, Stoerman-Chopard C, et al. Identi-
fication of plasma proteins adsorbed on hemodialysis tubing
that promote Staphylococcus aureus adhesion. J Lab Clin
References Med 2000;135:32e42.
20. Patti JM, Allen BL, McGavin MJ, Hook M. MSCRAMM-medi-
1. Raad I, Hanna H, Maki D. Intravascular catheter-related in- ated adherence of microorganisms to host tissues. Annu
fections: advances in diagnosis, prevention, and manage- Rev Microbiol 1994;48:585e617.
ment. Lancet Infect Dis 2007;7:645e657. 21. Gray ED, Peters G, Verstegen M, Regelmann WE. Effect of
2. Jarvis WR, Edwards JR, Culver DH, et al. Nosocomial infec- extracellular slime substance from Staphylococcus epider-
tion rates in adult and pediatric intensive care units in the midis on the human cellular immune response. Lancet
United States. National nosocomial infections surveillance 1984;1:365e367.
system. Am J Med 1991;91:185Se191S. 22. Rupp ME, Ulphani JS, Fey PD, Mack D. Characterization of
3. Raad II, Bodey GP. Infectious complications of indwelling Staphylococcus epidermidis polysaccharide intercellular
vascular catheters. Clin Infect Dis 1992;15:197e208. adhesin/hemagglutinin in the pathogenesis of intravascular
4. Lamas C, Boia M, Eykyn SJ. Osteoarticular infections com- catheter-associated infection in a rat model. Infect Immun
plicating infective endocarditis: a study of 30 cases be- 1999;67:2656e2659.
tween 1969 and 2002 in a tertiary referral centre. Scand J 23. O’Grady NP, Alexander M, Dellinger EP, et al. Guidelines for
Infect Dis 2006;38:433e440. the prevention of intravascular catheter-related infections.
5. Lamas CC, Eykyn SJ. Bicuspid aortic valve e a silent danger: Infect Control Hosp Epidemiol 2002;23:759e769.
analysis of 50 cases of infective endocarditis. Clin Infect Dis 24. Edgeworth JD, Treacher DF, Eykyn SJ. A 25-year study of
2000;30:336e341. nosocomial bacteremia in an adult intensive care unit. Crit
6. Fowler Jr VG, Justice A, Moore C, et al. Risk factors for Care Med 1999;27:1421e1428.
hematogenous complications of intravascular catheter- 25. Orsi GB, Di Stefano L, Noah N. Hospital-acquired, labora-
associated Staphylococcus aureus bacteremia. Clin Infect tory-confirmed bloodstream infection: increased hospital
Dis 2005;40:695e703. stay and direct costs. Infect Control Hosp Epidemiol 2002;
7. Casey AL, Mermel LA, Nightingale P, Elliott TS. Antimicro- 23:190e197.
bial central venous catheters in adults: a systematic review 26. Pittet D, Tarara D, Wenzel RP. Nosocomial bloodstream in-
and meta-analysis. Lancet Infect Dis 2008;8:763e776. fection in critically ill patients. Excess length of stay, extra
330 J. Edgeworth

costs, and attributable mortality. J Am Med Assoc 1994; 41. Betjes MG, van Agteren M. Prevention of dialysis catheter-
271:1598e1601. related sepsis with a citrateetaurolidine-containing lock
27. Rello J, Ricart M, Mirelis B, et al. Nosocomial bacteremia in solution. Nephrol Dial Transplant 2004;19:1546e1551.
a medicalesurgical intensive care unit: epidemiologic char- 42. Quarello F, Forneris G. Prevention of hemodialysis catheter-
acteristics and factors influencing mortality in 111 episodes. related bloodstream infection using an antimicrobial lock.
Intensive Care Med 1994;20:94e98. Blood Purif 2002;20:87e92.
28. Renaud B, Brun-Buisson C. Outcomes of primary and cath- 43. Marciante KD, Veenstra DL, Lipsky BA, Saint S. Which anti-
eter-related bacteremia. A cohort and caseecontrol study microbial impregnated central venous catheter should
in critically ill patients. Am J Respir Crit Care Med 2001; we use? Modeling the costs and outcomes of antimicrobial
163:1584e1590. catheter use. Am J Infect Control 2003;31:1e8.
29. Maki DG, Weise CE, Sarafin HW. A semiquantitative culture 44. Shorr AF, Humphreys CW, Helman DL. New choices for cen-
method for identifying intravenous-catheter-related infec- tral venous catheters: potential financial implications.
tion. N Engl J Med 1977;296:1305e1309. Chest 2003;124:275e284.
30. Pratt RJ, Pellowe CM, Wilson JA, et al. Epic2: National evi- 45. Veenstra DL, Saint S, Saha S, Lumley T, Sullivan SD. Efficacy
dence-based guidelines for preventing healthcare-associ- of antiseptic-impregnated central venous catheters in
ated infections in NHS hospitals in England. J Hosp Infect preventing catheter-related bloodstream infection: a
2007;65(Suppl. 1):S1e64. meta-analysis. J Am Med Assoc 1999;281:261e267.
31. Gastmeier P, Geffers C. Prevention of catheter-related 46. Beyersmann J, Gastmeier P, Grundmann H, et al. Use of
bloodstream infections: analysis of studies published be- multistate models to assess prolongation of intensive care
tween 2002 and 2005. J Hosp Infect 2006;64:326e335. unit stay due to nosocomial infection. Infect Control Hosp
32. Pronovost P, Needham D, Berenholtz S, et al. An interven- Epidemiol 2006;27:493e499.
tion to decrease catheter-related bloodstream infections 47. Blot SI, Depuydt P, Annemans L, et al. Clinical and economic
in the ICU. N Engl J Med 2006;355:2725e2732. outcomes in critically ill patients with nosocomial catheter-
33. Department of Health UK. Going further faster: implement- related bloodstream infections. Clin Infect Dis 2005;41:
ing the saving lives delivery programme. Saving lives: 1591e1598.
a delivery programme to reduce HCAI. London: DoH; 2006. 48. Digiovine B, Chenoweth C, Watts C, Higgins M. The attribut-
34. Ho KM, Litton E. Use of chlorhexidine-impregnated dressing able mortality and costs of primary nosocomial bloodstream
to prevent vascular and epidural catheter colonization and infections in the intensive care unit. Am J Respir Crit Care
infection: a meta-analysis. J Antimicrob Chemother 2006; Med 1999;160:976e981.
58:281e287. 49. Garrouste-Orgeas M, Timsit JF, Tafflet M, et al. Excess risk
35. Walder B, Pittet D, Tramer MR. Prevention of bloodstream of death from intensive care unit-acquired nosocomial
infections with central venous catheters treated with bloodstream infections: a reappraisal. Clin Infect Dis
anti-infective agents depends on catheter type and inser- 2006;42:1118e1126.
tion time: evidence from a meta-analysis. Infect Control 50. Rello J. Impact of nosocomial infections on outcome: myths
Hosp Epidemiol 2002;23:748e756. and evidence. Infect Control Hosp Epidemiol 1999;20:
36. Hanna H, Benjamin R, Chatzinikolaou I, et al. Long-term 392e394.
silicone central venous catheters impregnated with mino- 51. Soufir L, Timsit JF, Mahe C, Carlet J, Regnier B, Chevret S.
cycline and rifampin decrease rates of catheter-related Attributable morbidity and mortality of catheter-related
bloodstream infection in cancer patients: a prospective ran- septicemia in critically ill patients: a matched, risk-
domized clinical trial. J Clin Oncol 2004;22:3163e3171. adjusted, cohort study. Infect Control Hosp Epidemiol
37. Carratala J, Niubo J, Fernandez-Sevilla A, et al. Random- 1999;20:396e401.
ized, double-blind trial of an antibiotic-lock technique for 52. Smith K, Gemmell CG, Hunter IS. The association between
prevention of gram-positive central venous catheter- biocide tolerance and the presence or absence of qac genes
related infection in neutropenic patients with cancer. among hospital-acquired and community-acquired MRSA
Antimicrob Agents Chemother 1999;43:2200e2204. isolates. J Antimicrob Chemother 2008;61:78e84.
38. Garland JS, Alex CP, Henrickson KJ, McAuliffe TL, Maki DG. 53. Chatzinikolaou I, Hanna H, Graviss L, et al. Clinical experi-
A vancomycineheparin lock solution for prevention of noso- ence with minocycline and rifampin-impregnated central
comial bloodstream infection in critically ill neonates with venous catheters in bone marrow transplantation recipi-
peripherally inserted central venous catheters: a prospec- ents: efficacy and low risk of developing staphylococcal re-
tive, randomized trial. Pediatrics 2005;116:e198ee205. sistance. Infect Control Hosp Epidemiol 2003;24:961e963.
39. Henrickson KJ, Axtell RA, Hoover SM, et al. Prevention of 54. Leon C, Ruiz-Santana S, Rello J, et al. Benefits of minocy-
central venous catheter-related infections and thrombotic cline and rifampin-impregnated central venous catheters.
events in immunocompromised children by the use of van- A prospective, randomized, double-blind, controlled, multi-
comycin/ciprofloxacin/heparin flush solution: a random- center trial. Intensive Care Med 2004;30:1891e1899.
ized, multicenter, double-blind trial. J Clin Oncol 2000; 55. Sampath LA, Tambe SM, Modak SM. In vitro and in vivo
18:1269e1278. efficacy of catheters impregnated with antiseptics or
40. Allon M. Prophylaxis against dialysis catheter-related bac- antibiotics: evaluation of the risk of bacterial resistance
teremia with a novel antimicrobial lock solution. Clin Infect to the antimicrobials in the catheters. Infect Control Hosp
Dis 2003;36:1539e1544. Epidemiol 2001;22:640e646.
Journal of Hospital Infection (2009) 73, 331e337
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Noroviruses in healthcare settings: a challenging


problem
M. Koopmans a,b,*
a
Erasmus Medical Centre, Rotterdam, The Netherlands
b
Laboratory for Infectious Diseases, National Institute of Public Health and the Environment, Bilthoven,
The Netherlands

Available online 22 September 2009

KEYWORDS Summary Current knowledge about noroviruses in relation to healthcare-


Healthcare-associated associated infections (HCAIs) merely scratches the surface. Most data come
infections; from outbreak-based studies, which represent only a small piece of the puz-
Norovirus GII4;
zle. Nevertheless, the data available show that the clinical impact of noro-
Norovirus outbreak
viruses is particularly severe in outbreaks in healthcare settings, and that it
may be increasing. Coupled with the projected increases of the population
aged >65 years, especially those needing healthcare, it is timely to consider
noroviruses in discussions around HCAIs. In particular, broadening the scope
from a field mostly discussing bacteriological problems and a more holistic
approach to dealing with HCAIs may be needed to avoid introducing new
risks when trying to deal with the antimicrobial resistance problem.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

Introduction investigator from the National Institutes of Health


discovered small round particles in stool samples
One of the diseases that has been making headlines from children involved in an outbreak in a school
in the popular media in recent years is the condition in Norwalk, Ohio, USA, now known to be the aetio-
popularly known as ‘gastric flu’, caused by noro- logical agent.1 To date, the Norwalk virus is the proto-
viruses. In contrast to what the recent attention type of what has turned out to be a very diverse
would suggest, a clinical syndrome known as ‘winter family of viruses infecting humans and animals.
vomiting disease’ caused by these viruses was
described as early as 1928. Forty years later, an
Properties of noroviruses and
epidemiology
* Corresponding author. Address: Laboratory for Infectious
Diseases, National Institute of Public Health and the Environ-
ment, Antonie van Leeuwenhoeklaan 9, 3720BA Bilthoven,
Many of those active in the healthcare setting have
The Netherlands. Tel.: þ31 30 2743945; fax: þ31 30 2744418. experienced one or more norovirus outbreaks and
E-mail address: marion.koopmans@rivm.nl the challenges they pose to infection control.
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.06.028
332 M. Koopmans

These can be better understood when looking at when looking at aetiology of illnesses for which
some of the basic properties of noroviruses. The a physician is consulted, when the aetiological
typical person can become infected by ingesting fraction of bacterial pathogens is higher, reflecting
only a few virus particles, but then will start to more severe symptoms on average.14 Recent stud-
shed high loads of viruses through stool and vomit.2 ies have shown that clinical impact has been
Viral loads >109 particles/mL of stool have been underestimated, with reports of severe disease
found in the first days following infection.3e5 and mortality associated with norovirus outbreaks
There is some evidence that higher viral loads in risk groups.9,11,15,16 All of these factors merge
are associated with more severe symptoms, but when outbreaks develop in healthcare settings.
other studies have not confirmed this and found It is important to realise that most of what is
similar levels of shedding in persons with asymp- currently known comes from studies of outbreaks
tomatic norovirus infection.3,4,6 Vomiting can be of gastroenteritis caused by noroviruses, whereas
abrupt and violent (projectile vomiting) which data on clinical impact remain sparse.17 This is
contributes to contamination of the environ- particularly true for data on the role of noroviruses
ment.7,8 Virus shedding gradually decreases over as causes of childhood diarrhoea in developing
a period of several weeks, long after people have countries. Diarrhoeal disease remains a major
recovered from illness, but both infection and cause of childhood mortality in these regions,
shedding may become chronic (>1 month) in per- and viruses (notably rotaviruses) play an important
sons with comorbidities.5,9e11 Once in the environ- role. Whether the same applies for noroviruses
ment, noroviruses can remain infectious for a long remains to be seen, and this topic has been identi-
time, depending on the specific conditions and fied as a major data gap by an expert committee
cleaning and disinfection procedures used. commissioned by the World Health Organization
Population-based studies have demonstrated (WHO) to discuss viruses in relation to food safety.
that viral pathogens e particularly rotavirus, noro- In parts of the world with high standards of
virus and sapovirus e are by far the most com- living, the greatest impact is seen in the outbreaks
monly identified causes of gastroenteritis in the caused by noroviruses (Figure 1). As norovirus
community (Figure 1).12,13 This picture changes infection is not a notifiable disease in most

Outbreaks, mild illness


Outbreaks, severe illness
hospital
Community, mild illness
Staff, visitor, patients Virus transmissions
evolution Patients
staff
visitors

Day care center, Elderly Epidemic/


school Home pandemic spread

community

Virus evolution Novel strains


Restaurant,
caterer, Multiple infections,
retail recombination

Introductions via travel, food

Figure 1 Epidemiology of noroviruses.


Noroviruses in healthcare settings 333

countries, it is difficult to extrapolate trends from pathogens. With the globalisation of food produc-
available surveillance data which are mostly based tion and marketing, outbreaks linked to such prod-
on passive reporting of outbreaks with clear differ- ucts may be very difficult to detect and verify.
ences in the set-up in different countries.18 Never- Nevertheless, studies have shown that this is not
theless, the combined analysis of epidemiological uncommon, and recently the global organisation
and molecular data, as has been done by that provides guidance for food industry, the Co-
a Europe-wide collaborative research network dex Alimentarius (www.codexalimentarius.net)
(the Foodborne Viruses in Europe network; has decided to commission the production of
www.fbve.nl), has yielded some interesting new guidelines for control of virus infection via food.
findings.19 In past years, outbreaks frequently
have been reported from healthcare institutions,
both hospitals and homes for elderly (Figure 2). Molecular epidemiology
A sharp increase was witnessed in 2002, co-
inciding with emergence of a specific genogroup II It is important to realise that noroviruses consti-
genotype 4 strain (GII4). This increase was real, tute a widely diverse group. They belong to the
based on data from countries that already had Caliciviridae family, which segregates into five
stable reporting for several years beforehand.20e23 genera containing human and animal pathogens
Noroviruses can be transmitted by direct per- (Figure 3).30 The genus norovirus consists of five
son-to-person contact with persons shedding main genogroups, three of which have been found
viruses. Since the infection route is faecaleoral, to infect humans. Each genogroup can further be
personal hand-washing hygiene is an important segregated into genotypes. The clinical relevance
determinant of risk of spreading e as demon- of this is that population immunity does not even
strated, for example, from analysis of outbreaks protect from infection with related strains, and
on cruise ships.24 Pre- and post-symptomatic shed- infections remain possible. This, coupled with
ding may lead to outbreaks but it is unclear how relatively poor immunogenicity of noroviruses,
relevant this mode of transmission really is.25e28 probably explains why norovirus illness is observed
Outbreak control protocols for workers in health- in people of all age groups. The success of the GII4
care or food industry usually recommend sick leave strains was explained by additional analyses show-
until at least 48 h after resolution of symptoms, ing that they evolve rapidly by mutations in the
under the assumption that people without symp- parts of the viral genome that encode the most
toms are less likely to be spreaders. Whether this surface-exposed parts.31e33 By doing so, these vi-
assumption is correct is unknown. An extension ruses change their antigenic make-up and possibly
of this mode of spread is contamination by food also the way they bind to the human host
handlers preparing or serving food. At least 20% (Figure 1). This finding resembles what is known
of outbreaks reported in the FBVE network are about influenza A viruses, where human vaccines
thought to be food-related, many of them linked are updated frequently to match the immunity
to food handlers.29 More difficult to detect are needed to protect from illness due to circulating
diffuse outbreaks that may occur when a food is strains. Like influenza, the successful norovirus
contaminated higher up in the production chain, strains have been found across the world, spread-
for instance with oysters and mussels or berries. ing rapidly once they emerge in one continent. It
Here, contamination with sewage of water used is unclear what drives this evolutionary process
in production leads to presence of multiple but immune-driven selection of strains is the
most plausible explanation, somewhat contrasting
the prior notion of poor immunogenicity of norovi-
ruses. Detailed knowledge of the molecular diver-
sity in the norovirus genus is required to allow the
use of molecular typing in support of investigations
Elderly homes (Figure 3).
Hospitals
Daycare
Hotel/catering Clinical impact
Other
The disease caused by noroviruses is popularly
Figure 2 Most commonly reported settings for noro- known as ‘gastric flu’. Typically symptoms develop
virus outbreaks. Source: the Foodborne Viruses in abruptly with acute onset of vomiting, and some-
Europe network (1112 outbreaks).19 times fever as soon as 8 h after exposure. Vomiting
334 M. Koopmans

Example Relevance of typing Routine

Family Caliciviridae Identification of new viruses No

Genus(5) Norovirus Identification of new viruses No

Species / genogroup (5) GII Potential for zoonotic infection No

Subtype/ genotype (>40) GGIL4 Monitoring of trends, surveillance, Some countries


international comparison

Lineage / variant (only in GII4?) GGIL4 2002 Surveillance, new epidemics, Some countries
virulence, international comparison

Outbreak/ patient strain Hu/NV/Amsterdaml/2002/NL Hospital epidemiology, source Rare


tracing

Figure 3 Taxonomic structure of noroviruses within the family Caliciviridae, and possible questions to address using
genetic typing at different taxonomic levels.

may be violent, and is seen as one of the effective infection have been reported, but clinical research
ways of spreading viruses into/onto the envir- into these pathogens is limited. With the increased
onment of a patient. Studies in which environmen- media attention for outbreaks following the epi-
tal surfaces were tested for viral RNA by using demics with GII4 strains, the number of hospital-
reverse transcriptionepolymerase chain reaction based laboratories offering services for norovirus
tests on cotton swabs have confirmed widespread diagnostics is rapidly increasing, and new data on
contamination.7,8 Even if it is not possible to test clinical impact of noroviruses can be expected in
for infectivity, presence of viral RNA does demon- the coming years.17 Carefully designed studies
strate that the environment has been contami- are needed because noroviruses are such a diverse
nated with faeces or vomit. The consequences of group of pathogens, and viruses belonging to dif-
this may be protracted outbreaks such as those ferent genotypes may behave differently. For in-
demonstrated on some cruise ships.34 Here, new stance, GII4 strains are more often found as
groups of passengers have become infected re- causes of outbreaks in healthcare settings than
peatedly following an initial outbreak. non-GII4 noroviruses, whereas the genogroup I
Norovirus patients usually have vomiting for 1e2 strains are more common in outbreaks related to
days on average, but diarrhoea may last longer, food or environmental contamination, pointing to
especially in children. In a community-based study possible differences in virus behaviour that may
of acute gastroenteritis in The Netherlands, about affect their epidemiology.19 In waste water, geno-
25% of all cases had symptoms lasting a week or group I strains are found almost as abundantly as
more.35 Longer duration of illness has been re- genogroup II viruses, whereas the latter are much
ported for young children and elderly.10,36 Several more frequently found as causes of human disease.
studies have shown that symptoms may be more This indicates possible differences in virulence of
severe in patients with underlying illnesses, based viruses belonging to different clades.40 At present,
on outbreaks in hospitals.9,11,36,37 A recent finding research addressing clinical impact in healthcare
is that chronic infection with norovirus is much settings should focus on GII noroviruses, particu-
more common than previously recognised. Chronic larly the recombinant strains that have emerged
diarrhoea and shedding has been observed in im- as the most common norovirus in childhood diar-
munocompromised individuals, but it is unknown rhoea, recognised by a combination of genes la-
how common this is, or if it is associated with belled IIb pol/II3 capsid (IIbp/II3c).41 Remarkably,
specific deficiencies in the immune system. Non- IIbp/II3c strains were first recognised following
specific symptoms such as vertigo, thirst, and a large international foodborne outbreak in
headaches after resolution of vomiting and diar- 2001.42 The capsid gene sequences matching the
rhoea may delay full recovery in elderly.38,39 No IIb polymerase gene have never been identified
other extraintestinal manifestations of norovirus from human surveillance, raising questions about
Noroviruses in healthcare settings 335

its origin. Genetic recombination is common, throughout that time period.41 These numbers
occurring when viruses belonging to the same may be biased, however, and need to be evaluated
genogroup exchange parts of their genes when in other studies. Nevertheless, low estimates of the
a host cell is infected simultaneously with two proportion of the population with impaired immu-
parent viruses (Figure 1). Noroviruses clustering nity are 1e2%, and knowing that one in four persons
within genogroup II have been identified in pigs, is likely to contract a norovirus infection each year
and pigs can be infected with human noroviruses, suggests that there may be numerous patients with
raising speculation about their potential as reser- chronic infection. An important question to answer
voir for new viruses, or for viruses resulting from is what are the best ways of controlling the disease
recombination between human and pig norovirus in healthcare settings. Clearly, we are faced with
strains (Figure 3).43e45 At present, there is no evi- multiple possible sources of virus triggering out-
dence that recombination occurs between human breaks. These could be any of the patients, staff
and animal strains, but monitoring of trends of or visitors walking into the hospital given the high
human illness should include characterisation of incidence of norovirus in the community.49 Addi-
a systematic sample of strains to increase our tional sources may be long-stay patients in the hos-
understanding of the complex epidemiology of pital, or food brought in by visitors. Understanding
these viruses. An additional reason for doing so the details of norovirus epidemiology will help to
comes from studying related animal viruses design more evidence-based strategies to reduce
belonging to the same family: there are examples their impact.
of highly successful species jumps within the vesi-
virus genus and the lagovirus genus, demonstrating
that caliciviruses are quite flexible.46 Therefore, Issues around infection control
from a virological perspective it is prudent to
monitor what is going on. Because norovirus infection is so common, pre-
vention of introduction of the virus in a healthcare
setting is an unrealistic goal. One could speculate
Specific issues related to norovirus in about the possibility of screening patients at
healthcare settings admission, but that requires rapid tests with very
high sensitivity to be effective. A recent study
The above shows that there are three important comparing infection control protocols of different
reasons for wanting to control norovirus outbreaks level of stringency was inconclusive due to poor
in healthcare settings. First, nosocomial norovirus compliance to a particular protocol. The only
infection may prolong the duration of hospital- measure that seemed to have an effect was the
isation and cause complications in vulnerable per- speed of starting any protocol, i.e. taking di-
sons, which should be avoided. Second, control of arrhoea and vomiting in one or two patients as
outbreaks may be costly when not implemented a trigger for enhanced hygienic measures.50
immediately, not only in terms of added cost for Another issue of some debate among virologists
cleaning and disinfection, but also in terms of is whether alcohol-based hand hygiene products
absenteeism of staff, longer hospitalisation of have sufficient activity against these viruses.51 The
patients, long-term treatment of patients that problem is that noroviruses cannot be grown in cell
develop chronic disease, closure of wards including culture, so that experimental inactivation studies
intensive care units, and postponement of surgery.47,48 are not possible.52 Model viruses have been used
The third reason is that in patients with chronic in- but their validity has been disputed because
fection the viruses can evolve by accumulation of specific characteristics can differ substantially
mutations.11 Besides the health problems for the between highly related viruses. The WHO recom-
individual patient, these persons therefore may mends the use of alcohol-based hand sanitation
eventually be the reservoir in which new variants globally, because of its advantage over no hand
develop, thus perpetuating the cycle of infection sanitation at all, e.g. in areas where access to
(Figure 1). Although this is speculative at this stage, clean water is limited.53 Data on virus inactivation
the size of the group that is at risk for chronic dis- for the diarrhoeal viruses where available shows
ease due to this ‘simple’ diarrhoea virus is large some effect on virus infectivity and thus the use
and expanding. A five-year survey in which patients of alcohol-based hand hygiene products is likely
diagnosed with norovirus infection were tested to have some advantage for these viruses as
with at least a one-month interval found chronic in- well.54 Virologists, however, do recommend the
fection in 22% of all patients in this group, which use of regular hand washing with water and soap
was 6% of all patients diagnosed with norovirus during outbreak situations.
336 M. Koopmans

In conclusion, the recent increase in incidence 13. Wheeler JG, Sethi D, Cowden JM, et al. Study of infectious
of norovirus outbreaks has triggered studies show- intestinal disease in England: rates in the community,
presenting to general practice, and reported to national
ing that the clinical impact of noroviruses is surveillance. The Infectious Intestinal Disease Study Exe-
greater than previously recognised. A clear picture cutive. Br Med J 1999;318:1046e1050.
emerges: outbreaks of norovirus illness in health- 14. de Wit MA, Kortbeek LM, Koopmans MP, et al. A comparison of
care settings may be disruptive and costly, and can gastroenteritis in a general practice-based study and a com-
cause significant complications including chronic munity-based study. Epidemiol Infect 2001;127:389e397.
15. Jansen A, Stark K, Kunkel J, et al. Aetiology of community-
illness and deaths. This should be sufficient moti- acquired, acute gastroenteritis in hospitalised adults:
vation for swift action in the face of new out- a prospective cohort study. BMC Infect Dis 2008;8:143.
breaks, using stringent infection control measures. 16. Harris JP, Edmunds WJ, Pebody R, Brown DW, Lopman BA.
Deaths from norovirus among the elderly, England and
Wales. Emerg Infect Dis 2008;14:1546e1552.
17. Koopmans M. Progress in understanding norovirus epidemi-
Acknowledgements ology. Curr Opin Infect Dis 2008;21:544e552.
18. Kroneman A, Harris J, Vennema H, et al. Data quality of
I am greatly indebted to my coworkers at the RIVM, 5 years of central norovirus outbreak reporting in the Euro-
Erasmus MC, and the FBVE network for the contin- pean Network for food-borne viruses. J Public Health (Oxf)
uous efforts to advance our understanding of 2008;30:82e90.
19. Kroneman A, Verhoef L, Harris J, et al. Analysis of inte-
norovirus.
grated virological and epidemiological reports of norovirus
outbreaks collected within the foodborne viruses in Europe
Network from 1 July 2001 to 30 June 2006. J Clin Microbiol
References 2008;46:2959e2965.
20. Bruggink L, Marshall J. Molecular changes in the norovirus
1. Kapikian AZ. The discovery of the 27 nm Norwalk virus: an polymerase gene and their association with incidence of
historic perspective. J Infect Dis 2000;181:S295eS302. GII.4 norovirus-associated gastroenteritis outbreaks in Vic-
2. Teunis PF, Moe CL, Liu P, et al. Norwalk virus: how infec- toria, Australia, 2001e2005. Arch Virol 2008;153:729e732.
tious is it? J Med Virol 2008;80:1468e1476. 21. Widdowson MA, Cramer EH, Hadley L, et al. Outbreaks of
3. Lee N, Chan MC, Wong B, et al. Fecal viral concentration acute gastroenteritis on cruise ships and on land: identifica-
and diarrhea in norovirus gastroenteritis. Emerg Infect Dis tion of a predominant circulating strain of norovirus e
2007;13:1399e1401. United States, 2002. J Infect Dis 2004;190:27e36.
4. Atmar RL, Opekun AR, Gilger MA, et al. Norwalk virus shed- 22. Lopman B, Vennema H, Kohli E, et al. Increase in viral gas-
ding after experimental human infection. Emerg Infect Dis troenteritis outbreaks in Europe and epidemic spread of
2008;14:1553e1557. new norovirus variant. Lancet 2004;363:682e688.
5. Tu ET, Bull RA, Kim MJ, et al. Norovirus excretion in an 23. Siebenga JJ, Vennema H, Duizer E, Koopmans MP. Gastroen-
aged-care setting. J Clin Microbiol 2008;46:2119e2121. teritis caused by norovirus GGII.4, The Netherlands,
6. Ozawa K, Oka T, Takeda N, Hansman GS. Norovirus infection 1994e2005. Emerg Infect Dis 2007;13:144e146.
in symptomatic and asymptomatic food handlers in Japan. 24. Chimonas MA, Vaughan GH, Andre Z, et al. Passenger be-
J Clin Microbiol 2007;45:3996e4005. havior associated with norovirus infection on board a cruise
7. Wu HM, Fornek M, Schwab KJ, et al. A norovirus outbreak at ship e Alaska. J Travel Med 2008;15:177e183.
a long term care facility: the role of environmental surface 25. Lo SV, Connolly AM, Palmer SR, Wright D, Thomas PD,
contamination. Infect Control Hosp Epidemiol 2005;26: Joynson D. The role of the pre-symptomatic food handler
802e810. in a common source outbreak of food-borne SRSV gastroen-
8. Boxman IL, Dijkman R, te Loeke NA, et al. Environmental teritis in a group of hospitals. Epidemiol Infect 1994;113:
swabs as a tool in norovirus outbreak investigation, including 513e521.
outbreaks on cruise ships. J Food Prot 2009;72:111e119. 26. Parashar UD, Dow L, Fankhauser RL, et al. An outbreak of
9. Ludwig A, Adams O, Laws HJ, Schroten H, Tenenbaum T. viral gastroenteritis associated with consumption of sand-
Quantitative detection of norovirus excretion in pediat- wiches: implications for the control of transmission by food
ric patients with cancer and prolonged gastroenteritis handlers. Epidemiol Infect 1998;121:615e621.
and shedding of norovirus. J Med Virol 2008;80: 27. Götz H, de JB, Lindbäck J, et al. Epidemiological investiga-
1461e1467. tion of a food-borne gastroenteritis outbreak caused by
10. Murata T, Katsushima N, Mizuta K, Muraki Y, Hongo S, Norwalk-like virus in 30 day-care centres. Scand J Infect
Matsuzaki Y. Prolonged norovirus shedding in infants 6 Dis 2002;34:115e121.
months of age with gastroenteritis. Pediatr Infect Dis 28. Patterson T, Hutchings P, Palmer S. Outbreak of SRSV gas-
J 2007;26:46e49. troenteritis at an international conference traced to food
11. Siebenga JJ, Beersma MF, Vennema H, van Biezen P, handled by a post-symptomatic caterer. Epidemiol Infect
Hartwig NJ, Koopmans M. High prevalence of prolonged 1993;111:157e162.
norovirus shedding and illness among hospitalized 29. Verhoef LP, Kroneman A, van Duynhoven Y, Boshuizen H,
patients: a model for in vivo molecular evolution. J Infect van Pelt W, Koopmans M. Foodborne viruses in Europe Net-
Dis 2008;198:994e1001. Erratum in: J Infect Dis 2008; work. Selection tool for foodborne norovirus outbreaks.
198:1575. Emerg Infect Dis 2009;15:31e38.
12. de Wit MA, Koopmans MP, Kortbeek LM, et al. Sensor, a pop- 30. Green KY, Chanock RM, Kapikian AZ. Human caliciviruses.
ulation-based cohort study on gastroenteritis in the Nether- In: Knipe DM, Howley PM, editors. Fields’ virology. 4th
lands: incidence and etiology. Am J Epidemiol 2001;154: edn. Philadelphia: Lippincott Williams & Wilkins; 2001. p.
666e674. 841e874.
Noroviruses in healthcare settings 337

31. Lindesmith LC, Donaldson EF, Lobue AD, et al. Mechanisms 43. Sugieda M, Nakajima S. Viruses detected in the caecum con-
of GII.4 norovirus persistence in human populations. PLoS tents of healthy pigs representing a new genetic cluster in
Med 2008;5:e31. genogroup II of the genus ‘‘Norwalk-like viruses’’. Virus
32. Siebenga JJ, Vennema H, Renckens B, et al. Epochal evolu- Res 2002;87:165e172.
tion of GGII.4 norovirus capsid proteins from 1995 to 2006. 44. Wang QH, Han MG, Cheetham S, Souza M, Funk JA, Saif LJ.
J Virol 2007;81:9932e9941. Porcine noroviruses related to human noroviruses. Emerg
33. Allen DJ, Gray JJ, Gallimore CI, Xerry J, Iturriza-Gómara M. Infect Dis 2005;11:1874e1881.
Analysis of amino acid variation in the P2 domain of the 45. Cheetham S, Souza M, Meulia T, Grimes S, Han MG, Saif LJ.
GII-4 norovirus VP1 protein reveals putative variant-specific Pathogenesis of a genogroup II human norovirus in gnoto-
epitopes. PLoS ONE 2008;3:e1485. biotic pigs. J Virol 2006;80:10372e10381.
34. Verhoef L, Depoortere E, Boxman I, et al. Emergence of 46. Smith AW, Skilling DE, Cherry N, Mead JH, Matson DO. Cal-
new norovirus variants on spring cruise ships and prediction icivirus emergence from ocean reservoirs: zoonotic and
of winter epidemics. Emerg Infect Dis 2008;14:238e243. interspecies movements. Emerg Infect Dis 1998;4:13e20.
35. Rockx B, De Wit M, Vennema H, et al. Natural history of 47. Hansen S, Stamm-Balderjahn S, Zuschneid I, et al. Closure
human calicivirus infection: a prospective cohort study. Clin of medical departments during nosocomial outbreaks: data
Infect Dis 2002;35:246e253. from a systematic analysis of the literature. J Hosp Infect
36. Lopman BA, Reacher MH, Vipond IB, Sarangi J, Brown DW. 2007;65:348e353.
Clinical manifestations of norovirus in health care settings. 48. Lopman BA, Reacher MH, Vipond IB, et al. Epidemiology and
Clin Infect Dis 2004;39:318e324. cost of nosocomial gastroenteritis, Avon, England,
37. Mattner F, Sohr D, Heim A, Gastmeier P, Vennema H, 2002e2003. Emerg Infect Dis 2004;10:1827e1834.
Koopmans M. Risk groups for clinical complications of noro- 49. Mattner F, Mattner L, Borck HU, Gastmeier P. Evaluation of
virus infections: an outbreak investigation. Clin Microbiol the impact of the source (patient versus staff) on nosoco-
Infect 2006;12:69e74. mial norovirus outbreak severity. Infect Control Hosp
38. Tsang OT, Wong AT, Chow CB, Yung RW, Lim WW, Liu SH. Epidemiol 2005;26:268e272.
Clinical characteristics of nosocomial norovirus in Hong 50. Friesema IHM, Vennema H, Heijne JCM, et al. Norovirus
Kong. J Hosp Infect 2008;69:135e140. outbreaks in nursing homes. The evaluation of infection control
39. Goller JL, Dimitriadis A, Tan A, Kelly H, Marshall JA. Long measures. Epidemiol. Infect 2009 (e-pub ahead of print).
term features of norovirus gastroenteritis in the elderly. 51. Lages SL, Ramakrishnan MA, Goyal SM. In vivo efficacy of
J Hosp Infect 2004;58:286e291. hand sanitisers against feline calicivirus: a surrogate for
40. da Silva AK, Le Saux JC, Parnaudeau S, Pommepuy M, norovirus. J Hosp Infect 2008;68:159e163.
Elimelech M, Le Guyader FS. Evaluation of removal of noro- 52. Duizer E, Schwab KJ, Neill FH, Atmar RL, Koopmans MP,
viruses during waste water treatment, using RTePCR: dif- Estes MK. Laboratory efforts to cultivate noroviruses.
ferent behaviors of GI and GII. Appl Environ Microbiol J Gen Virol 2004;85:79e87.
2007;73:7891e7897. 53. World Alliance for Patient Safety. WHO guidelines on hand
41. Beersma MF, Schutten M, Vennema H, et al. Norovirus in hygiene in health care (advanced draft). Geneva: WHO;
a Dutch tertiary care hospital (2002e2007): frequent noso- 2006.
comial transmission and dominance of GIIb strains in young 54. Sandora TJ, Shih MC, Goldmann DA. Reducing absenteeism
children. J Hosp Infect 2009;71:199e205. from gastrointestinal and respiratory illness in elementary
42. Koopmans M, Vennema H, Heersma H, et al. Early identifi- school students: a randomized, controlled trial of an
cation of common-source foodborne virus outbreaks in infection-control intervention. Pediatrics 2008;121:
Europe. Emerg Infect Dis 2003;9:1136e1142. e1555ee1562.
Journal of Hospital Infection (2009) 73, 338e344
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Pseudomonas aeruginosa: a formidable and


ever-present adversary
K.G. Kerr a,*, A.M. Snelling b
a
Department of Microbiology, Harrogate District Hospital, Harrogate, North Yorkshire, UK
b
Division of Biomedical Sciences & Bradford Infection Group, University of Bradford, Bradford,
West Yorkshire, UK

Available online 21 August 2009

KEYWORDS Summary Pseudomonas aeruginosa is a versatile pathogen associated


Healthcare-associated with a broad spectrum of infections in humans. In healthcare settings
infection; the bacterium is an important cause of infection in vulnerable individuals
Hospital environment;
including those with burns or neutropenia or receiving intensive care. In
Infection control;
Pseudomonas
these groups morbidity and mortality attributable to P. aeruginosa infec-
aeruginosa tion can be high. Management of infections is difficult as P. aeruginosa is
inherently resistant to many antimicrobials. Furthermore, treatment is be-
ing rendered increasingly problematic due to the emergence and spread of
resistance to the few agents that remain as therapeutic options. A notable
recent development is the acquisition of carbapenemases by some strains
of P. aeruginosa. Given these challenges, it would seem reasonable to iden-
tify strategies that would prevent acquisition of the bacterium by hospital-
ised patients. Environmental reservoirs of P. aeruginosa are readily
identifiable, and there are numerous reports of outbreaks that have been
attributed to an environmental source; however, the role of such sources in
sporadic pseudomonal infection is less well understood. Nevertheless there
is emerging evidence from prospective studies to suggest that environmen-
tal sources, especially water, may have significance in the epidemiology of
sporadic P. aeruginosa infections in hospital settings, including intensive
care units. A better understanding of the role of environmental reservoirs
in pseudomonal infection will permit the development of new strategies
and refinement of existing approaches to interrupt transmission from these
sources to patients.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

* Corresponding author. Address: Department of Microbiology, Harrogate District Hospital, Lancaster Park Road, Harrogate, North
Yorkshire HG2 7SX, UK. Tel.: þ44 1423 553077.
E-mail address: kevin.kerr@hdft.nhs.uk

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.04.020
Prevention and control of P. aeruginosa 339

Introduction Patients in critical care units are particularly


at risk of these infections, partly due to the
Pseudomonas aeruginosa is associated with an presence of invasive devices and partly because of
ever-widening spectrum of infections. Some mani- underlying disease and comorbidities. The mortality
festations of P. aeruginosa infection occur pre- attributable to P. aeruginosa infection such as
dominantly in the community although the ventilator-associated pneumonia or bacteraemia
majority are seen in healthcare settings especially is substantial, especially for patients who receive
in immunosuppressed individuals and other highly inadequate empirical therapy.1e3 Morbidity also
vulnerable patients such as those in the intensive may be considerable. For example, post-surgical
care unit (ICU). Infections may be associated endophthalmitis is a feared complication which
with significant morbidity and mortality and thera- may, even if treated promptly, result in significant
peutic options are becoming increasingly limited visual impairment or even total loss of sight.4
with the continued emergence and spread of anti- As well as their undoubted clinical importance,
microbial resistant strains. Given the potential P. aeruginosa infections are also quantitatively sig-
severity of specific P. aeruginosa infections and nificant as has been noted in numerous reports.
problems in selecting optimal therapy for multi- Recently, the US National Healthcare Safety Net-
drug resistant (MDR) isolates, identification and work (which subsumes the National Nosocomial
implementation of effective strategies to prevent Infection Surveillance system) published data on
these infections are urgent priorities. >28 000 cases of nosocomial infections from 463
hospitals gathered over a 22 month period.5 Over-
all, P. aeruginosa was the sixth most frequently
occurring pathogen, the second commonest cause
Clinical features of P. aeruginosa of ventilator-associated pneumonia and the
infection seventh commonest cause of catheter-related
bloodstream infection.
There can be no doubt about the versatility of
P. aeruginosa as a pathogen. The list of infections
associated with this bacterium is long and continues Virulence factors of P. aeruginosa
to grow. Some infections are more likely to be en-
countered in community settings. These include An impressive arsenal of virulence factors can be
folliculitis and the green nail or hot foot syndromes deployed by P. aeruginosa. This extensive armoury
following recreational exposure to water sources partly explains why the range of infections caused
such as hot tubs, Jacuzzis and swimming pools, by the bacterium is so wide. A full discussion of
septic arthritis (especially of smaller joints) in established and putative virulence factors is beyond
injecting drug users and otitis externa. The last the scope of this paper, but they include enzymes
may manifest as severe, potentially life-threaten- such as proteases and elastases and phenazine pig-
ing malignant otitis externa in patients with ments, such as pyocyanin and rhamolipids. The bac-
diabetes and immunocompromised individuals. terium possesses several different export systems
Underlying pulmonary disease, such as bronchi- that are involved in the secretion of virulence fac-
ectasis and cystic fibrosis, is a predisposing factor tors. Of particular importance is a type III secretion
for P. aeruginosa respiratory tract infections. Most system which enables injection of effector proteins
serious P. aeruginosa infections are seen in health- directly into the cytoplasm of host cells.6 Four ef-
care settings and include bacteraemia, pneumonia, fector proteins have been identified: ExoS and
urosepsis and wound infection including secondary ExoT, which are bifunctional enzymes with GTPase
infection of burns. Some patient groups are more activating protein and ADP ribosylase activities
at risk of acquiring infection than others. These (which ultimately cause disruption of the host cell
include immunocompromised patients, especially cytoskeleton) and ExoU (a cytotoxin with phospho-
those with chemotherapy-induced neutropenia lipase activity) and Exo Y (an adenylate cyclase).
who are at particular risk of pseudomonal blood- Different combinations of these proteins have
stream infections; hence empiric antimicrobial profound and often devastating effects on epi-
therapy for febrile neutropenia should include at thelial barrier function and wound healing. It is
least one agent with anti-pseudomonal activity. also thought that they interfere with the function-
Physical breaches in host defences such as those ing of macrophages and neutrophils.
which follow surgical incisions, insertion of urinary The ability of P. aeruginosa to form biofilm is
and vascular catheters and endotracheal tubes also important for the ability of the bacterium to per-
predispose patients to P. aeruginosa infection. sist in environmental niches such as pipes and
340 K.G. Kerr, A.M. Snelling

taps.7 When growing as a complex mass of cells at- did not have an antibiotic policy and that 43%
tached to a surface, P. aeruginosa cells can be sig- (6/14) did not routinely isolate patients colonised
nificantly more resistant to biocides than when or infected with MDR strains of P. aeruginosa.11
they are in a planktonic (free-floating) state.8 Perhaps the most concerning development in
This may have significant consequences for hospi- recent years has been the emergence of carbapen-
tal cleaning regimens. emases in MDR strains of P. aeruginosa.15 Loss of
Biofilms can also occur inside the host and are the carbapenems, which have been the mainstay
associated with the pathogenesis of chronic lung of therapy for infections caused by MDR strains,
infections in patients with cystic fibrosis and severely limits therapeutic options, with colistin
bronchiectasis as well as infections associated becoming the ‘antibiotic of last resort’. The
with indwelling devices and other prosthetic emergence of colistin resistance in carbapenem-
material. resistant P. aeruginosa, creating effectively pan-
resistant strains, is thus a serious development.
This is highlighted by an outbreak of pan-resistant
Antimicrobial therapy of P. aeruginosa P. aeruginosa in the ICU of a cancer centre in
infection Bratislava. Five of six patients with P. aeruginosa
(colistin minimum inhibitory concentration:
Pseudomonas aeruginosa is intrinsically resistant to 4 mg/L) bacteraemia died.16
several classes of antibiotics, thus limiting
therapeutic options. Unfortunately, antimicrobial
therapy is becoming even more problematic due Pseudomonas aeruginosa in the
to acquired or mutational resistance. A wide range hospital environment
of resistance mechanisms have been identified
including multi-drug efflux pumps (which can confer Pseudomonas aeruginosa is capable of colonising
resistance to, among others, cephalosporins, a range of niches such as the phyllosphere and rhi-
ureidopenicillins, fluoroquinolones and aminogly- zosphere of plants, soil and a wide range of
cosides), aminoglycoside-modifying enzymes, aquatic habitats. Although the bacterium is gener-
b-lactamases, and target site modifications, e.g. ally not considered a component of the normal re-
of DNA gyrase and topoisomerase IV following gional flora of humans, faecal and e to a lesser
mutations in gyrA and parC, respectively.9 Resis- extent e oropharyngeal and skin carriage is not
tance to a particular class may be mediated by sev- uncommon.
eral mechanisms. For example, isolates may lose The range of reservoirs in healthcare environ-
susceptibility to a carbapenem due to production ments from which P. aeruginosa has been isolated
of a carbapenemase, loss of the OprD porin, or via is wide ranging, with examples listed in Table I.
a multidrug efflux pump. A number of recently pub- Sites most likely to harbour the bacterium are wa-
lished studies have highlighted the growing problem ter-related (e.g. taps and showers) or where mois-
of antimicrobial resistance in P. aeruginosa, and in ture or humidity is high (e.g. respiratory therapy
particular the emergence of MDR (resistance to 3 equipment). Several properties of P. aeruginosa
classes), extensively drug-resistant (resistance to favour persistence in the hospital environment.
all but one or two classes) or even pandrug-resistant The bacterium is inherently resistant to several
strains. Souli et al. recently published data from disinfectants such as biguanides and quaternary
23 countries on the European Antimicrobial ammonium compounds through the action of
Resistance Surveillance System (EARSS) website.10 multidrug efflux pumps. Furthermore, the ability
Resistance rates for aminoglycosides, carbape- to form biofilm on a range of inanimate surfaces
nems, quinolones and ceftazidime were 0e51.9%; also contributes to disinfectant resistance as well
9e50.5%; 7.2e51.9% and 4e48.5%, respectively. as impeding physical removal. Persistence in moist
Overall, 18% of isolates were recorded as being environments is also favoured by the use of the
MDR. type III secretion system to kill free-living amoeba
As noted earlier, patients in intensive care are which graze on environmental bacteria.17
especially vulnerable to P. aeruginosa infection The true importance of P. aeruginosa in the
and reports of increasing resistance in this setting hospital environment as it relates to healthcare-
give particular cause for concern.11e14 Given the associated infection (HCAI) remains a controversial
frequent use of antibiotics in this setting, high issue. Even when strains indistinguishable from
resistance rates in ICU-based studies are not unex- clinical isolates are isolated from the ward envir-
pected but Handberger et al. report that half of onment, it can be argued that patients have con-
the units (10/20) which responded to the survey taminated the environment directly or indirectly
Prevention and control of P. aeruginosa 341

hospital-wide intervention to reduce ciprofloxacin


Table I Environmental reservoirs of P. aeruginosa in
hospitals use was associated with a significant reduction in
the numbers of ciprofloxacin-resistant P. aerugi-
Potable water
nosa isolates, but this was tempered by an
Taps/sinks/sink traps
increase in carbapenem-resistant strains.19 In a
Showers
Disinfectants/sanitisers/antiseptics/bar soap five-year study involving 35 ICUs Jonas et al. exam-
Respiratory therapy equipment ined the genodiversity of ciprofloxacin- and imipe-
Ice makers nem-resistant P. aeruginosa isolates and found
Flower vases that this was significantly lower in ICUs with high
Shaving/toothbrushes resistance rates and low antimicrobial usage com-
Medication, e.g. eye drops; multi-dose vials; pared with ICUs with low resistance rates and high
mouthwash antimicrobial use.20 This suggests that high resis-
Mop heads/buckets tance rates in units with low antimicrobial use
Endoscopes/endoscope washers occurs due to more frequent opportunities for
Urometers
cross-transmission. Thus, efforts to reduce the
Water baths
emergence and propagation of antibiotic resis-
Hydrotherapy pools
Infant feeding basins tance in P. aeruginosa through prudent antimicro-
Bathing basins bial use may be undermined if infection control
Bath toys practice is suboptimal.
Cleaning equipment

Infection control
rather than vice versa. Nevertheless, as discussed As illustrated by reports from outbreak investiga-
below there is now accumulating evidence to sug- tions, the importance of standard infection control
gest that the environment can play an important techniques such as good hand hygiene for the
role in the epidemiology of HCAI caused by control and prevention of P. aeruginosa infection
P. aeruginosa. cannot be overemphasised. Following an outbreak
in a neonatal ICU in which 16 babies died from
P. aeruginosa infection, multivariate analysis iden-
P. aeruginosa infection in hospitals: tified two nurses as being significantly associated
prevention is better than cure with colonisation/infection of neonates with the
outbreak strains.21 Furthermore, isolates indistin-
Given the possible consequences of P. aeruginosa guishable on molecular typing from these strains
infection, particularly for vulnerable patients, were cultured from the nurses’ nails, one of
and the continued emergence of MDR strains which whom had long nails and the other who had artifi-
hamper effective antimicrobial therapy, it is clear cial nails. Outbreak control measures included in-
that strategies to prevent infection and reduce creased hand hygiene, banning artificial nails and
the likelihood of antimicrobial resistance should restricting the length of non-artificial nails. These
become a key priority. reduced but did not eliminate further cases of in-
fection.21 Similarly, a scrub nurse with onychmyco-
sis was identified as the likely source of an
Antimicrobial stewardship outbreak of P. aeruginosa infection in a cardio-
thoracic unit. Improved hand hygiene is often
One of the principal elements of an antimicrobial employed as a control measure during outbreaks;
stewardship programme is restriction of certain however, as other interventions, such as isolation
classes of antimicrobials, to try to delay the of colonised or infected patients, are usually insti-
emergence of resistance to these agents. Unfortu- tuted contemporaneously, it may be unclear as to
nately, recent studies have reported only partial which measure (or combination thereof) is most
success with this approach. Ntagiopolous et al. re- successful in preventing the spread of infection.22
stricted the use of ciprofloxacin and ceftazidime
over an 18 month period in an ICU.18 Although
resistance of P. aeruginosa to both agents fell The hospital environment
by the end of the intervention, this was at the
expense of resistance to meropenem which Given the widespread distribution of the bacter-
increased significantly. Similarly, a 34 month ium, it is not surprising that efforts are made to
342 K.G. Kerr, A.M. Snelling

identify the presence of P. aeruginosa in the hospi-


Table II Prospective investigations of P. aeruginosa
tal environment, especially water supplies, sinks, in hospital water: variables in study design
taps and showerheads. Earlier studies were ham-
Frequency of patient sampling
pered by reliance on phenotypic typing techniques
Patient body sites sampled (single versus multiple)
such as serotyping or pyocin typing to compare the
Frequency of water sampling
relatedness of clinical and environmental iso- Nature of environmental sampling (water only; taps
lates.23 The introduction of molecular typing only; water and taps)
methods has permitted improved characterisation Sampling methods (volume of water/pre- vs
of environmental and clinical isolates, strengthen- post-flush/culture media/temperature of
ing evidence that hospital water supplies and out- incubation, etc.)
lets may act as a source of infection.24e28 Number of colonies selected for molecular typing
Nevertheless, the question as to whether water
and water outlets are infecting patients or whether
patients contaminate water outlets cannot be an-
swered definitively by investigations performed in problem. Aumeran et al. reported that replace-
the outbreak setting. Furthermore, sources of pot- ment of shower heads and hoses and cleaning dis-
able water other than mains water should be taken infection failed to control an outbreak of infection
into account when trying to trace reservoirs for in- associated with both P. aeruginosa and P. putida
fection. For example, Eckmanns et al. recently re- on an oncohaematology unit.24 The outbreak was
ported an outbreak of P. aeruginosa infection in six terminated following the chlorination of the hospi-
ICUs in the same hospital.29 Extensive environmen- tal water system and the fitting of point-of-use fil-
tal sampling identified the outbreak strain in both ters on taps and showers. Subsequently, a new
opened and unopened units of commercially avail- distribution system for the unit, in which water
able bottled water. The outbreak was terminated was treated by daily chlorination and passed
when this water was withdrawn. through prefilters (5 mm and 1 mm) and terminal
Several prospective studies have been under- filtration (0.22 mm), was installed.24
taken that acknowledge the limitations of outbreak An outbreak of MDR P. aeruginosa serogroup 011
investigations.30e34 However, despite variations in infection on a neurosurgery ICU was not controlled
study design, these investigations, with some by descaling of taps and replacement of faulty
exceptions, have provided evidence of patient pipework. The outbreak ended only when the
acquisition of P. aeruginosa from hospital water unit was temporarily closed and all the sinks re-
(Table II).35 Rogues et al. investigated a 16-bedded placed.27 Replacement of taps also finally halted
ICU over a 6 month period.36 Weekly tap water spec- an outbreak of P. aeruginosa infection on a paedi-
imens were obtained, as were throat, rectal, spu- atric surgical unit after other measures including
tum and urine cultures from patients on admission chlorination of the water supply and disinfection
and weekly thereafter. Isolates of P. aeruginosa of sinks were unsuccessful.26 Similarly, Hota
were typed using SpeI pulsed-field gel electrophore- et al. were able to terminate an outbreak associ-
sis (PFGE). The authors concluded that P. aerugi- ated with contamination of sink drains only when
nosa carriage by patients was occurring following the sinks were replaced.25 An outbreak of MDR
exposure to contaminated tap water, and that P. aeruginosa infection on an ICU which proved re-
colonised patients were the source of contamina- fractory to control measures, such as enhanced
tion for taps.36 Another study conducted over a 12 barrier and hygiene precautions, cohorting of pa-
month period in five ICUs within the same hospital tients and changes to the unit’s antibiotic policy,
compared patient and tap isolates using the same were not sufficient to end the outbreak.37 Thermal
methodology.31 These were indistinguishable in disinfection of taps and the use of sterile water for
56/132 (42%) cases, again suggesting that in non- patient care purposes had to be instigated.37
outbreak situations water outlets may be an Following a study where P. aeruginosa colonisa-
important reservoir for P. aeruginosa. tion or infection in ICU patients was associated
with contaminated taps, Petignat et al. introduced
a range of measures: an increase in the water tem-
Prevention perature; copper and silver ionisation; replace-
ment of tap water by P. aeruginosa-free bottled
Growing recognition that hospital water represents water for patient use; and reinforcement of stan-
an important source of P. aeruginosa in both dard precautions and hand hygiene.38 Three groups
endemic or outbreak situations has prompted the of colonised/infected patients were identified.
development of interventions to counter this Group 1 comprised patients whose isolate was
Prevention and control of P. aeruginosa 343

genotypically indistinguishable from an isolate techniques have been employed with varying de-
recovered from a tap. Group 2 patients had grees of success, to reduce or eliminate the bacte-
isolates which were indistinguishable from that of rium from these sources and futher evaluations of
an isolate recovered from at least one other pa- these are clearly indicated. However, it should be
tient, but not grown from a tap. Group 3 consisted acknowledged that we cannot rely solely on these
of cases with a unique genotype. The control mea- control measures and that they should be employed
sures led to statistically significant reductions in as part of a comprehensive multifaceted approach
infection/colonisation in group 1 and 2 patients alongside other interventions such as good hand
but cases in group 3 patients, who were assumed hygiene and prudent use of antimicrobials.
to represent cases of endogenous infection,
remained unchanged.38
Vianelli et al. reported successful control of an Conflict of interest statement
outbreak of P. aeruginosa on a haematology unit K.K. and A.S. have received research funding
following the installation of point-of-use filtration. from PALL Europe; K.K. has received speaker
This approach has also been investigated in non- fees from PALL Europe.
outbreak situations.39 Trautman et al. introduced
filters on an 11-bedded surgical ICU.40 P. aerugi- Funding sources
nosa infections were initially deemed to be endog- None.
enous in origin and selective decontamination of
the digestive tract (SDD) was introduced. This
failed to make an impact on the numbers of infec-
tions, and other measures, such as use of sterile References
water for oral care, face washing and cleaning of
1. Lodise Jr TP, Patel N, Kwa A, et al. Predictors of 30-day
taps, were introduced without success. Point- mortality among patients with Pseudomonas aeruginosa
of-use filters were installed with water outlets bloodstream infections: impact of delayed appropriate
sampled at two-weekly intervals beforehand. In antibiotic selection. Antimicrob Agents Chemother 2007;
the pre-filter period 113/117 (97%) water speci- 51:3510e3515.
mens grew P. aeruginosa with [109/113 (96%)] 2. Micek ST, Lloyd AE, Ritchie DJ, Reichley RM, Fraser VJ,
Kollef MH. Pseudomonas aeruginosa bloodstream infection:
yielding numbers in excesss of 102 cfu/mL. Molec- importance of appropriate initial antimicrobial treatment.
ular typing revealed that all 113 water isolates Antimicrob Agents Chemother 2005;49:1306e1311.
and 25/27 (92.6%) patient isolates belonged to 3. Kollef KE, Schramm GE, Wills AR, Reichley RM, Micek ST,
the same clonotype. After introduction of filters Kollef MH. Predictors of 30-day mortality and hospital costs
0/52 samples yielded P. aeruginosa and the num- in patients with ventilator-associated pneumonia attributed
to potentially antibiotic-resistant Gram-negative bacteria.
ber of patients colonised or infected with the Chest 2008;134:281e287.
bacterium was significantly reduced, as was the 4. Eifrig CW, Scott IU, Flynn Jr HW, Miller D. Endophthalmitis
amount of anti-pseudomonal antibiotics used.40 caused by Pseudomonas aeruginosa. Ophthalmology 2003;
110:1714e1717.
5. Hidron AI, Edwards JR, Patel J, et al. NHSN annual update:
antimicrobial-resistant pathogens associated with health-
Discussion care-associated infections: annual summary of data
reported to the National Healthcare Safety Network at
Pseudomonas aeruginosa remains a major HCAI- the centers for disease control and prevention, 2006e
related pathogen and is associated with significant 2007. Infect Control Hosp Epidemiol 2008;29:996e1011.
morbidity and mortality, particularly for immuno- 6. Engel J, Balachandran P. Role of Pseudomonas aeruginosa type
III effectors in disease. Curr Opin Microbiol 2009;12:61e66.
compromised patients and vulnerable patients on 7. Tart AH, Wozniak DJ. Shifting paradigms in Pseudomonas
ICUs. Management of infections associated with aeruginosa biofilm research. Curr Top Microbiol Immunol
this bacterium is rendered increasingly difficult 2008;322:193e206.
due to the continued emergence of antibiotic resis- 8. Smith K, Hunter IS. Efficacy of common hospital biocides
tance and, more recently, the appearance of pan- with biofilms of multi-drug resistant clinical isolates.
J Med Microbiol 2008;57:966e973.
resistant strains. Against this backdrop, the need 9. Mesaros N, Nordmann P, Plésiat P, et al. Pseudomonas
for robust strategies for prevention and control of aeruginosa: resistance and therapeutic options at the turn
these infections is self-evident. Results from studies of the new millennium. Clin Microbiol Infect 2007;13:
which have employed molecular typing techniques 560e578.
undertaken as part of outbreak investigations or 10. Souli M, Galani I, Giamarellou H. Emergence of extensively
drug-resistant and pandrug-resistant Gram-negative bacilli
conducted prospectively have highlighted the im- in Europe. Eurosurveillance 2008;13:1e11.
portance of hospital water supplies as a potential 11. Hanberger H, Arman D, Gill H, et al. Surveillance of micro-
source of P. aeruginosa. A range of control bial resistance in European intensive care units: a first
344 K.G. Kerr, A.M. Snelling

report from the CareeICU programme for improved infec- due to Pseudomonas aeruginosa in a paediatric surgical unit
tion control. Intensive Care Med 2009;35:91e100. associated with tap-water contamination. J Hosp Infect
12. Jean SS, Hsueh PR, Lee WS, et al. Nationwide surveillance 1998;39:301e307.
of antimicrobial resistance among non-fermentative 27. Bert F, Maubec E, Bruneau B, Berry P, Lambert-Zechovsky N.
Gram-negative bacteria in intensive care units in Taiwan: Multi-resistant Pseudomonas aeruginosa outbreak associ-
SMART programme data 2005. Int J Antimicrob Agents ated with contaminated tap water in a neurosurgery inten-
2009;33:266e271. sive care unit. J Hosp Infect 1998;39:53e62.
13. Walkty A, Decorby M, Nichol K, Mulvey MR, Hoban D, 28. Kolmos HJ, Thuesen B, Nielsen SV, Lohmann M,
Zhanel G. Antimicrobial susceptibility of Pseudomonas Kristoffersen K, Rosdahl VT. Outbreak of infection in a burns
aeruginosa isolates obtained from patients in Canadian unit due to Pseudomonas aeruginosa originating from con-
intensive care units as part of the Canadian National Inten- taminated tubing used for irrigation of patients. J Hosp In-
sive Care Unit study. Diagn Microbiol Infect Dis 2008;61: fect 1993;24:11e21.
217e221. 29. Eckmanns T, Oppert M, Martin M, et al. An outbreak of
14. Rosenthal VD, Maki DG, Mehta A, et al. International Noso- hospital-acquired Pseudomonas aeruginosa infection
comial Infection Control Consortium report, data summary caused by contaminated bottled water in intensive care
for 2002e2007, issued January 2008. Am J Infect Control units. Clin Microbiol Infect 2008;14:454e458.
2008;36:627e637. 30. Trautmann M, Bauer C, Schumann C, et al. Common RAPD
15. Walsh TR. Clinically significant carbapenemases: an update. pattern of Pseudomonas aeruginosa from patients and tap
Curr Opin Infect Dis 2008;21:367e371. water in a medical intensive care unit. Int J Hyg Environ
16. Beno P, Krcmery V, Demitrovicova A. Bacteraemia in cancer Health 2006;209:325e331.
patients caused by colistin-resistant Gram-negative bacilli 31. Blanc DS, Nahimana I, Petignat C, Wenger A, Bille J,
after previous exposure to ciprofloxacin and/or colistin. Francioli P. Faucets as a reservoir of endemic Pseudomonas
Clin Microbiol Infect 2006;12:497e498. aeruginosa colonization/infections in intensive care units.
17. Matz C, Moreno AM, Alhede M, et al. Pseudomonas aerugi- Intensive Care Med 2004;30:1964e1968.
nosa uses type III secretion system to kill biofilm-associated 32. Berthelot P, Grattard F, Mahul P, et al. Prospective study of
amoebae. ISME J 2008;2:843e852. nosocomial colonization and infection due to Pseudomonas
18. Ntagiopoulos PG, Paramythiotou E, Antoniadou A, aeruginosa in mechanically ventilated patients. Intensive
Giamarellou H, Karabinis A. Impact of an antibiotic restric- Care Med 2001;27:503e512.
tion policy on the antibiotic resistance patterns of Gram- 33. Reuter S, Sigge A, Wiedeck H, Trautmann M. Analysis of
negative microorganisms in an intensive care unit in transmission pathways of Pseudomonas aeruginosa between
Greece. Int J Antimicrob Agents 2007;30:360e365. patients and tap water outlets. Crit Care Med 2002;30:
19. Cook PP, Das TD, Gooch M, Catrou PG. Effect of a program 2222e2228.
to reduce hospital ciprofloxacin use on nosocomial Pseudo- 34. Vallés J, Mariscal D, Cortés P, et al. Patterns of colonization
monas aeruginosa susceptibility to quinolones and other an- by Pseudomonas aeruginosa in intubated patients: a 3-year
timicrobial agents. Infect Control Hosp Epidemiol 2008;29: prospective study of 1,607 isolates using pulsed-field gel
716e722. electrophoresis with implications for prevention of ventila-
20. Jonas D, Meyer E, Schwab F, Grundmann H. Genodiversity of tor-associated pneumonia. Intensive Care Med 2004;30:
resistant Pseudomonas aeruginosa isolates in relation to an- 1768e1775.
timicrobial usage density and resistance rates in intensive 35. Cholley P, Thouverez M, Floret N, Bertrand X, Talon D. The
care units. Infect Control Hosp Epidemiol 2008;29: role of water fittings in intensive care rooms as reservoirs
350e357. for the colonization of patients with Pseudomonas aerugi-
21. Moolenaar RL, Crutcher JM, San Joaquin VH, et al. A pro- nosa. Intensive Care Med 2008;34:1428e1433.
longed outbreak of Pseudomonas aeruginosa in a neonatal 36. Rogues AM, Boulestreau H, Lashéras A, et al. Contribution
intensive care unit: did staff fingernails play a role in of tap water to patient colonisation with Pseudomonas aer-
disease transmission? Infect Control Hosp Epidemiol 2000; uginosa in a medical intensive care unit. J Hosp Infect 2007;
21:80e85. 67:72e78.
22. McNeil SA, Nordstrom-Lerner L, Malani PN, Zervos M, 37. Bukholm G, Tannaes T, Kjelsberg AB, Smith-Erichsen N. An
Kauffman CA. Outbreak of sternal surgical site infections outbreak of multidrug-resistant Pseudomonas aeruginosa
due to Pseudomonas aeruginosa traced to a scrub nurse associated with increased risk of patient death in an inten-
with onychomycosis. Clin Infect Dis 2001;33:317e323. sive care unit. Infect Control Hosp Epidemiol 2002;23:
23. Trautmann M, Lepper PM, Haller M. Ecology of Pseudomonas 441e446.
aeruginosa in the intensive care unit and the evolving role 38. Petignat C, Francioli P, Nahimana I, et al. Exogenous
of water outlets as a reservoir of the organism. Am J Infect sources of Pseudomonas aeruginosa in intensive care unit
Control 2005;33:S41eS49. patients: implementation of infection control measures
24. Aumeran C, Paillard C, Robin F, et al. Pseudomonas aerugi- and follow-up with molecular typing. Infect Control Hosp
nosa and Pseudomonas putida outbreak associated with Epidemiol 2006;27:953e957.
contaminated water outlets in an oncohaematology paedi- 39. Vianelli N, Giannini MB, Quarti C, et al. Resolution of a Pseu-
atric unit. J Hosp Infect 2007;65:47e53. domonas aeruginosa outbreak in a hematology unit with the
25. Hota S, Hirji Z, Stockton K, et al. Outbreak of multidrug- use of disposable sterile water filters. Haematologica 2006;
resistant Pseudomonas aeruginosa colonization and 91:983e985.
infection secondary to imperfect intensive care unit room 40. Trautmann M, Halder S, Hoegel J, Royer H, Haller M. Point-
design. Infect Control Hosp Epidemiol 2009;30:25e33. of-use water filtration reduces endemic Pseudomonas aeru-
26. Ferroni A, Nguyen L, Pron B, Quesne G, Brusset MC, ginosa infections on a surgical intensive care unit. Am J Infect
Berche P. Outbreak of nosocomial urinary tract infections Control 2008;36:421e429.
Journal of Hospital Infection (2009) 73, 345e354
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Extended-spectrum b-lactamase-producing
organisms
M.E. Falagas a,b,c,*, D.E. Karageorgopoulos a
a
Alfa Institute of Biomedical Sciences, Athens, Greece
b
Department of Medicine, Henry Dunant Hospital, Athens, Greece
c
Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA

Available online 10 July 2009

KEYWORDS Summary Extended-spectrum b-lactamases (ESBLs), which hydrolyse


Bacterial drug extended-spectrum cephalosporins and are inhibited by clavulanic acid,
resistance; are spreading among Enterobacteriaceae. The CTX-M enzymes are repla-
b-Lactam resistance;
cing SHV and TEM enzymes as the prevalent type of ESBLs, principally in
Cefotaximases;
Genetic techniques;
community-acquired infections caused by Escherichia coli. Associated in-
Microbiological fectious syndromes include mainly urinary tract infections, and secondly
techniques; bloodstream and intra-abdominal infections, and may be serious enough
Plasmids to warrant hospitalisation. Affected patients commonly have various un-
derlying risk factors. This is also observed in hospital-acquired infections.
The rates of ESBL-expression among nosocomial Enterobacteriaceae
isolates, particularly Klebsiella pneumoniae, have risen substantially in
several countries. The hospital epidemiology of these infections is often
complex; multiple clonal strains causing focal outbreaks may co-exist with
sporadic ones. Relevant infection-control measures should focus on reducing
patient-to-patient transmission via the inanimate environment, hospital
personnel, and medical equipment. Wise use of antibiotics is also essen-
tial. The available therapeutic options for the treatment of ESBL-
associated infections are limited by drug resistance conferred by the
ESBLs, along with frequently observed co-resistance to various antibiotic
classes, including cephamycins, fluoroquinolones, aminoglycosides, tetra-
cyclines, and trimethoprim/sulfamethoxazole. Relevant clinical data
regarding the effectiveness of different regimens for ESBL-associated in-
fections are limited. Although certain cephalosporins may appear active
in vitro, associated clinical outcomes are often suboptimal. b-Lactam/
b-lactamase inhibitor combinations may be of value, but the supporting
evidence is weak. Carbapenems are regarded as the agents of choice,

* Corresponding author. Address: Alfa Institute of Biomedical Sciences, 9 Neapoleos Street, 151 23 Marousi, Athens, Greece.
Tel.: þ30 210 683 9604; fax: þ30 210 683 9605.
E-mail address: m.falagas@aibs.gr

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.02.021
346 M.E. Falagas, D.E. Karageorgopoulos

and may be more effective than fluoroquinolones for serious infections. Ti-
gecycline and polymyxins have substantial antimicrobial activity against
ESBL-producing Enterobacteriaceae, and, along with fosfomycin, merit
further evaluation.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

Introduction on the Tigecycline Evaluation and Surveillance


Trial (TEST) global surveillance database, the
The main mechanism of bacterial resistance to the rate of ESBL production was highest among the
b-lactam class of antibiotics consists of the pro- K. pneumoniae isolates collected in Latin America,
duction of b-lactamases, which are hydrolytic followed by Asia/Pacific Rim, Europe, and North
enzymes with the ability to inactivate these America (44.0%, 22.4%, 13.3% and 7.5%, respec-
antibiotics before they reach the penicillin-binding tively).7 The same ranking order between the
proteins located at the cytoplasmic membrane. different geographical regions was observed
The extended-spectrum b-lactamases (ESBLs) are regarding the prevalence of ESBLs among the E.
classified in the molecular (Ambler) class A and coli isolates, although the corresponding rates
functional (BusheJacobyeMedeiros) group 2be; were lower (13.5%, 12.0%, 7.6%, and 2.2%, respec-
they are characterised by the ability to hydrolyse tively).7 It should be mentioned that the above
an oxyimino-b-lactam at a rate 10% of that for data refer to isolates related to hospital-acquired
benzylpenicillin along with inhibition by clavulanic infections obtained from various clinical
acid.1,2 The presence of ESBLs in various members specimens.
of the Enterobacteriaceae family, particularly Detailed data derived from the TEST database
Klebsiella pneumoniae and Escherichia coli, is of regarding the prevalence of ESBL production
great microbiological and clinical importance. among Enterobacteriaceae isolates in Europe
ESBLs are also found in non-fermentative Gram- have recently been presented.8 According to these
negative bacteria, such as Pseudomonas aerugi- data that refer to 22 European countries for the
nosa and Acinetobacter baumannii.3 period of 2004 to 2007, the rate of ESBL production
The ESBL enzymes were initially recognised in among 515 K. pneumoniae isolates and 794 E. coli
clinical isolates in the 1980s; they derived mainly isolates was 15.5% and 9.8%, respectively. Marked
from the TEM or SHV types of b-lactamases, by point differences were observed in the country-specific
mutations in the parent enzymes which did not data; the highest rate of ESBL production was
possess extended-spectrum b-lactam substrate noted in Greece, while the lowest was noted in
activity.3 The CTX-M type of ESBLs is becoming in- Denmark. Relevant data collected by the European
creasingly more prevalent, particularly in E. coli Antimicrobial Resistance Surveillance System
and K. pneumoniae.4,5 More than 50 enzymes of (EARSS Annual Report 2007. Bilthoven. The Nether-
the latter type have so far been identified, which lands. ISBN:978-90-6960-214-1. available at:
can be divided into five main groups on the basis http://www.rivm.nl/earss/) regarding resistance
of amino acid changes (CTX-M1, CTX-M2, CTX-M8, rates to third-generation cephalosporins of
CTX-M9 and CTX-M25, respectively).6 The origin of K. pneumoniae and E. coli clinical isolates col-
some of these enzymes has been traced to chromo- lected in 31 European countries are consistent
somally encoded enzymes of the Kluyvera spp. of with those of the TEST database.
environmental bacteria. The relevant genes are Particular attention should be paid to the in-
thought to have been mobilised into conjugative creasing prevalence of the CTX-M type ESBLs
plasmids and thus transferred to pathogenic bac- worldwide.6 This can be attributed to the spread
teria.6 Additional clinically relevant types of ESBLs of CTX-M genes among bacterial species by plas-
include mainly the VEB, PER, GES, TLA, IBC, mids or other mobile genetic elements, as well as
SFO-1, BES-1 and BEL-1 types.3 to the clonal expansion of epidemic strains carry-
ing these genes.5 The prevalence of specific types
or groups of CTX-M ESBLs has acquired endemic
Global epidemiology proportions in many countries. Among European
countries, relevant examples include CTX-M-1 en-
The rate of ESBL production among Enterobacter- zymes in Italy, CTX-M-9 and CTX-M-14 enzymes in
iaceae varies worldwide. In a recent study based Spain, CTX-M-3 enzymes in Poland, and CTX-M-15
ESBL-producing organisms 347

enzymes in the UK.5,6,9 Notably, the CTX-M-15 type of ESBL present in a micro-organism, which
ESBLs exhibit a nearly worldwide distribution.5 may be particularly useful for epidemiological
Nevertheless CTX-M enzymes have rarely been purposes.9 Moreover, they can detect low-level
found responsible for ESBL production among clin- resistance, and can be performed without prior
ical isolates collected in the USA. However, a re- culture of the microbiological specimen.
cent study highlights the increasing prevalence of
CTX-M type ESBLs in a large US institution.10
Clinical relevance and impact of ESBL-
associated infections
Laboratory detection
It is increasingly being recognised that the pro-
In the microbiological laboratory, detection of duction of ESBLs is not relevant to nosocomial
ESBLs can be done with phenotypic or genotypic infections only, but is becoming an important public
tests. The phenotypic tests are routinely used in health issue also with regard to infections acquired
clinical diagnostic laboratories, whereas the geno- in the community. Community-onset ESBL-associ-
typic tests are mainly used in reference or re- ated infections are principally caused by E. coli pro-
search laboratories. ducing CTX-M type ESBLs.9 Urinary tract infections
The phenotypic tests for ESBL detection involve constitute the main clinical syndrome observed in
screening and confirmatory steps. The screening this setting. Bloodstream infections may also be
step consists of testing for resistance to cefpodox- observed, mainly of urinary or biliary tract origin.
ime (which is hydrolysed by all TEM, SHV, and CTX-M Community-acquired ESBL-associated infections
types of ESBLs), cefotaxime, ceftazidime, ceftriax- typically affect patients with various complicating
one, or aztreonam.11 The confirmatory step is based factors. A relevant caseecontrol study identified
on the demonstration of synergy between the above various risk factors for community-acquired
agents and clavulanic acid.11 Several methods in- infection by ESBL-producing E. coli, including
cluding the double disc synergy test, the combina- increased age, female sex, diabetes mellitus, re-
tion disc method, or specific ESBL Etests can be current urinary tract infection, previous instru-
used in this regard.11,12 Poor sensitivity of these mentation of the urinary tract, follow-up in
tests may be observed when the evaluated ESBL- outpatient clinic, and previous receipt of amino-
producing isolate additionally produces a b-lacta- penicillins, cephalosporins, or fluoroquinolones.13
mase not inhibited by clavulanic acid, such as an Such findings raise the question whether commu-
AmpC b-lactamase or metallo-b-lactamase.12 nity-onset ESBL-associated infections are mainly
Methods to overcome this limitation include the healthcare-associated. However, reports of truly
use of cefepime, which is a weak substrate for community-acquired infections are increasing,
most AmpC b-lactamases, the use of chromogenic while clusters of cases in the community, particu-
agar, cloxacillin-containing agar, or the addition of larly among members of the same family, may be
EDTA to inactivate metallo-b-lactamases.11 observed.14 Additionally, faecal carriage of ESBLs
The above-mentioned principles for the detec- has been reported in a considerable percentage
tion of ESBLs by phenotypic methods have been of healthy individuals residing in the community.15e17
incorporated in most commercial semi-automated Potential transmission of ESBL-producing organ-
bacterial identification and antimicrobial suscep- isms from animal sources to humans through the
tibility testing systems. However, their perfor- food chain or patient-to-patient transmission of
mance in this regard is variable and their these organisms might contribute to the dissemi-
accuracy appears to be lower compared with the nation of ESBLs in the community, but these issues
conventional phenotypic methods.12 require further study.16
The genotypic tests for the detection of ESBLs ESBL-associated infections observed in hospitalised
primarily consist of polymerase chain reaction- patients represent either serious community-
based amplification of the specific genes. Regard- acquired infections requiring hospital admission or
ing the TEM and SHV type ESBLs, additional infections acquired during hospitalisation. The
molecular techniques, such as sequencing or degree that community-acquired infections con-
restriction fragment length polymorphism, are re- tribute to the isolation of ESBL-producing organisms
quired for the identification of specific point in hospitalised patients appears to be increasing.18
mutations that differentiate these enzymes from Furthermore, infection or colonisation of residents
parent enzymes without ESBL activity.9 Although in long-term care facilities by ESBL-producing
technically challenging, the genotypic methods organisms may provide a means for the dissemina-
have the advantage of identification of the specific tion of ESBLs between the community and hospitals.
348 M.E. Falagas, D.E. Karageorgopoulos

In this respect, the incidence of colonisation by modes of patient-to-patient transmission of ESBL-


ESBL-producing organisms of residents in long- producing organisms in the hospital setting, which
term care facilities appears to be substantially in- include transmission via colonisation of the
creasing, although relevant data are limited.19 inanimate environment, the hands of healthcare
Regarding hospital-acquired infections caused by personnel, and of medical equipment.14
ESBL-producing organisms, the majority of relevant The identification of patients colonised with ESBL-
studies refer to K. pneumoniae.9,14 Clinical syn- producing organisms can be done with surveillance
dromes observed in this setting include respiratory cultures of gastrointestinal tract specimens, partic-
tract and wound infections, in addition to urinary ularly with rectal swabs.14 It has been shown that
tract, bloodstream, and intra-abdominal ones.9 a substantial percentage of patients who develop
Risk factors for infection or colonisation of hospital- nosocomial ESBL-associated infections have preced-
ised patients by ESBL-producing organisms are simi- ing colonisation of the gastrointestinal tract. How-
lar to those referring to other common nosocomial ever, the identification of ESBL producers among
micro-organisms.20 Specifically, increasing length of commensal Enterobacteriaceae is technically de-
hospital or intensive care unit (ICU) stay, greater se- manding and requires the use of selective culture
verity of clinical status, insertion of various types of media. Whether a strategy of selective decontamina-
indwelling catheters, performance of certain types tion of the gastrointestinal tract of patients found to
of invasive procedures or surgical interventions, re- be colonised with ESBL-producing organisms is effec-
ceipt of renal replacement therapy or mechanical tive in terms of infection control remains a controver-
ventilatory support have all been associated with sial issue.14 Although effective decolonisation may
the isolation of ESBL-producing organisms from hos- reduce the likelihood of subsequent infection by
pitalised patients.3,21 The use of antibiotics, particu- these organisms, as well as horizontal spread to
larly of oxyimino-b-lactams or fluoroquinolones, neighbouring patients, increased resistance rates of
constitutes an important additional risk factor.3 nosocomial ESBL-producing Enterobacteriaceae iso-
Studies assessing the epidemiology of hospital- lates to agents commonly used in this regard, such
acquired ESBL-associated infections have often as neomycin and norfloxacin, limit the utility of this
recognised a complex pattern.18 Specifically, epi- approach. Moreover, the use of microbiologically ac-
demic strains may co-exist with sporadic ones,20 tive agents, such as polymyxins, may carry the risk of
while multiple predominant clones may also be ob- selection for Gram-negative organisms resistant to
served.18 Furthermore, the same types of ESBLs this class of agents, which is often used as a last resort
may be identified in clonally unrelated isolates, option for infections by highly resistant isolates.24
or, conversely, isolates with the same clonal origin It should be mentioned that the selection of proper
may encode different types of ESBLs.14 These ob- antibiotic therapy is a key factor relating to the
servations are attributed to horizontal spread of effectiveness of infection control. Specifically, limit-
ESBL genes through mobile genetic elements.15 ing the institutional use of third generation cephalo-
The clinical impact of ESBL-associated infections sporins has been shown to aid towards the reduction
has mainly been studied in hospitalised patients, of the prevalence of ESBL-producing organisms.25 Use
especially those with bloodstream infections. In this of fluoroquinolones may contribute to the selection of
respect, it has been found that bloodstream ESBL producers, because determinants of fluoro-
infections by ESBL-producing Enterobacteriaceae quinolone resistance are often carried in the same
isolates compared with non-producing isolates is mobile genetic elements as ESBL genes.15 Some stud-
associated with a delay in the institution of appro- ies have suggested that substitution of cephalosporins
priate antimicrobial therapy, as empirically insti- for piperacillin/tazobactam may be useful in limiting
tuted antibiotics may be inactive.22 The above the nosocomial isolation rates of ESBL-producing
factor is thought to be mainly responsible for the organisms.25,26 However, consideration should be given
increased mortality related to ESBL production in in preventing the emergence of Gram-negative organ-
K. pneumoniae or E. coli bloodstream infections.22 isms with advanced antimicrobial drug resistance.27

Hospital infection control Treatment

The general aspects of hospital infection control for Associated antimicrobial drug resistance
ESBL-producing Enterobacteriaceae are similar to patterns
those applied for other common nosocomial Gram-
negative organisms.14,23 Specifically, infection con- ESBLs hydrolyse penicillins, cephalosporins (with
trol measures should focus on preventing the main the exception of cephamycins), and aztreonam.14
ESBL-producing organisms 349

However, the degree of hydrolytic activity against cilastatin regarding the subgroup of patients
the above substrates may considerably differ for infected with ESBL-producing organisms.30
different types of ESBLs. Typically, the TEM and Additional studies, although small, have reported
SHV type ESBLs have greater hydrolytic activity suboptimal effectiveness of cephalosporins for
for ceftazidime than cefotaxime, in contrast to the treatment of ESBL-producing Enterobacteria-
the CTX-M type ESBLs.6,14 Consequently, ESBL-pro- ceae, even if in-vitro antimicrobial activity is
ducing organisms may appear susceptible to some shown.31,32 Thus, most experts argue against
of the above agents in vitro. The Clinical and Lab- cephalosporin use as treatment of choice for ESBL-
oratory Standards Institute (CLSI) recommends associated infections.
that ESBL-producing E. coli, K. pneumoniae, Kleb-
siella oxytoca and P. mirabilis should be reported
b-Lactams/b-lactamase inhibitor
as resistant to penicillins, true cephalosporins
combinations
and aztreonam, regardless of the in-vitro suscepti-
bility data. The level of in-vitro antimicrobial drug
The degree of inhibitory activity of b-lactamase
resistance conferred by the presence of ESBLs to
inhibitors against the hydrolysis of b-lactams by
b-lactam/b-lactamase inhibitor combinations is
the ESBL enzymes may vary by the type of inhibitor
variable.14 Relevant minimum inhibitory concen-
as well as by the type of ESBL. In this respect,
trations can be in the susceptible range. An inocu-
tazobactam has been found to be more potent
lum effect regarding the in-vitro susceptibility of
compared with clavulanic acid against certain
ESBL-producing isolates to agents that are hydro-
CTX-M type ESBLs,33 while both of the above
lysed by the ESBLs, including b-lactam/b-lacta-
agents appear to be more potent than sulbactam
mase inhibitor combinations, has also been
in inhibiting TEM and SHV type ESBLs.34
found. However, clinical relevance of this phenom-
The available clinical evidence regarding the
enon has not been firmly established, as it may
utility of b-lactam/b-lactamase inhibitor combina-
represent simply a laboratory artefact.21
tions for the treatment of ESBL-associated infec-
An important factor that limits the array of
tions is rather limited. Specifically, favourable
active antibiotics against ESBL-producing Entero-
patient outcomes have been related to piperacil-
bacteriaceae is the frequent co-expression of
lin/tazobactam treatment in some small studies,
resistance by these organisms to classes of antimi-
although such findings have not been consistently
crobial agents other than those hydrolysed by the
reproduced.26,35 One pharmacokinetic/pharmaco-
ESBLs. This has been shown for fluoroquinolones,
dynamic modelling study concluded that the prob-
aminoglycosides, tetracyclines (excluding glycylcy-
ability of pharmacodynamic target attainment
clines), and trimethoprim/sulfamethoxazole.9
against infections caused by ESBL-producing E. coli
Cephalosporins and K. pneumoniae is lower for piperacillin/tazo-
bactam than cefepime if conventional dosing regi-
Relatively few studies have assessed clinically mens are used.36 Moreover, increasing resistance
rates of ESBL Enterobacteriaceae to piperacillin/ta-
cephalosporin treatment for infections caused by
zobactam may limit the potential therapeutic util-
ESBL-producing organisms, with an agent of this
ity of this agent.26 Last but not least, it should be
class showing activity in vitro against the causative
mentioned that amoxicillin/clavulanate may have
organism. In a small relevant clinical trial, clinical
considerable antimicrobial activity against
success rates were similar in seven patients with
Enterobacteriaceae organisms isolated in the com-
CTX-M-producing E. coli bacteraemia treated with
munity, and it may constitute an effective thera-
ceftazidime compared with eight such patients
treated with imipenem/cilastatin (86% compared peutic option for community-acquired urinary tract
infections caused by ESBL-producing isolates.13,37
with 88%, respectively).28 Likewise, a retrospective
study did not find significant differences in the
clinical outcome of 44 ICU hospitalised patients Cephamycins
with TEM-24-producing Enterobacter aerogenes
infections, treated with cefepime-based versus car- The cephamycins, mainly including cefoxitin, ce-
bapenem-based therapy.29 However, the microbio- fotetan, and cefmetazole, do not by definition
logical outcome was inferior in the cefepime constitute substrates for hydrolysis by the ESBL
group. Moreover, in the context of a randomised enzymes. However, co-resistance to these agents
trial evaluating patients with nosocomial pneumo- in ESBL-producing Enterobacteriaceae may be
nia, cefepime treatment tended to be associated observed, mainly due to porin loss or concomitant
with worse outcome compared with imipenem/ expression of AmpC b-lactamases. Clinical data
350 M.E. Falagas, D.E. Karageorgopoulos

regarding the potential value of cephamycins for the use of carbapenems over fluoroquinolones,
the treatment of ESBL-associated infections are whereas the other found similar effectiveness
scarce. Specifically, a small retrospective study with both these antibiotic classes.41,44
evaluated treatment with flomoxef, which is
grouped along with latamoxef in the related
Tigecycline
oxacephem class of b-lactams, or a carbapenem
for a total of 27 patients with K. pneumoniae
Tigecycline, a derivative of minocycline, is the first
bacteraemia. Difference in mortality between
member of the glycylcycline class of antibiotics
the two treatment groups was not evident.38
available for clinical use. It has the property to
However, additional reports have noted that emer-
evade common mechanisms of resistance to
gence of resistance to cephamycins may be
tetracyclines expressed in Gram-negative and
observed during therapy with agents, and even
Gram-positive bacteria.45 Detailed data on the
co-resistance to carbapenems also.14,39
antimicrobial activity of tigecycline against ESBL-
producing Enterobacteriaceae, as reported in a re-
Carbapenems cent systematic review of the literature, are
summarised in Table II.46 Specifically, excellent
Carbapenems are considered to be the treatment activity of tigecycline has been shown against
of choice against serious ESBL-associated infec- ESBL-producing E. coli isolates. Additionally, sub-
tions.9 This is mainly because they are not inacti- stantial antimicrobial activity of tigecycline has
vated by these enzymes in vitro, and have been demonstrated against ESBL-producing K.
demonstrated adequate effectiveness for the pneumoniae isolates, although this depends on
treatment of serious Gram-negative infections at the interpretive breakpoints of susceptibility
various body sites. However, specific data for their elected. Data regarding the antimicrobial activity
clinical use against ESBL-associated infections are of tigecycline against other ESBL-producing En-
rather limited, although generally supportive of terobacteriaceae organisms are rather limited.
their effectiveness.30,32,35,40 A multicentre pro- Clinical data regarding the effectiveness of
spective cohort study that evaluated 85 cases of tigecycline for the treatment of infections caused
K. pneumoniae bacteraemia demonstrated that by ESBL-producing organisms are yet limited.46
use of a carbapenem in the initial five-day period Further, consideration of the pharmacokinetic
of the infection was a factor independently associ- and pharmacodynamic parameters of this agent
ated with lower mortality.41 In addition, in a small casts doubt on the potential effectiveness of tige-
clinical trial, ertapenem use for the treatment of cycline for the treatment of specific infectious
20 patients with early-onset ventilator-associated syndromes, such as urinary tract and bloodstream
pneumoniae caused by ESBL-producing Enterobac- infections.45 In this respect, only a fraction of
teriaceae resulted in a rather favourable overall 10e15% of the tigecycline dose appears to be ex-
clinical success rate of 80%.42 creted as active, unchanged drug in the urine.
Achievable serum concentrations of this agent
may also be inadequate (due to extensive tissue
Fluoroquinolones
drug distribution) for substantial antimicrobial
activity to be exerted against pathogenic micro-
As mentioned above, ESBL-producing organisms
organisms circulating in the bloodstream with
may carry resistance determinants that confer
minimum inhibitory concentrations close to the
low- or high-level resistance to fluoroquinolones.
susceptibility breakpoint.
Potential resistance to fluoroquinolones may relate
to suboptimal patient outcomes when these agents
are elected as empirical therapy for infections Considerations for further research
caused by ESBL-producing organisms. There are
some concerns also regarding the effectiveness of The re-evaluation of earlier-used antimicrobial
fluoroquinolones for the treatment of serious ESBL- agents, that have had low clinical use in recent
associated infections caused by fluoroquinolone- decades, for potential antimicrobial activity and
susceptible isolates, compared with carbapenems. clinical effectiveness against today’s resistant
Studies that have provided relevant data e albeit micro-organisms may provide a temporary solution
only for a small number of patients e are summar- to the problem of spreading and advancing bac-
ised in Table I.35,40,41,43,44 Regarding the findings of terial drug resistance. Agents that may be useful
the two largest relevant studies, which both address for the treatment of ESBL-associated infections
K. pneumoniae bacteraemia, one study favoured include polymyxins, fosfomycin, nitrofurantoin,
Table I Clinical outcome in patients with infections caused by ESBL-producing Enterobacteriaceae treated with fluoroquinolones, carbapenems, or other agents
showing in-vitro activity
ESBL-producing organisms

Study Study design Patient characteristics Outcome Specific agents: n/N (%)
Fluoroquinolones Carbapenems Other antibiotics
a
Kim et al. Retrospective Adult patients with Mortality Ciprofloxacin : 1/3 Imipenem: 2/12 (17) Aminoglycosidesa: 2/4 (50)
(2002)43 cohort study ESBL K. pneumoniae (33)
bacteraemia
Burgess et al. Retrospective Cases with infections by Clinical Fluoroquinolonesa: Carbapenemsa: 0/3 (0) Piperacillin/tazobactama: 1/3
(2003)35 cohort study ESBL-producing failure 0/3 (0) (33)
organisms (E. coli, K.
pneumoniae, K. oxytoca)
Endimiani et al. Retrospective Patients with TEM-52 Clinical Ciprofloxacina,b: Carbapenemsa,b: 3/11 Aminoglycosidesa,b: 0/2 (0)
(2004)40 cohort study ESBL-producing K. failure 2/7 (29) (27) [imipenem: 2/10
pneumoniae bacteraemia (20); meropenem: 1/1
(100)]
Kang et al. Retrospective Patients with ESBL- 30-day Ciprofloxacinc: Carbapenemsc: 8/62 NR
(2004)44 cohort study producing E. coli or K. mortality 3/29 (10) (12.9)
pneumoniae bacteraemia
Paterson et al. Prospective Patients aged >6 years Mortality Ciprofloxacina: Carbapenemsa: 1/27 (4) Cephalosporinsa: 2/5 (50); b-
(2004)41 multicentre with ESBL-producing K. 4/11 (36) [imipenem: 1/24 (4); lactam/b-lactamase inhibitora:
cohort study pneumoniae meropenem: 0/3 (0)] 2/4 (50)
ESBL, extended-spectrum b-lactamase; NR, not specifically reported.
a
Representing therapy as the only microbiologically active agents.
b
Patients who received adequate treatment and dose for 7 days.
c
Definitive therapy.
351
352 M.E. Falagas, D.E. Karageorgopoulos

Table II Cumulative susceptibility to tigecycline of ESBL-producing Enterobacteriaceae isolates identified in


different studiesa
Micro-organisms No. of studies Susceptibility, % (no. of isolates)
FDA criteriab EUCAST criteriab
E. coli 16 99.8% (1636) 99.7% (737)
Klebsiella spp. 17 92.3% (2030) 72.3% (1284)
Enterobacter spp. 4 91.3% (69) 77.6% (49)
a
Adapted from Kelesidis et al.46
b
Food and Drug Administration (FDA) and European Committee on Antimicrobial Susceptibility Testing (EUCAST) breakpoints of
susceptibility: minimum inhibitory concentrations 2 and 1 mg/L, respectively.

and temocillin. Furthermore, combining available a pragmatic new definition for ESBLs has been
cephalosporins with b-lactamase inhibitors could emphasised.51 In fact, re-definition of the ESBLs to
enhance the effectiveness of the former agents include enzymes of different classes of b-lacta-
against ESBL-associated infections. mases that confer resistance to extended-spectrum
With regard to polymyxins (of which colistin and cephalosporins has recently been proposed.52
polymyxin B are currently available for clinical
use), these have retained excellent antimicrobial Conclusion
activity against ESBL-producing organisms.47 How-
ever, reported clinical use of these agents for the The increasing prevalence and shifting epidemiol-
treatment of such infections is scarce, since they ogy of ESBL-producing organisms, particularly of K.
are typically reserved for the treatment of infec- pneumoniae and E. coli, render the infections
tions caused by Gram-negative bacteria with caused by these pathogenic micro-organisms an
more advanced resistance patterns.23 important public health problem. The resistance
Fosfomycin can also have good antimicrobial to extended-spectrum b-lactams, which by defini-
activity against ESBL-producing Enterobacteri- tion these enzymes confer, along with frequently
aceae.48 Despite concerns that increasing use of observed co-resistance to other antibiotics renders
this agent may lead to resistance development, re- ineffective many of the regimens traditionally used
sistance rates among urinary tract isolates have re- for the empirical therapy of various types of associ-
mained low.49 Moreover, a recent study revealed ated infections. This may be of particular impor-
high effectiveness of fosfomycin in the treatment tance for community-acquired infections, since
of community-acquired lower urinary tract infec- options for oral antibiotic therapy against ESBL-pro-
tion caused by ESBL-producing E. coli.13 The po- ducing organisms appear to be limited. Regarding
tential value of this drug for the treatment of nosocomial infections caused by these organisms,
systemic infections e of origin other than the carbapenems appear as the most reliable therapeu-
urinary tract e is of marked interest.49 With regard tic agents. Further research is required on appropri-
to nitrofurantoin, this agent may also be effective ate strategies to limit the emergence and spread of
in the treatment of ESBL-associated uncompli- resistant organisms, both in the community and the
cated urinary tract infections, but this could be hospital settings, as well as to evaluate the avail-
limited by co-resistance to this agent of ESBL- able therapeutic agents and identify new ones.
producing organisms.50

b-Lactamases viewed from a clinical Conflict of interest statement


None declared.
perspective
Funding sources
Clinicians involved in the treatment of infectious None.
diseases may be puzzled by the continuous expan-
sion of knowledge concerning b-lactamases, since
new enzymes with potentially varying properties
are increasingly being recognised. From a clinical
References
standpoint, translating and grouping the complex
1. Bush K, Jacoby GA, Medeiros AA. A functional classification
microbiological information into therapeutically scheme for beta-lactamases and its correlation with
meaningful categories might facilitate the choice molecular structure. Antimicrob Agents Chemother 1995;
of appropriate therapy. In this regard, the need for 39:1211e1233.
ESBL-producing organisms 353

2. Ambler RP, Meadway RJ. Chemical structure of bacterial 19. Nicolas-Chanoine MH, Jarlier V. Extended-spectrum beta-
penicillinases. Nature 1969;222:24e26. lactamases in long-term-care facilities. Clin Microbiol
3. Jacoby GA, Munoz-Price LS. The new beta-lactamases. N Infect 2008;14(Suppl. 1):111e116.
Engl J Med 2005;352:380e391. 20. Rodriguez-Bano J, Navarro MD, Romero L, et al. Clinical and
4. Jones CH, Tuckman M, Keeney D, Ruzin A, Bradford PA. molecular epidemiology of extended-spectrum beta-lacta-
Characterization and sequence analysis of extended spec- mase-producing Escherichia coli as a cause of nosocomial
trum b-lactamase encoding genes from Escherichia coli, infection or colonization: implications for control. Clin
Klebsiella pneumoniae and Proteus mirabilis isolates col- Infect Dis 2006;42:37e45.
lected during tigecycline phase 3 clinical trials. Antimicrob 21. Pfaller MA, Segreti J. Overview of the epidemiological pro-
Agents Chemother 2009;53:465e475. file and laboratory detection of extended-spectrum beta-
5. Livermore DM, Canton R, Gniadkowski M, et al. CTX-M: lactamases. Clin Infect Dis 2006;42(Suppl. 4):S153eS163.
changing the face of ESBLs in Europe. J Antimicrob Chemo- 22. Schwaber MJ, Carmeli Y. Mortality and delay in effective
ther 2007;59:165e174. therapy associated with extended-spectrum beta-
6. Bonnet R. Growing group of extended-spectrum beta-lacta- lactamase production in Enterobacteriaceae bacteraemia:
mases: the CTX-M enzymes. Antimicrob Agents Chemother a systematic review and meta-analysis. J Antimicrob Che-
2004;48:1e14. mother 2007;60:913e920.
7. Reinert RR, Low DE, Rossi F, Zhang X, Wattal C, Dowzicky MJ. 23. Karageorgopoulos DE, Falagas ME. Current control and
Antimicrobial susceptibility among organisms from the treatment of multidrug-resistant Acinetobacter baumannii
Asia/Pacific Rim, Europe and Latin and North America infections. Lancet Infect Dis 2008;8:751e762.
collected as part of TEST and the in vitro activity of tigecy- 24. Matthaiou DK, Michalopoulos A, Rafailidis PI, et al. Risk fac-
cline. J Antimicrob Chemother 2007;60:1018e1029. tors associated with the isolation of colistin-resistant gram-
8. Hackel M, Badal R, Bouchillon S, et al. Extended-spectrum negative bacteria: a matched caseecontrol study. Crit Care
beta-lactamase production in Europe. In: Abstracts of the Med 2008;36:807e811.
18th European Congress of Clinical Microbiology and Infec- 25. Kim JY, Sohn JW, Park DW, Yoon YK, Kim YM, Kim MJ. Con-
tious Diseases (ECCMID), Barcelona, Spain, 2008. trol of extended-spectrum beta-lactamase-producing Kleb-
9. Pitout JD, Laupland KB. Extended-spectrum beta-lacta- siella pneumoniae using a computer-assisted management
mase-producing Enterobacteriaceae: an emerging public- program to restrict third-generation cephalosporin use. J
health concern. Lancet Infect Dis 2008;8:159e166. Antimicrob Chemother 2008;62:416e421.
10. Lewis 2nd JS, Herrera M, Wickes B, Patterson JE, 26. Peterson LR. Antibiotic policy and prescribing strategies for
Jorgensen JH. First report of the emergence of CTX-M-type therapy of extended-spectrum beta-lactamase-producing
extended-spectrum beta-lactamases (ESBLs) as the Enterobacteriaceae: the role of piperacillin-tazobactam.
predominant ESBL isolated in a U.S. health care system. Clin Microbiol Infect 2008;14(Suppl. 1):181e184.
Antimicrob Agents Chemother 2007;51:4015e4021. 27. Falagas ME, Rafailidis PI, Kofteridis D, et al. Risk factors of
11. Drieux L, Brossier F, Sougakoff W, Jarlier V. Phenotypic carbapenem-resistant Klebsiella pneumoniae infections:
detection of extended-spectrum beta-lactamase produc- a matched case control study. J Antimicrob Chemother
tion in Enterobacteriaceae: review and bench guide. Clin 2007;60:1124e1130.
Microbiol Infect 2008;14(Suppl. 1):90e103. 28. Bin C, Hui W, Renyuan Z, et al. Outcome of cephalosporin
12. Wiegand I, Geiss HK, Mack D, Sturenburg E, Seifert H. treatment of bacteremia due to CTX-M-type extended-
Detection of extended-spectrum beta-lactamases among spectrum beta-lactamase-producing Escherichia coli. Diagn
Enterobacteriaceae by use of semiautomated microbiology Microbiol Infect Dis 2006;56:351e357.
systems and manual detection procedures. J Clin Microbiol 29. Goethaert K, Van Looveren M, Lammens C, et al. High-dose
2007;45:1167e1174. cefepime as an alternative treatment for infections caused
13. Rodriguez-Bano J, Alcala JC, Cisneros JM, et al. Community by TEM-24 ESBL-producing Enterobacter aerogenes in
infections caused by extended-spectrum beta-lactamase- severely-ill patients. Clin Microbiol Infect 2006;12:56e62.
producing Escherichia coli. Arch Intern Med 2008;168: 30. Zanetti G, Bally F, Greub G, et al. Cefepime versus imi-
1897e1902. penem-cilastatin for treatment of nosocomial pneumonia
14. Paterson DL, Bonomo RA. Extended-spectrum beta-lacta- in intensive care unit patients: a multicenter, evaluator-
mases: a clinical update. Clin Microbiol Rev 2005;18: blind, prospective, randomized study. Antimicrob Agents
657e686. Chemother 2003;47:3442e3447.
15. Canton R, Novais A, Valverde A, et al. Prevalence and 31. Paterson DL, Ko WC, Von Gottberg A, et al. Outcome of ceph-
spread of extended-spectrum beta-lactamase-producing alosporin treatment for serious infections due to apparently
Enterobacteriaceae in Europe. Clin Microbiol Infect 2008; susceptible organisms producing extended-spectrum
14(Suppl. 1):144e153. beta-lactamases: implications for the clinical microbiology
16. Rodriguez-Bano J, Lopez-Cerero L, Navarro MD, Diaz de laboratory. J Clin Microbiol 2001;39:2206e2212.
Alba P, Pascual A. Faecal carriage of extended-spectrum 32. Wong-Beringer A, Hindler J, Loeloff M, et al. Molecular
beta-lactamase-producing Escherichia coli: prevalence, risk correlation for the treatment outcomes in bloodstream
factors and molecular epidemiology. J Antimicrob Chemo- infections caused by Escherichia coli and Klebsiella pneu-
ther 2008;62:1142e1149. moniae with reduced susceptibility to ceftazidime. Clin
17. Falagas ME, Karageorgopoulos DE. Clinical Microbiology and Infect Dis 2002;34:135e146.
Infectious Diseases (ECCMID) e 18th European Congress. 33. Bush K, Macalintal C, Rasmussen BA, Lee VJ, Yang Y. Kinetic
Drug resistance among Gram-negative and Gram-positive interactions of tazobactam with beta-lactamases from all
bacteria. IDrugs 2008;11:409e411. major structural classes. Antimicrob Agents Chemother
18. Valverde A, Coque TM, Garcia-San Miguel L, Baquero F, 1993;37:851e858.
Canton R. Complex molecular epidemiology of extended- 34. Payne DJ, Cramp R, Winstanley DJ, Knowles DJ. Compara-
spectrum beta-lactamases in Klebsiella pneumoniae: tive activities of clavulanic acid, sulbactam, and
a long-term perspective from a single institution in Madrid. tazobactam against clinically important beta-lactamases.
J Antimicrob Chemother 2008;61:64e72. Antimicrob Agents Chemother 1994;38:767e772.
354 M.E. Falagas, D.E. Karageorgopoulos

35. Burgess DS, Hall 2nd RG, Lewis 2nd JS, Jorgensen JH, 43. Kim BN, Woo JH, Kim MN, Ryu J, Kim YS. Clinical implica-
Patterson JE. Clinical and microbiologic analysis of a hospi- tions of extended-spectrum beta-lactamase-producing
tal’s extended-spectrum beta-lactamase-producing isolates Klebsiella pneumoniae bacteraemia. J Hosp Infect 2002;
over a 2-year period. Pharmacotherapy 2003;23: 52:99e106.
1232e1237. 44. Kang CI, Kim SH, Park WB, et al. Bloodstream infections due to
36. Ambrose PG, Bhavnani SM, Jones RN. Pharmacokinetics- extended-spectrum beta-lactamase-producing Escherichia
pharmacodynamics of cefepime and piperacillin-tazobactam coli and Klebsiella pneumoniae: risk factors for mortality
against Escherichia coli and Klebsiella pneumoniae strains and treatment outcome, with special emphasis on antimicro-
producing extended-spectrum beta-lactamases: report from bial therapy. Antimicrob Agents Chemother 2004;48:
the ARREST program. Antimicrob Agents Chemother 2003; 4574e4581.
47:1643e1646. 45. Falagas ME, Karageorgopoulos DE, Dimopoulos G. Clinical
37. Falagas ME, Polemis M, Alexiou VG, Marini- significance of the pharmacokinetic and pharmacodynamic
Mastrogiannaki A, Kremastinou J, Vatopoulos AC. Antimicro- characteristics of tigecycline. Curr Drug Metab 2009;10:
bial resistance of Esherichia coli urinary isolates from 13e21.
primary care patients in Greece. Med Sci Monit 2008;14: 46. Kelesidis T, Karageorgopoulos DE, Kelesidis I, Falagas ME.
CR75eCR79. Tigecycline for the treatment of multidrug-resistant
38. Lee CH, Su LH, Tang YF, Liu JW. Treatment of ESBL-producing Enterobacteriaceae: a systematic review of the evidence
Klebsiella pneumoniae bacteraemia with carbapenems or from microbiological and clinical studies. J Antimicrob
flomoxef: a retrospective study and laboratory analysis of Chemother 2008;62:895e904.
the isolates. J Antimicrob Chemother 2006;58:1074e1077. 47. Gales AC, Jones RN, Sader HS. Global assessment of the an-
39. Lee CH, Chu C, Liu JW, Chen YS, Chiu CJ, Su LH. Collateral timicrobial activity of polymyxin B against 54 731 clinical
damage of flomoxef therapy: in vivo development of porin isolates of Gram-negative bacilli: report from the SENTRY
deficiency and acquisition of blaDHA-1 leading to ertape- antimicrobial surveillance programme (2001e2004). Clin
nem resistance in a clinical isolate of Klebsiella pneumo- Microbiol Infect 2006;12:315e321.
niae producing CTX-M-3 and SHV-5 beta-lactamases. J 48. Falagas ME, Kanellopoulou MD, Karageorgopoulos DE, et al.
Antimicrob Chemother 2007;60:410e413. Antimicrobial susceptibility of multidrug-resistant Gram
40. Endimiani A, Luzzaro F, Perilli M, et al. Bacteremia due to negative bacteria to fosfomycin. Eur J Clin Microbiol Infect
Klebsiella pneumoniae isolates producing the TEM-52 ex- Dis 2008;27:439e443.
tended-spectrum beta-lactamase: treatment outcome of 49. Falagas ME, Giannopoulou KP, Kokolakis GN, Rafailidis PI.
patients receiving imipenem or ciprofloxacin. Clin Infect Fosfomycin: use beyond urinary tract and gastrointestinal
Dis 2004;38:243e251. infections. Clin Infect Dis 2008;46:1069e1077.
41. Paterson DL, Ko WC, Von Gottberg A, et al. Antibiotic ther- 50. Garau J. Other antimicrobials of interest in the era of ex-
apy for Klebsiella pneumoniae bacteremia: implications of tended-spectrum beta-lactamases: fosfomycin, nitrofuran-
production of extended-spectrum beta-lactamases. Clin toin and tigecycline. Clin Microbiol Infect 2008;14
Infect Dis 2004;39:31e37. (Suppl. 1):198e202.
42. Bassetti M, Righi E, Fasce R, et al. Efficacy of ertapenem in 51. Livermore DM. Defining an extended-spectrum beta-
the treatment of early ventilator-associated pneumonia lactamase. Clin Microbiol Infect 2008;14(Suppl. 1):3e10.
caused by extended-spectrum beta-lactamase-producing 52. Giske CG, Sundsfjord AS, Kahlmeter G, et al. Redefining
organisms in an intensive care unit. J Antimicrob Chemo- extended-spectrum beta-lactamases: balancing science
ther 2007;60:433e435. and clinical need. J Antimicrob Chemother 2009;63:1e4.
Journal of Hospital Infection (2009) 73, 355e363
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Acinetobacter: an old friend, but a new enemy


K.J. Towner*

Department of Clinical Microbiology, Nottingham University Hospitals NHS Trust, Queen’s Medical
Centre, Nottingham NG7 2UH, UK

Available online 22 August 2009

KEYWORDS Summary Acinetobacter emerged as a significant nosocomial pathogen


Acinetobacter; during the late 1970s, probably as a consequence, at least in part, of in-
Epidemiology; creasing use of broad-spectrum antibiotics in hospitals. Most clinically sig-
Infection control
nificant isolates belong to the species Acinetobacter baumannii or its close
measures;
Nosocomial infection;
relatives, with many infections concentrated in intensive care, burns or
Therapeutic options high dependency units treating severely ill or debilitated patients. Large
outbreaks can occur in such units, involving the infection or colonisation
of numerous patients by specific epidemic strains of A. baumannii.
Recently, a particular problem has concerned cross-infection of injured
military patients repatriated from combat regions of the world (e.g. Iraq
and Afghanistan). Carbapenems have previously been the treatment of
choice for infected patients, but increasing reports worldwide now
describe A. baumannii isolates resistant to all conventional antimicrobial
regimens. Data to support therapeutic use of the limited number of new
antimicrobial agents (e.g. tigecycline) with in-vitro activity against these
pathogens are still very limited. Detailed advice concerning prevention
and control of outbreaks caused by multidrug-resistant strains of acineto-
bacter is available from the UK Health Protection Agency. In addition to an-
tibiotic prescribing policies and audit, these measures focus on reinforcing
standard infection control procedures and precautions, with particular
attention to thorough cleaning of patient areas to take account of the
long-term survival of acinetobacter after drying and inadequate disinfec-
tion. Despite these measures, the problem continues to escalate, with
many hospitals worldwide now reporting outbreaks caused by multidrug-
resistant strains of acinetobacter.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

* Tel.: þ44 115 9709163; fax: þ44 115 9422190.


E-mail address: Kevin.Towner@nuh.nhs.uk

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.03.032
356 K.J. Towner

Introduction antimicrobial agents, such as ampicillin (60e70% of


isolates susceptible), gentamicin (92.5%), chloram-
The genus Acinetobacter has a long and convo- phenicol (57%) and nalidixic acid (97.8%), so that in-
luted taxonomic history. It was in 1911 that Beijer- fections caused by these organisms could be treated
inck, a Dutch microbiologist working in Delft, relatively easily.2 However, multidrug-resistant
isolated and described the first example of an (MDR) clinical isolates of Acinetobacter spp. have
organism that would now be recognised as Acine- been reported increasingly during the last two de-
tobacter.1 Since then, members of the genus cades, almost certainly as a consequence of exten-
have been classified under a variety of different sive use of potent broad-spectrum antimicrobial
names (e.g. Bacterium anitratum, Herellea vagini- agents in hospitals throughout the world.
cola, Mima polymorpha, Moraxella lwoffi), and In the clinical environment, Acinetobacter bau-
this confusion resulted in difficulties in establish- mannii and its close relatives (genomic species 3
ing the epidemiology and true clinical importance and 13TU), together forming the ‘A. baumannii
of these organisms. Today, the use of molecular complex’, are the genomic species of greatest clin-
methods has established the identity of at least ical importance, together accounting for the vast
33 different species belonging to the genus Acine- majority of infections and hospital outbreaks in-
tobacter, of which 18 have now been assigned for- volving Acinetobacter spp.3e5 The members of
mal species names (Table I). A further 28 unnamed the complex are very difficult for routine diagnos-
groups have been identified that contain multiple tic laboratories to distinguish accurately; there-
strains, and there are also at least 21 ungrouped fore, reports of A. baumannii in the scientific
single strains. However, although the genus Acine- and medical literature should be assumed to in-
tobacter comprises an ever-growing number of de- clude the other members of the complex unless
fined bacterial species, only a minority of these this possibility has been specifically excluded.
species are of clinical significance (see below). Many of the infections caused by these organisms
Members of the genus Acinetobacter first began occur in intensive care or high dependency units
to be recognised as significant nosocomial pathogens in which severely ill or debilitated patients are
during the early 1970s. In early in-vitro studies, most treated extensively with broad-spectrum antimi-
clinical isolates were susceptible to commonly used crobial agents. Members of the A. baumannii com-
plex seem to have an extraordinary ability to
develop resistance to even the most potent antimi-
Table I Species with validly described names that crobial compounds, and are able to rapidly fill the
belong to the genus Acinetobacter a,b ecological niche left vacant by the elimination of
A. calcoaceticus competing bacteria with broad-spectrum com-
A. baumannii pounds. Genetic interchange with other bacterial
A. haemolyticus species that are capable of growing at lower envir-
A. junii onmental temperatures is also possible, with
A. johnsonii
significant implications for the wider spread of
A. lwoffii
A. radioresistens
antibiotic resistance genes in the environment.
A. ursingii
A. schindleri
A. parvus Common misconceptions
A. baylyi
A. bouvetii Many misconceptions concerning acinetobacter still
A. towneri appear repeatedly in the scientific and medical liter-
A. tandoii ature. Chief among these are the statements that
A. tjernbergiae A. baumannii is (i) ubiquitous or highly prevalent in
A. gerneri nature, (ii) that it can be recovered easily from
A. beijerinckii
soil, water and animals, and (iii) that it is a frequent
A. gyllenbergii
a
skin and oropharyngeal commensal of humans. While
A. grimontii has recently been demonstrated to be a het- these statements certainly apply to members of the
erotypic synonym of A. junii, while A. venetianus currently
remains as a provisional designation awaiting further
genus Acinetobacter when considered as a whole,
investigation. A. baumannii (and its close relatives of clinical
b
In addition to the 18 named species listed, a further 26 importance) are not ubiquitous organisms. While it
unnamed groups have been identified that contain multiple is certainly true that A. baumannii can be isolated
strains, and there are also at least 21 ungrouped single from patients and hospital environmental sources
strains that exist in culture collections.
during outbreaks, this species has no known natural
Acinetobacter 357

habitat outside the hospital.5 This species can be iso- by the high degree of resistance of these organisms
lated only very rarely from soil, water and other en- to drying and disinfectants, leading to long-term
vironmental samples; indeed, during non-outbreak persistence in the hospital environment and the
periods it is often isolated only rarely inside occurrence of outbreaks of infection involving
hospitals. many patients, and the ever-increasing proportion
In summary, three major overlapping popula- of isolates that are MDR. Numerous studies have in-
tions of the genus Acinetobacter been identified to dicated an upward trend in MDR A. baumannii, but
date: (i) MDR isolates found primarily in hospitals resistance rates can vary widely according to the
during outbreak situations e these isolates are individual hospital, city or country involved.
capable of colonising and infecting hospitalised Thus, in the UK, the latest available surveillance
patients, and comprise mainly A. baumannii and data show that resistance to carbapenems has in-
its close relatives; (ii) ‘sensitive’ isolates that creased from <0.5% in 1990 to 24% in 2007 (UK
form part of the normal commensal skin flora of Health Protection Agency; unpublished data).
humans (25e70% of individuals) and animals, and Even allowing for the distortions caused by re-
which can also be found as part of the spoilage peated sampling of predominant MDR epidemic
flora of many different foodstuffs e these isolates strains, it is clear that A. baumannii has a remark-
comprise mainly A. johnsonii, A. lwoffii and A. able ability to acquire resistance genes and to de-
radioresistens; and (iii) ‘sensitive’ isolates that velop resistance against virtually all available
can be found in the environment, soil or wastewa- antibiotic classes (see below).
ters e these isolates comprise mainly A. calcoace- The ‘reservoir’ for nosocomial infection with
ticus and A. johnsonii. The natural habitats of the A. baumannii still remains uncertain, and may vary
many other species belonging to the genus Acine- among healthcare facilities. Large numbers of
tobacter (Table I) are still poorly defined. Most A. baumannii infections have recently been re-
species have been isolated on an occasional basis ported among military casualties repatriated
from humans, including clinical specimens, but in from war zones in Iraq and Afghanistan.6 Based
terms of infection control and clinical importance, on the misconception that A. baumannii is ubiqui-
concern should be reserved for the MDR isolates tous in the environment, it was originally thought
belonging to the A. baumannii complex. Such or- that such infections might have originated from
ganisms are not ubiquitous and are generally iso- contact with contaminated soil in the war zones.
lated only from colonised or infected patients, or However, it has now become clear that the main
from the hospital environment during outbreaks. cause was contamination of the environment of
field hospitals and other healthcare facilities
involved in the repatriation of military casualties.7
Acinetobacter as a nosocomial pathogen Potential sources of infection in the hospital
environment from which these organisms have
The main problems caused by acinetobacter in the been isolated during outbreaks are numerous
hospital setting mostly concern critically ill pa- (Table II). Airborne transmission and patient-
tients in intensive care units (ICUs), particularly to-patient transmission have also been demon-
those requiring mechanical ventilation, and pa- strated. However, although the hands of hospital
tients with wound or burn injuries (trauma pa- personnel, coupled with contamination of environ-
tients). Infections associated with acinetobacter mental surfaces and medical equipment, may play
include ventilator-associated pneumonia, skin and a role in the spread of A. baumannii during an out-
soft-tissue infections, wound infections, urinary break, it seems likely that the infected patient
tract infections, secondary meningitis and blood- forms the primary reservoir of infection, with
stream infections.2,5 Such infections are caused such patients shedding extremely large numbers
predominantly by members of the A. baumannii of A. baumannii cells into their surrounding envir-
complex; nosocomial infections caused by other onment. Factors facilitating the spread of A. bau-
species belonging to the genus Acinetobacter are mannii are listed in Table III.
relatively unusual and are restricted mainly to
catheter-related bloodstream infections and rare
outbreaks related to point-source contamination. Infection control measures
There have also been a few reports of commu-
nity-acquired infections, usually in patients with Once endemic in a healthcare unit, A. baumannii is
comorbidities in tropical or subtropical areas.5 extremely difficult to eradicate. Nevertheless, it is
Problems caused by members of the A. bauman- still possible to eradicate these organisms from
nii complex in the hospital setting are exacerbated a unit when an uncompromising approach is taken
358 K.J. Towner

the environment, and the use of nebulised colistin


Table II Potential sources from which Acineto-
bacter baumannii has been isolated in the hospital for patients with evidence of mild-to-moderate
environment ventilator-associated pneumonia.8 Nevertheless,
there are also numerous examples in which it has
Hands of staff
been necessary to implement patient isolation
Ventilators and tubing
Oxygen analysers and/or ward closures for periods of up to four
Bronchoscopes weeks in order to combat A. baumannii out-
Bed frames breaks.9e16 Detailed guidance concerning contact
Sinks isolation precautions, risk factors for colonisation
Jugs or infection, antibiotic prescribing policies, patient
Soap transfer procedures (internal and external), use of
Plastic screens dedicated equipment, screening strategies, and
Bed linen, pillows and mattresses cleaning and decontamination procedures has been
Resuscitation bags made available by a Working Party of the UK Health
Blood pressure cuffs
Protection Agency (http://www.hpa.org.uk/web/
Parenteral nutrition solution
HPAwebFile/HPAweb_C/1194947325341).17
Gloves
Humidifiers To reiterate, the most important source of A.
Patients baumannii in a potential outbreak situation is the
Respirometers already colonised or infected patient. If an in-
Lotion dispensers crease in the number of cases is detected, the iso-
Rubbish bins lates should first be identified and typed, the
Air supply patients involved should be traced and isolated
Bowls where possible, hygiene and infection control pro-
Hand cream cedures should be re-emphasised and enhanced,
Bedside charts antibiotic policies should be reviewed, and the
Service ducts/dust
unit should be cleaned thoroughly.
Computer keyboards
Cell phones
Epidemiology

Numerous studies have investigated the epidemiol-


to infection control. Normal infection control mea-
ogy of A. baumannii. Various patterns of spread of
sures are often insufficient to halt the transmission
individual A. baumannii strains have been reported,
of MDR A. baumannii, but a range of enhanced
including outbreaks caused by the same strain in
measures that focused on controlling environmen-
multiple hospitals within a city, outbreaks in mul-
tal contamination of an outbreak strain proved to
tiple cities within a country, and outbreaks in hospi-
be successful in eradicating A. baumannii from
tals in several different countries.18e25 In the UK,
the ICU of a major London teaching hospital.8
a survey in 1999e2001 identified 34 different A.
Key measures included use of a closed tracheal
baumannii genotypes in 46 UK hospitals.26 These
suction system for all patients receiving mechani-
genotypes belonged to 10 different clusters, with
cal ventilation, improved hand decontamination
particular strains generally being characteristic of
using alcohol gels, clearer designation of responsi-
particular hospitals.26 Between 2003 and 2006,
bilities and strategies for cleaning equipment and
two carbapenem-resistant A. baumannii lineages
(SE clone and OXA-23 clone) became prevalent in
over 40 UK hospitals each, with these lineages being
Table III Factors facilitating the spread of Acineto- susceptible only to colistin and tigecycline.22 More
bacter baumannii recently, a further lineage (the Northwest strain)
has become prevalent in several hospitals in the
Increased length of hospital stay
northern/midland regions of the UK.
Prior antibiotics
Mechanical ventilation The question of whether specific carbapenem-
Exposure to patients colonised or infected with resistant clones are spreading in European hospi-
A. baumannii tals has also been examined. As part of the EU
Environmental contamination ARPAC project, 169 hospitals in 32 countries pro-
Understaffing vided data concerning MDR isolates of Acineto-
Poor adherence of staff to hand hygiene bacter spp.27 In total, 130 hospitals reported
encountering carbapenem-resistant isolates of
Acinetobacter 359

acinetobacter, ranging from rare sporadic isolates the results of properly performed antimicrobial
to an endemic/epidemic situation. Diverse clusters susceptibility tests. Antibiotic selection for empir-
were identified in European hospitals by random ical therapy is challenging and should rely,
amplified polymorphic DNA, pulsed-field gel elec- wherever possible, on institutional-level data con-
trophoresis and polymerase chain reaction-based cerning the phenotypes and genotypes of A. bau-
sequence typing, but three major European lin- mannii strains endemic in a particular hospital
eages were identified. As in the UK, multiple iso- environment. No large prospective controlled clin-
lates from a single hospital generally belonged to ical trials for the treatment of A. baumannii infec-
the same clone.27 tions have been reported to date. Knowledge of
the best therapeutic approaches is therefore
based on in-vitro susceptibility data, small case
Antimicrobial therapy series and retrospective analysis of observational
studies. Of the available options (Table IV), wide-
Table IV lists the classes of antimicrobial agents that spread resistance means that broad-spectrum
are currently considered to have potential activity penicillins, broad-spectrum cephalosporins and
against A. baumannii. However, successive surveys monobactams cannot be recommended as suitable
have shown increasing resistance among clinical candidates for the empirical treatment of severe
isolates, and high proportions of isolates are A. baumannii infections, although cefepime and
now resistant to most commonly used antibacterial cefpirome retain some potentially useful residual
agents, including aminopenicillins, ureido- activity.
penicillins, broad-spectrum cephalosporins, most
aminoglycosides, quinolones, chloramphenicol and
Aminoglycosides
tetracyclines.2e4 As a consequence, the emergence
of MDR strains of acinetobacter, probably following
Resistance rates to all the clinically useful amino-
the increasing use of broad-spectrum antibiotics in
glycoside antibiotics are higher in Acinetobacter
hospitals, has resulted in carbapenems (especially
spp. than in most other groups of pathogens.3,35
imipenem and meropenem) becoming the mainstay
Most aminoglycoside resistance in Acinetobacter
of treatment for acinetobacter infections. How-
spp. involves production of aminoglycoside-modi-
ever, reports of carbapenem resistance in clinical
fying enzymes, and all three classes of aminoglyco-
isolates of Acinetobacter spp. are now accumulat-
side-modifying enzymes have been found in
ing worldwide, with some isolates being resistant
Acinetobacter spp. Nevertheless, depending on
to all conventional antimicrobial agents.18,28e30 It
the results of susceptibility tests, aminoglycosides,
is therefore vital that therapeutic strategies
particularly amikacin, continue to show useful ac-
optimise the use of existing antimicrobial agents
tivity against 40e50% of strains in some centres.
and minimise the possibilities for the evolution of
drug resistance. Fluoroquinolones
Potential therapeutic approaches for treatment
of A. baumannii infections have been considered Until 1990, quinolones had reasonably good activity
in detail in several recent review articles.5,31e34 against A. baumannii, but resistance to these anti-
The antibiotic classes listed in Table IV have poten- biotics subsequently emerged rapidly in clinical iso-
tial activity against individual strains of acineto- lates. Newer fluoroquinolones, such as
bacter, but therapy should ideally be based on moxifloxacin, sometimes have increased activity
in vitro against A. baumannii in comparison with
older agents such as ciprofloxacin. Spence and
Table IV Currently available classes of antimicro- Towner demonstrated that isolates with a single
bial agents with potential activity against Acineto- mutation in the gyrA gene at codon Ser-83 had
bacter baumannii a ciprofloxacin minimum inhibitory concentration
Anti-pseudomonal broad-spectrum penicillins (MIC) of 2 to >32 mg/L, but a moxifloxacin MIC of
Anti-pseudomonal broad-spectrum cephalosporins 0.25e1 mg/L.36 All moxifloxacin-resistant (MIC
Monobactams >2 mg/L) clinical isolates possessed a second
Aminoglycosides mutation in codon Ser-80 of the parC gene. Addi-
Fluoroquinolones
tional single-step mutation studies with ciprofloxa-
Carbapenems
cin-resistant/moxifloxacin-susceptible isolates
Polymyxins
Sulbactam revealed that such stable moxifloxacin-resistant
Glycylcyclines mutants were difficult to generate, but that growth
could occur in the presence of moxifloxacin,
360 K.J. Towner

perhaps indicating the presence of an efflux pump antimicrobial properties of these agents against
that could be induced in the presence of A. baumannii. However, as currently formulated
subinhibitory concentrations of moxifloxacin. for clinical use, sulbactam is likely to be the only
one of these inhibitors to be active in vivo against
Carbapenems A. baumannii. Recent evidence has suggested that
the activity of sulbactam against A. baumannii iso-
As mentioned above, carbapenems (especially imi- lates has declined significantly, perhaps in response
penem and meropenem) have been a mainstay of to the increased clinical use of this compound.34
treatment for acinetobacter infections for the past
decade. However, although these agents are still Tigecycline
active against many strains, increasing numbers of
clinical isolates of A. baumannii resistant to carba- Tigecycline is a member of a new class of tetracy-
penems are now being reported worldwide, reach- cline-related antimicrobial agents, the glycylcy-
ing levels of 90% in some centres.34 In northern clines, and was found to have good in-vitro activity
European countries (e.g. the UK), carbapenem against some carbapenem-resistant acinetobacter
resistance is now reported in w15% of A. baumannii isolates in preliminary studies.43,44 However, clini-
isolates, but levels are much higher in southern cal experience regarding the use of tigecycline for
European countries (e.g. Spain, Italy, Greece). the treatment of patients with MDR A. baumannii
infections has been somewhat mixed. In the avail-
Polymyxins able clinical reports, tigecycline has mainly been
used, often in combination regimens, to treat
Polymyxins (e.g. colistin) show bactericidal activ- a small number of critically ill patients with infec-
ity against A. baumannii, and resistance rates have tions such as ventilator-associated pneumonia and
remained relatively low. Favourable clinical re- primary or secondary bacteraemia.45,46 Most pa-
sponses have been reported for intravenous colis- tients had a good outcome, but failure of tigecy-
tin therapy in case series of ICU patients with cline to clear A. baumannii bacteraemia was
various types of infections, including ventilator- noted in a few cases, perhaps because of subopti-
associated pneumonia and nosocomial meningi- mal concentrations of tigecycline in blood. In-vitro
tis.34 Perhaps surprisingly, reports of colistin toxic- studies have revealed that resistance to tigecy-
ity are relatively rare. Polymyxins can be cline can be mediated by overexpression of
administered in a nebulised form via the respira- a multidrug efflux pump, and the emergence of re-
tory tract in patients with nosocomial pneumonia, sistance to tigecycline during therapy has already
and several small studies have reported clinical ef- been reported.45,47
fectiveness.34 Some studies have indicated that
colistin may be useful for treating infections
caused by carbapenem-resistant isolates, but in- Combination therapy
creasing use of polymyxins in critically ill patients
may lead rapidly to the emergence of resistan- Various combinations of a carbapenem with sul-
ce.37e39 Indeed, high rates of resistance to colistin bactam, tobramycin, amikacin, colistin, rifampicin
have recently been reported in A. baumannii and aztreonam have been assessed, both in vitro
isolates from two South Korean hospitals.40 and in vivo, with somewhat mixed results.34 Other
reports have described the successful use of sul-
Sulbactam bactam, which has unusual intrinsic activity
against Acinetobacter spp. (see above), in combi-
A few reports have described the successful ther- nation with ampicillin, or have proposed unusual
apeutic use of sulbactam, which has unusual in- combinations of antibiotics, such as polymyxin B,
trinsic activity against Acinetobacter spp., either imipenem and rifampicin.48e50 Clinical data are
alone or in combination with ampicillin.41 Urban too few to recommend the use of specific combina-
et al. investigated the interaction of sulbactam, tion regimens for the treatment of infections
clavulanic acid and tazobactam with the penicil- caused by MDR strains of A. baumannii, but combi-
lin-binding proteins (PBPs) of imipenem-resistant nation regimens might be considered by clinicians
and -susceptible isolates of A. baumannii, and in order to achieve synergistic activity and to max-
showed in competition-binding experiments that imise antimicrobial effectiveness (as well as to
all three of these b-lactamase inhibitors bound to minimise the possibility of emergence of further
the PBPs of the imipenem-susceptible isolates.42 resistance) in severely ill patients for whom thera-
This observation may explain the in-vitro intrinsic peutic options are otherwise limited.
Acinetobacter 361

Discussion these lineages are selected primarily on the basis


of the resistance genes that they carry or because
A. baumannii and its close relatives have become there is something special about certain lineages
a major cause of hospital-acquired infections pri- that confers epidemic potential currently remains
marily because of the remarkable ability of these unanswered.
bacteria to survive and spread in the hospital en-
vironment and to rapidly acquire resistance deter-
minants to a wide range of antibacterial agents.
Epidemic spread of MDR strains among patients in Conflict of interest statement
hospitals, particularly in ICUs, has been observed None.
frequently, with infected patients disseminating
large numbers of these organisms into their en- Funding sources
vironment. The problem is then compounded by Work on acinetobacter in K.J.T.’s laboratory is
the long-term survival of these organisms on supported, in part, by a grant from the Medical
numerous surfaces and inanimate objects, and by Research Council (grant no. RA0119).
their high degree of resistance to drying,
disinfectants and antibiotics. Consequently, once
endemic, these organisms are extremely difficult References
to eradicate from a particular healthcare unit.
Other special features of the genus Acineto- 1. Dijkshoorn L, Nemec A. The diversity of the genus Acineto-
bacter include the fact that, perhaps by accident, bacter. In: Gerischer U, editor. Acinetobacter molecular
it has evolved a range of its own special resistance biology. Norfolk: Caister Academic Press; 2008. p. 1e34.
2. Bergogne-Bérézin E. Resistance of Acinetobacter spp. to an-
genes (particularly carbapenemases), as well as timicrobials e overview of clinical resistance patterns and
the capacity to overexpress them in response to therapeutic problems. In: Bergogne-Bérézin E, Joly-
antibiotic challenge. It has also evolved molecular Guillou ML, Towner KJ, editors. Acinetobacter, microbiol-
mechanisms to capture resistance genes from ogy, epidemiology, infections, management. Boca Raton:
other organisms, and has developed a range of ex- CRC Press; 1996. p. 133e183.
3. Bergogne-Bérézin E, Towner KJ. Acinetobacter spp. as nos-
pression mechanisms (e.g. provision of promoters ocomial pathogens: microbiological, clinical, and epidemio-
on insertion sequences) that enables ‘foreign’ re- logical features. Clin Microbiol Rev 1996;9:148e165.
sistance genes to be expressed. Consequently, an- 4. Van Looveren M, Goosens H, ARPAC Steering Group. Antimi-
timicrobial resistance now greatly limits the crobial resistance of Acinetobacter spp. in Europe. Clin
available therapeutic options, especially if isolates Microbiol Infect 2004;10:684e704.
5. Peleg AY, Seifert H, Paterson DL. Acinetobacter baumannii:
are resistant to the carbapenem class of antimicro- emergence of a successful pathogen. Clin Microbiol Rev
bial agents, which would otherwise be the agents 2008;21:538e582.
of choice for the treatment of severe infections 6. Centers for Disease Control and Prevention. Acinetobacter
caused by these organisms. Knowledge of the sus- baumannii infections among patients at military medical
ceptibility patterns of strains endemic in particular facilities treating injured US service members, 2002e
2004. Morb Mortal Wkly Rep 2004;53:1063e1066.
geographical areas is essential, but therapy of in- 7. Scott P, Deye G, Srinivasan A, et al. An outbreak of multi-
fections caused by carbapenem-resistant strains drug-resistant Acinetobacter baumannii e calcoaceticus
frequently requires the use of unusual drugs (e.g. complex infection in the US military health care system
colistin or sulbactam), often in novel combina- associated with military operations in Iraq. Clin Infect Dis
tions, or of drugs for which there is presently 2007;44:1577e1584.
8. Wilks M, Wilson A, Warwick S, et al. Control of an outbreak
very little clinical experience (e.g. tigecycline). of multidrug-resistant Acinetobacter baumannii coloniza-
Anecdotes, small case series and case reports sug- tion and infection in an intensive care unit (ICU) without
gest that such regimens can be efficacious in indi- closing the ICU or placing patients in isolation. Infect Con-
vidual patients, although reports of resistance to trol Hosp Epidemiol 2006;27:654e658.
new drugs and unusual combinations are already 9. Idzenga D, Schouten MA, van Zanten AR. Outbreak of Acine-
tobacter genomic species 3 in a Dutch intensive care unit.
beginning to appear. J Hosp Infect 2006;63:485e487.
Well-controlled clinical trials of existing and 10. Kraniotaki E, Manganelli R, Platsouka E, Grossato A,
new therapies, combined with greater emphasis Paniara O, Palù G. Molecular investigation of an outbreak
on the prevention of healthcare-associated A. bau- of multidrug-resistant Acinetobacter baumannii, with char-
mannii infections, are essential in order to combat acterisation of class 1 integrons. Int J Antimicrob Agents
2006;28:193e199.
the spread of these MDR organisms. Worldwide 11. Longo B, Pantosti A, Luzzi I, et al. An outbreak of Acineto-
spread of MDR lineages of A. baumannii is now be- bacter baumannii in an intensive care unit: epidemiological
ing observed, although the question of whether and molecular findings. J Hosp Infect 2006;64:303e305.
362 K.J. Towner

12. Carbonne A, Naas T, Blanckaert K, et al. Investigation of 30. Falagas ME, Bliziotis IA. Pandrug-resistant gram-negative
a nosocomial outbreak of extended-spectrum beta-lacta- bacteria: the dawn of the post-antibiotic era? Int J Antimi-
mase VEB-1-producing isolates of Acinetobacter baumannii crob Agents 2007;29:630e636.
in a hospital setting. J Hosp Infect 2005;60:14e18. 31. Gilad J, Carmeli Y. Treatment options for multidrug-resis-
13. Pimentel JD, Low J, Styles K, Harris OC, Hughes A, Athan E. tant Acinetobacter species. Drugs 2008;68:165e189.
Control of an outbreak of a multi-drug-resistant Acineto- 32. Maragakis LL, Perl TM. Acinetobacter baumannii: epidemi-
bacter baumannii in an intensive care unit and a surgical ology, antimicrobial resistance, and treatment options. Clin
ward. J Hosp Infect 2005;59:249e253. Infect Dis 2008;46:1254e1263.
14. De Jong G, Duse A, Richards G, Marais E. Back to basics e 33. Dijkshoorn L, Nemec A, Seifert H. An increasing threat in
optimizing the use of available resources during an out- hospitals: multidrug-resistant Acinetobacter baumannii.
break of multi-drug resistant Acinetobacter spp. J Hosp Nat Rev Microbiol 2007;5:939e951.
Infect 2004;57:186e187. 34. Karageorgopoulos DE, Falagas ME. Current control and
15. Denton M, Wilcox MH, Parnell P, et al. Role of environmen- treatment of multidrug-resistant Acinetobacter baumannii
tal cleaning in controlling an outbreak of Acinetobacter infections. Lancet Infect Dis 2008;8:751e762.
baumannii on a neurosurgical intensive care unit. J Hosp In- 35. Miller JH, Sabatelli FJ, Naples L, Hare RS, Shaw KJ. The
fect 2004;56:106e110. most frequently occurring aminoglycoside resistance mech-
16. Maragakis LL, Tucker MG, Miller RG, Carroll KC, Perl TM. In- anisms e combined results of surveys in eight regions of the
cidence and prevalence of multidrug-resistant acineto- world. J Chemother 1995;7(Suppl.):17e30.
bacter using targeted active surveillance cultures. J Am 36. Spence RP, Towner KJ. Frequencies and mechanisms of resis-
Med Assoc 2008;299:2513e2514. tance to moxifloxacin in nosocomial isolates of Acinetobacter
17. Health Protection Agency. Working party guidance on the baumannii. J Antimicrob Chemother 2003;52:687e690.
control of multi-resistant Acinetobacter outbreaks. 37. Garnacho-Montero J, Ortiz-Leyba C, Jiménez-Jiménez FJ,
18. Landman D, Quale JM, Mayorga D, et al. Citywide clonal et al. Treatment of multidrug-resistant Acinetobacter bau-
outbreak of multiresistant Acinetobacter baumannii and mannii ventilator-associated pneumonia (VAP) with intra-
Pseudomonas aeruginosa in Brooklyn, NY: the preanti- venous colistin: a comparison with imipenem-susceptible
biotic era has returned. Arch Intern Med 2002;162: VAP. Clin Infect Dis 2003;36:1111e1118.
1515e1520. 38. Michalopoulos A, Kasiakou SK, Rosmarakis ES, Falagas ME.
19. Turton JF, Kaufmann ME, Warner M, et al. A prevalent mul- Cure of multidrug-resistant Acinetobacter baumannii bac-
tiresistant clone of Acinetobacter baumannii in Southeast teraemia with continuous intravenous infusion of colistin.
England. J Hosp Infect 2004;58:170e179. Scand J Infect Dis 2005;37:142e145.
20. Marais E, de Jong G, Ferraz V, Maloba B, Dusé AG. Inter- 39. Matthaiou DK, Michalopoulos A, Rafailidis PI, et al. Risk
hospital transfer of pan-resistant Acinetobacter strains in factors associated with the isolation of colistin-resistant
Johannesburg, South Africa. Am J Infect Control 2004;32: gram-negative bacteria: a matched caseecontrol study.
278e281. Crit Care Med 2008;36:807e811.
21. Nemec A, Dijkshoorn L, van der Reijden TJ. Long-term pre- 40. Ko KS, Suh JY, Kwon KT, et al. High rates of resistance to
dominance of two pan-European clones among multi-resis- colistin and polymyxin B in subgroups of Acinetobacter
tant Acinetobacter baumannii strains in the Czech baumannii isolates from Korea. J Antimicrob Chemother
Republic. J Med Microbiol 2004;53:147e153. 2007;60:1163e1167.
22. Coelho JM, Turton JF, Kaufmann ME, et al. Occurrence of 41. Levin AS, Levy CE, Manrique AE, Medeiros EA, Costa SF.
carbapenem-resistant Acinetobacter baumannii clones at Severe nosocomial infections with imipenem-resistant
multiple hospitals in London and Southeast England. J Clin Acinetobacter baumannii treated with ampicillin/sulbac-
Microbiol 2006;44:3623e3627. tam. Int J Antimicrob Agents 2003;21:58e62.
23. Naas T, Coignard B, Carbonne A, et al. VEB-1 extended- 42. Urban C, Go E, Mariano N, Rahal JJ. Interaction of sulbac-
spectrum beta-lactamase-producing Acinetobacter tam, clavulanic acid and tazobactam with penicillin-binding
baumannii, France. Emerg Infect Dis 2006;12:1214e1222. proteins of imipenem-resistant and -susceptible Acineto-
24. van Dessel H, Dijkshoorn L, van der Reijden T, et al. Identi- bacter baumannii. FEMS Microbiol Lett 1995;125:193e198.
fication of a new geographically widespread multiresistant 43. Henwood CJ, Gatward T, Warner M, et al. Antibiotic resis-
Acinetobacter baumannii clone from European hospitals. tance among clinical isolates of Acinetobacter in the UK,
Res Microbiol 2004;155:105e112. and in vitro evaluation of tigecycline (GAR-936). J Antimi-
25. Seifert H, Dolzani L, Bressan R, et al. Standardization and crob Chemother 2002;49:479e487.
interlaboratory reproducibility assessment of pulsed-field 44. Pachón-Ibáñez ME, Jiménez-Mejı́as ME, Pichardo C,
gel electrophoresis-generated fingerprints of Acinetobacter Llanos AC, Pachón J. Activity of tigecycline (GAR-936)
baumannii. J Clin Microbiol 2005;43:4328e4335. against Acinetobacter baumannii strains, including those
26. Spence RP, Towner KJ, Henwood CJ, James D, Woodford N, resistant to imipenem. Antimicrob Agents Chemother
Livermore DM. Population structure and antibiotic resis- 2004;48:4479e4481.
tance of Acinetobacter DNA group 2 and 13TU isolates from 45. Karageorgopoulos DE, Kelesidis T, Kelesidis I, Falagas ME.
hospitals in the UK. J Med Microbiol 2002;51:1107e1112. Tigecycline for the treatment of multidrug-resistant
27. Towner KJ, Levi K, Vlassiadi M, ARPAC Steering Group. (including carbapenem-resistant) Acinetobacter infections:
Genetic diversity of carbapenem-resistant isolates of Acine- a review of the scientific evidence. J Antimicrob Chemo-
tobacter baumannii in Europe. Clin Microbiol Infect 2008; ther 2008;62:45e55.
14:161e167. 46. Gordon NC, Wareham DW. A review of clinical and microbi-
28. Coelho J, Woodford N, Turton J, Livermore DM. Multi-resis- ological outcomes following treatment of infections involv-
tant acinetobacter in the UK: how big a threat? J Hosp ing multidrug-resistant Acinetobacter baumannii with
Infect 2004;58:167e169. tigecycline. J Antimicrob Chemother, 2009;63:775e780.
29. Poirel L, Nordmann P. Carbapenem resistance in Acineto- 47. Peleg AY, Adams J, Paterson DL. Tigecycline efflux as a mech-
bacter baumannii: mechanisms and epidemiology. Clin anism for nonsusceptibility in Acinetobacter baumannii.
Microbiol Infect 2006;12:826e836. Antimicrob Agents Chemother 2007;51:2065e2069.
Acinetobacter 363

48. Levin AS. Multiresistant Acinetobacter infections: a role for Acinetobacter baumannii. Antimicrob Agents Chemother
sulbactam combinations in overcoming an emerging world- 2004;48:753e757.
wide problem. Clin Microbiol Infect 2002;8:144e153. 50. Wareham DW, Bean DC, Urban CM, Rahal JJ. In vitro activ-
49. Yoon J, Urban C, Terzian C, Mariano N, Rahal JJ. In vitro ities of polymyxin B, imipenem, and rifampin against multi-
double and triple synergistic activities of polymyxin B, drug-resistant Acinetobacter baumannii. Antimicrob Agents
imipenem, and rifampin against multidrug-resistant Chemother 2006;50:825e826.
Journal of Hospital Infection (2009) 73, 364e370
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Community-associated meticillin-resistant
Staphylococcus aureus as a cause of
hospital-acquired infections
R.L. Skov*, K.S. Jensen

National Centre for Antimicrobials and Infection Control, Statens Serum Institut, Copenhagen, Denmark

Available online 27 September 2009

KEYWORDS Summary The worldwide emergence of community-associated meticillin-


Community-associated resistant Staphylococcus aureus (CA-MRSA) during the last decade repre-
MRSA; sents a significant change in the biology of MRSA strains and is changing
Hospital-acquired
the epidemiology of MRSA infections. CA-MRSA infections are caused by
infections;
Mathematical
strains belonging to lineages distinct from HA-MRSA. In the community,
modelling; CA-MRSA strains typically cause skin and soft tissue infections in children
Staphylococcus aureus and younger adults. However, CA-MRSA strains increasingly cause health-
care-acquired infections including surgical site infections, ventilator-asso-
ciated pneumonia and bacteraemia. A mathematical model showing the
influence of MRSA transmission in the community on the prevalence of
MRSA in hospitals is presented. The increasing prevalence of MRSA in the
community also results in an increase in community-onset MRSA (CO-MRSA)
among S. aureus bacteraemia and other invasive infections. These patients
do not have typical risk factors for MRSA. Such changes may have profound
implications for the choice of empirical therapy for serious infections
where S. aureus is a possible cause. The new and potentially very large
reservoir of MRSA in production animals with subsequent transmission to
humans represents an additional serious threat to the control of MRSA both
in general and as a cause of healthcare-acquired infections. CA-MRSA is
thus a matter of serious concern and should be suppressed.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

* Corresponding author. Address: National Centre for Antimicrobials and Infection Control, Statens Serum Institut, 5 Artillerivej,
2300 Copenhagen, Denmark. Tel.: þ45 3268 8348; fax: þ45 3268 3231.
E-mail address: rsk@ssi.dk

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.07.004
CA-MRSA as a cause of HAI 365

Introduction University of Chicago Children’s Hospital with no


established HA risk factors.11 Again, the MRSA iso-
Meticillin-resistant Staphylococcus aureus (MRSA) lates differed from MRSA isolates causing nosoco-
is one of the most significant causes of health- mial infections in the same institution. However,
care-associated (HA) infections worldwide; it is as stated in the accompanying editorial, this was,
associated with significant morbidity and mortality at that time, not perceived as indicative of a gen-
and imposes a huge burden on healthcare eral change in the epidemiology of MRSA.12 Later,
resources.1,2 CA-MRSA from the same time period was reported
Meticillin resistance is encoded by the mecA as retrospective cases or case series from a number
gene which has no counterpart in meticillin- of European countries including The Netherlands
susceptible S. aureus. The origin of the mecA (1988), Denmark (1993), Finland (1997), Greece
gene remains to be established. The regulatory (1998) and France (1998).13e17
operons and mecA itself are carried on mobile However, the general change in the perception
genetic elements known as the staphylococcal that CA-MRSA actually constitutes a separate en-
chromosome cassettes mec (SCCmec).3,4 tity began with a report of the death of four
Recently, the epidemiology of MRSA infections children caused by CA-MRSA.18 Since then,
has changed significantly. Since the first description a steadily increasing number of papers have con-
of MRSA in 1961 and until the late 1990s, MRSA was firmed this significant change in the epidemiology
almost exclusively a HA infection.5 Multilocus se- of MRSA transmission and infection.
quence typing (MLST) has shown that >90% of these
MRSA infections were caused by MRSA strains that
belonged to only five genetic lineages/clonal com- Characteristics of CA-MRSA
plexes (CC), i.e. CC5, CC8, CC22, CC 30 and CC45
carrying SCCmec cassettes IeVI.6 Cases of CA-MRSA differ from the traditional HA-
Today, MRSA transmission and infections are no MRSA in several aspects, both microbiologically
longer confined to healthcare institutions but are and epidemiologically. CA-MRSA strains belong to
increasingly seen in the community, and MRSA distinct lineages and are not just feral descendants
strains are found in yet more genetic lineages. of HA-MRSA strains but seem to have evolved
Similarly new SCCmec types and subtypes are separately. Presently, five CA-MRSA lineages are
emerging rapidly and currently at least nine found worldwide: ST1-IV (USA400); ST8-IV
SCCmec types plus several subtypes have been (USA300); ST30-IV (Pacific/Oceania); ST59-IV and
described. V (USA1000, Taiwan) and ST80-IV (European).19 Re-
cently, MRSA of the CC398 lineage has emerged
worldwide. This lineage appears to have its habitat
Early reports of CA-MRSA in livestock e especially in pigs e from where
it spreads to people in close contact with MRSA-
Before this, MRSA in persons without connection to positive animals.20
healthcare institutions [community-associated CA-MRSA is characterised by possession of the
(CA)-MRSA] had been described only sporadically smaller SCC mec type IV and, to a lesser degree,
and was confined to particular groups or regions. type V or new variants. Depending on the lineage
The first description of CA-MRSA was by Savoralatz they are often less resistant to other classes of
et al. in 1982 when MRSA spread among intra- antibiotics than HA-MRSA strains.21 Furthermore,
venous drug abusers.7,8 This outbreak was followed CA-MRSA very often produce potent toxins/viru-
by secondary cases of hospital-acquired infections lence factors such as PantoneValentine leucocidin
(HAIs). In 1993, Udo et al. reported the endemic (PVL), arginine catabolic mobile element (ACME)
presence of CA-MRSA in aboriginal communities and phenol-soluble modulins.19
in Western Australia.9 This was followed by a report Epidemiologically, the type of infections, the
from New Zealand including isolates from 1988 to age distribution of patients with CA-MRSA infec-
1996.10 Common to all these reports was that the tions and the risk factors differ significantly from
causative strains, i.e. ST74-IVa in USA, WA MRSA HA-MRSA. Risk factors for CA-MRSA include: history
5 (ST8-IV) in Australia and ST30-IV in New Zealand, of colonisation/infection with CA-MRSA; close con-
were all different from the types causing health- tact (family) with a person colonised/infected
care-associated infections. with CA-MRSA; membership of an indigenous
In 1998, Herold et al. reported a marked in- community; and being a member of ‘special
crease in the number of community-acquired communities’, e.g. participation in contact sports,
MRSA infections in children admitted to the injection drug use, living in correctional facilities
366 R.L. Skov, K.S. Jensen

or shelters, military personnel, and men who have performed for only a fraction of MRSA cases, which
sex with men.21,22 Common for these ‘special com- means that the occurrence of HAI caused by CA-
munities’ is that they share the presence of one MRSA strains may be underestimated.
or more of the following ‘5 C’ risk factors: poor In other countries, HAI caused by CA-MRSA is
hygiene/Cleanliness; Compromised skin; frequent already a significant problem. In Greece, Chini
skin Contact; Contaminated surfaces and shared et al. found that 40% of HAI MRSA infections were
items; and Crowding in addition to frequent anti- caused by strains circulating in the community
biotic use and overuse.23 Furthermore, MRSA in (CC30-IVb and CC80-IV).28 In Denmark, indirect ev-
these groups tends to build up and eventually idence suggests that HAI is caused to a still higher
MRSA is likely to disseminate from these groups degree by CA-MRSA strain types since 72% of HAI
into the population in general. cases were caused by strains carrying SCCmec IV
Despite characteristic microbiological and epi- or cassette types not belonging to SCCmec types
demiological features, none of these are specific IeVI (our own unpublished data). The most striking
and there is no clear definition of CA-MRSA. This change has, however, been seen in the USA espe-
has led to the use of several different definitions cially after the emergence of USA 300 (ST8-IV).
including strain characteristics and/or epidemio- This strain was first recognised around 2000 as
logical criteria including: susceptibility to a significant cause of CA-MRSA, but within three
clindamycin/ciprofloxacin, being non-multidrug to four years it spread rapidly and became a fre-
resistant, carrying SCCmec IV or V, being PVL pos- quent cause of HAI.29e32 In fact, CA-MRSA might
itive; community onset combined with a lack of displace the traditional HA-MRSA in the future, as
HA risk factors and without previous history of be- indicated by Popovich et al. who found that the in-
ing MRSA positive.24 As mentioned previously, none cidence density rate of infections caused by strains
of the above criteria are exclusive for CA-MRSA and classified as community genotypes increased 20-
among the microbiological features it is worth not- fold from 0.05 to 0.1 in 2003e2006 compared
ing that EMRSA-15 (ST 22), which is one of the five with 2000e2003, whereas strains with hospital
large traditional HA-MRSA lineages, carries SCCmec genotypes decreased from 0.16 to 0.11.33
IV. Similarly, the characterisation of MRSA infec-
tions in persons previously found MRSA positive as
healthcare-related was very valuable in the begin- Mathematical modelling
ning of the CA-MRSA epidemic; however, it has
become increasingly problematic and, in several The consequences of changes in the epidemiology
cases, directly misleading with the increasing num- and/or the effect of different interventions can
ber of CA-MRSA cases.24,25 be investigated using mathematical models.34e37
A definition of CA-MRSA is complicated further Until very recently, most of the modelling studies
by the nature of S. aureus/MRSA carriage. The bac- (e.g. Cooper et al.34) looked at ‘the old scenario’
terium can be carried for a prolonged period of in which transmission was confined to the hospital
time without/before causing infections, this environment.
means that MRSA carriage can be acquired in the We use a slightly modified version of the de-
community, silently carried into the hospital and terministic model by Cooper et al.34 to simulate
then cause infection, which may then be classified the effect of community transmission of MRSA
as nosocomial and vice versa. (Figure 1, Table I and equations in Appendix).
The simulation model was programmed in Delphi
(Borland Delphi Professional Ver 7.0) using
CA-MRSA as a cause of HAI a fourth-order RungeeKutta method for numerical
integration of the differential equations.
CA-MRSA has mainly been associated with rela- In the model, persons can either be hospitalised
tively mild skin and soft tissue infections in other- (N1 þ z); recently discharged from hospital, i.e.
wise healthy people. An important question for the having a greater risk of transmission (N2) or belong
handling of the change in epidemiology is the to the general population (N3) either as colonised/
extent to which CA-MRSA strains can cause severe infected (y) or uncolonised (x). When MRSA car-
HAI. In UK hospitals, the situation is still dominated riers are identified in the hospital, they are moved
almost exclusively by EMRSA-15 (ST22-IV) and, to to ‘isolation’ (z) where no transmission is assumed.
a lesser degree, EMRSA-16 (ST36-II). However, The various transmission terms, bxy, are normal-
there have been reports of HAI caused by isolates ised with respect to the corresponding value of N
perceived to be CA-MRSA.26,27 Furthermore, full in order to facilitate comparison of b-values from
epidemiological investigation including typing is the three population layers.
CA-MRSA as a cause of HAI 367

Uncolonized Colonized Isolated Here b1 is unchanged (b1 ¼ 0.1115 per day), b3 is


chosen as 0.001 b1 (arbitrarily) and b2 is set to
double the community prevalence to 2% in 2 or 4
Hospital

years, respectively, for an isolation capacity of


20e50 beds.38
Finally, we examined the effect of active
screening by increasing 4 to 0.1 per day, i.e. an
average time of 10 days for MRSA-positive patients
to be identified and isolated (or a 45% reduction in
Community

non-isolated MRSA patient-days) with an isolation


N1 = x1+y1 capacity of 20, 40 and 50 beds. Effects of other in-
N2 = x2+y2 terventions have not been included. Throughout
N3 = x3+y3 the simulated time periods of intervention (cases
0r, 1ar, 1aR) the b-values were kept from the pre-
Figure 1 Flow diagram for the model with transmis- ceding period.
sion in hospital and community (adapted and modified Figure 2 shows the steeply rising prevalence of
from Cooper et al.34). MRSA after introduction of transmission in the
community at t ¼ 30 years. A new equilibrium level
is reached after 7 to 14 years, leading to a preva-
The simulations include two time periods, see lence of MRSA carriers in the hospital as high as
Table II and Figure 2: time course ‘0’ equals the 20% (case 1a; for a b2 ¼ 0.049 b1) if no interven-
period where transmission was almost exclusively tions are implemented. It further shows that by
confined to hospitals (as in the earlier model study, using active screening as the intervention
i.e. b2 ¼ b3 ¼ 0) with no active surveillance for (4 ¼ 0.1 per day), the demands to obtain control
MRSA, i.e. MRSA is only detected by clinical cul- over MRSA in the hospital varied from 5 years
tures resulting in 4 ¼ 0.02 per day.34 Hospital with 20 isolation beds (case 0r) to 11 years with
transmission is set to b1 ¼ 0.1115 per day, resulting 50 beds (case 1aR).
in a total prevalence in the community ((y2 þ y3)/ The model simulations above indicate that even
(N2 þ N3)) of 1% at equilibrium (reached after a very small b-value for community transmission
about 30 years).38 (b2 z 0.05 b1 in our case 1a) will have a major
Time segments ‘1 (a, b, c)’ denote the following impact on the overall dynamics in the hospitale
period with transmission also in the community. community system.

Table I Parameter values used in the simulations shown for the model in Figure 1
Symbol Value Description
b1 0.1115 Transmission in hospital, per day
b2 0 or adjusteda Transmission in community layer 2, per day
b3 0 or 0.001 b1 Transmission in community layer 3, per day
Nimax 20 or 50 Number of beds in isolation unit
4 0.02 or 0.1 Isolation rate, per day
p 0.25 Cleared fraction of patients leaving isolation
1/r 20 Mean length of stay in isolation, days
1/l 370 Mean duration of carriage, days
1/m 8 Mean length of hospital stay, days
n 0.0057 High readmission rate, per day
x 0.00063 Low readmission rate, per day
g 0.03 Rate of change from high to low readmission, per day
The above values give rise to the following population levels (for Ntotal ¼ 170 000):
N1 þ z z1000 Number of hospitalised patients
N2 z3500 Number of recently discharged persons
N3 z165 500 General population
Except for b1, b2, b3 and Nimax, the parameter values are copied from the model by Cooper et al.34
a
b2 was adjusted to give a doubling in community prevalence from 1% to 2% within 2 years (cases 1a and 1b in Table II) or within
4 years (case 1c).
368 R.L. Skov, K.S. Jensen

Table II Parameter values (Nimax and 4) and simulation results for a number of cases (time segments) of which
some are shown in Figure 2
Case Nimax 4 (/day) R0 Hospital Community Ttrans (years)
prevalence (%) prevalence (%)
Baseline 0 >18 0.02 1.049 4.6 1.0 z 30
Response 0 0r 20 0.10 0.662 0.0 0.0 5.0
Two-year doubling 1a 20 0.02 1.120 20.1 4.6 7.0
1b 50 0.02 1.162 15.9 4.1 14.1
Four-year doubling 1c 20 0.02 1.079 16.7 3.6 9.7
Response 1 1ar 40 0.10 0.731 14.1 3.4 11.3
1aR 50 0.10 0.731 0.0 0.0 10.5
The prevalences listed are the equilibrium values obtained when the simulated time period is sufficiently long.
Ttrans is the time for the community prevalence to reach 95% of the total change from its case initial value till the new equilibrium
value.
R0 was calculated under the assumption that isolation capacity (free beds) will always be available, which does not hold for long
periods of time in most of the simulated cases (case 0 being the exception).

The N2 population in the model consists of pa- further, not only in S. aureus, e.g. vancomycin-
tients recently discharged, including persons who resistant MRSA (VRSA), but also in other genera.
are residents in nursing homes; a sector where
transmission of MRSA is an increasing problem.39
In the simulations it is furthermore assumed that New Danish MRSA guidelines
the new MRSA strains have unchanged transmission
rates in hospitals compared with the old HA-MRSA Denmark has had a very low incidence of MRSA, i.e.
types; an increased intrahospital transmission <100 new cases per year (both infections and
rate of these strains would lead to even worse colonisations) since the 1970s. However, the num-
scenarios. ber of MRSA cases has increased steeply from 2002
to a maximum of 851 new cases in 2005.21 Some of
the increase was explained by a large outbreak of
Community-onset MRSA HA-MRSA in the six hospitals of Vejle County caused
by a ST22-IV strain, but general increase in MRSA, in
The increasing prevalence of MRSA in the commu- particular in CA-MRSA, was also seen in the rest of
nity also results in an increase in the proportion the country.21 Therefore it became apparent that
of community onset MRSA (CO-MRSA) among the policy which had successfully kept MRSA to
S. aureus bacteraemia (SAB) and other invasive a minimum in Denmark for decades simply was not
infections. In France during 2006e2007, 40% of sufficient to cope with the changing epidemiology.
invasive MRSA infections had community onset In consequence, new guidelines for MRSA were im-
and a recent large multicentre investigation in plemented in 2006 (www.sst.dk/publ/publ2008/
Australia and New Zealand reported that 18% of CFF/MRSA_veijl_en_19mar08.pdf). They reinforce
community-onset SAB were MRSA.40,41 the use of barrier precautions and single rooms for
Furthermore, to a still higher degree these all MRSA-positive hospital patients, but also include
patients do not have typical risk factors for MRSA interventions against MRSA in nursing homes and for
and that makes it difficult to predict whether persons receiving professional healthcare at home.
a given patient is at risk for having an MRSA Furthermore, all MRSA-positive persons are offered
infection. The increase in CO-MRSA combined eradication treatment as soon as they are in a condi-
with the lack of a typical patient profile may tion that makes eradication of MRSA possible, i.e.
have profound implications for the choice of no active infection and preferably no intravenous
empirical therapy for serious infections where lines or catheters. Since the implementation of
S. aureus is a possible cause, especially with the the new guidelines, the number of MRSA HAIs, espe-
increasing evidence that inappropriate initial anti- cially, has declined rapidly to a total of only eight
biotic treatment leads to higher mortality.42 The cases of MRSA bacteraemia in 2007.
increasing need for anti-MRSA coverage of empiri- In conclusion, the distinction between CA-MRSA
cal treatment will increase the overall consump- and HA-MRSA is becoming increasingly blurred and
tion of antibiotics and thereby drive resistance the typical patient at risk for carrying MRSA is
CA-MRSA as a cause of HAI 369

50 Appendix
1ar
Number in isolation

40 The applied deterministic model is described by


the following differential equations:
30 1aR dx1 b x1 y1
1a ¼ 1 þ ly1  mx1 þ vx2 þ xx3
20 dt x1 þy1
0
dy1 b1 x1 y1
10 ¼  ly1  my1 þvy2 þ xy3  Fðy1 ; zÞ
0r dt x1 þ y1
0 dz
¼ Fðy1 ; zÞ  rz
dt
20
Hospital prevalence ( )

dx2 b x2 y2
¼ 2 þ ly2 þ mx1  vx2  gx2 þ prz
1ar dt x2 þ y2
15

1a dy2 b2 x2 y2
10 ¼  ly2 þ my1  vy2  gx2 þ ð1  pÞrz
dt x2 þ y2
5 0 dx3 b x3 y3
¼ 3 þ ly3 þ gx2  xx3
0r 1aR dt x3 þ y3
0
dy3 b3 x3 y3
5 ¼  ly3 þ gy2  xy3
dt x3 þ y3
Community prevalence ( )

4 
1ar 4y1 if z < Nimax
Fðy1 ; zÞ ¼
3
minð4y1 ; rNimax Þ if z ¼ Nimax
1a
2 1aR

0 References
1
0r
1. Grundmann H, Aires-de-Sousa M, Boyce J, Tiemersma E.
0 Emergence and resurgence of meticillin-resistant Staphylo-
0 10 20 30 40 50
coccus aureus as a public-health threat. Lancet 2006;368:
Time (year)
874e885.
Figure 2 Simulation results for number of patients in 2. Boyce JM, Cookson B, Christiansen K, et al. Meticillin-resistant
isolation, hospital prevalence, and community preva- Staphylococcus aureus. Lancet Infect Dis 2005;5:653e663.
3. Katayama Y, Ito T, Hiramatsu K. A new class of genetic ele-
lence, respectively, during three scenarios: 0 þ 0r,
ment, staphylococcus cassette chromosome mec, encodes
0 þ 1a þ 1ar, and 0 þ 1a þ 1aR. (See text and Tables I
methicillin resistance in Staphylococcus aureus. Antimicrob
and II for corresponding parameter values.) Agents Chemother 2000;44:1549e1555.
4. Ito T, Katayama Y, Hiramatsu K. Cloning and nucleotide se-
difficult to define. In the UK, HA-MRSA still domi-
quence determination of the entire mec DNA of pre-methi-
nates. However, in other countries such as Greece cillin-resistant Staphylococcus aureus N315. Antimicrob
and USA and low-level countries like Denmark, CA- Agents Chemother 1999;43:1449e1458.
MRSA causes a significant portion of HAIs. Reports 5. Jevons MP. ‘‘Celbenin’’-resistant staphylococci. Br Med J
from several countries document that CA-MRSA is 1961;1:124e125.
6. Enright MC, Robinson DA, Randle G, Feil EJ, Grundmann H,
spreading from closed risk communities into the
Spratt BG. The evolutionary history of methicillin-resistant
general population. Mathematical modelling sug- Staphylococcus aureus (MRSA). Proc Natl Acad Sci U S A
gests that transmission of MRSA in the community 2002;99:7687e7692.
influences the occurrence of MRSA and thereby of 7. Saravolatz LD, Pohlod DJ, Arking LM. Community-acquired
HAI in hospitals significantly. CA-MRSA is thus a mat- methicillin-resistant Staphylococcus aureus infections:
a new source for nosocomial outbreaks. Ann Intern Med
ter of serious concern and should be suppressed.
1982;97:325e329.
8. Saravolatz LD, Markowitz N, Arking L, Pohlod D, Fisher E.
Conflict of interest statement Methicillin-resistant Staphylococcus aureus. Epidemiologic
None declared. observations during a community-acquired outbreak. Ann
Intern Med 1982;96:11e16.
9. Udo EE, Pearman JW, Grubb WB. Genetic analysis of com-
Funding sources munity isolates of methicillin-resistant Staphylococcus au-
None. reus in Western Australia. J Hosp Infect 1993;25:97e108.
370 R.L. Skov, K.S. Jensen

10. Mitchell JM, MacCulloch D, Morris AJ. MRSA in the commu- aureus: nosocomial transmission in a neonatal unit. J Hosp
nity. N Z Med J 1996;109:411. Infect 2006;64:244e250.
11. Herold BC, Immergluck LC, Maranan MC, et al. Community- 28. Chini V, Petinaki E, Meugnier H, et al. Emergence of a new
acquired methicillin-resistant Staphylococcus aureus in clone carrying PantoneValentine leukocidin genes and
children with no identified predisposing risk. J Am Med staphylococcal cassette chromosome mec type V among
Assoc 1998;279:593e598. methicillin-resistant Staphylococcus aureus in Greece.
12. Boyce JM. Are the epidemiology and microbiology of meth- Scand J Infect Dis 2008;40:368e372.
icillin-resistant Staphylococcus aureus changing? J Am Med 29. Anonymous. Methicillin-resistant Staphylococcus aureus in-
Assoc 1998;279:623e624. fections among competitive sports participants e Colorado,
13. Larsen AR, Bocher S, Stegger M, Goering R, Pallesen LV, Indiana, Pennsylvania, and Los Angeles County, 2000e2003.
Skov R. Epidemiology of European community-associated Morb Mortal Wkly Rep 2003;52:793e795.
methicillin-resistant Staphylococcus aureus clonal complex 30. Anonymous. Methicillin-resistant Staphylococcus aureus in-
80 type IV strains isolated in Denmark from 1993 to 2004. fections in correctional facilities e Georgia, California,
J Clin Microbiol 2008;46:62e68. and Texas, 2001e2003. Morb Mortal Wkly Rep 2003;52:
14. Chini V, Petinaki E, Foka A, Paratiras S, Dimitracopoulos G, 992e996.
Spiliopoulou I. Spread of Staphylococcus aureus clinical iso- 31. Klevens RM, Morrison MA, Fridkin SK, et al. Community-asso-
lates carrying PantoneValentine leukocidin genes during ciated methicillin-resistant Staphylococcus aureus and
a 3-year period in Greece. Clin Microbiol Infect 2006;12: healthcare risk factors. Emerg Infect Dis 2006;12:1991e1993.
29e34. 32. Seybold U, Kourbatova EV, Johnson JG, et al. Emergence of
15. Salmenlinna S, Lyytikainen O, Vuopio-Varkila J. Community- community-associated methicillin-resistant Staphylococcus
acquired methicillin-resistant Staphylococcus aureus, Fin- aureus USA300 genotype as a major cause of health care-
land. Emerg Infect Dis 2002;8:602e607. associated blood stream infections. Clin Infect Dis 2006;
16. Wannet WJB, Spalburg E, Heck MEOC, et al. Emergence of 42:647e656.
virulent methicillin-resistant Staphylococcus aureus strains 33. Popovich KJ, Weinstein RA, Hota B. Are community-associ-
carrying PantoneValentine leucocidin genes in The Nether- ated methicillin-resistant Staphylococcus aureus (MRSA)
lands. J Clin Microbiol 2005;43:3341e3345. strains replacing traditional nosocomial MRSA strains? Clin
17. Dufour P, Gillet Y, Bes M, et al. Community-acquired meth- Infect Dis 2008;46:787e794.
icillin-resistant Staphylococcus aureus infections in France: 34. Cooper BS, Medley GF, Stone SP, et al. Methicillin-resistant
emergence of a single clone that produces PantoneValen- Staphylococcus aureus in hospitals and the community:
tine leukocidin. Clin Infect Dis 2002;35:819e824. stealth dynamics and control catastrophes. Proc Natl Acad
18. Anonymous. Four pediatric deaths from community-acquired Sci USA 2004;101:10223e10228.
methicillin-resistant Staphylococcus aureus e Minnesota and 35. Bootsma MC, Diekmann O, Bonten MJ. Controlling methicil-
North Dakota, 1997e1999. Morb Mortal Wkly Rep 1999;48: lin-resistant Staphylococcus aureus: quantifying the effects
707e710. of interventions and rapid diagnostic testing. Proc Natl Acad
19. Diep BA, Otto M. The role of virulence determinants in com- Sci USA 2006;103:5620e5625.
munity-associated MRSA pathogenesis. Trends Microbiol 36. D’Agata EM, Webb GF, Horn MA, Moellering Jr RC, Ruan S.
2008;16:361e369. Modeling the invasion of community-acquired methicillin-
20. Wulf M, Voss A. MRSA in livestock animalsdan epidemic resistant Staphylococcus aureus into hospitals. Clin Infect
waiting to happen? Clin Microbiol Infect 2008;14:519e521. Dis 2009;48:274e284.
21. Larsen AR, Stegger M, Bocher S, Sorum M, Monnet DL, Skov RL. 37. Smith DL, Dushoff J, Perencevich EN, Harris AD, Levin SA.
Emergence and characterization of community-associated Persistent colonization and the spread of antibiotic
methicillin-resistant Staphylococcus aureus infections in resistance in nosocomial pathogens: resistance is a regional
Denmark, 1999 to 2006. J Clin Microbiol 2009;47:73e78. problem. Proc Natl Acad Sci U S A 2004;101:3709e3714.
22. Boucher HW, Corey GR. Epidemiology of methicillin-resistant 38. Gorwitz RJ, Kruszon-Moran D, McAllister SK, et al. Changes
Staphylococcus aureus. Clin Infect Dis 2008;46(Suppl. 5): in the prevalence of nasal colonization with Staphylococcus
S344eS349. aureus in the United States, 2001e2004. J Infect Dis 2008;
23. Tong SY, McDonald MI, Holt DC, Currie BJ. Global implica- 197:1226e1234.
tions of the emergence of community-associated methicil- 39. Mody L, Kauffman CA, Donabedian S, Zervos M, Bradley SF.
lin-resistant Staphylococcus aureus in indigenous Epidemiology of Staphylococcus aureus colonization in nurs-
populations. Clin Infect Dis 2008;46:1871e1878. ing home residents. Clin Infect Dis 2008;46:1368e1373.
24. David MZ, Glikman D, Crawford SE, et al. What is commu- 40. Turnidge JD, Nimmo GR, Pearson J, Gottlieb T, Collignon PJ.
nity-associated methicillin-resistant Staphylococcus au- Epidemiology and outcomes for Staphylococcus aureus bac-
reus? J Infect Dis 2008;197:1235e1243. teraemia in Australian hospitals, 2005e06: report from the
25. Skov R, Gudlaugsson O, Hardardottir H, et al. Proposal for Australian Group on Antimicrobial Resistance. Commun Dis
common Nordic epidemiological terms and definitions for Intell 2007;31:398e403.
methicillin-resistant Staphylococcus aureus (MRSA). Scand 41. Dauwalder O, Lina G, Durand G, et al. Epidemiology of inva-
J Infect Dis 2008;40:495e502. sive methicillin-resistant Staphylococcus aureus clones
26. Otter JA, French GL. The emergence of community-associ- collected in France in 2006 and 2007. J Clin Microbiol
ated methicillin-resistant Staphylococcus aureus at a Lon- 2008;46:3454e3458.
don teaching hospital, 2000e2006. Clin Microbiol Infect 42. Schramm GE, Johnson JA, Doherty JA, Micek ST, Kollef MH.
2008;14:670e676. Methicillin-resistant Staphylococcus aureus sterile-site in-
27. David MD, Kearns AM, Gossain S, Ganner M, Holmes A. Com- fection: the importance of appropriate initial antimicrobial
munity-associated meticillin-resistant Staphylococcus treatment. Crit Care Med 2006;34:2069e2074.
Journal of Hospital Infection (2009) 73, 371e377
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Screening and isolation for infection control


E. Tacconelli*

Department of Infectious Diseases, Università Cattolica Sacro Cuore, Rome, Italy

Available online 21 August 2009

KEYWORDS Summary Control measures aimed to reduce the prevalence of health-


Healthcare-associated care-associated infections include active surveillance cultures (ASCs), con-
infection; tact isolation of patients colonised with epidemiologically significant
Isolation;
pathogens, and pre-emptive isolation of high risk patients. However, the
Meticillin-resistant
Staphylococcus
benefits of these measures are questionable. A systematic review of isola-
aureus; tion policies demonstrated that intensive concerted interventions includ-
Screening; ing isolation can substantially reduce nosocomial meticillin-resistant
Vancomycin-resistant Staphylococcus aureus (MRSA) infection. Monitoring of interventions is fun-
enterococcus damental. Surveillance data should be presented and fed back appropria-
tely. International guidelines suggest that only intensive care units
should apply extensive ASCs. However, legislation for mandatory screening
at hospital admission has been advocated in many countries. Targeted
screening could be used to limit the potential for dissemination of anti-
biotic-resistant pathogens from otherwise unsuspected carriers from the
start of patients’ hospitalisation, as opposed to other strategies, in which
screening programmes target patients already hospitalised. Although the
influx of antibiotic-resistant pathogens into the hospital would not change,
early detection would reduce the time colonised patients might have to
disseminate pathogens. Recently, rapid methods for molecular detection
of MRSA have been developed. Data on the impact of these tests on
the MRSA acquisition rate are extremely heterogeneous. Published studies
differ according to the settings in which they have been evaluated, the
choice of patient population to be screened, other infection control measures
employed and, most importantly, study design and baseline prevalence
of MRSA. Based on these studies, definitive recommendations cannot be
made.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

* Address: Largo F. Vito 8, 00168, Rome, Italy. Tel.: þ39 06 30155527; fax: þ39 06 3054519.
E-mail address: etacconelli@rm.unicatt.it

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.05.002
372 E. Tacconelli

Introduction osteomyelitis, or septic arthritis. A systematic


review including ten studies and 1170 patients
Efforts to control healthcare-associated infections estimated the risk of infection following MRSA
(HCAIs) have, with few exceptions, failed, result- colonisation.7 The analysis showed a four-fold in-
ing in growing political and public alarm. Increased crease (95% confidence interval: 2e7) in the risk
morbidity and mortality due to antibiotic-resistant of infection after MRSA colonisation compared
bacteria acquired during hospital stay have also with meticillin-susceptible S. aureus (MSSA) colo-
been reported worldwide.1,2 The European Centre nisation. Significant heterogeneity was detected
for Disease Prevention and Control has estimated between studies. The majority of reports differed
that HCAIs affect about one in 20 hospital pa- in the choice of patient population, severity of
tients.3 In January 2009 the European Commission illness, and frequency of sampling employed to
published a Council Recommendation on patient detect colonisation, further suggesting the need for
safety, including the prevention and control of new research with appropriate statistical analysis.
HCAIs.4 The Commission advised that a national Olivier et al. studied the risk of VRE bacteraemia
strategy, complementary to strategies targeted among patients previously colonised with the bac-
towards the prudent use of antimicrobial agents, teria. Four percent of colonised patients devel-
should be developed incorporating prevention oped bacteraemia caused by the same strain, as
and control of HCAIs into national public health confirmed by the genotypic analysis.8
objectives and aiming to reduce the risk of such Important clues in deciding whether screening
infections within hospitals. Hence, the question and contact isolation are needed are the pro-
arises: how are public health professionals to portions of antibiotic resistance attributable to
decide what is the most cost-effective approach hospital antibiotic use and cross-transmission in
to preventing HCAIs? Unfortunately, a satisfactory hospitalised patients. In a multicentre prospective
answer from evidence-based data cannot be Italian study, we determined the incidence of new
provided. Up to now, most studies have focused on colonisation due to MRSA, VRE and ciprofloxacin-
meticillin-resistant Staphylococcus aureus (MRSA) resistant Pseudomonas aeruginosa per 1000 days of
and vancomycin-resistant enterococci (VRE) and antibiotic therapy.9 The highest risk was associ-
to a much lesser extent on multidrug-resistant ated with carbapenem use, with eight new cases
(MDR) Gram-negative bacteria. The control mea- of MRSA colonisation per 1000 days of therapy.
sures aimed to reduce the spread of HCAIs include: An accurate estimate of the attributable fraction
education of healthcare workers (HCWs) with im- of antibiotic resistance due to cross-transmission
plementation and observation of hand-washing is not available. Many factors have contributed to
practices; active surveillance cultures (ASCs); con- this lack of information. For example, Buke et al.
tact isolation of patients colonised with epidemio- demonstrated significant epidemiological differ-
logically important bacteria (such as MRSA and ences in the variables associated with colonisation
VRE); pre-emptive isolation of high risk patients; with MRSA and extended spectrum b-lactamase-
and restriction of antibiotic use. Numerous studies producing (ESBL) Gram-negative bacteria.10
have demonstrated that the best way to face the Another parameter useful for a better under-
problem is a multidisciplinary approach. Marshall standing of the attributable fraction due to
et al. summarised in four steps the actions needed cross-transmission is colonisation pressure, i.e.
to reduce the prevalence of HCAIs: (1) antibiotic the prevalence of resistance in the surrounding
stewardship; (2) reduction in the number of environment. Williams et al. demonstrated that
colonised patients; (3) prevention of infection in ongoing monitoring of colonisation pressure pro-
colonised patients; and (4) prevention of cross- vides the opportunity for early implementation
transmission between hospitalised patients.5 of enhanced infection prevention and control
Among the strategies aimed to reduce patient-to- practices, and can potentially decrease nosocomial
patient transmission, the role of screening and transmission of MRSA and prevent outbreaks.11
isolation is still under debate.
The first assumption underlying a screening
strategy is that colonisation increases the risk of Should mandatory screening for MRSA
infection. Huang et al. observed that MRSA infec- and VRE be performed?
tions developed in 29% of 209 colonised patients
in an 18 month follow-up period.6 Twenty-eight ‘Who’ should be screened for epidemiologically
percent of infections involved bacteraemia and important pathogens and ‘where’ and ‘when’ are
56% involved pneumonia, soft tissue infection, still open questions. In studies of infection control
Screening and isolation for HCAI prevention 373

measures, screening is never the primary interven- period in a 700-bed hospital with 23 000 admis-
tion and it is usually included in a multifaceted sions annually in Germany.17 The screening pro-
approach. The majority of studies relate to hospital- gramme was able to prevent 48% of predicted
acquired MRSA (HA-MRSA). A recent systematic nosocomial MRSA, saving a predicted V200,782
review of the literature performed by McGinigle with a net saving of V110,000 annually. The
et al. summarised the evidence of the efficacy of screening programme became cost-effective at
ASCs in the intensive care unit (ICU) setting on sev- a low MRSA incidence rate. The major limit of
eral clinical and economic outcomes.12 Although the study was that the authors did not use real
most of observation-based studies reported a signifi- costs calculation but instead hospital reimburse-
cant reduction in MRSA infection after the applica- ment as a surrogate parameter.
tion of ASCs, the evidence was of poor quality and For VRE control, it has been estimated that ASCs
could not allow definitive recommendations. The would be cost-beneficial for hospital units where
role of ASCs might be different according to the base- the number of patients with VRE bacteraemia is at
line prevalence of MRSA and the hospital setting. In least 6e9 patients per year or if the savings from
three intensive care units (ICUs) in France the acqui- fewer VRE bacteraemic patients in combination
sition of MRSA was reduced from 7% to 3% through with decreased antimicrobial use ranged from
multiple interventions, including ASC, contact pre- US$100,000 to 150,000 per year.18
cautions and use of alcohol hand rub.13 All patients Targeted surveillance based on patients’ risk
were screened for nasal carriage on hospital admis- factors in medical or surgical wards might be
sion and weekly thereafter and placed under contact preferred as the most effective use of hospital
precautions if screen positive. Similar results were resources. Risk factors for HCAIs due to MRSA at
obtained in a 18 month study in Colorado, USA.14 hospital admissions were analysed in 127 patients
All patients admitted to ICUs and medical wards with bacteraemia in an endemic setting in Boston,
were screened at admission and weekly thereafter. USA.19 Two logistic regression models were gener-
The overall rate of HA-MRSA infections compared ated. In the first model, previous MRSA infection or
with historical controls decreased significantly, colonisation, cellulitis, presence of a central ve-
although an increased number of community-ac- nous catheter (CVC), and skin ulcers were indepen-
quired MRSA skin infections was admitted to the dently associated with MRSA bacteraemia. Since
wards. The majority occurred in surgery ICU (SICU) prior MRSA colonisation or infection status may
because the medical ICU (MICU) had a higher not be known at the time of hospitalisation, a sec-
incidence of colonisation and lower percentage of ond model was generated which excluded prior
compliance with hand washing. Physicians in SICU history of MRSA colonisation or infection. This
were also less likely to enter isolation rooms. model identified the presence of a CVC, hospital-
Some countries have maintained low endemic isation(s) within the previous six months, use of
levels of MRSA by implementing nationwide control quinolones within the previous 30 days, and dia-
measures targeting MRSA such as the ‘search betes mellitus as independent risk factors for
and destroy’ (S&D) strategy.15 Measures of S&D MRSA bacteraemia. Both models had high sensitiv-
include: contact isolation for MRSA-positive ity and specificity. Harbarth et al. defined risk fac-
patients; pre-emptive isolation and screening for tors for unknown carriage of MRSA at hospital
high risk patients; screening of patients and admission in a university hospital in Geneva.20
personnel following an unexpected case of MRSA; Male sex, age >75 years, previous exposure to qui-
screening of HCWs with furlough of those found nolones, cephalosporins and carbapenems, previ-
to be carriers until decontamination is achieved; ous hospitalisation, intrahospital transfer and
closure of wards to new admissions if there urinary catheterisation were associated with
is more than one carrier among hospitalised MRSA colonisation at hospital admission.
patients. Interestingly, Bootsma et al. generated A prediction score for VRE identification was
a stochastic three-hospital model and an analytical derived and validated in a high-endemic setting in
one-hospital model to quantify the effectiveness the USA.21 A risk index was derived by using six in-
of different infection control measures and to dependent risk factors associated with VRE recov-
predict the effect of rapid diagnostic testing on ery within 48 h of hospital admission: previous
isolation needs.16 The analysis showed that the isolation of MRSA (4 points), whether the patient
application of the S&D strategy in a hospital with was receiving long-term haemodialysis (3), trans-
high endemicity would reduce the prevalence to fer from a long-term care facility (3), antibiotic ex-
<1% within 6 years. posure (3), prior hospitalisation (3), and age >60
The costs of a hospital-wide selective screen- years (2). On the basis of a point score 10, the
ing programme were analysed for a 19 month sensitivity, specificity, and positive and negative
374 E. Tacconelli

predictive values of this prediction rule were 44%, March 2009 and for all admissions by March 2011.
98%, 81%, and 90%, respectively. This extended the guidance issued as part of the
For several years conventional culture methods, ‘Saving Lives’ package in September 2006, which
which can take 48e72 h to obtain a result, have recommended a risk-based approach to screening
been considered the gold standard to detect to include high-risk surgical patients, renal dialysis
MRSA colonisation. In the absence of pre-emptive patients, those admitted to ICUs, high risk medical
room isolation this time might be enough to spread admissions, and transfers from other hospitals,
the bacterium among patients. Recently, rapid long-term care facilities, and nursing homes. In
methods for molecular detection of MRSA-colon- response to new legislation, the Society for
ised patients with available results in 2 h have Healthcare Epidemiology of America (SHEA) and
been developed with high sensitivity and specifi- the Association for Professionals in Infection
city.22 Major limits for the wide application of mo- Control and Epidemiology Inc. (APIC) developed
lecular diagnosis for MRSA are related to the high a position statement asserting that, although
costs and varying benefits in different studies. conspicuous evidence to support the use of ASCs
Nine studies (eight cohort studies and one clus- for high risk patients and during outbreaks is
ter-randomised, cross-over trial) analysed the effi- available, there is not sufficient evidence to justify
cacy of screening for MRSA using molecular tests the mandatory use of this control measure.32 The
compared with no screening or screening with con- lack of support for legislation is due to the uncer-
ventional cultures.23e30 Studies were performed in tainties and the potential unintended con-
different settings: ICUs (two studies), medical sequences of legislation mandating application of
and/or surgery wards (four) and throughout the ASCs, namely: exclusion of local infection control
hospital (two). Four papers reported a significant professionals from their role of leading risk assess-
decrease in the rate of HA-MRSA, two observed ment and resource allocation; unresolved contro-
no effects, and one showed uncertain results. versies regarding the epidemiological, biological
Variability in the efficacy of polymerase chain and clinical implications of ASCs; and the poten-
reaction (PCR) might be partially explained by tially negative effects on patients of contact
different comparisons applied in the studies. The isolation.
main limitations other than the study design
were systematic screening not performed at
Should contact isolation precautions be
discharge, PCR result not confirmed by conven-
tional culture, lack of analysis of possible variation implemented for patients colonised or
in MRSA epidemiology during the study period, infected with epidemiologically
absence of monitoring of compliance with decolo- important bacteria?
nisation treatment or contact precautions. Based
on these studies, definitive recommendations Sixty-five percent of HCWs contaminate their
cannot be made. uniforms or gowns during routine care of patients
A rapid real-time PCR test that detects the with MRSA. On >25% of occasions HCW’s clean
presence of vanA and/or vanB genes has been hands became recontaminated after contact with
proposed for rapid screening to identify patients their contaminated clothing. Cooper et al. demon-
harbouring VRE at hospital admission.31 For 502 strated that intensive concerted interventions,
rectal swabs and stool specimens, sensitivity and including isolation, can substantially reduce MRSA.33
specificity was 98% and 87%, and 95% and 87%, re- A before-and-after study evaluated the efficacy of
spectively. When the test is used in settings with contact and droplet precautions in reducing the
high prevalence of vanA isolates, results would incidence of HA-MRSA infections.34 After a baseline
not require conventional culture confirmation. On period, contact and droplet precautions were
the contrary, the high number of false positives implemented in all ICUs. Reductions in the inci-
for vanB would require culture confirmation. dence of HA-MRSA infection in ICUs also led to the
In the USA, several large organisations such as implementation of contact precautions in non-ICU
the Veterans Affairs and Evanston Northwestern patient care areas. Combined rates of HA-MRSA
University and a few states such as Illinois, New infection in the MICU and SICU decreased with no
Jersey and Pennsylvania introduced mandatory significant change in other ICUs. After the discon-
screening for both VRE and MRSA. In the UK it tinuation of droplet precautions, the combined
was announced in October 2007 that all patients rate in the MICU and SICU decreased further
admitted to National Health Service hospitals although not significantly. After the implementation
would be screened for carriage of MRSA. Screening of contact precautions the rate decreased signifi-
was to be in place for all elective admissions by cantly in non-ICU areas.
Screening and isolation for HCAI prevention 375

Although >100 studies have reported control of suggestions made by other studies that patients
MRSA with screening linked to isolation and cohort- in isolation receive less frequent care and may
ing, two studies suggested that reporting culture even suffer psychologically.
results and isolating colonised patients had no
impact on the prevalence of HA-MRSA.35 The first
study assessed the effect of daily microbiological Decolonisation
surveillance alone on the spread of S. aureus.36
During a 10 week period, surveillance cultures Following positive screening, a patient should be
were performed in 158 patients. Surveillance cul- placed in contact precaution and decolonised. The
tures and genotyping of MRSA and MSSA isolates majority of protocols for MRSA decolonisation in-
demonstrated the absence of cross-transmission clude mupirocin alone or in combination with body
among patients. The second study was a prospec- washes, and systemic therapy with one or two
tive one-year study in the ICUs of two teaching antibiotics such as rifampicin, doxycycline and tri-
hospitals. Admission and weekly screens were methoprim-sulfamethoxazole. Less popular proto-
used to ascertain the incidence of MRSA colonisa- cols include the tea tree oil, a popular natural
tion.37 In the middle six months, MRSA-positive antiseptic, topical gentian violet and arbekacin
patients were neither moved to a single room nor inhalation in patients with percutaneous endo-
cohort-nursed unless they were carrying other scopic gastrostomy. The duration of the decolonisa-
multiresistant bacteria. Patients’ characteristics tion treatment is usually 7e10 days. Decolonisation
and MRSA acquisition rates were similar in the has been proven to be efficacious in reducing
periods when patients were moved and not moved. infections in high risk groups.42 In dialysis patients
The crude (unadjusted) Cox proportional hazards the use of mupirocin reduced the rate of S. aureus
model showed no evidence of increased transmis- infections by 68%.43 Risk reduction was higher in
sion during the non-move phase. However, the peritoneal dialysis patients. Significant reduction
studies have many limitations. First, the time of S. aureus infections was also observed in patients
elapsed from admission to the results of cultures after cardiothoracic and orthopaedic surgery.42
was up to four days, and patients with a length Different protocols were used for VRE decoloni-
of hospitalisation <48 h were excluded. This time sation with poor response. After one month >50%
would have been sufficient, especially in highly en- of the patients are recolonised.44 The most prom-
demic situations, to allow person-to-person trans- ising agent for gastrointestinal decolonisation was
mission of MRSA. Second, the low number of ramoplanin, the first of a new class of antibiotics,
carers and patients might have had an important the glycolipodepsipeptides. The drug is bacteri-
role on the final outcome. cidal against Gram-positive bacteria and is not ab-
For VRE the role of contact precautions is still sorbed when taken orally and thus achieves high
under evaluation. Our group is currently reviewing faecal concentrations. In animal studies ramopla-
the available evidence within the Cochrane Wound nin inhibits VRE colonisation due to re-expansion
Group.38 Many reports showed that intensive infec- of the colonic microflora. A randomised placebo-
tion control strategies including isolation enabled controlled double-blind trial showed that VRE
the timely termination of VRE outbreaks, even decolonisation was unsuccessful in 79% of the
those involving VRE strains with high epidemic po- patients.44
tential on high risk wards. Premature discontinua- Screening and decolonisation of HCWs has been
tion of infection control measures caused attempted for MRSA but its use remains contro-
recurrence of VRE transmission.39 versial.45 The prevalence of MRSA among profes-
A combination of control measures including sionals is related to geographical regions (Eastern
ASCs, contact precautions for all colonised and Europe 1.6%, Africa 15.5%), location (ICU 4.7%,
infected patients and antimicrobial stewardship general wards 6.3%), type of HCW (nurse 8%, med-
has also been reported to significantly reduce the ical 7.4%), type of room (private cohorting 2.4%, no
incidence of ESBL-producing bacteria.40 private/no cohorting 7.7%), and contact pre-
Contact precautions have been associated with cautions in force (3.3%) or not applied (5.6%).46
several adverse effects. Their application should However, little evidence suggests that the
be preceded by accurate training of HCWs and it exclusion of MRSA-positive HCWs improves the
should not be accepted as a substitute for hand control of HA-MRSA with the exception of hospital
washing. In the MICU at Duke University Medical outbreaks. Screening might have psychological
Center it was observed that HCWs were approxi- implications for professionals who test positive.
mately half as likely to enter the rooms of patients Cost-effectiveness analysis for this approach in
in contact isolation.41 The results supported this population is also unavailable. Only staff
376 E. Tacconelli

members with colonised or infected hand lesions 7. Safdar N, Bradley EA. The risk of infection after nasal colo-
should be off work while receiving courses of nisation with Staphylococcus aureus. Am J Med 2008;12:
310e315.
clearance therapy.47 8. Olivier CN, Blake RK, Steed LL, Salgado CD. Risk of vanco-
mycin-resistant enterococcus (VRE) bloodstream infection
Discussion among patients colonized with VRE. Infect Control Hosp
Epidemiol 2008;29:404e409.
9. Tacconelli E, De Angelis G, Cataldo MA, et al. Usage of car-
Patients and the public increasingly view rates of bapenems significantly increases the rate of new colonisa-
HCAIs as indicators of quality of patient care. tion due to antibiotic-resistant bacteria in hospitalised
There has been much debate about the evidence patients. Barcelona: ECCMID; 2008. Oral communication.
10. Buke C, Armand-Lefevre L, Lolom I, et al. Epidemiology of
and cost-effectiveness of various infection control
multidrug-resistant bacteria in patients with long hospital
policies. A multidisciplinary approach should be stays. Infect Control Hosp Epidemiol 2007;28:1255e1260.
employed in all settings including: hand hygiene 11. Williams VR, Callery S, Vearncombe M, Simor AE. The role of
programmes, ASCs, training and feedback to colonization pressure in nosocomial transmission of methi-
HCWs, bundles for ventilator-associated pneumo- cillin-resistant Staphylococcus aureus. Am J Infect Control
2009;37:106e110.
nia and CVC-related infections and environmental
12. McGinigle KL, Gourlay ML, Buchanan IB. The use of active
programmes. Target pathogens should be locally surveillance cultures in adult intensive care units to reduce
defined. Programmes focused solely on MRSA might methicillin-resistant Staphylococcus aureus-related mor-
not be the appropriate answer to impact on HCAIs. bidity, mortality, and costs: a systematic review. Clin Infect
Intensive promotion of alcohol-based hand rubs Dis 2008;46:1717e1725.
13. Lucet JC, Paoletti X, Lolom I, et al. Successful long-term
and behavioural change interventions might rep-
program for controlling methicillin-resistant Staphylococcus
resent a more cost-effective approach compared aureus in intensive care units. Intensive Care Med 2005;31:
with universal screening policies. On the grounds 1051e1057.
of lack of evidence-based clinical and cost-effec- 14. Clancy M, Graepler A, Wilson M, Douglas I, Johnson J,
tiveness data, routine screening of all admissions Price CS. Active screening in high-risk units is an effective
and cost-avoidant method to reduce the rate of methicil-
to hospital is not advocated at this time.48e51
lin-resistant Staphylococcus aureus infection in the hospi-
Rapid molecular tests for MRSA screening are real- tal. Infect Control Hosp Epidemiol 2006;27:1009e1017.
istic only when combined with adequate infection 15. Vos MC, Ott A, Verbrugh HA. Successful search-and-destroy
control measures in high risk patients. policy for methicillin-resistant Staphylococcus aureus in
The Netherlands. J Clin Microbiol 2005;43:2034.
16. Bootsma MC, Diekmann O, Bonten MJ. Controlling methicil-
lin-resistant Staphylococcus aureus: quantifying the effects
Conflict of interest statement of interventions and rapid diagnostic testing. Proc Natl Acad
None. Sci USA 2006;103:5620e5625.
17. Wernitz MH, Keck S, Swidsinski S, Schulz S, Veit SK. Cost
analysis of a hospital-wide selective screening programme
Funding sources for methicillin-resistant Staphylococcus aureus (MRSA)
None. carriers in the context of diagnosis related groups (DRG)
payment. Clin Microbiol Infect 2005;11:466e471.
18. Montecalvo MA, Jarvis WR, Uman J, et al. Costs and savings
References associated with infection control measures that reduced
transmission of vancomycin-resistant enterococci in an
1. Boucher HW, Corey GR. Epidemiology of methicillin- endemic setting. Infect Control Hosp Epidemiol 2001;22:
resistant Staphylococcus aureus. Clin Infect Dis 2008;46: 437e442.
S344eS349. 19. Tacconelli E, Venkataraman L, De Girolami PC, D’Agata EM.
2. Harris AD, McGregor JC, Furuno JP. What infection control Methicillin-resistant Staphylococcus aureus bacteraemia
interventions should be undertaken to control multidrug- diagnosed at hospital admission: distinguishing between
resistant Gram-negative bacteria? Clin Infect Dis 2006; community-acquired versus healthcare-associated strains.
43(Suppl. 2):S57eS61. J Antimicrob Chemother 2004;53:474e479.
3. Anonymous. Healthcare-associated infections. European 20. Harbarth S, Sax H, Fankhauser-Rodriguez C, Schrenzel J,
Centre for Disease Prevention and Control. Available from: Agostinho A, Pittet D. Evaluating the probability of previ-
http://ecdc.gov; 2006. ously unknown carriage of MRSA at hospital admission. Am
4. Proposal for a Council Recommendation on patient safety, J Med 2006;119:275. e15e23.
including the prevention and control of healthcare associ- 21. Tacconelli E, Karchmer AW, Yokoe D, D’Agata EM. Prevent-
ated infections {COM(2008) 836 final} {SEC(2008) 3004} ing the influx of vancomycin-resistant enterococci into
{SEC(2008) 3005}. EC Brussels; 20 January 2009. health care institutions, by use of a simple validated predic-
5. Marshall C, Wesselingh S, McDonald M, Spelman D. Control tion rule. Clin Infect Dis 2004;39:964e970.
of endemic MRSA e what is the evidence? A personal view. 22. Carroll MC. Rapid diagnostics for methicillin-resistant
J Hosp Infect 2004;56:253e268. Staphylococcus aureus: current status. Mol Diagn Ther
6. Huang SS, Platt R. Risk of methicillin-resistant Staphylococ- 2008;12:15e24.
cus aureus infection after previous infection or coloniza- 23. Conterno LO, Shymanski J, Ramotar K, et al. Real-time
tion. Clin Infect Dis 2003;36:281e285. polymerase chain reaction detection of methicillin-
Screening and isolation for HCAI prevention 377

resistant Staphylococcus aureus: impact on nosocomial 37. Farr BM. What to think if the results of the National Insti-
transmission and costs. Infect Control Hosp Epidemiol tutes of Health randomized trial of methicillin-resistant
2007;28:1134e1141. Staphylococcus aureus and vancomycin-resistant entero-
24. Cunningham R, Jenks P, Northwood J, Wallis M, Ferguson S, coccus control measures are negative (and other advice to
Hunt S. Effect on MRSA transmission of rapid PCR testing of young epidemiologists): a review and an au revoir. Infect
patients admitted to critical care. J Hosp Infect 2007;65: Control Hosp Epidemiol 2006;27:1096e1106.
24e28. 38. Tacconelli E, Cauda R, Cataldo MAA, Carmeli Y, De
25. Harbarth S, Masuet-Aumatell C, Schrenzel J, et al. Evalua- Angelis G. Control interventions for preventing spread
tion of rapid screening and pre-emptive contact isolation of vancomycin-resistant enterococci (VRE) in hospitals.
for detecting and controlling methicillin-resistant Staphylo- Cochrane Database Syst Rev 2008;(4).
coccus aureus in critical care: an interventional cohort 39. Schmidt-Hieber M, Blau IW, Schwartz S, et al. Intensified
study. Crit Care 2006;10:R25. strategies to control vancomycin-resistant enterococci in
26. Jeyaratnam D, Whitty CJ, Phillips K, et al. Impact of rapid immunocompromised patients. Int J Hematol 2007;86:
screening tests on acquisition of meticillin resistant Staph- 158e162.
ylococcus aureus: cluster randomised crossover trial. Br 40. Conterno LO, Shymanski J, Ramotar K, Toye B, Zvonar R,
Med J 2008;336:927e930. Roth V. Impact and cost of infection control measures to re-
27. Jog S, Cunningham R, Cooper S, et al. Impact of reoperative duce nosocomial transmission of extended-spectrum beta-
screening for meticillin-resistant Staphylococcus aureus by lactamase-producing organisms in a non-outbreak setting.
real-time polymerase chain reaction in patients undergoing J Hosp Infect 2007;65:354e360.
cardiac surgery. J Hosp Infect 2008;69:124e130. 41. Kirkland KB, Weinstein JM. Adverse effects of contact isola-
28. Keshtgar MR, Khalili A, Coen PG, et al. Impact of rapid mo- tion. Lancet 1999;354:1177e1178.
lecular screening for meticillin-resistant Staphylococcus au- 42. Kallen AJ, Wilson CT, Larson RJ. Perioperative intranasal
reus in surgical wards. Br J Surg 2008;95:381e386. mupirocin for the prevention of surgical-site infections:
29. Robicsek A, Beaumont JL, Paule SM, et al. Universal surveil- systematic review of the literature and meta-analysis.
lance for methicillin-resistant Staphylococcus aureus in 3 Infect Control Hosp Epidemiol 2005;26:916e922.
affiliated hospitals. Ann Intern Med 2008;148:409e418. 43. Tacconelli E, Carmeli Y, Aizer A, Ferreira G, Foreman MG,
30. Harbarth S, Fankhauser C, Schrenzel J, et al. Universal D’Agata EM. Mupirocin prophylaxis to prevent Staphylococ-
screening for methicillin-resistant Staphylococcus aureus cus aureus infection in patients undergoing dialysis: a meta-
at hospital admission and nosocomial infection in surgical analysis. Clin Infect Dis 2003;37:1629e1638.
patients. J Am Med Assoc 2008;299:11. 44. Wong MT, Kauffman CA, Standiford HC, et al. Ramoplanin
31. Stamper PD, Cai M, Lema C, Eskey K, Carroll KC. Comparison VRE2 Clinical Study Group. Effective suppression of vanco-
of the BD GeneOhm VanR assay to culture for identification mycin-resistant Enterococcus species in asymptomatic gas-
of vancomycin-resistant enterococci in rectal and stool trointestinal carriers by a novel glycolipodepsipeptide,
specimens. J Clin Microbiol 2007;45:3360e3365. ramoplanin. Clin Infect Dis 2001;33:1476e1482.
32. Weber SG, Huang SS, Oriola S, et al. Legislative mandates for 45. Simpson AH, Dave J, Cookson B. The value of routine
use of active surveillance cultures to screen for meti- screening of staff for MRSA. J Bone Joint Surg Br 2007;89:
cillin-resistant Staphylococcus aureus and vancomycin- 565e566.
resistant enterococci: position statement from the Joint SHEA 46. Albrich WC, Harbarth S. Health-care workers: source, vec-
and APIC Task Force. Am J Infect Control 2007;35:73e85. tor, or victim of MRSA? Lancet Infect Dis 2008;8:289e301.
33. Cooper BS, Stone SP, Kibbler CC, et al. Isolation measures in 47. Nathwani D, Morgan M, Masterton RG, et al. British Society
the hospital management of methicillin resistant Staphylo- for Antimicrobial Chemotherapy Working Party on commu-
coccus aureus (MRSA): systematic review of the literature. nity-onset MRSA Infections. Guidelines for UK practice for
Br Med J 2004;329:533e538. the diagnosis and management of meticillin-resistant
34. Mangini E, Segal-Maurer S, Burns J, et al. Impact of contact Staphylococcus aureus (MRSA) infections presenting in the
and droplet precautions on the incidence of hospital- community. J Antimicrob Chemother 2008;61:976e994.
acquired methicillin-resistant Staphylococcus aureus infec- 48. Diekema DJ, Climo M. Preventing MRSA infections: finding it
tion. Infect Control Hosp Epidemiol 2007;28:1261e1266. is not enough. J Am Med Assoc 2008;299:1190e1192.
35. Cepeda JA, Whitehouse T, Cooper B, et al. Isolation of pa- 49. Gould IM. Control of methicillin-resistant Staphylococcus
tients in single rooms or cohorts to reduce spread of MRSA aureus in the UK. Eur J Clin Microbiol Infect Dis 2005;24:
in intensive-care units: prospective two-centre study. Lan- 789e793.
cet 2005;365:295e304. 50. Dancer SJ. Considering the introduction of universal MRSA
36. Nijssen S, Bonten MJ, Weinstein RA. Are active microbiolog- screening. J Hosp Infect 2008;69:315e320.
ical surveillance and subsequent isolation needed to pre- 51. Tacconelli E. Methicillin-resistant Staphylococcus aureus:
vent the spread of methicillin-resistant Staphylococcus risk assessment and infection control policies. Clin
aureus? Clin Infect Dis 2005;40:405e409. Microbiol Infect 2008;14:407e410.
Journal of Hospital Infection (2009) 73, 378e385
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

The role of environmental cleaning in the control


of hospital-acquired infection
S.J. Dancer*

Department of Microbiology, Hairmyres Hospital, East Kilbride, UK

Available online 1 September 2009

KEYWORDS Summary Increasing numbers of hospital-acquired infections have gener-


Acinetobacter; ated much attention over the last decade. The public has linked the so-
Environmental called ‘superbugs’ with their experience of dirty hospitals but the precise
cleaning;
role of environmental cleaning in the control of these organisms remains
C. difficile;
unknown. Until cleaning becomes an evidence-based science, with
Hospital-associated
infection; established methods for assessment, the importance of a clean environment
Infection control; is likely to remain speculative. This review will examine the links between
MRSA; the hospital environment and various pathogens, including meticillin-resis-
Norovirus; tant Staphylococcus aureus, vancomycin-resistant enterococci, norovirus,
VRE Clostridium difficile and acinetobacter. These organisms may be able to sur-
vive in healthcare environments but there is evidence to support their vulner-
ability to the cleaning process. Removal with, or without, disinfectants,
appears to be associated with reduced infection rates for patients. Unfortu-
nately, cleaning is often delivered as part of an overall infection control pack-
age in response to an outbreak and the importance of cleaning as a single
intervention remains controversial. Recent work has shown that hand-touch
sites are habitually contaminated by hospital pathogens, which are then de-
livered to patients on hands. It is possible that prioritising the cleaning of
these sites might offer a useful adjunct to the current preoccupation with
hand hygiene, since hand-touch sites comprise the less well-studied side of
the hand-touch site equation. In addition, using proposed standards for
hospital hygiene could provide further evidence that cleaning is a cost-effec-
tive intervention for controlling hospital-acquired infection.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

Introduction
* Address: Department of Microbiology, Hairmyres Hospital,
Eaglesham Road, East Kilbride G75 8RG, UK. Tel.: þ44 1355
585000; fax: þ44 1355 584350. There has been much debate over hospital clean-
E-mail address: stephanie.dancer@lanarkshire.scot.nhs.uk liness and increasing numbers of hospital-acquired
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.03.030
Environmental cleaning and HAI 379

infections (HAIs). The public have linked their infection control intervention for patients. There
visual experience of dirty hospitals with the risk will also be some discussion on the measurement
of HAI but there is little evidence to support this at of ‘cleanliness’ of the healthcare environment
present.1 Cleaning has never been regarded as an and why this is important for future work evaluat-
evidence-based science and consequently receives ing the role of hospital cleaning and HAI.
little attention from the scientific community.
Since there are no scientific standards to measure
the effect of an individual cleaner, or assess en- Meticillin-resistant Staphylococcus
vironmental cleanliness, finding the evidence for aureus
benefit in the control of infection is further ham-
pered.2 There are always basic aesthetic consider- MRSA resists desiccation and can survive in hospital
ations that cannot be disregarded; a perception of dust for up to a year.11 It is found throughout the
cleanliness, however defined, is expected for hospital environment, particularly around patients
patients, their relatives and staff from healthcare known to be colonised or infected with the bac-
environments. terium. Molecular fingerprinting of these strains
Cleaning is routinely monitored by visual audit shows that MRSA-positive patients tend to shed
in the UK. Looking to see if a ward is clean may their own strain of MRSA into the near-patient
fulfil aesthetic obligations but it does not provide environment.12 If staff enter a room containing
a reliable assessment of the infection risk for an an MRSA patient, two-thirds of them will acquire
individual patient on that ward.3 The organisms the patient’s strain on gloved hands or apron.12
that cause infection are invisible to the naked Even if they do not touch the patient directly,
eye and their existence is not necessarily associ- four in ten will still exit the room carrying the
ated with the presence of visual dirt. Furthermore, patient’s strain of MRSA on hands or apron.12
the impression of cleanliness is confounded by MRSA can be found on general surfaces such as
clutter, and fabric and maintenance deficits.4 floors and radiators, furniture such as beds and
Visual assessment will inevitably be subject to lockers, and clinical equipment.7e10,12e14 Some
bias under these circumstances. It is more difficult sites, e.g. linen, curtains, beds, lockers and
to clean a crowded, cluttered environment, overbed tables, tend to harbour MRSA more
perhaps related to a cleaner’s incentive, when frequently than others.7,8,13 It is thought that
confronted with peeling plaster, cracked tiles or contamination of near-patient hand-touch sites
worn floor coverings.4 provides the biggest risk of MRSA acquisition for
Sites that are frequently touched by hands are patients.2,6 In addition, there is a small but signif-
thought to provide the greatest risk for patients, icant increase in the risk of acquiring MRSA if a pa-
and those situated right beside patients provide tient is admitted into a room previously occupied
the biggest risk of all.5e7 The responsibility for by carrier patients.15
cleaning near-patient hand-touch sites does not There is some evidence that cleaning removes
always rest with the ward cleaners, however, since MRSA from the ward environment with benefit for
beds, drip stands, lockers and overbed tables are patients.6,16 An outbreak of MRSA lingered for sev-
more usually cleaned by nurses.7,8 Nurses are eral months on a urological ward, resisting all the
also responsible for the decontamination of more usual infection control interventions such as pro-
delicate clinical equipment. This overlapping of motion of hand hygiene and isolation of patients.16
cleaning responsibilities has created some confu- The investigating team found the outbreak strain
sion; it has also meant that cleaning opportunities of MRSA scattered throughout the ward environ-
of some items are missed or abandoned.9,10 ment and doubled the number of domestic clean-
The microbial pathogens that cause HAI have ing hours from 60 per week to 120. Following
two special properties: first, they are recognised this, there was no further isolation of the outbreak
as hospital pathogens; second, they have an innate strain from the environment and the number of pa-
ability to survive on surfaces in the hospital tients affected decreased immediately. The clean-
environment for long periods of time.1 They in- ing intervention was thought to have played
clude organisms such as meticillin-resistant Staph- a significant role in the termination of the out-
ylococcus aureus (MRSA), Clostridium difficile, break and was estimated to have saved at least
vancomycin-resistant enterococci (VRE), Acineto- £28,000.16
bacter spp. and norovirus.1,6 This mini-review Another outbreak of glycopeptide-intermediate
will summarise the evidence for the presence S. aureus (GISA) in an intensive therapy unit
and survival of these organisms in the clinical en- proved difficult to control until a wave of further
vironment as well as support for cleaning as a valid control measures, including enhanced cleaning,
380 S.J. Dancer

was introduced.17 The outbreak encompassed two improved environmental cleaning on the spread
patient clusters, although genotyping showed that of VRE in a medical intensive care unit (ICU),
all cases were caused by the same strain. The with and without promotion of hand hygiene com-
second cluster occurred despite the institution of pliance.24 In this study, there were four periods:
maximum contact-isolation procedures. This di- the first served as a baseline period without any
rected attention towards the inanimate environ- interventions; the second began after a 30 day
ment as the major source of contamination, since period during which there was an education and
it was thought that re-emergence of the strain intensified observation programme to improve
could be explained by its ability to survive on inert cleaning; the third served as a ‘washout’ period
surfaces. The meticulous cleaning procedures without further interventions; and the fourth in-
finally implemented probably helped to stop the cluded an intensive hand hygiene campaign. The
outbreak, although it was not possible to deter- study found that enforcing cleaning measures
mine the relative roles of barrier precautions and along with encouraging hand hygiene was associ-
environmental decontamination in eradicating ated with less surface contamination with VRE,
the strain.17 cleaner healthcare worker hands and a significant
Outwith the outbreak situation, another study reduction in VRE cross-transmission among
examined the effects of enhanced cleaning on two patients. The authors concluded that decreasing
matched surgical wards in a controlled cross-over environmental contamination might help to
trial for two six-month periods.18 There were nine control the spread of VRE in hospitals.24
ward-acquired MRSA infections during routine
cleaning periods, but only four when the wards re-
ceived extra cleaning, notably targeting hand- Clostridium difficile
touch sites and clinical equipment. More MRSA pa-
tient-days during the enhanced cleaning periods C. difficile is a spore-forming anaerobe that has
predicted at least thirteen new cases instead of been recovered in abundance from the environ-
the four that actually occurred. The study con- ment of symptomatic patients.1,25 The more
cluded that targeted cleaning using detergent patients there are with C. difficile on the ward,
wipes and water could be a cost-effective mech- the more likely it is that other patients will also ac-
anism of reducing MRSA infection on a surgical quire the organism.26 Environmental contamina-
ward.18 tion with spores is now well accepted as a risk
factor for the acquisition of C. difficile.27 Further-
more, as the level of environmental contamination
Vancomycin-resistant enterococci increases, so does the amount of C. difficile on the
hands of healthcare workers, and once again near-
VRE are not credited with the same degree of patient hand-touch sites are regarded as a particu-
pathogenicity as MRSA, but they may still cause lar risk.25,28
infections in vulnerable patients, including out- Having established that there is a dynamic
breaks that are difficult to control. In addition, the transmission cycle for C. difficile similar to that
vanA gene has been shown to transfer to MRSA, for MRSA, there is additional evidence to support
making the latter even more difficult to treat.19 the value of cleaning in its control.1,29e31 Follow-
Part of the problem of controlling VRE in hospitals ing a rise in C. difficile cases, one hospital intro-
is due to their extreme longevity in the hospital duced enhanced cleaning with hypochlorite into
environment and their resistance to routine clean- two ICUs.31 One of the ICUs applied the extra
ing.1 Even bleach-based cleaning sometimes fails cleaning to all areas, including rooms used solely
to eradicate these hardy organisms.20 Contact by staff, and sensitive clinical equipment was
with contaminated surfaces in the rooms of colon- wiped over twice daily using hypochlorite-impreg-
ised patients results in transfer of VRE to gloved nated cloths. The other unit introduced the inten-
hands, despite cleaning with disinfectants.5,21,22 sive hypochlorite clean into isolation rooms
In addition, patients admitted into rooms previ- housing patients already infected with C. difficile.
ously accommodating patients with VRE are them- Rates of infection decreased in both units over sev-
selves more likely to acquire the organism.15 eral months and appeared to be maintained at
It has already been suggested that environmen- a lower rate for at least two years after the clean-
tal cleaning might be important in the control of ing intervention, despite some relaxation of the
VRE.23 Additional work has highlighted its presence initial regimen.31
on hand-touch sites near the patient, just as for Infection control teams do not question the
MRSA.10,22 A recent paper describes the impact of importance of thorough environmental cleaning
Environmental cleaning and HAI 381

for controlling C. difficile, although whether infection control measures that included the use
cleaners should use disinfectants or detergents of gloves before using the computer and thorough
for routine cleaning continues to be hotly de- disinfection of the plastic covers effectively termi-
bated.25,29,30 Although bleach cleaning certainly nated the outbreak. Up until the outbreak oc-
appears to have an effect on both environmental curred, no one had thought to include the
contamination and patient acquisition of C. diffi- computer keyboards in a routine cleaning specifi-
cile, it could be the increased physical effort of cation.39 Similarly, during a period of high en-
enhanced cleaning that contributes towards the demicity of acinetobacter in another burns unit,
overall result rather than a direct effect of the the endemic strain was identified from surfaces
bleach itself.30 It is known that cleaning of toilets close to the patient, including bed linen, medica-
and other sites using appropriate disinfectants tion table and the display surface of an overbed
does not totally remove all traces of C. diffi- monitor.40
cile.27,30,32,33 It is likely that agreement on the
best way of removing C. difficile from the environ-
ment will elude us for some time to come. Norovirus

Norovirus can be found on a huge variety of surfaces


Acinetobacter both in hospitals and in the community.41e43 Several
studies cite the association of norovirus with hand-
Acinetobacter can also be recovered from the touch sites such as toilet taps, door-handles, hospi-
hospital environment with ease, including inani- tal equipment, elevator and microwave buttons,
mate hand-touch sites near the patient.34 Seeding switches and telephones.41,42,44 When fingers
clinical and environmental strains onto Formica come into contact with virus-contaminated ma-
surfaces demonstrated survival of between one terial, norovirus is consistently transferred to
and two weeks, although some strains are known typical hand-touch sites.44
to survive for much longer.35 The importance of The importance of environmental cleaning in
cleaning in controlling outbreaks of Acinetobacter the control of outbreaks of norovirus is widely
spp. has been emphasised in previous studies.36,37 accepted.41,45 This includes clinical equipment, as
One of these describes an outbreak caused by mul- well as floors, toilets and general surfaces. Cur-
tiply resistant strains of A. baumannii involving tains should be removed and sent to the laundry,
more than thirty patients in two ICUs.37 Environ- and other soft furnishings either washed down or,
mental contamination was recognised as an impor- preferably, steam-cleaned. All general cleaning,
tant reservoir of the epidemic strains and the especially the toilets and bathroom areas, should
outbreak ceased only after both ICUs were closed be with a chlorine-containing disinfectant or
for terminal cleaning and disinfection.37 bleach at a specified concentration. Cleaning poli-
Another study examined the levels of environmental cies should always include the use of these disin-
contamination with acinetobacter in a neurosurgical fectants since detergent-based cleaning often
ICU during a prolonged outbreak.38 As with MRSA fails to eradicate the virus from the environ-
and C. difficile, there were many near-patient ment.44 Without scrupulous attention to the en-
hand-touch sites that yielded the epidemic strain. vironment, outbreaks not only continue, but will
This study also demonstrated a significant associa- resume within a short space of time. This is
tion between the amount of environmental con- because norovirus survives in the warm clinical
tamination and patient colonisation. The environment with ease while retaining its infectivity,
conclusion was that high standards of cleaning readily transferring to patients from hands that
play an integral role in controlling outbreaks of have just touched a contaminated site.
acinetobacter in the intensive care setting, al- Outside hospitals, norovirus outbreaks can be
though once again, little is known about the best devastating in closed or semi-closed communi-
way to clean in non-outbreak settings.38 ties.46 These include sudden and extensive out-
A further study describes what happened fol- breaks in hotels or cruise-liners, but outbreaks can
lowing the introduction of bedside computers in also occur in nursing and residential homes, prisons
a paediatric burns ward.39 There was a sudden and schools. An outbreak reported recently in a pri-
increase in the number of patients acquiring acine- mary school involved 79 pupils and 24 members of
tobacter and environmental screening demon- staff.47 Subsequent investigation of the outbreak
strated the organism on various surfaces in the showed that person-to-person contact was a major
patients’ rooms, especially the plastic covers factor in the transmission of the virus, but there
over the bedside computer keyboards. Targeted was evidence that poorly cleaned computer
382 S.J. Dancer

equipment was also implicated. A strain of noro- using a range of techniques in a coordinated and in-
virus, identical by RNA sequencing to two strains tegrated approach.48 Any isolation of pathogens, or
retrieved from patients, was isolated from one com- pathogen indicators, causes concern and warrants
puter keyboard and mouse in one particular class- immediate action. By contrast, environmental sur-
room. This occurred even after instituting face sampling in hospitals usually only takes place
a bleach clean the previous day. Public health offi- in response to an outbreak e and then only if the
cials recommended good hand-washing practices, infection control team responsible has the motive,
exclusion of symptomatic persons and thorough en- means and interest to initiate environmental screen-
vironmental disinfection with a diluted (1:50 con- ing.48 It is time that high risk surfaces were subjected
centration) household bleach solution, to include to routine screening in order to monitor overall
sites that are shared but not commonly cleaned.47 levels of microbial dirt and the results used to gener-
ate increased or targeted cleaning before a hospital
outbreak occurs. It is false economy to wait until
Discussion an outbreak occurs before the clinical environment
receives the attention it deserves.
There is plenty of evidence supporting the role of There is no reason why cleaning should not
domestic cleaning in hospitals as an important become an evidence-based science, particularly
intervention in the control of HAI. Unfortunately, when we know that lack of it is associated with
it often constitutes part of an overall infection human infection. Microbiological standards have
control package in response to an outbreak and its been proposed, using long-established principles
importance as a stand-alone activity remains from the food industry as well as from standards
controversial. This does not encourage on-going governing media such as air and water.2 Since the
managerial support for cleaning services in the pathogens of interest are widely scattered in time
hospital, particularly if resources are limited. The and space, these standards make use of quantita-
situation is hampered by the lack of scientific tive and qualitative microbial indicators rather
standards for hospital cleaning, rather than the than focus on trying to find a discrete pathogen.
subjective visual assessment practised at present. Practical applications for high risk surfaces in hospi-
The following three sections argue for a measured, tals would not be difficult to institute although the
targeted and methodological approach to domes- finer details related to risk, and site, require further
tic cleaning in hospitals. evaluation.2,49 Surfaces in outpatient corridors do
not present a similar risk of infection as hand-touch
What is ‘clean’? sites might, situated beside a ventilated patient in
an ICU. Attempts have already been made to com-
If we state that a hospital is clean, we assume that pare benchmark visual, ATP bioluminescence and
it looks clean and that it is safe for patients. microbiological values against each other, and one
Unfortunately, the microbes responsible for HAI study has attempted to evaluate microbiological
are invisible to the naked eye. This means that standards against infection risk in an ICU.8,50
visual assessment is insufficient for defining clean-
liness, nor will it accurately predict the infection Where to clean?
risk for patients.2 Cleanliness should not actually
be defined without indicating how it is assessed. There is increasing evidence regarding the impor-
A recent study compared visual assessment against tance of hand-touch sites in the transmission of
both biochemical (ATP bioluminescence) and mi- pathogens to healthy persons, as well as to
crobiological screening of the hospital environ- patients.51,52 It is also becoming apparent that
ment.3 The results showed that whereas most the sites closer to the patient are more likely to
surfaces looked clean, less than a quarter were furnish an infection risk than those situated fur-
free from organic soil (ATP) and less than half ther away.7,8 The role of these near-patient
were microbiologically clean.3 Given the risk of ac- hand-touch sites in MRSA transmission and, indeed,
quiring hospital pathogens from a hospital ward, other hospital pathogens, has not been given the
visual assessment is outdated, inadequate and sci- priority that it deserves. Ward cleaners work to
entifically obsolete. The only benefit from a visual a set specification that encompasses general
inspection is to appease aesthetic obligations. surfaces and bathrooms, with emphasis on the
There has been suggestion that hospitals would cleaning of floors and toilets.53 These are not nec-
benefit from cleaning standards emulating those essarily near-patient hand-touch sites. Examples
implemented in the food industry.2,48 Food prep- of the latter include bed rails, bedside lockers,
aration surfaces are subjected to routine sampling infusion pumps, door handles and various switches,
Environmental cleaning and HAI 383

including the nurse-call button, and they rarely a veritable blunderbuss approach to cleaning in
feature in the domestic specification.2,7,8 the event of an outbreak and managerial reluc-
It is already known that traditional sites such as tance to protect, let alone prioritise, routine
toilets, general surfaces and sinks tend to attract standards of cleaning outwith the outbreak situa-
high rates of cleaning but that hand-touch sites, tion. There is a lot of work still to do to establish
which are more likely to harbour and transmit cleaning as an evidence-based science, and to
microbial pathogens, are only poorly cleaned.10,54 translate the evidence into cleaning practices in
The responsibility for cleaning many hand-touch healthcare environments. Given the importance of
sites usually rests with the ward nurses, who are controlling HAI, it makes sense to support current
often very busy and almost permanently under- cleaning practises until such time as sufficient
staffed in many hospitals. Two recent studies in evidence is forthcoming.6
ICUs have demonstrated an increased risk of infec- There is plenty of evidence to support basic
tion following periods of inadequate nurse staffing, cleaning in hospitals. More and better-directed
or conversely, excess workload.55,56 It may be that cleaning reduces the risk of acquisition of a variety
concentrating available cleaning resources on of hospital organisms. It might also affect the
high-risk hand-touch sites would be the most number of patients with these so-called ‘super-
cost-effective cleaning strategy at the present bugs’ in the community, since hospital acquisition
time.18 A recent study has shown that it is possible invariably leads to patients taking them home.57
to improve environmental cleaning following an More interest in basic hygiene is warranted be-
educational campaign and feedback on adequacy cause microbes are becoming increasingly resis-
of discharge cleaning.10 tant due to the inappropriate, inadequate and
uncontrolled use of antibiotics and there are fewer
How to clean? and fewer agents in the developmental pipeline.58
A century ago, people died from trivial wounds
Most of the studies describing the benefits from because there were no antibiotics.
cleaning in this review used disinfectants to clean Simple hygiene could be our only defence when
the hospital environment. Virtually all were re- Darwinian evolution finally terminates the antibi-
ported as part of the response to an outbreak. Only otic era.59 We need to raise the level of awareness
a few UK-based studies used detergent and water, regarding hygiene, and its importance, throughout
and even fewer reported cleaning benefits in the society and it is hospitals that should lead the way.
absence of an outbreak.18,30 It appears that when If they at least start by cleaning up the way they
reviewing the evidence for the role of cleaning in could, then this would at least offer some sem-
the control of HAI, there are several issues which blance of safety to patients.
still require clarification. First, is there any differ-
ence between the quantity, quality and methods Conflict of interest statement
for routine cleaning compared with what is needed The author has received research funding from
in the event of an outbreak; and second, is it suf- UNISON for studies on hospital cleaning.
ficient to proclaim the benefits from cleaning with
disinfectants without establishing what can be Funding sources
achieved using soap and water alone? These ques- None.
tions require an evidence-based approach before
we can set the best specification for cleaning in
our hospitals. In addition, no one has yet modelled References
different cleaning methods against the infection
risk for patients, their degree of vulnerability and 1. Dancer SJ. Mopping up hospital infection. J Hosp Infect
the clinical area in which they are exposed. 1999;43:85e100.
Most hospitals have their own domestic specifi- 2. Dancer SJ. How do we assess hospital cleaning? A proposal
for microbiological standards for surface hygiene in hospi-
cations for wards, operating theatres, outpatient
tals. J Hosp Infect 2004;56:10e15.
and non-clinical areas. More countries are pro- 3. Griffith CJ, Cooper RA, Gilmore J, Davies C, Lewis M. An
ducing national standards for environmental clean- evaluation of hospital cleaning regimes and standards. J
ing. These set a necessary and valuable precedent Hosp Infect 2000;45:19e28.
but they are not based on sufficient scientific 4. Maurer IM. Hospital hygiene. 3rd edn. Bristol: Wright PSG;
1985.
evidence to justify their contents. At the beginning
5. Bhalla A, Pultz NJ, Gries DM, et al. Acquisition of nosoco-
of the twenty-first century, we simply do not know mial pathogens on hands after contact with environmental
how to clean our hospitals in order to create the surfaces near hospitalised patients. Infect Control Hosp
safest environment for patient care e hence Epidemiol 2004;25:164e167.
384 S.J. Dancer

6. Dancer SJ. Importance of the environment in meticillin- 24. Hayden MK, Bonten MJM, Blom DW, Lyle EA, van de
resistant Staphylococcus aureus acquisition: the case for Vijver DAMC, Weinstein RA. Reduction in acquisition of van-
hospital cleaning. Lancet Infect Dis 2008;8:101e113. comycin-resistant enterococcus after enforcement of rou-
7. Dancer SJ, White L, Robertson C. Monitoring environmental tine environmental cleaning measures. Clin Infect Dis
cleanliness on two surgical wards. Int J Environ Health Res 2006;42:1552e1560.
2008;18:357e364. 25. Verity P, Wilcox MH, Fawley W, Parnell P. Prospective evalu-
8. White L, Dancer SJ, Robertson C, McDonald J. Are hygiene ation of environmental contamination by Clostridium diffi-
standards useful in assessing infection risk? Am J Infect cile in isolation side rooms. J Hosp Infect 2001;49:204e209.
Control 2008;36:381e384. 26. Dubberke ER, Reske KA, Olsen MA, et al. Evaluation of Clos-
9. Blythe D, Keenlyside D, Dawson SJ, Galloway A. Environ- tridium difficile-associated disease pressure as a risk factor
mental contamination due to methicillin-resistant Staphy- for C. difficile-associated disease. Arch Intern Med 2007;
lococcus aureus (MRSA). J Hosp Infect 1998;38:67e70. 167:1092e1097.
10. Goodman ER, Platt R, Bass R, Onderdonk AB, Yokoe DS, 27. Kaatz GW, Gitlin SD, Schaberg DR, et al. Acquisition of Clos-
Huang SS. Impact of an environmental cleaning intervention tridium difficile from the hospital environment. Am J Epi-
on the presence of methicillin-resistant Staphylococcus au- demiol 1998;127:1289e1294.
reus and vancomycin-resistant enterococci on surfaces in 28. Samore MH, Venkatamaran L, DeGirolami PC, Arbeit RD,
intensive care unit rooms. Infect Control Hosp Epidemiol Karchmer AW. Clinical and molecular epidemiology of spor-
2008;29:593e599. adic and clustered cases of nosocomial Clostridium difficile
11. Wagenvoort JH, Sluijsmans W, Penders RJ. Better environ- diarrhoea. Am J Med 1996;100:32e40.
mental survival of outbreak vs. sporadic MRSA isolates. J 29. Mayfield JL, Leet T, Miller J, Mundy LM. Environmental con-
Hosp Infect 2000;45:231e234. trol to reduce transmission of Clostridium difficile. Clin
12. Boyce JM, Potter-Bynoe G, Chenevert C, King T. Environ- Infect Dis 2000;31:995e1000.
mental contamination due to methicillin-resistant Staphy- 30. Wilcox MH, Fawley WN, Wrigglesworth N, Parnell P, Verity P,
lococcus aureus: possible infection control implications. Freeman J. Comparison of the effect of detergent versus
Infect Control Hosp Epidemiol 1997;18:622e627. hypochlorite cleaning on environmental contamination
13. Lemmen SW, Hafner H, Zolldan D, Stanzel S, Lutticken R. and incidence of Clostridium difficile infection. J Hosp
Distribution of multi-resistant Gram-negative versus Gram- Infect 2003;54:109e114.
positive bacteria in the hospital inanimate environment. J 31. McMullen KM, Zack J, Coopersmith CM, Kollef M,
Hosp Infect 2004;56:191e197. Dubberke E, Warren DK. Use of hypochlorite solution to de-
14. Hardy KJ, Oppenheim BA, Gossain S, Gao F, Hawkey PM. A crease rates of Clostridium difficile-associated diarrhoea.
study of the relationship between environmental contami- Infect Control Hosp Epidemiol 2007;28:205e207.
nation with methicillin-resistant Staphylococcus aureus 32. Eckstein BC, Adams DA, Eckstein EC, et al. Reduction of Clos-
(MRSA) and patients’ acquisition of MRSA. Infect Control tridium difficile and vancomycin-resistant Enterococcus con-
Hosp Epidemiol 2006;27:127e132. tamination of environmental surfaces after an intervention
15. Huang SS, Datta R, Platt R. Risk of acquiring antibiotic-resis- to improve cleaning methods. BMC Infect Dis 2007;7:61.
tant bacteria from prior room occupants. Arch Intern Med 33. Alfa MJ, Dueck C, Olson N, et al. UV-visible marker confirms
2006;166:1945e1951. that environmental persistence of Clostridium difficile
16. Rampling A, Wiseman S, Davis L, et al. Evidence that hospital spores in toilets of patients with C. difficile-associated diar-
hygiene is important in the control of methicillin-resistant rhea is associated with lack of compliance with cleaning
Staphylococcus aureus. J Hosp Infect 2001;49:109e116. protocol. BMC Infect Dis 2008;8:64e70.
17. de Lassence A, Hidri N, Timsit JF, et al. Control and out- 34. Getchell-White SI, Donowitz LJ, Groschel DH. The inani-
come of a large outbreak of colonization and infection with mate environment of an intensive care unit as a potential
glycopeptide-intermediate Staphylococcus aureus in an in- source of nosocomial bacteria: evidence for long survival
tensive care unit. Clin Infect Dis 2006;42:170e178. of Acinetobacter calcoaceticus. Infect Control Hosp Epi-
18. Dancer SJ, White LF, Lamb J, Girvan EK, Robertson C. Mea- demiol 1989;10:402e407.
suring the effect of enhanced cleaning in a UK hospital: 35. Wendt C, Dietze B, Dietz E, Ruden H. Survival of Acineto-
a prospective cross-over study. BMC Infect Med 2009;7:28. bacter baumannii on dry surfaces. J Clin Microbiol 1997;
19. Sievert DM, Rudrik JT, Patel JB, McDonald LC, Wilkins MJ, 35:1394e1397.
Hageman JC. Vancomycin-resistant Staphylococcus aureus 36. Scerpella EG, Wanger AR, Armitige L, Anderlini P,
in the United States, 2002e2006. Clin Infect Dis 2008;46: Ericsson CD. Nosocomial outbreak caused by a multiresistant
668e674. clone of Acinetobacter baumannii: results of the caseecon-
20. Noble MA, Isaac-Renton JL, Bryce EA, et al. The toilet as trol and molecular epidemiologic investigations. Infect Con-
a transmission vector of vancomycin-resistant enterococci. trol Hosp Epidemiol 1995;16:92e97.
J Hosp Infect 1998;40:237e241. 37. Tankovic J, Legrand P, de Gatines G, Chemineau V, Brun-
21. Ray AJ, Hoyen CK, Das SM, Taub TF, Eckstein EC, Donskey CJ. Buisson C, Duval J. Characterisation of a hospital outbreak
Nosocomial transmission of vancomycin-resistant enterococci of imipenem-resistant Acinetobacter baumannii by pheno-
from surfaces. J Am Med Assoc 2002;287:1400e1401. typic and genotypic typing methods. J Clin Microbiol
22. Hayden MK, Blom DW, Lyle EA, Moore CG, Weinstein RA. 1994;32:2677e2681.
Risk of hand or glove contamination after contact with pa- 38. Denton M, Wilcox MH, Parnell P, et al. Role of environmen-
tients colonized with vancomycin-resistant enterococcus or tal cleaning in controlling an outbreak of Acinetobacter
the colonized patients’ environment. Infect Control Hosp baumannii on a neurosurgical intensive care unit. J Hosp In-
Epidemiol 2008;29:149e154. fect 2004;56:106e110.
23. Martinez JA, Ruthazer R, Hansjosten K, Barefoot L, 39. Neely A, Maley MP, Warden GD. Computer keyboards as res-
Snydman DR. Role of environmental contamination as a risk ervoirs for Acinetobacter baumannii in a burn hospital. Clin
factor for acquisition of vancomycin-resistant enterococci Infect Dis 1999;29:1358e1359.
in patients treated in a medical intensive care unit. Arch 40. Seifert H, Boullion B, Schulze A, Pulverer G. Plasmid DNA
Intern Med 2003;163:1905e1912. profiles of Acinetobacter baumannii: clinical application
Environmental cleaning and HAI 385

in a complex endemic setting. Infect Control Hosp Epi- 50. Lewis T, Griffith C, Gallo M, Weinbren M. A modified ATP
demiol 1994;15:520e528. benchmark for evaluating the cleaning of some hospital
41. Wu HM, Fornek M, Schwab KJ, et al. A norovirus outbreak at environmental surfaces. J Hosp Infect 2008;69:156e163.
a long-term-care facility: the role of environmental surface 51. Oelberg DG, Joyner SE, Jiang X, Laborde D, Islam MP,
contamination. Infect Control Hosp Epidemiol 2005;26: Pickering LK. Detection of pathogen transmission in neona-
802e810. tal nurseries using DNA markers as surrogate indicators.
42. Gallimore CI, Taylor C, Gennery AR, et al. Environmental Pediatrics 2000;105:311e315.
monitoring for gastroenteric viruses in a pediatric primary 52. Rheinbaben F, Schunemann S, Gross T, Wolff H. Transmis-
immunodeficiency unit. J Clin Microbiol 2006;44:395e399. sion of viruses via contact in a household setting: experi-
43. Evans MR, Meldrum R, Lane W, et al. An outbreak of viral ments using bacteriophage straight phiX174 as a mode
gastroenteritis following environmental contamination at virus. J Hosp Infect 2000;46:61e66.
a concert hall. Epidemiol Infect 2002;129:355e360. 53. Anonymous. New model cleaning contract. London: NHS
44. Barker J, Vipond IB, Bloomfield SF. Effects of cleaning and dis- Estates; December 2004.
infection in reducing the spread of norovirus contamination 54. Carling PC, Briggs JL, Perkins J, Highlander D. Improved
via environmental surfaces. J Hosp Infect 2004;58:42e49. cleaning of patient rooms using a new targeting method.
45. Green J, Wright PA, Gallimore CI, Mitchell O, Morgan- Clin Infect Dis 2006;42:385e388.
Capner P, Brown DWG. The role of environmental contami- 55. Dancer SJ, Coyne M, Speekenbrink A, Samavedam S,
nation with small round structured viruses in a hospital out- Kennedy J, Wallace PGM. Methicillin-resistant Staphylococ-
break investigated by reverse-transcriptase polymerase cus aureus (MRSA) acquisition in an intensive care unit
chain reaction assay. J Hosp Infect 1998;39:39e45. (ICU). Am J Infect Control 2006;34:10e17.
46. Love SS, Jiang X, Barrett E, Farkas T, Kelly S. A large hotel 56. Hugonnet S, Chevrolet J-C, Pittet D. The effect of workload
outbreak of Norwalk-like virus gastroenteritis among three on infection risk in critically ill patients. Crit Care Med
large groups of guests and hotel employees in Virginia. Epi- 2007;35:76e81.
demiol Infect 2002;129:127e132. 57. Scanvic A, Denic L, Gaillon S, Giry P, Andremont A,
47. CDC. Norovirus outbreak in an elementary school e District Lucet JC. Duration of colonisation by methicillin-resistant
of Columbia, February 2007. Morb Mortal Wkly Rep 2008; Staphylococcus aureus after hospital discharge and risk fac-
56:1340e1343. tors for prolonged carriage. Clin Infect Dis 2001;32:
48. Griffith CJ. Hospital-acquired infection: are there lessons 1393e1398.
from the food industry? Biomed Sci 2006 August:697e699. 58. Budd R. Penicillin: triumph and tragedy. Oxford: Oxford
49. Al-Hamad A, Maxwell S. How clean is clean? Proposed University Press; 2007.
methods for hospital cleaning assessment. J Hosp Infect 59. Dancer SJ. Back to cleanliness. Rapid response. Br Med J
2008;70:328e334. 2008;336.
Journal of Hospital Infection (2009) 73, 386e391
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Controversies in infection: infection control or


antibiotic stewardship to control
healthcare-acquired infection?
I.M. Gould*

Department of Medical Microbiology, Aberdeen Royal Infirmary, Foresterhill, Aberdeen AB25 2ZN, UK

Available online 10 July 2009

KEYWORDS Summary Despite record resource being devoted to the control of health-
Antibiotic stewardship; care-acquired infection (HCAI), rates have never been higher. Although the
Healthcare-acquired discovery of the contagiousness of puerperal sepsis by Alexander Gordon
infection;
heralded the golden era of bacteriology and antibiotics, this led to a belief
Infection control
that infection was beaten. This in its turn may well have led us into a false
sense of security and an over-reliance on antibiotics. Modern medicine has
built many of its advances on a need for antibiotics, but their very success
has led to huge over-use and resulting problems of resistance. Compounded
by the absence of a good antibiotic pipeline we are now being forced to ad-
dress the paradox of antibiotics; namely that they may actually be causing
many HCAIs. Not only Clostridium difficile infection, but many others such
as those caused by meticillin-resistant Staphylococcus aureus, are more or
less completely contingent on antibiotic prescribing. Control of prescribing
would probably be just as effective a measure in our fight against HCAI as
conventional infection control measures. Arguably, traditional infection
control is akin to fire-fighting and antibiotic stewardship to prevention.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

Introduction infection (HCAI) seemingly uncontrollable, it is


pertinent to assess whether current strategies to
In a time of unparalleled resource devoted to contain HCAI are failing and, if so, why?
infection control, and healthcare-acquired Certainly, the public perception is of hospitals
as the dangerous places of Florence Nightingale’s
time.1 Meticillin-resistant Staphylococcus aureus
* Tel.: þ44 1224 554954; fax: þ44 1224 550632. (MRSA) rates continue to rise in most countries,
E-mail address: i.m.gould@abdn.ac.uk as do most of the multi-resistant Gram-negative
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.02.023
Control of healthcare-acquired infection 387

bacteria (see the European Antimicrobial Resis- But it is to the obstetrician Alexander Gordon of
tance Surveillance System website) and in many Aberdeen (1752e1798) that we must look for the
countries Clostridium difficile seems to be an in- next big breakthrough.5 Based on detailed diaries
creasing problem, not withstanding its ascertain- he kept of an outbreak of puerperal fever which
ment difficulties.2 With MRSA and C. difficile, at ran its course from 1789e1793 in Aberdeen, Gordon
least, it is quite clear that those infections deduced the relationship between erysipelas and
are an additional burden, with rates of meticillin- puerperal fever, its cause as being transferred
susceptible S. aureus (MSSA) infection remaining from the postmortem room to the lying-in room
stable or even increasing.3 by midwives and its prevention by adequate
What is also notable is that the majority of day- hand hygiene and disinfection of clothes. His
to-day infection control problems in the hospital treatise, first published in 1795, predates Sem-
are due to increasingly resistant bacteria. In this melweis by more than 50 years but despite being
bicentenary year of Charles Darwin’s birth it is reprinted several times over the next century, in
pertinent to reflect on the role that antibiotic use London and the USA, he has always been over-
may have as a selecting force for evolution of shadowed by Semmelweis.6 Interestingly, like
these ‘superbugs’. Semmelweis, Gordon made himself unpopular
In this short review I will reflect on our over- among his colleagues with his observations. Soon
reliance on antibiotics in modern medicine and try after, he left Aberdeen and died of tuberculosis.
to put antibiotic stewardship in historical perspec- Gordon was cited by Oliver Wendell Holmes but
tive alongside infection control, at least as an not by Semmelweis, so it is interesting to specu-
equal partner in our fight against HCAI. late on whether Semmelweis was actually aware
of Gordon’s work.7 Possibly he was and probably
he should have been, as the treatise had been
Are infection control policies failing? published so many times. Also, Scottish medicine
was well renowned during that period and com-
Ignaz Semmelweis is often cited as the father of munication with and travel to Europe frequent.
modern infection control, although such concepts On the other hand, Semmelweis was said not to
are actually more than 2000 years older!4 have a good grasp of the medical literature, un-
Marcus Terentius Varro (116e27 BC), a Roman like Gordon.8
soldier and Director of the Imperial Library in Further advance awaited Pasteur’s refutation of
Rome, warned against the building of homes near spontaneous generation of life, Koch’s recognition
swamps ‘because there are bred certain minute of the first pathogenic organism (anthrax), Lister’s
creatures which cannot be seen by the eyes, which implementation of Pasteur’s findings in the form of
float in the air and enter the body through the antiseptic surgery and its development into aseptic
mouth and nose and there cause serious diseases’. surgery.9 In the meantime, Florence Nightingale
Girolamo Fracastoro (1478e1553) was perhaps had introduced better sanitation and hand washing
the first to formulate a recognisable ‘modern at the Barrack hospital in Scutari, although her
theory’ of contagion. He stated that it could occur role in this has recently been debated.1
in three ways; by direct contact with the infected, It is well documented that antiseptic and sub-
by contact with their clothes, or through the air. sequently aseptic surgery and other procedures
He described the agents of contagion as germs or were rapidly taken up by virtually all informed
seeds, small imperceptible particles, each specific medical practitioners over the following years and
for a different disease. popular perception is that they were well adhered
It took the invention of the microscope for to, with the traditional matron overseeing good
theories to advance. Robert Hooke (1635e1703), practice.9 But does this still generally apply or has
Athanasius Kircher (1602e1680) and Antonie poor practice crept in, albeit perhaps just outside
van Leeuwenhoek (1632e1723) described micro- the operating theatre? Certainly this seems to be
organisms in numerous settings using microscopes. the case where hand hygiene adherence rates of
Hooke even described ‘little worms’ in the blood 20e40% are considered normal.10 If this be so,
of victims of bubonic plague in 1656! By 1764, Sir may it be due directly or indirectly to the great
John Pringle (1707e1782) had established the success of, and perhaps over-reliance on, anti-
basic principles of sanitation and ventilation of biotics e with a belief that antibiotics can negate
hospital wards and military quarters. He uses the the need for good hygiene, asepsis and cleanliness?
word ‘antiseptic’ in the context of Kircher’s Furthermore, could antibiotic use actually be
theory of contagion by microscopic germs harmful in some instances, actually increasing
(‘animalcula’). the number of infections? Increasingly we hear
388 I.M. Gould

calls for zero tolerance in failure to adhere to hand Much antibiotic over-use may seem justifiable
hygiene and other infection control policies. But for the individual patient when viewed in isolation;
perhaps some of the policies of recent years have nevertheless antibiotic use, unlike any other drug
been misplaced. Take universal precautions category, cannot be viewed in such isolation.
(UPs), for example. Such a blind faith in their
broad applicability has, I believe, been one of Antibiotic use as a cause of HCAI
the main reasons for the failure to control
MRSA.11 Over-reliance on UPs to the exclusion of Each antibiotic prescription has an environmental,
specific policies targeting the biology of problem ecological consequence.18 Only over the past
organisms such as MRSA is counterproductive, decade has this become widely accepted, although
even if UPs could be successfully implemented. A Fleming warned of this at an early stage. But the
problem organism such as MRSA should be targeted downside of antibiotics goes well beyond develop-
specifically according to its method of spread and ment of resistance as already mentioned with
biological weaknesses.12 In the case of MRSA this MRSA and C. difficile. Indeed, not only does anti-
means active surveillance cultures, admission cul- biotic use select for and maintain antibiotic resis-
tures, isolation, decolonisation and decontamina- tance, but it also enhances its spread.19,20 This
tion.13 This has been clear for at least three latter point is crucial to the control of modern
decades but with a few exceptions, for example HCAI and illustrates how potentially critical anti-
in Scandinavia and Holland, such policies have biotic stewardship is in the control of most HCAIs.
been very poorly implemented. Possibly it also illustrates why traditional infec-
tion control policies have not been as successful
The antibiotic era as we would have hoped.
Increased transmission of tetracycline-resistant
It is hard to imagine hospital medicine in the pre- organisms during tetracycline therapy was demon-
antibiotic era, but contemporary reports of the strated as long ago as 1960.21 In 2008 increased
difference that antibiotics made to individual cases numbers of MRSA were demonstrated in the noses
well describe the fantastic advance that they of carriers receiving quinolones or b-lactams (to
were.14 Indeed it is the case that many of the great which the MRSA strains were resistant) compared
advances of modern medicine and surgery would with controls.22 Presumably this is due to the com-
not have been possible without the ability to rely petitive advantage that was achieved by the anti-
on antibiotics to cure secondary infection. But at biotic administration, ablating the normal
what cost? The average hospital now uses, on aver- protective commensal flora (including MSSA), al-
age, levels approaching 100 defined daily doses per lowing multiplication of the MRSA with increased
100 occupied bed-days, a value equivalent to eco- potential for contaminating the environment. It
logical saturation.15 This is the equivalent of all pa- is easy to imagine adjacent patients being more
tients receiving a full daily dose of antibiotic from susceptible to acquisition of MRSA if they are also
the day of admission until discharge. Of course, on an appropriate ‘MRSA selecting’ antibiotic. Fur-
on average, only less than half of inpatients will ac- thermore, antibiotics such as the quinolones and
tually receive antibiotics at any one time, but those cephalosporins are well known to increase expres-
who do receive them are often on double doses or sion of fibronectin adhesins, facilitating adherence
combination therapy. Many guidelines advocate and ability to colonise, and also many other viru-
prolonged antibiotic prophylaxis, particularly for lence factors such as toxins which might cause col-
the ever-increasing number of immunosuppressed onisation to develop into infection.19,20 With the
patients, and often these uses are justified by a rea- increasing trend to admission screening for MRSA,
sonable evidence base.16 On many other occasions, such knowledge should be put to good use by
however, prescribing is definitely inappropriate e avoiding prescription of agents to which the
‘just in case’ or on the basis of a poor quality sever- MRSA is resistant, both in MRSA carriers and their
ity assessment or misdiagnosis. Current policies to contacts. This is not always an easy task when
shorten length of stay and curtail costs also dealing with multiply resistant MRSA.
encourage empiric use, often of unnecessarily In 1970 Sleigh et al. described inability to
broad-spectrum antibiotics. Combination therapy control an outbreak of multidrug-resistant (MDR)
is often used for a number of reasons, good Klebsiella pneumoniae in a neurosurgery unit.23
and bad, including broadening spectrum to accom- All conventional control measures had failed, so
modate increasing antibiotic resistance, thus the authors took the unusual, and probably never
completing the spiralling circle of therapeutic to be repeated, step of banning prescription of
empiricism.17 all antibiotics. Not only did the outbreak strain
Control of healthcare-acquired infection 389

disappear (actually it may have been declining all such encoded resistance determinants. Also,
anyway) but, more interestingly, the total number any loss of fitness can often be compensated for
of infections decreased dramatically and no one by mutations in the bacteria. So it may be that
died of uncontrolled infection during the period proper stewardship can only halt the current de-
of embargo on antibiotic prescriptions. velopment of resistance, although there are
So we have to ask ourselves, could antibiotic use enough examples of reversals to give us some
be increasing the incidence of most HCAIs due to hope.28
MDR bacteria, and not just MRSA and C. difficile? One of the main problems is how to reduce
Perhaps it is not just resistance that is caused by overall antibiotic use, as modern medical devel-
antibiotics but also HCAI? This is a frightening opments seem to know no end to the immunosup-
thought on one hand, as we have become so reliant pression of the patient. Currently, our efforts on
on antibiotics. On the other hand, this reliance has stewardship (which mainly revolve around formu-
probably become over-reliance, leading to poor laries and guidelines) achieve only uniformity of
quality infection control in the belief that infec- prescribing with adherence to policies, guidelines
tion has been beaten by antibiotics. But it is also and formularies. Paradoxically this may actually
a good time to take stock, as the antibiotic pipe- be more harmful, as the best defence against
line is dry and there are no significant develop- resistance is probably diversity of prescribing.29
ments expected for another 10e20 years.24 So Little effort is actually put into reducing prescrib-
the sooner we are more cautious in our use of ing with the possible exception of shortening dose
antibiotics the better, needing to preserve those duration. What is really required is better diagno-
that we have. sis (of course including better diagnostics), better
Returning to MRSA as an example, there are severity assessment and, where antibiotics are in-
many caseecontrol studies demonstrating prior dicated, better knowledge of pharmacokinetic/
antibiotic exposure, particularly with cephalo- pharmacodynamic dosing schedules to optimise
sporins and quinolones as a risk factor for both outcome and delay development of resistance.
MRSA colonisation and infection.25 The same These are difficult things to address in the middle
applies, of course, to many other MDR bacteria of the night when relatively junior doctors are
including most of the MDR Gram-negatives. For making the prescribing decision, often trying
MRSA there are also numerous ecological studies to protect themselves from the threat of under-
demonstrating such a relationship at a community treating (and perhaps of litigation) and trying to
and hospital level.14 There are even a dozen or so do their best for the patient. More is better or let’s
studies in the literature suggesting that MRSA rates prescribe ‘just in case’!
were decreased by antibiotic policies that reduced The example of recent guidelines from the
use of cephalosporins and quinolones.26 Usually, British and American Thoracic Societies for treat-
however, such studies demonstrate the concept ment of community-acquired pneumonia (CAP) are
of ‘squeezing the balloon’ whereby reduction in a good case in point.30,31 They make recommenda-
use of one drug is mirrored by increasing use of an- tions for various severities of illness but the natu-
other. Usually this will have its own resistance ral inclination of most clinicians is always to
problems, although it may be that the cephalo- ‘cover’ themselves or their patients with the rec-
sporins and quinolones, in particular, are more ommendation for treatment of severe cases. Argu-
prone to resistance developing than the penicil- ably, such resulting over-use of cephalosporins,
lins, for example. quinolones and macrolides may have triggered
the MRSA outbreaks in North America and the
UK.32 The guidelines themselves are not without
Antibiotic stewardship fault. While they purport to be evidence-based,
the antibiotic recommendations are definitely
Unfortunately, reducing total antibiotic use is not, leaning heavily to combination therapy and
even more difficult than modulating class use the perceived need to cover all aetiological
and there are very few robust studies in the agents. The Scandinavian countries, which by and
literature that demonstrate a reversal of resis- large have retained penicillin monotherapy as
tance.27 There are complex molecular reasons their treatment of choice for CAP, have not
why this might be the case, particularly the published worse outcomes and have many fewer
mobile gene cassette that can integrate on problems with antibiotic resistance.
chromosomes to encode multiple resistance Gone are the days when we can aspire to ‘cover’
determinents. Use of any one agent thus encoded 100% in the empiric treatment of every severe
can provide selection advantage for maintaining infection.33 Paradoxically, this is at a time when it
390 I.M. Gould

has never been clearer that immediate appropri- in intensive-care units: prospective two-centre study. Lan-
ate empiric therapy is beneficial to the septic cet 2005;365:295e304.
11. Gould IM. Can we control MRSA? Scott Med J 2007;52:3e4.
patient. Difficult decisions have to be made. For 12. Lindsay JA, Holden MT. Understanding the rise of the super-
instance, do we prescribe a carbapenem and bug: investigation of the evolution and genomic variation of
glycopeptide for all septic patients if we have an Staphylococcus aureus. Funct Integr Genomics 2006;6:
ESBL and MRSA problem? At what ‘level’ is MRSA 186e201.
or ESBL a problem that should impact on empiric 13. Gould IM, MacKenzie FM, MacLennan G, Pacitti D, Watson EJ,
Noble DW. Topical antimicrobials in combination with admis-
choice? What happens when carbapenem resis- sion screening and barrier precautions to control endemic
tance or glycopeptide resistance become problem- methicillin-resistant Staphylococcus aureus in an intensive
atic, as they already have in some parts of the care unit. Int J Antimicrob Agents 2007;29:536e543.
world? Colistin, tigecycline and fosfomycin are 14. Jeffrey JS, Thomson S. Penicillin in battle casualties.
possible alternatives but not so universally appli- Br Med J 1944;2(4356):1e4.
15. MacKenzie FM, Bruce J, Struelens MJ, Goossens H,
cable as carbapenems. Outcome with them or Mollison J, Gould IM, ARPAC Steering Group. Antimicrobial
with glycopeptides, even in susceptible organisms, drug use and infection control practices associated with
may be suboptimal, making decisions on empiric the prevalence of methicillin-resistant Staphylococcus au-
therapy even more difficult. While re-evaluation reus in European hospitals. Clin Microbiol Infect 2007;13:
and possible step-down are often an option within 269e276.
16. Gafter-Gvili A, Paul M, Fraser A, Leibovici L. Effect of quino-
the next 48 h, this will be too late for some lone prophylaxis in afebrile neutropenic patients on micro-
patients and in the majority there will not be any bial resistance: systemic review and meta-analysis.
positive cultures to guide follow-on therapy. J Antimicrob Chemother 2007;59:5e22.
Much more strategic thought needs to be given to 17. Kim JH, Gallis HA. Observations on spiraling empiricism: its
these important issues in the absence of new causes, allure, and perils, with particular reference to anti-
biotic therapy. Am J Med 1989;87:201e206.
antibiotics to help us out of this dilemma. 18. Sarkar P, Gould IM. Antimicrobial agents are societal drugs:
In conclusion, new thinking is needed in our use how should this influence prescribing? Drugs 2006;66:
of antibiotics, not only to delay the development 893e901.
of further antibiotic resistance but to help in the 19. Gould IM. Antibiotic policies to control hospital-acquired in-
control of HCAI. This will require widespread fection. J Antimicrob Chemother 2008;61:763e765.
20. Dancer SJ. The effect of antibiotics on methicillin-resistant
consultation and the time just might be now. Staphylococcus aureus. J Antimicrob Chemother 2008;61:
246e253.
Conflict of interest statement 21. Berntsen C, McDermott W. Increased transmissibility of
None declared. staphylococci to patients receiving an antimicrobial drug.
N Engl J Med 1960;262:637e642.
22. Cheng VCC, Li IWS, Wu AKL, et al. Effect of antibiotics
Funding sources on the bacterial load of meticillin-resistant Staphylococcus
None. aureus colonization in anterior nares. J Hosp Infect 2008;
70:27e34.
23. Price DJE, Sleigh JD. Control of infection due to Klebsiella
References aerogenes in a neurosurgical unit by withdrawal of all anti-
biotics. Lancet 1970;2:1213e1215.
1. Williams K. Reappraising Florence Nightingale. Br Med J 24. Gould IM. Antimicrobials: an endangered species? Int J Anti-
2008;337:1461e1466. microb Agents 2007;30:383e384.
2. Planche T, Aghaizu A, Holliman R, et al. Diagnosis of Clos- 25. Tacconelli E. Does antibiotic exposure increase the risk of
tridium difficile infection by toxin detection kits: a system- methicillin-resistant Staphylococcus aureus (MRSA) isola-
atic review. Lancet Infect Dis 2008;8:777e784. tion? A systematic review and meta-analysis. J Antimicrob
3. Gould IM. Costs of hospital-acquired methicillin-resistant Chemother 2008;61:26e38.
Staphylococcus aureus (MRSA) and its control. Int J Antimi- 26. Gould IM. Comment on: Interventions to control MRSA: high
crob Agents 2006;28:379e384. time for time-series analysis? J Antimicrob Chemother
4. Dallas J. Little worms which propagate plague. J R Coll 2009;63:224.
Physicians Edinb 2008;38:376e377. 27. Davey P, Brown E, Fenelon L, et al. Systematic review of
5. Porter IA. Alexander Gordon, M.D. of Aberdeen 1752e1799. antimicrobial drug prescribing in hospitals. Emerg Infect
Edinburgh: University of Aberdeen/Oliver & Boyd; 1959. Dis 2006;12:211e216.
6. Gordon A. Treatise on the epidemic puerperal fever of 28. Gould IM. The epidemiology of antibiotic resistance. Int J
Aberdeen. London; 1795. Antimicrob Agents 2008;32:S2eS9.
7. Holmes OW. The contagiousness of puerperal fever. N Engl 29. Sandiumenge A, Diaz E, Rodriguez A, et al. Impact of di-
Q J Med Surg 1843;i:503. versity of antibiotic use on the development of antimicro-
8. Loudon I. The tragedy of childbed fever. London: Oxford bial resistance. J Antimicrob Chemother 2006;57:
University Press; 2000. 1197e1204.
9. Bulloch W. The history of bacteriology. London: Oxford Uni- 30. Anonymous. Guidelines for the management of adults with
versity Press; 1960. hospital-acquired, ventilator-associated, and healthcare-
10. Cepeda JA, Whitehouse T, Cooper B, et al. Isolation of pa- associated pneumonia. Am J Respir Crit Care Med 2005;
tients in single rooms or cohorts to reduce spread of MRSA 171:388e416.
Control of healthcare-acquired infection 391

31. British Thoracic Society Standards of Care Committee. BTS aureus, Aberdeen, 1996e2000. Emerg Infect Dis 2004;10:
guidelines for the management of community acquired 1432e1441.
pneumonia in adults. Thorax 2001;56:iv1e64. 33. Masterton RG. The new treatment paradigm and the role
32. Monnet DL, MacKenzie FM, López-Lozano JM, et al. Antimi- of carbapenems. Int J Antimicrob Agents 2009;33:
crobial drug use and methicillin-resistant Staphylococcus 105e110.
Journal of Hospital Infection (2009) 73, 392e396
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Where does infection control fit into a hospital


management structure?
E.T. Brannigan a, E. Murray a,b, A. Holmes a,b,*
a
Imperial College Healthcare NHS Trust, London, UK
b
Imperial College, London, UK

Available online 20 August 2009

KEYWORDS Summary To be effective, infection prevention and control must be


Hospital-acquired integrated into the complex and multiple interlinking systems within a hos-
infection; pital’s management structure. Each of the systems must consider how
Organisational change;
activity associated with it can be optimised to minimise infection risk to
Patient safety;
Quality
patients. The components of an organisational structure to achieve these
quality assurance and patient safety aims are discussed. The use of perfor-
mance management tools in relation to infection control metrics is re-
viewed, and the use of hospital-acquired infection as a proxy indicator
for deficiencies of system management is considered. Infection prevention
and control cannot be the role and responsibility of a single individual or
a small dedicated team; rather it should be a priority at all levels and
integrated within all management systems, including the research and
educational agendas.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

Introduction and control. Any such structure should set its sights
beyond merely addressing any legislative and stat-
Where does infection control fit into a hospital utory requirements, and should provide a frame-
management structure? The question should first work for organisational change as well as the
be asked what a hospital management structure systems required to support this change. Key ele-
should provide in relation to infection prevention ments of this framework include a belief in a shared
purpose and aspiration to excellence, supporting
reinforcement systems, assurance that staff have
* Corresponding author. Address: Imperial College London,
Hammersmith Hospital Campus, Du Cane Road, London W12
the skills required for change and that staff have
0NN, UK. consistent and multiple role models within the
E-mail address: alison.holmes@imperial.ac.uk organisation. These elements could almost be
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.03.031
Hospital management and infection 393

considered together as an ‘organisational change Impact of human resources management


bundle’. The structure should also have the ability on infection control
to foster reliability and resilience.1,2
Hospital management must consider infection Areas in which HR management has a direct impact
control primarily as a core aspect of patient on infection control include occupational health,
safety, and as an indicator of quality of care. It health clearance and vaccination against prevent-
must be acknowledged that achieving quality able infection. However, manpower details such
patient care and safety is entirely dependent on as working patterns, team formation and skill-mix
implementation of best practice in individual are also inextricably linked to infection control
clinical care, and that this delivery requires outcomes. Infection risk has been shown to be
expert input at both operational and strategic strongly related to staffing levels and staff train-
levels. ing. These are underpinned by other HR activities
including recruitment and retention, staff ap-
Role and interplay of multiple praisal, job descriptions, induction, leave manage-
ment, agency staff and shift patterns. HR
management systems management has been shown to have particular
impact in the context of critical care areas,
Infection prevention and control relies upon the especially intensive care units (ICUs). Staffing
successful interplay and management of multiple levels among nursing staff especially have been
management systems, integrating infection pre- shown significantly to affect hospital-acquired
vention and control into management at all levels infection, both in neonatal and in adult ICUs.3,4
and forcing specific consideration of infection risk Physician staffing levels have also been studied;
for patients throughout multiple management infection-related outcomes are not addressed sep-
systems. To achieve optimally lowered infection arately, but findings relate to the overall adverse
risk for patients, action is required in a wide impact on LOS and mortality.5
variety of domains. Examples of the diverse areas
and aspects of a hospital infrastructure which must
be considered as having a role in infection pre-
vention and control include human resources (HR),
An organisation-wide approach
staff:patient ratios, bed management, patient
pathways, staff training, information and informa- A comprehensive organisational approach should
tion technology, contract management, procure- be adopted, one which recognises existing man-
ment, estates and facilities, capital planning, agement structures and facilitates development of
building, design, performance monitoring, anti- a model that fully integrates infection control into
biotic stewardship, organisational learning, adop- hospital management and the quality agenda.
tion of innovation, risk management, governance, Infection control must be a core part of gover-
priority setting, resource allocation, communica- nance and must maintain a high clinical profile
tions, and business planning among others. Given alongside a high management profile.
the complexity of assuring high quality standards
across such diverse groups and the contingency of
each of these systems one on the other, achieving Bridging the gap between managers and
effective infection prevention cannot therefore clinicians
be devolved to an individual specialist team or
to an isolated group of committed experts. Infection control provides an opportunity, agenda
Instead a comprehensive whole-scale organisa- and framework for such a bridging strategy. If
tional approach is required. Infection risk needs infection control sits solely within a separate
to be considered in almost all areas of hospital service or team, unlinked to management strat-
management, patient pathways and delivery of egy, or with little clinical and managerial influ-
care. Awareness of infection prevention priorities ence, it has limited effectiveness and impact.
must be embedded and be considered a prerequi- Strategic decision-makers without knowledge or
site in decision-making at all levels. Infection without systems to ensure consideration of in-
control must be fully integrated into the health- fection control put patients at risk. Managers
care structure and systems, into performance and need to be aware of the direct impact, any
patient outcome metrics and into the hospital knock-on effects and potential collateral damage
culture including in research and education that could occur as a result of any decision-
strategy. making, whether it be about capital planning,
394 E.T. Brannigan et al.

pathways, service reconfiguration, etc. Avoiding and metrics which drive continuous learning and
any compromise in infection prevention and pa- improvement. A critical component in all of these
tient safety depends on continuous awareness is strong corporate accountability and hospital
which can be achieved by improved dialogue and leadership. ‘Achieving safety requires more
working to a common agenda.6e9 Skills and training than individual carefulness. It is a corporate
needs of management and clinical staff must be responsibility.’13,14
realised, and there must be clear lines of account-
ability which follow funding structures, and which
Leadership
are supported by agreed performance indicators.10
These indicators should be recognised and incor-
Within a hospital structure leadership must come
porated in any balanced scorecards, dashboards
directly from the Chief Executive Officer (CEO)
or organisational performance monitoring tools.
who must have demonstrable commitment to
infection prevention and control and leader-
Balanced scorecard for infection ship.15 The Director of Infection Prevention and
prevention and patient safety Control (DIPC) is the person appointed by each
trust who has direct line of accountability to
the CEO and this role provides opportunities for
The use of the balanced scorecard provides
specialist leadership and composite roles.16 The
a framework to measure performance beyond
DIPC leads and champions infection prevention
finances in private industry. It allows alignment
and control at multiple levels within the organi-
of performance measures with an organisation’s
sation, ensuring that a consistent message is
strategic mission and goals, measuring perfor-
embedded in directorates, groups, teams, and
mance, and factors driving performance. In es-
networks. Consistent role models are key to
sence it is a tool that can provide a basis for
organisational change.1 The DIPC must have
executing good strategy well and managing
a profile and recognition of this role by peer
change successfully. There are caveats with such
groups and the organisation and provide a bridg-
an approach inasmuch as you get what you
ing role between managers and clinicians. The
measure, and use of a scorecard is known to
multiple layered approach allows the DIPC to
skew activity. The scorecard is not fixed; instead
address issues pertinent to patients and to public
it needs regular refreshing and updating as prior-
concerns first and foremost, to communicate
ities of the institution evolve.11 Many trusts use
a clear message to staff which states and pro-
the balanced scorecard approach for internal per-
motes the view of infection control as a shared
formance managing. Indicators used must have
goal involving all staff, across the multidisciplin-
credibility with frontline staff and have shared
ary spectrum. This includes delivering a strong
agreement of their value. Leaders must select
message to all prescribers relating to prudent
outcome or process measures that represent the
prescribing and ensuring that antibiotic steward-
interests of frontline healthcare workers, must
ship is fully addressed by the organisation. The
ensure that it has local face validity, and that per-
DIPC role must deliver internal reinforcement,
formance on the chosen measure will help focus
leadership, board-to-ward engagement, exper-
quality improvement.12
tise and organisational commitment, as well as
provide role modelling and exemplify corporate
Organisational learning and culture responsibility.

This requires creation of an environment which


permits continuous learning and quality improve- Lessons learned from approaches and
ment shared across professional bodies, and across processes from outside healthcare
directorates and units. Systems-based approaches
embedded in the framework of the organisation Like many other areas of hospital management and
must be adopted for sustainability. These systems patient safety, much can be learned from arenas
must be shaped and supported by clinical and outside healthcare. It is also worth noting that the
managerial expertise, within a culture that sup- significant reductions of MRSA bacteraemias made
ports and reinforces infection control as a corpor- by many NHS trusts were due not to new technol-
ate priority, continually reinforcing the required ogy or scientific advances but through better
behaviour and values. Interventions must be sup- application of existing knowledge using a variety
ported by regular feedback, monitoring systems of managerial approaches. These include the risk
Hospital management and infection 395

reduction and hazard management approaches HCAI as an indicator of complex systems


from high risk industries. management and organisational
Organisational approaches, as used in other
resilience
complex industries, can be applied to acute trusts
to embed infection control into the running of
There is pressure for greater use of quality in-
hospitals and delivery of clinical care, and organisa-
dicators and measures in healthcare, and infection
tional change management tools can be used to
rates and infection prevention is a major indicator
drive this. These include leadership and the
of quality in clinical care and quality in manage-
development of a ‘board-to-ward’ approach of
ment.21,22 Infection control can serve as a marker
engagement, and performance indicators with
of an organisation’s capacity to manage multiple,
scorecards or dashboards while maintaining aware-
complex systems, and HCAI rates can therefore
ness of their limitations. In addition to trusts
be proxy indicators of levels of staffing, levels
addressing internal reinforcement systems, exter-
of training, organisational stress, management
nal reinforcement provides the scrutiny and
failure, inadequate systems, reliability, and
incentives to achieve goals, and has been seen
resilience.
with public information campaigns, the introduc-
A resilient system is one that quickly recovers
tion of mandatory reporting, the intense media
stability after an unexpected event or in the face
interest and increased patient choice. Strong
of continuous significant stresses in a complex,
external reinforcement of CEO accountability for
dynamic environment; the stability is provided by
healthcare-associated infections (HCAIs) was pro-
constant change rather than continuous repeti-
vided not just by mandatory data submission in
tion.23 The challenge to the system is that it must
England, but also by the mandatory requirement
facilitate reliability while developing resilience to
that CEOs review and sign off data monthly. Finally
both internal and external threats and stressors,
the introduction of legislation made the delivery of
e.g. mergers, leadership change, financial deficit,
infection prevention and control and compliance
infection outbreaks, global recession, major policy
with core duties legal requirements for every NHS
shift, environmental incident and technology
trust.
change among others.
Valuable lessons from the safety arena include
Common to both the Maidstone and Stoke
the use of quality improvement tools and the
Mandeville Health Care Commission reports re-
development and adoption of ‘care bundles’ to
lating to C. difficile were themes related to organ-
ensure best practice and minimising risk in partic-
isational stressors; recent mergers, preoccupation
ular clinical interventions. Local improvement
with financial situation, service reconfigurations
campaigns can learn form advertising and politics,
and high bed occupancy levels (>90%). This was
as have the national patient safety and quality
in addition to the more obviously directly related
improvement campaigns such as those of the
issues such as poor antibiotic stewardship, issues
Institute of Health Improvement’s ‘1000 lives’
relating to isolation facilities and the state of the
campaign, the ‘5 million lives’ campaign, and the
hospital environment.24,25
DoH ‘Saving Lives’ campaign. These national cam-
Resilient organisations hold to key principles:
paigns also provided additional external reinforce-
acknowledging even small failings, resisting over-
ment to local programmes.
simplified accounts of events, remaining sensitive
The effective use of teams has been recognised
and responsive to front-line operations, maintain-
as a key aspect of delivering quality performance
ing reserves and capability in anticipation of
in other industries. This involves better use of
stressors, and are adaptable to shifting locations
team structures with an acknowledgement of in-
of expertise.23 Each of these principles should be
dividual skills, but with staff understanding that
addressed by a trust in its overarching approach
teamwork is valued rather than individual inter-
to infection prevention and control.
ventions. This relies on leadership, adaptability, as
well as mutual performance monitoring and sup-
port.17,18 The team approach has been particularly
valuable in reducing line-associated bacteraemia Discussion
in ICUs. Staff can be further supported by encour-
agement of use of decision aids, systems which Infection prevention and control should have
make the desired action the default, and clear de- a presence and profile throughout the hospital
lineation of processes and pathways, all of which management structure. From the top and down
seek to decrease the human factor and increase through every level of decision-making and care,
the system reliability.19,20 supported by clear leadership, systems, structures
396 E.T. Brannigan et al.

and expert input, an organisational approach to 8. Edwards N. Doctors and managers: building a new relation-
infection prevention is needed, involving manage- ship. Clin Med 2005;5:577e579.
9. Edwards N, Kornacki MJ, Silversin J. Unhappy doctors: what
ment structures and systems to change behaviour are the causes and what can be done? Br Med J 2002;324:
and culture, to drive quality improvement and 835e838.
support sustainable best practice in a resilient 10. Holmes A, Dinneen M, Murray E. Creating a new culture.
framework. In this context training needs must be Publ Serv Rev: Health 2006;8:19e22.
addressed, and further applied research is needed 11. Kaplan RS, Norton DP. Using the balanced scorecard as
a strategic management system. Harv Bus Rev 1996;1:
to determine which healthcare models best deliver 75e85.
effective, reliable and resilient infection 12. Pronovost PJ, Berenholtz SM, Needham DM. A framework for
prevention.26,27 health care organizations to develop and evaluate a safety
scorecard. J Am Med Assoc 2007;298:2063e2065.
13. Leape L, Epstein AM, Hamel MB. A series on patient safety.
N Engl J Med 2002;347:1272e1274.
Acknowledgements 14. Annas GJ. The patient’s right to safety e improving the
quality of care through litigation against hospitals. N Engl
We are grateful to the support from the National J Med 2006;354:2063e2066.
Institute of Health Research and the Biomedical 15. Institute of Medicine. In: Kohn LT, Corrigan J, Donaldson MS,
Research Centre at Imperial College. editors. To err is human: building a safer health system.
Washington, DC: National Academy Press; 1999.
16. Department of Health. Winning ways: working together to
Conflict of interest statement reduce healthcare associated infection in England. London:
None declared. DoH; 2003.
17. Baker DP, Day R, Salas E. Teamwork as an essential compo-
Funding sources nent of high-reliability organizations. Health Serv Res 2006;
41:1576e1598.
Support from the NIHR.
18. Wenzel RP, Edmond MB. Team-based prevention of cath-
eter-related infections. N Engl J Med 2006;355:2781e2783.
19. Resar R. Making noncatastrophic health care processes re-
References liable: learning to walk before running in creating
high-reliability organizations. Health Serv Res 2006;41:
1. Lawson E, Price C. Psychology of change management. 1677e1689.
McKinsey Q 2003;2:30e41. 20. Pronovost PJ. Creating high reliability in health care organi-
2. Holmes A. Working smarter: an organisational approach to zations. Health Serv Res 2006;41:1599e1617.
infection prevention. Bull R Coll Pathol 2008;142:106e109. 21. Darzi A. High quality care for all. NHS next stage review
3. Harbarth S, Sudre P, Dharan S, Cadenas M, Pittet D. Out- final report. London: Department of Health; 2008.
break of Enterobacter cloacae related to understaffing, 22. Vincent C, Aylin P, Dean Franklin B, et al. Is health care
overcrowding, and poor hygiene practices. Infect Control getting safer? Br Med J 2008;337:1205e1207.
Hosp Epidemiol 1999;20:598e603. 23. Weick K, Sutcliffe K. Managing the unexpected: assuring
4. Hugonnet S, Chevrolet JC, Pittet D. The effects of workload high performance in an age of complexity. San Francisco:
on infection risk in critically ill patients. Crit Care Med Jossey-Bass; 2001.
2007;35:296e298. 24. Healthcare Commission. Investigation into outbreaks of
5. Pronovost PJ, Angus DC, Dorman T, Robinson KA, Clostridium difficile at Maidstone and Tunbridge Wells
Dremsizov TT, Young TL. Physician staffing patterns and NHS trust. London: HC; 2007.
clinical outcomes in critically ill patients: a systematic re- 25. Healthcare Commission. Investigation into outbreaks of
view. J Am Med Assoc 2002;288:2151e2162. Clostridium difficile at Stoke Mandeville hospital, Bucking-
6. Silversin J, Kornacki M. Leading physicians through change: hamshire hospitals NHS trust. London: HC; 2006.
how to achieve and sustain results. Tampa, FL: American 26. McDonnell A, Wilson R, Goodacre S. Evaluating and imple-
College of Physician Executives; 2000. menting new services. Br Med J 2006;332:109e112.
7. Shortell SM, Waters TM, Clarke KWB, Budetti PP. Physicians 27. UK Clinical Research Collaboration. Translational infection
as double agents: maintaining trust in an era of multiple research initiative. London: Medical Research Council;
accountabilities. J Am Med Assoc 1998;280:1102e1108. 2008.
Journal of Hospital Infection (2009) 73, 397e399
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Hand hygiene and infection in hospitals: what do


the public know; what should the public know?
M. Fletcher*

National Patient Safety Agency, London, UK

Available online 26 September 2009

KEYWORDS Summary Healthcare-associated infection (HCAI) is a topic of increasing


Health care associated public interest, particularly to users of health services. At the same time,
infections; there is a move towards greater openness and transparency across the
Patients;
whole healthcare sector. Thus we see public reporting of performance in
Perception;
relation to rates of HCAI and infection control practices is increasingly well
Public;
Awareness; established in the NHS in England. So does any of this make a difference?
Information And how embedded is the ‘public right to know’? In this paper it is argued
that, although the public right to know about rates of HCAIs is well recog-
nised, the evidence base about the impact of such information is limited.
The paper suggests actions which can be taken by boards and senior
leaders in healthcare organisations to increase impact. Furthermore the
example of hand hygiene suggests that we have some way to go in creating
an environment in which patients feel empowered to ask questions that
may reduce their own vulnerability to infection.
ª 2009 Published by Elsevier Ltd on behalf of The Hospital Infection Society.

Introduction hospitalised.2 At the same time, there is a move


towards greater openness and transparency across
Healthcare-associated infection (HCAI) is a topic of the whole healthcare sector.3
increasing public interest, particularly to users of Arguably, the ‘public right to know’ is already
the National Health Service (NHS) in England.1 This an accepted fact by the NHS in England. Public
is accompanied by genuine public concern about reporting of performance in relation to rates of
the risk of becoming infected as a result of being HCAI and infection control practices is increasingly
well established. There is a large e and growing e
amount of information about HCAI in the public
* Address: National Patient Safety Agency, 4-8 Maple Street,
domain. For example, many NHS trusts’ websites
London WIT 5HD, UK. Tel.: þ44 20 7927 9527. routinely display data on their infection control
E-mail address: martin.fletcher@npsa.nhs.uk programme and performance. Such information is
0195-6701/$ - see front matter ª 2009 Published by Elsevier Ltd on behalf of The Hospital Infection Society.
doi:10.1016/j.jhin.2009.06.029
398 M. Fletcher

also increasingly common at the entrance to effectiveness, safety and patient-centeredness re-
hospitals and clinical areas. More widely, the mains uncertain.
Health Protection Agency routinely publishes Boards and senior leaders in healthcare organi-
data on national infection rates.4 sations have a key role to play in building a culture
So does any of this make a difference? And how of safety and openness in their organisations.8 How
embedded is the ‘public right to know’? can they make the ‘public right to know’ more
In this paper it is argued that, although the meaningful?
public right to know about rates of HCAIs is well Three actions are suggested. First, boards and
recognised, the evidence base about the impact of senior leaders need to pay close attention to the
such information is limited. The paper suggests purpose, design and implementation of informa-
actions which can be taken by boards and senior tion that is publicly reported. A key question to ask
leaders in healthcare organisations to increase is ‘What do we want to achieve by publishing
impact. Furthermore the example of hand hygiene patient care performance information?’ Second,
suggests that we have some way to go in creating the public need to be involved in developing and
an environment in which patients feel empowered designing measures on which to report. A key
to ask questions that may reduce their own question to ask is ‘How have we involved key
vulnerability to infection. audiences in the design of the measures?’ Third,
the presentation of information needs to be
The evidence on public reporting improved. A key question to ask is ‘How can we
present information in ways that are easy to use
Broader than the field of infection control, the and understand?’
research literature suggests a number of drivers
for publishing patient care performance data, The example of hand hygiene
including5:
At its most basic, it could be argued that the
e as a means of improving the quality of care
‘public right to know’ should commence with
through greater accountability and
a patient being able to ask healthcare workers
transparency;
(HCWs) whether they have cleaned their hands. In
e as a means of informing selection by patients or
practice, this does not appear to happen often,
their intermediaries in the context of select-
and what appears simple is, in fact, much more
ing, rewarding or incentivising better perform-
complex.
ing providers;
Surveys with HCWs undertaken by the National
e to help providers identify areas in which they
Patient Safety Agency’s ‘cleanyourhands’ cam-
are underperforming.
paign suggest that nearly two-thirds of these staff
feel that being prompted by patients could help to
There is little evidence about the effect of
improve their hand hygiene behaviour.9 At the
publication both in the context of HCAI and wider
same time, around one-quarter of these staff felt
health service performance.6 The growth in public
that such a prompt would create tension between
reporting has not been matched by a growth of re-
HCWs and patients.
search on impact. There are mixed signals on the
Surveys with hospital inpatients and members
effect of public reporting on patient outcomes.
of the public indicate that around half of the
Publication of performance data can be an impor-
respondents said that they were unlikely ever to
tant stimulus for quality improvement. This sug-
ask nurses or doctors whether they had cleaned
gests that the organisation which is publishing
their hands. In practice, many fewer respondents
data may itself be one of the key audiences. There
had actually asked this question of a doctor or
is emerging evidence on consumer selection of
nurseethis is despite the fact that patient
health plans in an American context with some
involvement has been an important feature of
suggestion of unintended consequences such as
the cleanyourhands campaign in England and
adverse selection. As Fung et al. conclude7:
Wales from its launch in 2004. Indeed, more
Evidence is scant particularly about individual pro- than 96% of trusts say that they involve patients
viders and practices. Rigorous evaluation of many in their work to improve infection control.9 The
major public reporting systems is lacking. Evidence Chief Medical Officer for England has also drawn
suggests that publicly releasing performance data attention to the importance of patient involve-
stimulates quality improvement activity at the hos- ment in influencing the hand hygiene behaviour
pital level. The effect of public reporting on of staff.10
Hospital hygiene and infection: what should the public know? 399

Discussion ‘public right to know’ has not yet fully reached the
bedside.
Despite the growth in publication of performance
data on HCAI, and public awareness of the risks of Conflict of interest statement
infection, it appears that many members of the None declared.
public are not comfortable to ask HCWs whether
they have cleaned their hands before touching Funding sources
them; further, that a significant proportion of None.
HCWs feel uncomfortable about being asked this
question. This would suggest that the ‘public right References
to know’ does not yet extend to this most basic
information. In short, awareness and information 1. Gould DJ, Drey NS, Millar M, Wilks M, Chamney M. Patients
do not appear to be leading to change. and the public: knowledge, sources of information and per-
ceptions about healthcare-associated infection. J Hosp
What lies behind this? Three factors are
Infect 2009;72:1e8.
suggested. 2. Gill J, Kumar R, Todd J, et al. Meticillin-resistant Staphylo-
First, patients want to believe that their HCWs coccus aureus: awareness and perceptions. J Hosp Infect
are doing the right thing and do not want to 2006;62:333e337.
think that they may not be getting anything less 3. Department of Health. Handbook to the NHS Constitution.
London: DoH; 2009.
than the best possible care.11 Second, the ques-
4. Health Protection Agency. Surveillance of healthcare
tioning of a HCW often feels for both parties associated infections report. London: HPA; 2008.
like a criticism of the clinical skills of the HCW 5. Berwick DM, James B, Coye MJ. Connections between
rather than a helpful part of ensuring the safest, quality measurement and improvement. Med Care
cleanest care. This reflects broader findings on 2003;41:30e38.
6. Shekelle PG, Yee-Wei L, Mattke S, Damberg C. Does public
patient involvement in patient safety.12 Third,
release of performance results improve quality of care? A
many healthcare organisations have not created systematic review. London: Health Foundation; 2008.
an environment in which both patients and 7. Fung CH, Lim Y-W, Mattke S, Damberg C, Shekelle PG.
HCWs feel that it is indeed ‘OK to ask’. In short, Systematic review: the evidence that publishing patient
for many patients, the risk of perceived harm of care performance data improves quality of care. Ann Intern
Med 2008;148:111e123.
such a question appears to outweigh the benefit
8. Pittet D, Donaldson L. Clean care is safer care: the first
of action. This is despite public concern about global challenge of the WHO World Alliance for Patient
HCAIs. Safety. Am J Infect Control 2005;33:476e479.
In conclusion, the public do have a right to know 9. National Patient Safety Agency. It’s OK to ask: empowering
about the infection rate in their hospital. More and the hospital patient to ask about hand hygiene. 2009
[Unpublished observations].
more information is being put into the public
10. Chief Medical Officer. On the state of public health: annual
domain. However, the availability of such infor- report of the Chief Medical Officer. London: Department of
mation is not enough on its own. Patient involve- Health; 2006.
ment in hand hygiene is a good example of the 11. Nuffield Trust. Involving people in public disclosure of
complexity and challenge. The fact that many clinical data. Nuffield Trust; 2003.
12. Davis RE, Koutantji M, Vincent CA. How willing are patients
patients do not feel able to ask HCWs whether
to question healthcare staff on issues related to the quality
they have cleaned their handseand that HCWs feel and safety of their healthcare? An exploratory study. Qual
uncomfortable being asked e suggests that the Saf Health Care 2008;17:90e96.
Journal of Hospital Infection (2009) 73, 400e407
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

What are the drivers of the UK media coverage of


meticillin-resistant Staphylococcus aureus, the
inter-relationships and relative influences?
T. Boyce a,*, E. Murray b, A. Holmes b
a
The King’s Fund, London, UK
b
Imperial College Healthcare NHS Trust, Imperial College, London, UK

Available online 20 August 2009

KEYWORDS Summary Meticillin-resistant Staphylococcus aureus (MRSA) infection in


Hospital-acquired the UK receives intense media attention. The nature of coverage, political
infection; responses and solutions offered has been questioned and the relationship
Hospital cleaning;
between health professionals, the media and government policy needs
Mass media;
Meticillin-resistant
greater understanding. We identified 2880 articles on MRSA published in
Staphylococcus 12 UK newspapers between 1994 and 2005, compared with 21 articles in
aureus; six major US newspapers. To investigate the relative influences and rela-
News; tionships further, 68 weeks of coverage from 1990 to 2004 were analysed.
Risk communication The dates were selected based on publication dates of the ten most
frequently cited articles on MRSA according the ISI Web of Science portal
of Department of Health press releases on MRSA since 1997. Within this
period, 351 news articles were published with members of the public and
politicians representing 60% of sources quoted. Scientific articles, even
those with the highest number of citations, have negligible influence on
newspaper coverage. Simple solutions quoted in the newspaper articles
focused almost exclusively on cleaning. The UK press exhibits a high interest
in MRSA compared with that of the USA. Healthcare workers, experts and
professional bodies have criticised the nature of media reporting, but have
had little influence or involvement in the press. This may facilitate journal-
ists, celebrities, the public and politicians to drive these stories unchecked
and allow politics to address only the simplistic solutions generated.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

* Corresponding author. Address: Research Fellow in Public Health, The King’s Fund, 11e13 Cavendish Square, London W1G 0AN,
UK. Tel.: þ44 20 7307 2663; fax: þ44 20 7307 2809.
E-mail address: t.boyce@kingsfund.org.uk

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.05.022
UK media coverage of MRSA 401

Introduction research were published and when the UK De-


partment of Health (DoH) issued press releases
In a 2008 UK-wide British Broadcasting Corporation about MRSA. Selecting these key dates when these
(BBC) poll on the National Health Service (NHS), different sources might have influenced press
acquiring an infection while in hospital ranked as coverage allowed us to explore the relationship
the main public concern about inpatient hospital and influence between the government, media and
care (BBC News online: Infections ‘the biggest NHS published medical research.
fear’; 30 June 2008). Concerns about infection
received a much higher rating (40%) than clinical
issues such as poor quality treatment (8%) or Methods
insufficient staff to meet patients’ needs (10%).1
Meticillin-resistant Staphylococcus aureus (MRSA) Twelve quality and popular newspapers with the
has become a major preoccupation of the UK highest circulation in the UK (Daily Telegraph,
media, with tabloid headlines promoting mops Sunday Telegraph, Guardian, Observer, Daily
and matrons as an overarching simple solution, Mail, Mail on Sunday, Mirror, Sunday Mirror, Sun,
but with more complex factors such as antibiotic News of the World, The Times, Sunday Times)
stewardship, bed management, isolation capacity, and six major US newspapers: New York Times,
staffing, behaviour and practice receiving little Los Angeles Times, Washington Post, USA Today,
coverage.2 This article explores these issues and Boston Globe and Chicago Tribune (widely
analyses the interactions and relative influences regarded as the elite of the US newspaper market
of scientists, clinicians, policymakers and public and highly influential on other media outlets) were
opinion on the media coverage of MRSA in the UK. examined for their coverage of MRSA including the
Since the early 1960s Western nations have number of articles published.
witnessed increasing numbers of infections associ-
ated with MRSA.3,4 In 2004 the UK National Audit Selection of time periods for further analysis
Office reported that from 2001 to 2004, there had of UK press
been an 8% increase in the number of reported
S. aureus bacteraemias (from 17 933 to 19 311) and From 1990 to 2004, a total of 30 separate dates in
an increase of 5% in the number of cases attributed 12 national UK newspapers were studied. 1990 is
to MRSA (from 7250 to 7647).5 The issue of MRSA in the start of the sample as this is the earliest date
England is unique for three reasons. First, in Octo- when most UK newspapers appear on the Lexis
ber 2000, the UK government announced that each Nexis subsription service database, which is
acute hospital trust in England would monitor and a searchable archive of international newspapers,
report numbers of MRSA bacteraemias. The first magazines, legal documents and other printed
hospital-acquired infection (HAI) for mandatory re- sources. The dates are based on the activities of
porting was to be MRSA bloodstream infection (BSI). two key sources of material on MRSA: medical and
Second, a reduction target was set by the govern- scientific journals and the UK Department of
ment of 50% in three years. Third, alongside the Health (DoH). The top ten cited articles published
increase in MRSA BSI cases that was then being between 1990 and 2004 with MRSA as a subject
seen in the UK, media coverage increased and inten- were identified on the ISI Web of Science portal.
sified, with MRSA often at the top of the UK news The publication dates of these papers were then
agenda. This was a level of coverage that appeared used as dates for analysis. ‘Most-cited’ status
to be unprecedented in other national media. may take several months, if not several years, to
Media representations of health issues are sig- be achieved; these articles are proxies to show
nificant as key sources of information for the public the type of coverage science research receives.
both as patients making health decisions and as an Tabloids and broadsheets can and do cover compli-
electorate. Media coverage can also drive health cated science stories, for example, coverage of
policy.6,7 The role of media in driving change and stem cell science demonstrates that tabloids do
providing external reinforcement to drive change report complicated science stories.11,12
must also be considered and understood, particu- All press releases about MRSA published on the
larly in light of the MRSA BSI reduction achieved in UK DoH website since 1997 provided an additional
England as reported in September 2008.8e10 19 dates. The Health Protection Agency (HPA) also
UK national newspaper coverage of MRSA was issues press releases related to MRSA; however,
examined over a 13 year period, covering 30 key most concern the rates of MRSA, some of which
dates when influential pieces of peer-reviewed appear on the DoH website. They issue their own
402 T. Boyce et al.

press releases, and two press releases in our more commonly used by journalists and there are
sample originated as HPA press releases. The HPA’s a larger number of articles using MRSA. This
own press releases are not included in the sample selection process resulted in a total of 233 news
as journalists primarily turn to the HPA for statis- articles which mentioned the DoH press releases or
tics on MRSA rates. They are not widely regarded journal articles (Table I).13e22
as key media agenda-setting bodies.12 The articles on MRSA were examined to identify:
An additional date when scientists proactively (i) story leads; (ii) quoted sources; (iii) reported
tried to engage the media about MRSA was also causes of the problem; and (iv) reported solutions.
selected. This was a press conference held in July
2004 at the Science Media Centre.
Each date was examined for a three-week Results
period; from one week before the press release
or journal article was published and the two weeks MRSA reporting in UK versus USA
afterwards. This period was used to include cover-
age before official press releases are issued (due to In 2004, a total of 737 articles about MRSA were
unofficial leaks) and also to gauge how coverage published in major UK national newspapers and by
evolves after information from science and gov- 2005 this had increased to 1514, a total of 2251
ernment sources has been released. Some dates articles in just two years. By contrast, reporting in
overlap, particularly in the summer and winter of the USA was significantly lower. Only 21 articles
2004 when a number of DoH press releases were were published in six major US newspapers from
published. Accounting for overlapping dates, a to- 1994 to 2005 (Figure 1). There were signs that this
tal of 68 weeks were analysed over 13 years. The was changing, with a marked increase from five
search term ‘MRSA’ was used, every article with articles in 2005 to 125 articles in 2007. The main
this term was analysed (duplicates articles were themes of HAI media coverage differed. In the
omitted). Whereas both MRSA and M.R.S.A. are UK coverage was primarily concerned with hospi-
used by journalists in both countries, for consis- tal-acquired infections, the NHS and governments
tency, MRSA was used as the search term e it is being blamed for poor response or lack of action

Table I Newspaper articles identified on 30 key dates (pertaining to publication of Department of Health or
journal articles) from 1990 to 2004 containing the term ‘MRSA’ where Department of Health or journal articles
were cited
Documents analysed No. of articles where Main theme
press release is mentioned
Science journal articles: 0 Medical research
Levine et al. (1991)13
Panlilio et al. (1992)14
Kreiswirth et al. (1993)15
Mulligan et al. (1993)16
Voss et al. (1994)17
Hiramatsu et al. (1997)18
Herold et al. (1998)19
Enright et al. (2000)20
Pittet et al. (2000)21
Kuroda et al. (2001)22 2 Medical research
Science Media Centre press briefing 3 Medical research
DoH press release dates:
12.07.04, 19.10.04, 04.11.04, 07.12.04 34 Cleaning
12.06.00, 09.06.03, 05.12.03, 15.10.04 21 Solutions
08.02.02, 14.07.04 18 Rates
22.11.99, 21.07.04, 05.11.04 15 Targets
01.12.04 3 Products
15.12.04 1 Science
23.04.98, 03.09.98, 17.12.98 0 Report
16.10.00 0 Monitoring
MRSA, meticillin-resistant Staphylococcus aureus; DoH, Department of Health.
UK media coverage of MRSA 403

1600

1400

1200
No. of articles on MRSA

1000

800

600

400

200

0
1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007
Year

Figure 1 Meticillin-resistant Staphylococcus aureus (MRSA)-related news reporting by newspapers in the UK (dashed
line) compared with the USA (solid line) from 1995 to 2007.

to reduce HAI. By contrast, US coverage focused on the episode ‘(e)veryday MRSA was in the newspa-
community-acquired infections, as there is no cen- pers.’22,23 Since 2002, MRSA has received substan-
tral healthcare system; newspapers focused on in- tial coverage in the UK national press, particularly
fections found in schools and sporting facilities, in the popular newspapers.c A total of 233 articles
and blamed individuals. containing the term ‘MRSA’ appeared in the 12
newspapers.
Analysis of UK press coverage over key dates Only five newspaper articles refer to scientific
journal articles: two cited Kuroda et al. (2001)22
Between 1986 and 1994, MRSA received sporadic (published in The Lancet) and referred to a Science
coverage in the UK media. By 2000, MRSA reporting Media Centre briefing. By contrast, 95 articles
had attained a momentum with celebrities de- (41%) included information from a DoH press re-
scribing their MRSA-related experiences. TV actress lease (Table I).
Lesley Ash had meticillin-susceptible S. aureus
(MSSA) but the media widely reported that she Story leads
had MRSA. Agony aunt and former nurse Claire
Rayner, also president of the Patients Association, The lead of most stories was not about science and
frequently appeared in the media as a vocal vic- data, but instead about victims or political state-
tim of MRSA. MRSA was also seeping into fictional ments about MRSA. This emphasis on personal
media. In the opening episode of the 2006 Dr Who narratives and political statements demonstrates
series, a race of nun-like catwomen ran a hospital the difficulty for scientists or scientific research to
with thousands of humans infected with a disease have a significant impact on how MRSA is covered.
secretly hidden in the basement. Screenwriter Instead of the story being about systematic prob-
Russell T. Davies said that when he was writing lems or public health challenges, it becomes

c
Traditionally UK newspapers were classified by size and hence quality: ‘tabloids’ were aimed at a ‘popular’ (mainly working class)
market whereas ‘broadsheets’ were ‘quality’ newspapers aimed at a more educated and wealthy market. In 2003 the first UK broad-
sheet changed its size to tabloid and many others soon followed. Hence the more common terms now used to describe UK newspapers
are ‘quality’ and ‘popular’.
404 T. Boyce et al.

a human interest story. Personal stories were more then reported by journalists is in contradiction to
common in the popular newspapers, where 44 those more complex solutions offered by health
stories led with this theme, compared with only professionals.
nine stories in the quality papers. Personal stories
put a ‘human face’ to medical or scientific stories Reported causes
and are viewed by journalists as a way to capture
the audience’s attention.24 Scientific research When the causes of MRSA were reported, jour-
made up 15% of all leads, but what makes this sta- nalists used simple discourses and rarely reported
tistic even more significant is that these stories the findings of scientific evidence or referred to
were chosen during periods when sources (the possible problems with NHS systems or structures.
DoH and scientists) were trying to control or con- Cleaners and cleanliness was the leading re-
tribute to the news agenda. ported cause and accounted for 102 (35.9%) of
all causes discussed. More complex ideas such as
Sources for quotations by journalists the challenges of the poor isolation capacity in
NHS hospitals were mentioned in only 4.9% of
An analysis of sources quoted directly by journal- articles. Other ideas such as staffing levels, use of
ists in their stories revealed that health profes- agency staff, screening policies, or intravenous
sionals were absent from much of the coverage of line care were not mentioned at all (Table III).
MRSA. The most common sources were members of Thirty-eight percent of stories contained no ref-
the public affected by MRSA, suggesting that the erences to the causes of MRSA, leaving a large
dominant discourse in stories about MRSA is not gap in terms of opportunities to build public
scientific, but rather a more general and access- awareness.
ible discourse (Table II).
Organisations such as the Hospital Infection Soci- Reported solutions
ety, the Infection Control Nurses Association (sub-
sequently known as the Infection Prevention With the emphasis on cleaning as the primary
Society), the Royal Colleges (of Physicians, Surgeons, cause of MRSA, it is therefore no surprise that
Pathologists and Nursing) and the British Medical the majority of reported solutions (Table IV) also
Association are largely absent from the coverage. related to cleaning. Although environmental
The public was the most common source: the cleaning has a role in contributing to the control
‘common-sense’ solution of cleaning accounted for of HAI levels, the politically determined target
69% of the solutions reported. This apparently was a reduction in a specific infection caused by
simple solution to MRSA offered by the public and a specific organism: hospital-acquired bacteraemia

Table II Newspaper articles identified on 30 key dates (pertaining to publication of Department of Health or
journal articles) from 1990 to 2004 containing the term ‘MRSA’ by sources directly quoted in all articles
Sources directly quoted in all articles No. of appearances % total sources
Public affected by MRSA 66 16.3
Politician (Labour Party) 52 12.9
Scientist/doctor 51 12.6
Politician other 42 10.4
DoH/HPA 41 10.2
Hospital representatives 36 8.9
Public e other 22 5.5
Pressure group 21 5.2
Other (e.g. lawyer, union, private health care, celebrity) 33 8.2
Health e other 12 3
Nurse 9 2.2
BMA/Royal Colleges 7 1.7
Infection Control Nurses Association 6 1.5
Hospital Infection Society 3 0.7
Cleaners 3 0.7
Total 404
MRSA, meticillin-resistant Staphylococcus aureus; DoH, Department of Health; HPA, Health Protection Agency; BMA, British
Medical Association.
UK media coverage of MRSA 405

Table III Newspaper articles identified on 30 key dates (pertaining to publication of Department of Health or
journal articles) from 1990 to 2004 containing the term ‘MRSA’ by reported causes of MRSA
Reported causes of MRSAa No. %
Cleaners/cleanliness 102 35.9
Hospital staff’s own hygiene/lack of knowledge 63 22.2
Other (e.g. government policy, visitors, shortage of consultants) 33 11.6
Government health targets 22 7.7
Invasive procedures 17 6
Over-prescription of antibiotics 16 5.6
Bed occupancy levels 14 4.9
Lack of isolation rooms 14 4.9
Shortage of consultants with infectious disease control expertise 2 0.7
Pressure on health professionals to prescribe antibiotics 1 0.4
Total 284
Articles not citing causes 88
MRSA, meticillin-resistant Staphylococcus aureus.
a
More than one cause can be included in a story.

caused by MRSA. It was this MRSA BSI data, sup- Considering the level of information, or how the
plied regularly by the mandatory reporting problem is defined, it is apparent that although the
scheme, that was being monitored. The near media did include some ‘facts’ about MRSA,
exclusive focus on environmental cleaning was the government cannot depend on the media to
unhelpful in that it suggested that hospital cleanli- do the job of informing the public about MRSA,
ness alone contributed to bacteraemia prevention especially the likely causes and potential solutions
and controlling antibiotic-resistant infection, and to the problem. This responsibility lies with health
that cleaning could ‘solve’ completely the problem professionals.
of increasing MRSA BSI. Some research also con-
cluded that there was ‘no direct link’ between Discussion
hospital cleanliness and the risk of MRSA bacterae-
mia.25 The way in which causes of the MRSA prob- When considering the paucity of stories resulting
lem and the potential solutions are reported is from the ten most highly cited scientific and
important because the media ‘can affect the types medical journal articles on MRSA, it is clear that
of policy solutions the public and policymakers science did not influence media coverage of MRSA.
consider’.26 Medical journals do not have a significant impact

Table IV Newspaper articles identified on 30 key dates (pertaining to publication of Department of Health or
journal articles) from 1990 to 2004 containing the term ‘MRSA’ by reported solutions of MRSA
Reported solutions of MRSA No. %
Cleaner wards/hospital 99 38.1
Other (e.g. research, private healthcare, products, suing) 42 16.2
Matron/inspections 37 14.2
Cleaning product (or self-cleaning product) 18 6.9
New drug 15 5.8
Hospital guidelines 14 5.4
Patients asking staff to wash hands 10 3.8
Isolation units 7 2.7
Reduce drug use 6 2.3
Related to pets 5 1.9
Ringfencing 3 1.2
Screening for MRSA 2 0.8
Publishing MRSA rates 2 0.8
Total 260
MRSA, meticillin-resistant Staphylococcus aureus.
406 T. Boyce et al.

on how the MRSA story is reported.2,25 In the time refutations. Unfortunately health professionals
frame we examined, the coverage of MRSA demon- did not see the value of engaging with the media
strates that the media can drive policy away from to ensure public understanding until it was too
scientific evidence and towards popular, common- late, when vaccination rates had fallen signifi-
sense solutions and continues to do so. Indeed, cantly and mumps and measles returned to schools
media coverage ‘can be an integral part of how across the UK.12 In terms of addressing MRSA and
policy is formed and re-formed as well as how it all HAIs, as a core aspect of patient safety and
is packaged in reports and then framed by press quality of care it is not too late to engage with
releases’.27 For example, at the UK Labour Party the media and the public, and ultimately, to influ-
conference in September 2007, the Secretary of ence policy.8
State for Health, Alan Johnson, announced that
each hospital would undergo a ‘deep clean’. But
are the media the only ones to blame? Medical
Conflict of interest
journals are also responsible for failing to influ-
None declared.
ence media coverage e less than half of the
medical journals promoted their research with
Funding sources
a press release. The combination of key govern-
The authors acknowledge the support from the
ment sources promoting cleaning as a solution
National Institute for Health Research (NIHR)
and scientific sources not promoting their own
Biomedical Research Centre Funding Scheme at
peer-reviewed research resulted in the story being
Imperial College and support from the Centre
reported in a particular way.
for Patient Safety and Service Quality (CPSSQ)
Health professionals might also argue that the
funded by the NIHR.
UK government’s agenda is incorrect or that MRSA
is being unnecessarily politicised. They might
believe that the problem is more about poor
management or lack of resources and therefore References
feel that contributing their opinions to the media
1. Jolley S. Assessing patients’ knowledge and fears about
is redundant because the problem is not rooted in MRSA infection. Nurs Times 2008;104:32e33.
‘medicine’ or ‘science’. There is little published 2. Anonymous. MRSA: Moving beyond mops and matrons.
research to verify these views, although lack of Lancet 2007;370:362.
training has been identified as the reason that 3. Reacher MH, Shah A, Livermore DM, et al. Bacteraemia and
scientists failed to participate in media cover- antibiotic resistance of its pathogens reported in England
and Wales between 1990 and 1998: trend analysis. Br Med
age.28 If health professionals fail to engage with J 2000;320:213e216.
the media or challenge the frames that the gov- 4. Grundmann H, Ires-de-Sousa M, Boyce J, Tiemersma E.
ernment or more ‘media-savvy’ sources try to Emergence and resurgence of methicillin-resistant Staphy-
set, then they are failing to participate in a very lococcus aureus as a public-health threat. Lancet 2006;
significant way in which the public learn and 368:874e885.
5. National Audit Office. Improving patient care by reducing
inform themselves about science and health is- the risk of hospital acquired infection: a progress report.
sues. The media are more influential and impor- Report No: HC876:3. London: Stationery Office; 2004.
tant when reporting new scientific stories, such 6. Miller D, Kitzinger J, Beharrell P. The circuit of mass com-
as MRSA.12,29,30 munication: media strategies, representation and audience
This is a health story which receives a great reception in the AIDS crisis. London: Sage Publications;
1998. pp. 128e149.
deal of coverage and can be regarded as an 7. Entwistle V, Sheldon T. The picture of health? Media cover-
opportunity for scientists and health professionals age of the health service. In: Franklin B, editor. Social
to engage with the public, to challenge the overt policy, the media and misrepresentation. 1st ed. London:
politicisation of MRSA, to promote basic knowl- Routledge; 1999. p. 118e134.
edge of infection and risk and a more comprehen- 8. Holmes AH. Can organisational change reduce hospital-
acquired infections? J Hosp Infect 2007;65:191e192.
sive understanding of HAIs and the threat of 9. Lawson E, Price C. The psychology of change management.
antibiotic resistance. At the same time, this is McKinsey Q 2003;2:30e41.
a story that recognises the ability of the media to 10. Health Protection Agency. Health protection report,
drive change and improvement regardless of sci- September 19. Wkly Rep 2008;2:38.
entific evidence. Some lessons can be learned 11. Williams C, Kitzinger J, Henderson L. Envisaging the embryo
in stem cell research: discursive strategies and media re-
from the MMR (measlesemumpserubella) vaccine porting of the ethical debates. Sociol Health Illn 2003;25:
story where many doctors did not get involved 793e814.
because they believed that the science was in- 12. Boyce T. Health, risk and news: the MMR vaccine and the
correct and thus did not require serious media. London: Peter Lang; 2007. pp. 186e188.
UK media coverage of MRSA 407

13. Levine D. Slow response to vancomycin or vancomycin plus 21. Pittet D, Hugonnet S, Harbarth S, et al. Effectiveness of
rifampin in methicillin-resistant Staphylococcus aureus a hospital-wide programme to improve compliance with
endocarditis. Ann Intern Med 1991;115:674e680. hand hygiene. Lancet 2000;356:1307e1312.
14. Panlilio A. Methicillin-resistant Staphylococcus aureus in 22. Kuroda M, Ohta T, Uchiyama I, et al. Whole genome
United States hospitals, 1975e1991. Infect Control Hosp sequencing of methicillin-resistant Staphylococcus aureus.
Epidemiol 1992;13:582e586. Lancet 2001;357:1225e1240.
15. Kreiswirth B. Evidence for a clonal origin of methicillin 23. Byrne C. Doctor who takes on flesh-eating bugs and the
resistance in Staphylococcus aureus. Science 1993;259: Almighty. The Independent March 2006;30:7.
227e230. 24. Henderson L, Kitzinger J. The human drama of genetics:
16. Mulligan M, Murray-Leisure KA, Ribner BS, et al. Methicillin- ‘hard’ and ‘soft’ media representations of inherited breast
resistant Staphylococcus aureus e a consensus review of cancer. Sociol Health Illn 1999;21:560e578.
the microbiology, pathogenesis, and epidemiology with im- 25. Green D. Does hospital cleanliness correlate with meticillin-
plications for prevention and management. Am J Med 1993; resistant Staphylococcus aureus bacteraemia rates? J Hosp
94:313e328. Infect 2006;64:184e186.
17. Voss A, Milatovic D, Wallrauch-Schwarz C, Rosdahl V, 26. Brown JD, Walsh-Childers K. Effects of media on personal
Braveny I. Methicillin-resistant Staphylococcus aureus in and public health. In: Bryant J, Zillmann D, Oliver MB, edi-
Europe. Eur J Clin Microbiol Infect Dis 1994;13:50e55. tors. Media effects: advances in theory and research. 2nd
18. Hiramatsu K, Aritaka N, Hanaki H, et al. Dissemination in ed. London: Routledge; 2002. p. 453e488.
Japanese hospitals of strains of Staphylococcus aureus 27. Davidson R. Radical blueprint for social change? Media
heterogeneously resistant to vancomycin. Lancet 1997; representations of new labour’s policies on public health.
350:1670e1673. Sociol Health Illn 2003;25:532e552.
19. Herold BC, Immergluck LC, Maranan MC, et al. Community- 28. Gascoigne T, Metcalfe J. Incentives and impediments to
acquired methicillin-resistant Staphylococcus aureus in scientists communicating through the media. Sci Comm
children with no identified predisposing risk. J Am Med 1997;18:265e282.
Assoc 1998;279:593e598. 29. Kitzinger J. Framing abuse: media influence and public
20. Enright MC, Day NPJ, Davies CE, Peacock SJ, Spratt BG. understandings of sexual violence against children. London:
Multi-locus sequence typing for characterization of meth- Pluto; 2004. pp. 30e39.
icillin-resistant and methicillin-susceptible clones of 30. Hornig-Priest S. Structuring public debate on biotechnology
Staphylococcus aureus. J Clin Microbiol 2000;38: media frames and public response. Sci Comm 1994;16:
1008e1015. 166e179.
Journal of Hospital Infection (2009) 73, 408e413
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Are national targets the right way to improve


infection control practice?
M. Millar*

Barts and The London NHS Trust, Pathology and Pharmacy Building, 80 Newark Street,
London E1 2ES, UK

Available online 21 August 2009

KEYWORDS Summary The ‘right way to improve infection control practice’ should be
Healthcare-acquired cost-effective and lead to a fair distribution of infection control resources.
infection; Cost-effectiveness is a measure of aggregate ‘good’, and fairness empha-
Risk perception;
sises similar treatment for individuals under similar circumstances. The
Targets
UK national meticillin-resistant Staphylococcus aureus (MRSA) bloodstream
infection (BSI) target encourages National Health Service trusts to
prioritise strategies aimed at MRSA BSI prevention. Under resource-limited
conditions, the MRSA BSI target inevitably encourages deprioritisation of
patients at risk of non-target healthcare-associated infection (HCAI), some
of which are associated with an equal or larger burden of adverse outcome.
Established healthcare improvement strategies, such as the Plan, Do,
Study, Act (PDSA) cycle advocated by the Health Foundation, require the
setting of aims (or targets). If we are to improve infection control practice
then we need to decide on what to measure, how to measure it, and what
the improvement (target) should be. In selecting targets for infection pre-
vention, account should be taken of the contribution of HCAI to adverse
health outcomes overall. Human risk compensation behaviour and
microbial adaptation may both counteract the overall benefit of infection
targets isolated from overall outcomes. Risk taking is part of a healthy
healthcare system. We must be careful not to isolate HCAI outcomes from
overall outcomes or to isolate ‘risk takers’ from ‘risk controllers’. We must
try to limit the scope for human risk compensation and we must watch out
for microbial adaptation. Targets should be set locally, taking account of
fairness and cost-effectiveness. Locally relevant information is key;
positive incentives work best.
ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights
reserved.

* Tel.: þ44 020 3246 0296; fax: þ44 020 3246 0325.
E-mail address: michael.millar@bartsandthelondon.nhs.uk

0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.02.022
National targets for infection control 409

Introduction covering the period 2008/9 to 2010/11.8 Two


targets are specified: (i) the annual numbers of
Leatherman and Sutherland trace the use of man- MRSA bacteraemias should be <50% of the 2003/4
agement targets back to the work of Drucker and level for each year covered by the agreement
the development of the goal-setting theory of mo- and (ii) the rates of Clostridium difficile in 2010/11
tivation.1e3 The Health of the Nation places health should be <30% of the 2007/8 number.
targets within the context of the management of
the UK National Health Service.4 Some of the cri-
teria for the selection of targets specified in The
Potential dangers associated with
Health of the Nation include that the area chosen ‘top-down’ targets
should be a major cause of disability or avoidable
death defined using mortality, morbidity and qual- Bevan and Hood state that regulation by targets
ity of life data; that effective interventions are assumes that priorities can be targeted; that the
available; and that the problem is quantifiable. part that can be measured stands for the whole;
Criteria and proposals for the selection of targets and that what is omitted doesn’t matter.9 This per-
were also developed through the 1990s in a number spective applies generally to targets but has par-
of other countries, for example Australia.5 ticular resonance for the infection targets which
Benefits of health targets include the consolida- focus on a very limited range of infections and
tion of higher level strategic objectives into local at-risk populations. Millar et al. argue that the cur-
policy and practice, consensus building (by opening rent UK National Health Service (NHS) MRSA bac-
dialogue about priorities and expectations), facili- teraemia target forces NHS trusts to prioritise
tation of the monitoring of progress (by identifying control of MRSA bacteraemia, which accounts for
target measures), and the provision of a basis for a small proportion (2%) of NHS HCAI.10 NHS trusts
performance contracting and incentives. In review- are inevitably resource-limited, so the MRSA target
ing the evidence that regulation has improved the inevitably leads to deprioritisation of patients at
quality of healthcare, Leatherman and Sutherland risk of other types of HCAI. In that sense the tar-
point out that attainment of targets has been gets are not fair and, by deprioritising, the larger
greatest in areas amenable to central command proportion of non-target HCAIs may also not be
and control performance management, but ‘less cost-effective. A recent paper from the USA
successful in those areas where intersectoral policy describes the focusing of resources on a single
and actions are needed or where complex social antibiotic-resistant pathogen (such as MRSA) as
behavioural change is required’.6 One of the prob- an ‘inherently flawed approach’ and goes on to
lems with assessment of the impact of health tar- show that resources would be better spent on con-
gets is that target measures do not necessarily trol of bloodstream infections overall rather than
provide information on the impact of the target MRSA bloodstream infections if we are trying to
overall. This point is taken up in a systematic review maximise numbers of lives saved for a given expen-
undertaken on behalf of the Health Foundation en- diture.11 Some additional concerns relating to
titled ‘Does public release of performance results national infection targets are that:
improve quality of care?’7 This report concludes
that public release of information stimulates e they distract focus and constrain flexibility in
change at the level of the hospital but that there response to non-target and emerging
is little evidence that public release improves pa- infections;
tient outcomes and that it may cause consumers e infection outcomes are dissociated from other
(on occasions) to make decisions that are inconsis- (arguably more important) patient outcomes;
tent with their own health goals. This discussion is e if they are too narrowly focused, they leave
primarily focused on how targets are selected, but scope for human compensation behaviour and
it is important to remember that the way in which for microbial adaptation.
targets that have been selected are then used is
another important consideration. It is almost impossible to predict future trends
in infectious diseases. This is well illustrated by
the over-optimism about the control of infectious
Current infection targets diseases during the 1960s, epitomised in the
statement by William Stewart, US Secretary
Current UK National Health Service Healthcare General in 1967, ‘It is time to close the book on
Associated Infection (HCAI) targets are specified infectious diseases.’ What we do know is that
in the Better Care for All Public Service agreement agents of infection are adaptable, that future
410 M. Millar

trends in infectious diseases are likely to have are to ensure that patients are cared for in a clean
a significant impact on humans, but that we cannot environment where the risks of healthcare associ-
predict why, where and in what form; so alertness ated infection are kept as low as possible’.16 For
and ability to respond flexibly are key to the future the risks of infection to be kept as low as possible
control of infectious diseases and in this respect would require that we do not take risks, imposing
narrowly focused targets are associated with many constraints and limits to the real and poten-
a potential danger of distracting alertness to new tial benefits of risk taking, such as might be associ-
and emerging infectious diseases. ated with the everyday activities of NHS operating
A narrowly focused infection target could be theatres.
achieved while the overall outcomes remain un- Individual perception of danger is a major
changed or deteriorate, so there are also potential psychological determinant of our response to
dangers associated with the dissociation of in- risks.12,13,17 Adams has done much to develop the
fection outcomes from overall outcomes. Arguably ideas and arguments underpinning our understand-
preventable infection is important through the ing of risk compensation and describes the rela-
contribution that preventable infection makes to tionship between perceived danger, rewards from
overall outcomes. taking risks, adverse events from taking risks,
and our propensity to take risks (see Figure 1).
It is important that those on the front line who
Risk perception and risk compensation provide healthcare benefits (such as those involved
behaviour in surgery) share the goals and objectives of those
whose jobs it is to work to minimise the risks of
Both individuals and institutions have to balance infection (infection control professionals). In prac-
risks and benefits, and there have been many tice, risk controllers (such as infection control pro-
studies which describe individual and institutional fessionals) tend to be geographically, managerially
behaviour in response to perceived risks and and operationally separated from the risk takers who
benefits associated with particular areas of provide healthcare. Insistence on the primacy of
activity.12,13 That there are benefits as well as risks overall outcomes (rather than narrowly focused in-
associated with healthcare activities is acknowl- fection outcomes) has the potential to facilitate the
edged within recent NHS guidelines and policy development of common goals between the ‘risk
statements; for example, the NHS Operating takers’ who provide healthcare and the ‘risk control-
Framework 2008/9 in section 2.16 states that ‘no lers’ who manage (infection) risks. Infection targets
healthcare system can be entirely risk-free’ and potentially bring conflict to this relationship by
‘Best Practice Risk’ describes ‘an aspiration emphasising narrowly focused infection targets
towards positive risk management which takes rather than overall outcomes for patients.
account of the beneficial as well as harmful con-
sequences of taking risks’.14,15 Even though the
concept that risks and benefits are associated Risk perception
with risk-taking behaviour is established in the
NHS literature, nevertheless the Health Act 2006 Slovic reports primary research and draws together
Part 2 states that ‘managers in NHS organisations much of the literature on the perception of risk.12

Propensity to take risks Rewards (from taking risks)

Balancing behaviour

Perception of risk of
adverse event Experience of adverse event(s)
(such as infection)
‘Cultural filter’

Figure 1 Balancing risks and benefits (modified from p. 15 of Adams13).


National targets for infection control 411

Several studies have quantified the relative risks specially designed children’s seats for cars, cycling
and benefits of different types of activity. For helmets and speed cameras. Risk compensation
example, risks and benefits can be quantified for has also been described as the Achilles’ heel of
motor vehicle driving by looking at how much we innovations in human immunodeficiency virus (HIV)
spend on motor vehicles and the risk of death asso- prevention. For example, improving treatment of
ciated with motor vehicles, with surgery, and with HIV reduces people’s perception of risk and may
vaccinations. Another way of quantifying percep- have the adverse consequence of leading to more
tions of risk is to ask people to rank risks and risk of HIV acquisition.18
benefits associated with particular activities. Gen- What perceptions are associated with the HCAI
erally speaking, perceptions of risks and benefits targets that might lead to risk compensation? Some
tend to show some positive correlation. Where relevant perceptions are given in Table II.
there is a lot of benefit associated with a particular Most of those with experience of the manage-
activity, people generally are prepared to carry ment of patients with infection are aware of the
a larger degree of risk than when there is less ben- potential for microbial adaptation, but there are
efit. Some of the findings from research on risk also dangers associated with separating infection
perception are shown in Table I. outcomes from overall outcomes because of
Risk perception is a major determinant of human risk compensation behaviour. Humans can
human risk behaviour; people often misperceive and often do change their behaviour in response to
risks, and the perception of the scale of risk can be safety measures, including (probably) HCAI control
amplified by social factors such as the media. That interventions.
the perception of risk is an important determinant
of human behaviour is well illustrated by a graph of
road accident deaths in Sweden in the 1960s.13 In Have things really got better with
September 1967 the laws were changed concerning respect to HCAI overall?
the side of the road on which people were allowed
to drive, and in association with this change there Many millions of pounds have been spent in the UK
was a dramatic decline in road accident deaths. In- on the control of target infections since 2000. If we
dividual motor vehicle drivers perceived a risk that accept that microbes can adapt to changes in human
others might forget which side of the road to drive behaviour and activities, and that humans can
on, so they changed their own behaviour e resulting change their behaviour in response to perceptions
in a dramatic reduction in road deaths. of risk, then we might also ask whether the NHS
Risk compensation occurs when people react to infection targets have produced an overall benefit.
a safety initiative by acting less safely e The National Prevalence Study of HCAI showed
frequently because of a change in the perception that in 1993/94, 9% of inpatients at a given time
of danger (risk of an adverse consequence). The were under treatment for an HCAI and that this
question arises: if we have narrowly focused in- figure had fallen to 8.2% in 2006.19 It is difficult to
fection targets, then does the human response to interpret this information because the duration of
those targets generate compensatory increases in hospital stay has steadily reduced over the last ten
risk outside of the target area(s)? There are many years, so it is not clear that the differences in
instances where safety interventions have not prevalence and in the results from different types
generated the expected benefits, for example of hospital could not be accounted for simply by

Table II Targets and perception of risk


Table I Risk perception
Perceptions:
e Risk perception is a major determinant of human
‘risk’ behaviour e the infection targets are the main priority
e Individuals have a perception of risk associated e failure to achieve the target is a threat (to the
with a certain activity (healthcare) or lifestyle individual and/or institution)
e Individuals accept a certain level of risk and e non-target infections do not matter (on a personal
benefit associated with a certain activity (health- or institutional level)
care) or lifestyle e HCAI is less of a problem (now) because HCAI
e Perception of the scale of risks can be amplified by control is on target
social factors such as the media e ‘others’ are doing something about HCAI
e The scale of risk may be misperceived HCAI, healthcare-associated infection.
412 M. Millar

changes in the denominator e those patients who UK Health Protection Agency data for 2003e2007
stay in hospital long enough to acquire a hospi- show an increase in the reports of bacteraemia
tal-acquired infection. The prevalence studies caused by Klebsiella and Enterobacter spp. against
have been based on all of the inpatients present a decline in the numbers of MRSA bacteraemias.
on the day of the study, whereas it is known that There were w4500 Klebsiella spp. bacteraemia
only a small and decreasing proportion of patients reports to the HPA in 2003 and >6000 in 2007
admitted to hospital stay for >24 h. For example, (>30% increase). This compares with 7698 MRSA
in one London teaching trust, of the 100 000 admis- bacteraemia reports between April 2003 and March
sions per year, 52 000 stay for less than a day, 2004 and 4926 reports in 2007 (36% reduction). That
15 000 stay for 24e48 h and only 9000 stay for the targets have led to changes in human behaviour
>48 h. Between 1995/6 and 2003/4 the mean is well illustrated by the observation that most
length of stay for NHS patients fell from 10 days trusts have an active process of root cause analysis
to 7.1 days. It is hard to know therefore if the for MRSA bloodstream infections; on the other
small reduction in prevalence of HCAI is attribut- hand, few NHS trusts routinely carry out root cause
able to improvements in HCAI prevention or in- analyses for other causes of bacteraemia.
stead reflects changes in the denominator
(resulting from shorter hospital stay).
Problems with reporting bias also make it diffi- How do we improve infection control
cult to assess whether HCAIs are being better practice?
controlled now than before the instigation of
targets. Although MRSA deaths mentioned on death If we are to improve infection control practice
certificates between 1993 and 2006 increased then we need to decide what to measure, how to
dramatically, this may reflect reporting bias. measure it and the scale of the improvement that
Unpublished data (derived from London teaching we are looking for. The national objective is to
trusts) suggest that MRSA bloodstream infection improve people’s overall life expectancy and
rates fell between 1998 and 2008, while rates of reduce health inequalities.14 I would argue that
bloodstream infection with other types of bacteria the objective of healthcare is to improve the
have been increasing. In one London teaching quality of life of patients, and that the objective
hospital, increases in E. coli bacteraemia have bal- of infection control professionals is to control
anced the decreases in MRSA bloodstream infection infection risks in a manner that best serves to
(Figure 2) during that period, so overall the rates of improve the overall well-being and interests
bacteraemia caused by significant pathogens (including quality of life) of patients. Some
(E. coli, Klebsiella spp., Pseudomonas aeruginosa, proposed ideal properties of infection targets
meticillin-sensitive and -resistant Staphylococcus are given in Table III.
aureus) are largely unchanged. Assurance of the quality of HCAI control
requires measures of both outcomes and process.
90
80
70
Table III Ideal properties of infection targets
Patient episodes

60
e Relevant (to local priorities)
50
e Transparent to stakeholders
40 e Fair (do not add to inequalities in health outcomes
30 or resource distribution)
20 e Linked with cost-effective interventions/actions
10 e Measurable (reliable data retrieval, definable
parameters)
0
e Sensitive to changes in local policy and practice
99

02

05

08
19

20

20

20

e Do not separate ‘risk takers’ from ‘risk controllers’


Quarter
e Take account of the possibility of risk compensation
Figure 2 Changes in the frequency of Escherichia coli e Do not constrain alertness and responsiveness to
and Staphylococcus aureus bacteraemia in a London non-target HCAIs
Teaching Hospital 1999e2008. Solid line: MRSA þ MSSA;
HCAI, healthcare-associated infection.
dashed line: E. coli.
National targets for infection control 413

HCAI varies from place to place and the contribu-


tion or otherwise of HCAI to overall outcomes also Conflict of interest statement
varies. HCAI performance can only be assessed None declared.
within a specific context, taking account of the
benefits as well as the risks of various healthcare Funding sources
activities. Local targets encourage local owner- None.
ship, have the potential to encourage (if selected
on the basis of overall outcomes) engagement of
‘risk takers’ with ‘risk controllers’, and are more References
likely to be locally relevant. An ethical argument
for local decision-making with respect to health- 1. Leatherman S, Sutherland K. The quest for quality: refining
the NHS reforms (1998e2008). London: The Nuffield Trust;
care resource allocation is made by Ashcroft.20 The 2008.
Health Foundation states that the right targets, 2. Drucker P. Management by objectives. New York: Harper
used in an appropriate way, can improve health Collins; 1993 (first published 1954).
quality and that it is important that systems and 3. Mitchell TR, Daniels D. Motivation. In: Borman WE, Ilgen DR,
services are designed in a way that ensures achiev- Klimoski RJ, editors. Handbook of psychology. Industrial
organizational psychology, vol. 12. New York: Wiley & Sons;
ing targets in one area does not lead to worsening 2003.
standards in others.21 Our Safer Patient Initiative 4. Anonymous. The health of the nation e a strategy for
demonstrates how much can be achieved when England. London: Department of Health; 1992.
hospitals set their own targets and are given the 5. Nutbeam D, Wise M, Bauman A, Harris E, Leeder S. Goals and
right technical support. targets for Australia’s health in the year 2000 and beyond.
Canberra: Australian Government Publishing Service; 1993.
The Plan, Do, Study, Act plan cycles proposed by 6. Sutherland K, Leatherman S. Regulation and quality
the Health Foundation are a simple approach that improvement: a review of the evidence. London: The
anyone can apply. They reduce risk by starting Health Foundation; 2006.
small; they can be used to plan, develop and 7. Shekelle PG, Yee-Wei L, Mattke S, Damberg C. Does public
implement change; and they are highly effective. release of performance results improve quality of care?
London: The Health Foundation; 2008.
Any government has responsibility to ensure 8. Anonymous. Better care for all public service agreement
that taxpayers’ money is well used, but the 2008/9e2010/11 objectives indicators. London: UK Depart-
question is: ‘Are national targets the best way to ment of Health Strategic Framework; July 2008.
ensure that taxpayers’ money is well used?’ I have 9. Bevan G, Hood C. Have targets improved performance in
argued that there are potential pitfalls with the English NHS? Br Med J 2006;332:419e422.
10. Millar M, Coast J, Ashcroft R. Are meticillin-resistant Staph-
national infection (control) targets. Wilde, who ylococcus aureus bloodstream infection targets fair to those
has done much to develop our understanding of with other types of healthcare associated infection or cost-
human psychology with respect to risk compensa- effective? J Hosp Infect 2008;69:1e5.
tion, describes four tactics to improve risk man- 11. Wenzel RP, Bearman G, Edmond MB. Screening for MRSA:
agement, and these are based on perceptions of a flawed hospital infection control intervention. Infect
Control Hosp Epidemiol 2008;29:1012e1018.
risk.17 He suggests that we can increase the per- 12. Slovic P. The perception of risk. Virgina: Earthscan; 2000.
ceived benefit of cautious behaviour, decrease the 13. Adams J. Risk. London: Routledge; 2001.
perceived cost of cautious behaviour, increase 14. Anonymous. NHS operating framework 2008/9. London:
the perceived cost of risky behaviour and decrease Department of Health; 2007.
the perceived benefit of risky behaviour. He states 15. Anonymous. Best practice in managing risk. London:
Department of Health; June 2007.
that, of all the accident countermeasures that are 16. Anonymous. The Health Act 2006. Code of practice for the
currently available, those affecting people’s moti- prevention and control of healthcare associated infections.
vation seem to be most successful, and those that London: Department of Health; January 2008.
reward people for accident-free performance 17. Wilde GJS. Target risk 2: a new psychology of safety and
seem to be the most promising. health. Toronto: PDE Publications; 2001.
18. Cassell MM, Halperin DT, Shelton JD, Stanton D. Risk
In conclusion, I have argued that risk taking is compensation: the Achilles’ heel of innovations in HIV
part of a healthy healthcare system, that we must prevention. Br Med J 2006;332:605e607.
be careful not to isolate HCAI outcomes from 19. Smythe ETM, McIlvenny G, Enstone JE, et al. Four country
overall outcomes or to isolate ‘risk takers’ from healthcare associated infection prevalence study 2006:
‘risk controllers’. We must try to limit the scope overview of the results. J Hosp Infect 2008;69:230e248.
20. Ashcroft R. Fair rationing is essentially local: an argument
for risk compensation and we must watch out for for postcode prescribing. Health Care Anal 2006;14:
microbial adaptation. Targets should be set 135e144.
locally, taking account of fairness and cost- 21. Anonymous. What impact does regulation have on the
effectiveness. Locally relevant information is quality of healthcare? Viewpoint. London: The Health
key, positive incentives work best. Foundation; 2007.
Journal of Hospital Infection (2009) 73, 414e417
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

REVIEW

Responsibility for managing healthcare-associated


infections: where does the buck stop?
B.I. Duerden*

Department of Health, London, UK

Available online 17 September 2009

KEYWORDS Summary The prevention and control of healthcare-associated infections


Bacteraemia; (HCAIs) requires a tripartite partnership between clinicians and carers,
HCAI; managers and government/Department of Health (DoH) across the whole
Legislation;
health and social care community. Mandatory surveillance of meticillin-
Performance
resistant Staphylococcus aureus bacteraemia and Clostridium difficile in-
management;
Surveillance fection has shown a significant fall from peak numbers in 2003/04 and
2006, respectively, and there is now a zero tolerance approach to prevent-
able infections and poor practice. Success so far has been based on senior
management commitment, enhanced real-time surveillance, implementa-
tion of clinical protocols (high impact interventions, prudent prescribing),
improved hand hygiene and environmental cleaning, and training and
audit, backed up by a heightened performance management focus through
targets and legislation (Code of Practice). DoH improvement teams have
supported National Health Service trusts in implementing change. Respon-
sibility for managing HCAI is a combination of managerial responsibility
based upon compliance assurance that procedures and protocols are being
implemented and personal professional responsibility of all clinicians and
other healthcare workers.
Crown Copyright ª 2009 Published by Elsevier Ltd on behalf of The Hospital
Infection Society. All rights reserved.

Infection is different from all other medical spe- and human populations, all of it heavily influenced
cialties e it spreads! It creates a fascinating in- by human behaviour. Infection knows no bound-
terplay of biology between microbial populations aries across health and social care as bacteria travel
with their human hosts and it is the behaviour of
patients and healthcare professionals that deter-
* Address: Department of Health, Wellington House, 133-155
Waterloo Road, London SE1 8UG, UK. Tel.: þ44 0207 972
mines the risk of healthcare-associated infection
4567; fax: þ44 0207 972 1001. (HCAI) and is the key to its prevention and control.
E-mail address: brian.duerden@dh.gsi.gov.uk HCAI affects all health and social care settings
0195-6701/$ - see front matter Crown Copyright ª 2009 Published by Elsevier Ltd on behalf of The Hospital Infection Society. All rights reserved.
doi:10.1016/j.jhin.2009.06.027
HCAI: where does the buck stop? 415

whether in the National Health Service (NHS) or control of HCAI, and for having systems in place to
the independent sector. This is a wide community ensure that it happens; and the government/DH
embracing all parts of the NHS [Department of that sets standards, ensures priority for HCAI
Health (DoH), strategic health authorities with prevention and control, sets targets and monitors
their performance management responsibilities, outcome, and uses the systems of performance
primary care trusts (PCTs) as commissioners and management to ensure that it happens throughout
providers of services, acute NHS trusts and foun- the NHS. Infection prevention and control became
dation trusts providing secondary and tertiary a statutory responsibility in the NHS with the
care, mental health trusts and ambulance trusts] Health Act 2006 which introduced the Code of
and the independent sector (hospitals, indepen- Practice (CoP) for HCAI that applied to all NHS
dent treatment centres and the great majority of bodies.1 Compliance was assessed by the Health-
nursing and care homes). The Health Protection care Commission through its annual health checks,
Agency (HPA) has a key role in supporting infection improvement notices and annual specialist inspec-
prevention and control activity across this wider tions against the Code (commonly known as the
community. ‘Hygiene Code’). The Health and Social Care Act
The HCAI challenge for the NHS in England is 2008 (coming into force in April 2009) built upon
exemplified by the results of mandatory surveil- the 2006 requirements by establishing a single
lance of meticillin-resistant Staphylococcus aureus regulator, the Care Quality Commission, and
(MRSA) bacteraemia (introduced in 2001) and Clos- extending the CoP to the independent sector and
tridium difficile infection (introduced in 2004 for all care settings (independent hospitals, nursing
patients aged >65 years and extended in 2007 to homes and care homes).2 Registration, which is
those aged between 2 and 64 years). The MRSA already a requirement for the independent sector,
bacteraemia reports peaked in 2003/04 at 7700 is extended to all NHS bodies and compliance with
cases and the C. difficile infections were 55 681 the CoP will be a requirement for registration.
in patients aged >65 years in 2006 (which repre- The current DoH HCAI programme was launched
sented 75% of cases). Why had these numbers risen with the Winning ways strategy published in
so high? My personal view is that between 1970 and December 2003.3 In July 2004, the action plan
2000 a dichotomy had developed between infec- Towards cleaner hospitals and lower rates of
tion prevention and control and the modern medical infection was published and this set a target for
practice that had delivered increased life expec- a 50% reduction in MRSA bacteraemias in the NHS
tancy, better treatment for many conditions such in England by 2008.4 The NHS deserves to be con-
as cancer and heart disease, increasingly complex gratulated on achieving this target and the first
surgery and better management of many chronic three-quarters of 2008/09 showed a reduction of
illnesses. Infection was considered to be a nuis- >60% from the quarterly average in 2003/04. The
ance, but we had antibiotics and vaccines. For target and performance management system
those in microbiology and infection control, this backed by legislation has ensured that infection
was a fascinating period in an interesting specialty prevention and control are a top priority, but the
with much to do. New infections were discovered NHS cannot be complacent and must continue to
and described at a rate of one per year in that improve. The next phase of the strategy was set
period, including C. difficile infection in 1978, out in Clean, safe care: reducing infections and
but the subject had become the province of the saving lives in January 2008.5 The target is a ceiling
infection specialists rather than mainstream and individual NHS trusts and the NHS generally
medicine. Reducing HCAI needed a change of the must aim to get as far below it as possible and to
corporate mindset from one which created a sys- maintain year-on-year reductions. This is led by
tem to deliver specialist clinical care and, some- a zero tolerance approach to aim for a minimum
times, as a secondary issue, took measures to number of infections. Zero tolerance does not
prevent infection, to one that creates a safe mean that ‘there will be no infections’ as this is
environment for patient care and then delivers biologically implausible. However, there should
specialist clinical care within that environment. be no tolerance towards preventable or avoidable
Responsibility for HCAI control is a tripartite infections or of poor clinical practice, whether this
partnership between clinicians and carers who are be in hand hygiene compliance, aseptic proce-
responsible for safe patient care and the diagnosis, dures or imprudent antibiotic prescribing. The
treatment, prevention and control of infection in aim is to get things right every time.
their patients; boards, chief executive and A new element of the programme is the decision
managers who are responsible for providing the to screen all patients admitted to NHS hospitals for
corporate environment for good prevention and MRSA colonisation. This requirement applies to all
416 B.I. Duerden

elective admissions (with some exceptions for very prevention and control. The HPA has supported the
low risk procedures) which must be effected by DoH with enhanced surveillance systems for MRSA
April 2009 and extended to all admissions by and C. difficile. At the clinicianepatient interface
2010/11 at the latest. The principle behind the there has been a strong focus on improved clinical
screening programme is that colonisation generally practice protocols through the Saving lives and
precedes infection, and that a colonised patient is Essential steps packages.7,8 These comprise self-
at risk of developing infection themselves and is assessment tools for NHS trusts and primary care
a possible source of transmission to others. If organisations that reflect the CoP core duties,
possible, MRSA-positive patients should be iso- and sets of ‘high impact interventions’ (care bun-
lated, but in any case, a positive screen should dles) for clinical procedures, backed up by several
be followed by decolonisation (alternatively guidance notes. The care bundless help staff to
known as suppressive treatment) to reduce the perform procedures properly, on every occasion,
risk to the individual and the risk of transmission. through sets of headline instructions and a simple
Decolonisation immediately reduces the bioburden audit tool focused on preventing the spread of
of MRSA to cover the period of highest risk and of infection and performing invasive procedures
clinical interventions; it needs to be done as close properly. There has also been a major emphasis
to the clinical treatment as possible. However, it on cleanliness and hygiene. The ‘cleanyourhands’
also needs to be recognised that re-emergence or campaign from the National Patient Safety Agency
recolonisation can occur over a period of weeks or continues to drive improved hand hygiene and
months. An effective screening programme needs there has been a revitalisation of environmental
partnership between hospitals and primary/com- cleaning, particularly for areas where there are
munity care settings to deliver the best service to or have been infected patients.9 All of this has
patients. been backed up by improved training and a height-
A similar partnership is needed to control C. dif- ened performance management focus through the
ficile infection. The unacceptably high levels targets and legislation. Management priority and
reached in 2006 started to come down in 2007/08 responsibility has recognised that HCAI prevention
but there is still much to be done. The government and control is not just the responsibility of the in-
set a target to reduce C. difficile infection by 30% fection control teams but extends from the board
by 2010/11 and the indications are that this will be and chief executive to all staff with clinical owner-
achieved with significant reductions occurring ship being key to delivery. All NHS bodies were
during 2008/09. However, the aim must be to drive required to appoint a director of infection preven-
these numbers down as far as possible, and a new tion and control, a senior person with clinical and
and updated guidance document, Clostridium managerial responsibility and authority who could
difficile infection: how to deal with the problem, ensure that infection prevention and control
was published in January 2009.6 The target is programmes were properly implemented.
based on PCT populations with a subset applied The measurement provided by the enhanced
to the acute hospital settings, so the two must surveillance systems was necessary to manage the
work together to achieve reductions. The numbers improvement programme. A national surveillance
for death certificates recording C. difficile infec- programme provides consistent definitions and is
tion as the underlying cause or a contributory important for performance management and target
factor to deaths reminds us that this is an infection monitoring. Data from enhanced surveillance
with significant mortality, especially in elderly and identifies risk factors. However, the key to local
vulnerable people. In 2007, C. difficile was men- delivery is having local surveillance data provided in
tioned 8324 times on death certificates, with a realistic and timely manner to all wards, units,
4056 (49%) of these being given as the underlying divisions, and specialties so that they can identify
cause. By this stage, the overall number of cases their problem areas, detect outbreaks and episodes
had started to fall but the awareness among of increased incidence, and monitor progress.
doctors of the contribution of C. difficile infection These local data need to be reviewed at all
to deaths was still leading to greater numbers management levels on a regular and frequent basis.
recorded on death certificates. The care bundles focused on key invasive
These improvements have been brought about clinical procedures that carried high risk of in-
by markedly changed practice across all NHS troducing infection e the insertion and care of
bodies from boardroom to ward and clinic. There central venous catheters, peripheral cannulae and
has been a sea-change in high level management renal dialysis catheters, management of surgical
involvement with an emphasis throughout NHS wounds, urinary catheters, patients who are
organisations on the requirements for infection incubated and ventilated, and those who have
HCAI: where does the buck stop? 417

C. difficile infection. The supporting guidance call for all involved in providing NHS services. It is
helps NHS trusts to implement MRSA screening pro- no longer acceptable for infections to be regarded
cedures, ensure that their blood culture protocols as normal. Patients can be very ill, some may die,
were giving reliable results, guided trusts in devel- many will stay in hospital longer and some will
oping their antimicrobial prescribing framework need major surgery as a result of infections. The
and identified the appropriate approaches to isola- treatment of these infections consumes significant
tion and cohorting of infected patients. NHS resources that can be better used. Against this
The programme has been promoted and sup- background, zero tolerance of HCAI is a profes-
ported through DoH improvement teams that have sional obligation for everyone, be they clinicians
visited most acute NHS trusts in England to help and healthcare support staff in all units, wards,
identify areas for action and support the imple- directorates, specialties, executive directors and
mentation of improved practice and procedures. board members in acute NHS trusts, PCTs and
This has shown clearly that the prevention and strategic health authorities, both commissioners
control of HCAI requires continued management and providers and extending to DoH and govern-
attention, including those commissioning and mon- ment. It requires a partnership across the whole of
itoring health and social care services, improved the health and social care community to provide
clinical care, antimicrobial stewardship to support an unbroken chain of care in order to break the
prudent antibiotic prescribing, audit of protocols chain of infection.
and procedures, and training for all health and
social care staff. Making this happen requires
a partnership between management responsibility, Conflict of interest statement
which requires compliance assurance from board None.
to ward, and personal responsibility of all staff
involved, which should be set out in job descrip- Funding sources
tions and job plans, be supported by mandatory This article describes the work of the Depart-
training and continued professional development, ment of Health/NHS HCAI programme of which
be included in appraisal in individual performance the author is Clinical Director. There is no
review, and at times backed up by disciplinary external funding of the programme.
measures if there is continued failure to im-
plement the required measures. The compliance
assurance requires a combination of surveillance
data to identify cases of infection, outbreaks and References
deaths, together with audit results of key activities
1. Health Act 2006eCode of practice for the prevention and
such as hand hygiene, implementation of clinical
control of healthcare associated infections. London: DoH;
protocols, isolation protocols, appropriate anti- 2006.
biotic prescribing and cleanliness inspections. All 2. Health and Social Care Act 2008eCode of practice for the
of these should be reviewed at all management prevention and control of healthcare associated infections.
levels from unit and ward to directorate, division London: DoH; 2009.
3. Department of Health. Winning ways: working together to
and board. Those responsible for commissioning
reduce healthcare associated infection in England. Report
services and monitoring delivery need to include from the Chief Medical Officer. London: DoH; 2003.
a requirement of infection prevention and control 4. Department of Health. Towards cleaner hospitals and
in all their commissioned service and their con- lower rates of infection: a summary of action. London:
tract monitoring should include a review of such DoH; 2004.
5. Department of Health. Clean, safe care: reducing infection
provision in terms of target numbers of infections
and saving lives. London: DoH; 2008.
and process monitoring. 6. Department of Health. Clostridium difficile infection: how
In implementing the HCAI programme the govern- to deal with the problem. London: DoH; 2009.
ment/DoH has used the political imperative, 7. Department of Health. Saving lives: reducing infection,
measurement through surveillance, target setting, delivering clean and safe care. London: DoH; 2007.
8. Department of Health. Essential steps to safe, clean care.
professional support, performance management
London: DoH; 2006.
and legislation, all with the aim of changing human 9. National Patient Safety Agency. Ready, steady, go! The full
behaviour (clinical and managerial) to overcome guide to implementing the cleanyourhands campaign in your
the challenge of HCAI. This has been a wake-up Trust. London: NPSA; 2004.
Journal of Hospital Infection (2009) 73, 418e419
Available online at www.sciencedirect.com

www.elsevierhealth.com/journals/jhin

LETTER TO THE EDITOR

Review of Lancet conference on healthcare- misinformation being published and to keep


associated infections the public better informed. It left me with the opin-
ion that microbiologists and other infection special-
ists should perhaps take time to actively engage
Madam, themselves with the media more often.
The programme discussing specific pathogens
I attended this Lancet conference at the Queen was well chosen e GRE, Pseudomonas aeruginosa,
Elizabeth II Conference Centre in London on 11 ESBL-producing coliforms and norovirus. I picked
and 12 December 2008 as a final year specialist reg- up some interesting facts from each of these; for ex-
istrar in medical microbiology and virology. Before ample, the reasons for the propensity for
attending the conference, I reviewed the pro- Enterococcal Clonal Complex 17 to cause hospital
gramme for the two day conference and saw that infections and the presence of specific virulence
it covered many of the infection control issues faced factors such as enterococcal surface protein, which
by infection control professionals in UK hospitals. is expressed on the cell surface and is responsible
The presentations were relevant to current for biofilm formation; also the fact that ampicillin-
problems in hospitals, such as epidemiology and resistant entercocci were the precedents of VRE in
management of meticillin-resistant Staphylococcus hospitals. The presentation on norovirus by Marion
aureus (MRSA) and Clostridium difficile infection, Koopmans from The Netherlands addressed the
specific pathogens such as norovirus, glycopep- causes for the current epidemics of this virus. Her
tide-resistant enterococci (GRE), extended- data demonstrated that the norovirus GII4 genotype
spectrum b-lactamase-producing coliforms (ESBLs) has dominated since 2002 and she outlined the dif-
and specific clinical problems such as ventilator- ferences in the epidemiology between GII4 and
associated pneumonia (VAP), catheter-related the non-GII4 isolates. The hypothesis proposed for
bloodstream infections (CRBSIs) and surgical site the current prevalence of GII4 and its variants is
infections (SSIs). In addition, the speakers were that GII4 strains differ in their host binding proper-
from various countries in Europe and the USA, which ties and therefore the population segment targeted
I found interesting as this would give a flavour of and level of immunity in that group (which was
what was happening outside the UK. based on molecular epidemiological data and math-
In the first couple of talks e the history and ematical modelling). I would have liked to hear
international epidemiology of healthcare-associ- more on molecular epidemiology and newer
ated infection (HAI) were presented, which I methods of diagnosis and management of some of
thought was a good starting point. The idea of these organisms.
developing a surveillance system in the UK and USA The next category of talks about common HAIs
for multiresistant organisms and information tech- (SSI, VAP and CRBSI) which are burning issues in UK
nology systems that can have an impact on reducing hospitals today was extremely relevant. Particu-
HAI and antimicrobial resistance was useful. The larly useful were the debate regarding the need
next talk entitled ‘International comparison of the for an agreed surveillance methodology allowing
social, media, and political context of HAIs’ was for the complexity of SSI and the data on post-
very different and the speaker pointed out the discharge diagnosis, as well as the current evi-
enormous amount of media interest, particularly in dence for the prevention of VAP and CRBSI.
issues relating to MRSA and C. difficile, and espe- The last set of presentations for the day carried
cially in the UK. There was an interesting debate several important messages regarding basic infec-
on the extent and frequency of doctors’ or scien- tion control practices: the use of alcohol hand
tists’ involvement with the media to prevent rub in developing countries; the concept of
0195-6701/$ - see front matter ª 2009 The Hospital Infection Society. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.jhin.2009.03.029
Letter to the Editor 419

‘hand-touch sites’; and the newer molecular amazed when I heard the talk about the Dutch
methods available for diagnosing specific patho- infection control experience, and although much
gens and identifying antibiotic resistance with of it is unthinkable in the UK or other countries, it
application of these in infection control practice, was fascinating to hear how aggressively MRSA was
e.g. multiplex polymerase chain reaction, IDI managed in The Netherlands.
MRSA assay for screening for MRSA, and finger- On the whole, although there was some repeti-
printing techniques (pulsed-field gel electrophore- tion of material that was presented especially on
sis and variable number tandem repeat typing). epidemiology of MRSA and C. difficile in the UK, and
The latter might better have been placed earlier in although some of the material was probably aimed
the day, perhaps before lunch! at the wider audience which included the media,
Day 2 started with a talk by Brian Duerden entitled administrators, regulators and policymakers as
‘Responsibility for managing HAIs: where does the well as representatives from commerce and indus-
buck stop?’ He presented the UK Department of try, I found this conference both useful and refresh-
Health’s current HAI programme with focus on MRSA ing. The conference was especially helpful to
bacteraemias and C. difficile infection. Key themes trainees planning to take their final Royal College
in his talk were ‘zero tolerance’ and ‘everyone’s of Pathologists exams e certainly the sessions that
responsibility’. This was followed by a session on covered specific pathogens and problems, and all
specific pathogens, and this time the topics were the presentations are freely available on the
hospital- and community-acquired MRSA, C. diffi- conference website (http://www.hai.thelancetcon-
cile and acinetobacter. The mathematical model- ferences.com/App/homepage.cfm?moduleid=4646
ling showing transmission of MRSA between 53&appname=100585).
hospital and community was novel and interesting.
The afternoon session was on controversies. The
talk on the importance of infection control versus
antibiotic manipulation was truly controversial M. Meda
and the contrast was made between ‘infection St George’s Hospital NHS Trust, London, UK
control and firefighting’ and ‘antibiotic stewardship Tel.: þ44 1483 766950.
and prevention’. The difficulties arising from the E-mail address: manjulasudheer@yahoo.com
absence of a standardised acceptable method for
measuring antibiotic use were noteworthy. I was Available online 9 July 2009

You might also like