ArticlePDF Available

Transient Neutropenia in Immunocompetent Infants with Respiratory Syncytial Virus Infection

MDPI
Viruses
Authors:

Abstract and Figures

The incidence of neutropenia and the association between neutropenia and severity of respiratory symptoms among infants with respiratory syncytial virus (RSV) infections remain to be elucidated. This single-center, retrospective study included immunocompetent infants (<10 months old) with laboratory-confirmed RSV infection admitted to our center between January 2012 and December 2019. Incidence of neutropenia (<1.0 × 109/L) within 10 days of onset and risk factors associated with subsequent neutropenia were evaluated. Among the 292 infants with RSV infection, including 232 (79%) with mild infection, neutropenia was observed in 31 (11%), with severe neutropenia (<0.5 × 109/L) in 3 (1.0%). No neutropenic infants developed serious infection or hematological disorder. Infants without neutropenia showed age <3 months at onset in 34%, C-reactive protein level <1.0 mg/L in 27%, and nasopharyngeal microbiota composition with any of Moraxella catarrhalis, Streptococcus pneumoniae, or Haemophilus influenzae in 63%. In comparison, infants with neutropenia showed age <3 months at onset in 74% (relative risk [RR] 2.15; 95% confidence interval [CI] 1.65–2.81), C-reactive protein level <1.0 mg/L in 55% (RR 2.02; 95% CI 1.38–2.94), and microbiota including Moraxella catarrhalis, Streptococcus pneumoniae, or Haemophilus influenzae in 15% (RR 0.24; 95% CI 0.10–0.61). Multiple logistic regression analyses showed that younger age at onset and absence of that nasopharyngeal microbiota profile were associated with development of neutropenia. In conclusion, age and airway microbiota are considered as risk factors for the development of transient neutropenia among infants with RSV infection. However, the neutropenia seems not to develop serious infection or hematological disorder.
Content may be subject to copyright.
viruses
Article
Transient Neutropenia in Immunocompetent Infants with
Respiratory Syncytial Virus Infection
Tatsuya Korematsu and Hiroshi Koga *


Citation: Korematsu, T.; Koga, H.
Transient Neutropenia in
Immunocompetent Infants with
Respiratory Syncytial Virus Infection.
Viruses 2021,13, 301. https://
doi.org/10.3390/v13020301
Academic Editor: Oliver Schildgen
Received: 5 January 2021
Accepted: 13 February 2021
Published: 15 February 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Department of Pediatrics, National Hospital Organization Beppu Medical Center,
1473 Oaza-Uchikamado, Beppu, Oita 874-0011, Japan; tatsuya.korematsu@gmail.com
*Correspondence: koga.hiroshi.ab@mail.hosp.go.jp; Tel.: +81-977-67-1111
Abstract:
The incidence of neutropenia and the association between neutropenia and severity of
respiratory symptoms among infants with respiratory syncytial virus (RSV) infections remain to be
elucidated. This single-center, retrospective study included immunocompetent infants
(<10 months
old) with laboratory-confirmed RSV infection admitted to our center between January 2012 and
December 2019. Incidence of neutropenia (<1.0
×
10
9
/L) within 10 days of onset and risk factors
associated with subsequent neutropenia were evaluated. Among the 292 infants with RSV infec-
tion, including 232 (79%) with mild infection, neutropenia was observed in 31 (11%), with severe
neutropenia (<0.5
×
10
9
/L) in 3 (1.0%). No neutropenic infants developed serious infection or hema-
tological disorder. Infants without neutropenia showed age <3 months at onset in 34%, C-reactive
protein level <1.0 mg/L in 27%, and nasopharyngeal microbiota composition with any of Moraxella
catarrhalis,Streptococcus pneumoniae, or Haemophilus influenzae in 63%. In comparison, infants with
neutropenia showed age <3 months at onset in 74% (relative risk [RR] 2.15; 95% confidence interval
[CI] 1.65–2.81), C-reactive protein level <1.0 mg/L in 55% (RR 2.02; 95% CI 1.38–2.94), and microbiota
including Moraxella catarrhalis,Streptococcus pneumoniae, or Haemophilus influenzae in 15% (RR 0.24;
95% CI 0.10–0.61
). Multiple logistic regression analyses showed that younger age at onset and ab-
sence of that nasopharyngeal microbiota profile were associated with development of neutropenia. In
conclusion, age and airway microbiota are considered as risk factors for the development of transient
neutropenia among infants with RSV infection. However, the neutropenia seems not to develop
serious infection or hematological disorder.
Keywords:
host microbial interactions; leukopenia; neutrophils; respiratory tract infections; severity
of illness index
1. Introduction
Respiratory syncytial virus (RSV) continues to cause significant morbidity and mor-
tality among children and the elderly worldwide [
1
]. Approximately 33 million annual
episodes of RSV-associated lower respiratory infection occur among children <5 years,
resulting in 3 million hospital admissions and 60,000 in-hospital deaths [
2
]. Nearly half of
these admissions and deaths occur in infants <6 months [2].
To reduce the global burden of RSV infection, RSV vaccines and antivirals have
been developed and are currently in clinical trials [
3
]. Lumicitabine (ALS-008176 or JNJ-
64041575), an oral nucleoside analogue against RSV polymerase, has shown successful
results in phase I and II trials in adults [
4
]. However, adverse effects of neutropenia were
observed in a phase II trial of infants <3 years old, and the trial was terminated [
5
]. To
clarify the causal relationship, additional nonclinical study (monkey study) was undertaken
and showed potential clastogenic effects (data not shown). In clinical practice, neutropenia
subsequent to RSV infection, particularly persistent neutropenia, influences the decision to
perform a diagnostic work-up for serious infectious and hematological diseases.
Transient neutropenia can be observed in immunocompetent infants following viral in-
fections, including RSV infection [
6
8
]. However, the cumulative incidence of neutropenia
Viruses 2021,13, 301. https://doi.org/10.3390/v13020301 https://www.mdpi.com/journal/viruses
Viruses 2021,13, 301 2 of 10
in RSV-infected infants remains to be elucidated. The aim of this study was to determine
the incidence of neutropenia among infants with RSV infection and to investigate risk
factors for neutropenia following RSV infection. Our results provide insights into clinical
practice for RSV-infected infants and pathways to further clinical trials of RSV antivirals.
2. Methods
2.1. Study Design and Setting
This was a retrospective observational study of infants admitted to a single pediatric
center in Japan for RSV infection. Beppu Medical Center is a pediatric center for the
100,000 inhabitants of the city of Beppu, in the southeast of Japan. In this city, all pediatric
patients requiring emergency care or hospital admission are transported to our center.
This study protocol, based on the 2007 version of the Strengthening the Reporting of
Observational Studies in Epidemiology (STROBE) statement [
9
], was approved by the
ethics committee of Beppu Medical Center. Passive informed consent was obtained from
the parents of participants through the hospital website and bulletin board.
2.2. Study Participants
In this study, we used a global respiratory severity score (GRSS) [
10
], a clinical severity
score validated for full-term infants <10 months of age with RSV infection. Thus, we
reviewed the information of full-term infants who were <10 months old at the time of
hospitalization for RSV infection at our center between January 2012 and December 2019.
RSV infection was diagnosed based on rapid immunochromatographic antigen testing of
nasopharyngeal swabs or aspirates for RSV. The date of consecutive fever (>37.5
C) or
respiratory symptom occurrence was considered as the first day of RSV infection. Infants
with at least one complete blood count performed within 10 days of illness were included.
Exclusion criteria were as follows: neutropenia diagnosed before onset of RSV infection,
other preceding hematological disorder, malignancy, immune disorder, inborn errors of
metabolism, administration of drugs associated with myelosuppression, administration
of palivizumab within the preceding month, or enrollment in a clinical trial for RSV
antiviral agents.
2.3. Data Collection and Definitions
Information about baseline characteristics, physical findings, laboratory data, and
treatments during the course of RSV infection were also obtained from the electronic
medical records of eligible infants. Clinical severity of RSV infection was assessed on
admission using a GRSS. GRSS is calculated using nine variables: general appearance;
wheezing; rales/rhonchi; chest wall retractions; cyanosis; lethargy; poor air movement;
worst room air SaO
2
; and maximum respiratory rate [
10
]. GRSS is reportedly associated
with length of hospital stay and to affect the CD8
+
T-cell profile in infants with RSV
infection [
10
,
11
]. Neutropenia in infants <1 year old is defined as an absolute neutrophil
count (ANC) <1.0
×
10
9
/L (severe: <0.5
×
10
9
/L) [
8
]. The nutritional condition of infants
was assessed by the z-score of weight-for-length, as calculated using the Anthro Survey
Analyser by the World Health Organization [
12
]. Abnormal values for body temperature
(>38.5
C), heart rate (>180 beats/min), and respiratory rate (>34 breaths/min) were
defined according to systemic inflammatory response syndrome criteria for infants [13].
Analysis of microbial genomic sequences from nasopharyngeal aspirates in infants
with RSV infection demonstrated Streptococcus-, Moraxella-, and Haemophilus-dominant
profiles as the three major microbial profiles, and these nasopharyngeal microbiota profiles
were associated with the clearance of RSV [
14
]. Based on this report, the microbiota profile
was evaluated from bacterial cultures of nasopharyngeal aspirate in this study.
Viruses 2021,13, 301 3 of 10
2.4. Outcome Measures
The primary outcome measure was neutropenia in infants with RSV infection within
10 days of onset. Secondary outcome measures were the potential risk factors including
GRSS associated with the occurrence of neutropenia.
2.5. Follow-Up Assessment
Infants showing neutropenia during hospitalization for RSV infection were evaluated
for clinical signs of underlying serious infectious and hematological disorder, including
pancytopenia and peripheral blood blast cells. When no clinical signs suggested underlying
serious infectious or hematological diseases, infants with neutropenia were discharged
after recovery from RSV infection. The parents of infants with neutropenia were sent a
questionnaire in May 2020 to identify the recovery of neutropenia and subsequent onset of
any hematological disorder.
2.6. Statistical Analysis
Descriptive statistics were used to summarize basic and clinical characteristics, as
percentages. Bivariate associations were assessed using Pearson’s
χ2
test or Fisher’s
exact test. Wilcoxon rank-sum test was used to compare continuous variables. Patient
characteristics, GRSS, laboratory data, and treatment during the course of RSV infection
were compared between infants with and without neutropenia. Logistic regression analysis
was used to identify independent risk factors for neutropenia. The multivariate model
was adjusted for GRSS, potential confounders on univariate analysis (p< 0.05), and factors
previously reported as relevant to neutropenia, such as age and sex [8,15].
JMP version 14 software (SAS Institute Inc., Cary, NC, USA) was used for all statistical
analyses. Values of p< 0.05 were considered significant. Measures of effect are presented
as the risk ratio or odds ratio with 95% confidence interval (CI).
3. Results
3.1. Basic Characteristics
During the study period, a total of 308 infants <10 months old were admitted to our
hospital because of RSV infection. RSV infection was confirmed by rapid immunochromato-
graphic antigen testing in all infants. Among these infants, complete blood counts were
performed within 10 days of onset in the 298 infants who met the inclusion criteria, then six
infants were excluded (due to palivizumab administration within 1 month in five infants;
and participation in a clinical trial of RSV antiviral in one infant). A total of 292 infants
were subsequently considered eligible for this study. Among the 292 infants, 60 (21%) had
GRSS scores of >3.5 [
10
], a threshold for predicting hospitalization, and 232 (79%) had
less severe GRSS scores of
3.5. All 292 infants underwent complete blood counts and
differential evaluations at admission and 70 (24%) underwent two times or more blood
tests within 10 days of onset.
The ethnicity of the 292 eligible infants comprised 290 (99%) Asian (286 Japanese,
2 Korean, 1 Chinese, and 1 Uzbek), 2 (0.7%) white, and no black. A seasonal trend
in admissions of RSV-infected infants was observed as an increase from September to
December in 151 infants (52%) and a decrease from April to July in 32 (11%).
Among the 292 infants with RSV infection, median age at admission was 4.2 months
(interquartile range [IQR] 2.1–7.3 months). Neutropenia with ANC <1.0
×
10
9
/L was identi-
fied in 31 (11%) of these 292 infants, including severe neutropenia with
ANC <0.5 ×109/L
in 3 (1.0%). In addition, neutropenia with ANC <1.0
×
10
9
/L was observed in 32 (8.7%)
of the 368 complete blood counts performed within 10 days of illness. The incidence of
neutropenia according to the day of illness in a series of complete blood counts is illustrated
in Figure 1. Neutropenia was observed in approximately 5–15% of blood samples from
days 2 through 9 of illness in RSV-infected infants.
Viruses 2021,13, 301 4 of 10
Viruses 2021, 13, x FOR PEER REVIEW 4 of 10
the 368 complete blood counts performed within 10 days of illness. The incidence of neu-
tropenia according to the day of illness in a series of complete blood counts is illustrated
in Figure 1. Neutropenia was observed in approximately 515% of blood samples from
days 2 through 9 of illness in RSV-infected infants.
Figure 1. Distribution of absolute neutrophil count (ANC) and proportion of neutropenia <1.0 ×
109/L in respiratory syncytial virus (RSV)-infected infants according to day of illness. Each box
bounds the interquartile range of ANC, divided by the median. Solid line denotes the proportion
of neutropenia.
Bacterial cultures of nasopharyngeal aspirates were collected from 234 (81%) of 292
infants (26 with neutropenia, 208 without neutropenia). Blood cultures collected from 26
infants were all negative. During hospitalization, apnea was observed in five infants
(1.7%). In total, 289 infants (99%) were treated with inhaled bronchodilator or nebulized
hypertonic saline, 282 (97%) with intravenous fluid administration, 88 (30%) with oxygen
therapy, 58 (20%) with antibiotic administration, 14 (4.8%) with corticosteroid administra-
tion, nine (3.1%) with high-flow nasal cannula, and one (0.3%) with endotracheal intuba-
tion. None of the infants developed subsequent leukemia or other bone marrow disorder,
or died during the course of RSV infection.
3.2. Potential Risk Factors for Neutropenia
Among the 292 infants, a comparison of clinical manifestations between those with
and without neutropenia is summarized in Table 1. Comorbidities were observed in 10
infants without neutropenia, including congenital heart defects in four, central nervous
system abnormalities in three, cleft lip and palate in one, osteogenesis imperfecta in one,
and anhidrotic ectodermal dysplasia in one. Younger age at onset, lower C-reactive pro-
tein level, and less frequent identification of any of Moraxella catarrhalis, Streptococcus pneu-
moniae, or Haemophilus influenzae in nasopharyngeal aspirates were observed in infants
with neutropenia, compared to those without neutropenia. Median ANC and C-reactive
protein level on admission were higher in the 135 infants with any of these three bacteria
identified from nasopharyngeal aspirates (4.0 × 109/L and 8.1 mg/L) than in the 99 infants
without such a microbiota profile (2.0 × 109/L and 0.9 mg/L, p < 0.0001 and p < 0.0001,
respectively). Age at onset of RSV infection <3 months represented an absolute risk in-
crease of 40% for subsequent neutropenia (number needed to diagnose = 2.5 infants). No
significant difference in corticosteroid use and severity of RSV infection as evaluated by
GRSS was apparent between groups.
Figure 1.
Distribution of absolute neutrophil count (ANC) and proportion of neutropenia <1.0
×
10
9
/L in respiratory
syncytial virus (RSV)-infected infants according to day of illness. Each box bounds the interquartile range of ANC, divided
by the median. Solid line denotes the proportion of neutropenia.
Bacterial cultures of nasopharyngeal aspirates were collected from 234 (81%) of 292 in-
fants (26 with neutropenia, 208 without neutropenia). Blood cultures collected from 26 in-
fants were all negative. During hospitalization, apnea was observed in five infants (1.7%).
In total, 289 infants (99%) were treated with inhaled bronchodilator or nebulized hypertonic
saline, 282 (97%) with intravenous fluid administration, 88 (30%) with oxygen therapy,
58 (20%) with antibiotic administration, 14 (4.8%) with corticosteroid administration, nine
(3.1%) with high-flow nasal cannula, and one (0.3%) with endotracheal intubation. None of
the infants developed subsequent leukemia or other bone marrow disorder, or died during
the course of RSV infection.
3.2. Potential Risk Factors for Neutropenia
Among the 292 infants, a comparison of clinical manifestations between those with
and without neutropenia is summarized in Table 1. Comorbidities were observed in
10 infants without neutropenia, including congenital heart defects in four, central nervous
system abnormalities in three, cleft lip and palate in one, osteogenesis imperfecta in
one, and anhidrotic ectodermal dysplasia in one. Younger age at onset, lower C-reactive
protein level, and less frequent identification of any of Moraxella catarrhalis,Streptococcus
pneumoniae, or Haemophilus influenzae in nasopharyngeal aspirates were observed in infants
with neutropenia, compared to those without neutropenia. Median ANC and C-reactive
protein level on admission were higher in the 135 infants with any of these three bacteria
identified from nasopharyngeal aspirates (4.0
×
10
9
/L and 8.1 mg/L) than in the 99 infants
without such a microbiota profile (2.0
×
10
9
/L and 0.9 mg/L, p< 0.0001 and p< 0.0001,
respectively). Age at onset of RSV infection <3 months represented an absolute risk
increase of 40% for subsequent neutropenia (number needed to diagnose = 2.5 infants). No
significant difference in corticosteroid use and severity of RSV infection as evaluated by
GRSS was apparent between groups.
Viruses 2021,13, 301 5 of 10
Table 1. Comparison of clinical manifestations between RSV-infected infants with and without neutropenia <1.0 ×109/L.
Neutropenic Infants (n= 31) Non-neutropenic Infants (n= 261) Relative Risk (95%CI) pValue
Basic Characteristics
Female infant 18 (58) 113 (43) 1.34 (0.96–1.87) 0.12
Age < 3 months at onset 23 (74) 90 (34) 2.15 (1.65–2.81) <0.0001 *
Weight-for-length z-score < 2 0 (0) 15 (5.8) not applicable 0.38
Admission before day 5 of illness 22 (71) 156 (60) 1.19 (0.93–1.52) 0.23
Vital Signs and Oxygen Saturation
on Admission
Axial temperature > 38.5 C 2 (6.5) 39 (15) 0.43 (0.11–1.70) 0.28
Heart rate > 180 beats/min 0 (0) 18 (6.9) not applicable 0.23
Respiratory rate > 34 breaths/min 25 (81) 185 (71) 1.14 (0.94–1.37) 0.25
Arterial oxygen saturation < 95% 2 (6.5) 37 (14) 0.46 (0.12–1.80) 0.40
Laboratory Findings on Admission
Absolute lymphocyte count < 1.5 ×109/L 0 (0) 2 (0.8) not applicable 1.0
C-reactive protein < 1.0 mg/L 17 (55) 71 (27) 2.02 (1.38–2.94) 0.0015 *
Consolidation on chest radiography 2 (6.5) 52 (20) 0.32 (0.08–1.26) 0.086
Culture of Nasopharyngeal Aspirate
Moraxella catarrhalis 1 (3.9) 87 (42) 0.09 (0.01–0.63) <0.0001 *
Streptococcus pneumoniae 1 (3.9) 51 (25) 0.16 (0.02–1.09) 0.013 *
Haemophilus influenzae 2 (7.7) 45 (22) 0.36 (0.09–1.38) 0.12
Any of these 3 bacteria 4 (15) 131 (63) 0.24 (0.10–0.61) <0.0001 *
Treatments
Antibiotic use during course of RSV
infection 5 (16) 57 (22) 0.74 (0.32–1.70) 0.64
Corticosteroid use during course of RSV
infection 1 (3.2) 14 (5.4) 0.60 (0.08–4.42) 1.0
Hospital stay >7 days 4 (13) 58 (22) 0.58 (0.23–1.49) 0.35
Severity of RSV Infectio
GRSS on admission > 3.5 8 (26) 52 (20) 1.30 (0.68–2.47) 0.44
CI: confidence interval; GRSS: global respiratory severity score; RSV: respiratory syncytial virus. * p< 0.05;
Denominator of 26 (bacterial cultures were not performed in 5);
denominator of 208 (bacterial
cultures were not performed in 53).
Viruses 2021,13, 301 6 of 10
Multiple logistic regression analysis was performed to evaluate associations between
neutropenia and relevant factors such as sex, age at onset, C-reactive protein level, na-
sopharyngeal microbiota profile, and GRSS (Table 2). Neutropenia within 10 days of onset
in RSV-infected infants was associated with younger age at onset and identification of any
of the three bacterial species mentioned above in cultures of nasopharyngeal aspirate, but
was no longer associated with sex, C-reactive protein level, or GRSS.
Table 2.
Logistic regression analysis for predicting neutropenia <1.0
×
10
9
/L in infants with RSV infection.
Odds Ratio (95%CI) pValue
Any of the 3 bacteria identified in cultures of
nasopharyngeal aspirate 0.11 (0.03–0.35) <0.0001 *
Age at onset of RSV infection, per
1-month increment 0.81 (0.65–0.97) 0.020 *
GRSS on admission, per 1-point increment 0.92 (0.67–1.24) 0.59
C-reactive protein, per 1-mg/L increment 1.02 (0.99–1.05) 0.10
Female infant 1.49 (0.61–3.70) 0.38
CI: confidence interval; GRSS: global respiratory severity score; RSV: respiratory syncytial virus. * p< 0.05;
Moraxella catarrhalis,Streptococcus pneumoniae, or Haemophilus influenzae.
3.3. Follow-up Assessment
For the 31 neutropenic infants, 21 questionnaires (68%) were returned. Median follow-
up period was 3.6 years (IQR 1.8–5.0 years). None of the 21 infants developed hematological
disorder or other serious diseases after hospital discharge. Complete blood count was
performed in 19 (90%) of the 21 infants after discharge, and all 19 infants had recovered
(ANC >1.5 ×109/L) from neutropenia.
4. Discussion
This study elucidated the incidence and the risk factors of transient neutropenia in
infants with RSV infection. Among immunocompetent infants with RSV infection, 11%
of the infants showed the transient neutropenia (ANC <1.0
×
10
9
/L), and the transient
neutropenia demonstrated a correlation with age at onset and nasopharyngeal microbiota
profile (presence of M. catarrhalis,S. pneumoniae, or H. influenzae). Among RSV-infected
infants with transient neutropenia, no subsequent serious secondary infections, bone
marrow disorders, or deaths were observed. Our results suggest that comorbid neutropenia
is a transient phenomenon without developing severe infection or hematologic malignancy
among infants with RSV infection.
4.1. RSV Infection and Neutropenia
The estimated incidence of transient neutropenia among RSV-infected infants is 11% in
this study, which raises concerns about potential serious bacterial infection or bone marrow
disorder, particularly in prolonged neutropenia. The incidence of serious bacterial infection
has been reported as 3.7–4.2% among young febrile infants with viral infections [
16
,
17
],
2.1–8.5% among immunocompetent children with febrile neutropenia [
7
,
18
], and 1.1–7.0%
among young infants with RSV infection [
17
,
19
,
20
]. These risks are uncommon, but
non-negligible. Based on these results, targeted antibiotic treatment may be needed in
RSV-infected infants with febrile neutropenia. Prophylactic antibiotic administration may
provide clinical benefit for RSV-infected infants with febrile neutropenia [
18
], especially
in young or ill-appearing infants [
19
,
20
]. Bone marrow disorders, including leukemia or
myelodysplastic syndrome, were found in 1.5–2.5% of previously healthy, infected children
with febrile neutropenia, and all of them showed chronic neutropenia >2 months
[6,21]
.
Given the safety concerns, a diagnostic work-up of underlying serious etiologies is neces-
sary for RSV-infected infants with febrile or prolonged neutropenia. Prophylactic antibiotic
use may then be withheld from RSV-infected infants with afebrile neutropenia.
Viruses 2021,13, 301 7 of 10
4.2. Age-Related Prevalence of Neutropenia
Normal values for ANC vary by age and ethnicity [8]. Except for the first two weeks
after birth, 1.0
×
10
9
/L is used as the lower limit of normal for ANC during the first year of
life, with 1.5
×
10
9
/L thereafter [
8
]. Benign ethnic neutropenia has been described in black
individuals [
8
,
15
], but no such individuals were present in this study. In our study, the
cumulative incidence of neutropenia (<1.0
×
10
9
/L) was 11% among RSV-infected infants,
higher than <5% among infants from the general population [
8
]. Febrile neutropenia
was reported as a serious adverse event in the clinical trial of lumicitabine, a nucleoside
analogue against RSV polymerase, in RSV-infected children <3 years old [
5
]. However,
neutropenic complications have not been reported in adult clinical trials of lumicitabine or
other nucleoside polymerase inhibitors, such as PC786 or EDP-938 [22,23].
4.3. Inflammatory Effect of Airway Microbiota in RSV Infection
In the present study, transient neutropenia occurring in infants with RSV infection
showed an association with nasopharyngeal microbiota, including M. catarrhalis,S. pneumo-
niae, or H. influenzae. Both ANC and C-reactive protein levels were higher in RSV-infected
infants with such microbiota than in those without. These results indicate that nasopha-
ryngeal microbiota profile can potentially induce systemic inflammation and neutrophil
recruitment. The effects of the airway microbiota profile on inflammatory response and on
the severity of RSV infection have been studied. Nasopharyngeal microbiota comprising
H. influenzae or Streptococcus have shown correlations with overexpression of inflammation-
related genes and the need for hospitalization in RSV-infected children <2 years old [
24
].
In addition, nasopharyngeal microbiota with H. influenzae was associated with delayed
clearance of RSV and the need for intensive care and for prolonged hospitalization in RSV-
infected infants [
14
,
25
]. Airway microbiota, including Moraxella or Haemophilus, evoked
the release of cytokines and chemokines among non-infected healthy neonates and among
RSV-infected infants, contributing to the development of recurrent wheezing until 3 years
of age [26,27].
4.4. Potential Role of Airway Microbiota Composition in Neutropenic Phenomenon
Our results demonstrated that nasopharyngeal microbiota composition without M.
catarrhalis,S. pneumoniae, or H. influenzae correlated with increased incidence of tran-
sient neutropenia and was not correlated with the severity of RSV infection in infants.
This suggests that decreased circulating neutrophil counts do not necessarily imply im-
munosuppression. RSV can be detected in peripheral blood in children, and infects bone
marrow stromal cells in children and adults [
28
,
29
]. RSV infection in infants also enhances
the release of hematopoietic cytokines, inflammatory cytokines, and proinflammatory
chemokines in nasal lavage fluids [
30
]. The release of these proinflammatory cytokines and
chemokines induced by RSV infection has an effect on the recruitment and the migration
of neutrophils to inflammatory sites. Decreased circulating neutrophils in the peripheral
blood of infants with RSV infection are therefore considered to result from facilitated trans-
migration and extravasation [
31
], in addition to the effect of transient myelosuppression
or neutrophil apoptosis. This postulated mechanism is consistent with the infrequent
occurrence of serious bacterial infections among immunocompetent children with febrile
neutropenia [
7
,
18
]. Transient neutropenia in RSV-infected infants has been reported to per-
sist for approximately 30 days [
6
]. Further research is required to reveal the pathogenesis
of transient neutropenia in infants with RSV infection.
4.5. Limitations
This type of retrospective observational study has inherent limitations. First, the
timing and the frequency of collecting blood samples within 10 days of illness were not
the same for all participants. This may underestimate the incidence of benign neutropenia
in RSV-infected infants. Second, 32% of patients did not complete follow-up assessment
in this study, and missing data were not included in the analyses. We could not confirm
Viruses 2021,13, 301 8 of 10
the recovery of neutropenia in all RSV-infected infants with neutropenia. Duration of
neutropenia was thus not analyzed in this study. Missed diagnoses of serious infection,
bone marrow disorder, and malignancy following neutropenia in RSV-infected infants were
presumed to be rare, because all children requiring emergency care or hospital admission
in this city were transported to our center. Third, subtypes of RSV were not identified in
the present study. The subtype of RSV has an effect on disease severity, and possibly on
ANC, through the enhanced release of proinflammatory cytokines and chemokines [
32
].
Finally, 80% of infants included in this study showed mild severity of RSV infection (GRSS
3.5) [
10
]. Our study results have limited generalizability to infants with severe RSV
infection or with non-Asian ethnicity.
5. Conclusions
Among infants hospitalized due to predominantly mild RSV infection, 11% showed
transient neutropenia <1.0
×
10
9
/L within 10 days of onset. However, no development of
serious disease was observed in the neutropenic infants. Age at onset and nasopharyngeal
microbiota profile were associated with the development of transient neutropenia. These
risk factors for benign neutropenia in RSV-infected infants should thus be considered in
clinical practice and clinical trials for RSV antivirals. Further investigation is needed to
determine the pathogenesis and the optimal management of transient neutropenia among
children with RSV infection.
Author Contributions:
Conceptualization: T.K. and H.K.; methodology: T.K. and H.K.; formal
analysis: H.K.; data curation: T.K.; original draft preparation: T.K.; review and editing: H.K. All
authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement:
The study was conducted according to the STROBE state-
ment, and approved by the Institutional Ethics Committee of Beppu Medical Center (BMC2019-015).
Informed Consent Statement:
Passive informed consent was obtained from the parents of partici-
pants through the hospital website and bulletin board.
Data Availability Statement: Not applicable.
Conflicts of Interest:
This research did not receive any specific grant from funding agencies in the
public, commercial, or not-for-profit sectors.
Abbreviations
ANC absolute neutrophil count
GRSS global respiratory severity score
RSV respiratory syncytial virus
References
1.
Griffiths, C.; Drews, S.J.; Marchant, D.J. Respiratory Syncytial Virus: Infection, Detection, and New Options for Prevention and
Treatment. Clin. Microbiol. Rev. 2017,30, 277–319. [CrossRef] [PubMed]
2.
Shi, T.; McAllister, D.A.; O’Brien, K.L.; Simoes, E.A.F.; Madhi, S.A.; Gessner, B.D.; Polack, F.P.; Balsells, E.; Acacio, S.;
Aguayo, C.; et al
. Global, regional, and national disease burden estimates of acute lower respiratory infections due to res-
piratory syncytial virus in young children in 2015: A systematic review and modelling study. Lancet
2017
,390, 946–958. [CrossRef]
3.
Xing, Y.; Proesmans, M. New therapies for acute RSV infections: Where are we? Eur. J. Pediatr.
2019
,178, 131–138. [CrossRef]
[PubMed]
4.
DeVincenzo, J.P.; McClure, M.W.; Symons, J.A.; Fathi, H.; Westland, C.; Chanda, S.; Lambkin-Williams, R.; Smith, P.;
Zhang, Q.
;
Beigelman, L.; et al. Activity of Oral ALS-008176 in a Respiratory Syncytial Virus Challenge Study. N. Engl. J. Med.
2015
,
373 2048–2058. [CrossRef]
5.
ClinicalTrials.gov. A Study to Evaluate the Antiviral Activity, Clinical Outcomes, Safety, Tolerability, and Pharmacokinetics of
Orally Administered Lumicitabine (JNJ-64041575) Regimens in Hospitalized Infants and Children Aged 28 Days to 36 Months
Infected with Respiratory Syncytial Virus. Available online: https://ClinicalTrials.gov/show/NCT03333317 (accessed on 7
July 2020).
Viruses 2021,13, 301 9 of 10
6.
Alexandropoulou, O.; Kossiva, L.; Haliotis, F.; Giannaki, M.; Tsolia, M.; Panagiotou, I.P.; Karavanaki, K. Transient neutropenia in
children with febrile illness and associated infectious agents: 2 years’ follow-up. Eur. J. Pediatr. 2013,172, 811–819. [CrossRef]
7.
Pascual, C.; Trenchs, V.; Hernandez-Bou, S.; Catala, A.; Valls, A.F.; Luaces, C. Outcomes and infectious etiologies of febrile
neutropenia in non-immunocompromised children who present in an emergency department. Eur. J. Clin. Microbiol. Infect Dis.
2016,35, 1667–1672. [CrossRef] [PubMed]
8. Segel, G.B.; Halterman, J.S. Neutropenia in pediatric practice. Pediatr. Rev. 2008,29, 12–23. [CrossRef] [PubMed]
9.
Von Elm, E.; Altman, D.G.; Egger, M.; Pocock, S.J.; Gotzsche, P.C.; Vandenbroucke, J.P.; Initiative, S. The Strengthening the
Reporting of Observational Studies in Epidemiology (STROBE) statement: Guidelines for reporting observational studies. Lancet
2007,370, 1453–1457. [CrossRef]
10.
Caserta, M.T.; Qiu, X.; Tesini, B.; Wang, L.; Murphy, A.; Corbett, A.; Topham, D.J.; Falsey, A.R.; Holden-Wiltse, J.; Walsh, E.E.
Development of a Global Respiratory Severity Score for Respiratory Syncytial Virus Infection in Infants. J. Infect Dis.
2017
,
215, 750–756. [CrossRef]
11.
Siefker, D.T.; Vu, L.; You, D.; McBride, A.; Taylor, R.; Jones, T.L.; DeVincenzo, J.; Cormier, S.A. Respiratory Syncytial Virus Disease
Severity Is Associated with Distinct CD8(+) T-Cell Profiles. Am. J. Respir. Crit. Care Med. 2020,201, 325–334. [CrossRef]
12.
World Health Organization. WHO Global Database on Child Growth and Malnutrition. The WHO Anthro Survey Analyser.
Available online: https://www.who.int/nutgrowthdb/software/en/ (accessed on 7 July 2020).
13.
Goldstein, B.; Giroir, B.; Randolph, A.; International Consensus Conference on Pediatric Sepsis. International pediatric sepsis
consensus conference: Definitions for sepsis and organ dysfunction in pediatrics. Pediatr. Crit. Care Med. 2005,6, 2–8.
14.
Mansbach, J.M.; Hasegawa, K.; Piedra, P.A.; Avadhanula, V.; Petrosino, J.F.; Sullivan, A.F.; Espinola, J.A.; Camargo, C.A.
Haemophilus-Dominant Nasopharyngeal Microbiota Is Associated with Delayed Clearance of Respiratory Syncytial Virus in
Infants Hospitalized for Bronchiolitis. J. Infect Dis. 2019,219, 1804–1808. [CrossRef]
15.
Hsieh, M.M.; Everhart, J.E.; Byrd-Holt, D.D.; Tisdale, J.F.; Rodgers, G.P. Prevalence of neutropenia in the U.S. population: Age,
sex, smoking status, and ethnic differences. Ann. Intern. Med. 2007,146, 486–492. [CrossRef] [PubMed]
16.
Mahajan, P.; Browne, L.R.; Levine, D.A.; Cohen, D.M.; Gattu, R.; Linakis, J.G.; Anders, J.; Borgialli, D.; Vitale, M.; Dayan, P.S.; et al.
Risk of Bacterial Coinfections in Febrile Infants 60 Days Old and Younger with Documented Viral Infections. J. Pediatr.
2018
,
203, 86–91.e2. [PubMed]
17.
Byington, C.L.; Enriquez, F.R.; Hoff, C.; Tuohy, R.; Taggart, E.W.; Hillyard, D.R.; Carroll, K.C.; Christenson, J.C. Serious bacterial
infections in febrile infants 1 to 90 days old with and without viral infections. Pediatrics 2004,113, 1662–1666. [CrossRef]
18.
Barg, A.A.; Kozer, E.; Mordish, Y.; Lazarovitch, T.; Kventsel, I.; Goldman, M. The Risk of Serious Bacterial Infection in Neutropenic
Immunocompetent Febrile Children. J. Pediatr. Hematol. Oncol. 2015,37, e347–e351. [CrossRef] [PubMed]
19.
Titus, M.O.; Wright, S.W. Prevalence of serious bacterial infections in febrile infants with respiratory syncytial virus infection.
Pediatrics 2003,112, 282–284.
20.
Levine, D.A.; Platt, S.L.; Dayan, P.S.; Macias, C.G.; Zorc, J.J.; Krief, W.; Schor, J.; Bank, D.; Fefferman, N.; Shaw, K.N.; et al. Risk
of serious bacterial infection in young febrile infants with respiratory syncytial virus infections. Pediatrics
2004
,113, 1728–1734.
[CrossRef] [PubMed]
21.
Karavanaki, K.; Polychronopoulou, S.; Giannaki, M.; Haliotis, F.; Sider, B.; Brisimitzi, M.; Dimitriou, C.; Scordias, G.;
Marangou, F.
;
Stamatiadou, A.; et al. Transient and chronic neutropenias detected in children with different viral and bacterial infections.
Acta Paediatr. 2006,95, 565–572. [CrossRef] [PubMed]
22.
Cass, L.; Davis, A.; Murray, A.; Woodward, K.; Ito, K.; Strong, P.; Rapeport, G. 1335. Safety and Pharmacokinetic Profile of PC786,
a Novel Inhibitor of Respiratory Syncytial Virus L-protein Polymerase, in a Single and Multiple-Ascending Dose Study in Healthy
Volunteer and Mild Asthmatics. Open Forum Infect Dis. 2018,5(Suppl. S1), S407–S408. [CrossRef]
23.
Coakley, E.; Ahmad, A.; Larson, K.; McClure, T.; Lin, K.; Lin, K.; Tenhoor, K.; Eze, K.; Noulin, N.; Horvathova, V.; et al. LB6.
EDP-938, a Novel RSV N-Inhibitor, Administered Once or Twice Daily Was Safe and Demonstrated Robust Antiviral and Clinical
Efficacy in a Healthy Volunteer Challenge Study. Open Forum Infect Dis. 2019,6(Suppl. S2), S995. [CrossRef]
24.
De Steenhuijsen Piters, W.A.; Heinonen, S.; Hasrat, R.; Bunsow, E.; Smith, B.; Suarez-Arrabal, M.C.; Chaussabel, D.; Cohen,
D.M.; Sanders, E.A.; Ramilo, O.; et al. Nasopharyngeal Microbiota, Host Transcriptome, and Disease Severity in Children with
Respiratory Syncytial Virus Infection. Am. J. Respir. Crit. Care Med. 2016,194, 1104–1115. [CrossRef] [PubMed]
25.
Hasegawa, K.; Mansbach, J.M.; Ajami, N.J.; Espinola, J.A.; Henke, D.M.; Petrosino, J.F.; Piedra, P.A.; Shaw, C.A.; Sullivan, A.F.;
Camargo, C.A., Jr.; et al. Association of nasopharyngeal microbiota profiles with bronchiolitis severity in infants hospitalised for
bronchiolitis. Eur. Respir. J. 2016,48, 1329–1339. [CrossRef] [PubMed]
26.
Folsgaard, N.V.; Schjorring, S.; Chawes, B.L.; Rasmussen, M.A.; Krogfelt, K.A.; Brix, S.; Bisgaard, H. Pathogenic bacteria colonizing
the airways in asymptomatic neonates stimulates topical inflammatory mediator release. Am. J. Respir. Crit. Care Med.
2013
,
187, 589–595. [CrossRef] [PubMed]
27.
Zhang, X.; Zhang, X.; Zhang, N.; Wang, X.; Sun, L.; Chen, N.; Zhao, S.; He, Q. Airway microbiome, host immune response and
recurrent wheezing in infants with severe respiratory syncytial virus bronchiolitis. Pediatr. Allergy Immunol.
2020
,31, 281–289.
[CrossRef] [PubMed]
28.
O’Donnell, D.R.; McGarvey, M.J.; Tully, J.M.; Balfour-Lynn, I.M.; Openshaw, P.J. Respiratory syncytial virus RNA in cells from the
peripheral blood during acute infection. J. Pediatr. 1998,133, 272–274. [CrossRef]
Viruses 2021,13, 301 10 of 10
29.
Rezaee, F.; Gibson, L.F.; Piktel, D.; Othumpangat, S.; Piedimonte, G. Respiratory syncytial virus infection in human bone marrow
stromal cells. Am. J. Respir. Cell Mol. Biol. 2011,45, 277–286. [CrossRef]
30.
Bennett, B.L.; Garofalo, R.P.; Cron, S.G.; Hosakote, Y.M.; Atmar, R.L.; Macias, C.G.; Piedra, P.A. Immunopathogenesis of
respiratory syncytial virus bronchiolitis. J. Infect Dis. 2007,195, 1532–1540.
31.
Russell, C.D.; Unger, S.A.; Walton, M.; Schwarze, J. The Human Immune Response to Respiratory Syncytial Virus Infection.
Clin. Microbiol. Rev. 2017,30, 481–502. [CrossRef]
32.
McConnochie, K.M.; Hall, C.B.; Walsh, E.E.; Roghmann, K.J. Variation in severity of respiratory syncytial virus infections with
subtype. J. Pediatr. 1990,117, 52–62. [CrossRef]
... We conducted an extensive analysis of patients with adenovirus pneumonia and observed that there was no significant difference in the average LOS and hospitalization costs between children with complications such as myocardial damage, abnormal liver function, and diarrhea, and those without any complications. This could be attributed to the fact that the aforementioned complications often cause transient damage (Korematsu and Koga 2021;Thorburn et al. 2018), which do not impact the duration of hospitalization or treatment costs. However, we noted that children who experienced respiratory failure had higher LOS and hospitalization costs compared to those with other complications. ...
Article
Full-text available
Adenovirus pneumonia is a prevalent form of community-acquired pneumonia among children. Research on the epidemiology and economic burden of this disease is crucial for public health, yet comprehensive data remains scarce, making it crucial to highlight on this topic. In this study, the data were extracted from the face sheet of discharge medical records collected from 26 tertiary children’s hospitals from January 2016 to December 2021. In total, 1854 children with laboratory-confirmed adenovirus pneumonia were hospitalized, accounting for 0.13% of the total number of hospitalized for pneumonia in the database during the period. In addition, this figure represents a meager 0.027% when compared to the total number of hospitalized children. The male-to-female ratio was 1.78:1. The 1–3-year age group had the highest number of inpatients for adenoviral pneumonia and the largest proportion of the total hospitalizations in the same age group. Overall, winter is the primary season for the prevalence of adenovirus pneumonia, however, in southern China, there are two peak seasons, winter and summer. Although patients with 3/4 adenovirus pneumonia had no significant complications, some patients had complications such as respiratory failure, diarrhea, and myocardial damage. The median length of stay of adenovirus pneumonia was 8 d [interquartile range (IQR) 6–11], and the median hospitalization cost was 1293.83 United States dollars (IQR 811.81–2472.51). These valuable epidemiological insights into adenovirus pneumonia in Chinese children can help direct the development of targeted prevention and control strategies and surveillance measures for HAdV infections in this demographic.
... The results for the mean LOS and hospitalization expenditure of patients with bronchiolitis showed that although LOS was comparable between children with and without complications, children without complications had lower hospitalization costs. As mentioned above, complications such as myocardial damage, abnormal liver function, and neutropenia are mostly transient and therefore do not prolong LOS [28,32]. However, patients with abnormal liver function had the highest LOS and expenditures. ...
Article
Full-text available
Background: Bronchiolitis is a common acute lower respiratory tract infection (ALRTI) and the most frequent cause of hospitalization of infants and young children with ALRTI. Respiratory syncytial virus is the main pathogen that leads to severe bronchiolitis. The disease burden is relatively high. To date, few descriptions of the clinical epidemiology and disease burden of children hospitalized for bronchiolitis are available. This study reports the general clinical epidemiological characteristics and disease burden of bronchiolitis in hospitalized children in China. Methods: This study included the face sheet of discharge medical records collected from 27 tertiary children's hospitals from January 2016 to December 2020 that were aggregated into the FUTang Update medical REcords (FUTURE) database. The sociodemographic variables, length of stay (LOS) and disease burden of children with bronchiolitis were analyzed and compared using appropriate statistical tests. Results: In total, 42,928 children aged 0-3 years were hospitalized due to bronchiolitis from January 2016 to December 2020, accounting for 1.5% of the total number of hospitalized children of the same age in the database during the period and 5.31% of the hospitalizations for ALRTI. The male to female ratio was 2.01:1. Meanwhile, more boys than girls were observed in different regions, age groups, years, and residences. The 1-2 year age group had the greatest number of hospitalizations for bronchiolitis, while the 29 days-6 months group had the largest proportion of the total inpatients and inpatients with ALRTI in the same age group. In terms of region, the hospitalization rate of bronchiolitis was the highest in East China. Overall, the number of hospitalizations from 2017 to 2020 showed a decreasing trend from that in 2016. Seasonally, the peak hospitalizations for bronchiolitis occurred in winter. Hospitalization rates in North China in autumn and winter were higher than those in South China, while hospitalization rates in South China were higher in spring and summer. Approximately, half of the patients with bronchiolitis had no complications. Among the complications, myocardial injury, abnormal liver function and diarrhea were more common. The median LOS was 6 days [interquartile range (IQR) = 5-8], and the median hospitalization cost was 758 United States dollars (IQR = 601.96-1029.53). Conclusions: Bronchiolitis is a common respiratory disease in infants and young children in China, and it accounts for a higher proportion of both total hospitalizations and hospitalizations due to ALRTI in children. Among them, children aged 29 days-2 years are the main hospitalized population, and the hospitalization rate of boys is significantly higher than that of girls. The peak season for bronchiolitis is winter. Bronchiolitis causes few complications and has a low mortality rate, but the burden of this disease is heavy.
... Recent nasal immune profiling of infants with RSV bronchiolitis showed increased levels of IL-8 [105], and global mRNA expression showed increased neutrophil signatures in severe vs. mild RSV infection in infants [106]. Further, transient neutropenia due to young age was not a risk factor for immunocompetent infants to develop serious RSV disease [107]. TLR-2 is also involved in pulmonary neutrophil during early infection (24 hpi) linked to the expression of CCL2 [108] which may be in part driven by RSV G protein [109]. ...
Article
Full-text available
RSV is a leading cause of respiratory tract disease in infants and the elderly. RSV has limited therapeutic interventions and no FDA-approved vaccine. Gaps in our understanding of virus–host interactions and immunity contribute to the lack of biological countermeasures. This review updates the current understanding of RSV immunity and immunopathology with a focus on interferon responses, animal modeling, and correlates of protection.
Article
Background and objective Serum procalcitonin ( PCT ) is a highly accurate biomarker for stratifying the risk of invasive bacterial infections ( IBIs ) in febrile infants ≤60 days old. However, PCT is unavailable in some settings. We explored the association of leukopenia and neutropenia with IBIs in non–critically ill febrile infants ≤60 days old, with and without PCT . Methods We conducted a secondary analysis of a prospective observational cohort consisting of 7407 non–critically ill infants ≤60 days old with temperatures ≥38°C. We focused on the risk of IBIs in patients with leukopenia (white blood cell [WBC] count <5000 cells/μL) or neutropenia (absolute neutrophil count [ANC] <1000 cells/μL), categorized to extremes of lower values, and the impact of PCT on these associations. Multiple logistic regression was used to identify independent predictors of IBIs. Results Final analysis included 6865 infants with complete data; 45% (3098) had PCT data available. Of the 6865, a total of 111 (1.6%) had bacteremia without bacterial meningitis, 18 (0.3%) had bacterial meningitis without bacteremia, and 19 (0.3%) had both bacteremia and bacterial meningitis. IBI was present in four of 20 (20%) infants with WBC counts ≤2500 cells/μL and four of 311 (1.3%) with ANC <1000 cells/μL. In multivariable logistic regression analysis not including PCT, a WBC count <2500 cells/μL was significantly associated with IBI (OR 13.48, 95% CI 2.92–45.35). However, no patients with leukopenia or neutropenia and PCT ≤0.5 ng/mL had IBIs. Conclusions Leukopenia ≤2500 cells/μL in febrile infants ≤60 days old is associated with IBIs. However, in the presence of normal PCT levels, no patients with leukopenia had IBIs. While this suggests leukopenia ≤2500 cells/μL is a risk factor for IBIs in non–critically ill young febrile infants only when PCT is unavailable or elevated, the overall low frequency of leukopenia in this cohort warrants caution in interpretation, with future validation required.
Article
Background Despite reassuring clinical safety data, thrombocytosis, anemia, lymphopenia, and liver function derangements have been observed in infants born to women with inflammatory bowel disease (IBD) treated with thiopurines and biologics. We aimed to define the prevalence, course, associations, and clinical impact of hematological and biochemical abnormalities in such infants. Methods This multicenter prospective cohort study assessed clinical, hematologic, and biochemical outcomes of infants exposed to thiopurines or biologics in utero for management of maternal IBD. Liver transaminases, full blood examination, and infant thiopurine metabolites (where exposed) were taken at delivery and 6 weeks of age. Abnormal results were repeated until normalization. Infants were followed clinically by a pediatric gastroenterologist up to 2 years of age. Results A total of 130 infants were included. Thrombocytosis and elevated alanine transaminase (ALT) were seen in over half of infants up to 6 months of age with no significant clinical impact. Elevated ALT was associated with increasing maternal C-reactive protein in second trimester, while thrombocytosis was associated with increasing maternal C-reactive protein and fecal calprotectin in third trimester. Preceding infection and vaccination were associated with an increased risk of elevated alkaline phosphatase at 3 months. In those exposed to thiopurines, increasing maternal 6-methylmercaptopurine at delivery was associated with increased ALT to 6 months. Conclusions Infants born to women with IBD commonly developed thrombocytosis, elevated alkaline phosphatase, and elevated ALT. These findings were associated with exposure to maternal inflammation, elevated 6-methylmercaptopurine at delivery, and infant vaccinations and infections, and had minimal clinical consequence.
Article
Introduction: Respiratory syncytial virus (RSV) is a common respiratory virus with a huge impact on patients, the healthcare system and society worldwide. Very few effective chances of prevention and treatment of RSV infection are available. Areas covered: In this paper, knowledge on RSV characteristics and current stage of development of new pharmacological measures against this virus are discussed. Expert opinion: In recent years the structure of RSV was explored in depth and several pharmacologic measures potentially effective for prevention and treatment of RSV infection and disease were identified. These new measures have the aim to overcome the limitations of palivizumab and ribavirin. Strategies to protect infants through immunization of pregnant women and/or the use of more effective monoclonal antibodies were developed. At the same time, it was defined which vaccines could be administered to unprimed infants to avoid the risk of enhanced respiratory disease and which vaccines could be effective in older patients and in subjects with reduced immune system efficiency. Finally, a great number of new antiviral drugs targeting the RSV proteins that allow RSV entering host cells or regulate virus replication were produced. Although further studies are needed, some preparations seem effective and safe, making the future of RSV infection prevention and treatment less gloomy.
Article
Full-text available
Background Early interactions between respiratory viruses and microbiota might modulate host immune responses and subsequently contribute to later development of recurrent wheezing and asthma in childhood. We aimed to study the possible association between respiratory microbiome, host immune response, and the development of recurrent wheezing in infants with severe respiratory syncytial virus (RSV) bronchiolitis. Methods Seventy‐four infants who were hospitalized at Beijing Children's Hospital during an initial episode of severe RSV bronchiolitis at 6 months of age or less were included and followed up until the age of 3 years. Sputum samples were collected, and their microbiota profiles, LPS, and cytokines were analyzed by 16S rRNA–based sequencing, ELISA, and multiplex immunoassay, respectively. Results Twenty‐six (35.1%) infants developed recurrent wheezing by the age of 3 years, and 48 (64.9%) did not. The relative abundance of Haemophilus, Moraxella, and Klebsiella was higher in infants who later developed recurrent wheezing than in those who did not (LDA score >3.5). Airway levels of LPS (P = .003), CXCL8 (P = .004), CCL5 (P = .029), IL‐6 (P = .004), and IL‐13 (P < .001) were significantly higher in infants who later developed recurrent wheezing than in those who did not. Moreover, high airway abundance of Haemophilus was associated with CXCL8 (r = 0.246, P = .037) level, and that of Moraxella was associated with IL‐6 level (r = 0.236, P = .046) and IL‐10 level (r = 0.266, P = .024). Conclusion Our study suggests that higher abundance of Haemophilus and Moraxella in airway microbiome might modulate airway inflammation during severe RSV bronchiolitis in infancy, potentially contributing to the development of subsequent recurrent wheezing in later childhood.
Article
Full-text available
Background Respiratory syncytial virus (RSV) represents an important global health challenge with significant morbidity and mortality in infants, elderly, and immunocompromised adults. No effective therapy is currently available. EDP-938 demonstrates potent in vitro activity against RSV Subtypes A and B. We report data from EDP 938-101, a double-blind, placebo-controlled, Phase 2a study that evaluated EDP-938 in adult volunteers inoculated with RSV-A Memphis 37b. Methods Subjects were healthy volunteers, 18–45 years, who were sero-suitable (i.e., lower 25th percentile). After RSV inoculation on Study Day 0, subjects had 12 hourly nasal wash monitored for RSV infection by qualitative RSV RT–PCR. On Study Day 5 or previously if qualitative RT–PCR was RSV+, subjects were randomized to receive 5 days of EDP-938 600 mg once daily (QD arm) or 500 mg loading dose then 300 mg twice daily (BID arm), or placebo twice daily. Assessments included 12 hourly nasal wash for quantitative RSV viral load, 8 hourly RSV Total Symptom Scoring (TSS) and daily mucus weights. Safety assessments were continued though Day 28 (last follow-up). The primary endpoint was the RSV viral load area under the curve (AUC) from first dose through Day 12 among RSV-infected subjects, defined as the Intent To Treat-Infected (ITT-I) population. The study was fully powered for both RSV viral load and TSS endpoints. Results A total of 115 subjects were randomized and inoculated; 86 were included in the ITT-I analysis. The primary and secondary efficacy endpoints were achieved with high statistical significance in QD and BID arms (figure and table). Among EDP-938 recipients all adverse events (AEs) were mild except for a single AE of moderate dyspepsia in the BID arm and events of moderate headache (n = 2) and hypoacusis (n = 1) in the placebo arm. All AEs resolved in follow-up. Conclusions In the RSV Challenge study, EDP-938 administered once or twice daily achieved primary and key secondary endpoints with robust reductions in RSV viral load (by both qRT–PCR and plaque assays), symptom scores and mucus weights. These data support the further clinical evaluation of EDP-938 in populations at risk of severe RSV disease. Disclosures Eoin Coakley, MD, Enanta Pharmaceuticals (Employee), Alaa Ahmad, PhD, Enanta Pharmaceuticals (Employee), Kajal Larson, PhD, Enanta Pharmaceuticals (Employee), Ty McClure, PhD, Enanta Pharmaceuticals (Employee), Kai Lin, PhD, Enanta Pharmaceuticals (Employee), Kursten Tenhoor, n/a, Enanta Pharmaceuticals (Consultant), Kingsley Eze, n/a, hVIVO Services Ltd. (Employee), Nicolas Noulin, PhD, No financial relationships or conflicts of interest, Veronika Horvathova, MBChB, MSc, hVIVO Services Limited (Other Financial or Material Support, Employed by hVIVO during the conduct of the study), Bryan Murray, MBBS, No financial relationships or conflicts of interest, Mark Baillet, PhD, S-CUBED (Employee), Julie Mori, PhD, hVIVO (Employee, Shareholder) Nathalie Adda, MD, Enanta Pharmaceuticals (Employee).
Article
Full-text available
Respiratory syncytial virus (RSV) infection is one of the main causes of infant hospitalization and mortality. The single-stranded RNA virus codes for 11 proteins of which the F protein, a surface epitope responsible for RSV fusion, is the most targeted for developing antiviral medicines and vaccines. The peak of symptoms occurs around day 4 to 6 of illness and the airway obstruction is merely caused by the host immune inflammatory response. Risk factors for severe bronchiolitis are prematurity, comorbidity, and/or being immunocompromised. At present, there are no curative therapies available for RSV infections and treatment is supportive only. Development of new antiviral medicines is however promising. The aim of this review is to give a summary of the most important new antiviral therapies in clinical development for RSV infection and to explain their mode of action. We therefore performed a literature search on this topic.Conclusion: There are currently at least eight antivirals being investigated in clinical trials. They all use different approaches to either focus on preventing viral fusion with host cells or inhibiting virus replication. Some target RSV surface epitopes like the F protein to halt fusion, others aim for RNA chain termination, while small interfering RNAs downregulate viral protein production. What is known: • RSV bronchiolitis is a very important pediatric disease as it is one of the main causes of infant hospitalization and mortality. By the age of 2 years, 95% of all the infants worldwide will have been infected. • The only recommended therapy is supportive since there are no existing curative therapies yet. What this study adds: • This review gives an overview of the current progress in the research field of RSV antivirals with background information on their mode of action.
Article
Full-text available
Background RSV is the most common cause of bronchiolitis in infants and is responsible for severe respiratory infections in the elderly and immunocompromised populations. RSV replicates in the columnar epithelial cells of the proximal and distal airways which are accessible to inhaled therapies. PC786 is a potent non-nucleoside RSV L-protein polymerase inhibitor designed for inhaled delivery. In preclinical studies, PC786 exhibits prolonged lung tissue residence with minimal systemic exposure, thus limiting the potential for adverse systemic effects. Methods A phase 1 study was conducted to evaluate the safety and pharmacokinetics of PC786 delivered in a suspension formulation by nebulizer (PARI LC SPRINT® device). Healthy volunteers (HVs) received placebo or PC786 as single ascending doses (0.5–20 mg, Cohort (C) 1), 5 mg BD for 7 days (C2), or 10 mg BD for 7 days (C3). Mild asthmatics received a single dose of PC786 5 mg or placebo (C4). PC786 PK was measured in plasma and in nasal mucosal lining fluid (MLF) collected using a synthetic absorptive matrix. Results PC786 was well tolerated, with no significant adverse clinical nor laboratory findings. Following single inhaled doses PC786 appeared rapidly in the plasma; mean plasma Cmax of 190, 571, 1,760, and 3,270 pg/mL, for the 0.5, 2, 8, and 20 mg doses, respectively, were measured on average at 0.68 to 0.93 hours (Tmax) post-inhalation. Following administration of 5 mg BD (C2) the extent of accumulation was approximately 2-fold. The geometric mean apparent terminal half-life measured following 10 mg BD (C3) was 97 hours. The ratio of MLF:plasma concentrations ranged from 6,347 (+2 hours) to 1,050 (+24h). Conclusion PC786 was well tolerated by HVs and asthmatics. The compound showed a rapid Tmax, suggesting rapid exposure of the respiratory epithelium. The PC786 concentrations in MLF exceed the IC90 for RSV, but circulating plasma concentrations were low. The MLF:plasma measured in this study was consistent with lung:plasma ratios measured in preclinical studies. The long plasma half-life is consistent with slow absorption from the lung being the dominant process controlling systemic kinetic behavior. The long t½ and 2-fold accumulation ratio observed on repeat dosing supports once daily dosing in subsequent studies. Disclosures L. Cass, Pulmocide Ltd.: Employee and Shareholder, Salary. A. Davis, Pulmocide Ltd.: Employee and Shareholder, Salary. A. Murray, Pulmocide Ltd.: Employee and Shareholder, Salary. K. Woodward, Pulmocide Ltd.: Consultant, Consulting fee. K. Ito, Pulmocide Ltd.: Employee and Shareholder, Salary. P. Strong, Pulmocide Ltd.: Board Member, Employee and Shareholder, Salary. G. Rapeport, Pulmocide Ltd.: Board Member, Employee and Shareholder, Salary.
Article
Full-text available
Background: We have previously estimated that respiratory syncytial virus (RSV) was associated with 22% of all episodes of (severe) acute lower respiratory infection (ALRI) resulting in 55 000 to 199 000 deaths in children younger than 5 years in 2005. In the past 5 years, major research activity on RSV has yielded substantial new data from developing countries. With a considerably expanded dataset from a large international collaboration, we aimed to estimate the global incidence, hospital admission rate, and mortality from RSV-ALRI episodes in young children in 2015. Methods: We estimated the incidence and hospital admission rate of RSV-associated ALRI (RSV-ALRI) in children younger than 5 years stratified by age and World Bank income regions from a systematic review of studies published between Jan 1, 1995, and Dec 31, 2016, and unpublished data from 76 high quality population-based studies. We estimated the RSV-ALRI incidence for 132 developing countries using a risk factor-based model and 2015 population estimates. We estimated the in-hospital RSV-ALRI mortality by combining in-hospital case fatality ratios with hospital admission estimates from hospital-based (published and unpublished) studies. We also estimated overall RSV-ALRI mortality by identifying studies reporting monthly data for ALRI mortality in the community and RSV activity. Findings: We estimated that globally in 2015, 33·1 million (uncertainty range [UR] 21·6-50·3) episodes of RSV-ALRI, resulted in about 3·2 million (2·7-3·8) hospital admissions, and 59 600 (48 000-74 500) in-hospital deaths in children younger than 5 years. In children younger than 6 months, 1·4 million (UR 1·2-1·7) hospital admissions, and 27 300 (UR 20 700-36 200) in-hospital deaths were due to RSV-ALRI. We also estimated that the overall RSV-ALRI mortality could be as high as 118 200 (UR 94 600-149 400). Incidence and mortality varied substantially from year to year in any given population. Interpretation: Globally, RSV is a common cause of childhood ALRI and a major cause of hospital admissions in young children, resulting in a substantial burden on health-care services. About 45% of hospital admissions and in-hospital deaths due to RSV-ALRI occur in children younger than 6 months. An effective maternal RSV vaccine or monoclonal antibody could have a substantial effect on disease burden in this age group. Funding: The Bill & Melinda Gates Foundation.
Article
Full-text available
Respiratory syncytial virus (RSV) is an important etiological agent of respiratory infections, particularly in children. Much information regarding the immune response to RSV comes from animal models and in vitro studies. Here, we provide a comprehensive description of the human immune response to RSV infection, based on a systematic literature review of research on infected humans. There is an initial strong neutrophil response to RSV infection in humans, which is positively correlated with disease severity and mediated by interleukin-8 (IL-8). Dendritic cells migrate to the lungs as the primary antigen-presenting cell. An initial systemic T-cell lymphopenia is followed by a pulmonary CD8 ⁺ T-cell response, mediating viral clearance. Humoral immunity to reinfection is incomplete, but RSV IgG and IgA are protective. B-cell-stimulating factors derived from airway epithelium play a major role in protective antibody generation. Gamma interferon (IFN-γ) has a strongly protective role, and a Th2-biased response may be deleterious. Other cytokines (particularly IL-17A), chemokines (particularly CCL-5 and CCL-3), and local innate immune factors (including cathelicidins and IFN-λ) contribute to pathogenesis. In summary, neutrophilic inflammation is incriminated as a harmful response, whereas CD8 ⁺ T cells and IFN-γ have protective roles. These may represent important therapeutic targets to modulate the immunopathogenesis of RSV infection.
Article
Rationale: Respiratory syncytial virus (RSV) presents causes significant morbidity and mortality in infants worldwide. Although T helper type 2 (Th2) pathology is implicated in severe disease, the mechanisms underlying the development of immunopathology are incompletely understood. Objectives: We aimed to identify local immune responses associated with severe respiratory syncytial virus disease in infants. Our hypothesis was that disease severity would correlate with enhanced Th2 cellular responses. Methods: Nasal aspirates were collected from infants hospitalized with severe (admitted to the pediatric intensive care unit) or moderate (maintained in the general ward) RSV disease at 7 days after enrollment. The immune response was investigated by evaluating T lymphocyte cellularity, cytokine concentration, and viral load. Measurements and main results: Severe patients had increased proportions of CD8+ T cells expressing IL-4 (Tc2) and reduced proportions of CD8+ T cells expressing IFNγ (Tc1). Nasal aspirates from severe patients had reduced concentrations of IL-17. Patients with greater frequencies of Tc1, CD8+ T cells expressing IL-17 (Tc17), and CD4+ T cells expressing IL-17 (Th17) had shorter durations of hospitalization. Conclusions: Severe RSV disease was associated with distinct T cell profiles. Tc1, Tc17, and Th17 may play protective roles in RSV infection, while Tc2 cells may play a previously underappreciated role in pathology.
Article
The relation of nasopharyngeal microbiota to the clearance of respiratory syncytial virus (RSV) in infants hospitalized for bronchiolitis is not known. In a multicenter cohort, we found that 106 of 557 infants (19%) hospitalized with RSV bronchiolitis had the same RSV subtype 3 weeks later (ie, delayed clearance of RSV). Using 16S ribosomal RNA gene sequencing and a clustering approach, infants with a Haemophilus-dominant microbiota profile at hospitalization were more likely than those with a mixed profile to have delayed clearance, after adjustment for 11 factors, including viral load. Nasopharyngeal microbiota composition is associated with delayed RSV clearance.
Article
Objective: To determine the risk of serious bacterial infections (SBIs) in young febrile infants with and without viral infections. Study design: Planned secondary analyses of a prospective observational study of febrile infants 60 days of age or younger evaluated at 1 of 26 emergency departments who did not have clinical sepsis or an identifiable site of bacterial infection. We compared patient demographics, clinical, and laboratory findings, and prevalence of SBIs between virus-positive and virus-negative infants. Results: Of the 4778 enrolled infants, 2945 (61.6%) had viral testing performed, of whom 1200 (48.1%) were virus positive; 44 of the 1200 had SBIs (3.7%; 95% CI, 2.7%-4.9%). Of the 1745 virus-negative infants, 222 had SBIs (12.7%; 95% CI, 11.2%-14.4%). Rates of specific SBIs in the virus-positive group vs the virus-negative group were: UTIs (33 of 1200 [2.8%; 95% CI, 1.9%-3.8%] vs 186 of 1745 [10.7%; 95% CI, 9.2%-12.2%]) and bacteremia (9 of 1199 [0.8%; 95% CI, 0.3%-1.4%] vs 50 of 1743 [2.9%; 95% CI, 2.1%-3.8%]). The rate of bacterial meningitis tended to be lower in the virus-positive group (0.4%) than in the viral-negative group (0.8%); the difference was not statistically significant. Negative viral status (aOR, 3.2; 95% CI, 2.3-4.6), was significantly associated with SBI in multivariable analysis. Conclusions: Febrile infants ≤60 days of age with viral infections are at significantly lower, but non-negligible risk for SBIs, including bacteremia and bacterial meningitis.
Article
Respiratory syncytial virus (RSV) infection is a significant cause of hospitalization of children in North America and one of the leading causes of death of infants less than 1 year of age worldwide, second only to malaria. Despite its global impact on human health, there are relatively few therapeutic options available to prevent or treat RSV infection. Paradoxically, there is a very large volume of information that is constantly being refined on RSV replication, the mechanisms of RSV-induced pathology, and community transmission. Compounding the burden of acute RSV infections is the exacerbation of preexisting chronic airway diseases and the chronic sequelae of RSV infection. A mechanistic link is even starting to emerge between asthma and those who suffer severe RSV infection early in childhood. In this article, we discuss developments in the understanding of RSV replication, pathogenesis, diagnostics, and therapeutics. We attempt to reconcile the large body of information on RSV and why after many clinical trials there is still no efficacious RSV vaccine and few therapeutics.