Download internal medicine journal

Document related concepts

Patient safety wikipedia , lookup

Adherence (medicine) wikipedia , lookup

Multiple sclerosis research wikipedia , lookup

Transcript
Volume 45, Issue 5, May 2015, Pages 471–596
INTERNAL
MEDICINE
JOURNAL
Volume
45
Issue
5
May
2015
ISSN 1444-0903
INTERNAL MEDICINE JOURNAL
Chronic obstructive pulmonary disease
Familial colorectal cancer
Colonoscopy in young women
SNAPSHOT acute coronary syndrome audit
Infection prevention practices in aged-care facilities
5/5/2015 5:16:42 PM
002-imj-v45-i5-ifc.indd 2
5/5/2015 3:15:57 PM
bs_bs_banner
Honorary Advisory Board
Editor-in-Chief
Jeff Szer, Melbourne
Continuing Education
Deputy Editor-in-Chief
Deputy Editor-in-Chief
Zoltan Endre, Sydney
Paul Bridgman, Christchurch
Subspecialty Editors
Cardiology (General)
Paul Bridgman, Christchurch
Infectious Diseases
David Gordon, Adelaide
Cardiology (Arrhythmias)
Andrew McGavigan, Adelaide
Intensive Care
Michael O’Leary, Sydney
Clinical Genetics
Les Sheffield, Melbourne
Internal Medicine
Ian Scott, Brisbane
Clinical Pharmacology
Jenny Martin, Newcastle
Yvonne Bonomo, Melbourne
(Addiction Medicine)
Nephrology
Zoltan Endre, Sydney
Continuing Education
(Clinical Perspectives)
Christopher Pokorny, Sydney
Emergency Medicine
Paul Middleton, Sydney
Endocrinology
Morton Burt, Adelaide
Anthony Russell, Brisbane
Ethics
Paul Komesaroff, Melbourne
Gastroenterology
David M. Russell, Melbourne
Geriatric Medicine
Leon Flicker, Perth
Haematology (General)
Peter Browett, Auckland
Peter Doherty, Melbourne
Kar Neng Lai, Hong Kong
Richard Larkins, Melbourne
Sir Gustav Nossal, Melbourne
Lawrie W. Powell, Brisbane
Nicholas Saunders, Newcastle
John Shine, Sydney
Chorh Chuan Tan, Singapore
Sir David Weatherall, Oxford
Judith Whitworth, Canberra
Editorial Ombudsman
Graham Macdonald, Sydney
Manager
Virginia Savickis, Sydney
Neurology
David Blacker, Perth
Nuclear Medicine
Frederick A. Khafagi, Brisbane
Occupational and
Environmental Medicine; Health
Economics; Editorials Editor
Des Gorman, Auckland
Oncology
Damien Thomson, Brisbane
Palliative Medicine
Janet Hardy, Brisbane
Editorial Assistant
Louise Young-Wilson, Sydney
Previous Editors-in-Chief
Internal Medicine Journal
Edward Byrne (1999–2004)
The Australian and New Zealand
Journal of Medicine
Graham Macdonald (1989–1999)
Michael O'Rourke (1981–1989)
Akos Z. Gyory (1975–1981)
Charles Kerr (1970–1975)
The Australasian Annals of Medicine
Ronald Winton (1957–1970)
Mervyn Archdall (1952–1956)
Public Health Medicine
Mark Ferson, Sydney
Respiratory Medicine
Matthew Naughton, Melbourne
Haemostasis/Thrombosis
Claire McLintock, Auckland
Rheumatology
Peter Youssef, Sydney
Immunology and Allergy
Marianne Empson, Auckland
Sexual Health Medicine
Darren Russell, Cairns
IMJ.JEBDec14
101-imj-v45-i5-jeb.indd fm_i
5/5/2015 3:16:38 PM
Up to 1.0FTE Permanent Staff Specialist
– Centre for Infectious Diseases
and Microbiology
PUSH
BOUNDARIES
Classification: Staff Specialist
Location: Westmead
Employment Status: Permanent Full-Time
Enquiries: Professor Jon Iredell (02) 9845 6012 or
(02) 8627 3410
Email: [email protected]
Reference Number: 12758
Closing Date: 31 May 2015
It is essential for all applicants to contact the Specialist
Medical and Dental Recruitment Unit on (02) 8838 2094 or
email [email protected] for an application
package. All selection criteria must be addressed in your
application. Applications must be received by 5pm on this
date. All applications are to be forwarded to the SMADR
Unit, Locked Bag 7118, Parramatta, BC NSW 2124.
Access this journal and thousands
of other essential resources.
Z02813
Please apply online by visiting:
nswhealth.erecruit.com.au
WILEY ONLINE LIBRARY
Featuring a clean and easy-to-use interface, this online service delivers intuitive navigation,
enhanced discoverability, expanded functionalities, and a range of personalization and
alerting options.
Sign up for content alerts and RSS feeds, access full-text, learn more about the journal,
find related content, export citations, and click through to references.
NSW Health Service: employer of choice
wileyonlinelibrary.com
Save Time and Let the
Research Come to You
9 Be the first to read Early View articles
9 Get notified of Accepted Articles when
they appear online
9 Receive table of contents details each
time a new issue is published
9 Never miss another issue!
wileyonlinelibrary.com
Follow these 3 easy steps to
register for alerts online:
1
2
3
Log into Wiley Online Library. If you are
not already a registered user, you can
create your profile for free.
Select “Get New Content Alerts” from
Journal Tools on the top left menu on any
journal page or visit the Publications page
to view all titles.
Submit your preferences and you are done.
You will now receive an email when a new
issue of the journal publishes.
12 - 4 6 0 6 0
Sign up for new content
ntent alerts
aler
for all of your favorite journals:
bs_bs_banner
aims and scope
The Internal Medicine Journal, formerly known as the
Australian and New Zealand Journal of Medicine, is the
official journal of the Adult Medicine Division of
The Royal Australasian College of Physicians (RACP).
Its purpose is to publish high-quality internationally
competitive peer-reviewed original medical research, both
laboratory and clinical, relating to the study and research
of human disease. Papers will be considered from all areas
of medical practice and science. The Journal also has a
major role in continuing medical education and publishes
review articles relevant to physician education. Except
where otherwise stated, articles are peer reviewed.
abstracting and indexing
This journal is indexed by Abstracts on Hygiene and
Communicable Diseases, AgBiotech News and Information,
AIDS Abstracts, Australian Medical Index, BIOBASE,
Biological Abstracts (BIOSIS), Biomedical Reference
(EBSCO), Cambridge Scientific Abstracts, Chemical
Abstracts Service, Current Contents/Clinical Medicine
(an ISI product), Derwent Biotechnology Abstracts,
EMBASE/Excerpta Medica, Environmental Sciences and
Pollution Management, Health and Safety Science Abstracts
(Online version), Helminthological Abstracts, InPharma
Weekly, International Pharmaceutical Abstracts (IPA),
Journals @ Ovid, MEDLINE, Nutrition Abstracts and
Reviews, Pharmacoeoconomics and Outcomes News,
Reactions Weekly, Science Citation Index, SCOPUS, Tropical
Diseases Bulletin, Vitis-Viticulture and Oenology Abstracts
(Online Edition), World Agricultural Economics and Rural
Sociology Abstracts, and CINAHL.
address for editorial correspondence
Editor-in-Chief, Internal Medicine Journal, The Royal
Australasian College of Physicians, 145 Macquarie Street,
Sydney, NSW 2000, Australia (tel: +61 2 9256 5431; fax:
+61 2 9252 3310). For enquiries regarding ScholarOne
Manuscripts (formerly known as ManuscriptCentral)
submissions please email [email protected]
(e.g. IMJ-0000-2014).
General enquiries should be directed to Virginia Savickis,
the Editorial Office, Internal Medicine Journal, using imj@
racp.edu.au
Comments on published papers are welcomed. Authors
are offered right of reply (no more than 500 words) at the
discretion of the Editor and discussion will not be entered
into. Given the current pressures on editorial space,
however, invited comments are restricted to one reply.
disclaimer
The Publisher, RACP and Editors cannot be held
responsible for errors or any consequences arising from the
use of information contained in this journal; the views and
opinions expressed do not necessarily reflect those of the
Publisher, RACP and Editors, neither does the publication
of advertisements constitute any endorsement by the
Publisher, RACP and Editors of the products advertised.
Copyright © 2015 Royal Australasian College of
Physicians.
For submission instructions, subscription and all other information visit
www.wileyonlinelibrary.com/journal/imj
This journal is available online at Wiley Online Library. Visit www.onlinelibrary.wiley.com to search the articles and
register for table of contents and email alerts.
Wiley’s Corporate Citizenship initiative seeks to address the environmental, social, economic, and ethical challenges
faced in our business and which are important to our diverse stakeholder groups. We have made a long-term commitment
to standardise and improve our efforts around the world to reduce our carbon footprint. Follow our progress at
www.wiley.com/go/citizenship
Access to this journal is available free online within institutions in the developing world through the HINARI initiative
with the WHO. For information, visit www.healthinternetwork.org
ISSN 1444-0903 (Print)
ISSN 1445-5994 (Online)
IMJ.JEBDec14
103-imj-v45-i5-jeb.indd fm_iii
5/5/2015 3:16:40 PM
Share your wealth of knowledge
Submit your manuscript today
Internal Medicine Journal
The Official Journal of the Adult Medicine Division
of The Royal Australasian College of Physicians (RACP)
Internal Medicine Journal actively recruits influential
and topical material in all areas of medical practice and
science for their upcoming publications. You are invited
to submit your original medical research, whether it be
laboratory and clinical, for consideration today.
To view author guidelines visit the Journal homepage at
www.wileyonlinelibrary.com/journal/imj
Edited by: Jeff Szer
Print ISSN: 1444-0903
Online ISSN: 1445-5994
Frequency: Monthly
Impact Factor (2009): 1.786
Submit your manuscript online at:
http://mc.manuscriptcentral.com/imj
bs_bs_banner
May 2015, Volume 45, Issue 5
Editorial
471
510
Domestic violence: it is time for the
medical profession to play its part
L. Piterman, P. A. Komesaroff, H. Piterman
and K. J. Jones
Review
474
C. Susanto and P. S. Thomas
517
Physical activity and sedentary behaviour:
applying lessons to chronic obstructive
pulmonary disease
482
527
Familial colorectal cancer
M. S. Lung, A. H. Trainer, I. Campbell
and L. Lipton
Clinical triage for colonoscopy is useful in
young women
K. D. Williamson, K. Steveling, G. Holtmann,
M. Schoeman and J. M. Andrews
497
Comparison of the management and
in-hospital outcomes of acute coronary
syndrome patients in Australia and New
Zealand: results from the binational
SNAPSHOT acute coronary syndrome 2012
audit
C. Ellis, C. Hammett, I. Ranasinghe, J. French,
T. Briffa, G. Devlin, J. Elliott, J. Lefkovitz,
B. Aliprandi-Costa, C. Astley, J. Redfern,
T. Howell, B. Carr, K. Lintern, S. Bloomer,
A. Farshid, P. Matsis, A. Hamer, M. Williams,
R. Troughton, M. Horsfall, K. Hyun,
G. Gamble, H. White, D. Brieger and D. Chew,
on behalf of Bi-National Acute Coronary
Syndromes (ACS) ‘SNAPSHOT’ Audit Group
105-imj-v45-i5-toc.indd fm_v
Efficacy of non-invasive mechanical
ventilation in the general ward in patients
with chronic obstructive pulmonary disease
admitted for hypercapnic acute respiratory
failure and pH < 7.35: a feasibility pilot study
S. Fiorino, L. Bacchi-Reggiani, E. Detotto,
M. Battilana, E. Borghi, C. Denitto,
C. Dickmans, B. Facchini, R. Moretti, S. Parini,
M. Testi, A. Zamboni, A. Cuppini, L. Pisani
and S. Nava
Original Articles
492
Obvious emphysema on computed
tomography during an acute exacerbation
of chronic obstructive pulmonary disease
predicts a poor prognosis
T. Cheng, H. Y. Wan, Q. J. Cheng, Y. Guo,
Y. R. Qian, L. Fan, Y. Feng, Y. Y. Song,
M. Zhou, Q. Y. Li, G. C. Shi and S. G. Huang
K. Hill, P. A. Gardiner, V. Cavalheri,
S. C. Jenkins and G. N. Healy
Clinical Perspectives
Assessing the use of initial oxygen therapy
in chronic obstructive pulmonary disease
patients: a retrospective audit of prehospital and hospital emergency
management
537
Prevalence and significance of CYP2C19*2
and CYP2C191*7 alleles in a New Zealand
acute coronary syndrome population
P. D. Larsen, L. R. Johnston, A. Holley,
A. C. La Flamme, L. Smyth, E. W. Chua,
M. A. Kennedy and S. A. Harding
546
Safety of coadministration of ezetimibe
and statins in patients with
hypercholesterolaemia: a meta-analysis
L. Luo, X. Yuan, W. Huang, F. Ren, H. Zhu,
Y. Zheng and L. Tang
557
Crescentic glomerulonephritis: data from
the Spanish Glomerulonephritis Registry
B. Quiroga, A. Vega, F. Rivera and J. M.
López-Gómez, on behalf of all members of the
Spanish Registry of Glomerulonephritis
5/5/2015 5:18:25 PM
WE MAKE YOUR RESEARCH EASY.
NOW WE MAKE JOB HUNTING EASY.
Let your partners in research energize your career.
Drawing on our expertise and relationships in the healthcare industry, Wiley-Blackwell invites you
to join Wiley Healthcare Jobs, the definitive job site for healthcare professionals.
đƫ premium jobs from the most respected names in healthcare
đƫ hundreds of healthcare-industry recruiters and employers
đƫ job alerts that match your criteria
đƫ expert career advice and candidate resources
Register and upload your resume/CV now to begin your job search!
Part of
wileyhealthcarejobs.com
bs_bs_banner
May 2015, Volume 45, Issue 5
Position Paper
563
Consensus guidelines for the investigation
and management of encephalitis in adults
and children in Australia and New Zealand
P. N. Britton, K. Eastwood, B. Paterson,
D. N. Durrheim, R. C. Dale, A. C. Cheng,
C. Kenedi, B. J. Brew, J. Burrow, Y. Nagree,
P. Leman, D. W. Smith, K. Read, R. Booy and
C. A. Jones, on behalf of the Australasian
Society of Infectious Diseases (ASID),
Australasian College of Emergency Medicine
(ACEM), Australian and New Zealand
Association of Neurologists (ANZAN) and the
Public Health Association of Australia (PHAA)
Brief Communications
576
Survey of infection control and
antimicrobial stewardship practices in
Australian residential aged-care facilities
R. L. Stuart, C. Marshall, E. Orr, N. Bennett,
E. Athan, D. Friedman and M. Reilly, on behalf
of Members of RACRIG (Residential Aged Care
Research Interest Group)
580
Long-term follow up of paediatric liver
transplant recipients: outcomes following
transfer to adult healthcare in New Zealand
R. Harry, C. Fraser-Irwin, S. Mouat, E. Gane,
S. Munn and H. M. Evans
583
588
S. K. Mallipattu, J. Ling and J. Uribarri
General correspondence
589
589
586
Author reply
M. Brown and D. Campbell
590
Leprosy and Australia
C. R. Boughton
590
Not only monoclonal antibodies …
J. C. Nossent
591
Author reply
R. J. Commons, R. Hannah and B. J. Currie
592
Redesign versus resources: continuity lost
C. P. Denaro
593
Author reply
R. Toomath, N. Szecket and P. Poole
594
The true prevalence of diabetes in hospital
patients and its implications
G. Somarajah, H. Karunajeewa, P. S. Hamblin,
E. Karahalios and E. Janus
595
Author reply
J. E. Cromarty, S. Parikh, T. J. Jackson,
W. K. Lim and S. Acharya
Letters to the Editor
Clinical-scientific notes
Audit of inpatient referrals
G. Simpson
Primary central nervous system
posttransplantation lymphoproliferative
disorder after heart and lung transplantation
G. Gifford, K. Fay, A. Jabbour and D. D. Ma
Serum creatinine is not the end-all, be-all
of renal impairment
Bilateral cordotomy post-failure of
intrathecal analgesia in a palliative care
setting
Systemic lupus erythematosus patients
and tertiary specialist care – simple
considerations dropping through the cracks:
osteoporosis monitoring as an example
S. P. M. Hosking, M. E. Franco, P. Poon
and L. William
M. Hew, E. J. McKinnon, B. Kirwin,
O. P. Martinez and M. Lucas
107-imj-v45-i5-toc.indd fm_vii
596
5/5/2015 5:18:26 PM
WE MAKE YOUR RESEARCH EASY.
NOW WE MAKE JOB HUNTING EASY.
Let your partners in research energize your career.
Drawing on our expertise and relationships across the research and business communities,
Wiley-Blackwell invites you to join Wiley Job Network, the definitive job site for professionals
in the sciences, technology, business, finance, healthcare and the arts.
đƫ premium jobs from the most respected names in your industry
đƫ hundreds of recruiters and employers in your field
đƫ job alerts that match your criteria
đƫ expert career advice and candidate resources
Register and upload your resume/CV now to begin your job search!
wileyjobnetwork.com
Internal Medicine Journal 45 (2015)
E D I TO R I A L
Domestic violence: it is time for the medical profession to play
its part
Ignored for a long time, domestic violence is now recognised as one of the major public health issues in Australia. The financial cost of the problem to the
community, estimated at $14.7 billion in 2013, is similar
to that of obesity and far in excess of diabetes.1 However,
beyond the monetary cost the personal and social
damage associated with domestic violence is regarded by
many as sufficient to constitute a national emergency.
Intensifying public interest in the problem is reflected
in the appointment as the 2015 Australian of the Year
of Rosie Batty, a courageous victim of a tragic case of
domestic violence, together with the establishment of a
special Domestic Violence Task Force in Queensland and
a Royal Commission in Victoria. While these initiatives
are most welcome, they will not on their own provide a
solution. What is needed is a national strategy that brings
together diverse groups and individuals from the community around a multidisciplinary programme that
includes information, support, education and research.
Such a programme will require support and resources
from government as well as active participation from the
medical profession.
Despite their potential influence and importance,
medical professionals, with rare exceptions, have been
strangely silent in this area. The Australian Medical Association has had an admirable policy on its books for 10
years and the Royal Australian College of General Practitioners has developed guidelines for family doctors.2
However, many other Colleges – including the Royal
Australasian College of Physicians – appear not even to
have developed policies on the subject, let alone effective
action strategies. It is time that the wider medical profession contributed actively to addressing this scourge on
Australian society.3
Any coordinated strategy on domestic violence will
need to be informed by accurate and reliable data. Unfortunately, such data are, at the present time relatively
limited. It is known that domestic violence extends to a
vast array of abusive settings, covering physical, sexual,
emotional and financial abuse among intimate partners,
same sex couples, elders and children. It crosses all socioeconomic, cultural, ethnic and religious boundaries. The
vast majority of victims are women, with 17% of all
Australian women aged more than 18 years having
experienced violence from a partner at some time, com-
pared with 5.3% for men,4 and that domestic violence is
especially marked in pregnancy, during which up to 36%
of all violence occurs,5 and when up to 20% of women
experience it for the first time.6
More than 65 000 cases of domestic incidents are
reported to police in Victoria each year,7 and nearly
35 000 intervention orders related to family violence are
issued, both of these numbers are rapidly increasing.8
Sadly, even death is a not infrequent outcome, with 185
domestic homicides having been reported across Australia in the 3 years to 2010.9
It is known that, despite its frequent occurrence,
domestic violence is recognised only rarely by medical
practitioners. Indeed, the Bettering the Evaluation and
Care of Health (BEACH) study,10 which monitors patterns
of consultations in general practice, reported that in more
than 95 000 consultations examined in 2013–2014,
domestic violence was never cited as a reason for
encounters by patients or a problem managed by general
practitioners (GP). Similarly, an Irish study of women
attending general practices showed that while 39% had
experienced violent behaviour from their partner, only
12% were questioned about it by their doctors.11
There is much, however, that remains to be explained.
In particular, it is not clear why doctors are doing so
badly. Contributing factors no doubt include a reluctance
of many patients themselves to volunteer that they have
been abused. However, a lack of awareness by practitioners is also important. Sufferers of violence frequently
make contact with GP, emergency medicine physicians,
obstetricians, psychiatrists, specialist physicians or in
other clinical settings. Indeed, full time GP may see up to
five women per week who have experienced partner
violence, of which two are severe.12 Patients may present
with unexplained physical injury, bruising, chronic
fatigue, anxiety, depression, insomnia or undifferentiated
somatic symptoms.13 Although each of these should raise
the possibility of domestic violence, very frequently the
warning signs are missed.
Nor is it well understood why victims are so reluctant
to report abuse and seek help. Power differences within
relationships, especially those between men and women,
and an associated sense of shame among the victims are
likely to be relevant here, perhaps leading women to
believe that the attacks on them were provoked by their
© 2015 Royal Australasian College of Physicians
471
Editorial
own failures as mothers and wives.14 Victims may also
feel that help is hard to obtain, based on the common
perception, that police officers are reluctant to become
involved in domestic violence, seeing the problem as one
of interpersonal conflict outside their legitimate responsibilities.15 The courts have contributed to this sense of a
lack of support by the application until recently of the
so-called ‘doctrine of provocation’ as a formal basis for
excusing violence against women.16 Doctors, frequently
lacking training, skills, confidence and practical resources
to enable them to respond effectively, are all too often
caught up in this sad labyrinth of unconscious collusion.
The final result of all these factors is that doctors mostly
don’t ask and women mostly don’t tell.
It is not known what social and psychological forces
drive some people to commit acts of violence against the
people who are closest to them and who trust them most.
And most importantly of all, evidence is lacking about
what interventions are effective for responding to domestic violence and for preventing its occurrence in the first
place.17
Up until now, attempts by the medical profession to
respond to the problem have largely focused on guidelines to assist in the detection, management and referral
of patients experiencing domestic violence.2,18 It is apparent that more than broad guidelines are needed. Also
required are society-wide approaches that mobilise individuals and groups within the community and draw on a
wide range of resources. The response must incorporate
support for victims to become empowered to speak out
and accept help, and understanding and management of
the underlying problems of those committing the violence. Here, a move away from the traditional approach
based on shame, recrimination and blame to a recognition that domestic violence is usually a symptom of deep
underlying social and psychological pathologies is more
likely than existing strategies to bear fruit.
It is clear from this formulation that there are many
ways in which doctors can help. They can provide safe
trusted spaces in which both the victims and their assailants can express their pain and explore options for
change. They can play a key part in detection, intervention and provision of specialised treatment of the
References
1 KPMG. Cost of Violence against women
and children. White ribbon international
conference. Sydney Australia 13–15 May
2013.
2 The Royal Australian College of General
Practitioners. Abuse and Violence:
physical, mental and emotional damage caused by
domestic violence. They can provide support and, where
appropriate, active treatment for the direct victims of the
violence as well as for all those harmed by being drawn
into its fatal web; this includes the child witnesses of the
violence, who often carry the damaging effects into their
own subsequent relationships. However, they cannot do
all this without a well coordinated, locally driven multidisciplinary team-based approach.
Doctors can participate in much-needed research into
the emotional and social roots of domestic violence, and
development and testing of intervention programmes for
both offenders and victims. The methodologies required
for such research are often complex, but the experience
gained from the study of other major public health problems, such as obesity, will provide a fecund resource.
Above all, they can help – along with many others – in
action to prevent the violence occurring in the first place.
This will involve work to change prevailing assumptions
that narrowly stereotype women and impose unrealistic
demands on all the parties in a relationship. Preventive
work must start early, involve both boys and girls, and
continue throughout school years. The media could play
a positive and ongoing role in promoting awareness of
the nature of the problem .
The contribution of domestic violence to both physical
and mental health problems should be included in undergraduate and postgraduate educational programmes,
including continuing professional development, to
enhance the possibility of effective responses.
Doctors can play an important role in developing and
implementing major changes in the social response to the
problem of domestic violence. It is time this was recognised and it is time they started doing so.
Received 28 February 2015; accepted 8 March 2015.
doi:10.1111/imj.12738
1
2
L. Piterman, P. A. Komesaroff, H. Piterman3 and
K. J. Jones4
1
Berwick and Peninsula Campuses, Monash University, 2Medicine,
Monash University, 3School of Primary Health Care, Monash
University, and 4Office of the Pro-Vice Chancellor, Peninsula
Campus, Monash University, Melbourne, Victoria, Australia
Working with our Patients in General
Practice. 4th edn. Melbourne: RACGP;
2014 [cited 2015 Feb 22]. Available
from URL: www.racgp.org.au/your
-practice/guidelines/whitebook
3 WHO. Responding to intimate partner
violence and sexual violence against
women: WHO clinical and policy
guidelines. Geneva WHO. 2013 [cited
2015 Feb 22]. Available from URL:
http://apps.who.int/iris/bitstream/
10665/85240/1/9789241548595_eng
.pdf?ua=1
4 Australian Bureau of Statistics. Personal
Safety, Australia, 2012. Cat No. 4906.
Canberra: ABS.
© 2015 Royal Australasian College of Physicians
472
Editorial
5 ABS. Personal Safety Survey, 2006. ABS
Cat. No. 4906.0. Canberra: Australian
Bureau of Statistics.
6 Walsh D. The hidden experience of
violence during pregnancy: a study
of 400 pregnant Australian women.
Aust J Prim Health 2008; 14:
97–105.
7 Victoria Police. Family Incident Reports,
Victoria Police Crime Statistics
2013–2014; 2015.
8 Magistrate’s Court of Victoria. Annual
Report 2012/2013, 2013 [cited 2013 Dec
18]. Available from URL: http://www
.magistratescourt.vic.gov.au/sites/
default/files/Default/MCV_Annual
_Report_2012-13.pdf
9 Chan A, Payne J. Homicide in Australia:
2008–2009 to 2009–2010. National
Monitoring Homicide Program annual
report. Canberra. Australian
Government, Australian Institute of
Criminology, 2013.
10 Britt H, Miller GC, Charles J, Henderson
J, Bayram C, Pan Y et al. General
Practice activity in Australia 2013–2014.
11
12
13
14
15
General practice series no. 36. Cat. no.
GEP 36.Canberra: AIHW, 2014.
Bradley F, Smith M, Long J, O’Dowd T.
Reported frequency of domestic
violence: cross sectional survey of
women attending general practice. BMJ
2002; 324: 271–8.
Hegarty K. What is intimate partner
abuse and how common is it? In:
Roberts G, Hegarty K, Feder G, eds.
Intimate Partner Abuse and Health
Professionals: New Approaches to Domestic
Violence. London: Elsevier; 2006; 19–40.
Hegarty K, Gunn J, Chrondos P, Small R.
Association between depression and
abuse by partners of women attending
general practice: descriptive cross sectional survey. BMJ 2004; 328: 612–24.
Australian Institute of Family Studies.
The many facets of shame in intimate
partner sexual violence. 2014 [cited
2014 Nov 24]. Available from URL:
http://www.aifs.gov.au/acssa/pubs/
researchsummary/ressum1/rs1a.html
Fagan J. The Criminalisation of Domestic
Violence: Promises and Limits. National
Institute of Justice Research Report.
Washington, DC: National Institute of
Justice; 1996.
16 The Law Institute of Victoria. Under the
Crimes (Homicide) Act 2005 provocation
as a defence to murder has been
abolished and a new defence, ‘defensive
homicide’, has been created. 2010 [cited
2015 Feb 22]. Available from URL:
http://www.lawreform.vic.gov.au/all
-projects/defences-homicide#sthash
.moy7bzwe.dpuf
17 Breckenridge J, Hamer J. Traversing the
maze of ‘evidence’ and ‘best practice’ in
domestic and family violence service provision
in Australia. Australian Domestic
and Family Violence Clearinghouse;
2014.
18 National Institute for Health and Care
Excellence (NICE). Domestic violence
and abuse: how services can respond
effectively. 2014 [cited 2015 Feb 22].
Available from URL: www.nice.org
.uk/guidance/ph50/chapter/
recommendations
© 2015 Royal Australasian College of Physicians
473
Internal Medicine Journal 45 (2015)
REVIEW
Physical activity and sedentary behaviour: applying lessons to
chronic obstructive pulmonary disease
K. Hill,1–3 P. A. Gardiner,4,5 V. Cavalheri,1,2 S. C. Jenkins1,2,6 and G. N. Healy1,4,7
1
School of Physiotherapy and Exercise Science, Faculty of Health Science, Curtin University, 2Lung Institute of Western Australia and Centre for
Asthma, Allergy and Respiratory Research, University of Western Australia, 3Physiotherapy Department, Royal Perth Hospital, 6Physiotherapy
Department, Sir Charles Gairdner Hospital, Perth, Western Australia, 4School of Population Health, The University of Queensland, 5Mater Research
Institute, The University of Queensland, Brisbane, Queensland, and 7Heart and Diabetes Institute, Baker IDI, Melbourne, Victoria, Australia
Key words
physical activity, sedentary behaviour, chronic
obstructive pulmonary disease.
Correspondence
Kylie Hill, School of Physiotherapy and Exercise
Science, Curtin University, GPO Box U1987,
Perth, WA 6845, Australia.
Email: [email protected]
Received 17 June 2014; accepted 20 August
2014.
doi:10.1111/imj.12570
Abstract
In health and disease, the benefits of regular participation in moderate to vigorous
intensity physical activity are well documented. However, individuals with chronic
conditions, such as those with chronic obstructive pulmonary disease (COPD), typically
do very little activity at a moderate or vigorous intensity. Much of their day is instead
spent in sedentary behaviour, such as sitting or reclining, which requires very little
energy expenditure. This high level of time spent in sedentary behaviour can have
serious health consequences, including increased risk of diabetes, cardiovascular disease
and premature mortality. There is emerging evidence to suggest that participation in
light intensity physical activities (e.g. standing or slow walking) may have benefits for
cardio-metabolic health. Given the low aerobic capacity of individuals with moderate to
severe COPD, increasing light intensity activity (through reducing sedentary time) may
be a feasible additional strategy to improve health in this population, alongside traditional recommendations to increase the time spent in moderate to vigorous intensity
physical activity. This review provides an overview of physical activity and sedentary
behaviour, with a particular emphasis on these behaviours for people with COPD. It
provides suggestions for the measurement of these behaviours within the clinical
setting, as well as for interventions that may be effective at increasing physical activity
and reducing sedentary behaviour in this population.
Introduction
The widespread benefits of regular participation in moderate to vigorous intensity physical activity are well established.1 However, consistent with international data, the
Funding: G. N. Healy was supported by a Heart Foundation [PH
12B 7054] Fellowship and by a NHMRC Centre for Research
Excellence Grant in the Translational Science of Sedentary
Behaviour (APP1041056). P. A. Gardiner was supported by a
NHMRC Center for Research Excellence Grant in Women’s
Health in the 21st Century (APP1000986).
Conflict of interest: All authors presented at the Airways annual
conference in 2013. Travel and accommodation expenses and an
honorarium were provided. G. N. Healy presented at an OERC
(Office Ergonomics Research Committee) meeting in 2013.
OERC covered travel and accommodation expenses and also
provided an honorarium. G. N. Healy also presented at the 2013
‘Juststand Wellness Summit’, a conference organised by
Ergotron. Ergotron covered travel and accommodation
expenses. No further honoraria or imbursements were received.
The funding bodies had no influence on the conduct or the
findings of the study.
majority of Australian adults fail to meet the recommended levels of physical activity to produce health benefits.2 This high level of inactivity contributes significantly
to healthcare costs.3 Recently, there has been a focus on
sedentary behaviour, or too much sitting. Specifically,
there is growing evidence that excessive sedentary time, in
particular time accumulated in uninterrupted bouts of
sedentary behaviour, is associated with adverse health
outcomes.4,5 Individuals with chronic obstructive pulmonary disease (COPD) typically engage in very little physical
activity due to exertional dyspnoea and fatigue. Although
pulmonary rehabilitation, which has a focus on exercise
training, has strong evidence for reducing symptoms,
improving exercise tolerance and quality of life,6 and
reducing healthcare utilisation7 in this patient population,
there is limited evidence that pulmonary rehabilitation
increases daily levels of physical activity and reduces sedentary time.
This review provides an overview of the health benefits
of physical activity across the spectrum, from light intensity through to moderate and vigorous intensity, as well
© 2014 Royal Australasian College of Physicians
474
Activity and sitting: lessons for COPD
Figure 1 An integrated and comprehensive
view of activity and inactivity. Adapted from
http://www.sedentarybehaviour.org/what-issedentary-behaviour.8
as the adverse health effects of too much time spent in
sedentary behaviour. It includes a summary of the
methods used to measure physical activity and sedentary
behaviour in research and clinical settings. Estimates of
time spent in physical activity and sedentary behaviour
by people with COPD are described as well as some direct
and ‘stealth’ interventions that aim to increase physical
activity and reduce sedentary behaviour.
Physical activity: definition
and measurement
Physical activity is defined as any bodily movement generated by skeletal muscle that results in energy expenditure.1 It is often classified as light, moderate or vigorous
intensity, according to the level of energy expenditure
required (Fig. 1).9 Multiple different behaviours fall
under these intensity classifications. For example, light
intensity physical activity would include activities, such
as showering and ironing.10 In contrast, vigorous intensity physical activity would include activities, such as
running and walking up hills.10 Physical activity may also
be classified as activities undertaken as part of daily
living, such as domestic and occupational tasks, or as
exercise, which is a form of physical activity that is
planned, structured and undertaken regularly with the
goal of improving or maintaining fitness (Table 1).1
Obtaining accurate and detailed measures of physical
activity are useful when designing and evaluating interventions to optimise activity levels. Measures of physical
activity can broadly be grouped into subjective (i.e. selfreport) and objective. Subjective measures rely on an
individual’s recall of their activity levels. Although data
obtained through subjective measures, such as questionnaires, may lack precision,11 detailed questioning over
recent time periods has been shown to improve the reliability of the data obtained.12 Subjective measures also
offer the opportunity to obtain detailed information
regarding the type of activities undertaken during daily
life, which allows clinicians to establish targets and goals
regarding participation in physical activity, based on individual preferences. The low cost associated with selfreport measures of physical activity has resulted in their
widespread use in clinical practice and epidemiological
research.
Objective measures involve using a device, commonly
a motion sensor, to capture physical activity. Devices
range in complexity and price. The most basic option is a
pedometer, which records the number of steps taken.
More sophisticated devices may use accelerometry to
measure movement and/or non-invasive physiological
sensors to estimate energy expenditure. The measurement properties of these devices and their output vary
Table 1 Definition of key terms9
Term
Physical activity
Light physical
activity
Moderate to
vigorous
physical activity
MET
Definition
Any bodily movement produced by skeletal
muscles that results in energy expenditure
above resting levels. Physical activity broadly
encompasses exercise, sports and physical
activities done as part of daily living,
occupation, leisure and active transportation.
Activity with a relative intensity of 20% to <40% of
VO2max. For the general population, it has
been defined as activities that have an energy
expenditure of >1.5 to 3 MET. It includes
activities, such as showering and ironing.
Activity with a relative intensity of 40% to <60%
(moderate) or ≥60% (vigorous) of VO2max. For
the general population, it has been defined as
activities that have an energy expenditure ≥ 3
MET. It includes activities, such as brisk walk,
cycling, walking uphill, rowing and running.
An index of energy expenditure. One MET is
equal to an oxygen uptake of 3.5 mL/kg/min,
which is the rate of energy expenditure while
sitting at rest.
MET, metabolic equivalent of tasks; VO2max, maximum rate of oxygen
uptake.
© 2014 Royal Australasian College of Physicians
475
Hill et al.
considerably. Most devices require technical expertise to
collect, download and interpret the data. Nevertheless,
technology in this area is advancing quickly, and it is
likely that the collection of robust physical activity data
through objective methods will be feasible for clinicians
in the near future. Further information on the measurement of physical activity is available elsewhere.13,14
Health effects of moderate to vigorous
physical activity
In adults, the benefits of regular participation in moderate to vigorous intensity physical activity have been well
established and include a reduction in the risk of cardiovascular disease as well as all-cause mortality.1 These
effects are likely to be mediated by several mechanisms,
including production, expression and release of
myokines by the skeletal muscle, improvement in
endothelial function, cardiovascular fitness and insulin
sensitivity, maintenance of a healthy body weight, preservation of fat-free mass and a reduction in circulating
systemic inflammatory biomarkers.1,15 Evidence of health
benefits has resulted in a range of public health messages
designed to promote participation in daily physical activity, with current guidelines from the United States recommending that adults perform a minimum of 150 min
of moderate intensity physical activity or 75 min of vigorous intensity physical activity each week.1 However,
despite the obvious health benefits of an active lifestyle,
31% of adults worldwide do not meet these guidelines
and are considered physically inactive.16 This high level of
inactivity has serious public health and economic consequences, with low levels of physical activity increasing
the risk of developing conditions, such as obesity and
type II diabetes.1 Further, there is evidence to suggest that
low levels of physical activity also play a part in the
development of some cancers, dementia and mood disturbances, such as depression.1 Overall, low levels of
physical activity have been estimated to account for 9%
of premature mortality, or more than 5.3 million deaths
worldwide each year.17
What about time spent in activity
other than moderate to vigorous
physical activity?
To date, much of the public health research and resources
have been targeted towards increasing population levels
of moderate to vigorous intensity activity. However,
on average, adults spend more than 90% of their waking day in activities other than those classified as moderate or vigorous intensity.3 Even if an individual was to
undertake the minimum of 30 min/day of moderate to
vigorous intensity activity specified in public health
guidelines,1 time in this activity intensity would still constitute less than 5% of a typical 16-h waking day. Accordingly, a more comprehensive view of inactivity has
increasingly penetrated research, policy and practice.
This approach considers activities across a spectrum from
sedentary, to light intensity activity to moderate and vigorous, with a focus on understanding the distribution and
health effects across this range of physical activity
(Fig. 1).
Sedentary behaviour: definition
and measurement
On average, the majority (46–59%) of adults spend their
waking hours at the low end of the spectrum, that is, in
sedentary behaviour.3 Sedentary behaviours are defined
both by low energy expenditure (<1.5 metabolic equivalent of tasks) and a sitting or reclining posture.18 They
occur throughout the waking day (i.e. sleep is not considered a sedentary behaviour), and across work, leisure,
domestic and transport domains. Common behaviours
that occur while sedentary include television viewing,
reading, driving, using a computer and playing cards.
Importantly, an individual can be both physically active
(i.e. meet the physical activity guidelines)1 and highly
sedentary; a concept coined ‘the active couch potato’.19
As outlined later, time spent in both physical activity and
sedentary behaviour contributes to health outcomes.
As is the case for physical activity, both subjective and
objective measures can be used to measure sedentary
time. In addition to measuring the total time spent in
sedentary behaviours, measures can also be used to assess
behaviours within individuals and groups, in the context
of the domains in which they occur. To date, self-report
measures of time spent in sedentary behaviour have typically being used, with generally good reliability, but poorto-modest validity.20 More recently, methods, such as past
day recall show improved validity over previous recall
periods, and may be useful for large-scale implementation.21 However, even a simple question, such as ‘in the
last week, how much time per day would you typically
spend sitting down?’ could be useful in a clinical setting
to provide tailored advice and monitor changes over
time.
Objective measures, such as those derived from accelerometers and inclinometers, have also been used to
measure sedentary time. Importantly, these devices
provide date and time stamped data, which enable analysis of not only the total amount of time spent in sedentary
behaviours, but also how and when the sedentary
time was accumulated. Ideally, such measures derive
sedentary time not only from low energy expenditure,
© 2014 Royal Australasian College of Physicians
476
Activity and sitting: lessons for COPD
but also posture in order to distinguish time spent sedentary (low energy, sitting or reclining posture) from
time spent standing (low energy, upright posture).
Postural-based measures, such as the activPAL monitor
(PAL Technologies, Glasgow, UK), have been shown to be
highly accurate compared to direct observation,22 and
their use is becoming more widespread within both intervention and observation research. However, these objective measures do not capture domain or behaviourspecific information; contextual information that is
useful for the development of intervention targets aimed
at individuals and public health messages on how to
reduce sedentary time. Therefore, it is recommended that
a combination of both self-report and objective measures
is used.20
Health impacts of too much sitting
The last decade has seen rapid advances in our understanding of the relationship between time spent in sedentary behaviours and health outcomes. A recent
review reported that those categorised in the most sedentary group, regardless of how it was measured, had
on average, twice the risk of developing type II diabetes
or cardiovascular disease, or of dying from cardiovascular disease, and 1.5 times the risk of dying prematurely
compared to those in the group who were the least sedentary.5 Detrimental associations with excessive sedentary time have also been observed with weight gain,
depressive symptoms, biomarkers of chronic disease risk
(including triglycerides, HDL cholesterol and insulin),
musculoskeletal symptoms, poor quality of life and
chronic kidney disease.23 Notably, although those who
are both inactive and have high sedentary time are at
the highest risk, even in those who met physical activity
guidelines (i.e. are ‘active’), detrimental associations
with sedentary time have been observed.19 This highlights the need to measure both sedentary time and
physical activity within lifestyle assessments. Mechanisms proposed for the associations observed include
the minimal muscular contractions in the large postural
muscles occurring during sitting,24 together with the
lower energy expenditure compared to non-sedentary
behaviours.25
Importantly, it is not just total sedentary time that
appears to be relevant for health, but also the manner in
which it is accumulated. Regularly interrupting sedentary
time, with either light or moderate intensity activity, has
been beneficially associated with biomarkers of chronic
disease.26 Conversely, long, unbroken periods of sitting
have been associated with increased insulin resistance
and poor glycaemic control.4,27 This evidence has
informed the development of national and international
recommendations to minimise the amount of time spent
in prolonged sitting and to break up sitting as often as
possible.1,28 Although sufficient robust evidence regarding ‘how often should we get up?’ is not yet available, a
practical message may be to ‘sit less throughout the day,
and stand up at least every 30 minutes’.
If not sedentary, then what?
The strong negative correlations observed between sedentary time and light intensity physical activity29 suggests
that if we are not sedentary, we are typically undertaking
light intensity activities. This highly heterogenous group
of behaviours includes standing, incidental movement
and slow walking; activities that are difficult to quantify
through self-report measurement tools. Correspondingly,
despite being high volume (on average, 37% to 46% of
adults’ waking hours),3 little is known about the health
effects of behaviours that fall within the light intensity
physical activity spectrum. Nevertheless, associations
observed with light intensity physical activity tend to be
opposite to those demonstrated with sedentary time.30 Of
note, there is preliminary evidence to suggest that there
are cardio-metabolic benefits for those who have a positive light-sedentary balance (i.e. more time is spent in
light intensity physical activity than sedentary), even if
recommended levels of moderate to vigorous intensity
physical activity are not achieved.30 Though it is ideal if
adults have both low sedentary time, and high moderate
to vigorous intensity physical activity time,30 these findings collectively suggest that there may also be benefit
from shifting sedentary time to light intensity activities; a
potentially more feasible and acceptable target for change
especially for those with chronic conditions, such as
COPD.
How are physical activity and sedentary
time affected in people with COPD?
Dyspnoea and fatigue during daily activities are frequently reported by people with COPD and appear to
contribute to the low levels of physical activity undertaken in this population.31 Specifically, there are now
robust data showing that people with COPD participate in
less physical activity when compared with healthy people
of a similar age. One of the first studies reporting this
difference using an objective measure of physical activity
showed that people with COPD spent less time standing
and walking when compared with healthy adults of a
similar age and gender proportion (Fig. 2).32 A review of
11 studies that measured physical activity levels in people
with COPD and healthy controls revealed that the proportion of time people with COPD spent participating in
© 2014 Royal Australasian College of Physicians
477
Hill et al.
Figure 2 Physical activity and sedentary
behaviour of people with chronic obstructive
pulmonary disease (COPD). Adapted from
Pitta et al. 2005.32
physical activity, relative to the healthy controls, was
57%.33 The level of physical activity of people with COPD
decreases with increased disease severity and in response
to an acute exacerbation.34,35
Besides engaging in lower levels of physical activity,
people with COPD spend a large proportion of their
waking hours sitting and lying down.32 That is, compared
to healthy controls, during waking hours, people with
COPD spend nearly 25% more time sitting and 200%
more time lying down (Fig. 2).32 In contrast to data on
physical activity, sedentary time does not seem to differ
across severities of COPD.36 Of note, it appears that sitting
time in this population is associated with lower exercise
capacity, lower motivation to exercise and higher
number of exacerbations in the past year.36
physical activity by individuals with a chronic health
condition is likely to have additional health consequences to those described in the general population.
That is, in addition to the impairments imposed by the
disease process itself, deconditioning of both the cardiovascular system and muscles of locomotion resulting
from participation in low levels of physical activity often
contributes to their decline in functional status.40 This has
led to an interest in the role of rehabilitative strategies
that aim to optimise participation in physical activity in
people with a chronic health condition.
Health benefits of physical activity and
consequences of low levels of physical
activity in people with COPD
There are broadly two approaches to increasing physical
activity; direct and ‘stealth’ interventions. Direct interventions use strategies to influence directly physical
activity, while ‘stealth’ interventions may target other
values and beliefs that extend beyond health to increase
physical activity. Data pertaining to interventions that
may improve sedentary behaviour in people with COPD
are scarce. Regarding physical activity, one direct intervention that has received attention in people with COPD
is the use of exercise training, within the framework of
pulmonary rehabilitation. Despite achieving strong evidence for reducing symptoms of dyspnoea and fatigue,
increasing exercise capacity, improving quality of life6
and reducing hospitalisations related to acute exacerbations of COPD,7 the effects of exercise training on physical
activity appear to be limited. A systematic review and
The benefits of participating in regular physical activity
are not limited to the general population. Specifically, in
people with COPD, regular participation in physical activity has been shown to reduce the risk of hospitalisation
and lower all-cause mortality.37 Higher levels of physical
activity in those with COPD also appear to minimise
extrapulmonary manifestations of the disease, such as
systemic inflammation and cardiac dysfunction.38 The
benefits of physical activity appear to be present prior to
the development of COPD as current smokers who participate in regular physical activity have a reduced rate of
decline in lung function.39 Participation in low levels of
How can we change physical activity
and sedentary behaviour in people
with COPD?
© 2014 Royal Australasian College of Physicians
478
Activity and sitting: lessons for COPD
meta-analysis of seven studies (two randomised trials and
five single-group interventional studies) examining the
effect of exercise training on physical activity in a total
of 472 people (419 males) with COPD demonstrated
minimal change, with an overall effect size of 0.12 (P =
0.01),41 which was equivalent to an increase of approximately 5 min per day. This small change may be because
pulmonary rehabilitation programmes lack an effective
behavioural component that targets changes in physical
activity outside of what people complete as part of their
structured exercise.
Examining the effects of embedding psychosocial interventions in pulmonary rehabilitation programs is a promising area for future research and may have real potential
for changing physical activity and sedentary time in
people with compromised lung function.42 A recent study
in overweight and obese adults showed that combining a
behavioural intervention with prescribed exercise
increased physical activity more so than exercise prescription alone.43 This would suggest the utility of this
approach in people with chronic conditions. The recent
Lancet series on physical activity contained a comprehensive review of approaches for increasing physical activity
within different population groups, and found strong evidence for behavioural and social approaches.44 Interventions within the primary care setting are successful at
increasing the self-reported physical activity levels of
inactive individuals at 12 months, with recent reviews of
physical activity interventions in adults45 and older
adults46 reporting that interventions containing behavioural strategies, such as goal setting, self-monitoring and
feedback were most effective. Nevertheless, in people
with COPD who are commencing a pulmonary rehabilitation programme, the timing of such interventions may
be critical given that for many people, it may be too much
to commence a regular exercise programme and at
the same time undertake more physical activity in their
daily life.
An example of an evidence-based behavioural
approach used in the primary care setting is the 5As
approach. This has been used widely in smoking cessation47 and was adopted in the 2013 National Health and
Medical Research Council clinical practice guidelines for
the management of overweight and obesity in adults,
adolescents and children in Australia as a useful framework for general practitioners to help obese patients
manage their weight and is based on: Assess level of
behaviour; Advise based on personal health risks; Agree
on a realistic set of goals; Assist to anticipate barriers and
develop a specific action plan; and, Arrange follow-up
support. Figure 3 contains an example of how this
approach may be used in clinical practice to influence
sedentary behaviour.
Figure 3 Example of using a ‘stealth’ behaviour to increase physical
activity by reducing sedentary time.
Rather than direct interventions to increase physical
activity, it is possible that ‘stealth’ interventions, such as
reducing time spent in sedentary behaviours (e.g. television viewing)48 in order to increase physical activity, may
offer greater success in people with COPD. This fits nicely
with the premise that sedentary behaviour is a new
health behaviour change target in its own right. While
most sedentary behaviour interventions have been conducted with children and adolescents, emerging evidence
suggests the utility of this stealth approach in adults.
Three studies (all in non-COPD populations) are worth
noting here. TView evaluated a 3-week programme using
an electronic television lock-out system with 36 overweight and obese participants aged 22–61 years.49 Stand
Up For Your Health50 took a whole-of-day approach to
reduce and interrupt prolonged sedentary time, targeting
television time as well as other sedentary behaviours,
such as sitting and reading, or engaging in computer use.
This single-group feasibility study conducted over 2
weeks with 59 older adults (aged 60–92 years) used a
face-to-face goal-setting consultation and one tailored
mailing.50 The final single group feasibility study was
conducted with 24 older adults (aged mean ± SD, 68 ± 6
years) and also used a face-to-face consultation and feedback on sedentary time as part of the intervention.51 All
three interventions achieved around a 30 min per day
reduction in sedentary time (24 to 37 min per day), of
which approximately one third (7 to 13 min/day) of this
time was reallocated to moderate to vigorous intensity
physical activity.
The findings from these studies suggest that changes in
sedentary time are achievable and that increases in physical activity are likely. Environmental changes, such
as devices to limit the amount of TV a person watches,
may be difficult to implement; however, behavioural
approaches produced similar changes in sedentary time.
© 2014 Royal Australasian College of Physicians
479
Hill et al.
The consultation sessions in the two feasibility studies49,50
used concepts from the 5As approach in that they:
assessed participants’ level of sedentary time (using
devices); advised participants of the pros and cons of
reducing sedentary time; agreed on a set of goals (in
conjunction with the participants); and assisted with overcoming barriers. No arrangements were made for
follow-up support. These interventions took an average
of 45 min50 and 30 min51 to deliver. The appeal of these
approaches is that they are simple, achievable and
unlikely to do any harm. However, randomised trials of
longer term interventions are needed to evaluate intervention efficacy in a range of populations. While these
studies were conducted in non-COPD populations, they
were in overweight and obese and older adult populations with a range of chronic conditions.
Earlier work has suggested that people with COPD
utilise 58% of their aerobic capacity to complete usual
activities of daily living.52 This is considerably more than
individuals with normal aerobic capacity, who have been
estimated to utilise 40% of their aerobic capacity during
usual activities of daily living.53 Given the limited aerobic
capacity of individuals with COPD, an intervention
focussed on increasing light intensity physical activity
References
1 Garber CE, Blissmer B, Deschenes MR,
Franklin BA, Lamonte MJ, Lee IM et al.
American College of Sports Medicine
position stand. Quantity and quality of
exercise for developing and maintaining
cardiorespiratory, musculoskeletal, and
neuromotor fitness in apparently
healthy adults: guidance for prescribing
exercise. Med Sci Sports Exerc 2011; 43:
1334–59.
2 Australian Bureau of Statistics. Let’s get
physical: how do adult Australians
measure up? Perspectives on Sport. 2013.
[cited 2015 Apr 11]. Available from
URL: http://www.abs.gov.au/ausstats/
[email protected]/Lookup/4156.0.55.001Main+
Features3Nov%202013
3 Owen N, Salmon J, Koohsari MJ, Turrell
G, Giles-Corti B. Sedentary behaviour
and health: mapping environmental and
social contexts to underpin chronic
disease prevention. Br J Sports Med 2014;
48: 174–7.
4 Dunstan DW, Kingwell BA, Larsen R,
Healy GN, Cerin E, Hamilton MT et al.
Breaking up prolonged sitting reduces
postprandial glucose and insulin
5
6
7
8
9
and breaking up time spent in sedentary behaviour may
be more appropriate in this population than one focussed
primarily on increasing time spent in moderate to vigorous intensity physical activity. The development of such
interventions – a key area for future research in individuals with COPD – should consider the approaches
described above (i.e. the 5As; stealth interventions) in
conjunction with evidence-based intervention strategies
(e.g. motivational interviewing; self-monitoring) for
behaviour change.
Conclusion
This paper has reviewed the benefits of physical activity
and the adverse effects of sedentary behaviour.
Exertional dyspnoea and fatigue pose additional challenges for people with COPD when attempting to undertake physical activity. Strategies are needed to assist both
healthy individuals and those with chronic conditions,
such as COPD to: (i) increase the time spent in physical
activity (which includes activity across the intensity spectrum); (ii) reduce total time spent sitting; and (iii) break
up any periods of prolonged sitting across the day.
responses. Diabetes Care 2012; 35:
976–83.
Wilmot EG, Edwardson CL, Achana FA,
Davies MJ, Gorely T, Gray LJ et al.
Sedentary time in adults and the
association with diabetes, cardiovascular
disease and death: systematic review
and meta-analysis. Diabetologia 2012; 55:
2895–905.
Lacasse Y, Goldstein R, Lasserson TJ,
Martin S. Pulmonary rehabilitation for
chronic obstructive pulmonary disease.
Cochrane Database Syst Rev 2006; (4):
CD003793.
Griffiths TL, Burr ML, Campbell IA,
Lewis-Jenkins V, Mullins J, Shiels K
et al. Results at 1 year of outpatient
multidisciplinary pulmonary
rehabilitation: a randomised controlled
trial. Lancet 2000; 355: 362–8.
Sedentary Behaviour Research Network.
What is sedentary behaviour? [cited
2014 Aug 14]. Available from URL:
http://www.sedentarybehaviour
.org/what-is-sedentary-behaviour/
Norton K, Norton L, Sadgrove D.
Position statement on physical activity
and exercise intensity terminology.
J Sci Med Sport 2010; 13: 496–502.
10 Ainsworth BE, Haskell WL, Whitt MC,
Irwin ML, Swartz AM, Strath SJ et al.
Compendium of physical activities: an
update of activity codes and MET
intensities. Med Sci Sports Exerc 2000;
32(9 Suppl): S498–504.
11 Prince SA, Adamo KB, Hamel ME, Hardt
J, Connor Gorber S, Tremblay M. A
comparison of direct versus self-report
measures for assessing physical activity
in adults: a systematic review. Int J
Behav Nutr Phys Act 2008; 5: 56.
12 Hunt T, Williams MT, Olds TS.
Reliability and validity of the
multimedia activity recall in children
and adults (MARCA) in people with
chronic obstructive pulmonary disease.
PLoS ONE 2013; 8: e81274.
13 Murphy SL. Review of physical activity
measurement using accelerometers in
older adults: considerations for research
design and conduct. Prev Med 2009; 48:
108–14.
14 van Poppel MN, Chinapaw MJ,
Mokkink LB, van Mechelen W, Terwee
CB. Physical activity questionnaires for
adults: a systematic review of
measurement properties. Sports Med
2010; 40: 565–600.
© 2014 Royal Australasian College of Physicians
480
Activity and sitting: lessons for COPD
15 Pedersen BK, Febbraio MA. Muscles,
exercise and obesity: skeletal muscle as a
secretory organ. Nat Rev Endocrinol 2012;
8: 457–65.
16 Hallal PC, Andersen LB, Bull FC,
Guthold R, Haskell W, Ekelund U et al.
Global physical activity levels:
surveillance progress, pitfalls, and
prospects. Lancet 2012; 380: 247–57.
17 Lee IM, Shiroma EJ, Lobelo F, Puska P,
Blair SN, Katzmarzyk PT et al. Effect of
physical inactivity on major
non-communicable diseases worldwide:
an analysis of burden of disease and life
expectancy. Lancet 2012; 380: 219–29.
18 Sedentary Behaviour Research Network.
Letter to the editor: standardized use of
the terms ‘sedentary’ and ‘sedentary
behaviours’. Appl Physiol Nutr Metab
2012; 37: 540–2.
19 Healy GN, Dunstan DW, Salmon J,
Shaw JE, Zimmet PZ, Owen N.
Television time and continuous
metabolic risk in physically active adults.
Med Sci Sports Exerc 2008; 40: 639–45.
20 Healy GN, Clark BK, Winkler EA,
Gardiner PA, Brown WJ, Matthews CE.
Measurement of adults’ sedentary time
in population-based studies. Am J Prev
Med 2011; 41: 216–27.
21 Matthews CE, Keadle SK, Sampson J,
Lyden K, Bowles HR, Moore SC et al.
Validation of a previous-day recall
measure of active and sedentary
behaviors. Med Sci Sports Exerc 2013; 45:
1629–38.
22 Kozey-Keadle S, Libertine A, Lyden K,
Staudenmayer J, Freedson PS. Validation
of wearable monitors for assessing
sedentary behavior. Med Sci Sports Exerc
2011; 43: 1561–7.
23 Tremblay MS, Colley RC, Saunders TJ,
Healy GN, Owen N. Physiological and
health implications of a sedentary
lifestyle. Appl Physiol Nutr Metabol 2010;
35: 725–40.
24 Hamilton MT, Hamilton DG, Zderic TW.
Role of low energy expenditure and
sitting in obesity, metabolic syndrome,
type 2 diabetes, and cardiovascular
disease. Diabetes 2007; 56: 2655–67.
25 Levine JA, Miller JM. The energy
expenditure of using a ‘walk-and-work’
desk for office workers with obesity. Br J
Sports Med 2007; 41: 558–61.
26 Healy GN, Matthews CE, Dunstan DW,
Winkler EA, Owen N. Sedentary time
and cardio-metabolic biomarkers in US
adults: NHANES 2003–06. Eur Heart J
2011; 32: 590–7.
27 Buckley JP, Mellor DD, Morris M,
Joseph F. Standing-based office work
shows encouraging signs of attenuating
post-prandial glycaemic excursion. Occup
Environ Med 2014; 71: 109–11.
28 Australian Government Department of
Health. Australia’s Physical Activity and
Sedentary Behaviour Guidelines. 2014.
29 Healy GN, Dunstan DW, Salmon J,
Cerin E, Shaw JE, Zimmet PZ et al.
Objectively measured light-intensity
physical activity is independently
associated with 2-h plasma glucose.
Diabetes Care 2007; 30: 1384–9.
30 Loprinzi PD, Lee H, Cardinal BJ. Daily
movement patterns and biological
markers among adults in the United
States. Prev Med 2014; 60: 128–30.
31 Vestbo J, Hurd SS, Agustí AG, Jones
PW, Vogelmeier C, Anzueto A et al.
Global strategy for the diagnosis,
management, and prevention of chronic
obstructive pulmonary disease: GOLD
executive summary. Am J Respir Crit Care
Med 2013; 187: 347–65.
32 Pitta F, Troosters T, Spruit MA, Probst
VS, Decramer M, Gosselink R.
Characteristics of physical activities in
daily life in chronic obstructive
pulmonary disease. Am J Respir Crit Care
Med 2005; 171: 972–7.
33 Vorrink SN, Kort HS, Troosters T,
Lammers JW. Level of daily physical
activity in individuals with COPD
compared with healthy controls. Respir
Res 2011; 12: 33.
34 Watz H, Waschki B, Meyer T,
Magnussen H. Physical activity in
patients with COPD. Eur Respir J 2009;
33: 262–72.
35 Pitta F, Troosters T, Probst VS, Spruit
MA, Decramer M, Gosselink R. Physical
activity and hospitalization for
exacerbation of COPD. Chest 2006; 129:
536–44.
36 Hartman JE, Boezen HM, de Greef MH,
Ten Hacken NH. Physical and
psychosocial factors associated with
physical activity in patients with chronic
obstructive pulmonary disease. Arch Phys
Med Rehabil 2013; 94: 2396–2402.
37 Garcia-Aymerich J, Lange P, Benet M,
Schnohr P, Anto JM. Regular physical
activity reduces hospital admission and
mortality in chronic obstructive
pulmonary disease: a population based
cohort study. Thorax 2006; 61: 772–8.
38 Watz H, Waschki B, Beoehme C,
Claussen M, Meyer T, Magnussen H.
Extrapulmonary effects of chronic
39
40
41
42
43
44
45
46
47
48
49
obstructive pulmonary disease on
physical activity. Am J Respir Crit Care
Med 2008; 177: 743–51.
Garcia-Aymerich J, Lange P, Benet M,
Schnohr P, Anto JM. Regular physical
activity modifies smoking-related lung
function decline and reduces risk of
chronic obstructive pulmonary disease: a
population-based cohort study. Am J
Respir Crit Care Med 2007; 175: 458–63.
Maltais F, LeBlanc P, Whittom F, Simard
C, Marquis K, Bélanger M et al.
Oxidative enzyme activities of the vastus
lateralis muscle and the functional status
in patients with COPD. Thorax 2000; 55:
848–53.
Cindy Ng LW, Mackney J, Jenkins S,
Hill K. Does exercise training change
physical activity in people with COPD?
A systematic review and meta-analysis.
Chron Respir Dis 2012; 9: 17–26.
Troosters T, van der Molen T, Polkey M,
Rabinovich RA, Vogiatzis I, Weisman I
et al. Improving physical activity in
COPD: towards a new paradigm. Respir
Res 2013; 14: 115.
Kozey-Keadle S, Staudenmayer J,
Libertine A, Mavilia M, Lyden K, Braun
B et al. Changes in sedentary time and
physical activity in response to an
exercise training and/or lifestyle
intervention. J Phys Act Health 2014; 11:
1324–33.
Heath GW, Parra DC, Sarmiento OL,
Andersen LB, Owen N, Goenka S et al.
Evidence-based intervention in physical
activity: lessons from around the world.
Lancet 2012; 380: 272–81.
Michie S, Abraham C, Whittington C,
McAteer J, Gupta S. Effective techniques
in healthy eating and physical activity
interventions: a meta-regression. Health
Psychol 2009; 28: 690–701.
King AC. Physical activity for an aging
population. Public Health Rev 2010; 32:
401–26.
Papadakis S, McDonald P, Mullen KA,
Reid R, Skulsky K, Pipe A. Strategies to
increase the delivery of smoking
cessation treatments in primary care
settings: a systematic review and
meta-analysis. Prev Med 2010; 51:
199–213.
Robinson T. Stealth Interventions for
Obesity Prevention and Control: Motivating
Behavior Change. Vol. 2010. New York,
NY: Elsevier Inc; 2010.
Otten JJ, Jones KE, Littenberg B,
Harvey-Berino J. Effects of television
viewing reduction on energy intake
© 2014 Royal Australasian College of Physicians
481
and expenditure in overweight and
obese adults: a randomized controlled
trial. Arch Intern Med 2009; 169:
2109–15.
50 Gardiner PA, Eakin EG, Healy GN,
Owen N. Feasibility of reducing older
adults’ sedentary time. Am J Prev Med
2011; 41: 174–7.
51 Fitzsimons CF, Kirk A, Baker G, Michie
F, Kane C, Mutrie N. Using an
individualised consultation and activPAL
feedback to reduce sedentary time in
older Scottish adults: results of a
feasibility and pilot study. Prev Med
2013; 57: 718–20.
52 Hill K, Dolmage TE, Woon L, Coutts D,
Goldstein R, Brooks D. Defining the
relationship between average daily
energy expenditure and field-based
walking tests and aerobic reserve in
COPD. Chest 2012; 141: 406–12.
53 Wasserman K, Hansen J, Sue D, Whipp
B. Principles of Exercise Testing and
Interpretation.Philadelphia, PA:
Lippincott, Williams and Wilkins; 2005.
C L I N I C A L P E R S P E CT I V E S
Familial colorectal cancer
M. S. Lung,1 A. H. Trainer,1,2,3 I. Campbell1 and L. Lipton3
1
Research Division and 2Familial Cancer Centre, Peter MacCallum Cancer Centre and 3Familial Cancer Centre, Royal Melbourne Hospital, Melbourne,
Victoria, Australia
Key words
colorectal neoplasm, adenomatous polyposis
coli, hereditary nonpolyposis, neoplastic
syndrome, hereditary.
Correspondence
Lara Lipton, Familial Cancer Clinic, Royal
Melbourne Hospital, Parkville, Vic. 3050,
Australia.
Email: [email protected]
Abstract
Identifying individuals with a genetic predisposition to developing familial colorectal
cancer (CRC) is crucial to the management of the affected individual and their family. In
order to do so, the physician requires an understanding of the different gene mutations
and clinical manifestations of familial CRC. This review summarises the genetics, clinical
manifestations and management of the known familial CRC syndromes, specifically
Lynch syndrome, familial adenomatous polyposis, MUTYH-associated neoplasia,
juvenile polyposis syndrome and Peutz–Jeghers syndrome. An individual suspected of
having a familial CRC with an underlying genetic predisposition should be referred to a
familial cancer centre to enable pre-test counselling and appropriate follow up.
Received 27 August 2014; accepted 24
February 2015.
doi:10.1111/imj.12736
Introduction
Australia and New Zealand have the highest incidence
rate of colorectal cancer (CRC) in the world, with an
Funding: M. S. Lung is funded by the Cancer Council of Victoria
and The University of Melbourne.
Conflict of interest: None.
Abbreviations
APC, adenomatous polyposis coli; CHRPE, congenital hypertrophy of
the retinal pigment epithelium; CI, confidence interval; CRC, colorectal
cancer; FAP, familial adenomatous polyposis; HHT, hereditary
haemorrhagic telangiectasia; HR, hazard ratio; IHC,
immunohistochemistry; JPS, juvenile polyposis syndrome; LS, Lynch
syndrome; MMR, mismatch repair; MSI, microsatellite instability; OR,
odds ratio; PJS, Peutz–Jeghers syndrome.
age-adjusted rate of 46 per 100 000 men and 32 per
100 000 women in 2008.1 The risk of developing CRC by
age 85 is 1 in 10 for men and 1 in 15 for women.2
Approximately 30% of the risk of sporadic CRC is
thought to be due to inherited genetic factors.3 Conversely, hereditary CRC syndromes with a known highrisk genetic aetiology make up approximately 5% of
CRC. Such disorders include Lynch syndrome (LS), familial adenomatous polyposis (FAP), MUTYH-associated
neoplasia and the hamartoma syndromes (Table 1).
Identification of individuals and families with these
syndromes allows the implementation of effective surveillance strategies which result in a reduction in cancer
incidence and death in many cases.
Here we discuss the genetics, clinical manifestations
and management of the known familial CRC syndromes,
© 2015 Royal Australasian College of Physicians
482
Familial colorectal cancer
Table 1 Summary of main familial colorectal cancer syndromes with associated genes and phenotypes
Syndrome
Associated gene(s)
Lynch syndrome
MLH1, MSH2,
MSH6, PMS2,
EPCAM
APC
Colonic phenotype
Major extra-colonic associations
Endometrial, ovarian, gastric, small bowel, urinary tract,
brain, biliary cancers; sebaceous gland tumours;
keratoacanthomas
Gastric fundic polyps; duodenal adenomas; papillary thyroid
cancer; medulloblastomas; desmoids; osteomas
Duodenal polyposis; other associations not fully defined
MUTYH-associated
neoplasia
Juvenile polyposis
syndrome
MUTYH
SMAD4, BMPR1A
CRC often R-sided; tends to be
poorly-differentiated; mucinous/signet
ring histology; lymphocytic infiltrate
Multiple adenomatous polyps
(10–1000 s)
adenomatous polyps or CRC without
adenomatous polyps
Juvenile polyps
Peutz–Jeghers
syndrome
STK11
Hamartomas
Classical FAP
and offer guidelines on whom to refer to a familial cancer
clinic for risk assessment and potential genetic testing.
Lynch syndrome
LS (hereditary non-polyposis CRC, HNPCC) is the most
common inherited CRC syndrome, accounting for
approximately 3% of all CRC diagnoses.4
Genetics
LS is an autosomal dominant syndrome caused by a
mutation in one of four mismatch repair (MMR) genes
(MLH1, MSH2, MSH6, PMS2) or the EPCAM gene, which is
situated upstream of MSH2. Interestingly, EPCAM is not
involved in MMR, but deletions in this gene indirectly
cause abrogation of MSH2 function.5,6 Germline MLH1
and MSH2 mutations account for >80% of LS cases,7
MSH6 for up to 10%,7 PMS2 for 2–3% and EPCAM for
1–3%.8 It is likely that PMS2 mutations make a significant
contribution to LS but the exact extent of this contribution is currently unclear because of difficulties in distinguishing genuine mutations in this gene from those
present in numerous pseudogenes.9
The types of inactivating mutations in the MMR
genes include frameshift, nonsense, splice site, missense/
in-frame deletions or genomic rearrangements which can
occur throughout each gene.10,11 Amino acid altering
missense mutations are more common in MLH1 than
MSH2.12 Large deletions of the 3′ end of the EPCAM gene
result in transcriptional read-through and epigenetic
silencing of MSH2.5 Founder mutations have also been
described such as an A→T transversion in the donor
splice site of intron 5 of MSH2, which occurs in Newfoundland,13 but also occurs as a recurrent mutation
worldwide and may account for up to 10% of LS cases.14
Juvenile polyps throughout gastrointestinal tract; increased
risk of small bowel, stomach and pancreas cancers
Hereditary haemorrhagic telangiectasia
Mucocutaneous pigmentation
Hamartomas throughout gastrointestinal tract; increased
risk of breast, stomach, pancreatic and gynaecological
cancers
LS tumours arise when the wild type copy of the gene
is lost in the tumour, resulting in inactivation of the
MMR pathway in the tumour. This causes an accumulation of errors in long repetitive stretches of DNA called
microsatellites, resulting in alleles of differing lengths or
‘microsatellite instability’ (MSI).15,16
Genes with microsatellites in their coding region often
undergo frameshift mutations in MSI tumours and
include genes involved in cell proliferation, apoptosis and
DNA repair such as TCF4, ILGFR-2, TGFBR2, AXIN2, BAX,
PTEN, CHK1, MLH3, MSH3 and MSH6.17
In LS, cancers may harbour somatic mutations in KRAS
and adenomatous polyposis coli (APC) but rarely BRAF.18
Clinical features and
genotype-phenotype correlations
Individuals with LS are at risk of developing CRC at an
earlier age (median age 45)4 than the general population (median age 71). They have a varying risk to age
70 years of developing CRC which depends upon the
gene that is mutated (Table 2). The majority (∼70%) of
CRC develop proximal to the splenic flexure.20 CRC in
LS tends to be poorly differentiated, have mucinous or
signet ring histology and have a marked lymphocytic
infiltrate.4
Female carriers of a LS mutation carry a 12–54% lifetime risk of endometrial cancer depending on the gene
mutated and a risk of ovarian cancer between 1% and
38% (Table 1).10 Other LS-associated cancers include
gastric cancer, small bowel adenocarcinoma, transitional cell carcinoma of bladder, ureters or renal pelvis,
biliary tract cancer, glioblastomas (previously known
as Turcot variant) and sebaceous gland tumours and
© 2015 Royal Australasian College of Physicians
483
Lung et al.
Table 2 Cumulative cancer risk to age 70 years by mutation type in Lynch syndrome
Mismatch repair
gene mutation
MLH110
MSH210
MSH610
PMS211
EPCAM19
CRC
risk
(%)
41
48
12
15–20
75
Endometrial
cancer
risk (%)
Ovarian
cancer
risk (%)
Gastric
cancer
risk (%)
Small bowel
cancer
risk (%)
Urinary tract
cancer
risk (%)
Brain
cancer
risk (%)
Biliary tract
cancer
risk (%)
54
21
16
15
12
20
38
1
6
0.2
0
0.4
1.1
0
0.2
2.2
0.7
≤3
≤3
≤3
1.9
keratoacanthomas (previously known as Muir–Torre
variant).4,21 The risk of these cancers varies with the
specific gene mutated.
Diagnosis of LS
The revised Bethesda guidelines22 were developed to help
clinicians decide on which CRC to test for MSI. These
guidelines are highly sensitive but have poor specificity.23
The Amsterdam criteria24 have been used since prior to
the discovery of MMR gene mutations to identify LS
families.24 They remain highly specific for LS mutations
(60–70%) but do not have the sensitivity to be used for
population screening.
Molecular testing of CRC using MSI or immunohistochemistry (IHC) for the four MMR proteins is used,
where possible, to aid in choosing patients for Lynch
syndrome genetic testing. The sensitivity of both methods
is high and comparable.25 These tests are also useful in
screening for LS in endometrial cancer.26
Certain caveats apply to IHC testing however. False
negatives may occur if the protein is present but not
functional, which can occur in the presence of germline
pathogenic missense mutations.27 Also, 15% of sporadic
CRC harbour somatic hypermethylation of the MLH1
promoter causing an MSI phenotype in the cancers with
loss of MLH1 IHC and may therefore be mistaken for LS.28
These patients do not have LS and 40% of these CRC may
harbour a V600E BRAF mutation that is very rare in LS.29
If a mutation is found in a MMR gene, predictive genetic
testing can be offered to unaffected family members before
the age at which screening should commence.
Surveillance and management
CRC surveillance in LS reduces CRC-related mortality,30,31 is estimated to increase life expectancy by 7
years32 and is cost-effective.32
There is non-randomised, controlled trial evidence that
three yearly colonoscopies reduce incidence and mortality from CRC in LS33; however, level IIIC evidence
favours 1–2-year intervals between colonoscopies due to
the rapid adenoma to carcinoma development in LS and
this is the standard practice in Australia.4,34–36
LS mutation carriers should start surveillance colonoscopy at the ages of 25 or 5 years prior to the earliest
diagnosis of CRC in the family, whichever is younger.36
There is no recommended age at which surveillance
colonoscopies should be stopped.
Guidelines based on expert consensus regarding the
management of LS34 suggest that screening for extracolonic cancers should ideally be done as part of a clinical
trial as no direct evidence for reduction in mortality from
screening is available. Prophylactic hysterectomy and
bilateral salpingo-oopherectomy is recommended for
female mutation carriers when they have completed
their families and reached the age of 40 years34 as no
effective surveillance exists for ovarian or endometrial
cancer.37,38 For patients with a family history of gastric
cancer or those from a high-risk background (Chinese,
Korean, Japanese, Chilean), second yearly gastroscopy
may be considered.
Surgical management of CRC in LS
When a LS mutation carrier develops CRC, several factors
need to be considered when deciding between a partial
colectomy and a subtotal colectomy. The risk of interval
cancers on surveillance colonoscopies post partial
colectomy is between 6% and 35%.34 However, no difference in 10-year survival was seen in a retrospective cohort
study of 382 patients who underwent either extended
colectomy or a segmental resection for their first CRC,
despite a metachronous CRC rate of 0% in those who had
an extended colectomy versus 22% for those who had a
segmental resection.39 In a separate study, quality of life
did not differ significantly between 23 patients who had a
segmental operation compared with 27 patients who had
extended surgery.40 Both the European Society of Medical
Oncology 2013 familial CRC guidelines36 and the LS management guidelines by the Mallorca group34 recommend
discussing the option of an extended colectomy with a
© 2015 Royal Australasian College of Physicians
484
Familial colorectal cancer
LS patient with CRC. Factors including patient wishes,
family planning and likely adherence to close follow-up
colonoscopic surveillance should be considered.
Chemoprevention
The Colorectal Adenoma/Carcinoma Prevention Program
2 trial randomised over 1000 individuals with known or
suspected LS to 600 mg per day of aspirin for 2 years
versus placebo.41 The study’s primary end-point was
development of colorectal adenoma or carcinoma.
There was no difference in the incidence of colorectal
adenomas or carcinomas between the two groups at a
mean follow-up time of 27 months. At a mean follow up
of 55.7 months, the hazard ratio (HR) for developing CRC
in the aspirin group was 0.63 (95% confidence interval
(CI) 0.35–1.13, P = 0.12) on an intention-to-treat analysis.
In the subgroup of patients who completed 2 years of
600 mg aspirin, the HR was 0.41 (95% CI 0.19–0.86, P =
0.02) compared with the placebo group. There was also a
trend to a decrease in non-CRC LS cancers, with the group
of patients who completed 2 years of aspirin having a
HR of 0.47 (95% CI 0.21–1.06, P = 0.07). Adverse events
did not differ while on treatment; however, data were
not collected on post-intervention adverse effects. The
ongoing Colorectal Adenoma/Carcinoma Prevention
Program 3 trial is determining the optimal dose of aspirin.
Constitutive MMR deficiency
Constitutive mismatch repair deficiency, a rare autosomal
recessive condition, has been described primarily in the
paediatric population.42 The inheritance of biallelic MMR
gene mutations results in lack of competent MMR in all
tissues. Individuals develop haematological malignancies
and rare brain tumours such as gliomas in childhood as
well as LS-associated tumours and café au-lait spots.43 It
can also present with colonic polyposis suggestive of
attenuated FAP or MUTYH-associated neoplasia.44 This
condition is diagnosed on IHC for MMR proteins, and
demonstrates absent MMR staining in both tumour and
surrounding normal tissue.
Familial adenomatous polyposis
Classical FAP results in the development of hundreds to
thousands of polyps in the colon and/or rectum by the
time of adolescence. Untreated, FAP results in CRC in
∼100% of cases by the fourth decade.45 Seventy to eighty
per cent of these CRC are left sided.46 FAP occurs in 1 in
8000–10 000 individuals and accounts for <1% of cases
of CRC.
Genetics
FAP is caused by germline mutations in the APC gene on
chromosome 5q. Ninety-five per cent of APC mutations
are protein truncating.47 While FAP is an autosomal
dominant syndrome, it has been estimated that 25% of
germline APC mutations occur de novo.45
APC is a tumour suppressor gene. It is thought of as a
‘gatekeeper’ gene as it inhibits cell growth by regulating
signalling through the Wnt pathway. It also plays a role in
other processes including cellular adhesion, cytoskeleton
stabilisation and possibly cell cycle and apoptosis.47
The position of a germline mutation within the APC
gene is associated with particular clinical features producing a phenotype/genotype correlation. Mutations
between codons 1250 and 1464 are associated with a
more severe form of the disease (>5000 polyps).48 Mutations in codon 1309 result in an earlier onset of CRC at a
mean age of 35 years.49 Mutations at or beyond codon
1444 confer an 11-fold increase in risk of developing
desmoids.50 Mutations between exons 9 and 15 (codons
311 to 1444) are associated with congenital hypertrophy
of the retinal pigment epithelium (CHRPE), which is the
commonest extra-intestinal manifestation of FAP.49 Once
a pathogenic mutation in the APC gene is found, presymptomatic genetic screening can be offered to blood
relatives.
An attenuated form of FAP is associated with mutations
in the 5′ end, exon 9 and 3′ ends of the APC gene.51–53 This
group of patients generally has a delayed appearance of
colonic adenomas and CRC by around 10–20 years.54 In
clinically determined attenuated FAP, with 30–100 polyps,
an APC mutation is found in 30% of cases.55
Extra-intestinal manifestations of FAP
FAP is associated with gastric fundic gland polyps and
duodenal adenomas, duodenal cancer, papillary thyroid
cancer (rare), hepatoblastoma in childhood (rare)
and central nervous system tumours (predominantly
medulloblastoma). Non-malignant associations include
desmoid tumours, CHRPE,56 epidermoid skin cysts and
osteomas (Gardner syndrome), lipomas and adrenal
tumours (very rarely malignant).56 CHRPE associated
with FAP tends to manifest as multiple bilateral lesions
with a depigmented halo.57
The main causes of death in FAP patients following
colectomy are duodenal cancer and desmoid tumours.58
Desmoid tumours are locally aggressive connective tissue
tumours that do not have metastatic potential. They
usually occur in the abdominal wall or mesentery or in
an abdominal surgical scar, and may cause morbidity and
mortality due to small bowel obstruction, ischaemia or
© 2015 Royal Australasian College of Physicians
485
Lung et al.
perforation, intra-abdominal abscesses and fistulas, or
ureteric obstruction.56
Surveillance and management
Since the advent of molecular diagnosis and prophylactic
surveillance and surgery, the mortality from CRC in FAP
has decreased significantly.59
For known APC mutation carriers and at risk relatives
from families where a clinical diagnosis has been made
but in which mutation testing is not possible, two yearly
flexible sigmoidoscopy should start at the age of 12–14
years of age.36 Yearly colonoscopy should be done when
polyps are seen and continued until colectomy.36
Colectomy is usually performed in late adolescence or
early adulthood. Surgical options are proctocolectomy
with ileal pouch-anal anastomosis in individuals with
a high rectal polyp burden or total colectomy with
ileorectal anastomosis. Those with a retained rectum
require annual endoscopy post-colectomy.
For attenuated FAP cases, two yearly colonoscopy is
recommended rather than sigmoidoscopy, starting at the
age of 18–20 years,36 as adenomas are found throughout
the colon.
Screening for extra-intestinal manifestations of FAP
should begin at diagnosis or at the age of 25–30 years,
whichever is earlier.36 Upper gastrointestinal endoscopy
using both front- and side-viewing endoscopy is performed every 5 years until the detection of adenomas.
The frequency of upper endoscopies once adenomas are
detected depends on the Spigelman stage of the polyps,60
which takes into account polyp number, size, histology
and dysplasia.
Symptomatic desmoid tumours may be treated
with medical therapy including non-steroidal antiinflammatory drugs with tamoxifen or raloxifene61 and
with chemotherapy.62 Surgical manipulation may result
in disease progression and should only be undertaken by
specialist surgeons after multidisciplinary consultation.36
Chemoprevention
Small trials have suggested that ascorbate,63 sulindac64
and the COX-2 inhibitors65 may result in a reduction in
adenoma burden of 10–40% while on treatment.66 Some
patients had regression of their adenomas; however,
the adenomas recurred when treatment was stopped.
The Concerted Action Polyp Prevention 1 trial did not
find any effect on polyp number with the use of aspirin
or resistant starch. Aspirin is not currently used for
chemoprevention of FAP.
MUTYH-associated neoplasia
MUTYH-associated polyposis (MAP) is an autosomal
recessive syndrome that results in the development of
several to hundreds of colorectal polyps and/or CRC67
and can present in a similar manner to FAP. MUTYH is an
important differential diagnosis in individuals with presumed de novo FAP, and genetic testing is the only means
to differentiate between these two conditions, which
have significantly different clinical implications to relatives of the affected individual. While the association of
MUTYH gene mutations with hereditary CRC was first
defined in patients with colorectal polyposis,67,68 subsequent population-based CRC studies have found that up
to one third of patients with biallelic MUTYH mutations
develop CRC without additional polyps being identified
at the time of diagnosis.69,70 Hence MUTYH-associated
CRC is not invariably associated with colorectal polyposis
and is a diagnosis that should be considered in young
onset CRC irrespective of polyp status. Patients with
biallelic mutations in MUTYH have a 93-fold increased
risk of CRC although this figure is subject to ascertainment bias.71 MAP is associated with duodenal polyposis
(<20%) and gastric fundic polyps (11%).72 Other extraintestinal manifestations have been observed in case
reports, including ovarian cancer and sebaceous gland
tumours but not desmoids.73
Genetics
The MUTYH gene is critical in the base excision repair
pathway. It is an adenine-specific DNA glycosylase
responsible for removing mispaired adenines. Biallelic
MUTYH mutations result in persistence of mispairing
between an oxidatively modified form of deoxyguanine
and adenine, leading to G:C→T:A transversion mutations.68 This is frequently seen in the APC and KRAS genes
in tumours from MAP patients.74
There are two founder mutations in MUTYH in
the Caucasian European population: p.Tyr179Cys and
p.Gly396Asp, which account for approximately 73% of
MUTYH mutations in these populations.75 Homozygosity
for the p.Tyr179Cys mutation is associated with a more
severe phenotype, with a significantly increased risk
of CRC (56-fold vs 19-fold) and an earlier mean age of
diagnosis (49.5 years vs 57.9 years) compared with
homozygosity for p.Gly396Asp.76 The mean age of onset of
the p.Gly396Asp/p.Tyr179Cys compound heterozygote is
in between the two homozygous groups at 52.5 years.76
Other pathogenic MUTYH mutations have been identified in different ethnic groups. For example, the
p.Val466Leu mutation was found to be homozygous in
four apparently unrelated affected Indian patients77
© 2015 Royal Australasian College of Physicians
486
Familial colorectal cancer
suggesting that this may be an important pathogenic
variant in the Indian population.
Under 1% of CRC is due to inherited biallelic mutations of the MUTYH gene.71,76
MUTYH heterozygotes
Multiple studies have attempted to ascertain if carriers of
one MUTYH gene mutation have an increased risk of
CRC.69,71,78,79 A large meta-analysis found a small increase
in CRC in monoallelic mutation carriers (odds ratio
(OR) 1.15 (95% CI = 0.98–1.36)).80 Several smaller welldesigned studies suggest a modestly increased risk.71,78,79
In this context, it is reasonable to offer individuals with
one MUTYH mutation population screening advice, or
five yearly colonoscopy from the age of 50 if they have a
family member affected with CRC before the age of 55
years.81
Surveillance and management for MAP
There are no controlled clinical trials of surveillance in
biallelic MUTYH mutation carriers to inform practice. The
European Society for Medical Oncology guidelines36
suggest following the surveillance and management recommendations for attenuated FAP. Upper gastrointestinal
endoscopy is reasonable but the optimal frequency is
unknown.
Hamartoma syndromes
Hamartomas are benign tumours consisting of cells
from the tissue of origin growing in a disorganised
way. The hamartomatous polyposis syndromes juvenile
polyposis syndrome (JPS) and Peutz–Jeghers syndrome
(PJS) are associated with an increased risk of various
malignancies.
JPS is an autosomal dominant condition. The word
‘juvenile’ in juvenile polyposis refers to the polyp histology, not the patient age. The diagnosis of juvenile
polyposis requires at least three juvenile polyps, a family
history of juvenile polyposis or juvenile polyps throughout the entire digestive tract. The incidence of JPS is
1:15 000–1:50 000. Children with juvenile polyposis may
present with rectal bleeding, anaemia, obstruction or
abdominal pain between the ages of 4 and 14 years of
age. Individuals with JPS have an increased risk of CRC
(39–68%) and small bowel, stomach and pancreas
cancers (21%).82,83 SMAD4 or BMPR1A gene mutations
are seen in approximately 40–60% of JPS cases.84–86
Around 20% of SMAD4 mutation carriers have associated
hereditary haemorrhagic telangiectasia (HHT),86 which
manifests as recurrent epistaxis, mucocutaneous telangi-
ectasia or pulmonary, cerebral or hepatic arteriovenous
malformations. A suggested approach to surveillance in
JPS is colonoscopy every 2 years from the age of 15 years
and upper gastrointestinal endoscopy every 2 years from
the age of 25 years.87 Upper and lower gastrointestinal
scopes should occur yearly once polyps are seen.87 Consideration of HHT is required for all individuals carrying a
SMAD4 mutation, and appropriate surveillance and management is required.87
PJS may present with typical mucocutaneous pigmentation in childhood and/or bowel intussusception,
obstruction or bleeding. Hamartomatous polyps are
found throughout the gastrointestinal tract, particularly
in the jejunum. They can also be found in extra-intestinal
sites such as the bladder and respiratory tract. The incidence of PJS is 1:50 000–1:200 000. There is an increased
risk of CRC (39%),88 breast cancer (45%), pancreatic
cancer (11%), stomach cancer (29%), gynaecological
cancer (18%) and Sertoli cell tumours of the testis.83,88
Mutations in the STK11 gene are found in up to 70% of
cases.88 It is inherited in an autosomal dominant fashion,
although up to 40% may occur de novo.89
Surveillance for cancer in asymptomatic individuals
with PJS should begin at age 30 years, and include
gastroduodenoscopy, colonoscopy and video capsule
endoscopy or barium follow through or magnetic resonance endoscopy at least three yearly.90 Recommendations for women with PJS include annual breast MRI
and bilateral mammography ± ultrasound from the age
of 30–50 years and annual mammography and clinical
breast examination from the age of 50 years.90 Bilateral
risk reducing mastectomy may be considered. Individuals with a family history of breast cancer under the age
of 35 years may require modified screening plans. A
pelvic examination, endoscopy and pap smear by a
gynaecologist every 2 years from the age of 18 years is
also recommended.90
Conclusion
Cancer is, in essence, a genetic disease.91 There is an
increasing focus on categorising cancers according to
their molecular aetiology, rather than their clinical manifestation, or phenotype, as has been done in the past.
Neither is sufficient on its own. We now know that the
same clinical picture can come about as a result of inherited mutations in different genes.55 Conversely, a mutation in the same gene can give rise to different
phenotypes, for example biallelic mutations in MUTYH
can present with polyposis or CRC without polyposis.71
Classifying a cancer by its phenotype alone may put
cancers with very different patterns of inheritance and
natural histories in the same group.
© 2015 Royal Australasian College of Physicians
487
Lung et al.
This has implications for inherited CRC as the diagnostic algorithms are now more complex. Germline DNA
tests should ideally be carried out at a familial cancer
centre to enable appropriate pre-test counselling and
follow up. The Cancer Council of New South Wales
has published referral guidelines for clinicians to refer
patients who may be at risk of familial CRC. These guidelines are available on the website www.eviq.org.au and
are summarised in Table 3.
Genetic counselling should be offered before a diagnostic genetic test is carried out. Genetic testing should begin
with an affected family member36 as this will allow more
focused genetic tests to be performed subsequently on
other family members. In the event that a germline
mutation is not identified, surveillance is still recommended and tailored to the individual and family.
There has been significant progress in the understanding of the genetic mechanisms underlying familial CRC
since the discovery of the APC gene in 1987.93 However,
there remains a group of affected families for which a
causative genetic mutation has yet to be identified.
Further research is required to delineate the hereditary
factors that may account for the increased risk of CRC in
these families. Identification of the hereditary factors
References
1 Jea F. GLOBOCAN 2008 v2.0, cancer
incidence and mortality worldwide:
IARC cancerbase No. 10 [Internet].
Lyon, France: International Agency for
Research on Cancer. 2010.
2 Australian Institute of Health and
Welfare. Cancer survival and prevalence
in Australia: period estimates from 1982
to 2010. Cancer Series no. 69. Cat. no.
CAN 65. Canberra: AIHW. 2012.
3 Lichtenstein P, Holm NV, Verkasalo PK,
Iliadou A, Kaprio J, Koskenvuo M et al.
Environmental and heritable factors in
the causation of cancer: analyses of
cohorts of twins from Sweden,
Denmark, and Finland. N Engl J Med
2000; 343: 78–85.
4 Lynch HT, Lynch PM, Lanspa SJ, Snyder
CL, Lynch JF, Boland CR. Review of the
Lynch syndrome: history, molecular
genetics, screening, differential diagnosis,
and medicolegal ramifications. Clin Genet
2009; 76: 1–18.
5 Ligtenberg MJL, Kuiper RP, Chan TL,
Goossens M, Hebeda KM, Voorendt M
et al. Heritable somatic methylation and
inactivation of MSH2 in families with
Lynch syndrome due to deletion of the
6
7
8
9
10
11
Table 3 CRC and polyposis referral guidelines based on eviQ Cancer
Treatment Online92
Individual characteristics
Blood relative of a known mutation carrier
Greater than 20 cumulative adenomatous polyps at any age
Greater than 3 cumulative adenomatous polyps by 30 years of age
Greater than 30 polyps of any type at any age
Multiple Lynch or Peutz–Jeghers syndrome-related cancers in the
same individual
Isolated CRC before 50 years of age
Family history characteristics
Three Lynch syndrome-related cancer cases (CRC, small bowel,
endometrial, ovarian, gastric, brain, urothelial transitional cell
carcinoma) in 1st or 2nd-degree relatives of any age
Two CRC cases in 1st or 2nd-degree relatives before the age of 50
years
Tumour pathology characteristics
Greater than 2 hamartomatous polyps at any age
Greater than 2 juvenile polyps at any age
Gastric fundic polyposis at any age
MSI or abnormal MMR IHC on CRC before the age of 60 years
responsible for this increased risk will lead to more
streamlined counselling and management of these
affected families.
3′ exons of TACSTD1. Nat Genet 2009;
41: 112–17.
Kovacs ME, Papp J, Szentirmay Z, Otto
S, Olah E. Deletions removing the last
exon of TACSTD1 constitute a distinct
class of mutations predisposing to Lynch
syndrome. Hum Mutat 2009; 30:
197–203.
Lynch HT, De la Chapelle A. Hereditary
colorectal cancer. N Engl J Med 2003;
348: 919–32.
Kuiper RP, Vissers LELM,
Venkatachalam R, Bodmer D,
Hoenselaar E, Goossens M et al.
Recurrence and variability of germline
EPCAM deletions in Lynch syndrome.
Hum Mutat 2011; 32: 407–14.
Vaughn CP, Robles J, Swensen JJ, Miller
CE, Lyon E, Mao R et al. Clinical analysis
of PMS2: mutation detection and
avoidance of pseudogenes. Hum Mutat
2010; 31: 588–93.
Bonadona V, Bonaïti B, Olschwang S,
Grandjouan S, Huiart L, Longy M et al.
Cancer risks associated with germline
mutations in mlh1, msh2, and msh6
genes in Lynch syndrome. JAMA 2011;
305: 2304–10.
Senter L, Clendenning M, Sotamaa K,
Hampel H, Green J, Potter JD et al. The
12
13
14
15
clinical phenotype of Lynch syndrome
due to germ-line PMS2 mutations.
Gastroenterology 2008; 135: 419–28,
e1.
Peltomaki P, Vasen HFA, Bisgaard ML,
Buerstedde JM, Friedl W, Grandjouan S
et al. Mutations predisposing to
hereditary nonpolyposis colorectal
cancer: database and results of a
collaborative study. Gastroenterology
1997; 113: 1146–58.
Green J, O’Driscoll M, Barnes A, Maher
E, Bridge P, Shields K et al. Impact of
gender and parent of origin on the
phenotypic expression of hereditary
nonpolyposis colorectal cancer in a large
newfoundland kindred with a common
MSH2 mutation. Dis Colon Rectum 2002;
45: 1223–32.
Hampel H, Frankel WL, Martin E,
Arnold M, Khanduja K, Kuebler P et al.
Screening for the Lynch syndrome
(hereditary nonpolyposis colorectal
cancer). N Engl J Med 2005; 352:
1851–60.
Aaltonen LA, Peltomaki P, Leach FS,
Sistonen P, Pylkkanen L, Mecklin JP
et al. Clues to the pathogenesis of
familial colorectal cancer. Science 1993;
260: 812–16.
© 2015 Royal Australasian College of Physicians
488
Familial colorectal cancer
16 Peltomaki P, Lothe RA, Aaltonen LA,
Pylkkanen L, Nystrom-Lahti M, Seruca
R et al. Microsatellite instability is
associated with tumors that characterize
the hereditary non-polyposis colorectal
carcinoma syndrome. Cancer Res 1993;
53: 5853–5.
17 Duval A, Hamelin R. Mutations at
coding repeat sequences in mismatch
repair-deficient human cancers: toward
a new concept of target genes for
instability. Cancer Res 2002; 62:
2447–54.
18 Tannergård P, Liu T, Weger A,
Nordenskjöld M, Lindblom A.
Tumorigenesis in colorectal tumors from
patients with hereditary non-polyposis
colorectal cancer. Hum Genet 1997; 101:
51–5.
19 Kempers MJE, Kuiper RP, Ockeloen
CW, Chappuis PO, Hutter P, Rahner N
et al. Risk of colorectal and endometrial
cancers in EPCAM deletion-positive
Lynch syndrome: a cohort study. Lancet
Oncol 2011; 12: 49–55.
20 Hampel H, Stephens JA, Pukkala E,
Sankila R, Aaltonen LA, Mecklin J-P
et al. Cancer risk in hereditary
nonpolyposis colorectal cancer
syndrome: later age of onset.
Gastroenterology 2005; 129: 415–21.
21 Watson P, Vasen HF, Mecklin JP,
Bernstein I, Aarnio M, Jarvinen HJ
et al. The risk of extra-colonic,
extra-endometrial cancer in the Lynch
syndrome. Int J Cancer 2008; 123:
444–9.
22 Umar A, Boland CR, Terdiman JP,
Syngal S, de la Chapelle A, Rüschoff J
et al. Revised Bethesda guidelines for
hereditary nonpolyposis colorectal
cancer (Lynch syndrome) and
microsatellite instability. J Natl Cancer
Inst 2004; 96: 261–8.
23 Hampel H. Point: justification for Lynch
syndrome screening among all patients
with newly diagnosed colorectal cancer.
J Natl Compr Canc Netw 2010; 8:
597–601.
24 Vasen HFA, Watson P, Mecklin JP,
Lynch HT. New clinical criteria for
hereditary nonpolyposis colorectal
cancer (HNPCC, Lynch syndrome)
proposed by the International
Collaborative Group on HNPCC.
Gastroenterology 1999; 116: 1453–6.
25 Hampel H, Frankel WL, Martin E,
Arnold M, Khanduja K, Kuebler P et al.
Feasibility of screening for Lynch
syndrome among patients with
26
27
28
29
30
31
32
33
34
colorectal cancer. J Clin Oncol 2008; 26:
5783–8.
Hendriks YMC, Wagner A, Morreau H,
Menko F, Stormorken A, Quehenberger
F et al. Cancer risk in hereditary
nonpolyposis colorectal cancer due to
MSH6 mutations: impact on counseling
and surveillance. Gastroenterology 2004;
127: 17–25.
Raevaara TE, Korhonen MK, Lohi H,
Hampel H, Lynch E, Lönnqvist KE et al.
Functional significance and clinical
phenotype of nontruncating mismatch
repair variants of MLH1. Gastroenterology
2005; 129: 537–49.
Jenkins MA, Hayashi S, O’Shea A-M,
Burgart LJ, Smyrk TC, Shimizu D et al.
Pathology features in Bethesda
guidelines predict colorectal cancer
microsatellite instability: a
population-based study. Gastroenterology
2007; 133: 48–56.
Domingo E, Laiho P, Ollikainen M,
Pinto M, Wang L, French AJ et al. BRAF
screening as a low-cost effective strategy
for simplifying HNPCC genetic testing.
J Med Genet 2004; 41: 664–8.
De Jong AE, Hendriks YMC, Kleibeuker
JH, De Boer SY, Cats A, Griffioen G et al.
Decrease in mortality in Lynch
syndrome families because of
surveillance. Gastroenterology 2006; 130:
665–71.
Renkonen-Sinisalo L, Aarnio M,
Mecklin JP, Jarvinen HJ. Surveillance
improves survival of colorectal cancer in
patients with hereditary nonpolyposis
colorectal cancer. Cancer Detect Prev 2000;
24: 137–42.
Vasen HFA, van Ballegooijen M,
Buskens E, Kleibeuker JK, Taal BG,
Griffioen G et al. A cost-effectiveness
analysis of colorectal screening for
hereditary nonpolyposis colorectal
carcinoma gene carriers. Cancer 1998;
82: 1632–7.
Järvinen HJ, Aarnio M, Mustonen H,
Aktan-Collan K, Aaltonen LA, Peltomäki
P et al. Controlled 15-year trial on
screening for colorectal cancer in
families with hereditary nonpolyposis
colorectal cancer. Gastroenterology 2000;
118: 829–34.
Vasen HF, Blanco I, Aktan-Collan K,
Gopie JP, Alonso A, Aretz S et al.
Revised guidelines for the clinical
management of Lynch syndrome
(HNPCC): recommendations by a group
of European experts. Gut 2013; 62:
812–23.
35 Vasen HF, Abdirahman M, Brohet R,
Langers AM, Kleibeuker JH, van
Kouwen M et al. One to 2-year
surveillance intervals reduce risk of
colorectal cancer in families with Lynch
syndrome. Gastroenterology 2010; 138:
2300–6.
36 Balmaña J, Balaguer F, Cervantes A,
Arnold D, ESMO Guidelines Working
Group. Familial risk-colorectal cancer:
ESMO Clinical Practice Guidelines.
Ann Oncol 2013; 24(Suppl 6): vi73–80.
37 Renkonen-Sinisalo L, Bützow R,
Leminen A, Lehtovirta P, Mecklin J-P,
Järvinen HJ. Surveillance for
endometrial cancer in hereditary
nonpolyposis colorectal cancer
syndrome. Int J Cancer 2007; 120:
821–4.
38 Stuckless S, Green J, Dawson L, Barrett
B, Woods MO, Dicks E et al. Impact of
gynecological screening in Lynch
syndrome carriers with an MSH2
mutation. Clin Genet 2013; 83:
359–64.
39 Parry S, Win AK, Parry B, Macrae FA,
Gurrin LC, Church JM et al.
Metachronous colorectal cancer risk for
mismatch repair gene mutation carriers:
the advantage of more extensive colon
surgery. Gut 2011; 60: 950–7.
40 Pollett WG, Marion K, Moeslein G,
Schneider C, Parry S, Veysey K et al.
Quality of life after surgery in
individuals with familial colorectal
cancer: does extended surgery have an
adverse impact? ANZ J Surg 2013; 84:
359–64.
41 Burn J, Gerdes A-M, Macrae F, Mecklin
J-P, Moeslein G, Olschwang S et al.
Long-term effect of aspirin on cancer
risk in carriers of hereditary colorectal
cancer: an analysis from the CAPP2
randomised controlled trial. Lancet 2011;
378: 2081–7.
42 Wimmer K, Etzler J. Constitutional
mismatch repair-deficiency syndrome:
have we so far seen only the tip of
an iceberg? Hum Genet 2008; 124:
105–22.
43 Felton KEA, Gilchrist DM, Andrew SE.
Constitutive deficiency in DNA
mismatch repair: is it time for Lynch III?
Clin Genet 2007; 71: 499–500.
44 Jasperson KW, Samowitz WS, Burt RW.
Constitutional mismatch
repair-deficiency syndrome presenting as
colonic adenomatous polyposis: clues
from the skin. Clin Genet 2011; 80:
394–7.
© 2015 Royal Australasian College of Physicians
489
Lung et al.
45 Bisgaard ML, Fenger K, Bülow S,
Niebuhr E, Mohr J. Familial
adenomatous polyposis (FAP):
frequency, penetrance, and mutation
rate. Hum Mutat 1994; 3: 121–5.
46 Björk J, Akerbrant H, Iselius L, Alm T,
Hultcrantz R. Epidemiology of familial
adenomatous polyposis in Sweden:
changes over time and differences
in phenotype between males and
females. Scand J Gastroenterol 1999; 34:
1230–5.
47 Fearnhead NS, Britton MP, Bodmer WF.
The ABC of APC. Hum Mol Genet 2001;
10: 721–33.
48 Nagase H, Miyoshi Y, Horii A, Aoki T,
Ogawa M, Utsunomiya J et al.
Correlation between the location of
germ-line mutations in the APC gene
and the number of colorectal polyps in
familial adenomatous polyposis patients.
Cancer Res 1992; 52: 4055–7.
49 Bertario L, Russo A, Sala P, Varesco L,
Giarola M, Mondini P et al. Multiple
approach to the exploration of
genotype-phenotype correlations in
familial adenomatous polyposis. J Clin
Oncol 2003; 21: 1698–707.
50 Bertario L, Russo A, Sala P, Eboli M,
Giarola M, D’Amico F et al. Genotype
and phenotype factors as determinants
of desmoid tumors in patients with
familial adenomatous polyposis. Int J
Cancer 2001; 95: 102–7.
51 Luijt R, Vasen HA, Tops CJ, Breukel C,
Fodde R, Khan PM. APC mutation in
the alternatively spliced region of exon
9 associated with late onset familial
adenomatous polyposis. Hum Genet
1995; 96: 705–10.
52 Matsubara N, Isozaki H, Tanaka N. The
farthest 3′ distal end APC mutation
identified in attenuated adenomatous
polyposis coli with extracolonic
manifestations. Dis Colon Rectum 2000;
43: 720–1.
53 Spirio L, Olschwang S, Groden J,
Robertson M, Samowitz W, Joslyn G
et al. Alleles of the APC gene: an
attenuated form of familial polyposis.
Cell 1993; 75: 951–7.
54 Knudsen AL, Bulow S, Tomlinson I,
Moslein G, Heinimann K, Christensen IJ
et al. Attenuated familial adenomatous
polyposis: results from an international
collaborative study. Colorectal Dis 2010;
12: e243–9.
55 Filipe B, Baltazar C, Albuquerque C,
Fragoso S, Lage P, Vitoriano I et al. APC
or MUTYH mutations account for the
56
57
58
59
60
61
62
63
64
65
66
majority of clinically well-characterized
families with FAP and AFAP phenotype
and patients with more than 30
adenomas. Clin Genet 2009; 76:
242–55.
Groen E, Roos A, Muntinghe F, Enting
R, Vries J, Kleibeuker J et al.
Extra-intestinal manifestations of
familial adenomatous polyposis. Ann
Surg Oncol 2008; 15: 2439–50.
Trainer AH. Extra-colonic manifestations
of familial adenomatous polyposis
coli. Adv Exp Med Biol 2009; 656:
119–27.
Lipton L, Tomlinson I. The genetics of
FAP and FAP-like syndromes. Fam
Cancer 2006; 5: 221–6.
Heiskanen I, Luostarinen T, Järvinen HJ.
Impact of screening examinations on
survival in familial adenomatous
polyposis. Scand J Gastroenterol 2000; 35:
1284–7.
Spigelman AD, Williams CB, Talbot IC,
Domizio P, Phillips RK. Upper
gastrointestinal cancer in patients with
familial adenomatous polyposis. Lancet
1989; 2: 783–5.
Vasen HFA, Möslein G, Alonso A, Aretz
S, Bernstein I, Bertario L et al.
Guidelines for the clinical management
of familial adenomatous polyposis (FAP).
Gut 2008; 57: 704–13.
Gega M, Yanagi H, Yoshikawa R, Noda
M, Ikeuchi H, Tsukamoto K et al.
Successful chemotherapeutic modality of
doxorubicin plus dacarbazine for the
treatment of desmoid tumors in
association with familial adenomatous
polyposis. J Clin Oncol 2006; 24:
102–5.
Bussey HJR, Decosse JJ, Deschner EE,
Eyers AA, Lesser ML, Morson BC et al. A
randomized trial of ascorbic acid in
polyposis coli. Cancer 1982; 50:
1434–9.
Giardiello FM, Hamilton SR, Krush AJ,
Piantadosi S, Hylind LM, Celano P et al.
Treatment of colonic and rectal
adenomas with sulindac in familial
adenomatous polyposis. N Engl J Med
1993; 328: 1313–16.
Steinbach G, Lynch PM, Phillips RKS,
Wallace MH, Hawk E, Gordon GB et al.
The effect of celecoxib, a
cyclooxygenase-2 inhibitor, in familial
adenomatous polyposis. N Engl J Med
2000; 342: 1946–52.
Lynch PM. Pharmacotherapy for
inherited colorectal cancer. Expert Opin
Pharmacother 2010; 11: 1101–8.
67 Sieber OM, Lipton L, Crabtree M,
Heinimann K, Fidalgo P, Phillips RKS
et al. Multiple colorectal adenomas,
classic adenomatous polyposis, and
germ-line mutations in MYH. N Engl J
Med 2003; 348: 791–9.
68 Al-Tassan N, Chmiel NH, Maynard J,
Fleming N, Livingston AL, Williams GT
et al. Inherited variants of MYH
associated with somatic G:C→T:A
mutations in colorectal tumors. Nat
Genet 2002; 30: 227–32.
69 Balaguer F, Castellví-Bel S, Castells A,
Andreu M, Muñoz J, Gisbert JP et al.
Identification of MYH mutation carriers
in colorectal cancer: a multicenter,
case-control, population-based study.
Clin Gastroenterol Hepatol 2007; 5:
379–87.
70 Grover S, Kastrinos F, Steyerberg EW,
Cook EF, Dewanwala A, Burbidge LA
et al. Prevalence and phenotypes of APC
and MUTYH mutations in patients with
multiple colorectal adenomas. JAMA
2012; 308: 485–92.
71 Farrington SM, Tenesa A, Barnetson R,
Wiltshire A, Prendergast J, Porteous M
et al. Germline susceptibility to colorectal
cancer due to base-excision repair gene
defects. Am J Hum Genet 2005; 77:
112–19.
72 Terdiman JP. MYH-associated disease:
attenuated adenomatous polyposis of
the colon is only part of the story.
Gastroenterology 2009; 137: 1883–6.
73 Vogt S, Jones N, Christian D, Engel C,
Nielsen M, Kaufmann A et al. Expanded
extracolonic tumor spectrum in
MUTYH-associated polyposis.
Gastroenterology 2009; 137: 1976–85, e10.
74 Boparai KS, Dekker E, van Eeden S,
Polak MM, Bartelsman JFWM,
Mathus-Vliegen EMH et al. Hyperplastic
polyps and sessile serrated adenomas as
a phenotypic expression of
MYH-associated polyposis.
Gastroenterology 2008; 135: 2014–18.
75 Cheadle JP, Sampson JR.
MUTYH-associated polyposis – from
defect in base excision repair to clinical
genetic testing. DNA Repair (Amst) 2007;
6: 274–9.
76 Lubbe SJ, Di Bernardo MC, Chandler IP,
Houlston RS. Clinical Implications of the
colorectal cancer risk associated with
MUTYH mutation. J Clin Oncol 2009; 27:
3975–80.
77 Sampson JR, Dolwani S, Jones S, Eccles
D, Ellis A, Evans DG et al. Autosomal
recessive colorectal adenomatous
© 2015 Royal Australasian College of Physicians
490
Familial colorectal cancer
78
79
80
81
82
83
polyposis due to inherited mutations of
MYH. Lancet 2003; 362: 39–41.
Jones N, Vogt S, Nielsen M, Christian D,
Wark PA, Eccles D et al. Increased
colorectal cancer incidence in obligate
carriers of heterozygous mutations in
MUTYH. Gastroenterology 2009; 137:
489–94, e1.
Cleary SP, Cotterchio M, Jenkins MA,
Kim H, Bristow R, Green R et al.
Germline MutY human homologue
mutations and colorectal cancer: a
multisite case-control study.
Gastroenterology 2009; 136: 1251–60.
Win A, Hopper J, Jenkins M.
Association between monoallelic
MUTYH mutation and colorectal cancer
risk: a meta-regression analysis. Fam
Cancer 2011; 10: 1–9.
Worthley DL, Suthers G, Lipton L.
Management of MUTYH-associated
neoplasia in Australia. Intern Med J 2008;
38: 644–50.
Schreibman IR, Baker M, Amos C,
McGarrity TJ. The hamartomatous
polyposis syndromes: a clinical and
molecular review. Am J Gastroenterol
2005; 100: 476–90.
Jasperson KW, Tuohy TM, Neklason
DW, Burt RW. Hereditary and familial
colon cancer. Gastroenterology 2010; 138:
2044–58.
84 Sayed MG, Ahmed AF, Ringold JR,
Anderson ME, Bair JL, Mitros FA
et al. Germline SMAD4 or BMPRIA
mutations and phenotype of juvenile
polyposis. Ann Surg Oncol 2002; 9: 901–6.
85 Calva-Cerqueira D, Chinnathambi S,
Pechman B, Bair J, Larsen-Haidle J,
Howe JR. The rate of germline
mutations and large deletions of SMAD4
and BMPR1A in juvenile polyposis. Clin
Genet 2009; 75: 79–85.
86 Aretz S, Stienen D, Uhlhaas S, Stolte M,
Entius MM, Loff S et al. High proportion
of large genomic deletions and a
genotype phenotype update in 80
unrelated families with juvenile
polyposis syndrome. J Med Genet 2007;
44: 702–9.
87 Cancer Institute NSW. eviQ cancer
treatments online. [updated 2014 Aug 7;
cited 2014 Aug 25]. Available from URL:
https://www.eviq.org.au/Protocol/tabid/
66/categoryid/66/id/751/Risk+
Management+for+Juvenile+Polyposis+
Syndrome.aspx
88 Hearle N, Schumacher V, Menko FH,
Olschwang S, Boardman LA, Gille JJP
et al. Frequency and spectrum of cancers
in the Peutz-Jeghers syndrome. Clin
Cancer Res 2006; 12: 3209–15.
89 Westerman AM, Entius MM, de Baar E,
Boor PPC, Koole R, van Velthuysen MLF
90
91
92
93
et al. Peutz-Jeghers syndrome: 78-year
follow-up of the original family. Lancet
1999; 353: 1211–15.
Cancer Institute NSW. eviQ cancer
treatments online. [updated 2014 Jul 9;
cited 2014 Aug 25]. Available from
URL: https://www.eviq.org.au/Protocol/
tabid/66/categoryid/66/id/395/Risk
+Management+for+Peutz-Jeghers
+Syndrome++.aspx
Vogelstein B, Kinzler KW. Cancer genes
and the pathways they control. Nat Med
2004; 10: 789–99.
Cancer Institute NSW. eviQ cancer
treatments online. [updated 2012 May
31; cited 2014 Aug 25]. Available from
URL: https://www.eviq.org.au/Protocol/
tabid/66/categoryid/439/id/657/Cancer
+Genetics+-+Colorectal+Cancer+and
+Polyposis+Referral+Guidelines.aspx
Mallinson EKL, Newton KF, Bowen J,
Lalloo F, Clancy T, Hill J et al. The
impact of screening and genetic
registration on mortality and colorectal
cancer incidence in familial
adenomatous polyposis. Gut 2010; 59:
1378–82.
© 2015 Royal Australasian College of Physicians
491
Internal Medicine Journal 45 (2015)
O R I G I N A L A RT I C L E S
Clinical triage for colonoscopy is useful in young women
K. D. Williamson,1,2,3 K. Steveling,4 G. Holtmann,5,6 M. Schoeman7 and J. M. Andrews3,8
1
Translational Gastroenterology Unit, John Radcliffe Hospital, 2Nuffield Department of Medicine, Oxford University, Oxford, UK, and 3Discipline of
Medicine, University of Adelaide, Adelaide, 5Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, 6Faculty of
Medicine and Biomedical Sciences, University of Queensland, Queensland and 7Gastrointestinal Endoscopic Services, Department of Gastroenterology
and Hepatology and 8Inflammatory Bowel Disease Service and Education, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital,
South Australia, Australia, and 4Department of Rheumatology, Knappschaftskrankenhaus, Püttlingen, Germany
Key words
colonoscopy, endoscopy, diagnosis, human,
female.
Correspondence
Kate Williamson, Translational Gastroenterology
Unit, John Radcliffe Hospital, Oxford, UK.
Email: [email protected]
Received 1 July 2014; accepted 28 January
2015.
doi:10.1111/imj.12703
Abstract
Background: Colonoscopy is an invasive procedure and a limited resource. It is therefore desirable to restrict its use to those in whom it yields an important diagnosis,
without missing pathology in others.
Aim: The aim of this study was to determine whether standard clinical criteria can be
used to reliably distinguish when colonoscopy is advisable in women 30 years and
younger.
Methods: A retrospective audit was performed at a single centre of 100 consecutive
colonoscopies performed in women 30 years old and younger. The indications for the
colonoscopy were recorded, and divided into clear and relative indications. The primary
outcome of whether an endoscopic diagnosis was made was compared between the two
groups. Clear indications for colonoscopy included overt rectal bleeding, elevated
inflammatory markers, anaemia, iron deficiency and strong family history of colorectal
cancer. Relative indications included abdominal pain or discomfort, bloating and altered
bowel habit/motions.
Results: The average age was 23 years. Sixty women had both relative and clear
indications. Eleven had only clear indications and 28 only relative indications. Altogether, 58 colonoscopies were normal, and 17 showed inflammatory bowel disease. No
subject with only relative indications had an abnormal finding (0/28). The diagnostic
yield was significantly different between those with only relative indications (0%)
versus those with at least one clear indication (59%; P < 0.0001).
Conclusions: Standard clinical criteria can be used to restrict safely the use of
colonoscopy in young women. This will avoid performing procedures in people without
clear indications, saving costs, resources and complications.
Introduction
The indications for colonoscopy are clearly defined by
various organisations.1 However, what is not clear is
whether we can use standard clinical criteria in low-risk
populations to restrict reliably the use of colonoscopy.
Colonoscopy is an invasive procedure with a documented
rate of serious complications, such as bleeding and
Funding/Conflict of interest: K. D. Williamson receives funding
from Merck Sharp & Dohme to carry out clinical research. J. M.
Andrews is in receipt of research support, lecture fees and advisory board membership from AbbVie, Abbott, Janssen, Ferring,
Nycomed, Orphan, Shire, Amgen, ImmunosanT, AstraZeneca,
Takeda, Hospira.
perforation, varying from 0.2–0.3%.2 Despite the risks of
the procedure staying relatively stable with age, the
pick-up rate of a serious finding is far lower in younger as
compared with older subjects – even without considering
symptoms. For example, the chance of a 30-year-old
patient presenting with colorectal cancer within the next
5 years is 1 in 7000, as compared with 1 in 100 for a 60
year old.3 In addition, resources for colonoscopy are
limited, and there are significant costs associated with it,
both direct and indirect.4
Common reasons given for requesting a colonoscopy,
especially in the young, are symptoms pertaining to irritable bowel syndrome (IBS) or other functional abdominal symptoms, with up to a quarter of colonoscopies
being performed in those with IBS-related symptoms.5
© 2015 Royal Australasian College of Physicians
492
Colonoscopy in young women
This is despite recommendations that a positive diagnosis
of IBS should be made on clinical grounds, with
colonoscopy specifically not indicated in those less than
50 years with typical IBS symptoms and no alarm
features.6
Therefore, it is important, based on a large number of
personal and population-based factors, to restrict the use
of colonoscopy to those for whom it is genuinely medically indicated. This has not been studied in a young
population. Despite this, we know anecdotally that many
individual clinicians find it hard to resist actively performing a procedure as they worry about the implications
of a missed diagnosis. Therefore, we set out to determine
the ability of standard clinical criteria to predict reliably a
clinical finding in young patients undergoing diagnostic
colonoscopy. Given that functional disorders, particularly
IBS, are more prevalent in females, we focussed on
females 30 years old and younger.
Methods
A retrospective audit was carried out of the last 100
consecutive colonoscopies in women aged 30 years old or
younger. The audit was carried out at the Royal Adelaide
Hospital, a large metropolitan teaching hospital in South
Australia. The endoscopy unit has an annual load of
approximately 1800 colonoscopies, with roughly 350
colonoscopies on the waiting list for a 6-month period.
Reports were obtained from the electronic recording
system, ‘Endoscribe’. Inclusion criteria were colonoscopy
reports of patients who were female and aged 30 years
old or less at the time of colonoscopy. There were no
exclusion criteria. The clinical indications documented on
the colonoscopy report, the request form or the case
notes were recorded as were any available blood test
results pre-procedure which were obtained from electronic sources and case notes. The clinical diagnosis, as
well as progress and complications immediately postcolonoscopy, was obtained from the same sources.
Indications for colonoscopy were classified as either
‘clear’ or ‘relative’ as generally accepted medically
(Table 1). Clear indications included overt rectal bleeding, elevated inflammatory markers, anaemia, iron deficiency or a strong family history affording at least
moderately elevated risk of colorectal cancer (according
to Australia’s National Health and Medical Record
Council (NHMRC) guidelines3). Relative indications
included abdominal pain or discomfort, bloating and
altered bowel habit.
The results were analysed to assess the diagnostic yield
of colonoscopies stratified according to indication
(relative vs clear). STATA version 10.0 (Statcorp, Texas,
USA) was used for statistical analysis. Binary variables
Table 1 Clear and relative indications†
Clear indications
Overt rectal bleeding
Iron deficiency
Anaemia
Elevated inflammatory markers
(temperature, ESR, CRP, WBC or
platelets)
Strong family history of colorectal
cancer
Relative indications
Abdominal pain or discomfort
Bloating
Altered bowel habit/motions
†
As per NHMRC guidelines.3 CRP, C-reactive protein; ESR, erythrocyte
sedimentation rate; WBC, white blood cells.
were arranged into two by two contingency tables, from
which positive predictive value, negative predictive
value, sensitivity and specificity were derived. Comparisons were made using Pearson’s Chi-squared statistics for
categorical variables. The study was approved by the
Royal Adelaide Hospital Human Research Ethics Committee as a clinical audit.
Results
One hundred sequential colonoscopy records were
reviewed. The average age of patients was 23 years
(range 16–30). Of the 100 patients, 11 patients had clear
indications, 28 patients had only relative indications, and
60 patients had clear as well relative indications. In one
case, insufficient data were available to determine the
indications fully. In the analysis, this case was assumed to
have a clear indication as it could not be safely judged
that she did not. Thus, in total, 72 patients had at least
one clear indication.
The most commonly noted relative indications were:
abdominal pain (n = 64), chronic diarrhoea (n = 33),
acute diarrhoea (n = 17), constipation (n = 14) and alternating stool form/frequency (n = 17). Of the clear indications, the most common was overt rectal bleeding (n =
48), followed by iron deficiency (n = 15), 11 of whom
were also anaemic (Table 2).
In the 100 colonoscopy reports audited, 58 procedures
were reported as normal. The most common single
abnormal finding was inflammatory bowel disease (IBD)
(n = 17). Other diagnoses included polyps, haemorrhoids,
anal fissure, rectal prolapse and one rectal carcinoma
(Table 3). In this cohort, no subject had more than one
diagnosis.
Of those with only relative indications (n = 28)
none had a positive finding at colonoscopy (0%, 95%
confidence interval (CI) 0–12.1%)). Of those with any
clear indication (n = 72), a positive finding was noted in
© 2015 Royal Australasian College of Physicians
493
Williamson et al.
Table 2 Distribution of indications
Indications for colonoscopy
Relative indications
Abdominal pain
Chronic diarrhoea
Acute diarrhoea
Constipation
Alternating bowel motions
Clear indications
Overt rectal bleeding
Iron deficiency
Anaemia
Family history CRC
n
64
33
17
14
17
48
15
11
0
Note that n > 100, as some patients had more than one indication. CRC,
colorectal cancer.
42/72 (58%, 95% CI 47–69%)). This difference in yield
between those with only relative versus any clear indication was statistically significant (P < 0.0001). The positive predictive value of an abnormal colonoscopy with
any clear indication was 58.5%, and the negative predictive value in the absence of any clear indication(s) was
100%. Even if findings of a non-serious nature were
excluded (n = 10: haemorrhoids, anal fissure or prolapse
– all of which could have been diagnosed with
proctoscopy or sigmoidoscopy), the difference in yield
remained highly clinically relevant and statistically significant – 0% versus 44% (P < 0.0001).
It is important to note that in those without any ‘clear’
indication for colonoscopy, no pathology would have
been missed if the colonoscopy had been avoided. The
single patient with rectal carcinoma presented with overt
rectal bleeding. It was also noted that a higher number of
clear indications denoted a higher chance of an abnormal
finding at colonoscopy (Table 3). Specifically, of the 17
patients with a finding of IBD, all had at least two clear
indications, and 11 of the 17 had three or more clear
indications.
Table 3 Findings at colonoscopy
Outcome colonoscopy
Normal
Inflammatory bowel disease
Polyp
Haemorrhoids
Anal fissure
Prolapse
Histological inflammation
Cancer†
Total
Number of clear indications
Total
0
1
2
3
4
5
6
28
–
–
–
–
–
–
–
28
21
–
6
2
3
3
3
1
39
7
6
–
1
–
–
3
–
17
2
5
–
1
–
–
1
–
9
–
2
–
–
–
–
1
–
3
–
1
–
–
–
–
–
–
1
–
3
–
–
–
–
–
–
3
†Presentation for this patient was overt rectal bleeding.
58
17
6
4
3
3
8
1
100
Two patients in the study had significant morbidity
associated with the procedure, predominantly pain postprocedure. Both patients had presented with only relative indications and had normal colonoscopies. One
patient required imaging, opiate analgesia and hospital
admission. As expected in such a small cohort, there
were no reported perforations or clinically significant
haemorrhage.
Discussion
In this audit, it was found that, of consecutive women
under 30 who had undergone colonoscopy in our unit,
42% overall had a positive finding. This is a very high
yield compared with what one might expect. This is likely
to be so, due to the fact that many patients were referred
for clinically accepted, clear indication(s). Therefore, our
data indicate that even in a young woman, any clear
indication (such as anaemia, bleeding or raised inflammatory markers) should justify a prompt referral.
However, perhaps the more interesting finding is that
the simple application of well-accepted triage criteria
would allow further limitation of colonoscopy in this
demographic, without any loss of safety or accuracy. This
would potentially free up some colonoscopy resources to
be used in those who would benefit most, and would
save unnecessary risk or morbidity in those who have no
medical need for the procedure. However, it needs to be
noted that only 28% of our colonoscopy cohort fell into
this group. Thus, utilising the criteria, only 28% of
colonoscopies could be avoided.
The diagnostic yield of colonoscopy varies greatly
depending on indication. One study of 736 colonoscopies
(average age 43.6 years), examined diagnostic yield of
colonoscopy according to American Society of Gastrointestinal Endoscopy (ASGE) indications.7 The yield ranged
from 38% for indications considered ‘generally indicated’, 20% for indications ‘generally not indicated’ and
13% for indications ‘not listed’. Generally indicated indications included such things as haematochezia, unexplained iron deficiency anaemia and acceptable colonic
cancer screening or surveillance, whereas ‘generally not
indicated’ included ‘chronic, stable, irritable bowel syndrome or chronic abdominal pain’. If we examine the
diagnostic yield of the patients in our study, we have
figures of 58% for those with a clear (or appropriate)
indication, and 0% for those with only relative indications. In a demographic where the prevalence of IBS is
relatively high, this audit suggests a conservative
approach be taken in the investigation of young women
with only relative symptoms, rather than referring
directly to colonoscopy given the absent diagnostic yield.
The fact that there were complications in two of the 28
© 2015 Royal Australasian College of Physicians
494
Colonoscopy in young women
(7%) patients coming to colonoscopy with only relative
indications is concerning enough in our audit to recommend holding off endoscopic evaluation in this low-risk
population.
A Scandinavian retrospective study analysed diagnostic
yield at colonoscopy according to indication/symptom.
They had a similar finding to us, with a high diagnostic
yield for clear indications, such as bleeding (67%) and
weight loss (33%), and a low diagnostic yield for patients
with non-specific gastrointestinal (GI) symptoms, including abdominal pain, change in bowel habit, constipation,
flatulence and weight loss (13.2%).8 We feel that weight
loss is a more clear indication for colonoscopy, rather
than a non-specific symptom, and the yield in this setting
was higher. They did find a significant yield for nonbloody diarrhoea – 31.2% of 176 patients. However, they
failed to mention whether there were abnormalities in
inflammatory markers, haemoglobin or weight loss,
which we feel may well have contributed to increase the
pre-test probability. Additionally, the mean age of their
patients was 54 years old, with half being male, two
factors which may arguably increase diagnostic yield. We
are not saying that diarrhoea is not an appropriate indication for colonoscopy, but rather that diarrhoea alone
may have a low diagnostic yield in this younger female
demographic, as supported by our findings, and that supportive features for organic pathology should be sought
before considering endoscopic investigation.
There is one caveat to mention regarding the above,
and that is to identify the minor subset of patients who
present with diarrhoea who may have a form of microscopic colitis (collagenous or lymphocytic). This is a diagnosis made in predominantly middle-aged to older
women (average age at diagnosis 53–69 years), and
invariably patients have very watery and frequent stools,
with a frequency above four times per day, and often the
presence of nocturnal diarrhoea.9 These specific features
of high frequency and nocturnal diarrhoea are unusual in
IBS. This small subset of patients would be best served
with a flexible sigmoidoscopy (rather than colonoscopy)
with colonic biopsies to look for this diagnosis, as recent
literature suggests that left-sided colonic biopsies are just
as sensitive as full colonic series biopsies in making a
diagnosis of microscopic colitis.10
The ASGE guidelines give recommendations for indications and contraindications for colonoscopy, as well as
settings in which colonoscopy is ‘generally not recommended’. As mentioned above, falling under this latter
heading is ‘chronic, stable, irritable bowel syndrome or
chronic abdominal pain’. Twenty-eight patients in our
audit had only relative indications, and based on our data
it might be concluded that colonoscopy may have been
reasonably avoided in these patients without missing
pathology. On the other hand, our study did not assess
the impact of the colonoscopy for the management of
these patients. We cannot exclude that the potential
‘therapeutic effects’ due to the reassurance of a normal
colonoscopy actually has a relevant influence on the
outcome or healthcare utilisation that would justify the
procedure.
If a colonoscopy is normal in a patient with symptoms,
it is possible that the very fact of doing the procedure may
make it more difficult to help the patient come to terms
with a functional diagnosis. The flip side of this is that a
negative colonoscopy in a young female may provide
reassurance, although this is unproven. Pertinently, this
issue has been addressed in two previous studies with
conflicting results. A prospective study of 59 patients
aged less than 50 years old undergoing colonoscopy
found a mean reduction in anxiety scores as well as
symptom scores, despite a minimal diagnostic yield.11
However, a second retrospective study of 458 patients
under 50 years old with IBS found the opposite, with no
association between a negative colonoscopy and reassurance or health-related quality of life.12 Hence, the justification of doing a colonoscopy in a young female with
only relative indications for the sake of reassurance might
not be necessarily justifiable. We additionally note that
procedure-related pain may be higher in those with functional GI disorders, such as IBS13 – in one study, 7%
needed admission for pain post procedure – which
further justifies restriction of use of colonoscopy in this
demographic unless there are real concerns for a positive
finding.
Another observation made in this study is that the
finding of IBD at colonoscopy in this young female
population was related to the number of clear indications they possessed. All patients with IBD had at least
two clear indications, with 64% of them having three
clear indications. This indicates that one should have a
higher suspicion of IBD in the patient with multiple
clear indications prior to colonoscopy in this demographic. While colonoscopy in this group is necessary to
confirm the diagnosis, the diagnosis itself should be
anticipated.
The limitations of the study were the retrospective
nature, and the small sample size. It was therefore not
powered to look at complications, nor cost-effectiveness,
but rather yield. One should therefore note, that
although none of 28 patients with only relative indications had a significant finding, the 95% CI was 0% to
12%. Hence, based on our sample size, up to 12% of
patients may still have had a significant finding. The use
of faecal calprotectin to enhance diagnostic accuracy may
be of benefit here, by minimising the risk in not performing colonoscopy where only ‘relative’ indications exist.14
© 2015 Royal Australasian College of Physicians
495
Williamson et al.
Conclusion
In women, under 30 years of age, after simple clinical and
laboratory parameters are found to be normal,
colonoscopy can be safely avoided. These data should
encourage gastroenterologists to be confident in making
a positive diagnosis of functional GI disease on clinical
grounds in the absence of alarm symptoms (or ‘clear
References
1 ASGE Standards of Practice Committee.
Appropriate use of GI endoscopy.
Gastrointest Endosc 2012; 75: 1127–31.
2 Dominitz JA, Eisen GM, Baron TH,
Goldstein JL, Hirota WK, Jacobson BC
et al. Complications of colonoscopy.
Gastrointest Endosc 2003; 57: 441–5.
3 Australian Cancer Network Colorectal
Cancer Guidelines Revision Committee.
Guidelines for the Prevention, Early Detection
and Management of Colorectal Cancer.
Sydney: The Cancer Council Australia
and Australian Cancer Network;
2005.
4 Sharara N, Adam V, Crott R, Barkun
AN. The costs of colonoscopy in a
Canadian hospital using a microcosting
approach. Can J Gastroenterol 2008; 22:
565–70.
5 Lieberman DA, Holub J, Eisen G,
Kraemer D, Morris CD. Utilization of
colonoscopy in the United States: results
from a national consortium. Gastrointest
Endosc 2005; 62: 875–83.
indications’), and reserve colonoscopy for subjects in
whom the medical need for the procedure is greater.
Acknowledgements
The authors acknowledge Dr Crispin Corte, Concord Hospital, Australia, for assistance with statistical analysis.
6 American College of Gastroenterology
Task Force on Irritable Bowel Syndrome,
Brandt LJ, Chey WD, Foxx-Orenstein
AE, Schiller LR, Schoenfeld PS et al. An
evidence-based position statement on
the management of irritable bowel
syndrome. Am J Gastroenterol 2009;
104(Suppl 1): S1–35.
7 Siddique I, Mohan K, Hasan F, Memon
A, Patty I, Al-Nakib B. Appropriateness
of indication and diagnostic yield of
colonoscopy: first report based on the
2000 guidelines of the American Society
for Gastrointestinal Endoscopy. World J
Gastroenterol 2005; 11: 7007–13.
8 Lasson A, Kilander A, Stotzer PO.
Diagnostic yield of colonoscopy based on
symptoms. Scand J Gastroenterol 2008; 43:
356–62.
9 Pardi DS, Kelly CP. Microscopic colitis.
Gastroenterology 2011; 140: 1155–65.
10 Chapman TP, Macfaul G, Abraham A.
Diagnosing microscopic colitis: is flexible
sigmoidoscopy a reliable alternative to
colonoscopy? Clin Gastroenterol Hepatol
2015; 13: 618.
11 Esfandyari T, Harewood GC. Value of a
negative colonoscopy in patients with
non-specific gastrointestinal symptoms. J
Gastroenterol Hepatol 2007; 22: 1609–14.
12 Spiegel BM, Gralnek IM, Bolus R,
Chang L, Dulai GS, Naliboff B et al. Is a
negative colonoscopy associated with
reassurance or improved health-related
quality of life in irritable bowel
syndrome? Gastrointest Endosc 2005; 62:
892–9.
13 Kim ES, Cheon JH, Park JJ, Moon CM,
Hong SP, Kim TI et al. Colonoscopy as
an adjunctive method for the diagnosis
of irritable bowel syndrome: focus on
pain perception. J Gastroenterol Hepatol
2010; 25: 1232–8.
14 Tibble J, Teahon K, Thjodleifsson B,
Roseth A, Sigthorsson G, Bridger S et al.
A simple method for assessing intestinal
inflammation in Crohn’s disease. Gut
2000; 47: 506–13.
© 2015 Royal Australasian College of Physicians
496
Comparison of the management and in-hospital outcomes of
acute coronary syndrome patients in Australia and New
Zealand: results from the binational SNAPSHOT acute coronary
syndrome 2012 audit
C. Ellis,1 C. Hammett,2 I. Ranasinghe,3 J. French,4 T. Briffa,5 G. Devlin,6 J. Elliott,7 J. Lefkovitz,8
B. Aliprandi-Costa,9 C. Astley,10 J. Redfern,3 T. Howell,11 B. Carr,12 K. Lintern,12 S. Bloomer,13 A. Farshid,14
P. Matsis,15 A. Hamer,16 M. Williams,17 R. Troughton,18 M. Horsfall,19 K. Hyun,3 G. Gamble,20 H. White,21
D. Brieger9 and D. Chew,19 on behalf of Bi-National Acute Coronary Syndromes (ACS) ‘SNAPSHOT’ Audit
Group
1
Department of Cardiology, Auckland City Hospital, 20Department of Medicine, University of Auckland, 21Department of Cardiology, Auckland City
Hospital, Auckland, 6Department of Cardiology, Waikato Hospital, Hamilton, 7Cardiology Department, Christchurch Hospital, Christchurch,
15
Cardiology Department, Wellington Hospital, Wellington, 16Department of Cardiology, Nelson Marlborough District Health Board, Nelson,
17
Cardiology Department, Dunedin Hospital, Dunedin, 18The Christchurch Heart Institute, Department of Medicine, University of Otago Christchurch,
Christchurch, New Zealand; and 2Royal Brisbane and Women’s Hospital, 11Clinical Access and Redesign Unit, Queensland Department of Health,
Brisbane, Queensland, 3The George Institute for Global Health, 4Department of Cardiology, Liverpool Hospital, 9Department of Cardiology, Concord
Hospital, 12Agency for Clinical Innovation, Sydney, 14Department of Cardiology, The Canberra Hospital, Canberra, ACT, New South Wales, 5School of
Population Health, University of Western Australia, 13Health Networks Branch, Department of Health of Western Australia, Perth, Western Australia,
8
Royal Melbourne Hospital, Melbourne, Victoria, 10Statewide Systems Strategy, SA Health, and 19Department of Cardiovascular Medicine, Flinders
University, Adelaide, South Australia, Australia
Key words
cardiology, audit, acute coronary syndrome.
Abstract
Background/Aims: We aimed to assess differences in patient management, and out-
Correspondence
Chris Ellis, Cardiology Department, Green Lane
CVS Services, Level 3, Auckland City Hospital,
Grafton, Auckland 1023, New Zealand.
Email: [email protected]
Received 20 December 2014; accepted 4
March 2015.
doi:10.1111/imj.12739
comes, of Australian and New Zealand patients admitted with a suspected or confirmed
acute coronary syndrome (ACS).
Methods: We used comprehensive data from the binational Australia and New Zealand
ACS ‘SNAPSHOT‘ audit, acquired on individual patients admitted between 00.00 h on
14 May 2012 to 24.00 h on 27 May 2012.
Results: There were 4387 patient admissions, 3381 (77%) in Australia and 1006
(23%) in New Zealand; Australian patients were slightly younger (67 vs 69 years, P =
0.0044). Of the 2356 patients with confirmed ACS, Australian patients were at a lower
cardiovascular risk with a lower median Global Registry Acute Coronary Events score
(147 vs 154 P = 0.0008), but as likely to receive an invasive coronary angiogram (58%
vs 54%, P = 0.082), or revascularisation with percutaneous coronary intervention (32%
vs 31%, P = 0.92) or coronary artery bypass graft surgery (7.0% vs 5.6%, P = 0.32). Of
the 1937 non-segment elevation myocardial infarction/unstable angina pectoris
(NSTEMI/UAP) patients, Australian patients had a shorter time to angiography (46 h vs
67 h, P < 0.0001). However, at discharge, Australian NSTEMI/UAP survivors were less
likely to receive aspirin (84% vs 89%, P = 0.0079, a second anti-platelet agent (57% vs
63%, P = 0.050) or a beta blocker (67% vs 77%, P = 0.0002). In-hospital death rates
were not different (2.7% vs 3.2%, P = 0.55) between Australia and New Zealand.
Conclusions: Overall more similarities were seen, than differences, in the management of suspected or confirmed ACS patients between Australia and New Zealand.
However, in several management areas, both countries could improve the service
delivery to this high-risk patient group.
© 2015 Royal Australasian College of Physicians
497
Ellis et al.
Introduction
The medical knowledge which directs the optimal management of patients with an acute coronary syndrome
(ACS) is the result of numerous clinical trials and is
summarised in local1–3 and international4–7 guidelines. A
key step in the management of ACS patients is for them to
access cardiac angiography in a timely manner.1–7 From
this investigation revascularisation with percutaneous
coronary intervention (PCI) or coronary artery bypass
graft (CABG) surgery can be undertaken, where appropriate. Another key area of management is in the provision of
proven secondary prevention medication and participation in a cardiac rehabilitation/secondary prevention
programme.1–7
The principal challenge for the management of ACS
patients is the translation of this robust evidence into
clinical practice.8–10 Better delivery of proven care has
more immediate potential to improve outcomes than
treatment innovations.11
To facilitate better service provision, there is a growing
recognition of the need for comparative effectiveness
research. The American Institute of Medicine identified
healthcare delivery systems and cardiovascular care as
among the highest priorities for comparative effectiveness research.12 However, there are few studies that
simultaneously examine care and outcomes between
countries,13 although international comparisons of
healthcare systems have the potential to yield important
insights and guide policies.14
Australia and New Zealand have many historical, cultural, economic and medical similarities. However, there
are also differences in the delivery of healthcare and
other variables between the two countries. For example
Australia has a major private healthcare contribution to
supplement public hospital care of ACS patients, with
significant inter-state differences,15 whereas much less
privately funded ACS care is seen in New Zealand.16
The ‘SNAPSHOT’ ACS study was a prospective audit
of the care provided to consecutive patients admitted
with suspected ACS during a 2-week period in Australia
and New Zealand in May 2012.15,16 The purpose of the
study was to identify current management and available
treatments, with the aim of better understanding the
ACS environment, in order subsequently to improve ACS
patient care. It provided a unique opportunity to
examine the delivery of care between the two countries.
Methods
Study group
The binational SNAPSHOT ACS study was a prospective audit of the care provided to consecutive patients
Australia and New Zealand SNAPSHOT ACS 2012 Steering Committee
David Brieger (NSW) (Co-chair), John French (NSW) (Co-chair), Derek Chew (SA) (Co-chair), Chris Ellis (NZ) (Co-Chair), Gerry
Devlin (NZ) (Co-Chair), Chris Hammett (QLD), Bernadette Aliprandi-Costa (NSW), Isuru Ranasinghe (NSW), Bridie Carr (NSW), Julie
Redfern (NSW), Fiona Turnbull (NSW), Carolyn Astley (SA), Tom Briffa (WA), Jamie Rankin (WA), Stephen Bloomer (WA), Ahmad
Farshid (ACT), Jeffrey Lefkovitz (VIC).
Project Managers – State-based Cardiac Clinical Networks
New Zealand – Greg Gamble, NSW/ACT – Bernadette Aliprandi-Costa, Bridie Carr, Karen Lintern, Queensland – Tegwen Howell,
Cindy Hall, Susanne Spencer, Dayna Williamson, Victoria – Hella Parker, Julie Plunkett, Wendy Wallace-Mitchell, South Australia,
TAS, NT Rosanna Tavella, Carolyn Astley, Western Australia – Samantha Thompson.
Data Management and Analysis – The George Institute for Global Health and the South Australian Health and Medical
Research Institute (SAHMRI)
Fiona Turnbull, Isuru Ranasinghe, Julie Redfern, Karice Hyun, Matthew Horsfall, Helen Hughes.
New Zealand SNAPSHOT ACS 2012 Steering Committee
Chris Ellis (Chair), Greg Gamble, Harvey White (Auckland), Gerald Devlin (Hamilton), Philip Matsis (Wellington), Andrew Hamer
(Nelson), John Elliott, Richard Troughton (Christchurch), Michael Williams (Dunedin).
Australia and New Zealand SNAPSHOT ACS 2012 Support
Funding: The SNAPSHOT ACS study was supported in part by: The Cardiac Society of Australia and New Zealand, The Heart Foundation
of Australia, The Agency for Clinical Innovation Cardiac Network (NSW), the Victorian Cardiac Clinical Network, the Queensland
Cardiac Clinical Network, the Western Australian Cardiovascular Health Network and the South Australian Cardiac Clinical Network.
Conflict of interest: Derek P. Chew: lecture fees AstraZeneca Australia; educational programme: Heart.org, John French: Advisory
Board Membership Sanofi Aventis Australia, AstraZeneca Australia, Eli Lilly Australia and Boehringer Ingelheim; Grant in Aid; the
Medicines Company. Tom Briffa: Grant In Aid WA Department of Health; travel support WA Department of Health. Chris Hammett:
Consultancy Bayer Australia and Eli Lilly Australia; lecture fees Boehringer Ingelheim and Eli Lilly Australia; travel assistance
AstraZeneca Australia, Bayer Australia, Boehringer Ingelheim Australia and Eli Lilly Australia, Schering Plough Australia, Abbott
Medical Australia. Greg Gamble: Grant Auckland Greenlane Fund. Tegwen Howell: travel assistance Heart Foundation Australia.
Stephen Bloomer: Grant WA Department of Health. David Brieger: Advisory Board AstraZeneca Australia, Boehringer Ingelheim
Australia, Bayer Australia, Pfizer, BMS Australia; Grants: AstraZeneca Australia, Sanofi Aventis Australia, Merck Schering Plough
Australia, Boehringer Ingelheim Australia; lecture fees AstraZeneca Australia, Bayer Australia; travel assistance Bayer Australia,
Boehringer Ingelheim Australia.
© 2015 Royal Australasian College of Physicians
498
Australia versus NZ ACS SNAPSHOT audit
admitted to an Australian or New Zealand hospital with
suspected ACS over a 2-week period: 00.00 h on Monday
14 May to 24.00 h on Sunday 27 May 2012. The study
methods are outlined in the initial publications.15,16
In summary, the study was designed and run by academic clinicians and researchers from both countries,
with widespread support from the Cardiac Society of
Australia and New Zealand, and from many associated
state and national groups, including cardiac clinical networks.15,16 The binational steering committee, project and
data management teams consisted of 31 representatives
(see Acknowledgements).
A 2-week audit period was accepted as a compromise
between the need to collect sufficient patient numbers to
obtain an accurate representative cohort versus the
ability of unfunded clinicians and nurses to collect consecutive patient data.
Data collection
Written study protocols were supplied to all participating
sites, along with definitions of the various data being
collected.
The data collection form recorded patient demographics, initial and discharge diagnosis, medication
use in hospital and at discharge, as well as investigations undertaken, invasive treatments received and
major adverse cardiovascular events (MACE) experienced by patients. Clinical variables enabled the calculation of the Global Registry Acute Coronary Events
(GRACE) risk score.17 Ethnicity was self-reported at
hospital admission.
changes consistent with ischaemia. ‘UAP’ and ‘chest-pain
unlikely ischaemic’ reflected local clinical determination.
Where the diagnosis remained uncertain in the absence
of definitive ECG changes and/or biomarker elevation,
but where the patient received in-hospital coronary
revascularisation (either PCI or CABG), the classification
of ‘chest-pain likely ischaemic’ was applied. Where a
clear alternative primary diagnosis emerged, or when
evidence of myonecrosis was considered secondary to
another disease process (e.g. pulmonary embolus, sepsis),
patients were grouped as ‘other diagnosis including secondary myonecrosis’.
Patient events and outcome
In-hospital MACE, previously defined,15,16 included the
occurrence of any one of the following events: death,
new or recurrent myocardial infarction (MI), MI following PCI or CABG, major bleeding, stroke, cardiac arrest or
worsening heart failure. Clinical event reporting relied on
local documentation using a standardised completion
note. Formal adjudication of events was not possible.
Ethical approval
Ethical approval (with opt-out consent) was obtained for
Australian sites, except for two hospitals in one state
where informed consent applied. In New Zealand’s case,
following review by the national multicentre ethics committee, the study was deemed to be an audit of clinical
management, and a consent waiver was given for all
participating sites.15,16
Patient eligibility and diagnosis
The inclusion criterion for the audit was ‘a patient admitted overnight with a suspected or confirmed ACS’.
Patients were tracked for the duration of the acute care
episode including all transfers between hospitals.
Following admission and investigations, a ‘discharge
diagnosis’ was subsequently determined by the local clinical team who confirmed the diagnosis of an ACS as an ST
– segment elevation myocardial infarction (STEMI), nonSTEMI (NSTEMI) or unstable angina pectoris (UAP) or
determined a ‘non-ACS’ presentation resultant on investigations undertaken in hospital and the patient’s clinical
course.
‘STEMI/LBBB’ required ST elevation or new onset left
bundle branch block (LBBB) on an electrocardiogram
(ECG) at any time during the admission, with elevation
of cardiac biomarkers except where patients had died
prior to biomarkers being taken. ‘NSTEMI’ required evidence of biomarker elevation with or without ECG
Statistical analysis
Data are presented as mean (standard deviation) or
median (interquartile range) as indicated. Comparisons
between groups for categorical variables were made using
Fisher’s exact test, or the Chi-squared test as appropriate.
Between groups comparison of non-normally distributed
data was made using Wilcoxon/Kruskall–Wallis test.
Confidence intervals for rates were calculated using a
mid-P method18 (www.openepi.com, cited 2014 Sept 10).
All tests were two tailed, P < 0.05 was considered
significant and since all comparisons had been prespecified no adjustment to the overall significance level
was made. All analyses were performed using SAS (v9.4,
SAS Institute Inc., Cary, NC, USA). A deliberate response
to some late data clarification from smaller sites led to
minor changes to the baseline data; hence, some data
have slightly changed from earlier publications.15,16,19
© 2015 Royal Australasian College of Physicians
499
Ellis et al.
Results
Patient demographics
Over the 2-week period of 14 to 27 May 2012, 4387
patients were admitted with a suspected or confirmed
ACS to an Australian (3381 (77%)) or a New Zealand
(1006 (23%)) hospital (Fig. 1). Australian patients were
2 years younger; there were mild differences in ethnicity
(P < 0.0001) and minor clinical differences (Table 1).
Of the 2356 patients with a final diagnosis of a locally
confirmed ACS, Australian patients were again younger
at 69 vs 72 years (P = 0.0004), and again had mild ethnic
differences (P < 0.0001) and modest clinical differences
(Table 2). Australian patients were at a lower cardiovascular risk, as assessed by the GRACE hospital admission
score,17 with a lower median GRACE score (147 vs 154,
P = 0.0008).
Investigations, revascularisations and
adverse events
For the entire cohort (n = 4387), Australian patients were
less likely to report the receipt of a chest X-ray (80% vs
91%, P < 0.0001), or a standard exercise treadmill test
(8.8% vs 23%, P < 0.0001), but slightly more likely to
receive a stress echocardiogram (2.7% vs 1.4%, P =
0.014), stress nuclear study (4.4% vs 0.1%, P < 0.0001)
or an invasive cardiac angiogram (38% vs 33%, P =
0.0012) (Table 3). Australian patients had a longer hospital length of stay (2.6 vs 2.2 days, P = 0.019).
For patients subsequently determined to have a
confirmed ACS (n = 2356), Australian patients were still
less likely to report the receipt of a chest x-ray (83% vs
92%, P < 0.0001) or a standard exercise treadmill test
AUSTRALIA
(n=3,381)
NT
(n=114)
WA
(n=352)
*Includes Tasmania
SA
(n=360)
QLD
(n=695)
NSW
(n=1138)
*Includes ACT
Vic
(n=722)
NEW ZEALAND
(n=1006)
(4.4% vs 17%, P < 0.0001) (Table 4). However, there
was no difference in the number receiving an invasive
cardiac angiogram (58% vs 54%, P = 0.082), or
revascularisation with PCI (32% vs 31%, P = 0.92) or
CABG surgery (7.0% vs 5.6%, P = 0.32). Further, the
hospital length of stay did not differ between the two
countries.
STEMI/LBBB patients
There were 419 STEMI/LBBB patients admitted over 2
weeks (Table 5). There was no statistically significant difference between Australia and New Zealand with regards
to standard patient demographics; however, Australian
patients had fewer exercise treadmill tests (0.6% vs
4.1%, P = 0.029) (Table 5).
Reperfusion management and time to
treatment of STEMI/LBBB patients
Primary PCI in 39% of STEMI/LBBB patients (Australian
37% vs New Zealand 43%, P = 0.34) was the most
common form of reperfusion therapy (Table 6). There
was no difference in the door to device time (DTDT) with
an overall median DTDT of 82 (53, 138) min. Further,
59% patients had a DTDT of ≤90 min (Australia 57% vs
New Zealand 67%, P = 0.28), the current target time.1–7
Some 29% of patients had a DTDT of >120 min (Australia
32% vs New Zealand 21%, P = 0.24).
Fibrinolytic therapy was received by 25% of STEMI/
LBBB patients (Australian 23% vs New Zealand 32%,
P = 0.42). There was no difference in the overall median
door to needle time (DTNT) of 42 (interquartile range 25,
70) min. Further, 34% patients had a DTNT of ≤30 min
(Australia 36% vs New Zealand 31%, P = 0.81) (Table 6),
the current target time.1–7
Australian patients were more likely to receive neither
fibrinolysis nor primary PCI as a reperfusion strategy
(40% vs 27%, P = 0.017). In addition, although the use of
glycoprotein 2b/3a inhibitor medication, bivalirudin and
unfractionated heparin were similar, Australian patients
were less likely to receive a low molecular weight
heparin (42% vs 56%, P = 0.020, Table 5).
In both countries, in-hospital death was seen in 7% of
patients, with 29% of patients experiencing a MACE
(death, new or recurrent MI, major bleeding, stroke,
cardiac arrest or worsening heart failure).
NSTEMI/UAP patients
Figure 1 Number of suspected ACS patients registered (n = 4387) in
Australia (n = 3381) and New Zealand (n = 1006).
Of the total 1937 NSTEMI/UAP patients, Australian
patients were younger (70 vs 73 years, P = 0.0006) and
more likely to be male (64% vs 59%, P = 0.048); there
© 2015 Royal Australasian College of Physicians
500
Australia versus NZ ACS SNAPSHOT audit
Table 1 Baseline demographic data of all of patients admitted with suspected or confirmed ACS (n = 4387)
Number of eligible hospitals
Participating hospitals (% eligible)
Age median (range) (years)
Sex (male)
Family history
Ethnicity
Caucasian
Maori
Aboriginal
Pacific Island
Torres Strait Islander
Asian
Indian
Other
Tobacco smoking
Current
Past
Never
Clinical factors
Hypertension
Diabetes mellitus
Dyslipidaemia
Atrial fibrillation
Renal impairment
On dialysis
Morbid obesity
Mechanical heart valve
Chronic liver disease
Chronic lung disease
Active cancer limiting life
Dementia/cognitive impairment
Significant impairment mobility/dependent for ADL
Prior vascular disease
Prior myocardial infarction
Prior PCI
Prior CABG
Prior TIA/Stroke
Prior PAD
Grace risk score (Granger)
GRACE risk score (med/IQR)
GRACE score ≥ 140
Killip class (I or II or III/IV) %
Australia
n = 3381 (77%)
New Zealand
n = 1006 (23%)
Total
n = 4387
525
478 (91%)
67 (18,99)
2035 (60%)
1172 (35%)
P-value
39
39 (100%)
69 (20,97)
585 (58%)
257 (26%)
564
517 (92%)
67 (18,99)
2620 (60%)
1429 (33%)
0.0044
0.26
<0.0001
2936 (87%)
10 (0.3%)
90 (2.7%)
16 (0.5%)
4 (0.1%)
45 (1.3%)
42 (1.2%)
238 (7.0%)
779 (77%)
86 (8.5%)
0
46 (4.6%)
0
35 (3.5%)
43 (4.3%)
17 (1.7%)
3715 (85%)
96 (2.2%)
90 (2.7%)
62 (1.4%)
4 (0.1%)
80 (1.8%)
85 (1.9%)
255 (5.8%)
646 (19%)
1197 (35%)
1538 (46%)
153 (15%)
432 (43%)
422 (42%)
799 (18%)
1629 (37%)
1956 (45%)
0.0058
<0.0001
0.046
2157 (64%)
879 (26%)
1844 (55%)
494 (15%)
335 (10%)
24
282 (8.3%)
70 (2.1%)
65 (1.9%)
452 (13.3%)
87 (2.6%)
114 (3.4%)
189 (5.6%)
620 (62%)
234 (23%)
539 (54%)
170 (17%)
167 (17%)
9
67 (6.7%)
12 (1.2%)
5 (0.5%)
121 (12%)
19 (1.9%)
34 (3.4%)
56 (5.6%)
2777 (63%)
1113 (25%)
2383 (54%)
664 (15%)
502 (11%)
33
349 (8.0%)
82 (1.9%)
70 (1.6%)
573 (13%)
106 (2.4%)
148 (3.4%)
245 (5.6%)
0.22
0.083
0.61
0.080
<0.0001
899 (27%)
686 (20%)
363 (11%)
326 (10%)
189 (5.6%)
292 (29%)
203 (20%)
103 (10%)
127 (13%)
78 (7.8%)
1191 (27%)
889 (20%)
466 (11%)
453 (10%)
267 (6.1%)
0.15
0.96
0.68
0.99
0.013
141 (115, 168)
504 (51%)
84%/13%/3%
139 (115, 165)
2085 (49%)
86%/11%3%
0.15
0.12
0.079
139 (115, 165)
1577 (48%)
87%/10%/3%
<0.0001
0.085
0.084
0.0008
0.29
0.13
0.99
0.99
ADL, activities of daily living; ACS, acute coronary syndrome; CABG, coronary artery bypass graft; GRACE, Global Registry Acute Coronary Events; IQR,
interquartile range; PAD, peripheral artery disease; PCI, percutaneous coronary intervention: TIA, transient ischaemic attack.
were minor clinical differences (Table 7). Australian
patients had a lower GRACE score (143 vs 151, P =
0.0014).
Anticoagulation management, GRACE scores
and time to treatment of NSTEMI/UAP patients
Overall, of the 1937 NSTEMI/UAP patients, 982 (51%)
received an invasive coronary angiogram (Table 8). Aus-
tralian patients had higher rates of coronary angiography
than New Zealand patients (52% vs 46%, P = 0.0011),
and Australian patients had a shorter time to angiography (46 h vs 67 h, P < 0.0001).
More high-risk Australian NSTEMI/UAP patients with
a GRACE score of ≥140 received an angiogram (51% vs
42%, P = 0.013) than equivalent New Zealand patients.
These high-risk Australian patients were also more
likely to receive their angiogram within 24 h (24% vs
© 2015 Royal Australasian College of Physicians
501
Ellis et al.
Table 2 Baseline demographic data of patients admitted with confirmed ACS (n = 2356)
% ACS admissions/All
Age median (range) (years)
Sex (male)
Family history
Ethnicity
Caucasian
Maori
Aboriginal
Torres Strait Islander
Pacific Island
Asian
Indian
Other
Tobacco smoking
Current
Past
Never
Clinical factors
Hypertension
Diabetes mellitus
Dyslipidaemia
Atrial fibrillation
Renal Impairment
On dialysis
Morbid obesity
Mechanical heart valve
Chronic liver disease
Chronic lung disease
Active cancer limiting life
Dementia/cognitive impairment
Significant impairment mobility/dependent for ADL
Prior vascular disease
Prior myocardial infarction
Prior PCI
Prior CABG
Prior TIA/Stroke
Prior PAD
Grace risk score (Granger)
GRACE score, median (IQR)
% GRACE score ≥ 140
Killip class (I or II or III/IV) %
Australia
n = 1825 (77%)
New Zealand
n = 531 (23%)
Total
n = 2356
54%
69 (20,99)
1194 (65%)
656 (36%)
P-value
53%
72 (21,97)
328 (62%)
137 (26%)
54%
70 (20,99)
1522 (65%)
793 (34%)
0.0004
0.12
<0.0001
1593 (87%)
5 (0.3%)
47 (2.6%)
2 (0.1%)
6 (0.3%)
21 (1.2%)
23 (1.2%)
128 (7.0%)
430 (81%)
43 (7.9%)
0
0
21 (4.0%)
10 (1.9%)
18 (3.6%)
9 (1.7%)
2024 (86%)
48 (2.0%)
47 (2.0%)
2 (0.1%)
27 (1.1%)
31 (1.3%)
41 (1.7%)
137 (5.8%)
354 (19%)
708 (39%)
763 (42%)
85 (16%)
232 (44%)
214 (41%)
439 (19%)
940 (40%)
977 (42%)
0.11
0.053
0.53
1240 (68%)
540 (30%)
1082 (59%)
234 (13%)
208 (11%)
17
155 (8.5%)
33 (1.8%)
38 (2.1%)
254 (14%)
48 (2.6%)
56 (3.1%)
95 (5.2%)
357 (67%)
144 (27%)
309 (58%)
92 (17%)
108 (20%)
5
29 (5.5%)
5 (0.9%)
2 (0.4%)
68 (13%)
8 (1.5%)
23 (4.3%)
34 (6.4%)
1597 (68%)
684 (29%)
1391 (59%)
328 (14%)
316 (13%)
22
184 (7.8%)
38 (1.6%)
40 (1.7%)
322 (14%)
56 (2.4%)
79 (3.4%)
129 (5.5%)
0.75
0.28
0.65
0.0054
<0.0001
573 (31%)
420 (23%)
222 (12%)
194 (11%)
126 (6.9%)
179 (34%)
118 (22%)
67 (13%)
80 (15%)
54 (10%)
752 (32%)
538 (23%)
289 (12%)
274 (12%)
180 (7.6%)
0.32
0.72
0.76
0.0070
0.016
147 (125, 173)
1035 (58%)
86%/11%/3.0%
154 (132, 181)
346 (66%)
81%/16%/3.5%
149 (126, 175)
1381 (60%)
85%/12%/3.2%
<0.0001
0.022
0.24
0.0040
0.57
0.15
0.17
0.28
0.0008
0.0001
0.053
ADL, activities of daily living; ACS, acute coronary syndrome; CABG, coronary artery bypass graft; GRACE, Global Registry Acute Coronary Events; IQR,
interquartile range; PAD, peripheral artery disease; PCI, percutaneous coronary intervention: TIA, transient ischaemic attack.
17%, P < 0.0001) or within 72 h (74% vs 57%, P =
0.0053) than similar New Zealand patients (Fig. 2). Australian patients were less likely to receive low molecular
weight heparin treatment (50% vs 68%, P < 0.0001)
(Table 7).
Overall 12% of patients experienced a MACE,
with a hospital death rate of 1.8%. There was no statistical difference in these outcomes between the two countries (Table 8).
Discharge medications
Of the 2356 confirmed ACS patients, 49 (2.6%) of 1825
Australian patients and 17 (3.2%) of 531 New Zealand
patients died in hospital, with 2290 patients subsequently
being discharged. Of the 388 surviving patients following
a STEMI/LBBB presentation, the use of aspirin, statins,
beta-blockers, angiotensin converting enzyme inhibitors/
angiotensin receptor blockers (ACE-I/ARB) and other
© 2015 Royal Australasian College of Physicians
502
Australia versus NZ ACS SNAPSHOT audit
Table 3 Investigations, revascularisations and events. (all patients, n = 4387)
Procedure
Chest X-ray
Echocardiogram
Stress echocardiogram
Exercise test
Stress nuclear study
CT coronary angiogram
Conventional angiogram
PCI
CABG
PCI or CABG
In-hospital events
Major bleed
Cardiac arrest
Worsening heart failure
Stroke
MI post-admission
In-hospital death
MACE†
Length of stay, median (IQR) (days)
Australia
n = 3381
2725 (80%)
1006 (30%)
92 (2.7%)
299 (8.8%)
148 (4.4%)
125 (3.7%)
1299 (38%)
582 (17%)
128 (3.7%)
706 (21%)
34 (1.0%)
65 (1.9%)
236 (7.0%)
16 (0.5%)
63 (1.9%)
63 (1.9%)
354 (10%)
2.6 (1.2, 4.8)
New Zealand
n = 1006
910 (91%)
287 (29%)
14 (1.4%)
229 (23%)
1 (0.1%)
39 (3.9%)
330 (33%)
169 (17%)
34 (3.4%)
202 (20%)
6 (0.6%)
18 (1.8%)
60 (6.0%)
5 (0.5%)
22 (2.2%)
20 (2.0%)
98 (9.7%)
2.2 (1.0, 4.6)
Total
n = 4387
P-value
3635 (83%)
1293 (29%)
106 (2.4%)
528 (12%)
149 (3.4%)
164 (3.7%)
1629 (37%)
751 (17%)
162 (3.7%)
908 (21%)
<0.0001
0.48
0.014
<0.0001
<0.0001
0.77
0.0012
0.78
0.63
0.60
40 (0.93)
83 (1.9%)
296 (6.7%)
21 (0.5%)
85 (1.9%)
83 (1.9%)
443 (10%)
2.5 (1.1, 4.8)
0.26
0.90
0.28
0.99
0.52
0.79
0.72
0.019
†MACE includes all cause death, new or recurrent MI, major bleeding, stroke, cardiac arrest, worsening heart failure. CABG, coronary artery bypass graft;
CT, computed tomography; IQR, interquartile range; MACE, major adverse cardiovascular events; PCI, percutaneous coronary intervention
Table 4 Investigations and revascularisations in patients with confirmed ACS (n = 2356): STEMI/LBBB (n = 419), NSTEMI (n = 1012) or UAP (n = 925)
Procedure
Chest X-ray
Echocardiogram
Stress echo
Exercise test
Stress nuclear study
CT coronary angiogram
Conventional angiogram
PCI
CABG
PCI or CABG
In-hospital events
Major bleed
Cardiac arrest
Worsening heart failure
Stroke
MI post-admission
In-hospital death
MACE†
Length of stay, median (IQR) (days)
Australia
n = 1825
1515 (83%)
662 (36%)
35 (1.9%)
81 (4.4%)
64 (3.5%)
67 (3.7%)
1054 (58%)
577 (32%)
127 (7.0%)
700 (38%)
28 (1.5%)
50 (2.7%)
189 (10%)
12 (0.6%)
58 (3.2%)
49 (2.7%)
272 (15%)
3.6 (2.0, 5.8)
New Zealand
n = 531
488 (92%)
210 (40%)
4 (0.8%)
89 (17%)
1 (0.2%)
23 (4.3%)
284 (54%)
166 (31%)
30 (5.6%)
195 (37%)
5 (0.9%)
14 (2.6%)
47 (8.9%)
4 (0.8%)
20 (3.8%)
17 (3.2%)
80 (15%)
3.5 (1.9, 6.0)
Total
n = 2356
P-value
2003 (85%)
872 (37%)
39 (1.7%)
170 (7.2%)
65 (2.8%)
90 (3.8%)
1338 (57%)
743 (32%)
157 (6.7%)
895 (38%)
<0.0001
0.18
0.080
<0.0001
<0.0001
0.52
0.082
0.92
0.32
0.51
33 (1.4%)
64 (2.7%)
236 (10%)
16 (0.7%)
78 (3.3%)
66 (2.8%)
352 (15%)
3.6 (2.0, 5.8)
0.40
0.99
0.33
0.77
0.49
0.55
0.95
0.65
†MACE includes all cause death, new or recurrent MI, major bleeding, stroke, cardiac arrest, worsening heart failure. ACS, acute coronary syndrome;
CABG, coronary artery bypass graft; CT, computed tomography; NSTEMI, non-segment elevation myocardial infarction; MACE, major adverse cardiovascular events; STEMI,/LBBB, segment elevation myocardial infarction/ left bundle branch block; PCI, percutaneous coronary intervention; UAP, unstable
angina pectoris.
© 2015 Royal Australasian College of Physicians
503
Ellis et al.
Table 5 Demographics, investigations and management of ‘STEMI/LBBB‘ patients
Demographics
Age, median (range)
Sex (male %)
Family history
Prior MI
Prior PCI
Prior CABG
Prior TIA/Stroke
GRACE dcore median (IQR)
% GRACE score ≥ 140
Investigations/Management
Chest X-ray
Echocardiogram
Stress echo
Exercise test
CT angiogram
Conventional angiogram
Total PCI
CABG
Anti-coagulation
GP IIb/IIIa inhibitor
Bivalirudin
UF heparin
LMW heparin
STEMI/LBBB
Australia
n = 321 (77%)
STEMI/LBBB
New Zealand
n = 98 (23%)
Total
P-value
n = 419
63 (30, 99)
228 (71%)
102 (32%)
59 (18%)
36 (11%)
18 (5.6%)
16 (5.0%)
170 (145, 199)
250 (80%)
68 (33, 97)
73 (75%)
22 (22%)
15 (15%)
11 (11%)
3 (3.1%)
7 (7.1%)
171 (149, 193)
83 (86%)
0.092
0.61
0.079
0.55
0.99
0.43
0.45
0.79
0.056
64 (30, 99)
301 (72%)
124 (30%)
74 (18%)
47 (11%)
21 (5.0%)
23 (5.5%)
171 (146, 197)
313 (76%)
268 (84%)
194 (60%)
2 (0.6%)
2 (0.6%)
12 (3.7%)
270 (84%)
208 (65%)
28 (8.7%)
87 (89%)
70 (71%)
1 (1%)
4 (4.1%)
4 (4.1%)
86 (88%)
66 (67%)
6 (6.1%)
0.26
0.056
0.55
0.029
0.77
0.42
0.72
0.53
355 (85%)
264 (63%)
3 (0.7%)
6 (1.4%)
16 (3.8%)
356 (85%)
274 (65%)
34 (8.1%)
90 (28%)
7 (2.2%)
219 (68%)
136 (42%)
28 (29%)
3 (3.1%)
73 (75%)
55 (56%)
0.90
0.70
0.26
0.020
118 (28%)
10 (2.4%)
292 (70%)
191 (46%)
CABG, coronary artery bypass graft; CT, computed tomography; GRACE, Global Registry Acute Coronary Events; IQR, interquartile range; LMW, low
molecular weight; MACE, major adverse cardiovascular events; MI, myocardial infarction; STEMI/LBBB, segment elevation myocardial infarction/left
bundle branch block; PCI, percutaneous coronary intervention; TIA, transient ischaemic attack; UAP, unstable angina pectoris; UF, unfractionated.
lipid medication was not statistically different, but fewer
Australian patients received dual anti-platelet therapy at
discharge (83% vs 98%, P < 0.0001) (Table 9).
Of the 1902 surviving patients after a NSTEMI/UAP
presentation, Australian patients were less likely to
receive aspirin (84% vs 89%, P = 0.0079) or a betablocker (67% vs 77%, P = 0.0002), but more likely to
receive an ACE-I/ARB medicine (65% vs 58%, P =
0.0060) or ‘other’ lipid lowering agent (8.2% vs 5.47%,
P = 0.024). Statin use was similar in Australia (80%) and
New Zealand (83%) (P = 0.21) (Table 9).
Rehabilitation/scheduled
outpatient investigations
Referrals for cardiac rehabilitation were low, with less
than half of discharged confirmed ACS patients having a
recorded referral for any of the six indicators assessed
(Table 10). Australian patients were less likely to be
referred as an in-patient in four of the five services,
whereas they were more likely to be referred to an outpatient service (Table 10).
Few patients were scheduled for outpatient investigations after hospital discharge, and there was no statistical
difference in these numbers of patients between the two
countries.
Discussion
The SNAPSHOT ACS study has enabled a comparison of
4387 Australian and New Zealand patients admitted to
hospital with a suspected or confirmed ACS.
Overall, the patient demographic differences between
the two countries were quite small, although the ethnic
mix gives an interesting insight into some minor population differences. Nonetheless, the similar ACS population does allow a reasonably valid comparison of ACS
management across the two countries.
Patients admitted with a suspected ACS require investigations to help to make an accurate medical diagnosis.
However, we found that less than half of patients received
an anatomical assessment of left ventricular function with
an echocardiogram, or of the coronary arteries with a
computed tomography coronary angiogram or an invasive
© 2015 Royal Australasian College of Physicians
504
Australia versus NZ ACS SNAPSHOT audit
Table 6 Reperfusion management and time to treatment of ‘STEMI/LBBB‘ patients
Reperfusion
Fibrinolysis†
Primary PCI
Number with rescue PCI (n)
Neither fibrinolysis nor Primary PCI
Reperfusion time frames
Number with pre-hospital fibrinolysis (n)
Number with in-hospital fibrinolysis (n)
Median DTNT (IQR) (min)‡
% DTNT ≤ 30 min
Primary PCI time frames
Number with primary PCI (PCI)
DTDT (All in-hospital PPCI) median (IQR) (min)
DTDT % ≤ 90 min
DTDT % ≤ 120 min
Start of angiogram to DT median (IQR) (min)
Angiogram time frames
Points with routine angiogram (excluding PPCI/Rescue PCI)
Time to routine angiogram (h)
Time to routine angiogram ≤ 24 h
In-hospital events
Major bleed
Cardiac arrest
Worsening heart failure
CVA
MI post-admission
In-hospital death
MACE§
Length of stay (median IQR)
STEMI/LBBB
Australia
n = 321 (77%)
STEMI/LBBB
New Zealand
n = 98 (23%)
P-value
n = 419
74 (23%)
120 (37%)
19 (6%)
129 (40%)
31 (32%)
42 (43%)
6 (6%)
26 (27%)
0.42
0.34
0.99
0.017
105 (25%)
162 (39%)
25 (6%)
155 (37%)
3 (3.1%)
28 (29%)
48 (28, 84)
9 (31%)
0.99
0.096
0.32
0.81
10 (3.1%)
64 (20%)
42 (25, 68)
22 (36%)
120 (37%)
82 (52, 143)
68/120 (57%)
82/120 (68%)
20 (14, 29)
42 (43)
82 (56, 93)
28/42 (67%)
33/42 (79%)
22 (13, 27)
Total
0.42
0.81
0.28
0.24
0.78
13 (3.1%)
92 (22%)
42 (25, 70)
31 (34%)
162 (39%)
82 (53, 138)
96/162 (59%)
115/162 (71%)
20 (14, 28)
65 (20%)
23 (14, 52)
34 (52%)
18 (18%)
36 (21, 67)
5 (28%)
0.99
0.098
0.11
83 (20%)
28 (15, 53)
38 (46%)
9 (2.8%)
25 (7.7%)
61 (19%)
7 (2.2%)
17 (5.3%)
24 (7.5%)
93 (29%)
4.4 (3.0, 6.6)
1 (1%)
9 (9%)
16 (16%)
1 (1%)
5 (5%)
7 (7%)
29 (30%)
3.7 (2.9, 6.1)
0.46
0.67
0.66
0.69
0.99
0.99
0.90
0.081
10 (2.4%)
34 (8.1%)
77 (18%)
8 (1.9%)
22 (5.3%)
31 (7.4%)
122 (29%)
4.1 (2.9, 6.6)
†Includes one NZ patient and two Australian treated with pharmaco-invasive intervention (fibrinolysis + PPCI). ‡Excludes pre-hospital fibrinolysis. §MACE
includes all cause death, new or recurrent MI, major bleeding, stroke, cardiac arrest, worsening heart failure. CVA, cerebrovascular accident; DTNT, door
to needle time; DTDT, door to device time; DT, device time; IQR, interquartile range; MACE, major adverse cardiovascular events; NZ, New Zealand; PCI,
percutaneous coronary intervention; PPCI, primary percutaneous coronary intervention. STEMI/LBBB, segment elevation myocardial infarction/left
bundle branch block.
cardiac angiogram. Further, less than one fifth received a
functional cardiac assessment with a standard treadmill
test, stress echocardiogram or a stress nuclear study. The
‘optimal’ level for these tests is unclear, but the numbers
do suggest a limited availability or underutilisation of
diagnostic investigations. This was seen across Australia
and New Zealand.
For patients with a confirmed ACS, optimal management is already defined in guidelines.1–7 Overall, a low
level of cardiac investigations was available to patients
admitted with an ACS. Many ACS patients are not
assessed with an echocardiogram or an invasive cardiac
angiogram. Even if some patients are particularly frail, and
it would be inappropriate for them to undergo an invasive
cardiac angiogram, just under half of the patients who did
not receive this test would not fit into this category.
Further, few patients would be so frail that a non-invasive
echocardiogram would not help to guide good management, but it was performed in only one third of the
confirmed ACS patients. There was again a similarly low
level of investigations seen in both countries.
It might have been thought that increased spending on
health would result in increased access to these investigations. The 2011 Organisation for Economic Cooperation and Development reported that Australia has about
twice the gross domestic product of New Zealand (US$
68 099.60 vs US$ 38 587.90 per capita, respectively) and
spends about 8.6% of gross domestic product (GDP) on
health compared with 10% of GDP spent on health in
New Zealand.20 Total health expenditure per capita (USD
purchasing power parity) was reported to be $3800 in
Australia and $3182 in New Zealand. However, despite
© 2015 Royal Australasian College of Physicians
505
Ellis et al.
Table 7 Demographics, investigations and management of ‘NSTEMI/UAP‘ patients
Demographics
Age, median (IQR) (years)
Sex (male %)
Family history
Prior MI
Prior PCI
Prior CABG
Prior TIA/Stroke
Grace score (median IQR)
% GRACE score ≥ 140
Investigations/Management
Chest X-ray
Echocardiogram
Stress echo
Stress nuclear study
Exercise test
CT angiogram
Conventional angiogram
Total PCI
CABG
Anti-Coagulation
Fibrinolysis
UF heparin
LMW heparin
GP IIb/IIIa inhibitor
Bivalirudin
NSTEMI/UAP
Australia
n = 1504 (78%)
NSTEMI/UAP
New Zealand
n = 433 (22%)
P-value
Total
NSTEMI/UAP
n = 1937
70 (60, 79)
966 (64%)
554 (37%)
514 (34%)
384 (26%)
204 (14%)
178 (12%)
143 (122, 166)
785 (53%)
73 (62, 82)
255 (59%)
115 (27%)
164 (38%)
107 (25%)
64 (15%)
73 (17%)
151 (127, 175)
263 (61%)
0.0006
0.048
<0.0001
0.17
0.75
0.53
0.0073
0.0014
0.0024
71 (61, 80)
1221 (63%)
669 (35%)
678 (35%)
491 (25%)
268 (14%)
251 (13%)
145 (123, 168)
1482 (78%)
401 (93%)
140 (32%)
3 (0.7%)
1 (0.2%)
85 (20%)
19 (4.4%)
198 (46%)
100 (23%)
24 (5.5%)
<0.0001
0.64
0.043
<0.0001
<0.0001
0.48
0.019
0.57
0.50
1648 (85%)
608 (31%)
36 (1.9%)
63 (3.2%)
164 (8.5%)
74 (3.8%)
982 (51%)
469 (24%)
123 (6.4%)
0
131 (30%)
293 (68%)
11 (2.5%)
3 (0.7%)
0.99
0.95
<0.0001
0.15
0.45
2 (0.10%)
583 (30%)
1043 (54%)
73 (3.8%)
23 (1.2%)
1247 (83%)
468 (31%)
33 (2.2%)
62 (4.0%)
79 (5.3%)
55 (3.7%)
784 (52%)
369 (25%)
99 (6.6%)
2 (0.10%)
452 (30%)
750 (50%)
62 (4.1%)
20 (1.3%)
CABG, coronary artery bypass graft; CT, computed tomography; GP, glycoprotein; GRACE, Global Registry Acute Coronary Events; IQR, interquartile
range; MI, myocardial infarction; NSTEMI/UAP, segment elevation myocardial infarction/unstable angina pectoris; PCI, percutaneous coronary intervention; TIA, transient ischaemic attack; UF, unfractionated.
Table 8 Anticoagulation management, GRACE scores and time to treatment of ‘NSTEMI/UAP‘ patients
Angiogram time frames†
Number of conventional angiograms
Time to angiography (median IQR) (h)
GRACE score ≥ 140‡
GRACE score ≥ 140 + angiogram
GRACE score ≥ 140 + angiogram ≤ 24 h
GRACE score < 140 (n)
GRACE score < 140 + angiogram
GRACE score < 140 + angiogram ≤ 72 h
In-hospital events
Major bleed
Cardiac arrest
Worsening heart failure
CVA
MI post-admission
In-hospital death
MACE§
Length of stay (median IQR) (days)
Australia
n = 1504 (78%)
New Zealand
n = 433 (22%)
P-value
Total
n = 1937
784 (52%)
46 (23, 77)
785 (53%)
398 (51%)
96 (24%)
697 (47%)
375 (54%)
278 (74%)
198 (46%)
67 (32, 96)
263 (61%)
110 (42%)
19 (17%)
166 (39%)
87 (52%)
50 (57%)
0.0011
<0.0001
0.0024
0.013
<0.0001
0.0024
0.75
0.0053
982 (51%)
49 (24, 85)
1048 (55%)
508 (48%)
115 (23%)
863 (45%)
462 (54%)
328 (71%)
19 (1.3%)
25 (1.7%)
128 (8.5%)
5 (0.3%)
41 (2.7%)
25 (1.6%)
179 (12%)
3.2 (1.8, 5.5)
4 (0.9%)
5 (1.2%)
31 (7.2%)
3 (0.7%)
15 (3.5%)
10 (2.3%)
51 (12%)
3.5 (1.8, 6.0)
0.80
0.66
0.43
0.39
0.42
0.41
0.99
0.34
23 (1.2%)
30 (1.5%)
159 (8.2%)
8 (0.4%)
56 (2.9)
35 (1.8%)
230 (12%)
3.3 (1.8, 5.7)
†Data from two angiograms are excluded because times unavailable. ‡GRACE score could not be calculated for 28 patients. §MACE includes all cause
death, new or recurrent MI, major bleeding, stroke, cardiac arrest, worsening heart failure. CVA, cerebrovascular accident; GRACE, Global Registry Acute
Coronary Events; IQR, interquartile range; MACE, major adverse cardiovascular events; MI, myocardial infarction.
© 2015 Royal Australasian College of Physicians
506
Australia versus NZ ACS SNAPSHOT audit
Figure 2 Proportion of ‘high risk’ NSTEMI/UAP patients (GRACE score ≥
140) with time to angiogram less than 24 or 72 h. □, New Zealand; ,
Australia.
this difference in healthcare spending, the investigations
received by suspected or confirmed ACS patients were
remarkably similar.
We found that for NSTEMI/UAP patients, there was a
slightly higher rate and more rapid access to cardiac angiography in Australia. This might be a reflection of higher
healthcare spending in Australia, although the differences between the two countries were not great. Further,
there are so many unknown variables, such as the relative costs of angiography, the availability and cost of
medical and non-medical staff, the relative costs of maintaining the healthcare facilities, the relative geography,
local and regional hospital provision, etc., which prevent
any firm conclusion being drawn. However, overall the
data are remarkable for the similarities seen between the
two countries rather than the differences.
We also found some other areas of ACS management
where there seemed to be suboptimal delivery of proven
treatments. Approximately one third of STEMI patients
did not receive reperfusion therapy. This figure is consistent with overseas experience. In a recent study describing
Table 9 Discharge medications of confirmed ACS patients (NZ n = 514: – 17 in hospital deaths, Australia n = 1776: – 49 in hospital deaths)
STEMI/LBBB
Aspirin (1)
Other antiplatelet (2)
Dual antiplatelet (1&2)
Beta blocker
ACE-I/ARB
Statin
Other lipid lowering
Australia
n = 297
New Zealand
n = 91
284 (96%)
245 (83%)
242 (82%)
242 (82%)
221 (74%)
270 (91%)
14 (5%)
90 (99%)
89 (98%)
90 (98%)
75 (82%)
74 (81%)
86 (95%)
1 (1%)
NSTEMI/UAP
P-value
Total
(n = 388)
Australia
n = 1429
New Zealand
n = 423
P-value
Total
n = 1902
0.20
<0.0001
<0.0001
0.99
0.21
0.38
0.13
374 (96%)
334 (86%)
331 (85%)
317 (82%)
295 (76%)
356 (92%)
15 (3.8%)
1246 (84%)
846 (57%)
768 (52%)
991 (67%)
962 (65%)
1189 (80%)
121 (8.2%)
378 (89%)
265 (63%)
260 (62%)
324 (77%)
244 (58%)
352 (83%)
23 (5.4%)
0.0079
0.050
0.0006
0.0002
0.006
0.21
0.024
1642 (85%)
1111 (58%)
1028 (54%)
1315 (69%)
1206 (63%)
1541 (81%)
144 (7.6%)
ACE-I/ARB, angiotensin converting enzyme inhibitors/angiotensin receptor blocker; ACS, acute coronary syndrome; NSTEMI/UAP, non-segment elevation
myocardial infarction/unstable angina pectoris; STEMI/LBBB, segment elevation myocardial infarction/ left bundle branch block.
Table 10 In-hospital cardiac rehabilitation/scheduled outpatient investigations in those discharged alive
In-hospital rehabilitation/screening
In-hospital cardiac rehabilitation
Smoking cessation advice/intervention
Screening for depression
Dietary modification advice
Physical activity advice
Referral to OP cardiac rehabilitation
Scheduled outpatient investigations
Coronary angiogram
PCI
CABG
Function test
Echocardiogram
Australia
n = 1776
New Zealand
n = 514
P-value
Total
n = 2290
703 (40%)
355 (20%)
163 (9.2%)
668 (37%)
775 (44%)
853 (48%)
258 (50%)
143 (28%)
65 (13%)
161 (31%)
212 (41%)
210 (41%)
<0.0001
0.0003
0.024
0.0092
0.34
0.0042
961 (42%)
499 (22%)
228 (10%)
829 (36%)
987 (43%)
1063 (46%)
101 (5.7%)
53 (3.0%)
42 (2.4%)
199 (11%)
145 (8.2%)
18 (3.5%)
9 (1.8%)
11 (2.1%)
49 (9.5%)
43 (8.4%)
0.055
0.16
0.53
0.30
0.86
119 (5.2%)
62 (2.7%)
53 (2.3%)
248 (11%)
188 (8.2%)
CABG, coronary artery bypass graft; OP, outpatient; PCI, percutaneous coronary intervention.
© 2015 Royal Australasian College of Physicians
507
Ellis et al.
the current situation in 30 European countries, no
reperfusion therapy was given to 7% to 63% of
patients.21 Why this occurs is unknown, and further
assessment as to how an increase in reperfusion in Australia and New Zealand can be achieved is certainly
required. Improvements in ACS patient care have been
reported22 with attention to detail in several clinical areas
resulting in improved patient outcomes.23
We also found that many areas of secondary prevention management seemed to be suboptimal. Up to half of
ACS events occur in individuals who have had a prior
hospital admission for coronary heart disease,24 emphasising the need for widespread use of secondary prevention medication as well as comprehensive cardiac
rehabilitation/secondary prevention programmes. There
are certainly some patients who are unsuitable candidates for individual medications, but the numbers of
patients not being discharged on proven secondary medication is of concern. Once more, although there was a
minor difference in uptake of these services between
Australia and New Zealand, the major issue is clearly one
of a missed opportunity to better deliver these proven
strategies to many patients in each country19).
The SNAPSHOT ACS Study has been an extensive programme designed to understand current ACS patient
management in Australia and New Zealand. Further in
depth assessment of the structure of the clinical services,
which are central to the delivery of healthcare to individual patients, is certainly warranted to try to find solutions to the limitations seen across the two countries.
These challenges of service delivery are common to all
References
1 Chew DP, Aroney CN, Aylward PE,
Kelly AM, White HD, Tideman PA et al.
2011 Addendum to the National Heart
Foundation of Australia/Cardiac Society
of Australia and New Zealand Guidelines
for the management of acute coronary
syndromes (ACS) 2006. Heart Lung Circ
2011; 20: 487–502.
2 ST-Elevation Myocardial Infarction
Guidelines Group and the New Zealand
Branch of the Cardiac Society of
Australia and New Zealand. ST-elevation
myocardial infarction: New Zealand
management guidelines, 2013.
N Z Med J 2013; 126: 127–64.
3 Non ST-Elevation Acute Coronary
Syndrome Guidelines Group and the
New Zealand Branch of the Cardiac
Society of Australia and New Zealand.
New Zealand 2012 guidelines for the
contemporary ACS management services, and warrant
the ongoing development of therapeutic advances, clinical trials, practice guidelines and performance and
outcome measures, termed the ‘cycle of quality’.25 The
advent of local clinical networks is likely to facilitate
greatly improvements in ACS care in many areas.26,27
There are several limitations to our study, including the
fact that although the majority of ACS patients were
identified, we could not guarantee consecutive recruitment over the 2-week period. The scope of this paper was
to report binational comparison data of acute in-hospital
management and outcomes, thus we have not included
follow-up data nor have we reported differences in ACS
management between city and more geographically isolated, rural populations. Neither has the scope of this
paper included a review of comparative cost structures
across Australia and New Zealand, although all of these
areas may importantly advance knowledge.
Conclusion
Overall the management of suspected or confirmed ACS
patients showed more similarities than differences
between Australia and New Zealand. However, some
limitation to important investigations and treatments
was seen in both countries. It is likely that most patient
benefit will come from a coordinated, ongoing assessment of the available clinical services within each
country, with timely feedback to focus improvements in
the management of ACS patients in both countries.
management of non ST-elevation acute
coronary syndromes. N Z Med J 2012;
125: 122–47.
4 Steg PG, James SK, Atar D, Badano LP,
Blömstrom-Lundqvist C, Borger MA
et al. ESC Guidelines for the
management of acute myocardial
infarction in patients presenting
with ST-segment elevation: the task
force on the management of
ST-segment elevation acute myocardial
infarction of the European Society of
Cardiology (ESC). Eur Heart J 2012; 33:
2569–619.
5 Hamm CW, Bassand J-P, Agewall S, Bax
J, Boersma E, Bueno H et al. ESC
Guidelines for the management of acute
coronary syndromes in patients
presenting without persistent
ST-segment elevation: the task force for
the management of acute coronary
syndromes (ACS) in patients presenting
without persistent ST-segment elevation
of the European Society of Cardiology
(ESC). Eur Heart J 2011; 32: 2999–3054.
6 O’Gara PT, Kushner FG, Ascheim DD,
Casey DE Jr, Chung MK, de Lemos JA
et al. 2013 ACCF/AHA guideline for the
management of ST-elevation myocardial
infarction: a report of the American
College of Cardiology Foundation/
American Heart Association Task Force
on Practice Guidelines. J Am Coll Cardiol
2013; 61: e78–140.
7 Anderson JL, Adams CD, Antman EM,
Bridges CR, Califf RM, Casey DE Jr et al.
2012 ACCF/AHA focused update
incorporated into the ACCF/AHA 2007
guidelines for the management of
patients with unstable angina/nonST-elevation myocardial infarction: a
report of the American College of
Cardiology Foundation/American Heart
Association Task Force on Practice
© 2015 Royal Australasian College of Physicians
508
Australia versus NZ ACS SNAPSHOT audit
8
9
10
11
12
13
14
15
Guidelines. J Am Coll Cardiol 2013; 61:
e179–347.
Astley CM, MacDougall CJ, Davidson
PM, Chew DP. Lost in translation:
health resource variability in the
achievement of optimal performance
and clinical outcome. Circ Cardiovasc Qual
Outcomes 2011; 4: 512–20.
Ellis CJ, Hamer AW. Cardiovascular
Health in New Zealand: areas of concern
and targets for improvement in 2008
and beyond. N Z Med J 2008; 121: 5–10.
White HD. Systems of care: need for
hub and spoke systems of care for
patients with myocardial infarction.
A call for action. N Z Med J 2010; 123:
7–10.
White HD, Chew DP. Acute myocardial
infarction. Lancet 2008; 372: 570–84.
Iglehart JK. Prioritizing
comparative-effectiveness research-IOM
recommendations. N Engl J Med 2009;
361: 325–8.
Chung SC, Gedeborg R, Owen N, James
S, Jeppsson A, Wolfe C et al. Acute
myocardial infarction: a comparison of
short-term survival in national outcome
registries in Sweden and the UK. Lancet
2014; 383: 1305–12.
Levy AR, Mitton C, Johnstone KM,
Harrigan B, Briggs AH. International
comparison of comparative effectiveness
research in five jurisdictions: insight for
the US. Pharmacoeconomics 2010; 28:
813–30.
Chew DP, French J, Briffa TG, Hammett
CJ, Ellis CJ, Ranasinghe I et al. Acute
coronary syndrome care across Australia
and New Zealand: the SNAPSHOT ACS
study. MJA 2013; 199: 1–7.
16 Ellis C, Gamble G, Devlin G, Elliott J,
Hamer A, Williams M et al. The
management of acute coronary
syndrome patients across New Zealand
in 2012: results of a third
comprehensive nationwide audit and
observations of current care. N Z Med J
2013; 126: 36–68.
17 Granger CB, Goldberg RJ, Dabbous O,
Pieper KS, Eagle KA, Cannon CP et al.
Predictors of hospital mortality in the
Global Registry of Acute Coronary
Events. Arch Intern Med 2003; 163:
2345–53.
18 Rothman KJ, Boice JD Jr. Epidemiologic
Analysis with a Programmable Calculator
(NIH Publication 79-1649). Washington,
DC: US Government Printing Office;
1979.
19 Redfern J, Hyun K, Chew DP, Astley C,
Chow C, Aliprandi-Costa B et al.
Prescription of secondary prevention
medications, lifestyle advice, and referral
to rehabilitation among acute coronary
syndrome inpatients: results from a large
prospective audit in Australia and New
Zealand. Heart 100: 1281–8.
20 OECD. Health at a Glance 2013: OECD
Indicators. France: OECD Publishing;
2013.
21 Widimsky P, Wijns W, Fajadet J, de
Belder M, Knot J, Aaberge L et al.
Reperfusion therapy for ST elevation
acute myocardial infarction in Europe:
description of the current situation in
30 countries. Eur Heart J 2010; 31:
943–57.
22 Gibson CM, Pride YB, Frederick PD,
Pollack CV Jr, Canto JG, Tiefenbrunn AJ
23
24
25
26
27
et al. Trends in reperfusion strategies,
door-to-needle and door-to-balloon
times, and in-hospital mortality among
patients with ST-segment elevation
myocardial infarction enrolled in the
National Registry of Myocardial
Infarction from 1990 to 2006. Am Heart
J 2008; 156: 1035–44.
Peterson ED, Shah BR, Parsons L,
Pollack CV Jr, French WJ, Canto JG
et al. Trends in quality of care for
patients with acute myocardial
infarction in the National Registry of
Myocardial Infarction from 1990 to
2006. Am Heart J 2008; 156: 1045–55.
Briffa T, Hobbs M, Tonkin A, Sanfilippo
FM, Hickling S, Ridout SC et al.
Population trends of current coronary
heart disease event rates remain high.
Circ Cardiovasc Qual Outcomes 2011; 4:
107–13.
Califf RM. The benefits of moving
quality to a national level. Am Heart J
2008; 156: 1019–22.
Kerr AJ, Lin A, Lee M, Ternouth I,
Killion B, Devlin G. Risk stratification
and timing of coronary angiography in
acute coronary syndromes: are we
targeting the right patients in a timely
manner? (ANZACS-QI 1). N Z Med J
2013; 126: 69–80.
Tideman PA, Tirimacco R, Senior DP,
Setchell JJ, Huynh LT, Tavella R et al.
Impact of a regionalised clinical cardiac
support network on mortality among
rural patients with myocardial
infarction. MJA 2014; 200: 157–60.
Supporting Information
Additional Supporting Information may be found in the online version of this article at the publisher’s web-site:
Appendix I Participating hospitals.
© 2015 Royal Australasian College of Physicians
509
Assessing the use of initial oxygen therapy in chronic
obstructive pulmonary disease patients: a retrospective audit
of pre-hospital and hospital emergency management
C. Susanto1 and P. S. Thomas1,2
1
Department of Respiratory Medicine, Prince of Wales Hospital and 2Inflammation and Infection Research Centre (IIRC), Faculty of Medicine, University
of New South Wales, Sydney, New South Wales, Australia
Key words
oxygen, emergency, chronic obstructive
pulmonary disease, ambulance, hospital.
Correspondence
Clarissa Susanto, Department of Respiratory
Medicine, Prince of Wales Hospital, PO Box
295, Sydney, NSW 2060, Australia.
Email: [email protected];
[email protected]
Received 25 August 2014; accepted 1 February
2015.
doi:10.1111/imj.12727
Abstract
Background: Carbon dioxide retention in chronic obstructive pulmonary disease
(COPD) exacerbations can be a complication of oxygen therapy. Current recommendations suggest an inspired oxygen level (FiO2) < 0.28, aiming for saturation (SpO2) of
88–92% until arterial blood gas analysis is available.
Aims: This study aims to assess the use of O2 therapy and FiO2 in the emergency
management of patients with a known diagnosis of COPD.
Methods: Retrospective audit of 150 COPD patients admitted over 18 months, data
being extracted from the hospital records.
Results: Of the records reviewed, 57% were male, mean age 75 years. COPD
was recognised in 53%. SpO2 recorded in 124 patients, with SpO2 < 88% seen in
40 patients. Oxygen was administered in 123 patients in ambulances; high flow
in 111 patients, and only 12 patients received O2 therapy in line with the recommended FiO2 < 0.28. In the emergency department (ED), 112 patients received
O2 supplementation; high flow given in 68 patients. Hypercapnia was seen in
71 patients; FiO2 > 0.28 given in 54 patients in ambulances and in 35 patients
in ED. Non-invasive ventilation was required in 53 patients; FiO2 > 0.28 given in 29
patients in the ED. Seven patients were admitted to intensive care unit, and 10 patients
died.
Conclusion: High-flow oxygen is used for the initial treatment of COPD exacerbations, but only 53% are recognised as having COPD. A FiO2 > 0.28 is
often initiated before admission and continued in the ED. A larger study would
be required to assess any possible harm of this approach, but education of those
involved in the care of COPD patients may reduce the risk of complications of
hypercapnia.
Introduction
The administration of oxygen (O2) is a common intervention during acute medical care in emergency situations. It
is often initially administered in high concentrations.
Hypoxaemia should be treated without delay, but the
development of carbon dioxide (CO2) retention following
O2 therapy is a complication seen in some patients with
lung diseases including chronic obstructive pulmonary
disease (COPD). Hypercapnia in COPD patients following
O2 therapy is a well-established risk that has been
Funding: None.
Conflict of interest: None.
described in many papers dating back more than 50
years.1–4
The current recommendation by the British Thoracic
Society (BTS) and the Australian COPDX Plan is that the
fractional inspired O2 (FiO2) should be no more than 0.28
when administered to patients with an exacerbation of
COPD, until the result of an arterial blood gas analysis
(ABG) is available, and for nebulised bronchodilators to
be given with compressed air rather than oxygen as the
driving gas.5,6 The importance of controlled O2 therapy
may be a lower priority in pre-hospital and emergency
settings as failure to correct inadequate oxygenation is
often perceived to be more dangerous than excessive
administration.7–9 Hypercapnic respiratory failure and
respiratory acidosis associated with a FiO2 > 0.28 are
© 2015 Royal Australasian College of Physicians
510
Audit of oxygen therapy in COPD patients
observed in patients admitted under the care of respiratory services.10
We conducted this study with the aims of assessing the
use of O2 therapy and FiO2 in the emergency management of COPD patients who were admitted to this hospital, and to assess any associated outcomes with regards
to this practice.
Aims
This study assessed retrospectively the use of O2 therapy
and FiO2 in the emergency management of COPD
patients who were admitted to hospital, and to document
the outcomes. We hypothesised that a higher FiO2
administration may be used than recommended and
might be associated with higher rates of morbidity and
mortality.
Methods
A retrospective audit was performed randomly on 150
medical records of patients who were admitted to Prince
of Wales Hospital Respiratory Department with an acute
exacerbation of COPD between 1 January 2011 and 30
June 2013. Patients included in the audit had to present
initially through the emergency department (ED), and
they also had to be brought into the hospital by ambulance. The audit included assessment of the ambulance
records in the paper hospital records, as well as the electronic medical records. Only patients who had a known
pre-existing COPD diagnosis were included in this study.
The diagnosis of COPD was obtained via the diseaserelated group coding provided by the medical records
department and a record of a diagnosis of COPD by a
respiratory physician on an outpatient clinic letter
together with previous appropriate spirometry results
and a history of current or previous smoking. The severity was assessed by the spirometry results based on the
COPDX criteria.7 In cases where the clinic letter did not
specify the severity of COPD, this was recorded as
unknown.
The patients’ demographics, oxygen saturations
(SpO2), oxygen delivery and types of oxygen supplementation, blood gas results, mortality and associated morbidity such as hypercapnia, non-invasive ventilation
(NIV) usage, intensive care unit (ICU) admission and
requirement for intubation were collated. Hypercapnia
was defined as a partial pressure of arterial carbon
dioxide (PaCO2) > 45 mmHg. Oxygen therapy was classified as high flow or low flow, with high flow being
defined as O2 delivery by Hudson Mask (HM), non-rebreather mask (NRB) or via nasal cannula with flow rate
>2 L/min. Oxygen delivery by nasal cannula is highly
variable, but 1–2 L/min of O2 delivers a FiO2 between
0.24–0.38, depending on whether the patient mouth
breathes during the administration.11,12
Results
In this audit of 150 medical records, 86 patients were
male and 64 were female. The mean age of the patient
cohort was 75 years old (SD = 8.7). Outside the hospital,
the initial recognition of COPD was noted in 80 patients
(53%), and the majority of the patients had severe COPD
(Table 1).
Thirty-six patients were on long-term home O2 supplementation and of these, 29 received O2 supplementation
at a flow rate higher than their usual rates. Twenty-one of
these patients had received the FiO2 > 0.28 via either HM
or NRB (HM = 15, NRB = 6). Out of the 29 patients, 11
patients had O2 saturations defined as low by standard
convention (i.e. SpO2 < 94%). The median SpO2 in these
20 patients was 89% (range 60–95%).
Oxygen saturations
The O2 saturation was recorded in 124 patients (83%)
prior to hospital. The mean O2 saturation in patients
audited was 88% (SD 7.73, CI 95% = 86.76–89.24%). An
oxygen saturation of <88% was observed in 40 patients,
SpO2 of 88–92% seen in 42 patients and SpO2 of >92% in
42 patients.
Initial oxygen therapy
Oxygen supplementation was administered at some stage
during the ambulance transfers in 123 patients (82%),
with 111 patients (75%) receiving a FiO2 > 0.28. The
most common device to deliver O2 therapy was via HM
(Fig. 1). Nebulised bronchodilators were given in 25
patients, but in 18 these were administered with 8 L/min
O2 rather than air.
A FiO2 of >0.28 was administered in 33 patients with
SpO2 < 88%, 27 patients with SpO2 88–92% and in 32
patients with SpO2 > 92% (Fig. 2). Of those with SpO2 >
92%, 22 patients received O2 supplementation via HM,
five patients via NRB, five patients via nasal cannula at
Table 1 Chronic obstructive pulmonary disease (COPD) severity in
patients audited (classified based on COPDX criteria of severity)
Severe
Moderate
Mild
Unknown
102 patients
28 patients
10 patients
10 patients
68%
18%
7%
7%
© 2015 Royal Australasian College of Physicians
511
Susanto & Thomas
Figure 1 Types of oxygen therapy given before hospital admission. ( ), Hudson mask (HM); ( ), non-re-breather mask (NRB); ( ), 3–4 L nasal cannula
(NC); ( ), ≤ 2 L NC; ( ), nebulised bronchodilator with air; ( ), other.
>2 L/min and only one patient received O2 supplementation via nasal cannula at ≤2 L/min (Fig. 2).
Of the 80 patients recognised as having a pre-existing
diagnosis of COPD, 51 of them received high flow O2
therapy. Overall, only 12 patients received O2 therapy in
line with the BTS and COPDX recommendation with
FiO2 < 0.28.
Types of oxygen therapy given in the
emergency department
Oxygen supplementation was administered in the ED in
112 patients (75%), and high flow O2 was administered
in 68 patients (45%). The most common type of O2
delivery device in the ED was also a HM (34 patients,
Figure 2 Types of oxygen therapy based on initial saturations. ( ), Hudson mask (HM); ( ), non-re-breather mask (NRB); ( ), 3–4 L nasal cannula (NC);
( ), ≤ 2 L NC; ( ), nebulised bronchodilators with air; ( ), unknown; ( ), no O2.
© 2015 Royal Australasian College of Physicians
512
Audit of oxygen therapy in COPD patients
Figure 3 Types of oxygen therapy given in ED. ( ), Hudson mask (HM); ( ), non-re-breather mask (NRB); ( ), 3–4 L nasal cannula (NC); ( ), bi-level
non-invasive ventilation (NIV); ( ), ≤2 L NC; ( ), Venturi mask (VM); ( ), other.
Fig. 3). Long-standing hypercapnia was previously
known to be present in 39 patients, and O2 supplementation with a FiO2 > 0.28 was administered in 24 of these
patients.
PCO2 level on a VBG. An initial ABG was performed in 52
patients (35%) in the ED.
Morbidity
Blood gas measurements
Blood gas analysis was performed in 93% of patients
arriving in the ED with an exacerbation of COPD. Venous
blood gas (VBG) was the most common type of blood gas
measurement, being performed in 88 patients (59%),
although recognised not to reflect accurately an arterial
blood gas (ABG).13 Of the 88 patients who had VBG
measurement, 41 patients had a venous CO2 level above
the range for a normal arterial value. ABG was performed
subsequently in 13 out of the 41 patients who were
deemed to have an elevated partial pressure of carbon
dioxide (PCO2) level on a VBG. These ABG measurements did confirm the presence of hypercapnia in
all of the patients shown to have an elevated
Hypercapnia was seen in 71 patients, and in these
patients a FiO2 > 0.28 was given in 54 patients outside the
hospital and continued in 35 patients in ED (Table 2).
NIV was required in 53 patients, of whom 29 were given
a FiO2 > 0.28, 19 patients were given a FiO2 < 0.28, and
five patients were given an unknown FiO2 in ED. Intubation was required in only one patient, and a FiO2 > 0.28
was administered in this patient in the ambulance as well
as in ED. The majority of patients were managed in the
ward, with only seven patients requiring admission to the
ICU or high dependency unit (HDU). The median length
of stay (LOS) for those receiving low flow O2 was 6.5 days
(range 2–27 days), and those receiving high flow O2 was
5 days (range 1–32 days).
Table 2 Morbidity based on FiO2 given in the ambulance and in emergency department (ED)
In ambulance
In ED
FiO2 < 0.28
FiO2 > 0.28
Unknown
FiO2 < 0.28
FiO2 > 0.28
Unknown
8 (36%)
6 (27%)
1 (5%)
54 (48%)
38 (34%)
5 (5%)
9 (56%)
9 (56%)
1 (6%)
29 (43%)
19 (28%)
2 (3%)
35 (52%)
29 (43%)
4 (6%)
7 (47%)
5 (33%)
1 (7%)
Acute hypercapnia (71 patients)
NIV (53 patients)
ICU/HDU (7 patients)
FiO2, fraction of inspired oxygen; ICU, intensive care unit; NIV, non-invasive.
© 2015 Royal Australasian College of Physicians
513
Susanto & Thomas
Mortality
There was a total of 10 deaths, of which nine patients had
known severe COPD. In those who died, a FiO2 > 0.28
was given in six patients in ambulances and in eight
patients in the ED.
Discussion
This audit confirms that oxygen administration is a
common medical intervention for emergency treatment
of COPD exacerbations, and that high flow oxygen is
often administered. Based on current guidelines, the
majority of the patients had not required the initial
oxygen supplementation given that their mean oxygen
saturation was ≥88% or could have been managed on a
lower FiO2.
This audit shows that a diagnosis of COPD was not
necessarily appreciated as being a relative contraindication for high-flow supplemental oxygen. Interestingly, a
previous audit performed in the United Kingdom had
also shown a similar problem indicating that a lack of
recognition of COPD is widespread.8 Initial assessments
outside hospital often recorded asthma, cardiac failure or
simply ‘shortness of breath’ as an alternative pre-existing
diagnosis. This lack of recognition of a pre-existing diagnosis of COPD could account for the high number of
uncontrolled oxygen administration episodes; however,
more than half of those patients recognised as having
pre-existing diagnosis of COPD still had received a
FiO2 > 0.28.
It is understandable that, when presented with an
acutely ill patient who cannot breathe or communicate,
oxygen supplementation takes priority, and this is appropriate. A long-term history of carbon dioxide retention
would not be obtained in such circumstances. This audit
also shows that while high-flow oxygen therapy is often
initiated in the ambulances, in more than half of cases
this was continued in the ED and probably a downtitration of this treatment was required.
Administration of high-flow oxygen in general leads to
an increase in minute ventilation, which leads to a lower
end-tidal carbon dioxide concentration.14 In patients with
COPD, however, hyperoxia leads to a decreased minute
ventilation and increase in transcutaneous carbon
dioxide.15 These changes have been postulated to be a
result of either depression of ventilation or due to worsening ventilation perfusion inequality relating to recruitment of poorly ventilated lung units by reversal of local
hypoxic pulmonary vasoconstriction with subsequent
release of sequestered carbon dioxide.16 The clinical
effects of supplemental oxygen-induced hypercapnia
have been known for a long time, and these include
Table 3 Variables associated with hypercapnia
Home O2
Severe COPD
Age >60 years
FiO2 > 0.28 in
ambulance
FiO2 > 0.28 in ED
Unadjusted
OR
Adjusted
OR
95%
CI
P-value
4.76
—
—
—
—
4.64
6.09
2.05
2.04–11.11
1.63–13.22
1.18–31.36
0.70–6.01
<0.01
<0.01
0.03
0.19
—
1.37
0.59–3.15
0.46
CI, confidence interval; FiO2, fraction of inspired oxygen; OR, odds ratio.
depression of neurological and cardiorespiratory
function.17,18
In a study of blood gas data of patients with COPD, a
negative correlation was shown between pH and partial
pressure of arterial oxygen (PaO2) after O2 therapy with
increased oxygenation associated with a lower pH, especially in those with hypercapnia.19 Several other studies
have also reported a greater incidence of adverse outcomes associated with higher flow oxygen in COPD
patients.8,10,20
While a higher proportion of those who were given
high-flow oxygen therapy developed hypercapnia and
required NIV in this study, logistic regression analysis
showed that the higher FiO2 administered was not necessarily a significant variable associated with the development of these morbidities (Tables 3, 4). This result
could however be related to missing data with an
unknown FiO2 and a lack of statistical power. This study
showed that the severity of COPD (OR 4.64, 95% CI
1.63–13.22) and older age group (OR 6.09, 95% CI 1.18–
31.36) were significant variables associated with the
development of hypercapnia. In an unadjusted OR analysis, pre-existing home oxygen usage was also significantly
associated with development of hypercapnia, likely to be
a reflection of the underlying severity of the COPD.
There was an overall mortality rate of 6.7%, which
is similar to the 6–10% rates quoted in other
studies.10,19–22 The length of stay in our study is longer
than the quoted average in the National Health Performance Authority (NHPA) of 5 days in COPD admission
Table 4 Variables associated with usage of NIV
Severe COPD
Age >60 years
FiO2 > 0.28 in ambulance
FiO2 > 0.28 in ED
Adjusted OR
95% CI
P-value
1.93
0.32
1.05
5.10
0.56–6.64
0.07–1.41
0.32–3.45
1.79–14.50
0.30
0.13
0.94
<0.01
CI, confidence interval; COPD, chronic obstructive pulmonary disease;
ED, emergency department; NIV, non-invasive; OR, odds ratio.
© 2015 Royal Australasian College of Physicians
514
Audit of oxygen therapy in COPD patients
without complications.23 However, the NHPA also
acknowledges that there is significant variation in the
LOS between 2.1 to 2.5 times longer depending on
the hospitals and underlying complications and comorbidities. In our study, there was a wide range in the
LOS in the low and high-flow oxygen group and obviously oxygen administration is not the sole determinant
factor of LOS. Duration of admission in these patients
was affected not only by their underlying COPD and
treatment, but also reflects contributions of other factors
such as other comorbidities, need for rehabilitation,
mobility, placement or social issues, etc.
Blood gas measurements were performed, but there
was a high proportion of VBG measurements rather than
the recommended arterial analysis. Although this study
shows that the VBG prediction of PCO2 level matches that
of the ABG in the 13 patients who had ABG repeated
following VBG sampling, a recent meta-analysis by Byrne
et al. showed that while VBG analysis compares well to
ABG for pH estimations, VBG results show an unacceptably large variability in the PCO2 and partial pressure of
oxygen (PO2). At present the accepted view is that VBGs
are unable to estimate the PaO2 and PaCO2 and furthermore, VBGs are not commonly used outside Australasia.13
A similar study performed by Joosten et al. in a Melbourne university teaching hospital found comparable
results to ours, with similar demographic characteristics
and findings that high flow O2 administration is a
common practice that is initiated in the ambulance and
then carried on in the ED.24 Also similar to our study,
Joosten pointed out the significant proportion of
patients who did not have ABG performed as part of
their COPD management, further highlighting this widespread practice.24 While the study by Joosten had a
smaller number of audit samples, they managed to show
significant relationship between higher PaO2 measured
(≥74.5 mmHg) and morbidity variables such as increased
LOS, NIV usage and HDU admission.24 These findings’
differences with ours likely reflect the different measurement of relationships with the variables rather than a
contradiction of findings. We did not use PaO2 as a
measurement variable given the high proportion of
VBG performed which limited our ability to document
PO2 confidently.
Overall, this study has confirmed that un-titrated
administration of oxygen in COPD patients is a common
practice. An audit that was recently performed in
Waikato hospital in New Zealand further highlights
that the practice of un-titrated oxygen administration is
common and is not only limited to the respiratory specialty.25 We acknowledge that given that this is an audit
study with multiple variables, it is not possible to
conclude that there is a causal relationship between a
higher FiO2 administration and adverse outcomes such
as hypercapnia or need for NIV. Indeed, other variables
such as COPD severity or age also play a significant part
in the development of morbidity and mortality in this
cohort and could have been detailed along with type of
exacerbation and medication. We acknowledge the limitations of this study being that of a retrospective study
with a relatively small sample size and a proportion of
missing data, all of which limit the analysis of this
study. To date, there has only been one randomised
controlled trial that studied the relationship between
high flow oxygen therapy and mortality in patients
with COPD which indicated a reduced risk of death
in patients with titrated oxygen therapy.20 Larger, prospective studies would be required to confirm the possible harm of un-titrated oxygen approach in COPD
patients.
The implementation of current oxygen guidelines for
use in the pre-hospital setting and education of those
who are involved in treating COPD patients in emergency care and other situations may reduce the risk
of complications in COPD patients.6,7 Oxygen is not
commonly titrated in the pre-hospital setting and
administered using high-flow delivery devices on the
perception that failure to correct oxygenation is more
dangerous than excessive administration.26 Perhaps consideration of developing oxygen administration guidelines depending on the initial SpO2 would be useful as
this could potentially prevent administration of high–
flow oxygen as first line treatment. Beasley et al. recommended no oxygen supplementation for those with
a SpO2 > 92%, 2–3 L O2 via nasal cannula in those with
SpO2 85–92% and usage of simple masks at higher
flows for those with SpO2 < 85%, titrated to aim for
a SpO2 > 92%.27 Other potential useful interventions
which can be considered include education of patients
regarding their diagnosis of COPD, the importance of
alerting ambulance and medical personnel to the diagnosis and the provision of a medical alert bracelet or
oxygen alert card, especially in those with known
hypercapnia. The utilisation of a dedicated oxygen prescription chart, which has been used in some hospitals,
may also be of some benefit.25
Conclusion
Our study highlights that high flow oxygen is commonly
used for the initial treatment of COPD exacerbations but
that the diagnosis of COPD is often not recognised. The
administration of oxygen supplementation with a FiO2
>0.28 is often initiated during ambulance transfers and
subsequently continued in the ED. A larger prospective
study would be required to confirm any possible harm of
© 2015 Royal Australasian College of Physicians
515
Susanto & Thomas
this approach, but education of those involved in the care
of COPD patients may reduce the risk of complications of
hypercapnia.
Hospital for their invaluable help in obtaining the
disease-related group coding and medical records. We
also thank Professor Jenny Peat for her invaluable statistical advice.
Acknowledgements
We thank Eilish Portelli, Annie Blenkinsopp and
the Medical Records Department at Prince of Wales
References
1 Campbell EJM. A method of controlled
oxygen administration which reduces
the risk of carbon dioxide retention.
Lancet 1960; 2: 12–14.
2 Donald KW. Neurological effects of
oxygen. Lancet 1949; 2: 1056–7.
3 Westlake EK, Simpson T, Kaye M.
Carbon dioxide narcosis in emphysema.
Q J Med 1955; 94: 155–73.
4 Smith J, Stone R, Muschenheim C.
Acute respiratory failure in chronic lung
disease: observations on controlled
oxygen therapy. Am Rev Respir Dis 1968;
97: 791–803.
5 Aubier M, Murciano D, Milic-Emili J,
Touaty E, Daghfous J, Pariente R et al.
Effects of administration of O2 on
ventilation and blood gases in patients
with chronic obstructive pulmonary
disease during acute respiratory failure.
Am Rev Respir Dis 1980; 122:
747–54.
6 O’Driscoll BR, Howard LS, Davison AG,
British Thoracic Society. British Thoracic
Society guideline for emergency oxygen
use in adult patients. Thorax 2008;
63(Suppl 6): vi1–68.
7 McKenzie DK, Frith PA, Burdon JGW,
Town GI. The COPDX Plan: Australia
and New Zealand Guidelines for the
management of Chronic Obstructive
Pulmonary Disease 2003. Med J Aust
2003; 178(Suppl): S1–40.
8 Durrington HJ, Flubacher M, Ramsay
CF, Howard LSGE, Harrison BDW.
Initial oxygen management in patients
with an exacerbation of chronic
obstructive pulmonary disease. Q J Med
2005; 98: 499–504.
9 Denniston AK, O’Brien C, David S. The
use of oxygen in acute exacerbations of
chronic obstructive pulmonary disease: a
prospective audit of pre-hospital and
hospital emergency management. Clin
Med 2002; 2: 449–51.
10 Cameron L, Pilcher J, Weatherall M,
Beasley R, Perrin K. The risk of serious
adverse outcomes associated with
hypoxaemia and hyperoxaemia in acute
exacerbations of COPD. Postgrad Med J
2012; 88: 684–9.
11 Wijesinghe M, Perrin K, Healy B, Hart
K, Clay J, Weatherall M et al.
Pre-hospital oxygen therapy in acute
exacerbations of chronic obstructive
pulmonary disease. Intern Med J 2011;
41: 618–22.
12 Bazuaye E, Stone T, Corris P, Gibson G.
Variability of inspired oxygen
concentration with nasal cannulas.
Thorax 1992; 47: 609–11.
13 Byrne A, Bennett M, Chatterji R,
Symons R, Pace NL, Thomas PS.
Peripheral venous and arterial blood gas
analysis in adults: are they comparable?
A systematic review and meta-analysis.
Respirology 2014; 19: 168–75.
14 Becker HF, Polo O, McNamara SG,
Berthon Jones M, Sullivan CE. Effect of
different levels of hyperoxia on
breathing in healthy subjects. J Appl
Physiol 1996; 81: 1683–90.
15 Sassoon CSH, Hassell KT, Mahutte CK.
Hypoxic induced hypercapnia in stable
chronic obstructive pulmonary disease.
Am Rev Respir Dis 1987; 135: 907–11.
16 Wettstein RB, Shelledy DC, Peters JI.
Delivery oxygen concentrations using
low-flow and high-flow nasal cannulas.
Respir Care 2005; 50: 604–9.
17 Seevers MH. The narcotic properties of
carbon dioxide. N Y State J Med 1944; 44:
597–602.
18 Sicker HO, Hickarn JB. Carbon dioxide
intoxication. Medicine (Baltimore) 1956;
35: 389–423.
19 Plant PK, Owen JL, Elliott MV. One year
prevalence study of respiratory acidosis
in acute exacerbations of COPD:
implications for provision of
non-invasive ventilation and oxygen
administration. Thorax 2000; 55: 550–4.
20 Austin M, Wills E, Blizzard L, Walters
EH, Wood-Baker R. Effect of high flow
oxygen on mortality on chronic
obstructive pulmonary disease patients
in prehospital setting: randomised
controlled trial. BMJ 2010; 341: c546.
21 Roberts CM, Stone RA, Buckingham RJ,
Pursey NA, Lowe D. Acidosis,
non-invasive ventilation and mortality
in hospitalized COPD exacerbations.
Thorax 2011; 66: 43–8.
22 Connors AF Jr, Dawson NV, Thomas C,
Harrell FE Jr, Desbisens N, Fulkerson
WJ et al. Outcomes following acute
exacerbation of severe chronic
obstructive lung disease. The SUPPORT
investigators (Study to Understand
Prognoses and Preferences for Outcomes
and Risks of Treatments). Am J Respir
Crit Care Med 1996; 154: 959–67.
23 National Health Performance Authority.
Hospital Performance: Length of Stay in
Public Hospital in 2011–12. Sydney, NSW:
NHPA; 2013; 16–17. Length of stay for
selected medical conditions, Chronic
Obstructive Pulmonary Disease.
24 Joosten SA, Koh MS, Bu X, Smallwood
D, Irving LB. The effects of oxygen
therapy in patients presenting to an
emergency department with
exacerbation of chronic obstructive
pulmonary disease. Med J Aust 2007;
185: 235–8.
25 Holbourn A, Wong J. Oxygen
prescribing practice at Waikoto Hospital
does not meet guideline
recommendations. Intern Med J 2014; 44:
1231–4.
26 Branson RD, Johannigman JA.
Pre-hospital oxygen therapy. Respir Care
2013; 58: 86–97.
27 Beasley R, Aldington S, Robinson G. Is it
time to change the approach to oxygen
therapy in the breathless patient? Thorax
2007; 62: 840–1.
© 2015 Royal Australasian College of Physicians
516
Obvious emphysema on computed tomography during an acute
exacerbation of chronic obstructive pulmonary disease predicts
a poor prognosis
T. Cheng,1 H. Y. Wan,1 Q. J. Cheng,1 Y. Guo,2 Y. R. Qian,2 L. Fan,2 Y. Feng,2 Y. Y. Song,3 M. Zhou,2 Q. Y. Li,2
G. C. Shi2 and S. G. Huang2
1
Department of Respiratory Medicine, Ruijin Hospital, North, 2Department of Respiratory Medicine, Ruijin Hospital and 3Department of Biostatistics,
Shanghai Jiaotong University School of Medicine, Shanghai, China
Key words
chronic obstructive, pulmonary disease, acute
exacerbation of chronic obstructive pulmonary
disease (AECOPD), X-ray computed,
tomography, emphysema, prognosis.
Correspondence
QiJian Cheng, Department of Respiratory
Medicine, Ruijin Hospital North, Shanghai
Jiaotong University School of Medicine, No.
999, Xiwang Road, Malu Town, Jiading,
Shanghai, 201800, China.
Email: [email protected];
[email protected]
HuanYing Wan, Department of Respiratory
Medicine, Ruijin Hospital North, Shanghai
Jiaotong University School of Medicine, No.
999, Xiwang Road, Malu Town, Jiading,
Shanghai, 201800, China.
Email: [email protected]
Received 20 August 2014; accepted 12 January
2015.
doi:10.1111/imj.12723
Abstract
Background: Emphysematous change on computed tomography (CT) during the
stable phase of chronic obstructive pulmonary disease (COPD) is reported to correlate
with COPD prognosis. Acute exacerbation of COPD (AECOPD) is associated with a high
risk of mortality and a poor prognosis.
Aims: This study aims to study the relationship between prognosis and emphysematous changes on CT during an AECOPD.
Methods: Histories were recorded, and CT acquired for 106 patients who visited the
emergency department for an AECOPD. Emphysematous change was quantified by
measuring the percentage of low-attenuation areas (LAA%) in the entire lung on CT
images with a threshold of –950 Hounsfield units. Other factors that could influence
AECOPD prognosis were also recorded on admission and analysed. At follow ups
conducted in 1 year, patient survival, the modified Medical Research Council (mMRC)
Dyspnoea Scale, and performance status (PS) were evaluated, and a COPD Assessment
Test (CAT) was completed.
Results: The 1-year follow up was completed by 103 of 106 patients. The median
LAA% was significantly higher in non-survivors (11%, n = 16) than in survivors
(5.69%, n = 87) (P = 0.006) at the 1-year follow up. LAA% was significantly correlated
with mMRC grade (r = 0.285, P = 0.008), PS (r = 0.397, P < 0.001) and CAT score (r =
0.27, P = 0.017) at the 3-month follow up, and with mMRC grade (r = 0.405, P < 0.001)
and PS (r = 0.377, P < 0.001) at the 1-year follow up. LAA% > 7.5% was a significant
predictor of 1-year mortality, higher mMRC and PS at the 3-month and 1-year follow
ups, after adjustment for other prognostic predictors.
Conclusion: Obvious emphysematous changes on CT (LAA% > 7.5%) during an
AECOPD predicts a poor prognosis independent of other known indicators.
Introduction
Chronic obstructive pulmonary disease (COPD) is characterised by persistent airflow limitation and is a leading
cause of morbidity and mortality worldwide.1 An acute
exacerbation of COPD (AECOPD) is an acute event
characterised by worsening of the patient’s respiratory
Funding: This study was funded by: (i) The Chronic Disease
Prevention and Treatment Programme of the Shanghai
ShenKang Hospital Development Centre, China (Shanghai
ShenKang Yi Yuan Fa Zhan Zhong Xin Man Xing Bing Zong He
Fang Zhi Xiang Mu) (SHDC12012305); and (ii) The 11th
National 5-year Development Plan (2008BAI52B00).
Conflict of interest: None.
symptoms beyond normal day-to-day variations, and
leads to changes in medication.1 It contributes to overall
disease severity in individual patients.1 The mean frequency of exacerbations is 0.9 times per year, according
to the ECLIPSE (Evaluation of COPD Longitudinally to
Identify Predictive Surrogate End-points) cohort.2 The
prognosis of an AECOPD is poor. In the United States, the
in-hospital mortality was 4.3% in 2006.3 In Chang’s
study, the 30-day mortality was 8.5% (21/248), and the
1-year mortality was 18.5% (42/227) among inpatients
in New Zealand.4
Screening high-risk patients helps in selecting more
aggressive treatments for certain patients while avoiding
unnecessary treatment in others. However, knowledge
about the determinants of prognosis is limited.
© 2015 Royal Australasian College of Physicians
517
Cheng et al.
In stable COPD, the severity of emphysema as measured by low-attenuation areas (LAA%) has been established to be a stronger predictor of mortality than lung
function, age or body mass index (BMI).5 The LAA% is
positively correlated with dyspnoea severity (the modified Medical Research Council (mMRC) Dyspnoea Scale)
and negatively correlated with forced expiratory volume
in 1 s (FEV1) % predicted, the 6-min walk test distance
(6MWT) and BMI.6 However, to the best of our knowledge, the prognostic value of LAA% in an AECOPD has
not yet been studied.
Many studies have shown that after an exacerbation,
most patients gradually return to their baseline level, and
almost no patient will attain better lung function than in
the stable phase of their disease.7,8 During the exacerbation, the aggravation of airflow limitations is mainly
caused by greater mucus production, airway wall oedema
and bronchoconstriction.9 These changes are reversed
during recovery. In contrast, emphysema is defined as the
abnormal, permanent enlargement of air spaces distal
to the terminal bronchioles, and accompanied by the
destruction of their walls.10 Therefore, we hypothesised
that the severity of emphysema may reflect the irreversibility and prognosis of an AECOPD.
The present study was designed to assess prospectively
the ability of LAA% measurements during COPD exacerbations to predict the mortality and severity of COPD
after the exacerbation.
Methods
Patients
Consecutive patients with a primary AECOPD diagnosis
who visited the emergency department from December
2011 to May 2012 were recruited. An AECOPD was
diagnosed by the admitting physician and defined as two
out of three of: an increase in dyspnoea, sputum volume
or sputum purulence from COPD beyond normal day-today variations that required emergency treatment.11
COPD was diagnosed on the basis of history and spirometry (Fig. 1) and confirmed by spirometry when the
patients were stable whenever possible. Exclusion criteria
were a history of other respiratory illnesses, such as lung
cancer, pneumothorax, hydrothorax, severe bronchiectasis, thorax malformation, destroyed lung, illness too
severe to undergo routine examinations (for example,
haemodynamic instability) and those who visited the
emergency department for reasons other than an
AECOPD. Only the first visit was included in the analysis,
even if the patient visited the emergency department
more than once during the study period. Patients
were treated according to Global Initiative for Chronic
Obstructive Lung Disease (GOLD) guidelines.1 The Ethics
Committee of Ruijin Hospital approved the study protocol (2009-23), and informed consent was obtained
from all participants.
Figure 1 Flow chart of cohort enrolment,
evaluation and follow up. AECOPD, acute
exacerbation of COPD; CAT, COPD Assessment Test; COPD, chronic obstructive pulmonary disease; mMRC, modified Medical
Research Council Dyspnoea Scale.
© 2015 Royal Australasian College of Physicians
518
Emphysema during AECOPD and prognosis
Clinical data collection
Table 1 The overall characteristics of the AECOPD patients
Medical history and physiological variables were recorded
during the first visit (Fig. S1). CRB-65 score was calculated
and one point given for each of the following features
present: confusion; respiratory rate ≥ 30/min; diastolic
blood pressure ≤ 60 mmHg or systolic blood pressure < 90
mmHg; and age ≥ 65 years.12 Follow ups were performed
at 1 month, 3 months and 1 year after the first visit. If the
patient had died, survival time and cause of death were
registered. At the 3-month and 1-year follow ups, if the
patient was alive, mMRC grade,1 performance status
(PS),13 COPD Assessment Test (CAT) score14 and severity
of cough and sputum were evaluated (Fig. S2).
Parameter
n/total
Mean ± SD (n)
Median (interquartile range) (n)
Male
Age (years)
BMI (kg/m2)
87/106
75.8 ± 9.8
21.0 ± 3.6
Smoking status
Current smoker
Ex-smoker
Pack-years
23/105
59/105
37.9 ± 24.5
Computed tomography emphysema evaluation
Chest computed tomography (CT) without contrast
media was performed during the first visit. Imaging was
performed during breath holding at full inflation with
the patient in supine positioning using the same scanner
(Light Speed 16; GE Medical Systems, Milwaukee, WI,
USA) and protocol (tube voltage, 120 kV; tube current,
220 mA; tube rotation time, 0.8 s; and collimation,
1.25 mm). Images were reconstructed with the ‘standard
algorithm’ at a 1.25-mm section thickness, 1.25-mm
interval and a 512 × 512 matrix, as in previous studies.15
Sixteen patients underwent follow-up CT with the same
parameters at the 3-month follow-up visit. The LAA%
was calculated automatically using commercial software
(Myrian, Intrasense; Montpellier, France) using a threshold of -950 Hounsfield units to determine emphysematous extent, as in previous studies.16
Statistical analysis
All statistical analyses were conducted with SPSS 17.0
(SPSS, Chicago, IL, USA). The LAA% of patients who died
within 1 month, between 1 month and 1 year, and those
who were alive at the 1-year follow up were rank transformed and compared using a one-way analysis of variance
and the least significant difference method. Spearman’s
correlation was used to evaluate the relationship between
LAA% and mMRC grade, performance status and CAT
score at the follow ups. A Cox regression was used to
examine risk factors for mortality. An ordinal logistic
regression was performed to examine factors influencing
mMRC grade and performance status at the follow ups.
Results
Population characteristics
One hundred and six patients were enrolled (Table 1).
Two patients were lost to follow up within 1 month, and
Comorbidities
Asthma
Diabetes mellitus
Hypertension
Coronary artery disease
Left heart failure
13/106
11/104
41/105
32/103
7/99
Clinical presentation
Increase in cough
Increase in sputum volume
Increase in sputum purulence
Increase in dyspnoea
Fever
92/104
90/103
51/103
94/105
44/98
CRB-65 score
LAA%
1 (1–1) (83)†
6.6 (2.4–12.1) (106)†
3-month follow up
Survival
mMRC
PS
CAT
93/104
2 (1–3) (85)†
1 (1–2) (94)†
11.9 ± 7.6 (78)
1-year follow up
Survival
mMRC
PS
CAT
87/103
2 (1–3) (83)†
1 (1–2) (97)†
13.5 ± 7.7 (73)
†Parameter is not normally distributed and is therefore presented as the
median and interquartile range. AECOPD, acute exacerbation of chronic
obstructive pulmonary disease; BMI, body mass index; CAT, COPD
Assessment Test; CRB-65 score, confusion, respiratory rate ≥ 30/min,
systolic blood pressure < 90 mmHg or diastolic blood pressure ≤ 60
mmHg and age ≥ 65 years; LAA%, the percentage of low-attenuation
areas (emphysema) on computed tomography imaging during an
AECOPD using a threshold of -950 HU; mMRC, modified Medical Research
Council Dyspnoea Scale; PS, performance status.
one patient was lost between the 3-month and 1-year
follow ups (follow-up rate: 98.11% at 1 month and 3
months; 97.17% at 1 year). Mortality was 6.73%
(7/104), 10.58% (11/104) and 15.53% (16/103) at 1
month, 3 months and 1 year, respectively, and a bit lower
than previously reported.3,4,17 The median LAA% was
6.6% (interquartile range: 2.4–12.1%, n = 106) (the
LAA% did not follow a normal distribution in these
patients). Sixteen patients underwent follow-up CT with
© 2015 Royal Australasian College of Physicians
519
Cheng et al.
Figure 2 (A) LAA% in patients who died within 1 month, between 1 month and 1 year, and 1-year survivors. **The LAA% during an acute exacerbation
of chronic obstructive pulmonary disease (AECOPD) was significantly higher in patients who died within 1 year than in 1-year survivors (median
(interquartile range) 11% (7.14–24.5) vs 5.69% (2.35–11.0), P = 0.006). *The LAA% during an AECOPD was significantly higher in patients who died
between 1 month and 1 year than in 1-year survivors (23.2% (2.64–29.9) vs 5.69% (2.35–11.0), P = 0.03). (B) A Kaplan–Meier survival curve for patients
with an AECOPD stratified according to LAA%. Survival was worse in patients with LAA% > 7.5% (P = 0.009, Cox regression). LAA%, the percentage of
low-attenuation areas (emphysema) on computed tomography (CT) imaging during an AECOPD using a threshold of –950 Hounsfield units; y, year; mo,
month; f/u, follow up.
the same parameter. A good correlation (r = 0.840, P <
0.001) and no significant difference (13.38% ± 9.04% vs
11.43% ± 7.1%, P = 0.135) was observed between the
LAA% measured during the exacerbation and during the
stable phase (the LAA% of these patients followed a
normal distribution) (Fig. S3). The median CRB-65 score
was 1 (interquartile range: 1–1, n = 83). The CRB-65
score could not be calculated for 23 patients because of
missing data. There was no significant difference between
patients with or without a CRB-65 score with respect to
LAA% or prognosis (Table S1). Twenty-nine patients
underwent standard pulmonary function tests (mean
FEV1 = 44.77 ± 18.54% predicted), and 13 underwent
bronchodilation tests (mean post-bronchodilator FEV1 =
48.65 ± 13.8% predicted) at follow up (Tables S2, S3).
Most AECOPD patients (86/105) have lung infiltration to
a greater or lesser extent on CT; however, the clinical
significance is limited. The presence of asthma or pneumonia was not significantly associated with any other
factor, including LAA%, CRB-65 score, smoking history
or prognosis (Table S4).
The risk of death and LAA%
The LAA% of patients who died within 1 year was significantly higher than that of survivors (median
(interquartile range), 11% (7.14–24.5%) vs 5.69%
(2.35–11.0%), P = 0.006, Mann–Whitney U-test). The
LAA% of patients who died between 1 month and 1 year
after exacerbation was significantly higher than those still
alive at the 1-year follow up (median (interquartile
range) 23.2% (2.64–29.9%) vs 5.69% (2.35–11.0%),
P = 0.03, Fig. 2a). The risk of death in patients with an
LAA% > 7.5% was significantly higher than in patients
with an LAA% ≤ 7.5% (RR = 4.5, P = 0.009; Fig. 2b).
Compared with the patients who died within 1 month,
patients who died between 1 month and 1 year had a
higher LAA% and lower incidence of fever, although the
results were not statistically significant. However, there
were no statistically significant differences during the first
visit to the emergency department between the group
who died within 1 month and the group who died later
(Table S5).
© 2015 Royal Australasian College of Physicians
520
Emphysema during AECOPD and prognosis
Table 2 An univariate Cox regression analysis of risk factors for 1-year
mortality in patients who came to the emergency department for an
AECOPD
Table 3 A multivariate Cox regression analysis of risk factors for 1-year
mortality in patients who came to the emergency department for an
AECOPD
Characteristics
Characteristics
RR
P value
Age (increment by 1 year)
BMI (kg/m2)
Male
1.063
0.913
1.55
0.047
0.209
0.562
CRB-65
LAA% > 7.5%
Fever (T > 37.3°C)
Clinical presentation
Increase in cough
Increase in sputum volume
Increase in sputum purulence
Increase in dyspnoea
Fever (T > 37.3°C)
0.385
0.389
1.361
1.861
0.279
0.098*
0.102
0.541
0.548
0.046*
AECOPD, acute exacerbation of COPD; CI, confidence interval; CRB-65
score, confusion, respiratory rate ≥ 30/min, systolic blood pressure < 90
mmHg or diastolic blood pressure ≤ 60 mmHg, and age ≥ 65 years; LAA%,
the percentage of low-attenuation areas (emphysema) on CT imaging
during an AECOPD using a threshold of -950 Hounsfield units; RR, risk ratio
of death in 1 year; T, body temperature.
Clinical scores
CRB-65 (increment by 1)
4.219
<0.001*
In the past 1 year
Exacerbations ≥ 2
Hospitalisation because of an AECOPD
1.569
2.147
0.368
0.139
Past medical history
Asthma
Hypertension
Diabetes
Coronary artery disease
Left heart failure
History of smoking
0.488
0.664
1.131
1.118
0.8
1.855
0.489
0.448
0.87
0.839
0.829
0.413
CT emphysema evaluation
LAA > 7.5%
4.545
0.009*
*Factors related to the prognosis with P < 0.1 in the univariate
analysis, which were selected for multivariate Cox regression
analysis. AECOPD, acute exacerbation of COPD; CI, confidence interval;
COPD, chronic obstructive pulmonary disease; CRB-65 score, confusion,
respiratory rate ≥ 30/min, systolic blood pressure < 90 mmHg
or diastolic blood pressure ≤ 60 mmHg, and age ≥ 65 years;
LAA%, the percentage of low-attenuation areas (emphysema)
on computed tomography imaging during an AECOPD using a threshold of -950 Hounsfield units; RR, risk ratio of death in 1 year; T, body
temperature.
RR
P
95% CI
2.836
3.891
0.259
0.001
0.039
0.085
1.55–5.19
1.07–14.1
0.06–1.21
The severity of symptoms after stabilisation
and LAA%
LAA% was significantly correlated with mMRC grade
(r = 0.285, P = 0.008), performance status (r = 0.397, P <
0.001) and CAT score (r = 0.27, P = 0.017) at the 3-month
follow up; and mMRC grade (r = 0.405, P < 0.001) and
performance status (r = 0.377, P < 0.001) at the 1-year
follow up (Table 4). However, the correlations were not
significant between LAA% and the CAT score at the
1-year follow up, exacerbation frequency, severity of
cough or severity of sputum (Table 4). In the stratified
analysis, relationships between LAA% and dyspnoea
symptoms after stabilisation were more significant
in men, patients with smoking histories and patients
without histories of left heart failure or asthma
(Table S6).
In univariate analyses, age, absence of increase
in cough during the exacerbation, CRB-65 score,
Table 4 The correlation between LAA% during an AECOPD and COPD
symptoms at follow up
3-month follow up
In the stratified analysis, the relationship between
LAA% and mortality was more significant in patients
with a history of smoking and without a history of left
heart failure or asthma (Table S6).
In the univariate Cox proportional hazard regression,
higher CRB-65 scores, ageing, absence of fever during
the exacerbation and LAA% > 7.5% were significantly
related to mortality (Table 2).
After performing a multivariate Cox regression using
CRB-65, LAA%, presence of fever and presence
of increase in cough, the following were mortality
predictors: higher CRB-65 scores (RR = 2.836, P = 0.001)
and LAA% > 7.5% (RR = 3.891, P = 0.039)
(Table 3).
mMRC grade
Performance status
CAT score
Severity of cough
Severity of sputum
Frequency of exacerbations
Frequency of hospitalisations
for exacerbations
1-year follow up
r
P
r
P
0.285
0.397
0.27
0.018
0.051
0.128
0.008
<0.001
0.017
0.869
0.644
0.247
0.405
0.377
0.18
-0.05
-0.045
0.09
0.219
<0.001
<0.001
0.127
0.66
0.689
0.429
0.051
CAT, COPD Assessment Test; COPD, chronic obstructive pulmonary
disease; LAA%, the percentage of low-attenuation areas (emphysema) on
computed tomography imaging during an AECOPD using a threshold of
-950 Hounsfield units; mMRC, modified Medical Research Council
Dyspnoea Scale.
© 2015 Royal Australasian College of Physicians
521
Cheng et al.
Table 5 Univariate and multivariate ordinal logistic regression analyses of factors influencing the mMRC scale and performance status at the 3-month
follow up in patients who came to emergency department for an AECOPD
Characteristics
M3 mMRC (increment by 1)
Univariate
analysis
M3 PS (increment by 1)
Multivariate ordinal
logistic regression
OR
P
OR
Age (increment by 1 year)
BMI
Male
1.126
0.991
1.332
<0.001
0.867
0.565
Clinical presentation
Increase in cough
Increase in sputum volume
Increase in sputum purulence
Increase in dyspnoea
Fever (T > 37.3°C)
0.121
0.334
0.416
3.102
0.732
0.003
0.089
0.032
0.081
0.447
0.134
2.638
0.263
2.329
0.063
0.315
0.014
0.351
Clinical scores
CRB-65 (increment by 1)
5.129
<0.001
4.034
In the past 1 year
Exacerbation ≥ 2
Hospitalisation due to an AECOPD
1.925
6.917
0.117
<0.001
7.434
Medical history
Asthma
Hypertension
Diabetes
Coronary artery disease
Left heart failure
History of smoking
0.64
1.13
1.059
1.571
4.693
0.742
0.463
0.759
0.928
0.29
0.035
0.534
CT emphysema evaluation
LAA% > 7.5%
2.91
0.01
10.47
3.195
P
95% CI
Univariate
analysis
OR
P
1.147
0.949
1.034
<0.001
0.31
0.946
0.02–1.11
0.40–17.5
0.09–0.76
0.39–13.8
0.230
0.217
0.764
5.807
0.696
0.008
1.44–11.3
0.002
2.11–26.2
0.018
1.49–73.8
0.036
1.08–9.45
Multivariate ordinal
logistic regression
OR
P
95% CI
0.023
0.008
0.489
0.017
0.37
1.412
0.599
0.723
0.545
0.21–9.46
0.11–3.14
1.27
0.792
0.21–7.54
5.646
<0.001
5.327
<0.001
2.29–12.4
1.968
3.337
0.094
0.013
1.13
2.713
0.805
0.076
0.43–2.98
0.90–8.18
0.382
1.716
1.323
1.271
2.079
1.095
0.132
0.890
0.964
0.569
0.308
0.846
3.662
0.001
3.619
0.014
1.30–10.0
Age was not utilised in the multivariate logistic regression because it was embodied in the CRB-65 score. Odds ratios (OR) refer to the change in OR of PS
or mMRC grade at least 1 unit higher at follow up with an increase in the predictor variables by 1 unit. Bold values refer to the OR of the independent
prognosis predictors. AECOPD, acute exacerbation of COPD; BMI, body mass index; CAT, COPD Assessment Test; CI, confidence interval; COPD, chronic
obstructive pulmonary disease; CRB-65 score, confusion, respiratory rate ≥ 30/min, systolic blood pressure < 90 mmHg or diastolic blood pressure ≤ 60
mmHg, and age ≥ 65 years; LAA%, the percentage of low-attenuation areas (emphysema) on computed tomography imaging during an AECOPD using a
threshold of -950 Hounsfield units; mMRC, modified Medical Research Council Dyspnoea Scale; M3, 3-month follow up; M12, 1-year follow up; PS,
performance status; T, body temperature.
AECOPD-related hospitalisations in the past year and
LAA% > 7.5% predicted poor mMRC grades and performance statuses at follow up. A history of left heart failure
also predicted a poor mMRC grade; an increase in dyspnoea, and no increase in sputum volume during exacerbations also predicted a poor performance status at follow
up (Tables 5,6).
In the multivariate analysis, LAA% > 7.5% remained
a significant predictor of higher mMRC grades at the
3-month (odds ratio (OR) = 3.195, P = 0.036) and
1-year follow ups (OR = 3.414, P = 0.028), and poor
performance statuses at the 3-month (OR = 3.619, P =
0.014) and 1-year follow ups (OR = 4.751, P = 0.017)
(Tables 5,6).
Discussion
Previous studies significantly correlated emphysematous
extent on CT with disease severity and prognosis.5,6
However, this is the first study that compared emphysema severity on CT during an exacerbation and prognosis to our knowledge. In the present study, there was a
very good correlation and no significant difference
between LAA% measured during the exacerbation and
in the stable phase. This suggested that LAA% during an
AECOPD has a similar prognostic predictive effect as
LAA% during the stable phase. CT is more often performed for patients with exacerbations to evaluate
lung consolidation, differentiate from pneumothorax or
© 2015 Royal Australasian College of Physicians
522
Emphysema during AECOPD and prognosis
Table 6 Univariate and multivariate ordinal logistic regression analyses of factors influencing the mMRC score and performance status at the 1-year
follow up in patients who came to emergency department for an AECOPD
Characteristics
M12 mMRC (increment by 1)
Univariate
analysis
OR
P
Age (increment by 1 year)
BMI
Male
1.070
0.949
1.679
0.001
0.368
0.301
Clinical presentation
Increase in cough
Increase in sputum volume
Increase in sputum purulence
Increase in dyspnoea
Fever (T > 37.3°C)
0.130
0.197
0.632
4.411
0.747
0.007
0.019
0.255
0.020
0.477
Clinical scores
CRB-65 (increment by 1)
6.521
In the past 1 year
Exacerbations ≥ 2
Hospitalisation due to an AECOPD
M12 PS (increment by 1)
Multivariate ordinal
logistic regression
OR
P
95% CI
Univariate
analysis
OR
P
1.089
0.937
1.182
<0.001
0.209
0.734
0.152
0.207
0.582
6.456
0.334
0.002
0.007
0.159
0.005
0.008
Multivariate ordinal
logistic regression
OR
P
95% CI
0.222
1.143
0.152
0.891
0.03–1.74
0.17–7.73
8.430
0.345
0.047
0.059
1.03–69.0
0.11–1.04
0.164
1.82
0.081
0.529
0.02–1.25
0.28–11.75
7.516
0.028
1.24–45.42
<0.001
4.428
0.007
1.5–13.07
9.300
<0.001
5.380
0.001
1.90–15.2
1.644
2.945
0.230
0.038
1.232
0.741
0.36–4.24
1.362
3.714
0.425
0.005
2.117
0.232
0.62–7.24
Past medical history
Asthma
Hypertension
Diabetes
Coronary artery disease
Left heart failure
History of smoking
0.800
1.026
1.071
1.837
17.64
1.808
0.702
0.949
0.917
0.160
0.001
0.220
0.494
0.934
1.296
2.305
3.522
1.322
0.226
0.86
0.671
0.047
0.082
0.547
2.450
2.728
0.174
0.327
0.67–8.91
0.37–20.2
CT emphysema evaluation
LAA% > 7.5%
3.987
0.001
3.770
0.001
4.751
0.017
1.32–17.1
19.53
3.414
0.007
2.24–170.4
0.028
1.14–10.24
The multivariate logistic regression was performed only on influencing factors related to the mMRC grade or performance status in the univariate analyses
with P < 0.1. Age was not utilised in the multivariate logistic regression because it was embodied in the CRB-65 score. Odds ratios (OR) refer to the change
in OR ratios of PS or mMRC grade at least 1 unit higher at follow up with an increase in the predictor variables by 1 unit. Bold values refer to the OR of the
independent prognosis predictors. AECOPD, acute exacerbation of chronic obstructive pulmonary disease; BMI, body mass index; CAT, COPD Assessment Test; CI, confidence interval; CRB-65 score, confusion, respiratory rate ≥ 30/min, systolic blood pressure < 90 mmHg or diastolic blood pressure ≤
60 mmHg, and age ≥ 65 years; LAA%, the percentage of low-attenuation areas (emphysema) on CT imaging during an AECOPD using a threshold of -950
HU; M3, 3-month follow up; M12, 1-year follow up; mMRC, the modified Medical Research Council Dyspnoea Scale; PS, performance status; T, body
temperature.
hydrothorax and screen for post-obstructive pneumonia
caused by neoplasms. Emergency CT imaging is inexpensive and readily available at our hospital.
The LAA% of patients who died between 1 month and
1 year was even higher than those who died within 1
month (although not statistically significant). LAA% may
reflect the mid-term risk of death to a greater extent than
the short-term risk. That is, patients without obvious
emphysema (LAA% ≤ 7.5%) may have better reversibility. LAA% differs from other indices mainly related to the
risk of death within 1 month.
In the present study, mMRC grade, performance status
and CAT score at follow up were registered as end-points
in addition to mortality. The mMRC scale is a 5-point
scale to evaluate COPD-related dyspnoea severity, recommended by GOLD guidelines,1 and correlates significantly
with respiratory symptom severity, disability18 and mortality.19 Performance status is a 6-point scale formulated
by the Eastern Cooperative Oncology Group to evaluate
performance and disability, formerly used in patients
with malignant tumours and validated to be a predictor
of mortality in COPD.20,21 In the present study, LAA%
was correlated with mMRC grade and performance status
at both the 3-month and 1-year follow ups, suggesting
that LAA% is correlated with COPD severity and mortality for more extended durations.
The correlation was not significant between LAA% and
chronic bronchitis symptoms, such as cough and sputum
© 2015 Royal Australasian College of Physicians
523
Cheng et al.
after stabilisation, or between LAA% and exacerbation
frequency. These results agree with findings of previous
studies showing that chronic bronchitis symptoms22 and
exacerbation risks6 are more related to bronchial wall
thickening than emphysematous changes.
The CAT is an 8-item measure of health status impairment from COPD, correlates closely with health status
measured by the St George’s Respiratory Questionnaire,
and is reliable and responsive.1,14 The correlation
between LAA% during exacerbations and CAT scores at
the 3-month follow up was limited but significant, while
the correlation between LAA% and CAT scores at the
1-year follow up was not significant. There are two possible reasons for this: first, the CAT includes each aspect
of COPD symptoms, while LAA% correlates to dyspnoea
rather than chronic bronchitis symptoms. Second, many
patients completed the CAT questionnaire with the help
of others because of low education and weakness, which
may have caused errors; this problem was also described
previously.23
The median LAA% (6.6%) and criterion for obvious
emphysematous change (7.5%) in the present research
were much lower than in other cohorts, such as in
Haruna’s study (22.1% and 32.3% respectively).5 This
may be because, first, patients with thickened bronchial
walls rather than severe emphysema tend to suffer from
frequent exacerbations.6,24 Second, the present study was
based on emergency department patients, some of whom
did not manifest severe dyspnoea in the stable phase2 and
did not regularly seek treatment for COPD, and therefore
were not registered in an outpatient department-based
cohort. Third, the severity of emphysema differs among
ethnic groups, even with similar lung function impairment.25 The COPD patients with hereditary alpha-1
antitrypsin deficiency, who often manifest severe emphysema, are extremely rare in China.26 Since we screened
almost all AECOPD patients who visited our emergency
department without selection, and the mortality was
only a little lower than in other cohorts,3,4,17 we consider
that this cohort covered a wide range of GOLD categories
and different degrees of emphysema.
In the present study, we diagnosed COPD based on
clinical presentation and bedside spirometry in addition
to previous diagnoses, because only a few patients with
COPD had been diagnosed previously in China, especially
those with mild COPD.27 Patients with a history of
asthma or left heart failure in addition to COPD were not
excluded, because we consider them to be representative
of some COPD patients. Many who reported to be suffering from asthma (‘xiao chuan’ in Chinese) were actually
suffering from COPD. In some patients with chronic
asthma, COPD may coexist.1 The incidence of concomitant COPD and asthma was reported to be 2604/6059 in
a Medicaid population.28 Left heart failure is another
common COPD comorbidity. Left ventricular dysfunction
was found in 32% of COPD patients with symptomatic
deterioration.29 Even in patients with stable COPD
and without a cardiologist-confirmed diagnosis of heart
failure, roughly 30% had heart failure to some extent.30
Although the cardinal symptoms of COPD and left heart
failure are similar, dyspnoea and exercise intolerance, left
heart failure does not cause obstructive airflow limitation.31 Therefore, FEV1/FVC < 70% indicates the existence of COPD, regardless of the presence of left heart
failure.
Patients with radiographic consolidation were not
excluded, because pneumonia is a common AECOPD
complication. In previous studies, 15–36.3% of inpatients
with an AECOPD had radiographic consolidation3,32,33
and were not excluded from a state audit.3,33 Furthermore, only a portion (12%/15%) would be diagnosed
with pneumonia based on paired radiographs.32
In the present study, the CRB-65 score was one of the
best predictors of mortality, severity of dyspnoea and
disability after stabilisation. The CRB-65 is a 4-point
score, originally used for risk stratification in communityacquired pneumonia. Recently, CRB-65 was reported to
predict in-hospital and 30-day mortality, but not 1-year
mortality.12,34 In our study, the CRB-65 score predicted
1-year mortality, mMRC grade and performance status at
the 1-year follow up. This may be because those enrolled
in the previous study were inpatients, while the CRB-65
score and CURB-65 score are portions of the admission
criteria.35 The present study enrolled almost all AECOPD
patients who visited the emergency department without
selection, thereby including different levels of severity,
making the predictive ability of the CRB-65 score more
obvious.
The present study showed that a history of left heart
failure predicts a higher mMRC grade at the 3-month
and 1-year follow ups independently. It is suggested
that left heart failure may aggravate dyspnoea in COPD,
which confuses severity assessments using the mMRC
grade. Indeed, the widely used New York Heart Association functional classification for left heart failure is
similar to the mMRC scale.36 These findings suggest the
necessity of grading criteria to assess COPD-related dyspnoea and left heart failure separately.
Limitations
The present study had some limitations. First, some prognostic predictors, such as blood gas analyses and lung
function tests were not analysed in this study, because
only a few patients completed these tests in the emergency department. Thus, it remained undetermined
© 2015 Royal Australasian College of Physicians
524
Emphysema during AECOPD and prognosis
whether the LAA% is a prognostic factor independent of
the blood gas analysis and lung function test. Second, the
CRB-65 score could not be calculated because of missing
data in some patients. However, there were no significant
differences in LAA% and prognosis between the patients
with or without CRB-65 score. Among the patients with
CRB-65 score, the LAA% was proven to be a prognosis
predictor independent of CRB-65. Third, only a few
patients underwent the follow-up CT with the same
parameter. Therefore, the CT manifestation of exacerbation could hardly be analysed in detail in this study. The
relationship between CT imaging and the aetiology of
exacerbation awaits further investigation. The impact of
exacerbation on emphysema progression, as shown in a
previous study,37 could not be analysed in the present
study. Fourth, among the several CT indices, only LAA%
References
1 Global Strategy for the Diagnosis,
Management and Prevention of COPD,
Global Initiative for Chronic Obstructive
Lung Disease (GOLD). 2011 [cited 2011
Feb 21]. Available from: http://www.
goldcopd.org/
2 Agusti A, Calverley PM, Celli B, Coxson
HO, Edwards LD, Lomas DA et al.
Characterisation of COPD heterogeneity
in the ECLIPSE cohort. Respir Res 2010;
11: 122.
3 Perera PN, Armstrong EP, Sherrill DL,
Skrepnek GH. Acute exacerbations of
COPD in the United States: inpatient
burden and predictors of costs and
mortality. COPD 2012; 9: 131–41.
4 Chang CL, Robinson SC, Mills GD,
Sullivan GD, Karalus NC, McLachlan JD
et al. Biochemical markers of cardiac
dysfunction predict mortality in acute
exacerbations of COPD. Thorax 2011; 66:
764–8.
5 Haruna A, Muro S, Nakano Y, Ohara T,
Hoshino Y, Ogawa E et al. CT scan
findings of emphysema predict mortality
in COPD. Chest 2010; 138: 635–40.
6 Han MK, Bartholmai B, Liu LX, Murray
S, Curtis JL, Sciurba FC et al. Clinical
significance of radiologic
characterizations in COPD. COPD 2009;
6: 459–67.
7 Niewoehner DE, Erbland ML, Deupree
RH, Collins D, Gross NJ, Light RW et al.
Effect of systemic glucocorticoids on
exacerbations of chronic obstructive
pulmonary disease. Department of
Veterans Affairs Cooperative Study
8
9
10
11
12
13
14
15
was analysed in the present study. Thus, it remains
unclear whether other CT indices were prognostic
factors. Lastly, the sample size of the present study was
small. Despite these limitations, the LAA% was still
proven to be associated with the prognosis, even after
adjustment for other co-variables.
Conclusions
The severity of emphysematous change on CT during
an acute exacerbation of COPD was correlated
with the risk of death in 1 year, and the severity of
dyspnoea and disability at 3 months and 1 year after
exacerbation. Obvious emphysema on CT during
an AECOPD is an independent predictor of a poor
prognosis.
Group. N Engl J Med 1999; 340:
1941–7.
Seemungal TA, Donaldson GC,
Bhowmik A, Jeffries DJ, Wedzicha JA.
Time course and recovery of
exacerbations in patients with chronic
obstructive pulmonary disease. Am J
Respir Crit Care Med 2000; 161:
1608–13.
Hurst JR, Wedzicha JA. The biology of a
chronic obstructive pulmonary disease
exacerbation. Clin Chest Med 2007; 28:
525–36.
The definition of emphysema. Report of
a National Heart, Lung, and Blood
Institute, Division of Lung Diseases
workshop. Am Rev Respir Dis 1985; 132:
182–5.
Burge S, Wedzicha JA. COPD exacerbations: definitions and classifications. Eur
Respir J Suppl 2003; 41: 46s–53s.
Edwards L, Perrin K, Wijesinghe M,
Weatherall M, Beasley R, Travers J. The
value of the CRB65 score to predict
mortality in exacerbations of COPD
requiring hospital admission. Respirology
2011; 16: 625–9.
Oken MM, Creech RH, Tormey DC,
Horton J, Davis TE, McFadden ET et al.
Toxicity and response criteria of the
Eastern Cooperative Oncology Group.
Am J Clin Oncol 1982; 5: 649–55.
Jones PW, Harding G, Berry P, Wiklund
I, Chen WH, Kline LN. Development
and first validation of the COPD
Assessment Test. Eur Respir J 2009; 34:
648–54.
Han MK, Kazerooni EA, Lynch DA, Liu
LX, Murray S, Curtis JL et al. Chronic
16
17
18
19
20
21
obstructive pulmonary disease
exacerbations in the COPDGene
study: associated radiologic
phenotypes. Radiology 2011; 261:
274–82.
Martinez CH, Chen YH, Westgate PM,
Liu LX, Murray S, Curtis JL et al.
Relationship between quantitative CT
metrics and health status and BODE in
chronic obstructive pulmonary disease.
Thorax 2012; 67: 399–406.
Roberts CM, Stone RA, Lowe D,
Pursey NA, Buckingham RJ.
Co-morbidities and 90-day outcomes in
hospitalized COPD exacerbations. COPD
2011; 8: 354–61.
Bestall JC, Paul EA, Garrod R, Garnham
R, Jones PW, Wedzicha JA. Usefulness
of the Medical Research Council (MRC)
dyspnoea scale as a measure of disability
in patients with chronic obstructive
pulmonary disease. Thorax 1999; 54:
581–6.
Nishimura K, Izumi T, Tsukino M, Oga
T. Dyspnea is a better predictor of 5-year
survival than airway obstruction in
patients with COPD. Chest 2002; 121:
1434–40.
Chen CZ, Ou CY, Wang WL, Lee CH,
Lin CC, Chang HY et al. Using
post-bronchodilator FEV(1) is better
than pre-bronchodilator FEV(1) in
evaluation of COPD severity. COPD
2012; 9: 276–80.
Roberts CM, Lowe D, Bucknall CE,
Ryland I, Kelly Y, Pearson MG. Clinical
audit indicators of outcome following
admission to hospital with acute
exacerbation of chronic obstructive
© 2015 Royal Australasian College of Physicians
525
Cheng et al.
22
23
24
25
26
27
pulmonary disease. Thorax 2002; 57:
137–41.
Mair G, Maclay J, Miller JJ, McAllister
D, Connell M, Murchison JT et al.
Airway dimensions in COPD:
relationships with clinical variables.
Respir Med 2010; 104: 1683–90.
Ringbaek T, Martinez G, Lange P. A
comparison of the assessment of quality
of life with CAT, CCQ, and SGRQ in
COPD patients participating in
pulmonary rehabilitation. COPD 2012; 9:
12–15.
Fujimoto K, Kitaguchi Y, Kubo K,
Honda T. Clinical analysis of chronic
obstructive pulmonary disease
phenotypes classified using
high-resolution computed tomography.
Respirology 2006; 11: 731–40.
Hansel NN, Washko GR, Foreman MG,
Han MK, Hoffman EA, DeMeo DL et al.
Racial differences in CT phenotypes in
COPD. COPD 2013; 10: 20–7.
Kwok JS, Lawton JW, Yew WW, Chau
CH, Lee J, Wong PC. Protease inhibitor
phenotypes and serum
alpha-1-antitrypsin levels in patients
with COPD: a study from Hong Kong.
Respirology 2004; 9: 265–70.
Fang X, Wang X, Bai C. COPD in China:
the burden and importance of proper
management. Chest 2011; 139: 920–9.
28 Shaya FT, Dongyi D, Akazawa MO,
Blanchette CM, Wang J, Mapel DW et al.
Burden of concomitant asthma and
COPD in a Medicaid population. Chest
2008; 134: 14–19.
29 Render ML, Weinstein AS, Blaustein AS.
Left ventricular dysfunction in
deteriorating patients with chronic
obstructive pulmonary disease. Chest
1995; 107: 162–8.
30 Rutten FH, Cramer MJ, Grobbee DE,
Sachs AP, Kirkels JH, Lammers JW et al.
Unrecognized heart failure in elderly
patients with stable chronic obstructive
pulmonary disease. Eur Heart J 2005; 26:
1887–94.
31 Wasserman K, Zhang YY, Gitt A,
Belardinelli R, Koike A, Lubarsky L et al.
Lung function and exercise gas
exchange in chronic heart failure.
Circulation 1997; 96: 2221–7.
32 Lieberman D, Lieberman D, Gelfer Y,
Varshavsky R, Dvoskin B, Leinonen M
et al. Pneumonic vs nonpneumonic
acute exacerbations of COPD. Chest
2002; 122: 1264–70.
33 Myint PK, Lowe D, Stone RA,
Buckingham RJ, Roberts CM. U.K.
National COPD Resources and Outcomes
Project 2008: patients with chronic
34
35
36
37
obstructive pulmonary disease
exacerbations who present with
radiological pneumonia have worse
outcome compared to those with
non-pneumonic chronic obstructive
pulmonary disease exacerbations.
Respiration 2011; 82: 320–7.
Chang CL, Sullivan GD, Karalus NC,
Mills GD, McLachlan JD, Hancox RJ.
Predicting early mortality in acute
exacerbation of chronic obstructive
pulmonary disease using the CURB65
score. Respirology 2011; 16:
146–51.
Lim WS, Baudouin SV, George RC, Hill
AT, Jamieson C, Le Jeune I et al. BTS
guidelines for the management of
community acquired pneumonia in
adults: update 2009. Thorax 2009;
64(Suppl 3): i1–55.
New York Heart Association. Diseases of
the Heart and Blood Vessels: Nomenclature
and Criteria for Diagnosis. Boston, MA:
Little, Brown and Co.; 1964.
Tanabe N, Muro S, Hirai T, Oguma T,
Terada K, Marumo S et al. Impact of
exacerbations on emphysema
progression in chronic obstructive
pulmonary disease. Am J Respir Crit Care
Med 2011; 183: 1653–9.
Supporting Information
Additional Supporting Information may be found in the online version of this article at the publisher’s web-site:
Figure S1 The questionnaire used during an acute exacerbation of COPD.
Figure S2 The questionnaire used at the 3-month and 1-year follow ups after an acute exacerbation of COPD.
Figure S3 The correlation between the LAA% on computed tomography (CT) during exacerbations and stable phases
(Pearson’s correlation). LAA%: the percentage of low-attenuation areas (emphysema) on CT imaging during an acute
exacerbation of chronic obstructive pulmonary disease (AECOPD) using a threshold of –950 HU.
Table S1 Comparisons between patients with and without a CRB-65 score.
Table S2 Main pulmonary function test data at follow up and during an exacerbation.
Table S3 Complete pulmonary function test data at the 3-month follow up.
Table S4a Comparisons between patients with and without asthma.
Table S4b Comparisons between patients with and without infiltration.
Table S5 Comparisons between patients who died within 1 month, who died between 1 month–1 year and survivors
at 1-year follow up.
Table S6 The correlation between low-attenuation areas (LAA%) during an acute exacerbation of chronic obstructive
pulmonary disease (AECOPD) and COPD symptoms at follow up stratified by other factors.
© 2015 Royal Australasian College of Physicians
526
Efficacy of non-invasive mechanical ventilation in the general
ward in patients with chronic obstructive pulmonary disease
admitted for hypercapnic acute respiratory failure and pH <
7.35: a feasibility pilot study
S. Fiorino,1 L. Bacchi-Reggiani,2 E. Detotto,1 M. Battilana,1 E. Borghi,1 C. Denitto,1 C. Dickmans,1 B. Facchini,1
R. Moretti,1 S. Parini,1 M. Testi,1 A. Zamboni,1 A. Cuppini,1 L. Pisani3 and S. Nava3
1
Unità Operativa di Medicina Interna, Ospedale di Budrio (Bologna), Department of Internal Medicine, Azienda Unità Sanitaria Locale di Bologna and
Istituto di Cardiologia and 3Terapia Intensiva Pneumologia S. Orsola, Azienda Ospedaliera Policlinico S. Orsola-Malpighi, Department of Specialist,
2
Diagnostic, and Experimental Medicine, School of Medicine, University of Bologna, Bologna, Italy
Key words
chronic obstructive pulmonary disease, acute
respiratory failure, non-invasive mechanical
ventilation.
Correspondence
Sirio Fiorino, Unità Operativa di Medicina
Interna, Ospedale di Budrio, Via Benni 44,
40065 Budrio (Bologna), Italy.
Email: sirio.fi[email protected]
Received 29 October 2014; accepted 12
January 2015.
doi:10.1111/imj.12726
Abstract
Aim: To date non-invasive (NIV) mechanical ventilation use is not recommended in
chronic obstructive pulmonary disease (COPD) patients with acute respiratory failure
(ARF) and pH < 7.30 outside a ‘protected environment’. We assessed NIV efficacy and
feasibility in improving arterial blood gases (ABG) and in-hospital outcome in patients
with ARF and severe respiratory acidosis (RA) admitted to an experienced rural
medical ward.
Methods: This paper is a prospective pilot cohort study conducted in the General
Medicine Ward of Budrio’s District Hospital. Two hundred and seventy-two patients
with ARF were admitted to our Department, 112, meeting predefined inclusion
criteria (pH < 7.35, PaCO2 > 45 mmHg). Patients were divided according to the
severity of acidosis into: group A (pH < 7.26), group B (7.26 ≤ pH < 7.30) and group C
(7.30 ≤ pH < 7.35). ABG were assessed at admission, at 2–6 h, 24 h, 48 h and at
discharge.
Results: Group A included 55 patients (24 men, mean age: 80.8 ± 8.3 years), group B
31 (12 men, mean age: 80.3 ± 9.4 years) and group C 26 (15 men, mean age:
78.6 ± 9.9 years). ABG improved within the first hours in 92/112 (82%) patients,
who were all successfully discharged. Eighteen per cent (20/112) of the patients
died during the hospital stay, no significant difference emerged in mortality rate
(MR) within the groups (23%, 16% and 8%, for groups A, B and C, respectively) and
between patients with or without pneumonia: 8/29 (27%) versus 12/83 (14%). On
multivariable analysis, only age and Glasgow Coma Scale had an impact on the clinical
outcome.
Conclusion: In a non-‘highly protected’ environment such as an experienced medical
ward of a rural hospital, NIV is effective not only in patients with mild, but also with
severe forms of RA. MR did not vary according to the level of initial pH.
Introduction
Funding: None.
Conflict of interest: S. Nava: speaking fees from PhilipsRespironics International – Resmed International – Weinman
International; travel grants: Weinman; research grants from
Starmed – Fisher and Paykel; free loan of equipment from:
Maquet – Air Liquide – Resmed – Philips-Respironics – Breas –
Siare.
Acute respiratory failure (ARF) with decompensated
respiratory acidosis represents a frequent reason for
hospital admission of patients with chronic obstructive
pulmonary disease (COPD).1 Recent research has shown
that COPD is very often associated with different
comorbidities that considerably impact on the management and the prognosis of these patients.2,3 Despite the
burden that COPD is increasing and that it will be the
third cause of death in a few years, the number of
patients admitted to the intensive care unit (ICU) has
© 2015 Royal Australasian College of Physicians
527
Fiorino et al.
decreased in the last decades. This may be partly due to
better pharmacological and rehabilitative strategies but
also to the early use of non-invasive mechanical ventilation (NIV) outside of a highly protected environment like
an ICU or a respiratory ICU (RICU). The early use of such
a therapeutic approach, when appropriate, may prevent
clinical deterioration of these patients, reducing risk of
endotracheal intubation (ETI) and mortality. However,
the pH level has been reported to be a crucial factor in
determining the NIV success rate.4,5 Although the confidence of healthcare providers with NIV has progressively
improved and although NIV has been applied with
success also outside the critical care environment,6,7 to
date NIV is not recommended outside ICU in patients
with severe hypercapnic ARF and a pH < 7.30.8 In particular, it was shown that NIV treatment failed in
approximately 50–60% of COPD-exacerbated subjects
with a baseline pH < 7.25. Indeed, concerns were also
placed on treating these patients outside a protected environment when they are affected by comorbidities.
We have prospectively performed a feasibility pilot
study with the aim to assess efficacy of NIV on mortality
rate in the ‘every day clinical practice’ at the General
Medical Ward of a small rural Italian hospital without
ICU, focusing our analysis on patients presenting acute or
acute-on-chronic respiratory failure and severe respiratory acidosis (pH < 7.35) Secondary outcomes were the
influence of acute and chronic comorbidities on patients’
outcomes. Therefore, the results may be useful to calculate sample sizes for future definitive studies.
Methods
We performed a prospective-cohort study in the General
Medicine Ward of Budrio’s District Hospital with the
‘external’ support of the RICU of S. Orsola-Malpighi Hospital at Bologna. If the patient met the intubation criteria,
it was agreed to transfer her/him promptly to that unit,
which is approximately 15 km far away.
Budrio District Hospital is a rural hospital of 80 beds
with medicine and surgery units as well as emergency
department but without ICU.
We enrolled consecutive COPD patients with
hypercapnic ARF or with exacerbations on chronic RF
(AHRF) admitted to our medical ward over a 13-month
period (from 1 January 2013 to 31 January 2014). All
these patients were first admitted to our emergency
room, and if they showed signs of AHRF, they were sent
to the doctor on duty at the medical ward. Only the
patients who met the inclusion criteria (see below) were
therefore consecutively enrolled.
Inclusion criteria were pH < 7.35 and PaCO2 >
45 mmHg, in association with a respiratory rate > 25
breaths/min and massive activation of accessory (or secondary) respiratory muscles. The patients were subsequently divided into three groups according to the
severity of acidosis:
• Group A included 55 patients with pH < 7.26;
• Group B included 31 patients with pH between 7.26
and 7.299;
• Group C included 26 patients with pH ≥ 7.30.
Exclusion criteria included multiple organ failure,
haemodynamic instability, acute ischaemic heart disease,
cerebrovascular accident, facial deformity preventing
adequate mask fitting, upper airway obstruction, any
causes of metabolic acidosis; gastrointestinal bleeding or
surgery, severe psychiatric conditions with psychomotor
agitation, cardiac or respiratory arrest with need of
urgent intubation and NIV refusal. Any type of metabolic
acidosis was excluded on the basis of the ‘classical’ arterial blood gases (ABG) criteria that is the presence of pH
< 7.35 and pCO2 < 45 mmHg with bicarbonate lower than
the normal range. In addition, we enrolled in our study
only patients with a body mass index < 30, to minimise
the chances that patients with overlap syndrome could be
included. Seven patients (four in group A, two in group B
and one in group C) had previously been treated with
NIV for an episode of acute COPD exacerbation, whereas
no patient was enrolled in a home care NIV programme.
COPD diagnosis was performed, according to a 1987
American Thoracic Society statement on the basis of pulmonary function tests (PFT) in 68% of our patients,
whereas in the remaining subset, it was carried out based
on the ‘usual criteria’ employed in most of the published
NIV trials4,5,8,9 to define COPD in absence of PFT, which
includes clinical history, physical examination and
imaging data, such as chest radiograph or high-resolution
computed tomography scan. In addition, health status of
each patient was assessed by Charlson Index comorbidity
score at hospital admission based on the clinical history
and patient’s previous and actual records.10 The protocol
was approved by our local Ethical Committee and written
consent was obtained from all patients. Before starting
NIV treatment, COPD patients received a standard
medical treatment, including aerosolised bronchodilator
drugs, intravenous steroid and, when necessary, furosemide and antibiotics.
NIV settings
Patients were treated with pressure support ventilation
with a fixed back-up rate (12 breaths/min) (Resmed
VSIII, ResMed, Sydney, NSW, Australia) supplied by
means of a double-tube circuit through an oro-nasal
non-vented mask (Performatrak total mask Respironics,
Pittsburgh, PA, USA).
© 2015 Royal Australasian College of Physicians
528
COPD: non-invasive ventilation
Different sizes of interfaces were available at patients’
bedside during NIV start. Peak inspiratory pressure was
initially set at 14 cmH2O and gradually increased to a
maximum of 34 cmH2O (median value was 22 cmH2O,
range 14–34 cmH2O) to obtain an expired tidal volume of
7–8 mL/kg, according to patients’ tolerance, whereas the
positive end-expiratory pressure was initially set at 4
cmH2O and increased to a maximum of 8 cmH2O or
reduced to resolve patients’ hypoxaemia or decrease their
discomfort respectively (median value was 6 cmH2O,
range 4–8 cmH2O). An arterial oxygen saturation ranging
between 90% and 95% was obtained with oxygen supply
by means of an adequate FiO2 setting, NIV was started
directly in the emergency department by the respiratory
physician on call. Patients were then transferred (average
time 2.4 ± 2.1 h) to the medical ward, where four beds
may be monitored and are utilised for very critically ill
patients. NIV was always prescribed and started by the
attending physician. The nurses were in charge to fix the
interfaces and to monitor the patients. NIV was applied
intermittently, for periods of at least 4 h, with a minimal
duration of 8 h per day, or continuously in case of
hypercapnic coma and was maintained until improvement of clinical signs and ABG parameters were obtained.
During NIV treatment in absence of clinical worsening,
both in day and night-time, each patient was examined
every 30 min by the attending nurses and every hour
by the attending physician, whereas arterial blood
pressure, oxygen saturation, respiratory rate and electrocardiogram were continuously monitored. NIV was
discontinued when considered clinically indicated (see
Measurements section).
Since we started the use of NIV in our unit several
years ago, our medical and nursing staff received
a specific training for NIV delivery with periodic retraining by a chest physician from our unit. The median
nurse/patient ratio in our unit is equal to 1:12. Lacking
an ICU, the decision to intubate the patient was taken by
the attending anaesthetist (24 h on duty) according to
our hospital guidelines (i.e. respiratory arrest, gasping for
air, deterioration or no improvement of ABG after 2 h of
NIV, sensorium deterioration, severe dyspnoea with sign
of incipient muscle fatigue). The adoption of limits on life
support and treatment (e.g. do not intubation order) was
left to patient’ or his relatives’ decision, in accordance
with the anaesthesiologist, because no specific Italian law
regulates this topic. Intubated patients were eventually
transferred to our ‘back-up’ RICU.
Measurements
The data recorded at hospital admission were age, sex
and Glasgow coma Scale at admission. Number and types
of acute and chronic non-respiratory comorbidities were
defined, according to the CI,10 ABG levels at admission
and within 2 h, 24 h, 48 h after NIV start and at discharge, as well as the length of in-hospital stay and
in-hospital mortality. The following pathological conditions were considered as causes of acute RF in the
enrolled patients: (i) acute exacerbations of COPD; (ii)
cardiogenic pulmonary oedema or congestive heart
failure; (iii) pneumonia; (iv) pneumothorax; and (v)
pulmonary embolism.
In accordance with previous studies, objective criteria
were used to define when to start the discontinuation
process of NIV: an increase in pH value ≥ 7.35, a decrease
in pCO2 of >15–20% and in respiratory rate ≥20%, with
oxygen saturation ≥90% in comparison with spontaneous breathing. SaO2 and ECG were continuously
monitored.9 Lung function test was carried out in 70/112
of patients in the preceding year.11
Statistical analysis
Continuous data were expressed as mean ± standard
deviation (SD) and categorical data as count and percentages. Baseline data and characteristics of A, B and C
groups of patients were compared with one-way analysis
of variance and Bonferroni post-hoc test for continuous
variables and with Chi-squared test for categorical data.
The repeated measures analysis of variance has been
performed in order to study the changes of pH, pCO2 and
pO2/FiO2, across time intervals until patient hospital discharge or death. Risk factors of in-hospital all cause mortality have been assessed by univariate and multivariable
logistic regression analysis. No correlated variables reaching a P-value less than 0.1 at univariate analysis were
included in the multivariable analysis. All P-values refer
to two-tailed tests of significance; P-values < 0.05 were
considered statistically significant. Statistical analyses
were performed using Stata/SE 12.1 (StataCorp LP,
College Station, TX, USA). The aim of the study was to
assess the feasibility of NIV to improve ABG and
in-hospital outcomes in these subjects. Therefore, in this
pilot study, we did not perform a power calculation.
Results
The flow chart of the study is shown in Figure 1.
A total of 112 COPD patients with hypercapnic RF,
meeting predefined inclusion criteria was enrolled in the
trial and then divided into three groups according
to the level of their acidosis. Among the COPD patients,
who were excluded by the study, 15 refused NIV, 39 had
pH ≥ 7.35 and were treated with only medical therapy,
whereas four required immediate intubation and were
© 2015 Royal Australasian College of Physicians
529
Fiorino et al.
Figure 1 The flow chart of the study.
246 patients with acute
respiratory failure
76 patients without
COPD
15 patients refused
NIV
170 patients with
COPD
39 patients with
pH≥7.35 treated with
medical therapy
112 patients meeting
criteria for NIV
NIV failure in 20
patients (20 deaths, no
ETI)
4 patients required
immediate
intubation
NIV success in 92
patients
transferred. Baseline characteristics of enrolled patients
and causes of acute RF are reported in Table 1. Obviously the severity of hypercapnia was significantly
worse in the more acidotic group together with the level
of consciousness. Numbers and types of acute and
chronic comorbidities, according to the Charlson Index,
are shown in Table 2. Overall the number of chronic
comorbidities did not differ between A, B and C groups.
All patients were ventilated at least one night after their
in-hospital admission.
Patients who responded initially well to NIV were 101
of 112 (first few hours), but six of them deteriorated
their pH after 48 h of NIV, and three died during NIV
because of stroke (n.2) and myocardial infarction (n.1)
Eighty-two per cent (92/112) of patients were therefore
discharged alive and considered NIV treated with
success: 42/55 (76%) in group A, 26/31 (84%) in group
B, 24/26 (92%) in group C. None of these patients was
considered intolerant to NIV. Overall 20/112 (18%) subjects died (NIV failure): 13 in group A, five in group B
and two in group C. (P = 0.21). None of them required
ETI because eight had a ‘do not intubation’ order (three
patients discontinued NIV for intolerance), whereas, in
the remaining 12 (four subjects discontinued NIV for
intolerance), ETI was considered inappropriate by the
attending anaesthetist, according to our hospital guidelines, because it was considered futile. The causes of
death in these patients were: stroke (two patients in
group A), myocardial infarction (one in group A), congestive heart failure (four in group A, four in group B
and one in group C), refractory hypoxia due to pneu-
monia (six in group A, one in group B and one in group
C) and are reported in Table 3. Table 4 depicts the
changes in ABG during the time course in the patients
of the three groups. A progressive improvement of arterial pH and pCO2 levels were observed in the subjects of
the three groups (Fig. 2).
Pneumonia was the cause of in-hospital admission in
29/112 (26%) patients (16 in patients with pH < 7.26,
eight in patients with pH between 7.26 and 7.299 and
five patients with pH ≥ 7.30, P = 0.64); 8/29 individuals
with pneumonia died versus 12/83 without pneumonia
(P = 0.19). None of our patients developed pneumonia
during their in-hospital stay. The left side of Table 5
shows the results of the univariate analysis to predict the
NIV failure (NIVF). Remarkably, renal failure was not
entered into the multivariable model due to the very
strong association with in-hospital survival. Only age and
GCS had an increased risk of NIVF, whereas, the prevalence of diabetes was higher in subjects with a better
clinical outcome, although this difference was not
significant (P = 0.08). Interestingly, the presence of
comorbidities, pneumonia and Charlson Index were not
associated with an increased probability of death in our
study. The right side of Table 5 illustrates the results of the
multivariate analysis: age and GCS were independently
associated with NIVF and/or death, whereas a history of
diabetes was associated with a better trend in clinical
outcome.
In-hospital stay was similar in the three groups (14.1 ±
10.71 in group A vs 16.6 ± 11.51 days in group B vs 14.6
± 6.8, P = 0.52).
© 2015 Royal Australasian College of Physicians
530
COPD: non-invasive ventilation
Table 1 Mean (SD) demographic and clinical characteristics of enrolled COPD patients with causes of acute respiratory failure, subdivided according to
pH arterial blood (pH < 7.26, pH between 7.26–7.299 or pH ≥ 7.30)
Characteristics at enrolment
Age (years)
Sex (M/F)
Glasgow Coma Scale
Respiratory rate (breaths/min)
Charlson Index
PaO2/FiO2
PaCO2 (mmHg)
Length of in-hospital stay
Bicarbonate
Patients with pH < 7.26
(group 1): 55
Patients with pH between
7.26–7.299
(group 2): 31
Patients with pH ≥ 7.30
(group 3): 26
P-value
80.8 ± 8.3
24/31
10.6 ± 3.0*
32.3 ± 3.6
3.54 ± 1.27
244.4 ± 93.9
84.7 ± 21.9*§
16.6 ± 11.5
29.9 ± 7.7
80.3 ± 9.4
12/19
12.0 ± 2.6
31.2 ± 2.2
3.48 ± 1.41
230.3 ± 66.3
66.3 ± 13.4
14.1 ± 10.71
31.8 ± 11.8
78.6 ± 9.9
15/11
13.0 ± 1.8
30.4 ± 3.8
2.23 ± 1.06
221.8 ± 69.2
65.6 ± 10.0
14.6 ± 6.8
33.8 ± 4.9
0.6
0.33
0.0009
0.06
0.0001
0.47
0.0001
0.52
0.15
Causes of ARF
Acute exacerbations of COPD
Pneumonia
Congestive heart failure/cardiogenic pulmonary oedema
Pulmonary embolism
Pneumothorax
22
16
15
2
0
14
8
8
1
0
9
5
11
1
0
0.85
Number of comorbidities
1
2
3
4
5
6
2
14
14
19
6
0
1
8
8
11
2
1
1
5
11
6
2
1
0.85
Bonferroni multiple-comparison test: *P < 0.05 group 1 versus group 3. §P < 0.05 group 1 versus group 2. ARF, acute respiratory failure; COPD, chronic
obstructive pulmonary disease.
Discussion
To our knowledge, our prospective observational pilot
study reports, for the first time in a rural hospital, the
feasibility and effectiveness of NIV in improving the
outcome of COPD patients with moderate to severe acidosis (pH < 7.35) outside a protected environment. Based
on the data obtained in the present investigation, with
the assumption to have an expected proportion of death
equal to 10% in the subset of patients at lower risk
(group C) and 25% in the subsets of patients at higher
risk, we need to enlist about 150 patients per group.
In the past few years, NIV use has been increased
worldwide, and it has become the ‘standard of care’ for
the treatment of most episodes of acute exacerbations in
COPD-hospitalised patients.12,13 A recent study performed in the United States over a time span of 11 years,
with the aim of assessing the prevalence and trends of
NIV for acute COPD using data from the Healthcare Cost
and Utilisation Project’s Nationwide Inpatient Sample,
has shown that the use of this type of respiratory support
has surpassed that of invasive mechanical ventilation.14
Data from this investigation did not allow depiction of the
location where patients were treated or their severity at
admission. However, it is very likely that not all the
612.000 patients requiring mechanical support were ventilated in a protected environment like an ICU or RICU,
and that a subset of individuals was also admitted nationwide in tertiary care hospitals. Most of the studies published in the literature were either conducted by an
experienced team and/or in large hospitals with an ICU,
and this may not reflect the ‘daily life’ situation of most of
the rural or small district hospitals.
The International Consensus Conference of NIV dated
back in 200015 was not specific in defining the ideal
location according to patients’ severity since it was stated
that the optimal environment depends on the capacity
for adequate monitoring, staff skill and experience.
In a multicentre randomised study, Plant et al.8, compared NIV with conventional medical therapy to treat
early an episode of ARF in COPD patients, using a
‘simple’ NIV ventilator, minimal monitoring and with a
median 1:11 patient/nurses ratio. NIV was initiated
and maintained by the ward staff according to a strict
© 2015 Royal Australasian College of Physicians
531
Fiorino et al.
Table 2 Number and types of acute and chronic comorbidities in enrolled patients, according to Charlson Index
Charlson
comorbidities
Patients with
Patients with pH
Patients with
pH < 7.26
between 7.26–7.299
pH ≥ 7.30
(group 1)
(group 2)
(group 3)
Myocardial infarction
history
Congestive heart
failure
Peripheral vascular
disease
Cerebrovascular
disease
Chronic pulmonary
disease
Dementia
Connective tissue
disease
Peptic ulcer disease
Mild liver disease
Diabetes
8
8
7
9
6
11
5
4
4
7
3
4
55
31
26
1
0
1
0
3
1
0
0
2
3
8
1
7
1
1
Charlson
comorbidities
Patients with
Patients with pH
Patients with
pH < 7.26
between 7.26–7.299
pH ≥ 7.30
(group 1)
(group 2)
(group 3)
Hemiplegia
1
1
3
Moderate or severe
renal disease
Diabetes with end
organ damage
Any tumour
7
2
1
4
1
3
7
4
1
Leukaemia
1
0
0
Lymphoma
Moderate or severe
liver disease
Metastatic solid
tumour
AIDS
2
2
0
0
0
1
0
0
0
0
0
0
AIDS, acquired immune deficiency syndrome.
protocol and after extensive training. Using prospectively defined criteria, NIV reduced the need for ETI and
hospital mortality. However, in a subgroup analysis,
patients with a pH < 7.30 after 4 h of therapy reported
a prognosis worse than that seen in comparable studies
conducted in the ICU and not better than in the conventional treatment group.
This study raised the concern that NIV could not be
safely used in the presence of moderate to severe acidosis
outside a protected location.
For example in the NIV guidelines, the British Thoracic
Society confirmed that ‘patients with more severe acidosis (pH < 7.30) should be managed in a higher dependency area such as a high dependency unit or ICU, as
should those in whom improvement in clinical state and
arterial blood gas tensions is not seen after 1–2 h of NIV
on a respiratory ward’.16
Interestingly in the same Country (UK) a survey on
more than 200 units revealed that NIV was initiated
in several non-ICU locations, including the emergency
department (54.6% of hospitals), respiratory wards
(51.4%) and general medical wards (18.8%), but once
more there was no mention about the severity of the
episode of ARF.17
To our knowledge, the only two large-sized investigations that tried to assess the percentage and clinical
outcome of patients with severe respiratory failure, who
were treated with NIV outside the wall of a protected
location on the basis of degree of acidosis observed at
hospital admission, were performed in UK and Canada.
The first one18 was an audit on 232 hospital units
obtaining data on 9716 patients, 20% of those with gases
recorded on admission were acidotic and among the 1077
receiving NIV, 55% had a pH < 7.26. Hospital mortality
Table 3 Causes of death in the three groups of patients, according to pH arterial blood (pH < 7.26, pH between 7.26–7.299 or pH ≥ 7.30)
Acute myocardial infarction
Acute ischaemic stroke
Pneumonia
Congestive heart failure
Patients with pH < 7.26
(group 1): 55
Patients with pH between
7.26–7.299 (group 2): 31
Patients with pH ≥ 7.30
(group 3): 26
P-value
1
2
6 (2 patients with NIV early and 1
with NIV late failure)
4 (2 patients with NIV early and 1
with NIV late failure)
0
0
1 (1 subject with NIV late failure)
0
0
1 (1 subject with NIV late failure)
0.59
0.35
0.31
4 (1 patient with NIV early and
1 with NIV late failure)
1 (1 subject with NIV late failure)
0.44
NIV, non-invasive.
© 2015 Royal Australasian College of Physicians
532
COPD: non-invasive ventilation
Table 4 Mean ± SD of arterial blood gas values of the patients with acute exacerbations of COPD
pH
P‡ = 0.0001
pCO2
P‡ = 0.0001
PO2/FiO2
P‡ = 0.37
Bicarbonate
Admission
2–6 h
24 h
48 h
At discharge
P-value†
Group A
Group B
Group C
7.17 ± 0.06
7.28 ± 0.01
7.31 ± 0.01
7.27 ± 0.08*
7.35 ± 0.05*
7.36 ± 0.04*
7.34 ± 0.09*
7.38 ± 0.05*
7.39 ± 0.05*
7.40 ± 0.06*
7.39 ± 0.09*
7.40 ± 0.04*
7.43 ± 0.05*
7.44 ± 0.05*
7.42 ± .04*
0.000
Group A
Group B
Group C
84.7 ± 21.9
66.3 ± 13.4
65.6 ± 10.0
69 ± 18.5*
57.2 ± 12.3*
60.6 ± 11.7*
59.7 ± 17*
55.3 ± 11.2*
59.8 ± 10.4*
55.2 ± 116.4*
55.1 ± 14.4*
56.5 ± 8.0*
47.6 ± 9*
46.9 ± 6.6*
52.2 ± 8.4*
0.000
Group A
Group B
Group C
244.4 ± 93.9
230.3 ± 66.3
221.8 ± 69.2
244.6 ± 84.1
254.9 ± 95.5
241.5 ± 63.9
270.3 ± 89.4
263.5 ± 65.1
239.2 ± 61.2
283.2 ± 87.2
292.8 ± 74.7*
296.6 ± 98.9*
312.1 ± 75.6*
327.3 ± 82.8*
303.6 ± 44.4*
0.000
Group A
Group B
Group C
29.9 ± 7.7
31.8 ± 11.8
33.8 ± 4.9
31.0 ± 6.8
32.0 ± 8.1
35.1 ± 4.5
32.0 ± 6.9
32.5 ± 6.2
36.1 ± 4.3
33.7 ± 7.2
32.6 ± 6.4
35.6 ± 3.8
32.3 ± 5.8
32.5 ± 5.6
34.6 ± 4.9
0.226
P‡ = 0.0001
Repeated measures analysis of variance. *<0.05 in comparison to admission. †From testing among time intervals. ‡From testing among groups A, B and
C. COPD, chronic obstructive pulmonary disease.
was 25% in these ventilated patients, and only 5% of all
acidotic patients in only 91 of 232 secondary care centres
received ETI. These data suggest that a large number of
units do not have in their hospitals ICU availability or
receive adequate critical care support.
The second study19 performed in a Canadian tertiary
care hospital showed that 13% and 23% of their COPD or
Cardiogenic Pulmonary Oedema patients received NIV in
the ward general or the observation ward respectively.
Indeed, 23 of 68 patients had a baseline pH ≤ 7.25 prior
to NIV initiation, 17 of whom had a pH from 7.15 to 7.24.
Of these patients, 16 improved after treatment, suggesting the feasibility of NIV outside a protected area even in
the case of severe acidosis.
Another interesting approach to treat patients with life
threatening ARF is that proposed by Cabrini et al.20 that
demonstrated how, under the supervision of a Medical
Emergency Team in our institution, NIV could be applied
in a wide variety of settings, outside the ICU, with a high
success rate and with few complications. Some years ago,
a previous study carried out in an ED had also suggested
that NIV is effective in the treatment of patients with
severe acidosis, due to acute hypercapnic COPD.21
Our study systematically showed the feasibility of NIV
even in patients with severe acidosis hospitalised in a
general ward with minimal instrumentation and monitoring, and with a patient : nurse rate similar to what was
described by Plant et al.8
However, it is important, to describe better the environment where this study was conducted. The rural hospital of Budrio is located in the bigger Bologna area,
serving alone a total of 100 000 inhabitants, and the
distance from the referring RICU is 15 km that can be
usually covered, in the case of need in <15 min by
ambulance. The Budrio’s hospital team has three
pulmonologists in its staff and a nursing team that initiated the use of NIV a decade ago. In 2011, the hospital
initiated a strict collaboration with the Respiratory and
Critical Care Unit of the Sant’Orsola Malpighi Hospital,
which included formal training of nurses and doctors not
only on the use of NIV ventilators and interfaces, but also
life supporting manoeuvres.
The encouraging data obtained in this study should
therefore not be generalised but placed in this particular
context.
In fact, the about-20% rate of NIV failure is in keeping
with most of the randomised controlled trial (RCT)
studies performed with a similar degree of acidosis.
Rather surprisingly though, we did not observe any statistical difference between the three groups of patients
divided according to their severity of pH. For example, in
patients with a mean pH of 7.17, Squadrone et al.22
reported a higher (>50%) failure rate than that reported
in group A of the present study (25%) with a similar
initial pH. However, they did not record the number and
types of comorbidities of their COPD patients. Several
studies23 have showed that coexisting comorbidities have
a high prevalence in patients with COPD, with percentages ranging from 40% to 95%, and that they represent
independent risk factors both for death in clinically stable
COPD individuals and for in-hospital mortality in COPD
patients after episodes of acute exacerbation. The types
© 2015 Royal Australasian College of Physicians
533
Fiorino et al.
200
250
pO2/FiO2
300
350
Figure 2 Arterial blood gases (ABG)
changes (pO2/FiO2, pCO2 and pH) during the
time course of the study. (
), pH ≥ 7.3;
(
), 7.26 < pH < 7.29; (
), pH <
7.26.
2-6 ore
24 ore
48 ore
dimissione
40
50
60
pCO2
70
80
90
ingresso
2
3
4
5
1
2
3
4
5
7.1
7.2
pH
7.3
7.4
7.5
1
and number of comorbidities in our three groups of
patients were very similar, and this may well explain the
similar outcomes of our patients despite the higher level
of acidosis and more severely compromised level of consciousness. Comorbidities have been shown to be one of
the major determinants of NIV success or failure.23–25 Very
few studies have considered the Charlson Index to assess
the overall severity of these patients, but interestingly the
mean index of our study (between 2.5 and 3.5, according
to the different groups) is rather similar to that described
by others and the overall NIV failure was fairly constant
(around 10–15%).26,27 The question of whether management of patients with a pH < 7.30 in a non-high dependency setting has equivalent outcomes to a high
dependency setting can be solved only with a RCT is very
difficult to perform.
© 2015 Royal Australasian College of Physicians
534
COPD: non-invasive ventilation
Table 5 Predictors of in hospital mortality at logistic regression analysis
Age (years)
Glasgow Coma Scale
pH
7.26–7.299
<7.26
Tumours
Sex
Charlson Index
Diabetes
Chronic heart failure
History of myocardial infarction
Cerebrovascular disease
Cor pulmonale
Univariate analysis
OR (95% CI)
P-value
Multivariable analysis
OR (95% CI)
P-value
1.13 (1.05–1.22)
0.82 (0.69–0.97)
0.001
0.022
1.14 (1.04–1.23)
0.79 (0.66–0.96)
0.003
0.017
2.30 (0.40–13.0)
3.71 (0.77–17.8)
1.44 (0.35–5.82)
0.97 (0.37–2.57)
1.23 (0.87–1.75)
0.16 (0.02–1.24)
1.71 (0.54–5.42)
1.02 (0.31–3.44)
1.35 (0.26–7.03)
2.44 (0.41–14.37)
0.34
0.10
0.60
0.96
0.24
0.08
0.36
0.96
0.72
0.32
CI, confidence interval; OR, odds ratio.
We also seek to find potential variables associated with
NIV failure. This has already been the objective of several
studies, but mostly performed in the ICU or RICU.
Changes in ABG after 1 or 2 h, respiratory rate and lack
of sensorium impairment were the most powerful predictors of success. We confirm the predictive power of
ABG and the rather surprising matter of age. An increasing age was associated with an elevated risk of NIV
failure. This appears to be in contrast with most of the
previous studies that showed a quite high success rate in
old patients, but it is of note that the average baseline pH
of the studies by Benhamou et al.28 and Nava et al.29 was
7.28 and 7.30, respectively, and that the mean age was 79
years and 81 years. Hence, overall the subjects enrolled in
the present study were more acidotic and older.
The overall results obtained in our study may have
important clinical and organisational implications. The
implementation of a NIV service in a tertiary hospital
without an ICU may decrease the number of urgent
admissions in a large community hospital where bed
availability in the critical care area, at least in some countries, may be problematic. Quite often, ICU admission is
denied in old patients especially when they have a
chronic illness or several comorbidities.30 The implementation of NIV in a hospital with limited resources should
be carefully evaluated, depending on the geographical
position, the expected number of patients to be treated in
1 year, the possibility of having a ‘back-up ICU’ in case of
a patient’s deterioration, dedicated equipment and most
important, adequate training for the doctors and nurses
to bring together, as in our case, an enthusiastic team of
motivated people.
In our hospital, we periodically meet (every 3 months)
for an audit aimed to update knowledge and to discuss
the problems that occurred in that time frame. Nurses
and doctors participate in national and international
courses on NIV. Lastly, we have specific protocols for
monitoring patients’ vital signs and tolerance and to
avoid nasal or skin abrasions.
Our study presents important limitations. In this
study, we did not record the effective hours of ventilation during nighttime, but it has been recently shown
that the amount of time that NIV is effectively applied
in ‘every day clinical practice’ is greater overnight than
during the day, with air leaks, disconnections and
desaturations not different between day and night.31 A
relatively small number of patients was enrolled in a
single centre for a limited amount of time (1 year).
Indeed, despite the anaesthesiologist on duty who
should theoretically follow the internal guidelines for
intubation, it is well possible that other factors may
have influenced his/her decision, such as the patient’s
refusal and severity/prognosis of related diseases. Therefore, all these factors raise the possibility of a type 2
error. However, at least from a general point of view,
our study has the merit to represent a snapshot of the
‘every day clinical practice’ in a rural hospital, where
usually RCT are not performed.
Conclusion
Our pilot study confirms the hypothesis that NIV is a
feasible and effective treatment also for patients with
severe forms of acute exacerbations of COPD, admitted to
a general ward of a rural hospital without dedicated critical care beds. However, the data need to be confirmed in
larger studies and in different geographical locations.
© 2015 Royal Australasian College of Physicians
535
Fiorino et al.
Acknowledgements
We are indebted to our nursing staff: I. Alberio, M.
Baccarini, M. Benetti, A. Brcanija, L. Carini, G. Cassini, D.
Dani, M. Dudojc, S. Francavilla, E. Leonelli, T. Leonetti,
C. Paternuosto, B. Morelli, N. Menandro, P. Passerò, M.
R. Pesce, O. Riccio, M. Skrzynecka, C. Testagrossa, I.
Turturiello, M. Vetrone, F. Caruso.
References
1 Vincent JL, Sakr Y, Ranieri VM.
Epidemiology and outcome of acute
respiratory failure in intensive care unit
patients. Crit Care Med 2003; 31(Suppl):
S296–9.
2 Divo M, Cote C, de Torres JP, Casanova
C, Marin JM, Pinto-Plata V et al.
Co-morbidities and risk of mortality in
pts with chronic obstructive pulmonary
disease. Am J Resp Crit Care Med 2012;
186: 155–61.
3 Chatila WM, Thomashow BM, Minai
OA, Criner GJ, Make BJ. Comorbidities
in chronic obstructive pulmonary
disease. Proc Am Thorac Soc 2008; 5:
549–55.
4 Ambrosino N, Foglio K, Rubini F, Clini
E, Nava S, Vitacca M. Non-invasive
mechanical ventilation in acute
respiratory failure due to chronic
obstructive pulmonary disease:
correlates for success. Thorax 1995; 50:
755–7.
5 Confalonieri M, Garuti G, Cattaruzza
MS, Osborn JF, Antonelli M, Conti G
et al. A chart of failure risk for
noninvasive ventilation in patients with
COPD exacerbation. Eur Respir J 2005;
25: 348–55.
6 Balami JS, Packham SM, Gosney MA.
Non-invasive ventilation for respiratory
failure due to acute exacerbations of
chronic obstructive pulmonary disease
in older patients. Age Ageing 2006; 35:
75–9.
7 Fiorino S, Detotto E, Battilana M,
Bacchi-Reggiani L, Moretti R, Benfenati
F et al. Severe exacerbations of chronic
obstructive pulmonary disease:
management with noninvasive
ventilation on a general medicine ward.
Ital J Med 2010; 4: 173–8.
8 Plant PK, Owen JL, Elliott MW. Early
use of non-invasive ventilation for acute
9
10
11
12
13
14
15
The authors thank Dr Simonetta Righi, Biblioteca
Centralizzata, Policlinico S. Orsola-Malpighi, Università
di Bologna, Bologna, Italy for her support in the scientific
bibliography searches and Dr Laura Pontoriero, Unità di
Cura Materno-Infantile, Distretto Lametino, A. S. P.
Catanzaro, Italy for her assistance in the English editing
of our manuscript.
exacerbations of chronic obstructive
pulmonary disease on general
respiratory wards: a multicentre
randomised controlled trial. Lancet 2000;
355: 1931–5.
Moretti M, Cilione C, Tampieri A,
Fracchia C, Marchioni A, Nava S.
Incidence and causes of non-invasive
mechanical ventilation failure after
initial success. Thorax 2000; 55:
819–25.
Charlson ME, Pompei P, Ales KL,
MacKenzie CR. A new method of
classifying prognostic comorbidity in
longitudinal studies: development and
validation. J Chronic Dis 1987; 40:
373–83.
Miller MR, Hankinson J, Brusasco V,
Burgos F, Casaburi R, Coates A et al.
ATS/ERS Task Force. Standardisation of
spirometry. Eur Respir J 2005; 26:
319–38.
Groenewegen KH, Schols AM, Wouters
EF. Mortality and mortality-related
factors after hospitalization for acute
exacerbation of COPD. Chest 2003; 124:
459–67.
Bustamante-Fermosel A,
De Miguel-Yanes JM, Duffort-Falcó M,
Muñoz J. Mortality-related factors after
hospitalization for acute exacerbation of
chronic obstructive pulmonary disease:
the burden of clinical features. Am J
Emerg Med 2007; 25: 515–22.
Stefan MS, Shieh MS, Pekow PS,
Rothberg MB, Steingrub JS, Lagu T et al.
Epidemiology and outcomes of acute
respiratory failure in the United States,
2001 – 2009: a national survey. J Hosp
Med 2013; 8: 76–82.
International Consensus Conference
Committee. International Consensus
Conferences in Intensive Care Medicine:
noninvasive positive pressure ventilation
in acute respiratory failure. Am J Respir
Crit Care Med 2001; 163: 283–91.
16 British Thoracic Society Standards of
Care Committee. Non-invasive
ventilation in acute respiratory failure.
Thorax 2002; 57: 192–211.
17 Sulaiman MI, Rodger KA, Hawkins M. A
survey of the use of non invasive
positive pressure ventilation in critical
care units in the United Kingdom. Am J
Respir Crit Care Med 2004; 169: A522.
18 Roberts CM, Stone RA, Buckingham RJ,
Pursey NA, Lowe D, National Chronic
Obstructive Pulmonary Disease
Resources and Outcomes Project
Implementation Group. Acidosis,
non-invasive ventilation and mortality
in hospitalised COPD exacerbations.
Thorax 2011; 66: 43–8.
19 Paus-Jenssen ES, Reid JK, Cockcroft
DW, Laframboise K, Ward HA. The use
of noninvasive ventilation in acute
respiratory failure at a tertiary care
center. Chest 2004; 126: 165–72.
20 Cabrini L, Moizo E, Nicelli E, Licini G,
Turi S, Landoni G. Noninvasive
ventilation outside the intensive care
unit from the patient point of view: a
pilot study. Respir Care 2012; 57: 704–9.
21 Crummy F, Buchan C, Miller B, Toghill
J, Naughton MT. The use of noninvasive
mechanical ventilation in COPD with
severe hypercapnic acidosis. Respir Med
2007; 101: 53–61.
22 Squadrone E, Frigerio P, Fogliati C,
Gregoretti C, Conti G, Antonelli M et al.
Noninvasive vs invasive ventilation in
COPD patients with severe acute
respiratory failure deemed to require
ventilatory assistance. Intensive Care Med
2004; 30: 1303–10.
23 Lindenauer PK, Stefan MS, Shieh MS,
Pekow PS, Rothberg MB, Hill NS.
Outcomes associated with invasive and
noninvasive ventilation among patients
hospitalized with exacerbations of
chronic obstructive pulmonary disease.
JAMA Intern Med 2014; 174: 1982–93.
© 2015 Royal Australasian College of Physicians
536
COPD: non-invasive ventilation
24 Pacilli AM, Valentini I, Carbonara P,
Marchetti A, Nava S. Determinants of
noninvasive ventilation outcomes during
an episode of acute hypercapnic
respiratory failure in chronic obstructive
pulmonary disease: the effects of
comorbidities and causes of respiratory
failure. Biomed Res Int 2014; 2014:
976783.
25 Walkey AJ, Wiener RS. Use of
noninvasive ventilation in patients with
acute respiratory failure, 2000–2009: a
population-based study. Ann Am Thorac
Soc 2013; 10: 10–17.
26 Lemyze M, Mallat J, Nigeon O,
Barrailler S, Pepy F, Gasan G et al.
Rescue therapy by switching to total
face mask after failure of face
mask-delivered noninvasive ventilation
in do-not-intubate patients in acute
respiratory failure. Crit Care Med 2013;
41: 481–8.
27 Carrillo A, Ferrer M, Gonzalez-Diaz G,
Lopez-Martinez A, Llamas N, Alcazar M
et al. Noninvasive ventilation in acute
hypercapnic respiratory failure caused
by obesity hypoventilation syndrome
and chronic obstructive pulmonary
disease. Am J Respir Crit Care Med 2012;
186: 1279–85.
28 Benhamou D, Muir JF, Melen B.
Mechanical ventilation in elderly
patients. Monaldi Arch Chest Dis 1998; 53:
547–51.
29 Nava S, Grassi M, Fanfulla F,
Domenighetti G, Carlucci A, Perren A
et al. Non-invasive ventilation in elderly
patients with acute hypercapnic
respiratory failure: a randomised
controlled trial. Age Ageing 2011; 40:
444–50.
30 Evans TW, Nava S, Mata GV, Guidet B,
Estenssoro E, Fowler R et al. Critical care
rationing: international comparisons.
Chest 2011; 140: 1618–24.
31 Olivieri C, Carenzo L, Vignazia GL,
Campanini M, Pirisi M, Della Corte F
et al. Does noninvasive ventilation
delivery in the ward provide early
effective ventilation? Respir Care 2015;
60: 6–11.
Prevalence and significance of CYP2C19*2 and CYP2C19*17
alleles in a New Zealand acute coronary syndrome population
P. D. Larsen,1,2 L. R. Johnston,1,3 A. Holley,1,3 A. C. La Flamme,3 L. Smyth,4 E. W. Chua,4 M. A. Kennedy4 and
S. A. Harding1,5
1
Wellington Cardiovascular Research Group and 5Cardiology Department, Wellington Hospital, 2Department of Surgery and Anaesthesia, University of
Otago, 3School of Biological Sciences, Victoria University of Wellington, Wellington and 4Carney Centre for Pharmacogenomics, University of Otago,
Christchurch, New Zealand
Key words
platelet activation/drug effect, acute coronary
syndrome, ticlopidine/*analogue and
derivative, ticlopidine/therapeutic use,
ethnicity.
Correspondence
Peter D. Larsen, University of Otago, PO Box
7343, Wellington, New Zealand.
Email: [email protected]
Received 27 October 2014; accepted 4 January
2015.
doi:10.1111/imj.12698
Abstract
Background: High on-treatment platelet reactivity has been associated with poor
outcomes following acute coronary syndromes (ACS). Both the loss of function
CYP2C19*2 allele and the gain of function CYP2C19*17 allele along with a range of
clinical characteristics have been associated with variation in the response to clopidogrel.
Aim: The study aims to examine the frequency of CYP2C19 variants and understand
the factors associated with on-treatment platelet reactivity in a New Zealand ACS
population.
Methods: We prospectively enrolled 312 ACS patients. We collected clinical characteristics and measured on-treatment platelet reactivity using two validated point-of-care
assays, VerifyNow and Multiplate. DNA was extracted and CYP2C19*2 and *17 alleles
were identified using real-time polymerase chain reaction.
Results: CYP2C19*2 or CYP2C19*17 alleles were observed in 101 (32%) and 106 (34%)
of patients, respectively, with significant differences in distribution by ethnicity. In Maori
and Pacific Island patients, 47% (confidence interval (CI) 31–63%) had CYP2C19*2 and
11% (CI 4–19%) CYP2C19*17 compared with 26% (CI 19–32%) and 41% (CI 32–49%)
in white people. Carriage of CYP2C19*2 alleles was associated with higher levels of
platelet reactivity measured by either assay, but we observed no relationship between
platelet reactivity and CYP2C19*17. In multivariate analysis diabetes, clopidogrel dose
and CYP2C19*2 status were all significant independent predictors of platelet reactivity.
Conclusions: Both CYP2C19*2 and *17 were common in a New Zealand ACS population, with CYP2C19*2 observed in almost half the Maori and Pacific Island patients.
CYP2C19*2, diabetes and clopidogrel dose were independent contributors to
on-treatment platelet reactivity.
© 2015 Royal Australasian College of Physicians
537
Larsen et al.
Introduction
There is considerable variability in the level of platelet
inhibition observed in patients treated with clopidogrel
and aspirin following acute coronary syndromes
(ACS).1–3 Patients with high levels of platelet reactivity on
clopidogrel have an increased risk of death, myocardial
infarction and stent thrombosis.4,5 In a New Zealand
context, we have previously demonstrated that high
levels of platelet reactivity are common, particularly in
Maori and Pacific Island patients.2
Previous studies have suggested that the variability in
clopidogrel’s pharmacodynamics is multifactorial in
origin. Clinical variables, such as diabetes and smoking,6
drug interactions including those with proton pump
inhibitors,7 functional genetic polymorphisms, most
importantly those in the hepatic CYP2C19 enzyme,8 and
variation in the level of pretreatment platelet reactivity,9
have all been implicated in contributing to the variability
in the response to clopidogrel. CYP2C19 is one of the
enzymes involved in converting clopidogrel to its active
metabolite. The CYP2C19*2 allele is a loss of function
(LOF) variant associated with reduced blood levels of the
active metabolite, while the *17 allele is a gain of function
variant associated with higher levels of the active
metabolite.8,10 CYP2C19*2 has been associated with an
increased risk of adverse cardiovascular outcomes.8,10,11
Understanding the frequency of the CYP2C19 variants
in a New Zealand ACS population and how this relates
to on-treatment levels of platelet reactivity is important
in determining optimal antiplatelet therapy in this
population.
Methods
Patient population
Patients presenting to Wellington Regional Hospital with
ACS between January and December 2012 were eligible
for inclusion in the study if there was an invasive
approach (coronary angiography ± percutaneous coronary intervention (PCI)) planned. All participants were
adequately pretreated with aspirin and clopidogrel. To
ensure the necessary statistical power to assess the impact
of ethnicity, we recruited 282 ACS patients (232 white
people, 40 Maori and Pacific Islanders) and enriched the
population with an additional 30 Maori and Pacific Island
ACS patients. Exclusion criteria included a platelet count
Funding: This work was supported by research grants from the
Heart Foundation and from the Capital and Coast District Health
Board.
Conflict of interest: None.
less than 100 × 109/L, known platelet function disorder,
administration of a fibrinolytic agent within 24 h of
enrolment, use of a glycoprotein IIb/IIIa receptor antagonist within 7 days, or administration of an oral
antiplatelet agent other than aspirin or clopidogrel within
2 weeks of enrolment. The study was reviewed and
approved by the Lower Regional South Ethics Committee
(LRS/11/09/035). All patients provided written informed
consent.
Definitions
An ACS was defined as symptoms suggestive of myocardial ischaemia lasting >10 min and either troponin elevation or ≥1 mm of new ST segment deviation or T wave
inversion on an electrocardiogram in at least two contiguous leads.12 Adequate pretreatment was defined as
chronic therapy (≥7 days) with aspirin (≥75 mg) and
clopidogrel (≥75 mg), and/or loading with aspirin
≥300 mg at least 2 h and clopidogrel ≥300 mg at least 6 h
prior to enrolment. The clopidogrel dose was defined as
‘high’ if patients had received a 600 mg loading dose
followed by a 150 mg daily maintenance dosing. Intermediate dose was defined as either a 600-mg loading
dose followed by 75-mg daily maintenance dose, or a
300-mg loading dose coupled with 150 mg maintenance
dose. Low dose was defined as a 300-mg loading dose
followed by 75-mg daily maintenance dose or chronic
therapy with 75-mg daily of clopidogrel.
Platelet function testing
Blood samples for platelet function testing were taken
prior to angiography. On average, this was 4 ± 2 days
after hospital presentation with ACS. The level of
on-treatment platelet reactivity was quantified using the
VerifyNow P2Y12 assay (Accumetrics, San Diego, CA,
USA) and the Multiplate analyser (Dynabyte, Munich,
Germany), both of which have been shown to be predictive of clinical outcomes.4,5 The VerifyNow P2Y12 assay, a
turbidimetric-based optical detection system, was used
according to the manufacturer’s instructions. This device
uses fibrinogen-coated microbeads, an agonist of adenosine diphosphate (20 mM ADP), and light transmittance
through whole blood, to measure platelet agglutination.
An optical signal, reported as P2Y12 reaction units (PRU),
was recorded. The Multiplate analyser is a multiple electrode impedance aggregometer that assesses platelet
function in whole blood as previously described.13 Briefly,
whole blood was added to the test cuvettes, diluted (1:2
with 0.9% NaCl solution), stirred and warmed to 37°C.
ADP was added to a final concentration of 6.4 mM, and
aggregation was then continuously recorded for 6 min.
© 2015 Royal Australasian College of Physicians
538
CYP2C19*2 in acute coronary syndromes
Aggregation values are quantified as area under the
aggregation curve expressed as aggregation units ×
minutes (AU). All material used for platelet function
testing was obtained from the manufacturer (Dynabyte).
High on-treatment platelet reactivity (HPR) was
defined as >208 PRU for VerifyNow and >47AU for
Multiplate measurement, while low on-treatment platelet reactivity (LPR) was defined as <85PRU for VerifyNow
and <19AU for Multiplate measurement of platelet
reactivity.14
Genotyping
Genomic DNA was isolated from EDTA blood, followed
by identification of CYP2C19*2, and *17 using validated
real-time polymerase chain reaction (PCR) highresolution melting assays. The assay for CYP2C19*2 was
based on the method described by Temesvari et al.15 For
testing the presence of CYP2C19*17, a 62-base-pair PCR
product was amplified with 0.2 μM each of the primers
AAATTTGTGTCTTCTGTTCTCAAA and TGCCCATCGTG
GCGCATTAT,16 in a 10-μL reaction mixture containing 1×
reaction buffer B1, 1.5 mM Mg2+, 0.2 mM of each dNTP,
1.5 mM SYTO 9 Green Fluorescent Nucleic Acid Stain
(Life Technologies, Carlsbad, CA, USA), 0.5 U of HOT
FIREPol DNA Polymerase (Solis BioDyne, Tartu, Estonia),
and 50 ng of input DNA. Thermal cycling was preceded
by 95°C for 10 min, then 45 cycles of 95°C for 10 s, 52°C
for 15 s and 72°C for 15 s. High-resolution melting profile
was obtained by initial heating at 95°C for 15 s,
re-annealing at 55°C for 15 s, and final denaturation with
gradual heating to 95°C, during which fluorescence data
were collected every 0.1°C. CYP2C19*2 and *17
genotyping for most samples was performed twice.
Ambiguous genotype calls were resolved by Sanger
sequencing or restriction digest.
Statistical analysis
Data are presented as mean (SD) for continuous variables
that were normally distributed and as counts (%) for
categorical variables. Differences in platelet reactivity by
genotype were examined using analysis of variance. In
univariate analysis using student t test, analysis of variance and correlation, we identified the factors associated
with at least one of the platelet function assay measurements at P < 0.10. These variables were then examined
using multivariate linear regression analysis with calculation of the adjusted β coefficient and coefficient of
determination (R2) to identify the independent contribution of each to the inter-individual variability in
on-treatment platelet reactivity as measured with the
Multiplate assay and the VerifyNow assay.
Results
Of the 312 patients enrolled in the study, 58 (18%)
presented with ST-segment elevation myocardial infarction, 239 (77%) with non-ST segment elevation myocardial infarction and 15 (5%) with unstable angina. The
mean age of the population was 63 (±10) years old, and
225 (72%) were male. The majority (237, 76%) of the
population were white people, with 70 (22%) of the
patients identifying as Maori or of Pacific Island descent.
There were 101 patients (32%) with carriage of at least
one CYP2C19*2 allele, and this more likely in Maori and
Pacific Island patients than white people (47%, confidence interval (CI) 31–63% compared with 26% CI
19–32%). There were 106 patients (34%) with at least
one CYP2C19*17 allele. Maori and Pacific Island patients
were less likely than white people to have a CYP2C19*17
allele (11% CI 4–19% compared with 41% CI 32–49%).
We examined platelet reactivity by CYP2C19*2 status
(Fig. 1), and the presence of the CYP2C19*17 allele
(Fig. 2). The presence of the CYP2C19*2 allele was associated with significant differences in platelet reactivity
with both platelet function assays. VerifyNow levels of
platelet reactivity were 174 (93), 210 (81) and 224 (68)
PRU in the no CYP2C19*2 alleles, 1 allele and 2
CYP2C19*2 alleles groups respectively (P = 0.001).
Bonferroni’s multiple comparison showed significant differences between no and 1 CYP2C19*2 allele groups, but
not between the other pairs. Multiplate levels of platelet
reactivity were 39 (23), 43 (23) and 64 (41) AU in the no
CYP2C19*2 alleles, 1 allele and 2 CYP2C19*2 alleles groups
respectively (P = 0.0003). Bonferroni’s multiple comparison showed significant differences between 2 CYP2C19*2
alleles and both 1 allele and no CYP2C19*2 alleles, but not
between the 1 allele and no CYP2C19*2 alleles groups.
There were no significant differences in platelet reactivity
measured by either assay and the presence of the
CYP2C19*17 allele.
On the basis of VerifyNow measurement, 138 (44%)
had HPR, and this was associated with CYP2C19*2 allele
carriage, with an observed rate of 26% in no CYP2C19*2,
54% with 1 CYP2C19*2 allele and 70% with 2 CYP2C19*2
alleles (P = 0.01, chi-squared test). Multiplate measurements led to the classification of 116 (37%) of the
patients as having HPR, and this was also associated with
CYP2C19*2 allele carriage, with a rate of 32% in the no
CYP2C19*2 allele group, 44% with one allele and 58%
with two CYP2C19*2 alleles (P = 0.03, chi-squared test).
LPR was observed in 53 (17%) and 56 (18%) patients
assessed by VerifyNow and Multiplate criteria, respectively, and this did not differ by CYP2C19*17 allele status
with either assay (P = 0.95 and P = 0.69, respectively, for
VerifyNow and Multiplate assays, chi-squared test).
© 2015 Royal Australasian College of Physicians
539
Larsen et al.
Figure 1 Individual values (n = 312) for on-treatment platelet reactivity stratified by CYP2C19*2 allele carriage as measured with VerifyNow P2Y12 assay
on the left and Multiplate ADP assay on the right. Differences in platelet reactivity were measured using analysis of variance. Mean and standard deviation
for each group are represented by the bars.
We examined clinical characteristics of the groups by
CYP2C19*2 allele status (Table 1). There were significant
differences in ethnicity between the three groups, with
higher rates of Maori and Pacific Island patients in 1 allele
and 2 CYP2C19*2 alleles groups. There were also significantly more diabetic patients in the 2 CYP2C19*2 alleles
group (0.02). There were no other significant differences
between the three groups.
Figure 2 Individual values (n = 312) for on-treatment platelet reactivity stratified by CYP2C19*17 allele carriage as measured with VerifyNow P2Y12 assay
on the left and Multiplate ADP assay on the right. Differences in platelet reactivity were measured using analysis of variance. Mean and standard deviation
for each group are represented by the bars.
© 2015 Royal Australasian College of Physicians
540
CYP2C19*2 in acute coronary syndromes
Table 1 Patient characteristics stratified by CYP2C19*2 status
No CYP2C19*2 alleles n = 211
1 CYP2C19*2 allele n = 84
2 CYP2C19*2 alleles n = 17
P-value
62 (11)
158 (75)
30 (5)
62 (11)
58 (69)
30 (5)
62 (11)
9 (53)
30 (6)
0.66
0.11
0.82
170 (81)
37 (18)
4 (2)
55 (65)
25 (30)
4 (5)
7 (41)
8 (47)
2 (12)
0.001
146 (69)
144 (68)
52 (25)
42 (20)
54 (65)
52 (62)
18 (21)
14 (17)
13 (77)
13 (77)
9 (53)
4 (23)
0.54
0.40
0.02
0.73
66 (31)
37 (18)
13 (6)
24 (29)
13 (15)
6 (7)
3 (18)
2 (12)
2 (12)
0.47
0.76
0.67
67 (32)
72 (34)
90 (42)
53 (25)
32 (15)
10 (4)
56 (26)
30 (36)
26 (31)
38 (45)
15 (18)
16 (19)
2 (2)
18 (21)
8 (47)
5 (30)
7 (41)
2 (12)
1 (6)
0 (0)
5 (30)
0.39
0.82
0.97
0.22
0.49
0.44
0.61
32 (15)
170 (81)
9 (4)
23 (27)
57 (68)
4 (5)
3 (17)
12 (70)
2 (12)
0.09
105 (81)
225 (69)
93 (84)
224 (54)
108 (90)
263 (82)
0.83
0.08
96 (45)
53 (25)
62 (30)
48 (57)
12 (14)
24 (29)
11 (65)
4 (23)
2 (12)
0.11
Age (years)
Male, n (%)
BMI
Ethnicity
European
Maori/Pacific Island
Other
Risk factors
Hypertension
Dyslipidaemia
Diabetes
Current smoker
Medical history
Previous MI
Previous PCI
Previous CABG
Admission medications
ACE inhibitor
Beta-blocker
Statin
Ca channel antagonist
Diuretic
SSRI
PPI
Clinical presentation
STEMI
NSTEMI
Unstable angina
Laboratory data
Creatinine (μmol/L)
Platelet count (109/L)
Clopidogrel dose
Low dose
Intermediate dose
High dose
ACE, angiotensin-converting enzyme; BMI, body mass index; CABG, coronary artery bypass graft; MI, myocardial infarction; NSTEMI, non-ST segment
elevation myocardial infarction; PCI, percutaneous coronary intervention; PPI, proton pump inhibitor; SSRI, selective serotonin reuptake inhibitor; STEMI,
ST-segment elevation myocardial infarction.
There were 14 variables that had a univariate relationship with either Multiplate or VerifyNow measured platelet reactivity at P < 0.10, and these were included in the
multivariate analysis shown in Table 2. CYP2C19*2 status,
platelet count, diabetes and clopidogrel dose were all
significant independent predictors of Multiplate measured platelet reactivity, while genotype, male gender,
diabetes and clopidogrel dose were predictors of
VerifyNow measured platelet reactivity. The proportion
of platelet reactivity predicted on the basis of CYP2C19*2
alone was low for both assays (Table 3), but improved the
overall predictive value above the use of clinical variables
alone for each assay.
Discussion
Both CYP2C19*2 and *17 had high prevalence in a New
Zealand ACS population. The frequency of these
polymorphisms differed significantly in Maori and Pacific
Island patients when compared with white people, with
higher CYP219*2 and lower CYP2C19*17 frequency in
Maori and Pacific Island patients. CYP2C19*2 alleles had a
modest impact on platelet reactivity, with diabetes and
clopidogrel dose having a greater influence. Clinical
factors and genotype combined only explained 20–21%
of the observed variance in on-treatment platelet
reactivity.
The observed frequency of CYP2C19*2 and *17 alleles
in this study were 32% and 34% respectively. The percentage of patients with CYP2C19*2 and *17 alleles in the
white people subpopulation, 26% and 41% respectively,
was within the range of previous studies, where 10–28%
with CYP2C19*27,8,17 and 21–41% with CYP2C19*178,18,19
have been described. The proportion of Maori and Pacific
Island patients with CYP2C19*2 was 47%. The only previous study to examine the incidence of CYP2C19*2 in
© 2015 Royal Australasian College of Physicians
541
Larsen et al.
Table 2 Contribution to on-treatment platelet reactivity
Multiplate ADP assay
CYP2C19*2 alleles
BMI
Male gender
Ethnicity
Hypertension
Diabetes
Creatinine
Dyslipidemia
Platelet count
Clopidogrel dose
ACE inhibitor
Beta-blocker
Diuretic
Statin
PPI
VerifyNow P2Y12 assay
βadj (95% CI)
P-value
βadj (95% CI)
P-value
7.1 (2.6 to 11.6)
0.08 (−0.4 to 0.6)
2.8 (−3.3 to 9.5)
−2.2 (−8.5 to 4.0)
−5.0 (−11.8 to 1.7)
6.5 (0.1 to 13.7)
−0.1 (−0.3 to 0.1)
2.2 (−4.4 to 8.9)
0.1 (0.07 to 0.15)
−3.6 (−6.9 to −0.5)
5.0 (−1.0 to 11.3)
1.9 (−4.2 to 8.0)
0.5 (−6.4 to 7.5)
5.1 (−1.3 to 11.5)
0.4 (−6.0 to 6.9)
0.002
0.75
0.36
0.47
0.15
0.05
0.57
0.51
0.001
0.02
0.10
0.54
0.87
0.12
0.90
25.9 (9.7 to 42.2)
1.3 (−0.5 to 3.2)
−26.3 (−48.5 to −4.0)
−4.1 (−26.5 to 18.3)
1.3 (−22.8 to 25.0)
40.9 (16.9 to 64.9)
−0.04 (−0.1 to 0.03)
15.1 (−8.9 to 39.2)
−0.1 (−0.2 to 0.13)
−15.4 (−26.9 to −3.8)
10.6 (−11.3 to 32.6)
11.8 (−10.1 to 33.7)
−8.8 (−33.6 to 16.1)
2.0 (−21.0 to 25.0)
17.8 (−5.4 to 41.0)
0.002
0.16
0.02
0.71
0.95
0.001
0.26
0.21
0.84
0.009
0.34
0.29
0.49
0.87
0.13
Adjusted B coefficients with 95% CI for on-treatment platelet reactivity in a multivariate linear regression model. BMI, body mass index; CI, confidence
interval; PPI, proton pump inhibitor.
Maori reported results in terms of allele frequency rather
than proportion of patients carrying the allele. Using this
measure, Lea et al. reported a frequency of 24%,20 and
our calculated frequency was similar at 29%. This is the
first study that we are aware of to examine the prevalence of CYP2C19*17 in Maori and Pacific Island patients,
where only 11% demonstrated this allele.
Several previous studies have demonstrated striking
differences in the proportion of patients with CYP2C19
variants.8 Within a Japanese cohort CYP2C19*2 was
observed in more than 57% of patients.3 A Korean study
reported 44% of patients had CYP2C19*2.21 This level of
variance between populations in a genotype known to
affect responsiveness to a commonly used drug and to be
associated with poor clinical outcomes highlights the
importance of examining clinically relevant genotypes in
a New Zealand population.
Table 3 Percentage of the variability in on-treatment platelet reactivity
explained
Multiplate
ADP assay
CYP2C19*2 alleles
Clinical variables†
LOF alleles and clinical variables
VerifyNow
P2Y12 assay
R2 (%)
P-value
R2 (%)
P-value
3
18
20
0.001
0.0001
0.0001
4
18
21
0.0001
0.0001
0.0001
The percentage of the variability in on-treatment platelet reactivity
explained was defined as the coefficient of determination (R2). †Clinical
variables included were the 14 variables listed in Table 2, all of which had
univariate associations with platelet reactivity with at least one of the
assays at P ≤ 0.10. LOF, loss of function.
The high prevalence of diabetes in Maori and Pacific
Island patients2 contributed to the high rate of diabetes in
the 2 CYP2C19*2 alleles group. Our previous observation
that on-treatment platelet reactivity was higher in Maori
and Pacific Island patients2 did not include genetic analysis, and so did not allow us to examine CYP2C19*2 alleles
and diabetes as covariates along with ethnicity. All three
were univariate predictors of platelet reactivity in the
current study. However, in the multivariate analysis, only
diabetes and CYP2C19*2 alleles but not ethnicity were
independent contributors to platelet reactivity, suggesting
that the ethnic differences are largely driven by these
factors.
The observed relationship between diabetes and platelet reactivity is consistent with previous reports.6,7,22 Similarly, lower doses of clopidogrel leading to lower levels of
active metabolite have been previously described as a
significant contributor to on-treatment platelet reactivity,
as observed in our cohort.7
The two different platelet function assays produced
slightly different models accounting for platelet reactivity.
Platelet count was associated with platelet reactivity
measured with the Multiplate system. Previous studies
have reported a relationship between platelet count and
platelet reactivity.7 The Multiplate assay is known to be
sensitive to platelet count,23 but whether this reflects a
more accurate assessment of how platelets behave in
blood or is an artefact of measurement is unclear. Male
gender has also been previously associated with higher
levels of platelet reactivity,7 consistent with the results
from the VerifyNow assay in our study.
© 2015 Royal Australasian College of Physicians
542
CYP2C19*2 in acute coronary syndromes
A large meta-analysis examining CYP2C19*2 allele carriage reported a significant increased risk of the composite end–point of cardiovascular death, myocardial
infarction and stroke associated with this allele.11 Several
studies have similarly reported a relationship between
HPR and death, myocardial infarction, and stent thrombosis.1,5,24 The findings of these studies raise the possibility that the high prevalence of CYP2C19*2 in Maori and
Pacific Island patients observed here, and the previously
described high rate of HPR in Maori and Pacific Island
patients,2 may contribute to the poor outcomes observed
in this patient group.
We did not observe a relationship between CYP2C19*17
and platelet reactivity in the current study, despite a high
prevalence of this allele. Larger studies than ours have
reported a lower rate of platelet reactivity in patients with
this genotype and an associated increased bleeding
risk,19,25 suggesting that our failure to observe any relationship here may be due to a lack of statistical power.
However, there are large studies that have failed to demonstrate a relationship between CYP2C19*17 and bleeding
risk.26
Alternative antiplatelet therapies are now available.
Ticagrelor and prasugrel are more potent P2Y12 receptor
antagonists, both of which have been associated with
improved clinical outcomes in ACS patients.27,28 Switching from clopidogrel to prasugrel2 or ticagrelor29 effectively reduces the rate of HPR, while simply increasing
the dose of clopidogrel appears to be a less effective strategy.30 In substudies from both PLATO31 and TRITON-TIMI
38,32 the adverse event rate was significantly higher in
the clopidogrel arm of the trials in subgroups with
CYP2C19*2 alleles. While a significant benefit remained
for ticagrelor compared with clopidogrel in those without
LOF alleles,31 there was no difference in the event rate
between those without LOF alleles treated with
clopidogrel compared with those treated with prasugrel.32
These studies raise the possibility that at least a portion of
the benefits associated with prasugrel and ticagrelor
could be achieved through a tailored strategy based on
genotype.
Given that clinical characteristics, medications and
genotype combined in this study to predict only 20% of
the measured platelet reactivity, it would appear that
there is considerable additional information that can be
gained from platelet function testing. However, it is not
currently clear how this information should best be used.
Tailored therapy driven by platelet function testing has
not been successful to date. The three largest randomised
trials using platelet function testing to tailor antiplatelet
therapy, the GRAVITAS trial,33 the ARCTIC study34 and
the TRIGGER-PCI trial,35 all failed to demonstrate any
benefit from this approach. In the GRAVITAS study,
patients with high levels of platelet function were given
higher dose of clopidogrel33 rather than a more potent
P2Y12 receptor antagonist, and this strategy has limited
effectiveness. This was also the case for the majority of
the ARCTIC study, with only 3.3% of patients switched to
prasugrel.34 The TRIGGER-PCI trial was halted early due
to a lower than expected event rate, suggesting that it
was not going to be adequately powered.35 A relatively
small (n = 600) randomised controlled trial that personalised antiplatelet therapy on the basis of CYP2C19 genotype demonstrated that this approach reduced MACE at 6
months in a Chinese population.36 This suggests that
using CYP2C19 status rather than phenotype to direct
therapy may have merit.
In the New Zealand environment, currently both
clopidogrel and ticagrelor are available and being used to
treat patients with ACS. Given that nearly half the Maori
and Pacific Island patients in this study had a CYP2C19*17
allele and we previously observed just over half had
HPR,2 it is reasonable to suggest that Maori and Pacific
Island patients with ACS are likely to have an increased
benefit from the use of ticagrelor and should be preferentially treated with this agent.
There are several limitations to this study. The sample
size of 312 patients was relatively small, and as a consequence our power to observe significant contributors
within a multivariate model was limited. This is seen in
the reasonably large CI around the βadj coefficients in
Table 2. Platelet function testing was performed prior to
angiography. The time between symptom onset, presentation and loading with antiplatelet agents will have
varied in the population, and we cannot exclude the
possibility that this contributes to the variance in
on-treatment platelet reactivity observed. We have not
measured platelet reactivity prior to treatment, and so
cannot examine how this relates to on-treatment variability. Other clinical variables that may contribute to
platelet reactivity, such as left ventricular function, were
not routinely measured. We have combined Maori and
Pacific Island patients into a single ethnic group within
this study, and this may be inappropriate as there may be
significant differences between Maori and Pacific Island
patients in terms of the frequency of CYP2C19 alleles.
Conclusion
Both CYP2C19*2 and *17 had high prevalence in a New
Zealand ACS population, and CYP2C19*2 was observed in
almost half the Maori and Pacific Island patients. While
CYP2C19*2 demonstrated a modest contribution to the
observed levels of platelet reactivity, we did not detect a
relationship between CYP2C19*17 and platelet reactivity.
Diabetes and clopidogrel dose were the most significant
© 2015 Royal Australasian College of Physicians
543
Larsen et al.
contributors to on-treatment platelet reactivity, but 80%
of the observed variance in platelet reactivity was not
accounted for.
References
1 Geisler T, Schaeffeler E, Dippon J,
Winter S, Buse V, Bischofs C et al.
CYP2C19 and nongenetic factors predict
poor responsiveness to clopidogrel
loading dose after coronary stent
implantation. Pharmacogenomics 2008; 9:
1251–9.
2 Johnston LR, Larsen PD, La Flamme AC,
Michel JM, Simmonds MB, Harding SA.
Suboptimal response to clopidogrel and
the effect of prasugrel in acute coronary
syndromes. Int J Cardiol 2013; 167:
995–9.
3 Nagashima Z, Tsukahara K, Morita S,
Endo T, Sugano T, Hibi K et al. Platelet
reactivity in the early and late phases of
acute coronary syndromes according to
cytochrome P450 2C19 phenotypes.
J Cardiol 2013; 62: 158–64.
4 El Ghannudi S, Ohlmann P, Meyer N,
Wiesel ML, Radulescu B, Chauvin M
et al. Impact of P2Y12 inhibition by
clopidogrel on cardiovascular mortality
in unselected patients treated by
percutaneous coronary angioplasty: a
prospective registry. JACC Cardiovasc
Interv 2010; 3: 648–56.
5 Sibbing D, Braun S, Morath T, Mehilli J,
Vogt W, Schˆmig A et al. Platelet
reactivity after clopidogrel treatment
assessed with point-of-care analysis and
early drug-eluting stent thrombosis.
J Am Coll Cardiol 2009; 53:
849–56.
6 Morel O, Kessler L, Ohlmann P, Bareiss
P. Diabetes and the platelet: toward new
therapeutic paradigms for diabetic
atherothrombosis. Atherosclerosis 2010;
212: 367–76.
7 Bouman HJ, Harmsze AM, van Werkum
JW, Breet NJ, Bergmeijer TO, Ten Cate
H et al. Variability in on-treatment
platelet reactivity explained by
CYP2C19*2 genotype is modest in
clopidogrel pretreated patients
undergoing coronary stenting. Heart
2011; 97: 1239–44.
8 Scott SA, Sangkuhl K, Gardner EE, Stein
CM, Hulot JS, Johnson JA et al. Clinical
pharmacogenetics implementation
consortium guidelines for cytochrome
P450-2C19 (CYP2C19) genotype and
9
10
11
12
13
14
15
16
clopidogrel therapy. Clin Pharmacol Ther
2011; 90: 328–32.
Gurbel PA, Bliden KP, Hiatt BL,
O’Connor CM. Clopidogrel for coronary
stenting: response variability, drug
resistance, and the effect of pretreatment
platelet reactivity. Circulation 2003; 107:
2908–13.
Scott SA, Sangkuhl K, Stein CM, Hulot
JS, Mega JL, Roden DM et al. Clinical
Pharmacogenetics Implementation
Consortium guidelines for CYP2C19
genotype and clopidogrel therapy: 2013
update. Clin Pharmacol Ther 2013; 94:
317–23.
Mega JL, Simon T, Collet JP, Anderson
JL, Antman EM, Bliden K et al.
Reduced-function CYP2C19 genotype
and risk of adverse clinical outcomes
among patients treated with clopidogrel
predominantly for PCI a meta-analysis.
JAMA 2010; 304: 1821–30.
Thygesen K, Alpert JS, Jaffe AS,
Simoons ML, Chaitman BR, White HD
et al. Third universal definition of
myocardial infarction. Circulation 2012;
126: 2020–35.
Johnston LR, Larsen PD, La Flamme AC,
Harding SA. Methodological
considerations for the assessment of
ADP induced platelet aggregation using
the Multiplate(R) analyser. Platelets
2013; 24: 303–7.
Tantry US, Bonello L, Aradi D, Price MJ,
Jeong YH, Angiolillo DJ et al. Consensus
and update on the definition of
on-treatment platelet reactivity to
adenosine diphosphate associated with
ischemia and bleeding. J Am Coll Cardiol
2013; 62: 2261–73.
Temesvari M, Paulik J, Kobori L,
Monostory K. High-resolution melting
curve analysis to establish CYP2C19 *2
single nucleotide polymorphism:
comparison with hydrolysis SNP
analysis. Mol Cell Probes 2011; 25:
130–3.
Santos PC, Soares RA, Santos DB,
Nascimento RM, Coelho GL, Nicolau JC
et al. CYP2C19 and ABCB1 gene
polymorphisms are differently
distributed according to ethnicity in the
Brazilian general population. BMC Med
Genet 2011; 12: 13.
17 Brackbill ML, Kidd RS, Abdoo AD,
Warner JG, Harralson AF. Frequency of
CYP3A4, CYP3A5, CYP2C9, and
CYP2C19 variant alleles in patients
receiving clopidogrel that experience
repeat acute coronary syndrome. Heart
Vessels 2009; 24: 73–8.
18 Cuisset T, Loosveld M, Morange PE,
Quilici J, Moro PJ, Saut N et al.
CYP2C19*2 and *17 alleles have a
significant impact on platelet response
and bleeding risk in patients treated
with prasugrel after acute coronary
syndrome. JACC Cardiovasc Interv 2012;
5: 1280–7.
19 Sibbing D, Koch W, Gebhard D,
Schuster T, Braun S, Stegherr J et al.
Cytochrome 2C19*17 allelic variant,
platelet aggregation, bleeding events,
and stent thrombosis in
clopidogrel-treated patients with
coronary stent placement. Circulation
2010; 121: 512–18.
20 Lea RA, Roberts RL, Green MR,
Kennedy MA, Chambers GK. Allele
frequency differences of cytochrome
P450 polymorphisms in a sample of
New Zealand Maori. N Z Med J 2008;
121: 33–7.
21 Jeong YH, Tantry US, Kim IS, Koh JS,
Kwon TJ, Park Y et al. Effect of
CYP2C19*2 and *3 loss-of-function
alleles on platelet reactivity and adverse
clinical events in East Asian acute
myocardial infarction survivors treated
with clopidogrel and aspirin. Circ
Cardiovasc Interv 2011; 4: 585–94.
22 Siller-Matula JM, Lang IM, Neunteufl T,
Kozinski M, Maurer G, Linkowska K
et al. Interplay between genetic and
clinical variables affecting platelet
reactivity and cardiac adverse events in
patients undergoing percutaneous
coronary intervention. PLoS ONE 2014;
9: e102701.
23 Hanke AA, Roberg K, Monaca E,
Sellmann T, Weber CF, Rahe-Meyer N
et al. Impact of platelet count on results
obtained from multiple electrode platelet
aggregometry (MultiplateTM). Eur J Med
Res 2010; 15: 214–19.
24 Price MJ, Endemann S, Gollapudi RR,
Valencia R, Stinis CT, Levisay JP et al.
Prognostic significance of
© 2015 Royal Australasian College of Physicians
544
CYP2C19*2 in acute coronary syndromes
25
26
27
28
29
post-clopidogrel platelet reactivity
assessed by a point-of-care assay on
thrombotic events after drug-eluting
stent implantation. Eur Heart J 2008; 29:
992–1000.
Harmsze AM, van Werkum JW,
Hackeng CM, Ruven HJT, Kelder JC,
Bouman HJ et al. The influence of
CYP2C19*2 and*17 on on-treatment
platelet reactivity and bleeding events in
patients undergoing elective coronary
stenting. Pharmacogenet Genom 2012; 22:
169–75.
Simon T, Verstuyft C, Mary-Krause M,
Quteineh L, Drouet E, Meneveau N et al.
Genetic determinants of response to
clopidogrel and cardiovascular events. N
Engl J Med 2009; 360: 363–75.
Wallentin L, Becker RC, Budaj A,
Cannon CP, Emanuelsson H, Held C
et al. Ticagrelor versus clopidogrel in
patients with acute coronary syndromes.
N Engl J Med 2009; 361: 1045–57.
Wiviott SD, Braunwald E, McCabe CH,
Montalescot G, Ruzyllo W, Gottlieb S
et al. Prasugrel versus clopidogrel in
patients with acute coronary syndromes.
N Engl J Med 2007; 357: 2001–15.
Alexopoulos D, Galati A, Xanthopoulou
I, Mavronasiou E, Kassimis G,
Theodoropoulos KC et al. Ticagrelor
versus prasugrel in acute coronary
syndrome patients with high
on-clopidogrel platelet reactivity
following percutaneous coronary
intervention: a pharmacodynamic
study. J Am Coll Cardiol 2012; 60:
193–9.
30 Sardella G, Calcagno S, Mancone M,
Palmirotta R, Lucisano L, Canali E et al.
Pharmacodynamic effect of switching
therapy in patients with high
on-treatment platelet reactivity and
genotype variation with high clopidogrel
Dose versus prasugrel: the RESET GENE
trial. Circ Cardiovasc Interv 2012; 5:
698–704.
31 Wallentin L, James S, Storey RF,
Armstrong M, Barratt BJ, Horrow J et al.
Effect of CYP2C19 and ABCB1 single
nucleotide polymorphisms on outcomes
of treatment with ticagrelor versus
clopidogrel for acute coronary
syndromes: a genetic substudy of the
PLATO trial. Lancet 2010; 376:
1320–8.
32 Mega JL, Close SL, Wiviott SD, Shen L,
Walker JR, Simon T et al. Genetic
variants in ABCB1 and CYP2C19 and
cardiovascular outcomes after treatment
with clopidogrel and prasugrel in
the TRITON-TIMI 38 trial: a
pharmacogenetic analysis. Lancet 2010;
376: 1312–19.
33 Price MJ, Berger PB, Teirstein PS,
Tanguay JF, Angiolillo DJ, Spriggs D
et al.. Standard- vs high-dose clopidogrel
based on platelet function testing after
percutaneous coronary intervention: the
GRAVITAS randomized trial. JAMA
2011; 305: 1097–105.
34 Collet JP, Cuisset T, Range G, Cayla G,
Elhadad S, Pouillot C et al. Bedside
monitoring to adjust antiplatelet therapy
for coronary stenting. N Engl J Med
2012; 367: 2100–9.
35 Trenk D, Stone GW, Gawaz M, Kastrati
A, Angiolillo DJ, Muller U et al. A
randomized trial of prasugrel versus
clopidogrel in patients with high platelet
reactivity on clopidogrel after elective
percutaneous coronary intervention
with implantation of drug-eluting stents
results of the TRIGGER-PCI (testing
platelet reactivity in patients undergoing
elective stent placement on clopidogrel
to guide alternative therapy with
prasugrel) study. J Am Coll Cardiol 2012;
59: 2159–64.
36 Xie X, Ma YT, Yang YN, Li XM, Zheng
YY, Ma X et al. Personalized antiplatelet
therapy according to CYP2C19 genotype
after percutaneous coronary
intervention: a randomized control trial.
Int J Cardiol 2013; 168: 3736–40.
© 2015 Royal Australasian College of Physicians
545
Safety of coadministration of ezetimibe and statins in patients
with hypercholesterolaemia: a meta-analysis
L. Luo, X. Yuan, W. Huang, F. Ren, H. Zhu, Y. Zheng and L. Tang
Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
Key words
ezetimibe, statin, hypercholesterolaemia,
adverse event, meta-analysis.
Correspondence
Lin Tang, Department of Nephrology, The
Second Affiliated Hospital of Chongqing
Medical University, Chongqing 400010, China.
Email: [email protected]
Received 20 October 2014; accepted 19
December 2014.
doi:10.1111/imj.12706
Abstract
Background: Hypercholesterolaemia is a pivotal risk factor for cardiovascular
and cerebrovascular disease and is treated with many effective lipid-lowering agents.
Statins are often used alone or in combination with ezetimibe. Combination
therapy is more effective because of its complementary approach, which has major
benefits for patients with unmanageable lipid levels. Extensive application of combination therapy has resulted in an increased incidence of side-effects, which has raised our
concern.
Aim: To evaluate the evidence associated with the safety of coadministration of
ezetimibe with statins.
Methods: Three electronic databases were searched (PubMed, EMBASE and
Cochrane Library) from January 2002 to October 2014. Two independent reviewers
critically identify randomised controlled trials (RCT), extracted the data and assessed
trial quality. A total ot 20 RCT met inclusion criteria, including 14 856 patients. A
fixed-effects model was used for meta-analysis to assess the safety of combination
therapy.
Results: Coadministration of ezetimibe and statins did not result in significant
increases in total adverse events (30% vs 29%, P = 0.34), serious adverse events (2% vs
1.6%, P = 0.81), treatment discontinuations (3.5% vs 2.9%, P = 0.22), gastrointestinal
adverse events (5% vs 4%, P = 0.08), allergic reactions or rashes (0.9% vs 1.3%, P =
0.33), creatine kinase > 10 × upper limit of normal (ULN) (0.2% vs 0.2%, P = 0.86),
alanine aminotransferase > 3 × ULN (0.5% vs 0.4%, P = 0.96) and aspartate
aminotransferase > 3 × ULN (0.4% vs 0.4%, P = 0.58).
Conclusion: The incidence of adverse events was similar between ezetimibe–statin
combination therapy and statin monotherapy; thus, we recommend combination
therapy for patients with hypercholesterolaemia at high risk for cardiovascular and
cerebrovascular disease.
Introduction
Statins are inhibitors of hydroxymethylglutaryl-CoA
(HMG-CoA) reductase, which lowers cholesterol
through inhibiting the sterol biosynthetic pathway and
are first-line lipid-altering medications.1 Ezetimibe is a
cholesterol-absorption inhibitor that potently and selectively reduces intestinal absorption of dietary and biliary
cholesterol, which is an option for patients with hypercholesterolaemia.2 The complementary mechanisms of
action of statins and ezetimibe have greater lipidaltering efficacy than either agent alone, which is an
Funding: This work was supported by a grant from the National
Natural Science Foundation of China (No. 81200519; L. Tang).
Conflict of interest: None.
option for patients at high cardiovascular and cerebrovascular risk whose lipid levels do not reach the recommended standard of the National Cholesterol Education
Program Adult Treatment Panel (NCEP ATP) III.3
However, in addition to lipid-modifying efficacy, the
safety of coadministration of ezetimibe with statins has
been a major concern.
This meta-analysis aimed to examine critically the evidence for safety by analysing trials comparing ezetimibe–
statin combination therapy with statin monotherapy.
Methods
We searched PubMed, EMBASE and Cochrane Library
databases from January 2002 to October 2014, using the
following terms ‘ezetimibe’, ‘zetia’, ‘ezetrol’, ‘statin’,
© 2015 Royal Australasian College of Physicians
546
Safety of ezetimibe and statins
‘simvastatin’, ‘atorvastatin’, ‘rosuvastatin’, ‘lovastatin’,
‘pravastatin’, ‘cerivastatin’, ‘fluvastatin’, ‘hyperlipidaemia’, ‘dyslipidaemia’ and ‘hypercholesterolaemia’. The
language was limited to English.
Studies were included if they met the following criteria: (i) double-blind RCT; (ii) patients >18 years of age
diagnosed with hypercholesterolaemia, whose lowdensity lipoprotein cholesterol (LDL-C) levels were above
NCEP ATP III guidelines;4 (iii) comparison of safety of
coadministation of ezetimibe and statins versus statins
monotherapy; and (iv) treatment duration >4 weeks.
Any disagreement about the inclusion of an article was
resolved by discussion.
Identified articles were screened to ensure that they
met the predetermined inclusion criteria. Titles were
reviewed initially, followed by abstracts and then full-text
publications. Detailed information of whole articles was
acquired by two reviewers independently. The detailed
data were extracted as follows: study characteristics (first
author’s name, publication year, number of participants),
invention and control measures (type and dosage of
active drug, duration of follow up), individual characteristics (number of patients with hypertension or diabetes
mellitus) and outcome indicators (numbers of serious
adverse events, treatment discontinuations, allergic reactions or rashes, patients with alanine aminotransferase
(ALT) >3 × upper limit of normal (ULN), patients with
aspartate aminotransferase (AST) >3 × ULN, gastrointestinal adverse events and patients with creatine kinase
(CK) >10 × ULN.
We put the Cochrane Risk of Bias tool into effect,
including assessing random sequence generation;
allocation concealment; participant, personnel and
outcome assessor blinding; incomplete outcome data;
selective outcome reporting; and other bias. In
addition, risk of bias was not used as an exclusion criterion for individual studies in the meta-analysis. The
risk of bias was demonstrated graphically: blank means
not clear; red means high risk, and green means low
risk; that is, the greener the graph, the higher quality
the trials.
For evaluation of safety, we analysed the total
number of adverse events, and the numbers of serious
adverse events, treatment discontinuations, allergic
reactions or rashes, patients with ALT >3 × ULN,
patients with AST >3 × ULN, gastrointestinal adverse
events, and patients with CK >10 × ULN in each group,
and compared the values between the two groups. The
statistical analysis was performed by Software Review
Manager 5.2 (Cochrane Collaboration, Oxford, UK). To
assess heterogeneity for RCT, χ2 test and its results, P
value and I2 statistics were analysed to assess the incidence of adverse events. Fixed-effects models were used
for the meta-analysis. The P values were two tailed, and
statistical significance was set at P = 0.05.
Results
A flow diagram of the selection process for the metaanalysis is shown in Figure 1. A total of 568 studies was
hit initially, and 49 articles were retrieved for evaluation
at length; and finally, 20 RCT that satisfied the inclusion
criteria were analysed. The characteristics of these 20
studies are listed in Table 1.5–24 The quality of the
reporting is shown in Figure 2; 13 trials had complete
information for assessing randomisation, and five trials
had adequate blinding. Results of the quality assessment
for individual studies are shown in Figure 3.
Total adverse events were reported in 16 studies, with
1165 events occurring in 3856 patients (30%) treated
with ezetimibe and statins, compared with 1198 events in
4171 patients (29%) treated with statins alone. There
was no significant difference in the two arms (95% confidence interval (CI), 0.85–1.06; P = 0.34; I2 = 0%), as
shown in Figure 4. In the subgroup analysis of ezetimibe
with atorvastatin, incidence of total adverse events in the
combination therapy and statin monotherapy arms was
568 citations reviewed
209 excluded for duplication
359 potentially relevant studies
149 excluded for title
210 abstracts reviewed
164 excluded based on
abstracts
46 full-text articles reviewed
20 randomised controlled trials
included in the meta-analysis
3 additional trials
29 excluded after full article review
2 not relevant
3 with other medications agents
2 studies on the same population
2 with unavailable extracted data
3 cross-over tails
5 post-hoc analysis
2 pooled analysis
2 extension studies
3 without NCEP ATPIII
5 no full text
Figure 1 Flow diagram of screening process for studies included in the
meta-analysis. NCEP ATP III, National Cholesterol Education Program
Adult Treatment Panel III.
© 2015 Royal Australasian College of Physicians
547
Luo et al.
Table 1 Characteristics of included studies.
Study
Participants
Duration
Intervention
E+S
Ballantyne et al. 20035
Melani et al. 20037
Kerzner et al. 20036
Feldman et al. 20049
Bays et al. 20048
Goldberg et al. 200410
Cruz-Fernandez et al. 200513
Barrios et al. 200512
Ballantyne et al. 200511
Farnier et al. 200514
Catapano et al. 200615
Zubaid et al. 200818
Conard et al. 200816
Leiter et al. 200817
Robinson et al. 200920
Farnier et al. 200919
Foody et al. 201022
Averna et al. 201021
Bays et al. 201123
Hing Ling et al. 201224
PHC
PHC
PHC
HC
PHC
PHC
HC, CHD
PHC, CHD or AS
HC
HC, CHD
HC
PHC, CAD
HC
HC
MS, HC
HC
HC
HC, CHD
HC
PHC
12 w
12 w
12 w
6w
12 w
12 w
6w
6w
6w
6w
6w
6w
6w
6w
6w
6w
12 w
6w
6w
6w
Hypertension
S
DM
E+S
S
E+S
S
Dose (mg)
num
Dose (mg)
num
num
num
num
num
E10+A10,20,40,80
E10+P10,20,40
E10+L10,20,40
E + S10,20,40
E + S10,20,40,80
E + S10,20,40,80
E10+A10,20
E10+S20
E + S10,20,40,80
E + S10,20
E10+S20,40,80
E10+S20
E10+A20
E10+A40
E10+S20,S40
E10+S20
E10+S20,S40
E10+S20
E10+R5,R10
E10+S40
255
204
192
457
609
353
220
221
951
181
1474
74
98
288
457
314
516
60
221
120
A10,20,40,80
P10,20,40
L10,20,40
S20
S10,20,40,80
S10,20,40,80
A10,20
A20
A10,20,40,80
S10,20
R10,20,40
S20
A40
A80
A10,20,40
R10
A10,20,40
S40
R10,R20
A40
248
205
220
253
622
349
230
214
951
191
1477
70
98
291
686
304
773
60
219
130
85
66
53
NR
NR
NR
127
141
NR
NR
NR
29
89
266
NR
203
NR
NR
NR
NR
80
64
64
NR
NR
NR
124
116
NR
NR
NR
30
87
273
NR
189
NR
NR
NR
NR
17
11
12
NR
NR
NR
38
59
NR
22
NR
21
NR
155
236
95
NR
77
NR
42
11
14
19
NR
NR
NR
41
53
NR
37
NR
26
NR
153
381
78
NR
97
NR
45
num, number; PHC, primary hypercholesterolaemia; HC, hypercholesterolaemia; CHD, coronary heart disease; CAD, coronary artery disease; DM,
diabetes mellitus; MS, metabolic syndrome; AS, atherosclerosis; E, ezetimibe; S, simvastatin; A, atorvastatin; R, rosuvastatin; L, lovastatin; P, pravastatin;
F, fluvastatin; w, week; NR, not reported.
27.6% and 27.7%, respectively, which did not represent
a significant difference (95% CI, 0.77–1.19; P = 0.69; I2 =
0%) (Fig. 4). In the subgroup analysis of simvastatin,
four trials reported the occurrence of total adverse events
during follow up. There were no treatment differences in
the proportion of patients reporting total adverse events
in both groups (95% CI, 0.65–1.10; P = 0.22; I2 = 0%)
(Fig. 4). This lack of significance persisted in the subgroup analysis of other statins (95% CI, 0.85–1.13; P =
0.75; I2 = 27%) (Fig. 4). Total adverse events were generally similar between the treatments in both groups.
Eighteen studies were assessed in terms of treatment
discontinuation, 169 of 4818 patients (3.5%) discon-
tinued treatment with ezetimibe and statins and 148 of
5142 patients (2.9%) discontinued statins alone. There
was no significant difference between combination
therapy and statin monotherapy (95% CI, 0. 92–1.44; P =
0.22; I2 = 0%), as shown in Figure 5. In a subgroup
analysis of atorvastatin, discontinuation in the combination therapy and atorvastatin monotherapy arms was
2.2% and 2.6%, respectively. There was no significant
difference in discontinuation between the groups (95%
CI, 0. 45–1.43; P = 0.46; I2 = 0%) (Fig. 5). In the subgroup
analysis of simvastatin, there were six trials that reported
the proportion of patients with treatment discontinuation during follow up, although there was no significant
Figure 2 Risk of bias of included studies.
( ), Low risk of bias; ( ), unclear risk of bias;
( ), high risk of bias.
© 2015 Royal Australasian College of Physicians
548
Other bias
Selective reporting (reporting bias)
Incomplete outcome data (attrition bias)
Blinding of outcome assessment (detection bias)
Blinding of paticipants and personnel (performance bias)
Allocation concealment (selection bias)
Random sequence generation (selection bias)
Safety of ezetimibe and statins
Averna et al. 201021
Ballantyne et al. 20035
Ballantyne et al. 200511
Barrios et al. 200512
Bays et al. 20048
Bays et al. 201123
Catapano et al. 200615
Conard et al. 200816
Cruz-Fernandez et al. 200513
Farnier et al. 200514
Farnier et al. 200919
Feldman et al. 20049
Foody et al. 201022
Goldberg et al. 200410
Hing Ling et al. 201224
Kerzner et al. 20036
Leiter et al. 200817
Melanie et al. 20037
Robinson et al. 200920
Zubaid et al. 200818
Figure 3 Risk of bias of individual studies. Blank, not clear; red, high risk;
green, low risk.
difference when comparing combination therapy with
simvastatin monotherapy (95% CI, 0. 81–1.58; P = 0.48;
I2 = 0%) (Fig. 5). This lack of significance persisted in the
subgroup analysis of other statins (95% CI, 0. 95–1.94; P
= 0.10; I2 = 18%) (Fig. 5).
Serious adverse events were reported in 13 studies,
with 76 events occurring in 3997 patients (2%) treated
with ezetimibe and statins, compared with 69 events in
4301 patients (1.6%) treated with statins alone. This end
point was not higher with combination therapy compared with statin monotherapy (95% CI, 0.75 – 1.45; P =
0.81; I2 = 0%), as shown in Figure 6. In the subgroup
analysis of atorvastatin, incidences of serious adverse
events in the combination therapy and atorvastatin
monotherapy arms were 2.1% and 1.5%, respectively,
although this difference was not significant (95% CI, 0.
58–3.09; P = 0.49; I2 = 0%) (Fig. 6). In the subgroup
analysis of simvastatin, five trials reported the proportion
of patients with serious adverse events during follow up.
Both treatments had similar tolerability and safety profiles (95% CI, 0.68–1.78; P = 0.7; I2 = 0%) (Fig. 6). Incidence of serious adverse events was generally
similar between treatments in the subgroup analysis of
other statins (95% CI, 0. 49–1.52; P = 0.60; I2 = 38%)
(Fig. 6).
Nine studies were assessed for gastrointestinal adverse
events. A total of 123 events occurred in 2446 patients
(5%) treated with ezetimibe and statins, compared with
122 events in 2957 patients (4%) treated with statins
alone. There was no significant difference between combination therapy and statin monotherapy (95% CI, 0.
97–1.63; P = 0.08; I2 = 24%), as shown in Figure 7. In the
subgroup analysis of atorvastatin, the incidences of gastrointestinal adverse events in the combination therapy
and atorvastatin monotherapy arms were 4% and 5%
respectively, which did not represent a significant difference (95% CI, 0.51–1.35; P = 0.46; I2 = 53%) (Fig. 7). In
the subgroup analysis of other statins, there were five
trials that reported the occurrence of gastrointestinal
adverse events during follow up. Treatment with other
statins alone was associated with a significant reduction
in gastrointestinal adverse events compared with combination of ezetimibe and statins (95% CI, 1.10–2.02; P =
0.01; I2 = 0%) (Fig. 7).
Six trials reported allergic reactions or rashes. Seventeen events occurred in 1903 patients (0.9%) treated
with ezetimibe and statins, compared with 31 events in
2391 patients (1.3%) treated with statins alone. There
was no significant difference between the groups (95%
CI, 0.41–1.35; P = 0.33; I2 = 0%) (Fig. 8).
CK >10 × ULN, was reported in 11 studies. Eleven events
occurred in 5579 patients (0.2%) treated with ezetimibe
and statins, compared with 10 events in 5850 patients
© 2015 Royal Australasian College of Physicians
549
Luo et al.
Figure 4 Forest plots to show the incidence of total adverse events. CI, confidence interval; OR, odds ratio.
(0.2%) treated with statins alone. There was no significant
difference between the groups (95% CI, 0.51–2.23; P =
0.86; I2 = 0%), as shown in Figure 9. In the subgroup
analysis of atorvastatin, the incidence of CK >10 × ULN in
the combination therapy and statin monotherapy arms
was 0.3% in both. There was no significant difference
between the groups (95% CI, 0.25–2.88; P = 0.79; I2 = 0%)
(Fig. 9). Both treatments had generally similar tolerability
and safety profiles in the subgroup analysis of other statins
(95% CI, 0.48–3.06; P = 0.68; I2 = 0%) (Fig. 9).
The incidence of ALT >3 × ULN was reported in 11
studies and did not reach statistical significance when
compared between the combination therapy and statin
monotherapy groups (95% CI, 0.58–1.77; P = 0.96; I2 =
0%) (Fig. 10). In the subgroup analysis of atorvastatin, 6
events occurred in 629 patients (1%) treated with
ezetimibe and atorvastatin, and 4 in 625 patients (0.6%)
treated with atorvastatin alone. There was no significant
difference between the groups (95% CI, 0.42–5.33; P =
0.54; I2 = 0%) (Fig. 10). There were generally similar
rates reported for the occurrence of ALT >3 ULN in the
subgroup of other statins (95% CI, 0.49–1.72; P = 0.80; I2
= 28%) (Fig. 10).
Seven trials reported the proportion of patients
with AST >3 × ULN. A total of 17 events occurred in
3864 patients (0.4%) treated with ezetimibe and statins,
© 2015 Royal Australasian College of Physicians
550
Safety of ezetimibe and statins
Figure 5 Forest plots to show the incidence of treatment discontinuation. CI, confidence interval; OR, odds ratio.
compared with 16 events in 4335 patients (0.4%) treated
with statins alone. There was no significant difference
between the groups (95% CI, 0.61–2.39; P = 0.58; I2 =
35%) (Fig. 11).
Discussion
Hypercholesterolaemia is one of the risk factors for atherosclerosis and has an indirect relationship with severe
complications of atherosclerosis, such as transient
ischaemic attacks, acute myocardial infarction and
thrombotic stroke, which can have a poor prognosis.
Elevated plasma LDL-C, especially, is a major risk factor
in the development and prevention of atherosclerotic
coronary heart disease and cerebrovascular disease.25
Reduction in LDL-C levels in individuals with preexisting coronary heart disease has been shown to reduce
cardiovascular and cerebrovascular disease morbidity and
total mortality.26–28
Statins have pleiotropic activities, including lowering
cholesterol level and stabilising atherosclerotic plaques
on the blood vessel walls, which can help to prevent
© 2015 Royal Australasian College of Physicians
551
Luo et al.
Figure 6 Forest plots to show the incidence of serious adverse events. CI, confidence interval; OR, odds ratio.
ischaemic events.29 Statins have also made a major
contribution to the treatment of hypercholesterolaemia
and play a key role in comprehensive strategies for
treating cardiovascular diseases in the 21st century.
In addition, statins are the most commonly prescribed
lipid-lowering drugs for patients with elevated
plasma LDL-C and play a major part in the reduction
of
cardiovascular
and
cerebrovascular
disease
morbidity.
Ezetimibe has been shown to be the first selective
inhibitor of cholesterol absorption by reducing overall
delivery to the liver, and the mean decrease in LDL-C
with ezetimibe was 19.1% versus placebo. Phase III
studies have shown a mean additional decrease of LDL-C
(12.1–13.8%) and increase in high-density lipoprotein
cholesterol (1.4–4.5%) with ezetimibe and various statins
at increasing dosages (vs pooled statins).30 Coadministration of ezetimibe and statins results in greater
reductions in LDL-C, which is similar to using the highest
dose of statin monotherapy.
Several studies have found that when the dosage of
statin is doubled, the decrease in LDL-C increases by
5–6%, along with efficacy; patients’ perceived intolerance of statin therapy increases, which is often dose
related and may include elevations in blood levels of liver
or muscle enzymes.31 A previous meta-analysis found
that intensive-dose statin therapy was associated with an
increased risk of adverse drug events, compared with
moderate dose therapy.32 Many patients at high risk for
coronary heart disease and cerebrovascular disease fail to
© 2015 Royal Australasian College of Physicians
552
Safety of ezetimibe and statins
Figure 7 Forest plots to show the incidence of gastrointestinal adverse events. CI, confidence interval; OR, odds ratio.
reach LDL-C targets with statin monotherapy, and
several patients cannot tolerate high doses of statins,
because of dose-related adverse events.33
In addition to efficacy, safety is an important issue
influencing the selection of ezetimibe for combination
with a statin. Ezetimibe and statins are associated with
liver function test abnormalities and can cause
rhabdomyolysis and myositis. Although the incidence of
adverse effects was low, the elevation of liver enzymes
was observed more frequency in ezetimibe–statin combination therapy than statin monotherapy in the overall
population. These rates for statin monotherapy (0.4%)
Figure 8 Forest plots to show the incidence of allergic reactions or rash. CI, confidence interval; OR, odds ratio.
© 2015 Royal Australasian College of Physicians
553
Luo et al.
Figure 9 Forest plots to show the number of patients with creatine kinase (CK) >10 × upper limit of normal (ULN). CI, confidence interval; OR,
odds ratio.
and ezetimibe-statin combination therapy (1.4%) were
consistent with those reported in the prescribing
information.34
Based on these studies, our meta-analysis clarifies the
evidence for safety from RCT comparing ezetimibe–
statin combination therapy with statin monotherapy. In
our study, combination therapy and monotherapy generally had a similar incidence of total adverse events
(30% vs 29%, P = 0.34), serious adverse events (2% vs
1.6%, P = 0.81), treatment discontinuations (3.5% vs
2.9%, P = 0.22), gastrointestinal adverse events (5%
vs 4%, P = 0.08), allergic reactions or rashes (0.9% vs
1.3%, P = 0.33), CK >10 × ULN (0.2% vs 0.2%, P =
0.86), ALT >3 × ULN (0.5% vs 0.4%, P = 0.96) and AST
>3 × ULN (0.4% vs 0.4%, P = 0.58). The incidence of
adverse effects with coadministration of ezetimibe and
statins did not differ significantly from those with statin
monotherapy.
Conclusion
The present meta-analysis shows that ezetimibe–statin
combination therapy is tolerated as well as statin
monotherapy. Our meta-analysis shows that the addition
of ezetimibe to statins is safe, and the safety profile is
similar between coadministration and monotherapy.
Combination therapy has greater efficacy through differing mechanisms of action, can lower doses of individual
drugs and alleviate adverse effects generated with high
doses of single agents. Thus, we recommend combination
therapy for patients with hypercholesterolaemia at high
risk for cardiovascular and cerebrovascular disease.
Acknowledgement
The authors express their gratitude to those who provided their time and assistance for this study.
© 2015 Royal Australasian College of Physicians
554
Safety of ezetimibe and statins
Figure 10 Forest plots to show the number of patients with alanine aminotransferase (ALT) >3 × upper limit of normal (ULN). CI, confidence interval; OR,
odds ratio.
Figure 11 Forest plots to show the number of patients with aspartate aminotransferase (AST) >3 × upper limit of normal (ULN). CI, confidence interval;
OR, odds ratio.
© 2015 Royal Australasian College of Physicians
555
Luo et al.
References
1 Sirtori CR. The pharmacology of statins.
Pharmacol Res 2014; 88: 3–10.
2 Jeu L, Cheng JW. Pharmacology and
therapeutics of ezetimibe (SCH 58235),
a cholesterol-absorption inhibitor. Clin
Ther 2003; 25: 2352–87.
3 Stein EA. An investigative look: selective
cholesterol absorption inhibitors –
embarking on a new standard of care.
Am J Manag Care 2002; 8: S36–9,
discussion S45–37.
4 Third Report of the National Cholesterol
Education Program (NCEP). Expert
panel on detection, evaluation, and
treatment of high blood cholesterol in
adults (Adult Treatment Panel III) final
report. Circulation 2002; 106: 3143–421.
5 Ballantyne CM, Houri J, Notarbartolo A,
Melani L, Lipka LJ, Suresh R et al. Effect
of ezetimibe coadministered with
atorvastatin in 628 patients with
primary hypercholesterolemia: a
prospective, randomized, double-blind
trial. Circulation 2003; 107: 2409–15.
6 Kerzner B, Corbelli J, Sharp S, Lipka LJ,
Melani L, LeBeaut A et al. Efficacy and
safety of ezetimibe coadministered
with lovastatin in primary
hypercholesterolemia. Am J Cardiol 2003;
91: 418–24.
7 Melani L, Mills R, Hassman D, Lipetz R,
Lipka L, LeBeaut A et al. Efficacy and
safety of ezetimibe coadministered with
pravastatin in patients with primary
hypercholesterolemia: a prospective,
randomized, double-blind trial. Eur Heart
J 2003; 24: 717–28.
8 Bays HE, Ose L, Fraser N, Tribble DL,
Quinto K, Reyes R et al. A multicenter,
randomized, double-blind,
placebo-controlled, factorial design
study to evaluate the lipid-altering
efficacy and safety profile of the
ezetimibe/simvastatin tablet compared
with ezetimibe and simvastatin
monotherapy in patients with primary
hypercholesterolemia. Clin Ther 2004;
26: 1758–73.
9 Feldman T, Koren M, Insull W,
McKenney J, Schrott H, Lewin A et al.
Treatment of high-risk patients
with ezetimibe plus simvastatin
co-administration versus simvastatin
alone to attain National Cholesterol
Education Program Adult Treatment
Panel III low-density lipoprotein
cholesterol goals. Am J Cardiol 2004; 93:
1481–6.
10 Goldberg AC, Sapre A, Liu J, Capece R,
Mitchel YB. Efficacy and safety of
ezetimibe coadministered with
simvastatin in patients with primary
hypercholesterolemia: a randomized,
double-blind, placebo-controlled trial.
Mayo Clin Proc 2004; 79: 620–9.
11 Ballantyne CM, Abate N, Yuan Z, King
TR, Palmisano J. Dose-comparison study
of the combination of ezetimibe and
simvastatin (Vytorin) versus atorvastatin
in patients with hypercholesterolemia:
the Vytorin Versus Atorvastatin (VYVA)
study. Am Heart J 2005; 149: 464–73.
12 Barrios V, Amabile N, Paganelli F, Chen
JW, Allen C, Johnson-Levonas AO et al.
Lipid-altering efficacy of switching
from atorvastatin 10 mg/day to
ezetimibe/simvastatin 10/20 mg/day
compared to doubling the dose of
atorvastatin in hypercholesterolaemic
patients with atherosclerosis or coronary
heart disease. Int J Clin Pract 2005; 59:
1377–86.
13 Cruz-Fernandez JM, Bedarida GV,
Adgey J, Allen C, Johnson-Levonas AO,
Massaad R et al. Efficacy and safety of
ezetimibe co-administered with ongoing
atorvastatin therapy in achieving
low-density lipoprotein goal in patients
with hypercholesterolemia and coronary
heart disease. Int J Clin Pract 2005; 59:
619–27.
14 Farnier M, Volpe M, Massaad R, Davies
MJ, Allen C. Effect of co-administering
ezetimibe with on-going simvastatin
treatment on LDL-C goal attainment in
hypercholesterolemic patients with
coronary heart disease. Int J Cardiol
2005; 102: 327–32.
15 Catapano AL, Davidson MH, Ballantyne
CM, Brady WE, Gazzara RA, Tomassini
JE et al. Lipid-altering efficacy of
the ezetimibe/simvastatin single
tablet versus rosuvastatin in
hypercholesterolemic patients. Curr Med
Res Opin 2006; 22: 2041–53.
16 Conard SE, Bays HE, Leiter LA, Bird SR,
Rubino J, Lowe RS et al. Efficacy and
safety of ezetimibe added on to
atorvastatin (20 mg) versus uptitration
of atorvastatin (to 40 mg) in
hypercholesterolemic patients at
moderately high risk for coronary heart
disease. Am J Cardiol 2008; 102:
1489–94.
17 Leiter LA, Bays H, Conard S, Bird S,
Rubino J, Hanson ME et al. Efficacy and
safety of ezetimibe added on to
atorvastatin (40 mg) compared with
18
19
20
21
22
23
24
uptitration of atorvastatin (to 80 mg) in
hypercholesterolemic patients at high
risk of coronary heart disease. Am J
Cardiol 2008; 102: 1495–501.
Zubaid M, Shakir DK, Bazargani N,
Binbrek A, Gopal R, Al-Tamimi O et al.
Effect of ezetimibe coadministration with
simvastatin in a Middle Eastern
population: a prospective, multicentre,
randomized, double-blind,
placebo-controlled trial. J Cardiovasc Med
(Hagerstown) 2008; 9: 688–93.
Farnier M, Averna M, Missault L,
Vaverkova H, Viigimaa M, Massaad R
et al. Lipid-altering efficacy of
ezetimibe/simvastatin 10/20 mg
compared with rosuvastatin 10 mg in
high-risk hypercholesterolaemic
patients inadequately controlled with
prior statin monotherapy – The
IN-CROSS study. Int J Clin Pract 2009;
63: 547–59.
Robinson JG, Ballantyne CM, Grundy
SM, Hsueh WA, Parving HH, Rosen JB
et al. Lipid-altering efficacy and safety of
ezetimibe/simvastatin versus atorvastatin
in patients with hypercholesterolemia
and the metabolic syndrome (from the
VYMET study). Am J Cardiol 2009; 103:
1694–702.
Averna M, Zaninelli A, Le Grazie C,
Gensini GF. Ezetimibe/simvastatin
10/20 mg versus simvastatin 40 mg in
coronary heart disease patients. J Clin
Lipidol 2010; 4: 272–8.
Foody JM, Brown WV, Zieve F, Adewale
AJ, Flaim D, Lowe RS et al. Safety and
efficacy of ezetimibe/simvastatin
combination versus atorvastatin alone in
adults >/=65 years of age with
hypercholesterolemia and with or at
moderately high/high risk for coronary
heart disease (the VYTELD study). Am J
Cardiol 2010; 106: 1255–63.
Bays HE, Davidson MH, Massaad R,
Flaim D, Lowe RS, Tershakovec AM
et al. Safety and efficacy of ezetimibe
added on to rosuvastatin 5 or 10 mg
versus up-titration of rosuvastatin in
patients with hypercholesterolemia (the
ACTE Study). Am J Cardiol 2011; 108:
523–30.
Hing Ling PK, Civeira F, Dan AG,
Hanson ME, Massaad R,
De Tilleghem Cle B et al.
Ezetimibe/simvastatin 10/40 mg versus
atorvastatin 40 mg in high
cardiovascular risk patients with primary
hypercholesterolemia: a randomized,
double-blind, active-controlled,
© 2015 Royal Australasian College of Physicians
556
Safety of ezetimibe and statins
multicenter study. Lipids Health Dis 2012;
11: 18–25.
25 LaRosa JC. Low-density lipoprotein
cholesterol reduction: the end is more
important than the means. Am J Cardiol
2007; 100: 240–2.
26 Pedersen TR, Kjekshus J, Berg K,
Haghfelt T, Faergeman O, Faergeman G
et al. Randomised trial of cholesterol
lowering in 4444 patients with coronary
heart disease: the Scandinavian
Simvastatin Survival Study (4S). 1994.
Atheroscler Suppl 2004; 5: 81–7.
27 Heart Protection Study Collaborative
Group. MRC/BHF Heart Protection
Study of cholesterol lowering with
simvastatin in 20 536 high-risk
individuals: a randomised
placebo-controlled trial. Lancet 2002;
360: 7–22.
28 Shepherd J. Preventing coronary artery
disease in the West of Scotland:
implications for primary prevention. Am
J Cardiol 1998; 82: 57T–59T.
29 Morishita R, Tomita N, Ogihara T.
HMG-Co A reductase inhibitors in the
treatment of cardiovascular diseases:
stabilization of coronary artery
plaque. Curr Drug Targets 2002; 3:
379–85.
30 Tellier P. Ezetimibe: from pharmacology
to clinical trials. Ann Endocrinol (Paris)
2003; 64: 442–7.
31 Bays H, Dujovne C. Colesevelam HCl: a
non-systemic lipid-altering drug. Expert
Opin Pharmacother 2003; 4: 779–90.
32 Silva M, Matthews ML, Jarvis C, Nolan
NM, Belliveau P, Malloy M et al.
Meta-analysis of drug-induced adverse
events associated with intensive-dose
statin therapy. Clin Ther 2007; 29:
253–60.
33 Daskalopoulou SS, Mikhailidis DP.
Reaching goal in hypercholesterolaemia:
dual inhibition of cholesterol synthesis
and absorption with simvastatin plus
ezetimibe. Curr Med Res Opin 2006; 22:
511–28.
34 Davidson MH, Ballantyne CM, Kerzner
B, Melani L, Sager PT, Lipka L et al.
Efficacy and safety of ezetimibe
coadministered with statins: randomised,
placebo-controlled, blinded experience
in 2382 patients with primary
hypercholesterolemia. Int J Clin Pract
2004; 58: 746–55.
Crescentic glomerulonephritis: data from the Spanish
Glomerulonephritis Registry
B. Quiroga,1 A. Vega,1 F. Rivera2 and J. M. López-Gómez,1 on behalf of all members of the Spanish Registry
of Glomerulonephritis
1
Hospital Gregorio Marañón, Madrid and 2Hospital General de Ciudad Real, Ciudad Real, Spain
Key words
age, crescentic glomerulonephritis,
Goodpasture, vasculitis, registry.
Correspondence
Borja Quiroga, Hospital Gregorio Marañón,
C/Doctor Esquerdo 46, Madrid, 28007, Spain.
Email: [email protected]
Received 17 November 2014; accepted 1
February 2015.
doi:10.1111/imj.12725
Abstract
Background: Crescentic glomerulonephritis (CGN) is a histological finding that implies
rapid deterioration of renal function and can be related to different diseases, such as type
1 or anti-glomerular basement membrane antibody (Goodpasture) disease, type 2 or
immune complex CGN and type 3 or pauci-immune disease.
Aim: The present study describes CGN and its characteristics based on the data from the
Spanish Glomerulonephritis Registry.
Methods: An analysis was made of all native renal biopsies obtained from patients
during 1994–2013 and classified as CGN. A patient epidemiological and clinical data
questionnaire was completed by the 120 centres involved.
Results: A total of 21 774 biopsies was performed, of which 2089 (8.1%) corresponded
to CGN (211 type 1, 177 type 2 and 1701 type 3). Renal function was poorer in type 1
compared with types 2 and 3, and proteinuria was higher in type 2 compared to types
1 and 3. Patients diagnosed with CGN type 3 were older than those with types 1 and 2,
but less hypertensive than the type 2 patients. No differences in the urine test findings
were found between types 1 and 2. Microhaematuria was the most frequent feature in
general, as well as in type 3 compared with types 1 and 2. The main indication for biopsy
was acute renal injury. Age was the only difference between type 1 patients with and
without alveolar haemorrhage (53 [33–67] vs 64 [46–73], P = 0.008).
Conclusion: Although classified as the same entity, the different types of CGN have
different features that must be taken into account.
© 2015 Royal Australasian College of Physicians
557
Quiroga et al.
Introduction
Crescentic glomerulonephritis (CGN) is defined by the
presence of more than 50% of glomerular crescents in a
renal biopsy. These histological findings imply rapid
deterioration of renal function, but can be related
to different diseases with different clinical, diagnostic,
treatment and prognostic features.1 There are three
main types of CGN: type 1 or anti-glomerular basement
membrane (GBM) antibody (Goodpasture) disease; type
2 or immune complex CGN; and type 3 or pauci-immune
disease.2 Recently, a double-antibody positive disease has
been accepted as CGN type 4. The disease typically manifests as impaired renal function, proteinuria (usually
<3 g/day) and haematuria. Types 1 and 3 usually also
show systemic symptoms.3,4 Although included within
the same type of glomerulonephritis (GN), CGN is not a
specific disease and can be caused by different mechanisms. Furthermore, the differences among them have
not been fully described.5 At present, immunological
tests, including anti-neutrophil cytoplasmic antibodies
(ANCA) and anti-GBM antibodies, can help clinicians to
assess patient diagnosis, relapse and treatment response.
Renal biopsies are the gold standard for diagnosing
kidney diseases. Histopathological data give nephrologists
important information regarding not only the diagnosis
but also the prognosis, with a view to making appropriate
management decisions. CGN and its pathological features
are very important for establishing proper management,
because treatment includes aggressive drugs, such
as immunosuppressive agents. In the early stages,
inflammatory cells and the expression of cytokines
(interleukin-1 and tumour necrosis factor alpha) are the
main findings. Based on the natural history of the
disease, the next step is the formation of glomerular
crescents that are firstly epithelial and lead to fibrosis and
irreversible renal failure.6,7 The presence of crescents in a
biopsy usually implies rapid progression of the disease
and also the need to decide treatment.8 Only if irreversible changes are found in the biopsy (e.g. fibrosis) should
non-aggressive immunosuppressive therapy be chosen.
Medical registries and networks are useful tools for
describing diseases, due to the large number of centres
involved and the important size of the samples. Studies of
diseases that are not particularly frequent, such as GN,
stand to benefit most from such registries.9 The United
States, Brazil, Europe (Spain, Italy, the Czech Republic,
France and Hungary) and Asia have their own biopsy
registries.10–12 The Spanish Glomerulonephritis Registry
has been collecting data in this regard since 1994, and now
Funding: None.
Conflict of interest: None.
has registered over 20 000 biopsies.13–15 Thanks to the
large records of biopsies, we now know that the natural
history of GN exhibits a changing pattern. Immunosuppressive drugs are improving, and histological techniques
supported by serum tests yield prompt results, thereby
allowing treatments to be started earlier. Descriptions of
these changes and the evaluation of correlations between
clinical data and histological patterns are essential in order
to improve outcomes in serious diseases, such as GN.
Aims
Based on the data of the Spanish Glomerulonephritis
Registry covering the period between 1994 and 2013, the
present study describes and analyses the different types
on CGN, including their incidence, epidemiological data,
clinical manifestations and laboratory features.
Methods
We analysed all native renal biopsies obtained from
patients classified as corresponding to CGN and included
in the Spanish Glomerulonephritis Registry between
1994 and 2013, together with the corresponding clinical
indications. Each sample was analysed by pathologists
from the 120 participating hospitals using specific techniques, mainly light microscopy and direct immunofluorescence (IgG, IgA, IgM, C3, C1q, fibrinogen and lightchain antibodies). For sending the biopsies to the registry
(through www.senefro.org, official webpage of the
Spanish Society of Nephrology) a questionnaire (available only in Spanish) was sent by the participant centres
(including year, hospital, name of the sender, age of the
patient, gender, renal function (serum creatinine, estimated glomerular filtration rate, proteinuria, description
of the sediment), main syndrome of the patient, history
of hypertension and data regarding the biopsy (number
of glomeruli and main diagnosis). The registry only
included the main diagnosis, so superimposed syndromes
could not be identified. Also, in the registry the diagnosis
was codified, but we do not have access to the pathological description.
A questionnaire on the patient epidemiological and
clinical data was completed. The following definitions
were established: (i) acute renal injury: rapid deterioration of the glomerular filtration rate (GFR), with or
without oligoanuria or rapidly progressive renal failure,
including a worsening of chronic kidney disease, (ii)
nephrotic syndrome: proteinuria >3.5 g/day/1.73 m2 and
serum albumin <2.5 g/dL, (iii) acute nephritic syndrome,
oliguric acute renal injury with oedema, haematuria and
hypertension, (iv) asymptomatic urinary abnormalities:
proteinuria <3.5 g/day and/or haematuria with more
© 2015 Royal Australasian College of Physicians
558
Crescentic glomerulonephritis in Spain
Table 1 Baseline characteristics
Age (years)
Gender (% males)
Hypertension (%)
Creatinine (mg/dL)
24-h creatinine
clearance (mL/min)
Proteinuria (g/day)
Glomeruli (number)
Total (n = 2089)
CGN 1 (n = 211)
CGN 2 (n = 177)
CGN 3 (n = 1701)
P CGN 1 vs 2
P CGN 1 vs 3
P CGN 2 vs 3
66 (52–73)
57.3
54.0
4.1 (2.5–6.9)
15 (8–30)
58 (39–71)
60.2
55.2
6.9 (3.9–10)
10 (5–20)
60 (41–72)
64.4
65.7
4.8 (3.0–7.5)
15 (7–24)
66 (55–74)
56.1
52.6
3.9 (2.3–6.2)
17 (9–32)
NS
NS
0.059
<0.001
0.029
<0.001
NS
NS
<0.001
<0.001
<0.001
0.054
0.003
0.001
0.034
1.7 (0.9–3.3)
13 (9–20)
2.0 (0.7–4.0)
13 (8–20)
3.0 (1.7–5.4)
13 (9–20)
1.6 (0.8–3.0)
14 (9–20)
0.001
0.067
<0.001
CGN, crescentic glomerulonephritis; NS, non-significant.
than three red blood cells per field, in the absence of
clinical manifestations, (v) arterial hypertension: blood
pressure >140/90 mmHg or antihypertensive treatment
irrespective of blood pressure and (vi) chronic kidney
disease: persistent serum creatinine >1.5 mg/dL.
The questionnaire was applied to obtain the following
information: identification code, date of birth, gender,
hospital, presence of hypertension and/or antihypertensive treatment, serum creatinine (mg/dL), 24-h urine
creatinine clearance (mL/min), proteinuria (g/day) and
urinary sediment. We also noted the main renal syndrome, the histological methods applied to the sample
and the number of glomeruli obtained. CGN was defined
as the presence of crescents in >50% of the glomeruli,
and was classified as follows:
• Type 1: accompanied by anti-glomerular basal membrane antibodies, with or without alveolar haemorrhage.
• Type 2: presence of immune complexes.
• Type 3: necrotising GN with or without ANCA or systemic vasculitis symptoms.
As this registry exists from 1994, and no changes in
definitions have been performed to avoid confusion in
data, some of them are outdated. The cut-off of 50% of
crescent formation for achieving the definition was due
to definition of the entity in 1994.16,17 No data regarding
the type of crescent are available in this registry.
SPSS version 16.0 statistical package for Microsoft
Windows (SPSS, Chicago, IL, USA). Statistical significance was considered for P < 0.05.
Results
Baseline characteristics are shown in Table 1. The
frequencies of each type of CGN and the overall percentages divided into five periods of 4 years each are
shown in Figure 1. Taking into account all the renal
biopsies performed (n = 21 774), 8.1% (n = 2089) corresponded to CGN; 0.8% (n = 211) corresponded to
type 1, 0.7% (n = 177) to type 2, and 6.5% (n = 1701)
to type 3. CGN type 1 was characterised by poorer renal
function than types 2 and 3. Proteinuria was higher in
CGN type 2 than in types 1 and 3. In turn, patients
diagnosed with CGN type 3 were older than those with
types 1 and 2 disease, but less hypertensive than
patients with CGN type 2.
The urine test results are shown in Table 2. The
most frequent characteristic in all types of CGN was
microhaematuria. We found no significant differences
Statistical analysis
The data were entered in a Microsoft Access database.
Values are expressed as the mean (SD) or median
(interquartile range). The normal distribution of the
samples was determined using the Kolmogorov–Smirnov
test. Categorical data were compared using the chisquared test or the Fisher’s exact test, while continuous
variables were compared using the Student t-test or
Mann–Whitney U-test. Analysis of variance was used
when several parameters of the two groups were
compared. Statistical analysis was performed using the
Figure 1 Evolution of the incidence of crescentic glomerulonephritis
(CGN).
© 2015 Royal Australasian College of Physicians
559
Quiroga et al.
Table 2 Urine test findings at diagnosis
Gross haematuria
Microhaematuria
Leukocyturia
Cylindruria
Telescoped sediment
Normal
Total (n = 2089)
CGN 1 (n = 211)
CGN 2 (n = 177)
CGN 3 (n = 1701)
P CGN 1 vs 2
P CGN 1 vs 3
P CGN 2 vs 3
334 (16%)
1170 (56%)
23 (1.1%)
23 (1.1%)
310 (14.8%)
65 (3.1%)
47 (26.3%)
83 (46.4%)
0 (0%)
3 (1.7%)
23 (12.8%)
4 (2.2%)
34 (22.5%)
71 (47%)
0 (0%)
1 (0.7%)
32 (21.2%)
4 (2.2%)
211 (14.6%)
826 (57.2%)
20 (1.4%)
12 (0.8%)
219 (15.2%)
45 (3.1%)
NS
NS
NS
NS
0.054
NS
<0.001
0.006
NS
NS
NS
NS
0.010
0.016
NS
NS
0.054
NS
CGN, crescentic glomerulonephritis; NS, non-significant.
between CGN types 1 and 2. At diagnosis, CGN type 3
had less gross haematuria, but microhaematuria was
more frequent than in the other two types.
Clinical features are shown in Table 3. The main indication of biopsy was acute renal injury, followed by
nephritic syndrome. At diagnosis, hypertension was
more frequent as the indication of biopsy in CGN type 3
in comparison to types 1 and 2. Also, chronic kidney
disease was a more frequent indication of biopsy in CGN
type 3 than in type 1 disease.
A comparison between Goodpasture syndrome with
and without alveolar haemorrhage (Table 4) only identified differences in age (older patients presenting less
alveolar haemorrhage) (P = 0.008).
The results among the elderly patients were analysed,
stratifying age into three groups (<65 years, 65–75 years
and >75 years). We found 61.7% of the patients diagnosed with CGN type 1 to be under 65 years of age;
22.9% were between 65–75 years of age, and 7.3% were
over 75 years of age. In patients with CGN type 2, 62.8%
were under 65 years of age, 21.6% were between 65–75
years of age and 15.5% over 75 years of age. Lastly,
among the patients who developed CGN type 3, 44.6%
were under 65 years of age, 32.7% were between 65–75
years of age, and 22.6% were over 75 years of age. The
differences in the distribution were statistically significant
when comparing CGN type 3 with types 1 and 2
(P < 0.0001 for both).
Discussion
Considering the large number of biopsies included in our
study, this is the largest series of CGN published to date.
A total of 8.1% of the biopsies included in the Spanish
Glomerulonephritis Registry corresponded to CGN. Of
these cases, 10% corresponded to CGN type 1, 8% to
type 2 and 81% to type 3. Considering a minimum of 10
glomeruli as adequate to the effects of diagnosis, our
series presented a median of 13 (all biopsies included).18
In coincidence with previous studies,19 patients developing CGN types 1 and 2 were younger, with a median age
of 58 and 60 years, respectively, when compared with
type 3 (66 years). On analysing the patients stratified by
age, and focusing on the elderly, we found that patients
over 75 years of age developed CGN type 3 more often
than the other groups. We recently published data
regarding the elderly (>75 years of age), showing a higher
prevalence of CGN type 3 in comparison to types 1 and
2.18 No gender differences were found between groups
(56.1–64.4% were males), though males tended to be
less frequent in CGN type 3.20
Since the introduction of laboratory tests to assess
ANCA/anti-GBM antibodies, biopsies for establishing a
diagnosis have decreased in number. In this context,
biopsies are presently more relevant for defining a prognosis, quantifying epithelial and fibrotic crescents to
decide whether to continue with immunosuppression
Table 3 Clinical features at diagnosis
Nephrotic syndrome
Nephritic syndrome
Hypertension
Asymptomatic urinary
abnormalities
Acute renal failure
Chronic kidney disease
Others
Unknown
Total (n = 2089)
CGN 1 (n = 211)
CGN 2 (n = 177)
CGN 3 (n = 1701)
P CGN 1 vs 2
P CGN 1 vs 3
P CGN 2 vs 3
144 (6.9%)
346 (16.6%)
146 (7%)
3 (0.1%)
12 (6.7%)
34 (19%)
3 (1.7%)
0 (0%)
15 (9.9%)
29 (19.2%)
6 (4.0%)
0 (0%)
85 (5.9%)
232 (16.1%)
128 (8.9%)
3 (0.2%)
NS
NS
NS
NS
NS
NS
<0.001
NS
0.051
NS
0.039
NS
112 (62.6%)
3 (1.7%)
3 (1.7%)
2 (1.1%)
84 (55.6%)
9 (6.0%)
2 (1.3%)
1 (0.7%)
820 (56.8%)
97 (6.7%)
11 (0.8%)
6 (0.4%)
NS
0.072
NS
NS
NS
<0.001
NS
NS
NS
NS
NS
NS
1211 (58%)
126 (6%)
26 (1.2%)
87 (4.2%)
CGN, crescentic glomerulonephritis; NS, non-significant.
© 2015 Royal Australasian College of Physicians
560
Crescentic glomerulonephritis in Spain
Table 4 Comparison between Goodpasture syndrome with and without alveolar haemorrhage
Age (years)
Gender (male)
Hypertension (presence)
Creatinine (mg/dL)
24-h creatinine clearance (mL/min)
Proteinuria (g/day)
Urine test findings
Gross haematuria
Microhaematuria
Leukocyturia
Cylindruria
Telescoped sediment
Normal
Clinical features
Nephrotic syndrome
Nephritic syndrome
Hypertension
Asymptomatic urinary abnormalities
Acute renal failure
Chronic kidney disease
Others
With alveolar haemorrhage (n = 92)
Without alveolar haemorrhage (n = 87)
P
53 (33–67)
55 (64.7%)
41 (51.3%)
7 (3.5–9.9)
10 (5–19.5)
2.1 (0.7–4.5)
64 (46–73)
51 (56%)
50 (58.8%)
6.8 (4.6–10)
9 (5–23)
2.0 (0.7–4.0)
0.008
NS
NS
NS
NS
NS
NS
22 (28.6%)
39 (50.6%)
0
1 (1.2%)
12 (15.5%)
2 (2.6%)
25 (30.1%)
44 (53%)
0
1 (1.2%)
11 (13.3%)
2 (2.4%)
6 (7.1%)
15 (17.6%)
2 (2.4%)
0
58 (68.2%)
0
4 (4.7%)
6 (7.1%)
19 (22.6%)
1 (1.2%)
0
54 (64.3%)
3 (3.6%)
1 (1.2%)
NS
NS, non-significant.
therapy or to prescribe more conservative treatment.21
However, while the mentioned immune tests yield high
(99%) positive predictive values, the negative predictive
values are only in the order of 80% (and even lower in
older patients). A renal biopsy is therefore mandatory if
CGN is suspected.1
CGN is one of the leading histopathologically diagnosed aetiologies underlying acute or rapidly progressive
renal injury.22 The indication of a renal biopsy for this
reason was registered in 58% of the cases, followed by
nephritic and nephrotic syndrome (16.6% and 6.9%
respectively). Interestingly, as shown in Table 3, hypertension accounted for 8.9% of the indications of biopsy in
CGN type 3, showing significant differences with respect
to types 1 and 2. Hypertension usually precedes flare-up
or relapse in ANCA-associated vasculitis, and renal biopsy
may be useful for diagnosing and starting treatment
before renal injury has developed.19 Chronic kidney
disease accounted for 6% and 6.7% of the indications in
CGN types 2 and 3 versus only 1% in CGN type 1 – thus
showing that this disorder usually debuts more aggressively than the other two types. This condition is confirmed by the fact that renal function is poorer in type 1
than in types 2 and 3, as a consequence of the higher
frequency of crescent formation at the time of diagnosis.19 The strongest predictor for renal and survival outcomes in CGN is renal function at the time of diagnosis,
which is correlated to the histological findings.23,24
Probably, as a result of the introduction of serum tests,
renal function at debut or onset has improved when
compared with previous reports.1,19
Important deterioration of renal function can lead to
low levels of proteinuria, and this is probably the main
reason for finding of sub-nephrotic protein excretion in
CGN.25 Our results indicate a median proteinuria of 1.7 g/
day, with higher protein urinary excretion in CGN type 2
(median 3.0 g/day) compared with the other disease
types. Haematuria is a constant finding in all types of
CGN16 and also in our series, with 85% of the patients
presenting gross haematuria, microhaematuria or telescoped sediment. However, the kind of haematuria differs
between groups. In this sense, microhaematuria appears
frequently in CGN type 3, unlike gross haematuria,
which is more often seen in types 1 and 2.
As observed in other series, anti-GBM antibody disease
could be associated with pulmonary bleeding in one-half
of all cases.5,17 The presence of lung bleeding was more
common in young patients, as published elsewhere;
however, no differences were observed in terms of
gender, clinical features, urine test findings or renal
function.17,25
Our study has a series of limitations. First, biopsy complications were not recorded. Second, no data were collected on more complex analytical parameters, such as
autoimmunity and the presence of ANCAs. Lastly, the
clinical questionnaire accompanying each renal biopsy
© 2015 Royal Australasian College of Physicians
561
Quiroga et al.
did not include treatment or patient outcome. CGN type
4 is not included in the registry, since it dates from 1994.
presentation, renal function, degree of proteinuria and
the urine test characteristics differ.
Conclusion
Acknowledgements
Although the three types of CGN are usually classified as
the same entity, some features, such as patient age at
The authors thank the participating hospitals for submitting the results of their renal biopsies.
References
1 Kambham N. Crescentic
Glomerulonephritis: an update on
pauci-immune and anti-GBM
diseases. Adv Anat Pathol 2012; 19:
111–24.
2 Seo P, Stone JH. The antineutrophil
cytoplasmic antibody-associated
vasculitides. Am J Med 2004; 117: 39–50.
3 Hauer HA, Bajema IM, van
Houwelingen HC, Ferrario F, Noël LH,
Waldherr R et al. Renal histology in
ANCA-associated vasculitis: differences
between diagnostic and serologic
subgroups. Kidney Int 2002; 61: 80–9.
4 Haas M, Eustace JA. Immune complex
deposits in ANCA-associated crescentic
glomerulonephritis: a study of 126 cases.
Kidney Int 2004; 65: 2145–52.
5 Jennette JC, Thomas DB. Crescentic
glomerulonephritis. Nephrol Dial
Transplant 2001; 16(Suppl 6):
80–2.
6 Tarzi RM, Cook HT, Pusey CD.
Crescentic glomerulonephritis: new
aspects of pathogenesis. Semin Nephrol
2011; 31: 361–8.
7 Atkins RC, Nikolic-Paterson DJ, Song Q,
Lan HY. Modulators of crescentic
glomerulonephritis. J Am Soc Nephrol
1996; 7: 2271–8.
8 Singh SK, Jeansson M, Quaggin SE.
New insights into the pathogenesis of
cellular crescents. Curr Opin Nephrol
Hypertens 2011; 20: 258–62.
9 Pesce F, Schena FP. Worldwide
distribution of glomerular diseases: the
role of renal biopsy registries. Nephrol
Dial Transplant 2010; 25: 334–6.
10 Rychlík I, Jancová E, Tesar V, Kolsky A,
Lácha J, Stejskal J et al. The Czech
11
12
13
14
15
16
17
registry of renal biopsies. Occurrence of
renal diseases in the years 1994–2000.
Nephrol Dial Transplant 2004; 19: 3040–9.
Schena FP. Survey of the Italian Registry
of Renal Biopsies. Frequency of the
renal diseases for 7 consecutive years.
The Italian Group of Renal
Immunopathology. Nephrol Dial
Transplant 1997; 12: 418–26.
Simon P, Ramee MP, Boulahrouz R,
Stanescu C, Charasse C, Ang KS et al.
Epidemiologic data of primary
glomerular diseases in western France.
Kidney Int 2004; 66: 905–8.
López-Gómez JM, Rivera F, on behalf of
Spanish Registry of Glomerulonephritis.
Renal biopsy findings in acute renal
failure in the cohort of patients in the
Spanish Registry of Glomerulonephritis.
Clin J Am Soc Nephrol 2008; 3: 674–81.
Goicoechea M, Rivera F, López-Gómez
JM, Spanish Registry of
Glomerulonephritis. Increased
prevalence of acute tubulointerstitial
nephritis. Nephrol Dial Transplant 2013;
28: 112–15.
Verde E, Quiroga B, Rivera F,
López-Gómez JM. Renal biopsy
in very elderly patients: data
from the Spanish Registry of
Glomerulonephritis. Am J Nephrol 2012;
35: 230–7.
Bruns FJ, Adler S, Fraley DS, Segel DP.
Long-term follow-up of aggressively
treated idiopathic rapidly progressive
glomerulonephritis. Am J Med 1989; 86:
400–6.
Wilson CB, Dixon FJ. Anti-glomerular
basement membrane antibody-induced
glomerulonephritis. Kidney Int 1973; 3:
74–89.
18 Berden AE, Ferrario F, Hagen EC, Jayne
DR, Jennette JC, Joh K et al.
Histopathologic classification of
ANCA-associated glomerulonephritis.
J Am Soc Nephrol 2010; 21: 1628–36.
19 Robson J, Doll H, Suppiah R, Flossmann
O, Harper L, Höglund P et al. Damage in
the anca-associated vasculitides:
long-term data from the European
Vasculitis Study group (EUVAS)
therapeutic trials. Ann Rheum Dis 2013.
doi:10.1136/annrheumdis
-2013-203927
20 Jennette JC, Falk RJ. Small-vessel
vasculitis. N Engl J Med 1997; 337:
1512–23.
21 Lee RW, D’Cruz DP. Pulmonary renal
vasculitis syndromes. Autoimmun Rev
2010; 9: 657–60.
22 Jennette JC. Rapidly progressive
crescentic glomerulonephritis. Kidney Int
2003; 63: 1164–77.
23 Levy JB, Turner AN, Rees AJ, Pusey CD.
Long-term outcome of anti-glomerular
basement membrane antibody disease
treated with plasma exchange and
immunosuppression. Ann Intern Med
2001; 134: 1033–42.
24 Hogan SL, Nachman PH, Wilkman AS,
Jennette JC, Falk RJ. Prognostic markers
in patients with antineutrophil
cytoplasmic autoantibody-associated
microscopic polyangiitis and
glomerulonephritis. J Am Soc Nephrol
1996; 7: 23–32.
25 Floege J, Johnson RJ, Feehally J.
Comprehensive Clinical Nephrology, 4rd.
St Louis, Missouri: Saunders Elsevier;
2010.
© 2015 Royal Australasian College of Physicians
562
Internal Medicine Journal 45 (2015)
P O S I T I O N PA P E R
Consensus guidelines for the investigation and management
of encephalitis in adults and children in Australia and
New Zealand
P. N. Britton,1,2 K. Eastwood,3,4 B. Paterson,4 D. N. Durrheim,4 R. C. Dale,1,5 A. C. Cheng,6,7 C. Kenedi,8,9,10
B. J. Brew,11,12 J. Burrow,13 Y. Nagree,14,15 P. Leman,14,16 D. W. Smith,14 K. Read,17 R. Booy1,2,18 and
C. A. Jones,1,2 on behalf of the Australasian Society of Infectious Diseases (ASID), Australasian College of
Emergency Medicine (ACEM), Australian and New Zealand Association of Neurologists (ANZAN) and the
Public Health Association of Australia (PHAA)
1
Discipline of Paediatrics and Child Health and Marie Bashir Institute for Emerging Infectious Diseases and Biosecurity, Sydney Medical School,
University of Sydney, Departments of 2Infectious Diseases and Microbiology and 5Neurology, and 18National Centre for Immunisation Research and
Surveillance, The Children’s Hospital at Westmead, 11St Vincent’s Centre for applied medical research, University of New South Wales, 12Department of
Neurology, St Vincent’s Hospital, Sydney, 3Health Protection, Hunter New England Population Health, 4Biopreparedness, Hunter Medical Research
Institute, Newcastle, New South Wales, 6Department of Infectious Diseases, The Alfred Hospital, 7Department of Epidemiology and Preventive
Medicine, Monash University, Melbourne, Victoria, 13Department of Neurology, Royal Darwin Hospital, Darwin, Northern Territory, 14Faculty of
Medicine, Dentistry and Health Sciences, University of Western Australia, 16Emergency Department, Royal Perth Hospital, Perth, 15Emergency
Department, Fremantle Hospital, Fremantle, Western Australia, Australia; Departments of 8General Medicine and 9Liaison Psychiatry, Auckland City
Hospital, 17Department of Infectious Diseases, North Shore Hospital, Auckland, New Zealand; and 10Department of Medicine and Department of
Psychiatry, Duke University Medical Center, Durham, North Carolina, USA
Key words
encephalitis, guideline, Australia, New Zealand.
Correspondence
Cheryl Jones, Discipline of Paediatrics and Child
Health, Sydney Medical School, C/o The
Children’s Hospital at Westmead, Locked Bag
4001, Westmead, NSW 2145, Australia.
Email: [email protected]
Received 2 November 2014; accepted 17
February 2015.
doi:10.1111/imj.12749
Abstract
Encephalitis is a complex neurological syndrome caused by inflammation of the brain
parenchyma. The management of encephalitis is challenging because: the differential
diagnosis of encephalopathy is broad; there is often rapid disease progression; it often
requires intensive supportive management; and there are many aetiologic agents for
which there is no definitive treatment. Patients with possible meningoencephalitis are
often encountered in the emergency care environment where clinicians must consider
differential diagnoses, perform appropriate investigations and initiate empiric antimicrobials. For patients who require admission to hospital and in whom encephalitis is
likely, a staged approach to investigation and management is preferred with the potential involvement of multiple medical specialties. Key considerations in the investigation
and management of patients with encephalitis addressed in this guideline include:
Which first-line investigations should be performed?; Which aetiologies should be
considered possible based on clinical features, risk factors and radiological features?;
What tests should be arranged in order to diagnose the common causes of encephalitis?;
When to consider empiric antimicrobials and immune modulatory therapies?; and What
is the role of brain biopsy?
Introduction
Encephalitis is a complex condition caused by brain
inflammation that is challenging to manage. The diagnoFunding: P. N. Britton is personally funded by NHMRC postgraduate scholarship 2014 (APP1074547), the Royal Australasian College of Physicians (RACP), Sydney Medical School and
the Marie Bashir Institute (MBI), the University of Sydney and
the Arkhadia Fund/Norah Theresa Hayes-Ratcliffe Fellowship.
Conflict of interest: None.
sis is rarely confirmed by brain biopsy and instead is
inferred by the presence of acute central nervous system
(CNS) dysfunction, fever and/or inflammation in the cerebrospinal fluid (CSF) and/or on neuroimaging.1 Differentiation from encephalopathy due to other causes is
difficult. There is a wide variety of presentations and a
myriad of possibile aetiologies but in most cases a cause is
not identified.2–4 There is often no definitive treatment1,5
and a high rate of mortality and morbidity.6 The investigation and management of encephalitis worldwide are of
© 2015 Royal Australasian College of Physicians
563
Britton et al.
variable quality.7–9 While several international guidelines
exist10–13 and the International Encephalitis Consortium
consensus included Australian authors,14 a concise guideline for Australian and New Zealand clinicians is required
due to differences in the epidemiology of encephalitis.
Box 2 Selected differential diagnoses
of suspected meningo-encephalitis
Meningitis without parenchymal involvement: bacterial, viral,
other (e.g. TB, cryptococcus).
Cerebral abscess and other forms of intra-cranial suppuration.
Methods
We reviewed the literature and sought expert opinions in
the development of the Consensus guidelines. The guidelines were peer reviewed by the Australasian Society for
Infectious Diseases Encephalitis Special Interest Group
and Guidelines Committee, the Public Health Association
of Australia (PHAA), the Australian and New Zealand
Association of Neurologists (ANZAN), and the Australasian College of Emergency Medicine (ACEM).
Epidemiology
In Australia, the annual hospitalisation rate for encephalitis has been calculated as 5.2/100 000 and case fatality
rate is estimated to be 4.6%.4 The highest admission
rates are observed in males, and those aged less than 9 or
over 60 years of age.4 This is similar to international
findings.15–17
Case definition
Infection associated encephalopathy (e.g. septic encephalopathy,
acute nectrotising encephalopathy (ANE)).
Vascular disease: ischaemic/haemorrhagic cerebro-vascular
accident (CVA), cerebral vasculitides (e.g. systemic lupus
erythematosus).
Hypertensive encephalopathy including posterior reversible
encephalopathy syndrome (PRES).
Neoplastic: primary
malignancies.
CNS
or
metastatic,
haematologic
Toxin induced encephalopathy: alcohol, illicit drugs, other drugs
(especially neuroleptics, cyclosporin).
Metabolic encephalopathy: hepatic, renal, hypoglycaemia,
hyponatraemia, hypocalcaemia, thiamine deficiency, Wilson
disease.
Neurodegenerative: fronto-temporal dementia, Creutzfeld-Jacob
disease (other prion disease), neuroacanthocytosis.
Demyelinating disease: multiple sclerosis (MS), neuromyelitis
optica (NMO or Devic disease).
Endocrine: Hashimoto’s encephalopathy/steroid
encephalopathy associated with autoimmune
(SREAT), Addisonian crisis.
responsive
thyroiditis
Psychiatric: psychosis, catatonia.
The international case definition of encephalitis
(Box 1)10 requires the presence of altered mental status
Seizure disorder.
Traumatic brain injury.
Intussusception.
Box 1 Encephalitis case definition
from the international encephalitis
consortium14
Major Criterion (required):
Patients presenting to medical attention with altered mental
status – defined as decreased or altered level of consciousness, or
lethargy or personality change – lasting ≥24h.
Minor Criteria (2 for possible encephalitis; ≥3 for probable or
confirmed encephalitis):
1 Documented fever ≥38°C (100.4°F) within the 72h before or
after presentation.
2 Generalised or partial seizures not fully attributable to a preexisting seizure disorder.
3 New onset of focal neurologic findings.
4 CSF WBC count ≥5/mm3.
5 Abnormality of brain parenchyma on neuroimaging suggestive
of encephalitis that is either new from prior studies or appears
acute in onset.
6 Abnormality on electroencephalography that is consistent
with encephalitis and not attributable to another cause.
AND Exclusion of encephalopathy caused by trauma, metabolic
disturbance, tumour, alcohol abuse, sepsis and other noninfectious causes.
lasting at least 1 day, and exclusion of encephalopathy
from other causes (Box 2). Confirmed diagnosis requires
meeting additional criteria such as CSF pleocytosis,
neuroimaging and electroencephalography (EEG)
changes consistent with encephalitis, and the presence
of seizures and new onset of focal neurological signs. Of
note, in individual cases, expected features of encephalitis such as headache, fever and CSF pleocytosis may be
absent.14
Aetiology
Multiple infectious agents have been associated with
encephalitis, but the syndrome is an uncommon manifestation of most. Viruses are the most commonly identified agent in all settings.5 Immune-mediated aetiologies
are increasingly recognised in up to one third of cases,
and are important because they are often treatable
(Box 3).
© 2015 Royal Australasian College of Physicians
564
Consensus guidelines for encephalitis
Box 3 Selected immune-mediated encephalitides
ADEM18,19
Acute disseminated encephalomyelitis is an inflammatory, multi-focal, demyelinating condition of the central nervous system. It presents
with encephalopathy and multi-focal neurological deficits. It is most common in children (mean age 5–8 years old), with a slight male
predominance. Rarely it may occur in adults. A temporal association following infection or, less commonly, vaccination is often identified.
Magnetic resonance imaging (MRI) is central to the diagnosis. Features include multi-focal, high signal lesions most evident on T2 weighted
and fluid-attenuated inversion recovery (FLAIR) sequences involving the sub-cortical, central and periventricular white matter and deep
grey matter. Approximately one quarter of children with ADEM will have serum antibodies to myelin oligodendrocyte glycoprotein (MOG).
Persistence of anti-MOG IgG is associated with recurrent central nervous system demyelination in this group. Corticosteroids are the
established first-line therapy, with other immune-modulatory therapies used in refractory cases. Acute haemorrhagic leuco-encephalopathy
(AHLE) is a rare, hyper-acute form of ADEM that overlaps with cerebral vasculitis.
Anti-NMDAR20–22
Anti-N-methyl-D-aspartate receptor encephalitis has been shown to be one of the principal causes of encephalitis in recent large prospective
studies. It typically presents with psychiatric symptoms, seizures, memory loss and mutism. The syndrome evolves to include movement
disorders, dysautonomia and sometimes hypoventilation. Although initially described as a para-neoplastic disorder with ovarian teratoma
in young adults (usually female), this tumour association is uncommon in young children where the female gender predominance is also
less pronounced. MRI is most often normal. It is diagnosed by identifying CSF or serum antibodies against the NR1 subunit of the NMDA
receptor. Anti-NMDAR can be identified in a proportion of relapsing HSV encephalitis, in particular if associated with chorea. Immunomodulatory therapy improves outcomes.
Anti-VGKC23–26
Anti-voltage-gated potassium channel-complex (including antibodies against leucine-rich glioma-inactivated 1 protein (Lgi1) and
contactin-associated protein 2(Caspr2)) encephalitis includes a broad clinical spectrum. In adults it typically presents in older male (>40 yrs)
patients with ‘limbic encephalitis’; sub-acute evolution of memory loss, confusion, medial-temporal lobe seizures and psychiatric features;
hyponatraemia is common. Lgi1 antibodies are often identified and it is rarely associated with malignancy. In children it presents as
temporal lobe focal seizures, status epilepticus and encephalopathy (behavioural disturbance, hallucinations) and cognitive decline. Specific
Lgi1 or Caspr2 antibodies may not be identified. Diagnosis is by identifying serum antibodies that bind to the VGKC-complex, although low
titre antibodies are of questionable significance. Immuno-modulatory therapy should probably be similar to NMDAR encephalitis although
there is less evidence.
Para-neoplastic ‘limbic encephalitis27,28
‘Limbic encephalitis’ (see above for clinical features) occurring in adults is often associated with malignancy. The encephalitis may occur
prior to the diagnosis, or during the course of cancer treatment. The tumours most often associated with limbic encephalitis are small cell
lung carcinomas (SCLC), testicular germ cell tumours, breast cancer, ovarian teratoma, Hodgkin lymphoma and thymoma. The most
commonly identified antibodies in this group are against intracellular neuronal antigens: anti-Hu, anti-Ma2(Ta), anti-CV2/CRMP5, and
anti-amphiphysin. A spectrum of neurological syndromes may overlap with ‘limbic encephalitis’ including features of brainstem encephalitis, basal ganglia syndromes, cerebellar ataxia and peripheral neuropathies. Specific antibodies associate with specific tumours, clinical
features and neurological outcome; for example, anti-Hu with SCLC, isolated limbic encephalitis and poorer prognosis, and anti-Ma2 with
testicular tumour, brainstem features and better prognosis. Treatment is directed towards the underlying tumour; immuno-modulatory
treatments are often used adjunctively.
Other
Increasing numbers of serum auto-antibodies are being associated with paraneoplastic and non-paraneoplastic limbic encephalitis. These
include: anti-Ri, anti-Yo, anti-glutamic acid decarboxylase (GAD), anti-gamma-amino-butyric acid B receptor (GABA-B-R), anti-alphaamino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA-R), anti-glycine receptor (GlyR), anti-dipeptidyl-peptidase-like
protein-6 (DPPX), anti- metabotropic glutamate receptor 5 (mGlu-R5).29 An algorithm addressing approaches to testing and management
has been recently published.30
These include acute disseminated encephalomyelitis
(ADEM), primarily seen in children and antibodymediated encephalitides (e.g. anti-N-methyl-D-aspartate
receptor (NMDAR) and anti-voltage-gated potassiumchannel (VGKC) complex).17,18,20
Aetiology varies with age, immune status, geography,
climate and pathogen endemicity, and has changed over
time due to changes in immunisation, changing behaviours (Box 4), improved testing and discovery of novel
aetiologies. Herpes simplex virus (HSV), varicella zoster
virus (VZV), toxoplasma, ADEM and enteroviruses
are the most commonly identified encephalitides from
studies based on hospital admission records. These
studies also demonstrate that deaths from toxoplasmosis,
HSV and measles-related encephalitis and subacute sclerosing panencephalitis (SSPE) have declined in recent
decades.4,31
A confirmed laboratory diagnosis is frequently not
obtained. Almost 70% of cases in a retrospective Australian study did not have an identified aetiology, although
© 2015 Royal Australasian College of Physicians
565
Britton et al.
Box 4 Clinical, risk factor and radiologic pointers to direct targeted (second line)
investigation
Clinical features
• Psychosis, movement disorder, hypoventilation: anti-NMDAR.
• Cognitive dysfunction, seizures: anti-VGKC, anti-NMDAR, HSV, HHV6, anti-GAD, anti-Hu, anti-Ma (other antibody- mediated see
Box 3).
• Subacute behavioural/personality change: HSV, anti-NMDAR, anti-VGKC, HIV, Treponema pallidum (syphilis), Whipple disease, trypanosomiasis#, SSPE, anti-GAD, anti-Hu, anti-Ma (other antibody mediated see Box 3).
• Hydrophobia, hypersalivation, delerium: rabies, ABLV.
• Parkinsonian features: flaviviruses (esp. JEV), anti-DR2 (basal ganglia encephalitis).
• Brainstem dysfunction: enteroviruses (esp. Ev71), flaviviruses (esp. MVEV, JEV, KUNV), Nipah#, Listeria monocytogenes, Burkholderia
pseudomallei, MTB, anti-Hu, anti-Ma (other paraneoplastic see Box 3).
• Associated limb weakness (flaccid paralysis) or tremor: enteroviruses (esp. Ev71, poliovirus), flaviviruses.
• Rash: enteroviruses, VZV, HHV6, measles , dengue, rickettsiae, Neisseria meningitidis (Meningococcus).
• Associated pneumonia: Mycoplasma pneumoniae, influenza, Nipah#, Hendra, Coxiella burnetii (Q fever).
• Parotitis, testicular pain: mumps.
• Cervical lymphadenopathy: EBV, CMV.
• SIADH: anti-VGKC, SLEV†.
• Chronic symptoms: HIV, JCV, BKV, trypanosomiasis#, SSPE, T. pallidum (syphilis), Whipple disease.
• Cranial nerve palsy: neuroborreliosis†.
Risk factors
• Neonate (<4 weeks): HSV-2, CMV, toxoplasmosis, T. pallidum (syphilis), L. monocytogenes, enteroviruses parechovirus.
• Infant/Child: HSV, VZV, enteroviruses, HHV6/7, M. pneumoniae, EBV, parechovirus, Bartonella sp., ADEM.
• >60 years: L. monocytogenes, VZV, HSV.
• Female: anti-NMDAR.
• Immunocompromised patient: HHV6, CMV, EBV, measles, VZV, LCMV, toxoplasma, cryptococcus, JCV, BKV, Bartonella sp.
• Tropical Australia: JEV, dengue, MVEV, KUNV, B. pseudomallei.
• Travel history‡
• Asia: JEV, dengue, malaria, MTB, Nipah, Angiostrongylus cantonensis.
• Pacific: JEV, dengue, malaria, MTB, Angiostrongylus cantonensis.
• North America: WNV, LACV, SLEV, CTFV, EEEV, neuroborreliosis, Rickettsia rickettsii (RMSF), ehrlichiosis (HME), anaplasmosis (HGA),
babesiosis, coccidiomycosis.
• South America: WNV, VEEV, dengue, MTB, trypanosomiasis (Chagas).
• Europe: TBEV, TOSV, neuroborreliosis, anaplasmosis (HGA).
• Africa: malaria, trypanosomiasis, MTB.
• Animal exposure
• Monkeys: herpes B, rabies†.
• Bats: rabies†, ABLV.
• Dogs and other canids outside Australia: rabies.
• Cats: Bartonella hensellae.
• Horse: Hendra, KUNV.
• Rodents: LCMV, leptospirosis.
• Snails/other moluscs: Angiostrongylus cantonensis.
• Swine: Nipah.
• Mosquito or Tick bite history.
• Arboviruses: MVEV, KUNV, JEV, dengue in Australia + by region.
• Rickettsiae: Rickettsia typhi, R. australis, R.honei, Orientalis tsutsugamushi in Australia + by region.
• Other: neuroborreliosis†, ehrlichiosis (HME)†, anaplasmosis (HGA)†.
• Recreational
• Sexually transmitted: HIV.
• Fresh water§: leptospirosis, Naegleria fowleri.
• Soil/mud§: Balamuthia mandrillis.
• Occupational
• Animal husbandry, farming: C. burnetii (Q fever), leptospirosis.
• Abbatoir workers: C. burnetii (Q fever).
• Unvaccinated: measles, mumps, rubella, VZV.
© 2015 Royal Australasian College of Physicians
566
Consensus guidelines for encephalitis
Box 4 Continued
Radiologic features
• Brainstem: enteroviruses (esp. Ev71), MVEV, JEV, WNV, nipah†, B. pseudomallei, L. monocytogenes, anti-NMO (anti-AQP4), anti-Hu,
anti-Ma (other paraneoplastic see Box 3).
• ‘Limbic’: HSV, HHV6, anti-NMDAR, anti-VGKC, anti-GAD, anti-Hu, anti-Ma (other antibody mediated see Box 3).
• Cerebellum: EBV, VZV, enteroviruses, M. pneumoniae.
• Subcortical grey matter (basal ganglia, thalami): EBV, flaviviruses (esp. JEV, MVEV), Influenza, MTB, post-streptococcal, M. pneumoniae,
anti-DR2.
• Frontal lobe: N. fowleri, B. mandrillis.
• ‘Vasculitic’: VZV, systemic lupus erythematosis (SLE) and other cerebral vasculitides.
• White matter lesions: ADEM, JCV-PML.
†If travelled to an endemic region. ‡Other important aspects of the travel history include the season (especially spring/summer for vector
borne pathogens) and specific activites engaged in. §In New Zealand natural geothermal pools pose a particular risk for amoebic
meningo-encephalitis, particularly those where there is the direct contact of the water with soil or run-off of water into the pool from soil.
ABLV, Australian bat lyssavirus; ADEM, acute disseminated encephalomyelitis; CMV, cytomegalovirus; CTFV, Colarado tick fever virus; DR2,
dopamine-2 receptor; EBV, Epstein–Barr virus; EEEV, eastern equine encephalitis virus; GAD, glutamic acid decarboxylase; HGA, human
granulocytotropic anaplasmosis; HHV6, human herpes virus-6; HIV, human immunodeficiency virus; HME, human monocytotropic
ehrlichiosis; HSV, herpes simplex virus; JCV, John Cunningham virus; JEV, Japanese encephalitis virus; KUNV, Kunjin virus; LACV, Lacrosse
virus; LMCV, lymphocytic choriomeningitis virus; MTB, Mycobacterium tuberculosis; MVEV, Murray valley encephalitis virus; NMDAR,
N-methyl-D-aspartate receptor; NMO, neuromyelitis optica (AQP4, aquaporin-4); RMSF, Rocky Mountain spotted fever; SLEV, St Louis
encephalitis virus; SSPE, sub-acute sclerosing panencephalitis; TBEV, tick bourne encephalitis virus; TOSV, Toscana virus; VEEV, Venezeulan
equine encephalitis virus; VGKC, voltage-gated potassium channel; VZV, varicella zoster virus; WNV, West Nile virus.
testing for immune-mediated and vector-borne causes
was limited.4 Rigorous implementation of systematic
testing will likely reduce this proportion but in many
cases the cause will remain unknown.17
In Australia, endemic viruses, including Hendra virus,
Australian bat lyssavirus (ABLV), Murray Valley encephalitis virus (MVEV) and West Nile virus (WNV) (Kunjin
clade (KUNV) – WNV/KUNV), should be considered as
possible aetiologies as well as regional infections such as
Japanese encephalitis virus (JEV), enterovirus 71 (EV71),
dengue and Nipah virus.32 Key differences to note when
applying this guideline in New Zealand are that there are
currently no endemic flaviviruses, nor are Hendra virus,
ABLV and Q fever endemic. Novel agents, particularly
viruses, or a changing geographical distribution of diseases
should be considered where unexplained encephalitis
clusters occur.
Causality
Experts agree that identification of an infectious agent
that is an established cause of encephalitis from a CNS
specimen is strong evidence of causality.33 Identification
of an encephalitic infectious agent outside of the CNS is
less conclusive. Identification of a specific antibody
response within the CSF in temporal association with an
episode of encephalitis is more convincing evidence of
causality than identification of a systemic antibody
response, especially on a single specimen. Causality may
be classified as confirmed/definite, probable or possible to
reflect the level of evidence achieved.2,3,14,17 Investigation
of patients may require specimens from multiple sites,
with repeated sampling for pathogen identification and
to identify a specific serologic response. This is necessary
to avoid missing treatable causes,17 especially where there
are two or more potential infectious agents and/or
autoantibodies.
Clinical assessment
We present two algorithms to assist clinicians with diagnosis and management. The first (Fig. 1) will assist clinicians to: identify possible meningoencephalitis patients,
consider differential diagnoses, initiate empiric acyclovir
and antibiotic therapy, and discriminate between patients
in whom encephalitis can be excluded from those where
a more rigorous assessment is required. Table 1 details
first-line investigations. The second algorithm (Fig. 2)
follows on from the first and is applied when encephalitis
is likely. It provides a multidisciplinary, staged approach
to investigation and management.
History
Collecting a comprehensive history is essential to enable
a diagnosis. The onset and evolution of altered consciousness, lethargy, cognition, behaviour or personality
change, seizures, weakness, abnormal movements and
© 2015 Royal Australasian College of Physicians
567
Britton et al.
Table 1 Recommended first-line investigation of encephalitis in Australia
and New Zealand
Specimen/Investigation
CSF†
Serum||
Respiratory
Faeces
Skin swabs (where
lesions present)
Neuroimaging
Tests
Opening pressure, microscopy, Gram stain
and bacterial culture
Cell count and type‡
Biochemistry: protein, glucose
PCR: HSV§, enterovirus, VZV
Antibodies: oligoclonal bands, VZV IgG¶
Antigen: cryptococcal Ag
Other: VDRL (adult); consider cytology; to
store
Serology: HIV††, flavivirus (Australia)‡‡,
M. pneumoniae, EBV (child/adolescent),
T. pallidum (syphilis – adult)
PCR testing for enterovirus, influenza A and
B, adenovirus
PCR or antigen testing for enterovirus,
adenovirus, rotavirus (child); enterovirus
culture/typing
PCR testing for HSV 1/2, VZV, enterovirus
MRI (sequences to include: T1,T2, FLAIR,
DWI, gradient-echo, gadolinium contrast)
or if unavailable CT with contrast
EEG
†Collect up to 10 mL, if able, in four tubes in adults and children, and up
to 5 mL in a small child (<2 years old). ‡Formal cytological examination is
required to reliably differentiate eosinophils from other leukocytes and
identify malignant cells. §HSV PCR is highly sensitive (>96%) between days
3 and 7 of the illness, its sensitivity decreases slightly in the second week
of the illness. False negatives prior to day 3 have been described. After
day 10, CSF HSV IgG can be used to make a late diagnosis. ¶Where
available, CSF VZV IgG may be more sensitive than PCR. Testing requires
the demonstration of intrathecal synthesis of VZV IgG, that is a reduced
serum/CSF ratio of VZV IgG compared with the serum/CSF ratio of
albumin. ||Collect up to 20 mL blood in a clotting tube in adults and
children; and up to 5 mL in a small child (<2 years old). ††HIV is very
uncommon in children in Australia, and encephalopathy is an uncommon
presentation; some experts would still undertake HIV testing as the diagnosis impacts upon possible aetiologies of encephalitis, and is treatable.
‡‡Flaviviral IgM should be tested after 5 days of symptoms. A negative
result makes the diagnosis unlikely. CSF IgM is specific for these viruses
and should be performed in patients in whom the diagnosis is likely in
terms of risk factors, clinical and radiologic features (see Boxes 4 and 5).
Ag, antigen; CSF, cerebro-spinal fluid; CT, computed tomography scan;
DWI, diffusion-weighted imaging; EEG, electro-encephalogram; FLAIR,
fluid-attenuated inversion recovery; HIV, human immunodeficiency virus;
HSV, herpes simplex virus; MRI, magnetic resonance imaging; PCR,
polymerase chain reaction; VZV, varicella zoster virus; WBC, white blood
cell.
altered sensation should be elicited and any localisation
(focality) recorded. Details of current or recent fever,
headache, rash or any other prodromal illness, and an
exposure history should be sought including contact with
sick persons, immunisation history, travel, mosquito, tick
or other insect bites, animal exposures (wild, farm or
domestic), and occupation and outdoor activities (e.g.
hiking, camping, water sport). Public health authorities
should be consulted about seasonal/epidemic activity of
infectious agents (e.g. flaviviruses and other arboviruses,
enteroviruses).36 Risk factors (Box 4) including age,
immunisation and immune status (e.g. immune suppressive treatment, immunodeficiency virus (HIV) risk
factors) should be considered.
Examination
Physical examination should include an objective assessment of the level of consciousness, and look for subtle
seizure activity, meningism, abnormal movements
(e.g. chorea, parkinsonism), weakness, sensory loss and
cranial nerve involvement (including deafness and
anosmia), noting any focal findings and for features suggesting other diagnoses (Box 2). Temperature and other
vital signs should be assessed for features of raised intracranial pressure or autonomic dysfunction. Mental status
examination should be recorded, particuarly if there are
psychotic features (hallucinations and delusions). A rash
or other skin lesions (e.g. bite marks, eschar, mouth/
palate ulcers, lymphadenopathy and shingles lesions),
respiratory or gastrointestinal signs may give clues to the
aetiology. Clusters of clinical features (e.g. psychosis and
movement disorder and anti-NMDAR encephalitis, or
hydrophobia, delirium and hypersalivation with rabies/
ABLV) (Boxes 3, 4) may be strongly indicative of a specific cause.
Investigations
First-line investigation of all patients with
suspected/probable encephalitis
Investigations to exclude differential diagnoses (Box 2)
and guide initial management are listed in Table 1 and
Figure 1. Blood cultures should be taken prior to the
administration of empiric antibiotics. A lumbar puncture
(LP) should be performed if there is no contraindication or following appropriate imaging and/or clinical
observation. CSF analysis is needed to confirm encephalitis (Fig. 1) and identify a cause. Sufficient volumes
should be sampled (Table 1) to enable microscopy and
cell counts, Gram stain, bacterial culture (mycobacterial
culture or fungal cultures if indicated), biochemistry
(protein, glucose) and exclusion of HSV, Cryptococcus,
VZV and syphilis in those patients meeting the more
rigorous definition of encephalitis (Fig. 2). Where
avaiable, other biomarkers of CNS inflammation including CSF oligoclonal bands37 and CSF neopterin38 should
© 2015 Royal Australasian College of Physicians
568
Consensus guidelines for encephalitis
be considered. A serum specimen (Table 1) should be
stored for testing with convalescent sera. All patients
(Fig. 2) should have serology for HIV, mycoplasma and
flaviviruses, and syphilis serology in adults and Epstein–
Barr virus (EBV) serology in children. A respiratory tract
specimen and stool for viral testing, and viral and bacterial swabs from any skin lesions should be collected.
CNS imaging (Fig. 1) should be performed on all
patients, by magnetic resonance imaging (MRI) wherever
possible using T1, T2 and fluid-attenuated inversion
recovery, diffusion-weighted imaging, gradient echo or
similar sequences and gadolinium contrast. A chest X-ray
is needed to detect associated lung disease (e.g. tuberculosis (TB) and Cryptococcus). EEG is highly sensitive in
encephalitis, but often non-specific. It is particularly
important in patients with chronic symptoms and those
with psychiatric presentations to identify encephalopathy
or to diagnose subtle seizure activity and non-convulsive
status epilepticus.11,12 Localised EEG activity may suggest
specific aetiologies (e.g. temporal localisation with HSV).
Targeted testing of patients with encephalitis
Where encephalitis is likely, second and third-line testing
of patients should be guided by risk factors, clinical
and radiologic features (Boxes 4, 5; Fig. 2) in consultation with specialists in neurology, infectious diseases,
microbiology/virology and neuroradiology. Remote consultations (by telephone) may be necessary, and transfer
to a referral centre considered.
Patient subgroups
Children
Encephalitis is challenging to identify in very young
infants as features are non-specific (lethargy, excessive
irritability, poor feeding). Diagnosis requires a high index
of suspicion and consultation with experienced clinicians. The most common causes of childhood encephalitis globally are HSV-1, VZV, enteroviruses and JEV in
endemic regions. Other causes include: ADEM, EBV and
adenovirus in children, and HSV-2 and parechovirus in
neonates. Human herpesvirus (HHV)-6 and HHV-7 may
be associated with febrile seizures and encephalopathy in
immune-competent children and may uncommonly
cause encephalitis in the immunocompromised.42
Mycoplasma pneumoniae has been associated with childhood encephalitis (less commonly in adults) when a positive M. pneumoniae IgM is detected in blood, although
causality remains controversial without concurrent
pathogen identification.33,43
Box 5 Tests of choice for the more
common and regionally important
aetiologies33,39–41
For an extensive review of indicated tests for other aetiologies see
Granerod et al., 2010 and Tunkel et al., 2008.10,33
Direct discussion with a medical microbiologist/virologist and
local laboratory scientist is good practice before ordering uncommon tests.
HSV: CSF PCR 3–10 days into illness. May be negative < 3 days,
repeat lumbar puncture and re-test CSF PCR (between days 3
and 10) if other features suggest HSV (see Fig. 2). CSF IgG (in
combination with serum IgG) > 10 days.
Enteroviruses: CSF PCR, stool and upper respiratory tract specimen for PCR/viral culture.
VZV: CSF PCR†, CSF IgG (in combination with serum IgG),
Acute serum IgM, serum IgG acute/convalescent‡.
EBV/CMV/HHV6: CSF PCR†, Acute serum IgM, serum IgG
acute/convalescent‡.
Flaviviruses§: CSF IgM after 5 days into illness, Acute serum
IgM, serum IgG acute/convalescent‡.
WNV/KUNV: As for other flaviviruses and CSF PCR.
Dengue: As for other flaviviruses and acute blood NS1
Antigen, PCR.
Measles: CSF IgM, acute serum IgM and acute/convalescent
serology, urine or upper respiratory specimen for PCR or antigen
testing.
Rabies or ABLV: CSF PCR. Serum and CSF IgG. DFA on saliva,
nuchal skin, corneal impression, brain.
Hendra or Nipah: PCR on CSF, serum, respiratory, urine specimens ± serum IgM/G.
Ab-mediated: Serum and/or CSF anti-NMDAR Ab; serum antiVGKC complex Ab; serum anti-Hu, anti-Ma2 Ab¶.
†Quantitave PCR may contribute to diagnosis. Exclusion of
HHV6 chromosomal integration may be required to confirm its
aetiologic role.
‡Convalescent for practical purposes is 2–4 weeks following
symptom onset.
§Includes: Japanese encephalitis virus (JEV), Murray valley
encephalitis virus (MVEV), West Nile virus/Kunjin virus (WNV/
KUNV), dengue, St Louis encephalitis virus (SLEV), tick-borne
encephalitis virus (TBEV). Other encephalitic arthropod-borne
viruses (arboviruses) are investigated in the same way including:
togaviruses (eastern equine encephalitis virus (EEEV), western
equine encephalitis virus (WEEV), Venezuelan equine encephalitis virus (VEEV)), bunyaviruses (Lacrosse virus (LACV), Toscana
virus (TOSV)), and reoviruses (Colorado tick fever virus (CTFV)).
¶Other serum antibodies that have been implicated in
paraneoplastic and non-para-neoplastic limbic encephalitis
include: anti-amphiphysin, anti-CV2/CRMP5, anti-Ri, anti-Yo,
anti-glutamic acid decarboxylase (GAD), anti-gamma-aminobutyric acid A and receptor B (GABA-A-R, GABA-B-R),
anti-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic
acid receptor (AMPA-R), anti-glycine receptor (GlyR), antidipeptidyl-peptidase-like protein-6 (DPPX), anti- metabotropic
glutamate receptor 5 (mGlu-R5).
CSF, cerebrospinal fluid; DFA, direct fluorescent antibody test;
HSV, herpes simplex virus; NMDAR, N-methyl-D-aspartate
receptor; PCR, polymerase chain reaction; VGKC, voltage-gated
potassium channel.
© 2015 Royal Australasian College of Physicians
569
Britton et al.
Who has ‘Suspected’ meningo-encephalitis?
An adult or child who presents with:
Encephalopathy: defined by some or all of the following features: altered level of consciousness, altered
cognition, altered personality/behaviour†, and lethargy‡;
In combination with: current or recent history of fever, and/or new onset seizures, and/or new onset focal
neurological signs/symptoms and/or headache.
*Conditions that may present
like meningo-encephalitis
- Meningitis
- Cerebral abscess
- Traumatic brain injury
- Encephalopathy:
Early investigation and Management
•
•
•
•
•
•
•
•
•
(See Box2 for detail)
Comprehensive history, including exposures, and examination (See box 4); Consider
other causes*
Bloods: full blood count, electrolytes, glucose, urea, creatinine, calcium, liver function tests, blood
culture, serum to store (5-10 mL). In adults consider early HIV testing with appropriate pre-test
Infection-associated
Toxin induced
Metabolic
Neurodegenerative
Seizure
Endocrine
Neoplastic
Psychiatric†
counselling.
•
•
•
•
Judicious fluid and electrolyte management as required.
Acute seizure management (follow local and state based guidelines).
Consider if Lumbar Puncture (LP) can be performed and role of preceding CT scan (see below).
If patient septic or LP or CT delayed§, commence antibiotics promptly as per national guidelines¶.
Performance of LP and role of prior CNS imaginggII
CNS imaging (most commonly CTΔ) should be performed prior to lumbar puncture in the following circumstances:
•
Impairment of consciousness; abnormal, fluctuating or declining GCS.
•
Signs of raised intra-cranial pressure (papilloedema, relative bradycardia with hypertension, oculomotor palsy or abnormal pupillary response).
•
Focal neurological deficits.
•
New onset seizures until stabilised.
•
Immunocompromised state (HIV/AIDS, immunosuppressive therapy, transplantation).
•
Previous history of a CNS lesion (mass lesion, stroke, or focal infection).
LP is relatively contra-indicated in the following circumstances:
•
Haemodynamic instability or acute respiratory failure.
•
Coagulation disorders e g disseminated intravascular coagulation use of anticoagulant drugs thrombocytopenia (<100 x106/L)
Contraindication to LP or LP deferred pending CNS
imaging or CT unavailable:
•
•
•
Bacterial meningitis possible. Commence antibiotics promptly§
for possible bacterial meningitis as per local and national
guidelines¶.
Encephalitis is possible, start acyclovir (see doses below).
Perform LP as soon as possible: if no radiologic
contraindication or reconsider daily while an inpatient.
Acyclovir dose:
Adults/Children >12 years:
10mg/kg IV 8 hourly.
Children:
<3 mo- 20mg/kg IV 8
hourly;
3mo-12years 500mg/m2
10mg/kg IV 8 hourly.
No clinical contraindication. LP performed:
•
•
Record CSF opening pressure.
Take 10mL if able (5mL in a small child <15kg); ideally in 4 tubes (1 - 2.5mL
in 4th tube).
Send for microscopy (cell count, Gram stain), culture and biochemistry
(protein, glucose), HSV and enterovirus PCR.
Check CSF microscopy values (see below).
•
•
adjust dose for renal
impairment.
CSF microscopy abnormal
•
Commence antibiotics for possible bacterial
meningitis as per local and national
guidelines2.
•
Encephalitis is possible, start acyclovir.
•
Consider tuberculous or fungal meningitis
depending on CSF parameters (see below) and
risk factors.
•
Arrange CNS imaging : MRI within 24-48h
(CT with contrast if MRI unavailable).
MRI a nd/or CSF abnormalconsistent with encephalitis (see below)
and no other diagnosis made.
Encephalitis probable
(See Figure 2)
MRI normal and CSF normal, but
clinical findings persist, and no
other diagnosis made.
Encephalitis remains possible.
(See Figure 2)
***Typical cerebro-spinal fluid patterns††11,34,35
Normal
Viral meningoencephalitis
Opening pressure
<25cm H20
Normal-high
<5
5–1000
Cell count (uL or
106/L)
Cell type
Lymphocyte/m Lymphocytes
onocytes, no
predominate
neutrophils or
(neutrophils if
RBC
early)
>0.5
Normal
Glucose
(CSF:plasma)
(mmol/L)
Protein (g/L)
<0.5
0.5–1.0
CSF microscopy normal
•
Bacterial / tuberculous / fungal meningitis
excluded.
•
Encephalitis remains possible, start acyclovir.
•
Arrange CNS imaging : MRI within 24-48h
(CT with contrast if MRI unavailable).
MRI normal, CSF normal and
clinical findings resolved.
Encephalitis excluded.
Manage as per alternative
diagnosis.
Alternative diagnosis made.
Encephalitis excluded.
Manage as per alternative
diagnosis.
Bacterial
meningitis
High
100–50 000
Tuberculous
meningitis
High
5–500
Fungal meningitis
Neutrophils
predominate
Lymphocytes
predominate
Lymphocytes
predominate
Low (<0.4)
Very Low (<0.3)
Normal-low
>1.0
1.0–5.0
0.2–5.0
Very High
5–1000
© 2015 Royal Australasian College of Physicians
570
Consensus guidelines for encephalitis
Figure 1 Algorithm for the assessment and management of a patient with suspected meningo-encephalitis.
Where traumatic sampling occurs with elevated red blood cells on microscopy, the WBC can be corrected using the formula: True CSF WBC = actual CSF
WBC – (WBC blood × RBC CSF/RBC blood) or approximately 1 WBC per 500 RBC. The ratio of WBC types in the CSF can be compared with that in blood.
1000 × 106/L RBC in CSF raises CSF protein by approximately 0.1 g/L. †Particular note should be made of apparently psychiatric presentations. ‡In young
children, the clinical features of encephalopathy may be difficult to discern and may include poor feeding, excessive irritability and unusual crying. §The
Australasian College of Emergency Medicine (ACEM) recommends that antibiotics may be delayed if the lumbar puncture will be performed within 20 min
of presentation. Antibiotics should be administered prior to LP where: there is no doctor present, there will be a delay to a required CT, lumbar puncture
is not able to be performed (due to other contraindication or the healthcare professional does not have the requisite skills) or possible systemic sepsis.
¶Consult Therapeutic Guidelines: Antibiotic: Version 15. Therapeutic Guidelines Limited, Melbourne (2014). ||If CT or MRI are unavailable locally, consultation with specialists in neurology and infectious diseases should be pursued and collaborative discussion as to the need for transfer to a referral centre
should be included in these discussions. Δ Early MRI should be advocated because of its increased sensitivity, particularly in children where stroke is less
common. ††There are exceptions to these ‘typical patterns’; infectious diseases consultation should be sought where particular risk factors or clinical
features suggest a specific aetiology and CSF findings are inconsistent with this. CNS, central nervous system; CSF, cerebrospinal fluid; CT, computed
tomography scan; GCS, Glasgow coma score; HIV/AIDS, human immunodeficiency syndrome/acquired immune deficiency syndrome; LP, lumbar puncture; MRI, magnetic resonance imaging; RBC, red blood cell; WBC, white blood cell.
◀
Immunocompromised hosts
The aetiology of encephalitis in the immunocompromised varies depending on the timing, nature and
intensity of immunosuppression. CSF pleocytosis may be
lacking in these patients. CNS reactivation of latent infection (e.g. VZV, cytomegalovirus (CMV), HHV-6, EBV) can
occur, but is less common than systemic reactivation.
HHV-6 post-transplant limbic encephalitis is now well
described.44,45 VZV reactivation and primary infection in
immunocompromised hosts causes a small vessel vasculitis. Encephalitis may be caused by opportunistic pathogens (e.g. Toxoplasma, Cryptococcus). HIV testing is essential
as encephalitis may be the presenting illness of HIV/
AIDS. A variety of neurological syndromes is associated
with HIV; patients at highest risk are those with severe
immune suppression (CD4 count < 200). Toxoplasma
gondii, Cryptococcus neoformans and CMV are the most
important pathogens. John Cunningham virusassociated progressive multifocal leucoencephalopathy
(PML) can present in a variety of ways including fulminant encephalopathy.46,47 Initiation of anti-retroviral
therapy can result in CNS immune reconstitution inflammatory syndrome (IRIS) encephalitis, including a nonpathogen-associated CNS IRIS, so-called CD8-positive
encephalitis.48 The role of corticosteroids in this group
should be discussed with an HIV specialist.
International travellers or immigrants
Overseas travellers may be exposed to a wide array of
infections that can cause encephalitis. Common aetiologies as well as the exotic should be considered. Testing
should be guided by a detailed history including timing
of symptom onset in relation to travel, destination and
in-location movements and activities, pre-departure
immunisation, antimicrobial prophylaxis and adherence,
animal and vector exposure, and ingestion of raw or
unusual foods. Cerebral malaria is a potential cause in the
febrile returned traveller with encephalopathy. Tuberculous meningoencephalitis should be considered, especially in young children and vector-borne pathogens (e.g.
flaviviruses, Rickettsia spp.) after travel to overseas rural
locations, especially in summer/spring. A history of insect
bites is not always given.
Tropical Australia
In those living in or returning from tropical Australia,
dengue, JEV and the endemic flaviviruses (MVEV and
KUNV) should be considered. Parts of the Northern Territory (above 21oS) are endemic for melioidosis, which
can present as brainstem encephalitis with cranial nerve
palsies and limb weakness.49 Leptospirosis may occur following flooding events and exposure to water.50
Unknown or ‘cryptic’ encephalitis
Brain biopsy is not necessary in most encephalitis
patients; however, it should be considered in patients
without a diagnosis who remain unwell or are deteriorating, especially if there are focal lesions on imaging or
where CNS vasculitis is suspected.51 Potentially treatable
aetiologies may be diagnosed using biopsy in these
circumstances,51–54 and other occult diagnoses can be
made, for example, CNS Whipple disease, TB, PML
and neurosarcoidosis.52 Liaison with histopathology and
microbiology prior to sampling is essential to ensure
correct specimen handling, transport and testing.
A proportion of patients will not have an aetiological
diagnosis made despite extensive investigation. Patients
© 2015 Royal Australasian College of Physicians
571
Britton et al.
Encephalitis(14)
An adult or child with:
1.
Encephalopathy: defined by the presence of some or all of the following features:
altered level of consciousness, altered cognition, personality/behavioural change,
lethargy; lasting >24 h.
2.
In combination with two or more of the following:
•
Fever or history of fever (>38C) within 72h before/after presentation.
•
Generalised or partial seizures not fully attributable to a pre-existing seizure disorder.
•
New onset focal neurologic findings.
•
CSF pleocytosis (≥ 5WBC/uL).
•
Abnormal results of neuroimaging suggestive of encephalitis.
•
EEG abnormality consistent with encephalitis and not attributable to another cause.
3.
AND no alternative cause identified/diagnosis made.
Arrange first-line investigations if not performed already (see also Table 1):
Commence empiric acyclovir:
•
Adults/Children >12 years: 10mg/kg IV 8
hourly.
Children: <3 mo- 20mg/kg IV 8 hourly;
3mo-12years 500mg/m2 10mg/kg IV 8 hourly.
CSF: microscopy, culture, protein, glucose, HSV PCR, enterovirus PCR, VZV PCR and IgG,
cryptococcal Ag, VDRL (adult), consider cytology.
•
Serology: HSV, flavivirus (Australia), HIV, M. pneumoniae, EBV (child/adolescent), Treponema
pallidum (Syphilis - adult)
•
Respiratory viral testing: PCR or antigen testing for enterovirus, influenza A and B,
adenovirus.
•
Stool viral testing: PCR or antigen for enterovirus, adenovirus, rotavirus (child).
•
MRI brain: sequences to include T1,T2, FLAIR, DWI, gradient-echo, gadolinium contrast or if
unavailable CT with contrast.
•
Chest X-ray.
•
EEG (particularly useful in certain circumstances†).
Consider addition of empiric antimicrobials for Listeria monocytogenes (penicillin or ampicillin) and
rickettsiae (doxycycline in adults)
adjust dose for renal impairment.
Arrange specialist consultation:
Neurology
Infectious Diseases
Microbiology/Virology
Radiology
Identify clinical features, risk factors, radiologic features (see Box 4) to guide second-line investigation:
•
•
In consultation with neurologist, infectious diseases specialist, radiologist, microbiologist (Box 5).
Especially note the following risk groups:
o
Children and neonates
o
Immunocompromised (including HIV/AIDS, immunosuppressive therapy, transplantation)
o
International travellers or immigrants
o
Those residing in or having travelled to tropical regions of Australia
Consider if HSV encephalitis is excluded and therefore determine duration of acyclovir therapy:
1.
2.
In a patient without neuroimaging suggestive of HSV encephalitis‡, cease empiric acyclovir if:
o
A negative CSF PCR for HSV is obtained, if the CSF was sampled between days 3 and 7 of the clinical illness.
o
Two negative CSF PCRs for HSV are obtained, if the first PCR was taken in the first 72 h of the clinical illness.
In a patient with neuroimaging suggestive of HSV encephalitis‡, irrespective of CSF PCR result:
o
Continue acyclovir for 21 days if < 3 mo; 14-21 days if >3 mo, child or adult and,
o
Consider CSF HSV IgG testing after day 10 of the clinical illness (unless a definitive alternative diagnosis made).
Definitive treatment of aetiology if identified
(see Box 6)
Where relevant, report case to public health or other
statutory authorities and perform contact tracing
If no aetiology identified, consider empiric treatment of
possible aetiologies, including immune therapy
(corticosteroids and/or IVIG) based on clinical features,
risk factors, radiologic features in consultation with
neurologist and infectious diseases specialist (see Box
6).
Consider third-line investigations if:
1.
The patient remains unwell and other investigations are negative or
2.
The patient is deteriorating and an aetiologic diagnosis has not yet been made.
•
•
Repeat CSF sampling: microscopy, CSF wet mount, cytology, repeat HSV PCR, CSF immunoglobulin testing (HSV, VZV, flavivirus, IgG index, ABLV
(Australia) and other epidemiologically relevant viruses if international travel).
Repeat MRI brain: sequences to include T1,T2, FLAIR, DWI, gradient-echo, gadolinium contrast. It is essential to liaise with a (neuro)radiologist with
regards to planning these and any additional sequences.
•
•
All patients in this circumstance should be tested for anti-NMDAR, anti-VGKC and in Australia, ABLV.
Brain biopsy: it is essential to liaise with histopathology and microbiology prior to sampling with regards to specimen handling, transport and testing –
especially note that specimens should not be formalin fixed prior to transfer to the laboratory.
© 2015 Royal Australasian College of Physicians
572
Consensus guidelines for encephalitis
Figure 2 Algorithm for the investigation and management of encephalitis.
†Patients with chronic symptoms, those with primarily psychiatric presentations or to diagnose subtle seizure activity and non-convulsive status
epilepticus. ‡Imaging findings characteristic of HSV are medial temporal lobe and inferior frontal cortex involvement; lesions may be unilateral or bilateral.
Ab, antibody; ABLV, Australian bat lyssavirus; Ag, antigen; AIDS, acquired immunodeficiency syndrome; CSF, cerebrospinal fluid; CT, computed tomography scan; DWI, diffusion weighted index; EEG, electroencephalogram; HIV, human immunodeficiency virus; HSV, herpes simplex virus; mo, months;
NMDAR, N-methyl D aspartate receptor; PCR, polymerase chain reaction ; VZV, varicella zoster virus; WBC, white blood cell; yo, years old; VGKC, voltage
gated potassium channel; MRI, magnetic resonance imaging; FLAIR, fluid attenuated inversion recovery.
◀
Box 6 Directed management of viral
and immune-mediated encephalitis
For an extensive review of antimicrobial treatments for other
aetiologies see Tunkel et al., 2008.10
HSV: Minimum 14 days intravenous acyclovir for immunocompetent patients and 21 days for immunocompromised patients
(adults and children > 12 years: 10 mg/kg 8 hourly; children:
<3 mo 20 mg/kg 8 hourly; 3 mo-12 yo 500 mg/m2 8 hourly).
Consider repeat lumbar puncture for CSF HSV PCR at planned
completion of treatment especially in immunocompromised and
children.
VZV: Consider 7–14 days intravenous acyclovir (adults and children > 12 years: 10 to 12.5 mg/kg 8 hourly; children: 500 mg/m2
8-hourly (approximately 20 mg/kg for child 5 years or less,
15 mg/kg for child 5–12 years)) with or without corticosteroids in
consultation with an infectious diseases specialist.
Enterovirus: Intravenous immunoglobulin if hypogammaglobulinaemic. Intravenous immunoglobulin is used widely in
Asia for enterovirus 71.
CMV/HHV6: Reduce immunosuppression and consider
ganciclovir and/or foscarnet in consultation with infectious
diseases specialist.
Rabies or ABLV: Consider Milwaukee protocol55,56 in consultation with infectious diseases specialist.
ADEM: Methylprednisolone 30 mg/kg daily in children up to
1000 mg (adult daily dose) for 3–5 days in consultation with
a neurologist. Second-line treatments in consultation with a
neurologist.
Ab-mediated: Immunosuppressive therapy in consultation
with a neurologist. Investigation for underlying tumour and
removal (where indicated). Ongoing tumour surveillance.
ADEM, acute disseminated encephalomyelitis; CMV, cytomegalovirus; CSF, cerebrospinal fluid; DFA, direct fluorescent antibody test; HHV, human herpes virus; HSV, herpes simplex virus;
VZV, varicella zoster virus.
with ‘cryptic’ encephalitis should be tested for antiNMDAR and anti-VGKC complex encephalitis and in
Australia for ABLV.
Management (Figs 1,2; Box 6)
Supportive and empiric
Seizure control, management of raised intracranial pressure (occasionally by surgical decompression), circulatory
and respiratory support, fluid and electrolyte balance,
nutritional support, skin integrity and pressure area care,
and prevention of hospital-acquired infections should all
be addressed.
Patients with ‘suspected meningo-encephalitis’ should
be commenced on acyclovir. Antibiotics for possible meningitis or sepsis should be administered promptly as per
local and national guidelines (ACEM advise within
20 min of presentation) and should not be delayed if LP
is contraindicated or neuroimaging delayed.
Directed management of encephalitis with an
identified aetiology (Box 6)
When a cause is identified, directed therapy (Box 6)
should be determined in consultation with relevant
specialists and national/international antimicrobial
guidelines.10
With the exception of the herpesviruses, most viral
causes have no specific treatment. For HSV and VZV
encephalitis, guidelines regarding acyclovir duration
and corticosteroids vary,57–64 as does advice regarding
antivirals for CMV or HHV-6 encephalitis.10 Antivirals
are not recommended for EBV encephalitis.10 Pleconaril
for enteroviral encephalitis has limited documented efficacy and is not widely availabile. Intravenous immunoglobulin is used, without strong evidence of efficacy, to
treat EV71-associated encephalo-myelitis,65 and also for
chronic enteroviral infections in antibody deficient
hosts. It is increasingly being used as adjunctive therapy
for other encephalitides. Corticosteroids have an established role in the management of ADEM, although this
is not based on high-quality evidence18,66; intravenous
immunoglobulin and plasma exchange may be used
where there is steroid resistance.18 Evidence of benefit
from immune suppression in NMDAR encephalitis in
increasing21 and similar approaches are recommended
for other immune-mediated encephalitides.67 An extensive search for an underlying malignancy should be performed whenever NMDAR encephalitis is diagnosed68
and in adults with ‘limbic encephalitis’69 (Box 3). There
is no evidence that antimicrobials are beneficial in
M. pneumoniae-associated encephalitis.
© 2015 Royal Australasian College of Physicians
573
Britton et al.
Outcome, prognosis and follow-up
Overall mortality of encephalitis is approximately
10%.2,3,17 Up to 50% of patients experience short-term
deficits with 20% experiencing severe sequelae; longterm outcome is poorly characterised, and neurocognitive sequelae likely underestimated.70,71 Depression
of consciousness at presentation is the main adverse
prognostic feature; poor outcome has also been associated with refractory status epilepticus, intensive care unit
admission, focal neurologic signs, abnormal MRI findings, extremes of age and immune compromise, a diagnosis of HSV in adults, and JEV or Mycoplasma pneumoniae
in children,5 or delay in the initiation of directed
therapy.
References
1 Lewis P, Glaser CA. Encephalitis. Pediatr
Rev 2005; 26: 353–63.
2 Mailles A, Stahl J-P, Steering C, Steering
Committee and Investigators Group.
Infectious encephalitis in France in
2007: a national prospective study. Clin
Infect Dis 2009; 49: 1838–47.
3 Glaser CA, Honarmand S, Anderson LJ,
Schnurr DP, Forghani B, Cossen CK
et al. Beyond viruses: clinical profiles
and etiologies associated with
encephalitis. Clin Infect Dis 2006; 43:
1565–77.
4 Huppatz C, Durrheim DN, Levi C,
Dalton C, Williams D, Clements MS et al.
Etiology of encephalitis in Australia,
1990–2007. Emerg Infect Dis 2009; 15:
1359–65.
5 Granerod J, Crowcroft NS. The
epidemiology of acute encephalitis.
Neuropsychologists 2007; 17: 406–28.
6 Khetsuriani N, Holman RC, Anderson
LJ. Burden of encephalitis-associated
hospitalizations in the United States,
1988–1997. Clin Infect Dis 2002; 35:
175–82.
7 Huppatz C, Gawarikar Y, Levi C, Kelly
PM, Williams D, Dalton C et al. Should
there be a standardised approach to the
diagnostic workup of suspected adult
encephalitis? A case series from
Australia. BMC Infect Dis 2010; 10:
353.
8 Kneen R, Jakka S, Mithyantha R,
Riordan A, Solomon T. The
management of infants and children
treated with aciclovir for suspected viral
encephalitis. Arch Dis Child 2010; 95:
100–6.
Recovery from encephalitis reaches a plateau at
approximately 6–12 months. Rehabilitation assessment
(medical and non-medical) should be considered, especially in those with neurological or neuropsychological
deficits at discharge. We recommend early formal discharge planning to facilitate referrals and follow-up
including development and learning in children, and
seizure management.
Conclusion
Further research is needed to inform better local management guidelines; however, many patients will
benefit from the optimal application of existing
knowledge.
9 Kelly C, Sohal A, Michael BD, Riordan
A, Solomon T, Kneen R. Suboptimal
management of central nervous system
infections in children: a multi-centre
retrospective study. BMC Pediatr 2012;
12: 145.
10 Tunkel AR, Glaser CA, Bloch KC, Sejvar
JJ, Marra CM, Roos KL et al. The
management of encephalitis: clinical
practice guidelines by the Infectious
Diseases Society of America. Clin Infect
Dis 2008; 47: 303–27.
11 Kneen R, Michael BD, Menson E,
Mehta B, Easton A, Hemingway C et al.
Management of suspected viral
encephalitis in children – Association of
British Neurologists and British
Paediatric Allergy, Immunology and
Infection Group National Guidelines.
J Infect 2012; 64: 449–77.
12 Solomon T, Michael BD, Smith PE,
Sanderson F, Davies NW, Hart IJ et al.
Management of suspected viral
encephalitis in adults – Association
of British Neurologists and British
Infection Association National
Guidelines. J Infect 2012; 64:
347–73.
13 Steiner I, Budka H, Chaudhuri A,
Koskiniemi M, Sainio K, Salonen O et al.
Viral meningoencephalitis: a review of
diagnostic methods and guidelines for
management. Eur J Neurol 2010; 17:
999–e57.
14 Venkatesan A, Tunkel AR, Bloch KC,
Lauring AS, Sejvar J, Bitnun A et al.
Case definitions, diagnostic algorithms,
and priorities in encephalitis: consensus
statement of the International
Encephalitis Consortium. Clin Infect Dis
2013; 57: 1114–28.
15 Davison KL, Crowcroft NS, Ramsay ME,
Brown DWG, Andrews NJ. Viral
encephalitis in England, 1989–1998:
what did we miss? Emerg Infect Dis 2003;
9: 234–40.
16 Kulkarni MA, Lecocq AC, Artsob H,
Drebot MA, Ogden NH. Epidemiology
and aetiology of encephalitis in Canada,
1994–2008: a case for undiagnosed
arboviral agents? Epidemiol Infect 2013;
141: 2243–55.
17 Granerod J, Ambrose HE, Davies NW,
Clewley JP, Walsh AL, Morgan D et al.
Causes of encephalitis and differences in
their clinical presentations in England:
a multicentre, population-based
prospective study. Lancet Infect Dis 2010;
10: 835–44. Erratum appears in Lancet
Infect Dis 2011 11: 79.
18 Tenembaum S, Chitnis T, Ness J,
Hahn JS. Acute disseminated
encephalomyelitis. Neurology 2007;
68(16 Suppl 2): S23–36.
19 Young NP, Weinshenker BG,
Lucchinetti CF. Acute disseminated
encephalomyelitis: current
understanding and controversies. Semin
Neurol 2008; 28: 84–94.
20 Gable MS, Sheriff H, Dalmau J, Tilley
DH, Glaser CA. The frequency of
autoimmune N-methyl-D-aspartate
receptor encephalitis surpasses that of
individual viral etiologies in young
individuals enrolled in the California
Encephalitis Project. Clin Infect Dis 2012;
54: 899–904.
21 Titulaer MJ, McCracken L, Gabilondo I,
Armangue T, Glaser C, Iizuka T et al.
Treatment and prognostic factors for
long-term outcome in patients with
anti-NMDA receptor encephalitis: an
© 2015 Royal Australasian College of Physicians
574
Consensus guidelines for encephalitis
22
23
24
25
26
27
28
29
30
31
32
observational cohort study. Lancet Neurol
2013; 12: 157–65.
Hacohen Y, Wright S, Waters P, Agrawal
S, Carr L, Cross H et al. Paediatric
autoimmune encephalopathies: clinical
features, laboratory investigations and
outcomes in patients with or without
antibodies to known central nervous
system autoantigens. J Neurol Neurosurg
Psychiatry 2013; 84: 748–55.
Vincent A, Buckley C, Schott JM, Baker
I, Dewar BK, Detert N et al. Potassium
channel antibody-associated
encephalopathy: a potentially
immunotherapy-responsive form of
limbic encephalitis. Brain 2004; 127(Pt
3): 701–12.
Suleiman J, Brenner T, Gill D, Brilot F,
Antony J, Vincent A et al. VGKC
antibodies in pediatric encephalitis
presenting with status epilepticus.
Neurology 2011; 76: 1252–5.
Wong SH, Saunders MD, Larner AJ, Das
K, Hart IK. An effective immunotherapy
regimen for VGKC antibody-positive
limbic encephalitis. J Neurol Neurosurg
Psychiatry 2010; 81: 1167–9.
Lai M, Huijbers MGM, Lancaster E,
Graus F, Bataller L, Balice-Gordon R
et al. Investigation of LGI1 as the antigen
in limbic encephalitis previously
attributed to potassium channels: a case
series. Lancet Neurol 2010; 9: 776–85.
Gultekin SH, Rosenfeld MR, Voltz R,
Eichen J, Posner JB, Dalmau J.
Paraneoplastic limbic encephalitis:
neurological symptoms, immunological
findings and tumour association in 50
patients. Brain 2000; 123(Pt 7):
1481–94.
Dalmau J, Rosenfeld MR. Paraneoplastic
syndromes of the CNS. Lancet Neurol
2008; 7: 327–40.
Lancaster E, Dalmau J. Neuronal
autoantigens – pathogenesis, associated
disorders and antibody testing. Nat Rev
Neurol 2012; 8: 380–90.
Lancaster E, Martinez-Hernandez E,
Dalmau J. Encephalitis and antibodies to
synaptic and neuronal cell surface
proteins. Neurology 2011; 77: 179–89.
Huppatz C, Kelly PM, Levi C, Dalton C,
Williams D, Durrheim DN. Encephalitis
in Australia, 1979–2006: trends and
aetiologies. Commun Dis Intell 2009; 33:
192–7.
Paterson BJ, Mackenzie JS, Durrheim
DN, Smith D. A review of the
epidemiology and surveillance of viral
zoonotic encephalitis and the impact on
33
34
35
36
37
38
39
40
41
42
43
44
human health in Australia. N S W Public
Health Bull 2011; 22: 99–104.
Granerod J, Cunningham R, Zuckerman
M, Mutton K, Davies NWS, Walsh AL
et al. Causality in acute encephalitis:
defining aetiologies. Epidemiol Infect
2010; 138: 783–800.
Beaman MH, Wesselingh SL. 4: Acute
community-acquired meningitis and
encephalitis. Med J Aust 2002; 176:
389–96.
Nigrovic LE, Shah SS, Neuman MI.
Correction of cerebrospinal fluid protein
for the presence of red blood cells in
children with a traumatic lumbar
puncture. J Pediatr 2011; 159:
158–9.
Selvey LA, Dailey L, Lindsay M,
Armstrong P, Tobin S, Koehler AP et al.
The Changing Epidemiology of Murray
Valley Encephalitis in Australia: the
2011 outbreak and a review of the
literature. PLoS Negl Trop Dis 2014; 8:
e2656.
Sinclair AJ, Wienholt L, Tantsis E, Brilot
F, Dale RC. Clinical association of
intrathecal and mirrored oligoclonal
bands in paediatric neurology. Dev Med
Child Neurol 2013; 55: 71–5.
Dale RC, Brilot F, Fagan E, Earl J.
Cerebrospinal fluid neopterin in
paediatric neurology: a marker of active
central nervous system inflammation.
Dev Med Child Neurol 2009; 51: 317–23.
Debiasi RL, Tyler KL. Molecular methods
for diagnosis of viral encephalitis. Clin
Microbiol Rev 2004; 17: 903–25, table of
contents.
Bloch KC, Glaser C. Diagnostic
approaches for patients with suspected
encephalitis. Curr Infect Dis Rep 2007; 9:
315–22.
Thomson RB Jr, Bertram H. Laboratory
diagnosis of central nervous system
infections. Infect Dis Clin North Am 2001;
15: 1047–71.
Ward KN, Andrews NJ, Verity CM,
Miller E, Ross EM. Human
herpesviruses-6 and -7 each cause
significant neurological morbidity in
Britain and Ireland. Arch Dis Child 2005;
90: 619–23.
Bitnun A, Ford-Jones E, Blaser S,
Richardson S. Mycoplasma pneumoniae
encephalitis. Semin Pediatr Infect Dis 2003;
14: 96–107.
Wainwright MS, Martin PL, Morse RP,
Lacaze M, Provenzale JM, Coleman RE
et al. Human herpesvirus 6 limbic
encephalitis after stem cell
45
46
47
48
49
50
51
52
53
54
55
56
transplantation. Ann Neurol 2001; 50:
612–19.
Seeley WW, Marty FM, Holmes TM,
Upchurch K, Soiffer RJ, Antin JH et al.
Post-transplant acute limbic encephalitis:
clinical features and relationship to
HHV6. Neurology 2007; 69: 156–65.
Kranick SM, Nath A. Neurologic
complications of HIV-1 infection and its
treatment in the era of antiretroviral
therapy. Continuum (N Y) 2012; 18:
1319–37. 6 Infectious Disease.
Ferenczy MW, Marshall LJ, Nelson CD,
Atwood WJ, Nath A, Khalili K et al.
Molecular biology, epidemiology, and
pathogenesis of progressive multifocal
leukoencephalopathy, the JC
virus-induced demyelinating disease of
the human brain. Clin Microbiol Rev
2012; 25: 471–506.
Lescure FX, Moulignier A, Savatovsky J,
Amiel C, Carcelain G, Molina JM et al.
CD8 encephalitis in HIV-infected
patients receiving cART: a treatable
entity. Clin Infect Dis 2013; 57: 101–8.
Currie BJ, Fisher DA, Howard DM,
Burrow JN. Neurological melioidosis.
Acta Trop 2000; 74: 145–51.
Smith JK, Young MM, Wilson KL, Craig
SB. Leptospirosis following a major flood
in Central Queensland, Australia.
Epidemiol Infect 2013; 141: 585–90.
Venkateswaran S, Hawkins C, Wassmer
E. Diagnostic yield of brain biopsies in
children presenting to neurology. J Child
Neurol 2008; 23: 253–8.
Wong SH, Jenkinson MD, Faragher B,
Thomas S, Crooks D, Solomon T. Brain
biopsy in the management of neurology
patients. Eur Neurol 2010; 64: 42–5.
Pulhorn H, Quigley DG, Bosma JJ,
Kirollos R, du Plessis DG, Jenkinson MD.
Impact of brain biopsy on the management of patients with nonneoplastic
undiagnosed neurological disorders. Neurosurgery 2008; 62: 833–7, discussion 7–8.
Hall WA. The safety and efficacy of
stereotactic biopsy for intracranial
lesions. Cancer 1998; 82: 1749–55.
Willoughby RE Jr, Tieves KS, Hoffman
GM, Ghanayem NS, Amlie-Lefond CM,
Schwabe MJ et al. Survival after
treatment of rabies with induction
of coma. N Engl J Med 2005; 352:
2508–14.
Milwaukee Protocol. Version 4.0
(updated 2012 Sep 13) [cited 2015 Apr
12]. Available from URL: http://www
.mcw.edu/Pediatrics/InfectiousDiseases/
PatientCare/Rabies.htm
© 2015 Royal Australasian College of Physicians
575
57 Openshaw H, Cantin EM.
Corticosteroids in herpes simplex virus
encephalitis. J Neurol Neurosurg Psychiatry
2005; 76: 1469.
58 Steiner I, Kennedy PG, Pachner AR. The
neurotropic herpes viruses: herpes
simplex and varicella-zoster. Lancet
Neurol 2007; 6: 1015–28.
59 Tyler KL. Herpes simplex virus infections
of the central nervous system:
encephalitis and meningitis, including
Mollaret’s. Herpes 2004; 11(Suppl 2):
57A–64A.
60 Kimberlin DW. Herpes simplex virus
infections in neonates and early
childhood. Semin Pediatr Infect Dis 2005;
16: 271–81.
61 Whitley RJ, Kimberlin DW. Herpes
simplex encephalitis: children and
adolescents. Semin Pediatr Infect Dis 2005;
16: 17–23.
62 Martinez-Torres F, Menon S, Pritsch M,
Victor N, Jenetzky E, Jensen K et al.
Protocol for German trial of
Acyclovir and corticosteroids in
Herpes-simplex-virus-encephalitis
63
64
65
66
(GACHE): a multicenter, multinational,
randomized, double-blind, placebocontrolled German, Austrian and Dutch
trial [ISRCTN45122933]. BMC Neurol
2008; 8: 40.
Gilden D. Varicella zoster virus and
central nervous system syndromes.
Herpes 2004; 11(Suppl 2): 89A–
94A.
Kamei S, Sekizawa T, Shiota H,
Mizutani T, Itoyama Y, Takasu T et al.
Evaluation of combination therapy using
aciclovir and corticosteroid in adult
patients with herpes simplex virus
encephalitis. J Neurol Neurosurg Psychiatry
2005; 76: 1544–9.
Ooi MH, Wong SC, Lewthwaite P,
Cardosa MJ, Solomon T. Clinical
features, diagnosis, and management of
enterovirus 71. Lancet Neurol 2010; 9:
1097–105.
Dale RC, Brilot F, Banwell B. Pediatric
central nervous system inflammatory
demyelination: acute disseminated
encephalomyelitis, clinically isolated
syndromes, neuromyelitis optica, and
67
68
69
70
71
multiple sclerosis. Curr Opin Neurol 2009;
22: 233–40.
Dale RC, Brilot F, Duffy LV, Twilt M,
Waldman AT, Narula S et al. Utility and
safety of rituximab in pediatric
autoimmune and inflammatory CNS
disease. Neurology 2014; 83: 142–50.
Dalmau J, Gleichman AJ, Hughes EG,
Rossi JE, Peng X, Lai M et al.
Anti-NMDA-receptor encephalitis: case
series and analysis of the effects of
antibodies. Lancet Neurol 2008; 7:
1091–8.
Florance NR, Davis RL, Lam C, Szperka
C, Zhou L, Ahmad S et al.
Anti-N-methyl-D-aspartate receptor
(NMDAR) encephalitis in children and
adolescents. Ann Neurol 2009; 66: 11–18.
Chaudhuri A, Kennedy PG. Diagnosis
and treatment of viral encephalitis.
Postgrad Med J 2002; 78: 575–83.
Britton PN, Dale RC, Booy R, Jones CA.
Acute encephalitis in children: Progress
and priorities from an Australasian
perspective. J Paediatr Child Health 2015;
51: 147–58.
B R I E F C O M M U N I C AT I O N S
Survey of infection control and antimicrobial stewardship
practices in Australian residential aged-care facilities
R. L. Stuart,1,2 C. Marshall,3,4 E. Orr,1 N. Bennett,5 E. Athan,6,7 D. Friedman6,7 and M. Reilly,8 on behalf of
Members of RACRIG (Residential Aged Care Research Interest Group)*
1
Department of Infectious Diseases, Monash Health and 2Department Medicine, Monash University and 3Department of Infectious Diseases, Royal
Melbourne Hospital and 4Department Medicine, University of Melbourne and 5VICNISS Coordinating Centre, Melbourne and 6Department Infectious
Disease, Barwon Health and 7Department of Medicine, Deakin University, Geelong, Victoria and 8Hands-On Infection Control, Perth, Western Australia,
Australia
Key words
residential care, infection control, antibiotic
stewardship.
Correspondence
Rhonda L. Stuart, Infectious Diseases, Monash
Health, 246 Clayton Road, Clayton, Melbourne,
Vic. 3168, Australia.
Email: [email protected]
Abstract
This study assessed infection prevention and antimicrobial stewardship (AMS) practices
in Australian residential aged-care facilities (RACF). Two hundred and sixty-five surveys
(15.6%) were completed with all states represented and the majority (177 (67.3%))
privately run. Only 30.6% RACF had infection control trained staff on site. Few facilities
had AMS policies, only 14% had antimicrobial prescribing restrictions. Most facilities
offered vaccination to residents (influenza vaccination rates >75% in 73% of facilities),
but pneumococcal vaccination was poor.
Received 25 August 2014; accepted 29 January
2015.
doi:10.1111/imj.12740
© 2015 Royal Australasian College of Physicians
576
Brief Communications
The term residential aged-care facilities (RACF) refers to
a group of residential facilities designed to meet the needs
of the elderly.1 These include nursing homes, skilled
nursing facilities and assisted living facilities. The population in RACF is vulnerable to infection due to frailty,
poor functional status, multiple comorbidities and compromised immune systems.2 Bed-bound residents are at
greater risk of skin and soft tissue infections, while those
with urine and/or faecal incontinence have an increased
risk of urinary tract infections.3 In addition, close living
proximity and frequent carer-resident contact facilitate
the spread of organisms among RACF residents.2 This,
coupled with frequent transfers to the acute hospital
setting, promotes a higher infection burden among residents in RACF compared to those living at home.4
Although the infection burden among the RACF population has long been recognised, infection prevention
efforts are often limited to sentinel infection surveillance
activity. Of particular concern is the widespread empiric
antibiotic prescribing in RACF that may lead to the emergence of antibiotic resistance. Studies have reported
increasing use of broad-spectrum oral antibiotics, such as
fluoroquinolones, among this population, with up to
75% of use judged to be inappropriate.5,6 In an era where
multidrug-resistant organisms (MDRO) are emerging in
the community, RACF residents have been increasingly
identified as important reservoirs of such bacteria.7,8
In Australia, RACF are operated by not-for-profit
organisations, church and charitable organisations, commercial organisations and some state and governments.
To receive Australian Government subsidies, RACF are
required to be accredited and meet four Accreditation
Standards. ‘An effective infection control programme’
and ‘care recipients’ medication is managed safely and
correctly’ are two of the 44 expected outcomes.9
The aim of this study was to document the infection
prevention and antimicrobial stewardship practices in
RACF around Australia. This will, in-turn, lend itself
to developing an understanding of gaps to inform an
ongoing research agenda.
All RACF caring for low or high-level care residents (as
defined by the Aged Care Funding Instrument),10 and
having more than 50 beds were identified from a data*This group comprises members from The Healthcare Infection
Control Special Interest Group (HICSIG), which is part of the
Australian Society of Infectious Diseases (ASID), as well as
members from the Australasian College for Infection Prevention
and Control (ACIPC). Other Members of RACRIG: David Looke,
John Ferguson, Judy Forrest, Luke Chen, Michelle Callard, Mike
Richards, Mona Schousboe, Sandy Berenger, Daryl Pye, Mary
Smith, Michael Wishart, Kirsty Buising.
Funding: None.
Conflict of interest: None.
base held by the Commonwealth Department of Health.
Managers of these facilities were contacted by email, and
then a follow-up phone call, to ask if they would like to
be involved in the survey. Details of contact numbers and
email addresses for these sites are all publicly available on
the Commonwealth Department of Health web site.10
The survey could be completed online or on paper and
mailed back to the principal investigator. Surveys were
sent to all available addresses, but we were unable to
confirm that the intended addressee received the email
unless they responded. Completion and submission of
the survey implied consent.
The survey was developed to enable RACF to answer
questions in a de-identified manner on aspects of infection control and antimicrobial stewardship, that the
members of the working group felt important. This
included questions (in a checkbox format) on the population demographics of residents, the expertise and availability of infection control personnel, the availability of
policies and procedures around infection control and
antimicrobial stewardship, the type of infection surveillance that was undertaken in the facility and whether
vaccination programmes were available for staff and
residents. There were 40 questions in total, with the
survey taking 5–10 min to complete. The RACF were
invited once to complete the survey, there were no
inducements offered for completion. Analysis was
descriptive. The Commonwealth Departmental Ethics
Committee approved the project.
There were 265 surveys returned from a possible total
pool of 1700 RACF (15.6%) with the majority (177
(67.3%)) being from private facilities. All States were
represented with 81 (30.6%), 88 (33.2%), 43 (16.2%),
22 (8.3%), 16 (6%) and 14 (5.3%) of returned surveys
coming from New South Wales, Victoria, Queensland,
South Australia, Western Australia and Tasmania respectively. This total number of RACF beds per facility ranged
from 50 to 250 (mean 90, median 77 beds). The total
number of RACF beds covered by the survey was 22 335,
with 15 725 (70.4%) being high care. Occupancy at the
time of the study was 95% (range 45–100%; median
98%). Male residents accounted for 7230 occupants
(32.4%) with 13 972 (62.6%) being greater than 85
years of age. Over the entire cohort, there were 776
residents (3.5%) with urinary catheters in situ and 22
(0.1%) with a vascular catheter in place, while 910
(4.1%) had been in an acute care facility in the previous
30 days.
Two hundred and forty-two (91.3%) facilities had designated infection control personnel although only 30.6%
(74) of these staff had any certification in infection
control. Two hundred and sixteen (81.5%) facilities had
an infection control committee. The availability of poli-
© 2015 Royal Australasian College of Physicians
577
Brief Communications
Table 1 Reported availability of procedures, surveillance undertaken and
vaccine availability
n (%)
Procedure availability
Management/isolation of MRSA carriers
Management/isolation of VRE carriers
Management/isolation of MDR-GN
Wound management
Management of urinary catheters
Management of vascular catheters
Management of enteral feeding
Standard precautions
Management/isolation of gastroenteritis
Management/isolation of Clostridium difficile
Cleaning rooms of MRO carriers
Hand hygiene
Antibiotic use
Surveillance undertaken
Non-catheter-associated urinary tract infection
Catheter-associated urinary tract infection
Blood stream infection
Common cold/pharyngitis
Influenza-like illness
Lower respiratory tract infection/pneumonia
Skin and soft tissue infection
Skin condition, wounds and ulcers
Eye infection
Gastrointestinal infection
Clostridium difficile infection
MRSA infection
VRE infection
Use of transmission-based precautions where appropriate
Contact (single room, gloves, gown)
Droplet (single room, surgical mask)
Airborne (negative pressure room, N95/P2 mask
None
Resident vaccinations available
Influenza
Pneumococcal
Tetanus/diphtheria/pertussis
Varicella-zoster
No vaccines available
Staff vaccines available
Influenza
Tetanus/diphtheria/pertussis
Varicella-zoster
Hepatitis B
Measles/mumps/rubella
QuantiFeron Gold/Mantoux testing
No vaccines available
226 (85.9)
206 (78.3)
174 (66.2)
259 (98.5)
250 (95.1)
73 (27.8)
218 (82.9)
253 (96.2)
258 (98.1)
156 (59.3)
228 (86.7)
256 (97.3)
106 (40.3)
240 (92.7)
211 (81.5)
171 (66)
195 (75.3)
229 (88.4)
242 (93.4)
242 (93.4)
251 (96.9)
250 (96.5)
242 (93.4)
151 (58.3)
197 (76.1)
182 (70.3)
239 (91.6)
183 (70.1)
34 (13)
23 (8.8)
245 (96.1)
170 (66.7)
11 (4.3)
6 (2.6)
10 (3.9)
217 (86.5)
21 (8.4)
77 (30.7)
20 (8)
26 (10.4)
16 (6.4)
31 (12.4)
MDR-GN, multidrug resistant Gram negative; MDRO, multidrug resistant
organism; MRSA, methicillin resistant Staphylococcus aureus; VRE,
vancomycin-resistant enterococcus.
cies or procedures within the facility and the surveillance
performed are outlined in Table 1.
Alcohol-based hand rub (ABHR) was available in all
publically accessible areas in 246 (95.4%) of facilities and
Table 2 Recommendations for RACF in Australia
Recommendation
Align accreditation of
RACF with national
standards for the
acute sector
Develop database of
policies and
procedures for
facilities to access
Develop antimicrobial
guidelines for
common infections in
RACF
Develop surveillance
programme for MDRO
Enhanced immunisation
Develop education
packages for nursing
staff
Hand hygiene
Cleaning and
disinfection
Examples
Include a focus on
• Infection prevention and control
• Antimicrobial stewardship
• Immunisation
Standardised procedures for managing
• MRSA colonisation
• VRE colonisation
• Management of urinary catheters
• Multidrug-resistant Gram-negative
colonisation
• Clostridium difficile infection
Suggested
• Urinary sepsis with and without indwelling
catheter
• Respiratory tract infection
• Skin and soft tissue infection
Include Australia-wide focus on
• Clostridium difficile
• Multidrug-resistant Gram-negative bacteria
• MRSA and VRE
Increase vaccination rates
• Influenza – resident
• Influenza – healthcare workers
• Pneumococcal vaccine – residents
Diploma opportunities
• Nurse practitioner role in RACF
• Infection control in RACF
Guideline development specific for RACF
Guideline development specific for RACF
MDRO, multidrug resistant organism; MRSA, methicillin resistant Staphylococcus aureus; RACF, residential aged-care facilities; VRE, vancomycinresistant enterococcus.
resident bedrooms in 72 (27.9%). While four facilities
(1.6%) did not have ABHR available at all.
Antimicrobial prescribing was the role of general practitioners in all facilities, while 88 (33.6%) also had specialist consultants prescribing. Notification to the medical
officer about resident illness and need for antibiotics was
made by telephone in 254 (98.5%) via a communication
book in 118 (45.7%) and in 16 cases (6.2%), the facility
waited until the doctor made a routine visit. Notably,
policies for antibiotic use were only available in 106
(40.3%) of facilities (Table 1), with only 36 (13.9%)
stating there were any restrictions to prescribing. Local
private pharmacies dispensed the majority of medications
(222, 86%) with the remainder distributed through a
hospital-associated pharmacy.
Vaccination for residents and staff is summarised in
Table 2. Influenza vaccination was available in 245
(96.1%) of facilities with vaccination rates in 2012–2013
reported as >75% in 190 (72.8%) facilities, 50–75% in 45
© 2015 Royal Australasian College of Physicians
578
Brief Communications
(17.3%) and <50% in 26 (11%) of facilities. Although
170 (66.7%) facilities claimed that pneumococcal
vaccination was available for residents, only 46 (20.7%)
reported vaccination rates >75%, with 99 facilities
(44.6%) reporting that the pneumococcal vaccination
rate for residents was unknown. Staff influenza vaccination rates were >75% in 35 (13.6%), 50–75% in 66
(25.7%), 20–50% in 105 (40.9%) and <20% in 51
(19.8%) of facilities.
This large survey of infection control and antimicrobial
stewardship practices in RACF across Australia is
the first of its kind to our knowledge. Infection control
resources were evident across the services; however,
most facilities did not have nursing staff possessing a
higher certification in this area. Hand hygiene education
and ABHR availability across all but a few facilities
implies the importance placed on this as an infection
control intervention, similar to that seen in the acute care
sector.11 Surveillance activities for common infections in
RACF was also generally undertaken although a lack of
surveillance for Clostridium difficile was notable given the
increasing concern there is for this disease.12 Additionally,
we were unable to assess if standardised criteria were
used for surveillance in these facilities – an area that
requires further work.
While the majority of facilities had procedures in place
for common infections, the currency of these documents
was not assessed in this study. In fact, some services
commented that it would be beneficial for Australian
RACF to have access to a bank of policies and procedures
that could be modified to suit individual facilities. This
could be an area for future investment and one that may
assist RACF accreditation. The survey did not address
auditing of these procedures.
Antimicrobial stewardship is recognised as a vital component of infection control activities and has become part
of National Standards for Accreditation in the Australian
acute health sector. Recognition of AMS importance is
References
1 Siegel JD, Rhinehart E, Jackson M,
Chiarello L, the Healthcare Infection
Control Practices Advisory Committee.
2007 guideline for isolation precautions:
preventing transmission of infectious
agents in healthcare settings. June, 2007
[cited 2014 Jul]. Available from URL:
http://www.cdc.gov/ncidod/dhqp/pdf/
isolation2007.pdf
2 Garibaldi RA. Residential care and
the elderly: the burden of infection.
J Hosp Infect 1999; 43(Suppl): S9–18.
also growing in residential care with increasing studies
showing an urgent need for research in this area.6,13 Our
survey supports the need to develop guidelines for antimicrobial use in RACF, to support facilities to enforce
restrictions on prescribing and to develop a nationwide
infection surveillance programme for MDRO.
Improving influenza vaccination rates in the elderly
decreases lower respiratory tract infection and decreases
hospitalisation.14 This survey has shown room for
improvement with 28% facilities declaring that <75% of
their residents were vaccinated in the previous year.
More concerning is the low influenza vaccination rates of
healthcare worker in these facilities (only 13.6% achieving vaccination rates >75%). This is a trend seen Australia wide,15 but given that most evidence for the
healthcare worker vaccine efficacy is in the residential
care setting,16 it is an area that deserves urgent attention.
Pneumococcal vaccination is recommended for those
>65 years of age,17 and it is concerning that RACF do not
place importance on knowing if residents are vaccinated.
More research in the area of vaccination in RACF is
required. A summary of recommendations from this
study is presented in Table 2.
A weakness of this study is the low response rate
(15.6%) of total available services such as these data
may not reflect all RACF across Australia. However,
given that the survey does include 265 sites and over
22 000 beds, it is the largest study of its kind. Furthermore, the response rate is estimated from a possible pool
of 1700 facilities with greater than 50 beds; however, we
were not able to confirm that all facilities received the
email invitation, thus the response rate is likely to be
much higher.
In summary, this large survey has uncovered some
important areas for future research and quality improvement in RACF, namely AMS, immunisation of both residents and staff and the role that accreditation changes
may have in these areas.
3 Nicolle LE, Strausbaugh LJ, Garibaldi
RA. Infections and antibiotic resistance
in nursing homes. Clin Microbiol Rev
1996; 9: 1–17.
4 Ingarfield SL, Finn JC, Jacobs IG, Gibson
NP, Holman CDJ, Jelinek GA et al. Use
of emergency departments by older
people from residential care: a
population based study. Age Ageing 2009;
38: 314–18.
5 Stuart RL, Kotsanas D, Webb B,
Vandergraaf S, Gillespie EE, Hogg GG
et al. Prevalence of antimicrobialresistant organisms in residential aged
care facilities. Med J Aust 2011; 195:
530–3.
6 Lim CJ, McLellan SC, Cheng AC, Culton
JM, Parikh SN, Peleg AY et al.
Surveillance of infection burden in
residential aged care facilities. Med J Aust
2012; 196: 327–31.
7 Rogers BA, Ingram PR, Runnegar N,
Pitman MC, Freeman JT, Athan E et al.
Community-onset Escherichia coli
infection resistant to expanded-spectrum
cephalosporins in low-prevalence
countries. Antimicrob Agents Chemother
2014; 58: 2126–34.
© 2015 Royal Australasian College of Physicians
579
Brief Communications
8 Pop-Vicas A, Tacconelli E, Gravenstein S,
Lu B, D’Agata EMC. Influx of
multidrug-resistant, gram-negative
bacteria in the hospital setting and the
role of elderly patients with bacterial
bloodstream infection. Infect Control Hosp
Epidemiol 2009; 30: 325–31.
9 Australian Government. Quality of care
principles. 2014 [cited 2014 Jul].
Available from URL: http://www
.comlaw.gov.au/Details/F2014L00830
10 Australian Government. Aged care. 2014
[cited 2014 Jul]. Available from URL:
http://www.myagedcare.gov.au/
aged-care-homes
11 Grayson ML, Russo PL, Cruickshank M,
Bear JL, Gee CA, Hughes CF et al.
Outcomes from the first 2 years of the
Australian National Hand Hygiene
Initiative. Med J Aust 2011; 195:
615–19.
12 Slimings C, Armstrong P, Beckingham
WD, Bull AL, Hall L, Kennedy KJ et al.
Increasing incidence of Clostridium difficile
infection, Australia, 2011–2012. Med J
Aust 2014; 200: 272–6.
13 Stuart RL, Wilson J, Bellaard-Smith E,
Brown R, Wright L, Vandergraaf S et al.
Antibiotic use and misuse in residential
aged care facilities. Intern Med J 2012;
42: 1145–9.
14 Loeb M, McGeer A, McArthur M, Walter
S, Simor AE. Risk factors for pneumonia
and other lower respiratory tract
infections in elderly residents of
long-term care facilities. Arch Intern Med
1999; 159: 2058–64.
15 Seale H, MacIntyre CR. Seasonal
influenza vaccination in Australian
hospital health care workers: a review.
Med J Aust 2011; 195: 336–8.
16 Talbot TR, Babcock H, Caplan AL, Cotton
D, Maragakis LL, Poland GA et al. Revised
SHEA position paper: influenza vaccination of healthcare personnel. Infect Control
Hosp Epidemiol 2010; 31: 987–95.
17 Australian Government. The Australian
Immunisation Handbook. Canberra:
Australian Government, Department of
Health; 2014; 1–546.
Long-term follow up of paediatric liver transplant recipients:
outcomes following transfer to adult healthcare in New Zealand
R. Harry,1 C. Fraser-Irwin,2 S. Mouat,2 E. Gane,1 S. Munn1 and H. M. Evans2
1
New Zealand Liver Transplant Unit (NZLTU), Auckland City Hospital and 2Department of Paediatric Gastroenterology, Starship Hospital, Auckland,
New Zealand
Key words
paediatrics, liver transplantation, transition to
adult care, patient non-adherence.
Correspondence
Rachael Harry, New Zealand Liver Transplant
Unit, Auckland City Hospital, 1023, Auckland,
New Zealand.
Email: [email protected]
Abstract
Poor outcomes are reported in young people with chronic health conditions. We performed a retrospective notes review of New Zealand paediatric liver transplant recipients
transferred to adult services. Two patients were lost to follow up. Out of 20, 12 were
non-adherent, and out of 12, 7 developed rejection. Other risk behaviours were
common in the non-adherent group. We conclude that dedicated services for these
young people may be needed to optimise outcomes.
Received 22 October 2014; accepted 28
January 2015.
doi:10.1111/imj.12721
Adolescents and young people who have chronic health
conditions are at increased risk of poor health and social
outcomes compared with those who do not.1,2,3 This has
been demonstrated among young people who were
recipients of solid organ transplants as children. Among
Funding: None.
Conflict of interest: None.
young people who have received renal transplants in
childhood, graft loss is reported as between 35% at 2
years following transfer to adult services4 and 67% at 4
years5. In liver transplant populations, others have
shown adherence with investigations, clinic appointments and medication is compromised in patients who
transfer to adult care.6 Such non-adherence is not
uniform but has been shown to be associated with low
© 2015 Royal Australasian College of Physicians
580
Brief Communications
socioeconomic status, single parenting, psychiatric diagnoses, school dropout, substance abuse and child abuse.7
Ultimately, non-adherence in liver transplant recipients
may be associated with late rejection, graft loss,
re-transplantation or death.8
Young people with chronic illness are also more likely
to engage in risk-taking behaviours such as smoking,
drinking alcohol, taking drugs and unprotected sex and
are more likely to come to harm as a result of them.2
The long-term outcome of paediatric liver transplant
recipients after transfer to adult services in New Zealand
has not been previously reported.
As such, the clinical notes were reviewed retrospectively of New Zealand paediatric liver transplant recipients born before 1998 who had been planned to transfer
to adult services prior to 31 December 2012 (i.e. aged
over 14 years). Data were retrieved from New Zealand
Liver Transplant Unit (NZLTU) and Starship Hospital
databases whether transplanted in New Zealand or
abroad. Prior to 2002, when the paediatric liver transplant service started at Starship Hospital, Auckland, paediatric liver transplant recipients underwent assessment
and transplantation in Brisbane, Australia.
This was granted institutional approval by the Auckland DHB research review committee.
Demographic data were recorded from clinical record
or NZLTU databases including ethnicity, age and diagnosis
at transplant.
The following outcomes occurring following transfer to
adult services were sought:
1. Non-adherence with medication as evidenced by
undetectable tacrolimus levels, self-report or report by
others of non-adherence in the clinical record or episodes of rejection attributed to non-adherence by treating
physicians.
2. Health outcomes such as rejection (biopsy proven or
as defined by treating physicians) admissions with rejection, mental health diagnoses, graft loss and death
3. Social outcomes, such as drug and alcohol use,
unplanned pregnancy and legal issues as reported in the
clinical record.
The data are reported in two groups based on the
presence or absence of evidence of non-adherence as
median and ranges.
A total of 60 New Zealand paediatric liver transplant
recipients who underwent liver transplantation before
1998 was identified from NZLTU and Starship Hospital
databases. Of these, nine had died post-transplant, six
had moved abroad, four had been lost to follow up (presumed to be abroad) and six were still in paediatric care.
There were 13 young people in the process of transition
through the newly established young person’s liver
clinic.
Table 1 Demographic data and health outcomes
Number
NZE
Age at Tx
Time Tx to transfer
Age at transfer
NA bloods/clinics
Late rejection
Multiple episodes
Mental health
Ongoing healthcare
engagement
Adherent
Non-adherent
8
8 (100%)
5 years (1–16)
5 years (1–9)
16.5 years (16–19)
0
0
0
0
8 (100%)
12
8 (66%)
2 years (1–13)
3 years (1–9)
17 years (15–18)
10 (83%)
7 (58%)
3 (25%)
5 (42%)
5 (42%)
NA, non-attendance; NZE, New Zealand European; Tx, transplant.
The remaining 22 patients had planned to be transferred directly from paediatrics to adult health services
without being involved in any formal transition pathway
or youth specific service.
Data were available on 20 of these, as two were lost to
follow up at or shortly after transfer. These data are
summarised in Table 1.
A total of 16 young people was New Zealand European
(NZE) and four were New Zealand Maori or Pacific
Islander. The median time after transplant was 17 years
(range 4–27 years) and the median age was 21.5 years
(range 17–30 years).
Out of 20, 12 (60%) of these patients had evidence of
non-adherence following transfer. The four non-NZE
young people all had evidence of non-adherence in their
record.
Of those who were non-adherent, 7/12 (58%) had
either suspected or biopsy proven rejection. Three of
these young people had multiple episodes of rejection
requiring hospitalisation. Overall, 35% of the nonadherent group had evidence of rejection, whereas no
patients who were thought to be adherent had late rejection. No patients in this series required re-transplantation
or died.
Other risk behaviours, such as alcohol and substance
use, are common in the group of patients with evidence
of non-adherence and are not reported in the group
presumed to be adherent (Fig. 1).
Young people in the non-adherent group were also
suboptimally engaged with healthcare with 10 young
people in the non-adherent group (83%) reported to be
suboptimally adherent with attendance at clinics or
monitoring blood tests. At the time of data collection,
only 42% of the non-adherent group were receiving
regular secondary or tertiary healthcare in New Zealand
(Table 1).
Other health risks were reported in the nonadherent group. Of this group, 42% carried mental
© 2015 Royal Australasian College of Physicians
581
Brief Communications
Figure 1 Substance use in the non-adherent group. ( ), Alcohol; ( ),
alcohol and drugs; ( ), alcohol, drugs and legal issues; ( ), no substance
use.
health diagnoses resulting in one intentional overdose.
Six pregnancies are reported in four young women in the
non-adherent group. Two young women had terminations aged 18 years, one also had an unplanned pregnancy at 19 years. Three live births were reportedin
patients aged 20, 22 and 22 years where intent related to
pregnancy was not reported. In the group of patients
with no evidence of non-adherence, there was one pregnancy reported to be intentional.
In conclusion, these data comprise a retrospective
notes review of patients who underwent paediatric liver
References
1 Sawyer SM, Drew S, Yeo MS, Britto MT.
Adolescents with a chronic condition:
challenges living, challenges treating.
Lancet 2007; 369: 1481–9.
2 Denny S, Farrant B, Cosgriff J, Harte M,
Sheridan J, Robinson E. Forgone health
care among secondary school students in
New Zealand. J Prim Health Care 2013; 5:
11–18.
3 Denny S, de Silva M, Fleming T, Clark T,
Merry S, Ameratunga S et al. The
prevalence of chronic health conditions
transplantation and were subsequently transferred
to adult services. There are limitations to these data,
including the under reporting of non-adherence and
other risk behaviours that are inherent in retrospective
clinical record reviews and the potential over reporting of
risk behaviours in the non-adherent group, as these may
have been specifically sought where they were not in the
adherent group.
Notwithstanding the limitations of this retrospective
study, we found that reported non-adherence with medication is common in paediatric liver transplant recipients
following transfer to adult services in New Zealand. Nonadherence was reported in two out of three patients, of
whom two out of three subsequently suffered harm as a
consequence of rejection.
Despite the flaws in the data, we confirm that in this
population, non-adherence occurs as part of a constellation of risk behaviours, including alcohol and drug use,
unintentional pregnancy, mental health diagnoses and
legal issues.
Dedicated services for young people have been shown
to improve outcomes for paediatric renal transplant
recipients in the UK.5 These data demonstrate that
there may be a need for improved services for young
people with chronic health conditions, including
solid organ transplants, in New Zealand. A holistic
approach that not only addresses the presenting
medical condition, but also provides culturally and
developmentally appropriate care may improve longterm medical and social outcomes for these young
people.
impacting on daily functioning and the
association with emotional well-being
among a national sample of high school
students. J Adolesc Health 2014; 54:
410–15.
4 Watson AR. Non-compliance and transfer
from paediatric to adult kidney transplant
unit. Pediatr Nephrol 2000; 14: 469–72.
5 Harden PN, Walsh G, Bandler N,
Bradley S, Lonsdale D, Taylor J et al.
Bridging the gap: an integrated
paediatric to adult clinical service for
young adults with kidney failure. BMJ
2012; 344: e3718.
6 Burra P. The adolescent and liver
transplantation. J Hepatol 2012; 56:
714–22.
7 Lurie S, Shemesh E, Sheiner PA, Emre S,
Tindle HL, Melchionna L et al.
Non-adherence in pediatric liver
transplant recipients – an assessment of
risk factors and natural history. Pediatr
Transplant 2000; 4: 200–6.
8 Mohammad S, Hormaza L, Neighbors K,
Boone P, Tierney M, Azzam RK et al.
Health status in young adults two decades
after pediatric liver transplantation. Am J
Transplant 2012; 12: 1486–95.
© 2015 Royal Australasian College of Physicians
582
Brief Communications
Primary central nervous system posttransplantation
lymphoproliferative disorder after heart and lung
transplantation
G. Gifford,1 K. Fay,1,2 A. Jabbour3,4,5 and D. D. Ma1,4,6
1
Haematology Department and 3Cardiology Department, St Vincent’s Hospital, 2Haematology Department, Royal North Shore Hospital, 4Faculty of
Medicine, University of New South Wales, 5Clinical Faculty, Victor Chang Cardiac Research Institute, and 6St Vincent’s Centre for Applied Medical
Research, Sydney, New South Wales, Australia
Key words
posttransplant lymhpoproliferative disorder,
central nervous system, lymphoma, heart,
lung, transplantation.
Correspondence
David D. Ma, Haematology, St Vincent’s
Hospital, 390 Victoria Street, Darlinghurst,
Sydney, NSW 2010, Australia.
Email: [email protected]
Received 4 September 2014; accepted 28
February 2015.
Abstract
Primary central nervous system posttransplantation lymphoproliferative disorder
(PCNS-PTLD) is uncommon, especially after heart or lung transplantation. Database
analysis from a single heart and lung transplantation centre and a literature review
pertaining to PCNS-PTLD was performed. In this study, the prevalence of PCNS-PTLD
was 0.18% after heart and/or lung transplants. Of 1674 transplants, three cases of
PCNS-PTLD developed 14 months, 9 years and 17 years posttransplant, and all were
Epstein–Barr virus driven malignancies. Literature review of the topic revealed predominantly retrospective studies, with most reported cases after renal transplantation.
The overall survival is poor, and it may be improved by early diagnosis and treatment.
There are no published guidelines on the management of PCNS-PTLD; immunechemotherapy in conjunction with reduction of immune suppression is preferred based
on available evidence.
doi:10.1111/imj.12735
Primary central nervous system posttransplantation
lymphoproliferative disorder (PCNS-PTLD) is uncommon,
and its occurrence after heart or lung transplantation is
unknown. PCNS-PTLD presents in a plethora of ways with
broad differential diagnoses. Management of this condition in transplant recipients is challenging. There is no
consensus on treatment due to its rarity. This report
describes all identified cases of PCNS-PTLD in a single
institution that performs heart and lung transplantation
over 28 years and its prevalence. The approaches to the
management of PCNS-PTLD based on published evidence
are summarised.
Case 1: A 64-year-old man presented with urinary incontinence, dysphasia, personality changes and memory loss
14 months after heart transplantation for transthyretin
amyloid cardiomyopathy. He was on prednisolone,
mycophenolate and cyclosporin. Magnetic resonance
imaging (MRI) revealed bilateral lesions in the frontotemporal areas. Neurosurgery relieved the associated
raised intracranial pressure and obtained histology;
monomorphic PTLD, variant diffuse large B cell lymphoma (DLBCL) that was angioinvasive and Epstein-Barr
Funding: St Vincent’s Haematology Research Trust.
Conflict of interest: St Vincent’s Hospital.
virus (EBV)–encoded RNA (EBER) positive. There was no
systemic involvement on computed tomography (CT) and
positron electron tomography (PET). Cyclosporin and
prednisone were reduced, mycophenolate replaced by
everolimus. Intravenous rituximab was administered
weekly for 4 weeks resulting in rapid clinical and radiological improvement. The patient then completed six
cycles of high-dose methotrexate (HD-MTX). The patient
remains alive at 18 months, with complete clinical recovery, no evidence of tumour on MRI and a normal functioning cardiac allograft.
Case 2: A 56-year-old man who received double lung
transplantation for α1-antitrypsin deficiency developed
gradual diplopia and ataxia 9 years posttransplant. He was
immunosuppressed with prednisolone, tacrolimus and
mycophenolate mofetil. Imaging showed a circumferential cerebellar lesion surrounding the fourth ventricle.
Emergency decompressive neurosurgery to relieve
raised intracranial pressure also provided a histological
diagnosis of monomorphic PTLD, variant DLBCL. The
lymphoma was angio-invasive and EBER positive. There
was no systemic involvement CT and PET. Tacrolimus
and mycophenolate were reduced and the patient
received one dose of intravenous rituximab. The patient
rapidly deteriorated and died within a month of his
diagnosis.
© 2015 Royal Australasian College of Physicians
583
Brief Communications
Case 3: A 70-year-old man presented with a 4-month
history of progressive ataxia and headache 17 years after
heart transplantation for idiopathic dilated cardiomyopathy. He was immunosuppressed with mycophenolate and
cyclosporin. Multiple cerebral masses with surrounding
oedema and midline shift were detected on cerebral
imaging. An EBER positive, large B cell PCNS-PTLD was
diagnosed, subtype not classified due to extensive necrosis. There was no systemic involvement on CT and PET.
Everolimus replaced cyclosporin. His performance status
was poor and he received one fraction of palliative whole
brain radiation therapy. He died within a month of his
diagnosis.
Immunosuppression for heart and lung transplantation
typically included corticosteroids, a calcineurin inhibitor
and a nucleotide antagonist. Immunosuppression is typically more intense and prolonged for recipients for heart
and lung transplantation, due to the higher incidence
of allograft rejection. For all patients in our institution,
immunosuppressive drugs were dosed according to
pharmacokinetic measurement, with therapeutic ranges
for heart and lung transplant for tacrolimus 10–15 ug/L;
cyclosporin trough therapeutic level varied according to
time after transplant, with a target of 100–180 ug/L a
year after transplantation; mycophenolate mofotil 2.5–
4.5 mg/L; sirolimus 3.5–15 ug/L.
In this single centre study of 1674 heart and/or lung
transplantation recipients (857 heart, 151 single lung,
583 bilateral lung and 83 heart-lung), three were identified to have PCNS-PTLD during a 28-year period
from 1984 to 2012, yielding a prevalence of 0.18%. The
transplantation recipients are routinely reviewed in a
multidisciplinary clinic with a prospectively maintained
departmental database, and local registry, so that the
prevalence is accurate. This study is the first to establish
prevalence for PCNS-PTLD after solid organ transplant.
PTLD ranges from polyclonal proliferations to
monomorphic forms indistinguishable from those which
occur in immunocompetent individuals. The major risk
factors for PTLD are immunosuppression and primary
EBV infection.1 Higher incidences of PTLD are thus
observed in lung transplantation recipients who
often remain heavily immunosuppressed, and in EBVseronegative patients receiving an allograft from an EBVseropositive donor. PTLD has a bimodal distribution; most
cases of PTLD occur within a year of transplantation,1 and
these cases are more likely to involve the allograft.2 PTLD
that occur late are more commonly disseminated, EBVnegative, monomorphic and carry a worse prognosis.3–5
The incidence of PCNS-PTLD is unknown, and older
series have included systemic PTLD with CNS involvement. In a series of 639 heart and/or lung transplantation
recipients, two developed PCNS-PTLD.6 PCNS-PTLD in
the published literature is predominantly reported in
small case series after renal transplantation, the commonest solid organ transplanted. The interval from transplantation to diagnosis of PCNS-PTLD has been reported
to range from 3 months to 5 years.7,8 The prognosis of
PCNS-PTLD is varied and best described after renal transplantation. A case series of 25 patients reported overall
survival of 40% with a median survival time of 26
months.9 In recent international reports, median survival
was 47 months in a series of 34 patients,8 while the
largest series of 84 PCNS-PTLD reported an overall survival of 43%.10
In the joint British Committee for Standards in Haematology and British Transplantation Society Guidelines for
Management of PTLD in Adult Solid Organ Transplant
Recipients,11 reduction in immunosuppression followed
by local radiotherapy with or without steroids, and addition of HD-MTX for young, fit patients are recommended
as grade C, level 3 evidence. Otherwise, survey of the
literature yields neither guidelines nor consensus documents on how to treat PCNS-PTLD. It may not be unreasonable to extrapolate from the treatment of PCNSL to
PCNS-PTLD. PCNSL is chemosensitive and radiosensitive,
and its treatment has been rigorously studied. However,
patients with PCNS-PTLD often have comorbidities
precluding the application of standard PCNSL treatment
protocols.
Recommendations for the treatment of systemic PTLD
might be insufficient or inappropriate for PCNS-PTLD.
Reducing immunosuppression in PTLD is recommended,1 as the attenuation or withdrawal of immunosuppression allows restitution of immunity, especially
against EBV. Reducing immunosuppression alone has
been reported to induce remission in low grade, systemic
PTLD.12 Reduction of immunosuppression needs to be
gradual to prevent allograft rejection, a timeframe that
is not feasible in PCNS-PTLD due to raised intracranial
pressure.
Surgery is directed at obtaining histological diagnosis
and for cerebral decompression. It otherwise has little
therapeutic role.
Whole-brain or focal radiation therapy is administered
alone or in combination for treatment of PCNSL or to
palliate symptoms.13 For PCNS-PTLD, radiation dosages
and schedules have been extrapolated from PCNSL and
human immunodeficiency virus and acquired immunodeficiency syndrome-related lymphoma. Acute and longterm neurotoxicity from radiation therapy limits this
treatment.
Systemic chemotherapy has improved outcomes for
PTLD,3 but the blood brain barrier limits the choice
of agents for PCNS-PTLD. Extrapolating from PCNSL
studies, HD-MTX and cytarabine have been successfully
© 2015 Royal Australasian College of Physicians
584
Brief Communications
used to treat PTLD-CNS.12,14 Complex medical comorbidities and poor performance status prevent the administration of systemic chemotherapy in many PCNS-PTLD
patients.
Therapy with intravenous rituximab has improved
overall survival for patients who develop systemic PTLD.15
Although only a small fraction of the systemic dose is
detectable in cerebrospinal fluid,16 rituximab remains
effective as monotherapy in PCNSL because of its long
half-life, resulting in low yet stable therapeutic concentrations over time.17 Sequential treatment of rituximab
followed by chemotherapy has also been validated.18
Direct administration of rituximab into the central
nervous system either through an Ommaya reservoir or
repeat lumbar punctures is the topic of ongoing research.
A small study showed that intrathecal rituximab was
efficacious in heavily pretreated patients with nonHodgkin lymphoma who have CNS disease.19 Intrathecal
rituximab has been successful in some paediatric sufferers
with PCNS-PTLD.20 However, accessing the intrathecal
space might be inadvisable in patients with raised intrac-
References
1 Heslop HE. How I treat EBV
lymphoproliferation. Blood 2009; 114:
4002–8.
2 Bakker NA, van Imhoff GW,
Verschuuren EA, van Son WJ,
Homan van der Heide JJ, Veeger NJ
et al. Early onset post-transplant
lymphoproliferative disease is associated
with allograft localization. Clin Transplant
2005; 19: 327–34.
3 Quinlan SC, Pfeiffer RM, Morton LM,
Engels EA. Risk factors for early-onset
and late-onset post-transplant
lymphoproliferative disorder in U.S.
kidney recipients. Am J Hematol 2011;
86: 206–9.
4 Paranjothi S, Yusen RD, Kraus MD,
Lynch JP, Patterson GA, Trulock EP.
Lymphoproliferative disease after lung
transplantation: comparison of
presentation and outcome of early and
late cases. J Heart Lung Transplant 2001;
20: 1054–63.
5 Ghobrial IM, Habermann TM, Macon
WR, Ristow KM, Larson TS, Walker RC
et al. Differences between early and late
posttransplant lymphoproliferative
disorders in solid organ transplant
patients: are they two different diseases?
Transplantation 2005; 79: 244–7.
6 Wudhikarn K, Holman CJ, Linan M,
Blaes AH, Dunitz JM, Hertz ME et al.
7
8
9
10
11
ranial pressure. In summary, the current literature
suggests that in conjunction with reduction in immunosuppression, immune-chemotherapy should be first line
treatment due to its efficacy and minimisation of
neurotoxicity.
In our series, the prevalence of PCNS-PTLD was low.
PCNS-PTLD were high-grade, aggressive, monomorphic,
EBV-driven malignancies that presented with neurological symptoms and occurred late after transplantation.
Histology was required for diagnosis. PCNS-PTLD after
heart and/or lung transplantation may become more
common in the future, as recipients live longer due to
improvements in transplantation techniques and
posttransplant care. Although PCNS-PTLD is rare, it
should be included in the differential diagnosis of heart
and/or lung transplantation recipients who present with
neurological symptoms and signs. The management of
PCNS-PTLD is currently extrapolated from PCNSL and
PTLD as clinical trials are very difficult to conduct in this
patient population.
Post-transplant lymphoproliferative
disorders in lung transplant recipients:
20-yr experience at the University of
Minnesota. Clin Transplant 2011; 25:
705–13.
Phan TG, O’Neill BP, Kurtin PJ.
Post-transplant primary CNS lymphoma.
Neuro-Oncol 2000; 2: 229–38.
Cavaliere R, Petroni G, Lopes MB, Schiff
D, the International Primary Central
Nervous System Lymphoma
Collaborative Group. Primary central
nervous system post-transplantation
lymphoproliferative disorder. Cancer
2010; 116: 863–70.
Snanoudj R, Durrbach A, Leblond V,
Caillard S, Hurault De Ligny B, Noel C
et al. Primary brain lymphomas after
kidney transplantation: presentation and
outcome. Transplantation 2003; 76:
930–7.
Evens AM, Choquet S, Kroll-Desrosiers
AR, Jagadeesh D, Smith SM,
Morschhauser F et al. Primary CNS
posttransplant lymphoproliferative
disease (PTLD): an international
report of 84 cases in the modern
era. Am J Transplant 2013; 13:
1512–22.
Parker A, Bowles K, Bradley JA, Emery
V, Featherstone C, Gupte G et al.
Diagnosis of posttransplant
lymphoproliferative disorder in solid
organ transplant recipients – BCSH and
12
13
14
15
16
BTS guidelines. Br J Haematol 2010; 149:
674–95.
Tsai DE, Hardy CL, Tomaszewski JE,
Kotloff RM, Oltoff KM, Somer BG et al.
Reduction in immunosuppression as
initial therapy for posttransplant
lymphoproliferative disorder: analysis of
prognostic variables an long-term
follow-up of 42 adult patients.
Transplantation 2001; 71: 1076–88.
Ferreri AJM. How I treat primary
CNS lymphoma. Blood 2011; 118:
510–22.
Ferreri AJ, Reni M, Foppoli M, Martelli
M, Pangalis GA, Frezzato M et al.
High-dose cytarabine plus high-dose
methotrexate versus high-dose
methotrexate alone in patients
with primary CNS lymphoma: a
randomised phase 2 trial. Lancet 2009;
31: 1512–20.
Choquet S, Leblond V, Herbrecht R,
Socié G, Stoppa AM, Vandenberghe P
et al. Efficacy and safety of rituximab in
B-cell post-transplantation
lymphoproliferative disorders: results of
a prospective multicenter phase 2 study.
Blood 2006; 107: 3053–7.
Kikuchi A, Kawada H, Iwaki Y, Machida
S, Tsuchiya T, Fukuda R et al.
Measurement of rituximab
concentration in the cerebrospinal fluid
in CNS lymphoma. Rinsho Ketsueki 2004;
45: 1255–7.
© 2015 Royal Australasian College of Physicians
585
17 Rubenstein JL, Combs D, Rosenberg J,
Levy A, McDermott M, Damon L et al.
Rituximab therapy for CNS lymphomas:
targeting the leptomeningeal
compartment. Blood 2003; 101: 466–8.
18 Trappe R, Oertel S, Leblond V, Mollee P,
Sender M, Reinke P et al. Sequential
treatment with rituximab followed by
CHOP chemotherapy in adult B-cell
post-transplant lymphoproliferative
disorder (PTLD): the prospective
international multicentre phase 2
PTLD-1 trial. Lancet Oncol 2012; 13:
196–206.
19 Rubenstein JL, Fridlyand J, Abrey L,
Shen A, Karch J, Wang E et al. Phase I
study of intraventricular administration
of rituximab in patients with recurrent
CNS and intraocular lymphoma. J Clin
Oncol 2007; 25: 1350–6.
20 Bonney DK, Htwe EE, Turner A, Kelsey
A, Shabani A, Hughes S et al. Sustained
response to intrathecal rituximab in EBV
associated post-transplant
lymphoproliferative disease confined to
the central nervous system following
haematopoietic stem cell transplant.
Paediatr Blood Cancer 2012; 58:
459–61.
L E T T E R S TO T H E E D I TO R
Clinical-scientific notes
Bilateral cordotomy post-failure of
intrathecal analgesia in a palliative
care setting
A 60-year-old man was referred to a tertiary palliative
care unit with poorly controlled right hip pain.
He was diagnosed in 2009 with a large solitary right
hip metastasis from a poorly differentiated pulmonary
neuroendocrine tumour. Over a 5-year period he was
given seven lines of chemotherapy and three courses of
radiotherapy (total of 121 Gy in 63 fractions) to this
metastasis.
The patient’s analgesic regimens failed to stabilise his
pain, despite opioid rotations and recognised coanalgesics. Unfortunately, analgesic up-titration caused
increasing drowsiness, which limited his mobility. He also
had an aversion to hospitals, exacerbated by anxiety, but
increasing pain persuaded him to consent to inpatient
intrathecal analgesia. Ongoing support from his family
was vital during his admission.
An intrathecal catheter was inserted in late January
2014. While initially effective, the management lacked
sustained efficacy despite significant dose increases. The
infusion was ceased after 12 days, following a multidisciplinary plan for a cordotomy.
An open bilateral cordotomy via a T1/T2 laminectomy
was performed with an excellent result. The patient had
no right leg pain post-procedure, and only minor wound
pain; postoperative analgesia is outlined in Table 1.
Lower limb power remained intact; however, due to
deconditioning, he was limited to stand-transfers and
also developed urinary incontinence. Following a 5-week
period of rehabilitation, he was discharged home with his
family, remaining comfortable and with improved mobility. Unfortunately, after 2 months he deteriorated and
required a 2-week hospital admission for end-of-life care.
Despite the best systemic management of cancer
pain, up to 10% of patients have inadequate analgesia.1
Intrathecal catheter insertion and cordotomy are two
analgesic procedures appropriate for medically refractory
cancer pain. There is no current consensus of when and
in whom cordotomies should be performed. However,
medically refractory, unilateral nociceptive pain, where
prognosis is less than 1 year is deemed appropriate.2
Holistic multidisciplinary care is essential to manage pain
and recognise suitable cordotomy candidates.2,3 While
percutaneous cordotomy is recognised as an effective and
less invasive technique, the neurosurgical expertise at
our centre is in open cordotomy.4 The open cordotomy
procedure is performed under general anaesthetic via a
laminectomy technique. Open surgical exposure allows
mechanical interruption of the lateral spinothalamic tract
at the upper thoracic level.1,3 In contrast, the percutaneous procedure is performed under local anaesthetic,
where a radiofrequency heat lesion interrupts the lateral
spinothalamic tract at the C1/C2 level.4 Open cordotomy
is less commonly performed, causes more extensive
lesions and is less selective than the percutaneous procedure; however, it decreases the risk of respiratory compromise.1 Complications of both procedures include:
paresis or ataxia, worsening micturition control (as in the
present case), post-cordotomy dysesthesia and mirror
pain.5 The patient was appropriate for a bilateral
approach given his high risk of developing contralateral
© 2015 Royal Australasian College of Physicians
586
Letters to the Editor
Table 1 Analgesic requirements
Drugs
Oral morphine
equivalent dose†
Morphine (oral)
Morphine (subcut)
Morphine (intrathecal)
Hydromorphone (oral)
Hydromorphone (subcut)
Bupivocaine – intrathecal
Methadone – oral
Midazolam (subcut)
Midazolam (intrathecal)
Ketamine – CSCI
Diclofenac
Pre-intrathecal
maximum dose
622.5‡
Post-intrathecal
Pre-cordotomy
3 days
post-cordotomy
3307.75 mg§ (1489–9503 mg) 120 mg (90–180 mg)
60 mg
70 mg
—
—
—
—
75 mg
—
—
—
50 mg
—
11.25 mg (5–45 mg)
10 mg (4–30 mg)
—
12 mg (0 mg–12 mg)
150 mg (120–200 mg)
20 mg (20–40 mg)
20 mg (10–35 mg)
10 mg (5–10 mg)
200 mg¶ (0–200 mg)
50 mg (50–75 mg)
—
30 mg (20–40 mg)
—
—
2 mg (2–4 mg)
—
—
5 mg (0–5 mg)
—
150 mg (100–200 mg)
100 mg (50–100 mg)
2 weeks
post-cordotomy
EOLC – 3 months
post-cordotomy
176.5 mg (112–194 mg) 1230 mg (255–1500 mg)
—
20 mg (20–20 mg)
—
4.5 mg (1–8 mg)
—
—
20 mg (10–20 mg)
—
—
—
—
—
—
—
—
82 mg (17–100 mg)
—
—
47.5 mg (7.5–85 mg)
—
—
—
†Morphine – oral : subcut: intrathecal = 3:1:0.01; hydromorphone : morphine = 1:5; methadone oral : morphine oral 1:4.7 (Knotkova, Fine & Portenoy –
see references). ‡Calculated from admission notes – median dose not available. §All doses are presented as median dose and range unless otherwise
specified. ¶Started post-intrathecal failure. CSCI, continuous subcutaenous infusion; EOLC, end-of-life care.
pain requiring a further cordotomy.6 This case study
demonstrates the beneficial use of cordotomy, once
intrathecal and other non-procedural analgesia have
failed. Unfortunately, the availability of cordotomy
remains dependent on appropriately skilled neurosurgeons.2 Further research should better elucidate
the use of cordotomy in this important group of
patients.
References
1 Atkin N, Jackson KA, Danks RA. Bilateral
open thoracic cordotomy for refractory
cancer pain: a neglected technique?
J Pain Symptom Manage 2010; 39: 924–9.
2 Honey C, Yeomans W, Isaacs A, Honey
CM. The dying art of percutaneous
cordotomy in Canada. J Palliat Med 2014;
17: 624–8.
Received 10 September 2014; accepted 2 October 2014.
doi:10.1111/imj.12743
S. P. M. Hosking,1 M. E. Franco,1,2 P. Poon1,2,4 and
L. William1,2,3
1
Supportive and Palliative Care Unit, Monash Medical Centre,
Monash Health and 2Faculty of Medicine, Monash University and
3
Palliative Care Department, Eastern Health and 4Eastern Palliative
Care Association, Melbourne, Victoria, Australia
3 Viswanathan A, Bruera E. Cordotomy for
treatment of cancer-related pain: patient
selection and intervention timing.
Neurosurg Focus 2013; 35: E6.
4 Bain E, Hugel H, Sharma M.
Percutaneous cervical cordotomy for the
management of pain from cancer: a
prospective review of 45 cases. J Palliat
Med 2013; 16: 901–7.
5 Jones B, Finlay I, Ray A, Simpson B. Is
there still a role for open cordotomy in
cancer pain management? J Pain Symptom
Manage 2003; 25: 179–84.
6 Vissers KCP, Besse K, Wagemans M,
Zuurmond W, Giezeman MJ, Lataster A
et al. 23. Pain in patients with cancer.
Pain Pract 2011; 11: 453–75.
© 2015 Royal Australasian College of Physicians
587
Letters to the Editor
Serum creatinine is not the end-all,
be-all of renal impairment
A 52-year-old African American man with no medical
history presented as a referral to the nephrology clinic
for evaluation of renal dysfunction. The patient was an
active body builder in good health. He had blood pressure of 110/70 mmHg and heart rate of 70 b.p.m., and
his physical examination was remarkable for an
extremely muscular physique with no evidence of
central obesity. He weighed 220 lb with a body mass
index of 30.4 kg/m2 and body surface area of 2.25 m2.
He reported taking injectable testosterone cypionate
to build muscle for the past 1 year. Initial laboratory
measurements revealed a blood urea nitrogen (BUN) of
3.9 mmol/L, a serum creatinine of 141.4 μmol/L and an
estimated glomerular filtration rate (eGFR) of 50 mL/
min/1.73 m2.
Repeat serum BUN/creatinine/eGFR did not reveal a
significant variation from the initial evaluation of renal
function on day 1 (Table 1). Urinalysis was negative for
haematuria, proteinuria or pyuria. Since the patient’s
only abnormality was an elevated serum creatinine, we
used several methods to measure his ‘true GFR’. First, the
24-h urine collection revealed a creatinine clearance of
131 mL/min (urine volume 1500 mL). Second, the GFR
was estimated at 140.2 mL/min from a dynamic
radionuclide study by measuring Tc-99m DTPA uptake
within the kidneys. Last, cystatin C was measured at
Table 1 Laboratory parameters during clinic visit
BUN (mmol/L)
Creatinine (μmol/L)
Urine protein/creatinine ratio (mg/mmol)
24 h protein (mg/day)
24 h creatinine excretion (mmol)
24 h volume (L)
eGFR MDRD (mL/min/1.73 m2)
Tc GFR (mL/min)
24 h creatinine clearance (mL/min)
24 h creatinine clearance (mL/min/1.73 m2)
Cystatin C (mg/L)
Day 1
Day 5
Day 21
3.9
141.4
6.7
4.6
141.4
6.7
157.5
26.5
1.5
50.3
140
131
110
5.0
132.6
5.6
50.3
1 Wang ZM, Gallagher D, Nelson ME,
Matthews DE, Heymsfield SB. Total-body
skeletal muscle mass: evaluation of
24-h urinary creatinine excretion by
computerized axial tomography. Am J Clin
Nutr 1996; 63: 863–9.
Received 25 September 2014; accepted 22 October 2014.
57.5
doi:10.1111/imj.12733
1
S. K. Mallipattu, J. Ling2 and J. Uribarri3
0.76
BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate;
MDRD, modification of diet in renal disease.
References
0.76 mg/L (normal range 0.5–1.0 mg/L). These three
independent measurements of a normal GFR confirmed
our suspicion that this patient had an elevated serum
creatinine in the absence of renal impairment.
In clinical practice, a serum creatinine greater than the
upper limit of normal is taken as prima facie evidence of
decreased GFR and therefore impaired renal function.
The use of the modification of diet in renal disease
(MDRD) equation to estimate GFR strengthens this interpretation since a high serum creatinine will invariably
calculate a decreased GFR. The diagnosis of decreased
GFR will label the patient as having chronic kidney
disease, a diagnosis with significant clinical, psychological
and financial repercussions.
The rate of creatinine production is directly associated
with skeletal muscle mass and to a lesser extent with
meat intake. For instance, cross-sectional studies in
healthy adult males demonstrate that the mean ratio of
skeletal muscle mass to 24-h creatinine excretion is
21.8 kg/g.1 Also, the administration of 3 mg/kg/week of
testosterone increases the skeletal muscle mass and 24-h
creatinine excretion by 20% within a short 12-week
period.2 Furthermore, muscle mass is a strong determinant of eGFR in normal healthy male subjects, independent of body fat or protein intake.3 This is clearly
exaggerated in our patient who had a significantly large
muscle mass with a high 24-h creatinine excretion
(Table 1). This concept is not accounted for in the widely
used eGFR from the MDRD equation. In addition to
the radioisotope studies and 24-h creatinine clearance
measurements to measure GFR, cystatin C production is
a better marker of renal function in these clinical scenarios since its measurement is independent of age, sex
or muscle mass.4 The use of these other measures of GFR
can often be neglected in the primary care setting, leading
to the misdiagnosis of renal dysfunction in a given patient
with elevated serum creatinine.
1
Division of Nephrology, 2Department of Medicine, Stony Brook
University, and 3Division of Nephrology, Department of Medicine,
Icahn School of Medicine at Mount Sinai, New York, USA
2 Griggs RC, Kingston W, Jozefowicz RF,
Herr BE, Forbes G, Halliday D. Effect of
testosterone on muscle mass and muscle
protein synthesis. J Appl Physiol 1989; 66:
498–503.
3 Chew-Harris JS, Florkowski CM, Elmslie
JL, Livesey J, Endre ZH, George PM. Lean
mass modulates glomerular filtration rate
in males of normal and extreme body
composition. Intern Med J 2014; 44:
749–56.
4 Laterza OF, Price CP, Scott MG. Cystatin
C: an improved estimator of glomerular
filtration rate? Clin Chem 2002; 48:
699–707.
© 2015 Royal Australasian College of Physicians
588
Letters to the Editor
General correspondence
Audit of inpatient referrals
I was very interested in the recent article by Brown et al.1
comparing speciality referrals made from inpatient
general medical units in regional and metropolitan hospitals. The authors state that there is no available
research on inpatient referrals published. In fact, there is
one historical comparator which we published in the
1980s.2 This was a somewhat similar study but was done
over a 6-month period prospectively when two units,
both of which were general medical units with a special
interest in respiratory medicine, recorded all of their
inpatient and outpatient activity. There are some interesting comparisons with Brown et al.’s study. The Teaching Hospital unit in our study (consisting of one
consultant, one senior registrar, one registrar and one
intern) averaged 16.4 admissions per week compared
with Monash’s 11.6 and the District General Hospital’s
(one consultant, one registrar and one intern) 19.7
admissions as opposed to West Gippsland Hospital’s 10.7.
In our study, there was no difference in the range of
pathology seen between the two hospitals, but we also
found a significant difference between the Teaching Hospital and the District General Hospital in the number
of referrals made to other specialists. The Teaching Hospital averaged 0.26 referrals per admission (including
References
1 Brown MG, Campbell D, Maydom BW.
The undivided patient: a retrospective
geriatrics) compared with Monash’s 1.74 and the District
General Hospital’s 0.13 compared with West Gippsland’s
0.69.
I would suspect that the units involved in Brown’s et al.
study were more generously staffed than those in Yorkshire in the 1980s, and together with the much smaller
number of admissions, this documents the reduction in
clinical experience now offered to junior doctors. Brown
et al. correctly point out the dangers of over-referral and
speculate on possible causes including greater availability
of subspecialists and fear of litigation. Our feeling in the
1980s was that the disparity largely reflected better
general medical skills and greater confidence in dealing
with a wider variety of conditions among physicians
working in District General Hospitals rather than major
metropolitan teaching centres, together with a reluctance
to bother one’s busy colleagues with routine or banal
medical conditions which any physician should be
capable of managing.
Received 23 October 2014; accepted 31 October 2014.
doi:10.1111/imj.12692
G. Simpson
Thoracic Medicine, Cairns Base Hospital, Cairns, Queensland,
Australia
cohort analysis of speciality referrals
made from inpatient general medical
units comparing regional to metropolitan
practice. Intern Med J 2014; 44: 884–9.
Author reply
We thank Simpson1 for commenting on our paper2 and
also for alerting us to his historical comparator study3,
which has also compared inpatient referrals in a Teaching
Hospital and District General Hospital in the 1980s. It is
indeed interesting to note that this study also showed a
significant difference between the Teaching Hospital and
District General Hospital in the number of referrals made
to other specialists. We agree that in the 1980s, this
difference may have reflected greater confidence and
general medical skills in dealing with a wide variety of
conditions among physicians working in District General
Hospitals rather than major teaching centres.
It is certainly interesting to note the change in patient
load over the last two decades. We do not entirely agree
2 Simpson FG, Wilson J, Peake MD, Cooke
NJ. Audit of workload of physicians with
a special interest in respiratory medicine.
Br J Dis Chest 1987; 81: 150–4.
with Simpson that this correlates with a reduction on
clinical experience offered to junior doctors. There have
certainly been important moves in recent years to ensure
safer working hours for junior doctors that may in some
ways have limited clinical exposure compared with two
decades ago. However, we suspect that the change in
patient load is more a reflection on the increasing complexity and expectation of patients along with advances
in medicine, which have made management more
complex as the years go by.
Received 22 February 2015; accepted 22 February 2015.
doi:10.1111/imj.12741
M. Brown and D. Campbell
General Medicine, Monash Health, Melbourne, Victoria, Australia
© 2015 Royal Australasian College of Physicians
589
Letters to the Editor
References
1 Simpson G. Audit of inpatient referrals.
Intern Med J 2015; 45: 589.
2 Brown MG, Campbell D, Maydom BW.
The undivided patient: a retrospective
cohort analysis of speciality referrals
made from inpatient general medical
units comparing regional to metropolitan
practice. Intern Med J 2014; 44: 884–9.
3 Simpson FG, Wilson J, Peake MD, Cooke
NJ. Audit of work-load of physicians with
Leprosy and Australia
While syphilis has been hailed as ‘the great imitator’,
leprosy can also qualify, as illustrated by Turner et al.1 in
a summary of cases treated in a tertiary centre in Victoria.
The authors make the point that little has been published
about aspects of the disease in developed settings. Some
readers may be interested in two accounts of leprosy2,3 as
seen in New South Wales from the 1950s to the 1970s,
revealing confusion in clinical assessments and differential diagnoses in these patients. In this country, leprosy in
References
1 Turner D, McGuinness SL, Leder K.
Leprosy: diagnosis and management in a
its clinical manifestations is seen infrequently, and by
many clinicians not at all, so that the diagnosis is often
simply not considered.
Received 9 February 2015; accepted 10 February 2015.
doi:10.1111/imj.12745
C. R. Boughton
Division of Infectious Diseases, Prince of Wales Hospital, University
of New South Wales, Sydney, New South Wales, Australia
developed setting. Intern Med J 2015; 45:
109–12.
2 Boughton CR. Leprosy and Australia. Med
J Aust 1972; 2: 837–42.
Not only monoclonal antibodies . . .
The recent case report by Commons et al.1 suggests that
monoclonal antibody therapy was the triggering factor
for the development of melioidosis in a patient with
psoriatic arthritis.
As the physician caring for this patient at the time, I
feel the authors have jumped to a conclusion regarding
the possible connection with anti-tumour necrosis factor
and anti-IL12/23 treatment in this case. While this
patient’s spondylitis responded well to biologic therapy,
he also suffered from severe, extensive plaque psoriasis
that was largely unresponsive to treatment (aside
from a short lived initial improvement after etanercept
therapy).
Given the fact that there is a considerable (>100)
number of patients maintained on biologic therapy in
a special interest in respiratory medicine.
Br J Dis Chest 1987; 81: 150–4.
3 Boughton CR. Leprosy in Sydney. Med J
Aust 1977; 2: 351–3.
Darwin in the last 5–10 years and the fact that this is the
first case of melioidosis observed in this group, I remain
unconvinced that treatment was a triggering factor in this
case. Working in the garden during the wet season with
extensive psoriatic skin lesions and without the use of
protective gear seems to have been the most likely route
of inoculation. Therefore, in my opinion, it would have
been more appropriate to add active psoriasis to the
author’s list of risk factors for acquiring melioidosis in
endemic areas.
Received 15 January 2015; accepted 4 February 2015.
doi:10.1111/imj.12742
J. C. Nossent
School of Medicine and Pharmacology, The University of Western
Australia, Perth, Western Australia, Australia
Reference
1 Commons RJ, Grivas R, Currie BJ.
Melioidosis in a patient on monoclonal
antibody therapy for psoriatic arthritis.
Intern Med J 2014; 44: 1245–46.
© 2015 Royal Australasian College of Physicians
590
Letters to the Editor
Author reply
We thank Nossent1 for his response to our article on
melioidosis in a patient on monoclonal antibody therapy.2
Nossent raises the important point that skin exposure is a
common method of inoculation and patients with psoriatic skin disease are at heightened risk for inoculation. The
patient we reported sustained a recent cut while gardening after heavy rainfall, which was felt to be the likely time
of inocuation. As described in the Darwin Prospective
Melioidosis Study,3 20% of patients who contract
melioidosis do not have recognised risk factors, and we
acknowledge that any association with adalimumab in
this patient remains speculative.
However, there are several features that heighten our
suspicion that monoclonal antibody therapy may have
predisposed our patient to melioidosis. First, the rapid
onset presentation with bacteraemia is very similar to
patients who are immunosuppressed or receiving highdose corticosteroids and is rarely seen in otherwise well
patients.4
In addition, we have now had a second patient diagnosed with melioidosis who presented in a very similar
fashion while on adalimumab for ankylosing spondylitis.
This 68-year-old man presented early this year with 24 h
of headache, lethargy, nausea and high fevers, with onset
several days after exposure to wet season rains in the Top
End of the Northern Territory. He had chronic lung
disease but no other risk factors for melioidosis. Blood
cultures grew Burkholderia pseudomallei and he responded
well to standard therapy, initially with intravenous
ceftazidime.
References
1 Nossent JC. Not only monoclonal
antibodies . . . Intern Med J 2015; 45:
590.
2 Commons RJ, Grivas R, Currie BJ.
Melioidosis in a patient on monoclonal
antibody therapy for psoriatic arthritis.
Intern Med J 2014; 44: 1245–6.
3 Currie BJ, Ward L, Cheng AC. The
epidemiology and clinical spectrum of
Finally, animal data have demonstrated that neutralisation of interleukin-12 and tumour necrosis factor
(TNF)-α led to increased susceptibility to melioidosis,
and adalimumab acts as a TNF-α inhibitor.5 It is apparent
that specific risks for individual monoclonal antibody
therapies may only be identified through post-marketing
surveillance. In particular, the links between natalizumab
and progressive multifocal leukoencephalopathy6 and
TNF-α inhibitors and listeria7 have now been well
documented.
As noted by Nossent, melioidosis had not been documented in >100 patients on biological therapy in
Darwin until our report. However, it is possible that
adalimumab has a particular ability to interfere specifically with critical immune pathways in the host defence
against B. pseudomallei. While we cannot claim that
adalimumab was definitively a causative risk factor for
melioidosis in these two cases, we feel it is important to
highlight the potential link. Further surveillance for
melioidosis is warranted in patients being treated
with adalimumab and other monoclonal antibody
therapies who live in or travel to melioidosis-endemic
locations.
Received 4 March 2015; accepted 5 March 2015.
doi:10.1111/imj.12744
R. J. Commons,1 R. Hannah1,2 and B. J. Currie1,3
Departments of 1Infectious Diseases and Northern Territory
Medical Program and 2Immunology, Royal Darwin Hospital and
3
Global and Tropical Health Division, Menzies School of Health
Research, Darwin, Northern Territory, Australia
melioidosis: 540 cases from the 20 year
Darwin prospective study. PLoS Negl Trop
Dis 2010; 4: e900.
4 Currie BJ. Melioidosis: evolving concepts
in epidemiology, pathogenesis and
treatment. Semin Respir Crit Care Med
2015; 36: 111–25.
5 Santanirand P, Harley VS, Dance DA,
Drasar BS, Bancroft GJ. Obligatory role of
gamma interferon for host survival in a
murine model of infection with
Burkholderia pseudomallei. Infect Immun
1999; 67: 3593–600.
6 Langer-Gould A, Atlas SW, Green AJ,
Bollen AW, Pelletier D. Progressive
multifocal leukoencephalopathy in a
patient treated with natalizumab. N Engl J
Med 2005; 353: 375–81.
7 Bodro M, Paterson DL. Listeriosis in
patients receiving biologic therapies.
Eur J Clin Microbiol Infect Dis 2013; 32:
1225–30.
© 2015 Royal Australasian College of Physicians
591
Letters to the Editor
Redesign versus resources:
continuity lost
I read with interest the results of the redesign of General
Medical Units at Auckland Hospital in your journal.1
The reduction seen in length of stay and bed days saved
actually occurred when two simultaneous interventions
occurred, namely service redesign but in addition the
service attained increased staffing. So in this pre/posttrial design, it is difficult to ascertain which of these
interventions had the most effect. Clearly before 2011,
the Auckland Hospital General Medical Service was overwhelmed with patient numbers and therefore increasing
both senior and junior medical staffing 24 h/7 days a
week would no doubt assist with looking after the
patients in a more timely manner, improve educational
opportunities for junior medical staff, and there is no
medical department in the world that does not want
extra resourcing to improve morale.
All general medical units around the world are reviewing their model of care to improve patient care and efficiency and external drivers like the 4-h rule in Australia
and similar rules in New Zealand are key influences.
However, there are aspects of the new model of care
described in Auckland that trouble me greatly.
First, the authors should be congratulated in improving
senior consultant supervision during admitting days by
rostering physicians in the evening. This can only
improve patient care, junior medical staff supervision and
efficiency. The real concern I have is with the loss of
continuity of care that occurs with the introduction of
separate admitting and receiving teams as well as the lack
of single point accountability in decision making when
multiple consultants care for a particular patient. There
are many methods of load sharing without having to
break these two key tenets of good physician care.
Our acute inpatients are becoming more complex with
many comorbidities, polypharmacy, complex social and
functional issues, and thus it takes significant time and
effort to understand the nuances of each particular
patient. There are higher expectations in quality of care
from patients and relatives. We are entering the era of
personalised therapies. So good care is very difficult to
achieve when medical staff are changing during a very
short admission and there are multiple handovers.
There is another important factor. With good continuity of care one receives constant feedback on all of the
decisions made during the patient’s journey; not only the
big decisions but also the tiny small ones as well. We all
make hundreds of decisions every day. The outcomes of
your decision making may not be apparent by the next
day; it may take till discharge or review in the clinic or
with the readmission! This constant feedback over years
refines our consultant skills and I believe makes physicians a superior clinician compared with disciplines that
do not have this opportunity. It would be a great loss to
lose this intrinsic advantage.
Space precludes me from commenting on the ducking
and weaving that can occur when single point accountability is lost; or the hindrance to mentorship and role
modelling when junior staff have to deal with multiple
consultants with differences in opinions on the same
case; or the flawed claim of no change in case mix by
referencing a US study; or commenting on a 30-day mortality without any methodology or results presented in
the paper.
In conclusion, there is no evidence in this paper that
the model of care was the key driver in improving the
process indicator of length of stay. The patient you know
best is the one that you have admitted, and I want my
admitting registrar to be my discharging registrar.
Received 24 September 2014; accepted 25 September 2014.
doi:10.1111/imj.12734
C. P. Denaro
Department of Internal Medicine & Aged Care, Royal Brisbane &
Women’s Hospital, School of Medicine, University of Queensland,
Brisbane, Queensland, Australia
Reference
1 Toomath R, Szecket N, Nahill A, Denison
T, Spriggs D, Lay C et al. Medical service
redesign shares the load saving 6000 bed
days and improving morale. Intern Med J
2014; 44: 785–90.
© 2015 Royal Australasian College of Physicians
592
Letters to the Editor
Author reply
We are very pleased that Denaro took an interest in
carefully reading our paper.1
We agree that it is difficult to conclude which specific
components of our new work model contributed most
to the benefits we observed. If another department was
interested in reproducing these improvements to their
own length of stay (LOS), the simple solution would be to
adopt all of the components from our bundle of interventions. As such, we were careful in our conclusions to
list all of the component changes as being responsible for
the improvement in LOS.2 Having said this, we are confident that the reorganisation of our work model, rather
than our increase in staffing, was the major contributor.
Many studies have already shown that reconfiguration, rather than addition, of hospital resources
has significant effects on efficiency. It has been demonstrated that the on-call schedules of medical personnel
have strong effects on the variation in daily discharges,3
and removing such variation improves patient flow.4
Additionally, smoothing (not increasing) patient discharges over the course of the week increases capacity
in the emergency department.5 Finally, spreading the
load of daily admissions over the whole service reduces
median LOS.6 Therefore, our expectation of decreased
LOS from ‘line-averaging’, and stratification of the
patient journeys, was well founded.
Although the increase in Senior Medical Officer (SMO)
staffing was a requirement of the new model, the number
of SMO on clinical duties at any one time is unchanged;
we had 12 working SMO teams in the old model, and 12
in the new one. The addition of the SMO evening shift
References
1 Denaro CP. Redesign versus resources;
continuity lost. Intern Med J 2015; 45:
592.
2 Toomath R, Szecket N, Nahill A, Denison
T, Spriggs D, Lay C et al. Medical service
re-design shares the load saving 6000 bed
days and improving morale. Intern Med J
2014; 44: 785–90.
3 Wong H, Wu RC, Tomlinson G, Caesar M,
Abrams H, Carter MW et al. How much
actually occurred 6 months prior to the implementation
of the new work model, and was not associated with any
change in median LOS. Conversely, there was an
immediate step change in median LOS with the introduction of the new model. These observations suggest that
the re-configured inpatient team structure was largely
responsible for the benefits in LOS.
Denaro cherishes patient continuity with good reason.
It makes sense for the patient as well as for the continuing education and development of the training doctors.
However, the first problem is that true continuity is an
illusion. An audit in our own hospital in the old model
showed that 62% of patients were discharged by a different team than the one who did the admission. There
are many forces that disrupt continuity that we have
learned to manage, not least of which is SMO going off
service for annual leave. Geographic, unit-based teams
are another such force, one that is accompanied by many
benefits including better coordinated multidisciplinary
care. Finally, the change of ownership of a patient’s care
is an opportunity for better diagnostic performance, a
built-in system to provide a second opinion.
Notwithstanding this defence of our findings and our
work model, we are continuously searching for changes
that optimise the many positive principles at play that
make for better patient care, including doctor–patient
and physician–trainee continuity.
Received 5 March 2015; accepted 6 March 2015.
doi:10.1111/imj.12737
R. Toomath, N. Szecket and P. Poole
Auckland City Hospital, General Medicine, Auckland, New Zealand
do operational processes affect hospital
inpatient discharge rates? J Public Health
(Oxf) 2009; 31: 546–53.
4 Institute for Healthcare Improvement.
Optimizing patient flow: moving patients
smoothly through acute care settings.
2003 [cited 2015 Mar 3]; Available from
URL: http://www.ihi.org
5 Szecket N, Wong HJ, Wu RC, Berman
HD, Morra D. Implementation of a
continuous admission model reduces the
length of stay of patients on an internal
medicine clinical teaching unit. J Hosp
Med 2012; 7: 55–9.
6 Wong HJ, Wu RC, Caesar M, Abrams H,
Morra D. Smoothing inpatient discharges
decreases emergency department
congestion: a system dynamics simulation
model. Emerg Med J 2010; 27: 593–8.
© 2015 Royal Australasian College of Physicians
593
Letters to the Editor
The true prevalence of diabetes in
hospital patients and its implications
We read with interest the article by Cromarty et al.1 on
the effect of diabetes on hospital acquired conditions and
length of stay. They should be commended for addressing
issues of crucial and increasing importance to the modern
Australian health system. However, this paper raises
methodological issues that require careful consideration
if we are to interpret and apply its findings. The authors
acknowledge that their coding-based strategy (using a
pre-2012 coding standard that restricted recording diagnoses to ‘those conditions actively monitored or treated
in the episode’) could potentially underestimate diabetes
prevalence. However, the likely scale and importance of
this effect were not made explicit. In particular, their
reported 4.5% diabetes prevalence contrasts with rates of
15–35% in a recent cross-sectional diabetes prevalence
survey that used active case detection in inpatients at
11 Melbourne metropolitan hospitals.2 Our institution
Western Health (Footscray and Sunshine Hospitals),
which has similar catchment population demographics to
Northern Hospital (i.e. a rapidly expanding metropolitan
population with high proportions from non-Englishspeaking backgrounds and lower socioeconomic strata),3
had the highest prevalence (35.1% at Western Health).
We have also found similar rates (33%) in General
Medical patients, using retrospective methods employing
a post-2012 coding strategy to an administrative dataset.
Assuming prevalence at Northern to be similar to our
own, we estimate that their study may in fact have
wrongly defined as many as six out of seven patients with
diabetes as being ‘non-diabetic’. Therefore, the potential
for their study results to be subject to differential
misclassification bias is large. In particular, we wonder
whether this effect may have diluted observed differences
in study end-points between ‘diabetic’ and ‘non-diabetic’
groups. Notably, the ranking of the top six hospitalacquired diagnoses was the same in the ‘diabetes
References
1 Cromarty J, Parikh S, Lim WK, Acharya
S, Jackson TJ. Effects of hospital-acquired
conditions on length of stay for patients
with diabetes. Intern Med J 2014; 44:
1109–16.
with end-organ sequelae’ and ‘non-diabetes (Charlson
Comorbidity score ≥1)’ groups. Conversely, their allocation strategy could also have selected patients with more
advanced or complicated diabetes and therefore exaggerated between-group differences (such as the overall
much higher rate of hospital-acquired diagnoses in the
diabetic group). As the authors also acknowledge, their
pre-2012 coding definition for diabetes creates significant
issues of generalisability. How are we to identify prospectively and then apply future interventions only to inpatients whose diabetes is ‘actively monitored or treated in
the episode’? A simple, broad and inclusive definition
(i.e. ‘patients with known diabetes’) is much easier to
apply. We also question the appropriateness of analysis of
variance and linear regression to analyse length of stay
data (which usually has a highly skewed distribution).
The authors do not comment on whether necessary
assumptions (linearity, normality, independence and
homoscedasticity) were met.
Unfortunately, the sorts of research questions that
arise in this area are generally poorly amenable to conventional prospective research designs. By contrast,
retrospective studies utilising administrative datasets are
logistically feasible but throw up a host of methodological challenges. It is important that studies such as this
are published so that we can address these challenges
and work to improve study designs and analytical
methods.
Received 29 January 2015; accepted 3 February 2015.
doi:10.1111/imj.12746
1
G. Somarajah, H. Karunajeewa,1,2 P. S. Hamblin,3,4
E. Karahalios5 and E. Janus1,4
1
General Internal Medicine Unit and 3Endocrinology and Diabetes
Unit, Western Health, 4Department of Medicine, Western
Academic Centre and 5Centre for Epidemiology and Biostatistics,
Melbourne School of Population and Global Health, University of
Melbourne, and 2The Walter and Eliza Hall Institute of Medical
Research, Melbourne, Victoria, Australia
2 Bach LA, Ekinci EI, Engler D, Gilfillan C,
Hamblin PS, MacIsaac RJ et al. The high
burden of inpatient diabetes: the
Melbourne Public Hospitals Diabetes
Inpatient Audit. MJA 2014; 201: 334–8.
3 Victorian Department of Health. 2012
Local Government area Profiles: North and
West Metropolitan Region [Internet].
Melbourne: Victorian Government
Department of Health; 2012. [cited 2012
Dec 20]. Available from URL: http://docs
.health.vic.gov.au/docs/doc/North-and
-West-Metro-Region
© 2015 Royal Australasian College of Physicians
594
Letters to the Editor
Author reply
We agree with Somarajah et al.1 that our study2 should
not be used to estimate the prevalence of diabetes in the
inpatient population; however, this was not our aim. We
sought instead to estimate the rates and types of hospitalacquired diagnoses and their effects on length of stay for
patients identified with the most severe diabetes in
routine hospital data. Our study is clearly not a basis for
estimating prevalence of diabetes, which the contemporaneous paper by Bach et al.3 does very well.
As their letter points out, and as we acknowledge at
several points in our research report, the cases we identified were those where diabetes required active treatment or monitoring. As such, we did not include in our
sample those cases of diabetic patients controlled with
diet or oral medications. We do not consider this to be a
‘mis-classification’ of diabetes patients, but rather an
issue in generalising our findings to the broader diabetes
population in hospital.
However, we have every confidence that patients in
our sample with end-organ sequelae of diabetes or other
symptoms that require active in-hospital treatment are
clinically identifiable and were in fact correctly classified.
This, we believe, is the most valuable group to identify
and assess, as these patients attract the largest proportion
of health funding and hence are the group that has
the greatest potential for improvement. Interventions
focused on changing care of all diabetes patients may not
be useful or cost-effective. As the authors of the letter
note, the effect of any ‘under-identification’ of the
broader diabetes population would be a conservative one,
that is, to ‘dilute’ the differences between our sample
patients and the general inpatient population. We regret
References
1 Somarajah G, Karunajeewa H, Hamblin
PS, Karahalios E, Janus E. The true
prevalence of diabetes in hospital patients
and its implications. Intern Med J 2015;
45: 594.
2 Cromarty J, Parikh S, Lim WK,
Shamsunder A, Jackson TJ. Effects of
terming the latter the ‘non-diabetes group’ but found it
difficult to come up with a concise alternative.
This is a foundation study assessing the inpatient complications among the diabetic population, which aimed to
provide a basis for future in-depth analysis of these
patients. The next stage of our research is to collaborate
with clinicians on interventions to identify and reduce
occurrences of inpatient complications, and to introduce
hospital-based auditing systems to better identify patients
most at risk.
We did not extend the manuscript with a defence of
our statistical approach to the analysis. We rely on
Lumley et al.’s evidence from simulations4 that parametric assumptions can be relaxed for large public health
data sets such as ours, allowing use of analysis of variance
and linear regression on skewed data.
We wholeheartedly endorse the authors’ observation
that ‘studies such as this’ should be published to improve
study designs and analytical methods for research that is
not amenable to conventional prospective research
designs. We stand by our design and methods in finding
that patients with end organ sequelae of diabetes suffer
common hospital-acquired complications at 7–10 times
the rate of similarly comorbid patients and have lengths
of stay significantly extended both by their diabetes and
by their hospital-acquired complications.
Received 3 March 2015; accepted 11 March 2015.
doi:10.1111/imj.12750
1
1
J. E. Cromarty, S. Parikh, T. J. Jackson,1 W. K. Lim2
and S. Acharya3
1
Northern Clinical Research Centre and 2Aged Care Department,
The Northern Hospital, Melbourne, Victoria, and 3John Hunter
Hospital, Newcastle, New South Wales, Australia
hospital acquired conditions on length of
stay for diabetic patients. Intern Med J
2014; 44: 1109–16.
3 Bach LA, Ekinci EI, Engler D, Gilfillian C,
Hamblin PS, MacIsaac RJ et al. The high
burden of inpatient diabetes mellitus: the
Melbourne Public Hospitals Diabetes
Inpatient Audit. Med J Aust 2014; 201:
334–8.
4 Lumley T, Diehr P, Emerson S, Chen L.
The importance of the normality
assumption in large public health data
sets. Annu Rev Public Health 2002; 23:
151–69.
© 2015 Royal Australasian College of Physicians
595
Letters to the Editor
Systemic lupus erythematosus patients
and tertiary specialist care – simple
considerations dropping through the
cracks: osteoporosis monitoring as
an example
We read with interest the paper by Nikpour et al.1 noting
the lack of Australian epidemiological data for lupus
treatment.
As lupus and its treatments directly impact on osteoporosis, we expect that tertiary lupus specialists be
actively aware of this common comorbidity and their
patients’ routine bone monitoring investigations followed, whether these are conducted in hospital or externally by general practitioners. We recently reviewed rates
of osteoporosis monitoring in a tertiary hospital cohort of
well-defined systemic lupus erythematosus patients
under non-endocrinological specialist care, identifying
significant gaps in practice in this at-risk group.
The monitoring aspects were presence of documented
bone densitometry (DXA) and biochemical markers of
bone turnover to determine the level of active awareness
of this co-morbidity in our patient cohort.
Between 2002 and 2010, 2325 positive anti-dsDNA
antibody tests were performed by the state public pathology provider in Western Australia. Removing serial
results on the same patient, and patients not seen in the
tertiary hospital system, we identified 271 potential subjects, for whom 190 medical records were available for
review. This confirmed 103 subjects had systemic lupus
erythematosus and ongoing tertiary hospital care. Paediatric patients, patients not seen at least four times in the
tertiary clinics and patients with discoid lupus, druginduced lupus, rheumatoid arthritis, overlap syndrome
or inflammatory bowel disease were excluded.
Our population was mostly female (87.4%) with a
median age of 44 years (interquartile range 34.5–55.5
years) and rheumatological (82%), cutaneous (79%),
bone marrow (56%), renal (51%) or serosal (29%)
disease manifestations. This cohort was cared for by
immunology and/or rheumatology (87%), with renal
medicine (33%) or other specialities (dermatology,
haematology, neurology) in 12% of patients overall.
Reference
1 Nikpour M, Bridge JA, Richter S. A
systematic review of prevalence, disease
Overall, 30% of patients were under the care of more
than one treating team.
A single DXA was performed in 27% of subjects, and
more than once in 37% with no difference in frequency
between males and females. A comparison of the percentage of patients with multiple serum measures, as
opposed to a single measurement, of calcium (71% vs
88%), vitamin D (47% vs 63%), parathyroid hormone
(18% vs 37%) and fasting metabolic bone studies (14%
vs 23%) suggests that monitoring over time for these
indices may also be lacking. The relative frequency of
serial DXA scans correlated positively with the duration
of prednisolone treatment (normalised to time under
observation since 2002) (Spearman’s R = 0.51, P <
0.0001) indicating that awareness of glucocorticoidinduced osteoporosis may at least be present. If traditional risk factors for osteoporosis were documented,
then we noted a correlation with ordering of all monitoring tests at least once (R = 0.23, P = 0.03) or serially (R
= 0.32, P = 0.003); however, patients were not more
regularly monitored if under shared care compared with
being under a single non-endocrinological speciality (P =
0.9).
As the purpose of this audit was evaluation of tertiary
hospital care, there is potential bias in patient selection,
which may not reflect care provided outside of the
hospital setting; however, as an indicator of nonendocrinological specialist awareness and management
of a co-morbidity associated with active lupus, it indicates
management we can improve on in Western Australia.
Received 9 January 2015; accepted 31 January 2015.
doi:10.1111/imj.12748
M. Hew,1 E. J. McKinnon,2 B. Kirwin,3
O. P. Martinez3,4,5 and M. Lucas1,2,3
1
Department of Clinical Immunology, PathWest Laboratory
Medicine WA, Queen Elizabeth II Medical Centre, 2Institute for
Immunology and Infectious Diseases, Murdoch University, 3School
of Pathology and Laboratory Medicine, Faculty of Medicine,
Dentistry and Health Sciences, University of Western Australia,
4
Department of Clinical Immunology, Royal Perth Hospital, and
5
Department of Clinical Immunology, Fiona Stanley Hospital,
Perth, Western Australia, Australia
characteristics and management of
systemic lupus erythematosus in
Australia: identifying areas of unmet
need. Intern Med J 2014; 44: 1170–9.
© 2015 Royal Australasian College of Physicians
596
OnlineOpen
The Open Access option
for your research
Wiley offers an optional open access model: OnlineOpen, in over 1250
journals. OnlineOpen is available to authors who wish to make their article
available to non-subscribers on publication, or whose funding agency
requires grantees to archive the final version of their article.
Open access: freely available on Wiley Online Library
and PubMed Central
Fully compliant with open access mandates – meeting the
requirements of funding organizations and institutions
where these apply
Option available for over 1250 Wiley journals
An icon clearly signals that your article is OnlineOpen
Authors can publish OnlineOpen retroactively
For research articles, short communications and
review articles
wileyonlinelibrary.com/onlineopen
12-50104
For more information on OnlineOpen including details of fees visit:
WE MAKE YOUR RESEARCH EASY.
NOW WE MAKE JOB HUNTING EASY.
Let your partners in research energize your career.
Drawing on our expertise and relationships across the research and business communities,
Wiley-Blackwell invites you to join Wiley Job Network, the definitive job site for professionals
in the sciences, technology, business, finance, healthcare and the arts.
đƫ premium jobs from the most respected names in your industry
đƫ hundreds of recruiters and employers in your field
đƫ job alerts that match your criteria
đƫ expert career advice and candidate resources
Register and upload your resume/CV now to begin your job search!
wileyjobnetwork.com
available. For other pricing options, including access information and
terms and conditions, please visit: wileyonlinelibrary.com/access.
Publisher
Internal Medicine Journal is published by Wiley Publishing Asia
Pty Ltd
155 Cremorne Street
Richmond, Victoria, 3121
Australia
Tel: +61 3 9274 3100
Fax: +61 3 9274 3101
Journal Customer Services
For ordering information, claims and any enquiry concerning your
journal subscription please go to www.wileycustomerhelp.com/ask or
contact your nearest office.
Americas: Email: [email protected]; Tel: +1 781 388 8598 or 1 800 835
6770 (toll free in the USA & Canada).
Europe, Middle East and Africa: Email: [email protected]; Tel:
+44 (0) 1865 778315.
Asia Pacific: Email: [email protected]; Tel: +65 6511 8000.
Japan: For Japanese speaking support, email: [email protected]; Tel (toll
free): 005 316 50 480.
Visit our Online Customer Help available in 7 languages at www.
wileycustomerhelp.com/ask
Production Editor
Yvonne Lim (email: [email protected])
Information for Subscribers
Internal Medicine Journal, (ISSN: 1444-0903), is published monthly. Institutional subscriptions on prices for 2015 are: Print & Online: US$1746 (US),
US$2114 (Rest of World), @1370 (Europe). Prices are exclusive of tax. AsiaPacific GST, Canadian GST/HST and European VAT will be applied at the
appropriate rates. For more information on current tax rates, please go to
wileyonlinelibrary.com/tax-vat. The price includes online access from
current content and all online back files to January 1st 2011, where
Delivery Terms and Legal Title
Where the subscription price includes print issues and delivery is to the
recipient’s address, delivery terms are Delivered at Place (DAP); the recipient is responsible for paying any import duty or taxes. Title to all issues
transfers FOB our shipping point, freight prepaid. We will endeavour to
fulfil claims for missing or damaged copies within six months of publication, within our reasonable discretion and subject to availability.
Printing and Despatch
Printed in Singapore by Markono Print Media Pte Ltd.
All journals are normally despatched direct from the country in which they
are printed by surface air-lifted delivery.
Offprints
C.O.S. Printers Pte Ltd, 9 Kian Teck Crescent, Singapore 628875. Fax:
+65 6265 9074. Email: [email protected]
Back Issues
Single issues from current and recent volumes are available at the
current single issue price from [email protected]. Earlier issues
may be obtained from Periodicals Service Company, 11 Main Street,
Germantown, NY 12526, USA. Tel: +1 518 537 4700, Fax: +1 518 537 5899,
Email: [email protected]
Copyright and Copying
© 2015 Royal Australasian College of Physicians. All rights reserved. No
part of this publication may be reproduced, stored or transmitted in any
form or by any means without the prior permission in writing from the
copyright holder. Authorisation to copy items for internal and personal
use is granted by the copy-right holder for libraries and other users registered with their local Reproduction Rights Organisation (RRO), e.g. Copyright Clearance Center (CCC), 222 Rosewood Drive, Danvers, MA 01923,
USA (www.copyright.com), provided the appropriate fee is paid directly
to the RRO. This consent does not extend to other kinds of copying such
as copying for general distribution, for advertising or promotional pruposes, for creating new collective works or for resale. Special requests
should be addressed to [email protected]
IMJ.PI.DEC14
003-imj-v45-i5-ibc.indd 3
5/5/2015 3:15:58 PM
VOLUME 45
issue 5
May 2015
Editorial
Position Paper
471
563
Domestic violence: it is time for the medical
profession to play its part
L. Piterman, P. A. Komesaroff, H. Piterman and K. J. Jones
Review
474
Physical activity and sedentary behaviour:
applying lessons to chronic obstructive
pulmonary disease
K. Hill, P. A. Gardiner, V. Cavalheri, S. C. Jenkins
and G. N. Healy
Clinical Perspectives
482
Familial colorectal cancer
M. S. Lung, A. H. Trainer, I. Campbell and L. Lipton
Brief Communications
576
Original Articles
492
497
510
517
527
537
546
557
Clinical triage for colonoscopy is useful in
young women
K. D. Williamson, K. Steveling, G. Holtmann,
M. Schoeman and J. M. Andrews
Comparison of the management and in-hospital
outcomes of acute coronary syndrome patients
in Australia and New Zealand: results from the
binational SNAPSHOT acute coronary syndrome
2012 audit
C. Ellis, C. Hammett, I. Ranasinghe, J. French, T. Briffa,
G. Devlin, J. Elliott, J. Lefkovitz, B. Aliprandi-Costa,
C. Astley, J. Redfern, T. Howell, B. Carr, K. Lintern,
S. Bloomer, A. Farshid, P. Matsis, A. Hamer, M. Williams,
R. Troughton, M. Horsfall, K. Hyun, G. Gamble,
H. White, D. Brieger and D. Chew, on behalf of
Bi-National Acute Coronary Syndromes (ACS)
‘SNAPSHOT’ Audit Group
Assessing the use of initial oxygen therapy in
chronic obstructive pulmonary disease patients:
a retrospective audit of pre-hospital and
hospital emergency management
C. Susanto and P. S. Thomas
Obvious emphysema on computed tomography
during an acute exacerbation of chronic
obstructive pulmonary disease predicts a poor
prognosis
T. Cheng, H. Y. Wan, Q. J. Cheng, Y. Guo, Y. R. Qian,
L. Fan, Y. Feng, Y. Y. Song, M. Zhou, Q. Y. Li, G. C. Shi
and S. G. Huang
Efficacy of non-invasive mechanical ventilation
in the general ward in patients with chronic
obstructive pulmonary disease admitted for
hypercapnic acute respiratory failure and pH <
7.35: a feasibility pilot study
S. Fiorino, L. Bacchi-Reggiani, E. Detotto, M. Battilana,
E. Borghi, C. Denitto, C. Dickmans, B. Facchini,
R. Moretti, S. Parini, M. Testi, A. Zamboni, A. Cuppini,
L. Pisani and S. Nava
Prevalence and significance of CYP2C19*2 and
CYP2C191*7 alleles in a New Zealand acute
coronary syndrome population
P. D. Larsen, L. R. Johnston, A. Holley, A. C. La Flamme,
L. Smyth, E. W. Chua, M. A. Kennedy and S. A. Harding
Safety of coadministration of ezetimibe and
statins in patients with hypercholesterolaemia:
a meta-analysis
L. Luo, X. Yuan, W. Huang, F. Ren, H. Zhu, Y. Zheng
and L. Tang
Crescentic glomerulonephritis: data from the
Spanish Glomerulonephritis Registry
B. Quiroga, A. Vega, F. Rivera and J. M. López-Gómez,
on behalf of all members of the Spanish Registry of
Glomerulonephritis
001-imj-v45-i5-oc-4.44mm.indd 1
Consensus guidelines for the investigation and
management of encephalitis in adults and
children in Australia and New Zealand
P. N. Britton, K. Eastwood, B. Paterson, D. N. Durrheim,
R. C. Dale, A. C. Cheng, C. Kenedi, B. J. Brew, J. Burrow,
Y. Nagree, P. Leman, D. W. Smith, K. Read, R. Booy and
C. A. Jones, on behalf of the Australasian Society of
Infectious Diseases (ASID), Australasian College of
Emergency Medicine (ACEM), Australian and New
Zealand Association of Neurologists (ANZAN) and the
Public Health Association of Australia (PHAA)
580
583
Survey of infection control and antimicrobial
stewardship practices in Australian residential
aged-care facilities
R. L. Stuart, C. Marshall, E. Orr, N. Bennett, E. Athan,
D. Friedman and M. Reilly, on behalf of Members of
RACRIG (Residential Aged Care Research Interest Group)
Long-term follow up of paediatric liver
transplant recipients: outcomes following
transfer to adult healthcare in New Zealand
R. Harry, C. Fraser-Irwin, S. Mouat, E. Gane, S. Munn
and H. M. Evans
Primary central nervous system
posttransplantation lymphoproliferative
disorder after heart and lung transplantation
G. Gifford, K. Fay, A. Jabbour and D. D. Ma
Letters to the Editor
Clinical-scientific notes
586 Bilateral cordotomy post-failure of intrathecal
analgesia in a palliative care setting
S. P. M. Hosking, M. E. Franco, P. Poon and L. William
588 Serum creatinine is not the end-all, be-all of
renal impairment
S. K. Mallipattu, J. Ling and J. Uribarri
General correspondence
589 Audit of inpatient referrals
G. Simpson
589 Author reply
M. Brown and D. Campbell
590 Leprosy and Australia
C. R. Boughton
590 Not only monoclonal antibodies …
J. C. Nossent
591 Author reply
R. J. Commons, R. Hannah and B. J. Currie
592 Redesign versus resources: continuity lost
C. P. Denaro
593 Author reply
R. Toomath, N. Szecket and P. Poole
594 The true prevalence of diabetes in hospital
patients and its implications
G. Somarajah, H. Karunajeewa, P. S. Hamblin,
E. Karahalios and E. Janus
595 Author reply
J. E. Cromarty, S. Parikh, T. J. Jackson, W. K. Lim
and S. Acharya
596 Systemic lupus erythematosus patients and
tertiary specialist care – simple considerations
dropping through the cracks: osteoporosis
monitoring as an example
M. Hew, E. J. McKinnon, B. Kirwin, O. P. Martinez and
M. Lucas