improving uitnodiging diagnosis of bacterial infections€¦ · and bacterial infections is not...

218
Improving diagnosis of bacterial infections Research without gold standard Chantal van Houten

Upload: others

Post on 20-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Improving diagnosis of

bacterial infections

Improving diagnosis of bacterial infections

Chantal van Houten

Research without gold standard

Chantal van Houten

UITNODIGINGvoor het bijwonen van

de openbare verdedigingvan het proefschrift

Improving diagnosis of bacterial infections

Research without gold standard

door

Chantal van Houten

Op dinsdag 22 januari 2019om 14.30 uur

in het Academiegebouw,Domplein 29 te Utrecht

Aansluitend bent u van harte welkom op

de receptie ter plaatse

Chantal van HoutenPieter Saenredamstraat 11

3583 TA [email protected]

ParanImfenStephanie Smetsers

[email protected]

Hester de [email protected]

Page 2: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,
Page 3: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Improving diagnosis of bacterial infections

Research without gold standard

Chantal van Houten

15873_CvHouten_BNW_v2.indd 1 26-11-18 12:42

Page 4: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Improving diagnosis of bacterial infections.©2018, Chantal van Houten, Utrecht, The Netherlands

The copyright of the articles that have been published or accepted for publication has been transferred to the respective journals. All rights reserved. No part of this thesis may be reproduced, stored in a retrieval system or transmitted in any form or by any means without prior written permission of the author.

ISBN: 978-94-6375-154-4

Cover design: Esther Scheide - www.proefschriftomslag.nlLay-out: Nikki Vermeulen - Ridderprint BV Printing: Ridderprint BV - www.ridderprint.nl

Publication of this thesis was kindly supported by MeMed and PhD programme Infection & Immunity Utrecht.

15873_CvHouten_BNW_v2.indd 2 26-11-18 12:42

Page 5: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Improving diagnosis of bacterial infections

Research without gold standard

Verbetering van de diagnose van bacteriële infectiesOnderzoek zonder gouden standaard

(met een samenvatting in het Nederlands)

Proefschrift

ter verkrijging van de graad van doctor aan de Universiteit Utrecht

op gezag van de rector magnificus, prof. dr. H.R.B.M. Kummeling,

ingevolge het besluit van het college voor promotiesin het openbaar te verdedigen op 22 januari 2019 des middags te 2.30 uur

door

Chantal Brigitte van Houtengeboren op 11 april 1989 te Soest

15873_CvHouten_BNW_v2.indd 3 26-11-18 12:42

Page 6: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Promotoren: Prof. dr. L.J. Bont Prof. dr. E.A.M. Sanders

15873_CvHouten_BNW_v2.indd 4 26-11-18 12:42

Page 7: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Contents

Chapter 1 General introduction 7

Chapter 2 Antibiotic overuse in children with respiratory syncytial virus 21 lower respiratory tract infection Chapter 3 Antibiotic misuse in respiratory tract infections in children and 41 adults – a prospective, multicentre study (TAILORED Treatment) Chapter 4 A host-protein based assay to differentiate between bacterial 65 and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study Chapter 5 Optimizing prediction of serious bacterial infections by a 101 host-protein assay

Chapter 6 Observational multi-centre, prospective study to characterize 123 novel pathogen-and host-related factors in hospitalized patients with lower respiratory tract infections and/or sepsis – The “TAILORED-Treatment” study

Chapter 7 Reproducibility of an expert panel diagnosis as reference 141 standard in infectious diseases

Chapter 8 A new diagnostic strategy for children with lower respiratory 159 tract infections Chapter 9 Summary, general discussion and future perspectives 173

Chapter 10 Nederlandse samenvatting (Dutch summary) 193

Addendum Contributing authors 201 Dankwoord (Acknowledgments) 209 List of publications 213 Curriculum vitae 215

15873_CvHouten_BNW_v2.indd 5 26-11-18 12:42

Page 8: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

1General introduction

15873_CvHouten_BNW_v2.indd 6 26-11-18 12:42

Page 9: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

1General introduction

15873_CvHouten_BNW_v2.indd 7 26-11-18 12:42

Page 10: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 8 26-11-18 12:42

Page 11: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

1

General introduction

9

Infec tious diseases in childrenIn the Netherlands, emergency departments (EDs) are annually visited by 300,000 children younger than 14 years of age.1 At the age of 18 months, children have suffered on average from eight infective episodes, mostly respiratory tract infections (RTIs).2 At the ED, fever is therefore one of the main presenting problems and accounts for about 10-30% of all visits by children.3-5 Most acute febrile illnesses and RTIs are caused by self-limiting viral infections, which do not require antibiotic treatment. However, a minority develops a serious bacterial infection (SBI), such as meningitis, sepsis, pneumonia or urinary tract infection, for which timely diagnosis and targeted therapy are necessary to prevent harm.6 At the hospital ED about 15% to 20% of febrile children are diagnosed with an SBI.7,8 Although viral infections are usually self-limiting, it has been recognized for approximately a century that respiratory viruses predispose to bacterial superinfections.9 Mixed (i.e. viral and bacterial) infections pose a medical problem for several reasons. First, bacterial superinfections in viral respiratory disease are associated with antibiotic treatment failure.9 In 51% of the children with a dual otitis media infection, the infection persisted for up to 12 days after initiation of antibiotic treatment, compared to 35% of those with only bacterial otitis and to 19% in patients with viral otitis.10 Second, in case of severe illness physicians are afraid to miss secondary bacterial infections, even if a viral pathogen is found, and therefore start pre-emptive antimicrobial treatment, leading to high numbers of antibiotic overuse.11,12 Indicating that there is an urgent need for better diagnostics to distinguish between simple viral infections and bacterial co-infections.

D rivers of antibiotic prescriptionsChildren with fever and RTIs are a significant cause of concern for parents because of the impact on both the child and the family, and concerns about serious illnessess.13 Parents who make decisions about their child’s health take into account not only the health risks to their child, but also the risk of appearing to be a bad or irresponsible parent to others.14 As clinicians desire to maintain a good relationship with the parent, parental pressure is a strong predictor for antibiotic prescription.13 However, parents are also aware of the disadvantages of antibiotics and might be willing to work together with healthcare providers to improve appropriate use.15 Furthermore, repeated consultations for the same episode increase clinician anxiety that they might ‘missing something’ and make prescription of antibiotics more likely.13 The prescription of antibiotics is also influenced by clinical uncertainty, in many cases it is difficult to differentiate between bacterial and non-bacterial disease based on clinical judgment alone and available blood tests include a large range of uncertain test results. This range of uncertainty should be reduced to a minimum to make diagnostics useful for clinicians.

15873_CvHouten_BNW_v2.indd 9 26-11-18 12:42

Page 12: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Chapter 1

10

The burden of antibiotic misuse Clinical uncertainty regarding the diagnosis of the infection and parental influences lead to inappropriate use of antibiotics (Table 1). Antibiotics are prescribed almost twice as often as required in children with acute RTI in the US.16 Most antibiotics for children in the US are prescribed by paediatricians (39%) and family practitioners (15%), however the prescription of antibiotics depends on geographic region.17 Importantly, antibiotic overuse has been linked to the emergence of resistant strains of bacteria, causing an estimated 25 000 deaths in Europe annually.18,19 Besides health consequences, antibiotic overuse has several economic consequences due to direct cost of unnecessarily prescribed antibiotics, indirect costs for antibiotic-related adverse events and costs for treating patients with antibiotic-resistant bacteria.20 Antimicrobial resistance is a phenomenon considered to be “one of the world’s most pressing health problems in the 21st century” by the Center for Disease Control.21,22 Conversely, it is estimated that 22-47% of all patients with a bacterial infection fail to get antibiotic treatment within the recommended timeframe due to delayed or missed diagnosis. This delay leads to medical complications (e.g. organ dysfunction, acute mastoiditis, and increased mortality).23-26 The enormous health and economic consequences of antibiotic misuse highlight the high need for a solution that would empower physicians to use antibiotics more appropriately.

Table 1. Facts on antibiotic misuse.

Overuse

• Almost 50% of the children in the US with acute RTI receive antibiotics unnecessarily16

• Antibiotic overuse leads to:

o Preventable antibiotic related adverse events

o Antimicrobial resistant bacteria

· 25 000 deaths in Europe annually18,19

· Direct costs: 20 billion US$ per year in the US20

· Indirect costs: 35 billion US$ per year in the US20

Underuse

• 22-47% of the patients with a bacterial infection fail to get antibiotics on time23-25

• Antibiotic underuse leads to:

o Medical complications23-26

o Increased mortality: odds ratio 3.59 for PICU patients26

15873_CvHouten_BNW_v2.indd 10 26-11-18 12:42

Page 13: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

1

General introduction

11

Conventional diagnosticsCulture of microorganisms is the most definitive way to confirm bacterial infections. Unfortunately, this reference standard is time consuming, hampered by inaccessibility of the site of infection and influenced by several factors, including previous antibiotic usage. The search for simple and accurate diagnostic tools to differentiate between viral and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28, both clinical scores to predict bacterial infections in febrile children, were implemented to support physicians in the identification and management of children at risk of SBI. However, after the introduction of the conjugate vaccines against Haemophilus influenza type B, Neisseria meningitides type C, and Streptococus pneumonia, the incidence rates of SBIs (i.e. mainly sepsis, meningitis and pneumonia) significantly dropped and clinical presentations changed.29-31 Consequently, experience with serious bacterial infections declined, possibly making it even more difficult to recognize rare but serious infections, and an urgent need for new diagnostic research arose in the post-vaccination era.32

The ideal biomarker for bacterial infections should allow a rapid and accurate diagnosis at or near the site of care and facilitate cost effective decision making.33 Currently, white blood cell (WBC) count, C-reactive protein (CRP) level and procalcitonin (PCT) level are the most frequently used biomarkers of infection. The complex response to tissue injury, infection and inflammatory diseases is characterized by an increase in the synthesis of hepatic acute-phase proteins, including CRP.34 PCT, the precursor molecule of calcitonin, is a 116-amino-acid acute phase peptide and mainly synthesized by the thyroid.34 PCT is detectable in healthy neonates, but is low or undetectable in serum of other healthy children.35 The dynamics of PCT is comparable with CRP, however, the peak concentration of CRP is seen 36 hours after infection, whereas PCT reach the peak concentration after 8 hours.36 Unfortunately, both WBC, CRP and PCT cannot differentiate accurately enough between bacterial and viral infections for ruling in or ruling out SBI, even when combined with clinical signs and symptoms.37-40 A Dutch research group found a discriminative ability of 0.81-0.86 for a prediction model that combines clinical variables and CRP.40 Recently, they showed there are no differences in accuracy of the prediction model when CRP was replaced by PCT.41

A study by Oved and colleagues42 showed that a novel host–response-based diagnostic assay, ImmunoXpert, distinguishes bacterial infection from viral infection with high accuracy. This assay integrates the concentrations of three biomarkers with different dynamics: tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), interferon gamma induced protein-10 (IP-10), and CRP. This combination of biomarkers leads to a decrease in cases with an uncertain diagnosis. Unfortunately, cases with equivocal

15873_CvHouten_BNW_v2.indd 11 26-11-18 12:42

Page 14: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Chapter 1

12

test results remain. The diagnostic value of TRAIL is noteworthy since its dynamics are complementary to traditionally studied bacteria-induced proteins such as CRP and PCT; TRAIL concentrations increase in viral infection and decrease during bacterial infection. The increase of TRAIL in viral patients may be related to TRAIL’s role in programmed cell death associated with viral infections. The mechanism for its reduction in bacterial infections warrants further study.43 The protein IP-10 is secreted by several cell types in response to INF-γ 44 and has a function in chemo-attraction of monocytes and T-cells.45 It also plays a role in the response to chronic viral infections including hepatitis C viruses46, human immunodeficiency viruses47, as well as a broad range of acute viral infections.42

Inter ventions to minimize antibiotic overuseBesides the search for new biomarkers, there are several studies performed to investigate other strategies to reduce antibiotic overuse. Systematic reviews found that passive strategies targeting only parents, such as waiting room posters or pamphlets, do not appear to alter prescribing rates significantly.48,49 The most effective interventions involve targeting both parents and clinicians during a consultation. Examples of such interventions include automatic computer prompts for evidence-based prescribing, printed materials with personalised information and integration of interventions into routine clinical processes.48-50 Large randomised controlled trials show that social norm feedback and online training with an information booklet for parents can substantially reduce antibiotic prescribing at low cost and at national scale.51,52 In hospitals most interventions focus on antimicrobial stewardship programs.53,54 Successful programs have achieved a 20% increase in clinical cure rates and a 10%–15% reduction in treatment failures, while decreasing antimicrobial use by 20%–35%.55 Most hospitals have challenges in implementing centralized antimicrobial stewardship programs due to lack of dedicated personnel and lack of financial resources.53 Even when implemented, centralized approaches to stewardship may fail to affect the many episodes of antimicrobial use not subject to scrutiny by the stewardship team.53 In conclusion, there are many interventions to reduce antibiotic overuse with positive results, but, prescribing antibiotics only to patients who really need them, remains a large challenge.

Reference standard to diagnose bac terial infec tion if a gold standard is lackingThe most important challenge in diagnostic studies in the field of infectious diseases, is the lack of a gold standard. A final diagnosis is necessary to calculate the accuracy measures of the diagnostic test(s) or model(s) in question. Clinical suspicion confirmed by microbiological results is an approach often used, but this method can be hampered

15873_CvHouten_BNW_v2.indd 12 26-11-18 12:42

Page 15: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

1

General introduction

13

by technical issues such as inaccessibility of the infection site (e.g. middle-ear, sinusitis), culture contamination and uncertainty of the diagnosis due to colonisation. Therefore, the cohorts most often studied tend to include more severely ill patients. One potential solution to the lack of a gold standard is the use of panel diagnosis as reference standard, i.e., a group of experts who assess the results from multiple tests to reach a final diagnosis in each patient. The use of an expert panel diagnosis has the advantage of capturing a wider spectrum of illness severities in the resulting cohort—including difficult-to-diagnose cases—and is therefore more likely to be generalizable to clinical practice.56 The use of a panel diagnosis appears to be increasing.57 Unfortunately, there is neither theoretical evidence, nor practical guidance on the preferred methodology to conduct panel diagnoses. It is therefore not surprising that a review shows that methods of panel diagnosis varied substantially across studies.57

Aims and outlines of this thesisThe overarching aim of this thesis is to improve diagnostic care for children with possible bacterial infection in order to optimize antibiotic use in this group of patients. In young children most infections are caused by viral pathogens, whereas bacterial pathogens are the main cause of infection in adults. We expected the amount of antibiotic misuse, and thus the need for new diagnostics, to be highest in children. For that reason, this thesis focusses mainly on children, aiming to improve diagnosis of acute infections and to optimise management decisions for children with fever or RTI presenting to hospital settings.

The first aim of this thesis is to study antibiotic misuse in RTI. Previously mentioned percentages of antibiotic misuse are based on clinical suspicion with confirmation by microbiological results or national datasets with general codes. This methodology can result in a cohort with the more severally ill patients and the use of guidelines for assessing antibiotic misuse can result in contradictory analyses. Therefore, we aim to get insight in antibiotic misuse using an expert panel as reference standard. In chapter 2 the proportion of bacterial co-infections and the burden of antibiotic overuse is studied in children with RSV infections. As mentioned above, we expected that antibiotic overuse is a larger problem in children compared to adults. However, exact numbers are missing. In chapter 3 antibiotic misuse in children and adults with RTI are compared, using an expert panel to determine the aetiology of the infection (i.e. viral or bacterial).

The second aim is to address the need for better diagnostics to differentiate between viral and bacterial infections. As mentioned earlier in this general introduction, currently available diagnostics are often insufficient to differentiate between viral

15873_CvHouten_BNW_v2.indd 13 26-11-18 12:42

Page 16: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Chapter 1

14

and bacterial infections. A novel host–response-based diagnostic assay combining three different proteins showed promising results. However, these results are based on internal validation performed by the manufactory itself. Chapter 4 reports on the accuracy of this new diagnostic tool studied in the OPPORTUNITY study. This study is an international, external validation of the assay to differentiate between bacterial and viral infections in preschool children. Since clinical signs and symptoms play an important role in diagnosing febrile children, it is also important to evaluate potential added value of a new diagnostic test in combination with clinical features. In chapter 5 the above-mentioned combination assay is integrated in an existing clinical prediction model for febrile children presenting at the hospital. Chapter 6 describes a prospective, international study (TAILORED study) aiming to generate a multi-parametric model for distinguishing between bacterial and viral aetiologies in children and adults.

Third, this thesis aims to assess the reproducibility of an expert panel diagnosis as reference standard in infectious diseases diagnostic studies. It is well known that single expert diagnoses have poor interrater agreement and lead to an over- or underestimation of the diagnostic test under evaluation. To overcome these limitations, expert panel diagnoses have been used. In chapter 7 the results of a validation study of such an expert panel in an infectious diseases diagnostic study are described (IMPRIND study).

Using the results of this thesis, an overview of diagnostics for children with lower respiratory tract infections and suggestions for a future approach are provided in chapter 8. Finally, an overall discussion of the main findings of this thesis can be found in chapter 9.

15873_CvHouten_BNW_v2.indd 14 26-11-18 12:42

Page 17: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

1

General introduction

15

References1. Nationaal Kompas, Acute care numbers and context. Available from:

https://www.volksgezondheidenzorg.info/onderwerp/acute-zorg/cijfers-context/gebruik-acute-zorg#node-gebruik-seh-naar-leeftijd

2. Singhal A, Morley R, Abbott R, Fairweather-Tait S, Stephenson T, Lucas A. Clinical safety of iron-fortified formulas. Pediatrics 2000; 105(3): E38.

3. van Veen M, Steyerberg EW, Ruige M, et al. Manchester triage system in paediatric emergency care: prospective observational study. Bmj 2008; 337: a1501.

4. Armon K, Stephenson T, Gabriel V, et al. Determining the common medical presenting problems to an accident and emergency department. Archives of disease in childhood 2001; 84(5): 390-2.

5. Bouwhuis CB, Kromhout MM, Twijnstra MJ, Buller HA, Moll HA. Few ethnic differences in acute pediatric problems: 10 years of acute care in the Sophia Children’s Hospital in Rotterdam. Nederlands tijdschrift voor geneeskunde 2001; 145(38): 1847-51.

6. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013; 41(2): 580-637.

7. Van den Bruel A, Haj-Hassan T, Thompson M, Buntinx F, Mant D. Diagnostic value of clinical features at presentation to identify serious infection in children in developed countries: a systematic review. Lancet 2010; 375(9717): 834-45.

8. Van den Bruel A, Thompson MJ, Haj-Hassan T, et al. Diagnostic value of laboratory tests in identifying serious infections in febrile children: systematic review. BMJ 2011; 342: d3082.

9. Hament JM, Kimpen JL, Fleer A, Wolfs TF. Respiratory viral infection predisposing for bacterial disease: a concise review. FEMS immunology and medical microbiology 1999; 26(3-4): 189-95.

10. Korppi M, Katila ML, Jaaskelainen J, Leinonen M. Role of non-capsulated Haemophilus influenzae as a respiratory pathogen in children. Acta paediatrica 1992; 81(12): 989-92.

11. van Woensel JB, von Rosenstiel IA, Kimpen JL, Spanjaard L, van Aalderen WM. Antibiotic use in pediatric intensive care patients with lower respiratory tract infection due to respiratory syncytial virus. Intensive care medicine 2001; 27(8): 1436.

12. Chertow DS, Memoli MJ. Bacterial coinfection in influenza: a grand rounds review. Jama 2013; 309(3): 275-82.

13. Cabral C, Lucas PJ, Ingram J, Hay AD, Horwood J. “It’s safer to ...” parent consulting and clinician antibiotic prescribing decisions for children with respiratory tract infections: An analysis across four qualitative studies. Social science & medicine (1982) 2015; 136-137: 156-64.

14. Casiday RE. Children’s health and the social theory of risk: insights from the British measles, mumps and rubella (MMR) controversy. Social science & medicine (1982) 2007; 65(5): 1059-70.

15. Szymczak JE, Klieger SB, Miller M, Fiks AG, Gerber JS. What Parents Think About the Risks and Benefits of Antibiotics for Their Child’s Acute Respiratory Tract Infection. Journal of the Pediatric Infectious Diseases Society 2017.

16. Kronman MP, Zhou C, Mangione-Smith R. Bacterial prevalence and antimicrobial prescribing trends for acute respiratory tract infections. Pediatrics 2014; 134(4): e956-65.

15873_CvHouten_BNW_v2.indd 15 26-11-18 12:42

Page 18: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Chapter 1

16

17. Fleming-Dutra KE, Demirjian A, Bartoces M, Roberts RM, Taylor TH, Jr., Hicks LA. Variations in Antibiotic and Azithromycin Prescribing for Children by Geography and Specialty-United States, 2013. Pediatr Infect Dis J 2018; 37(1): 52-8.

18. Laxminarayan R, Duse A, Wattal C, et al. Antibiotic resistance-the need for global solutions. Lancet Infect Dis 2013; 13(12): 1057-98.

19. ECDC Technical Report: The bacterial challenge: time to react, Sept 2009. Available from: http://ecdc.europa.eu/en/publications/Publications/0909_TER_The_Bacterial_Challenge_Time_to_React.pdf (accessed 11-8-2016)

20. WHO: Antibiotic resistance; frequently asked question. 2011. Available from: http://www.euro.who.int/en/health-topics/disease-prevention/antimicrobial-resistance/antibiotic-resistance/frequently-asked-questions (accessed 7-7-2017).

21. CDC: About Antimicrobial Resistance. Available from: https://www.cdc.gov/drugresistance/about.html (accessed 7-7-2017).

22. Arias CA, Murray BE. Antibiotic-resistant bugs in the 21st century--a clinical super-challenge. N Engl J Med 2009; 360(5): 439-43.

23. Craig JC, Williams GJ, Jones M, et al. The accuracy of clinical symptoms and signs for the diagnosis of serious bacterial infection in young febrile children: prospective cohort study of 15 781 febrile illnesses. BMJ 2010; 340: c1594.

24. Metersky ML, Sweeney TA, Getzow MB, Siddiqui F, Nsa W, Bratzler DW. Antibiotic timing and diagnostic uncertainty in Medicare patients with pneumonia: is it reasonable to expect all patients to receive antibiotics within 4 hours? Chest 2006; 130(1): 16-21.

25. Van Zuijlen DA, Schilder AG, Van Balen FA, Hoes AW. National differences in incidence of acute mastoiditis: relationship to prescribing patterns of antibiotics for acute otitis media? Pediatr Infect Dis J 2001; 20(2): 140-4.

26. Weiss SL, Fitzgerald JC, Balamuth F, et al. Delayed antimicrobial therapy increases mortality and organ dysfunction duration in pediatric sepsis. Crit Care Med 2014; 42(11): 2409-17.

27. McCarthy PL, Sharpe MR, Spiesel SZ, et al. Observation scales to identify serious illness in febrile children. Pediatrics 1982; 70(5): 802-9.

28. Dagan R, Powell KR, Hall CB, Menegus MA. Identification of infants unlikely to have serious bacterial infection although hospitalized for suspected sepsis. J Pediatr 1985; 107(6): 855-60.

29. Thigpen MC, Whitney CG, Messonnier NE, et al. Bacterial meningitis in the United States, 1998-2007. N Engl J Med 2011; 364(21): 2016-25.

30. Gladstone RA, Jefferies JM, Faust SN, Clarke SC. Continued control of pneumococcal disease in the UK - the impact of vaccination. Journal of medical microbiology 2011; 60(Pt 1): 1-8.

31. Chandran A, Watt JP, Santosham M. Prevention of Haemophilus influenzae type b disease: past success and future challenges. Expert review of vaccines 2005; 4(6): 819-27.

32. Gill PJ, Goldacre MJ, Mant D, et al. Increase in emergency admissions to hospital for children aged under 15 in England, 1999-2010: national database analysis. Archives of disease in childhood 2013; 98(5): 328-34.

33. Keitel K, Lacroix L, Gervaix A. Point-of-care Testing in Pediatric Infectious Diseases. Pediatr Infect Dis J 2018; 37(1): 108-10.

34. Hedlund J, Hansson LO. Procalcitonin and C-reactive protein levels in community-acquired pneumonia: correlation with etiology and prognosis. Infection 2000; 28(2): 68-73.

15873_CvHouten_BNW_v2.indd 16 26-11-18 12:42

Page 19: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

1

General introduction

17

35. van Rossum AM, Wulkan RW, Oudesluys-Murphy AM. Procalcitonin as an early marker of infection in neonates and children. Lancet Infect Dis 2004; 4(10): 620-30.

36. Simon L, Gauvin F, Amre DK, Saint-Louis P, Lacroix J. Serum procalcitonin and C-reactive protein levels as markers of bacterial infection: a systematic review and meta-analysis. Clin Infect Dis 2004; 39(2): 206-17.

37. Principi N, Esposito S. Biomarkers in Pediatric Community-Acquired Pneumonia. International journal of molecular sciences 2017; 18(2).

38. Baer G, Baumann P, Buettcher M, et al. Procalcitonin guidance to reduce antibiotic treatment of lower respiratory tract infection in children and adolescents (ProPAED): a randomized controlled trial. PLoS One 2013; 8(8): e68419.

39. Kapasi AJ, Dittrich S, Gonzalez IJ, Rodwell TC. Host Biomarkers for Distinguishing Bacterial from Non-Bacterial Causes of Acute Febrile Illness: A Comprehensive Review. PLoS One 2016; 11(8): e0160278.

40. Nijman RG, Vergouwe Y, Thompson M, et al. Clinical prediction model to aid emergency doctors managing febrile children at risk of serious bacterial infections: diagnostic study. BMJ 2013; 346: f1706.

41. Nijman RG, Vergouwe Y, Moll HA, et al. Validation of the Feverkidstool and procalcitonin for detecting serious bacterial infections in febrile children. Pediatric research 2017.

42. Oved K, Cohen A, Boico O, et al. A Novel Host-Proteome Signature for Distinguishing between Acute Bacterial and Viral Infections. PLoS One 2015; 10(3): e0120012.

43. Falschlehner C, Schaefer U, Walczak H. Following TRAIL’s path in the immune system. Immunology 2009; 127(2): 145-54.

44. Luster AD, Unkeless JC, Ravetch JV. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature 1985; 315(6021): 672-6.

45. Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD. IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. Journal of immunology (Baltimore, Md : 1950) 2002; 168(7): 3195-204.

46. Romero AI, Lagging M, Westin J, et al. Interferon (IFN)-gamma-inducible protein-10: association with histological results, viral kinetics, and outcome during treatment with pegylated IFN-alpha 2a and ribavirin for chronic hepatitis C virus infection. The Journal of infectious diseases 2006; 194(7): 895-903.

47. Falconer K, Askarieh G, Weis N, Hellstrand K, Alaeus A, Lagging M. IP-10 predicts the first phase decline of HCV RNA and overall viral response to therapy in patients co-infected with chronic hepatitis C virus infection and HIV. Scandinavian journal of infectious diseases 2010; 42(11-12): 896-901.

48. Andrews T, Thompson M, Buckley DI, et al. Interventions to influence consulting and antibiotic use for acute respiratory tract infections in children: a systematic review and meta-analysis. PLoS One 2012; 7(1): e30334.

49. Vodicka TA, Thompson M, Lucas P, et al. Reducing antibiotic prescribing for children with respiratory tract infections in primary care: a systematic review. The British journal of general practice : the journal of the Royal College of General Practitioners 2013; 63(612): e445-54.

50. Blair PS, Turnbull S, Ingram J, et al. Feasibility cluster randomised controlled trial of a within-consultation intervention to reduce antibiotic prescribing for children presenting to primary care with acute respiratory tract infection and cough. BMJ open 2017; 7(5): e014506.

15873_CvHouten_BNW_v2.indd 17 26-11-18 12:42

Page 20: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Chapter 1

18

51. Hallsworth M, Chadborn T, Sallis A, et al. Provision of social norm feedback to high prescribers of antibiotics in general practice: a pragmatic national randomised controlled trial. Lancet 2016; 387(10029): 1743-52.

52. Dekker ARJ, Verheij TJM, Broekhuizen BDL, et al. Effectiveness of general practitioner online training and an information booklet for parents on antibiotic prescribing for children with respiratory tract infection in primary care: a cluster randomized controlled trial. The Journal of antimicrobial chemotherapy 2018.

53. Doron S, Davidson LE. Antimicrobial stewardship. Mayo Clinic proceedings 2011; 86(11): 1113-23.

54. Hamilton KW, Gerber JS, Moehring R, et al. Point-of-prescription interventions to improve antimicrobial stewardship. Clin Infect Dis 2015; 60(8): 1252-8.

55. Septimus EJ, Owens RC, Jr. Need and potential of antimicrobial stewardship in community hospitals. Clin Infect Dis 2011; 53 Suppl 1: S8-s14.

56. Reitsma JB, Rutjes AW, Khan KS, Coomarasamy A, Bossuyt PM. A review of solutions for diagnostic accuracy studies with an imperfect or missing reference standard. J Clin Epidemiol 2009; 62(8): 797-806.

57. Bertens LC, Broekhuizen BD, Naaktgeboren CA, et al. Use of expert panels to define the reference standard in diagnostic research: a systematic review of published methods and reporting. PLoS Med 2013; 10(10): e1001531.

15873_CvHouten_BNW_v2.indd 18 26-11-18 12:42

Page 21: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 19 26-11-18 12:42

Page 22: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

2Antibiotic overuse in children with respiratory syncytial virus lower respiratory tract Infection

CB van Houten, CA Naaktgeboren, BJM Buiteman, M van der Lee, A Klein, I Srugo, I Chistyakov, W de Waal, CB Meijssen, PW Meijers, KM de Winter-de Groot, TFW Wolfs, Y Shachor-Meyouhas, M Stein, EAM Sanders and LJ Bont

Pediatr Infect Dis J. 2018 Nov;37(11):1077-1081

15873_CvHouten_BNW_v2.indd 20 26-11-18 12:42

Page 23: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

2Antibiotic overuse in children with respiratory syncytial virus lower respiratory tract Infection

CB van Houten, CA Naaktgeboren, BJM Buiteman, M van der Lee, A Klein, I Srugo, I Chistyakov, W de Waal, CB Meijssen, PW Meijers, KM de Winter-de Groot, TFW Wolfs, Y Shachor-Meyouhas, M Stein, EAM Sanders and LJ Bont

Pediatr Infect Dis J. 2018 Nov;37(11):1077-1081

15873_CvHouten_BNW_v2.indd 21 26-11-18 12:42

Page 24: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

22

Chapter 2

Abstrac tBackground Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infections (LRTI) during the first year of life. Antibiotic treatment is recommended in cases suspected of bacterial coinfection. The aim of this prospective study was to estimate the incidence of bacterial coinfections and the amount of antibiotic overuse in children infected with RSV using expert panel diagnosis.

MethodsChildren 1 month of age and over with LRTI or fever without source were prospectively recruited in hospitals in the Netherlands and Israel. Children with confirmed RSV infection by Polymerase Chain Reaction (PCR) on nasal swabs were evaluated by an expert panel as reference standard diagnosis. Three experienced paediatricians distinguished bacterial coinfection from simple viral infection using all available clinical information, including all microbiologic evaluations and a 28-day follow-up evaluation.

ResultsA total of 188 children (24% of all 784 recruited patients) were positive for RSV. From these, 92 (49%) were treated with antibiotics. All 27 children (29%) with bacterial coinfection were treated with antibiotics. Fifty-seven patients (62%) were treated with antibiotics without a diagnosis of bacterial coinfection. In 8 of the 92 (9%), the expert panel could not distinguish simple viral infection from bacterial coinfection.

Conclusion This is the first prospective international multicentre RSV study using an expert panel as reference standard to identify children with and without bacterial coinfection. All cases of bacterial coinfections are treated, whereas as many as one-third of all children with RSV LRTI are treated unnecessarily with antibiotics.

15873_CvHouten_BNW_v2.indd 22 26-11-18 12:42

Page 25: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

23

2

Antibiotic overuse in children with RSV

Introduc tionRespiratory syncytial virus (RSV) is the most common cause of viral lower respiratory tract infections (LRTI) during the first year of life. In winter seasons, 20% of hospitalizations and 18% of emergency department visits for acute respiratory infections of young children is associated with RSV infection.1 A Cochrane review found insufficient evidence to support the use of antibiotics for bronchiolitis.2 Accordingly, antibiotics are not recommended for RSV LRTI unless there is clear evidence of secondary bacterial disease or respiratory failure.2 There is a large variation in the reported percentages of bacterial coinfection in RSV patients, though most studies report less than 11%.3–10 Despite the low incidence of bacterial coinfections in patients infected with RSV, antibiotic prescriptions range between 29% and 80%.3,4,9–12 Antibiotic overuse is associated with an expansion in antibiotic resistance. Yearly the numbers of patients dying as a direct result of infections caused by antibiotic-resistant microorganisms in the United States and Europe are 23,000 and 25,000, respectively. The total economic burden is estimated to be $20 billion direct health care costs and $35 billion a year additional costs for lost productivity in the United States.13,14 The aim of the current prospective, international multicentre study, was to investigate the frequency of bacterial coinfections, and antibiotic overuse in children with RSV infections using expert panel diagnosis on bacterial causes of disease as the reference standard.

MethodsSetting and participantsThe patient population consisted of 2 similar prospective cohort studies, OPPORTUNITY and TAILORED. The aim of both studies was to differentiate bacterial from viral infections. Participants of OPPORTUNITY were described previously.15 Patient recruitment for both studies took place at the emergency department and ward of 3 secondary and 1 university hospital in the Netherlands and 2 secondary hospitals in Israel. In both studies, children 1 month of age and older presenting with fever (peak temperature ≥ 38.0°C) or symptoms of LRTI were eligible for inclusion. Patients were excluded in case of a previous episode of fever in the past 3 weeks, psychomotor retardation, moderate-to-severe metabolic disorder, primary or secondary immunodeficiency, HIV-, HBV- or HCV infection and active malignancies. Antibiotic use before inclusion was noted. Parents gave written informed consent before sampling. The studies are registered on ClinicalTrials.gov, NCT01931254 and NCT02025699 and were approved by the ethics committees in the participating countries.

15873_CvHouten_BNW_v2.indd 23 26-11-18 12:42

Page 26: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

24

Chapter 2

Data collectionWithin 24 hours after presentation, a nasal swab (Universal Transport Medium, Copan) was collected for multiplex RT-PCR testing of the most common respiratory pathogens, including RSV and stored at ˗80°C. The swabs were performed for research and were analysed when patient recruitment was finished; therefore, the attending physician was not informed about the results of the nasal swab. Samples from the OPPORTUNITY study were transferred to MeMed Diagnostics (Tirat Carmel, Israel) (RV15 test kit, Seegene, KR), samples from the TAILORED study were transferred to the laboratory of Utrecht University Hospital (Magna Pure 96 DNA and Viral NA Large Volume and Magna Pure LC Total Nucleic Acid Isolation Kit, both Roche). After 28 days, we took a questionnaire for clinical follow-up. Clinical data were collected from medical records using an electronic Case Report Form (eCRF). The eCRF consisted of demographics, medical history, physical examination findings, radiological and laboratory data, microbiological test results (including study-specific nasal swab data) and follow-up information.

Reference outcomeCurrently, no single reference standard test exists to determine the aetiology of an infection.16 Therefore, we followed the England’s National Health Service’s standard for evaluating diagnostic tests and composed an expert panel reference standard.17 We have recently reported on the use of an expert panel as reference standard.15 The panel comprised paediatricians with at least 10 years of clinical experience. Every recruited patient was diagnosed by 3 panel members using all available eCRF information, including the 28-day follow-up, but blinded for the labels of their peers. Each expert assigned 1 of the following aetiologies to each patient: bacterial infection, viral infection, bacterial coinfection (i.e., viral and bacterial coinfection) or indeterminate. When at least 2 panel members assigned the same aetiology, that aetiology was considered to be present. Patients were assigned an inconclusive outcome if each panel member assigned a different aetiology or when at least 2 panel members diagnosed the patients as indeterminate. By design, these patients could not be used for the objectives of this study.

Statistical analysisChildren were first stratified according to the result of the Polymerase Chain Reaction (PCR) test for RSV. Thereafter, we stratified the children according to the reference diagnosis (i.e., simple viral, bacterial coinfection or inconclusive) and studied the use of antibiotic treatment for every reference standard outcome separately. Sub-cohort analyses were performed on patients admitted to the paediatric intensive care unit (PICU), per age group and for the Dutch and Israeli cohort separately. For baseline

15873_CvHouten_BNW_v2.indd 24 26-11-18 12:42

Page 27: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

25

2

Antibiotic overuse in children with RSV

characteristics, univariate comparisons were performing Fisher exact test, t tests and Mann-Whitney test as appropriate. Statistical analysis was performed by using SPSS version 21.0 for Windows (IBM Corp., Armonk, NY). A P value <0.05 was considered statistically significant.

ResultsPatient characteristicsA total of 784 children were recruited between October 2013 and May 2016 (Figure 1). In 188 (24%) patients, the nasal swab was positive for RSV. The median age of RSV patients was significantly lower when compared with RSV-negative patients (Supplemental Table 1). Of these RSV patients, median age was 9 months, and the majority (60%) of the patients were boys (Table 1). As shown in Supplemental Table 2a, there were some differences between RSV patients recruited in the different countries. When comparing patients recruited in secondary care centres only, the cohorts were more similar (Supplemental Table 2b). In RSV patients with and without antibiotic treatment, many other viruses were detected, rhinovirus was most common (Supplemental Table 3).

Bacterial co-infection during RSV LRTIThe expert panel assigned reference standard outcomes to all 188 RSV patients (Figure 1) of which 27 (14%) were diagnosed with bacterial coinfection. In 10 (5%) patients, the reference standard outcome was inconclusive; clinical characteristics are shown in Table 1. At presentation, patients with bacterial coinfection appeared more ill than RSV patients without bacterial coinfection (P < 0.001; Table 1). RSV patients with bacterial coinfection were significantly more often admitted at the PICU compared with patients diagnosed with viral infection (63% vs. 7%, respectively; P < 0.001). When looking at the cohort in which all 3 expert panels agree on the diagnosis, (unanimous cohort) 16 (9%) of the patients was diagnosed with a bacterial coinfection (Supplemental Table 4).

Use of antibioticsOf all 188 RSV patients, 92 (49%) patients were treated with antibiotics (Figure 1). Amoxicillin/clavulanate was prescribed most often in the Netherlands; in Israel, most children received amoxicillin (Supplemental Table 5). All 27 RSV patients with a bacterial coinfection were treated with antibiotics. The most common bacterial diagnoses are shown in Table 1. A bacterial pathogen was detected in only 5 (19%) of the patients. Antibiotics were also administered to 57 (30%) RSV patients with viral infection as reference standard outcome (Figure 1). Of them, 54 (95%) received antimicrobial treatment within 72 hours after presentation at the hospital (Figure 1). Even when using

15873_CvHouten_BNW_v2.indd 25 26-11-18 12:42

Page 28: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

26

Chapter 2

our more strict unanimous reference standard, 40 patients (21%) without bacterial coinfection received antibiotics (Supplemental Table 3). Off all RSV patients with a simple viral LRTI receiving antibiotics, 15 (33%) patients had pneumonia and 29 (63%) bronchiolitis as clinical syndrome. Eight RSV children with viral upper respiratory tract infection received also antibiotics (1 case of tonsillitis, 7 cases with non-specified upper respiratory tract infection). In RSV patients without a bacterial coinfection, patients treated with antibiotics were more often admitted to the hospital and had more often pulmonary infiltrates on chest x-ray than patients without antibiotics (Table 2).

Total

recruited

n=784

Nasal

swab

RSV +

n=188

Nasal

swab

RSV -

n=596

Reference

standard

Inconclusive*

n=10

Reference

standard

Bacterial

co-infection

n=27

Reference

standard

Simple viral

infection

n=151

AB +

n=57

AB -

n=94

AB +

n=27

AB -

n=0

AB +

n=8

AB -

n=2

Primary

(<72 hours)

n=54

Secondary

(>72 hours)

n=3

Figure 1. Recruitment and flow of children presenting at the hospital with fever without source and acute respiratory tract infections. *Inconclusive expert panel diagnosis was assigned to patients where each panel member gave a different diagnosis or where 2 or more members diagnosed the patient as indeterminate. AB: antibiotics.

15873_CvHouten_BNW_v2.indd 26 26-11-18 12:42

Page 29: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

27

2

Antibiotic overuse in children with RSV

Table 1. Baseline characteristics RSV cohort.

Totaln=188

Simple viral

infectionn=151

Bacterial co-infection

n=27Inconclusive

n=10 p-value*At presentation

Age (months) (median, IQR) 9(4-18) 9(5-19) 5(2-15) 12(4-25) 0.06

Gender, male (n,%) 112(60) 90(60) 15(56) 7(70) 0.83

Site of presentation <0.001

Secondary care centre (n,%) 144(77) 126(83) 8(30) 10(100)

Tertiary care centre (n,%) 16(8) 14(9) 2(7) 0(0)

PICU (n,%) 28(15) 11(7) 17(63) 0(0)

Duration of symptoms (days) (mean, SD) 3(1.7) 3(1.8) 3(1.4) 3(1.3) 0.33

Cough (n,%) 162(86) 134(89) 19(70) 9(90) 0.42

Ill-appearing (n,%) 67(36) 45(30) 18(67) 4(40) <0.001

Crepitation by auscultation (n,%) 99(53) 79(52) 13(48) 7(70) 0.35

Accessory muscle use / retractions (n,%) 64(34) 49(32) 14(52) 1(10) 0.06

Respiratory rate (mean, SD) 50(15) 50(15) 50(15) 44(12) 0.96

Saturation (%) (mean, SD) 96(5) 96(5) 94(8) 97(2) 0.12

Maximal temperature (°C) (mean, SD) 39.2(0.8) 39.1(0.8) 39.3(0.8) 39.3(0.7) 0.33

CRP (mg/L) (median, IQR) 18(7-43) 15(7-36) 50(8-151) 31(11-96) 0.002

Chest X-ray performed (n,%) 104(55) 75(50) 24(89) 5(50) <0.001

Infiltrate on chest radiography (n,%) 41(39) 25(33) 13(54) 3(60) <0.001

At discharge

Hospital admission (n,%) 121(64) 93(63) 24(89) 4(40) 0.001

Hospitalization duration (days) (median, IQR)

5(3-8) 4(3-6) 13(6-19) 4(3-8) <0.001

Need of mechanical ventilation (n,%) 23(12) 9(6) 14(52) 0(0) <0.001

Deaths (n,%) 1(0) 0(0) 1(4) 0(0) NA

CRP max (mg/L) (median, IQR) 24(10-64) 18(8-40) 152(105-212) 31(11-96) <0.001

Clinical syndrome (n,%) 0.001

LRTI 116(62) 88(58) 21(78) 7(70)

URTI 46(24) 41(27) 2(7) 3(30)

Unknown 18(10) 17(11) 1(4) 0(0)

Other 3(1) 3(2) 0(0) 0(0)

Urinary tract infection 2(1) 0(0) 2(7) 0(0)

Gastro-enteritis 2(1) 2(2) 0(0) 0(0)

Bacteremia 1(1) 0(0) 1(4) 0(0)

Clinical syndrome is based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. *p-value between bacterial and viral reference standard. CRP: C-reactive protein, IQR: interquartile range, PICU: paediatric intensive care units, LRTI: lower respiratory tract infection, NA: not applicable, URTI: upper respiratory tract infection.

15873_CvHouten_BNW_v2.indd 27 26-11-18 12:42

Page 30: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

28

Chapter 2

Table 2. Baseline characteristics RSV patients with viral reference standard.

Totaln=151

Antibiotic treatment

n=57

No antibiotic treatment

n=94 p-valueAge (months) (median, IQR) 9(5-19) 10(5-19) 9(4-18) 0.81

Gender, male (n,%) 90(60) 33(58) 57(61) 0.86

Maximal temperature (°C) (mean, SD) 39.1(0.8) 39.2(0.7) 39.1(0.8) 0.28

Duration of symptoms (days) (mean, SD) 3(1.8) 3(1.6) 3(1.9) 0.67

Ill-appearing (n,%) 45(30) 23(40) 22(23) 0.08

Hospital admission (n,%) 93(63) 45(79) 48(51) <0.001

Hospitalization duration (days) (median, IQR) 4(3-6) 5(3-7) 4(2-5) 0.004

CRP (mg/L) at admission (median, IQR) 15(7-36) 23(8-50) 13(6-26) 0.02

Chest X-ray performed (n,%) 75(50) 40(70) 35(37) <0.001

Infiltrate on chest radiography (n,%) 25(33) 20(50) 5(14) 0.001

Disease severity

Need of mechanical ventilation (n,%) 9(6) 7(12) 2(2) 0.03

Deaths (n,%) 0(0) 0(0) 0(0) NA

Recruiting site 0.009

Secondary care centre (n,%) 126(83) 41(72) 85(90)

Tertiary care centre (n,%) 14(9) 8(14) 6(7)

PICU (n,%) 11(7) 8(14) 3(3)

Clinical syndrome (n,%) <0.0001

LRTI 88(58) 46(81) 42(45)

URTI 41(27) 8(14) 33(35)

Unknown 17(11) 1(2) 16(17)

Other 3(2) 2(3) 1(1)

Gastro-enteritis 2(2) 0(0) 2(2)

Comparison of simple viral RSV infections with and without antibiotic treatment.Clinical syndrome is based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. CRP: C-reactive protein, IQR: interquartile range, PICU: paediatric intensive care units, LRTI: lower respiratory tract infection, NA: not applicable, URTI: upper respiratory tract infection.

Subgroup analysisOf the 188 RSV patients 28 (47%), children required PICU admission. Of these, 17 (61%) had a bacterial coinfection and were treated with antibiotics. One RSV patient with a bacterial coinfection died due to respiratory and circulatory insufficiency. Of the patients with a viral infection admitted to the PICU, 8 (29%) received antibiotics. In the overall cohort, the percentage of bacterial coinfection was higher in children under 3 months of age (28.6%) than in children older than 3 months (12.4%), P = 0.034. Bacterial coinfection and PICU admissions were seen more often in the Dutch cohort than the Israeli cohort (25% vs. 7%, respectively, P <0.001 and 34% vs. 1%, P <0.001).

15873_CvHouten_BNW_v2.indd 28 26-11-18 12:42

Page 31: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

29

2

Antibiotic overuse in children with RSV

The percentage of antibiotic overuse was similar in the Israeli and Dutch RSV cohort, 32% versus 27%, respectively (P = 0.52). When comparing the non-PICU cohort, the percentages of bacterial coinfections in the Dutch cohort decreases to 7.5% and 5.5% in the Israeli cohort (P = 0.74). The percentage of antibiotic overuse in this cohort is similar to the overall cohort.

D isc ussionIn this study, we prospectively evaluated the frequency of bacterial coinfections in patients diagnosed with RSV LRTI and antibiotic overuse by using an expert panel as the reference standard. We showed that bacterial coinfections are rare with 14%, but that about one-third of all children with RSV LRTI were treated unnecessarily with antibiotics.

In general, viral LRTI is a risk factor for bacterial coinfection, with increasing severity of respiratory illness.18,19 The percentage of bacterial coinfections of 14% in our study is higher compared with other studies, most of them report bacterial coinfections up to 10%.3–10 The high proportion of bacterial coinfection is probably related to recruitment at our PICU. Previous research also showed that infants with severe bronchiolitis requiring admission on the intensive care unit have higher rates of bacterial coinfection.11,12,20–22 When selecting only the non-PICU cohort, the percentage of bacterial coinfections decreased to the more common percentage of 6%.

The prescription rate of antibiotics in patients with RSV in our cohort was low when compared with other studies. Patient recruitment was performed in both Israel and The Netherlands. Country-specific antibiotic prescription rates of the countries in which previous studies are performed, are higher in comparison with Israel and The Netherlands.3,9,23 Especially in The Netherlands, antibiotic consumption per person is relatively low.24 The percentage of antibiotic use on the PICU is also lower when compared with previous reported data on other PICU cohorts, but in line with 2 studies performed on other Dutch PICUs.11,12,20–22

Antibiotic overuse was similar in The Netherlands and Israel. Dutch and Israeli guidelines for management of pneumonia in children are similar and advise to start amoxicillin in children with clinical evidence of pneumonia.

A recent study showed that physicians are more likely to use antibiotics in non-RSV–infected patients compared with RSV.25 In our study, the nasal swabs were performed for research and were analysed when patient recruitment was finished; therefore, the attending physician was not informed about the results of the nasal swab.

15873_CvHouten_BNW_v2.indd 29 26-11-18 12:42

Page 32: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

30

Chapter 2

Strengths of this study deserve further discussion. First, a strength of the present study is the thorough nature of the reference standard. To our knowledge, the OPPORTUNITY and TAILORED studies are the first studies using an expert panel as the preferred reference standard to distinguish bacterial from viral infection to investigate the amount of antibiotic overuse in children with RSV LRTI. Clinical suspicion confirmed by microbiological results is an approach often employed in other studies, but this method can be hampered by technical issues (e.g., culture contamination and uncertainty due to colonization) and microbiological investigations were not performed in all patients, particularly in children with lower disease severity. Therefore, reported numbers of bacterial pneumonia in infants with RSV infections are mostly from studies performed on the PICU.11,12,20–22 Using an expert panel has the advantage of capturing a wider spectrum of illness severities in the resulting cohort, and therefore, is more likely to be generalizable to clinical practice.26,27 Second, we study the number of correct and incorrect antibiotic use in children with RSV infections. Other studies report on the number of bacterial coinfections separately from the antibiotic treatment numbers or excluded patients positive for bacterial pathogens.9,20,25 Therefore, it is in these studies not possible to calculate the amount of suspected antibiotic overuse. Limitations of our study should also be discussed. First, children below the age of 1 month were not included. Children younger than 30 days are at high risk of occult serious bacterial infections.28,29 A second limitation of the present study is that due to logistical reasons not all eligible children participated in this study, which may have introduced bias. Third, 4 (5%) of the RSV patients with a simple viral infection received antibiotics more than 72 hours after presentation. The expert panel was instructed to diagnose the patients at the moment of presentation. Therefore, we cannot exclude that these 4 patients developed a bacterial coinfection later on. Even when considering these antibiotic treatment as correct, still 28% of the RSV patients is treated unnecessary with antibiotics. Fourth, the expert panel was not able to diagnose all patients, although we found a high level of agreement (95%) between experts within the panels. Employing adjudication of an expert panel diagnosis is an approach commonly used in this area of research.27,30 Additionally, due to the low antibiotic prescribing rates in Israel and The Netherlands, the reported amount of antibiotic overuse is not generalizable to all other countries.23 The results of our study cannot be extrapolated to other viruses, the analysis of non-RSV patients was beyond the scope of this study. Finally, because of the relatively small number of patients with a bacterial coinfection, it was not possible to perform an accurate multivariate regression to define criteria for starting antibiotics in children infected with RSV. However, as described previously in other studies as well, the number of bacterial coinfections is low.

15873_CvHouten_BNW_v2.indd 30 26-11-18 12:42

Page 33: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

31

2

Antibiotic overuse in children with RSV

In conclusion, our data show that although bacterial coinfections in children with RSV infections are relatively rare, high percentages of antibiotic prescriptions are common. Further research is needed to develop accurate and practical tools to help physicians recognizing bacterial coinfections in children infected with RSV. In the meantime, a judicious approach to antimicrobial treatment is warranted for 1 of the most common diseases during early childhood.

15873_CvHouten_BNW_v2.indd 31 26-11-18 12:42

Page 34: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

32

Chapter 2

References1. Hall CB, Weinberg GA, Iwane MK, et al. The burden of respiratory syncytial virus infection in

young children. N Engl J Med. 2009;360:588–598.

2. Farley R, Spurling GK, Eriksson L, et al. Antibiotics for bronchiolitis in children under two years of age. Cochrane Database Syst Rev. 2014;10:Cd005189.

3. Hall CB, Powell KR, Schnabel KC, et al. Risk of secondary bacterial infection in infants hospitalized with respiratory syncytial viral infection. J Pediatr. 1988;113:266–271.

4. Hishiki H, Ishiwada N, Fukasawa C, et al. Incidence of bacterial coinfection with respiratory syncytial virus bronchopulmonary infection in pediatric inpatients. J Infect Chemother. 2011;17:87–90.

5. Levine DA, Platt SL, Dayan PS, et al.; Multicenter RSV-SBI Study Group of the Pediatric Emergency Medicine Collaborative Research Committee of the American Academy of Pediatrics. Risk of serious bacterial infection in young febrile infants with respiratory syncytial virus infections. Pediatrics. 2004;113:1728–1734.

6. Titus MO, Wright SW. Prevalence of serious bacterial infections in febrile infants with respiratory syncytial virus infection. Pediatrics. 2003;112:282–284.

7. Purcell K, Fergie J. Concurrent serious bacterial infections in 2396 infants and children hospitalized with respiratory syncytial virus lower respiratory tract infections. Arch Pediatr Adolesc Med. 2002;156:322–324.

8. Bilavsky E, Shouval DS, Yarden-Bilavsky H, et al. A prospective study of the risk for serious bacterial infections in hospitalized febrile infants with or without bronchiolitis. Pediatr Infect Dis J. 2008;27:269–270.

9. Bloomfield P, Dalton D, Karleka A, et al. Bacteraemia and antibiotic use in respiratory syncytial virus infections. Arch Dis Child. 2004;89:363–367.

10. Cebey-López M, Pardo-Seco J, Gómez-Carballa A, et al.; GENDRES network. Bacteremia in children hospitalized with respiratory syncytial virus infection. PLoS One. 2016;11:e0146599.

11. Randolph AG, Reder L, Englund JA. Risk of bacterial infection in previously healthy respiratory syncytial virus-infected young children admitted to the intensive care unit. Pediatr Infect Dis J. 2004;23:990–994.

12. van Woensel JB, von Rosenstiel IA, Kimpen JL, et al. Antibiotic use in pediatric intensive care patients with lower respiratory tract infection due to respiratory syncytial virus. Intensive Care Med. 2001;27:1436.

13. CDC report: Antibiotic resistance threats in the United States 2013. Available from: http://www.cdc.gov/drugresistance/threat-report-2013/pdf/ar-threats-2013-508.pdf (accessed 3-6-2015).

14. Laxminarayan R, Duse A, Wattal C, et al. Antibiotic resistance-the need for global solutions. Lancet Infect Dis. 2013;13:1057–1098.

15. van Houten CB, de Groot JAH, Klein A, et al. A host-protein based assay to differentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study. Lancet Infect Dis. 2017;17:431–440.

16. Wacker C, Prkno A, Brunkhorst FM, et al. Procalcitonin as a diagnostic marker for sepsis: a systematic review and meta-analysis. Lancet Infect Dis. 2013;13:426–435.

17. Rutjes AW, Reitsma JB, Coomarasamy A, et al. Evaluation of diagnostic tests when there is no gold standard. A review of methods. Health Technol Assess. 2007;11:iii, ix–51.

15873_CvHouten_BNW_v2.indd 32 26-11-18 12:42

Page 35: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

33

2

Antibiotic overuse in children with RSV

18. Hament JM, Kimpen JL, Fleer A, et al. Respiratory viral infection predisposing for bacterial disease: a concise review. FEMS Immunol Med Microbiol. 1999;26:189–195.

19. Hendaus MA, Jomha FA, Alhammadi AH. Virus-induced secondary bacterial infection: a concise review. Ther Clin Risk Manag. 2015;11:1265–1271.

20. Thorburn K, Harigopal S, Reddy V, et al. High incidence of pulmonary bacterial co-infection in children with severe respiratory syncytial virus (RSV) bronchiolitis. Thorax. 2006;61:611–615.

21. Kneyber MC, Blussé van Oud-Alblas H, van Vliet M, et al. Concurrent bacterial infection and prolonged mechanical ventilation in infants with respiratory syncytial virus lower respiratory tract disease. Intensive Care Med. 2005;31:680–685.

22. Duttweiler L, Nadal D, Frey B. Pulmonary and systemic bacterial co-infections in severe RSV bronchiolitis. Arch Dis Child. 2004;89:1155–1157.

23. The center for disease dynamics: Antibiotic prescribing rates by country. Available from: http://www.cddep.org/tool/antibiotic_prescribing_rates_country#sthash.kiRPO6aH.dpbs. (accessed 5-2-2018).

24. van de Sande-Bruinsma N, Grundmann H, Verloo D, et al. Antimicrobial drug use and resistance in Europe. Emerg Infect Dis. 2008;14:1722–1730.

25. Goriacko P, Saiman L, Zachariah P. Antibiotic use in hospitalized children with respiratory viruses detected by multiplex polymerase chain reaction. Pediatr Infect Dis J. 2017.

26. Reitsma JB, Rutjes AW, Khan KS, et al. A review of solutions for diagnostic accuracy studies with an imperfect or missing reference standard. J Clin Epidemiol. 2009;62:797–806.

27. Bertens LC, Broekhuizen BD, Naaktgeboren CA, et al. Use of expert panels to define the reference standard in diagnostic research: a systematic review of published methods and reporting. PLoS Med. 2013;10:e1001531.

28. Bachur RG, Harper MB. Predictive model for serious bacterial infections among infants younger than 3 months of age. Pediatrics. 2001;108:311–316.

29. Milcent K, Faesch S, Gras-Le Guen C, et al. Use of procalcitonin assays to predict serious bacterial infection in young febrile infants. JAMA Pediatr. 2016;170:62–69.

30. Oved K, Cohen A, Boico O, et al. A novel host-proteome signature for distinguishing between acute bacterial and viral infections. PLoS One. 2015;10:e0120012

15873_CvHouten_BNW_v2.indd 33 26-11-18 12:42

Page 36: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

34

Chapter 2

S upplementar y materialSupplemental Table 1. Baseline characteristics total cohort.

Totaln=784

RSV+n=188

RSV-n=596 p-value

Age (months) (median, IQR) 15(6-30) 9(4-18) 17(8-35) <0.001

Gender, male (n,%) 438(56) 112(60) 326(55) 0.27

Maximal temperature (°C) (mean, SD) 39.3(0.8) 39.2(0.8) 39.4(0.8) 0.003

Duration of symptoms (days) (mean, SD) 2.6(1.8) 3(1.7) 2(1.8) <0.001

Ill-appearing (n,%) 297(38) 67(36) 230(39) 0.72

Hospital admission (n,%) 506(65) 121(64) 385(65) 1.00

Hospitalization duration (days) (median, IQR) 3(2-5) 5(3-8) 3(2-5) <0.001

CRP (mg/L) at admission (median, IQR) 21(7-54) 18(7-43) 23(7-57) 0.20

Chest X-ray performed (n,%) 278(36) 104(55) 174(29) 0.002

Infiltrate on chest radiography (n,%) 100(36) 41(39) 59(34) 0.37

Disease severity

Need of mechanical ventilation (n,%) 43(6) 23(12) 20(3) <0.001

Deaths (n,%) 1(0) 1(0) 0(0) NA

Use of antibiotics (n,%) 333(42) 92(49) 241(40) 0.052

Recruiting site <0.001

Secondary care centre (n,%) 615(78) 144(77) 471(79)

Tertiary care centre (n,%) 110(14) 16(8) 94(16)

PICU (n,%) 59(8) 28(15) 31(5)

Clinical syndrome (n,%) <0.001

LRTI 224(28) 116(62) 108(18)

URTI 218(28) 46(24) 172(29)

Unknown 194(25) 18(10) 176(30)

Other 60(8) 3(1) 57(10)

UTI 32(4) 2(1) 30(5)

GE 23(3) 2(1) 21(4)

Bacteremia 4(1) 1(1) 3(1)

Viremia 9 (1) 0(0) 9(2)

CNS 20(2) 0(0) 20(3)

Clinical syndrome is based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis; CNS included cerebellitis, encephalitis, meningitis and epilepsy. CRP: C-reactive protein, PICU: paediatric intensive care units, CNS: central nervous system, GE: gastro-enteritis, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection, UTI: urinary tract infection.

15873_CvHouten_BNW_v2.indd 34 26-11-18 12:42

Page 37: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

35

2

Antibiotic overuse in children with RSV

Supplemental Table 2. Baseline characteristics of RSV patients per country.

a. Total

n=188NL

n=80IL

n=108 p-value

Age (months) (median, IQR) 9(4-18) 6(2-12) 12(6-24) <0.001

Gender, male (n,%) 112(60) 44(55) 68(63) 0.29

Maximal temperature (°C) (mean, SD) 39.2(0.8) 39.1(0.8) 39.2(0.7) 0.21

Duration of symptoms (days) (mean, SD) 3(1.7) 3(1.8) 3(1.6) 0.17

Ill-appearing (n,%) 67(36) 45(56) 22(20) <0.001

Hospital admission (n,%) 121(64) 71(89) 51(47) <0.001

Hospitalization duration (days) (median, IQR) 5(3-8) 6(3-13) 4(3-5) <0.001

CRP (mg/L) at admission (median, IQR) 18(7-43) 24(9-58) 15(7-37) 0.06

Chest X-ray performed (n,%) 104(55) 40(50) 64(59) 0.30

Infiltrate on chest radiography (n,%) 41(39) 16(40) 25(39) 1.00

Disease severity

Need of mechanical ventilation (n,%) 23(12) 23(29) 0(0) <0.001

Deaths (n,%) 1(0) 1(1) 0(0) NA

Recruiting site <0.001

Secondary care centre (n,%) 144(77) 37(46) 107(99)

Tertiary care centre (n,%) 16(8) 16(20) 0(0)

PICU (n,%) 28(15) 27(34) 1(1)

Clinical syndrome (n,%) 0.03

LRTI 116(62) 49(61) 67(62)

URTI 46(24) 27(34) 19(18)

Unknown 18(10) 3(4) 15(14)

Other 3(1) 0(0) 3(3)

UTI 2(1) 1(12) 1(1)

GE 2(1) 0(0) 2(2)

Bacteremia 1(1) 0(0) 1(1)

15873_CvHouten_BNW_v2.indd 35 26-11-18 12:42

Page 38: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

36

Chapter 2

Supplemental Table 2. Baseline characteristics of RSV patients per country. (Continued)

b. Total

n=144NL

n=37IL

n=107 p-valueAge (months) (median, IQR) 12(6-22) 8(5-19) 12(6-24) 0.10

Gender, male (n,%) 86(60) 18(49) 68(63) 0.12

Maximal temperature (°C) (mean, SD) 39.2(0.8) 39.1(0.7) 39.2(0.7) 0.64

Duration of symptoms (days) (mean, SD) 3(1.8) 3(1.8) 3(1.6) 0.81

Ill-appearing (n,%) 37(26) 16(43) 22(20) 0.011

Hospital admission (n,%) 78(54) 28(76) 51(47) 0.002

Hospitalization duration (days) (median, IQR) 4(3-5) 3(2-5) 4(3-5) 0.45

CRP (mg/L) at admission (median, IQR) 16(7-38) 23(10-57) 15(7-37) 0.11

Chest X-ray performed (n,%) 73(51) 10(27) 64(59) 0.002

Infiltrate on chest radiography (n,%) 31(42) 7(70) 25(39) 0.09

Clinical syndrome (n,%) 0.007

LRTI 83(58) 16(43) 67(62)

URTI 37(26) 18(49) 19(18)

Unknown 17(12) 2(5) 15(14)

Other 3(2) 0(0) 3(3)

UTI 2(1) 1(3) 1(1)

GE 2(1) 0(0) 2(2)

a. Comparison of all enrolees between the Dutch and Israeli cohort. b. Comparison of participants recruited in secondary hospitals between the Dutch and Israeli cohort.Clinical syndrome is based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. CRP: C-reactive protein, PICU: paediatric intensive care units, GE: gastro-enteritis, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection, UTI: urinary tract infection.

15873_CvHouten_BNW_v2.indd 36 26-11-18 12:42

Page 39: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

37

2

Antibiotic overuse in children with RSV

Supplemental Table 3. Microbiology results RSV patients with simple viral reference standard.

Totaln=151

Antibiotic treatment

n=57

No antibiotic treatment

n=94

Study nasal swab PCR

1 virus 76(50) 27(47) 50(53)

2 virus 46(31) 16(28) 30(32)

3 virus 27(18) 13(23) 14(15)

4 virus 1(1) 1(2) 0(0)

Viruses

Adenovirus 15(10) 5(9) 10(11)

Bocavirus 16(11) 7(12) 9(10)

Coronavirus 229E/NL63/OC43 14(9) 7(12) 7(7)

Enterovirus* 8(5) 4(7) 4(4)

Influenza A/B 7(5) 5(9) 2(2)

Metapneumovirus* 6(4) 3(5) 3(3)

Parainfluenza 3(2) 1(2) 2(2)

Respiratory syncytial virus A/B 151(100) 57(100) 94(100)

Rhinovirus A/B/C 31(21) 13(23) 18(19)

Routine care

Positive blood tests

Respiratory syncytial virus 25(17) 12(22) 13(16)

Positive nasopharyngeal tests

Metapneumovirus 1(1) 1(2) 0(0)

Respiratory syncytial virus 9(6) 4(7) 3(3)

Adenovirus 2(1) 1(2) 1(1)

Positive sputum culture

Haemophilus influenzae 1(1) 1(2) 0(0)

Group A streptococcus 1(1) 1(2) 0(0)

Respiratory syncytial virus 5(3) 4(7) 1(1)

Moraxella Catarrhalis 2(1) 0(0) 2(2)

Positive urine culture

E.coli 1(1) 1(2)** 0(0)

Klebsiella oxytoca 1(1) 1(2)** 0(0)

Positive eye culture

Haemophilus influenzae 4(3) 3(5) 1(1)

Blood tests including: culture, PCR, serology, antigen test.*Only tested in patients participating in the OPPORTUNITY study. ** Same patient, CFU 10^4.

15873_CvHouten_BNW_v2.indd 37 26-11-18 12:42

Page 40: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

38

Chapter 2

Supplemental Table 4. Expert panel agreement.

Antibiotics + Antibiotics -

Simple viral reference standard

3 out of 3 (n,%) 40(21) 89(47)

2 out of 3 (n,%) 17(9) 5(3)

Bacterial co-infection reference standard

3 out of 3 (n,%) 16(9) NA

2 out of 3 (n,%) 11(6) NA

Inconclusive reference standard (n,%) 8(4) 2(1)

Given numbers are number of patients, in parentheses percentages of the total cohort.

Supplemental Table 5. Prescribed antibiotics.

Overalln=92*

The Netherlands

n=43*Israeln=49*

Viral infection

n=57*

Bacterial infection

n=27*Inconclusive

n=10Amoxicillin 40 9 31 29 5 6Amoxicillin/clavulanate 40 32 8 16 22 2Cefuroxime 10 0 10 8 1 1Azithromycin 6 2 4 5 1 0Ceftriaxone 4 3 1 2 2 0Gentamicin 3 2 1 0 2 1Meropenem 3 2 1 0 3 0Cefazolin 2 1 1 1 1 0Cefotaxime 1 1 0 1 0 0Ciprofloxacin 1 1 0 0 1 0Clarithromycin 1 1 0 1 0 0Unknown 4 0 4 4 0 0

* Some patients received 2 or 3 different antibiotics, therefore the total number is higher compared to the number of patients.

15873_CvHouten_BNW_v2.indd 38 26-11-18 12:42

Page 41: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 39 26-11-18 12:42

Page 42: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

3

15873_CvHouten_BNW_v2.indd 40 26-11-18 12:42

Page 43: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

3Antibiotic misuse in respiratory tract infections in children and adults:a prospective, multicentre study (TAILORED Treatment)

CB van Houten, A Cohen, D Engelhard, JP Hays, R Karlsson, ERB Moore, D Fernández, R Kreisberg, LV Collins, W de Waal, KM de Winter-de Groot, TFW Wolfs, P Meijers, B Luijk, JJ Oosterheert, R Heijligenberg, SUC Sankatsing, AWJ Bossink, AP Stubbs, M Stein, S Reisfeld, A Klein, R Rachmilevitch, J Ashkarq, I Braverman, V Kartun, I Chistyakov, E Bamberger, I Srugo, M Odeh, E Schiff, Y Dotan, O Boico, R Navon, T Friedman, L Etshtein, M Paz, TM Gottlieb, E Pri-Or, G Kronenfeld, E Simon, K Oved, E Eden and LJ Bont

Submitted

15873_CvHouten_BNW_v2.indd 41 26-11-18 12:42

Page 44: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

42

Chapter 3

Abstrac tBackground Respiratory tract infections (RTI) are more commonly caused by viral pathogens in children than in adults. Surprisingly little is known about antibiotic use in children as compared to adults with RTI. This prospective, international study aimed to determine antibiotic consumption in children and adults with RTI in order to prioritize the target age population for antibiotic stewardship interventions.

MethodsWe recruited children, aged 1 month and older, and adults who presented at the emergency department or were hospitalized with clinical presentation of RTI. A panel of three experienced physicians adjudicated a reference standard diagnosis (i.e., bacterial or viral infection) for all the patients using all available clinical and laboratory information, including a 28-day follow-up assessment.

ResultsThe cohort included 284 children and 232 adults with RTI (median age, 1.3 years and 64.5 years, respectively). The proportion of viral infections was larger in children than in adults (209 (74%) versus 89 (38%), p<0.001). In case of viral RTI, antibiotics were prescribed (i.e., overuse) less frequently in children than in adults (77/209 (37%) versus 74/89 (83%), p<0.001). Only 1 (1%) child and 3 (2%) adults with bacterial infection were not treated with antibiotics (i.e. underuse); all were mild cases.

Conclusion This international, prospective study confirms major antibiotic overuse in patients with RTI. Viral infection is more common in children, but antibiotic overuse is more frequent in adults with viral RTI. Together, these findings support the need for effective interventions to decrease antibiotic overuse in RTI patients of all ages.

15873_CvHouten_BNW_v2.indd 42 26-11-18 12:42

Page 45: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

43

3

Antibiotic misuse in children and adults

Introduc tionAcute respiratory tract infections (RTIs) are one of the leading causes of emergency department (ED) visits and are often due to viral pathogens.1-4 Although viral infections are more common in children, studies based on national datasets show that the problem of antibiotic overuse in RTI is largest in adults.4-6 Unfortunately, it is often not possible to differentiate between viral and bacterial diseases on clinical judgment alone.7 Consequently, antibiotics are prescribed almost twice as often as required in children with acute RTIs in the US.1 Falsey and colleagues show that 90% of adults hospitalized with a viral respiratory illness were treated with antibiotics.8 Antibiotic overuse is associated with an increasing prevalence of antibiotic resistance.9 In Europe, 25,000 patients die annually due to infections with antibiotic-resistant microorganisms, with estimated costs of €1.5 billion.10-12 Therefore, there are increasing efforts to study host-biomarkers that could discriminate bacterial from non-bacterial infections.13 A prospective, international study (The “Tailored-Treatment” (TTT) study, www.tailored-treatment.eu) was designed to generate a multi-parametric model for distinguishing between bacterial and viral infections based on new host- or pathogen-related biomarkers. The current study is the first subgroup analysis of this TTT-study and aimed to determine antibiotic misuse in children and adults with RTI, using an expert panel to assign viral and bacterial diagnoses. This study will be instrumental to analyse strategies for new diagnostics to differentiate between viral and bacterial infections.

MethodsStudy design Patient recruitment for this prospective biomarker TTT-study took place in convenience and consecutive series at the ED and wards of secondary and tertiary hospitals in The Netherlands and Israel. For this subgroup analyses paediatric patients (aged ≥1 month), and adult patients, (aged >18 years), with a suspected upper and/or lower RTI and a maximal disease duration of eight days, were selected. RTI was defined as presence of two or more of the following signs: tachypnea, cough, nasal flaring, chest retractions, rales, expiratory wheeze and/or decreased breath sounds. For children WHO age-specific criteria for tachypnea were used.14 Patients were excluded in case of: previous episode of fever in the past three weeks; nosocomial RTI (>3days after hospitalization); psychomotor retardation; moderate to severe metabolic disorder; primary or secondary immunodeficiency; proven or suspected HIV, HBV or HCV infection and active malignancies. To participate in the study (parenteral) informed consent was required. The TTT-study is registered on ClinicalTrials.gov, NCT02025699, and was approved by the ethics committees in the participating countries.

15873_CvHouten_BNW_v2.indd 43 26-11-18 12:42

Page 46: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

44

Chapter 3

Data collectionWithin 24 hours after presentation a nasal swab (Universal Transport Medium, Copan) was collected for the establishment of patient reference standard diagnosis. As shown in the clinical protocol, other samples were also collected for additional research questions of the TTT-study. All nasal swabs were stored at -80°C until transfer to the UMCU laboratory (The Netherlands). A multiplex PCR-based assay of the 14 most common respiratory pathogens (nine viruses, five bacteria) was performed (MagnaPure LC total nucleic acid kit and MagnaPure 96 DNA, Roche Diagnostics, Mannheim, Germany).15 The PCR results were not available for the attending physician, since this assay was performed after completion of the recruitment process. Clinical data sourced from medical records was recorded in an electronic Case Report Form (eCRF). The eCRF included details of demographics, medical history, physical examination, radiological and laboratory data performed for routine care, microbiological results (including results from routine care and study-specific multiplex PCR on nasal swab), if antimicrobial and/or antiviral treatment was started and information from a 28-day follow-up assessment. All available data were uploaded to a study-specific central, secured database, to which access was granted only to TTT-research partners.

OutcomesCurrently, no single reference standard test exists for determining the aetiology of an infection.16 Therefore, we followed the UK’s National Health Service standard for evaluating diagnostic tests and employed an expert panel reference standard.17 We established expert panels with paediatricians (i.e. general paediatricians and sub-specialists) for the paediatric cohort and specialists in internal medicine, pulmonology and infectious diseases for the adult cohort. All physicians had more than 10 years of clinical experience. They received a review of paediatric, emergency medicine, and infectious disease literature, written by the study team as background information, without any instructions or decision rules. Every recruited patient was diagnosed by three panel members affiliated to the country of recruitment, using all available eCRF information, but blinded to the diagnoses of their peers. Each expert assigned one of the following classifications to each patient: simple viral infection; bacterial infection; mixed infection (i.e., viral and bacterial co-infection); non-infectious disease; or indeterminate. A majority consensus was applied for the final diagnosis. Patients assigned as ‘mixed infection’ were subsequently classified as bacterial because they are clinically managed similarly. Cases were labelled as ‘inconclusive’ if each panel member assigned a different aetiology or when at least two panel members diagnosed the case as ‘indeterminate’. A microbiologically confirmed diagnosis was predefined as an unanimous panel diagnosis plus the detection of at least one virus for simple viral cases or for bacterial cases a positive blood culture, excluding the following probable contaminants: Coagulase-

15873_CvHouten_BNW_v2.indd 44 26-11-18 12:42

Page 47: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

45

3

Antibiotic misuse in children and adults

negative staphylococci; Corynebacterium spp.; Bacillus spp.; Propionibacterium acnes; Micrococcus spp. and Viridans group streptococci. For the detection of viruses and bacteria microbiological diagnostics performed for routine care (e.g. blood cultures, sputum cultures and serology) and study specific nasal swab PCR results were reviewed.

Statistical analysis Patients from this convenience cohort of the TTT biomarker study were first stratified according to the reference diagnosis (e.g. viral, bacterial, non-infectious and inconclusive). For the purpose of this study, we excluded non-infectious and inconclusive cases. For the primary objective of this study we then calculated and compared the percentage of antibiotic use per reference diagnosis for children and adults separately. A sensitivity analysis was performed on the microbiologically confirmed sub-cohort. Secondary analyses were performed for children and adults separately to compare patient characteristics between simple viral and bacterial infections, antibiotic use per virus, patient characteristics of viral cases receiving and not-receiving antibiotics, and different antibiotic agents per country. Sub-cohort analyses were performed for the Dutch and Israeli cohorts and for patients admitted to the intensive care unit (ICU). A post hoc analysis was performed on the timing of antibiotic administration in patients with bacterial outcomes to see whether there is delayed antibiotic prescribing (i.e., antibiotics started >72 hours after administration). For baseline characteristics, univariate comparisons were performed using the Fisher’s exact test, the Students t-test, and Mann-Whitney test, as appropriate. Statistical analysis was performed using the SPSS version 22.0 for Windows software. A p-value <0.05 was considered statistically significant.

ResultsPatient characteristicsBetween April 2014 and September 2016 a total of 616 patients with RTI (302 children and 314 adults) were recruited (Figure 1). The panel diagnosed 516 patients as having a bacterial or a viral infection, encompassing 284 children and 232 adults (median ages, 1.3 years and 64.5 years, respectively) (Table 1). Mixed infection was considered as bacterial. The expert panel diagnosed 12 adults as having a non-infectious disease (predominantly, chronic obstructive pulmonary disease or asthma exacerbation). The reference standard diagnosis was inconclusive for 18 children and 70 adults. In 215 (76%) children and 120 (52%) adults, the study nasal swab (to help the expert panel establishing the outcome) was positive for one or more microorganisms (Supplemental Table 1). In most of the patients with a bacterial reference standard, a bacterial pathogen was not found (Supplemental Table 1).

15873_CvHouten_BNW_v2.indd 45 26-11-18 12:42

Page 48: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

46

Chapter 3

Table 1. Baseline of bacterial and viral respiratory tract infections in children and adults.

Children n=284

Adults n=232

Age, years 1.3[0.6-3.0] 64.5[52-75]

Male sex 167(59) 131(57)

Presence of comorbidity 125(44) 199(86)

Ill-appearing 113(40) 114(53)

Maximum temperature, °C 39.2 (0,9) 38.6(1,0)

Duration of symptoms, days 3(2) 4(2)

Hospital admission 208(75) 217(94)

Hospitalization duration, days 4[3-8] 5[3-8]

CRP (mg/L) at admission 16[4-43] 34[9-136)

Disease severity

Oxygen saturation, % 95[92-98] 94[91-96]

Needed mechanical ventilation 31(11) 3(1)

Deaths 1(1) 3(1)

Admission site

Secondary care centre 198(70) 173(75)

Tertiary care centre 47(16) 53(23)

ICU 39(14) 6(2)

Country

The Netherlands 136(48) 131(56)

Israel 148(52) 101(44)

Clinical syndrome

COPD/Asthma exacerbation 4(1) 45(19)

LRTI 150(53) 172(74)

URTI 130(46) 15(7)

Data are presented as n(%), mean (SD), or median [IQR]. LRTI included: pneumonia, acute bronchitis and bronchiolitis, URTI included: laryngitis, pharyngitis, otitis media, sinusitis, epiglottitis and tonsillitis. Ill-appearing based on attending physician’s impression. CRP: C-reactive protein, ICU: intensive care unit, COPD: Chronic Obstructive Pulmonary Disease, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection.

Patient outcomesThe proportion of simple viral infections was larger in children than in adults (209/284 (74%) versus 89/232 (38%), respectively, p<0.001). Most bacterial co-infections were observed in children infected with rhinovirus (17/62, 27%) and respiratory syncytial virus (RSV) (23/98, 23%), whereas influenza was most frequently associated with bacterial co-infection in adults (17/52, 33%, Table 2). Children and adults with bacterial

15873_CvHouten_BNW_v2.indd 46 26-11-18 12:42

Page 49: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

47

3

Antibiotic misuse in children and adults

infections were more often hospitalized (p-values respectively <0.0001 and 0.009) and had higher CRP values (p-value 0.001 and <0.0001 respectively) compared with patients with a simple viral infection (Table 3). In 172/284 (61%) of the paediatric cohort and 114/232 (49%) of the adult patients the expert panel diagnosis can be confirmed microbiologically. This microbiologically confirmed sub-cohort includes in total 286 patients, 145 children and 58 adults with simple viral infection and, 27 children and 56 adults with bacterial infection (Supplemental Figure 1).

Table 2. Appropriate and inappropriate antibiotic usage per virus.

a.

PaediatricViral

n=209Bacterial

n=75

Viruses detected* Antibiotic use*** Viruses detected Antibiotic use

Adenovirus 28 12(43) 2 2(100)

Bocavirus 22 7(32) 5 5(100)

Influenza virus 30 10(33) 6 6(100)

Rhinovirus 45 16(36) 17 16(94)

RSV 75 32(43) 23 22(96)

Other** 26 11(42) 8 8(100)

b.

AdultViral n=89

Bacterial n=143

Viruses detected Antibiotic use*** Viruses detected Antibiotic use

Influenza virus 35 30(86) 17 16(94)

Rhinovirus 16 12(75) 6 6(100)

RSV 14 13(93) 4 4(100)

Other**** 11 10(91) 8 8(100)

a. Paediatric cohort. b. Adult cohort. Viral and bacterial diagnoses based on expert panel diagnoses. Mixed infection was considered as bacterial. Data shown represent the numbers of positive PCR of nasal swabs performed for the study and n(%) of patients in this group receiving antibiotics. RSV: respiratory syncytial virus. *As some patients tested positive for more than one virus, the total number of detected viruses is higher than the number of patients. ** Includes coronavirus, human metapneumovirus, and parainfluenza virus. *** Numbers of antibiotic usages are given per virus. As some patients tested positive for more than one virus, the total antibiotic usage is different with respect to the numbers given in Figure 1. **** Includes adenovirus, bocavirus, coronavirus, human metapneumovirus, and parainfluenza virus.

Antibiotic usageThe overall antibiotic prescription rate for viral and bacterial RTI was 71%, and the antibiotic overuse rate (i.e., antibiotic prescription for simple viral RTI) was 51%. Antibiotics were administered less frequently to children than adults with a simple viral

15873_CvHouten_BNW_v2.indd 47 26-11-18 12:42

Page 50: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

48

Chapter 3

infection (77/209 (37%) versus 74/89 (83%), p<0.001, (Figure 1). This difference was similar across different viral pathogens, including influenza and RSV (Table 2). Within the microbiologically confirmed sub-cohort, similar percentages of antibiotic overuse were observed (50/145 (34%) children versus 50/58 (86%) adults, (Supplemental Figure 1). Children receiving antibiotics for simple viral RTI were more often admitted to the ICU (p-value 0.032) and had more often lower RTI (p-value 0.001), compared with children not receiving antibiotics (Table 4). Adults with simple viral RTI receiving antibiotics were more often male (p-value 0.033), had higher temperatures (p-value 0.004) and also had more often lower RTI (p-value 0.003), compared with adults not receiving antibiotics (Table 4). Among the patients with bacterial RTI (n=218), only one (1%) child and three (2%) adults were not treated with antibiotics (Supplemental Table 2). Dutch children received mostly amoxicillin/clavulanate, whereas Israeli children received mostly amoxicillin. Among adults the most prescribed antibiotic agents were amoxicillin (The Netherlands) and roxithromycin (Israel, Supplemental Figure 2). From patients with bacterial outcome information on antibiotic timing was available for 107 patients (49%). In eight children (7%) antibiotics were prescribed >72 hours after admission, seven of these children were admitted on the ICU. All adults received antibiotics within 72 hours after presentation.

616 patients

recruited

307 patients

with RTI

recruited in

the Netherlands

309 patients

with RTI

recruited in

Israel

218 diagnosed as

bacterial by

expert panel

(reference standard)

298 diagnosed as

viral by

expert panel

(reference standard)

88 diagnosed as

inconclusive by

expert panel

(reference standard)

12 diagnosed as

non-infectious by

expert panel

(reference standard)

209

child

89

adult

AB +

n=77

AB -

n=132

75

child

143

adult

AB +

n=74

AB -

n=15

AB +

n=74

AB -

n=1

AB +

n=140

AB -

n=3

0

child

12

adult

AB +

n=8

AB -

n=4

18

child

70

adult

AB +

n=17

AB -

n=1

AB +

n=69

AB -

n=1

Figure 1. Flowchart of patients.AB -: antibiotics not prescribed, AB +: antibiotics prescribed, RTI: respiratory tract infection.

15873_CvHouten_BNW_v2.indd 48 26-11-18 12:42

Page 51: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

49

3

Antibiotic misuse in children and adults

Subgroup analysisWe analysed the Dutch (n=267) and Israeli (n=249) cohort separately (Supplemental Table 3). Antibiotic overuse in children with simple viral infections was similar in the Dutch and Israeli cohort (32/104 (31%) versus 45/105 (43%), p=0.07). In adults with simple viral infection, the proportion of patients receiving antibiotics was lower in The Netherlands, when compared with Israel (35/47 (74%) versus 39/42 (93%), p=0.021). Of all 284 children, 39 (14%) children were admitted to the ICU. Thirty-three (85%) ICU patients received antibiotics, 11 (33%) had simple viral infection. Six (2%) adults were admitted to the ICU, all received antibiotics. Five adult ICU patients had simple viral infections, and one patient had a bacterial infection. Influenza virus was detected in four of them. Table 3. Comparison of patients with viral and bacterial reference standards.a

Paediatric cohortViral

n=209Bacterial

n=75 p-valueAge, years 1.2[0.6-2.8] 1.3[0.5-5.8] 0.102

Male sex 119(57) 48(64) 0.122

Presence of comorbidity 86(41) 39(52) 0.104

Ill-appearing 75(36) 38(51) 0.059

Maximum temperature, °C 39.1(0,9) 39.3(0,9) 0.150

Duration of symptoms, days 3(2) 3(2) 0.497

Hospital admission 144(70) 64(91) <0.0001

Hospitalization duration, days 4[3-6] 4[2-16] 0.050

CRP (mg/L) at admission 13[4-38] 22[6-131] 0.001

Oxygen saturation, % 96[92-98] 95[91-98] 0.523

Need mechanical ventilation 12(6) 19(25) <0.0001

Admission site <0.0001

Secondary care centre 152(73) 46(61)

Tertiary care centre 40(19) 7(9)

ICU 17(8) 22(29)

Country 0.291

The Netherlands 104(50) 32(43)

Israel 105(50) 43(57)

Clinical syndrome <0.0001

Asthma exacerbation 4(2) 0(0)

LRTI 110(53) 55(73)

URTI 95(45) 20(27)

15873_CvHouten_BNW_v2.indd 49 26-11-18 12:42

Page 52: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

50

Chapter 3

Table 3. Comparison of patients with viral and bacterial reference standards. (Continued)

b.

Adult cohortViraln=89

Bacterialn=143 p-value

Age, years 61[46-72] 67[53-75] 0.061

Male sex 46(52) 85(59) 0.247

Presence of comorbidity 79(89) 120(84) 0.304

Ill-appearing 38(43) 76(59) 0.023

Maximum temperature, °C 38,3(0,9) 38,7(1,0) 0.015

Duration of symptoms, days 4(2) 4(3) 0.495

Hospital admission 79(89) 138(97) 0.009

Hospitalization duration, days 4[3-6] 6[3-9] 0.010

CRP (mg/L) at admission 14[4-43] 67[16-193] <0.0001

Oxygen saturation, % 95[91-96] 94[92-97] 0.779

Needed mechanical ventilation 2(2) 1(1) 0.310

Admission site 0.007

Secondary care centre 71(80) 102(71)

Tertiary care centre 13(15) 40(28)

ICU 5(5) 1(1)

Country 0.376

The Netherlands 47(53) 84(59)

Israel 42(47) 59(41)

Clinical syndrome 0.001

COPD/Asthma exacerbation 23(26) 22(15)

LRTI 55(62) 117(82)

URTI 11(12) 4(3)

a. Paediatric cohort. b. Adult cohort. Viral and bacterial diagnoses based on expert panel diagnoses. Mixed infection was considered as bacterial. Data are presented as n(%), mean (SD), or median [IQR]. CRP: C-reactive protein, ICU: intensive care unit, COPD: Chronic Obstructive Pulmonary Disease, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection.

Most studies of antibiotic misuse rates are based on national datasets and classify infections, using general codes, such as the International Classification of Diseases.4-6 Using guidelines for assessing antibiotic misuse can result in contradictory analyses. For example, Donnelly et al.4 have classified pharyngitis and tonsillitis as diseases for which antibiotic treatment is appropriate, whereas Barlam et al.5 have proposed that antibiotic use for these illnesses is inappropriate. Using an expert panel as reference standard has the advantage of individual outcomes (i.e. bacterial or viral infection) for every patients, resulting in more accurate percentages of antibiotic misuse. In the present prospective study, we confirmed that antibiotic overuse in viral RTI is more prevalent among

15873_CvHouten_BNW_v2.indd 50 26-11-18 12:42

Page 53: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

51

3

Antibiotic misuse in children and adults

adult patients. In our study, children less often had comorbidity, appeared to be less ill at presentation, and had lower a priori probabilities of having a bacterial infection, compared with adults. Physicians are more inclined to initiate antibiotic treatment if the patient appears to be ill upon presentation, even if a viral pathogen was detected, and do often not adhere to related recommendations.19,20 Therefore, in addition to effective diagnostics tests, education and prescribing feedback are needed to reduce antibiotic overuse.21,22

Table 4. Baseline of viral respiratory tract infections children and adults, antibiotics versus no antibiotics.

a. AB + n=77

AB -n=132 p-value

Age, years 1.0[0.5-2.7] 1.2[0.6-2.8] 0.945

Male sex 42(55) 77(58) 0.594

Presence of comorbidity 24(31) 62(47) 0.025

Ill-appearing 30(39) 45(35) 0.473

Maximum temperature, °C 39,2(0,9) 39,1(0,8) 0.479

Duration of symptoms, days 3(2) 3(2) 0.352

Hospital admission 60(79) 84(65) 0.030

Hospitalization duration, days 5[3-9] 3[2-4] <0.001

CRP (mg/L) at admission 14[3-32] 10[3-26] 0.294

Disease severity

Oxygen saturation, % 95[88-97] 97[93-99] 0.051

Needed mechanical ventilation 9(12) 3(2) 0.005

Death 0(0) 0(0) NA

Admission site 0.032

Secondary care centre 50(65) 102(77)

Tertiary care centre 16(21) 24(18)

ICU 11(14) 6(5)

Country 0.070

The Netherlands 32(42) 72(55)

Israel 45(58) 60(45)

Clinical syndrome 0.001

COPD/Asthma exacerbation 0(0) 4(3)

LRTI 48(62) 47(36)

URTI 29(38) 81(61)

15873_CvHouten_BNW_v2.indd 51 26-11-18 12:42

Page 54: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

52

Chapter 3

Table 4. Baseline of viral respiratory tract infections children and adults, antibiotics versus no antibiotics. (Continued)

b.AB +

n=74AB -

n=15 p-valueAge, years 64[47-75] 56[51-60] 0.086

Male sex 42(57) 4(27) 0.033

Presence of comorbidity 66(89) 13(87) 0.778

Ill-appearing 34(47) 4(27) 0.156

Maximum temperature, °C 38,5(0,9) 37,8(0,6) 0.004

Duration of symptoms, days 4(2) 3(2) 0.478

Hospital admission 66(89) 13(87) 0.778

Hospitalization duration, days 4[3-6] 4[2-7] 0.805

CRP (mg/L) at admission 15[5-45] 7[3-35] 0.332

Disease severity

Oxygen saturation, % 95[91-96] 95[91-98] 0.317

Needed mechanical ventilation 2(3) 0(0) 0.520

Death 1(1) 0(0) 0.651

Admission site 0.234

Secondary care centre 60(81) 11(73)

Tertiary care centre 9(12) 4(27)

ICU 5(7) 0(0)

Country 0.021

The Netherlands 35(47) 12(80)

Israel 39(53) 3(20)

Clinical syndrome 0.003

COPD/Asthma exacerbation 14(19) 9(60)

LRTI 51(69) 4(27)

URTI 9(12) 2(13)

a. Paediatric cohort. b. Adult cohort. Data are presented as n(%), mean (SD), or median [IQR]. LRTI included pneumonia, acute bronchitis and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. AB +: antibiotics prescribed, AB -: antibiotics not prescribed, CRP: C-reactive protein, ICU: intensive care unit, COPD: Chronic Obstructive Pulmonary Disease, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection.

15873_CvHouten_BNW_v2.indd 52 26-11-18 12:42

Page 55: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

53

3

Antibiotic misuse in children and adults

D isc ussionThis convenience cohort of patients from the TTT biomarker study is the first prospective study comparing the burden of antibiotic misuse in both children and adults diagnosed with RTI, using an expert panel adjudication as the reference standard.18 We observed that antibiotic overuse was less common in children than in adults with a simple viral RTI (37% versus 83%), regardless of viral aetiology. Only 1 (1%) child and 3 (2%) adults with bacterial infection were not treated with antibiotics (i.e., underuse), all “untreated patients” were mild cases with full spontaneous recovery. The percentages of antibiotic underuse in our study were low. In literature, underuse up to 31% for children with pneumonia has been described.23, 24 Therefore, we performed a post hoc analysis on a selection of patients for whom information about the timing of antibiotic administration was available. We found delayed antibiotic prescribing (i.e., antibiotics started >72 hours after admission) in only seven children who were admitted to the ICU and one non-ICU child; there were no delayed antibiotic prescriptions in adults. The expert panel may have underestimated bacterial infections in patients recovering without antibiotics. We did not observe differences in antibiotic overuse for either children or adults with respect to viral aetiology. In contrast, a recent single-centre, retrospective cohort study of 1400 children found that physicians were more likely to use antibiotics for non-RSV-infected children.25 It is possible that different methodologies, as well as a lack of power in our study, may explain the contradictory findings. The current study, aiming to study differences in antibiotic misuse between children and adults, is a sub-analysis of a biomarker study. Therefore, no sample size calculation for this objective was made. The number of recruited patients are sufficient for the primary objective, but we lack power for statistical analyses regarding antibiotic overuse per virus.Existing literature shows that antibiotic use is higher in Israel, compared with the Netherlands.26 Nevertheless, we did not observe significant differences in antibiotic overuse between Dutch and Israeli children. A relatively high rate of antibiotic overuse in the Netherlands may be related to the high proportion of severely ill children in the Dutch cohort.A strength of our study is that the cohort comprised both children and adults, enabling a direct comparison of findings without any confounding issues related to the methodology. A second strength is the thorough nature of our reference standard to distinguish viral from bacterial infections.16,27 Clinical suspicion confirmed by microbiological results is an approach often employed in other studies as a reference standard, although this method can restrict the analysis to the easy-to-diagnose patients, and is not always technically applicable to RTIs. Using an expert panel has the advantage of capturing a wider spectrum of illness severities and, therefore, is more

15873_CvHouten_BNW_v2.indd 53 26-11-18 12:42

Page 56: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

54

Chapter 3

likely to be generalizable to clinical practice.16,28 The expert panel was provided with all available clinical information, including information about the course of the disease, all microbiological results (including study-specific multiplex PCR on nasal swabs), and information from a 28-day follow-up assessment. This information was not available to the attending physicians when deciding to start antibiotics or not. A limitation of this study is that not all eligible patients participated in this study for practical reasons (e.g., attending physician didn’t have time to recruit the patient at the ED, parents or patients didn’t want phlebotomy only for study proposes), which may have introduced a selection bias in favour of more severely ill patients and could lead to an overestimation of antibiotic overuse. A second limitation is that, by design, patients with an inconclusive panel diagnosis were excluded. Although it is notable that 98% of whom were receiving antibiotics. Therefore, including these patients would probably not change the results. Third, we collected a nasal swab for every patient for the establishment of patient diagnosis, other microbiological diagnostics (e.g., sputum and blood cultures) were only performed if indicated for routine care. Standardizing more microbiological diagnostics might have led to fewer inconclusive panel diagnoses. A fourth limitation is that we don’t have information on the use of influenza and pneumococcal vaccines available. As a consequence, we cannot exclude that information on vaccine history of participants would have allowed for a more accurate panel diagnosis. Fifth, we cannot exclude that some possible confounders (e.g. comorbidity, hospital admission and site specific protocols) might drive some of the difference in prescribing practices between children and adults. Finally, defined daily antibiotic dosages per 1000 patient days in France, Greece, the UK and the USA is 1.5–3.3-times higher than in The Netherlands.29 Due to the low antibiotic prescription rates in The Netherlands, it is plausible that antibiotic overuse will be even higher in other countries. In the present prospective study, we confirmed that antibiotic overuse in viral RTI is more prevalent among adult patients. In our study, children had less often comorbidity, appeared to be less ill at presentation, and had lower a priori probabilities of having a bacterial infection, compared with adults. Physicians are more inclined to initiate antibiotic treatment if the patient appears to be ill upon presentation, even if a viral pathogen was detected, and do often not adhere to related recommendations.19,20 Therefore, in addition to effective diagnostics tests, education and prescribing feedback are needed to reduce antibiotic overuse in RTI patients.21,22

In conclusion, viral RTI is more common in children, whereas antibiotic overuse is more common in adult patients with RTI, supporting the need for better diagnostics to differentiate between viral and bacterial infection across all ages.

15873_CvHouten_BNW_v2.indd 54 26-11-18 12:42

Page 57: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

55

3

Antibiotic misuse in children and adults

References1. Kronman MP, Zhou C, Mangione-Smith R. Bacterial prevalence and antimicrobial prescribing

trends for acute respiratory tract infections. Pediatrics. 2014;134(4):e956-65.

2. Chancey RJ, Jhaveri R. Fever without localizing signs in children: a review in the post-Hib and postpneumococcal era. Minerva Pediatr. 2009;61(5):489-501.

3. Massin MM, Montesanti J, Gerard P, Lepage P. Spectrum and frequency of illness presenting to a pediatric emergency department. Acta Clin Belg. 2006;61(4):161-5.

4. Donnelly JP, Baddley JW, Wang HE. Antibiotic utilization for acute respiratory tract infections in U.S. emergency departments. Antimicrobial agents and chemotherapy. 2014;58(3):1451-7.

5. Barlam TF, Soria-Saucedo R, Cabral HJ, Kazis LE. Unnecessary Antibiotics for Acute Respiratory Tract Infections: Association With Care Setting and Patient Demographics. Open forum infectious diseases. 2016;3(1):ofw045.

6. Fleming-Dutra KE, Hersh AL, Shapiro DJ, Bartoces M, Enns EA, File TM, Jr., et al. Prevalence of Inappropriate Antibiotic Prescriptions Among US Ambulatory Care Visits, 2010-2011. Jama. 2016;315(17):1864-73.

7. Van den Bruel A, Haj-Hassan T, Thompson M, Buntinx F, Mant D. Diagnostic value of clinical features at presentation to identify serious infection in children in developed countries: a systematic review. Lancet (London, England). 2010;375(9717):834-45.

8. Falsey AR, Becker KL, Swinburne AJ, Nylen ES, Formica MA, Hennessey PA, et al. Bacterial complications of respiratory tract viral illness: a comprehensive evaluation. The Journal of infectious diseases. 2013;208(3):432-41.

9. O’Neill J. Review on Antimicrobial Resistance: Tackling drug-resistant infections globally: final report and recommendations. London 2016. Available from: https://amr-review.org/sites/default/files/AMR%20Review%20Paper%20%20Tackling%20a%20crisis%20for%20the%20health%20and%20wealth%20of%20nations_1.pdf (accessed 4-4-2018).

10. CDC report: Antibiotic resistance threats in the United States 2013. Available from: http://www.cdc.gov/drugresistance/threat-report-2013/pdf/ar-threats-2013-508.pdf (accessed 3-6-2015)

11. Laxminarayan R, Duse A, Wattal C, Zaidi AK, Wertheim HF, Sumpradit N, et al. Antibiotic resistance-the need for global solutions. Lancet Infect Dis. 2013;13(12):1057-98.

12. WHO: Antimicrobial resistance: global report on surveillance. 2014. Available from: http://www.who.int/drugresistance/documents/surveillancereport/en/ (Accessed 26-2-2018).

13. Kapasi AJ, Dittrich S, Gonzalez IJ, Rodwell TC. Host Biomarkers for Distinguishing Bacterial from Non-Bacterial Causes of Acute Febrile Illness: A Comprehensive Review. PLoS One. 2016;11(8):e0160278.

14. Organization WH. Pocket Book of Hospital Care for Children. Geneva, Switzerland: World Health Organization; 2005.

15. Mengelle C, Mansuy JM, Sandres-Saune K, Barthe C, Boineau J, Izopet J. Prospective evaluation of a new automated nucleic acid extraction system using routine clinical respiratory specimens. Journal of medical virology. 2012;84(6):906-11.

16. Bertens LC, Broekhuizen BD, Naaktgeboren CA, Rutten FH, Hoes AW, van Mourik Y, et al. Use of expert panels to define the reference standard in diagnostic research: a systematic review of published methods and reporting. PLoS Med. 2013;10(10):e1001531.

15873_CvHouten_BNW_v2.indd 55 26-11-18 12:42

Page 58: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

56

Chapter 3

17. Rutjes AW, Reitsma JB, Coomarasamy A, Khan KS, Bossuyt PM. Evaluation of diagnostic tests when there is no gold standard. A review of methods. Health Technol Assess. 2007;11(50):iii, ix-51.

18. van Houten CB, de Groot JA, Klein A, Srugo I, Chistyakov I, de Waal W, et al. A host-protein based assay to differentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study. Lancet Infect Dis. 2017;17(4):431-40.

19. Oosterheert JJ, van Loon AM, Schuurman R, Hoepelman AI, Hak E, Thijsen S, et al. Impact of rapid detection of viral and atypical bacterial pathogens by real-time polymerase chain reaction for patients with lower respiratory tract infection. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2005;41(10):1438-44.

20. Lacroix L, Manzano S, Vandertuin L, Hugon F, Galetto-Lacour A, Gervaix A. Impact of the lab-score on antibiotic prescription rate in children with fever without source: a randomized controlled trial. PLoS One. 2014;9(12):e115061.

21. Hallsworth M, Chadborn T, Sallis A, Sanders M, Berry D, Greaves F, et al. Provision of social norm feedback to high prescribers of antibiotics in general practice: a pragmatic national randomised controlled trial. Lancet. 2016;387(10029):1743-52.

22. Dekker ARJ, Verheij TJM, Broekhuizen BDL, Butler CC, Cals JWL, Francis NA, et al. Effectiveness of general practitioner online training and an information booklet for parents on antibiotic prescribing for children with respiratory tract infection in primary care: a cluster randomized controlled trial. The Journal of antimicrobial chemotherapy. 2018.

23. Kornblith AE, Fahimi J, Kanzaria HK, Wang RC. Predictors for under-prescribing antibiotics in children with respiratory infections requiring antibiotics. The American journal of emergency medicine. 2017.

24. Craig JC, Williams GJ, Jones M, Codarini M, Macaskill P, Hayen A, et al. The accuracy of clinical symptoms and signs for the diagnosis of serious bacterial infection in young febrile children: prospective cohort study of 15 781 febrile illnesses. BMJ. 2010;340:c1594.

25. Goriacko P, Saiman L, Zachariah P. Antibiotic Use in Hospitalized Children with Respiratory Viruses Detected by Multiplex Polymerase Chain Reaction. Pediatr Infect Dis J. 2017.

26. The center for disease dynamics: Antibiotic prescribing rates by country. Available from: http://www.cddep.org/tool/antibiotic_prescribing_rates_country#sthash.kiRPO6aH.dpbs (accessed 5-2-2018).

27. Oved K, Cohen A, Boico O, Navon R, Friedman T, Etshtein L, et al. A Novel Host-Proteome Signature for Distinguishing between Acute Bacterial and Viral Infections. PLoS One. 2015;10(3):e0120012.

28. Reitsma JB, Rutjes AW, Khan KS, Coomarasamy A, Bossuyt PM. A review of solutions for diagnostic accuracy studies with an imperfect or missing reference standard. J Clin Epidemiol. 2009;62(8):797-806.

29. van de Sande-Bruinsma N, Grundmann H, Verloo D, Tiemersma E, Monen J, Goossens H, et al. Antimicrobial drug use and resistance in Europe. Emerging infectious diseases. 2008;14(11):1722-30.

15873_CvHouten_BNW_v2.indd 56 26-11-18 12:42

Page 59: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

57

3

Antibiotic misuse in children and adults

S upplementar y materialSupplemental Table 1. Microbiology results. a.

Totaln=284

Viral infectionn=209

Bacterial infectionn=75

Study nasal swab PCR(performed on all patients)0 microorganism 69(24) 44(21) 25(33)

1 microorganism 151(53) 114(55) 37(50)

2 microorganisms 48(17) 38(18) 10(13)

3 microorganisms 16(6) 13(6) 3(4)

Microorganism

Adenovirus 30(11) 28(13) 2(3)

Bocavirus 27(10) 22(11) 5(7)

Coronavirus 13(5) 8(4) 5(7)

Influenza virus A/B 36(13) 30(14) 6(8)

Metapneumovirus 11(4) 9(4) 2(3)

Parainfluenza virus 10(4) 9(4) 1(1)

Respiratory syncytial virus A/B 98(35) 75(36) 23(31)

Rhinovirus A/B/C 62(22) 45(22) 17(23)

Mycoplasma pneumonia 3(1) 1(0.5) 2(5)

Bordetella pertussis 3(1) 0(0) 3(7)

Bordetella parapertussis 1(0.5) 0(0) 1(2)

Routine care

Positive blood culture

Staphylococcus, coagulase-negative 2(1) 1(0.5) 1(1)

Streptococcus pneumoniae 1(0.5) 0(0) 1(1)

Haemophilus influenzae 2(1) 0(0) 2(3)

Fusobacterium necrophorum 1(0.5) 0(0) 1(1)

Positive serology

Respiratory syncytial virus A/B 3(1) 2(1) 1(1)

Positive sputum culture/PCR

Respiratory syncytial virus A/B 12(4) 6(3) 6(8)

Moraxella catarrhalis 11(4) 1(0.5) 10(13)

Haemophilus influenzae 9(3) 2(1) 7(9)

Streptococcus pneumoniae 5(2) 0(0) 5(7)

Enterobacter cloacae 4(1) 1(0.5) 3(4)

Group A streptococcus 1(0.5) 1(0.5) 0(0)

Pseudomonas aeruginosa 1(0.5) 0(0) 1(1)

15873_CvHouten_BNW_v2.indd 57 26-11-18 12:42

Page 60: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

58

Chapter 3

Supplemental Table 1. Microbiology results. (Continued)

a.

Totaln=284

Viral infectionn=209

Bacterial infectionn=75

Positive nasopharyngeal culture/PCR

Group A streptococcus 5(2) 1(0.5) 4(5)

Streptococcus pneumoniae 1(0.5) 1(0.5) 0(0)

Haemophilus influenzae 1(0.5) 1(0.5) 0(0)

Mycoplasma pneumoniae 1(0.5) 0(0) 1(1)

Bordetella pertussis 1(0.5) 0(0) 1(1)

Respiratory syncytial virus A/B 19(7) 13(6) 6(8)

Adenovirus 5(2) 3(1) 2(5)

Influenza virus 5(2) 4(2) 1(1)

Rhinovirus 3(1) 1(0.5) 2(5)

Metapneumovirus 3(1) 3(1) 0(0)

Coronavirus 1(0.5) 1(0.5) 0(0)

Parainfluenza virus 1(0.5) 1(0.5) 0(0)

b. Total

n=232Viral infection

n=89Bacterial infection

n=143Study nasal swab PCR(performed on all patients)0 microorganism 112(48) 17(19) 95(66)

1 microorganism 115(50) 68(76) 47(33)

2 microorganisms 5(2) 4(5) 1(1)

Microorganism

Adenovirus 3(1) 2(2) 1(1)

Bocavirus 1(0.5) 0(0) 1(1)

Coronavirus 4(2) 1(1) 3(2)

Influenza virus A/B 52(22) 35(39) 17(12)

Metapneumovirus 6(3) 4(5) 2(1)

Parainfluenza virus 5(2) 4(5) 1(1)

Respiratory syncytial virus A/B 18(8) 14(16) 4(3)

Rhinovirus A/B/C 22(10) 16(18) 6(4)

Mycoplasma pneumoniae 11(5) 0(0) 11(8)

Bordetella pertussis 3(1) 0(0) 3(2)

Routine care

Positive blood culture/PCR

Streptococcus pneumoniae 5(2) 0(0) 5(3)

Moraxella osloensis 1(0.5) 0(0) 1(1)

Chlamydophila pneumoniae 1(0.5) 0(0) 1(1)

15873_CvHouten_BNW_v2.indd 58 26-11-18 12:42

Page 61: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

59

3

Antibiotic misuse in children and adults

Supplemental Table 1. Microbiology results. (Continued)

b.

Totaln=232

Viral infectionn=89

Bacterial infectionn=143

Positive serology

Bordetella pertussis 1(0.5) 0(0) 1(1)

Mycoplasma pneumoniae 1(0.5) 0(0) 1(1)

CMV 1(0.5) 1(1) 0(0)

EBV 1(0.5) 1(1) 0(0)

Mycobacterium tuberculosis 1(0.5) 0(0) 1(1)

Positive sputum culture

Haemophilus influenzae 15(6) 1(1) 14(10)

Moraxella catarrhalis 3(1) 0(0) 3(2)

Serratia ureilytica 1(0.5) 0(0) 1(1)

Staphylococcus aureus 5(2) 1(1) 4(3)

Streptococcus pneumoniae 4(1.5) 0(0) 4(3)

Acinetobacter baumannii 3(1) 0(0) 3(2)

Candida albicans 5(2) 0(0) 5(3)

Candida tropicalis 3(1) 1(1) 2(1)

Candida glabrata 2(1) 0(0) 2(1)

Klebsiella oxytoca 1(0.5) 0(0) 1(1)

Pseudomonas aeruginosa 5(2) 1(1) 4(3)

Positive nasopharyngeal culture/PCR

Group A streptococcus 1(0.5) 0(0) 1(1)

Staphylococcus aureus 1(0.5) 0(0) 1(1)

Mycoplasma pneumoniae 7(3) 0(0) 7(5)

Candida albicans 2(1) 1(1) 1(1)

Metapneumovirus 4(1.5) 3(3) 1(1)

Respiratory syncytial virus A/B 4(1.5) 3(3) 1(1)

Adenovirus 3(1) 2(2) 1(1)

Rhinovirus 3(1) 3(3) 0(0)

Influenza virus 26(11) 18(20) 8(6)

Parainfluenza virus 1(0.5) 1(1) 0(0)

a. Paediatric cohort. b. Adult cohort. Microbiology results from study specific nasal swab PCR (performed on all patients) and positive results from microbiology diagnostics performed for routine care. Reference standard outcomes are based on the majority consensus of the expert panel. Mixed infection was considered as ‘bacterial’. Data are presented as n(%).

15873_CvHouten_BNW_v2.indd 59 26-11-18 12:42

Page 62: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

60

Chapter 3

Supp

lem

enta

l Tab

le 2

. Cas

es w

ith

a re

fere

nce

stan

dard

dia

gnos

is o

f bac

teri

al in

fect

ion

not t

reat

ed w

ith

anti

biot

ics.

Pt.

Clin

ical

synd

rom

eA

ge

(y)

Sex

Coun

try

Max

imal

te

mpe

ratu

re

(°C)

CRP

Hos

pita

lizat

ion

dura

tion

(d)

Com

orbi

dity

Mic

robi

olog

yCl

inic

al d

etai

lsCh

ildre

n

1Br

onch

iolit

is0.

25M

IL37

.421

3Pu

lmon

ary

valv

e st

enos

isN

asal

sw

ab: B

orde

tella

pa

rape

rtus

sis,

RSV

and

rhin

oviru

s.

Pres

ente

d w

ith c

ough

an

d dy

spne

a si

nce

5 da

ys.

Clin

ical

exa

min

atio

n: b

ilate

ral

lym

phad

enop

athy

cer

vica

l, cr

epita

tion,

whe

eze

and

syst

olic

mur

mur

. Che

st X

-ray

: no

rmal

.Ad

ults

1CO

PD

Exac

erba

tion

66F

NL

36.8

5

9D

M2,

CO

PD, T

IA,

schi

zoph

reni

c,

fibro

mya

lgia

Sput

um c

ultu

re:

Hae

mop

hilu

s infl

uenz

aePr

esen

ted

with

cou

gh a

nd

dysp

nea

sinc

e 7

days

. Clin

ical

ex

amin

atio

n: w

heez

e an

d pr

olon

ged

expi

ratio

n. C

hest

X-

ray:

nor

mal

. Req

uire

d ox

ygen

eve

n af

ter d

isch

arge

.2

COPD

Ex

acer

batio

n40

MN

L38

.86

7CO

PD, a

sthm

a,

dust

alle

rgy,

ad

dict

ion

to

ciga

rett

es,

hero

in a

nd

coca

ine

Sput

um c

ultu

re:

Stre

ptoc

occu

s pne

umon

ia,

Citr

obac

ter k

oser

i, Ca

ndid

a al

bica

ns, I

nflue

nza

viru

s. Bl

ood

cultu

re: n

egat

ive

Pres

ente

d w

ith d

yspn

ea,

dry

coug

h, h

eada

che

and

mya

lgia

sin

ce 4

day

s. Cl

inic

al

exam

inat

ion:

whe

eze

only

. Ch

est X

-ray

: nor

mal

(tw

ice)

.Re

quire

d ox

ygen

for 3

day

s.3

COPD

Ex

acer

batio

n47

MIL

36.5

11

Non

eBr

onch

oalv

eola

r lav

age:

St

aphy

loco

ccus

aur

eus

Pres

ente

d w

ith d

yspn

ea

and

coug

h si

nce

14 d

ays.

CT-

ches

t: In

filtr

ativ

e in

flam

mat

ion

and

cons

olid

atio

n.D

urin

g ad

mis

sion

, CO

PD w

as

diag

nose

d.CR

P: C

-rea

ctiv

e pr

otei

n, F

: fem

ale,

M:m

ale,

NL:

The

Net

herla

nds,

IL: I

srae

l, CO

PD: C

hron

ic O

bstr

uctiv

e Pu

lmon

ary

Dis

ease

, DM

2: D

iabe

tes M

ellit

us T

ype

2, T

IA: T

rans

ient

Is

chem

ic A

ttac

k.

15873_CvHouten_BNW_v2.indd 60 26-11-18 12:42

Page 63: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

61

3

Antibiotic misuse in children and adults

Supplemental Table 3. Baseline of bacterial and viral RTI children and adults, The Netherlands versus Israel.

Children Adults

NLn=136

ILn=148

NLn=131

ILn=101

Age, years 0.9[0.2-3.1] 1.7[0.9-3.0] 66[56-74] 61[40-76]

Male sex 81(60) 86(58) 71(54) 60(59)

Presence of comorbidity 74(54) 51(35) 122(93) 77(76)

Ill-appearing 77(58) 36(24) 50(42) 64(66)

Maximal temperature, °C 39,1(0,8) 39,3(0,9) 38,7(0,9) 38,4(1,1)

Duration of symptoms, days 3(2) 3(2) 3(2) 4(3)

Hospital admission 125(92) 83(59) 124(95) 93(93)

Hospitalization duration, days 5[3-14] 3[2-4] 6[4-9] 3[2-6]

CRP (mg/L) at admission 22[7-53] 11[3-34] 92[35-203] 11[4-24]

Disease severity

Oxygen saturation, %

Needed mechanical ventilation 31(23) 0(0) 3(2) 0(0)

Death 1(1) 0(0) 1(1) 2(2)

Admission site

Secondary care centre 50(37) 148(100) 74(57) 99(98)

Tertiary care centre 47(34) 0(0) 53(40) 0(0)

ICU 39(29) 0(0) 4(3) 2(2)

Clinical syndrome

COPD/Asthma exacerbation 0(0) 4(3) 39(30) 6(6)

LRTI 80(59) 70(47) 87(66) 85(84)

URTI 56(41) 74(50) 5(4) 10(10)

Antibiotic treatment 64(47) 87(59) 117(89) 97(96)

Study nasal swab PCR

0 microorganism 16(12) 55(37) 67(51) 58(57)

1 microorganism 90(66) 63(43) 63(48) 40(40)

2 microorganisms 21(15) 24(16) 1(1) 3(3)

3 microorganisms 9(7) 6(4) 0(0) 0(0)

Microorganism

Adenovirus 8(6) 22(15) 0(0) 3(3)

Bocavirus 16(12) 11(7) 1(1) 0(0)

Influenza virus 21(15) 15(10) 33(25) 19(19)

Rhinovirus 34(25) 28(19) 12(9) 10(10)

RSV 65(48) 33(22) 9(7) 9(9)

Other* 14(10) 20(14) 10(8) 5(5)

15873_CvHouten_BNW_v2.indd 61 26-11-18 12:42

Page 64: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

62

Chapter 3

Supplemental Table 3. Baseline of bacterial and viral RTI children and adults, The Netherlands versus Israel. (Continued)

Children Adults

NLn=136

ILn=148

NLn=131

ILn=101

Expert panel diagnosis

Bacterial infection 32(24) 43(29) 84(64) 59(58)

Viral infection 104(76) 105(71) 47(46) 42(42)

Antibiotic overuse 32(31) 45(43) 35(74) 39(93)

Data are presented as n(%), mean (SD), or median [IQR]. LRTI included pneumonia, acute bronchitis and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. NL: The Netherlands, IL: Israel, CRP: C-reactive protein, ICU: intensive care unit, COPD: Chronic Obstructive Pulmonary Disease, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection, RSV: respiratory syncytial virus. *Includes coronavirus, human metapneumovirus, and parainfluenza virus.

616 patients

recruited

307 patients

with RTI

recruited in

the Netherlands

309 patients

with RTI

recruited in

Israel

83 microbiologically

confirmed bacterial

diagnosis

203 microbiologically

confirmed viral

diagnosis

145

child

58

adult

AB +

n=50

AB -

n=95

27

child

56

adult

AB +

n=50

AB -

n=8AB +

n=27

AB -

n=0

AB +

n=54

AB -

n=2

516 unanimous

diagnosis by

expert panel

(reference standard)

Supplemental Figure 1. Flowchart of microbiologically confirmed patients.AB - : antibiotics not prescribed, AB +: antibiotics prescribed, RTI: respiratory tract infection.

15873_CvHouten_BNW_v2.indd 62 26-11-18 12:42

Page 65: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

63

3

Antibiotic misuse in children and adults

0

10

20

30

40

50

60N

umbe

r of a

gent

pre

scrip

tions

Antibiotic agent

Children NL

Children IL

Adults NL

Adults IL

Supplemental Figure 2. Number of antibiotic agents used per country for children and adults with RTIs.

NL: The Netherlands, IL; Israel.

15873_CvHouten_BNW_v2.indd 63 26-11-18 12:42

Page 66: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

4

15873_CvHouten_BNW_v2.indd 64 26-11-18 12:42

Page 67: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

4A host-protein based assay to differentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study

CB van Houten, JAH de Groot, A Klein, I Srugo, I Chistyakov, W de Waal, CB Meijssen, W Avis, TFW Wolfs, EAM Sanders and LJ Bont

Lancet Infect Dis. 2017 Apr;17(4):431-440

15873_CvHouten_BNW_v2.indd 65 26-11-18 12:42

Page 68: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

66

Chapter 4

Abstrac tBackground A physician is frequently unable to distinguish bacterial from viral infections. ImmunoXpert is a novel assay combining three proteins: tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), interferon gamma induced protein-10 (IP-10), and C-reactive protein (CRP). We aimed to externally validate the diagnostic accuracy of this assay in differentiating between bacterial and viral infections and to compare this test with commonly used biomarkers.

MethodsIn this prospective, double-blind, international, multicentre study, we recruited children aged 2–60 months with lower respiratory tract infection or clinical presentation of fever without source at four hospitals in the Netherlands and two hospitals in Israel. A panel of three experienced paediatricians adjudicated a reference standard diagnosis for all patients (i.e., bacterial or viral infection) using all available clinical and laboratory information, including a 28-day follow-up assessment. The panel was masked to the assay results. We identified majority diagnosis when two of three panel members agreed on a diagnosis and unanimous diagnosis when all three panel members agreed on the diagnosis. We calculated the diagnostic performance (i.e., sensitivity, specificity, positive predictive value, and negative predictive value) of the index test in differentiating between bacterial (index test positive) and viral (index test negative) infection by comparing the test classification with the reference standard outcome.

ResultsBetween Oct 16, 2013 and March 1, 2015, we recruited 777 children, of whom 577 (mean age 21 months, 56% male) were assessed. The majority of the panel diagnosed 71 cases as bacterial infections and 435 as viral infections. In another 71 patients there was an inconclusive panel diagnosis. The assay distinguished bacterial from viral infections with a sensitivity of 86.7% (95% CI 75.8–93.1), a specificity of 91.1% (87.9–93.6), a positive predictive value of 60.5% (49.9–70.1), and a negative predictive value of 97.8% (95.6–98.9). In the more clear cases with unanimous panel diagnosis (n=354), sensitivity was 87.8% (74.5–94.7), specificity 93.0% (89.6–95.3), positive predictive value 62.1% (49.2–73.4), and negative predictive value 98.3% (96.1–99.3).

Conclusion This external validation study shows the diagnostic value of a three-host protein-based assay to differentiate between bacterial and viral infections in children with lower respiratory tract infection or fever without source. This diagnostic based on CRP, TRAIL, and IP-10 has the potential to reduce antibiotic misuse in young children.

15873_CvHouten_BNW_v2.indd 66 26-11-18 12:42

Page 69: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

67

4

Validation of a host-protein based assay

Introduc tionThe proportion of bacterial infections is low in children presenting at the hospital emergency department with fever without source (range, 0.02–0.7%) and acute respiratory tract infections (26.5–28.3%).1,2 Unfortunately, it is often not possible to differentiate between bacterial and non-bacterial disease on the basis of clinical judgment alone. Consequentially, antibiotics are prescribed almost twice as often as required in children with acute respiratory tract infections in the USA.1 Antibiotic overuse is associated with antibiotic resistance expansion, causing 25 000 deaths in Europe annually.3,4 Conversely, it is estimated that 22–47% of all patients with a bacterial infection fail to get antibiotic treatment within the recommended timeframe due to delayed or missed diagnosis, leading to medical complications and increased mortality (odds ratio [OR] 1.15).5–7 These data create an incentive to introduce diagnostic tools that can aid in distinguishing between bacterial and non-bacterial causes of infection. A study by Oved and colleagues8 showed that a novel host–response-based diagnostic assay, ImmunoXpert, distinguishes bacterial infection from viral infection with high accuracy. This assay integrates the concentrations of three biomarkers: tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), interferon gamma induced protein-10 (IP-10), and C-reactive protein (CRP). The diagnostic value of TRAIL is noteworthy since its dynamics are complementary to traditionally studied bacteria-induced proteins such as CRP and procalcitonin; TRAIL concentrations increase in viral infection and decrease during bacterial infection. A systematic review9 by Kapasi and colleagues showed that the study8 from Oved and colleagues describing our index test was of high quality. In the present investigator-initiated study, named the OPPORTUNITY study, we aimed to externally validate the diagnostic importance of this assay for discrimination of bacterial versus viral disease in children aged between 2 and 60 months presenting at the hospital with fever without source or lower respiratory tract infection, and we compared its diagnostic accuracy with the commonly applied CRP and procalcitonin.

MethodsStudy design and participants Patient recruitment for this prospective, double-blind study took place in convenience series at the emergency departments and paediatric wards of three secondary and one university hospital in the Netherlands, and two secondary hospitals in Israel. Patients aged between 2 and 60 months presenting with fever (peak temperature ≥38.0°C measured by axillary, rectal, or ear thermometer) and symptoms of lower respiratory tract infection or fever without source existing for a maximum of 6 days were eligible for inclusion. Children were recruited consecutively; if a child was eligible for recruitment, a member of the study team would ask parents for permission to participate in the study

15873_CvHouten_BNW_v2.indd 67 26-11-18 12:42

Page 70: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

68

Chapter 4

until the predefined number of patients was achieved. Children between 1 and 2 months of age were recruited for secondary analyses because the assay had not previously been assessed in this age group. Patients were excluded in case of a previous episode of fever in the past 3 weeks; psychomotor retardation; moderate to severe metabolic disorder; primary or secondary immunodeficiency; HIV, infection by hepatitis B or hepatitis C viruses; and active malignancies. Patients who received antibiotics at any time before the beginning of the study were not excluded. To obtain reference values of this novel diagnostic, a non-infectious control population was recruited before elective procedures (e.g., cardiothoracic surgery, hernia correction). The index test was done at the same time as the reference test. Parents gave written informed consent before sampling. The OPPORTUNITY study was approved by the ethics committees in the participating countries. The report of this study is written following the STARD reporting guidelines. The full study protocol is with the investigators and can be requested.

Procedures24 h after presentation, a venous blood sample and a nasal swab (universal transport medium, Copan, Brescia, Italy) were collected. Within 2 h after collection, blood was fractionated into serum and−together with the nasal swab−stored at -80°C until transfer to MeMed Diagnostics (Tirat Carmel, Israel). Staff at MeMed Diagnostics did the assay and measured additional procalcitonin concentrations on the serum sample by ELISA using LIASON BRAHMS PCT (DiaSorin, Saluggia, Italy), and Elecsys BRAHMS PCT (Roche Diagnostics, Indianopolis, IN, USA). They also used multiplex PCR testing for 15 common respiratory viruses (Seeplex RV15, Seegene, Seoul, South Korea) on the nasal swab.The index test combines the concentrations of TRAIL, IP-10, and CRP using a predetermined logistic regression formula to compute the likelihood score for a bacterial or viral infection as described previously.8 On the basis of the likelihood score of the index test, each patient was classified as having a bacterial or a viral immune response. According to the current ConformitéEuropéenne−In Vitro Diagnostic (CE-IVD) label, patients with an index test score below 0.35 were classified as viral, between 0.35 and 0.65 as equivocal, and above 0.65 as bacterial (Supplemental Figure 1).8 An equivocal or intermediate score is used more often in diagnostic tests.10 To provide a classification for all patients, thus without equivocal test results, we additionally analysed test results using a single cut-off value of 0.5, whereby the test predicts 50% chance of bacterial infection and 50% chance of viral infection.We did a 28-day post recruitment phone call for clinical follow-up. Clinical data were collected from medical records by use of an electronic case report form (eCRF). The eCRF consisted of demographics, medical history, physical examination findings, all available

15873_CvHouten_BNW_v2.indd 68 26-11-18 12:42

Page 71: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

69

4

Validation of a host-protein based assay

radiological and laboratory data collected for routine care (including procalcitonin, white blood cell, and absolute neutrophil count), microbiological investigation results (including study-specific nasal swab data), and follow-up information.Currently, no single reference standard test exists for identifying the cause of an infection.11 Therefore, we followed England’s National Health Service’s standard for assessing diagnostic tests and compiled an expert panel reference standard.12 The panel comprised paediatricians with more than 10 years of clinical experience. They received a review of paediatric, emergency medicine, and infectious disease literature written by the study team as background information without any instructions or decision rules. Every recruited patient was diagnosed by three panel members affiliated to the country of recruitment using all available eCRF information, but masked for the index test results (which were done simultaneously) and for the decisions of their peers. Each expert assigned one of the following causes to each patient: bacterial infection, viral infection, mixed infection (i.e., bacterial and viral co-infection), non-infectious disease, or indeterminate. Patients assigned as mixed infection were later classified as bacterial because they are clinically managed similarly. The expert panel was instructed to give their diagnosis in relation to the moment of sampling.We distinguished two types of bacterial infection diagnosis: bacterial infection by majority agreement and bacterial infection by unanimous agreement. Majority agreement was established when at least two panel members classified disease cause as bacterial, which was considered the preferred primary outcome (Supplemental Figure 1). This approach was predicted to be conservative, and it could be argued the estimation of diagnostic assessment would be more accurate if obtained using cases for which all panel members assign the same cause. We therefore established bacterial infection by unanimous agreement when all three panel members classified disease cause as bacterial, which was a more stringent primary outcome. The subset of patients with a microbiologically confirmed infection (Supplemental Figure 2) was analysed separately. Microbiologically confirmed bacterial diagnosis was predefined in the statistical analysis plan as unanimous panel diagnosis plus a bacteraemia with positive blood culture, bacterial meningitis with positive CSF culture, urinary tract infection with positive urine culture, or tonsillitis with throat culture positive for streptococci. Microbiologically confirmed viral diagnosis was defined as detection of at least one virus plus unanimous panel diagnosis.Cases were assigned an inconclusive outcome if each panel member assigned a different cause for infection and when at least two panel members diagnosed the case as indeterminate. By design, these cases could not be used to assess diagnostic performance.

15873_CvHouten_BNW_v2.indd 69 26-11-18 12:42

Page 72: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

70

Chapter 4

Blinding was a critical component of the OPPORTUNITY study. The index test was done on anonymous serum samples in the absence of any clinical or other patient related information. Final expert panel diagnosis was made using all available eCRF information, but blinded for the decisions of their peers and for the results of the index test. Only after all data were obtained and the definite database was locked, were the index test results unblinded, enabling the pre-specified analyses to be done.The primary outcome was the diagnostic performance of the index test in differentiating between bacterial and viral causes in patients aged 2–60 months. The diagnostic performance of the index test compared with CRP, and with procalcitonin was assessed as well. The secondary outcome according to the protocol would have been the performance of the index test in discriminating infectious and non-infectious causes of disease, but these data are not reported.

Statistical analysisFor the primary outcome, we calculated the diagnostic performance (i.e., sensitivity, specificity, positive predictive value and negative predictive value, positive and negative likelihood ratios, diagnostic odds ratio, and the area under the receiver operating curves [AUC]) of the index test in differentiating between bacterial (index test positive) and viral (index test negative) causes in patients aged 2–60 months by comparing the test classification with the reference standard outcome. These test outcomes were completed for two cohorts, the majority consensus cohort and the unanimous consensus cohort, which were decided by the expert panel. For these analyses, patients with an index test score below 0.35 were classified as viral, between 0.35 and 0.65 as equivocal, and above 0.65 as bacterial (Supplemental Figure 1). Accuracy measures were calculated from outcomes from the CE-IVD cut-offs, excluding equivocal results.For the comparison between the index test and CRP, and with procalcitonin, we compared the cut-off independent discriminative outcome of the index test by calculating the AUC. To examine the clinical effect of the index test, we assessed improvement in classification, by identifying the number of patients correctly and incorrectly reclassified using the index test instead of CRP or procalcitonin tests.13 PWe used predefined cut-off values (CRP 40 μg/mL, procalcitonin 0.5 ng/mL) as commonly used by others.14–18 Additionally, we analysed the accuracy of the index test when an alternative cut-off of 0.5 was used, such that the test predicts 50% chance of bacterial infection and 50% chance of viral infection. Finally, we did specific subgroup analyses of patients in the microbiologically confirmed sub-cohort, patients admitted to hospital, non-admitted patients, children aged between 1 and 2 months, and patients admitted to the paediatric intensive-care unit per clinical syndrome, and patients treated with and without antibiotics at the moment of recruitment. To prevent overestimations we

15873_CvHouten_BNW_v2.indd 70 26-11-18 12:42

Page 73: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

71

4

Validation of a host-protein based assay

did the secondary analyses on only the majority cohort. A p value of less than 0.05 was considered significant. We used SPSS version 21.0 for Windows and R version 3.2.2 to analyse the statistical data. The OPPORTUNITY study is registered with ClinicalTrials.gov, number NCT01931254.

Role of the funding sourceThe funder of the OPPORTUNITY study had no role in the study design, data collection, data analysis, data interpretation, or the writing of the report. The corresponding author had full access to all the data in the study and had final responsibility for the decision to submit for publication.

ResultsA total of 777 children in Dutch and Israeli hospitals were recruited between Oct 16, 2013, and Jan 28, 2015, with final follow-up done on March 1, 2015. Children aged between 1 and 2 months (n=34) and non-infectious patients (n=103) were excluded from primary analysis. In 63 children the index test was not measured, either because the children did not fulfil inclusion criteria, did not have an available blood sample, or consent was withdrawn (Figure 1). Baseline characteristics of these patients are shown in Supplemental Table 1a. Eventually, 577 patients were available for primary analysis (Table 1), and had a mean age of 21 months, with boys making up 56% of the participants. In most patients one or two microorganisms were found in the study nasal swab. In these swabs rhinovirus and respiratory syncytial virus were the most commonly found pathogens. In routine care 21 children had a positive urine culture and 13 children had a positive blood culture (Supplemental Table 2). There were some differences between the patients recruited in the different countries (Supplemental Table 1b). Overall, the Dutch cohort included children with more severe disease presentation than did the Israeli cohort with longer duration of hospital stay and higher need for admission to intensive-care units.

The expert panel assigned reference standard outcomes to all 577 cases included in the primary analysis (Figure 1). Of 506 cases with a conclusive outcome, 406 had unanimous agreement, and 100 a majority agreement from the expert panel. The panel diagnosed 71 patients as having a bacterial infection and 435 as having a viral infection. In another 71 cases the reference standard outcome was inconclusive; in 16 cases each panel member assigned a different diagnosis and in 55 cases the majority assigned the case as indeterminate (Supplemental Table 3). Cases with inconclusive panel diagnosis were excluded from the primary analysis (Table 1).

15873_CvHouten_BNW_v2.indd 71 26-11-18 12:42

Page 74: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

72

Chapter 4

202 patients recruited in the Netherlands 575 patients recruited in Israel

777 recruited

640 in primary study population

137 excluded from primary analyses*

34 aged 1-2 months

103 non-infectious patients

577 in primary analysis

63 excluded before unblinding

41 did not fulfil inclusion criteria

20 did not have blood sample

2 other

71 diagnosed as bacterial

by expert panel

(reference standard)

435 diagnosed as viral

by expert panel

(reference standard)

71 diagnosed as inconclusive

by expert panel†

(reference standard)

71 index test

52 bacterial infections

8 viral infections

11 equivocal infections

435 index test

34 bacterial infections

349 viral infections

52 equivocal infections

71 index test

27 bacterial infections

37 viral infections

7 equivocal infections

Figure 1. Trial design.Recruitment and flow of children presenting at the hospital with fever without source and acute respiratory tract infections to differentiate between bacterial and viral origin. Expert panel diagnoses can be divided into majority, unanimous, and microbiologically confirmed groups. *Children between 1 and 2 months of age and non-infectious patients were recruited for sub-analyses to study the test outcomes in these populations. †Inconclusive expert panel diagnosis was assigned to cases in which each panel member gave a different diagnosis or in which two or more members diagnosed the case as indeterminate. These inconclusive diagnoses were excluded from the analysis.

15873_CvHouten_BNW_v2.indd 72 26-11-18 12:42

Page 75: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

73

4

Validation of a host-protein based assay

Table 1. Baseline characteristics of patients included in primary analysis.

Totaln=577

Viral infection

n=435

Bacterial infection

n=71Inconclusive

n=71 p-value*Age (months)(mean, SD)

21(16) 20(16) 24(17) 25(17) 0.044

Gender, male (n,%) 324(56) 246(57) 36(51) 42(59) 0.370

Maximal temperature (°C)(mean, SD)

39.4(0.8) 39.3(0.8) 39.7(0.8) 39.4(0.9) <0.0001

Duration of symptoms (days) (mean, SD)

2.8(1.7) 2.7(1.7) 3.0(1.8) 2.7(1.8) 0.277

Hospital admission (n,%) 316(55) 219(50) 59(83) 38(53) <0.0001

Hospitalization duration (days) (median, IQR)

3(2-4) 3(2-4) 4(3-5) 3(3-5) <0.0001

Antibiotic treatment prescribed (n,%) 224(39) 100(23) 71(100) 53(75) <0.0001

CRP (mg/L) at admission(median, IQR)

21(7-52)

14.4(6-32)

123(52-226)

50.4(18-80)

<0.0001

Disease severity

Need of mechanical ventilation (n,%)

8(1) 4(1) 3(4) 1(1) 0.062

Deaths (n,%) 0(0) 0(0) 0(0) 0(0) NA

Recruiting site 0.032

Secondary care centre (n,%) 528(92) 400(92) 63(89) 65(92)

Tertiary care centre (n,%) 35(6) 27(6) 3(4) 5(7)

PICU (n,%) 14(2) 8(2) 5(7) 1(1)

Clinical syndrome <0.0001

Bacteraemia /viraemia 7(1) 3(1) 3(4) 1(1)

CNS 11(2) 10(2) 1(1) 0(0)

FWS 173(30) 153(35) 4(6) 16(23)

GE 18(3) 17(4) 0(0) 1(1)

LRTI 141(24) 100(23) 24(34) 17(24)

URTI 155(27) 120(28) 13(18) 22(31)

UTI 18(3) 0(0) 16(23) 2(4)

Other 54(9) 32(7) 10(14) 12(17)

Reference standard outcomes were based on majority agreement of the expert panel. Mixed infection was considered as bacterial. Clinical syndrome was based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. Baseline characteristics of excluded patients are shown in the appendix. *p-values of differences between bacterial and viral infections. CRP: C-reactive protein, PICU: paediatric intensive care units, CNS: central nervous system, FWS: fever without source, GE: gastro-enteritis, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection, UTI: urinary tract infection. NA = not applicable.

15873_CvHouten_BNW_v2.indd 73 26-11-18 12:42

Page 76: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

74

Chapter 4

Table 2 shows the results of the index test for different cohorts, which occurred at the same time as the reference test. When using the majority cohort (n=443) for the primary outcome, the sensitivity and specificity of the index test for diagnosing bacterial infections was 86.7% (95% CI 75.8–93.1) and 91.1% (87.9–93.6; Table 3). The positive predictive value was 60.5% (49.9–70.1) and the negative predictive value 97.8% (95.6–98.9). The accuracy of the index test for the second primary outcome, the unanimous cohort (n=354), was slightly better than for the majority cohort (Table 3). The receiver operating characteristics curve of the majority cohort showed good discriminative ability of the index test with an AUC of 0.90 (0.86–0.95), and 0.92 (0.87–0.97) with the unanimous cohort (Figure 2).

Table 2. Results of the index test per used reference standard cohort using the CE-IVD label.Number of

cases classified as bacterial by reference test

Number of cases classified

as viral by reference test

CE-IVD classification cut-off

Reference test: majority cohort (n=506)

Viral (<0.35) 8(1.8%) 349(78.8%)

Equivocal (>0.35-<0.65) 11(2.5%) 52(11.7%)

Bacterial (>0.65) 52(11.7%) 34(7.7%)

Reference test: unanimous cohort (n=406)

Viral (<0.35) 5(1.4%) 291(82.2%)

Equivocal (>0.35-<0.65) 8(2.3%) 44(12.4%)

Bacterial (>0.65) 36(10.2%) 22(6.2%)

Reference test: microbiologically confirmed cohort (n=335)

Viral (<0.35) 2(0.7%) 257(87.1%)

Equivocal (>0.35-<0.65) 1(0.3%) 39(13.2%)

Bacterial (>0.65) 16(5.4%) 20(6.8%)

Alternative cut-off of 0.5

Reference test: majority cohort (n=506)

Viral (<0.5) 11(2.2%) 379(74.9%)

Bacterial (>0.5) 60(11.9%) 56(11.1%)

According to the current CE-IVD label, patients with an index test score below 0.35 were classified as viral, between 0.35 and 0.65 as equivocal, and above 0.65 as bacterial. An alternative cut-off of 0.5 was also used for the index test classification, such that the test predicted a 50% chance of bacterial infection and 50% chance of viral infection using the majority cohort.

15873_CvHouten_BNW_v2.indd 74 26-11-18 12:42

Page 77: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

75

4

Validation of a host-protein based assay

a.

Specificity

Sens

itivi

ty

0.0

0.2

0.4

0.6

0.8

1.0

1.0 0.8 0.6 0.4 0.2 0.0

Index test AUC = 0.90 (95% CI 0.86 − 0.95 )CRP AUC = 0.89 (95% CI 0.84 − 0.94 )PCT AUC = 0.84 (95% CI 0.79 − 0.90 )

b.

Specificity

Sens

itivi

ty

0.0

0.2

0.4

0.6

0.8

1.0

1.0 0.8 0.6 0.4 0.2 0.0

Index test AUC = 0.90 (95% CI 0.86 − 0.95 )CRP AUC = 0.89 (95% CI 0.84 − 0.94 )PCT AUC = 0.84 (95% CI 0.79 − 0.90 )

Figure 2. Receiver operating characteristics (ROC) curve of sensitivity versus specificity for the index test, CRP and PCT. a. Area under the receiving operating curve (AUC) for the index test, CRP and PCT for all patients that are part of the majority cohort and had CRP and PCT values available (n=479). The AUC difference between the index test and PCT is 0.06 (p=0.02). b. Area under the receiving operating curve (AUC) for the index test, CRP and PCT for all patients that are part of the unanimous cohort and had CRP and PCT values available (n=387). The AUC difference between the index test and PCT is 0.08 (p=0.01). CRP: C-reactive protein, PCT: procalcitonin

We compared the AUC of CRP and procalcitonin to diagnose bacterial infection with the index test. We found that the AUC from the index test was not different from the AUC for CRP, but significantly higher compared with the AUC for procalcitonin (p=0.02 majority cohort, p=0.01 unanimous cohort; Figure 2). The index test showed similar performance results in the microbiologically confirmed (n=295) sub-cohort (Table 3). Test performance was also similar when using a cut-off of 0.5 of the index test (Table 3). In children with bacterial infections, mean CRP con centration was higher (p<0.0001) and TRAIL con centration lower (p<0.0001) than in children with viral infections (Supplemental Figure 3). In non-infectious control patients, CRP and IP-10 concentrations were lower than in infectious patients, TRAIL concentrations were lower in non-infectious control patients than in children with viral infections, but higher than in those with bacterial infections.

We closely examined the clinical characteristics of the patients in which the index test results and the panel diagnosis were discordant (i.e., false-negative and false positive index test results). Details of all eight cases (2%) that were incorrectly classified as viral by the index test are described in table 4. One patient had blood culture positive for Kingella kingae, one patient had Bordetella pertussis infection. In 34 cases (7%) in which the index test was false positive for bacterial infection, we did not identify a clear clinical pattern. In these 34 cases, 24 (71%) patients recovered without antibiotic treatment, mean hospitalisation duration was 2 days, and two children needed mechanical

15873_CvHouten_BNW_v2.indd 75 26-11-18 12:42

Page 78: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

76

Chapter 4

ventilation (not shown). Five (15%) of these patients received antibiotic treatment before recruitment.

Table 3. Diagnostic test accuracy measures of the index test per used reference standard.Majority

cohortUnanimous

cohortMicrobiologically confirmed cohort

CE-IVD cut-off

Number of patients in subgroup 443 354 295

Sensitivity 86.7(75.8-93.1) 87.8(74.5-94.7) 88.9(67.2-96.9)

Specificity 91.1(87.9-93.6) 93.0(89.6-95.3) 92.8(89.1-95.3)

Positive predictive value 60.5(49.9-70.1) 62.1(49.2-73.4) 44.4(29.5-60.4)

Negative predictive value 97.8(95.6-98.9) 98.3(96.1-99.3) 99.2(97.2-99.8)

Positive likelihood ratio 9.8(7.0-13.7) 12.5(8.2-19.0) 12.3(7.8-19.4)

Negative likelihood ratio 0.1(0.1-0.3) 0.1(0.1-0.3) 0.1(0.0-0.4)

Diagnostic odds ratio 66.7(29.3-152.0) 95.2(34.0-267.0) 102.8(22.1-478.9)

CE-IVD cut-off

Number of patients in subgroup 506 - -

Sensitivity 84.5(74.3-91.1) - -

Specificity 87.1(83.7-90.0) - -

Positive predictive value 51.7(42.7-60.6) - -

Negative predictive value 97.2(95.0-98.4) - -

Positive likelihood ratio 6.6(5.0-8.5) - -

Negative likelihood ratio 0.2(0.1-0.3) - -

Diagnostic odds ratio 36.9(18.3-74.4) - -

Data are accuracy measures (95% CI), calculated according to the CE-IVD label for the majority cohort, the unanimous cohort, and the microbiologically confirmed cohort to diagnose bacterial infection. Also shown are the data calculated according to the alternative cut-off of 0.5 for the majority cohort. Accuracy measures were calculated from table 2, excluding equivocal results.

To assess the potential added value of the index test in correctly classifying bacterial and viral infections, we did a head-to-head comparison of the index test and CRP (cut-off 40 mg/mL) in a reclassification table stratified by the majority reference standard outcomes (Table 5). The index test significantly improved classification of patients with viral infection (8.6%; p<0.0001) but did not significantly improve in patients with bacterial infection (5%, p=0.321) compared with CRP. Similarly, the index test improved diagnostic classification for viral (6.3%, p=0.002) but not bacterial infection (5.4%, p=0.364) compared with procalcitonin (Table 5). The index test reduced the number of false positives from 67 to 34 (51%) compared with CRP and from 55 to 32 (58%) compared with procalcitonin (both p<0.0001). For comparison, diagnostic accuracy for different CRP and procalcitonin cut-offs are provided in Supplemental Table 4.

15873_CvHouten_BNW_v2.indd 76 26-11-18 12:42

Page 79: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

77

4

Validation of a host-protein based assay

Tabl

e 4.

Cas

es w

ith a

refe

renc

e st

anda

rd o

utco

me

of b

acte

rial

that

wer

e cl

assi

fied

by th

e in

dex

test

as v

iral (

fals

e ne

gativ

e).

Pt.

Inde

x te

st

scor

eCl

inic

al

synd

rom

eAg

e (m

)Se

x (F

/M)

Coun

try

(IL/N

L)

Max

imal

te

mpe

ratu

re

(C)

CRP

Hos

pita

lizat

ion

dura

tion

(d)

AB

Com

orbi

dity

Mic

robi

olog

yCl

inic

al d

etai

ls1*

0.00

2Ba

cter

aem

ia12

MIL

39.0

2.7

0Ye

sN

one

Bloo

d cu

lture

: Ki

ngel

la K

inga

e.;

Nas

al sw

ab:

ente

rovi

rus a

nd

boca

viru

s

Pres

enta

tion

with

rhin

orrh

oea,

fa

tigue

, ano

rexi

a an

d fe

ver;

by c

linic

al e

xam

inat

ion

wel

l ap

pear

ing,

rash

and

stom

atiti

s; no

t hos

pita

lized

20.

03Re

spira

tory

tr

act

infe

ctio

n

20F

NL

39.8

1832

Yes

Lung

ag

enes

is,

trea

chea

- br

onch

o-m

alac

ie,

vent

ricul

ar

sept

al d

efec

t

Nas

al sw

ab: R

SV;

bloo

d cu

lture

ne

gativ

e

Adm

itted

on

the

PICU

und

er

antib

iotic

pro

phyl

axes

with

co

-trim

oxaz

ole;

pre

sent

atio

n w

ith d

yspn

oea

and

coug

h;

no re

cent

surg

ery;

ill

appe

arin

g, ra

les a

nd c

ervi

cal

lym

phad

enop

athy

; CRP

m

axim

al 9

4; c

hest

x-ra

y:

cons

olid

atio

n pa

rahi

lar;

prol

onge

d ho

spita

lizat

ion

due

to a

bac

teria

l sup

er-in

fect

ion

30.

05Bo

rdet

ella

pe

rtus

sis

2M

NL

39.0

835

Yes

Non

eN

asop

hary

ngea

l sw

ab: B

orde

tella

pe

rtus

sis;

Nas

al

swab

: infl

uenz

a vi

rus a

nd

rhin

oviru

s

Pres

enta

tion

with

cou

gh,

sput

um, d

iarr

hoea

and

vo

miti

ng; i

ll ap

pear

ing,

rale

s, us

e of

acc

esso

ry m

uscl

es; n

o ch

est x

-ray

40.

07Pn

eum

onia

6M

IL39

.819

.26

Yes

Hyp

er-

reac

tive

airw

ay

and

give

n sa

lbut

amol

an

d flu

ticas

one

Nas

al sw

ab: R

SV;

bloo

d cu

lture

ne

gativ

e

Pres

enta

tion

with

cou

gh,

rhin

orrh

oea,

dys

pnoe

a an

d vo

miti

ng; c

repi

tatio

n an

d pr

olon

ged

expi

ratio

n du

ring

clin

ical

exa

min

atio

n;

cons

olid

atio

n on

the

ches

t x-

ray

15873_CvHouten_BNW_v2.indd 77 26-11-18 12:42

Page 80: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

78

Chapter 4

Tabl

e 4.

Cas

es w

ith a

refe

renc

e st

anda

rd o

utco

me

of b

acte

rial

that

wer

e cl

assi

fied

by th

e in

dex

test

as v

iral (

fals

e ne

gativ

e). (

Cont

inue

d)

Pt.

Inde

x te

st

scor

eCl

inic

al

synd

rom

eAg

e (m

)Se

x (F

/M)

Coun

try

(IL/N

L)

Max

imal

te

mpe

ratu

re

(C)

CRP

Hos

pita

lizat

ion

dura

tion

(d)

AB

Com

orbi

dity

Mic

robi

olog

yCl

inic

al d

etai

ls5

0.08

Pneu

mon

ia52

MN

L39

.913

3Ye

sEp

isodi

c dy

spno

ea

and

give

n sa

lbut

amol

Nas

al sw

ab: p

ara-

influ

enza

and

rh

inov

irus

Pres

enta

tion

with

dys

pnoe

a,

coug

h an

d fe

ver.

Ill a

ppea

ring,

ra

les,

whe

ezin

g, d

ecre

ased

br

eath

soun

ds, u

se o

f ac

cess

ory

mus

cles

, non

-ex

udat

e to

nsill

itis.

Max

imal

CR

P 82

. No

ches

t X-ra

y.

6*0.

10U

rinar

y tr

act

infe

ctio

n10

FIL

40.0

60.8

3Ye

sN

one

Urin

e cu

lture

: Es

cher

ichi

a co

li >1

0^5

colo

ny

form

ing

units

pe

r mL

Pres

enta

tion

with

feve

r an

d de

crea

sed

solid

inta

ke;

norm

al c

linic

al e

xam

inat

ion;

ul

tras

ound

of t

he k

idne

y sh

owed

hyd

rone

phro

sis;

sedi

men

t: hi

gh c

once

ntra

tion

of le

ukoc

ytes

; abs

ence

of

nitr

ite

70.

22O

titis

med

ia6

MN

L40

.861

2Ye

sN

one

Nas

al sw

ab:

rhin

oviru

sFe

ver,

coug

h an

d de

crea

sed

inta

ke. I

ll ap

pear

ing,

non

-ex

udat

e to

nsill

itis,

norm

al

otos

copy

. 8

0.23

Phar

yngi

tis40

MIL

38.5

8.8

3Ye

sN

one

Nas

opha

ryng

eal

cultu

re: g

roup

A

Stre

ptoc

occu

s; na

sal s

wab

: in

fluen

za v

irus

and

rhin

oviru

s

Pres

enta

tion

with

febr

ile

conv

ulsio

ns; m

acul

opap

ular

ra

sh a

nd p

hary

ngiti

s; CR

P m

axim

al 1

7.9

All

of t

hese

pat

ient

s re

ceiv

ed a

ntib

iotic

s. CR

P=C-

reac

tive

prot

ein.

IL=I

srae

l. N

L=N

ethe

rland

s. PI

CU=p

aedi

atric

inte

nsiv

e ca

re u

nit.

RSV=

resp

irato

ry s

yncy

tial v

irus.

*Pat

ient

s w

ith a

mic

robi

olog

ical

ly c

onfir

med

dia

gnos

is.

15873_CvHouten_BNW_v2.indd 78 26-11-18 12:42

Page 81: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

79

4

Validation of a host-protein based assay

Head-to-head comparison of the index test and CRP and procalcitonin in the unanimous cohort (Supplemental Table 5) showed even larger reclassification improvement for children with bacterial infection and similar results for children with viral infection compared with the majority cohort.

Table 5. Diagnostic reclassification by the index test compared with CRP or procalcitonin.

Index test outcome

Viral Bacterial Total

Bacterial infection outcome from reference standard

CRP (<40 μg/mL; n=60)

Viral 6 5 11

Bacterial 2 47 49

Total 8 52 60

p value of improved classification* p=0.321

Procalcitonin (<0.5 ng/mL; n=56)

Viral 5 6 11

Bacterial 3 42 45

Total 8 48 56

p value of improved classification* p=0.364

Viral infection outcome from reference standard

CRP (<40 μg/mL; n=383)

Viral 305 11 316

Bacterial 44 23 67

Total 349 34 383

p value of improved classification* p<0.0001

Procalcitonin (<0.5 ng/mL; n=363)

Viral 293 15 308

Bacterial 38 17 55

Total 331 32 363

p value of improved classification* p=0.002The index test classification was compared to the majority reference standard outcome. Patients with an index test score below 0.35 were classified as viral, between 0.35 and 0.65 as equivocal (n=63), and above 0.65 as bacterial. Head-to-head comparison of CRP or procalcitonin and the index test results. As a cut-off for CRP we used <40 μg/mL and for PCT we used <0.5 ng/mL. A PCT value could not be measured for all patients, therefore the total numbers of patients is lower when compared with the CRP reclassification table. *In patients with a bacterial infection as agreed by the majority of the expert panel, five patients were correctly reclassified from viral to bacterial infections using the index test instead of CRP and two patients were incorrectly reclassified from bacterial to viral infections. In patients with a viral infection this reclassification from bacterial to viral infections was 44 patients and 11 patients for the incorrect reclassification from viral to bacterial. Reclassification improvement was 5% for patients with bacterial infection (5 minus 2 of 60) and 8.6% for patients with a viral infection (44 minus 11 of 383). In patients with a bacterial infection as agreed by the majority of the expert panel, six patients were correctly reclassified from viral to bacterial infections using the index test instead of procalcitonin and three patients were incorrectly reclassified from bacterial to viral infections. In patients with a viral infection this reclassification from bacterial to viral infections was 38 patients and 15 patients for the incorrect reclassification from viral to bacterial, respectively. Reclassification improvement was 5.4% for patients with bacterial infection (6 minus 3 of 56) and 6.3% for patients with a viral infection (38 minus 15 of 363).

15873_CvHouten_BNW_v2.indd 79 26-11-18 12:42

Page 82: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

80

Chapter 4

We analysed data from children aged between 1 and 2 months (n=34) as a predefined secondary analysis (Figure 1). Six children were excluded because of unsuccessful blood sampling (four) or inconclusive panel diagnosis (two). All 28 children (22 viral infections, six bacterial infections) were classified correctly by the index test. For patients admitted to the intensive care unit (ten, 30% bacterial infection), the test accuracy to diagnose bacterial infection was lower when compared with overall study population (AUC 0.67, 95% CI 0.13–1.00), although no meaningful statistical comparison was possible. Five paediatric intensive-care unit patients (50%) needed mechanical ventilation. There were no significant differences in age (24 months vs 20 months), previous antibiotic use (n=4 [40%] vs n=77 [19%]), and mean duration of pre-existent symptoms (1.4 days vs 2.8 days) between patients admitted to the paediatric intensive-care unit and other patients. The diagnostic accuracy of the assay was similar in patients with antibiotic treatment compared with patients without antibiotic treatment at the moment of recruitment and for patients admitted to hospital compared with non-admitted patients (Supplemental Table 6). Subgroup analyses per clinical syndrome showed no major differences in index test performance (Supplemental Table 7).

D isc ussionIn this double-blind, prospective study we validated a host-protein based assay, combining three blood circulating proteins: CRP, TRAIL, and IP-10. We showed that the assay significantly improved diagnosis of bacterial infection in children aged 2 to 60 months presenting with lower respiratory tract infection or fever without source at the hospital compared with CRP and procalcitonin. In an exploratory analysis in children between 1 and 2 months of age, the assay also showed promising diagnostic accuracy.Biomarkers such as CRP and procalcitonin are used in routine practice to help physicians distinguish between bacterial and viral infections, but cannot be used as a solitary predictor.14,18 Clinical prediction models have often shown accurate diagnostic performance but most of them have not been validated or have a large number of variables, limiting their usefulness.5,14,19 Nijman and colleagues19 validated a model combining clinical variables and CRP to predict serious bacterial infection in febrile children on the hospital emergency department. They reported that CRP substantially improved risk estimates compared with estimates based on clinical signs and symptoms only.19 A review of various studies9 has assessed additional markers in the search for diagnostic tools that offer better discrimination. Engelmann and colleagues20 reported that myxovirus resistance protein A (MxA), a specific viral marker, discriminates bacterial and viral infections with an AUC of 0.89. Furthermore, when combining CRP with MxA the AUC increased to 0.94.20 Our study confirms the diagnostic value of combining CRP with a viral marker (in this case TRAIL).

15873_CvHouten_BNW_v2.indd 80 26-11-18 12:42

Page 83: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

81

4

Validation of a host-protein based assay

To compare the accuracy of the assay in diagnosing bacterial infections compared with CRP and procalcitonin we used the most commonly applied cut-offs (CRP 40 μg/mL and procalcitonin 0.5 ng/mL) for the reclassification, but also compared the results of the index test with other cut-off values (CRP, 20, 60, and 80 μg/mL, procalcitonin, 0.25 and 1.0 ng/mL). We showed that the diagnostic assay also outperformed both CRP and procalcitonin using these cut-offs. We did the power analysis for the validation of the assay and not to detect differences in AUC between the assay and CRP or procalcitonin as this was a secondary outcome.The high diagnostic value of the index test is founded on the distinct dynamics of TRAIL, CRP, and IP-10 during viral and bacterial infections. TRAIL is the first blood-borne protein to be identified as increasing in concentration during viral infection and decreasing during bacterial infection. The increase of TRAIL in viral patients could be related to TRAIL’s role in programmed cell death associated with viral infections.21,22 The mechanism for the reduction of TRAIL in bacterial infections warrants further study. The protein IP-10 is secreted by several cell types in response to interferon-γ23 and has a function in chemo attraction of monocytes and T cells.24 It also has a role in the response to chronic viral infections including hepatitis C viruses,25 HIV,26 and a broad range of acute viral infections.8,27

A strength of the present study is the double-blind design. To our knowledge, this is the first prospective validation study for a diagnostic assay differentiating between bacterial and viral infection that was double blinded. A second strength is the thorough nature of the reference standard: the expert panel approach described by Oved and colleagues8 was modified; we included 1 month follow-up data and expert panel membership was restricted to non-attending physicians.8 Another strength of this study is the heterogeneous population. The Dutch cohort included children with more severe disease than the Israeli cohort. We showed that the assay did well in these quite different populations.

Limitations of our study should also be discussed. First, the reference standard used in the present study has limitations as no gold standard is available. There are various approaches to assigning reference bacterial versus viral outcomes described in the literature.18–20,28,29 Clinical suspicion confirmed by microbiological results is an approach often used, but this method can be hampered by technical issues (e.g., culture contamination and uncertainty because of colonisation) and the resulting cohort tends to include more severely ill patients. In our study, this approach reduces the subgroup with bacterial infection for performance analysis and tends to increase the proportion of patients who are more easy to diagnose. The use of an expert panel diagnosis is an approach commonly applied in this area of research.8,10 It has the advantage of capturing

15873_CvHouten_BNW_v2.indd 81 26-11-18 12:42

Page 84: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

82

Chapter 4

a wider spectrum of illness severities in the resulting cohort—including difficult-to-diagnose cases—therefore, is more likely to be generalizable to clinical practice.30 We found a high level of agreement between experts within the panels (88% agreement).A second limitation of the present study is that we excluded children with severe comorbidity, with viral chronic infections (e.g., HIV and hepatitis B virus) and with immunodeficiency. Diez-Padrisa and colleagues31 showed that procalcitonin and CRP cannot differentiate viral from invasive bacterial pneumonia in children admitted to hospital with malaria parasites. Further validation studies in high-risk populations are required. We have already started a separate validation study in children with cancer. The low number of patients with distinct clinical syndromes is another limitation, and means that we are underpowered to establish the accuracy of the index test in these subgroups. The same holds for ten patients admitted to the intensive care unit, for which we noted lower diagnostic performance of the index test. Not all eligible children participated in this study because of logistical reasons, which could have introduced bias. Additionally, we did not record the exact time interval between presentation at the hospital and study recruitment. Therefore, we are unclear of the influence of this variable in test accuracy. We did not identify a clear association between the duration of symptoms and the accuracy of the assay.An additional limitation was that the expert panel was blinded to the index test score, but was provided with CRP values when tested for routine care, as this parameter is routinely used to help identify the cause of the infection.32 Although the assay was shown to be superior to CRP in both specificity and sensitivity, we cannot exclude that incorporation bias—in which part of the test being assessed is included in the reference standard and thus can lead to overestimating test accuracy -has resulted in some degree of overestimation of the test outcome.Finally, there were 71 patients for whom the expert panel could not assign a final diagnosis and accordingly were excluded from the analysis. Such inconclusive cases are inherent to studies without a gold standard and this was taken into account when calculating the sample size. Implementation studies are needed to investigate the clinical use of the assay in these cases. Although the test shows clinical promise, follow-up studies should also examine cost-effectiveness considerations since these would affect the test’s potential adoption.

Timely identification of bacterial infections in children remains challenging, leading to antibiotic overuse and underuse with its profound health and economic consequences. To address the need for better diagnostics, we have validated, in a double-blind, prospective, multicentre study, a host-based assay that combines novel and traditional blood-borne proteins for differentiatingbetween bacterial and viral infections. Our findings of the OPPORTUNITY study are

15873_CvHouten_BNW_v2.indd 82 26-11-18 12:42

Page 85: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

83

4

Validation of a host-protein based assay

promising, supporting the need for implementation research to examine the added clinical utility of ImmunoXpert to diagnose bacterial infection in clinical care for children with lower respiratory tract infection and fever without source presenting at the hospital.

15873_CvHouten_BNW_v2.indd 83 26-11-18 12:42

Page 86: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

84

Chapter 4

References1. Kronman MP, Zhou C, Mangione-Smith R. Bacterial prevalence and antimicrobial prescribing

trends for acute respiratory tract infections. Pediatrics 2014; 134: e956–65.

2. Chancey RJ, Jhaveri R. Fever without localizing signs in children: a review in the post-Hib and postpneumococcal era. Minerva Pediatr 2009; 61: 489–501.

3. Laxminarayan R, Duse A, Wattal C, et al. Antibiotic resistance—the need for global solutions. Lancet Infect Dis 2013; 13: 1057–98.

4. ECDC Technical Report: The bacterial challenge: time to react, Sept 2009. Available from: http://ecdc.europa.eu/en/publications/Publications/0909_TER_The_Bacterial_Challenge_Time_to_React.pdf (accessed 11-8-2016).

5. Craig JC, Williams GJ, Jones M, et al. The accuracy of clinical symptoms and signs for the diagnosis of serious bacterial infection in young febrile children: prospective cohort study of 15 781 febrile illnesses. BMJ 2010; 340: c1594.

6. Metersky ML, Sweeney TA, Getzow MB, Siddiqui F, Nsa W, Bratzler DW. Antibiotic timing and diagnostic uncertainty in Medicare patients with pneumonia: is it reasonable to expect all patients to receive antibiotics within 4 hours? Chest 2006; 130: 16–21.

7. Van Zuijlen DA, Schilder AG, Van Balen FA, Hoes AW. National differences in incidence of acute mastoiditis: relationship to prescribing patterns of antibiotics for acute otitis media? Pediatr Infect Dis J 2001; 20: 140–44.

8. Oved K, Cohen A, Boico O, et al. A novel host-proteome signature for distinguishing between acute bacterial and viral infections. PLoS One 2015; 10: e0120012.

9. Kapasi AJ, Dittrich S, González IJ, Rodwell TC. Host biomarkers for distinguishing bacterial from non-bacterial causes of acute febrile illness: a comprehensive review. PLoS One 2016; 11: e0160278.

10. Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med 2015; 373: 2005–14.

11. Bertens LC, Broekhuizen BD, Naaktgeboren CA, et al. Use of expert panels to define the reference standard in diagnostic research: a systematic review of published methods and reporting. PLoS Med 2013; 10: e1001531.

12. Rutjes AW, Reitsma JB, Coomarasamy A, Khan KS, Bossuyt PM. Evaluation of diagnostic tests when there is no gold standard. A review of methods. Health Technol Assess 2007; 11: iii, ix–51.

13. Pencina MJ, D’Agostino RB Sr, Steyerberg EW. Extensions of net reclassification improvement calculations to measure usefulness of new biomarkers. Stat Med 2011; 30: 11–21.

14. Lacour AG, Zamora SA, Gervaix A. A score identifying serious bacterial infections in children with fever without source. Pediatr Infect Dis J 2008; 27: 654–56.

15. Galetto-Lacour A, Zamora SA, Gervaix A. Bedside procalcitonin and C-reactive protein tests in children with fever without localizing signs of infection seen in a referral center. Pediatrics 2003; 112: 1054–60.

16. Korppi M, Remes S. Serum procalcitonin in pneumococcal pneumonia in children. Eur Respir J 2001; 17: 623–27.

17. Gervaix A, Galetto-Lacour A, Gueron T, et al. Usefulness of procalcitonin and C-reactive protein rapid tests for the management of children with urinary tract infection. Pediatr Infect Dis J 2001; 20: 507–11.

15873_CvHouten_BNW_v2.indd 84 26-11-18 12:42

Page 87: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

85

4

Validation of a host-protein based assay

18. Nijman RG, Moll HA, Smit FJ, et al. C-reactive protein, procalcitonin and the lab-score for detecting serious bacterial infections in febrile children at the emergency department: a prospective observational study. Pediatr Infect Dis J 2014; 33: e273–79.

19. Nijman RG, Vergouwe Y, Thompson M, et al. Clinical prediction model to aid emergency doctors managing febrile children at risk of serious bacterial infections: diagnostic study. BMJ 2013; 346: f1706.

20. Engelmann I, Dubos F, Lobert PE, et al. Diagnosis of viral infections using myxovirus resistance protein A (MxA). Pediatrics 2015; 135: e985–93.

21. Falschlehner C, Schaefer U, Walczak H. Following TRAIL’s path in the immune system. Immunology 2009; 127: 145–54.

22. Gyurkovska V, Ivanovska N. Distinct roles of TNF-related apoptosis-inducing ligand (TRAIL) in viral and bacterial infections: from pathogenesis to pathogen clearance. Inflamm Res 2016; 65: 427–37.

23. Luster AD, Unkeless JC, Ravetch JV. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature 1985; 315: 672–76.

24. Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD. IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. J Immunol 2002; 168: 3195–204.

25. Romero AI, Lagging M, Westin J, et al. Interferon (IFN)-gamma-inducible protein-10: association with histological results, viral kinetics, and outcome during treatment with pegylated IFN-alpha 2a and ribavirin for chronic hepatitis C virus infection. J Infect Dis 2006; 194: 895–903.

26. Falconer K, Askarieh G, Weis N, Hellstrand K, Alaeus A, Lagging M. IP-10 predicts the first phase decline of HCV RNA and overall viral response to therapy in patients co-infected with chronic hepatitis C virus infection and HIV. Scand J Infect Dis 2010; 42: 896–901.

27. van der Does Y, Tjikhoeri A, Ramakers C, Rood PP, van Gorp EC, Limper M. TRAIL and IP-10 as biomarkers of viral infections in the emergency department. J Infect 2016; 72: 761–63.

28. Chen HL, Hung CH, Tseng HI, Yang RC. Plasma IP-10 as a predictor of serious bacterial infection in infants less than 4 months of age. J Trop Pediatr 2009; 55: 103–08.

29. Tsalik EL, Henao R, Nichols M, et al. Host gene expression classifiers diagnose acute respiratory illness etiology. Sci Transl Med 2016; 8: 322ra11.

30. Reitsma JB, Rutjes AW, Khan KS, Coomarasamy A, Bossuyt PM. A review of solutions for diagnostic accuracy studies with an imperfect or missing reference standard. J Clin Epidemiol 2009; 62: 797–806.

31. Diez-Padrisa N, Bassat Q, Machevo S, et al. Procalcitonin and C-reactive protein for invasive bacterial pneumonia diagnosis among children in Mozambique, a malaria-endemic area. PLoS One 2010; 5: e13226.

32. NICE: Fever in under 5: assessment and initial management. Available from: https://www.nice.org.uk/guidance/cg160/resources/fever-in-under-5s-assessment-and-initial-management-35109685049029 (accessed 19-7- 2016).

15873_CvHouten_BNW_v2.indd 85 26-11-18 12:43

Page 88: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

86

Chapter 4

S upplementar y materialSupplemental Table 1. Baseline characteristics of excluded cases and comparison between NL and IL.

a.

Excluded cases n=63

Primary analysis cohort n=577 p-value

Age (months) (mean, SD) 27(24) 21(16) 0.094

Gender, male (n,%) 26(55) 324(56) 0.127

Maximal temperature (°C) (mean, SD) 39.4(0.8) 39.4(0.8) 0.918

Duration of symptoms (days) (mean, SD) 4.1(4.0) 2.8(1.7) 0.057

Hospital admission (n,%) 28(56) 316(55) 1.000

Hospitalization duration (days) (median, IQR) 4(3-5) 3(2-4) 0.153

Antibiotic treatment prescribed (n,%) 25(50) 224(39) 0.135

CRP (mg/L) at admission (median, IQR) 19.2( 7-59) 21( 7-52) 0.860

Disease severity

Need of mechanical ventilation (n,%) 0(0) 8(1) 1.000

Deaths (n,%) 0(0) 0(0) NA

Recruiting site 0.151

Secondary care centre (n,%) 56(89) 528(92)

Tertiary care centre (n,%) 7(11) 35(6)

PICU (n,%) 0(0) 14(2)

Clinical syndrome n=51 0.964

Bacteraemia/viraemia 0(0) 7(1)

CNS 1(2) 11(2)

FWS 13(25) 173(30)

GE 2(4) 18(3)

LRTI 11(22) 141(24)

URTI 16(31) 155(27)

UTI 2(4) 18(3)

Other 6(12) 54(9)

Positive nasopharyngeal culture/PCR

Group A streptococcus 5(2) 1(0.5) 4(5)

Streptococcus pneumoniae 1(0.5) 1(0.5) 0(0)

Haemophilus influenzae 1(0.5) 1(0.5) 0(0)

Mycoplasma pneumoniae 1(0.5) 0(0) 1(1)

Bordetella pertussis 1(0.5) 0(0) 1(1)

Respiratory syncytial virus A/B 19(7) 13(6) 6(8)

Adenovirus 5(2) 3(1) 2(5)

Influenza virus 5(2) 4(2) 1(1)

Rhinovirus 3(1) 1(0.5) 2(5)

Metapneumovirus 3(1) 3(1) 0(0)

Coronavirus 1(0.5) 1(0.5) 0(0)

Parainfluenza virus 1(0.5) 1(0.5) 0(0)

b. Total

n=232Viral infection

n=89Bacterial infection

n=143Study nasal swab PCR(performed on all patients)0 microorganism 112(48) 17(19) 95(66)

1 microorganism 115(50) 68(76) 47(33)

2 microorganisms 5(2) 4(5) 1(1)

Microorganism

Adenovirus 3(1) 2(2) 1(1)

Bocavirus 1(0.5) 0(0) 1(1)

Coronavirus 4(2) 1(1) 3(2)

Influenza virus A/B 52(22) 35(39) 17(12)

Metapneumovirus 6(3) 4(5) 2(1)

Parainfluenza virus 5(2) 4(5) 1(1)

Respiratory syncytial virus A/B 18(8) 14(16) 4(3)

Rhinovirus A/B/C 22(10) 16(18) 6(4)

Mycoplasma pneumoniae 11(5) 0(0) 11(8)

Bordetella pertussis 3(1) 0(0) 3(2)

Routine care

Positive blood culture/PCR

Streptococcus pneumoniae 5(2) 0(0) 5(3)

Moraxella osloensis 1(0.5) 0(0) 1(1)

15873_CvHouten_BNW_v2.indd 86 26-11-18 12:43

Page 89: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

87

4

Validation of a host-protein based assay

Supplemental Table 1. Baseline characteristics of excluded cases and comparison between NL and IL. (Continued)

b.

The Netherlands n=134

Israel n=443 p-value

Age (months) (mean, SD) 19(16) 22(17) 0.065

Gender, male (n,%) 80(60) 244(55) 0.372

Maximal temperature (°C) (mean, SD) 39.4(0.8) 39.4(0.8) 0.689

Duration of symptoms (days) (mean, SD) 2 (1.6) 3 (1.7) <0.0001

Hospital admission (n,%) 110(82) 206(47) <0.0001

Hospitalization duration (days) (median, IQR) 3(2-5) 3(2-4) 0.921

Antibiotic treatment prescribed (n,%) 70(52) 154(35) 0.0004

CRP (mg/L) at admission (median, IQR) 22.5(6-63) 20.3(7-49) 0.295

Disease severity

Need of mechanical ventilation (n,%) 8(6) 1(0) <0.0001

Deaths (n,%) 0(0) 0(0) NA

Recruiting site <0.0001

Secondary care centre (n,%) 86(64) 442(100)

Tertiary care centre (n,%) 35(26) 0(0)

PICU (n,%) 13(10) 1(0)

Clinical syndrome <0.0001

Bacteraemia/viraemia 4(3) 3(1)

CNS 5(4) 6(1)

FWS 32(24) 141(32)

GE 6(4) 12(3)

LRTI 14(10) 127(29)

URTI 50(37) 105(24)

UTI 11(8) 7(2)

Other 12(9) 42(9)

Clinical syndrome was based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. a. Excluded cases. Some baseline characteristics are not available for all patients. Given data are percentages of the available numbers. b. Comparison between the patients recruited in the Netherlands and Israel.CRP: C-reactive protein, PICU: paediatric intensive care units, CNS: central nervous system, FWS: fever without source, GE: gastro-enteritis, LRTI: lower respiratory tract infection, URTI: upper respiratory tract infection, UTI: urinary tract infection. NA = not applicable.

15873_CvHouten_BNW_v2.indd 87 26-11-18 12:43

Page 90: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

88

Chapter 4

Supplemental Table 2. Microbiology results.

Total n=577

Viral infection

n=435

Bacterial infection

n=71Inconclusive

n=71Study nasal swab PCR (performed on all patients*)

0 microorganism 104(18) 63(15) 22(31) 19(27)

1 microorganism 268(46) 204(47) 32(45) 32(45)

2 microorganisms 145(25) 115(26) 15(21) 15(21)

3 microorganisms 51(9) 45(10) 1(1) 5(7)

4 microorganisms 7(1) 6(1) 1(1) 0(0)

5 microorganisms 1(0) 1(0) 0(0) 0(0)

Microorganism

Adenovirus 91(16) 72(17) 5(7) 14(20)

Bocavirus 64(11) 48(11) 7(10) 9(13)

Coronavirus229E/NL63/OC43 50(9) 36(8) 7(10) 7(10)

Enterovirus 78(13) 69(16) 4(6) 5(7)

Influenza virus A/B 65(11) 55(13) 3(4) 7(10)

Metapneumovirus 58(10) 49(11) 3(4) 6(8)

Parainfluenza virus 42(7) 36(8) 6(8) 1(1)

Respiratory syncytial virus A/B 120(21) 101(23) 10(14) 9(13)

Rhinovirus A/B/C 169(29) 127(29) 23(32) 19(27)

Routine care

Positive blood culture 13(3) 4(1) 6(8) 3(4)

Staphylococcus coagulase negative 8(1) 4(1) 1(1.4) 3(4)

Streptococcus pneumonia 1(0.2) 0(0) 1(1.4) 0(0)

Haemophilus influenza 1(0.2) 0(0) 1(1.4) 0(0)

Kingella kingae 1(0.2) 0(0) 1(1.4) 0(0)

Group A streptococcus 1(0.2) 0(0) 1(1.4) 0(0)

Enterococcus faecalis 1(0.2) 0(0) 1(1.4) 0(0)

Positive blood PCR 4(0.8) 4(1) 0(0) 0(0)

Enterovirus 3(0.6) 3(0.7) 0(0) 0(0)

Parechovirus 1(0.2) 1(0.3) 0(0) 0(0)

Positive serology

Respiratory syncytial virus A/B 4(1) 3(0.7) 0(0) 1(1)

Positive CSF culture/PCR 5(1) 4(1) 1(1.4) 0(0)

Neisseria meningitides 1(0.2) 0(0) 1(1.4) 0(0)

Enterovirus 2(0.4) 2(0.5) 0(0) 0(0)

Human herpes virus type 6 1(0.2) 1(0.2) 0(0) 0(0)

Varicella zoster virus 1(0.2) 1(0.2) 0(0) 0(0)

15873_CvHouten_BNW_v2.indd 88 26-11-18 12:43

Page 91: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

89

4

Validation of a host-protein based assay

Supplemental Table 2. Microbiology results. (Continued)

Total n=577

Viral infection

n=435

Bacterial infection

n=71Inconclusive

n=71

Positive nasopharyngeal culture/PCR 41(7) 24(6) 8(11) 9(13)

Group A streptococcus 9(2) 1(0.2) 4(6) 4(6)

Streptococcus anginosus 1(0.2) 0(0) 0(0) 1(1)

Streptococcus dysgalactiae 1(0.2) 0(0) 0(0) 1(1)

Staphyloccocus aureus 1(0.2) 0(0) 0(0) 1(1)

Bordetella pertussis 2(0.4) 0(0) 1(1.4) 1(1)

Metapneumovirus 10(2) 7(2) 2(3) 1(1)

Respiratory syncytial virus A/B 5(1) 4(1) 1(1.4) 0(0)

Adenovirus 5(1) 5(1) 0(0) 0(0)

Enterovirus 3(0.5) 3(0.7) 0(0) 0(0)

Rhinovirus 1(0.2) 1(0.2) 0(0) 0(0)

Bocavirus 1(0.2) 1(0.2) 0(0) 0(0)

Influenza B virus 1(0.2) 1(0.2) 0(0) 0(0)

Parainfluenza virus 1(0.2) 1(0.2) 0(0) 0(0)

Positive antigen 32(6) 24(6) 5(7) 3(4)

Respiratory syncytial virus A/B 28(5) 23(5) 2(3) 3(4)

Influenza virus 2(0.4) 0(0) 2(3) 0(0)

Group A streptococcus 2(0.4) 1(0.2) 1(1.4) 0(0)

Positive sputum culture 5(1) 3(0.6) 2(3) 0(0)

Candida albicans 1(0.2) 1(0.2) 0(0) 0(0)

Haemophilus influenza 1(0.2) 0(0) 1(1.4) 0(0)

Streptococcus dysgalactiae 1(0.2) 1(0.2) 0(0) 0(0)

Staphyloccocus aureus 1(0.2) 1(0.2) 0(0) 0(0)

Rhinovirus 1(0.2) 0(0) 1(1.4) 0(0)

Positive stool culture/PCR 11(2) 8(1.6) 1(1.4) 2(3)

Enterovirus 4(1) 4(0.8) 0(0) 0(0)

Adenovirus 2(0.4) 1(0.2) 1(1.4) 0(0)

Parechovirus 1(0.2) 1(0.2) 0(0) 0(0)

Norovirus 1(0.2) 1(0.2) 0(0) 0(0)

Clostridium difficile 1(0.2) 1(0.2) 0(0) 0(0)

E. coli 1(0.2) 0(0) 0(0) 1(1)

Campylobacter 1(0.2) 0(0) 0(0) 1(1)

Positive urine culture 21(4) 1(0.2) 17(24) 3(4)

E.coli 17(3) 1(0.2)** 15(21) 1(1)

Enterococcus faecalis 2(0.4) 0(0) 2(3) 0(0)

15873_CvHouten_BNW_v2.indd 89 26-11-18 12:43

Page 92: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

90

Chapter 4

Supplemental Table 2. Microbiology results. (Continued)

Total n=577

Viral infection

n=435

Bacterial infection

n=71Inconclusive

n=71

Candida albicans 1(0.2) 0(0) 0(0) 1(1)

Pseudomonas A 1(0.2) 0(0) 0(0) 1(1)

Positive wound culture/PCR 4(1) 2(0.4) 2(3) 0(0)

Staphyloccocus aureus 1(0.2) 0(0) 1(1.4) 0(0)

Group A streptococcus 1(0.2) 0(0) 1(1.4) 0(0)

Staphylococcus coagulase negative 1(0.2) 1(0.2) 0(0) 0(0)

Enterovirus 1(0.2) 1(0.2) 0(0) 0(0)

Positive tympanocentesis culture

Haemophilus influenza 1(0.2) 0(0) 1(1.4) 0(0)

Positive perineum culture

Streptococcus pneumoniae 1(0.2) 0(0) 1(1.4) 0(0)

Positive eye culture

Haemophilus influenza 7(1) 6(1.2) 0(0) 1(1)

Table includes all patients eligible for primary analysis. Reference standard outcomes were based on majority agreement of the expert panel. Mixed infection was considered as bacterial.*For one patient (viral reference standard outcome) a study nasal swab was not available.**CRP=1, recovered without antibiotic treatment, clinical diagnosis was a gastro-enteritis, during admission interpreted as contamination.

Supplemental Table 3. Expert panel agreement and reference standard outcomes per country.

Totaln=577

Netherlandsn=134

Israeln=443

Expert panel agreement 506(88) 114(85) 392(89)

3 out of 3 406(71) 89(66) 317(72)

2 out of 3 100(17) 25(19) 75(17)

Inconclusive outcome 71(12) 20(15) 51(11)

No agreement 16(3) 6(5) 10(2)

Indeterminate* 55(9) 14(10) 41(9)

Outcome n=506 n=114 n=392

Bacterial 71(14) 26(23) 45(11)

Viral 435(86) 88(77) 347(89)

Table includes all patients eligible for primary analysis.*The majority of the expert panel diagnosed these cases as indeterminate.

15873_CvHouten_BNW_v2.indd 90 26-11-18 12:43

Page 93: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

91

4

Validation of a host-protein based assay

Supplemental Table 4. Diagnostic performance of the index test compared to CRP and PCT.

a.Accuracy measure

Index test n=443

CRP 20 n=443

CRP 40 n=443

CRP 60 n=443

CRP 80 n=443

Sensitivity 86.7( 75.8-93.1) 88.3( 77.8-94.2) 81.7( 70.1-89.4) 75.0( 62.8-84.2) 68.3( 55.8-78.7)

Specificity 91.1( 87.9-93.6) 62.7( 57.7-67.4) 82.5( 78.4-86.0) 93.2( 90.2-95.3) 96.9( 94.6-98.2)

PPV 60.5( 49.9-70.1) 27.0( 21.3-33.7) 42.2( 33.6-51.3) 63.4( 51.8-73.6) 77.4( 64.5-86.5)

NPV 97.8( 95.6-98.9) 97.2( 94.3-98.6) 96.6( 94.1-98.1) 96.0( 93.5-97.5) 95.1( 92.5-96.9)

LR + 9.8( 7.0-13.7) 2.4( 2.0-2.8) 4.7( 3.6-6.0) 11.0( 7.4-16.5) 21.8( 12.2-39.1)

LR - 0.1( 0.1-0.3) 0.2( 0.1-0.4) 0.2( 0.1-0.4) 0.3( 0.2-0.4) 0.3( 0.2-0.5)

DOR 66.7(29.3-152.0) 12.7( 5.6-28.7) 21.0( 10.4-42.5) 41.2( 20.3-83.5) 66.7(30.2-147.2)

b.Accuracy measure

Index test n=419

PCT 0.25 n=419

PCT 0.5 n=419

PCT 1.0 n=419

Sensitivity 85.7(74.3-92.6) 100(93.6-100) 80.4(68.2-88.7) 71.4(58.5-81.6)

Specificity 91.2(87.8-93.7) 2.2(1.1-4.3) 84.8(80.8-88.2) 90.1(86.6-92.8)

PPV 60.0(49.0-70.0) 13.6(10.6-17.3) 45.0(35.6-54.8) 52.6(41.6-63.5)

NPV 97.6(95.4-98.8) 100(67.6-100) 96.6(93.9-98.1) 95.3(92.6-97.1)

LR + 9.7(6.9-13.8) 1.0(1.0-1.0) 5.3(4.0-7.0) 7.2(5.1-10.2)

LR - 0.2(0.1-0.3) 0.0(0-NA) 0.2(0.1-0.4) 0.3(0.2-0.5)

DOR 62.1(27.0-142.6) NA 22.9(11.2-47.0) 22.7(11.6-44.6)

Diagnostic performance measures were calculated by comparing majority reference standard outcome to the index test classification (left column) and classification derived using different CRP cut-offs (a) or different PCT cut-offs, in the subset of patients where both the index test and PCT were measured (b). According to the current CE-IVD label, patients with an index test score below 0.35 were classified as viral, between 0.35 and 0.65 as equivocal, and above 0.65 as bacterial.Accuracy measures are given with 95% CI.CRP: C-reactive protein, PCT: procalcitonin, PPV: positive predicted value, NPV: negative predicted value, LR+: positive likelihood ratio, LR-: negative likelihood ratio, DOR: diagnostic odds ratio. NA = not applicable.

15873_CvHouten_BNW_v2.indd 91 26-11-18 12:43

Page 94: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

92

Chapter 4

Supplemental Table 5. Diagnostic reclassification by the index test compared to CRP and PCT in patients according to unanimous reference standard outcomes.The final diagnosis was based on the unanimous agreement of the expert panel. The index test classification was compared to the unanimous reference standard outcome. Patients with an index score below 0∙35 were classified as viral, between 0∙35 and 0∙65 as equivocal (n=52), and above 0∙65 as bacterial. a. Head-to-head comparison of CRP and the index test results. As a cut-off for CRP we used <40µg/ml. b. Head-to-head comparison of PCT and the index test results. As a cut-off for PCT we used <0∙5ng/ml. A PCT value could not be calculated for all patients, therefore the total numbers of patients is lower than in the CRP reclassification table.

a.Reference standard outcome:

Bacterial infection (n=41)Reference standard outcome :

Viral infection (n=313)Index test result Index test result

Viral Bacterial Total Viral Bacterial Total

CRP result(<40µg/ml)

Viral 4 5 9 261 8 269

Bacterial 1 31 32 30 14 44

Total 5 36 41 291 22 313

* Among patients with a bacterial infection as determined by an unanimous expert panel, 5 patients were correctly reclassified from viral to bacterial using the index test instead of CRP and 1 patient was incorrectly reclassified from bacterial to viral. For patients with a viral infection this was 30 and 8, respectively. Reclassification improvement was 9.8% for patients with bacterial infection (5-1 of 41; p=0.142) and 7.0% for patients with a viral infection (30-8 of 313; p<0.0001).

b.Reference standard outcome:

Bacterial infection (n=41)Reference standard outcome:

Viral infection (n=296)Index test result Index test result

Viral Bacterial Total Viral Bacterial TotalPCT result(<0.50g/ml)

Viral 4 4 8 245 11 256

Bacterial 1 32 33 30 10 40

Total 5 36 41 275 21 296

* Among patients with a bacterial infection as determined by an unanimous expert panel, 4 patients were correctly reclassified from viral to bacterial using the index test instead of PCT and 1 patient was incorrectly reclassified from bacterial to viral. For patients with a viral infection this was 30 and 11, respectively. Reclassification improvement was 7.3% for patients with bacterial infection (4-1 of 41; p=0.251) and 6∙4% for patients with a viral infection (30-11 of 296; p=0.004).

15873_CvHouten_BNW_v2.indd 92 26-11-18 12:43

Page 95: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

93

4

Validation of a host-protein based assay

Supplemental Table 6. Diagnostic performance of the index test in patients treated with and without antibiotic treatment at the moment of recruitment and admitted and non-admitted patients.

Accuracy measure

Antibiotictreatment at the

moment of recruitmentn=81

No antibiotic treatment at the

moment of recruitmentn=341

Admittedpatientsn=237

Non admittedpatientsn=205

Sensitivity 93.3(70.2-99.7) 83.3(69.4-91.7) 85.7(73.3-92.9) 90.9(62.3-99.5)

Specificity 92.4(83.5-96.7) 90.6(86.8-93.4) 88.3(82.9-92.1) 94.3(90.1-96.8)

PPV 73.7(51.2-88.2) 55.6(43.3-67.2) 65.6(53.4-76.1) 47.6(28.3-67.6)

NPV 98.4(91.4-99.9) 97.5(94.9-98.8) 96.0(91.9-98.0) 99.5(97.0-100)

LR + 12.3( 5.2-28.9) 8.9( 6.1-13.0) 7.3( 4.9-11.0) 16.0( 8.8-29.3)

LR - 0.1( 0.0-0.5) 0.2( 0.1-0.4) 0.2( 0.1-0.3) 0.1( 0-0.6)

DOR 170.8(18.5-1579.2) 48.4(19.7-119.0) 45.3(18.1-113.1) 166.4(19.5-1419.4)

Diagnostic performance measures were calculated by comparing majority reference standard outcome to the index test classification. According to the current CE-IVD label, patients with an index test score below 0.35 were classified as viral, between 0.35 and 0.65 as equivocal, and above 0.65 as bacterial. Accuracy measures are given with 95% CI.PPV: positive predicted value, NPV: negative predicted value.

15873_CvHouten_BNW_v2.indd 93 26-11-18 12:43

Page 96: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

94

Chapter 4

Supp

lem

enta

l Tab

le 7

. Dia

gnos

tic

perf

orm

ance

of t

he in

dex

test

per

clin

ical

syn

drom

e.

Acc

urac

y m

easu

re

Bact

erae

mia

/vir

aem

ia

n=6

CNS

n=9

FWS

n=13

9G

E n

= 15

LRTI

n=10

8U

RTI

n= 1

14U

TIn=

15O

ther

n=37

Bact

eria

l cas

es (

n)3

13

021

815

9

Sens

itivi

ty

66.7

(20.

8-98

.3)

100(

5.1-

100)

100(

43.9

-100

)N

A90

.5(7

1.1-

97.3

)50

.0 (2

1.5-

78.5

)93

.3(7

0.2-

99.7

)10

0(70

.1-1

00)

Spec

ifici

ty10

0(43

.9-1

00)

75.0

(40.

9-92

.9)

92.6

(87.

0-96

.0)

100(

79.6

-100

)92

.0(8

4.3-

96.0

)88

.7(8

1.2-

93.4

)N

A89

.3(7

2.8-

96.3

)

PPV

100(

34.2

-100

)33

.3(1

.7-7

9.2)

23.1

(8.2

-50.

3)N

A73

.1(5

3.9-

86.3

)25

.0(1

0.2-

49.5

)N

A75

.0(4

6.8-

91.1

)

NPV

75(3

0.1-

98.7

)10

0(61

.0-1

00)

100(

97.0

-100

)N

A97

.6(9

1.5-

99.3

)95

.9(9

0.0-

98.4

)N

A10

0(86

.7-1

00)

LR +

NA

4.0(

1.2-

13.3

)13

.6(7

.5-2

4.7)

NA

11.2

(5.5

-23.

2)4.

4(1.

8-10

.6)

NA

9.3(

3.2-

27.2

)

LR -

0.3(

0.1-

1.7)

0.0(

0-N

A)

0.0(

0-N

A)

NA

0.1(

0.0-

0.4)

0.6(

0.3-

1.1)

NA

0.0(

0-N

A)

DO

RN

AN

AN

AN

A10

8.6

(20.

9-56

4.9)

7.8

(1.7

-35.

5)N

AN

A

Dia

gnos

tic p

erfo

rman

ce w

as e

valu

ated

by

com

parin

g th

e in

dex

test

cla

ssifi

catio

n to

the

maj

ority

ref

eren

ce s

tand

ard

outc

ome.

Pat

ient

s w

ith a

n in

dex

test

sco

re

belo

w 0

.35

wer

e cl

assi

fied

as v

iral,

betw

een

0.35

and

0.6

5 as

equ

ivoc

al, a

nd a

bove

0.6

5 as

bac

teria

l. Cl

inic

al s

yndr

ome

was

bas

ed o

n th

e di

agno

sis

of th

e at

tend

ing

phys

icia

n at

dis

char

ge f

rom

the

hos

pita

l. LR

TI in

clud

ed p

neum

onia

and

bro

nchi

oliti

s; U

RTI i

nclu

ded

lary

ngiti

s, ph

aryn

gitis

, otit

is m

edia

, sin

usiti

s an

d to

nsill

itis.

Accu

racy

mea

sure

s ar

e gi

ven

with

95%

CI.

PPV:

pos

itive

pre

dict

ed v

alue

, NPV

: neg

ativ

e pr

edic

ted

valu

e, L

R+: p

ositi

ve li

kelih

ood

ratio

, LR-

: neg

ativ

e lik

elih

ood

ratio

, DO

R: d

iagn

ostic

odd

s ra

tio. C

NS:

cen

tral

ne

rvou

s sy

stem

, FW

S: f

ever

with

out

sour

ce, G

E: g

astr

o-en

terit

is, L

RTI:

low

er r

espi

rato

ry t

ract

infe

ctio

n, U

RTI:

uppe

r re

spira

tory

tra

ct in

fect

ion,

UTI

: urin

ary

trac

t in

fect

ion.

NA

= n

ot a

pplic

able

.

15873_CvHouten_BNW_v2.indd 94 26-11-18 12:43

Page 97: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

95

4

Validation of a host-protein based assay

Equivocal

Viral/Other Bacterial

0 10 20 35 65 80 90 100

Supplemental Figure 1. The index test score.Classifi cation based on the index test score. According to the current CE-IVD label, patients with an index test score below 0.35 were classifi ed as viral, between 0.35 and 0.65 as equivocal, and above 0.65 as bacterial.

Entire cohort

Majority cohort (>=2/3)

Unanimous cohort (3/3)

Microbiologically confirmed cohort

Supplemental Figure 2. Types of diagnostic reference standards.Patients with majority agreement of the expert panel (majority cohort) are used as the predefi ned preferred reference standard. The accuracy of the index test is also calculated for patients with unanimous agreement (unanimous cohort) and for a sub-cohort of patients within the unanimous cohort with a microbiologically confi rmed diagnosis.

15873_CvHouten_BNW_v2.indd 95 26-11-18 12:43

Page 98: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

96

Chapter 4

a.

Bacterial Viral Non-infectious0

50

100

150

200

250

300

ug/ml

CRP

>350

b.

15873_CvHouten_BNW_v2.indd 96 26-11-18 12:43

Page 99: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

97

4

Validation of a host-protein based assay

c.

Supplemental Figure 3. Expression of CRP, TRAIL and IP-10.Box plots for CRP (a), TRAIL (b) and IP-10 (c) concentrations measured among patients with bacterial infections (n=71), patients with viral infections (n=435) and non-infectious controls (n=103). The analysis was performed on patients with a reference standard outcome based on majority agreement of the expert panel. The red bar corresponds to the group mean with standard deviation. TRAIL: TNF-related apoptosis inducing ligand, IP-10: interferon gamma induced protein-10, CRP: C-reactive protein.

15873_CvHouten_BNW_v2.indd 97 26-11-18 12:43

Page 100: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

98

Chapter 4

Supp

lem

enta

l Fig

ure

4. In

dex

test

sco

re d

istr

ibut

ion

per r

efer

ence

sta

ndar

d ou

tcom

e.

Una

nim

ous

agre

emen

t bac

teria

l/vira

l: al

l pan

el m

embe

rs d

iagn

osed

thes

e pa

tient

s as

bac

teria

l or v

iral.

Maj

ority

agr

eem

ent b

acte

rial/v

iral:

at le

ast t

wo

out o

f thr

ee p

anel

mem

bers

dia

gnos

ed th

ese

patie

nts

as b

acte

rial o

r vira

l. In

conc

lusi

ve: c

ases

with

inde

term

inat

e pa

nel d

iagn

osis

and

cas

es w

ithou

t pan

el a

gree

men

t. Th

e re

d ba

r cor

resp

onds

to th

e gr

oup

mea

n w

ith s

tand

ard

devi

atio

n.

15873_CvHouten_BNW_v2.indd 98 26-11-18 12:43

Page 101: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 99 26-11-18 12:43

Page 102: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

5

15873_CvHouten_BNW_v2.indd 100 26-11-18 12:43

Page 103: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

5Optimizing prediction of serious bacterial infections by a host-protein assay

CB van Houten, JS van de Maat, CA Naaktgeboren, LJ Bont and R Oostenbrink

Submitted

15873_CvHouten_BNW_v2.indd 101 26-11-18 12:43

Page 104: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

102

Chapter 5

Abstrac tBackgroundTo determine whether updating a diagnostic prediction model by adding a combination assay (tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), interferon gamma induced protein-10 (IP-10), and C-reactive protein (CRP)) can accurately identify children with pneumonia or other serious bacterial infection (SBI).

MethodsObservational double-blind diagnostic study in hospitals in Israel and the Netherlands. A total of 591 children, aged 1 to 60 months, presenting with lower respiratory tract infections or fever without source were recruited. Ninety-two of them had SBI. The original Feverkidstool, a polytomous logistic regression model including clinical variables and CRP, was recalibrated and thereafter updated by using the assay. Main outcome measures were pneumonia, other SBI or no SBI.

Results The recalibrated original Feverkidstool discriminated well between SBI and viral infections, with a c-statistic for pneumonia of 0.84 (95% CI: 0.77 to 0.92) and 0.82 (95% CI: 0.77 to 0.86) for other SBI. The discriminatory ability increased when CRP was replaced by the combination assay; c-statistic for pneumonia increased to 0.89 (95% CI: 0.82 to 0.96) and for other SBI to 0.91 (95% CI: 0.87 to 0.94). This updated Feverkidstool improved diagnosis of SBI mainly in children with low-moderate risk estimates of SBI.

Conclusion We improved the diagnostic accuracy of the Feverkidstool by replacing CRP with a combination assay to predict pneumonia or other SBI in febrile children. The updated Feverkidstool has the largest potential to rule out bacterial causes and thus to decrease unnecessary antibiotic prescription in children with low to moderate predicted risk of SBI.

15873_CvHouten_BNW_v2.indd 102 26-11-18 12:43

Page 105: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

103

5

Optimizing prediction of bacterial infections

Introduc tionSuspicion of infectious disease is one of the most common cause of paediatric emergency department (ED) visits.1 The proportion of bacterial infections in children with fever without source (FWS) and acute respiratory tract infections (RTI) is however low (respectively 0.02–13% and 26–28%).2-4 Next, identifying patients benefitting from antibiotic treatment remains a major diagnostic challenge. Consequentially, children with acute RTI receive antibiotics almost twice as often as the estimated prevalence.2,

5, 6 Antibiotic overuse is associated with increased antibiotic resistance, causing 25.000 deaths in Europe annually.7, 8 This underlines the need for new diagnostics to better differentiate between viral and bacterial infections. Therefore, several prediction models have been developed.9, 10 The Feverkidstool, a clinical prediction model including both clinical parameters and C-reactive protein (CRP), is considered a validated tool for supporting clinical decision-making on e.g. whether or not to start antibiotics.11-13 However, further improvement of this diagnostic tool is warranted as it does not provide an accurate diagnosis for all patients. We recently showed that a novel blood assay, combining concentrations of CRP with tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and interferon gamma induced protein-10 (IP-10), could diagnose bacterial infections more accurate compared to CRP alone.14-16 The aim of this study is to investigate whether updating the Feverkidstool by replacing CRP with the combination assay can improve the diagnosis of SBI in preschool children.

MethodsThe current study builds on the analysis from the prospective observational Opportunity study at hospitals in the Netherlands and Israel.14 In short, this study included clinical data, a host-protein based assay, nasal swab PCR and 28-day follow-up data from children aged 1 to 60 months with lower RTI or FWS. A total of 777 children were recruited. Because the Feverkidstool was developed for febrile patients presenting at the ED, patients from the intensive care unit (n=16) and non-infectious control patients (n=103) were excluded.14 In 67 children the assay was not measured for various reasons (Supplemental Figure 1). The Opportunity study was approved by the ethics committees in the participating countries. This manuscript follows the TRIPOD reporting guidelines.

Prediction modelThe Feverkidstool is a polytomous prediction model that predicts the risk of pneumonia or other SBI, based on the following variables: age, body temperature, heart rate, respiratory rate, oxygen saturation, ill-appearance, peripheral capillary refill, chest wall retractions and CRP (definitions presented in the Statistical Analysis Plan). Peripheral

15873_CvHouten_BNW_v2.indd 103 26-11-18 12:43

Page 106: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

104

Chapter 5

capillary refill was not recorded in the Opportunity study, therefore these systematic missing values were replaced by the mean prevalence of prolonged capillary refill in the initial Feverkidstool cohort (=0.039).

Combined host-protein based assayThe assay is currently ELISA based (a point of care test is being developed) and combines the concentrations of TRAIL, IP-10 and CRP using a predetermined logistic regression formula to compute the likelihood score for a bacterial infection.14, 15 The assay was performed on anonymous serum samples in absence of any patient-related information.

Reference standard diagnosisAs part of the Opportunity study, an expert panel assigned one of the following aetiologies to each patient: bacterial infection, viral infection, mixed infection (i.e., bacterial and viral co-infection) or indeterminate.14 The panel based their diagnosis on all available clinical and laboratory information, including a 28-day follow-up assessment. Panel members were blinded for the decisions of their peers and for the assay and Feverkidstool results. Patients with a bacterial or mixed reference standard diagnosis were divided into pneumonia or other SBI (e.g. meningitis, urinary tract infections, bacteraemia) based on the diagnosis at hospital discharge.

Statistical analysisGeneral approach To find out whether the prediction of SBI would improve when the original Feverkidstool was updated with the assay, we compared the diagnostic accuracy of both models (hereafter called original and updated Feverkidstool). Because one element of the assay, CRP, was already a predictor in the original Feverkidstool, we essentially removed CRP from the updated Feverkidstool by assigning the median CRP value for all participants. P<0.05 was considered statistically significant. Statistical analyses were performed in SPSS version 21.0 for Windows and R version 3.2.2.

Model development and performance First, we recalibrated the original Feverkidstool on our data using logistic regression. Then, we updated the Feverkidstool by replacing CRP with the combination assay. The discriminative ability of the recalibrated original and updated model was expressed using pairwise c-statistics.17

15873_CvHouten_BNW_v2.indd 104 26-11-18 12:43

Page 107: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

105

5

Optimizing prediction of bacterial infections

Predicted risk thresholds A decision curve analysis (DCA) was performed to assess the relative harm of false positives and false negatives per probability threshold and helped to interpret the differences between the models along the wide range of predicted probabilities.18 For a set of predefined risk thresholds we calculated sensitivity, specificity, positive and negative predictive value, positive and negative likelihood ratio (LR+, LR-) for the original and updated models.

Reclassification To assess the potential added value of the updated Feverkidstool in correctly classifying SBI and viral infections using defined thresholds, we did a head-to-head comparison for the updated Feverkidstool and the recalibrated original Feverkidstool in a reclassification table.19

Missing values Multiple imputation techniques enabled analysing all available data. Missing Feverkidstool variables were therefore imputed 10 times using the MICE algorithm, R statistical software.20 Variables used in the imputation model are presented in the Statistical Analyses Plan. Inconclusive diagnoses (n=71), were also imputed, using the variables selected by the MICE algorithm. Analyses were performed separately in the 10 imputed data sets and combined using Rubin’s rules.21 A sensitivity analysis was performed by leaving out patients with an inconclusive diagnosis.

ResultsPopulation characteristics A total of 591 patients were available for analyses; 30 pneumonia, 66 other SBI and 495 viral infections (Supplemental Figure 1). Children with pneumonia were older (median age 2 years) than children with other SBI or with viral infections (median age 1.3 years) and children with pneumonia or other SBI were hospitalized more often than children with viral infections, respectively 73% and 77% versus 52% (Table 1). Children with an inconclusive reference standard diagnosis differed from children with a conclusive diagnosis on age, biomarker values and antibiotic prescription (Supplemental Table 1).

Model performance The recalibrated original Feverkidstool discriminate well between pneumonia and other infections (c-statistic 0.84, 95% CI: 0.77-0.92), and between other SBI and other infections (c-statistic 0.82, 95% CI 0.77-0.86) (Supplemental Figure 2). This performance

15873_CvHouten_BNW_v2.indd 105 26-11-18 12:43

Page 108: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

106

Chapter 5

Table 1. Characteristics of patients included in the primary analysis.Pneumonia

(n=30)Other SBI

(n=66)Viral

(n=495)Predictor variables

Age (years) 2.0[1.1-3.4] 1.3[0.7-2.8] 1.3[0.6-2.3]

Gender, male 19(63%) 32(49%) 280(57%)

Duration of fever (days) 3[2-5] 2[1-4] 2[1-4]

Temperature (°C) 38.6[38.2-39.8] 38.7[37.8-39.4] 38.5[37.6-39.2]

n=30(100%) n=66(100%) n=493(99%)

Respiratory rate 50[34-70] 40[31-52] 38[30-52]

n=15(50%) n=31(47%) n=252(51%)

Tachypnea 12(80%) 17(55%) 130(52%)

Heart rate 160(24) 152(27) 151(24)

n=29(97%) n=57(86%) n=453(92%)

Tachycardia 21(72%) 29(51%) 232(51%)

Oxygen saturation (%O2) 98[97-99] 99[97-100] 98[96-100]

n=28(93%) n=48(73%) n=417(84%)

Desaturation (<94%O2) 1(4%) 0(0%) 24(6%)

Chest wall retractions 6(20%) 4(6%) 60(12%)

n=30(100%) n=63(95%) n=484(98%)

Ill appearance 13(43%) 25(38%) 141(29%)

C-reactive protein (mg/l) 176[72-224] 102[55-151] 15[5-36]

Assay score 98[76-100] 88[68-98] 4[1-26]

Other variables

Hospital admission 22(73%) 51(77%) 255(52%)

Hospitalization duration (days) 4[3-6] 4[3-5] 3[2-4]

Antibiotic treatment prescribed 30(100%) 63(96%) 140(29%)

Recruiting site

Secondary care centre 27(90%) 63(96%) 463(94%)

Tertiary care centre 3(10%) 3(4%) 32(6%)

Focus of infection

Central nervous system 0(0%) 0(0%) 9(2%)

Gastrointestinal tract 0(0%) 1(2%) 19(4%)

Other 3(10%) 11(17%) 39(8%)

Respiratory tract 26(87%) 18(27%) 250(50%)

Systemic 0(0%) 2(3%) 9(2%)

Unknown 1(3%) 10(15%) 169(34%)

Urinary tract 0(0%) 24(36%) 0(0%)Data are presented as n (%), median [IQR] or mean (SD). This table includes imputed reference standard diagnoses, data are based on 1 of the 10 imputed datasets. If data were not available for all patients, the total number of available data are noted. Clinical syndrome was based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis.

15873_CvHouten_BNW_v2.indd 106 26-11-18 12:43

Page 109: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

107

5

Optimizing prediction of bacterial infections

is similar to numbers observed in previous Feverkidstool validations.11, 12 After updating the Feverkidstool by replacing CRP with the assay, discrimination between bacterial and other infections improved, reflected by an improved c-statistic for pneumonia to 0.89 (95% CI: 0.82 to 0.96), and for other SBI to 0.91 (95% CI: 0.87 to 0.94) (Supplemental Figure 2). A sensitivity analysis of the cohort without imputed reference standard diagnoses showed similar results, with improved prediction of pneumonia (increased c-statistic from 0.87 (95% CI: 0.79 to 0.95) to 0.91 (95% CI: 0.83 to 0.98)) and an improved prediction of other SBI (from 0.82 (95% CI: 0.76 to 0.88) to 0.91 (95% CI: 0.86 to 0.95)).

Predicted risk thresholds The decision curve analysis shows the net benefit of starting antibiotics using predictions of the updated Feverkidstool instead of the original Feverkidstool, depending on the choice of probability threshold. Children with low-moderate predicted risks ≤40% for pneumonia or other SBI had most benefit from the updated Feverkidstool (Figure 1). Table 2 gives more detailed insight on effects of the updated model in diagnostic value using several thresholds. Using a rule of thumb of LR+ of 5 and LR- of 0.217, thresholds of 10% and 2.5% using the updated model seem better applicable for ruling in and out of both pneumonia and other SBI.

0.0 0.2 0.4 0.6 0.8 1.0

−0.2

0.0

0.2

0.4

0.6

0.8

1.0

Sta

ndar

dize

d N

et B

enef

it

Original FKT Updated FKTAllNone

Predicted Risk Threshold

Pneumonia

0.0 0.2 0.4 0.6 0.8 1.0

−0.2

0.0

0.2

0.4

0.6

0.8

1.0

Sta

ndar

dize

d N

et B

enef

it

FKT Updated FKTAllNone

Predicted Risk Threshold

Other Serious Bacterial Infection

Figure 1. Decision curve analysis.Decision curve analysis with the net benefit of starting antibiotics to none of the patients (black line), to all patients (grey line), the original Feverkidstool (red line), and the updated Feverkidstool (blue line), depending on the choice of probability threshold for starting antibiotics for pneumonia (a) and other SBI (b). FKT; Feverkidstool.

15873_CvHouten_BNW_v2.indd 107 26-11-18 12:43

Page 110: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

108

Chapter 5

Tabl

e 2.

Dia

gnos

tic

perf

orm

ance

of p

redi

ctio

n m

odel

s in

clud

ing

clin

ical

sig

ns a

nd s

ympt

oms

and

eith

er C

-rea

ctiv

e pr

otei

n or

the

assa

y.

Sens

itiv

ity

(95%

CI)

Spec

ifici

ty (9

5% C

I)PP

V (9

5% C

I)N

PV (

95%

CI)

LR+

(95%

CI)

LR-

(95%

CI)

Orig

inal

Fev

erki

dsto

ol

Pne

umon

ia

Ri

sk o

f ≥ 2

.5%

0.85

(0.7

7-0.

94)

0.58

(0.5

4-0.

63)

0.10

(0.0

6-0.

14)

0.99

(0.9

8-0.

99)

2.06

(1.3

9-3.

07)

0.25

(0.0

8-0.

76)

Ri

sk o

f ≥ 5

%0.

81(0

.71-

0.91

)0.

76(0

.73-

0.79

)0.

15(0

.09-

0.22

)0.

99(0

.98-

0.99

)3.

41(2

.23-

5.20

)0.

24(0

.10-

0.62

)

Ri

sk o

f ≥ 1

0%0.

62(0

.47-

0.78

)0.

89(0

.87-

0.92

)0.

24(0

.13-

0.34

)0.

98(0

.97-

0.99

)5.

83(3

.90-

8.71

)0.

42(0

.25-

0.69

)

Ri

sk o

f ≥ 1

5%0.

48(0

.31-

0.65

)0.

93(0

.91-

0.95

)0.

27(0

.14-

0.40

)0.

97(0

.96-

0.98

)6.

80(3

.87-

11.9

7)0.

56(0

.37-

0.84

)

Ri

sk o

f ≥ 2

0%0.

40(0

.23-

0.58

)0.

95(0

.94-

0.97

)0.

31(0

.16-

0.47

)0.

97(0

.96-

0.98

)8.

52(4

.57-

15.9

0)0.

63(0

.45-

0.87

)

Ri

sk o

f ≥ 3

0%0.

27(0

.10-

0.45

)0.

99(0

.98-

1.00

)0.

60(0

.39-

0.81

)0.

96(0

.95-

0.98

)28

.09(

8.16

-96.

67)

0.73

(0.5

7-0.

94)

Oth

er S

BI

Ri

sk o

f ≥ 2

.5%

0.98

(0.9

7-1.

00)

0.19

(0.1

5-0.

22)

0.13

(0.1

0-0.

16)

0.99

(0.9

8-1.

00)

1.21

(1.1

3-1.

30)

0.09

(0.0

1-0.

65)

Ri

sk o

f ≥ 5

%0.

96(0

.94-

0.99

)0.

41(0

.37-

0.46

)0.

17(0

.13-

0.21

)0.

99(0

.98-

1.00

)1.

64(1

.48-

1.83

)0.

08(0

.02-

0.43

)

Ri

sk o

f ≥ 1

0%0.

85(0

.78-

0.92

)0.

70(0

.67-

0.74

)0.

26(0

.20-

0.33

)0.

98(0

.96-

0.99

)2.

87(2

.37-

3.48

)0.

21(0

.10-

0.42

)

Ri

sk o

f ≥ 1

5%0.

54(0

.42-

0.66

)0.

82(0

.79-

0.85

)0.

27(0

.19-

0.36

)0.

94(0

.92-

0.95

)3.

03(2

.24-

(4.1

2)0.

56(0

.41-

0.76

)

Ri

sk o

f ≥ 2

0%0.

38(0

.26-

0.50

)0.

89(0

.86-

0.91

)0.

30(0

.19-

0.40

)0.

92(0

.90-

0.94

)3.

37(2

.23-

5.09

)0.

70(0

.57-

0.86

)

Ri

sk o

f ≥ 3

0%0.

25(0

.13-

0.36

)0.

94(0

.92-

0.95

)0.

32(0

.18-

0.46

)0.

91(0

.89-

0.93

)3.

82(2

.15-

6.77

)0.

81(0

.69-

0.93

)

15873_CvHouten_BNW_v2.indd 108 26-11-18 12:43

Page 111: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

109

5

Optimizing prediction of bacterial infections

Tabl

e 2.

Dia

gnos

tic

perf

orm

ance

of p

redi

ctio

n m

odel

s in

clud

ing

clin

ical

sig

ns a

nd s

ympt

oms

and

eith

er C

-rea

ctiv

e pr

otei

n or

the

assa

y.

(Con

tinue

d)

Sens

itiv

ity

(95%

CI)

Spec

ifici

ty (9

5% C

I)PP

V (9

5% C

I)N

PV (

95%

CI)

LR+

(95%

CI)

LR-

(95%

CI)

Upd

ated

Fev

erki

dsto

ol

Pne

umon

ia

Ri

sk o

f ≥ 2

.5%

0.88

(0.8

0-0.

95)

0.75

(0.7

1-0.

78)

0.16

(0.0

9-0.

22)

0.99

(0.9

9-1.

00)

3.48

(2.3

9-5.

07)

0.16

(0.0

5-0.

49)

Ri

sk o

f ≥ 5

%0.

85(0

.76-

0.94

)0.

83(0

.80-

0.86

)0.

21(0

.13-

0.29

)0.

99(0

.99-

1.00

)4.

93(3

.55-

6.84

)0.

18(0

.07-

0.49

)

Ri

sk o

f ≥ 1

0%0.

76(0

.64-

0.88

)0.

89(0

.86-

0.91

)0.

27(0

.17-

0.37

)0.

99(0

.98-

0.99

)6.

84(4

.49-

10.4

1)0.

27(0

.14-

0.53

)

Ri

sk o

f ≥ 1

5%0.

72(0

.60-

0.85

)0.

92(0

.90-

0.94

)0.

33(0

.21-

0.45

)0.

98(0

.98-

0.99

)9.

27(5

.47-

15.7

2)0.

30(0

.15-

0.58

)

Ri

sk o

f ≥ 2

0%0.

66(0

.52-

0.80

)0.

94(0

.93-

0.96

)0.

39(0

.25-

0.53

)0.

98(0

.97-

0.99

)11

.88(

7.07

-19.

97)

0.36

(0.2

0-0.

63)

Ri

sk o

f ≥ 3

0%0.

50(0

.34-

0.67

)0.

97(0

.96-

0.98

)0.

48(0

.31-

0.65

)0.

97(0

.96-

0.98

)17

.45(

8.88

-34.

27)

0.50

(0.3

1-0.

82)

Oth

er S

BI

Ri

sk o

f ≥ 2

.5%

0.97

(0.9

4-0.

99)

0.62

(0.5

8-0.

66)

0.24

(0.1

9-0.

30)

0.99

(0.9

9-1.

00)

2.57

(2.2

2-2.

99)

0.05

(0.0

1-0.

26)

Ri

sk o

f ≥ 5

%0.

92(0

.88-

0.97

)0.

75(0

.72-

0.79

)0.

32(0

.25-

0.39

)0.

99(0

.98-

1.00

)3.

71(3

.04-

4.51

)0.

10(0

.04-

0.26

)

Ri

sk o

f ≥ 1

0%0.

88(0

.82-

0.94

)0.

83(0

.80-

0.86

)0.

39(0

.31-

0.47

)0.

98(0

.97-

0.99

)5.

05(4

.04-

6.32

)0.

15(0

.07-

0.30

)

Ri

sk o

f ≥ 1

5%0.

84(0

.77-

0.91

)0.

86(0

.84-

0.89

)0.

43(0

.35-

0.52

)0.

98)0

.97-

0.99

)6.

16(4

.78-

7.93

)0.

18(0

.10-

0.34

)

Ri

sk o

f ≥ 2

0%0.

80(0

.72-

0.88

)0.

89(0

.86-

0.91

)0.

46(0

.37-

0.56

)0.

97(0

.96-

0.98

)6.

95(5

.27-

9.17

)0.

22(0

.14-

0.37

)

Ri

sk o

f ≥ 3

0%0.

67(0

.57-

0.77

)0.

91(0

.89-

0.93

)0.

49(0

.39-

0.59

)0.

96(0

.94-

0.97

)7.

70(5

.49-

10.8

2)0.

36(0

.25-

0.52

)

CI: c

onfid

ence

inte

rval

, PPV

: pos

itive

pre

dict

ive

valu

e, N

PV: n

egat

ive

pred

ictiv

e va

lue,

LR+

: pos

itive

like

lihoo

d ra

tio, L

R-: n

egat

ive

likel

ihoo

d ra

tio.

15873_CvHouten_BNW_v2.indd 109 26-11-18 12:43

Page 112: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

110

Chapter 5

Table 3. Diagnostic reclassification by the updated Feverkidstool compared to the original Feverkidstool.The Feverkidstool classification was compared to the majority reference standard for the prediction of pneumonia (a) and other SBI (b). Head-to-head comparison of the original Feverkidstool and the updated Feverkidstool. Predicted risks of pneumonia (a) and other SBI (b) were low if the predicted risk was below 2.5%, intermediate between 2.5% and 10%, and high above 10%.

a.

Reference standard diagnosis:Pneumonia (n=30)

Reference standard diagnosis:No pneumonia (n=561)**

Updated Feverkidstool predicted risk for pneumonia

Updated Feverkidstool predicted risk for pneumonia

Low Intermediate High Low Intermediate High

Original Feverkidstool predicted risk for pneumonia

Low 3 1 1 304 20 3

Intermediate 1 2 4 100 46 27

High 0 0 18 14 14 33

* Among the patients with a pneumonia as determined by the majority of the expert panel, 6 patients were

correctly reclassified as being at higher risk using the updated Feverkidstool instead of the original Feverkidstool

and 1 patient was incorrectly reclassified. For patients in whom pneumonia was absent, these numbers are respectively 128 and 50. Reclassification improvement was 17% for patients with pneumonia (6 minus 1 of 30) and 14% for patients without pneumonia (128 minus 50 of 561).** As the comparison in this table is between the presence or absence of pneumonia, it should be noted that ‘no pneumonia’ includes viral and other SBI.

b.Reference standard diagnosis:

Other SBI (n=66)Reference standard diagnosis:

No other SBI (n=525)**Updated Feverkidstool

predicted risk for other SBIUpdated Feverkidstool

predicted risk for other SBI

Low Intermediate High Low Intermediate High

Original Feverkidstool predicted risk for other SBI

Low 1 0 0 98 0 0

Intermediate 1 2 6 190 55 25

High 0 4 52 40 51 66

* Among the patients with other SBI as determined by the majority of the expert panel, 6 patients were

correctly reclassified using the updated Feverkidstool instead of the original Feverkidstool and 5 patients were incorrectly reclassified. For patients in who other SBI is absent, these numbers are respectively 281 and 25. Reclassification improvement was 2% for patients with other SBI (6 minus 5 of total 66) and 49% for patients without other SBI (281 minus 25 of 525). ** As the comparison in this table is between the presence or absence of other SBI, it should be noted that ‘no other SBI’ includes viral and pneumonia.

Reclassification Table 3 shows the clinical consequences if the updated Feverkidstool was used instead of the original Feverkidstool. In total, the updated Feverkidstool for pneumonia reduced the number children with a falsely predicted high or intermediate risk from 234 to 143 (39%) compared to the original Feverkidstool, and from 427 to 197 (54%) for children with other SBI.

15873_CvHouten_BNW_v2.indd 110 26-11-18 12:43

Page 113: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

111

5

Optimizing prediction of bacterial infections

D isc ussionIn this study we showed that when a clinical prediction model including CRP was updated with a combined host-protein based assay, serious bacterial infections were predicted more accurately in children presenting with a lower RTI or FWS at the hospital. We showed that children with a low to moderate predicted risk benefit most from this updated model.

Strengths and weaknesses of the studyA strength of the present study is the used combination of variables. To our knowledge, this is the first study combining clinical parameters with both bacterial and viral biomarkers. A second strength is the use of an expert panel reference standard, which has the advantage of capturing a wide spectrum of illness severities, including difficult to diagnose cases, therefore likely to be generalizable to clinical practice.18 Another strength is the prospective patient recruitment, leading to assay results for all febrile children and not only for those with blood sampling indicated by routine care, representing the heterogeneous paediatric population presenting at the hospital. In addition, a strength of the Feverkidstool is its polytomous character. This enables the discrimination between different diseases: pneumonia and other SBI versus no SBI.19 Finally, in clinical practice different thresholds are needed depending on the setting. Therefore we performed a decision curve analysis to help interpret the differences between the models along the wide range of predicted probabilities.20

Limitations of our study should also be addressed. First, the number of bacterial cases was relatively low. Therefore it was not possible to refit the individual coefficients for all Feverkidstool variables, but, the aim of the study was not to build an optimal diagnostic model, but to see whether the new assay had additional value to the original well validated Feverkidstool. Second, one of the Feverkidstool variables, capillary refill, was not available for any of the participants. However, this dichotomous variable capillary refill has little diagnostic value within the Feverkidstool (regression coefficient 0.18 and 0.30 for the prediction of pneumonia and other SBI respectively), therefore the effect on the accuracy will be limited. Third, the reclassification is based on arbitrarily chosen thresholds (2.5% and 10%). These thresholds, however, mostly corresponds with LR that have been reported to be meaningful in decisions for febrile children: LR+ >5.0 for ruling-in SBI and LR- <0.2 for ruling-out SBI.17 Fourth, the Feverkidstool includes important clinical variables that are used by every physician when deciding to start antibiotics or not. Following clinical care, the expert panel was provided with a wide range of clinical information, including the clinical Feverkidstool variables. The panel, however was not informed about the algorithm. We, however, cannot exclude that incorporation bias—in which part of the test being assessed is included in the reference standard—has resulted

15873_CvHouten_BNW_v2.indd 111 26-11-18 12:43

Page 114: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

112

Chapter 5

in some degree of overestimation of the outcome. Finally, there were 71 patients for whom the expert panel could not assign a final diagnosis. Such inconclusive cases are inherent to studies using outcomes lacking a gold standard. To make optimal use of the data from all recruited patients, we have imputed these reference standard diagnoses. As the imputed diagnoses are used for both the original and the updated Feverkidstool, we do not expect this influenced the results of updating the Feverkidstool. In addition, a sensitivity analysis in which all cases with imputed diagnoses were excluded showed comparable results.

Comparison with existing literatureThe study in which the Feverkidstool was developed and the Opportunity study had comparable inclusion criteria, most important, both studies included children suspected of infection based on increased temperature.12, 14 Differences in inclusion criteria should also be discussed. The Feverkidstool derivation cohort included Dutch children aged 1 month to 15 years, whereas the Opportunity study included Dutch and Israeli children aged 1 month to 5 years. In addition, for the Feverkidstool development study children who received antibiotics before the ED visit were excluded, for the Opportunity study antibiotic use wasn’t an exclusion criteria.12, 14

We recently confirmed the external validity of the Feverkidstool, but when procalcitonin (PCT) was added to the prediction model or when CRP was replaced by PCT the accuracy for predicting SBI in febrile children did not improve.11 Another study also evaluated updating strategies for the Feverkidstool by adding PCT and resistin.13 They found that changes in positive and negative likelihood ratios for different risk thresholds are minimal. The Opportunity study showed that the combination assay significantly improved the diagnosis of bacterial infection compared to CRP and PCT.14 In contrast with the two above mentioned Feverkidstool updates, our current study has shown that updating the Feverkidstool with the assay does meaningfully improve the accuracy of the model. This further confirms our previous observation that the combination of CRP, TRAIL and IP-10 had higher diagnostic value in differentiating between bacterial and viral infections compared to CRP alone.14 The diagnostic value of TRAIL is noteworthy since its dynamics are complementary to traditionally studied bacteria-induced proteins such as CRP and PCT; TRAIL concentrations increase in viral infection and decrease during bacterial infection.

Implications for clinical practiceClinical signs and symptoms play an important role when physicians diagnose febrile children, but do not sufficiently differentiate between viral and bacterial infections. Therefore, the use of diagnostic prediction models that include both clinical parameters and biomarkers is intuitive and helpful. We showed that the updated Feverkidstool has most added value for patients in the low-moderate risk group, with predicted risk for SBI

15873_CvHouten_BNW_v2.indd 112 26-11-18 12:43

Page 115: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

113

5

Optimizing prediction of bacterial infections

below 40%. At thresholds of 2.5% and 10%, the reclassification table showed substantial improvement in diagnosis. The cases with predicted risks between low to moderate (2.5 vs 40%) may be characterized by having intermediate values of CRP with higher diagnostic uncertainty. Adding two viral biomarkers to the prediction rule will provide an extra dimension to the model and therefore improves diagnosis for especially those cases. As a point of care test is under development, the manufacturer has not given an indication to the eventual cost yet. To optimize cost-effective use of the combination test, our results suggest that the added value of the assay is the highest in children with a predicted risk <40% as predicted by the original Feverkidstool.

Implications for future researchSince we have proven the accuracy of the updated Feverkidstool, the next step is to perform a prospective and external validation el, and to evaluate its impact on resource use and antibiotic treatment. An important aspect is to define risk cut-offs for different settings in clinical practice, e.g. young children and children with co-morbidities. In addition, in order to optimize resource use, new biomarkers may benefit selected patient subgroups in particular (e.g. selected on a set of clinical characteristics/predicted risk) rather than in all febrile children. This targeted risk approach may also be applied to position the role of e.g. Myxovirus resistance protein A (MxA) and CRP.21

ConclusionIn conclusion, a new blood assay including viral and bacterial biomarkers, combined with a clinical prediction model, is in this study cohort superior to the model with CRP only for predicting serious bacterial infection in preschool children. In children with low to moderate predicted risk of SBI in particular, the updated Feverkidstool with the assay has the potential to optimize targeted antibiotic prescription and to prevent unnecessary use of antibiotics.

15873_CvHouten_BNW_v2.indd 113 26-11-18 12:43

Page 116: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

114

Chapter 5

References1. Alpern ER, Stanley RM, Gorelick MH, Donaldson A, Knight S, Teach SJ, et al. Epidemiology of

a pediatric emergency medicine research network: the PECARN Core Data Project. Pediatric emergency care. 2006;22(10):689-99.

2. Kronman MP, Zhou C, Mangione-Smith R. Bacterial prevalence and antimicrobial prescribing trends for acute respiratory tract infections. Pediatrics. 2014;134(4):e956-65.

3. Chancey RJ, Jhaveri R. Fever without localizing signs in children: a review in the post-Hib and postpneumococcal era. Minerva Pediatr. 2009;61(5):489-501.

4. Hubert-Dibon G, Danjou L, Feildel-Fournial C, Vrignaud B, Masson D, Launay E, et al. Procalcitonin and C-reactive protein may help to detect invasive bacterial infections in children who have fever without source. Acta paediatrica. 2018;107(7):1262-9.

5. Van Boeckel TP, Gandra S, Ashok A, Caudron Q, Grenfell BT, Levin SA, et al. Global antibiotic consumption 2000 to 2010: an analysis of national pharmaceutical sales data. Lancet Infect Dis. 2014;14(8):742-50.

6. Kraus EM, Pelzl S, Szecsenyi J, Laux G. Antibiotic prescribing for acute lower respiratory tract infections (LRTI) - guideline adherence in the German primary care setting: An analysis of routine data. PLoS ONE. 2017;12(3).

7. Laxminarayan R, Duse A, Wattal C, Zaidi AK, Wertheim HF, Sumpradit N, et al. Antibiotic resistance-the need for global solutions. Lancet Infect Dis. 2013;13(12):1057-98.

8. ECDC Technical Report: The bacterial challenge: time to react, Sept 2009. Available from: http://ecdc.europa.eu/en/publications/Publications/0909_TER_The_Bacterial_Challenge_Time_to_React.pdf (accessed 11-8-2016).

9. Craig JC, Williams GJ, Jones M, Codarini M, Macaskill P, Hayen A, et al. The accuracy of clinical symptoms and signs for the diagnosis of serious bacterial infection in young febrile children: prospective cohort study of 15 781 febrile illnesses. BMJ. 2010;340:c1594.

10. Lacour AG, Zamora SA, Gervaix A. A score identifying serious bacterial infections in children with fever without source. Pediatr Infect Dis J. 2008;27(7):654-6.

11. Nijman RG, Vergouwe Y, Moll HA, Smit FJ, Weerkamp F, Steyerberg EW, et al. Validation of the Feverkidstool and procalcitonin for detecting serious bacterial infections in febrile children. Pediatr Res. 2017.

12. Nijman RG, Vergouwe Y, Thompson M, van Veen M, van Meurs AH, van der Lei J, et al. Clinical prediction model to aid emergency doctors managing febrile children at risk of serious bacterial infections: diagnostic study. Bmj. 2013;346:f1706.

13. Irwin AD, Grant A, Williams R, Kolamunnage-Dona R, Drew RJ, Paulus S, et al. Predicting Risk of Serious Bacterial Infections in Febrile Children in the Emergency Department. Pediatrics. 2017;140(2).

14. van Houten CB, de Groot JA, Klein A, Srugo I, Chistyakov I, de Waal W, et al. A host-protein based assay to differentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study. Lancet Infect Dis. 2017;17(4):431-40.

15. Oved K, Cohen A, Boico O, Navon R, Friedman T, Etshtein L, et al. A Novel Host-Proteome Signature for Distinguishing between Acute Bacterial and Viral Infections. PLoS One. 2015;10(3):e0120012.

16. Srugo I, Klein A, Stein M, Golan-Shany O, Kerem N, Chistyakov I, et al. Validation of a Novel Assay to Distinguish Bacterial and Viral Infections. Pediatrics. 2017;140(4).

15873_CvHouten_BNW_v2.indd 114 26-11-18 12:43

Page 117: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

115

5

Optimizing prediction of bacterial infections

17. Thompson M, Van den Bruel A, Verbakel J, Lakhanpaul M, Haj-Hassan T, Stevens R, et al. Systematic review and validation of prediction rules for identifying children with serious infections in emergency departments and urgent-access primary care. Health Technol Assess. 2012;16(15):1-100.

18. Reitsma JB, Rutjes AW, Khan KS, Coomarasamy A, Bossuyt PM. A review of solutions for diagnostic accuracy studies with an imperfect or missing reference standard. J Clin Epidemiol. 2009;62(8):797-806.

19. Biesheuvel CJ, Vergouwe Y, Steyerberg EW, Grobbee DE, Moons KG. Polytomous logistic regression analysis could be applied more often in diagnostic research. J Clin Epidemiol. 2008;61(2):125-34.

20. Vickers AJ, Van Calster B, Steyerberg EW. Net benefit approaches to the evaluation of prediction models, molecular markers, and diagnostic tests. Bmj. 2016;352:i6.

21. Engelmann I, Dubos F, Lobert PE, Houssin C, Degas V, Sardet A, et al. Diagnosis of viral infections using myxovirus resistance protein A (MxA). Pediatrics. 2015;135(4):e985-93

15873_CvHouten_BNW_v2.indd 115 26-11-18 12:43

Page 118: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

116

Chapter 5

S upplementar y material

777 patients

recruited

202 patients

recruited in the

Netherlands

575 patients

recruited in

Israel

591 in primary

study population

186 excluded

103 non-infectious

41 did not fulfill

inclusion criteria

24 no blood sample

16 ICU admission

2 other

72 diagnosed as

bacterial by

expert panel

(reference standard)

448 diagnosed as

viral by

expert panel

(reference standard)

71 diagnosed as

inconclusive by

expert panel

(reference standard)

26

Pneumonia

66

Other SBI

499

viral reference

standard

92

bacterial reference

standard

51

viral

20

bacterial

Majority diagnosis

10 times imputation

Supplemental Figure 1. Flowchart of included patients.

15873_CvHouten_BNW_v2.indd 116 26-11-18 12:43

Page 119: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

117

5

Optimizing prediction of bacterial infections

Specificity

Sen

sitiv

ity

0.0

0.2

0.4

0.6

0.8

1.0

1.0 0.8 0.6 0.4 0.2 0.0

Original FKT for pneumonia, c-statistic= 0.84 (95% CI 0.77-0.92) Updated FKT for pneumonia, c-statistic= 0.89 (95% CI 0.82-0.96) Original FKT for other SBI, c-statistic= 0.82 (95% CI 0.77-0.86) Updated FKT for other SBI, c-statistic= 0.91 (95% CI 0.87-0.94)

Supplemental Figure 2. Receiver operating characteristics (ROC) curve of sensitivity versus specificity for the original and updated Feverkidstool. Area under the receiving operating curve (c-statistic) for the original and for the updated Feverkidstool for pneumonia and other SBI are shown in the figure. The c-statistic difference for pneumonia is 0.09 and 0.05 for other SBI. FKT: Feverkidstool, SBI: serious bacterial infection.

15873_CvHouten_BNW_v2.indd 117 26-11-18 12:43

Page 120: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

118

Chapter 5

Supplemental Table 1. Characteristics of patients with and without reference standard.Viral and bacterial reference standard

diagnosis(n=520)

Inconclusive reference standard

diagnosis(n=71) p-value

Predictor variables

Age (years) (median, IQR) 1.2(0.6-2.4) 1.7(1.0-3.0) 0.01

Gender (male) (n,%) 289(56%) 42(59%) 0.61

Duration of fever (days) (median, IQR) 2(1-4) 2(1-4) 0.80

Temperature (°C) (median, IQR) 38.5(37.7-39.2) 38.6(37.8-39.3) 0.49

Respiratory rate (mean, SD) 42(16) 39(12) 0.35

Tachypnea (n,%) 140(27%) 19(27%) 0.74

Heart rate (mean, SD) 152(24) 151(25) 0.92

Tachycardia (n,%) 248(48%) 34(48%) 0.77

Oxygen saturation (%O2) (median, IQR) 98(97-100) 98(96-100) 0.41

Desaturation (<94%O2) (n,%) 25(5%) 0(0%) 0.10

Chest wall retractions (n,%) 67(13%) 3(4%) 0.045

Ill appearanc (n, %) 153(29%) 26(37%) 0.22

C-reactive protein (mg/l) (median, IQR) 18(7-46) 53(18-83) <0.001

Assay score (median, IQR) 7(1-40) 35(5-88) <0.001

Other variables

Hospital admission (n,%) 290(56%) 38(54%) 0.70

Hospitalization duration (days) (median, IQR) 3(2-4) 3(2-5) 0.08

Antibiotic treatment prescribed (n,%) 180(35%) 53(75%) <0.001

Recruiting site 0.80

Secondary care centre (n,%) 487(94%) 66(93%)

Tertiary care centre (n,%) 33(6%) 5(7%)

Focus of infection 0.21

Central nervous system 9(2%) 0(0%)

Gastrointestinal tract 19(4%) 1(1%)

Other 42(8%) 11(16%)

Respiratory tract 254(48%) 40(56%)

Systemic 10(2%) 1(1%)

Unknown 164(32%) 16(23%)

Urinary tract 22(4%) 2(3%)

Clinical syndrome was based on the diagnosis of the attending physician at discharge from the hospital. LRTI included pneumonia and bronchiolitis; URTI included laryngitis, pharyngitis, otitis media, sinusitis and tonsillitis. Baseline characteristics of excluded patients are shown in the appendix. *p-values of differences between bacterial and viral infections.

15873_CvHouten_BNW_v2.indd 118 26-11-18 12:43

Page 121: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

119

5

Optimizing prediction of bacterial infections

Supplemental Table 2. Description of recalibration of the Feverkidstool and the updated model.

Linear predictors for pneumonia and other SBI Feverkidstool are calculated as defined previously [1]:

LP (pneumonia Feverkidstool) = -17.9 (Intercept) + 1.02 * Age (max 1 year, in years) + 0.01 * Age (if >1

year: age in years - 1)+ 0.13 * Sex (female) + 0.29 * Temperature (ºC) + 0.21 * Duration of fever (days)

+ 0.44 * Presence of tachypnoea - 0.04 * Presence of tachycardia + 1.59 * Oxygen saturation <94%

- 0.18 * Capillary refill time (>3s) + 0.47 * Presence of chest wall retractions + 0.16* ill appearance +

0.64 * Ln(CRP) (mg/l)

LP (other SBI Feverkidstool) = -4.7 (Intercept) -1.73 * Age (max 1 year, in years) + 0.11 * Age (if >1 year:

age in years − 1)+ 0.70 * Sex (female) - 0.02 * Temperature (ºC) - 0.03 * Duration of fever (days) - 0.11

* Presence of tachypnoea - 0.02 * Presence of tachycardia - 3.29 * Oxygen saturation <94% + 0.30 *

Capillary refill time (>3s) - 3.78 * Presence of chest wall retractions + 0.27 * ill appearance + 1.14 *

Ln(CRP) (mg/l)

where LP refers to the linear predictor in a (polytomous) logistic regression model.

We used the fixed intercept and coefficients within the original Feverkidstool, but used the outcome as a linear predictor in our logistic recalibration model and in the updated model. This resulted in the following (polytomous) logistic regression models:

Recalibration of original Feverkidstool:LP1 = -0.58 + 1.28 (LP pneumonia Feverkidstool)LP2 = -0.02 + 0.54 (LP other SBI Feverkidstool)

Updated Feverkidstool, including combination assay:LP3 = -4.19 + 0.59 (LP pneumonia Feverkidstool, with median CRP for all patients)+ 0.05 (score Assay)LP4 = -3.57 + 0.24 (LP other SBI Feverkidstool, with median CRP for all patients)+ 0.05 (score Assay)

Probabilities of the outcomes are calculated with:Original Feverkidstool Risk (pneumonia) = eLP1/ (1 + eLP1 + eLP2) Original Feverkidstool Risk (other SBI) = eLP2 / (1 + eLP1 + eLP2),Updated Feverkidstool Risk (pneumonia) = eLP3 / (1 + eLP3 + eLP4) Updated Feverkidstool Risk (other SBI) = eLP4 / (1 + eLP3 + eLP4), [2, 3]

15873_CvHouten_BNW_v2.indd 119 26-11-18 12:43

Page 122: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

120

Chapter 5

References1. Nijman RG, Vergouwe Y, Thompson M, van Veen M, van Meurs AH, van der Lei J, et al. Clinical

prediction model to aid emergency doctors managing febrile children at risk of serious bacterial infections: diagnostic study. Bmj. 2013;346:f1706.

2. Begg CB, Gray R. Calculation of polychotomous logistic-regression parameters using individualized regressions. Biometrika. 1984(71):11-8.

3. Wijesinha A, Begg CB, Funkenstein HH, McNeil BJ. Methodology for the differential diagnosis of a complex data set. A case study using data from routine CT scan examinations. Medical decision making : an international journal of the Society for Medical Decision Making. 1983;3(2):133-54.

15873_CvHouten_BNW_v2.indd 120 26-11-18 12:43

Page 123: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 121 26-11-18 12:43

Page 124: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

6

15873_CvHouten_BNW_v2.indd 122 26-11-18 12:43

Page 125: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

6Observational multi-centre, prospective study to characterize novel pathogen- and host-related factors in hospitalized patients with lower respiratory tract infections and/or sepsis:the “TAILORED-Treatment” study

CB van Houten, K Oved, E Eden, A Cohen, D Engelhard, SA Boers, R Kraaij, R Karlsson, D Fernandez, E Gonzalez, Y Li, AP Stubbs, ERB Moore, JP Hays and LJ Bont

BMC Infectious Diseases 2018; 18:377

15873_CvHouten_BNW_v2.indd 123 26-11-18 12:43

Page 126: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

124

Chapter 6

Abstrac tBackground The emergence and spread of antibiotic resistant micro-organisms is a global concern, which is largely attributable to inaccurate prescribing of antibiotics to patients presenting with non-bacterial infections. The use of ‘omics’ technologies for discovery of novel infection related biomarkers combined with novel treatment algorithms offers possibilities for rapidly distinguishing between bacterial and viral infections. This distinction can be particularly important for patients suffering from lower respiratory tract infections (LRTI) and/or sepsis as they represent a significant burden to healthcare systems. Here we present the study details of the TAILORED-Treatment study, an observational, prospective, multi-centre study aiming to generate a multi-parametric model, combining host and pathogen data, for distinguishing between bacterial and viral aetiologies in children and adults with LRTI and/or sepsis.

MethodsA total number of 1200 paediatric and adult patients aged 1 month and older with LRTI and/or sepsis or a non-infectious disease are recruited from Emergency Departments and hospital wards of seven Dutch and Israeli medical centres. A panel of three experienced physicians adjudicate a reference standard diagnosis for all patients (i.e., bacterial or viral infection) using all available clinical and laboratory information, including a 28-day follow-up assessment. Nasal swabs and blood samples are collected for multi-omics investigations including host RNA and protein biomarkers, nasal microbiota profiling, host genomic profiling and bacterial proteomics. Simplified data is entered into a custom-built database in order to develop a multi-parametric model and diagnostic tools for differentiating between bacterial and viral infections. The predictions from the model will be compared with the consensus diagnosis in order to determine its accuracy.

DiscussionThe TAILORED-Treatment study will provide new insights into the interplay between the host and micro-organisms. New host- or pathogen-related biomarkers will be used to generate a multi-parametric model for distinguishing between bacterial and viral infections. This model will be helpful to better guide antimicrobial therapy for patients with LRTI and sepsis. This study has the potential to improve patient care, reduce unnecessary antibiotic prescribing and will contribute positively to institutional, national and international healthcare economics.

15873_CvHouten_BNW_v2.indd 124 26-11-18 12:43

Page 127: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

125

6

The “TAILORED-Treatment” study

Introduc tionThe burden of antibiotic misuse Antibiotics are the most prescribed class of drugs world-wide.1 However, 30–50% of antibiotics are estimated to be prescribed inappropriately, making antibiotics also the most misused drug class.1-5 Antibiotic overuse, i.e. prescribing antibiotics to treat a non-bacterial disease, is a serious problem. For example, in the US over 80 million antibiotic prescriptions are given annually for viral infections in the outpatient setting.6 This may cause the emergence of adverse events (AEs) such as allergic reactions and antibiotic-associated diarrhoea.7 Importantly, antibiotic overuse has been linked to the emergence of resistant strains of bacteria, by the Center for Disease Control considered as “one of the world’s most pressing health problems in the 21st century”.8,9 Annually, 23,000 deaths due to resistant bacteria are expected in the US alone.8,10 Resistant bacteria are projected to cause over 10 M annual deaths worldwide by 2050, surpassing cancer as the leading cause of death.8,11 A second type of antibiotic misuse is underuse, i.e. delayed or no antibiotic treatment in case of a bacterial disease, from which a patient could have benefited.12–17 Although reducing the risk of antibiotic-related AEs18,19, underuse of antibiotics may lead to a prolonged disease duration and an increased rate of complications that could have been avoided with early antimicrobial treatment.19–21 For example, up to 15–40% of adult patients hospitalized for bacterial pneumonia in the US receive delayed or no antibiotic treatment.14,15 Finally, applying the wrong antibiotic spectrum to treat a bacterial disease is the third type of antibiotic misuse. For example, administering third generation cephalosporins instead of macrolides to treat a respiratory infection caused by atypical bacteria.

Health economics consequencesAntibiotic overuse has also several health economics consequences. For instance, the annual cost of unnecessary antibiotic prescriptions for adult respiratory infections in the US is estimated to be $1.1 billion.22 In addition, there are also indirect costs, including the costs of treating preventable antibiotic-related AEs and the costs of prolonged hospital stay as a result of AEs. Lastly, are the costs related to the emergence of antibiotic-resistant bacterial strains. The cost of treating patients with antibiotic-resistant bacteria is estimated at $16-26 billion annually in the US23-25 and over €1.5 billion in the EU.26 The health economics consequences of underuse of antibiotics include the costs of treating preventable complications and a prolonged disease duration resulting from a delayed or no antibiotic treatment.19-21

15873_CvHouten_BNW_v2.indd 125 26-11-18 12:43

Page 128: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

126

Chapter 6

Antibiotics in respiratory tract infections and sepsisRespiratory tract infections are one of the most common causes of hospital and outpatient visits in the EU, comprising 1 out of 3 admissions annually.9 In patients with mild respiratory tract infections, over prescription of antibiotics in general and prescription of broad spectrum antibiotics are large problems. Broad spectrum antibiotics account for approximately half of all antibiotic prescribing for children with acute RTI27, with the above mentioned consequences.8 Sepsis-related hospital admissions are less frequent but represent a significant burden to healthcare systems in terms of adverse patient outcomes and the need for rapid, but effective, intervention strategies.28 In septic patients the key clinical challenge is to ensure that the correct antimicrobial treatment is administered, and that this therapy is administered as soon as possible.29

Limitations of current diagnostic toolsAlthough the current diagnostic tools for facilitating appropriate use of antibiotics (such as culture-, PCR-, and immunoassay-based) may be valuable in some clinical situations, they have major limitations (Figure 1). First, existing diagnostic tools often require hours to days to provide information, whereas physicians need to decide whether to prescribe antibiotics within shorter time periods. Second, available diagnostic technologies usually require direct sampling of the pathogen. Such sampling is often not feasible if the infection site is inaccessible (e.g., pneumonia, sinusitis, middle-ear infection, etc.). A third limitation is that available technologies usually search for the presence of specific bacteria or viruses. However, many types of bacteria and viruses could be present as part of the natural flora without causing an infection. For example, the important respiratory tract pathogenic bacteria S. pneumoniae is also part of the upper respiratory tract natural flora in up to 68% and 15% of healthy children and adults, respectively30 and respiratory viruses were detected in 35.4% of a-symptomatic children.31 Finally, diagnostic solutions that were developed for detection of a specific pathogenic strain, often fail to detect the constantly evolving and emerging strains of the same family of pathogens, for example rapid Influenza tests showed low sensitivity, 45%, during the 2009 H1N1 influenza A viral outbreak.32 These limitations lead to a ‘diagnostic gap’, which in turn often leads physicians to either over-prescribe (e.g., ‘just-in-case’ approach) or under-prescribe (e.g., ‘watchful waiting’ approach) antibiotics18-20, both of which adversely influence patient care and health economics.

15873_CvHouten_BNW_v2.indd 126 26-11-18 12:43

Page 129: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

127

6

The “TAILORED-Treatment” study

1

Figure 1. Limitations of current diagnostic tools.

The TAILORED-Treatment projectThe health and health economics consequences of antibiotic misuse highlight the need for a diagnostic tool that would help physicians to use antibiotics appropriately. Ideally, this diagnostic tool should provide a diagnosis that can accurately and rapidly differentiate between bacterial and viral infections. However, as previously described, current diagnostic tools are often inadequate (Figure 1). Therefore, the TAILORED-Treatment consortium was established to develop new tools aimed at increasing the effectiveness of antibiotic therapy, reduce adverse events, and limit the emergence of antimicrobial resistance in children and adults. In reality, targeted antimicrobial therapy can most effectively be achieved by utilizing personalized data to facilitate a tailored and optimized approach to individual patient treatment. This can best be achieved by utilizing knowledge gained from both host-centred and pathogen-centred parameters during health and disease. Unfortunately, these parameters have traditionally, tended to be measured independently, and used on an ad hoc basis without careful integration for the best treatment of the patient. However, recent advances in the development of high-throughput and sensitive molecular-based technologies, improved databases, and bioinformatics analysis tools, rapidly enabling that the goal of personalized medicine and treatment can be reached. Unfortunately, however, there currently exists

15873_CvHouten_BNW_v2.indd 127 26-11-18 12:43

Page 130: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

128

Chapter 6

a ‘technological gap’ between recent state-of-the-art methodologies (for example with respect to gaining new insights into novel host-pathogen interactions) and laboratory-to-bedside results to benefit patients, physicians and society as a whole.33 The observational TAILORED-Treatment project is designed to bridge this technological gap in order to generate novel insights and innovations that are readily exploitable in the field of personalized medicine and infectious diseases.

Study ObjectivesPrimary ObjectiveTo develop a novel multi-parametric diagnostic model for the management of patients with LRTI and/or sepsis that is based on novel pathogen- and host-related factors.

Secondary Objectives1. Identify individual as well as sets of blood host biomarkers for differentiating

between bacterial, and viral aetiologies and determine the sensitivity and specificity of these biomarkers in distinguishing between the different infection types;

2. Identify individual as well as sets of blood host biomarkers for differentiating between bacterial subtypes, namely Gram positive, Gram negative and atypical bacteria;

3. Characterize the temporal dynamics of the host-pathogen interactions based on multiple sampling before and during antimicrobial treatment;

4. Characterize the respiratory microbiome in patients presenting with LRTI and/or sepsis;

5. Develop a mass spectrometry based, rapid detection technique for identification of microbial pathogens and antimicrobial resistances in clinical samples;

6. Develop a personalized treatment algorithm for tailored antimicrobial therapy that integrates clinical, molecular and biochemical data;

7. Define genetic mechanisms underlying the differential host response between patients with viral versus bacterial infection.

MethodsGeneral study design In this prospective, multi-centre, observational study, an expected total number of 1200 paediatric and adult patients aged 1 month and older with suspected LRTI and/or sepsis or a non-infectious disease are recruited, in order to develop a diagnostic model to differentiate between bacterial and viral infections. The study will generate a model for patients with LRTI and a separate model for patients with sepsis. The prediction of the model obtained from each patient will be compared with a consensus diagnosis by

15873_CvHouten_BNW_v2.indd 128 26-11-18 12:43

Page 131: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

129

6

The “TAILORED-Treatment” study

a panel of three experts in order to determine its accuracy (Figure 2). The study is a non-interventional study, and therefore, the diagnostic prediction of the model will not be used for any clinical or diagnostics decision making. Local data monitor plans were used and executed by local, independent monitors.

Study participants Patients aged 1 month and older that attend the Emergency Department (ED) and hospital wards of seven Dutch and Israel medical centres (secondary and tertiary) due to a suspected LRTI and/or sepsis that agree (or their legal guardians agrees) to sign an informed consent and meet the inclusion criteria and none of the exclusion criteria (Table 1), are eligible to participate in the study. Informed consent is asked by the study team (i.e. research nurses or medical doctor). Sepsis criteria are defined according to published criteria.34 Patients receive care as usual. The non-infectious disease group include patients with clinical signs of a non-infectious disease.

Table 1. Eligible criteria for The TAILORED Treatment study.

Inclusion criteria Exclusion criteria

Age ≥ 1 month Proven or suspected HIV, HBV, or HCV infection

Symptoms ≤ 8 days Post-transplant

LRTI; ≥ 2 signs Congenital immune deficiency

Respiratory rate > 20/min* Obvious alternative causes of respiratory distress

Chest cough Nosocomial LRTI (> 3days after hospitalization)

Nasal flaring Other febrile infection during the past 3 weeks

Retractions Active (haematological) malignancy

Rales Immune-suppressive or immune-modulating therapies

Expiratory wheeze Moderate to severe psychomotor retardation

Decreased breath sounds Moderate to severe congenital metabolic disorder

Sepsis**; ≥ 2 signs***

Heartrate > 90/min

Respiratory rate > 20/min

Core body temperature >38°C or <36°C

White blood cell count >12,000 cells/ μl or <4,000/ μl

* WHO age-specific criteria for tachypnea will be used for children.41

** Age-specific criteria for sepsis criteria according to the guidelines of the International Paediatric Sepsis Consensus Conference 2005 will be used for children.42

*** One of which must be abnormal temperature or white blood cell count. HIV: human immunodeficiency virus, HBV: hepatitis B virus, HCV: hepatitis C virus

15873_CvHouten_BNW_v2.indd 129 26-11-18 12:43

Page 132: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

130

Chapter 6

Medical

recordsPCR results

Identify potential participants

Enrollment (informed consent)

Clinical evaluation (care as usual)

Blood samples Nasal swabsStool sample

(in case of diarrhea)

Repeated sampling 3-5 days

from inclusion in subgroup

Follow-up phone call 28 days

from inclusion

Electronic Case

Record Form (eCRF)

Physician #1 Physician #3Physician #2

Diagnosis #1 Diagnosis #3Diagnosis #2

eCRF

Consensus decision3 ouf of 3

No decisionMarjority decision

2 ouf of 3

Decision will be

used to determine

model accuracy

Data cannot be

used to determine

model accuracy

Decision will be

analyzed as a

seprate sub-group

Figure 2. Schematic diagram of the study workflow.

15873_CvHouten_BNW_v2.indd 130 26-11-18 12:43

Page 133: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

131

6

The “TAILORED-Treatment” study

Data/sample collectionClinical dataClinical evaluation and management is performed according to standard institutional procedures (Figure 2). Collected clinical data, includes demographics, medical history, clinical symptoms, physical examination, disease course, laboratory measurements, as well as more advanced diagnostic information including microbiological (including an additional multiplex PCR), and serological investigations. A follow-up phone call 28-day after recruitment, defines the end of follow-up and generates data to be used by the expert panel (e.g. maximum disease severity, total use of antibiotics).

Sample collectionFor study purposes two blood samples, two nasal swabs and (in case of diarrhoea) a stool sample are collected (Figure 2). Blood and nasal swabs sampling at two sampling points (at inclusion and after 3-5 days) is performed in hospitalized Dutch children in order to monitor the temporal dynamics of the host-pathogen interactions. One nasal swab sample (Universal Transport Medium, Copan, USA) is used for PCR based microbiological investigations performed for the establishment of patient diagnosis (only at inclusion). The PCR results are included in the eCRF and are available to the TAILORED-Treatment expert panel while determining the diagnosis of the patient (Figure 2). The second nasal swab is collected in Transport Medium suitable for bacterial culturing (e-swab, Liquid Amies Medium, Copan), this sample is used for respiratory microbiome investigations and to develop a rapid detection technique for identification of microbial pathogens and antimicrobial resistances, based on mass spectrometry and proteomics for discovery of peptide biomarkers. Serum samples are used for different proteomic measurements, leukocytes are isolated for discovery of RNA-based biomarkers. Following RNA isolation, remaining DNA is used for GWAS studies. GWAS analyses is performed in the Netherlands, as Institutional review board (IRB) approval for genetic testing is not obtained in Israel. DNA collection is mentioned explicitly in the patient information. Blood samples are collected from children with a non-infectious disease only if blood sampling is required as part of their routine care. In cases of diarrhoea on enrolment, stool samples are collected. These samples will be evaluated to identify Clostridium difficile infections.

‘Proteotyping’Shotgun proteomic detection and identification of microorganisms, or ‘proteotyping’, relies on recognition of species-unique peptides by tandem mass spectrometry (MS), allowing discrimination of species facilitated at amino acid level resolution. The recent evolution of mass spectrometers, having high sensitivity, accuracy and resolution, enables detection of almost the complete proteome of a microorganism. With such

15873_CvHouten_BNW_v2.indd 131 26-11-18 12:43

Page 134: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

132

Chapter 6

analytical means, it is feasible to determine directly, within a clinical sample, the species identity, the sub-species strain type and the factors expressing virulence and antimicrobial resistance.35 Clinical samples are processed by first removing human biomass and subsequently collecting bacteria. The proteins of the collected bacteria were processed by the LPITM FlowCell or in-solution digestion protocols followed by liquid chromatography tandem MS (LC-MS/MS) analysis. The recently developed technology called Lipid-based Protein Immobilization (LPI™) is based on immobilization of biological material within a flow cell, followed by digestion of exposed proteins by an enzyme, such as trypsin.36-38 The MS-based proteomics methodology has been applied successfully, without prior cultivations to the analyses of clinical nasal swab samples from the Sahlgrenska University Hospital that have been confirmed positive for a respiratory infectious bacterial species, i.e., S. pneumoniae, H. influenza, M. catarrhalis and S. aureus (unpublished data).

Central DatabaseAll clinical data (eCRFs) and research data (host biomarkers, host microbiome, pathogen identity and resistance) is coded and uploaded to a central, secured database (HoPOIT database) to which access is granted only to research partners (Figure 3). The centralized clinical, microbiological, molecular and biochemical data is used to develop a multi-parametric model and diagnostic tools for differentiating between bacterial and viral infections.

1

Central database Developing and validating multi-parametric model for differentiating

between bacterial and viral aetiologies.

Blood samples Nasal swabs

Protein- and RNA-based biomarker

screen

GWAS

Multiplexed PCR Respiratory pathogens

Microbiome

analysis

Pathogen proteomics

Medical records

Electronic Case Record Form

(eCRF)

Expert panel diagnosis

Figure 3. Development of the multi-parametric model.

15873_CvHouten_BNW_v2.indd 132 26-11-18 12:43

Page 135: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

133

6

The “TAILORED-Treatment” study

Aetiology determinationCurrently, no single reference standard exists for differentiating between bacterial and viral aetiology in an acute infectious disease. Therefore, we follow the Standards for Reporting of Diagnostic Accuracy (STARD) recommendation39, and create a highly rigorous reference standard based on expert panel adjudication (Figure 2). The eCRF of each patient is available to a panel of three independent physicians that are affiliated to the country of recruitment. Based on the information included in the eCRF, each member of the panel assigns one of the following diagnostic labels to each one of the patients: (i) bacterial infection; (ii) viral infection; (iii) mixed infection (i.e., bacterial and viral co-infection); (iv) non-infectious disease; or (v) undetermined. Of note, each expert is blinded to the research results and to the labels of his peers on the expert panel. For this purpose, mixed infection (bacterial and viral co-infection) is considered as bacterial infections, as they often elicit similar patient management. A final diagnosis is determined based on consensus agreement between all three experts (Figure 2). The final diagnosis is used for the purpose of calculating the accuracy of the newly developed diagnostic tools. Cases with only majority agreement (2 out of 3 experts) are analysed as a separate sub-group. The remaining cases are labelled as undetermined. Specifically, this label is assigned in cases where each of the experts assigns a different diagnosis or in cases where 2 or more of the experts assign an “undetermined” diagnosis. These patients are used for data analysis and are expected to comprise roughly 10% of all patients.

Study Stages The study is conducted in two consecutive stages. Stage A is used to collect molecular, biochemical, microbiological and clinical data on a group of approximately 900 patients. Collected data is used to develop unique diagnostic signatures able to distinguish between bacterial and viral aetiologies, a mass spectrometry based rapid detection technique for identification of microbial pathogens and antimicrobial resistances and a personalized treatment algorithm for tailored antimicrobial therapy that integrates clinical, molecular and biochemical data. In stage B, a second, independent cohort of approximately 300 patients is recruited for the purpose of testing and validating the performances of the multi-parametric model.

OutcomesPrimary EndpointSensitivity and specificity for a multi-parametric diagnostic model, incorporating different pathogen- and host-related factors, in differentiating between bacterial and viral aetiology in patients with LRTI and/or sepsis.

15873_CvHouten_BNW_v2.indd 133 26-11-18 12:43

Page 136: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

134

Chapter 6

Secondary Endpoints1. Sensitivity and specificity for host-related individual blood biomarkers, in

differentiating between bacterial and viral aetiology from other aetiologist in patients with LRTI and/or sepsis;

2. Sensitivity and specificity for host-related sets of blood biomarkers, in differentiating Gram positive or Gram negative or atypical aetiology from other disease aetiologist in patients with LRTI and/or sepsis;

3. Monitoring the temporal dynamics of host-related blood biomarkers levels during the course of disease in patients with LRTI and/or sepsis;

4. A list of significant bacterial microbiome components that are associated with poor or favourable clinical outcome in patients with LRTI and/or sepsis;

5. Sensitivity and specificity for liquid chromatography-mass spectrometry (LC-MS/MS) and lipid-based Protein Immobilization (LPI) proteomics-based rapid detection technique in identifying pathogens in clinical samples of patients with LRTI and/or sepsis;

6. A web-based application that recommends physicians with a preferred antimicrobial treatment based on patients clinical, molecular and biochemical data;

7. To define genetic mechanisms underlying the different host response between patients with viral versus bacterial infection.

Sample size calculation The following power analysis aims to achieve the primary study objective of developing a new diagnostic model for classifying patients with viral and bacterial aetiologist and treatment algorithms to guide antimicrobial prescribing.

Paediatric population analysisFirst, we estimate the sample size required to reject the null hypothesis that the sensitivity over the entire population, P, is lower than P0=70% (H0: P ≤ P0, H1: P > P0) with significance level of 1% (α = 0.01), power of 80% (=1-β) for a true sensitivity P1 of 90% (P1 - P0 ≥ 0.2). For sufficiently large sample sizes (n >~30) the sample distribution of the sensitivity, is approximately normally distributed, (p ̂ ~ N{P, P(1-P)/n}). Thus, the number of bacterial patients that are required is:

𝑛𝑛 =(𝑍𝑍1−𝛼𝛼√𝑃𝑃0(1 − 𝑃𝑃0) + 𝑍𝑍1−𝛽𝛽√𝑃𝑃1(1 − 𝑃𝑃1))

2

(𝑃𝑃1 − 𝑃𝑃0)2≤ 76

We estimate that the ratio between bacterial and viral paediatric patients is 1:3 and that expert consensus is achieved in 65% of the cases. Thus, the number of patients with an acute infection required in order to reach 76 bacterial patients is 468 = (76 / 25% / 65%).

15873_CvHouten_BNW_v2.indd 134 26-11-18 12:43

Page 137: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

135

6

The “TAILORED-Treatment” study

Using similar considerations for computing specificity the sample size for viral patients is also 76. Of note, we anticipate that the above mentioned 468 patients will include 229 (= 468 x 75% x 65%) viral patients, and thus there is no need for additional patients. To assess the host-response based diagnostics accuracy in differentiating patients with an acute infection (both viral and bacterial) and non-infectious patients, assuming P0=70% (H0: P ≤ P0, H1: P > P0) with significance level of 1% (α = 0.01), power of 80% (=1-β) for a true sensitivity P1 of 90% (P1 - P0 ≥ 0.2) requires 76 non-infectious patients. Overall, the paediatric cohort should include at least 468 patients with a suspected infectious disease and 76 patients with non-infectious.

Adult population analysisUsing the same model as for the paediatric populations (assuming α = 0.01, 1-β=0.2, P1=90%, P0=70% and a consensus rate of 65%) and assuming a viral to bacterial rate of 3:7 yields 390 patients with an infectious disease. Additionally, 76 non-infectious patients will be enrolled (based on similar calculations as for the paediatric population).

Methods for computing the accuracy of a multi-parametric model The multi-parametric model integrates various host-related and pathogen-related parameters into a single score that determines the likelihood of bacterial and viral aetiology (Figure 3). The score is computed using a multinomial logistic regression formula that is developed during Stage A of the study. Using predetermined cut-offs, each of the patients is classified as bacterial, viral, or inconclusive (i.e., a marginal immune response that is neither clearly bacterial nor viral). The model diagnosis will be compared with the consensus diagnosis in order to determine its accuracy. The diagnostic accuracy is quantified by computing the following measures: sensitivity, specificity, positive predictive value, negative predictive value, total accuracy, positive likelihood ratio, negative likelihood ratio and diagnostic odds ratio. The area under the receiver operation curve is computed to perform cut-off independent comparisons of different diagnostic methods.

D isc ussionThe inappropriate use of antibiotics has severe global health and economic consequences, including the emergence of antibiotic-resistant bacteria.33 A major driver of antibiotic misuse is the inability to accurately distinguish between bacterial and viral infections based on currently available diagnostic solutions.33 Many different biomarkers have been investigated in previous studies, none of them being accurate enough to establish or rule out bacterial infections.40 The TAILORED-Treatment study is a novel collaboration between (university) hospitals, academic institutions and

15873_CvHouten_BNW_v2.indd 135 26-11-18 12:43

Page 138: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

136

Chapter 6

(diagnostic) industry, aimed to collect and analyse ‘multi-omics’ data from patients suffering from LRTI and/or sepsis. Univariate and multivariate analysis of the data will generate new biomarkers to distinguish between bacterial, viral, and non-infectious disease. The multi-omics approach to generate infectious disease algorithms utilised by this study is currently unique within this area of medicine and provides a template for follow-up combined infectious disease data studies in the future. Some challenges remain for this type of study. First of all, as described previously, in a subset of patients, sampling of blood and nasal swabs at two sampling points (at presentation and after 3-5 days) should allow to measure data on the temporal dynamics of the host-pathogen interaction. However, if patients are not admitted to the hospital or are already discharged after one or two days, it is hard to collect a second sample. However, consecutive samples are not necessarily needed in order to achieve the primary objective. A second challenge is how to best interpret and efficiently act on the wealth of data available. Therefore, dedicated bioinformatics professionals are part of the TAILORED-Treatment consortium, for building the unique (HoPOIT) database and to develop the treatment algorithms. Finally, the study is a non-interventional study, and therefore, the diagnostic prediction of the model is not used for any clinical, diagnostic or prognostic decision making. After completing the study, utility studies will be needed to establish the clinical impact of any TAILORED-Treatment algorithms developed. In summary, the aim of this prospective international (TAILORED-Treatment) study is to develop algorithms to differentiate between bacterial and viral infections in children and adults with LRTI and/or sepsis. The study has the potential to improve patient care, reduce unnecessary antibiotic prescribing (thereby reducing the spread of antibiotic resistances) and will contribute positively to institutional, regional and national healthcare economics.

AcknowledgementThe authors would like to acknowledge the following co-workers on the TAILORED-Treatment project, who together with the authors are helping to realize the project goals: Racheli Kreisberg, Vincent Collins, Hans Hoogeveen, Erik Kristiansson, Anders Karlsson, Astrid Heikema, Deborah Horst-Kreft and Julio Font Perez.

15873_CvHouten_BNW_v2.indd 136 26-11-18 12:43

Page 139: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

137

6

The “TAILORED-Treatment” study

References1. Get Smart: Fast facts about antibiotic resistance. Available from: https://www.cdc.gov/

antibiotic-use/community/about/fast-facts.html

2. Davey P, Brown E, Fenelon L, Finch R, Gould I, Holmes A, Ramsay C, Taylor E, Wiffen P, Wilcox M: Systematic review of antimicrobial drug prescribing in hospitals. Emerging infectious diseases 2006, 12(2):211-216.

3. Linder JA, Stafford RS: Antibiotic treatment of adults with sore throat by community primary care physicians: a national survey, 1989-1999. JAMA 2001, 286(10):1181-1186.

4. Pulcini C, Cua E, Lieutier F, Landraud L, Dellamonica P, Roger PM: Antibiotic misuse: a prospective clinical audit in a French university hospital. Eur J Clin Microbiol Infect Dis 2007, 26(4):277-280.

5. Cadieux G, Tamblyn R, Dauphinee D, Libman M: Predictors of inappropriate antibiotic prescribing among primary care physicians. CMAJ 2007, 177(8):877-883.

6. CDC: Antibiotic Use in the United States, 2017: Progress and Opportunities. Available from: https://www.cdc.gov/antibiotic-use/stewardship-report/outpatient.html

7. Bartlett JG: Clinical practice. Antibiotic-associated diarrhea. N Engl J Med 2002, 346(5):334-339.

8. CDC: About Antimicrobial Resistance. Available from: https://www.cdc.gov/drugresistance/about.html (accessed 7-7-2017).

9. Arias CA, Murray BE: Antibiotic-resistant bugs in the 21st century--a clinical super-challenge. N Engl J Med 2009, 360(5):439-443.

10. FDA: Battle of the Bugs: Fighting Antibiotic Resistance. Available from: https://www.fda.gov/drugs/resourcesforyou/consumers/ucm143568.htm

11. O’Neill J. Review on Antimicrobial Resistance: Tackling drug-resistant infections globally: final report and recommendations. London 2016. Available from: https://amr-review.org/sites/default/files/AMR%20Review%20Paper%20%20Tackling%20a%20crisis%20for%20the%20health%20and%20wealth%20of%20nations_1.pdf (accessed 4-4-2018).

12. Battleman DS, Callahan M, Thaler HT: Rapid antibiotic delivery and appropriate antibiotic selection reduce length of hospital stay of patients with community-acquired pneumonia: link between quality of care and resource utilization. Arch Intern Med 2002, 162(6):682-688.

13. Craig JC, Williams GJ, Jones M, Codarini M, Macaskill P, Hayen A, Irwig L, Fitzgerald DA, Isaacs D, McCaskill M: The accuracy of clinical symptoms and signs for the diagnosis of serious bacterial infection in young febrile children: prospective cohort study of 15 781 febrile illnesses. BMJ 2010, 340:c1594.

14. Houck PM, Bratzler DW, Nsa W, Ma A, Bartlett JG: Timing of antibiotic administration and outcomes for Medicare patients hospitalized with community-acquired pneumonia. Arch Intern Med 2004, 164(6):637-644.

15. Houck PM, Bratzler DW, Niederman M, Bartlett JG: Pneumonia treatment process and quality. Arch Intern Med 2002, 162(7):843-844.

16. De Miguel-Yanes JM, Andueza-Lillo JA, Gonzalez-Ramallo VJ, Pastor L, Munoz J: Failure to implement evidence-based clinical guidelines for sepsis at the ED. The American journal of emergency medicine 2006, 24(5):553-559.

17. Metersky ML, Sweeney TA, Getzow MB, Siddiqui F, Nsa W, Bratzler DW: Antibiotic timing and diagnostic uncertainty in Medicare patients with pneumonia: is it reasonable to expect all patients to receive antibiotics within 4 hours? Chest 2006, 130(1):16-21.

15873_CvHouten_BNW_v2.indd 137 26-11-18 12:43

Page 140: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

138

Chapter 6

18. Little PS, Williamson I: Are antibiotics appropriate for sore throats? Costs outweigh the benefits. Bmj 1994, 309(6960):1010-1011.

19. Spiro DM, Tay KY, Arnold DH, Dziura JD, Baker MD, Shapiro ED: Wait-and-see prescription for the treatment of acute otitis media: a randomized controlled trial. Jama 2006, 296(10):1235-1241.

20. Little P: Delayed prescribing of antibiotics for upper respiratory tract infection. Bmj 2005, 331(7512):301-302.

21. Zwart S, Sachs AP, Ruijs GJ, Gubbels JW, Hoes AW, de Melker RA: Penicillin for acute sore throat: randomised double blind trial of seven days versus three days treatment or placebo in adults. Bmj 2000, 320(7228):150-154.

22. Fendrick AM, Monto AS, Nightengale B, Sarnes M: The economic burden of non-influenza-related viral respiratory tract infection in the United States. Arch Intern Med 2003, 163(4):487-494.

23. John JF, Jr., Fishman NO: Programmatic role of the infectious diseases physician in controlling antimicrobial costs in the hospital. Clin Infect Dis 1997, 24(3):471-485.

24. Study: Antibiotics problems cost U.S. between $17B and $26B a year Available from: http://www.bizjournals.com/triangle/stories/2009/10/19/daily4.html

25. Roberts RR, Hota B, Ahmad I, Scott RD, 2nd, Foster SD, Abbasi F, Schabowski S, Kampe LM, Ciavarella GG, Supino M et al: Hospital and societal costs of antimicrobial-resistant infections in a Chicago teaching hospital: implications for antibiotic stewardship. Clin Infect Dis 2009, 49(8):1175-1184.

26. WHO: Antibiotic resistance; frequently asked question. 2011. Available from: http://www.euro.who.int/en/health-topics/disease-prevention/antimicrobial-resistance/antibiotic-resistance/frequently-asked-questions (accessed 7-7-2017)

27. Gerber JS, Ross RK, Bryan M, Localio AR, Szymczak JE, Wasserman R, Barkman D, Odeniyi F, Conaboy K, Bell L et al: Association of Broad- vs Narrow-Spectrum Antibiotics With Treatment Failure, Adverse Events, and Quality of Life in Children With Acute Respiratory Tract Infections. Jama 2017, 318(23):2325-2336.

28. Sterling SA, Miller WR, Pryor J, Puskarich MA, Jones AE: The Impact of Timing of Antibiotics on Outcomes in Severe Sepsis and Septic Shock: A Systematic Review and Meta-Analysis. Crit Care Med 2015, 43(9):1907-1915.

29. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, Sevransky JE, Sprung CL, Douglas IS, Jaeschke R et al: Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013, 41(2):580-637.

30. Greenberg D, Broides A, Blancovich I, Peled N, Givon-Lavi N, Dagan R: Relative importance of nasopharyngeal versus oropharyngeal sampling for isolation of Streptococcus pneumoniae and Haemophilus influenzae from healthy and sick individuals varies with age. Journal of clinical microbiology 2004, 42(10):4604-4609.

31. Rhedin S, Lindstrand A, Rotzen-Ostlund M, Tolfvenstam T, Ohrmalm L, Rinder MR, Zweygberg-Wirgart B, Ortqvist A, Henriques-Normark B, Broliden K et al: Clinical utility of PCR for common viruses in acute respiratory illness. Pediatrics 2014, 133(3):e538-545.

32. Cruz AT, Demmler-Harrison GJ, Caviness AC, Buffone GJ, Revell PA: Performance of a rapid influenza test in children during the H1N1 2009 influenza a outbreak. Pediatrics 2010, 125(3):e645-650.

15873_CvHouten_BNW_v2.indd 138 26-11-18 12:43

Page 141: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

139

6

The “TAILORED-Treatment” study

33. Cohen A, Bont L, Engelhard D, Moore E, Fernandez D, Kreisberg-Greenblatt R, Oved K, Eden E, Hays JP: A multifaceted ‘omics’ approach for addressing the challenge of antimicrobial resistance. Future microbiology 2015, 10(3):365-376.

34. Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, Cohen J, Opal SM, Vincent JL, Ramsay G: 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Intensive care medicine 2003, 29(4):530-538.

35. Karlsson R, Gonzales-Siles L, Boulund F, Svensson-Stadler L, Skovbjerg S, Karlsson A, Davidson M, Hulth S, Kristiansson E, Moore ER: Proteotyping: Proteomic characterization, classification and identification of microorganisms--A prospectus. Systematic and applied microbiology 2015, 38(4):246-257.

36. Karlsson R, Davidson M, Svensson-Stadler L, Karlsson A, Olesen K, Carlsohn E, Moore ER: Strain-level typing and identification of bacteria using mass spectrometry-based proteomics. Journal of proteome research 2012, 11(5):2710-2720.

37. Gonzales-Siles L, Karlsson R, Kenny D, Karlsson A, Sjoling A: Proteomic analysis of enterotoxigenic Escherichia coli (ETEC) in neutral and alkaline conditions. BMC microbiology 2017, 17(1):11.

38. Karlsson R, Thorell K, Hosseini S, Kenny D, Sihlbom C, Sjoling A, Karlsson A, Nookaew I: Comparative Analysis of Two Helicobacter pylori Strains using Genomics and Mass Spectrometry-Based Proteomics. Frontiers in microbiology 2016, 7:1757.

39. Cohen JF, Korevaar DA, Altman DG, Bruns DE, Gatsonis CA, Hooft L, Irwig L, Levine D, Reitsma JB, de Vet HC et al: STARD 2015 guidelines for reporting diagnostic accuracy studies: explanation and elaboration. BMJ open 2016, 6(11):e012799.

40. Kapasi AJ, Dittrich S, Gonzalez IJ, Rodwell TC: Host Biomarkers for Distinguishing Bacterial from Non-Bacterial Causes of Acute Febrile Illness: A Comprehensive Review. PLoS One 2016, 11(8):e0160278.

41. Organization WH: Pocket Book of Hospital Care for Children. Geneva, Switzerland: World Health Organization; 2005.

42. Goldstein B, Giroir B, Randolph A: International pediatric sepsis consensus conference: definitions for sepsis and organ dysfunction in pediatrics. Pediatr Crit Care Med 2005, 6(1):2-8.

15873_CvHouten_BNW_v2.indd 139 26-11-18 12:43

Page 142: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

7

15873_CvHouten_BNW_v2.indd 140 26-11-18 12:43

Page 143: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

7Reproducibility of an expert panel diagnosis as reference standard in infectious diseases

CB van Houten, CA Naaktgeboren, L Ashkenazi-Hoffnung, S Ashkenazi, W Avis, I Chistyakov, T Corigliano, A Galetto, I Gangoiti, A Gervaix, D Glikman, I Ivaskeviciene, AA Kuperman, L Lacroix, Y Loeffen, F Luterbacher, CB Meijssen, S Mintegi, B Nasrallah, C Papan, AMC van Rossum, H Rudolph, M Stein, R Tal, T Tenenbaum, V Usonis, W de Waal, S Weichert, JG Wildenbeest, KM de Winter-de Groot, TFW Wolfs, N Mastboim, TM Gottlieb, A Cohen, K Oved, E Eden, PD Feigin, L Shani, and LJ Bont

Submitted

15873_CvHouten_BNW_v2.indd 141 26-11-18 12:43

Page 144: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

142

Chapter 7

Abstrac tBackground If a gold standard is lacking in a diagnostic test accuracy study, expert diagnosis is frequently used as reference standard. However, inter- and intra-observer agreements are imperfect. The aim of this study was to quantify the reproducibility of a panel diagnosis for paediatric infectious diseases.

MethodsPaediatricians from six countries adjudicated a diagnosis (i.e. bacterial infection, viral infection or undetermined) for febrile children. Diagnosis was reached when the majority of panel members came to the same diagnosis, leaving others inconclusive. We evaluated intra-observer and intra-panel agreement with six weeks and three years’ time intervals. We calculated the proportion of inconclusive diagnosis for a three-, five- and seven-expert panel.

ResultsFor both time intervals (i.e. six weeks and three years) intra-panel agreement was higher (kappa 0.88, 95%CI: 0.81-0.94 and 0.80, 95%CI: NA) compared to intra-observer agreement (kappa 0.77, 95%CI: 0.71-0.83 and 0.65, 95%CI: 0.52-0.78). After expanding the three-expert panel to five or seven experts, the proportion of inconclusive diagnoses (11%) remained the same.

ConclusionA panel consisting of three experts provides more reproducible diagnoses than an individual expert in children with lower respiratory tract infection or fever without source. Increasing the size of a panel beyond three experts has no major advantage.

15873_CvHouten_BNW_v2.indd 142 26-11-18 12:43

Page 145: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

143

7

Reproducibility of panel diagnosis

Introduc tionIntroduction of novel diagnostics into routine care is usually based on clinical validation studies. Evidence on the diagnostic accuracy is produced by comparing results of the test under evaluation (the index test, e.g. a newly developed blood test) with the actual presence or absence of a target condition. Ideally, a gold standard to determine the presence of this target condition is available, which is an error-free classification in all patients, blinded from the index test result, and performed within a short interval of time.1 There are, however, many conditions, such as certain infectious diseases2, for which such a gold standard does not exist. Using expert diagnosis as reference standard is therefore commonly applied in validation studies. The experts identify those with the target condition among the persons being tested, based on the available information.1,3 Several studies have assessed agreement between different experts and found a poor to moderate inter-observer agreement.4-10 Due to imperfect reproducibility of the expert diagnosis, the estimated accuracy of the diagnostic test under evaluation may be an over- or underestimation.11,12 Based on these results regulators and clinicians will decide to use novel diagnostics in clinical practice or not. To overcome these limitations, expert panels have been employed. However, to our knowledge, the reproducibility of such panel diagnoses in infectious diseases has not been examined. The current study was designed to quantify the reproducibility of an expert panel diagnosis for infectious diseases during childhood.

MethodsThis study had two primary objectives to assess reproducibility of an expert panel diagnosis. The first objective was to evaluate the reproducibility of an expert panel diagnosis in comparison to a single expert diagnosis. For this objective we calculated intra-observer and intra-panel agreement, comparing the diagnoses changed after six weeks and three years’ time intervals. The second objective was to evaluate the proportion of children in whom diagnosis changed after expanding the panel size (i.e. three, five or seven experts). We included uneven numbers of panel sizes as this is most commonly used in diagnostic studies.13 For both objectives we used data from our previously published OPPORTUNITY study.14 Briefly, we validated a host-protein based diagnostic using data from children aged 1 to 60 months presenting with lower respiratory tract infections or fever without source at the emergency department or on paediatric wards in the Netherlands and Israel.14

15873_CvHouten_BNW_v2.indd 143 26-11-18 12:43

Page 146: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

144

Chapter 7

Participating experts The IMPRIND consortium (‘Improving diagnostics in infectious diseases’) consisted of 25 paediatricians from six different European countries with at least 10 years of clinical experience since accredited as medical doctors. All experts were blinded to the diagnoses of their peers. Panel members did not receive other training than an explanation of the adjudication process and a review of paediatric, emergency medicine, and infectious disease literature written by the study team as background information without any instructions or decision rules. Before reviewing the cases, experts completed a short online survey on baseline characteristics (e.g. age, hours of clinical work per week).

Reviewing processEach expert reviewed two batches of up to 50 cases (i.e., in total there were a maximum of 100 patient records per expert). The review process, as well as the definitions employed, have been previously described.14 In short, each expert received an electronic Case Record Form (eCRF) including demographics, medical history, physical examination findings, all available radiological and laboratory data collected for routine care, microbiological investigation results (including study-specific nasal swab data), and a 28-day follow-up information. Based on the eCRF data, each expert assigned one of the following etiologies to each case: bacterial infection, viral infection, mixed infection (i.e., bacterial and viral co-infection), non-infectious disease, or indeterminate. Cases assigned as mixed infection were later classified as bacterial, because the clinical management is similar.

Case selection In total, 611 cases from the OPPORTUNITY study were available for the current study. Overall, case selection was based on a convenient sample size. For the first objective, evaluating the reproducibility of an expert panel diagnosis in comparison to a single expert diagnosis, we used two different time intervals (i.e. six weeks and three years, Figure 1). Randomly selected cases were used to calculate intra-observer agreement. After six weeks or three years the same cases were diagnosed by the same experts. To quantify reproducibility of panel diagnosis, a random selection of cases was diagnosed by three experts and was used to calculate intra-panel agreement (Figure 1). To minimize the possibility that experts had recognized cases from their first assignment (i.e., six weeks or three years ago), we did not inform the experts about the objective to measure reproducibility. For the second objective, i.e. effect of panel size on reproducibility, case selection was performed in two ways (Figure 1). First, we took a random selection of available cases. In the OPPORTUNITY study, 70% of the cases were assigned an unanimous panel diagnosis. To avoid overestimation of the reproducibility, we added ‘harder to diagnose’ cases with a majority (2 out of 3 experts agreed on the diagnosis) or

15873_CvHouten_BNW_v2.indd 144 26-11-18 12:43

Page 147: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

145

7

Reproducibility of panel diagnosis

inconclusive diagnosis, thus excluding unanimous diagnosis.14 Cases were assigned an inconclusive diagnosis if each panel member assigned a different diagnosis or when at least two panel members diagnosed the case as indeterminate. The selected cases were reviewed by at least seven experts allowing analyses of different panel compositions.

611 Opportunity cases available

Objective 1Intra-observer and

intra-panel agreement

Objective 2Panel size analyses

6 week time-interval

3 year time-interval

Intra-observer agreement

99 cases

Intra-panel agreement

63 cases

Random selection

Random selection

Intra-observer agreement224 cases

Intra-panel agreement113 cases

Random selection

Random selection

193 cases

Random selection (n=138) plus selection of difficult to

diagnose cases (n=55)*

* Majority or inconclusive consensus in Opportunity study. Majority consensus was defined if 2 out of 3 experts agreed on the diagnosis, inconclusive diagnosis if each panel member assigned a different diagnosis and when at least two panel members diagnosed the case as indeterminate.

Figure 1. Case selection.Case selection to assess reproducibility of panel diagnoses. Objective 1: to compare reproducibility of individual expert versus expert panel diagnosis. Objective 2: to compare diagnosis and agreement for expert panels of different size (i.e. three, five or seven experts). Case selection was based on a convenient sample size.

Statistical analysisIntra- and inter-observer and intra- and inter-panel agreements were calculated with Fleiss kappa statistics (κ). Kappa statistics measure the observed level of agreement between diagnoses for a set of nominal ratings and corrects for agreement that would be expected by chance.15 The degree of agreement was rated as suggested by Landis and Koch: fair (0.21-0.40), moderate (0.41-0.60), substantial (0.61-0.80), or almost perfect

15873_CvHouten_BNW_v2.indd 145 26-11-18 12:43

Page 148: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

146

Chapter 7

(0.81-1.0).16 The value of kappa statistics is a relative measure, while clinician’s question of reference standard variation calls for an absolute measure.11 Therefore, we also calculated absolute agreement. In addition, we calculated the proportion of bacterial, viral and inconclusive diagnoses and the alterations in diagnosis for all possible panel compositions if the panel size increased (i.e. three, five or seven experts). To analyze the reproducibility of different panel sizes, we calculated inter-observer and inter-panel agreement for all possible panel compositions (e.g. 193 patients x 35 possible panels when constructing a panel with three members from seven options). Analyses of inter-panel agreement was performed for three- and five-expert panels, each panel consists of three and five unique panel members. When calculating the proportion of bacterial, viral and inconclusive diagnoses, and the intra-panel and inter-panel agreement, the majority diagnosis was used. Majority diagnosis was defined as at least two out of three, three out of five or five out of seven panel members agreeing on a diagnosis. We used R version 3.4.4 for the statistical analysis.

ResultsExperts and cases characteristicsTwenty-five paediatricians from six different countries participated in the study, of who the majority had special interest in infectious diseases (Supplemental Table 1). Thirteen experts (52%) had participated in an expert panel for a clinical study in the past. The experts made a total of 2375 individual diagnoses. In total, 355 unique cases were used with some cases being used more than once for the different objectives. The median age of cases used for objective 1 (i.e. intra-observer vs intra-panel agreement) was 14 months (Table 1). Cases used for objective 2 (i.e. reproducibility for different panel sizes) were slightly older (median age 18 months). In both groups approximately 60% were male (Table 1).

Intra-observer and intra-panel agreementAs shown in Figure 2, when the time interval was six weeks, intra-observer agreement was substantial (mean κ=0.77, 95% CI: 0.71-0.83), when using Landis’ cut-off values. When a three-expert panel was employed, with the diagnosis defined by majority, reproducibility was higher with an almost perfect agreement (mean κ=0.88, 95% CI: 0.81-0.94). Intra-panel agreement with a three year time interval was also higher in comparison to intra-observer agreement (mean κ= 0.80, 95% CI: NA and mean κ= 0.65, 95% CI: 0.52-0.78, respectively). Similar to kappa statistics, the absolute proportion of intra-panel agreement was higher compared to the intra-observer agreement for both six weeks and three years’ time intervals (Table 2). Intra-observer agreement as well as intra-panel agreement was modestly higher when the time interval between

15873_CvHouten_BNW_v2.indd 146 26-11-18 12:43

Page 149: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

147

7

Reproducibility of panel diagnosis

assessments was shorter (six weeks versus three years, Table 2). We constructed reclassification tables to obtain deeper insight into diagnostic changes (Supplemental Table 2). With a six weeks’ time interval single expert diagnoses changed in 10% of the individual cases, and panel diagnoses in 5% of the cases. For a three years’ time interval these percentages were respectively 15% and 10%.

Table 1. Baseline characteristics of the included cases.Objective 1

(n=296)Objective 2

(n=193)

Age (months) (median, IQR) 14(7-30) 18(10-31)

Gender (male) (n,%) 175(59%) 117(61%)

Max. temperature (°C) (median, IQR) 39.3(38.8-40.0) 39.5(39.0-40.0)

Duration of symptoms (d) (median, IQR) 2(1-3) 2(1-4)

Hospital admission (n,%) 185(63%) 127(66%)

Hospitalization duration (d) (median, IQR) 3(2-5) 3(2-5)

Antibiotics prescribed (n,%) 164(55%) 108(56%)

CRP (mg/L) (median, IQR) 21(7-58) 27(10-62)

Need of mechanical ventilation (n,%) 2(1%) 3(2%)

Recruiting site

Secondary care centre (n,%) 260(88%) 169(87)

Tertiary care centre (n,%) 33(11%) 19(10%)

PICU (n,%) 3(1%) 5(3%)

Clinical syndrome

URTI (n,%) 89(30%) 57(29%)

FWS (n,%) 75(25%) 46(24%)

LRTI (n,%) 66(22%) 55(29%)

Other (n,%) 66(22%) 35(18%)

Objective 1: to compare reproducibility of individual expert versus expert panel diagnosis. Objective 2: to compare diagnosis and agreement for expert panels of different size (i.e. three, five or seven experts). IQR: interquartile range, CRP: C-reactive protein, PICU: paediatric intensive care unit, FWS: fever without source, URTI: upper respiratory tract infection, LRTI: lower respiratory tract infection.

Table 2. Absolute proportion of intra-observer and intra-panel agreements.

% agreement(min-max)

Number of expert (or panels)

6 weeks interval

Intra-observer agreement, mean (min-max) 91% (85-94%) 6 experts

Intra-panel agreement, mean (min-max) 95% (94%-97%) 2 panels

3 years interval

Intra-observer agreement, mean (min-max) 84% (76-92%) 6 experts

Intra-panel agreement 90% 1 panel

Presented percentages are mean proportions with minimum and maximum values from different experts or three-expert panels.

15873_CvHouten_BNW_v2.indd 147 26-11-18 12:43

Page 150: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

148

Chapter 7

Figure 2. Box plot for the intra-observer and intra-panel agreements. Each dot corresponds to the kappa per expert or per three-expert panel. In box plots, the band inside the box represents the median kappa. The bottom and top of the box represent the fi rst and third quartiles. The whiskers refl ect the minimum and maximum kappa.

Panel size The overall proportion of inconclusive diagnoses for a panel of three experts, using majority consensus, was 11%; this proportion did not decrease when experts were added to the panel (Figure 3). Moreover, the overall proportion of bacterial and viral diagnoses did not change when the panel was expanded to fi ve members. Nevertheless, the diagnosis changed in 11% of the individual cases when the size of the panel increased from three to fi ve experts and in 10% when expanding the panel size from fi ve to seven experts. Inter-observer agreement within a panel of three experts was fair with a mean kappa of 0.39 (95% CI: 0.38-0.40). Inter-observer agreement did not improve when the panel was expanded to fi ve experts (Supplemental Figure 1). Inter-panel agreement

15873_CvHouten_BNW_v2.indd 148 26-11-18 12:43

Page 151: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

149

7

Reproducibility of panel diagnosis

between diff erent panels was substantial (κ=0.75, 95% CI: 0.74-0.75) using the majority diagnosis. Inter-panel agreement was higher (κ=0.86, 95% CI: 0.85-0.86) when the three-expert panels were expanded to fi ve experts (majority diagnosis defi ned as at least three out of fi ve panel members agreed on a diagnosis).

Figure 3. Expert panel diagnosis using panels of diff erent sizeThe waves show the change in diagnoses if the panel was expanded from three to fi ve and seven experts (e.g. panel size analysis). Light green waves; cases with a viral diagnosis by a panel existing of three experts, purple waves; cases with an inconclusive diagnosis by a panel existing of three experts, dark green waves: cases with a bacterial diagnosis by a panel existing of three experts. Overall, diagnosis changed in 11% of the individual cases when the size of the panel increased from three to fi ve experts and in 10% when expanding the panel size from fi ve to seven experts.

D isc ussionIn the present study a panel consisting of three experts as a reference standard in a diagnostic infectious diseases study provided more reproducible diagnoses than individual expert’s diagnoses and showed a high level of intra-panel agreement.

15873_CvHouten_BNW_v2.indd 149 26-11-18 12:43

Page 152: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

150

Chapter 7

Expanding the panel to five or seven experts led to a substantial change in diagnoses and did not lead to a decrease in the number of inconclusive diagnoses.

A review by Tuijn et al. evaluated the agreement between health care professionals and examined the effectiveness of planned interventions for improving reproducibility. They found overall a fair agreement (κ=0.31) between health care professionals before implementing various interventions.17 This agreement is in line with ours and other studies which found a poor to moderate interrater agreement.4-10 Several radiological and histological studies evaluated the intra-observer agreement. Most of these studies found high level of agreement.18-22 However, it should be noted that these studies used relative short time intervals (i.e. several weeks) to assess intra-observer agreement. In our study intra-observer agreement was lower for the three years’ time interval compared to the six years’ time interval. This result shows that it is crucial to mention the time interval when presenting intra-observer agreements. None of the previously mentioned studies evaluated the reproducibility of individual expert diagnosis compared to a panel. We found only two, non-infectious diseases related studies that compared single observer diagnoses with panel diagnoses.23,24 Both studies concluded, similarly to the findings in our study, that the reproducibility and accuracy of the diagnosis was higher for panel diagnosis and recommended this approach at least for research purposes.

Our findings may be valuable for researchers, but also for organizations as the Food and Drug Administration (FDA) and European Medicines Agency (EMA), as these organizations approve drugs related to outcomes based on studies using single expert assessments.25,26 However, our results demonstrated that if a panel diagnosis was used instead of individual expert diagnosis, the outcome of a study (i.e. the effect of the drug) might have changed significantly.

The major strength of our study is the comprehensive procedure of assessing reproducibility of a panel diagnosis in infectious diseases. To our knowledge, this is the first study that evaluated intra-observer agreement with such a long time-interval (i.e. three years) and that examined the effect of expanding the size of the panel from three to seven experts. A second strength worth mentioning, is the prospective character of the OPPORTUNITY study, from which cases were derived, leading to complete, longitudinal and high-quality data for every case.

Limitations of our study should also be discussed. First, we cannot exclude that experts included in the intra-observer and intra-panel analyses recognized cases from their first review. Obviously, the shorter the time interval, the more likely it is to recognize cases and to remember the previously assigned diagnosis. Although we have changed

15873_CvHouten_BNW_v2.indd 150 26-11-18 12:43

Page 153: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

151

7

Reproducibility of panel diagnosis

the study numbers, this may have contributed to the higher intra-observer and intra-panel agreement with the six weeks interval compared to the three years’ time interval. Second, we presented an arbitrary set of data to the experts. We used the same eCRF as in our, previously published, validation study of a new diagnostic. Third, there are several aspects of panel diagnosis we didn’t evaluate in the current study. For example, the correlations between expert characteristics (i.e., experience years and country of origin) or eCRF variables (including specific biomarkers) and outcome. Fourth, the sample-size estimation was based on a convenient sample size. We feel this was acceptable according to the rule of thumb that 50–100 patients are needed in order to obtain an adequate power for a study on reproducibility.27 Due to logistical reasons, intra-panel agreement could only be calculated for a limited number of panels. Fifth, the review process of the cases by the experts required a significant effort from the expert panel members. Therefore, it was not feasible to have all 193 cases reviewed by the same seven experts for the second objective. This non-uniformity in the cases experts classified possibly impacted the results. Sixth, in our study individual diagnosis changed in 11% of the cases when the size of the panel increased from three to five experts and in 10% when expanding the panel size from five to seven experts. We consider this as acceptable, as the proportion of inconclusive cases did not change. However, these differences might still have significant impact on the sensitivity and specificity of the panel diagnosis. Finally, given the wide variety of consensus panels, our findings cannot be generalized to non-infectious patients.

In conclusion, this study suggests that a panel consisting of three experts provides more reproducible diagnosis than an individual expert, while increasing the size of experts’ panel further has no major advantage. In the absence of a gold standard to differentiate viral from bacterial infections in children, a panel diagnosis of experts, instead of the diagnosis of a single expert, is therefore highly preferred as a reference standard. Further prospective validation and optimization of panel diagnosis is needed to obtain accurate results of diagnostic studies without a gold standard.

15873_CvHouten_BNW_v2.indd 151 26-11-18 12:43

Page 154: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

152

Chapter 7

References1. Reitsma JB, Rutjes AW, Khan KS, Coomarasamy A, Bossuyt PM. A review of solutions for

diagnostic accuracy studies with an imperfect or missing reference standard. J Clin Epidemiol 2009; 62(8): 797-806.

2. Lynch T, Bialy L, Kellner JD, et al. A systematic review on the diagnosis of pediatric bacterial pneumonia: when gold is bronze. PLoS One 2010; 5(8): e11989.

3. Rutjes AW, Reitsma JB, Coomarasamy A, Khan KS, Bossuyt PM. Evaluation of diagnostic tests when there is no gold standard. A review of methods. Health Technol Assess 2007; 11(50): iii, ix-51.

4. DiGiorgio MJ, Vinski J, Bertin M, Sun Z, Bena JF, Albert NM. Single-center study of interrater agreement in the identification of central line-associated bloodstream infection. American journal of infection control 2014; 42(6): 638-42.

5. McBryde ES, Brett J, Russo PL, Worth LJ, Bull AL, Richards MJ. Validation of statewide surveillance system data on central line-associated bloodstream infection in intensive care units in Australia. Infection control and hospital epidemiology 2009; 30(11): 1045-9.

6. Bada C, Carreazo NY, Chalco JP, Huicho L. Inter-observer agreement in interpreting chest X-rays on children with acute lower respiratory tract infections and concurrent wheezing. Sao Paulo medical journal = Revista paulista de medicina 2007; 125(3): 150-4.

7. Fischer JE, Seifarth FG, Baenziger O, Fanconi S, Nadal D. Hindsight judgement on ambiguous episodes of suspected infection in critically ill children: poor consensus amongst experts? European journal of pediatrics 2003; 162(12): 840-3.

8. Greenwald PW, Schaible DD, Ruzich JV, Prince SJ, Birnbaum AJ, Bijur PE. Is single observer identification of wound infection a reliable endpoint? The Journal of emergency medicine 2002; 23(4): 333-5.

9. Loeb MB, Carusone SB, Marrie TJ, et al. Interobserver reliability of radiologists’ interpretations of mobile chest radiographs for nursing home-acquired pneumonia. Journal of the American Medical Directors Association 2006; 7(7): 416-9.

10. Klompas M. Interobserver variability in ventilator-associated pneumonia surveillance. American journal of infection control 2010; 38(3): 237-9.

11. de Vet HC, Mokkink LB, Terwee CB, Hoekstra OS, Knol DL. Clinicians are right not to like Cohen’s kappa. Bmj 2013; 346: f2125.

12. Gauvin F, Dassa C, Chaibou M, Proulx F, Farrell CA, Lacroix J. Ventilator-associated pneumonia in intubated children: comparison of different diagnostic methods. Pediatr Crit Care Med 2003; 4(4): 437-43.

13. Bertens LC, Broekhuizen BD, Naaktgeboren CA, et al. Use of expert panels to define the reference standard in diagnostic research: a systematic review of published methods and reporting. PLoS Med 2013; 10(10): e1001531.

14. van Houten CB, de Groot JA, Klein A, et al. A host-protein based assay to differentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study. Lancet Infect Dis 2017; 17(4): 431-40.

15. Hallgren KA. Computing inter-rater reliability for observational data: an overview and tutorial. Tutorials in quantitative methods for psychology 2012; 8(1): 23-34.

16. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics 1977; 33(1): 159-74.

15873_CvHouten_BNW_v2.indd 152 26-11-18 12:43

Page 155: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

153

7

Reproducibility of panel diagnosis

17. Tuijn S, Janssens F, Robben P, van den Bergh H. Reducing interrater variability and improving health care: a meta-analytical review. Journal of evaluation in clinical practice 2012; 18(4): 887-95.

18. Muhlhofer HM, Lenze U, Lenze F, et al. Inter- and intra-observer variability in biopsy of bone and soft tissue sarcomas. Anticancer research 2015; 35(2): 961-6.

19. van Buijtenen JM, van Tunen ML, Zuidema WP, et al. Inter- and intra-observer agreement of the AO classification for operatively treated distal radius fractures. Strategies in trauma and limb reconstruction (Online) 2015; 10(3): 155-9.

20. Imerci A, Aydogan NH, Tosun K. Evaluation of inter- and intra-observer reliability of current classification systems for subtrochanteric femoral fractures. European journal of orthopaedic surgery & traumatology : orthopedie traumatologie 2018; 28(3): 499-502.

21. Okizaki A, Nakayama M, Nakajima K, et al. Inter- and intra-observer reproducibility of quantitative analysis for FP-CIT SPECT in patients with DLB. Annals of nuclear medicine 2017; 31(10): 758-63.

22. Ominde M, Sande J, Ooko M, et al. Reliability and validity of the World Health Organization reading standards for paediatric chest radiographs used in the field in an impact study of Pneumococcal Conjugate Vaccine in Kilifi, Kenya. PLoS One 2018; 13(7): e0200715.

23. Stroink H, van Donselaar CA, Geerts AT, et al. Interrater agreement of the diagnosis and classification of a first seizure in childhood. The Dutch Study of Epilepsy in Childhood. Journal of neurology, neurosurgery, and psychiatry 2004; 75(2): 241-5.

24. Gabel MJ, Foster NL, Heidebrink JL, et al. Validation of consensus panel diagnosis in dementia. Arch Neurol 2010; 67(12): 1506-12.

25. Meltzer HY, Risinger R, Nasrallah HA, et al. A randomized, double-blind, placebo-controlled trial of aripiprazole lauroxil in acute exacerbation of schizophrenia. The Journal of clinical psychiatry 2015; 76(8): 1085-90.

26. Citrome L, Du Y, Risinger R, et al. Effect of aripiprazole lauroxil on agitation and hostility in patients with schizophrenia. International clinical psychopharmacology 2016; 31(2): 69-75.

27. Terwee CB, Mokkink LB, Knol DL, Ostelo RW, Bouter LM, de Vet HC. Rating the methodological quality in systematic reviews of studies on measurement properties: a scoring system for the COSMIN checklist. Quality of life research : an international journal of quality of life aspects of treatment, care and rehabilitation 2012; 21(4): 651-7.

15873_CvHouten_BNW_v2.indd 153 26-11-18 12:43

Page 156: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

154

Chapter 7

S upplementar y materialSupplemental Table 1. Baseline characteristics of expert

Experts(n=25)

Gender (male) (n,%) 13(52)

Country

The Netherlands (n,%) 8(32)

Israel (n,%) 7(28)

Switzerland (n,%) 4(16)

Germany (n,%) 3(12)

Lithuania (n,%) 2(8)

Spain (n,%) 1(4)

Age

30-39 years (n,%) 5(20)

40-49 years (n,%) 9(36)

50-59 years (n,%) 8(32)

≥ 60 years (n,%) 3(12)

Clinical experience*

10-19 years (n,%) 10(40)

≥ 20 years (n,%) 15(60)

Special interest

Infectious diseases (n,%) 15(60)

Emergency Medicine (n,%) 4(16)

Pulmonology (n,%) 1(4)

Other (n,%) 5(20)

Hours per week working as clinician

10-19 hours (n,%) 4(16)

20-29 hours (n,%) 4(16)

30-39 hours (n,%) 4(16)

≥ 40 hours (n,%) 13(52)

Participated in an expert panel before

No (n,%) 12(48)

Yes, once (n,%) 7(28)

Yes, more than once (n,%) 6(24)

* Since accredited as medical doctors

15873_CvHouten_BNW_v2.indd 154 26-11-18 12:43

Page 157: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

155

7

Reproducibility of panel diagnosis

Supplemental Table 2. Reclassification of expert and panel diagnoses over timea.

Expert diagnoses(n=284) in 99 cases

Assignment 6 weeks later

Bacterial Viral Inconclusive

First assignment Bacterial 54 4 2

Viral 3 191 9

Inconclusive 6 4 11

b.

Panel diagnoses(n=63) in 63 cases

Assignment 6 weeks later

Bacterial Viral Inconclusive

First assignment Bacterial 14 1 0

Viral 0 46 1

Inconclusive 1 0 0

c.

Expert diagnoses(n=449) in 224 cases

Assignment 3 years later

Bacterial Viral Inconclusive

First assignment Bacterial 65 6 11

Viral 5 290 25

Inconclusive 8 14 25

d.

Panel diagnoses(n=113) in 113 cases

Assignment 3 years later

Bacterial Viral Inconclusive

First assignment Bacterial 19 0 3

Viral 0 73 4

Inconclusive 2 2 10

If cases had been diagnosed by multiple experts or panels the diagnoses were taken together. a. Expert reclassification six weeks’ time interval, b. Three-expert panel reclassification six weeks’ time interval, c. Expert reclassification three years’ time interval, d. Three-expert panel reclassification three years’ time interval.

15873_CvHouten_BNW_v2.indd 155 26-11-18 12:43

Page 158: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

156

Chapter 7

Supplemental Figure 1. Box plot for the inter-observer agreement for a three- and fi ve-expert panel. Each dot represents the kappa value for inter-observer agreement within a panel. All possible panel compositions were analyzed.

15873_CvHouten_BNW_v2.indd 156 26-11-18 12:43

Page 159: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 157 26-11-18 12:43

Page 160: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

8

15873_CvHouten_BNW_v2.indd 158 26-11-18 12:43

Page 161: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

8A new diagnostic strategy for children with lower respiratory tract infections

CB van Houten and LJ Bont

Nederlands tijdschrift voor Medische Microbiologie,2018;26:nr3

15873_CvHouten_BNW_v2.indd 159 26-11-18 12:43

Page 162: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

160

Chapter 8

Abstrac tLower respiratory tract infections (LRTIs) occur frequently, especially in children below the age of five. Approximately 75% of these infections in children have a viral aetiology, however, about 25% of these viral infections are treated with antibiotics. This antibiotic overuse shows that the diagnostic process of children with LRTIs is a difficult process for physicians. Due to the increasing antimicrobial resistance, correct prescription of antibiotics becomes even more important. It is therefore not surprising that there have been several developments in the field of diagnostic tests, especially in biomarkers to differentiate between viral and bacterial infections. This article shows a recent overview and provides a recommendation for diagnostics in children with LRTIs that could guide physicians prescribing antibiotics. For children with an uncomplicated LRTI, diagnostics and antimicrobial treatment are not indicated. If antibiotic treatment is considered, measuring a combination of viral and bacterial biomarkers can be helpful in ruling out a bacterial infection and thereby reduce unnecessary antibiotic use.

15873_CvHouten_BNW_v2.indd 160 26-11-18 12:43

Page 163: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

161

8

A new diagnostic strategy

Introduc tionLower respiratory tract infections (LRTIs) are among the most common acute clinical diseases in paediatrics. Worldwide, 15% of the children under the age of five die from pneumonia, making it the most common infectious cause of death in children.1 In the Netherlands, the risk of death is low, partly due to improvements in supportive care and the introduction of the National Vaccination Program. However, in the Netherlands, LRTIs still cause high morbidity with a national incidence in children below the age of five of 70 per 1,000 persons.2 A LRTI is a diagnosis made on the basis of symptoms of fever, cough and increased respiratory effort caused by inflammation of the lower airways, rather than using diagnostic tests. This definition includes bronchiolitis and typical and atypical pneumonia. Approximately 75% of the respiratory infections in children have a viral cause, including Respiratory Syncytial Virus (RSV), Human Metapneumovirus, Adenovirus, Bocavirus or Parainfluenza virus.3 The typical bacteria that are the most common cause of pneumonia are Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis. Because most LRTIs have a viral origin, a cautious policy can often be applied for antibiotic prescription. Nevertheless, about a quarter of all viral infections are treated with antibiotics.3 This overuse of antibiotics shows that it is hard for physicians to diagnose children with LRTIs. Given the increasing antibiotic resistance among respiratory pathogens, the importance of prescribing antibiotics correctly has become increasingly important. Therefore, it is not surprising that in recent years there have been several developments in the field of diagnostic tests, mainly focussing on biomarkers, to distinguish between viral and bacterial infections. This article provides a recent overview and recommendation for diagnostics in children with LRTIs, and could be used as a guideline for physicians. The article focuses primarily on children with LRTI presenting at the hospital, but can certainly be applied in outpatient settings by general practitioners.

The diagnostic processClinical featuresWhen a child presents with fever, signs of increased respiratory effort (chest wall retractions, use of respiratory muscles, nasal flaring) and an increased respiratory rate, there is a substantial chance of a pneumonia.4 Unfortunately, it is hard to differentiate between viral and bacterial pathogens based on clinical signs and symptoms.5,6 Clinical symptoms are important when estimating the severity of the infection. Therefore, the severity criteria shown in table 1 could be helpful.7

15873_CvHouten_BNW_v2.indd 161 26-11-18 12:43

Page 164: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

162

Chapter 8

Radiological diagnosticsIn many epidemiological studies, the result of a chest X-ray is an important criterion for classifying pneumonia. A chest X-ray is therefore regularly made in children with LRTIs. Although, often the result has no influence on treatment in clinical practice. First, studies show that the inter-observer agreement is imperfect in reporting radiological abnormalities in young children with pneumonia, consequentially, it is difficult to make the correct diagnosis.8 In addition, radiological abnormalities are difficult to correlate with the aetiology of the infection. It is thought that alveolar infiltrates are more suited to bacterial pathogens. However, several studies show that alveolar infiltrates also occur in 40-50% of viral LRTIs.9,10 Therefore, it can be concluded that a chest X-ray does not contribute to determining the pathogen of a pneumonia11, and that there is thus no indication for making a chest X-ray in children with an uncomplicated LRTI. A chest X-ray should be considered if there is suspicion of a recurrent or a complicated pneumonia (pleural effusion or lung abscess). Several studies investigated the diagnostic value of ultrasound in children with the suspicion of pneumonia. A recent study emphasizes that there are (yet) no standardized methods for the use in children and that ultrasound as a diagnostic tool for interstitial pneumonia is still inaccurate.11,12

Table 1. Criteria to estimate the severity of a pneumonia.

Mild to moderate severe pneumonia Severe pneumonia

Children<2 year

Respiratory rate <50/minute Respiratory rate >70/minute

Mild chest wall retractions Mild to severe chest wall retractions, moaning and /or nasal flaringApnoea

Saturation ≥ 92% Cyanosis, saturation <92%

Normal heart rate* Tachycardia*

Capillary refill <2 sec Capillary refill ≥ 2 sec

Normal intake Does not drink anymore

Temperature <38.5 ˚C Temperature ≥ 38.5 ˚C

Children ≥ 2 year

Respiratory rate <50/minute Respiratory rate >50/minute

Mild dyspnoea Severe dyspnoea, nasal flaring and/or moaning breathing

Saturation ≥ 92% Cyanosis, saturation <92%

Normal heart rate* Tachycardia*

Capillary refill <2 sec Capillary refill ≥ 2 sec

Normal intake Signs of dehydration, vomiting

Temperature <38.5 ˚C Temperature ≥ 38.5 ˚C

* Corrected for age and temperature.

15873_CvHouten_BNW_v2.indd 162 26-11-18 12:43

Page 165: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

163

8

A new diagnostic strategy

Microbiological diagnosticsIn adults with a LRTI, a sputum culture is frequently used to identify the pathogen and to adjust the antibiotics policy based on the culture results. In young, non-intubated, children it is not possible to obtain an adequate sputum sample. A nasopharyngeal swab for Polymerase Chain Reaction (PCR) testing is often taken as an alternative. However, children have nasopharyngeal colonization with the same pathogens that also cause pneumonia. Therefore, in the interpretation of positive test results of both the viral PCR and the PCR on Mycoplasma pneumoniae, asymptomatic carriage should be taken into account.13 For RSV and Influenza there is strong evidence of causal attribution in case of LRTI, thus PCR for these viruses is more useful than for other viral pathogens. Therefore, Influenza and RSV PCR is recommended during the endemic season in admitted children with an uncomplicated LRTI, if a proven Influenza or RSV infection is needed for isolation precautions.14,15 In addition, one must realize that a positive viral PCR does not exclude a bacterial superinfection. Pneumococcal antigen detection in urine in children is also often positive due to carriage in the upper airways.16 Several studies evaluated the effect of rapid tests on antibiotic use, they found conflicting results.17-20 A recent study shows that physicians prescribe antibiotics more often for non-RSV infected children compared to children with RSV.21 Blood cultures can detect pathogens of a bacterial pneumonia. Unfortunately, the sensitivity of a blood culture in pneumonia is low, because a large proportion of the patients with pneumonia do not have bacteraemia and the results are often influenced by previous antibiotic treatment.22,23 Therefore, microbiological testing is not recommended in children with a mild LRTI, unless detection of the pathogen is important for the isolation precautions in the ward.24 Several researchers are currently studying the microbiome. It is known that once the balance in the microbiome is disturbed pathogenic bacteria have the chance to spread to the lungs and cause LRTI.25 Currently, little is known about the microbiome during infection in children with LRTIs26, but in the future the microbiome of the patient may play a role in (personalized) antibiotic treatment.27

Markers of infection in blood A recent study shows that laboratory measurements (e.g. C-reactive protein (CRP) and leukocyte count) are strong drivers of antibiotic prescribing in children with LRTIs.28 However, it is also well known that the current diagnostic tests do not differentiate accurately between viral and bacterial LRTIs and there is an urgent need for new, reliable biomarkers. Therefore, both at the level of fundamental research and at the level of the pharmaceutical industry there is an increasing effort to develop point of care diagnostic tests using easy-to-measure host biomarkers to assist physicians in correct prescription

15873_CvHouten_BNW_v2.indd 163 26-11-18 12:43

Page 166: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

164

Chapter 8

of antibiotics. Unfortunately, many of these biomarkers have not (yet) been studied in children, and if this is the case, conflicting results are often found, mainly caused by different cut-off values and uncertainties in the gold standard. Consequentially, these biomarkers cannot be leading in the decision to start antibiotics or not. Below we will discuss the most studied and the most promising, new markers for children with LRTIs (Table 2).

Haematological markersInitially, haematological biomarkers such as erythrocyte sedimentation rate and leukocyte and neutrophil count were used to differentiate between viral and bacterial LRTIs. However, literature shows that these biomarkers do not differentiate accurately, because both sensitivity and specificity are too low to apply these haematological markers in clinical practice. Even when these markers are combined, no reliable distinction can be made between viral and bacterial pneumonia.29-31

Acute phase proteinsCRP is one of the most used biomarkers in infectious diseases. However, there are several factors that complicate the interpretation of CRP concentrations (e.g. low specific character, large “grey area” and the fact that the peak concentration is only reached after 1-2 days). In addition, a review shows that the diagnostic accuracy of CRP in children with pneumonia is limited.30 In recent years, many clinical studies have investigated the role of procalcitonin (PCT). The dynamics of PCT are similar to those of CRP, but where the peak concentration of CRP is seen only after approximately 36 hours, PCT reaches a peak concentration after 8 hours.32 A randomized controlled trial from Switzerland showed that the use of PCT in children with LRTIs does not affect the antibiotic prescription rate, but does reduce the duration of the antibiotic treatment.33 These findings are in line with other studies, which show that PCT has mainly added value in determining the duration of antibiotic treatment (especially in neonatal- and paediatric intensive care units). However, like CRP, it does not differentiate accurately between viral and bacterial pathogens to base antibiotic treatment on its concentration.

Cytokine markersSeveral cytokine markers (both pro- and anti-inflammatory) have been studied in adult patients, but research on these markers in children is limited. Recently, researchers showed that the concentrations of various cytokines (including various interleukins and Tumour Necrosis Factor alpha) differ significantly between viral and bacterial infections in children with fever.34 Interleukine 6 (IL-6) appears to be most promising: in many studies this marker reaches a significantly higher concentration in children

15873_CvHouten_BNW_v2.indd 164 26-11-18 12:43

Page 167: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

165

8

A new diagnostic strategy

with bacterial infections compared to viral infections. One study found a sensitivity and negative predictive value of 100%, a specificity of 99% and a positive predictive value of 98% when IL-6 was used in children to differentiate between pneumonia caused by RSV and Mycoplasma pneumoniae.35 However, more research is needed before this biomarker can be used in all children with LRTIs.

Cell surface markersIn children, the most researched cell surface biomarker is CD64. This immunoglobulin receptor is expressed on polymorphonuclear neutrophils (PMN), and its expression seems to increase as PMNs are activated by bacterial infections. A review shows that this marker can therefore be of value in distinguishing viral from bacterial infections in children. However, all included studies were of low methodological quality and analysed febrile patients with a wide variety of clinical syndromes.29 Hereafter, another study confirmed the diagnostic value of CD64 specifically in children with LRTIs. They found an area under the ROC curve of 0.90 (95% confidence interval: 0.83-0.98) for the discriminative power of this marker in paediatric LRTIs.36 On the other hand, a prospective study from the Netherlands found only a small difference in median CD64 concentrations between children with viral and bacterial LRTIs.37 Currently, there is insufficient evidence to use this biomarker in clinical practice.

Table 2. Overview of the most studied and most potential, new, host biomarkers for children with lower respiratory tract infections to differentiate between viral and bacterial infections.

Examples Advantages Disadvantages

Haematological markers

Erythrocyte sedimentation rate,

leukocytes, neutrophils

Easy to measureCommonly used

Differentiates insufficiently

Acute-phase protein CRP, PCT Easy to measureCommonly used

Differentiates insufficientlyDifferent cut-off values

Cytokine markers IL-6, TNF-α Promising study results Insufficiently investigated

Cell surface markers CD64, CD35 Promising study resultsin neonatal infections

Conflicting resultsin LRTI

New biomarkers RNA profiles,MxA

Multiplecandidate markers

Complex technique (RNA)Unknown cut-off values (MxA)

Insufficiently investigatedCombination of markers

MxA+CRP, CRP+TRAIL+IP-10

Increased discriminating power

Utility unknown

CRP: C-reactive protein, PCT: procalcitonin, IL-6: interleukin 6, TNF-α: Tumour Necrosis Factor alfa, MxA: Myxovirus Resistance Protein A, TRAIL: TNF-related apoptosis inducing ligand, IP-10: interferon gamma inducible protein-10, LRTI: lower respiratory

15873_CvHouten_BNW_v2.indd 165 26-11-18 12:43

Page 168: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

166

Chapter 8

New markersNew, but promising, are studies that investigate genetic markers. Different pathogens activate specific host responses, whereby specific pattern-recognizing receptors are expressed on leukocytes. RNA profiles can be identified with microarray analyses of these leukocytes. With the pathogen specific profiles it seems that a reliable distinction can be made between viral and bacterial infections.38-42 Already in 2007 a combination of 35 genes was identified in patients, mostly children, with respiratory infections.41 These genes had an overall accuracy of 95% in distinguishing Influenza A virus infections from infections with E. coli or S. pneumoniae. Recently a new combination of genetic markers in patients with respiratory infections was published. The authors found an accuracy of 87% for identifying viral and bacterial infection. In addition, an external validation in five other datasets (three of which also included children) showed an area under the ROC curve of 0.90 to 0.99.40 Despite these promising results, this technique is still too complex and insufficiently studied to be used in daily practice.New proteins in blood have also been considered as a diagnostic tool to distinguish between viral and bacterial infections. Myxovirus resistance protein A (MxA) is a protein that is induced by type 1 interferon and is secreted during viral infection and is not present in a bacterial infection. The largest study in this area, in which 553 children with an infection were included, showed a sensitivity of 96% and a specificity of 67% for the differentiation between viral and bacterial infections.43 More studies are needed to confirm these results, to examine the protein in children with lower respiratory tract infections and to establish accurate cut-off values.

Combination of host-response biomarkersIn addition to the identification of new biomarkers, combinations of different host biomarkers are studied. The combination of viral and bacterial markers appears to have added value in the diagnostic process. For example, in the before mentioned study on MxA, the MxA / CRP ratio was also evaluated.43 The researchers show that the area under the ROC-curve for distinguishing bacterial from viral infections for MxA alone is 0.89 (95% confidence interval: 0.82-0.96). When the MxA / CRP ratio is used, this area under the curve increases to 0.94 (95% confidence interval: 0.88-1.00). The test has not been tested specifically in children with a LRTIs. In a previous publication we showed that the combination of CRP with TNF-related apoptosis inducing ligand (TRAIL) and interferon gamma inducible protein-10 (IP-10) is significantly more accurate in distinguishing between viral and bacterial infections in children than CRP alone.3,44,45 Especially the addition of TRAIL is promising, because this is the first published host biomarker whose concentration increases in viral infections and decreases during bacterial infections.45 Once this combination test (ImmunoXpert) becomes available as a point of care device, clinical studies are needed to show whether the use of this test actually leads to a

15873_CvHouten_BNW_v2.indd 166 26-11-18 12:43

Page 169: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

167

8

A new diagnostic strategy

decrease in unnecessary antibiotics in children with lower respiratory tract infections without under treatment of serious bacterial infections.

Prediction models Multiple prediction models including clinical variables, with and without biomarkers, have been published in recent years. A review from 2012 found some diagnostic value in a pneumonia prediction model, but it was insufficient to widely implement this model.46 Hereafter, a robust prediction model was developed in a large observational study with an area under the ROC-curve of 0.84 for predicting bacterial pneumonia.47 However, this model includes 26 variables, making their usefulness in clinical practice limited. A Dutch research group showed that their prediction model, with clinical variables and CRP, has a distinctive capacity (c-statistic) of 0.81 for predicting bacterial pneumonia.48 In a recent validation study, they showed that especially in bacterial infections other than pneumonia, the addition of CRP to clinical features increases the model’s distinctive character. They found no difference in accuracy between CRP and PCT in combination with their prediction model.49 Although this is the first, validated prediction model including biomarkers, the results are not sufficient to base antibiotic treatment on the predicted risk. In the end, it is important that biomarkers are integrated into these clinical prediction models. That is why we are currently working on a study with the Sophia Children’s Hospital in which we investigate the diagnostic value of the addition of the biomarker combination test (CRP, TRAIL and IP-10)3,45 to the above-mentioned Dutch clinical prediction model.48

D isc ussionThe current available diagnostic tests differentiate insufficient between viral and bacterial infections in children with LRTI. At the moment, the Dutch Society for Paediatrics therefore recommends to perform no additional diagnostics and to have a ‘wait and see’ policy for children younger than two years of age with mild symptoms of a LRTI, including bronchiolitis (Figure 1a).14,15 Following the guideline, children with a clinically evident pneumonia should receive antibiotics, because viral and bacterial pneumonia cannot be accurately distinguished. In the absence of fast and accurate biomarkers, many children are treated unnecessary with antibiotics. With this article, we show that there are several, potentially useful, biomarkers in development. Diagnostic tests combining viral and bacterial biomarkers (MxA and CRP, TRAIL, CRP and IP-10) appear to be meaningful in ruling-out bacterial infections. If these diagnostic tests become widely available, they can be used as an intermediate step in the guideline for uncomplicated LRTIs before starting antibiotics. When there is a suspicion of a viral infection based on these host biomarkers, a wait-

15873_CvHouten_BNW_v2.indd 167 26-11-18 12:43

Page 170: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

168

Chapter 8

and-see policy with suffi cient follow-up is justifi ed in non-life-threatening infections (Figure 1b). In this way, antibiotic use can be limited to the group that really needs it. Until these new host biomarkers are available, additional diagnostics for uncomplicated LRTIs are not recommended and restrained antibiotic use is justifi ed given the low a priori chance of a bacterial infection.

a.

Life- threatening

Not life- threatening

Severe

Mild to moderate

Figure 1. Flowchart for children with lower respiratory tract infections. a. Current NVK guideline for lower respiratory tract infections. b. Future guideline. LRTI: lower respiratory tract infections, AB: antibiotics.

Viral infection

Bacterial infection

‘Wait and see’

<2 years

≥2 years

Biomarker

AB

AB

AB

LRTI

LRTI ‘Wait and see’

AB

AB

AB

AB

b.

Life- threatening

Not life- threatening

Severe

Mild to moderate

Figure 1. Flowchart for children with lower respiratory tract infections. a. Current NVK guideline for lower respiratory tract infections. b. Future guideline. LRTI: lower respiratory tract infections, AB: antibiotics.

Viral infection

Bacterial infection

‘Wait and see’

<2 years

≥2 years

Biomarker

AB

AB

AB

LRTI

LRTI ‘Wait and see’

AB

AB

AB

AB

Figure 1. Flowchart for children with lower respiratory tract infections.a. Current guideline “lower respiratory tract infections” from the Dutch association for paediatrics.14

b. Future vision for treatment of lower respiratory tract infections. LRTI: lower respiratory tract infections, AB: antibiotics.

15873_CvHouten_BNW_v2.indd 168 26-11-18 12:43

Page 171: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

169

8

A new diagnostic strategy

References1. WHO Fact sheet pneumonia. Available from: http://www.who.int/mediacentre/factsheets/fs3

31/en/ (accessed 2-1-2018).

2. Volksgezondheidenzorg, Lower respiratory tract infections numbers and context. Available from: https://www.volksgezondheidenzorg.info/onderwerp/infecties-van-de-onderste-luchtwegen/cijfers-context/huidige-situatie#!node-aantal-nieuwe-gevallen-van-infecties-van-de-onderste-luchtwegen (accessed 02-01-2018).

3. van Houten CB, de Groot JA, Klein A, et al. A host-protein based assay to diff erentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study. Lancet Infect Dis 2017; 17(4): 431-40.

4. Nijman RG, Thompson M, van Veen M, Perera R, Moll HA, Oostenbrink R. Derivation and validation of age and temperature specifi c reference values and centile charts to predict lower respiratory tract infection in children with fever: prospective observational study. Bmj 2012; 345: e4224.

5. Rambaud-Althaus C, Althaus F, Genton B, D’Acremont V. Clinical features for diagnosis of pneumonia in children younger than 5 years: a systematic review and meta-analysis. Lancet Infect Dis 2015; 15(4): 439-50.

6. Van den Bruel A, Haj-Hassan T, Thompson M, Buntinx F, Mant D. Diagnostic value of clinical features at presentation to identify serious infection in children in developed countries: a systematic review. Lancet 2010; 375(9717): 834-45.

7. Harris M, Clark J, Coote N, et al. British Thoracic Society guidelines for the management of community acquired pneumonia in children: update 2011. Thorax 2011; 66 Suppl 2: ii1-23.

8. Elemraid MA, Muller M, Spencer DA, et al. Accuracy of the interpretation of chest radiographs for the diagnosis of paediatric pneumonia. PLoS One 2014; 9(8): e106051.

9. Esposito S, Daleno C, Prunotto G, et al. Impact of viral infections in children with community-acquired pneumonia: results of a study of 17 respiratory viruses. Infl uenza and other respiratory viruses 2013; 7(1): 18-26.

10. Don M, Valent F, Korppi M, Canciani M. Diff erentiation of bacterial and viral community-acquired pneumonia in children. Pediatrics international 2009; 51(1): 91-6.

11. Chiappini E, Venturini E, Galli L, Novelli V, de Martino M. Diagnostic features of community-acquired pneumonia in children: what’s new? Acta paediatrica 2013; 102(465): 17-24.

12. Principi N, Esposito A, Giannitto C, Esposito S. Lung ultrasonography to diagnose community-acquired pneumonia in children. BMC pulmonary medicine 2017; 17(1): 212.

13. van der Zalm MM, van Ewijk BE, Wilbrink B, Uiterwaal CS, Wolfs TF, van der Ent CK. Respiratory pathogens in children with and without respiratory symptoms. J Pediatr 2009; 154(3): 396-400, .e1.

14. Richtlijn ‘Diagnostiek en behandeling van ongecompliceerde pneumonie bij kinderen in de tweede en derde lijn’ , Nederlandse Vereniging voor Kindergeneeskunde 2015. Available from: https://www.nvk.nl/Kwaliteit/Richtlijnen-overzicht/Details/articleType/ArticleView/articleId/1291/Onderste-Luchtweginfecties#tab15 (accessed 5-3-2018).

15. Richtlijn ‘Bronchiolitis’, Nederlandse Vereniging voor Kindergeneeskunde 2012. Available from: https://www.nvk.nl/Kwaliteit/Richtlijnen-overzicht/Details/articleType/ArticleView/articleId/698/Bronchiolitis#tab15 (accessed 25-7-2018).

15873_CvHouten_BNW_v2.indd 169 26-11-18 12:43

Page 172: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

170

Chapter 8

16. Dominguez J, Blanco S, Rodrigo C, et al. Usefulness of urinary antigen detection by an immunochromatographic test for diagnosis of pneumococcal pneumonia in children. Journal of clinical microbiology 2003; 41(5): 2161-3.

17. Doan QH, Kissoon N, Dobson S, et al. A randomized, controlled trial of the impact of early and rapid diagnosis of viral infections in children brought to an emergency department with febrile respiratory tract illnesses. J Pediatr 2009; 154(1): 91-5.

18. Bonner AB, Monroe KW, Talley LI, Klasner AE, Kimberlin DW. Impact of the rapid diagnosis of influenza on physician decision-making and patient management in the pediatric emergency department: results of a randomized, prospective, controlled trial. Pediatrics 2003; 112(2): 363-7.

19. Poehling KA, Zhu Y, Tang YW, Edwards K. Accuracy and impact of a point-of-care rapid influenza test in young children with respiratory illnesses. Archives of pediatrics & adolescent medicine 2006; 160(7): 713-8.

20. van de Pol AC, Wolfs TF, Tacke CE, et al. Impact of PCR for respiratory viruses on antibiotic use: theory and practice. Pediatric pulmonology 2011; 46(5): 428-34.

21. Goriacko P, Saiman L, Zachariah P. Antibiotic Use in Hospitalized Children with Respiratory Viruses Detected by Multiplex Polymerase Chain Reaction. Pediatr Infect Dis J 2017.

22. Shah SS, Dugan MH, Bell LM, et al. Blood cultures in the emergency department evaluation of childhood pneumonia. Pediatr Infect Dis J 2011; 30(6): 475-9.

23. Resti M, Micheli A, Moriondo M, et al. Comparison of the effect of antibiotic treatment on the possibility of diagnosing invasive pneumococcal disease by culture or molecular methods: a prospective, observational study of children and adolescents with proven pneumococcal infection. Clinical therapeutics 2009; 31(6): 1266-73.

24. Gill PJ, Richardson SE, Ostrow O, Friedman JN. Testing for Respiratory Viruses in Children: To Swab or Not to Swab. JAMA pediatrics 2017; 171(8): 798-804.

25. Bosch AATM, Bogaert D. Kinderneuzen vol met bacterien en virussen - het microbioom. Januari 2014. Available from: https://rivm.nl/Documenten_en_publicaties/Algemeen_Actueel/Uit gaven/Infectieziekten_Bulletin/Jaargang_25_2014/Januari_2014/Inhoud_Januari_2014/Kinderneuzen_vol_met_bacterien_en_virussen_het_microbioom (accessed 14-3-2018).

26. Esposito S, Principi N. Impact of nasopharyngeal microbiota on the development of respiratory tract diseases. European journal of clinical microbiology & infectious diseases 2018; 37(1): 1-7.

27. Cohen A, Bont L, Engelhard D, et al. A multifaceted ‘omics’ approach for addressing the challenge of antimicrobial resistance. Future microbiology 2015; 10(3): 365-76.

28. Gotta V, Baumann P, Ritz N, et al. Drivers of antibiotic prescribing in children and adolescents with febrile lower respiratory tract infections. PLoS One 2017; 12(9): e0185197.

29. Kapasi AJ, Dittrich S, Gonzalez IJ, Rodwell TC. Host Biomarkers for Distinguishing Bacterial from Non-Bacterial Causes of Acute Febrile Illness: A Comprehensive Review. PLoS One 2016; 11(8): e0160278.

30. Principi N, Esposito S. Biomarkers in Pediatric Community-Acquired Pneumonia. International journal of molecular sciences 2017; 18(2).

31. Galetto-Lacour A, Gervaix A. Identifying severe bacterial infection in children with fever without source. Expert review of anti-infective therapy 2010; 8(11): 1231-7.

32. Simon L, Gauvin F, Amre DK, Saint-Louis P, Lacroix J. Serum procalcitonin and C-reactive protein levels as markers of bacterial infection: a systematic review and meta-analysis. Clin Infect Dis 2004; 39(2): 206-17.

15873_CvHouten_BNW_v2.indd 170 26-11-18 12:43

Page 173: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

171

8

A new diagnostic strategy

33. Baer G, Baumann P, Buettcher M, et al. Procalcitonin guidance to reduce antibiotic treatment of lower respiratory tract infection in children and adolescents (ProPAED): a randomized controlled trial. PLoS One 2013; 8(8): e68419.

34. Yusa T, Tateda K, Ohara A, Miyazaki S. New possible biomarkers for diagnosis of infections and diagnostic distinction between bacterial and viral infections in children. Journal of infection and chemotherapy 2017; 23(2): 96-100.

35. Zhou JM, Ye Q. Utility of Assessing Cytokine Levels for the Differential Diagnosis of Pneumonia in a Pediatric Population. Pediatr Crit Care Med 2017; 18(4): e162-e6.

36. Zhu G, Zhu J, Song L, Cai W, Wang J. Combined use of biomarkers for distinguishing between bacterial and viral etiologies in pediatric lower respiratory tract infections. Infectious diseases 2015; 47(5): 289-93.

37. van Veen M, Nijman RG, Zijlstra M, et al. Neutrophil CD64 expression is not a useful biomarker for detecting serious bacterial infections in febrile children at the emergency department. Infectious diseases 2016; 48(5): 331-7.

38. Mahajan P, Kuppermann N, Mejias A, et al. Association of RNA Biosignatures With Bacterial Infections in Febrile Infants Aged 60 Days or Younger. Jama 2016; 316(8): 846-57.

39. Herberg JA, Kaforou M, Wright VJ, et al. Diagnostic Test Accuracy of a 2-Transcript Host RNA Signature for Discriminating Bacterial vs Viral Infection in Febrile Children. Jama 2016; 316(8): 835-45.

40. Tsalik EL, Henao R, Nichols M, et al. Host gene expression classifiers diagnose acute respiratory illness etiology. Sci Transl Med 2016; 8(322): 322ra11.

41. Ramilo O, Allman W, Chung W, et al. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood 2007; 109(5): 2066-77.

42. Hu X, Yu J, Crosby SD, Storch GA. Gene expression profiles in febrile children with defined viral and bacterial infection. Proceedings of the National Academy of Sciences of the United States of America 2013; 110(31): 12792-7.

43. Engelmann I, Dubos F, Lobert PE, et al. Diagnosis of viral infections using myxovirus resistance protein A (MxA). Pediatrics 2015; 135(4): e985-93.

44. Srugo I, Klein A, Stein M, et al. Validation of a Novel Assay to Distinguish Bacterial and Viral Infections. Pediatrics 2017; 140(4).

45. Oved K, Cohen A, Boico O, et al. A Novel Host-Proteome Signature for Distinguishing between Acute Bacterial and Viral Infections. PLoS One 2015; 10(3): e0120012.

46. Thompson M, Van den Bruel A, Verbakel J, et al. Systematic review and validation of prediction rules for identifying children with serious infections in emergency departments and urgent-access primary care. Health Technol Assess 2012; 16(15): 1-100.

47. Craig JC, Williams GJ, Jones M, et al. The accuracy of clinical symptoms and signs for the diagnosis of serious bacterial infection in young febrile children: prospective cohort study of 15 781 febrile illnesses. BMJ 2010; 340: c1594.

48. Nijman RG, Vergouwe Y, Thompson M, et al. Clinical prediction model to aid emergency doctors managing febrile children at risk of serious bacterial infections: diagnostic study. BMJ 2013; 346: f1706.

49. Nijman RG, Vergouwe Y, Moll HA, et al. Validation of the Feverkidstool and procalcitonin for detecting serious bacterial infections in febrile children. Pediatric research 2017.

15873_CvHouten_BNW_v2.indd 171 26-11-18 12:43

Page 174: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

9

15873_CvHouten_BNW_v2.indd 172 26-11-18 12:43

Page 175: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

9Summary, general discussion and future perspectives

15873_CvHouten_BNW_v2.indd 173 26-11-18 12:43

Page 176: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 174 26-11-18 12:43

Page 177: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

175

9

Summary, general discussion and future perspectives

S ummar y of the main finding of this thesisTimely antibiotic treatment improves the outcome of patients with bacterial infections, but antibiotics are not indicated in case of viral infections. Unfortunately, it is often not possible to differentiate between bacterial and viral infections based on clinical judgement alone, even when using available blood tests. This thesis describes our research to the different aspects of this diagnostic dilemma.

First, we evaluated the consequences of this complicated diagnostic process on antibiotic use. We used an expert panel of paediatric infectious disease physicians to assign a viral or bacterial diagnosis to each individual case. In chapter 2, we present the characteristics of 188 children positive for respiratory syncytial virus (RSV). Only 14% of them were diagnosed with a bacterial co-infection. Most of these children were admitted to the paediatric intensive care unit (PICU). All bacterial co-infections were treated with antibiotics. One third of all children with viral RSV infections without presumed bacterial co infections were treated unnecessarily with antibiotics. The only clinical feature that differed significantly between children with and without bacterial co-infections was “ill-appearing”. This result emphasizes that clinical signs and symptoms cannot help physicians distinguish between patients who benefit from antibiotics and those who do not.

The aim of the study described in chapter 3 was to compare antibiotic overuse in children and adults with respiratory tract infections (RTI) using expert panel diagnoses. The proportion of viral infections was larger in children than in adults (74% versus 38%). However, antibiotic overuse was less frequent in children than adults with viral RTI (37% versus 83%). These findings support the need for effective diagnostics to decrease antibiotic overuse in RTI patients of all ages.

Second, we aimed to address the need for better diagnostics to differentiate between viral and bacterial infections. In chapter 4 we describe the results of a double-blinded, prospective, international, validation study (the OPPORTUNITY study) in children below the age of five. We studied a novel assay combining three proteins: tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), interferon gamma induced protein-10 (IP-10), and C-reactive protein (CRP). We showed that this combination of biomarkers upregulated by viral and bacterial infections had a high diagnostic value. The assay is especially helpful for ruling out bacterial infections. The number of false positives decreased with 51% if the combination assay was used instead of CRP alone. This result suggests that this assay has the potential to reduce antibiotic misuse in young children. Our findings are promising and support the need for implementation research to examine the added clinical utility on unnecessary antibiotic prescriptions.

15873_CvHouten_BNW_v2.indd 175 26-11-18 12:43

Page 178: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

176

Chapter 9

In addition to biomarkers, clinical signs and symptoms play an important role when physicians diagnose febrile children. In chapter 5, we therefore integrated the above-mentioned combination assay in an existing clinical prediction model for febrile children presenting at the hospital. This model includes clinical signs and symptoms and CRP and was updated with the combination assay. The updated model was more accurate in predicting serious bacterial infections compared to the model with CRP as the only biomarker. Our results show that the added value of the assay is highest in children with a predicted risk <40% for serious bacterial infection as predicted by the original prediction rule. This cut-off point can therefore be used to optimize cost-effective use of the combination assay.

As the aforementioned combination assay is not perfect and as antibiotic resistance has important health consequences, more research is needed. In chapter 6, we have therefore taken the OPPORTUNITY study forward to an international prospective study (The TAILORED Treatment study) to develop a multi-parametric algorithm to distinguish viral from bacterial infections in paediatric and adult patients. The multi-omics approach to generate infectious disease algorithms utilised by this study is currently unique to diagnose bacterial infections and provides a template for infectious disease studies in the future.

Third, the absence of a gold standard to diagnose bacterial infections is troublesome in diagnostic studies. All studies presented in this thesis used an expert panel reference standard to establish patient outcomes (i.e. bacterial or viral infection). In chapter 7, we aimed to evaluate the reproducibility of such an expert panel reference standard. We found that a panel consisting of three experts provides more reproducible diagnosis than individual expert diagnosis. Even with a time interval of three years, the intra-panel agreement was high (kappa 0.80). Increasing the size of the expert panel to seven experts has no major advantage. Our findings supporting the need for further validation and optimization of panel diagnoses to obtain accurate results of diagnostic studies without gold standard.

Finally, in chapter 8, we provide an overview of current and new diagnostics in children with RTIs. Protein- and RNA-based blood host response biomarkers show promising results. Techniques that identify RNA profiles are still too complex and the study results too preliminary to be used in clinical practice. However, the combination of biomarkers that are upregulated by viral and bacterial infections are almost ready to use in clinical practice and may help physicians become more restrictive in antibiotic use in patients with mild or moderately severe infection.

15873_CvHouten_BNW_v2.indd 176 26-11-18 12:43

Page 179: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

177

9

Summary, general discussion and future perspectives

To interpret the findings of the studies presented in this thesis in a larger context, several issues and suggestions for future research need to be discussed. First, we discuss the balance between too much and too little antibiotics. Then, we elaborate on opportunities and challenges to improve diagnosis of bacterial infection. Finally, we discuss lessons learned from this thesis regarding expert panel reference standard diagnoses.

Too much and too l ittle antibioticsThe impact of antibiotic overuse in the NetherlandsAs mentioned in the introduction of this thesis, antibiotic overuse is a major problem worldwide.1,2 Compared to other countries, antibiotic prescription rates in Dutch patients are relatively low, and when it comes to reducing antibiotic use, the Netherlands is seen as an example.3,4 The impact of antibiotic overuse in Dutch patients with lower respiratory tract infections (LRTI) is unclear. Therefore, we combined the estimated percentages of viral infections and antibiotic prescriptions in these viral infection (this thesis) and the numbers of newly diagnosed LRTI (Dutch public health data). We calculated that in the Netherlands annually almost 190,000 antibiotic prescriptions are prescribed unnecessary for patients with LRTI alone (Table 1). This number is probably an underestimation, as at the general practitioner level more children are diagnosed with a viral infection (i.e. 90%).5,6 The percentages of antibiotic overuse are similar for both hospital and general practitioner.5,6 Table 1 shows that the absolute number of annually antibiotic overuse is higher in adults compared to children. Based on these numbers, it may be argued that further research should focus mainly on adult patients. However, the problem of antimicrobial resistance is more complex. For example, due to a high carrier rate of bacteria in the airways and day-care facilities the transmission of bacteria is higher in children compared to adults. In addition, it is unknown if antibiotic resistance is a reversible process, and thus it is unknown if children will carry antibiotic resistance bacteria for the rest of their lives or not.7 Altogether, we need to conduct diagnostic studies in both children and adults.

Paucity of antibioticsWhereas there is a major overconsumption of antibiotics in high income countries (HICs), there is often a scarcity of antibiotics in low and middle income countries (LMICs). Balancing improvement of antibiotic access for therapeutic uses and minimisation of antibiotic misuse is critical.9 More children in LMICs countries die from inadequate access to antibiotics than from drug-resistant infections10, yet resistance threatens the long-term viability of these drugs.11 A recent randomized controlled trial performed in sub-Saharan Africa showed that all-cause mortality was 14% lower among almost

15873_CvHouten_BNW_v2.indd 177 26-11-18 12:43

Page 180: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

178

Chapter 9

200,000 preschool children who received a twice-yearly dose of azithromycin than among children who received placebo.12 Nonspecific use of antibiotics is discouraged, but in these settings there is also lack of access to instruments necessary to make the correct diagnosis.9 The need for LMICs to develop actionable strategies to combat antibiotic resistance and improve antibiotic stewardship has been recognized by major global public health institutions, including the US Centers for Disease Control and Prevention and the World Health Organization.13,14 Due to structural, cultural and socioeconomic factors contributing to the development of antibiotic resistance, it is widely acknowledged that LMICs require a distinct approache.15-17 Collective actions are needed to prioritize in national action plans on antimicrobial resistance based on costs, benefits, and situational context.18

Table 1. Calculation of annual antibiotic overuse in The Netherlands for LRTI.

Percentage Absolute number

Children

Episodes of LRTI NA 110,7508

Viral aetiology 63% 69,773

Viral infections treated with antibiotics 51% 35,584

Adults

Episodes of LRTI NA 518,6368

Viral aetiology 32% 165,964

Viral infections treated with antibiotics 93% 154,346

Total population

Viral infections treated with antibiotics NA 189,930

Percentages of viral infections (based on expert panel diagnosis) and antibiotic prescriptions (this thesis) and numbers of newly diagnosed lower respiratory tract infections (Dutch public health data). LRTI: lower respiratory tract infection. NA: not applicable.

Modelling to assess the impact of vaccinesIn addition to improvements in diagnostic testing, antibiotic stewardship and new antibiotics, vaccines have been proposed as an essential intervention to reduce antibiotic misuse.19,20 At different levels, vaccination affects both the number of cases of bacterial resistant disease and the amount of antibiotic use.21 At the direct level bacterial vaccines provide immunological protection against carriage and infection, and therefore less often requires antimicrobial treatment. In addition, at the population level, vaccination leads to a reduction in the transmission of drug-resistant bacteria.21 Consequently, less people are infected which leads to a reduction of antibiotic use. Current mathematical models identified some of the effects of bacterial vaccines on antibiotic resistance dynamics. Existing models, however, do not include economic evaluations of vaccinations. These assessments might be important to inform public

15873_CvHouten_BNW_v2.indd 178 26-11-18 12:43

Page 181: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

179

9

Summary, general discussion and future perspectives

health policy makers. Next to bacterial vaccines, viral vaccines, such as influenza and measles vaccines, might also reduce antibiotic use.22,23 This could also be true for RSV as we showed that almost half of the children infected with RSV received antibiotics.24 Therefore, if an RSV vaccine becomes available25, implementation may result in a reduction of antibiotic use. However, no models exist so far that show how vaccinations against viral infections can impact antimicrobial resistance.

Future perspectives• New diagnostic tools to differentiate between viral and bacterial infections, developed in

HIC, should be validated in LMIC.

• Mathematical models for vaccines should be improved to inform public health policy makers in determining strategies against antibiotic overuse.

• Future research is needed to expand knowledge about the resistome and reversibility of antibiotic resistant bacteria.

D iagnosing bac terial infec tionsNew opportunitiesA unique biomarker to differentiate between viral and bacterial infections is the protein TNF-related apoptosis inducing ligand (TRAIL). TRAIL is the first published host biomarker in which concentrations increase in every viral infection and decrease in bacterial infections.26 TRAIL is a small protein with concentrations in the range of picograms per millilitre. Until recently, this protein was primarily studied in tumour cells.27 In the last years, it has become more clear that TRAIL also plays an important role in the modulation of both the innate and adaptive inflammatory responses.28 TRAIL is produced and secreted by most normal tissue cells and after activation by interferons (IFN), TRAIL is upregulated on the cell surface of various cells of the immune system (e.g. natural killer cells, T cells, dendritic cells and macrophages).29 There are five known receptors for TRAIL: two signalling receptors and three decoy receptors.30 TRAIL is capable of initiating apoptosis through at least six known, caspase dependant, pathways. These complex pathways presents many opportunities for regulation by the host, but also for manipulation by a virus. TRAIL may limit or enhance viral replication dependent on the specific virus. Some viruses (e.g. rhinovirus, adenovirus) can exploit mechanisms of cell death to spread progeny to neighbouring cells, thereby increasing their pathogenicity.30 In this thesis, we focus on TRAIL as a diagnostic marker while TRAIL may also serve as a prognostic marker. Soluble TRAIL levels are associated with immune paralysis and a high risk of mortality in adults with sepsis.28 In the Opportunity study a similar preliminary trend was seen: the more severely ill children had lowest TRAIL concentrations.In addition to TRAIL, there is IFN-γ induced protein-10 (IP-10). This protein is secreted

15873_CvHouten_BNW_v2.indd 179 26-11-18 12:43

Page 182: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

180

Chapter 9

by several cell types in response to IFN-γ31 and has a function in chemo attraction of monocytes and T cells.32 IP-10 concentrations increase both in viral and bacterial infection, but increases to significantly higher concentrations in viral infections compared to bacterial infections.26

Another promising host blood biomarker that is upregulated in viral infections is Myxovirus resistance protein A (MxA).33-35 The MxA gene is expressed in blood mononuclear cells and epithelial cells, and expression is upregulated exclusively by type 1 interferons (IFNs) and does not respond to other cytokines such as IL-1 or TNF-α.36,37 MxA prevents virus transport to the nucleus, so that the primary transcription of the viral genome cannot take place. In most cases of acute viral infections, type 1 IFN and MxA are released into the peripheral blood. Detection of IFNs in serum is difficult and unreliable, mainly due to their short half-life.38 In contrast, MxA has a long half-life of 2.3 days (19 hours for CRP) and shows a fast induction within 1-2 hours after infection (36 hours for CRP and 8 hours for procalcitonin (PCT)).39,40

More complex, but also auspicious, host biomarkers to differentiate between viral and bacterial infections are the RNA profiles. RNA profiles can be identified with microarray analyses of leukocytes, with the help of these pathogen specific profiles, a reliable diagnosis of specific infections can be made.41-45 Currently there are several limitations of this technique (e.g. identifying an accurate and limited subset of RNA markers, developing a point of care test and making the test cost effective), and it will take probably many years before these problems will be solved. However, the advantage of pathogen discrimination makes it worthwhile to continue studying these RNA profiles.

Combining biomarkers is the key to successCurrently used host biomarkers to differentiate between viral and bacterial infections are mostly biomarkers upregulated primarily by bacterial infections (e.g. CRP and PCT). The concentrations of the above-mentioned new biomarkers (TRAIL and MxA) increase primarily in viral infections. These distinct responses to viral and bacterial infection can be helpful to diagnose viral or bacterial infections. It is therefore not surprising that current research focuses on combinations of these different biomarkers. In this thesis, we have studied the combination of markers upregulated by viral infections (i.e. TRAIL and IP-10) with CRP. This combination assay has a negative predictive value of 98% and is more accurate in differentiating between bacterial and viral infections in children compared to all three markers separately. The high diagnostic value of this combination assay is founded on the distinct dynamics of TRAIL, CRP, and IP-10 during viral and bacterial infections (Figure 1). Later on, similar results were found in other validation studies of this combination assay (Table 2).

15873_CvHouten_BNW_v2.indd 180 26-11-18 12:43

Page 183: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

181

9

Summary, general discussion and future perspectives

Figure 1. Three-dimensional view showing the strength of a combination assay. The addition of each biomarker with distinct responses to viral and bacterial infection leads to an increase in accurate viral or bacterial diagnosis (in green and red). CRP: C-reactive protein, TRAIL: TNF-related apoptosis inducing ligand, IP-10: interferon gamma induced protein-10.

Another promising diagnostic test combines concentrations of CRP and MxA. An utilization study of this assay in 21 adult patients presenting at the general practice showed diagnostic accuracy and positive impact on appropriate antibiotic prescribing.50 Especially, the high negative predictive value (97%) reduces clinician’s fear of missing a serious bacterial infection and supports a watchful waiting approach.50

In addition to these blood biomarkers, we should not forget the diagnostic value of clinical signs and symptoms. Diagnostic algorithms based solely on clinical data are inaccurate to diff erentiate between viral and bacterial infections.51 Even when CRP or PCT is added to these clinical predictions rules, the algorithms are not accurate enough

15873_CvHouten_BNW_v2.indd 181 26-11-18 12:43

Page 184: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

182

Chapter 9

to be used in clinical settings.52,53 However, in this thesis we showed that the addition of the combination assay (CRP, TRAIL and IP-10) to a previously published clinical prediction rule, results in a more accurate algorithm compared to the prediction rule with CRP alone. Incorporating point-of-care testing of a combination of biomarkers that are upregulated in viral and bacterial infections in clinical prediction rules can be helpful in the decision to start antibiotics or not.

Table 2. Overview of studies analysing the combination of CRP, TRAIL and IP-10.

Author Year Cohort

Number of evaluated patients Sensitivity Specificity PPV NPV

Oved et al.26 2015 Children and adults

653 92% 89% 76-98% 78-98%

van Houten et al.46 2016 Children 506 86.7% 91.1% 60.5% 97.8%

Srugo et al.47 2017 Children 307 93.8% 89.8% 74.4% 97.9%

Stein et al.48 2017 Children and adults

124 93% 91% 96% 86%

Ashkenazi et al.49 2018 Children and adults

314 93.5% 94.3% 92.7% 94.9%

Remaining challengesDiagnostics for life-threatening infectionsIn children admitted to the PICU, the risk of poor outcomes is highest if appropriate antimicrobial therapy is delayed. PICU admission represents a clinical scenario in great need of improved diagnostics. In this thesis, we showed that the combination of CRP, TRAIL and IP-10 was inaccurate in differentiating between viral and bacterial infections in children admitted to the PICU. Also other biomarkers such as CRP and PCT do not differentiate accurately between bacterial and viral RTI in children admitted to the PICU.54,55 We found that bacterial co-infections occurred more often in RSV infected children admitted to the PICU compared to children not admitted to the PICU.24 The mixed origin of most PICU admissions may explain the low accuracy of biomarkers in these severely ill patients. Especially, in mechanically ventilated patients high rates of false positive cases are seen.55 Mechanical ventilation can cause lung injury and thereby activation of the immune system.56 This exogenous immune system activation may influence the accuracy of host-biomarkers. Taken together, we need to conclude that a pre-emptive approach regarding antibiotic use seems unavoidable in children with life-threatening infections.

15873_CvHouten_BNW_v2.indd 182 26-11-18 12:43

Page 185: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

183

9

Summary, general discussion and future perspectives

Diagnostic thresholdsAnother challenge in the diagnosis of a bacterial infection is to determine thresholds for new diagnostic tests. Different clinical contexts require distinct analysis and thresholds to determine when to initiate antibiotic therapy. Therefore, different cut-offs for biomarker concentrations are used for ruling in and ruling out a bacterial infection. Most of these cut-offs points include a range of equivocal results. With the combination assay studied in this thesis the number of patients with an equivocal test results decreased, but for some patients a diagnosis was still not available (Figure 1). Equivocal test results seem to be unavoidable, however, the number of patients with an equivocal test result should be as low as possible. Expert panel consensus meetings can be considered to determine thresholds for the most common clinical contexts (e.g. non-admitted, admitted, PICU admission, immunocompromised). The duration of disease need to be taken into account when determining these thresholds, as some biomarkers, especially CRP, research their peak concentration after several hours.40 These thresholds should be used and evaluated in utility studies, based on the results of these studies recommendations can be made.

Point-of-care technology At the moment the combination assay of CRP, TRAIL and IP-10 is an ELISA based test. It is important to develop a bedside point-of-care (POC) test to implement the assay in clinical practice. Despite the fact that a POC-test must be rapid and easy to use, this POC technology must also have the same precision as the ELISA. Most challenging therefore is the detection of TRAIL, as this is a small protein with a range of picograms per millilitre. Finally, the performed accuracy studies are performed on serum samples, while a capillary blood sample is preferred. Many test samples are needed to see whether the measurement of TRAIL, IP-10 and CRP in capillary samples is as accurate as the measurement of these proteins in serum samples.

RegulationWhile developing the POC technology, a regulatory appraisal must be performed. At the moment the assay has a Conformité Européene (CE) mark as In Vitro Diagnostics (IVD), meaning that it meets the Essential Requirements regarding safety. However, this mark doesn’t say anything about efficacy of the test. The Food and Drug Administration (FDA) is looking at the wider picture and asking ‘Does healthcare really need it?’. This regulation process has a high degree of scrutiny and is therefore time consuming and expensive.

15873_CvHouten_BNW_v2.indd 183 26-11-18 12:43

Page 186: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

184

Chapter 9

ImplementationSince the accuracy of the new assay has been proven, the next step is to perform implementation research to examine the added clinical utility to diagnose bacterial infections in clinical care. This final phase leads also to another challenge, namely the adherence to antibiotic prescription recommendations.57 Non-compliance to recommendations is higher if children are hospitalized, have younger age and have pneumococcal infection risk factors.57 Risk factors for pneumococcal infections included in this study are immune deficiency, asplenia, sickle cell disease and chronic lung disease.57 From other studies it is known that training of physicians and providing them with feedback regarding their antibiotic prescriptions are effective in reducing the number of unnecessary prescriptions.58,59 Therefore, even if a fast and accurate diagnostic test is commercially available, it will take time and effort before the test will lead to a reduction in antibiotic misuse.

Health technology assessmentAfter the implementation phase it is important to evaluate the healthcare, patient and societal cost-benefit. As the combination assay is not yet available as a POC test, the manufacturer has not given an indication of the eventual cost. Evaluation of the test benefits regarding the direct costs (e.g. antibiotic exposure, resource use, adverse clinical outcomes) will be relatively easy, however, to calculate the benefits of the test concerning indirect costs (e.g. the cost of antimicrobial resistance) will be a challenge.

Future perspectives• Future diagnostic research should focus on combinations of viral and bacterial host

biomarkers with clinical signs and symptoms.

• The combination of CRP, TRAIL and IP-10 should be tested in a utility study if a point of care test is available.

• There is a need for new biomarkers that can differentiate between viral and bacterial infections in children admitted to the paediatric intensive care unit.

Research without gold standardDefining the standard to diagnose bacterial infectionsAs discussed in this thesis, there is no gold standard to diagnose bacterial infections. An expert panel diagnosis is a widely used method to provide a solution.60 Unfortunately, many variations of expert panel reference standards are used because clear guidelines are lacking.61 Based on the experiences gained in this thesis, we highlight some important considerations when using an expert panel reference diagnosis.

15873_CvHouten_BNW_v2.indd 184 26-11-18 12:43

Page 187: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

185

9

Summary, general discussion and future perspectives

First, it is important that the expert panel is blinded to the result of the index test and for the diagnoses of their peers.62 A limitation of the Opportunity study is that the expert panel was provided with CRP values when tested for routine care, as this parameter is routinely used to help identify the cause of infection.63 Panel members were also informed about the biomarkers included in the index test. To avoid incorporation bias, it is recommended to not disclose index test information with the expert panel. Including the test being assessed in the reference standard can lead to overestimating test accuracy.64 Therefore, we cannot exclude some degree of overestimation of the study results. Second, performing follow-up for all patients and providing the expert panel this information is strongly recommended. A follow-up phone call 2-4 weeks after recruitment can help expert panels to distinguish between viral and bacterial aetiologies, especially for non-admitted patients. Several cases that are difficult to diagnose at presentation will become easy to diagnose for the panel when receiving information about the disease course. It has the advantage of capturing a wider spectrum of diseases illness severities in the study cohort, including difficult-to-diagnose cases.60 Finally, as shown in this thesis, there is large intra- and inter observer variability. Nevertheless, we showed that a panel consisting of three experts provides a more reproducible diagnosis than individual expert diagnosis. This finding confirms the conclusion of other studies, a panel diagnosis is more stable, and therefore probably more accurate, than an individual assessment.60,62 In our study increasing the size of an expert panel beyond three members seems to have limited added value. In conclusion, the preferred reference standard in absence of a gold standard is a blinded, expert panel consisting of three experts who receive follow-up information from every patient.

What if the expert panel is not conclusive?Inconclusive reference standards are inherent to studies without a gold standard. Complete transparency regarding the handling of inconclusive diagnoses in the analysis is essential for the reader to understand how the results have been derived. First, researchers should report sufficient information on these inconclusive cases, including the answers to the following questions: When is the reference standard considered to be inconclusive? How many patients had an inconclusive reference standard? Where the characteristics of these patients similar to the rest of the cohort?61 It can thereby be helpful to extended the standard 2x2 (i.e. reference standard positive or negative versus index test result positive or negative) classification matrix to a 3x2 matrix (i.e. reference standard positive, inconclusive or negative versus index test result positive or negative). If there are also inconclusive index tests results the matrix should be a 3x3 matrix.65

15873_CvHouten_BNW_v2.indd 185 26-11-18 12:43

Page 188: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

186

Chapter 9

Table 3. Overview of possible approaches to handle inconclusive diagnosis.

Approach Advantage Disadvantage

Exclusion from primary analysis Only ‘true’ diagnoses in primary analysis.

Some difficult to diagnose cases are left out.

Imputation Increase in number of available cases.

Possibility of incorrect diagnoses.

“Best and worst case” Provide a range within the real accuracy can be expected.

Both scenarios aren’t realistic.

Discussion between experts Most similar to routine care. Increases workload for experts. Dominated by a single member.

Thereafter, researchers should predefine how inconclusive cases will be handled. Unfortunately, there is no single “optimal” approach to analyse inconclusive reference standard results. Table 3 shows an overview of the possible approaches. If inconclusive cases are excluded from the primary analysis, it is important to report the test results for these cases as well. The imputation model is especially useful when comparing different tests (e.g. like we did when updating the clinical prediction rule). With a “best and worst case” scenario, the accuracy analyses are first performed considering all inconclusive cases as having a viral outcome and second with all inconclusive cases assigned as a bacterial infection.65 These analyses can be performed secondarily. A commonly used method for discussion between experts is the Delphi method.66 The Delphi method is a staged procedure in which first the diagnosis per individual member is generated followed by a plenary discussion of those cases with disagreement.67,68 To conclude, for pragmatic reasons excluding inconclusive cases from the primary analysis seems the most suitable approach.

Future perspectives• More validation studies for expert panels as reference standard in infectious disease

diagnostic studies should be performed to optimize the use of this reference standard.

• Based on validation studies, a guideline for the use an expert panel reference standard in diagnostic studies should be made.

• Advantages and disadvantages of different approached to analyse inconclusive reference standards should be studied in more detail.

Concluding remarks It is important to realize that the large amount of inappropriate use of antibiotics that we found in this thesis has global health and economic consequences. However, in a large proportion of the patients it remains hard to identify which patients benefit from antibiotics and which do not. To find solutions for this diagnostic dilemma, we studied

15873_CvHouten_BNW_v2.indd 186 26-11-18 12:43

Page 189: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

187

9

Summary, general discussion and future perspectives

a new combination assay (CRP, TRAIL and IP-10). The promising results we found during the first external validation of this assay in children and the improved accuracy after adding the assay to an existent clinical prediction rule, brings the search for an accurate diagnostic test a step further. The ultimate goal in the evaluation of a diagnostic test is to demonstrate health improvement. Validation studies (as performed in this thesis) do not provide information about clinical utility (e.g. antibiotic use and clinical outcome). Therefore, as sufficient diagnostic value of the combination assay has been established, utility studies are now indicated. Unfortunately, regulators do not require utility studies before allowing implementation of novel diagnostics in routine care. This results in the implementation of diagnostics of which we do not know whether or not there is benefit for the patient or society.In the absence of a gold standard, we used an expert panel reference standard and found that the reproducibility of a panel consisting of three experts provides more reproducible diagnosis than individual expert diagnosis. Our findings are important for researchers, but also for regulatory organisations, as these organisations approve drugs based on studies using expert opinions as the outcome. Due to imperfect reproducibility of the expert diagnosis the estimated accuracy of the diagnostic test under evaluation may be an overestimation or an underestimation. Therefore, further studies are needed to optimise the expert panel reference standard.My vision for the future of timely identification of patients with bacterial infection is one in which clinical judgement is combined with accurate protein-based or RNA-based blood host response biomarkers. This combination will be sensitive enough to allow for restrictive antibiotic use in patients with mild or moderately severe infection. For life-threatening infections liberal use of antibiotics will remain required as we are still searching for sufficiently sensitive diagnostics in this population.

15873_CvHouten_BNW_v2.indd 187 26-11-18 12:43

Page 190: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

188

Chapter 9

References1. CDC: About Antimicrobial Resistance. Available from: https://www.cdc.gov/drugresistance/

about.html (accessed 7-7-2017).

2. O’Neill J. Review on Antimicrobial Resistance: Tackling drug-resistant infections globally: final report and recommendations. London 2016. Available from: https://amr-review.org/sites/default/files/AMR%20Review%20Paper%20%20Tackling%20a%20crisis%20for%20the%20health%20and%20wealth%20of%20nations_1.pdf (accessed 4-4-2018).

3. The center for disease dynamics: Antibiotic prescribing rates by country. Available from: http://www.cddep.org/tool/antibiotic_prescribing_rates_country#sthash.kiRPO6aH.dps (accessed 5-2-2018).

4. Sheldon T. Saving antibiotics for when they are really needed: the Dutch example. Bmj 2016; 354: i4192.

5. Kool M. Serious infections and healthcare in children with fever prsenting at a general practice oout-of-hours service. In: Febrile children at a general practice out-of-hours service [thesis]. Rotterdam: Erasmus University; 2015.

6. Dekker AR, Verheij TJ, van der Velden AW. Inappropriate antibiotic prescription for respiratory tract indications: most prominent in adult patients. Family practice 2015; 32(4): 401-7.

7. Sundqvist M. Reversibility of antibiotic resistance. Upsala journal of medical sciences 2014; 119(2): 142-8.

8. Volksgezondheidzorg, Lower respiratory tract infections numbers and context. Available from: https://www.volksgezondheidenzorg.info/onderwerp/infecties-van-de-onderste-luchtwegen/cijfers-context/huidige-situatie#!node-aantal-nieuwe-gevallen-van-infecties-van-de-onderste-luchtwegen (accessed 02-01-2018).

9. Mendelson M, Rottingen JA, Gopinathan U, et al. Maximising access to achieve appropriate human antimicrobial use in low-income and middle-income countries. Lancet 2016; 387(10014): 188-98.

10. Laxminarayan R, Matsoso P, Pant S, et al. Access to effective antimicrobials: a worldwide challenge. Lancet 2016; 387(10014): 168-75.

11. Nadimpalli M, Delarocque-Astagneau E, Love DC, et al. Combating Global Antibiotic Resistance: Emerging One Health Concerns in Lower- and Middle-Income Countries. Clin Infect Dis 2018; 66(6): 963-9.

12. Keenan JD, Bailey RL, West SK, et al. Azithromycin to Reduce Childhood Mortality in Sub-Saharan Africa. N Engl J Med 2018; 378(17): 1583-92.

13. National action plan for combating antibiotic-resistant bacteria. Atlanta, GA: US Centers for Disease Control and Prevention. 2015. Available from: https://www.cdc.gov/drugresistance/pdf/national_action_plan_for_combating_antibotic-resistant_bacteria.pdf. (accessed 28-04-2018).

14. Organization. WH. Global action plan on antimicrobial resistance. Geneva, Switzerland: WHO, 2015.

15. Okeke IN, Lamikanra A, Edelman R. Socioeconomic and behavioral factors leading to acquired bacterial resistance to antibiotics in developing countries. Emerging infectious diseases 1999; 5(1): 18-27.

16. Laxminarayan R, Duse A, Wattal C, et al. Antibiotic resistance-the need for global solutions. Lancet Infect Dis 2013; 13(12): 1057-98.

15873_CvHouten_BNW_v2.indd 188 26-11-18 12:43

Page 191: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

189

9

Summary, general discussion and future perspectives

17. Byarugaba DK. A view on antimicrobial resistance in developing countries and responsible risk factors. International journal of antimicrobial agents 2004; 24(2): 105-10.

18. Kakkar M, Sharma A, Vong S. Developing a situation analysis tool to assess containment of antimicrobial resistance in South East Asia. Bmj 2017; 358: j3760.

19. Fortanier AC, Venekamp RP, Stellato RK, et al. Outpatient antibiotic use in Dutch infants after 10-valent pneumococcal vaccine introduction: a time-series analysis. BMJ open 2018; 8(6): e020619.

20. Ginsburg AS, Klugman KP. Vaccination to reduce antimicrobial resistance. The Lancet Global health 2017; 5(12): e1176-e7.

21. Atkins KE, Lafferty EI, Deeny SR, Davies NG, Robotham JV, Jit M. Use of mathematical modelling to assess the impact of vaccines on antibiotic resistance. Lancet Infect Dis 2017.

22. Marchisio P, Esposito S, Bianchini S, et al. Efficacy of injectable trivalent virosomal-adjuvanted inactivated influenza vaccine in preventing acute otitis media in children with recurrent complicated or noncomplicated acute otitis media. Pediatr Infect Dis J 2009; 28(10): 855-9.

23. Hansen NS, Byberg S, Hervig Jacobsen L, et al. Effect of early measles vaccine on pneumococcal colonization: A randomized trial from Guinea-Bissau. PLoS One 2017; 12(5): e0177547.

24. van Houten CB, Naaktgeboren C, Buiteman BJM, et al. Antibiotic Overuse in Children with Respiratory Syncytial Virus Lower Respiratory Tract Infection. Pediatr Infect Dis J 2018.

25. Mazur NI, Higgins D, Nunes MC, et al. The respiratory syncytial virus vaccine landscape: lessons from the graveyard and promising candidates. Lancet Infect Dis 2018.

26. Oved K, Cohen A, Boico O, et al. A Novel Host-Proteome Signature for Distinguishing between Acute Bacterial and Viral Infections. PLoS One 2015; 10(3): e0120012.

27. MacFarlane M. TRAIL-induced signalling and apoptosis. Toxicology letters 2003; 139(2-3): 89-97.

28. Tian Y, Tao T, Zhu J, et al. Soluble tumor necrosis factor related apoptosis inducing ligand level as a predictor of severity of sepsis and the risk of mortality in septic patients. PLoS One 2013; 8(12): e82204.

29. Gyurkovska V, Ivanovska N. Distinct roles of TNF-related apoptosis-inducing ligand (TRAIL) in viral and bacterial infections: from pathogenesis to pathogen clearance. Inflammation research 2016; 65(6): 427-37.

30. Cummins N, Badley A. The TRAIL to viral pathogenesis: the good, the bad and the ugly. Current molecular medicine 2009; 9(4): 495-505.

31. Luster AD, Unkeless JC, Ravetch JV. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature 1985; 315(6021): 672-6.

32. Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD. IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. Journal of immunology (Baltimore, Md : 1950) 2002; 168(7): 3195-204.

33. Engelmann I, Dubos F, Lobert PE, et al. Diagnosis of viral infections using myxovirus resistance protein A (MxA). Pediatrics 2015; 135(4): e985-93.

34. Self WH, Rosen J, Sharp SC, et al. Diagnostic Accuracy of FebriDx: A Rapid Test to Detect Immune Responses to Viral and Bacterial Upper Respiratory Infections. Journal of clinical medicine 2017; 6(10).

35. Joseph P, Godofsky E. Outpatient Antibiotic Stewardship: A Growing Frontier-Combining Myxovirus Resistance Protein A With Other Biomarkers to Improve Antibiotic Use. Open forum infectious diseases 2018; 5(2): ofy024.

15873_CvHouten_BNW_v2.indd 189 26-11-18 12:43

Page 192: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

190

Chapter 9

36. Kawamura M, Kusano A, Furuya A, et al. New sandwich-type enzyme-linked immunosorbent assay for human MxA protein in a whole blood using monoclonal antibodies against GTP-binding domain for recognition of viral infection. Journal of clinical laboratory analysis 2012; 26(3): 174-83.

37. Nakabayashi M, Adachi Y, Itazawa T, et al. MxA-based recognition of viral illness in febrile children by a whole blood assay. Pediatric research 2006; 60(6): 770-4.

38. Halminen M, Ilonen J, Julkunen I, Ruuskanen O, Simell O, Makela MJ. Expression of MxA protein in blood lymphocytes discriminates between viral and bacterial infections in febrile children. Pediatric research 1997; 41(5): 647-50.

39. Ronni T, Melen K, Malygin A, Julkunen I. Control of IFN-inducible MxA gene expression in human cells. Journal of immunology (Baltimore, Md : 1950) 1993; 150(5): 1715-26.

40. Simon L, Gauvin F, Amre DK, Saint-Louis P, Lacroix J. Serum procalcitonin and C-reactive protein levels as markers of bacterial infection: a systematic review and meta-analysis. Clin Infect Dis 2004; 39(2): 206-17.

41. Mahajan P, Kuppermann N, Mejias A, et al. Association of RNA Biosignatures With Bacterial Infections in Febrile Infants Aged 60 Days or Younger. Jama 2016; 316(8): 846-57.

42. Herberg JA, Kaforou M, Wright VJ, et al. Diagnostic Test Accuracy of a 2-Transcript Host RNA Signature for Discriminating Bacterial vs Viral Infection in Febrile Children. Jama 2016; 316(8): 835-45.

43. Tsalik EL, Henao R, Nichols M, et al. Host gene expression classifiers diagnose acute respiratory illness etiology. Sci Transl Med 2016; 8(322): 322ra11.

44. Ramilo O, Allman W, Chung W, et al. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood 2007; 109(5): 2066-77.

45. Hu X, Yu J, Crosby SD, Storch GA. Gene expression profiles in febrile children with defined viral and bacterial infection. Proceedings of the National Academy of Sciences of the United States of America 2013; 110(31): 12792-7.

46. van Houten CB, de Groot JA, Klein A, et al. A host-protein based assay to differentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study. Lancet Infect Dis 2017; 17(4): 431-40.

47. Srugo I, Klein A, Stein M, et al. Validation of a Novel Assay to Distinguish Bacterial and Viral Infections. Pediatrics 2017; 140(4).

48. Stein M, Lipman-Arens S, Oved K, et al. A novel host-protein assay outperforms routine parameters for distinguishing between bacterial and viral lower respiratory tract infections. Diagnostic microbiology and infectious disease 2018; 90(3): 206-13.

49. Ashkenazi-Hoffnung L, Oved K, Navon R, et al. A host-protein signature is superior to other biomarkers for differentiating between bacterial and viral disease in patients with respiratory infection and fever without source: a prospective observational study. European journal of clinical microbiology & infectious diseases 2018; 37(7): 1361-71.

50. Davidson M. FebriDx point-of-care testing to guide antibiotic therapy for acute respiratory tract infection in UK primary care: a retrosepctive outcome analysis. J Infect Dis Preve Med 2017; 5:165.

51. Thompson M, Van den Bruel A, Verbakel J, et al. Systematic review and validation of prediction rules for identifying children with serious infections in emergency departments and urgent-access primary care. Health Technol Assess 2012; 16(15): 1-100.

15873_CvHouten_BNW_v2.indd 190 26-11-18 12:43

Page 193: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

191

9

Summary, general discussion and future perspectives

52. Nijman RG, Vergouwe Y, Moll HA, et al. Validation of the Feverkidstool and procalcitonin for detecting serious bacterial infections in febrile children. Pediatric research 2017.

53. Nijman RG, Vergouwe Y, Thompson M, et al. Clinical prediction model to aid emergency doctors managing febrile children at risk of serious bacterial infections: diagnostic study. BMJ 2013; 346: f1706.

54. Matha SM, Rahiman SN, Gelbart BG, Duke TD. The utility of procalcitonin in the prediction of serious bacterial infection in a tertiary paediatric intensive care unit. Anaesthesia and intensive care 2016; 44(5): 607-14.

55. Lautz AJ, Dziorny AC, Denson AR, et al. Value of Procalcitonin Measurement for Early Evidence of Severe Bacterial Infections in the Pediatric Intensive Care Unit. J Pediatr 2016; 179: 74-81.e2.

56. Pinhu L, Whitehead T, Evans T, Griffiths M. Ventilator-associated lung injury. Lancet 2003; 361(9354): 332-40.

57. Launay E, Levieux K, Levy C, et al. Compliance with the current recommendations for prescribing antibiotics for paediatric community-acquired pneumonia is improving: data from a prospective study in a French network. BMC pediatrics 2016; 16(1): 126.

58. Dekker ARJ, Verheij TJM, Broekhuizen BDL, et al. Effectiveness of general practitioner online training and an information booklet for parents on antibiotic prescribing for children with respiratory tract infection in primary care: a cluster randomized controlled trial. The Journal of antimicrobial chemotherapy 2018.

59. Hallsworth M, Chadborn T, Sallis A, et al. Provision of social norm feedback to high prescribers of antibiotics in general practice: a pragmatic national randomised controlled trial. Lancet 2016; 387(10029): 1743-52.

60. Reitsma JB, Rutjes AW, Khan KS, Coomarasamy A, Bossuyt PM. A review of solutions for diagnostic accuracy studies with an imperfect or missing reference standard. J Clin Epidemiol 2009; 62(8): 797-806.

61. Bertens LC, Broekhuizen BD, Naaktgeboren CA, et al. Use of expert panels to define the reference standard in diagnostic research: a systematic review of published methods and reporting. PLoS Med 2013; 10(10): e1001531.

62. Moons KG, Grobbee DE. When should we remain blind and when should our eyes remain open in diagnostic studies? J Clin Epidemiol 2002; 55(7): 633-6.

63. NICE: Clinical guideline Fever in under 5s: assessment and initial management. Available from: https://www.nice.org.uk/guidance/cg160/resources/fever-in-under-5s-assessment-and-initial-management-35109685049029 (accessed 19-07-2016).

64. Bossuyt PM, Reitsma JB, Bruns DE, et al. The STARD statement for reporting studies of diagnostic accuracy: explanation and elaboration. The Standards for Reporting of Diagnostic Accuracy Group. Croat Med J 2003; 44(5): 639-50.

65. Shinkins B, Thompson M, Mallett S, Perera R. Diagnostic accuracy studies: how to report and analyse inconclusive test results. Bmj 2013; 346: f2778.

66. Handels RL, Wolfs CA, Aalten P, et al. Optimizing the use of expert panel reference diagnoses in diagnostic studies of multidimensional syndromes. BMC neurology 2014; 14: 190.

67. Bertens LC, van Mourik Y, Rutten FH, et al. Staged decision making was an attractive alternative to a plenary approach in panel diagnosis as reference standard. J Clin Epidemiol 2015; 68(4): 418-25.

68. Gabel MJ, Shipan CR. A social choice approach to expert consensus panels. Journal of health economics 2004; 23(3): 543-64.

15873_CvHouten_BNW_v2.indd 191 26-11-18 12:43

Page 194: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

10

15873_CvHouten_BNW_v2.indd 192 26-11-18 12:43

Page 195: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

10Nederlandse samenvatting(Dutch summary)

15873_CvHouten_BNW_v2.indd 193 26-11-18 12:43

Page 196: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 194 26-11-18 12:43

Page 197: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

195

10

Nederlandse samenvatting

Infecties zijn veelvoorkomend en vooral kinderen hebben hier met grote regelmaat last van. Op de leeftijd van 18 maanden heeft een kind gemiddeld acht episodes met een infectie doorgemaakt. Tijdige behandeling met antibiotica verbetert de uitkomst van patiënten met een infectie veroorzaakt door een bacterie. Echter, het merendeel van de infecties op de kinderleeftijd wordt door een virus veroorzaakt en ook bij volwassenen zijn veel infecties van virale aard. Antibiotica werken niet tegen virussen en zijn daarom niet geïndiceerd als er sprake is van een virale infectie. Helaas is het vaak niet mogelijk om op basis van klinische kenmerken onderscheid te maken tussen virale en bacteriële infecties. Zelfs als er gebruik wordt gemaakt van beschikbare bloedtesten is dit onderscheid niet goed te maken. Het missen van een bacteriële infectie kan voor de patiënt ernstige gevolgen hebben. Aan de andere kant, overmatig gebruik van antibiotica heeft ook nadelen, zowel voor de patiënt als voor de wereldwijde gezondheidszorg. Dit proefschrift beschrijft ons onderzoek naar de verschillende aspecten van dit diagnostische dilemma bij patiënten met infecties.

In de eerste twee hoofdstukken van dit proefschrift evalueerden we de gevolgen van het complexe diagnostische proces op het gebruik van antibiotica. Hierbij maakten we gebruik van een expert panel bestaande uit artsen met speciale interesse voor infectieziekten. We vroegen het panel om een virale of bacteriële diagnose toe te wijzen aan elke patiënt die deelnam aan onze onderzoeken. In hoofdstuk 2 presenteren we de karakteristieken van 188 kinderen die in het ziekenhuis kwamen met een infectie met het respiratoir syncytieel virus (RSV). Dit virus is een veelvoorkomende oorzaak van luchtweginfecties bij jonge kinderen. Het stellen van een goede diagnose bij kinderen met een RSV-infectie is lastig, omdat er naast de virale infectie ook een bacteriële infectie kan optreden (bacteriële co-infectie). Veertien procent van de kinderen werd door het expert panel gediagnosticeerd met een bacteriële co-infectie. De meeste van deze kinderen werden opgenomen op de intensive care. Alle bacteriële co-infecties werden behandeld met antibiotica. Een derde van alle kinderen met een RSV-infectie zonder veronderstelde bacteriële co-infecties werd onnodig behandeld met antibiotica. Kinderen met een bacteriële co-infectie maakten vaker een zieke indruk op de behandelend arts dan kinderen zonder bacteriële co-infectie. Er waren geen andere klinische kenmerken dan deze zieke indruk die significant verschilden tussen kinderen met en zonder bacteriële co-infectie. Dit resultaat benadrukt dat klinische kenmerken artsen onvoldoende kunnen helpen om onderscheid te maken tussen patiënten die baat hebben bij antibiotica en degenen die dat niet hebben.

Het doel van het onderzoek beschreven in hoofdstuk 3 was om overmatig antibiotica gebruik te vergelijken tussen kinderen en volwassenen met luchtweginfecties. Ook bij dit onderzoek maakten we gebruik van expert panels voor het stellen van de diagnose.

15873_CvHouten_BNW_v2.indd 195 26-11-18 12:43

Page 198: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

196

Chapter 10

Van alle luchtweginfecties was het aandeel virale infecties groter bij kinderen dan bij volwassenen (74% versus 38%). Daarentegen, kwam overmatig gebruik van antibiotica minder vaak voor bij kinderen dan bij volwassenen met virale luchtweginfecties (37% versus 83%). Deze bevindingen ondersteunen de behoefte aan effectieve diagnostiek om overmatig gebruik van antibiotica te voorkomen bij luchtweginfecties in patiënten van alle leeftijden.

Ten tweede onderzochten we de betrouwbaarheid van een diagnostische test en van een voorspelmodel om virale en bacteriële infecties van elkaar te onderscheiden. In hoofdstuk 4 beschrijven we de resultaten van een dubbelblinde, prospectieve, internationale validatiestudie (de OPPORTUNITY studie). De studie werd uitgevoerd bij kinderen onder de leeftijd van vijf jaar die zich in het ziekenhuis presenteerden met koorts. We bestudeerden een combinatie van eiwitten die in de bloedbaan worden afgegeven bij virale en bacteriële infecties. Deze eiwitten zijn: tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), interferon gamma induced protein-10 (IP-10) en C-reactive protein (CRP). Met dit onderzoek hebben we vastgesteld dat deze combinatietest een hoge diagnostische waarde heeft. Met name de dynamiek van TRAIL is veelbelovend. Dit is de eerste gepubliceerde biomarker waarvan de concentratie toeneemt bij virale infecties en daalt bij bacteriële infecties. De test is vooral nuttig voor het uitsluiten van bacteriële infecties. Het aantal fout-positieve diagnoses nam met 51% af als de combinatietest werd gebruikt in plaats van alleen CRP. Dit resultaat suggereert dat deze test de potentie heeft om overmatig antibiotica bij jonge kinderen te verminderen. Er zijn nu implementatiestudies nodig om te onderzoeken wat in de praktijk de toegevoegde waarde van deze combinatietest zal zijn op het aantal onnodige antibiotica voorschriften.

Naast biomarkers spelen klinische kenmerken een belangrijke rol voor artsen als ze een diagnose stellen bij kinderen met een infectie. In het onderzoek beschreven in hoofdstuk 5 hebben we daarom de bovengenoemde combinatietest geïntegreerd in een bestaand klinisch voorspelmodel. Dit gevalideerde voorspelmodel is speciaal ontwikkeld voor kinderen die zich met koorts in het ziekenhuis presenteren. Het model bevat klinische kenmerken en de biomarker CRP. We hebben dit voorspelmodel geüpdatet door CRP te vervangen door de combinatietest. Het nieuwe model was nauwkeuriger in het voorspellen van ernstige bacteriële infecties in vergelijking met het model met CRP als enige biomarker. Onze resultaten tonen aan dat de toegevoegde waarde van de combinatietest het grootst is bij kinderen met een laag tot matig risico (<40%) op ernstige bacteriële infecties zoals voorspeld door het oorspronkelijke voorspelmodel. Dit afkappunt kan daarom worden gebruikt om kosteneffectief gebruik van de combinatietest te optimaliseren.

15873_CvHouten_BNW_v2.indd 196 26-11-18 12:43

Page 199: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

197

10

Nederlandse samenvatting

In hoofdstuk 6 hebben we de OPPORTUNITY studie voortgezet in een internationale, prospectieve studie (The TAILORED Treatment studie). Het doel van deze studie was om een algoritme te ontwikkelen om virale en bacteriële infecties te onderscheiden bij zowel kinderen als volwassenen. Dit algoritme zal gebaseerd zijn op diverse biologische processen in het lichaam en wordt daarom ook wel een multi-omics aanpak genoemd. Deze multi-omics benadering is momenteel uniek voor het diagnosticeren van bacteriële infecties en kan als voorbeeld dienen voor toekomstige studies naar infectieziekten.Ten derde onderzochten we een referentiestandaard voor bacteriële infectie, namelijk een expert panel. De afwezigheid van een gouden standaard om bacteriële infecties te diagnosticeren maakt het lastig om betrouwbaar diagnostisch onderzoek uit te voeren. In alle onderzoeken gepresenteerd in dit proefschrift werd daarom gebruik gemaakt van een expert panel als referentiestandaard. Dit expert panel stelde vast of er sprake was van een bacteriële of virale infectie. In hoofdstuk 7 hebben we de reproduceerbaarheid van een dergelijke expert panel als referentiestandaard geëvalueerd. Individuele experts en expert panels diagnosticeerden een casus twee keer, met tijdsintervallen van zes weken en drie jaar. De experts stelden de diagnose zonder te weten dat ze dezelfde casus eerder beoordeeld hadden. We ontdekten dat een panel bestaande uit drie experts een meer reproduceerbare diagnose biedt dan de diagnose van een individuele expert. Zelfs met een tijdsinterval van drie jaar was de overeenkomst tussen de panel diagnoses hoog (kappa 0.80). Uitbreiding van het expert panel naar vijf of zeven experts had geen grote meerwaarde. Onze bevindingen ondersteunen de behoefte aan verdere validatie en optimalisatie van panel diagnoses om nauwkeurige resultaten te verkrijgen in geval van diagnostische studies zonder gouden standaard.

Tot slot geven we in hoofdstuk 8 een overzicht van de huidige en nieuwe diagnostische testen bij kinderen met luchtweginfecties. Vooral eiwit- en RNA-gebaseerde biomarkers, die in het bloed meetbaar zijn na een infectie, laten veelbelovende resultaten zien. Technieken die RNA-profielen identificeren zijn echter nog te complex en onvoldoende onderzocht om in de klinische praktijk te gebruiken. De combinatietest van biomarkers die aan de bloedbaan worden afgegeven bij virale en bacteriële infecties, is echter bijna klaar voor gebruik in de klinische praktijk. Deze combinatietest kan artsen helpen om terughoudender te worden bij het gebruik van antibiotica bij patiënten met milde of matig ernstige infecties.

15873_CvHouten_BNW_v2.indd 197 26-11-18 12:43

Page 200: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Addendum

15873_CvHouten_BNW_v2.indd 198 26-11-18 12:43

Page 201: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

AddendumContributing authors

Dankwoord(Acknowledgments)

List of publications

Curriculum vitae

15873_CvHouten_BNW_v2.indd 199 26-11-18 12:43

Page 202: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 200 26-11-18 12:43

Page 203: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

201

11

Contributing authors

Contributing authors

Jalal AshkarDepartment of Paediatrics, Hillel Yaffe Medical Centre, Hadera, Israel

Liat Ashkenazi-HoffnungSchneider Children’s Medical Center, Petach Tikva, Sackler Faculty of Medicine, Tel Aviv, Israel

Shai AshkenaziSchneider Children’s Medical Center, Petach Tikva, Sackler Faculty of Medicine, Tel Aviv, Israel

Wim AvisDepartment of Paediatrics, Gelderse Vallei Hospital, Ede, the Netherlands

Ellen BambergerDepartment of Paediatrics, Bnai Zion Medical Centre, Haifa, Israel

Stefan A. BoersDepartment of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, the Netherlands

Olga BoicoMeMed, Tirat Carmel, Israel

Louis J. BontDivision of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Aik W.J. BossinkDepartment of Respiratory Medicine, Diakonessenhuis Utrecht, Utrecht, the Netherlands

Itzhak BravermanDepartment of Paediatrics, Hillel Yaffe Medical Centre, Hadera, Israel

Brigitte J.M. BuitemanDivision of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Irena ChistyakovDepartment of Paediatrics, Bnai Zion Medical Centre, Haifa, Israel

15873_CvHouten_BNW_v2.indd 201 26-11-18 12:43

Page 204: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

202

Addendum

Asi CohenMeMed, Tirat Carmel, Israel

Laurence V. CollinsIBEXperts Ltd, Ra’anana, Israel

Teresa CoriglianoDepartment of Paediatrics, Geneva University Hospitals, Geneva, Switzerland

Yaniv DotanDepartment of Internal Medicine, Bnai Zion Medical Centre, Haifa, Israel

Eran EdenMeMed, Tirat Carmel, Israel

Dan EngelhardDivision of Paediatric Infectious Disease Unit, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel

Liat EtshteinMeMed, Tirat Carmel, Israel

Paul D. FeiginFaculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa, Israel

David FernándezNoray Bioinformatics, Derio, Spain

Tom FriedmanMeMed, Tirat Carmel, Israel

Annick GalettoDepartment of Paediatrics, Geneva University Hospitals, Geneva, Switzerland

Iker GangoitiDepartment of Paediatric Emergency Medicine, Cruces University Hospital, Bilbao, Basque Country, Spain

Alain GervaixDepartment of Paediatrics, Geneva University Hospitals, Geneva, Switzerland

15873_CvHouten_BNW_v2.indd 202 26-11-18 12:43

Page 205: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

203

11

Contributing authors

Daniel GlikmanThe faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel

Eva GonzalezNoray Bioinformatics, Derio, Spain

Tanya M. GottliebMeMed, Tirat Carmel, Israel

Joris A.H. de GrootDivision Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, the Netherlands

John HaysDepartment of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, the Netherlands

Rik HeijligenbergDepartment of Internal Medicine, Gelderse Vallei Hospital, Ede, the Netherlands

Inga IvaskevicieneDepartment of Paediatrics, Vilnius University Hospital, Vilnius, Lithuania

Roger KarlssonDepartment of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy,

University of Gothenburg, Gothenburg, Sweden

Valery KartunDepartment of Paediatrics, Hillel Yaffe Medical Centre, Hadera, Israel

Adi KleinDepartment of Paediatrics, Hillel Yaffe Medical Centre, Hadera, Israel

Robert KraaijDepartment of Internal Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands

Racheli KreisbergIBEXperts Ltd, Ra’anana, Israel

Gali KronenfeldMeMed, Tirat Carmel, Israel

15873_CvHouten_BNW_v2.indd 203 26-11-18 12:43

Page 206: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

204

Addendum

Amir A. KupermanBlood Coagulation Service and Paediatric Haematology Clinic, Galilee Medical Centre, Nahariya, and Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel

Laurence LacroixDepartment of Paediatrics, Geneva University Hospitals, Geneva, Switzerland

Maaike van der LeeDivision of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Yunlei LiClinical Bioinformatics Unit, Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands

Yvette LoeffenDivision of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Bart LuijkDepartment of Respiratory Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Fanny LuterbacherDepartment of Paediatrics, Geneva University Hospitals, Geneva, Switzerland

Niv MastboimMeMed, Tirat Carmel, Israel

Josephine S. van de MaatDepartment of Paediatrics, Erasmus MC University, Medical Centre Rotterdam, the Netherlands

Pieter W. MeijersDepartment of Paediatrics, Gelderse Vallei Hospital, Ede, the Netherlands

Clemens B. MeijssenDepartment of Paediatrics, Meander Medical Centre, Amersfoort, the Netherlands

Santiago MintegiDepartment of Paediatric Emergency Medicine, Cruces University Hospital, Bilbao, Basque Country, Spain

15873_CvHouten_BNW_v2.indd 204 26-11-18 12:43

Page 207: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

205

11

Contributing authors

Edward R.B. MooreDepartment of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy,

University of Gothenburg, Gothenburg, Sweden

Christiana A. NaaktgeborenDivision Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, the Netherlands

Basheer NasrallahDepartment of Paediatrics, Bnai Zion Medical Centre, Haifa, Israel

Roy NavonMeMed, Tirat Carmel, Israel

Majed OdehDepartment of Paediatrics, Bnai Zion Medical Centre, Haifa, Israel

Rianne OostenbrinkDepartment of Paediatrics, Erasmus MC University, Medical Centre Rotterdam, the Netherlands

Jan Jelrik OosterheertDepartment of Internal Medicine and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Kfir OvedMeMed, Tirat Carmel, Israel

Cihan PapanPaediatric Infectious Diseases, University Children’s Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany

Meital PazMeMed, Tirat Carmel, Israel

Ester Pri-OrMeMed, Tirat Carmel, Israel

Ronit RachmilevitchDepartment of Paediatrics, Hillel Yaffe Medical Centre, Hadera, Israel

15873_CvHouten_BNW_v2.indd 205 26-11-18 12:43

Page 208: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

206

Addendum

Sharon ReisfeldDepartment of Paediatrics, Hillel Yaffe Medical Centre, Hadera, Israel

Annemarie M.C. van RossumDepartment of Paediatrics, Erasmus MC University, Medical Centre Rotterdam, the Netherlands

Henriette RudolphDepartment of Paediatrics, University Children’s Hospital Mannheim, Mannheim, Germany

Elisabeth A.M. SandersDivision of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Sanjay U.C. SankatsingDepartment of Internal Medicine, Diakonessenhuis Utrecht, Utrecht, the Netherlands

Yael Schachor-MeyouhasDepartment of Paediatric Infectious Disease, Ruth Rappaport Children’s Hospital, Rambam Health Care Campus, Haifa, Israel

Elad SchiffDepartment of Paediatrics, Bnai Zion Medical Centre, Haifa, Israel

Liran ShaniMeMed, Tirat Carmel, Israel

Einav SimonMeMed, Tirat Carmel, Israel

Isaac SrugoDepartment of Paediatrics, Bnai Zion Medical Centre, Haifa, Israel

Michal SteinDepartment of Paediatrics, Hillel Yaffe Medical Centre, Hadera, Israel

Andrew P. StubbsClinical Bioinformatics Unit, Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands

15873_CvHouten_BNW_v2.indd 206 26-11-18 12:43

Page 209: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

207

11

Contributing authors

Roei TalDepartment of Paediatrics, Galilee Medical Centre, Nahariya and Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel

Tobias TenenbaumPaediatric Infectious Diseases, University Children’s Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany

Vytautas UsonisDepartment of Paediatrics, Vilnius University Hospital, Vilnius, Lithuania

Wouter de WaalDepartment of Paediatrics, Diakonessenhuis, Utrecht, the Netherlands

Stefan WeichertPaediatric Infectious Diseases, University Children’s Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany

Joanne G. WildenbeestDivision of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Karin M. de Winter-de GrootDepartment of Paediatric Respiratory Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

Tom F.W. WolfsDivision of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands

15873_CvHouten_BNW_v2.indd 207 26-11-18 12:43

Page 210: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 208 26-11-18 12:43

Page 211: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

209

11

Dankwoord

D ankwoord

‘Grote dingen worden nooit door één persoon gedaan.’ Dit proefschrift is dan ook met de steun en inspanningen van velen tot stand gekomen en ik wil iedereen daarvoor heel erg bedanken. Een aantal mensen wil ik in het bijzonder bedanken.

Allereerst natuurlijk alle patiënten en hun ouders die hebben deelgenomen aan mijn onderzoeken, jullie vormen de basis van dit proefschrift. Ik vond het bijzonder om te zien hoe iedereen zonder eigenbelang wilde meewerken aan mijn onderzoek (inclusief de bloedafnames). Door jullie wist ik altijd weer precies waarom ik voor dit promotietraject heb gekozen.

Mijn promotoren, wat is jullie enthousiasme en positieve instelling aanstekelijk!Prof. Dr. L.J. Bont, beste Louis. Ik heb heel veel bewondering voor hoe jij jouw passie voor patiëntenzorg en wetenschappelijk onderzoek combineert. Je betrokkenheid is noemenswaardig en ondanks je overvolle agenda maakte je altijd tijd voor me vrij. Je gaf me de ruimte om mijn ideeën voor onderzoek uit te voeren en me hierbij te helpen wanneer dat nodig was. Ik vond het bijzonder om te merken dat in elk overleg en bij elke beslissing mijn ontwikkeling en carrière bij jou voorop stonden. Heel erg bedankt voor de kansen die je me hebt gegeven en ik hoop nog vele jaren van je te mogen leren. Prof. Dr. E.A.M. Sanders, beste Lieke. Al bij onze eerste afspraak gaf je aan graag betrokken te zijn bij mijn promotietraject en zo gebeurde het. Ondanks je vertrek naar het RIVM kon ik altijd bij je aankloppen voor overleg of advies. Wat was het een feest om met jou naar nieuwe data te kijken, jij zorgde ervoor dat ik alle data nog beter wilde begrijpen. Bedankt voor je motiverende ideeën en oprechte visie op het onderzoek.

De leden van de leescommissie; Prof. dr. A.I.M. Hoepelman (voorzitter), Prof. dr. T.J.M. Verheij, Prof. dr. A.M.C. van Rossum, Prof. dr. K.G.M. Moons en Prof. dr. N.M. Wulffraat, wil ik hartelijk bedanken voor hun tijd en bereidheid om mijn proefschrift te beoordelen.

Alle kamergenoten, heel erg bedankt voor al jullie gezelligheid en steun. Samen met de arts-onderzoekers op de gang (Maartje, Charlotte en Natalie) maakten jullie onderzoek doen nog leuker. Linde, ik ben blij dat ik van begin tot eind met jou een kamer heb mogen delen. Wat heb ik bewondering voor je enthousiasme, je luisterend oor en de positieve instelling waarmee je in het leven staat. Stephanie, Mieke en Maarten, wat was het fijn om als onervarener onderzoeker bij jullie op de kamer te komen en van jullie te mogen leren. Laura, wat konden we het vanaf moment één goed met elkaar vinden. Met plezier denk ik terug aan onze gesprekken over hoe we wetenschap in het WKZ beter

15873_CvHouten_BNW_v2.indd 209 26-11-18 12:43

Page 212: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

210

Addendum

op de kaart konden zetten en kijk waar we nu staan! Koos, Merel en Annefleur, bedankt voor jullie support bij het afronden van mijn promotie. De, soms wat competitieve, koffiemomenten zijn goud waard en hopelijk houden jullie die er op de flexplekken in!

Onderzoeksverpleegkundigen Maaike en Brigitte, zonder jullie was dit proefschrift er nooit gekomen! Wat hebben jullie je ingezet om alle patiënten in de studies te krijgen en om alle data te verzamelen. Bedankt voor al het geduld en het teamgevoel dat jullie wisten te creëren. Wouter, nog nooit heb ik zo’n zorgvuldige en betrokken student gezien als jij, bedankt dat je onderdeel van het team wilde zijn.

De RSV-onderzoeksgroep, door de parel van de week werd elke mini-succesje waardevol en was er altijd wel een reden voor taart. Bedankt voor alle feedback op presentaties, posters en artikelen. Nienke, wat was het fijn om dit hele traject tegelijk met doorlopen te hebben, een blik zei vaak al genoeg. Ik zal onze congresbezoeken nooit vergeten. Loes, wat heb jij een groot sociaal hart. Bedankt dat je altijd oog had voor mij als persoon achter de onderzoeker. Joris en Christiana, ‘R scripts’ blijven voor mij alsof ik een Chinees boek lees, enorm bedankt voor al jullie statistische hulp en adviezen.

Alle collega’s uit de verschillende deelnemende ziekenhuizen; UMC Utrecht, Diakonessenhuis Utrecht, Meander Medisch Centrum Amersfoort, Ziekenhuis Gelderse Vallei en Jeroen Bosch Ziekenhuis. Bedankt voor alle hulp bij het includeren van patiënten en voor het diagnosticeren van de casuïstiek. Het was een groot plezier om met jullie te werken en om te mogen leren van jullie expertise.Special thanks go to MeMed’s team (under supervision of Kfir and Eran) for the collaboration in all projects presented in this thesis. I really enjoyed the fruitful discussions and it was a great experience to visit your company and the participating centers in Israel.

Ik ben enorm dankbaar voor alle kansen die ik door bovenstaande collega’s heb gekregen. Toch had ik dit alles niet kunnen doen zonder de belangrijkste mensen in mijn leven; mijn familie en vrienden.

Mijn paranimfen Stephanie en Hester, wat ben ik blij dat jullie op deze belangrijke dag naast me staan! Hester, al meer dan 15 jaar delen we lief en leed met elkaar. Op de belangrijke momenten ben je er voor me en ik wist dan ook gelijk dat jij mijn paranimf moest worden. Wat heb ik een bewondering voor je enorme doorzettingsvermogen, je laat je door niets tegenhouden om je doelen te bereiken. Ik heb er alle vertrouwen in dat je inzet, enthousiasme en collegialiteit beloond gaat worden. Stephanie, jij begrijpt me en weet precies wat ik nodig heb. Ik koester de avonden waarop we samen grote

15873_CvHouten_BNW_v2.indd 210 26-11-18 12:43

Page 213: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

211

11

Dankwoord

stappen in ons onderzoek wisten te zetten en wat was het bijzonder om vorig jaar jouw paranimf te mogen zijn. Je enorme inzet voor je werk, gezin en vrienden is geweldig. Ik kijk er naar uit om over enige tijd samen te werken in de patiëntenzorg.

Al mijn lieve vrienden en vriendinnen die zorgen voor de ontspanning in mijn leven. Lisa en Sarah, wat was het fijn om met elkaar de paniek- en pleziermomenten van promoveren te delen. Bedankt voor jullie enorme steun, maar vooral voor alle gezellige borrels, etentjes, feestjes en alle andere momenten waarop we werk even heel ver achter ons konden laten. Marjolein, ook jij was er altijd voor de nodige afleiding. Een musicalavond of bijkletsen met wijn en chocoladerozijnen (met als toetje een raketje), een avond samen met jou is altijd een succes.

Mijn tennisteam, door jullie staat gezelligheid elke zaterdag weer voorop. Bedankt dat jullie me op deze dagen helpen herinneren dat ik niet altijd hoef te presteren.

Mijn schoonfamilie, Stef, Joke en Manouk. Altijd weer geïnteresseerd in mijn promotieonderzoek én in mij. Bedankt dat ik onderdeel mag zijn van jullie familie.

Opa en Lenny, wat ben ik blij dat jullie aanwezig zijn bij mijn promotie! Bedankt voor jullie enorme steun, vertrouwen en betrokkenheid. Ik hoop nog veel aangeklede borrels met jullie te mogen hebben.

Patrick, ‘broertje’, al vanaf mijn eerste coschap moest jij niets hebben van al mijn medische verhalen, wat zal jij dan ook blij geweest zijn toen ik ging promoveren. Bedankt voor je humor en je nuchtere kijk op de wereld waar ik nog veel van kan leren. Diewertje, jij maakt de familie compleet en met jouw bakkunsten worden de feestdagen een nog groter feest.

Lieve papa en mama, eigenlijk zijn er geen woorden die beschrijven hoe dankbaar ik ben om jullie als ouders te hebben. Jullie hebben altijd in me geloofd en steken jullie trots niet onder stoelen of banken. Bedankt dat jullie echt altijd voor me klaar staan en ervoor zorgen dat ik met volle overtuiging mijn dromen naleef.

Lieve Yannick, samen met jou ben ik het gelukkigst. Dank je wel voor al je hulp en geduld als ik weer eens gefrustreerd was, omdat het niet (zo snel) ging als ik wilde. Je relativeringsvermogen en droge humor had ik tijdens dit promotietraject niet willen missen. Dank je wel voor het enorme vertrouwen dat je me geeft. Ik heb heel veel zin in onze verdere toekomst samen!

15873_CvHouten_BNW_v2.indd 211 26-11-18 12:43

Page 214: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

15873_CvHouten_BNW_v2.indd 212 26-11-18 12:43

Page 215: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

213

11

List of publications

List of public ations

In this thesisvan Houten CB, Naaktgeboren CA, Buiteman BJM, van der Lee M, Klein A, Srugo I, Chistyakov I, de Waal W, Meijssen CB, Meijers PW, de Winter-de Groot KM, Wolfs TFW, Shachor-Meyouhas Y, Stein M, Sanders EAM, Bont LJ. Antibiotic Overuse in Children with Respiratory Syncytial Virus Lower Respiratory Tract Infection. Pediatr Infect Dis J. 2018 Nov;37(11):1077-1081.

van Houten CB, Cohen A, Engelhard D, Hays JP, Karlsson R, Moore ERB, Fernández D, Kreisberg R, Collins LV, de Waal W, de Winter-de Groot KM, Wolfs TFW, Meijers P, Luijk B, Oosterheert JJ, Heijligenberg R, Sankatsing SUC, Bossink AWJ, Stubbs AP, Stein M, Reisfeld S, Klein A, Rachmilevitch R, Ashkarq J, Braverman I, Kartun V, Chistyakov I, Bamberger E, Srugo I, Odeh M, Schiff E, Dotan Y, Boico O, Navon R, Friedman T, Etshtein L, Paz M, Gottlieb TM, Pri-Or E, Kronenfeld G, Simon E, Oved K, Eden E, Bont LJ. Antibiotic misuse in respiratory tract infections in children and adults – a prospective, multicentre study (TAILORED Treatment) Submitted.

van Houten CB, de Groot JAH, Klein A, Srugo I, Chistyakov I, de Waal W, Meijssen CB, Avis W, Wolfs TFW, Sanders EAM, Bont LJ. A host-protein based assay to differentiate between bacterial and viral infections in preschool children (OPPORTUNITY): a double-blind, multicentre, validation study. Lancet Infect Dis. 2017 Apr;17(4):431-440.

van Houten CB, van de Maat JS, Naaktgeboren CA, Bont LJ, Oostenbrink R. Optimizing prediction of serious bacterial infections by a host-protein assay. Submitted.

van Houten CB, Oved K, Eden E, Cohen A, Engelhard D, Boers SA, Kraaij R, Karlsson R, Fernandez D, Gonzalez E, Li Y, Stubbs AP, Moore ERB, Hays JP, Bont LJ. Observational multi-centre, prospective study to characterize novel pathogen-and host-related factors in hospitalized patients with lower respiratory tract infections and/or sepsis – The “TAILORED-Treatment” Study. BMC Infect Dis. (2018) 18:377.

van Houten CB, Shani L, Naaktgeboren CA, Ashkenazi-Hoffnung L, Ashkenazi S, , Avis W, Chistyakov I, Corigliano T, Galetto A, Gangoiti I, Gervaix A, Glikman D, Ivaskeviciene I, Kuperman AA, Lacroix L, Loeffen Y, Luterbacher F, Meijssen CB, Mintegi S, Nasrallah B, Papan C, van Rossum AMC, Rudolph H, Stein M, Tal R, Tenenbaum T, Usonis V, de Waal W, Weichert S, Wildenbeest JG, de Winter-de Groot KM, Wolfs TFW, Mastboim N, Gottlieb TM, Cohen A, Oved K, Eden E, Bont LJ. Reproducibility of an expert panel diagnosis as reference standard in infectious diseases. Submitted.

15873_CvHouten_BNW_v2.indd 213 26-11-18 12:43

Page 216: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

214

Addendum

van Houten CB, Bont LJ. A new diagnostic strategy for children with lower respiratory tract infections. Nederlands tijdschrift voor Medische Microbiologie, 2018;26:nr3.

Other publicationsHeikema AP, Horst-Kreft D, Boers SA, Jansen R, de Ridder MAJ, van Houten CB, Bont LJ, Stubbs AP, Hays JP. Nanopore 16S rRNA gene sequencing of the human nasal microbiota indicates the (bioinformatics) loss of established genera and shows high bacterial species diversity. Submitted.

Hopmans TE, van Houten CB, Kasius A, Kouznetsova OI, Nguyen LA, Rooijmans SV, Voormolen DN, van Vliet EO, Franx A, Koster MP. Increased risk of type II diabetes mellitus and cardiovascular disease after gestational diabetes mellitus: a systematic review. Ned Tijdschr Geneeskd. 2015;159:A8043.

Chapters in booksde Groot RCA, van Houten CB, van Rossum AMC, Bont LJ. Lower respiratory tract infections. Book: Werkboek Kinderinfectieziekten, 2018.

15873_CvHouten_BNW_v2.indd 214 26-11-18 12:43

Page 217: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

215

11

Curriculum vitae

Curric ulum vitae

Chantal van Houten was born on the 11th of April 1989 in Soest, the Netherlands, where she grew up and attended primary school. She graduated from secondary school in 2007 at the Meridiaan college, Het Nieuwe Eemland, Amersfoort. In that same year she started medical training at Utrecht University. Her passion for paediatric medicine was born during her paediatric internship and grew throughout her final internship on the ward of the Wilhelmina Children’s Hospital (WKZ) in Utrecht.

After graduating from medical school in 2013, Chantal started a PhD project in the Department of Paediatric Immunology and Infectious Diseases at the WKZ. The research described in this thesis was performed under the supervision of Prof. dr. L.J. Bont and Prof. dr. E.A.M. Sanders. In 2016 she started the research committee in the WKZ to connect researchers and to close the gap between physicians and researchers. She interrupted her PhD project in 2017 to work as a resident not in training (ANIOS) in the Department of Paediatrics in the Meander Medical Centre in Amersfoort under supervision of Dr. J. Jansen. In 2018 she returned to the WKZ to finish this thesis.

In February 2019 Chantal will start the paediatric residency-training (AIOS) at the WKZ, Utrecht (supervisors Prof. Dr. J. Frenkel and Drs. J. Smal) and at the Gelre Hospital, Apeldoorn (supervisors Drs. A.H. Robroch and Dr. J.H. Oudshoorn).

Chantal lives with Yannick ‘t Mannetje in Utrecht, the Netherlands.

15873_CvHouten_BNW_v2.indd 215 26-11-18 12:43

Page 218: Improving UITNODIGING diagnosis of bacterial infections€¦ · and bacterial infections is not new. Starting in the 1980’s, the Yale Observation Scale27 and the Rochester Criteria28,

Improving diagnosis of

bacterial infections

Improving diagnosis of bacterial infections

Chantal van Houten

Research without gold standard

Chantal van Houten

UITNODIGINGvoor het bijwonen van

de openbare verdedigingvan het proefschrift

Improving diagnosis of bacterial infections

Research without gold standard

door

Chantal van Houten

Op dinsdag 22 januari 2019om 14.30 uur

in het Academiegebouw,Domplein 29 te Utrecht

Aansluitend bent u van harte welkom op

de receptie ter plaatse

Chantal van HoutenPieter Saenredamstraat 11

3583 TA [email protected]

ParanImfenStephanie Smetsers

[email protected]

Hester de [email protected]