Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

JPT-107712; No of Pages 18

Pharmacology & Therapeutics xxx (2020) xxx

Contents lists available at ScienceDirect

Pharmacology & Therapeutics

journal homepage: www.elsevier.com/locate/pharmthera

Pharmacological targets and emerging treatments for respiratory


syncytial virus bronchiolitis
Farah Elawar a,1, Ahmed K. Oraby b,c,1, Quinten Kieser a,1, Lionel D. Jensen a, Tyce Culp a,
Frederick G. West b,⁎, David J. Marchant a,⁎⁎
a
Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
b
Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
c
Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Misr University for Science &Technology, Al-Motamayez District, 6th of October City, P.O. Box 77, Egypt

a r t i c l e i n f o a b s t r a c t

Available online xxxx RSV infection of the lower respiratory tract in infants is the leading cause of pediatric hospitalizations and second
to malaria in causing infant deaths worldwide. RSV also causes substantial morbidity in immunocompromised
and elderly populations. The only available therapeutic is a prophylactic drug called Palivizumab that is a human-
Keywords: ized monoclonal antibody, given to high-risk infants. However, this intervention is expensive and has a limited
Therapeutic impact on annual hospitalization rates caused by RSV. No vaccine is available, nor are efficacious antivirals to
Prevention treat an active infection, and there is still no consensus on how infants with bronchiolitis should be treated during
Respiratory syncytial virus hospital admission. In this comprehensive review, we briefly outline the function of the RSV proteins and their
Respiratory tract infection suitability as therapeutic targets. We then discuss the most promising drug candidates, their inhibitory mecha-
Pediatric
nisms, and whether they are in the process of clinical trials. We also briefly discuss the reasons for some of the
failures in RSV therapeutics and vaccines. In summary, we provide insight into current antiviral development
and the considerations toward producing licensed antivirals and therapeutics.
© 2020 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://
creativecommons.org/licenses/by-nc-nd/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. New antiviral therapeutics are needed to stem the burden of RSV disease. . . . . . . . . . . . . . . . . . . . 0
3. RSV spread and adaptation in the community . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. RSV proteins, their functions and potential as therapeutic targets . . . . . . . . . . . . . . . . . . . . . . . 0
5. An overview of RSV therapeutic development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
6. Vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7. Prophylaxis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
8. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Funding sources . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

Abbreviations: CLD, chronic lung disease; CPE, cytopathic effect; ERD, enhanced respiratory disease; GE, gene end; GS, gene start; ICU, intensive care unit; IAV, influenza A virus; IBV,
influenza B virus; IFN, interferon; LRTI, lower respiratory tract infection; MAVS, mitochondria antiviral signaling molecule; MeV, measles virus; PFU, plaque forming unit; PKR, protein
kinase R; PVM, pneumonia virus of mice; RdRp, RNA-dependent RNA polymerase; RSV, respiratory syncytial virus; RV, rhinovirus; HRB, heptad repeat B.
⁎ Corresponding author at: Department of Chemistry, University of Alberta, Edmonton, Alberta T6G-2G2, Canada.
⁎⁎ Corresponding author.
E-mail address: marchant@ualberta.ca (D.J. Marchant).
1
These authors contributed equally.

https://doi.org/10.1016/j.pharmthera.2020.107712
0163-7258/© 2020 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Please cite this article as: F. Elawar, A.K. Oraby, Q. Kieser, et al., Pharmacological targets and emerging treatments for respiratory syncytial virus
bronchiolitis, Pharmacology & Therapeutics, https://doi.org/10.1016/j.pharmthera.2020.107712
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

1. Introduction efficacy of therapeutics for the treatment and prevention of RSV


means that development of therapeutics in this arena is ongoing and
Respiratory Syncytial Virus (RSV) is a member of the Orthopn- rapidly gaining more attention.
eumovirus genus, Pneumoviridae family, and Mononegavirales order
(Rima et al., 2017). RSV has two major sub-types, type A and type B, 1.3. Treating the symptoms of RSV infections: reducing inflammation and
that share disease manifestations and 95% sequence identity. Originally maintaining an open airway
referred to as Chimpanzee Coryza Agent, RSV was first identified in
1955 in a chimpanzee colony at the Walter Reed Army Institute of Humans are born with almost all of the airways and alveoli air sacs
Research (Blount Jr., Morris, & Savage, 1956). RSV was immediately that they will ever have (Hislop, 2002). Consequently, airways are a
suspected as a human pathogen, as the authors noted that RSV neutral- smaller diameter in children, placing them at higher risk of being oc-
izing antibodies were present in a laboratory worker who contracted an cluded during respiratory infection. The main cause of airway occlusion
upper respiratory tract infection after working closely with infected is the inflammation resulting from infection of the airways. Thus, in the
chimpanzees. Soon after, pre-existing neutralizing antibodies were emergency room preventing and or alleviating airway inflammation is
detected in the serum of patients who had no contact with the chimpan- the primary form of therapeutic intervention that is available to pediat-
zee colony. RSV was successfully isolated from a human patient in 1957 ric patients with severe RSV infection (R. B. Wright, Pomerantz, & Luria,
(R. Chanock & Finberg, 1957).In subsequent years, the importance of 2002). The treatment of croup and bronchiolitis caused by RSV is now
RSV as a major contributor of disease in pediatric (Andrew & Gardner, largely treated with corticosteroids and beta agonists. These are most
1963) and elderly (Morales, Calder, Inglis, Murdoch, & Williamson, frequently in the form of inhaled aerosols and come in many different
1983) populations became apparent. beta-agonist-corticosteroid combinations. In a recent review of cortico-
steroid use and emergency room visits, the data concluded corticoste-
1.1. The purpose of this review roids are effective and alleviate relapse events (Kirkland, Cross,
Campbell, Villa-Roel, & Rowe, 2018).
Despite the discovery of RSV dating back over six decades, no vac-
cine to prevent infection or efficacious antiviral for treating infection 1.4. Long and short acting beta-adrenergic agonists
are available. Lack of licensed medication is not due to a lack of drug de-
velopment activity as there have been several high-profile failures of Constriction of airways occurs during inflammation and is mediated
RSV vaccines and therapeutics in the last four years (Table 1). In the primarily by smooth muscle cells that surround the epithelial lined air-
fall of 2018 and winter of 2020, three promising compounds that all ways tubes. The observation that beta-agonist stimulation could allevi-
targeted different stages of RSV replication were withdrawn from clini- ate constriction in the airways during asthma was first reported in the
cal trials (Marty et al., 2019). Furthermore, there were two high-profile 1960s (Kiyomoto, Iwasawa, & Harigaya, 1970; Schuster & Baum,
failed vaccine trials of different strategies (Ruckwardt, Morabito, & 1969), and beta-agonists have since been in use as inhaled rescue and
Graham, 2019). While these vaccines saw little success in a primary set- maintenance medications for airway constriction, particularly in asthma
ting, there was limited achievement in reducing both hospitalization (Kiyomoto et al., 1970; Schuster & Baum, 1969). The ‘beta-agonists’
due to lower respiratory tract infection and severe hypoxemia. As a re- come in two main classes, the short and long-acting beta-adrenergic ag-
sult, these vaccines may see restricted use exclusively for high-risk pop- onists, frequently referred to as their SABA and LABA acronyms,
ulations (Jares Baglivo & Polack, 2019). Therefore, RSV continues to respectively. Given their success in asthma they are also used as emer-
impose a significant global burden of disease with few experimental gency administered medications for occluded and inflamed airways
therapeutic options. In this review, we discuss the classes of compounds resulting from respiratory infections. These are often provided as in-
in clinical trials, their targets, and some of the recent and historical fail- haled powder or even nebulised in saline vapour. The vapour helps
ures of RSV therapeutics. It is only after learning from the mistakes of loosen mucus in lungs and so they are frequently used in combination
the past and incorporating a thorough understanding of RSV replication with glucocorticoids (Newton & Giembycz, 2016).
that we will develop efficacious therapies and vaccines. It is important to note that not all SABAs and LABAs may be used as
rescue medications where rapid onset of action is most desirable
1.2. The current state of RSV vaccines, therapeutics and prophylaxis (Cazzola, Beeh, Price, & Roche, 2015). For instance, salbutamol (albute-
rol), a SABA, and formoterol, a LABA, can be used as rescue medication
Currently, there are two therapeutics licensed for managing disease due to their quick onset of action. However salmeterol, another LABA,
due to severe RSV infection. Ribavirin, the first licensed therapeutic, is should not be used as a rescue medication due to slow inset of action.
an antiviral nucleoside analogue that was recommended for the treat- Rapid onset SABAs and LABAs may be used as rescue, although it is
ment of severe RSV infection in 1993. However, clinical trials leading only LABAs that are currently paired with corticosteroids in combina-
to this recommendation were methodologically flawed, and the efficacy tion therapies (Sinha et al., 2020). The purpose of keeping SABA treat-
of ribavirin was exaggerated (Smith, 1991; Smith et al., 1991). Subse- ments separate from that of inhaled corticosteroids (ICS) is to reduce
quent studies found that the benefits of ribavirin were limited or non- over-use of SABA therapy, as increased SABA reliance can lead to exac-
existent (Law et al., 1997; Meert, Sarnaik, Gelmini, & Lieh-Lai, 1994), erbation of asthma symptoms (O'Byrne et al., 2018; Sinha et al., 2020).
and ribavirin is no longer included as a standard of care for RSV infec- Common ICS/LABA combinations are budesonide/formoterol or
tion. The only currently licensed prophylaxis, Palivizumab, a humanized fluticasone furoate/salmeterol (Newton & Giembycz, 2016). Many
monoclonal antibody against RSV, was licensed in 1998 for the prophy- other combinations have also been reported, including beclomet-
lactic prevention of RSV infections. Due to its high cost, limited efficacy, hasone/formoterol, fluticasone/formoterol, and fluticasone/vilanterol
and the necessity of monthly prophylactic injections, Palivizumab is (Chantaphakul & Ruxrungtham, 2016).
only administered to high-risk infants. Since a majority of hospitalized
infants are not eligible for Palivizumab, there is no decrease in hospital- 1.5. The question of efficacy of inhaled beta-agonists in infants during se-
ization rates in full term infants that contract RSV from older siblings vere RSV bronchiolitis
(Hardelid, Verfuerden, McMenamin, Smyth, & Gilbert, 2019). Paliviz-
umab is not efficacious in the treatment of ongoing RSV infection and It is currently debated as to whether beta-agonists should be used as
has a less significant effect on overall RSV transmission rates than previ- rescue therapy for RSV infections in infants. A study by Derish et al.
ously thought (Murray et al., 2014; Robinson, Le Saux, Canadian showed that infants who were treated with inhaled albuterol during
Paediatric Society, & Immunization, 2015). In summary, the limited the acute phase of severe RSV infection benefited (Derish, Hodge, Dunn,

2
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Table 1
Recent RSV antivirals in devlopment or in clinical trivals.

Compound Name Development stage Mechanism of action Citation

JNJ-53718678 Phase 2 CT completed Small molecule RSV-F inhibitor {Stevens, 2018 #842}
(JNJ-678) NCT02387606 NCT03379675
GS-5806 (Presatovir) Phase 2b CT completed Small molecule RSV-F inhibitor {Perron, 2015 #453;DeVincenzo, 2014
#841} NCT02135614
BTA-C585 Phase 2 CT completed Small molecule RSV-F inhibitor {Heylen, 2017 #663} NCT02718937
ALX-0171 Phase 1 and 2 CT ongoing Antibody-like RSV-F inhibitor {Detalle, 2016 #459} NCT02979431
NCT03418571
AK-0529 Phase 1b and 2 CT ongoing Small molecule RSV-F inhibitor NCT02654171 NCT02460016
MDT 637 Phase 1 CT completed; results not released Small molecule RSV-F inhibitor {Douglas, 2005 #461} NCT01355016
(VP-14637)
BMS-433771 No investigation in CT Small molecule RSV-F inhibitor {Cianci, 2005 #462}
TMC-353121 No investigation in CT Small molecule RSV-F inhibitor {Bonfanti, 2008 #464}
ALN-RSV01 Phase 2b CT; partial success siRNA targeting RSV mRNA encoding RSV-N {Gottlieb, 2016 #456} NCT01065935
RSV604 Phase 1 CT completed; results not released Binds RSV-N protein; blocks RNA synthesis {Challa, 2015 #420} NCT00416442
ALS-008176 Phase 2 CT completed; development on hold, Targets RSV-L; causes chain termination {Deval, 2015 #418;DeVincenzo, 2015
(lumicitabine) results not released #457}
NCT02673476
PC786 Phase 2 CT ongoing Targets RSV-L {Coates, 2017 #662}
NCT03382431
AZ-27 No investigation in CT Inhibits de novo RNA synthesis initiation by RSV {Noton, 2015 #466}
BI-D No investigation in CT Inhibits RSV mRNA elongation and capping {Fearns, 2016 #463}
producing IFN response
YM-53403 No investigation in CT Targets RSV L {Sudo, 2005 #419}
GS-5734 Phase 2 CT ongoing* Inhibits polymerization by RSV-L in vitro {Warren, 2016 #478} NCT02818582
BCX-4430 Phase 1 CT* Inhibits polymerization by RSV-L in vitro {Warren, 2014 #465} NCT02319772

& Ariagno, 1998), while others have argued that inhaled albuterol given antivirals. In fact, RSV has been recognized by the World Health Organi-
to infants was of “limited value as a bronchodilator” (Hammer, Numa, zation (WHO) as the number one vaccine priority (R. M. Chanock et al.,
& Newth, 1995). The most recent Meta-analyses that studied the ques- 1988). Anti-infectives are likely to be utilized most during neonatal in-
tion of inhaled beta-agonist treatment during severe acute RSV infection tensive care unit (NICU) stays and shortly after discharge (Leader
concluded that there was no benefit in improving oxygen saturation or et al., 2002; Mitchell, Defoy, & Grubb, 2017). While individuals will be
length of hospital stay, and that they should be avoided as treatments re-infected with the same strain of RSV throughout life, the primary in-
for infant bronchiolitis (Cai, Lin, & Liang, 2020; Gadomski & Scribani, fection during infancy is typically the most severe and so is of the
2014). Of primary concern is the tachycardia side effects of beta- highest priority. In the community, approximately 40% of all primary in-
agonists in the very young. Both meta-analysis studies added that large fections in infancy result in lower respiratory tract infections (LRTI),
scale multi-center clinical trials were required to better address the ques- which manifest as bronchiolitis or pneumonia. However, not all of
tion of beta-agonist bronchodilator use during infant bronchiolitis. these cases will be reported to the hospital and 0.5-2% of all otherwise
healthy infants will require hospitalization due to RSV LRTI (B. Paes,
1.6. Against the use of corticosteroids for rescue treatment of infant Mitchell, Li, & Lanctot, 2012). In Canada and the United States, RSV
bronchiolitis LRTI in infants results in approximately 12 000 and 77 000 annual hos-
pitalizations, respectively (B. A. Paes, Mitchell, Banerji, Lanctot, &
Inhaled corticosteroids are administered regularly in those who suf- Langley, 2011).
fer from moderate to severe asthma and in those with chronic obstruc- In 2010, Nair et al. estimated that LRTI caused by RSV in children
tive pulmonary disease. These are provided in the form of inhaler, under five years old resulted in 3.4 million hospitalizations and 66 000
frequently as a colloid delivered with either inhaled breath or to 199 000 deaths annually worldwide (examining data from 2005)
hydrofluoroalkane propellant gas. It was logical to assume that these (Nair et al., 2010). A decade later, the RSV Global Epidemiology Network
medications could be beneficial for the treatment of bronchiolitis observed a comparable worldwide burden of disease in children under
where acute airway inflammation is a primary cause of airway constric- five years of age, with RSV LRTI responsible for 3.2 million hospital
tion. However, whether inhaled corticosteroids like fluticasone and admissions and 94 600 to 149 400 deaths annually (Shi et al., 2017).
budesonide are beneficial in the treatment of acute respiratory distress RSV-related mortality disproportionately affects developing countries
in adults, remains unclear, and even less so in pediatric patients (Peter relative to Western nations. Unfortunately, in these settings newly
et al., 2008). A recent report on the management of admitted bronchiol- licensed therapeutics will likely not be available initially due to the tre-
itis cases advised against the use of inhaled glucocorticoid corticosteroids mendous financial cost of patent-protected non-generic drugs. Never-
(Karampatsas, Kong, & Cohen, 2019; Mecklin, Heikkila, & Korppi, 2018), theless, it is developing countries where RSV drugs are needed most.
an advisory that is consistent with bronchodilators in bronchiolitis. However, if treatment of the human immunodeficiency virus (HIV)
In summary, there is still no consensus on how infants with bronchi- pandemic is to be a model, then generic production of any licensed
olitis should be treated in the hospital setting. Therefore, there is a great RSV drugs would be beneficial. Generic drugs that were produced in
need to develop emergency rescue medications for infants admitted to the Indian, Brazilian and South African biotechnology complex were of
hospital with RSV bronchiolitis. tremendous benefit in resource poor settings (Ruckwardt et al., 2019).
They contributed greatly to reducing the burden of HIV in sub-
2. New antiviral therapeutics are needed to stem the burden of RSV Saharan Africa. Although, efforts such as these typically don’t come
disease without legal challenges to possible patent infringement (Ruckwardt
et al., 2019). Nevertheless, generically manufactured drugs at a fraction
RSV is a leading cause of both infant mortality and hospitalization of the cost of the name brand product may help to fulfil the need to stem
worldwide (Shi et al., 2019), so there is a great need for direct-acting the burden of RSV in these settings.

3
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

2.1. The RSV viral-load infection profile compared to influenza and the im- Aries, Argentina) clade emerged in 1999 and became the dominant
plications for treatment global RSV type B clade in the ensuing decades (Trento et al., 2010).
RSV BA strain continued to evolve with four genotypes emerging in
Acute viral infections can be difficult to treat with antivirals as the the ten years following its emergence. We have proposed that this be-
patient typically must be treated shortly after the onset of symptoms haviour supports our own proposed model of the emergence of RSV
for the intervention to be effective. In other words, intervention into a community by high-titer clades (Elawar et al., 2017). The high-
must be effective before the viral load reaches its peak, thus titer clade hypothesis will be described in further detail below. These as-
preventing viral spread and damage to the host’s cells from both di- pects of RSV adaptation/mutation suggest that it will be important to
rect, virally induced cytopathic effects and the immune response to modify or even replace drugs as new RSV strains evolve.
infected cells. Of course, antivirals only offer therapeutic potential
if viral replication drives disease; if viral load has already been con-
3.2. RSV transmission
trolled by the immune response at the time of symptom onset then
an antiviral will be of little benefit. This important consideration is
RSV is primarily transmitted via large nasopharyngeal droplets
exemplified by influenza A virus (IAV), where peak viral shedding
from infected individuals (B. A. Paes et al., 2011). Large droplets
precedes peak symptom severity (Carrat et al., 2008). Neuramini-
(>100 μm in size) remain in the air for only a matter of seconds,
dase inhibitors such as zanamivir (Relenza) and oseltamivir
quickly falling onto surfaces due to gravity (Morawska, 2006).
(Tamiflu) effectively inhibit IAV replication in vitro and have been
Therefore, direct contact with an infected individual, or contact
stockpiled to address the threat posed by pandemic influenza out-
with contaminated surfaces, is required for transmission via large
breaks. However, the clinical benefits of these antivirals are limited.
droplets. Infectious RSV can be isolated from solid surfaces 6 hours
Zanamivir and oseltamivir have no effect on hospitalization rates or
after transfer and can survive on hands for 25 minutes (Hall,
serious complications, and facilitate only a minor decrease in the
Douglas Jr., & Geiman, 1980). Rubbing one’s eyes and face with
time to symptom alleviation (Jefferson et al., 2014). This could be
hands contaminated by these surfaces can lead to infection, as RSV
due tothe rapid rise and then fall of influenza viral load that reaches
gathers a foothold on the host here by first replicating in the con-
a crescendo within two days of symptom onset. It is within this two-
junctiva of the eyes or in the nose (HaHall et al., 1980). It then goes
day window that patients must receive zanamivir and oseltamivir.
on to infect the upper and lower respiratory tracts.
After this point the damage has been done by viral replication and
Recently, it was observed that viable RSV is present in aerosols pro-
the immune system suppresses infection, matching infection
duced by infants with bronchiolitis (Kulkarni et al., 2016). Air was sam-
magnitude.
pled at distances of 1 metre, 5 metres and 10 metres from infected
Human challenge studies that compared the viral load progression
infants. Air was also sampled in both the presence of infants and 2
of RSV to influenza showed there was a significantly longer window of
hours after discharge. RSV titers from airborne particles were highest
viral shedding to treat RSV infection (Bagga et al., 2013), suggesting
at a distance of 1 metre; however, RSV was detected up to a distance
that RSV infections will be more responsive to antivirals than influenza.
of 5 metres from patients. RSV titers were highest when sampling was
Furthermore, in experimentally inoculated adult volunteers, RSV repli-
conducted in the presence of the patient, yet aerosols containing viable
cation and titers mirrored symptom severity (J. P. DeVincenzo et al.,
RSV remained detectable at 2 hours post-discharge. Furthermore, RSV
2010). Likewise, in hospitalized children, RSV load paralleled disease se-
was found in particles less than 4.7 μm in diameter. While large diame-
verity (El Saleeby, Bush, Harrison, Aitken, & Devincenzo, 2011), unlike
ter particles are typically caught in the upper respiratory tract, particles
influenza where peak symptoms followed after the peak of viral load
less than 5 μm in size may be directly inhaled into the lower airways
(Carrat et al., 2008). Therefore, when an RSV antiviral becomes avail-
(Kulkarni et al., 2016; Roy & Milton, 2004). The possibility of airborne
able, the drug is expected to have a reasonable window of opportunity
transmission of RSV has implications for public health measures used
to treat the infected individual.
to prevent nosocomial RSV transmission.
Surfaces contaminated with RSV represent another major source of
3. RSV spread and adaptation in the community
transmission (Hall et al., 1980). RSV conjugated with air pollution can
last for weeks on a dry surface (Cruz-Sanchez et al., 2013), and serves
3.1. RSV seasonality
as a significant source of nosocomial transmission. Thus, the best pro-
phylaxis measures to protect from RSV infection are likely hand-
In temperate climates, such as North America, the RSV season lasts
washing, stifled coughs-sneezes, and social distancing.
3-5 months; typically beginning in autumn, peaking in winter, and ta-
pering off in the spring. RSV transmission is significantly reduced, or
not detected, during the summer months (C. Griffiths, Drews, & 3.3. Adaptation of RSV and the potential for drug resistance
Marchant, 2017; Janet, Broad, & Snape, 2018; Sloan et al., 2017;
Welliver Sr., 2007). In contrast, very cold or tropical climates do not Owing to the low fidelity of its RNA-dependent RNA polymerase
obey this pattern, and RSV transmission occurs year-round (Janet (RdRp), RSV evolves rapidly. The constant population-level evolution
et al., 2018; Welliver Sr., 2007). A variety of hypotheses have been pro- of RSV results in the replacement of antigenic epitopes and contributes
posed to explain the transmission patterns observed across these differ- to the explanation of how RSV successfully reinfects individuals
ent regions, including changes to viral stability in the environment that throughout life (Zlateva, Lemey, Vandamme, & Van Ranst, 2004). This
are dependent on temperature, humidity, and ultraviolet light intensity. viral evolution occurs within the individual host due to selective pres-
Alternatively, changes in human behavior throughout the seasons have sures imposed by the immune response (Grad et al., 2014). In an infant
been proposed to play a role (Griffiths et al., 2017; Janet et al., 2018; lacking an adaptive immune response, RSV genetic diversity was rela-
Sloan et al., 2017; Welliver Sr., 2007). No animal reservoir is known or tively stagnant but increased rapidly upon reconstitution of the adap-
suspected for RSV, and the reintroduction of RSV in regions with sea- tive response with a bone marrow transplant (Grad et al., 2014). Thus,
sonal transmission cycles is not understood (C. Griffiths et al., 2017) the rapid generation of genetic diversity following the introduction of
(Bilawchuk, Griffiths, Jensen, Elawar, & Marchant, 2017; Collins & a selective pressure, such as the host immune response, will likely trans-
Graham, 2008). late to the emergence of drug resistance following the introduction of a
When novel clades emerge, they have been observed to displace an- new therapeutic. In summary, similar to other RNA viral infections, drug
cestral clades and spread globally. The current prevalent RSV type A resistance will represent a significant hurdle in the efficacy of treatment
clade is the ON1 (Ontario, Canada) clade. The RSV type B BA (Buenos over time.

4
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

3.4. Evolution of RSV in the community toward decreased virulence into a homodecameric ring with each individual subunit interacting
with 7 bases of the RSV genome (Tawar et al., 2009). The RNA itself is
While novel clades spread rapidly, they are not necessarily bound to the decameric ring in a basic groove near the external surface
more virulent. The association of pathogenesis with RSV subtype such that RNA can be readily accessed by the viral RdRp, which is critical
has remained a pressing question since RSV was first divided into since RSV-N has been implicated in viral RNA replication (Grosfeld, Hill,
type A and type B in the 1980s. Conflicting evidence suggests that & Collins, 1995). Interestingly, the function of RSV-N during infection by
RSV subtype does, (Laham et al., 2017; Martinello, Chen, Weibel, RSV extends beyond genomic packaging to antagonism of the host in-
& Kahn, 2002; Walsh, McConnochie, Long, & Hall, 1997) or does nate immune response (Lifland et al., 2012).
not, (J. P. Devincenzo, 2004; Fodha et al., 2007) predict disease se-
verity. The ability to further organize subtypes into genetic clades 4.2.1. Antagonism of the immune response by nucleoprotein
may help address this question. Recently, a pilot study observed There are two mechanisms by which RSV-N modulates the host
that RSV strains which replicate to high titers in patients and immune response. The first mechanism relies on the interaction of
have high replication kinetics in cell culture are genetically similar. RSV-N with a multitude of host proteins, including: retinoic-acid-
These were termed High Titre clades (Elawar et al., 2017). This inducible gene-I (RIG-I), melanoma differentiation-associated
study did not directly examine the severity of illness; however, gene 5 (MDA5), and mitochondrial antiviral signaling protein
an association of RSV viral load and disease severity has been ob- (Lifland et al., 2012) (MAVS; also known as IPS-1, CARDIF, or
served previously (Buckingham, Bush, & Devincenzo, 2000). Thus, VISA (Mogensen, 2009)). RIG-I and MDA5 are cytosolic pattern
identification of high-titer clades may enable forecasting of domi- recognition receptors (PRRs) (Yoneyama et al., 2004; Yoneyama
nant strains with potential to cause severe infections in the subse- et al., 2005). RIG-I detects 5’-triphosphorylated RNA and double-
quent RSV season. stranded RNA (dsRNA), while MDA5 detects only the latter
Our group also showed that newly emerging RSV strains into (Hornung et al., 2006). Following activation by their cognate li-
communities were driven by strains that produced the highest titers gands, both RIG-I and MDA5 signal through the MAVS adaptor
in patients and tissue culture (Elawar et al., 2017). These may be qua- protein to stimulate an interferon (IFN)-β response. Transfection
sispecies that are imported, and so they have emerged with an ability of RSV-N into cells infected with Newcastle Virus (which elicits a
to replicate to a higher titer in a naïve population. However, we also strong IFN-β response) attenuates the IFN-β response via inhibi-
noted the presence of ‘resident clade’ RSV strains that circulate at a tion of RIG-I and MDA5 signaling through sequestration of MAVS
lower titer relative to high-titer clades. While resident clade viruses to inclusion bodies (Lifland et al., 2012). This represents one strat-
produced lower titers in patients and tissue culture, we found that egy by which the RSV-N protein antagonizes the innate immune
they were more infectious in culture. Thus, we propose that there is a response, supporting the argument that it is a bona fide therapeu-
natural evolution of RSV toward strains that linger in the community, tic target.
replicate slower, and have less propensity to cause disease. This evolu- The second known mechanism hinges upon interaction between
tion is driven by the characteristics of resident strains, which are more RSV-N and a protein kinase termed PKR (Clemens et al., 1993). PKR is
infectious but cannot replicate to high titres. Any virus that undergoes produced in response to type I IFNs as a means of inducing an antivi-
fitness adaptation through mutation will likely evolve drug resistance. ral state in cells (Garcia et al., 2006). PKR is activated by cytosolic
This is especially true for direct-acting antivirals where evolutionary dsRNA (reviewed in (Clemens & Elia, 1997)). Briefly, binding to
pressure is placed upon the population of quasispecies; a member of dsRNA results in activation of PKR via phosphorylation of multiple
the population that is resistant to drug pressure will outgrow the serine and threonine residues. Phosphorylated PKR inactivates eu-
susceptible species. karyotic initiation factor 2 α (eIF-2α) via phosphorylation. In the ab-
sence of eIF-2α, cellular translation of mRNA into protein ceases.
4. RSV proteins, their functions and potential as therapeutic targets During RSV infection, there is an accumulation of phosphorylated
PKR; however, eIF-2α is not inactivated. Instead, phosphorylated
4.1. Non-structural protein 1 and non-structural protein 2 PKR is bound by RSV-N (Groskreutz, Babor, Monick, Varga, &
Hunninghake, 2010). Given the localization of RSV-N to inclusion
The RSV non-structural 1 (RSV-NS1) and non-structural 2 (RSV- bodies during RSV infection, binding of PKR by RSV-N likely seques-
NS2) proteins demonstrate a remarkable number of immunomodula- ters PKR into inclusion bodies (Lifland et al., 2012).
tory functions centered around antagonism of the host IFN response A viral protein such as RSV-N that is indispensable for viral replica-
(reviewed in (Barik, 2013)), delaying apoptosis (Bitko et al., 2007), tion is a good therapeutic target, such that suppressing RSV-N activity
and arresting the cell cycle in the G0/G1 phase (Wu et al., 2012). They may release innate immune mechanisms from inhibition while decreas-
are the first proteins to be expressed during infection, underlining the ing RSV replication.
importance of shutting down the IFN response to the virus. Therefore,
therapeutic potential may exist in blocking NS protein-mediated
immune-suppression to augment the innate immune response against 4.3. RSV-P phosphoprotein
RSV. Targeting NS1 and NS2 may decrease viral titres as a prophylactic
or therapeutic setting in infants, similar to what has already been dem- The phosphoprotein in RSV (RSV-P) helps make up the RNA-
onstrated in preclinical studies (Zhang et al., 2005). Recently, studies dependent RNA polymerase complex RSV needs to transcribe and repli-
suggested NS2 could be targeted and inhibited by small molecule cate its genome. Since the polymerase complex is made up of 3 different
drugs (Vasou et al., 2018). Thus, it may be worthwhile to pursue non- proteins, targeting the polymerase complex is an option that has not
structural proteins in antiviral discovery and perhaps in combination been comprehensively explored. The polymerase complex structure
with drugs targeting other RSV proteins. has recently been resolved, thereby opening opportunities for designing
antivirals to target novel RSV sites (Gilman et al., 2019). Furthermore,
4.2. The RSV Genome and Nucleoprotein RSV-P has been implicated as a strong target for antiviral compound de-
sign (Hara et al., 2020). The position RSV-P fits in the polymerase com-
RSV has a single-stranded negative-sense RNA genome of approxi- plex demonstrates that antivirals can inhibit RSV-P function because it
mately 15.2kb that encodes 10 genes and 11 proteins (Collins & would have access to a binding pocket. RSV-P is essential for viral repli-
Melero, 2011). The RNA genome is tightly bound to RSV nucleoprotein cation and transcription, therefore, halting the P protein function will
(RSV-N) in a helical ribonucleoprotein (RNP) complex. RSV-N arranges halt RSV-L activity as well.

5
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

4.4. RSV-M Matrix protein 4.5.3. RSV-SH small hydrophobic protein


Like RSV-G, RSV-SH is not essential for RSV replication in cell culture
The matrix protein (RSV-M) is a structural protein which lines the (Bukreyev, Whitehead, Murphy, & Collins, 1997; Karron et al., 1997),
inside of the viral envelope and associates with the RNP via RSV-M2-1 and recombinant RSV strains lacking RSV-SH were only slightly attenu-
(Kiss et al., 2014). RSV-M does not have characterized antigenic regions ated in mice (Bukreyev et al., 1997). In chimpanzees, RSV lacking RSV-
that are accessible to the host immune system for neutralization; there- SH were attenuated in the lower, but not upper, respiratory tract
fore, it is not currently a suitable RSV protein target in a vaccine or (Whitehead et al., 1999). While the role of RSV-SH in RSV replication
antiviral. is not well characterized, these observations have precipitated investi-
gations of RSV-SH deletion vaccine candidates (Karron et al., 2005). Fur-
thermore however, RSV-SH does not physically protrude as far as RSV-F
4.5. Envelope proteins: RSV-F fusion protein, RSV-G glycoprotein, and RSV-
and RSV-G, therefore it is not an ideal neutralization target.
SH small hydrophobic protein
4.6. RSV-M2
The RSV viral envelope proteins include fusion protein (RSV-F), gly-
coprotein (RSV-G), and small hydrophobic protein (RSV-SH). RSV entry
The RSV polymerase consists of a complex formed by RSV-L, RSV-P,
is mediated by a complex interaction between RSV-F and RSV-G enve-
and RSV-M2-1 to transcribe and replicate its RNA genome (Reviewed
lope proteins and host cell proteins (reviewed by (C. Griffiths et al.,
in (Cowton, McGivern, & Fearns, 2006)). RSV-M2-1 is primarily in-
2017)). Recently, a paper by Griffiths et al. showed that RSV first binds
volved in the complex as transcription terminator. There are reports of
to a main receptor, insulin-like growth factor-1 receptor (IGF1R) (C. D.
RSV-M2-1 interacting substantially with RSV-P in the complex
Griffiths et al., 2020). This binding to IGF1R triggers trafficking of the
(Selvaraj et al., 2018). Therefore, RSV-M2-1 is an essential part of the
RSV coreceptor, nucleolin, to RSV virions waiting at the cell surface, trig-
RSV lifecycle and can be targeted with an antiviral either by directly
gering fusion and viral entry (C. D. Griffiths et al., 2020). Other host cell
targeting the protein or the complex that the protein folds into. Further-
proteins have been proposed as receptors, including CX3 chemokine re-
more, recent studies have suggested that M2-1 has a role in down-
ceptor 1 (CX3CR1) (S. M. Johnson et al., 2015), epidermal growth factor
stream transcription to translation (Bouillier et al., 2019). The
receptor (EGFR) (Krzyzaniak, Zumstein, Gerez, Picotti, & Helenius,
importance of M2-1 in transcription and translation of RSV makes M2-
2013; Lingemann et al., 2019), annexin II (Malhotra et al., 2003),
1 a more attractive therapeutic target.
calcium-dependent lectins, Toll-like receptor 4 (TLR4) (Marchant
et al., 2010), intercellular adhesion molecule 1 (ICAM-1) (Behera et al.,
4.7. RNA-dependent RNA polymerase
2001), nucleolin (Tayyari et al., 2011), and heparan sulfate proteogly-
cans (HSPGs) (Feldman, Audet, & Beeler, 2000). The functions of these
The RSV RdRp is encoded by the RSV-L gene and is packaged within
proteins include tethering RSV to the cell surface and triggering fusion
the virion in conjunction with the genome. This protein, in complex
(C. Griffiths et al., 2017; Griffiths et al., 2020).
with RSV-P, catalyses transcription and replication of the RSV genome
(Figs. 1 and 2). The polymerase complex of a virus is critical because it
4.5.1. RSV-F fusion protein produces the mRNA from which viral proteins are made and it replicates
RSV-F has been the focus point for many pharmaceutical companies the genome to be packaged into progeny viruses. These activities are
as a vaccine candidate and as the target of prophylactic antibodies like crucial for an infection to happen and so the polymerase makes for a
palivizumab. This is because it is the principle neutralizing determinant strong therapeutic target.
of RSV, and necessary for viral entry and infection (C. Griffiths et al.,
2017). RSV-F triggers the fusion of virion and host cell membranes 4.7.1. The structure of the RSV polymerase and implications for therapeutic
that allow delivery of the virus capsid contents into the host cell. Before development
interaction with the host cell, it exists as a trimer in a pre-fusion confor- Even though the RSV-L protein is the core polymerase enzyme, the
mation (McLellan et al., 2013). RSV-F binds to the RSV receptor IGF1R RSV-L and -P proteins are the minimal constituents of the RSV polymer-
that triggers focal trafficking of the coreceptor, nucleolin, to RSV virions ase that are required for transcription and replication activity (Fearns &
at the cell’s surface (C. D. Griffiths et al., 2020). Host-cell nucleolin was Collins, 1999; Tchesnokov, Raeisimakiani, Ngure, Marchant, & Gotte,
first identified as a candidate coreceptor for RSV-F via a virus overlay 2018). Recently, the structure of the RSV-L and -P polymerase complex
protein binding assay and is necessary for optimal RSV entry (Tayyari has been elucidated to 3.2-Å resolution by cryoelectron microscopy
et al., 2011). We hypothesize that NCL interaction with RSV-F triggers (Gilman et al., 2019) (Fig. 3). This provides a high-resolution picture
the fusion of RSV with the host cell that is driven by a conformational of the RSV-L and -P core RdRp complex that on its own has in vitro
shift in multiple spring-loaded, metastable, RSV-F proteins (Griffiths RNA-polymerase activity. The RSV-P protein encompasses the RSV-L
et al., 2017). Until recently, the more stable RSV-F in its post-fusion core catalytic protein in a “tentacular” formation in a 4:1 stoichiometry,
state was predominantly used in vaccine development. However, after meaning that the four RSV-P proteins emanate from the core protein
multiple years of clinical trial failure, we know the RSV-F post-fusion structure like tentacles and each RSV-P molecule on the RSV-L takes
conformation does not confer protective antibodies against infectious on a distinct conformation (Fig. 3). There are two primary functions of
RSV particles (Mazur et al., 2018). RSV-L, transcription of individual mRNA transcripts and genome repli-
cation, the regulation of which was only elucidated recently (Noton,
4.5.2. RSV-G glycoprotein Deflube, Tremaglio, & Fearns, 2012). The structure of the polymerase
Depending on the type of immortalized cell line, RSV-G is dispens- provides a model from which new compounds can be made and ex-
able for infectivity in cell culture (Karron et al., 1997). However, interac- plains the generation of resistance to antiviral drugs.
tion between RSV-G and the cellular membrane protein CX3CR1 is The RSV-L/-P complex can initiate transcription or replication and
necessary for optimal infection of in vivo models (S. M. Johnson et al., produce short transcripts up to 200 nucleotides in length (Fearns &
2015). Soluble RSV-G is secreted by virions to serve as an immune and Collins, 1999; Noton et al., 2012). Expression of RSV-L, -P and -N is
antibody decoy (Bukreyev, Yang, & Collins, 2012). RSV-G is also the sufficient for minigenome replication, indicating that concurrent encap-
most variable protein expressed in the RSV genome and, as such, has sidation of the growing genome by RSV-N is required for optimal RdRp
been used to characterize RSV strains. The variability of this protein replicase processivity (Grosfeld et al., 1995). Conversely, transcriptase
makes it unattractive as a vaccine target because the vaccine would activity by RSV RdRp requires the RSV-M2-1 anti-termination factor
have to be adapted frequently. (Collins, Hill, Cristina, & Grosfeld, 1996). As a result, a full complement

6
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Fig. 1. Experimental therapeutic strategies target different points in the RSV replication cycle. The RSV replication cycle from receptor binding, entry, to genome replication, viral budding
and spread to adjacent cells are shown. Drugs targeting each of the steps or RSV proteins are shown in parentheses.

of RSV-L, -P, -M2-1, and -N is required to produce full infectious RSV M2-1 (Asenjo, Calvo, & Villanueva, 2006). Thus, blocking these interac-
from a cDNA antigenome (Collins et al., 1995). tions or the phosphorylation of RSV-P could lead to new classes of
therapeutics.

4.7.2. Drugs targeting the RNA-dependent RNA polymerase


RSV-L is the catalytic subunit of RSV RdRp, responsible for polymer- 5. An overview of RSV therapeutic development
izing genome and transcript chain elongation (Poch, Sauvaget, Delarue,
& Tordo, 1989), polyadenylation (reviewed by (Cowton et al., 2006)), 5.1. Development of RSV antivirals
and 5’ capping (Barik, 1993). The RSV RdRp, as with all RNA viruses, is
central to viral replication and is thus a logical therapeutic target. Millions of compounds have been screened for antiviral activity
The RSV-P protein is also a necessary subunit of RdRp along with against RSV in high throughput platforms and a small handful of these
RSV-L. Phosphorylation of RSV-P at serine-232 is required for chain compounds have entered clinical trials (Table 1). Despite RSV antiviral
elongation (Dupuy, Dobson, Bitko, & Barik, 1999), and phosphorylation drug discovery spanning decades, no efficacious antivirals have entered
of RSV-P at threonine-108 enables interaction of the RdRp with RSV- the market. The absence of efficacious RSV antivirals in the clinic

Fig. 2. The structure and configuration of the RSV polymerase consisting of RSV-L and P proteins.

7
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Fig. 3. Structures of RSV fusion and entry inhibitors.

highlights the need for creative efforts to identify novel chemical enti- activity, we will discuss those compounds with the most promise or
ties with antiviral activity against RSV. that have failed clinical trials.
The diverse group of antiviral compounds against RSV that have en-
tered clinical trials includes nucleoside analogues and non-nucleoside 5.2. Fusion inhibitors
analogues which target the RSV polymerase complex, an siRNA which
targets RSV-N, and small molecules which interfere with viral fusion 5.2.1. GS-5806
(Table 1) (Broadbent, Groves, Shields, & Power, 2015). Alternatively, GS-5806, also referred to as presatovir, is an RSV-F inhibitor (Perron
therapeutics such as Danirixin, an oral small molecule drug that inhibits et al., 2015). Gilead Sciences employed a high-throughput antiviral
the CXCR2 receptor, seek to mitigate the damage due to inflammation screening campaign in their efforts to discover and optimize hits against
which occurs during RSV infection (Broadbent et al., 2015). Rather RSV (Mackman et al., 2015a, 2015b). A result was the discovery of a
than describe the myriad of compounds that have in vitro antiviral structurally distinct class of RSV fusion inhibitors bearing a pyrazolo

8
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

[1,5-a]-pyrimidin-2-yl moiety. A phenotypic screening of ~400,000 Phe140 and Phe488 located in the RSV-F fusion peptide and HRB
compounds, using human epithelial type-2 (Hep-2) cells to assess CPE domains (Fig. 4A) (Douglas et al., 2005). The fusion peptide and
(cytopathic effects) by RSV A2 virus identified a racemic hit 1 with an HRB domains are located in the N-terminus and C-terminus of RSV-F,
in vitro EC50 = 202 nM (Fig. 4A). respectively, that undergo conformational rearrangements during the
GS-5806 functions by locking RSV-F in the pre-fusion conformation fusion process.
and preventing the subsequent conformational change required for fu- Moreover, the positively charged piperidine nitrogen of JNJ-
sion with the host cell membrane, thereby preventing entry of the virus 2408068 reaches down to the negatively charged Asp 486 in the bind-
(Samuel et al., 2015). In healthy adult volunteers inoculated with RSV, ing pocket (Fig. 4A). This observation showed that the inhibitor acted
GS-5806 reduced symptom scores and viral replication (DeVincenzo as an antagonist, with both hydrophobic and electrostatic interactions,
et al., 2014; Mackman et al., 2015a, 2015b) and in a direct comparison, to prevent RSV-F rearrangement and stabilize the RSV-F pre-fusion
GS-5806 offered a greater therapeutic index than VP-14637, TMC- conformation. However, JNJ-2408068 was found to have a long tissue
353121, BMS-433771, BI-D, YM-53403, RSV604, and ribavirin (Perron retention time (half-life in the lung is about 153 h), which raised
et al., 2015). Mutations that confer resistance to GS-5806 map to the safety concerns and led to a decision to halt further development
fusion peptide (L138F and F140L) or heptad repeat B (HRB) (F488L/S) (Sun, Pan, Jiang, & Lu, 2013).
regions of RSV-F. GS-5806 resistant mutants also exhibited resistance
to other inhibitors of RSV-F such as VP-14637. Similarly, mutations 5.2.4. JNJ-53718678
within the RSV-F fusion peptide and HRB domains that confer resistance JNJ-53718678 (JNJ-678) is another potent RSV-F protein inhibitor
to other fusion inhibitors reduced the efficacy of GS-5806 (Stray et al., developed by Johnson & Johnson (Fig. 3D). Isothermal titration calorim-
2020). Interestingly, GS-5806 resistant mutants are still susceptible to etry (ITC) has been used to assess the binding of JNJ-678 to the RSV
palivizumab and ribavirin and the converse is true as well. This means fusion protein, involving both pre- and post-fusion F protein conforma-
that should resistance to one treatment option arise, an efficacious ther- tions (Roymans et al., 2017a, 2017b). Results showed that the com-
apeutic in the form of either palivizumab or GS-5806 will remain. In pound bound tightly to the pre-fusion conformation of RSV-F. JNJ-678
April of 2017, GS-5806 completed phase 2b clinical trials in adults hos- exhibited exceptionally potent in vitro activity against RSV A2 strain
pitalized with RSV, however, results have not been released with an EC50 of 480 pM. Investigation of the binding mode and
(NCT02135614). structure-activity relationship of this compound compared to other re-
Recently, in a phase 2 clinical trial, presatovir was unsuccessful at ported fusion inhibitors showed that the same pocket was being
improving virological and clinical outcomes (Marty et al., 2019). Fur- targeted. The replacement of the benzimidazole, which has been re-
thermore, the need for supplemental oxygen and mechanical ventila- ported to be essential for the activity of JNJ-2408068, with an indole
tion were not different between the placebo and presatovir treatment moiety, retained the antiviral activity. The chloroindole moiety of JNJ-
groups. As with many antivirals that target virus entry, one must con- 678 was shown by X-ray crystallographic analysis to also be involved
sider that failure of the trial may have been due to treatment delay dur- in a π-π stacking interaction with Phe488 and Phe140 (Fig. 4B). In addi-
ing the course of patients’ infection. We would suggest that entry tion, once the compound was bound to the F protein, the chlorine atoms
inhibitor such as presatovir could be more useful as prophylactic established a halogen bond with the backbone of Thr397 in a
drugs that would be administered during RSV seasons to those at risk sub-pocket formed by amino acids Asp489, Asp486, Thr397 and
from infection. Phe137. These results confirm that members of this class of compounds
share similar mechanisms of binding. Several studies have reported the
5.2.2. MDT-637 use of an oral treatment against RSV infections involving the use of
MDT-637, formerly known as VP-14637 (Fig. 3B), is a substituted JNJ-53718678. These studies supported the efficacy and safety of this
bis-tetrazole-benzhydryl phenol, which has been reported to inhibit compound in inhibiting viral replication and reducing disease severity
RSV cell entry (Fig. 1). Evaluation of MDT-637 against RSV clinical iso- in infected adults (Israel et al., 2016; Stevens et al., 2018).
lates in cotton rats showed that the compound had a hundred- to Like GS-5806, JNJ-678 stabilizes the pre-fusion conformation of
thousand-fold greater potency than ribavirin (Kim et al., 2017a, RSV-F to inhibit viral fusion (Roymans et al., 2017a, 2017b). Due to
2017b). Moreover, MDT-637 showed broad in vitro antiviral activity the similar mechanisms of action, it is unsurprising that mutations
on clinical strains of different RSV genotypes. Douglas and coworkers that confer resistance to JNJ-678 map to regions in the fusion peptide
used an RSV-induced cell fusion assay to confirm that MDT-637 acts (L141W) or HRB (D489Y) domains of RSV-F. In HeLa cells, the average
by inhibiting viral fusion (Douglas et al., 2003; Douglas et al., 2005). concentration at which viral infection was reduced by 50% was 0.46
Drug-resistant variants have been selected and genotypic analysis of nM. Additionally, JNJ-678 was observed to reduce viral titer and
the resistant viruses revealed various mutations in the F protein. Struc- RSV-induced lung inflammation, two major contributors to RSV dis-
tural analysis of the F protein revealed a hydrophobic cavity formed by ease severity, in mouse, cotton rat, and lamb models. As was observed
six amino acids. This pocket can accommodate two hydrophobic resi- for GS-5806, treatment with JNJ-678 also reduced viral load and
dues (F483 and F488) during the formation of the six-helix bundle symptom scores in healthy adult volunteers experimentally infected
(6HB) (Roymans et al., 2010). Identifying the mechanism of action of with RSV (Stevens et al., 2018).
this compound opened the door for a detailed understanding of the
mechanism surrounding RSV fusion protein inhibition and hence the 5.2.5. TMC-353121
progress toward more potent compounds with different scaffolds. Molecular modeling and pharmacokinetics studies were carried out
by Johnson & Johnson to identify critical substructures responsible for
5.2.3. JNJ-2408068 the long tissue retention time of JNJ-2408068. As a result, TMC-
JNJ-2408068 is a benzimidazole derivative that has been reported to 353121, a morpholinopropylaminobenzimidazole RSV fusion inhibitor
have potent RSV fusion inhibitory activity (Fig. 3C). Battles and (Fig. 3E), has been developed as an improved derivative of JNJ-
coworkers assessed the antiviral activity of JNJ-2408068 in different cel- 2408068 (Bonfanti et al., 2008). TMC-353121 retained the antiviral ac-
lular RSV assays (Battles et al., 2016). JNJ-2408068 binds to a three-fold- tivity against RSV-F that was seen in the earlier compounds, with a
symmetric pocket in the metastable pre-fusion conformation of RSV-F. pEC50= 9.9 nM and a half-life around 14h in lung tissues. Preclinical
This binding stabilizes the RSV-F pre-fusion conformation by tethering studies in cotton rats and African monkeys revealed a dose-dependent
two regions that undergo a structural arrangement which facilitates antiviral activity of TMC-353121 varying from one log10 reduction of
membrane fusion (Battles et al., 2016). Binding of JNJ-2408068 has re- viral load to complete inhibition of RSV replication (Ispas et al., 2015).
vealed a π-π stacking interaction with the aromatic side chains of TMC-353121 has been co-crystallized with a construct of the RSV-F

9
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Fig. 4. Mechanisms of inhibition of two RSV-F entry inhibitors. A, JNJ-2408068 bound to the RSV-F envelope glycoprotein of RSV with important amino acids involved in the binding with
hydrogen bonds shown in dashed red lines, ion pairing shown in magenta and π-cation interaction in cyan (PDB accession = 5EA3). B, Ligand interaction diagram of JNJ-678 indicating the
key binding interactions of the compound with F protein (π-π stacking represented by circled green arrows, Hydrogen and halogen bonds by green arrows). Adapted with permission from
(Isreal S. et al. 2016). Copyright 2017 Springer.

protein (Roymans et al., 2010). Analysis of the binding mode of TMC- interactions between RdRp constituents are mechanisms by which anti-
353121 within the prefusion F protein conformation has shown that virals may inhibit RSV replication (Table 1).
this binding did not eliminate all the interactions between the heptad
repeats, known as HR1 and HR2, of the F protein, which can be 5.3.2. ALN-RSV01
envisioned as a zipper that can close completely in the absence of the in- Host cell-based RNA interference has been explored as a potential
hibitor. However, the binding of TMC-353121 and other benzimidazole RSV antiviral. The RNA interference response in host cells is triggered
RSV-F protein inhibitors, such as JNJ-2408068, induced a distorted form by siRNA loaded onto argonaute proteins in mammals to form the
of the RSV fusion protein trimer which rendered the zipper only par- RNA-induced Silencing complex (RISC), an endoribonuclease that
tially closed. This distortion has been proved to be sufficient to potently degrades sequence specific mRNA transcripts and is responsible for
inhibit RSV viral replication (Douglas et al., 2005; Roymans et al., 2010). translation repression (Lam, Chow, Zhang, & Leung, 2015).
ALN-RSV01 is the first antiviral siRNA approved for testing in
5.2.6. RV521 clinical trials that targets RSV-N transcripts. In a phase 2 clinical
A fusion inhibitor called RV521 targets the RSV-F protein to inhibit trial using healthy adult volunteers inoculated with RSV, ALN-RSV01
the fusion steps of viral entry (Fig. 1). It has a favourable in vitro 50% in- given prophylactically reduced the number of volunteers infected (J. P.
hibitory concentration of about 1 nM. This drug candidate, developed by DeVincenzo et al., 2010). In a separate phase 2 clinical trial, ALN-
ReViral in the United Kingdom, has recently undergone a phase 2a, dou- RSV01 was administered to adult lung transplant recipients with RSV
ble blind, placebo controlled human challenge study of antiviral efficacy infections. While ALN-RSV01 reduced progression of bronchiolitis
(DeVincenzo et al., 2020). Participants were treated with the experi- obliterans syndrome, no reduction was observed in symptom severity,
mental medication five days after challenge with RSV or when infection viral shedding, or viral load (Gottlieb et al., 2016). ALN-RSV01 com-
was detected. Both viral load and disease severity were reduced signif- pleted phase 2b clinical trials in May 2012 but the results have not yet
icantly, and recovery of viral load back to detection threshold was faster been posted (NCT01065935).
in those treated with both 350 mg and 200 mg doses of the inhibitor.
The reported treatment emergent adverse events were mild and tran- 5.4. Nucleoside and non-nucleoside inhibitors of RSV-L
sient. In summary, the RSV research and drug development community
will be watching the development of this compound in further clinical Nucleoside analogues have long been a cornerstone of antiviral re-
trials in the years to come. search. In 1963, the deoxyuridine analogue idoxuridine, used to treat
HSV eye infections, became the first licensed antiviral. In 1985, azido-
thymidine (AZT) became the first nucleoside reverse transcriptase in-
5.3. Replication inhibitors hibitor and drug licensed to treat HIV infection. Most recently, the
hepatitis C virus (HCV) antiviral nucleotide analogue, Sofosbuvir, was li-
5.3.1. RSV604 censed. HCV is a blood borne pathogen in which chronic infection can
RSV-N nucleoprotein of RSV plays indispensable roles in RNA tran- lead to a lifelong disease. Sofosbuvir offers the first curative treatment
scription and replication given that it is an essential component of the for HCV infection (De Clercq & Li, 2016). Of the nucleoside analogues
RSV polymerase complex. RSV604 is a candidate drug that targets the our group has tested expermentally, modest antiviral activity against
RSV-N protein. One mechanism of action proposed for RSV604 involves RSV was noted for cytarabine (Tchesnokov et al., 2018), a chemothera-
direct binding of RSV-N by RSV604 to prevent interaction of viral and peutic cytosine analogue (Lichtman, 2013).
host accessory proteins required for optimal viral transcription, in vivo To date, 32 compounds derived from nucleosides have been ap-
(Challa et al., 2015). It is not unexpected that interference with a variety proved for clinical use against HIV, HBV, HCV, HSV, VZV, HCMV, and
of target proteins inhibits RSV RdRp, given the essential nature of mul- IAV (De Clercq & Li, 2016; De Clercq & Schinazi, 2016). Nucleoside ana-
tiple viral and cellular proteins in forming a functional RSV RdRp com- logues typically take advantage of the reliance of viruses on viral poly-
plex. Specifically, cellular proteins, including heat shock protein 90 merase complexes for replication. Interfering with these polymerase
(HSP90), are necessary for optimal RdRp processivity (Munday et al., complexes can result in chain termination or catastrophic mutagenesis
2015). Antagonism of any RdRp enzymatic process or antagonism of through non-complementary base pairing (De Clercq & Li, 2016).

10
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Nucleoside analogues have previously been investigated for antiviral ac- 2005). In the clinic, ribavirin plus pegylated IFN was the standard of
tivity against RSV. Two nucleoside analogues which inhibit RSV replica- care for HCV infection for many years until this treatment was surpassed
tion, ribavirin and lumicitabine, have been described (DeVincenzo et al., by curative direct-acting antivirals (Feld & Liang, 2005; Houghton,
2015). Ribavirin is a potent inhibitor of RSV replication in cell culture 2014) due to its toxicity and limited efficacy, and has since fallen out
while lumicitabine has demonstrated efficacy in adult human volun- of favour in the clinic.
teers but it is currently halted after phase 2 clinical trials (Table 1) Ribavirin is effective against RSV in cell culture, with reported effec-
(DeVincenzo et al., 2015). tive concentration 50% (EC50) values of 6.3 to 28.38 μM (Chapman et al.,
2007; Kim et al., 2017a, 2017b; Noah et al., 2010; Sudo et al., 2005;
5.4.1. Nucleoside analogues as RSV antivirals Tiong-Yip et al., 2014). Given the efficacy of ribavirin in cell culture, it
Nucleoside analogues have proven effective as a cure for chronic in- has been widely used to validate screening assays, provide a point of ref-
fections like hepatitis-C and controlling HIV infection and transmission. erence for compounds identified in these screens, and offers a standard
However, they are not without notable side-effects including through which different screening assays can be compared.
lipodystrophy, headaches, bone loss, and kidney, liver, and pancreatic
damage. Therefore, it is important to determine the side-effects of nu- 5.4.3. PC786
cleoside analogues in children. Subsequently, we must then determine PC786 contrasts lumicitabine as it is not a nucleoside analogue, how-
whether the benefits of treating an acute infection like RSV with a nucle- ever it does target RSV-L for inhibition (Coates et al., 2017). During serial
oside analogue outweigh any undesirable side-effects in infants and passaging of PC786, escape mutants (RSV-L Y1631H or Y1631L)
children. One must remember that, with respect to pharmacology, in- emerged at passage three. As PC786 is not a nucleoside analogue, it is
fants and children can respond quite differently to drugs compared to not surprising that the locations of mutations differed between
adults. However, nucleoside analogues like zidovudine (azidothymi- lumicitabine and PC786. The RSV-L Y1631H mutation also conferred re-
dine) have proven safe and effective at preventing vertical HIV trans- sistance to PC786 parent compounds AZ-27 and YM-53403 (Noton
mission in newborns. Whether they will be effective and safe in the et al., 2015; Sudo et al., 2005). While the mechanisms of inhibition dif-
treatment of acute infectious disease in infants and children remains fer, lumicitabine and PC786 both target RSV-L. It will be interesting to
unclear. learn whether similar compounds target RSV-L or whether they can tar-
get another component of the RdRp complex (Noton et al., 2015; Plant
5.4.2. Ribavirin et al., 2015). PC786 development has been halted after phase 2 clinical
In 1972, synthesis of 1,2,4-triazole nucleosides in the pursuit of trials (NCT03382431).
antiviral compounds led to the discovery of ribavirin (1-β-D-
ribofuranosyl-1,2,4-triazole-3-carboxamide) (Fig. 5A). In this initial 5.4.4. ALS-8112
study, in vitro antiviral activity was described for a broad array of Wang et al. (Alios BioPharma) developed a series of novel sugar-
many different viruses: VSV, PIV-3, RV, herpes simplex virus type 1 modified nucleosides that inhibited RSV polymerase in a cell-based
(HSV1), herpes simplex virus type 2 (HSV2), pseudorabies virus, mu- assay (Fig. 5B) (Wang et al., 2015). The modified nucleoside analogues
rine cytomegalovirus (mCMV), vaccinia virus, myxoma virus, adenovi- were converted to their 5’-triphosphate counterparts and tested against
rus (AdV), parainfluenza virus type 1 (PIV-1), influenza A virus (IAV), RSV isolates for their activity against the viral polymerase. Four′-azido-
influenza B virus (IBV), coxsackievirus, and poliovirus (Witkowski, 2′-deoxy-2′,2′-difluorocytidine was found to be a potent inhibitor of
Robins, Sidwell, & Simon, 1972). Ribavirin was later found to also have RSV-induced cytopathic effect with an EC50 of 1 μM. Similar analogues
antiviral activity against RSV, HCV, HIV, Sendai virus, MeV, and Newcas- have previously been reported to inhibit HCV polymerase (D. B. Smith
tle virus (Chapman et al., 2007; Hong & Cameron, 2002; Hruska, et al., 2009). However, due to reports of potential safety concerns
Bernstein, Douglas Jr., & Hall, 1980; Huffman et al., 1973; Kim et al., caused by 4’-azidocytidine prodrugs in phase 2 clinical trials, the search
2017a, 2017b; Scholtissek, 1976; Sidwell et al., 1972; Sudo et al., for another analogue(s) with specific RSV inhibitory activity and safety

Fig. 5. Structures of RSV-L polymerase inhibitors.

11
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

margin was necessary (Nelson et al., 2012). Optimization processes led 2018; Zhu et al., 2017). This new monoclonal antibody will be an im-
to the identification of novel nucleoside analogues, including the 4’- provement over palivizumab in that it has a longer half-life of about
chloromethylene compound ALS-8112 (Fig. 5B), with improved po- 70 days (as opposed to palivizumab’s 20 days), requiring only one intra-
tency and selectivity against RSV polymerase. The RSV-L protein was muscular injection per season. Furthermore, whereas palivizumab
identified as the molecular target of ALS-8112 by the characterization binds to site II on the side of pre- and post-fusion RSV-F protein,
of drug resistance-associated mutations in the L gene (Deval et al., nirsevimab neutralises RSV by binding to the prefusion RSV-F glycopro-
2015a, 2015b). The 5’-triphosphate form of ALS-8112 (ALS-8112-TP) tein at the more sensitive neutralisation site ∅, located at the apex of the
caused chain termination of RNA synthesis and inhibition of RSV viral RSV-F glycoprotein. It has completed a phase 1 trial in adults (Griffin
polymerization activity in enzymatic assays. ALS-8112-TP did not et al., 2017) and a phase 1/2a trial in infants (Domachowske et al.,
show any inhibitory activity against host or HCV polymerases, proving 2018). The phase 2b trial was conducted on infants between 29 weeks
its specificity against RSV polymerase. 0 days and 34 weeks 6 days gestational age, and those who were enter-
ing their first full RSV season at the time of screening. The efficacy and
5.4.5. Lumicitabine (ALS-8176) safety data for the infant trial were not available at the time of writing
Poor oral bioavailability of ALS-8112 necessitated the use of a this review.
prodrug strategy which resulted in the development of ALS-8176, also
named lumicitabine, which is a 3’,5’-di-O-isobutyryl-2’-fluorocytidine 6. Vaccines
prodrug of ALS-8112 (Wang et al., 2015) (Fig. 5C). Following entry
into the host cell, ALS-8176 is triphosphorylated and competes with cy- A fundamental challenge to the development of RSV vaccines is that
tidine triphosphate for access to RSV RdRp. Lumicitabine binds RSV natural infection with RSV in immunocompetent adults offers very little
RdRp near the active site and results in chain termination. The parent protection (Hall, Walsh, Long, & Schnabel, 1991). Any vaccine will need
compound of lumicitabine also inhibits PIV-3 and VSV RdRp in vitro. to elicit a stronger immune response than natural infection and do so in
Interestingly, slight structural modifications to lumicitabine conferred the absence of deleterious side effects. RSV is able to either evade or
activity against HCV RdRp, a positive sense ssRNA virus (Deval et al., suppress memory IgA B cells. Measles virus infection is similar in this re-
2015a, 2015b). Serial passaging of RSV in the presence of increasing gard, although that was overcome with a replication competent live-
concentrations of lumicitabine yielded resistant mutants. These mu- attenuated vaccine. So far, all attenuated RSV vaccines strategies have
tants possessed 4 amino acid substitution mutations, each mapping to failed clinical trials.
the third conserved region of RSV RdRp within motif B which lies near
the catalytic active site. These mutations included RSV-L M628L,
A789V, L795I, and I796V. 6.1. The first failed RSV vaccine
There were promising results from lumicitabine clinical trials where
adult volunteers were experimentally challenged with RSV. Participants Shortly after the discovery of RSV, Kim et al. in the 1960s attempted
were inoculated with RSV and began treatment with lumicitabine after to develop a vaccine through formalin inactivation of RSV (Kim et al.,
infection was confirmed by RT-PCR. This scenario resembled a more re- 1969). This approach had been successful for Jonas Salk, who had re-
alistic scenario where infection would be confirmed prior to treatment. cently developed the world’s first vaccine against poliovirus (Baicus,
Here, it was found that lumicitabine reduced viral load and duration of 2012). A clinical trial was completed with a group of 31 infants less
infection (J. P. DeVincenzo et al., 2015). It should be noted that this is than one year of age. These infants were primarily from African
the furthest RSV-L inhibitors have progressed in clinical trials to date. American and families of low socioeconomic status (Kim et al., 1969).
Unfortunately, clinical trials studying lumicitabine were recently can- The increase in neutralizing antibodies following immunization was
celled (late 2018). An ascending dose study in infants hospitalized limited to a 4-fold increase in 43% of immunized infants and this rise
with RSV infection showed treatment-emergent neutrophil abnormali- in neutralizing antibodies afforded no protection from natural infection
ties. This result led to a cessation of further clinical trials with during the subsequent RSV season. Instead, enhanced respiratory dis-
lumicitabine (Beigel et al., 2019). ease (ERD) was observed upon natural infection. 80% of RSV-infected
vaccinated infants required hospitalization while only 5% of the control
5.5. Biologicals; therapeutic antibodies group required hospitalization. In the vaccinated group the duration of
hospitalization was significantly increased, the rate of serious complica-
Biologicals have been extensively investigated for potential thera- tions increased, and two infants died. Understandably, this tragedy
peutic use against RSV infection. Palivizumab was originally licensed stunted RSV vaccine development campaigns.
for prophylactic treatment against RSV in high-risk infants and failed
to demonstrate efficacy as a therapeutic intervention during ongoing in- 6.2. Enhanced respiratory disease
fection ("Palivizumab, a Humanized Respiratory Syncytial Virus Mono-
clonal Antibody, Reduces Hospitalization From Respiratory Syncytial Over the ensuing decades, mechanisms of ERD have been proposed.
Virus Infection in High-risk Infants," 1998; Saez-Llorens, et al., 2004). One explanation for ERD is based on the pathogenic deposition of
Motavizumab is a humanized monoclonal antibody which was derived antibody-antigen immune complexes (IC) in the lungs. While IC forma-
from palivizumab. Motavizumab failed to gain regulatory approval for tion can be beneficial towards virus neutralization during viremia, the
prophylactic or therapeutic treatment against RSV infection after hyper- deposition of ICs into tissue drives the pathogenesis of a variety of infec-
sensitivity reactions were observed and no improved efficacy versus tious and autoimmune diseases (Jancar & Sanchez Crespo, 2005). In
palivizumab was observed. The most recent attempt at producing a bi- mice vaccinated with formalin-inactivated RSV (FI-RSV), ERD (mea-
ologic intervention was ALX-0171; heavy chain antibody fragments sured as an increase in airway hyper-response) and IC formation in
generated in llamas were genetically linked via glycine-serine segments the lungs was observed. ERD was absent in complement-deficient
to form trimeric nanobodies (Detalle et al., 2016; Hultberg et al., 2011). mice and in B cell-deficient mice (which lack antibodies required for
Results in cotton rats were promising, with 2-log reductions in viral ti- IC formation) (Polack et al., 2002). The authors concluded that IC depo-
ters observed. A phase 1 clinical trial is ongoing (NCT02979431). sition in the lungs and subsequent complement activation drives ERD
following vaccination with FI-RSV. This study relies on airway hyper-
5.5.1. Nirsevimab MEDI 8897 responsiveness acting as an appropriate measure of ERD, which is only
A new prophylactic monoclonal called nirsevimab (MEDI 8897) is modestly appropriate. However, the authors bolster their conclusions
being developed by MedImmune, Astra Zeneca (Domachowske et al., by noting extensive complement activation in lung biopsies obtained

12
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

from the two infants who succumbed to ERD in the vaccine trial by Kim virus candidates including MEDI-ΔM2-2, which lacks the RSV-M2-2
et al. (Kim et al., 1969). gene (NCT01459198), or a genetically stabilized version of MEDI-599
Building on these results, Delgado et al. observed that FI-RSV elicited (NCT01852266). Lastly, results are not available for RSV ΔNS2 Δ1313
non-protective low-avidity antibodies (Delgado et al., 2009). Insuffi- I1314L which lacks RSV-NS2, has a deletion of the 1313 codon in the
cient toll-like receptor (TLR) stimulation in plasmacytoid dendritic RSV-L protein, and a temperature sensitive mutation introduced
cells (pDCs) resulted in reduced CD4+ T lymphocyte proliferation and through an I1314L mutation in the RSV-L protein (NCT01893554).
activation. Resultantly, B cells failed to form germinal centers and un-
dergo the affinity maturation process necessary for increasing antibody 6.5. Subunit vaccines in clinical trials
avidity. In this study, complementing FI-RSV vaccination with UV-
inactivated RSV was sufficient to induce a protective antibody response. Novavax has led the development of nanoparticle-based RSV vac-
cines. Nanoparticles were created through infection of Sf9 insect cells
6.3. Lessons learned about enhanced respiratory disease with a recombinant baculovirus containing a modified RSV-F gene.
RSV-F nanoparticles consist of multiple peptide homotrimers joined
In the wake of the failed vaccine attempt by Kim et al., candidate vac- via a micelle core (G. Smith et al., 2012). The RSV-F on these nanoparti-
cines are now carefully monitored for ERD in animal models (the cotton cles most likely consist of the post-fusion form of RSV-F that is a weaker
rat model is especially useful for detecting ERD) prior to clinical trials. neutralization determinant than the stabilized prefusion form of RSV-F
While no vaccine has been developed to treat RSV, neither has any vac- (Krarup et al., 2015; McLellan et al., 2013; Ngwuta et al., 2015; Rossey,
cine which elicits ERD entered clinical trials in infants. Modern RSV vac- McLellan, Saelens, & Schepens, 2018). A phase 3 clinical trial in adults
cination attempts have included the production of immunogenic over 60 years of age observed no protection against lower respiratory
nanoparticles (G. Smith et al., 2012) or generation of live-attenuated tract infection (0.47% in the vaccinated group versus 0.44% in the pla-
RSV virus vaccines through random mutagenesis, guided attenuation cebo arm) (NCT02608502).
through recombinant RSV generation, or generation of recombinant bo- A separate phase 3 clinical trial was recently completed that investi-
vine RSV or PIV expressing human RSV-F and RSV-G (reviewed (Karron, gated whether immunization of pregnant women in their third trimes-
Buchholz, & Collins, 2013)). Owing to the difficult challenge of generat- ter with the RSV-F nanoparticle confers protection to infants via
ing an immune response in the first months of life when protection maternal antibodies (NCT02624947)(Madhi et al., 2020). The primary
against RSV is most important, maternal vaccination strategies are also endpoint was to test whether RSV-associated, medically significant
currently being explored in a phase 3 clinical trial (NCT02624947). lower respiratory tract infection could be reduced through maternal
vaccination, to protect neonates in their first 90 days of life (Madhi
6.4. Live attenuated RSV vaccines in clinical trials et al., 2020). This trial failed to achieve efficacy but the data suggest
that the RSV vaccination strategy is worthy of further investigation.
6.4.1. cpts248/404
cpts248/404 was the last live attenuated vaccine candidate to enter 7. Prophylaxis
clinical trials prior to the utilization of recombinant genetic engineering
technologies to introduce specific mutations. “cp” and “ts” refer to 7.1. Efficacy and cost-effectiveness of palivizumab
mutations generated through cold passaging or chemical mutagene-
sis-induced temperature sensitive mutations, respectively (Karron Palivizumab is a humanized monoclonal antibody containing
et al., 2013). In phase I clinical trials, cpts248/404 was tested in steadily murine-origin complementarity-determining regions specific for the
younger age cohorts, culminating in administration to infants less than pre-fusion conformation of RSV-F (S. Johnson et al., 1997). Palivizumab
two months of age (Wright et al., 2000). In seropositive children be- was licensed for prophylactic treatment against RSV infection in high-
tween 15 and 59 months of age cpts248/404 live virus was not shed, risk infants following the IMpact clinical trial. This clinical trial included
and antibody responses were nearly non-existent. In seronegative chil- 1502 infants born at less than 35 weeks gestational age ("Prevention
dren less than 24 months of age, cpts248/404 live virus was shed and of respiratory syncytial virus infections: indications for the use of
neutralizing antibodies against RSV were observed in serum. However, palivizumab and update on the use of RSV-IGIV. American Academy
this study was not powered to determine the efficacy of protection of Pediatrics Committee on Infectious Diseases and Committee of
against natural RSV infection. In the youngest cohort of 1 to 2-month Fetus and Newborn," 1998). Monthly administration of palivizumab in
old infants, neutralizing antibody development was rare – as is the premature infants without bronchopulmonary dysplasia, a type of
case for natural infection in this age group. As expected, maternal anti- chronic lung disease (CLD), reduced the rate of hospitalization in this
body levels antagonized the development of most infant antibody re- cohort from 8.1 % in the placebo arm to 1.8% in the palivizumab arm
sponses, including IgG antibodies against RSV-F and RSV-G, and IgA (P < 0.001). For infants with bronchopulmonary dysplasia the rate of
antibodies against RSV-F. Finally, airway congestion associated with hospitalization was reduced from 12.8% to 7.9% between placebo and
peak cpts248/404 shedding precluded the vaccine candidate from fur- treated groups, respectively (P = 0.038). Shortly thereafter, another
ther study in the 1 to 2-month old cohort (Wright et al., 2000). large clinical trial was focussed specifically on 1287 infants with hemo-
dynamically significant chronic heart disease (CHD). This study ob-
6.4.2. MEDI-559 served a reduction in hospitalization from 9.7% to 5.3% (P = 0.003)
MEDI-559 is an attenuated mutant RSV molecular virus clone that (Feltes, et al., 2003). Subsequent population-based studies typically
was created via reverse genetics. Cpts248/404 was further attenuated confirmed that palivizumab effectively reduced the rate of hospitaliza-
by deletion of RSV-SH and introduction of a temperature sensitive tion due to RSV infection (Geskey, Thomas, & Brummel, 2007). Owing
Y1321K mutation in RSV-L (Malkin et al., 2013). In RSV-seronegative to these successes in clinical trials, palivizumab is widely prescribed to
children aged 5 to 24 months, MEDI-559 produced neutralizing high-risk infants.
antibody responses in 59% of infants versus 9% for the placebo group. Many limitations reduce the impact of palivizumab on the annual
Protection from natural RSV infection was not measured. Lower respira- RSV burden of disease. These include prohibitive cost, the time burden
tory tract illness was higher in the MEDI-559 arm than placebo yet was on families and physicians associated with monthly injections through-
comparable to the rate observed in placebo groups in other studies out the RSV season, the lack of efficacy against ongoing infection, and
making the safety of MEDI-559 difficult to interpret (Malkin et al., the development of resistant RSV strains. Cost-benefit analyses vary
2013). To date, subsequent clinical trials on MEDI-559 have not been widely between studies and study populations. Hampp et al. found
initiated. Results are not available for other live attenuated vaccine that for premature infants in Florida, 30 infants must be treated to

13
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

avoid a single hospitalization; thus, the cost per hospitalization avoided efforts, there are no available efficacious antivirals to treat RSV infection,
in this population was approximately $302,103. This is dramatically nor are vaccines available to prevent infection or any consensus on how
more expensive than the cost of hospitalization in this region, at ap- infants with RSV bronchiolitis should be treated. The discovery of com-
proximately $8,910 per infant in this population (Hampp, Kauf, Saidi, pounds with antiviral activity against RSV is of paramount importance.
& Winterstein, 2011). In contrast, for inhabitants of Baffin Island in Nu- Despite several decades of research, we have yet to develop an anti-
navut where medical evacuation to the Children’s Hospital of Eastern viral drug for the treatment of RSV infection. Therefore, there is still
Ontario drastically increases the cost of hospitalization, administering room to design new protocols which could rapidly identify compounds
palivizumab to all infants less than 6 months of age (as opposed to with antiviral activity against RSV, and ultimately address the substan-
only high-risk infants) may be a cost-effective strategy (Banerji et al., tial burden of disease it imposes.
2009). The Canadian Pediatric Society notes that palivizumab treatment
in Canada costs approximately $5,600 per infant, and the number Funding sources
needed to treat to prevent hospitalization ranges from 16-23 for various
high-risk groups (Robinson et al., 2015). As such, outside of Baffin Is- FE is supported by a graduate scholarship from the Women’s and
land, palivizumab is not a cost-effective treatment. Currently, the Cana- Children’s Health Research Institute, QK is supported by a graduate
dian Pediatric Society recommends prophylactic palivizumab treatment scholarship from Alberta Innovates, and DJM is supported by a Canada
for preterm infants, infants with CHD, immunocompromised infants, Research Chair in Viral Pathogenesis, and grants from the Canadian
and infants with CLD (Robinson et al., 2015). Institutes of Health Research. FGW is supported by a grant from the
Palivizumab has been examined as an intervention to treat serious Natural Sciences and Engineering Research Council of Canada.
RSV infection, and a significant reduction in tracheal RSV concentration
was observed (Malley et al., 1998). However, treatment with paliviz- Declaration of Competing Interest
umab did not produce clinically relevant benefits as there was no reduc-
tion in the duration of hospitalization, duration of mechanical The authors declare that there are no conflicts of interest.
ventilation, or duration of supplemental oxygen therapy (Malley et al.,
1998). Similarly, other studies have found no clinically relevant benefits Acknowledgments
of treatment of RSV infection with palivizumab (Helmink, Ragsdale,
Peterson, & Merkel, 2016). We thank Dr. Robert Newton for helpful discussion and expertise
provided in the Long and Short Acting Beta-adrenergic Agonists section.
7.2. RSV drug resistance

As an RNA virus with an error-prone RdRp, RSV exists as a References


quasispecies (Lauring & Andino, 2010). Therefore, RSV is expected to
have the capacity to rapidly develop mutations which confer resistance Andrew, J. D., & Gardner, P. S. (1963). Occurrence of Respiratory Syncytial Virus in Acute
Respiratory Diseases in Infancy. British Medical Journal 2, 1447–1448.
to antiviral interventions. The clinical implications of these resistant Asenjo, A., Calvo, E., & Villanueva, N. (2006). Phosphorylation of human respiratory syn-
mutants depend on the fitness cost associated with each mutation. cytial virus P protein at threonine 108 controls its interaction with the M2-1 protein
Prior to the licensure of palivizumab, RSV mutations in the antigenic A in the viral RNA polymerase complex. The Journal of General Virology 87, 3637–3642.
Bagga, B., Woods, C. W., Veldman, T. H., Gilbert, A., Mann, A., Balaratnam, G., ...
site of the RSV-F protein had already been observed to confer resistance
Devincenzo, J. P. (2013). Comparing influenza and RSV viral and disease dynamics
to antibody peptide fragments (Crowe et al., 1998). Resistance to in experimentally infected adults predicts clinical effectiveness of RSV antivirals.
palivizumab was first noted via the K272M mutation in RSV-F which de- Antiviral Therapy 18, 785–791.
veloped readily in cell culture during serial passaging in increasing Baicus, A. (2012). History of polio vaccination. World J Virol 1, 108–114.
Banerji, A., Lanctot, K. L., Paes, B. A., Masoud, S. T., Tam, D. Y., Macdonald, W. A., & Roberts,
palivizumab concentrations (Zhao, Chen, & Sullender, 2004). This muta- A. (2009). Comparison of the cost of hospitalization for respiratory syncytial virus dis-
tion was not associated with a reduction in fitness as growth kinetics in ease versus palivizumab prophylaxis in Canadian Inuit infants. The Pediatric Infectious
cell culture were not affected. Resistance to palivizumab via the K272M Disease Journal 28, 702–706.
Barik, S. (1993). The structure of the 5' terminal cap of the respiratory syncytial virus
mutation was confirmed in vivo in the Cotton Rat model. The known list
mRNA. The Journal of General Virology 74(Pt 3), 485–490.
of RSV-F mutations which confer resistance to palivizumab has grown Barik, S. (2013). Respiratory syncytial virus mechanisms to interfere with type 1 inter-
to include N262S, N268I, K272N, K272T, K272Q, S275F, and S275L ferons. Current Topics in Microbiology and Immunology 372, 173–191.
(Zhu et al., 2012). An RSV strain with an RSV-F K272E mutation was Battles, M. B., Langedijk, J. P., Furmanova-Hollenstein, P., Chaiwatpongsakorn, S., Costello,
H. M., Kwanten, L., ... McLellan, J. S. (2016). Molecular mechanism of respiratory syn-
the first clinic isolate with suspected resistance to palivizumab (Boivin cytial virus fusion inhibitors. Nature Chemical Biology 12, 87–93.
et al., 2008). In untreated populations, mutations associated with resis- Behera, A. K., Matsuse, H., Kumar, M., Kong, X., Lockey, R. F., & Mohapatra, S. S. (2001).
tance are less common, yet remain present. Of 145 untreated infants in- Blocking intercellular adhesion molecule-1 on human epithelial cells decreases respi-
ratory syncytial virus infection. Biochemical and Biophysical Research Communications
fected with RSV, palivizumab-resistant isolates RSV-F N262D and S275F
280, 188–195.
were identified with a frequency of 0.7% each (Zhu et al., 2012). Some Beigel, J. H., Nam, H. H., Adams, P. L., Krafft, A., Ince, W. L., El-Kamary, S. S., & Sims, A. C.
studies have found that wild type RSV strains outcompete (2019). Advances in respiratory virus therapeutics - A meeting report from the 6th
palivizumab-resistant RSV strains in direct competition assays (Zhu isirv Antiviral Group conference. Antiviral Research 167, 45–67.
Bilawchuk, L. M., Griffiths, C. D., Jensen, L. D., Elawar, F., & Marchant, D. J. (2017). The sus-
et al., 2011). This has likely limited the spread of palivizumab- ceptibilities of respiratory syncytial virus to nucleolin receptor blocking and antibody
resistant strains in the general population. Studies specifically examin- neutralization are dependent upon the method of virus purification. Viruses 9.
ing whether palivizumab efficacy has been reduced since licensure, Bitko, V., Shulyayeva, O., Mazumder, B., Musiyenko, A., Ramaswamy, M., Look, D. C., &
due to the circulation of resistant strains, have not been conducted. Barik, S. (2007). Nonstructural proteins of respiratory syncytial virus suppress pre-
mature apoptosis by an NF-kappaB-dependent, interferon-independent mechanism
These results on palivizumab, which was once thought to target a con- and facilitate virus growth. Journal of Virology 81, 1786–1795.
served epitope on RSV (DeVincenzo et al., 2004), suggest that resistance Blount, R. E., Jr., Morris, J. A., & Savage, R. E. (1956). Recovery of cytopathogenic agent
mutations to new biologics will likely emerge and surveillance will have from chimpanzees with coryza. Proceedings of the Society for Experimental Biology
and Medicine 92, 544–549.
to continue to understand the effect on prophylaxis. Boivin, G., Caouette, G., Frenette, L., Carbonneau, J., Ouakki, M., & De Serres, G. (2008).
Human respiratory syncytial virus and other viral infections in infants receiving
8. Conclusions palivizumab. Journal of Clinical Virology 42, 52–57.
Bonfanti, J. -F., Meyer, C., Doublet, F., Fortin, J., Muller, P., Queguiner, L., ... Andries, K.
(2008). Selection of a respiratory syncytial virus fusion inhibitor clinical candidate.
RSV infection imposes a significant burden of disease on infants, the 2. discovery of a morpholinopropylaminobenzimidazole derivative (TMC353121).
elderly, and immunocompromised individuals. Despite intensive Journal of Medicinal Chemistry 51, 875–896.

14
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Bouillier, C., Cosentino, G., Leger, T., Rincheval, V., Richard, C. A., Desquesnes, A., ... Rameix- the Treatment of Respiratory Syncytial Virus Infection. Antimicrobial Agents and
Welti, M. A. (2019). The Interactome analysis of the Respiratory Syncytial Virus pro- Chemotherapy 60, 6–13.
tein M2-1 suggests a new role in viral mRNA metabolism post-transcription. Scientific Deval, J., Hong, J., Wang, G., Taylor, J., Smith, L. K., Fung, A., ... Beigelman, L. (2015a). Mo-
Reports 9, 15258. lecular basis for the selective inhibition of respiratory syncytial virus RNA polymerase
Broadbent, L., Groves, H., Shields, M. D., & Power, U. F. (2015). Respiratory syncytial virus, by 2’-fluoro-4’-chloromethyl-cytidine triphosphate. PLoS Pathogens 11, e1004995.
an ongoing medical dilemma: an expert commentary on respiratory syncytial virus Deval, J., Hong, J., Wang, G., Taylor, J., Smith, L. K., Fung, A., ... Beigelman, L. (2015b). Mo-
prophylactic and therapeutic pharmaceuticals currently in clinical trials. Influenza lecular basis for the selective inhibition of respiratory syncytial virus RNA polymerase
and Other Respiratory Viruses 9, 169–178. by 2’-fluoro-4’-chloromethyl-cytidine triphosphate. PLoS Pathogens 11, e1004995.
Buckingham, S. C., Bush, A. J., & Devincenzo, J. P. (2000). Nasal quantity of respiratory DeVincenzo, J., Tait, D., Efthimiou, J., Mori, J., Kim, Y. I., Thomas, E., ... Littler, E. (2020). A
syncytical virus correlates with disease severity in hospitalized infants. The Pediatric randomized, placebo-controlled, respiratory syncytial virus human challenge study
Infectious Disease Journal 19, 113–117. of the antiviral efficacy, safety, and pharmacokinetics of RV521, an inhibitor of the
Bukreyev, A., Whitehead, S. S., Murphy, B. R., & Collins, P. L. (1997). Recombinant respira- RSV-F protein. Antimicrobial Agents and Chemotherapy 64.
tory syncytial virus from which the entire SH gene has been deleted grows efficiently Devincenzo, J. P. (2004). Natural infection of infants with respiratory syncytial virus sub-
in cell culture and exhibits site-specific attenuation in the respiratory tract of the groups A and B: a study of frequency, disease severity, and viral load. Pediatric
mouse. Journal of Virology 71, 8973–8982. Research 56, 914–917.
Bukreyev, A., Yang, L., & Collins, P. L. (2012). The secreted G protein of human respiratory DeVincenzo, J. P., Hall, C. B., Kimberlin, D. W., Sanchez, P. J., Rodriguez, W. J., Jantausch, B.
syncytial virus antagonizes antibody-mediated restriction of replication involving A., ... Piazza, F. M. (2004). Surveillance of clinical isolates of respiratory syncytial virus
macrophages and complement. Journal of Virology 86, 10880–10884. for palivizumab (Synagis)-resistant mutants. The Journal of Infectious Diseases 190,
Cai, Z., Lin, Y., & Liang, J. (2020). Efficacy of salbutamol in the treatment of infants with 975–978.
bronchiolitis: A meta-analysis of 13 studies. Medicine (Baltimore) 99, e18657. DeVincenzo, J. P., McClure, M. W., Symons, J. A., Fathi, H., Westland, C., Chanda, S., ... Fry, J.
Carrat, F., Vergu, E., Ferguson, N. M., Lemaitre, M., Cauchemez, S., Leach, S., & Valleron, A. J. (2015). Activity of oral ALS-008176 in a respiratory syncytial virus challenge study.
(2008). Time lines of infection and disease in human influenza: a review of volunteer The New England Journal of Medicine 373, 2048–2058.
challenge studies. American Journal of Epidemiology 167, 775–785. DeVincenzo, J. P., Whitley, R. J., Mackman, R. L., Scaglioni-Weinlich, C., Harrison, L., Farrell,
Cazzola, M., Beeh, K. M., Price, D., & Roche, N. (2015). Assessing the clinical value of fast E., ... Chien, J. W. (2014). Oral GS-5806 activity in a respiratory syncytial virus chal-
onset and sustained duration of action of long-acting bronchodilators for COPD. lenge study. The New England Journal of Medicine 371, 711–722.
Pulmonary Pharmacology & Therapeutics 31, 68–78. DeVincenzo, J. P., Wilkinson, T., Vaishnaw, A., Cehelsky, J., Meyers, R., Nochur, S., ...
Challa, S., Scott, A. D., Yuzhakov, O., Zhou, Y., Tiong-Yip, C. L., Gao, N., ... Yu, Q. (2015). Lambkin-Williams, R. (2010). Viral load drives disease in humans experimentally in-
Mechanism of action for respiratory syncytial virus inhibitor RSV604. Antimicrobial fected with respiratory syncytial virus. American Journal of Respiratory and Critical
Agents and Chemotherapy 59, 1080–1087. Care Medicine 182, 1305–1314.
Chanock, R., & Finberg, L. (1957). Recovery from infants with respiratory illness of a virus Domachowske, J. B., Khan, A. A., Esser, M. T., Jensen, K., Takas, T., Villafana, T., ... Griffin, M.
related to chimpanzee coryza agent (CCA). II. Epidemiologic aspects of infection in in- P. (2018). Safety, tolerability and pharmacokinetics of MEDI8897, an extended half-
fants and young children. American Journal of Hygiene 66, 291–300. life single-dose respiratory syncytial virus prefusion F-targeting monoclonal antibody
Chanock, R. M., Murphy, B. R., Collins, P. L., Coelingh, K. V., Olmsted, R. A., Snyder, M. H., ... administered as a single dose to healthy preterm infants. The Pediatric Infectious
Flores, J., et al. (1988). Live viral vaccines for respiratory and enteric tract diseases. Disease Journal 37, 886–892.
Vaccine 6, 129–133.
Douglas, J. L., Panis, M. L., Ho, E., Lin, K. -Y., Krawczyk, S. H., Grant, D. M., ... Cihlar, T. (2005).
Chantaphakul, H., & Ruxrungtham, K. (2016). Fixed-Dose combination of the inhaled cor- Small molecules VP-14637 and JNJ-2408068 inhibit respiratory syncytial virus fusion
ticosteroid and long-acting beta2-agonist therapy in adults with persistent asthma. by similar mechanisms. Antimicrobial Agents and Chemotherapy 49, 2460.
Expert Opinion on Pharmacotherapy 17, 631–642.
Douglas, J. L., Panis, M. L., Ho, E., Lin, K. -Y., Krawczyk, S. H., Grant, D. M., ... Cihlar, T. (2003).
Chapman, J., Abbott, E., Alber, D. G., Baxter, R. C., Bithell, S. K., Henderson, E. A., ... Powell, K.
Inhibition of Respiratory Syncytial Virus Fusion by the Small Molecule VP-14637 via
L. (2007). RSV604, a novel inhibitor of respiratory syncytial virus replication.
Specific Interactions with F Protein. Journal of Virology 77, 5054.
Antimicrobial Agents and Chemotherapy 51, 3346–3353.
Dupuy, L. C., Dobson, S., Bitko, V., & Barik, S. (1999). Casein kinase 2-mediated phosphor-
Clemens, M. J., & Elia, A. (1997). The double-stranded RNA-dependent protein kinase
ylation of respiratory syncytial virus phosphoprotein P is essential for the transcrip-
PKR: structure and function. Journal of Interferon & Cytokine Research 17, 503–524.
tion elongation activity of the viral polymerase; phosphorylation by casein kinase 1
Clemens, M. J., Hershey, J. W., Hovanessian, A. C., Jacobs, B. C., Katze, M. G., Kaufman, R. J., ...
occurs mainly at Ser(215) and is without effect. Journal of Virology 73, 8384–8392.
Williams, B. R. (1993). PKR: proposed nomenclature for the RNA-dependent protein
El Saleeby, C. M., Bush, A. J., Harrison, L. M., Aitken, J. A., & Devincenzo, J. P. (2011). Respi-
kinase induced by interferon. Journal of Interferon Research 13, 241.
ratory syncytial virus load, viral dynamics, and disease severity in previously healthy
Coates, M., Brookes, D., Kim, Y. I., Allen, H., Fordyce, E. A. F., Meals, E. A., ... Rapeport, G.
naturally infected children. The Journal of Infectious Diseases 204, 996–1002.
(2017). Preclinical characterization of PC786, an inhaled small-molecule respiratory
Elawar, F., Griffiths, C. D., Zhu, D., Bilawchuk, L. M., Jensen, L. D., Forss, L., ... Marchant, D. J.
syncytial virus l protein polymerase inhibitor. Antimicrobial Agents and
(2017). A Virological and Phylogenetic Analysis of the Emergence of New Clades of
Chemotherapy 61.
Respiratory Syncytial Virus. Scientific Reports 7, 12232.
Collins, P. L., & Graham, B. S. (2008). Viral and host factors in human respiratory syncytial
virus pathogenesis. Journal of Virology 82, 2040–2055. Fearns, R., & Collins, P. L. (1999). Model for polymerase access to the overlapped L gene of
respiratory syncytial virus. Journal of Virology 73, 388–397.
Collins, P. L., Hill, M. G., Camargo, E., Grosfeld, H., Chanock, R. M., & Murphy, B. R. (1995).
Production of infectious human respiratory syncytial virus from cloned cDNA con- Feld, J. J., & Liang, T. J. (2005). HCV persistence: cure is still a four letter word. Hepatology
firms an essential role for the transcription elongation factor from the 5’ proximal 41, 23–25.
open reading frame of the M2 mRNA in gene expression and provides a capability Feldman, S. A., Audet, S., & Beeler, J. A. (2000). The fusion glycoprotein of human respira-
for vaccine development. Proceedings of the National Academy of Sciences of the tory syncytial virus facilitates virus attachment and infectivity via an interaction with
United States of America 92, 11563–11567. cellular heparan sulfate. Journal of Virology 74, 6442–6447.
Collins, P. L., Hill, M. G., Cristina, J., & Grosfeld, H. (1996). Transcription elongation factor of Feltes, T. F., Cabalka, A. K., Meissner, H. C., Piazza, F. M., Carlin, D. A., Top, F. H., Jr., ... Cardiac
respiratory syncytial virus, a nonsegmented negative-strand RNA virus. Proceedings of Synagis Study, G (2003). Palivizumab prophylaxis reduces hospitalization due to re-
the National Academy of Sciences of the United States of America 93, 81–85. spiratory syncytial virus in young children with hemodynamically significant con-
Collins, P. L., & Melero, J. A. (2011). Progress in understanding and controlling respiratory genital heart disease. The Journal of Pediatrics 143, 532–540.
syncytial virus: still crazy after all these years. Virus Research 162, 80–99. Fodha, I., Vabret, A., Ghedira, L., Seboui, H., Chouchane, S., Dewar, J., ... Freymuth, F. (2007).
Cowton, V. M., McGivern, D. R., & Fearns, R. (2006). Unravelling the complexities of respi- Respiratory syncytial virus infections in hospitalized infants: association between
ratory syncytial virus RNA synthesis. The Journal of General Virology 87, 1805–1821. viral load, virus subgroup, and disease severity. Journal of Medical Virology 79,
Crowe, J. E., Firestone, C. Y., Crim, R., Beeler, J. A., Coelingh, K. L., Barbas, C. F., ... Murphy, B. 1951–1958.
R. (1998). Monoclonal antibody-resistant mutants selected with a respiratory syncy- Gadomski, A. M., & Scribani, M. B. (2014). Bronchodilators for bronchiolitis. Cochrane
tial virus-neutralizing human antibody fab fragment (Fab 19) define a unique epitope Database of Systematic Reviews 6, 1–63 CD001266.
on the fusion (F) glycoprotein. Virology 252, 373–375. Garcia, C. S., Curiel, R. E., Mwatibo, J. M., Pestka, S., Li, H., & Espinoza-Delgado, I. (2006).
Cruz-Sanchez, T. M., Haddrell, A. E., Hackett, T. L., Singhera, G. K., Marchant, D., Lekivetz, R., The antineoplastic agent bryostatin-1 differentially regulates IFN-gamma receptor
... Agnes, G. R. (2013). Formation of a stable mimic of ambient particulate matter con- subunits in monocytic cells: transcriptional and posttranscriptional control of IFN-
taining viable infectious respiratory syncytial virus and its dry-deposition directly gamma R2. Journal of Immunology 177, 2707–2716.
onto cell cultures. Analytical Chemistry 85, 898–906. Geskey, J. M., Thomas, N. J., & Brummel, G. L. (2007). Palivizumab: a review of its use in
De Clercq, E., & Li, G. (2016). Approved Antiviral Drugs over the Past 50 Years. Clinical the protection of high risk infants against respiratory syncytial virus (RSV). Biologics
Microbiology Reviews 29, 695–747. 1, 33–43.
De Clercq, E., & Schinazi, R. F. (2016). Editorial overview: Antiviral strategies. Current Gilman, M. S. A., Liu, C., Fung, A., Behera, I., Jordan, P., Rigaux, P., ... McLellan, J. S. (2019).
Opinion in Virology 18, v–vi. Structure of the respiratory syncytial virus polymerase complex. Cell 179(193–204),
Delgado, M. F., Coviello, S., Monsalvo, A. C., Melendi, G. A., Hernandez, J. Z., Batalle, J. P., ... e114.
Polack, F. P. (2009). Lack of antibody affinity maturation due to poor Toll-like recep- Gottlieb, J., Zamora, M. R., Hodges, T., Musk, A. W., Sommerwerk, U., Dilling, D., ... Glanville,
tor stimulation leads to enhanced respiratory syncytial virus disease. Nature Medicine A. R. (2016). ALN-RSV01 for prevention of bronchiolitis obliterans syndrome after re-
15, 34–41. spiratory syncytial virus infection in lung transplant recipients. The Journal of Heart
Derish, M., Hodge, G., Dunn, C., & Ariagno, R. (1998). Aerosolized albuterol improves air- and Lung Transplantation 35, 213–221.
way reactivity in infants with acute respiratory failure from respiratory syncytial Grad, Y. H., Newman, R., Zody, M., Yang, X., Murphy, R., Qu, J., ... DeVincenzo, J. (2014).
virus. Pediatric Pulmonology 26, 12–20. Within-host whole-genome deep sequencing and diversity analysis of human respi-
Detalle, L., Stohr, T., Palomo, C., Piedra, P. A., Gilbert, B. E., Mas, V., ... Depla, E. (2016). Gen- ratory syncytial virus infection reveals dynamics of genomic diversity in the absence
eration and Characterization of ALX-0171, a Potent Novel Therapeutic Nanobody for and presence of immune pressure. Journal of Virology 88, 7286–7293.

15
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Griffin, M. P., Khan, A. A., Esser, M. T., Jensen, K., Takas, T., Kankam, M. K., ... Dubovsky, F. Karron, R. A., Buchholz, U. J., & Collins, P. L. (2013). Live-attenuated respiratory syncytial
(2017). Safety, tolerability, and pharmacokinetics of MEDI8897, the respiratory syn- virus vaccines. Current Topics in Microbiology and Immunology 372, 259–284.
cytial virus prefusion f-targeting monoclonal antibody with an extended half-life, in Karron, R. A., Buonagurio, D. A., Georgiu, A. F., Whitehead, S. S., Adamus, J. E., Clements-
healthy adults. Antimicrobial Agents and Chemotherapy 61. Mann, M. L., ... Sidhu, M. S. (1997). Respiratory syncytial virus (RSV) SH and G pro-
Griffiths, C., Drews, S. J., & Marchant, D. J. (2017). Respiratory syncytial virus: infection, teins are not essential for viral replication in vitro: clinical evaluation and molecular
detection, and new options for prevention and treatment. Clinical Microbiology characterization of a cold-passaged, attenuated RSV subgroup B mutant. Proceedings
Reviews 30, 277–319. of the National Academy of Sciences of the United States of America 94, 13961–13966.
Griffiths, C. D., Bilawchuk, L. M., McDonough, J. E., Jamieson, K. C., Elawar, F., Cen, Y., ... Karron, R. A., Wright, P. F., Belshe, R. B., Thumar, B., Casey, R., Newman, F., ... Collins, P. L.
Marchant, D. J. (2020). IGF1R is an entry receptor for respiratory syncytial virus. (2005). Identification of a recombinant live attenuated respiratory syncytial virus
Nature 583, 615–619. vaccine candidate that is highly attenuated in infants. The Journal of Infectious
Grosfeld, H., Hill, M. G., & Collins, P. L. (1995). RNA replication by respiratory syncytial Diseases 191, 1093–1104.
virus (RSV) is directed by the N, P, and L proteins; transcription also occurs under Kim, H. W., Canchola, J. G., Brandt, C. D., Pyles, G., Chanock, R. M., Jensen, K., & Parrott, R. H.
these conditions but requires RSV superinfection for efficient synthesis of full- (1969). Respiratory syncytial virus disease in infants despite prior administration of
length mRNA. Journal of Virology 69, 5677–5686. antigenic inactivated vaccine. American Journal of Epidemiology 89, 422–434.
Groskreutz, D. J., Babor, E. C., Monick, M. M., Varga, S. M., & Hunninghake, G. W. (2010). Kim, Y. I., Pareek, R., Murphy, R., Harrison, L., Farrell, E., Cook, R., & DeVincenzo, J. (2017b).
Respiratory syncytial virus limits alpha subunit of eukaryotic translation initiation The antiviral effects of RSV fusion inhibitor, MDT-637, on clinical isolates, vs its
factor 2 (eIF2alpha) phosphorylation to maintain translation and viral replication. achievable concentrations in the human respiratory tract and comparison to ribavi-
The Journal of Biological Chemistry 285, 24023–24031. rin. Influenza and Other Respiratory Viruses 11, 525–530.
Hall, C. B., Douglas, R. G., Jr., & Geiman, J. M. (1980). Possible transmission by fomites of Kim, Y. -I., Pareek, R., Murphy, R., Harrison, L., Farrell, E., Cook, R., & DeVincenzo, J.
respiratory syncytial virus. The Journal of Infectious Diseases 141, 98–102. (2017a). The antiviral effects of RSV fusion inhibitor, MDT-637, on clinical isolates,
Hall, C. B., Walsh, E. E., Long, C. E., & Schnabel, K. C. (1991). Immunity to and frequency of vs its achievable concentrations in the human respiratory tract and comparison to ri-
reinfection with respiratory syncytial virus. The Journal of Infectious Diseases 163, bavirin. Influenza and Other Respiratory Viruses 11, 525–530.
693–698. Kirkland, S. W., Cross, E., Campbell, S., Villa-Roel, C., & Rowe, B. H. (2018). Intramuscular
Hammer, J., Numa, A., & Newth, C. J. (1995). Albuterol responsiveness in infants with re- versus oral corticosteroids to reduce relapses following discharge from the emer-
spiratory failure caused by respiratory syncytial virus infection. The Journal of gency department for acute asthma. Cochrane Database of Systematic Reviews 6,
Pediatrics 127, 485–490. CD012629.
Hampp, C., Kauf, T. L., Saidi, A. S., & Winterstein, A. G. (2011). Cost-effectiveness of respi- Kiss, G., Holl, J. M., Williams, G. M., Alonas, E., Vanover, D., Lifland, A. W., ... Wright, E. R.
ratory syncytial virus prophylaxis in various indications. Archives of Pediatrics & (2014). Structural analysis of respiratory syncytial virus reveals the position of M2-
Adolescent Medicine 165, 498–505. 1 between the matrix protein and the ribonucleoprotein complex. Journal of
Hara, K., Yaita, K., Khamrin, P., Kumthip, K., Kashiwagi, T., Eleouet, J. F., ... Watanabe, H. Virology 88, 7602–7617.
(2020). A small fragmented P protein of respiratory syncytial virus inhibits virus in- Kiyomoto, A., Iwasawa, Y., & Harigaya, S. (1970). Studies on tetrahydroisoquinolines
fection by targeting P protein. The Journal of General Virology 101, 21–32. (THI). VI. Effects of trimetoquinol on tracheal and some other smooth muscles.
Hardelid, P., Verfuerden, M., McMenamin, J., Smyth, R. L., & Gilbert, R. (2019). The contri- Arzneimittelforschung 20, 46–52.
bution of child, family and health service factors to respiratory syncytial virus (RSV) Krarup, A., Truan, D., Furmanova-Hollenstein, P., Bogaert, L., Bouchier, P., Bisschop, I. J., ...
hospital admissions in the first 3 years of life: birth cohort study in Scotland, 2009 to Langedijk, J. P. (2015). A highly stable prefusion RSV F vaccine derived from structural
2015. Euro Surveillance 24. analysis of the fusion mechanism. Nature Communications 6, 8143.
Helmink, B. J., Ragsdale, C. E., Peterson, E. J., & Merkel, K. G. (2016). Comparison of Intra- Krzyzaniak, M. A., Zumstein, M. T., Gerez, J. A., Picotti, P., & Helenius, A. (2013). Host cell
venous Palivizumab and Standard of Care for Treatment of Respiratory Syncytial entry of respiratory syncytial virus involves macropinocytosis followed by proteolytic
Virus Infection in Mechanically Ventilated Pediatric Patients. Journal of Pediatric activation of the F protein. PLoS Pathogens 9, e1003309.
Pharmacology and Therapeutics 21, 146–154. Kulkarni, H., Smith, C. M., Lee Ddo, H., Hirst, R. A., Easton, A. J., & O’Callaghan, C. (2016).
Hislop, A. A. (2002). Airway and blood vessel interaction during lung development. Evidence of respiratory syncytial virus spread by aerosol. Time to revisit infection
Journal of Anatomy 201, 325–334. control strategies? American Journal of Respiratory and Critical Care Medicine 194,
Hong, Z., & Cameron, C. E. (2002). Pleiotropic mechanisms of ribavirin antiviral activities. 308–316.
Progress in Drug Research 59, 41–69. Laham, F. R., Mansbach, J. M., Piedra, P. A., Hasegawa, K., Sullivan, A. F., Espinola, J. A., &
Hornung, V., Ellegast, J., Kim, S., Brzozka, K., Jung, A., Kato, H., ... Hartmann, G. (2006). 5'- Camargo, C. A., Jr. (2017). Clinical profiles of respiratory syncytial virus subtypes A
Triphosphate RNA is the ligand for RIG-I. Science 314, 994–997. AND B among children hospitalized with bronchiolitis. The Pediatric Infectious
Houghton, M. (2014). Hepatitis C: the next 25years. Antiviral Research 110, 77–78. Disease Journal 36, 808–810.
Hruska, J. F., Bernstein, J. M., Douglas, R. G., Jr., & Hall, C. B. (1980). Effects of ribavirin on Lam, J. K., Chow, M. Y., Zhang, Y., & Leung, S. W. (2015). siRNA versus miRNA as therapeu-
respiratory syncytial virus in vitro. Antimicrobial Agents and Chemotherapy 17, tics for gene silencing. Molecular Therapy–Nucleic Acids 4, e252.
770–775. Lauring, A. S., & Andino, R. (2010). Quasispecies theory and the behavior of RNA viruses.
Huffman, J. H., Sidwell, R. W., Khare, G. P., Witkowski, J. T., Allen, L. B., & Robins, R. K. PLoS Pathogens 6, e1001005.
(1973). In vitro effect of 1-beta-D-ribofuranosyl-1,2,4-triazole-3-carboxamide Law, B. J., Wang, E. E., MacDonald, N., McDonald, J., Dobson, S., Boucher, F., ... Stephens, D.
(virazole, ICN 1229) on deoxyribonucleic acid and ribonucleic acid viruses. (1997). Does ribavirin impact on the hospital course of children with respiratory syn-
Antimicrobial Agents and Chemotherapy 3, 235–241. cytial virus (RSV) infection? An analysis using the pediatric investigators collabora-
Hultberg, A., Temperton, N. J., Rosseels, V., Koenders, M., Gonzalez-Pajuelo, M., Schepens, tive network on infections in Canada (PICNIC) RSV database. Pediatrics 99, E7.
B., ... de Haard, H. J. (2011). Llama-derived single domain antibodies to build multiva- Leader, S., Jacobson, P., Marcin, J., Vardis, R., Sorrentino, M., & Murray, D. (2002). A method
lent, superpotent and broadened neutralizing anti-viral molecules. PLoS One 6, for identifying the financial burden of hospitalized infants on families. Value in Health
e17665. 5, 55–59.
Ispas, G., Koul, A., Verbeeck, J., Sheehan, J., Sanders-Beer, B., Roymans, D., ... Verloes, R. Lichtman, M. A. (2013). A historical perspective on the development of the cytarabine
(2015). Antiviral Activity of TMC353121, a Respiratory Syncytial Virus (RSV) Fusion (7days) and daunorubicin (3days) treatment regimen for acute myelogenous leuke-
Inhibitor, in a Non-Human Primate Model. PLoS One 10, e0126959. mia: 2013 the 40th anniversary of 7+3. Blood Cells, Molecules & Diseases 50, 119–130.
Israel, S., Rusch, S., DeVincenzo, J., Boyers, A., Fok-Seang, J., Huntjens, D., ... Verloes, R. Lifland, A. W., Jung, J., Alonas, E., Zurla, C., Crowe, J. E., Jr., & Santangelo, P. J. (2012). Human
(2016). Effect of Oral JNJ-53718678 (JNJ-678) on Disease Severity in Healthy Adult respiratory syncytial virus nucleoprotein and inclusion bodies antagonize the innate
Volunteers Experimentally Inoculated With Live Respiratory Syncytial Virus (RSV): immune response mediated by MDA5 and MAVS. Journal of Virology 86, 8245–8258.
A Placebo-Controlled Challenge Study. Open Forum Infectious Diseases, 3. Lingemann, M., McCarty, T., Liu, X., Buchholz, U. J., Surman, S., Martin, S. E., ... Munir, S.
Jancar, S., & Sanchez Crespo, M. (2005). Immune complex-mediated tissue injury: a mul- (2019). The alpha-1 subunit of the Na+,K+-ATPase (ATP1A1) is required for
tistep paradigm. Trends in Immunology 26, 48–55. macropinocytic entry of respiratory syncytial virus (RSV) in human respiratory epi-
Janet, S., Broad, J., & Snape, M. D. (2018). Respiratory syncytial virus seasonality and its thelial cell. PLoS Pathogens 15, e1007963.
implications on prevention strategies. Human Vaccines & Immunotherapeutics 14, Mackman, R. L., Sangi, M., Sperandio, D., Parrish, J. P., Eisenberg, E., Perron, M., ... Cihlar, T.
234–244. (2015a). Discovery of an oral respiratory syncytial virus (RSV) fusion inhibitor (GS-
Jares Baglivo, S., & Polack, F. P. (2019). The long road to protect infants against severe RSV 5806) and clinical proof of concept in a Human RSV Challenge Study. Journal of
lower respiratory tract illness. F1000Res, 8. Medicinal Chemistry 58, 1630–1643.
Jefferson, T., Jones, M. A., Doshi, P., Del Mar, C. B., Hama, R., Thompson, M. J., ... Heneghan, Mackman, R. L., Sangi, M., Sperandio, D., Parrish, J. P., Eisenberg, E., Perron, M., ... Cihlar, T.
C. J. (2014). Neuraminidase inhibitors for preventing and treating influenza in (2015b). Discovery of an oral respiratory syncytial virus (RSV) fusion inhibitor (GS-
healthy adults and children. Cochrane Database of Systematic Reviews 4, 1–370 5806) and clinical proof of concept in a human RSV challenge study. Journal of
CD008965. Medicinal Chemistry 58, 1630–1643.
Johnson, S., Oliver, C., Prince, G. A., Hemming, V. G., Pfarr, D. S., Wang, S. C., ... Young, J. F. Madhi, S. A., Polack, F. P., Piedra, P. A., Munoz, F. M., Trenholme, A. A., Simoes, E. A. F., ...
(1997). Development of a humanized monoclonal antibody (MEDI-493) with potent Prepare Study, G. (2020). Respiratory syncytial virus vaccination during pregnancy
in vitro and in vivo activity against respiratory syncytial virus. The Journal of Infectious and effects in infants. The New England Journal of Medicine 383, 426–439.
Diseases 176, 1215–1224. Malhotra, R., Ward, M., Bright, H., Priest, R., Foster, M. R., Hurle, M., ... Bird, M. (2003). Iso-
Johnson, S. M., McNally, B. A., Ioannidis, I., Flano, E., Teng, M. N., Oomens, A. G., ... Peeples, lation and characterisation of potential respiratory syncytial virus receptor(s) on ep-
M. E. (2015). Respiratory Syncytial Virus Uses CX3CR1 as a Receptor on Primary ithelial cells. Microbes and Infection 5, 123–133.
Human Airway Epithelial Cultures. PLoS Pathogens 11, e1005318. Malkin, E., Yogev, R., Abughali, N., Sliman, J., Wang, C. K., Zuo, F., ... Dubovsky, F. (2013).
Karampatsas, K., Kong, J., & Cohen, J. (2019). Bronchiolitis: an update on management and Safety and immunogenicity of a live attenuated RSV vaccine in healthy RSV-
prophylaxis. British Journal of Hospital Medicine (London, England) 80, 278–284. seronegative children 5 to 24 months of age. PLoS One 8, e77104.

16
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

Malley, R., DeVincenzo, J., Ramilo, O., Dennehy, P. H., Meissner, H. C., Gruber, W. C., ... Plant, H., Stacey, C., Tiong-Yip, C. L., Walsh, J., Yu, Q., & Rich, K. (2015). High-throughput
Ambrosino, D. M. (1998). Reduction of respiratory syncytial virus (RSV) in tracheal hit screening cascade to identify respiratory syncytial virus (RSV) inhibitors. Journal
aspirates in intubated infants by use of humanized monoclonal antibody to RSV F of Biomolecular Screening 20, 597–605.
protein. The Journal of Infectious Diseases 178, 1555–1561. Poch, O., Sauvaget, I., Delarue, M., & Tordo, N. (1989). Identification of four conserved mo-
Marchant, D., Singhera, G. K., Utokaparch, S., Hackett, T. L., Boyd, J. H., Luo, Z., ... Hegele, R. tifs among the RNA-dependent polymerase encoding elements. The EMBO Journal 8,
G. (2010). Toll-like receptor 4-mediated activation of p38 mitogen-activated protein 3867–3874.
kinase is a determinant of respiratory virus entry and tropism. Journal of Virology 84, Polack, F. P., Teng, M. N., Collins, P. L., Prince, G. A., Exner, M., Regele, H., ... Karron, R. A.
11359–11373. (2002). A role for immune complexes in enhanced respiratory syncytial virus disease.
Martinello, R. A., Chen, M. D., Weibel, C., & Kahn, J. S. (2002). Correlation between respi- The Journal of Experimental Medicine 196, 859–865.
ratory syncytial virus genotype and severity of illness. The Journal of Infectious Prevention of respiratory syncytial virus infections (1998). Indications for the use of
Diseases 186, 839–842. palivizumab and update on the use of RSV-IGIV. American Academy of Pediatrics
Marty, F. M., Chemaly, R. F., Mullane, K. M., Lee, D. G., Hirsch, H. H., Small, C. B., ... Dadwal, Committee on Infectious Diseases and Committee of Fetus and Newborn. Pediatrics
S. S. (2019). A phase 2b, randomized, double-blind, placebo-controlled multicenter 102, 1211–1216.
study evaluating antiviral effects, pharmacokinetics, safety, and tolerability of Rima, B., Collins, P., Easton, A., Fouchier, R., Kurath, G., Lamb, R. A., ... Ictv Report, C. (2017).
presatovir in hematopoietic cell transplant recipients with respiratory syncytial ICTV Virus Taxonomy Profile: Pneumoviridae. The Journal of General Virology 98,
virus (RSV) infection of the lower respiratory tract. Clinical Infectious Diseases, 1–9 2912–2913.
Ahead of print. Robinson, J. L., Le Saux, N., Canadian Paediatric Society, I. D., & Immunization, C. (2015).
Mazur, N. I., Higgins, D., Nunes, M. C., Melero, J. A., Langedijk, A. C., Horsley, N., ... Preventing hospitalizations for respiratory syncytial virus infection. Paediatrics &
Respiratory Syncytial Virus Network, F. (2018). The respiratory syncytial virus vac- Child Health 20, 321–333.
cine landscape: lessons from the graveyard and promising candidates. The Lancet Rossey, I., McLellan, J. S., Saelens, X., & Schepens, B. (2018). Clinical Potential of Prefusion
Infectious Diseases 18, e295–e311. RSV F-specific Antibodies. Trends in Microbiology 26, 209–219.
McLellan, J. S., Chen, M., Leung, S., Graepel, K. W., Du, X., Yang, Y., ... Graham, B. S. (2013). Roy, C. J., & Milton, D. K. (2004). Airborne transmission of communicable infection–the
Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing elusive pathway. The New England Journal of Medicine 350, 1710–1712.
antibody. Science 340, 1113–1117. Roymans, D., Alnajjar, S. S., Battles, M. B., Sitthicharoenchai, P., Furmanova-Hollenstein, P.,
Mecklin, M., Heikkila, P., & Korppi, M. (2018). The change in management of bronchiolitis Rigaux, P., ... Koul, A. (2017a). Therapeutic efficacy of a respiratory syncytial virus fu-
in the intensive care unit between 2000 and 2015. European Journal of Pediatrics 177, sion inhibitor. Nature Communications 8, 167.
1131–1137. Roymans, D., Alnajjar, S. S., Battles, M. B., Sitthicharoenchai, P., Furmanova-Hollenstein, P.,
Meert, K. L., Sarnaik, A. P., Gelmini, M. J., & Lieh-Lai, M. W. (1994). Aerosolized ribavirin in Rigaux, P., ... Koul, A. (2017b). Therapeutic efficacy of a respiratory syncytial virus fu-
mechanically ventilated children with respiratory syncytial virus lower respiratory sion inhibitor. Nature Communications 8, 167.
tract disease: a prospective, double-blind, randomized trial. Critical Care Medicine Roymans, D., De Bondt, H. L., Arnoult, E., Geluykens, P., Gevers, T., Van Ginderen, M., ...
22, 566–572. Andries, K. (2010). Binding of a potent small-molecule inhibitor of six-helix bundle
Mitchell, I., Defoy, I., & Grubb, E. (2017). Burden of Respiratory Syncytial Virus Hospitali- formation requires interactions with both heptad-repeats of the RSV fusion protein.
zations in Canada. Canadian Respiratory Journal 2017, 4521302. Proceedings of the National Academy of Sciences 107, 308.
Mogensen, T. H. (2009). Pathogen recognition and inflammatory signaling in innate im- Ruckwardt, T. J., Morabito, K. M., & Graham, B. S. (2019). Immunological Lessons from Re-
mune defenses. Clinical Microbiology Reviews 22, 240–273 (Table of Contents). spiratory Syncytial Virus Vaccine Development. Immunity 51, 429–442.
Morales, F., Calder, M. A., Inglis, J. M., Murdoch, P. S., & Williamson, J. (1983). A study of Saez-Llorens, X., Moreno, M. T., Ramilo, O., Sanchez, P. J., Top, F. H., Jr., Connor, E. M., &
respiratory infections in the elderly to assess the role of respiratory syncytial virus. Group, M.-S (2004). Safety and pharmacokinetics of palivizumab therapy in children
The Journal of Infection 7, 236–247. hospitalized with respiratory syncytial virus infection. The Pediatric Infectious Disease
Journal 23, 707–712.
Morawska, L. (2006). Droplet fate in indoor environments, or can we prevent the spread
of infection? Indoor Air 16, 335–347. Samuel, D., Xing, W., Niedziela-Majka, A., Wong, J. S., Hung, M., Brendza, K. M., ...
Sakowicz, R. (2015). GS-5806 inhibits pre- to postfusion conformational changes of
Munday, D. C., Wu, W., Smith, N., Fix, J., Noton, S. L., Galloux, M., ... Hiscox, J. A. (2015). In-
the respiratory syncytial virus fusion protein. Antimicrobial Agents and
teractome analysis of the human respiratory syncytial virus RNA polymerase com-
Chemotherapy 59, 7109–7112.
plex identifies protein chaperones as important cofactors that promote L-protein
Scholtissek, C. (1976). Inhibition of influenza RNA synthesis by virazole (ribavirin).
stability and RNA synthesis. Journal of Virology 89, 917–930.
Archives of Virology 50, 349–352.
Murray, J., Bottle, A., Sharland, M., Modi, N., Aylin, P., Majeed, A., ... Medicines for Neonates
Schuster, H. P., & Baum, P. (1969). New isoprenaline-derivative (1-(3,5-
Investigator, G. (2014). Risk factors for hospital admission with RSV bronchiolitis in
dihydroxyphenyl)-1-hydroxy-2-1(4-hydroxyphenyl)-isopropyl-aminoethane. Th
England: a population-based birth cohort study. PLoS One 9, e89186.
1165 a) in therapy of acute cardiovascular and respiratory emergencies.
Nair, H., Nokes, D. J., Gessner, B. D., Dherani, M., Madhi, S. A., Singleton, R. J., ... Campbell, H.
Arzneimittelforschung 19, 1905–1914.
(2010). Global burden of acute lower respiratory infections due to respiratory syncy-
Selvaraj, M., Yegambaram, K., Todd, E., Richard, C. A., Dods, R. L., Pangratiou, G. M., ...
tial virus in young children: a systematic review and meta-analysis. Lancet 375,
Edwards, T. A. (2018). The Structure of the Human Respiratory Syncytial Virus M2-
1545–1555.
1 Protein Bound to the Interaction Domain of the Phosphoprotein P Defines the Ori-
Nelson, D. R., Zeuzem, S., Andreone, P., Ferenci, P., Herring, R., Jensen, D. M., ... Hill, G.
entation of the Complex. MBio 9.
(2012). Balapiravir plus peginterferon alfa-2a (40KD)/ribavirin in a randomized
Shi, T., Denouel, A., Tietjen, A. K., Campbell, I., Moran, E., Li, X., ... Investigators, R. (2019).
trial of hepatitis C genotype 1 patients. Annals of Hepatology 11, 15–31.
Global disease burden estimates of respiratory syncytial virus-associated acute respi-
Newton, R., & Giembycz, M. A. (2016). Understanding how long-acting beta2 ratory infection in older adults in 2015: A systematic review and meta-analysis. The
-adrenoceptor agonists enhance the clinical efficacy of inhaled corticosteroids in Journal of Infectious Diseases 222(7), S577–S583.
asthma - an update. British Journal of Pharmacology 173, 3405–3430. Shi, T., McAllister, D. A., O’Brien, K. L., Simoes, E. A. F., Madhi, S. A., Gessner, B. D., ...
Ngwuta, J. O., Chen, M., Modjarrad, K., Joyce, M. G., Kanekiyo, M., Kumar, A., ... Graham, B. Network, R. S. V. G. E. (2017). Global, regional, and national disease burden estimates
S. (2015). Prefusion F-specific antibodies determine the magnitude of RSV neutraliz- of acute lower respiratory infections due to respiratory syncytial virus in young chil-
ing activity in human sera. Science Translational Medicine 7, 309ra162. dren in 2015: a systematic review and modelling study. Lancet 390, 946–958.
Noah, J. W., Severson, W., Chung, D. H., Moore, B., Jia, F., Xu, X., ... Aube, J. (2010). A cell Sidwell, R. W., Huffman, J. H., Khare, G. P., Allen, L. B., Witkowski, J. T., & Robins, R. K.
based hts approach for the discovery of new inhibitors of RSV. Probe Reports from (1972). Broad-spectrum antiviral activity of Virazole: 1-beta-D-ribofuranosyl-1,2,4-
the NIH Molecular Libraries Program. Bethesda (MD). triazole-3-carboxamide. Science 177, 705–706.
Noton, S. L., Deflube, L. R., Tremaglio, C. Z., & Fearns, R. (2012). The respiratory syncytial Sinha, I. P., Dodd, S. R., Grime, C., Hawcutt, D. B., Fernandes, R. M., Roberts, M. R., ...
virus polymerase has multiple RNA synthesis activities at the promoter. PLoS Williamson, P. R. (2020). Where next for inhaled corticosteroids in childhood
Pathogens 8, e1002980. asthma? The Lancet Respiratory Medicine 8, 345.
Noton, S. L., Nagendra, K., Dunn, E. F., Mawhorter, M. E., Yu, Q., & Fearns, R. (2015). Respi- Sloan, C., Heaton, M., Kang, S., Berrett, C., Wu, P., Gebretsadik, T., ... Hartert, T. (2017). The
ratory Syncytial Virus Inhibitor AZ-27 Differentially Inhibits Different Polymerase Ac- impact of temperature and relative humidity on spatiotemporal patterns of infant
tivities at the Promoter. Journal of Virology 89, 7786–7798. bronchiolitis epidemics in the contiguous United States. Health & Place 45, 46–54.
O’Byrne, P. M., FitzGerald, J. M., Bateman, E. D., Barnes, P. J., Zhong, N., Keen, C., ... Reddel, Smith, D. B., Kalayanov, G., Sund, C., Winqvist, A., Maltseva, T., Leveque, V. J. P., ... Smith, M.
H. K. (2018). Inhaled Combined Budesonide-Formoterol as Needed in Mild Asthma. (2009). The design, synthesis, and antiviral activity of monofluoro and difluoro ana-
The New England Journal of Medicine 378, 1865–1876. logues of 4′-Azidocytidine against hepatitis C virus replication: The discovery of 4′-
Paes, B., Mitchell, I., Li, A., & Lanctot, K. L. (2012). Respiratory hospitalizations and respira- Azido-2′-deoxy-2′-fluorocytidine and 4′-Azido-2′-dideoxy-2′,2′-difluorocytidine.
tory syncytial virus prophylaxis in special populations. European Journal of Pediatrics Journal of Medicinal Chemistry 52, 2971–2978.
171, 833–841. Smith, D. W., Frankel, L. R., Mathers, L. H., Tang, A. T., Ariagno, R. L., & Prober, C. G. (1991).
Paes, B. A., Mitchell, I., Banerji, A., Lanctot, K. L., & Langley, J. M. (2011). A decade of respi- A controlled trial of aerosolized ribavirin in infants receiving mechanical ventilation
ratory syncytial virus epidemiology and prophylaxis: translating evidence into every- for severe respiratory syncytial virus infection. The New England Journal of Medicine
day clinical practice. Canadian Respiratory Journal 18, e10–e19. 325, 24–29.
Perron, M., Stray, K., Kinkade, A., Theodore, D., Lee, G., Eisenberg, E., ... Cihlar, T. (2015). Smith, E. A. (1991). Management of the infant with respiratory syncytial virus. Journal of
GS-5806 inhibits a broad range of respiratory syncytial virus clinical isolates by Pediatric Nursing 6, 434.
blocking the virus-cell fusion process. Antimicrobial Agents and Chemotherapy 60, Smith, G., Raghunandan, R., Wu, Y., Liu, Y., Massare, M., Nathan, M., ... Glenn, G. (2012).
1264–1273. Respiratory syncytial virus fusion glycoprotein expressed in insect cells form protein
Peter, J. V., John, P., Graham, P. L., Moran, J. L., George, I. A., & Bersten, A. (2008). Cortico- nanoparticles that induce protective immunity in cotton rats. PLoS One 7, e50852.
steroids in the prevention and treatment of acute respiratory distress syndrome Stevens, M., Rusch, S., DeVincenzo, J., Kim, Y. I., Harrison, L., Meals, E. A., ... Verloes, R.
(ARDS) in adults: meta-analysis. BMJ 336, 1006–1009. (2018). Antiviral activity of oral JNJ-53718678 in healthy adult volunteers challenged

17
F. Elawar, A.K. Oraby, Q. Kieser et al. Pharmacology & Therapeutics xxx (2020) xxx

with respiratory syncytial virus: A placebo-controlled study. The Journal of Infectious either the NS2 or SH gene is attenuated in chimpanzees. Journal of Virology 73,
Diseases 218, 748–756. 3438–3442.
Stray, K., Perron, M., Porter, D. P., Anderson, F., Lewis, S. A., Perry, J., ... Jordan, R. (2020). Witkowski, J. T., Robins, R. K., Sidwell, R. W., & Simon, L. N. (1972). Design, synthesis, and
Drug resistance assessment following administration of RSV fusion inhibitor broad spectrum antiviral activity of 1- -D-ribofuranosyl-1,2,4-triazole-3-carboxamide
presatovir to participants experimentally infected with respiratory syncytial virus. and related nucleosides. Journal of Medicinal Chemistry 15, 1150–1154.
The Journal of Infectious Diseases 9, 1468–1477. Wright, P. F., Karron, R. A., Belshe, R. B., Thompson, J., Crowe, J. E., Jr., Boyce, T. G., ...
Sudo, K., Miyazaki, Y., Kojima, N., Kobayashi, M., Suzuki, H., Shintani, M., & Shimizu, Y. Murphy, B. R. (2000). Evaluation of a live, cold-passaged, temperature-sensitive, re-
(2005). YM-53403, a unique anti-respiratory syncytial virus agent with a novel spiratory syncytial virus vaccine candidate in infancy. The Journal of Infectious
mechanism of action. Antiviral Research 65, 125–131. Diseases 182, 1331–1342.
Sun, Z., Pan, Y., Jiang, S., & Lu, L. (2013). Respiratory syncytial virus entry inhibitors Wright, R. B., Pomerantz, W. J., & Luria, J. W. (2002). New approaches to respiratory infec-
targeting the F protein. Viruses 5, 211–225. tions in children. Bronchiolitis and croup. Emergency Medicine Clinics of North America
Tawar, R. G., Duquerroy, S., Vonrhein, C., Varela, P. F., Damier-Piolle, L., Castagne, N., ... Rey, 20, 93–114.
F. A. (2009). Crystal structure of a nucleocapsid-like nucleoprotein-RNA complex of Wu, W., Tran, K. C., Teng, M. N., Heesom, K. J., Matthews, D. A., Barr, J. N., & Hiscox, J. A.
respiratory syncytial virus. Science 326, 1279–1283. (2012). The interactome of the human respiratory syncytial virus NS1 protein high-
Tayyari, F., Marchant, D., Moraes, T. J., Duan, W., Mastrangelo, P., & Hegele, R. G. (2011). lights multiple effects on host cell biology. Journal of Virology 86, 7777–7789.
Identification of nucleolin as a cellular receptor for human respiratory syncytial Yoneyama, M., Kikuchi, M., Matsumoto, K., Imaizumi, T., Miyagishi, M., Taira, K., ... Fujita, T.
virus. Nature Medicine 17, 1132–1135. (2005). Shared and unique functions of the DExD/H-box helicases RIG-I, MDA5, and
Tchesnokov, E. P., Raeisimakiani, P., Ngure, M., Marchant, D., & Gotte, M. (2018). Recom- LGP2 in antiviral innate immunity. Journal of Immunology 175, 2851–2858.
binant RNA-Dependent RNA Polymerase Complex of Ebola Virus. Scientific Reports 8, Yoneyama, M., Kikuchi, M., Natsukawa, T., Shinobu, N., Imaizumi, T., Miyagishi, M., ...
3970. Fujita, T. (2004). The RNA helicase RIG-I has an essential function in double-
Tiong-Yip, C. L., Aschenbrenner, L., Johnson, K. D., McLaughlin, R. E., Fan, J., Challa, S., ... Yu, stranded RNA-induced innate antiviral responses. Nature Immunology 5, 730–737.
Q. (2014). Characterization of a respiratory syncytial virus L protein inhibitor. Zhang, W., Yang, H., Kong, X., Mohapatra, S., San Juan-Vergara, H., Hellermann, G., ...
Antimicrobial Agents and Chemotherapy 58, 3867–3873. Mohapatra, S. S. (2005). Inhibition of respiratory syncytial virus infection with intra-
Trento, A., Casas, I., Calderon, A., Garcia-Garcia, M. L., Calvo, C., Perez-Brena, P., & Melero, J. nasal siRNA nanoparticles targeting the viral NS1 gene. Nature Medicine 11, 56–62.
A. (2010). Ten years of global evolution of the human respiratory syncytial virus BA Zhao, X., Chen, F. P., & Sullender, W. M. (2004). Respiratory syncytial virus escape mutant
genotype with a 60-nucleotide duplication in the G protein gene. Journal of Virology derived in vitro resists palivizumab prophylaxis in cotton rats. Virology 318, 608–612.
84, 7500–7512. Zhu, Q., McAuliffe, J. M., Patel, N. K., Palmer-Hill, F. J., Yang, C. F., Liang, B., ... Suzich, J. A.
Vasou, A., Paulus, C., Narloch, J., Gage, Z. O., Rameix-Welti, M. A., Eleouet, J. F., ... Adamson, (2011). Analysis of respiratory syncytial virus preclinical and clinical variants resis-
C. S. (2018). Modular cell-based platform for high throughput identification of com- tant to neutralization by monoclonal antibodies palivizumab and/or motavizumab.
pounds that inhibit a viral interferon antagonist of choice. Antiviral Research 150, The Journal of Infectious Diseases 203, 674–682.
79–92. Zhu, Q., McLellan, J. S., Kallewaard, N. L., Ulbrandt, N. D., Palaszynski, S., Zhang, J., ... Suzich,
Walsh, E. E., McConnochie, K. M., Long, C. E., & Hall, C. B. (1997). Severity of respiratory J. A. (2017). A highly potent extended half-life antibody as a potential RSV vaccine
syncytial virus infection is related to virus strain. JID 175, 814–820. surrogate for all infants. Science Translational Medicine, 9.
Wang, G., Deval, J., Hong, J., Dyatkina, N., Prhavc, M., Taylor, J., ... Beigelman, L. (2015). Dis- Zhu, Q., Patel, N. K., McAuliffe, J. M., Zhu, W., Wachter, L., McCarthy, M. P., & Suzich, J. A.
covery of 4'-chloromethyl-2'-deoxy-3',5'-di-O-isobutyryl-2'-fluorocytidine (ALS- (2012). Natural polymorphisms and resistance-associated mutations in the fusion
8176), a first-in-class RSV polymerase inhibitor for treatment of human respiratory protein of respiratory syncytial virus (RSV): effects on RSV susceptibility to
syncytial virus infection. Journal of Medicinal Chemistry 58, 1862–1878. palivizumab. The Journal of Infectious Diseases 205, 635–638.
Welliver, R. C., Sr. (2007). Temperature, humidity, and ultraviolet B radiation predict com- Zlateva, K. T., Lemey, P., Vandamme, A. M., & Van Ranst, M. (2004). Molecular evolution
munity respiratory syncytial virus activity. The Pediatric Infectious Disease Journal 26, and circulation patterns of human respiratory syncytial virus subgroup a: positively
S29–S35. selected sites in the attachment g glycoprotein. Journal of Virology 78, 4675–4683.
Whitehead, S. S., Bukreyev, A., Teng, M. N., Firestone, C. Y., St Claire, M., Elkins, W. R., ...
Murphy, B. R. (1999). Recombinant respiratory syncytial virus bearing a deletion of

18

You might also like