132
INVESTIGATION OF INHALED NITRIC OXIDE AND MESENCHYMAL STROMAL CELLS AS NOVEL THERAPEUTIC STRATEGIES TO IMPROVE CLINICAL OUTCOME IN EXPERIMENTAL SEVERE INFLUENZA by Ilyse Darwish A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Ilyse Darwish (2012)

INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

INVESTIGATION OF INHALED NITRIC OXIDE AND MESENCHYMAL STROMAL CELLS AS NOVEL

THERAPEUTIC STRATEGIES TO IMPROVE CLINICAL OUTCOME IN EXPERIMENTAL SEVERE INFLUENZA

by

Ilyse Darwish

A thesis submitted in conformity with the requirements for the degree of Master of Science

Institute of Medical Science University of Toronto

© Copyright by Ilyse Darwish (2012)

Page 2: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

ii

Investigation of Inhaled Nitric Oxide and Mesenchymal Stromal Cells as

Novel Therapeutic Strategies to Improve Clinical Outcome in

Experimental Severe influenza

Ilyse Darwish

Masters of Science

Institute of Medical Science University of Toronto

2012

Abstract

Severe influenza, recognized as a clinical syndrome characterized by hyper-induction of pro-

inflammatory cytokine production, results in approximately 250–500 thousand deaths annually

worldwide. Current influenza research is focused on therapeutics to target the influenza virus or

modulate influenza virus-induced inflammation as potential treatment options to improve clinical

outcome in experimental influenza A (H1N1) virus infection. The goals of this work were: (1) to

evaluate the utility of inhaled nitric oxide (iNO) for decreasing influenza virus production in the

lungs, and (2) investigate the use of mesenchymal stromal (stem) cells (MSCs) for mitigating

deleterious host responses to influenza infection. Here, we report that MSCs and iNO,

administered alone either prophylactically or post-influenza virus infection, fail to modulate host

inflammation, fail to improve acute lung injury, fail to dampen lung viral load, and fail to improve

survival of infected mice.

Page 3: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

iii

Acknowledgments

I am grateful to my supervisor Dr. W. Conrad Liles for his mentorship, guidance and support, and

to my committee members Dr. Kevin Kain, Dr. Samira Mubareka, and Dr. David Kelvin, for their

guidance and support. I am very thankful to the Kain lab for creating a supportive learning

environment which allowed me to excel throughout my graduate training, with particular thanks to

Dr. Hani Kim, Dr. Laura Erdman, Dr. Constance Finney, Dr. Lena Serghides and Beata Majczrak

for their patient teaching. Thank you to my parents, my sister and my partner for their support

throughout my studies.

Page 4: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

iv

Table of Contents

Page

Chapter 1: Literature Review

1.1 Global burden of influenza 1

1.2 Structure and life cycle of the influenza virion 2

1.3 Uncomplicated and severe influenza 5

1.4 Viral determinants of pathogenesis 6 1.4.1 Hemagglutinin…………………..…………………………………………………..... 6 1.4.2 Polymerase proteins including PB1-F2 peptide……………………………………… 7 1.4.3 Other viral determinants…………………..………………………………………….. 8

1.5 Antiviral Therapy 9

1.5.1. Neuraminidase inhibitors, M2 ion channel inhibitors and other antivirals…….......... 9 1.6 Investigational antiviral therapy – inhaled nitric oxide 13

1.7 Host determinants of pathogenesis

1.7.1 Cytokine dysregulation ………..……………………………………………............... 15

1.8 Immunomodulatory therapy 19 1.8.1 Glucocorticoids…………………..……………………………………………........... 21 1.8.2 Macrolides…………………..……………………………………………………...... 22 1.8.3 TNF inhibitors…………………..…………………………………………………... . 23 1.8.4 Cyclooxygenase-2 inhibitors…………………..…………...……………………….... 24 1.8.5 HMG-CoA reductase inhibitors…………………..………………...………………... 26 1.8.6 High mobility group box 1 antagonists……………………………….…………...…. 27 1.8.7 Peroxisome proliferator activated receptor agonists.………………..……………….. 28 1.8.8 AMP-activated protein kinase agonists……………………………..………………... 30 1.9 Investigational immunomodulatory therapy – mesenchymal stromal cells (MSCs) 31 1.9.1 MSCs and the immune system……………………………………………..………... 32 1.9.2 MSC duality – immunostimulant or immunosuppressant? …………………………. 36 1.9.3 Effects of MSCs in pre-clinical models……………...……………………………… 38 1.9.4 MSC derived paracrine mediators…………………………………………………… 39 1.10 Mouse models of influenza 41

1.11 Research aims and objectives 44

Page 5: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

v

Chapter 2: Inhaled nitric oxide antiviral therapy f or severe influenza

2.1 Introduction………………………………………………………………………………... 46

2.2 Materials and methods………………………………………………………..……………. 48

2.3 Results……………………………………………………………………………………… 50

2.4 Discussion..…………………………………………………………………………………. 55

Chapter 3: Mesenchymal stromal cell immunomodulatory therapy for severe influenza

3.1 Introduction……………………………………………………………………………........ 57

3.2 Materials and methods……………………………………………………………………... 59

3.3 Results……………………………………………………………………………………… 63

3.4 Discussion………………………………………………………………………………...... 81

Chapter 4: Summary and future directions

4.1 Summary ………………...………………………………..…………………………..…… 91

4.2 Future Directions……...………………………………………………………………........ 93

4.3 Final Considerations………………………………………………………………….......... 96

Chapter 5: References 98

Page 6: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

vi

List of Tables

Table 1.1. Summary of investigational immunomodulatory agents for treatment 20

of influenza

Page 7: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

vii

List of Figures

Chapter 1

Fig. 1.1. Structure and life cycle of influenza A virus 4

Fig. 1.2. Mesenchymal stem cell immune cell targets 34

Chapter 2

Fig. 2.1. Prophylactic iNO therapy increased weight loss and decreased survival 51

of C57Bl/6 mice infected with influenza A/WSN/33 virus

Fig. 2.2. Prophylactic and post-infection intermittent iNO does not alter weight 53

loss kinetics or survival of C57Bl/6 mice infected with influenza A/WSN/33 virus

Fig. 2.3. Intermittent high dose iNO prophylactic therapy failed to decrease viral 54

load of C57Bl/6 mice infected with influenza A/WSN/33 virus

Chapter 3

Fig. 3.1. Male C57BL/6 mice infected with 425 EID50 influenza A/PR/8 virus develop 63

increased morbidity and mortality

Fig. 3.2. Male C57BL/6 mice infected with 425 EID50 influenza A/PR/8 virus develop 65

lung inflammation and acute lung injury

Fig. 3.3. hMSCs fail to improve survival, alter weight loss kinetics or affect lung 67

viral load in experimental severe influenza

Fig. 3.4. hMSCs fail to alter lung inflammation in experimental severe influenza 69

Fig. 3.5. hMSCs fail to alter acute lung injury in experimental severe influenza 70

Fig. 3.6. hMSCs fail to alter weight loss kinetics in experimental sub-lethal 71

influenza

Fig. 3.7. Cytokine stimulated hMSC prophylaxis fails to alter weight loss kinetics 73

or improve survival in experimental severe influenza

Fig. 3.8. hMSC adjunctive therapy may improve survival in experimental influenza 75

A/PR/8 virus infection but not influenza A/Mex/4108 virus infection and hMSC

adjunctive therapy does not alter weight loss kinetics in both models of

experimental severe influenza

Fig. 3.9. hMSCs do not decrease BAL fluid or lung parenchymal inflammation when 77

used as adjunctive therapy in mice infected with influenza A/PR/8 virus

Page 8: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

viii

Fig. 3.10. hMSCs fail to decrease acute lung injury when used as adjunctive 78

therapy in mice infected with influenza A/PR/8 virus

Fig. 3.11. hMSCs do not decrease BAL fluid or lung parenchymal inflammation 79

when used in adjunctive to antiviral therapy in mice infected with influenza

A/Mex/4108 virus

Fig. 3.12. hMSCs fail to decrease acute lung injury when used as adjunctive 80

therapy in mice infected with influenza A/Mex/4108 virus

Page 9: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

ix

List of Abbreviations

A/Mex/4108: A/Mexico/4108/09

A/PR/8: A/PuertoRico/8/34

A/WSN/33: A/Wisconsin/33

AICAR: aminoimidazole carboxamide ribonucleotide

ALI: acute lung injury

AMPK: AMP-activated protein kinase

Ang-1: angiopoietin-1

APC: antigen presenting cell

ARDS: acute respiratory distress syndrome

BAL: bronchoalveolar lavage

CFU: colony forming unit

COX: cyclooxygenase

CV-N: cyanovirin-N

DC: dendritic cell

eNOS: endothelial nitric oxide synthase

FDA: Food and Drug Administration

GFP: green fluorescent protein

GvHD: graft-vs-host disease

HA: hemagglutinin

HLA: human leukocyte antigen

HMGB-1: high mobility group box 1

HMG-CoA: 3-hydroxy-3-methylglutaryl coenzyme A

HPAIV: high pathogenic avian influenza

IDO: indoleamine 2,3-dioxygenase

IFN: interferon

IL: interleukin

ILI: influenza-like-illness

IL-1R: interleukin-1 receptor

iNO: inhaled nitric oxide

Page 10: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

x

iNOS: inducible nitric oxide synthase

KGF: keratinocyte growth factor

LPAIV: low pathogenic avian influenza

LPS: lipopolysaccharide

MDCK cell: Madin-Darby canine kidney cell

MHC: major histocompatability complex

M1: matrix 1

M2: matrix 2

M2 macrophage: alternatively activated macrophage

NA: neuraminidase

NAI: neuraminidase inhibitor

NEP: nuclear export protein

NF-κB: nuclear factor kappa B

NK cell: natural killer cell

NO: nitric oxide

NOS: nitric oxide synthase

NP: nucleoprotein

NSAID: non-steroidal anti-inflammatory

NS1: non-structural protein 1

NS2: non-structural protein 2

PA: polymerase acidic protein

PB1: polymerase basic protein 1

PB2: polymerase basic protein 2

PB1-F2: polymerase basic protein 1 – F2

PFU: plaque forming unit

PGE2: prostaglandin E2

PPAR: peroxisome proliferator activated receptor

ROS: reactive oxygen species

SARS: severe acute respiratory syndrome

siRNA: short interfering RNA

SNAP: S-nitroso-N-acetylpenicillamine

sTNFR: soluble TNF receptor

Page 11: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

xi

TGF-β: transforming growth factor beta

TNF: tumour necrosis factor

TSG-6: TNF inducible protein 6

VCAM-1: vascular cell adhesion molecule-1

vRNP: viral ribonucleoprotein

WHO: world health organization

Page 12: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

1

Chapter 1 : Literature Review

1.1 Global burden of severe influenza

Influenza viruses A, B, and C belong to the Orthomyxoviridae family of single stranded

negative-sense RNA viruses. Influenza B viruses are known to infect only humans and seals,

while influenza A viruses can infect a wide range of avian and mammalian hosts. Influenza B

viruses can cause mild to severe respiratory tract infections and can account for a significant

proportion of laboratory confirmed cases of influenza; however, they do not cause pandemics

[299]. This is likely due to their host range, which limits the generation of new strains by

reassortment. Influenza C viruses are known to infect both humans and pigs and may cause mild

upper respiratory tract illness in humans which can rarely lead to infection of the lower

respiratory tract; nearly all adults have been infected with these viruses [300].

While wild aquatic birds are the primary reservoir for influenza A viruses, periodic reassortment

between avian and mammalian influenza viruses or adaptation to novel host species can give rise

to influenza epidemics or pandemics [1]. During annual influenza epidemics in humans,

approximately 5–15% of the population will be infected with influenza A virus [2]. Of those

infected, an estimated 3–5 million of these cases are classified as severe illness, leading to 250–

500 thousand deaths each year [2]. In the United States alone, seasonal influenza accounts for

more than 226,000 hospital admissions annually [3]. The spectre of an influenza pandemic,

however, poses a drastically worse scenario.

Page 13: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

2

Up to 50% of the population can be infected with influenza A virus during a pandemic year [4].

The most devasting pandemic to date, the 1918 Spanish influenza, was estimated to have killed

50 to 100 million people [5], with an estimated case fatality rate of 2–3% [6]. Case fatality rates

for the 1957/1958 Asian flu and the 1968 Hong Kong flu pandemics were estimated to be

approximately 0.2% [6], while the most recent pandemic, the 2009 swine origin influenza virus

(H1N1) pandemic, had a relatively low case fatality rate of 0.026% [7].

Most recently, 602 cases of avian influenza virus (H5N1) have been reported to the World

Health Organization (WHO) between 2003 and April 2012, of which 355 cases resulted in

fatalities [8]. However, the WHO employs stringent criteria to ascertain H5N1 disease

excluding asymptomatic individuals. A recent meta-analysis which calls the WHO H5N1

disease criteria into question reveals that 1–2% of study participants from 20 studies were

seropositive for H5N1 [10]. Therefore, the existence of mild or subclinical H5N1 infection

means the true case fatality rate is likely to be less than 60% as reported by WHO.

Furthermore, individuals with limited access to healthcare may not receive a laboratory-

confirmed diagnosis of H5N1 influenza virus infection [9].

1.2 Structure and life cycle of the influenza virion

The influenza virus genome is comprised of eight single-stranded negative-sense viral RNA

segments which encode the ten following gene products: hemagglutinin (HA), neuraminidase

(NA), matrix 2 (M2), polymerase basic protein 1 (PB1), polymerase basic protein 2 (PB2),

polymerase acidic protein (PA), nucleoprotein (NP), matrix 1 (Ml), non-structural protein 1

Page 14: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

3

(NS1) and non-structural protein 2 (NS2). An alternate reading frame in the PB1 polymerase

gene segment which encodes the protein PB1-F2 may also be generated in select influenza A

viruses [33].

The eight influenza viral segments are encapsulated by a host-derived lipid bilayer envelope in

which viral encoded glycoproteins HA and NA, as well as M2 are embedded [11]. Currently

there are 17 known subtypes of HA and 10 known subtypes of NA, which can be found in a

number of combinations on the influenza A virus surface. Subtypes H1N1, H2N2, and H3N2 are

known to circulate in humans while other subtypes are commonly found in other species

including 9 different subtypes of H5, H7, and H9 found mainly in wild birds and poultry. H7N7

influenza A viruses can infect horses and H3N8 viruses can infect both horses and dogs. Other

subtypes have been isolated from bats, pigs, marine mammals and guinea pigs [301,302].

Each influenza A viral RNA segment is structurally bound to NP to form viral ribonucleoprotein

(vRNPs), which are associated with the 3 viral polymerase proteins that form the polymerase

complex (PB1, PB2, and PA) located at the ends of the nucleocapsids. Finally, M1 forms a shell

surrounding the virion nucleocapsids, underneath the virion envelope [1].

Page 15: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

4

Figure 1.1. Structure and life cycle of influenza A virus (© Richard Burke Squires, courtesy of the Biology Image Library, ref. [12].)

The process by which influenza A virus infects host cells begins with the binding of HA to sialic

acid on host epithelial cells. The HA0 precursor protein undergoes post-translational cleavage

into the HA1-HA2 complex, a process critical to influenza virulence [13]. The virus is then

internalized to form a host cell endosome containing each RNA segment. Hydrogen ions in the

endosome are pumped through the opening of a proton pore, the M2 ion channel, which results

in endosomal acidification and subsequent release of vRNPs into the cytoplasm. Via facilitated

translocation with the aid of at least two nuclear localization sequences (NLS) contained by the

NP known as NLS1 and NLS2, the vRNPs are then imported into the nucleus of the host cell

where RNA replication, transcription and assembly of progeny vRNPs occurs [291]. Newly-

made vRNPs assemble in the nucleus and are exported into the cytoplasm. HA, NA and M2

Page 16: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

5

migrate via the golgi apparatus and anchor in the plasma membrane forming the new virion

membrane. Viral segments are then arranged so that each of the eight segments is included in a

newly formed virion. The process by which this occurs is not well understood however it is

thought to involve several protein-protein interactions between the cytoplasmic tails of the viral

integral membrane proteins, the matrix protein and the vRNPs [297]. Finally, sialylated progeny

virions gather at the host cell membrane where their sialic acid is cleaved by NA which frees the

newly assembled infectious influenza progeny virions from the host cell [11]. This process of

influenza respiratory tract virus replication peaks within 24–72 hours after illness onset [88].

1.3 Uncomplicated and severe influenza

Most influenza virus infections are uncomplicated and resolve within 7 – 10 days. Symptoms of

acute mild infection are sudden onset of high fever, coryza, cough, headache, malaise and

transient tracheo-bronchitis [4]. Individuals who are more susceptible to developing severe

disease include the elderly, children, pregnant women, immunocompromised individuals and

individuals with known underlying health conditions including asthma, chronic obstructive

airway disease, cardiovascular disease, obesity, and diabetes [4,14]. Clinically, severe influenza

is characterized by dyspnea, cyanosis, bloody (or coloured) sputum, chest pain, persistent fever

(duration > 3 days), altered mental status, and/or hypotension [15]. Severe influenza can lead to

diffuse alveolar damage with histopathologic features of acute lung injury (ALI) and the acute

respiratory distress syndrome (ARDS) [16–20], which directly contributes to influenza-

associated morbidity and mortality. ARDS is characterized by increased permeability of the

microvascular endothelium and disruption of the alveolar-capillary barrier, leading to a

proteinaceous exudate in the alveolar airspaces, accompanied by neutrophil, macrophage, and

Page 17: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

6

erythrocyte infiltration [21,22]. Post-mortem examination of the lungs of patients who

succumbed to infection with the 2009 pandemic H1N1 influenza virus revealed diffuse alveolar

damage with or without a hemorrhagic component or necrotizing bronchiolitis; severe

neutrophilic inflammation was noted in a substantial number of cases [23]. Severe infection may

also result in renal dysfunction and multi-organ failure, and secondary bacterial pneumonia may

also cause substantial illness and death, as was observed with the 1918 influenza pandemic.

1.4 Viral determinants of influenza pathogenesis 1.4.1 Hemagglutinin

High and low pathogenic avian influenza viruses (HPAIV and LPAIV, respectively) differ in

their HA protein cleavage sequence. HPAIV possesses a polybasic cleavage site

(QRERRRKKR/G) while LPAIV possesses one to two basic amino acids at this site [24–26].

The multibasic cleavage motif found in HPAIV is recognized by subtilisen-like endoproteases,

such as furin, which are present in all tissues, while the LPAIV lacking this motif is recognized

by trypsin-like enzymes preferentially expressed at the surface of respiratory and gastrointestinal

epithelia [23]. Therefore, LPAIV, although infectious, replicates in a limited number of cell

types and is therefore restricted in its ability to spread in the host, while HPAIV is able to

replicate in all tissues leading to widespread lethal systemic infection [23,26]. Hatta et al. also

found that high cleavability of HA was essential for lethal murine H5N1 infection as conversion

of the multibasic amino acid HA site to the sequence typical of avirulent avian influenza viruses

highly attenuated an H5N1 modified virus [27].

The importance of the avian HA glycoprotein in the induction of pro-inflammatory cytokines has

been demonstrated in vitro [28]. The importance of influenza virus HA in immunomodulation is

Page 18: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

7

also illustrated by the 1918 influenza pandemic, in which virus containing the 1918 HA/NA was

associated with enhanced pulmonary infiltrates and production of IFN-γ, TNF, MIP-2, and CCL3

[29].

1.4.2 Polymerase proteins including PB1-F2 peptide

PB1-F2 is a critical influenza virulence determinant for some influenza virus strains in certain

hosts [30]. The PB1-F2 peptide, encoded in the +1 reading frame of the PB1 segment of the

trimeric viral RNA-dependant RNA polymerase complex, is expressed in the highly virulent

1918 pandemic influenza virus, highly virulent murine-adapted influenza A/Puerto Rico/8/34

(A/PR/8; H1N1) virus and HPAIV [31]. It is pro-apoptotic [32] and has been associated with

increased pathogenicity and mortality in a mouse model [33]. PB1-F2 has also been implicated

in copathogenesis with bacterial pathogens such as Streptococcus pneumonia; the C-terminal

portion of the peptide contributed to acute lung injury, even in the absence of viral replication

[34]. The PB1-F2 peptide of the 2009 pandemic H1N1 influenza virus is truncated due to the

presence of several stop codons in the PB1 gene [35,36]. This may partially account for the

relatively low virulence associated with the 2009 pandemic virus. However, 2009 pandemic

H1N1 influenza virus genetically modified to express PB1-F2 had similar virulence compared to

wild-type 2009 pandemic H1N1 influenza virus in mouse and ferret models, despite elevated

levels of IFN-γ, CCL2, CCL4, and CCL5 [36]. Since the initial viral strains were sequenced,

several isolates circulating during the pandemic were found to express a 57 amino acid PB1-F2

due to a stop-to-leucine substitution at position 12. This appeared to increase virus replication in

cell culture but not pathogenicity in a mouse model [37].

Page 19: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

8

Expression of PB1-F2 was also shown to confer increased pathogenicity without altering virus

replication [38] when a serine amino acid is present at position 66 [39]. Influenza viruses with

the N66S mutation in PB1-F2 have been shown to cause enhanced production of cytokines and

associated cellular infiltrates [40]. Thus, the presence of PB1-F2 influenza virus expression is an

important contributor to influenza A virulence and is involved in immunopathology. PB2

mutations have also been noted to promote virulence and transmission, including D701N and

E627K [27,41,42]. More recently, the PB2-E158G mutation was associated with increased viral

replication and mortality in 2009 pandemic H1N1 influenza virus-infected mice as well as H5N1

virus-infected chickens [43].

1.4.3 Other viral determinants

Other virulence determinants, including influenza virus NS1 and NA, have been associated with

increased virus replication as well as inflammation [44,45]. NS1 in particular has been well-

studied, and an NS1-truncated live attenuated virus has been proposed as a vaccine candidate

[46–48]. NS1 is a multifunctional protein and several reviews outlining its role in immune

evasion, viral replication, and host cell modulation have been published [49–52]. Interferon

antagonism is a primary function of the NS1 protein, acting at both the pre- and post-

transcriptional levels of type I interferon production [53–55]. NS1 has also been shown to

directly inhibit the actions of antiviral proteins [56,57]. NS1 activity and virulence varies

depending on viral strain. For example, the NS1 in a highly pathogenic H5N1 virus from an

outbreak involving humans was shown to play a central role to the high mortality associated with

this virus [58]. Finally, NS1 has been implicated in host cell signaling, whereby the C-terminal

Page 20: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

9

domain of avian influenza viruses contains a PDZ ligand capable of binding host cell proteins

and leading to decreased survival and increased lung damage in a mouse model [59,60].

1.5 Antiviral therapy

1.5.1 Neuraminidase inhibitors, M2 ion channel inhibitors and other antivirals

While influenza vaccines may only provide moderate protection against influenza [292], annual

immunization remains the primary means of protection against influenza. However, in the event

of a pandemic, strain-specific vaccine production can take up to six months. During this time,

entire unvaccinated populations remain susceptible to a novel pandemic strain. In the interim,

antiviral drugs are important for patient management and the abrogation of transmission.

The United States Food and Drug Administration (FDA) has approved two classes of antiviral

drugs for the prevention and treatment of influenza. These include the M2 ion channel

inhibitors, also known as the adamantanes (amantadine and rimantadine) and the neuraminidase

inhibitors (NAIs) (oseltamivir and zanamivir). The first class of drugs, the adamantanes, block

the influx of H+ ions through the M2 ion channel, preventing the acid-triggered fusion reaction

and thus interfering with viral endosomal release inside the cell [61]. The second class of drugs,

the NAIs, competitively bind to the highly conserved enzyme NA active site by mimicking sialic

acid, the natural substrate of NA. This inhibits the enzyme’s key function by destroying

neuraminic acid-containing receptors, which prevents newly assembled virion cleavage and

release of progeny virions from infected cells [61].

Page 21: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

10

Because the licensure of NAIs was obtained based on studies in healthy adults with

uncomplicated seasonal influenza, it is not clear that these drugs are effective for treatment of

severe disease. A major Cochrane review which amalgamated two large reviews of oseltamivir

use for influenza in healthy adults and children, respectively, found that oseltamivir shortens the

duration of symptoms by less than a day in people with influenza-like illness (ILI) but there is no

evidence of an effect on hospitalizations [62]. It is uncertain whether oseltamivir has an effect on

complications. However, treatment with oseltamivir has been associated with a reduction in

mortality in a number of studies [91,294,295].

Because influenza A virus possesses an error-prone RNA polymerase, a high incidence of

mutation occurs and variants resistant to antiviral drugs may be selected; permissive mutations

restoring viral fitness ensure ongoing transmission and infection [65,66]. With the development

of viral resistance, the utility of adamantanes has been all but eliminated. Since 2005, nearly all

influenza A (H3N2) viruses isolated globally were resistant to adamantanes [67,68]. All 2009

pandemic influenza A (H1N1) viruses isolated globally from September 2010 to January 2011

were also resistant to adamantanes [68]. As the continued use of adamantanes further exerts

selective pressure on the development of drug resistance, the use of these drugs as mono-therapy

should be eliminated from current clinical practice.

Unlike adamantanes, the frequency of resistance to NAIs has been minimal, until recently. Since

2007, a significant increase in seasonal influenza A (H1N1) virus mutations conferring resistance

to oseltamivir have been observed [69]. The majority of these mutations have been characterized

by a histidine to tyrosine mutation at residue 274 of the NA; the glutamine at position 276 is

pushed away by the bulkier tyrosine substitute, thus disrupting the hydrophobic pocket which

Page 22: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

11

otherwise accommodates the pentyloxy portion of the oseltamivir molecule [69,70]. During the

2008–2009 influenza season, nearly 100% of tested seasonal influenza A/H1N1 virus strains in

the United States and in over a dozen other countries were found to be oseltamivir-resistant

[71,72]. As of October 2010, 313 cases of oseltamivir-resistant 2009 pandemic H1N1 influenza

virus have been identified, and these cases were mostly linked to prophylactic or therapeutic use

of oseltamivir [68,73]. Oseltamivir resistance has also been found in H5N1 virus-infected and

oseltamivir-treated patients [74,75]. However, the majority of 2009 pandemic H1N1 influenza

viruses tested from September 2010 to January 2011 remained susceptible to oseltamivir [68].

Oseltamivir is restricted to oral administration, though a phase I trial is planned by Hoffmann La

Roche for an intravenous (IV) formulation [76]. Clinical trials have also demonstrated the safety

of peramivir, an NAI developed for IV administration. Japan has recently licensed peramivir

under the name Rapiacta [77,78].

Another licensed NAI, zanamivir, has a low incidence of resistance and is currently an

alternative treatment strategy for oseltamivir-resistant strains [79]. Zanamivir’s low resistant rate

may be attributable to a low prescription rate as well as differences in the way it binds to NA

[61]. However, zanamivir is delivered to the lungs by a dry powder inhaler which may pose

difficulties for patients on mechanical ventilation [80], individuals with severe cough or dyspnea,

small children and the elderly [81]. There is also concern of sub-optimal delivery to sites of

infection in influenza virus-infected patients with pneumonia or pulmonary disease [16,80,82].

Therefore, H5N1 virus-infected patients who are viremic may not respond to NAIs as the

antiviral is unlikely to reach the necessary end-target sites, as has been observed where patients

Page 23: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

12

shed virus for many days despite combined NAI treatment [293]. Nebulization of zanamivir is

not recommended due to the formulation’s potential to cause mechanical ventilator obstruction,

which may result in death [83]. Zanamivir is currently undergoing phase II trials for parenteral

I.V. use [84,85]. CS-8958 (laninamivir prodrug), an NAI similar to zanamivir but with a

prolonged half-life, is also being studied for use by inhalation and is currently commercially

available as Inavir in Japan [86,87].

In addition to the development of influenza virus drug resistance, another important caveat to

consider with respect to antivirals includes limitations in the timing of administration [88–92].

Initiation of oseltamivir treatment within the first 48–72 hours after symptom onset is associated

with lower mortality and greater antiviral efficacy compared with treatment initiated beyond the

first 48 hours of symptom onset for H1N1, H3N2 and H5N1 infected patients [89,90]. This may

be attributed to the peak in influenza respiratory tract viral replication at 24–72 hours after illness

onset [88]. Furthermore, influenza patients in the intensive care unit or those who died from

severe influenza were less likely to have received antiviral treatment within 48 hours after the

onset of symptoms [91]. Late antiviral treatment may also be unable to prevent the development

of ARDS; 2009 pandemic H1N1 influenza virus-infected patients with ARDS who died began

oseltamivir treatment an average of 5 days after the onset of symptoms compared to infected

patients who survived without ARDS and those with mild-disease, where treatment was initiated

an average of 4 and 2 days post symptom onset, respectively [92]. However, late antiviral

treatment is still associated with a significant reduction in mortality compared to no treatment

[294].

Page 24: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

13

Other caveats include limited efficacy in cases of H5N1 virus infection [75] and limited access to

stockpiles during a pandemic [93,94]. More effective, affordable and accessible drugs to treat

severe influenza are urgently required. Other antiviral agents which may target different stages of

the viral life cycle are currently being investigated, including the polymerase inhibitor favipiravir

(T-705), HA inhibitor prokaryotic cyanovirin-N (CV-N), short interfering RNA (siRNA), and a

sialic acid receptor inhibitor, fludase (DAS-181) [95].

1.6 Investigational antiviral therapy - inhaled nitric oxide

Nitric oxide (NO) is an important cellular signaling molecule synthesized from L-arginine by

NO synthase (NOS). In the airways, NOS is present in a variety of cells, including macrophages,

vascular endothelial cells, airway epithelial cells and neurons, where NOS activity is known to

mediate neurotransmission, smooth muscle contraction and mucin secretions. There are three

types of NOS: constituent and calcium-dependant isoforms (both principally present in

endothelial cells and neuronal cells), and the inducible or calcium-independent isoform (iNOS)

[96]. NO is also a well known biological mediator in the host response to infection [96,97].

Various inflammatory stimuli such as lipopolysaccharide (LPS) and cytokines including IFN-γ

and TNF can cause high and sustained NO production by iNOS. Depending on the species,

strain, infection dose and pathogen entry route, iNOS activity can result in pro- or anti-

inflammatory responses, cytotoxicity, or cytoprotection [reviewed in 96].

In vitro, NO antimicrobial activity has been demonstrated against a variety of viruses including

ectromilia virus, vaccinia virus, herpes simplex type 1 viruses, coronavirus, and influenza A and

B viruses [98–102]. In these studies, administration of the NO donor S-nitroso-N-

Page 25: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

14

acetylpenicillamine (SNAP) to virus-infected cells significantly reduced viral burden. Lin et al.

reported that SNAP did not have any direct anti-viral capability since pre-treatment of viral

stocks with SNAP for 1 hour failed to affect virus infectivity, but rather biochemical analysis

revealed that NO exerts its antiviral effects by profound inhibition of viral RNA synthesis, viral

protein accumulation, and viral release from infected cells [103].

Severe cases of influenza are often associated with multisystem organ failure and hypoxemic

respiratory failure, including ALI/ARDS requiring ventilatory support [104,105]. Affected

individuals may receive ‘rescue’ therapies, including inhaled nitric oxide (iNO), in an attempt to

improve outcome [105]. However, iNO administration for ARDS secondary to viral pneumonia

has not been specifically reported to improve clinical outcome [104,105].

iNO therapy is currently FDA approved for the treatment of term and near-term neonates with

hypoxemic respiratory failure associated with clinical or echocardiographic evidence of

pulmonary arterial hypertension [106,107]. Variable findings have been reported for iNO

efficacy when administered at 1 ppm and up to 80 ppm. For its indicated use, iNO has been

found to increase vasodilation, transiently improve arterial oxygenation, reduce length of

mechanical ventilation, reduce oxygen requirement, and decrease length of stay in the intensive

care unit [107–109]. One study found iNO, at 30 ppm or less, decreased the spread and intensity

of lung infiltrates and improved arterial oxygen saturation in patients with severe acute

respiratory syndrome (SARS) [110]. However, systematic reviews and meta-analysis of

randomized controlled trials have shown that iNO, when used therapeutically in the management

of ARDS, does not reduce mortality [111–115]. Moreover, iNO therapy for ARDS may increase

Page 26: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

15

the risk of iNO treated patients developing renal dysfunction [114,115]. Despite this, 39% of

critical care specialists surveyed reported using iNO for the management of patients with ARDS

in Ontario, Canada [116].

1.7 Host determinants of pathogenesis

1.7.1 Cytokine dysregulation

Various host factors can influence susceptibility to influenza and progression to severe disease

including immune status, underlying health conditions and age. Recently, it has become clear

that one of the most important factors contributing to influenza-related morbidity and mortality is

inflammation. Severe influenza can be characterized as a disease which induces hyper-

production of inflammatory cytokines during host inflammation. In physiologic conditions, anti-

inflammatory cytokines are immunoregulatory molecules that control pro-inflammatory

responses. In pathological conditions, insufficient control of anti-inflammatory responses over

pro-inflammatory responses may occur and the net response of this imbalance determines

individual patient outcome. Specifically, the extent of the host inflammatory response triggered

by various influenza virus strains is positively correlated with clinical severity of influenza

including HPAIV infection and infection with reconstructed 1918 pandemic H1N1 influenza

virus in animal models. Importantly, the degree or magnitude of dysregulated cytokine

production fails to correlate with viral titers in both animal models [117–123] and in humans

[123,124].

Page 27: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

16

Cytokine dysregulation has repeatedly been observed in human influenza virus infection from

the earliest H5N1 outbreaks in 1997 onward [13,123,124]. H5N1-induced hypercytokinemia has

also been observed in mouse models [120] and in cell culture [125]. In these H5N1 studies,

levels of cytokines, including CXCL10, CCL5, CXCL9, CCL2, IL-8, IL-10, IL-6, IFN-γ, TNF,

CCL3 and soluble IL-2, were elevated compared to other influenza virus strains.

Influenza caused by seasonal H1N1 and H3N2 virus strains and the recent 2009 pandemic H1N1

influenza virus have not been associated with cytokine dysregulation, which may partially

account for the low case fatality rate of 2009 pandemic H1N1 influenza virus [7,126–128]. In

vitro experiments with seasonal H3N2 influenza virus and other non-pandemic H1N1 influenza

viruses showed no evidence of hypercytokinemia [126,127]. In vitro [128] and ex vivo [127]

experiments did not reveal cytokine dysregulation induced by 2009 pandemic H1N1 influenza

virus infection. However, To et al. found higher levels of pro-inflammatory cytokines in patients

with 2009 pandemic H1N1 influenza virus infection and ARDS [92]. This often resulted in

death compared to patients who survived without ARDS or those who suffered mild forms of the

disease, independent of viral load amongst the three groups. Taken together, these studies

suggest that 2009 pandemic H1N1 influenza virus may be associated with cytokine dysregulation

in the setting of ARDS.

The significance of each elevated cytokine in influenza virus-induced cytokine dysregulation is

not well defined. For example, Peiris et al. found significantly elevated levels of CXCL10 and

CXCL9 but no significant differences in levels of CCL2, CCL5 and IL-8 in patients with H5N1

virus infection compared to patients infected with other influenza viruses [123], while Perrone et

Page 28: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

17

al. did not observe significantly elevated levels of CXCL10 or CXCL9, but rather significantly

elevated levels of CCL3, KC (mouse IL-8), IL-1α, IFN-γ, and IL-6, in HPAIV infected mice

[120].

In order to elucidate which cytokines are most important in mediating lung pathology,

investigators have taken advantage of mice with targeted deletions of specific cytokine or

cytokine receptor genes (i.e., cytokine ‘knock-out’ mice). Research involving cytokine knock-

out mice has revealed protective or deleterious roles of certain cytokines in the host response to

influenza, which seems to vary depending on the specific strain or subtype of influenza virus

studied. CCR5-/- mice (receptor for CCL3, CCL4 and CCL5) developed massive inflammation

and severe pulmonary damage associated with decreased survival when infected with influenza

A/PR/8 virus [117]. Similarly, mortality was significantly increased or accelerated in IL-1R1-/-

mice (receptor for IL-1α and IL-1β) compared to wild-type mice when infected with influenza

A/PR/8 virus or reconstructed 1918 virus, respectively [129,130]. This suggests signaling

through IL-1R1 is protective and these observations indicate that a proper balance in immune-

mediated inflammation is necessary to achieve eradication of influenza virus while avoiding

deleterious host-mediated inflammatory tissue injury. Other research has also found similar

morbidity and mortality in CCL3-/-, IL-1R-/-, IL-6-/-, TNFR1-/-, IL-6-/-, TNF-/- or CCL2-/- mice

inoculated with avian influenza virus H5N1 compared to wild-type mice [131,132].

To the contrary, other studies have shown improved outcome in cytokine or cytokine receptor

knock-out mice. CCR2-/- mice infected with influenza A/PR/8 virus had reduced lung injury and

inflammation and mortality compared to wild-type controls [117]. The chemokine receptor

Page 29: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

18

CCR2 binds its ligand CCL2 to cause the migration of monocytes and the development of

monocyte-derived inflammatory cells in the lungs which has been associated with influenza virus

induced lung injury and subsequent mortality [117,122,133]. This finding is in contrast to that

observed with CCL2-/- mice reported by Salomon et al. [132]. A further study by Lin et al.

showed reduced lung pathology and an 89% survival increase of CCR2-/- mice compared to

influenza virus-infected wild-type controls [133]. However, Aldridge et al. showed CCR2-/- mice

infected with A/PR/8 had comparable morbidity and mortality to infected wild-type controls

[134]. Subsequent studies using a CCR2 inhibitor, PF-04178903, administered prophylactically

and twice daily post-infection resulted in 100% survival of influenza A/PR/8 virus-infected mice

compared to 25% survival and decreased pulmonary immune pathology in untreated mice [122].

Therefore, CCR2 inhibition may be effective in reducing influenza virus-induced lung pathology

and overall mortality.

It is important to bear in mind that conclusions based on experiments employing mice with

specific targeted genetic deletions (‘knock-out’) may be misleading as other downstream factors

may be modified and ‘hidden from view’. Functional redundancy amongst signaling pathways

may result in host compensation which could mask the immunomodulatory benefits of single

cytokine depletion. To account for such host responses, Perrone et al. examined TNF-R1, TNF-

R2, and IL-1R1 triple knock-out mice [121]. When infected with a lethal dose of H5N1, these

mice exhibited decreased inflammatory cell infiltrates and cytokine levels, reduced morbidity,

and a significant delay in mortality compared to wild-type controls [121]. Taken together, this

research suggests that reducing overall inflammation rather than targeting individual cytokines

may prove to be a superior strategy for decreasing influenza-related morbidity and mortality.

Page 30: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

19

1.8 Immunomodulatory therapy

The outcome of influenza virus infection is determined by both viral and host factors; therefore,

it would be logical to investigate the efficacy of therapeutic strategies which target the host in

combination with antiviral therapy. Such therapeutic strategies could potentially target either

signaling pathways to reduce viral replication or host-mediated inflammation in an effort to

dampen deleterious tissue/organ dysfunction and injury. Importantly, several experimental

studies in mice show that mortality is reduced by immunomodulatory agents without

concomitant antiviral treatment and without any decrease in virus replication, implying

modulation of host inflammation alone may be sufficient to improve influenza virus-infected

patient outcome or provide additional benefit when used in combination with antiviral agents.

A number of FDA-approved drugs have received investigational interest as immunomodulatory

strategies for the treatment of influenza (described in sections 1.8.1-1.8.8). All of these drugs,

with the exception of CCR-2 antagonist PF-04178903, are produced as generic agents and, thus,

would be relatively inexpensive and available for deployment in the event of a pandemic. In

addition, these agents could potentially circumvent waning efficacy in the face of increasing

antiviral resistance. Chapter 1.8 will focus on immunomodulatory agents for the treatment of

severe influenza which target dysregulated host inflammatory responses (summarized in Table

1.1).

Page 31: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

20

Table 1.1. Summary of investigational immunomodulatory agents for treatment of influenza (reprinted with permission from © Expert Reviews Ltd, ref. [135].)

Page 32: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

21

1.8.1 Glucocorticoids

Glucocorticoids are a group of corticosteroids which exert anti-inflammatory activity via binding

to cytoplasmic receptors that subsequently interact with glucocorticoid response elements to

regulate transcription of a number of genes [136]. Clinical trials using prolonged low-to-

moderate dose glucocorticoid therapy for ARDS, a prominent feature of severe influenza, have

demonstrated that glucocorticoid therapy enhances the anti-inflammatory activity of the

glucocorticoid-activated glucocorticoid receptor; a major regulator of inflammation in ARDS

[reviewed in 137]. Consistent with these clinical observations, glucocorticoids suppress cytokine

levels [138] and IFN-γ induced pro-inflammatory gene expression including cyclooxygenase-2

(COX-2) expression in human bronchial epithelial cells in vitro [136].

Evidence to date supporting the use of glucocorticoids for the treatment of severe influenza is

inconclusive. In a rat model of experimental H3N2 influenza, daily doses of the glucocorticoid

triamcinolone acetonide combined with an NAI suppressed IFN-γ induction, resulting in

decreased macrophage activation and pulmonary inflammation [139]. However, survival

benefits were not reported in this study, and a study of experimental H5N1 influenza in mice

found no difference in mortality between daily corticosterone-treated mice and controls [132]. In

humans, glucocorticoid therapy has yielded equivocal H5N1 infection with uncertain effects. In a

randomized trial in Vietnam, all 4 H5N1 virus-infected patients who received glucocorticoids

died [15]. Moreover, in a clinical series reported from Thailand, only 2 of 8 H5N1-infected

patients treated with methylprednisolone survived [140]. In contrast, a recent study, while

lacking an ‘influenza infected with ARDS’ control group, reported that glucocorticoid treatment

improved ALI in individuals with ARDS secondary to severe 2009 pandemic H1N1 influenza

Page 33: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

22

[141]. In view of these mixed results, the use of glucocorticoids as adjunctive therapy to

antiviral drugs for severe influenza may warrant further study in the form of a randomized

clinical trial, but there is currently insufficient evidence to endorse its routine use.

1.8.2 Macrolides

Macrolides, such as erythromycin, azithromycin, and clarithromycin, are bacteriostatic protein

synthesis inhibitors that exert broad immunomodulatory activities including downregulation and

upregulation of pro- and anti-inflammatory cytokines, respectively, modulation of leukocyte

recruitment, and improvement of macrophage phagocytic function, but without causing global

immunosuppression [142–144]. In vitro, clarithromycin reduces cytokine content in culture

supernatant of seasonal H3N2 influenza virus-infected tracheal epithelial cells [145].

Erythromycin treatment of H2N2 influenza virus-infected mice increased survival from 14% to

57%, and reduced inflammatory cell counts [146].

Secondary bacterial pneumonia is frequently observed during influenza pandemics, resulting in

increased influenza viral titers and lung inflammation [147,148]. As non-lytic antibiotics,

macrolides may be important in clearing influenza bacterial co-infection in addition to

decreasing host inflammation. Ampicillin, a β-lactam antibiotic targeting the bacterial cell wall

has been shown to clear bacterial infection from influenza virus-infected lungs, however

ampicillin use did not alter mortality rate in a mouse model of severe pneumococcal pneumonia

with bacteremia following influenza [149]. This prompted investigation into the use of

azithromycin, which significantly improved outcome of a milder bacteria-influenza co-infection

compared to ampicillin by dampening inflammatory responses caused by ampicillin-induced

Page 34: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

23

bacterial lysis, due to their non-lytic antimicrobial activity [150]. Therefore, macrolides may be

advantageous when used in secondary bacterial pneumonia following influenza, and clinical

trials are required to demonstrate a benefit from macrolide use in severe influenza. However, a

note of caution is warranted because of the risk that widespread routine use of antibiotics for

treatment of individuals with influenza may promote the development of problematic

antimicrobial resistance in the long-term [151]. Also, safety concerns including increased risk of

cardiovascular death have been associated with macrolides [296].

1.8.3 TNF inhibitors

Researchers have explored the use of therapeutic agents to lower host inflammation by directly

targeting specific cytokines. TNF-neutralizing monoclonal antibodies and soluble TNF-receptor

fusion proteins are important in managing inflammation caused by immune-mediated disorders,

such as inflammatory bowel disease and rheumatoid arthritis [152,153] and increasing survival

of LPS injected mice [154]. In contrast, anti-TNF strategies have not been beneficial in the

treatment of other inflammatory conditions such as sepsis [155].

Experimental use of anti-TNF therapy in mouse models of influenza has shown inconsistent

results. Few studies have reported that TNF neutralizing strategies for severe influenza in mouse

models reduce lung lesion severity, pulmonary inflammatory cell infiltrates, and T-cell cytokine

production, however with no survival benefit reported [131,132,156,157]. TNFR-1-/- mice

survived longer than wild-type mice when infected with reconstructed 1918 influenza virus,

suggesting that anti-TNF therapy might provide some therapeutic benefit in severe influenza.

However, it should be noted that all mice died in both groups [130].

Page 35: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

24

Anti-TNF therapy for severe influenza in humans has not been studied in a controlled clinical

trial, and its failure for the treatment of sepsis casts doubt on its potential benefit in severe

clinical influenza. Additionally, increased risk of bacterial infection and infections with rare

intracellular pathogens are well-described complications of anti-TNF therapy [158]. Thus,

overall experimental evidence to support the clinical use of anti-TNF therapy for severe

influenza is currently lacking, especially when considering the attendant risks.

1.8.4 Cyclooxygenase-2 inhibitors

Cyclooxygenase (COX) enzymes, which catalyze the conversion of arachidonic acid into

prostaglandins, play important roles in modulating immune responses and inflammation

[118,159,160]. Widely used clinically for their analgesic, anti-inflammatory and antipyretic

properties, the COX inhibitors consist of the well known non-selective non-steroidal anti-

inflammatory drugs (NSAIDs), such as ibuprofen, which inhibit both COX-1 and COX-2, and

the selective COX-2 inhibitors, such as celecoxib, which target COX-2 specifically [118].

In vitro but not necessarily in vivo data support the use of COX-2 inhibitors for severe influenza.

Pronounced COX-2 upregulation has been reported following in vitro infection of macrophages

with H5N1 virus and in lung tissue at autopsy from patients who died of H5N1 virus-associated

disease [161]. In vitro, COX-2 inhibitors, including celecoxib, have been shown to suppress

influenza virus-induced inflammatory cytokine production [128,161]. In mouse models of H3N2

influenza, inflammation was reduced and survival increased in COX-2-/- compared to wild-type

mice [118], suggesting a benefit from COX-2 deficiency. However, experiments in knock-out

mice may yield misleading results secondary to effects on unidentified downstream pathways. In

Page 36: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

25

murine models of influenza, the use of the pharmacological COX-2 inhibitor celecoxib did not

confer any survival advantage [162,163]. These studies suggest that COX-2 inhibitors alone

may not be effective agents for severe influenza therapy; however, retrospective studies to

determine whether COX-2 inhibitor use was associated with improved clinical outcome may still

be warranted.

Research examining the use of COX-2 inhibitors in combination with anti-viral drugs has shown

promising results. Combination therapy including zanamivir, celecoxib and the anti-

inflammatory drug mesalazine significantly improved survival of mice infected with H5N1

influenza virus compared to treatment with zanamivir alone [162]. Importantly, the triple

combination therapy reduced levels of IL-6, IFN-γ, TNF and MIP-1 in infected mice compared

to zanamivir treatment alone. Single daily use of either immunomodulator or the mesalazine and

celecoxib combination failed to confer any survival advantage. It should also be noted that

combination therapy of mesalazine and celecoxib was not investigated further in this study. The

experimental data suggests that COX-2 inhibitors may provide additional benefit when used

adjunctively to existing antiviral strategies. Further research is necessary to strengthen the

evidence in support of combined celecoxib, mesalazine, and antiviral therapy use for severe

influenza.

Animal models of ALI have shown COX-2-derived mediators to exert a protective role against

inflammation-induced lung injury, in part via enhanced lipoxin signaling [164]. Lipoxins are a

unique class of arachidonic acid-derived lipid pro-inflammatory mediators of inflammation

which display in vivo inhibition of polymorphonuclear cell activation, cytokine release and

Page 37: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

26

angiogenesis [reviewed in 165]. Inhibition of lipoxin signaling has been associated with

enhanced lethality of H5N1 virus infection in mice [166]. This finding, when considering the

unconvincing in vivo evidence to support the use of COX-2 inhibitors as monotherapy, provides

indication of the potential use for lipoxin agonists to reduce inflammation in severe influenza and

improve patient outcome.

1.8.5 HMG-CoA reductase inhibitors

A group of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors,

commonly known as statins [167], were proposed as a potential therapeutic strategy to reduce

influenza virus-induced inflammation in 2005 [93]. Statins have been shown to modulate host

immunity by inhibiting activation of immune effector cells, such as macrophages, and antigen

presentation by macrophages to T cells, as well as affecting endothelial function through the

inhibition of reactive oxygen species (ROS) production and upregulation of endothelial nitric

oxide synthase (eNOS) [168, reviewed in 169]. Statins also antagonize high mobility group box

1 protein (HMGB-1), a potent pro-inflammatory cytokine, and suppress CCR2 gene expression

[170–173]. Advantages of this class of drug include its widespread availability, relatively

inexpensive cost, and long shelf life [174].

No study has reported statin reduced mortality in murine models of influenza. In humans, a

cohort study conducted during the 1996–2006 influenza seasons revealed a 16% reduction in 30

day pneumonia mortality with the use of statins, although interpretation of these results may

have been complicated by confounding factors [94,174]. Among patients admitted with

laboratory-confirmed influenza, those who used statins before or during hospital stay had lower

Page 38: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

27

odds of dying within 30 days and after adjusting for other variables they reported a 41%

reduction in mortality [175]. It is important to note that adverse drug reactions, such as

autoimmune hepatitis and myopathy, are associated with statin therapy. Taken together, there is

currently insufficient evidence to recommend routine clinical use of statins for the management

of influenza. A randomized clinical trial of acute statin therapy in hospitalized statin-naïve,

influenza virus-infected patients may be warranted [176].

1.8.6 High mobility group box 1 protein antagonists

High mobility group box 1 (HMGB-1) is a 215 amino acid protein that functions intracellularly

as a transcription factor and is highly conserved among species [177]. When released either

passively from dying, necrotic cells or actively from macrophages, HMGB-1 functions in the

extracellular milieu as a signal of tissue injury/damage [178]. Implicated in the pathogenesis of

severe sepsis [179,180] and arthritis [181], HMGB-1 is a potent mediator of pro-inflammatory

activity in the presence of inflammatory cytokines such as IL-1β, IFN-γ or TNF [171,172].

Given these properties, HMGB-1 antagonists could potentially prove useful in the treatment of

‘the cytokine storm’ in severe influenza.

However, HMGB-1 is characterized as a relatively late mediator of inflammation in sepsis

[171,180,182] and, thus, its inhibition may not impact clinical outcome in severe influenza. For

example, plasma levels of HMGB-1 in H2N2 influenza virus-infected mice peaked at day 9 post-

infection (P.I.), a full 2 days after peak mortality was reached [183]. Furthermore, treatment

with the HMGB-1 antagonist ethyl pyruvate failed to reduce mortality [183]. Further studies of

Page 39: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

28

other HMGB-1 antagonists will provide a better understanding of the role of HMGB-1 in severe

influenza.

Glycyrrhizin, a compound extracted from liquorice root, has been reported to bind and inhibit

HMGB-1 [184,185], increase T-cell IFN-γ production, and reduce endocytic viral uptake [186–

188]. Glycyrrhizin treatment increased survival in murine models of influenza when

administered prophylactically 1 day prior to infection and continued on days 1 and 4 P.I. [187].

However, protection against LPS induced lung injury in mice receiving glycyrrhizin pre-

treatment has been attributed in part to suppression of COX-2 and iNOS expression [189].

Therefore, additional study is needed to determine whether inhibition of HMGB-1 is in fact the

mechanism by which glycyrrhizin increases survival in experimental influenza. Metformin and

statins also antagonize HMGB-1 [170,190]. Molecular modeling of the HMGB-1-glycyrrhizin

interaction may aid in the development of potent anti-HMGB-1 specific drugs which could

potentially be useful as adjunctive therapy to antiviral drugs in the treatment of severe influenza.

1.8.7 Peroxisome proliferator activated receptor agonists

Peroxisome proliferator activated receptors (PPARs) are a family of lipid activated transcription

factors, consisting of PPAR-α, PPAR-δ, and PPAR-γ, which are key regulators of inflammation

and lipid metabolism [reviewed in 191]. PPAR activation antagonizes crucial inflammatory

pathways such as NF-κB, AP1 and STAT [reviewed in 192], which results in a reduction of

inflammatory cytokines such as IL-6 and IFN-γ [193].

Page 40: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

29

Gemfibrozil, a PPAR-α agonist belonging to the fibrate family of drugs, was discovered over 40

years ago to reduce plasma lipid and cholesterol level and is widely used as treatment for

hypertriglyceridemia [194,195]. Gemfibrozil reduces the release of inflammatory cytokines

including TNF, IL-6 and IFN-γ [196,197] and has been proposed for the treatment of severe

influenza [194,196]. Daily injections of gemfibrozil administered in a treatment strategy

following the onset of experimental H2N2 influenza in mice increased survival from 26% to

52% [194]. In contrast, gemfibrozil failed to increase survival of H5N1 influenza virus-

infected mice when administered 48 hours post-inoculation [162]. These studies suggest the need

to conduct further research on the use of gemfibrozil for severe influenza.

Thiazolidinediones, also known as glitazones, are a class of PPAR-γ agonists, including

pioglitazone and rosiglitazone. Pioglitazone has been shown to suppress inflammation in sepsis

[198] and improve insulin sensitivity in patients with impaired glucose tolerance [199] by

reducing CCL2 expression, which is the primary ligand for CCR2 and an important mediator of

host inflammation [199,200]. In experimental murine models, prophylactic administration of

pioglitazone to A/PR/8 influenza virus-infected mice has been reported to increase survival by

20–40% [134,201]. Aldridge et al. found the benefits of pioglitazone administration pre- and

post-influenza virus inoculation in mice to be independent of pulmonary viral load [134].

Therefore, the benefit of pioglitazone therapy can be attributed to modulation of host

inflammatory responses.

Rosiglitazone has also recently been shown to increase survival in experimental murine

influenza. Specifically, Moseley et al. reported 100% survival of A/PR/8 influenza virus-

Page 41: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

30

infected mice treated with rosiglitazone compared to 30% survival of influenza virus-infected

controls [201]. Moseley et al. also reported 20% survival of influenza A/California/04/09 (a

2009 pandemic H1N1 strain) virus-infected mice treated with rosiglitazone compared to 100%

fatality of infected controls. These results are by far the most promising reported to date in the

search for an effective immunomodulatory strategy to improve clinical outcome in severe

influenza. Because of the encouraging results observed with PPAR agonists in the treatment of

influenza, biochanin A, a component of red clover with both PPAR-α and PPAR-γ agonistic

properties, has been proposed as a potentially effective immunomodulatory agent for the

treatment of influenza [195].

1.8.8 AMP-activated protein kinase agonists

Increases in the intracellular AMP-to-ATP ratio trigger the activation of AMP activated protein

kinase (AMPK), an enzyme which exerts anti-inflammatory effects upon activation [202].

Aminoimidazole carboxamide ribonucleotide (AICAR) is an AMP-activated protein kinase

activator. In a murine model of LPS-induced ALI, administration of AICAR resulted in AMPK

activation in the lungs which diminished the severity of lung injury as shown by decreased

pulmonary edema, and reduced bronchoalveolar lavage fluid levels of IL-6 and TNF [202].

Moseley et al. reported a 40% increase in survival of A/PR/8 influenza virus-infected mice

treated with AICAR while combination therapy including AICAR and pioglitazone improved

survival by 60% [201].

Metformin, another AMPK agonist and known HMGB-1 antagonist, is routinely used in

combination with pioglitazone for patients with diabetes mellitus and has been shown to improve

Page 42: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

31

survival in murine endotoxemia [190]. Metformin and pioglitazone have been shown to have

synergistic effects in type 2 diabetes [203]. This compound is also commercially available.

Therefore, metformin and AICAR might prove effective in the treatment of severe influenza

when used either alone, or in combination with PPAR-γ agonists and/or antiviral drugs.

1.9 Investigational immunomodulatory therapy – mesenchymal stromal cells (MSCs)

In 1968, Friedenstein and colleagues described a murine bone marrow cell population with the

ability to form colonies and differentiate into several cell types [204]. These cells, now referred

to as mesenchymal stromal (stem) cells (MSCs), represent a heterogenous subset of non-

hematopoeitic pluripotent stromal cells with multilineage potential. MSCs can be isolated from

embryonic tissue, adipose tissue, liver, muscle and dental pulp; however, adult bone marrow

remains the most common source of MSCs for pre-clinical and clinical studies [205,206]. A

position paper released by the International Society for Cellular Therapy [207], defines MSCs by

the following 3 minimum criteria:

1. MSC’s must adhere to plastic when maintained in standard culture conditions.

2. MSCs must possess the ability to differentiate into osteoblasts, chondroblasts and

adipocytes in vitro.

3. MSCs must lack several lineage markers including haematopoietic and endothelial

markers (CD34, CD45, CD11b, CD14, CD79α, CD19, and HLA-DR), and must possess

several mature stromal cell markers (CD105, CD73, CD90).

Page 43: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

32

While the first two criteria are mostly universally accepted, the third criteria is complicated by

varying MSC markers identified between species and strains of species; therefore, the ability to

identify a unique MSC cell surface marker remains unresolved [208].

Initial clinical interest in MSCs focused on their ability to differentiate into injured cell types as a

means to improve outcome in pre-clinical models of disease. However, more recent pre-clinical

studies demonstrate that while MSCs can reduce injury, engraftment rates are low [209-211].

Rather, MSCs are now thought to exert their effects by controlling inflammatory and

immunological reactions, and secreting multiple paracrine factors.

1.9.1 MSCs and the immune system

In vitro, MSCs have been shown to suppress both innate and adaptive immune responses

including an ability to suppress effector and cytotoxic T-cell functions, impair cytolytic potential

of natural killer (NK) cell functions, induce regulatory T-cells, and modulate dendritic cell (DC)

and macrophage function. A number of studies have reported that the addition of MSCs to

primary mixed lymphocyte co-cultures can suppress T-cell proliferation [212-214]. MSCs may

also inhibit T-cell proliferation indirectly by inhibiting monocytes from maturing into DCs [215].

DCs play a fundamental role in antigen presentation to naïve T-cells following DC maturation,

which can be induced by pro-inflammatory cytokines or pathogen associated molecules [216].

In a study by Jiang et al., the allostimulatory ability of MSC-treated mature DCs on allogeneic T-

cells was impaired [215]. Here, MSCs were shown to induce an immunosuppressive DC

phenotype by inhibiting their production of pro-inflammatory cytokines TNF, IFN-γ, and IL-12

and inducing DC production of anti-inflammatory cytokine IL-10 [215].

Page 44: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

33

MSCs may also affect NK cells of the innate immune system. NK cells play an important role in

antiviral and anti-tumor immune responses due to their cytolytic properties and pro-

inflammatory cytokine production. Under low NK-to-MSC ratios, MSCs alter the phenotype of

NK cells and suppress proliferation, cytokine secretion, and cytotoxicity against major

histocompatability complex (MHC) class I expressing targets [217]. MSC-mediated inhibition of

NK cells is related to the downregulation of NK cell-activating surface receptors, while

conversely, cytokine-activated NK cells can kill MSCs in vitro [218]. Some of these effects

require cell-to-cell contact, whereas others are mediated by soluble factors, including

transforming growth factor - β (TGF-β) and prostaglandin E2 (PGE2). This suggests diverse

mechanisms may be responsible for MSC-mediated NK-cell suppression. Neutrophils are also

important cells in innate immunity which are rapidly mobilized and activated to kill

microorganisms during infection. After binding to bacterial products MSCs have been shown to

delay neutrophil apoptosis through an IL-6 dependant mechanism [219].

Finally, MSCs have been shown to alter macrophage phenotype, in vitro [220] and in vivo [221].

Monocytes preferentially differentiate into IL-10 secreting alternatively activated

(immunosuppressive) macrophages (M2 macrophages) in the presence of MSCs in vitro [220].

Macrophages isolated from MSC-treated septic mice produced significantly higher amounts of

anti-inflammatory cytokine IL-10 than those from non-MSC treated mice, suggesting a

temporary reprogramming of monocyte and therefore macrophage function [221].

Page 45: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

34

Figure 1.2. Mesenchymal stem cell immune cell targets (reprinted with permission from © Bentham Science Publishers, ref. [222].)

The clinical use of MSC therapy is likely to rely heavily on the use of allogeneic and not

autologous MSCs. This is because autologous cells may require several weeks to expand in

culture before they could be used in the clinic; by contrast, allogeneic cells would be readily

available “off-the-shelf” as they could be manufactured in advance for immediate use in the

treatment of acute disease. Also, allogeneic transplantation would ensure that the cells are

derived from healthy immunocompetent individuals and thus improving their likelihood of

functionality.

Page 46: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

35

However, the use of allogeneic MSCs would require “universality” – that is, a low immunogenic

profile so that the cells could evade the recipient’s immune responses. Expression of human

leukocyte antigens (HLAs) – the major histocompatability complex (MHC) molecules I and II –

present antigenic peptides generated in the cytosol to CD8+ T-cells or in intracellular vesicles to

CD4+ T-cells, respectively. Low human MSC expression of MHC Class I and lack of MHC

class II and classic co-stimulatory molecules such as B7-1, B7-2, CD80, CD86, CD40 and

CD40L allow human MSCs to evade host immune responses [223]. Therefore, human MSCs

could potentially be administered to patients without the need for HLA matching. Indeed,

patients who underwent MSC allotransplants experienced no anti-allogeneic MSC antibody

formation or T-cell sensitization [224].

However, under certain stimuli including low level IFN-γ co-culture, MSCs may upregulate

expression of MHC class II to the cell surface (can detect MHC Class II intracellularly in basal

conditions) and therefore act as antigen-presenting cells (APCs) to activate antigen specific

immune responses [225,226]. Studies have also demonstrated that infusion of allogeneic mouse

MSC can elicit a host immune response and lead to MSC rejection [227,228]. Therefore, MSCs

may not be intrinsically immunopriviliged and may be unable to serve as a “universal donor” in

MHC mismatched recipients, but this may be specific to mouse rather than human-derived cells.

Further investigation is necessary to understand the complex interaction between MSCs and the

immune system.

The use of MSCs for therapeutic applications has also attracted attention for their ability to

“home” to sites of inflammation upon IV injection. Homing has been described as the process by

Page 47: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

36

which cells migrate to and engraft in the tissue which they will exert their effects. Since MSCs

may localize to the injured tissue, concerns of MSCs inducing systemic immunosuppression -

which could lead to major complications - may be lessened. In a mouse model of multiple organ

failure, green fluorescent protein (GFP)-tagged MSCs homed to sites of injury within numerous

tissues, with MSCs proportionally homing to regions of most severe disease [229]. Akin to

leukocyte trafficking, a key indicator of MSC homing is the increase in inflammatory

chemokines at the site of injury.

In basal conditions, MSCs express several chemokine receptors located on their surface,

including CCR2, CXCR3, CXCR4, CXCR5, and CX3CR1 as well as RNA encoding the

following chemokines – CCL2, CXCL8, SDF-1(CXCL12), and cytokines TGF-β and IL-6 [230-

232]. Studies have shown that MSCs respond to chemoattractant factors including CCL2,

CXCL12, CX3CL1, CCL3, and IL-8 leading to MSC chemotaxis [233,234]. However

chemokine / cytokine and receptor expression varies in the literature, possibly due to different

culture conditions, including passage and the initial heterogeneity of the isolated cells obtained

from bone marrow [232,235,236]. It is not known which chemokines and their receptors are

most critical for MSC migration.

1.9.2 MSC duality – immunostimulant or immunosuppressant?

MSCs are located at the interface of the bone marrow cavity and on the peripheral surface of the

main sinusoidal vessels [237]. This position gives MSCs a unique ability to function as

“gatekeepers” to the marrow by interacting with all cells transiting in and out of the marrow and

by regulating immune responses as the first line of defense. The role of MSCs in immune

Page 48: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

37

regulation has been show to be homeostatic, namely, they possess an ability to serve as either

immunostimulatory or immunosuppressive, depending on the environment.

The presence of IFN-γ and other cytokines in the MSC microenvironment play an important role

in induction of immunosuppression and MSC homing. Ren et al. have demonstrated that the

immunosuppressive effect of human MSCs depends on IFN-γ in the co-presence of either TNF,

IL-1α or IL-1β [238]. Under co-stimulation with these cytokines in vitro, human MSCs secreted

large amounts of indoleamine 2,3-dioxygenase (IDO) and chemokines which in turn drove T-cell

migration in proximity with MSCs, whereas high level IDO production suppressed T-cell

function. Aggarwal and Pittenger demonstrated that the level of PGE2 - an important MSC-

secreted protein - is significantly upregulated by MSCs in the presence of TNF [213]. IFN-γ

mediated MSC activation has also been demonstrated in vivo in a model of graft-vs-host disease

(GvHD) where recipients of IFN-γ-/- T-cells did not respond to MSC treatment [239].

Mechanistically, IFN-γ may act directly on MSCs by upregulating B7-H1, an inhibitory

molecule on the surface of MSCs [240].

On the contrary, low levels of IFN-γ or the absence of specific proteins in the MSC

microenvironment may cause MSCs to behave as APCs [241]. Human MSCs may also act as

immune-enhancers when IDO is knocked-down. Therefore, a clinical scenario where MSCs may

encounter insufficient pro-inflammatory cytokines in vivo could potentially lead to MSC-

mediated immunostimulation. It may then be difficult to determine the point in the disease

progression at which administering MSCs could achieve an immunsuppressive response.

Page 49: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

38

1.9.3 Effects of MSCs in pre-clinical models

MSC mediated immunopsuppresion after infusion in vivo has been documented in a diverse

array of pre-clinical animal models of disease. MSC-based prophylactic and treatment strategies

have yielded significant therapeutic benefits in pre-clinical models of a variety of inflammatory

diseases, including rheumatoid arthritis [243], sepsis [221,244] and ALI [209,210,245–248].

However, several studies report no MSC-mediated immunosuppression or therapeutic benefit in

pre-clinical models of GvHD, organ transplantation and rheumatoid arthritis [249-251].

Furthermore, MSCs used in combination with the universal immunosuppressant cyclosporine A

for the treatment of GvHD resulted in accelerated graft rejection in mice [251]. The clinical trial

registry at the National Institute of Health indicates over 200 registered clinical trials currently

investigating the use of MSCs in humans as a therapy for diverse diseases including Crohn’s

disease, GvHD, osteogenesis imperfecta and multiple sclerosis [252].

Most importantly, MSCs have been shown to reduce dysregulated inflammatory responses, to

improve alveolar fluid clearance, and to maintain lung epithelial and endothelial integrity in the

lung during pulmonary inflammation and injury in murine models [209,210,245–248, reviewed

in 253]. These models consist of lung injury induced by either: (1) bleomycin - an antitumour

agent known to cause lung injury by inducing oxidative stress and elevated inflammation, or (2)

endotoxin / LPS – a major constituent of the outer cell wall of gram-negative bacteria. In these

murine models of ALI, the beneficial effects of MSC therapy have been attributed to the

secretion of growth factors, cytokines and lipid mediators [reviewed in 253 and discussed in

section 1.9.3].

Page 50: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

39

1.9.4 MSC derived paracrine mediators

While the initial driving force behind therapeutic interest in MSCs was for their regenerative

capacity, it is their paracrine properties and ability to modulate host inflammation that markedly

increased the known range of MSC therapeutic applications and attracted even greater scientific

interest in MSC research. MSCs secrete a vast array of soluble mediators and growth factors

which can modulate the host response. Importantly, these findings have been observed

independent of direct MSC-to-cell contact. For example, concentrated MSC-conditioned media

reversed multi-organ dysfunction syndrome in a rat model [254]. While many

immunosuppressive mediators are suspected to be important for treating lung injury, the proteins

under greatest investigation include IL-1 receptor antagonist (IL-1ra) [255], soluble TNF

receptor (sTNFR) [256], IDO [214], IL-10 [221], angiopoeitin-1 (Ang-1)[298], TNF stimulated

gene 6 protein (TSG-6) [258,259] TGF-β [260], PGE2 [213,221], and keratinocyte growth factor

(KGF) [248]. These proteins may exert their positive effects by immunomodulation, improving

alveolar fluid clearance, or reducing lung endothelial permeability.

IL-1ra, sTNFR-1, IL-10, IDO, PGE2 and TSG-6 may be important in MSC-mediated

immunomodulation. In a bleomycin-induced animal model of lung injury, injected MSCs

decreased inflammatory responses and prevented the development of lung fibrosis. These effects

were attributed to MSC secretion of IL-1ra [255]. In another pre-clinical mouse model of

disease, human MSCs administered to LPS induced sepsis in mice attenuated systemic

inflammation by secreting sTNFR1 [256]. sTNFR1 binds to TNF to neutralize its inflammatory

activity. MSC secretion of IL-10 is also important. MSCs can mediate downregulation of pro-

inflammatory cytokines TNF and MIP-2 and upregulation of anti-inflammatory cytokine IL-10

Page 51: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

40

in BAL (BAL) fluid of endotoxin induced lung injury in mice [247]. MSC secretion of PGE2

may be implicated in reprogramming macrophages to increase IL-10 production resulting in

reduced mortality and improved organ function in pre-clinical sepsis [221]. Human MSC

released IDO has been attributed to the immunosuppressive properties of MSCs as well [214]. In

vitro, MSC released IDO – an important protein in tryptophan metabolism - inhibits T-cell

proliferation directly and indirectly via IDO induced monocyte differentiation into M2–like

immunosuppressive cells [220]. Improved myocardial infarction and lung injury in MSC treated

mice has also been attributed to MSC expression of TSG-6 [258,259]. TSG-6 is a TNF induced

anti-inflammatory protein.

In addition to MSC secreted immunomodulatory proteins, the effects of MSCs may be mediated

by endothelial/epithelial specific growth factors including Ang-1 and KGF which are potentially

important in maintaining lung-vascular endothelial integrity. The integrity of the lung

microvascular endothelium is essential to prevent the influx of protein-rich fluid from the plasma

which may further aggravate the ability of the lung epithelium to reduce alveolar edema. Ang-1,

a ligand for the endothelial-specific receptor tyrosine kinase Tie2 [261], is a potent mediator of

angiogenesis that functions to prevent leakage and promote blood vasculature quiescence via

strengthening of endothelial cell junctions and downregulation of surface adhesion molecules,

such as vascular cell adhesion molecule-1 (VCAM-1) and E-selectin [262–265]. In vitro, the

effects of MSCs have been shown to be mediated by Ang-1 which resulted in reduced epithelial

protein permeability in cultured human alveolar type II cells [298]. KGF, another important

MSC soluble mediator, has also been shown to reduce lung injury in animal models of

pulmonary edema [266]. KGF may stimulate the proliferation and differentiation of alveolar type

Page 52: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

41

II cells, which in turn may promote lung repair during injury. In an ex vivo perfused human

lung, intra-bronchial instillation of human MSCs 1 hour following endotoxin-induced lung injury

restored alveolar fluid clearance, in part by the secretion of KGF [248].

It is important to note that inhibition of most MSC secreted molecules does not result in

complete loss of MSC immunosuppressive function. For example, upon MSC knockdown of

TSG-6, Danchuk et al. reported a decrease but not complete loss of MSC mediated

immunosuppression in vivo [259]. Also the relative importance of each MSC secreted protein is

dramatically varied between studies. Lastly, administering isolated proteins implicated as the

most important MSC soluble factor does not usually fully recapitulate the immunosuppressive

function of MSC administration. For example, MSCs were more effective in mouse model of

bleomycin induced lung injury, than recombinant or virally delivered IL-1ra [255]. Therefore, it

is likely that no single molecule exerts an exclusive role and MSC mediated effects are likely the

result of concerted actions mediated by many important soluble mediators in combination.

1.10 Mouse models of influenza

To date, mouse models of influenza have demonstrated utility in the guidance of novel

therapeutic investigation and intervention for the treatment of severe influenza in humans. As in

humans, influenza virus virulence in mice is dependent on the virus subtype and strain. In

addition, the lethal dose, replication kinetics, and inflammatory response are affected by a

complex and multigenic host component [303]. Symptoms of severe disease are comparable in

mice and humans as both may feature reduced blood oxygen saturation, lung edema, increased

Page 53: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

42

lung viral load, pulmonary immune cell infiltrates and elevated cytokine production and

hemorrhage [267]. Also, onset of symptoms positively correlates with viral titers [268].

However, experimental mice may not show signs of fever, and both cyanosis and dyspnea are

not easily detectable in many models aside from H5N1 influenza [267]. The most widely

accepted indicator of disease progression in influenza murine models is weight loss [269,270]

Antiviral compounds as well as several investigational immunomodulatory agents have shown

efficacy against influenza and host inflammation in mice [271, previously discussed].

Advantages of mouse models include well characterized immune responses and a wide

availability of murine immune reagents. Additionally, mice are inbred so experimental results are

usually reproducible; however, results may be misleading due to genetic variability in the human

population. For example, while zanamivir has demonstrated efficacy against H5N1 in mice

[271], antivirals have shown limited efficacy against H5N1 in humans [75] and the efficacy of

oseltamivir is not entirely clear [62]. Conversely, statins have been shown to reduce disease

severity in humans when their use was evaluated in retrospective studies, while statins are

ineffective at improving outcome in murine models (previously discussed). Therefore, while

mouse models are important in guiding the development of novel therapies for use in humans,

results should be interpreted with caution.

Historically, influenza A/PR/8 virus (H1N1) and A/WSN/33 virus (H1N1) are the most

commonly used strains in experimental influenza [272]. While both A/PR/8 and A/WSN/33 are

known to cause severe inflammation, A/WSN/33 is better known for its neurotropism and its

capacity to cause systemic infection [273]. Both influenza A/PR/8 virus and A/WSN/33 virus

have been serially passaged and adapted to mice in order to cause severe disease. Therefore, it is

important to bear in mind that conclusions based on results obtained using these strains may be

Page 54: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

43

misleading due to molecular changes acquired during adaptation. Nonetheless, A/PR/8 has been

employed in the evaluation of many immunomodulatory agents (e.g. PPARs) for the treatment of

severe influenza [134,201].

Page 55: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

44

1.11 Research aims and objectives

While the vast majority of influenza virus infections resolve without complications, millions of

individuals develop severe disease each year and are at risk of death. Given the importance of

the innate host response in severe influenza – particularly inflammation - adjunctive therapies to

complement anti-viral treatment strategies are in need. In the face of rising antiviral drug

resistance, novel anti-influenza drugs are also sorely in need. The aim of this work was to

investigate two novel therapeutic treatment strategies for severe influenza. This included an

assessment of iNO therapy as an antiviral agent and exploration of mesenchymal stem cell

therapy as an immunomodulatory strategy.

In Chapter 2, we examined whether iNO administration could reduce viral load and improve

survival in a murine model of severe influenza. Our specific aims were as follows:

1. To determine if iNO delivered either continuously or intermittently could improve

survival in an experimental murine model of severe influenza;

2. To establish whether iNO therapy could reduce the influenza viral burden in an

experimental murine model of severe influenza.

We found that iNO therapy failed to improve survival or decrease viral load in experimental

influenza.

In Chapter 3 we examined whether MSC therapy could modulate the host response to infection

in an in vivo mouse model of severe influenza and therefore improve outcome. The specific

aims of this work were as follows:

Page 56: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

45

1. To establish and characterize the influenza A/PR/8 virus mouse model of severe

influenza which develops inflammation and acute lung injury;

2. To determine whether MSC therapy can improve survival and/or dampen host

inflammation and lung injury in a mouse model of severe influenza;

3. To assess the ability of MSC prophylaxis, with or without MSCs pre-incubated with

cytokines, to improve survival in experimental influenza model;

4. To test whether combining oseltamivir with MSCs is effective in reducing mortality

and/or dampening host inflammation and lung injury in experimental influenza model.

We found that MSCs were not effective in dampening host inflammation and acute lung injury in

experimental influenza. Thus, MSCs were unable to decrease morbidity and mortality in our

experimental model of influenza.

Page 57: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

46

Chapter 2: Inhaled Nitric Oxide Antiviral Therapy f or Severe Influenza

2.1 INTRODUCTION

Further investigation of antiviral strategies which have shown promising results against influenza

A viruses in vitro but have not been investigated in vivo are warranted. One of these agents

which may be effective when used against influenza virus is NO. In vitro, NO has antimicrobial

activity against a wide range of viruses, including influenza A virus; therefore, we hypothesized

that inhaled NO (iNO) would increase survival in vivo by reducing the viral load in C57Bl/6

mice infected with a lethal dose of influenza A/WSN/33 virus (H1N1). iNO delivery would

provide a safer and easier delivery method than administration of NO donors such as SNAP, as

iNO is currently approved for treating term and near-term neonates with hypoxemic respiratory

failure [106,107].

iNO was delivered to influenza virus-infected mice either continuously or intermittently at 80 or

160 ppm, respectively, using both prophylactic and post-infection treatment strategies. These

dosing regimens were chosen for the following reasons:

1. iNO delivery in hypoxemic respiratory failure is already approved up to a dose of 80

parts per million (ppm) [106,107].

2. Gaseous NO at a high dose of no less than 160 ppm and with five hours of continuous

exposure, can elicit a non-specific antimicrobial response against a broad range of

microorganisms in vitro [274].

Page 58: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

47

In vivo, 160 ppm iNO treatment would result in NO binding to hemoglobin to form

methemoglobin, resulting in reduced oxygen transport and hypoxemia, as well as the potential

for elevated levels of the harmful NO metabolite NO2. However, Miller et al. have shown that

iNO in an intermittent delivery regimen of 160 ppm for 30 min every 3.5 hours can prevent

methemoglobinemia and reduce the potential of host cell toxicity in vivo [Miller C, personal

communication]. Therefore, we chose to deliver 160 ppm iNO in a similar manner.

We chose to investigate the use of iNO over other antiviral agents for several reasons. Firstly, it

will be available off patent as of 2013 and therefore will be relatively cheap to administer as

treatment for severe influenza. Secondly, it is relatively easy to administer with a breathing

mask in resource poor areas. Lastly, iNO would likely evade the development of influenza drug

resistance that is often associated with antiviral therapy, due to its non-specific antiviral effects

including enzyme nitrosylation, such as nitrosylation of proteases (e.g. reverse transciptases and

ribonucleotide reductase) containing cysteine residues and viral-encoded transcription factors

that are involved in replication [97, 98].

Page 59: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

48

2.2 MATERIALS AND METHODS

Murine influenza model

Animal use protocols were reviewed and approved by the University Health Network Ontario

Cancer Institute Animal Care Committee, and all experiments were conducted in accordance

with institutional guidelines in an animal biosafety level 2 facility. Female C57Bl/6 mice, 9–11

weeks old, were obtained from Jackson Laboratories (Bar Harbor, ME, USA) and maintained

under pathogen-free conditions with a 12-hour light cycle. On day 0, while under light

isofluorane anesthesia, experimental mice were infected via nasal instillation with 1000 plaque

forming units (PFU) of influenza A/WSN/33 virus (H1N1) (stock kindly provided by Dr.

Eleanor Fish, University Health Network/University of Toronto) in 50 µl PBS. Weight was

recorded daily for a maximum of twelve days P.I., and mice were sacrificed when euthanasia

criteria was met (greater than 20% weight loss). Lung tissue was harvested for analysis on day 5

P.I..

In Vivo NO Delivery

Prophylactic or post-infection iNO therapy was initiated either 1 hour prior to or 4 hours P.I.,

respectively. Mice were placed in flow-through chambers with free access to food and water and

received either compressed room air, continuous NO at 80 ppm +/-5ppm mixed with compressed

room air, or intermittent NO for 30 min every 3.5 hours at 160 ppm+/-5ppm mixed with

compressed room air. Soda lime (200 g) was supplied to each chamber, and gas flow was

maintained at 10–12 L/min to scavenge and minimize NO2 levels, respectively. NO2 levels were

Page 60: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

49

limited to <2 ppm for continuous iNO therapy and <8 ppm for intermittent iNO therapy. NO and

NO2 levels were measured using an AeroNOX machine (Pulmonox Medical, AB, CA).

Lung influenza viral load analysis

Lungs were harvested and frozen at -80°C. Lungs were thawed, weighed, and homogenized in 1

ml PBS for 30 sec using a Tissue Miser homogenizer (Fisher Scientific, ON, CA). Lung

homogenates were spun at 10,000xg for 10 min, aliquoted, and stored at -80°C for viral yield

titration. Influenza A/WSN/33 viral yield in lung homogenates was quantified by plaque assay in

Madin-Darby canine kidney (MDCK) cells (ATCC, VA, USA). MDCK cells (ATCC, Manassas,

VA, USA) were maintained in Eagle’s MEM (ATCC) supplemented with 10% FBS and 0.25%

gentamicin. All cell lines were cultured at 37oC with 5% CO2. MDCK cells were plated at a

concentration of 8x106 cells/plate in 6 well culture plates. 12–24 hours later, medium was

removed and MDCK cells were washed twice with PBS. 10-fold dilutions of lung homogenates

were added to MDCK cells in 500 µL Eagle’s MEM, in duplicate, and incubated at 37°C with

5% CO2 for 1 hour with plates rocked every 15 min. After incubation, 1 mL of serum-free 2X

Eagle’s MEM supplemented with 8 µl/ml trypsin, 60 µl/ml of 7.5% sodium bicarbonate and 20

µl/ml antibiotics, combined with 1 ml of 1.2% agarose, was added to each well. Once the agarose

set, plates were incubated at 37°C for 42–72 hours until syncytia were observed. Plates were

fixed with Carnoy’s fixative (3:1, methanol:glacial acetic acid) for 30 min then stained with

0.1% crystal violet in 20% ethanol to visualize plaques. Viral load was expressed as plaque

forming units per gram of lung tissue (PFU/g).

Page 61: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

50

Statistical Analysis

Logrank tests were performed on Kaplan-Meier survival curves. Significant differences in

weight loss between groups were assessed by two-way analysis of variance (ANOVA) and

Bonferroni post-tests were performed. Student’s t-tests were carried out on viral yield data to

assess significant differences (p ≤ 0.05) between experimental groups.

Page 62: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

51

2.3 RESULTS

2.3.1 Continuous iNO at 80 ppm decreased survival and intermittent high dose iNO at 160 ppm did not increase survival of influenza A virus-infected mice

We evaluated the ability of iNO to improve survival of influenza A/WSN/33 virus-infected mice.

Experimental C57Bl/6 mice were inoculated intranasally with an 80–100% lethal dose of

A/WSN/33 virus (1000 PFU). At 5 days P.I., the majority of mice in all experimental groups

experienced weight loss (Figure 2.1A and 2.2A). At 7 days P.I., mice began to reach euthanasia

criteria (≤80% of day 0 weight), and by day 10 P.I., most mice were euthanized (Figure 2.1B and

2.2B). If 20% weight loss was not met by day 10 P.I., the infection typically resolved, and

surviving mice gained weight.

Figure 2.1. Prophylactic iNO therapy increased weight loss and decreased survival of C57Bl/6 mice infected with influenza A/WSN/33 virus. C57Bl/6 male mice were infected with 1000 PFU A/WSN/33 virus and administered continuous NO at 80 ppm or compressed room air starting 1 hour prior to infection (n=17–18/group, 2 independent pooled experiments). (A) Shown are the weight loss data up to the day of first death. Mice receiving iNO displayed a significant reduction in weight compared to infected controls (Two-way ANOVA p < 0.001 with Bonferroni post-tests; **, p < 0.01 on day 6 and 7 P.I.). Error bars represent standard deviations. (B) Survival curve. iNO significantly reduced survival of treated mice compared to infected controls as shown by Kaplan-Meir survival curves; **, logrank test: P < 0.01).

0 2 4 6 880

85

90

95

100

105

110

iNO prophylaxisAIR

**

**

Day P.I.

% o

f o

rig

inal

wei

gh

t (m

ean

±± ±± S

D)

0 2 4 6 8 10 120

20

40

60

80

100

iNO prophylaxisAIR

**

Day P.I.

Per

cen

t su

rviv

al

A B

Page 63: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

52

Weight loss over the course of infection was accelerated in mice administered continuous iNO at

80 ppm starting 1 hour prior to inoculation compared to infected control mice administered

compressed room air (P < 0.001) (Figure 2.1A). Continuous iNO administered at 80 ppm starting

1 hour prior to inoculation significantly decreased survival of A/WSN/33 virus-infected mice

compared to infected control mice administered compressed room air (P < 0.01). During the

course of infection, 100% of continuous iNO treated mice were euthanized compared to 80% of

infected control mice (Figure 2.1B). Intermittent iNO administrated at 160 ppm for 30 min

intervals every 3.5 hours starting either 1 hour prior to or 4 hours P.I. resulted in similar weight

loss kinetics (Figure 2.2A) and consequent survival kinetics (Figure 2.2B) of infected mice

compared to infected control mice administered compressed room air.

Page 64: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

53

Figure 2.2. Prophylactic and post-infection intermittent iNO does not alter weight loss kinetics or survival of C57Bl/6 mice infected with influenza A/WSN/33 virus. C57Bl/6 male mice were infected with 1000 PFU A/WSN/33 virus and administered NO intermittently at 160 ppm for 30 min intervals every 3.5 hours starting either 1 hour prior to infection or 4 hours P.I.. Infected control mice were administered compressed room air (n=9–10/group). (A) Shown are the weight loss data up to the day of first death. No difference was observed (Two-way ANOVA). Error bars represent standard deviations. (B) Survival curve. No difference was observed (logrank test).

2.3.2 Continuous or intermittent iNO administration does not reduce lung viral load

As NO at high concentrations has been shown to decrease the viral load of infected cells in vitro

(Miller C, personal communication), we examined whether iNO could reduce the viral load of

influenza virus-infected mice. iNO was administered starting 1 hour prior to influenza

A/WSN/33 virus infection and continued either continuously at 80 ppm or intermittently at 160

ppm for 30 min every 3.5 hours until mouse lungs were harvested at peak influenza viral load in

the lungs (determined to be day 5 P.I. based on preliminary studies, data not shown). Since iNO

was administered both prior to and for 5 days P.I., we were able to test whether iNO at

intermediate (80 ppm) or high concentration (160 ppm) could prevent either viral entry or viral

0 2 4 6 870

80

90

100

110iNO prophylaxisiNO post-infection

AIR

Day P.I.

% o

f o

rig

inal

wei

gh

t (m

ean

±± ±± S

D)

0 2 4 6 8 10 120

20

40

60

80

100

AIRiNO prophylaxisiNO post-infection

Day P.I.

Per

cen

t su

rviv

al

A B

Page 65: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

54

replication in vivo, and thereby reduce viral load. Continuous iNO administered at 80 ppm,

intermittent iNO administered at 160 ppm, and administered compressed room air yielded

similar lung viral loads of infected mice on day 5 P.I. (Figure 2.3A and B, respectively).

Therefore, iNO administered both continuously and intermittently failed to reduce lung viral load

of infected mice, compared to infected control mice administered compressed room air.

Figure 2.3. Intermittent high dose iNO prophylactic therapy failed to decrease viral load of C57Bl/6 mice infected with influenza A/WSN/33 virus. Lungs were collected 5 days post-influenza A/WSN/33 virus infection (1000 PFU) from mice treated with (A) continuous iNO at 80 ppm or compressed room air starting 1 hour prior to infection (n=5/group) or (B) intermittent iNO at 160 ppm or compressed room air for 30 min intervals every 3.5 hours starting 1 hour prior to infection (n=5/group). Error bars represent standard deviations. Lung viral load was quantified for all experimental groups by plaque assay on MDCK cells.

105

106

107

intermittent iNO AIR

Day 5 P.I.

PF

U/g

lun

g t

issu

e

105

106

107

continuous iNO AIR

Day 5 P.I.

PF

U/g

lun

g t

issu

e

A B

Page 66: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

55

2.4 DISCUSSION

In this report, we investigated the application of iNO therapy as an antiviral strategy for

prophylaxis and treatment of severe influenza. We found that iNO was unable to reduce the viral

burden in the lungs of influenza virus infected mice when administered continuously or

intermittently throughout the course of infection. We also showed that iNO, administered either

intermittently or continuously at varying concentration, was unable to reduce morbidity and

mortality in experimental severe influenza.

Typically, iNO is administered at initial doses of 5–20 ppm in randomized controlled trials and

observational studies for neonatal hypoxic respiratory failure [107]. Although FDA-approved at

concentrations up to 80 ppm, no specific dose of iNO has been proven more advantageous than

another [107,113]. Rather, methemoglobinemia, defined as 7% methemoglobin by Davidson et

al. was more likely to occur at higher concentrations [276]. We speculated that

methemoglobinemia may account for the decrease in survival observed in our study with

continuous iNO administration at 80 ppm. However, we could not draw this conclusion as

methemoglobin levels were not measured. NO2 concentrations were measured daily over the

course of infection and kept below 2 ppm as is acceptable in humans, however, lung toxicity may

still explain these results as the toxic threshold in mice may be lower. On the other hand, given

previous in vitro findings by McMullen et al. [274], 80 ppm may also have been too low of a

concentration to provide an antiviral effect.

A high dose of NO at 160 ppm was administered intermittently, not to target the airway vessels

specifically, but rather to induce an antimicrobial effect while avoiding the harmful effects of

high dose continuous iNO delivery. iNO administered to influenza virus-infected mice in this

Page 67: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

56

manner, either prophylactically or therapeutically, failed to improve survival of infected mice,

change the course of weight loss, or decrease the lung viral load, compared to control mice

administered compressed air. Therefore, although administration of high dose intermittent iNO

may have reduced the harmful side-effects of NO, antimicrobial activity was not observed in

vivo. However, the variation in weight loss over the course of infection within either the iNO

treated or untreated groups, as indicated by large and overlapping standard deviations, may

suggest that perhaps too few animals were studied to identify a true effect of iNO therapy.

In conclusion, despite the demonstrated antimicrobial activity of NO against influenza A virus in

vitro, the results of this study do not support the use of iNO as a prophylactic or treatment

strategy to reduce viral burden or improve clinical outcome in severe influenza in vivo.

Furthermore, it may be difficult to achieve virucidal concentrations of NO in the airways using

iNO at concentrations that are safe in the living host.

Page 68: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

57

Chapter 3: Mesenchymal Stromal Cell Immunomodulatory Therapy for Severe Influenza

3.1 INTRODUCTION

MSCs offer considerable promise as a novel influenza treatment strategy. While previous

strategies to inhibit a specific component of the immune response in influenza have met with all

but failure, a more complex overall immunomodulatory effect demonstrated by MSCs may be

more effective in decreasing influenza associated morbidity and mortality. In addition, MSCs

may be able to reprogramme the immune response to reduce destructive host inflammation while

maintaining a sufficient inflammatory response to control virus replication. Also, MSCs may

enhance the repair of lung injury and restore epithelial and endothelial integrity, either by direct

MSC cell contact or secretion of various paracrine mediators including KGF and Ang-1.

Indeed, several reports have suggested that mesenchymal stem cells (MSCs) could exert a potent

immunosuppressive effect in pre-clinical models of acute lung injury, and thus may have a

therapeutic potential for other inflammatory / lung injury dependent pathologies [209,210,245-

248]. Therefore, we aimed to establish whether MSCs could be used to control overzealous host

inflammatory responses and decrease acute lung injury in experimental severe influenza, a major

cause of morbidity and mortality in severe influenza.

Most experimental evidence to date regarding the use of MSC treatment for ALI comes from

studies that employed mouse MSCs (mMSCs) [209,210,245,247]. However, it is critical that the

Page 69: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

58

efficacy and mechanism of human MSCs (hMSCs) be studied in depth before proceeding to

clinical trials so that the human physiological response could be accurately predicted. Evidence

suggests that the mechanism by which hMSCs exert their effects varies from that of mMSCs

[238]. Therefore, MSC investigators have more recently employed hMSCs in pre-clinical study

[256,258,259]. Likewise, we chose to evaluate the use of hMSCs rather than mMSCs in our

experimental model of severe influenza.

As basal levels of MSC chemokines / cytokine expression can be modified by pre-stimulation

with IFN-γ and TNF [231,238], we sought to assess whether cytokine pre-incubated hMSCs

could improve survival in our experimental influenza model. We also examined the effect of

hMSCs in combination with antiviral therapy, oseltamivir, for the two following reasons. Firstly,

any future MSC therapy for severe influenza patients will be administered in combination with

antiviral therapy, as is standard treatment. Secondly, oseltamivir might have important effects on

the host inflammatory environment and therefore effect MSC functionality and

migration/homing. Because inflammation and ALI/ARDS are major determinants of morbidity

and mortality in severe influenza, MSC therapy represents a promising adjunctive

immunomodulatory strategy to improve outcome in severe influenza and warrants investigation.

Page 70: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

59

3.2 MATERIALS AND METHODS

Murine influenza model

Animal use protocols were reviewed and approved by the University Health Network Ontario

Cancer Institute Animal Care Committee, and all experiments were conducted in accordance

with institutional guidelines in an animal biosafety level 2 facility. Male C57Bl/6 mice, 8–10

weeks old, were obtained from Jackson Laboratories (Bar Harbor, ME, USA) and maintained

under pathogen-free conditions with a 12-hour light cycle. On day 0, while under light

isofluorane anesthesia, experimental mice were infected by nasal instillation with 425 EID50 of

influenza A/PR/8 virus (H1N1) (stock kindly provided by Dr. David Kelvin, University Health

Network/University of Toronto) in 50 ul PBS. Weight was recorded daily for a maximum of

twelve days P.I., and mice were sacrificed when euthanasia criteria was met (greater than 20%

weight loss). BAL fluid, lung tissue, and blood were harvested for analysis.

Cells

Human mesenchymal stromal cells (hMSCs) were provided by the Texas A&M Health Science

Center College of Medicine Institute for Regenerative Medicine at Scott & White. hMSCs

(isolated from a 24 year old male donor) were quickly thawed to 37°C and plated for 24 hours in

α-MEM with 2–4 mM L-glutamine supplemented with 100 U/ml penicillin, 100 ug/ml

streptomycin and 16.5% Fetal Bovine Serum (FBS). After 24 hrs, plates were washed with PBS

and cells were lifted by incubation with trypsin/EDTA for 2 min at 37°C and re-plated at 60

cells/cm2. Cells were incubated for 7–10 days for each subsequent passage (P). P3–P4 hMSCs

were lifted as described above and spun at 300xg for 5 min at 4°C with 10 ml of media. Media

Page 71: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

60

was then discarded and cells were spun twice at 300xg, 4°C with 10 ml of fresh Dulbecco’s

Phosphate Buffered Saline (PBS; Gibco/Invitrogen, Burlington ON, CA). After last wash, cells

were re-suspended in 1 ml fresh PBS and total cell count was determined via hemocytometry.

hMSCs were re-suspended in PBS at 2.5x 106 cells/ml and 100 ul was injected via the tail vein

into experimental mice on day -2, 0, 2, and 5 P.I.. Injections were performed using 26 1/2 gauge

needles and a typical mouse restrainer.

MDCK cells (ATCC, Manassas, VA, USA) were maintained in Eagle’s MEM (ATCC)

supplemented with 10% FBS and 0.25% gentamycin. All cell lines were cultured at 37oC with

5% CO2.

Reagents

Oseltamivir phosphate was extracted from Tamiflu capsules (Roche, Switzerland) and

administered to mice via oral gavage. Experimental mice were administered 5 mg/kg in 100 ul

ddH20, twice daily, every 12 hours, beginning either 2 or 5 days P.I. for a maximum of 5 days

[as described in 277]. Recombinant human IFN-γ and TNF were purchased from R&D Systems

(Minneapolis, MN, USA) and hMSCs were pre-incubated for 24 hours prior to cell harvest at 10

and 3 ng/ml, respectively [as described in 220].

BAL fluid analysis

BAL fluid of both lungs was obtained by instillation and aspiration of three consecutive 500 ul

aliquots of PBS. BAL fluid was spun at 800xg at 4°C for 5 min. Supernatant from the first

lavage was removed and stored at -80°C for further analysis. Red blood cells in BAL fluid cell

pellet were lysed with 250 ul red blood cell lysis buffer for 5 min then spun again at 800xg for 5

Page 72: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

61

min at 4°C. Combined cell pellets from all washes per mouse were re-suspended in 200 ul PBS.

Total cell numbers were determined using a hemocytometer. BAL fluid concentrations of

CCL3, CCL2, CXCL10, CCL5, mouse keratinocyte-derived cytokine (KC) and IFN-γ were

determined by sandwich ELISA (R&D Systems, Minneapolis, MN, USA) as per manufacturer’s

instructions. BAL fluid total protein concentration was measured using a BCA protein assay,

and BAL fluid IgM and albumin concentration was determined by sandwich ELISA (Bethyl

Laboratories, Montgomery, TX, USA) as per manufacturer’s instructions.

Lung homogenate analysis

Lungs were harvested and frozen at -80°C. Lungs were thawed, weighed, and homogenized in 1

ml PBS for 30 sec using a Tissue Miser homogenizer (Fisher Scientific, ON, CA). Lung

homogenates were spun at 10,000xg for 10 min, aliquoted, and stored at -80°C for viral yield

titration. Influenza A/PR/8 viral yield in lung homogenates was quantified by plaque assay in

MDCK cells (ATCC, VA, USA) as described in section 2.2.

Blood Analysis

Infected mice and uninfected controls were anesthetized with intraperitoneal (IP) injection of

ketamine (100 mg/kg) and xylazine (5 mg/kg) on day 7 post infection. Cardiac puncture was

performed where approximately 600 µL of whole blood was isolated from each mouse. Samples

were kept at room temperature for 30 min then stored on ice until centrifugation at 13,000 rpm,

4°C, for 10 minutes. Serum was aliquoted and stored at -80°C for further cytokine analysis.

Cytokine levels were determined as described above.

Page 73: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

62

Statistical Analysis

Analysis was performed using GraphPad Prism software. Kaplan-Meier survival curves were

compared using the logrank test. Differences between groups were assessed by one- or two-way

analysis of variance (ANOVA) with Bonferroni post-tests.

Page 74: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

63

3.3 RESULTS

3.3.1 Influenza A/PR/8 virus infection is characterized by a high mortality rate, elevated inflammation and acute lung injury

To establish a lethal model of severe influenza, experimental C57Bl/6 mice were infected

intranasally with 425 EID50 influenza A/PR/8 virus. This dose resulted in a mortality rate of

80%, where death was established according to standard euthanasia criteria (≤80% of day 0

weight) (Figure 3.1B). At 6 days P.I., the majority of mice in all experimental groups

experienced weight loss while at 7 days P.I., mice began to reach euthanasia criteria (Figure

3.1A and B). By 10 days P.I., most mice were euthanized. If 20% weight loss was not met by

day 10 P.I., the infection typically resolved, and surviving mice gained weight.

Figure 3.1. Male C57BL/6 mice infected with 425 EID50 influenza A/PR/8 virus develop increased morbidity and mortality. Eight week-old male C57Bl/6 mice were infected with 425 EID50 influenza A/PR/8 virus. (A) Weight loss is expressed in percentage of original weight over the course of infection (n=10). Error bars represent standard deviation. (B) Kaplan-Meir survival curve (n=10).

0 2 4 6 8 10 1270

80

90

100

110

Day P.I.

% o

f ori

gin

al w

eig

ht (

mea

n±± ±±

SD

)

0 2 4 6 8 10 120

20

40

60

80

100

Day P.I.

Per

cen

t su

rviv

al

A B

Page 75: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

64

In order to characterize influenza A/PR/8 virus infection as severe influenza which develops

elevated host inflammation, BAL fluid, lung homogenate, and serum were collected from

C57BL/6 mice on days 0, 2, 4, 5, and 6 over the course of infection and examined for total

number of inflammatory cells, as well as cytokine and chemokine protein content. Specifically,

CCL2, CCL5, KC, CXCL10, and IFN-γ levels were quantified, as these proteins have been

shown to be significantly elevated in and most commonly associated with severe influenza

compared to mild or uncomplicated influenza [135]. BAL fluid inflammatory cells began to

increase on day 4 P.I. until 6 days P.I., one day prior to the first death (Figure 3.2A). BAL fluid

levels of CCL2, CCL5, KC and CXCL10 began to rise at approximately 4 days P.I. and

continued to increase until 6 days P.I. (Figure 3.2B). BAL fluid IFN-γ began to increase 6 days

P.I., 2 days following the increase in CCL2, CCL5, KC and CXCL10. Lung homogenate levels

of CCL2, CCL5, KC, CXCL10, and IFN-γ followed a similar trend as is observed in BAL fluid,

while serum levels remained low or below the limit of detection throughout the course of

infection.

In order to characterize influenza A/PR/8 virus infection as severe influenza which develops

acute lung injury (ALI), BAL was performed on C57BL/6 mice on days 0, 2, 4, 5, and 6 P.I.

Total protein, and more specifically, IgM, are indicative of alveolar-capillary membrane barrier

leak and, therefore, were quantified in BAL fluid [18]. Levels of total protein and IgM were

elevated starting 4 days P.I., and continued to rise until the first day when euthanasia criteria was

met (Figure 3.2C).

Page 76: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

65

Figure 3.2. Male C57BL/6 mice infected with 425 EID50 influenza A/PR/8 virus develop lung inflammation and acute lung injury. (A) Total inflammatory cell count was measured by haemocytometry in BAL fluid of influenza virus-infected mice on days 0,2,4,5, and 6 P.I. (n=5-7/group). (B) A panel of cytokines and chemokines was examined by ELISA in the serum, lung homogenate and BAL fluid of influenza virus-infected mice on days 0,2,4,5, and 6 P.I. (n=5-7/group). (C) Total protein and IgM, standard markers of acute lung injury, were measured by BCA assay and ELISA, respectively, on days 0,2,4,5, and 6 P.I. (n=5-7/group). Protein levels and cell count are presented as median with interquartile range (IQR).

0 1 2 3 4 5 60

1000

2000

3000 IFNγγγγ

Day P.I.

BA

L/S

eru

m/L

ung

pg

/ml

±± ±± I

QR

0 1 2 3 4 5 60

500

1000

1500

2000

0

1000

2000

3000

4000

5000

6000CXCL10

Day P.I.

BA

L/S

eru

m p

g/m

l±± ±±

IQR

Lu

ng

pg

/ml±± ±± S

D

0 1 2 3 4 5 60

200

400

600

800

0

5000

10000

CCL5

Day P.I.

BA

L/S

eru

m p

g/m

l±± ±±

IQR

Lung

pg/m

l±± ±± SD

0 1 2 3 4 5 60

100

200

300

400

500

600

700

0

250

500

750

1000

1250CCL2

Day P.I.

BA

L/S

eru

m p

g/m

l±± ±±

IQR

Lun

g p

g/m

l + S

D

0 1 2 3 4 5 60

100

200

300

400

500

0

1000

2000

3000

4000

5000KC

Day P.I.

BA

L/S

eru

m p

g/m

l±± ±±

IQR

Lu

ng

pg

/ml +

SD

0 1 2 3 4 5 60

500

1000

1500

2000

2500

Total Protein

Day P.I.

BA

L u

g/m

l±± ±±

IQR

0 1 2 3 4 5 60

500

1000

1500

2000IgM

Day P.I.

BA

L n

g/m

l±± ±±

IQR

CBAL

SerumBAL

Lung Homogenate

BAL

SerumBAL

Lung Homogenate

SerumBAL

Lung Homogenate

SerumBAL

Lung HomogenateSerumBAL

Lung Homogenate

A

0 1 2 3 4 5 60

250

500

750

1000Inflammatory Cell Count

Day P.I.To

tal

# o

f ce

lls

in B

AL

(x1

03 )±± ±±

IQR

B

Page 77: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

66

3.3.2 Mesenchymal stem cell therapy fails to improve survival and/or dampen host inflammation and lung injury in experimental severe influenza

2.5x105 hMSCs were administered either 4 hours prior to infection (day 0), early (day 2), or late

(day 5) in the course of infection. A dose of 2.5x105 hMSCs was chosen since this dose was

effective in a report by Mei et al. [210]. MSCs were obtained from the Texas A&M Health

Science Center College of Medicine Institute for Regenerative Medicine at Scott & White and

were reported by the Centre as meeting the MSC defining criteria proposed by the International

Society for Cellular Therapy (207, data not shown).

In the lethal influenza A/PR/8 virus mouse model, hMSCs administered prior to, early, or late in

the course of infection, resulted in similar weight loss kinetics (Figure 3.3A) and consequent

survival kinetics (Figure 3.3B) of infected mice compared to infected control mice administered

PBS. No difference in lung viral titer was observed between experimental groups (Figure 3.3C).

Page 78: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

67

Figure 3.3. hMSCs fail to improve survival, alter weight loss kinetics or affect lung viral load in experimental severe influenza. Eight week-old male C57Bl/6 mice were administered 2.5 x 105 hMSCs (passage 3), via the tail vein, either 4 hours prior to influenza A/PR/8 virus infection (425 EID50), 2 days post infection (P.I.), or 5 days P.I. Weight was recorded daily. No significant differences in (A) weight loss kinetics (Two-way ANOVA, n=18-20/group, 2 pooled experiments) or (B) survival (logrank test, n=18-20/group, 2 pooled experiments) were observed. Error bars represent standard deviation. (C) lungs were collected 7 days P.I and quantified via plaque assay on MDCK cells. No significant differences were observed (One-way ANOVA, n=5/group). Error bars represent standard deviation.

hMSC Day 0 P.I.hMSC Day 2 P.I.hMSC Day 5 P.I.PBS

survival mice weight loss

A B

hMSC Day 0 P.I.hMSC Day 2 P.I.hMSC Day 5 P.I.PBS

survival mice weight loss

103

104

105

106

107

Day 7 P.I.

hMSCs Day 0 P.I.hMSCs Day 2 P.I.hMSCs Day 5 P.I.PBS

PF

U/g

lu

ng

tis

sue

+ S

D

C

0 2 4 6 8 10 120

20

40

60

80

100

Day P.I.

Per

cen

t su

rviv

al

0 2 4 6 8 10 1270

75

80

85

90

95

100

105

Day P.I.

% o

f ori

gin

al w

eig

ht (

mea

n±± ±±

SD

)

Page 79: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

68

To assess whether hMSCs could dampen lung inflammation, experimental mice administered

hMSCs or PBS were sacrificed on day 7 P.I. and BAL fluid levels of CCL2, CXCL10, KC,

CCL5, and IFN-γ were quantified. All BAL fluid cytokine and chemokine levels were similar

for hMSC treated mice compared to control mice (Figure 3.4). Cytokine and chemokine levels

were below the limit of detection in BAL fluid of uninfected mice. To assess whether hMSCs

could decrease acute lung injury in experimental influenza, we tested the BAL fluid for markers

of acute lung injury. Similarly, there was no difference in BAL fluid total protein, IgM, and

albumin levels on day 7 P.I. between experimental groups (Figure 3.5).

Page 80: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

69

Figure 3.4. hMSCs fail to alter lung inflammation in experimental severe influenza. Eight week-old male C57Bl/6 mice infected with 425 EID50 influenza A/PR/8 virus and treated with 2.5x105 hMSCs (passage 3) were sacrificed on day 7 P.I. BAL was performed. No significant difference in BAL fluid level of selected cytokines / chemokines was observed between mice treated early (day 2 P.I.) or late (day 5 P.I.) in the course of infection, compared to infected control mice (One-way ANOVA, n=5/group). Error bars represent standard deviation. ND = nondetectable.

0

200

400

600

800

1000

1200

1400

Day 7 P.I.

CCL2

ND

BA

L

pg

/ml

+

SD

0

250

500

750

1000

Day 7 P.I.

CXCL10

ND

BA

L p

g/m

l +

SD

0

100

200

300

400

500

Day 7 P.I.

CCL5

ND

BA

L p

g/m

l +

SD

0

25

50

75

100

125

150

175

Day 7 P.I.

KC

ND

BA

L p

g/m

l +

SD

0

1000

2000

3000

4000IFNγγγγ

ND

Day 7 P.I.

BA

L p

g/m

l +

S

D

hMSC Day 5 P.I.hMSC Day 2 P.I.PBSUninfected

Page 81: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

70

Figure 3.5. hMSCs fail to alter acute lung injury in experimental severe influenza. Eight week-old male C57Bl/6 mice infected with 425 EID50 influenza A/PR/8 virus and treated with 2.5x105 hMSCs (passage 3) were sacrificed on day 7 P.I. BAL was performed. No significant difference in total protein, IgM, or albumin, was observed between mice treated early (day 2 P.I.) or late (day 5 P.I.) in the course of infection, compared to infected control mice (One-way ANOVA, n=5/group). Error bars represent standard deviation. ND = nondetectable.

Next we tested whether hMSCs could decrease morbidity in a non-lethal model of A/PR/8

influenza virus infection. Since a dose of 425 EID50 A/PR/8 virus resulted in 80% mortality, we

hypothesized that administering hMSCs in a milder infection model would allow greater

opportunity for hMSCs to provide a therapeutic effect. Mice were administered a sub-lethal dose

of A/PR/8 virus (150 EID50) and hMSCs were administered to infected mice in both prophylactic

(2 days prior to infection) and therapeutic (2 and 5 days P.I.) experimental groups. Weight loss

over the course of infection was recorded (Figure 3.6). There was no difference in weight loss

between experimental groups.

0

1

2

3

4

5

Day 7 P.I.

Total Protein

BA

L m

g/m

l +

SD

0

100

200

300

400

500

600

700ALBUMIN

Day 7 P.I.

ND

BA

L u

g/m

l +

SD

0

1

2

3

4

5

6 IgM

Day 7 P.I.

NDBA

L u

g/m

l +

SD

hMSC Day 5 P.I.hMSC Day 2 P.I.PBSUninfected

Page 82: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

71

Figure 3.6. hMSCs fail to alter weight loss kinetics in experimental sub-lethal influenza. Eight week-old male C57Bl/6 mice were administered 2.5 x 105 hMSCs (passage 3), via the tail vein, 2 days prior to a sub-lethal influenza A/PR/8 virus infection (150 EID50), 2 days post infection (P.I.), or 5 days P.I. Weight was recorded daily. No significant difference in weight loss over the course of infection was observed (Two-way ANOVA, n=11/group). Error bars represent standard deviation.

Page 83: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

72

3.3.3 Mesenchymal stem cell prophylaxis, with or without MSC cytokine pre-incubation, fails to improve survival in experimental influenza

Several important findings in MSC biology have centered around the micro-immuno

environment for which hMSCs persist. One key observation is the requirement of IFN-γ in

combination with TNF, IL-1α or IL-1β, to induce human MSC-mediated immunosuppression

[238]. Therefore, we tested whether pre-incubating hMSCs with both IFN-γ and TNF [as

described in 220], could enhance the immunosuppressive properties of hMSCs and therefore

improve survival when administered to infected mice in our experimental model of severe

influenza. 2.5x105 cytokine stimulated or non-stimulated hMSCs (passage 3) were administered

to influenza virus-infected mice as prophylaxis (2 days prior to infection) or therapy (5 days

P.I.). Weight loss kinetics (Figure 3.7A) and consequent survival kinetics (Figure 3.7B) were

unchanged with administration of cytokine stimulated or non-stimulated hMSCs 2 days prior to

infection, compared to infected control mice administered PBS. Similarly, no improvement in

weight loss or survival was observed with cytokine stimulated hMSCs administered on day 5

P.I., compared to infected control mice administered PBS (data not shown).

Page 84: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

73

Figure 3.7. Cytokine stimulated hMSC prophylaxis fails to alter weight loss kinetics or improve survival in experimental severe influenza. Eight week-old male C57Bl/6 mice were administered 2.5 x 105 hMSCs, pre-incubated for 24 hours with or without 10 ng/ml IFN-γ and 3 ng/ml TNF, 2 days prior to infection with 425 EID50 influenza A/PR/8 virus. Weight was recorded daily. No significant difference in (A) weight loss kinetics (Two-way ANOVA, n=10/group) or (B) survival (logrank test, n=10/group) were observed. Error bars represent standard deviation.

3.3.4 Mesenchymal stem cell therapy fails to reduce mortality and/or dampen host inflammation and lung injury when used as an adjunctive therapy in two models of experimental influenza

Influenza virus-infected individuals seeking clinical care will likely be treated with antiviral

drugs upon presentation. Therefore, we tested the hypothesis that the combination of a

neuraminidase inhibitor, oseltamivir, together with hMSCs, could be effective in reducing

mortality. To reflect a common clinical scenario, we delayed combination therapy for 5 days

after challenging C57Bl/6 with 550 EID50 influenza A/PR/8 virus. Mice receiving oseltamivir

and control mice receiving PBS had a survival rate of 18% and 17%, respectively. With the

addition of hMSC therapy to oseltamivir treatment, the survival rate increased to 35% (Figure

0 2 4 6 8 10 12 140

20

40

60

80

100

Day P.I.

Per

cen

t su

rviv

al

0 1 2 3 4 5 6 7 870

80

90

100

110

Day P.I.

% o

f ori

gina

l wei

ght

(mea

n±± ±±

SD

)

hMSC Day -2 P.I.cytokine stimulated hMSC Day -2 P.I.PBSA B

hMSC Day -2 P.I.cytokine stimulated hMSC Day -2 P.I.PBS

Page 85: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

74

3.8A right). However, this result was not statistically significant (logrank test; p=0.47). Weight

loss over the course of infection was similar between experimental groups (Figure 3.8A left).

It is important to employ a variety of influenza virus strains to ascertain that the experimental

effects can be applicable to human infection by various strains. Therefore, a similar experiment

to assess hMSCs as an adjunctive treatment strategy was performed with a lethal dose (104

EID50) of influenza A/Mexico/4108/09 (A/Mex/4108) virus, a 2009 pandemic H1N1 human

influenza strain that can directly infect mice without undergoing serial passage (Figure 3.8B).

As the onset of weight loss over the course of infection occurs more rapidly in our influenza

A/Mex/4108 virus model compared to our influenza A/PR/8 virus model, oseltamivir and hMSC

treatment was initiated on day 2 P.I. No difference in weight loss (Figure 3.8B left) or survival

(Figure 3.8B right) between experimental groups was observed.

Page 86: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

75

Figure 3.8. hMSC adjunctive therapy may improve survival in experimental influenza A/PR/8 virus infection but not influenza A/Mex/4108 virus infection and hMSC adjunctive therapy does not alter weight loss kinetics in both models of experimental severe influenza. (A) Eight- week old male C57Bl/6 mice infected with 550EID50 influenza A/PR/8 virus were administered 5 mg/kg of oseltamivir in 100 ul ddH2O via gavage, twice daily for 5 days, either with or without hMSC treatment (2.5x105 cells), starting 5 days P.I. Left Weight loss kinetics over the course of infection were measured to assess morbidity in mice. Shown are the weight loss data up to day 8 P.I., when most mice reach euthanasia criteria. No difference between experimental groups was observed (Two-way ANOVA, n=29-32/group; 3 pooled experiments). Error bars represent standard deviation. Right Survival curves. Survival of mice treated with hMSCs and oseltamivir in combination was increased compared to control group; however, this was not significant. (logrank test p=0.47, n=29-32/group; 3 pooled experiments). (B) Eight- week old male C57Bl/6 mice infected with 104 EID50 influenza A/Mex/4108 virus were administered 5 mg/kg of oseltamivir in 100 ul ddH2O via gavage, twice daily for 5 days, either with or without hMSC treatment (2.5x105 cells), starting 2 days P.I. Left Weight loss kinetics over the course of infection were measured to assess morbidity in mice. Shown are the weight loss data up until day 7 P.I., when most mice reach euthanasia criteria. No difference between experimental groups was observed (Two-way ANOVA, n=9-12/group). Error bars represent standard deviation. Right Survival curve. No difference was observed (logrank test, n=9-12/group).

0 1 2 3 4 5 6 7 870

80

90

100

Day P.I. 550 EID50 A/PR/8

% o

f o

rig

inal

wei

gh

t (m

ean

±± ±± S

D)

0 2 4 6 8 10 120

20

40

60

80

100

Day P.I. 550 EID50 A/PR/8

Per

cent

su

rviv

al

0 1 2 3 4 5 6 770

80

90

100

Day P.I. 104 EID50 A/Mex/4108

% o

f o

rig

inal

wei

gh

t (m

ean

±± ±± S

D)

0 2 4 6 80

20

40

60

80

100

Day P.I. 104 EID50 A/Mex/4108

Per

cen

t su

rviv

al

A

B

OseltamivirOseltamivir + hMSC

PBSOseltamivirOseltamivir + hMSC

PBS

OseltamivirOseltamivir + hMSC

PBS OseltamivirOseltamivir + hMSC

PBS

Page 87: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

76

In the A/PR/8 experimental model, mice from each experimental group as described above were

sacrificed on day 7 P.I. BAL fluid and lung homogenate levels of cytokines and chemokines

CCL2, CXCL10, KC, CCL5, and IFN-γ were quantified. BAL fluid and lung homogenate

cytokine and chemokine levels were similar for combination therapy treated mice compared to

control mice that received oseltamivir alone or PBS (Figure 3.9). CXCL10 was significantly

decreased in the lung parenchyma of mice that received oseltamivir, in combination with or

without hMSCs (One-way ANOVA, p<0.05). No difference in BAL fluid markers of acute lung

injury (total protein, IgM, and albumin) was observed on day 7 P.I. for hMSC treated mice

compared to control mice that received oseltamivir or PBS alone (Figure 3.10).

In our influenza A/Mex/4108 virus experimental model, mice from each experimental group as

described above were sacrificed on day 5 P.I. BAL fluid level of cytokines and chemokines

CCL2, CXCL10, KC, CCL5, and IFN-γ were quantified and no difference was observed on day

5 P.I. (Figure 3.11). No difference in BAL fluid markers of acute lung injury (total protein, IgM,

and albumin) was observed on day 5 P.I. for hMSC treated mice compared to control mice that

received oseltamivir or PBS alone (Figure 3.12).

Page 88: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

77

Figure 3.9. hMSCs do not decrease BAL fluid or lung parenchymal inflammation when used as adjunctive therapy in mice infected with influenza A/PR/8 virus. Eight week-old male C57Bl/6 mice infected with 550 EID50 influenza A/PR/8 virus were sacrificed on day 7 P.I. Mice treated with antivirals were administered 5 mg/kg oseltamivir in 100 ul ddH20 once every 12 hours via gavage on day 5 P.I. for a maximum of 5 days. For the combined treatment group, 2.5x105 hMSCs were also administered via the tail vein, 5 days P.I. BAL was performed and lungs were excised. No significant difference in BAL fluid or lung parenchymal level of selected cytokines / chemokines were observed between hMSC treated mice in combination with oseltamivir treatment, compared to oseltamivir alone (one-way ANOVA, n=7/group). Error bars represent standard deviation. ND = nondetectable.

0

500

1000

1500

2000

2500

0

2

4

6IFNγγγγ

BAL LUNG

ND ND

Day 7 P.I.

BA

L p

g/m

l +

S

D

Lu

ng

ng

/ml +

SD

0

1

2

3

4

5

0

5

10

15

20

BAL LUNG

CCL2

ND

Day 7 P.I.B

AL ng/m

l + S

D

Lung n

g/m

l + S

D

0

300

600

900

1200

0

1

2

3

4p<0.05CXCL10

BAL LUNG

ND

Day 7 P.I.

BA

L p

g/m

l +

S

D

Lu

ng

ng

/ml +

SD

0

100

200

300

400

0

10

20

30

40

50

BAL LUNG

CCL5

ND

Day 7 P.I.

BA

L p

g/m

l +

S

D

Lu

ng

ng

/ml +

SD

PBSOseltamivirOseltamivir + hMSC

Uninfected

0

25

50

75

100

125

150

175

0

50

100

150

200

250

300

350KC

BAL LUNG

BA

L p

g/m

l +

SD

Lu

ng pg

/ml +

SD

Day 7 P.I.

Page 89: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

78

Figure 3.10. hMSCs fail to decrease acute lung injury when used as adjunctive therapy in mice infected with influenza A/PR/8 virus. Eight week-old male C57Bl/6 mice infected with 550 EID50 influenza A/PR/8 virus were sacrificed on day 7 P.I. Mice treated with antivirals were administered 5 mg/kg oseltamivir in 100 ul ddH20 once every 12 hours via gavage on day 5 P.I. for a maximum of 5 days. For the combined treatment group, 2.5x105 hMSCs were also administered via the tail vein, 5 days P.I. BAL was performed. No significant difference in BAL fluid total protein, IgM or albumin was observed between hMSC treated mice in combination with oseltamivir treatment, compared to oseltamivir alone (one-way ANOVA, n=7/group). Error bars represent standard deviation. ND = nondetectable.

0

1

2

3

4TOTAL PROTEIN

Day 7 P.I.

BA

L m

g/m

l +

SD

0

300

600

900

1200ALBUMIN

Day 7 P.I.

NDBA

L u

g/m

l +

S

D

0

1

2

3

4

5

6IgM

Day 7 P.I.

NDBA

L u

g/m

l +

S

D

PBSOseltamivirOseltamivir + hMSC

Uninfected

Page 90: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

79

Figure 3.11. hMSCs do not decrease BAL fluid or lung parenchymal inflammation when used as adjunctive therapy in mice infected with influenza A/Mex/4108 virus. Eight week-old male C57Bl/6 mice infected with 104 EID50 A/Mex/4108 virus were sacrificed on day 5 P.I. Mice treated with antivirals were administered 5 mg/kg oseltamivir in 100 ul ddH20 once every 12 hours via gavage on day 2 P.I. for a maximum of 4 days. For the combined treatment group, 2.5x105 hMSCs were also administered, via the tail vein, 2 days P.I. BAL was performed. No significant difference in BAL fluid or lung parenchymal level of selected cytokines / chemokines was observed between hMSC treated mice in combination with oseltamivir treatment, compared to oseltamivir alone (one-way ANOVA, n=5/group). Error bars represent standard deviation. ND = nondetectable.

0

500

1000

1500

2000

2500 IFNγγγγ

Day 5 P.I.

ND

BA

L p

g/m

l + S

D

0

50

100

150

200

250 KC

ND

Day 5 P.I.

BA

L p

g/m

l + S

D

0

200

400

600

800 CCL2

ND

Day 5 P.I.

BA

L p

g/m

l + S

D

0

100

200

300

400 CCL5

ND

Day 5 P.I.

BA

L p

g/m

l + S

D

0

500

1000

1500 CXCL10

ND

Day 5 P.I.

BA

L p

g/m

l + S

D

PBSOseltamivirOseltamivir + hMSC

Uninfected

Page 91: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

80

Figure 3.12. hMSCs fail to decrease acute lung injury when used as adjunctive therapy in mice infected with influenza A/Mex/4108 virus. Eight week-old male C57Bl/6 mice infected with 104 EID50 influenza A/Mex/4108 virus were sacrificed on day 5 P.I. Mice treated with antivirals were administered 5 mg/kg oseltamivir in 100 ul ddH20 once every 12 hours via gavage on day 2 P.I. for a maximum of 4 days. For the combined treatment group, 2.5x105 hMSCs were also administered, via the tail vein, 2 days P.I. BAL was performed. No significant difference in BAL fluid total protein, IgM or albumin was observed between hMSC treated mice in combination with oseltamivir treatment, compared to oseltamivir alone (one-way ANOVA, n=5/group). Error bars represent standard deviation. ND = nondetectable.

0

1000

2000

3000

4000 ALBUMIN

ND

BA

L u

g/m

l + S

D

Day 5 P.I.0

250

500

750

1000 IgM

ND

BA

L n

g/m

l +

SD

D ay 5 P .I .0.0

0.5

1.0

1.5

2.0 TOTAL PROTEIN

BA

L m

g/m

l +

SD

Day 5 P.I.

PBSOseltamivirOseltamivir + hMSC

Uninfected

Page 92: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

81

3.4 DISCUSSION

MSC-based prophylactic and treatment strategies have yielded significant therapeutic benefits in

various pre-clinical models of acute lung injury [209,210,245-248]. These encouraging findings

in which MSCs were efficacious led us to hypothesize that MSCs might dampen host

inflammation, decrease acute lung injury, and improve survival in an experimental model of

severe influenza, characterized by heightened inflammation and acute lung injury. Despite these

positive pre-clinical findings, our study demonstrated that hMSCs were unable to affect

inflammation, lung injury and survival in experimental influenza. This finding is in accordance

with few published reports where MSCs were unable to dampen host inflammation and improve

outcome in vivo [249-251].

Clinical management of severe influenza complicated by ARDS is often guided by the

ALI/ARDS literature. For example, it is recommended that lung-protective ventilations

strategies such as low tidal volume ventilation be used in managing patients with severe

influenza complicated by ARDS, based on recommendations of the ARDSNet trial [278]. Given

the demonstrated utility of drawing from ALI/ARDS literature to inform potential treatment

strategies for influenza, it was reasonable to evaluate the therapeutic potential of MSCs in the

context of severe influenza. Despite similarities in clinical presentation and pathophysiology of

ALI and severe influenza, our negative findings reinforce that viral-induced ALI is not wholly

comparable with other forms and etiologies of ALI, such as bacterial derived ALI.

We first established the influenza A/PR/8 virus mouse model of severe influenza, characterized

by the development of inflammation and acute lung injury, and evaluated the use of hMSCs for

the prevention or treatment of severe disease (Fig. 3.1,3.2). While influenza A/PR/8 virus

Page 93: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

82

infection induced a host response and lung injury in vivo, we do not know whether the host

response was specifically dysregulated, as we did not compare host responses between various

influenza virus infection models. However, in our model, approximately 80% of mice reached

euthanasia criteria - defined by 20% weight loss - where such weight loss was associated with

elevated inflammation.

The failure of hMSCs to suppress inflammation and decrease lung injury could be attributed to

myriad factors. Firstly, the hMSCs may have rapidly differentiated in culture which rendered

them ineffective as immunosuppresive agents. In this study, hMSCs were cultured per the

“manufacturer’s guidelines”, in terms of low passage number, medium and FBS stocks and

concentrations, and cell harvesting at log phase of growth, as these conditions have been deemed

optimal [279]. Despite these attempts to create optimal conditions, it is possible that the cultures

spontaneously differentiated in vitro rendering them ineffective in vivo. Also, the structure and

function of hMSCs tend to vary between donors. For example, Francois et al. found that the

amount of IDO produced by each hMSC donor varied, thereby influencing their

immunosuppressive potential [220]. Nevertheless, culture-expanded MSCs tested in our

experimental model always exhibited a spindle-shaped fibroblastic morphology and were

deemed to be of high quality as indicated by measuring the number of colony forming units (a

generally accepted indicator of MSC quality [279]) (data not shown).

Our results may also be explained if the hMSCs were unable to home to the lungs in our

experimental model. Varied protocols to isolate and expand MSC populations have been

reported to influence MSC homing capacity. For example, protocols which employ progressive

subculturing are associated with a loss of chemokine receptors and decreased chemotactic

Page 94: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

83

responses [232,235,236]. MSCs delivered intravenously would reach the pulmonary

microcirculation first; therefore, homing might not be of concern when lungs are the end target

organ. For example, MSCs may be trapped in the lung as microemboli after IV infusion [258].

Mei et al. also found an average 47% of injected MSCs in the lungs 15 minutes after MSC

delivery into LPS-challenged mice [210]. However, we did not test for the presence of MSCs in

the lung after IV delivery in our model, therefore, we can not definitively conclude that the cells

did in fact reach the lungs.

If the cells were indeed delivered to the lungs when administered intravenously in our model of

influenza, must they have remained in the lungs in order to elicit a beneficial effect? Since we

employed allogeneic MSCs, HLA mismatching may have led to rapid rejection after injection.

This result would be consistent with other reports where allogeneic murine MSCs were rejected

when administered in MHC mismatched mice compared to autologous cell transplant [227,228].

Additionally our cells were derived from human donors, rather than extracted from mice, which

could have exacerbated MSC rejection in vivo. This strategy was employed because human

clinical trials involving MSCs employ MSCs derived from human donors and a similar cross-

species strategy was employed by others in an attempt to elucidate the mechanism of human

MSC mediated effects [258,259]. They found that hMSCs attenuated lung inflammation and

injury (similar to mouse MSCs) which suggests that the beneficial effects are not restricted to

mouse MSCs in mice. Furthermore, MHC class II antigens are not expressed on human MSCs

[223] but are constitutively expressed on murine MSCs [227]. Therefore, human MSC rejection

may be less likely than murine MSC rejection. Indeed, patients who underwent MSC

allotransplant experienced no anti-allogeneic MSC antibody formation or T-cell sensitization

Page 95: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

84

[224], suggesting human MSCs may not be rejected by the recipient and are indeed

immunoprivileged.

Even if the cells were not rejected, their in vivo life span may nonetheless have been too short to

mediate outcome. The majority of MSCs were lost from the lung at 3 and 14 days, respectively,

after delivery to mice with LPS induced lung injury [209-211]. These findings could suggest that

in our experimental model of severe influenza, MSCs did not persist long enough to induce a

beneficial response. However, these studies found MSCs to improve outcome in experimental

lung injury despite low levels of MSC retention in the lungs. Therefore, the reported benefit in

response to cell-based therapy in lung injury models does not necessarily require a high level of

long-term MSC persistence in the lung. Furthermore, MSCs may need not reach the lungs to

exert their effects. For example, intraperitoneal administration of hMSCs attenuated LPS-

induced lung inflammation, despite being less effective than IV delivery or oropharyngeal

aspiration [259].

Alternatively, our ascribed cell dose could have been too low to exert an immunosuppressive

effect, regardless of where the cells localize and/or persist in vivo. We consistently administered

a 2.5x105 dose of hMSCs, via the tail vein, in our experimental influenza model as this dose was

effective in a report by Mei et al. [210]. However, in that report cells were administered via the

jugular vein and not the tail vein, which could potentially account for the observed difference;

cells introduced via the jugular vein have less distance to travel and therefore less potential to be

destroyed before reaching the lungs, compared to cells administered via the tail vein. Indeed,

Kim et al. found that a dose of 1x106 cells resulted in a significantly prolonged life span and a

significantly higher number of motor neurons in mice with amyotrophic lateral sclerosis

compared to untreated mice or mice treated with 1x104 cells [280]. Additionally, several positive

Page 96: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

85

findings regarding the use of MSC therapy in lung injury models reported administration of 5 x

105 cells and 7.5 x 105 cells [245,247]. Therefore, it is possible that, as opposed to having too few

cells persist in the lung, the problem was created at the outset with an insufficient dosage of

MSCs delivered via the tail vein. This issue could be circumvented by injecting more cells.

However, rats administered 5 million cells at one time point or 2 million cells at two time points

in a transplantation model did not suppress immunity but rather accelerated graft rejection [251].

As alluded to earlier, the route of MSC delivery may have affected our findings. MSCs

delivered directly into the lung airspaces have been shown to decrease cytokines in BAL fluid

and improve survival after endotoxin induced lung injury [247]. However, MSCs delivered

intravenously in a similar model did not reduce BAL inflammatory cytokines [246]. Therefore,

it is possible that direct rather than IV MSC delivery to the lungs of influenza virus-infected mice

might have improved MSC mediated immunosupression and outcome in our experimental

model. However, MSCs administered by various methods in addition to intrapulmonary delivery

- including tail vein, jugular vein, and intraperitoneal delivery - were effective in improving

outcome in experimental lung injury [209,210,245,246,259]. Therefore, while route of delivery

may be important, it is likely insufficient to explain our negative results.

Given the vast amounts of literature describing the immunosuppressive functions of MSCs, it is

reasonable to question whether MSC immunosuppression could diminish the ability of the host

to control virus replication and mediate viral clearance. However, we did not observe any

increase in viral load with hMSC therapy (Fig. 3.3C). To the contrary, recent in vivo studies

have provided evidence that MSCs may enhance antimicrobial immune effector cell function and

reduce microbial burden during infectious challenge [221,244]. For example, Mei et al. reported

Page 97: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

86

decreased total bacterial counts in septic mice, which was hypothesized to be attributed to

increased splenic monocyte and macrophage phagocytosis [244]. In our model, influenza viral

titer might have been too overwhelming for any anti-microbial effects to have occurred, or the

hMSCs may have been unable to exert anti-viral compared to anti-bacterial effects.

Alternatively, it is possible that the influenza virus in the lung itself interacted with the MSCs in

a manner which prevented the MSCs from exerting their immunosuppressive activity.

Temporary inactivation of MSC mediated immunosuppression induced by viral activation of

toll-like receptors (TLRs) 3 and 4 on MSCs has been described [281]. This finding may

represent a mechanism that allows the immune system to function effectively in the presence of

virus or bacteria. This finding could account for reduced MSC functionality in the course of

infection since influenza RNA can serve as a ligand for TLR3.

As the induction of MSC immunosuppression highly depends on the microenvironment into

which the cells are introduced, when is the ideal time to administer MSCs in severe influenza?

hMSCs were delivered early (day 2 P.I.) or late (day 5 P.I.) in the course of infection when lung

inflammation was minimal or elevated, respectively. In both regimens, MSCs failed to dampen

host lung inflammation, decrease acute lung injury, or improve morbidity and mortality (Fig.

3.3-3.5). Despite many in vitro findings of MSC mediated immunosuppression, the

microenvironment in vivo is much more variable and unpredictable. When applying MSCs to an

animal model of disease, MSC immunosuppression might be easily turned off in vivo, or rather

immunostimulatory, as has been reported in other models of disease [250]. This may account for

the inability of MSCs to dampen host inflammation in our animal model of severe influenza.

Page 98: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

87

Unfortunately, there is no validated method of measuring MSC bioactivity in vivo, which

generates further challenges [282].

Another discrepancy which may account for observed differences between the effects of MSCs

in our model of lung injury and those with reported MSC therapeutic benefit is the length of time

between introducing the injury-causing agent, and the inflammatory response / onset of disease.

Most models of acute lung injury result in acute lung inflammation which peaks within 24-48

hours then resolves shortly thereafter, such as in the endotoxin lung injury model. In our model

of severe influenza virus-induced lung injury, the host response did not ensue until

approximately 4 days post-influenza virus infection - and mice could remain alive for up to 9

days P.I. before they succumbed to death. This may have important implications for our

findings. MSCs introduced to the lung on day 5 post-influenza virus infection may not have

exerted effects which lasted long enough to affect the outcome of influenza virus-infected mice.

Additionally, MSCs administered prophylactically or early in the course of infection may not

have been sufficiently stimulated by the host microenvironment, as the host inflammatory

response was only detected beginning around 4 days post-influenza virus infection (Fig. 3.2).

Contrary to our hypothesis, we found that MSCs administered prophylactically increased

morbidity of influenza virus-infected mice; however, this finding was not significant (Fig. 3.7).

An explanation for these effects could be that insufficient in vivo levels of pro-inflammatory

cytokines rendered MSCs immune-enhancing, despite MSC ex-vivo pre-stimulation for one

group of MSC treated mice in an attempt to counter this effect. Nevertheless, MSCs administered

concurrently with lung injury or before lung injury in experimental models have been effective

[209,245,246]. In fact, Ortiz et al. reported that MSCs were only effective when administered at

Page 99: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

88

the same time as bleomycin [245]. Therefore, MSCs can exert their beneficial effects in the

initial stages of injury, albeit in a model of bleomycin induced injury and not viral induced

injury. Although timing of MSC administration is likely to be an important factor, it is difficult

to conclude that the non- or minimally inflammatory environment into which the MSCs were

introduced when administered prophylactically or early in the course of influenza virus infection

is the reason for which the cells lacked efficacy in improving morbidity and mortality.

MSCs administered in combination with oseltamivir also had no effect (Fig. 3.8 – 3.12). Since

the mice treated with oseltamivir only did not show improved survival in our influenza A/PR/8

virus or influenza A/Mex/4108 virus experimental models, we postulated that the infection may

simply have been too severe for any agent to improve outcome. In particular, the absence of

clinical effect could have been because the high levels of inflammation present in severe

influenza may have overcome the immunosuppressive properties of MSCs in vivo. This is

consistent with the absence of clinical effect in experimental GvHD and rheumatoid arthritis,

both diseases characterized by overwhelming inflammation [249,250]. However, this may not

necessarily be the case in our study since MSCs have proven effective in pre-clinical sepsis, a

disease characterized by an extreme systemic inflammatory response [221].

While antiviral treatment was initiated late in the course of infection to re-capitulate the clinical

scenario, the timing of administration was likely too late to inhibit viral replication, since viral

load has been shown to peak in the lungs 24-72 hours after infection [88]. Oseltamivir is also

known to have limited efficacy when administered beyond 48 hours post-onset of symptoms –

mice begin to lose weight around day 5 P.I. which could be deemed the onset of influenza

Page 100: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

89

symptoms, however, it is difficult to correlate symptom onset in humans with our experimental

animal model.

Despite a lack of efficacy with oseltamivir treatment, we observed decreased levels of IFN-γ,

CCL2, CCL5 and KC in BAL fluid and significantly decreased CXCL10 in lung interstitium of

oseltamivir-treated mice, independent of MSC treatment (Fig. 3.9). Wong et al. noted

oseltamivir could dampen host inflammation independent of an antiviral effect, which may

explain our findings [283]. Similarly, others have also hypothesized that a dampened immune

response, including decreased cytokine production, may be the mechanism of action of

oseltamivir, rather than influenza virus replication inhibition [284]. However, it is difficult to

determine whether our finding was attributable to oseltamivir immunomodulatory effects or to an

indirect effect of oseltamivir on viral replication, as lung viral load was not quantified in this set

of experiments.

Noteworthy, decreased lung inflammation (albeit insignificant) with oseltamivir treatment did

not correlate with improved survival in our experimental model. This may indicate that our

experimental model was too severe, as not even a modest ability to dampen host inflammation

was able to translate into decreased morbidity and mortality. Related, neither oseltamivir or

oseltamivir/MSC combination treatment administered starting day 2 post infection in the 2009

pandemic H1N1 experimental influenza (A/Mex/4108) virus model had any effect. Thus,

although influenza A/Mex/4108 virus was a human strain which is not murine-adapted (thereby

having possibly greater clinical relevance to human influenza virus infection than influenza

A/PR/8 virus) our dose of 104 EID50 influenza A/Mex/4108 virus was probably too

Page 101: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

90

overwhelming to observe any therapeutic effects of either oseltamivir or MSCs as the influenza

virus dose proved to be 100% lethal regardless of the treatments applied.

Finally, although MSC-secreted proteins have been attributed to MSC-mediated improved

outcome in pre-clinical models of lung injury, they may not have been useful in the treatment of

severe influenza, despite underlying pathophysiology common to lung injury derived from viral

or bacterial derived pneumonia, sepsis, or trauma. For example, althought it may be beneficial to

administer recombinant TSG-6 as treatment for LPS-induced lung injury in mice [259], TSG-6

treatment for severe influenza may be detrimental due to its ability to upregulate COX-2 [285],

as COX-2 upregulation may contribute to increased morbidity and mortality in severe influenza

(discussed in chapter 1). The same reasoning is applicable to MSC upregulation of PGE2, which

is generated by conversion of arachidonic acid via COX-2. Furthermore, IFN-γ and TNF-

induced upregulation of MSC secreted factors may have exacerbated disease severity; this may

explain why MSCs pre-incubated with these cytokines resulted in increased morbidity and

mortality (Fig. 3.7). Further evaluation of individual MSC-secreted proteins for treating severe

influenza may be warranted.

In conclusion, our study was not able to demonstrate the therapeutic potential of MSCs in

experimental severe influenza in mice. Given the overwhelming number of publications

identifying MSCs as an effective treatment in a variety of disease models, we believe that our

findings are valuable to the field of MSC research as they provide a noteworthy contrast to some

of the existing literature in this field. Ongoing clinical trials should reveal the true potential of

these cells and provide insight as to whether further investigation of MSCs for severe influenza

treatment is warranted.

Page 102: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

91

Chapter 4: Summary and Future Directions

4.1 Summary

Antiviral drugs currently approved for the treatment of influenza have inherent therapeutic

limitations, including unknown efficacy in treating severe disease or complications, timing of

administration (i.e. limited clinical impact when administered beyond the first 48 hours of

symptom onset) and rising antiviral resistance. Therefore, we investigated the use of iNO as a

new antiviral treatment for severe influenza (Chapter 2). Unfortunately, we found that iNO was

unable to dampen the viral load and thus improve outcome in experimental severe influenza.

This may have been due to the instability and short half-life of NO and the toxic effects of NO

metabolites. In future, there may be merit to exploring another vehicle of NO delivery which

could perhaps stabilize nitric oxide without generating toxic effects. Overall, our findings

emphasize the limitations of drawing conclusions from in vitro experimental findings, as the host

response in vivo is much more complex; in vitro findings may not reflect what will be

correspondingly observed in vivo where myriad pathways are activated during infection.

Severe influenza is associated with an overexuberant host-mediated innate immune response,

leading to excessive inflammation, tissue injury and acute lung injury. It is clear that this

deleterious inflammatory host response is a major contributor to influenza-related morbidity and

mortality. Development of novel and innovative adjunctive treatment strategies that target the

host response to the influenza virus may be critically important for clinical outcome in the case

of severe disease. It is clear that severe influenza with acute lung injury is a highly complex

disease process involving many pathophysiologic manifestations. Given this, it is not surprising

Page 103: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

92

that strategies targeting one aspect of the disease process have yet to prove efficacious. This

suggests that a new multifaceted therapeutic approach, aimed at reducing lung injury while

maintaining a balanced immune response and facilitating lung repair, could be of greatest value.

In this regard, mesenchymal stem cells were an obvious choice warranting investigation as a new

treatment approach to lung injury and enhanced inflammation developed in severe influenza due

to their previous demonstrated effects in pre-clinical models of lung injury (Chapter 3).

However, contrary to our hypothesis, we found that MSCs did not reduce cytokine/chemokine

responses or improve ALI in our murine influenza model. Our experimental findings reinforce

that, despite similarities in clinical presentation and pathobiology of acute lung injury caused by

various sources, beneficial effects in other models of ALI/ARDS may not necessarily be

translated to influenza-virus induced ALI.

Page 104: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

93

4.2 Future directions

Our main focus was to test whether MSCs, given alone or as adjunctive therapy, could reduce

host inflammation, specifically, inflammatory cytokines correlated with severe disease and

protein level in the lungs - a hallmark of ALI. We were unable to identify any beneficial effect;

therefore, a number of plausible explanations merit further investigation. For example, were

MSCs in fact mediating the host response? We did not provide any indication for MSC

biological activity in vivo and host interaction. To determine this, we could investigate

biomarkers of MSC activity in vivo. Specifically, BAL fluid levels of important MSC-secreted

proteins, including KGF, Ang-1 and PGE2, could be measured following MSC delivery.

In light of the fact that oseltamivir did not decrease morbidity and mortality in our experimental

model, we could not conclude that MSCs were ineffective as an adjunctive therapeutic agent.

Therefore, studies to optimize oseltamivir dosing were recently completed (data not shown).

Further examination of an MSC and oseltamivir combination treatment strategy - with a dose of

oseltamivir that improves survival by ~30% when used alone - is underway.

Further studies could address the hypothesis that insufficient MSC dosing led to sub-optimal in

vivo effects, as the optimal MSC dose necessary to obtain a therapeutic benefit remains

undefined. A report by Kim et al. found MSC mediated effects in vivo to positively correlate

with increasing cell dose [280]. Therefore, we could determine whether provision of 1 million

MSCs to influenza virus-infected mice improves outcome. We could also try intratracheal or

intrapulmonary delivery of cells to influenza virus-infected mice, as there is no standardized

method of MSC delivery that achieves greatest clinical efficacy (as discussed in Chapter 3).

Page 105: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

94

While it would be favourable to develop a reliable protocol to culture MSCs, it may also be

valuable to assess other in vitro culturing systems. Specifically, MSCs could be grown as 3D

spheroids under the “hanging drop” method as this form of culture has been shown to enhance

MSC mediated immunosuppression, compared to monolayer culture, in a co-culture system with

LPS-activated macrophages and in a mouse model of zymosan induced peritonitis [286]. It may

also be valuable to assess MSC delivery in a more stable form. To protect against MSC

degradation, delivery of hydrogel-encapsulated MSCs as described by Karoubi et al. could

provide enhanced cell survival in the lungs [287]. Alternatively, MSC cultures could potentially

be modified to improve their immunosuppressive properties.

Lastly, if these investigations reveal a role for MSC-mediated improvement in severe disease,

therapeutic strategies based on enhancement of MSC-secreted proteins could be considered. We

have not found any literature that reports a therapeutic benefit with the use of MSC-secreted

mediators in severe influenza. However, single MSC-released mediators have been effective in

improving outcome in pre-clinical models of lung injury. For example, intrapulmonary delivery

of recombinant human TSG-6, a known MSC mediator, reduced LPS-induced inflammation in

the lung [259]. In particular, proteins which could affect the endothelial barrier may be of

interest as inhibition of endothelial activation/dysfunction and associated capillary leak has

recently been shown to increase survival of H5N1 virus-infected mice, without altering cytokine

levels or viral load [288]. Binding of the ligand Slit to its endothelial receptor Robo4 inhibits

hyperpermeability induced by VEGF and promotes cell-surface expression of adherens junction

protein VE cadherin [288,289]. London et al. significantly improved survival in experimental

H5N1 influenza murine models by administering the active fragment of Slit, Slit2N, which is

released via proteolytic cleavage upon binding to Robo4 [288]. This study suggests that

Page 106: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

95

strengthening the vascular-endothelial barrier may be an important therapeutic target for severe

influenza treatment.

With this in mind, we could evaluate the role of MSC-secreted protein Ang-1, a potent mediator

of angiogenesis that functions to prevent leakage and promote blood vasculature quiescence, in

the course of severe influenza virus infection. Should Ang-1 be dysregulated, we could assess

Ang-1 rescue strategies in restoring endothelial integrity in the lungs, such as administration of

Ang-1 via overexpression in a viral vector. Increasing Ang-1 levels to preserve endothelial

integrity may prove to be an effective strategy to prevent or ameliorate ALI caused by severe

influenza.

Page 107: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

96

4.3 Final considerations

Antiviral resistance against influenza, including dual (adamantane and oseltamivir) resistance

[290], is likely to continue to increase, posing further public health concerns. Therapeutic

strategies employing antiviral agents alone are unlikely to significantly improve overall clinical

outcome of individuals with severe influenza. Therefore, interest in adjunctive

immunomodulatory strategies for the treatment of influenza needs to grow, and innovative

approaches targeting the host response to influenza need to emerge.

The current, limited experimental evidence to support the use of immunomodulatory therapy

must be strengthened. Promising approaches to combined antiviral and immunomodulatory

therapy for influenza have been demonstrated by Zheng et al. using celecoxib, mesalazine and

zanamivir in pre-clinical models of severe influenza [162]. Further study of such approaches is

clearly warranted. Based on the promising results in pre-clinical models of severe influenza,

combination therapy employing PPAR agonists and/or COX-2 inhibitors with conventional

antiviral agents should be studied in randomized clinical trials of severe influenza.

Infection and subsequent death due to avian influenza A (H5N1) is likely to continue. In the

event that the H5N1 virus develops the ability to transmit from human-to-human, a pandemic

may occur. Furthermore, as evidenced by 2009 pandemic H1N1 influenza, it is not currently

possible to predict which influenza virus strain will give rise to the next pandemic, and how

virulent that strain will be. New adjunctive treatment strategies, in addition to new antiviral

therapy, will likely be required to significantly improve clinical outcome.

Page 108: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

97

Finally, immunomodulatory strategies that have yielded positive results in pre-clinical and

clinical studies of ALI/ARDS may have therapeutic implications for the management of severe

influenza. Targeted strategies to preserve vascular-endothelial integrity in an effort to prevent

influenza-related end-organ dysfunction/injury, including ALI/ARDS, should emerge as an

innovative approach to improve clinical outcome in severe influenza.

Page 109: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

98

Chapter 5: References

1. Webster RG, Bean WJ, Gorman OT, Chamber TM, Kawaoka Y. Evolution and ecology of influenza A viruses. Microbiol. Rev. 56(1), 152–79 (1992). 2. World Health Organization. Fact sheet 211, Influenza: Available at: http://www.who.int/mediacentre/factsheets /2003/fs211/en/. Accessed November 15, 2010. 3. Thompson WW, Shay DK, Weintraub E et al. Influenza-associated hospitalizations in the United States. JAMA 292, 1333–40 (2004). 4. Taubenberger JK, Morens DM. The pathology of influenza virus infections. Ann. Rev. Pathol. 3, 499–522 (2008). 5. Johnson NP, Mueller J. Updating the accounts: Global mortality of the 1918–1920 “Spanish” influenza pandemic. Bull. Hist. Med. 76(1), 105–15 (2002). 6. World Health Organization. Pandemic influenza preparedness and response: Available at: http://www.who.int/csr/ disease/influenza/PIPGuidance09.pdf. Accessed November 15, 2010. 7. Donaldson LJ, Rutter PD, Ellis BM et al. Mortality from pandemic A/H1N1 2009 influenza in England: public health surveillance study. BMJ. 339, b5213 (2009). 8. World Health Organization. Cumulative number of confirmed human cases for avian influenza A (H5N1) reported to WHO, 2003–2012. Available at: http://www.who.int/influenza/human_animal_interface/EN_GIP_20120412CumulativeNumberH5N1cases.pdf. Accessed April 15, 2012. 9. World Health Organization. WHO case definitions for human infections with influenza A (H5N1) virus. Available at: http://www.who.int/influenza/resources/documents/case_definition2006_08_29/en/. Accessed April 15, 2012. 10. Wang TT, Parides MK, Palese P. Seroevidence for H5N1 influenza infections in humans: meta-analysis. Science 335(6075), 1463 (2012). 11. Baigent SJ, McCauley JW. Influenza type A in humans, mammals and birds: Determinants of virus virulence, host-range and interspecies transmission. Bioessays. 25(7), 657–671 (2003). 12. Influenza virus life cycle © Richard Burke Squires, courtesy of the Biology Image Library. Available at: http://gallus.reactome.org/cgi-bin/eventbrowser?DB=test_gallus_reactome_release_2_myisam&ID=168255&. Accessed April 15, 2012.

Page 110: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

99

13. Zambon MC. The pathogenesis of influenza in humans. Rev. Med. Virol. 11(4), 227–241 (2001). 14. Medina RA, Garcia-Sastre A. Influenza A viruses: new research developments. Nat. Rev. Microbiol. 9, 590–603 (2011). 15. World Health Organization. Global alert and response (GAR): Recommended use of antivirals: Available at: http://www.who.int/csr/disease/swineflu/notes/h1n1_ use_antivirals_20090820/en/index.html. Accessed December 15, 2010. 16. To K, Chan PK, Chan K et al. Pathology of fatal human infection associated with avian influenza A H5N1 virus. J. Med. Virol. 63(3), 242–246 (2001). 17. Tran TH, Nguyen TL, Nguyen TD et al. Avian influenza A (H5N1) in 10 patients in Vietnam. N. Engl. J. Med. 350(12), 1179–1188 (2004). 18. Beigel JH, Farrar J, Han AM et al. Avian Influenza A (H5N1) Infection in humans. N. Engl. J. Med. 353(13), 1374–1385 (2005). 19. Abdel-Ghafar A-N, Chotpitayasunondh T, Gao Z et al. Update on avian influenza A (H5N1) virus infection in humans. N. Engl. J. Med. 358(3), 261–273 (2008). 20. Gill JR, Sheng Z-M, Ely SF et al. Pulmonary pathological findings of fatal 2009 pandemic influenza A/H1N1 viral infections. Arch. Pathol. Lab. Med. 134(2) 235–253 (2010). 21. Ware LB, Matthay MA. The acute respiratory distress syndrome. N. Engl. J. Med. 342, 1334–1349 (2000). 22. Bernard GR. Acute respiratory distress syndrome: A historical perspective. Am. J. Respir. Crit. Care Med. 172(7), 798–806 (2005). 23. Mauad T, Hajjar LA, Callegari GD et al. Lung pathology in fatal novel human influenza A (H1N1) infection. Am. J. Respir. Crit. Care Med. 181, 72–79 (2010). 24.Webster RG, Rott R. Influenza virus A pathogenicity: the pivotal role of hemagglutinin. Cell 50(5), 665–666 (1987). 25. Puthavathana, P, Auewarakul P, Charoenying PC et al. Molecular characterization of the complete genome of human influenza H5N1 virus isolates from Thailand. J. Gen. Virol. 86(Pt 2), 423–433 (2005). 26. Kalthoff D, Globig A, Beer M. (Highly pathogenic) avian influenza as a zoonotic agent. Vet. Microbiol. 140(3–4), 237–245 (2010). 27. Hatta M, Gao P, Halfmann P, Kawaoka Y. Molecular basis for high virulence of Hong Kong H5N1 influenza A viruses. Science 293(5536), 1840–1842 (2001).

Page 111: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

100

28. Cheng X, Xu Q, Song E, Yang C-F, Kemble G, Jin H. The hemagglutinin protein of influenza A/Vietnam/1203/2004 (H5N1) contributes to hyperinduction of proinflammatory cytokines in human epithelial cells. Virology 406(1), 28–36 (2010). 29. Tumpey TM, García-Sastre A, Taubenberger JK et al. Pathogenicity of influenza viruses with genes from the 1918 pandemic virus: functional roles of alveolar macrophages and neutrophils in limiting virus replication and mortality in mice. J. Virol. 79(23), 14933–14944 (2005). 30. Chen CJ, Chen GW, Wang CH, Huang CH, Wang YC, Shih SR. Differential localization and function of PB1-F2 derived from different strains of influenza A virus. J. Virol. 84(19), 10051–62 (2010). 31. Basler CF, Aguilar PV. Progress in identifying virulence determinants of the 1918 H1N1 and the Southeast Asian H5N1 influenza A viruses. Antiviral Res. 79(3), 166–178 (2008). 32. Chen W, Calvo PA, Malide D et al. A novel influenza A virus mitochondrial protein that induces cell death. Nat. Med. 7(12), 1306–1312 (2001). 33. Zamarin D, Ortigoza MB, Palese P. Influenza A virus PB1-F2 protein contributes to viral pathogenesis in mice. J. Virol. 80(16), 7976–7983 (2006). 34. McAuley JL, Hornung F, Boyd KL et al. Expression of the 1918 influenza A virus PB1-F2 enhances the pathogenesis of viral and secondary bacterial pneumonia. Cell Host Microbe 2(4), 240–249 (2007). 35. Garten RJ, Davis T, Russell CA et al. Antigenic and genetic characteristics of swine-origin 2009 A (H1N1) influenza viruses circulating in humans. Science 325 (5939), 197–201 (2009). 36. Hai R, Schmolke M, Varga ZT et al. PB1-F2 expression by the 2009 pandemic H1N1 influenza virus has minimal impact on virulence in animal models. J. Virol. 84(9), 4442–4450 (2010). 37. Ozawa M, Basnet S, Burley LM, Neumann G, Hatta M, Kawaoka Y. Impact of amino acid mutations in PB2, PB1-F2, and NS1 on the replication and pathogenicity of pandemic (H1N1) 2009 influenza viruses. J. Virol. 85(9), 4596–4601 (2011). 38. McAuley JL, Zhang K, McCullers JA. The effects of influenza A virus PB1-F2 protein on polymerase activity are strain specific and do not impact pathogenesis. J. Virol. 84(1), 558–564 (2010). 39. Conenello GM, Zamarin D, Perrone LA et al. A single mutation in the PB1-F2 of H5N1 (HK/97) and 1918 influenza A viruses contributes to increased virulence. PLoS Pathog. 3(10), 1414–1421 (2007).

Page 112: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

101

40. Conenello GM, Tisoncik JR, Rosenzweig E, Varga ZT, Palese P, Katze MG. A single N66S mutation in the PB1-F2 protein of influenza A virus increases virulence by inhibiting the early interferon response in vivo. J. Virol. 85(2), 652–662 (2011). 41. Steel J, Lowen AC, Mubareka S, Palese P. Transmission of influenza virus in a mammalian host is increased by PB2 amino acids 627K or 627E/701N. PLoS Pathog. 5(1), e1000252 (2009). 42. Fornek JL, Gillim-Ross L, Santos C et al. A single-amino-acid substitution in a polymerase protein of an H5N1 influenza virus is associated with systemic infection and impaired T-cell activation in mice. J. Virol. 83(21), 11102–11115 (2009). 43. Zhou B, Li Y, Halpin R, Hine E, Spiro DJ, Wentworth DE. PB2 residue 158 is a pathogenic determinant of pandemic H1N1 and H5 influenza a viruses in mice. J. Virol. 85(1), 357–365 (2011).

44. Soubies SM, Volmer C, Croville G et al. Species-specific contribution of the four C-terminal amino acids of influenza A virus NS1 protein to virulence. J. Virol. 84(13), 6733–6747 (2010). 45. Illyushina NA, Seiler JP, Rehg JE, Webster RG, Govorkova EA. Effect of neuraminidase inhibitor–resistant mutations on pathogenicity of clade 2.2 A/Turkey/15/06 (H5N1) influenza virus in ferrets. PLoS Pathog. 6(5), e1000933 (2010). 46. Steel J, Lowen AC, Pena L et al. Live attenuated influenza viruses containing NS1 truncations as vaccine candidates against H5N1 highly pathogenic avian influenza. J. Virol. 83(4), 1742–1753 (2009). 47. Hai R, Martínez-Sobrido L, Fraser KA, Ayllon J, García-Sastre A, Palese P. Influenza B virus NS1-truncated mutants: live-attenuated vaccine approach. J. Virol. 82(21), 10580–10590 (2008).

48. Zhou B, Li Y, Belser JA, Pearce MB et al. NS-based live attenuated H1N1 pandemic vaccines protect mice and ferrets. Vaccine 28(50), 8015–8025 (2010). 49. Volmer R, Mazel-Sanchez B, Volmer C, Soubies SM, Guérin J-L. Nucleolar localization of influenza A NS1: striking differences between mammalian and avian cells. J. Gen. Virol. 89(10), 2359–2376 (2009). 50. Fernandez-Sesma A. The influenza virus NS1 protein: inhibitor of innate and adaptive immunity. Infect. Disord. Drug Targets 7(4), 336–343 (2007). 51. Chen Z, Krug RM. Selective nuclear export of viral mRNAs in influenza-virus-infected cells. Trends Microbiol. 8(8), 376–383 (2000). 52. Kochs G, García-Sastre A, Martínez-Sobrido L. Multiple anti-interferon actions of the influenza A virus NS1 protein. J. Virol. 81(13), 7011–7021 (2007).

Page 113: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

102

53. Mibayashi M, Martínez-Sobrido L, Loo YM, Cárdenas WB, Gale M Jr, García-Sastre A. Inhibition of retinoic acid-inducible gene I-mediated induction of beta interferon by the NS1 protein of influenza A virus. J. Virol. 81(2), 514–524 (2007). 54. Hayman A, Comely S, Lackenby A et al. NS1 proteins of avian influenza A viruses can act as antagonists of the human alpha/beta interferon response. J. Virol. 81(5), 2318–2327 (2007). 55. Nemeroff ME, Barabino SM, Li Y, Keller W, Krug RM. Influenza virus NS1 protein interacts with the cellular 30 kDa subunit of CPSF and inhibits 3'end formation of cellular pre-mRNAs. Mol. Cell 1(7), 991–1000 (1998). 56. Min JY, Krug RM. The primary function of RNA binding by the influenza A virus NS1 protein in infected cells: Inhibiting the 2'-5' oligo (A) synthetase/RNase L pathway. Proc. Natl. Acad. Sci. U.S.A. 103(18), 7100–7105 (2006). 57. Min JY, Li S, Sen GC, Krug RM. A site on the influenza A virus NS1 protein mediates both inhibition of PKR activation and temporal regulation of viral RNA synthesis. Virology 363(1), 236–243 (2007). 58. Seo SH, Hoffmann E, Webster RG. Lethal H5N1 influenza viruses escape host anti-viral cytokine responses. Nat. Med. 8(9), 950–954 (2002). 59. Obenauer JC, Denson J, Mehta PK et al. Large-scale sequence analysis of avian influenza isolates. Science 311(5767), 1576–1580 (2006). 60. Jackson D, Hossain MJ, Hickman D, Perez DR, Lamb RA. A new influenza virus virulence determinant: the NS1 protein four C-terminal residues modulate pathogenicity. Proc. Natl. Acad. Sci. U.S.A. 105(11), 4381–4386 (2008). 61. Moscona A. Medical management of influenza infection. Annual Rev. Med. 59, 397–413 (2008). 62. Jefferson T, Jones MA, Doshi P et al. Neuraminidase inhibitors for preventing and treating influenza in healthy adults and children. Cochrane Database of Systematic Reviews. Issue 1 Art. No: CD008965 (2012). 63. Doshi P. Neuraminidase inhibitors: the story behind the Cochrane review. BMJ. 339, 1348–1351 (2009). 64. Mazur I, Wurzer WJ, Ehrhardt C et al. Acetylsalicylic acid (ASA) blocks influenza virus propagation via its NF-kappaB-inhibiting activity. Cell. Microbiol. 9(7), 683–1694 (2007). 65. Bouvier NM, Lowen AC, Palese P. Oseltamivir-resistant influenza A viruses are transmitted efficiently among guinea pigs by direct contact but not by aerosol. J. Virol. 82(20), 10052–10058 (2008).

Page 114: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

103

66. Bloom JD. Permissive secondary mutations enable the evolution of influenza oseltamivir resistance. Science 328(5983), 1272–1275 (2010). 67. Deyde V, Xu X, Bright R et al. Surveillance of resistance to adamantanes among influenza A(H3N2) and A(H1N1) viruses isolated worldwide. J. Infect. Dis. 196(2), 249–257 (2007). 68. World Health Organization. Weekly epidemiological record 4 March 2011, 86th year No. 10, 2011, 86, 81–92: Available at: http://www.who.int/wer/2011/wer8610.pdf. Accessed April 12, 2011. 69. Sheu TG, Deyde VM, Okomo-Adhiambo M et al. Surveillance for neuraminidase inhibitor resistance among human influenza A and B viruses circulating worldwide from 2004 to 2008. Antimicrob. Agents Chemother. 52(9), 3284–3292 (2008). 70. Collins PJ, Haire LF, Lin YP et al. Crystal structures of oseltamivir-resistant influenza virus neuraminidase mutants. Nature 453(7199), 1258–61 (2008). 71. Centers for Disease Control and Prevention. 2008–2009 Influenza season week 39 ending October 3, 2009: Available at: http://www.cdc.gov/flu/weekly/weeklyarchives2008–2009/weekly39.htm. Accessed October 10, 2010. 72. World Health Organization. Influenza A (H1N1) virus resistance to oseltamivir: Available at: http://www.who.int/csr/disease/influenza/h1n1_table/en/index.html. Accessed Nov 22, 2010. 73. World Health Organization. Weekly update on oseltamivir resistance to influenza H1N1 (2009) viruses: Available at: http://www.who.int/csr/disease/influenza/weekly_update_oseltamivir_resistant_ 20101108.pdf. Accessed Nov 22, 2010. 74. Le QM, Kiso M, Someya K et al. Avian flu: Isolation of drug-resistant H5N1 virus. Nature 437(7062), 1108 (2005). 75. de Jong MD, Thanh TT, Khanh TH et al. Oseltamivir resistance during treatment of influenza A (H5N1) infection. N. Engl. J. Med. 353(25), 2667–2672 (2005). 76. A study of intravenous oseltamivir [Tamiflu] in infants with influenza: Available at: http://clinicaltrials.gov/ct2/show/NCT01053663. Accessed April 12, 2011. 77. Kohno S, Kida H, Mizuguchi M, Shimada J. Efficacy and safety of intravenous peramivir for treatment of seasonal influenza virus infection. Antimicrob. Agents Chemother. 54(11), 4568–74 (2010). 78. Castillo R, Holland LE, Boltz DA. Peramivir and its use in H1N1 influenza. Drugs Today (Barc). 46(6), 399–408 (2010). 79. Gaur AH, Bagga B, Barman S et al. Intravenous zanamivir for oseltamivir-resistant 2009

Page 115: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

104

H1N1 Influenza. N. Engl. J. Med. 362(1), 88–89 (2010). 80. Smith JR, Ariano RE, Toovey S. The use of antiviral agents for the management of severe influenza. Crit. Care Med. 38(4), e43–e51 (2010). 81. Armitage JM, Williams SJ. Inhaler technique in the elderly. Age and Ageing 17(4), 275–278 (1988). 82. Medeiros R, Rameix-Welti MA, Lorin V et al. Failure of zanamivir therapy for pneumonia in a bone-marrow transplant recipient infected by a zanamivir-sensitive influenza A (H1N1) virus. Antiviral Ther. 12(4), 571–576 (2007). 83. Food and Drug Administration. Safety letter from GlaxoSmithKline regarding Relenza (zanamivir). Silver Spring, MD, October 8, 2009: Available at: http://www.fda. gov/downloads/safety/MedWatch/ SafetyInformation/SafetyAlertsForHumanMedical Products/UCM186224.pdf. Accessed October 12, 2010. 84. U.S. National Institute of Health. Safety study to assess IV zanamivir for treatment of influenza infection in patients who are in hospital (NAI113678). Phase II. (2011): Available at: http://clinicaltrials.gov/ct2/show/NCT01014988. Accessed January 15, 2011. 85. Da Dalt L, Calistri A, Chillemi C et al. Oseltamivir resistant pandemic (H1N1) 2009 treated with nebulized zanamivir (letter). Emerg. Infect. Dis. 16 (11), (2010). Available at: http://www.cdc.gov/EID/content/16/11/1813.htm. Accessed December 3, 2010. 86. Hayden F. Developing new antiviral agents for influenza treatment: What does the future hold? Clin. Infect. Dis. 48(S1), S3–S13 (2009). 87. Yamashita M. Laninamivir and its prodrug, CS-8958: long-acting neuraminidase inhibitors for the treatment of influenza. Antivir. Chem. Chemother. 21(2),71–84 (2010). 88. Murphy BR, Baron S, Chalhub EG, Uhlendorf CP, Chanock RM. Temperature-sensitive mutants of influenza virus. IV. Induction of interferon in the nasopharynx by wild-type and a temperature- sensitive recombinant virus. J. Infect. Dis. 128(4), 488–493 (1973). 89. Aoki FY, Macleod MD, Paggiaro P et al. Early administration of oral oseltamivir increases the benefits of influenza treatment. J. Antimicrob. Chemother. 51(1), 123–129 (2003). 90. Kandun IN, Tresnaningsih E, Purba WH et al. Factors associated with case fatality of human H5N1 virus infections in Indonesia: A case series. Lancet 372, 744–749 (2008). 91. Jain S, Kamimoto L, Bramley AM et al. Hospitalized patients with 2009 H1N1 influenza in the United States, April–June 2009. N. Engl. J. Med. 361(20), 1935–1944 (2009). 92. To K , Hung I , Li I et al. Delayed clearance of viral load and marked cytokine activation in severe cases of pandemic H1N1 2009 influenza virus infection. Clin. Infect. Dis. 50(6), 850–859

Page 116: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

105

(2010). 93. Enserink M. Old drugs losing effectiveness against flu; could statins fill gap? Science 309(5743), 1976–1977 (2005). 94. Fedson DS. Meeting the challenge of influenza pandemic preparedness in developing countries. Emerg. Infect. Dis. 15(3), 365–371 (2009). 95. Moscona A. Global transmission of oseltamivir-resistant influenza. N. Engl. J. Med. 36(10), 953–956 (2009). 96. Bogdan C. Nitric oxide and the immune response. Nat. Immunol. 2, 907–16 (2001). 97. Fang FC. Antimicrobial reactive oxygen and nitrogen species: concepts and controversies (Review). Nat. Rev. Microbiol. 2, 820–32 (2004). 98. Fang FC, De Groote MA. NO inhibitions: antimicrobial properties of nitric oxide. Clin. Infect. Dis. 21, S162–5 (1995). 99. Akerstrom S, Mousavi-Jazi M, Klingstrom J, Leijon M, Lundkvist A, Mirazimi A. Nitric oxide inhibits the replication cycle of severe acute respiratory syndrome coronavirus. J. Virol. 79, 1966-69 (2005). 100. Rimmelzwaan G, Baars M, Lijster P, Fouchier R, Osterhaus A. Inhibition of influenza virus replication by nitric oxide. J. Virol. 73, 8880–3 (1999). 101. Karupiah G, Xie QW, Buller RML et al. Inhibition of viral replication by IFNg induced NO synthase. Science. 261, 1445–8 (1993). 102. Croen KD. Evidence for antiviral effect of nitric oxide. Inhibition of herpes simplex virus type 1 replication. J. Clin. Invest. 91, 2446–52 (1993). 103. Lin YL, Huang YL, Ma SH et al. Inhibition of Japanese encephalitis infection by nitric oxide: antiviral effect of nitric oxide on RNA virus replication. J. Virol. 71(7), 5227–5235 (1997). 104. Napolitano LM, Park PK, Raghavendran K, Bartlett RH. Nonventilatory strategies for patients with life-threatening 2009 H1N1 influenza and severe respiratory failure. Crit. Care Med. 38, e74–90 (2010). 105. Kumar A, Zarychanski R, Pinto R. et al. Critically ill patients with 2009 influenza A (H1N1) infection in Canada. JAMA. 302, 1872–79 (2009). 106. Food and Drug Administration. Approval of NDA 20-846 INOmax nitric oxide gas. USA: FDA. 1999.

Page 117: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

106

107. DiBlasi RM, Myers TR, Hess DR. Evidence-based clinical practice guideline: inhaled nitric oxide for neonates with acute hypoxic respiratory failure. Respir. Care. 55, 1717–45 (2010). 108. Clark RH, Kueser TJ, Walker MW et al. Low-dose nitric oxide therapy for persistent pulmonary hypertension of the newborn. Clinical inhaled nitric oxide research group. N. Engl. J. Med. 342, 469–74 (2000). 109. The Franco-Belgium Collaborative NO Trial Group. Early compared with delayed inhaled nitric oxide in moderately hypoxaemic neonates with respiratory failure: a randomised controlled trial. Lancet. 354, 1066–71 (1999). 110. Chen L, Liu P, Gao H et al. Inhalation of nitric oxide in the treatment of severe acute respiratory syndrome: a rescue trial in Beijing. Clin. Infect. Dis. 39, 153135 (2004). 111. Barrington KJ, Finer N. Inhaled nitric oxide for respiratory failure in preterm infants. Cochrane Database Syst. Rev. 12, CD000509 (2010). 112. Donohue PK, Gilmore MM, Cristofalo E et al. Inhaled nitric oxide in preterm infants: a systematic review. Pediatrics. 127, e414–22 (2011). 113. Sokol J, Jacobs SE, Bohn D. Inhaled nitric oxide for acute hypoxemic respiratory failure in children and adults: a meta-analysis. Anesth. Analg. 1, CD002787 (2003). 114. Adhikari NKJ, Burns KEA, Friedrich JO, Granton JT, Cook DJ, Meade MO. Effect of nitric oxide on oxygenation and mortality in acute lung injury: systematic review and meta-analysis. BMJ. 334, 779 (2007). 115. Afshari A, Brok J, Møller AM, Wetterslev J. Inhaled nitric oxide for acute respiratory distress syndrome and acute lung injury in adults and children: a systematic review with meta-analysis and trial sequential analysis. Anesth. Analg. 112, 1411–21 (2011). 116. Meade MO, Jacka MJ, Cook DJ et al. Survey of interventions for the prevention and treatment of acute respiratory distress syndrome. Crit. Care Med. 32, 946–54 (2004). 117. Dawson TC, Beck MA, Kuziel WA, Henderson F, Maeda N. Contrasting effects of CCR5 and CCR2 deficiency in the pulmonary inflammatory response to influenza A virus. Am. J. Pathol. 156(6), 1951–1959 (2000) 118. Carey MA, Bradbury JA, Seubert JM et al. Contrasting effects of cyclooxygenase-1 (COX-1) and COX-2 deficiency on the host response to influenza A viral infection. J. Immunol. 175(10), 6878 –6884 (2005). 119. Kobasa D, Jones SM, Shinya K et al. Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus. Nature 445(7125), 319–323 (2007).

Page 118: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

107

120. Perrone LA, Plowden JK, García-Sastre A, Katz JM, Tumpey TM. H5N1 and 1918 pandemic influenza virus infection results in early and excessive infiltration of macrophages and neutrophils in the lungs of mice. PLoS Pathog. 4(8), e1000115 (2008). 121. Perrone L, Szretter K, Katz J, Mizgerd J, Tumpey T. Mice lacking both TNF and IL-1 receptors exhibit reduced lung inflammation and delay in onset of death following infection with a highly virulent H5N1 virus. J. Infect. Dis. 202(8), 1161–1170 (2010). 122. Lin KL, Sweeney S, Kang BD, Ramsburg E, Gunn MD. CCR2-antagonist prophylaxis reduces pulmonary immune pathology and markedly improves survival during influenza infection. J. Immunol. Available at: http://jimmunol.org/content/early/2010/11/19/jimmunol.1001002.abstract. 123. Peiris J, Yu WC, Leung CW et al. Re-emergence of fatal human influenza A subtype H5N1 disease. Lancet 363(9409), 617–619 (2004). 124. de Jong MD, Simmons CP, Thanh TT et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat. Med. 12(10), 1203–1207 (2006). 125. Chan MCW, Cheung CY, Chui WH et al. Proinflammatory cytokine responses induced by influenza A (H5N1) viruses in primary human alveolar and bronchial epithelial cells. Respir. Res. 6, 135–147 (2005). 126. Cheung C, Poon L, Lau A et al. Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease? Lancet 360(9348), 1831–1837 (2002). 127. Chan MC, Chan RW, Yu WC et al. Tropism and innate host responses of the 2009 pandemic H1N1 influenza virus in ex vivo and in vitro cultures of human conjunctiva and respiratory tract. Am. J. Pathol. 176(4), 1828–1840 (2010). 128. Woo P, Tung E, Chan K et al. Cytokine profiles induced by the novel swine origin influenza A/H1N1 virus: Implications for treatment strategies. J. Infect. Dis. 201(3), 346–353 (2010). 129. Schmitz N, Kurrer M, Bachmann MF, Kopf M. Interleukin-1 Is responsible for acute lung immunopathology but increases survival of respiratory influenza virus infection. J. Virol. 79(10), 6441–6448 (2005). 130. Belisle SE, Tisoncik JR, Korth MJ et al. Genomic profiling of tumor necrosis factor alpha (TNF-α) receptor and interleukin-1 receptor knockout mice reveals a link between TNF-α signaling and increased severity of 1918 pandemic influenza virus infection. J. Virol. 84(24), 12576–12588 (2010). 131. Szretter KJ, Gangappa S, Lu X et al. Role of host cytokine responses in the pathogenesis of avian H5N1 Influenza Viruses in Mice. J. Virol. 81(6), 2736–2744 (2007).

Page 119: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

108

132. Salomon R, Hoffmann E, Webster RG. Inhibition of the cytokine response does not protect against lethal H5N1 influenza infection. Proc. Natl. Acad. Sci. U.S.A. 104(30), 12479 –12481 (2007). 133. Lin KL, Suzuki Y, Nakano H, Ramsburg E, Gunn MD. CCR2+ monocyte-derived dendritic cells and exudate macrophages produce influenza-induced pulmonary immune pathology and mortality. J. Immunol. 180(4), 2562–2572 (2008). 134. Aldridge JR, Moseley CE, Boltz DA et al. TNF/iNOS-producing dendritic cells are the necessary evil of lethal influenza virus infection. Proc. Natl. Acad. Sci. U.S.A. 106(13), 5306–5311 (2009). 135. Darwish I, Mubareka S, Liles WC. Immunomodulatory therapy for severe influenza. Expert Rev. Anti-Infect. Ther. 9(7), 807–22 (2011). 136. Pawliczak R, Logun C, Madara P et al. Influence of IFN-γ on gene expression in normal human bronchial epithelial cells: modulation of IFN-γ effects by dexamethasone. Physiol. Genomics 23(1), 28–45 (2005). 137. Meduri GU, Annane D, Chrousos GP, Marik PE, Sinclair SE. Activation and regulation of systemic inflammation in ARDS. Chest 136(6), 1631–1643 (2009). 138. Almawi WY, Beyhum HN, Rahme AA, Rieder MJ. Regulation of cytokine and cytokine receptor expression by glucocorticoids. J. Leukocyte Biol. 60(5), 563–572 (1996). 139. Ottolini M, Blanco J, Porter D et al. Combination anti-inflammatory and antiviral therapy of influenza in a cotton rat model. Pediatr. Pulmonol. 36(4), 290–294 (2003). 140. Chotpitayasunondh T, Ungchusak K, Hanshaoworakul W et al. Human disease from influenza A (H5N1), Thailand, 2004.(Research). Emerg. Infect. Dis. 11(2), 201(9) (2005). 141. Quispe-Laime A, Bracco J, Barberio P et al. H1N1 influenza A virus-associated acute lung injury: response to combination oseltamivir and prolonged corticosteroid treatment. Intensive Care Med. 36(1), 33–41 (2010). 142. Sugiyama K, Shirai R, Mukae H et al. Differing effects of clarithromycin and azithromycin on cytokine production in murine dendritic cells. Clin. Exp. Immunol. 147,540–546 (2007). 143. Hodge S, Hodge G, Jersmann H et al. Azithromycin improves macrophage phagocytic function and expression of mannose receptor in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 178, 139–148 (2008). 144. Amsden GW. Anti-inflammatory effects of macrolides—an underappreciated benefit in the treatment of community-acquired respiratory tract infections and chronic inflammatory pulmonary conditions? J. Antimicrob. Chemother. 55(1), 10–21 (2005).

Page 120: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

109

145. Yamaya M, Shinya K, Hatachi Y et al. Clarithromycin inhibits type A seasonal influenza virus infection in human airway epithelial cells. J. Pharmacol. Exp. Therapeutics 333(1), 81–90 (2010). 146. Sato K, Suga M, Akaike T et al. Therapeutic effect of erythromycin on influenza virus-induced lung injury in mice. Am. J. Respir. Crit. Care Med. 157(3), 853–857 (1998). 147. McCullers JA. Insights into the interaction between influenza virus and pneumococcus. Clin. Microbiol. Rev. 19(3), 571–582 (2006). 148. Smith MW, Schmidt JE, Rehg JE, Orihuela C, McCullers JA. Induction of pro- and anti- inflammatory molecules in a mouse model of pneumococcal pneumonia following influenza. Comp. Med. 57(1), 82–89 (2007). 149. McCullers JA. Effect of antiviral treatment on the outcome of secondary bacterial pneumonia after influenza. J. Infect. Dis. 190(3), 519–526 (2004). 150. Karlström Å, Boyd KL, English BK, McCullers JA. Treatment with protein synthesis inhibitors improves outcomes of secondary bacterial pneumonia after influenza. J. Infect. Dis. 199(3), 311–319 (2009). 151. Low D. Reducing antibiotic use in influenza: challenges and rewards. Clin. Microbiol. Infect. 14(4), 298–306 (2008).

152. Feldmann M. Development of anti-TNF therapy for rheumatoid arthritis. Nat. Rev. Immunol. 2(5), 364–371 (2002).

153. Hanauer SB, Sandborn WJ, Rutgeerts P et al. Human anti-tumor necrosis factor monoclonal antibody (Adalimumab) in Crohn's disease: the CLASSIC-I trial. Gastroenterology. 130(2), 323–333 (2006). 154. Beutler B, Milsark I, Cerami A. Passive immunization against cachectin/tumor necrosis factor protects mice from lethal effect of endotoxin. Science 229(4716), 869–871 (1985). 155. Fisher CJ, Agosti JM, Opal SM et al. Treatment of septic shock with the tumor necrosis factor receptor: Fc fusion protein. N. Engl. J. Med. 334(26), 1697–1702 (1996). 156. Peper RL, Van Campen H. Tumor necrosis factor as a mediator of inflammation in influenza A viral pneumonia. Microb. Pathog. 19(3), 175–183 (1995). 157. Hussell T, Pennycook A, Openshaw P. Inhibition of tumor necrosis factor reduces the severity of virus-specific lung immunopathology. Eur. J. Immunol. 31(9), 2566–2573 (2001).

Page 121: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

110

158. Askling J, Dixon W. The safety of anti-tumor necrosis factor therapy in rheumatoid arthritis. Curr. Opin. Rheumatol. 20(2), 138–144 (2008). 159. Tilley SL, Coffman TM, Koller BH. Mixed messages: modulation of inflammation and immune responses by prostaglandins and thromboxanes. J. Clin. Invest. 108(1), 15–23 (2001). 160. Rocca B, FitzGerald GA. Cyclooxygenases and prostaglandins: shaping up the immune response. Internat. Immunopharmacol. 2(5), 603–630 (2002). 161. Lee S, Cheung C, Nicholls J et al. Hyperinduction of cyclooxygenase-2–mediated proinflammatory cascade: A mechanism for the pathogenesis of avian influenza H5N1 infection. J. Infect. Dis. 198(4), 525–535 (2008). 162. Zheng B, Chan K, Lin Y et al. Delayed antiviral plus immunomodulator treatment still reduces mortality in mice infected by high inoculum of influenza A/H5N1 virus. Proc. Natl. Acad. Sci. U.S.A. 105(23), 8091–8096 (2008). 163. Carey MA, Bradbury JA, Rebolloso YD et al. Pharmacologic inhibition of COX-1 and COX-2 in influenza A viral infection in mice. PLoS ONE 5(7), e11610 (2010). 164. Fukunaga K, Kohli P, Bonnans C, Fredenburgh LE, Levy BD. Cyclooxygenase 2 plays a pivotal role in the resolution of acute lung injury. J. Immunol. 174(8), 5033–5039 (2005). 165. Spite M, Serhan CN. Novel lipid mediators promote resolution of acute inflammation. Circ. Res. 107(10), 1170–1184 (2010). 166. Cilloniz C, Pantin-Jackwood MJ, Ni C et al. Lethal dissemination of H5N1 influenza virus is associated with dysregulation of inflammation and lipoxin signaling in a mouse model of infection. J. Virol. 84(15), 7613–7624 (2010). 167. Fedson DS. Pandemic influenza: a potential role for statins in treatment and prophylaxis. Clin. Infect. Dis. 43(2), 199–205 (2006). 168. Rosenson RS. Pluripotent mechanisms of cardioprotection with HMG-CoA reductase inhibitor therapy. Am. J. Cardiovasc. Drugs 1(6), 411–420 (2001). 169. Jacobson JR. Statins in endothelial signaling and activation. Antiox. Redox. Signal. 11(4), 811–821 (2009). 170. Yang J, Huang C, Yang J, Jiang H, Ding J. Statins attenuate high mobility group box-1 protein induced vascular endothelial activation: a key role for TLR4/NF-κB signaling pathway. Mol. Cell. Biochem. 345(44), 189–195 (2010). 171. Wang H, Bloom O, Zhang M et al. HMGB-1 as a late mediator of endotoxin lethality in mice. Science 285, 248–251 (1999).

Page 122: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

111

172. Sha Y, Zmijewski J, Xu Z, Abraham E. HMGB1 develops enhanced proinflammatory activity by binding to cytokines. J. Immunol. 180(4), 2531–2537 (2008). 173. Han KH, Ryu J, Hong KH et al. HMG-CoA reductase inhibition reduces monocyte CC chemokine receptor 2 expression and monocyte chemoattractant protein-1-mediated monocyte recruitment in vivo. Circulation 111,1439–1447 (2005). 174. Kwong JC, Li P, Redelmeier DA. Influenza morbidity and mortality in elderly patients receiving statins: A cohort study. PLoS ONE. 4(11), e8087 (2009). 175. Vandermeer M, Thomas A, Kamimoto L et al. Association between use of statins and mortality among patients hospitalized with laboratory-confirmed influenza virus infections: a multistate study. J. Infect. Dis. 205(1), 13–19 (2012). 176. Walsh E. Statins and influenza: can we move forward? J. Infect. Dis. 205 (1), 1–3 (2012). 177. Muller S, Scaffidi P, Degryse B et al. The double life of HMGB1 chromatin protein: architectural factor and extracellular signal. EMBO J. 20(16), 4337–4340 (2001). 178. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 418(6894), 191–195 (2002). 179. Wang H, Yang H, Tracey KJ. Extracellular role of HMGB1 in inflammation and sepsis. J. Internal Med. 255(3), 320–331 (2004). 180. Yang H, Ochani M, Li J et al. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc. Natl. Acad. Sci. U.S.A. 101(1), 296–301 (2004). 181. Andersson U, Erlandsson-Harris H. HMGB1 is a potent trigger of arthritis. J. Internal Med. 255(3), 344–350 (2004). 182. Yang H, Wang H, Czura CJ, KJ Tracey. HMGB1 as a cytokine and therapeutic target. J. Endotoxin Res. 8(6), 469–472 (2002). 183. Alleva LM, Budd AC, Clark IA. Systemic release of high mobility group box 1 protein during severe murine influenza. J. Immunol. 181(2), 1454–1459 (2008). 184. Finney RSH, Somers GH. The anti-inflammatory activity of glycyrrhetinic acid and derivatives. J. Pharmacol. 10, 613–620 (1959). 185. Mollica L, De Marchis F, Spitaleri A et al. Glycyrrhizin binds to high-mobility group box 1 protein and inhibits its cytokine activities. Chem. Biol. 14(4), 431–441 (2007). 186. Abe N, Ebina T, Ishida N. Interferon induction by glycyrrhizin and glycyrrhetinic acid in mice. Microbiol. Immunol. 26(6), 535–539 (1982).

Page 123: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

112

187. Utsunomiya T, Kobayashi M, Pollard R, Suzuki F. Glycyrrhizin, an active component of licorice roots, reduces morbidity and mortality of mice infected with lethal doses of influenza virus. Antimicrob. Agents Chemother. 41(3), 551–556 (1997). 188. Wolkerstorfer A, Kurz H, Bachhofner N, Szolar OH. Glycyrrhizin inhibits influenza A virus uptake into the cell. Antiviral Res. 83(2), 171–178 (2009). 189. Ni YF et al. Glycyrrhizin inhibits COX-2 and iNOS in LPS-induced ALI. J. Surg. Res. 165, 329–335 (2011). 190. Tsoyi K, Jang HJ, Nizamutdinova IT et al. Metformin inhibits HMGB1 release in LPS-treated RAW 264.7 cells and increases survival rate of endotoxaemic mice. Brit. J. Pharmacol. 162(7), 1498–1508 (2011). 191. Bensinger SJ, Tontonoz P. Integration of metabolism and inflammation by lipid-activated nuclear receptors. Nature 454(7203), 470–477 (2008). 192. Bassaganya-Riera J, Song R, Roberts PC, Hontecillas R. PPAR-γ activation as an anti-inflammatory therapy for respiratory virus infections. Viral Immunol. 23(4), 343–352 (2010). 193. Cunard R, Ricote M, DiCampli D et al. Regulation of cytokine expression by ligands of peroxisome proliferator activated receptors. J. Immunol. 168(6), 2795–2802 (2002). 194. Budd A, Alleva L, Alsharifi M et al. Increased survival after gemfibrozil treatment of severe mouse influenza. Antimicrob. Agents Chemother. 51(8), 2965–2968 (2007). 195. Alleva LM, Cai C, Clark IA. Using complementary and alternative medicines to target the host response during severe influenza. eCAM 7(4), 501–510 (2010). 196. Zhao S, Ye H, Zhou H, Nie S, Li Q. Gemfibrozil reduces release of tumor necrosis factor-[alpha] in peripheral blood mononuclear cells from healthy subjects and patients with coronary heart disease. Clinica Chimica Acta. 332(1–2), 61–67 (2003). 197. Chinetti G, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm. Res. 49(10), 497–505 (2000). 198. Haraguchi G, Kosuge H, Maejima Y et al. Pioglitazone reduces systematic inflammation and improves mortality in apolipoprotein E knockout mice with sepsis. Intensive Care Med. 34(7), 1304–1312 (2008). 199. Di Gregorio GB, Yao-Borengasser A, Rasouli N et al. Expression of CD68 and macrophage chemoattractant protein-1 genes in human adipose and muscle tissues. Diabetes 54(8), 2305–2313 (2005). 200. Haraguchi G, Kosuge H, Maejima Y et al. Pioglitazone reduces systemic inflammation and

Page 124: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

113

improves mortality in apolipoprotein E knockout mice with sepsis. Intensive Care Med. 34, 1304–1312 (2008). 201. Moseley CE, Webster RG, Aldridge JR. Original Article: Peroxisome proliferator-activated receptor and AMP-activated protein kinase agonists protect against lethal influenza virus challenge in mice. Influenza Other Respi. Viruses 4(5), 307–311 (2010). 202. Zhao X, Zmijewski JW, Lorne E et al. Activation of AMPK attenuates neutrophil proinflammatory activity and decreases the severity of ALI. Am. J. Physiol. Lung Cell. Mol. Physiol. 295(3), L497–L504 (2008). 203. Pitocco D, Giubilato S, Zaccardi F et al. Pioglitazone reduces monocyte activation in type 2 diabetes. Acta. Diabetol. 46, 75–77 (2009). 204. Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 6(2), 230–47 (1968). 205. Prockop DJ. Marrow stromal cells as stem cells for nonhematopoetic tissue. Science. 276(5309), 71–4 (1997).

206. Bianco P, Robey PG. Marrow stromal stem cells. J. Clin. Invest. 105(12), 1663–8 (2000). 207. Dominici M, Le Blanc K, Mueller I et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytother. 8(4), 315–7 (2006). 208. Peister A, Mellad JA, Larson BL, Hall BM, Gibson LF, Prockop DJ. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood 103(5), 1662–8 (2004). 209. Rojas M, Xu J, Woods CR et al. Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am. J. Respir. Cell Mol. Biol. 33, 145–52 (2005). 210. Mei SH, McCarter SD, Deng Y, Parker CH, Liles CW, Stewart DJ. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 4(9), 1525–37 (2007). 211. Xu J, Qu J, Cao L et al. Mesenchymal stem cell-based angiopoietin-1 gene therapy for acute lung injury induced by lipopolysaccharide in mice. J. Pathol. 214, 472–81 (2008). 212. Bartholomew A, Sturgeon C, Siatskas M et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp. Hematol. 30, 42–48 (2002).

Page 125: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

114

213. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 105(4), 1815–22 (2005). 214. Meisel R, Zibert A, Laryea M, Gobel U, Daubener W, Dilloo D. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase -mediated tryptophan degredation. Blood 103, 4619–21 (2004). 215. Jiang XX, Zhang Y, Liu B et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte derived dendritic cells. Blood 105(10), 4120–6 (2005). 216. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 8(9), 726–736 (2008). 217. Sotiropolou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells 24(1), 74–85 (2006). 218. Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L. Mesenchymal stem cell natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, where as MSCs can inhibit IL-2 induced NK cell proliferation. Blood 107(4), 1484–90 (2006). 219.Raffaghello L, Bianchi G, Bertolotto M et al. Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem cells 26(1), 151–62 (2008). 220. Francois M, Romieu-Mourez R, Li M, Galipeau J. Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation. Mol. Ther. 20(1), 187–95 (2012). 221. Nemeth K, Leelahavanichkul A, Yuen PST et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E2-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 15(1), 42–49 (2009). 222. Newman R, Yoo D, LeRoux MA, Danilkovitch-Miagkova A. Treatment of inflammatory diseases with mesenchymal stem cells. Inflamm. All. Drug Targets. 8(2), 110 – 123 (2009). 223. Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic t-cells proliferation by human marrow stromal cells: implications in transplantation. Transplantation 75(3), 389–97 (2003). 224. Sundin M, Ringden O, Sundberg B, Nava S, Gotherstrom C, Le Blanc K. No alloantibodies against mesenchymal stromal cells, but presence of anti-fetal calf serum antibodies, after transplantation in allogeneic hematopoietic stem cell recipients. Haematologica. 92(9), 1208–15 (2007).

Page 126: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

115

225. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp. Hematol. 31, 890–896 (2003). 226. Stagg et al IFNg stimulated marrow stromal cells: a new type of non-hematopoetic APC. Blood 107(6), 2570–7 (2006). 227. Eliopoulos, N., J. Stagg, L. Lejeune, S. Pommey, J. Galipeau. Allogeneic marrow stromal cells are immune rejected by MHC class I and II mismatched recipient mice. Blood 106, 4057–4065 (2005). 228. Prigozhina TB, Khitrin S, Elkin G, Eizik O, Morecki S, Slavin S. Mesenchymal stromal cells lose their immunosuppressive potential after allotransplantation. Exp. Hematol. 36, 1370–6 (2008). 229. Chapel A, Bertho JM, Bensidhoum M et al. Mesenchymal stem cells home to injured tissues when co-infused with hematopoietic cells to treat a radiation-induced multi-organ failure syndrome. J. Gene Med. 5, 1028–38 (2003). 230. Spring H, Schuler T, Arnold B, Hammerling G, Ganss R. Chemokines direct endothelial progenitors into tumour noevessels. Proc. Natl. Acad. Sci. U.S.A. 102(50), 18111–6 (2005).

231. Croitoru-Lamoury J, Lamoury FMJ, Zaunders JJ, Veas LA, Brew BJ. Human mesenchymal stem cells constitutively express chemokines and chemokine receptors that can be upregulated by

cytokines, IFN-β, and copaxone. J. Interferon Cytokine Res. 27, 53–64 (2007). 232. Lee RH, Hsu SC, Munoz J. A subset of human rapidly self renewing marrow stromal cells preferentially engraft in mice. Blood 107(5), 2153–61 (2006). 233. Wang L, Li Y, Chen X et al. MCP-1, MIP-1, IL-8 and ischemic cerebral tissue enhance human bone marrow stromal cell migration in interface culture. Hematol. 7(2), 113–17 (2002).

234. Ji JF, He BP, Dheen T, Tay SSW. Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury. Stem cells 22(3), 415–27 (2004). 235. Honczarenko M, Le Y, Swierkowski M, Ghiran I, Glodek AM, Silberstein LE. Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells 24(4), 1030–41 (2006). 236. Son BR, Marquez-Curtis LA, Kucia M et al. Migration of bone marrow and cord blood mesenchymal stem cells in vitro is regulated by stromal derived factor – 1- cxcr4 and hepatocyte growth factor factor-c-met axes and involves matrix metalloproteinases. Stem Cells 24(5), 1254–64 (2006).

Page 127: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

116

237. Bianco P, Riminucci M, Gronthos S, Robey PG. Bone marrow stromal cells: nature, biology, and potential applications. Stem Cells 19(3),180–192 (2001). 238. Ren G, Su J, Zhang L et al. Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells 27, 1954–62 (2009). 239. Polchert D, Sobinsky J, Douglas G , Kidd M, Moadsiri A, Reina E et al. IFN-gamma actiation of mesenchymal stem cells for treatment and prevention of graft versus host disease. Eur. J. Immunol. 38, 1745–1755 (2008). 240. Sheng H, Wang Y, Jin Y et al. A critical role of IFNg in priming MSC-mediated suppression of T cell proliferation through up-regulation of B7-H1. Cell Res. 18, 846–57 (2008). 241. Chan JL, Tang KC, Patel AP et al. Antigen-presenting property of mesenchymal stem cells occurs during a narrow window at low levels of interferon-y. Blood 107(12), 4817–24 (2006). 242. Li W, Ren G, Huang Y et al. Mesenchymal stem cells: a double-edged sword in regulating immune responses. Cell Death Differ. epub ahead of print 16 March (2012). 243. Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheumat. 56(4), 1175–1186 (2007). 244. Mei SHJ, Haitsma JJ, Dos Santos CC et al. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am. J. Respir. Crit. Care Med. 182(8), 1047–57 (2010). 245. Ortiz LA, Gambelli F, McBride C. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc. Natl. Acad. Sci. U.S.A. 100(14), 8407–11 (2003). 246. Xu J, Woods CR, Mora AL et al. Prevention of endotoxin-induced systemic response by bone marrow-derived mesenchymal stem cells in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 293(1), L131–41 (2007).

247. Gupta N, Su X, Popov B, Lee JW, Serikov V, Matthay MA. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J. Immunol. 179(3), 1855–63 (2007).

248. Lee JW, Fang X, Gupta N, Serikov V, Matthay MA. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc. Natl. Acad. Sci. U.S.A. 106(38), 16357–62 (2009). 249. Djouad F, Fritz V, Apparailly F et al. Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor α in collagen-induced arthritis. Arthritis Rheum. 52, 1595–1603 (2005).

Page 128: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

117

250. Sudres M, Norol F, Trenado A et al. Bone marrow mesenchymal stem cells suppress lymphocyte proliferation in vitro but fail to prevent graft vs host disease in mice. J. Immunol. 176, 7761–67 (2006). 251. Inoue S, Popp FC, Koehl GE. Immunomodulatory effects of MSCs in a rat organ transplant model. Transplantation 81, 1589–96 (2006). 252. http://clinicaltrials.gov/ct2/home 253. Lee JW, Gupta N, Serikov V, Matthay MA. Potential application of mesenchymal stem cells in acute lung injury. Expert Opin. Biol. Ther. 9(10), 1259–1270 (2009). 254. Van Poll D, Parekkadan B, Cho CH et al. Mesenchymal stem cell-derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo. Hepatol. 47, 1634–43 (2008). 255. Ortiz LA, DuTreil M, Fattman C et al. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc. Natl. Acad. Sci. U.S.A. 104(26), 11002–7 (2007). 256. Yagi H, Soto-Gutierrez A, Navarro-Alvarez N et al. Reactive Bone Marrow Stromal Cells Attenuate Systemic Inflammation via sTNFR1. Mol. ther. 18(10), 1857–64 (2010). 257. McCarter SD, Mei SH, Lai PF et al. Cell-based angiopoietin-1 gene therapy for acute lung injury. Am. J. Respir. Crit. Care Med. 175, 1014–26 (2007). 258. Lee RH, Pulin AA, Seo MJ et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 5, 54–63 (2009). 259. Danchuk S, Ylostalo J, Hossain F et al. Human multipotent stromal cells attenuate lipopolysaccharide-induced acute lung injury in mice via secretion of tumor necrosis factor-α-induced protein 6. Stem Cell Res. Ther. 2(3), 27 (2011). 260. Nemeth et al. Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proc. Natl. Acad. Sci. U.S.A. 107, 5652–57 (2010). 261. Yancopoulos GD, Davis S, Gale NW et al. Vascular-specific growth factors and blood vessel formation. Nature 407(6801), 242–248 (2000). 262. Davis S, Aldrich TH, Jones PF et al. Isolation of angiopoietin-1, a ligand for the tie2 receptor, by secretion-trap expression cloning. Cell 87(7), 1161–1169 (1996). 263. Thurston G, Suri C, Smith K et al. Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1. Science 286(5449), 2511–2514 (1999).

Page 129: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

118

264. Thurston G, Rudge JS, Ioffe E et al. Angiopoietin-1 protects the adult vasculature against plasma leakage. Nat. Med. 6(4), 460–463 (2000). 265. Gamble JR, Drew J, Trezise L et al. Angiopoietin-1 is an antipermeability and anti-inflammatory agent in vitro and targets cell junctions. Circ. Res. 87(7), 603–7 (2000). 266. Sugahara K, Iyama K, Kuroda MJ, Sano K. Double intratracheal instillation of keratinocyte growth factor prevents bleomycin-induced lung fibrosis in rats. J. Pathol. 186, 90–8 (1998). 267. Bernard DL. Animal models for the study of influenza pathogenesis and therapy. Antiviral Res. 82(2), A110–22 (2009). 268. Van Reeth K. Cytokines in the pathogenesis of influenza. Vet. Microbiol. 74(1–2), 109–16 (2000). 269. Matsuoka Y, Lamirande EW, Subbarao K. The mouse model for influenza. Curr. Protoc. Microbiol. Chapter 15, Unit 15G.3 (2009). 270. O'Donnell CD, Subbarao K. The contribution of animal models to the understanding of the host range and virulence of influenza A viruses. Microb. Infect. 13, 502–15 (2011). 271. Gubareva LV, McCullers JA, Bethell RC, Webster RG. Characterization of influenza /Hong Kong/156/97 (H5N1) virus in a mouse model and protective effect of zanamivir on H5N1 infection in mice. J. Infect. Dis. 178(6), 1592–96 (1998). 272. Belser JA, Szretter KJ, Katz JM, Tumpey TM. Use of animal models to understand the pandemic potential of highly pathogenic avian influenza viruses. Adv. Virus Res. 73, 55– 97 (2009). 273. Castrucci MR, Kawaoka Y. Biologic importance of neuraminidase stalk length in influenza A viruses. J. Virol. 67(2), 759–764 (1993). 274. McMullin B, Chittock D, Roscoe D, Garcha H, Wang L, Miller C. The antimicrobial effect of nitric oxide on the bacteria that cause nosocomial pneumonia in mechanically ventilated patients in the ICU. Resp Care. 50, 1451–56 (2005). 275. Miller C, McMullin B, Ghaffari A, Miller J, Stenzler A, Pick N, Roscoe D, Ghahary A, Road J, Av Gay Y. Gaseous nitric oxide bactericidal activity retained during intermittent high-dose short duration exposure. Nitric Oxide. 20, 16–23 (2009). 276. Davidson D, Barefield ES, Kattwinkel J et al. Inhaled nitric oxide for the early treatment of persistent pulmonary hypertension of the term newborn: a randomized, double-masked, placebo-controlled, dose-response, multi-center study. The I-NO/PPHN Study Group. Pediatrics. 1, 325–34 (1998).

Page 130: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

119

277. Leibbrandt A, Meier C, Kong-Schuster et al. Iota-carrageenan is a potent inhibitor of influenza A virus infection. PLoS One. 5(12), e14320 (2010). 278. Brower RG, Matthay MA, Morris A et al. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N. Engl. J. Med. 342(18), 1301–1308 (2000). 279. Sekiya I, Larsen B, Smith J, Pochampally R, Cui J-G, Prockop D. Expansion of Human Adult Stem Cells from Bone Marrow Stroma: Conditions that Maximize the Yields of Early Progenitors and Evaluate Their Quality. Stem Cells 20, 530–541 (2002). 280. Kim H, Kim HY, Choi MR et al. Dose-dependent efficacy of ALS-human mesenchymal stem cells transplantation into cisterna magna in SOD1-G93A ALS mice Neurosci. lett. 468 (3), 190–194 (2010). 281. Liotta F, Angeli R, Cosmi L et al. Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing Notch signaling. Stem Cells 26(1), 279–89 (2008). 282. Ahrlundt-Richter L, De Luca M, Marshak DR, Munsie M, Veiga A, Rao M. Isolation and production of cells suitable for human therapy: challenges ahead. Cell Stem Cell 4(1), 20–6 (2008). 283. Wong ZX, Jones JE, Anderson GP, Gualano RC. Oseltamivir treatment of mice before or after mild influenza infection reduced cellular and cytokine inflammation in the lung. Influenza Other Resp. Viruses. 5(5), 343–50 (2011). 284. Jefferson T, Jones M, Doshi P, Del Mar C, Dooley L, Foxlee R. Neuraminidase inhibitors for preventing and treating influenza in healthy adults. Cochrane Database Syst. Rev. 17(2), CD001265 (2010). 285. Mindrescu C, Le J, Wisniewski HG, Vilcek J. Up-regulation of cyclooxygenase-2 expression by TSG-6 protein in macrophage cell line. Biochem. Biophys. Res. Commun. 330, 737–745 (2005). 286. Bartosh TJ, Ylostalo JH, Mohammadipoor A et al. Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. Proc. Natl. Acad. Sci. U.S.A. 107 (31), 13724–13729 (2010). 287. Karoubi G, Ormiston ML, Stewart DJ, Courtman DW. Single-cell hydrogel encapsulation for enhanced survival of human marrow stromal cells. Biomaterials. 20(29), 5445–55 (2009). 288. London NR, Zhu W, Bozza FA et al. Targeting robo4-dependent slit signaling to survive the cytokine storm in sepsis and influenza. Science Translat. Med. 2(23), 23ra19 (2010). 289. Jones CA, London NR, Chen H et al. Robo4 stabilizes the vascular network by inhibiting

Page 131: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

120

pathologic angiogenesis and endothelial hyperpermeability. Nat. Med. 14(4), 448–453 (2008). 290. Sheu TG, Fry AM, Garten RJ et al. Dual resistance to adamantanes and oseltamivir among seasonal influenza A (H1N1) viruses: 2008–2010. J. Infect. Dis. 203(1), 13–17 (2011). 291. Ozawa M, Fujii K, Muramoto Y et al. Contributions of two nuclear localization signals of influenza A virus nucleoprotein to viral replication. J. Virol. 81(1), 30–41 (2007). 292. Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect. Dis. 12(1), 36–44 (2012). 293. Peterson E, Keld DB, Ellermann-Eriksen S et al. Failure of combination oral oseltamivir and inhaled zanamivir antiviral treatment in ventilator- and ECMO-treated critically ill patients with pandemic influenza A (H1N1). Scand. J. Infect. Dis. 43(6–7), 495–503 (2011). 294. McGeer A, Green KA, Plevneshi A et al. Antiviral therapy and outcomes of influenza requiring hospitalization in Ontario, Canada. Clin. Infect. Dis. 45(12), 1568–75 (2007). 295. Lee N, Choi KW, Chan PKS et al. Outcomes of adults hospitalized with severe influenza. Thorax 65(6), 510–5 (2010). 296. Ray WA, Murray KT, Hall K, Arbogast PG, Stein CM. Azithromycin and the risk of cardiovascular death. N. Engl. J. Med. 366, 1881–90 (2012). 297. Schmitt AP, Lamb RA. Influenza virus assembly and budding at the viral budozone. Adv. Virus Res. 64, 383–416 (2005). 298. Fang X, Neyrinck AL, Matthay MA, Lee JW. Allogeneic human mesenchymal stem cells restore epithelial protein permeability in cultured human alveolar type II cells by secretion of Angiopoeitin-1. J. Biol. Chem. 285, 26211-22 (2010). 299. Centers for Disease Control and Prevention: Available at: http://www.cdc.gov/flu/weekly/. Accessed July 4, 2012.

300. Wagaman PC, Spence HA, O'Callaghan RJ. Detection of influenza C virus by using an in situ esterase assay. J. Clin. Microbiol. 27(5), 832–6 (1989).

301. Centers for Disease Control and Prevention: Available at: http://www.cdc.gov/flu/avianflu/influenza-a-virus-subtypes.htmhttp://www.cdc.gov/flu/weekly/. Accessed July 4, 2012.

302. Leyva-Grado VH, Mubareka S, Krammer F, Cardenas WB, Palese P. Influenza virus

infection in guinea pigs raised as livestock, Ecuador. Emerg. Infect. Dis.18(7), [epub ahead of

print] (2012).

Page 132: INVESTIGATION OF INHALED NITRIC OXIDE AND … · 2013-10-25 · Ilyse Darwish Masters of Science Institute of Medical Science University of Toronto 2012 Abstract Severe influenza,

121

303. Boon ACM, deBeachamp J, Hollmann A et al. Host genetic variation affects resistance to

infection with a highly pathogenic H5N1 influenza A virus in mice. J. Virol. 83(20), 10417–26

(2009).