chisha teza sikazwe - research-repository.uwa.edu.au · the effective contribution of viral...

213
The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe BSc (Biomedical Sciences and Molecular biology) Master of Infectious Diseases This thesis is presented for the degree of Doctor of Philosophy in Microbiology of The University of Western Australia School of Biomedical Sciences 2018

Upload: others

Post on 20-Sep-2019

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

The Effective Contribution of Viral Respiratory Infection to Wheezing Illness

in Hospitalised Young Children

Chisha Teza Sikazwe

BSc (Biomedical Sciences and Molecular biology)

Master of Infectious Diseases

This thesis is presented for the degree of Doctor of Philosophy in Microbiology of

The University of Western Australia

School of Biomedical Sciences

2018

Page 2: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

ii

Page 3: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

iii

Thesis declaration

I, Chisha Teza Sikazwe, certify that:

This thesis has been substantially accomplished during enrolment in the degree.

This thesis does not contain material which has been accepted for the award of any

other degree or diploma in my name, in any university or other tertiary institution.

No part of this work will, in the future, be used in a submission in my name, for an y

other degree or diploma in any university or other tertiary institution without the

prior approval of The University of Western Australia and where applicable, any

partner institution responsible for the joint-award of this degree.

This thesis does not contain any material previously published or written by another

person, except where due reference has been made in the text.

The work(s) are not in any way a violation or infringement of any copyright,

trademark, patent, or other rights whatsoever of any person.

Technical assistance was kindly provided by Tom Chung for Cytokine multiplex bead

experiment that is described in Chapter seven of this thesis.

This thesis contains only sole-authored work, some of which has been published

and/or prepared for publication under sole authorship.

XChisha Teza Sikazwe

Date: 25.06.2018

Page 4: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

iv

Abstract

Respiratory tract infections are a leading cause of morbidity and mortality worldwide. Our

emerging understanding of the importance of acute lower respiratory viral infe ction in

early childhood predisposing to chronic inflammatory respiratory disease, coupled with the

complex interplay between virus and host underscores the need for investigations to

understand the clinical manifestation and the interplay between the magnitude of

infection and the corresponding host response. Rhinoviruses, specifically RV -C have been

identified as an important contributor to wheezing illness in paediatric medicine. Though,

little is known about its contribution to infection. Thus, this the sis aims to improve

understanding of the underlying pathophysiological mechanisms in the context of RV -C

wheezing illness through the development of a reliable method of quantifying RV -C load

and characterisation of the host response following infection.

RV-C was the most common virus detected in preschool aged children hospitalised with an

acute asthma exacerbation (Chapter 6). This was also apparent in preschool aged non-

asthmatic children hospitalised with a wheezing illness (chapter 7). Interestingly, i n young

infants under the age of two years hospitalised (Chapter 5) with a wheezing illness, RSV

rather than RV-C was found to be the most common virus detected. Viral load studies

revealed that both RSV and RV-C replicate to significantly higher levels in hospitalised

patients than in non-respiratory disease controls. In asthmatics and non-asthmatic

patients, RV-C induced respiratory wheeze appears to be driven by a Th2 response that is

independent of viral load. The magnitude of the host immune response is more apparent

in children with asthma compared to those without asthma. In addition, RV-C infection in

pre-school aged children with wheeze appears to induce a selective increase in neutrophil

chemokines and a significant increase in neutrophil numbers with a simultaneous increase

in illness severity.

Page 5: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

v

The findings in this thesis extend the existing knowledge on RV-C mediated wheezing

illness and demonstrate that magnitude of replication does not significantly contribute to

disease outcome. Conversely, it appears that the host immune response for which in part

is driven by a pro-inflammatory, neutrophilic inflammation may play a substantial role in

severity of disease. Host directed therapy targeting neutrophil pathways may prove to be

beneficial in young children infected with RV-C.

Page 6: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

vi

Table of contents

Thesis declaration ......................................................................................................... iii

Abstract........................................................................................................................ iv

List of Figures................................................................................................................. x

List of Tables ................................................................................................................ xii

Acknowledgments....................................................................................................... xiii

AUTHORSHIP DECLARATION: SOLE AUTHOR PUBLICATIONS .................................... xv

Peer reviewed papers and Conference presentations .................................................... xvi

List of Abbreviations................................................................................................... xvii

1 Literature review .................................................................................................... 2

1.1 Introduction ............................................................................................ 3

1.1.1 Respiratory syncytial virus ........................................................................ 6

1.1.2 Rhinoviruses ............................................................................................ 6

1.1.3 Human Metapneumovirus ........................................................................ 8

1.1.4 Influenza virus ......................................................................................... 8

1.1.5 Parainfluenza virus ................................................................................... 9

1.1.6 Adenovirus ............................................................................................ 10

1.1.7 Human Coronaviruses ............................................................................ 10

1.2 Laboratory Diagnosis of Respiratory Infections ................................................... 11

1.2.1 Upper and Lower airway samples............................................................ 12

1.2.2 Virus isolation by cell culture .................................................................. 13

1.2.3 Direct detection of respiratory viruses by immunofluorescence ................ 14

1.2.4 Nucleic Acid Tests .................................................................................. 15

1.3 Viral kinetics of acute respiratory tract infections................................................ 19

1.4 Viral respiratory infection and asthma................................................................ 20

1.5 Innate immune response to viral respiratory infection ........................................ 23

Page 7: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

vii

1.6 Aims of Project ................................................................................................. 26

2 Materials and Methods ......................................................................................... 27

2.1 Sample collection .............................................................................................. 27

2.2 Nucleic acid extraction and viral detection.......................................................... 28

2.3 Design of primers and probes ............................................................................ 29

2.4 Production and quantification of transcribed RNA standards ............................... 31

2.5 Quantitative Real time PCR (Viral load)............................................................... 32

2.6 Digital Droplet PCR ............................................................................................ 33

2.7 PCR reagents .................................................................................................... 34

2.8 Gel Electrophoresis ........................................................................................... 34

2.9 Thermocyclers .................................................................................................. 34

2.10 Computational analysis ..................................................................................... 35

2.11 Multiplex immunoassay..................................................................................... 35

2.12 Quality Control ................................................................................................. 36

2.13 Statistical Analysis ............................................................................................. 36

2.14 Ethics Approval ................................................................................................. 36

3 The design and development of quantitative detection assays for the common

causative viral pathogens of acute lower respiratory tract infection ................................ 38

3.1 Introduction ..................................................................................................... 39

3.2 Samples............................................................................................................ 41

3.3 Results ............................................................................................................. 41

3.4 Discussion......................................................................................................... 55

4 The development of a reliable PCR assay to measure RV-C load in clinical samples ... 61

4.1 Introduction ..................................................................................................... 62

4.2 Samples............................................................................................................ 64

4.3 Results ............................................................................................................. 66

4.4 Discussion......................................................................................................... 75

5. The Quantitative Detection of Respiratory Syncytial Virus in Hospitalized Young South

African Children ........................................................................................................... 78

5.1 Introduction ..................................................................................................... 79

5.2 Samples............................................................................................................ 81

5.3 Results ............................................................................................................. 82

5.3.1 Baseline characteristics .......................................................................... 82

5.3.2 RSV infection and clinical outcome .......................................................... 84

Page 8: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

viii

5.3.3 RSV disease and HIV infection ................................................................. 87

5.3.4 RSV load .................................................................................................... 88

5.4 Discussion......................................................................................................... 91

6 Determinants of acute asthma exacerbation severity following RV-C infection ......... 95

6.1 Introduction ..................................................................................................... 96

6.2 Samples............................................................................................................ 97

6.3 Results ............................................................................................................. 99

6.3.1 Study participants characteristics ............................................................ 99

6.3.2 Virus Detection ...................................................................................... 99

6.3.3 RV-C load..............................................................................................101

6.3.4 Surrogate markers of inflammation........................................................104

6.3.5 Performance of RV-C load, neutrophils and eosinophils in predicting the

severity AAE ........................................................................................................108

6.4 Discussion........................................................................................................109

7. Cytokine profiles in nasal secretions of patients hospitalised with Rhinovirus Species C

associated respiratory wheeze .....................................................................................115

7.1 Introduction ....................................................................................................116

7.2 Samples...........................................................................................................117

7.3 Results ............................................................................................................119

7.3.1 Virus Detections....................................................................................119

7.3.2 Nasal cytokine profiles of wheezing patients following RV-C infection ......124

7.3.3 Relationships between cytokines, RV-C load and clinical outcomes ..........132

7.4 Discussion........................................................................................................135

8. General Discussion and Conclusions......................................................................142

8.1 Introduction .........................................................................................143

8.2 Epidemiology of respiratory viruses in young children under the age of five

years 145

Page 9: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

ix

8.3 Reliable methods of accurately determining viral load in RV-C infected

patients 148

8.4 Viral Determinants of severity of RV-C induced wheezing illness ..............150

8.6 Conclusion .......................................................................................................154

Bibliography ...............................................................................................................156

Appendices .................................................................................................................176

Appendix 1 ..........................................................................................................176

Appendix 2 .................................................................................................................181

Published work completed during PhD .........................................................................181

Page 10: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

x

List of Figures

Figure 4.3-1 A BioEdit sequence alignment of primer and probe regions that were targeted

by assays one to four. Sequences of forward primer region (left box), probe region (center

box) and reverse primer region (right box). Identical bases at the same position are

represented by dots whereas capitalized bases indicate mismatches between sequences.

................................................................................................................................... 66

Figure 4.3-2 the algorithm for the determination of RV-C viral load in clinical samples ..... 72

Figure 4.3-3: Box plots of RV-C load in samples from young children presenting to the

Emergency Department with acute wheeze. .................................................................. 74

Figure 5.3-1: Viruses detected in NPAs collected from ALRI cases and NRD controls. RV

(RV-A, RV-B, RV-C), HCoV (OC43, 229E, HKU-1, NL63), HPIV (PIV I-IV), IFV (A/H1N1, A/H3N2,

B and C) ....................................................................................................................... 84

Figure 5.3-2: The distribution of RSV positive and RSV-negative ALRI cases by age. RSV

disease was more prevalent in children within their first year of life. Peak hospitalization

rate was observed in the 0-2 month age group. ............................................................ 86

Figure 5.3-3: Distribution of respiratory viruses detected in HIV infected ALRI patients .... 87

Figure 5.3-4: A box plot comparing RSV load between ALRI cases and NRD controls......... 88

Figure 5.3-5 A box plot comparing RSV load by subtype and clinical diagnosis.................. 89

Figure 5.3-6: A box plot comparing RSV loads in ALRI cases with a viral co-infection [n=12;

RSV with either hAdV (75%, n=9), hCoV (17%, n=2) or RV (8%, n=1)] versus sole RSV

infection (n=15). .......................................................................................................... 90

Figure 6.3-1 A bar graph comparing the frequency (%) of viruses detected in cases and

controls. Rhinovirus-C (RV-C), Rhinovirus-A (RV-A), parainfluenza virus (PIV), respiratory

syncytial virus (RSV), human adenovirus (hAdV), Influenza viruses (IFV), human corona

virus (hCoV) Rhinovirus-B (RV-B) and human metapneumovirus (hMPV) ........................100

Fig 6.3-2: Box plot summarising RV-C load in children hospitalised with an acute asthma

exacerbation (cases) and otherwise healthy individuals with a non-respiratory disease

(controls). Median RV-C load of AAE cases was 2.6 log10 copies/mL higher than that of the

non-respiratory disease control group . ........................................................................102

Figure 6.3-3: A boxplot of RV-C loads for cases(n=21) stratified by disease severity and

controls (n=2). Groups were compared using the Mann-Whitney U test,. median viral load

between the two severity groups did not differ significantly. .........................................103

Figure 6.3-4 Surrogate markers of asthma exacerbation in acute samples from patients

infected with RV-C stratified by illness severity. A) Absolute neutrophil peripheral blood

counts B) absolute eosinophils peripheral blood counts C) Total serum IgE levels counts.

Mann- Whitney U or Kruksall-Wallis test were used for testing on the apporpriate group of

subjects. All data are expressed as box and whisker plots. *:p=0.05 (severe vs non-severe)

** p<0.001 (all AAE cases vs NRD controls) ...................................................................105

Figure 6.3-6 Receiver operator characteristic curves (ROC) of markers of AAE severity. A)

Absolute neutrophil count and b) RV-C load, eosinophil count and serum IgE. ................108

Figure 7.3-1: Virus detection rates from samples of children hospitalised with respiratory

wheeze. RV-rhinoviruses, respiratory syncytial virus (RSV), adenovirus (ADV), human

parainfluenza virus (HPIV), Influenza viruses (IFV) and HBoV (human bocavirus) .............119

Page 11: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xi

Figure 7.3-2: RV detection rates stratified by species. RV-C was the predominant species

detected in children hospitalised with respiratory wheeze ............................................120

Figure 7.3-3- A comparison of virus detection rates between patients with classified with

asthma compared to those not classified with asthma. .................................................121

Figure 7.3-4: Area under the curve analysis of RV-C load to predict hospitalisation. RV-C

load was poor predictor of hospitalisation with an AUC of 0.5 (95%CI, 0.24-0.74) ...........122

Figure 7.3-5 Levels of IL-4 (a) and IL-13 (b) in nasal secretions of non-respiratory disease

controls, RV-C infected patients with asthma and without asthma. IL-4 and IL-13 were both

significantly elevated in the asthmatic group but IL-13 levels did not differ significantly in

the non asthmatic group compared to controls.* p<0.05. ..............................................125

Figure 7.3-6 Levels of IL-12 (a) and IL-2 (b) in nasal secretions of non-respiratory disease

controls, RV-C infected patients with asthma and without asthma. Levels of IL-2 and IL-12

did not significantly differ when each group (asthmatics and non-asthmatics patients) were

individually compared to controls. ...............................................................................126

Figure 7.3-7 Levels of Interferon (IFN)-γ (a), IFN-λ (b), IFN-α (c), in nasal secretions of

controls, hospitalised RV-C infected patients with asthma and without asthma. IFN-γ was

significantly attenuated in both patient groups compared to controls. The l evels of IFN- λ,

and IFN-α were not significantly different in either patient group compared to controls.

*p<0.05. .....................................................................................................................127

Figure 7.3-8 Levels of IL-1β (a) and IL-6 (b) in nasal secretions of controls, hospitalised RV-C

infected patients with asthma and without asthma. IL-1β was significantly elevated in the

asthma group but not in the non-asthma group compared to healthy controls.*p<0.05. .128

Figure 7.3-9 Levels of IL-8 (a) and IP-10 (b) in nasal secretions of non-respiratory disease

controls, RV-C infected patients with asthma and without asthma. IL-8 and IP-10 were both

significanlty elevated both patient groups comapred to controls.* p<0.05, **p<0.001 ...129

Figure 7.3-10 Levels of IL-10 (a) and IL-17 (b) in nasal secretions of non-respiratory disease

controls, RV-C infected patients with asthma and without asthma. IL-10 and IL-17 were

only significantly elevated in the asthma patient group compared to controls.* p<0.05 ..130

Page 12: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xii

List of Tables Table 1-1: Seasonal and clinical profiles of the commonly detected viruses associated with

acute respiratory tract infection....................................................................................... i

Table 4.3-1 A comparison of RNA transcript concentration and Cq values for the different

RV-C assays.................................................................................................................. 68

Table 4.3-2 the performance of the individual PCR assays for the detection of matched RV -

C RNA transcript........................................................................................................... 69

Table 4.3-3 Intra and Inter assay variability of the four RV-C qRT-PCR assays (Assay 1-4) .. 70

Table 4.3-4 Variation in calculated copy number yield (%) of transcripts 1-4 compared to

the number of probe mismatches ................................................................................. 71

Table 5.3-1 Demographic and clinical details of study participants .................................. 83

Table 6.3-1 Baseline characteristics of children with acute asthma exacerbation (AAE) and

controls ....................................................................................................................... 99

Table 6.3-2: A statistical summary of the risk of being diagnosed with acute asthma

exacerbation follwoing respiratory virus detection........................................................101

Table 7.3-1 Summary of clinical and demographic data of hospitalised children with RV -C

respiratory wheeze .....................................................................................................123

Table 7.3-2 nasal cytokine levels of healthy non-respiratory disease controls, RV-C

infected patients with asthma and without asthma. ....................................................130

Table 7.3-3: An illustration of the relationship between RV-C load and inflammatory

mediator production in the nasal secretions of children with asthma .............................133

Table 7.3-4: An illustration of the relationship between RV-C load and inflammatory

mediator production in the nasal secretions of children without asthma ........................133

Table 7.3-5: Association between cytokine production and hospitalisation of children

hospitalised following RV-C infection............................................................................134

Table 0-1 The performance of the individual PCR assays for the detection of matched RV -C

RNA transcript ............................................................................................................176

Table 0-2 A comparison of RNA transcript concentration and Cq values for the different RV-

C assays ......................................................................................................................177

Table 0-3 Intra and Inter assay variability of the four RV-C qRT-PCR assays (Assay 1-4) ....178

Table 0-4 The RV-C load determinations for patients enrolled in the PREVIEW study. Clinical

samples were tested in triplicate and mean viral load calculated. ..................................179

Page 13: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xiii

Acknowledgments

First and foremost, I want to thank Prof. David Smith, A/Prof. Allison Imrie, Dr. Glenys

Chidlow and Dr. Gerald Harnett. It has been an honour to be your Ph.D. student. You all

have taught me, both consciously and unconsciously, how good research is conducted. I

appreciate all your contributions of time, ideas, and funding to make my Ph.D. experience

productive and stimulating. The joy and enthusiasm you all had for this research was

contagious and motivational for me, even during tough times. I am also thankful for the

excellent example you have provided as successful leaders in your respective fields. Special

thanks to Robyn Thompson for her countless efforts to ensure I had access to everything

(conferences, meetings and training courses) and everyone I needed to accomplish my

objectives.

I would like to thank all the members of the PathWest Molecular Diagnostics Laboratory

for accommodating me into your busy laboratory. You have been a source of friendships as

well as good advice. Many thanks to our collaborators at the Telethon Kids Institute,

especially Professor Peter Le Souef, Dr. Ingrid Laing and Dr Kim Khoo. I would also like to

thank Dr. Abha Chopra at the Institute of Immunology and Infectious diseases for providing

access to their digital PCR instrument.

Last but not the least, I want to thank my partner Emma, my family and my closest friend’s

you guys have been amazing during this time. Mum you have been my biggest supporter

from the get go. Dad thank you for your wise words, my brothers Kapembwa and

Kamando, you are the best brothers and friends one could ever ask for. Jarrad, thank you

for your friendship during this phase it was one where we learnt a lot from each other and

I will forever cherish the times spent at coffee. My dearest Emma, thank you for being so

patient and supportive during this time.

Page 14: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xiv

I gratefully acknowledge the funding sources that made my Ph.D. work possible. I was

funded by University of Western Australia Postgraduate award and University of Western

Australia Top-up Scholarship. Material, consumables and software were made available by

PathWest Laboratory Medicine WA. This research was supported by an Australian

Government Research Training Program (RTP) Scholarship.

Page 15: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xv

AUTHORSHIP DECLARATION: SOLE AUTHOR PUBLICATIONS

This thesis contains the following sole-authored work that has been published and/or prepared for publication.

Deta ils of the work: Comparison of Droplet Digital RT-PCR to qPCR for the quantitative detection of Respiratory

Syncytia l Virus Chisha T. Sikazwe, Angela Fonceca, Avram Levy, Glenys R. Chidlow, Al lison Imrie, Mark Everard,

David W. Smith. RSV16: 10th International Respiratory syncytial vi rus symposium, Sept 2016, Patagonia,

Argentina. Location in thesis: Chapter Three

Deta ils of the work: Reliable quantification of rhinovirus species C using real-time PCR. Sikazwe CT, Chidlow GR,

Imrie A, Smith DW. J Vi rol Methods. 2016 May 20;235:65-72. doi :10.1016/j.jvi romet.2016.05.014. [Epub ahead

of print] PMID: 27216896. Location in thesis: Chapter Four

Deta ils of the work: The Quantitative Detection of Respiratory Syncytial Virus in Hospitalized Young South

African Children Chisha T. Sikazwe, Al icia A. Annamalay, Glenys R. Chidlow, Salome Abbott, Siew -Kim Khoo,

Joelene Bizzintino, Robin Green, Allison Imrie, Peter LeSouëf, David W. Smith. Submitted to Influenza and other

Respiratory vi ruses. Location in thesis: Chapter Five

Deta ils of the work: Sikazwe CT, Chidlow GR, Imrie A, Smith DW. Determinants of acute asthma exacerbation

severity following RV-C infection. Location in thesis: Chapter Six

Deta ils of the work: Sikazwe CT, Chidlow GR, Imrie A, Smith DW. Cytokine profiles in nasal secretions of

patients hospitalised with Rhinovirus Species C associated respiratory wheeze, Cytokine. (In preparation).

Location in thesis: Chapter Seven

XChisha Teza Sikazwe

Date:

XAllison Imrie

Co-ordinating Supervisor

Date:

25/06/2018

25/06/2018

Page 16: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xvi

Peer reviewed papers and Conference presentations

1 Rachael Lappan; Kara Imbrogno; Chisha Sikazwe; Denise Anderson; Danny Mok;

Harvey Coates; Shyan Vijayasekaran; Paul Bumbak; Christopher Blyth; Sarra Jamieson;

Christopher Peacock A microbiome case-control study on recurrent acute otitis media

identified potentially protective bacterial genera. Microbiome, (In submission)

2 Bjerregaard, A., Laing, I.A., Backer, V., Fally, M., Khoo, S.-K., Chidlow, G., Sikazwe, C.,

Smith, D.W., Le Souëf, P. and Porsbjerg, C. (2016) Clinical characteristics of eosinophilic

asthma exacerbations. Respirology, 22: 295–300. doi: 10.1111/resp.12905.

3 Bjerregaard, A., Laing, I.A., Backer, V., Fally, M., Khoo, S.-K., Chidlow, G., Sikazwe, C.,

Smith, D.W., Le Souëf, P. and Porsbjerg, C. (2017)-High fractional exhaled nitric oxide

and sputum eosinophils are associated with an increased risk of future virus-induced

exacerbations – a prospective cohort study. Clinical and Experimental Allergy

4 The Quantitative Detection of Respiratory Syncytial Virus in Hospitalized Young South

African Children Chisha T. Sikazwe, Alicia A. Annamalay, Glenys R. Chidlow, Salome

Abbott, Siew-Kim Khoo, Joelene Bizzintino, Robin Green, Allison Imrie, Peter LeSouëf,

David W. Smith. Submitted to Influenza and other Respiratory viruses (in submission)

5 Reliable quantification of rhinovirus species C using real -time PCR. Sikazwe CT,

Chidlow GR, Imrie A, Smith DW. J Virol Methods. 2016 May 20;235:65-72.

doi:10.1016/j.jviromet.2016.05.014. [Epub ahead of print] PMID: 27216896

6 Respiratory viruses in young South African children with acute lower respiratory

infections and interactions with HIV. Annamalay AA, Abbott S, Sikazwe CT, Khoo SK,

Bizzintino J, Zhang G, Laing I, Chidlow GR, Smith DW, Gern J, Goldblatt J, Lehmann D,

Green RJ, Le Souëf PN. J Clin Virol. 2016 Aug;81:58-63. doi: 10.1016/j.jcv.2016.06.002.

Epub 2016 Jun 4. PMID: 27317881

7 Comparison of Droplet Digital RT-PCR to qPCR for the quantitative detection of

Respiratory Syncytial Virus Chisha T. Sikazwe, Angela Fonceca, Avram Levy, Glenys R.

Chidlow, Allison Imrie, Mark Everard, David W. Smith. RSV16: 10th International

Respiratory syncytial virus symposium, Sept 2016, Patagonia, Argentina.

8 Reliable Quantification of Rhinovirus Species C using a Taqman Real -time PCR Based

Approach Chisha T Sikazwe · Glenys R. Chidlow · Allison Imrie · David W. Smith. 1st

International Meeting for Respiratory Pathogens, Sep 2015 Singapore.

9 The Relationship between RSV Load and Clinical Disease in South African Children

Chisha T Sikazwe, Glenys R. Chidlow, Allison Imrie, David W Smith. 9th International

Symposium on Respiratory syncytial virus Nov 2014, Cape Town, South Africa

Page 17: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xvii

List of Abbreviations

Abbreviation Definition

µ Micro

µL Microlitre

µM Micromolar

AAE Acute Asthma Exacerbation

ALF Australian Lung Foundation

ARTI Acute Respiratory Tract Infection

BAL Bronchoalveolar Lavage

BHQ Black Hole Quencher

BLAST Basic Local Alignment Search Tool

bp Base Pair

cDNA Complementary DNA

CFT Complement Fixation Test

CMV Cytomegalovirus

COPD Chronic Obstructive Pulmonary Disease

CPE Cytopathic Effect

Cq Cycle Quantification Value

Ct Cycle Threshold

Page 18: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xviii

CXCL- Chemokine Ligand

ddPCR Digital Droplet PCR

DNA Deoxyribonucleic Acid

EIA Enzyme Immunoassay

GINA Global Initiative For Asthma

HAdV Human Adenovirus

HBoV Human Bocavirus

HCoV Human Coronavirus

HMPV Human Metapneumovirus

HPIV Human Parainfluenza virus

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

IF Immunofluorescence

IFAV Influenza A Virus

IFBV Influenza B Virus

IFN- Interferon

IL- Interleukin

LNA Locked Nucleic Acid

LRTI Lower Respiratory Tract Infection

MERS-CoV Middle Eastern Respiratory Syndrome Coronavirus

Page 19: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xix

MGB Minor Groove Binding

mL Milliliter

NA Nasal Aspirate

NAT Nucleic Acid Test

NPA Nasopharyngeal Aspirate

NPS Nasopharyngeal swab

NT Neutralisation Test

NW Nasal Wash

PCR Polymerase Chain Reaction

PMH Princess Margaret Hospital

PWLM PathWest Laboratory Medicine WA

qPCR Quantitative Real Time PCR

RNA Ribonucleic Acid

RSV Respiratory Syncytial Virus

RT-ddPCR Reverse Transcription Digital Droplet PCR

RT-PCR Reverse Transcription PCR

RT-qPCR Reverse Transcription Quantitative Real Time PCR

RV Rhinovirus

SARS-CoV Severe Acute Respiratory Syndrome Coronavirus

Page 20: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

xx

Th T Helper Cells

TKI Telethon Kids Institute

TLR Toll Like Receptors

TNF- Tumor Necrosis Factor

URTI Upper Respiratory Tract Infection

UTR Untranslated Region

UWA The University Of Western Australia

VL Viral Load

WHO World Health Organisation

α Alpha

β Beta

Page 21: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe
Page 22: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

2

1 Literature review

Page 23: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

1.1 Introduction

Acute respiratory tract infections (ARTIs) contribute substantially to the global burden of

illness from communicable pathogens. ARTIs are a leading cause of morbidity and mortality

accounting for approximately four million deaths per year globally (Bryce et al., 2005).

Children under the age of five years, adults over the age of sixty-five, individuals with an

underlying chronic condition and immunocompromised individuals are population groups

in whom poor outcomes occur following infection (Falsey et al., 2003; Graham and Gibb,

2002; Nair et al., 2011a; Nair et al., 2010; Nair et al., 2013). Estimates of the global burden

of acute respiratory illness indicate that there are vast differences between developed and

developing countries. According to recent reports, ARTIs are the leading cause of childhood

death in developing countries (Nair et al., 2011b). Pneumonia alone accounts for 1.4

million childhood deaths per year in these regions. Conversely, in developed nations

deaths due to ARTIs represent a negligible percentage of total deaths, accounting for less

than two percent of deaths but are predominantly responsible for absenteeism and

enormous financial costs to the healthcare system (Denny Jr., 2001). Reports from the US

estimate that direct financial costs of ARTIs to the healthcare system approach $40 billion

annually. Similarly, in Europe the expenditure on patients with ARTI is over €15 billion. The

direct and indirect cost of ARTIs to the Australian health care system is estimated to be up

to AUD 600 million each year (The Australian Lung Foundation, 2007).

All groups of microbes are capable of establishing an infection in the respiratory tract.

However, viruses predominate as aetiologic agents of ARTIs and can contribute to

respiratory disease either directly through frank infection or indirectly by exacerbating

pre-existing illness (Bafadhel et al., 2011; Bandi et al., 2003; Busse, Lemanske, and Gern,

2010; Hosseini et al., 2015) and increasing the risk of secondary bacterial infection

(Bellinghausen et al., 2016; Ewijk et al., 2007). Influenza virus (IFV), respiratory syncytial

Page 24: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

4

virus (RSV), rhinovirus (RV), human metapneumovirus (hMPV), human parainfluenza virus

(hPIV) and human adenovirus (hAdV) are the most prevalent viruses in hospitalised

patients and are all linked to ALRI (Lukšić et al., 2013). The distribution of these viruses is

influenced by season, geographic region and age group (Lukšić et al., 2013). They are at

least 6 families and more than 150 viruses that are associated with ARTI. The clinical and

seasonal profiles of the most common types are listed in Table 1.

Page 25: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

Table 1-1: Seasonal and clinical profiles of the commonly detected viruses associated with acute respiratory tract infection

Virus

Incubation period (days)

Seasonality Clinical manifestations Laboratory diagnostic method

Respiratory syncytial virus (RSV) 2-8 Winter bronchiolitis, pneumonia Virus isolation in cell culture, RT-PCR, IF, EIA,

Human metapneumovirus (HMPV) 2-8 Winter to spring bronchiolitis, pneumonia RT-PCR, IF

Rhinovirus (RV) 2 All year round coryza, COPD and asthma exacerbation, bronchiolitis, pneumonia

RT-PCR, NT and Virus isolation in cell culture (for some types)

Coronavirus (HCoV) 2 All year round coryza, pneumonia (rarely in non-SARS or MERS infection)

RT-PCR,ELISA,HA,IF

Influenza virus (IFV) 2-8 Winter Virus isolation in cell culture, RT-PCR, CFT, HI,IF, EIA,NT

Human parainfluenza viruses (HPIV) 2-8 Autumn to winter coryza, croup, bronchitis, bronchiolitis, and pneumonia

Virus isolation in cell culture, RT-PCR, CFT, HI,IF, EIA,NT

Adenovirus (AdV) 5-7 All year round pneumonia, bronchitis, pharyngitis, tonsillitis, and pharyngoconjunctival

fever

RT-PCR, HI, IF,EIA and NT

Human bocavirus (HBoV) 5-7 All year round Coryza, bronchiolitis, pneumonia

RT-PCR

Abbreviations: RT-PCR; reverse transcription PCR, IF; immunofluorescence, EIA; enzyme immunoassay, HA; haemagglutination, NT; neutralisation test, HI; haemagglutination inhibition, CFT; complement fixation

test.

Page 26: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

1.1.1 Respiratory syncytial virus

RSV is a member of the paramyxoviridae family and has a single strand negative sense

enveloped RNA genome. RSV genome encodes two non-structural proteins and nine

structural proteins. The attachment (G) and fusion (F) glycoproteins are the

immunodominant antigens of RSV and are essential in infectivity and antigenicity (Fodha et

al., 2007). RSV is considered to be serologically monotypic, but consists of two genetic

subgroups, RSV-A and RSV-B (Papadopoulos et al., 2004). RSV A is more prevalent than RSV

B but there is no consensus on which of the subtypes is more pathogenic.

RSV accounts for 40-50% of all viral infections requiring hospitalisation in young infants

(Nair et al., 2011b). Virtually all children are exposed to RSV before the age of 3 years

(Henderson et al., 2005). Acute bronchiolitis is the typical syndrome associated with RSV

infection in young infants and is characterised by a predominantly neutrophilic pattern of

inflammation and mucus in the airways, which often results in some degree of pulmonary

obstruction. Young infants are predisposed to severe acute bronchiolitis because of the

small diameter of their airways. Congenital heart disease, prematurity, bronchopulmonary

dysplasia or cystic fibrosis are all risk factors for severe infection and possibly death

(PREVENT, 1997).

1.1.2 Rhinoviruses

Rhinoviruses (RVs) are non-enveloped positive sense single stranded RNA viruses

belonging to the family Picornaviridae. They are genetically and antigenically diverse

consisting of three species (A, B and C). Historically, extensive cross neutralisation test

were utilised for the classification of RV-A and B isolates into 100 serotypes but these

isolates (types) are now assigned solely on genomic sequence (McIntyre, Knowles, and

Page 27: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

7

Simmonds, 2013). In 2006, utilisation of molecular techniques led to the discovery of RV-C,

which had been entirely unrecognised using traditional rhinovirus detection techniques

(Arden et al., 2006). Cell lines that were conventionally used to isolate rhinovirus are not

permissive to RV-C infection and as a result preclude serotype assignment. Sequence

typing has revealed that RV-C is genetically more diverse than the other two species and

currently consists of 65 distinct genotypes (McIntyre et al., 2013).

Historically, RV-A and RV-B serotypes were stratified into two groups (major and minor) on

the basis of cellular receptor utilisation (intracellular adhesion molecule [major] or low-

density lipoprotein receptor [minor]) whereas the RV-C receptor remained unidentified

until recently. A recent report has shown that the human cadherin-related family member

3 (CDHR3), a member of the cadherin family of transmembrane proteins, facilitates RV -C

attachment and replication (Bochkov et al., 2015). This receptor is highly expressed in the

lower respiratory tract but its biological function remains unknown. A single nucleotide

polymorphism in CDHR3 gene is associated with RV-C wheezing illness in infancy and has

been shown to be risk factor for asthma inception (Bonnelykke et al., 2014).

The spectrum of disease exhibited by RVs range from asymptomatic infections, mild upper

respiratory infections (common cold) to severe or fatal lower respiratory tract infections

(pneumonia and bronchiolitis). RV infection is also associated with the exacerbation of

chronic respiratory conditions such as asthma, cystic fibrosis and other chronic obstructive

pulmonary disorders (Blomqvist et al., 2002; Çalışkan et al., 2013; Camargo et al., 2012;

Choi et al., 2015). In addition, RV induced wheeze in early childhood is associated with an

increased risk of developing asthma later on in life (Jackson et al., 2008a). Recent studies

have also revealed that RV-C species are more closely associated with asthma exacerbation

and illness requiring hospitalization than the other RV species, suggesting that RV-C may be

more pathogenic than RV-A and RV-B (Bizzintino et al., 2011a; Cox et al., 2013). Host and

Page 28: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

8

viral factors associated with RV-C pathogenesis and illness severity are yet to be

completely elucidated.

1.1.3 Human Metapneumovirus

HMPV is classified as a negative sense single stranded RNA virus belonging to the

Paramyxoviridae family. Sequence analysis of the fusion (F) and attachment (G) genes has

identified two genotypes namely A and B. Both genotypes may co-circulate but only one

genotype is dominant during an epidemic (Tsukagoshi et al., 2013a; van den Hoogen et al.,

2002).

HMPV is associated with hospitalisation in young children, the elderly and individuals w ith

an underlying chronic condition. It is distributed worldwide and mimics the seasonal

distribution of influenza and RSV in that infection rates tend to peak in winter and early

spring (Kahn, 2003). Much like other respiratory viruses the spectrum of clinical disease

ranges from a mild upper respiratory tract infection to severe pneumonia. Observational

studies report that elderly people infected with hMPV display more severe clinical

symptoms compared to younger patients and those elderly infected with RSV or Influenza

virus (Falsey et al., 2003; Widmer et al., 2012).

1.1.4 Influenza virus

The influenza viruses are a major causative agent of ARTI in both children and adults,

Infection can cause mild to severe illness, and can occasionally lead to death (Pretorius et

al., 2016). Influenza accounts for a substantial burden of ARTI and current estimates

suggest that it is responsible for approximately five million severe cases and between

291,000-646,000 deaths per annum (Iuliano et al., 2018). Three Influenza types (type A,

type B and type C) are known to infect humans. Only the influenza A/H1N1, A/H3N2 and

influenza B viruses are considered as being significant contributors to seasonal influenza.

Page 29: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

9

Influenza C virus infection is seasonal but has limited genetic diversity and is not thought to

cause epidemics (Nicholson, Wood, and Zambon, 2003).

Genomes of influenza A and B viruses are composed of eight single-stranded negative

sense RNA segments whereas the influenza C virus is composed of seven segments of

single stranded negative sense (Nicholson et al., 2003).

The genomes of influenza viruses specifically the Influenza A virus (IFAV) regularly undergo

changes under immune pressure. The genes encoding surface glycoproteins

haemagglutinin and neuraminidase may undergo subtle genetic changes (antigenic drift) or

abrupt major changes (antigenic shift).

Influenza therapeutics comes in the form of vaccines and anti -viral compounds. Vaccines

are recommended for annual boosting especially in high risk groups (Fiore, Bridges, and

Cox, 2009; Matsushita et al., 2017). Most Influenza antiviral compounds inhibit viral

replication and release, and can be utilised for treatment and prophylaxis (Dobson et al.,

2015; Jefferson et al., 2014b).However, adverse events reported from use of these

compounds suggested that they should be administered judiciously(Jefferson et al.,

2014a).

1.1.5 Parainfluenza virus

Human parainfluenza viruses (HPIV), of which they are four types (HPIV1-4) are negative

sense RNA viruses of the Paramyxoviridae family. HPIVs typically cause mild, self-limited

upper respiratory tract infections in adults, but can result in severe, life -threatening LRTIs

in immunocompromised patients (Guzmán-Suarez et al., 2012). HPIVs 1-3 are a common

cause of croup syndrome and are among the most commonly detected viruses in

hospitalised young infants after RSV. HPIV-4 is known to cause mild upper respiratory tract

Page 30: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

10

infections (Tsukagoshi et al., 2013b). However, due to a lack of epidemiological data on

HPIV-4 the extent of its pathogenicity is largely unknown.

1.1.6 Adenovirus

Human Adenoviruses (HAdV) are a major cause of viral disease in both children and adults

(Lynch, Fishbein, and Echavarria, 2011). HAdV have linear double-stranded DNA genomes

and are classified into seven species (HAdV A to G) and a total of 55 types based on a

combination of serological characteristics and phylogenetic analysis (Robinson et al., 2011).

Certain species are known to cause respiratory illnesses including, acute febrile pharyngitis

(HAdV-B and -C; types 1,3,5,7), pharyngoconjunctival fever (HAdV-B; types 3,7,14), acute

respiratory tract infections (HAdV-B and -E; types 3,4,7,14,21), pneumonia (HAdV-B, -C,

and -E; serotypes 1-4 and 7), pertussis-like syndrome (HAdV-C; type 5) (Demian et al.,

2014). PCR is the method of choice in detecting adenoviruses.

1.1.7 Human Coronaviruses

Human coronaviruses (HCoVs) are enveloped viruses with a single-strand positive sense

RNA genome. Coronaviruses possess the largest known genomes among any of the RNA

viruses (Lau et al., 2013). Six species of HCoVs are known to infect humans; they are

classified into four genera based on proteomic analysis (Lau et al., 2013). HCoV-229E and

HCoV-NL63 belong to the Alphacoronavirus genus, HCoV-OC43 and HCoV-HKU1 belong to

lineage A Betacoronavirus, while severe acute respiratory syndrome associated

coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV)

belong to lineages B and C Betacoronavirus, respectively (Annan et al., 2013). HCoV-OC43,

HCoV-229E, hCoV-HKU1 and hCoV-NL63 are commonly associated with self-limiting mild

upper respiratory tract infections but on some occasions may be causative agents of

severe LRTIs especially in young infants, the elderly, and individuals with compromised

Page 31: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

11

immunity (Lu et al., 2012). HCoVs are detected frequently with other respiratory viruses;

the significance of this phenomenon in relation to illness severity is unclear.

In the last decade SARS-CoV and MERS-CoV have been responsible for epidemics around

the world (Cheng et al., 2007). SARS-CoV was identified as the causative agent of the SARS

global epidemic from 2002 to 2003. MERS-CoV was initially reported in Saudi Arabia in

2012 (Zaki et al., 2012). Since then it has caused illness in people in countries with links to

the Arabian Peninsula (de Groot et al., 2013). Both MERS-CoV and SARS-CoV are emerging

zoonotic pathogens that crossed the species barrier with case fatality rates of 35% and

10% respectively (To et al., 2013). Bats appear to be the natural reservoir of both MERS-

CoV and SARS-CoV, transmission to humans occurs via dromedary camels and civets (live

market animals) respectively (Dijkman et al., 2013; Hu et al., 2015; Pfefferle et al., 2009;

Yusof et al., 2015). Nucleic acid tests are the method of choice for the laboratory diagnosis

of HCoVs (Gaunt et al., 2010) and MERS-CoV(Feikin et al., 2015).

1.2 Laboratory Diagnosis of Respiratory Infections

Laboratory diagnostic methods are the cornerstone in accurately attributing pathogen to

clinical presentation. Early detection of the aetiologic agent is pertinent for providing

optimal clinical management, which may include patient isolation, determining

appropriate therapy and/or cessation of inappropriate therapeutic interventions. In

addition, diagnostics are also essential for outbreak detection and response, and public

health surveillance. Characteristics of the ideal diagnostic test include being accurate

(between tests and laboratories), high throughput, cost-effective, suitable for a wide

spectrum of clinical samples, excellent sensitivity and specificity. Current diagnostic tests

fulfil some, but not all of these ideal characteristics.

Page 32: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

12

They are several methods that are available for the identification of the aetiologic agent

from patients with an ARTI. The traditional laboratory diagnostic methods such as virus

isolation in cell culture and serology tests perform well but have some inherent limitations

in aspects of sensitivity and/or specificity. Molecular based diagnostic techniques such as

PCR have led to clinicians and scientists re-evaluating the role of certain viruses in disease

outcome. PCR based tests are a relatively new tool in the laboratory diagnostics. These

tests allow for rapid screening of clinical samples for multiple aetiologies with the added

benefit of excellent sensitivity and specificity. Further, these diagnostic approaches

facilitate further understanding of the viral kinetics in respiratory infections as well as the

ability to identify respiratory viruses that have been entirely missed by traditional

diagnostic approaches.

1.2.1 Upper and Lower airway samples

A variety of specimens have been used for directly sampling the respiratory tract including

nasal swabs (NS), nasopharyngeal swabs (NPS), nasal washings (NW), nasal aspirates (NA),

nasopharyngeal aspirates (NPA), sputum, bronchoalveolar lavage (BAL) or biopsies. The

technique utilised to collect a sample is based on clinical presentation and the method to

be utilised to identify the pathogen.

NWs are conventionally used on children and are not well tolerated in adults. NW can be

unpleasant to the patient, in addition, NW can be technically demanding, as the technique

requires the use of a solution such as saline. A NS is commonly taken from the mid-inferior

portion of the inferior turbinates for optimal virus recovery. Though this collection

technique maximises virus recovery, it causes some discomfort to the patient.

Nonetheless, both sampling techniques are suitable for detection of virus by cell culture.

NPA is especially useful for virus culture, antigen detection, and polymerase chain reaction

(PCR) based assays and is collected using a mucus trap, attached to a disposable suction

Page 33: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

13

catheter. This technique requires a skilled operator, so it may be unsuitable for widespread

clinical practice. Studies that have compared NS and NPA sampling techniques for

detection by PCR report that the overall sensitivity of nasal swabs was inferior to that of

NPAs, but the authors noted that obtaining a NPA was invasive, uncomfortable and

significantly more distressful than a nasal swab (Heikkinen et al., 2002; Lambert et al.,

2008). With the recent development of flocked nasal swabs and advanced molecular

techniques, the sensitivities have increased to a level comparable to NPAs with the added

advantage of the flocked nasal swab being less painful and more convenient than NPAs, as

no supplementary tools are required (Ortiz de la Tabla et al., 2010). Sputum samples are

more representative of lower respiratory tract and are collected non-invasively. Despite

the aforementioned advantages of sputum collection, practical reasons preclude the

effective collection of sputum from infants and children (Abdullahi et al., 2008; Grant et al.,

2012). Children have difficulty producing sufficient sputum for laboratory evaluation

compared to adults and are more inclined to swallow the specimen than expectorate it

(Grant et al., 2012). Further the viscosity of sputum and the likely presence of

contaminating bacteria as well as the toxic effects sputum can have on cell culture

preclude its widespread use for viral diagnostic purposes (Falsey, Formica, and Walsh,

2012). BAL and lung biopsy specimen are collected directly from lower respiratory tract

sites. Previous studies on MERS-CoV have shown that quantification cycle (Cq) values were

lower in lower respiratory tract samples compared to upper respiratory tract samples and

more accurately reflect the apparent viral kinetics in the lower respiratory tract. However,

these specimen types are usually collected from the severely ill (Falsey et al., 2012) and too

few of these specimens would be available to conduct proper population level studies.

1.2.2 Virus isolation in cell culture

Page 34: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

14

Historically, virus isolation by cell culture was the diagnostic method used to screen

respiratory samples for the presence of viruses. Virus isolation in cell culture is regarded as

the "gold standard" to which all other detection methods have been compared since the

isolation of virus in cell culture indicates the presence of an infectious, viable, and

replication competent virus, an occurrence which is unachievable using other detection

techniques (Leland and Ginocchio, 2007). This method is labour intensive, and often

requires prolonged incubation periods (3-14 days) to provide results, limiting its use in the

acute management of patients. A broad range of respiratory viruses can be grown by using

at least 4 cell lines, which includes human epithelial cell lines (HEp-2, A-549, HeLa),

human fibroblast cell lines (HLF, HELF, MCR-5, WI-38) and primary monkey kidney cells

(Denny Jr., 2001). For most respiratory virus applications, the presence of virus is

commonly detected by a characteristic cytopathic effect (CPE) under light microscopy. The

combination of conventional cell culture and commercial viral -antigen detection systems

can be used to accelerate diagnostic turn-around times, since viral antigens are detected in

the cell monolayer by immunofluorescence, usually before a distinct CPE is observable

(Terletskaia-Ladwig et al., 2008). In recent times, more rapid and powerful tools to detect

the presence of virus in clinical samples collected from patients following ARTI have

gradually superseded virus culture techniques.

1.2.3 Direct detection of respiratory viruses by immunofluorescence

Laboratory diagnosis can be performed by direct detection of virus in clinical specimen.

Direct antigen tests assist in the diagnosis of respiratory infection by providing evidence of

the antigen in respiratory specimen. These tests are instrumental in informing pertinent

therapeutic decisions in a short time frame (Gomez et al., 2016). Direct and indirect

immunofluorescence are the two sub-forms of the immunofluorescence test. The direct

test is composed of a virus specific monoclonal antibody that is conjugated directly to

Page 35: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

15

fluorescent dye. If the virus is present in the clinical sample the monoclonal antibody

attaches to the targeted viral antigen and the conjugated fluorescent dye can be visualised

under a fluorescent microscope. In the indirect method, two antibodies are used, a specific

primary monoclonal antibody that attaches to the viral antigen and a secondary antibody

labelled with a fluorescent dye to bind to the primary monoclonal antibody. The indirect

method is more sensitive than the direct method because of the signal amplification from

multiple secondary antibodies binding to a single primary antibody.

Antigen detection methods such as the enzyme immunoassay (EIA) are performed by

adding patient specimen onto a surface that is pre-coated with an antibody that captures

the virus specific antigen if present in the specimen. EIA is a highly sensitive assay and

utilises an enzyme system to produce a colour reaction that can be quantified.

1.2.4 Nucleic Acid Tests

Diagnostic laboratories detect respiratory viruses in clinical samples by using molecular

techniques to detect virus genome. PCR based methods are the most favourable and the

most widely used approach for rapid and accurate respiratory virus detection from clinical

specimens. The advent of nucleic acid based tests has led to the identification of viruses

that were entirely missed using conventional diagnostic techniques. Most respiratory

viruses have their genetic information primarily stored in the form of RNA, with the

notable exception of HAdV and HBoV. Amplification of an RNA target requires a reverse

transcription (RT) step in order to convert RNA to cDNA followed by conventional PCR (RT-

PCR). DNA targets do not require an RT step. The repetition of three successive reactions

is the basis of PCR:

1. Denaturation of double stranded DNA (dsDNA) into si ngle stranded DNA (ssDNA)

between 94 and 95⁰C

Page 36: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

16

2. Annealing of 2 synthetic oligonucleotides (primers) to each ssDNA at each termini at a

variable temperature ranging from 37-72⁰C

3. Synthesis of new DNA strands; the PCR method util izes a thermostable DNA polymerase

to add nucleotides at the 3' of end of the primers at 68 - 72⁰C.

These three steps entail a single PCR cycle, since each synthesized DNA strand becomes a

template for amplification there is an exponential increase in amplicon at the end of each

cycle. The length of the PCR product is equivalent to that of the two primers plus the

distance separating the two primers. Several variations of PCR are applicable to the

diagnosis of respiratory viral infection including nested PCR, real -time PCR (qPCR) and

multiplex qPCR.

1.2.4.1 Real time PCR (qPCR)

Utilisation of modern molecular techniques has enabled detection of viral nucleic acid

during the exponential phase of the reaction rather than waiting for the endpoint of the

reaction to register detection. The fundamental principle of qPCR is based on the detection

and quantification of fluorescent reporter molecules whose signal can be registered in

each cycle of the PCR. The cycle number when the fluorescence becomes detectable is

referred to as the cycle threshold/quantification value (Ct/Cq), and is proportional to the

logarithm of the initial amount in the clinical sample. Unlike conventional PCR, qPCR does

not require any post PCR amplification manipulations thereby minimizing any issues

related to contamination. However, similar to conventional PCR is the addition of a reverse

transcription step when screening clinical samples for respiratory viruses with an RNA

genome.

A DNA binding fluorescent dye, such as SYBR green represents the simplest method to

detect amplified product using qPCR, given that this dye binds to any double stranded DNA

Page 37: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

17

in the reaction. A shortfall of SYBR green chemistry is its lack of sequence specific DNA

binding activity. So this type of chemistry will also bind to non-specific products that the

qPCR reaction may generate and may lead to false positive results. Sequence specific

hydrolysis probes are alternative detection chemistry and mitigate the problem of false

positive results because fluorescence signal is only detected when the probe binds to the

target area. Sequence specific hydrolysis probes are the most widely used and published

detection chemistry available for qPCR. These probes are beneficial because they are

designed to increase both the sensitivity and specificity of the assay. qPCR provides

excellent sensitivity and a wide dynamic range and for this reason can be utilised to

measure viral load in clinical samples.

Viral load determination may increase current understanding of host viral interactions

(Quinn, 2011) and may help predict disease progression (Jartti et al., 2013). Viral load

determination from clinical samples is performed by interpolating the cycle quantification

value (Cq) into a standard curve. The standard curve is constructed by standards of target

nucleic acid that encompass a wide range (5-9 logs) of known concentrations. It is

important to note that there is inherent variability in construction of a standard curve

between batches. As such it is imperative that the reference standards used are scrutinised

thoroughly for precision and reproducibility during the validation process , in order to

understand the limitations of the assays and to provide valid standards for RNA

quantification.

Specimen collection is an important source of variability that may obscure real changes

and consequently unreliable quantification results. The reliability of any PCR-based

quantification experiment can be improved by including an invariant endogenous control

(mRNA that is stably expressed and is not affected by experimental conditions) in the

experiment to correct for sample to sample variation that may arise.

Page 38: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

18

1.2.4.2 Multiplex PCR

Multiplex PCR refers to the use of multiple sequence specific primers and probes to detect

multiple targets in a single reaction tube. This type of assay is efficient and economical but

requires substantial assay optimization to ensure optimal amplification of the different

targets if present. The availability of multiplex PCR makes the detection of co-infections

feasible (Franz et al., 2010). Multiplex PCR is an important tool in the diagnostic laboratory

when screening for potential etiologic contributions to disease.

1.2.4.3 Droplet digital PCR

Droplet digital PCR (ddPCR) is a relatively new approach that improves on qPCR by making

external standards unnecessary in viral quantification. In a similar approach to qPCR,

ddPCR involves the detection of template sequence with either a SYBR green or hydrolysis

probe reaction chemistry but quantification is conducted differently. It involves the

generation of a large library of emulsion based droplets (~20,000), also termed partitions

(Markey, Mohr, and Day, 2010). These partitions are generated from sample-reagent

mixtures and are distributed in such a way that there may be either one or zero template

molecules in the each partition (Mojtahedi, Fouquier d'Herouel, and Huang, 2014). This is

the fundamental principle underpinning digital PCR. Further, and in contrast to qPCR the

thermal cycling is performed to endpoint (Markey et al., 2010). The total initial number of

template is obtained by tallying partitions in which the template is detected compared to

the number of partitions in which the reaction is unreactive (Markey et al., 2010). A

Poisson correction is applied to the final copy number to compensate for the possibility

that more than one template molecule may be present in some partitions. ddPCR is highly

sensitive, reproducible, provides enhanced accuracy and precision thus lending itself to a

number of favourable applications such as low copy target detection, quantitative

detection of minor genotypes. Moreover, ddPCR does not rely on factors such as

Page 39: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

19

amplification efficiency and arbitrarily assigned threshold values and therefore reduces the

amount of bias in the sample resulting in confident quantification (Hall Sedlak and Jerome,

2014; Huggett et al., 2013).

Recent work from a study comparing ddPCR to qPCR for quantitative detection of

cytomegalovirus (CMV) demonstrated that ddPCR is equivalent for the accurate

quantification of CMV load in clinical samples over a wide dynamic range (Hayden et al.,

2013). Recent work that evaluated ddPCR for influenza vaccine development has

demonstrated a high throughput ddPCR method for very precise and accurate influenza

virus titre quantification (Palatnik de Sousa et al., 2015). The authors also noted several key

issues that are determinants of variability in qPCR were circumvented with the ddPCR

approach.

1.3 Viral kinetics in acute respiratory tract infections

Viral kinetic studies have improved our understanding of the interplay between host and

virus in human immunodeficiency virus (HIV), CMV, hepatitis B and C infections.

Experimental human infection studies on RSV and IFAV have shown the unique ways in

which viral kinetics modulate illness severity and disease course (Bagga et al., 2013;

DeVincenzo et al., 2010; El Saleeby et al., 2011). In addition, viral load studies have shed

light on critical treatment time points used in order to minimise the risk of severe illness.

The recent discovery of novel respiratory pathogens has stimulated investigations into viral

load as a surrogate marker of disease severity in hospitalised patients. For ex ample,

evidence from a recent study conducted in France reports that a high hMPV viral load is an

important predictor of disease severity in young children (Roussy et al., 2014). Indeed,

previous RSV studies indicate that high viral load is associated with symptom severity

(DeVincenzo et al., 2010; El Saleeby et al., 2011; Houben et al., 2010) . Furthermore, a

Page 40: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

20

reduction in RSV and influenza viral loads in patients as a consequence of antiviral therapy

has been found to be associated with improved clinical outcomes (Boivin, Coulombe, and

Wat, 2003; Shadman and Wald, 2011). This paradigm of direct virus damage to host tissue

shaping clinical outcome has been supported further by investigations on SARS-CoV and

MERS-CoV (Feikin et al., 2015; Hung et al., 2004; Min et al., 2016). In RV infection, evidence

in the published literature suggests that viral load is predictive of the severity of clinical

illness, given that individuals with a lower respiratory tract infection harbour higher viral

loads that those with an upper respiratory tract infection (Utokaparch et al., 2011). This

finding is further supported by an Italian study that reported that an abundant RV viral

load (>106copies/mL) in the absence of other viral pathogens is strongly associated with

LRTI (Piralla et al., 2012). However, other studies have not reproduced these findings (Jartti

et al., 2008) and thus the relevance of RV viral load in lower respiratory tract infections is

not yet completely understood.

1.4 Viral respiratory infection and asthma

Asthma is one of the most common chronic respiratory inflammatory conditions and a

substantial contributor to morbidity worldwide (GINA, 2017). It is characterised by airway

inflammation, remodelling, reversible airway blockage, and increased airway smooth

muscle tone (Zhao et al., 2002). It can be difficult to diagnose asthma with certainty in

children aged between 0-5 years because there are no standardised diagnostic criteria for

asthma (GINA, 2017). In addition, respiratory infections predominant in childhood such as

bronchiolitis share many clinical features with exacerbations of asthma, including

wheezing, shortness of breath and respiratory distress (Sigurs et al., 2000). Diagnosis of

asthma in children may involve the individual’s history of recurrent wheeze, family history

of atopy and objective investigations that support the diagnosis. Several stimuli can trigger

Page 41: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

21

exacerbations of asthma symptoms in young children including smoking, viral infections,

air pollution and environmental allergens (Pelaia, Vatrella, and Maselli, 2012; Riccio et al.,

2012; Saraya et al., 2014; Sykes et al., 2014). The advent of molecular methods has shown

that respiratory viruses are present more commonly in acute asthma than previously

acknowledged, and are detected in up to 85% of asthma exacerbations (Saraya et al.,

2014). RV and RSV are the predominant viruses that appear to be involved with these

exacerbations (Saraya et al., 2014; Sigurs et al., 2000; Soto-Quiros et al., 2012). It is still

not yet clear if infection causes long term changes in the airways which subsequently

increase the risk of developing asthma. An alternative hypothesis is that severe RSV and/or

RV disease (bronchiolitis) may be an early marker of a predisposition for childhood asthma

(Miller et al., 2011; Moore, Stokes, and Hartert, 2013; Moore et al., 2007). The children at

high risk of developing asthma are those who wheeze and develop allergic sensitization

before the age of 2 years (Jackson et al., 2008a; Sigurs et al., 2010; Sigurs et al., 2000). Long

term prospective studies have linked severe RSV disease in young infants to subsequent

recurrent wheeze and asthma in susceptible children (Sigurs et al., 2010). However other

studies do not confirm the association between medically attended RSV disease and

subsequent asthma development (Poorisrisak et al., 2010).

RV infection in young infants is an independent risk factor for subsequent wheeze and

childhood asthma (Jackson et al., 2008a). Several reports have shown that RVs are the

most common virus detected in hospitalised young children with exacerbations of asthma,

suggesting an etiologic role (Liu et al., 2016; Luchsinger et al., 2014; Message and Johnston,

2002; Miller et al., 2011). Children classified as asthmatic are more likely to have poor

outcomes following RV infection compared to normal individuals (Corne et al., 2002). RV-C

appears to be the most commonly detected species in hospitalised asthmatic children with

some studies suggesting that it is also the most pathogenic (Bizzintino et al., 2011a; Cox et

al., 2013; Liu et al., 2016). However, determinants of RV-C associated asthma severity are

Page 42: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

22

yet to be clearly defined, and the contribution of RV-C in asthma exacerbation severity is

still not clear.

Page 43: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

23

1.5 Innate immune response to viral respiratory infection

In response to infection, germ line encoded receptors (pattern recognition receptors,

PRRs) that are resident on sentinel cells of the immune system sense conserved pathogen-

associated molecular patterns (PAMPs) on microbes (Gommerman and Ng, 2013; Kim and

Lee, 2014; Malmgaard, 2004; Scagnolari et al., 2009). There are three known PRR families

and they include toll-like receptors (TLRs), retinoic acid inducible gene 1 (RIG-1)-like RNA

helicases (RLHs) and nucleotide-binding oligomerization domain (NOD) like receptors

(NLRs). Signalling downstream of each PRR family results in the activation of important

pathways that regulate the expression of chemokines, pro-inflammatory cytokines, type 1

interferon (type 1 IFN) and antimicrobial peptides (Thompson and Locarnini, 2007). RLHs,

TLR 3, 7, 8 and 9 identify viral nucleic acid and induce innate type 1 IFN responses with or

without the production of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IL-12, IL-18).

Type 1 IFN is known to block viral replication at an early step in replication and to be very

important for the induction of an anti-viral state against common respiratory viruses

(Scagnolari et al., 2009). Type I IFN deficient mice are more prone to delayed viral

clearance and severe disease following viral challenge. Further, impaired IFN response to

viral infection has been postulated as a pathogenic mechanism for poor outcomes in

asthmatic patients (Baraldo et al., 2012) and MERS-CoV infection (Faure et al., 2014). In

animal models of hMPV infection it has been shown that a TLR 4 mediated inflammatory

response does not facilitate an adaptive immune response important for viral clearance

and protection against reinfection but predicts the progression of clinical disease

(Velayutham et al., 2013). In RSV infection, PRRs correlate with viral load and there is an

increased pulmonary expression of PRRs compared to healthy controls or infants with RV

or hBoV infection (Scagnolari et al., 2009).

Page 44: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

24

Different viral infections can elicit different inflammatory responses, and these

inflammatory responses can generally be grouped into T-helper 1 (Th1) and T-helper 2

(Th2) responses. These inflammatory responses are classified based on the type of

chemokines and cytokines produced. Th1 responses are characterised by production of

IFN-γ, IL-1β, IL-2, IL-12, IL-18, and TNF-α. The Th1 response is pro-inflammatory and is

important in the generation of immune responses necessary for the control and clearance

of intracellular pathogens, thus it is the most suitable response to viral infection. The Th2

response on the other hand is characterised by secretion of IL-4, IL-5, IL-6, IL-9, IL-10, and

IL-13. This type of response is involved in antibody production (including IgE) and

eosinophilic inflammation. This response is strongly associated with atopy and protection

against parasitic infection. It has also been demonstrated that Th2 inflammation counter

regulates Th1 mediated responses (Gill et al., 2010). Respiratory viral infections provoke

differing inflammatory profiles. For instance, IFAV can induce the overproduction of Th1

inflammatory mediators and this is associated with poor clinical outcomes. Dysregulated

secretion of TNF-α and IL-1β maybe critical for pathogenicity and may also hinder the

development of an effective adaptive immune response following infection (Han et al.,

2014). In RSV disease, sequential increases of IFN-γ coincide with improved clinical

outcomes and provide support for the protective role of IFN-γ following infection

(Bermejo-Martin et al., 2007a). Conversely, severe acute viral respiratory infection in

infants is associated with augmented Th2 responses (Bermejo-Martin et al., 2007b). In

addition, individuals with an underlying Th2 bias, such as those with an allergic pulmonary

condition, are more likely to have poor clinical outcomes compared with those without an

underlying Th2 bias (Gern et al., 2000; Mahmutovic Persson et al., 2016; Monick et al.,

2007).

Many types of cells have been implicated in the pathogenesis of ARTI and of note is the

quick and robust pulmonary and systemic neutrophil response following infection

Page 45: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

25

(Nagarkar et al., 2009; Sugamata et al., 2012). This robust neutrophil response correlates

with disease severity and is mediated primarily by IL-8 (Cortjens et al., 2016). Neutrophilia

is a common response to both bacterial and viral infections. While there are clear

protective roles for neutrophils against bacterial infection, the evidence for an anti-viral

role is less clear. The role of neutrophils has been investigated in RSV infection and they

are believed to play a role in pathogenesis (Cortjens et al., 2016; Linden, 2001;

Mahmutovic Persson et al., 2016). The role of neutrophils in influenza mouse models is

reported to be both protective and pathogenic. Some papers suggest neutrophils can

reduce viral load by phagocytosis of infected cells and trapping of free virus in neutrophil

extracellular traps. However, other reports have demonstrated that neutrophil

myeloperoxidase facilitates acute respiratory distress syndrome following influenza

infection and that mice lacking myeloperoxidase were more competent in reducing viral

load (Sugamata et al., 2012). Neutrophil involvement has also been demonstrated in RV-

induced asthma exacerbations and high counts are observed in the airways of individuals

following fatal exacerbations (Fahy, 2009; Fahy et al., 1995; Linden, 2001). In a RV mouse

model, IL-8 knock-out mice infected with RV showed significantly reduced neutrophil

responses, and demonstrated reduced airway hyper-responsiveness (Nagarkar et al.,

2009). Functional neutrophil activity has been demonstrated to be enhanced in the airways

of asthmatics, particularly during exacerbations, and correlates with reductions in lung

function and increases in symptom score (Proud, 2011). However, the precise contribution

of neutrophils to asthma has yet to be established.

Page 46: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

26

1.6 Aims of Project

The general aim of this thesis was to develop reliable quantitative PCR (viral load) assays to

investigate the contribution of certain respiratory viruses to clinical outcome of young

children hospitalised with an acute viral respiratory tract infection.

The specific aims were as follows:

To develop and validate the use of reliable in-house qPCR (viral load) assays for

different respiratory viruses including IFAV, hMPV, RSV-A&B, and RV-C.- Chapter 3

and 4 (RV-C only)

To compare the analytical performance of real time qPCR to digital PCR in the

quantitative measurement of RSV load in clinical samples - Chapter 3

To examine the contribution of RSV to clinical disease in young infants in a high HIV

prevalence setting- Chapter 5

To investigate the contribution of RV-C to disease severity in young children

presenting to emergency department with acute asthma exacerbations - Chapter

6.

To understand the interplay between RV-C infection and the innate immune

response in young children hospitalised with a RV-C associated wheezing illness -

Chapter 7

Page 47: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

27

2 Materials and Methods

2.1 Sample collection

Clinical specimens included flocked nasal swabs, per nasal aspirates, sputum and

nasopharyngeal aspirates samples. Swabs were placed in virus transport medium (VTM)

which contains Hanks balanced salt solution, with 50mg gentamicin, 0.1% bovine serum

albumin, 0.1% sodium bicarbonate and 8mM HEPES buffer).

Nasopharyngeal aspirates (NPAs) were collected from 105 ALRI cases and 53 controls from

Pretoria, South Africa between July 2011 and November 2012 (Chapter 5). Children 0-2

years of age hospitalized with an ALRI at the Steve Biko Academic Hospital or Tshwane

District Hospital in Pretoria were enrolled as cases. A diagnosis of pneumonia (respiratory

distress and either chest X-ray changes (e.g. consolidation or effusion), or auscultatory

findings (e.g. crepitations or bronchial breathing) or bronchiolitis (respiratory distress and

at least one of the following; wheeze, chest X-ray changes (e.g. hyperinflation) or

Hoovers’s sign (inward movement of the lower rib cage during inspiration) was determined

by the clinician in charge. Fifty-three age-matched children presenting to the same

hospitals with a non-respiratory illness over the same period were enrolled as controls

(NRD controls). The study protocol was approved by the University of Western Australia

Human Ethics Committee and the University of Pretoria Ethics Committee. Written

informed consent from parent/guardian was obtained prior to participation. NPAs were

stored at -80°C in Pretoria, South Africa until transfer on dry ice to Telethon Kids Institute,

Perth, Australia for processing and long term storage. An aliquot of each sample was

transported on dry ice to PathWest Laboratory Medicine WA, Perth, Australia.

For Chapter 6, flocked nasal swabs were collected from young children presenting with an

acute asthma exacerbation as part of the MAVRIC (Mechanisms of Acute Viral Respiratory

Page 48: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

28

Infection in Children) study carried out at Princess Margaret Hospital in Perth, Western

Australia. Flocked nasal swab specimens were also collected from children with no

evidence of respiratory disease. These children were selected to match the acute asthma

exacerbation cases for age, and season of birth. An aliquot of each sample was

transported on dry ice to PathWest Laboratory Medicine WA, Perth, Australia and stored

at -80oC until further use.

Study participants for the cytokine study (Chapter 7) had a flocked swab sample collected

upon inpatient admission. These samples were placed on ice and transported to the

PathWest laboratory as soon as practically possible for storage in -80oC freezer.

2.2 Nucleic acid extraction and viral detection

Total nucleic acids were extracted from 200µL of respiratory specimen using the MagMAX

viral RNA isolation kit (Thermo-Fisher Scientific, Australia) according to the manufacturer’s

instructions. All automated liquid transfer procedures utilised the CAS 1200 instrument

and conductive tips (Corbett Life Science. Australia)

An in-house multiplex respiratory pathogen assay was used to screen study samples for

respiratory pathogens such as influenza A, B and C viruses (FLUAV, FLUBV,FLUCV),

parainfluenza virus I-IV (PIV), RSV-A & B, Coronaviruses (hCoV- HKU1, -NL63, -OC-43, -

229E), human metapneumovirus (hMPV), and human adenoviruses (hAdV) (Chidlow et al.,

2009). Every multiplex PCR run performed included positive (PCR, inhibition and extraction

positive controls) and negative controls (method blanks interspersed by 5 test samples).

Nucleic acid extracts were stored at -80oC. A few minor adjustments were made to the

multiplex PCR real-time assay (Chidlow et al., 2009). Adjustments included the addition of

a 2.5 µM ROX reference dye (ThermoFisher), which was to be used for the 1:10 dilution of

amplicon generated from the enrichment PCR assays. Further, extra primer pairs were

Page 49: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

29

included to screen for hMPV (Chidlow et al., 2009). The primer pairs reported by Lee and

colleagues (Lee et al., 2007) were used for RV identification and genotyping. Respective

monoplex quantitative PCR assays were used to confirm any discordant results obtained

from the respiratory multiplex PCR assay. Thermal cycling programmes for the enrichment

PCR assays and the real time multiplex PCR assay were adopted from (Chidlow et al.,

2009).

2.3 Design of primers and probes

Assay oligonucleotides were designed using primer express v3.0 software (primers and

MGB probes) (ThermoFisher Scientific, Australia or the Exiqon microRNA PCR online

service (LNA probes).Primers were synthesised by Integrated DNA Technologies (IDT,

Australia), MGB probes were synthesized by Applied Biosystems (ThermoFisher Scientific,

Australia) and Locked Nucleic Acid (LNA) probes were synthesized by Sigma-Aldrich (Sigma-

Aldrich, Australia). The primers and probes used in the multiplex PCR to screen initially for

viral nucleic acid in respiratory samples were as previously described (Chidlow, 2013;

Chidlow et al., 2009).

Viral load from clinical samples was determined by real-time PCR targeting specific regions

of nucleoprotein gene of RSV (A&B) and HMPV, the matrix gene of IFAV and the

5’untranslated region (UTR) of RV-C (Table 2.1). These sequences were used because they

are highly conserved across all the respective strains investigated. Table 2.1 lists primers

and probes details for IFAV, RSV (A&B), HMPV, RV-C and Glyceraldehyde-3-phosphate

dehydrogenase (GAPDH) assay

Page 50: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

30

Table 2.1: Primers and probes used in quantitative real-time PCR assays

aLNA bases are underlined.

Target Target region Oligonucleotide sequence (Position) a

HMPV F (Forward Primer) Nucleoprotein gene 5’-ATCATCAGGYAAYATYCCACAAA-3’ (420 - 442)

HMPV R (Reverse Primer ) 5’- TATTAARGCACCTACACATAATAA-3’ (518 -542)

HMPV (Probe) 5’-FAM-CCTGCGTGGCTGCC-MGBNFQ-3’ (481- 497)

RSV-A F (Forward Primer) Nucleoprotein gene 5’CAACTTCTGTCATCCAGCAAA3’(1117 -1137)

RSV-A R (Reverse Primer) 5’TGCACATCATAATTAGGAGTATCAAT3’ (1166-1191)

RSV-A Probe 5’-FAM-CACCATCCAACGGAGC`3’-BHQ-1 (1140 – 1155)

RSV-B F (Forward Primer) Nucleoprotein gene 5’ATTCAACGTAGTACAGGAGATAATA3’ (1141 - 1165)

RSV-B R (Reverse Primer ) 5’CCACATAGTTTGTTTAGGTGTTT’ (1193 -1214)

RSV-B Probe 5’-FAM-TGACACTCCCAATTAT3’-BHQ-1 (1167 – 1182)

IFAV (Forward Primer) Matrix gene 5’CTTCTAACCGAGGTCGAAACGTA3’ (7-29)

IFAV (Reverse Primer ) 5’-GGTGACAGGATTGGTCTTGTCTTTA--3’ (137-161)

IFAV (Probe) 5’FAM-TCAGGCCCCCTCAAAGCCGAG3’-BHQ-1 (49-69)

RV-C IrlonS (Forward Primer) 5’UTR 5’-GCACTTCTGTTTCCCC-3’ (165 - 180)

RV-C EntA (Reverse Primer) 5’- GCATTCAGGGGCCGGAG-3’ (461 -445)

RV-C Probe 1 5’-FAM-CCTGCGTGGCTGCC-MGBNFQ-3’ (358 - 371)

RV-C Probe 2 5’-FAM-CCCGCGTGGCTGCC3’-BHQ-1 (359 - 372)

RV -C Probe 3 5’-FAM-CCCGCGTGGTGCCC-MGBNFQ-3’ (354 - 367)

RV -C Probe 4 5’ -FAM-CCTGCGTGGTGCCC3’-BHQ-1 (354 – 367)

GAPDH (forward) GAPDH mRNA 5’GAAGGTGAAGGTCGGAGTC3’ (7-25)

GAPDH (reverse) 5’AAATCCCATCACCATCTTC3’ (213-231)

GAPDH probe 5’-FAM-GGCTGAGAACGGGAAGCTTG-MGBNFQ

Page 51: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

31

2.4 Production and quantification of transcribed RNA

standards Nucleotide sequences matching a segment of the 5' UTR of RV-C2 [EF077280.1], RV-C6

[EF582387], RV-C51 [JF317015],RV-C25 [JF317013], RSV A [KJ627348]&B [KU950585] NP gene,

HMPV NP gene [AHV79765], IFAV matrix gene [CY056296] were incorporated into individual

plasmid constructs manufactured by Integrated DNA Technologies (IDT, Australia). M13

forward (5′-GTA AAA CGA CGG CCA GT-3′) and reverse (CAG GAA ACA GCT ATG ACC) primers

were used to amplify the target sequence from each individual plasmid construct. The PCR

reaction mix (20µL) contained PCR buffer (Thermo Fisher Scientific Australia Pty Ltd), 2mM

MgCl2, 0.2 mM dNTP, 0.2µM primers (IDT, Australia), 0.5 U AmpliTaq Gold® (ThermoFisher

Scientific, Australia) and cycling conditions were as follows: 10 minutes at 95°C followed by 45

cycles of 30 seconds at 94°C, 45 seconds at 50 °C and 60 seconds at 72°C. Successful

amplification was confirmed by the detection of PCR products by agarose gel electrophoresis.

Post PCR purification was completed using ExoSAP-It reagent (Affymetrix, Ohio, USA) following

the manufacturer’s protocol.

The Megashortscript T7 high yield transcription kit (ThermoFisher Scientific, Australia) was

used to synthesize RNA in vitro. All transcription reactions were completed at 37°C for 16

hours followed by TURBO DNA-free™ DNase treatment, DNAse Removal and MEGAclear™

Transcription Clean-Up (ThermoFisher Scientific, Australia) following the manufacturer’s

instructions. The RNA transcripts were eluted in THE RNA Storage solution (ThermoFisher

Scientific, Australia), and stored in single-use aliquots at -80°C.

The RNA transcript was quantified using the Qubit RNA Broad Range assay on the Qubit 2.0

fluorometer (Life Technologies, USA). To assess the quantification accuracy of the Qubit 2.0

Page 52: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

32

fluorometer all RNA transcripts were measured in triplicate. The conversion of RNA

concentration into RNA copies/µL was done with the following formulae:

1. M.W. of ssRNA = (RNA transcript length (bp) x 320.5) + 159.0

2. Number of molecules (copies) per ug ssRNA = Avogadros number (6.022*10^23) *

(M.W. of ssRNA)

3. RNA copies/µL= [RNA transcript concentration as per Qubit * number of molecules

(copies) per ug ssRNA] / (RNA transcript length)

2.5 Quantitative Real time PCR (Viral load)

The qScript XLT One-Step qRT-PCR Toughmix kit (Quanta Biosciences Gaithersburg, USA) was

utilized for the qRT-PCR assays. The 20 μL reaction volume contained 8 μL of template,

forward primer, reverse primer and hydrolysis probe. Thermocycler conditions were as

follows: 10 minutes at 50°C, followed by 3 minute incubation at 95°C then 40 cycles of 95oC for

20 seconds and 60°C for 60 seconds (80 seconds for RV-C assays to accommodate larger

amplicon size) using the Rotor Gene 6000 real-time thermocycler (Qiagen, Australia). All

experiments were performed in triplicate including positive controls and non-template

controls. We determined quantification cycle (Cq) values for each reaction using a manual Cq

threshold of 0.10 on the Rotor Gene 6000 application software.

In all experiments, a standard curve was generated by comparing Cq values and the copy

number. The reaction efficiencies of the assays were calculated according to the equation:

E=10(-1/M)-1, where M is the slope of the standard curve. An eight point standard curve (101 -

108 copies/µL), positive and negative control were mandatory on all verified runs. The viral

load in clinical samples was determined by interpolation of the quantification cycle value into

the appropriate standard curve.

Page 53: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

33

The reliability and reproducibility of viral load quantification by RT-qPCR was assessed using

the Minimum Information for Publication of Quantitative Real-Time PCR Experiments (MIQE)

guidelines (Bustin et al., 2009). Tenfold dilutions of each cRNA transcript were run in triplicate

to assess intra-assay variation. Inter-assay variation of Cq values was determined by analysing

data from five independent assays. Assay analytical sensitivity and specificity were also

evaluated.

2.6 Digital Droplet PCR

A laboratory developed test for RSV detection was used, together with a QX100 droplet digital

PCR system (Bio-Rad, USA). The ddPCR reaction mixture consisted of 7.5µL of a 5x ddPCR super

mix (Bio-Rad), 3µl of 2X reverse transcriptase, 15mM of DTT, 0.8mM of primers and 0.15mM of

probe mix and 8µL of sample nucleic acid solution in final volume of 20µL. The reaction mix

together with the 70µL of droplet generation oil was loaded into a di sposable plastic cartridge

and placed in the droplet generator (Bio-Rad). Following droplet generation each reaction-

sample mix was transferred to a 96 well PCR plate (BioRad). PCR amplification was performed

on a CFX-96 thermocycler (Bio-Rad) using a thermal profile beginning with a reverse

transcription step of 50oC for 30 minutes, then 95oC for 10 minutes followed by 40 cycles of

95°C for 30 seconds and 55°C for 60 seconds, 1 cycle of 98°C for 10 minutes, and ending at 4°C.

Following amplification, the plate was loaded on the droplet reader (Bio-Rad) and the droplets

from each well of the plate were read automatically at a rate of 32 wells per hour. DdPCR data

were analysed with QuantaSoft analysis software (Bio-Rad), and the quantification of the

target molecule was presented as the number of copies per µl of PCR mixture.

Page 54: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

34

2.7 PCR reagents

One step cDNA synthesis and PCR was performed using Superscript III one step RT-PCR system

(Thermofisher, USA) or Quanta qScript XLT one step RT-qPCR tough mix (QuantaBio,USA).

Modifications to the manufacturers instruction included reducing the reaction volume to 20 µL

and the enzyme to 0.5 µL and the addition of iStarTaq DNA polymerase (0.5 units/ reaction)

(Intron Biotechnology, South Korea). Further modification included the addition of 0.9mM of

DTT. Asymmetrical primer concentrations (0.4mM and 0.8mM for forward and reverse primers

respectively) were used for all RV-C assays.

2.8 Gel Electrophoresis

Nucleic acids were mixed with loading buffer and run on a 1 % agarose gel . Electrophoresis

was performed in 1x TAE buffer with a voltage of 200 volts for the time required to obtain

satisfactory migration, typically 30 minutes. DNA was visualised under UV light on a

transilluminator and digitally photographed using LabWorks Image Acquisition and Analysis

Software (UVP Bioimaging Systems).

2.9 Thermocyclers

Conventional PCR assays were performed in either 96 well plates (Axygen, USA) sealed with

Aluminium sealing film (Axygen, USA) or 0.2mL PCR tubes (ThermoFisher, USA) cycled using an

AB 2700 thermocyclers (Applied Biosystems, USA).

Real-time PCR experiments were conducted in either 0.2ml PCR tubes (Axygen, USA), 72 or

100 tube rings (Qiagen, Australia) or 96 well plates (Bio-Rad) or 384 well plates (Axygen, USA).

The rings and plates were sealed with a heat treated film. All experiments were conducted on

Page 55: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

35

the Rotorgene6000 thermocycler (Qiagen), the CFX-96 thermocycler (Bio-Rad) or The Applied

Biosystems ViiA 7 system (ThermoFisher).

2.10 Computational analysis

Specificity analysis was conducted using MegaBlast against human, viral and bacterial

sequences from the Genbank nucleotide collection.

Collection of target sequence for computational analysis was conducted using the virus

pathogen resource database (https://www.viprbrc.org/brc/home.spg?decorator=vipr), the

Picornavirus study group website (http://www.picornastudygroup.com/types/enterovirus/hrv-

c.html) and PathWest molecular diagnostics in-house respiratory virus database. Sequence of

the respective viral strains were downloaded and aligned to the appropriate primers and

probe sets. For each set of viral sequences, we determined the corresponding assays

numerical coverage at the strain level.

2.11 Multiplex immunoassay

The multiplex cytokine immunoassay kit was purchased from eBiosciences. Study samples

were sent to the manufacturer for processing. All samples were analysed for IFN-γ, IL-12p70,

IL-13, IL-1β, IL-2, IL-4, IL-5, IL-6, TNF-α, GM-CSF, IL-18, IL-10, IL-17A, IL-21, IL-22, IL-23, IL-27, IL-

9, IFN-α, IL-31, IL-15, IL-1α, IL-1RA, IL-7, TNF-β, Eotaxin, GRO-a, IL-8, IP-10, MCP-1, MIP-1b,

SDF-1 a, RANTES, IL-29 using a human cytokine 34-plex panel (eBiosciences, Australia)

Page 56: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

36

2.12 Quality Control

Appropriate personal protective equipment was put on at all times. All equipment was

maintained and serviced at regular intervals. Molecular grade reagents were used at all time.

Prepared reagents were made in large volumes and stored in single use aliquots where

appropriate. Batches of reagents were quality checked before use.

Contamination control measures included a unidirectional work flow and separate rooms with

dedicated equipment. All experiments were performed in triplicate including positive controls

and non-template controls. Detection of glyceraldehyde 3-phosphate dehydrogenase (GAPDH)

mRNA was utilized to ensure adequate specimen collection, RNA extraction and detection of

PCR inhibitors. Good quality samples were considered to be those with GAPDH Cq values

below 31.5. Standard laboratory procedure was followed and validated according to the

National Pathology Accreditation Advisory Council guidelines.

2.13 Statistical Analysis

All statistical analyses were carried out using SPSS statistical software version 16.0 (IBM Inc

Chicago, USA.). Significant differences between groups were evaluated using One Way-ANOVA

test, Chi-square or Fisher’s exact tests. Associations of RSV load with ALRI, RSV subtype or

multiple infections were examined using the Mann-Whitney U test. In all statistical analysis a

p-value <0.05 was considered statistically significant.

2.14 Ethics Approval

The RSV study protocol (chapter 5) was approved by the University of Western Australia

Human Ethics Committee and the University of Pretoria Ethics Committee. Written informed

Page 57: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

37

consent from parent/guardian was obtained prior to participation. The RV-C and disease

severity study (Chapter 6) was part of a larger study which was approved by the ethics

committee and Princess Margaret hospital in Perth, Australia where recruitment was

undertaken. Informed consent was obtained before inclusion in the study and the collection of

samples. Ethics approval was also obtained to conduct the study protocol used in chapter 7.

This approval was granted by Princess Margaret Hospital for Children Ethics Committee and

the Research Governance Office.

Page 58: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

38

3 The design and development of quantitative detection assays

for the common causative viral pathogens of acute lower

respiratory tract infection

Page 59: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

39

3.1 Introduction

Respiratory tract infections are a major cause of childhood morbidity and mortality worldwide

(Walker et al., 2013). Young infants, the elderly and the immunocompromised are population

groups most at risk of severe ALRI. Respiratory syncytial virus (RSV), influenza A viruses (IFAV)

and human metapneumovirus (HMPV) are the most common respiratory viruses identified in

these groups (Ruuskanen et al., 2011). These viruses are strongly associated with a wide range

of clinical manifestations ranging from mild upper respiratory tract infection to severe

pneumonia and death (Chidlow et al., 2012; Cohen et al., 2015b; Jafri et al., 2013; Moyes et al.,

2013; Pretorius et al., 2016; Takeyama et al., 2015a). The role of the virus in determining

disease severity is not clearly understood but several lines of evidence suggest that viral load is

an important contributor (Houben et al., 2010; Jansen et al., 2010; Li et al., 2010; Martin et al.,

2012). Therefore being able to accurately measure the amount of virus during illness may

provide insight into the effective contribution of the virus to disease outcome.

In many diagnostic laboratories reverse transcription quantitative PCR (RT-qPCR) is the gold

standard for the quantitation of respiratory viruses among hospitalized patients with ALRI

(Alsaleh et al., 2014; Chidlow et al., 2009; Do et al., 2010; Gueudin et al., 2003) . RT-qPCR is

preferred to other detection techniques because it provides superior detection sensitivities

and specificities and, of prime importance to this study, is the ability to quantify viral load in

clinical samples. Quantification of viral nucleic acid in clinical samples relies on the

amplification of the target sequence which in turn results in a detectable fluorescence signal

known as the quantification cycle value (Cq) (Niesters, 2001). Sample viral load is determined

by comparing the Cq value to a series of external standards representing serial dilutions of

known copy number. A crucial limitation of this approach is that accuracy of the calibration

Page 60: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

40

curve is highly dependent on reaction efficiency, a parameter that can be skewed by inhibitors

(Verhaegen et al., 2016). Droplet digital PCR (ddPCR) on the other hand is relatively new

approach that improves on qPCR by making external standards obsolete in viral quantification

(Huggett, Cowen, and Foy, 2015). In a similar approach to qPCR, ddPCR involves the detection

of template sequence with either a SYBR green or hydrolysis probe reaction chemistry but

quantification is conducted differently. It involves the generation of a large library of emulsion

based droplets (~20,000), also termed partitions (Markey et al., 2010). These partitions are

generated from sample-reagent mixtures and are distributed in such a way that at least a

proportion of them contain no template molecules (this is done in order to se parate and

isolate single molecules). Results are obtained by tallying the number of partitions in which the

template sequence is detected compared to the number of partitions in which there is no

amplification (Huggett et al., 2013).

The genetic diversity observed in the genome of most RNA viruses is a major challenge that

precludes the design of accurate and reliable viral load assays. As such, the aim of this chapter

was to design PCR based quantification assays that provide overall coverage of the known

genetic variability and subsequently evaluate the analytical and clinical performances.

Additionally, to further understand the utility of ddPCR in the clinical virology setting this

chapter thoroughly evaluates the differences in analytical and clinical performances between

ddPCR and RT-qPCR using synthetic RSV RNA and clinical samples from patients following RSV

infection.

Page 61: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

41

3.2 Samples

Clinical samples received at PathWest Laboratory Medicine WA were utilised for assay

validation. Specimen types included nasopharyngeal swabs, flocked nasal swabs and sputum.

Nucleic acid was extracted from samples using standard procedure (Chapter 2.2).

3.3 Results

3.3.1 Assay Design

The primer and probes sets of each assay were designed to target a highly conserved region of

the appropriate target sequence. Based on high quality multiple sequence alignment,

primer/probe sets were designed for the quantitative detection of HMPV (NP gene), RSV A and

B (NP gene) and Influenza A (matrix gene). Primer and probe sequence information including

target position for each assay are given in Table 3.1. Each assay consisted of two sequence

specific primers and a hydrolysis probe.

Page 62: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

42

Table 3.1 Primers and probes used for respiratory virus quantification assays

Oligonucleotide Oligonucleotide sequence (Position) a

HMPV F (Forward Primer) 5’-ATCATCAGGYAAYATYCCACAAA-3’ (420 - 442)

HMPV R (Reverse Primer ) 5’- TATTAARGCACCTACACATAATAA-3’ (518 -542)

HMPV (Probe) 5’-FAM-CCTGCGTGGCTGCC-MGBNFQ-3’ (481- 497)

RSV-A F (Forward Primer) 5’CAACTTCTGTCATCCAGCAAA-3’(1117 -1137)

RSV-A R (Reverse Primer) 5’TGCACATCATAATTAGGAGTATCAAT-3’ (1166-1191)

RSV-A Probe 5’-FAM-CACCATCCAACGGAGC-3’-BHQ-1 (1140 – 1155)

RSV-B F (Forward Primer) 5’ATTCAACGTAGTACAGGAGATAATA-3’ (1141 - 1165)

RSV-B R (Reverse Primer ) 5’CCACATAGTTTGTTTAGGTGTTT-3’ (1193 -1214)

RSV-B Probe 5’-FAM-TGACACTCCCAATTAT-3’-BHQ-1 (1167 – 1182)

FLUA MAT F (Forward Primer) 5’CTTCTAACCGAGGTCGAAACGTA-3’ (7-29)

FLUA MAT (Reverse Primer ) 5’-GGTGACAGGATTGGTCTTGTCTTTA-3’ (137-161)

FLUA MAT Probe 5’FAM-TCAGGCCCCCTCAAAGCCGAG-3’-BHQ-1 (49-69)

Page 63: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

43

3.3.2 In silico coverage analysis

In silico analysis was performed against 60 HMPV nucleocapsid sequences, 80 RSV -A and 75

RSV-B nucleocapsid sequences, and 80 IFAV matrix gene sequences retrieved from GenBank.

For each assay, representative sequences from different geographical regions over the last five

years were selected to provide coverage for most of the sequence diversity observed

worldwide. In silico analysis of the respective viral target sequences revealed negligible

sequence mismatches between target and primers/ probe sets. Subsequently, analytical and

clinical performances of each assay were evaluated to ensure satisfactory laboratory

performance.

3.3.3 The impact of PCR Master mixes on the variability of Cq values

In an attempt to identify which PCR master mix facilitates superior quantification results, a PCR

Master Mix comparison was performed between the Superscript III RT-PCR System and Quanta

qScript XLT one step RT-qPCR tough mix using 10 fold serial dilution of synthetic RNA of the

appropriate viral target. It can be seen in Fig. 3.1 that the two master mixes produced

comparable quantification values (Cq), ranging between 0.1 and 2 per dilution. When

amplification efficiencies of the two master mixes were compared, it was found that

amplification efficiency was consistently higher among assays prepared using the qScript XLT

one step RT-qPCR tough mix compared to assays prepared using Superscript III RT-PCR System

(Fig.3.2). Accordingly, the Quanta qScript XLT one step RT-qPCR tough mix was selected as the

master mix of choice to evaluate the laboratory performance of all the RT-qPCR assays

developed.

Page 64: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

44

0

5

10

15

20

25

30

35

40

45

Qu

anti

fica

tio

n c

ycle

val

ue

(C

q)

Standard diluton

qScript XLT

SSIII

A

0

5

10

15

20

25

30

35

40

Qu

anti

fica

tio

n c

ycle

val

ue

(C

q)

Standard diluton

qScript XLT

SSIII

C

0

5

10

15

20

25

30

35

40

45

Qu

anti

fica

tio

n c

ycle

val

ue

(C

q)

Standard diluton

qScript XLT

SSIII

B

0

5

10

15

20

25

30

35

40

Qu

anti

fica

tio

n c

ycle

val

ue

(C

q)

Standard diluton

qScript XLT

SSIII

D

Figure 3.1 A comparison of two commercial master mixes for the quantitative detection of RSV-A (A), IFAV (B), HMPV (C), RSV-B (D). Purified synthetic RNA of each respiratory virus

was prepared and the assays were perfomed in triplicate in serial 10 fold dilutions. For each assay, the data is plotted as the average Cq value for the triplicate samples prepared in the respective mastermixes.

Page 65: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

45

Figure 3.2 Comparison of amplification efficiency between qScript XLT mastermix and Superscript III RT-PCR System master mix. The assays prepared in the qScript XLT one step RT-qPCR tough mix generated significantly

higher amplification efficiencies than assays prepared in the Superscript III RT-PCR System (p=0.014).

3.3.4 Analytical Sensitivity

In order to evaluate the analytical performances of the developed quantification assays, it was

necessary to assess linearity, limit of detection, amplification efficiency and goodness of fit.

This experiment was performed using serial dilutions of the appropriate synthetic RNA

transcripts prepared in PCR grade water. The analyses shown in Table 3.2 indicate that the

respiratory quantification assays herein provided excellent amplification efficiencies with

minimal variation between runs, a wide linear dynamic range (>7 orders of magnitude) and

good agreement between theoretical values and experimental values.

88

90

92

94

96

98

100

RSV-A RSV-B IFAV HMPV

Am

plif

icat

ion

eff

icie

ncy

(%

)

Assay

qScript

SSIII

Page 66: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

46

Table 3.2 Analytical performance data on IFAV, RSVA, RSVB and HMPV quantitative assays using synthetic R NA

transcript

IFAV RSV-A RSV-B HMPV

Mean ± SD %CV Mean ± SD %CV Mean ± SD %CV Mean ± SD %CV

Slope -3.33 ± 0.04 1.01 -3.35± 0.01 0.06 -3.32 ± 0.02 0.46 -3.31 ± 0.04 1.15

Efficiency 1.00 ± 0.01 1.46 0.99 ± 0.01 0.08 0.99 ± 0.01 0.64 1.01 ± 0.02 1.62

Y-intercept 39.77± 2.86 7.20 41.71 ± 1.57 3.75 40.01 ± 3.20 8.00 38.36 ± 1.33 3.46

Goodness of fit (R2) 0.999 0.01 0.999 0.00 0.999 0.01 0.999 0.00

Range of Linearity 101-108 101-108 101-108 101-108

Limit of Detection

(copies/µL)

7 7 6 4

3.3.5 Analytical specificity

The specificity of each assay was evaluated with nucleic acid extracts from other respiratory

pathogens, including influenza A and B, human respiratory syncytial virus, parainfluenza

viruses 1-4, human adenovirus, human bocavirus, human coronaviruses (HCoV-229E, HCoV-NL-

63, HCoV-OC43, and HCoV-HKU-1), Mycoplasma pneumoniae and Streptococcus pneumoniae.

All assays were non-reactive when tested against these other respiratory pathogens. In

addition, a BLASTn search performed to check the specificity of the primer and probe sets used

in the assays showed no genomic cross-reactivity with other virus families, bacteria or cells.

Page 67: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

47

3.3.6 Repeatability and Reproducibility

Assay reproducibility and repeatability were evaluated using four artificial respiratory samples

constructed from high (106), moderate (104 and 102) and low (101) copy number for each viral

type. Repeatability and reproducibility of each quantitative detection assay was evaluated in

triplicate and over five independent experiments respectively. Repeatability which was

defined as the degree of variation between replicates within the same run demonstrated

relatively low coefficient of variation (CV) for all assays(table 3.3). As can be clearly seen from

table 3.3 repeatability was a factor of input copy number. Reproducibility, which was defined

as the degree of variation between runs demonstrated a coefficient of variation of less than

20%, with variability increasing proportionally with dilution of transcript.

Page 68: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

48

Table 3.3 Repeatability and reproducibility of quantitative detection assays for HMPV, IFAV, RSV-A and RSV-B

Repeatability a Reproducibility b

RNA target and input target copies

Quantity range (Calculated copies/ reaction)

%CV range

Quantity Mean (Calculated copies/reaction)

%CV

HMPV 106 1270000-1420000 0.50-3.01 1,327,400 3

HMPV 104 10260-12230 0.65- 3.76 13,443 4

HMPV 102 100-140 0.91-4.20 121 7

HMPV 101 11-15 6.09-14.89

15 11

RSV-A 106 1370000-1460000 0.70-2.30 1,435,000 7

RSV-A 104 13500-14200 0.58-2.90 13,898 10

RSV-A 102 135-143 0.89-3.60 139 14

RSV-A 101 12-17 8.03-15.40

15 16

RSV-B 106 1301000 - 1408000 0.65-2.48 1,376,667 5

RSV-B 104 13300-15060 0.48-2.78 14,118 8

RSV-B 102 101-140 0.91-3.74 123 12

RSV-B 101 10-19 5.03-10.06

14 14

IFAV 106 7390000-8180000 0.91-2.63 7807500 6

IFAV 104 75900-83900 1.02-3.05 79848 11

IFAV 102 705-778 1.34-4.83 741 15

IFAV 101 59-83 4.99-11.48

69 18

% CV – Percentage coefficient of variation a Assays were performed in triplicate b Five independent experiments

Page 69: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

49

3.3.7 Evaluation of Clinical specimens

Respiratory samples (n=100) were obtained from hospitalised patients with an acute lower

respiratory tract previously screened for a broad range of respiratory pathogens using

standard diagnostic PCRs, that were reactive for either HMPV (n=20), RSV-A (n=20) , RSV-B

(n=20) or IFAV (H1N1[n=20] and H3N2 [n=20]). All samples met the criteria for adequate

sample collection and nucleic acid extraction with GAPDH Cq values of less than 31. Fig.3

clearly shows that each assay is capable of determining a broad range of viral loads in clinical

samples.

Figure 3.3 Box plots of viral loads from patients infected with either IFAV (n=40), HMPV (n=20), RSV-A (n=20),

RSV-B (n=20). Viral load values ranged between 2 log10 copies/mL and 10 log10 copies/mL.

Page 70: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

50

3.3.8 Real-time PCR vs Digital droplet PCR

It was of interest to compare the analytical and clinical performance of the current gold

standard for molecular quantification (RT-qPCR) to the relatively new digital droplet PCR (RT-

ddPCR). It was too costly an exercise to test all 4 assays thus the RSV-A assay was selected

arbitrarily for this comparison.

Dilutions of in vitro transcribed RSV-A RNA were tested to evaluate analytical performance

characteristics such as accuracy, limit of detection, linearity, and repeatability. Figure 3.4

shows the relationship between measured RNA concentration and nominal RNA concentration

of the two PCR approaches using in vitro synthesized RSV-A RNA. It can be seen from this

figure that both methods showed a strong linear (>0.98) relationship between predicted and

observed values and that RT-qPCR was slightly more accurate than RT-ddPCR (as determined

by the slope value of the trend line). It was also noted that the quantitative value assigned by

both PCR instruments to the RNA standards across the concentration range was approximately

a 1.0 log10 lower than the expected copy number.

Page 71: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

51

Further analytical performance characteristics are illustrated in Fig.3.5. It is evident from these

data that the qPCR assay demonstrated a wider linear dynamic range (spanning more than 6

orders of magnitude) compared to ddPCR assay, which was subject to droplet saturation at

every instance where input RNA concentration was higher than 105 copies. However, the RT-

ddPCR assay demonstrated a lower detection limit compared to the RT-qPCR assay (2 copies vs

7 copies). Importantly for the validity and reliability of the ddPCR data was the clear

discrimination between positive and negative droplets at all measurable RNA input

concentrations, specifically at low concentrations (Fig. 3.5C).

RT-qPCR

y = x - 0.9075

R² = 1

RT-ddPCR y = 0.8843x - 0.4405

R² = 0.9856 0

2

4

6

8

0 2 4 6 8 10

Me

asu

red

RN

A c

on

cen

trat

ion

(lo

g10

co

pie

s/m

L)

Nominal RNA concentration (Log10 copies/mL)

RT-qpcr

RT-ddpcr

Linear (RT-qpcr)

Linear (RT-ddpcr)

Fig. 3.4 A comparison of accuracy between RSV-A RT-qPCR assay and RSV– A RT-ddPCR assay. Each point signifies the mean log10 copy number of duplicate samples. Accuracy was determined using the slope value of the trend line.

Page 72: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

52

Figure 3.5 Linear quantitative range of RSV-A assay constructed using serial dilutions of artificially synthesized RSV-A RNA and performed using either the RT-ddPCR assay (A) or RT-qPCR (B). RT-qPCR demonstrated a broader linear dynamic range but ddPCR demonstrated higher analytical sensitivity. RFU (Relative fluorescence units) measured in nanometres. (C) A 2D amplitude plot for varying RNA input concentrations (100-105) on the ddPCR instrument. The red line designates the threshold used to discriminate between clusters of positive and

negative droplets.

B

A

C

Page 73: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

53

Repeatability of each method was evaluated using replicates of in vitro RSV RNA transcript at

varying dilutions (104, 103,101,100). It is clear from Fig. 3.6 that the ddPCR assay exhibited

superior precision compared to the qPCR assay, especially at lower concentrations. The high

CV observed at 1 copy (RT-ddPCR) can be attributed to the stochasticity of the platform when

approaching its limit of detection.

Figure 3.6 Precision evaluations between RT-ddPCR and RT-qPCR. %CV was calculated by dividing the standard deviation over the mean of the replicate values. Overall and at each

concentration, RT-ddPCR demonstrated superior precision compared to RT-qPCR.

0

10

20

30

40

50

60

70

1.00E+04 1.00E+03 1.00E+01 1.00E+00

Co

eff

. o

f va

riat

ion

(%

)

RSV-A copies/µL

RT-ddPCR RT-qPCR

Page 74: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

54

3.3.9 Clinical evaluation of RT-ddPCR

Finally, to evaluate the clinical applicability of RT-ddPCR, 22 known RSV-A positive clinical

samples were each tested in duplicate with the two PCR methods and compared for positivity

rate and goodness of agreement (Fig 3.7). The analysis showed no difference in positivity rate

between the two methods. However, RT-ddPCR assay could not accurately assign viral copy

number to clinical samples (n=5) with input concentration greater than 105 copies but this was

not a problem with the qPCR assay. Figure 3.7 shows a Bland-Altman plot employed to assess

goodness of agreement between the two platforms. As may be seen from the plot, there was

no significant difference in copy number. The difference values of the measurable samples

(n=17) were within ± 1.96 standard deviations (0.31 log10 copies/mL) of the mean difference.

Further, we found no significant trend in proportions above or below the mean.

Figure 3.7 Bland-Altman plot. Viral loads of known RSV-A positive clinical samples were used for agreement evaluation between the RT-ddPCR and RT-qPCR. Mean differences and the

95% limits of agreement were calculated and are illustrated in the graph above.

Page 75: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

55

3.4 Discussion

This study describes the development and validation of accurate and robust qPCR assays that

can be utilized for the quantitative determination of viral load for a range of clinically relevant

viral respiratory pathogens. These viruses are most commonly associated with severe lower

respiratory tract infections and as such it is imperative that the virological factors driving

disease severity are understood. Indeed, recent reports emphasize that viral load maybe a

significant risk factor associated with disease outcome in hospitalized children (DeVincenzo et

al., 2010; El Saleeby et al., 2011; Roussy et al., 2014; To et al., 2010). Thus, the development of

reliable quantification assays provides an essential tool to examine the effective contribution

of the viral pathogen to disease outcome.

The design of primer and probe sets for any given assay should be a cautious approach to

ensure the assays developed are sensitive for the target and specific enough to be non-

reactive to unsuitable genomic material. Moreover, when designing assays it is important to

target highly conserved regions and not regions with a high incidence of mutations; since

mismatches between assay and target sequence may result in grossly inaccurate viral load

measurements (Hoffman et al., 2008; Letowski, Brousseau, and Masson, 2004; Randhawa et

al., 2011; Whiley and Sloots, 2005). For that reason we chose the nucleocapsid gene (HMPV,

RSV-A and RSV-B) and the matrix gene of IFAV as amplification targets. In silico analysis

indicated that the primer and probe sequences of all the assays provided satisfactory

coverage when aligned to the appropriate sequences that represented the known gene tic

variation worldwide over the last five years. Further, in silico analysis indicated that our assays

provided coverage for the known genetic variation within the appropriate target sequence

with minimal mismatches, crucially no mismatches in the probe target region. It has to be

Page 76: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

56

acknowledged that In in silico sequence homology between target and assay primer and probe

sets is not an accurate predictor of test performance (Morales and Holben, 2009). Therefore it

was important to evaluate performance of these assays using clinical material. Indeed, the

assays herein demonstrated excellent clinical performance indicating that these assays can be

utilised in a diagnostic setting.

Owing to an increase in the availability of commercial master mix kits, diagnostic laboratories

face the challenge of identifying the most appropriate reagents for their real time PCR

applications. Indeed, it has been demonstrated that different reaction components can affect

assay performance including assay sensitivity and reproducibility (Stephens, Hutchins, and

Dauphin, 2010; West and Sawyer, 2006). This chapter examined two master mixes for use in

the quantitative detection of HMPV, IFAV, RSV-A and B. Comparison of Quanta qScript XLT one

step RT-qPCR tough mix and the Superscript III RT-PCR System, after the application of the

appropriate synthetic RNA to the respective assays revealed no difference between the two

kits in terms of linear dynamic range. This was not surprising as the technical bulletin provided

with each system generally supported this. However, a significant discrepancy in amplification

efficiency was observed between the assays prepared in Quanta qScript XLT and the

superscript III system. We found that Quanta qScript XLT yielded consistently better

amplification efficiencies in comparison to assays prepared in Superscript III RT-PCR System.

One may speculate that the variability in amplification efficiency maybe a result of

thermocycler performance (Stephens et al., 2010), alternatively it may also indicate that

Quanta qScript XLT one step RT-qPCR tough mix facilitates a more efficient cDNA synthesis

compared to Superscript III system. In concordance with our finding is a previous study that

evaluated the laboratory performance of four RT systems and showed that the Superscript III

system was one of the worst performing RT kits in terms RNA-to-cDNA conversion capacity

Page 77: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

57

(Levesque-Sergerie et al., 2007). Taken together, this finding underscores the importance of

the reverse transcription step in developing accurate quantification assays. It al so emphasizes

the importance of evaluating PCR master mixes to ensure a suitable selection is made based

on the individual laboratory needs.

All assays demonstrated high amplification efficiencies, broad dynamic range and were non-

reactive when tested against other respiratory pathogens which suggest that all assays can

accurately quantify a wide range of viral loads with remarkably high specificity. Further, the

assays herein demonstrated relatively low intra-assay and inter-assay variation indicating that

data generated from these assays are reliable and reproducible.

Contention still exists about which PCR technology provides accurate and reliable viral load

data. Thus, performance characteristics of ddPCR were compared to qPCR for the quantitative

detection of RSV in clinical samples was evaluated using an already optimized RT-qPCR

protocol adapted for RT-ddPCR. Hayden et al. (2013) showed in their work that qPCR

demonstrated superior sensitivity and less variability across the concentration range for

clinical samples and reference material compared to ddPCR. Yet, in direct contrast to the

Hayden et al. (2013) report are recent studies that indicate that ddPCR demonstrates superior

precision and sensitivity across the measurable concentration range. For example, work

comparing ddPCR to qPCR for quantitative determination of hepatitis B viral load

demonstrated that ddPCR provides improved analytical sensitivity and specificity and as such,

is suitable for hepatitis B viral load determination in clinical samples (Tang et al., 2016). In

other recent work Palatnik de Sousa et al. (2015) evaluated ddPCR for influenza vaccine

development and demonstrated a high throughput ddPCR method for very precise and

accurate influenza virus titre quantification. The authors also noted several key issues that are

Page 78: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

58

determinants of variability in qPCR were avoided with the ddPCR approach. Coudray-Meunier

et al. (2016) report that ddPCR provided improved precision and analytical sensitivity and

concluded that digital PCR may have an important tool in human pathogenic virus surveillance

and outbreak investigation and may be beneficial to public health. The finding herein is

consistent with reports that suggest that sample partitioning provides superior sensitivity and

precision compared to qPCR. Our analyses also demonstrated an excellent level of agreement

for viral load values in clinical samples between the two PCR approaches which indicates that

ddPCR can be utilized as an alternative platform for the reliable absolute quantification of RSV

(and possibly other pathogenic respiratory viruses) in clinical samples (Coudray-Meunier et al.,

2016; Hayden et al., 2013; Tang et al., 2016). A major drawback of ddPCR is the finite amount

of droplets (~15,000) that can be generated by the current ddPCR instrument. This drawback

results in a relatively narrow linear dynamic range with a remarkably low upper limit of

quantification (105copies; ~21 Cq) compared to qPCR. Other work report similar inaccurate

measurements when sample copy number exceeds 105 copies (Coudray-Meunier et al., 2016;

Palatnik de Sousa et al., 2015). Indeed, sample dilution to an input concentration that falls

within the linear dynamic range of the ddPCR assay may mitigate this problem (Palatnik de

Sousa et al., 2015). Additionally, RT-ddPCR has relatively larger turnaround times compared to

qPCR (6.5hrs vs 3.5 hrs), thus may need to be streamlined prior to being introduced into the

clinical setting. Nonetheless, the findings of this study showed that ddPCR fulfils most of the

requirements of a reliable molecular quantification tool for the field of clinical virology, with

the added value of avoiding the need for calibrators. Theoretically the high precision and

sensitivity provided by ddPCR lends itself to detection of rare variants in mixed vi ral

populations that may arise from adapting to challenges such as the host immune response and

anti-viral therapeutics. However, to achieve its full potential in the clinical setting ddPCR needs

Page 79: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

59

to be further optimized to match the linear dynamic range of RT-qPCR (>9 orders of

magnitude).

There are limitations to acknowledge. Firstly there is a wide range of respiratory samples and

collection methods, which may have an impact on the resulting quantitative analysis.

Secondly, it has previously been shown that mismatch between primer and probe sequences

arising from sequence variability in the target region is an important determinant of inaccurate

viral load. Although a thorough analysis was undertaken to accommodate spatial and temporal

sequence variation for the respective assay target regions. Sequence analysis cannot predict

future point mutations thus ongoing surveillance is required to monitor changes in the target

sequence. Thirdly, accurate quantitative detection of viral target sequence relies on properly

constructed standard curves. This means technical errors in pipetting and preparation of

standards may substantially contribute to inaccuracy. Also it is important to note that optimal

RNA to cDNA conversion is a crucial determinant of accurate quantification measurement

because both RT-ddPCR and RT-qPCR can only measure the amount of DNA that is present.

Developing an accurate and reliable method for viral quantification is essential when

attempting to establish standardized definitions for clinical disease and for therapeutic

response. Pertinent to the correct interpretation of data generated from these assays is a

thorough interrogation of the developed assays prior to implementation in the clinical setting.

It appears that qPCR remains the gold standard for the molecular quantification of viral nucleic

acid in clinical samples but with further instrument optimization ddPCR can provide a useful

alternative.

Page 80: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

60

Page 81: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

61

4 The development of a reliable PCR assay to measure RV-C load

in clinical samples

Page 82: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

62

4.1 Introduction

Rhinoviruses (RV) are a common cause of acute respiratory infection in people of all ages

(Fawkner-Corbett et al., 2015; Puro et al., 2005). RVs are antigenically diverse, thus people can

be infected with different RV types over the course of a lifetime (Cooney, Fox, and Kenny,

1982; Cooney et al., 1975). The spectrum of disease associated with this group of viruses can

range from asymptomatic infection, mild upper respiratory tract infection (common cold) to

severe lower respiratory tract infections which may include bronchiolitis or pneumonia (Choi

et al., 2015; Iwane et al., 2011; Luchsinger et al., 2014). RV has been identified as an important

contributor to acute asthma (Bizzintino et al., 2011) and wheezing illness in young children and

is an important risk factor in the development of asthma later in life (Jackson et al., 2008).

Further, RV exacerbates pre-existing airway diseases such as asthma, cystic fibrosis and

chronic obstructive pulmonary diseases (Camargo et al., 2012; Kennedy et al., 2014;

Luchsinger et al., 2014).

Genome arrangement, capsid properties and conserved sequences are the current basis of RV

species classification, of which there are three recognized species (RV -A, RV-B, and RV-C)

(McIntyre, Knowles, and Simmonds, 2013). RV-C is the most recently described species and to

date there are 55 recognised genotypes. Clinical significance of RV-C is still debated as some

studies report that RV-C causes more severe disease than the other two species (Bizzintino et

al., 2011; Cox et al., 2013; Miller et al., 2009) but others have not found this association (Iwane

et al., 2011; Linsuwanon et al., 2009). Inaccurate quantitative methods complicate the

evaluation of viral factors that may contribute to disease severity (Schibler et al., 2012). Viral

load studies of other viruses have shown that the amount of replicating virus is an important

Page 83: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

63

contributor to disease severity (Franz et al., 2010; Jansen et al., 2010), thus an accurate and

reliable method of quantifying RV-C load may be an important tool in understanding the

contribution of RV-C to disease pathogenesis, disease progression and clinical management.

Conventional culture methods used to measure viral load in clinical samples are not suitable

for RV-C types as this virus is non-cultivable using traditional techniques. Molecular methods

have overcome this problem, but the inter-genotype sequence variation within the target

region prevents the design of a single quantitative assay that quantifies all genotypes at equal

efficiencies (Schibler et al., 2012). Furthermore, developing specific primer and probe

combinations for each genotype would be impractical in a diagnostic setting. This study aims

to develop and validate a minimum set of PCR assays required to quantify circulating RV-C

genotypes found in children.

Page 84: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

64

4.2 Samples

Nasopharyngeal aspirates NPAs (n = 40) were collected between June 2013 and April 2015

from children presenting to the Emergency Department of Princess Margaret Hospital for

Children in Perth. These samples represented a subset of children with episodic wheeze

enrolled in The Prednisolone Response Evaluation in Viral Induced Episodic Wheeze (PREVIEW)

study. This study was approved by the Princess Margaret Hospital for Children Ethics

Committee (1970/EP).

Total nucleic acids were extracted from 200µL of each respiratory specimen using the

MagMAX viral RNA isolation kit (Chapter 2.2) according to the manufacturer’s instructions.

Detection of the glyceraldehyde 3-phosphate dehydrogenase (GAPDH) reference mRNA

(Gueudin et al., 2003) was utilized to ensure adequate specimen collection, RNA extraction

and removal of PCR inhibitors. Good quality samples were considered those with GAPDH

quantification cycle values (Cq) no higher than 2 standard deviations (6.4) of the mean Cq value

(25.1). Analysis was not performed on samples above the predetermined accepted range.

The primers used in this study were based on previously published primer sequences (Table 1)

which amplify a 296bp region within the 5’ untranslated region (UTR) of RV species (Gama et

al., 1988; Ireland, Kent, and Nicholson, 1993). Rhinovirus species identification was performed

using a published semi nested PCR assay (Ireland et al., 1993) followed by sequencing.

Two hundred and thirty four RV-C 5’ UTR sequences which represented 34/55 of the currently

known RV-C genotypes were evaluated for the design of the minimum amount of probe

sequences required to overcome the inter-genotypic variation within the target region . These

sequences were obtained from our in-house RV-C database (n=218) and the Picornaviridae

Page 85: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

65

study group website (n=16) (http://www.picornastudygroup.com). Nucleotide sequence

alignments were analysed using BioEdit Sequence Alignment Editor Version 7.2.5 (Hall, 1999).

The reliability and reproducibility of RV-C viral load quantification by RT-qPCR was assessed

using the Minimum Information for Publication of Quantitative Real -Time PCR Experiments

(MIQE) guidelines (Bustin et al., 2009). Tenfold dilutions of each cRNA transcript were tested in

triplicate to assess intra-assay variation. Inter-assay variation of Cq values was determined by

analysing data from five independent assays.

Using the appropriate RNA transcript for each RV-C assay, a tenfold dilution series of twelve

concentrations was prepared. The second last dilution of the tenfold dilution series was used

to prepare a two-fold dilution series of 10 concentrations. 8µL of each dilution from the two-

fold dilution series was added to a 12 µL PCR reaction mix and run for 50 PCR cycles using the

Rotor Gene 6000 real-time thermocycler (Qiagen, Australia). Twenty-four PCR replicates were

tested at each concentration. Poisson regression analysis was used to determine the limit for a

95% confidence of detection. To evaluate variability these experiments were repeated on five

different occasions.

Analytical specificity was assessed using BLAST searches against other virus families, bacteria

and cell sequences from the Genbank nucleotide collection. In addition, an in -house cross

reactivity panel was used to assess the specificity of our RV-C assays against other respiratory

pathogens.

Page 86: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

66

4.3 Results

4.3.1 Validation of real-time PCR assay for RV-C viral load quantification

We designed four assays based on RV-C 5’UTR sequences belonging to the 34 RV-C genotypes

for which 5’UTR sequences were available. All assays used a common primer pair, but with

different specific probe sequences (Fig 4.3-1). In silico analysis demonstrated that the probe

sequence of assay-1 was homologous to the probe target region of 22 of the 34 RV-C

genotypes, while the probe target region of the remaining 12 genotypes aligned completely to

the probe in either assay- 2, -3 or-4 (Appendix 10.1). These assays were unable to be assessed

against the other 21 known genotypes because the 5’UTR sequence information was

unavailable.

Figure 4.3-1 A BioEdit sequence alignment of primer and probe regions that were targeted by assays one to four. Sequences of

forward primer region (left box), probe region (center box) and reverse primer region (right box). Identical bases at the same position are represented by dots whereas capitalized bases indicate mismatches between sequences.

All qPCR assays were optimized for primer concentration and annealing/extension

temperature. The optimal annealing temperature was determined to be 60oC with a

denaturation time of 20 seconds and an annealing/extension time of 80 seconds. The PCR

conditions were selected to produce the maximum fluorescent signal generated after 40

amplification cycles.

Serial 2-fold dilutions of 10 concentrations of each RNA transcript was prepared in PCR-grade

water and tested to determine the limit of detection of the assays. Using Poisson regression

Page 87: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

67

analysis, the limit for a 95% probability of detection was estimated to be 1147 copies/mL for

assay-1, and 4765 copies/mL, 1138 copies/mL and 1470 copies/mL respectively for assays 2-4.

Nucleic acid extracts from other respiratory pathogens, including influenza A and B, human

respiratory syncytial virus, human metapneumovirus, parainfluenza viruses 1-4, human

adenovirus, human bocavirus, human coronaviruses (HCoV-229E, HCoV-NL-63, HCoV-OC43,

and HCoV-HKU-1), Mycoplasma pneumoniae and Streptococcus pneumoniae were non-

reactive in each of the RV-C real-time assays. In addition, a BLASTn search performed to check

the specificity of the primer and probe sets used in the assays showed no genomic cross -

reactivity with other virus families, bacteria or cells. However as anticipated there was cross

reactivity with other enterovirus species.

Linearity was assessed in triplicate over five independent experiments, and in all assays it

spanned more than 7 orders of magnitude (Table 4.3-1). All assays demonstrated a strong

linear relationship (r2=>0.995) between Cq values and RNA copy number (Table 4.3-2). All

assays demonstrated amplification efficiencies of more than 95% (Table 4.3-2).

Page 88: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

68

Table 4.3-1 A comparison of RNA transcript concentration and Cq values for the different RV-C assays

RV-C Assay-1 RV-C Assay-2 RV-C Assay-3 RV-C Assay-4

RNA transcript

concentration

Mean Cq +/-

SD

Mean Cq +/-

SD

Mean Cq +/-

SD

Mean Cq +/-

SD

100 32.77+/-0.14 33.25+/-1.48 32.66+/-0.31 31.84+/-1.01

101 29.11+/-0.12 28.22+/-0.46 29.62+/-0.06 27.37+/-0.07

102 25.86+/-0.11 25.37+/-0.06 25.95+/-0.07 24.20+/-0.14

103 22.07+/-0.06 22.25+/-0.07 22.52+/-0.04 20.96+/-0.03

104 18.68+/-0.06 18.91+/-0.06 19.08+/-0.08 17.53+/-0.10

105 15.35+/-0.04 15.39+/-0.04 15.52+/-0.11 14.20+/-0.14

106 11.73+/-0.1 13.13+/-0.11 12.05+/-0.08 10.79+/-0.04

107 8.5+/-0.04 8.78+/-0.02 8.55+/-0.02 7.81+/-0.06

108 5.27+/-0.12 5.17+/-0.2 5.19+/-0.16 4.77+/-0.07

% CV – Percentage coefficient variation, Cq- quanti fication cycle va lue , SD- s tandard deviation

Page 89: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

69

Table 4.3-2 the performance of the individual PCR assays for the detection of matched RV-C RNA transcript

RV-C Assay-1 RV-C Assay-2 RV-C Assay-3 RV-C Assay-4

Mean +/- SD CV% Mean +/- SD CV% Mean +/- SD CV% Mean +/- SD CV%

Slope (n=) -3.32+/-0.11 3.33 -3.38+/-0.11 3.23 -3.44+/-0.09 2.72 -3.37+/-0.05 1.52

Efficiency 0.98+/-0.05 5.02 0.97+/-0.05 4.68 0.95+/-0.04 3.93 0.97+/-0.03 3.12

Y-intercept 34.00+/-2.46 7.23 38.00+/-2.72 7.17 36.12+/- 1.00 7.55 33.45 +/- 1.59 8.42

Goodness of fit (R2) 0.999 0.11 0.999 0.47 0.999 0.09 0.999 0.08

Range of Linearity 100-108 copies/ml 100-108 copies/ml 100-108 copies/ml 100-108 copies/ml

Page 90: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

70

4.3.2 Repeatability and reproducibility

To evaluate repeatability and reproducibility of each assay, dilutions (101,102,104,106) of RNA

transcript were tested in triplicate. Intra-assay %CV of the four RV-C assays ranged from 0.10%

- 8.58% and in most cases variability increased proportionally with dilution (Table 4.3). Inter-

assay variability was evaluated using results from five independent experiments and

demonstrated %CV of less than 15% (Table 4.3).

Table 4.3-3 Intra and Inter assay variability of the four RV-C qRT-PCR assays (Assay 1-4)

Intra-assay variation a

Inter-assay variation b

RNA target and input target copies

Quantity range (Calculated copies/ reaction)

%CV range Quantity Mean (Calculated copies/reaction)

%CV

RV-C1 106 3030000-3560000 0.16-2.39 3480000 6.89

RV-C1 104 30000-36700 0.27-2.33 37500 5.67

RV-C1 102 125-409 0.16-7.07 361 8.92

RV-C1 101 21-40 2.1-7.33 39 5.88

.

RV-C2 106 3560000-4210000 0.22-0.61 3810000 7.50

RV-C2 104 39100-49700 1.70 -2.42 43400 10.56

RV-C2 102 379-405 1.44 -2.75 396 3.04

RV-C2 101 35-42 3.89-5.82 39 7.22

RV-C3 106 4080000-5790000 0.37-2.30 4760000 14.58

RV-C3 104 35700-46200 0.23-1.23 42000 9.26

RV-C3 102 372-470 0.40-8.58 440 9.21

RV-C3 101 42-59 0.93-7.78 47 14.57

RV-C4 106 4020000-4860000 0.10-1.77 4360000 8.22

RV-C4 104 48500-52800 0.28-1.61 50000 4.89

RV-C4 102 423-608 1.07-4.76 525 11.05

RV-C4 101 42-49 1.47-5.16 46 5.36

% CV – Percentage coefficient variation a Assays were performed in tripl icate b five independent experiments

Page 91: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

71

4.3.3 Probe mismatch

To demonstrate the need for separate assays this study examined the impact of probe -target

sequence mismatches on viral load. Each RV-C transcript was prepared at seven different

concentrations ranging from 107 to 101 copies/µL with the calculated copy numbers (means of

three experiments) for each transcript expressed as percentages of the copy number obtained

with the perfectly matched RV-C transcript-1. At concentrations between 107 and 102

copies/µL there was minimal (<15%) difference in copy number yield between transcript-1 and

transcript-2. However, at the lowest copy number (101), a single nucleotide mismatch (near

the 5’end) in the probe target region (Fig. 4.3-1) resulted in an inaccurate viral load

determination (Table 4.3-4). Multiple mismatches between the probe and target (transcript-3

and-4) resulted in substantial inaccuracy in RV-C load measurement across the

concentration range (Table 4.3-4).

Table 4.3-4 Variation in calculated copy number yield (%) of transcripts 1-4 compared to the number of

probe mismatches

1Each RV-C transcript was tested at 7 di fferent concentrations ranging from 10

1 – 10

7 copies/ µL in RV Assay-1. The

ca lculated copy numbers (means of three independent experiments) for each transcript i s presented as a percentage of the perfectly matched RV-transcript 1.

Calculated copy number of transcripts 2-4 1

Probe mismatches

107 106 105 104 103 102 101

Transcripts

1 100% 100% 100% 100% 100% 100% 100%

2 95% 90% 76% 72% 90% 87% 28% 1

3 <1% <1% <1% <1% <1% <1% <1% 4

4 7% 10% 12% 5% 10% 3% 3% 3

Page 92: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

72

4.3.4 Clinical studies

An algorithm was developed to guide viral load determination for RV-C positive samples (Fig.

4.3-2). Using this algorithm RV-C positive samples (n=40) from children presenting with acute

wheeze were matched to the appropriate assay.

Figure 4.3-2 the algorithm for the determination of RV-C viral load in clinical samples

Sample

Picornavirus PCR

Species Identification by sequence analysis

RV-C matched to appropriate probe (in silico) sequence

Viral Load determined

Detected

RV-C

Page 93: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

73

In this group of patients, a total of 23 genotypes were identified with the most commonly

detected genotypes being C-16 (n=5), C-35 (n=3), C-42 (n=3), C-14 (n=3) and C-11 (n=3) (see

Appendix 10.1). In silico analysis demonstrated that assay-1 aligned completely with the target

region of 24/40 (16/23 genotypes) samples. Assays 2-4 were suitable for the remaining

genotypes (n=7) (see appendix 10.1). As can be clearly s seen in Figure 4.3-3, all 40 samples

had there RV-C load determined using one of the assays developed herein. Assay 1 provided

coverage for the majority of samples (27/40, 67.5%) . With assays 2-4 providing coverage for

the remaining samples (Fig.4.3-3). All samples met criteria for adequate sample collection and

nucleic acid extraction with GAPDH Cq values within the accepted range (see Appendix 10.1).

Page 94: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

74

Figure 4.3-3: Box plots of RV-C load in samples from young children presenting to the Emergency Department

with acute wheeze.

Page 95: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

75

4.4 Discussion

This study presents the development and validation of four qRT-PCR assays that, in

combination are able to accurately and reliably measure the viral load of circulating RV -C

genotypes.

Reports of an association between RV-C infection and severe respiratory disease have been

mixed as some studies have found an association (Bizzintino et al., 2011; Bochkov et al., 2011;

Camargo et al., 2012; Piralla et al., 2009) but others have not (Iwane et al., 2011; Linsuwanon

et al., 2009). Similar to other acute viral respiratory tract infections where a correlation exists

between viral load and disease severity (DeVincenzo et al., 2010; Li et al., 2010; Roussy et al.,

2014; To et al., 2010), it is suspected that RV-C load may also drive disease severity. Inaccurate

quantitative methods complicate the evaluation of viral factors that may contribute to disease

severity (Schibler et al., 2012). Previously published quantitative assays have tried to address

the genetic heterogeneity of RV-C by either using an intercalating dye in place of a specific

hydrolysis probe or adding degenerate bases in either the primer or probe sequence (Bochkov

et al., 2011; Granados et al., 2012). However, accuracy maybe impaired since these techniques

may lead to non-specific amplification and reduced amplification efficiencies of the assays

(Chemidlin Prevost-Boure et al., 2011; Schibler et al., 2012).

In this study, performance evaluation of each assay was conducted in accordance with the

MIQE guidelines (Bustin et al., 2009). A portion in the 5’UTR was chosen as a target for our

assays since previous work has demonstrated that 5’UTR sequence can be used for RV -C

genotypic assignment at almost identical accuracy to the VP4/VP2 and VP1 and with superior

clinical sensitivity (Lee et al., 2012). All assays demonstrated a broad dynamic range, high

sensitivity, efficiency and performance in RV-C viral load determination with both clinical

Page 96: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

76

samples and in vitro RNA transcripts. Viral load in our respiratory samples were within the 1x

103 and 1x1012 copies/ mL range which is in concordance with previous publications (Li et al.,

2010; Roussy et al., 2014; Schibler et al., 2012). All assays demonstrated high repeatability and

reproducibility with CV of below 8% and 15% respectively. All assays were nonreactive with a

range of other potential respiratory pathogens but cross-reactivity with other picornavirus

species requires sequencing of the 5’UTR to confirm RV -C identification prior to quantification.

This was also needed to determine the appropriate primer-probe combination.

To accurately measure viral load in clinical samples it is vital that primers and probes are

designed to match the target sequence. Indeed, previously published studies have shown that

the positioning of the mismatch is a crucial determinant of probe binding affinity (Benovoy,

Kwan, and Majewski, 2008; Letowski, Brousseau, and Masson, 2004) which may in turn impact

upon the accuracy of the calculated viral load. Mismatches throughout or near the middle of

the probe target region destabilize hybridization more than those near the ends (Letowski et

al., 2004). Another recent study demonstrated that multiple mismatches in the probe target

region may have a greater impact on accuracy than a single mismatch (Randhawa et al., 2011).

This is consistent with our findings which showed that at most dilutions a single probe

mismatch between assay probe and transcript material had minimal impact on accuracy but

when multiple mismatches were present there was a substantial effect on viral load

measurement across the dilution range. Together, these findings demonstrate the need for

multiple qRT-PCR assays to achieve accurate RV-C loads for the different genotypes. However,

we were able to show that this could be achieved with a small numbe r of assays, requiring

only four different probes to cover the 34 genotypes with known 5’UTR sequences. It is

anticipated that these four assays will cover more of the RV-C genotypes, but that awaits

further sequence data.

Page 97: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

77

Fortunately other studies have demonstrated that while a large proportion of RV-C genotypes

circulate simultaneously in various geographical regions worldwide they are dominated by C-1,

C-2, C-6, C-16, C-17, C-18 and C-43 genotypes (Lu et al., 2014; McIntyre et al., 2013). All of

these genotypes were quantified at equal efficiencies in this study, suggesting that our assays

can be used to accurately determine RV-C load of various genotypes from different

geographical regions as well as to properly investigate differences in pathogenesis between

RV-C genotypes. A limitation of the current method is it cannot reveal the presence of a mixed

infection and therefore may not be able to accurately quantify the viral load of each genotype

present.

In conclusion, this study describes a reliable and accurate PCR based method of quantifying

RV-C load in clinical samples containing a wide range of RV-C genotypes. These assays will

provide a reliable tool for investigating the role of RV-C in respiratory illness, and for

evaluating the effectiveness of future antiviral therapies.

Page 98: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

78

5. The Quantitative Detection of Respiratory Syncytial Virus

in Hospitalized Young South African Children

Page 99: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

79

5.1 Introduction

Acute lower respiratory tract infections (ALRIs) are a leading cause of morbidity and

mortality in infants worldwide. ALRIs add an extensive health and economic burden

especially in developing nations, where 99% of all ALRI-related deaths occur (Denny

Jr., 2001; Nair et al., 2013). Respiratory syncytial virus (RSV) is the predominant

etiological agent of acute lower respiratory tract infection in children under the age of

five years. In children under the age of five years, RSV contributes to approximately 33

million episodes per year (Nair et al., 2010). RSV is estimated to account for 85% of

bronchiolitis cases and 20% of childhood pneumonia cases (Nair et al., 2011b). The

peak age of RSV bronchiolitis is 1—2 months and it is the most common cause of

hospitalization during infancy (Kim, Lee, and Lee, 2000). It is well established that

virtually all children have been exposed to RSV by the age of three years, with the

majority of children being infected in their first year of life. Furthermore between the

ages of one and 12 years-old RSV is associated with more deaths than influenza

(Fleming, Pannell, and Cross, 2005). Several risk factors have been associated with

severe RSV disease including malnutrition (Paynter et al., 2014), immunodeficiency

(Englund, Anderson, and Rhame, 1991), premature birth, and congenital heart and

lung disease (Zhang et al., 2014). Several lines of evidence suggest that severe RSV

bronchiolitis during infancy (Henderson et al., 2005; Schauer et al., 2002; Sigurs et al.,

2010; Wennergren and Kristjánsson, 2001) and early childhood (Sigurs et al., 2000)

may increase the risk of developing asthma.

Page 100: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

80

RSV is classified into antigenic subtypes (RSV-A and RSV-B) based on amino acid

sequence composition of the attachment G glycoprotein (Mufson et al., 1985). Both

antigenic subtypes frequently circulate simultaneously but the proportion of infection

due to each differs each season (Akerlind and Norrby, 1986). Although epidemiological

studies have shown that RSV-A dominant seasons are generally associated with more

reports of severe illness (McConnochie et al., 1990) and suggest that RSV subtype may

influence pathogenicity, there are conflicting reports about which subtype is more

pathogenic (Hirsh et al., 2014; Xiang et al., 2013).

The presence of co-infecting pathogens has been suggested as a possible

pathogenicity determinant (Franz et al., 2010). Associations between higher viral loads

and poorer clinical outcomes in patients with RSV and other respiratory infections

have been reported (Bagga et al., 2013; Drews et al., 1997; Foulongne et al., 2006;

Franz et al., 2010; Li et al., 2010; Roussy et al., 2014), but this remains contentious

(Martin et al., 2012; Wang et al., 2014).

Several studies have described the epidemiology of RSV in South Africa (Cohen et al.,

2015a; Cohen et al., 2015c; Cohen et al., 2015d; Pretorius et al., 2013; Tempia et al.,

2015; van Niekerk and Venter, 2011), but none have focused on the importance of

viral load as a potential predictor of clinical outcome among hospitalized patients. We

undertook this study to investigate the relationship between RSV load and clinical

disease among young children (<2 years) in a country with a high HIV prevalence, to

examine the role of RSV subtype and viral co-infections in the pathogenesis of ALRI in

infants.

Page 101: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

81

5.2 Samples

Nasopharyngeal aspirates (NPAs) were collected from 105 ALRI cases and 53 controls

from Pretoria, South Africa between July 2011 and November 2012. Children 0-2 years

of age hospitalized with an ALRI at the Steve Biko Academic Hospital or Tshwane

District Hospital in Pretoria were enrolled as cases. A diagnosis of pneumonia

(respiratory distress and either chest X-ray changes (e.g. consolidation or effusion), or

auscultatory findings (e.g. crepitations or bronchial breathing) or bronchiolitis

(respiratory distress and at least one of the following; wheeze, chest X-ray changes

(e.g. hyperinflation) or Hoovers’s sign ( inward movement of the lower rib cage during

inspiration) was determined by the clinician in charge. Fifty-three age-matched

children presenting to the same hospitals with a non-respiratory illness over the same

period were enrolled as controls (NRD controls). The ARCHITECT HIV Ag/Ab Combo

assay (Abbott Diagnostics) was used to screen patients for the presence of human

immunodeficiency virus (HIV). The Amplicor HIV-1 DNA PCR assay (Roche Diagnostics,

Branchburg, NJ) was used to confirm HIV status. At enrolment, baseline characteristics

and clinical symptoms were collected by the physician using a detailed questionnaire.

The study protocol was approved by the University of Western Australia Human Ethics

Committee and the University of Pretoria Ethics Committee. Written informed consent

from parent/guardian was obtained prior to participation.

All RSV positive samples (RSV A/RSV B) by multiplex PCR (Chidlow et al., 2009) were

confirmed and viral load determined using an in-house monoplex RT-qPCR protocol

targeting the nucleoprotein gene as described in chapter 3. RT-qPCR analysis was

performed on the Rotor Gene 6000 cycler (QIAGEN, Australia). All experiments were

Page 102: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

82

performed in triplicate including positive controls and non-template controls.

Detection of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNA (Gueudin et

al., 2003) was utilized to ensure adequate specimen collection, RNA extraction and

detection of PCR inhibitors. Good quality samples were considered to be those with

GAPDH CT values below 31.5.

5.3 Results

5.3.1 Baseline characteristics

A nasopharyngeal aspirate (NPA) was obtained from 158 children (105 cases and 53

NRD controls). Of the 105 cases, 57 (54%) children were diagnosed with pneumonia

and 48 (46%) children were diagnosed with bronchiolitis (Table 5.3-1).

Page 103: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

83

Table 5.3-1 Demographic and clinical details of study participants

ALRI cases (n=105) NRD Controls (n=53) P

RSV-positive (n=27)

RSV-negative (n=78)

RSV-positive (n=9)

RSV-negative (n=44)

Age (wks.) mean ± SD 21 ± 14 32 ± 28 33 ± 26 39 ± 31 0.03#

Gender (%) NS

Male 18 (66.7) 53 (67.9) 5 (55.6) 25 (56.8)

Weight (kg) mean± SD 6.6 ± 2.4 6.4 ± 2.8 6.0 ± 3.1 6.6 ± 2.8 NS¶

Race (%) NS¶

Black 15 (55.6) 68 (87.2) 8 (88.9) 36 (81.8)

White 10 (37) 5 (6.4) 1(11.1) 5 (11.4)

Other 2 (7.4) 5 (6.4) 0 3 (6.8)

HIV status (%) 0.018¶

Positive 0 15^ (19.2) 1 (11.1) 1 (2.3)

Negative 25 (92.6) 60 (76.9) 8 (88.9) 39 (88.6)

Unknown 2 (7.4) 3 (3.8) 0 4 (9.1)

Allergies (%) NS

Yes 0 (0) 3 (2.9) 0 2 (4.5)

No 27 (100) 75 (96.2) 9 (100) 41 (93.2)

Unsure 0 (0) 0 (0) 0 1 (2.3)

Clinical Diagnosis (%) <0.01¶

Pneumonia 7 (25.9) 50 (64) - -

Bronchiolitis 20 (74.1) 28 (36) - -

# Comparison of ages between RSV positive ALRI cases, RSV negative ALRI cases and NRD controls (ANOVA) ¶ Comparisons of proportions between RSV positive ALRI cases vs RSV negative ALRI cases and NRD controls (Fisher’s exact test) ^ No respiratory virus was detected in 67% (10/15) of HIV infected children NS: non-significant, SD - standard deviation.

Page 104: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

84

A respiratory virus was detected in 88/105 (84%) ALRI patients compared with 37/53

(70%) NRD controls (p=0.041). Rhinoviruses (51/105, 49%), hAdV (33/105, 31%) and

RSV (27/105, 27%) were the most frequently detected viral pathogens in children with

an ALRI, but were also found in 40%, 28% and 17% of NRD control samples

respectively (Fig. 5.3-1). None of the differences for the individual viruses reached

statistical significance.

Figure 5.3-1: Viruses detected in NPAs collected from ALRI cases and NRD controls. RV (RV-A, RV-B, RV-

C), HCoV (OC43, 229E, HKU-1, NL63), HPIV (PIV I-IV), IFV (A/H1N1, A/H3N2, B and C)

5.3.2 RSV infection and clinical outcome

In patients with ALRI, age was significantly associated with RSV disease (Table 5.3-1),

45% of RSV associated ALRI cases were infected within their first two months of life

(Fig.5.3-2). In addition, children in their first year of life were at a significantly

increased risk of RSV associated ALRI (OR: 9.6, 95%CI: 1.2, 75.7, p=0.011).

Page 105: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

85

Twenty of 27 (74%) RSV-positive ALRI cases were classified as bronchiolitis and 7/27

(26%) were classified as pneumonia. RSV was significantly more common in

bronchiolitis cases than pneumonia cases (p<0.001). In non-RSV associated ALRI cases,

50/78 (64%) patients were classified as clinical pneumonia and 28/78 (36%) were

classified as bronchiolitis (Table 5.3-1).

Analysis by antigenic subtype found that RSV-A (18/105, 17%) was more common than

RSV-B (9/105, 9%) in ALRI cases, but RSV-A (8/53, 9%) was also found more often than

RSV-B (1/53, 2%) in NRD controls. Therefore, the proportion of RSV -B detected in

patients with an ALRI compared to that in NRD controls suggest a tendency for RSV-B

to cause more disease than RSV-A, although the association did not reach statistical

significance. However both RSV-A (n=18, 72% vs 28%, p=0.01) and RSV-B (n=9, 71% vs

29%, p=0.04) were significantly more common in bronchiolitis cases than in

pneumonia cases.

Page 106: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

86

Figure 5.3-2: The distribution of RSV positive and RSV-negative ALRI cases by age. RSV disease was more

prevalent in children within their first year of life. Peak hospitalization rate was observed in the 0-2 month age group.

Page 107: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

87

5.3.3 RSV disease and HIV infection

Interestingly, RSV was significantly less common among HIV infected ALRI patients

compared to HIV uninfected ALRI patients (Table 5.3-1). Clinical pneumonia was the

more likely diagnosis in HIV infected ALRI patients [14/15, (93%) vs 1/15, (7%);

p=0.0013]. As demonstrated in fig. 5.3-3, a respiratory virus was encountered in 33%

(5/15) of cases with parainfluenza viruses being the predominant viral pathogen in this

group of patients.

Figure 5.3-3: Distribution of respiratory viruses detected in HIV infected ALRI patients

hCoV 7% hAdV

7%

hPIV 22%

virus not detected

64%

Page 108: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

88

5.3.4 RSV load

To obtain a more accurate estimate of the association between RSV infection and

clinical outcome, RSV load studies were performed. Median RSV load in ALRI patients

(8.2 log10 RSV RNA copies/mL [IQR: 6.3-9.1] n=27) was 1.9 log10 copies higher than in

NRD controls (6.3 log10 RSV RNA copies/mL [IQR: 4.6-7.4] n=9) p=0.031 (figure 5.3-4).

Figure 5.3-4: A box plot comparing RSV load between ALRI cases and NRD controls

This analysis revealed no significant difference in median viral load between ALRI

patients infected with RSV-B compared to those infected with RSV-A (8.2 log10 RSV-B

copies/mL [IQR: 6.4-9.3] vs 8.2 log 10 RSV-A copies/ mL, IQR [6.9-8.6]) (Fig 5.3-5).

Overall RSV load was not associated with a type of ALRI (pneumonia or bronchiolitis),

with the median RSV load being 7.3 log10 copies/mL (IQR: 5.6 – 8.2) in children

diagnosed with pneumonia compared to 8.6 log10 copies/mL (IQR: 6.7 – 9.4) in

children diagnosed with bronchiolitis (Fig.5.3-5).

Page 109: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

89

Figure 5.3-5 A box plot comparing RSV load by subtype and clinical diagnosis

Finally, we sought to understand the contribution of viral co-infection to patient

outcome. Viral co-infections were detected in 38/105 (36%) ALRI patient samples and

in 18/53 (34%) NRD control samples with RSV, hAdV or RV being the most frequently

detected viruses in both sample groups. Our analysis revealed a significant association

between viral coinfection and RSV detection in NRD controls (cases: 12/27 (44%) vs

NRD controls: 8/9 (89%) p=0.026). RSV load did not significantly differ among ALRI

patients when RSV was detected alone or in the presence of other viruses (Fig.5.3-6).

Page 110: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

90

Figure 5.3-6: A box plot comparing RSV loads in ALRI cases with a viral co-infection [n=12; RSV with either hAdV

(75%, n=9), hCoV (17%, n=2) or RV (8%, n=1)] versus sole RSV infection (n=15).

Page 111: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

91

5.4 Discussion

We conducted a study to assess the impact of RSV load on clinical outcomes in a

previously unexamined population of South African children, and where there is a high

prevalence of HIV infection. As in previous studies in other populations we found that

higher RSV loads were associated with clinical illness (El Saleeby et al., 2011; Fodha et

al., 2007; Houben et al., 2010; Utokaparch et al., 2011). This is also consistent with the

suggestion that clinical illness due to RSV infection is a direct virus-mediated

phenomenon (Bagga et al., 2013; DeVincenzo et al., 2010).

Interestingly, we found that RSV and other viral pathogens were found less commonly

in HIV infected children than in non-HIV infected children, and when a viral pathogen

was detected, nearly all of the children had pneumonia rather than bronchiolitis. This

was surprising given that viral pathogens especially RSV contribute substantially to

ALRI in children under five (Nair et al., 2010). However, as most of the HIV-infected

children had pneumonia, the low RSV rates may reflect the high number of alternative

causes in these children. Aetiological studies of pneumonia in HIV infected children

reveal that non-viral pathogens in particular Streptococcus pneumoniae,

Staphylococcus aureus, gram-negative bacteria and Pneumocystis jirovecii

predominantly contribute to hospitalization and death from pneumonia (B-Lajoie et

al., 2016; Gray and Zar, 2010; Lanaspa et al., 2015), presumably related to

immunosuppression (Madhi et al., 2000; Marais et al., 2006; Zash et al., 2016). It is not

clear why RSV-associated ALRI was so uncommon in these children. However we

acknowledge that current findings are limited by sample size and therefore further

investigations are needed to clarify and determine clinical implications.

Page 112: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

92

Our analysis revealed that the prevalence of RSV is relatively high in asymptomatic

South African children. Studies in other geographical settings report much lower RSV

prevalence rates (Jartti et al., 2008; Self et al., 2015). However, similar asymptomatic

detection rates (17% and 42%) have been reported in Kenyan children (Matu et al.,

2014; Munywoki et al., 2015) possibly indicating that asymptomatic RSV detection may

be important in the epidemiology of RSV, at least in some populations. We also found

that in asymptomatic individuals, RSV A was more commonly detected and at

comparatively higher titres than RSV-B. RSV-A has been found to shed at higher titres

and for a lengthier period than RSV-B (Takeyama et al., 2015b). Collectively, this may

suggest that the RSV subtype circulating in a population and its association with illness

may be determined by both host characteristics and viral titre.

Differences in disease severity between RSV subtypes are controversial; some studies

have reported that RSV A is more virulent than RSV-B (Hornsleth et al., 1998; Jafri et

al., 2013; Papadopoulos et al., 2004), whilst other studies have reported contrary

results (Fodha et al., 2007; Oliveira et al., 2008; Xiang et al., 2013). Our analysis in

hospitalized cases revealed no significant difference in viral load between the

antigenic subtypes.

The role of viral co-infections in disease outcome remains a controversial topic,

especially in population groups where viral co-infections are common. Some studies

have reported that viral co-infections result in poorer clinical outcome compared to

single respiratory virus infections (Drews et al., 1997; Foulongne et al., 2006; Franz et

al., 2010; Greensill et al., 2003), but this has not been supported in other publications

(Martin et al., 2012; Wang et al., 2014). Since we found that RSV load was related to

Page 113: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

93

disease, we expected that if viral co-infections contribute to disease causation, then

the RSV load in co-infected patients (ALRI cases) would be lower than in RSV mono-

infected ALRI cases. This was not the case and RSV load was similar whether RSV was

detected alone or in the presence of other viruses, suggesting that the co-infecting

virus did not contribute to disease. HAdVs were the most common co-infecting virus,

being detected in 75% of the RSV co-infections. HAdVs were frequently detected in

NPA specimens from both our ALRI cases and NRD controls. Previous studies have

found that hAdV pathogenicity may be species-specific (Chidlow et al., 2012) and that

hAdV DNA persists for several weeks following clinical or subclinical infection (Demian

et al., 2014; Robinson et al., 2011). It is therefore possible that any effect of mixed

infections compared to monoinfection may have been masked by a high incidence of

detection of non-pathogenic hAdV species. Overall, additional investigations with

measures of the immunological response in combination with disease severity

indicators will be required to extend our understanding of this phenomenon.

Our study was limited in several ways. First, the small sample size limited our ability to

reach statistical significance for many findings. Second, we only had clinical diagnoses

without detailed data on disease severity. Third, no follow up of the NRD controls was

conducted after enrolment thus we do not know whether some individuals developed

ALRI in the days after sample collection. Fourth, we were unable to consider the role

of bacterial-viral interactions which may have a pertinent role in disease outcome.

Fifth, RSV data was collected over a single season and hence may not be

representative of long term transmission dynamics of RSV in this region. Lastly, studies

of hospitalized children may not reflect wider community impact of this virus.

Page 114: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

94

In conclusion, our study shows that RSV is a significant contributor to infant morbidity

in young South African children, and viral load maybe an important predictor of

disease. Although a number of observations in this chapter are confirmatory, this

confirmation is important. The study was conducted in South Africa, a region

previously with little data on the viral kinetics of RSV disease in young children.

Furthermore, it is in a part of the world widely recognised to have a high HIV

prevalence among young children. The findings of this chapter clearly demonstrate

that the causative agents of acute lower respiratory tract infection in young children

infected with HIV are diverse and not isolated to the commonly detected respiratory

viruses in the uninfected HIV population. In addition, our study provides valuable

region and cultural specific data which is pertinent to accurately defining the global

burden of childhood RSV disease. Given the small samples size and the lack of precise

clinical and laboratory data, future study is needed to determine confirm the observed

regional differences and to further describe the importance of regional factors (i.e.

HIV infection, nutritional status, asthma prevalence and bacterial co-infection) on RSV

disease severity.

Page 115: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

95

6 Determinants of acute asthma exacerbation severity following

RV-C infection

Page 116: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

96

6.1 Introduction

Acute Asthma Exacerbation (AAE) is a leading cause of severe morbidity and hospital ization in

children with airway disease (Murray et al., 2006). AAE is characterized by airway

inflammation, airflow obstruction and airway hyper-responsiveness that results in the rapid

decline in lung function. A substantial recruitment of inflammatory cells to the respiratory

airways is the fundamental basis of airway inflammation. These inflammatory cells and the

products they secrete contribute to airway obstruction in part through plugging of the airways

(Fahy, 2009; Zhao et al., 2002). Respiratory viral infection is the most frequent trigger of AAE

(Johnston et al., 2005a). Clinical and epidemiological findings have shown that viral induced

exacerbations associated with as many as 80% of cases in children (Johnston et al., 2005a).

Other triggers that may result in an AAE include allergens, environmental pollutants, and

occupational irritants. The type of trigger uniquely shapes the pattern of airway infl ammation

during an AAE and this may have implication on clinical management (Berry et al., 2007; Holt

et al., 2010; Leung et al., 2013; Oommen, McNally, and Grigg, 2003; Zhao et al., 2002).

The relationship between respiratory viral infection and exacerbation of asthma disease has

been suspected for a long time, though maybe underestimated. Sensitive molecular diagnostic

methods have substantially improved the detection of respiratory viruses in AAE cases and

consequently reinforced the association between respiratory viral infections and AAE. Viruses

are detected in up to 85% of AAE, the most common type being rhinovirus (RV). RV as a sole

pathogen is detected in approximately 60% of AAE (Denlinger et al., 2011; Jackson et al.,

2008b; Khetsuriani et al., 2008; Miller et al., 2012). In fact, rhinovirus is the most common

virus in near fatal asthma exacerbations. The pathogenesis of AAE triggered by viral infections

is yet to be elucidated. Furthermore, a causal relationship is difficult to establish, since not

Page 117: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

97

every asthmatic patient with a viral respiratory tract infection will experience AAE. Further

complicating this are studies that have demonstrated asymptomatic detection of the same

viral types in healthy children (Camargo et al., 2012; Jartti et al., 2008).

Previous studies report that RV-C infected asthmatic patients were more likely to be

hospitalized than patients infected with other RV species (Bizzintino et al., 2011a; Linsuwanon

et al., 2009). The lack of a reliable method to quantify the many genotypes of RV -C has

precluded investigations into the contribution of RV-C on severity of disease. However we

have previously developed (see chapter 4) a comprehensive RV -C quantification assay to

facilitate investigation in to whether severity of AAE is driven by the amount of virus in the

airways.

We hypothesized that RV-C load significantly differs between children with a severe AAE and

non-severe AAE and that the amount RV-C in the airways is driving an inflammatory response

that determines the severity of disease.

6.2 Samples

Participants (n=121) up to the age of 10 were recruited from Princess Margaret Hospital

(Perth, Australia). Of the 121 study participants, ninety-nine presented with an acute asthma

exacerbation of varying severity. Twenty-two otherwise healthy children from the community

matched for age and sex attending clinic formed the healthy control group. All AAE-related ED

encounters during the study period were classified as "mild", "moderate" or "severe",

according clinical practice guidelines (Royal Children's Hospital, 2015) that include vital and

readily available signs and symptoms, including pulse rate, presence of respiratory wheezes,

rales, oxygen saturation, and the use of accessory muscles, measured upon arrival to the ED.

Page 118: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

98

All medical records of ED and hospital admissions were reviewed for treatment and outcome s.

Blood was collected from each participant for full blood count analysis by clinical staff.

Nucleic acid was extracted from 200µL of clinical samples using automated extraction method

described in chapter 2. Nucleic acid was tested using a multiplex PCR for the common

respiratory pathogens including respiratory syncytial virus, human metapneumovirus, human

parainfluenza viruses, influenza viruses and adenovirus (Chidlow et al., 2009). Rhinovirus

screening and genotyping was performed at the Telethon Kids Institute using previously

published primers (Lee et al., 2012b). The algorithm described in chapter 4 for RV-C viral load

determination was applied to any sample that was picornavirus positive. Viral load

determination was performed on the Rotor Gene 6000 cycler (QIAGEN, Australia). All

experiments were performed in triplicate including positive controls and non-template

controls. Detection of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNA (Gueudin

et al., 2003) was utilized to ensure adequate specimen collection, RNA extraction and

detection of PCR inhibitors. Good quality samples were considered to be those with GAPDH CT

values below 31.5.

Page 119: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

99

6.3 Results

6.3.1 Study participants characteristics

A total of 121 children were enrolled; 99 presented to hospital with an acute asthma

exacerbation (AAE) (cases) and 22 were non-respiratory disease community controls (NRD-

controls). We encountered no statistically significant difference between cases and controls

for age and sex (Table 6.3-1). Furthermore no significant differences in age, sex, atopy status

and history of wheeze were encountered when cases were stratified by RV -C detection. As

can be seen from Table 1, children presenting with acute asthma exacerbation following RV-C

infection were generally atopic (73%). The majority of these children were classified with a

moderate (40%) or severe (48%) asthma exacerbation. Only 4% of cases were admitted to the

ICU, the remaining cases were either discharged at ED (64%) or admitted into an in-patient

ward (28%).

Table 6.3-1 Baseline characteristics of children with acute asthma exacerbation (AAE) and controls

RV-C positive AAE (n=29)

RV-C negative AAE (n=70)

Controls without asthma (n=22)

p-value

Mean age ± SD, yr. 5.1 ± 3.3 4.86 ± 3.7 5 ± 4.2 NS

Male % 56 57% 52 NS

Previous wheezing

episodes (%)

36 31 - NS

Atopic % 73 60 45 NS Definition of abbreviations: SD= standard deviation, yr. = year, atopy determined by skin prick test, NS: not significant. Mann-Whitney U testwas used to test comparisons of continuous variables ¶ Fisher’s exact test was used to test comparisons of categorical variables.

6.3.2 Virus Detection

We examined NPAs of children with acute asthma exacerbation (AAE) symptoms and non-

respiratory disease community controls for the presence of viral pathogens and found that one

Page 120: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

100

or more viral types were detected in 75% (74/ 99) of samples from children with an AAE and in

50% (11/22) NRD control samples (p=0.03). As can be seen in figure 6.3-1, RVs (47/74, 63%),

parainfluenza virus (HPIV) (11/74, 15%) and respiratory syncytial virus (RSV) (9/74, 12%) were

the most prevalent viruses in patients with AAE (Fig.6.3-1).

Figure 6.3-1 A bar graph comparing the frequency (%) of viruses detected in cases and controls. Rhinovirus-C (RV-C), Rhinovirus-A (RV-A), parainfluenza virus (PIV), respiratory syncytial virus (RSV), human adenovirus (hAdV), Influenza viruses (IFV), human corona virus (hCoV) Rhinovirus-B (RV-B) and human metapneumovirus (hMPV)

Interestingly, RV-C was found to be the most prevalent of the RV species when cases were

stratified by species type. Further, this analysis revealed that study participants infected with

RV-C were 2.6 times more likely to be AAE patients than NRD controls (Table 6.3-2). RSV and

HPIV were the other viruses that may have increased the risk of asthma exacerbation following

infection.

0

5

10

15

20

25

30

35

RV-C RV-A PIV RSV hAdV IFV hCoV RV-B hMPV

Fre

qu

en

cy o

f d

ete

ctio

ns

(%)

Virus

Cases

Controls

Page 121: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

101

Table 6.3-2: A statistical summary of the risk of being diagnosed with acute asthma exacerbation follwoing

respiratory virus detection

Controls (n=22)

Cases (n=99)

OR (95%CI) p-value

RV-C 3 29 2.6 (0.9-9.6) 0.07

RV-A 4 17 0.9 (0.3-3.1) 0.5

RV-B 0 1 N/A N/A

ADV 0 6 N/A N/A

Influenza 2 4 0.4 (0.07-2.5) 0.3

RSV 1 9 2.1 (0.2-17.5) 0.4

HMPV 2 1 0.2 (0.13-3.6) 0.3

HCOV 3 3 0.2 (0.037-1.1) 0.07

HPIV 2 11 1.3 (0.3-6.1) 0.9

This study also aimed to understand if viral coinfection may be a risk factor of asthma

exacerbation. Overall, the detection rate for viral coinfection was not significantly different

between in NRD controls (5/22, 23%) and AAE cases (14/99, 14%). Further, of the 29 RV-C

infected AAE case, six were identified as coinfections but the combination of viruses detected

did not increase the risk of disease or severity (p>0.1).

6.3.3 RV-C load

In order to gain further understanding on the contribution of RV-C infection on asthma

exacerbation, viral load was determined in 21/29 RV-C infected AAE cases and 2/3 NRD

controls. As clearly demonstrated in figure 6.3-2, viral load was 2.6 log10 copies/mL higher in

children presenting with AAE compared to NRD controls (6.6 log10 copies/mL; IQR: 5.1-8.2 vs

4.0 log10 copies/mL; p=0.0235).

Page 122: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

102

Fig 6.3-2: Box plot summarising RV-C load in children presenting to emergency department with an acute asthma exacerbation (cases) and otherwise healthy individuals with a non-respiratory disease (controls). Median RV-C load of AAE cases was 2.6 log10 copies/mL higher than that of the non-respiratory disease control group .

In order to investigate the contribution of RV-C load to disease severity, we stratified AAE

cases into mild, moderate and severe groups based on the Royal Children’s Hospital clinical

guidelines (Royal Children's Hospital, 2015). Because of inadequate number of cases in the

mild group, we categorised mild and moderate cases as non- severe. Using this stratification

system we did not observe any significant difference in RV-C load between the severe group

compared to the non-severe group (6.6 log10 copies/mL; IQR: 4.7-8.3 vs 6.5 log10 copies/mL;

IQR: 5.2-8.2, p=0.98) figure 6.3-3).

Page 123: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

103

Figure 6.3-3: A boxplot of RV-C loads for cases(n=21) stratified by disease severity and controls (n=2). Groups were compared using the Mann-Whitney U test,. median viral load between the two severity groups did not

differ significantly.

Page 124: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

104

6.3.4 Surrogate markers of inflammation

Recruitment of inflammatory cells to the airways is central to the cascade of events that result

in AAE. Neutrophil counts and eosinophil counts have previously been shown to be important

markers of illness severity in AAE (Wenzel et al., 1999). Therefore we analysed levels of these

factors to understand their role in illness severity. Our analysis revealed that peripheral blood

neutrophil counts (median: 83% IQR: 67-92% vs 62% IQR: 59-75%, p=0.049) but not eosinophil

counts (median: 0.1% IQR: 0-4% vs 1% IQR: 0.04-2%, P>0.1) were associated with severe

disease (Figure 6.3-4A,B). An elevated serum IgE was observed in patients with severe AAE

compared to patients with non-severe AAE (median: 35 kU/L IQR:5-136 kU/L vs median: 16

kU/L IQR: 3-76 kU/L) but this observation did not reach statistical significance (Figure 6.3-4C).

Despite the association of high neutrophil count and severe AAE following RV -C infection, no

corresponding statistically significant correlation was observed between RV-C load and

neutrophil count.

Page 125: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

105

A

*

*

*

Figure 6.3-4 Surrogate markers of asthma exacerbation in acute samples from patients infected with RV-C stratified by illness severity. A) Absolute neutrophil peripheral blood counts B) absolute eosinophils peripheral blood counts C) Total serum IgE levels counts. Mann- Whitney U or Kruksall-Wallis test were used for testing on the apporpriate group of

subjects. All data are expressed as box and whisker plots. *:p=0.05 (severe vs non-severe) ** p<0.001 (all AAE cases vs NRD controls)

Page 126: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

106

Figure 6.3-4 B) A comparison of absolute eosinophils peripheral blood counts patients with severe AAE, non-

severe AAE and control group.

B

Page 127: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

107

Figure 6.3-4 C) A comparison of total serum IgE levels between severe patients and non- severe patients

C

Page 128: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

108

6.3.5 Performance of RV-C load, neutrophils and eosinophils in predicting

the severity AAE

The performance characteristics of RV-C load, absolute neutrophils counts, eosinophil counts

and serum IgE levels as single biomarkers of severe AAE were analysed. The ROC curves of the

four biomarkers are shown in figure 5a and 5b. Area under the curve (AUCs) for RV-C load,

absolute neutrophil count, eosinophil count and serum IgE levels were 0.58 (95%CI; 0.243-

0.912, p=NS), 0.92 (95%CI; 0.81-1.0, p= 0.03), 0.51 (95% CI; 0.1-0.9, p=NS), 0.53 (95%CI; 0.2-

0.88, p=NS) respectively. Neutrophil counts alone were an excellent predictor of severe AAE

(Figure 6.3-5a). Importantly, RV-C load, absolute eosinophil count and serum IgE levels were

not added to the prediction model because of their poor performance as single biomarkers

(Figure 6.3-5b).

Figure 6.3-5 Receiver operator characteristic curves (ROC) of markers of AAE severity. A) Absolute neutrophil count and b) RV-C load, eosinophil count and serum IgE.

Page 129: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

109

6.4 Discussion

This study revealed that RV is the most common viral agent detected in hospitali sed patients

with an AAE and that RV-C appears to be the predominant of the three species. Furthermore,

the RV-C load in the airways of asthmatic children during an acute exacerbation does not

appear to be associated with illness severity and is a poor predictor of severe AAE. However, a

host response that includes a predominantly neutrophil inflammatory pattern and elevated

levels of IgE was associated with severity of illness.

Several previous studies report viral detection rates of between 62% and 85% in exacerbating

asthmatic children (Heymann et al., 2004; Johnston et al., 2005a; Leung et al., 2013; Rakes et

al., 1999) as opposed to detection rates of 3-12% when asthmatic children were asymptomatic

(Message and Johnston, 2002). Our detection rate of 75% percent falls well within the range of

those studies and suggests that acute respiratory viral infections contribute to the initiation of

AAE. Our analysis also revealed that RV are the predominant viruses detected in children with

AAE thus corroborating other studies that report RV as the major viral trigger of AAE (Johnston

et al., 2005a; Lee et al., 2012a; Monto, 2002).

RV-C was the major RV species in the AAE cases indicating that RV-C strains may have a greater

propensity to trigger AAE than the other two species. Our finding is in concordance with

studies from Australia (Bizzintino et al., 2011c) and Costa Rica (Soto-Quiros et al., 2012) but in

contrast to studies in the United States that suggest RV-A maybe an equally important trigger

of asthma exacerbation (Kennedy et al., 2014; Khetsuriani et al., 2008). Indeed this is an area

in need of further research.

This study is one of the first observational studies to investigate the role of RV -C load in AAE

severity in a paediatric population using a reliable RV-C load quantification technique. Previous

Page 130: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

110

studies attempting to address this issue have generalised their findings based on RV -A studies

(Kennedy et al., 2014). However, this approach may not provide an accurate account of the

contribution of RV-C to disease severity. Using the RV-C quantification protocol developed in

chapter four, we found that viral load was not significantly different between severe and non -

severe AAE groups which points towards a limited pathogenic role for the virus in symptom

severity. This finding is in support of previous reports that demonstrate that RV mediated

cytotoxicity does not contribute to illness severity (Kennedy et al., 2014). Interestingly, our

finding is in direct contrast to what we encounter in hospitalized paediatric patients with RSV

bronchiolitis, a disease with similar clinical features to AAE but in which viral load appears to

be the primary driver of disease severity (chapter 4) (Bagga et al., 2013; DeVincenzo et al.,

2010; El Saleeby et al., 2011). Taken together, one may postulate that that the mere presence

of actively replicating RV may be enough to initiate the cascade of inflammatory events. It also

suggests RV-C replication kinetics may not drive asthma symptom severity in a similar manner

to RSV in paediatric bronchiolitis cases.

Serum IgE is a surrogate marker for the activation of allergic inflammation, increased level of

this marker is a characteristic feature in severe AAE (D’Amato et al., 2014; Holt et al., 2010).

Recent studies demonstrate that upon viral infection IgE cross linking on plasmacytoid

dendritic cells from patients with asthma results in an impaired antiviral response (Durrani et

al., 2012; Gill et al., 2010). Elevated levels of IgE have been shown to enhance the expression

of a T-helper 2 (Th2) polarized response and in turn strongly down regulate the expression of

T-helper 1 (Th1) inflammatory response in the asthmatic (Baraldo et al., 2012). This counter-

regulation has been hypothesised to be an important determinant of severe AAE (Durrani et

al., 2012; Gill et al., 2010; Pritchard et al., 2012). Our analysis revealed a trend that suggested

elevated serum IgE levels in RV-C infected patients with severe AAE and potentially points to

Page 131: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

111

the existence of a synergistic pathomechanism between viral infection and allergen to

promote severe disease (Murray et al., 2006; Soto-Quiros et al., 2012) however, a lack of

numbers may have prevented us from reaching a statistically meaningful conclusion.

Although an eosinophilic inflammation is a characteristic feature in many asthma phenotypes

it is not associated with viral induced asthma exacerbation. Moreover, RV induced AAE is

strongly associated with a neutrophilic inflammatory pattern in human and animal studies

(Berry et al., 2007; Clarke et al., 2014; Denlinger et al., 2011; Fahy et al., 1995). Thus our

finding of elevated neutrophil counts but not eosinophil counts in children presenting to ED

with AAE fits well with the current understanding of the contribution of inflammatory cells to

virus induced allergic airway disease. This study also revealed an association between

peripheral blood neutrophil count and severe AAE. These findings are in accordance with a

growing body of research demonstrating associations between viral induced severe AAE and

an abundance of neutrophils in peripheral blood and respiratory airways. A previous study

found that several participants who reported upper respiratory tract infection as a trigger for

their AAE at presentation to the emergency department had substantial neutrophilia in their

sputum (25). Another study reported greater than fivefold more neutrophils in the sputum of

patients with RV associated AAE compared to those with RV-associated URTI (Denlinger et al.,

2011) . This relationship was also confirmed in a study where reduction in lung function

correlated strongly with elevated neutrophil counts (Message et al., 2008).

Although we did not investigate pathogenesis in our study, potential mechanisms deserve

comment. Prolonged neutrophil activation in the airways is associated with severe asthma

(Fahy, 2009). Naturally, aged neutrophils spontaneously undergo apoptosis are recognised and

phagocytised by macrophages leading to the resolution of inflammation. However, previous

studies have reported that impaired clearance of aged neutrophils by macrophages may drive

Page 132: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

112

a sustained inflammatory environment (Fitzpatrick et al., 2008; Huynh et al., 2005). Moreover,

impaired clearance of these aging neutrophils leads to necrosis with the subsequent loss of

membrane integrity resulting in the release of cytotoxic compounds such as reactive oxygen

species and granule enzymes that contributes to local tissue damage and ultimately airway

obstruction (Cortjens et al., 2016; Li et al., 2009; Obermayer et al., 2014). Another potential

mechanism that may account for neutrophil dominated severe asthma relates to the impaired

production of IL-10 in the lung microenvironment of asthmatics (Kearley et al., 2005). IL-10 is

an anti-inflammatory mediator that effectively suppresses the production of pro-inflammatory

inflammatory mediators and enzymes in activated macrophages, T cells, and

polymorphonuclear cells (Moore et al., 1993). A previous study demonstrated that patients

with severe asthma had significantly lower IL-10 levels compared to controls and patients with

mild disease (Lim et al., 1998). Another study that evaluated the effect of IL-10 producing

interstitial macrophages on allergen-induced asthma in a mouse model demonstrated that

allergen challenged IL-10 deficient mice exhibited severe, neutrophil dominant, lung pathology

compared to wild type mice. Furthermore, transplantation of wild type macrophages reduced

biomarkers of neutrophilic inflammation (Kawano et al., 2016). Clearly, neutrophils play an

important role in the severity of viral induced asthma exacerbation and further study should

focus on the therapeutic implications of targeting these cells.

Our analysis also revealed that absolute neutrophil count (ANC) provided excellent

discrimination of severe AAE from non-severe AAE following RV-C infection. This finding is of

interest given that almost 60% of neutrophilic asthma patients are administered high dose

inhaled corticosteroid therapy or are classified with steroid refractory asthma(Bel et al., 2011).

Addition of absolute neutrophil count to current or future predictive models for severe asthma

Page 133: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

113

exacerbation following RV-C infection may facilitate optimised treatment strategies that may

cater to this sub-group of patients to improve clinical outcomes .

There are several limitations to acknowledge first, we did not have a cohort of non-atopic RV-C

infected patients so we could not exclude the effect of atopy independent of RV -C infection on

disease severity. Second, small sample size meant that we could not reach statistical

significance for many of our findings. Thus our data requires confirmation in a larger sample

size. Third, single time point sampling meant it was difficult to ascertain whether exacerbation

severity is a function of viral load. It would be Ideal to sequentially sample participants to

establish possible correlations between symptom severity and RV-C load. Fourth, given that

blood markers may reflect systemic level of inflammation, morbidities external of the

pulmonary environment may skew biomarker concentrations, making it difficult to interpret

the findings. Therefore, the surrogate markers for airway inflammation used in this study may

not be as accurate as analogous biomarkers derived from lower respiratory tract samples.

Fifth, we did not characterise the inflammatory mediators present in the nasal secretions of

our samples meaning we did not comprehensively explore the host's contribution to asthma

severity. This assumes importance because cytokine molecules can sustain and amplify the

inflammatory response through several mechanisms.

In conclusion, RV-C is an important agent in asthma exacerbation but the quantity of virus may

not have a significant influence on the severity of illness. Severe exacerbation induced by RV -C

is associated with a neutrophilic inflammatory pattern, and therapeutic interventions directed

at host related factors may be more important than those directed at controlling viral

replication. Therapeutic strategies such as administering vaccines may yield the greatest

Page 134: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

114

health benefit given that respiratory viruses especially rhinoviruses substantially contribute to

the burden of AAE.

Page 135: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

115

7. Cytokine profiles in nasal secretions of patients

hospitalised with Rhinovirus Species C associated

respiratory wheeze

Page 136: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

116

7.1 Introduction

Rhinoviruses are the most frequently detected viruses in pre-school aged children (3-5 years)

hospitalised with respiratory wheeze (Cox et al., 2013; Iwane et al., 2011). RV-C is

disproportionately the most commonly detected of the three rhinovirus species and is of

particular importance because it is strongly associated with asthma related hospitalisations

(Bizzintino et al., 2011a; Cox et al., 2013; Linsuwanon et al., 2009). Acute asthma exacerbations

(AAE) can be severe and are a major contributor to asthma related morbidity. Asthma related

costs pose an enormous financial burden on health resources, with annual cost in Australia

estimated to be more than $700 million (NAC, 2015). Further, RV associated early wheezing

episodes is an independent risk factor for recurrent wheezing and asthma inception (Jackson

et al., 2008b; Saraya et al., 2014).

Factors which contribute to RV-C associated clinical outcome have not been completely

elucidated but appear to have little or no association with viral load (Kennedy et al., 2014)(see

chapter 6). Evidence is accumulating that the host immune response may be the most

important factor in clinical outcome following infection (Holgate et al., 2005; Pritchard et al.,

2012; Quint, 2008). As yet, little data exists on the cytokine profile of hospitalised children

following RV-C infection. Previous studies with other respiratory viruses have shown that

several host cytokines correlate with duration of hospitalisation (Peiris, Hui, and Yen, 2010).

Further, human challenge studies using RV-16 (rhinovirus species A) report differences in

cytokine profiles between asthmatic and non-asthmatic patients suggesting the host immune

response is determined by atopic status (Hansel et al., 2017). However, little data exist on the

differences in cytokine profiles of asthmatic and non-asthmatic patients hospitalised following

RV-C infection. As such, the prime focus of this chapter was to characterise the nasal cytokine

Page 137: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

117

profile of hospitalised pre-school aged children with asthma as opposed to those without

asthma following RV-C infection. In addition, we planned to investigate the differences in RV-C

load and clinical outcomes among hospitalised children with asthma compared to hospitalised

non-asthmatic children. Knowing the cytokine profile provides further understanding of

pathogenic mechanisms involved and facilitates the development of better targeted

therapeutic options for the management of RV-C associated disease.

7.2 Samples

Flocked nasal swabs were collected from 605 children between the ages of 24 to 72 months of

age presenting to the Emergency Department at Princess Margaret Hospital with a clinical

diagnosis of acute wheeze. Eligible patients had clinical signs of wheeze on physicial

examination and were deemed clinically to have features in keeping with an acute upper

respiratory tract viral infection (URTI) or a history of URTI symptoms preceding the onset of

wheeze. The flocked nasal swabs were collected in 5mL of virus transport media (VTM)

transported to PWLM and stored at -80ºC pending further use. Samples (n=10) from otherwise

healthy children matched for age and sex were used as controls for nasal cytokines studies. All

samples were collected and tested between 2013 and 2016.

Nucleic acid was extracted from 200µL of clinical samples using automated extraction method

described in chapter 2. Nucleic acid was tested using a multiplex PCR for the common

respiratory pathogens including respiratory syncytial virus, human metapneumovirus, human

parainfluenza viruses, influenza viruses and adenovirus (Chidlow et al., 2009). RV screening

and genotyping was performed at the Telethon Kids Institute using previously published

primers (Lee et al., 2012a). The algorithm described in chapter 4 for RV-C viral load

Page 138: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

118

determination was applied to any sample that was RV-C positive. Viral load determination was

performed on the Rotor Gene 6000 cycler (QIAGEN, Australia). All experiments were

performed in triplicate including positive controls and non-template controls. Detection of

glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNA (Gueudin et al., 2003) was

utilized to ensure adequate specimen collection, RNA extraction and detection of PCR

inhibitors. Good quality samples were considered to be those with GAPDH CT values below

31.5. Cytokine bead array was used to obtain levels of cytokines from nasal secretions.

Page 139: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

119

7.3 Results

7.3.1 Virus Detections

This study enrolled 605 children aged between 24 and 72 months presenting to the emergency

department with respiratory wheeze. Nasal secretions were obtained prior to discharge or

when the patient was admitted into a hospital ward. Respiratory specimens were

subsequently screened for a range of respiratory pathogens using a validate d in-house

respiratory pathogen multiplex PCR panel. Respiratory pathogen detection results are shown

in figure 7.3-1. A respiratory pathogen was detected in 65% (n=393) of samples and as may be

seen from figure 7.3-1 rhinovirus (RV), respiratory syncytial virus (RSV) and adenovirus (ADV)

were the most frequently detected pathogens. RV-C (207/271, 76%) was the predominant

rhinovirus species detected (Fig.7.3-2). In 91% (n=188) of RV-C positive samples, RV-C was the

only pathogen detected.

Figure 7.3-1: Virus detection rates from samples of children hospitalised with respiratory wheeze. RV-

rhinoviruses, respiratory syncytial virus (RSV), adenovirus (ADV), human parainfluenza virus (HPIV), Influenza viruses (IFV) and HBoV (human bocavirus)

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

80.0

RV RSV ADV HPIV HCoV HMPV IFV HBoV

viru

s d

ete

ctio

n r

ate

s (%

)

Page 140: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

120

Figure 7.3-2: RV detection rates stratified by species. RV-C was the predominant species detected in children

hospitalised with respiratory wheeze

0%

10%

20%

30%

40%

50%

60%

70%

80%

90%

RV-A RV-B RV-C

RV

sp

eci

es

de

tect

ion

rat

es

(%)

Page 141: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

121

Virus detections were stratified for presence of physician diagnosed asthma in order to

determine whether RV-C is more commonly detected in asthma patients compared to non-

asthma patients. This analysis demonstrated that RV-C was the most common virus detected

in each group and no statistically significant difference in in detection rates was observed

(Fig.7.3-3).

Figure 7.3-3- A comparison of virus detection rates between patients with classified with asthma compared to those not classified with asthma.

Page 142: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

122

In this part of this study it was of importance to determine the contribution of RV -C infection

to clinical outcome. Length of hospitalisation was used as a marker of illness severity. Overall,

length of hospitalisation was relatively short with the majority of RV-C infected patients

discharged within a median time 10 hours. Mann-Whitney U test analysis showed that RV-C

load was not associated with hospitalisation given that viral load in the outpatient group

(6.00log10 copies/mL) were not significantly different (p>0.1) compared to the in-patient

group (5.82log10 copies/mL). Further, Area under the curve analysis showed that RV-C load is

not a suitable predictor of hospitalisation (Figure 7.3-4).

Figure 7.3-4: ROC assessment for RV-C load as a predictor of hospitalisation. AUC values shown in the legend (0.5, 95%CI, 0.24-0.74) demonstrated that RV-C load was poor predictor of hospitalisation.

Page 143: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

123

Next, we stratified RV-C infected patients by asthma diagnosis to get a better understanding of

the differences following RV-C infection. RV-C infected patients with asthma had a longer

duration of hospitalisation compared to RV-C non-asthmatics but this observation did not

reach statistical significance (Table 7.3-1). Further, RV-C load was evaluated between patients

with asthma and those without asthma in order to compare the replication kinetics of the virus

between these patients groups. As can be clearly seen in Figure 7.3-4 there was no difference

in RV-C load between the two patient groups.

Table 7.3-1 Summary of clinical and demographic data of hospitalised children with RV-C respiratory wheeze

# Comparison of continuous variables (Independent samples T-Test/Mann-Whitney U test) ¶ Comparisons of proportions (Fisher’s exact test) NS: non-significant, SD - standard deviation, IQR: Interquartile range

Asthmatics (n=57) Non-asthmatics (n=134) p-value

Age (mean ± SD) 2.9 ± 1.2 2.8 ± 1 n.s#

Sex (male %) 78 73 n.s ¶

History of eczema (No %) 45 20 n.s ¶

History of hay fever 11 0 n.s ¶

Median RV-C Log10 copies/mL (IQR) 5.8 (3.1 – 6.4) 5.8 (3.8 - 6.8) n.s#

length of stay (median hours, IQR) 7 (1-16) 10 (2-16) n.s#

Page 144: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

124

7.3.2 Nasal cytokine profiles of wheezing patients following RV-C infection

In order to gain further insight into the host response following RV -C infection, this study

explored the nasal cytokine profiles of RV-C infected children with asthma and without asthma

who were hospitalised with wheezing illness. Samples from 19 RV-C positive patients with

asthma and 15 RV-C positive patients without asthma were randomly selected for further

analyses. All 34 patients were steroid naïve and hospitalised with a RV -C only infection.

Community controls matched for age and sex were included in this part of the study. These

non-respiratory disease healthy controls were screened for respiratory pathogens and four

were detected with RV and one with RSV. These samples were excluded from the final

cytokine analysis.

Overall, our analysis demonstrated that there were no significant differences between the

groups for IL-2, IL-12, IFN- λ, IFN- α, IL-5, TNF- α, MIP-1β, and GM-CSF. When we examined

inflammatory mediators that were common in both the asthma and non-asthma groups

compared to controls, it was found that levels of IL-4, IL-6, IL-8, IL-9, IL-15, IL-27 and IP-10

were significantly elevated (p<0.05) (figures 7.3-5 to 10 and Table 7.3-2). Both groups also

demonstrated similar attenuation of CXCL-1 and the IFN-γ response.

When we investigated the inflammatory mediator changes that were unique to asthma

groups, they showed significantly elevated levels (p < 0.05) of IL-1β, IL-10, IL-13, IL-17 and IL-22

compared to controls. When the same analysis was applied to the non-asthma group, they

only showed significant attenuation of IL-18, IL-21, IL-31, CXCL-12, RANTES and eotaxin (figures

7.3-5 to 10 and Table 7.3-2).

Page 145: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

125

* *

*

*

*

A B

Figure 7.3-5 Levels of IL-4 (a) and IL-13 (b) in nasal secretions of non-respiratory disease controls, RV-C infected patients with asthma and without asthma. IL-4 and IL-13 were both significantly elevated in the asthmatic group but IL-13 levels did not differ significantly in the non asthmatic group compared to controls.* p<0.05.

Page 146: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

126

n.s

n.s

A B

Figure 7.3-6 Levels of IL-12 (a) and IL-2 (b) in nasal secretions of non-respiratory disease controls, RV-C infected patients with asthma and without asthma. Levels of IL-2 and IL-12 did not significantly differ when each group (asthmatics and non-asthmatics patients) were individually compared to controls.

Page 147: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

127

*

*

n.s

n.s

A B C

Figure 7.3-7 Levels of Interferon (IFN)-γ (a), IFN-λ (b), IFN-α (c), in nasal secretions of controls, hospitalised RV-C infected patients with asthma and without asthma. IFN-γ was significantly

attenuated in both patient groups compared to controls. The levels of IFN- λ, and IFN-α were not significantly different in either patient group compared to controls. *p<0.05.

Page 148: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

128

A B

*

*

*

Figure 7.3-8 Levels of IL-1β (a) and IL-6 (b) in nasal secretions of controls, hospitalised RV-C infected patients with asthma and without asthma. IL-1β was significantly elevated in the asthma group but not in the non-asthma group compared to healthy controls.*p<0.05.

Page 149: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

129

*

**

*

*

B A

Figure 7.3-9 Levels of IL-8 (a) and IP-10 (b) in nasal secretions of non-respiratory disease controls, RV-C infected patients with asthma and without asthma. IL-8 and IP-10 were both significanlty elevated both patient groups comapred to controls.* p<0.05, **p<0.001

Page 150: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

130

B A

*

*

Figure 7.3-10 Levels of IL-10 (a) and IL-17 (b) in nasal secretions of non-respiratory disease controls, RV-C infected patients with asthma and without asthma. IL-10 and IL-17 were only

significantly elevated in the asthma patient group compared to controls.* p<0.05

Page 151: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

131

Table 7.3-2 nasal cytokine levels of healthy non-respiratory disease controls, RV-C infected patients with asthma and without asthma.

Participants IL-5 IL-9 IL-15 IL-18 IL-21 IL-22 IL-23 IL-27 IL-31 TNF-α CXCL-12 CXCL-1 MIP-1β RANTES GM-CSF Eotaxin

Ctrls (n=5) 17 0 0 66 89 48 32 0 59 8 390 426 19 8 5 10

IQR 13-16 0-36 0-2 62-78 88-97 45-53 13-34 0-91 56-67 6-9 358-458 344-676 17-26 7-9 5-7 9-17

Asth (n=18) 12 51 10 38 114 157 38 103 77 8 331 193 45 9 7 8

IQR 7-23 20-95 6-19 12-76 32-191 45-389 16-99 25-241 31-147 5-15 124-453 93-315 17-67 3-21 2-15 5-14

Non-Asth (n=15) 14 36 7 22 50 91 16 47 38 5 142 103 25 4 5 6

IQR 10-16 18-87 4-10 13-26 29-81 35-174 15-35 11-107 23-55 5-11 79-228 62-240 10-87 3-5 2-8 5-9

p values

Asth vs Ctrls n.s 0.009 <0.001 n.s n.s 0.048 n.s 0.005 n.s n.s n.s 0.002 n.s n.s n.s n.s

Non-asth vs Ctrls n.s 0.024 <0.001 0.004 0.025 n.s n.s 0.047 0.025 n.s 0.005 0.001 n.s 0.007 n.s 0.004

Asth vs Non-asth n.s n.s 0.049 n.s n.s n.s 0.01 0.031 n.s n.s n.s n.s n.s n.s n.s n.s

Abbreviations: IL-interleukin, TNF-α- Tumour necrosis factor alpha, CXCL- chemokine ligand, MIP- Macrophage Inflammatory protein, RANTES- Regulated on Activation, Normal T Cell Expressed and

Secreted, GM-CSF-Granulocyte-macrophage colony-stimulating factor. Asth-asthmatics, Non.Asth-Non-asthmatics, Ctrls-controls, IQR- interquartile range

Page 152: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

132

7.3.3 Relationships between cytokines, RV-C load and clinical outcomes

In order to understand whether the level of viral replication influenced the magnitude of

cytokine response, a univariate analysis was conducted separately for asthma and non-

asthma patient groups. For this analysis, we selected inflammatory mediators that

demonstrated significant changes (either attenuated/elevated) compared to controls

(figures 7.3-5 to 10 and Table 7.3-2). Thus, for the asthma group we selected IFN-γ, IL-1β,

IL-4, IL-6, IL-8, IL-9, IL-10, IL-13, IL-15, IL-17, IL-27, IL-22, IP-10, and CXCL-1. Inflammatory

mediators selected to conduct the analysis in the non-asthma group included IFN-γ, IL-4,

IL-6, IL-8, IL-9, IL-15, IL-27 and IP-10, IL-18, IL-21, IL-31, CXCL-12, CXCL-1 RANTES and

eotaxin. The data in table 7.3-3 and 7.3-4 clearly shows weak non-significant relationships

between the selected cytokines and RV-C load for both groups.

Page 153: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

133

Table 7.3-3: An illustration of the relationship between RV-C load and inflammatory mediator production in the nasal secretions of children with asthma

Table 7.3-4: An illustration of the relationship between RV-C load and inflammatory mediator production in the nasal secretions of children without asthma

IFN-α IL-1β IL-4 IL-6 IL-10 IL-17 IL-9 IL-15 CXCL-1 IL-8 IP-10 IL-22 IL-27

Correlation Coefficient 0.05 -0.03 -0.10 -0.07 0.11 -0.06 -0.02 -0.08 -0.05 0.00 0.02 -0.18 -0.19

p- value 0.85 0.92 0.71 0.79 0.69 0.83 0.93 0.77 0.85 1.00 0.95 0.52 0.47

IFN-α IL-4 IL-6 IL-8 IL-9 IL-15 IP-10 IL-18 IL-21 IL-27 IL-31 Eotaxin CXCL-1 CXCL-12 RANTES

Correlation Coefficient -0.16 0.14 -0.27 -0.01 -0.03 -0.29 -0.18 -0.31 -0.20 0.02 -0.19 -0.15 -0.06 -0.10 -0.42

p-value 0.59 0.64 0.35 0.97 0.92 0.32 0.53 0.28 0.50 0.95 0.51 0.62 0.83 0.74 0.13

Page 154: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

134

Mann-Whitney U test was used in order to investigate whether hospitalisation was

associated with the aforementioned cytokines for either the asthmatic group or the non-

asthmatic group. As can be seem from table 7.3-5, in either group hospitalisation was not

associated any of the analysed cytokines.

Table 7.3-5: Association between cytokine production and hospitalisation of children hospitalised following RV-C infection

p-value (Asthmatics) p-value (Non-Asthmatics)

IFN-γ 0.26 0.947

IL-4 0.259 0.601

IL-6 0.212 1.00

IL-8 0.26 0.361

IL-9 0.109 0.744

IP-10 0.594 0.896

IL-15 0.191 0.512

IL-27 0.138 0.554

CXCL-1 0.515 0.794

IL-13 0.313 -

IL-1B 0.441 -

IL-17 0.233 -

IL-22 0.26 -

IL-18 - 0.794

IL-21 - 1.00

Eotaxin - 0.946

CXCL-12 - 0.896

RANTES - 0.647

IL-31 - 0.793

- denotes that analysis was not performed in that group.

Page 155: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

135

7.4 Discussion

Evaluating the contribution and the interplay between viral factors and host response is of

prime importance to our understanding of the pathogenic mechanisms involved in viral

induced respiratory wheeze. The results just described indicate that RV -C is the

predominant virus associated with respiratory illness with wheezing in hospitalised

preschool aged children. However, the amount of that virus does not appear to predict

clinical outcome. In both the asthmatic and non-asthmatic group the nasal cytokine

profiles indicated a Th2-type (allergic) response, which was stronger in the asthmatic

patients, while in both groups there was the same degree of suppression of the Th1-type

(microbicidal) responses. These responses were unrelated to virus load. This indicates that

the pathogenesis of wheeze in all RV-C infected patients was due to an allergic-type

cytokine response, triggered by infection but not driven by ongoing viral replication but by

the underlying tendency of the host to mount an allergic-type response. The nasal

cytokine profiles of RV-C infected patients showed a Th2-type cytokine response.

Furthermore, RV-C infection in asthmatics induces a more intense Th2 cytokine response

compared to infected non-asthmatic patients.

The present results indicate that rhinoviruses are the most important cause of respiratory

wheeze in hospitalised pre-school aged children. The proportion of RV in this group of

children is similar to the range reported in the previous chapter (see chapter five) and

within the range reported in the published literature (Johnston et al., 2005a; Miller et al.,

2007b; Rakes et al., 1999). Interestingly, RV-C as a sole pathogen was the most common

pathogen detected in hospitalised children with respiratory wheeze thus suggesting an

important role for RV-C in both asthma exacerbation and non-asthma respiratory wheeze.

Other studies have reported similar findings in hospitalised patients (Bizzintino et al.,

2011a). In our study, length of hospital stay was used as measure of clinical outcome. Our

Page 156: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

136

data showed that that length of hospitalization did not statistically significantly differ

between asthmatic patients and non-asthmatic patients following RV-C infection. That was

surprising as the cytokine data indicated that the asthmatic patients had stronger allergic-

type responses. However, the median length of stay was shorter in the non-asthmatic

group (7 hours versus 10 hours) but did not reach statistical significance. Nearly all patients

stayed less than 48 hours, so length of stay measured in hours is probably too crude a

measure of severity. Unfortunately other indicators, such as clinical disease severity

grading, were not available for these patients. However our data does suggest that RV-C

induces a relatively short clinical illness with few severe complications, which is consistent

with a recent Finnish multicentre study, which reported that children with a rhinovirus only

infection had shorter acute clinical course compared to children with RSV only infection

(Hasegawa et al., 2014).

It appears that RV-C load is not a risk factor for hospitalisation, since the findings herein

were unable to demonstrate significantly different RV-C loads between patients that were

hospitalised compared to patients discharged. This finding is in concordance with a recent

study reporting that rhinovirus load does not significantly correlate with short term clinical

outcome (Jartti et al., 2015). Furthermore, area under the curve analysis demonstrated

that RV-C load is not an accurate predictor of poor short term clinical outcome suggesting

that presentation factors other than the level of replication are more accurate predictors

of poor clinical outcome. That supports our findings that RV-C load was not related to the

strength of the Th2-type responses and presumably, the severity of wheeze.

The findings in relation to RV-C are in direct contrast to those found in RSV wheezing illness

in young infants, where viral load is an independent predictor of poor clinical outcomes

(Utokaparch et al., 2011). Altogether, the findings in this thesis suggest that the

mechanism underlying clinical outcome in wheezing illness unlike RSV are not predicated

Page 157: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

137

on viral replication but are determined by the host immune response and the triggering of

Th2-type cytokine responses.

The results in this chapter show immunological differences between RV-C infected patients

with asthma and without asthma. Among the Th2 cytokines IL-4, IL-9 were significantly

elevated in both patient groups but IL-10 and IL-13 were only significantly elevated in the

asthmatic group. In contrast, in both patient groups the levels of Th1 cytokines were either

significantly attenuated (IFN-y, IL-18) or showed no differences (IL-12, IL-23, IFN-α and IFN-

λ) compared to controls. Altogether, this suggests that RV -C infection promotes the

induction of a host response that is skewed to the Th2-type and away from the antiviral

Th1 type responses (Message et al., 2008; Pritchard et al., 2012). Signature Th2 cytokines

IL-4, IL-9 and IL-13 mediate recruitment of inflammatory cells to the lung, IgE isotype class

switching, upregulation of high affinity IgE receptor on mast cells and basophils, and IgE

dependent mast cell activation which results in the development of immediate allergic

reactions and mucus hypersecretion (Kau and Korenblat, 2014; Kearley et al., 2011).

The ability of the host to mount an effective interferon response typically contributes to

protection against viral illness (Pritchard et al., 2012). Our finding of blunted interferon

responses in asthmatics and non-asthmatics suggests RV-C either directly or indirectly

attenuates interferon production and secretion. It is more likely that the production of

Th2-type cytokines downregulate the induction of antiviral interferons and other Th1-type

cytokines important for the induction of an antiviral response (Machado et al., 2009).

The observation of type 2 response following RV-C infection in pre-school aged children

without asthma raises an interesting point in the context of early viral infection and

childhood asthma. It may simply be that wheeze can be triggered in any child infected with

RV-C, and the pathways for induction of wheeze are the same, irrespective of whether

they have an asthmatic predisposition. However, my finding that the Th2-type responses

Page 158: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

138

are greater in asthmatic than in non-asthmatic patients, suggest a synergetic relationship

between RV-C and host factors in the genesis of wheeze. It is therefore possible that the

observed skewing towards Th2-type responses following RV-C infection in early childhood

may predispose to later onset of asthma in susceptible children. Especially because

previous reports demonstrate that early life RV ALRI is an independent predictor of

childhood asthma (Lemanske et al., 2005). Alternatively, It could be that children classified

as non-asthmatic in this study may in fact be asthmatic and have not yet been diagnosed.

Diagnosing asthma in children below the age of six years can be difficult because episodic

respiratory symptoms such as wheezing are very common in children below this age, which

includes the children in the studies used in this thesis. Therefore, the observed dominance

of Th2-type response following RV-C infection in “non-asthmatics” may in fact be a marker

of predisposition to asthma. In addition, this finding also highlights the complexity of the

virus-host interactions in the context of asthma and indicates that developing asthma likely

necessitates recurrent infections, a suitable genetic predisposition and allergen exposure

(Ahanchian et al., 2012). Novel therapy that reduces recurrent respiratory viral infection

(specifically RV-C and RSV) which skew host immune response to an allergic phenotype

could help prevent the development of childhood asthma.

My findings also demonstrated elevated levels of IL-10 in patients with asthma but not in

non-asthmatics. Some studies have reported that IL-10 prevents dysregulated

inflammatory processes that cause airway narrowing (Kawano et al., 2016; Message et al.,

2008), but other studies report the contrary suggesting that IL-10 may contribute to

altered airway function (Mäkelä et al., 2000). One may speculate that following RV-C

infection, the elevated levels of IL-10 in patients with asthma but not in non-asthmatic

patients is not indicative of a protective role but one that facilitates Th2-type responses

possibly by promoting IL-4/IL-13 mediated responses (Schopf et al., 2002).

Page 159: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

139

This analysis also revealed significantly elevated levels of IL-6 and IL-8 in both asthmatic

and non-asthmatic groups. IL-6 is a known pro-inflammatory cytokine that is increased

during symptomatic RV infections, and it promotes the acute phase response in part by

neutrophil activation, as well as stimulating T cell responses and antibody production (Zhu

et al., 1996). IL-8 is a potent chemoattractant of neutrophils, elevated levels of IL- 8

induced by RV infection correlate with increases in bronchial hyper-reactivity and severity

of respiratory symptoms (Zhu et al., 1997). Interestingly, levels of RANTES, eotaxin and IL-5

(all potent chemoattractants of eosinophils) did not significantly differ in either patient

group compared to healthy controls. Altogether these analyses suggest neutrophilic

inflammatory pattern, but not eosinophilic inflammation contributes to disease

pathogenesis following RV-C infection.

Previous studies have associated IL-17 with promoting a Th2-type inflammatory

environment by enhancing production of IL-13 (Jin and Dong, 2013). IL-13 and IL-17 are

reported to function synergistically to regulate the epithelial cell response that controls

mucus production following viral infection (Jin and Dong, 2013). IL-17 also facilitates

neutrophil activation and proliferation in non-eosinophilic asthma in part by enhancing the

production of the pro-inflammatory cytokines IL-6 and IL-8 (Linden, 2001). Animal studies

modelling the interplay in the lung between RV and IL-17 report that IL-17 is detected at

higher levels in the lung of asthmatics compared to healthy controls following RV infection

(Al-Ramli et al., 2009). It is therefore possible that IL-17 may contribute to RV-induced

disease in asthma patients.

In both patient groups RV-C infection was associated with elevated levels of IP-10

compared to controls which is in agreement with other reports (Culley et al., 2006;

Matsumoto and Inoue, 2014; Quint, 2008). IP-10 is a chemoattractant that mediates

inflammatory response by recruitment of circulating leucocytes to the site of

Page 160: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

140

inflammation. Increased levels of IP-10 are generally considered as a non-specific response

to viral infection (Quint, 2008).

This analysis also demonstrated a significant elevation of IL-1β levels in asthma patients

compared to controls but this was not evident in the children without asthma. IL-1β is a

proinflammatory cytokine that is released following RV infection and contributes to

pathogenesis of respiratory disease by recruitment of inflammatory cells and enhancing

production of IL-8 (Kluijver et al., 2003). Experimental human models of asthma

exacerbations demonstrate that an early rise in IL-1β in respiratory secretions is temporally

associated with clinical symptoms (Kluijver et al., 2003; Liu et al., 2013). However, IL-1β

was not significantly correlated with any of the markers of severity used in this study.

However, it remains possible that levels of IL-1β in children with asthma following RV

infection may be a marker of more severe illness compared to non-asthmatics, and that

the markers used in our studies were not appropriate for detecting this effect.

This study has potential limitations. Firstly, only one time point was used for the

measurement of cytokines and viral load, and although early evaluations are useful for the

acute phase response, they do not permit further insights such as those provided by

sequential measurements. Even though it is tempting to speculate that there is a direct

association between cytokine concentrations and illness severity, the full burden of disease

cannot be solely attributed to this phenomenon because cytokines may behave as markers

of tissue damage, without necessarily contributing to pathology directly.

In summary, RV-C appears to be the predominant pathogen associated with respiratory

wheeze in hospitalised preschool aged children. A short-lived clinical course appears to be

a hallmark of RV-C infection. RV-C load is neither a risk factor nor a reliable marker for

hospitalisation following infection. Further, RV-C infection is more important than the level

of replication because it appears that factors other than viral load drive clinical course. RV -

Page 161: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

141

C associated respiratory wheeze in hospitalized children is characterized by a dominant

Th2-type inflammatory response. Furthermore, RV-C promotes a more intense cytokine

response in children with asthma compared to children without asthma. The induction of

cytokines that mediate recruitment and activation of neutrophils may be an important

underlying pathogenic mechanism associated with RV-C disease. Thus, potential

therapeutic interventions should be aimed at modulating the host response following

infection.

Page 162: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

142

8. General Discussion and Conclusions

Page 163: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

143

8.1 Introduction

Acute lower respiratory infections are an important cause of morbidi ty and mortality in

children worldwide. Furthermore, respiratory tract infections also deliver an enormous

financial burden on healthcare for which the direct and indirect cost to the Australian

health care system is estimated to be up to AUD 600 million each year (The Australian Lung

Foundation, 2007). Within the last few decades it has become abundantly clear that

viruses are the leading cause of ALRI, yet therapeutic options are limited. In addition to

ALRI, a considerable amount of evidence implicates respiratory viral infection as a major

trigger of asthma exacerbations (Johnston et al., 2005b; Matsumoto and Inoue, 2014;

Message and Johnston, 2002). Further, RV and RSV are implicated in asthma pathogenesis

in children. RSV is the leading cause of lower respiratory tract infection and death in

children below the age of two years old (Nair et al., 2013). Severe RSV bronchiolitis in

infancy is consistently implicated with persistent wheeze to age six (Moore et al., 2013)

and although the picture is still incomplete, the current evidence suggests that severe RSV

bronchiolitis may be an independent risk factor for allergic sensitisation (Wu and Hartert,

2011).

RVs are disproportionately the most common cause of upper respiratory tract infection i n

humans and are now recognised as an important pathogen of the lower respiratory tract.

RV species C especially, has been identified as an important contributor to wheezing illness

in paediatric medicine (Bizzintino et al., 2011b; Cox et al., 2013; Linsuwanon et al., 2009).

Unlike the other RV species that have been extensively studied over the years, RV -C cannot

be grown in conventional cell culture. The lack of a conventional system allowing the

propagation of RV-C in vitro has precluded investigations by traditional virological studies.

This has meant there is little understanding of the pathogenic properties of RV -C. It is

through the use of molecular techniques that all of the currently recognised RV -C

Page 164: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

144

genotypes have been identified and their diversity established (Khetsuriani et al., 2008;

McIntyre et al., 2013). However, their pathogenic properties are still largely undefined,

and their association with disease severity still yet to be clearly elucidated. It is well

established in other infections that viral load plays an essential role in disease progression

and clinical outcome (Bagga et al., 2013; Feikin et al., 2015; Fleming et al., 2005). For

example, in chapter 5 of this thesis we demonstrated that RSV load is higher in

symptomatic children. In the literature, viral load HMPV is associated with disease severity

in young children (Roussy et al., 2014). Similarly, a recent study conducted in China

demonstrated that high HBoV load is associated with more severe lower respiratory tract

symptoms, longer duration of wheezing and hospitalisation (Deng et al., 2012). The high

genetic variability in the PCR target region of RV-C has in the past impeded the

development of reliable molecular based viral load assays to investigate this association.

Other studies have utilised degenerate bases in the PCR primer and probe sets to

overcome the inter-genotypic variability of RV-C (Granados et al., 2012). However, these

efforts are known to compromise accuracy of measurements (Chemidlin Prevost-Boure et

al., 2011). Thus, this thesis aims to improve understanding of the underlying

pathophysiological mechanisms in the context of RV-C wheezing illness through the

development of a reliable molecular based method for quantifying RV-C load and then the

characterisation of the host response following infection.

Page 165: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

145

8.2 Epidemiology of respiratory viruses in young

children under the age of five years

RV-C and RSV are the predominant aetiological agents of wheezing illness in young children

(chapter 5-7). Virtually all children are estimated to have been infected by RSV before the

age of three, with bronchiolitis being the predominant clinical manifestation (chapter 5).

RSV predominates as the aetiologic agent of medically attended ALRI in the first year of

life, with peak hospitalisation rates observed in the first two months of life (Bont et al.,

2016; Hasegawa et al., 2014; Henderson et al., 2005)(chapter 5). This finding supports

previous reports in the literature demonstrating that hospitalisation rates for RSV disease

increases with decreasing age, peaking in the first few months of life (Bont et al., 2016).

Indeed, this study also contributes to the global understanding of RSV morbidity

specifically in the first year of life, which is pertinent for future RSV vaccination strategies.

Further, given that the burden of RSV disease has been well described in many high income

settings but data from low income countries is sparse. The findings in chapter 5 come from

a low-middle income country and extend the current knowledge of the global impact of

RSV, and indicate that the inverse relationship between RSV infection and age is a universal

phenomenon in children.

Contrary to the data from many previous epidemiological studies (Camargo et al., 2012;

Jartti et al., 2008; Self et al., 2015), the findings in chapter 5 demonstrated a relatively high

percentage of samples from NRD controls that tested positive for RSV (17%). Indeed this

finding may represent by-stander nucleic acid from previous infection, given that PCR can

detect viral nucleic acid in respiratory secretions even after the resolution of

infection(Camargo et al., 2012). Unfortunately no attempt was made to isolate virus in cell

culture and as such precluded the determination of viability. However, even in the absence

of the viability data, the high rate of detection of RSV-RNA in healthy children suggests that

Page 166: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

146

RSV infection is frequent in those communities. Alternatively and biologically plausible,

these data may also suggest individuals with asymptomatic RSV may be an important

transmission pathway in this region. A recent study in Kenya (a region similar to where our

study samples were collected) reported similar findings showing a high percentage (40%)

of asymptomatic RSV detection in the community (Munywoki et al., 2015). It is unknown

whether these frequent asymptomatic cases are a result of unique culturally specific

factors, differences in host immunity, or differences in the infecting RSV strains.

It also appears from this study that RVs are the predominant aetiologic agent of wheezing

illness in preschool aged children, in concordance with previous reports (Miller et al.,

2007a). Chapter 6 and 7 demonstrate that RV-C is the most commonly detected of the RV

species in pre-school aged children (3-5 years old) hospitalised with a wheezing illness or

asthma exacerbation. Table 6.3-2 also indicates that RV-C may be more inherently

pathogenic than RV-A (Unfortunately a lack of numbers for RV-C could not permit this

analysis to draw a reliable condition). Overall, our data suggests that species specific

differences may be important determinants of RV epidemiology (Cox et al., 2013; Liu et al.,

2016). RV-C utilises a different cell surface receptor to RV-A and RV-B for attachment.

Recent studies have identified cadherin-related family member 3 (CDHR3) as the most

likely receptor for RV-C entry and replication (Bochkov et al., 2015). CDHR3 is ubiquitously

expressed in the airway epithelium including areas of the lower respiratory tract.

Interestingly, a variant of the CDHR3 gene which has been associated with wheezing illness

and childhood asthma (Bonnelykke et al., 2014) has also been shown to enhance RV-C

binding and progeny yields in vitro (Bochkov et al., 2015). It is unknown whether the same

relationship exists in vivo and also what implication this may have on the risk of persistent

wheeze and asthma inception.

Page 167: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

147

In this thesis, children hospitalised with RV-C infection infrequently required intensive care

treatment and were discharged within 48 hours of admission (chapter 7), supporting other

data that RV-C associated AAE results in a self-limiting illness without serious complications

(Jartti et al., 2015). However, we may have missed more subtle differences in clinical

course that were not reflected in duration of stay or ICU admission rates.

The findings of this thesis also demonstrated that there is an age -related shift in the

epidemiology of medically attended respiratory viral illness. RSV predominates as the most

common aetiologic agent of viral wheeze (bronchiolitis) in the early years of life especially

in the first year of life, which is in agreement with previous reports (Lukšić et al., 2013;

Takeyama et al., 2015a). The findings in chapter 6 and 7 demonstrate that RVs, especially

species C, appear to assume predominance thereafter. An explanation of this observed

shift was not investigated in this study but one may speculate that it is in part driven by a

decline in RSV disease severity as a result of previous exposure in the first years of life, and

also in part by the large number of genetically and likely antigenically distinct RV -C types

capable of infection. Given that RV infection has been reported to be an independent risk

factor for subsequent childhood asthma and decreased lung function (Guilbert et al., 2011;

Tovey et al., 2015) future study should investigate the influence of this sequential infection

in the development of persistent wheeze and asthma onset. It may be that in susceptible

children severe RSV infection primes them for asthma, possibly by interfering with normal

development of the lung (Gern and Busse, 2002) so that subsequent RV-C infection may

permanently shift the host immune response towards a more allergic phenotype.

This thesis also highlighted the difficulty in assessing etiologic contribution of viruses to

clinical disease using qualitative PCR analysis. Chapter 5 and 6, both of which were case

control studies demonstrated a high rate of respiratory virus detections in the non -

respiratory disease control groups. These findings may reflect incidental detection of

Page 168: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

148

nucleic acid from a recent previous infection, frequent acute infections, or possibly long

term persistence of infection. As demonstrated in this study, quantitative results may be

helpful in determining the significance of these viruses as a cause of ARTI.

8.3 Reliable methods of accurately determining viral

load in RV-C infected patients

Accurate methods to quantify viral load can be a powerful tool for improving our

understanding of the kinetics and pathogenesis of viral replication (Bagga et al., 2013),

establishment of clinical correlates and evaluating the effectiveness of antiviral therapy

(Boivin et al., 2003). This study, like others has demonstrated the frequency of infections

with RVs (Bruning et al., 2015; Camargo et al., 2012; Piralla et al., 2009), especially RV-C.

Accurately measuring viral loads has been hampered by two major challenges. Firstly, RV-

C is uncultivable in conventional cell culture, so viral load cannot be measured in that way.

Therefore, molecular methods, especially techniques that are capable of accurately

determining viral load in clinical samples become necessary. However, the extensive

variability in the PCR target region of RV-C genotypes presents the second challenge in

developing reliable methods of quantifying RV-C in clinical samples. No single primer probe

pair could be expected to provide accurate quantification of the wide range of RV-C types.

In this study, viral loads assays for RV-C were developed as a tool to obtain an accurate

understanding of the contribution of RV-C to disease.

Given that several RV-C genotypes can circulate concurrently (Chapter 7) (McIntyre et al.,

2013) it was imperative for this project to design an assay that would provide coverage for

all the currently known genotypes. Computational analysis was used to assess the range of

coverage of each assay against sequences representative of the diverse range of RV-C

genotypes. This analysis revealed that four real-time PCR probes were required to

Page 169: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

149

overcome the inter-genotypic variation within the target region. This highlights the

importance of computational analysis before proceeding to laboratory evaluations. This

study also demonstrated the importance of target – assay homology on the robustness of

the assay in two respects. Firstly, complete homology between target and probe enhances

the quantitative accuracy of the assay, and secondly, it improves assay specificity.

This study also demonstrated that there are specific factors which must be taken into

account when developing a reliable PCR assay to quantify viral load. Firstly, it was shown

that the reaction conditions provided by the detection reagent is a determinant of

accurate measurement. Our findings indicate that choice of reaction mixture, whether

commercial or in-house, should be evaluated prior to use, as some reaction mixes are

superior to others for the purpose of viral load determination. Secondly, careful primer

and probe optimisation is necessary, and this should also undergo vigorous assessment. In

our study it was necessary to optimise and use asymmetric PCR rather than conventional

PCR conditions to enable better sensitivity and reproducibility. Thirdly, specimen collection

is a source of result variability and may influence the accuracy and the interpretation of the

viral load measurement (Hayden et al., 2012). This is because the quality (amount of cells)

and the quantity (volume per sample) of the different sampling methods vary considerably.

Therefore, the addition of an invariant endogenous control (GAPDH) in the assay was

essential to correct for any sample to sample variation.

The clinical and public health importance of the assays developed in this study is that it is

now possible to accurately and reliably investigate the viral replication kinetics of RV -C. In

so doing these assays will stimulate further discussion and insight into the kinetics and

pathogenesis of RV-C replication. Like RSV (Bagga et al., 2013) and influenza (Boivin et al.,

2003), for which the pattern of viral load indicates when antiviral therapy would be most

Page 170: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

150

effective, an accurate understanding of the natural history of RV-C infection will be

important in the development of future therapeutic strategies.

8.4 Viral Determinants of severity of RV-C induced

wheezing illness

Early studies in a range of viruses have shown a relationship between the magnitude of

replication and clinical outcome. For example in HIV infection, a high viral load is

associated with poor clinical outcomes (Attia et al., 2009). In the context of respiratory

viruses, evidence suggests that RSV disease in young children is a virus mediated

phenomenon (chapter 5) and virus load is an independent predictor of disease severity

(Houben et al., 2010; Utokaparch et al., 2011). It appears to be different for RV-C.

Although the clinical manifestations in young children hospitalised following RV -C and RSV

infection are indistinguishable (predominantly a wheezing illness), the findings in this

thesis reveal that the drivers of illness severity may be different. We found that illness

severity in hospitalised children with an exacerbation of asthma or frank whee zing illness is

not associated with RV-C load. This may suggest that a pathogenic process triggered by

infection rather than the ongoing viral replication determines the severity of illness .

Interestingly, while the evidence herein suggests that illness severity is not associated with

viral load, a previous experimental study using RV-A demonstrated that viral load appeared

to drive symptom severity in study participants (Message et al., 2008). Indeed, this may

suggest species specific differences in mechanistic pathways in the pathogenesis of

disease. Taken together, these findings have implications for novel therapy initiatives given

that for a specific type of virus, virus specific interventions may be necessary while on the

other hand and in the context of RV-C these interventions may not be as beneficial to high

risk populations. Indeed, a RV vaccine would have substantial benefits for the community

Page 171: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

151

as a whole, especially in individuals with underlying chronic respiratory diseases such as

asthma. However, technical feasibility of composing a vaccine with over 100 or more

serologically distinguishable antigens and also capable of generating a broad long lasting

immune response are the two main challenges that have in the past precluded RV vaccine

development. Encouraging results in recent animal studies has demonstrated that it may

be immunologically possible to develop a polyvalent RV vaccine (Lee et al., 2016). Indeed

an issue that is till yet to be resolved is the reliable propagation RV-C in cell culture. The

viruses used by Lee et al. (2016) were generally RV-A species. As this thesis has shown RV-C

is the most frequently detected virus in medically attended wheezing illness in preschool

aged children and thus any RV vaccine developed should also include antigens that broadly

represent C species.

8.5 Host response following RV-C infection

This thesis also provides an insight into the interplay between RV-C and the host immune

response in hospitalised young children following infection. The findings herei n suggest

that the immune response following RV-C infection shapes illness severity in asthmatic

patients, and most likely in non-asthmatic children (chapter 7). This phenomenon is not

associated with RV-C load and suggests an independent role for immunopathogenesis in

the clinical outcome of RV-C infection. RV-C infection promotes Th2-biased responses in

asthmatic and non-asthmatic children hospitalised with RV-C wheeze, similar to what is

observed in severe RSV bronchiolitis (Zeng et al., 2011). Our work demonstrated that the

dominant Th2 cytokines including IL-4 and IL-13 are more pronounced in asthmatic

children compared to non-asthmatic children; possibly a result of the underlying allergic

pulmonary environment. However, this analysis did not demonstrate any associations

between this pronounced cytokine response and poor clinical outcomes. Other studies

have shown that these cytokines are major mediators of exaggerated airway responses

Page 172: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

152

following infection (Mukherjee and Lukacs, 2010; Pala et al., 2002). Future study would

benefit from understanding whether this Th2 biased response relates in any way to

subsequent asthma development later on in life.

RV-C infection induced a robust acute phase neutrophil response but this response was

independent of viral load. Further, increasing neutrophil numbers correlated with disease

severity (chapter 6). Despite neutrophils being important in the engulfment and

subsequent elimination of invading extracellular microorganisms, controversy still

surrounds their role in viral infections. Neutrophils can only be of benefit if they assist in

viral clearance, which in our study does not appear to be the case, as the neutrophil

response was highest in the sickest children but viral load remain unchanged (chapter 6).

Previous reports have also shown that an increase in neutrophil numbers in asthmatic

patients following viral infection corresponds with an increase in symptom score (Louis et

al., 2000). Furthermore, widespread neutrophil infiltration is seen in the lung tissue from

fatal cases of AAE (Wenzel et al., 1999) and RSV LRTI (Johnson et al., 2006). Neutrophilic

inflammation, specifically in asthma patients assumes importance because of the

refractory nature of the neutrophilic asthma phenotype to standard asthma treatment

(Alam et al., 2017). It also assumes importance because it suggests that different triggers

of asthma may induce different inflammatory patterns, which in turn means that

therapeutic interventions may need to be modified according to the response type.

Macrolide antibiotic treatment is a potential intervention for dampening excessive

neutrophilic inflammation following RV-C induced respiratory wheeze (Brusselle and

Pavord, 2017; Gibson et al., 2017). Studies performed with RV-C in this context are

currently scarce but will be necessary for acute treatment and to mitigate long term

asthma like symptoms. Macrolides have demonstrated anti-neutrophilic activities in vitro

models of pulmonary infection and inflammation (Gielen, Johnston, and Edwards, 2010;

Menzel et al., 2016; Tamaoki et al., 1999). Furthermore, in a recent proof of concept study

Page 173: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

153

macrolide treatment in RSV induced bronchiolitis demonstrated anti -neutrophilic activity

and consequently better clinical outcomes in comparison to placebo treated patients

(Beigelman et al., 2015). A similar finding has also been reported for influenza infection

(Lee et al., 2017).

Chapter 7 provided insights into the underlying mediators of the observed neutrophil

infiltration. This robust neutrophil infiltration appears to be mediated by the neutrophil

chemoattractant IL-8 (chapter 7), which was observed to be higher in asthma patients

compared to non-asthma patients suggesting a more vigorous inflammatory response

(Alam et al., 2017). It also appears likely that IL-17 may contribute to enhanced neutrophil

recruitment specifically in children with asthma (chapter 7) but its role in clinical outcome

could not be established in this study. In vitro experiments have shown that exacerbation

of neutrophil responses is one of the main consequences of IL-17 secretion following

rhinovirus infection (Wiehler and Proud, 2007). Further, models of rhinovirus induced AAE

have demonstrated that IL-17 secretion is associated with increased airway hyper-

responsiveness (Al-Ramli et al., 2009). Future study should be designed to assess the

therapeutic potential of monoclonal antibody targeting these cytokines in response to RV -

C infection.

One issue that was not investigated in this thesis and is of potential importance is the

contribution the airway microbiome to natural course of vi ral respiratory illness. It is well

established that the presence of the microbiota is pivotal for the development and

maintenance of the host defence. Evidence in the literature suggests that respiratory

commensal bacteria can play both a protective role and a pathological role. For instance,

the presence of a commensal nasopharyngeal microbiota protected mice against RSV -

induced airway hyper-responsiveness (Ni et al., 2012). On the hand a pathological role has

also been described in which certain bacterial pathogens can provoke a strong infection as

Page 174: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

154

well as an exaggerated host immune response (Sajjan et al., 2006). The evidence available

suffices to show that the complexity of microbiome interactions in the airways, possibly

contributes to the susceptibility to exacerbations and the natural course of airway

diseases. Future study must consider how RV-C interacts with the airway microbiome to

modulate clinical outcome.

8.6 Conclusion

Overall, RV-C predominates as the most important viral pathogen in preschool aged

children hospitalised with a wheezing illness. The accurate quantification method used to

measure viral load in this project has provided a novel tool for obtaining insight into

replication kinetics of RV-C and enabled further study into its contribution to disease. This

thesis has also demonstrated that the existing platform used to determine viral load has its

limitations and advanced techniques/platforms such as digital PCR may well be used as the

optimal quantification method for viruses with high sequence diversity. Nonetheless, the

accurate method of viral load determination developed in this thesis has demonstrated

that RV-C load does not drive severity of infection; it merely triggers the disease process.

There is strong evidence that RV-C infection is characterised by a strong Th2 biased

response. It appears that the magnitude of neutrophilia within the airways may in part

modulate severity of illness in young children hospitalised following RV-C induced

wheezing illness. Further experimental study is required to understand more fully the

interplay between RV-C and the host immune response in shaping the outcome of RV-C

infection. A better understanding will help guide therapeutic approaches and the

development of new treatment and preventive strategies.

Page 175: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

155

Page 176: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

156

Bibliography

Abdullahi, O., Nyiro, J., Lewa, P., Slack, M. and Scott, J.A., 2008. The descriptive epidemiology of Streptococcus pneumoniae and Haemophilus influenzae nasopharyngeal carriage in children and adults in Kilifi district, Kenya. Pediatr. Infect. Dis. J. 27, 59-64.

Ahanchian, H., Jones, C.M., Chen, Y.-s. and Sly, P.D., 2012. Respiratory viral infections in children with asthma: do they matter and can we prevent them? BMC Pediatr. 12, 147.

Akerlind, B. and Norrby, E., 1986. Occurrence of respiratory syncytial virus subtypes A and B strains in Sweden. J. Med. Virol. 19, 241-7.

Al-Ramli, W., Prefontaine, D., Chouiali, F., Martin, J.G., Olivenstein, R., Lemiere, C. and Hamid, Q., 2009. T(H)17-associated cytokines (IL-17A and IL-17F) in severe asthma. J. Allergy Clin. Immunol. 123, 1185-7.

Alam, R., Good, J., Rollins, D., Verma, M., Chu, H., Pham, T.H. and Martin, R.J., 2017. Airway and serum biochemical correlates of refractory neutrophilic asthma. J. Allergy Clin. Immunol.

Alsaleh, A.N., Grimwood, K., Sloots, T.P. and Whiley, D.M., 2014. A retrospective performance evaluation of an adenovirus real-time PCR assay. J. Med. Virol. 86, 795-801.

Annan, A., Baldwin, H.J., Corman, V.M., Klose, S.M., Owusu, M. and Nkrumah, E.E., 2013. Human betacoronavirus 2c EMC/2012-related viruses in bats. Ghana and Europe Emerg Infect Dis 19.

Arden, K., McErlean, P., Nissen, M., Sloots, T. and Mackay, I., 2006. Frequent detection of human rhinoviruses, paramyxoviruses, coronaviruses, and bocavirus during acute respiratory tract infections. J. Med. Virol. 78, 1232 - 1240.

Attia, S., Egger, M., Muller, M., Zwahlen, M. and Low, N., 2009. Sexual transmission of HIV according to viral load and antiretroviral therapy: systematic review and meta-analysis. AIDS 23, 1397-404.

B-Lajoie, M.-R., Drouin, O., Bartlett, G., Nguyen, Q., Low, A., Gavriilidis, G., Easterbrook, P. and Muhe, L., 2016. Incidence and prevalence of opportunistic and other infections and the impact of antiretroviral therapy among HIV-infected children in low and middle-income countries: a systematic review and meta-analysis. Clin. Infect. Dis.

Bafadhel, M., McKenna, S., Terry, S., Mistry, V., Reid, C., Haldar, P., McCormick, M., Haldar, K., Kebadze, T. and Duvoix, A., 2011. Acute exacerbations of chronic obstructive pulmonary disease: identification of biologic clusters and their biomarkers. Am. J. Respir. Crit. Care Med. 184.

Bagga, B., Woods, C.W., Veldman, T.H., Gilbert, A., Mann, A., Balaratnam, G., Lambkin-Williams, R., Oxford, J.S., McClain, M.T., Wilkinson, T., Nicholson, B.P., Ginsburg, G.S. and Devincenzo, J.P., 2013. Comparing influenza and RSV viral and disease dynamics in experimentally infected adults predicts clinical effectiveness of RSV antivirals. Antivir. Ther. 18, 785-91.

Bandi, V., Jakubowycz, M., Kinyon, C., Mason, E.O., Atmar, R.L., Greenberg, S.B. and Murphy, T.F., 2003. Infectious exacerbations of chronic obstructive pulmonary disease associated with respiratory viruses and non-typeable Haemophilus influenzae. FEMS Immunol. Med. Microbiol. 37.

Baraldo, S., Contoli, M., Bazzan, E., Turato, G., Padovani, A., Marku, B., Calabrese, F., Caramori, G., Ballarin, A., Snijders, D., Barbato, A., Saetta, M. and Papi, A., 2012. Deficient antiviral immune responses in childhood: Distinct roles of atopy and asthma. J. Allergy Clin. Immunol. 130, 1307-1314.

Page 177: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

157

Beigelman, A., Isaacson-Schmid, M., Sajol, G., Baty, J., Rodriguez, O.M., Leege, E., Lyons, K., Schweiger, T.L., Zheng, J., Schechtman, K.B., Castro, M. and Bacharier, L.B., 2015. Randomized trial to evaluate azithromycin's effects on serum and upper airway IL-8 levels and recurrent wheezing in infants with respiratory syncytial virus bronchiolitis. J. Allergy Clin. Immunol. 135, 1171-8.e1.

Bel, E.H., Sousa, A., Fleming, L., Bush, A., Chung, K.F., Versnel, J., Wagener, A.H., Wagers, S.S., Sterk, P.J. and Compton, C.H., 2011. Diagnosis and definition of severe refractory asthma: an international consensus statement from the Innovative Medicine Initiative (IMI). Thorax 66, 910-917.

Bellinghausen, C., Gulraiz, F., Heinzmann, A.C.A., Dentener, M.A., Savelkoul, P.H.M., Wouters, E.F., Rohde, G.G. and Stassen, F.R., 2016. Exposure to common respiratory bacteria alters the airway epithelial response to subsequent viral infection. Respir. Res. 17, 68.

Bermejo-Martin, J.F., Garcia-Arevalo, M.C., Alonso, A., De Lejarazu, R.O., Pino, M., Resino, S., Tenorio, A., Bernardo, D., Leon, A.J., Garrote, J.A., Ardura, J., Dominguez-Gil, M., Eiros, J.M., Blanco-Quiros, A., Munoz-Fernandez, M.A., Kelvin, D.J. and Arranz, E., 2007a. Persistence of proinflammatory response after severe respiratory syncytial virus disease in children. J. Allergy Clin. Immunol. 119, 1547-1550.

Bermejo-Martin, J.F., Garcia-Arevalo, M.C., De Lejarazu, R.O., Ardura, J., Eiros, J.M., Alonso, A., Matias, V., Pino, M., Bernardo, D., Arranz, E. and Blanco-Quiros, A., 2007b. Predominance of Th2 cytokines, CXC chemokines and innate immunity mediators at the mucosal level during severe respiratory syncytial virus infection in children. Eur. Cytokine Netw. 18, 162-7.

Berry, M.A., Morgan, A., Shaw, D.E., Parker, D., Green, R.H., Brightling, C.E., Bradding, P., Wardlaw, A.J. and Pavord, I.D., 2007. Pathological features and inhaled corticosteroid response of eosinophilic and non-eosinophilic asthma. Thorax.

Bizzintino, J., Lee, W.-M., Laing, I.A., Vang, F., Pappas, T., Zhang, G., Martin, A.C., Khoo, S.-K., Cox, D.W., Geelhoed, G.C., McMinn, P.C., Goldblatt, J., Gern, J.E. and Le Souëf, P.N., 2011a. Association between human rhinovirus C and severi ty of acute asthma in children. Eur. Respir. J. 37, 1037-1042.

Bizzintino, J., Lee, W.M., Laing, I.A., Vang, F., Pappas, T., Zhang, G., Martin, A.C., Khoo, S.K., Cox, D.W. and Geelhoed, G.C., 2011b. Association between human rhinovirus C and severity of acute asthma in children. Eur. Respir. J. 37.

Bizzintino, J.A., Lee, W.M., Laing, I.A., Vang, F., Pappas, T., Zhang, G., Martin, A.C., Geelhoed, G.C., McMinn, P.C., Goldblatt, J., Gern, J.E. and Le Souëf, P.N., 2011c. Association between human rhinovirus C and severity of acute asthma in children. The European respiratory journal 37, 1037-1042.

Blomqvist, S., Roivainen, M., Puhakka, T., Kleemola, M. and Hovi, T., 2002. Virological and serological analysis of rhinovirus infections during the first two years of life in a cohort of children. J. Med. Virol. 66, 263-268.

Bochkov, Y.A., Watters, K., Ashraf, S., Griggs, T.F., Devries, M.K., Jackson, D.J., Palmenberg, A.C. and Gern, J.E., 2015. Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication. Proceedings of the National Academy of Sciences 112, 5485-5490.

Boivin, G., Coulombe, Z. and Wat, C., 2003. Quantification of the Influenza Virus Load by Real-Time Polymerase Chain Reaction in Nasopharyngeal Swabs of Patients Treated with Oseltamivir. The Journal of Infectious Diseases 188, 578-580.

Bonnelykke, K., Sleiman, P., Nielsen, K., Kreiner-Moller, E., Mercader, J.M., Belgrave, D., den Dekker, H.T., Husby, A., Sevelsted, A., Faura-Tellez, G., Mortensen, L.J., Paternoster, L., Flaaten, R., Molgaard, A., Smart, D.E., Thomsen, P.F., Rasmussen, M.A., Bonas-Guarch, S., Holst, C., Nohr, E.A., Yadav, R., March, M.E., Blicher, T.,

Page 178: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

158

Lackie, P.M., Jaddoe, V.W.V., Simpson, A., Holloway, J.W., Duijts, L., Custovic, A., Davies, D.E., Torrents, D., Gupta, R., Hollegaard, M.V., Hougaard, D.M., Hakonarson, H. and Bisgaard, H., 2014. A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat. Genet. 46, 51-55.

Bont, L., Checchia, P.A., Fauroux, B., Figueras-Aloy, J., Manzoni, P., Paes, B., Simões, E.A.F. and Carbonell-Estrany, X., 2016. Defining the Epidemiology and Burden of Severe Respiratory Syncytial Virus Infection Among Infants and Children in Western Countries. Infectious Diseases and Therapy 5, 271-298.

Bruning, A.H.L., Thomas, X.V., van der Linden, L., Wildenbeest, J.G., Minnaar, R.P., Jansen, R.R., de Jong, M.D., Sterk, P.J., van der Schee, M.P., Wolthers, K.C. and Pajkrt, D., 2015. Clinical, virological and epidemiological characteristics of rhinovirus infections in early childhood: A comparison between non-hospitalised and hospitalised children. J. Clin. Virol. 73, 120-126.

Brusselle, G. and Pavord, I., 2017. Azithromycin in uncontrolled asthma. The Lancet 390, 629-630.

Bryce, J., Boschi-Pinto, C., Shibuya, K. and Black, R., 2005. WHO estimates of the causes of death in children. Lancet 365, 1147 - 1152.

Busse, W.W., Lemanske, R.F. and Gern, J.E., 2010. Role of viral respiratory infections in asthma and asthma exacerbations. Lancet 376.

Bustin, S.A., Benes, V., Garson, J.A., Hellemans, J., Huggett, J., Kubista, M., Mueller, R., Nolan, T., Pfaffl, M.W., Shipley, G.L., Vandesompele, J. and Wittwer, C.T. 2009. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments, Clin. Chem., pp. 611-22.

Çalışkan, M., Bochkov, Y.A., Kreiner-Møller, E., Bønnelykke, K., Stein, M.M., Du, G., Bisgaard, H., Jackson, D.J., Gern, J.E., Lemanske, R.F., Nicolae, D.L. and Ober, C., 2013. Rhinovirus Wheezing Illness and Genetic Risk of Childhood-Onset Asthma. New Engl. J. Med. 368, 1398-1407.

Camargo, C.N., Carraro, E., Granato, C.F. and Bellei, N., 2012. Human rhinovirus infections in symptomatic and asymptomatic subjects. Braz. J. Microbiol. 43, 1641-5.

Chemidlin Prevost-Boure, N., Christen, R., Dequiedt, S., Mougel, C., Lelievre, M., Jolivet, C., Shahbazkia, H.R., Guillou, L., Arrouays, D. and Ranjard, L., 2011. Validation and application of a PCR primer set to quantify fungal communities in the soil environment by real-time quantitative PCR. PLoS One 6, e24166.

Cheng, V.C., Lau, S.K., Woo, P.C. and Yuen, K.Y., 2007. Severe acute respiratory syndrome coronavirus as an agent of emerging and reemerging infection. Clin. Microbiol. Rev. 20, 660-94.

Chidlow, G. 2013. The Role of Estalished and Emerging Pathogens in Respiratory Disease, Medicine and Dentistry, The University of Western Australia, Perth, Australia.

Chidlow, G.R., Harnett, G.B., Shellam, G.R. and Smith, D.W., 2009. An Economical Tandem Multiplex Real-Time PCR Technique for the Detection of a Comprehensive Range of Respiratory Pathogens. Viruses 1, 42-56.

Chidlow, G.R., Laing, I.A., Harnett, G.B., Greenhill, A.R., Phuanukoonnon, S., Siba, P.M., Pomat, W.S., Shellam, G.R., Smith, D.W. and Lehmann, D., 2012. Respiratory viral pathogens associated with lower respiratory tract disease among young children in the highlands of Papua New Guinea. J. Clin. Virol. 54-540, 235-239.

Choi, S.-H., Hong, S.-B., Kim, T., Kim, S.-H., Huh, J.W., Do, K.-H., Lee, S.-O., Kim, M.-N., Lim, C.-M., Kim, Y.S., Koh, Y., Woo, J.H., Choi, S.-H. and Sung, H., 2015. Clinical and molecular characterization of rhinoviruses A, B, and C in adult patients with pneumonia. J. Clin. Virol. 63, 70-75.

Page 179: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

159

Clarke, D.L., Davis, N.H., Majithiya, J.B., Piper, S.C., Lewis, A., Sleeman, M.A., Corkill, D.J. and May, R.D., 2014. Development of a mouse model mimicking key aspects of a viral asthma exacerbation. Clin. Sci. (Lond.) 126, 567-80.

Cohen, C., Moyes, J., Tempia, S., Groome, M., Walaza, S., Pretorius, M., Dawood, H., Chhagan, M., Haffejee, S., Variava, E., Kahn, K., von Gottberg, A., Wolter, N., Cohen, A.L., Malope-Kgokong, B., Venter, M. and Madhi, S.A., 2015a. Mortality amongst Patients with Influenza-Associated Severe Acute Respiratory Illness, South Africa, 2009-2013. PLoS One 10, e0118884.

Cohen, C., Walaza, S., Moyes, J., Groome, M., Tempia, S., Pretorius, M., Hellferscee, O., Dawood, H., Chhagan, M., Naby, F., Haffejee, S., Variava, E., Kahn, K., Nzenze, S. , Tshangela, A., von Gottberg, A., Wolter, N., Cohen, A.L., Kgokong, B., Venter, M. and Madhi, S.A., 2015b. Epidemiology of viral-associated acute lower respiratory tract infection among children <5 years of age in a high HIV prevalence setting, South Africa, 2009-2012. Pediatr. Infect. Dis. J. 34, 66-72.

Cohen, C., Walaza, S., Moyes, J., Groome, M., Tempia, S., Pretorius, M., Hellferscee, O., Dawood, H., Chhagan, M., Naby, F., Haffejee, S., Variava, E., Kahn, K., Nzenze, S., Tshangela, A., von Gottberg, A., Wolter, N., Cohen, A.L., Kgokong, B., Venter, M. and Madhi, S.A., 2015c. Epidemiology of Viral-associated Acute Lower Respiratory Tract Infection Among Children <5 Years of Age in a High HIV Prevalence Setting, South Africa, 2009–2012. Pediatr. Infect. Dis. J. 34, 66-72.

Cohen, C., Walaza, S., Moyes, J., Groome, M., Tempia, S., Pretorius, M., Hellferscee, O., Dawood, H., Haffejee, S., Variava, E., Kahn, K., Tshangela, A., von Gottberg, A., Wolter, N., Cohen, A.L., Kgokong, B., Venter, M. and Madhi, S.A., 2015d. Epidemiology of Severe Acute Respiratory Illness (SARI) among Adults and Children Aged ≥5 Years in a High HIV-Prevalence Setting, 2009–2012. PLoS One 10, e0117716.

Corne, J.M., Marshall, C., Smith, S., Schreiber, J., Sanderson, G., Holgate, S.T. and Johnston, S.L., 2002. Frequency, severity, and duration of rhinovirus infections in asthmatic and non-asthmatic individuals: a longitudinal cohort study. Lancet 359, 831-4.

Cortjens, B., Boer, O.J.d., Jong, R.d., Antonis, A.F.G., Piñeros, Y.S.S., Lutter, R., Woensel, J.B.M.v. and Bem, R.A., 2016. Neutrophil extracellular traps cause airway obstruction during respiratory syncytial virus disease. The Journal of Pathology 238, 401-411.

Coudray-Meunier, C., Fraisse, A., Martin-Latil, S., Delannoy, S., Fach, P. and Perelle, S., 2016. A Novel High-Throughput Method for Molecular Detection of Human Pathogenic Viruses Using a Nanofluidic Real-Time PCR System. PLoS One 11, e0147832.

Cox, D.W., Bizzintino, J., Ferrari, G., Khoo, S.K., Zhang, G., Whelan, S., Lee, W.M., Bochkov, Y.A., Geelhoed, G.C., Goldblatt, J., Gern, J.E., Laing, I.A. and Le Souëf, P.N., 2013. Human Rhinovirus Species C Infection in Young Children with Acute Wheeze Is Associated with Increased Acute Respiratory Hospital Admissions. Am. J. Respir. Crit. Care Med. 188, 1358-1364.

Culley, F.J., Pennycook, A.M., Tregoning, J.S., Hussell, T. and Openshaw, P.J., 2006. Differential chemokine expression following respiratory virus infection reflects Th1- or Th2-biased immunopathology. J. Virol. 80, 4521-7.

D’Amato, G., Stanziola, A., Sanduzzi, A., Liccardi, G., Salzillo, A., Vitale, C., Molino, A., Vatrella, A. and D’Amato, M., 2014. Treating severe allergic asthma with anti -IgE monoclonal antibody (omalizumab): a review. Multidisciplinary Respiratory Medicine 9, 1-6.

de Groot, R.J., Baker, S.C., Baric, R.S., Brown, C.S., Drosten, C., Enjuanes, L., Fouchier, R.A., Galiano, M., Gorbalenya, A.E., Memish, Z.A., Perlman, S., Poon, L.L., Snijder, E.J.,

Page 180: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

160

Stephens, G.M., Woo, P.C., Zaki, A.M., Zambon, M. and Ziebuhr, J., 2013. Middle East respiratory syndrome coronavirus (MERS-CoV): announcement of the Coronavirus Study Group. J. Virol. 87, 7790-2.

Demian, P., Horton, K., Kajon, A., Siam, R., Hasanin, A.M., Elgohary Sheta, A., Cornelius, C. and Gaynor, A., 2014. Molecular identification of adenoviruses associated with respiratory infection in Egypt from 2003 to 2010. BMC Infect. Dis. 14, 50.

Denlinger, L.C., Sorkness, R.L., Lee, W.-M., Evans, M.D., Wolff, M.J., Mathur, S.K., Crisafi, G.M., Gaworski, K.L., Pappas, T.E., Vrtis, R.F., Kelly, E.A., Gern, J.E. and Jarjour, N.N., 2011. Lower Airway Rhinovirus Burden and the Seasonal Risk of Asthma Exacerbation. Am. J. Respir. Crit. Care Med. 184, 1007-1014.

Denny Jr., F.W., ed, 2001. The Clinical Impact of Human Respiratory Vi rus Infections. Marcel Dekker, New York.

DeVincenzo, J.P., Wilkinson, T., Vaishnaw, A., Cehelsky, J., Meyers, R., Nochur, S., Harrison, L., Meeking, P., Mann, A., Moane, E., Oxford, J., Pareek, R., Moore, R., Walsh, E., Studholme, R., Dorsett, P., Alvarez, R. and Lambkin-Williams, R., 2010. Viral load drives disease in humans experimentally infected with respiratory syncytial virus. Am. J. Respir. Crit. Care Med. 182, 1305-14.

Dijkman, R., Jebbink, M.F., Koekkoek, S.M., Deijs, M., Jonsdottir, H.R., Molenkamp, R., Ieven, M., Goossens, H., Thiel, V. and van der Hoek, L., 2013. Isolation and characterization of current human coronavirus strains in primary human epithelial cell cultures reveal differences in target cell tropism. J. Virol. 87, 6081-90.

Do, D.H., Laus, S., Leber, A., Marcon, M.J., Jordan, J.A., Martin, J.M. and Wadowsky, R.M., 2010. A one-step, real-time PCR assay for rapid detection of rhinovirus. J. Mol. Diagn. 12, 102-8.

Dobson, J., Whitley, R.J., Pocock, S. and Monto, A.S., 2015. Oseltamivir treatment for influenza in adults: a meta-analysis of randomised controlled trials. Lancet 385, 1729-1737.

Drews, A.L., Atmar, R.L., Glezen, W.P., Baxter, B.D., Piedra, P.A. and Greenberg, S.B., 1997. Dual respiratory virus infections. Clin. Infect. Dis. 25, 1421-9.

Durrani, S.R., Montville, D.J., Pratt, A.S., Sahu, S., DeVries, M.K., Rajamanickam, V., Gangnon, R.E., Gill, M.A., Gern, J.E., Lemanske, R.F., Jr. and Jackson, D.J., 2012. Innate immune responses to rhinovirus are reduced by the high-affinity IgE receptor in allergic asthmatic children. J. Allergy Clin. Immunol. 130, 489-95.

El Saleeby, C.M., Bush, A.J., Harrison, L.M., Aitken, J.A. and Devincenzo, J.P., 2011. Respiratory syncytial virus load, viral dynamics, and disease severity in previously healthy naturally infected children. J. Infect. Dis. 204, 996-1002.

Englund, J.A., Anderson, L.J. and Rhame, F.S., 1991. Nosocomial transmission of respiratory syncytial virus in immunocompromised adults. J. Clin. Microbiol. 29, 115-119.

Ewijk, B.E., Wolfs, T.F., Aerts, P.C., Kessel, K.P., Fleer, A., Kimpen, J.L. and Ent, C.K., 2007. RSV mediates Pseudomonas aeruginosa binding to cystic fibrosis and normal epithelial cells. Pediatr. Res. 61.

Fahy, J.V., 2009. Eosinophilic and Neutrophilic Inflammation in Asthma. Proc. Am. Thorac. Soc. 6, 256-259.

Fahy, J.V., Kim, K.W., Liu, J. and Boushey, H.A., 1995. Prominent neutrophilic inflammation in sputum from subjects with asthma exacerbation. J. Allergy Clin. Immunol. 95, 843-52.

Falsey, A.R., Erdman, D., Anderson, L.J. and Walsh, E.E., 2003. Human metapneumovirus infections in young and elderly adults. J. Infect. Dis. 187, 785-90.

Falsey, A.R., Formica, M.A. and Walsh, E.E., 2012. Yield of Sputum for Viral Detection by Reverse Transcriptase PCR in Adults Hospitalized with Respiratory Illness. J. Clin. Microbiol. 50, 21-24.

Page 181: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

161

Faure, E., Poissy, J., Goffard, A., Fournier, C., Kipnis, E., Titecat, M., Bortolotti, P., Martinez, L., Dubucquoi, S., Dessein, R., Gosset, P., Mathieu, D. and Guery, B., 2014. Distinct Immune Response in Two MERS-CoV-Infected Patients: Can We Go from Bench to Bedside? PLoS One 9, e88716.

Feikin, D.R., Alraddadi, B., Qutub, M., Shabouni, O., Curns, A., Oboho, I.K., Tomczyk, S.M., Wolff, B., Watson, J.T. and Madani, T.A., 2015. Association of Higher MERS-CoV Virus Load with Severe Disease and Death, Saudi Arabia, 2014. Emerging Infect. Dis. 21, 2029-2035.

Fiore, A.E., Bridges, C.B. and Cox, N.J., 2009. Seasonal influenza vaccines. Curr. Top. Microbiol. Immunol. 333, 43-82.

Fitzpatrick, A.M., Holguin, F., Teague, W.G. and Brown, L.A.S., 2008. Alveolar macrophage phagocytosis is impaired in children with poorly controlled asthma. The Journal of allergy and clinical immunology 121, 1372-1378.e3.

Fleming, D.M., Pannell, R.S. and Cross, K.W., 2005. Mortality in children from influenza and respiratory syncytial virus. J. Epidemiol. Community Health 59, 586-590.

Fodha, I., Vabret, A., Ghedira, L., Seboui, H., Chouchane, S., Dewar, J., Gueddiche, N., Trabelsi, A., Boujaafar, N. and Freymuth, F., 2007. Respiratory syncytial virus infections in hospitalized infants: Association between viral load, virus subgroup, and disease severity. J. Med. Virol. 79, 1951-1958.

Foulongne, V., Guyon, G., Rodiere, M. and Segondy, M., 2006. Human metapneumovirus infection in young children hospitalized with respiratory tract disease. Pediatr. Infect. Dis. J. 25, 354-9.

Franz, A., Adams, O., Willems, R., Bonzel, L., Neuhausen, N., Schweizer-Krantz, S., Ruggeberg, J.U., Willers, R., Henrich, B., Schroten, H. and Tenenbaum, T., 2010. Correlation of viral load of respiratory pathogens and co-infections with disease severity in children hospitalized for lower respiratory tract infection. J. Clin. Virol. 48, 239-245.

Gaunt, E.R., Hardie, A., Claas, E.C., Simmonds, P. and Templeton, K.E., 2010. Epidemiology and clinical presentations of the four human coronaviruses 229E, HKU1, NL63, and OC43 detected over 3 years using a novel multiplex real-time PCR method. J. Clin. Microbiol. 48, 2940-7.

Gern, J.E. and Busse, W.W., 2002. Relationship of viral infections to wheezing illnesses and asthma. Nat. Rev. Immunol. 2, 132-8.

Gern, J.E., Vrtis, R., Grindle, K.A., Swenson, C. and Busse, W.W., 2000. Relationship of Upper and Lower Airway Cytokines to Outcome of Experimental Rhinovirus Infection. Am. J. Respir. Crit. Care Med. 162, 2226-2231.

Gibson, P.G., Yang, I.A., Upham, J.W., Reynolds, P.N., Hodge, S., James, A.L., Jenkins, C., Peters, M.J., Marks, G.B., Baraket, M., Powell, H., Taylor, S.L., Leong, L.E.X., Rogers, G.B. and Simpson, J.L., 2017. Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): a randomised, double-blind, placebo-controlled trial. The Lancet 390, 659-668.

Gielen, V., Johnston, S.L. and Edwards, M.R., 2010. Azithromycin induces anti-viral responses in bronchial epithelial cells. Eur. Respir. J. 36, 646-54.

Gill, M.A., Bajwa, G., George, T.A., Dong, C.C., Dougherty, II, Jiang, N., Gan, V.N. and Gruchalla, R.S., 2010. Counterregulation between the FcepsilonRI pathway and antiviral responses in human plasmacytoid dendritic cells. J. Immunol. 184, 5999-6006.

GINA, 2017. Global Initiate for Asthma. Global Strategy for Asthma Management and Prevention NHLBI/WHO Workshop Report, Bethesda, MD.

Page 182: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

162

Gomez, S., Prieto, C., Vera, C., J, R.O. and Folgueira, L., 2016. Evaluation of a new rapid diagnostic test for the detection of influenza and RSV. Enferm. Infecc. Microbiol. Clin. 34, 298-302.

Gommerman, J. and Ng, D., 2013. The regulation of immune responses by DC derived Type I IFN. Front. Immunol. 4.

Graham, S.M. and Gibb, D.M., 2002. HIV disease and respiratory infection in children. Br. Med. Bull. 61, 133-50.

Granados, A., Luinstra, K., Chong, S., Goodall, E., Banh, L., Mubareka, S., Smieja, M. and Mahony, J., 2012. Use of an improved quantitative polymerase chain reaction assay to determine differences in human rhinovirus viral loads in different populations. Diagn. Microbiol. Infect. Dis. 74, 384-387.

Grant, L.R., Hammitt, L.L., Murdoch, D.R., O’Brien, K.L. and Scott, J.A., 2012. Procedures for Collection of Induced Sputum Specimens From Children. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America 54, S140-S145.

Gray, D.M. and Zar, H.J., 2010. Community-acquired pneumonia in HIV-infected children: a global perspective. Curr. Opin. Pulm. Med. 16, 208-16.

Greensill, J., McNamara, P.S., Dove, W., Flanagan, B., Smyth, R.L. and Hart, C.A., 2003. Human Metapneumovirus in Severe Respiratory Syncytial Virus Bronchiolitis. Emerging Infect. Dis. 9, 372-375.

Gueudin, M., Vabret, A., Petitjean, J., Gouarin, S., Brouard, J. and Freymuth, F., 2003. Quantitation of respiratory syncytial virus RNA in nasal aspirates of children by real-time RT-PCR assay. J. Virol. Methods 109, 39-45.

Guilbert, T.W., Singh, A.M., Danov, Z., Evans, M.D., Jackson, D.J., Burton, R., Roberg, K.A., Anderson, E.L., Pappas, T.E., Gangnon, R., Gern, J.E. and Lemanske, R.F., Jr., 2011. Decreased lung function after preschool wheezing rhinovirus illnesses in children at risk to develop asthma. J. Allergy Clin. Immunol. 128, 532-8.e1-10.

Guzmán-Suarez, B.B., Buckley, M.W., Gilmore, E.T., Vocca, E., Moss, R., Marty, F.M., Sanders, R., Baden, L.R., Wurtman, D., Issa, N.C., Fang, F. and Koo, S., 2012. Clinical potential of DAS181 for treatment of parainfluenza-3 infections in transplant recipients. Transpl. Infect. Dis. 14, 427-433.

Hall Sedlak, R. and Jerome, K.R., 2014. The potential advantages of digital PCR for clinical virology diagnostics. Expert Rev. Mol. Diagn. 14, 501-7.

Han, Y., Ling, M.T., Mao, H., Zheng, J., Liu, M., Lam, K.T., Liu, Y., Tu, W. and Lau, Y.-L., 2014. Influenza Virus-Induced Lung Inflammation Was Modulated by Cigarette Smoke Exposure in Mice. PLoS One 9, e86166.

Hansel, T.T., Tunstall, T., Trujillo-Torralbo, M.-B., Shamji, B., del-Rosario, A., Dhariwal, J., Kirk, P.D.W., Stumpf, M.P.H., Koopmann, J., Telcian, A., Aniscenko, J., Gogsadze, L., Bakhsoliani, E., Stanciu, L., Bartlett, N., Edwards, M., Walton, R., Mallia, P., Hunt, T.M., Hunt, T.L., Hunt, D.G., Westwick, J., Edwards, M., Kon, O.M., Jackson, D.J. and Johnston, S.L., 2017. A Comprehensive Evaluation of Nasal and Bronchial Cytokines and Chemokines Following Experimental Rhinovirus Infection in Allergic Asthma: Increased Interferons (IFN-γ and IFN-λ) and Type 2 Inflammation (IL-5 and IL-13). EBioMedicine 19, 128-138.

Hasegawa, K., Mansbach, J.M., Teach, S.J., Fisher, E.S., Hershey, D., Koh, J.Y., Clark, S., Piedra, P.A., Sullivan, A.F. and Camargo, C.A., 2014. Multicenter Study of Viral Etiology and Relapse in Hospitalized Children with Bronchiolitis. The Pediatric infectious disease journal 33, 809-813.

Hayden, R.T., Gu, Z., Ingersoll, J., Abdul-Ali, D., Shi, L., Pounds, S. and Caliendo, A.M., 2013. Comparison of droplet digital PCR to real-time PCR for quantitative detection of cytomegalovirus. J. Clin. Microbiol. 51, 540-6.

Page 183: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

163

Hayden, R.T., Yan, X., Wick, M.T., Rodriguez, A.B., Xiong, X., Ginocchio, C.C., Mitchell, M.J. and Caliendo, A.M., 2012. Factors Contributing to Variability of Quantitative Viral PCR Results in Proficiency Testing Samples: a Multivariate Analysis. J. Clin. Microbiol. 50, 337-345.

Heikkinen, T., Marttila, J., Salmi, A.A. and Ruuskanen, O., 2002. Nasal Swab versus Nasopharyngeal Aspirate for Isolation of Respiratory Viruses. J. Clin. Microbiol. 40, 4337-4339.

Henderson, J., Hilliard, T.N., Sherriff, A., Stalker, D., Al Shammari, N. and Thomas, H.M., 2005. Hospitalization for RSV bronchiolitis before 12 months of age and subsequent asthma, atopy and wheeze: a longitudinal birth cohort study. Pediatr. Allergy Immunol. 16, 386-92.

Heymann, P.W., Carper, H.T., Murphy, D.D., Platts-Mills, T.A., Patrie, J., McLaughlin, A.P., Erwin, E.A., Shaker, M.S., Hellems, M., Peerzada, J., Hayden, F.G., Hatley, T.K. and Chamberlain, R., 2004. Viral infections in relation to age, atopy, and season of admission among children hospitalized for wheezing. J. Allergy Clin. Immunol. 114, 239-47.

Hirsh, S., Hindiyeh, M., Kolet, L., Regev, L., Sherbany, H., Yaary, K., Mendelson, E. and Mandelboim, M., 2014. Epidemiological Changes of Respiratory Syncytial Virus (RSV) Infections in Israel. PLoS One 9, e90515.

Hoffman, N.G., Cook, L., Atienza, E.E., Limaye, A.P. and Jerome, K.R., 2008. Marked Variability of BK Virus Load Measurement Using Quantitative Real-Time PCR among Commonly Used Assays. J. Clin. Microbiol. 46, 2671-2680.

Holgate, S., Casale, T., Wenzel, S., Bousquet, J., Deniz, Y. and Reisner, C., 2005. The anti -inflammatory effects of omalizumab confirm the central role of IgE in allergic inflammation. J. Allergy Clin. Immunol. 115.

Holt, P.G., Rowe, J., Kusel, M., Parsons, F., Hollams, E.M., Bosco, A., McKenna, K., Subrata, L., de Klerk, N., Serralha, M., Holt, B.J., Zhang, G., Loh, R., Ahlstedt, S. and Sly, P.D., 2010. Toward improved prediction of risk for atopy and asthma among preschoolers: a prospective cohort study. J. Allergy Clin. Immunol. 125, 653-9, 659 e1-659 e7.

Hornsleth, A., Klug, B., Nir, M., Johansen, J., Hansen, K.S., Christensen, L.S. and Larsen, L.B., 1998. Severity of respiratory syncytial virus disease related to type and genotype of virus and to cytokine values in nasopharyngeal secretions. Pediatr. Infect. Dis. J. 17, 1114-21.

Hosseini, S.S., Ghasemian, E., Jamaati, H., Tabaraie, B., Amini, Z. and Cox, K., 2015. Association between respiratory viruses and exacerbation of COPD: a case–control study. Infect Dis (Lond) 47.

Houben, M.L., Coenjaerts, F.E., Rossen, J.W., Belderbos, M.E., Hofland, R.W., Kimpen, J.L. and Bont, L., 2010. Disease severity and viral load are correlated in infants with primary respiratory syncytial virus infection in the community. J. Med. Virol. 82, 1266-71.

Hu, B., Ge, X., Wang, L.-F. and Shi, Z., 2015. Bat origin of human coronaviruses. Virol J. 12, 221.

Huggett, J.F., Cowen, S. and Foy, C.A., 2015. Considerations for digi tal PCR as an accurate molecular diagnostic tool. Clin. Chem. 61, 79-88.

Huggett, J.F., Foy, C.A., Benes, V., Emslie, K., Garson, J.A., Haynes, R., Hellemans, J., Kubista, M., Mueller, R.D., Nolan, T., Pfaffl, M.W., Shipley, G.L., Vandesompele, J., Wittwer, C.T. and Bustin, S.A., 2013. The digital MIQE guidelines: Minimum Information for Publication of Quantitative Digital PCR Experiments. Clin. Chem. 59, 892-902.

Page 184: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

164

Hung, I.F.N., Cheng, V.C.C., Wu, A.K.L., Tang, B.S.F., Chan, K.H., Chu, C.M., Wong, M.M.L., Hui, W.T., Poon, L.L.M., Tse, D.M.W., Chan, K.S., Woo, P.C.Y., Lau, S.K.P., Peiris, J.S.M. and Yuen, K.Y., 2004. Viral Loads in Clinical Specimens and SARS Manifestations. Emerging Infect. Dis. 10, 1550-1557.

Huynh, M.-L.N., Malcolm, K.C., Kotaru, C., Tilstra, J.A., Westcott, J.Y., Fadok, V.A. and Wenzel, S.E., 2005. Defective Apoptotic Cell Phagocytosis Attenuates Prostaglandin E2 and 15-Hydroxyeicosatetraenoic Acid in Severe Asthma Alveolar Macrophages. Am. J. Respir. Crit. Care Med. 172, 972-979.

Iuliano, A.D., Roguski, K.M., Chang, H.H., Muscatello, D.J., Palekar, R., Tempia, S., Cohen, C., Gran, J.M., Schanzer, D., Cowling, B.J., Wu, P., Kyncl, J., Ang, L.W., Park, M., Redlberger-Fritz, M., Yu, H., Espenhain, L., Krishnan, A., Emukule, G., van Asten, L., Pereira da Silva, S., Aungkulanon, S., Buchholz, U., Widdowson, M.-A. and Bresee, J.S., 2018. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. The Lancet 391, 1285-1300.

Iwane, M.K., Prill, M.M., Lu, X., Miller, E.K., Edwards, K.M., Hall, C.B., Griffin, M.R., Staat, M.A., Anderson, L.J., Williams, J.V., Weinberg, G.A., Ali, A., Szilagyi, P.G., Zhu, Y. and Erdman, D.D., 2011. Human rhinovirus species associated with hospitalizations for acute respiratory illness in young US children. J. Infect. Dis. 204, 1702-10.

Jackson, D.J., Gangnon, R.E., Evans, M.D., Roberg, K.A., Anderson, E.L., Pappas, T.E., Printz, M.C., Lee, W.-M., Shult, P.A., Reisdorf, E., Carlson-Dakes, K.T., Salazar, L.P., DaSilva, D.F., Tisler, C.J., Gern, J.E. and Lemanske, R.F., 2008a. Wheezing Rhinovirus Illnesses in Early Life Predict Asthma Development in High-Risk Children. Am. J. Respir. Crit. Care Med. 178, 667-672.

Jackson, D.J., Gangnon, R.E., Evans, M.D., Roberg, K.A., Anderson, E.L., Pappas, T.E., Printz, M.C., Lee, W.M., Shult, P.A., Reisdorf, E., Carlson-Dakes, K.T., Salazar, L.P., DaSilva, D.F., Tisler, C.J., Gern, J.E. and Lemanske, R.F., Jr., 2008b. Wheezing rhinovirus illnesses in early life predict asthma development in high-risk children. Am. J. Respir. Crit. Care Med. 178, 667-72.

Jafri, H.S., Wu, X., Makari, D. and Henrickson, K.J., 2013. Distribution of respiratory syncytial virus subtypes A and B among infants presenting to the emergency department with lower respiratory tract infection or apnea. Pediatr. Infect. Dis. J. 32, 335-40.

Jansen, R.R., Schinkel, J., Dek, I., Koekkoek, S.M., Visser, C.E., de Jong, M.D., Molenkamp, R. and Pajkrt, D., 2010. Quantitation of respiratory viruses in relation to clinical course in children with acute respiratory tract infections. Pediatric Infectious Diseases Journal 29, 82-4.

Jartti, T., Hasegawa, K., Mansbach, J.M., Piedra, P.A. and Camargo, C.A., Jr., 2015. Rhinovirus-induced bronchiolitis: Lack of association between virus genomic load and short-term outcomes. J. Allergy Clin. Immunol. 136, 509-12.e11.

Jartti, T., Jartti, L., Peltola, V., Waris, M. and Ruuskanen, O., 2008. Identification of respiratory viruses in asymptomatic subjects: asymptomatic respiratory viral infections. Pediatr. Infect. Dis. J. 27, 1103-7.

Jartti, T., Soderlund-Venermo, M., Hedman, K., Ruuskanen, O. and Makela, M.J., 2013. New molecular virus detection methods and their clinical value in lower respiratory tract infections in children. Paediatr. Respir. Rev. 14, 38-45.

Jefferson, T., Jones, M., Doshi, P., Spencer, E.A., Onakpoya, I. and Heneghan, C.J., 2014a. Oseltamivir for influenza in adults and children: systematic review of clinical study reports and summary of regulatory comments. BMJ : British Medical Journal 348.

Jefferson, T., Jones, M.A., Doshi, P., Del Mar, C.B., Hama, R., Thompson, M.J., Spencer, E.A., Onakpoya, I., Mahtani, K.R., Nunan, D., Howick, J. and Heneghan, C.J., 2014b.

Page 185: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

165

Neuraminidase inhibitors for preventing and treating influenza in healthy adults and children. Cochrane Database Syst Rev, Cd008965.

Jin, W. and Dong, C., 2013. IL-17 cytokines in immunity and inflammation. Emerg Microbes Infect 2, e60.

Johnson, J.E., Gonzales, R.A., Olson, S.J., Wright, P.F. and Graham, B.S., 2006. The histopathology of fatal untreated human respiratory syncytial virus infection. Mod. Pathol. 20, 108-119.

Johnston, N.W., Johnston, S.L., Duncan, J.M., Greene, J.M., Kebadze, T., Keith, P.K., Roy, M., Waserman, S. and Sears, M.R., 2005a. The September epidemic of asthma exacerbations in children: a search for etiology. J. Allergy Clin. Immunol. 115, 132-8.

Johnston, N.W., Johnston, S.L., Duncan, J.M., Greene, J.M., Kebadze, T., Keith, P.K., Roy, M., Waserman, S. and Sears, M.R., 2005b. The September epidemic of asthma exacerbations in children: a search for etiology. J. Allergy Clin. Immunol. 115.

Kahn, J.S., 2003. Human metapneumovirus: a newly emerging respiratory pathogen. Curr. Opin. Infect. Dis. 16, 255-8.

Kau, A.L. and Korenblat, P.E., 2014. Anti-Interleukin 4 and 13 for Asthma Treatment in the Era of Endotypes. Curr. Opin. Allergy Clin. Immunol. 14, 570-575.

Kawano, H., Kayama, H., Nakama, T., Hashimoto, T., Umemoto, E. and Takeda, K., 2016. IL-10-producing lung interstitial macrophages prevent neutrophilic asthma. Int. Immunol. 28, 489-501.

Kearley, J., Barker, J.E., Robinson, D.S. and Lloyd, C.M., 2005. Resolution of airway inflammation and hyperreactivity after in vivo transfer of CD4 regulatory T cells is interleukin 10 dependent. The Journal of Experimental Medicine 202, 1539.

Kearley, J., Erjefalt, J.S., Andersson, C., Benjamin, E., Jones, C.P., Robichaud, A., Pegorier, S., Brewah, Y., Burwell, T.J., Bjermer, L., Kiener, P.A., Kolbeck, R., Lloyd, C.M., Coyle, A.J. and Humbles, A.A., 2011. IL-9 governs allergen-induced mast cell numbers in the lung and chronic remodeling of the airways. Am. J. Respir. Crit. Care Med. 183, 865-75.

Kennedy, J.L., Shaker, M., McMeen, V., Gern, J., Carper, H., Murphy, D., Lee, W.M., Bochkov, Y.A., Vrtis, R.F., Platts-Mills, T., Patrie, J., Borish, L., Steinke, J.W., Woods, W.A. and Heymann, P.W., 2014. Comparison of viral load in individuals with and without asthma during infections with rhinovirus. Am. J. Respir. Crit. Care Med. 189, 532-9.

Khetsuriani, N., Lu, X., Teague, W., Kazerouni, N., Anderson, L. and Erdman, D., 2008. Novel human rhinoviruses and exacerbation of asthma in children. Emerg. Infect. Dis. 14, 1793 - 1796.

Kim, M.R., Lee, H.R. and Lee, G.M., 2000. Epidemiology of acute viral respiratory tract infections in Korean children. J. Infect. 41, 152-8.

Kim, T.H. and Lee, H.K., 2014. Innate immune recognition of respiratory syncytial virus infection. BMB Rep.

Kluijver, J.D., Grünberg, K., Pons, D., Klerk, E.P.A.D., Dick, C.R., Sterk, P.J. and Hiemstra, P.S., 2003. Interleukin‐1β and interleukin‐1ra levels in nasal lavages during experimental rhinovirus infection in asthmatic and non‐asthmatic subjects. Clin. Exp. Allergy 33, 1415-1418.

Lambert, S.B., Whiley, D.M., O'Neill, N.T., Andrews, E.C., Canavan, F.M., Bletchly, C., Siebert, D.J., Sloots, T.P. and Nissen, M.D., 2008. Comparing Nose-Throat Swabs and Nasopharyngeal Aspirates Collected From Children With Symptoms for Respiratory Virus Identification Using Real-Time Polymerase Chain Reaction. Pediatrics 122, e615-e620.

Page 186: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

166

Lanaspa, M., O'Callaghan-Gordo, C., Machevo, S., Madrid, L., Nhampossa, T., Acacio, S., de la Horra, C., Friaza, V., Campano, E., Alonso, P.L., Calderon, E.J., Roca, A. and Bassat, Q., 2015. High prevalence of Pneumocystis jirovecii pneumonia among Mozambican children <5 years of age admitted to hospital with clinical severe pneumonia. Clin. Microbiol. Infect. 21, 1018.e9-1018.e15.

Lau, S.K., Li, K.S., Tsang, A.K., Lam, C.S., Ahmed, S. and Chen, H., 2013. Genetic characterization of Betacoronavirus lineage C viruses in bats reveals marked sequence divergence in the spike protein of pipistrellus bat coronavirus HKU5 in Japanese pipistrelle: Implications for the origin of the novel Middle East respiratory syndrome coronavirus. J. Virol. 87.

Lee, N., Wong, C.K., Chan, M.C.W., Yeung, E.S.L., Tam, W.W.S., Tsang, O.T.Y., Choi, K.W., Chan, P.K.S., Kwok, A., Lui, G.C.Y., Leung, W.S., Yung, I.M.H., Wong, R.Y.K., Cheung, C.S.K. and Hui, D.S.C., 2017. Anti-inflammatory effects of adjunctive macrolide treatment in adults hospitalized with influenza: A randomized controlled trial. Antiviral Res. 144, 48-56.

Lee, S., Nguyen, M.T., Currier, M.G., Jenkins, J.B., Strobert, E.A., Kajon, A.E., Madan-Lala, R., Bochkov, Y.A., Gern, J.E., Roy, K., Lu, X., Erdman, D.D., Spearman, P. and Moore, M.L., 2016. A polyvalent inactivated rhinovirus vaccine is broadly immunogenic in rhesus macaques. Nature Communications 7, 12838.

Lee, W.-M., Lemanske, R.F., Evans, M.D., Vang, F., Pappas, T., Gangnon, R., Jackson, D.J. and Gern, J.E., 2012a. Human Rhinovirus Species and Season of Infection Determine Illness Severity. American Journal of Respiratory and Critical Care Medicine 186, 886-891.

Lee, W.-M., Lemanske, R.F., Evans, M.D., Vang, F., Pappas, T., Gangnon, R., Jackson, D.J. and Gern, J.E., 2012b. Human Rhinovirus Species and Season of Infection Determine Illness Severity. Am. J. Respir. Crit. Care Med. 186, 886-891.

Lee, W.M., Kiesner, C., Pappas, T., Lee, I., Grindle, K., Jartti, T., Jakiela, B., Lemanske, R.F., Jr., Shult, P.A. and Gern, J.E., 2007. A diverse group of previously unrecognized human rhinoviruses are common causes of respiratory illnesses in infants. PLoS One 2, e966.

Leland, D.S. and Ginocchio, C.C., 2007. Role of Cell Culture for Virus Detection in the Age of Technology. Clin. Microbiol. Rev. 20, 49-78.

Lemanske, R.F., Jackson, D.J., Gangnon, R.E., Evans, M.D., Li, Z., Shult, P.A., Kirk, C.J., Reisdorf, E., Roberg, K.A., Anderson, E.L., Carlson-Dakes, K.T., Adler, K.J., Gilbertson-White, S., Pappas, T.E., DaSilva, D.F., Tisler, C.J. and Gern, J.E., 2005. Rhinovirus illnesses during infancy predict subsequent childhood wheezing. J. Allergy Clin. Immunol. 116, 571-577.

Letowski, J., Brousseau, R. and Masson, L., 2004. Designing better probes: effect of probe size, mismatch position and number on hybridization in DNA oligonucleotide microarrays. Journal of Microbiology Methods 57, 269-78.

Leung, T.F., Chan, P.K., Wong, G.W., Fok, T.F. and Ng, P.C., 2013. Respiratory viruses and atypical bacteria triggering severe asthma exacerbation in children. Hong Kong Med. J. 19 Suppl 4, 11-4.

Levesque-Sergerie, J.-P., Duquette, M., Thibault, C., Delbecchi, L. and Bissonnette, N., 2007. Detection limits of several commercial reverse transcriptase enzymes: impact on the low- and high-abundance transcript levels assessed by quantitative RT-PCR. BMC Mol. Biol. 8, 1-18.

Li, C.C., Wang, L., Eng, H.L., You, H.L., Chang, L.S., Tang, K.S., Lin, Y.J., Kuo, H.C., Lee, I.K., Liu, J.W., Huang, E.Y. and Yang, K.D., 2010. Correlation of pandemic (H1N1) 2009 viral load with disease severity and prolonged viral shedding in children. Emerging Infect. Dis. 16, 1265-72.

Page 187: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

167

Li, H.-N., Barlow, P.G., Bylund, J., Mackellar, A., Björstad, Å., Conlon, J., Hiemstra, P.S., Haslett, C., Gray, M., Simpson, A.J., Rossi, A.G. and Davidson, D.J., 2009. Secondary necrosis of apoptotic neutrophils induced by the human cathelicidin LL-37 is not proinflammatory to phagocytosing macrophages. J. Leukocyte Biol. 86, 891-902.

Lim, S., Crawley, E., Woo, P. and Barnes, P.J., 1998. Haplotype associated with low interleukin-10 production in patients with severe asthma. The Lancet 352, 113.

Linden, A., 2001. Role of interleukin-17 and the neutrophil in asthma. Int. Arch. Allergy Immunol. 126, 179-84.

Linsuwanon, P., Payungporn, S., Samransamruajkit, R., Posuwan, N., Makkoch, J., Theanboonlers, A. and Poovorawan, Y., 2009. High prevalence of human rhinovirus C infection in Thai children with acute lower respiratory tract disease. J. Infect. 59, 115-121.

Liu, Y., Hill, M.G., Klose, T., Chen, Z., Watters, K., Bochkov, Y.A., Jiang, W., Palmenberg, A.C. and Rossmann, M.G., 2016. Atomic structure of a rhinovirus C, a virus species linked to severe childhood asthma. Proceedings of the National Academy of Sciences 113, 8997-9002.

Liu, Y., Li, S., Zhang, G., Nie, G., Meng, Z., Mao, D., Chen, C., Chen, X., Zhou, B. and Zeng, G., 2013. Genetic variants in IL1A and IL1B contribute to the susceptibility to 2009 pandemic H1N1 influenza A virus. BMC Immunol. 14, 37-37.

Louis, R., Lau, L.C., Bron, A.O., Roldaan, A.C., Radermecker, M. and Djukanovic, R., 2000. The relationship between airways inflammation and asthma severity. Am. J. Respir. Crit. Care Med. 161, 9-16.

Lu, R., Yu, X., Wang, W., Duan, X., Zhang, L., Zhou, W., Xu, J., Xu, L., Hu, Q., Lu, J., Ruan, L., Wang, Z. and Tan, W., 2012. Characterization of Human Coronavirus Etiology in Chinese Adults with Acute Upper Respiratory Tract Infection by Real -Time RT-PCR Assays. PLoS One 7, e38638.

Luchsinger, V., Ampuero, S., Palomino, M.A., Chnaiderman, J., Levican, J., Gaggero, A. and Larranaga, C.E., 2014. Comparison of virological profiles of respiratory syncytial virus and rhinovirus in acute lower tract respiratory infections in very young Chilean infants, according to their clinical outcome. J. Clin. Virol. 61, 138-44.

Lukšić, I., Kearns, P.K., Scott, F., Rudan, I., Campbell, H. and Nair, H., 2013. Viral etiology of hospitalized acute lower respiratory infections in children under 5 years of age – a systematic review and meta-analysis. Croat. Med. J. 54, 122-134.

Lynch, J.P., 3rd, Fishbein, M. and Echavarria, M., 2011. Adenovirus. Semin. Respir. Crit. Care Med. 32, 494-511.

Machado, E.R., Carlos, D., Lourenco, E.V., Sorgi, C.A., Silva, E.V., Ramos, S.G., Ueta, M.T., Aronoff, D.M. and Faccioli, L.H., 2009. Counterregulation of Th2 immunity by interleukin 12 reduces host defenses against Strongyloides venezuelensis infection. Microbes Infect 11, 571-8.

Madhi, S.A., Schoub, B., Simmank, K., Blackburn, N. and Klugman, K.P., 2000. Increased burden of respiratory viral associated severe lower respiratory tract infections in children infected with human immunodeficiency virus type-1. The Journal of Pediatrics 137, 78-84.

Mahmutovic Persson, I., Akbarshahi, H., Menzel, M., Brandelius, A. and Uller, L., 2016. Increased expression of upstream TH2-cytokines in a mouse model of viral-induced asthma exacerbation. J. Transl. Med. 14, 52.

Mäkelä, M.J., Kanehiro, A., Borish, L., Dakhama, A., Loader, J., Joetham, A., Xing, Z., Jordana, M., Larsen, G.L. and Gelfand, E.W., 2000. IL-10 is necessary for the expression of airway hyperresponsiveness but not pulmonary inflammation after allergic sensitization. Proc. Natl. Acad. Sci. U. S. A. 97, 6007-6012.

Page 188: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

168

Malmgaard, L., 2004. Induction and regulation of IFNs during viral infections. J. Interferon Cytokine Res. 24, 439-54.

Marais, B.J., Rabie, H., Schaaf, S.H. and Cotton, M.F., 2006. Common opportunistic infections in HIV infected infants and children Part 1—respiratory infections. South African Family Practice 48, 52-56.

Markey, A.L., Mohr, S. and Day, P.J., 2010. High-throughput droplet PCR. Methods 50, 277-81.

Martin, E.T., Kuypers, J., Wald, A. and Englund, J.A., 2012. Multiple versus single virus respiratory infections: viral load and clinical disease severity in hospitalized children. Influenza Other Respir Viruses 6, 71-7.

Matsumoto, K. and Inoue, H., 2014. Viral infections in asthma and COPD. Respir Investig 52, 92-100.

Matsushita, M., Takeuchi, S., Kumagai, N., Morio, M., Matsushita, C., Arise, K. and Awatani, T., 2017. Booster influenza vaccination confers additional immune responses in an elderly, rural community-dwelling population. Am. J. Infect. Control.

Matu, M., Kikuvi, G., Wanzala, P., Karama, M. and Symekher, S., 2014. Aetiology of Acute Respiratory Infections in Children under Five Years in Nakuru, Kenya. Journal of Microbiology & Experimentation 1.

McConnochie, K.M., Hall, C.B., Walsh, E.E. and Roghmann, K.J., 1990. Variation in severity of respiratory syncytial virus infections with subtype. J. Pediatr. 117, 52-62.

McIntyre, C.L., Knowles, N.J. and Simmonds, P., 2013. Proposals for the classification of human rhinovirus species A, B and C into genotypically assigned types. The Journal of General Virology 94, 1791-1806.

Menzel, M., Akbarshahi, H., Bjermer, L. and Uller, L., 2016. Azithromycin induces anti-viral effects in cultured bronchial epithelial cells from COPD patients. Sci. Rep. 6, 28698.

Message, S.D. and Johnston, S.L., 2002. Viruses in asthma: The role of viruses in childhood respiratory infections. Br. Med. Bull. 61, 29-43.

Message, S.D., Laza-Stanca, V., Mallia, P., Parker, H.L., Zhu, J., Kebadze, T., Contoli, M., Sanderson, G., Kon, O.M., Papi, A., Jeffery, P.K., Stanciu, L.A. and Johnston, S.L., 2008. Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production. Proc. Natl. Acad. Sci. U. S. A. 105, 13562-13567.

Miller, E.K., Hernandez, J.Z., Wimmenauer, V., Shepherd, B.E., Hijano, D., Libster, R., Serra, M.E., Bhat, N., Batalle, J.P., Mohamed, Y., Reynaldi, A., Rodriguez, A., Otello, M., Pisapia, N., Bugna, J., Bellabarba, M., Kraft, D., Coviello, S., Ferolla, F.M., Chen, A., London, S.J., Siberry, G.K., Williams, J.V. and Polack, F.P., 2012. A Mechanistic Role for Type III IFN-λ1 in Asthma Exacerbations Mediated by Human Rhinoviruses. Am. J. Respir. Crit. Care Med. 185, 508-516.

Miller, E.K., Lu, X., Erdman, D.D., Poehling, K.A., Zhu, Y., Griffin, M.R., Hartert, T.V., Anderson, L.J., Weinberg, G.A., Hall, C.B., Iwane, M.K. and Edwards, K.M., 2007a. Rhinovirus-Associated Hospitalizations in Young Children. J. Infect. Dis. 195, 773-781.

Miller, E.K., Lu, X., Erdman, D.D., Poehling, K.A., Zhu, Y., Griffin, M.R., Hartert, T.V., Anderson, L.J., Weinberg, G.A., Hall, C.B., Iwane, M.K., Edwards, K.M. and New Vaccine Surveillance, N., 2007b. Rhinovirus-associated hospitalizations in young children. J. Infect. Dis. 195, 773-81.

Miller, E.K., Williams, J.V., Gebretsadik, T., Carroll, K.N., Dupont, W.D., Mohamed, Y.A., Morin, L.L., Heil, L., Minton, P.A., Woodward, K., Liu, Z. and Hartert, T.V., 2011. Host and viral factors associated with severity of human rhinovirus-associated infant respiratory tract illness. J. Allergy Clin. Immunol. 127, 883-91.

Page 189: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

169

Min, C.-K., Cheon, S., Ha, N.-Y., Sohn, K.M., Kim, Y., Aigerim, A., Shin, H.M., Choi, J.-Y., Inn, K.-S., Kim, J.-H., Moon, J.Y., Choi, M.-S., Cho, N.-H. and Kim, Y.-S., 2016. Comparative and kinetic analysis of viral shedding and immunological responses in MERS patients representing a broad spectrum of disease severity. Sci. Rep. 6, 25359.

Mojtahedi, M., Fouquier d'Herouel, A. and Huang, S., 2014. Direct elicitation of template concentration from quantification cycle (Cq) distributions in digital PCR. Nucleic Acids Res. 42, e126.

Monick, M.M., Powers, L.S., Hassan, I., Groskreutz, D., Yarovinsky, T.O., Barrett, C.W., Castilow, E.M., Tifrea, D., Varga, S.M. and Hunninghake, G.W., 2007. Respiratory Syncytial Virus Synergizes with Th2 Cytokines to Induce Optimal Levels of TARC/CCL17. The Journal of Immunology 179, 1648-1658.

Monto, A.S., 2002. The seasonality of rhinovirus infections and its implications for clinical recognition. Clin. Ther. 24, 1987-97.

Moore, K.W., O'Garra, A., Malefyt, R.W., Vieira, P. and Mosmann, T.R., 1993. Interleukin-10. Annu. Rev. Immunol. 11, 165-190.

Moore, M.L., Stokes, K.L. and Hartert, T.V., 2013. The impact of viral genotype on pathogenesis and disease severity: respiratory syncytial virus and human rhinoviruses. Curr. Opin. Immunol. 25, 761-768.

Moore, W.C., Bleecker, E.R., Curran-Everett, D., Erzurum, S.C., Ameredes, B.T., Bacharier, L., Calhoun, W.J., Castro, M., Chung, K.F., Clark, M.P., Dweik, R.A., Fitzpatrick, A.M., Gaston, B., Hew, M., Hussain, I., Jarjour, N.N., Israel, E., Levy, B.D., Murphy, J.R., Peters, S.P., Teague, W.G., Meyers, D.A., Busse, W.W. and Wenzel, S.E., 2007. Characterization of the severe asthma phenotype by the National Heart. Lung and Blood Institute’s Severe Asthma Research Program. J. Allergy Clin. Immunol. 119.

Morales, S.E. and Holben, W.E., 2009. Empirical testing of 16S rRNA gene PCR primer pairs reveals variance in target specificity and efficacy not suggested by in silico analysis. Appl. Environ. Microbiol. 75, 2677-83.

Moyes, J., Cohen, C., Pretorius, M., Groome, M., von Gottberg, A., Wolter, N., Walaza, S., Haffejee, S., Chhagan, M., Naby, F., Cohen, A.L., Tempia, S., Kahn, K., Dawood, H., Venter, M., Madhi, S.A. and Group, f.t.S.A.S.A.R.I.S., 2013. Epidemiology of Respiratory Syncytial Virus–Associated Acute Lower Respiratory Tract Infection Hospitalizations Among HIV-Infected and HIV-Uninfected South African Children, 2010–2011. J. Infect. Dis. 208, S217-S226.

Mufson, M.A., Orvell, C., Rafnar, B. and Norrby, E., 1985. Two distinct subtypes of human respiratory syncytial virus. J. Gen. Virol. 66 ( Pt 10), 2111-24.

Mukherjee, S. and Lukacs, N.W., 2010. Association of Interleukin- 13 in RSV induced pulmonary disease: Still a promising target. Expert Rev. Anti Infect. Ther. 8, 617-621.

Munywoki, P.K., Koech, D.C., Agoti, C.N., Bett, A., Cane, P.A., Medley, G.F. and Nokes , D.J., 2015. Frequent Asymptomatic Respiratory Syncytial Virus Infections During an Epidemic in a Rural Kenyan Household Cohort. J. Infect. Dis. 212, 1711-1718.

Murray, C.S., Poletti, G., Kebadze, T., Morris, J., Woodcock, A., Johnston, S.L. and Custovic, A., 2006. Study of modifiable risk factors for asthma exacerbations: virus infection and allergen exposure increase the risk of asthma hospital admissions in children. Thorax 61, 376-382.

NAC, A.A.A.a.N.A.C.A. 2015. The Hidden Cost of Asthma. Nagarkar, D.R., Wang, Q., Shim, J., Zhao, Y., Tsai, W.C., Lukacs, N.W., Sajjan, U. and

Hershenson, M.B., 2009. CXCR2 is required for neutrophilic airways inflammation and hyperresponsiveness in a mouse model of human rhinovirus infection. Journal of immunology (Baltimore, Md. : 1950) 183, 6698-6707.

Page 190: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

170

Nair, H., Brooks, W.A., Katz, M., Roca, A., Berkley, J.A., Madhi, S.A., Simmerman, J.M., Gordon, A., Sato, M., Howie, S., Krishnan, A., Ope, M., Lindblade, K.A., Carosone-Link, P., Lucero, M., Ochieng, W., Kamimoto, L., Dueger, E., Bhat, N., Vong, S., Theodoratou, E., Chittaganpitch, M., Chimah, O., Balmaseda, A., Buchy, P., Harris, E., Evans, V., Katayose, M., Gaur, B., O'Callaghan-Gordo, C., Goswami, D., Arvelo, W., Venter, M., Briese, T., Tokarz, R., Widdowson, M.-A., Mounts, A.W., Breiman, R.F., Feikin, D.R., Klugman, K.P., Olsen, S.J., Gessner, B.D., Wright, P.F., Rudan, I., Broor, S., Simões, E.A.F. and Campbell, H., 2011a. Global burden of respiratory infections due to seasonal influenza in young children: a systematic review and meta-analysis. The Lancet 378, 1917-1930.

Nair, H., Nokes, D.J., Gessner, B.D., Dherani, M., Madhi, S.A., Singleton, R.J., O'Brien, K.L., Roca, A., Wright, P.F., Bruce, N., Chandran, A., Theodoratou, E., Sutanto, A., Sedyaningsih, E.R., Ngama, M., Munywoki, P.K., Kartasasmita, C., Simoes, E.A., Rudan, I., Weber, M.W. and Campbell, H., 2010. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet 375, 1545-55.

Nair, H., Simões, E.A.F., Rudan, I., Gessner, B.D., Azziz-Baumgartner, E., Zhang, J.S.F., Feikin, D.R., Mackenzie, G.A., Moiïsi, J.C., Roca, A., Baggett, H.C., Zaman, S.M.A., Singleton, R.J., Lucero, M.G., Chandran, A., Gentile, A., Cohen, C., Kri shnan, A., Bhutta, Z.A., Arguedas, A., Clara, A.W., Andrade, A.L., Ope, M., Ruvinsky, R.O., Hortal, M., McCracken, J.P., Madhi, S.A., Bruce, N., Qazi, S.A., Morris, S.S., El Arifeen, S., Weber, M.W., Scott, J.A.G., Brooks, W.A., Breiman, R.F. and Campbell, H., 2013. Global and regional burden of hospital admissions for severe acute lower respiratory infections in young children in 2010: a systematic analysis. The Lancet 381, 1380-1390.

Nair, H., Verma, V., Theodoratou, E., Zgaga, L., Huda, T., Simoes, E., Wright, P., Rudan, I. and Campbell, H., 2011b. An evaluation of the emerging interventions against Respiratory Syncytial Virus (RSV)-associated acute lower respiratory infections in children. BMC Public Health 11, S30.

Ni, K., Li, S., Xia, Q., Zang, N., Deng, Y., Xie, X., Luo, Z., Luo, Y., Wang, L., Fu, Z. and Liu, E., 2012. Pharyngeal microflora disruption by antibiotics promotes airway hyperresponsiveness after respiratory syncytial virus infection. PLoS One 7, e41104.

Nicholson, K.G., Wood, J.M. and Zambon, M., 2003. Influenza. The Lancet 362, 1733-1745. Niesters, H.G., 2001. Quantitation of viral load using real-time amplification techniques.

Methods 25, 419-29. Obermayer, A., Stoiber, W., Krautgartner, W.-D., Klappacher, M., Kofler, B., Steinbacher, P.,

Vitkov, L., Grabcanovic-Musija, F. and Studnicka, M., 2014. New Aspects on the Structure of Neutrophil Extracellular Traps from Chronic Obstructive Pulmonary Disease and <italic>In Vitro</italic> Generation. PLoS One 9, e97784.

Oliveira, T.F., Freitas, G.R., Ribeiro, L.Z., Yokosawa, J., Siqueira, M.M., Portes, S.A., Silveira, H.L., Calegari, T., Costa, L., Mantese, O.C. and Queiroz, D.A., 2008. Prevalence and clinical aspects of respiratory syncytial virus A and B groups in children seen at Hospital de Clinicas of Uberlandia, MG, Brazil. Mem. Inst. Oswaldo Cruz 103, 417-22.

Oommen, A., McNally, T. and Grigg, J., 2003. Eosinophil activation and preschool viral wheeze. Thorax 58, 876-879.

Ortiz de la Tabla, V., Masiá, M., Antequera, P., Martin, C., Gazquez, G., Buñuel, F. and Gutiérrez, F., 2010. Comparison of Combined Nose-Throat Swabs with Nasopharyngeal Aspirates for Detection of Pandemic Influenza A/H1N1 2009 Virus by Real-Time Reverse Transcriptase PCR. J. Clin. Microbiol. 48, 3492-3495.

Page 191: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

171

Pala, P., Bjarnason, R., Sigurbergsson, F., Metcalfe, C., Sigurs, N. and Openshaw, P.J., 2002. Enhanced IL-4 responses in children with a history of respiratory syncytial virus bronchiolitis in infancy. Eur. Respir. J. 20, 376-82.

Palatnik de Sousa, C.B., Veach, A.J., Beard, C., Porter, F., Wilson, M. and Scorza, F.B., 2015. Procedia of the 8th Vaccine & ISV Congress, Philadelphia, USA, 2015Digital Droplet PCR for Influenza Vaccine Development. Procedia in Vaccinology 9, 96-103.

Papadopoulos, N.G., Gourgiotis, D., Javadyan, A., Bossios, A., Kallergi, K., Psarras, S., Tsolia, M.N. and Kafetzis, D., 2004. Does respiratory syncytial virus subtype influences the severity of acute bronchiolitis in hospitalized infants? Respir. Med. 98, 879-882.

Paynter, S., Ware, R.S., Lucero, M.G., Tallo, V., Nohynek, H., Weinstein, P., Williams, G., Sly, P.D. and Simoes, E.A., 2014. Malnutrition: a risk factor for severe respiratory syncytial virus infection and hospitalization. Pediatr. Infect. Dis. J. 33, 267-71.

Peiris, J.S.M., Hui, K.P.Y. and Yen, H.-L., 2010. Host response to Influenza virus: protection versus immunopathology. Curr. Opin. Immunol. 22, 475-481.

Pelaia, G., Vatrella, A. and Maselli, R., 2012. The potential of biologics for treatment of asthma. Nat. Rev. Drug Discov. 11.

Pfefferle, S., Oppong, S., Drexler, J.F., Gloza-Rausch, F., Ipsen, A. and Seebens, A., 2009. Distant relatives of severe acute respiratory syndrome coronavirus and close relatives of human coronavirus 229E in bats. Ghana Emerg Infect Dis 15.

Piralla, A., Lilleri, D., Sarasini, A., Marchi, A., Zecca, M., Stronati, M., Baldanti, F. and Gerna, G., 2012. Human rhinovirus and human respiratory enterovirus (EV68 and EV104) infections in hospitalized patients in Italy, 2008–2009. Diagn. Microbiol. Infect. Dis. 73, 162-167.

Piralla, A., Rovida, F., Campanini, G., Rognoni, V., Marchi, A., Locatelli, F. and Gerna, G., 2009. Clinical severity and molecular typing of human rhinovirus C strains during a fall outbreak affecting hospitalized patients. J. Clin. Virol. 45, 311-7.

Poorisrisak, P., Halkjaer, L.B., Thomsen, S.F., Stensballe, L.G., Kyvik, K.O., Skytthe, A., Schioetz, P.O. and Bisgaard, H., 2010. Causal direction between respiratory syncytial virus bronchiolitis and asthma studied in monozygotic twins. Chest 138, 338-44.

Pretorius, M.A., Tempia, S., Walaza, S., Cohen, A.L., Moyes, J., Variava, E., Dawood, H., Seleka, M., Hellferscee, O., Treurnicht, F., Cohen, C. and Venter, M., 2016. The role of influenza, RSV and other common respiratory viruses in severe acute respiratory infections and influenza-like illness in a population with a high HIV sero-prevalence, South Africa 2012-2015. J. Clin. Virol. 75, 21-6.

Pretorius, M.A., van Niekerk, S., Tempia, S., Moyes, J., Cohen, C., Madhi, S.A., Venter, M. and Group, f.t.S.S., 2013. Replacement and Positive Evolution of Subtype A and B Respiratory Syncytial Virus G-Protein Genotypes From 1997–2012 in South Africa. J. Infect. Dis. 208, S227-S237.

PREVENT, 1997. Reduction of respiratory syncytial virus hospitalization among premature infants and infants with bronchopulmonary dysplasia using respiratory syncytial virus immune globulin prophylaxis. The PREVENT Study Group. Pediatrics 99, 93-9.

Pritchard, A.L., White, O.J., Burel, J.G. and Upham, J.W., 2012. Innate interferons inhibit allergen and microbial specific TH2 responses. Immunol. Cell Biol. 90, 974-977.

Proud, D., 2011. Role of rhinovirus infections in asthma. Asian Pac. J. Allergy Immunol. 29, 201-8.

Quinn, T.C., 2011. Viral Kinetics of Genital Herpes: A Molecular Probe Into Host-Viral Interactions. The Journal of Infectious Diseases 204, 495-498.

Quint, J.K., 2008. IP-10 as a biomarker for rhinoviral infections in asthma. Thorax 63, 200. Rakes, G.P., Arruda, E., Ingram, J.M., Hoover, G.E., Zambrano, J.C., Hayden, F.G., Platts-

Mills, T.A.E. and Heymann, P.W., 1999. Rhinovirus and Respiratory Syncytial Virus

Page 192: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

172

in Wheezing Children Requiring Emergency Care. Am. J. Respir. Crit. Care Med. 159, 785-790.

Randhawa, P., Kant, J., Shapiro, R., Tan, H., Basu, A. and Luo, C., 2011. Impact of Genomic Sequence Variability on Quantitative PCR Assays for Diagnosis of Polyomavirus BK Infection. J. Clin. Microbiol. 49, 4072-4076.

Riccio, A.M., Dal Negro, R.W., Micheletto, C., De Ferrari, L., Folli, C., Chiappori, A. and Canonica, G.W., 2012. Omalizumab modulates bronchial reticular basement membrane thickness and eosinophil infiltration in severe persistent allergic asthma patient. Int. J. Immunopathol. Pharmacol. 25.

Robinson, C.M., Singh, G., Henquell, C., Walsh, M.P., Peigue-Lafeuille, H., Seto, D., Jones, M.S., Dyer, D.W. and Chodosh, J., 2011. Computational analysis and identification of an emergent human adenovirus pathogen implicated in a respiratory fatality. Virology 409, 141-147.

Roussy, J.F., Carbonneau, J., Ouakki, M., Papenburg, J., Hamelin, M.E., De Serres, G. and Boivin, G., 2014. Human metapneumovirus viral load is an important risk factor for disease severity in young children. J. Clin. Virol. 60, 133-40.

Royal Children's Hospital, R. 2015. Clinical Practice Guidline, Melbourne, Australia. Ruuskanen, O., Lahti, E., Jennings, L.C. and Murdoch, D.R., 2011. Viral pneumonia. The

Lancet 377, 1264-1275. Sajjan, U.S., Jia, Y., Newcomb, D.C., Bentley, J.K., Lukacs, N.W., LiPuma, J.J. and

Hershenson, M.B., 2006. H. influenzae potentiates airway epithelial cell responses to rhinovirus by increasing ICAM-1 and TLR3 expression. FASEB J. 20, 2121-3.

Saraya, T., Kurai, D., Ishii, H., Ito, A., Sasaki, Y., Niwa, S., Kiyota, N., Tsukagoshi, H., Kozawa, K., Goto, H. and Takizawa, H., 2014. Epidemiology of virus-induced asthma exacerbations: with special reference to the role of human rhinovirus. Front. Microbiol. 5.

Scagnolari, C., Midulla, F., Pierangeli, A., Moretti, C., Bonci, E., Berardi, R., De Angelis, D., Selvaggi, C., Di Marco, P., Girardi, E. and Antonelli, G., 2009. Gene Expression of Nucleic Acid-Sensing Pattern Recognition Receptors in Children Hospitalized for Respiratory Syncytial Virus-Associated Acute Bronchiolitis. Clin. Vaccine Immunol. 16, 816-823.

Schauer, U., Hoffjan, S., Bittscheidt, J., Kochling, A., Hemmis, S., Bongartz, S. and Stephan, V., 2002. RSV bronchiolitis and risk of wheeze and allergic sensitisation in the first year of life. Eur. Respir. J. 20, 1277-83.

Schopf, L.R., Hoffmann, K.F., Cheever, A.W., Urban, J.F., Jr. and Wynn, T.A., 2002. IL-10 is critical for host resistance and survival during gastrointestinal helminth infection. J. Immunol. 168, 2383-92.

Self, W.H., Williams, D.J., Zhu, Y., Ampofo, K., Pavia, A.T., Chappell, J.D., Hymas, W.C., Stockmann, C., Bramley, A.M., Schneider, E., Erdman, D., Finelli, L., Jain, S., Edwards, K.M. and Grijalva, C.G., 2015. Respiratory Viral Detection in Children and Adults: Comparing Asymptomatic Controls and Patients With Community-Acquired Pneumonia. J. Infect. Dis.

Shadman, K.A. and Wald, E.R., 2011. A review of palivizumab and emerging therapies for respiratory syncytial virus. Expert Opin. Biol. Ther. 11, 1455-67.

Sigurs, N., Aljassim, F., Kjellman, B., Robinson, P.D., Sigurbergsson, F., Bjarnason, R. and Gustafsson, P.M., 2010. Asthma and allergy patterns over 18 years after severe RSV bronchiolitis in the first year of life. Thorax 65, 1045-52.

Sigurs, N., Bjarnason, R., Sigurbergsson, F. and Kjellman, B., 2000. Respiratory Syncytial Virus Bronchiolitis in Infancy Is an Important Risk Factor for Asthma and Allergy at Age 7. Am. J. Respir. Crit. Care Med. 161, 1501-1507.

Page 193: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

173

Soto-Quiros, M., Avila, L., Platts-Mills, T.A.E., Hunt, J.F., Erdman, D.D., Carper, H., Murphy, D.D., Odio, S., James, H.R., Patrie, J.T., Hunt, W., O'Rourke, A.K., Davis, M.D., Steinke, J.W., Lu, X., Kennedy, J. and Heymann, P.W., 2012. High titers of IgE antibody to dust mite allergen and risk for wheezing among asthmatic children infected with rhinovirus. J. Allergy Clin. Immunol. 129, 1499-1505.

Stephens, K.W., Hutchins, R.J. and Dauphin, L.A., 2010. Cross-platform evaluation of commercial real-time reverse transcription PCR master mix kits using a quantitative 5′nuclease assay for Ebola virus. Mol. Cell. Probes 24, 370-375.

Sugamata, R., Dobashi, H., Nagao, T., Yamamoto, K.-i., Nakajima, N., Sato, Y., Aratani, Y., Oshima, M., Sata, T., Kobayashi, K., Kawachi, S., Nakayama, T. and Suzuki, K., 2012. Contribution of neutrophil-derived myeloperoxidase in the early phase of fulminant acute respiratory distress syndrome induced by influenza virus infection. Microbiol. Immunol. 56, 171-182.

Sykes, A., Macintyre, J., Edwards, M.R., del Rosario, A., Haas, J., Gielen, V., Kon, O.M., McHale, M. and Johnston, S.L., 2014. Rhinovirus-induced interferon production is not deficient in well controlled asthma. Thorax 69, 240-246.

Takeyama, A., Hashimoto, K., Sato, M., Kawashima, R., Kawasaki, Y. and Hosoya, M., 2015a. Respiratory syncytial virus shedding by children hospitalized with lower respiratory tract infection. J. Med. Virol.

Takeyama, A., Hashimoto, K., Sato, M., Kawashima, R., Kawasaki, Y. and Hosoya, M., 2015b. Respiratory syncytial virus shedding by children hospitalized with lower respiratory tract infection. J. Med. Virol., n/a-n/a.

Tamaoki, J., Kondo, M., Kohri, K., Aoshiba, K., Tagaya, E. and Nagai, A., 1999. Macrolide antibiotics protect against immune complex-induced lung injury in rats: role of nitric oxide from alveolar macrophages. J. Immunol. 163, 2909-15.

Tang, H., Cai, Q., Li, H. and Hu, P., 2016. Comparison of droplet digital PCR to real -time PCR for quantification of hepatitis B virus DNA. Biosci. Biotechnol. Biochem., 1-6.

Tempia, S., Walaza, S., Viboud, C., Cohen, A.L., Madhi, S.A., Venter, M., von Mollendorf, C., Moyes, J., McAnerney, J.M. and Cohen, C., 2015. Deaths Associated with Respiratory Syncytial and Influenza Viruses among Persons ≥5 Years of Age in HIV-Prevalent Area, South Africa, 1998–2009(1). Emerging Infect. Dis. 21, 600-608.

Terletskaia-Ladwig, E., Meier, S., Hahn, R., Leinmüller, M., Schneider, F. and Enders, M., 2008. A convenient rapid culture assay for the detection of enteroviruses in clinical samples: comparison with conventional cell culture and RT-PCR. J. Med. Microbiol. 57, 1000-1006.

The Australian Lung Foundation, A. 2007. Respiratory Infectious Disease in Australia, The Australian Lung Foundation, Brisbane, Australia.

Thompson, A.J.V. and Locarnini, S.A., 2007. Toll-like receptors, RIG-I-like RNA helicases and the antiviral innate immune response. Immunol Cell Biol 85.

To, K.K.W., Chan, K.-H., Li, I.W.S., Tsang, T.-Y., Tse, H., Chan, J.F.W., Hung, I.F.N., Lai, S.-T., Leung, C.-W., Kwan, Y.-W., Lau, Y.-L., Ng, T.-K., Cheng, V.C.C., Peiris, J.S.M. and Yuen, K.-Y., 2010. Viral load in patients infected with pandemic H1N1 2009 influenza A virus. J. Med. Virol. 82, 1-7.

To, K.K.W., Hung, I.F.N., Chan, J.F.W. and Yuen, K.-Y., 2013. From SARS coronavirus to novel animal and human coronaviruses. J. Thorac. Dis., S103-S108.

Tovey, E.R., Stelzer-Braid, S., Toelle, B.G., Oliver, B.G., Reddel, H.K., Willenborg, C.M., Belessis, Y., Garden, F.L., Jaffe, A., Strachan, R., Eyles, D., Rawlinson, W.D. and Marks, G.B., 2015. Rhinoviruses significantly affect day-to-day respiratory symptoms of children with asthma. J. Allergy Clin. Immunol. 135, 663-9.e12.

Tsukagoshi, H., Ishioka, T., Masahiro, N., Kozawa, K. and Kimura, H., 2013a. Molecular epidemiology of respiratory viruses in virus-induced asthma. Front. Microbiol. 4.

Page 194: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

174

Tsukagoshi, H., Ishioka, T., Noda, M., Kozawa, K. and Kimura, H., 2013b. Molecular epidemiology of respiratory viruses in virus-induced asthma. Front. Microbiol. 4, 278.

Utokaparch, S., Marchant, D., Gosselink, J.V., McDonough, J.E., Thomas, E.E., Hogg, J.C. and Hegele, R.G., 2011. The relationship between respiratory viral loads and diagnosis in children presenting to a pediatric hospital emergency department. Pediatr. Infect. Dis. J. 30, e18-23.

van den Hoogen, B.G., Bestebroer, T.M., Osterhaus, A.D. and Fouchier, R.A., 2002. Analysis of the genomic sequence of a human metapneumovirus. Virology 295, 119-32.

van Niekerk, S. and Venter, M., 2011. Replacement of Previously Circulating Respiratory Syncytial Virus Subtype B Strains with the BA Genotype in South Africa. J. Virol. 85, 8789-8797.

Velayutham, T.S., Kolli, D., Ivanciuc, T., Garofalo, R.P. and Casola, A., 2013. Critical role of TLR4 in human metapneumovirus mediated innate immune responses and disease pathogenesis. PLoS One 8, e78849.

Verhaegen, B., De Reu, K., De Zutter, L., Verstraete, K., Heyndrickx, M. and Van Coillie, E., 2016. Comparison of Droplet Digital PCR and qPCR for the Quantification of Shiga Toxin-Producing Escherichia coli in Bovine Feces. Toxins (Basel) 8.

Walker, C.L.F., Rudan, I., Liu, L., Nair, H., Theodoratou, E., Bhutta, Z.A., O'Brien, K.L., Campbell, H. and Black, R.E., 2013. Global burden of childhood pneumonia and diarrhoea. Lancet 381, 1405-1416.

Wang, Y., Ji, W., Chen, Z., Yan, Y.D., Shao, X. and Xu, J., 2014. Comparison of severe pneumonia caused by Human metapneumovirus and respiratory syncytial virus in hospitalized children. Indian J. Pathol. Microbiol. 57, 413-7.

Wennergren, G. and Kristjánsson, S., 2001. Relationship between respiratory syncytial virus bronchiolitis and future obstructive airway diseases. Eur. Respir. J. 18, 1044-1058.

Wenzel, S.E., Schwartz, L.B., Langmack, E.L., Halliday, J.L., Trudeau, J.B., Gibbs, R.L. and Chu, H.W., 1999. Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am. J. Respir. Crit. Care Med. 160, 1001-8.

West, D.M. and Sawyer, J., 2006. Freezing Complete Polymerase Chain Reaction Master Mix Reagents for Routine Molecular Diagnostics. J. Vet. Diagn. Invest. 18, 580-582.

Whiley, D.M. and Sloots, T.P., 2005. Sequence variation in primer targets affects the accuracy of viral quantitative PCR. J. Clin. Virol. 34, 104-107.

Widmer, K., Zhu, Y., Williams, J.V., Griffin, M.R., Edwards, K.M. and Talbot, H.K., 2012. Rates of hospitalizations for respiratory syncytial virus, human metapneumovirus, and influenza virus in older adults. J. Infect. Dis. 206, 56-62.

Wiehler, S. and Proud, D., 2007. Interleukin-17A modulates human airway epithelial responses to human rhinovirus infection. Am. J. Physiol. Lung Cell Mol. Physiol. 293, L505-15.

Wu, P. and Hartert, T.V., 2011. Evidence for a causal relationship between respiratory syncytial virus infection and asthma. Expert Rev. Anti Infect. Ther. 9.

Xiang, Z., Gonzalez, R., Ren, L., Xiao, Y., Chen, L., Zhang, J., Wang, W., Yang, Q., Li, J., Zhou, H., Vernet, G., Paranhos-Baccala, G., Wang, Z. and Wang, J., 2013. Prevalence and clinical characteristics of human respiratory syncytial virus in Chinese adults with acute respiratory tract infection. J. Med. Virol. 85, 348-53.

Yusof, M.F., Eltahir, Y.M., Serhan, W.S., Hashem, F.M., Elsayed, E.A. and Marzoug, B.A., 2015. Prevalence of Middle East respiratory syndrome coronavirus (MERS-CoV) in dromedary camels in Abu Dhabi Emirate. United Arab Emirates Virus genes 50.

Page 195: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

175

Zaki , A.M., van Boheemen , S., Bestebroer , T.M., Osterhaus , A.D.M.E. and Fouchier , R.A.M., 2012. Isolation of a Novel Coronavirus from a Man with Pneumonia in Saudi Arabia. New Engl. J. Med. 367, 1814-1820.

Zash, R.M., Shapiro, R.L., Leidner, J., Wester, C., McAdam, A.J., Hodinka, R.L., Thior, I., Moffat, C., Makhema, J., McIntosh, K., Essex, M. and Lockman, S., 2016. The aetiology of diarrhoea, pneumonia and respiratory colonization of HIV-exposed infants randomized to breast- or formula-feeding. Paediatr Int Child Health, 1-9.

Zeng, R., Li, C., Li, N., Wei, L. and Cui, Y., 2011. The role of cytokines and chemokines in severe respiratory syncytial virus infection and subsequent asthma. Cytokine 53, 1-7.

Zhang, X.-B., Liu, L.-J., Qian, L.-L., Jiang, G.-L., Wang, C.-K., Jia, P., Shi, P., Xu, J. and Wang, L.-B., 2014. Clinical characteristics and risk factors of severe respiratory syncytial virus-associated acute lower respiratory tract infections in hospitalized infants. World J. Pediatr. 10, 360-364.

Zhao, J., Takamura, M., Yamaoka, A., Odajima, Y. and Iikura, Y., 2002. Altered eosinophil levels as a result of viral infection in asthma exacerbation in childhood. Pediatr. Allergy Immunol. 13, 47-50.

Zhu, Z., Tang, W., Gwaltney, J.M., Jr., Wu, Y. and Elias, J.A., 1997. Rhinovirus stimulation of interleukin-8 in vivo and in vitro: role of NF-kappaB. Am. J. Physiol. 273, L814-24.

Zhu, Z., Tang, W., Ray, A., Wu, Y., Einarsson, O., Landry, M.L., Gwaltney, J. and Elias, J.A., 1996. Rhinovirus stimulation of interleukin-6 in vivo and in vitro. Evidence for nuclear factor kappa B-dependent transcriptional activation. J. Clin. Invest. 97, 421-

430.

Page 196: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

176

Appendices

Appendix 1

Table 0-1 The performance of the individual PCR assays for the detection of matched RV-C RNA transcript

RV-C Assay-1 RV-C Assay-2 RV-C Assay-3 RV-C Assay-4

Mean +/- SD CV% Mean +/- SD CV% Mean +/- SD CV% Mean +/- SD CV%

Slope (n=) -3.32+/-0.11 3.33 -3.38+/-0.11 3.23 -3.44+/-0.09 2.72 -3.37+/-0.05 1.52

Efficiency 0.98+/-0.05 5.02 0.97+/-0.05 4.68 0.95+/-0.04 3.93 0.97+/-0.03 3.12

Y-intercept 34.00+/-2.46 7.23 38.00+/-2.72 7.17 36.12+/- 1.00 7.55 33.45 +/- 1.59 8.42

Goodness of fit (R2) 0.999 0.11 0.999 0.47 0.999 0.09 0.999 0.08

Range of Linearity 100-108 copies/ml 100-108 copies/ml 100-108 copies/ml 100-108 copies/ml

Page 197: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

177

Table 0-2 A comparison of RNA transcript concentration and Cq values for the different RV-C assays

RV-C Assay-1 RV-C Assay-2 RV-C Assay-3 RV-C Assay-4

RNA transcript

concentration

Mean Cq +/-

SD

Mean Cq +/-

SD

Mean Cq +/-

SD

Mean Cq +/-

SD

100 32.77+/-0.14 33.25+/-1.48 32.66+/-0.31 31.84+/-1.01

101 29.11+/-0.12 28.22+/-0.46 29.62+/-0.06 27.37+/-0.07

102 25.86+/-0.11 25.37+/-0.06 25.95+/-0.07 24.20+/-0.14

103 22.07+/-0.06 22.25+/-0.07 22.52+/-0.04 20.96+/-0.03

104 18.68+/-0.06 18.91+/-0.06 19.08+/-0.08 17.53+/-0.10

105 15.35+/-0.04 15.39+/-0.04 15.52+/-0.11 14.20+/-0.14

106 11.73+/-0.1 13.13+/-0.11 12.05+/-0.08 10.79+/-0.04

107 8.5+/-0.04 8.78+/-0.02 8.55+/-0.02 7.81+/-0.06

108 5.27+/-0.12 5.17+/-0.2 5.19+/-0.16 4.77+/-0.07

% CV – Percentage coefficient variation, Cq- quanti fication cycle va lue, SD- s tandard deviation

Page 198: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

178

Table 0-3 Intra and Inter assay variability of the four RV-C qRT-PCR assays (Assay 1-4)

Intra -ass ay variation a

Inter-assay variation b

RNA target and input target copies

Quantity range (Calculated copies/ reaction)

%CV range Quantity Mean (Calculated copies/reaction)

%CV

RV-C1 106 3030000-3560000 0.16-2.39 3480000 6.89

RV-C1 104 30000-36700 0.27-2.33 37500. 5.67

RV-C1 102 125-409 0.16-7.07 361. 8.92

RV-C1 101 21-40 2.1-7.33 39 5.88

.

RV-C2 106 3560000-4210000 0.22-0.61 3810000. 7.50

RV-C2 104 39100-49700 1.70 -2.42 43400. 10.56

RV-C2 102 379-405 1.44 -2.75 396. 3.04

RV-C2 101 35-42 3.89-5.82 39 7.22

RV-C3 10

6 4080000-5790000 0.37-2.30 4760000 14.58

RV-C3 104 35700-46200 0.23-1.23 42000 9.26

RV-C3 102 372-470 0.40-8.58 440 9.21

RV-C3 101 42-59 0.93-7.78 47 14.57

RV-C4 106 4020000-4860000 0.10-1.77 4360000 8.22

RV-C4 104 48500-52800 0.28-1.61 50000 4.89

RV-C4 102 423-608 1.07-4.76 525 11.05

RV-C4 101 42-49 1.47-5.16 46 5.36

% CV – Percentage coefficient variation a Assays were performed in triplicate b Five independent experiments

Page 199: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

179

Table 0-4 The RV-C load determinations for patients enrolled in the PREVIEW study. Clinical samples were tested in triplicate and mean viral load calculated.

Sample ID

RNA copies/mL

SD %CV

Log10 RNA copies/mL

Genotype

Assay

GAPDH Mean Cq

GAPDH SD

1 1.35E+05 4.73E+04

0.35

5.13 C-04 98% 1 30 0.8

2 2.98E+08 4.47E+07

0.15

8.47 C-06 95% 1 24.5 0.1

3 5.33E+09 7.46E+08

0.14

9.73 C-06 95% 1 25.6 0.8

4 3.15E+06 8.19E+05

0.26

6.5 C-08 100%

2 27.7 0.6

5 6.44E+04 1.67E+04

0.26

4.81 C-08 98% 2 27.7 0.3

6 1.06E+07 2.65E+06

0.25

7.02 C-14 96% 3 23 0.3

7 4.20E+07 7.56E+06

0.18

7.62 C-14 96% 3 22.8 0.3

8 3.55E+06 8.17E+05

0.23

6.55 C-14 97% 3 28.8 0.3

9 5.33E+09 8.53E+08

0.16

9.73 C-16 95% 1 27.6 0.6

10 4.28E+06 1.03E+06

0.24

6.63 C-16 94% 1 27.7 0.2

11 2.96E+07 6.51E+06

0.22

7.47 C-16 97% 1 25.4 0.4

12 1.91E+08 3.44E+07

0.18

8.28 C-16 97% 1 27.3 0.5

13 1.76E+08 2.82E+07

0.16

8.25 C-16 98% 1 28.2 0.1

14 3.28E+07 6.56E+06

0.2 7.52 C-23 97% 1 25.6 0.3

15 7.53E+08 1.28E+08

0.17

8.88 C-24 96% 1 21.4 0.6

16 1.98E+06 6.73E+05

0.34

6.3 C-25 97% 1 25.8 0.8

17 5.60E+08 8.40E+07

0.15

8.75 C-25 99% 1 20.2 0.5

18 2.28E+09 3.42E+08

0.15

9.36 C-28 98%)

1 21.6 0.4

19 9.66E+06 1.55E+06

0.16

6.99 C-3 97% 1 26.6 0.4

20 3.48E+05 1.08E+05

0.31

5.54 C-30 99% 1 27 0.4

21 1.99E+03 6.17E+02

0.31

3.3 C-35 96% 4 29.8 0.4

22 1.69E+06 6.76E+05

0.4 6.23 C-35 96% 4 27.3 0.4

23 3.19E+06 8.61E+05

0.27

6.5 C-35 97% 4 25.6 0.3

24 1.25E+07 2.13E+06

0.17

7.1 C-35 97% 4 22.3 1.3

25 3.26E+09 5.87E+08

0.18

9.51 C-38 96% 1 23.7 0.5

26 4.45E+06 1.29E+06

0.29

6.65 C-39 99% 1 24.2 0.1

27 1.04E+05 2.08E+04

0.2 5.02 C-42 95% 2 28.8 0.1

28 7.88E+05 2.99E+05

0.38

5.9 C-42 96% 2 30 0.8

29 4.15E+05 1.66E+04

0.04

5.62 C-42 97% 2 29.9 0.2

30 1.11E+06 3.66E+05

0.33

6.05 C-43 95% 1 28.2 0.4

31 2.65E+07 5.04E+06

0.19

7.42 C-46 96% 3 25.1 0.6

32 1.18E+08 1.53E+07

0.13

8.07 C-46 96% 3 23.5 0.2

33 4.73E+04 1.32E+0 0.2 4.67 C-51 96% 4 24.8 0.8

Page 200: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

180

4 8

34 8.51E+08 1.62E+08

0.19

8.93 C-04 99% 1 27.5 0.1

35 1.70E+07 3.74E+06

0.22

7.23 C-19 96% 4 29.8 0.1

36 2.93E+05 7.62E+04

0.26

5.47 C-11 99% 1 24.5 0.6

37 3.29E+05 8.23E+04

0.25

5.52 C-11 99% 1 28.6 0.4

38 1.23E+07 2.34E+06

0.19

7.09 C-11 99% 1 23.2 0.1

39 2.04E+06 4.90E+05

0.24

6.31 C-24 97% 1 25.3 0.2

40 3.95E+03 1.46E+03

0.37

3.6 C-13 96% 1 24.6 0.1

SD- Standard deviation %CV- coefficient of variation GAPDH- Glyceraldehyde 3-phosphate dehydrogenase; internal control

Page 201: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

181

Appendix 2

Published work completed during PhD

Page 202: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

182

Page 203: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

183

Page 204: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

184

Page 205: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

185

Page 206: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

186

Page 207: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

187

Page 208: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

188

Page 209: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

189

Page 210: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

190

Page 211: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

191

Page 212: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

192

Page 213: Chisha Teza Sikazwe - research-repository.uwa.edu.au · The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe

193