Download as pdf or txt
Download as pdf or txt
You are on page 1of 50

r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

Available online at www.sciencedirect.com

Respiratory Investigation

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

Guideline

ARDS clinical practice guideline 2021*

Sadatomo Tasaka a,*, Shinichiro Ohshimo b, Muneyuki Takeuchi c,


Hideto Yasuda d, Kazuya Ichikado e, Kenji Tsushima f, Moritoki Egi g,
Satoru Hashimoto h, Nobuaki Shime i, Osamu Saito j,
Shotaro Matsumoto k, Eishu Nango l, Yohei Okada m, Kenichiro Hayashi n,
Masaaki Sakuraya o, Mikio Nakajima p, Satoshi Okamori q,
Shinya Miura r, Tatsuma Fukuda s, Tadashi Ishihara t, Tetsuro Kamo u,
Tomoaki Yatabe v, Yasuhiro Norisue w, Yoshitaka Aoki x, Yusuke Iizuka y,
Yutaka Kondo t, Chihiro Narita z, Daisuke Kawakami aa, Hiromu Okano ab,
Jun Takeshita ac, Keisuke Anan e, Satoru Robert Okazaki ad,
Shunsuke Taito ae, Takuya Hayashi af, Takuya Mayumi ag,
Takero Terayama ah, Yoshifumi Kubota ai, Yoshinobu Abe aj,
Yudai Iwasaki ak, Yuki Kishihara al, Jun Kataoka am,
Tetsuro Nishimura an, Hiroshi Yonekura ao, Koichi Ando ap,
Takuo Yoshida aq, Tomoyuki Masuyama d, Masamitsu Sanui y, ARDS
Clinical Practice Guideline Committee 2021 from the Japanese Respiratory
Society, the Japanese Society of Intensive Care Medicine, and the Japanese
Society of Respiratory Care Medicine1
a
Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Aomori, Japan
b
Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences,
Hiroshima University, Hiroshima, Japan
c
Department of Intensive Care Medicine, Osaka Women's and Children's Hospital, Osaka, Japan
d
Department of Emergency and Critical Care Medicine, Jichi Medical University, Saitama Medical Center, Saitama,
Japan
e
Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
f
International University of Health and Welfare, Tokyo, Japan
g
Department of Anesthesiology, Kobe University Hospital, Hyogo, Japan
h
Department of Anesthesiology and Intensive Care Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
i
Department of Emergency and Critical Care Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima
University, Hiroshima, Japan
j
Department of Pediatric Emergency and Critical Care Medicine, Tokyo Metropolitan Children's Medical Center,
Tokyo, Japan
k
Division of Critical Care Medicine, National Center for Child Health and Development, Tokyo, Japan
l
Department of Family Medicine, Seibo International Catholic Hospital, Tokyo, Japan

*
This article is co-published in Journal of Intensive Care (https://doi.org/10.1186/s40560-022-00615-6) and Respiratory Investigation.
* Corresponding author. Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki 036-
8562, Japan.
E-mail address: tasaka@hirosaki-u.ac.jp (S. Tasaka).
https://doi.org/10.1016/j.resinv.2022.05.003
2212-5345/© 2022 The Authors. Published by Elsevier B.V. on behalf of The Japanese Respiratory Society. This is an open access article
under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 447

m
Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto,
Japan
n
Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan
o
Department of Emergency and Intensive Care Medicine, JA Hiroshima General Hospital, Hiroshima, Japan
p
Emergency and Critical Care Center, Tokyo Metropolitan Hiroo Hospital, Tokyo, Japan
q
Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
r
Paediatric Intensive Care Unit, The Royal Children's Hospital Melbourne, Melbourne, Australia
s
Department of Emergency and Critical Care Medicine, Graduate School of Medicine, University of the Ryukyus,
Okinawa, Japan
t
Department of Emergency and Critical Care Medicine, Juntendo University, Urayasu Hospital, Chiba, Japan
u
Department of Critical Care Medicine, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
v
Department of Anesthesiology, Nishichita General Hospital, Aichi, Japan
w
Tokyo Bay Urayasu Ichikawa Medical Center, Chiba, Japan
x
Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Shizuoka,
Japan
y
Department of Anesthesiology and Critical Care Medicine, Jichi Medical University Saitama Medical Center,
Saitama, Japan
z
Department of Emergency Medicine, Shizuoka General Hospital, Shizuoka, Japan
aa
Department of Anesthesia and Critical Care, Kobe City Medical Center General Hospital, Hyogo, Japan
ab
Department of Critical Care and Emergency Medicine, National Hospital Organization Yokohama Medical Center,
Kanagawa, Japan
ac
Department of Anesthesiology, Osaka Women's and Children's Hospital, Osaka, Japan
ad
Department of Intensive Care Medicine, Kameda Medical Center, Chiba, Japan
ae
Division of Rehabilitation, Department of Clinical Practice and Support, Hiroshima University Hospital, Hiroshima,
Japan
af
Pediatric Emergency and Critical Care Center, Saitama Children's Medical Center, Saitama, Japan
ag
Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa,
Japan
ah
Department of Psychiatry, School of Medicine, National Defense Medical College, Saitama, Japan
ai
Department of Infectious Diseases, Kameda Medical Center, Chiba, Japan
aj
Division of Emergency and Disaster Medicine, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
ak
Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine,
Miyagi, Japan
al
Department of Emergency Medicine, Japanese Red Cross Musashino Hospital, Tokyo, Japan
am
Department of Critical Care Medicine, Nerima Hikarigaoka Hospital, Tokyo, Japan
an
Department of Traumatology and Critical Care Medicine, Osaka City University Graduate School of Medicine,
Osaka, Japan
ao
Department of Anesthesiology and Pain Medicine, Fujita Health University Bantane Hospital, Aichi, Japan
ap
Division of Respiratory Medicine and Allergology, Department of Medicine, Showa University School of Medicine,
Tokyo, Japan
aq
Intensive Care Unit, Department of Anesthesiology, Jikei University School of Medicine, Tokyo, Japan

article info abstract

Article history: Background: The joint committee of the Japanese Society of Intensive Care Medicine/Japa-
Received 19 April 2022 nese Respiratory Society/Japanese Society of Respiratory Care Medicine on ARDS Clinical
Received in revised form Practice Guideline has created and released the ARDS Clinical Practice Guideline 2021.
7 May 2022 Methods: The 2016 edition of the Clinical Practice Guideline covered clinical questions (CQs)
Accepted 13 May 2022 that targeted only adults, but the present guideline includes 15 CQs for children in addition
Available online 24 June 2022 to 46 CQs for adults. As with the previous edition, we used a systematic review method
with the Grading of Recommendations Assessment Development and Evaluation (GRADE)
Keywords: system as well as a degree of recommendation determination method. We also conducted
ARDS systematic reviews that used meta-analyses of diagnostic accuracy and network meta-
Acute lung injury analyses as a new method.
Systematic review Results: Recommendations for adult patients with ARDS are described: we suggest against
Clinical practice guideline using serum C-reactive protein and procalcitonin levels to identify bacterial pneumonia as

1
The members of ARDS Clinical Practice Guideline Committee
are listed at Appendix A section.
448 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

the underlying disease (GRADE 2D); we recommend limiting tidal volume to 4e8 mL/kg for
mechanical ventilation (GRADE 1D); we recommend against managements targeting an
excessively low SpO2 (PaO2) (GRADE 2D); we suggest against using transpulmonary pres-
sure as a routine basis in positive end-expiratory pressure settings (GRADE 2B); we suggest
implementing extracorporeal membrane oxygenation for those with severe ARDS (GRADE
2B); we suggest against using high-dose steroids (GRADE 2C); and we recommend using
low-dose steroids (GRADE 1B). The recommendations for pediatric patients with ARDS are
as follows: we suggest against using non-invasive respiratory support (non-invasive posi-
tive pressure ventilation/high-flow nasal cannula oxygen therapy) (GRADE 2D); we suggest
placing pediatric patients with moderate ARDS in the prone position (GRADE 2D); we
suggest against routinely implementing NO inhalation therapy (GRADE 2C); and we suggest
against implementing daily sedation interruption for pediatric patients with respiratory
failure (GRADE 2D).
Conclusions: This article is a translated summary of the full version of the ARDS Clinical
Practice Guideline 2021 published in Japanese (URL: https://www.jrs.or.jp/publication/jrs_
guidelines/). The original text, which was written for Japanese healthcare professionals,
may include different perspectives from healthcare professionals of other countries.
© 2022 The Authors. Published by Elsevier B.V. on behalf of The Japanese Respiratory So-
ciety. This is an open access article under the CC BY license (http://creativecommons.org/
licenses/by/4.0/).

1. Background the present guideline, a joint committee was formed from


the preparatory stage to unify the intentions of the guideline
In Japan, the Japanese Society of Respiratory Care Medicine creation policy and methods.
published the first and second editions of the Clinical Practice Treatment outcomes have been improving mainly due to
Guideline for acute respiratory distress syndrome (ARDS) in advances in respiratory management, but the mortality rate of
1999 [1] and 2004 [2], respectively, after which the Japanese ARDS remains high at approximately 25e40% [6]. The primary
Respiratory Society published the Guideline for ARDS Practice objective of this guideline was to provide support so that
in 2005 [3] and 2010 [4]. These could be referred to as guide- healthcare professionals from multiple disciplines, including
books, so to speak, in the form of narrative reviews. Funda- non-specialist physicians, could make appropriate decisions
mentally, clinical guidelines need to go through a process of to improve the outcomes of patients with ARDS. Therefore, a
objectively determining recommendations for clinical ques- diverse range of areas need to be covered, from diagnosis to
tions (CQ) based on systematic reviews (SR) for evaluating the respiratory management, drug therapy, and physical therapy.
quality of evidence for a CQ. As such, both the content and scale far exceed the previous
The Japanese Society of Respiratory Care Medicine and edition. We hope that this guideline will serve as the basis for
Japanese Society of Intensive Care Medicine jointly estab- a platform that disseminates evidence relating to ARDS across
lished the ARDS Clinical Guideline creation committee in July clinical department disciplines.
2014 with the aim of providing SRs and recommendations
using the Grading of Recommendations Assessment, Devel-
opment and Evaluation (GRADE) system. At the same time, 2. Methods
the Japanese Respiratory Society had established a guideline
creation committee; therefore, the three societies and two 2.1. Composition of ARDS clinical practice guideline
committees decided to create a domestically integrated creation committee
clinical guideline. Specifically, the Japanese Respiratory So-
ciety was responsible for Part 1, which described the narra- The ARDS clinical practice guideline creation committee is
tive review section in the form of a so-called scope. The composed of the following.
Japanese Society of Respiratory Care Medicine and Japanese
Society of Intensive Care Medicine were primarily respon-  Governing committee: mainly responsible for managing
sible for Part 2, which focused on the SRs and recommen- and operating the overall clinical practice guideline, from
dations for the 13 CQs; however, the SR reviewers were CQ proposals mainly for adults to the creation of the final
recruited from all three societies. The completed ARDS draft of the clinical practice guideline.
Clinical Guideline 2016 [5] was highly regarded as an inter-  Pediatric steering committee: responsible for managing
national standard by the GRADE system even in the and operating the overall clinical practice guideline, from
Appraisal of Guidelines for Research and Evaluation II CQ proposals in the pediatric area to the creation of the
(AGREEII) by the Japan Council for Quality Health Care. final draft of the clinical practice guideline.
Meanwhile, some issues, such as inconsistent descriptions  SR steering and governing committee (area-governing team,
between Parts 1 and 2, remained. Therefore, when creating education team, methodology team): responsible for leading
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 449

the SR team that implements SRs in accordance with PICO KQ5: Is there a link between intermediate and final
and the CQs determined by the governing committee and outcomes?
panel meeting and also responsible for determining the SR
methodology and its education.  Key clinical issues related to intervention
 Support team: plays various auxiliary roles across the
overall clinical practice guideline creation process (e.g., KQ1 (comprehensive question): Can intervention improve
panel meeting operation, creation of various publications, final prognosis?
management of various created files). KQ2: Who should be subject to interventions?
 SR team: responsible for implementing SRs in accordance KQ3: What is the effect of interventions on outcomes?
with PICO and the CQs determined by the governing KQ4: What harms are there in interventions?
committee and panel meeting, as well as creating evidence KQ5: Is there a link between intermediate and final
profiles to be explained later. outcomes?
 Clinical practice guideline consultant: plays a supervisory
2.4. Creation of PICO sheet
and consultative role as a clinical practice guideline expert
across the entire process.
The patients, their diagnosis/prediction, interventions, out-
comes, etc., in the CQs, which were organized and formulated
above, were organized in a sheet called a PICO sheet. This PICO
2.2. Setting of important CQs
sheet was proposed mainly by the general committee members
and the responsible committee members of each SR area, and it
The governing committee selected 61 (adult: 46; children: 15)
was decided with the approval of the panel meeting. As the
CQs with reference to the previous guideline and public
targets of this guideline are patients with ARDS, so the patients
comments (JulyeSeptember 2019). The CQs were divided into
were set as patients with ARDS as a general rule. Some of the
the following five areas, with an area manager assigned for
clinical questions targeted patients with acute respiratory
each.
failure suspected with ARDS according to the analytic frame-
work. Furthermore, in this guideline, there are CQs relating to
 How should the diagnosis and prognosis prediction of
clinical practice areas where there were few reports of clinical
ARDS be conducted? (area A)
trials targeting only patients with ARDS, but where the results
 Should non-invasive respiratory support be used for pa-
of clinical trials targeting intensive care patients or mechanical
tients with ARDS? (area B)
ventilation patients are applicable to patients with ARDS; for
 How should invasive respiratory support be conducted for
such CQs, the target patients were set as patients with similar
patients with ARDS? (area C)
pathological conditions such as those receiving intensive care
 What and how should treatment adjacent to ventilator use
or mechanical ventilation.
be conducted for patients with ARDS? (area D)
 What and how should drug and non-drug therapy be con- 2.5. Relative importance of outcomes
ducted for patients with ARDS? (area E)
Outcomes involved listing beneficial (benefits) and harmful
(harms) aspects (no more than seven aspects), and a score of
2.3. Analytic framework 1e9 (1: least important; 9: most important) was relatively
assigned according to the GRADE system. The scoring was
To extract and organize the components of the CQs that
proposed mainly by the general committee members and the
should undergo SR in the abovementioned key clinical issues,
responsible committee members of each SR area, and it was
we created an analytic framework according to the clinical decided with the approval of the panel meeting.
flow and set key questions (KQ) 1e5 based on the following *In terms of scoring, if only a given outcome can occur,
definitions. This analytic framework was proposed mainly by then scores were given for cases where each outcome is crit-
the general committee members and responsible committee ical (7e9 points), important (4e6 points), or not important (1e3
members for each SR area, and it was decided with the points) for decision making. As a general rule for recommen-
approval of the panel meeting. The CQs that underwent SR for dation, decisions were made using outcomes that were eval-
each KQ in these key clinical issues were formulated using the uated as “critical.”
PICO format (alternatively, PECO, PICOT, or other formats).
Please see the attachment for the list of CQs. 2.6. SR policy

 Key clinical issues related to diagnosis/prediction For each CQ, each SR team conducted a preliminary search
and selected one of the following four policies.
KQ1 (comprehensive question): Does diagnosis/prediction
improve final prognosis?  Evaluating existing SRs and adapt their results (see below).
KQ2: Who should be subject to diagnosis/prediction?  Conducting a new SR.
KQ3: What is the diagnostic/predictive performance of  No SR conduction due to very scarce relevant literature
diagnosis/prediction outcomes? available. .
KQ4: What harms are there in diagnosis/prediction?  No SR conduction due to low clinical importance of the CQ.
450 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

2.7. GRADE-ADOLOPMENT (evaluation and adaptation Step 2: Primary screening


of existing SRs) All titles and abstracts specified in Step 1 were down-
loaded. The automatic duplicate deletion function of the
When evaluating existing SRs and adapting their results, such literature management software EndNote (Clarivate Analytics,
results are adapted when the following criteria are met. USA) or Mendeley (Mendeley Ltd., UK) were used to remove
duplicates, with duplicate articles further deleted manually.
 The clinical question matches. The article screening work was conducted online using
 A comprehensive search of research is conducted. Rayyan (https://rayyan.qcri.org/welcome). Two independent
 The eligibility criteria for the selection and exclusion of reviewers reviewed the titles and abstracts of the literature,
research are clearly defined. excluding those which clearly were different on the following
 The risk of bias of research is properly and critically points: intervention studies were selected on the basis of
examined research design, patients, intervention method, and language;
(e.g., Cochrane risk of bias tool in interventional studies, and diagnostic/predictive studies were selected on the basis of
ROBINS, in observational studies). research design, patients, index test, reference standard,
 A quantitative integration in which estimates effects availability of 2x2 table, and language. If there were conflicting
(meta-analysis) is conducted (when appropriate). opinions between the reviewers, a third reviewer conducted
 There is no problem with the quality of the article after an evaluation, with a decision made after discussion.
evaluating with AMSTAR2 or ROBINS.
Step 3: Secondary screening (full-text review).
If the publication of the existing SR is more than 2 years
The full text was ordered based on the screening results of
before the search formula creation start date of April 2020,
Step 2, and the target literature was selected using the entire
then an additional search is conducted with the same
main text. Intervention studies were selected on the basis of
search formula.
research design, patients, and intervention method. Diag-
 For an intervention-based SR, if there is a newer randomized
nostic/predictive studies were selected on the basis of
controlled trial (RCT) published than the existing SR, then a
research design, patients, index test, reference standard, and
new SR is conducted as a general rule.
availability of a 2x2 table. If there were conflicting opinions
 The existing SR articles and evaluated results (PICO,
between the reviewers, a third reviewer conducted an evalu-
AMSTAR2) are presented to the methodology team.
ation, with a decision made after discussion. A flow diagram
 The content is reviewed by two members of the method-
was created for the literature search and extraction process
ology team, who will confirm whether those articles can
through the primary and secondary screening.
truly be used. Those results will be presented to the gov-
erning committee, and an approval (or rejection) will be Step 4: Data extraction
presented as the final decision. Each team created a sheet in advance for extracting in-
 The use of the existing SR will be determined according to formation relating to the studies; information was then
the final decision of the governing committee. Even if the extracted based on this sheet. The data were listed separately
existing SR cannot be used, the use of the search formula or for each outcome determined in each CQ. When there was
risk of bias will be left to the discretion of each team. insufficient data, the authors were contacted as necessary.
 When using the existing articles for the SR of RCTs, whether
Step 5: Qualitative evaluation, quantitative integration
observational studies will be included will be determined on
The quality of evidence was evaluated according to the
the basis of the outcome. If the RCTs include many large-
method advocated by the GRADE working group. For primary
scale studies where a sufficient sample size/event occur-
studies to be incorporated in the meta-analysis for each CQ,
rence size is ensured for the harms, then there is no need to
the standard bias evaluation tools such as the Cochrane risk of
evaluate observational studies. If there is no evaluation for
bias tool (Ver. 1) and QUADAS-2 tool were used to grade the
the outcomes of harms, then observational studies will also
evidence across the four stages of high, moderate, low, and
be examined.
very low. For the meta-analysis of each outcome data, the
Cochrane Review Manager (RevMan5) software ver. 5.4 (http://
tech.cochrane.org/revman) or general statistical software
2.8. SR and meta-analysis
such as STATA, R, and SAS were used to conduct an appro-
priate quantitative integration according to the type of esti-
Step 1: Literature search
mated value of the effect.
A literature search is conducted based on previous studies,
After making a final decision of the quality of evidence and
previous ARDS guidelines, etc., from which key articles
conducting a meta-analysis, a summary of finding table (SoF
thought to be applicable to the research question are extrac-
table) and GRADE evidence profile table were created. GRADEpro
ted, and a literature search formula is created so that such key
GDT (http://gdt.guidelinedevelopment.org/) was used for the
articles are always included. When creating the literature
creation of both tables. After the SoF table and evidence profile
search formula, a final decision was made under the super-
were created by each SR team, mutual peer review was then
vision of the Kyoto Prefectural University of Medicine Library.
conducted by the guideline supervisor, education team, and
As a general rule, the literature search was conducted using
support team, centered on the methodology team. For area B,
MEDLINE (PubMed), CENTRAL, and Ichushi-Web as the data-
“Should non-invasive respiratory support be used for patients
bases. As a general rule, the search period was not limited.
with ARDS?” a network meta-analysis was conducted, and the
The languages used were either English or Japanese.
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 451

quality of evidence evaluation was conducted based on the In our practice statement
GRADE working group method. When there was insufficient evidence for a recommenda-
tion and a GPS was not applicable, but guidance based on
Step 6: Creation of body of evidence
current clinical practice patterns was considered appropriate,
Recommendations for each CQ were determined by
we provided descriptions in the current standard clinical
examining the quality of evidence and the balance between
practice as an “In our practice statement” [8,9]. An “In our
benefits and harms, as well as patient values, costs, and re-
practice statement” was intended to describe current care and
sources. Specifically, a table called the evidence-to-decision
was not intended to be interpreted as a recommendation [8].
(EtoD) framework was created based on the evidence profile,
and this was submitted to the panel meeting for drafting the Recommendation decision in panel meeting
recommendations. The modified Delphi method was used for consensus
building at the panel meeting.
2.9. Creation of recommendations
Step 1: Advance voting
Each committee member independently gave a score of
When an SR was conducted, or when an existing SR was
1e9 (1: disagree, 9: agree) for the created recommendation
evaluated and its results were adapted, the committee
draft. Committee members who gave a score of <7 also pro-
members (guideline supervisor, governing committee SR area
vided the reasons for the decision. Voting was conducted
supervisor, responsible committee members of each area,
online and anonymously. An independent member who was
methodology team, education team, support team) collabo-
not involved in voting (support team or governing committee
rated in advance of the decision of recommendation and
secretariat) aggregated the results and calculated the score
created drafts of the evidence to decision table (EtoD table)
median, inter-percentile range, inter-percentile range
and recommendation text. The EtoD table and recommenda-
adjusted for symmetry, and disagreement index (DI).
tion text drafts underwent mutual peer review with other
supervisors and the support team, centered on the method- Step 2: Panel meeting
ology team, and their contents were refined. The Minds Panel meetings were conducted based on the aggregated
guideline creation manual 2017 [7] was referenced in the results as shown below to reach consensus.
creation of the recommendation text draft. Afterwards, the
four factors of certainty of evidence, balance of effects, values (1) When there was no agreement (Median <6.5 or DI  1)
and acceptability, and feasibility were examined based on the Discussions were held within the committee, after which
draft, and the recommendations were formulated by the amendments were made to the EtoD and recommended
panel meeting using the following procedure. text, and a second vote was held.
Voting was conducted up to three times.
When there is no reliable evidence or when the SR was not
(2) When there was agreement (Median 6.5 and DI < 1)
conducted
When SR was conducted but there was no reliable evidence
When a serious opinion was present during voting for a
(e.g., lack of applicable literature), or when an SR was not
comment or recommendation presented by committee
conducted, this was not subject to GRADE evaluation (un-
member, discussions were held within the committee, and a
graded). In these cases, the responsible committee members
consensus was reached. CQs for which a consensus was not
of the area as well as members of the SR governing committee
reached within the committee resulted in amendments to the
(SR governing committee of each area, methodology team,
EtoD and recommended text, after which a second vote was
education team) collaborated to create a recommendation
held.
text draft with reference to the following criteria, and a policy
of (1) or (2) was decided by the panel meeting. Strength of recommendation
The strength of recommendation shown by the GRADE
(1) Recommendation as good practice statement (GPS) system was classified into four categories: 1, recommended/
(2) Description of current state of standard clinical practice recommended against, and 2, suggested/suggested against
without recommendation (in our practice statement). (Table 1).
Additionally, the certainty of evidence was classified as
Good practice statement shown in Table 2.
When confident that the certainty of the net benefit of a
Creation of recommendation text and public comments
medical practice shown by indirect evidence is at the same
After the recommendation text and strength of recom-
level as this example even without a formal literature search,
mendation were determined, the governing committee wrote
or when it is thought that the task of collecting linked indirect
the commentary text. Afterwards, public comments were
evidence supporting the applicable recommendation would
solicited from June 11th to June 24th, 2021, on the websites of
be difficult and unproductive, then we considered presenting
each academic society. There were 10 comments during the
this as a GPS [8,9]. In these cases, the recommendation was
designated period, and descriptions were added in the text.
“recommended,” and the quality of evidence was “ungraded”
(ungraded recommendation) [8,9]. The recommendation text Recommendations on COVID-19
clarified “Good practice statement.” The panel meeting Recommendations for COVID-19 were not included in this
decided whether to support the GPS by a “yes/no” vote, with guideline because there is insufficient evidence for their
use of the GPS decided at 90% agreement. inclusion.
452 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

Table 1 e The categories of the strength of recommendation shown by the GRADE system.
Recommendation Example Notation
Recommended “We recommend conducting xx (medical practice).” 1
Suggested “We suggest conducting xx (medical practice).” 2
Recommended against “We recommend against conducting xx (medical practice).” 1
Suggested against “We suggest against conducting xx (medical practice).” 2

Table 2 e The definitions of the certainly of evidence.


Certainty Definition Notation
High Highly confident that the effect estimates supports the recommendation A
Moderate Moderate confidence that the effect estimates supports the B
recommendation
Low Limited confidence that the effect estimates supports the C
recommendation
Very low Almost no confidence in the effect estimates to support the D
recommendation

furthermore, there is a possibility that the prognosis improves


3. Results as a result of conducting evidence-based recommendations
presented in this guideline. Therefore, there are large benefits
Below, the CQs and their recommendations are described for in ARDS diagnosis in patients with acute respiratory failure.
the five adult areas and the pediatric area. Please refer to ARDS diagnosis is based on the Berlin Definition proposed in
additional files 1e6 for the search strategies, flow diagram, 2012. The minimum requirements for the diagnostic items in
risk of bias summaries, forest plot, EP and EtD of each CQ. addition to medical history and physical findings are chest
imaging tests (simple chest X-rays, chest CT) and arterial
A. Area A: Diagnosis/severity of illness evaluation/type blood gas analysis; the harm to patients and additional costs
evaluation to medical institutions as a result of these actions are thought
to be trivial.
CQ1: Should ARDS diagnoses be conducted for patients
with acute respiratory failure? CQ2: Should blood brain natriuretic peptide (BNP) and NT-
proBNP levels be used for identifying cardiogenic pulmonary
Background edema as the causative disease of acute respiratory failure?
ARDS is a serious and urgent pathological condition that
causes acute respiratory failure. Its diagnosis can affect the Background
understanding of these pathological conditions, severity of Identifying cardiogenic pulmonary edema is important in
illness evaluation, and treatment policy, and whether to ARDS clinical practice. Blood BNP and NT-proBNP levels are
confirm the diagnosis of ARDS is an important clinical widely used as a supplementary diagnosis for heart failure,
question. and whether to use these for identifying the causative disease
of acute respiratory failure is an important clinical question.
Recommendation
We recommend that ARDS should be suspected for pa- Recommendation
tients with acute respiratory failure (GPS). We suggest using blood BNP or NT-proBNP levels for
identifying cardiogenic pulmonary edema in patients with
Supplementary item acute respiratory failure (weak recommendation/very low
Not only is the diagnosis of ARDS important for improving certainty of evidence: GRADE 2D).
prognosis but also the diagnosis and treatment of diseases
that causes ARDS. Supplementary conditions
Changes in clinical conditions (e.g., patient characteristics,
Rationale testing characteristics and timing, pre-test probability, pa-
Summary of evidence: tient/medical staff values) may change the balance of effects
There are no clinical studies that compare whether con- and result in a different option being recommended.
firming or not confirming ARDS in a patient with acute res-
piratory failure directly improves patient outcomes. When Rationale
ARDS is diagnosed, there is a possibility that the prognosis Summary of evidence:
improves as a result of conducting the evidence-based rec- We found 6 studies on BNP and NT-proBNP levels in the
ommendations presented in this guideline. blood. Blood BNP (cutoff value: 400e500 pg/mL) (3 studies
[10e12]: N ¼ 252) had an integrated sensitivity of 0.77 (95%
Balance of effects, acceptability, and feasibility determination: confidence interval [CI]: 0.65e0.85) and integrated specificity
The diagnosis of ARDS enables important clinical decision- of 0.62 (95% CI: 0.53e0.70). Blood BNP (cutoff value: 1,000 pg/
making, such as understanding the pathological condition, mL) (2 studies [11,12]: N ¼ 128) had an integrated sensitivity of
evaluating illness severity, and deciding on a treatment policy;
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 453

0.50 (95% CI: 0.36e0.64) and integrated specificity of 0.82 (95% CQ4: Should pneumococcal urinary antigen tests and
CI: 0.72e0.89). Additionally, blood NT-proBNP (cutoff value: sputum Gram staining be used for identifying pneumococcal
4,000 pg/mL) (1 study [13]: N ¼ 121) had an integrated sensi- pneumonia as the causative disease of ARDS?
tivity of 0.71 (95% CI: 0.52e0.85) and integrated specificity of
Background
0.89 (95% CI: 0.80e0.94). The implementation of testing is
Pneumococcal pneumonia is a cause of acute respiratory
generally supported when taking into account the balance
failure and ARDS, and early medical intervention is important.
between desirable and undesirable effects due to the imple-
Whether to use urinary pneumococcal capsular antigen tests
mentation of testing.
and sputum Gram staining for conducting the early diagnosis
Certainty of evidence: of pneumococcal pneumonia is an important clinical question.
The quality of evidence for the diagnostic performance of
Recommendation
testing was judged as “very low” for almost all cases.
We suggest the use of pneumococcal urinary antigen
Balance of effects, acceptability, and feasibility determination: testing and sputum Gram staining for identifying pneumo-
This is already used in routine clinical practice, and there coccal pneumonia as the causative disease of ARDS (weak
are no problems with acceptability and feasibility. recommendation/very low certainty of evidence: GRADE 2D.

CQ3: Should serum C-reactive protein (CRP) and procalci- Supplementary conditions
tonin (PCT) levels be used for identifying bacterial pneumonia Changes in clinical conditions (e.g., patient characteristics,
as the underlying disease of ARDS? testing characteristics and timing, pre-test probability, pa-
tient/medical staff values) may change the balance of effects
Background
and result in a different option being recommended.
Pneumonia is a common causative disease of ARDS, most
of which is bacterial pneumonia. Whether to use CRP and PCT Rationale
for early recognition of this bacterial pneumonia is an Summary of evidence:
important clinical question. Pneumococcal urinary antigen testing (23 studies [36e58]:
N ¼ 10,900) had an integrated sensitivity of 0.65 (95% CI:
Recommendation
0.61e0.68) and integrated specificity of 0.91 (95% CI: 0.85e0.95).
We suggest against identifying bacterial pneumonia as the
Sputum Gram staining (11 studies [55,59e68]: N ¼ 1,794) had
underlying disease of ARDS only with serum CRP and PCT
an integrated sensitivity of 0.69 (95% CI: 0.56e0.80) and inte-
results (weak recommendation/very low certainty of evi-
grated specificity of 0.91 (95% CI: 0.83e0.96).
dence: GRADE 2D).
Certainty of evidence:
Supplementary conditions
The certainty of evidence relating to diagnostic perfor-
Changes in clinical conditions (e.g., patient characteristics,
mance for each test was judged as “very low.”
testing characteristics and timing, pre-test probability, pa-
tient/medical staff values) may change the balance of effects Balance of effects, acceptability, and feasibility determination:
and result in a different option being recommended. When comparing the effects and harms of testing with the
case where the treatment policy was decided without testing,
Rationale
it is thought that the desirable effects of testing are large. This
Summary of evidence:
is a generally accepted medical practice with no problems
We found 14 studies relating to serum CRP and 21 studies
regarding feasibility.
relating to serum PCT. Serum CRP (14 studies [14e27]:
N ¼ 3,093) had a sensitivity of 0.76 (95% CI: 0.63e0.89) and CQ5: Should Legionella urinary antigen testing be used for
specificity of 0.78 (fixed). Serum PCT (21 studies [14e26,28e35]: identifying Legionella pneumonia as the causative disease of
N ¼ 4,721) had a sensitivity of 0.64 (95% CI: 0.56e0.73) and ARDS?
specificity of 0.83 (fixed) (estimates using HSROC model, fixed
Background
at median value of specificity of primary studies).
Legionella pneumonia presents with rapid respiratory fail-
Certainty of evidence: ure and is important as a causative disease of ARDS. Mean-
In consideration of the risk of bias, indirectness, inconsis- while, there is a possibility that an improved prognosis can be
tency, and inaccuracy, and certainty of evidence relating to obtained with Legionella pneumonia with an appropriate
the effects of testing was judged as “very low.” administration of antibiotics, so whether to use Legionella
urinary antigen testing is an important clinical question.
Balance of effects, acceptability, and feasibility determination:
When comparing patients who are suspected of having Recommendation
bacterial pneumonia based on the results of serum CRP or We suggest using Legionella urinary antigen testing for
serum PCT tests and those who are treated as if they have identifying Legionella pneumonia as the causative disease of
bacterial pneumonia regardless of the results of these tests, ARDS (weak recommendation/very low certainty of evidence
the latter may be more beneficial in situations where the pre- GRADE 2D).
test probability is estimated to be above a certain level.
Supplementary conditions
Complications from testing are thought to be negligible in
Changes in clinical conditions (e.g., patient characteristics,
terms of clinical decision-making. This is a widely used test
testing characteristics and timing, pre-test probability,
with probably no problem regarding feasibility.
454 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

patient/medical staff values) may change the balance of ef- acceptability and feasibility since these tests are conducted in
fects and result in a different option being recommended. daily clinical practice.

Rationale CQ7: Should antigen tests of the pharyngeal/nasopharyn-


Summary of evidence: geal swabs and PCR tests of the bronchoalveolar lavage fluid
We found 21 studies [41,69e88] relating to Legionella urinary be used for identifying influenza pneumonia as the causative
antigen testing (N ¼ 11,724); the integrated sensitivity was 0.79 disease of ARDS?
(95% CI: 0.71e0.85) and the integrated specificity was 1.00 (95%
Background
CI: 0.99e1.00).
It has been reported that the administration of neur-
Certainty of evidence: aminidase inhibitors reduced hospitalization, pneumonia
Considering the risk of bias, indirectness, and inconsis- onset, and mortality rate in influenza virus infections. Antigen
tency for both sensitivity and specificity, the certainty of evi- tests based on rapid diagnostic kits using pharyngeal swabs or
dence was judged to be “very low.” nasopharyngeal swabs are commonly used in the diagnosis of
influenza virus infections. Meanwhile, there are some reports
Balance of effects, acceptability, and feasibility determination:
that indicated that antigen tests using upper respiratory tract
When comparing the effects and harms of Legionella uri-
specimens could not diagnose novel Influenza A (H1N1)
nary antigen testing with the case where the treatment policy
pneumonia, with diagnoses made using PCR tests of bron-
was decided without testing, the desirable effects of testing
choalveolar lavage (BAL) fluid. Diagnosing influenza pneu-
are thought to be large. This is a generally accepted medical
monia with these tests and conducting therapeutic
practice with no problems regarding feasibility.
intervention based on the administration of the appropriate
CQ6: Should antigen and PCR tests of the pharyngeal antiviral medication is important.
swabs and serum antibody tests be used to identify Myco-
Recommendation
plasma pneumonia as the causative disease of ARDS?
We cannot provide a specific recommendation regarding
Background whether to use antigen tests of the pharyngeal swabs and/or
Mycoplasma pneumonia is relatively common as a form of PCR tests of the BAL fluid for identifying influenza pneumonia
community-acquired pneumonia and can rarely progress to as the causative disease of ARDS. The current status of these
ARDS and present with serious respiratory failure. Antigen tests is that they are used based on the experience of clini-
and PCR tests of the pharyngeal swabs and serum antibody cians (in our practice statement).
tests are generally used for the diagnosis of Mycoplasma
Supplementary conditions
pneumonia. Verifying the benefits and harms due to the
None
implementation of these tests is an important issue in ARDS
clinical practice. Rationale
Summary of evidence:
Recommendation
No applicable studies
We cannot provide a specific recommendation regarding
whether to use antigen and PCR tests of the pharyngeal swabs Certainty of evidence:
or serum antibody tests for identifying Mycoplasma pneu- Since there are no applicable studies, the quality of evi-
monia as a causative disease of ARDS. The current status of dence cannot be evaluated.
these tests is that they are used based on the experience of
Balance of effects, acceptability, and feasibility determination:
clinicians (in our practice statement).
The balance between desirable effects and undesirable
Supplementary conditions effects is unclear. It is thought that there are no problems
None regarding acceptability and feasibility.

Rationale CQ8: Should PCR tests of the bronchoalveolar lavage fluid


Summary of evidence: and blood antigenemia methods be used for identifying
No applicable studies cytomegalovirus pneumonia as the causative disease of
ARDS?
Since there is no high-quality evidence that investigated
the diagnostic accuracy of tests, no clear recommendations Background
can be made for this CQ. Therefore, we provided a description The cytomegalovirus (CMV) pneumonia blood antigenemia
of the current clinical practice rather than an evidence-based method and CMV-PCR test of the BAL fluid are used for the
recommendation for this CQ. early diagnosis of CMV infection and determination of the
effect of treatment, and they are widely used as an index for
Certainty of evidence:
monitoring, determining the effect of antiviral agents, and the
Since there are no applicable studies, the quality of evi-
discontinuation period.
dence cannot be evaluated.
Recommendation
Balance of effects, acceptability, and feasibility determination:
We suggest using PCR tests of BAL fluid and blood anti-
The balance between benefits and harms is unclear. It is
genemia methods for identifying CMV pneumonia as the
thought that there are no problems with regard to
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 455

causative disease of ARDS (weak recommendation/very low and integrated specificity of PCP diagnosis were 0.84 (95% CI:
certainty of evidence: GRADE 2D). 0.66e0.93) and 0.79 (95% CI: 0.69e0.87), respectively.

Supplementary conditions Certainty of evidence:


Changes in clinical conditions may change the balance of The certainty of evidence relating to the diagnostic per-
effects and result in a different option being recommended. formance of PCP tests using serum b-D-glucan was judged as
“very low.”
Rationale
Summary of evidence: Balance of effects, acceptability, and feasibility determination:
PCR tests using BAL fluid (5 studies [89e93]: N ¼ 353) had an The implementation of testing is generally supported when
integrated sensitivity of 0.94 (95% CI: 0.86e0.97) and integrated taking into account the balance between benefits and harms
specificity of 0.84 (95% CI: 0.52e0.96). due to the implementation of testing. Meanwhile, the certainty
Blood antigenemia methods (3 studies [94e96]: N ¼ 91) of evidence relating to testing in general is very low. There is no
had an integrated sensitivity of 0.67 (95% CI: 0.54e0.79) and problem with feasibility, and it is a generally accepted method.
integrated specificity of 0.87 (95% CI: 0.73e0.95) when the
CQ10: Should serum b-D-glucan and galactomannan an-
definition of “positive” was at least one positive cell per
tigens of the blood or BAL fluid be used for identifying inva-
200,000. Furthermore, the frequency of adverse events with
sive pulmonary aspergillosis as the causative disease of
BAL was as follows: mortality, 0.000% (95% CI: 0.000e0.035).
ARDS?
Certainty of evidence:
Background
For both PCR tests of the BAL fluid and blood antigenemia
Invasive pulmonary aspergillosis (IPA) rapidly progresses
methods, the certainty of evidence was judged as “very low”
and presents with respiratory failure and can also result in
after considering the risk of bias, etc. The certainty of evidence
fatal outcomes. Therefore, whether to use biomarkers for
regarding the harm of BAL was judged as “moderate” or “low.”
identifying the causative disease of ARDS is an important
Balance of effects, acceptability, and feasibility determination: clinical question.
Benefits included appropriate treatment based on diag-
Recommendation
nosis, and harms included unnecessary treatment due to
We suggest against identifying IPA as the causative disease
misdiagnosis (false positive) and adverse events. When
of ARDS using only serum b-D-glucan test results (weak
compared with cases where the treatment policy was decided
recommendation/very low certainty of evidence: GRADE 2D).
without testing, the benefits were thought to be large. This is a
We suggest identifying IPA as the causative disease of
feasible medical practice.
ARDS using galactomannan antigen tests in blood and BAL
CQ9: Should serum b-D-glucan be used for identifying fluid (weak recommendation/very low certainty of evidence:
Pneumocystis pneumonia as the causative disease of ARDS? GRADE 2D).

Background Supplementary conditions


Pneumocystis pneumonia (PCP) causes acute respiratory Changes in clinical conditions (e.g., patient characteristics,
failure, and it is anticipated that early diagnosis with serum b- testing characteristics and timing, pre-test probability, pa-
D-glucan testing would contribute to the improved prognosis tient/medical staff values) may change the balance of effects
of patients. Meanwhile, misdiagnosis with PCP is thought to and result in a different option being recommended.
worsen patient prognosis through the administration of un-
Rationale
necessary drugs and its side effects, as well as the lack of
Summary of evidence:
implementation of treatments for other causative diseases.
When setting a cutoff value of 80 pg/mL for serum b-D-
Verifying the benefits and harms of test implementation is an
glucan (9 studies [100e108]: N ¼ 757), the integrated sensitivity
important issue in ARDS clinical practice.
was 0.70 (95% CI: 0.49e0.85) and the integrated specificity was
Recommendation 0.73 (95% CI: 0.58e0.84). Additionally, when setting a cutoff
We suggest using serum b-D-glucan tests for identifying value of 1.0 optical density index (ODI) for blood gal-
PCP as the causative disease of ARDS (weak recommendation/ actomannan antigen tests (8 studies [109e116]: N ¼ 145), the
very low certainty of evidence: GRADE 2D). integrated sensitivity was 0.76 (95% CI: 0.60e0.91) and the in-
tegrated specificity was 0.88 (95% CI: 0.79e0.94).
Supplementary conditions
Changes in clinical conditions (e.g., patient characteristics, Certainty of evidence:
testing characteristics and timing, pre-test probability, pa- Considering the risk of bias, indirectness, inconsistency,
tient/medical staff values) may change the balance of effects and inaccuracy for both sensitivity and specificity in either
and result in a different option being recommended. test, the certainty of evidence was judged to be “very low.”

Rationale Balance of effects, acceptability, and feasibility determination:


Summary of evidence: It was thought that there were limited clinical conditions
When setting a cutoff value (80 pg/mL) for serum b-D- that could benefit from serum b-D-glucan tests, whereas there
glucan (3 studies [97e99]: N ¼ 148), the integrated sensitivity were many clinical conditions for galactomannan antigen
456 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

tests. Both tests are generally accepted medical practices with integrated specificity of 0.69 (fixed). Chest CT (6
no problems with regard to feasibility. studies [128e133]: N ¼ 409) had an integrated sensitivity of
0.62 (95% CI: 0.32e0.91) and an integrated specificity of 0.76
CQ11: Should plain chest X-rays, chest high-resolution CT,
(fixed).
and interferon g release assays be used for identifying miliary
tuberculosis as the causative disease of ARDS? Certainty of evidence:
In consideration of the risk of bias, indirectness, inconsis-
Background
tency, and inaccuracy for both lung biopsies and chest CT, the
Not only can miliary tuberculosis be the cause of ARDS and
certainty of evidence was judged as “very low.”
be fatal, the disease also requires prevention measures for
airborne infection. Therefore, whether chest imaging tests or Balance of effects, acceptability, and feasibility determination:
interferon g release assays (IGRAs) should be conducted for When considering the net benefits after including adverse
identifying miliary tuberculosis as the causative disease of events associated with tests, it is thought that there was a very
ARDS is an important question. limited number of clinical conditions where determining the
treatment policy using lung biopsies or chest CT test results is
Recommendation
useful. It was judged that a general statement could not be
We cannot provide a specific recommendation regarding
made regarding the feasibility of lung biopsies and chest CT
whether to use plain chest X-rays, chest high-resolution CT
tests.
(HRCT), and IGRAs for identifying miliary tuberculosis as the
causative disease of ARDS. Plain chest X-ray is conducted for CQ13: Should PaO2/FIO2 (P/F) ratio be used for predicting
almost all patients with respiratory failure; HRCT and IGRA prognosis of patients with ARDS?
are currently conducted based on the experience of clinicians
Background
(in our practice statement).
The Berlin Definition stipulated the severity of ARDS ac-
Rationale cording to the P/F ratio, but its prognosis prediction perfor-
Summary of evidence: mance is unclear. Whether to use the P/F ratio for ARDS
No applicable studies patient prognosis prediction was thought to be clinically
important.
Certainty of evidence:
Since there are no applicable studies, the quality of evi- Recommendation
dence cannot be evaluated. We suggest against ARDS patient prognosis prediction
using only P/F ratio (cutoff: 100, 200) results (weak recom-
Balance of effects, acceptability, and feasibility determination:
mendation/very low certainty of evidence: GRADE 2D).
The balance between benefits and harms is unclear. It is
thought that there are no problems with regard to accept- Supplementary conditions
ability and feasibility. The P/F ratio is used to determine the severity of illness and
treatment policy in clinical practice, but this recommendation
CQ12: Should lung pathology or chest CT imaging be used
does not reject the use of the P/F ratio in such circumstances.
for predicting prognosis of ARDS patients?
Additionally, changes in clinical conditions (e.g., patient
Background characteristics, testing characteristics and timing, pre-test
ARDS typically presents with pathologically diffuse alve- probability, patient/medical staff values) may change the
olar damage, but the Berlin Definition does not necessarily balance of effects and result in a different option being
refer to pathological conditions. Whether to use lung pathol- recommended.
ogy and chest CT imaging for predicting prognosis of patients
Rationale
with ARDS is an important clinical question.
Summary of evidence:
Recommendation When the cutoff value is 100 (19 studies [129,134e151]:
We suggest against using only lung pathology or only chest N ¼ 15,040), the integrated sensitivity was 0.43 (95% CI:
CT imaging for predicting prognosis of patients with ARDS 0.37e0.50) and integrated specificity was 0.70 (95% CI:
(weak recommendation/very low certainty of evidence: 0.66e0.74). When the cutoff value was 200 (20 studies
GRADE 2D). [129,134,135,137e150,152e154]: N ¼ 15,489), the integrated
sensitivity was 0.85 (95% CI: 0.81e0.88) and integrated speci-
Supplementary conditions
ficity was 0.24 (95% CI: 0.21e0.29).
Changes in clinical conditions (e.g., patient characteristics,
testing characteristics and timing, pre-test probability, pa- Certainty of evidence:
tient/medical staff values, decision-making following test re- In consideration of the risk of bias, inconsistency, and in-
sults) may result in a different option being recommended. accuracy, the certainty of evidence was judged as “very low.”

Rationale Balance of effects, acceptability, and feasibility determination:


Summary of evidence: When considering the net benefits, there was a very
Lung biopsies (11 studies [117e127]: N ¼ 616) had an limited number of clinical conditions where determining the
integrated sensitivity of 0.42 (95% CI: 0.21e0.57) and treatment policy using P/F ratio test results was useful. The P/
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 457

F ratio is a commonly conducted medical practice and is Considering the ranking results, there may be little differ-
thought to be feasible. ence in the effects of reducing mortality between NPPV and
HFNC among the types of non-invasive respiratory support.
B. Area B: Non-invasive respiratory support
CQ 15: Should NPPV be used over oxygen therapy for pa-
tients with ARDS?
CQ 14: Should non-invasive respiratory support be used
for patients with ARDS? Background
NPPV is used for patients with acute respiratory failure to
Background
avoid complications due to tracheal intubation, but delays in
Non-invasive respiratory support such as non-invasive
tracheal intubation increase mortality risk. NPPV is effective
positive pressure ventilation (NPPV) and high-flow nasal
for cardiogenic pulmonary edema and acute exacerbations of
cannula oxygen therapy (HFNC) is expected to avoid compli-
chronic obstruction pulmonary disease (COPD), but NPPV is
cations due to tracheal intubation, but delays in tracheal
not an established treatment for patients with ARDS.
intubation have been reported to increase mortality rate.
However, NPPV and HFNC are not established treatments in Recommendation
the respiratory management of patients with ARDS. We suggest conducting NPPV over conventional oxygen
therapy as an initial respiratory management for adult pa-
Recommendation
tients with acute respiratory failure suspected of having ARDS
We suggest conducting non-invasive respiratory
if there are no contraindications for non-invasive respiratory
support (NPPV, HFNC) instead of conventional oxygen
support or if organ failure other than respiratory failure is
therapy as an initial respiratory management for adult pa-
absent (weak recommendation/moderate certainty of evi-
tients with acute respiratory failure suspected of having ARDS
dence: GRADE 2B).
if there are no contraindications for non-invasive respiratory
support or if organ failure other than respiratory failure is Supplementary item
absent. Because delays in necessary tracheal intubation could in-
NPPV (weak recommendation/moderate certainty of evi- crease mortality, patients should be carefully managed in an
dence: GRADE 2B). environment where tracheal intubation can be conducted
HFNC (weak recommendation/moderate certainty of evi- after the start of NPPV.
dence: GRADE 2B). Contraindications for non-invasive respiratory support
We suggest conducting non-invasive respiratory support include the inability to protect the airway, high risk of vom-
(NPPV, HFNC) over tracheal intubation. iting, impaired consciousness, uncooperative patients, and
NPPV (weak recommendation/moderate certainty of evi- unstable hemodynamics.
dence: GRADE 2B).
HFNC (weak recommendation/moderate certainty of evi- Rationale
dence: GRADE 2B). Summary of evidence:
Network meta-analysis was conducted using 19 RCTs
Supplementary item (N ¼ 2,777) [155e173]. Short-term mortality (direct compari-
Delays in necessary tracheal intubation could increase son 14 RCTs [155,158,160,162e171,173]: N ¼ 1,494) occurred in
mortality, so patients should be carefully managed in an 107 fewer/1,000 patients (166 fewere31 fewer patients);
environment where tracheal intubation could be conducted tracheal intubation (direct comparison 14 RCTs [155,158,160,
after the start of non-invasive respiratory support. 163e171,173]: N ¼ 1,495) was performed in 135 fewer/1,000
Contraindications for non-invasive respiratory support patients (188 fewere69 fewer patients); pneumonia (direct
include the inability to protect the airway, high risk of vom- comparison 8 RCTs: N ¼ 804) occurred in 50 fewer/1,000 pa-
iting, impaired consciousness, uncooperative patients, and tients (75 fewere11 fewer patients. From the above, the
unstable hemodynamics. desirable effect of NPPV was judged to be “moderate.”
Rationale There were 2 RCTs [167,170] that reported on skin disor-
Summary of evidence: ders, which was an important outcome relating to harm; this
This CQ included examinations using network meta- did not occur with conventional oxygen therapy, whereas
analyses (see CQ15e18 for details). The surface under the 3.2%e25.5% of patients undergoing NPPV experienced skin
cumulative ranking curve (SUCRA), which was quantified as disorders. From the above, the undesirable effect of NPPV was
the average rating, was used to plot the SUCRA values of each judged to be “trivial.”
device (from a minimum of 0 to maximum of 100), with short- Certainty of evidence:
term mortality on the horizontal axis and pneumonia on the The certainty of evidence and direction of all effects were
vertical axis. consistent, and the certainty of evidence for the overall
In order of descending favorability, the ranking of short- outcome was judged as “moderate” adopting the highest
term mortality and pneumonia was 1. HFNC, 2. NPPV, 3. me- certainty of evidence.
chanical ventilation (MV), and 4. Oxygen therapy. The SUCRA
values for short-term mortality were 72.0 for HFNC, 66.3 for Values, balance of effects, acceptability, and feasibility
NPPV, 49.9 for MV, and 11.7 for oxygen therapy; the SUCRA determination:
values for pneumonia were 92.3 for HFNC, 71.9 for NPPV, 33.1 A dedicated device is needed for NPPV, and this may in-
for oxygen therapy, and 2.7 for MV. crease the burden on nurses; however, this is already
458 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

implemented in the daily clinical practice of many acute care CQ 17: Should NPPV be used prior to conducting tracheal
hospitals. As such, even when considering cost, harmfulness, intubation in patients with ARDS?
etc., it was judged that it was “probably yes” acceptable and
Background
feasible.
NPPV is used for patients with acute respiratory failure to
CQ 16: Should HFNC be used over conventional oxygen avoid complications due to tracheal intubation, but delays in
therapy for patients with ARDS? tracheal intubation increase mortality risk. NPPV is effective
for cardiogenic pulmonary edema and acute exacerbations of
Background
COPD, but NPPV is not an established treatment for patients
HFNC is used for patients with acute respiratory failure to
with ARDS.
avoid complications due to tracheal intubation, but delays in
tracheal intubation increase mortality risk. However, HFNC is Recommendation
not an established treatment in the respiratory management We suggest conducting NPPV over tracheal intubation
of patients with ARDS. as an initial respiratory management for adult patients
with acute respiratory failure suspected of having ARDS if
Recommendation
there are no contraindications for non-invasive respiratory
We suggest conducting HFNC over oxygen therapy as
support or if organ failure other than respiratory failure is
an initial respiratory management for adult patients
absent (weak recommendation/moderate certainty of evi-
with acute respiratory failure suspected of having ARDS if
dence: GRADE 2B).
there are no contraindications for non-invasive respiratory
support or if organ failure other than respiratory failure is Supplementary item
absent (weak recommendation/moderate certainty of evi- Because delays in necessary tracheal intubation could in-
dence: GRADE 2B). crease mortality, patients should be carefully managed in an
environment where tracheal intubation can be conducted
Supplementary item
after the start of NPPV.
Because delays in necessary tracheal intubation could in-
Contraindications for non-invasive respiratory support
crease mortality, patients should be carefully managed in an
include the inability to protect the airway, high risk of vom-
environment where tracheal intubation can be conducted
iting, impaired consciousness, uncooperative patients, and
after the start of HFNC.
unstable hemodynamics.
Contraindications for non-invasive respiratory support
include the inability to protect the airway, high risk of vom- Rationale
iting, impaired consciousness, uncooperative patients, and Summary of evidence:
unstable hemodynamics. Network meta-analysis was conducted using 19
RCTs (N ¼ 2,777) [155e173]. Short-term mortality (direct
Rationale
comparison 2 RCTs [157,171]: N ¼ 129) occurred in 31 fewer/
Summary of evidence:
1,000 patients (203 fewere296 more patients); pneumonia
Network meta-analysis was conducted using 19 RCTs
(direct comparison 2 RCTs [157,171]: N ¼ 129) occurred in 401
(N ¼ 2,777) [155e173]. Short-term mortality direct compari-
fewer/1,000 patients (450 fewere295 fewer patients). From the
son 2 RCTs: N ¼ 976) occurred in 117 fewer/1,000 pa-
above, the desirable effect of NPPV was judged to be
tients (215 fewere44 more patients); intubation (direct
“moderate.”
comparison 4 RCTs [156,159e161]: N ¼ 1176) was performed
There was 1 RCT [157] that reported on skin disorders,
in 89 fewer/1,000 patients (187 fewere127 more); pneumonia
which was an important outcome relating to harm; there were
(direct comparison 1 RCT [160]: N ¼ 200) occurred in 82
more cases of skin disorders with NPPV (3/32 [9.4%] vs. 0/32
fewer/1,000 patients (110 fewere4 more patients). From the
[0%]). From the above, the undesirable effect of NPPV was
above, the desirable effect of HFNC was judged to be
judged to be “trivial.”
“moderate.”
Certainty of evidence:
Certainty of evidence:
The direction of all effects was consistent, and the cer-
The direction of all effects was consistent, and the cer-
tainty of evidence for the overall outcome was judged as
tainty of evidence for the overall outcome was judged as
“moderate” adopting the highest certainty of evidence.
“moderate” adopting the highest certainty of evidence.
Values, balance of effects, acceptability, and feasibility
Values, balance of effects, acceptability, and feasibility
determination:
determination:
The balance of effects of NPPV was judged as “probably
The balance of effects of HFNC was judged as
favors the intervention.” A dedicated device is needed for
“probably favors the intervention.” A dedicated device is needed
NPPV, but this is relatively cheap compared with a ventilator.
for HFNC, and a large amount of oxygen is needed compared
It is already implemented in the daily clinical practice of many
with oxygen therapy, but this is already implemented in the
acute care hospitals, so it was judged that it was “probably
daily clinical practice of many acute care hospitals, so it was
yes” acceptable and feasible.
judged that it was “probably yes” acceptable and feasible.
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 459

CQ 18: Should HFNC be used prior to conducting tracheal C. Area C: Invasive respiratory support
intubation in patients with ARDS?
CQ 19: Should low tidal volume be used in mechanically
Background
ventilated adult patients with ARDS?
HFNC is used for patients with acute respiratory failure to
avoid complications due to tracheal intubation, but delays in Background
tracheal intubation increase mortality risk. However, HFNC is Several studies have suggested that mechanical ventilation
not an established treatment in the respiratory management may cause lung damage and bleeding. Limiting tidal volume is
of patients with ARDS. one of the lung protection strategies. Its effects have been
verified in several large-scale RCTs, the results of which,
Recommendation
however, are inconsistent.
We suggest conducting HFNC over tracheal intubation as
an initial respiratory management for adult patients with Recommendation
acute respiratory failure suspected of having ARDS if there We recommend limiting tidal volume to 4e8 mL/kg for
are no contraindications for non-invasive respiratory sup- mechanically ventilated adult patients with ARDS (strong
port or if organ failure other than respiratory failure is absent recommendation/very low certainty of evidence: GRADE 1D).
(weak recommendation/moderate certainty of evidence:
Supplementary item
GRADE 2B).
The criteria for adoption of the studies used in the SR were
Supplementary item 4e8 mL/kg for the low tidal volume ventilation group and
Because delays in necessary tracheal intubation could in- >8 mL/kg for the conventional tidal volume ventilation group,
crease mortality, patients should be carefully managed in an from which the present results were obtained, so the above
environment where tracheal intubation can be conducted recommendations were made. The SR results had a very low
after the start of HFNC. certainty of evidence, but low tidal volume is thought to be
Contraindications for non-invasive respiratory support commonly used in daily clinical practice, so the recommen-
include the inability to protect the airway, high risk of vom- dation was changed to “strong recommendation” as a result of
iting, impaired consciousness, uncooperative patients, and re-voting at the panel meeting.
unstable hemodynamics.
Rationale
Rationale Summary of evidence:
Summary of evidence: By limiting tidal volume, short-term mortality (11 RCTs
Network meta-analysis was conducted using 19 RCTs [174e184]: N ¼ 1,795) occurred in 85 fewer/1,000 patients
(N ¼ 2,777) [155e173]. Short-term mortality (no direct com- (95% CI: 138 fewere24 fewer), ventilator-free days (VFD) (4
parison) occurred in 56 fewer/1,000 patients (248 fewere386 RCTs [178,180,181,183]: N ¼ 1,045) was extended by an
more patients); pneumonia (no direct comparison) occurred in average of 3.28 days (95% CI: 0.73 days shortere5.82 days
440 fewer/1,000 patients (478 fewere295 fewer patients). From longer), and barotrauma (7 RCTs [174e176,178,179,181,183]:
the above, the desirable effect of HFNC was judged to be N ¼ 1,551) was 1 more/1,000 patients (95% CI: 27 fewere37
“moderate.” more). From the above, the predicted desirable effect was
The risk of aspiration during HFNC is unclear, but pneu- judged as “moderate.” Meanwhile, there were no significant
monia incidence decreased, and this was not an undesirable outcomes for harms, and the undesirable effects were
effect. Positive pressure due to HFNC (60 L/min) was approx- judged as “do not know.” From the above, the balance of
imately 4 cmH2O, and the possibility of lung damage was low. effects and harms was judged to be “probably favors the
From the above, the undesirable effect of HFNC was judged to intervention.”
be “trivial.”
Certainty of evidence:
Certainty of evidence: Because the direction of outcomes among desirable effects
The direction of all effects was consistent, and the was not the same, the certainty of evidence for the overall
certainty of evidence for the overall outcome was outcome was judged as “very low,” which was the lowest
judged as “moderate” adopting the highest certainty of certainty of evidence of all the outcomes.
evidence.
Values, balance of effects, acceptability, and feasibility
Values, balance of effects, acceptability, and feasibility determination:
determination: The increased burden and cost to the patient of limiting the
The balance of effects of HFNC was judged as “probably amount of ventilation is considered to be small, and the
favors the intervention.” A dedicated device is needed for feasibility was judged as “yes”.
HFNC, but this is relatively cheap compared with a ventilator.
CQ 20: Should high levels of positive end-expiratory pres-
It is already implemented in the daily clinical practice of many
sure (PEEP) be used for mechanically ventilated adult patients
acute care hospitals, so it was judged that it was “probably
with ARDS?
yes” acceptable and feasible.
460 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

Background tidal volume or transpulmonary pressure is appropriately


The use of high levels of PEEP is a strategy for reducing restricted, then it may not be possible to state that high
ventilator-associated lung injury (VALI). plateau pressure is always harmful.

Recommendation Rationale
We suggest using high levels of PEEP for mechanically Summary of evidence:
ventilated adult patients with ARDS. A total of 6 RCTs [175,178,181,183,187,188] (N ¼ 2,882) were
(weak recommendation/very low certainty of evidence: found. By limiting plateau pressure, the following outcomes
GRADE 2D). were found to be beneficial: short-term mortality occurred in 6
more/1,000 patients (95% CI: 54 fewere81 more patients)
Supplementary item
[178,183,187,188], long-term mortality occurred in 4 fewer/1,000
In the studies included in this SR, the PEEP values on day 1
patients (95% CI: 99 fewere118 more patients) [175,181,188],
in the intervention group ranged from 10 to 16.3 cmH2O and
VFD was extended by an average of 0.82 days (95% CI: 4 days
from 6.5 to 12.0 cmH2O in the control group. The most com-
shortere5.65 days longer) [181,183,188], and barotrauma
mon methods of setting PEEP in the intervention group were
occurred in 11 fewer/1,000 patients (95% CI: 30 fewere12 more
setting by Pflex and setting by ARDSnet PEEP table.
patients) [175,178,181,187,188]. Therefore, the desirable effect of
Rationale the intervention was judged to be trivial. Meanwhile, with re-
Summary of evidence: gard to harmful outcomes, there were no reports of death due
With the use of high levels of PEEP, short-term mortality (15 to hypercapnia. In the RCT [187] included in this meta-analysis,
RCTs [174,180,183,185e196]: N ¼ 4,108) occurred in 55fewer/ adverse events included refractory acidosis (42/508) and deaths
1,000 patients (95% CI: 103 fewere0 fewer patients), VFD (11 RCTs due to refractory acidosis (38/508). Therefore, the undesirable
[180,183,185,186,188,189,191e195]: N ¼ 2,988) was extended by effects of the intervention was judged as “trivial.” Therefore,
an average of 1.82 days (95% CI: 0.37 days shortere4.01 days the balance of effects and harms was judged as “do not know.”
longer), and the length of hospitalization (6 RCTs [187,190e194]:
Certainty of evidence:
N ¼ 2,392) was extended by an average of 0.86 days (95% CI: 3.08
Because the direction of outcomes in desired and unde-
days shortere4.8 days longer). From the above, the predicted
sirable effects was inconsistent, the certainty of evidence for
desirable effect was judged as “moderate.” Meanwhile, baro-
the overall outcome was judged as “very low” by adopting the
trauma (10 RCTs [174,183,185e188,191e193,195]: N ¼ 2,861)
lowest certainty of evidence.
occurred in 1 more/1,000 patients (95% CI: 23 fewere39 more
patients). From the above, the undesirable effect was judged as Balance of effects, acceptability, and feasibility determination:
“trivial.” Therefore, the balance of effects and harms due to This is already used in daily clinical practice and is thought
intervention was judged as “probably favors the intervention.” to have sufficiently high acceptability and feasibility.

Certainty of evidence: CQ22: Which, between pressure-control ventilation (PCV)


Because the directions of desired and undesirable effects and volume-control ventilation (VCV), is desirable for me-
were not consistent, the certainty of evidence for the overall chanical ventilation in adult patients with ARDS?
outcome was judged as “very low” by adopting the lowest
Background
certainty of evidence.
The ventilation modes of commonly used ventilators are
Values, balance of effects, acceptability, and feasibility VCV and PCV, but it is unclear which between the ventilation
determination: modes is more beneficial for patient prognosis.
Because the increased burden on patients and cost of using
Recommendation
higher PEEP is considered to be small, the cost was judged as
From the results of this SR, we cannot provide a recom-
“small” compared with the benefit.
mendation on which ventilation mode between PCV and VCV
CQ21: Should plateau pressure be limited for mechanically should be used for adult patients with ARDS (in our practice
ventilated adult patients with ARDS? statement).

Background Supplementary item


Increased airway pressure is thought to be one of the fac- VCV has the advantage of detecting changes in compliance
tors causing VALI, and limiting plateau pressure is expected to and airway resistance and may be considered for use in fa-
suppress the development of VALI [5,178]. Meanwhile, cilities that are familiar with its use. The panel voted to make
limiting the plateau pressure may induce adverse events such this an “in our practice statement” without providing a
as hypercapnia. recommendation.

Recommendation Rationale
We suggest plateau pressure restrictions when using a Summary of evidence:
ventilator on adult patients with ARDS (weak recommenda- The use of PCV as a ventilation mode resulted in the length
tion/very low certainty of evidence: GRADE 2D). of mechanical ventilation shortening by an average of 4.1 days
(95% CI: 6.84 days shortere1.37 days shorter) [197e199], and
Supplementary item
mortality decreasing by 149/1,000 patients (95% CI: 270 few-
The present results may not suggest that desirable effects
ere11 more patients) [197e199]. From the above results, the
would always outweigh the undesirable effects, and if the
desirable effect of the intervention was judged to be
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 461

“moderate.” Meanwhile, VALI was not reported in the reports analysis using only classical APRV, the certainty of evidence
adopted in this SR. Therefore, we cited the meta-analysis of for the overall outcome was judged as “very low.”
Chacko et al. [200], which included 2 RCTs, which reported that
Balance of effects, acceptability, and feasibility determination:
VALI occurred in 23/1,000 patients (95% CI: 12 fewere72 more
Since it is difficult to conduct a typical APRV with other
patients) due to PCV, and the undesirable effect was judged to
than a specific model of ventilator, additional costs are
be trivial. As a result, it was judged that the effect of the
required. In addition, the appropriate use of APRVs may
intervention was probably greater than the harm.
require skilled staff, and therefore, the use of APRVs should be
Certainty of evidence: addressed on a facility-specific basis.
Because the direction of desirable and undesirable effects
CQ24: Which is preferable for ventilatory management of
was inconsistent, the certainty of evidence for the overall
adult patients with ARDS: synchronized intermittent manda-
outcome was judged as “very low” by adopting the lowest
tory ventilation (SIMV) or assisted controlled ventilation (A/C)?
certainty of evidence.
Background
Balance of effects, acceptability, and feasibility determination:
SIMV and A/C are available in standard ventilators, but it is
Since the potential for PCV to increase VALI is small, the
unclear which between the two is more effective.
benefits of PCV were considered to outweigh the harms. In
addition, the use of PCV does not increase costs or burdens, so Recommendation
it is considered to be sufficiently feasible. We cannot provide a recommendation from the results of
the present SR on which mode should be used in adult pa-
CQ23: Should airway pressure release ventilation (APRV)
tients with ARDS (in our practice statement).
be used in the mechanical ventilation of adult patients with
ARDS? Supplementary item
Considering the risks of increased asynchrony and
Background
extended time until ventilator removal, A/C is used more
APRV is frequently used for ARDS because it can maintain
often than SIMV.
high airway pressures, but it is unclear if APRV is more
beneficial than commonly used ventilation modes, such as Rationale
VCV and PCV. Summary of evidence:
With SIMV use, the length of mechanical ventilation
Recommendation
decreased by 0 days (7.41 fewer dayse7.41 more days) [208],
We cannot provide a recommendation on whether APRV or
and mortality decreased by 100/1,000 patients (272 fewere308
conventional ventilation modes should be used in adult pa-
more patients) [208]. From the above results, the expected
tients with ARDS. APRV could be considered when using me-
desirable effect was judged as “small.” Meanwhile, there was
chanical ventilation with spontaneous respiration (in our
an observational study that showed a high incidence of
practice statement).
asynchrony with the use of SIMV and a study demonstrating
Supplementary item that the time to ventilator weaning was longer when SIMV
The subgroup analysis that excluded inverse ratio venti- was used as a ventilator weaning mode [209e211]. From the
lation showed desirable effects on VFD, mortality, and baro- above, the expected undesirable effect was judged as “do not
trauma in the APRV group. However, the panel meeting could know.” Therefore, the balance of effects and harms was
not come to an agreement about making recommendation judged to be “probably favors the intervention.”
including acceptability and feasibility and decided to present
Certainty of evidence:
this as an in our practice statement.
The only RCT included in the meta-analysis had a small
Rationale sample size of 40, and mortality and length of mechanical
Summary of evidence: ventilation were secondary outcomes [208]. There was a trend
A meta-analysis was conducted with 7 RCTs [201e207]. toward higher midazolam use in the A/C group, which may
APRV resulted in VFD being extended by an average of 3.64 have influenced delirium, asynchrony, ventilator days, and
days (0.02 days shortere7.3 days longer) [201e207], mortality mortality. These findings made it difficult to make recom-
being decreased by 72/1,000 patients (125 fewere5 more pa- mendations for the use of SIMV and A/C in this meta-analysis.
tients) [201e207], and barotrauma occurring in 23 fewer/1,000
Balance of effects, acceptability, and feasibility determination:
patients (41 fewere42 more patients) [201,202,207]. From the
It is thought that the costs of changing the mode of me-
above results, the desirable effect due to the intervention was
chanical ventilation are low.
judged as “moderate.” Meanwhile, no clear harmful outcomes
were observed, and the undesirable effect was judged as “do CQ25: When using mechanical ventilation in adult pa-
not know.” From the above results, the effect of APRV was tients with ARDS with spontaneous breathing, is pressure
judged to be probably favours. The same results were obtained support ventilation (PSV) or A/C preferred?
in a meta-analysis of only classical APRV.
Background
Certainty of evidence: It is unclear which is more effective, PSV or A/C, which is
Because the direction of the desirable and undesirable ef- commonly used in patients with preserved spontaneous
fects was consistent in both the overall analysis and the breathing.
462 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

Recommendation Rationale
We cannot provide a recommendation on whether PSV or Summary of evidence:
A/C should be used in adults with ARDS. It is common practice When conducting the recruitment maneuver, VFD was
to select the ventilation mode based on the individual pa- extended by an average of 0.91 days (1.56 days shortere3.37
tient's condition and status (in our practice statement). days longer) [191e193,212e214], 28-day mortality decreased by
16/1,000 patients (61 fewere41 more patients) [174,187,
Supplementary item
190e193,212e217], and the intensive care unit (ICU) length of
The appropriate mode should be selected considering the
stay was shortened by an average of 1.03 days (2.58 days
presence of spontaneous breathing, use of sedatives or muscle
shortere0.53 days longer) [187,190e193,212e217]. From the
relaxants, and severity of ARDS in each patient.
above, the desirable effect was judged to be “trivial.” Mean-
Rationale while, with regard to harm outcomes, barotrauma occurred
Asynchrony between the patient and ventilator can lead to in 4 fewer/1,000 patients (27 fewere35 more patients)
lung injury from mechanical ventilation, which can adversely [174,187,190e193,212e214,216,217], and circulatory failure
affect hemodynamics and ultimately lead to patient death. occurred in 90 more/1,000 patients (3 fewere218 more patients)
PSV is a ventilation mode that preserves the patient's [191,193,218]. From the above, harms of the intervention were
spontaneous breathing and provides support during inspira- judged to be small. The balance of desired effects and harms
tion. Compared with the A/C mode, PSV may be less likely to was judged to be “probably favors the comparison.”
cause patient-ventilator asynchrony and result in lung injury.
Certainty of evidence:
Additionally, the dose of sedatives could also be reduced due
Because the outcome directions for the desirable and un-
to the increased comfort of breathing in PSV. However,
desirable effects were not consistent, the certainty of the ev-
because PSV is a ventilation mode that relies on spontaneous
idence for the overall outcome was judged to be “very low.”
breathing, if the patient's own spontaneous respiratory effort
is too strong, it may increase the tidal volume and compro- Balance of effects, acceptability, and feasibility determination:
mise the lung-protective ventilation. The burden and cost of the recruitment procedure is not
significant, as it only requires education and training of
Summary of evidence:
medical staff. In addition, there is a tendency that circulatory
No applicable studies
failure including cardiac arrest increases. Implementation of
Certainty of evidence: the maneuver should be carefully considered.
The quality of the evidence cannot be assessed because of
CQ27: Are ventilator weaning protocols useful in patients
the lack of relevant studies.
with mechanical ventilation?
Balance of effects, acceptability, and feasibility determination:
Background
Both ventilation modes are included in standard ventila-
Prolonged ventilation management may increase adverse
tors, and the required costs and resources are probably low.
events such as ventilator-associated pneumonia, whereas
CQ26: Should we perform a recruitment maneuver when premature extubation could increase reintubation and mor-
ventilating an adult patient with ARDS? tality rate [219]. Therefore, it is important to recognize the
timing of ventilator weaning at an early stage and in a reliable
Background
manner. Ventilator weaning is often done at the discretion of
The recruitment maneuver is a low-cost intervention that
the clinician, but protocolized weaning may be effective in
can be implemented at bedside and may improve oxygenation
reducing ventilator days [219].
and lung compliance, reducing the need for rescue therapies
such as veno-venous extracorporeal membrane oxygenation Recommendation
(VV-ECMO). Meanwhile, positive pressure ventilation at high We suggest protocolized ventilator weaning in adult pa-
pressure may cause circulatory failure and barotrauma. tients with ARDS (weak recommendation/very low certainty
of evidence: GRADE 2D).
Recommendation
We suggest against the routine use of the recruitment Supplementary item
maneuver in adult patients with ARDS It is important to educate medical staff when introducing a
(weak recommendation/very low certainty of evidence: protocol and making new changes to ventilator settings with
GRADE 2D). multidisciplinary medical staff. It is also necessary to pay
close attention to patient monitoring.
Supplementary item
Adequate education and training are required to perform Rationale
the recruitment maneuver. If the circulation is adequately Summary of evidence:
monitored and critical situations, such as cardiopulmonary Because there were no RCTs that included only patients
arrest, can be adequately addressed, a recruitment maneuver with ARDS, we included studies of patients who were on
may be performed to improve the P/F ratio and avoid rescue mechanical ventilation for more than 24 h [220e232]. With the
therapy. use of the protocol, in-hospital mortality occurred in 10 more/
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 463

1,000 patients (30 fewere57 more) [221,223e226,228,232], ICU ventilator-free days were shortened by an average of 0.5 days
mortality occurred in 47 fewer/1,000 patients (80 fewere2 (1 RCT: N ¼ 795; 95% CI: 1.71 days shortere0.71 days longer)
more patients) [222,224,227,232], duration of mechanical [236]; and barotrauma occurred in 30 more/1,000 patients (4
ventilation was shortened by an average of 29.33 h (47.98 h RCTs: N ¼ 784; 95% CI: 15 fewere96 more patients)
shortere10.69 h shorter) [220e230,232], hospital stay was [233e235,237]. From these, the undesirable effect was judged
shortened by an average of 1.19 days (2.92 days shortere0.55 to be “small.” From the above results, the balance of effects
days longer) [221,223,226,228,229], ICU length of stay was was judged to be that “probably favors the comparison.”
shortened by an average of 17.84 h (29.67 h shortere6.02 h
Certainty of evidence:
shorter) [221,222,224,226e230,232], tracheostomy was per-
The direction of outcomes of desirable effects and unde-
formed in 34 fewer/1,000 patients (58 fewere1 fewer patients)
sirable effects was consistent, and the certainty of evidence
[221,225e228,230], and reintubation was performed in 6 fewer/
for the overall outcome was judged as “high” by adopting the
1,000 patients (36 fewere36 more patients) [220,221,223,
highest of all the outcomes.
224,226,227,230,232]. Therefore, the expected desirable effect
was judged to be “trivial.” Meanwhile, there were no harm Values, balance of effects, acceptability, and feasibility
outcomes, so the undesirable effect was judged to be “do not determination:
know.” From the above results, we concluded that the effects The use of HFOV in adult patients requires a specialized
were probably favors the intervention. mechanical ventilator and skilled medical staff. Judging from
the overall balance of equipment, human cost, and effective-
Certainty of evidence:
ness, there would be little benefit in introducing HFOV to a
The direction of the outcomes within the desirable effect
facility that is not familiar with its use.
was inconsistent, so the certainty of the evidence for the
overall outcome was judged to be “very low.” CQ29: Should driving pressure be used as an index when
implementing mechanical ventilation in adult patients with
Balance of effects, acceptability, and feasibility determination:
ARDS?
Except for the use of special ventilatory modes, the costs of
implementing the protocol are trivial. Meanwhile, creating Background
protocols and training medical staff may take time and have Driving pressure is associated with survival in the venti-
associated costs. latory management of patients with ARDS [238]. Limiting the
driving pressure may reduce VALI rate [239]. Meanwhile,
CQ28: Should high-frequency oscillatory ventilation
limiting driving pressure may increase harms such as hyper-
(HFOV) be used for adult patients with ARDS?
capnia and acid-base imbalance. Therefore, there is clinical
Background significance in investigating the usefulness of setting driving
It is important to prevent VALI in adult patients with ARDS. pressure as an index when conducting mechanical ventilation
HFOV is a mechanical ventilation mode that restricts the tidal in patients with ARDS.
volume and enables lung recruitment, and it is thought to be a
Recommendation
type of lung protective ventilation. However, it is not a com-
We cannot provide a recommendation on whether driving
mon mechanical ventilation method for adult patients with
pressure should be used as an index when conducting me-
ARDS, and there is a need to verify its effectiveness and safety.
chanical ventilation in adult patients with ARDS. There is
Recommendation increased importance of restricting driving pressure in
We suggest against implementing HFOV for adult patients ensuring standard lung protective ventilation that restricts
with moderate or severe ARDS (weak recommendation/high tidal volume or plateau pressure based on ideal weight while
certainty of evidence: GRADE 2A). paying attention to the harms such as hypercapnia and acid-
base imbalance (in our practice statement).
Supplementary item
This recommendation is for the use of HFOV in adult pa- Rationale
tients with typical moderate to severe ARDS (P/F ratio Summary of evidence:
200 mmHg). A total of 13 observational studies met the inclusion
criteria, and there were no RCTs that met the inclusion criteria
Rationale
[145,238e249]. Low driving pressure is expected to reduce
Summary of evidence:
VALI rate; meanwhile, there are concerns of harms such as
Mechanical ventilation by HFOV affected the following
increased respiratory rate, atelectasis, hypercapnia, and res-
items defined as important outcomes: refractory hypoxemia,
piratory acidosis. Low driving pressure is associated with
38 more/273 patients to 19 fewer/275 patients [233]. Therefore,
prevention of lung overextension or hyperinflation, decrease
the desirable effect was judged as “trivial.” Meanwhile, the
in VALI rate, and decrease in mortality rate. However,
effects on harmful outcomes were as follows: short-term
although driving pressure is a prognostic indicator, it is un-
mortality occurred in 27 more/1,000 patients (5 RCTs:
clear whether using driving pressure as a marker for ventila-
N ¼ 1,612; 95% CI: 61 fewere133 more patients) [233e237]; in-
tion settings improves outcomes for patients with ARDS. The
hospital mortality occurred in 64 more/1,000 patients (2 RCTs:
balance between the positive and negative effects of using
N ¼ 1,343; 95% CI: 47 fewere215 more patients) [233,236]; ICU
driving pressure as an index for management is unclear, and
length of stay was extended by an average of 1.5 days (1 RCT:
no clear recommendations can be made in this CQ. For this
N ¼ 795; 95% CI: 0.71 days shortere3.71 days longer) [236];
464 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

reason, this CQ is not an evidence-based recommendation, Values, balance of effects, acceptability, and feasibility
but rather an in our practice statement. determination:
Intervention is change in SpO2 target and has no cost.
Certainty of evidence:
Management of patients with chronic obstructive pulmonary
Since there are no relevant studies, the quality of evidence
disease and others with low SpO2 is common and may be
cannot be evaluated.
feasible in patients with ARDS.
Values, balance of effects, acceptability, and feasibility
determination: D. Area D: Treatment adjacent to ventilator use
Since there is no cost to buy additional equipment to
measure driving pressure, it can be implemented with edu- CQ31: Should neuromuscular blockers be used at an early
cation and manpower. phase in adult patients with moderate or severe ARDS?

CQ30: Is low SpO2 (PaO2) a target for management in adult Background


patients with ARDS? The use of neuromuscular blockers for adult patients
with moderate or severe ARDS has been suggested to
Background
improve the prognosis of patients with ARDS by reducing
Oxygen therapy is essential in adult patients with ARDS,
VALI rate [257,258]. However, there are concerns such as
and SpO2 monitoring during treatment is essential. Recent
delays in early mobilization, ICU-AW, and impaired quality
RCTs have shown that management that targets lower SpO2
of line (QOL).
improves mortality, whereas some studies have shown an
increase in mortality, and the effect of management targeting Recommendation
low SpO2 is inconsistent [250]. Therefore, it is necessary to We suggest administering neuromuscular blockers at an
verify the benefits and harms of management targeting low early phase for adult patients with moderate or severe ARDS
SpO2 in patients with ARDS and to define a target SpO2 value. (weak recommendation/very low certainty of evidence:
GRADE 2D).
Recommendation
We suggest against conducting management that targets Supplementary item
an excessively low SpO2 (PaO2) in adult patients with ARDS The administration of neuromuscular blockers should be
(weak recommendation/very low certainty of evidence: limited to early-onset patients with moderate or worse ARDS
GRADE 2D). for no longer 48 h. The drug mainly used in international RCTs
(cisatracurium) is not available in Japan. Alternative drugs in
Supplementary item
Japan include the aminosteroid neuromuscular blockers
The optimal SpO2 (PaO2) is currently unknown. Manage-
(rocuronium and vecuronium). Care should be taken when
ment that avoids excessively low or high oxygen levels should
using aminosteroids because the risk of ICU-AW could in-
be conducted.
crease. Also, particular attention should be given to concom-
Rationale itant use with corticosteroids.
Summary of evidence:
Rationale
There were 6 RCTs [251e256] (N ¼ 2,337) comparing man-
Summary of evidence:
agement of adult ventilated patients with low SpO2 (PaO2)
There were 5 RCTs [257e261] that were in line with PICO. As
versus high SpO2 (PaO2) goals. Favorable outcomes of man-
for the effect estimates, survival (5 RCTs: N ¼ 1,461) [257e261]
agement targeting low SpO2 included long-term mortality (18
had a risk difference of 84 more/1,000 patients (95% CI: 39
fewer/1,000 patients; 95% CI: 77 fewere51 more patients)
fewere236 more/1,000 patients), QOL (EQ-5D, etc.) (1 RCT:
[251e256], VFD (0.25 days longer; 95% CI: 1.76 days short-
N ¼ 246) [258] had a mean of 0.02 points lower (95% CI: 0.09
ere2.27 days longer) [251,254,255], and ICU-acquired weak-
lesse0.05 more points), and barotrauma (4 RCTs: N ¼ 1,437)
ness (ICU-AW) (51 fewer/1,000 patients; 95% CI: 80 fewere4
[257e260] had a risk difference of 31 fewer/1,000 patients (95%
more patients) [251]. As a result, the desirable effect was
CI: 46 fewere9 fewer/1,000 patients). The desirable effect due
judged to be “small.” Meanwhile, the results were as follows
to intervention was judged as “moderate”. ICU-AW (4 RCTs:
for harmful outcomes: new arrythmia (26 more/1,000 patients;
N ¼ 747) [257e260] had a risk difference of 25 more/1,000 pa-
85% CI: 7 fewere84 more patients) [251,252] and intestinal
tients (95% CI: 58 fewere125 more/1,000 patients). The unde-
ischemia (21 more/1,000 patients; 95% CI: 20 fewere866 more
sirable effects were judged as “small.” From the above, the
patients) [251,252]. As a result, the undesirable effect was
balance of effects was judged as “probably favors the
judged to be “small.” Based on the above results, the balance
intervention.”
of effects was judged to be “do not know.”
Certainty of evidence:
Certainty of evidence:
The certainty of evidence of the overall outcome
Because the direction of outcomes was inconsistent, the
used the lowest certainty of evidence and was judged as “very
certainty of evidence for the overall outcome was judged as
low.”
“very low” by adopting the lowest of all the outcomes.
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 465

Values, balance of effects, acceptability, and feasibility Values, balance of effects, acceptability, and feasibility
determination: determination:
Neuromuscular blockers are drugs that are easy to acquire Measuring transpulmonary pressure requires a specific
and use, and they have minimal costs. They are already used ventilator, so its implementation is difficult for some facilities.
in clinical practice and are thought to be sufficiently feasible.
CQ33: Should electrical impedance tomography (EIT) be
CQ32: Should transpulmonary pressure be used when used in PEEP settings for patients with ARDS?
setting PEEP in patients with ARDS?
Background
Background There is no established method regarding appropriate
Excessive PEEP in patients with ARDS enhanced VALI and PEEP settings in patients with ARDS. EIT, which visualizes
also has a negative effect on hemodynamics [189,262]. gas distribution in the lungs by measuring impedance
Meanwhile, excessively low PEEP has also been reported to changes in the body, can be used to extract the ventilation
increase lung collapse, which may enhance VALI and worsen status of each lung site, and PEEP could be appropriately set
oxygenation [189,262]. Nevertheless, there is no established by searching for PEEP where the lungs are uniformly aerated
method for setting an appropriate PEEP. Clarifying a useful [263]. Clarifying a useful method for PEEP settings is a key
method for PEEP settings is a key clinical issue clinical issue.

Recommendation Recommendation
We suggest against using transpulmonary pressure as a We cannot recommend the use of EIT in PEEP settings for
routine basis in PEEP settings for patients with ARDS. patients with ARDS in our daily practice (in our practice
(weak recommendation/moderate certainty of evidence: statement).
GRADE 2B).
Supplementary item
Supplementary item As of February 2021, EIT has not been released in Japan.
The results of this SR indicated that transpulmonary There is no evidence that compares EIT to conventional PEEP
pressure-based PEEP setting is associated with decreased 28- decision methods for setting the appropriate PEEP, but there is
day mortality and long-term mortality. However, because its a possibility that EIT may help to set the appropriate PEEP for
measurement requires a specific ventilator, it is difficult to uniform lung ventilation.
recommend this procedure for routine measures. Therefore,
Rationale
the above recommendation was given. This recommendation
Summary of evidence:
does not reject the use of transpulmonary pressure under
A literature research was systematically conducted, but
circumstances where the esophageal pressure could be
there were no studies that compared EIT-based PEEP decisions
measured.
and other methods as an important outcome. Therefore, a
Rationale clear recommendation cannot be suggested for this CQ. This
Summary of evidence: CQ is not an evidence-based recommendation but instead a
The results of this SR showed that there were 2 RCTs description of the current state of clinical practice.
[189,262] that compared transpulmonary pressure-based PEEP
Certainty of evidence:
settings and conventional ARDSnet table-based PEEP decision
Since there are no applicable studies, the quality of evi-
methods in patients with ARDS. As for the effect estimates for
dence cannot be evaluated.
short-term mortality (2 RCTs: N ¼ 261) [189,262] with trans-
pulmonary pressure-based PEEP settings, the risk difference Values, balance of effects, acceptability, feasibility
was 85 fewer/1,000 patients (95% CI: 225 fewere247 more/ determination:
1,000 patients) [189,262]; and for long-term mortality (2 RCTs: As of February 2021, the EIT has not been released in Japan
N ¼ 259) [189,262], the risk difference was 72 fewer/1,000 pa- and is an experimental device. When it is released in the
tients (95% CI: 207 fewere148 more/1,000 patients). From the future, the cost is expected to be excessive.
above, the desirable effect transpulmonary pressure-based
CQ34: Should pulmonary ultrasound be used for PEEP
PEEP setting was judged as “small.” As for harms, the fre-
settings in patients with ARDS?
quency of pneumothorax or barotrauma did not increase with
transpulmonary pressure-based PEEP settings (pneumo- Background
thorax: 2/98 vs. 3/102, absolute difference of 0.9% (3.4%e In patients with ARDS, excessive PEEP can increase VALI
5.2%); barotrauma: 5/98 vs. 6/102, absolute difference of 0.8% and adversely affect circulatory dynamics [189,262]. On the
(5.5%e7.1%); it was also reported that there were no compli- other hand, extremely low PEEP also increases collapsed lung,
cations associated with esophageal pressure balloon place- and lower PEEP also has been reported to affect VALI and
ment [262]. Therefore, the undesirable effect was judged as worsen oxygenation [189,262]. There is no established method
“small.” From the above, the balance of effects was judged as for setting an appropriate PEEP. Clarifying a useful method for
“do not know.” PEEP settings is a key clinical issue.

Certainty of evidence: Recommendation


The certainty of evidence of the overall outcome adopted It is not common to use pulmonary ultrasound in the PEEP
the lowest certainty of evidence of all outcomes and was settings of patients with ARDS (in our practice statement).
judged as “moderate.”
466 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

Supplementary item intervention was judged to be “moderate.” Meanwhile, as for


Evaluating the non-ventilated area of the lung with pulmo- harmful outcomes, the effect estimates for tracheal tube
nary ultrasound requires technique and skill, and there is also problems (2 RCTs: N ¼ 808) [264,270] was 83 more/1,000 patients
no evidence that compares this with usual PEEP setting in the prone position when compared to patients not in the
methods. There is also the possibility of excessive PEEP levels prone position (95% CI: 30 moree151 more/1,000 patients), and
based on the results of pulmonary ultrasound. Meanwhile, the number of pressure-induced skin disorders and ulcers (2
pulmonary ultrasound can help assess PEEP configuration RCTs: N ¼ 344) [265,267] increased by 114/1,000 patients (95% CI:
because it can assess collapsed lung recruitment due to elevated 19 moree237 more/1,000 patients), and the predicted unde-
PEEP, is non-invasive, and does not require additional costs. sirable effect was judged to be “small.” From the above, the
balance of effects was judged as “Probably favors the
Rationale
intervention.”
Summary of evidence:
No applicable studies Certainty of evidence:
Because the direction of desirable and undesirable effects
Balance of effects, acceptability, feasibility determination:
is not consistent, the certainty of evidence for the overall
Ultrasounds are equipped in each facility that conducts
outcome was judged as “very low,” which was the lowest of all
clinical practice for ARDS, and it is thought that additional costs
the outcomes.
of resources are not needed. Pulmonary ultrasound itself is non-
invasive and does not require additional costs, so it is easy to Balance of effects, acceptability, and feasibility determination:
implement; however, it is thought that evaluating lung aeration Repositioning the patient to the prone position requires more
with pulmonary ultrasound requires technique and skill, and it manpower than usual, but there is no need to purchase addi-
cannot be said that this method is unequivocally feasible. tional expensive equipment, so cost is minimal. Meanwhile,
there are concerns of the onset of tracheal tube displacement or
CQ35: Should prone positioning be used in adult patients
dislocation and skin damage due to the prone position, but its
with moderate or severe ARDS?
harms are trivial. Although there is no evidence in Japan, this is
Background already implemented in clinical practice. Even when consid-
Changing the position of patients with ARDS from supine ering costs, it is thought to be feasible if conducted in a facility
to prone position is thought to be a remedy for severe hyp- where there are well-trained staff in ARDS management.
oxemia because it improves oxygenation. Additionally, it has
CQ36: Should ECMO be conducted in adult patients with
been suggested that prone positioning has the effects of pre-
severe ARDS?
venting the onset or progression of VALI, and it has the pos-
sibility of improving patient prognosis [264]. The prone Background
positioning itself does not require special equipment, but Introducing ECMO to patients with severe ARDS is ex-
complications could occur by changing the body repositioning pected to reduce the mortality rate through rescue effects for
and long-term management [264]. Given these contexts, severe respiratory failure, where maintaining oxygenation is
clarifying the benefits and harms of the prone positioning in difficult with normal mechanical ventilation [271,272]. ECMO
patients with ARDS is a key clinical issue. may also minimize VALI. However, its effectiveness has not
been established due to the uncertainty of indications and
Recommendation
timing as well as the presence of many complications. Clari-
We suggest performing the prone positioning in adult pa-
fying the relationship between ECMO introduction and mor-
tients with moderate or severe ARDS for long periods of time
tality as well as adverse events are key clinical issues.
(weak recommendation/very low certainty of evidence:
GRADE 2D). Recommendation
We suggest implementing ECMO for adult patients with
Supplementary item
severe ARDS (weak recommendation/moderate certainty of
Prone positioning needs to be done in hospitals with
evidence: GRADE 2B).
experienced staffs. Additionally, long implementation times
(over 12 h) should be considered when placing patients in the Supplementary item
prone position. It is desirable to consult an experienced facility or
specialist regarding ECMO indications and patient transport in
Rationale
cases of severe hypoxemia or hypercapnia that is resistant to
Summary of evidence:
standard lung protective ventilation or adjuvant therapies
The results of SR for beneficial outcomes showed that the
such as muscle relaxants and prone positioning.
effect estimates for short-term mortality (7 RCTs: N ¼ 2,118)
[264e270] was 69 fewer/1,000 patients in the prone position Rationale
when compared to patients not in the prone position (95% CI: Summary of evidence:
135 fewere13 more/1,000 patients), mechanical ventilation There were 2 RCTs [271,272] that comparatively investi-
duration (3 RCTs: N ¼ 871) [266,267,269] was shortened by an gated whether ECMO or standard treatment with conven-
average of 0.48 days (95% CI: 1.61 days shortere0.65 days tional mechanical ventilation should be conducted for severe
longer), and ICU length of stay (3 RCTs: N ¼ 518) [268e270] was patients with ARDS. For beneficial outcomes, the effect esti-
extended by an average of 1.03 days (95% CI: 1.7 days short- mates for 60-day mortality (2 RCTs: N ¼ 429) [271,272] was 133
ere3.75 days longer), and the desirable effect due to fewer/1,000 patients with ECMO implementation (95% CI: 204
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 467

fewere43 fewer/1,000 patients), and the desirable effect due to Rationale


intervention was judged as “moderate.” Meanwhile, for Summary of evidence:
harmful outcomes, the effect estimates for stroke (1 RCTs: As for beneficial outcomes, the effect estimates for short-
N ¼ 249) [271] was 40 fewer/1,000 patients (95% CI: 58 fewere25 term mortality (14 RCTs: N ¼ 2,887) [276e289] was 37 fewer/
more/1,000 patients). However, considering the possibility 1,000 patients with early tracheostomy compared to pa-
that there were 25 more patients/1,000 patients due to inter- tients without early tracheostomy (95% CI: 70 fewere0
vention, the undesirable effect was judged as “small.” As an fewer/1,000 patients), ventilator-associated pneumonia
“important” outcome, bleeding-related complications tended (VAP) (8 RCTs: N ¼ 1,527) [279,281e284,286e288] was 67
to increase. Therefore, the balance of effects was judged as fewer/1,000 patients (95% CI: 120 fewere5 more/1,000 pa-
“probably favors the intervention.” tients), and VFD (4 RCTs: N ¼ 1,243) [281,282,284,286] was
extended by an average of 1.2 days (95% CI: 0.57 days lon-
Certainty of evidence:
gere1.82 days longer). From the above, the desirable effect
Because the direction of desirable and undesirable effects
due to intervention was judged as “small.” Meanwhile, for
is not consistent, the certainty of evidence for the overall
the harmful outcomes, the effect estimates associated with
outcome was judged as “moderate,” which was the highest of
tracheostomy-related bleeding (5 RCTs: N ¼ 1,715)
all the outcomes.
[278,281,282,284,285] was 7 more/1,000 patients with early
Balance of effects, acceptability, and feasibility determination: tracheostomy compared to patients without early trache-
There are cost concerns with ECMO implementation. The ostomy (95% CI: 5 fewere30 more/1,000 patients), and tra-
introduction and management of ECMO requires staff from cheostomy infection (4 RCTs: N ¼ 816) [278,281,282,284] was
multiple disciplines with specialized knowledge and skill, and 12 more/1,000 patients (95% CI: 6 fewere54 more/1,000 pa-
human resource must also be considered. In the Western tients), and the predicted harm was judged as “trivial.”
countries, when a patient indicated for ECMO was present in a Therefore, the balance of effects and harms was judged as
facility where ECMO cannot be conducted, the patient is “probably favors the intervention.”
transferred to an ECMO center, thereby centralizing ECMO
Certainty of evidence:
procedures. Attempts are also made in Japan toward this
Because the direction of desirable and undesirable effects
centralization of ECMO treatment.
is not consistent, the certainty of evidence for the overall
CQ37: Should early tracheostomy be performed in adult outcome was judged as “very low,” which was the lowest of all
patients with ARDS? the outcomes.

Background Balance of effects, acceptability, and feasibility determination:


Tracheostomy has the advantage of reducing the admin- Tracheostomy has relatively low costs, is a procedure
istration of sedatives or analgesics as well as avoiding vocal within the scope of usual clinical practice, and does not
cord injury when compared to tracheal intubation [273,274]. require the purchase of additional equipment or the addi-
However, tracheostomy may be associated with bleeding or tional human resources, so the adverse effects due to
removal difficulties due to airway narrowing. It has also been intervention are thought to be small. Meanwhile, early tra-
reported that early tracheostomy within 14 days after intu- cheostomy may result in unnecessary tracheostomies being
bation may improve prognosis [275]. Therefore, clarifying conducted.
whether early tracheostomy improves patient prognosis in
CQ38: Should a VAP prevention bundle be routinely con-
patients with ARDS where the length of mechanical ventila-
ducted for adult patients with ARDS?
tion can be extended to a long period of time is a key clinical
issue. Background
VAP is pneumonia that newly occurs 48 h after
Recommendation
the initiation of mechanical ventilation and onwards, and it
We suggest performing early tracheostomy in adult pa-
also includes pneumonia that occurred within 48 h
tients with ARDS (weak recommendation/very low certainty
following patient withdrawal from a ventilator [290]. VAP is
of evidence: GRADE 2D).
one of the most important complications in mechanically
Supplementary item ventilated patients, and it is important to implement
There is no clear definition of early tracheostomy, with appropriate preventive measures for VAP. The VAP pre-
many studies defining this from within 48 h to within 10 days vention bundle, which involves conducting a series of care
after the initiation of mechanical ventilation, and early tra- measures as a set, has been proposed as a preventive
cheostomy could be considered if the general condition of the method for VAP, but its effects and certainty of evidence are
patient is stable. Meanwhile, there are concerns with early unclear.
tracheostomy that it may be conducted on patients who did
Recommendation
not require it in the first place. Additionally, the tracheostomy
We recommend conducting a VAP prevention bundle
itself may involve high levels of risk in patients with ARDS
routinely in adult patients with ARDS on a ventilator (GPS).
with high oxygen concentrations, airway pressure, or PEEP;
therefore, careful consideration is needed for adaptation to Supplementary item
the patient in this recommendation. Bundle contents should be considered in each facility.
468 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

Rationale CQ40: Should nitric oxide inhalation be used for patients


Summary of evidence: with ARDS?
The predicted desirable effect due to VAP prevention
Background
bundle implementation was judged as moderate. We were not
It has been reported that nitric oxide (NO) inhalation,
able to find any RCTs that directly compared whether to
which has a pulmonary vessel dilatation effect, was effective
perform VAP prevention bundles for adult patients with ARDS
in improving oxygenation in ARDS; meanwhile, it has also
requiring mechanical ventilation. However, multiple obser-
been reported that it did not improve the length of mechanical
vational studies reported that the occurrence of VAP was
ventilation or mortality [294]. NO is sometimes used as a
reduced through the implementation of VAP prevention
rescue therapy when oxygenation does not improve [295];
bundles, and the balance of effects was judged as “probably
therefore, conducting an SR regarding NO inhalation is a key
favors the intervention.”
clinical issue.
Balance of effects, acceptability, and feasibility determination: Recommendation
Additionally, the balance of desirable and undesirable ef- We suggest against using NO inhalation in patients with
fects was judged as “probably favors the intervention.” The ARDS (weak recommendation/very low certainty of evidence:
VAP prevention bundle has been taken up in the joint action GRADE 2D).
on medical safety in Japan and has spread nationwide, so it
would be accepted by major stakeholders. There are many Supplementary item
facilities implementing VAP prevention bundles as part of This recommendation does not reject the use of NO inha-
daily clinical practice, and it was judged to be feasible. lation as rescue therapy in facilities that already use this
treatment.
E. Area E: Drug therapy/non-drug therapy Rationale
Summary of evidence:
CQ39: Should thrombomodulin be used in patients with As a result of implementation of NO inhalation, the length
ARDS? of mechanical ventilation (2 RCTs: N ¼ 77) [296,297] shortened
Background by an average of 1.77 days (95% CI: 3.55 days shortere0.02 days
It has been reported that thrombomodulin, which is used longer) and ICU length of stay (1 RCT: N ¼ 40) [296] shortened
for disseminated intravascular coagulation treatment, may by an average of 0.7 days (95% CI: 38.28 days shortere36.88
have anti-inflammatory effects as well as anticoagulant ef- days longer). Therefore, it was thought that there were mini-
fects [291e293]. Therefore, thrombomodulin may be effective mal predicted desirable effects (judged as “trivial” benefit).
in suppressing inflammation in ARDS. Meanwhile, as a result of implementation of NO inhalation,
mortality (7 RCTs: N ¼ 878) [295e301] occurred in 40 more/
Recommendation 1,000 patients (95% CI: 16 fewere109 more patients) and renal
We cannot provide a recommendation for the adminis- dysfunction (1 RCT: N ¼ 180) [299] in 132 more/1,000 patients
tration of thrombomodulin in patients with ARDS. (in our (95% CI: 10 moree373 more patients). As a result, the predicted
practice statement). undesirable effect was judged as “moderate.”
Rationale
Certainty of evidence:
Summary of evidence:
Although NO inhalation increased the harms (mortality
We conducted an SR, but found no RCTs to assess the effect
and renal dysfunction), the length of mechanical ventilation
of thrombomodulin in only patients with ARDS.
and ICU length of stay were shortened. Because the direction
Certainty of evidence: of desirable and undesirable effects is not consistent, the
Since there are no applicable studies, the quality of evi- certainty of evidence was judged as “very low” that was the
dence cannot be evaluated. lowest among all outcomes.

Balance of effects, acceptability, and feasibility determination: Balance of effects, acceptability, and feasibility determination:
Since there are no applicable studies, the balance of effects Given that mortality increased when NO was used, the
is unknown. Considering the lack of evidence and its off-label harms were judged to outweigh the benefits. Given the need
use, thrombomodulin use in patients with ARDS is likely un- for ethics committee approval and specialized equipment, it is
acceptable. Additionally, ethics committee approval will be predicted that there are a limited number of medical in-
needed when administering thrombomodulin for ARDS, since stitutions where this is feasible.
this such use will not be covered by the national health in-
CQ41: Should sivelestat be used for patients with ARDS?
surance. Therefore, an unequivocal statement cannot be
made regarding feasibility. The cost of 6 days of thrombo- Background
modulin administration is approximately $2100 (¥240,000). The pathogenesis of ARDS is a permeability pulmonary
There is a possibility of increased burdens on patients, hos- edema due to non-specific inflammation where neutrophil
pitals, and payment institutions. elastase is involved as an important mediator [302,303]. It is an
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 469

important clinical issue to test whether sivelestat, a neutro- administration at an early stage and continuing this for at
phil elastase inhibitor, is effective in ARDS. least 7 days [310].

Recommendation Rationale
We suggest against using sivelestat in patients with ARDS. Summary of evidence:
(weak recommendation/very low certainty of evidence: High-dose corticosteroids
GRADE 2D). For desirable effects, long-term mortality (1 RCT [311]:
N ¼ 99) decreased by 32/1,000 patients (95% CI: 196 fewere183
Rationale
more patients), so this was judged as “trivial.” For undesirable
Summary of evidence:
effects, infection (1 RCT [311]: N ¼ 99) occurred in 58 more/
As a result of sivelestat use, mortality (4 RCTs [304e307]:
1,000 patients (95% CI: 46 fewere353 more patients), so this
N ¼ 557) occurred in 5 fewer/1,000 patients (95% CI: 67 few-
was judged as “small.”
ere78 more patients), length of mechanical ventilation (2
Low-dose corticosteroids
RCTs [306,308]: N ¼ 38) was shortened by an average of 4.7
For desirable effects, long-term mortality (5 RCTs [312e316]:
days (95% CI: 13.11 days shortere3.64 days shorter), and the
N ¼ 769) decreased by 105/1,000 patients (95% CI: 178 fewere7
ICU length of stay (1 RCT [304]: N ¼ 24) was shortened by an
fewer patients), infection (5 RCTs [312e316]: N ¼ 769) occurred
average of 5 days (95% CI: 21.26 days shortere11.26 days
in 50 fewer/1,000 patients (95% CI: 97 fewere6 more patients),
shorter). The predicted desirable effect of sivelestat was
and VFD (5 RCTs [312e316]: N ¼ 767) increased by an average of
judged as “small,” and the harms were also thought to be
4.75 days (95% CI: 2.97 days longere6.54 days longer), hospital
“small.”
length of stay (2 RCTs [313,314]: N ¼ 271) shortened by an
Certainty of evidence: average of 5.04 days (95% CI: 9.43 days shortere0.65 days
There was no effect of the intervention on mortality, shorter), and ICU length of stay (2 RCTs [313,314]: N ¼ 271)
whereas there was a beneficial effect of the intervention on shortened by an average of 5.23 days (95% CI: 9.64 days short-
the lengths of mechanical ventilation and ICU stay. Therefore, ere0.82 days shorter), so this was judged as “moderate.” No
the direction of desirable and undesirable effects is not undesirable effects were observed at low doses. Therefore,
consistent, and the certainty of evidence was judged as “very when considered together with the description in the addi-
low.” tional considerations, it was judged to be “trivial.”

Balance of effects, acceptability, and feasibility determination: Certainty of evidence:


The balance of effects of intervention was judged as “do High dose corticosteriods
not know.” The benefits of the intervention were not clear, Because the direction of desirable effects and undesirable
whereas the costs could increase. Thus, the intervention was effects was not consistent, the certainty of evidence was
judged to be likely unacceptable. The drug is readily acces- judged as “very low,” which was the lowest of all the
sible, and the feasibility of the intervention was set as high. outcomes.
Low dose corticosteriods
CQ42: Should corticosteroids be used for adult patients
As the direction of desirable effects and undesirable effects
with ARDS?
was consistent, the certainty of evidence was judged as
Background “moderate,” which was the highest of all the outcomes.
The pathogenesis of ARDS is a permeability pulmonary
Balance of effects, acceptability, and feasibility determination:
edema due to non-specific inflammation [309], and whether
High-dose steroid administration can be harmful, and is
anti-inflammatory corticosteroids improve clinical outcomes
therefore thought to be unacceptable. Effects of the inter-
is an important clinical question.
vention such as decreased mortality, VFD extension, short-
Recommendation ened ICU length of stay, and shortened in-hospital stay could
We suggest against using high-dose corticosteroids for be expected for low-dose steroid administration, and it was
adult patients with ARDS. judged as economically acceptable. Steroid administration is
(weak recommendation/very low certainty of evidence: covered by insurance for septicemia and severe pneumonia,
GRADE 2C). which are the major causes of ARDS, and its feasibility was
judged to be high.
We recommend using low-dose corticosteroids for adult
patients with ARDS. CQ43: Should early rehabilitation intervention be con-
(strong recommendation/moderate certainty of evidence: ducted for adult patients with ARDS?
GRADE 1B).
Background
Supplementary item Post-intensive care syndrome (PICS) is more likely to be a
The types and dose of steroids for ARDS vary from study to problem for patients with severe illnesses such as ARDS [317].
study. The high dose is approximately 30 mg/kg in terms of Early rehabilitation is provided during ICU stay to prevent
methylprednisolone, and the low dose is approximately PICS. It has been reported that early rehabilitation improved
1e2 mg/kg. Sub-group analyses and previous meta-analyses physical function in severely ill patients and mechanically
suggested the usefulness of starting low-dose steroid ventilated patients [318,319]. Therefore, whether early
470 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

rehabilitation intervention should be conducted for patients excluding patients requiring deep sedation and should be
with ARDS is an important clinical question. noted during implementation. Because there were also no
studies focused only on patients with ARDS, care must be
Recommendation
taken in indications for the interventions.
We suggest conducting early (i.e., within 72 h) rehabilita-
tion for adult patients with ARDS (weak recommendation/low Rationale
certainty of evidence: GRADE 2C). Summary of evidence:
No studies were identified that included only patients with
Rationale
ARDS. As a result of non-sedative/light-sedative management,
Summary of evidence:
the number of patients who survived and got discharged (2
With implementation of early rehabilitation, mortality (6
RCTs [327,328]; N ¼ 242) increased by 28/1,000 patients (90
RCTs [320e325]: N ¼ 883) decreased by 2/1,000 patients (95%
fewere172 more patients), and ventilator-free days (2 RCTs
CI: 46 fewere57 more patients), ICU-AW (2 RCTs [325,326];
[328,329]; N ¼ 1,287) was extended by an average of 2 days (0.91
N ¼ 211) occurred in 238 fewer/1,000 patients (95% CI: 325
days shortere4.88 days longer). From the above, the predicted
fewere114 fewer patients), and length of mechanical
desirable effect was judged as “small.”
ventilation (6 RCTs [320e322,324e326]; N ¼ 652) was short-
Meanwhile, as a result of non-sedative/light-sedative
ened by an average of 2.39 days (95% CI: 3.73 days short-
management, the fraction of those undergoing tracheos-
ere1.06 days shorter). Additionally, the number of
tomy (3 RCTs [327e329]: N ¼ 1,416) decreased by 4/1,000 pa-
individuals experiencing adverse events that were thought
tients (24 fewere25 more patients), and accidental extubation
to be undesirable effects (2 RCTs [322,323]; N ¼ 612)
(3 RCTs [327e329]; N ¼ 1,416) increased by 33/1,000 patients (2
decreased by 20/1,000 patients (95% CI: 42 fewere23 more
moree76 more patients). From the above, the predicted un-
patients). From the above, the predicted desirable effect was
desirable effect was judged as “small.”
judged as “moderate.”
Meanwhile, harms involved in conducting early rehabili- Certainty of evidence:
tation included one case each of unplanned tracheal tube Because the direction of desirable and undesirable effects
extubation, accidental removal of the arterial catheter, and is not consistent, the certainty of evidence was judged as “very
allergy due to electrical stimulation pad reported in RCTs in low,” which was the lowest of all the outcomes.
the intervention group. Therefore, the undesirable effect of
Values, balance of effects, acceptability, and feasibility
early rehabilitation was judged as “trivial.”
determination:
Certainty of evidence: When considering in terms of point estimates, the direc-
The certainty of evidence was “low” as for all outcomes. tion of predicted desirable effect of this intervention was
almost consistent, and the direction can be changed when the
Values, balance of effects, acceptability, and feasibility
upper and lower limits of the confidence interval were
determination:
considered. From the above, the balance of benefits and harms
Feasibility is thought to be high in facilities that are
was judged as “do not know.” However, the number of pa-
familiar with the implementation of early rehabilitation; fa-
tients who survived and got discharged increased by up to 172/
cilities that have a physiotherapist, occupational therapist,
1,000 patients, and costs may also be reduced. Therefore, it
and speech therapist in charge of intensive care; and envi-
was judged that the effects of intervention likely outweighed
ronments with sufficient manpower.
the harms.
CQ44: Should non-sedative or light-sedative management
CQ45: Should restrictive fluid management strategies be
be conducted for adult patients with ARDS?
implemented for adult patients with ARDS?
Background
Background
Sufficient sedation may sometimes be necessary for the
Pulmonary edemas caused by vascular endothelial damage
management of mechanical ventilation, but high-dose seda-
and vascular hyperpermeability are problems in ARDS, but the
tives may increase the duration of mechanical ventilation and
direct cause of death in ARDS is not hypoxemia but non-lung
the risk of delirium. Minimal to no sedation is expected to
organ failure [330]. In other words, there is room for debate on
have a beneficial effect or avoid complications.
whether restrictive fluid management, which can lead to
Recommendation organ perfusion disorders, should be used in fluid manage-
We suggest conducting no sedation or minimal sedation ment in ARDS with the aim of improving lung oxygenation.
management for adult patients with ARDS (weak recom-
Recommendation
mendation/very low certainty of evidence: GRADE 2D).
We suggest performing the restrictive fluid management
Supplementary item strategies for adult patients with ARDS (weak recommenda-
In contrast to deep sedation, where subjects do not tion/moderate certainty of evidence: GRADE 2B).
respond to stimuli, light sedation refers to sedation where
Supplementary item
subjects could be awakened by physical or verbal stimuli. This
There are several methods of restrictive fluid management
recommendation is based on the results of clinical studies
such as reduced infusion and use of diuretics, and an
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 471

appropriate decision should be made according to the pa- [340,342,343]: N ¼ 113) increased by 11.84 (95% CI: 31.2 low-
tient's condition. ere54.88 higher). From this, the predicted desirable effect was
judged to be “moderate.”
Rationale
Meanwhile, as a result of using u3 fatty acid preparations,
Summary of evidence:
gastrointestinal intolerance (1 RCT [340]: N ¼ 272) occurred in
As a result of restrictive fluid management, mortality (6
77 more/1,000 patients (95% CI: 21 fewere228 more patients).
RCTs [331e336]: N ¼ 1,214) decreased by 28/1,000 patients (95%
From this, the predicted undesirable effect was judged to be
CI: 74 fewere23 more patients), and VFD (1 RCT [336]:
“small.”
N ¼ 1,000) was extended by an average of 2.5 days (95% CI: 1.12
days longere3.88 days longer). The number of days where Certainty of evidence:
hemodynamic events assumed to be harmful did not occur (1 Because the direction of desirable and undesirable effects
RCT [336]: N ¼ 1,000) was shortened by an average of 0.3 days is not consistent, the certainty of evidence was judged as “very
(95% CI: 0.57 days shortere0.03 days shorter) as a result of low,” which was the lowest of all the outcomes.
restrictive infusion management, so the predicted desirable
Values, balance of effects, acceptability, and feasibility
effect was judged to be “small.”
determination:
Meanwhile, as a result of restrictive fluid management, the
Regarding values, the predicted desirable effect of this
proportion of late-onset cognitive dysfunction, which is an
intervention is likely significant compared with the undesir-
important outcome, occurred in 117 more/1,000 patients (1
able effect when considering not only the point estimates but
RCT [337]; certainty of evidence, “low”). Therefore, the pre-
also the upper and lower limits of the confidence interval.
dicted undesirable effect was judged to be “trivial.”
Regarding acceptability, benefits from the intervention
Certainty of evidence: outweigh the harms, and there is the possibility of cost
Because the direction of desirable and undesirable effects reduction; however, the time and effort needed for interven-
is the same, the certainty of evidence was judged to be tion would increase, so it cannot be unequivocally stated one
“moderate,” which was the highest of all the outcomes. way or another. Regarding feasibility, enteral nutrition itself is
a common treatment, and common enteral preparations are
Values, balance of effects, acceptability, and feasibility
easily available, but there are limits to hospitals that adopt u3
determination:
fatty acid preparations, so it was judged that feasibility cannot
The predicted desirable effect of this intervention was
be unequivocally stated one way or another.
judged to outweigh the harms. Since the possibility that the
judgment may differ depending on the restrictive fluid man-
F. Children
agement method, it was judged that this would likely be
accepted.
As in adults, the mechanism of ARDS onset in children is
CQ46: Should enteral nutrition with high u3 fatty acid also pulmonary edema due to hyperpermeability of pulmo-
content be given to patients with ARDS? nary capillaries. However, its causes, background diseases,
and respiratory characteristics (physiological, anatomical,
Background
immunological) are different from those of adults [344,345].
Experiments with sepsis and septic acute lung injury
Therefore, conceptualizing pediatric ARDS (PARDS) separately
model animals showed that the administration of fats with
from adult ARDS is justified. A total of 15 CQs were selected in
high u3 fatty acid content could suppress pulmonary vascular
this area, leading to 3 weak recommendations for and 6 weak
permeability, pulmonary edema, and pulmonary hyperten-
recommendations against interventions, and 6 in our practice
sion [338]. Clarifying the effectiveness of giving enteral
statements (Table 3). Particular attention must be paid to the
nutrition with high u3 fatty acid content such as fish oil is an
following 2 points.
important clinical issue.

Recommendation 1) Definition
We suggest giving enteral nutrition with high u3 fatty acid
content to patients with ARDS (weak recommendation/very The Pediatric Acute Lung Injury Consensus Conference
low certainty of evidence: GRADE 2D). (PALICC) definition were proposed in 2015 as a definition of
PARDS [346]. The frequency of PARDS according to the PALICC
Supplementary item
definition was reported as 6.1% of mechanically ventilated
Enteral nutritional supplements with high u3 fatty acid
patients in the pediatric intensive care unit (PICU), and the
content have been withdrawn from the market in Japan, and
mortality rate was reported as 17.1% [347]. Important differ-
providing enteral nutrition with high u3 fatty acid content
ences between the PALICC definition and the Berlin definition
requires the addition of u3 fatty acid preparations to normal
[348] include the following: SpO2 could be used in diagnosis;
enteral nutrition.
oxygenation index, in preference to the P/F ratio, is the pri-
Rationale mary metric of lung disease severity to define PARDS for
Summary of evidence: patients treated with invasive mechanical ventilation; infil-
As a result of using u3 fatty acid preparations, mortality (7 tration shadows on chest imaging do not have to be bilateral;
RCTs [339e341]: N ¼ 672) decreased by 42/1,000 patients (95% patient groups who may develop ARDS are defined as at risk,
CI: 117 fewere77 more patients), and the P/F ratio (3 RCTs etc. There may be insufficient evidence to conclude whether
472 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

Table 3 e Comparison of pediatric recommendation to adult one.


CQ number Question Direction of recommendation
PCQ adult pediatric adult
1 14 Should non-invasive respiratory support (NPPV/HFNC) be used for 1 þ1
pediatric patients with ARDS?
2 19 Should tidal volume be restricted in pediatric patients with ARDS? 0 þ2
3 20 Should high PEEP be used in pediatric patients with moderate to severe 0 þ1
ARDS?
4 21 Should plateau pressure be restricted in pediatric patients with ARDS? 0 þ1
5 27 Should a protocol be used when liberating pediatric patients with acute þ1 þ1
respiratory failure from mechanical ventilator?
6 28 Should HFOV be used for pediatric patients with moderate to severe ARDS? 1 1
7 23 Should APRV be used for pediatric patients with ARDS? 1 0
8 30 How should target SpO2 values be set in pediatric patients with ARDS? 0 1
(Adult: Is low SpO2 (PaO2) a target for management in adult patients with
ARDS?
9 31 Should muscle relaxants be used at an early stage in pediatric patients with 0 þ1
moderate or severe ARDS?
10 35 Should pediatric patients with moderate to severe ARDS be placed in the þ1 þ1
prone position?
11 40 Should nitric oxide inhalation therapy be used in pediatric patients with 1 1
ARDS?
12 N/A Should surfactant be used in pediatric patients with ARDS? 1 N/A
13 42 Should steroids be used in pediatric patients with ARDS? 0 high dose: 1
low dose: þ2
14 N/A Should a protocol be used for the sedation of pediatric respiratory failure þ1 N/A
patients?
15 44 Should daily sedation interruption (DSI) be implemented for pediatric 1 þ1
respiratory failure patients?
(Adult: Should non-sedative or light-sedative management be conducted
for adult patients with ARDS?)

þ2, strong recommendation for using an intervention; þ1, weak recommendation for using an intervention.
0, no recommendation; 1, weak recommendation against using an intervention.
N/A, not applicable.
CQ, clinical question; PCQ, pediatric clinical question.

the PALICC or the Berlin definition is more useful for the children with lung injuries associated with congenital lung
decision-making of therapeutic interventions in PARDS. PaO2 disease and perinatal abnormalities were excluded.
measurements are not easy in children, so SpO2 could be used
instead. It is known that some PARDS diagnosed by the PAL- b) Pathology
ICC definition could be overlooked by the Berlin definition,
suggesting the superiority of the PALICC definition [346,347]. Although this is an ARDS guideline, the scope of patients
Meanwhile, PALICC definition does not have a long history of (¼P) was expanded according to the contents of CQs. We
use, and many studies have been using the Berlin definition. decided to extract as many articles as possible that contained
In the extraction of articles for the systematic review in this information on PARDS at the literature search stage. In defi-
guideline, we included PARDS diagnosed by any one of the nitions other than PALICC, ARDS cannot be diagnosed if
PALICC definition, Berlin definition, and American-European invasive positive pressure ventilation is not conducted;
Consensus Conference Criteria. therefore, for pediatric CQ (PCQ) 1, all patients with acute
respiratory failure were targeted, but articles only including
2) Patients bronchiolitis were excluded. For PCQ5, PCQ14, and PCQ15, as
a) Age there were few differences between ARDS and non-ARDS
patients, all mechanically ventilated children were included.
There is no definite threshold for the age to distinguish For the other PCQs, articles in which at least half of the causes
children and adults. PALICC also does not define an upper or of acute respiratory failure were ARDS, were included.
lower age limit. Meanwhile, respiratory disorders in newborns
PCQ1: Should non-invasive respiratory support (NPPV/
due to perinatal abnormalities such as respiratory distress
HFNC) be used for pediatric patients with ARDS?
syndrome, meconium aspiration syndrome, respiratory dis-
orders secondary to congenital malformations (e.g., congenital Background
diaphragmatic hernia) are excluded. Like in the PALICC defi- Non-invasive respiratory support such as NPPV or HFNC
nition, the upper and lower age limits for children were not set for patients with ARDS is expected to avoid complications due
in this guideline; when a study included children based on the to tracheal intubation. Meanwhile, there are concerns that the
definition for age in each study, it was included, whereas application of non-invasive respiratory support could delay
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 473

tracheal intubation and, if applied to patients with severe several studies that mechanical ventilation itself could be the
ARDS, could increase mortality rate. Furthermore, there are cause of lung injury [174,176]. Under such circumstances, tidal
tolerability issues in children, so non-invasive respiratory volume restriction could be a part of lung protective strategies
support is not an established treatment in the respiratory for reducing adverse events due to mechanical ventilation.
management of patients with ARDS, and clarifying its effec- Whether to restrict tidal volume during mechanical ventila-
tiveness is a key clinical issue. tion for pediatric patients with ARDS is a key clinical issue.
Note: The target population in the SR of this CQ was not
Recommendation
limited to just patients with ARDS and included all pediatric
We cannot provide a recommendation regarding tidal
cases with acute respiratory failure, but populations that
volume restrictions for pediatric patients with ARDS. It is
targeted only acute bronchiolitis were excluded.
generally accepted not to exceed tidal volume in accordance
Recommendation with the treatment strategy of adult patients with ARDS (in
We suggest against using non-invasive respiratory support our practice statement).
(NPPV/HFNC) in pediatric patients with ARDS.
Supplementary item
(weak recommendation/very low certainty of evidence:
Currently, it is unclear whether to use standard weight or
GRADE 2D).
actual weight for the tidal volume per body weight. There is a
Supplementary item reference physiological tidal volume value of <8 mL/kg [354].
This recommendation does not reject the use of non- Rationale
invasive respiratory support in the early stage of pediatric Summary of evidence:
acute respiratory failure in cases other than ARDS. No applicable studies
Rationale Certainty of evidence:
Summary of evidence: There are no applicable studies, so a description of the
As for beneficial outcomes, the tracheal intubation rate (4 certainty of evidence cannot be provided.
RCTs [349e352]; N ¼ 276) was lower by 46/1,000 patients (95% CI:
150 fewer to 227 more patients). The length of hospital stay (2 Balance of effects, acceptability, and feasibility determination:
RCTs [351,352]; N ¼ 92) was shortened by an average of 0.25 Strain is considered to be an important factor in VALI [355].
days (95% CI: 3.86 days shorter to 3.37 days longer). The pre- Restricting tidal volume so as not to overextend the alveoli is
dicted desirable effect was judged as “trivial.” As for harmful thought to reduce strain and alleviate lung injury [355]. Mean-
outcomes due to intervention, the mortality rate (4 RCTs while, restricting tidal volume is associated with harmful ef-
[349e351,353]; N ¼ 870) increased by 65/1,000 patients (95% CI: fects such as hypercapnia. However, there are no studies that
61 fewer to 545 more patients), and ventilator-free days (1 RCT directly evaluated these aspects, and it is not possible to
[351]; N ¼ 42) was shortened by an average of 6 days (95% CI: determine the balance of effects. Restricting tidal volume is
13.37 days shorter to 1.37 days longer). Pressure ulcers occurred possible by changing the settings of the ventilator, and it is
in 11.6% of the intervention group and was not observed in the thought to be sufficiently acceptable and feasible.
non-intervention group. The predicted undesirable effect was PCQ3: Should high PEEP be used in pediatric patients with
judged to be “small.” Considering the importance of clinical moderate to severe ARDS?
intubation rate and mortality rate, the balance of effects was
judged as “probably favors the comparison.” Background
Mechanical ventilation is often needed in patients with
Certainty of evidence: ARDS, but ventilator use can induce VALI [174]. Therefore, the
The direction of desirable and undesirable effects were mechanical ventilation of these patients should be provided
inconsistent, so the certainty of evidence was judged to be based on lung protective strategies. Application of high-level
“very low.” PEEP is a strategy that aims to reduce VALI [356]. Meanwhile,
Balance of effects, acceptability, and feasibility determination: high PEEP is thought to have adverse effects on hemody-
When focusing on the possibility that mortality rate in- namics. Whether to use high PEEP for mechanical ventilation
creases, undesirable effects outweigh desirable effects. There- in pediatric patients with moderate to severe ARDS is a key
fore, the balance of effects was set as “probably favors the clinical issue.
comparison.” Acceptability was set as “varies” due to the pos- Recommendation
sibility that interface wearing or positive pressure may not be We could not provide a recommendation on whether to
accepted in children. Feasibility in pediatric intensive care units use high PEEP in pediatric patients with moderate to severe
is high, but specialized knowledge is also necessary, and this ARDS, but high PEEP is commonly used based on clinician's
was set as “varies.” experience and findings from adult patients with ARDS (in our
PCQ2: Should tidal volume be restricted in pediatric pa- practice statement).
tients with ARDS? Supplementary item
Background Care should be taken to avoid excessive plateau pressure
Mechanical ventilation could be used in patients with se- when using high PEEP (especially >10 cmH2O). Additionally,
vere ARDS to buy time as a rescue therapy for ARDS, but the careful monitoring of adverse events such as hemodynamic
mortality rate remains high. It has also been suggested in instability is needed. The effects of PEEP differ according to
474 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

lung pathophysiology, and high PEEP does not necessarily restrictions could be implemented by changing only the
improve oxygenation or lung injury. ventilator settings, which is considered to be sufficiently
acceptable and feasible.
Rationale
Summary of evidence: PCQ5: Should a protocol be used when liberating pediatric
No applicable studies patients with acute respiratory failure from mechanical
ventilator?
Certainty of evidence:
There are no applicable studies, so the certainty of evi- Background
dence could not be evaluated. Prolonged mechanical ventilation increases the incidence
of adverse events; meanwhile, premature extubation in-
Balance of effects, acceptability, and feasibility determination:
creases the reintubation rate and mortality [359]. Whether to
There are no applicable studies, and the balance of effects
use a protocol when liberating pediatric patients with acute
could not be determined. PEEP can be controlled by changing
respiratory failure from mechanical ventilator is a key clinical
the ventilator settings, which is considered acceptable or
issue.
feasible.
Recommendation
PCQ4: Should plateau pressure be restricted in pediatric
We suggest to use a protocol when liberating pediatric
patients with ARDS?
patients with acute respiratory failure who have been me-
Background chanically ventilated for more than 24 h from ventilator.
There are concerns that mechanical ventilation in adult (weak recommendation/very low certainty of evidence:
patients with ARDS could lead not only to extended ventilator GRADE 2D).
use time due to VALI but also increased mortality rate.
Supplementary item
Increased airway pressure is thought to be one of the factors
None
that cause VALI, and it is expected that VALI is suppressed by
restricting plateau pressure [357]. Meanwhile, restricting Rationale
plateau pressure may induce adverse events such as hyper- Summary of evidence:
capnia. Whether to restrict plateau pressure in the mechani- Results of the 2 adopted RCTs [360,361] (N ¼ 479) showed
cal ventilation of pediatric patients with ARDS is a key clinical that, for beneficial outcomes, 30-day mortality (1 RCT [360];
issue. N ¼ 260) decreased by 1/1,000 patients (95% CI: 14 fewer to 88
more patients) and duration of mechanical ventilation (1 RCT
Recommendation
[361]; N ¼ 219) was shortened by an average of 2.9 days (95% CI:
We cannot provide a recommendation regarding plateau
5.91 days shorter to 0.11 days longer). As a result, the desirable
pressure restrictions for pediatric patients with ARDS, but
effect was judged as “small.”
respiratory management with restrictions on plateau pres-
There were no reports on critical harmful outcomes; for
sure is implemented in accordance with the treatment strat-
other harmful outcomes, reintubation (2 RCTs [360,361];
egy of adult patients with ARDS (in our practice statement).
N ¼ 479) decreased by 2/1,000 patients (95% CI: 48 fewer to 103
Supplementary item more patients) and unplanned extubation (1 RCT [360];
The standard has been 28 cmH2O (more pressure may be N ¼ 260) occurred in 41 fewer/1,000 patients (95% CI: 57 fewer
required in conditions with decreased chest wall compliance) to 19 more patients). As a result, the undesirable effect was
[358]. It should be noted that restricting plateau pressure alone judged as “trivial.”
is not sufficient in controlling overdistention if there is strong From the above, the balance of effects was judged as
spontaneous breathing. It should also be noted that the “probably favors the intervention.”
plateau pressure and maximum airway pressure are different
Certainty of evidence:
even in the case of pressure-control ventilation.
For the desirable effects, 30-day mortality and duration of
Rationale mechanical ventilation were both set with a certainty of evi-
Summary of evidence: dence of “very low.” For the undesirable effects, there were no
No applicable studies reports of critical outcomes. As the direction of desirable and
undesirable effects was consistent, the certainty of evidence
Certainty of evidence:
of all outcomes was judged as “very low.”
There are no applicable studies, so the certainty of evi-
dence could not be evaluated. Balance of effects, acceptability, and feasibility determination:
The balance of effects was judged as “probably favors
Balance of effects, acceptability, and feasibility determination:
the intervention,” and it was judged that patients and
Plateau pressure restrictions in patients with ARDS could
families would accept this method. It is thought that its
suppress VALI, but there are concerns that changes in blood
implementation including evaluations of sedation level may
acid-base balance associated with hypercapnia could result in
not be easy in ICUs that are not specialized for children.
adverse effects on hemodynamics and tissue metabolism.
Therefore, the feasibility of this intervention varies from fa-
However, there are no applicable studies, and it is not possible
cility to facility.
to determine the balance of effects. Plateau pressure
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 475

PCQ6: Should HFOV be used for pediatric patients with Recommendation


moderate to severe ARDS? We suggest against implementing APRV as a ventilation
mode in pediatric patients with ARDS (weak recommenda-
Background
tion/very low certainty of evidence: GRADE 2D).
HFOV is a mechanical ventilation mode where both lung
recruitment and restricted tidal volume can be achieved. Supplementary item
Though it cannot be said to be a common mechanical venti- This recommendation does not reject the implementation
lation mode for children. Whether to use HFOV for pediatric of APRV in facilities familiar with its use.
patients with moderate to severe ARDS is a key clinical issue.
Rationale
Recommendation Summary of evidence:
We suggest against implementing HFOV for pediatric pa- SR results showed that there was 1 adopted RCT [367]
tients with ARDS. (N ¼ 52) in line with PICO. For beneficial outcomes, devel-
(weak recommendation/very low certainty of evidence: opmental prognosis (PCPC after 180 days) showed 0 change
GRADE 2D). (95% CI: 0.4 worsened to 0.4 improved), ventilator-free days
shortened by an average of 4.5 days (95% CI: 10.35 days
Supplementary item
shorter to 1.35 days longer), and length of hospital stay was
This recommendation does not reject the implementation
shortened by an average of 5 days (95% CI: 10.73 days
of this procedure in facilities that already have an HFOV-
shorter to 0.73 days longer). Twenty-eight-day mortality rate
specialized ventilator and are familiar with its use.
was initially assumed as a desirable effect, but as described
Rationale later, mortality rate increased, so this was considered as an
Summary of evidence: undesirable effect. Therefore, the desirable effect due to
Results of the 4 adopted RCTs [362e365](N ¼ 292) showed intervention was judged as “trivial.” For harmful outcomes,
that, for beneficial outcomes, mortality (4 RCTs [362e365]; 28-day mortality rate increased by 269/1,000 patients (95%
N ¼ 292) decreased by 12/1,000 patients (95% CI: 108 fewer to CI: 8 fewer to 845 more patients) for the intervention
116 more patients), and ventilator-free days (1 RCT [362]; compared to the control, and exacerbated hemodynamics
N ¼ 102) was shortened by an average of 0 days (95% CI: 0.82 was observed in 77 fewer/1,000 patients (95% CI: 111 fewer
days shorter to 0.82 days longer). From the above, the desir- to 231 more patients). Therefore, the undesirable effect was
able effect was judged as “trivial.” For harmful outcomes, judged as “moderate.” From the above, the balance of ben-
hemodynamic instability (2 RCTs [363,365]; N ¼ 76) occurred in efits and harms was judged as “probably favors the
53 more/1,000 patients (95% CI: 18 fewer to 689 more patients), comparison.”
and the undesirable effect was judged as “small.” Therefore,
Certainty of evidence:
the balance of effects was judged as “probably favors the
The direction of desirable and undesirable effects was not
comparison.”
the same, and the certainty of evidence for the overall
Certainty of evidence: outcome adopted the certainty of the outcome with the lowest
The direction of desirable and undesirable effects was not certainty and set as “very low.”
the same, and the certainty of evidence of the overall outcome
Balance of effects, acceptability, and feasibility determination:
was judged as “very low.”
The balance of effects was judged as “probably favors the
Balance of effects, acceptability, and feasibility determination: comparison.” There was the possibility that this had a nega-
The balance of effects was judged as “probably favors the tive effect on mortality rate, and intervention may not be
comparison,” and it was judged that patients and families acceptable. APRV is feasible if a specific ventilator is available,
would accept this method. HFOV cannot be implemented but if not, then feasibility is low, and the overall feasibility is
without a specialized ventilator. Moreover, in facilities that do set as “varies”.
not use HFOV on a regular basis, purchasing the dedicated
PCQ8: How should target SpO2 values be set in pediatric
ventilators and providing training on how to use them will
patients with ARDS?
increase on-site burden. Therefore, feasibility was judged as
likely low. Background
Management with high oxygen concentration has been
PCQ7: Should APRV be used for pediatric patients with
reported to induce adverse events that promote pulmonary
ARDS?
fibrogenesis [368]. Meanwhile, it is unclear whether low SpO2
Background management improves survival rate or worsens prognosis
Alongside treatment of underlying diseases, mechanical through adverse events. Therefore, the target SpO2 value that
ventilation in patients with ARDS is important [366]. The should be set in pediatric patients with ARDS is a key clinical
optimal ventilation mode in mechanical ventilation is not issue.
clear. APRV is used in some cases of pediatric ARDS to main-
Recommendation
tain a high airway pressure, but its effectiveness in children is
We cannot provide a specific target SpO2 value for pediatric
unclear. Therefore, whether to use APRV in pediatric patients
patients with ARDS, but target SpO2 value are set so as to avoid
with ARDS is a key clinical issue.
476 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

excessively high or low oxygen levels, which could cause practice, and it is thought that there are no problems with
organ damage (in our practice statement). acceptability or feasibility.

Supplementary item PCQ10: Should pediatric patients with moderate to severe


None ARDS be placed in the prone position?

Rationale Background
Summary of evidence: The prone position may be effective as treatment for mod-
No applicable studies erate to severe ARDS [370], as it improves oxygenation and
prevents VALI, but there is room for debate on its clinical ef-
Certainty of evidence:
fects based on previous RCTs and meta-analyses [264,371,372].
There are no applicable studies, so a description of the
The prone position can be implemented without special
certainty of evidence cannot be provided.
equipment, but complications can occur with repositioning of
Balance of effects, acceptability, and feasibility determination: the patients and prolonged management. Therefore, whether
There are no applicable studies, and it is not possible to judge to place pediatric patients with moderate to severe ARDS in the
the balance of effects. Management with a specific SpO2 target is prone position is a key clinical issue.
possible with only changes to ventilator settings and is already
Recommendation
implemented in daily clinical practice, so it is thought that its
We suggest placing pediatric patients with moderate to
acceptability and feasibility are sufficiently high.
severe ARDS in the prone position (weak recommendation/
PCQ9: Should muscle relaxants be used at an early stage in very low certainty of evidence: GRADE 2D).
pediatric patients with moderate or severe ARDS?
Supplementary item
Background Placing children in the prone position requires the facility
The use of muscle relaxants for patients with ARDS has been to be familiar with the procedure, and applications need to be
reported to avoid excessive stress to alveoli, reduce barotrauma, considered for each facility, including the degree of sedation.
and improve oxygenation, suggesting an improved prognosis
Rationale
[369]. However, it has also often been reported that complica-
Summary of evidence:
tions could occur due to muscle relaxants, which reduce or
There was 1 adopted RCT [373] (N ¼ 101; patients with P/F
eliminate spontaneous breathing [369]. Whether to use muscle
ratio <300 targeted). For beneficial outcomes, 28-day mortality
relaxants at an early stage for pediatric patients with moderate
decreased by 2/1,000 patients (95% CI: 59 fewer to 217 more
or severe ARDS is a key clinical issue.
patients), poor developmental prognosis decreased by 103/
Recommendation 1,000 patients (95% CI: 174 fewer to 75 more patients), and
We cannot provide a recommendation on the early use of ventilator-free days was shortened by an average of 0.3 days
muscle relaxants for pediatric patients with moderate or se- (95% CI: 3.63 days shorter to 3.03 days longer). As a result, the
vere ARDS, but muscle relaxants have been used at an early desirable effect due to intervention was judged as “small.” For
stage in accordance with the treatment strategy of adult harmful outcomes, endotracheal tube troubles occurred in 22
patients with moderate or worse ARDS (in our practice fewer/1,000 patients (95% CI: 78 fewer to 175 more patients),
statement). and the predicted undesirable effect was judged as “trivial.”
From the above, for whether benefits due to intervention
Supplementary item
would outweigh harms, it was judged that “probably favors
None
the intervention.”
Rationale
Certainty of evidence:
Summary of evidence:
The direction of outcomes for desirable and undesirable
No applicable studies
effects were not the same, and the certainty of evidence was
Certainty of evidence: judged to be “very low.”
There are no applicable studies, so no description of the
Balance of effects, acceptability, and feasibility determination:
certainty of evidence can be provided.
The balance of effects was judged as “probably favors the
Balance of effects, acceptability, and feasibility determination: intervention.” It is thought that there are not many human
Muscle relaxants in patients with ARDS requiring me- resources needed for repositioning small children to the prone
chanical ventilation are thought to have the advantages of position, but the facility will need to be familiar with the
decreased patient-ventilator asynchrony, decreased oxygen procedure for safe management, so it was judged that
consumption, increased respiratory compliance and func- acceptability and feasibility of this procedure was likely
tional residual capacity, and prevention of local alveolar possible.
overdistention due to strong spontaneous breathing [369].
PCQ11: Should nitric oxide inhalation therapy be used in
Meanwhile, there are concerns of harmful effects such as the
pediatric patients with ARDS?
onset of ICU-AW due to the use of muscle relaxants. How-
ever, there are no studies that directly evaluated these as- Background
pects, so it is not possible to judge the balance of effects. Various pathological conditions such as hypoxic pulmo-
Muscle relaxants themselves are used in daily clinical nary vasospasm and ventilation-perfusion imbalance are
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 477

involved in ARDS [374]. There is a possibility that NO inhala- Supplementary item


tion, which has a pulmonary vessel dilatation effect [375], may None
be effective, but its clinical effects vary according to reports.
Rationale
Therefore, whether to use NO inhalation therapy for pediatric
Summary of evidence:
patients with ARDS is a key clinical issue.
Meta-analysis results of the 9 incorporated RCTs [379e387]
Recommendation showed that, for the beneficial outcomes due to surfactant
We suggest against routinely implementing NO inhalation use, mortality (9 RCTs [379e387]; N ¼ 658) decreased by 64/
therapy for pediatric patients with ARDS (weak recommen- 1,000 patients (95% CI: 130 fewer to 45 more patients), 28-day
dation/very low certainty of evidence: GRADE 2C). ventilator-free days (2 RCTs [386,387]; N ¼ 261) were short-
ened by an average of 0.65 days (95% CI: 5.26 days shorter to
Supplementary item
3.95 days longer), and the length of hospital stay (2 RCTs
This treatment can be acceptable in limited circumstances.
[385,386]; N ¼ 194) was shortened by an average of 1.67 days
Examples include temporary use until the introduction of
(95% CI: 7.82 days shorter to 4.49 days longer). Therefore, the
extracorporeal membrane oxygenation, and situations where
desirable effect was judged as “small.” For harmful outcomes,
there are no other treatments and a high mortality rate is
drug-related adverse events (7 RCTs [379,380,383e387];
predicted. It should be noted that NO inhalation therapy for
N ¼ 589) occurred in 180 more/1,000 patients (95% CI: 81 more
ARDS is not covered by insurance.
to 360 more patients). Many of the drug-related adverse events
Rationale were transient hypoxemia and hemodynamic compromises,
Summary of evidence: and few were serious, so the undesirable effect was judged as
Meta-analysis results of the 3 adopted RCTs [376e378] “small.” Therefore, the balance of effects was judged as “does
(N ¼ 177) showed that, for beneficial outcomes, mortality rate not favor either the intervention or the comparison.”
(3 RCTs [376e378]; N ¼ 177) decreased by 54/1,000 patients
Certainty of evidence:
(95% CI: 237 fewer to 409 more patients), and 28-day
The direction of critical outcomes was not the same, and
ventilator-free days (2 RCTs [376,377]; N ¼ 71) were
the certainty of evidence was set as “very low.”
extended by an average of 4.90 days (95% CI: 0.78 days longer
to 9.03 days longer). However, there is the possibility that Balance of effects, acceptability, and feasibility determination:
mortality increased by 409/1,000 patients, so the desirable The balance of effects was judged as “does not favor either
effect was judged as “small.” For harmful outcomes, serious the intervention or the comparison.” Given that surfactants
side effects (3 RCTs [376e378]; N ¼ 177) decreased by 10/1,000 are expensive drugs, interventions could increase drug-
patients (95% CI: 21 fewer to 99 more patients). However, related adverse events, and resources are needed for imple-
there is the possibility that it could increase 99/1,000 patients, mentation, it was judged that acceptability was “probably no,”
so the undesirable effect was judged as “trivial.” Therefore, and feasibility was “probably no.”
the balance of effects was judged as “probably favors the
PCQ13: Should steroids be used in pediatric patients with
intervention.”
ARDS?
Certainty of evidence:
Background
The direction of critical outcomes was the same, and the
ARDS is a pathological condition caused by inflammation
certainty of evidence was judged as “low.”
that has spread to the lungs due to various causes [374]. Ste-
Balance of effects, acceptability, and feasibility determination: roids have an suppressing effect on inflammation mediators,
The balance of effects was judged as “probably favors the but they can also decrease the immune system. Therefore,
intervention,” and it was judged that there were no problems whether to use steroids is a key clinical issue.
with patient and family acceptability. However, the predicted
net effect was uncertain, and there are many facilities where Recommendation
the feasibility of this treatment would be low, so we decided We cannot provide a recommendation on the administra-
not to propose that this be implemented routinely. tion of steroids for pediatric patients with ARDS. Steroid
administration is examined in consideration of patient back-
PCQ12: Should surfactant be used in pediatric patients ground and pathophysiology (in our practice statement).
with ARDS?
Supplementary item
Background This is a description of the current clinical practice, and
Various factors such as alveolar epithelial injury and this does not reject the use of steroids.
alveolar surfactant dysfunction are involved in the patho-
physiology of ARDS [374]. Surfactant administration can be Rationale
effective, but its clinical effects vary according to reports. Summary of evidence:
Therefore, whether to use surfactant on pediatric patients There was only 1 RCT [388] (N ¼ 35). For beneficial out-
with ARDS is a key clinical issue. comes, the mortality rate decreased by 88/1,000 patients (95%
CI: 110 fewer to 344 more patients), ventilator-free days was
Recommendation extended by an average of 1.32 days (95% CI: 3.32 days shorter
We suggest against using surfactants for pediatric patients to 5.96 days longer), and length of hospital stay was shortened
with ARDS (weak recommendation/very low certainty of evi- by an average of 6.87 days (95% CI: 15.32 days shorter to 1.58
dence: GRADE 2D).
478 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

days longer). The confidence intervals were large, and there developmental prognosis. Therefore, the desirable effect was
was the possibility that mortality rate increased by 344 pa- judged as “small.” Meanwhile, for harmful outcomes, there
tients, so the desirable effect was judged as “trivial.” For were no reports of absolute numbers of adverse events such
harmful outcomes, the infectious disease incidence decreased as unplanned extubation or worsening of respiratory status.
by 219/1,000 patients (95% CI: 296 fewer to 175 more patients), The report (Curley, 2015 [389]) taken up in the meta-analysis
and hyperglycemia increased by 142/1,000 patients (95% CI: indicated that withdrawal syndrome was more by 29/1,000
138 fewer to 684 more patients). The critical harmful out- patients (95% CI: 4 fewer to 60 more patients), and the un-
comes of developmental prognosis and presence of myop- desirable effect was judged as “trivial.” Therefore, the bal-
athy/neuropathy were not evaluated, and the undesirable ance of effects was judged as “probably favors the
effect was judged as “varies.” The balance of effects was intervention.”
judged as “varies.”
Certainty of evidence:
Quality of evidence: The direction of critical outcomes was the same, and the
The sample size of the applicable study was extremely certainty of evidence was judged as “moderate.”
small, and patient backgrounds also varied significantly.
Balance of effects, acceptability, and feasibility determination:
Therefore, it was thought difficult to provide a recommenda-
The balance of effects was judged as “probably favors the
tion from this meta-analysis.
intervention,” and it was judged that there were no major
Balance of effects, acceptability, and feasibility determination: problems with patient and family acceptability. There is a
The balance of effects was judged as “varies.” As steroids possibility that sedation/analgesia scale introduction for
are frequently used in daily clinical practice, it was thought children in ICUs with few pediatric patients would be a
that there were no problems with feasibility. However, the burden, and feasibility was judged as “varies.”
critical undesirable effects of developmental prognosis and
PCQ15: Should daily sedation interruption (DSI) be imple-
presence of myopathy/neuropathy were not evaluated.
mented for pediatric respiratory failure patients?
Therefore, the acceptability of this was thought to vary by
stakeholder. Background
DSI is expected to shorten the length of mechanical
PCQ14: Should a protocol be used for the sedation of pe-
ventilation and reduce the risk of delirium and withdrawal
diatric respiratory failure patients?
symptoms, but this is not an established treatment in pedi-
Background atric patients with ARDS. Therefore, whether to implement
The introduction of sedation protocols is expected to DSI in pediatric patients with ARDS is a key clinical issue.
shorten the length of mechanical ventilation and reduce the As DSI is an intervention for patients ventilated with
risk of onset of delirium and withdrawal symptoms, but its various diseases to be liberated from mechanical ventilation,
usefulness in the mechanical ventilation of pediatric patients target populations were not limited to patients with ARDS, but
with ARDS has not been established. Therefore, whether to included all mechanically ventilated children.
use a protocol for the sedation of pediatric patients with ARDS
Recommendation
is a key clinical issue.
We suggest against implementing DSI for pediatric respi-
As sedation protocols are interventions for patients with
ratory failure patients (weak recommendation/very low cer-
various diseases to be liberated from mechanical ventilation,
tainty of evidence: GRADE 2D).
target populations were not limited to patients with ARDS but
included all mechanically ventilated children. Supplementary item
There is currently insufficient evidence for providing a
Recommendation
recommendation for DSI. It is important to evaluate on a daily
We suggest the use of protocols for the sedation of pedi-
basis whether withdrawal from a ventilator is possible.
atric respiratory failure patients (weak recommendation/
moderate certainty of evidence: GRADE 2B). Rationale
Summary of evidence:
Supplementary item
There were 3 RCTs [390e392] (N ¼ 261) that compared the
The sedation protocol examined was based on an algo-
use of DSI in sedation management. For beneficial outcomes,
rithm in which nurses assess and adjust sedation and anal-
the length of mechanical ventilation was shortened by an
gesia on a scale.
average of 1.48 days (95% CI: 3.48 days shorter to 0.51 days
Rationale longer), and the length of hospital stay was shortened by an
Summary of evidence: average of 3.4 days (95% CI: 8.84 days shorter to 2.04 days
There was 1 RCT [389] (N ¼ 2,449) that compared a seda- longer); there were no reports of developmental prognosis.
tion protocol group with a normal clinical practice group. For From the above, the desirable effect was judged as “small.”
beneficial outcomes, mortality was lower by 13/1,000 patients Meanwhile, for harmful outcomes, mortality increased by 166/
(95% CI: 25 fewer to 4 more patients), length of mechanical 1,000 patients (95% CI: 92 fewer to 1,000 more patients), and
ventilation changed by an average of 0 days (95% CI: 0.46 days unplanned extubation decreased by 26/1,000 patients (95% CI:
shorter to 0.46 days longer), and length of hospital stay was 41 fewer to 39 more patients). From the above, the undesirable
shortened by an average of 2 days (95% CI: 3.04 days shorter effect was judged as “moderate.” As a result, the balance of
to 0.96 days longer). There were no reports of the effects was judged as “probably favors the comparison.” The
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 479

causal relationship between intervention and increased


mortality in the present meta-analysis is unclear. Availability of data and materials

Certainty of evidence: Additional file 1e6 contains Modified Preferred Reporting


The direction of outcomes was not the same, and the cer- Items of Systematic Reviews and Meta-Analyses (PRISMA)
tainty of evidence was set as “very low.” flow-chart, risk of bias table, forest plots, evidence profiles,
Balance of effects, acceptability, and feasibility determination: and evidence to decision table for each CQ according to the
The balance of effects was judged as “probably favors the GRADE system.
comparison.” The personnel costs after DSI are large in ICUs Additional file 7 contains tables disclosing intellectual and
that have few opportunities to hospitalize pediatric patients. financial conflicts of interest for each person who participated
Additionally, the increased discomfort in children due to DSI in the creation of this guideline.
may result in situations where family acceptability is difficult.
Therefore, acceptability and feasibility were set as “cannot be
unequivocally stated.” Funding

The Japanese Society of Intensive Care Medicine (JSICM), the


4. Conclusions Japanese Respiratory Society (JRS), and the Japanese Society of
Respiratory Care Medicine (JSRCM) task force on clinical
Continuing from the 2016 edition, we created a clinical prac- practice guideline for ARDS collaborated to perform system-
tice guideline using the GRADE system. Considering our atic review and to provide practical recommendations for the
experience from the previous edition, we established a crea- health care providers. Thus, funding was shared between
tion governing committee in close collaboration with the these three societies.
three academic societies from the preparation stage. There
were many more CQs, so there were also more SR reviewers,
and we were forced to increase the complexity of our opera- Authors' contributions
tions. Therefore, we established an SR governing committee,
with responsible committee members assigned to each area; This guideline document was prepared by the 9 Guideline
we also formed a support team, which provided operational steering committee members and directors, 1 member of
support for the governing committee. There were some adviser for GRADE system, and 38 members of the guideline
changes in the SR committee members, but committee support members. MS (JSICM) and ST (JRS) are the chairmen of
members who were involved in the creation of the 2016 edi- this work, and both contributed equally to the creation of
tion played a leading role, and we were able to complete the SR these guidelines. SO (JSRCM), HY (JSICM), and YO (JSICM) are
work almost as planned. Some of the recommendation con- the organizers of the whole project and manuscript prepara-
tents, such as those for steroid administration, became more tion. The names of the members are listed in the title page and
in-depth than existing guidelines; it is thought that we were authors' information. Each member's contributions are shown
able to provide a certain degree of originality as a result. in Additional file 7. Systematic review members from the
Meanwhile, many of the newly set CQs did not have RCTs; in committee engaged in the making of systematic review.
addition, despite the fact that this was a clinical practice Guideline panelists held the conference and voted for the
guideline for medical professionals in Japan, it could be seen recommendations of each clinical question. All authors read
that there was almost no evidence from Japan, and this has and approved the final manuscript.
reaffirmed the need to provide evidence from Japan. We
would like to reflect on the points clarified in the present
creation process, and we would like to create a better clinical Conflict of Interest
practice guideline in the next revision.
All committee members and panelists submitted disclosure
forms of financial and academic conflict of interest (COI) prior
Ethics approval and consent to participate to being requested to participate in individual activities. If
panelists have any COI concerning each CQ, other panelists
Not applicable were assigned to replace the vacancy. All COI were collected
according to the guideline written by Japanese Society of
Intensive Care Medicine. Detailed information of COI and the
Consent for publication roles in creating this clinical guideline are summarized in
Additional file 7.
Not applicable
480 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

VCV volume-control ventilation


Acknowledgements VFD ventilator-free days
VV-ECMO veno-venous extracorporeal membrane
We would like to thank Editage (www.editage.jp) for their oxygenation
English language editing service. We also gratefully
acknowledge Ms. Fukue Yamamoto in the JSICM Secretariat
and Ms. Kumiko Imazu in the JRS Secretariat for their kind Appendix A. Authors' information
supports.
Detailed information of members of ARDS clinical practice
guideline committee other than appeared at the top of this
List of abbreviations
article.
Systematic review committee members
A/C assisted controlled ventilation
Takuro Nakashima (Department of Intensive Care Medi-
AGREEII Appraisal of Guidelines for Research and Evaluation
cine, Kumamoto University Hospital), Aiko Masunaga
II
(Department of Respiratory Medicine, Kumamoto University
APRV airway pressure release ventilation
Hospital), Aiko Tanaka (Department of Anesthesiology and
ARDS acute respiratory distress syndrome
Intensive Care Medicine, Graduate School of Medicine, Osaka
BAL bronchoalveolar lavage
University), Akihiko Inoue (Department of Emergency and
BNP brain natriuretic peptide
Critical Care Medicine, Hyogo Emergency Medical Center),
CI confidence interval
Akiko Higashi (Department of Emergency and Critical Care
CMV cytomegalovirus
Medicine/Chiba University Graduate School of Medicine),
COPD chronic obstructive pulmonary disease
Atsushi Tanikawa (Department of Emergency and Critical
CQ clinical question
Care, Tohoku University Hospital), Atsushi Ujiro (Department
CRP C-reactive protein
of critical care, Osaka city general hospital), Chihiro Takayama
DI disagreement index
(Takatsuki General Hospital), Daisuke Kasugai (Department of
DSI daily sedation interruption
Emergency and Critical Care Medicine, Nagoya University
EIT electrical impedance tomography
Graduate School of Medicine), Daisuke Kawakami (Depart-
GRADE Grading of Recommendations Assessment,
ment of Anesthesia and Critical Care, Kobe City Medical
Development and Evaluation
Center General Hospital), Daisuke Ueno (Department of Acute
GPS good practice statement
Medicine, Kawasaki Medical School), Daizoh Satoh (Depart-
HFNC high-flow nasal cannula oxygen therapy
ment of Anesthesiology and Pain Medicine, Juntendo Univer-
HFOV high-frequency oscillatory ventilation
sity School of Medicine), Shinichi Kai (Department of
HRCT high-resolution CT
Anesthesia, Kyoto University Hospital), Kohei Ota (Depart-
ICU intensive care unit
ment of Emergency and Critical Care Medicine, Graduate
ICU-AW ICU-acquired weakness
School of Biomedical and Health Sciences, Hiroshima Uni-
IGRA interferon g release assay
versity), Yoshihiro Hagiwara (Department of Emergency and
IPA Invasive pulmonary aspergillosis
Critical Care Medicine, Saiseikai Utsunomiya Hospital), Jun
KQ key question
Hamaguchi (Department of Emergency and Critical Care
MV mechanical ventilation
Medicine, Tokyo Metropolitan Tama Medical Center), Ryo Fujii
NO nitric oxide
(Department of Emergency Medicine and Critical Care Medi-
NPPV non-invasive positive pressure ventilation
cine, Tochigi prefectural emergency and critical care center,
ODI optical density index
Imperial Foundation Saiseikai Utsunomiya Hospital), Takashi
PALICC Pediatric Acute Lung Injury Consensus Conference
Hongo (Department of Emergency Medicine Okayama Saisei-
Criteria
kai General Hospital), Yuki Kishihara (Department of emer-
PARDS pediatric ARDS
gency medicine, Japanese Red Cross Musashino Hospital),
PCP Pneumocystis pneumonia
Naohisa Masunaga (Department of Healthcare Epidemiology,
PCT procalcitonin
School of Public Health in the Graduate School of Medicine,
PCV pressure-control ventilation
Kyoto University), Ryohei Yamamoto (Department of Health-
PCQ pediatric clinical question
care Epidemiology, School of Public Health in the Graduate
PEEP positive end-expiratory pressure
School of Medicine, Kyoto University, Kyoto, Japan), Satoru
P/F PaO2/FIO2
Robert Okazaki (Department of Intensive Care Medicine,
PICS post-intensive care syndrome
Kameda Medical Center), Ryo Uchimido (Department of
PICU pediatric intensive care unit
Intensive Care Medicine, Tokyo Medical and Dental University
QOL quality of line
Hospital), Tetsuro Terayama (Department of Psychiatry, Na-
RCT randomized controlled trial
tional Defense Medical College), Satoshi Hokari (Department
SIMV synchronized intermittent mandatory ventilation
of Respiratory Medicine and Infectious Diseases, Niigata Uni-
SR systematic review
versity Graduate School of Medical and Dental Sciences),
SUCRA surface under the cumulative ranking curve
Hitoshi Sakamoto (Department of Thoracic Surgery, Shimane
VALI ventilator-associated lung injury
Prefectural Central Hospital), Dongli (Department of Anes-
VAP ventilator-associated pneumonia
thesia, Kyoto University Hospital; Department of Healthcare
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 481

Epidemiology, Kyoto University Graduate School of Medicine Hospital), Kengo Asano (Intensive Care Unit, Department of
and Public Health), Emiko Nakataki (Department of Critical Anesthesiology, Jikei University School of Medicine), Kenichi
care medicine, Tokushima Central Prefectural Hospital), Erina Hondo (Department of Trauma and Acute Critical Care Center,
Tabata (Department of Respiratory Medicine, Kanagawa Car- Tokyo Medical and Dental University Hospital), Kenji Ishii
diovascular and Respiratory Medicine), Seisuke Okazawa (Intesive Care unit, St Lukes international hospital), Kensuke
(First Department of Internal Medicine, Toyama University Fujita (Department of Emergency Medicine and Critical Care
Hospital, Toyama), Futoshi Kotajima (Department of Intensive Medicine, Tochigi prefectural emergency and critical care
Care Medicine, Saitama Medical School International Medical center, Imperial Foundation Saiseikai Utsunomiya Hospital),
Center), Go Ishimaru (Soka Municipal Hospital), Haruhiko Kenta Ogawa (Intensive Care Unit, Department of Anesthesi-
Hoshino (Department of nursing, International University of ology, Jikei University School of Medicine), Kentaro Ito (Res-
Health and Welfare), Hideki Yoshida (Department of Emer- piratory Center, Matsusaka Municipal Hospital), Kentaro
gency and Critical Care Medicine, St. Marianna University Tokunaga (Department of Intensive Care Medicine, Kuma-
School of Medicine), Hidetaka Iwai (Department of Anes- moto University Hospital), Kenzo Ishii (Department of Anes-
thesia, Social medical corporation Hakuaikai Kaisei Hospital), thesiology, Intensive Care Unit, Fukuyama City Hospital),
Hiroaki Nakagawa (Division of Respiratory Medicine, Depart- Kohei Kusumoto (Department of pediatric critical care medi-
ment of Internal Medicine, Shiga University of Medical Sci- cine, Hyogo prefectural Amagasaki General Medical Center),
ence), Hiroko Sugimura (Department of Critical Care, Chiba Kohei Takimoto (Department of Intensive Care Medicine,
Children's Hospital), Hiromichi Narumiya (Division of Emer- Kameda Medical Center), Kohei Yamada (Department of
gency and Critical Care, Japanese Red Cross Kyoto Daini Traumatology and Critical Care Medicine, National Defense
Hospital), Hiromu Okano (Department of Critical care and Medical College), Koichi Naito (Department of Cardiac Reha-
Emergency Medicine, National Hospital Organization Yoko- bilitation, Iwama Cardiovascular and Dental Clinic for Pre-
hama Medical Center), Hiroshi Nakamura (Department of vention and Care), Koichi Yamashita (Division of Critical Care
General Medicine, Kure Medical Center and Chugoku Cancer Center, Kochi Red Cross Hospital), Koichi Yoshinaga (Depart-
Center), Hiroshi Sugimoto (Department of Respiratory Medi- ment of Anesthesiology and Critical Care, Jichi Medical Uni-
cine, Konan Medical Center), Hiroyuki Hashimoto (Depart- versity, Saitama Medical Center), Kota Yamauchi (Department
ment of Pharmacoepidemiology, Graduate School of Medicine of Rehabilitation, Steel Memorial Yawata Hospital), Maki
and Public Health, Kyoto University), Hiroyuki Ito (Department Murata (Department of Healthcare Epidemiology, Kyoto Uni-
of Pulmonology, Kameda Medical Center), Hisashi Dote versity of Graduate School of Medicine), Makiko Konda
(Department of Emergency and Critical Care Medicine, Seirei (Department of Anesthesiology, Nara Medical University),
Hamamatsu General Hospital), Hisashi Imahase (Graduate Manabu Hamamoto (Pediatric Emergency and Critical Care
school of medicine, Tokyo University), Hitoshi Sato (Depart- Center, Saitama Children's Medical Center, Saitama, Japan),
ment of critical care, National Cerebral and Cardiovascular Masaharu Aga (Department of Respiratory Medicine, Yoko-
Center), Masahiro Katsurada (Hyogo Prefectural Tamba Med- hama Municipal Citizen's Hospital), Masahiro Kashiura
ical Center), Ichiro Osawa (Department of Critical Care and (Department of Emergency and Critical Care Medicine, Jichi
Anesthesia, National Center for Child Health and Develop- Medical University Saitama Medical Center), Masami Ishi-
ment), Jun Kamei (Emergency and Critical Care Center, Kura- kawa (Department of Anesthesiology and Critical Care Medi-
shiki Central Hospital), Jun Maki (Department of Critical Care cine of Kure Kyousai Hospital), Masayuki Ozaki (Department
Medicine, Kyushu University Hospital), Jun Sugihara (Depart- of Emergency and Critical Care Medicine, Komaki City Hos-
ment of pulmonology, Kashiwa municipal hospital), Jun pital), Michihiko Kono (Department of intensive Care Unit,
Takeshita (Department of Anesthesiology, Osaka Women's Sakai City Medical CenterMedicalcare unit), Michihito Kyo
and Children's Hospital), Junichi Fujimoto (Department of (Department of Emergency and Critical Care Medicine, Grad-
intensive care medicine, Yokohama Rosai Hospital), Junichi uate School of Biomedical and Health Sciences, Hiroshima
Ishikawa (Emergency and Critical Care Medical Center & Pe- University), Minoru Hayashi (Fukui Prefectural Hospital),
diatric emergency medicine Osaka City General Hospital), Mitsuhiro Abe (Respiratory Medicine, Chiba University Hos-
Junko Kosaka (Department of Anesthesiology and Resusci- pital), Mitsunori Sato (Department of pediatric critical care,
tology, Okayama University Hospital), Junpei Shibata Shizuoka children's hospital), Mizu Sakai (Department of
(Department of Anesthesiology and Critical Care Medicine, Respiratory medicine and Allergology, Kochi Medical School,
Fujita Health University School of Medicine), Katsuhiko Kochi University), Motoshi Kainuma (Anesthesiology, Emer-
Hashimoto (Department of Minimally Invasive Surgical and gency Medicine, and Intensive Care Division Inazawa Munic-
Medical Oncology, Fukushima Medical University, Fukush- ipal Hospital), Naoki Tominaga (Department of Emergency
ima, Japan), Yasushi Nakano (Kawasaki Municipal Ida Hospi- and Critical Care Medicine, Nippon Medical School), Naoya
tal), Kazuki Kikuyama (Department of Intensive Care Unit, Iguchi (Department of Anesthesiology and Intensive Care
Showa University Hospital), Kazushige Shimizu (Department Medicine, Graduate School of Medicine, Osaka University),
of Clinical Engineer), Kazuya Okada (Department of intensive Natsuki Nakagawa (Department of respiratory medicine,
care, Yokosuka General Hospital Uwamachi), Keishi Kawano Kanto Central Hospital of the Mutual Aid of Association of
(Department of Anesthesiology and Resuscitology, Okayama Public School Teachers), Nobumasa Aoki (Department of
University Hospital), Keisuke Anan (Division of Respiratory Respiratory Medicine and Infectious Diseases, Niigata Uni-
Medicine, Saiseikai Kumamoto Hospital), Keisuke Ota (Inten- versity Graduate School of Medical and Dental Sciences),
sive care center, Shizuoka General Hospital), Ken-ichi Kano Norihiro Nishioka (Department of Preventive Services, Kyoto
(Department of Emergency medicine, Fukui Prefectural University Graduate School of Medicine), Norihisa Miyashita
482 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

(Division of Pediatric Critical Care Medicine, Hyogo Prefectural Nagasaki University Hospital), Takeshi Umegaki (Department
Kobe Children's Hospital), Nozomu Seki (Department of of Anesthesiology, Kansai Medical University), Taku Fur-
Emergency, Toyama University Hospital), Ryo Ikebe (Depart- ukawa (Department of Anesthesiology and Critical Care, Jichi
ment of Nursing, Osaka Women's and Children's Hospital), Medical University Saitama Medical Center), Taku Omura
Ryosuke Imai (Department of Pulmonary Medicine, St. Luke's (Department of Emergency and Critical Care Medicine, Chiba
International Hospital), Ryota Tate (Department of Critical University Graduate School of Medicine), Takumi Nagao
Care Medicine, Tokyo Bay Urayasu-Ichikawa medical center), (Department of Sakakibara Heart Institute), Takuya Mayumi
Ryuhei Sato (Department of Critical Care Nursing, Graduate (Department of Cardiovascular Medicine, Graduate School of
School of Medicine Kyoto University), Sachiko Miyakawa Medical Science, Kanazawa University), Takuya Taniguchi
(Emergency Life-Saving Technique Academy of Kyushu, Pro- (Department of Cardiovascular Medicine, Otsu City Hospital),
fessor), Satoshi Kazuma (Department of Intensive Care Med- Takuya Yoshida (Department of Emergency and Critical Care
icine, Sapporo Medical University School of Medicine), Satoshi Medicine, Komaki City Hospital), Tatsutoshi Shimatani (Chu-
Nakano (Pediatric Emergency and Critical Care Center, Sai- goku Rosai Hospital), Teppei Murata (Department of Cardiol-
tama Children's Medical Center, Saitama, Japan), Satoshi ogy Tokyo Metropolitan Geriatric Hospital and Institute of
Tetsumoto (Department of Respiratory Medicine and Clinical Gerontology), Tetsuya Sato (Department of Emergency and
Immunology, Suita Municipal Hospital), Satoshi Yoshimura Critical Care Medicine, Tohoku University Hospital), Tohru
(Department of Preventive Services, Kyoto University Grad- Sawamoto (Department of Emergency and Critical Care Med-
uate School of Medicine), Shigenori Yoshitake (Department of icine, Tokai University School of Medicine), Yoshifumi Koukei
Health Science, Kyusyu University of Health and Welfare), (Tokyo Metropolitan Tama Medical Center Department of
Shin-etsu Hoshi (Iwate Prefectural Kamaishi Public Health Emergency Medicine Trauma and Resuscitation Center),
Center), Shingo Ohki (Department of Emergency and Critical Tomohiro Takehara (Division of Pulmonary Medicine,
Care Medicine, Graduate School of Biomedical and Health Department of Medicine, Keio University School of Medicine),
Sciences, Hiroshima University), Shintaro Sato (Department Tomomi Ueda (Department of Anesthesiology, Saiseikai
of Respirology, Saitama Red Cross Hospital), Shodai Yoshihiro Yokohamashi Tobu Hospital), Tomoya Katsuta (Department
(Department of Pharmacy, Onomichi General Hospital), Shoi- of respiratory medicine, Ehime Prefecture Central Hospital),
chi Ihara (Osaka Police Hospital, Respiratory medicine), Shota Tomoya Nishino (Department of Emergency and Critical Care,
Yamamoto (Department of Radiology, Tokai University Tokai University School of Medicine), Toshiki Yokoyama
Hachioji Hospital, Tokyo, Japan), Shunichi Koide (Department (Department of Respiratory Medicine and Allergy and
of Emergency and Critical Care Medicine, Urasoe General Department of Emergency and Intensive Care Medicine,
Hospital), Shunsuke Kimata (Department of Preventive Med- Intensive Care Unit/Tosei General Hospital), Ushio Higa-
icine, Kyoto University Graduate School of Public Health), shijima (Department of Anesthesiology and Intensive Care
Shunsuke Saito (Department of emergency, Okinawa Prefec- Medicine, Nagasaki University Graduate school of Biomedical
tural Chubu Hospital), Shunsuke Yasuo (Department of Sciences), Wataru Iwanaga (Department of emergency and
emergency medicine, Kyoto Katsura Hospital), Shusuke critical care medicine, Urasoe general hospital), Yasushi Inoue
Sekine (Department of Anesthesiology, Tokyo Medical Uni- (International University of Health and Welfare, Mita Hospital,
versity), Soichiro Mimuro (Department of Anesthesiology and Respiratory Diseases Center), Yoshiaki Iwashita (Emergency
Intensive Care Hamamatsu University School of Medicine), and Critical Care Medicine, Shimane University), Yoshie
Soichiro Wada (Department of pediatrics, Teine Keijinkai Yamada (Department of Healthcare Epidemiology, School of
Hospital), Sosuke Sugimura (Department of Medical Engi- Public Health in the Graduate School of Medicine, Kyoto Uni-
neering, Faculty of Health Care Science, Himeji Dokkyo Uni- versity), Yoshifumi Kubota (Kameda medical center depart-
versity), Tadashi Ishihara (Department of Emergency and ment of infectious diseases), Yoshihiro Suido (Department of
Critical Care Medicine, Juntendo University, Urayasu Hospi- Respiratory Medicine, Asao General Hospital), Yoshihiro
tal), Tadashi Kaneko (Emergency and Critical Care Center, Mie Tomioka (Department of Anesthesiology, Todachuo General
University Hospital), Tadashi Nagato (Department of Respi- Hospital), Yoshihisa Fujimoto (TMG Asaka Medical Center),
ratory Medicine, JCHO Tokyo Yamate Medical Center), Takaaki Yoshihito Fujita (Department of Anesthesiology and Intensive
Maruhashi (Department of Emergency and Critical care Med- Care Medicine, Aichi Medical University), Yoshikazu Yama-
icine, Kitasato University school of Medicine), Takahiro guchi (Department of Anesthesiology, Yokohama Municipal
Tamura (Department of Anesthesiology, Nagoya University Citizen's Hospital), Yoshimi Nakamura (Department of Emer-
Graduate School of Medicine), Takanori Ohno (Department of gency and Critical Care Medicine, Japanese Red Cross Kyoto
Emergency, SHOWA University Fujigaoka hospital), Takashi Daini Hospital), Yoshinobu Abe (Division of Emergency and
Ichiyama (Intensive Care Unit, Shinshu University Hospital), Disaster Medicine Tohoku Medical and Pharmaceutical Uni-
Takashi Niwa (Department of Respiratory Medicine, Kana- versity), Yoshitomo Eguchi (Saiseikai Kumamoto Hospital),
gawa Cardiovascular and Respiratory Center), Takashi Ueji Yoshiyasu Oshima (Bachelor of Pharmacy/Department of
(National Hospital Organization Osaka National Hospital), Pharmacy, Kobe Tokushukai Hospital), Yosuke Fukuda
Takayuki Ogura (Department of Emergency Medicine and (Department of Medicine, Division of Respiratory Medicine
Critical Care Medicine, Tochigi Prefectural Emergency and and Allergology, Showa University School of Medicine), Yudai
Critical Care Center, Imperial Gift Foundation SAISEIKAI, Iwasaki (Department of Anesthesiology and Perioperative
Utsunomiya Hospital), Takeshi Kawasaki (Department of Medicine, Tohoku University Graduate School of Medicine),
Respirology, Graduate School of Medicine, Chiba University), Yuichi Yasufuku (Department of Health Policy and Manage-
Takeshi Tanaka (Infection Control and Education Center, ment, School of Medicine, Keio University), Yuji Shono
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 483

(Emergency and Critical Care Center, Kyushu University [12] Karmpaliotis D, Kirtane AJ, Ruisi CP, Polonsky T,
Hospital), Yuka Nakatani (Department of Internal Medicine, Malhotra A, Talmor D, et al. Diagnostic and prognostic
Showa Inan General Hospital), Yuki Nakamori (Department of utility of brain natriuretic Peptide in subjects admitted to
the ICU with hypoxic respiratory failure due to
Molecular Pathobiology and Cell Adhesion Biology, Mie Uni-
noncardiogenic and cardiogenic pulmonary edema. Chest
versity Graduate School of Medicine), Yukie Ito (Department of 2007;131:964e71.
Intensive Care Medicine, Osaka Women's and Children's [13] Lin Q, Fu F, Chen H, Zhu B. Copeptin in the assessment of
Hospital), Yuko Tanabe (Department of Clinical Oncology, acute lung injury and cardiogenic pulmonary edema. Respir
Toranomon hospital), Yusuke Nagamine (Department of Med 2012;106:1268e77.
Anesthesiology and Critical Care Medicine, Yokohama City [14] Wussler D, Kozhuharov N, Tavares Oliveira M, Bossa A,
University Hospital), Yuta Nakamura (Emergency and Critical Sabti Z, Nowak A, et al. Clinical utility of procalcitonin in
the diagnosis of pneumonia. Clin Chem 2019;65:1532e42.
Care Centre, Saiseikai Kumamoto Hospital), and Yutaro Kur-
[15] Titova E, Christensen A, Henriksen AH, Steinshamn S,
ihara (Department of Emergency and Critical Care Medicine, 
Asberg A. Comparison of procalcitonin, C-reactive protein,
Kitasato University School of Medicine). white blood cell count and clinical status in diagnosing
pneumonia in patients hospitalized with acute
exacerbations of COPD: a prospective observational study.
Appendix B. Supplementary data Chron Respir Dis 2019;16:1479972318769762.
[16] Legriel S, Grigoresco B, Martel P, Henry-Lagarrigue M,
Lvovschi V, Troche  G, et al. Diagnostic accuracy of
Supplementary data to this article can be found online at
procalcitonin for early aspiration pneumonia in critically ill
https://doi.org/10.1016/j.resinv.2022.05.003. patients with coma: a prospective study. Neurocrit Care
2019;30:440e8.
[17] Ding HG, Zhou HF, Diao MY, Xu Y, Pan QM, Shen XH. A
references novel biomarker of serum Histidine-Rich Glycoprotein
(HRG) for diagnosing and predicting prognosis of ventilator-
associated pneumonia (VAP): a pilot study. Eur Rev Med
[1] Japanese Society of Respiratory Care Medicine. ARDS Pharmacol Sci 2018;22:7920e7.
clinical practice guidelines 1st edition. (In Japanese). Jpn J [18] Chen C, Yan M, Hu C, Lv X, Zhang H, Chen S. Diagnostic
Respir Care 1999;16:95e115. efficacy of serum procalcitonin, C-reactive protein
[2] Japanese Society of Respiratory Care Medicine. ARDS concentration and clinical pulmonary infection score in
clinical practice guidelines 2nd edition. (In Japanese). Jpn J Ventilator-Associated Pneumonia. Med Sci (Paris)
Respir Care 2004;21:44e61. 2018;34:26e32.
[3] Japanese Respiratory Society ARDS Guideline Committee. [19] Berge K, Lyngbakken MN, Einvik G, Winther JA, Brynildsen J,
Clinical practical guideline for acute lung injury and acute Røysland R, et al. Diagnostic and prognostic properties of
respiratory distress syndrome (In Japanese). 2005. procalcitonin in patients with acute dyspnea: data from the
[4] Japanese Respiratory Society ARDS Guideline Committee. ACE 2 Study. Clin Biochem 2018;59:62e8.
Clinical practical guideline for acute lung injury and acute [20] Habib SF, Mukhtar AM, Abdelreheem HM, Khorshied MM,
respiratory distress syndrome (In Japanese). second ed. 2010. Sayed RE, Hafez MH, et al. Diagnostic values of CD64, C-
[5] Hashimoto S, Sanui M, Egi M, Ohshimo S, Shiotsuka J, Seo R, reactive protein and procalcitonin in ventilator-associated
et al. The clinical practice guideline for the management of pneumonia in adult trauma patients: a pilot study. Clin
ARDS in Japan. J Intensive Care 2017;5:50. Chem Lab Med 2016;54:889e95.
[6] Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, [21] Le Bel J, Hausfater P, Chenevier-Gobeaux C, Blanc F-X,
et al. Epidemiology, patterns of care, and mortality for Benjoar M, Ficko C, et al. Diagnostic accuracy of C-reactive
patients with acute respiratory distress syndrome in protein and procalcitonin in suspected community-
intensive care units in 50 countries. JAMA 2016;315:788e800. acquired pneumonia adults visiting emergency department
[7] Japan Council for Quality Health Care. The Minds guideline and having a systematic thoracic CT scan. Crit Care
creation manual. 2017. Available at: https://minds.jcqhc.or. 2015;19:366.
jp/s/guidance_2017_0_h. [22] Porfyridis I, Georgiadis G, Vogazianos P, Mitis G, Georgiou A.
[8] Weiss SL, Peters MJ, Alhazzani W, Agus MSD, Flori HR, C-reactive protein, procalcitonin, clinical pulmonary
Inwald DP, et al. Surviving sepsis campaign international infection score, and pneumonia severity scores in nursing
guidelines for the management of septic shock and sepsis- home acquired pneumonia. Respir Care 2014;59:574e81.
associated organ dysfunction in children. Pediatr Crit Care [23] Bafadhel M, Clark TW, Reid C, Medina M-J, Batham S,
Med 2020;21:e52e106. Barer MR, et al. Procalcitonin and C-reactive protein in
[9] Guyatt GH, Alonso-Coello P, Schünemann HJ, Djulbegovic B, hospitalized adult patients with community-acquired
Nothacker M, Lange S, et al. Guideline panels should seldom pneumonia or exacerbation of asthma or COPD. Chest
make good practice statements: guidance from the GRADE 2011;139:1410e8.
Working Group. J Clin Epidemiol 2016;80:3e7. [24] Ramirez P, Garcia MA, Ferrer M, Aznar A, Valencia M,
[10] Komiya K, Ishii H, Teramoto S, Takahashi O, Eshima N, Sahuquillo JM, et al. Sequential measurements of
Yamaguchi O, et al. Diagnostic utility of C-reactive protein procalcitonin levels in diagnosing ventilator-associated
combined with brain natriuretic peptide in acute pneumonia. Eur Respir J 2008;31:356e62.
pulmonary edema: a cross sectional study. Respir Res [25] Müller B, Harbarth S, Stolz D, Bingisser R, Mueller C,
2011;12:83. Leuppi J, et al. Diagnostic and prognostic accuracy of
[11] Levitt JE, Vinayak AG, Gehlbach BK, Pohlman A, Van clinical and laboratory parameters in community-acquired
Cleve W, Hall JB, et al. Diagnostic utility of B-type pneumonia. BMC Infect Dis 2007;7:10.
natriuretic peptide in critically ill patients with [26] Holm A, Pedersen SS, Nexoe J, Obel N, Nielsen LP,
pulmonary edema: a prospective cohort study. Crit Care Koldkjaer O, et al. Procalcitonin versus C-reactive protein
2008;12:R3. for predicting pneumonia in adults with lower respiratory
484 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

tract infection in primary care. Br J Gen Pract [43] Fukushima K, Nakamura S, Inoue Y, Higashiyama Y,
2007;57:555e60. Ohmichi M, Ishida T, et al. Utility of a sputum antigen
[27] Adnet F, Borron SW, Vicaut E, Giraudeaux V, Lapostolle F, detection test in pneumococcal pneumonia and lower
Bekka R, et al. Value of C-reactive protein in the detection of respiratory infectious disease in adults. Intern Med
bacterial contamination at the time of presentation in drug- 2015;54:2843e50.
induced aspiration pneumonia. Chest 1997;112:466e71. [44] Sorde  R, Falco  V, Lowak M, Domingo E, Ferrer A, Burgos J,
[28] Shokri M, Ghasemian R, Bayani M, Maleh PA, Kamrani M, et al. Current and potential usefulness of pneumococcal
Sadeghi-Haddad-Zavareh M, et al. Serum and alveolar urinary antigen detection in hospitalized patients with
procalcitonin had a weak diagnostic value for ventilator- community-acquired pneumonia to guide antimicrobial
associated pneumonia in patients with pulmonary therapy. Arch Intern Med 2011;171:166e72.
infection score  6. Rom J Intern Med 2018;56:9e14. [45] Abdeldaim G, Herrmann B, Korsgaard J, Olce n P, Blomberg J,
[29] Self WH, Balk RA, Grijalva CG, Williams DJ, Zhu Y, Strålin K. Is quantitative PCR for the pneumolysin (ply) gene
Anderson EJ, et al. Procalcitonin as a marker of etiology in useful for detection of pneumococcal lower respiratory
adults hospitalized with community-acquired pneumonia. tract infection? Clin Microbiol Infect 2009;15:565e70.
Clin Infect Dis 2017;65:183e90. [46] Weatherall C, Paoloni R, Gottlieb T. Point-of-care urinary
[30] Heining L, Giesa C, Ewig S. MR-proANP, MR-proADM, and pneumococcal antigen test in the emergency department
PCT in patients presenting with acute dyspnea in a medical for community acquired pneumonia. Emerg Med J
emergency unit. Lung 2016;194:185e91. 2008;25:144e8.
[31] Alba GA, Truong QA, Gaggin HK, Gandhi PU, De [47] Tzeng DH, Lee YL, Lin YH, Tsai CA, Shi ZY. Diagnostic value
Berardinis B, Magrini L, et al. Diagnostic and prognostic of the Binax NOW assay for identifying a pneumococcal
utility of procalcitonin in patients presenting to the etiology in patients with respiratory tract infection. J
emergency department with dyspnea. Am J Med Microbiol Immunol Infect 2006;39:39e44.
2016;129:96e104. [48] Lasocki S, Scanvic A, Le Turdu F, Restoux A, Mentec H,
[32] Dallas J, Brown SM, Hock K, Scott MG, Skrupky LP, Bleichner G, et al. Evaluation of the Binax NOW
Boyle 3rd WA, et al. Diagnostic utility of plasma Streptococcus pneumoniae urinary antigen assay in
procalcitonin for nosocomial pneumonia in the intensive intensive care patients hospitalized for pneumonia.
care unit setting. Respir Care 2011;56:412e9. Intensive Care Med 2006;32:1766e72.
[33] Luyt CE, Combes A, Reynaud C, Hekimian G, [49] Genne  D, Siegrist HH, Lienhard R. Enhancing the etiologic
Nieszkowska A, Tonnellier M, et al. Usefulness of diagnosis of community-acquired pneumonia in adults
procalcitonin for the diagnosis of ventilator-associated using the urinary antigen assay (Binax NOW). Int J Infect Dis
pneumonia. Intensive Care Med 2008;34:1434e40. 2006;10:124e8.
[34] Boussekey N, Leroy O, Georges H, Devos P, d'Escrivan T, [50] Briones ML, Blanquer J, Ferrando D, Blasco ML, Gimeno C,
Guery B. Diagnostic and prognostic values of admission Marı́n J. Assessment of analysis of urinary pneumococcal
procalcitonin levels in community-acquired pneumonia in antigen by immunochromatography for etiologic diagnosis
an intensive care unit. Infection 2005;33:257e63. of community-acquired pneumonia in adults. Clin Vaccine
[35] Duflo F, Debon R, Monneret G, Bienvenu J, Chassard D, Immunol 2006;13:1092e7.
Allaouchiche B. Alveolar and serum procalcitonin: [51] Strålin K, Kaltoft MS, Konradsen HB, Olce n P, Holmberg H.
diagnostic and prognostic value in ventilator-associated Comparison of two urinary antigen tests for establishment
pneumonia. Anesthesiology 2002;96:74e9. of pneumococcal etiology of adult community-acquired
[36] Lee J, Song JU. Performance of pneumococcal urinary pneumonia. J Clin Microbiol 2004;42:3620e5.
antigen test in patients with community-onset pneumonia: [52] Ishida T, Hashimoto T, Arita M, Tojo Y, Tachibana H,
a propensity score-matching study. Korean J Intern Med Jinnai M. A 3-year prospective study of a urinary antigen-
2020;35:630e40. detection test for Streptococcus pneumoniae in
[37] Zhou F, Gu L, Qu JX, Liu YM, Cao B. Evaluating the utility of community-acquired pneumonia: utility and clinical
Binax NOW Streptococcus pneumoniae urinary antigen test impact on the reported etiology. J Infect Chemother
in adults with community acquired pneumonia in China. 2004;10:359e63.
Clin Respir J 2018;12:425e32. [53] Marcos MA, Jime nez de Anta MT, de la Bellacasa JP,
[38] Burgos J, Garcia-Pe rez JN, di Lauro SG, Falco  V, Pumarola T, Gonza  lez J, Martı́nez E, Garcı́a E, et al. Rapid urinary antigen
Almirante B, et al. Usefulness of Sofia Pneumococcal FIA® test for diagnosis of pneumococcal community-acquired
test in comparison with BinaxNOW® Pneumococcal test in pneumonia in adults. Eur Respir J 2003;21:209e14.
urine samples for the diagnosis of pneumococcal [54] Gutierrez F, Masia  M, Rodrı́guez JC, Ayelo A, Solda  n B,
pneumonia. Eur J Clin Microbiol Infect Dis 2018;37:1289e95. Cebria  n L, et al. Evaluation of the immunochromatographic
[39] Laijen W, Snijders D, Boersma WG. Pneumococcal urinary Binax NOW assay for detection of Streptococcus
antigen test: diagnostic yield and impact on antibiotic pneumoniae urinary antigen in a prospective study of
treatment. Clin Respir J 2017;11:999e1005. community-acquired pneumonia in Spain. Clin Infect Dis
[40] Ikegame S, Nakano T, Otsuka J, Yoshimi M, Matsuo T, 2003;36:286e92.
Kubota M, et al. The evaluation of the sputum antigen kit in [55] Butler JC, Bosshardt SC, Phelan M, Moroney SM,
the diagnosis of pneumococcal pneumonia. Intern Med Tondella ML, Farley MM, et al. Classical and latent class
2017;56:1141e6. analysis evaluation of sputum polymerase chain reaction
[41] €
Athlin S, Iversen A, Ozenci V. Comparison of the ImmuView and urine antigen testing for diagnosis of pneumococcal
and the BinaxNOW antigen tests in detection of pneumonia in adults. J Infect Dis 2003;187:1416e23.
Streptococcus pneumoniae and Legionella pneumophila in [56] Farina C, Arosio M, Vailati F, Moioli F, Goglio A. Urinary
urine. Eur J Clin Microbiol Infect Dis 2017;36:1933e8. detection of Streptococcus pneumoniae antigen for
[42] Molinos L, Zalacain R, Mene ndez R, Soledad Reyes S, diagnosis of pneumonia. New Microbiol 2002;25:259e63.
Capelastegui A, Cillo  niz C, et al. Sensitivity, specificity, and [57] Murdoch DR, Laing RT, Mills GD, Karalus NC, Town GI,
positivity predictors of the pneumococcal urinary antigen Mirrett S, et al. Evaluation of a rapid
test in community-acquired pneumonia. Ann Am Thorac immunochromatographic test for detection of
Soc 2015;12:1482e9. Streptococcus pneumoniae antigen in urine samples from
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 485

adults with community-acquired pneumonia. J Clin [73] Diederen BMW, Bruin JP, Scopes E, Peeters MF, IJzerman EP.
Microbiol 2001;39:3495e8. Evaluation of the Oxoid Xpect Legionella test kit for
[58] Burel E, Dufour P, Gauduchon V, Jarraud S, Etienne J. detection of Legionella pneumophila serogroup 1 antigen in
Evaluation of a rapid immunochromatographic assay for urine. J Clin Microbiol 2009;47:2272e4.
detection of Streptococcus pneumoniae antigen in urine [74] Diederen BM, Peeters MF. Evaluation of the SAS Legionella
samples. Eur J Clin Microbiol Infect Dis 2001;20:840e1. Test, a new immunochromatographic assay for the
[59] Fukuyama H, Yamashiro S, Kinjo K, Tamaki H, Kishaba T. detection of Legionella pneumophila serogroup 1 antigen in
Validation of sputum Gram stain for treatment of urine. Clin Microbiol Infect 2007;13:86e8.
community-acquired pneumonia and healthcare- [75] Koide M, Higa F, Tateyama M, Nakasone I, Yamane N,
associated pneumonia: a prospective observational study. Fujita J. Detection of legionella species in clinical samples:
BMC Infect Dis 2014;14:534. comparison of polymerase chain reaction and urinary
[60] Akter S, Shamsuzzaman SM, Jahan F. Community acquired antigen detection kits. Infection 2006;34:264e8.
bacterial pneumonia: aetiology, laboratory detection and [76] Diederen BM, Peeters MF. Evaluation of two new
antibiotic susceptibility pattern. Malays J Pathol immunochromatographic assays (Rapid U Legionella
2014;36:97e103. antigen test and SD Bioline Legionella antigen test) for
[61]  L, Llopis R, Jacob J, Juan A, Palom X, Barde
Ferre s I, et al. Is detection of Legionella pneumophila serogroup 1 antigen in
sputum Gram staining useful in the emergency urine. J Clin Microbiol 2006;44:2991e3.
department's management of pneumonia. Emergencias [77] Diederen BM, Peeters MF. Evaluation of Rapid U
2011;23:108e11. Legionella Plus Test, a new immunochromatographic assay
[62] Miyashita N, Shimizu H, Ouchi K, Kawasaki K, Kawai Y, for detection of Legionella pneumophila serogroup 1
Obase Y, et al. Assessment of the usefulness of sputum antigen in urine. Eur J Clin Microbiol Infect Dis
Gram stain and culture for diagnosis of community- 2006;25:733e5.
acquired pneumonia requiring hospitalization. Med Sci [78] Helbig JH, Uldum SA, Lück PC, Harrison TG. Detection of
Monit 2008;14:Cr171e6. Legionella pneumophila antigen in urine samples by the
[63] Yang S, Lin S, Khalil A, Gaydos C, Nuemberger E, Juan G, BinaxNOW immunochromatographic assay and
et al. Quantitative PCR assay using sputum samples for comparison with both Binax Legionella Urinary Enzyme
rapid diagnosis of pneumococcal pneumonia in adult Immunoassay (EIA) and Biotest Legionella Urin Antigen EIA.
emergency department patients. J Clin Microbiol J Med Microbiol 2001;50:509e16.
2005;43:3221e6. [79] Domı́nguez J, Galı́ N, Blanco S, Pedroso P, Prat C, Matas L,
[64] Garcı́a-Va zquez E, Marcos MA, Mensa J, de Roux A, Puig J, et al. Assessment of a new test to detect Legionella urinary
Font C, et al. Assessment of the usefulness of sputum antigen for the diagnosis of Legionnaires' Disease. Diagn
culture for diagnosis of community-acquired pneumonia Microbiol Infect Dis 2001;41:199e203.
using the PORT predictive scoring system. Arch Intern Med [80] Kazandjian D, Chiew R, Gilbert GL. Rapid diagnosis of
2004;164:1807e11. Legionella pneumophila serogroup 1 infection with the
[65] Roso n B, Carratala J, Verdaguer R, Dorca J, Manresa F, Binax enzyme immunoassay urinary antigen test. J Clin
Gudiol F. Prospective study of the usefulness of sputum Microbiol 1997;35:954e6.
Gram stain in the initial approach to community-acquired [81] Plouffe JF, File Jr TM, Breiman RF, Hackman BA, Salstrom SJ,
pneumonia requiring hospitalization. Clin Infect Dis Marston BJ, et al. Reevaluation of the definition of
2000;31:869e74. Legionnaires' disease: use of the urinary antigen assay.
[66] Fine MJ, Orloff JJ, Rihs JD, Vickers RM, Kominos S, Community Based Pneumonia Incidence Study Group. Clin
Kapoor WN, et al. Evaluation of housestaff physicians' Infect Dis 1995;20:1286e91.
preparation and interpretation of sputum Gram stains for [82] Leland DS, Kohler RB. Evaluation of the L-CLONE Legionella
community-acquired pneumonia. J Gen Intern Med pneumophila serogroup 1 urine antigen latex test. J Clin
1991;6:189e98. Microbiol 1991;29:2220e3.
[67] Zhang XP, Deng KE, Ye YQ, Luo WT. Rapid detection of [83] Samuel D, Harrison TG, Taylor AG. Detection of Legionella
pneumococcal antigens in sputa in patients with pneumophila serogroup 1 urinary antigen using an
community-acquired pneumonia by coagglutination. Med enhanced chemiluminescence ELISA. J Biolumin
Microbiol Immunol 1988;177:333e8. Chemilumin 1990;5:183e5.
[68] Rein MF, Gwaltney Jr JM, O'Brien WM, Jennings RH, [84] Birtles RJ, Harrison TG, Samuel D, Taylor AG. Evaluation of
Mandell GL. Accuracy of Gram's stain in identifying urinary antigen ELISA for diagnosing Legionella
pneumococci in sputum. JAMA 1978;239:2671e3. pneumophila serogroup 1 infection. J Clin Pathol
[69] Peci A, Winter AL, Gubbay JB. Evaluation and comparison of 1990;43:685e90.
multiple test methods, including real-time PCR, for [85] Aguero-Rosenfeld ME, Edelstein PH. Retrospective
Legionella detection in clinical specimens. Front Public evaluation of the Du Pont radioimmunoassay kit for
Health 2016;4:175. detection of Legionella pneumophila serogroup 1
[70] Gadsby NJ, Helgason KO, Dickson EM, Mills JM, Lindsay DSJ, antigenuria in humans. J Clin Microbiol 1988;26:1775e8.
Edwards GF, et al. Molecular diagnosis of Legionella [86] Tang PW, Toma S. Broad-spectrum enzyme-linked
infections–Clinical utility of front-line screening as part of a immunosorbent assay for detection of Legionella soluble
pneumonia diagnostic algorithm. J Infect 2016;72:161e70. antigens. J Clin Microbiol 1986;24:556e8.
[71] Jørgensen CS, Uldum SA, Sørensen JF, Skovsted IC, Otte S, [87] White A, Kohler RB, Wheat LJ, Sathapatayavongs B,
Elverdal PL. Evaluation of a new lateral flow test for detection Winn Jr WC, Girod JC, et al. Rapid diagnosis of Legionnaires'
of Streptococcus pneumoniae and Legionella pneumophila disease. Trans Am Clin Climatol Assoc 1982;93:50e62.
urinary antigen. J Microbiol Methods 2015;116:33e6. [88] Sathapatayavongs B, Kohler RB, Wheat LJ, White A,
[72] Svarrer CW, Lück C, Elverdal PL, Uldum SA. Winn Jr WC, Girod JC, et al. Rapid diagnosis of Legionnaires'
Immunochromatic kits Xpect Legionella and BinaxNOW disease by urinary antigen detection. Comparison of ELISA
Legionella for detection of Legionella pneumophila urinary and radioimmunoassay. Am J Med 1982;72:576e82.
antigen have low sensitivities for the diagnosis of [89] Honda J, Yonemitsu J, Kitajima H, Yosida N, Fumirori T,
Legionnaires' disease. J Med Microbiol 2012;61:213e7. Oizumi K. Clinical utility of capillary polymerase chain
486 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

reaction for diagnosis of Cytomegalovirus pneumonia. neutropenic patients with haematological malignancies.
Scand J Infect Dis 2001;33:702e5. Mycoses 2018;61:650e5.
[90] de la Hoz RE, Byrne SK, Hayashi S, Sherlock C, Cook D, [104] Dobias R, Jaworska P, Tomaskova H, Kanova M, Lyskova P,
Hogg JC. Diagnosis of cytomegalovirus infection in HIV- Vrba Z, et al. Diagnostic value of serum galactomannan,
infected patients with respiratory disease. Clin Diagn Virol (1,3)-b-d-glucan, and Aspergillus fumigatus-specific IgA and
1998;10:1e7. IgG assays for invasive pulmonary aspergillosis in non-
[91] Hansen KK, Vestbo J, Benfield T, Lundgren JD, Mathiesen LR. neutropenic patients. Mycoses 2018;61:576e86.
Rapid detection of cytomegalovirus in bronchoalveolar [105] Boch T, Reinwald M, Spiess B, Liebregts T, Schellongowski P,
lavage fluid and serum samples by polymerase chain Meybohm P, et al. Detection of invasive pulmonary
reaction: correlation of virus isolation and clinical outcome aspergillosis in critically ill patients by combined use of
for patients with human immunodeficiency virus infection. conventional culture, galactomannan, 1-3-beta-D-glucan
Clin Infect Dis 1997;24:878e83. and Aspergillus specific nested polymerase chain reaction
[92] Liesnard C, De Wit L, Motte S, Brancart F, Content J. Rapid in a prospective pilot study. J Crit Care 2018;47:198e203.
diagnosis of cytomegalovirus lung infection by DNA [106] Lahmer T, Rasch S, Schnappauf C, Beitz A, Schmid RM,
amplification in bronchoalveolar lavages. Mol Cell Probes Huber W. Comparison of serum galactomannan and 1,3-
1994;8:273e83. beta-D-glucan determination for early detection of invasive
[93] Eriksson BM, Brytting M, Zweygberg-Wirgart B, Hillerdal G, pulmonary aspergillosis in critically ill patients with
Olding-Stenkvist E, Linde A. Diagnosis of cytomegalovirus hematological malignancies and septic shock.
in bronchoalveolar lavage by polymerase chain reaction, in Mycopathologia 2016;181:505e11.
comparison with virus isolation and detection of viral [107] Bo €
€ lük G, Kazak E, Ozkalemkas  ca H,
‚ F, Ener B, Akalin H, Ag
antigen. Scand J Infect Dis 1993;25:421e7. et al. Comparison of galactomannan, beta-D-glucan, and
[94] Moon SM, Sung H, Kim MN, Lee SO, Choi SH, Kim YS, et al. Aspergillus DNA in sera of high-risk adult patients with
Diagnostic yield of the cytomegalovirus (CMV) antigenemia hematological malignancies for the diagnosis of invasive
assay and clinical features in solid organ transplant aspergillosis. Turk J Med Sci 2016;46:335e42.
recipients and hematopoietic stem cell transplant [108] Metan G, Koç AN, Kaynar LG, Atalay A, Oztürk A, Eser B,
recipients with CMV pneumonia. Transpl Infect Dis et al. What is the role of the (1/3)-b-D-glucan assay in the
2012;14:192e7. screening of patients undergoing autologous
[95] Uberti-Foppa C, Lillo F, Terreni MR, Puglisi A, Guffanti M, haematopoietic stem-cell transplantation? Mycoses
Gianotti N, et al. Cytomegalovirus pneumonia in 2013;56:34e8.
AIDS patients: value of cytomegalovirus culture from BAL [109] Sun YQ, Ji Y, Xu LP, Liu DH, Liu KY, Huang XJ. Combination
fluid and correlation with lung disease. Chest of real-time polymerase chain reaction assay and serum
1998;113:919e23. galactomannan in the diagnosis of invasive aspergillosis in
[96] Angelici E, Contini C, Sebastiani G, Folgori F, Delia S, Serra P, patients with hematological malignancies and recipients of
et al. Cytomegalovirus in bronchoalveolar lavage hematopoietic stem cell transplantation. Zhonghua Yi Xue
specimens from patients with AIDS: comparison with Za Zhi 2010;90:375e8.
antigenaemia and viraemia. J Med Microbiol [110] Busca A, Locatelli F, Barbui A, Limerutti G, Serra R,
1996;45:149e52. Libertucci D, et al. Usefulness of sequential Aspergillus
[97] Engsbro AL, Najat S, Jørgensen KM, Kurtzhals JAL, galactomannan antigen detection combined with early
Arendrup MC. Diagnostic accuracy of the 1,3-b-D-glucan radiologic evaluation for diagnosis of invasive pulmonary
test for pneumocystis pneumonia in a tertiary university aspergillosis in patients undergoing allogeneic stem cell
hospital in Denmark: a retrospective study. Med Mycol transplantation. Transplant Proc 2006;38:1610e3.
2019;57:710e7. [111] Marr KA, Balajee SA, McLaughlin L, Tabouret M, Bentsen C,
[98] Passos AIM, Dertkigil RP, Ramos MC, Busso-Lopes AF, Walsh TJ. Detection of galactomannan antigenemia by
Tararan C, Ribeiro EO, et al. Serum markers as an aid in the enzyme immunoassay for the diagnosis of invasive
diagnosis of pulmonary fungal infections in AIDS patients. aspergillosis: variables that affect performance. J Infect Dis
Braz J Infect Dis 2017;21:606e12. 2004;190:641e9.
[99] Lahmer T, da Costa CP, Held J, Rasch S, Ehmer U, Schmid RM, [112] Pinel C, Fricker-Hidalgo H, Lebeau B, Garban F, Hamidfar R,
et al. Usefulness of 1,3 Beta-D-Glucan detection in non-HIV Ambroise-Thomas P, et al. Detection of circulating
immunocompromised mechanical ventilated critically ill Aspergillus fumigatus galactomannan: value and limits of
patients with ARDS and suspected Pneumocystis jirovecii the Platelia test for diagnosing invasive aspergillosis. J Clin
pneumonia. Mycopathologia 2017;182:701e8. Microbiol 2003;41:2184e6.
[100] Garnham K, Halliday CL, Rai NJ, Jayawardena M, Hasan T, [113] Becker MJ, Lugtenburg EJ, Cornelissen JJ, Van Der Schee C,
Kok J, et al. Introducing 1,3-Beta-D-glucan for screening and Hoogsteden HC, De Marie S. Galactomannan detection in
diagnosis of invasive fungal diseases in Australian high risk computerized tomography-based broncho-alveolar lavage
haematology patients: is there a clinical benefit? Intern Med fluid and serum in haematological patients at risk for
J 2022;52:426e35. invasive pulmonary aspergillosis. Br J Haematol
[101] Chang E, Kim TS, Kang CK, Jun KI, Shin D, Koh Y, et al. 2003;121:448e57.
Limited positive predictive value of b-d-Glucan in [114] Maertens J, Van Eldere J, Verhaegen J, Verbeken E,
hematologic patients receiving antimold prophylaxis. Open Verschakelen J, Boogaerts M. Use of circulating
Forum Infect Dis 2020;7:ofaa048. galactomannan screening for early diagnosis of invasive
[102] Heldt S, Prattes J, Eigl S, Spiess B, Flick H, Rabensteiner J, aspergillosis in allogeneic stem cell transplant recipients. J
et al. Diagnosis of invasive aspergillosis in hematological Infect Dis 2002;186:1297e306.
malignancy patients: performance of cytokines, Asp LFD, [115] Ulusakarya A, Chachaty E, Vantelon JM, Youssef A,
and Aspergillus PCR in same day blood and bronchoalveolar Tancre de C, Pico JL, et al. Surveillance of Aspergillus
lavage samples. J Infect 2018;77:235e41. galactomannan antigenemia for invasive aspergillosis by
[103] Furfaro E, Giacobbe DR, Del Bono V, Signori A, Guolo F, enzyme-linked immunosorbent assay in neutropenic
Minetto P, et al. Performance of serum (1,3)-ß-d-glucan patients treated for hematological malignancies. Hematol J
screening for the diagnosis of invasive aspergillosis in 2000;1:111e6.
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 487

[116] Bretagne S, Costa JM, Bart-Delabesse E, Dhe din N, Rieux C, [131] Chung JH, Kradin RL, Greene RE, Shepard JA,
Cordonnier C. Comparison of serum galactomannan Digumarthy SR. CT predictors of mortality in pathology
antigen detection and competitive polymerase chain confirmed ARDS. Eur Radiol 2011;21:730e7.
reaction for diagnosing invasive aspergillosis. Clin Infect [132] Ichikado K, Suga M, Muranaka H, Gushima Y, Miyakawa H,
Dis 1998;26:1407e12. Tsubamoto M, et al. Prediction of prognosis for acute
[117] Arrieta E, Sangiovanni S, Garcia-Robledo JE, Vela  squez M, respiratory distress syndrome with thin-section CT:
Sua LF, Ferna  ndez-Trujillo L. Video-assisted thoracoscopic validation in 44 cases. Radiology 2006;238:321e9.
lung biopsy in critically ill patients with hematologic [133] Rouby JJ, Puybasset L, Cluzel P, Richecoeur J, Lu Q, Grenier P.
malignancy and acute respiratory distress syndrome: a case Regional distribution of gas and tissue in acute respiratory
series report. J Investig Med High Impact Case Rep distress syndrome. II. Physiological correlations and
2020;8:2324709620912101. definition of an ARDS Severity Score. CT Scan ARDS Study
[118] Lipps KM, Bharat A, Walter JM. Lung biopsy in patients with Group. Intensive Care Med 2000;26:1046e56.
acute respiratory distress syndrome supported on [134] Song M, Liu Y, Lu Z, Luo H, Peng H, Chen P. Prognostic
extracorporeal membrane oxygenation: a 2 Year factors for ARDS: clinical, physiological and atypical
experience. ASAIO J 2019;65:e92e4. immunodeficiency. BMC Pulm Med 2020;20:102.
[119] Cardinal-Fernandez P, Ortiz G, Chang CH, Kao KC, [135] Fujishima S, Gando S, Saitoh D, Kushimoto S, Ogura H,
Bertreau E, Philipponnet C, et al. Predicting the impact of Abe T, et al. Demographics, treatments, and outcomes of
diffuse alveolar damage through open lung biopsy in acute acute respiratory distress syndrome: the focused outcomes
respiratory distress syndrome-The PREDATOR study. J Clin research in emergency care in acute respiratory distress
Med 2019;8:829. syndrome, sepsis, and trauma (FORECAST) study. Shock
[120] Park J, Lee YJ, Lee J, Park SS, Cho YJ, Lee SM, et al. 2020;53:544e9.
Histopathologic heterogeneity of acute respiratory distress [136] Shen Y, Cai G, Gong S, Dong L, Yan J, Cai W. Interaction
syndrome revealed by surgical lung biopsy and its clinical between low tidal volume ventilation strategy and severity
implications. Korean J Intern Med 2018;33:532e40. of acute respiratory distress syndrome: a retrospective
[121] Willetts L, Parker K, Wesselius LJ, Protheroe CA, Jaben E, cohort study. Crit Care 2019;23:254.
Graziano P, et al. Immunodetection of occult eosinophils in [137] Chinh LQ, Manabe T, Son DN, Chi NV, Fujikura Y, Binh NG,
lung tissue biopsies may help predict survival in acute lung et al. Clinical epidemiology and mortality on patients with
injury. Respir Res 2011;12:116. acute respiratory distress syndrome (ARDS) in Vietnam.
[122] Charbonney E, Robert J, Pache JC, Chevrolet JC, Eggimann P. PLoS One 2019;14:e0221114.
Impact of bedside open lung biopsies on the management [138] Chan MC, Chao WC, Liang SJ, Tseng CH, Wang HC,
of mechanically ventilated immunocompromised patients Chien YC, et al. First tidal volume greater than 8 mL/kg is
with acute respiratory distress syndrome of unknown associated with increased mortality in complicated
etiology. J Crit Care 2009;24:122e8. influenza infection with acute respiratory distress
[123] Baumann HJ, Kluge S, Balke L, Yekebas E, Izbicki JR, syndrome. J Formos Med Assoc 2019;118:378e85.
Amthor M, et al. Yield and safety of bedside open lung [139] Neuschwander A, Lemiale V, Darmon M, Pe  ne F,
biopsy in mechanically ventilated patients with acute lung Kouatchet A, Perez P, et al. Noninvasive ventilation during
injury or acute respiratory distress syndrome. Surgery acute respiratory distress syndrome in patients with
2008;143:426e33. cancer: trends in use and outcome. J Crit Care
[124] Papazian L, Doddoli C, Chetaille B, Gernez Y, Thirion X, 2017;38:295e9.
Roch A, et al. A contributive result of open-lung biopsy [140] Kallet RH, Zhuo H, Ho K, Lipnick MS, Gomez A, Matthay MA.
improves survival in acute respiratory distress syndrome Lung injury etiology and other factors influencing the
patients. Crit Care Med 2007;35:755e62. relationship between dead-space fraction and mortality in
[125] Canzian M, Soeiro Ade M, Taga MF, Barbas CS, Capelozzi VL. ARDS. Respir Care 2017;62:1241e8.
Correlation between surgical lung biopsy and autopsy [141] DesPrez K, McNeil JB, Wang C, Bastarache JA, Shaver CM,
findings and clinical data in patients with diffuse Ware LB. Oxygenation saturation index predicts clinical
pulmonary infiltrates and acute respiratory failure. Clinics outcomes in ARDS. Chest 2017;152:1151e8.
(Sao Paulo) 2006;61:425e32. [142] Bellani G, Laffey JG, Pham T, Madotto F, Fan E, Brochard L,
[126] Patel SR, Karmpaliotis D, Ayas NT, Mark EJ, Wain J, et al. Noninvasive ventilation of patients with acute
Thompson BT, et al. The role of open-lung biopsy in ARDS. respiratory distress syndrome. Insights from the LUNG
Chest 2004;125:197e202. SAFE study. Am J Respir Crit Care Med 2017;195:67e77.
[127] Papazian L, Thomas P, Bregeon F, Garbe L, Zandotti C, Saux P, [143] Lazzeri C, Bonizzoli M, Cozzolino M, Verdi C, Cianchi G,
et al. Open-lung biopsy in patients with acute respiratory Batacchi S, et al. Serial measurements of troponin and
distress syndrome. Anesthesiology 1998;88:935e44. echocardiography in patients with moderate-to-severe
[128] Nishiyama A, Kawata N, Yokota H, Sugiura T, Matsumura Y, acute respiratory distress syndrome. J Crit Care
Higashide T, et al. A predictive factor for patients with acute 2016;33:132e6.
respiratory distress syndrome: CT lung volumetry of the [144] Lai CC, Sung MI, Liu HH, Chen CM, Chiang SR, Liu WL, et al.
well-aerated region as an automated method. Eur J Radiol The ratio of partial pressure arterial oxygen and fraction of
2020;122:108748. inspired oxygen 1 Day after acute respiratory distress
[129] Kamo T, Tasaka S, Suzuki T, Asakura T, Suzuki S, Yagi K, syndrome onset can predict the outcomes of involving
et al. Prognostic values of the Berlin definition criteria, patients. Medicine (Baltimore) 2016;95:e3333.
blood lactate level, and fibroproliferative changes on high- [145] Laffey JG, Bellani G, Pham T, Fan E, Madotto F, Bajwa EK,
resolution computed tomography in ARDS patients. BMC et al. Potentially modifiable factors contributing to outcome
Pulm Med 2019;19:37. from acute respiratory distress syndrome: the LUNG SAFE
[130] Ichikado K, Muranaka H, Gushima Y, Kotani T, Nader HM, study. Intensive Care Med 2016;42:1865e76.
Fujimoto K, et al. Fibroproliferative changes on high- [146] Chen W, Janz DR, Shaver CM, Bernard GR, Bastarache JA,
resolution CT in the acute respiratory distress syndrome Ware LB. Clinical characteristics and outcomes are similar
predict mortality and ventilator dependency: a prospective in ARDS diagnosed by oxygen saturation/Fio2 ratio
observational cohort study. BMJ Open 2012;2:e000545. compared with PaO2/FiO2 ratio. Chest 2015;148:1477e83.
488 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

[147] Choi WI, Shehu E, Lim SY, Koh SO, Jeon K, Na S, et al. [162] Brambilla AM, Aliberti S, Prina E, Nicoli F, Del Forno M,
Markers of poor outcome in patients with acute hypoxemic Nava S, et al. Helmet CPAP vs. oxygen therapy in severe
respiratory failure. J Crit Care 2014;29:797e802. hypoxemic respiratory failure due to pneumonia. Intensive
rez-Me
[148] Villar J, Pe ndez L, Blanco J, An~o
 n JM, Blanch L, Belda J, Care Med 2014;40:942e9.
et al. A universal definition of ARDS: the PaO2/FiO2 ratio [163] Zhan Q, Sun B, Liang L, Yan X, Zhang L, Yang J, et al. Early
under a standard ventilatory setting–a prospective, use of noninvasive positive pressure ventilation for acute
multicenter validation study. Intensive Care Med lung injury: a multicenter randomized controlled trial. Crit
2013;39:583e92. Care Med 2012;40:455e60.
[149] Hernu R, Wallet F, Thiollie re F, Martin O, Richard JC, [164] Wermke M, Schiemanck S, Ho € ffken G, Ehninger G,
Schmitt Z, et al. An attempt to validate the modification of Bornha € user M, Illmer T. Respiratory failure in patients
the American-European consensus definition of acute lung undergoing allogeneic hematopoietic SCT–a randomized
injury/acute respiratory distress syndrome by the Berlin trial on early non-invasive ventilation based on standard
definition in a university hospital. Intensive Care Med care hematology wards. Bone Marrow Transplant
2013;39:2161e70. 2012;47:574e80.
[150] Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, [165] Squadrone V, Massaia M, Bruno B, Marmont F, Falda M,
Caldwell E, Fan E, et al. Acute respiratory distress Bagna C, et al. Early CPAP prevents evolution of acute lung
syndrome: the Berlin Definition. JAMA 2012;307:2526e33. injury in patients with hematologic malignancy. Intensive
[151] Cooke CR, Kahn JM, Caldwell E, Okamoto VN, Heckbert SR, Care Med 2010;36:1666e74.
Hudson LD, et al. Predictors of hospital mortality in a [166] Cosentini R, Brambilla AM, Aliberti S, Bignamini A, Nava S,
population-based cohort of patients with acute lung injury. Maffei A, et al. Helmet continuous positive airway pressure
Crit Care Med 2008;36:1412e20. vs oxygen therapy to improve oxygenation in community-
[152] Bhadade R, de'Souza R, Harde M, Asgaonkar D, acquired pneumonia: a randomized, controlled trial. Chest
Tuplondhe N. Mortality predictors of ARDS in medical 2010;138:114e20.
intensive care unit of a tertiary care Centre in a tropical [167] Ferrer M, Esquinas A, Leon M, Gonzalez G, Alarcon A,
country. J Assoc Physicians India 2015;63:16e22. Torres A. Noninvasive ventilation in severe hypoxemic
[153] Britos M, Smoot E, Liu KD, Thompson BT, Checkley W, respiratory failure: a randomized clinical trial. Am J Respir
Brower RG. The value of positive end-expiratory pressure Crit Care Med 2003;168:1438e44.
and Fio₂ criteria in the definition of the acute respiratory [168] Hilbert G, Gruson D, Vargas F, Valentino R, Gbikpi-
distress syndrome. Crit Care Med 2011;39:2025e30. Benissan G, Dupon M, et al. Noninvasive ventilation in
rez-Me
[154] Villar J, Pe ndez L, Lo pez J, Belda J, Blanco J, immunosuppressed patients with pulmonary infiltrates,
Saralegui I, et al. An early PEEP/FIO2 trial identifies different fever, and acute respiratory failure. N Engl J Med
degrees of lung injury in patients with acute respiratory 2001;344:481e7.
distress syndrome. Am J Respir Crit Care Med [169] Martin TJ, Hovis JD, Costantino JP, Bierman MI,
2007;176:795e804. Donahoe MP, Rogers RM, et al. A randomized, prospective
[155] He H, Sun B, Liang L, Li Y, Wang H, Wei L, et al. A evaluation of noninvasive ventilation for acute respiratory
multicenter RCT of noninvasive ventilation in pneumonia- failure. Am J Respir Crit Care Med 2000;161:807e13.
induced early mild acute respiratory distress syndrome. [170] Delclaux C, L'Her E, Alberti C, Mancebo J, Abroug F, Conti G,
Crit Care 2019;23:300. et al. Treatment of acute hypoxemic nonhypercapnic
[156] Azoulay E, Lemiale V, Mokart D, Nseir S, Argaud L, Pe ne F, respiratory insufficiency with continuous positive airway
et al. Effect of high-flow nasal oxygen vs standard oxygen pressure delivered by a face mask: a randomized controlled
on 28-day mortality in immunocompromised patients with trial. JAMA 2000;284:2352e60.
acute respiratory failure: the HIGH randomized clinical trial. [171] Antonelli M, Conti G, Bufi M, Costa MG, Lappa A, Rocco M,
JAMA 2018;320:2099e107. et al. Noninvasive ventilation for treatment of acute
[157] Belenguer-Muncharaz A, Cubedo-Bort M, Blasco-Asensio D, respiratory failure in patients undergoing solid organ
Mateu-Campos L, Vidal-Tegedor B, Madero-Perez J, et al. transplantation: a randomized trial. JAMA 2000;283:235e41.
Non-invasive ventilation versus invasive mechanical [172] Antonelli M, Conti G, Rocco M, Bufi M, De Blasi RA, Vivino G,
ventilation in patients with hypoxemic acute respiratory et al. A comparison of noninvasive positive-pressure
failure in an intensive care unit. A randomized controlled ventilation and conventional mechanical ventilation in
study. Minerva Pneumol 2017;56:1e10. patients with acute respiratory failure. N Engl J Med
[158] Lemiale V, Mokart D, Resche-Rigon M, Pe ne F, Mayaux J, 1998;339:429e35.
Faucher E, et al. Effect of noninvasive ventilation vs oxygen [173] Wysocki M, Tric L, Wolff MA, Millet H, Herman B.
therapy on mortality among immunocompromised patients Noninvasive pressure support ventilation in patients with
with acute respiratory failure: a randomized clinical trial. acute respiratory failure. A randomized comparison with
JAMA 2015;314:1711e9. conventional therapy. Chest 1995;107:761e8.
[159] Lemiale V, Mokart D, Mayaux J, Lambert J, Rabbat A, [174] Amato MB, Barbas CS, Medeiros DM, Magaldi RB,
Demoule A, et al. The effects of a 2-h trial of high-flow Schettino GP, Lorenzi-Filho G, et al. Effect of a protective-
oxygen by nasal cannula versus Venturi mask in ventilation strategy on mortality in the acute respiratory
immunocompromised patients with hypoxemic acute distress syndrome. N Engl J Med 1998;338:347e54.
respiratory failure: a multicenter randomized trial. Crit Care [175] Brochard L, Roudot-Thoraval F, Roupie E, Delclaux C,
2015;19:380. Chastre J, Fernandez-Monde jar E, et al. Tidal volume
[160] Frat JP, Thille AW, Mercat A, Girault C, Ragot S, Perbet S, reduction for prevention of ventilator-induced lung injury
et al. High-flow oxygen through nasal cannula in acute in acute respiratory distress syndrome. The Multicenter
hypoxemic respiratory failure. N Engl J Med Trail Group on Tidal Volume reduction in ARDS. Am J Respir
2015;372:2185e96. Crit Care Med 1998;158:1831e8.
[161] Bell N, Hutchinson CL, Green TC, Rogan E, Bein KJ, Dinh MM. [176] Stewart TE, Meade MO, Cook DJ, Granton JT, Hodder RV,
Randomised control trial of humidified high flow nasal Lapinsky SE, et al. Evaluation of a ventilation strategy to
cannulae versus standard oxygen in the emergency prevent barotrauma in patients at high risk for acute
department. Emerg Med Australas 2015;27:537e41. respiratory distress syndrome. Pressure- and Volume-
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 489

Limited Ventilation Strategy Group. N Engl J Med distress syndrome: a pilot, randomized controlled trial. Crit
1998;338:355e61. Care Med 2016;44:32e42.
[177] Wu G, Lu B. The application of low tidal volume pressure- [192] Cavalcanti AB, Suzumura EA,  Laranjeira LN, Cavalcanti AB,
controlled ventilation in patients with acute respiratory Suzumura EA,  Laranjeira LN, et al. Effect of Lung
distress syndrome. Hunan Yi Ke Da Xue Xue Bao Recruitment and Titrated, et al. Effect of lung recruitment
1998;23:57e8. and titrated positive end-expiratory pressure (PEEP) vs low
[178] Brower RG, Shanholtz CB, Fessler HE, Shade DM, White Jr P, PEEP on mortality in patients with acute respiratory distress
Wiener CM, et al. Prospective, randomized, controlled syndrome: a randomized clinical trial. JAMA
clinical trial comparing traditional versus reduced tidal 2017;318:1335e45.
volume ventilation in acute respiratory distress syndrome [193] Hodgson CL, Cooper DJ, Arabi Y, King V, Bersten A, Bihari S,
patients. Crit Care Med 1999;27:1492e8. et al. Maximal recruitment open lung ventilation in acute
[179] East TD, Heermann LK, Bradshaw RL, Lugo A, Sailors RM, respiratory distress syndrome (PHARLAP). A phase II,
Ershler L, et al. Efficacy of computerized decision support multicenter randomized controlled clinical trial. Am J
for mechanical ventilation: results of a prospective multi- Respir Crit Care Med 2019;200:1363e72.
center randomized trial. Proc AMIA Symp 1999:251e5. [194] Lam NN, Hung TD, Hung DK. Impact of "opening the lung"
[180] Ranieri VM, Suter PM, Tortorella C, De Tullio R, Dayer JM, ventilatory strategy on burn patients with acute respiratory
Brienza A, et al. Effect of mechanical ventilation on distress syndrome. Burns 2019;45:1841e7.
inflammatory mediators in patients with acute respiratory [195] Salem MS, Eltatawy HS, Abdelhafez AA, Alsherif SE-dI. Lung
distress syndrome: a randomized controlled trial. JAMA ultrasound- versus FiO2-guided PEEP in ARDS patients.
1999;282:54e61. Egypt J Anaesth 2020;36:31e7.
[181] Brower RG, Matthay MA, Morris A, Schoenfeld D, [196] Li J, Luo Z, Li X, Huang Z, Han J, Li Z, et al. Effect of different
Thompson BT, Wheeler A. Ventilation with lower tidal transpulmonary pressures guided mechanical ventilation
volumes as compared with traditional tidal volumes for on respiratory and hemodynamics of patients with ARDS: a
acute lung injury and the acute respiratory distress prospective randomized controlled trial. Zhonghua Wei
syndrome. N Engl J Med 2000;342:1301e8. Zhong Bing Ji Jiu Yi Xue 2017;29:39e44.
[182] Orme Jr J, Romney JS, Hopkins RO, Pope D, Chan KJ, [197] Ali AAE-R, El Wahsh RAE-R, Agha MAE-S, Tawadroos BB.
Thomsen G, et al. Pulmonary function and health-related Pressure regulated volume controlled ventilation versus
quality of life in survivors of acute respiratory synchronized intermittent mandatory ventilation in COPD
distress syndrome. Am J Respir Crit Care Med patients suffering from acute respiratory failure. Egypt J
2003;167:690e4. Chest Dis Tuberc 2016;65:121e5.
[183] Villar J, Kacmarek RM, Pe rez-Mendez L, Aguirre-Jaime A. A [198] Esteban A, Alı́a I, Gordo F, de Pablo R, Suarez J, Gonza  lez G,
high positive end-expiratory pressure, low tidal volume et al. Prospective randomized trial comparing pressure-
ventilatory strategy improves outcome in persistent acute controlled ventilation and volume-controlled ventilation in
respiratory distress syndrome: a randomized, controlled ARDS. For the Spanish Lung Failure Collaborative Group.
trial. Crit Care Med 2006;34:1311e8. Chest 2000;117:1690e6.
[184] Sun JJ, Yang MW, Wang CH, Li CH, Qin WH, Li YL, et al. [199] Rappaport SH, Shpiner R, Yoshihara G, Wright J, Chang P,
Clinical effects of low-stretch ventilation on acute Abraham E. Randomized, prospective trial of pressure-
respiratory distress syndrome. Zhongguo Wei Zhong Bing Ji limited versus volume-controlled ventilation in severe
Jiu Yi Xue 2009;21:609e12. respiratory failure. Crit Care Med 1994;22:22e32.
[185] Brower RG, Lanken PN, MacIntyre N, Matthay MA, Morris A, [200] Chacko B, Peter JV, Tharyan P, John G, Jeyaseelan L.
Ancukiewicz M, et al. Higher versus lower positive end- Pressure-controlled versus volume-controlled ventilation
expiratory pressures in patients with the acute respiratory for acute respiratory failure due to acute lung injury (ALI) or
distress syndrome. N Engl J Med 2004;351:327e36. acute respiratory distress syndrome (ARDS). Cochrane
[186] Long Y, Liu DW, Zhou X, Liu HZ, Guo ZJ, Huang H, et al. The Database Syst Rev 2015;1:Cd008807.
application of individualized ventilation strategies in acute [201] Li JQ, Li N, Han GJ, Pan CG, Zhang YH, Shi XZ, et al. Clinical
respiratory distress syndrome]. Zhonghua Jie He He Hu Xi research about airway pressure release ventilation for
Za Zhi 2006;29:549e53. moderate to severe acute respiratory distress syndrome.
[187] Meade MO, Cook DJ, Guyatt GH, Slutsky AS, Arabi YM, Eur Rev Med Pharmacol Sci 2016;20:2634e41.
Cooper DJ, et al. Ventilation strategy using low tidal [202] Maxwell RA, Green JM, Waldrop J, Dart BW, Smith PW,
volumes, recruitment maneuvers, and high positive end- Brooks D, et al. A randomized prospective trial of airway
expiratory pressure for acute lung injury and acute pressure release ventilation and low tidal volume
respiratory distress syndrome: a randomized controlled ventilation in adult trauma patients with acute respiratory
trial. JAMA 2008;299:637e45. failure. J Trauma 2010;69:501e11.
[188] Mercat A, Richard JC, Vielle B, Jaber S, Osman D, Diehl J-L, [203] Putensen C, Zech S, Wrigge H, Zinserling J, Stüber F, Von
et al. Positive end-expiratory pressure setting in adults with Spiegel T, et al. Long-term effects of spontaneous breathing
acute lung injury and acute respiratory distress syndrome: during ventilatory support in patients with acute lung
a randomized controlled trial. JAMA 2008;299:646e55. injury. Am J Respir Crit Care Med 2001;164:43e9.
[189] Talmor D, Sarge T, Malhotra A, O'Donnell CR, Ritz R, [204] Varpula T, Jousela I, Niemi R, Takkunen O, Pettila € V.
Lisbon A, et al. Mechanical ventilation guided by esophageal Combined effects of prone positioning and airway pressure
pressure in acute lung injury. N Engl J Med release ventilation on gas exchange in patients with acute
2008;359:2095e104. lung injury. Acta Anaesthesiol Scand 2003;47:516e24.
[190] Hodgson CL, Tuxen DV, Davies AR, Bailey MJ, Higgins AM, [205] Varpula T, Valta P, Niemi R, Takkunen O, Hynynen M,
Holland AE, et al. A randomised controlled trial of an open Pettila€ VV. Airway pressure release ventilation as a primary
lung strategy with staircase recruitment, titrated PEEP and ventilatory mode in acute respiratory distress syndrome.
targeted low airway pressures in patients with acute Acta Anaesthesiol Scand 2004;48:722e31.
respiratory distress syndrome. Crit Care 2011;15:R133. [206] Varpula T, Valta P, Markkola A, Pohjanen K, Halavaara J,
[191] Kacmarek RM, Villar J, Sulemanji D, Montiel R, Ferrando C, Hynynen M, et al. The effects of ventilatory mode on lung
Blanco J, et al. Open lung approach for the acute respiratory aeration assessed with computer tomography: a
490 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

randomized controlled study. J Intensive Care Med ventilation of identifying patients capable of breathing
2009;24:122e30. spontaneously. N Engl J Med 1996;335:1864e9.
[207] Zhou Y, Jin X, Lv Y, Wang P, Yang Y, Liang G, et al. Early [222] Fan L, Su Y, Elmadhoun OA, Zhang Y, Zhang Y, Gao D, et al.
application of airway pressure release ventilation may Protocol-directed weaning from mechanical ventilation in
reduce the duration of mechanical ventilation in acute neurological patients: a randomised controlled trial and
respiratory distress syndrome. Intensive Care Med subgroup analyses based on consciousness. Neurol Res
2017;43:1648e59. 2015;37:1006e14.
[208] Luo J, Wang MY, Liang BM, Yu H, Jiang FM, Wang T, et al. [223] Kollef MH, Shapiro SD, Silver P, St John RE, Prentice D,
Initial synchronized intermittent mandatory ventilation Sauer S, et al. A randomized, controlled trial of protocol-
versus assist/control ventilation in treatment of moderate directed versus physician-directed weaning from
acute respiratory distress syndrome: a prospective mechanical ventilation. Crit Care Med 1997;25:567e74.
randomized controlled trial. J Thorac Dis 2015;7:2262e73. [224] Krishnan JA, Moore D, Robeson C, Rand CS, Fessler HE. A
[209] Robinson BR, Blakeman TC, Toth P, Hanseman DJ, prospective, controlled trial of a protocol-based strategy to
Mueller E, Branson RD. Patient-ventilator asynchrony in a discontinue mechanical ventilation. Am J Respir Crit Care
traumatically injured population. Respir Care Med 2004;169:673e8.
2013;58:1847e55. [225] Marelich GP, Murin S, Battistella F, Inciardi J, Vierra T,
[210] Brochard L, Rauss A, Benito S, Conti G, Mancebo J, Rekik N, Roby M. Protocol weaning of mechanical ventilation in
et al. Comparison of three methods of gradual withdrawal medical and surgical patients by respiratory care
from ventilatory support during weaning from mechanical practitioners and nurses: effect on weaning time and
ventilation. Am J Respir Crit Care Med 1994;150:896e903. incidence of ventilator-associated pneumonia. Chest
[211] Esteban A, Frutos F, Tobin MJ, Alı́a I, Solsona JF, Valverdú I, 2000;118:459e67.
et al. A comparison of four methods of weaning patients [226] Namen AM, Ely EW, Tatter SB, Case LD, Lucia MA, Smith A,
from mechanical ventilation. Spanish Lung Failure et al. Predictors of successful extubation in neurosurgical
Collaborative Group. N Engl J Med 1995;332:345e50. patients. Am J Respir Crit Care Med 2001;163:658e64.
[212] Khan NA, Saleem M, Ashfaq A, Yusuf M. Is the lung [227] Navalesi P, Frigerio P, Moretti MP, Sommariva M, Vesconi S,
recruitment and titrated positive end expiratory pressure a Baiardi P, et al. Rate of reintubation in mechanically
better strategy as compare to low PEEP on mortality in ventilated neurosurgical and neurologic patients:
patients with acute respiratory distress syndrome. Med evaluation of a systematic approach to weaning and
Forum Mon 2018;29:93e7. extubation. Crit Care Med 2008;36:2986e92.
[213] Kung SC, Hung YL, Chen WL, Wang CM, Chang HC, Liu WL. [228] Roh JH, Synn A, Lim CM, Suh HJ, Hong S-B, Huh JW, et al. A
Effects of stepwise lung recruitment maneuvers in patients weaning protocol administered by critical care nurses for
with early acute respiratory distress syndrome: a the weaning of patients from mechanical ventilation. J Crit
prospective, randomized, controlled trial. J Clin Med Care 2012;27:549e55.
2019;8:231. [229] Rose L, Presneill JJ, Johnston L, Cade JF. A randomised,
[214] Xi XM, Jiang L, Zhu B. Clinical efficacy and safety of controlled trial of conventional versus automated weaning
recruitment maneuver in patients with acute respiratory from mechanical ventilation using SmartCare/PS. Intensive
distress syndrome using low tidal volume ventilation: a Care Med 2008;34:1788e95.
multicenter randomized controlled clinical trial. Chin Med J [230] Simeone F, Biagioli B, Scolletta S, Marullo ACM, Marchet-
(Engl). 2010;123:3100e5. Ti L, Caciorgna M, et al. Optimization of mechanical
[215] Chung FT, Lee CS, Lin SM, Kuo CH, Wang TY, Fang YF, et al. ventilation support following cardiac surgery. J Cardiovasc
Alveolar recruitment maneuver attenuates extravascular Surg (Torino) 2002;43:633e41.
lung water in acute respiratory distress syndrome. [231] Strickland Jr JH, Hasson JH. A computer-controlled
Medicine (Baltimore) 2017;96:e7627. ventilator weaning system. Chest 1991;100:1096e9.
[216] Huh JW, Jung H, Choi HS, Hong SB, Lim CM, Koh Y. Efficacy [232] Stahl C, Dahmen G, Ziegler A, Muhl E. Comparison of
of positive end-expiratory pressure titration after the automated protocol-based versus non-protocol-based
alveolar recruitment manoeuvre in patients with acute physician-directed weaning from mechanical ventilation.
respiratory distress syndrome. Crit Care 2009;13:R22. Intensivmed Notfallmed 2009;46:441e6.
[217] Yu S, Hu TX, Jin J, Zhang S. Effect of protective lung [233] Ferguson ND, Cook DJ, Guyatt GH, Mehta S, Hand L,
ventilation strategy combined with lung recruitment Austin P, et al. High-frequency oscillation in early acute
maneuver in patients with acute respiratory distress respiratory distress syndrome. N Engl J Med
syndrome (ARDS). J Acute Dis 2017;6:163e8. 2013;368:795e805.
[218] Constantin JM, Futier E, Cherprenet AL, Chanques G, [234] Bollen CW, van Well GT, Sherry T, Beale RJ, Shah S,
Guerin R, Cayot-Constantin S, et al. A recruitment Findlay G, et al. High frequency oscillatory ventilation
maneuver increases oxygenation after intubation of compared with conventional mechanical ventilation in
hypoxemic intensive care unit patients: a randomized adult respiratory distress syndrome: a randomized
controlled study. Crit Care 2010;14:R76. controlled trial [ISRCTN24242669]. Crit Care 2005;9:R430e9.
[219] Blackwood B, Alderdice F, Burns KE, Cardwell CR, Lavery G, [235] Derdak S, Mehta S, Stewart TE, Smith T, Rogers M,
O'Halloran P. Protocolized versus non-protocolized weaning Buchman TG, et al. High-frequency oscillatory ventilation
for reducing the duration of mechanical ventilation in for acute respiratory distress syndrome in adults: a
critically ill adult patients. Cochrane Database Syst Rev randomized, controlled trial. Am J Respir Crit Care Med
2010;5:Cd006904. 2002;166:801e8.
[220] Chaiwat O, Sarima N, Niyompanitpattana K, Komoltri C, [236] Young D, Lamb SE, Shah S, MacKenzie I, Tunnicliffe W,
Udomphorn Y, Kongsayreepong S. Protocol-directed vs. Lall R, et al. High-frequency oscillation for acute respiratory
physician-directed weaning from ventilator in intra- distress syndrome. N Engl J Med 2013;368:806e13.
abdominal surgical patients. J Med Assoc Thai [237] Mohamed SA-R, Mohamed NN. Efficacy and adverse events
2010;93:930e6. of early high-frequency oscillatory ventilation in adult burn
[221] Ely EW, Baker AM, Dunagan DP, Burke HL, Smith AC, patients with acute respiratory distress syndrome. Egypt J
Kelly PT, et al. Effect on the duration of mechanical Anaesth 2019;32:421e9.
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 491

[238] Chiu LC, Hu HC, Hung CY, Chang CH, Tsai FC, Yang CT, et al. care unit: the oxygen-ICU randomized clinical trial. JAMA
Dynamic driving pressure associated mortality in acute 2016;316:1583e9.
respiratory distress syndrome with extracorporeal [254] Mackle D, Bellomo R, Bailey M, Beasley R, Deane A,
membrane oxygenation. Ann Intensive Care 2017;7:12. Eastwood G, et al. Conservative oxygen therapy during
[239] Amato MB, Meade MO, Slutsky AS, Brochard L, Costa ELV, mechanical ventilation in the ICU. N Engl J Med
Schoenfeld DA, et al. Driving pressure and survival in the 2020;382:989e98.
acute respiratory distress syndrome. N Engl J Med [255] Panwar R, Hardie M, Bellomo R, Barrot L, Eastwood GM,
2015;372:747e55. Young PJ, et al. Conservative versus liberal oxygenation
[240] Burrell AJC, Lubnow M, Enger TB, Nanjayya VB, Philipp A, targets for mechanically ventilated patients. A pilot
Malfertheiner MV, et al. The impact of venovenous multicenter randomized controlled trial. Am J Respir Crit
extracorporeal membrane oxygenation on cytokine levels Care Med 2016;193:43e51.
in patients with severe acute respiratory distress syndrome: [256] Yang X, Shang Y, Yuan S. Low versus high pulse oxygen
a prospective, observational study. Crit Care Resusc saturation directed oxygen therapy in critically ill patients:
2017;19:37e44. a randomized controlled pilot study. J Thorac Dis
[241] Parhar KKS, Zjadewicz K, Soo A, Sutton A, Zjadewicz M, 2019;11:4234e40.
Doig L, et al. Epidemiology, mechanical power, and 3-year [257] Papazian L, Forel JM, Gacouin A, Penot-Ragon C, Perrin G,
outcomes in acute respiratory distress syndrome patients Loundou A, et al. Neuromuscular blockers in early acute
using standardized screening. An observational cohort respiratory distress syndrome. N Engl J Med
study. Ann Am Thorac Soc 2019;16:1263e72. 2010;363:1107e16.
[242] Villar J, Martı́n-Rodrı́guez C, Domı́nguez-Berrot AM, [258] Moss M, Huang DT, Brower RG, Ferguson ND, Ginde AA,
Ferna ndez L, Ferrando C, Soler JA, et al. A quantile analysis Gong MN, et al. Early neuromuscular blockade in the acute
of plateau and driving pressures: effects on mortality in respiratory distress syndrome. N Engl J Med
patients with acute respiratory distress syndrome receiving 2019;380:1997e2008.
lung-protective ventilation. Crit Care Med 2017;45:843e50. [259] Gainnier M, Roch A, Forel JM, Thirion X, Arnal J-M, Donati S,
[243] Toufen Junior C, De Santis Santiago RR, Hirota AS, et al. Effect of neuromuscular blocking agents on gas
Carvalho ARS, Gomes S, Amato MBP, et al. Driving pressure exchange in patients presenting with acute respiratory
and long-term outcomes in moderate/severe acute distress syndrome. Crit Care Med 2004;32:113e9.
respiratory distress syndrome. Ann Intensive Care [260] Forel JM, Roch A, Marin V, Michelet P, Demory D, Blache J-L,
2018;8:119. et al. Neuromuscular blocking agents decrease
[244] Fukumoto M. Theory and indications of fat emulsions. inflammatory response in patients presenting with acute
INTENSIVIST 2017;9:618e25. respiratory distress syndrome. Crit Care Med
[245] Guerin C, Papazian L, Reignier J, Ayzac L, Loundou A, 2006;34:2749e57.
Forel JM. Effect of driving pressure on mortality in ARDS [261] Guervilly C, Bisbal M, Forel JM, Mechati M, Lehingue S,
patients during lung protective mechanical ventilation in Bourenne J, et al. Effects of neuromuscular blockers on
two randomized controlled trials. Crit Care 2016;20:384. transpulmonary pressures in moderate to severe acute
[246] Liu L, Yang Y, Gao Z, Li M, Mu X, Ma X, et al. Practice of respiratory distress syndrome. Intensive Care Med
diagnosis and management of acute respiratory distress 2017;43:408e18.
syndrome in mainland China: a cross-sectional study. J [262] Beitler JR, Sarge T, Banner-Goodspeed VM, Gong MN,
Thorac Dis 2018;10:5394e404. Cook D, Novack V, et al. Effect of titrating positive
[247] Mekontso Dessap A, Boissier F, Charron C, Be got E, end-expiratory pressure (PEEP) with an esophageal
Repesse  X, Legras A, et al. Acute cor pulmonale during pressure-guided strategy vs an empirical high PEEP-FiO2
protective ventilation for acute respiratory distress strategy on death and days free from mechanical
syndrome: prevalence, predictors, and clinical impact. ventilation among patients with acute respiratory distress
Intensive Care Med 2016;42:862e70. syndrome: a randomized clinical trial. JAMA
[248] Morales-Quinteros L, Schultz MJ, Bringue  J, Calfee CS, 2019;321:846e57.
Camprubı́ M, Cremer OL, et al. Estimated dead space [263] Zhao Z, Chang MY, Chang MY, Gow CH, Zhang JH, Hsu YL,
fraction and the ventilatory ratio are associated with et al. Positive end-expiratory pressure titration with
mortality in early ARDS. Ann Intensive Care 2019;9:128. electrical impedance tomography and pressure-volume
[249] Chiumello D, Carlesso E, Brioni M, Cressoni M. Airway curve in severe acute respiratory distress syndrome. Ann
driving pressure and lung stress in ARDS patients. Crit Care Intensive Care 2019;9:7.
2016;20:276. [264] Guerin C, Reignier J, Richard JC, Beuret P, Gacouin A,
[250] Zhao X, Xiao H, Dai F, Brodie D, Meng L. Classification and Boulain T, et al. Prone positioning in severe acute
effectiveness of different oxygenation goals in respiratory distress syndrome. N Engl J Med
mechanically ventilated critically ill patients: network 2013;368:2159e68.
meta-analysis of randomised controlled trials. Eur Respir J [265] Gattinoni L, Tognoni G, Pesenti A, Taccone P, Mascheroni D,
2021;58:2002928. Labarta V, et al. Effect of prone positioning on the survival
[251] Asfar P, Schortgen F, Boisrame -Helms J, Charpentier J, of patients with acute respiratory failure. N Engl J Med
Guerot E, Megarbane B, et al. Hyperoxia and hypertonic 2001;345:568e73.
saline in patients with septic shock (HYPERS2S): a two-by- [266] Guerin C, Gaillard S, Lemasson S, Ayzac L, Girard R,
two factorial, multicentre, randomised, clinical trial. Lancet Beuret P, et al. Effects of systematic prone positioning in
Respir Med 2017;5:180e90. hypoxemic acute respiratory failure: a randomized
[252] Barrot L, Asfar P, Mauny F, Winiszewski H, Montini F, controlled trial. JAMA 2004;292:2379e87.
Badie J, et al. Liberal or conservative oxygen therapy for [267] Voggenreiter G, Aufmkolk M, Stiletto RJ, Baacke MG,
acute respiratory distress syndrome. N Engl J Med Waydhas C, Ose C, et al. Prone positioning improves
2020;382:999e1008. oxygenation in post-traumatic lung injury–a prospective
[253] Girardis M, Busani S, Damiani E, Donati A, Rinaldi L, randomized trial. J Trauma 2005;59:333e41.
Marudi A, et al. Effect of conservative vs conventional [268] Mancebo J, Ferna  ndez R, Blanch L, Rialp G, Gordo F,
oxygen therapy on mortality among patients in an intensive Ferrer M, et al. A multicenter trial of prolonged prone
492 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

ventilation in severe acute respiratory distress syndrome. critically ill patients anticipated requiring prolonged
Am J Respir Crit Care Med 2006;173:1233e9. mechanical ventilation. Chin Med J 2012;125:1925e30.
[269] Fernandez R, Trenchs X, Klamburg J, Castedo J, Serrano JM, [285] Young D, Harrison DA, Cuthbertson BH, Rowan K. Effect of
Besso G, et al. Prone positioning in acute respiratory early vs late tracheostomy placement on survival in
distress syndrome: a multicenter randomized clinical trial. patients receiving mechanical ventilation: the TracMan
Intensive Care Med 2008;34:1487e91. randomized trial. JAMA 2013;309:2121e9.
[270] Taccone P, Pesenti A, Latini R, Polli F, Vagginelli F, Mietto C, [286] Diaz-Prieto A, Mateu A, Gorriz M, Ortiga B, Truchero C,
et al. Prone positioning in patients with moderate and Sampietro N, et al. A randomized clinical trial for the timing
severe acute respiratory distress syndrome: a randomized of tracheotomy in critically ill patients: factors precluding
controlled trial. JAMA 2009;302:1977e84. inclusion in a single center study. Crit Care 2014;18:585.
[271] Combes A, Hajage D, Capellier G, Demoule A, Lavoue  S, [287] Dunham CM, Cutrona AF, Gruber BS, Calderon JE,
Guervilly C, et al. Extracorporeal membrane oxygenation for Ransom KJ, Flowers LL. Early tracheostomy in severe
severe acute respiratory distress syndrome. N Engl J Med traumatic brain injury: evidence for decreased mechanical
2018;378:1965e75. ventilation and increased hospital mortality. Int J Burns
[272] Peek GJ, Mugford M, Tiruvoipati R, Wilson A, Allen E, Trauma 2014;4:14e24.
Thalanany MM, et al. Efficacy and economic assessment of [288] Mohamed KAE, Mousa AY, ElSawy AS, Saleem AM. Early
conventional ventilatory support versus extracorporeal versus late percutaneous tracheostomy in critically ill adult
membrane oxygenation for severe adult respiratory failure mechanically ventilated patients. Egypt J Chest Dis Tuberc
(CESAR): a multicentre randomised controlled trial. Lancet 2014;63:443e8.
2009;374:1351e63. [289] Yadav S, Yadav G, Bharti AK, Shrivastav A, Verma RK.
[273] Cheung NH, Napolitano LM. Tracheostomy: epidemiology, Effects of early verses late percutaneous dilatational
indications, timing, technique, and outcomes. Respir Care tracheostomy on mechanically ventilated ICU patients. Res
2014;59:895e916. J Pharm Biol Chem Sci 2017;8:2078e82.
[274] Freeman BD, Morris PE. Tracheostomy practice in adults [290] American Thoracic Society; Infectious Diseases Society of
with acute respiratory failure. Crit Care Med America. Guidelines for the management of adults with
2012;40:2890e6. hospital-acquired, ventilator-associated, and healthcare-
[275] de Franca SA, Tavares WM, Salinet ASM, Paiva WS, associated pneumonia. Am J Respir Crit Care Med
Teixeira MJ. Early tracheostomy in severe traumatic brain 2005;171:388e416.
injury patients: a meta-analysis and comparison with late [291] Kudo D, Toyama M, Aoyagi T, Akahori Y, Yamamoto H,
tracheostomy. Crit Care Med 2020;48:e325e331. Ishii K, et al. Involvement of high mobility group box 1 and
[276] Sugerman HJ, Wolfe L, Pasquale MD, Rogers FB, O'Malley KF, the therapeutic effect of recombinant thrombomodulin in a
Knudson M, et al. Multicenter, randomized, prospective mouse model of severe acute respiratory distress
trial of early tracheostomy. J Trauma 1997;43:741e7. syndrome. Clin Exp Immunol 2013;173:276e87.
[277] Saffle JR, Morris SE, Edelman L. Early tracheostomy does not [292] Suzuki K, Okada H, Takemura G, Takada C, Tomita H,
improve outcome in burn patients. J Burn Care Rehabil Yano H, et al. Recombinant thrombomodulin protects
2002;23:431e8. against LPS-induced acute respiratory distress syndrome
[278] Bouderka MA, Fakhir B, Bouaggad A, Hmamouchi B, via preservation of pulmonary endothelial glycocalyx. Br J
Hamoudi D, Harti A. Early tracheostomy versus prolonged Pharmacol 2020;177:4021e33.
endotracheal intubation in severe head injury. J Trauma [293] Hayakawa S, Matsuzawa Y, Irie T, Rikitake H, Okada N,
2004;57:251e4. Suzuki Y. Efficacy of recombinant human soluble
[279] Rumbak MJ, Newton M, Truncale T, Schwartz SW, thrombomodulin for the treatment of acute exacerbation of
Adams JW, Hazard PB. A prospective, randomized, study idiopathic pulmonary fibrosis: a single arm, non-
comparing early percutaneous dilational tracheotomy to randomized prospective clinical trial. Multidiscip Respir
prolonged translaryngeal intubation (delayed tracheotomy) Med 2016;11:38.
in critically ill medical patients. Crit Care Med [294] Park KJ, Lee YJ, Oh YJ, Lee KS, Sheen SS, Hwang SC.
2004;32:1689e94. Combined effects of inhaled nitric oxide and a recruitment
[280] Blot F, Similowski T, Trouillet JL, Chardon P, Korach J-M, maneuver in patients with acute respiratory distress
Costa M-A, et al. Early tracheotomy versus prolonged syndrome. Yonsei Med J 2003;44:219e26.
endotracheal intubation in unselected severely ill ICU [295] Taylor RW, Zimmerman JL, Dellinger RP, Straube RC,
patients. Intensive Care Med 2008;34:1779e87. Criner GJ, Davis Jr K, et al. Low-dose inhaled nitric oxide in
[281] Terragni PP, Antonelli M, Fumagalli R, Faggiano C, patients with acute lung injury: a randomized controlled
Berardino M, Pallavicini FB, et al. Early vs late tracheotomy trial. JAMA 2004;291:1603e9.
for prevention of pneumonia in mechanically ventilated [296] Gerlach H, Keh D, Semmerow A, Busch T, Lewandowski K,
adult ICU patients: a randomized controlled trial. JAMA Pappert DM, et al. Dose-response characteristics during
2010;303:1483e9. long-term inhalation of nitric oxide in patients with severe
[282] Trouillet JL, Luyt CE, Guiguet M, Ouattara A, Vaissier E, acute respiratory distress syndrome: a prospective,
Makri R, et al. Early percutaneous tracheotomy versus randomized, controlled study. Am J Respir Crit Care Med
prolonged intubation of mechanically ventilated patients 2003;167:1008e15.
after cardiac surgery: a randomized trial. Ann Intern Med [297] Troncy E, Collet JP, Shapiro S, Guimond JG, Blair L,
2011;154:373e83. Ducruet T, et al. Inhaled nitric oxide in acute respiratory
[283] Koch T, Hecker B, Hecker A, Brenck F, Preuß M, distress syndrome: a pilot randomized controlled study. Am
Schmelzer T, et al. Early tracheostomy decreases J Respir Crit Care Med 1998;157:1483e8.
ventilation time but has no impact on mortality of intensive [298] Dellinger RP, Zimmerman JL, Taylor RW, Straube RC,
care patients: a randomized study. Langenbecks Arch Surg Hauser DL, Criner GJ, et al. Effects of inhaled nitric oxide in
2012;397:1001e8. patients with acute respiratory distress syndrome: results
[284] Zheng Y, Sui F, Chen XK, Zhang GC, Wang XW, Zhao S, et al. of a randomized phase II trial. Inhaled Nitric Oxide in ARDS
Early versus late percutaneous dilational tracheostomy in Study Group. Crit Care Med 1998;26:15e23.
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 493

[299] Lundin S, Mang H, Smithies M, Stenqvist O, Frostell C. respiratory distress syndrome: a multicentre, randomised
Inhalation of nitric oxide in acute lung injury: results of a controlled trial. Lancet Respir Med 2020;8:267e76.
European multicentre study. The European Study Group of [317] Herridge MS, Tansey CM, Matte  A, Tomlinson G, Diaz-
Inhaled Nitric Oxide. Intensive Care Med 1999;25:911e9. Granados N, Cooper A, et al. Functional disability 5 years
[300] Michael JR, Barton RG, Saffle JR, Mone M, Markewitz BA, after acute respiratory distress syndrome. N Engl J Med
Hillier K, et al. Inhaled nitric oxide versus conventional 2011;364:1293e304.
therapy: effect on oxygenation in ARDS. Am J Respir Crit [318] Tipping CJ, Harrold M, Holland A, Romero L, Nisbet T,
Care Med 1998;157:1372e80. Hodgson CL. The effects of active mobilisation and
[301] Cuthbertson BH, Galley HF, Webster NR. Effect of inhaled rehabilitation in ICU on mortality and function: a
nitric oxide on key mediators of the inflammatory response systematic review. Intensive Care Med 2017;43:171e83.
in patients with acute lung injury. Crit Care Med [319] Doiron KA, Hoffmann TC, Beller EM. Early intervention
2000;28:1736e41. (mobilization or active exercise) for critically ill adults in the
[302] Donnelly SC, MacGregor I, Zamani A, Gordon MW, intensive care unit. Cochrane Database Syst Rev
Robertson CE, Steedman DJ, et al. Plasma elastase 2018;3:CD010754.
levels and the development of the adult respiratory [320] Abu-Khaber HA, Abouelela AMZ, Abdelkarim EM. Effect of
distress syndrome. Am J Respir Crit Care Med electrical muscle stimulation on prevention of ICU acquired
1995;151:1428e33. muscle weakness and facilitating weaning from
[303] Moraes TJ, Chow C-W, Downey GP. Proteases and lung mechanical ventilation. Alexandria J Med 2013;49:309e15.
injury. Crit Care Med 2003;31:S189e94. [321] Dong Z-H, Yu B-X, Sun Y-B, Fang W, Li L. Effects of early
[304] Kadoi Y, Hinohara H, Kunimoto F, Saito S, Goto F, Kosaka T, rehabilitation therapy on patients with mechanical
et al. Pilot study of the effects of ONO-5046 in patients with ventilation. World J Emerg Med 2014;5:48e52.
acute respiratory distress syndrome. Anesth Analg [322] Fossat G, Baudin F, Courtes L, Bobet S, Dupont A,
2004;99:872e7. Bretagnol A, et al. Effect of in-bed leg cycling and electrical
[305] Nakayama H. Evaluation of the usefulness of civelestat stimulation of the quadriceps on global muscle strength in
sodium in patients with ALI/ARDS under NPPV (In Japanese) critically ill adults: a randomized clinical trial. JAMA
Prog Med 2013;33:2223e7. 2018;320:368e78.
[306] Sato N. Treatment of ALI/ARDS caused by pneumonia with [323] Morris PE, Berry MJ, Files DC, Thompson JC, Hauser J,
civelestat sodium (In Japanese) Prog Med 2008;28:437e9. Flores L, et al. Standardized rehabilitation and hospital
[307] Zeiher BG, Artigas A, Vincent J-L, Dmitrienko A, Jackson K, length of stay among patients with acute respiratory
Thompson BT, et al. Neutrophil elastase inhibition in acute failure: a randomized clinical trial. JAMA
lung injury: results of the STRIVE study. Crit Care Med 2016;315:2694e702.
2004;32:1695e702. [324] Dos Santos FV, Cipriano Jr G, Vieira L, Güntzel Chiappa AM,
[308] Ryugo M, Sawa Y, Takano H, Matsumiya G, Iwai S, Ono M, Cipriano GBF, Vieira P, et al. Neuromuscular electrical
et al. Effect of a polymorphonuclear elastase inhibitor stimulation combined with exercise decreases duration of
(sivelestat sodium) on acute lung injury after mechanical ventilation in ICU patients: a randomized
cardiopulmonary bypass: findings of a double-blind controlled trial. Physiother Theory Pract 2020;36:580e8.
randomized study. Surg Today 2006;36:321e6. [325] Schweickert WD, Pohlman MC, Pohlman AS, Nigos C,
[309] Pelosi P, D'Onofrio D, Chiumello D, Paolo S, Chiara G, Pawlik AJ, Esbrook CL, et al. Early physical and occupational
Capelozzi VL, et al. Pulmonary and extrapulmonary acute therapy in mechanically ventilated, critically ill patients: a
respiratory distress syndrome are different. Eur Respir J randomised controlled trial. Lancet 2009;373:1874e82.
2003;42:48se56s. [326] Yu L, Jiang J-X, Zhang Y, Chen Y-Z, Shi Y. Use of in-bed
[310] Ruan SY, Lin HH, Huang CT, Kuo PH, Wu HD, Yu CJ. cycling combined with passive joint activity in acute
Exploring the heterogeneity of effects of corticosteroids on respiratory failure patients receiving mechanical
acute respiratory distress syndrome: a systematic review ventilation. Ann Palliat Med 2020;9:175e81.
and meta-analysis. Crit Care 2014;18:R63. [327] Treggiari MM, Romand JA, Yanez ND, Deem SA, Goldberg J,
[311] Bernard GR, Luce JM, Sprung CL, Rinaldo JE, Tate RM, Hudson L, et al. Randomized trial of light versus deep
Sibbald WJ, et al. High-dose corticosteroids in patients with sedation on mental health after critical illness. Crit Care
the adult respiratory distress syndrome. N Engl J Med Med 2009;37:2527e34.
1987;317:1565e70. [328] Strøm T, Martinussen T, Toft P. A protocol of no sedation for
[312] Meduri GU, Headley AS, Golden E, Carson SJ, Umberger RA, critically ill patients receiving mechanical ventilation: a
Kelso T, et al. Effect of prolonged methylprednisolone randomised trial. Lancet 2010;375:475e80.
therapy in unresolving acute respiratory distress syndrome: [329] SRLF Trial Group. Impact of oversedation prevention in
a randomized controlled trial. JAMA 1998;280:159e65. ventilated critically ill patients: a randomized trial-the
[313] Meduri GU, Golden E, Freire AX, Taylor E, Zaman M, AWARE study. Ann Intensive Care 2018;8:93.
Carson SJ, et al. Methylprednisolone infusion in early severe [330] Ware LB, Matthay MA. The acute respiratory distress
ARDS: results of a randomized controlled trial. Chest syndrome. N Engl J Med 2000;342:1334e49.
2007;131:954e63. [331] Han F, Sun R, Ni Y, Hu X, Chen X, Jiang L, et al. Early
[314] Steinberg KP, Hudson LD, Goodman RB, Hough CL, initiation of continuous renal replacement therapy
Lanken PN, Hyzy R, et al. Efficacy and safety of improves clinical outcomes in patients with acute
corticosteroids for persistent acute respiratory distress respiratory distress syndrome. Am J Med Sci
syndrome. N Engl J Med 2006;354:1671e84. 2015;349:199e205.
[315] Tongyoo S, Permpikul C, Mongkolpun W, Vattanavanit V, [332] Meng JB, Lai ZZ, Xu XJ, Ji CL, Hu MH, Zhang G. Effects of
Udompanturak SU, Kocak M, et al. Hydrocortisone early continuous venovenous hemofiltration on E-selectin,
treatment in early sepsis-associated acute respiratory hemodynamic stability, and ventilatory function in patients
distress syndrome: results of a randomized controlled trial. with septic-shock-induced acute respiratory distress
Crit Care 2016;20:329. syndrome. Biomed Res Int 2016;2016:7463130.
[316] Villar J, Ferrando C, Martı́nez D, Ambro  s A, Mun
~ oz T, [333] Martin GS, Mangialardi RJ, Wheeler AP, Dupont WD,
Soler JA, et al. Dexamethasone treatment for the acute Morris JA, Bernard GR. Albumin and furosemide therapy in
494 r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5

hypoproteinemic patients with acute lung injury. Crit Care [349] Cam BV, Tuan DT, Fonsmark L, Poulsen A, Tien NM,
Med 2002;30:2175e82. Tuan HM, et al. Randomized comparison of oxygen mask
[334] Martin GS, Moss M, Wheeler AP, Mealer M, Morris JA, treatment vs. nasal continuous positive airway pressure in
Bernard GR. A randomized, controlled trial of furosemide dengue shock syndrome with acute respiratory failure. J
with or without albumin in hypoproteinemic patients with Trop Pediatr 2002;48:335e9.
acute lung injury. Crit Care Med 2005;33:1681e7. [350] Chisti MJ, Salam MA, Smith JH, Ahmed T, Pietroni MAC,
[335] Phillips C. Hemodynamics and extravascular lung water in Shahunja KM, et al. Bubble continuous positive airway
acute lung injury: a prospective randomized controlled pressure for children with severe pneumonia and
multicentered trial of goal directed treatment of EVLW hypoxaemia in Bangladesh: an open, randomised
versus standard management for the treatment of acute controlled trial. Lancet 2015;386:1057e65.
lung injury. Available at: https://www.clinicaltrials.gov/ct2/ [351] Peters MJ, Agbeko R, Davis P, Klein N, Zenasni Z, Jones A,
show/NCT00624650. et al. Randomized study of early continuous positive
[336] Wiedemann HP, Wheeler AP, Bernard GR, Thompson BT, airways pressure in acute respiratory failure in children
Hayden D, deBoisblanc B, et al. Comparison of two fluid- with impaired immunity (SCARF) ISRCTN82853500. Pediatr
management strategies in acute lung injury. N Engl J Med Crit Care Med 2018;19:939e48.
2006;354:2564e75. [352] Yanez LJ, Yunge M, Emilfork M, Lapadula M, Alca  ntara A,
[337] Mikkelsen ME, Christie JD, Lanken PN, Biester RC, Ferna  ndez C, et al. A prospective, randomized, controlled
Thompson BT, Bellamy SL, et al. The adult respiratory trial of noninvasive ventilation in pediatric acute
distress syndrome cognitive outcomes study: long-term respiratory failure. Pediatr Crit Care Med 2008;9:484e9.
neuropsychological function in survivors of acute lung [353] McCollum ED, Mvalo T, Eckerle M, Smith AG, Kondowe D,
injury. Am J Respir Crit Care Med 2012;185:1307e15. Makonokaya D, et al. Bubble continuous positive airway
[338] Chang Y-F, Hou Y-C, Pai M-H, Yeh S-L, Liu J-J. Effects of u-3 pressure for children with high-risk conditions and severe
polyunsaturated fatty acids on the homeostasis of CD4þ T pneumonia in Malawi: an open label, randomised,
cells and lung injury in mice with polymicrobial sepsis. J controlled trial. Lancet Respir Med 2019;7:964e74.
Parenter Enteral Nutr 2017;41:805e14. [354] Pediatric Acute Lung Injury Consensus Conference Group.
[339] Elamin EM, Miller AC, Ziad S. Immune enteral nutrition can Pediatric acute respiratory distress syndrome: consensus
improve outcomes in medical-surgical patients with ARDS: recommendations from the Pediatric Acute Lung Injury
a prospective randomized controlled trial. J Nutr Disord Consensus Conference. Pediatr Crit Care Med
Ther 2012;2:109. 2015;16:428e39.
[340] Rice TW, Wheeler AP, Thompson BT, deBoisblanc BP, [355] Marini JJ, Rocco PRM, Gattinoni L. Static and dynamic
Steingrub J, Rock P, et al. Enteral omega-3 fatty acid, contributors to ventilator-induced lung injury in clinical
gamma-linolenic acid, and antioxidant supplementation in practice. Pressure, energy, and power. Am J Respir Crit Care
acute lung injury. JAMA 2011;306:1574e81. Med 2020;201:767e74.
[341] Gadek JE, DeMichele SJ, Karlstad MD, Pacht ER, Donahoe M, [356] Fan E, Brodie D, Slutsky AS. Acute respiratory distress
Albertson TE, et al. Effect of enteral feeding with syndrome: advances in diagnosis and treatment. JAMA
eicosapentaenoic acid, gamma-linolenic acid, and 2018;319:698e710.
antioxidants in patients with acute respiratory distress [357] Nieman GF, Satalin J, Andrews P, Aiash H, Habashi NM,
syndrome. Enteral Nutrition in ARDS Study Group. Crit Care Gatto LA. Personalizing mechanical ventilation according to
Med 1999;27:1409e20. physiologic parameters to stabilize alveoli and minimize
[342] Parish M, Valiyi F, Hamishehkar H, Sanaie S, Jafarabadi MA, ventilator induced lung injury (VILI). Intensive Care Med
Golzari SE, et al. The effect of omega-3 fatty acids on ARDS: Exp 2017;5:8.
a randomized double-blind study. Adv Pharm Bull [358] Kneyber MCJ, de Luca D, Calderini E, Jarreau P-H,
2014;4:555e61. Javouhey E, Lopez-Herce J, et al. Recommendations for
[343] Stapleton RD, Martin TR, Weiss NS, Crowley JJ, Gundel SJ, mechanical ventilation of critically ill children from the
Nathens AB, et al. A phase II randomized placebo- Paediatric Mechanical Ventilation Consensus Conference
controlled trial of omega-3 fatty acids for the treatment of (PEMVECC). Intensive Care Med 2017;43:1764e80.
acute lung injury. Crit Care Med 2011;39:1655e62. [359] Melsen WG, Rovers MM, Groenwold RHH, Bergmans DCJJ,
[344] Sapru A, Flori H, Quasney MW, Dahmer MK. Pediatric acute Camus C, Bauer TT, et al. Attributable mortality of
lung injury consensus conference group. Pathobiology of ventilator-associated pneumonia: a meta-analysis of
acute respiratory distress syndrome. Pediatr Crit Care Med individual patient data from randomised prevention
2015;16:S6e22. studies. Lancet Infect Dis 2013;13:655e71.
[345] Khemani RG, Smith LS, Zimmerman JJ, Erickson S, Pediatric [360] Foronda FK, Troster EJ, Farias JA, Barbas CS, Ferraro AA,
Acute Lung Injury Consensus Conference Group. Pediatric Faria LS, et al. The impact of daily evaluation and
acute respiratory distress syndrome: definition, incidence, spontaneous breathing test on the duration of pediatric
and epidemiology: proceedings from the Pediatric Acute mechanical ventilation: a randomized controlled trial. Crit
Lung Injury Consensus Conference. Pediatr Crit Care Med Care Med 2011;39:2526e33.
2015;16:S23e40. [361] Schultz TR, Lin RJ, Watzman HM, Durning SM, Hales R,
[346] Khemani RG, Smith L, Lopez-Fernandez YM, Kwok J, Morzov R, Woodson A, et al. Weaning children from mechanical
Klein MJ, et al. Paediatric acute respiratory distress syndrome ventilation: a prospective randomized trial of protocol-
incidence and epidemiology (PARDIE): an international, directed versus physician-directed weaning. Respir Care
observational study. Lancet Respir Med 2019;7:115e28. 2001;46:772e82.
[347] Gupta S, Sankar J, Lodha R, Kabra SK. Comparison of [362] El-Nawawy A, Moustafa A, Heshmat H, Abouahmed A. High
prevalence and outcomes of pediatric acute respiratory frequency oscillatory ventilation versus conventional
distress syndrome using pediatric acute lung injury mechanical ventilation in pediatric acute respiratory
consensus conference criteria and Berlin definition. Front distress syndrome: a randomized controlled study. Turk J
Pediatr 2018;6:93. Pediatr 2017;59:130e43.
[348] ARDS Definition Task Force. Acute respiratory distress ~
[363] Arnold JH, Hanson JH, Toro-Figuero LO, GutiA©rrez J,
syndrome: the Berlin Definition. JAMA 2012;307:2526e33. Berens RJ, Anglin DL. Prospective, randomized comparison
r e s p i r a t o r y i n v e s t i g a t i o n 6 0 ( 2 0 2 2 ) 4 4 6 e4 9 5 495

of high-frequency oscillatory ventilation and conventional [379] Luchetti M, Ferrero F, Gallini C, Natale A, Pigna A,
mechanical ventilation in pediatric respiratory failure. Crit Tortorolo L, et al. Multicenter, randomized, controlled study
Care Med 1994;22:1530e9. of porcine surfactant in severe respiratory syncytial virus-
[364] Samransamruajkit R, Prapphal N, Deelodegenavong J, induced respiratory failure. Pediatr Crit Care Med
Poovorawan Y. Plasma soluble intercellular adhesion 2002;3:261e8.
molecule-1 (sICAM-1) in pediatric ARDS during high [380] Mo€ ller JC, Schaible T, Roll C, Schiffmann J-H, Bindl L,
frequency oscillatory ventilation: a predictor of mortality. Schrod L, et al. Treatment with bovine surfactant in severe
Asian Pac J Allergy Immunol 2005;23:181e8. acute respiratory distress syndrome in children: a
[365] Samransamruajkit R, Rassameehirun C, Pongsanon K, randomized multicenter study. Intensive Care Med
Huntrakul S, Deerojanawong J, Sritippayawan S, et al. A 2003;29:437e46.
comparison of clinical efficacy between high frequency [381] Rodriguez-Moya VS, Gallo-Borrero CM, Santos-Areas D,
oscillatory ventilation and conventional ventilation with Prince-Martinez IA, Diaz-Casanas E, Lopez-Herce Cid J.
lung volume recruitment in pediatric acute respiratory Exogenous surfactant and alveolar recruitment in the
distress syndrome: a randomized controlled trial. Indian J treatment of the acute respiratory distress syndrome. Clin
Crit Care Med 2016;20:72e7. Respir J 2017;11:1032e9.
[366] Facchin F, Fan E. Airway pressure release ventilation and [382] Rodriguez-Moya VS, Del Carmen Machado-Lubian M,
high-frequency oscillatory ventilation: potential strategies 
Barrese-Perez Y, Avila-Albuerne Y, Uranga-Pin ~ a R, Blanco-
to treat severe hypoxemia and prevent ventilator-induced Hidalgo O, et al. Cuban exogenous pulmonary surfactant in
lung injury. Respir Care 2015;60:1509e21. treatment of pediatric acute respiratory distress syndrome.
[367] Ganesan SL, Jayashree M, Singhi SC, Bansal A. Airway MEDICC Rev 2017;19:24.
pressure release ventilation in pediatric acute respiratory [383] Thomas NJ, Guardia CG, Moya FR, Cheifetz IM, Markovitz B,
distress syndrome: a randomized controlled trial. Am J Cruces P, et al. A pilot, randomized, controlled clinical trial
Respir Crit Care Med 2018;198:1199e207. of lucinactant, a peptide-containing synthetic surfactant, in
[368] Walters DM, Cho H-Y, Kleeberger SR. Oxidative stress and infants with acute hypoxemic respiratory failure. Pediatr
antioxidants in the pathogenesis of pulmonary fibrosis: a Crit Care Med 2012;13:646e53.
potential role for Nrf2. Antioxid Redox Signal [384] Thomas NJ, Spear D, Wasserman E, Pon S, Markovitz B,
2008;10:321e32. Singh AR, et al. CALIPSO: a randomized controlled trial of
[369] Hraiech S, Yoshida T, Annane D, Duggal A, Fanelli V, calfactant for acute lung injury in pediatric stem cell and
Gacouin A, et al. Myorelaxants in ARDS patients. Intensive oncology patients. Biol Blood Marrow Transplant
Care Med 2020;46:2357e72. 2018;24:2479e86.
[370] Claude Gue rin C, Albert RK, Beitler J, Gattinoni L, Jaber S, [385] Willson DF, Zaritsky A, Bauman LA, Dockery K, James RL,
Marini JJ, et al. Prone position in ARDS patients: why, when, Conrad D, et al. Instillation of calf lung surfactant extract
how and for whom. Intensive Care Med 2020;46:2385e96. (calfactant) is beneficial in pediatric acute hypoxemic
[371] Munshi L, Del Sorbo L, Adhikari NKJ, Hodgson CL, respiratory failure. Members of the Mid-Atlantic Pediatric
Wunsch H, Meade MO, et al. Prone position for Critical Care Network. Crit Care Med 1999;27:188e95.
acute respiratory distress syndrome. A systematic [386] Willson DF, Thomas NJ, Markovitz BP, Bauman LA,
review and meta-analysis. Ann Am Thorac Soc DiCarlo JV, Pon S, et al. Effect of exogenous surfactant
2017;14:S280e8. (calfactant) in pediatric acute lung injury: a randomized
[372] Robak O, Schellongowski P, Bojic A, Laczika K, Locker GJ, controlled trial. JAMA 2005;293:470e6.
Staudinger T. Short-term effects of combining upright and [387] Willson DF, Thomas NJ, Tamburro R, Truemper E, Truwit J,
prone positions in patients with ARDS: a prospective Conaway M, et al. Pediatric calfactant in acute respiratory
randomized study. Crit Care 2011;15:R230. distress syndrome trial. Pediatr Crit Care Med
[373] Curley MA, Hibberd PL, Fineman LD, Wypij D, Shih MC, 2013;14:657e65.
Thompson JE, et al. Effect of prone positioning on clinical [388] Drago BB, Kimura D, Rovnaghi CR, Schwingshackl A,
outcomes in children with acute lung injury: a randomized Rayburn M, Meduri GU, et al. Double-blind, placebo-
controlled trial. JAMA 2005;294:229e37. controlled pilot randomized trial of methylprednisolone
[374] Thompson BT, Chambers RC, Liu KD. Acute respiratory infusion in pediatric acute respiratory distress syndrome.
distress syndrome. N Engl J Med 2017;377:562e72. Pediatr Crit Care Med 2015;16:e74e81.
[375] Westphal K, Strouhal U, Byhahn C, Hommel K, Behne M. [389] Curley MA, Wypij D, Watson RS, Grant MJ, Asaro LA,
Inhalation of nitric oxide in severe lung failure. Cheifetz IM, et al. Protocolized sedation vs usual care in
Anaesthesiol Reanim 1998;23:144e8. pediatric patients mechanically ventilated for acute
[376] Bronicki RA, Fortenberry J, Schreiber M, Checchia PA, respiratory failure: a randomized clinical trial. JAMA
Anas NG. Multicenter randomized controlled trial of 2015;313:379e89.
inhaled nitric oxide for pediatric acute respiratory distress [390] Gupta K, Gupta VK, Jayashree M, Singhi S. Randomized
syndrome. J Pediatr 2015;166:365e9. controlled trial of interrupted versus continuous sedative
[377] Ream RS, Hauver JF, Lynch RE, Kountzman B, Gale GB, infusions in ventilated children. Pediatr Crit Care Med
Mink RB. Low-dose inhaled nitric oxide improves the 2012;13:131e5.
oxygenation and ventilation of infants and children with [391] Vet NJ, de Wildt SN, Verlaat CWM, Knibbe CAJ, Mooij MG,
acute, hypoxemic respiratory failure. Crit Care Med van Woensel JBM, et al. A randomized controlled trial of
1999;27:989e96. daily sedation interruption in critically ill children.
[378] Dobyns EL, Cornfield DN, Anas NG, Fortenberry JD, Intensive Care Med 2016;42:233e44.
Tasker RC, Lynch A, et al. Multicenter randomized [392] Verlaat CWM, Heesen GP, Vet NJ, de Hoog M, van der
controlled trial of the effects of inhaled nitric oxide therapy Hoeven JG, Kox M, et al. Randomized controlled trial of daily
on gas exchange in children with acute hypoxemic interruption of sedatives in critically ill children. Paediatr
respiratory failure. J Pediatr 1999;134:406e12. Anaesth 2014;24:151e6.

You might also like