You are on page 1of 271

Applied Technologies in Pulmonary Medicine

This page is intentionally left blank


Applied Technologies in

Pulmonary
Medicine
Editor

Antonio M. Esquinas Murcia

56 figures, 9 in color and 29 tables, 2011

Basel · Freiburg · Paris · London · New York · Bangalore ·


Bangkok · Shanghai · Singapore · Tokyo · Sydney
This page is intentionally left blank
‘To Rosario, my wife, inspiration and love for all.’
Antonio M. Esquinas
Antonio M. Esquinas
Intensive Care Unit
Hospital Morales Meseguer
Murcia, Spain

Library of Congress Cataloging-in-Publication Data

Applied technologies in pulmonary medicine / editor, Antonio M. Esquinas.


p. ; cm.
Includes bibliographical references and indexes.
ISBN 978-3-8055-9584-1 (hard cover: alk. paper)  ISBN 978-3-8055-9585-8 (e-ISBN)
1. Respiratory therapy  Technological innovations. 2. Respirators (Medical equipment)
I. Esquinas, Antonio M.
[DNLM: 1. Pulmonary Medicine  Instrumentation. 2. Pulmonary Medicine – methods. 3. Biomedical Technology.
4. Respiratory Physiological Phenomena. 5. Respiratory Tract Diseases. 6. Technology, Medical  instrumentation. WF 100]
RC735.I5A67 2011
615.8'36  dc22
2010036628

Bibliographic Indices. This publication is listed in bibliographic services.


Disclaimer. The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not
of the publisher and the editor(s). The appearance of advertisements in the book is not a warranty, endorsement, or approval of the products or
services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or
property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.
Drug Dosage. The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord
with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations,
and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for
any change in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a
new and/or infrequently employed drug.
All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means electronic
or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing
from the publisher.
© Copyright 2011 by S. Karger AG, P.O. Box, CH–4009 Basel (Switzerland)
www.karger.com
Printed in Switzerland on acid-free and non-aging paper (ISO 9706) by Reinhardt Druck, Basel
ISBN 978–3–8055–9584–1
e–ISBN 978–3–8055–9585–8
Contents

XIII Preface
Esquinas, A.M. (Murcia)

Applied Technologies in Mechanical Ventilation

1 Proportional Assist Ventilation and Neurally Adjusted Ventilatory Assist


Beck, J.; Sinderby, C. (Toronto, Ont.)
6 Time-Adaptive Mode: A New Ventilation Form for the Treatment of
Respiratory Insufficiency
Dellweg, D.; Barchfeld, T.; Kerl, J.; Koehler, D. (Schmallenberg)
10 Influence of Ventilation Strategies on Hemodynamics in
Hypovolemic Shock
Herff, H. (Innsbruck)
15 Gas Exchange during Perfluorocarbon Liquid Immersion
Davies, M.W.; Dunster, K.R. (Brisbane, Qld.)
19 Extracorporeal Membrane Oxygenation for Respiratory and Heart Failure in Adults
Herlihy, J.P.; Loyalka, P.; Connolly, T.; Kar, B.; Gregoric, I. (Houston, Tex.)
28 Automatic Control of Mechanical Ventilation Technologies
Tehrani, F.T. (Fullerton, Calif.)

Weaning Mechanical Ventilation

35 Corticosteroids to Prevent Post-Extubation Upper Airway Obstruction


Epstein, S.K. (Boston, Mass.)
39 FLEX: A New Weaning and Decision Support System
Tehrani, F.T. (Fullerton, Calif.)

VII
Applied Technologies in Specific Clinical Situations

Technology in Sleep Pulmonary Disorders

46 Thermal Infrared Imaging during Polysomnography: Has the Time Come to Unwire the
‘Wired’ Subjects?
Murthy, J.N.; Pavlidis, I. (Houston, Tex.)

Technology in Cardiopulmonary Resuscitation

51 Cardiopulmonary Resuscitation with the Boussignac System


Boussignac, G. (Antony)
53 Respiratory Function Monitoring during Simulation-Based Mannequin Teaching
Schmölzer, G.M. (Melbourne, Vic./Graz); Morley, C.J. (Melbourne, Vic.)

Technology in Inhalation Therapy

60 Technological Requirements for Inhalation of Biomolecule Aerosols


Siekmeier, R.; Scheuch, G. (Gemünden)
67 Problems and Examples of Biomolecule Inhalation for Systemic Treatment
Siekmeier, R.; Scheuch, G. (Gemünden)
77 Diabetes Treatment by Inhalation of Insulin – Shine and Decline of a
Novel Type of Therapy
Siekmeier, R.; Scheuch, G. (Gemünden)

Technology in Diagnosis and Pulmonary Problems

84 Endobronchial Ultrasound
Anantham, D.; Siyue, M.K. (Singapore)

Technology in Anesthesiology

89 Minimally Invasive Thoracic Surgery for Pulmonary Resections


Salati, M.; Rocco, G. (Naples)
96 Acute Lung Collapse during Open-Heart Surgery
Neema, P.K.; Manikandan, S.; Rathod, R.C. (Trivandrum)
102 Anesthesia in the Intraoperative MRI Environment
Bergese, S.D.; Puente, E.G. (Columbus, Ohio)
107 Awake Thoracic Epidural Anesthesia Pulmonary Resections
Pompeo, E.; Tacconi, F.; Mineo, T.C. (Rome)

VIII Contents
Technology in Transplants

114 Preservation of Organs from Brain-Dead Donors with Hyperbaric Oxygen


Bayrakci, B. (Ankara)

Technology and Monitoring

119 Teleassistance in Chronic Respiratory Failure Patients


Vitacca, M. (Lumezzane)
126 Capnography: Gradient PACO2 and PETCO2
Donnellan, M.E. (Needham, Mass.)

Technology and Equipment in Transport

132 Problems of Air Travel for Patients with Lung Disease


Robson, A.G. (Edinburgh)

Respiratory Technology in Abdominal Syndromes

136 Intra-Abdominal Hypertension and Abdominal Compartment Syndrome:


Measuring Techniques and the Effects on Lung Mechanics
De Keulenaer, B.L. (Fremantle, W.A.)

Critical Care Problems

Applied Technologies in Pulmonary Diagnosis

145 Pleural Effusions in Critically III Patients


Papaioannou, V.; Pneumatikos, I. (Alexandroupolis)

Infections – Ventilator-Associated Pneumonia

151 Gravitational Force and Respiratory Colonization in Mechanical Ventilation


Aly, H. (Washington, D.C.); Berra, L. (Boston, Mass.); Kolobow, T. (Bethesda, Md.)
156 Use of Gram Stain or Preliminary Bronchoalveolar Lavage Culture Results to
Guide Discontinuation of Empiric Antibiotics in Ventilator-Associated Pneumonia
Swanson, J.M.; Wood, G.C. (Memphis, Tenn.)
163 Patterns of Resolution in Ventilator-Associated Pneumonia
Myrianthefs, P.M.; Evagelopoulou, P.; Baltopoulos, G.J. (Athens)

Contents IX
168 Viral Infections in the Intensive Care Unit
Luyt, C.E. (Paris)
172 Healthcare-Associated Pneumonia among Hospitalized Patients
Shindo, Y.; Hasegawa, Y. (Nagoya)

Oncology and Pulmonary Complications

178 Critical Care Outcome of Lung Cancer Patients


Adam, A.K.; Soubani, A.O. (Detroit, Mich.)

Prognosis and Readmission

185 Readmission to the Intensive Care Unit for Patients with Lung Edema or Atelectasis
Matsuoka, Y.; Zaitsu, A.; Hashizume, M. (Fukuoka)

Pulmonary Rehabilitation and Technology

192 Early Mobilization in the Intensive Care Unit: Safety, Feasibility, and Benefits
Korupolu, R.; Gifford, J.M.; Zanni, J.M.; Truong, A.; Vajrala, G.; Lepre, S.; Needham, D.M. (Baltimore, Md.)
197 Evidence-Based Guidelines in Pulmonary Rehabilitation
Ries, A.L. (San Diego, Calif.)
202 Ward Mortality in Patients Discharged from the ICU with Tracheostomy
Fernandez, R.F. (Manresa)

Pulmonary Medicine in Pediatrics and Neonatology Critical Care

205 Exogenous Surfactant in Respiratory Distress Syndrome


Calkovska, A. (Martin); Herting, E. (Lübeck)
210 Hypercapnia and Hypocapnia in Neonates
Tao, L.; Zhou, W. (Guangzhou)

Respiratory Health Problems and Environment

217 Air Pollution and Health


Carvalho-Oliveira, R.; Nakagawa, N.K.; Saldiva, P.H.N. (São Paulo)
223 Air Pollution and Non-Invasive Respiratory Assessments
Nakagawa, N.K.; Nakao, M.; Goto, D.M.; Saraiva-Romanholo, B.M. (São Paulo)
231 Air Pollution, Oxidative Stress and Pulmonary Defense
Macchione, M.; Bueno Garcia, M.L. (São Paulo)

X Contents
Organization in Pulmonary Medicine

238 Mechanical Ventilation in Disaster Management


Branson, R.; Blakeman, T.C.; Rodriquez, D. (Cincinnati, Ohio)
246 Postoperative Intensive and Intermediate Care?
Weissman, C. (Jerusalem)

250 Author Index


251 Subject Index

Contents XI
This page is intentionally left blank
Preface

Technological innovations in the treatment of Further topics that the readers will find in this
respiratory diseases involve a critical vision of book include the outcome of patients with lung
all aspects from basic physics, pathophysiology, cancer admitted to the ICU, new results on pul-
diagnosis and treatments, to clinical experience. monary rehabilitation and technologies, evidence-
Applied Technologies in Pulmonary Medicine is based guidelines and the basics on discharge from
an updated selection of the most current original the ICU, how to optimize the problems in pedi-
articles published on new technological advan- atric and neonatal critical care telemonitoring and
ces in the diagnosis and treatment of respiratory assistance in chronic respiratory failure and cap-
problems. The analytical methodology is a very nography innovations, the newest options for dia-
original aspect of this book in comparison to gnosis of pulmonary diseases (polysomnography,
other textbooks on respiratory medicine, where ultrasound), technology in emergency medicine
invited authors critically present their results and such as cardiopulmonary resuscitation, and new
the clinical implications of their findings. options in inhalation therapies (macromolecules
The analysis includes basic areas such as pul- such as insulin).
monary and critical care medicine, mechanical Recently, two new major topics have gained
ventilation, ventilator modes (extracorporeal the interest of all specialists engaged in the field
membrane oxygenation, time-adaptive modes, of pulmonary medicine and related technologies:
proportional assist ventilation, automatic con- firstly the diagnosis of health respiratory problems
trol mechanical ventilation, etc.), new pharma- and the environment, and secondly new concepts
cological treatments during mechanical ven- of organizational issues in global disaster manage-
tilation (weaning options and post-extubation ment and the role of mechanical ventilation, gui-
failure), the basics of pathophysiology, treat- delines and options.
ment and how to prevent ventilator-associated The major topics in Applied Technologies in
pneumonia (new antibiotics, viral infections and Pulmonary Medicine and their clinical implica-
healthcare-associated pneumonia). We have also tions have involved hard and meticulous work. It
included original advances in technologies that presents a novel approach to help clinicians ea-
are applied in anesthesiology and postoperative sily understand the technologies provided in the
critical care (minimally invasive thoracic surge- numerous papers. We hope that the reader and
ry, open-heart surgery, intraoperative and pul- younger researchers will acquire practical ideas
monary resections) and in the preservation of when carrying out their laboratory and clinical
organs. trials on a daily basis.

XIII
I would like to thank all the authors as well as Siempos, MD, Athens, Greece; Giovanni Vento,
the following collaborators: Penny Andrews, BSN, MD, Rome, Italy, and M.Terese Verklan, PhD,
RN, Baltimore, Md., USA; Melissa Brown, RRT- CCNS, RNC, Houston, Tex., USA. Their efforts
NPS, San Diego, Calif., USA; Andrea Calkovska, to reach these objectives are greatly appreciated. I
MD, PhD, Martin, Slovakia; Ettore Capoluongo, personally believe that the knowledge and ‘appli-
MD, Rome, Italy; Bart L. De Keulenaer, MD, cation of technologies in pulmonary medicine’ will
FJFICM, East Fremantle, W.A., Australia; become a continuous dynamic process of ideas
Emmanuel Douzinas, MD, Athens, Greece; Lothar and experiences of trial and error, where the fi-
Engelmann, MD, Leipzig, Germany; J. Pat Herlihy, nal conclusions can be drawn once they become
MD, Houston, Tex., USA; Pavlos M. Myrianthefs, routine.
MD, PhD, Kifissia/Athens, Greece; Naomi Kondo Antonio M. Esquinas
Nakagawa, BSc, PhD, São Paulo, Brazil; Catherine Murcia, Spain
S. Sassoon, MD, Long Beach, Calif., USA; Ilias I.

XIV Preface
Applied Technologies in Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 1–5

Proportional Assist Ventilation and Neurally


Adjusted Ventilatory Assist
Jennifer Becka ⭈ Christer Sinderbyb
a
Department of Pediatrics, University of Toronto, and bKeenan Research Centre, Li Ka Shing Knowledge Institute and
Department of Critical Care Medicine, St. Michael’s Hospital, Toronto, Ont., Canada

Abbreviations This article describes the concepts related to PAV


EAdi Electrical activity of the diaphragm
and NAVA, their similarities and their differences,
NAVA Neurally adjusted ventilatory assist and the recent physiological studies. For a more
PAV Proportional assist ventilation detailed review of this topic, the reader is referred
to Sinderby and Beck [3].

Historically, patients in need of mechanical venti- Patient-Ventilator Interaction


lation were often heavily sedated or paralyzed and
placed on time-cycled modes of ventilation. There Ideally, a mechanical ventilator should behave
is now clear evidence showing that reduced seda- as a respiratory prosthesis, providing air in tan-
tion and spontaneous breathing improve patient dem with the patient’s breathing in terms of tim-
outcome in terms of days on ventilation and mor- ing and the magnitude of the inspiration. The
tality [1]. Under these conditions (less sedation prevalence of patient-ventilator asynchrony has
and more spontaneous breathing), unless the pa- recently been revealed [2] and it is now read-
tient ‘entrains’ themselves to the rate of the breath ily accepted that during conventional ventila-
delivery, time-cycled modes may not be the most tion, such as pressure support ventilation, poor
appropriate, especially in light of the recent work patient-ventilator asynchrony does occur [4,
demonstrating that patient-ventilator asynchrony 5]. Patient-ventilator asynchrony ranges at its
increases the duration of mechanical ventilation worst from completely missed patient efforts
and mortality [2]. (so-called wasted efforts) and auto-triggering in
Two new modes of mechanical ventilation are the absence of spontaneous efforts, to delays in
now available on the market that can synchro- ventilator triggering and cycling-off. Modes with
nize not only the timing, but also the level of as- fixed levels of assist (such as assist control, pres-
sist to the patient’s own effort, PAV and NAVA. sure control, and pressure support ventilation)
Proportional Assist Ventilation
Brain (respiratory centers) Step 1
Phrenic nerve and neuromuscular transmission Step 2
In PAV [9], the ventilator generates airway pres-
Diaphragm activation Step 3
Diaphragm contraction Step 4
sure in proportion to instantaneous flow and vol-
Lung expansion Step 5 ume (Step 6). The flow assist, which is a percent-
Airway flow and volume Step 6 age of the airway resistance, dictates how much
airway pressure is delivered per unit of flow. The
Fig. 1. Simplified chain of events involved with spon- volume assist, which is a percent of the pulmo-
taneous breathing, beginning with central respiratory nary elastance, dictates how much pressure is de-
drive (Step 1), to the final ventilatory output at the air- livered per unit of volume. The degree of assist
way (Step 6).
can range to provide unloading between 0 and
100%. During PAV, knowledge about respirato-
ry system mechanics and endotracheal tube re-
sistance are required. This is especially important
may also be asynchronous since the ventilator in preterm neonates as there is a large breath-to-
cannot respond to changes in patient demand breath variability in resistance and compliance of
on a breath-by-breath basis. Patient-ventilator the respiratory system. Recently, ‘PAV+’, with up-
asynchrony has now been shown to affect pa- dated measurements of resistance and elastance
tient outcome in terms of prolonged weaning and implementation of load-adjustable gain fac-
[6], poorer sleep quality [7], longer duration tors, has the potential to account for this.
of mechanical ventilation and tracheotomy [2], With regard to physiological studies [for a re-
and in infants, higher incidence of pneumotho- view, see 3], PAV has been shown to improve pa-
rax [8]. tient comfort, improve patient-ventilator inter-
action, improve sleep quality, and allows greater
variability in breathing pattern (i.e. more physi-
From Brain to Breath: Spontaneous Breathing ological) in comparison with pressure support.
In both adults and neonates, PAV has been dem-
A simplified schematic of the chain of events that onstrated to unload the respiratory muscles, with
occur during spontaneous breathing is presented lower mean airway pressures than pressure sup-
in figure 1. The signal for spontaneous breathing port ventilation – with similar clinical short-term
originates in the respiratory centers (Step 1) and outcomes (gas exchange and hemodynamics).
in the case of the diaphragm – the most important Recently, PAV with load-adjustable gain factors
muscle of respiration – travels down the phrenic has been shown to be feasible in critically ill pa-
nerves, then passes through the neuromuscular tients, and to require fewer interventions with re-
junction (Step 2) to activate the diaphragm elec- spect to sedation and ventilator settings [10] com-
trically (Step 3). It is only after this step of electri- pared to pressure support.
cal activation of the diaphragm, that cross-bridge
cycling is initiated and the muscle contracts (Step
4). Contraction of the diaphragm results in lung Neurally Adjusted Ventilatory Assist
expansion (Step 5), resulting in flow and volume
at the airway (Step 6). Depending on the type and NAVA uses the EAdi (Step 3) – a signal represen-
severity of the disease, the final output of airflow tative of the output from the respiratory centers
and volume at the airway may not represent the – to control both the timing and the magnitude
true neural respiratory output. of delivered pressure [11]. The EAdi is obtained

2 Beck ⭈ Sinderby
by an array of miniaturized sensors placed on a with excessively leaky non-invasive interfaces
conventional nasogastric (or orogastric) feed- does not affect patient-ventilator synchrony.
ing tube. The electrode array is positioned in
the esophagus at the level of the gastroesopha-
geal junction, where the spontaneous activity Discussion
of the crural diaphragm is sensed. Standardized
signal processing algorithms automatically take The similarities and differences between PAV and
into account diaphragm displacement, motion NAVA can only be discussed theoretically as there
artifacts, filtering of the electrocardiogram, and are no studies in the literature comparing these
cross-talk from other active muscles [12]. The two modes of ventilation. The lack of a single de-
processed signal, known as the EAdi waveform, vice providing both modes of ventilation is likely
can be characterized by its amplitude on inspira- the responsible factor.
tion (phasic EAdi) and expiration (tonic EAdi) as In principle, NAVA and PAV are similar in
well as its timing (neural inspiratory time, neural that they are both modes of assisted ventilation
expiratory time, neural respiratory rate). When where the applied airway pressure is servo-con-
compared to the airway pressure waveform in trolled continuously throughout spontaneous
other modes of ventilation, the EAdi provides inspiration, changing in proportion to the pa-
information about patient-ventilator synchrony. tient’s breathing effort and allowing the patient
In the absence of the EAdi signal (and the cath- to control the extent and timing of lung inflation.
eter position has been deemed appropriate), it is During both NAVA and PAV, the amplification
an indication of central apnea, or suppression of ‘gain’ between patient effort and delivered pres-
spontaneous breathing activity. Hence, the EAdi sure can be adjusted, in order to achieve more
signal has monitoring capabilities as well as con- or less unloading of the respiratory muscles. This
trolling the ventilator. is very different from modes of ventilation that
In infant and adult patients, NAVA has been are volume- or pressure-targeted, where fixed lev-
shown to significantly improve patient-ventilator els of assist are delivered independent of patient
interaction compared to conventional modes of effort.
assist [4, 5, 13], in terms of both improved tim- Both PAV and NAVA require that the patient
ing and proportionality. Neural triggering and is spontaneously breathing. However, it should
cycling-off on non-invasive (helmet) ventilation be noted that NAVA uses the neural output signal
in healthy adults has demonstrated that this im- (EAdi), whereas PAV has no monitoring capabili-
proved synchrony improves comfort [14]. ties for quantifying respiratory drive. This means
During NAVA, the assist levels are adjusted by that, similar to other patient-triggered modes of
changing the proportionality between the EAdi ventilation, a back-up mode of ventilation is re-
and delivered pressure (the so-called ‘NAVA lev- quired in the case of central apnea. As well, upper
el’). Stepwise increases in the NAVA level cause a pressure limits should be adjusted accordingly,
gradual reduction in respiratory drive, and there- in the case of large and central respiratory drive.
fore the expected increase in pressure is not nec- The fact that PAV and NAVA require some de-
essarily achieved. Due to this physiological down- gree of spontaneous breathing may actually be a
regulation of the EAdi signal, airway pressure and clinical advantage, in that the respiratory muscles
tidal volume ‘plateau’ at adequate levels of un- are encouraged to be used during partial ventila-
loading [15]. tor assist. Inactivity of respiratory muscles dur-
Since the EAdi controller signal for NAVA is ing mechanical ventilation (due to too high lev-
pneumatically independent, application of NAVA els of sedation or too high levels of assist) has a

Proportional Assist Ventilation and Neurally Adjusted Ventilatory Assist 3


negative impact on diaphragm muscle fiber in- flow and volume (Step 6). In the case of dynamic
tegrity and prolongs the duration of mechanical hyperinflation, if the respiratory drive stays the
ventilation [16]. same (i.e. same EAdi), the flow and volume will
Unlike pressure support ventilation, increas- be lower, and the controller signal for PAV may
ing levels of assist with PAV and NAVA have little reduce the airway pressure delivery.
effect on respiratory rate and tidal volume when
unloading is sufficient. In modes of ventilation
that allow the patient the freedom to control the Conclusion
rate and depth of inspiration, it seems that there
is a desired minute ventilation, rate and volume. PAV and NAVA are both modes of partial venti-
When unloading is adequate to satisfy the patient’s lator assist delivering assist in proportion to pa-
demand, if the assist is increased during PAV or tient effort. During NAVA, the diaphragm elec-
NAVA, patient effort decreases and therefore, so trical activity – a true signal of neural respiratory
does the amount of assist. output – is the controller signal for delivered ven-
The major differences between these two tilation. During PAV, a surrogate measurement
modes lie in how the disease processes affect the of respiratory drive is used to control the venti-
controller signals. During NAVA, the EAdi (the lator. The inherent benefits of these two modes
neural respiratory drive to the diaphragm from lie in the fact that these modes require sponta-
the respiratory centers, Step 3 in figure 1) is the neous breathing and offer synchronized delivery
controller signal. PAV uses airway flow and vol- of assist.
ume (Step 6), which is a surrogate measurement
of respiratory drive, and further down the chain of
events involved with spontaneous breathing. Recommendations
In the presence of a leak – for example in in-
fants with leaks around the endotracheal tube, or • Implement spontaneous mode of ventilation
during non-invasive ventilation – the flow and as soon as possible/tolerable.
volume signal in Step 6 will be misinterpreted • Ensure that respiratory drive is not suppressed
as patient flow and volume. For triggering and by too high levels of sedation or too high
delivering proportional assist during PAV, the levels of assist, i.e. ensure that patients are
leak may auto-cycle the ventilator and may call spontaneously breathing.
for increased flow delivery during inspiration. • Optimize patient-ventilator synchrony.
In sharp contrast, NAVA, using a neural trigger,
is not affected by leaks for obtaining synchrony.
Depending on the size of the leak, an increase in
the NAVA level however may be required to un-
load the respiratory muscle sufficiently.
The major difference between NAVA and PAV
might be observed in the case of dynamic hy-
perinflation, where shortening of the respiratory
muscles affects the force output for a given neural
activation. In fact, any disease process affecting
the contractile properties of the diaphragm (Step
4) will in theory cause an ‘uncoupling’ between
neural respiratory drive (Steps 1–3) and patient

4 Beck ⭈ Sinderby
References
1 Girard TD, Kress JP, Fuchs BD, et al: Effi- 6 Chao DC, Scheinhorn DJ, Stearn-Has- 12 Aldrich T, Sinderby C, McKenzie D, et al:
cacy and safety of a paired sedation and senpflug M: Patient-ventilator trigger Electrophysiologic techniques for the
ventilator weaning protocol for mechan- asynchrony in prolonged mechanical assessment of respiratory muscle func-
ically ventilated patients in intensive ventilation. Chest 1997:112:1592–1599. tion; in ATS/ERS Statement on Respira-
care (Awakening and Breathing Con- 7 Bosma K, Ferreyra G, Ambrogio C, et al: tory Muscle Testing. Am J Respir Crit
trolled trial): a randomised controlled Patient-ventilator interaction and sleep Care Med 2002;166:518–624.
trial. Lancet 2008;371:126–134. in mechanically ventilated patients: 13 Beck J, Reilly M, Grasselli G, et al:
2 Thille AW, Rodriguez P, Cabello B, et al: pressure support versus proportional Patient-ventilator interaction during
Patient-ventilator asynchrony during assist ventilation. Crit Care Med 2007;35: neurally adjusted ventilatory assist in
assisted mechanical ventilation. Inten- 1048–1054. low birth weight infants. Pediatr Res
sive Care Med 2006;32:1515–1522. 8 Greenough A, Wood S, Morley CJ, Davis 2009;65:663–668.
3 Sinderby C, Beck J: Proportional assist JA: Pancuronium prevents pneumotho- 14 Moerer O, Beck J, Brander L, et al: Sub-
ventilation and neurally adjusted venti- races in ventilated premature babies ject-ventilator synchrony during neural
latory assist – better approaches to who actively expire against positive pres- versus pneumatically triggered non-
patient-ventilator synchrony? Clin Chest sure inflation. Lancet 1984;1:1–3. invasive helmet ventilation. Intensive
Med 2008;29:329–342. 9 Younes M: Proportional assist ventila- Care Med 2008;34:1615–1623.
4 Colombo D, Cammarota G, Bergamaschi tion, a new approach to ventilatory sup- 15 Brander L, Leong-Poi H, Beck J, et al:
V, et al: Physiologic response to varying port. Theory. Am Rev Respir Dis 1992; Titration and implementation of neu-
levels of pressure support and neurally 145:114–120. rally adjusted ventilatory assist in criti-
adjusted ventilatory assist in patients 10 Xirouchaki N, Kondili E, Vaporidi K, et cally ill patients. Chest 2009;135:695–
with acute respiratory failure. Intensive al: Proportional assist ventilation with 703.
Care Med 2008;34:2010–2018. load-adjustable gain factors in critically 16 Levine S, Nguyen T, Taylor N, et al:
5 Spahija J, De Marchie M, Albert M, et al: ill patients: comparison with pressure Rapid disuse atrophy of diaphragm
Patient-ventilator interaction during support. Intensive Care Med 2008;34: fibers in mechanically ventilated
pressure support ventilation and neu- 2026–2034. humans. N Engl J Med 2008;358:1327–
rally adjusted ventilatory assist. Crit 11 Sinderby C, Navalesi P, Beck J, et al: Neu- 1335.
Care Med 2010;38:518–526. ral control of mechanical ventilation in
respiratory failure. Nat Med 1999;5:
1433–1436.

Jennifer Beck, PhD


Keenan Research Centre, Li Ka Shing Knowledge Institute
St. Michael’s Hospital, 30 Bond Street, Queen Wing 4-072
Toronto, ON M5B1W8 (Canada)
Tel. +1 416 880 3664, E-Mail beckj@smh.toronto.on.ca

Proportional Assist Ventilation and Neurally Adjusted Ventilatory Assist 5


Section Title
Applied Technologies in Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 6–9

Time-Adaptive Mode: A New Ventilation Form for


the Treatment of Respiratory Insufficiency
D. Dellweg ⭈ T. Barchfeld ⭈ J. Kerl ⭈ D. Koehler
Pneumonology 1, Kloster Grafschaft, Schmallenberg, Germany

Abbreviations produce a linear or curvilinear flow profile during


AC Assist control
inspiration that might not match the patient’s own
NIV Non-invasive ventilation inspiratory flow profile. This may cause discom-
S Spontaneous fort and increase patient-ventilator asynchrony.
ST Spontaneous-timed In view of these problems, Weinmann GmbH
TA Timed automated (Hamburg, Germany) introduced a new mode of
NIV (TA incorporated into the Ventilogic© venti-
lator) that automatically captures the patient’s own
NIV has gained substantial importance and is flow profile and adjusts ventilation with preselect-
considered standard medical care for hypercap- ed pressure levels in a controlled fashion [6].
nic respiratory failure of different etiologies [1].
It also has a proven benefit for certain forms of
hypoxemic respiratory failure [2]. The main ratio- Description of the TA Algorithm
nale of NIV is the unloading effect on the respira-
tory muscles during ventilation [3]. This results Inspiratory and expiratory pressures must be se-
in reduction of dyspnea, increased mobility and lected on the ventilator-interactive display prior
better quality of life for the patient. NIV is usu- to ventilation. The operator has also to select the
ally applied using a S, ST or AC mode. In either type of underlying airway disease (R = restrictive,
mode, patients have to trigger the ventilator un- O = obstructive, N = normal) and set upper and
less the programmed backup rate (ST mode) ex- lower limits to the respiratory rate by selecting a
ceeds the patient’s respiratory rate. The work that target rate and a range of allowance. The patient
is necessary to trigger the ventilator can be sub- must be connected (via mask) to the ventilator
stantial and might be as high as 39% of the total prior to activation of the mode.
work of breathing [4]. Controlled NIV is feasible Once activated, TA-mode ventilation begins
[5] and might decrease the respiratory workload, with an analysis phase. During this phase, a con-
eliminating the need to trigger the ventilator. tinuous pressure of 4 hPa is delivered by the venti-
Controlled NIV however is not frequently used lator turbine to guarantee effective carbon dioxide
and might potentially increase patient-ventilator washout through the whisper valve of the mask in-
asynchrony or mismatch. Non-invasive ventilators terface. During this phase, the ventilator analyzes
85

70 P1 (hPa)
Run-in period of autoadaptive controlled ventilation

60
50
40
30
20
10
0
–10
–20
–30
–40
–50
–60 Flow (/s) Poes (hPa)
–70
–80
0 200 400 600 800 1,000 1,200 1,400 1,600 1,800 2,000 2,200 2,400 2,600 2,800 3,000 3,200
Seconds × 10–2

Fig. 1. Pressure and flow tracings during the run-in period of autoadaptive controlled ventilation (TA mode). According
to the measured flow profile of spontaneous breathing, the ventilator slowly increases inspiratory pressure (PI) dur-
ing the run-in period, resulting in increased inspiratory flow (Flow) and raised esophageal pressure (Poes), a marker of
respiratory muscle unloading.

the patient’s own flow profile by integration of flow P(t) represents the pressure integral, flow and
and time. Once the ventilator senses a stable profile volume arise from averaged flow pattern data from
(time and flow measurements within a predefined the analysis phase. The selection R = restrictive, O
range of allowance), the ventilator will increase in- = obstructive and N = normal allocates distinct
spiratory pressure over five consecutive breaths in constant numbers for resistance (R given in hPa/
steps of 60–70–80–90–100% of preset value (fig. (l/s)) and compliance (C given in ml/hPa) into the
1). During the inspiratory phase, inspiratory pres- equation. The system software calculates P(t) ac-
sure will be adjusted over inspiratory time in or- cording to the individual preselected maximal in-
der to obtain a flow profile matching the patient’s spiratory pressures.
own pattern. One has to note that the preselected Inspiratory time refers to the average inspira-
inspiratory pressure will only be achieved during tory time recorded during the analysis phase. We
peak inspiratory flow, and that the pressure level selected a broad range of the respiratory rate to
during inspiration will be adjusted to mimic the allow each individual to achieve his or her natu-
previously detected flow profile. ral respiratory rate. The target rate range selection
The inspiratory pressure curve is calculated can be used to prevent an inept and non-physio-
according to the following motion equation: logical breathing pattern.
Inspiratory to expiratory time ratio (I:E ratio)
P(t) = R × flow + 1/C × volume is determined by the subject’s I:E ratio measured

TA Mode: A New Form of Self-Adjusting Controlled Ventilation 7


0.7

0.6

0.5

0.4
Vt ()

0.3

0.2

Ventilation
Analysis

0.1

0
1 5 9 13 17 21 25 29 33 37 41 45 49 53 57 61 65 69 73 77 81 85 89 93
Breath

Fig. 2. Onset of ventilation (red line) increases tidal volume (Vt) initially, since patient effort is supported by gas de-
livery from the ventilator. While the latter is constant, patient effort decreases as indicated by the subsequent de-
crease in Vt.

during the analysis phase. At the end of the run-in Clinical Implication of TA-Mode NIV
period, ventilator gas delivery will increase (Vt) as
long as the patient has not decreased his or her re- TA-mode ventilation has been compared to
spiratory effort. Patient effort will then consecu- S-mode ventilation in healthy individuals and
tively decrease as a sign of respiratory muscle rest achieved a higher degree of respiratory muscle
and decreasing oxygen cost of breathing (fig. 2). unloading (see fig. 3) [7]. It therefore represents
TA mode does not allow for additional trig- a promising mode to better unload the respira-
gered breaths, however if the ventilator senses tory muscles in patients who require NIV. Clinical
subject-ventilator asynchrony, and it will reana- studies in patients are being currently conduct-
lyze the patient’s flow pattern. Asynchrony is de- ed but have not been published to date. From our
fined by inspiratory and/or expiratory fighting in experience, TA-mode ventilation is well tolerated
four consecutive breaths. Inspiratory fighting is and effective in the majority of patients.
defined by a flow reduction of at least 20 l/min be- Patients with a markedly irregular breathing
low the mean inspiratory flow inside the middle pattern during sleep might experience recurrent
60% of the inspiratory time (between TI 20 and TI phases of breathing pattern reanalysis if fighting
80). Expiratory fighting is defined by the presence criteria are fulfilled. This might cause sleep dis-
of flow rise of 10 l/min above leak-compensation turbances and can compromise compliance and
inside the middle 40% of the expiratory time (be- practicability of this type of ventilation. According
tween TE 30 and TE 70). to our personal experience, the latter applies only

8 Dellweg ⭈ Barchfeld ⭈ Kerl ⭈ Koehler


their usual quiet and normal breathing pattern
p < 0.001
during the analysis period. Patients however can
manually select reanalysis, if they are not satisfied
0.6 with the ventilator-generated breathing pattern.
TA-mode ventilation is promising and an in-
Work of breathing (J/)

p < 0.001
novative new mode of ventilation with improved
0.4 unloading of respiratory muscles. Clinical studies
in respiratory failure of different etiologies how-
ever are required to prove clinical feasibility and
0.2 evaluate the clinical benefit.

p < 0.001
0 Conclusion
Unassisted S-mode TA-mode
TA-mode ventilation offers the opportunity of
Fig. 3. Work of breathing during unassisted breathing, additional respiratory muscle unloading because
S-mode and TA-mode NIV. it reduces the work required to trigger the ven-
tilator. The mode is well tolerated by the major-
ity of patients, however asynchrony with frequent
to a minority of patients (<5%). In general, pa- phases of re-analysis might compromise the qual-
tients have to understand the functioning of the ity of ventilation and user compliance in some
TA mode and should be instructed to breathe patients.

References
1 Ozsancak A, D’Ambrosio C, Hill NS: 4 Vitacca M, Barbano L, D’Anna S, Porta 6 Kohler D, Dellweg D, Barchfeld T,
Nocturnal noninvasive ventilation. Chest R, Bianchi L, Ambrosino N: Comparison Klauke M, Tiemann B: Time-adaptive
2008;133:1275–1286. of five bilevel pressure ventilators in mode, a new ventilation form for the
2 Ferrer M, Esquinas A, Leon M, Gonzalez patients with chronic ventilatory failure: treatment of respiratory insufficiency –
G, Alarcon A, Torres A: Noninvasive a physiologic study. Chest 2002;122: a self-learning system (in German).
ventilation in severe hypoxemic respira- 2105–2114. Pneumologie 2008;62:527–532.
tory failure: a randomized clinical trial. 5 Dellweg D, Schonhofer B, Haidl PM, et 7 Dellweg D, Barchfeld T, Klauke M, Eiger
Am J Respir Crit Care Med 2003;168: al: Short-term effect of controlled G: Respiratory muscle unloading during
1438–1444. instead of assisted noninvasive ventila- auto-adaptive non-invasive ventilation.
3 Girault C, Chevron V, Richard JC, et al: tion in chronic respiratory failure due to Respir Med 2009;103:1706–1712.
Physiological effects and optimisation of chronic obstructive pulmonary disease.
nasal assist-control ventilation for Respir Care 2007;52:1734–1740.
patients with chronic obstructive pul-
monary disease in respiratory failure.
Thorax 1997;52:690–696.

Dr. med. Dominic Dellweg


Pneumonology 1, Kloster Grafschaft
Annostrasse 1, DE–57392 Schmallenberg (Germany)
Tel. +49 2972 791 00, Fax +49 2972 791 2526
E-Mail d.dellweg@fkkg.de

TA Mode: A New Form of Self-Adjusting Controlled Ventilation 9


Section Title
Applied Technologies in Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 10–14

Influence of Ventilation Strategies on


Hemodynamics in Hypovolemic Shock
Holger Herff
Department of Anesthesiology and Critical Care Medicine, Innsbruck Medical University, Innsbruck, Austria

Abbreviations blood volume has the potential to reduce effects


CPR Cardiopulmonary resuscitation
of high PEEP levels on blood flow, as long as the
ITD Inspiratory threshold device right ventricle does not fail [5]. Vice versa, nega-
ITPR Intrathoracic pressure controller tive PEEP effects are more pronounced if the cen-
NEEP Negative end-expiratory pressure tral volume state is hypovolemic [6]. Thus, central
PEEP Positive end-expiratory pressure hypovolemic patients are at high risk of cardio-
vascular collapse even at moderate PEEP levels.
Right atrial pressures <10 cm H2O in intubated
Influence of Intrathoracic Pressure on patients seem to be critical if PEEP levels are in-
Blood Flow – Physiological Background creasing [5]. It is noteworthy that while PEEP may
be used in cardiogenic shock to reduce central
High positive ventilation pressures being induced volume and subsequently left ventricular preload
by high PEEP levels or recruitment maneuvers [7], in cardiac arrest increased central volume im-
are standard tools to improve oxygenation in proves blood flow during CPR [6, 8, 9]. Although
emergency and critical care patients, especially in cardiac arrest often may be the final form of car-
acute lung injury [1, 2]. However, positive pres- diogenic shock, the mechanisms determining
sure ventilation with PEEP increases intrathorac- blood flow in cardiac arrest and during CPR are
ic pressure which is subsequently transmitted to similar to those in hypovolemic shock. Since heart
intrathoracic vessels. Thus, venous return into the function is bridged by chest compressions during
thorax is decreased which has a significant nega- CPR, blood flow mainly depends on the quality of
tive impact on cardiac preload, cardiac output and chest compressions and central volume status and
subsequently on blood pressure [3]. PEEP levels thus central (relative) hypervolemia may increase
>15 cm H2O increase pulmonary arterial resis- blood flow whereas hypovolemia may reduce it.
tance directly which may additionally result in In consequence, ventilation strategies improving
right ventricular failure [4]. Normovolemic pa- blood flow in cardiac arrest [10, 11] are similar to
tients tolerate PEEP well and hypervolemic central those in hypovolemic shock [12, 13].
Conventional Therapeutic Consequences – we have to be aware that a strategy of high respi-
Reducing Mean Airway Pressures ratory rates and low tidal volumes may fail in ob-
structive patients such as asthmatics. Due to short
In a recent study, a strong correlation between expiratory times, incomplete expiration might
mean intrapulmonary pressure and blood flow lead to intrinsic PEEP levels that may again have
and cardiac output was demonstrated in a sim- detrimental effects on blood flow [14].
ulated hemorrhagic shock animal model [14].
Thus, ventilation strategies that may decrease
mean intrathoracic pressure either by reducing Experimental Therapeutic Consequences –
PEEP, lower tidal volumes, or lower respiratory Reducing Mean Airway Pressures Sub-Zero
rates may increase venous return to the heart and
subsequently cardiac output. The easiest step to Another more aggressive strategy to reduce mean
achieve reduced mean airway pressures is to re- airway pressure further is to apply NEEP. In a pig
duce PEEP. Such a reduction of PEEP from 10 to model of severe hemorrhagic shock, intermit-
5 or 0 cm H2O PEEP in severe hemorrhagic shock tent negative pressure was applied by suction-
increased short-term survival in pigs substantial- ing gas out of the airways during the expiratory
ly [15]. Another method may be to reduce tidal phase [16]. Ventilation was applied using normal
volumes; despite retaining PEEP and lung min- tidal volumes and respiratory rates. As a result,
ute volume constant by doubling respiratory rate all NEEP animals survived the 120-min observa-
in an animal model, survival rate was improved tional period whereas only half of the 0 PEEP and
by 40–60% for as much as 20 min longer due to none of the 5 PEEP animals survived. Reducing
reduced tidal volumes. In this experiment, mean the baseline pressure by reducing EEP to negative
airway pressure was reduced substantially by the levels is the most effective way to reduce mean air-
lower inflated tidal volumes despite higher respi- way pressure. Thus, it is not surprising that NEEP
ratory rates. Further, mean airway pressure cor- ventilation significantly increases blood flow in
related strongly with blood pressure and cardi- hemorrhagic shock [16].
ac output [14]. Thus, despite remaining minute While this strategy needed a special technical
ventilation and subsequently blood gases con- apparatus to apply NEEP in ventilated animals
stant and maintaining PEEP levels, the influence this could be achieved with less technical effort
of ventilation on blood pressure could have been in spontaneously breathing individuals. A spe-
decreased by reducing tidal volumes and subse- cial valve, ITD, was originally designed to be used
quently mean intrapulmonary pressure. Mean in- during CPR [8, 17]. The ITD closes the airway up
trapulmonary pressure as an important force that to a preset pressure during the chest decompres-
interferes with venous return explains apparently sion phase of CPR [8]. The forces of the recoil-
discrepancies to previous studies reporting higher ing chest that would suction air into the patient’s
respiratory rates being detrimental to blood flow lungs if the ventilation tube remains open, result
in acute shock states [12, 13]. In these studies, tid- in a subatmospheric pressure in the patient’s lungs
al volumes were constant and thus higher respi- if this tube is closed by the ITD [8]. The recoil-
ratory rates resulted in higher mean intrapulmo- ing forces can be further enhanced during CPR
nary pressures, whereas reduced respiratory rates if in addition to the forces generated by passive
with constant tidal volumes automatically result- recoiling of the chest wall, active decompression
ed in lower mean intrapulmonary pressures which is applied to the chest [18]. These negative airway
due to our data improve blood flow and survival pressures substantially increase venous return to
chances of experimental animals [14]. However, the heart resulting in a central hypervolemic state,

Ventilation in Hypovolemic Shock 11


thus ‘priming the pump’ for the next chest com- [26, 27]. Thus, despite still lacking experience in
pression. This strategy resulted in better blood humans in severe hemorrhagic shock, spontane-
flow in CPR models and increased survival rates ously breathing through an ITPR seems to be a
in clinical studies [17, 19]. promising concept to improve blood flow in hu-
In non-CPR states, e.g. during hemorrhagic man hypovolemic shock.
hypovolemic shock, such a negative airway pres-
sure cannot be generated by force on the chest.
However, the forces generated by the diaphragm Negative Side Effects of Decreasing
can be used in spontaneously breathing patients Intrapulmonary Pressures and Limitations
to generate subatmospheric intrapulmonary pres-
sures. Special valves for shock (ITPR) had been PEEP is used to improve oxygenation in ventilat-
developed comparable to the CPR-ITD. Within ed patients since it has the potential to remain the
general lower cracking pressures <10 cm H2O, alveoli open state [1]. Further, since many shock
the ITPR may allow patients to ventilate freely patients may suffer from thoracic trauma as well
against some tolerable resistance which gener- lung-protective ventilation, strategies including
ates negative intrapulmonary pressures [20]. The PEEP are efficient against pulmonary failure [28].
ITPR can be set on facemasks that then have to Thus, concepts that omit PEEP or induce NEEP
be sealed to patients’ faces tightly. These devices may gain some bargain in the immediate shock
improved substantially blood flow in shock states state while in the aftermath patients may be lost
in spontaneously breathing animals that were be- due to increased rates of pulmonary failure [29].
ing intubated due to anesthesia for animal pro- Thus, concepts that postulate to omit well-estab-
tection. Thus, short-term survival rates were sub- lished clinical concepts such as PEEP have to be
stantially increased in hemorrhagic shock models well deliberated and need more clinical evidence
[21, 22]. In a recent study the ITPR further im- in controlled prospective studies.
proved blood flow and short-term survival in a Especially high negative intrapulmonary pres-
pig model of acute heat stroke [23]. Although heat sures may result in pulmonary atelectasis and thus
shock may have a different etiology compared to right-to-left shunts that may endanger patients of
hemorrhagic shock, subatmospheric intrapulmo- hypoxia. In a study performed in 2001, we used
nary pressure seems to improve hemodynamics an ITD model with a cracking pressure of 35 cm
in other forms of hypovolemic shock, too. H2O in a CPR model [30]. Without active ven-
Testing the ITPR in healthy adult volunteers tilation the airway was completely obstructed
increased cardiac output up to 20%; further, heart which resulted in resorption atelectasis; although
rate increased as well as stroke volume indicat- the oxygen concentration in the alveoli was 100%
ing better venous return to the heart that forces before the experiment, the animals were severely
the (healthy) heart to increased work [24, 25]. In hypoxic after 2 min. In contrast, if the airway re-
further recent studies in human volunteers using mained open the SaO2 was still >95% after 5 min
the ITPR, hypotension was artificially induced experimental time [30]. Thus, if these valves are
by central hypovolemia due to progressive lower used in intubated patients during CPR, the best
body negative pressure. This was achieved by a way to avoid complete resorption atelectasis is in-
special garment applying –7 cm H2O on the lower termittent active positive pressure ventilation for
body half. In such a simulated central hypovolem- recruitment of atelectatic pulmonary areas [31].
ic state, spontaneous ventilation through an ITPR In spontaneously breathing patients, airway ob-
resulted in significantly improved cardiac out- struction may be especially dangerous due to
put and more stable cardiocirculatory conditions rapidly developing negative pressure pulmonary

12 Herff
edema [32]. Further, to avoid resorption atelecta- Conclusion
sis, subatmospheric pressures in spontaneously
breathing patients generated by special valves as Reducing mean airway pressure may be a strat-
the ITPR have to be so low that the valves open egy to improve venous return to the heart and
during every inspiratory effort. Last, breathing subsequently blood flow in hypovolemic shock
through an ITPR in shock increases work of pow- states. One method may be to omit PEEP or to
er for breathing; while healthy volunteers did not decrease tidal volumes. Experimental approaches
have any problems to compensate, this may be to reduce intrapulmonary pressures to subatmo-
different in multiple trauma patients [33]. spheric levels, either in spontaneously breathing
patients or in the expiratory phase during artifi-
cially ventilation, are not evidenced yet and need
further research.

References
1 Barbas C, de Matos G, Pincelli M, da 6 Ido Y, Goto H, Lavin M, Robinson J, 13 Pepe P, Raedler C, Lurie K, Wigginton
Rosa Borges E, Antunes T, de Barros J, Mangold J, Arakawa K: Effects of posi- J: Emergency ventilatory management
Okamoto V, Borges J, Amato M, de Car- tive end-expiratory pressure on carotid in hemorrhagic states: elemental or
valho C: Mechanical ventilation in acute blood flow during closed-chest cardio- detrimental? J Trauma 2003;54:1048–
respiratory failure: recruitment and high pulmonary resuscitation in dogs. Anesth 1055.
positive end-expiratory pressure are nec- Analg 1982;61:557–560. 14 Herff H, Paal P, von Goedecke A, Lind-
essary. Curr Opin Crit Care 2005;11: 7 Hoffmann B, Welte T: Non-invasive pos- ner KH, Severing AC, Wenzel V: Influ-
18–28. itive pressure ventilation in cardiogenic ence of ventilation strategies on sur-
2 Borges J, Okamoto V, Matos G, Caramez pulmonary edema (in German). Med vival in severe controlled hemorrhagic
M, Arantes P, Barros F, Souza C, Victo- Klin (Munich) 1999;94:58–61. shock. Crit Care Med 2008;36:2613–
rino J, Kacmarek R, Barbas C, Carvalho 8 Lurie K, Coffeen P, Shultz J, McKnite S, 2620.
C, Amato M: Reversibility of lung col- Detloff B, Mulligan K: Improving active 15 Krismer AC, Wenzel V, Lindner KH,
lapse and hypoxemia in early acute compression-decompression cardiopul- Haslinger CW, Oroszy S, Stadlbauer KH,
respiratory distress syndrome. Am J monary resuscitation with an inspira- Konigsrainer A, Boville B, Hormann C:
Respir Crit Care Med 2006;174:268–278. tory impedance valve. Circulation Influence of positive end-expiratory
3 Gernoth C, Wagner G, Pelosi P, Luecke T: 1995;91:1629–1632. pressure ventilation on survival during
Respiratory and haemodynamic changes 9 Lurie K, Mulligan K, McKnite S, Detloff severe hemorrhagic shock. Ann Emerg
during decremental open lung positive B, Lindstrom P, Lindner K: Optimizing Med 2005;46:337–342.
end-expiratory pressure titration in standard cardiopulmonary resuscitation 16 Krismer AC, Wenzel V, Lindner KH, von
patients with acute respiratory distress with an inspiratory impedance threshold Goedecke A, Junger M, Stadlbauer KH,
syndrome. Crit Care 2009;13:R59. valve. Chest 1998;113:1084–1090. Konigsrainer A, Strohmenger HU,
4 Biondi JW, Schulman DS, Soufer R, Mat- 10 Aufderheide T, Lurie K: Death by hyper- Sawires M, Jahn B, Hormann C: Influ-
thay RA, Hines RL, Kay HR, Barash PG: ventilation: a common and life-threaten- ence of negative expiratory pressure ven-
The effect of incremental positive end- ing problem during cardiopulmonary tilation on hemodynamic variables dur-
expiratory pressure on right ventricular resuscitation. Crit Care Med ing severe hemorrhagic shock. Crit Care
hemodynamics and ejection fraction. 2004;32(suppl):S345–S351. Med 2006;34:2175–2181.
Anesth Analg 1988;67:144–151. 11 Aufderheide T, Sigurdsson G, Pirrallo R, 17 Plaisance P, Lurie K, Payen D: Inspira-
5 Jellinek H, Krafft P, Fitzgerald R, Yannopoulos D, McKnite S, von Briesen tory impedance during active compres-
Schwarz S, Pinsky M: Right atrial pres- C, Sparks C, Conrad C, Provo T, Lurie K: sion-decompression cardiopulmonary
sure predicts hemodynamic response to Hyperventilation-induced hypotension resuscitation: a randomized evaluation
apneic positive airway pressure. Crit during cardiopulmonary resuscitation. in patients in cardiac arrest. Circulation
Care Med 2000;28:672–678. Circulation 2004;109:1960–1965. 2000;101:989–994.
12 Pepe P, Lurie K, Wigginton J, Raedler C,
Idris A: Detrimental hemodynamic
effects of assisted ventilation in hemor-
rhagic states. Crit Care Med
2004;32(suppl):S414–S420.

Ventilation in Hypovolemic Shock 13


18 Plaisance P, Lurie K, Vicaut E, Adnet F, 22 Yannopoulos D, McKnite S, Metzger A, 28 Laudi S, Donaubauer B, Busch T, Kerner
Petit J, Epain D, Ecollan P, Gruat R, Cav- Lurie K: Intrathoracic pressure regula- T, Bercker S, Bail H, Feldheiser A, Haas
agna P, Biens J, Payen D: A comparison tion improves 24-hour survival in a por- N, Kaisers U: Low incidence of multiple
of standard cardiopulmonary resuscita- cine model of hypovolemic shock. organ failure after major trauma. Injury
tion and active compression-decompres- Anesth Analg 2007;104:157–162. 2007;38:1052–1058.
sion resuscitation for out-of-hospital 23 Voelckel W, Yannopoulos D, Zielinski T, 29 Schwartz D, Maroo A, Malhotra A, Kes-
cardiac arrest. French Active Compres- McKnite S, Lurie K: Inspiratory imped- selman H: Negative pressure pulmonary
sion-Decompression Cardiopulmonary ance threshold device effects on hemorrhage. Chest 1999;115:1194–
Resuscitation Study Group. N Engl J hypotension in heat-stroked swine. 1197.
Med 1999;341:569–575. Aviat Space Environ Med 2008;79:743– 30 Herff H, Raedler C, Zander R, Wenzel V,
19 Wolcke B, Mauer D, Schoefmann M, 748. Schmittinger CA, Brenner E, Rieger M,
Teichmann H, Provo T, Lindner K, Dick 24 Convertino V, Ratliff D, Ryan K, Doerr Lindner KH: Use of an inspiratory
W, Aeppli D, Lurie K: Comparison of D, Ludwig D, Muniz G, Britton D, Clah S, impedance threshold valve during chest
standard cardiopulmonary resuscitation Fernald K, Ruiz A, Lurie K, Idris A: compressions without assisted ventila-
versus the combination of active com- Hemodynamics associated with breath- tion may result in hypoxaemia. Resusci-
pression-decompression cardiopulmo- ing through an inspiratory impedance tation 2007;72:466–476.
nary resuscitation and an inspiratory threshold device in human volunteers. 31 Dorph E, Wik L, Strømme T, Eriksen M,
impedance threshold device for out-of- Crit Care Med 2004;32(suppl):S381– Steen P: Oxygen delivery and return of
hospital cardiac arrest. Circulation S386. spontaneous circulation with ventila-
2003;108:2201–2205. 25 Convertino V, Ratliff D, Crissey J, Doerr tion: compression ratio 2:30 versus chest
20 Lurie K, Zielinski T, McKnite S, Idris A, D, Idris A, Lurie K: Effects of inspiratory compressions only CPR in pigs. Resusci-
Yannopoulos D, Raedler C, Sigurdsson impedance on hemodynamic responses tation 2004;60:309–318.
G, Benditt D, Voelckel W: Treatment of to a squat-stand test in human volun- 32 Fremont R, Kallet R, Matthay M, Ware L:
hypotension in pigs with an inspiratory teers: implications for treatment of Postobstructive pulmonary edema: a
impedance threshold device: a feasibility orthostatic hypotension. Eur J Appl case for hydrostatic mechanisms. Chest
study. Crit Care Med 2004;32:1555– Physiol 2005;94:392–399. 2007;131:1742–1746.
1562. 26 Convertino V, Ryan K, Rickards C, 33 Idris A, Convertino V, Ratliff D, Doerr D,
21 Yannopoulos D, Metzger A, McKnite S, Cooke W, Idris A, Metzger A, Holcomb J, Lurie K, Gabrielli A, Banner M: Imposed
Nadkarni V, Aufderheide T, Idris A, Adams B, Lurie K: Inspiratory resistance power of breathing associated with use
Dries D, Benditt D, Lurie K: Intratho- maintains arterial pressure during cen- of an impedance threshold device.
racic pressure regulation improves vital tral hypovolemia: implications for treat- Respir Care 2007;52:177–183.
organ perfusion pressures in normov- ment of patients with severe hemor-
olemic and hypovolemic pigs. Resuscita- rhage. Crit Care Med
tion 2006;70:445–453. 2007;35:1145–1152.
27 Ryan K, Cooke W, Rickards C, Lurie K,
Convertino V: Breathing through an
inspiratory threshold device improves
stroke volume during central hypov-
olemia in humans. J Appl Physiol
2008;104:1402–1409.

Dr. Holger Herff


Department of Anesthesiology and Critical Care Medicine
Innsbruck Medical University, Anichstrasse 35
AT–6020 Innsbruck (Austria)
Tel. +43 512 504 80375, Fax +43 512 504 6780375, E-Mail holger.herff@i-med.ac.at

14 Herff
Applied Technologies in Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 15–18

Gas Exchange during Perfluorocarbon Liquid


Immersion
Mark W. Daviesa–c ⭈ Kimble R. Dunstera,d
a
Grantley Stable Neonatal Unit, bPerinatal Research Centre, and cDepartment of Paediatrics and Child Health, Royal Brisbane and
Women’s Hospital, The University of Queensland, and dMedical Engineering Research Facility, Queensland University of Technology,
Brisbane, Qld., Australia

Abbreviations two key principles: (1) extremely preterm infants


PFC Perfluorocarbon
have very thin, poorly keratinized skin across
CPAP Continuous positive airway pressure which, it is well known, gas exchange can occur,
and (2) PFC liquid is an excellent carrier of both
oxygen and carbon dioxide.
Immersion in PFC liquid provides other po-
Given that there is potential for transcutaneous tential advantages: (1) water is not soluble in,
respiration in the extremely premature newborn or miscible with, PFC, so water will not evapo-
infant, it has been proposed that immersion of rate from the skin possibly slowing skin kerati-
these infants in PFC liquid will allow augmenta- nization (thus prolonging skin respiration); (2)
tion of gas exchange. This is possible because PFC it may allow tidal breathing of PFC liquid in a
liquid has an excellent oxygen- and carbon-diox- spontaneously breathing infant providing non-
ide-carrying capacity. Immersion in PFC may injurious lung gas exchange, and (3) tempera-
allow a spontaneously breathing infant to also ture regulation and control in the immersed in-
achieve lung gas exchange with the combination fant [1].
of skin and lung gas exchange being sufficient to
support life. Alternatively, immersion with some Experimental Studies
skin gas exchange may allow other forms of me- Only two papers have explored the concept of
chanical ventilation (with or without PFC) to be nursing whole subjects immersed in PFC liquid.
used but in a far less injurious way. Hiroma et al. [1] nursed adult rats in PFC liquid to
investigate the temperature control possibilities of
their ‘liquid incubator’. They found that the tem-
Analysis Main Topics Related to Title perature of rats two-thirds submerged in FC-43
was readily controlled and manipulated by chang-
Immersion in PFC and the Potential for Skin Gas ing the temperature of the FC-43.
Exchange In a small experimental pilot study, Davies et
The potential for gas exchange through the skin al. [2] explored the possibility of using whole-
when immersed in PFC liquid is feasible given body immersion in PFC to provide some degree
E F

B A

A D

Fig. 1. A lamb immersed in PFC liquid (B) circulated – via inlet and outlet pipes (A) – with an exter-
nal pump and heater. 100% oxygen (C) flows through a ‘bubble curtain’ (D). Fluids and drugs are
given by syringe pumps (E) into the umbilical vessels. Vital signs (incl. arterial pressure and oxygen
saturations) are monitored (F).

of skin gas exchange combined with tidal liquid infants also have a significant risk of brain in-
breathing in preterm lambs (fig. 1). They did not jury from intracerebral bleeding (intraventricu-
achieve any meaningful gas exchange. They did lar hemorrhage) and ischemia (porencephaly and
demonstrate that the central circulation remains periventricular leukomalacia). Any lung disease,
relatively intact immediately after birth, and and its required respiratory support, along with
whilst significant initial heat loss occurs before accompanying hemodynamic disturbance will
immersion, the fetus can be adequately warmed contribute to this increased risk of brain injury.
and temperature maintained with immersion in Survivors are also at risk of neonatal chronic lung
warm PFC liquid. disease which will lead to prolonged need for re-
spiratory support and oxygen as well as a con-
siderably longer stay in hospital. They also have
Discussion significant post-discharge respiratory morbidity
and mortality.
Extremely preterm newborn infants are often The extremely preterm newborn infant has
critically ill from birth. Mortality in this group very immature lungs and will almost certainly
of infants is high and a large part of that mortal- need respiratory support. The incidence of severe
ity is primarily due to lung immaturity and acute hyaline membrane disease is very high, but even
lung disease. Even when the baby’s death is not in the absence of lung disease they may require re-
directly due to lung disease it is often a major spiratory support because of their pulmonary im-
contributor to it. Extremely preterm newborn maturity and extremely compliant chest wall.

16 Davies ⭈ Dunster
Any respiratory support given has the potential breathing on CPAP, tidal PFC breathing or any
to cause lung injury, especially mechanical venti- form of mechanical ventilation.
lation and its associated ventilator-induced lung
injury. This lung injury can make the acute and Immersion in PFC with Spontaneous Liquid
chronic lung disease worse. Many forms of less in- Breathing
jurious respiratory support have been postulated If it is possible to support a spontaneously breath-
and used including nasal CPAP, high-frequency ing preterm infant immersed in PFC and achieve
ventilation, triggered and synchronized ventila- both lung gas exchange through tidal flow of PFC
tion, volume-targeted ventilation and liquid ven- in and out of the lungs and skin gas exchange from
tilation. Despite this, babies born at the margins direct contact with the PFC, then an extremely
of viability continue to have a high mortality and simple form of advanced life support would be
significant neurodevelopmental and respiratory available for the extremely preterm infant.
morbidity. A number of hurdles would have to be over-
If an alternative means of gas exchange could come before this technique would be remotely
be provided then we might be able to ventilate possible, and a number of conditions would need
in a less injurious way. The provision of gas ex- to be optimized for its success. These include: (a)
change requires adequate gas exchange surfaces prompt delivery and immersion in PFC; (b) en-
and sufficient oxygen and carbon dioxide gradi- suring adequate lung expansion – may require
ents with a functioning circulation. Alternative lung recruitment maneuvers prior to immersion
means of gas exchange include: (1) extracorporeal in PFC (with or without endotracheal intubation);
– whilst this has the potential to provide all the (c) ensuring adequate respiratory effort; (d) en-
gas exchange requirements it has so far not been suring an adequate circulation (may require cir-
possible in preterm infants and will likely result culatory support, e.g. adrenaline infusion), and
in intolerable fluctuations in hemodynamics and (e) the problem of the baby floating on the top of
an even greater risk of brain injury; (2) gut/peri- the very dense PFC.
toneum – gas exchange is possible over the gut
and peritoneum and better oxygenation has been Immersion in PFC as an Adjunct to Conventional
demonstrated with intra-abdominal PFC liquid Forms of Respiratory Support
in rabbits [3, 4], and (3) skin – the transfer of oxy- It may not be possible to achieve adequate gas ex-
gen and carbon dioxide is possible across the skin change through the lungs with spontaneous tid-
of the newborn human and the more immature al liquid breathing. However, the degree of skin
the infant, the greater the degree of gas transfer gas exchange achieved may be sufficient to aug-
[5–7]. ment that provided by more conventional means.
Davies et al. [2] have proposed, and tested in a Because some of the gas exchange is provided via
small experimental pilot study, that immersion in the skin, less will be needed through the lungs. In
PFC will provide some degree of skin gas exchange. the infant ventilated with conventional mechan-
The gas exchange provided by this means would ical ventilation this will allow the use of lower
hopefully supplement that available through the pressures and smaller tidal volumes and decrease
lungs. Thus the degree of respiratory support that baro- and volutrauma and its sequelae.
will be needed will be less because of that occur-
ring through the skin and therefore should be able Other Uses of Immersion in PFC
to be provided with a much lower risk of lung in- Apart from providing the capacity for skin gas
jury. The lung gas exchange could be provided exchange, immersion may have other advantag-
in a number of ways: with normal spontaneous es. Because there is no skin-air interface the skin

Gas Exchange during PFC Liquid Immersion 17


will not dry out and keratinization will be delayed. infant’s temperature to be manipulated, for exam-
Any skin gas exchange can therefore be main- ple for therapeutic hypothermia in infants with
tained. Immersion in PFC may provide a better hypoxic ischemic encephalopathy.
barrier to skin colonization with potentially in-
fecting organisms. Bacteria are not able to be car-
ried in PFC liquid and cannot be supported by it. Conclusion
Control of the temperature of the PFC liquid will
allow the maintenance of a normal temperature Although the initial studies in PFC immersion
and a thermo-neutral environment thus minimiz- have been unsuccessful in augmenting gas ex-
ing metabolic demand, oxygen consumption and change, the large potential for its use remains. A
carbon dioxide production. Manipulation of the large amount of research is needed to progress this
temperature of the PFC liquid will also allow an from an embryological technique to clinical use.

References
1 Hiroma T, Baba A, Tamura M, Naka- 3 Chiba T, Harrison MR, Ohkubo T, Rol- 6 Cartlidge PHT, Rutter N: Percutaneous
mura T: Liquid incubator with perfluo- lins MD, Albanese CT, Jennings RW: oxygen delivery to the preterm infant.
rochemicals for extremely premature Transabdominal oxygenation using per- Lancet 1988;1:315–317.
infants. Biol Neonate 2006;90:162–167. fluorocarbons. J Pediatr Surg 7 Cartlidge PHT, Rutter N: Percutaneous
2 Davies MW, Dunster KR, Wilson K: Gas 1999;34:895–901. respiration in the new-born infant.
exchange during perfluorocarbon liquid 4 Miyaguchi N, Nagahiro I, Kotani K, Effect of ambient oxygen concentration
immersion: life-support for the ex utero Nakanishi H, Mori H, Osaragi T, Shi- on pulmonary oxygen uptake. Biol Neo-
fetus. Med Hypotheses 2008;71:91–98. mizu N: Transintestinal systemic oxy- nate 1988;54:68–72.
genation using perfluorocarbon. Surg
Today 2006;36:262–266.
5 Evans NJ, Rutter N: Percutaneous respi-
ration in the newborn infant. J Pediatr
1986;108:282–286.

Mark W. Davies, FRACP


Grantley Stable Neonatal Unit, Royal Brisbane and Women’s Hospital, The University of Queensland
Brisbane, QLD 4029 (Australia)
Tel. +61 736 368 918, Fax +61 736 365 259
E-Mail Mark_Davies@health.qld.gov.au

18 Davies ⭈ Dunster
Applied Technologies in Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 19–27

Extracorporeal Membrane Oxygenation for


Respiratory and Heart Failure in Adults
J. Pat Herlihya ⭈ Pranav Loyalkab ⭈ Timothy Connollya ⭈
Biswajit Karb ⭈ Igor Gregoricc
a
Department of Pulmonary and Critical Care, and bDepartment of Cardiology, and cDepartment of Cardiovascular Surgery,
The Texas Heart Institute at St. Luke’s Episcopal Hospital, Baylor College of Medicine, University of Texas Health Science Center,
Houston, Tex., USA

Abbreviations witnessing a failed embolectomy of massive pul-


ARDS Acute respiratory distress syndrome
monary embolism in a young woman. In 1953, his
CESAR Conventional Ventilatory Support vs. Extra- ‘heart and lung apparatus’ successfully supported
corporeal Membrane Oxygenation for an 18-year-old through surgical closure of a large
Severe Adult Respiratory Failure atrial septal defect by pumping the patient’s blood
CHF Congestive heart failure from the vena cava through an oxygenator and
ECMO Extracorporeal membrane oxygenation
back into the aorta by way of intravascular cannu-
ELSO Extracorporeal Life Support Organization
ILA Interventional Lung Assist
lae and tubing [2]. Gibbon’s innovations, together
LA Left atrium with the work of other investigators from many
LV Left ventricle different institutions, resulted in practical, ef-
LVADs Left ventricular assist devices fective ‘cardiopulmonary bypass’ and allowed all
MV Mechanical ventilation manners of ‘open heart’ surgery to be performed
SIRS Systemic inflammatory response syndrome by the 1960s [3, 4].
VA Veno-arterial Efforts to bring the newly successfully extra-
VILI Ventilator induced lung injury corporeal oxygenation technology to the rescue
VV Veno-venous
of patients suffering from primary respiratory
failure, who were beyond the help of mechani-
cal ventilation (MV), began in 1965 [5]. The first
By the late 19th century, French and German sci- successful report of such was by Hill et al. [6], in
entists had developed devices that effectively ox- 1972, using a novel ‘membrane oxygenator’. A
ygenated blood outside of the body [1]. In 1931, series of trials beginning in the 1970s and con-
John Gibbon, then a Harvard fellow in surgery tinued through the 1990s demonstrated the ef-
at The Massachusetts General Hospital, began fectiveness of ECMO in the neonatal population
a lifetime of work to bring ‘extracorporeal oxy- (77% survival rate), as well as for pediatric pa-
genation’ into the service of cardiac surgery after tients (55% survival rate) suffering from a variety
of pathologies causing primary respiratory failure support respiratory failure [1, 3]. The 1980s saw
[7, 8]. However, a large trial of ECMO for adult the widespread introduction of microporous hol-
respiratory distress syndrome in the 1970s result- low polypropylene fibers into the design of mem-
ed in a 90% mortality [9]. Reasons for the stark brane oxygenators. Oxygen gas flows through
outcome differences were not clear at the time. the center of these fiber membranes and blood
Nevertheless, as such, ECMO use for adults, the around the oxygen, allowing much more efficient
primary subject of this review, was relegated to gas transfer [3]. Plasma leakage through these
rare and desperate circumstances over the next 30 membranes, however, complicated their use and
years. However, the successful use of ECMO to necessitated their frequent change, a situation not
treat ARDS in the recently completed CESAR trial ideal for long-term ECMO [14]. Within the last
[10, 11], as well as recent reports of effective use several years however, microfiber, microporous,
of ECMO to support acute heart failure, and re- polymethylpentene membrane oxygenators have
fractory cardiac arrest [12, 13], promises to bring been commercially deployed which provide high-
ECMO into more common and widespread use performance gas exchange with low resistance to
for adult patients. Before reviewing these stud- flow and relatively little blood trauma, but with-
ies and their implications for practice, it would out the plasma leakage. These devices have been
seem helpful to first look at several of the techni- found to perform very well for ECMO for more
cal advances in key tools for ECMO delivery, i.e. than a week at a time [15–17]. Very recently, these
oxygenators, vascular access catheters and blood oxygenators have been produced with protein and
pumps, that to a large degree have allowed the heparin molecules lining the membrane in order
current successes of ECMO. We will also brief- to decrease the systemic inflammation and blood
ly touch on some advances in understanding the clotting associated with passage of blood through
pathophysiology of lung injury and CHF, as well extrinsic surfaces. Certainly, this has led to de-
as innovations in MV and cardiac support that creased level of systemic anticoagulation required
have additionally contributed to ECMO success. to prevent thrombus formation in the oxygenator.
The original extracorporeal oxygenators used Whether these innovations will promote better
were ‘direct contact’ oxygenators wherein oxy- outcomes for ECMO is, of yet, unclear [14, 18].
gen gas was bubbled through blood, or applied Originally, vascular access for the creation of
to blood drawn into a thin layer. These were the the ECMO circuit in open heart surgery involved
so-called ‘bubble’, ‘screen’, and ‘film’ oxygenators. ‘central cannulation’ of either the vena cava or
These devices functioned adequately to oxygenate right atrium and the ascending aorta with large
blood for heart surgery, but their use was com- bore cannulae placed under direct visualization
plicated by the creation of gas bubbles and blood in an open chest. Initial protocols for ECMO in
trauma, and could therefore only be used for a the support of respiratory failure used peripher-
few hours. With the help of advancing material ally placed cannulae [19, 20]. These catheters used
science, the 1970s ushered in ‘membrane oxygen- a femoral or internal jugular vein insertion site to
ators’ which employed ultrathin, gas-permeable allow catheter access to blood from the vena cava
sheets, initially of silicon rubber, to create tiny as inflow to the oxygenator and a carotid or femo-
channels through which extracorporeal blood ral arterial cannula to provide outflow of the oxy-
flowed, juxtaposed to an oxygen gas source. This genated blood to the proximal aorta. However, by
technology allowed oxygen to diffuse into blood the 1990s, VA ECMO was largely abandoned for
without direct contact and its associated problems, use in respiratory failure in deference to a VV ap-
thus promoting better clinical tolerance and facil- proach whereby venous blood from the distal in-
itating the longer use of oxygenators necessary to ferior vena cava was pumped to the oxygenator

20 Herlihy ⭈ Loyalka ⭈ Connolly ⭈ Kar ⭈ Gregoric


and returned to the proximal vena cava or right of CHF may offer some advantages that we will
atrium, using two different femoral venous cath- touch on later [23].
eters or both a femoral and an internal jugular Pumps to move blood through the ECMO cir-
venous catheter. This was because thromboem- cuit were originally of the roller pump design.
bolic events from arterial catheters were found Two problems with this technique were direct
to be common and potentially devastating, as in blood trauma caused by squeezing the blood be-
the circumstance of a stroke. Furthermore, VV tween the tubing walls, and spalling, hence embo-
ECMO, aided by catheter systems with ever bet- lization, of plastic microfragments from the tub-
ter flow characteristics, proved just as effective, in ing wall [24]. The blood trauma led to hemolysis,
work largely done with the neonatal and pediatric intravascular coagulation and activation of sys-
populations [21, 22]. temic inflammation. Design innovations to mini-
ECMO, when deployed to support of heart fail- mize these complications have included impeller
ure, utilizes a VA circuit [20]. The circuit can utilize blades which, though an improvement, still cre-
a central vascular approach with catheters already ate significant blood turbulence and can promote
in place for the circumstance of post-cardiotomy, air retention in the blood returning to the patient.
or heart transplant failure, or a peripheral can- Most current ECMO protocols incorporate cen-
nulae approach for rapid deployment for cardiac trifugal pumps which promote laminar flow with
arrest or medically refractory CHF. Central cath- lessened blood trauma and air retention [13].
eters provide inflow to the oxygenator from the Catheters and pumps produced currently for
right atrium and outflow to the proximal ascend- ECMO have biologic and antithrombotic coatings
ing aorta with the advantage of providing oxygen- to further reduce thromboembolic events and, as
ated blood in a position where it carries well to the in the case of oxygenators, to lessen systemic in-
coronary and cranial arteries. Of course, bleeding flammation that results from blood contact with
and infectious complications are of high concern an extracorporeal surface. Less systemic anticoag-
when VA ECMO is so configured for any signifi- ulation needs to be used with these systems, miti-
cant length of time. Peripheral VA ECMO circuits gating bleeding, a major complication of ECMO
consist of jugular or femoral venous access to the [20].
vena cava as inflow to the oxygenator and carotid Several major advances in the understanding
or femoral artery catheter access to the aorta for of acute lung injury, ARDS, and MV in support of
outflow delivery. Carotid catheters have the ad- these pathologies have been made since the 1970s,
vantage of delivering oxygenated blood to the as- which, no doubt, have contributed in a major way
cending aorta, but their use can be complicated by to the current success with ECMO. In the 1980s it
thromboembolic stroke. Femoral artery catheters was discovered that positive pressure MV settings
terminate in the proximal descending aorta, and that were heretofore considered safe and effective
provide antegrade flow of oxygenated blood into were in fact often injurious to lungs, especially
the aortic arch. With this approach, well-oxygen- those already injured [25]. In fact, ventilators can
ated blood may not carry well to the coronary ar- promote ARDS and even SIRS. Current genera-
teries, brain or upper extremities. For this reason, tion ventilators are built with the principles of pre-
when such a circuit is used, it is recommended to venting VILI, and clinical protocols for avoiding
place a right radial arterial line as a monitor for ‘volutrauma’ and other forms of VILI are in wide-
upper extremity, and hence heart and brain, blood spread use. Interestingly, MV of neonates and pe-
oxygenation. New catheter devices that permit ac- diatric patients has, since almost the beginning of
cess to the left atrium promise another route of positive pressure ventilation, incorporated pres-
inflow to the oxygenator and in the circumstance sure limitation strategies [26]. One wonders if the

Extracorporeal Membrane Oxygenation for Respiratory and 21


Heart Failure in Adults
difference between adult volume-cycled ventila- of the study was 63% as opposed to those receiv-
tion and pressure-cycled ventilation for pediatrics ing conventional MV for whom it was 47%. These
accounted in some significant degree for the out- data are consistent with that reported in the ELSO
come differences between pediatric and adult se- registry maintained by the University of Michigan
ries of ECMO in the 1970s. During the late 1980s [www.elso.med.umich.edu] which shows current
and early 1990s there also came a realization that survival rates of ECMO for adult ARDS of >50%
ARDS progressed through two pathophysiolog- [28].
ic phases, the early ‘exudative’ phase and subse- These data suggest that ECMO has arrived
quently the ‘fibroproliferative’ phase [27]. If the as therapy for early and severe ARDS, and other
lung did not go down the path of normal archi- potentially acute, reversible severe lung injuries
tecture restoration, but instead to fibrosis and scar such as cryptogenic organizing pneumonia [23],
formation, outcomes were much worse. This un- among other pathologies [29]. The CESAR trial
derstanding has figured in selection of patients concept was not only to support patients through
more likely to benefit from ECMO. respiratory failure but to intervene early to pre-
vent toxic use of MV and allow the lung its best
chance of recovery. Late ARDS, presumably in the
ECMO for ARDS and Primary Respiratory fibroproliferative phase, with its poorer prognosis,
Failure was notably excluded. It is also salient that 22 of
90 patients, essentially a quarter of those eligible
In February 2008, results of the CESAR trial were for the ECMO protocol, never received it because
released at the international Society of Critical they improved. Separating those patients with
Care Medicine meeting [10]. The full report was early severe ARDS who will benefit from ECMO
published in The Lancet in late 2009 [11]. This and those who will spontaneously improve will
study, however, may already be promoting a para- likely prove very difficult, if not impossible, at the
digm shift in the management of severe lung inju- bedside with our current diagnostic and prognos-
ry. Essentially this randomized prospective study tic tools.
showed significantly improved outcomes using
ECMO to manage severe and early ARDS com-
pared to the standard current approach of MV, at ECMO for Primary Cardiac Failure
what is currently thought to be noninjurious lev-
els. Specifically, the entry criteria to the study was Management of the failing heart has come a long
potentially reversible lung injury of high severity way from the 1960s when the first successful heart
as determined by a Murray score >3.0 (severe hy- transplant was performed, and the first attempt at
poxia, extensive pulmonary infiltrates by chest x- artificial heart support was made. Percutaneous
ray, need for high levels of MV, and poor pulmo- coronary intervention has, of course, revolution-
nary compliance) or uncompensated hypercarbia ized the management of acute cardiogenic shock
resulting in a pH <7.2. Exclusion criteria includ- from coronary occlusion. LVADs have come into
ed late ARDS as determined by a requirement of frequent use in heart transplant centers as a bridge
high FiO2 (>80%), or high ventilator measured to transplant, and the current generation of de-
airway pressure (plateau pressure >30 cm H2O) vices is now even approved as destination ther-
for >7 days. Moribund patients and those with apy [30]. Other mechanical support devices in
contraindications to heparin were also exclud- early clinical application or development, such
ed. For patients randomized to ECMO, survival as the total artificial heart, are encouraging [31].
without severe disability, the primary endpoint Understanding of ventricular remodeling and

22 Herlihy ⭈ Loyalka ⭈ Connolly ⭈ Kar ⭈ Gregoric


heart recovery from injury has also come a long that the patient is beyond recovery for reasons of
way since the early days when damaged heart neurologic injury or multiple system organ fail-
tissue was thought to be adynamic and fixed in ure, or cardiac transplantation/mechanical heart
dysfunction [31, 32]. Current clinical standards assist device placement can be performed [38].
of CHF management incorporate many of these We have even reported using ECMO as bridge to
concepts, such as the critical importance of after- heart lung transplant in a patient with severe re-
load reduction for optimal cardiac function and fractory primary pulmonary hypertension [39].
recovery from injury. Potentially revolutionary The last several years have seen ECMO applied to
new therapies, such as stem cell implantation and cases of refractory cardiac arrest, and data from
myocardial cell apoptosis suppression, are being ELSO show the survival rate in this circumstance
investigated by us at the Texas Heart Institute and to be an amazing 27% [40].
many others throughout the world [32].
These advances have spurred the burgeoning
interest in ECMO as a temporary support device Practical Aspects
for medically refractory CHF, cardiogenic shock,
and failed resuscitation of cardiac arrest, until ei- At this point, some practical aspects of provid-
ther heart recovery or implementation of a longer ing ECMO for lung or heart patients are worth
term solution such as cardiac transplant or LVAD reviewing. First of all, ECMO is an extraordinary
insertion. It seems a bit ironic that ECMO is com- resource and labor-intensive process. Institutions
ing back to serve its original purpose of cardiac and individual providers need to be prepared
support, but outside of the operating room, and for such. That being said, there are indications
for a much more extended period of time. that, at least for ARDS, ECMO may prove ec-
Following initial successes in supporting neo- onomically viable relative to current standard
natal and pediatric respiratory failure, cardiolo- therapy [11]. In addition to the equipment for
gists have been using ECMO in this age group ECMO a dedicated team of specialists experi-
over the past 20 years for severe CHF, pulmonary enced in ECMO is recommended. ELSO recom-
hypertension, and intracardiac shunts. Survival to mends that institutions perform 20 cases a year
discharge for these patients, who of course benefit to achieve and maintain optimal skill levels. The
from further interventions in addition to ECMO, team should include: perfusionists, for contin-
is 40% [12]. Data for ECMO use in adult CHF ual maintenance and monitoring of the ECMO
and cardiogenic shock that has come out within system; interventionalists, for catheter place-
the last decade have consistently shown remark- ment; pulmonologists and respiratory therapists,
able survival rates of 50–60% [13, 33–36]. Disease to monitor lung injury and maintain appropri-
states that ECMO alone may be sufficient for, giv- ate ventilator settings; cardiologists, to monitor
en the oftentimes temporary nature of the pathol- and manage central hemodynamics, hematolo-
ogy, include: the low cardiac output syndrome gists, to help for anticoagulation, and to monitor
post-cardiotomy, post-partum cardiomyopathy, for coagulopathy, thrombocytopenia and bleed-
viral myocarditis, arrhythmia-induced cardio- ing complications; nephrologists, to monitor and
myopathy, and severe pulmonary hypertension manage renal function and volume status; infec-
with right ventricular failure resulting from pul- tious disease specialists, to monitor the many
monary embolism or post-pulmonary artery sur- potential sources or infections and to treat such
gical thrombectomy [37]. In patients who do not early and aggressively; nutritionists, to prevent
recover sufficient intrinsic heart function, ECMO malnutrition; potentially neurologists, to asses
has served well as bridge therapy until it is clear neurologic change; and of course, the bedside

Extracorporeal Membrane Oxygenation for Respiratory and 23


Heart Failure in Adults
ICU nurse, whose array of assessment, interven- the pump, the countercurrent gas flow through
tion and communication skills make it all work. the oxygenator, known as the ‘sweep’, and the
Patients who survive ECMO need a variety of FiO2 of the gas. Initial blood flow is approxi-
support services, as do their families. Social ser- mately 70 ml/kg/min (or 5 l/min for an aver-
vice support is key over the long haul of illness age-sized man). For large individuals a second
and recovery. High level rehabilitation services ECMO system may be required. Initial sweep
are typically involved at the earliest opportu- should match the blood flow. So if 5 l/min of
nity in our patients’ post-ECMO as they often blood is flowing through the system, then the
demonstrate severe deconditioning or myopathy, sweep should be 5 l/min. Initially the sweep gas
neuropathy of critical illness. should be 100% FiO2. Flow through the oxygen-
Vascular access for ECMO has been discussed ator can be increased to improve oxygenation
above. Essentially, VV ECMO is the current stan- of the blood or improve O2 delivery. Increasing
dard for support of primary respiratory failure. sweep can also improve oxygenation of the
VA ECMO is used for primary cardiac failure. blood as well as offload CO2. A primary goal
However, it is worth noting here that this con- of ECMO for respiratory failure is to allow lung
figuration causes significant afterload for the left injury recovery. A secondary goal of ECMO in
ventricle [13]. This is at least counterproductive the circumstance of cardiac failure is to allow
for a ventricle with potential for recovery and ventilator settings that are noninjurious to the
may be catastrophic resulting in pulmonary hem- lung. Both goals are served by adjusting MV to
orrhage. Often, in this circumstance, an IABP is a Vt of 4–8 ml/kg of ideal body weight (in the
placed to off-load the ventricle. Unfortunately, case of volume-cycled ventilation), disallowing
this counterpulsation device may interfere with a plateau pressure of >30 cm H2O, and using an
the ECMO arterial catheter delivering oxygenat- FiO2 of 30% or less. This is usually accomplished
ed blood. Several solutions have been proposed gradually while adjusting ECMO to compensate
for this problem. Creation of an atrial shunt to by for lost gas exchange.
percutaneous transeptal perforation, and direct A trial of wean from ECMO, in the circum-
cannulation of the LA or LV as a vent to unload stance of primary respiratory failure, is made when
the LV have been reported [13]. Additionally, a the chest x-ray has sufficiently cleared and pulmo-
percutaneous LVAD, specifically the Impella nary compliance is has sufficiently improved [11].
Recovery (Abiomed Inc., Danvers, Mass., USA), At that point, sweep is turned down, and eventu-
has been placed to effectively unload the LV in ally off, to see if gas exchange can be maintained
conjunction with ECMO to good effect [41]. We by the ventilator alone, on nontoxic settings (as
have reported the use of the TandemHeart System delineated above) that limit volutrauma and an
(CardiacAssist Inc., Pittsburg, Pa., USA) catheters FiO2 of 60% or less. In the case of ECMO for car-
to access the LA as the delivery point for ECMO- diac support the key test of weanability is whether
treated blood in a patient with severe pulmonary CO can be maintained at a sufficient level for tis-
hypertension and right ventricular failure [23]. It sue O2 delivery, as determined by a combination
may be possible to use the LA access of the TH of objective measurements of (>2.2 l/min/m2) and
system as inflow to ECMO and thereby reduce LV clinical markers of sufficient oxygenation, such
load and wall stress. as lactic acid level and organ function, while the
Once vascular access for ECMO has been es- pump flow is decreased. Blood flow of <2 l/min
tablished, the three key components of ECMO through the ECMO circuit can prompt stasis in
requiring fairly continuous adjustment are the the system, so that is the usual lower limit of pump
blood flow through the oxygenator, regulated by flow in a wean trial.

24 Herlihy ⭈ Loyalka ⭈ Connolly ⭈ Kar ⭈ Gregoric


Continuous sedation of patients undergoing of sweep gas the ILA can result in dramatic im-
ECMO is necessary for tolerance. Fentanyl, with provements in gas exchange. It has been used
its relatively favorable hemodynamic profile, has in primary hypoxemic and hypercarbic respira-
worked well for us. Paralysis is also sometimes tory failure with survival rates of 41%, a signif-
required. Potential complications of ECMO are icant improvement over expected mortality in
many [8, 14, 20] but include, in order of practical these patients using conventional therapy [42].
importance: bleeding from administered antico- Interestingly it has been applied to chest trauma
agulation as well as coagulopathy and thrombocy- and blast victims because of its rapid application
topenia induced by the ECMO system; intracranial and lack of need for a pump, to excellent effect
bleeds are of particular concern; thromboembo- [14, 43]. It has also been used successfully as a
lic events, both venous and arterial; deep venous bridge to lung transplant [43].
thrombosis can occur at venous catheter inser- The first clinical case reports using paracor-
tion sites and embolize; arterial access sites can poreal artificial lung have just come out [44, 45].
thrombose and embolize causing, depending This device is essentially a membrane oxygenator
upon the location of the catheter, limb ischemia placed in parallel to the pulmonary circulation
or vital organ infarcts including CVAs (10% inci- with connection to the pulmonary artery and to
dence in adults on VA ECMO); hepatopathy and the left atrium. It has been used in the specific cir-
ischemic bowel from poor splanchnic perfusion, cumstance of right ventricular failure secondary to
sepsis from lung, catheter, urinary and abdomi- pulmonary hypertension, when standard ECMO
nal sources; SIRS, and multisystem organ failure. is failing. It performed well in these circumstances
Device malfunctions which occur in up to 18% supporting 1 patient for 63 days. There are hopes
of cases include: oxygenator failure requiring that, at a now distant point, this approach may ul-
change out (usually due to thrombus formation), timately result in a device that can be used long
and oxygenator or tubing rupture, resulting in air term, even out of the hospital [46].
emboli. Intravascular oxygenator systems such as the
intravascular oxygenator [47, 48] and the Hattler
catheter [49] are in the early stages of develop-
Promising Developments ment, or subclinical testing. These systems are de-
signed for implantation in the vena cava with or
There are several exciting and very promising without blood pumps and using an external gas
developments in the area of lung assist devices. source for oxygenation.
The ILA (Novalung GmbH, Talheim, Germany)
has recently been approved for clinical use in
Europe. This polymethylpentene membrane ox- Conclusion
ygenator is significantly smaller than traditional
oxygenators, has excellent gas exchange charac- ECMO appears poised to take a place in best prac-
teristics, and is constructed in such a way that tice clinical algorithms for managing severe acute
it presents very little resistance to blood flow lung injury, and medically refractory heart failure
through it. As such, it is pumpless, and driven as a bridge to recovery, implantable mechanical
by arterial flow in patients with intact hemody- cardiac assist device, or heart, lung or heart lung
namics, when placed in a femoral artery to ILA transplantation.
to femoral vein circuit. Typically only 1–2 l/min
of blood flows through the oxygenator or about
20% of cardiac output. However, with high flow

Extracorporeal Membrane Oxygenation for Respiratory and 25


Heart Failure in Adults
References
1 Lim MW: The history of extracorporeal 13 Matsumiya G, Saitoh S, Sawa Y: Extra- 24 Kopp R, Steinseifer U, Rossant R:
oxygenators. Anaesthesia 2006;61:984– corporeal assist circulation for heart fail- Extracorporeal lung assist for ARDS:
995. ure. Circ J 2009;73(suppl A). past, present and future; in Vincent, J-L
2 Zareba KM: John H. Gibbon, Jr, MD: a 14 Pesenti A, Zanelli A, Patroniti N: Extra- (ed): Intensive Care Medicine Annual
poet with an idea (1903–1973). Cardiol J corporeal gas exchange. Curr Opin Crit Update 2008. New York, Springer Sci-
2009;16:98–100. Care 2009;15:52–58. ence + Business Media, 2008, pp 235–
3 Haworth WS: Gibbon’s heart-lung 15 Toomasian JM, Schreiner RJ, Meyer 247.
machine. The development of the mod- DE, et al: A polymethylpentene fiber 25 Tremblay LN, Slutsky AS: Ventilator-
ern oxygenator. Ann Thorac Surg 2003; gas exchanger for long-term extracor- induced lung injury: from the bench to
76:S2216–S2219. poreal life support. ASAIO J the bedside. Intensive Care Med 2006;
4 Daly RC, Dearani JA, McGregor CG, 2005;51:390–397; erratum in ASAIO J 32:24–33.
Mullany CJ, Orszulak TA, Puga FJ, et al: 2008;54:137. 26 AARC Clinical Practice Guidelines: Neo-
Fifty years of open heart surgery at the 16 Formica F, Avalli L, Martino A, Mag- natal time-triggered, pressure-limited,
Mayo Clinic. Mayo Clinic Proc 2005;80: gioni E, Muratore M, Ferro O, et al: time-cycled mechanical ventilation.
636–640. Extracorporeal membrane oxygenation Respir Care 1994;39:808–816.
5 Rashkind WJ, Freeman A, Klein D, et al: with a poly-methylpentene oxygenator 27 Luh S, Chiang C: Acute lung injury/acute
Evaluation of a disposable plastic, low (Quadrox D). The experience of a single respiratory distress syndrome: the
volume, pumpless oxygenator as a lung Italian centre in adult patients with mechanism, present strategies and
substitute. J Pediatr 1965;66:94–102. refractory cardiogenic shock. ASAIO J future perspectives of therapies. J Zheji-
6 Hill JD, O’Brien TG, Murray JJ, Dontigny 2008;54:89–94. ang Univ Sci B 2007;8:60–69.
L, Bramsom ML, Osborn JJ, et al: Pro- 17 Agati S, Ciccarello G, Fachile N, et al: 28 Conrad SA, Rycus PT, Dalton H: Extra-
longed extracorporeal oxygenation for DIDECMO: a new polymethylpentene corporeal Life Support Registry Report
acute post-traumatic respiratory failure oxygenator for pediatric extracorporeal 2004. ASAIO J 2005;51:4–10.
(shock-lung syndrome). Use of the membrane oxygenation. ASAIO J 2006; 29 Kahn J, Muller H, Marte W, Rehak P,
Bramson membrane lung. N Engl J Med 52:509–512. Wasler A, Prenner G, et al: Establishing
1972;286:629–634. 18 Palatianos GM, Foroulos CN, Vassili MI, extracorporeal membrane oxygenation
7 Dalton HJ, Rycus PT, Conrad SA: Update Astras G, Triantafillou K, Papadakis E, et in a university clinic: case series. J Car-
on extracorporeal life support 2004. al: A prospective, double-blind study on diothorac Vasc Anesth 2007;21:384–387.
Semin Perinatol 2005;29:24–33. the efficacy of the bioline surface-hepa- 30 Lahpor J: State of the art: implantable
8 Schuerer DJ, Kolovos NS, Boyd KV, Coo- rinized extracorporeal perfusion circuit. ventricular assist devices. Curr Opin
persmith CM: Extracorporeal membrane Ann Thorac Surg 2003;76:129–135. Organ Transplant 2009;14:554–559.
oxygenation: current clinical practice, 19 Hill JD: Acute pulmonary failure: treat- 31 Jugdutt BI, Current and novel cardiac
coding, and reimbursement. Chest ment with extracorporeal oxygenation. support therapies. Curr Heart Fail Rep
2008;134:179–184. Med Instrum 1977;11:198–201. 2009;6:19–27.
9 Zapol WM, Snider MT, Hill JD, et al: 20 Marasco SF, Lukas G, McDonald M, 32 Khovnezhad A, Jalali Z, Tortolani AJ A
Extracorporeal membrane oxygenation McMillan J, Ihle B: Review of extracor- synopsis of research in cardiac apoptosis
in severe acute respiratory failure: a ran- poreal membrane oxygenation support and its application to congestive heart
domized prospective study. JAMA 1979; in critically Ill adult patients. Heart Lung failure. Tex Heart Inst J 2007;34:352–
242:2193–2196. Circ 2008;17(suppl 4);S41–S47. 359.
10 Peek G: CESAR: adult ECMO vs. conven- 21 Knight GR, Dudell GG, Evans ML, 33 Koerner MM, Jahanyar J: Assist devices
tional ventilation trial. Society of Critical Grimm PS: A comparison of venovenous for circulatory support in therapy-
Care Medicine – 37th Critical Care Con- and venoarterial extracorporeal mem- refractory acute heart failure. Curr Opin
gress, Honolulu, February 2–6, 2008. brane oxygenation in the treatment of Cardiol 2008;23:399–406.
11 Peek GJ, Mugford M, Tiruvoipati R, et al: neonatal respiratory failure. Crit Care 34 Luo XJ, Wang W, Hu SS, Gao HW, Long
Efficacy and economic assessment of Med 1996;24:1678–1683. C, Song YH, et al: Extracorporeal mem-
conventional ventilatory support versus 22 Pettignano R, Fortenberry JD, Heard brane oxygenation for treatment of car-
extracorporeal membrane oxygenation ML, Labuz MD, Kesser KC, Tanner AJ, et diac failure in adult patients. Interact
for severe adult respiratory failure al: Primary use of the venovenous Cardiovasc Thorac Surg 2009;9:296–
(CESAR): a multicentre randomised approach for extracorporeal membrane 300.
controlled trial. Lancet 2009;374:1351– oxygenation in pediatric acute respira- 35 Hoefer D, Ruttman E, Poelzl G, Kilo J,
1363. tory failure. Pediatr Crit Care Med Hoermann C, Margreiter R, et al: Out-
12 Cooper DS, Jacobs JP, Moore L, Stock A, 2003;4:291–298. come evaluation of the bridge-to-bridge
Gaynor JW, Chancy T, et al: Cardiac 23 Herlihy JP, Loyalka P, Jayaraman G, Kar concept in patients with cardiogenic
extracorporeal life support: state of the B, Gregoric, ID: Extracorporeal mem- shock. Ann Thorac Surg 2006;82:28–33.
art in 2007. Cardiol Young 2007;17(suppl brane oxygenation using the Tandem-
2):104–115. Heart System’s catheters. Tex Heart Inst J
2009;36:337–341.

26 Herlihy ⭈ Loyalka ⭈ Connolly ⭈ Kar ⭈ Gregoric


36 Murashita T, Eya K, Miyatake T, et al: 40 Thiagarajan RR, Brogan TV, Scheurer 45 Camboni D, Philipp A, Arit M, Pfeiffer
Outcome of the perioperative use of per- MA, Laussen PC, Rycus PT, Bratton SL: M, Hilker M, Schmid C: First clinical
cutaneous cardiopulmonary support for Extracorporeal membrane oxygenation experience with a paracorporeal artifi-
adult cardiac surgery: factors affecting to support cardiopulmonary resuscita- cial lung in humans. ASAIO J 2009;55:
hospital mortality. Artif Organs 2004;28: tion in adults. Ann Thorac Surg 2009;87: 304–306.
189–195. 778–785. 46 Zwischenberger BA, Clemson LA, Zwis-
37 Ogino H, Ando M, Matsuda K, Sasaki H, 41 Vlasselaers D, Desmet M, Desmet L, chenberger JB: Artificial lung: progress
Nakanishi N, et al: Japanese single-cen- Meyns B, Dues J: Ventricular unloading and prototypes. Expert Rev Medical
ter experience of surgery for chronic with a miniature axial flow pump in Devices 2006;3:485–497.
thromboembolic pulmonary hyperten- combination with extracorporeal mem- 47 Zwischenberger JB, Tao W, Bidani A:
sion. Ann Thorac Surg 2006;82:630–636. brane oxygenation. Intensive care Med Intravascular membrane oxygenator and
38 Copeland J: Invited commentary. Ann 2006;32:329–333. carbon dioxide removal devices: a
Thorac Surg 2006;82:33–34. 42 Bein T, Weber F, Philipp A, et al: A new review of performance and improve-
39 Gregoric ID, Chandra D, Myers TJ, Shei- pumpless extracorporeal interventional ments. ASAIO J 1999;45:41–46.
nin SA, Loyalka P, Kar B: Extracorporeal lung assist in critical hypoxemia/hyper- 48 Cattaneo G, Strauss A, Reul H: Compact
membrane oxygenation as a bridge to capnia. Crit Care Med 2006;34:1372– intra- and extracorporeal oxygenator
emergency heart-lung transplantation in 1377. developments. Perfusion 2004;19:251–
a patient with idiopathic pulmonary 43 Meyer A, Struber M, Fisher S: Advances 255.
arterial hypertension. J Heart Lung in extracorporeal ventilation. Anesthe- 49 Hattler BG, Lund LW, Golob J, et al: A
Transplant 2008;27:466–468. siol Clin 2008;26:381–391. respiratory gas exchange catheter: in
44 Schmid C, Philip A, Hiker M, Arit M, vitro and in vivo tests in large animals. J
Trabald B, Pfeiffer M, et al: Bridge to Thorac Cardiovasc Surg 2002;124:520–
lung transplantation through a pulmo- 530.
nary artery to left atrial oxygenator cir-
cuit. Ann Thorac Surg. 2008;85:1202–
1205.

J. Pat Herlihy, MD
Department of Pulmonary and Critical Care
The Texas Heart Institute at St. Luke’s Episcopal Hospital
Baylor College of Medicine, University of Texas Health Science Center
6624 Fannin Street, Suite 1730
Houston, TX 77030 (USA)
Tel. +1 713 255 4000, Fax +1 713 255 4050, E-Mail jph@houstonlungdocs.com

Extracorporeal Membrane Oxygenation for Respiratory and 27


Heart Failure in Adults
Section Title
Applied Technologies in Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 28–34

Automatic Control of Mechanical Ventilation


Technologies
Fleur T. Tehrani
Department of Electrical Engineering, California State University, Fullerton, Calif., USA

Abbreviations and morbidity rates of patients on this medical


ASV Adaptive support ventilation
treatment. Therefore, any useful innovation in
ATC Automatic tube compensation mechanical ventilation technology that can im-
Edi Neural drive signal to the diaphragm prove and expedite the treatment can significantly
FiO2 Fraction of inspired oxygen impact the quality of healthcare for ICU patients.
HFJV High-frequency jet ventilation The following provides a brief overview of dif-
HFOV High-frequency oscillatory ventilation ferent mechanical ventilation technologies and a
NAVA Neurally adjusted ventilatory assist discussion of several innovative techniques that
PAV Proportional assist ventilation have been employed in practice in mechanical
PEEP Positive end-expiratory pressure
ventilators in recent years. The potential impacts
PPS Proportional pressure support ventilation
on healthcare by several newer techniques includ-
VAPS Volume-assured pressure support
ing closed-loop ventilation technologies will be
analyzed by discussing the applications of those
techniques along with their main strengths and
Mechanical ventilation is a life-saving technolo- weaknesses.
gy that has evolved remarkably in the past sev-
eral decades. In particular, positive pressure me-
chanical ventilation in many different forms and An Overview of Various Mechanical
by using various modalities is employed as an es- Ventilation Technologies
sential medical treatment in the ICU settings of
hospitals all over the world. Without this tech- Mechanical ventilation is used in various forms
nology, the treatment of many illnesses and most and by using different technologies. In what is
major surgical operations would not be possible. called negative pressure ventilation, the patient’s
However, if this treatment is not provided prop- thoracic area or his entire body up to his neck is
erly or the duration of mechanical ventilation is enclosed in an airtight chamber and the volume
unduly prolonged due to non-optimal treatment, of the chamber is expanded, thereby inflating the
many medical complications such as ventilator- lungs using negative pressure. Examples of such
associated pneumonia may result. Those compli- devices are tank ventilators, jacket ventilators, and
cations can significantly increase the mortality cuirasses. However, negative pressure ventilation
devices are not used in many applications today. limitations of the advanced ventilators, and based
Instead, positive pressure ventilation in which on the patient’s illness and requirements chooses
the patient’s lungs are inflated by application of a suitable treatment option among many alterna-
positive pressure to his airways is considered as tives and sets the main ventilatory parameters.
today’s dominant technology. This kind of treat- The clinician’s decisions which in many cases can
ment can be applied non-invasively, by use of a make the difference between life and death for the
facial mask or a mouthpiece, or it can be given in- patient need to be made in the ICU setting within
vasively by using an endotracheal tube or by use a short period of time.
of tracheostomy. It has been reported that in the USA alone there
Positive pressure ventilation can be provided by are between 44,000 and 98,000 injuries resulting
using high-frequency ventilation techniques such from medical errors per year [2]. The human costs
as HFJV and HFOV. In high-frequency ventila- of serious injuries and mortalities associated with
tion, the oxygenated air is delivered at high rates medical errors cannot be measured financially
and small volumes to the patient’s airways, vibrat- and the financial costs of such incidents are be-
ing the alveoli at high frequencies to avoid injur- tween USD 17 and 29 billion per year in the USA
ing the lung tissue. High-frequency ventilation is alone [2]. It is needless to say that by expecting
used predominantly in the neonatal and pediat- so much from clinicians, such errors become in-
ric patient populations with less frequent appli- evitable. Medical technology has to be employed
cations in adult patient groups. The more com- more aggressively to address this issue. As was
monly used type of positive pressure ventilation is discussed above, mechanical ventilation is an es-
referred to as conventional ventilation. This kind sential treatment technique that is mainly used in
of ventilation can be given in both invasive and the ICU settings of hospitals. Provision of optimal
non-invasive forms. In conventional mechanical ventilatory treatment can expedite weaning and
ventilation, the respiration frequency can be se- prevent prolonged ventilation therapy. Such treat-
lected to be close to the natural breathing rate of ments not only will improve the quality of care
the patient [1]. The methods used in conventional and help reduce medical complications associ-
ventilation are many, and it is not the purpose of ated with prolonged mechanical ventilation such
this article to describe those modalities. The focus as incidents of ventilator-associated pneumonias,
of this article is on newer automated modalities of but they will significantly reduce the costs of ICU
conventional ventilation that have impacted ven- ventilatory treatments at the same time.
tilatory treatment in recent years. Automatic control of this technology can be
used to reduce errors associated with the treat-
Importance of Automation in Conventional ment. Several innovative automatic ventilation
Positive Pressure Mechanical Ventilation techniques have been employed in mechanical
In the past few decades, mechanical ventilators ventilators to this date [1]. Those technologies
have become more advanced with many options and their strengths and weaknesses are discussed
and modalities. The purpose of offering various below.
options in these machines is to make them more
responsive to patients’ needs and thereby improve Advanced Mechanical Ventilation Technologies
treatment. However, the majority of advanced ven- There have been many advanced features includ-
tilators are still open-loop controlled machines in ed in mechanical ventilators in recent years. In
which the main outputs of the ventilator are man- a mode called VAPS, which is available in Bird
ually set by an operator. In using these ventilators, 8400STi model, the ventilator’s output is con-
the clinician needs to know various options and trolled automatically during each breath to assure

Automatic Control of Mechanical Ventilation Technologies 29


the delivery of a target volume set by the clini- in different phases of treatment. Many research-
cian. In this mode, the first portion of the breath ers have developed systems for automatic control
is pressure-limited. However, if the target tidal of patient’s oxygenation by closed-loop control of
volume cannot be delivered, inspiration is con- FiO2, PEEP, or both. Examples of such systems
tinued based on a peak flow setting to deliver have been reviewed [3]. A number of other re-
the target volume to the patient. In another tech- searchers have designed automatic systems for
nique called ATC that is incorporated in a num- adjusting ventilation, respiration rate, or control
ber of different ventilators including the Drager of weaning, and in some more recent systems
Evita series, the Viasys Avea ventilators, and the automatic control of ventilation or weaning has
Nellcor Puritan Bennett 840 model, the ventilator been combined with closed-loop adjustment of
is designed to compensate for the imposed addi- PEEP and/or FiO2 [3].
tional work of breathing due to the endotrache- Despite many attempts by a number of re-
al tube. By using ATC, the pressure drop across searchers to develop automatic ventilation sys-
the endotracheal tube is calculated based on the tems for control of the main outputs of ventila-
measured flow and the tube characteristics, and tors, only a few of such systems have been used in
the ventilator compensates for that pressure drop. practice to this date. Those major automatic sys-
Use of ATC reduces the work of breathing and tems are: (i) ASV used in Hamilton Medical ven-
makes it easier for the patient to start breathing tilators; (ii) PAV (or PPS) used in several different
on his own. While VAPS is designed to assure de- ventilators including Drager Evita series, Puritan
livery of adequate ventilation to the patient and Bennett 840 model, and Respironics BiPAP ven-
to prevent the untoward effects of hypoxemia and tilators; (iii) SmartCare offered by Drager Evita
hypercapnia, ATC is designed to facilitate wean- ventilators, and (iv) NAVA designed to use the pa-
ing patients from mechanical ventilation. These tient’s own neural respiratory drive that is adapted
techniques like other innovative modes of venti- in Maquet ventilators. An overview and discus-
lation such as synchronized intermittent manda- sion of these commercially available systems are
tory ventilation, pressure-regulated volume con- provided below.
trol and many others have been used in practice
in ventilators for some time and are designed to
improve mechanical ventilation treatment and Major Automatic Ventilation Technologies
expedite weaning.
Despite the application of automation in vari- Adaptive Support Ventilation
ous forms in today’s ventilators and their many ASV is a closed-loop technique for automatic ad-
advantages, still the main outputs of these ma- justment of tidal volume and respiratory rate in
chines such as tidal volume, respiratory rate, min- mechanical ventilation. This technique was in-
ute ventilation, FiO2, and PEEP are manually set vented in 1980s and was described as one of the
by clinicians in most advanced modes of venti- embodiments of a patent issued in 1991 [1, 4].
lation. Therefore, using such techniques still re- ASV is used by Hamilton Medical ventilators.
quires the clinician to make the important choices Figure 1 shows a schematic block diagram of this
about the treatment by trial and error. However, system.
due to the advancements in sensor technology In the ASV mode, the required minute ventila-
and the pressing need for more automation of tion is calculated from an input called %MinVol
ventilation, there have been many attempts by a and the patient’s ideal body weight. The patient’s
number of researchers to more aggressively con- respiratory mechanics are measured on a breath-
trol the main outputs of ventilators automatically by-breath basis and provided to the controller to

30 Tehrani
% minvol

ASV Mechanical
Ideal body weight To patient
algorithm ventilator
Control signals for
PEEP inspiratory
pressure and
respiratory rate

pressure, and respiratory


Patient volume, flow,

mechanics data

From patient
Volume, flow,
and respiratory
mechanics monitor

Fig. 1. Schematic diagram of the ASV system.

calculate the required tidal volume and respira- to be manually adjusted in this mode. ASV does
tory rate of the patient. In this mode, if the pa- not provide continuous automatic adjustment of
tient triggers the breaths and can breathe sponta- minute ventilation, and PEEP and FiO2 are manu-
neously, the machine provides additional pressure ally controlled.
support to meet the calculated tidal volume tar-
get. For passive patients, the ventilator delivers Proportional Assist Ventilation
the required tidal volume by using pressure con- PAV is a patented technique in which the venti-
trol ventilation at the calculated optimal rate of lator measures the patient’s ongoing volume and
breathing. The optimal frequency of breathing in rate of flow of inspiratory gas and applies ad-
this method is calculated on a breath-by-breath ditional pressure support in proportion to the
basis by using the measured respiratory mechan- patient’s own inspiratory effort [5]. Some varia-
ics data to minimize the respiratory work rate. By tions of this mode are offered in Puritan Bennett
providing a more natural breathing rate to the pa- 840 ventilators as PAV+, in Drager Evita series as
tient, ASV aims at reducing asynchrony between PPS, and as a non-invasive mode in Respironics
the machine and the patient and thereby to stim- BiPAP ventilators. Figure 2 shows a schematic
ulate spontaneous breathing and expedite wean- block diagram of PAV. Proper use of this tech-
ing. The expiration time in ASV is also adjusted nique requires constant and accurate measure-
when necessary to make sure that lungs are effec- ment of respiratory elastance and airway resis-
tively emptied during expiration and the buildup tance in order to prevent a runaway situation.
of intrinsic PEEP is prevented. ASV can be used In that case, the PAV controller of figure 2 also
for passive as well as active patients and a mini- receives the respiratory mechanics data and pro-
mum required ventilation can be guaranteed by cesses that along with volume and flow rate data
using this mode. Despite the advantages of ASV, to adjust the level of pressure support to the
still several main outputs of the ventilator need patient.

Automatic Control of Mechanical Ventilation Technologies 31


Volume and
PAV Mechanical
flow rate
controller ventilator
monitor
Inspiratory Volume Control To
gas from and signal patient
patient flow rate for
data inspiratory
pressure

Fig. 2. Schematic diagram of the PAV system.

Patient’s measured
end-tidal PCO2

Patient’s measured Smart Care Mechanical


To patient
respiratory rate controller ventilator
Pressure
support control
Patient’s measured
signal
tidal volume

Fig. 3. Schematic diagram of the SmartCare system.

The main advantage of PAV is the synchrony a schematic block diagram of this system. In this
between the patient and the machine. This syn- technique which was first introduced in 1992 [6],
chrony is quite significant since it tends to reduce three patient parameters are monitored by the
the fighting that occurs between the ventilator ventilator, namely tidal volume, respiratory rate,
and a spontaneously breathing patient. Therefore, and the end-tidal pressure of carbon dioxide. The
by providing this synchrony, PAV can give more system’s function is to keep these variables within
comfort to the patient and expedite weaning. a predefined ‘comfort zone’. Using SmartCare, the
PAV is fundamentally a weaning method and ventilator that operates in the pressure support
cannot be used for passive patients. Its proper op- mode, increases the level of support incremental-
eration requires accurate measurement of respi- ly if respiratory rate increases beyond a prescribed
ratory mechanics during spontaneous breathing. range, or tidal volume decreases below a certain
It is suited to patients with relatively strong spon- level, or end-tidal pressure of CO2 increases above
taneous activity and should be watched carefully an acceptable value. If the measured values of these
for ventilatory leaks. PEEP and FiO2 need to be three parameters remain within their predefined
manually adjusted in this mode. ranges, then the level of support is reduced incre-
mentally until the patient is ready to be extubated.
SmartCare This system provides automated support during
SmartCare is a weaning technique available in weaning and is designed to expedite the proce-
Drager Medical Evita ventilators. Figure 3 shows dure by using established protocols. The system in

32 Tehrani
Edi signal
from patient
NAVA
Mechanical
filter and To patient
ventilator
amplifier Ventilation
drive signal

Amplifier gain
adjusted by
clinician

Fig. 4. Schematic diagram of the NAVA system.

SmartCare does not take into account the effects develops between the ventilator and a spontane-
of variations in patient’s respiratory mechanics ously breathing patient due to asynchrony, and
on the acceptable ranges of breathing frequency can expedite weaning. NAVA cannot be used in
and tidal volume and there is no guarantee in the all patients and its effectiveness may be affected if
system that the patient receives adequate minute strong sedatives are administered. The insertion of
ventilation during weaning. PEEP and FiO2 are a nasogastric tube is an additional required inva-
manually adjusted in this mode. sive procedure in this mode that needs to be done
with care and precision [9]. The amplification fac-
Neurally Adjusted Ventilatory Assist tor applied to the Edi signal which determines the
NAVA is a fundamentally different ventilato- level of support provided by the ventilator is ad-
ry mode compared with the other methods de- justed manually by the clinician. PEEP and FiO2
scribed above, in the sense that it uses the patient’s values are also controlled manually in NAVA.
own respiratory neural drive signal to control the
ventilator. The principles of this technology were
first introduced in 1970 [7]. That early technique Conclusion
was enhanced and patented many years later in
1990s [8], and has been adapted in Maquet venti- Significant progress has been made in the design
lators in recent years. Figure 4 shows a schematic and development of automatic ventilation tech-
block diagram of NAVA. In this technique, the pa- niques in the past few decades. Several modalities
tient’s respiratory neural drive signal to the dia- in which some of the main outputs of ventilators
phragm (Edi) is detected by electrodes mounted are automatically controlled are currently in use
on a nasogastric tube which needs to be properly in the ICU settings. When applied properly, these
positioned at the lower esophagus. This signal is technologies tend to improve treatment and help
filtered to eliminate noise due to moving artifacts, facilitate the complex tasks handled by ICU cli-
electrocardiogram, and other disturbances and nicians. Despite these significant improvements,
is amplified by a gain set manually by the clini- there are still some drawbacks associated with the
cian before being applied to drive the ventilator. available automatic techniques of ventilation that
The main advantage of NAVA is the synchrony can be addressed by newer technologies and there
between the machine and the patient which im- are a number of remaining ventilatory tasks that
proves patient comfort, reduces the fighting that can be automated.

Automatic Control of Mechanical Ventilation Technologies 33


Recommendations designed an automatic ventilation system may be,
it is still an aide to the clinician and not a substi-
The following are some recommendations for tute for expert medical care. The automatic tech-
improvement of automation of mechanical nologies that have been and will likely continue to
ventilation: be developed in mechanical ventilation, serve the
• Inclusion of more innovative technologies in purpose of helping to provide more optimal treat-
mechanical ventilation to address the short- ments to patients that can be more responsive to
comings of the present automatic techniques. their needs. These technologies also serve to help
• Emphasizing on technologies that increase clinicians further by constantly monitoring pa-
synchrony between the patient and the tients in a hectic environment and providing the
ventilator without increasing the invasiveness ICU nurses, therapists, and physicians with more
of the treatment. time to give higher attention to the kinds of care
• Incorporation of more innovative sensor that machines cannot provide. Innovations in me-
technologies to prevent measurement errors chanical ventilation technology can improve ICU
during automatic control of ventilation. care by helping to prevent medical errors and un-
• More emphasis on non-invasive ventilation necessary prolongation of mechanical ventilation.
when such treatment is applicable. The innovations in this life-saving technology will
• Inclusion of more effective automatic weaning likely have a significant impact on the quality of
technologies in mechanical ventilation. healthcare as they pertain to ICU treatments in
In summary, there is little doubt that the trend the years to come.
of automation in mechanical ventilation will con-
tinue in the future. However, no matter how well

References
1 Tehrani FT: Automatic control of 4 Tehrani FT: Method and apparatus for 7 Huszczuk A: A respiratory pump con-
mechanical ventilation. Part 2: The controlling an artificial respirator, US trolled by phrenic nerve activity. J Phys-
existing techniques and future trends. J Patent No 4,986,268, issued January 22, iol 1970;210:183P.
Clin Monit Comput 2008;22:417–424. 1991. 8 Sinderby C, Grassino A, Friberg S, et al:
2 Kohn LT, Corrigan JM, Donaldson MS 5 Younes M: Proportional assist ventila- Inspiratory proportional pressure assist
(eds): To Err Is Human: Building a Safer tion, a new approach to ventilatory sup- ventilation controlled by a diaphragm
Health System. Washington, Institute of port. Am Rev Respir Dis 1992;145:114– electromyographic signal, US Patent No
Medicine, The National Academies 120. 5,820,560, issued October 13, 1998.
Press, 2000. 6 Dojat M, Brochard L, Lemaire F, et al: A 9 Barwing J, Ambold M, Linden, N, et al:
3 Tehrani FT: Automatic control of knowledge-based system for assisted Evaluation of the catheter positioning
mechanical ventilation. Part 1: Theory ventilation of patients in intensive care for neutrally adjusted ventilatory assist.
and history of the technology. J Clin units. Int J Clin Monit Comput 1992;9: Intensive Care Med 2009;35:1809–1814.
Monit Comput 2008;22:409–415. 239–250.

Prof. Fleur T. Tehrani, PhD, PE


Department of Electrical Engineering, California State University, Fullerton
800 N. State College Blvd, Fullerton, CA 92831 (USA)
Tel. +1 657 278 2658, Fax +1 714 281 1360
E-Mail ftehrani@fullerton.edu

34 Tehrani
Weaning Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 35–38

Corticosteroids to Prevent Post-Extubation Upper


Airway Obstruction
Scott K. Epstein
Division of Pulmonary, Critical Care, and Sleep Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston,
Mass., USA

Abbreviations failure after removal of the endotracheal tube.


CLV Cuff leak volume
Children are particularly susceptible to post-
UAO Upper airway obstruction extubation UAO because: the larynx is smaller;
fluid easily accumulates in the loose subglottic
submucosal connective tissue, and the cricoid
cartilage ring is less expandable. From 5 to 35%
Extubation failure (reintubation) is associated of patients will experience post-extubation UAO.
with increased ICU mortality, increased length This wide range results from the different patient
of hospital stay, greater need for tracheostomy populations studied, the variability in the defini-
and for long-term acute care, and increased tion of the entity, and the treatments employed
costs [1]. Clinical deterioration between the after extubation (e.g. racemic epinephrine, non-
time of extubation and the re-establishment of invasive ventilation). Most studies use the pres-
ventilatory support may provide the best expla- ence of stridor as a surrogate for UAO or laryn-
nation for increased morbidity and mortality. geal edema. Others will only categorize a patient
Therefore, strategies designed to prevent or treat as having UAO if treatment was required (e.g.
extubation failure have the potential to improve corticosteroids, racemic epinephrine, reintuba-
outcome. tion). Relatively few studies have used direct lar-
yngoscopic assessment of the airway to define
the presence of UAO. Up to 50% of patients who
Prevalence of Post-Extubation UAO manifest post-extubation UAO will require rein-
tubation though in some studies as few as 2–10%
Intubation, with placement of an endotrache- require reinsertion of an airway. Post-extubation
al tube, often results in laryngotracheal injury UAO usually manifests within 8 h of extubation
manifested as inflammation, mucosal ulceration, and almost always within 24–48 h of extubation.
edema, or granuloma formation. If severe, this Numerous factors have been associated with in-
results in glottic or subglottic narrowing lead- creased risk for post-extubation UAO in adults
ing to stridor, respiratory distress, or respiratory (table 1) [2].
Table 1. Factors associated with post-extubation UAO in adults

Female gender (e.g. small tracheal diameter)


Trauma to upper airway
Multiple intubation attempts
Recent unplanned extubation
Age >70–80 years
Excessively mobile or overly large endotracheal tube size
Decreased ratio of patient height to tube diameter (e.g. large tube diameter placed in a short patient)
Ratio of endotracheal tube size to laryngeal diameter >45%
Increased duration of intubation (e.g. >6 days)
Tracheal infection
Low Glasgow Coma Scale score
Excess endotracheal tube cuff pressure
Reduced CLV (<24% of inspired tidal volume or <110 ml)

Detection of Increased Risk for Post- to tidal volume delivered by the ventilator. This ad-
Extubation UAO ditional inspired tidal volume is not measured by
the ventilator leading to a falsely low measurement
Because it is difficult to visualize the trachea in of inspired tidal volume. The resulting difference
the presence of an endotracheal tube, detection of between inspired and expired tidal volume will be
UAO is best performed using the quantitative cuff falsely low. Alternatively, if lung compliance is de-
leak test [3]. During this maneuver the patient is creased, some of the inspired tidal volume imme-
ventilated on assist control (e.g. set tidal volume) diately moves cephalad around the endotracheal
with the endotracheal cuff deflated. The differ- tube rather than entering the lung. Thus delivered
ence (CLV) between inspired and expired tidal inspiratory tidal volume is falsely low resulting in a
volume, averaged over approximately six consecu- falsely low CLV. In this case, the risk for UAO may
tive breaths is then compared. An obstructed up- be underappreciated. Either of these sources of er-
per airway results in similar inspiratory and expi- ror can be eliminated by delivering the machine
ratory tidal volumes while a patent airway results breath with the cuff inflated and then deflating the
in a substantial difference as a large volume of gas cuff just prior to expiration [4].
escapes around the tube. This quantitative cuff leak In pediatric patients the air leak test identifies
is then reported as either a percentage of inspired the pressure (e.g. >30 mm Hg), measured using a
tidal volume or as an absolute CLV. The risk for manometer, at which an audible leak around the
post-extubation stridor and reintubation is higher endotracheal tube occurs. The test has met with
when CLV is less than approximately 12–25% of in- variable success [5].
spired volume or an absolute value of <110–130 ml.
Some patients who appear to have UAO based on
the cuff leak test can nevertheless be successfully Corticosteroids to Prevent Post-Extubation
extubated. This falsely low CLV can occur when se- UAO
cretions adhere to or pool around the external sur-
face of the endotracheal tube. Another explanation Corticosteroids are a logical therapeutic strategy
is that with the cuff deflated, additional tidal vol- to preventing UAO as inflammation is frequent-
ume may be inspired around the tube thus adding ly the underlying cause. Studies published from

36 Epstein
1989 to 1997 in pediatric and neonatal patients 48 h and had a CLV <110 ml. Patients either re-
demonstrate that corticosteroids reduce post- ceived placebo or four doses of dexamethasone (5
extubation UAO by nearly 40%, and may reduce mg) every 6 h for four doses. Patients were extubat-
the need for reintubation [6]. In contrast, three ed 24 h after the last dose. Corticosteroids resulted
randomized controlled trials in adults, published in reduced post-extubation stridor (10 vs. 28%).
prior to 2000, all found intravenous corticoster- There was no difference in the need for reintuba-
oids administered prior to extubation did not re- tion (2.5 vs. 5%) but a much larger cohort would
duce the incidence of post-extubation UAO or have been necessary to demonstrate benefit given
need for reintubation. Corticosteroids may not the low baseline event rate. An important observa-
have worked in these studies for several reasons. tion is that treatment with dexamethasone led to a
The cohorts studied were not at high risk as rein- significant increase in CLV that persisted 24 h after
tubation rates in the control arms ranged from 0 the last dose (e.g., at the time of extubation).
to 2.6%. Corticosteroids were given immediate- The third trial randomized 761 patients who
ly (30–60 min) prior to extubation and therefore had been intubated for ≥36 h. Although no formal
were not likely to have sufficient time to exert a process was used to identify a high-risk cohort, the
clinically significant anti-inflammatory effect. 22% incidence of stridor in the control group sug-
Lastly, only a single dose of corticosteroids was gests an appropriate study population [9]. These
administered and the doses were relatively low. investigators compared 20 mg of methylpredniso-
Since 2006 there have been three well-con- lone, given every 4 h for 12 h prior to extubation, to
ducted published randomized controlled trials placebo. Corticosteroid pretreatment was associat-
that demonstrate that corticosteroids can effec- ed with decreased risk for post-extubation UAO (3
tively prevent post-extubation UAO and reduce vs. 22%), need for reintubation (4 vs. 8%), and need
the need for reintubation in adults. These stud- for reintubation secondary to UAO (0.3 vs. 4%).
ies differ from the older studies in that a high- Since the publication of these trials a num-
risk cohort was selected and corticosteroids were ber of systematic reviews and meta-analyses have
given at higher doses, multiple doses were deliv- been published. The analysis of Jaber et al. [10]
ered, and administration began at least 12–24 h included five published randomized controlled
prior to extubation. The first study by Cheng et al. trials and two abstracts totaling 1,846 patients.
[7] randomized 128 medical and surgical patients, Overall, corticosteroids significantly reduced the
ventilated for at least 24 h, who were identified risk of stridor (relative risk 0.48) and reintubation
as being at elevated risk for post-extubation UAO (relative risk 0.58) but the benefit was most pro-
based on a CLV <24% of inspired tidal volume. nounced in those deemed to be at high risk based
Three groups were studied: one group received on reduced CLV (stridor 35 vs. 19%, reintubation
40 mg of methylprednisolone 24 h prior to extu- 20 vs. 9%). No clear benefit could be demonstrat-
bation (one injection); one group received 40 mg ed in patients not deemed to be at high risk (us-
of methylprednisolone every 6 h beginning 24 h ing the CLV) or in those who only received corti-
prior to extubation (4 injections), and one group costeroids 1 h prior to extubation. A subsequent
received placebo injections. Corticosteroids sig- meta-analysis included 14 studies of adults, chil-
nificantly decreased the percentage of patients dren and neonates (approx. 2,600 patients) [6].
with stridor (30% placebo, 2% one injection, 7% Overall, corticosteroids reduced post-extubation
four injections) and the need for reintubation (19, UAO and need for reintubation, with results con-
5, 7%). sistent across all three patient populations. The ef-
The second study by Lee et al. [8] randomized fect was most pronounced when administration
86 medical patients who were ventilated for at least occurred at least 12 h prior to extubation. These

Corticosteroids to Prevent Post-Extubation UAO 37


meta-analyses identified no major complications made using the factors in table 1, in the absence
associated with the use of corticosteroids. of reduced CLV, is unknown. Nevertheless, based
on the safety of the published corticosteroid regi-
mens, the threshold for treatment should be low.
Recommendations For adults, 20–40 mg of intravenous methylpred-
nisolone should be administered every 6 h begin-
Corticosteroids should be given to patients at ning at least 12–24 h prior to planned extubation.
high risk for post-extubation respiratory fail- For pediatric and neonatal patients, intravenous
ure. High risk is best assessed by the quantita- dexamethasone 0.25–0.5 mg/kg should be given
tive cuff leak test using a reduced CLV of <24% every 6 h at least 12 h prior to planned extuba-
of the inspired tidal volume or <110 ml. It must tion. Of note, most pediatric/neonatal studies
be remembered that some patients with a reduced have continued corticosteroids for approximately
CLV can be extubated successfully without other 12 h after extubation.
intervention though identifying these patients is
difficult. Whether selection for high risk can be

References
1 Epstein SK: Decision to extubate. Inten- 5 Mhanna MJ, Zamel YB, Tichy CM, et al: 8 Lee CH, Peng MJ, Wu CL: Dexametha-
sive Care Med 2002;28:535–546. The ‘air leak’ test around the endotra- sone to prevent postextubation airway
2 Roberts RJ, Welch SM, Devlin JW: Corti- cheal tube, as a predictor of postextuba- obstruction in adults: a prospective, ran-
costeroids for prevention of postextuba- tion stridor, is age dependent in chil- domized, double-blind, placebo-con-
tion laryngeal edema in adults. Ann dren. Crit Care Med 2002;30:2639–2643. trolled study. Crit Care 2007;11:R72–R79.
Pharmacother 2008;42:686–691. 6 McCaffrey J, Farrell C, Whiting P, et al: 9 Francois B, Bellissant E, Gissot V, et al:
3 Miller RL, Cole RP: Association between Corticosteroids to prevent extubation 12-Hour pretreatment with methylpred-
reduced cuff leak volume and postextu- failure: a systematic review and meta- nisolone versus placebo for prevention
bation stridor. Chest 1996;110:1035– analysis. Intensive Care Med of postextubation laryngeal oedema: a
1040. 2009;35:977–986. randomised double-blind trial. Lancet
4 Prinianakis G, Alexopoulou C, Mamida- 7 Cheng KC, Hou CC, Huang HC, et al: 2007;369:1083–1089.
kis E, et al: Determinants of the cuff-leak Intravenous injection of methylpredniso- 10 Jaber S, Jung B, Chanques G, et al:
test: a physiological study. Crit Care lone reduces. the incidence of postextuba- Effects of steroids on reintubation and
2005;9:R24–R31. tion stridor in intensive care unit patients. post-extubation stridor in adults: meta-
Crit Care Med 2006;34:1345–1350. analysis of randomised controlled trials.
Crit Care 2009;13:R49–R59.

Scott K. Epstein, MD
Office of Educational Affairs, Sackler 317
136 Harrison Avenue, Boston, MA 02111 (USA)
Tel. +1 617 636 2191, Fax +1 617 636 0894
E-Mail Scott.Epstein@tufts.edu

38 Epstein
Weaning Mechanical Ventilation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 39–45

FLEX: A New Weaning and Decision Support System


Fleur T. Tehrani
Department of Electrical Engineering, California State University, Fullerton, Calif., USA

Abbreviations although many techniques have been developed


CO2 Carbon dioxide
to this date [4], most of them have not been im-
FiO2 Concentration of oxygen in the inspiratory gas plemented in commercial ventilators. This may be
O2 Oxygen attributed to lack of training for the systems or
PEEP Positive end-expiratory pressure that they may not have been accessible to clini-
PS Pressure support cians and researchers other than the groups that
developed them. Another drawback of such sys-
tems has been the concern over their noise immu-
Choosing the right ventilatory mode and param- nity and error propagation. Nonetheless, effective
eters for ICU patients can be a challenging task decision support systems as well as closed-loop
for many medical personnel. ICU clinicians are systems for mechanical ventilation can be quite
required to carefully review the patients’ under- useful tools in helping clinicians with ventilatory
lying illnesses as well as many available modes treatment of their patients in the ICU environ-
of ventilation, and set the ventilatory param- ment where correct and timely decision on treat-
eters to deliver optimum treatment to their pa- ment can often make the difference between life
tients in a timely manner. These tasks can be sig- and death.
nificantly simplified by use of effective automatic
ventilation modes, or with the aide of properly
designed decision support systems. Use of ad- Analysis
vanced closed-loop techniques or effective open-
loop advisory systems for ventilation can lead to Choice of Ventilatory Parameters and the
better care for critically ill patients, which can in Weaning Technique
turn result in significant reduction in the mortal- Mechanical ventilation can be provided using
ity and morbidity rates associated with provision various techniques such as negative pressure
of inappropriate or unduly prolonged mechanical ventilation, high-frequency oscillatory ventila-
ventilation. tion, or conventional positive pressure ventila-
Many different techniques for automatic con- tion [5]. Among these techniques, conventional
trol of mechanical ventilation have been devel- positive pressure ventilation provided invasively
oped and there are several such technologies (i.e. by use of an endotracheal tube or a tracheo-
used in commercial ventilators today [1–3]. With stomy tube), or non-invasively (e.g., by use of a
regard to open-loop decision support systems, facial or nasal mask) are the more common types
of ventilation adapted today. Conventional posi- When used as a closed-loop control technique,
tive pressure ventilation can be provided by us- FLEX takes input data from the clinician as well
ing various modalities [2]. Advanced mechanical as from a data monitoring unit that receives pa-
ventilators are designed to provide wide ranges of tient’s data from the ventilator and provides it to
ventilatory parameters in various modes of venti- the FLEX algorithm automatically. The algorithm
lation. Open-loop decision support systems can computes the optimal ventilatory parameters for
be helpful to clinicians to set the required ventila- the patient that include the amount of required
tory parameters manually. Also, automatic venti- ventilation, the peak inspiratory pressure, the rate
lation and weaning technologies that have become of respiration if applicable, the level of PEEP, the
available in recent years can provide valuable as- required FiO2 and any needed adjustment in the
sistance to clinicians by constant monitoring of inspiratory to expiratory time ratio. The system
the patients’ conditions and automatically adjust- then provides control signals to the ventilator in
ing ventilatory parameters in response to their accordance with the computed parameter values
changing requirements. Furthermore, weaning to adjust the machine’s outputs automatically.
from mechanical ventilation can be a difficult task If FLEX is used as an open-loop advisory sys-
especially for hard-to-wean patients. Mechanical tem, the patient and ventilatory data do not need
ventilation can cause serious complications if un- to be provided automatically and can be input
necessarily prolonged, while its premature dis- by the clinician intermittently. In the open-loop
continuation can lead to reintubation which may mode, the system makes treatment recommen-
have detrimental consequences. Therefore, a com- dations to the clinician and does not control the
puterized system that can wean patients safely and ventilator automatically. Figure 1 shows the re-
effectively can be quite helpful to patients and cli- cord window of FLEX in the advisory mode for
nicians and lead to reduction in the ICU stay and an adult patient.
healthcare costs at the same time. Figure 2 shows a flowchart of the FLEX algo-
In this article, the main features of a more re- rithm. In the beginning, the input and internal
cent computerized weaning and decision sup- parameters along with their acceptable ranges are
port system for mechanical ventilation known as defined. At this stage, the input data is processed,
FLEX are described and a brief discussion of fu- artifact detection methods are used, and errone-
ture trends in this field is provided. ous data is discarded. Also, abstraction techniques
are used to prepare input data for processing. This
data abstraction is needed to prevent provision of
FLEX Features inappropriate outputs or abrupt changes in venti-
lation to the patient.
FLEX is a computerized system for mechanical At the next step, the required optimum venti-
ventilation that can be used as an open-loop advi- lation and respiratory rate are computed. This is
sory system or as a closed-loop control technique. done based on patient’s ideal body weight, his/her
It can be used in positive pressure ventilation in- temperature, patient’s blood CO2 and O2 levels,
cluding invasive and non-invasive modes. It can and respiratory resistance and compliance data.
be applied in volume control/assist as well as pres- The patient’s CO2 data which can be obtained
sure control/assist modalities in the management by end-tidal CO2 monitoring is optional for adult
or weaning phases of ventilation. FLEX can be and pediatric patients but needs to be provided
used for adult and pediatric patient populations for neonates. The patient’s O2 level is monitored
and a modified version of the system can be em- by using pulse oximetry, the respiratory mechan-
ployed in neonatal ventilatory treatment. ics data is monitored and provided to the system

40 Tehrani
Fig. 1. A patient’s record window of FLEX when used as an advisory system.

continuously or intermittently based on the mode in a mode of ventilation known as adaptive sup-
of ventilation, and the patient’s ideal body weight port ventilation. The system further adjusts the
and temperature are input by the clinician. The inspiratory to expiratory time ratio if necessary
patient’s respiratory dead space is either mea- to prevent the build up of intrinsic PEEP based on
sured or estimated by using empirical equations. the patient’s respiratory mechanics data.
The algorithm uses the above-mentioned data to In the next step of the algorithm, the opti-
compute the required alveolar ventilation and the mal values of FiO2 and PEEP are computed. This
optimum rate of respiration which is found to is done to prevent hypoxemia, hyperoxemia,
minimize the work rate of breathing. The tech- barotraumas, and reduction in the cardiac out-
nique of optimizing the respiratory rate and tidal put. If the system is used in a closed-loop mode,
volume to minimize the breathing work rate is a a fine proportional- integral-derivative control
patented technology [6] that has been available scheme can be added to the algorithm to adjust

FLEX: A New Weaning and Decision Support System 41


START
A

Define the input and internal parameters and their ranges, discard
erroneous data due to artifact. Smooth the input data for processing.

Compute the required ventilation and the optimum repiratory rate for minimum respiratory work rate based
on patient’s oxygen and carbon dioxide levels in the blood, respiratory mechanics data, patient’s ideal body
weight and temperature. Apply safety rules to the results and adjust the expiration time if necessary.

Compute the required values of PEEP and FIO2.

Calculate the required peak inspiratory pressure and apply safety rules.

Define the threshold levels for spontaneous breathing rate and tidal
volume based on the computed optimal values.

Check the patient’s blood gases, the strength of spontaneous effort, and the present values of PEEP and FIO2.

Yes
Are the patient’s conditions
acceptable for weaning?

Weaning can start and the


No level of mandatory
ventilation can be reduced.
Yes
Are the patient’s
conditions alarming?
B
Generate an alarm.
No

Weaning is not allowed No


Full mechanical Has weaning already started?
ventilation is needed.

Yes
B
Continue with weaning, but increase the level of mandatory ventilation.

Send the output messages to display. If in the automatic mode, send the control signals to the ventilator.

If in the automatic mode, wait for a predefined period and then go to A Otherwise, wait until reset.

Fig. 2. A flowchart of the FLEX algorithm.

42 Tehrani
FiO2 and PEEP [7]. Next, the required inspirato- In one of the more recent of these technologies
ry pressure is computed and safety rules are ap- known as neurally adjusted ventilatory assist ven-
plied. Then the threshold levels of tidal volume tilation [9], the patient’s own respiratory neural
and respiratory rate are defined based on the com- drive signal is used to drive the ventilator. This
puted required values. These threshold levels are signal which is detected by electrodes mount-
needed in later steps to determine the strength of ed on a nasogastric tube positioned at the lower
the patient’s spontaneous breathing and to assess esophagus of the patient, needs to be effectively
his/her readiness for weaning. Next, the program cleared of artifact noise and amplified by a man-
checks a series of conditions including the pa- ually controlled gain factor to control the venti-
tient’s blood gases, the strength of his/her spon- lator. Use of the patient’s own respiratory drive
taneous effort, and the levels of FiO2 and PEEP signal provides good synchronization between
to determine whether the patient is ready to be the machine and the patient’s respiratory system.
weaned. If the patient’s conditions are acceptable, However, this technique cannot be used in all pa-
weaning is allowed and is started at the discretion tients, administration of neural depressants can
of the clinician by reducing the level of ventilato- interfere with the treatment, and the requirement
ry support. Otherwise, weaning is not started but of having a well-positioned nasogastric tube is
the program checks to see whether the patient’s not always convenient especially in non-invasive
conditions are alarming and also if weaning has ventilation. In another technique known as pro-
already begun. If the patient’s conditions are not portional assist ventilation [10] the pressure ap-
alarming but the ventilatory treatment is already plied by the ventilator is controlled by and fol-
in the weaning phase, the level of support is in- lows the pressure developed by the patient’s own
creased. However, if the patient’s conditions are respiratory system. This technique is suitable for
found to be alarming, warnings are generated and use in the weaning phase of ventilatory treatment
full mechanical ventilation is provided. and cannot be used for passive patients or those
This procedure is repeated automatically at whose spontaneous breathing effort is not reason-
prescribed intervals in the closed-loop mode and ably strong. Proper application of this technique
is performed intermittently at the discretion of which can be employed in both invasive and non-
the clinician in the open-loop advisory mode. invasive ventilation requires measurement of re-
The details of the control scheme and the math- spiratory elastance and airway resistance in spon-
ematical equations and procedures of the algo- taneously breathing patients.
rithm have been described in detail elsewhere [8], FLEX is a system which is designed to be flex-
and therefore are not repeated here for brevity. ible for use in a wide range of ventilatory modes.
This system which is the subject of a new patent
application can be used in the management phase
Discussion of treatment for passive patients as well as wean-
ing phase by application of PS. It incorporates the
Various open-loop techniques for adjustment of features of a patented mode known as adaptive
ventilatory parameters have been introduced in support ventilation by automatically controlling
the past [4]. Also, a number of technologies for tidal volume and respiratory rate and augments
automatic control of mechanical ventilation have the features of that mode by closed-loop control
been developed that are designed to control pa- of several additional ventilatory parameters in-
tients’ ventilation, oxygenation, or weaning [1– cluding minute ventilation, PEEP and FiO2. In
3]. Several automatic ventilatory techniques are the weaning phase of treatment, using modes
already available in commercial ventilators [3]. such as PS, FLEX controls the pressure applied by

FLEX: A New Weaning and Decision Support System 43


the machine automatically while adjusting PEEP needs of the patient in mind. Although non-
and FiO2 at the same time. It can be used in non- invasive ventilation is becoming increasingly
invasive modes of ventilation such as continuous popular, clinical indications should be clearly
positive airway pressure, or bilevel positive airway observed to choose between invasive and non-
pressure by controlling the PS level while improv- invasive ventilatory treatments. Likewise, in
ing oxygenation by automatically adjusting FiO2. choosing a ventilatory mode or an automatic
FLEX has shown good potential in closed-loop technology, it is essential to know what kind
setups as well as open-loop clinical evaluations of patients the technique is intended for, for
[8, 11]. However, more extensive clinical assess- example (1) whether the mode is restricted to
ments are needed to evaluate the strengths and weaning or if it can be used for passive patients
weaknesses of the system for different patient pro- as well; (2) whether the ventilatory mode is
files and populations. targeted towards certain patient profiles such as
those with acute respiratory distress syndrome
or chronic obstructive pulmonary disease, or it
Recommendations can be used for various patient groups.
• Before using an automatic ventilatory technique,
Application of automation in mechanical ventila- it is necessary to know what parameters it
tion has gained increasing momentum in recent is designed to control automatically, which
years and this trend will likely continue in the fu- inputs are monitored and used by the system,
ture. Despite the existence of several major auto- and whether certain aspects of ventilation
matic techniques in commercial ventilators, most or parameters need to be carefully watched
ventilatory parameters are still manually adjusted. during the treatment.
It is likely that more parameters for control of ven- • It is important to note that no automatic
tilation, weaning, or oxygenation will be automat- technique is designed to substitute for the
ically controlled using newly developed modes in expert clinical care. Rather, such systems are
the near future. The automatic techniques offer designed to assist the clinicians to choose
clear advantages over manual systems and are de- better treatments for their patients. No matter
signed to prevent clinical errors, improve health- how well an automatic system might have been
care and reduce costs. However, the following developed, careful attention to the conditions
points may be worth considering in choosing and of the patient and his/her response to treatment
using such systems: are essential for a successful outcome.
• Every system has its advantages as well as
drawbacks and needs to be chosen with the

References
1 Branson RD, Johannigman JA, Camp- 3 Tehrani FT: Automatic control of 5 Pilbeam SP, Cairo JM: Mechanical venti-
bell RS, et al: Closed-loop mechanical mechanical ventilation. Part 2: The lation, physiological and clinical applica-
ventilation. Respir Care 2002;47:427– existing techniques and future trends. J tions, ed 4. St Louis, Mosby Elsevier,
451. Clin Monit Comput 2008;22:417–424. 2006, pp 81–103.
2 Chatburn RL: Classification of ventilator 4 Tehrani FT, Roum JH: Intelligent deci- 6 Tehrani FT: Method and apparatus for
modes: update and proposal for imple- sion support systems for mechanical controlling an artificial respirator. US
mentation. Respir Care 2007;52:301– ventilation. Artif Intell Med 2008;44: Patent No 4,986,268, issued January 22,
323. 171–182. 1991.

44 Tehrani
7 Tehrani FT: Method and apparatus for 9 Sinderby C: Neurally adjusted ventila- 11 Tehrani FT, Abbasi S: Evaluation of a
controlling a ventilator. UK Patent Serial tory assist (NAVA). Minerva Anestesiol computerized system for mechanical
No 2423721, granted October 14, 2008 2002;68:378–380. ventilation of infants. J Clin Monit Com-
(patent pending in several other coun- 10 Younes M: Proportional assist ventila- put 2009;23:93–104.
tries). tion, a new approach to ventilatory sup-
8 Tehrani FT, Roum JH: FLEX: a new com- port. Am Rev Respir Dis 1992;145;114–
puterized system for mechanical ventila- 120.
tion. J Clin Monit Comput 2008;22:121–
130.

Prof. Fleur T. Tehrani, PhD, PE


Department of Electrical Engineering, California State University, Fullerton
800 N. State College Blvd, Fullerton, CA 92831 (USA)
Tel. +1 657 278 2658, Fax +1 714 281 1360
E-Mail ftehrani@fullerton.edu

FLEX: A New Weaning and Decision Support System 45


SectionTitle
Applied
Section Title
Technologies in Specific Clinical Situations
Section
TechnologyTitlein Sleep Pulmonary Disorders
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 46–50

Thermal Infrared Imaging during


Polysomnography: Has the Time Come to
Unwire the ‘Wired’ Subjects?
Jayasimha N. Murthya ⭈ Ioannis Pavlidisb
aDivisions of Pulmonary, Critical Care and Sleep Medicine, University of Texas Health Science Center, and
bDepartment of Computer Science, University of Houston, Houston, Tex., USA

Abbreviations fatalities associated with motor vehicle accidents


ATHEMOS Automated thermal monitoring system
[3]. Untreated SDB is also associated with cardio-
BCI Bayesian credible interval vascular diseases [4].
κ (kappa) Chance-corrected agreement Diagnosis of sleep apnea typically involves an
Pn Nasal pressure – airflow monitoring device overnight sleep study with simultaneous monitor-
routinely used in polysomnography ing of airflow channels (Pn, oro-nasal thermis-
REM Rapid eye movement tor), electrocardiogram, sleep staging by electro-
RERA Respiratory effort-related arousals
encephalography, electrooculography, and chin
SDB Sleep disordered breathing – encompasses
a wide variety of disorders characterized by and leg electromyography in accordance with
breathing abnormalities during sleep such as the Level 1 recommendations of the American
obstructive sleep apnea, central sleep apnea, Academy of Sleep Medicine [5]. However, con-
etc.
ventional polysomnography has the potential to
TIRI Thermal infrared imaging
interfere with the ‘usual’ sleep pattern. A sizeable
proportion of patients and normal volunteers
who have never had a prior sleep study, experi-
Untreated SDB is a very prevalent problem and ence the ‘first night effect’ characterized mainly
can cause daytime sleepiness as well as have far- by a decrease in sleep efficiency, prolongation of
reaching socioeconomic and health-related con- sleep-onset time, increase in REM sleep latency
sequences. A summary of statistics from three and a reduction in the total amount of REM sleep
pooled studies estimated that 20% of adults with [6]. Additionally, it has been demonstrated that
a body mass index between 25 and 28 kg/m2 have instrumentation during polysomnography affects
SDB based on an apnea-hypopnea index of ≥5 per body position during sleep [7] and thus impacts
hour [1]. Sleepiness is believed to have played a ma- the diagnosis of SDB, the severity of which can
jor role in causing disasters such as the Chernobyl increase during supine sleep. Moreover, bedside
and the Three Mile Island tragedies [2] and has manipulation of sensors, in an effort to obtain
been reported to be the most common cause of high-quality data, may further disturb sleep and
hinder our ability to obtain a true representation to the detection of subtle airflow abnormalities.
of the patient’s usual sleep pattern. Thus, decreas- TIRI, on the other hand, is an array (imaging sen-
ing subject contact with monitoring equipment sor) and not just a point sensor-like thermistor.
and testing in the ‘usual’ sleep environment may Therefore, it can be used to obtain a thermistor-
help counteract polysomnography’s interference like signal across time by averaging the thermal
with sleep and SDB. signal from all the points in the cross-sectional
American Academy of Sleep Medicine recom- area of the visible nares, as well as provide thermal
mends the use of thermistor as the airflow sensor information over an extended two-dimensional
for the diagnosis of apnea and Pn for the diagno- surface. Analysis of signal as an evolving two-di-
sis of hypopnea as well as RERA [5]. While Pn can mensional surface across increases the sensitivity
overestimate pathological events, especially when of TIRI and may increase its ability to detect sub-
subjects change from nasal to predominantly oral tle airflow-related phenomena.
breathing, the thermistor on the other hand lacks Thus, on theoretical grounds alone, one would
the sensitivity as compared to Pn, to detect subtle expect TIRI to be considered a ‘virtual thermis-
flow abnormalities such as hypopnea and RERAs. tor’ to perform at least as good as the oro-nasal
Non-contact airflow monitoring technology such thermistor [8]. Despite these inherent advantages
as TIRI has the capability to monitor airflow dur- of TIRI as compared to the thermistor, the follow-
ing sleep without subject contact. This concept ing significant challenges had to be overcome to
can be extended to include both laboratory-based realize the translation of this technology for air-
and home-based sleep studies. flow monitoring:
(1) Virtual probing: Since there is no physical
probe involved in TIRI, a computational method
Description of TIRI is needed to segment the nostrils in the image, thus
creating a ‘virtual’ probe, where the measurement
The principle of operation of TIRI is close to that can be performed. The difference in the tempera-
of the oro-nasal thermistor in that both these ture of inspired and expired air brings about a tem-
methods sense thermal radiation. However, it is poral variation in the thermal signature of the nos-
important to note the following differences be- trils. This fluctuating thermal signal helps in the
tween the thermistor and TIRI: differentiation of the nostrils (or mouth, if the sub-
(1) TIRI can sense thermal information at ject is mouth-breathing) from the rest of the facial
a distance (contact-free), while the thermistor tissue. The nostrils can thus be segmented from
needs to be physically placed and tethered to pa- the face with the nasal cartilages forming ‘colder’
tient’s oro-nasal area, in the proximity of the ther- boundaries around the thermal signal (fig. 1).
mal signal. (2) Tracking: Because there is no tethering and
(2) TIRI acquires thermal information through the subject is free to move, a computational meth-
natural radiation from the source as opposed to the od is needed to track changes in position, so that
thermistor which acquires thermal information the virtual probe stays always in place and the ac-
by conduction when placed in the path of airflow. curacy of measurement is preserved. Functional
Theoretically, conduction is bidirectional and has imaging such as TIRI that maps randomly chang-
an associated measurement error based on where ing thermophysiological function in a non-re-
and when the measurements are made. The accu- strained subject requires a collaborative network
racy also depends on the intensity as well as the of particle filter trackers based on advanced sta-
magnitude of change in the thermal signal that tistics (coalitional tracking [9]) to compensate for
is being measured. This error can be detrimental subject movement.

Thermal Infrared Imaging during Polysomnography 47


Boundaries

Fig. 1. Temporal variance of nos-


tril region in thermal imagery dur-
ing breathing: (a) inspiration phase,
a b c
(b) transition phase, (c) expiration
phase, and (d) thermal color map.
Note the cold boundaries (labeled
‘Boundaries’) of the nasal cartilages
d 25.55 °C 35.92 °C
that allow the segmentation of nos-
trils from the rest of the facial tissue.

(3) Signal extraction: Assuming that virtual for an average recording time of around 110 min.
probing and tracking work well, they create the There was excellent κ (κ = 0.92, 95% BCI 0.86,
opportunity for a good measurement, but not the 0.96; probability of κ being ≥0.70 [pκ] = 0.99) be-
measurement in itself. Since there is no physical tween TIRI and thermistor for the detection of
transducer that can produce an electronic sig- apnea (defined as a ≥90% decrease in airflow for
nal in response to thermal signal, the signal has at least 10 s) and hypopnea (defined as a decrease
to be computationally generated. This has been in airflow signal by at least 50% from the baseline
achieved by continuous wavelet transformation with a ≥4% oxygen desaturation from pre-event
on the normalized thermal signal under the as- baseline). Likewise, there was a high degree of κ
sumption that the breathing component is the between TIRI and Pn (κ = 0.83, 95% BCI 0.70,
strongest part of the varying thermal signal. 0.90; pκ = 0.98). When the performance of therm-
istor, Pn and TIRI was compared, the thermistor
missed the most number of concordant events de-
Integration of TIRI with Polysomnography tected in the other two channels, while Pn missed
the least. However, it is intriguing to note that TIRI
The integrated hardware and software system missed only 3 concordant events detected by the
we used in our preliminary study [10] is called thermistor and Pn while the thermistor missed 21
ATHEMOS. The thermal signal acquired by the concordant events detected by TIRI and Pn. The
infrared camera and processed by ATHEMOS as better performance of TIRI as compared to the
an airflow signal was recorded into an existing thermistor can be explained by the increased ef-
polysomnography system as an airflow channel, ficiency of detection of thermal signal by natural
using a custom-made digital to analog converter. radiation, as opposed to conduction.
This allowed for easy display and comparison of
all the airflow channels on a single screen. The
camera was kept at about 2.45 m away from the Discussion
patient during the recording.
Fourteen subjects (9 men and 5 women) with- There appears to be need to further advance the
out sleep apnea and 13 subjects (7 men and 6 field of polysomnography with the use of non-
women) with obstructive sleep apnea were studied contact-sensing methods in laboratory and home-

48 Murthy ⭈ Pavlidis
based sleep studies. The need to obtain a true rep- signal acquisition can still continue if the nostrils
resentation the usual sleep pattern motivates this are within the camera frame. In such a scenar-
shift in paradigm. Non-contact-sensing methods io, the operator can remotely change the camera
such as TIRI have the potential to advance the field position on a tilt-pan stand, redefine the region
in this direction. However, this technology should of interest and resume data collection. Extreme
be thoroughly tested prior to its widespread clini- changes in body position can also be handled by
cal use. Even though the results of our initial eval- interfacing multiple cameras at different positions
uation of TIRI appear to be optimistic, significant in the room. Only when a subject buries his or her
concerns in the study design, including a small face into a pillow or pulls a sheet over the face, will
sample and a limited monitoring period, remain. the signal be lost. This scenario will need a techni-
TIRI, to our knowledge, has never been evaluated cian intervention to instruct the patient appropri-
during an overnight polysomnography. A study ately. On a similar note, masks also interfere with
of this nature is of vital importance to establish thermal signal acquisition and thus TIRI cannot
the resilience and accuracy of thermal imaging as be used simultaneously with a continuous posi-
airflow-sensing method during routine nocturnal tive airway pressure titration at the present time.
sleep studies. Even if contact airflow-sensing de- TIRI has the potential to make a significant im-
vices are replaced by non-contact technology, we pact in pediatric polysomnography, where contact
still have to contend with the remaining contact oro-nasal sensors are difficult to place and main-
sensors that may continue to interfere with sleep tain. Sterilization of equipment or consumables is
during polysomnography. On the other hand, the not necessary for TIRI to operate.
role of TIRI in conducting home sleep studies Future studies should validate TIRI in labora-
should be explored and validated. Since TIRI is tory-based nocturnal polysomnography in adults
currently a prototype, the expense associated with and children. Since the accuracy of TIRI in the de-
this technology is relatively high. However, with tection RERAs and its resilience during overnight
further development and widespread use of this polysomnography are still uncertain, this tech-
technology, here is potential for cost reduction. nology at the present time should only be used for
Analysis of thermal imaging signal using al- investigational purposes.
gorithms that preserve the array structure of the
sensor may help in the detection of subtle airflow
abnormalities such as RERAs. In such a situa- Recommendations
tion, direct comparison of TIRI with Pn in scor-
ing RERAs would be necessary to further validate • There is a need to develop non-contact-sensing
this technology. modalities such as TIRI during polysomno-
TIRI is the only technology that can perform graphy to not only improve patient comfort
signal acquisition in a retrospective manner since and experience, but also to obtain a represent-
signal transduction is done computationally. The ative sample of the subject’s usual sleep.
operator can redefine a new region of interest and • Even though the preliminary study with TIRI
computationally transduce the thermal signal. is encouraging, further validation is required
This concept can be expanded to simultaneously before this technology can be used in a clinically
monitor airflow from the oro-nasal region and ar- meaningful way.
tificial airways such as tracheostomies. The cur- • TIRI has the potential to be used for medical
rent tracking algorithms automatically compen- applications beyond polysomnography where
sate for mild to moderate subject movement. In airflow monitoring in a non-contact manner
the event of a drastic change in subject position, is necessary.

Thermal Infrared Imaging during Polysomnography 49


References
1 Young T, Peppard PE, Gottlieb DJ: Epide- 4 Takama N, Kurabayashi M: Influence of 8 Fei J, Pavlidis I: Thermistor at a distance:
miology of obstructive sleep apnea: a untreated sleep-disordered breathing on unobtrusive measurement of breathing.
population health perspective. Am J the long-term prognosis of patients with IEEE Trans Biomed Eng 2010;57:988–
Respir Crit Care Med 2002;165:1217– cardiovascular disease. Am J Cardiol 998.
1239. 2009;103:730–734. 9 Dowdall J, Pavlidis I, Tsiamyrtzis P:
2 Mitler MM, Carskadon MA, Czeisler CA, 5 Iber C, Ancoli-Israel S, Chesson A, et al: Coalitional tracking. Comput Vis Image
et al: Catastrophes, sleep, and public The AASM Manual for the Scoring of Underst 2007;106:205–219.
policy: consensus report. Sleep 1988;11: Sleep and Associated Events: Rules, Ter- 10 Murthy JN, van Jaarsveld J, Fei J, et al:
100–109. minology and Technical Specifications, Thermal infrared imaging: a novel
3 Young T, Blustein J, Finn L, et al: Sleep- ed 1. Westchester/IL, American Acad- method to monitor airflow during poly-
disordered breathing and motor vehicle emy of Sleep Medicine, 2007. somnography. Sleep 2009;32:1521–1527.
accidents in a population-based sample 6 Tamaki M, Nittono H, Hayashi M, et al:
of employed adults. Sleep 1997;20:608– Examination of the first-night effect
613. during the sleep-onset period. Sleep
2005;28:195–202.
7 Metersky ML, Castriotta RJ: The effect of
polysomnography on sleep position:
possible implications on the diagnosis of
positional obstructive sleep apnea. Res-
piration 1996;63:283–287.

Jayasimha N. Murthy, MD
Divisions of Pulmonary, Critical Care and Sleep Medicine
University of Texas Health Science Center
6432 Fannin MSB 1.274, Houston, TX 77030 (USA)
Tel. +1 713 500 6828, Fax +1 713 500 6829, E-Mail Jayasimha.Murthy@uth.tmc.edu

50 Murthy ⭈ Pavlidis
Applied Technologies in Specific Clinical Situations
Technology in Cardiopulmonary Resuscitation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 51–52

Cardiopulmonary Resuscitation with the


Boussignac System
Georges Boussignac
Antony, France

More than 600,000 people suffer a cardiac arrest Analysis


every year in Europe and the USA. This pathol-
ogy has a very severe prognosis. Only 3–5% of Ideal ventilation must be (i) automatic and (ii)
the people who suffer a cardiovascular accident help fill the lungs during decompression in order
survive. to avoid barotraumas. This is the concept of the
Guidelines (ILCOR/AHA, 2005) for cardio- CPR Boussignac system, which uses positive pres-
pulmonary resuscitation (CPR) have advanced sure to fill oxygen into the lungs during decom-
considerably in the last 30 years: (i) in the 1980s pression. During each compression, the O2 ad-
there were 5 compressions for 1 ventilation; (ii) in ministered is expelled towards the outside because
the 1990s there were 15 compressions for 2 ven- the CPR Boussignac system is an open system. In
tilations, and (iii) nowadays the ratio is 30 com- each compression the lung receives an insufflation
pressions for 2 ventilations, and once the patient without needing a ventilator or an oxygen cylin-
is intubated a constant external cardiac massage der facemask. Also, being an open system a risk of
follows at 10–12 V/min. barotraumas is not possible. This concept is that
Recommendations have advanced more and of passive oxygenation. Passive oxygenation ad-
more towards the predominance of the massage ministered by the CPR Boussignac system can be
considered the best way of maintaining correct performed by a specially designed intratracheal
hemodynamics. However, two problems arise: tube or by a supraglottic device that improves the
(1) making the blood flow without oxygen is not interesting form of the gaseous interchanges [1] as
an interesting option if we want to guarantee sur- well as hemodynamics [2, 3].
vival of the vital organs that are great consumers
of oxygen, and (2) the type of present ventilation
(oxygen cylinder facemask or ventilator) is not Discussion
adapted to chest compression (desynchronization
massage/ventilation can induce a barotrauma). For several years, application of a positive pressure
It will be interesting to find a new way to give during CPR has been rejected for being a factor of
oxygen to patients with CPR and maintain or im- reduction of the venous return (reperfusion). The
prove hemodynamics. studies of Steen et al. [2] have demonstrated that
the concept of passive oxygenation for the CPR during CPR; (iii) there is 15–20 cm of water in the
Boussignac system did not reduce the venous re- lung during compression and –5 to –8 cm during
turn, but otherwise it was improved. These same decompression, and (iv) in the mouth, pressure
studies in an animal model have demonstrated variability is very low with between 2 and 3 cm
a 50% survival rate using classic ventilation and of water.
a 100% survival rate using passive oxygenation
according to Boussignac. A study carried out in
Belgium has demonstrated that the association Recommendations
of continuous chest compressions with a cardiac
compressor with the concept of passive oxygen- • Simplification of the attention to the patient:
ation by the CPR Boussignac system has made only massage.
it possible to obtain a survival rate of 26.3% as • Defibrillate as soon as possible.
opposed to 10% obtained with continuous chest • Use passive oxygenation to supply the patient
compressions combined with classic ventilation. oxygen.
To conclude: (i) a patient with cardiac ar- • Execute all these steps as quickly as possible
rest must be considered as an inert body; (ii) in (<10 min).
the thoracic cavity there is a very important ΔP

References
1 Brochard L, Boussignac G, Dubois 2 Steen S, Liao Q, Pierre L, Paskevicius A, 3 Gillis M, Keirens A, Steinkamm C, Ver-
Randé JL, Harf A, Bertrand C, Sjöberg T: Continuous intratracheal belen J, Muysoms W, Reynders N: The
Geschwind H: Cardiopulmonary resusci- insufflation of oxygen improves the effi- use of Lucas and the Boussignac tube in
tation without a ventilator using a novel cacy of mechanical chest compression- the prehospital setting. ERC Congress,
endotracheal tube in a human. Anesthe- active decompression cardiopulmonary Ghent 2008.
siology 1990;72:389. resuscitation. Resuscitation
2004;62:219–227.

Georges Boussignac, MD
1, rue de Provence
FR–92160 Antony (France)
Tel. +33 6 60 99 12 83, E-Mail georgesboussignac@orange.fr

52 Boussignac
Applied Technologies in Specific Clinical Situations
Technology in Cardiopulmonary Resuscitation
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 53–59

Respiratory Function Monitoring during


Simulation-Based Mannequin Teaching
Georg M. Schmölzera–d ⭈ Colin J. Morleya,c,e
aNeonatal Services, Royal Women’s Hospital, bThe Ritchie Centre, Monash Institute for Medical Research, Monash University,
cMurdoch Children Research Institute, Melbourne, Vic., Australia, dDivision of Neonatology, Department of Paediatrics, Medical
University, Graz, Austria, and eDepartment of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Vic., Australia

Abbreviations and-mask’ ventilation, the evaluation of how well


PEEP Positive end-expiratory pressure
the trainee applies the mask and ventilates the
PIP Peak inflation pressure mannequin is subjective.
PPV Positive pressure ventilation Facemask leak is both a common and unrec-
RFM Respiratory function monitor ognised problem during PPV which can lead to
tE Expiratory time failure of ventilation. Recently, a RFM was used
tI Inflation time to evaluate ‘bag-and-mask’ ventilation during
VT Tidal volume simulation-based mannequin teaching [3–6].
V Te Expiratory tidal volume Participants had large and unrecognised leaks be-
V Ti Inspiratory tidal volume
tween the face and the mask [6]. When the par-
ticipants used a RFM they were able to adjust the
mask position and facemask leak was halved [4].
Adequate ventilation remains the cornerstone During resuscitation the spontaneous VT or
of respiratory support during neonatal transi- the VT being delivered during PPV is unknown.
tion [1]. Internationally agreed consensus state- Various studies suggest a VT within a range of
ments on neonatal resuscitation recommend that 4–8 ml/kg. Insufficient VT can lead to inad-
breathing should be assisted by giving PPV with equate gas exchange, whereas excessive VT can
a manual inflation device [2]. However, PPV can lead to volutrauma [7–11]. PPV is always pres-
be difficult because the assessment of facemask sure-limited, however the purpose of applying a
seal, tidal volume (VT) and effective ventilation peak pressure is to inflate the lung with an ap-
is subjective relying on clinical impression of ad- propriate VT. However, the VT delivered is not
equate chest rise and an increase in heart rate fixed but dependent on the size, compliance,
[2]. and resistance of the lungs, and the applied pres-
Neonatal life support courses are a mandato- sure. During resuscitation as the lung aerates
ry part of neonatal training in many countries. the VT delivered will change as the infant starts
Although they emphasise the importance of ‘bag- breathing. Recent studies have been shown that
50 tI tE PIP of
20 cm H2O

PEEP of
5 cm H2O
Ventilation pressure
(cm H2O)

0
100
Flow towards the
infant

Gas flow
(ml/s)

Flow away from the


–100
infant
30

1s

Tidal volume VTi


(ml)

VTe

Fig. 1. PPV with a facemask and no leak. PPV with a set PEEP and PIP of 5 and 20 cm H2O. Gas flow curves of infla-
tion and expiration are returning to the baseline. This indicates sufficient inspiration and expiration time. The areas
underneath both inflation and expiration gas flow curves are similar reflecting an equal amount of gas entering and
leaving the lung. The V T curve displays V Ti and V Te, showing an equal volume of gas entering and leaving the lung. In
addition, no leak is present. V Ti has reached a plateau indicating no further gas continues to enter the lung as inflation
is continued.

operators are unable to deliver accurate and ap- teaching. A RFM can assist during mask ventila-
propriate VTs during simulation-based PPV or tion: (i) it can help to identify correct mask hold
during neonatal resuscitation [12, 13]. When the and positioning techniques to reduce leak of gas
participants could see a display of the VT they between the mask and face; (ii) continuously mea-
were able to achieve the desired volume more ac- sure and display the PIP and PEEP, (iii) enable the
curately [12]. operator to adjust the pressure to deliver an ap-
In this chapter we discuss the potential use propriate VT, and (iv) provide an objective assess-
of a RFM during simulation-based mannequin ment of the trainee’s performance.

54 Schmölzer ⭈ Morley
50

Ventilation pressure
(cm H2O)

0
100

Gas flow
(mL/s)

–100
-100 1s VTi VTe
VTe
30

Tidal volume
(ml)

Leak Leak
0

End of inflation and start of expiration

Fig. 2. Airway leak during PPV. The area underneath the inflation gas flow curve is larger than that under the expiratory
gas flow curves. The VT curve displays a larger VTi compared to VTe and leak is shown as a straight line in the VT curve.

Respiratory Function Monitor and displays numerical values for PIP, PEEP, VTi,
VTe, respiratory rate, expiratory minute ventila-
Any RFM which measures and displays airway tion, tI, and tE (fig. 1). The percentage of leak be-
pressure, gas flow and VT can be used during simu- tween mask and face is calculated and displayed
lation-based mannequin training. The flow sensor with the following equation: [(VTi – VTe) ÷ VTi] ×
is placed between the facemask and the ventilation 100 (fig. 2) [3].
device [3–6]. By integrating the flow signal, the VTi All figures were obtained during PPV of a
and VTe passing through the sensor are automat- Laerdal neonatal mannequin (Laerdal Medical
ically calculated. An airway pressure line is con- AS, Stavanger, Norway), which was made inter-
nected to measure and display the PIP and PEEP. nally leak-free. PPV was performed using a round
A RFM continuously displays airway pressure, silicone facemask (Laerdal Medical AS) and a
gas flow and VT waves. In addition, it measures Neopuff Infant T-Piece Resuscitator (Fisher &

Respiratory Function Monitoring during Simulation-Based Mannequin 55


Teaching
50 Correction of mask position

Ventilation pressure
(cm H2O)

0
150

Flow towards equals


Gas flow flow away from the
(ml/s) infant

Less flow away


Flow towards
from the infant
the infant
–150
30
#
1s
Tidal volume *
#
(ml) + *

0
* VTi # VTe + Leak

Fig. 3. Correction of facemask position. During PPV the airway pressure curve fails to achieve the set PIP of 30 cm H2O. A
returning inspiratory gas flow curve to baseline indicates gas flow towards the facemask. In contrast, there is much less
expiratory gas flow indicating a leak around the facemask. The VT curve reflects the gas flow curve and displays a leak of
around 80%. There is almost no gas entering or leaving the lung. A significant reduction in the facemask leak is achieved
after correction of facemask position. Adequate gas flow is entering and leaving the lung and the set PIP is delivered.

Paykel Healthcare, Auckland, New Zealand), a using a resuscitation mannequin that is internal-
continuous-flow, pressure-limited, T-piece device ly leak-free and a flow sensor placed between the
with a built-in manometer and a PEEP valve. facemask and the ventilation device [3–5]. Any
leak will be displayed as the difference between
VTi and VTe or the difference in area of the flow
Mask Hold and Positioning Techniques curve above and below zero (fig. 2) [3].
During PPV the trainee receives constant
During simulation-based mannequin training a visual feedback because the RFM displays the
RFM can be used to teach the best technique of amount of leak and the trainee can adjust the
positioning, holding and ventilating with a face- position of the facemask to minimise leak (fig. 3)
mask (fig. 1). This can be easily demonstrated [4].

56 Schmölzer ⭈ Morley
PIP 50 cm H2O

50 PIP 42 cm H2O

PIP 28 cm H2O PIP 30 cm H2O

Ventilation pressure
(cm H2O)

0
100

Gas flow
(ml/s)

1s

–100
30
VTe = 12 ml/kg
VTe = 6 ml/kg VTe = 7 ml/kg
Tidal volume
(ml) VTe = 2 ml/kg

Fig. 4. Adjusting PIP to achieve appropriate V Ts. Initially, the inflation and expiratory gas flow curves are small; the
measured V Te is approximately 2 ml/kg. An increase in V Te to 6 ml/kg is achieved with an increase in PIP to 42 cm H2O.
The consequence of a further PIP increase to 50 cm H2O is an increase in V Te to 12 ml/kg, which is excessive. Decreasing
the PIP to 30 cm H2O resulting in a delivered V Te of 7 ml/kg.

Assessment of PIP and PEEP and falls rapidly, it can be difficult to see the PIP
and PEEP [17]. During simulation-based manne-
The purpose of applying any inflating pressure quin training with a RFM the trainee can easily
during PPV is to inflate the lungs with an appro- assess the whole pressure wave and the numerical
priate VT to create a functional residual capacity values of PIP and PEEP (fig. 1).
and thereby facilitate gas exchange. With a self-
or flow-inflating bag the pressure delivered is un-
known unless it is measured and displayed using a Adjusting PIP to Achieve Appropriate VTs
manometer [14–17]. The applied pressure is usu-
ally shown on a dial during PPV with a T-piece Once a trainee is able to obtain leak-free facemask
device. Although this is useful, as pressure rises ventilation, he then can concentrate on assessing

Respiratory Function Monitoring during Simulation-Based Mannequin 57


Teaching
the delivered VT by adjusting the inflating pres- neonatal resuscitation, operators were unable to
sure. With a self- or flow-inflating bag they will judge their delivered VTe or facemask leak by look-
learn how hard they need to squeeze and with a ing at chest rise. They showed that the majority of
T-piece device adjust the PIP they should set to operators underestimated their delivered VTe and
deliver an appropriate VT (fig. 4). Although man- facemask leak. A RFM can assist the ‘instructor’ to
nequins are different from infants, the principles objectively assess the provider’s performance dur-
can be learned and then applied during real resus- ing ‘bag-and-mask’ ventilation.
citation [18].

Conclusion
Assessment of the Trainee’s Performance
A RFM can aid during simulation-based manne-
Assessment of a trainee’s performance remains quin training providing a quantitative assessment
a substantial part of any neonatal resuscitation of the trainee’s technique. A RFM can assist dur-
training courses. However, the assessment of ade- ing ‘bag-and-mask’ ventilation: identifying cor-
quate ‘bag-and-mask’ ventilation remains subjec- rect mask hold and positioning techniques, as-
tive. The ‘instructor’ judges mask position, venti- sessment of PIP and PEEP, and adjusting the PIP
lation rate, and chest rise to assess the adequacy to deliver an appropriate VT provide objective as-
of the trainees’ performance of PPV. Recently, sessment of a trainee’s performance.
Schmölzer et al. [13] have shown that during

References
1 Singhal N, McMillan DD, Yee WH, 6 Wood FE, Morley CJ, Dawson JA, et al: 11 Vilstrup CT, Bjorklund LJ, Werner O, et
et al: Evaluation of the effectiveness Assessing the effectiveness of two round al: Lung volumes and pressure-volume
of the standardized neonatal resuscita- neonatal resuscitation masks: study 1. relations of the respiratory system in
tion program. J Perinatol 2001;21: Arch Dis Child Fetal Neonatal Ed small ventilated neonates with severe
388–392. 2008;93:F235–F237. respiratory distress syndrome. Pediatr
2 2005 International Consensus on Car- 7 Bjorklund LJ, Ingimarsson J, Curstedt T, Res 1996;39:127–133.
diopulmonary Resuscitation and Emer- et al: Manual ventilation with a few 12 Kattwinkel J, Stewart C, Walsh B, et al:
gency Cardiovascular Care Science with large breaths at birth compromises Responding to compliance changes in a
Treatment Recommendations. Part 7: the therapeutic effect of subsequent lung model during manual ventilation:
Neonatal resuscitation. Resuscitation surfactant replacement in immature perhaps volume, rather than pressure,
2005;67:293–303. lambs. Pediatr Res 1997;42: should be displayed. Pediatrics 2009 vol,
3 O’Donnell CP, Kamlin CO, Davis PG, et 348–355. 123:e465–e470.
al: Neonatal resuscitation 1: a model to 8 Hillman NH, Moss TJ, Kallapur SG, et al: 13 Schmölzer GM, Kamlin OC, O’Donnell
measure inspired and expired tidal vol- Brief, large tidal volume ventilation initi- CP, Dawson JA, Morley CJ, Davis PG:
umes and assess leakage at the face ates lung injury and a systemic response Assessment of tidal volume and gas leak
mask. Arch Dis Child Fetal Neonatal Ed in fetal sheep. Am J Respir Crit Care during mask ventilation of preterm
2005;90:F388–F391. Med 2007;176:575–581. infants in the delivery room. Arch Dis
4 Wood FE, Morley CJ, Dawson JA, et al: 9 Wada K, Jobe AH, Ikegami M. Tidal vol- Child Fetal Neonatal Ed 2010 (in press).
A respiratory function monitor ume effects on surfactant treatment 14 Oddie S, Wyllie J, Scally A: Use of self-
improves mask ventilation. Arch responses with the initiation of ventila- inflating bags for neonatal resuscitation.
Dis Child Fetal Neonatal Ed tion in preterm lambs. J Appl Physiol Resuscitation 2005;67:109–112.
2008;93:F380–F381. 1997;83:1054–1061. 15 Bennett S, Finer NN, Rich W, et al: A
5 Wood FE, Morley CJ, Dawson JA, et al: 10 Polglase GR, Hillman NH, Pillow JJ, et al: comparison of three neonatal resuscita-
Improved techniques reduce face mask Positive end-expiratory pressure and tion devices. Resuscitation 2005;67:113–
leak during simulated neonatal resusci- tidal volume during initial ventilation of 118.
tation: study 2. Arch Dis Child Fetal preterm lambs. Pediatr Res
Neonatal Ed 2008;93:F230–F234. 2008;64:517–522.

58 Schmölzer ⭈ Morley
16 Hussey SG, Ryan CA, Murphy BP: Com- 17 O’Donnell CP, Davis PG, Lau R, et al: 18 Schmölzer GM, Kamlin COF, Dawson
parison of three manual ventilation Neonatal resuscitation. 3. Manometer JA, et al: Respiratory function monitor-
devices using an intubated mannequin. use in a model of face mask ventilation. ing of neonatal resuscitation. Arch Dis
Arch Dis Child Fetal Neonatal Ed Arch Dis Child Fetal Neonatal Ed Child Fetal Neonatal Ed 2010;95:
2004;89:F490–F493. 2005;90:F397–F400. F295–F303.

Georg M. Schmölzer, MD
Department of Newborn Research, The Royal Women’s Hospital
20 Flemington Road, Parkville, VIC 3052 (Australia)
Tel. +61 3 8345 3775, Fax +61 3 8345 3789
E-Mail georg.schmoelzer@me.com

Respiratory Function Monitoring during Simulation-Based Mannequin 59


Teaching
SectionTitle
Applied
Section Title
Technologies in Specific Clinical Situations
Section
TechnologyTitlein Inhalation Therapy
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 60–66

Technological Requirements for Inhalation of


Biomolecule Aerosols
R. Siekmeiera ⭈ G. Scheuchb
aFederal Institute for Drugs and Medical Devices (BfArM), and bActivaero GmbH, Gemünden, Germany

Abbreviations surface and blood between 0.5 and 1.0 μm which


DPI Dry powder inhaler
is much less than in the bronchial tract where the
MDI Metered dose inhaler deposited substances have to pass a distance of
30–40 μm and more between mucus surface and
blood), low expression of proteases and peptidas-
es when compared to the intestine, perfusion vol-
In the last decades, methods for recombinant syn- ume at rest of about 5 l/min and missing hepatic
thesis of peptides and proteins were developed al- first pass effect (fig. 1, 2) [1–5].
lowing the production of large amounts of these
compounds for treatment of systemic diseases.
Due to their biochemical properties (high mo- General Problems of Inhalation Therapy
lecular weight, hydrophilic properties, sensitivity
against chemicals and proteolytic enzymes) they Problems in inhalation therapy can be caused
require parenteral administration resulting in by the pharmaceutical compound, the nebuliz-
negative effects on convenience and compliance er used or the patient to be treated. Therefore,
of the patients in cases of chronic diseases (e.g. depending on the underlying cause, various ap-
diabetes mellitus). Therefore, inhalant adminis- proaches exist for optimization of the therapy.
tration via the nose or mouth seems to be an al- If the pharmaceutical compound is subject of
ternate method. The best conditions for drug ab- rapid inactivation within the nebulization pro-
sorption are found in the lung periphery (i.e. the cess (e.g. by aggregation and oxidation) or after
alveolar region) making it an important target for pulmonary deposition (e.g. by proteolytic cleav-
inhalant administration of drugs with a systemic age) or it is too large to penetrate the alveolar
mode of action: size of the alveolar surface about membrane, these problems can be solved for ex-
the half of a tennis court (132 m2) depending ample by addition of stabilizers, proteinase in-
on lung distension (much larger than that of the hibitors as well as absorption enhancers [5–7].
nose of about 180 cm2), thin alveolar epithelium Nebulizer-related problems, e.g. low aerosol out-
(thickness in most regions between 0.1 and 0.2 put, inadequate aerosol distribution width or
μm resulting in a total distance between epithelial high variability of the aerosol particle spectrum,
Airway
1–2 17, 18 23
generation
Bronchi Terminal bronchioles Alveoli
3–5 mm diameter 0.5–1 mm diameter 250 μm diameter

8 μm

Aerosol particle
2 μm diameter

58 μm

3 μm 0.07 μm fluid
10 μm

Ciliated cell Brush cell 0.1–0.2 μm


Basal cell
Goblet cell Basement membrane Type I cell

Mechanism of Impaction Sedimentation Diffusion


particle deposition (inertia) (gravity) (Brownian motion)

Fig. 1. Lung epithelium and mechanisms of particle deposition at different sites within the lungs
[3]. Lung epithelial cells of the different lung regions are drawn at their relative sizes. The higher the
number of the airway generation the deeper the particle is inspired into the lung (0: trachea, 1–2:
bronchi, 3–5: bronchioles, 17–18: terminal bronchioles, 19–20: respiratory bronchioles, 21–22: al-
veolar ducts, 23: alveolar sacs). Mechanisms of particle depositions depending on the aerodynam-
ic particle diameters (dae) are impaction (inertia), sedimentation (gravity) and diffusion (Brownian
motion) in bronchi, terminal bronchioles and alveoli, respectively. A typical aerosol particle (dae:
2 μm) contains tens to hundreds of millions of insulin molecules or hundreds of millions/billions
small molecules depending on its physical character (liquid or solid). Solid aerosol particles are too
large to be absorbed in total and must dissolve to release their drugs for absorption. The deeper an
aerosol particle penetrates into the lung the thinner becomes the airway epithelium and the larger
becomes the lung surface. In consequence, the function of the epithelial absorption barrier de-
creases and the absorption increases as a function of the particle penetration depth into the lung.
Typical cells in the bronchi are basal cells which serve as the stem or progenitor cells for the other
epithelial cells in case of injury or apoptosis, ciliated cells which provide the mechanism for mov-
ing the mucus blanket, goblet cells which secrete the mucus and brush cells which are involved in
drug metabolism. The same cells and the mucus layer are also found in the smaller airways but not
as tall. The thinnest absorption barrier is found in lung alveoli. The basement membrane is not a
membrane but an extracellular matrix of different biopolymers to which epithelial cells attach.

can be solved by use of another type of nebuliz- nebulizer and optimization of the breathing ma-
er, e.g. ultrasonic nebulizer or DPI. Finally, pa- neuver [2, 8–10]. However, different problems
tient-related problems (e.g. lung function, pul- may occur together and in consequence the op-
monary morphometry, breathing technique and timal solution has to consider all underlying as-
compliance) can be improved by use of another pects (table 1).

Technological Requirements for Inhalation of Biomolecule Aerosols 61


Systemic Bound
circulation drug

Pulmonary Conductive Gastrointestinal


Inhaled circulation airways tract
drug

Drug Drug
in alveoli in blood

Deposition
upper RT
Pulmonary
deposition Metabolic
degradation products

RT: Respiratory tract Protein or peptide pharmaceutical

Fig. 2. Uptake of inhaled drugs after peripheral/alveolar deposition [1]. Most of the drug deposit-
ed in the alveoli is absorbed into the blood. Minor proportions are the subject of local degradation
and transport into the conductive airways. From the latter the most is swallowed and degraded in
the gastrointestinal tract and only a minor proportion is absorbed into the blood.

Physical Methods for Aerosol Administration Dry Powder Inhalers

Inhalant drug administration requires high In DPIs, aerosols are produced by disaggregation
efficiency of drug delivery, reproducible dosing, of preformed (e.g. milled or spray-dried) micron-
targeted delivery of the inhaled drug to the site ized particles. The energy required for disaggrega-
of action, ease of device operation, short dura- tion is supplied by the inhalation maneuver or al-
tion of treatment, minimized risk to the patient ternatively by means of an external energy source
and the medical personnel, environmental pro- [2, 12]. The advantages of DPIs are the environ-
tection and cost-effectiveness [11]. A number of mental sustainability (due to a propellant-free de-
products have been developed for this purpose. sign) and the ease to use (not much patient coor-
However, there are strong differences regarding dination is needed). However, the disadvantages
their suitability for nebulization and adminis- are the dependency of the deposition efficiency
tration of the various compounds. In the past, on the patient’s inspiratory airflow, the potential
often low rates of pulmonary drug absorption for dose uniformity problems and the relative high
were achieved because the nebulizers used were complexity and costs for development and manu-
not qualified for production of an adequate aero- facture. DPIs are established for treatment of asth-
sol particle spectrum and the breathing patterns ma and chronic obstructive pulmonary disease by
of the patients were not taken into account [4, means of β-mimetics, anticholinergics or steroids,
7, 10]. but up to now there is only little experience on

62 Siekmeier ⭈ Scheuch
Table 1. Problems in aerosol therapy and their solution [2, 4, 5, 8–10]

Source of the problem Type of the problem Solution of the problem

Compound Formulation: Optimization of manufacturing:


Chemical/physical stability Refinement of production processes
Aerodynamic diameter Addition of stabilizing excipients
Adhesion force Improvement of galenics:
Electrical charge Addition of absorption enhancers
Biology: and proteinase inhibitors and packing
Permeability into particles
Metabolism
Safety
Immunogenicity

Device Low aerosol output Vibrating mesh nebulizers


Aerosol distribution width Dry powder inhalers (DPIs)
Variability of aerosol particle spectrum Metered dose inhalers (MDIs)
Long treatment times for Vibrating mesh nebulizers
drug solutions

Patient Lung function Optimization of nebulizers (including


Lung morphology their handling)
Pulmonary diseases Standardization of the breathing maneuver
Breathing maneuver
Compliance

inhalant administration of biomolecules except techniques, fast dissolving dry powders might be
insulin (Exubera®) for systemic treatment [1, 12, considered as carriers for nanoparticles [14].
13]. This is caused by specific problems for the use
of proteins or peptides occurring in the processes
of lyophilization or spray drying, micronization, Metered Dose Inhalers
completeness of dispersion and disaggregation as
well as the surveillance of the latter. In MDIs, compounds are dissolved or suspended
In passive systems the inspiratory air flow of in a pressurized propellant (nowadays typically
the patient is an essential parameter. If it is not hydrofluoroalkanes) which has to be nontoxic,
sufficient for complete disaggregation, large ag- noninflammable, compatible with drugs formu-
gregates are inhaled which cannot reach the al- lated as suspensions or solutions and has appropri-
veolar region. On the other hand, a high air flow ate boiling points and densities. Consistent dosing
rate increases oropharyngeal deposition which is requires a constant vapor pressure throughout the
also followed by a reduction of pulmonary aero- product’s life. After its release with high velocity
sol deposition. Furthermore, humidity can be a the mixture rapidly expands forming an aerosol.
large problem because it impairs protein stabil- Therefore, MDIs often require spacers for optimi-
ity and also affects disaggregation and dispersion zation of aerosol deposition [2, 15]. Aerosols from
[2, 12]. However, if the underlying problems espe- MDIs are well established in clinical treatment
cially in particle engineering are solved by novel of patients with asthma or chronic obstructive

Technological Requirements for Inhalation of Biomolecule Aerosols 63


pulmonary disease (e.g. by β-mimetics, parasym- residual volume of medication left in the device
patholytics and steroids) for about 50 years [2, 15]. (cost economic effect) and a breath-actuated
Unfortunately, MDIs cannot be used for treatment aerosol delivery which limits the release of aero-
with macromolecules (e.g. peptides and proteins) solized drug into the environment [11]. However,
because a number of prerequisites (stability of the it is difficult to aerosolize suspensions (exception:
drug within storage in the inhaler, no denatur- nano-suspensions) and they sometimes fail when
ation of the drug within the nebulization process, liposomal formulations should be aerosolized.
production of an aerosol with appropriate particle
distribution pattern) are not sufficiently fulfilled.
Novel Devices for Enhanced Pulmonary Drug
Delivery
Nebulizers
Numerous devices are based on the bolus inha-
The appropriateness of nebulizers for adminis- lation technique [9, 10]. For example, MDIs and
tration of macromolecular compounds depends DPIs are supposed to deliver the aerosol cloud at
on their performance (e.g. aerosol output, dis- the beginning of a breath which leads to a more
tribution width and variability of the aerosol efficient lung deposition than an aerosol inhala-
particle spectrum) as well as the stability of the tion over the entire inspiration because the clean
compounds used for nebulization. In air-jet neb- air that follows the aerosol cloud transports the
ulizer’s protein structure and function can be particles deeply into the lungs and extends their
compromised independently from the molecu- pulmonary residence time [9, 10]. The AERx®
lar weight of the protein by surface denaturation, Pulmonary Drug Delivery System of Aradigm
shear-stress-induced denaturation as well as des- (USA) uses a bolus of aerosol particles produced
iccation of the aerosol droplets within the nebuli- by a piston that empties a small liquid reservoir
zation process [2]. However, additives like lipids, into the inhalation air. The release of the bolus
surfactant, amino acids, albumin, polyols as well can be activated during a certain time point dur-
as packing into liposomes may enhance protein ing an inspiration. Other devices, like the AKITA®
stability [2, 16]. Inhalation System (Activaero, Germany) and the
Ultrasonic nebulizers act by a disruption of liq- I-neb® AAD® System (Philips Respironics, The
uid surfaces by means of ultrasound and allow a Netherlands) use liquid nebulizer systems oper-
production of high concentration aerosols [16]. ating only at a certain time during an inhalation
Usually, aerosol particles are produced with such cycle and therefore also use the bolus inhalation
nebulizers which are not appropriate for deep technique to increase pulmonary particle depo-
lung delivery. sition [10]. Currently, the AKITA® technology is
In vibrating mesh nebulizers, a liquid aerosol the most advanced technology as it is the only
with a high fine-particle fraction is produced by one that actively controls the entire inhalation
means of a vibrating mesh or plate with multiple maneuver of the patient. This is done by means
apertures. Aerosols are generated as a fine mist of a positive air pressure delivered by a computer-
without requirement of an internal baffling sys- controlled compressor which is programmed on
tem [11, 16]. In comparison to conventional jet the basis of the patient’s individual lung function
nebulizers and ultrasonic nebulizers, they have data. By Activaero’s SmartCard technology the
a number of advantages which are a higher effi- lung function data can be submitted quickly after
ciency for drug delivery to the respiratory tract, a prior lung function test to the AKITA® device.
an effective aerosolization of solutions, a minimal However, the design has been further improved in

64 Siekmeier ⭈ Scheuch
Table 2. Advantages of individualized controlled inhalation for research and routine therapy

Advantages of individualized Benefit for clinical trials Benefit for outpatient treatment
controlled inhalation

Reduction of side effects Lower number of dropouts Higher quality of life and compliance

Lower lung drug dose variability Reduction in the number of Increase of efficiency
patients to be included

Better drug targeting Increase of efficiency

Electronic compliance control Reduction in the number of Extended feedback for physicians
patients to be included

Higher drug exploitation Lower drug costs

the AKITA-2®, the next generation of this technol- good convenience and compliance of the treated
ogy. The AKITA-2® operates with ultrasonic and patients using the Akita® inhalation device [17].
ultrasonic mesh nebulizers. This new nebulizer is
able to nebulize up to 99% of the filled dose into
particles with mass median aerodynamic diam- Conclusions
eter <4 μm – measured for 0.9% NaCl solution.
The improved understanding of aerosol physics
and mechanisms of pulmonary aerosol deposition
Advantages of Individualized Controlled have been followed by development of modern de-
Inhalation vices for inhalant drug delivery. Both modern de-
vices and optimized breathing patterns are prereq-
The optimization of the breathing maneuver by uisites for optimal pulmonary drug delivery. The
individualized controlled inhalation has a number most recent step for optimization of pulmonary
of advantages. These include clinical trials with drug deposition is the consideration of the indi-
pulmonary drug administration as well as clini- vidual lung function which is a critical factor in in-
cal routine of hospital and outpatient treatment. halation therapy. Likely, future aerosol therapy in
In brief, in clinical studies, improved pulmonary contrast to the past will not be based on a standard
dosage, better drug targeting and reduced side therapy with fixed application volumes and times
effects may result in lower required drug doses, in a large number of patients but on patient’s in-
lower number of dropouts and a lower number of dividual anthropometric data and lung function.
patients to be included which is followed by a rel- This will be followed by a further extension of clin-
evant reduction of costs. Corresponding benefits ical indications for aerosol therapy, e.g. inhalation
for outpatient treatment also include an improved of pharmaceuticals and biomolecules for treatment
efficiency, lower costs and, important for many of local (e.g. tobramycin in cystic fibrosis patients)
patients, a better quality of life (due to lower rate of and systemic (e.g. insulin in diabetes mellitus and
side effects and shorter inhalation times) (table 2). heparin for anticoagulation) diseases.
A number of studies have demonstrated the im-
proved pulmonary drug deposition as well as the

Technological Requirements for Inhalation of Biomolecule Aerosols 65


References
1 Agu RU, Ugwoke MI, Armand M, Kinget 6 Hussain A, Arnold JJ, Khan MA, Ahsan 12 Telko MJ, Hickey AJ: Dry powder inhaler
R, Verbeke N: The lung as a route for F: Absorption enhancers in pulmonary formulation. Respir Care 2005;50:1209–
systemic delivery of therapeutic proteins protein delivery. J Control Release 2004; 1227.
and peptides. Respir Res 2001;2:198– 94:15–24. 13 Siekmeier R, Scheuch G: Inhaled insulin
209. 7 Yu J, Chien YW: Pulmonary drug deliv- – Does it become reality? J Physiol Phar-
2 Niven RW: Delivery of biotherapeutics ery: physiologic and mechanistic macol 2008;59(suppl 6):81–113.
by inhalation aerosol. Crit Rev Ther aspects. Crit Rev Ther Drug Carrier Syst 14 Shoyele SA, Cawthorne S: Particle engi-
Drug Carrier Syst 1995;12:151–231. 1997;14:395–453. neering techniques for inhaled biophar-
3 Patton JS, Byron PR: Inhaling medicines: 8 Byron PR, Patton JS: Drug delivery via maceuticals. Adv Drug Deliv Rev 2006;
delivering drugs to the body through the the respiratory tract. J Aerosol Med 58:1009–1029.
lungs. Nat Rev Drug Discov 2007;6:67– 1994;7:49–75. 15 Newman SP: Principles of metered-dose
74. 9 Scheuch G, Kohlhaeufl MJ, Brand P, inhaler design. Respir Care 2005;50:
4 Wolff RK: Safety of inhaled proteins for Siekmeier R: Clinical perspectives on 1177–1190.
therapeutic use. J Aerosol Med 1998;11: pulmonary systemic and macromolecu- 16 Köhler D, Fleischer W (eds): Theorie
197–219. lar delivery. Adv Drug Deliv Rev 2006; und Praxis der Inhalationstherapie.
5 Siekmeier R, Scheuch G: Systemic treat- 58:996–1008. München, Arcis Verlag, 2000.
ment by inhalation of macromolecules – 10 Scheuch G, Siekmeier R: Novel 17 Fischer A, Stegemann J, Scheuch G,
principles, problems and examples. J approaches to enhance pulmonary deliv- Siekmeier R: Novel devices for individu-
Physiol Pharmacol 2008;59(suppl 6):53– ery of proteins and peptides. J Physiol alized controlled inhalation can opti-
79. Pharmacol 2007;58(suppl 5):615–625. mize aerosol therapy in efficacy, patient
11 Dhand R: New frontiers in aerosol deliv- care and power of clinical trials. Eur J
ery during mechanical ventilation. Med Res 2009;14(suppl IV):71–77.
Respir Care 2004;49:666–677.

PD Dr. med. habil. Rüdiger Siekmeier


Federal Institute for Drugs and Medical Devices (BfArM)
Kurt-Georg-Kiesinger-Allee 3, DE–53175 Bonn (Germany)
Tel. +49 228 207 5360, Fax +49 228 207 5300
E-Mail r.siekmeier@bfarm.de

66 Siekmeier ⭈ Scheuch
Applied Technologies in Specific Clinical Situations
Technology in Inhalation Therapy
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 67–76

Problems and Examples of Biomolecule Inhalation


for Systemic Treatment
R. Siekmeiera ⭈ G. Scheuchb
aFederal Institute for Drugs and Medical Devices (BfArM), and bActivaero GmbH, Gemünden, Germany

Abbreviations Stability of Biomolecules


BAL Bronchoalveolar lavage
G-CSF Granulocyte colony-stimulating factor Independent from the type and the MW of the
GM-CSF Granulocyte-monocyte colony-stimulating biomolecule, aerosolization may result in dena-
factor turation and loss of functionality, for instance by
IL-2 Interleukin-2 oxidation. For example, aerosolization by means
LMWH Low molecular weight heparin of an air-jet nebulizer may cause an inactivation
MW Molecular weight
of recombinant human G-CSF (MW: 18.8 kDa),
interferon-α (MW: 19 kDa) and growth hormone
(MW: 22 kDa), whereas α1-antitrypsin (MW: 51
In recent decades a number of biomolecules, most- kDa), desoxyribonuclease (MW: 30 kDa) and
ly peptides and proteins, have come into the focus heparin (MW: 3–20 kDa) are more stable [1, 2].
of interest for inhalant administration as an alter- For enhancement of stability within the nebuliza-
native of parenteral delivery. Novel inhalation de- tion process, various additives may be added or a
vices and optimization of the breathing technique dry powder aerosol may be used instead of a liq-
enabled researchers to develop methods for the uid aerosol [1, 2].
non-invasive administration of these compounds,
especially for treatment of chronic systemic dis-
orders (e.g. diabetes mellitus, anticoagulation). Physiological Inhibitors of Absorption
However, beside aspects of aerosol delivery and
inhalation, a number of special biochemical and The lung has been exposed to microorganisms
biological aspects must be considered. These in- and foreign substances from the environment
clude the physical and biochemical stability of the for millions of years within the evolution pro-
biomolecules within the nebulization process as cess. In consequence, a complex defense system
well as after pulmonary deposition, and the alveo- has been developed protecting the respiratory
lar absorption of the compound. tract from the nostrils down until the alveoli. In
upper airways and bronchi, defense mechanisms mostly CD4+ lymphocytes) play a crucial role in
consist of anatomic barriers, cough, mucociliary the immunological response after antigen pre-
apparatus, airway epithelium, secretory immuno- sentation by macrophages and dendritic cells.
globulin A, dendritic cell network and lymphoid However, lymphocytes can also phagocytose and
structure [3]. About 90% of inhaled particles with include granules containing proteases and prote-
diameters >2–3 μm are deposited in the central olytic enzymes [1–3]. Type I pneumocytes which
airways on the mucus overlying the cilial epithe- cover about 97% of the alveolar surface (the re-
lium from where they are rapidly transported to maining area consists of type II pneumocytes)
the trachea by means of the mucociliary escalator express carboxypeptidase which degrades a num-
and then swallowed into the gastrointestinal tract ber of peptides and proteins. However, the total
[3–5]. The absorption of biomolecules deposited distance between the respiratory tract and circu-
there is further reduced by the thickness of mucus lation is only 0.5 μm facilitating the diffusion of
layer and respiratory epithelium as well as local gasses as well as penetration and transport of flu-
peroxidases. ids and (inhaled) macromolecules. The latter can
Much better conditions for absorption are pass alveolar epithelium via different transport
found in the lung periphery, i.e. the alveoli [1, 2, mechanisms, which are intracellular tight junc-
6–8]. However, even there a number of defense tions, membrane pores and vesicular transport by
mechanisms exist (fig. 1). The first barriers after type I and type II pneumocytes (fig. 1) [1, 2].
contact are the mucus layer (a complex mixture Another transport mechanism serving for the
of lipids and glycoproteins, but also surfactant exchange of fluids and macromolecules are mem-
from the lower respiratory tract) and the alveo- brane pores. It is assumed that pores of different
lar lining fluid (includes a large amount of sur- sizes exist, which can increase their diameter in
factant with phospholipids and surfactant apoli- case of an existing hydrostatic pressure gradient
poproteins acting as a surface-active substance). [1, 2].
Amount, composition and thickness of the mucus In pneumocytes types I and II another mech-
layer depend on its localization in the respiratory anism of vesicular transport has been described,
tract, inflammatory and neuronal factors where- which is comparable to that in epithelial and en-
as synthesis and release of surfactant from type dothelial cells. In detail, the vesicular transport
II pneumocytes are modulated by hyperventila- mechanism of type I pneumocytes is pressure-in-
tion, endogenous and exogenous factors (phar- dependent and allows the transcellular transport
maceuticals) [1, 2]. Cells located in the respirato- of fluids and macromolecules. However, an esti-
ry tract (mostly macrophages representing about mation of the functional capacity of this transport
85% of cells retrieved by BAL in healthy individu- mechanism is difficult, because (1) the number of
als) also counteract the absorption of deposited vesicles increases in liquid-filled lung indicating
substances. They serve as an unspecific defense their role in the transport of fluids, (2) the glyco-
mechanism (e.g. against bacteria and inhaled calix affects the uptake of proteins via specific or
particles) and act via phagocytosis, secretion of unspecific binding mechanisms and a number of
reactive oxygen species by means of respiratory receptors and binding proteins were identified on
burst and release of mediators of inflammation. capillary endothelia, (3) the definite processing of
However, even granulocytes (about 1–2% in nor- the vesicles inside the cells and the mechanisms
mal BAL) may invade rapidly and serve as potent for their movement (e.g. Brownian movement)
inhibitors of absorption, e.g. by phagocytosis, res- are not conclusively identified, (4) the energet-
piratory burst and secretion of proteases. Last but ic mechanisms of membrane displacement and
not least, lymphocytes (10–20% in normal BAL, fusion of the vesicles are not yet conclusively

68 Siekmeier ⭈ Scheuch
Globular

Air
Surfactant surface
Particle protein

Solution

Surfactant-coated
protein (?)

Alveolar hypophase
Macrophage Denaturated
protein (?)

Transmembrane proteins,
surface proteins,
ion channels,
Released adhesion molecules
protein and others

Proteases and Vesicles (?)


Glycocalix
peptidases

Type I pneumocyte
Cytosol

Interstitial matrix of variable thickness

Endothelial cell
Cytosol
Blood

Endothelial Epithelial Epithelial


Glycocalix surface basal lamina basal lamina

Fig. 1. Barriers for absorption of peptides and proteins after peripheral/alveolar deposition
[1, 2].

Problems and Examples of Biomolecule Inhalation for Systemic Treatment 69


elucidated, and (5) different types of vesicles (e.g. of proteins used in clinical therapy) is between 20
clathrin-coated and clathrin-uncoated) exist, and 50%. However, because of proteolytic degra-
which both play a role in transcytosis, but differ dation the bioavailability of some proteins is much
in respect to their characteristics of protein up- smaller [1, 2, 7–10]. Other variables affecting the
take [1, 2]. absorption are pH value, electrical charge, surface
Surfactant produced from pneumocytes type activity, solubility and stability in the alveolar en-
II together with proteins plays an important role vironment [1, 2, 7, 9]. In hydrophilic compounds
for the clearance of macromolecules by means of (e.g. carbohydrates, peptides and proteins) the
the alveolar lining fluid. Further cellular process- half-life time of alveolar absorption (t0.5) as well
ing can take place with or without binding of the as the time to reach the maximum serum concen-
macromolecules on the cellular surface and de- tration (tmax) increase as a function of their MW
pends strongly on the charge of the molecules. [2, 9, 10].
A large proportion of the material absorbed by
endocytosis from type II pneumocytes is depos-
ited in lamellar bodies. In addition, transcellular Improvement of Macromolecule Absorption
transport represents another mechanism for ab-
sorption of macromolecules [1, 2]. Biomolecules deposited in the alveoli can be ab-
The basal lamina (thickness of about 20–25 sorbed by four distinct mechanisms: phagocyto-
nm) placed below the epithelium predominant- sis by alveolar macrophages, paracellular diffu-
ly consists of glycoproteins (e.g. laminin and fi- sion via tight junctions, vesicular endocytosis or
bronectin) and has an anionic charge on its out- pinocytosis, and receptor-dependent transcytosis
er surface. Presumably, the latter regulates the [1, 2, 8]. Accordingly, the functional role of bar-
permeation dependent on size and charge of the riers and transport mechanisms and their control
molecules. After their passage through the alveo- by physiological and pharmacological factors is
lar wall and alveolar basal lamina, inhaled sub- very different and in consequence a large num-
stances reach the interstitium, where proteins can ber of very different compounds and techniques
be bound by macromolecules or inactivated or for absorption enhancement were investigated [1,
phagocytosed by macrophages or transported to 2, 8, 11], some of which are described below in
the lymphatic system. In the latter case, proteins more detail.
can be detected after some hours in the circula- Even though the activity of proteases and pep-
tion. Endothelial basal lamina and endothelium tidases in the alveolar region is much lower than in
are also barriers for the absorption of macromol- the gastrointestinal tract, proteolytic degradation
ecules. However, compared to the other barriers of susceptible proteins can cause a relevant reduc-
described before they are less effective in inhibit- tion of the bioavailability [9]. Therefore, bioavail-
ing the absorption of biomolecules (fig. 1) [1, 2]. ability can be increased by addition of protease
inhibitors (e.g. nafamostat mesilate, aprotinin and
p-amidinophenylmethanesulfonyl fluoride•HCl).
Factors Affecting the Absorption of However, the effects of the various protease inhib-
Macromolecules itors are very specific to the type of the protease or
peptidase [2, 6, 10, 11].
After alveolar deposition, proteins with a low MW The heterogenous group of surface-active
are absorbed more rapidly than those with a high compounds (e.g. bile acids, fatty acids, non-ion-
MW. The bioavailability of proteins with a MW ic detergents) is assumed to increase the alveolo-
up to 30 kDa (which includes the vast majority capillary transport by an interaction with the cell

70 Siekmeier ⭈ Scheuch
membrane resulting in a liquefaction followed size and chemical properties (charge, MW) of the
by an increased permeability and/or a modula- consisting phospholipids [2, 6, 11].
tion of cellular tight junctions followed by an in- Liposomes are particles (size range from some
creased paracellular permeability. Presumably, nanometers up to a few micrometers) consisting
bile acids increase the absorption by alteration of hydrophobic lipids and phospholipids form-
of the mucus layer, protection of proteins against ing a closed, concentric, bilayer-membrane ves-
enzymatic degradation, disaggregation of pro- icle with a hydrophilic aqueous center [2, 11, 12].
tein multimers, opening of epithelial tight junc- According to this structure, both hydrophobic
tions as well as solubilization of phospholipids and hydrophilic compounds can be packed into
and proteins out of the cell membrane followed liposomes prior to transportation into the lung.
by formation of micelles. However, a strong ab- Hydrophilic compounds (e.g. pharmaceuticals
sorption-enhancing effect can result in damage and larger biomolecules) are entrapped into the
to the epithelial surfaces, especially after treat- vesicle in the inner of the liposome, whereas li-
ment with higher doses and longer treatment pophilic (hydrophobic) compounds are encapsu-
times [2, 10, 11]. lated into the membrane bilayer. Because of their
Cyclodextrins are cyclic polymers of glucose, strong chemical and structural similarity, lipo-
which can form complexes with molecules fitting somes merge with cell membranes and facilitate
into their lipophilic inner structure. The underly- drug delivery into the interior of the cell (fig. 2).
ing modes for absorption enhancement are solu- However, especially small liposomes are also ab-
bilization and complexation of membrane lipids sorbed via cellular phagocytosis [12]. Depending
and proteins of epithelial cells, inhibition of pro- on their structure, liposomes have a high trans-
teolytic enzymes and modification of the physi- port capacity and allow the transport of a large
cochemical properties (e.g. solubility and parti- number of very different compounds. One more
tion coefficient) of the administered substances. characteristic is the sustained release of the com-
However, the toxicity increases with the intensity pounds transported by liposomes [2, 10–12].
of absorption enhancement, both depending on Even though the majority of studies revealed no
the structure of the compound [2, 6, 11]. toxic effect of liposomes, it must be considered
Based on the observation that smaller parti- that both effects, absorption enhancement and
cles are more rapidly phagocytosed than larger lung toxicity, depend on their physicochemical
ones, methods were developed to bind macro- properties (concentration, charge, chain length
molecules to microparticles [6, 9]. For this pur- and MW of phospholipids) [2, 6, 11, 12].
pose, proteins are packed into the inner of bio- Another approach is the modification of pro-
logically degradable polymers or lipids. This teins by fusion to the Fc domain of an IgG1 (IgG
results in a reduction of physiological clearance subtype 1) [2, 13, 14]. In contrast to rodents where
in the alveolar region and proteolytic degradation the expression of the Fc receptor in the gut rapidly
of proteins after phagocytosis by alveolar mac- decreases after weaning and remains low in tis-
rophages. In addition, a sustained release of the sues of adult animals, the Fc receptor in humans
compounds from the microparticles is achieved keeps expressed in several absorptive tissues (e.g.
[9, 10]. Microparticles for drug administration lung, kidney, intestine) even in adulthood. IgG
can be classified into porous particles and lipo- Fc fusion proteins are taken up into epithelial
somes [2, 6, 9–11]. Pharmacological properties cells by pinocytosis. In detail, a coated vesicle is
of porous particles depend on the used materi- formed by invagination of the plasma membrane
al, particle size, porosity and surface structure, entrapping IgG and other solutes in its lumen.
whereas those of liposomes depend on particle Obviously, only a small proportion of IgG binds

Problems and Examples of Biomolecule Inhalation for Systemic Treatment 71


Phospholipid

Liposome Liposome
inclusion

Cell membrane
Outside Membrane
of cell protein

Inside of cell

Fig. 2. Acceptance of a liposome into a cell. Liposomes consist of lipids and


phospholipids [2, 12]. Each phospholipid has a polar hydrophilic ‘head group’
and two hydrophilic ‘tails’. When phospholipid molecules are hydrated un-
der low-shear conditions they spontaneously arrange themselves in sheets
with their heads up and tails down. These sheets then join tails-to-tails and
form a bilayer membrane that encloses water and – if added – water-soluble
compounds (e.g. pharmaceuticals and larger biomolecules) in the center of
the sphere. If liposomes come into contact with cell membranes consisting
of phospholipids, lipids and proteins, the liposome membrane fuses with the
cell membrane facilitating the entry of the encapsulated drug into the inte-
rior of the cell.

to FcRn at the plasma membrane, whereas most alkaline pH value of the interstitial space. Passage
of the binding takes place intracellularly, because of IgG into the circulation is most likely primarily
the majority of FcRn is localized in acidic endo- paracellular because of the absence of tight junc-
somal vesicles inside the cell. The transport ves- tions between endothelial cells. The FcRn recep-
icles containing IgG bound to FcRn do not fuse tor is also responsible for the long half-life time
with degradative lysosomes but rather pass uni- of IgG in the bloodstream, because it protects
directionally through the epithelial cell, driven IgG from degradation. As in epithelial cells, IgG
by the pH gradient between luminal and serosal is taken up from vascular endothelial cells by pi-
exposures of the epithelial cells. As the binding nocytosis. However, in contrast to epithelial cells,
of IgG to FcRn is pH-dependent (tight binding at IgG there is not subject of transcytosis, because
slightly acidic pH), there is a release of IgG from the endocytic vesicles containing IgG bound to
FcRn after fusion of the transport vesicles with FcRn return to the plasma membrane of the en-
the plasma membrane at the basolateral site of the dothelial cells, so that IgG is released back into
epithelial cells because of the neutral to slightly the bloodstream resulting in a recycling process

72 Siekmeier ⭈ Scheuch
for IgG protecting IgG from lysosomal degrada- produced aerosol particles in older nebulizers and
tion. Fc fusion proteins can be efficiently admin- a not sufficiently standardized inhalation maneu-
istered as liquid aerosols and several studies have ver. A more recent study investigated the antico-
demonstrated a good tolerability, a high bioavail- agulative effect (determined by measurement of
ability even of larger proteins (e.g. erythropoietin) the anti-FXa activity) of different doses of inhaled
and an increased half-life time in the circulation certoparin in healthy humans in comparison to
in animals or humans [2, 13, 14]. subcutaneous administration. The study revealed
a rapid onset of the anticoagulative effect after
certoparin inhalation, a decrease of the interindi-
Systemic Treatment with Inhaled vidual variabilities after administration of higher
Macromolecules doses (up to 9,000 IU) when compared to lower
doses, a satisfactory anticoagulative effect after
A number of studies investigated the feasibility inhalation of 9,000 IU and a longer duration af-
of macromolecule inhalation for systemic treat- ter inhalation of 9,000 IU when compared to sub-
ment. A focus was set on hormones (insulin, cal- cutaneous administration of 3,000 IU which was
citonin, growth hormones, somatostatin, thyroid- achieved without side effects [16].
stimulating hormone, and follicle-stimulating Erythropoietin, an erythrogenic growth factor
hormone), growth factors (G-CSF and GM-CSF), (epoetin α: MW: 14.7 kDa; epoetin β, γ, δ, ε, ω:
different interleukins (e.g. IL-2) and heparin (un- MW: 18.2 kDa) is used for stimulation of eryth-
fractionated and LMWH) [6, 15–18]. However, in ropoiesis in the bone marrow, e.g. in patients with
humans, most data are available for insulin, hepa- end-stage renal failure and cancer. Due to its large
rin and IL-2. In the following, some examples, ex- MW the pulmonary absorption without methods
cept insulin which is described elsewhere in this for absorption enhancement is low [2, 14]. An in-
book, are compiled. teresting method based on Fc fusion proteins has
Heparin, an acidic sulfated mucopolysaccha- been developed to improve pulmonary uptake
ride, is characterized by a MW of unfractionated and pharmacokinetics of erythropoietin (and also
heparin between 2.8 and 58 kDa (mean value: 15 other proteins) [2, 13, 14]. In brief, in cynomol-
kDa) and between 2 and 6 kDa in case of fraction- gus monkeys a better absorption of Epo-Fc di-
ated LMWH. Both require parenteral adminis- mers was observed after a shallow breathing pat-
tration and serve as an anticoagulant due to their tern than after a deep inhalation due to the higher
binding to antithrombin III resulting in a confor- expression of Fc receptors in the central airways.
mation change of this protein. Beside this, a lot of After inhalation of the Epo-Fc dimer, the Epo-Fc
other properties of heparin (e.g. interaction with monomer and unconjugated erythropoietin bio-
growth factors, regulation of cell proliferation availabilities of 5%, 35% (which is similar to that
and angiogenesis, modulation of proteases and after subcutaneous administration) and 15% were
antiproteases) are of interest in medical research. observed, respectively. The low bioavailability of
Since 1965, a number of studies had investigated the Epo-Fc dimer was assumed to be caused by
safety and feasibility of the inhalation of heparin its higher MW or steric hindrance of IgG and
and LMWH for anticoagulation. It was found that erythropoietin. Independent from the mode of
inhalant administration was effective, well tolerat- administration (inhalation, intravenous adminis-
ed and not followed by relevant pulmonary or sys- tration) the observed plasma half-life times (t0.5)
temic side effects [16, 19]. However, some studies were higher for the Epo-Fc dimer and the Epo-
showed a strong variability of the anticoagulative Fc monomer than for unconjugated erythropoi-
effect, e.g. due to an inadequate diameter of the etin, demonstrating an increase of t0.5 due to the

Problems and Examples of Biomolecule Inhalation for Systemic Treatment 73


fusion to the Fc residual of the immunoglobin. patients to recover neutrophils (e.g. after induc-
Functional analysis by measurement of reticulo- tion chemotherapy for treatment of acute mye-
cytes in blood revealed that all types of inhaled logenous leukemia, mobilization of hematopoietic
erythropoietin and both fusion proteins were bi- progenitor cells prior to cytapheresis and follow-
ologically active, although there were differences ing transplantation of hematopoietic progenitor
with respect to the strength of the reticulocyte- cells) [23]. However, several other potential clini-
increasing effect [13, 18]. Some further experi- cal indications, e.g. use in antitumor therapy and
ments including measurement of pharmacoki- treatment of alveolar proteinosis, have been inves-
netics after administration of different doses were tigated [23]. The glycoprotein is usually admin-
also performed in healthy human volunteers and istered parenterally but in studies for treatment
confirmed the absorption and a dose-dependent of cancer or alveolar proteinosis it has been ad-
biological effect after inhalation of the Epo-Fc fu- ministered by means of inhalation [18, 23]. The
sion protein [14, 18]. feasibility of aerosol administration of GM-CSF
IL-2 is produced from T lymphocytes after an- was proven in cynomolgus monkeys more than 15
tigen stimulation and serves as an immune mod- years ago [18]. A number of studies investigated
ulator, e.g. activating cytotoxic T cells and natu- the effect of GM-CSF on pulmonary alveolar pro-
ral killer cells making it to an interesting target teinosis which is an orphan disease (less than 500
in tumor research [20]. It was observed that IL-2 reported cases until 2006 and first described in
caused a suppression of metastases of malignant 1958). Since a first case report from 1996, several
tumors, especially metastases from melanoma investigators demonstrated that inhalant admin-
and kidney cancer. In the latter, persisting remis- istration of GM-CSF aerosol alone or if necessary
sions were achieved in a number of patients. In in combination with whole lung lavage is a safe
consequence, IL-2 treatment received approval and efficient therapy for treatment of pulmonary
from the Food and Drug Administration (FDA) alveolar proteinosis even for a longer treatment
for treatment of metastasizing kidney cancer and period resulting in a therapy-dependent improve-
melanoma in 1992 and 1998, respectively [20]. ment of clinical behavior and lung function pa-
Most frequently, IL-2 was subject of systemic rameters [18, 24, 25]. Other studies investigated
administration, however in a number of studies the effect of various study regimens and doses of
there was also an inhalant administration of IL-2 inhaled recombinant GM-CSF on different types
liposomes alone or in combination with other cy- of cancer, e.g. metastases of kidney carcinoma, os-
tokines (e.g. interferon-α). Most data were pub- teosarcoma and melanoma [18]. Usually, inhala-
lished for patients with advanced kidney cancer, tion of lower doses was not followed by relevant
especially those with pulmonary or mediastinal changes of lung function and side effects. In ad-
metastases [20–23]. The inhalation (first-line and dition, there were also only minor increases of
second-line therapy) was well tolerated showing white blood cells and neutrophils under therapy.
fewer side effects than systemic treatment with However, treatment with aerosolized GM-CSF re-
cytokines and was followed by a relevant increase sulted in a relevant disease regression or reduc-
of survival time. Inhalation of high IL-2 doses tion of disease progression in some patients [18].
(second-line therapy) was also followed by tem- In a more recent study, escalating doses of up to
porary regression of pulmonary metastases but 2,000 μg b.i.d. were administered in patients with
not extrapulmonary metastases in patients with lung metastases of melanoma. The investigators
melanoma [21, 22]. observed an acceptable toxicity of inhaled GM-
Treatment with recombinant GM-CSF (MW: CSF, the greatest changes of antitumor immunity
14.6 kDa) has been approved for therapy of in patients receiving the highest drug doses and

74 Siekmeier ⭈ Scheuch
longer times of progression-free survival in pa- induction of specific effects on the target organ
tients developing an immune response [26]. lung in case of hormones) as well as damage of
Cyclosporin A, a cyclic peptide (MW: 1,200 Da) lung epithelium directly (e.g. bile acids, cyclo-
serves as an immunosuppressant for prevention dextrins and other absorption enhancers) or via
of graft rejection in patients after organ transplan- production of reactive oxygen species (e.g. in
tation as well as in patients with autoimmune dis- case of cationic liposomes). Last but not least,
eases [27]. In a number of studies predominantly pulmonary diseases may complicate or prevent
performed in lung transplant recipients, the effect inhalant drug therapy under some circumstances
of cyclosporin liposome inhalation was investigat- [2, 6, 11, 17].
ed [27]. After inhalation the lipophilic compound
is rapidly absorbed. However, the pharmacokinet-
ics indicate a temporary uptake of the compound Conclusions
by alveolar macrophages as well as its interaction
with pulmonary surfactant or membranes of alve- In recent decades, inhalation of biomolecules has
olar epithelium [28]. Depending on the deposited come into the focus of interest. However, prior
lung dose of inhaled cyclosporin A, an improved to studies investigating the inhalation of these
transplant function (determined by measurement compounds, the physical and physiological back-
of the forced expiratory volume in 1 s) was ob- ground for reproducible administration of suffi-
served in lung transplanted patients with graft cient drug doses into the lung had to be elucidated.
rejection. At the same time, patients treated with After solving these basic questions, the feasibility
aerosolized cyclosporin A required lower doses of inhalative administration was investigated for
of other immunosupressants when compared to a large number of biomolecules (mainly peptides
the control group. However, inhalant immuno- and proteins). However, up to now, data regarding
suppressive therapy was well tolerated and there the long-time effects of inhaled macromolecules
were no higher rates of pulmonary infections as except insulin and heparin are sparse. In addi-
well as no hepatotoxic or nephrotoxic effects [29]. tion, there are also few data regarding the feasibil-
More recent data of the same study group dem- ity and safety of carriers (e.g. microparticles and
onstrated that deposition of sufficient pulmonary liposomes) as well as stabilizers and absorption
doses of cyclosporin A can prevent the decrease of enhancers for pulmonary drug administration.
graft function and the occurrence of bronchioli- Therefore, future studies are required for further
tis obliterans (which is largely affecting the long- investigation of long-time effects and optimiza-
time prognosis after lung transplantation) and in tion of inhalative drug administration. Then it is
consequence improve the long-time outcome in likely that inhalation-based methods for drug ad-
lung transplanted patients [30]. ministration will serve as a safe and convenient
alternative of subcutaneous injection in patients
with systemic diseases.
Safety of Macromolecule Inhalation

Safety and tolerability of pulmonary adminis-


tered compounds can be largely different from
those after subcutaneous administration. Inhaled
pharmaceuticals as well as additives for absorp-
tion enhancement may induce an incompatibil-
ity (e.g. immunization in case of proteins and

Problems and Examples of Biomolecule Inhalation for Systemic Treatment 75


References
1 Niven RW: Delivery of biotherapeutics 13 Bitonti AJ, Dumont JA, Low SC, Peters 23 Thipphawong J: Inhaled cytokines and
by inhalation aerosol. Crit Rev Ther RT, Kropp KE, Palombella VJ, Stattel JM, cytokine antagonists. Adv Drug Delivery
Drug Carrier Syst 1995;12:151–231. Lu Y, Tan CA, Song JJ, Garcia AM, Simis- Rev 2006;58:1089–1105.
2 Siekmeier R, Scheuch G: Systemic treat- ter NE, Spiekermann GM, Lencer WI, 24 Seymour JF, Presneill JJ: Pulmonary
ment by inhalation of macromolecules Blumberg RS: Pulmonary delivery of an alveolar proteinosis. Progress in the first
– principles, problems and examples. J erythropoietin Fc fusion protein in non- 44 years. Am J Respir Crit Care Med
Physiol Pharmacol 2008;59(suppl 6): human primates through an immuno- 2002;166:215–235.
53–79. globulin transport pathway. Proc Natl 25 Wylam ME, Ten R, Prakash UBS,
3 Nicod LP: Pulmonary defence mecha- Acad Sci USA 2004;101:9763–9768. Nadrous HF, Clawson ML, Anderson
nisms. Respiration 1999;66:2–11. 14 Bitonti AJ, Dumont JA: Pulmonary PM: Aerosol granulocyte-macrophage
4 Scheuch G, Kohlhaeufl MJ, Brand P, administration of therapeutic proteins colony-stimulating factor for pulmonary
Siekmeier R: Clinical perspectives on using an immunoglobulin transport alveolar proteinosis. Eur Respir J 2006;
pulmonary systemic and macromolecu- pathway. Adv Drug Deliv Rev 2006; 27:585–593.
lar delivery. Adv Drug Deliv Rev 2006; 58:1106–1118. 26 Markovic SN, Suman VJ, Nevala WK,
58:996–1008. 15 Laube BL: The expanding role of aero- Geeraerts L, Creagan ET, Erickson LA,
5 Scheuch G, Siekmeier R: Novel app- sols in systemic drug delivery, gene ther- Rowland KM, Morton RF, Horvath WL,
roaches to enhance pulmonary delivery apy, and vaccination. Respir Care 2005; Pittelkow MR: A dose-escalation study
of proteins and peptides. J Physiol Phar- 50:1161–1176. of aerosolized sargramostim in the treat-
macol 2007;58(suppl 5):615–625. 16 Scheuch G, Brand P, Meyer T, Herpich C, ment of metastatic melanoma: an
6 Agu RU, Ugwoke MI, Armand M, Kinget Müllinger B, Brom J, Weidinger G, Kohl- NCCTG Study. Am J Clin Oncol 2008;31:
R, Verbeke N: The lung as a route for häufl M, Häussinger K, Spannagl M, 573–579.
systemic delivery of therapeutic proteins Schramm W, Siekmeier R: Anticoagula- 27 Corcoran TE: Inhaled delivery of aero-
and peptides. Respir Res 2001;2:198– tive effects of the inhaled low molecular solized cyclosporine. Adv Drug Deliv
209. weight heparin certoparin in healthy Rev 2006;58:1119–1127.
7 Patton JS, Byron PR: Inhaling medicines: subjects. J Physiol Pharmacol 2007;58 28 Burckart GJ, Smaldone GC, Eldon MA,
delivering drugs to the body through the (suppl 5):603–614. Venkataramanan R, Dauber J, Zeevi A,
lungs. Nat Rev Drug Discov 2007;6: 67– 17 Siekmeier R, Scheuch G: Inhaled insulin McCurry K, McKaveney TP, Corcoran
74. – does it become reality? J Physiol Phar- TE, Griffith BP, Iacono AT: Lung deposi-
8 Wolff RK: Safety of inhaled proteins for macol 2008;59(suppl 6):81–113. tion and pharmacokinetics of cyclo-
therapeutic use. J Aerosol Med 1998;11: 18 Siekmeier R, Scheuch G: Treatment of sporine after aerosolization in lung
197–219. systemic diseases by inhalation of bio- transplant patients. Pharm Res 2003;20:
9 Byron PR, Patton JS: Drug delivery via molecule aerosols. J Physiol Pharmacol 252–256.
the respiratory tract. J Aerosol Med 2009;60(suppl 5):15–26. 29 Iacono AT, Smaldone GC, Keenan RJ,
1994;7:49–75. 19 Köhler D: Aerosolized heparin. J Aerosol Diot P, Dauber JH, Zeevi A, Burckart GJ,
10 Yu J, Chien YW: Pulmonary drug deliv- Med 1994;7:307–314. Griffith BP: Dose-related reversal of
ery: physiologic and mechanistic aspects. 20 Eklund JW, Kuzel TM: A review of recent acute lung rejection by aerosolized
Crit Rev Ther Drug Carrier Syst 1997;14: findings involving interleukin-2-based cyclosporine. Am J Respir Crit Care Med
395–453. cancer therapy. Curr Opin Oncol 2004; 1997;155:1690–1698.
11 Hussain A, Arnold JJ, Khan MA, Ahsan 16:542–546. 30 Iacono AT, Johnson BA, Grgurich WF,
F: Absorption enhancers in pulmonary 21 Enk AH, Nashan D, Rübben A, Knop J: Youssef JG, Corcoran TE, Seiler DA,
protein delivery. J Control Release 2004; High-dose inhalation interleukin ther- Dauber JH, Smaldone GC, Zeevi A,
94:15–24. apy for lung metastases in patients with Yousem SA, Fung JJ, Burckart GJ,
12 Dhand R: New frontiers in aerosol deliv- malignant melanoma. Cancer 2000;88: McCurry KR, Griffith BP: A randomized
ery during mechanical ventilation. 2042–2046. trial of inhaled cyclosporine in lung-
Respir Care 2004;49:666–677. 22 Huland E, Heinzer H: Renal cell carci- transplant recipients. N Engl J Med
noma – innovative medical treatments. 2006;354:141–150.
Curr Opin Urol 2004;14:239–244.

PD Dr. med. habil. Rüdiger Siekmeier


Federal Institute for Drugs and Medical Devices (BfArM)
Kurt-Georg-Kiesinger-Allee 3, DE–53175 Bonn (Germany)
Tel. +49 228 207 5360, Fax +49 228 207 5300
E-Mail r.siekmeier@bfarm.de

76 Siekmeier ⭈ Scheuch
Applied Technologies in Specific Clinical Situations
Technology in Inhalation Therapy
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 77–83

Diabetes Treatment by Inhalation of Insulin –


Shine and Decline of a Novel Type of Therapy
R. Siekmeiera ⭈ G. Scheuchb
aFederal Institute for Drugs and Medical Devices (BfArM), and bActivaero GmbH, Gemünden, Germany

In recent decades, techniques for protein produc- in comparison to subcutaneously (sc) adminis-
tion by means of recombinant DNA technology tered insulin in rabbits. The authors observed a
have been developed and largely improved en- glucose-lowering effect and a more rapid onset of
abling us to produce sufficient quantities of pep- action after intratracheal than after sc adminis-
tides and proteins for novel medical treatment tration, but higher drug doses were required [7].
and administration options. Many decades were A first study on inhaled insulin in patients was
required to solve the problems of a reproducible performed by Heubner et al. [8], also in 1924.
and efficient drug dosing by inhalation, even in They reported a dose-dependent effect of insu-
the case of insulin. However, shortly after approv- lin inhalation on blood glucose. Again, the re-
al and market launch, distribution was stopped quired dose was 30 times higher after inhalation.
due to low market penetration. This article briefly Independently from this group, Gänsslen [9] also
describes the history, problems and experience on studied the effect of insulin inhalation in patients.
insulin inhalation until market withdrawal. He also observed that inhaled insulin effectively
lowered the concentration of blood glucose. His
required amounts of insulin were not as high as
History of Insulin Inhalation those described above by Heubner et al. [8]. It
took 46 more years until Wigley et al. [10] pub-
Insulin was isolated in 1921 by Banting and Best, lished their pivotal study of insulin inhalation of-
and 1 year later on January 11, 1922, it was in- fering the proof of principle of this therapy. They
troduced into medical treatment [1–3]. Initially were able to demonstrate that pork-beef insulin
it was the subject of intramuscular injection. administered by a nebulizer caused a prompt in-
However, other techniques for drug application crease in plasma-immunoreactive insulin and
(transdermal, ocular, oral, buccal, nasal, pulmo- that hypoglycemia showed a temporal relation-
nal, rectal, vaginal and transuterine) were investi- ship. However, pulmonary delivered insulin was
gated [4–6]. In 1924 and 1925 – only 2 years after far away from approval and several studies per-
the start of the therapeutic insulin era – the first formed in the next decades were necessary to de-
studies on insulin inhalation were published. The velop this therapy option [6]. For example, devic-
first paper described the effect of intratracheally es were developed for optimized drug deposition
in the alveoli (standard nebulizers mainly de- these particles. However, prerequisites for use of
posit the aerosol in the bronchial system) [1, 11, these excipients are rapid degradation after in-
12]. At about 1990 the technical prerequisites for halation and immunological and toxicological
the pulmonary delivery of insulin were estab- inertness [6, 13].
lished. Based on different technical and pharma- Liposomes are only used experimentally for
cological solutions (e.g. different inhalation sys- pulmonary insulin administration so far, but may
tems, powder aerosol or liquid aerosol), several be an alternative [6, 13–15].
companies developed inhalation devices (table Solid particles (microspheres or large porous
1) [1, 6]. Exubera® from Pfizer/Nektar was the particles) are chemically and physically more sta-
first to receive approval from the US Food and ble than liposomes and allow higher drug loading
Drug Administration and the European Drug [13, 16]. Pharmacological properties of micropar-
Agency in early 2006 for patients with diabe- ticles (size range: <500 nm) depend on the used
tes mellitus types 1 and 2. However, 1 year af- material, preparation technique, particle size, po-
ter marketing start, Pfizer announced it would rosity and surface structure as well as the delivery
withdraw Exubera® from the market in October device [6, 14, 16, 17]. Microspheres can be pro-
2007, citing that the drug had failed to gain mar- duced by a number of distinct methods based on
ket acceptance. A few months later most other supercritical fluid technology, emulsion-solvent
developers stopped their programs on inhaled evaporation, spray-drying and phase separation.
insulin. However, one company, Pharmaceutical Examples for the clinical use of microspheres for in-
Discovery Corp./Mannkind Pharmaceuticals, sulin inhalation are (ProMaxx®, Epic Therapeutics;
continued [6]. Technosphere®, Pharmaceutical Discovery Corp.,
and calcium phosphate-polyethylene glycol parti-
cles, BioSante Pharmaceuticals) [6, 14].
Carriers of Insulin for Inhalation Large porous particles are characterized by
geometric diameters >5 μm, low particle density
Most of the recent pulmonary delivery methods (generally <0.1 g/ml) and aerodynamic diameters
were based on insulin powder aerosols. One ex- <5 μm. They have good flow and aerosolization
ception was (AERx® iDMS) where a liquid pack- properties due to their low aerodynamic diam-
aging was used (table 1). In some of the novel eter. There was one system for insulin inhalation
techniques, insulin is formulated into micro- based on large porous particles (AIR®, Alkermes)
spheres (liposomes, particles, large porous par- [6, 14].
ticles) in order to improve its pharmacological
properties (e.g. inhalation, absorption). Most of
the published clinical data regarding the inhala- Improvement of Insulin Bioavailability by
tion of particles were based on Technosphere®. Addition of Absorption Enhancers
An advantage of microparticles is that they are
phagocytosed less rapidly [1, 6, 13]. Insulin The bioavailability of biomolecules after alveolar
packed in the inner part of biologically degrad- deposition can be improved by addition of com-
able polymers and lipids (microparticles and li- pounds affecting the absorption (e.g. bile acids) and
posomes, respectively) is subject of a slow alveo- compounds inhibiting the proteolytic degradation
lar clearance and peptide degradation by alveolar (e.g. aprotinin). However, many of these substanc-
macrophages resulting in an increased bioavail- es were found to be toxic [2, 6, 13, 16, 18].
ability. There may also be an altered pharma- Only few studies have investigated the ef-
cokinetics which is subject of a slow release from fect of absorption enhancers in humans or other

78 Siekmeier ⭈ Scheuch
Table 1. Devices for inhalant administration of insulin [modified according to 1, 6]: the table reflects the situation in
2006, i.e. before market withdrawal of Exubera®

Trade name Status of Principle or pharmaceutical form


(developer/partner) development

Exubera® Market Dry powder insulin packed into blisters of 1 mg (≈3 U insulin) or
(Nektar/Pfizer) approval 3 mg. Dosing via the number of blisters. Pneumatic release of the
2006 aerosol out of the blister in an inhalation chamber. Particle
diameter of 2.5 μm.

AERx® iDMS Phase III Liquid insulin packed into single strips and dosed in single units.
(Aradigm/ Regulation of breathing maneuver by means of microprocessors and
NovoNordisk) electronic optimization of insulin release within inspiratory flow.
Particle diameter of 1–3 μm.

HIIP® Phase III Dry powder insulin packed into blisters. Mechanical system with breath
(Alkermes/Eli Lilly) activated release of particles. Porous particles of low density with a
geometric diameter of 5–30 μm (aerodynamic diameter of <5 μm).

Technosphere® Phase III Dry powder recombinant insulin combined with a derivative of
(Pharmaceutical diketopiperazine. Self-assembly into an ordered lattice array at low
Discovery Corp./ pH value. Mass median aerodynamic diameter 2–4 μm. Dissolution
Mannkind of particles and insulin release at neutral pH value on alveolar surface.
Pharmaceuticals) Dry powder inhaler and passive disagglomeration (MedTone®).

Microdose DPI® Phase II Dry powder insulin packed into blisters. Disaggregation of drug
(Microdose powder by means of a piezo vibrator. Mass median aerodynamic
Technologies/ diameter approximately 1.5 μm, 84% of the particles <4.7 μm.
Elan Corp.)

Unknown Phase II Dry crystals of a recombinant insulin formulation. Administration by


(Abbott (formerly means of a handheld breath actuated inhaler (BAI) driven by a
Kos Pharma.)/–) propellant.

Aerodose® Phase II Liquid insulin. Administration by means of a breath activated multiple


(Aerogen/–) (stopped dose inhaler. Mean particle diameter of 3.2 μm, 87% of the particles
in 2003) between 1 and 6 μm.

Bio-Air® Phase I Coated dry particles based on calcium phosphate nanoparticle carriers.
(BioSante Administration by means of a calcium phosphate nanoparticulate
Pharmaceuticals/–) delivery system.

Alveair® Phase I Liquid insulin. Administration by means of a generic handheld device


(CoreMed/Fosun delivering inhaled insulin with the same units as injected insulin.
and Xuzhou)

Unknown Phase I Microspheres of recombinant human insulin (ProMaxx®).


(Epic Therapeutics/–) Administration by means of a dry powder inhalation device (Cyclohaler)
>90% insulin. 95% of the microspheres with diameters 0.95–2.1 μm
(mean: 1.5 μm), 95% of the particles <4.7 μm.

Spiros® Phase I Dry powder insulin blisters. Release by means of a hand-held,


(Elan Pharma (stopped battery-driven, multiple-dose inhalator. Novel powder dispersion
(formerly in 2004) system (Spiros-S2) without electromechanical components of the Spiros
Dura Pharma)/–) for administration at low inspiratory flow rates (15–30 l/min).

Diabetes Treatment by Inhalation of Insulin 79


mammals except rats. For example, sodium cit- indicates the existence of a dose-dependent uptake
rate, mannitol and glycine are excipients used in mechanism [6, 18, 22]. A number of studies were
Exubera® [6, 19]. However, even these data are also performed with the insulin derivative lispro.
unequivocal. For example, Heinemann et al. [20] Lower doses of lispro were required to achieve the
observed only a small increase in bioeffectivity same insulin concentration and onset of action
if a powder aerosol of insulin was administered was more rapid. Both effects may be caused by a
in combination with an endogenous bile acid in breakup of the hexamer into monomers.
healthy humans, whereas Johansson et al. [21] Many studies determined relative bioavailabil-
found a strongly increased bioavailability of in- ity by comparing of AUC after inhalation with
sulin in dogs if the substance was administered sc administration. Other investigators calculated
as a fluidic aerosol containing also taurocholate the bioeffectivity (hypoglycemic effect of inhaled
[16, 18]. insulin compared to a defined sc administered
insulin dose). However, both methods result in
an underestimation of the therapeutic effect of
Pharmacokinetics of Inhaled Insulin in the inhaled insulin fraction itself (only a small in-
Individuals without Lung Diseases sulin fraction is deposited in the lung periphery
where it can be absorbed). In controlled studies,
Many studies investigated pharmacokinetics bioavailabilities and bioeffectivities for inhaled
and pharmacodynamics of inhaled insulin in insulin were found between 9 and 22% as well as
healthy subjects and found significant differ- 8 and 16%, respectively. In consequence, the in-
ences caused by inhaler use, administered for- haled insulin doses required to achieve the same
mulations and doses of insulin. After inhalation, therapeutic effect are up to 11 times higher com-
regular insulin is usually absorbed faster than sc pared to that after injection [6, 18]. Only about
administered insulin (time to peak concentra- 30–40% of the filled insulin doses reach the lungs
tion (tmax): 7–90 min vs. 42–274 min [1, 6, 11, and from these over 50% are deposited in the
18, 22]. A biphasic pharmacokinetics has been airways (bronchial system) and underlie muco-
found with a first peak rapidly after inhalation ciliary transport and/or degradation. And from
followed by a slow release. Within the first hour the particles reaching the alveoli, about 40–50%
after administration the area under the concen- are rapidly absorbed into the circulatory system
tration vs. time curve (AUC) is larger for inhaled [6, 18].
insulin than for sc insulin. On the other hand, This shows that deep lung deposition is a ma-
a larger total AUC is observed for sc insulin if jor predictor of bioavailability. The best depo-
an observation period of 6 h is used, suggest- sition will be achieved if the aerosol is inhaled
ing a therapeutic benefit of inhaled insulin in at the beginning of a slow and deep breath, en-
treating of prandial or postprandial hyperglyce- abling the particles to reach the alveolar region.
mia. Additionally, inhaled insulin bears a lower However, not only the total amount of alveolar
risk for postprandial hypoglycemia because of insulin deposition, but also the intraindividual
increased clearance [18]. reproducibility of this therapy is affected by the
Dose-response studies in patients with diabe- individual breathing of the patients. A number
tes mellitus type 1 revealed a widely linear dose of studies have shown a similar or even better
response. However, even when the maximum reproducibility of insulin administration by in-
insulin concentration (Cmax) increased with ad- halation than sc injection. This shows that sc ad-
ministered dose there was an increasing time de- ministration also has a high variability [2, 6, 18,
lay to peak concentration in plasma (tmax), which 22–25].

80 Siekmeier ⭈ Scheuch
Effect of Smoking on the Pharmacokinetics of compared to healthy subjects without chronic ob-
Inhaled Insulin structive pulmonary disease [6]. In consequence,
due to the high frequency of chronic obstructive
A number of studies have described a higher ab- pulmonary disease in the population, its effect on
sorption (up to 3–5 times) of inhaled insulin in insulin absorption should be investigated prior to
smokers compared to non-smokers (higher ab- a re-launch.
sorption (Cmax), AUC and bioavailability as well
as a higher peak concentration (Cmax) and a
shorter time to peak (tmax)) [1, 5, 6, 18, 22, 25, Safety of Inhalant Insulin
26]. Smoking cessation reduced absorption, an ef-
fect which was reversed by smoking resumption The safety of inhaled insulin was subject of many
[6, 26]. These changes are caused by an increased investigations. Most data regarding the long-term
permeability of the alveolo-capillary barrier due tolerability were published for the Exubera® sys-
to chronic cigarette smoking. However, acute cig- tem and the AERx iDMS® system for study pe-
arette smoking significantly blunts the enhanced riods of up to 2 years and more in patients with
insulin absorption. The underlying mechanisms diabetes mellitus types 1 and 2 [2, 6, 18, 22, 25,
of these effects are not understood. It is assumed 27]. Inhalation of insulin caused only a minimal
that inhaled nicotine and pulmonary neutrophils change of spirometric parameters of lung func-
may play a relevant role. Because of these complex tion (e.g. forced expiratory volume in 1 s, forced
effects, inhalant insulin therapy was not approved vital capacity) as well as parameters of diffusion
in current smokers and ex-smokers [6, 19]. capacity for carbon monoxide and blood gas anal-
ysis [1, 2, 5, 6, 18, 25, 27]. However, the manu-
facturer recommended spirometric measurement
Effect of Pulmonary Diseases on the of lung function before treatment, after 6 months
Pharmacokinetics of Inhalant Insulin and thereafter at least annually in the product in-
formation for Exubera® [28]. In principle, the ob-
Because it was assumed that lung diseases may served low pulmonary toxicity may be explained
affect pulmonary drug absorption, such patients by the distribution of the inhaled doses of 4–5 mg
were excluded from routine treatment with in- t.i.d. on a total alveolar surface of about 80–120
haled insulin. However, acute respiratory infec- m2 and the following rapid decrease of its concen-
tions had no relevant effect on pharmacokinet- tration due to absorption and distribution in the
ics and pharmacodynamics in otherwise healthy body fluid as well as proteolytic degradation [1, 2,
individuals [6, 18]. In contrast, asthma patients 5, 6, 18, 23, 24].
showed a mild decrease of Cmax and a distinct de- Beside its strong metabolic effect, insulin also
crease of AUC (bioavailability) and plasma glu- acts as a weak growth factor (effectivity only
cose concentration (bioeffectivity). Furthermore, 1/100 of insulin-like growth factor-1) after bind-
asthma patients showed a higher variability of ing to the receptor for insulin-like growth factor-1
Cmax and AUC after insulin inhalation. In prin- [18]. About 6 months after the end of marketing
ciple, these changes can be caused by increased of Exubera® the Food and Drug Administration
drug deposition in the bronchi. In contrast, pub- published a press release reporting a potentially
lished data on the pharmacokinetics of inhaled increased risk for bronchial cancer in ex-smok-
insulin in patients with chronic obstructive pul- ers treated with inhaled insulin [29]. However,
monary disease are limited and conflicting. It the number of reported cases is extremely small
was found higher or lower absorption of insulin and even though investigations were continued,

Diabetes Treatment by Inhalation of Insulin 81


no relevant data seem to confirm the theory of an (2) repeated pulmonary function tests (both re-
increased lung cancer risk in smokers due to in- sulting in extra costs depending on the required
sulin inhalation. doses [32]), (3) an ongoing requirement of blood
In a number of insulin inhalation studies, in- sampled by finger punction for the measurement
creased serum titers of non-neutralizing IgG an- of glucose concentration, and (4) no relevant im-
tibodies were found which had no therapeutic provement of metabolic control [19, 27, 32]. Based
relevance, i.e. there were no correlations to the on these arguments, the National Institute for
metabolic control, lung function and adverse Health and Clinical Excellence (NICE, UK) and
events [1, 2, 4, 6, 18, 25, 27, 30, 31]. Most likely the the Institute for Quality and Efficiency in Health
induction of these antibodies is caused by insulin Care (IQWiG, Germany) declined funding for
formulation and dose (inhaled insulin is given in inhalant insulin in Great Britain and Germany,
higher doses and more frequently for treatment respectively. It is likely that these decisions were
of postprandial hyperglycemia than sc insulin) as substantial reasons that this therapy option failed
well as the lung as the target of delivery (presence in the market.
of macrophages, dendritic cells and lymphocytes
in the lung) [5, 6, 25, 30, 31].
Mild to moderate cough occurred rapidly after Conclusions
inhalation of dry powder insulin (seconds to min-
utes) in up to 20–30% of patients. However, the More than 80 years after the first studies, inhaled
symptoms were transient, settled with continua- insulin (Exubera®) was approved and introduced
tion of the therapy and seldom resulted in treat- into the market. Marketing has been rapidly
ment withdrawal [6]. stopped because of the unexpectedly low sales. In
In summary, there was no relevant difference the short time of its marketing, Exubera® was ac-
regarding the risk for occurrence of hypoglyce- cepted by the patients (although not reimbursed
mia between inhalant and sc insulin. However, by most health insurance companies) and well
the risk was expectedly higher in patients treat- tolerated without adverse effects. Beside Exubera®
ed with inhalant insulin when compared to those a number of other devices for inhalation of insu-
treated with oral antidiabetics [6]. lin were under development. However, due to the
rapid stop of the distribution of Exubera® and
the consecutive stop of the development of simi-
Acceptance and Costs of Inhaled Insulin lar products by all competitors except Mannkind
Pharmaceuticals, some questions are not com-
The development of pen injector systems has pletely answered. These include the effect of long-
largely improved diabetes treatment in the last time inhalation on lung function and the effect
decades. However, all these medical products are of pulmonary diseases on deposition, absorption
invasive which is important in patients suffer- and pharmacokinetics of inhaled insulin as well
ing from needle phobia (about 10% of patients). as the potentially increased risk for lung cancer in
Therefore, pulmonary administration of insulin ex-smokers. However, if there will be a re-launch
was assumed to be a revolution in the treatment of this interesting non-invasive method of drug
of insulin-dependent diabetes. Indeed, patients administration, the open questions should be fur-
favored the inhaler even though it is more com- ther investigated before.
plex to handle [19, 27]. However, these advantag-
es are opposed by (1) the small bioavailability of
inhaled insulin and higher required insulin doses,

82 Siekmeier ⭈ Scheuch
References
1 Cefalu WT: Concept, strategies, and fea- 14 Cryan SA: Carrier-based strategies for 24 Scheuch G, Siekmeier R: Novel
sibility of non-invasive insulin delivery. targeting protein and peptide drugs to approaches to enhance pulmonary deliv-
Diabetes Care 2004;27:239–246. the lungs. AAPS J 2005;7:E20-E41. ery of proteins and peptides. J Physiol
2 Owens DR: New horizons – alternative 15 Sakagami M, Byron PR: Respirable Pharmacol 2007;58(suppl 5):615–625.
routes for insulin delivery. Nat Rev Drug microspheres for inhalation. The poten- 25 Valente AXCN, Langer R, Stone HA,
Discov 2002;1:529–540. tial of manipulating pulmonary disposi- Edwards DA: Recent advances in the
3 Wilde MI, McTavish D: Insulin lispro: a tion for improved therapeutic efficacy. development of an inhaled insulin prod-
review of its pharmacological properties Clin Pharmacokinet 2005;44:263–277. uct. Biodrugs 2003;17:9–17.
and therapeutic use in the management 16 Hussain A, Arnold JJ, Khan MA, Ahsan 26 Becker RHA, Sha S, Frick AD, Fountaine
of diabetes mellitus. Drugs 1997;54: F: Absorption enhancers in pulmonary RJ: The effect of smoking cessation and
597–614. protein delivery. J Control Release 2004; subsequent resumption on absorption of
4 Belmin J, Valensi P: Novel drug delivery 94:15–24. inhaled insulin. Diabetes Care 2006;29:
systems for insulin. Clinical potential for 17 Agu RU, Ugwoke MI, Armand M, Kinget 277–282.
use in the elderly. Drugs Aging 2003;20: R, Verbeke N: The lung as a route for 27 Ceglia L, Lau J, Pittas AG: Meta-analysis:
303–312. systemic delivery of therapeutic proteins Efficacy and safety of inhaled insulin
5 Heinemann L, Pfützner A, Heise T: and peptides. Respir Res 2001;2:198– therapy in adults with diabetes mellitus.
Alternative routes of administration as 209. Ann Intern Med 2006;145:665–675.
an approach to improve insulin therapy: 18 Patton JS, Bukar JG, Eldon MA: Clinical 28 NDA 21-868/Exubera®: US package
update on dermal, oral, nasal and pul- pharmacokinetics and pharmacodynam- insert, 2006. New York, Pfizer Labs
monary insulin delivery. Curr Pharm ics of inhaled insulin. Clin Pharmacoki- (available from http://www.pfizer.com/
Des 2001;7:1327–1351. net 2004;43:781–801. pfizer/download/uspi_exubera.pdf).
6 Siekmeier R, Scheuch G: Inhaled insulin 19 Guntur VP, Dhand R: Inhaled insulin: 29 Food and Drug Administration Med-
– does it become reality? J Physiol Phar- extending the horizons of inhalation Watch Alert, 2008. Exubera® Inhalation
macol 2008;59(suppl 6):81–113. therapy. Respir Care 2007;52:911–922. Powder (available from http://www.
7 Laqueur E, Grevenstuk A: Über die 20 Heinemann L, Klappoth W, Rave H, drugs.com/fda/exubera-insulin-human-
Wirkung intratrachealer Zuführung von Hompesch B, Linkeschowa R, Heise T: rdna-origin-inhalation-powder-12372.
Insulin. Klin Wochenschr 1924;3:1273– Intra-individual variability of the meta- html).
1274. bolic effect of inhaled insulin together 30 Fineberg SE, Kawabata T, Finco-Kent D,
8 Heubner W, de Jongh SE, Laqueur E: with an absorption enhancer. Diabetes Liu C, Krasner A: Antibody response to
Über Inhalation von Insulin. Klin Care 2000;23:1343–1347. inhaled insulin in patients with type 1 or
Wochenschr 1924;3:2342–2343. 21 Johansson F, Hjertberg E, Eirefelt S, type 2 diabetes. An analysis of initial
9 Gänsslen M: Über Inhalation von Insu- Tronde A, Hultkvist Bengtsson U: Mech- phase II and III inhaled insulin (Exu-
lin. Klin Wochenschr 1925;4:71. anisms for absorption enhancement of bera®) trials and a two-year extension
10 Wigley FW, Londono JH, Wood SH, Ship inhaled insulin by sodium taurocholate. trial. J Clin Endocrinol Metab 2005;90:
JC, Waldman RH: Insulin across respira- Eur J Pharm Sci 2002;17:63–71. 3287–3394.
tory mucosae by aerosol delivery. Diabe- 22 Sakagami M: Insulin disposition in the 31 Stoever JA, Palmer JP: Inhaled insulin
tes 1971;20:552–556. lung following oral inhalation in and insulin antibodies: a new twist to an
11 Niven RW: Delivery of biotherapeutics humans. A meta-analysis of its pharma- old debate. Diabetes Technol Ther 2002;
by inhalation aerosol. Crit Rev Ther cokinetics. Clin Pharmacokinet 2004;43: 4:157–161.
Drug Carrier Syst 1995;12:151–231. 539–552. 32 Black C, Cummins E, Royle P, Philip S,
12 Gonda I: The ascent of pulmonary drug 23 Scheuch G, Kohlhaeufl MJ, Brand P, Waugh N: The clinical effectiveness and
delivery. J Pharm Sci 2000;89:940–945. Siekmeier R: Clinical perspectives on cost-effectiveness of inhaled insulin in
13 Siekmeier R, Scheuch G: Systemic treat- pulmonary systemic and macromolecu- diabetes mellitus: a systematic review
ment by inhalation of macromolecules lar delivery. Adv Drug Deliv Rev 2006; and economic evaluation. Health Tech-
– principles, problems and examples. J 58:996–1008. nol Assess 2007;11:1–126.
Physiol Pharmacol 2008;59(suppl 6):53–
79.

PD Dr. med. habil. Rüdiger Siekmeier


Federal Institute for Drugs and Medical Devices (BfArM)
Kurt-Georg-Kiesinger-Allee 3, DE–53175 Bonn (Germany)
Tel. +49 228 207 5360, Fax +49 228 207 5300
E-Mail r.siekmeier@bfarm.de

Diabetes Treatment by Inhalation of Insulin 83


Section Titlein Diagnosis and Pulmonary Problems
Technology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 84–88

Endobronchial Ultrasound
Devanand Anantham ⭈ Mariko Koh Siyue
Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Singapore

Abbreviations lesions for biopsy. In radial EBUS, a 360° image


EBUS Endobronchial ultrasound
that is perpendicular to the long axis of the airway
EUS Endoscopic ultrasound is produced (fig. 1). The resolution of the image is
TBNA Transbronchial needle aspiration <1 mm and a depth of 40–50 mm is scanned.
In thoracic tumors, EBUS has been shown to be
superior to even computed tomography in distin-
guishing airway wall invasion from extrinsic com-
EBUS has revolutionized bronchoscopy because pression with a sensitivity of 89% and specificity
it extends the endoscopist’s vision beyond the air- of 100% [1]. When airway walls are examined, up
way walls. EBUS is made possible by miniaturized to 7 layers can be distinguished. The mucosa, en-
ultrasound transducers that can be inserted via the dochondium, perichondium and adventitia are
working channel of a flexible bronchoscope. This hyperechoic (white) while the intervening sub-
is termed radial EBUS and facilitates evaluation mucosa, cartilage and connective tissue layer are
of airway walls, as well as location of peripheral hypoechoic (gray) [1]. The relationship of lymph
lung lesions. Alternatively, a linear transducer has nodes relative to the airways can also be visual-
been built in a dedicated bronchoscope to permit ized using radial EBUS. Once a target lesion is
real-time TBNA biopsies. This is termed EBUS- identified, the probe is removed from the bron-
TBNA and has become an accepted modality for choscope and regular TBNA can be performed to
the invasive staging of the mediastinum in non- obtain biopsy specimens. Although radial EBUS
small cell lung cancer. facilitates such guided biopsies, it is limited by the
absence of real-time sampling. The reported di-
agnostic yields with radial EBUS directed TBNA
Radial EBUS is 72–86% and this procedure has been shown to
improve diagnostic rates over ‘blind’ conventional
Radial EBUS has a 20-MHz (12–30 MHz available) TBNA in obtaining a histological diagnosis from
rotating transducer. ‘Central’ probes with balloon paratracheal lymph node stations [2, 3].
sheaths are used in the proximal airways for either When radial EBUS is used to guide broncho-
bronchial wall assessment or to guide TBNA of scopic biopsies of peripheral lung lesions, it in-
lymph nodes. ‘Peripheral’ probes without balloon creases the diagnostic yield of transbronchial lung
sheaths are used to identify parenchymal lung biopsies of small (<30 mm) lesions [4]. Diagnostic
Radial EBUS image

‘Central’ radial EBUS with balloon sheath

Fig. 1. ‘Central’ and ‘peripheral’ ra-


dial EBUS probes. Inset: ultrasound
image of a peripheral lung lesion ‘Peripheral’ radial EBUS
(arrow).

sensitivities of 61–80% that are independent of le- extending 20–50 mm in depth (fig. 2). The EBUS-
sion size have been reported [3, 4]. EBUS also ap- TBNA scope has an external diameter of 6.9 mm
pears to remove variables that usually influence which is larger than a standard flexible broncho-
the ability to obtain a positive histological diag- scope. Therefore, oral rather than nasal intubation
nosis. These variables include underlying disease, is necessary. Furthermore, the endoscopic view-
presence of the bronchus sign and lobar location ing optics is at a 30° oblique angle and operator
[5]. The transducer probe is inserted through a compensation is required during bronchoscopy.
guide sheath into the bronchi where the lesion is This bronchoscope has a 2.0-mm working chan-
suspected based on pre-procedure computed to- nel that is designed to house a dedicated 21- to
mography imaging. Normal air-filled alveoli pro- 22-gauge biopsy needle with a depth of penetra-
duce a whitish snowstorm image with ‘comet-tail’ tion can be varied up to 40 mm by a safety lock.
artifacts. Lung lesions are usually more hypoecho- There is a balloon fitted over the transducer probe
ic or gray. When a tumor is located, the transducer that can be filled with saline to facilitate coupling.
is removed and regular biopsy forceps are used to However, this is not always needed if good contact
take biopsies through the guide sheath that has can be maintained between the airway mucosa
been maintained in position within the lesion. and the probe. Lymph nodes appear more echoic
(gray) than blood vessels and Doppler can be used
in making the distinction. Once the target lymph
Linear EBUS-TBNA nodes are identified on EBUS, real-time TBNA
is performed. The needle sheath that houses the
The EBUS-TBNA bronchoscope has a 7.5-MHz TBNA needle is pushed forward first such that it
transducer mounted on its distal tip. This trans- is visualized on the endoscopic image before the
ducer is convex and produces a 50° sector view ‘jabbing’ technique is used to perform TBNA un-
parallel to the long axis of the bronchoscope der real-time guidance. Once the TBNA needle is

Endobronchial Ultrasound 85
EBUS-TBNA image

Fig. 2. EBUS-TBNA bronchoscope EBUS-TBNA bronchoscope


with balloon sheath (arrow) and
22-gauge needle. Inset: ultrasound
image of tumor (right arrow) invad-
ing a blood vessel (left arrow).

within the target, the stylet of the needle is agitat- that the results are true negatives. Re-staging of
ed to dislodge any airway debris. The stylet is then the mediastinum with EBUS-TBNA after neoad-
removed before biopsies can be aspirated. All me- juvant chemotherapy has had less success with a
diastinal and hilar lymph nodes are accessible to much lower diagnostic sensitivity [3].
EBUS-TBNA except the aortopulmonary (station The histological staging of a radiologically
5), paraaortic (station 6), paraesophageal (station normal mediastinum with either lymphadenop-
8) and pulmonary ligament (station 9) nodes. For athy ≤10 mm or negative positron emission to-
mediastinal staging of non-small cell lung cancer, mography has been shown to be feasible with sen-
3 cytology aspirations per lymph node station is sitivities approaching 90% [9]. These data would
recommended. If adequate core specimens are suggest that systematic staging is possible as op-
obtained, then 2 passes will suffice [6]. posed to the targeted sampling that is widely prac-
EBUS-TBNA has been shown to have a higher ticed. However, such studies were performed by
diagnostic yield than conventional ‘blind’ TBNA experts on patients under general anesthesia and
and may be comparable in sensitivity to cervi- the widespread applicability of the data is still not
cal mediastinoscopy [3, 7]. The pooled diagnos- established because EBUS-TBNA is usually per-
tic sensitivity of EBUS-TBNA is 90% and speci- formed under moderate sedation. Nevertheless,
ficity is 100%, but the false negative rate is about EBUS clearly has the potential to identify lymph-
20% [8]. This discordance between sensitivity and adenopathy that has been missed on computed
negative predictive value is attributed to the high tomography. In addition, the 22-gauge needle can
prevalence of malignancy in the reported studies. obtain samples that are sufficient for genetic and
The high false negative rates mandate that all neg- molecular analysis such as epidermal growth fac-
ative results need to be either followed up clini- tor receptor mutations.
cally or subject to further testing using alternative By combining EBUS-TBNA with another en-
modalities such as mediastinoscopy to confirm doscopic procedure, i.e. transesophageal EUS-

86 Anantham ⭈ Siyue
guided fine-needle aspiration, complete staging of bronchoscopes with careless use are complications
the mediastinum is possible with access to lymph that need to be avoided with the appropriate train-
node stations not accessible to either technique on ing. Radial probes should not be inserted into the
its own. EBUS offers greater access to the paratra- working channel when ‘active’ and spinning. By
cheal and hilar lymph nodes while EUS can tar- ensuring that the needle sheath is clearly visible
get inferior mediastinal lymph nodes and adre- before attempting TBNA, inadvertent perforation
nal metastases. Diagnostic sensitivities of 93–94% of the EBUS-TBNA scope can be prevented.
have been reported for the combined EBUS and EBUS can also add to endoscopy time and this
EUS procedure [3]. has implications for what can be achieved under
EBUS-TBNA has also been used to successfully moderate sedation. However, in expert hands,
obtain biopsy specimens from primary tumors lo- radial EBUS adds less than 3 min to TBNA and
cated in the paratracheal and peribronchial region about 60 s to the transbronchial biopsy of periph-
with a diagnostic sensitivity of 82–94% [3, 10]. As eral lesions. Targeted EBUS-TBNA of enlarged
long as there is no intervening aerated lung, these lymph nodes has been reported to take a mean of
tumors can be identified as soft tissue structures 12.5 (range 8–21) min [3]. However, a longer pro-
on ultrasound. The procedure is then similar to cedure duration is likely to be needed if complete
sampling lymph nodes and is especially useful mediastinal staging is attempted with evaluation
in diagnosing centrally located tumors without of non-enlarged lymph nodes as well.
any airway involvement. In the demonstration
of non-caseating granulomatous inflammation,
EBUS-TBNA has a diagnostic yield for sarcoi- Conclusion
dosis of 85–94% [3]. This has been shown to be
more effective than standard TBNA. If bigger tis- EBUS has the potential to become part of stan-
sue specimens are needed for histological analysis dard bronchoscopy because of negligible com-
in conditions such as lymphoma, it is even pos- plications, improved diagnostic yield and a short
sible to insert a 1.15-mm miniforceps through the learning curve. It enhances histopathological
EBUS scope and push it past the airway wall via staging of the mediastinum in non-small cell lung
a needle puncture [3]. This can potentially obtain cancer, as well as increases the diagnostic yields
real-time forceps biopsies of mediastinal lesions. of both peribronchial and peripheral lung lesions.
Furthermore, EBUS-TBNA has been used thera- Ultrasonographic reflections enable the endosco-
peutically to drain mediastinal, as well as bron- pist to view beyond the surface of the airway walls
chogenic cysts and consequently relieve symp- and identify deeper structures. This ability to ‘see
tomatic central airway obstruction [3]. through walls’ without the need for ionizing ra-
Complications have rarely been attributed diation remains the premise on which EBUS has
directly to either radial EBUS or EBUS-TBNA. been developed. It is also the reason why EBUS
Radial EBUS-guided lung biopsies have a re- continues to gain widespread acceptance.
ported rate of pneumothorax of 0–5% and risk of
moderate bleeding of 1% which is in keeping with
the complication rates expected from convention- Recommendations
al transbronchial lung biopsies [3, 4]. Therefore,
these cases are considered complications of for- • Radial EBUS facilitates airway wall assessment,
ceps biopsy rather than that of EBUS imaging. TBNA of paratracheal lymph nodes and
However, damage to the delicate radial trans- biopsies of peripheral lung lesions via a guide
ducer probes and perforation of the EBUS-TBNA sheath.

Endobronchial Ultrasound 87
• Linear EBUS-TBNA improves diagnostic yield if the necessary equipment and expertise are
compared to conventional ‘blind’ TBNA of available.
mediastinal and hilar lymph node stations, as • Negative biopsies from EBUS-guided pro-
well as peribronchial tumors. cedures need to have subsequent biopsies from
• EBUS-TBNA is a recognized modality for the alternative procedures because of the risk of
invasive staging of the mediastinum in non- false negative results.
small cell lung cancer and is recommended

References
1 Herth F, Ernst A, Schulz M, Becker H: 5 Yamada N, Yamazaki K, Kurimoto N, et 8 Detterbeck FC, Jantz MA, Wallace M, et
Endobronchial ultrasound reliably dif- al: Factors related to diagnostic yield of al: American College of Chest Physi-
ferentiates between airway infiltration transbronchial biopsy using endobron- cians: Invasive Mediastinal Staging of
and compression by tumor. Chest chial ultrasonography with a guide Lung Cancer: ACCP Evidence-Based
2003;123:458–462. sheath in small peripheral pulmonary Clinical Practice Guidelines, ed 2. Chest
2 Herth F, Becker HD, Ernst A: Conven- lesions. Chest 2007;132:603–608. 2007;132(suppl):202S–220S.
tional vs. endobronchial ultrasound- 6 Lee HS, Lee GK, Lee HS, et al: Real-time 9 Herth FJ, Eberhardt R, Krasnik M, et al:
guided transbronchial needle aspiration: endobronchial ultrasound-guided trans- Endobronchial ultrasound-guided trans-
a randomized trial. Chest 2004;125: bronchial needle aspiration in mediasti- bronchial needle aspiration of lymph
322–325. nal staging of non-small cell lung can- nodes in the radiologically and positron
3 Anantham D, Koh MS, Ernst A: Endo- cer: how many aspirations per target emission tomography – normal medi-
bronchial ultrasound. Respir Med 2009; lymph node station? Chest 2008;134: astinum in patients with lung cancer.
103:1406–1414. 368–374. Chest 2008;133:887–891.
4 Paone G, Nicastri E, Lucantoni G, et al: 7 Ernst A, Anantham D, Eberhardt R, et 10 Nakajima T, Yasufuku K, Fujiwara T, et
Endobronchial ultrasound-driven biopsy al: Diagnosis of mediastinal adenopathy al: Endobronchial ultrasound-guided
in the diagnosis of peripheral lung real-time endobronchial ultrasound- transbronchial needle aspiration for the
lesions. Chest 2005;128:3551–3557. guided needle aspiration versus medias- diagnosis of intrapulmonary lesions. J
tinoscopy. J Thorac Oncol 2008;3:577– Thorac Oncol 2008;3:985–988.
582.

Devanand Anantham
Consultant
Department of Respiratory and Critical Care Medicine
Singapore General Hospital
Outram Road, Singapore 169608 (Singapore)
Tel. +65 6321 4700, Fax +65 6227 1736, E-Mail anantham.devanand@sgh.com.sg

88 Anantham ⭈ Siyue
Technology in Anesthesiology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 89–95

Minimally Invasive Thoracic Surgery for Pulmonary


Resections
Michele Salati ⭈ Gaetano Rocco
Division of Thoracic Surgery, Department of Thoracic Surgery and Oncology, National Cancer Institute,
Pascale Foundation, Naples, Italy

Abbreviations Analysis
MITS Minimally invasive thoracic surgery
NSCLC Non-small cell lung cancer Traditional Technique to Perform VATS and the
VATS Video-assisted thoracic surgery Indication of This Approach
VATS emerged as a routine procedure to treat
several thoracic diseases after 1990, as it is testi-
fied by a multitude of papers inherent to this topic
The era of MITS began almost 100 years ago when published since this date. Reports from different
Hans Christian Jacobaeus first introduced a cysto- institutions and the concomitant effort by the in-
scope through the chest wall to explore the pleural dustries to improve the efficiency of the surgical
cavity and to cauterize pleural adhesions favoring instruments gave a contribution to further define
lung collapse in the treatment of pulmonary tu- in most recent years the precise role of this tech-
berculosis [1]. Since then the evolution of tech- nique and in treating all those pathologic condi-
nologies with consequent development of more tions previously approached via open surgery.
sophisticated and reliable surgical procedures has From a practical point of view, VATS is a MITS
led to the modern concepts and practice of VATS procedure that usually requires three small inci-
[2]. sions (fig. 1) on the chest wall (ports) through
In this paper we present the technical aspects which the instruments can be placed to see in-
of a currently minimally invasive approach in the side the pleural space (thoracoscope) and to per-
scenario of modern thoracic surgery, emphasiz- form a large variety of procedures (operative in-
ing its benefits in comparison to conventional struments). As a rule, the VATS technique can be
open techniques. In addition, considering the performed only in those patients in whom the
fact that the process of technologic improvement placement of an endotracheal double-lumen tube
is still ongoing and will probably become even by the anesthesiologist allows the collapse of the
more rapid than in the past, we also describe the affected lung obtaining an effective space with-
frontiers that nowadays have been reached by in the pleural cavity. However, recent evidence
MITS. seems to support the feasibility of VATS in the
Fig. 1. Usual localization of the ports
on the chest wall for conventional
VATS technique and the ‘baseball
diamond’.

Table 1. Absolute and relative contraindication for VATS

Absolute Medical contraindication for general anesthesia (i.e. recent cardiac disease, insufficient
contraindications pulmonary function test, severe coagulopathy)

Anatomic deformity of the airways or inability to tolerate a single lung ventilation

Prior decortication or pleurodesis

Relative Extensive pleural adhesions or previous intrathoracic procedures


contraindications
Target lesion not accessible (i.e. small central tumors)

Masses with a large involvement of the chest wall

Thoracic anatomic deformities

awake patient [3]. Table 1 shows the contraindi- obtain a strategic ideal triangulation that allows
cations usually reported in the literature for safely the maximal visibility and manipulation of the
performing this surgical approach. pathologic site (fig. 1). It is also important to take
Technical Aspects. The positions reported of into consideration that during the operation the
the three incisions can vary depending on the site sites of the scope and of the instruments can be
of the target lesion and it should respect the con- swapped around in order to maximize the visibil-
cept of the so-called ‘baseball diamond theory’ ity and to optimize the operative angles [5].
[4]. Accordingly, the thoracoscope and the oth- Over the last two decades the surgical indus-
er instruments should be positioned in order to tries have multiplied and refined a multitude of

90 Salati ⭈ Rocco
instruments that now make it possible to perform chest tube duration and length of stay [13, 20, 21].
nearly all the procedures traditionally approached In addition, the oncologic results are comparable
with open surgery [6, 7]. Obviously, the introduc- between traditional surgery and MITS approach
tion of any new technique in thoracic surgery is a to cure patients affected by NSCLC [22, 23].
challenge that involves not only surgeons, but the
entire surgical team (surgeons, anesthesiologists Recent Evolutions of the MITS
and nurses). Before starting a VATS program the Nowadays the MITS is evolving towards frontiers
center must have acquired a large and consolidat- that in some centers are already the preferred
ed experience in open thoracic surgery [8]. treatment option for specific thoracic diseases. In
this scenario the robotically assisted thoracic sur-
Applications and Clinical Advantages of the VATS gery and the uniportal VATS probably represent
Procedure the most important evolutions. In, 2008, Melfi
The applications of the VATS approach are in- and Mussi [24] published a detailed paper about
creasing in modern thoracic surgery as shown the learning curve and complications related to
and today the VATS approach is the standard of the robotically assisted lobectomy. Table 2 reports
care for many intrathoracic diseases [9–13]. The the advantages and disadvantages of VATS com-
technical features of VATS lobectomies compared pared to robotic surgery as clearly defined in this
to open surgery have been investigated in several study. Although some other papers [25, 26] dem-
studies [14], although the applied methodology onstrated the feasibility and safety of this tech-
was not always rigorous. nique for performing several thoracic procedures,
VATS lobectomy as well as minor resections this high technologic approach appears to be still
of the lung performed through MITS techniques in an evolving phase.
seem to warrant the same effectiveness of the con- At present the most complete device for the ro-
ventional thoracotomy procedures. Moreover, botically assisted surgery is the Da Vinci Robotic
VATS offers advantages in some clinical aspects System (Surgical Intuitive, Mountain View, Calif.,
when compared to the previous thoracotomy ap- USA) [27]. This high-tech and expensive system
proach. When the results of VATS for minor re- is made of a console where the surgeon sits and
sections (i.e. wedge resections with curative or manipulates two joysticks looking through binoc-
diagnostic purposes or for pneumothorax) are ulars that give a three-dimensional view of the op-
compared to open surgery, it is clear that the VATS erative field. The console is connected to the sur-
techniques are associated with a shorter hospital gical manipulator which offers two arms directly
stay and a reduced need for pain medication as a moved by the surgeon at the console (instrument
consequence of less postoperative pain [15–18]. arms) and another arm to guide the endoscope.
At the same time, one randomized controlled trial Moreover, the instrument is provided with a mo-
showed that such advantages were obtained with tion scaling that increases the precision of the sur-
higher costs in case of lung biopsies for interstitial geon’s original movement and at the same time
lung disease [19]. reduces the natural human tremor.
Recently, large case series of patients submitted The theoretic and practical phases of the train-
to VATS lobectomy to treat early-stage NSCLC ing program should provide the knowledge of a
have been published. Although the information new and extremely technological surgical arma-
derived from randomized controlled trials is mentarium and develop new capabilities inherent
scarce, it appears that VATS lobectomy is associ- to the robotic surgery such as the binocular three-
ated to less postoperative pain, reduced postop- dimensional vision, the sensibility of the joystick
erative complications and mortality rate, shorter and the degrees of the robotic arm movement.

MITS for Pulmonary Resection 91


Table 2. Advantages and disadvantages of video-thoracoscopic surgery vs. robotic surgery [from 24, with permis-
sion]

Video-thoracoscopic surgery Robotic surgery

Advantages Well-developed technology Three-dimensional imaging


Affordable and ubiquitous Dexterity
Proved efficacy Seven degrees of freedom
No fulcrum effect
No physiologic tremors
Scale motions
Ergonomic position

Disadvantages Loss of touch sensation No tactile feedback


Compromised dexterity Expensive
Limited degrees of motion Unproved benefit
Fulcrum effect
Amplification of physiologic tremors

Table 3. Technical recommendations to perform different uniportal VATS procedures [from 28, with permission]

Procedure Intercostal space Line Decubitus

Bullectomy 5th Posterior-median lateral


axillary line

Lung biopsy or lung upper lobe 5th Median axillary line lateral
resection
middle lobe-lingula 5th–6th Posterior axillary- lateral
scapular line

lower lobe 4th Median-posterior lateral


axillary line

Mediastinal nodes middle mediastinum 5th Scapular line lateral


biopsy
posterior mediastinum 5th Posterior axillary line supine

Sympathectomy 3rd Axillary hair line lateral

Pericardial window 5th Axillary line supine-


semilateral

The uniportal VATS represents the evolution purposes, given its high versatility to reach differ-
of MITS towards the least invasive approach. It ent targets inside the thoracic cavity [28]. The op-
can be used to treat several intrathoracic condi- erative steps and the feasibility of performing lung
tions (table 3) with both diagnostic and curative wedge resection using the uniportal approach were

92 Salati ⭈ Rocco
that the use of roticulating instruments with very
small diameter (5.0–3.0 mm) is necessary to re-
duce at a minimum their mutual interference.
Obviously this approach does not allow to mutu-
ally change the position of the operative instru-
ments and the optical source during the opera-
tion. So it is fundamental to carefully choose the
right intercostal space where to place the single
a incision (table 3).
Considering the clinical benefits of this tech-
nique, it seems that the uniportal VATS magnifies
the results already obtained with the conventional
VATS. In fact, several studies have shown that in
comparison to the three-portal VATS, the unipor-
tal VATS can offer clinical advantages for the pa-
tient, such as a reduction of pain and paraesthesia,
a shorter hospital stay and a shorter time for return
to work [29–31]. Interestingly, at least one paper
published by Salati et al. [32] that compares the
b results of uniportal VATS vs. three-portal VATS
for treating primary spontaneous pneumothorax
demonstrated that the uniportal approach mini-
Fig. 2. Diagram representing the different geometric ap-
proaches to the pulmonary lesion by (a) standard three-
mizes the global hospital costs.
port VATS compared with (b) uniportal VATS [from 29,
with permission].
Discussion

The technical improvements achieved in recent


initially reported in a paper published in 2004 by years have made VATS a valuable option for per-
Rocco et al. [29]. In the uniportal VATS, a single- forming minor and major lung resection and a
port incision (about 2.0–2.5 cm long) is used as the MITS approach has become the first choice to
unique access to insert the optical source and all treat many intrathoracic diseases, particularly be-
the operative instruments. To perform uniportal nign conditions (i.e. primary spontaneous pneu-
VATS a radical change of the traditional concepts mothorax and emphysematous bullae) or lesions
related to the conventional three-portal VATS is without diagnosis (i.e. solitary pulmonary nod-
necessary. In fact, in the uniportal VATS technique ule and interstitial lung disease) [9]. The use of
the geometric approach to reach the target lesion VATS is more controversial in case of primary or
is completely different and it is represented by a secondary pulmonary tumors, due to the lack of
sagittal plane where the lesion and the operative strong scientific evidence [14].
instruments should ideally lie (fig. 2). Although several papers have investigated the
Taking into consideration this new perspec- benefits of VATS vs. open surgery, demonstrat-
tive and the fact that the instruments are inserted ing that the latter is superior in many aspects, we
through the same port incision, which can be con- think that the widespread use of VATS would need
sidered the fulcrum for all of them, it is intuitive more prospective randomized trials on several

MITS for Pulmonary Resection 93


Table 4. Ideal terms of comparison between the con- Possibly, one or two VATS lobectomists should be
ventional thoracic surgery and VATS part of a surgical team of dedicated thoracic sur-
Clinical terms of Pain quantification
geons [35]. Costs of VATS may not necessarily
comparison Postoperative respiratory function represent an issue when the reduction in length
recovery of hospitalization is taken into account [35]. For
Complication rate reduction that reason it seems important to include a VATS
Ability to work impairment training program during the thoracic surgery res-
Chest tube duration
Immunocompetence involvement
idency in order to acquire the technical skills to
Oncologic benefit perform a minimally invasive procedure indepen-
Quality of life impact dently and safely [36].
Cost comparison Postoperative hospital stay
duration
Operative cost evaluation Recommendations
Postoperative cost evaluation
Recovery to work and social cost • VATS should be considered the standard of care
to treat several lung disease in modern thoracic
surgery (diagnosis of pulmonary nodules,
surgical lung biopsies for interstitial lung
disease, bullectomy for primary spontaneous
related aspects (table 4). In particular, the poten- pneumothorax).
tial oncologic benefit of approaching lung cancer • VATS offers substantial clinical benefits for
by VATS is still not completely known. The dem- the patients in comparison to the conventional
onstration that MITS procedures allow to main- open approach (reduction of pain and shorter
tain a high immunocompetence against lung tu- hospital stay).
mors could provide further trust for its use [33]. • A VATS lobectomy program should be
Undoubtedly, nowadays VATS should be made developed in every thoracic surgery center
available in every thoracic surgical unit perform- given the proved clinical and oncological
ing an adequate yearly volume of procedures [34]. benefits for treating early-stage NSCLC.

References
1 Jacobaeus HC: The practical impor- 4 Landreneau RJ, Mack MJ, Hazelrigg SR, 7 Asamura H, Suzuki K, Kondo H, et al:
tance of thoracoscopy in surgery of the et al: Video-assisted thoracic surgery: Mechanical vascular division in lung
chest. Surg Gynecol Obstet 1922;34: basic technical concepts and intercostals resection. Eur J Cardiothorac Surg 2002;
289–296. approach strategies. Ann Thorac Surg 21:879–882.
2 Landreneau RJ, Mack MJ, Dowling RD, 1992;54:800–807. 8 McKenna RJ: Complications and learn-
et al: The role of thoracoscopy in lung 5 Sasaki M, Hirai S, Kawabe M, et al: Tri- ing curves for video-assisted thoracic
cancer management. Chest 1998;113:6S– angle target principle for the placement surgery lobectomy. Thorac Surg Clin
12S. of trocars during video-assisted thoracic 2008;18:275–280.
3 Pompeo E, Mineo D, Rogliani P, et al: surgery. Eur J Cardiothorac Surg 9 Yim APC: VATS major pulmonary resec-
Feasibility and results of awake thoraco- 2005;27:307–312. tion revisited – controversies, tech-
scopic resection of solitary pulmonary 6 Ng CSH, Yim APC: Video-assisted thora- niques, and results. Ann Thorac Surg
nodules. Ann Thorac Surg coscopic surgery bullectomy for emphy- 2002;74:615–623.
2004;78:1761–1768. sematous/bullous lung disease. MMCTS
April 2005:265.

94 Salati ⭈ Rocco
10 Solli P, Spaggiari L: Indications and 19 Miller JD, Urschel JD, Cox G, et al: A 28 Salati M, Brunelli A, Rocco G: Uniportal
developments of video-assisted thoracic randomized, controlled trial comparing video-assisted thoracic surgery for diag-
surgery in the treatment of lung cancer. thoracoscopy and limited thoracotomy nosis and treatment of intrathoracic
Oncologist 2007;12:1205–1214. for lung biopsy in interstitial lung dis- conditions. Thorac Surg Clin 2008;18:
11 McKenna RJ Jr, Houck W, Fuller CB: ease. Ann Thorac Surg 2000;70:1647– 305–310.
Video-assisted thoracic surgery lobec- 1650. 29 Rocco G, Martin-Ucar A, Passera E: Uni-
tomy: experience with 1,100 cases. Ann 20 Kirby TJ, Mack MJ, Landreneau RJ, et al: portal VATS wedge pulmonary resec-
Thorac Surg 2006;81:421–426. Lobectomy – video-assisted thoracic tions. Ann Thorac Surg 2004;77:726–
12 Loscertales J, Jimenez-Merchan R, Con- surgery versus muscle-sparing thoraco- 728.
gregado M, et al: Video-assisted surgery tomy. A randomized trial. J Thorac Car- 30 Rocco G: VATS lung biopsy: the unipor-
for lung cancer. State of the art and per- diovasc Surg 1995;109:997–1002. tal technique. MMCTS January 2005:356
sonal experience. Asian Cardiovasc Tho- 21 Flores RM, Park BJ, Dycoco J, et al: 31 Jutley RS, Khalil MW, Rocco G: Unipor-
rac Ann 2009;17:313–326. Lobectomy by video-assisted thoracic tal vs. standard three-port VATS tech-
13 Tomaszek SC, Cassivi SD, Shen KR, et al: surgery versus thoracotomy for lung nique for spontaneous pneumothorax:
Clinical outcomes of video-assisted tho- cancer. J Thorac Cardiovasc Surg July comparison of post-operative pain and
racoscopic lobectomy. Mayo Clin Proc. 2009;138:11–18. residual paraesthesia. Eur J Cardiothorac
2009;84:509–513. 22 Walker WS, Codispoti M, Soon SY, et al: Surg 2005;28:43–46.
14 Sedrakyan A, van der Meulen J, Lewsey Long-term outcomes following VATS 32 Salati M, Brunelli A, Xiume F, et al: Uni-
J, et al: Video-assisted thoracic surgery lobectomy for non-small cell broncho- portal video-assisted thoracic surgery
for treatment of pneumothorax and lung genic carcinoma. Eur J Cardiothorac for primary spontaneous pneumothorax:
resections systematic review of ran- Surg 2003;23:397–402. clinical and economic analysis in com-
domised clinical trials. BMJ 2004;329: 23 Sugi K, Kaneda Y, Esato K: Video- parison to the traditional approach.
1008. assisted thoracoscopic lobectomy Interact Cardiovasc Thorac Surg 2008;7:
15 Waller DA, Forty J, Morritt GN: Video- achieves a satisfactory long-term prog- 63–66.
assisted thoracoscopic surgery versus nosis in patients with clinical stage IA 33 Craig SR, Leaver HA, Yap PL, et al: Acute
thoracotomy for spontaneous pneu- lung cancer. World J Surg 2000;24: phase responses following minimal
mothorax. Ann Thorac Surg 1994;58: 27–31. access and conventional thoracic sur-
372–377. 24 Melfi FMA, Mussi A: Robotically gery. Eur J Cardiothorac Surg 2001;20:
16 Ayed AK, Al-Din HJ: Video-assisted tho- Assisted Lobectomy: Learning Curve 455–463.
racoscopy versus thoracotomy for pri- and Complications. Thorac Surg Clin 34 Klepetko W, Aberg TH, Lerut AE, et al:
mary spontaneous pneumothorax: a 2008;18:289–295. EACTS/ESTS Working Group on Struc-
randomized controlled trial. Med Prin- 25 Bodner J, Wykypiel H, Wetscher G, et al: tures in Thoracic Surgery Structure of
ciples Pract 2000;9:113–118. First experiences with the da VinciTM General Thoracic Surgery in Europe. Eur
17 Santambrogio L, Nosotti M, Bellaviti N, operating robot in thoracic surgery. Eur J Cardiothorac Surg 2001;20:663–668.
et al: Videothoracoscopy versus thoraco- J Cardiothorac Surg 2004;25:844–851. 35 Walker WS, Casali G: The VATS lobecto-
tomy for the diagnosis of the indetermi- 26 Park BJ, Flores RM, Rusch VW: Robotic mist: analysis of costs and alterations in
nate solitary pulmonary nodule. Ann assistance for video-assisted thoracic the traditional surgical working pattern
Thorac Surg 1995;59:868–871. surgical lobectomy: technique and initial in the modern surgical unit. Thorac Surg
18 Landreneau RJ, Mack MJ, Dowling RD, results. J Thorac Cardiovasc Surg 2006; Clin 2008;18:281–287.
et al: The role of thoracoscopy in lung 131:54–59. 36 McKenna RJ: Complications and learn-
cancer management. Chest 27 Melfi FMA, Ambrogi MC, Lucchi M, et ing curves for video-assisted thoracic
1998;113(suppl):6S-12S. al: Video robotic lobectomy. MMCTS surgery lobectomy. Thorac Surg Clin
June 2005:448. 2008;18:275–280.

Michele Salati, MD
Via A. De Gasperi 17/c
IT–60020 Offagna, Ancona (Italy)
Tel. +39 349 2599 060, Fax +39 071 5964 481
E-Mail michelesalati@hotmail.com

MITS for Pulmonary Resection 95


Section Titlein Anesthesiology
Technology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 96–101

Acute Lung Collapse during Open-Heart Surgery


Praveen Kumar Neema ⭈ S. Manikandan ⭈ Ramesh Chandra Rathod
Department of Anaesthesiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India

Abbreviations Mechanisms of Acute Lung Collapse


CPAP Continuous positive-airway pressure
CPB Cardiopulmonary bypass Passive recoil of the lungs occurs when ventilation
ECMO Extracorporeal membrane oxygenation is discontinued on CPB; passive recoil returns
ET Endotracheal tube lung volume to its functional residual capacity.
IPPV Intermittent positive pressure ventilation The inherent tendency of lungs to collapse is due
OHS Open-heart surgery to fluid lining the alveoli and the presence of elas-
PEEP Positive end-expiratory pressure tin fibers in the lung parenchyma; the tendency of
RV Right ventricle collapse is opposed by outward pull by the chest
wall, and by the presence of negative intrapleu-
ral pressure [1]. The presence of surfactant in the
Patients undergoing OHS invariably suffer either alveolar fluid lining reduces surface tension and
total or partial lung collapse on discontinuation decreases the tendency of alveolar collapse; simi-
of ventilation during CPB. The collapse of the larly, presence of nitrogen in the alveolar gas de-
lung is total if the pleura or chest wall is opened. lays gaseous absorption, provides stability to the
Acute unyielding lung collapse during OHS is alveoli, and delays alveolar collapse (atelectasis).
very rare, but if it develops it can seriously com- Despite these opposing forces the lungs gradual-
promise respiratory and circulatory functions ly collapse on discontinuation of ventilation. The
that may pose life-threatening challenges. This collapsed lungs generally re-expand on manual
review describes mechanisms of acute lung col- inflation and resumption of IPPV. Occasionally,
lapse during OHS, the pathophysiology of car- because of the obstruction, compression, or col-
diovascular and respiratory complications in the lapse of the main airway or its branches or pneu-
presence of acute lung collapse, the measures mothorax (table 1), it is not possible to achieve
that help re-expansion of the collapsed lung, the complete re-expansion of the lung parenchyma.
measures that help separation of the patient from With extraluminal airway compression, lung
CPB in spite of persistent lung collapse and the collapse occurs only if the obstruction is com-
measures that may prevent acute lung collapse plete, otherwise it results in air trapping and hy-
during CPB. perinflation of the lung supplied by the partially
Table 1. Causes of acute lung collapse during open heart surgery

Intraluminal
Mucous plugs
Inspissations of accumulated secretions
Clotted blood in the airway
Intraluminal lesion

Extraluminal
Left lower lobe bronchus compression due to enlarged left ventricle
Left lower lobe bronchus compression due to aneurysmal left atrium
Left main bronchus compression due to enlarged and tense left pulmonary artery
Left bronchus compression due to transesophageal echocardiography probe
Right middle lobe bronchus compression due to enlarged right pulmonary artery
Right airway compression due to aneurysm of ascending aorta
Direct origin of right upper lobe bronchus from trachea
Vascular ring

Others
Inadvertent advance of the ET tube into the right or left bronchus
Pneumothorax secondary to:
Central venous cannulation
Pleural injury during internal mammary artery dissection
Pleural and lung injury during adhesiolysis in redo open heart surgery
Pleural injury during sternal closure and wiring

compressed airway and compression collapse of the ventricular function. During IPPV the car-
the surrounding normal lung parenchyma. The diopulmonary interactions depend on cardiac re-
CPB-associated inflammatory responses and loss serve, circulating blood volume, autonomic tone,
of surfactant, myocardial dysfunction, and trans- lung volume, attained intrathoracic pressure and
fusion-related acute lung injury, etc., are other its effects on pulmonary vasculature and juxta-
important mechanisms that decrease the lung cardiac pleural pressure [2]. The cardiopulmo-
compliance, increase the inherent tendency of nary effects of lung collapse and incomplete lung
lung collapse and might necessitate application of expansion depend on the magnitude of the lung
increased inflation pressures and PEEP to ensure collapse, the integrity of the pleura, the tidal vol-
sustained lung expansion. ume chosen for ventilation, the circulating blood
volume, ventricular preload and the contractil-
ity of the heart, etc. It should be noted that the
Pathophysiology of Cardiovascular and small areas of lung collapse are usually of no clin-
Respiratory Complications during Acute Lung ical consequence. Cardiopulmonary interactions
Collapse manifest as mechanical and physiologic effects.
The mechanical effects are due to heterogeneity of
It is important to understand various aspects of airway resistance and lung compliance, and lead
cardiopulmonary interactions between IPPV and to differential ventilation of lung parenchyma

Acute Lung Collapse during Open-Heart Surgery 97


including overdistension of compliant alveoli with attempt to improve hemodynamics and arterial
normal airways, poor ventilation of compliant al- blood gases.
veoli with compromised airways and air trapping
of alveoli with grossly compromised airways and
no ventilation of alveoli with obstructed airways. Measures That May Help Re-Expansion of the
Additionally, the alveoli are repeatedly subjected Collapsed Lung
to volutrauma as well as barotrauma. The venti-
lation-perfusion effects vary from no perfusion Careful repositioning of the ET tube and/or
in overdistended alveoli to absence of ventilation ET suction generally restores airway integrity.
in collapsed alveoli with varying effects on gas- Sustained inflation (up to 45 cm H2O for approx-
eous exchange. The overdistended alveoli act as imately 20 s in patients with noncompliant lungs
dead space whereas collapsed alveoli as shunt and and 40 cm H2O for approximately 8 s in healthy
others lie within this wide spectrum; this hetero- patients) has been shown to recruit collapsed al-
geneity of ventilation-perfusion leads to varying veoli [3]. Rarely, it may not be possible to establish
degrees of hypoxemia and hypercapnia. The over- a clear airway and in such situations it is not possi-
distended alveoli act as Starling’s resistor because ble to achieve complete re-expansion of the lungs.
of collapse of the surrounding capillary network In the presence of proximal thick airway secre-
and increased pulmonary vascular resistance. The tions, the ability to deliver a high inspiratory pres-
peak and plateau airway pressures are increased sure to recruit collapsed lung units is inhibited.
but do not offer a clue to the extent of existing Fiberoptic bronchoscopy offers numerous advan-
heterogeneity of ventilation and perfusion. The tages: it allows the anesthesiologist to visualize the
cardiovascular effects are secondary to increased tracheobronchial anatomy, the physical charac-
afterload to RV, raised catecholamine levels due to teristics, the quantity, and the source of the secre-
hypoxemia and hypercarbia, and impaired filling tions. In addition, extraluminal obstructions can
of the left ventricle due to decreased RV output be recognized. The limitation to this technique
and possibly due to a shift of the interventricular in the pediatric population is related to the size
septum. The presence of increased peak airway of the bronchoscope. Pediatric bronchoscopes fit
pressure, decreased air entry on auscultation, ab- through a 5.0-mm ET tube. However, suctioning
sent movement of the pleura and presence of vis- through small scopes may be suboptimal, espe-
ibly collapsed lung confirms the diagnosis. The cially when thick secretions are present. The clini-
clinical manifestations during separation from cal scenario of full CPB can be taken advantage
CPB may include raised central venous pres- of, however, and allow for temporary removal of
sure, visible or echocardiographic demonstra- the ET tube from the trachea (recognizing that it
tion of RV distension with or without interven- may be challenging to re-intubate a patient un-
tricular septum shift with or without impaired der the surgical drapes). Room for passage of a
left ventricular filling, increased pulmonary ar- larger bronchoscope into the trachea and bron-
tery pressure, systemic hypotension, tachycardia, chi then becomes available. The larger size scope,
decreased saturation, visible differential ventila- with greater suctioning capabilities, will ensure
tion of the lung if the pleura is opened, increasing more effective bronchial lavage. Bronchoscopy
requirement of inotropes to support the circula- also assists in repositioning a new ET tube endo-
tion and possible failure to separate the patient bronchially for direct ventilation and recruitment
from CPB. The afterload to the RV and the pos- of the collapsed lung. An example of this clinical
sibility of RV failure is greater if the tidal volume scenario is the cystic fibrosis patient population
and/or the RV preload are increased further in an in whom secretion management, bronchial toilet,

98 Neema ⭈ Manikandan ⭈ Rathod


Cardiopulmonary effects of high tidal Cardiopulmonary effects of modest or low
volume ventilation tidal volume ventilation
1. Overdistension of compliant alveoli 1. Decreased afterload to RV
2. Diversion of blood flow to collapsed alveoli 2. Improved oxygenation and ventilation
causing shunt effect 3. Decreased shunt effect
3. Increased RV afterload

Fig. 1. Schematic diagram explaining the mechanism of improvement in arterial blood gases and
decrease in RV afterload with changes in tidal volume in the presence of acute lung collapse.

and lung recruitment are components of the dis- and failure, and failure to wean the patient from
ease treatment [4]. CPB [7]. Logically, the appropriate management
in such a situation is to facilitate perfusion of the
well-ventilated areas of the lung by avoiding hy-
Measures That May Help Separation of the perinflation of the ventilated areas of the lung
Patient from CPB (fig. 1). However, how to select an appropriate
tidal volume in such a situation is not known. We
It is necessary to have a clear understanding of the believe that with modest tidal volume (5–7 ml/
underlying pathophysiology. Areas of lung col- kg) ventilation, the compression of the alveolar
lapse result in asymmetric lung compliance within vessels in the well-ventilated areas is likely to be
and between the two lungs and altered respiratory minimal. Perhaps visual assessment of the effects
mechanics [5]. In this situation, during IPPV, a of chosen tidal volume on airway pressure, lung
greater proportion of the tidal volume distributes inflation, and RV volume/distension may provide
to the more compliant areas of the lung. Hypoxic the key to the appropriateness of the chosen tidal
pulmonary vasoconstriction [6] in the areas of volume. A decreasing peak airway pressure indi-
lung collapse tends to divert blood flow toward cates alveolar recruitment and allows further in-
the ventilated areas of the lung and decreases the creases in tidal volume and RV preload. An im-
shunt effect. However, this beneficial effect, di- portant critical decision in the presence of acute
version of blood flow toward well-ventilated ar- lung collapse is the timing of reversal of antico-
eas of the lung, would be negated if the ventilated agulation. With protamine administration, pul-
areas are hyperinflated (fig. 1). Therefore, in the monary vasoconstriction is known that may re-
presence of significant lung collapse, IPPV with sult in increased afterload to RV and its failure;
normal as well as with larger tidal volume is ex- therefore, the administration of protamine should
pected to result in hyperinflation of ventilated ar- be delayed until hemodynamics and pulmonary
eas, increased afterload to the RV, its distention functions show improvement.

Acute Lung Collapse during Open-Heart Surgery 99


It should also be realized that full hemody- Besides these mechanical issues that result in
namic support for a certain period of time may be pulmonary dysfunction, there are systemic se-
needed; therefore, switching to an ECMO circuit quelae related to CPB that can have deleterious
should be considered as an alternative. Femoral physiologic effects as well. The inflammatory re-
vessels accept the cannulae required for full cir- sponse includes activation of mediators such as
culatory support, however in small children these interleukins, leukotrienes, and polymorphonu-
vessels are often small. In such situations, use of clear cells that have been implicated in lung in-
the carotid artery and internal jugular vein for jury. Leukocyte depletion, hemofiltration, use
vascular access provides an alternative access for of asanguineous prime, preoperative treatment
cannulation and ECMO support. The ECMO is with steroids, and intraoperative aprotinin have
a closed circuit, needs less anticoagulation (acti- all been used to attenuate the inflammatory re-
vated coagulation times ~160 to 180 s), relies on sponse. However, attempts to mitigate the inflam-
kinetic drainage, and allows for easy testing for matory response or its sequelae have not been
successful weaning. successful. In fact, Chaney et al. [12] showed that
patients receiving methylprednisolone actually
had worsened post-CPB pulmonary function. It
Measures That May Prevent Acute Lung was believed that aprotinin had anti-inflammato-
Collapse during CPB ry properties that might reduce some of the end-
organ damage seen, but given the controversies
During CPB the common practice to deflate lungs surrounding this drug, its routine use has been
can contribute to some of the pulmonary dysfunc- questioned.
tion seen post-CPB; strategies such as mechani-
cal ventilation [8] and CPAP have been tried [9,
10]. However, there has been little proven ben- Recommendations
efit to these measures. Earlier we use to deflate
the lungs during CPB, but of late we routinely ap- • Lung collapse during CPB, on discontinuation
ply 2–5 cm of CPAP during CPB depending on of ventilation, occurs invariably in patients
the surgeon’s comfort; a higher CPAP often forc- undergoing OHS. The collapsed lungs generally
es the lung in the surgical field particularly if the re-expand on manual inflation and resumption
pleura is opened. The value of CPAP lies in easy of IPPV. Minor areas of lung collapse are of no
re-expansion of the partially collapsed lungs on clinical significance.
manual inflation. ET suction, particularly in the • The clinical manifestations of significant lung
presence of left heart failure, chronic obstructive collapse during separation from CPB may
and restrictive pulmonary disease, and in poten- include raised central venous pressure, visible
tially infected patients, would ensure a clear air- or echocardiographic demonstration of RV
way and is useful. For thoracotomy approaches, distension with or without interventricular
Mittnacht et al. [11] describe lung isolation by us- septum shift with or without impaired left
ing a double-lumen endobronchial tube and ap- ventricular filling, increased pulmonary artery
plication of CPAP, during CPB, to the dependent pressure, systemic hypotension, tachycardia,
lung. In patients where insertion of a double-lu- decreased peripheral saturation, visible
men endobronchial tube is not possible, use of a differential ventilation of the lung if the pleura
bronchial blocker allows lung isolation, allowing is opened.
for both surgical exposure and dependent lung • Careful ET suction and/or repositioning of
ventilation. the ET tube generally resolves the airway

100 Neema ⭈ Manikandan ⭈ Rathod


issues. The fiberoptic bronchoscopy allows help separation of the patient from CPB. A
the anesthesiologist to visualize the ET tube decreasing peak airway pressure indicates
position, tracheobronchial anatomy, the alveolar recruitment and allows further
physical characteristics, the quantity, and the increases in tidal volume and RV preload. In
source of the secretions. However, suctioning the presence of lung collapse and increased
through small scopes may be suboptimal, central venous pressures, protamine should be
especially when thick secretions are present. administered carefully.
Bronchoscopy also assists in repositioning a new • Application of CPAP of 2–5 cm during CPB
ET tube endobronchially for direct ventilation allows easy re-expansion of the collapsed lung
and recruitment of the collapsed lung. on manual inflation.
• Ventilation with modest tidal volume (5–7
ml/kg) and careful preloading of the RV

References
1 West JB: Mechanics of breathing; in West 5 Carlon GC, Ray C Jr, Klein R, et al: Crite- 10 Loeckinger A, Kleinasser A, Linner KH,
JB (ed): Best and Taylor’s Physiological ria for selecting positive end expiratory et al: Continuous positive airway pres-
Basis of Medical Practice. New Delhi, pressure and independent synchronized sure at 10 cm H2O during cardiopulmo-
B.I. Waverly Pvt Ltd, 1996, pp 560–587. ventilation of each lung. Chest 1978;74: nary bypass improves postoperative gas
2 Pinsky MR: Heart-lung interactions; in 501–507. exchange. Anesth Analg 2000;91:522–
Grenvik A, Ayres SM, Holbrook PR, 6 Benumof JL: General respiratory physi- 527.
Shoemaker WC (eds): Textbook of Criti- ology and respiratory function during 11 Mittnacht AJ, Jashi U, Nguyen K, et al:
cal Care, ed 4. Noida/India, Harcourt anesthesia, in Benumof JL (ed): Anes- Continuous positive airway pressure and
Asia Pte Ltd, 2000, pp 1204–1221. thesia for Thoracic Surgery. Philadel- lung separation during cardiopulmonary
3 Rothen HU, Neumann P, Berglund JE, et phia, Saunders, 1995, pp 43–122. bypass to facilitate congenital heart sur-
al: Dynamics of re-expansion atelectasis 7 Neema PK, Manikandan S, Ahuja A, et gery via the right thorax in children.
during general anesthesia. Br J Anaesth al: Difficult weaning from cardiopulmo- Pediatr Anesth 2007;17:693–696.
1999;82:551–556. nary bypass in the lateral position 12 Chaney MA, Durazo-Arvizu RA,
4 Slieker MG, Van Gestel JP, Heijerman caused by lung collapse. J Cardiothorac Nikolov MP, et al: Methylprednisolone
HG, et al: Outcome of assisted ventila- Vasc Anesth 2008;22:616–624. does not benefit patients undergoing
tion for acute respiratory failure in cystic 8 Lake C, Booker PD: Pediatric Cardiac coronary artery bypass graft surgery. J
fibrosis. Intensive Care Med 2006;32: Anesthesia, ed 4. Philadelphia, Lippin- Thorac Cardiovasc Surg 2001;121:561–
754–758. cott Williams & Wilkins, 2005. 569.
9 Altmay E, Karaca P, Yurtseven N, et al:
Continuous positive airway pressure
does not improve lung function after
cardiac surgery. Can J Anesth 2006;53:
919–925.

Dr. Praveen Kumar Neema


B-9, NFH, Sree Chitra Residential Quarters
Poonthi Road, Kumarpuram, Trivandrum, Kerala 695011 (India)
Tel. +91 471 2557 321, Fax +91 471 2446 433
E-Mail neema@sctimst.ac.in, praveenneema@yahoo.co.in

Acute Lung Collapse during Open-Heart Surgery 101


Section Titlein Anesthesiology
Technology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 102–106

Anesthesia in the Intraoperative MRI Environment


Sergio D. Bergese ⭈ Erika G. Puente
Department of Anesthesiology, The Ohio State University, Columbus, Ohio, USA

Abbreviations is MRI-compatible or not, patient monitoring,


ASA American Society of Anesthesiologists
safety and personnel.
ECG Electrocardiography The design of the OR requires special consid-
iMRI Intraoperative magnetic resonance imaging erations in terms of size, logistics, and distribution
MR Magnetic resonance of space and equipment. The room must be larger
MRI Magnetic resonance imaging than the standard ORs in order to allow enough
OR Operating room space for the equipment and allow proper access to
the patient. The challenge of being able to balance
the requirements of access to the patient and the
MRI technology has gradually acquired a promi- magnetic field strength in and around the room
nent role in the iMRI environment. This modern must be taken into consideration. A larger room
equipment can provide substantial benefits when will allow enough space to install an MRI ma-
incorporated as a diagnostic, monitoring and chine, a portable shield, if required, and to move
treatment tool in the iMRI setting. At the same the patient in and out of the MRI core. A larger
time, incorporating the MRI technology can add room can also facilitate the transit of an increased
an extra challenge for the anesthesiologist and number of hospital personnel, who are required
healthcare providers involved. Safety, for both the in the iMRI suite to provide adequate care of the
patient and the OR staff, and adequate monitoring patient. The size of the room should also allow for
are the main concerns for the anesthesia provider the incorporation of special ‘MR-safe’ equipment,
when dealing with MRI equipment into the OR. designed to minimize interference from the mag-
As in many surgical environments, the primary netic field, and to ensure an adequate distance
goals of the anesthetic management in the iMRI from the location where this field is the stron-
setting are to avoid heart rate and blood pressure gest. Since these devices have some ferromagnet-
fluctuations, hypoxia, hypercapnia, to prevent in- ic properties, maintaining a specific distance from
creased venous pressure, and to allow for a rapid the magnet is required in order to avoid attraction
recovery, which in turn allows for an early neuro- of these objects into the magnet.
logical assessment [1]. The specific room design will be determined
In order to accomplish optimal anesthesia according to the use of the scanner and the types
management in the iMRI setting, there are several of procedures to be conducted in that OR. MRI
special considerations that need to be addressed, in the OR may be used for solely monitoring pur-
including the OR design, whether the equipment poses or for surgical guidance during a procedure.
These applications need to be taken into consid- noise trace. The Faraday cage has been specially
eration for the final design. Monitoring surgi- designed to minimize the impact of this low-ener-
cal progress and tissue changes, and confirming gy electric noise on the magnetic field and there-
the accomplishment of the surgical goal can be fore the quality of the images [3].
achieved with intermittent imaging performed In order to decrease the interference with the
at predetermined points during the operation. magnetic field as much as possible and achieve
Conversely, in the case of continuous real-time high-quality interpretation, the set-up of the OR
MR guidance of surgical tools such as biopsy nee- in preparation for the scanning is crucial. The set-
dles, endoscopes, or laser fibers, throughout the up usually involves a multi-step process from pa-
procedure demands a more sophisticated MRI tient positioning, to portable shield preparation,
system [2]. to monitoring equipment placement. The anes-
Currently, there are two types of ORs that thesiologist must take into consideration that the
have been designed which incorporate the MRI use of tubing extension is required in order to ad-
technology. In the first, the MRI is incorporat- equately reach the patient. It is also necessary to
ed into the permanent construction of the OR. have more than one intravenous access, teleme-
This is more frequently used with high-field MRI try ECG monitoring, fiberoptic temperature skin
technology. This structure is permanent and al- probes, pulse oximeter, blood pressure cuff, and
locates the OR entirely for iMRI use. The sec- vital signs monitors. All of these must be arranged
ond is a mobile design, which is compatible with properly so as to minimize the possibility of mov-
most models of low-field MRI scanners. This de- ing, interrupting, or disconnecting the breathing
sign requires the use of a portable shield or por- circuit, infusion lines, or monitoring cables with
table Faraday cage. This design renders the OR the Faraday cage. This preparation requires the
suitable for two different uses, with the iMRI and involvement of numerous trained hospital staff
without, the latter allows its use as a regular OR. in order to have the patient safely ready for the
The unique design of the portable cage requires a magnet to be positioned and be able to initiate
closed shield so that it collapses into the shape of the scanning. Barua et al. [4] described a learning
an accordion when not in use. The bottom of the curve effect in the set-up time that significantly
cage is a permanent stainless steel shield, which decreased the set-up time with increased training
is located under the OR table. The shiny finish of the staff.
of the stainless steel plate below the OR table can There are also certain constraints and hazards
reflect onto the images causing artifacts and cor- to consider when incorporating MRI technology
ruption on image appearance. Therefore, a matte into the OR environment. These are mainly re-
finish is recommended for the floor shield of the lated to the monitoring equipment, anesthesia
Faraday cage. machine, and infusion devices. According to the
Additionally, the bottom of the accordion- ASA, monitoring standards of the patient’s venti-
shaped shield must have an aluminum foil type lation, circulation, temperature, and oxygenation
wrapping that when making contact with the should be continuously evaluated throughout the
floor will act as an impermeable shield. It is criti- entire procedure. A restriction of the portable
cal to ensure that the bottom of the Faraday cage iMRI setting is that during the time the patient
surrounding the stainless steel plate is proper- is inside the Faraday cage, within the bore of the
ly positioned to provide adequate sealing of the scanner, they are not under direct supervision or
cage. Devices, such as infusion pumps, plasma visibility of the anesthesiologist for the duration
and liquid crystal displays, physiologic monitors, of the scan. During this period, it is essential to
and computers can generate a detectable electric assure adequate temperature control, continuous

Anesthesia in the Intraoperative MRI Environment 103


ventilatory support, intravenous drugs or con- equipment [6]. At the same time, the monitoring
trast media infusions, and inhalation anesthetics, equipment can interfere with the nuclear mag-
in compliance with the ASA guidelines [5]. The netic signals, leading to poor-quality images [1].
anesthesiologist must also be certain that the ap- Special non-ferromagnetic equipment has been
propriate drugs and equipment are readily avail- designed over time to decrease interference as
able in case of encountering unexpected airway or much as possible with the iMRI system, and in
cardiovascular complications [1]. turn obtaining the highest quality imaging as
The role of the anesthesiologist is most im- possible. Unfortunately, this results in consider-
portant when promoting safety in order to mini- able cost increments.
mize accidents associated with the iMRI technol- Non-magnetic MRI equipment available in-
ogy. To ensure adequate patient monitoring and cludes continuous infusion pumps and vital signs
supervision, the anesthesiologist must perform a monitors. There are also non-magnetic MRI lar-
detailed MRI compatibility preoperative exami- yngoscope handles and blades that function with
nation. This exam should include exploring for non-magnetic MRI lithium laryngoscope batter-
history of acquired or implanted metallic devices ies. It is important to point out that the recharge-
in the patient, such as cerebrovascular clips, car- able batteries should never be recharged in the
diac pacemakers, stents, bullets, braces, dentures, presence of the electromagnetic field.
or even extensive tattoos. These metallic devices Other important aspects to take into consid-
may generate imaging artifacts, may move from eration when providing anesthesia in the iMRI
their place and cause trauma, or may even heat up setting is the length of the different tubing sys-
and produce severe burns. tems required. In order for the intravenous tub-
The anesthesiologist must be hasty to request ing to reach the patient within the Faraday cage,
MR-compatible medical supplies to avoid such it is necessary to use extension tubing. Adequately
complications. For instance, a specific detail to long breathing tubes are also required due to the
remember when fastening the endotracheal tube remote location of the anesthesia machine. This
by inflating the safety balloon, the balloon needs extended anesthesia circuit may pose additional
to be drawn away from the patient’s face. The challenges for the anesthesia provider to maintain
safety balloon from the endotracheal tube has a adequate ventilation and administration of drugs.
metallic component that can cause serious burns The dead space from the extensions creates a time
to the patient’s skin. In some institutions the use delay upon the administration of volatile anes-
of temperature Foley catheters is very common. thetics and drugs before the expected onset of ef-
The anesthesiologist must be aware that for iMRI fects can be observed [3].
these catheters must not be used and instead, re- One of the most vulnerable signals affected by
placed with a standard Foley catheter without the high-energy radiofrequency pulses and elec-
a temperature probe. The temperature probe is tric noise generated by the iMRI technology is the
also metallic and can produce severe local burns ECG signaling. This can be decreased by the use
in the bladder and proximal urethra. For tem- of safeguarded cables, telemetry, or fiberoptic ma-
perature monitoring, a MRI-compatible probe is chinery [1]. The use of telemetry for ECG elimi-
recommended. nates the use of wires. The ECG electrodes must
During the scan, the magnet will generate be non-magnetic or ‘MR-safe’ to protect the pa-
a strong magnetic field that can interfere with tient from potential injuries and should be placed
monitoring and anesthetic equipment. This may carefully to minimize MRI-related artifacts [7].
affect or cause failure of electrical, electron- The use of special shielded non-magnetic MRI
ic, or mechanical life support and monitoring pulse oximeters is also recommended.

104 Bergese ⭈ Puente


Equipment can be modified so that it can be Recommendations
used within a range from the MRI scanner and
therefore can be designated as either ‘MR-safe’, • Ensure adequate patient safety by performing
‘MR-unsafe’ or ‘MR-conditional’. Equipment that a detailed MRI compatibility preoperative
is ‘MR-compatible’ not only is proven safe but examination (verify if the patient does not have
does not interfere with the quality of the scanner. acquired or implanted metallic device, such
Some special equipment considerations when as cerebrovascular clips, cardiac pacemakers,
monitoring a patient during iMRI [8–10] include stents, bullets, braces, dentures, or even
the following: (1) Blood pressure: utilize MRI-safe extensive tattoos).
or MRI-compatible blood pressure monitors. (2) • Ensure that the patients are aware of the
ECG: only MRI-compatible ECG pads should be procedure and know what to expect from the
used and the use of special MRI-compatible elec- use of the scanner during such procedure,
trocardiographic leads is recommended in or- e.g. use of additional tubing, loud noise of the
der to minimize interference with the readings. magnet during the scanning periods, colder
(3) Ventilation: consider visibility impairment of room temperature setting.
chest movements. We recommend the use of re- • Ensure that the anesthesia and monitoring
spiratory capnography that is usually available in equipment is MR-safe before being utilized
MR-compatible systems. (4) Oxygenation: to pre- and introduced into the iMRI setting.
vent severe burns from the pulse oximeter cables • Ensure that during the time the patient is
we recommend the use of MR-specific fiberop- not under the direct visual supervision of
tic pulse oximeters which do not overheat. (5) the anesthesiologist there is reliable and
Temperature: the magnet may produce overheat- accurate monitoring of pulse oximetry,
ing during the scan or the temperature may de- electroencephalographic signaling, blood
crease from the air conditioning required to pro- pressure and temperature control, and
tect the superconductors. continuous ventilatory support in compliance
Finally, patients should be educated on this with the ASA standard.
procedure prior to undergoing their surgery un- • Ensure that intravenous drug administration,
der iMRI techniques, as well as the hospital staff contrast media infusions, inhalation anes-
should be adequately trained and requested to thetics, and supplemental oxygen availability
complete a detailed MRI compatibility exam, and delivery follow ASA guidelines.
when required to ambulate within the iMRI • Ensure that the appropriate drugs and
setting. equipment are readily available in case of
encountering unexpected airway or cardio-
vascular complication.

References
1 Kraayenbrink M, McAnulty G: Neuroan- 2 Scott A: Interventional and Intraopera- 3 Bergese SD, Puente EG: Anesthesia in
esthesia; in Moore JA, Newell DW (eds): tive Magnetic Resonance Imaging. the intraoperative MRI environment.
Neurosurgery: Principles and Practice. Copyright Alberta Heritage Foundation Neurosurg Clin N Am 2009;20:155–162.
London, Springer, 2005. for Medical Research (2004). Available at
http://www.ihe.ca/documents/ip17.pdf
(accessed March 2010).

Anesthesia in the Intraoperative MRI Environment 105


4 Barua E, Johnston J, Fujii J, et al: Anes- 6 Bendo AA, Kass IS, Hartung J, et al: 9 Dorsch JA, Dorsch SE (eds): Equipment
thesia for brain tumor resection using Anesthesia for neurosurgery; in Barash for the Magnetic Resonance Imaging
intraoperative magnetic resonance with PG, Cullen BF, Stoelting RK (eds): Clini- Environment; in Understanding Anes-
the polestar N-20 system: experience cal Anesthesia, ed 5. Philadelphia, Lip- thesia Equipment, ed 5. Philadelphia,
and challenges. J Clin Anesth 2009;21: pincott-Raven, 2006. Lippincott Williams & Wilkins, 2008.
371–376. 7 Trankina MF: Neurodiagnostic proce- 10 American Society of Anesthesiologists:
5 Standards for Basic Anesthetic Monitor- dures; in Cucchiara RF, Black S, Michen- Practice advisory on anesthetic care for
ing Committee of Origin: Standards and felder JD (eds): Clinical Neuroanesthe- magnetic resonance imaging. A report
practice parameters. Approved by the sia, ed 2. New York, Churchill by the ASA task force on anesthetic care
House of Delegates of October 21, 1986, Livingstone, 1998. for magnetic resonance Imaging. Anes-
and last amended on October 25, 2005. 8 Stoelting RK, Miller RD: Basics of Anes- thesiology 2009;110:459–479.
Available at http://www.asahq.org/publi- thesia, ed 5. Philadelphia, Churchill Liv-
cationsAndServices/sgstoc.htm ingstone, 2007.
(accessed September 2009).

Erika G. Puente, MD
Department of Anesthesiology, The Ohio State University
410 W 10th Ave, N411 Doan Hall, Columbus, OH 43210 (USA)
Tel. +1 614 293 8487, Fax +1 614 293 8153
E-Mail Erika.puente@osumc.edu

106 Bergese ⭈ Puente


Technology in Anesthesiology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 107–113

Awake Thoracic Epidural Anesthesia Pulmonary


Resections
Eugenio Pompeo ⭈ Federico Tacconi ⭈ Tommaso Claudio Mineo
Cattedra di Chirurgia Toracica, Policlinico Università Tor Vergata, Rome, Italy

Abbreviations pathophysiological aspects remain to be elucidat-


TEA Thoracic epidural anesthesia
ed. Moreover, currently, few surgeons do perform
VATS Video-assisted thoracoscopic surgery awake thoracic surgery procedures and some
skepticism still survives regarding their feasibility
and potential advantages.
In this article we sought to review the current
VATS is now routinely employed to perform non- state of the art of this fascinating, newly available
anatomical pulmonary resections in a number of surgical option.
neoplastic and non-neoplastic conditions. In these
instances, general anesthesia with one-lung ven-
tilation is considered mandatory to accomplish a Background
safe operation. However, the use of this type of an-
esthesia can induce several adverse effects [1] that Videothoracoscopy
increase the overall invasiveness of the procedure The first thoracoscopy probably dates back to 1865
with a potentially negative impact on morbidity when the Irish physician Francis Richard Cruise
and hospitalization time. employed a modified Désormeaux’s cystoscope to
In order to avoid these detrimental effects, we perform a binocular thoracoscopy in an 11-year-
have started in 2001 a clinical research program old girl with empyema. Nonetheless, the Swedish
of VATS pulmonary resections performed by sole internist Hans Jacobaeus contributed to dissemi-
TEA in spontaneously ventilating, awake patients nate this minimally invasive technique that he ini-
[2–4]. So far, awake VATS resection of undeter- tially employed to sever pleural adhesions and thus
mined nodules [2], solitary metastases [3], and help lung collapse during the Forlanini’s artificial
even non-small cell lung cancer have been re- pneumothorax [5]. Following this pioneering work,
ported [4]. In these early series, results have been thoracoscopy remained underused for decades be-
encouraging although awake VATS pulmonary ing relegated to a minor role in management of un-
resections must be still considered investigation- determined pleural diseases and effusions.
al procedures since indications, comparative re- The eclipse of thoracoscopy lasted until the
sults with standard surgical approaches and many early 1990s when the introduction of video
technology and magnified imaging led to the ex- collapsed lung during one-lung ventilation pos-
plosive birth of the modern videothoracoscop- sibly due to generation of oxygen-derived free
ic surgery. This minimally invasive surgical ap- radicals as a consequence of a sort of ischemia
proach was immediately thought to mandate reperfusion tissue injury caused by intraoper-
general anesthesia and one-lung ventilation to al- ative-atelectasis-postoperative-reventilation of
low adequate surgical manipulation of the lung lung tissue. Moreover, Hemmerling et al. [12]
and easy accomplishment of several types of sur- have recently shown that 70% of patients under-
gical procedures including lung resections. going thoracic surgery with one-lung ventilation
longer than 1 h suffered from cerebral desatura-
Awake Thoracic Surgery tion >20% from their baseline. This level of deox-
As far as the use of regional anesthesia in thoracic ygenation is accepted as the threshold of cerebral
surgery is concerned, in 1950, Buckingham et al. ischemia and can be associated with a high inci-
[6] reported on 617 thoracic surgical operations dence of postoperative cognitive and major cere-
including thoracoplasties, pneumonectomies, bral dysfunction.
plumbage with acrylate pack, lobectomies, and Physiologic disturbances associated with sin-
open pneumolysis, performed through sole epidu- gle-lung ventilation in the lateral position include
ral anesthesia. More recently, use of regional anes- hypoxic pulmonary vasoconstriction in the non-
thesia in thoracoscopy was advocated in 1984 by dependent lung, an overall increase in venous ad-
Boutin et al. [7] in France and in 1987 by Rusch and mixture from about 10% to more than 27% [13], a
Mountain [8] in the USA to diagnose and manage decrease in oxygen partial pressure, and a change
pleural diseases. Reports of awake VATS pulmo- in alveolar-arterial oxygen tension gradient [14].
nary resection are much more recent and scant; In addition, one-lung ventilation has shown to
wedge resection was first reported in 1997 by Nezu enhance the hypothalamus-pituitary-adrenal axis
et al. [9] to resect blebs in patients with sponta- response to surgical stress [15], which eventually
neous pneumothorax, whereas more recently our impairs activity of natural killer cells. Yet, a com-
group reported on videothoracoscopic resection plex series of compartmental changes in both the
of undetermined pulmonary nodules [2]. ventilated and the non-ventilated lung can stimu-
late local release of pro-inflammatory mediators
and oxidative stress products [1].
Anesthesia The rationale for performing videothoraco-
scopic operations in awake patients through re-
Adverse Effects of General Anesthesia gional anesthesia is to avoid the adverse effects
The birth of modern thoracic surgery coincided of general anesthesia and mechanical ventilation,
with the development of double-lumen endo- maintain more physiologic muscular, neurolog-
bronchial tubes permitting single-lung ventila- ic and cardiopulmonary conditions, and create
tion that were adapted for surgery by Björk and a satisfactory environment for surgical maneu-
Carlens [10] in 1950. Unfortunately, despite some vering while providing optimal thoracic analge-
indisputable advantages, general anesthesia with sia. Nonetheless, some theoretical concerns arise
single-lung ventilation can cause several adverse in this regard since the need of operating on a
effects including hemodynamic disturbances and ventilating lung may reveal more technically de-
a multifactorial mechanical-ventilation-related manding and the idea still survives that surgi-
injury [1]. cal pneumothorax in a spontaneously ventilating
Tekinbas et al. [11] have recently shown that patient is poorly tolerated and increases opera-
biochemical and histopathologic injury occur in tive risk.

108 Pompeo ⭈ Tacconi ⭈ Mineo


Physiologic Effects of Epidural Anesthesia rib-cage-abdominal motion, and preserved abil-
In awake patients under local or epidural anes- ity to cough are all contributing factors that justify
thesia, we can attain the best type of monitoring, the better ventilation observed in these patients
namely neurologic vigilance. The net effect of TEA when compared to those operated on through
on lung function is mainly determined by the ex- general anesthesia.
tension of the motor blockade depending on the Cardiovascular effects of epidural anesthesia
height of the insertion of the catheter, the choice include decreased determinants of myocardial
of local anesthetic and its concentration. Evidence oxygen demand, improved myocardial blood flow
exists that TEA may potentially impair to some and left ventricular function, and reduced throm-
extent dynamic lung volumes. It has been sug- botic-related complications. Furthermore, it has
gested that following limited sensory block from been shown that epidural anesthesia can reduce
dermatome T1 to T5, vital capacity is decreased heart rate and occurrence of arrhythmias in pul-
by 5.6% and FEV1 by 4.9% [16], an effect which monary resections [16, 18].
can be explained by a direct motor blockade of
intercostal muscles. Yet, the effect of sympathi- Epidural Anesthesia in Awake VATS
colysis could result in an unopposed vagal tone TEA is carried out to achieve somatosensory and
leading to increased bronchial tone and reactiv- motor block from T1 to T8 level, while preserv-
ity. Nevertheless, we did not find any remarkable ing diaphragmatic motion. The epidural catheter
change in small airway flows and Tiffeneau index is inserted at the T4 level to achieve an optimal
during spirometric examinations performed dur- analgesia to the targeted hemithorax. Analgesia is
ing awake VATS. This seems to suggest that TEA induced with a 20-cc bolus of 2% ropivacaine +
had no significant detrimental effects on bronchi- sufentanil 5 μg/ml and the patient is placed in lat-
al tone in these patients. eral decubitus position for about 15–20 min with
TEA improves diaphragmatic contractility the side targeted for surgery in a dependent po-
[17] and breathing pattern, and provides better sition to help gravity distribution of anesthetics.
postoperative analgesia than patient-controlled Sensory level is tested every 5 min with warm-
intravenous administration of opioids. In fact, cold discrimination and/or pinprick tests. Care is
we have reasoned that one of the effects that con- taken to avoid extending sympathetic block over
tribute to keep respiratory function satisfactory T1 level, which can be disclosed by the develop-
throughout awake VATS, is the maintained dia- ment of a Bernard-Horner syndrome.
phragmatic motion that might decrease the det- Oral or intravenous premedication with ben-
rimental effect of the abdominal pressure leading zodiazepines is useful in anxious patients. We
the paralyzed diaphragm to compress the depen- recommend usage of short-acting sedatives at
dent lung during general anesthesia. minimum dosage to assure a light sedation while
Following general anesthesia, early postopera- maintaining adequate patient’s vigilance and
tive lung function is influenced by residual mus- awakeness. Intravenous opioids are preferably
cular relaxation, the time of extubation, pain ther- avoided due to their recently described potential
apy, and vigilance. In particular, immediately after depression of cortical centers controlling volun-
an operation, the ability to cough seems to be one tary respiration [19].
of the most important factors affecting lung func- Intraoperatively, continuous infusion of ropi-
tion and depends in great part on the efficacy of vacaine 0.5% with sufentanil 1.66 μg/ml is deliv-
diaphragmatic contraction and pain relief. ered into the epidural space at a rate of 5–7 ml/h
Conversely, in awake VATS, the absence of dia- via an elastomeric device. Peripheral oxygen satu-
phragmatic relaxation, better synchronization of ration is continuously monitored and additional

Awake VATS Pulmonary Resections 109


O2 is given through a Venturi mask only if room collapsed lung, which alters the physiologic re-
air ventilation leads to a drop of saturation below gional ventilation/perfusion ratios within the lung
90%. A potential intraoperative adverse effect is and seems to play a major role in determining the
the development of a panic attack, which can be degree of intraoperative hypoxia. However, in our
triggered by either an unsatisfactory analgesia or clinical experience, we have found that following
by dyspnea that may occur following induction the surgical pneumothorax, the decrease in arteri-
of the surgical pneumothorax, particularly in pa- al oxygenation is usually of limited extent and can
tients with poor pulmonary function and hyper- be corrected by oxygen administration through a
capnia. Although the exact etiology of panic at- Venturi mask. A hypothetical explanation of this
tacks is still unknown, it has been suggested that effect might be that a certain ventilatory excursion
an abnormal sensitivity to CO2 and disturbances is still maintained in the collapsed lung despite
of acid-base balance in brainstem could trigger an the presence of intrapleural atmospheric pressure
erroneous ‘suffocation alarm’ by the locus coerule- [22]. In addition, it is likely that in most instanc-
us neurons [20]. es a sufficient degree of compensatory ventilation
During wound closure, the epidural anesthet- is assured by the dependent lung whose efficien-
ic regimen is changed to ropivacaine 0.16% and cy is increased by the maintained diaphragmatic
sufentanil 1 μg/ml at 2–5 ml/h for postoperative function.
analgesia. The epidural catheter is removed 24–48 A frequent finding in awake VATS is the devel-
h after surgery. Postoperatively, liquids infusion is opment of intraoperative hypercarbia [23]. In this
stopped immediately and drinking, meal intake, respect, although the reduced tidal ventilation
ambulation and physiotherapy can be started on may be regarded as the major causative mecha-
the same day of surgery [4]. nism, this fails to explain why sometimes hyper-
carbia is not paralleled by a relevant decrease in
oxygenation. We hypothesize that perioperative-
Surgery ly, a certain degree of rebreathing effect may oc-
cur due to the existence of inter-pleural pressure
Physiolologic Effects of Surgical Pneumothorax gradients. Nonetheless, the increase in PaCO2 is
In physiologic conditions, intrapleural pressures usually well tolerated even in patients with severe
are negative throughout most of the respiratory emphysema [23] and rarely requires conversion
cycle. Instead, during awake videothoracoscopic to general anesthesia and mechanical ventilation.
surgery procedures, atmospheric air enters the Yet, we have shown that it resolves within 1 h after
pleural cavity and induces an immediate collapse the completion of the awake surgical procedure
of the non-dependent lung, the degree of which is [24] (fig. 1).
related to a series of factors including absence of
pleural adhesions anchoring the lung to the chest Awake VATS Resection of Undetermined
wall, the elastic properties of the lung and resis- Pulmonary Nodules
tances of the airways. Overall, all candidates suitable of videothoraco-
Regarding the effect of an open pneumotho- scopic resection of a pulmonary nodule and who
rax on oxygenation, some human and animal in- have no contraindications for TEA are theoreti-
vestigations suggested that changes in ventilation/ cally eligible for an awake approach. In a random-
perfusion ratio are more relevant that impair- ized comparison of 60 patients with undetermined
ment in ventilation. Anthonisen [21] found that solitary pulmonary nodules who underwent tho-
a pneumothorax of 25–40% in extent is associated racoscopic wedge resection through either sole
with an even redistribution of ventilation in the TEA or general anesthesia with double-lumen

110 Pompeo ⭈ Tacconi ⭈ Mineo


450 Normal lung Normal lung
Fibrosis Fibrosis
400 COPD 55 COPD
PaO2/FiO2 (mm Hg)

PaCO2 (mm Hg)


350 50

300 45

250 40

200 35

150
T1 T2 T3 T4 T1 T2 T3 T4

Fig. 1. Perioperative behavior of PaO2/FiO2 ratio and PaCO2 assessed at four fixed time intervals: T1 = closed chest in
lateral decubitus; T2 = after creation of the surgical pneumothorax; T3 = end of the surgical procedure; T4 = 1 h after
the operation.

intubation plus TEA [11], we had found no differ- Our surgical strategy in patients with pul-
ence in technical feasibility although 2 patients in monary metastases is based on the assumption
each group required conversion due to unexpect- that since iterative operations can be required to
ed lung cancer requiring lobectomy. Comparative prolong survival, it might be better to employ
results amongst study groups showed that anes- a less invasive surgical approach initially, while
thesia satisfaction score, changes in arterial oxy- deserving more aggressive ones for eventual re-
genation, need of nursing care and median hos- explorations in case of relapse. For this reason,
pital stay were significantly better in the awake in 1999 we had developed the transxiphoid ap-
group [2]. proach [25] that allowed to accomplish biman-
It is likely that in spontaneously ventilating ual palpation of the lung during VATS metas-
awake patients, avoidance of general anesthe- tasectomy to help identify all lung lesions that
sia- and single-lung-ventilation-related adverse might have been missed by sole instrumental
effects resulted in a more physiologic lung re- palpation.
expansion and a faster recovery with immediate More recently, we have proposed a new sur-
resumption of normal daily life activities. As a re- gical approach entailing awake VATS metasta-
sult, 47% of the patients in the awake group could sectomy through TEA [3] in order to minimize
be discharged within the second postoperative surgical and immunological stress that has been
day whereas this was possible only in 17% of pa- hypothesized as a potential cause of further can-
tients in the control group. cer metastasization.
Eligibility criteria include complete control of
Awake VATS Pulmonary Metastasectomy the primary tumor and absence of extrapulmo-
Videothoracoscopic pulmonary metastasectomy nary metastases, a newly discovered solitary pul-
is advocated in patients with single peripheral monary nodule localized in the peripheral one
lung lesions although the accuracy of digital and third of the lung and measuring <3 cm at the
instrumental lung palpation by VATS has been helical computed tomography. Exclusion crite-
questioned. ria included the presence of multiple metastases,

Awake VATS Pulmonary Resections 111


radiologic evidence of pleural scarring and/or a Conclusions
history of previous thoracic surgery on side tar-
geted for metastasectomy. Awake VATS pulmonary resections have now
In a 14-patient cohort undergoing awake VATS been successfully performed in many patients and
metastasectomy at our institution, the procedure in several pathologic conditions. Early series have
was easily and safely accomplished in all patients suggested that these procedures can assure a new
under sole TEA with no operative mortality or patient-friendly approach that minimizes opera-
major morbidity. Awake pulmonary metastasec- tive risks and permits short hospitalizations and
tomy resulted in optimal patient acceptance and immediate resumption of daily life activities.
satisfaction which was rated as excellent to good TEA has been preferred by our group as well
in 12 patients (86%). Hospital stay was signifi- as by other surgeons and allowed to achieve op-
cantly shorter than in a control group operated timal thoracic analgesia and few adverse effects.
through general anesthesia, while oncologic re- The main concern raised against awake VATS
sults and survival were similar [3]. procedures is the fear that surgical pneumothorax
is poorly tolerated by spontaneously ventilating
Awake VATS Resection of Lung Cancer awake patients. However, data that is progressive-
Resection of lung cancer represents the most pro- ly accumulating seems to contradict this empir-
vocative indication for awake VATS pulmonary ic thought showing that adequate ventilation is
resection although it is worth noting that in a re- assured in most of patients whereas satisfactory
cent series even awake anatomic lung resections oxygenation can be easily maintained through-
through thoracotomy have been reported by Al- out these procedures. Nonetheless, this fascinat-
Abdullatief et al. [26]. ing surgical option has opened several new doors
Proposed inclusion criteria for awake resec- that need light to be shed through. Many physio-
tion of lung cancer include peripheral stage I le- pathology aspects need to be better elucidated and
sions, poor pulmonary function or other impor- real advantages, disadvantages and cost-effective-
tant comorbidity leading to consider the patient ness still require further investigation to be defini-
high-risk for anatomical resection and/or even tively indicated. Surgery is changing and several
sole general anesthesia [4]. A further indication valid surgical procedures are likely to become ob-
for this approach might entail the resection of pe- solete within few years. Innovative non-surgical
ripheral tumors in medically inoperable patients options are being actively investigated to cure dis-
already treated with non-surgical local therapies eases avoiding surgical trauma and even the need
such as percutaneous radiofrequency ablation or for hospitalization. Thoracic surgeons of the third
stereotactic radioablation. millennium have to pick up the gauntlet and jump
Our initial results have been encouraging and on board of new less and less invasive surgical op-
in the first patients operated on there was no mor- tions. Awake thoracic surgery is expected to rep-
tality or major morbidity with satisfactory 2-year resent one strong answer to this challenge.
survival.

References
1 Gothart J: Lung injury after thoracic 2 Pompeo E, Mineo D, Rogliani P, et al: 3 Pompeo E, Mineo TC: Awake pulmonary
surgery and one-lung ventilation. Curr Feasibility and results of awake thoraco- metastasectomy. J Thorac Cardiovasc
Opin Anaesthsiol 2006;19:5–10. scopic resection of solitary pulmonary Surg 2007;133:960–966.
nodules. Ann Thorac Surg 2004;78:
1761–1768.

112 Pompeo ⭈ Tacconi ⭈ Mineo


4 Pompeo E, Mineo TC: Awake operative 12 Hemmerling TM, Bluteau MC, Kazan R, 20 Harvey H, Hayashi J, Speigel DR:
videothoracoscopic pulmonary resec- Bracco D: Significant decrease of cere- Chronic obstructive pulmonary disease
tions. Thorac Surg Clin 2008;18:311–320. bral oxygen saturation during single- and panic disorder: their interrelation-
5 Hoksch B, Birken-Bertsch H, Muller JM: lung ventilation measured using abso- ships and a unique utilization of beta-
Thoracoscopy before Jacobaeus. Ann lute oximetry. Br J Anaesth 2008;101: receptor agonists. Psychosomatics 2008;
Thorac Surg 2002;74:1288–1290. 870–875. 49:546.
6 Buckingham WW, Beatty AJ, Brasher 13 Bardoczky GI, Szegedi LL, d’Hollander 21 Anthonisen NR: Regional lung function
GA, Ottosen P: The technique of admin- AA, et al: Two-lung and one-lung venti- in spontaneous pneumothorax. Am Rev
istering epidural anesthesia in thoracic lation in patients with chronic obstruc- Respir Dis 1977;115:873–876.
surgery. Dis Chest 1950;17:561–568. tive pulmonary disease: the effects of 22 Subotic D, Mandaric D, Gajic M,
7 Boutin C, Velardocchio JM, position and FiO2. Anesth Analg 2000; Vukcevic M: Uncertainties in the current
Prud’homme A: Value of thoracoscopy. 90:35–41. understanding of gas exchange in spon-
Presse Med 1984;13:2635–2639. 14 Karzai W, Schwarzkopf K: Hypoxemia taneous pneumothorax: effective lung
8 Rusch VW, Mountain C: Thoracoscopy during one-lung ventilation. Prediction, ventilation may persist in smaller-sized
under regional anesthesia for the diag- prevention, and treatment. Anesthesiol- pneumothorax. Med Hypoth 2005;64:
nosis and management of pleural dis- ogy 2009;110:1402–1411. 1144–1149.
ease. Am J Surg 1987;154:274–278. 15 Tonnesen E, Hohndorf K, Lerbjerg G, 23 Mineo TC, Pompeo E, Mineo D, et al:
9 Nezu K, Kushibe K, Tojo T, et al: Thora- Christensen NJ, Huttel S, Andersen K: Awake non-resectional lung volume
coscopic wedge resection of blebs under Immunological and hormonal response reduction surgery. Ann Surg 2006;243:
local anesthesia with sedation for treat- to lung surgery during one-lung ventila- 131–136.
ment of a spontaneous pneumothorax. tion. Eur J Anaesth 1993;10:189–195. 24 Mineo TC: Epidural anesthesia in awake
Chest 1997;111:230–235. 16 Groeben H: Epidural anesthesia and pul- thoracic surgery. Eur J Cardiothorac
10 Björk VO, Carlens E: The prevention of monary function. J Anesth 2006;20:290– Surg 2007;32:13–19.
spread during pulmonary resection by 299. 25 Mineo TC, Pompeo E, Ambrogi V, et al:
use of a double-lumen catheter. J Thorac 17 Pansard JL, Mankikian B, Bertrand M, et Transxiphoid video-assisted approach
Surg 1950;20:151–157. al: Effects of thoracic extradural block for bilateral pulmonary metastasectomy.
11 Tekinbas C, Ulusoy H, Yulug E, et al: on diaphragmatic electrical activity and Ann Thorac Surg 1999;67:1808–1810.
One-lung ventilation: for how long? J contractility after upper abdominal sur- 26 Al-Abdullatief M, Wahood A, Al-Shirawi
Thorac Cardiovasc Surg 2007;134:405– gery. Anesthesiology 1993;78:63–71. N, et al: Awake anaesthesia for major
410. 18 Oka T, Ozawa Y, Ohkubo Y: Thoracic thoracic surgical procedures: an obser-
epidural bupivacaine attenuates vational study. Eur J Cardiothorac Surg
supraventricular tachyarrhythmias after 2007;32:346–350.
pulmonary resection. Anesth Analg
2001;93:253–259.
19 Pattinson KT, Governo RJ, MacIntosh BJ,
et al: Opioids depress cortical centers
responsible for the volitional control of
respiration. J Neurosci 2009;29:8177–
8186.

Prof. Eugenio Pompeo


Cattedra di Chirurgia Toracica, Policlinico Università Tor Vergata
Viale Oxford, 81, IT–00133 Rome (Italy)
Tel. +39 06 2090 2877, Fax +39 06 2090 2881
E-Mail pompeo@med.uniroma2.it

Awake VATS Pulmonary Resections 113


Section Titlein Transplants
Technology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 114–118

Preservation of Organs from Brain-Dead Donors


with Hyperbaric Oxygen
Benan Bayrakci
Pediatric Intensive Care Unit, Hacettepe University School of Medicine, Ankara, Turkey

Abbreviations techniques are limited. Organ preservation is a


eNOS Endothelial nitric oxide synthase
field of increasing importance due to the advances
iNOS Inducible nitric oxide synthase in transplantation medicine but unfortunately an
excellent system for the preservation of harvested
organs has not yet been defined. Hopefully, hy-
perbaric oxygen seems to be a promising candi-
Organ transplantation is an established therapeu- date as a bridge to transplantation by keeping the
tic modality for the end-stage organ failures. It has donated organs viable. A number of studies have
been used as a life-saving treatment modality for demonstrated that hyperbaric oxygen therapy
decades. As a matter of fact, the incremental ap- influences reperfusion injury and consequential
plication of organ transplantation increased the acute cellular rejection. Hyperbaric oxygen thera-
demand for donor organs. The main limitation of py is a technology that involves oxygen treatment
its wider application is the availability of suitable at supra-atmospheric pressures in high concen-
donor organs. This limitation put forwards the use trations, generating increased levels of physically
of marginal donor organs in order to increase the dissolved oxygen in plasma. This form of trans-
donor pool. Especially brain-dead donors consti- ported oxygen, compared with oxygen chemically
tute an alternative organ pool. However, the irre- bound to hemoglobin, is able to enter tissues with
versible loss of the brain functions inevitably leads almost no blood flow. Recent evidence also sug-
to a progressive deterioration in function of the gests a correlation between the severity of reper-
donor organs. The various described mechanisms fusion injury and acute cellular rejection which
of organ injury and cellular death are predomi- further causes organ damage. Acute cellular re-
nantly related to ischemia and reperfusion which jection affects approximately 30–40% of patients
account for the majority of graft loss, although after transplantation and adds significantly to the
the importance of a well-functioning organ in the postoperative morbidity and overall cost of trans-
recipient is of utmost importance for the success plantation. Hyperbaric oxygen therapy has been
of transplantation. The present methods of pre- shown to reduce the severity of reperfusion inju-
venting ischemia are regional or total body per- ry as well as modulate both humoral and cellular
fusion with chemicals and application of hypo- immune response. Organs procured from brain-
thermia. However, the beneficial effects of these dead hyperbaric oxygen-treated donors have less
cellular injury from ischemia, reperfusion, and particularly nitric oxide appears to play a role in
the no-reflow phenomenon, thus yielding organs this event. The cumulative effect of warm isch-
in an optimized state for transplantation. Treating emia and cold preservation is an energy deficien-
the harvested organs with hyperbaric oxygen car- cy within the endothelial cells leading to intracel-
ries the potential benefit of better preservation lular edema and exacerbating the hypoxic injury
prior to reimplantation. to the donor organ [3]. These detrimental events
are of paramount relevance in the global under-
standing of tissue injury, as transplanted organs
Main Topics undergo this process prior to, during and follow-
ing brain death, cold storage and finally at the time
Brain Death. The death of the brain is associated of reperfusion in the recipient [1]. Hyperbaric ox-
to an autonomic storm causing the massive release ygen treatment gives the tissues the opportunity
of circulating endogenous catecholamines. Light to be kept oxygenated throughout all entire pro-
and electron microscopy of organs procured from cess so neither a hypoxic nor a reperfusion state
experimental animals and human brain-dead or- can occur.
gan donors shows typical catecholamine-calcium- Acute Cellular Rejection. Acute cellular rejec-
induced injury, which results from the tissue isch- tion is an additional effect of reperfusion injury
emia-reperfusion [1]. Brain death leads to a series due to its potential to stimulate the host response.
of pathophysiological changes referred to as the The pathophysiology of acute cellular rejection is
autonomic storm (an initial period of excessive complex and has not been fully understood yet.
parasympathetic activity followed by a significant Many antigens that are not normally present are
adrenergic activity) which cause a significant im- expressed in response to reperfusion injury. This
pact on major organs used for transplantation exposure results in an innate immune response
[2]. The catecholamines activate lipases, proteas- that involves humoral components such as cy-
es, and endonucleases, while nitric oxide synthase tokines, complement and coagulation proteases
leads to further decay in membrane channel in- as well as cellular components made up of mac-
tegrity and increased oxygen free radical produc- rophages and neutrophils. This initial response is
tion. Oxygen free radicals participate in the per- followed by an adaptive immune response which
oxidation of lipids and injure the endothelium leads to the activation of T and B cells acting spe-
and cell membranes, resulting in a net loss of cel- cifically against exposed alloantigens. The host re-
lular integrity. Membrane permeability causes ex- sponse to donor alloantigens is centered on the
cess Ca2+ influx to the cell which results in further major histocompatibility complex antigens [4].
injury. Cellular aerobic oxidative respiration col- Activation of cytokines leads to the transforma-
lapses and the cellular oxygen lack results in in- tion of the T cells to take on immunoregulation
tracellular inhibition of cellular energy, although and cytotoxicity [5]. Hyperbaric oxygen therapy
ion gradients require constant energy at cellular has previously been shown to alter cell surface
channels. The failure of the Ca2+ channels precipi- major histocompatibility complex class I antigen
tate the Ca2+-induced injury [2]. expression and ultimately inhibit the transcrip-
Reperfusion Injury. After reimplantation of the tion of immunomodulatory cytokines, especially
organ the re-establishment of blood flow causes IL-2 along with its ability to reduce the expression
oxygenation of the ischemic tissues which adds a of MHC class I antigens [6].
new injury related to toxic oxygen free radicals. Hyperbaric Oxygen Therapy. Hyperbaric oxy-
The amount of tissue injury caused by reperfusion gen therapy has been used since 1662 for a wide
seems to be proportional to the ischemic time and spectrum of applications including diabetes

Preservation of Organs from Brain-Dead Donors with Hyperbaric Oxygen 115


mellitus, atherosclerosis, microangiopathy and injury, but it is still doubtful whether or not the
blood clotting disturbances, decompression sick- addition of hyperbaric oxygen to organ preserva-
ness, carbon monoxide poisoning, gas embolism tion algorithms might greatly prolong the storage
and radionecrosis. Hyperbaric oxygen therapy period.
increases the oxygen supply to the target tissue. Hyperbaric oxygen therapy has recently been
Recent experimental studies have suggested that shown to be a useful adjunct in several models
hyperoxemia provided by hyperbaric oxygen may of ischemic reperfusion injury, but its mechanism
be beneficial in the treatment of reperfusion in- of action remains mysterious. Oxygen is a criti-
jury. Hyperbaric oxygen was found to limit infarct cal substrate in the alleviation of hypoxia, anoxia
size in the reperfused rabbit heart and further evi- and ischemia, but paradoxically it also functions
dence that hyperbaric oxygen plays a role in resto- as a deleterious metabolite during the reperfu-
ration of oxidative enzyme activity was shown in a sion of previously ischemic tissues. In addition to
dog model [3, 7]. Hyperbaric oxygen therapy has this controversy, a hyperoxygenation and its me-
been used in enhancing the survival of the skin tabolites demonstrate cellular and clinical benefit,
grafts with approximately 50% less graft failures particularly in the field of ischemic reperfusion
[8]. Delivery of oxygen at high partial pressures in injury. Hyperbaric oxygen has been used with a
the plasma may attenuate endothelial cell hypox- beneficial therapeutic effect in a wide variety of
ia and reverse a potentially self-propagating cycle, models of reperfusion injury including myocar-
ischemia and response to ischemia that compro- dium, skeletal muscle, small intestine and the liv-
mises microvascular flow [9]. er. The majority of in vivo studies have shown a
reduction in the tissue injury when reperfusion is
performed in the presence of hyperbaric oxygen.
Discussion Hyperbaric oxygen interacts with the effects of
reperfusion injury at numerous interfaces, which
Organs used for transplantation undergo a se- include the endothelium, neutrophils, mediators
ries of injuries starting from the period before the of inflammation, microvascular blood flow, lipid
event of brain death, although relevance of a well- peroxidation and cellular energy levels. A number
functioning transplanted organ is clearly crucial of studies have confirmed the ability of hyperbaric
for the success of organs requiring immediate oxygen to selectively induce eNOS while inhibit-
function. The viability of any organ will depend ing iNOS production and the reduction of lipid
on how soon it depletes its oxygen and finally its peroxidation [6].
energy stores. Currently harvested organ pres- Another important role of hyperbaric treat-
ervation consists of using preservation solutions ment is immunomodulation. The effect of mini-
and cooling the organ to slow down metabolism mizing reperfusion injury is also known to reduce
but these only allow a limited time before trans- immune activation as it prevents the upregulation
plantation. Time of survival for the stored organ is of major histocompatibility antigen class II mol-
limited (maximum 48 h for kidneys and 12 h for ecules in the donor organ [6]. The mainstay of
liver). One other concern following a prolonged controlling acute cellular rejection remains with
anaerobic tissue state is reperfusion injury where immunosuppressive therapies. If the laboratory
oxygen radicals and superoxides destroy cellu- studies were proven in the clinical situation, it
lar components and compromise the success of may provide the opportunity to minimize drug-
transplantation. A hyperbaric preservation tech- based immunosuppression.
nique facilitates that the organ remains in an aero- Organs procured from brain-dead, hyper-
bic state thus preventing reperfusion and related baric oxygen-treated donors have less cellular

116 Bayrakci
injury from ischemia, reperfusion and the no- of hyperbaric oxygen therapy in the clinical set-
reflow phenomenon, thus yielding organs in an ting. Today, mini-hyperbaric oxygen chambers
optimized state for transplantation. Our clinical are available for biological materials. Such cham-
observation in 2 brain-dead donors supports the bers may help us overcome these current obsta-
fact that hyperbaric oxygen improves transplanta- cles. Treating the donor organ itself with hyper-
tion success. Both of the donors were CO intoxi- baric oxygen has the potential to carry the good
cation cases who had received hyperbaric oxygen effects of hyperbaric oxygen described above to
treatment and had to wait 72 h in brain-dead state the transplantation process. These mini organ
before the harvesting process took place because chambers have the ability to be mobile giving do-
of the toxic etiology. Due to the prolonged pro- nor organs the opportunity to receive hyperbaric
cess they suffered multiple cardiac arrest episodes oxygen throughout the whole harvesting to im-
causing additional ischemia-reperfusion injury to plantation process compromising the transport
the donor organs. Abundant organ functioning time as well.
after transplantation suggests a possible therapeu-
tic effect of hyperbaric oxygen treatment limiting
the ischemic insult generated from brain death Conclusions
and repetitive cardiac arrests. The frozen biopsy
obtained from the liver during transplantation Based on the available scientific evidence, hyper-
surgery, showing no evidence of necrosis and in- baric oxygen application seems to have the capac-
flammation, also gives a countenance to this sug- ity to influence the outcomes of transplantation at
gestion [10]. Hyperbaric oxygen is a promising multiple levels. Adding hyperbaric oxygen therapy
treatment as a bridge to transplantation, keep- into the organ preservation algorithm may be of
ing the donated organs viable until the harvest- considerable benefit, not only by keeping the or-
ing procedure take place for potential brain-dead gans oxygenated but also impairing the inevitable
donors. reperfusion injury. Keeping organs in a more op-
Hyperbaric oxygen therapy seems to reduce timal condition for transplantation is crucial for
the effect of reperfusion injury by various mecha- the success of transplantation. Hyperbaric oxygen
nisms depending on where it is applied, including greatly increases the viability of the organ while
exposure to the donor organ prior to reimplanta- awaiting a host either applied to the donor prior
tion as well as the recipient both before and after to the harvesting process or to the recipients after
transplantation [6]. Hyperbaric oxygen chambers reimplantation. Nevertheless, the most promis-
are both cumbersome and expensive. The diffi- ing type of application seems to be the hyperbaric
culty of moving critically ill patients in and out of treatment of the isolated donor organ because of
the chambers and the high infrastructure require- its ease of use, being handy for transport and of
ments have been major obstacles to the delivery relatively low cost.

References
1 Novitzky D: Donor management: state of 3 Thomas MP, Brown LA, Sponseller DR, 4 Pober JS, Orosz CG, Rose ML, Savage
the art. Transplant Proc 1997;29:3773– et al: Myocardial infarct size reduction CO: Can graft endothelial cells initiate a
3775. by the synergistic effect of hyperbaric host anti-graft immune response? Trans-
2 Novitzky D: Detrimental effects of brain oxygen and recombinant tissue plasmi- plantation 1996;61:343–349.
death on the potential organ donor. nogen activator. Am Heart J 1990;120:
Transplant Proc 1997;29:3770–3772. 791–800.

Preservation of Organs from Brain-Dead Donors with Hyperbaric Oxygen 117


5 Lipman ML, Stevens AC, Strom TB: 7 Sterling DL, Thornton JD, Swafford A, et 9 Glazier JJ: Attenuation of reperfusion
Heightened intragraft CTL gene expres- al: Hyperbaric oxygen limits infarct size microvascular ischemia by aqueous oxy-
sion in acutely rejecting renal allografts. in ischemic rabbit myocardium in vivo. gen: experimental and clinical observa-
J Immunol 1994;152:5120–5127. Circulation 1993;88:1931–1936. tions. Am Heart J 2005;149:580–584.
6 Muralidharan V, Christophi C: Hyper- 8 Svehlik J Jr, Zábavniková M, Guzanin S, 10 Bayrakci B: Preservation of organs from
baric oxygen therapy and liver trans- et al: Hyperbaric oxygenotherapy as a brain-dead donors with hyperbaric oxy-
plantation. HPB (Oxford) 2007;9:174– possible means of preventing ischemic gen. Pediatr Transplant 2008;12:506–
182. changes in skin grafts used for soft tissue 509.
defect closure. Acta Chir Plast 2007;49:
31–35.

Benan Bayrakci, MD
Pediatric Intensive Care Unit, Hacettepe University School of Medicine
Yeni Ladin Sitesi No: 19, Angora Caddesi
TR–06532 Beytepe, Çankaya, Ankara (Turkey)
Tel. +90 533 749 33 99, Fax +90 312 311 23 98
E-Mail benan@hacettepe.edu.tr

118 Bayrakci
Technology and Monitoring
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 119–125

Teleassistance in Chronic Respiratory Failure


Patients
Michele Vitacca
Divisione di Pneumologia Riabilitativa, Fondazione Salvatore Maugeri, IRCCS, Lumezzane, Italy

Abbreviations the same time, the additional time required for


COPD Chronic obstructive pulmonary disease
caregivers to provide transportation to physician
e-health Electronic health visits is usually not included in the cost analysis
GP General practitioner [3]. The family burden of HMV patients has been
HMV Home mechanical ventilation described to be particularly high with regard to
MV Mechanical ventilation money, which was reported to be excessive in
TA Teleassistance 17.3% of the cases [3].

Socioeconomic trends have led to a markedly Homecare Systems


increased interest in improving society’s abil-
ity to deliver effective care to older and chroni- Homecare for respiratory patients is a complex ar-
cally ill patients at home [1]. Challenges in the ray of services delivered in an uncontrolled set-
care of frail elders include the organization and ting in which patients and families are integral
sustainability of the continuum of services, re- members of the healthcare team [4]. Complexity,
source allocation and cultural competence in ser- lack of direct control, and acute exacerbations
vice delivery as case management programs and on chronic conditions all likely contribute to
partnerships with families [1]. In patients sub- the difficulty in organizing homecare assistance
mitted under HMV, underlying disease, level of [4]. Among homecare programs, HMV shows a
their dependency, hours spent under MV, pres- great prevalence in European countries. A recent
ence of tracheotomy, distance from home to hos- American Thoracic Society statement [4] has em-
pital, and hospital accesses are the causes of the phasized the need for a strict follow-up of these
major care burden [2] for the family and health- frail patients. In particular, homecare should fo-
care system. The time spent by caregivers to pro- cus on a patient-centered perspective and patient
vide assistance for patients with poor daily life and family satisfaction: reduction of complica-
activities has been described as enormous [3]. At tions resulting from hospitalization, maintaining
Integrated tele-chronic care

Second
opinion
(pulmo, cardio,
Patient-centered neurol, psychol,
pharmacy)

Devices
Fig. 1. Operative chain for the Instruments
tele-chronic care program from our
group, with permission. The inte-
grated tele-chronic care program
offers a network between the GP,
Basal
patient, family, call center and nurse network: GP,
tutor. According to different dis- patient,
eases and level of severity, devices call center
are prescribed and inserted into the nurse tutor
program. A multidisciplinary sani-
tary staff is available for a second
opinion 24 hours a day.

an acceptable quality of life, and enabling a com- enables remote, isolated and rural areas to have
fortable and dignified death have been proposed clinical support from those hospitals and medi-
as major endpoints [4]. cal systems with a higher level of medical exper-
tise [5–7]. Telemedicine can be classified on the
basis of the mode of operation or on the basis of
Telemedicine − Telesupport − Teleassistance application [5]. According to the mode of opera-
tion it can be divided into two groups: real-time
Recent advances in technology make it possible (interactive) mode and store-and-forward mode.
to reach patients in their homes through e-health In the real-time interactive mode, the patient is
or telemedicine. Telemedicine is a very useful present with an attending physician or paramedi-
and critical application of information and com- cal personnel, and a specialist is present at a re-
munication technologies, and may deliver high- mote medical center. In the store-and-forward
quality healthcare services regionally, national- mode, all relevant information (data, graphics,
ly, or globally. Telemedicine is a combination of images, etc.) is transmitted electronically to the
medical expertise, medical equipment, comput- specialist. It is possible to distinguish two similar
er hardware and software, and communication methods: one in which telemedicine is an aid for
technology, by which a patient can be examined, helping the nurse in the management of patients
investigated, monitored and treated by a medi- with chronic diseases (telenursing), and the oth-
cal expert in a distant place [5]. ‘Tele’ is a Greek er, completely automated, in which a computer-
word meaning ‘distance’ and ‘mederi’ is a Latin ized voice-answering system and a computer al-
word meaning ‘to heal’. Hence, telemedicine is gorithm checked the patient’s vital signs with an
‘medicine practiced at a distance’ [5]. Temedicine acceptable range previously set by the physician

120 Vitacca
NMV IMV

RR

VE

VTe

VTi

Paw

Fig. 2. From top to bottom: representative tracings sent by phone to call center of respiratory rate, minute ventilation,
expiratory tidal volume, inspiratory tidal volume, airway pressure, pSat O2 and heart rate trends during non-invasive
mechanical ventilation (NMV) (a left, b bottom) and during invasive mechanical ventilation (IMV) (a right).

(telemanagement). Nurse-telephone triage lines program, while figure 2 shows representative


have been running for over 10 years. The nurses tracings of biological signs transmitted by means
who answer the phones are specially trained and of TA. The three main types of benefit arising
use algorithms on computers to steer the conversa- from the e-health investment are quality, access
tion to a particular conclusion. Figure 1 shows an and efficiency. Table 1 summarizes the main ben-
example of an operative chain for a telemedicine efits and opportunities of e-health.

Teleassistance in Chronic Respiratory Failure Patients 121


Table 1. Main benefits and opportunities of e-health

Supporting the delivery of care tailored to individual patients


Improving transparency and accountability of care processes and facilitating shared care across boundaries
Aiding evidence-based practice and error reduction
Improving diagnostic accuracy and treatment appropriateness
Improving access to effective healthcare by reducing barriers created
Facilitating patient empowerment for self-care and health decision-making
Improving cost efficiency by streamlining processes, reducing waiting times and waste
More responsibility of referral center
More information among actors
Educational model
‘Self-responsibility’ of patient and caregiver
Transmission of data and real-time consultation
Complimentary to classical models
Reducing omission of care
Increasing continuity
Increasing effectiveness
Prompt attention to emergencies
Improving patient satisfaction
Improving effectiveness
Reducing costs
Reducing communication errors

Results from the Literature admissions and days of hospital stay. Young et
al. [10] described the general application of tele-
The Kaiser Permanente Organization study [8] phone-linked communication systems in the
reported the first formal randomized controlled healthcare setting for COPD patients discuss-
trial of home videophones. Patients in the inter- ing the rationale for expected improvements
vention group were equipped with home video- in disease control and quality of life, and for a
phones, an electronic stethoscope and a digital reduction in acute healthcare utilization. Cost
blood pressure monitor; over 18 months, pa- and effectiveness data from the Milwaukee and
tients in the telemedicine group received 17% Iron Mountain Veterans Affairs Medical Centers
fewer home visits by nurses than the control telepulmonary program [11] were collected for
group and the average cost of care in this group a period of 1 year. Telemedicine was found to be
was 27% less than in the control group [8]. In an more cost effective (USD 335 per patient/year)
intervention group of 46 patients, Farrero et al. compared to routine care (USD 585 per patient/
[9] studied monthly telephone calls, home visits year) and on-site care (USD 1,166 per patient/
every 3 months, and home or hospital visits on a year). Hernandez et al. [12] tested a home vs.
demand basis. 48 patients as control group com- conventional hospitalization with a random-
pleted the 1-year follow-up period. The authors ized study of 8 weeks. 222 COPD patients ad-
found a decrease in the mean number of emer- mitted to the emergency room received an in-
gency department visits, decrease in hospital tegrated care with a specialized nurse, a home

122 Vitacca
visit within 24 h and patient free phone access. this perspective, we [17] should first ask whether
Rehospitalization, emergency room visits, days a TA program could be effective also in severe
of hospitalization and quality of life were signifi- patients on long-term oxygen therapy and HMV.
cantly in favor of the homecare group. Bourbeau In line with Casas et al. [16] and Maiolo et al.
et al. [13] compared a treated group with an edu- [14], our study confirms that an integrated mul-
cation program, nurse weekly visit, monthly tele- tidisciplinary monitoring and care with the aid
phone follow-up with a control group with usual of information technologies can reduce hospital-
care. Maiolo et al. [14] for 12 months followed izations by about 36%, GP urgent calls by 65%,
23 patients with chronic respiratory failure with home relapses by 71%, even in more severe pa-
a fixed twice-a-week transmission of pSat and tients. According to our primary endpoint (re-
heart rate. Comparing the healthcare utilization duction of hospitalizations), this study also con-
in the year before the study they demonstrated firmed that patients affected with COPD seem
a reduction in hospital admission and home re- to take a greater advantage from TA. We [17]
lapses of 50 and 55% respectively. Patients were have confirmed the key role of nurses as a spe-
satisfied with the quality of the personal telem- cialized figure able to educate patients and their
onitoring process in 96% of cases. Eighteen [15] families/caregivers before discharge, to screen
well-motivated patients with advanced COPD, all requests and to coordinate all actors involved
who had had at least four hospitalizations dur- in the follow-up.
ing the previous 2 years, were included in a
home-based telemedicine program including
nurse visits, a laptop computer, an electrocardio- Conclusion
gram, spirometer, oximeter and blood pressure
monitor, and a videoconference camera. After 9 The real ‘technology’ includes human resources
months there was a decrease in hospitalizations, available in hospitals and home and social health
emergency department visits and use of health organizations; TM models need to respond to cri-
services. Casas at al. [16] demonstrated with a teria of equity, simplicity, efficiency, efficacy, and
randomized controlled trial on 155 exacerbated have to be patient-centered and safe. The chal-
COPD patients that a standardized integrated lenge that telemedicine will become part of the
care intervention, based on shared care arrange- standard of care remains open.
ments among different levels of the system with
support of information technologies, effective-
ly prevents hospitalizations for exacerbations in Recommendations
COPD patients. In their study [16], only 24%
of patients were on long-term oxygen therapy • Hospitalization of chronically ill patients is
while none was mechanically ventilated. In this a ‘failure’ for the health system and chronic
study, periodic phone calls were scheduled ev- diseases are becoming the case for the large-
ery 3 months as ‘store-and-forward necessity’. scale deployment of e-health.
We [17] have recently demonstrated that the TA • e-Health may be an opportunity for health
program is effective in preventing hospitaliza- organization, new strategic politician vision
tions, home acute exacerbations, and urgent GP and clinical reorganization. Table 2 sum-
calls, and may be cost-effective in severe chronic marizes the recommendations and limitations
respiratory failure patients needing home oxy- for telecare.
gen therapy and/or MV. The COPD group seems • The future of homecare and telemedicine will
to take the greater advantage from TA [17]. In depend on (i) human factors, (ii) economics,

Teleassistance in Chronic Respiratory Failure Patients 123


and (iii) technology. Patients need healthcare Table 2. Recommendations and limitations for telecare
and they want to remain at home for as long as
possible. Recommendations
Clearly and articulated mission
• While intervention trials have utilized different Specific goals to be achieved
designs, the nurse practitioner is common to Accountable governance structure
all the trials and appears to have a key role in Well-defined service
the management of chronic disease. Well-defined target population
• Results on long-term effect, cost-effective Well-structured service providers
Detailed procedures and protocols (scheduled
analysis, quality of life, and public health
quality controls)
burden reduction remain to be demonstrated. Correct choice of technology adequate for each
• Nowadays a lot of applications of homecare and situation
telemedicine are possible and operative. Home Well-structured outcomes perspective
telemedicine is not strictly ‘technology’ but an Self-sustaining programs
innovative instrument (based on health figures Limitations
more than high-tech instruments) which will Many different software, hardware and telecom-
help the daily job of doctors with patients and munication options
their families: telemedicine changes the way we Poor specification design for each condition
deliver care to patients by changing the space Poor uniformity for standards
No clear strategies for promoting wide utilization
and time in the relations between patients and
Limited technological and clinical research
health professionals. Legal problems between subjects involved
Costs
Poor knowledge and culture
Skepticism of doctors
Absence of reimbursements

References
1 Young HM: Challenges and solutions for 4 American Thoracic Society Docu- 8 Johnston B, Wheeler L, Deuser J, Sousa
care of frail older adults. Online J Issues ments: Statement on home care for KH: Outcomes of the Kaiser Permanente
Nurs 2003;8:5. WHO Global Observatory patients with respiratory disorders. Am Tele-Home Health Research Project.
for e-Health. Global e-Health Survey, J Respir Crit Care Med 2005;171,1443– Arch Fam Med 2000;9:40–45.
Geneva, July 2005. 1464. 9 Farrero E, Escarrabill J, Prats E, Maderal
2 Vitacca M, Escarrabill J, Galavotti G, 5 Scalvini S, Vitacca M, Paletta L, Gior- M, Manresa F: Impact of a hospital-
Vianello A, Prats E, Scala R, Peratoner A, dano A, Balbi B: ‘Telemedicine: a new based homecare program on the man-
Guffanti E, Maggi L, Barbano L, Balbi B: frontier for effective healthcare services. agement of COPD patients receiving
Home mechanical ventilation patients: a Monaldi Arch Chest Disease long-term oxygen therapy. Chest 2001;
retrospective survey to identify level of 2004;61:226–233. 19:364–369.
burden in real life. Monaldi Arch Chest 6 Wootton R: Telemedicine: clinical 10 Young M, Sparrow D, Gottlieb D, Selim
Dis 2007;67:142–147. review. BMJ 2001;323:557–560. A, Friedman R: A telephone-linked com-
3 Langa KM, Fendrick AM, Flaherty KR, 7 Wooton R, Dimmick SL, Kvedar JC puter system for COPD care. Chest 2001;
Martinez FJ, Kabeto MU, Saint S: Infor- (eds): Home Telehealth: Connecting 119:1565–1575.
mal caregiving for chronic lung disease Care within the Community. London,
among older Americans. Chest 2002; The Royal Society Medicine Press,
122:2197–2203. 2006.

124 Vitacca
11 Agha Z, Schapira RM, Maker AH: Cost- 13 Bourbeau J, Julien M, Maltais F, Rouleau 16 Casas A, Troosters T, Garcia-Aymerich J,
effectiveness of telemedicine for the M, Beaupré A, Bégin R, Renzi P, Nault D, Roca J, Hernández C, Alonso A, del Pozo
delivery of outpatient pulmonary care to Borycki E, Schwartzman K, Singh R, F, de Toledo P, Antó JM, Rodríguez-Roi-
a rural population. Telemed J E Health Collet JP: Chronic Obstructive Pulmo- sín R, Decramer M: Integrated care pre-
2002;8:281–291. nary Disease Axis of the Respiratory vents hospitalizations for exacerbations
12 Hernandez C, Casas A, Escarrabill J, Network Fonds de la Recherche en Santé in COPD patients. Eur Respir J 2006;
Alonso J, Puig-Junoy J, Farrero E, Vilagut du Québec: Reduction of hospital utili- 28:123–130.
G, Collvinent B, Rodriguez-Roisin R, zation in patients with chronic obstruc- 17 Vitacca M, Bianchi L, Guerra A, Fracchia
Roca J: Home hospitalisation of exacer- tive pulmonary disease a disease-specific C, Spanevello A, Balbi B, Scalvini S: Tele-
bated chronic obstructive pulmonary self-management intervention. Arch assistance in chronic respiratory failure
disease patients. Eur Respir J 2003; Intern Med 2003;163:585–591. patients: a randomised clinical trial Eur
21:58–67. 14 Maiolo C, Mohamed EI, Fiorani CM, De Respir J 2009;33:411–418.
Lorenzo A: Home tele-monitoring for
patients with severe respiratory illnesses:
the Italian experience. J Telemed Tele-
care 2003;9:67–71.
15 Vontetsianos T, Giovas P, Katsara T,
Rigopoulou A, Mpirmpa G, Giaboudakis
P, Koyrelea S, Kontopyrgias G, Tsoulkas
B: Telemedicine-assisted home support
for patients with advanced chronic
obstructive pulmonary disease: prelimi-
nary results after a nine-month follow-
up. J Telemed Telecare 2005;11(suppl 1):
86–88.

Michele Vitacca, MD
Divisione di Pneumologia Riabilitativa, Fondazione Salvatore Maugeri
IRCCS, Via Mazzini 129, IT–25066 Lumezzane/BS (Italy)
Tel. +39 030 825 3168, Fax +39 030 825 3188
E-Mail michele.vitacca@fsm.it

Teleassistance in Chronic Respiratory Failure Patients 125


Section Titleand Monitoring
Technology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 126–131

Capnography: Gradient PACO2 and PETCO2


Michael E. Donnellan
Oridion Capnography Inc., Needham, Mass., USA

Abbreviations a normal healthy individual can be closely corre-


ARDS Adult respiratory distress syndrome
lated suggesting the PETCO2 may be able to pre-
CO2 Carbon dioxide dict the PaCO2.
FRC Functional residual capacity The gradient is largely dependent upon the
PaCO2 Partial pressure arterial carbon dioxide physiological dead space and the slope of the al-
P(a-ET)CO2 Gradient veolar plateau displayed in phase lll of the graphic
PaO2 Partial pressure of oxygen waveform in figure 1. An increase in dead space
PEEP Positive end-expiratory pressure can result in a corresponding increase in the
PETCO2 End-tidal carbon dioxide PaCO2-PETCO2 gradient. However, a negative
Qsp/QT Physiological shunt
relationship can also exist between the PaCO2-
VD/VT Dead-space tidal volume ratio
PETCO2 and the slope in phase lll where an in-
V/Q ratio Ventilation-perfusion ratio
crease can indicate a reduction in the gradient.
Subsequently, it may be stated that the net change
in the PaCO2-PETCO2 is the result of changes in
Capnography provides a numeric and graphic dead space and the patterns of the various V/Q
wave display of measured fractional CO2 in real alveoli [4–6].
time with breath-to-breath feedback known as P(a-ET)CO2 is dependent and influenced
PETCO2 monitoring providing feedback on pa- upon underlying pulmonary disease in patients
tient’s clinical standing by determining and track- where the PETCO2 can differ from the PaCO2
ing the baseline ventilatory status over time [1, because of V/Q mismatching. There also may
2]. A capnometer displays a number (PETCO2) be wide variations in gradients when a patient‘s
whereas a capnograph displays a number and a condition does not remain constant over time
waveform (capnogram) (fig. 1), both monitoring [7, 8].
changes in CO2 concentration during the respira-
tory cycle [3].
The difference between the PETCO2 and Trending
PaCO2 is referred to as the gradient which is a re-
sult of the relationship between V (ventilation) A constant PETCO2 does not insure a constant
airflow to the alveoli and Q (perfusion) blood flow PaCO2, especially in pre-hospital ventilatory
to the capillaries. This is the V/Q ratio which in management where fluctuations in the gradient
% CO2 max Normal capnogram Normal EtCO2: 35–45 mm Hg
I II III IV
CO2 (mm Hg)
100 S4
50 Real-time Trend
S3
75 CV D
37 C
A1 AB E
50 0
S2 A2
Waveform characteristic:
25 • A–B Baseline •D ETCO2 value
• B–C Expiratory upstroke • D–E Inspiration
V • C–D Expiratory plateau
V2550
V5075

Fig. 2. A ‘normal’ capnogram is a waveform representing


the varying CO2 level throughout the breath cycle.
Fig. 1. Capnogram displaying a number and a
waveform.

are common over time. In mechanically ventilat- Case Reviews


ed patients, changes in the individual parameters
such as rate and tidal volumes can contribute to Chronic Obstructive Pulmonary Disease
the variability in the PaCO2-PETCO2 gradient Figure 3 is the breath-to-breath tracing of a se-
[8]. However, although these variations in the vere tachypneic chronic obstructive pulmonary
gradient may not be used to predict the PaCO2, disease patient. The breaths appear compressed
it remains useful in documenting changes in V/Q displaying a slight variation in PETCO2. The
mismatches and can alert the clinician to chang- PaCO2 was 74 mm Hg resulting in a gradient dif-
ing physiological conditions [9]. ference of 24 mm Hg. This is an example where
There are other reviews and opinions claiming the diseased alveoli are not emptying evenly re-
the PETCO2 will closely approximate the PaCO2 sulting in the end-tidal sample containing con-
or may differ by an acceptable constant or range, siderable dead-space air. While the 24 mm Hg
especially in intubated patients where a closed difference in this gradient is considered high (a
ventilatory system exists. In these scenarios, the normal value being between 2 and 6 mm Hg), it
PaCO2 may no longer require as frequent moni- renders the PETCO2 useful in determining the
toring and comparisons [9] (fig. 2). It is gener- PaCO2 when subject to occasional validation in
ally accepted that a sizeable difference in the gra- light of the complete clinical picture in real time.
dient does not diminish or preclude the value of Given the assumption that a verified PETCO2 can
the PETCO2 and that a rise in the PETCO2 indi- act as a proxy for PaCO2, it can be a valuable tool
cates a rise in the PaCO2. However, equating the in intensive care and will reduce arterial blood
exact numeric values is not advised and at least sampling [10].
two comparisons should be obtained between the
PaCO2-PETCO2 before completely relying upon ARDS PEEP Titration
the PETCO2, especially in patients that are clini- V/Q mismatching is considered a primary cause
cally stable [10]. for hypoxemia in the ARDS patient where the dis-
tribution of V/Q ratios are altered as shown by

Capnography: Gradient PACO2 and PETCO2 127


ICU patients with respiratory distress, not requir-
ing immediate tracheal intubation, were man-
aged in a standard manner to include arterial
blood gas monitoring (i-Stat® Analyzer; Abbott
Laboratories, Ill., USA) and the PETCO2 by a
PCO2 = 50 cannula (Microstream Smart Capnoline Plus®;
Oridion Inc., Needham, Mass., USA) in order to
determine and monitor the PaCO2-PETCO2 gra-
dient [15]. Of the 49 patients reviewed, the gra-
PCO2 = 0
dient between the PaCO2-PETCO2 was >5 mm
Hg in 41 of the patients and >10 mm Hg in 25 of
the patients. The maximum gradient was 34 mm
Fig. 3. Breath-to-breath tracing of a severe tachypneic
Hg. It was determined that although capnography
chronic obstructive pulmonary disease.
monitoring in this non-intubated patient popu-
lation with respiratory distress presented mini-
mal problems, the PETCO2 poorly reflected the
multiple inert elimination techniques [11]. PEEP PaCO2 and the PaCO2-PETCO2 experienced the
has been shown to increase FRC and directly effect largest gradient in the hypersonic and tachypne-
intrapulmonary shunting Qsp/QT (physiological ic patients. The accuracy was incompatible with
shunt) while improving oxygenation in ARDS pa- routine clinical use of PETCO2 and as a predictor
tients when applying various levels of PEEP [12]. of the PaCO2 [16].
However, determining the level of PEEP where the
beneficial effects supersede harmful ones is impor- Massive Pulmonary Emboli
tant when titrating the PEEP, improving the PaO2 Six patients with hypertension undergoing
and minimizing the effect upon the cardiac out- thrombolysis for massive pulmonary emboli were
put, dead-space volume, and barotraumas [13]. evaluated by PaCO2-PETCO2 gradient monitor-
It has been observed that the PaCO2-PETCO2 ing for several hours after the treatment. Two of
gradient decreases when PEEP is applied and the patients experienced a large decrease in the
minimized at 15 cm H2O, but increased again at PaCO2-PETCO2 gradient (from 17 mm Hg be-
20 cm H2O. The VD/VT value had a direct and fore treatment to 1.5 mm Hg) at 2–4 h and 12–24
corresponding cause of action with the PaCO2- h respectively. In the initial attempt to evaluate
PETCO2 gradient. The PaO2 is correspondingly the use of capnography and a gradient during the
at its highest when the PaCO2-PETCO2 gradient treatment of massive pulmonary emboli, it was
is minimal and intrapulmonary shunts decrease suggested that the decrease in the gradient sug-
with increasing levels of PEEP and was minimal gests the efficacy of the thrombolysis and the el-
at 20 cm H2O [14]. The values are easily moni- evated persistent PaCO2-PETCO2 gradient may
tored and a trend can be seen with simple arterial question the efficacy of the treatment. However,
blood gas analysis and simple capnograms. The capnography shows promise as a non-invasive
PaCO2-PETCO2 gradient is a useful and simple bedside monitoring application in this treatment
value too. [17].

Respiratory Distress Non-Intubated Patient Newborns


In a study conducted in the outpatient emergen- Two groups of mechanically ventilated new-
cy unit of a French University Hospital, 5 mobile borns at Children’s Hospital Boston, Mass., were

128 Donnellan
separated into two sections: group A with pulmo- A homogeneous lung model that generated
nary disease and group B without pulmonary dis- five non-linear first-order differential equations
ease. 20 infants were selected and gas sampling and two equations for gas exchange implemented
performed using a sidestream hand-held device mathematical modeling describing variations in
(Microcap®; Oridion Inc.) with microstream sam- CO2 and O2 compartmental fractions and alve-
pling technology [18]. olar volumes to include pulmonary capillary gas
PaCO2-PETCO2 gradients were collected from exchange. The PaCO2-PETCO2 between adjust-
13 of the patients with pulmonary disease and 7 ed experimental and sinusoidal ventilatory flow
without. The patients in group A were diagnosed rates while at rest and during exercise indicated
with persistent pulmonary hypertension, respi- similar values in PaCO2 and PETCO2 for differ-
ratory distress syndrome and pneumonia, while ent flow dynamics. The model studies regarding
group B was free of pulmonary disease. Birth the effects of metabolic, circulatory and cardiac
weight, gestational age, postnatal age and mode output and respiratory parameters indicated a sig-
of ventilation were similar. The mean PaCO2- nificant difference in the PaCO2-PETCO2 during
PETCO2 gradients were significantly different exercise [21].
between groups A and B, demonstrating wide
gradients in group A with pulmonary disease.
However, it is noted that because tidal volumes Discussion
are small and respiratory rates are high in this
patient population, PaCO2 was diluted with dead- Monitoring the physiological dead space is a criti-
space gas from the ventilator circuit. As a result, cal value in determining a pathway of clinical care
sidestream technology that continuously samples for the patient. Specific numeric determinations
exhaled gases from a side port on the proximal of the VD/VT, and other formulas, are interrelat-
endotracheal tube before entry into the breath- ed as dead-space determinants project direct cor-
ing circuit lowered dead-space gas dilution and relations to the status and effectiveness of ventila-
is preferred over mainstream monitoring devices tory efforts to include ventilator management.
because of positioning and increased dead space Capnography is accepted by many as a use-
in the system which competes with the already ful parameter by continuously monitoring the
small tidal volumes of this patient population PETCO2 which can be used as an indicator of the
[19]. PaCO2 even in patients with significant pulmo-
nary disease. However, many studies have indi-
Analysis cated that the PaCO2-PETCO2 gradient will in-
Four independent VD/VT ranges and the cor- crease as the VD/VT ratios increase, therefore the
responding relationship between the PaCO2 and strength of the correlation between the two may
PETCO2 using Pearson’s correlation coefficient decrease as the VD/VT increases [20].
were assessed. The mean PaCO2-PETCO2 differ- There are other opinions based on studies that
ence in each VD/VT range was also calculated us- maintain that there is no way of knowing how
ing multivariable linear regression models to re- large the PaCO2-PETCO2 will be in a given pa-
view the relationships between all dependent and tient and what conditions may exist at any partic-
independent variables to appraise the concurrence ular time to impact the gradient on rapid intervals
between PaCO2 and PETCO2. StataCorp (College [10]. For instance, in a severe ARDS patient, the
Station, Tex., USA) statistics software was used in homodynamic and pulmonary status can create
the evaluation of the data presented [20]. consistent and rapid V/Q shifts which do not al-
low for consistent or constant correlation between

Capnography: Gradient PACO2 and PETCO2 129


PaCO2-PETCO2 monitoring [12]. There are also Conclusions
physiological circumstances where negative val-
ues of the PaCO2-PETCO2 gradient are present Physiological dead-space ventilation is a ma-
which affects its ability in assessing dead-space jor factor in determining the relationship of the
ventilation, for example in the terminal part of the PaCO2-PETCO2 gradient. The difference between
expiration phase, PCO2 can rise rapidly and may the PaCO2-PETCO2 gradient is directly propor-
exceed PaCO2. The slope of phase III (fig. 1) in- tional to the degree of physiological dead space
creases and the likelihood of sampling a PETCO2 and maintains a direct relationship or constant as
greater than the PaCO2 exists. Wide ranges of the VD/VT ratio increases. It remains a useful in-
V/Q mismatching and reduced FRC in patients dicator of PaCO2 within the stable patient as well
after cardiopulmonary bypass surgery for exam- as with those with substantial lung disease with
ple may result in a negative P(a-ET)CO2 value and elevated VD/VT ratios, or under controlled an-
reduced FRC, and increased CO2 production may esthesia, as long as the elevations in the gradient
cause a negative P(a-ET)CO2 value in infants [22, remain predictable or correlate with the increase
23]. in physiological dead space. However, as the clin-
A non-related dead-space issue that will di- ical status of the patient deteriorates, the gradi-
rectly affect the accuracy of the PaCO2-PETCO2 ent become less predictable, especially in that pa-
monitoring and gradient is the level of secretions tient population experiencing extremely elevated
being produced by the patient and the amount of physiological dead space from severe lung dis-
suctioning required. Accumulation of thick secre- ease, hemodynamic instability and/or multisys-
tions in the CO2 sensor is not unusual and will re- tem failure. At this point, the PETCO2 becomes
quire sensor cleaning, removal and in many cases the predictor only when verified by periodic arte-
result in inaccurate PETCO2 if any reading can be rial blood gases which is the denominator and as
taken. Reliance of the PaCO2-PETCO2 gradient such will be less required as PaCO2-PETCO2 gra-
with acute or chronic, intubated or non-intubat- dients are utilized.
ed patients as a definitive diagnostic parameter is
controversial requiring more analysis, especially
in the pulmonary acute patient population.

References
1 Breen PH: Carbon dioxide kinetics dur- 4 Tyburski J, Carlin AM, Harvey EH, et al: 7 Yamanaka MK, Sue DY: Comparison of
ing anesthesia: pathophysiology and End-tidal CO2 and arterial CO2 differ- arterial end-tidal PCO2 difference and
monitoring; in Breen PH (ed): Respira- ences: a useful intraoperative mortality EAD space/tidal volume ration in respi-
tion in Anesthesia: Pathophysiology and marketing trauma surgery. J Trauma ratory failure. Chest 1987;92:832–835.
Clinical Update. Anesthesiology Clinics 2003;55:892–897 8 Belpomme V, Ricard-Hibon A, Devoir C,
of North America. Philadelphia, Saun- 5 Shankar KB, Moseley H, Kumar Y: Nega- et al: Correlation of arterial PCO2-
ders, 1998, vol 16, pp 259–293. tive arterial to end-tidal gradients. Can J PETCO2 in prehospital controlled ventila-
2 Swedlow DB: Capnometry and capnog- Anesth 1991;38:260–261. tion. Am J Emerg Med 2005;23:852–859.
raphy. The anesthesia disaster early 6 Fletcher R, Janson B, Cumming G, Brew 9 Anderson CT, Breen PH: Carbon dioxide
warning system. Semin Anesth 1986;3: J: The concept of dead space with special kinetics and capnography during critical
194–205. reference to the single breath test for care. Crit Care 2000;4:207–215.
3 Krauss B, Hess DR: Capnography for carbon dioxide. Br J Anaesth 1981;53: 10 Lawrence MM: All You Really Need to
procedural sedation and analgesia in the 77–88. Know to Interpret Arterial Blood Gases
emergency department. Ann Emerg – Non-Invasive Blood Gas Interpreta-
Med 2007;50:172–181. tion, ed 2. Philadelphia, Lippincott,
1999.

130 Donnellan
11 Rodriguez-Roisen R, Wagner PD: Clini- 16 Plewa MC, Sikora S, Engoren M, et al: 20 McSwain SD, Hamel DS, Smith PB, Gen-
cal relevance of ventilation-perfusion Evaluation of capnography in non-intu- tile MA, Srinivasan S, Meliones JN,
inequality determined by inert gas elim- bated emergency department, patients Cheifetz IM: End-tidal and arterial car-
ination. Eur Respir J 1998;3:469–482. with respiratory distress. Acad Emerg bon dioxide measurements correlated
12 Dall’ava-Santucci J, Armaganidis A, Bru- Med 1995;2:901–908. across all levels of physiologic dead
net F, et al: Mechanical effects of PEEP 17 Clause D, Liistro G, Thys F, et al: Arterial space. Respir Care 2010;55;350–352.
in adult respiratory distress syndrome. J to end-tidal CO2 (PaCO2-EtCO2) gradi- 21 Benallal H, Busso T: Analysis of end-
Appl Physiol 1990;3:843–848. ent as a monitoring parameter of effi- tidal and arterial PCO2 using a breath-
13 Petty TI: The use, abuse and mystique of cacy during thrombolytic therapy for ing model. Eur J Appl Physiol
positive and emergency pressure. Am massive pulmonary embolism in sponta- 2000;83:402–408.
Rev Respir Dis 1998;138:875–878. neously breathing patients. 23rd Int 22 Russell GB, Graybeal JM, Strout JC: Sta-
14 Coffey RL, Albert RK, Robertson HT: Symp Intensive Care & Emergency Med- bility of arterial to end-tidal carbon
Mechanism of physiological dead-space icine, Brussels, March 2003. Critical dioxide gradients during postoperative
response to PEEP after oleic acid Care 2003(suppl 12):P114. cardiorespiratory support. Can J Anaesth
induced lung injury. J Appl Physiol 1983; 18 Hagerty JJ, Kleinman ME, Zurakowski 1990;37:560–566.
55:1550–1557. D, Lyons AC, Krauss B: Accuracy of a 23 Rich GF, Sconzo JM: Continuous end-
15 Yosefy C, Hay E, Nasri Y, Magen E, Rei- new low-flow sidestream capnography tidal CO2 difference sampling within
sin L: End-tidal carbon dioxide as a pre- technology in newborns: a pilot study. J proximal end tracheal tube estimates
dictor of the arterial PCO2 in the emer- Perinatol 2002;22:219–225. arterial CO2 tension in infants. Can J
gency department setting. Emerg Med J 19 Badgwell JM, McLead ME, Lerman J, Anaesth 1991;38:201–203.
2004;21:557–559. Creighton RE: End-tidal PCO2 measure-
ments sampled at the distal and proxi-
mal ends of the endotrachael tube in
infants and children. Anesth Analg
1987;66:959–664.

Michael E. Donnellan, MBA


Oridion Capnography Inc.
160 Gould Street, Suite 205
Needham, MA 02494 (USA)
Tel. +1 707 422 8538, Fax +1 707 435 9231
E-Mail michael_resp@yahoo.com

Capnography: Gradient PACO2 and PETCO2 131


Section Titleand Equipment in Transport
Technology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 132–135

Problems of Air Travel for Patients with Lung


Disease
Andrew G. Robson
Respiratory Function Service, Western General Hospital, Edinburgh, UK

Abbreviations assessment protocols have been developed. These


COPD Chronic obstructive pulmonary disease
protocols either expose patients to the conditions
FEV1 Forced expiratory volume in one second they are likely to encounter during air travel or
FiO2 Inspired fraction of oxygen use sea level arterial blood gas analysis to estimate
PaO2 Arterial partial pressure of oxygen altitude PO2.
PO2 Partial pressure of oxygen

High Altitude Physiology


Air travel is a rapidly expanding mode of travel
worldwide. By analogy, many more passengers The sigmoid shape of the oxygen dissociation
with chronic respiratory disease will be flying as a curve allows individuals with no respiratory dis-
result of this increase. ease to ascend to moderate altitude without any
At normal cruising altitudes (9,000–12,500 m appreciable hypoxaemia. Beyond this altitude the
for commercial traffic) most aircraft are designed fall in alveolar PO2 is steep and significant hypox-
to maintain a reduced cabin pressure which is the ia can quickly develop. Patients with respiratory
equivalent to an altitude of not greater than 2,438 disease may have a rightward shift in the oxygen
m (8,000 ft) above sea level [1]. At this altitude, dissociation curve due to chronic respiratory aci-
PO2 is ~14.4 kPa and FiO2 is the equivalent of dosis which will result in a decreased affinity of
15.1% at sea level. Because of the sigmoid shape haemoglobin for oxygen, increasing the possibil-
of the oxygen dissociation curve, most passengers ity for the development of desaturation.
can tolerate this reduction in PO2 without experi- The normal response to increasing altitude is
encing any respiratory distress, but patients with an increase in cardiac output and minute ventila-
chronic respiratory disease may develop clinically tion, which will compensate for the reduced PO2.
significant hypoxia. However, passengers with either cardiac or re-
The accurate prediction of symptoms at al- spiratory limitation may experience difficulty in
titude is not currently possible, so most studies increasing either of these parameters sufficiently
have focused on attempts to predict the possibil- to compensate for the increased altitude. Patients
ity of hypoxia at altitude and a number of different with chronic respiratory disease may also have a
blunted ventilatory response to hypoxia and this is for the patient to breathe a hypoxic gas mix-
may lead to alveolar and tissue hypoxia, which is ture (commonly referred to as a hypoxic chal-
of considerable clinical significance. lenge) which will replicate the PO2 experienced in
a pressurised commercial airliner. Ideally the pa-
tient’s FiO2 should be 15.1% as this replicates the
Existing Guidelines PO2 likely to be experienced at 2,438 m (8,000 ft).
Most commercial flights actually have a simulated
Two comprehensive guides giving advice on the cabin altitude lower than this figure (~2,000 m),
management of patients with chronic lung disease but it is prudent to assess a patient for the ‘worst-
have been published [2, 3]. These publications case’ scenario.
aim to provide guidance for physicians who are Hypoxic challenge can be carried out using a
involved in assessing patients who are consider- specially prepared gas mixture either from a gas
ing commercial air travel. Both publications stress cylinder or by utilising a Douglas bag, which acts
that they are not intended to provide inflexible as a reservoir for the hypoxic gas mixture which
rules for air travel but should be used as a basis a patient breathes from using a non-rebreathing
for advising each patient on an individual basis. valve [4]. A simplification of the hypoxic inhala-
There are very few absolute contraindications tion challenge method was described by Vohra
to commercial air travel, but they include infec- and Klocke [5], who used a 40% Venturi-type ox-
tious tuberculosis and unresolved pneumotho- ygen mask driven by 100% nitrogen, which lowers
rax, the latter because air trapped in the pleural the FiO2 to 15.1%. This method has the advantage
space will expand at altitude. Following thoracic that very little specialist equipment is required to
surgery some airlines will accept patients after 2 perform the challenge and this method is being
weeks of recovery but individual assessment is re- used in a number of respiratory function labora-
quired. All patients in these categories will require tories within Europe.
individual assessment. The ideal method to assess a patient’s fitness to
The British Thoracic Society guidelines sug- fly is to take them to a hypobaric chamber, where
gest that anybody with a resting ground level O2 simulated altitude can be reproduced, but these
saturation >95% will be able to fly without the risk chambers are uncommon and the author is not
of developing significant hypoxia. The Society’s aware of any chambers which are used for the
guidelines also recommend that anybody with a routine clinical assessment of patients consider-
resting O2 saturation <92% should fly with sup- ing commercial air travel.
plemental O2. Patients already receiving supple- Cramer et al. [6] have used a modified body pl-
mental O2 (i.e. patients on long-term oxygen ther- ethysmograph as an exposure chamber, a method
apy) should have their usual flow rate increased which has the advantage of eliminating the need
by 2 l/min for the duration of the flight to com- for the patient to breathe through a mask which
pensate for the reduction in PO2. some patients find uncomfortable or inhibiting.
The FiO2 within the exposure chamber needs to
be closely monitored to ensure that it remains
Preflight Assessment of Patients with with the desired range.
Respiratory Disease Considering Air Travel A number of authors have developed pre-
diction equations, which use a patient’s sea-lev-
In order to assess a patient’s risk of hypoxia during el PaO2 and FEV1 to estimate PaO2 at altitude.
air travel, a number of different assessment pro- Although the prediction equations described
tocols have been developed. The easiest method will predict hypoxia in groups of patients, their

Problems of Air Travel for Patients with Lung Disease 133


accuracy in individual patients has been ques- reached cruising altitude which was maintained
tioned. For example, Christensen et al. [7] found throughout the flight.
that despite adequate sea level PaO2, significant Most published data have been concerned with
hypoxia occurred in simulated flight in 33% of investigating the response of patients with COPD.
patients at rest and 66% during light exercise. This is perhaps understandable as COPD is prob-
These and other data support the view that the ably the most common condition amongst airline
most precise way to predict hypoxia in individ- passengers but research has been published in oth-
uals is by hypoxic challenge of the individual er patient groups as well. Oades et al. [9] have as-
patient. sessed flight fitness in children with cystic fibrosis
and found the hypoxic challenge to be a good pre-
dictor of which patients were at risk of significant
Oxygen Supplementation during Air Travel desaturation during flight. Christensen et al. [10]
have studied patients with restrictive lung dis-
Most of the studies described above have not only ease from a variety of different causes in a hypo-
used hypoxic challenge or hypobaric chamber baric chamber and have demonstrated that these
exposure to investigate the development of hy- patients may become hypoxic when exposed to
poxia, but have also explored the use of oxygen 2,438 m of simulated altitude. Further research
supplementation to reverse the hypoxia induced. on the effects of commercial air travel in a range
In most cases the use of supplemental oxygen at of different respiratory diseases is required.
low flow rates (2–4 l/min) has proved effective in In summary, a number of methods to predict
restoring PaO2 to ground level values. It is impor- hypoxia have been developed to assess patient
tant to remember that not all airlines will agree to with chronic respiratory disease who wish to fly.
carry passengers requiring supplemental oxygen Further research is required to answer the harder
and some airlines will not provide supplemental question of how to predict which patients will de-
oxygen at a flow rate >4 l/min. velop symptoms or come to actual harm through
hypoxia during flight. Fortunately, for patients it
is clear that commercial air travel is safe for the
Discussion great majority of passengers and with adequate
planning for in-flight oxygen even patients with
Because very few flights are diverted because more advanced disease can now safely enjoy the
of in-flight respiratory emergencies, we can as- benefits of air travel.
sume that commercial air travel is a safe method
of transport for the vast majority of patients with
respiratory disease. Not all patients require in- Recommendations
depth assessment before flying.
Are long flights more hazardous? Most hypo- • Assess each patient individually – not all
xic challenges used for the clinical assessment of patients will require hypoxic challenge.
a patient’s fitness to fly are of 20–30 min dura- • Where hypoxic challenge is indicated, the
tion. The assumption has always been made that patients FiO2 should ideally be 15.1%.
this is long enough for any physiological changes • Supplemental oxygen can be titrated to keep
to have taken place. Akerø et al. [8] have inves- patient’s PaO2 within the desired range.
tigated hypoxia in group of patients with COPD
during a 6-hour commercial flight who found that
there was an initial fall in PaO2 once the flight had

134 Robson
References
1 Cottrell JJ: Altitude exposures during 5 Vohra KP, Klocke RA: Detection and 8 Akerø A, Christensen CC, Edvardsen A,
aircraft flight. Flying higher. Chest 1988; correction of hypoxaemia associated et al: Hypoxaemia in chronic obstructive
92:81–84. with air travel. Am Rev Respir Dis 1993; pulmonary disease patients during a
2 British Thoracic Society: Managing pas- 148:1215–1219. commercial flight. Eur Respir J 2005;
sengers with respiratory disease plan- 6 Cramer D, Ward S, Geddes D: Assess- 25:725–730.
ning air travel: BTS recommendations. ment of oxygen supplementation during 9 Oades PJ, Buchdahl RM, Bush A: Predic-
Thorax 2002;57:289–304 (2004 update air travel. Thorax 1996;51:202–203. tion of hypoxaemia at high altitude in
available at www.brit-thoracic.org.uk). 7 Christensen CC, Ryg M, Refvem OK, et children with cystic fibrosis. BMJ 1994;
3 Aerospace Medical Association: Medical al: Development of severe hypoxaemia 308:15–18.
Guidelines for Airline Travel, ed 2. Aviat in chronic obstructive pulmonary dis- 10 Christensen CC, Ryg M, Refvem OK, et
Space Environ Med 2003;74:A1–A19. ease patients at 2,438 m (8,000 ft) alti- al: Effect of hypobaric hypoxia on blood
4 Gong H, Tashkin DP, Lee EY, et al: tude. Eur Respir J 2000;15:635–639. gases in patients with restrictive lung
Hypoxia-altitude simulation test: evalua- disease. Eur Respir J 2002;20:300–305.
tion of patients with chronic airway
obstruction. Am Rev Respir Dis 1984;
130:980–986.

Andrew G. Robson
Senior Clinical Scientist
Respiratory Function Service, Western General Hospital
Crewe Road South, Edinburgh EH4 2XU (UK)
Tel. +44 131 537 2575, Fax +44 131 537 2351
E-Mail Andy.Robson@luht.scot.nhs.uk

Problems of Air Travel for Patients with Lung Disease 135


Section TitleTechnology in Abdominal Syndromes
Respiratory
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 136–144

Intra-Abdominal Hypertension and Abdominal


Compartment Syndrome: Measuring Techniques
and the Effects on Lung Mechanics
Bart L. De Keulenaer
Intensive Care Unit, Fremantle Hospital, Fremantle, W.A., Australia

Abbreviations raised IAP [3, 4]. Recently, Cheatham [5] showed


ACS Abdominal compartment syndrome
a significant increased patient survival to hospi-
ALI Acute lung injury tal discharge from 50 to 72% in 478 consecutive
APP Abdominal perfusion pressure patients requiring an open abdomen when a con-
ARDS Acute respiratory distress syndrome tinued revised IAH/ACS management algorithm
CW Chest wall was used.
FRC Functional residual capacity The WSACS (www.wsacs.org) has published
HOB Head of bed angle definitions, guidelines and recommendations
IAH Intra-abdominal hypertension for patients at risk for IAH/ACS. IAH is defined
IAP Intra-abdominal pressure
as a sustained pathologic elevation of IAP of 12
MV Mechanical ventilation
mm Hg, whereas ACS is defined as a sustained
PEEP Positive end-expiratory pressure
PVR Pulmonary vascular resistance IAP >20 mm Hg with or without an APP (APP =
V-P curve Volume-pressure curve MAP-IAP) of <60 mm Hg that is associated with
WSACS World Society of Abdominal Compartment new organ dysfunction or organ failure [6, 7].
Syndrome Keeping this in mind, the application of an IAH/
ACS algorithm (see WSACS website) is recom-
mended which includes a non-surgical and surgi-
This overview will give an insight into the effects cal approach in patients at risk.
raised IAP can have on respiratory mechanics, its
implications and what treatment strategies should
be used when faced with IAH or ACS. IAH/ACS Measuring Techniques
is associated with increased morbidity and mor-
tality [1, 2] but despite this good evidence, clini- The golden standard to measure IAP is still via
cians are still reluctant to accept the importance the bladder where the pressure is measured in
of measuring IAP in patients at risk of developing the supine position, along the mid-axillary line
IAH/ACS. Studies have shown that clinical exam- at the level of the superior iliac crest at end-ex-
ination alone has a poor sensitivity in detecting piration and is expressed in mm Hg [6]. There
20
IAP

Tap on Rapid oscillation test


belly

Respiration

Fig. 1. Rapid oscillation flush test.

are several commercial sets available for intermit- • Zero transducer at the level of the iliac crest
tent IAP measurement like the AbViser (Wolfe- along the mid-axillary line (mark with a
Tory Medical, Salt Lake City, Utah, USA) or the ruler).
Foley manometer technique (Holtech Medical, • Instill 20 ml of sterile saline into the bladder
Charlottenlund, Denmark). Some departments (green diaphragm will automatically inflate to
will have their own in-house set-up. prevent the sterile saline to drain into the urine
It is quite easy to measure the IAP once it is bag).
all set up and ready to go. It is simple and easy to • Wait 30 s and record the IAP in mm Hg at end-
familiarize the nursing staff with this technique. expiration.
However, problems can arise when using a hydro- • Normally after approximately 1 min the
static fluid column including changes with posi- diaphragm (green) will deflate automatically
tioning, over- or underdamping of the system, to allow urine and saline to drain again into
and clinicians should be aware of these pitfalls. the urine bag.
It is therefore recommended for correct measure- • Repeat procedure for every IAP
ment of the IAP to look for respiratory variations measurement.
during measurement and the presence of oscilla-
tions when gently tapping the abdomen. In case Measuring IAP Step-by-Step Foley Manometer
of a damped signal the rapid flush test should be (fig. 4)
instigated (fig. 1). Initial set-up and preparations (use aseptic
technique):
Measuring IAP via AbViser Kit (fig. 2) • Open the Foley manometer pouch and close
• First connect the AbViser kit to the patient’s the tube clamp.
urinary catheter (fig. 3), then place the patient • Place the urine collection device under the
in the supine position and make sure the patient’s bladder and tape the drainage tube to
patient’s abdominal contractions are absent. the bed sheet as shown in figure 3.

Intra-Abdominal Hypertension and Abdominal Compartment Syndrome 137


Saline bag
Monitor Monitor ‘One-step’
early often infuse saline

Double
check valve
Zeroing
Pressure stopcock
transducer
Transduction
Fluid
Ab viser injection
auto valve Drain
Foley Drain Foley
Ballon inflates with saline injection–
Balloon automatically deflates –
fluid drains normally IAH occluding drain and directing fluid
into bladder

Patient’s Urinary Urinary


bladder catheter drain tubing

IAP
displays on
patient
monitor

Early
Data Reduce
recognition
trending risk of ACS
of IAH

Fig. 2. AbViser diagram on how to measure IAP via the bladder technique and the AbViser kit.

• Insert the Foley manometer between the • Open clamp, and read IAP (end-expiration)
catheter and drainage device. when the meniscus has stabilized.
• Prime the Foley manometer with 20 ml of • Close clamp and place the Foley manometer in
sterile saline through its needle-free injection/ its drainage position.
sampling port. Prime only once, i.e. at initial • Slow descent (>20–30 s) of the meniscus during
set-up, or subsequently to remove any air in an IAP determination suggests a blocked or
the manometer tube. kinked Foley catheter.
Remember that a surgical or non-surgical in-
IAP Monitoring tervention based on one absolute value of IAP
• Place the ‘0 mm Hg’ mark of the manometer should be avoided, it is the trend over time that
tube at the symphysis pubis level and elevate is important.
the filter vertically above the patient.

138 De Keulenaer
1 Closed clamp, inject 2 Open clamp
20 ml of saline

40 mm Hg

IAP, mm Hg

3 Symphysis
pubis = 0 mm Hg
Close clamp

Fig. 3. Foley manometer (Holtech)


showing three easy steps to mea-
sure IAP.

Increased pleural P
Lungs

Reduced chest
TP wall compliance

Atelectasis

IAP Diaphragm
Fig. 4. Effects of raised IAP on respi-
ratory function. TP = Intrathoracic
pressure.

Intra-Abdominal Hypertension and Abdominal Compartment Syndrome 139


Table 1. Effect of body positioning on IAP measurements

Number Supine HOB 15° HOB 30° HOB 45° Lateral Reverse
(observations) Trendelenberg

Malbrain [11] 37 (79) 8.8±3.9 NA NA 17.1±6.1 6.6±2.9 13.3±4.8

Chionh [12]1 58 (174) 7 (0.7–13.2) NA 8.5 (2.2–14) 10.3 NA NA


(2.9–16.2)

McBeth [13] 37 (300) 13.4±4.2 NA 18.4±4.8 21.5±5.0 NA NA

Vasquez [14]2,3 45 (675) 10.2 12.4 14.0 16.7 NA 19.3


(8.7–11.8) (10.7– (12.3–15.8) (14.8–18.5) (16.8–21.8)
14.1)

Cheatham [15]4 132 (396) 11.8 13.3 15.4 NA NA NA


(11.4–12.2) (12.8– (14.9–15.9)
13.8)

De Keulenaer 10 (60) 6.6±2.9 NA NA NA 11.2±3.3 NA


[16]

Cobb 20 (180) 1.8±2.0 NA NA 16.7 NA NA


[17]

Data expressed as: 1 Means ± SD, 2 Median ± range, 3 Means with 95% CI, 4 Means ± range.

Raised IAP and Respiratory Mechanics morbidity and mortality. Therefore, patients at
risk of developing IAH or ACS should have their
Does the Abdomen Behave as a Hydraulic System? IAP measured every 4 h. It is common practice to
First we have to establish if the abdomen behaves measure IAP in the supine position via the blad-
as a hydraulic system. der (see above) but several authors have shown
Pressures in the abdomen were recognized to that body position significantly affects IAP (ta-
be atmospheric or positive when Rushmer [8] ble 1). This is an important observation as most
showed that the magnitude of pressure at various ICU patients will be nursed in a semirecumbent
levels in the abdomen were related to the height to lateral position rather than supine causing an
of the hydrostatic column of abdominal contents underestimation of the measured IAP that could
above the point of measurement. In other words, be significant in the critically ill patient. Based
it was recognized that the abdomen behaved as a on the available literature, IAP in the semirecum-
hydraulic system and the pressures within were bent position at HOB 30° and 45° is on average 4
hydrostatic in nature. and 9 mm Hg respectively higher than the stan-
Most patients in the ICU are nursed with a dard bladder pressure measurement in the su-
HOB elevation of 30–45° to reduce the risk of pine position and that in patients with impend-
venous arterial pressure. IAH and ACS, as men- ing ACS or grade 3–4 IAH, this should be taken
tioned before, are associated with increased into account [9].

140 De Keulenaer
Table 2. Pulmonary effects of increased IAP

Properties lung/CW Ventilation values Bloods Lung volumes Others

Diaphragm elevation ↑ Auto-PEEP ↑ Hypercarbia FRC ↓ Alveolar edema ↑

Intrathoracic pressure ↑ Peak airway PaO2 ↓ and All lung volumes Activated lung
pressure ↑ PaO2/FiO2 ↓ (TLC, TV, …) ↓ neutrophils ↑

Pleural pressure ↑ Mean airway A-a gradient ↓ Dynamic Pulmonary


pressure ↑ compliance ↓ inflammatory
infiltration ↑

Upper inflection point Plateau Static respiratory Pulmonary


on V-P curve ↓ pressure ↑ system infection rate ↑
compliance ↓

Lower inflection point PVR ↑ Static CW Metabolic cost and


on V-P curve ↑ compliance ↓↓ work of breathing ↑

Extravascular Dead space


lung water ↑ ventilation ↑

IAP and Respiratory Physiology onto the chest cavity, subsequently increasing the
intrathoracic pressure and pulmonary vasculature
Atelectasis eventually leading to pulmonary hypertension
An increased IAP results in a cephalad displace- (table 2).
ment of the diaphragm leading to compression Clinical implications: Raised IAP leads to hy-
atelectasis in the dependent lung fields. Such at- poxia and hypercarbia and can be prevented by
electasis and subsequently arterial hypoxemia PEEP.
(increase in physiologic shunt) are common find-
ings in mechanically ventilated or postopera- Patient Population
tive patients in the ICU. The incidence of these We know from previous data that the occurrence
complications has been reported to be as high as of increased IAP and IAH is not limited to surgery
70% after abdominal surgery and much lower af- or trauma cases alone (45.6%). The incidence of
ter extra-abdominal non-thoracic procedures. IAH in critically ill medical patients is even higher
In fact, after anesthesia with muscular paralysis, (54.4%). Therefore, it affects the entire ICU popu-
crest-shaped changes of increased densities in lation which makes it quite a challenge in terms
the dependent regions of both lungs on CT have of MV when raised IAP and their effects are not
been found. As a result of the cephalad displace- accounted for. We have published a case report of
ment of the diaphragm, there will be a decrease a patient on non-invasive positive pressure ven-
in functional residual capacity, increased alveolar tilation experiencing a cardiac arrest whilst he
dead space and an increase in physiological shunt was put into the semirecumbent position, which
all leading to hypoxemia and hypercarbia. These instantly increased his IAP and led to ACS [10].
changes can be reduced or prevented when PEEP High IAP will lead to increased intrathoracic pres-
is applied. Increased IAP are also superimposed sures, reduced venous return, increased afterload,

Intra-Abdominal Hypertension and Abdominal Compartment Syndrome 141


increased PVR, reduced cardiac contractility and to high permeability edema. MV can result in
eventually cardiac arrest. We identified the prob- end-inspiratory alveolar overstretching and/or
lem early being severe ACS with an IAP >30 mm repeated alveolar collapse subsequently disturb-
Hg due to massive gastric insufflation causing hy- ing the normal fluid balance across the alveolo-
percarbia and asystole. As soon as a nasogastric capillary membrane. The effects (disturbance of
tube was inserted, the dramatic events were re- the integrity of the endothelium, epithelium and
versed almost instantly. impairment of the surfactant system) of MV are
Clinical implications: Raised IAP can affect similar to those seen in ARDS. There is some evi-
the entire ICU population and early intervention dence that MV may result in translocation of bac-
(surgical or non-surgical) should be instigated. teria from the lungs into the bloodstream and the
release of inflammatory mediators from the lung
High IAP and Increased edema Formation tissue into the systemic circulation. Therefore,
High IAP in oleic acid lung injury in pigs signif- MV may contribute to the development of mul-
icantly increased the amount of edema. In their tiple organ failure. Recent animal studies have
animal study they investigated the impact of an shown that abdominal compression followed by
IAP of 0 and 20 mm Hg on respiration and he- abdominal decompression significantly increased
modynamics [18]. They found that after applying the neutrophils in the lungs. Histopathological
20 mm Hg IAP to healthy lungs the gas content findings showed dense pulmonary inflammato-
significantly decreased but lung tissue mass was ry infiltration including atelectasis and alveolar
unaffected. In oleic acid-injured lungs, not only edema.
was there a further decrease in the gas content, Clinical implications: Increased IAP can pro-
but there was also a significant further increase voke the release of proinflammatory cytokines
in tissue weight. This is probably due to increased leading to multiple organ failure.
edema formation and decreased fluid clearance.
The former, due to increased thoracic central ve- IAP and Decreased CW Compliance
nous and pulmonary artery pressures in injured ARDS is characterized by a reduction in FRC
lungs with increased permeability, the latter due and a decrease in static compliance of the respi-
to increased intrathoracic pressure and its effects ratory system. Given the underlying pulmonary
on the lymphatic flow. injury that is present in patients with ARDS, the
Clinical Implications: In patients with ALI/ decrease in static compliance is thought to reflect
ARDS, raised IAP can promote edema formation. mainly alterations of the mechanical properties of
Management includes improving abdominal wall the lung rather than those of the CW. However, a
compliance, evacuating intraluminal contents, number of studies have reported a decrease in CW
evacuating abdominal fluid collections and cor- compliance in mechanically ventilated patients
recting positive fluid balance when IAP is raised. with ALI. Mutoh et al. [19] demonstrated in an
An adequate PEEP level to counteract raised animal model that abdominal distension marked-
IAP is warranted and subsequently will improve ly altered respiratory mechanics by its effect on the
hypoxia. mechanical properties of the CW. On the basis of
these results, the decrease in CW compliance found
IAP and the Effects in ARDS and ALI? in patients with ARDS was ascribed to abdominal
ARDS has become a well-recognized entity and distension. It also seems that alterations in the me-
results from a number of different initiating in- chanics of the respiratory system are based on the
sults with the final common pathway damage to underlying disease process for ARDS. In patients
the alveolar epithelium and endothelium leading with medical ARDS, the inspiratory V-P curve of

142 De Keulenaer
the respiratory system and lung showed a progres- In the early stages of ARDS, PEEP regional-
sive reduction in elastance with inflating volume ly overstretches the pulmonary units that are al-
because of alveolar recruitment (upward concav- ready open, increasing the elastance. PEEP keeps
ity) [20]. In patients in whom ARDS followed ma- the alveoli open at expiration (preventing them
jor abdominal surgery, abdominal distension with to collapse), thereby decreasing compliance of
increased values of CW elastance was observed the respiratory system. In extrapulmonary ARDS
(upward convexity indicating a progressive in- data have shown that PEEP opens collapsed al-
crease in elastance with inflating volume caused veoli and induces recruitment, whereas in pul-
by alveolar overdistension). When abdominal monary ARDS, PEEP leads more to overstretch
pressure was normalized by surgical re-explora- and increase in elastance (improvement of gas
tion, improvement of the mechanical properties exchange is more likely to be via regional diver-
of the respiratory system, lung, and CW was ob- sion of ventilation or perfusion). Therefore, in ex-
served. These data suggest that the flattening of trapulmonary ARDS, some have suggested best
the V-P curve at high pressures observed in some PEEP equals IAP.
patients with ARDS may be due to increase in CW Clinical implications: Raised IAP decreases
elastance related to abdominal distension. These CW compliance. In ARDS secondary to abdom-
results may also have importance for the optimal inal surgery, reduced CW compliance results in
ventilatory management of critically ill patients flattening of V-P curve at high pressures, whereas
with ARDS with respect to the selection of opti- in medical ARDS due to lung recruitment the op-
mal PEEP and VT levels to minimize ventilator- posite occurs.
induced lung injury.
The key point is that for a given applied pres-
sure, the transpulmonary pressure falls when the Conclusion
pleural pressure rises. The diaphragm is mechani-
cally coupled to the abdominal wall and contents. Raised IAP can have a significant impact on the
If IAP increases, FRC decreases, which will shift respiratory function and hemodynamic parame-
the V-P curve of the respiratory system/CW/lung ters in the critically ill patient and when not ac-
to a lower volume (downward) and rightward. counted for can increase morbidity and mortal-
Abdominal distension causes flattening of the in- ity. Future studies should focus on outcome and
spiratory V-P curve of the respiratory system due mortality trials and the WSACS has successfully
to increased CW elastance and or further atelecta- endorsed many multicenter trials in an attempt
sis. Decompression of the abdomen shows an up- to promote research in this particular area in the
ward displacement along the volume axis of V-P critically ill patient.
curves which reflects recruited volume. Those
findings are important with regard to PEEP.

References
1 Malbrain ML, Chiumello D, Pelosi P, et 2 Malbrain ML, Chiumello D, Pelosi P, et 3 Sugrue M, Bauman A, Jones F, et al:
al: Prevalence of intra-abdominal hyper- al: Incidence and prognosis of intra- Clinical examination is an inaccurate
tension in critically ill patients: a multi- abdominal hypertension in a mixed pop- predictor of intra-abdominal pressure.
centre epidemiological study. Intensive ulation of critically ill patients: a multi- World J Surg 2002;26:1428–1431.
Care Med 2004;30:822–829. ple-center epidemiological study. Crit
Care Med 2005;33:315–322.

Intra-Abdominal Hypertension and Abdominal Compartment Syndrome 143


4 Kirkpatrick AW, Brenneman FD, 9 De Keulenaer BL, De Waele JJ, Powell B, 15 Cheatham ML DWJ, De Keulenaer B,
McLean RF, et al: Is clinical examination et al: What is normal intra-abdominal Widder S, et al: Effect of body position
an accurate indicator of raised intra- pressure and how is it affected by posi- on intra-abdominal pressure measure-
abdominal pressure in critically injured tioning, body mass and positive end- ment: A multicenter analysis. Acta Clin
patients? Can J Surg 2000;43:207–211. expiratory pressure? Intensive Care Med Belg 2007;62:246
5 Cheatham ML: Is the evolving manage- 2009;35:969–976. 16 De Keulenaer BL CG, Maddox I, Powell
ment of intra-abdominal hypertension 10 De Keulenaer BL, De Backer A, Schep- B, et al: Intra-abdominal pressure mea-
and abdominal compartment syndrome ens DR, et al: Abdominal compartment surements in lateral decubitus and
improving survival? Crit Care Med 2010; syndrome related to noninvasive ventila- supine position. Acta Clin Belg Suppl
38:402–407. tion. Intensive Care Med 2003;29:1177– 2007;62:269.
6 Cheatham ML, Malbrain ML, Kirkpat- 1181. 17 Cobb WS, Burns JM, Kercher KW, et al:
rick A, et al: Results from the Interna- 11 Malbrain ML: Different techniques to Normal intra-abdominal pressure in
tional Conference of Experts on Intra- measure intra-abdominal pressure: time healthy adults. J Surg Res 2005;129:231–
Abdominal Hypertension and for a critical re-appraisal. Intensive Care 235.
Abdominal Compartment Syndrome. II. Med 2003;30:357–371. 18 Quintel M, Pelosi P, Caironi P, et al: An
Recommendations. Intensive Care Med 12 Chionh JJ, Wei BP, Martin JA, et al: increase of abdominal pressure increases
2007;33:951–962. Determining normal values for intra- pulmonary edema in oleic acid-induced
7 Malbrain ML, Cheatham ML, Kirkpat- abdominal pressure. ANZ J Surg 2006; lung injury. Am J Respir Crit Care Med
rick A, et al: Results from the Interna- 76:1106–1109. 2004;169:534–541.
tional Conference of Experts on Intra- 13 McBeth PB Zygun D, Widder S, 19 Mutoh T, Lamm WJ, Embree LJ, et al:
Abdominal Hypertension and Cheatham M, et al: Effect of patient Volume infusion produces abdominal
Abdominal Compartment Syndrome. I. positioning on intra-abdominal pressure distension, lung compression, and chest
Definitions. Intensive Care Med 2006;32: monitoring. Am J Surg 2007;193:644– wall stiffening in pigs. J Appl Physiol
1722–1732. 647. 1992;72:575–582.
8 Rushmer R: The nature of intraperito- 14 Vasquez DG, Berg-Copas GM, Wetta- 20 Chiumello D, Carlesso E, Aliverti A, et
neal and intrarectal pressure Am J Phys- Hall R, et al: Influence of semi-recum- al: Effects of volume shift on the pres-
iol 1946:242–249. bent position on intra-abdominal pres- sure-volume curve of the respiratory
sure as measured by bladder pressure. J system in ALI/ARDS patients. Minerva
Surg Res 2007;139:280–285. Anesthesiol 2007;73:109–118.

Bart L. De Keulenaer, MD, FJFICM


Intensive Care Unit, Fremantle Hospital
Alma Street, East Fremantle, WA 6160 (Australia)
Tel. +61 8 9431 3333, Fax +61 8 9431 3009
E-Mail bdekeul@hotmail.com

144 De Keulenaer
Critical Care Problems
Applied Technologies in Pulmonary Diagnosis
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 145–150

Pleural Effusions in Critically III Patients


Vasilios Papaioannou ⭈ Ioannis Pneumatikos
Intensive Care Unit, Alexandroupolis University Hospital, Democritus University of Thrace, Alexandroupolis, Greece

Abbreviations studies. Hemothorax and chylothorax will not be


CL Lung compliance
discussed in this article.
CT Computed tomography
CW Chest wall compliance
CXRs Chest x-rays Pathophysiology
LDH Lactate dehydrogenase
LUS Lung ultrasound The pleura space is normally filled with a very
PEEP Positive end-expiratory pressure small amount of colorless alkaline fluid witch
PE(s) Pleural effusion(s) serves as a coupling system between the chest
SCXR Semirecumbent position chest x-ray
wall and the lung. In cases of both hydrostatic
and high-permeability pulmonary edema, fluid
can enter the pleural cavity from the interstitial
PEs are defined as the excessive accumulation of spaces of the lung via the visceral pleura. The lym-
fluid in the pleural space indicating an imbalance phatics have the capacity to absorb 20 times more
between pleural fluid formation and removal. The fluid than is normally formed [2]. Accordingly,
incidence of PEs in the ICU seems to be common a PE may occur when excessive formation of
and varies with screening methods, from approxi- fluid can overwhelm absorptive mechanisms or
mately 8% for physical examination to more than when there is decreased fluid absorption by the
60% for routine LUS [1]. Patients in the ICU may lymphatics.
develop PEs due to either their primary disease Lung collapse associated with PEs may lead
or as a result of several supportive and therapeu- to hypoxemia due to ventilation-perfusion mis-
tic interventions. In particular, hypotension and match or true shunt. However, different studies
hemodynamic compromise requiring aggressive have failed to demonstrate severe oxygenation im-
hydration may lead to fluid overload and inter- pairment due to PEs, whereas unilateral drainage
stitial edema formation with subsequent pleural of large effusions had mild effects on gas exchange
transudation. The current review focuses on the [3]. Furthermore, even if pleural fluid accumula-
PE pathophysiology, diagnosis and management tion may reduce lung volumes, it has been demon-
in the ICU, based on recent findings from clinical strated that the effects of both fluid accumulation
and removal upon respiratory mechanics depend
on the ratio of lung/CW [4]. The more compliant
the lung, the greater the change in lung function-
al residual capacity; the more compliant the chest
Liver
wall, the greater the thoracic cage adjustment with
smaller effect upon lung volumes. Pleural
Diaphragm
fluid

Etiology

PEs are traditionally classified as either transu-


dates or exudates. A transudative PE develops
Fig. 1. LUS in an ICU patient showing a PE. Its appearance
when the systemic factors influencing the for- is anechoic, implying rather a transudate.
mation or absorption of pleural fluid are altered.
Examples of conditions producing transudative
PE are left ventricular failure, pulmonary embo-
lism and cirrhosis. On the contrary, an exudative
PE develops due to mismatch between local fac-
tors influencing the formation and absorption of or mediastinal displacement are missing until
pleural fluid [5]. Examples of conditions produc- the effusion is massive. Other common signs are
ing exudative PEs are bacterial or viral pneumo- blunted of costophrenic angle and loss of hemidi-
nia, malignancy and pulmonary embolism. Only aphragm silhouette. A normal SCXR does not ex-
three studies have prospectively referred to the clude the presence of a PE. The overall accuracy
frequency and etiology of PEs in the ICU [1, 6, 7]. of a SCXR for detecting a PE with reference to
Table 1 describes the most common causes of PEs decubitus view has been estimated to 0.67–0.95. A
in critically ill patients. more recent study using the LUS as gold standard
estimated the accuracy of SCXR to 82% [8].

Diagnosis Lung Ultrasound


LUS shows better sensitivity and reliability for di-
Chest X-Ray Imaging agnosis of PEs than bedside SCXR, it rules out
Nearly all CXRs taken in the ICU are obtained other etiologies such as atelectasis or consolida-
with patients lying in a semirecumbent posi- tion, takes less time than SCXR and can be repeat-
tion – SCXR. This technique is limited by the ed serially at the bedside. In addition, the skills
fact that the film cassette is placed posterior to required to detect PEs are easy to acquire. A PE is
the thorax, so the x-ray beam originates anteri- described as an anechoic or hypoechoic layer be-
or, at a shorter distance than recommended and tween two pleural layers (fig. 1). PEs can be easily
quite often not tangentially to the diaphragmatic distinguished from spleen and liver by visualiza-
dome, thereby making difficult the correct inter- tion of the so-called ‘sinusoid sign’ which indi-
pretation of the silhouette sign. The most com- cates the centrifugal shifting of the lung towards
mon radiographic finding of a PE in a SCXR is in- the chest wall during inspiration. LUS evalua-
creased homogeneous density over the lower lung tion of PEs in mechanically ventilated patients
field without obliterating normal bronchovascu- is very important because it allows the quanti-
lar markings, whereas air bronchogram, and hilar fication of pleural fluid, supporting the decision

146 Papaioannou ⭈ Pneumatikos


Table 1. Causes of PEs in ICU patients reported in three studies.

Ca uses of PEs Mattison Fartoukh Chih-Yen


et al. [1] et al. [6] et al. [7]
(n = 62) (n = 113) (n = 94)
Congestive heart failure 22 (35.5%) 28 (24.8%) 9 (10%)
Atelectasis 14 (23%) 2 (1.7%) –
Cirrhosis 5 (8%) 6 (5.3%) 2 (2.1%)
Hypoalbuminemia 5 (8%) – 7 (7.4%)
Nephrotic syndrome – 1 (0.8%) –
Parapneumonic effusions 7 (11%) 29 (25.6%) 36 (38.3%)
Pulmonary embolism – 5 (4.4%) –
Empyema 1 (2%) 12 (10.6%) 15 (16%)
Malignancies 2 (3%) 11 (9.7%) 1 (1%)
Tuberculosis – 2 (1.7%) –
Pancreatitis 1 (2%) 2 (1.7%) 2 (2%)
Postsurgical effusions – – 1 (1%)
Collagen vascular disease – – 1 (1%)
Uremic pleurisy 1(2)% – –
Hemothorax – 4 (3.4%) –
Trauma – – –
Sepsis – – 10 (10.6%)
Other 1 (2%) – –
Unknown 3 (5%) 6 (5.3%) 8 (9%)

whether or not thoracocentesis should be per- shaped opacities in the posterior and basal por-
formed and providing visual guidance for pleu- tions of the hemithorax. Moreover, chest CT can
ral fluid evacuation [9]. In the supine position, a aid in differential diagnosis between empyema
maximum interpleural distance at the lung base and lung abscess [1].
in end-expiration (Sep) ≥50 mm is highly predic-
tive of a PE ≥500 ml [10]. Another important Pleural Fluid Examination
contribution of LUS is to provide non-invasive Transudates are generally clear with a slightly yel-
information about the nature of PEs. Transudates low tint. Exudates are usually cloudy with large
are always anechoic whereas a liquid with mo- numbers of cells. If pus is aspirated, an empye-
bile particles or septa is suggestive of exudates or ma is established. Pus is determined by its gross
hemothorax. appearance, which is a thick, viscous and opaque
fluid.
Chest CT Scan Exudative PEs meet at least one of the Light’s
CT scan is the ‘gold standard’ examination for criteria, whereas transudative PEs meet none: (1)
detecting and characterizing PEs. CT scan has pleural fluid protein/serum protein >0.5; (2) pleu-
the advantage over LUS in that it can evaluate ral fluid LDH/serum LDH >0.6; (3) pleural fluid
the pleural surface better, and it is ideal to assess LDH more than two-thirds of the normal upper
the lung parenchyma and tracheobronchial tree. limit for serum. Light’s criteria may label approx-
In simple uncomplicated PEs, it shows crescent- imately 25% of transudates as exudates [5]. In

Pleural Effusions in Critically III Patients 147


PES suspicion on portable CXR

Lung ultrasound

Large PEs, Small PEs,


respiratory failure, >15 mm thick on LUS
infection, hemothorax

Yes No

LUS-guided diagnostic
thoracocentesis
(biochemistry, stain, cultures

Exudate Transudate

Fig. 2. Flowchart for the manage-


Chest tube pH <7.2 pH >7.2 Observation
ment of PE in critically ill patients drainage
based on LUS and pleural fluid bio-
chemistry findings.

these conditions, a serum to pleural fluid albumin Diseases with low pleural fluid pH may also have
gradient >1.2 g/dl indicates a transudative PE. A a low pleural fluid glucose concentration, defined
combination of PE cholesterol (criterion for exu- as <60 mg/dl or a pleural fluid/serum glucose ra-
dates: PE-CHOL >60 mg/dl) and PE LDH con- tio of <0.5 [5].
centrations (criterion for exudates: PE-LDH >280
UI/l) seems to have the same or highest discrimi-
natory potential than Light’s criteria [11]. The pH Management of PEs in the ICU
of normal pleural fluid is approximately 7.62 ow-
ing to active transport of HCO3– into the pleural Pleural fluid characteristics remain the major re-
space. A low pH is seen in inflammatory and infil- liable diagnostic test for guiding management in
trative disorders such as infected parapneumonic critically ill patients. Diagnostic sampling (diag-
effusions, empyema, malignancies, and collagen nostic thoracocentesis) is recommended in all
vascular disease. A parapneumonic pleural fluid cases with PEs associated with a lung disease or
with pH <7.2 is an indication for pleural drain- recent chest trauma or surgery. Figure 2 depicts
age. The glucose concentration in transudates and different steps for the management of PEs in the
in most exudates is similar to that in the serum, ICU setting, based on LUS and pleural fluid anal-
as glucose is of low molecular weight and moves ysis findings. LUS is especially valuable in guiding
from blood to pleural fluid by simple diffusion. drainage of loculated or very small effusions. With

148 Papaioannou ⭈ Pneumatikos


the patient adequately positioned on his back or in and no benefit results from recruitment maneu-
supine position, one must check for an inspiratory ver, lung collapse from PEs is minor and drainage
distance of at least 15 mm with the effusion visible is not warranted.
at the adjacent upper and lower intercostals spac-
es. Optimally, the puncture should be done within
seconds to minutes of the marking. Recommendations
The main indications of therapeutic thoracen-
tesis in critically ill patients include drainage of • PEs are common in ICU patients but they can
empyema or hemothorax, and improvement of easily go unrecognized due to technical and
ventilatory compromise in cases of massive effu- diagnostic limitations of supine portable CXRs.
sions. Small PEs do not usually require therapeu- They are usually small, uncomplicated and
tic thoracocentesis and typically will resolve with postoperative, and resolve with conservative
conservative management (i.e., aggressive fluid management. However, if infection is con-
removal). However, in patients with acute respi- sidered, a thoracocentesis should be done
ratory failure, catheter drainage of even a small PE without delay.
can improve oxygenation significantly [12]. • Thoracocentesis in critically ill patients
In a recent review, Graf [13] proposed a thera- should be performed with the help of LUS
peutic algorithm based on respiratory mechanics. or CT scan guidance to decrease the risk of
In cases of low CW, the same PEs will compress complications.
more the adjacent lung, inducing significant col- • Oxygenation effects of PE drainage may
lapse that has been shown to be resistant to in- vary from lack of response to significant
creasing levels of PEEP. Drainage is likely benefi- amelioration during mechanical ventilation.
cial in these cases. If chest wall is normal related In these patients, the ratio of CW/CL has a
to reduced CL PEEP or any recruitment maneuver major role on lung volume and gas exchange
can reduce the impact of PEs on lung volume and response to pleural fluid drainage.
improve oxygenation. Conversely, if CW is normal

References
1 Mattison LE, Coppage L, Alderman DF, 4 Brown NE, Zamel N, Aberman A: 8 Emamian SA, Kaasbol MA, Olsen JF, et
et al: Pleural effusions in the medical Changes in pulmonary mechanics and al: Accuracy of the diagnosis of pleural
ICU: prevalence, causes, and clinical gas exchange following thoracocentesis. effusion on supine chest x-ray. Eur
implications. Chest 1997;111:1018– Chest 1978;74:540–542. Radiol 1997;7:57–60.
1023. 5 Light RW, MacGregor MI, Luschsinger 9 Eibenberger KL, Dock WI, Ammann
2 Miserocchi G: Physiology and PC, et al: Pleural effusions: the diag- ME, et al: Quantification of pleural effu-
pathophysiology of pleural fluid turn- nostic separation of transudates and sions: sonography versus radiography.
over. Eur Respir J 1997;10:219–225. exudates. Ann Intern Med 1972;62: Radiology 1994;191:681–684.
3 Nishida O, Arellano R, Cheng DC, et al: 57–63. 10 Roch A, Bojan M, Michelet P, et al: Use-
Gas exchange and hemodynamics in 6 Fartoukh M, Azoulay E, Galliot R, et al: fulness of ultrasonography in predicting
experimental pleural effusion. Crit Care Clinically documented pleural effusions pleural effusions >500 ml in patients
Med 1999;27:583–587. in medical ICU patients: how useful is receiving mechanical ventilation. Chest
routine thoracentesis? Chest 2005;127:224–232.
2002;121:178–184.
7 Tu CY, Hsu WH, Hsia TC, et al: Pleural
effusions in febrile medical ICU patients.
Chest ultrasound study. Chest
2004;126:1274–1280.

Pleural Effusions in Critically III Patients 149


11 Romero S, Martinez A, Hernandez L, et 12 Miller KS, Sahn SA: Chest tubes. Indica- 13 Graf J: Pleural effusion in the mechani-
al: Light’s criteria revisited: consistency tions, technique, management and com- cally ventilated patient. Curr Opin Crit
and comparison with new proposed plications. Chest 1987;91:258–264. Care 2009;15:10–17.
alternative criteria for separating pleural
transudates from exudates. Respiration
2000;67:18–23.

Ioannis Pneumatikos, MD
Intensive Care Unit, Alexandroupolis University Hospital
Democritus University of Thrace
GR–68000 Dragana, Alexandroupolis (Greece)
Tel. +30 25 5107 5081, Fax +30 25 5103 0423, E-Mail ipnevmat@med.duth.gr

150 Papaioannou ⭈ Pneumatikos


Critical Care Problems
Infections – Ventilator-Associated Pneumonia
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 151–155

Gravitational Force and Respiratory Colonization in


Mechanical Ventilation
Hany Alya ⭈ Lorenzo Berrab ⭈ Theodor Kolobowc
aDivision of Newborn Services, The George Washington University and Children’s National Medical Center, Washington, D.C.;
bMassachusetts General Hospital, Boston, Mass., and cPulmonary and Critical Care Medicine Branch, Heart Lung and Blood Institute,
National Institute of Health, Bethesda, Md., USA

Abbreviations ETT cuff always form, allowing passage of bacte-


ria-colonized secretions from the oropharynx or
CASS Continuous aspiration of subglottic secretions
ETT Endotracheal tube by gastric reflux from the stomach, into the lower
VAP Ventilation-associated pneumonia airways [2]. Leakage of secretions around the ETT
cuff has been observed in more than 80% of in-
tubated patients undergoing surgery [2, 3]. It is
VAP that develops in patients receiving mechani- likely that the semirecumbent position increases
cal ventilation is the most common nosocomial the pooling of oral secretions in the oropharynx.
infection in patients with acute respiratory failure, The hydrostatic pressure of secretions that collect
and is associated with prolonged hospitalization, in the subglottic space (area above the ETT cuff)
increase in healthcare costs and with a 20–70% may facilitate the leakage across the ETT cuff into
mortality [1]. The aerodigestive tract above the the lower airways.
vocal cords is always heavily colonized by several Aspiration of a large inoculum of pathogenic
species of bacteria. However, in the healthy hu- bacteria is the primary cause in the development
man, the lower respiratory tract remains free of of VAP [1–3]. The stomach of critically ill patients
pathogenic bacteria. The major defense mecha- often becomes colonized with Gram-negative
nisms in preventing colonization of the lower air- bacteria. With aspiration of gastric contents into
ways and the development of pneumonia center the oropharynx, these organisms can overgrow
on the cough reflex and mucociliary clearance. All the normal oropharynx flora, and through as-
such mechanisms are greatly impaired in the in- piration across the ETT cuff colonize the lower
tubated and mechanically ventilated patient. The airways and lead to lung infection. Furthermore,
ETT itself allows direct access to the lower respi- during mechanical ventilation, formation of wa-
ratory tract and thwarts the cough reflex. The in- ter droplets on the inner surface of the ETT, due
flated ETT cuff blocks the normal flow of mucous to moisture condensation, is unavoidable. A bio-
clearance which is necessary to remove particles film rapidly develops that covers the inner lumen
and microbes from the bronchial tree. When the of the ETT. The biofilm is a potential reservoir of
ETT cuff inflates within the trachea, folds on the bacteria [4] that will flow into the lungs when the
30°
30°

a Head up

Orogastric feeding tube


Fig. 1. Illustration of the two study
positions: (a) the ‘head-up’ position
with orientation of the trachea/ETT 10°
upward against gravity (to mimic
the semirecumbent position), and
(b) the ‘head-down’ position, with 10°
the orientation of the trachea/ETT b Head down
downward [7].

patient is in the semirecumbent position or out of development of VAP. In this experiment, sheep
the ETT when the patient is in the Trendelenburg- were randomized either to the ‘head-up’ posi-
lateral body position. tion to mimic the semirecumbent position, or the
Despite these concerns, the US Center for ‘head-down’ position, with the orientation of the
Disease Control recommends positioning the pa- trachea/ETT respectively above, or below the hor-
tient in semirecumbent position with the head ele- izontal (fig. 1). All sheep in the ‘head-down’ po-
vated 30–45° to decrease aspiration of gastric con- sition were placed in a rotational system that al-
tent and colonization of the upper aerodigestive lowed turning sheep from one side to the other in
tract by pathogens from the stomach [5, 6]. The a timed manner. Half of the sheep with the head
level of evidence of this recommendation to pre- down received enteral feeding and half did not.
vent VAP is supposedly category II: ‘recommenda- Animals were ventilated for 72 h and then sac-
tion suggested for implementation and supported rificed. No antibiotics were administered at any
by suggestive clinical or epidemiologic studies or time. All sheep in the ‘head-up’ position had a
by strong theoretical rationale.’ However, no ani- significant decrease in PaO2/FiO2 and heavy bac-
mal or human studies were carried out to under- terial colonization of the lungs. All sheep in the
stand role pathogenic mechanisms of VAP related ‘head-down’ rotating position retained excellent
to gravity until recently. lung function with normal PaO2/FiO2 and no evi-
dence of VAP or bacterial lung colonization.
In a subsequent study, Berra et al. [8] exam-
Animal Studies ined the effects of body position when the ETT
is coupled with a side channel just above the cuff
The study conducted by Panigada et al. [7] was for CASS on the development of VAP. Twenty-
the first VAP animal model to provide a com- two sheep were randomized into three groups.
prehensive assessment of the effect of gravity on Eight sheep were kept prone, with the head

152 Aly ⭈ Berra ⭈ Kolobow


a b

Fig. 2. Illustrations of the two study positions: (a) supine position: the infant is maintained on his/her back while the
ETT is held upright in the vertical position and the bed is kept horizontal, and (b) lateral position: the infant is main-
tained on his or her side, with the ETT resting on the bed at the same level as the trachea [10].

elevated 30° above the horizontal plane (control speed of 2.1 ± 1.1 mm/min. Mucus flow, constant-
group); 14 sheep were intubated with an ETT for ly moving within the trachea towards the glottis,
CASS; 7 of these 14 sheep were kept prone with was mechanically obstructed by inflated ETT cuff
the head elevated (CASS-up group), and 7 sheep and accumulated at the cuff of the ETT, and was
with the trachea horizontal (CASS-down group). either spontaneously drained through the ETT lu-
The lower respiratory tract in all sheep kept with men, or was cleared during routine tracheal suc-
the head elevated 30° above horizontal was highly tion. In sheep in the semirecumbent position, tra-
colonized in both of the control and the CASS-up cheal mucus flow was first towards the glottis in
groups. However, in the CASS-down group, the the non-dependent (dorsal) part of the trachea,
lower respiratory tract was not colonized in 6 of then mucus accumulated at the inflated ETT cuff
7 sheep. One sheep showed low levels of bacte- and gravitated to the dependent (ventral) part of
rial growth. the trachea, and ultimately revered direction and,
In yet another study, Bassi et al. [9] investi- by the force of gravity, moved towards the lungs.
gated the effects of gravitational force on trache- Six out of 8 sheep in the semirecumbent position
al mucus transport, which is a primary defense developed pneumonia, while no pneumonia was
mechanisms to prevent lung bacterial coloni- acquired by any of the sheep positioned with the
zation. Sixteen intubated sheep were again ran- trachea oriented below horizontal. Similar results
domized to be positioned with the orientation of were reproduced in pigs ventilated for 168 h (fig.
the trachea above (30–45° from horizontal), or 1) to overcome concerns with sheep (ruminants)
below (5°) horizontal (Trendelenburg position). that may not be a clinically relevant model of in-
Tracheal mucus velocity was measured via radio- fection because of their high colonization of the
graphic tracking of tantalum disks. Tracheal mu- gastrointestinal tract and abundant production of
cus flow in sheep in the Trendelenburg position oropharyngeal secretions [oral commun., unpubl.
was always towards the ETT cuff with an average results].

Gravitational Force and Respiratory Colonization in Mechanical Ventilation 153


Table 1. Number (%) of infants and types of microorganisms recovered in tracheal aspirates at days 2 and 5 of
mechanical ventilation (χ2 value = 19.8) [10]

Supine Lateral p value


n = 30 n = 30

Microbiological results on day 2 20 (67) 14 (47) 0.12


Gram-negative rods 13 (43) 11(37)
Klebsiella 9 (30) 8 (27)
Pseudomonus 3 (10) 2 (7)
Enterobacter 1 (3) 1 (3)
Gram-positive cocci
Staphylococcus 1(3) 0 (0)
Mixed organisms 6(20) 3 (10)

Microbiological results on day 5 26 (87) 9 (30) <0.01


Gram-negative rods 18 (60) 6 (20)
Klebsiella 10 (33) 4 (13)
Pseudomonus 6 (20) 2 (7)
Enterobacter 2 (7) 0 (0)
Gram-positive cocci 0 (0) 2 (7)
Staphylococcus 0 (0) 1 (3)
Streptococcus 0 (0) 1 (3)
Candida 2 (7) 0 (0)
Mixed 6 (20) 1 (3)

Clinical Studies circuit resting horizontally on the bed as opposed


to suspended in the air for supine patients.
The first clinical trial on the relationship between A pilot study [11] was recently conducted to test
gravity and bacterial colonization in the lower re- the feasibility of maintaining ventilated patients in
spiratory system was published in 2008. In that the lateral position in an adult ICU. Ten patients
prospective, controlled trial, Aly et al. [10] ran- were put in a lateral position and a similar number
domly assigned 60 intubated infants to either su- of patients remained in the semirecumbent posi-
pine position (n = 30), or to lateral position (n = tion. They examined the incidence of aspiration by
30), to keep the orientation of the neck/trachea at, detecting pepsin in the tracheal aspirate. The inci-
or below horizontal (fig. 2). After 5 days of me- dence of aspiration did not differ much between
chanical ventilation, tracheal cultures were pos- groups; 3 patients in the lateral position and 5 pa-
itive in 26 infants (87%) in the supine position tients in the semirecumbent position had aspira-
group, and in 9 infants (30%) in the lateral group tion. Of note, patients in the lateral position had
(p < 0.05). Gram-negative bacteria were the main a better progression of arterial oxygenation and a
bacteria isolated from the trachea (table 1). In ad- significantly shorter duration of mechanical ven-
dition to decreased colonization, the lateral posi- tilation compared to the semirecumbent position
tion group had a significant decrease in the in- Therefore, this pilot study showed that lateral-hor-
cidence of self-extubation. It was concluded that izontal orientation of the body and of the ETT is
when patients were in the lateral position it was feasible and safe in intubated adult patients.
more stable to keep the ETT and the breathing

154 Aly ⭈ Berra ⭈ Kolobow


Conclusion advantageous. However, large clinical trials are
still needed to test the long-term clinical outcomes
From this review, we conclude that gravity is a key related to positioning of ventilated patients. Until
component in bacterial colonization of the low- then it is important to understand that supine
er respiratory tract. Keeping ventilated patients and/or semirecumbent positioning is not optimal
in the lateral position is feasible and possibly and such practice is unfounded.

References
1 Chastre J, Fagon JY: Ventilator-associ- 6 Drakulovic MB, Torres A, Bauer TT, 9 Bassi GL, Zanella A, Cressoni M,
ated pneumonia. Am J Respir Crit Care Nicolas JM, Nogue S, Ferrer M: Supine Stylianou M, Kolobow T: Following tra-
Med 2002;165:867–903. body position as a risk factor for nosoco- cheal intubation, mucus flow is reversed
2 Young PJ, Pakeerathan S, Blunt MC, Sub- mial pneumonia in mechanically venti- in the semirecumbent position: possible
ramanya S: A low-volume, low-pressure lated patients: a randomised trial. Lancet role in the pathogenesis of ventilator-
tracheal tube cuff reduces pulmonary 1999;354:1851–1858. associated pneumonia. Crit Care Med.
aspiration. Crit Care Med 2006;34:632– 7 Panigada M, Berra L, Kolobow T: Bacte- 2008;36:518–525.
639. rial colonization of the respiratory tract 10 Aly H, Badawy M, El-Kholy A, Nabil R,
3 Rello J, Sonora R, Jubert P, Artigas A, under artificial ventilation: trachea and Mohamed A: Randomized, controlled
Rue M, Valles J: Pneumonia in intubated tracheal tube orientation. Crit Care Med trial on tracheal colonization of venti-
patients: role of respiratory airway care. 2003;31:2715. lated infants: can gravity prevent ventila-
Am J Respir Crit Care Med 8 Berra L, De Marchi L, Panigada M, Yu tor-associated pneumonia. Pediatrics
1996;154:111–115. ZX, Baccarelli A, Kolobow T: Evaluation 2008;122:770–774.
4 Inglis TJ, Millar MR, Jones JG, Robinson of continuous aspiration of subglottic 11 Mauri T, Berra L, Kumwilaisak K, Pivi S,
DA: Tracheal tube biofilm as a source of secretion in an in vivo study. Crit Care Ufberg J, Kueppers F, Pesenti A, Bigatello
bacterial colonization of the lung. J Clin Med 2004;32:2071–2078. L: Lateral-horizontal patient position
Microbiol. 1989;27:2014–2018. and horizontal orientation of the endo-
5 Recommendations of the CDC and the tracheal tube to prevent aspiration in
Healthcare Infection Control Practices adult surgical intensive care unit
Advisory Committee: Guidelines for pre- patients: a feasibility study Respir Care
venting health-care-associated pneumo- 2010;55:294–302.
nia, 2003. Respir Care 2004;49:926–939.

Hany Aly, MD
Division of Newborn Services, The George Washington University
900 23rd Street, NW, Suite G-2092, Room G-132
Washington, DC 20037 (USA)
Tel. +1 202 715 5236, Fax +1 202 715 5354, E-mail haly@mfa.gwu.edu

Gravitational Force and Respiratory Colonization in Mechanical Ventilation 155


SectionTitle
Critical
Section Care
Title Problems
Section
InfectionsTitle
– Ventilator-Associated Pneumonia
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 156–162

Use of Gram Stain or Preliminary Bronchoalveolar


Lavage Culture Results to Guide Discontinuation
of Empiric Antibiotics in Ventilator-Associated
Pneumonia
Joseph M. Swansona,b,c ⭈ G. Christopher Wooda
aDepartments of Clinical Pharmacy, bPharmaceutical Sciences, and cPharmacology, College of Pharmacy and Medicine, University of

Tennessee Health Science Center, Memphis, Tenn., USA

Abbreviations characterization of sputum, oxygenation, etc.) to


BAL Bronchoalveolar lavage
invasive diagnostic procedures (BAL, PSB, PTC).
HFCC Hold for colony count Additionally, the lack of a standard diagnostic
NG No growth method has prompted research efforts toward im-
pBAL Preliminary BAL cultures proving the sensitivity and specificity of diagnos-
PSB Protected specimen brushing tic tools for VAP.
PTC Protected telescoping catheter Additional important considerations related
VAP Ventilator-associate pneumonia to therapy of VAP include (1) ensuring appropri-
ate empirical antibiotic therapy and (2) avoiding
overexposure to antibiotics. Appropriate empirical
VAP remains a significant infectious complica- therapy is essential because inappropriate empiri-
tion in the ICU, occurring in up to 27% of me- cal antibiotic therapy is associated with increased
chanically ventilated patients, increasing hospital mortality. Unfortunately, inappropriate empirical
stay by up to 9 days, hospital costs by USD 40,000, antibiotic therapy generally requires broad-spec-
and attributable mortality by 33–50%. The pro- trum antibiotics and final culture results are gen-
found affect of VAP necessitates the use of a rapid erally not available until 72–96 h following culture
and accurate diagnostic method. Unfortunately, obtainment [2, 3]. Since only approximately 40%
the diagnosis of VAP is problematic. The lack of of patients with clinically suspected VAP are ac-
a ‘gold standard’ for diagnosis poses a significant tually diagnosed with VAP, many may receive un-
problem when comparing different approaches needed antibiotics. Increased exposure to antibi-
[1]. As a result, practicing clinicians use meth- otics can increase bacterial resistance, leaving the
ods ranging from clinical parameters alone (chest clinician with a delicate balance to maintain [4].
radiograph, temperature, white blood cell count, A potential way to decrease antibiotic use would
be to rapidly identify patients without VAP so that ‘(e.g. Gram stain G+, culture with G– alone or
empiric antibiotics could be discontinued before G+ and G– together) and did not use the terms
the final culture results are reported. This review ‘complete correlation’ or ‘partial correlation. Not
will focus on the use of preliminary BAL culture all studies evaluated correlation when the Gram
results and Gram stain results to adjust empirical stain was negative, but Croce et al. [6] described
antibiotic therapy. the best correlation when patients did not have
VAP (88%). When a correlation between Gram
stain and organism causing VAP were reported,
Gram Stain and Preliminary Culture Results results varied. Goldberg et al. [14] demonstrated
the best correlation of Gram stain with VAP or-
Gram Stain ganisms at 71%, while others ranged from 46 to
Logic dictates that the presence or absence of bac- 60% [8, 11, 13]. Two studies reported statistical
teria in pulmonary cultures alludes to the diagno- scores for correlation; Raghavendran et al. [15]
sis of VAP. Gram stain can be used to immediately reported a fair correlation with a κ score of 0.314
identify bacteria in pulmonary cultures. Studies and Croce et al. [6] reported φ coefficients rang-
have investigated the ability of the Gram stain to ing from –0.297 to 0.484 depending on the com-
enhance the diagnosis of VAP. parison. Overall, these results were disappointing
In 1994, Marquette et al. [5] were the first to and led most investigators to suggest against the
report the ability of Gram stain to identify VAP use of Gram stain to guide empirical therapy. This
prior to final culture results. Definitions for VAP resulted in research into other methods to predict
or absence thereof were complex, including post- final culture results earlier.
mortem examination, PSB (threshold of 103 cfu/
ml), and chest radiograph ‘rapid cavitation’. This Preliminary Results
report was followed by other studies evaluating Based on published data, use of Gram stain has
Gram stain as a diagnostic tool for VAP. All of been abandoned by many clinicians. Without
the studies used clinical criteria as an indicator Gram stain, many wait for final culture results to
of VAP and confirmed the diagnosis using vari- adjust or discontinue empirical antibiotic ther-
ous procedures including BAL, PSB, and PTC. apy for VAP. Mueller et al. [2] first investigated
Microbiologic thresholds used to confirm the di- the ability of pBAL cultures to predict the final
agnosis of VAP were the same for PSB and PTC cultures. This study was a retrospective analysis
(103), whereas thresholds for BAL ranged from using a pharmacist-maintained microbiologic
103 to 105. The use of Gram stain for diagnosis culture database. Due to the nature of the data-
of VAP produced sensitivities ranging from 67 to base, data were only saved in 24-hour increments.
90%, specificities ranging from 49 to 100%, posi- Thus, preliminary results >24 h following sam-
tive predictive values from 45 to 100%, and neg- ple procurement were evaluated. Bronchoscopic
ative predictive values from 42 to 96% (table 1) BALs were performed in all patients with suspect-
[5–14]. Several studies addressed the important ed VAP using techniques previously described [6,
factor of correlating Gram stain with all pathogens 16, 17]. Preliminary results were categorized as no
identified on final culture results. This correlation growth (NG), insignificant (1–99,999 cfu/ml), or
is described differently based on the study. For ex- significant (≥100,000 cfu/ml). Diagnosis of VAP
ample, Duflo et al. [10] described partial corre- was ruled out if final cultures yielded <100,000
lation as meaning some of the Gram stain mor- cfu/ml or VAP was confirmed if ≥100,000 cfu/
photypes grew on final culture, while Goldberg ml. Preliminary culture results demonstrated an
et al. [14] provided a more detailed description overall sensitivity of 90% and specificity >99% for

Use of Gram Stain or Preliminary BAL Culture Results to Guide 157


Discontinuation of Empiric Antibiotics in VAP
Table 1. Studies evaluating Gram stain for VAP diagnosis

Reference Sample size Gold Sensitivity Specificity Positive Negative


(first author) standard % % predictive predictive
value, % value, %

Marquette 75 PSB, 103 85 94 88 92


1994 [5]

Croce 232 BAL, 105 57 60 47 69


1998 [6]

Prekates 75 BAL, 105 77 87 71 90


1998 [7]

Allaouchiche 118 BAL, PSB 90.2 73.7 64.8 93.3


1999 [8]

Mimoz 186 PSB/ 74, 81 94, 100 91,100 82, 88


2000 [9] PTC, 103

Duflo 116 BAL, 103 76 100 100 75


2001 [10]

Davis 155 BAL, 105 GN 73 GN 49 GN 78 GN 42


2005 [11] GP 87 GP 59 GP 68 GP 83

Veinstein 76 PTC, 103 83 74 79 79


2006 [12]

Kopelman 227 BAL, 104 GN 67 GN 74 GN 69 GN 71


2006 [13] GP 80 GP 66 GP 48 GP 70

Goldberg 309 BAL, 105 90 67 45 96


2008 [14]

GN = Gram-negative; GP = Gram-positive.

the presence of VAP. Preliminary results accurate- could potentially reduce antibiotic exposure in a
ly predicted the presence or absence of VAP in significant number of patients.
96.5% of cases. Interestingly, pBAL results dem- This study prompted changes to the original
onstrating NG had a higher false negative rate Presley Memorial Trauma Center VAP clinical
when compared to those demonstrating insignifi- guideline (fig. 1). The modified guideline includ-
cant growth (7.9 vs. 2.4%, p < 0.001). These results ed the use of pBAL results to determine empirical
were counterintuitive and a clear explanation was antibiotic duration (fig. 2).
not identified. These data suggested the use of in- Subsequently, a prospective validation of the
significant pBAL culture results could potentially retrospective study was conducted with the ad-
reduce empirical antibiotic duration by 2–3 days. ditional goal of evaluating duration of empirical
Considering that only ~40% of patients with clin- antibiotics in cases where VAP is ultimately ruled
ical signs are actually diagnosed with VAP, this out by using pBAL results [3]. Prospective data

158 Swanson ⭈ Wood


Clinical suspicion of VAP

Bronchoscopy with BAL

Empiric antibiotic therapy based on timing of ICU admission

>7 days in ICU:


≤7 days in ICU:
Vancomycin 20 mg/kg IV q 12 h +
Ampicillin/sulbactam 3 g IV q 6 h
Cefepime 2 g IV q 8 h

Final culture results (72–96 h)

<100,000 cfu/ml ≥100,000 cfu/ml

Empiric therapy Definitive antibiotic


Fig. 1. Original Presley Memorial discontinued therapy
Trauma Center VAP clinical guide-
line using final BAL culture results.

collection revealed that the retrospective database Interestingly, the presence of antibiotics did not
had combined two different preliminary culture alter these results. Unfortunately, not all prelimi-
results into the category of NG. In fact, prelimi- nary results were helpful in determining the final
nary BAL culture results could be reported as NG, diagnosis. The preliminary culture result HFCC
HFCC, insignificant growth (1–99,999 cfu/ml), or was indeterminate for the final results, yielding
significant growth (≥100,000 cfu/ml). Using these final results significant for VAP in only 40% of
new classifications of preliminary culture results, cases. Additionally, significant preliminary re-
474 BALs from 176 patients were analyzed. The sults always yielded significant final results, but
positive predictive value of significant prelimi- in 22% of cases significant growth of an addi-
nary culture results was 100%. The negative pre- tional pathogen was identified between the first
dictive values of insignificant and NG prelimi- preliminary result and final results. Use of insig-
nary culture results were 95 and 99% respectively. nificant preliminary culture results resulted in

Use of Gram Stain or Preliminary BAL Culture Results to Guide 159


Discontinuation of Empiric Antibiotics in VAP
Clinical suspicion of VAP

Bronchoscopy with BAL

Empiric antibiotic therapy based on timing of ICU admission

>7 days in ICU:


≤7 days in ICU:
Vancomycin 20 mg/kg IV q 12 h +
Ampicillin/sulbactam 3 g IV q 6 h
Cefepime 2 g IV q 8 h

Preliminary culture results (>24 h)

Insignificant Significant
No growth to date
(1–99,999 cfu/ml) (≥100,000 cfu/ml)

Continue empiric Discontinue Continue empiric


antibiotic therapy antibiotic therapy antibiotic therapy

Final culture results

<100,000 cfu/ml 100,000 cfu/ml

Fig. 2. Updated Presley Memorial


Empiric therapy Definitive
Trauma Center VAP clinical guide-
discontinued antibiotic therapy
line using preliminary BAL culture
results.

significantly shorter durations of empirical anti- Discussion


biotics when compared to the use of final results
[median 1.5 (1.25, 2) vs. 3 (3, 4) days, p < 0.001]. The intent of using Gram stain or pBAL culture
These data resulted in an additional change to results was to provide a more rapid diagnosis of
the Presley Memorial Trauma Center VAP clini- VAP, with the ultimate goal being the guidance of
cal guideline. empirical antibiotic therapy. The ability of Gram
stain to predict VAP is debatable. Several studies

160 Swanson ⭈ Wood


demonstrated acceptable sensitivities, specifici- results were known (1.5 [3] vs. 6 [18] days). Thus,
ties, positive predictive and negative predictive using pBAL to discontinue antibiotics is another
values [5, 9]. However, others reported less fa- tool to potentially reduce the development of re-
vorable results. More importantly, Gram stain sistant bacteria.
was not shown to be effective in identifying final However, pBAL culture results are not infal-
VAP pathogen morphotypes. This renders Gram lible. In fact, preliminary results yielding HFCC
stain less helpful in guiding empirical antibiotic or significant quantities do not provide enough
therapy, including discontinuing or narrowing of information to completely determine final culture
broad-spectrum antibiotics. Current guidelines results. This causes the potential misinterpreta-
recommend using quantitative culture results for tion of final results and thus requires empirical
VAP diagnosis when possible rather than Gram antibiotics to remain broad until final results are
stain. available. Additionally, the validity of preliminary
Preliminary culture results provide more in- culture results has not been studied outside of one
formation than Gram stain, mostly related to the institution. Variations in diagnostic techniques,
quantity of pathogens identified. This additional quantitative thresholds, and laboratory practices
information is provided at the cost of time. The may alter the ability of preliminary culture results
two studies addressing preliminary cultures inves- to accurately predict final results. Overall, the use
tigated results reported ≥24 h post-BAL, whereas of preliminary culture results to predict final re-
Gram stain is available in a few hours. Preliminary sults and alter empirical therapy are promising.
results demonstrated excellent positive and nega-
tive predictive values in both studies. Finally, in-
significant preliminary culture results were used Recommendations
to significantly reduce antibiotic exposure in pa-
tients where VAP is ultimately ruled out. This is • The ability of Gram stain to predict VAP and
an important result because it is the first to dem- organisms causing VAP is questionable.
onstrate a reduction of unnecessary empirical an- • Preliminary culture results demonstrating NG
tibiotic therapy. The prospective study evaluating and insignificant growth predict final culture
preliminary results demonstrated NG results had a results.
>99% negative predictive value. This suggests that • Preliminary culture results demonstrating
combining insignificant and NG preliminary cul- insignificant growth have been used to reduce
ture results could further reduce empirical antibi- antibiotic exposure in patients suspected of
otic exposure in patients that ultimately have VAP having VAP.
ruled out. Indeed, the most important finding of
the prospective study was a significant reduction
in the duration of empiric antibiotic therapy when
pBAL was used to guide empiric therapy. The de-
crease of 1.5 days seen in this study may not seem
immediately impressive, but there will be a large
cumulative reduction in antibiotic use with this
method because of the large number of patients
who receive empiric antibiotics for a VAP work-
up. Also, the use of pBAL to guide discontinua-
tion of antibiotics compares very favorably to an-
other recent study that waited until final culture

Use of Gram Stain or Preliminary BAL Culture Results to Guide 161


Discontinuation of Empiric Antibiotics in VAP
References
1 American Thoracic Society; Infectious 7 Prekates A, Nanas S, Argyropoulou A, et 13 Kopelman TR: Can empiric broad-spec-
Diseases Society of America: Guidelines al: The diagnostic value of Gram stain of trum antibiotics for ventilator-associated
for the management of adults with hos- bronchoalveolar lavage samples in pneumonia be narrowed based on
pital-acquired, ventilator-associated, and patients with suspected ventilator-asso- Gram’s stain results of bronchoalveolar
healthcare-associated pneumonia. Am J ciated pneumonia. Scand J Infect Dis lavage fluid. Am J Surg 2006;192:812–
Respir Crit Care Med 2005;171:388–416. 1998;30:43–47. 816.
2 Mueller EW, Wood GC, Kelley MS, et al: 8 Allaouchiche B, Jaumain H, Chassard D, 14 Goldberg AE, Malhotra AK, Riaz OJ, et
The predictive value of preliminary bac- et al: Gram stain of bronchoalveolar al: Predictive value of bronchoalveolar
terial colony counts from bronchoalveo- lavage fluid in the early diagnosis of ven- lavage fluid Gram’s stain in the diagnosis
lar lavage in critically ill trauma patients. tilator-associated pneumonia. Br J of ventilator-associated pneumonia: a
Am Surg 2003;69:749–755. Anaesth 1999;83:845–849. prospective study. J Trauma 2008;65:
3 Swanson JM, Wood GC, Croce MA, et al: 9 Mimoz O, Karim A, Mazoit JX, et al: 871–876.
Utility of preliminary bronchoalveolar Gram staining of protected pulmonary 15 Raghavendran K, Wang J, Belber C, et al:
lavage results in suspected ventilator- specimens in the early diagnosis of ven- Predictive value of sputum gram stain
associated pneumonia. J Trauma 2008; tilator-associated pneumonia. Br J for the determination of appropriate
65:1271–1277. Anaesth 2000;85:735–739. antibiotic therapy in ventilator-associ-
4 Trouillet JL, Chastre J, Vuagnat A, et al: 10 Duflo F, Allaouchiche B, Debon R, et al: ated pneumonia. J Trauma
Ventilator-associated pneumonia caused An evaluation of the Gram stain in pro- 2007;62:1377–1382.
by potentially drug-resistant bacteria. tected bronchoalveolar lavage fluid for 16 Mueller EW, Croce MA, Boucher BA, et
Am J Respir Crit Care Med 1998;157: the early diagnosis of ventilator-associ- al: Repeat bronchoalveolar lavage to
531–539. ated pneumonia. Anesth Analg 2001;92: guide antibiotic duration for ventilator-
5 Marquette CH, Wallet F, Neviere R, et al: 442–447. associated pneumonia. J Trauma
Diagnostic value of direct examination 11 Davis KA, Eckert MJ, Reed RL 2nd, et al: 2007;63:1329–1337.
of the protected specimen brush in ven- Ventilator-associated pneumonia in 17 Croce MA, Fabian TC, Schurr MJ, et al:
tilator-associated pneumonia. Eur Respir injured patients: do you trust your Using bronchoalveolar lavage to distin-
J 1994;7:105–113. Gram’s stain? J Trauma 2005;58:462– guish nosocomial pneumonia from sys-
6 Croce MA, Fabian TC, Waddle-Smith L, 466. temic inflammatory response syndrome:
et al: Utility of Gram’s stain and efficacy 12 Veinstein A, Brun-Buisson C, Derrode a prospective analysis. J Trauma 1995;39:
of quantitative cultures for posttrau- N, et al: Validation of an algorithm based 1134–1139.
matic pneumonia: a prospective study. on direct examination of specimens in 18 Micek ST, Ward S, Fraser VJ, et al: A ran-
Ann Surg 1998;227:743–751. suspected ventilator-associated pneumo- domized controlled trial of an antibiotic
nia. Intensive Care Med 2006;32:676– discontinuation policy for clinically sus-
683. pected ventilator-associated pneumonia.
Chest 2004;125:1791–1799.

Joseph M. Swanson, PharmD


Department of Clinical Pharmacy, College of Pharmacy and Medicine
University of Tennessee Health Science Center
901 Madison Ave., Suite 308, Memphis, TN 38163 (USA)
Tel. +1 901 448 1418, Fax +1 901 448 1221, E-Mail jswanson@uthsc.edu

162 Swanson ⭈ Wood


Critical Care Problems
Infections – Ventilator-Associated Pneumonia
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 163–167

Patterns of Resolution in Ventilator-Associated


Pneumonia
Pavlos M. Myrianthefs ⭈ Penelope Evagelopoulou ⭈ George J. Baltopoulos
Intensive Care Unit, KAT Hospital, Athens University School of Nursing, Athens, Greece

Abbreviations mm3) and the deterioration of gas exchange (de-


ARDS Acute respiratory distress syndrome
creased PaO2/FiO2 ratio, increased needs in oxy-
cfu Colony-forming units gen or increased need for positive end-expiratory
CPIS Clinical pulmonary infection score pressure).
CRP C-reactive protein The clinical criteria of VAP diagnosis could
MRSA Methicillin-resistant Staphylococcus aureus also be used as patterns of VAP resolution. Beyond
ODIN Organ dysfunctions and/or infection their diagnostic role, they could also contribute for
PCT Procalcitonin the monitoring of VAP evolution and to the evalu-
SOFA Sepsis-related organ failure assessment ation of the efficacy of the appropriate antibiotic
VAP Ventilator-associated pneumonia
therapy and recognition of patients with clinical
WBC White blood cells
deterioration or antibiotic failure. The application
of the optimum duration of antibiotic therapy de-
creases relapse rates and prevents reinfection and
VAP is the most frequent ICU-acquired infection other complications. As literature is focused on
among patients receiving mechanical ventilation. the optimal duration of antibiotic treatment, the
VAP prolongs the length of ICU stay and increas- study of the natural history of the response to ap-
es the costs and the risk of death in critically ill propriate therapy may help to better justify short-
patients. It has been associated with an attribut- term antibiotic therapy for VAP.
able mortality of approximately 30% depending
on the pathogen isolated, especially when initial
antibiotic treatment is inappropriate. Main Topics
The American Thoracic Society [1] suggests a
combination of clinical and microbiological cri- The Infectious Diseases Society of America
teria for VAP diagnosis. These include the emer- guidelines suggest 72 h of therapy as the time to
gence of a new or progressive radiographic infil- record ‘failure to improve’ in patients with com-
trate, the presence of fever >38.3°C, the presence munity-acquired pneumonia. Sensitivity infor-
of increased purulent tracheal aspirates, leuko- mation from microbiological cultures is usually
cytosis or leukopenia (>12,000 or <4,000 cells/ available in most patients with pneumonia after
48 h of therapy; at this timepoint the patient is as demonstrated by repeated protective specimen
usually reassessed and de-escalation is discussed. brushes. The authors found that microbial growth
Resolution of infectious variables after follow- of ≤103 cfu/ml after several days of the initiation
up of 27 patients with VAP was first evaluated in of antimicrobial therapy is accompanied by clini-
1997. These authors reported that resolution oc- cal failure in <7% of the patients and that micro-
curred in a median of 6 days after diagnosis. bial growth of >103 cfu/ml is related with clinical
The patterns that have been studied for VAP failure in 55.8% of the patients.
resolution include the temperature, the WBC Dennesen et al. [3] demonstrated that among
count, the PaO2/FiO2 ratio and the CPIS. CPIS patients treated with initial appropriate antibiotic
includes parameters such as oxygenation, radio- therapy, significant improvements were observed
graphic findings and their evolution, tempera- for all clinical parameters (temperature, PaO2/
ture, WBC count, tracheal aspirates and microbi- FiO2, leukocyte counts) starting from day 1, most
ological culture results with a maximum score of apparently and rapidly for the PaO2/FiO2 ratio
12. Values of 6 or higher are considered diagnos- within the first 6 days. Little further improvement
tic for VAP. Chest radiographs are generally con- in fever, WBC count or the PaO2/FiO2 ratio oc-
sidered to be of limited value for defining clinical curred beyond 7 days of antibiotic treatment. The
improvement in patients with pneumonia, since microbiologic monitoring with bronchoalveolar
comorbidities (chronic obstructive pulmonary lavage was also correlated with VAP resolution,
disease, chronic heart failure) exist in many of as a gradual fall in the mean number of cfu in en-
them. Although radiologic findings do not reflect dotracheal aspirates was observed. Appropriate
VAP severity, deteriorating abnormalities (pro- antimicrobial therapy rapidly eradicates endo-
gressive infiltrates and evolution of abscess) are tracheal colonization with Streptococcus pneumo-
suggestive of progressive or recurrent episodes of niae, Haemophilus influenzae and Staphylococcus
VAP. CRP and PCT have also been evaluated in aureus, but fails to eradicate colonization with
some studies as markers for VAP monitoring. Enterobacteriaceae and Pseudomonas aerugino-
sa, indicating that follow-up of this parameter
is an unreliable parameter for therapy success
Discussion when these pathogens are involved. These find-
ings, which are completely different from those
Hypoxemia and fever are clinical variables that reported by Montravers et al. [2] emphasize that
can be easily followed at the bedside of the pa- endotracheal colonization is not equivalent to in-
tient simply by physical examination to monitor fection of distal airways and that bacterial eradica-
clinical response to antibiotic therapy. Most stud- tion from endotracheal aspirates is a poor marker
ies report that the oxygenation index (PaO2/FiO2 for determination of clinical response of VAP, es-
ratio) is the earliest parameter to be improved. pecially when caused by Gram-negative bacteria.
PaO2/FiO2 ratio improves from the first day of the Only when VAP is caused by H. influenzae or S.
initiation of appropriate antibiotic therapy and its pneumoniae may follow-up of endotracheal aspi-
improvement goes on until 7 days later, so identi- rates be useful.
fying patients with a good prognosis. Luna et al. [4] found that patients who survived
Montravers et al. [2] observed a significant VAP due to different pathogens had a clinical im-
decrease in temperature and increase in PaO2/ provement by days 3–5 reflected by increased
FiO2 ratio within 3 days of antibiotic treatment, PaO2/FiO2 ratio which was the first parameter
which was accompanied by eradication of bacte- achieving ‘normal’ values by day 3 and best cor-
ria from distal airways in the majority of patients, related to outcome. They also reported that PaO2/

164 Myrianthefs ⭈ Evagelopoulou ⭈ Baltopoulos


FiO2 improvement within the first 3 days distin- authors attributed the good correlation between
guished survivors from non-survivors. CPIS and outcome to one component of it, the
Shorr et al. [5] in a large randomized trial con- index of PaO2/FiO2 with its value improving rap-
firmed that the PaO2/FiO2 ratio may be the most idly in patients on adequate therapy. They found
valuable tool in assessing response to therapy in that improvement in the PaO2/FiO2 ratio was best
VAP. Failure of the PaO2/FiO2 ratio to improve correlated to clinical response, whereas changes
by day 3 or the temperature to fall identifies pa- in the radiographic infiltrate, secretions, fever and
tients with clinical failure and at increased risk for leukocytes were not helpful.
poor outcomes. By day 3, failure of improvement Combes et al. [8], using data from a large
should be considered as a sign for revaluation of prospective multicenter randomized trial, the
the patient. Therefore, intensivists should not fo- PNEUMA trial, studied the predictors for infec-
cus on the precise value for the PaO2/FiO2 but tion recurrence and death in VAP patients. They
should track it over time. concluded that persistent fever on day 8 after VAP
Core temperature as a clinical variable of VAP onset was strongly associated with infection recur-
resolution is useful mainly in patients with VAP rence whereas persistently elevated WBC count
and ARDS, although the feverish period is lon- was not. In contrast, leukocyte count increased
ger in these patients. Vidaur et al. [6] observed for patients who ultimately died, whereas tem-
that fever and hypoxemia were the best indicators perature did not discriminate between survivors
of resolution in patients with VAP and absence and non-survivors. WBCs which are persistently
of ARDS within 48 h of therapy. In patients with increased after 2 weeks of therapy may indicate
ARDS, monitoring core temperature is the most infection relapse. Also, failure of temperature im-
useful indicator although fever takes twice as long provement by day 8 or weak improvement in ox-
to resolve, while the oxygenation index should be ygenation after 2 weeks may indicate reinfection
ignored. The evolution of hypoxemia is the main or relapse. In that study [8] it was also patients
differential factor in the resolution of VAP be- without infection recurrence who had a rapid im-
tween patients with and without ARDS. provement of SOFA, ODIN and radiologic scores.
Garrard et al. [7] investigated the clinical re- The type of bacteria responsible for VAP, non-fer-
sponse to antimicrobial therapy in 83 patients menting Gram-negative bacteria and MRSA, af-
with VAP due to different pathogens using CPIS. fected outcomes independently despite appropri-
In this study, CPIS which was evaluated daily, in- ate antibiotic therapy.
creased progressively over the 2 days prior to the In terms of outcome, it was found [8] that pro-
clinical diagnosis of pneumonia (CPIS ≥6) and gressive WBCs increase despite antibiotic admin-
gradually fell after the commencement of anti- istration which is a bad prognostic factor as well
biotic therapy over the next 9 days. The authors as oxygenation decline after 2 weeks of treatment.
concluded that CPIS may be useful in identify- Temperature was not found as an accurate prog-
ing pneumonia and evaluating response to ther- nostic factor. On the other hand, rapid improve-
apy. In another study, Luna et al. [4] confirmed ment in SOFA and ODIN as well as chest x-ray
the same findings in 63 patients with VAP diag- scores are predictors of good outcome. A bad
nosis. They found that serial measurements of prognostic factor is the failure of improvement
the CPIS could be used to identify survivors who of chest x-ray infiltrates which further deteriorate
differed from non-survivors as early as day 3 of within 2 weeks of antibiotic administration.
therapy. In addition, the evolution of CPIS cor- In another recent prospective observational
related with mortality rate making it an impor- study trial by Vidaur et al. [9] it was reported that
tant variable to monitor during VAP therapy. The the rate of VAP resolution depends on the pathogen

Patterns of Resolution in Ventilator-Associated Pneumonia 165


isolated and its susceptibility to treatment. This inflammatory process as indicated by the persis-
study which included 60 VAP episodes showed tently elevated PCT.
that the clinical variables of VAP resolution (fever, The value of daily measurements of PCT and
PaO2/FiO2, WBC count) were improved during CRP and other biological markers as well as solu-
the first 3 days in >70% of patients on appropriate ble triggering receptor expressed on myeloid cells
antibiotic therapy when VAP was due to methicil- as reliable and useful markers in VAP resolution
lin-sensitive S. aureus, H. Influenzae and suscep- must be elucidated in further studies. In conclu-
tible P. aeruginosa. However, the resolution of the sion, monitoring of VAP evolution can be easily
same clinical variables was significantly delayed applied by the same parameters used for the diag-
(8–10 days) in the group of patients with VAP nosis including oxygenation index (PaO2/FiO2),
due to MRSA while receiving appropriate antibi- temperature, WBCs and CPIS. The most useful
otic therapy and due to P. aeruginosa with inap- parameter to assess VAP resolution seems to be
propriate antibiotic therapy. Independently of the the oxygenation improvement which is obvious
disease severity, MRSA VAP was significantly as- from the first days and continuous for at least 7
sociated with poor clinical resolution and a longer days. CPIS seems to be the second most useful
duration of mechanical ventilation. parameter which should be improved by day 3.
Several studies have shown that CRP is useful Failure of oxygenation and CPIS to improve by
in the diagnosis of sepsis in different clinical situ- day 3 are signs for patients’ re-evaluation and
ations. Pόvoa et al. [10] evaluated the CRP levels possibly clinical failure. Temperature may be use-
in the clinical resolution of VAP. In a cohort of 47 ful only in ARDS patients. Persistently increased
VAP patients, CRP levels were monitored daily. WBCs and temperature as well as failure of oxy-
The authors observed that on day 4 of the antibi- genation to improve after 2 weeks may be indica-
otic therapy, the CRP level of survivors was 0.62 tors of relapse or reinfection.
times the initial value, whereas in non-survivors
it was 0.98. By day 4, a CRP >0.6 times the initial
level or a CRP that relapses or has biphasic kinet- Recommendations
ics was a marker of poor outcome and was associ-
ated with the diagnosis of non-resolving VAP. • The optimum duration of antibiotic therapy in
PCT, the precursor molecule of calcitonin, is VAP imposes the monitoring of clinical vari-
another biomarker that could be used as a tool for ables for the definition of VAP resolution. The
VAP prognosis. PCT is a 116-amino-acid peptide best indicator of VAP resolution is the oxy-
that is devoid of known hormonal activity. Serum genation index (PaO2/FiO2) which improves
levels of PCT are very low in healthy individuals. from the first day of antibiotic therapy and
PCT levels rise during bacterial infections. The reaches ‘normal’ values within the first 3 days.
role of PCT kinetics as a prognostic marker dur- • In patients with VAP and ARDS, monitoring
ing VAP has been recently investigated by Luyt core temperature is the most useful indicator
et al. [11]. The authors observed that higher PCT although the feverish period is much longer
values during VAP therapy identified patients at in these patients, while the oxygenation ratio
higher risk for death or clinical failure. Serum must be ignored.
PCT levels decreased during the clinical course • Serial measurements of CPIS can define the
of VAP but were significantly higher from day 1 clinical course of VAP resolution, identifying
to day 7 in patients with unfavorable outcome. those with good outcome as early as day 3 and
Therefore, failure of PaO2/FiO2 to improve by day could possibly be of help to define strategies to
3 may be the clinical manifestation of the same shorten the duration of therapy.

166 Myrianthefs ⭈ Evagelopoulou ⭈ Baltopoulos


• Daily CRP measurements after onset of anti- • Clinical resolution of VAP is associated with
biotic therapy are useful in the identification the implemented pathogen. MRSA VAP
as early as day 4 of VAP patients with poor resolution is slower than in episodes due to
outcome. Retained PCT serum levels on day other pathogens when receiving appropriate
7 of antibiotic therapy are strong predictors of antibiotic therapy. The progression of radio-
unfavorable outcome. Their role in everyday logic infiltrates, the persistence of increased
clinical use must be elucidated. WBCs and a non-improved ratio of PaO2/
FiO2 after 2 weeks of treatment are predictors
of poor outcome.

References
1 American Thoracic Society, Infectious 4 Luna CM, Blanzaco D, Niederman MS, 8 Combes A, Luyt CE, Fagon JY, et al:
Diseases Society of America: Guidelines et al: Resolution of ventilator-associated Early predictors for infection recurrence
for the management of adults with hos- pneumonia: prospective evaluation of and death in patients with ventilator-
pital-acquired, ventilator-associated, and the clinical pulmonary infection score as associated pneumonia. Crit Care Med
healthcare-associated pneumonia. Am J an early clinical predictor of outcome. 2007;35:146–154.
Respir Crit Care Med 2005;171:388–416. Crit Care Med 2003;31:676–682. 9 Vidaur L, Planas K, Sierra R, et al: Venti-
2 Montravers P, Fagon JY, Chastre J, et al: 5 Shorr AF, Cook D, Jiang X, et al, for the lator-associated pneumonia impact of
Follow-up protected specimen brushes Canadian Critical Care Trials Group: organisms on clinical resolution and
to assess treatment in nosocomial pneu- Correlates of clinical failure in ventila- medical resources utilization. Chest
monia. Am Rev Respir Dis 1993;147: tor-associated pneumonia: insights from 2008;133:625–632.
38–44. a large, randomized trial. J Crit Care 10 Pόvoa P, Coelho L, Almeida E, et al:
3 Dennesen PJ, Van der Ven AJ, Kessels 2008;23:64–73. C-reactive protein as a marker of ventila-
AG, et al: Resolution of infectious 6 Vidaur L, Gualis B, Rodriguez A, et al: tor-associated pneumonia resolution: a
parameters after antimicrobial therapy Clinical resolution in patients with sus- pilot study. Eur Respir J 2005;25:804–
in patients with ventilator-associated picion of ventilator associated pneumo- 812.
pneumonia. Am J Respir Crit Care Med nia: A cohort study comparing patients 11 Luyt CE, Guerin V, Combes A, et al: Pro-
2001;163:1371–1375. with and without acute respiratory dis- calcitonin kinetics as a prognostic
tress syndrome. Crit Care Med 2005;33: marker of ventilator-associated pneumo-
1248–1253. nia. Am J Respir Crit Care Med 2005;
7 Garrard CS, A’Court CD: The diagnosis 171:48–53.
of pneumonia in the critically ill. Chest
1995;108(suppl 2):17S–25S.

Pavlos M. Myrianthefs, MD, PhD


Intensive Care Unit, KAT Hospital, Athens University School of Nursing
Sokratous 4A’
GR–14561 Kifissia, Athens (Greece)
Tel. +30 210 623 4434, Fax +30 210 623 1949
E-Mail pmiriant@nurs.uoa.gr

Patterns of Resolution in Ventilator-Associated Pneumonia 167


SectionTitle
Critical
Section Care
Title Problems
Section
InfectionsTitle
– Ventilator-Associated Pneumonia
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 168–171

Viral Infections in the Intensive Care Unit


C.E. Luyt
Service de Réanimation Médicale, Institut de Cardiologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France

Abbreviations HIV-related infections and those occurring in


ARDS Acute respiratory distress syndrome
bone-marrow or solid-organ transplants.
CAP Community-acquired pneumonia
CMV Cytomegalovirus
HSV Herpes simplex virus Analysis
PCR Polymerase chain reaction
RSV Respiratory syncytial virus Community-Acquired Viral Infections
Community-acquired viral infections are com-
mon diseases that can lead to ICU admission.
The incidence of viral infection has been long un- HSV encephalitis is rare, but it should be prompt-
derestimated because of the scarcity of diagnostic ly recognized because of the availability of a cura-
tests. Modern diagnosis methods, such as PCR, tive treatment. Infection of the respiratory tract
that can detect small amounts of viral nucleic is more frequent: either infection of the upper re-
acid, have markedly improved the identification spiratory tract, which can lead to ICU admission
of viral infections. Viral diseases are in fashion, by triggering exacerbation of chronic obstruc-
and the recent A(H1N1) influenza pandemic will tive pulmonary disease or heart failure decom-
not attenuate it. However, it is important to bear pensation [3], or infection of the lower respira-
in mind that viral detection does not necessar- tory tract, corresponding to a true pneumonia [1].
ily mean viral infection: actually, viral detection Respiratory viruses can trigger up to 40% of acute
can correspond to asymptomatic carriage, or vi- exacerbation of chronic obstructive pulmonary
ral replication without organ involvement, and disease requiring ICU [4], and can be involved in
true viral infection. In the ICU, viral disease can cardiopulmonary failure of elderly [3].
be roughly divided in two categories: community- Viruses were found to cause CAP in roughly
acquired viral disease, with respiratory viruses at 10% of cases, with some studies reaching up to
the head [1], and/or nosocomial viral infection, 40% [1]. In those studies, influenza and rhinovi-
represented by infection due to Herpesviridae, rus were the most frequent viruses detected, fol-
namely HSV and CMV [2]. We will not discuss lowed by other respiratory viruses, like parainflu-
viral infections in specific populations, namely enza, adenovirus, RSV, coronaviruses and, more
recently, human metapneumovirus [2]. These vi- 20% had a CMV viremia >1,000 copies/ml [8].
ruses can cause severe pneumonia with ARDS re- These data confirmed other previously published
quiring mechanical ventilation, but the frequency studies that underlined the role of CMV in un-
of this complication is not known. explained fever, pneumonia or ARDS [9–11]. In
ICU patients, CMV can be detected in the blood
Nosocomial Viral Infections after a median of ICU stay of 12 days, the highest
Nosocomial viral infections are due to latent vi- viremia being detected after a median of 26 days
ruses that are reactivated in critically ill individu- of ICU stay. For patients with CMV lung disease,
als. Actually, after the initial phase of the disease the infection occurs after prolonged mechanical
(in which the inflammatory response predomi- ventilation, roughly after a mean of 3 weeks of
nates), the anti-inflammatory response becomes mechanical ventilation [8, 11].
predominant, leading to ‘immunoparalysis’, noso- The role of respiratory viruses as a cause of
comial infections and viral reactivation [5]. The nosocomial pneumonia is probably limited.
viruses responsible for these episodes are mostly Daubin et al. [12], in a prospective study on 139
Herpesviridae. mechanically ventilated patients, showed that
HSV reactivation in the throat occurs in 22– only 2 out of the 39 patients suspected of having
54% of ICU patients [2, 6, 7]. In a recent study on developed VAP had a respiratory sample (tracheal
201 non-immunocompromised patients ventilat- aspirate) positive for respiratory viruses (1 entero-
ed for at least 5 days, HSV reactivation in the throat virus and 1 influenza). Notably, in that study, 12
was diagnosed in 109 patients (54%), asymptom- (31%) patients had a sample positive for HSV and
atically in 56% of them, whereas it was associated 1 for CMV [12]. Another study found that among
with herpetic ulceration of the lip or gingivostom- 185 patients whose BAL fluid was tested for sev-
atitis in 48 (44%) [6]. HSV can be detected in the eral respiratory viruses (influenza A and B, rhi-
lower respiratory tract of 5–64% of ICU patients, novirus, RSV, adenovirus, human metapneumo-
depending on the population and the diagnostic virus), 21 (11%) [6] were positive: 1 BAL sample
method used. In most cases, HSV recovery from was positive for influenza A, 1 for RSV, 5 for rhi-
lower respiratory tract samples of non-immuno- novirus and 4 for adenovirus; none were positive
compromised ventilated patients corresponds to for human metapneumovirus [13].
viral contamination of the lower respiratory tract Recently, Acanthamoeba polyphaga mimivirus
from mouth and/or throat, but for some patients, has been considered a possible cause of pneumo-
a real HSV bronchopneumonitis (corresponding nia, but its responsibility as pathogen responsible
to HSV infection of the lung parenchyma) can de- for real cases of pneumonia has never been cat-
velop. In 201 non-immunocompromised patients egorically demonstrated [14]. Because of conflict-
with prolonged mechanical ventilation, HSV ing data, no definite conclusion can be drawn con-
bronchopneumonitis was diagnosed in 42 (21%) cerning a possible role of A. p. mimivirus in CAP
[6]. Although HSV reactivation in the throat can or nosocomial viral pneumonia at present [2, 15].
occur early in ICU patients [6, 7], HSV broncho-
pneumonitis occurs generally later, after a mean
of 14 days of mechanical ventilation [6]. Discussion
CMV reactivation seems also frequent in ICU
patients. A recent large multicenter study of crit- Community-Acquired Viral Infections
ically ill non-immunocompromised patients Although encephalitis (with HSV encephalitis at
found that up to 33% of them experienced CMV the head) are not common, every patient suspect-
viremia at any level during their ICU stay, and that ed should be promptly treated with acyclovir until

Viral Infections in the ICU 169


the result of cerebrospinal fluid testing for HSV is not meaningful. It is justified to look for HSV re-
obtained. activation and/or infection in patients with sus-
Viral CAP is common, but as discussed above, pected pneumonia, either immunocompromised,
it is important to bear in mind that viral detec- non-immunocompromised with clinical reacti-
tion does not necessarily mean viral infection. vation of HSV (herpes labialis or gingivostoma-
The identification of viral infection is mostly done titis), or those with unexplained acute respiratory
by PCR. However, the reliability of PCR-positive distress syndrome (ARDS) [6]. The diagnosis of
samples could be doubtful. Like other diagnos- HSV reactivation in the throat can be done by vi-
tic tests, there are false positive results, caused by rus isolation in cell culture. For lower respiratory
contamination of PCR reactions with amplifica- tract infection, HSV can be isolated in tracheal
tion products from previous tests or by carryover aspirate, BAL or mini-BAL samples by cell culture
of homologous genomic DNA. Alternatively, false or PCR. Recently, the virus load, performed by
positive results may arise from non-specific bind- real-time PCR, has been highlighted, and might,
ing of primers to irrelevant sequences. On the in the future, become a way to diagnose HVS lung
other side, the relevance of viral detection in diag- involvement [6].
nostic samples may be also doubtful. Viral excre- For CMV reactivation in the blood, the exact
tion does not mean viral infection; some patients timing and frequency of tests remain to be deter-
can have asymptomatic carriage, or a viral excre- mined for non-immunocompromised patients,
tion without organ involvement. Thus, the results whereas a weekly assay is sufficient in immuno-
of a positive viral sample must be confronted with compromised patients [8]. DNA can be detected
the clinical status of the patient. in the blood by PCR, and quantified by real-time
Another issue is the relevance of performing PCR [8]. For CMV pneumonia in non-immuno-
extensive investigations to diagnose viral infec- compromised patients, BAL fluid should be tested
tion, whereas except for influenza, no specific an- for CMV in case of unexplained ARDS or pneu-
tiviral drug is available. It is probably relevant to monia symptoms without a pathogen being iden-
perform rapid tests for influenza in specific con- tified. CMV can be isolated in the BAL fluid by
ditions, during seasonal influenza, or during epi- cell culture or PCR [9–11].
demic or pandemic. Influenza can be treated by However, virus isolation in BAL fluid does not
neuraminidase inhibitors, oseltamivir and zan- necessarily mean viral infection or viral disease.
amivir. Oseltamivir is the first-choice drug, al- For example, HSV recovery in the lower respi-
though its efficacy is probably limited, even when ratory tract might be a local virus excretion or a
given in the first 48 h of symptom onset. It should contamination from the mouth and/or throat. For
be given only in patients with proven influenza, suspected viral pneumonia, cytologic examination
or in special situations (epidemic or pandemic in- of the cells collected during bronchoscopic BAL
fluenza). Vaccine is probably the best way to fight is the cornerstone for HSV bronchopneumonitis
against seasonal influenza, especially in selected and CMV pneumonia diagnosis [2, 6]. Actually,
populations (elderly, care workers, immunocom- Herpesviridae lung infections can be confirmed
promised), by decreasing the number of severe by a histological involvement, i.e. a HSV- or
cases, including viral pneumonia requiring ICU CMV-specific cytopathic effect on cells collected
admission, and by reducing the mortality rate. during bronchoscopic BAL [2, 6]. The cytopathic
effect seen depends on the Herpesviridae consid-
Nosocomial Viral Infections ered: nuclear inclusions are specific of HSV infec-
Although Herpesviridae infections are frequent in tion whereas cytoplasmic inclusions are specific
ICU patients, screening every patient is probably of CMV infection.

170 Luyt
Recommendations compromised patients with clinical reactivation
of HSV (herpes labialis or gingivostomatis),
• In patients suspected of having developed viral patients with no bacteria but clinical signs
CAP, testing for influenza should be during an of pneumonia, and those with unexplained
epidemic or pandemic. ARDS.
• In ICU patients suspected of having developed • CMV reactivation and/or pneumonia should
ventilator-associated pneumonia, testing for be considered also in ICU patients with
HSV reactivation should be performed in unexplained pneumonia, ARDS.
immunocompromised patients, non-immuno-

References
1 Jennings LC, Anderson TP, Beynon KA, 6 Luyt CE, Combes A, Deback C, et al: 12 Daubin C, Parienti JJ, Vincent S, et al:
et al: Incidence and characteristics of Herpes simplex virus lung infection in Epidemiology and clinical outcome of
viral community-acquired pneumonia patients undergoing prolonged mechan- virus-positive respiratory samples in
in adults. Thorax 2008;63:42–48. ical ventilation. Am J Respir Crit Care ventilated patients: a prospective cohort
2 Luyt CE, Combes A, Nieszkowska A, et Med 2007;175:935–942. study. Crit Care 2006;10:R142.
al: Viral infections in the ICU. Curr Opin 7 Bruynseels P, Jorens PG, Demey HE, et 13 Luyt CE, Le Goff J, Deback C, et al:
Crit Care 2008;14:605–608. al: Herpes simplex virus in the respira- Atteintes respiratoires virales nosocomi-
3 Carrat F, Leruez-Ville M, Tonnellier M, tory tract of critical care patients: a pro- ales autres qu’à Herpesviridae
et al: A virologic survey of patients spective study. Lancet 2003;362:1536– (abstract). XXXVI Conference of the
admitted to a critical care unit for acute 1541. Société de Réanimation de Langue Fran-
cardiorespiratory failure. Intensive Care 8 Limaye AP, Kirby KA, Rubenfeld GD, et çaise, Paris, 2008, suppl 58.
Med 2006;32:156–159. al: Cytomegalovirus reactivation in criti- 14 Vincent A, La Scola B, Forel JM, et al:
4 Cameron RJ, de Wit D, Welsh TN, et al: cally ill immunocompetent patients. Clinical significance of a positive serol-
Virus infection in exacerbations of JAMA 2008;300:413–422. ogy for mimivirus in patients presenting
chronic obstructive pulmonary disease 9 Papazian L, Fraisse A, Garbe L, et al: a suspicion of ventilator-associated
requiring ventilation. Intensive Care Cytomegalovirus. An unexpected cause pneumonia. Crit Care Med 2009;37:
Med 2006;32:1022–1029. of ventilator-associated pneumonia. 111–118.
5 Hotchkiss RS, Coopersmith CM, Anesthesiology 1996;84:280–287. 15 Dare RK, Chittaganpitch M, Erdman
McDunn JE, et al: The sepsis seesaw: tilt- 10 Papazian L, Doddoli C, Chetaille B, et al: DD: Screening pneumonia patients for
ing toward immunosuppression. Nat A contributive result of open-lung mimivirus. Emerg Infect Dis 2008;14:
Med 2009;15:496–497. biopsy improves survival in acute respi- 465–467.
ratory distress syndrome patients. Crit
Care Med 2007;35:755–762.
11 Chiche L, Forel JM, Roch A, et al: Active
cytomegalovirus infection is common in
mechanically ventilated medical inten-
sive care unit patients. Crit Care Med
2009;37:1850–1857.

C.E. Luyt, MD, PhD


Service de Réanimation Médicale, Institut de Cardiologie
Groupe Hospitalier Pitié-Salpêtrière
FR–75651 Paris Cedex 13 (France)
Tel. +33 42 16 38 16, Fax +33 42 16 38 17, E-Mail charles-edouard.luyt@psl.aphp.fr

Viral Infections in the ICU 171


SectionTitle
Critical
Section Care
Title Problems
Section
InfectionsTitle
– Ventilator-Associated Pneumonia
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 172–177

Healthcare-Associated Pneumonia among


Hospitalized Patients
Yuichiro Shindo ⭈ Yoshinori Hasegawa
Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan

Abbreviations Analysis of Main HCAP-Related Topics


CAP Community-acquired pneumonia
HAP Hospital-acquired pneumonia Definition of HCAP
HCAP Healthcare-associated pneumonia Details of the definition of HCAP differ among
MDR Multidrug-resistant recent studies [2–6]. The following are current
MRSA Methicillin-resistant Staphylococcus aureus consensus-derived criteria for HCAP: (1) prior
PDR Potentially drug-resistant hospitalization for ≥2 days in the preceding 90
days; (2) residence in a nursing home or extend-
ed care facility; (3) recent receipt of intravenous
HCAP is a relatively new designation [1]. A sub- antibiotic therapy, chemotherapy, or wound care
stantial number of HCAP patients were previ- in the preceding 30 days, or (4) attending a hospi-
ously defined as having CAP. However, recent tal or dialysis clinic [7]. These criteria were devel-
reports suggest that HCAP should be recog- oped mainly in North America. However, there
nized as a distinct entity because of differences are several differences in epidemiological fac-
in epidemiological patterns of HCAP and CAP tors associated with HCAP (e.g., age distribution
patients [2–6]. Patients with HCAP have great- and frequency of occurrence of PDR pathogens)
er risks of infection with PDR or MDR patho- among countries and institutions, probably as a
gens (e.g., Pseudomonas aeruginosa and MRSA). result of differences in healthcare systems. When
These patients also have a higher mortality rate discussing the definition of HCAP, it should be
than patients with CAP. HCAP is a heterogeneous noted that physicians are required to immediate-
disease; its epidemiological findings are different ly identify patients with risks of PDR pathogen
among countries and institutions (fig. 1) [2–5]. infection in order to approach the optimal de-
In this chapter we consider the current issues that cision on the initial appropriate antibiotic treat-
need to be discussed and clinical implications re- ment. Therefore, considering the epidemiological
garding HCAP, based on our results and other differences among countries, minor modifica-
recent reports [2–6]. tions of the criteria of HCAP should be allowed,
Mortality

Risk of multidrug-resistant pathogens

CAP HCAP HAP/VAP

Fig. 1. Position of HCAP in all pneu-


monias. HCAP is a heterogeneous
disease. There are epidemiological Carratalà [2] Shindo [5] Kollef [3]
differences in the position of HCAP (Spain) (Japan) Micek [4]
in each country. VAP = Ventilator- (USA)
associated pneumonia.

keeping in mind their different healthcare sys- S. pneumoniae and H. influenzae tend to occur
tems. Further validation studies are needed. among CAP patients, whereas S. aureus, P. aerug-
inosa, and MRSA occur among HAP patients.
Baseline Characteristics of Patients with HCAP Comparing the frequencies of S. pneumoniae, H.
Common findings of HCAP patients in recent pa- influenzae, S. aureus, P. aeruginosa, and MRSA
pers were that they had the following character- in four studies, we found differences in the po-
istics more often than CAP patients: elderly, se- sition of HCAP among countries or institutions,
vere pneumonia, central nervous system disorder, i.e. whether HCAP was closer to CAP or HAP, as
chronic renal disease, confusion, respiratory fail- shown in figure 1. This is one of the reasons why
ure, aspiration, and previous use of antibiotics [2, HCAP differs among countries and institutions in
3, 5]. Furthermore, we found that the following its epidemiological pattern.
findings were more often detected in patients with
HCAP than those with CAP: high level of blood Clinical Outcomes
urea nitrogen, acidemia, hyponatremia, and ane- Table 2 shows a comparison of clinical outcomes
mia [5]. Therefore, we suggest that patients with among patients with HCAP in five studies [2–6].
HCAP require more active intervention for system- In most of these studies, the proportion of 30-day
ic management and appropriate treatment of their or in-hospital mortality and that of inappropri-
underlying diseases in addition to early initiation ate initial antibiotic treatment were significantly
of appropriate and adequate antibiotic therapy. higher in HCAP patients compared to CAP pa-
tients. Furthermore, we reported that in-hospital
Pathogen Distribution mortality among HCAP patients with and with-
In our study, PDR pathogens (MRSA, Pseudomo- out initial treatment failure was 62.9% (22/35)
nas species, Acinetobacter species, and extend- and 7.5% (8/106), respectively (p < 0.001), and
ed-spectrum β-lactamase-producing Enterobac- that among CAP patients with and without ini-
teriaceae) occurred more frequently among tial treatment failure was 32.5% (13/40) and 2.5%
HCAP patients than CAP patients [5], as shown in (4/163), respectively (p < 0.001) [5]. Our findings
other studies [3, 4]. However, pathogen distribu- suggest that in-hospital mortality among HCAP
tion differed among reports (table 1). In general, patients with initial treatment failure was more

Healthcare-Associated Pneumonia among Hospitalized Patients 173


Table 1. Etiology of culture-positive HCAP in four studies*

Microbes Carratalà Shindo Kollef Micek


et al. [2] et al. [5] et al. [3] et al. [4]
(n = 85†) (n = 77†) (n = 988) (n = 431)
Spain Japan USA USA
single-center single-center multicenter single-center
prospective retrospective retrospective retrospective
Gram-negative pathogens
Pseudomonas species 2.6 10.4 25.3 25.5
Acinetobacter species – 2.1 2.6 –
ESBL‡-producing Gram- 0 1.3 – –
negative bacteria
Klebsiella species 0 13.0 7.6 6.5
Escherichia coli 3.5 6.5 5.2 4.2
Haemophilus influenzae 17.6 5.2 5.8 4.2
Other Gram-negative bacteria – 10.4 13.0 19.0

Gram-positive pathogens
Streptococcus pneumoniae 41.2 24.7 5.5 10.4
Staphylococcus aureus 3.5 18.2 46.7 44.5
MRSA§ 1.2 6.5 26.5 30.6
Streptococci other than S. pneumoniae – 7.1 7.8 –
Other Gram-positive bacteria – 2.8 7.7 –

*
Data are presented as percentages.

Cases without identified pathogens were excluded.

Extended-spectrum β-lactamase.
§
Methicillin-resistant Staphylococcus aureus.

Table 2. Clinical outcomes among HCAP patients in five studies*

Outcomes Carratalà Venditti Shindo Kollef Micek


et al. [2] et al. [6] et al. [5] et al. [3] et al. [4]
(n = 126) (n = 90) (n = 141) (n = 988†) (n = 431†)
Spain Italy Japan USA USA
single-center multicenter single-center multicenter single-center
prospective prospective retrospective retrospective retrospective

30-Day mortality 10.3 – 15.6 – –


In-hospital mortality – 17.8 21.3 19.8 24.6
Inappropriate antibiotic 5.6 18.9 20.8 – 28.3
treatment

* Data are presented as percentages.


† Culture-positive pneumonia.

174 Shindo ⭈ Hasegawa


serious than that among CAP patients and indi- as patients with CAP. However, recent reports re-
cate that physicians should pay careful attention vealed that a substantial number of patients with
to the initial treatment of HCAP. Occurrence of risk of PDR pathogen infection and high mortality,
PDR pathogens was associated with initial treat- similar to those with HAP, were included among
ment failure and inappropriate initial antibiotic patients with traditionally defined CAP. Such pa-
treatment [5]. In particular, HCAP patients with tients are essential to the concept of HCAP. Both
PDR pathogens were 4.2 and 14.0 times as likely patients with newly defined HCAP and CAP pres-
to have initial treatment failure and inappropri- ent from the community. However, the fact that
ate initial antibiotic treatment, respectively, than HCAP was more life-threatening than CAP to pa-
those without PDR pathogens [5]. Therefore, we tients with initial treatment failure suggests the
suggest that physicians should strongly consider importance of identifying HCAP patients during
PDR pathogens while determining the initial em- diagnosis of pneumonia, especially in the emer-
pirical antibiotic treatment of HCAP patients in gency department, to improve their outcome.
order to improve their management. Therefore, physicians need to understand that
HCAP should be identified as a distinct entity.
Risk Factors for PDR Pathogens After identifying HCAP patients, how should
Which population should be targeted for com- we select the initial empirical antibiotic agents?
bination therapy with broad-spectrum antibiot- The initial empirical antibiotic strategy for HCAP
ics? First, in our study, the proportion of occur- is currently under discussion because of differenc-
rence of PDR pathogens among HCAP patients es in the etiology of HCAP in each country or in-
was 22.1% [5]. Therefore, it is not necessary that stitute. The current priorities that need to be dis-
all HCAP patients receive combination therapy cussed are encapsulated in the following questions:
with broad-spectrum antibiotics. Furthermore, (1) What are the acceptable risk factors for infection
the frequency of PDR pathogens was not depen- with PDR or MDR pathogens? (2) Should we strat-
dent on the severity of pneumonia in HCAP pa- ify those risk factors? and (3) Should we consider
tients, as indicated in our study [5]. Thus, identi- the severity of illness in selection of initial empiri-
fying risk factors for PDR pathogens is important cal antibiotic agents? Additional epidemiological
in selecting initial empirical antibiotic agents. In studies are needed to answer the above questions,
our analysis of risk factors for PDR pathogen oc- and interventional studies should be performed to
currence, use of broad-spectrum antibiotics on assess the antibiotic treatment strategy. Until such
more than 2 days within the previous 90 days and studies are performed, physicians should consider
tube feeding were found to be significant, the risk the local risk factors for infection with PDR patho-
ratios being 3.1 and 2.5, respectively [5]. gens, such as use of broad-spectrum antibiotics on
more than 2 days within the previous 90 days and
tube feeding, as shown in our study.
Discussion Figure 2 shows a schematic for identifying pa-
tients with HCAP and the potential implications
As the first step in clinical management of HCAP, for initial antibiotic treatment. This algorithm is
the most important process for physicians is to a proposal and needs validation, particularly with
identify HCAP patients during diagnosis of pneu- regard to selection of initial empirical antibiotic
monia. Pneumonia in hospitalized patients was agents after identification of HCAP patients.
previously divided into two categories: CAP and Recently, some authors have stated that the
HAP. Most patients admitted to hospital with a concept of HCAP is not needed, because it poten-
diagnosis of pneumonia were simply categorized tially leads to overtreatment [8]. In considering

Healthcare-Associated Pneumonia among Hospitalized Patients 175


Pneumonia patients requiring hospitalization

HCAP is present including any of the following:


1) Hospitalization for 2 days or more in the preceding 90 days
2) Residence in a nursing home or extended care facility
3) Receipt of intravenous antibiotic therapy, chemotherapy, or wound
care in the preceding 30 days
4) Long-term dialysis

No Yes
Treat for CAP Assess risk factors* for PDR or MDR pathogens
e.g.:
Potential pathogens in HCAP – Use of broad-spectrum antibiotics on more than 2 days
† Group A within the previous 90 days
S. pneumoniae –Tube feeding
MSSA *Physicians should consider local risk factors for
Antibiotic-sensitive enteric Gram- infection with PDR or MDR pathogens
negative bacteria
No Yes
H. Influenzae
Atypical pathogens Group A † Group B‡
‡ Group B Treat for non-PDR or non- Treat for PDR or MDR pathogens
Pathogens listed in Group A MDR pathogens
PDR pathogens Antipseudomonal ␤-lactam
MRSA ␤-Lactam + macrolide +
Pseudomonas species or Antipseudomonal fluoroquinolone
Acinetobacter species Respiratory fluoroquinolone or aminoglycoside
ESBL-producing Enterobacteriaceae +/–
Stenotrophomas maltophilia Linezolid or vancomycin

Fig. 2. Schema for evaluation of pneumonia patients requiring hospitalization. This proposed algorithm needs to be
validated. The following issues need to be discussed with regard to selection of initial empirical antibiotic agents: (1)
What are the acceptable risk factors for infection with PDR or MDR pathogens? (2) Should we stratify those risk factors?
and (3) Should we consider the severity of illness? Although use of broad-spectrum antibiotics on more than 2 days
within the previous 90 days and tube feeding were found to be significant risk factors for PDR pathogens in our recent
study, other factors such as immune suppression may also be possible risk factors.

categories of pneumonia, the most important key In conclusion, many countries are facing the
perspective is to provide the appropriate initial problem of an aging society, and the number of el-
assessment and empirical treatment for patients derly people in close contact with healthcare ser-
with pneumonia. Therefore, identifying HCAP vices will increase in the near future. Therefore,
patients is one of the most important approaches the number of patients with HCAP is also expect-
to assure appropriate treatment for patients with ed to increase, and it is therefore becoming an im-
pneumonia. In order to provide the appropriate portant challenge for physicians to improve the
treatment or to avoid overtreatment, we consider quality of management for patients with HCAP.
that it is important to answer three questions, as
described above, regarding the selection of initial
empirical antibiotic agents in HCAP patients.

176 Shindo ⭈ Hasegawa


Recommendations • To assess risk factors for infection with
PDR or MDR pathogens in selecting initial
• To recognize that patients with HCAP have empirical antibiotic agents after identifying
greater risks of infection with PDR or MDR HCAP patients.
pathogens and higher mortality than patients
with CAP.
• To identify patients with HCAP during
diagnosis of pneumonia, especially in the
emergency department.

References
1 American Thoracic Society, Infectious 3 Kollef MH, Shorr A, Tabak YP, et al: Epi- 6 Venditti M, Falcone M, Corrao S, et al:
Diseases Society of America: Guidelines demiology and outcomes of healthcare- Outcomes of patients hospitalized with
for the management of adults with hos- associated pneumonia: results from a community-acquired, healthcare-associ-
pital-acquired, ventilator-associated, large US database of culture-positive ated, and hospital-acquired pneumonia.
and healthcare-associated pneumonia. pneumonia. Chest 2005;128:3854–3862. Ann Intern Med 2009;150:19–26.
Am J Respir Crit Care Med 2005;171: 4 Micek ST, Kollef KE, Reichley RM, et al: 7 Kollef MH, Morrow LE, Baughman RP,
388–416. Healthcare-associated pneumonia and et al: Healthcare-associated pneumonia:
2 Carratalà J, Mykietiuk A, Fernandez- community-acquired pneumonia: a sin- a critical appraisal to improve identifica-
Sabe N, et al: Healthcare-associated gle-center experience. Antimicrob tion, management, and outcomes. Pro-
pneumonia requiring hospital admis- Agents Chemother 2007;51:3568–3573. ceedings of the HCAP Summit. Clin
sion: epidemiology, antibiotic therapy, 5 Shindo Y, Sato S, Maruyama E, et al: Infect Dis 2008;46(suppl 4):S296–S334.
and clinical outcomes. Arch Intern Med Healthcare-associated pneumonia 8 Ewig S, Welte T, Chastre J, et al: Rethink-
2007;167:1393–1399. among hospitalized patients in a Japa- ing the concepts of community-acquired
nese community hospital. Chest 2009; and health-care-associated pneumonia.
135:633–640. Lancet Infect Dis 2010;10:279–287.

Yuichiro Shindo, MD
Department of Respiratory Medicine
Nagoya University Graduate School of Medicine
65 Tsurumai-cho, Showa-ku, Nagoya 466-8550 (Japan)
Tel. +81 52 744 2167, Fax +81 52 744 2176, E-Mail yshindo@med.nagoya-u.ac.jp

Healthcare-Associated Pneumonia among Hospitalized Patients 177


SectionTitle
Critical
Section Care
Title Problems
Section
Oncology Title
and Pulmonary Complications
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 178–184

Critical Care Outcome of Lung Cancer Patients


Abdulgadir K. Adama ⭈ Ayman O. Soubanib
aDivision of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, and bSection of Pulmonary and

Critical Care Medicine, Karmanos Cancer Center, Wayne State University School of Medicine, Detroit, Mich., USA

Lung cancer is the third most common malig- patients, and discusses the recent data on the out-
nancy, but remains the leading cause of cancer come and predictors of outcome of this care. The
mortality in both men and women in the USA discussion is not directed towards patients who
and throughout the world. An estimated 159,390 are admitted to the ICU postoperatively after re-
deaths, accounting for about 28% of all cancer section of lung cancer rather to those patients
deaths, are expected to occur in 2009 in the USA. who develop critical illness associated with their
Since 1987, more women have died each year from underlying malignancy.
lung cancer than from breast cancer. Death rates
among men decreased by 2.0% per year from 1994
to 2005. Female lung cancer death rates have been Indications for Medical ICU Admission
stable since 2003 after continuously increasing for
several decades. The 1-year relative survival for In general, lung cancer patients are admitted to
lung cancer has increased from 35% in 1975–1979 the medical ICU for diagnoses that are similar to
to 41% in 2001–2004, largely due to improvement other patient populations. These include acute
in surgical techniques and combined therapies. respiratory failure, sepsis, cardiovascular prob-
However, the 5-year survival rate for all stages lems, neurologic impairments, renal or metabol-
combined is only 15% [1–5]. Patients with lung ic abnormalities, or bleeding disorders. In a study
cancer, regardless of type or stage, commonly re- of critically ill lung cancer patients by Adam and
quire ICU care for a variety of acute illnesses re- Soubani [8], the main causes for admission to the
lated to the underlying malignancy, treatment, or medical ICU were acute respiratory failure (49%),
co-morbid illnesses. Lung cancer is the third most cardiovascular problems (25%), sepsis (8%), and
common cancer in critically ill patients, and ac- neurological impairment (5%). The indications
counts for 16% of all cancer-related admissions to for admission to the medical ICU for lung cancer
the ICU [6, 7]. There is an ongoing controversy patients may be classified to cancer-related com-
about the value of ICU care to this patient popu- plications, treatment-related complications, and
lation given the poor overall prognosis associated co-morbid illnesses. These indications are sum-
with lung cancer. This chapter reviews the indi- marized in table 1.
cations for medical ICU care for the lung cancer
Table 1. Main indications for medical ICU admission secondary to metastasis to the central nervous
system leading to altered mental status, seizures,
Cancer-related complications
and spinal cord compression with paraplegia or
Superior vena cava obstruction
Airways obstruction or infiltrations
quadriplegia.
Hemoptysis
Pleural effusion Treatment-Related Complications
Pulmonary embolism Radiation pneumonitis is not an uncommon com-
Electrolyte abnormalities plication, especially when used with concurrent
Neurologic complications chemotherapy. The severity of this complication
Treatment-related complications is variable, however may progress rapidly to acute
Radiation pneumonitis respiratory failure in spite of institution of corti-
Chemotherapy-induced pulmonary toxicity costeroid therapy. Diffuse lung injury consistent
Bronchoscopic or other pulmonary procedure with acute respiratory distress syndrome has been
complications
described in some patients following radiation
Postoperative complications for cancer resection
therapy for lung cancer [9]. Several chemothera-
Infections peutic agents used in the treatment of lung cancer
Directly related to the cancer, e.g. obstructive are associated with pulmonary toxicity and acute
pneumonia
respiratory failure. These agents include gemcit-
Immunosuppression secondary to chemotherapy
abine, gefitinib, etoposide, paclitaxel, docetaxel,
Underlying co-morbid illnesses and bevacizumab. Furthermore, chemotherapy
Cardiovascular diseases may lead to immunosuppression with increased
Pulmonary diseases
risk of severe infections, including respiratory in-
Others
fections. The differentiation between infectious
and treatment-related pulmonary complications
may be challenging.
Also, some patients might develop complica-
Cancer-Related Complications tions following diagnostic or therapeutic surgi-
Lung cancer can cause obstruction of the supe- cal or bronchoscopic procedures that may require
rior vena cava causing superior vena cava syn- admission to the medical ICU. For example, pa-
drome, which may be a life-threatening emer- tients may develop acute respiratory distress syn-
gency. Tumors may cause airways infiltration and drome following pneumonectomy [10]. In ad-
obstruction, resulting in respiratory failure and dition, patients may develop acute respiratory
postobstructive pneumonia. Hemoptysis due to failure or significant bleeding following bron-
bleeding tumors in the airways or vascular inva- choscopy, biopsies, or palliative endobronchial
sion can be life-threatening. Cancer-related or procedures.
malignant pleural effusion can lead to respiratory
decompensation. Pulmonary embolism second- Co-Morbid Illnesses
ary to a hypercoagulable state due to malignan- Lung cancer patients are generally at increased
cy is another cause of severe respiratory distress. risk for a variety of respiratory and cardiovascular
Electrolyte abnormalities such as hypercalcemia, problems that are related to their age and cigarette
hyponatremia and hypokalemic alkalosis, are not smoking. These patients may need medical ICU
uncommon in these patients and may be the rea- care for acute exacerbation of chronic obstructive
sons for admission to the medical ICU. A vari- pulmonary disease, cardiogenic pulmonary ede-
ety of neurological complications might develop ma, acute coronary syndrome or other illnesses

Critical Care Outcome of Lung Cancer Patients 179


Table 2. Mortality of lung cancer patients admitted to the medical ICU and literature review

Reference Patients Mechanical ICU Hospital Long-term


(first author) n ventilation, n mortality % mortality, % mortality, %1

Ewer, 1986 [28] 46 46 85 91 98


Boussat, 2000 [29] 57 52 67 75 NR
Jennens, 2002 [32] 20 9 NR NR 85
Lin, 2003 [30] 81 81 72.8 85 NR
Reichner, 2006 [31] 47 23 43 60 NR
Soares, 2007 [33] 143 100 42 59 67
Adam, 2008 [8] 139 68 22 40 48

NR = Not reported.
1
Long-term survival signifies survival >6 months after medical ICU admission.

such as acute renal failure. Finally, the indication of 42%, and 69% for those receiving mechanical
for medical ICU may be due to a combination of ventilation. More recently, Adam and Soubani
the above conditions. [8] reported the outcome of 139 lung cancer pa-
tients, including 68 mechanically ventilated pa-
tients who were admitted to the medical ICU.
Critical Care Outcome of Lung Cancer Patients The overall ICU mortality was 22%, and 38% for
those who were on mechanical ventilation. The
Studies have shown that the ICU outcome of can- hospital mortality for the whole group was 40%,
cer patients in general is poor [11–27]. There are and 52% were still alive more than 6 months af-
few studies that specifically evaluated lung can- ter medical ICU admission. These data show that
cer patients and they suggested that the medical there is a clear trend towards improved overall
ICU outcome of these patients is also guarded survival of lung cancer patients admitted to the
[8, 28–34] (table 2). Ewer et al. [28] in 1986 re- medical ICU, and while those who are mechan-
ported that the ICU mortality among lung can- ically ventilated had a higher mortality; never-
cer patients requiring mechanical ventilation theless, the survival rate for this subgroup has
was 85%. Boussat et al. [29] in 2000 reported also improved. The other observation is that the
an overall ICU mortality of 66%, and 71% for ICU outcome of lung cancer patients generally
those requiring mechanical ventilation. Lin et al. approaches that of other patient populations ad-
[30] in 2003 reported an ICU mortality of 73% mitted to the medical ICU.
in those patients requiring mechanical ventila- It is difficult to establish the specific reasons
tion. However, recent literature suggests that for the improved ICU outcome of lung cancer
the outcome of lung cancer patients admitted to patients, however factors that have been shown
the medical ICU has been steadily improving. to improve ICU outcome in other patient popu-
Reichner et al. [31] in 2006 reported an overall lations, probably apply to these patients as well.
ICU mortality of 43%, and 74% for those requir- These factors may be related to improved me-
ing mechanical ventilation. Another study by chanical ventilation strategies that minimize fur-
Soares et al. [33] in 2007 reported ICU mortality ther lung injury and increased utilization of non-

180 Adam ⭈ Soubani


Table 3. Predictors of critical care outcome

Predictors Reference(s)

Mechanical ventilation 8, 28, 30, 31


Severity of co-morbidities 33
Number of organ system failures 8, 33
Cancer recurrence or progression 31, 33
High sepsis-related or sequential organ failure assessment score 31
Performance status 29
Hemodynamic instability requiring vasopressors 8

invasive ventilation. A few studies have reported emotional and physical toll on these patients and
that the early use of this strategy has resulted in their families. In addition, such care is very expen-
improved gas exchange, decreased dyspnea, less sive and will consume scarce resources. It would
mechanical ventilation, and lower overall mor- be useful to be able to predict, prior to admission
tality rates [24, 35]. In addition, better manage- to the ICU, whether the patient is going to benefit
ment of sepsis and implementation of patient from this aggressive and expensive therapy.
safety bundles of care (such as the ventilator bun- Several retrospective studies have tried to iden-
dle and sepsis bundle) may play a role in better tify the clinical variables that are associated with
ICU outcome. The improvement in the differ- poor ICU outcome (table 3). In a study by Soares et
ent therapeutic options for lung cancer and the al. [33], the predictors of poor ICU outcome were
increased knowledge and experience that devel- severity of co-morbidities, number of organ sys-
oped in those units that routinely deal with can- tem failures, cancer recurrence or progression, and
cer patients may contribute to improved outcome airway infiltration or obstruction by cancer. Some
of these patients. Furthermore, the multidisci- of the predictors that were reported in the study
plinary approach to the management of these pa- by Reichner et al. [31] were the need for mechani-
tients with the involvement of intensivists, oncol- cal ventilation, advanced lung cancer stage, and a
ogists, and other specialists probably contributes higher sepsis-related or sequential organ failure
to the better management of critically ill lung can- assessment score. The death predictive factors in
cer patients. the study by Boussat et al. [29] were acute pulmo-
nary disease and Karnofsky performance status
<70. In the study by Adam and Soubani [8], sev-
Predictors of Critical Care Outcome of Lung eral predictors correlated with poor ICU outcome.
Cancer Patients These include high admission Acute Physiology
and Chronic Health Evaluation (APACHE) III
Despite the improved ICU and hospital outcome score, the need for mechanical ventilation, the use
of lung cancer patients reported in the recent lit- of vasopressors, positive blood cultures, high se-
erature, not all patients benefit from this aggres- rum lactate, the presence of two or more multi-
sive care. For many of the patients with lung can- organ system failure, and the need for advanced
cer, ICU care is futile and will not prolong their cardiac life support protocol for cardiopulmonary
lives. Such therapy may be associated with a huge arrest. On multi-regression analysis only the use

Critical Care Outcome of Lung Cancer Patients 181


of vasopressors and the presence of two or more not responding to therapy, continue to require me-
organ system failures predicted poor ICU out- chanical ventilation, or develop multiorgan sys-
come, with an odds ratio 8.7 and 40.8 respectively. tem failure, then the patients and/or their family
The stage of lung cancer or the presence of metas- should be approached regarding limiting aggres-
tasis did not correlate with poor medical ICU out- sive therapy and considering palliative care. While
come. This was similar to the findings in the study aggressive therapy is appropriate in patients with
by Boussat et al. [29], but in contrast to the find- lung cancer, it appears that performing advanced
ings of Soares et al. [33] and Reichner et al. [31]. cardiac life support on those with critical illness
Regarding the type of lung cancer the studies have is futile. Few studies have shown that it is unusual
shown variable results. The latest study observed for those patients to survive their hospitalization
that patients with non-small cell lung cancer had [8, 33]. This observation may be conveyed to pa-
a favorable ICU outcome [8], which is contrary to tients and their family members to avoid subject-
the findings of other studies [29, 31]. ing the patients to this measure if their conditions
Age was not found to be a predictor of out- deteriorate.
come in most of these studies, and should not be An important aspect in the care of the lung
a criterion by itself against ICU care. Performance cancer patient should be an early discussion about
status, on the other hand, appears to be more a sig- the extent of treatment the patient wishes and
nificant factor. A recent study by Christodoulou et end-of-life issues. It has been our experience, and
al. [36], of critically ill cancer patients, including documented in the literature, that the majority of
lung cancer, reported that only performance sta- the lung cancer patients admitted to the medi-
tus (3–4) was a predictor of short-term negative cal ICU did not have discussions with their on-
ICU outcome. cologists about the extent of treatment they wish,
It is clear from the available studies that there nor have they addressed their code status. This
are no absolute predictors of ICU outcome of lung adds an extra burden, and sometimes confusion,
cancer patients. It appears the outcome of these among physicians and family members about the
patients is dependent on clinical variables that are patient’s wishes. Critically ill lung cancer patients
similar to other patient populations and include may not be able to make informed decisions and
the performance status, the severity of the acute their family may not know the patients’ wishes.
illness and the number of organ system failures. Thoughtful decisions about end-of-life issues
Given the difficulty in determining the futility are difficult under these stressful situations. In
of ICU care for patients with lung cancer upon ini- a study by Reichner et al. [31], only 26% of lung
tial evaluation of their critical illness, it is reason- cancer patients admitted to the medical ICU had
able to offer this care to all critically ill lung cancer ‘do-not-resuscitate’ orders, and 64% of patients
patients except those who have advanced malig- with stage IV non-small cell lung cancer were full
nancy unresponsive to therapy, or have made a code. Of those patients who were full code on ad-
decision against aggressive therapy. Treatment of mission to the medical ICU, there was no record
these patients who are admitted to the medical of end-of-life discussions documented in any of
ICU should include appropriate diagnostic stud- the available outpatient records. The code status
ies, ventilatory support (either by non-invasive was subsequently changed to do-not-resuscitate
positive pressure ventilation or mechanical ven- in 49% of the patients a mean of 7 days after ad-
tilation), hemodynamic support and, when nec- mission to the medical ICU. The pulmonary and
essary, renal replacement therapy. These patients critical care physician was solely responsible for
should be re-evaluated in a few (3–5) days to as- the change in the code status in 65% of the cas-
sess their response to therapy [19–21]. If they are es. Another study by Lamont and Christakis [37]

182 Adam ⭈ Soubani


showed that physicians provide a frank estimate Recommendations
of prognosis in only one third of cases, and in the
rest provide no estimate or consciously over- or Lung cancer patients are increasingly being ad-
underestimate survival time. It may be under- mitted to the medical ICU for conditions relat-
standable for the oncologists not to discuss end- ed to their malignancy, side effects of treatment,
of-life care as they are starting treatment for these or co-morbid illnesses. The outcome of these
patients, however they are in the best position to patients has been progressively improving, and
have frank discussions with their patients and is approximating that of the general critically ill
family members, in the outpatient setting, after population. There are no absolute predictors of
they had a good rapport with them and had a time critical care outcome of these patients. The se-
to assess the patients’ health and response to ther- verity and the acuity of illness and the number
apy. An important resource to physicians caring of organ system failures appear to be more im-
for lung cancer patients is consultation with pal- portant than the characteristics of the underlying
liative care services that support the patient, fam- malignancy. Full ICU care should be considered
ily and physicians as they are going through treat- for all lung cancer patients who wish to have it, or
ment and in making end-of-life decisions. Recent those whose wishes are not known, except those
observations have shown that such services are who clearly have an advanced refractory disease.
underutilized, especially in critically ill patients. A multidisciplinary team approach including an
Furthermore, making use of the palliative care intensivist, medical oncologist, and palliative care
services has been associated with lowered cost for service is important for the appropriate manage-
care of cancer patients [38]. The recent American ment of these patients. It is reasonable to reassess
College of Chest Physicians guidelines for the di- the clinical status of these patients after 3–5 days
agnosis and management of lung cancer recom- of their care in the ICU to decide on further man-
mend that palliative care, including a consultation agement. Every effort should be made to discuss
with a palliative care team, be integrated into the the patient’s end-of-life wishes and code status by
treatment of patients with advanced lung cancer, their oncologist in the outpatient setting.
even those pursuing curative or life-prolonging
therapies [39].

References
1 American Cancer Society: Cancer Facts 5 Thun MJ, Henley SJ, Burns D, et al: Lung 9 Byhardt RW, Abrams R, Almagro U: The
and Figures 2009. Atlanta, American cancer death in lifelong nonsmoker. J association of adult respiratory distress
Cancer Society, 2009 (www.cancer.org). Natl Cancer Inst 2006;98:691–699. syndrome with thoracic irradiation. Int J
2 Jemal A, Siegel R, Ward E, et al: Cancer 6 Griffin JP, Nelson JE, Koch KA, et al: Radiat Oncol Biol Phys 1988;15:1441–
statistics, 2009. CA Cancer J Clin 2009; End-of-life care in patients with lung 1446.
59:225–249. cancer. Chest 2003;123:312S–31S. 10 Tang SS, Redmond K, Griffiths M, et al:
3 Jemal A, Chu KC, Tarone RE: Recent 7 Kress JP, Christenson J, Pohlman AS, et The mortality from acute respiratory
trends in lung cancer mortality in the al: Outcomes of critically ill cancer distress syndrome after pulmonary
United States. Cancer surveillance series. patients in a university hospital setting. resection is reducing: a 10-year single
J Natl Cancer Inst 2001;93:277–283. Am J Respir Crit Care Med 1999;160: institutional experience. Eur J Cardio-
4 Jemal A, Travis WD, Tarone RE, et al: 1957–1961. thorac Surg 2008;34:898–902.
Lung cancer rates convergence in young 8 Adam AK, Soubani AO: Outcome and 11 Groeger JS, White P, Nierman DM, et al:
men and women in the United States: prognostic factors of lung cancer Outcome for cancer patients requiring
analysis by birth cohort and histologic patients admitted to the medical inten- mechanical ventilation. J Clin Oncol
type. Int J Cancer 2003;105:101–107. sive care unit. Eur Respir J 2008;31: 1999;17:991–997.
47–53.

Critical Care Outcome of Lung Cancer Patients 183


12 Staudinger T, Stoiser B, Mullner M, et al: 21 Kongsgaard UE, Meidell NK: Mechanical 31 Reichner CA, Thompson JA, O’Brien S,
Outcome and prognostic factors in criti- ventilation in critically ill cancer et al: Outcome and code status of lung
cally ill cancer patients admitted to the patients: outcome and utilization of cancer patients admitted to the medical
intensive care unit. Crit Care Med resources. Support Care Cancer 1999;7: ICU. Chest 2006;130:719–723.
2000;28:1322–1328. 95–99. 32 Jennens RR, Rosenthal MA, Mitchell P,
13 Thiery G, Azoulay E, Darmon M, et al: 22 Benoit DD, Vandewoude KH, Decruyen- Presneill JJ: Outcome of patients admit-
Outcome of cancer patients considered aere JM, et al: Outcome and early prog- ted to the intensive care unit with newly
for intensive care unit admission: a hos- nostic indicators in patients with a diagnosed small cell lung cancer. Lung
pital-wide prospective study. J Clin hematologic malignancy admitted to the Cancer 2002;38:291–296.
Oncol 2005;23:4406–4413. intensive care unit for a life-threatening 33 Soares M, Darmon M, Salluh JI, et al:
14 Groeger JS, Lemeshow S, Price K, et al: complication. Crit Care Med 2003;31: Prognosis of lung cancer patients with
Multicenter outcome study of cancer 104–112. life-threatening complications. Chest
patients admitted to the intensive care 23 Soubani AO, Kseibi E, Bander JJ, et al: 2007;13:840–846.
unit: a probability of mortality model. J Outcome and prognostic factors of 34 Sharma G, Freeman J, Zhang D, Good-
Clin Oncol 1998;16:761–770. hematopoietic stem cell transplantation win JS: Trends in end-of-life ICU use
15 Azoulay E, Moreau D, Alberti C, et al: recipients admitted to a medical ICU. among older adults with advanced lung
Predictors of short-term mortality in Chest 2004;126:1604–1611. cancer. Chest 2008;133:72–78.
critically ill patients with solid malig- 24 Azoulay E, Alberti C, Bornstain C, et al: 35 Cuomo A, Delmastro M, Ceriana P, et al:
nancies. Intensive Care Med 2000;26: Improved survival in cancer patients Noninvasive mechanical ventilation as a
1817–1823. requiring mechanical ventilatory sup- palliative treatment of acute respiratory
16 Ghosen M, Kanso C, Kattan J, et al: Out- port: impact of noninvasive mechanical failure in patients with end-stage solid
come of cancer patients admitted to the ventilatory support. Crit Care Med 2001; cancer. Palliat Med 2004;18:602–610.
intensive care unit. J Med Liban 2002;50: 29:519–525. 36 Christodoulou C, Rizos M, Galani E, et
132–136. 25 Azoulay E, Thiery G, Chevret S, et al: al: Performance status: a simple predic-
17 Groeger JS, Glassman J, Nierman DM, et The prognosis of acute respiratory fail- tor of short-term outcome of cancer
al: Probability of mortality of critically ill ure in critically ill cancer patients. Medi- patients with solid tumors admitted to
cancer patients at 72 h of intensive care cine (Baltimore) 2004;83:360–370. the intensive care unit. Anticancer Res
unit management. Support Care Cancer 26 Mendoza V, Lee A, Marik PE: The hospi- 2007;27:2945–2948.
2003;11:686–695. tal – survival and prognostic factors of 37 Lamont EB, Christakis NA: Prognostic
18 Hauser MJ, Tabak J Baier H: Survival of patients with solid tumors admitted to disclosure to patients with cancer near
patients with cancer in a medical critical an ICU. Am J Hosp Palliat Care the end of life. Ann Intern Med
care unit. Arch Intern Med 1982;142: 2008;25:240–243. 2001;134:1096–105.
527–529. 27 Taccone FS, Artigas AA, Sprung CL, et 38 Fadul N, Elsayem A, Palmer JL, et al:
19 Schapira DV, Studnicki J, Bradham DD, al: Characteristics and outcomes of can- Predictors of access to palliative care
et al: Intensive care, survival and cer patients in European ICUs. Crit Care services among patients who died at a
expense of treating critically ill cancer 2009;13:R15. comprehensive cancer center. J Palliat
patients. JAMA 1993;269:783–786. 28 Ewer MS, Ali MK, Atta MS, et al: Out- Med 2007;10:1146–52.
20 Maschmeyer G, Bertschat FL, Moesta come of lung cancer patients requiring 39 Griffin JP, Koch KA, Nelson JE, Cooley
KT, et al: Outcome analysis of 189 con- mechanical ventilation for pulmonary ME: Palliative care consultation, quality-
secutive cancer patients referred to the failure. JAMA 1986;256:3364–3366. of-life measurements, and bereavement
intensive care unit as emergencies dur- 29 Boussat S, El’rini T, Dubiez A, et al: Pre- for end-of-life care in patients with lung
ing a 2-year period. Eur J Cancer 2003; dictive factors of death in primary lung cancer: ACCP evidence-based clinical
39:783–792. cancer patients on admission to the practice guidelines, ed 2. Chest
intensive care unit. Intensive Care Med 2007;132:404S–422S.
2000;26:1811–1816.
30 Lin YC, Tsai YH, Huang CC, et al: Out-
come of lung cancer patients with acute
respiratory failure requiring mechanical
ventilation. Respir Med 2004;98:43–51.

Ayman O. Soubani, MD
Division of Pulmonary, Critical Care and Sleep Medicine
Wayne State University School of Medicine
3990 John R Street, 3 Hudson, Detroit, MI 48201 (USA)
Tel. +1 313 745 8471, Fax +1 313 993 0562, E-Mail asoubani@med.wayne.edu

184 Adam ⭈ Soubani


Prognosis and Readmission
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 185–191

Readmission to the Intensive Care Unit for Patients


with Lung Edema or Atelectasis
Yoshinori Matsuoka  Akinoro Zaitsu  Makoto Hashizume
Department of Emergency and Critical Care Center, School of Medicine, Kyushu University Japan, Fukuoka, Japan

Abbreviations It is generally thought that weight reduction by


PaO2 Partial pressure of arterial oxygen
muscle contraction of 0–0.5 kg/day occurs during
P/F ratio PaO2/FiO2 ratio bed rest [1–3] and body weight gain due to im-
FiO2 Function of inspired O2 concentration proved nourishment occurs about 4 weeks later
PEEP Positive-pressure respiration [4–6]; therefore, we thought that the body weight
gain may have occurred due to internal surplus
water. In turn, this might cause lung edema or at-
Of 1,835 patients who entered the ICU during electasis and worsening of the respiratory state.
the 36 months from January 2003 to December This suggests that surplus lung water that was not
2005, 141 were readmitted within 1 month of removed during the first ICU stay may be associ-
their initial discharge. Acute respiratory failure ated with subsequent development of lung edema
by lung edema or atelectasis was present in 21 or atelectasis and early readmission to the ICU.
of these patients (14.9% of the readmitted pa- Therefore, we performed a retrospective inves-
tients, and 1.1% of all ICU patients). For these tigation of patient management during the first
21 patients, the body weight on the last day of ICU stay for patients readmitted to the ICU with
the first ICU stay was markedly higher than that lung edema or atelectasis.
on the first day. In addition, most of the patients
had normalized respiratory states at the time of
initial ICU discharge, but aggravation of the re- Analysis Main Topics Related with Your Title
spiratory state occurred immediately after dis-
charge, with lung consolidation. Patients requir- During the 36-month period from January 1, 2003
ing a longer period of artificial respiration and to December 31, 2005, 1,835 patients were admit-
those with a shorter ICU stay after extubation ted into our ICU. Of these patients, 141 (7.6% of
had a shorter time to ICU readmission. After re- the total number of admissions) were readmis-
admission, their respiratory state was improved sion cases, and 21 of the 141 patients were suffer-
by decreasing the body weight based on strict ing from acute respiratory failure due to lung ede-
water management. ma or atelectasis. Thus, patients with lung edema
or atelectasis comprised 14.9% of readmitted
Table 1. Clinical data during the first ICU admission period for the 21 readmitted acute respiratory failure patients
(1)

Initial body Final body Body weight Time to ICU readmission,


weight, kg weight, kg gain, % days

1 56.1 62.7 12 1
2 60.3 71.1 11.8 2
3 49.9 55.3 11 2
4 53.2 58.9 10.8 2
5 55.9 61.7 10.3 2
6 64.1 70.6 10.2 2
7 67.3 74 10 3
8 66 71.9 9 4
9 49.2 53.1 8 4
10 55 59.4 8 4
11 67 71.7 7 8
12 60.3 63.9 6 10
13 61.5 64.6 5 10
14 54 56.6 4.8 10
15 46.7 48.6 4 11
16 58.3 60.5 3.7 11
17 62.2 64.1 3 15
18 63 64.3 2.1 16
19 50.3 51.3 1.9 16
20 52.6 53.2 1.2 17
21 54.1 54.6 1 18

Body weight gain (%) = Final body weight (kg)  Initial body weight (kg).

patients and 1.1% of the total number of ICU pa- Acute respiratory failure was defined as PaO2
tients. We investigated the patients who had lung <60 Torr for indoor inspiration or a respiratory
consolidation and were diagnosed with acute re- disorder equivalent to PaO2 <60 Torr [7]. This
spiratory failure due to lung edema or atelecta- threshold is the definition of respiratory failure in
sis. Patients with a diagnosis of a disease due to a Japan. Respiratory function was evaluated using
specific bacterial cause, such as pneumonia, were the P/F ratio [8]. The initial and final body weight
excluded from the study. Since almost all patients during the first ICU period, body weight gain at
readmitted to the ICU more than 1 month after the time of ICU discharge (weight upon discharge
their initial ICU stay did not have a condition from the ICU ÷ weight when entering the ICU),
that was connected directly with the disease that the time to ICU readmission, the P/F ratio on the
caused the first ICU admission, we only investi- last day of the first ICU stay and on the first day
gated patients who were readmitted to the ICU of the second admission, the period using the
within 1 month of their initial ICU stay. respirator, the length of the initial ICU stay after

186 Matsuoka  Zaitsu  Hashizume


Table 2. Clinical data during the first ICU admission period for the 21 readmitted acute respiratory failure patients
(2)
P/F ratio 1 P/F ratio 2 R value E value R/E ratio

1 388 120 18 1 18
2 368 132 17 2 8.5
3 378 123 16 2 8
4 400 149 16 2 8
5 410 161 13 2 6.5
6 441 176 15 2 7.5
7 399 155 12 2 6
8 387 166 10 2 5
9 412 185 8 2 4
10 433 199 12 2 6
11 412 195 11 2 5.5
12 450 200 10 3 3.33
13 399 211 8 3 2.66
14 421 253 8 3 2.66
15 419 258 7 4 1.75
16 367 345 5 4 1.25
17 421 309 9 4 2.25
18 403 321 9 4 2.25
19 367 339 12 5 2.4
20 410 297 11 6 1.83
21 400 344 10 6 1.66

P/F ratio 1 = P/F ratio on the last day of the first ICU stay (mm Hg); P/F ratio 2 = P/F ratio on the first day of ICU
readmission (mm Hg); R value = the period using a respirator (days); E value = length of ICU stay after extubation
(days); R/E ratio = R value ÷ E value.

extubation, and the R/E ratio (the period using failure patients are shown in tables 1 and 2. A neg-
a respirator (R) ÷ the length of the ICU stay af- ative linear relationship was found between body
ter extubation (E)) were measured retrospective- weight gain at the time of initial ICU discharge
ly. Correlations were investigated between body and the time to ICU readmission (fig. 1). A weight
weight gain at the time of ICU discharge and the increase of more than 10% at the time of ICU dis-
time to ICU readmission, between body weight charge was likely to cause readmission to the ICU
gain and the P/F ratio at ICU readmission, be- within 3 days. A negative linear relationship was
tween the R/E ratio and the time to ICU readmis- also found between body weight gain at the time
sion, between the R/E ratio and body weight gain, of ICU discharge and the P/F ratio at ICU read-
and between body weight gain until extubation mission (fig. 2). A weight increase of more than
and the time to extubation. 10% at ICU discharge and a P/F ratio <150 were
Clinical data during the first ICU admission likely to result in readmission to the ICU due to
period for the 21 readmitted acute respiratory severe respiratory failure.

Readmission to the ICU for Patients with Lung Edema or Atelectasis 187
25

Time to ICU readmission (days)


Time to ICU readmission (days) 20
20
15

10 15

5 10

0 5

–5 0
0 5 10 15 0 5 10 15 20
Body weight gain at initial ICU discharge (%) R/E ratio

Fig. 1. A negative linear relationship (R = –0.98) was found Fig. 3. An inverse proportional relationship (R = –0.74)
between body weight gain at the initial ICU discharge and was found between the R/E ratio and the time to ICU re-
the time to ICU readmission. A weight increase of more admission. A large R/E ratio correlated with a short time
than 10% at the time of ICU discharge suggested readmis- to ICU readmission.
sion to the ICU within 3 days was likely.

16
400 Body weight gain (%)
readmission (mm Hg)

350 14
12
P/F ratio at ICU

300
250 10
200 8
150 6
100 4
50 2
0 0
0 5 10 15 0 5 10 15 20
Body weight gain at initial ICU discharge (%) R/E ratio

Fig. 2. A negative linear relationship (R = –0.96) was Fig. 4. A proportional relationship (R = 0.81) was found
found between body weight gain at the initial ICU dis- between the R/E ratio and body weight gain. A large R/E
charge and the P/F ratio at ICU readmission. A weight ratio correlated with a large body weight gain at the time
increase of more than 10% at ICU discharge or a P/F ra- of initial ICU discharge.
tio below 150 indicated probable readmission to the ICU
with severe respiratory failure.

An inverse relationship was found between low possibility of readmission to the ICU. A di-
the R/E ratio and the time to ICU readmission rect relationship was found between the R/E ra-
(fig. 3). A large R/E ratio indicates that the pa- tio and body weight gain (fig. 4); hence, a large
tient left the ICU soon after long-term respira- R/E ratio correlated with high body weight gain
tor management, and this was associated with at the time of ICU discharge. As shown in figure
a short period before ICU readmission. An R/E 1 or 2, a large body weight gain at ICU discharge
ratio of over 5 indicated that a patient was likely or a poor P/F ratio were associated with readmis-
to return to the ICU within 5 days. Conversely, a sion to the ICU due to severe respiratory failure.
patient with an R/E ratio of around 1 had a very A positive linear relationship was found between

188 Matsuoka  Zaitsu  Hashizume


Time to extubation (days)
permeability. The relative increase in the amount
14
of fluid outside the lung blood vessels causes al-
12 veolar collapse, a fall in lung compliance, lung
10
8 edema, and atelectasis, and this leads to pre-lung
6 failure. This kind of lung failure reflects a state in
4
2 which alveolar regions with poor ventilation are
0 formed locally. Even if breathing movement is sat-
0 5 10 15 20
Body weight gain until extubation (%) isfactory and alveolar ventilation is maintained,
full inspiration of oxygen cannot occur because a
Fig. 5. A positive linear relationship (R = 0.75) was found
blood-flow shunt has arisen in the lungs [19–22].
between body weight gain until extubation and the time Once the lung blood-vessel endothelium is re-
to extubation. As the weight increased during respirator stored, the permeability is reduced and the cir-
use, the time to extubation was prolonged. culation is stabilized, the transfusion procedure
should enter the refilling stage [23]. At this stage,
since extracellular fluid has returned to the lung
blood vessels, diuresis is used to normalize the
body weight gain until extubation and the time to amount of fluid outside the blood vessels. This re-
extubation (fig. 5); as weight increased during the quires immediate reduction of transfusion, per-
period of respiratory support, the time to extuba- formance of diuresis and prompt discharge of the
tion lengthened. extracellular fluid from the body. Improvement of
oxygenation is also important for recovery from
pre-lung failure. However, judgment of the tim-
Discussion ing of this procedure is not easy in practice, and
often there is a failure to normalize the extracellu-
Cytokine release is stimulated by inflammation, lar fluid due to continuation of superfluous trans-
and these cytokines stimulate adherence of leuko- fusion and inadequate diuresis. Consolidation of
cytes and monocytes to endothelial cells of blood the lungs may therefore arise, and this kind of lung
vessels. Elastase and oxygen radicals released by failure might be viewed as iatrogenic respiratory
leukocytes may then cause damage to the en- failure. Many cases of lung edema in the ICU may
dothelial cells [9–11], causing circulation failure not also be due to bacteria [24, 25].
and increased blood-vessel permeability [12–15]. The development of respirators has allowed
Transfusion can be performed to compensate maintenance of oxygenation, even in patients
for the insufficient amount of plasma due to in- with severe lung failure, by providing the correct
creased blood-vessel permeability and this may amount of ventilation. PEEP is particularly effec-
stabilize the circulation dynamics. tive for atelectasis caused by lung edema, because
The lungs are especially prone to leukocyte-in- it increases the lung expiration capacity, widens
duced damage. Once the blood-vessel endothelial the peripheral respiratory tract, and re-expands
cells are damaged by activated leukocytes in the the collapsed alveolar space. As a result, oxygen-
acute phase, increased blood-vessel permeability ation appears to improve, but if PEEP is stopped
in the lungs allows plasma to move into the al- the respiratory state worsens; hence, this proce-
veolar space from outside the lung blood vessels dure does not address the fundamental causes of
[16–18]. Transfusion can be performed to stabi- lung failure. The basic treatment for lung failure
lize the circulation, but plasma may leak outside is simply to normalize surplus extracellular flu-
of the lung blood vessels due to the increased id outside the lung blood vessels. Extubation by

Readmission to the ICU for Patients with Lung Edema or Atelectasis 189
PEEP before the extracellular fluid is normalized Reduction of the R value (the period of respira-
does allow oxygenation to be maintained at a high tory support) would also have a favorable effect
concentration of oxygen, but the alveolar space on the R/E ratio.
opened by PEEP collapses in a short time and this From figure 5, losing weight during the period
may cause lung consolidation in this region. of respiratory support is likely to lead to early ex-
In the current work, a negative linear relation- tubation, since the time to extubation was related
ship was found between body weight gain at the to body weight gain. In other words, losing body
time of ICU discharge and the time to ICU re- weight at the refilling stage prevents ICU read-
admission (fig. 1) and between body weight gain mission and may decrease the length of the ICU
at the time of ICU discharge and the P/F ratio at stay. Fluid management failure during the first
the time of ICU readmission (fig. 2). A weight in- ICU stay may result in ICU readmission due to
crease of more than 10% at ICU discharge or a lung edema or atelectasis. A key for prevention
P/F ratio of below 150 was associated with read- of ICU readmission is to obtain complete reversal
mission to the ICU within 3 days. Body weight of lung failure before ICU discharge, using strict
should decrease steadily from entering the ICU water management based on weight measure-
by about 0.5 kg/day due to muscle wasting by ments and with care not to assume that an appar-
bed rest, as mentioned above; therefore, the ac- ent improvement in the respiratory state will be
tual body weight gain at ICU discharge is larger sustained, since this improvement may be due to
than the measured value, since some weight loss respiratory support.
should have occurred in the ICU.
A large R/E ratio, which indicates that a patient
left the ICU soon after long-term respirator man- Recommendations
agement, was associated with a large body weight
gain at the time of ICU discharge (fig. 4). Since a • Prevention of ICU readmission requires
large body weight gain tends to lead to ICU read- complete recovery from lung failure before
mission (fig. 1 or 2), it is desirable to have a small initial ICU discharge.
R/E ratio. From figure 3, readmission to the ICU • Strict water management based on body weight
is extremely unlikely when the R/E ratio is around measurement is essential at the refilling stage.
1, suggesting that it is preferable to keep patients • A decrease in body weight at this stage prevents
in the ICU after extubation for the same period ICU readmission and may decrease the length
for which a respirator was used. However, in prac- of the initial ICU stay.
tice this is difficult due to medical costs and the
desire to limit the number of hospitalization days.

References
1 Forbes GB: Weight loss during fasting: 3 Drenick EJ, Swendseid ME, Blahd WH, 5 Long CL: Energy balance and carbohy-
implications for the obese. Am J Clin Tuttle SG: Prolonged starvation as treat- drate metabolism in infection and sep-
Nutr 1970;23:1212–1219. ment for severe obesity. JAMA 1964;187: sis. Am J Clin Nutr 1977;30:1301–1310.
2 Benedict FG: A Study of Prolonged Fast- 100–105. 6 Long CL, Kinney JM, Geiger JW: Non-
ing. Washington, Carnegie Institute of 4 Spady DW, Payne PR, Picou D, Waterlow suppressability of gluconeogenesis by
Washington, 1915, pp 69–87. JC: Energy balance during recovery from glucose in septic patients. Metabolism
malnutrition. Am J Clin Nutr 1976;29: 1976;25:193–201.
1073–1088.

190 Matsuoka  Zaitsu  Hashizume


7 Zaitsu A: The latest trends in the treat- 13 Endo S, Inada K, Ceska M, Takakuwa T, 19 Dantzger DR: Pulmonary gas exchange;
ment of acute respiratory failure. Fuku- Yamada Y, Nakae H, Kasai T, Yamashita in Dantzger DR (ed): Cardiopulmonary
oka Igaku Zasshi 1990;81:391–295. H, Taki K, Yoshida M: Plasma interleu- Critical Care, ed 2. Philadelphia, Saun-
8 Covelli HD, Nessan VJ, Tuttle WK: Oxy- kin-8 and polymorphonuclear leukocyte ders, 1991, pp 25–43.
gen derived variables in acute respira- elastase concentrations in patients with 20 Lanken PN: Ventilation-perfusion rela-
tory failure. Crit Care Med 1983;11: septic shock. J Inflamm 1995;45:136– tionships; in Grippi MA (ed): Pulmonary
646–649. 142. Pathophysiology. Philadelphia, Lippin-
9 Nakae H, Endo S, Inada K, Kasai T, 14 Petty TL: Protease mechanisms in the cott, 1995, pp 195–210.
Yoshida M: Significance of α-tocopherol pathogenesis of acute lung injury. Ann 21 Buohuys A: Respiratory dead space; in
and interleukin-8 in septic adult respira- NY Acad Sci 1991;624:267–277. Fenn WO, Rahn H (eds): Handbook of
tory distress syndrome. Res Commun 15 Edens HA, Parkos CA: Neutrophil Physiology: Respiration. Bethesda,
Chem Pathol Pharmacol 1994;84:197– transendothelial migration and altera- American Physiological Society, 1964,
202. tion in vascular permeability: focus on pp 699–714.
10 Endo S, Inada K, Inoue Y, Kuwata Y, neutrophil-derived azurocidin. Curr 22 D’Alonzo GE, Dantzker DR: Mechanisms
Suzuki M, Yamashita H, Hoshi S, Opin Hematol 2003;10:25–30. of abnormal gas exchange. Med Clin
Yoshida M: Two types of septic shock 16 Su X, Camerer E, Hamilton JR, Coughlin North Am 1983;67:557–571.
classified by the plasma levels of cytok- SR, Matthay MA: Protease-activated 23 Matsui M, Kudo T, Kudo M, Ishihara H,
ines and endotoxin. Circ Shock 1992;38: receptor-2 activation induces acute lung Matsuki A: The endocrine response after
264–274. inflammation by neuropeptide-depen- burns. Agressologie 1991;32:233–235.
11 Endo S, Inada K, Yamada Y, Kasai T, dent mechanisms. J Immunol 2005;175: 24 Meduri GU, Mauldin GL, Wunderink
Takakuwa T, Nakae H, Kikuchi M, Hoshi 2598–2605. RG, et al: Causes of fever and pulmonary
S, Suzuki M, Yamashita H, et al: Plasma 17 Guo M, Wu MH, Granger HJ, Yuan SY: densities in patients with clinical mani-
tumour necrosis factor-α levels in Focal adhesion kinase in neutrophil- festations of ventilator-associated pneu-
patients with burns. Burns 1993; induced microvascular hyperpermeabil- monia. Chest 1994;106:221–235.
19:124–127. ity. Microcirculation 2005;12:223–232. 25 Bates JH, Campbell GD, Barron AL, et al:
12 Endo S, Inada K, Yamashita H, 18 Brigham KL, Meyrick B: Endotoxin and Microbial etiology of acute pneumonia
Takakuwa T, Nakae H, Kasai T, Kikuchi lung injury. Am Rev Respir Dis 1986; in hospitalized patients. Chest 1992;101:
M, Ogawa M, Uchida K, Yoshida M: 133:913–927. 1005–1012.
Platelet-activating factor acetylhydrolase
activity, type II phospholipase A2, and
cytokine levels in patients with sepsis.
Res Commun Chem Pathol Pharmacol
1994;83:289–295.

Yoshinori Matsuoka, MD
Department of Emergency and Critical Care Center
School of Medicine, Kyushu University Japan
3-1-1 Maidashi Higashiku, Fukuokashi, Fukuoka 812-8582 (Japan)
Tel. +81 92 642 5871, Fax +81 92 642 5874, E-Mail yoshinori216@h2.dion.ne.jp

Readmission to the ICU for Patients with Lung Edema or Atelectasis 191
Section TitleRehabilitation and Technology
Pulmonary
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 192–196

Early Mobilization in the Intensive Care Unit:


Safety, Feasibility, and Benefits
Radha Korupolua ⭈ Jeneen M. Giffordb ⭈ Jennifer M. Zannic ⭈ Alex Truonga ⭈
Gangadhar Vajralaa ⭈ Scott Lepred ⭈ Dale M. Needhama,d
a
Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, bDepartment of Internal Medicine, Johns Hopkins
Bayview Medical Center, cDepartment of Physical Medicine and Rehabilitation, Johns Hopkins Hospital, and dDepartment of
Physical Medicine and Rehabilitation, Johns Hopkins University, Baltimore, Md., USA

Deconditioning and weakness are problems com- (6%) patients receiving ‘usual care’ in the ICU
monly experienced by ICU patients [1–4]. Bed rest [11]. In another study of 150 acute lung injury pa-
and immobility contribute to the development of tients in the ICU, only 27% received physical ther-
these neuromuscular complications, which can apy which occurred on only 6% of all ICU days
be severe and long lasting in ICU survivors [1– [12]. Likewise, in a smaller study of 20 physiologi-
3]. The benefits of physical medicine and reha- cally stable ICU patients, physical activity was ob-
bilitation in patients with prolonged mechanical served over two separate 4-hour periods that were
ventilation have been well recognized [5–8], and variably scheduled throughout the day. During a
recently there has been growing interest in evalu- total of 156 h of observation, only two instances of
ating early initiation of mobilization activities in sitting and one instance of standing were record-
the acute ICU setting [4, 9, 10]. Existing studies ed. No patient ambulated, and all other activities
have demonstrated that early mobilization is safe, consisted solely of a passive range of motion and
feasible, and associated with short-term benefits turning the patient in bed [13].
in critically ill patients. One systematic review included 24 studies
In this chapter, we will briefly review the rel- which evaluated neuromuscular abnormalities in
evant epidemiology and pathophysiology of im- 1,421 adult ICU patients with sepsis, multiorgan
mobility and bed rest. Thereafter, we will discuss dysfunction, or prolonged mechanical ventilation
early mobilization of critically ill patients specifi- [14]. In this review, 46% of subjects had critical ill-
cally addressing its safety, feasibility and potential ness neuromuscular abnormalities diagnosed us-
benefits. ing electrophysiological testing, with or without
associated physical examination. A separate study
used physical examination to diagnose neuromus-
Epidemiology cular weakness in awake patients who required
>7 days of mechanical ventilation. In this cohort,
Routine use of physical medicine and rehabilita- ICU-acquired weakness was clinically diagnosed
tion therapy in the ICU is relatively rare. In one in 25% of 95 patients [1]. Neuromuscular weakness
study, physical therapy was given to only 8 of 135 acquired in the ICU can persist for months or years
after hospital discharge, causing impairments in recovery. The term ‘early’ is used to emphasize the
physical function and quality of life [2, 3, 15]. important difference in timing between this strat-
egy and current ICU practice where mechanically
ventilated patients frequently are heavily sedated
Pathophysiology of Bed Rest and Immobility and unable to actively participate in mobilization
activities until after extubation or ICU discharge.
Immobility has deleterious effects on muscle that By contrast, with early mobilization, heavy seda-
may contribute to the development of ICU-acquired tion is avoided, with a goal of balancing patient
weakness. Experimental studies in animals have alertness and comfort. In this setting, physical
demonstrated that immobility activates multiple activity may commence immediately after ma-
molecular pathways which result in an overall im- jor physiological derangements have been stabi-
balance between protein degradation and protein lized, often within 24–72 h of ICU admission [28].
synthesis, resulting in a net loss of muscle fibers Mobilization of patients may occur despite their
[16–18]. Studies in healthy individuals have dem- ongoing requirements for moderate levels of sup-
onstrated that long-term bed rest (i.e. >30 days) port from mechanical ventilation, vasopressor in-
leads to a state of systemic inflammation, with in- fusions, and other ICU therapies.
creased reactive oxide species [19] and inflamma-
tory cytokines [20], as well as a transient decrease
in protective antioxidants [21]. Together, these Safety
changes create an inflammatory environment that
is toxic to muscles through inhibition of anabolic The safety of early physical medicine and rehabili-
pathways, activation of catabolic pathways, and di- tation activities is of particular concern in patients
rect interaction with muscle fibers to interrupt con- who are critically ill. Three published studies have
tractile force. Insulin resistance and increased sen- specifically analyzed this issue. In one study con-
sitivity to the catabolic effects of cortisol are other ducted in a respiratory ICU, 103 mechanically
mechanisms that have been implicated in muscle ventilated patients with acute respiratory failure
atrophy associated with bed rest [22–24]. performed a total of 1,449 activity events, includ-
Bed rest also results in important cardiovascu- ing sitting on the edge of the bed, sitting in a chair,
lar changes that can decrease exercise tolerance. and ambulation. Safety-related issues occurred in
Underlying physiological mechanisms include less than 1% of all activity events, and all were cor-
abnormal baroreceptor response contributing to rected promptly without any negative sequelae at-
postural hypotension and tachycardia, and de- tributable to the issue. Of all activities, 41% were
creased stroke volume, cardiac output, and peak conducted in intubated patients without any extu-
oxygen uptake [25, 26]. In addition, patients ex- bations occurring during therapy [28]. Similarly,
posed to prolonged periods of bed rest often expe- a controlled trial in a medical ICU analyzed the
rience changes in mood which may also contrib- effect of an early mobility protocol, initiated with-
ute to one’s decreased physical function [27]. in 48 h of mechanical ventilation. None of the 106
patients in the early mobility protocol arm experi-
enced accidental dislodgement of any medical de-
Early Mobilization in the ICU vice or any life-threatening situation [11]. Finally,
in a third study conducted in a medical ICU, 69
The goals of early mobilization in the ICU are to mobilization activity events were performed on
mitigate the effects of decreased physical activity, 31 patients. Only three adverse events were re-
preserve patients’ physical function, and expedite ported: all were desaturation episodes which only

Early Mobilization in the ICU 193


required a temporary increase in oxygen require- rehabilitation and promoting safety and efficien-
ment [29]. cy. Challenging long-standing assumptions about
Based on these studies, we have proposed an the need for heavy sedation and bed rest in criti-
algorithm for selecting ICU patients who are ap- cally ill patients is important in instituting ‘culture
propriate for early mobilization [10]. Patients are change’ in the ICU. Successful culture change in-
assessed daily for the ability to actively participate. volves interdisciplinary training, support from all
Early mobilization is deferred in patients who are levels of leadership, and a commitment from the
deeply sedated or comatose. Criteria for cardiac ICU clinical staff and administration to make mo-
and pulmonary stability include (1) FiO2 ≤0.6, (2) bilization a priority in patient care. If successful,
PEEP ≤10 cm H2O, (3) no increase in vasopressor such changes can improve teamwork, safety and
dose in the past 2 h, (4) no continuous infusion consistency of ICU care, as well as minimize ICU-
of a vasodilator medication, (5) no new anti-ar- acquired weakness and impairments in physical
rhythmic agent, and (6) no new cardiac ischemia. function [10, 30, 31].
Clinical judgment from all clinicians participat-
ing in patient care is necessary to determine oth-
er contraindications and to identify patients who Potential Benefits of Early Mobilization
may benefit from a trial of therapy despite failing
to meet these specific criteria. Mobilization of patients in the ICU can prevent
the negative sequelae of immobility and enhance
physical function in the hospital. In patients with
Feasibility chronic pulmonary disease or requirement for
long-term mechanical ventilation, physical medi-
Common mobilization activities that are feasible cine and rehabilitation therapy improves mobil-
for mechanically ventilated patients in the ICU ity, increases overall strength, improves weaning
setting include transferring from the supine posi- from mechanical ventilation, increases indepen-
tion to an unsupported sitting position at the edge dence with activities of daily living, and reduces
of the bed, standing, transferring into a chair, and ICU length of stay [5–8]. There is growing evi-
ambulation [11, 28–30]. Ambulation in the typi- dence that these benefits may also occur with
cal mechanically ventilated patient requires the physical medicine and rehabilitation therapy in
combined efforts of a physical therapist, nurse, re- ICU patients who have a greater acuity of illness.
spiratory therapist and technician to safely man- Recent studies of early mobility in the ICU have
age the patient and the associated lines, tubes and demonstrated improved physical function [11, 28,
life support equipment [11, 28]. In addition to 30], shortened length of stay in ICU and hospital
mobilization activities, patients may benefit from [11, 31], and improved weaning from mechanical
resistance training and range of motion activities ventilation [31].
to promote muscle strengthening and joint mo- One study of an early mobility program involv-
bility. As previously outlined, these activities are ing 103 mechanically ventilated patients demon-
feasible and safe, even in ICU patients requiring strated that 69% of patients walked more than
mechanical ventilation via an endotracheal tube 100 ft by ICU discharge. A subsequent study at
[28, 31]. Additionally, this therapy can be accom- the same medical center used a different cohort
plished without any increase in cost [28, 31]. of 104 mechanically ventilated patients and dem-
Promoting early mobilization in the ICU re- onstrated that 91 patients (88%) walked a medi-
quires a multidisciplinary team approach to pa- an (interquartile range) distance of 200 (0–800)
tient care with the goal of minimizing barriers to ft by ICU discharge [31]. During a 6-year period

194 Korupolu ⭈ Gifford ⭈ Zanni ⭈ Truong ⭈ Vajrala ⭈ Lepre ⭈ Needham


in which the early mobility program was ongo- Conclusion
ing at this hospital, ICU and hospital length of
stay steadily decreased, the proportion of patients Neuromuscular complications in survivors of
receiving tracheotomy decreased from 29 to 5%, critical illness are common and may cause long-
and the proportion of patients with weaning fail- lasting impairments in physical function and
ure decreased from 12 to 3% [31]. Collectively, quality of life. Prolonged bed rest and immobility
these reports demonstrate mobilization in the are important factors in the development of these
ICU can be beneficial for patient outcomes and complications. Early mobilization may diminish
hospital resource utilization. Beneficial results the detrimental effects of bed rest. Recent studies
were observed at another hospital where a con- have demonstrated that early mobilization is safe,
trolled trial demonstrated that an early mobility feasible, and beneficial for mechanically ventilat-
protocol was associated with a shorter time from ed ICU patients. Large multicenter randomized
ICU admission to getting out of bed (5.0 days in trials will enhance the validity and generalizabili-
the mobility group vs. 11.3 days in the ‘usual care’ ty of these existing studies. Future research should
control group, p < 0.001), a shorter risk-adjusted address the effects of early mobility on ICU pa-
ICU length of stay (5.5 vs. 6.9 days, p = 0.025), and tients’ long-term outcomes, including muscle
a decreased risk-adjusted hospital length of stay strength, physical function, quality of life, and re-
(11.2 vs. 14.5 days, p = 0.006) [11]. This controlled source utilization after hospital discharge.
trial again demonstrates that early mobilization
in the ICU is associated with improved physical
function and decreased resource utilization.

References
1 De Jonghe B, Sharshar T, Lefaucheur JP, 7 Make B, Gilmartin M, Brody JS, Snider 12 Needham DM, Wang W, Desai SV, et al:
et al: Paresis acquired in the intensive GL: Rehabilitation of ventilator-depen- Intensive care unit exposures for long-
care unit: a prospective multicenter dent subjects with lung diseases. The term outcomes research: development
study. JAMA 2002;288:2859–2867. concept and initial experience. Chest and description of exposures for 150
2 Fletcher SN, Kennedy DD, Ghosh IR, et 1984;86:358–365. patients with acute lung injury. J Crit
al: Persistent neuromuscular and neuro- 8 Chiang LL, Wang LY, Wu CP, Wu HD, Care 2007;22:275–284.
physiologic abnormalities in long-term Wu YT: Effects of physical training on 13 Winkelman C, Higgins PA, Chen YJ:
survivors of prolonged critical illness. functional status in patients with pro- Activity in the chronically critically ill.
Crit Care Med 2003;31:1012–1016. longed mechanical ventilation. Phys Dimens Crit Care Nurs 2005;24:281–
3 Herridge MS, Cheung AM, Tansey CM, Ther 2006;86:1271–1281. 290.
et al: One-year outcomes in survivors of 9 Herridge MS: Mobile, awake and criti- 14 Stevens RD, Dowdy DW, Michaels RK,
the acute respiratory distress syndrome. cally ill. CMAJ 2008;178:725–726. Mendez-Tellez PA, Pronovost PJ, Need-
N Engl J Med 2003;348:683–693. 10 Korupolu R, Gifford JM, Needham DM: ham DM: Neuromuscular dysfunction
4 Needham DM: Mobilizing patients in Early mobilization of critically ill acquired in critical illness: a systematic
the intensive care unit: improving neu- patients: reducing neuromuscular com- review. Intensive Care Med
romuscular weakness and physical func- plications after intensive care. Contemp 2007;33:1876–1891.
tion. JAMA 2008;300:1685–1690. Crit Care 2009;6:1–12. 15 Dowdy DW, Eid MP, Dennison CR, et al:
5 Nava S: Rehabilitation of patients admit- 11 Morris PE, Goad A, Thompson C, et al: Quality of life after acute respiratory
ted to a respiratory intensive care unit. Early intensive care unit mobility ther- distress syndrome: a meta-analysis.
Arch Phys Med Rehabil 1998;79:849– apy in the treatment of acute respiratory Intensive Care Med 2006;32:1115–1124.
854. failure. Crit Care Med 2008;36:2238– 16 Paddon-Jones D, Sheffield-Moore M,
6 Martin UJ, Hincapie L, Nimchuk M, 2243. Cree MG, et al: Atrophy and impaired
Gaughan J, Criner GJ: Impact of whole- muscle protein synthesis during pro-
body rehabilitation in patients receiving longed inactivity and stress. J Clin Endo-
chronic mechanical ventilation. Crit crinol Metab 2006;91:4836–4841.
Care Med 2005;33:2259–2265.

Early Mobilization in the ICU 195


17 Taillandier D, Aurousseau E, Meynial- 22 Ferrando AA, Lane HW, Stuart CA, 27 Hough CL, Needham DM: The role of
Denis D, et al: Coordinate activation of Davis-Street J, Wolfe RR: Prolonged bed future longitudinal studies in ICU survi-
lysosomal, Ca2+-activated and ATP- rest decreases skeletal muscle and vors: understanding determinants and
ubiquitin-dependent proteinases in the whole-body protein synthesis. Am J pathophysiology of weakness and neuro-
unweighted rat soleus muscle. Biochem J Physiol 1996;270:E627–E633. muscular dysfunction. Curr Opin Crit
1996;316:65–72. 23 Hamburg NM, McMackin CJ, Huang AL, Care 2007;13:489–496.
18 Tidball JG, Spencer MJ: Calpains and et al: Physical inactivity rapidly induces 28 Bailey P, Thomsen GE, Spuhler VJ, et al:
muscular dystrophies. Int J Biochem Cell insulin resistance and microvascular Early activity is feasible and safe in
Biol 2000;32:1–5. dysfunction in healthy volunteers. Arte- respiratory failure patients. Crit Care
19 Pawlak W, Kedziora J, Zolynski K, Kedzi- rioscler Thromb Vasc Biol Med 2007;35:139–145.
ora-Kornatowska K, Blaszczyk J, Wit- 2007;27:2650–2656. 29 Stiller K, Phillips A, Lambert P: The
kowski P: Free radicals generation by 24 Wang X, Hu Z, Du J, Mitch WE: Insulin safety of mobilisation and its effects on
granulocytes from men during bed rest. resistance accelerates muscle protein haemodynamics and respiratory status
J Gravit Physiol 1998;5:131–132. degradation: activation of the ubiquitin- of intensive care patients. Physiother
20 Schmitt DA, Schwarzenberg M, Tkaczuk proteasome pathway by defects in mus- Theory Pract 2004;20:175–185.
J, et al: Head-down tilt bed rest and cle cell signaling. Endocrinology 30 Thomsen GE, Snow GL, Rodriguez L,
immune responses. Pflugers Arch 2006;147:4160–4168. Hopkins RO: Patients with respiratory
2000;441(suppl):R79–R84. 25 Convertino VA, Bloomfield SA, Green- failure increase ambulation after transfer
21 Pawlak W, Kedziora J, Zolynski K, et al: leaf JE: An overview of the issues: physi- to an intensive care unit where early
Effect of long-term bed rest in men on ological effects of bed rest and restricted activity is a priority. Crit Care Med 2008;
enzymatic antioxidative defence and physical activity. Med Sci Sports Exerc 36:1119–1124.
lipid peroxidation in erythrocytes. J 1997;29:187–190. 31 Hopkins RO, Spuhler VJ, Thomsen GE:
Gravit Physiol 1998;5:163–164. 26 Fortney SM, Schneider VS, Greenleaf JE: Transforming ICU culture to facilitate
The physiology of bed rest; in Fregly MJ, early mobility. Crit Care Clin
Blatteis CM (eds): Handbook of Physiol- 2007;23:81–96.
ogy. New York, Oxford University Press,
1996, pp 889–939.

Dale M. Needham, MD, PhD


Division of Pulmonary and Critical Care Medicine, Johns Hopkins University
1830 E. Monument St, 5th Floor, Baltimore, MD 21205 (USA)
Tel. +1 410 955 3467, Fax +1 410 955 0036
E-Mail dale.needham@jhmi.edu

196 Korupolu ⭈ Gifford ⭈ Zanni ⭈ Truong ⭈ Vajrala ⭈ Lepre ⭈ Needham


Pulmonary Rehabilitation and Technology
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 197–201

Evidence-Based Guidelines in Pulmonary


Rehabilitation
Andrew L. Ries
School of Medicine, University of California, San Diego, Calif., USA

Abbreviations then, in conjunction with ACCP in 1997, the first


AACVPR American Association of Cardiovascular and
evidence-based guidelines [5]. Since that time, the
Pulmonary Rehabilitation published literature on pulmonary rehabilitation
ACCP American College of Chest Physicians has increased substantially, providing justifica-
ATS American Thoracic Society tion for recommending pulmonary rehabilitation
COPD Chronic obstructive pulmonary disease as part of standards of care for the management of
ERS European Respiratory Society patients with COPD and other chronic lung dis-
eases [3, 6, 7]. Therefore, ACCP and AACVPR de-
cided to update the 1997 guidelines with a system-
Pulmonary rehabilitation has emerged as a rec- atic, evidence-based review of the literature that
ommended standard of care for patients with was published recently [8].
chronic lung disease based on a growing body
of scientific evidence. Several organizations have
taken a lead role in championing pulmonary re- Main Topics
habilitation and developing comprehensive state-
ments, practice guidelines, and evidence-based The ATS and ERS recently adopted the following
guidelines. Documenting the scientific evidence definition of pulmonary rehabilitation [3]:
underlying clinical practice has been important Pulmonary rehabilitation is an evidence-based,
in overcoming skepticism and convincing health multidisciplinary, and comprehensive intervention
professionals, healthcare institutions, third-party for patients with chronic respiratory diseases who
payors, and regulatory agencies to support pul- are symptomatic and often have decreased daily life
monary rehabilitation. activities. Integrated into the individualized treat-
The first definition of pulmonary rehabilita- ment of the patient, pulmonary rehabilitation is
tion was developed in 1974 by the ACCP, and the designed to reduce symptoms, optimize functional
first comprehensive statement published by ATS status, increase participation, and reduce health-
in 1981 [1]. ATS subsequently updated its state- care costs through stabilizing or reversing systemic
ment in 1999 [2] and then, in conjunction with manifestations of the disease.
ERS, in 2006 [3]. The first systematic review of This definition focuses on three important fea-
the scientific basis of pulmonary rehabilitation tures of successful rehabilitation: (1) a multidisci-
was published by the AACVPR in 1990 [4] and plinary approach; (2) an individualized program
Table 1a. Relationship of strength of the supporting evidence to the balance of benefits to risks and burdens [10]

Strength of evidence Balance of benefits to risks and burdens

benefits outweigh risks/burdens outweigh evenly balanced uncertain


risks/burdens benefits
High 1A 1A 2A
Moderate 1B 1B 2B
Low or very low 1C 1C 2C 2C

1A: strong recommendation. 2A: weak recommendation.


1B: strong recommendation. 2B: weak recommendation.
1C: strong recommendation. 2C: weak recommendation.

Table 1b. Description of balance of benefits to risks/burdens scale

Benefits clearly outweigh the risks and burdens Certainty of imbalance


Risks and burdens clearly outweigh the benefits Certainty of imbalance
Risks/burdens and benefits are closely balanced Less certainty
Balance of benefits to risks and burdens is uncertain Uncertainty

tailored to the patients needs, and (3) attention to patients with diseases other than COPD such as
physical, psychological and social function. interstitial diseases, cystic fibrosis, bronchiectasis,
Rehabilitation programs for patients with and thoracic cage abnormalities.
chronic lung disease are well established as a In the recently updated evidence-based guide-
means of enhancing standard therapy in order lines, the ACCP/AACVPR Panel focused on stud-
to control and alleviate symptoms and optimize ies published since the previous 1997 review, con-
functional capacity [3, 5, 7, 9]. The primary goal is centrating on published literature in patients with
to restore the patient to the highest possible level COPD. Because of the many advances and new
of independent function which is accomplished areas of investigation, the Panel not only updat-
by helping patients learn more about their disease, ed the areas reviewed and recommendations in
treatments, and coping strategies. the previous guideline [5] but also reviewed addi-
Pulmonary rehabilitation is appropriate for tional new topics. Recommendations were devel-
any patient with stable chronic lung disease who oped for several outcomes of comprehensive pul-
is disabled by respiratory symptoms. Programs monary rehabilitation programs including lower
typically include components such as patient as- extremity exercise training, dyspnea, health-relat-
sessment, exercise training, education, nutri- ed quality of life, healthcare utilization, survival,
tional intervention, and psychosocial support. psychosocial outcomes, and long-term benefits.
These programs have been successfully applied to Additional topics reviewed include duration of

198 Ries
Table 2. Recommendations and ratings of the evidence-based guidelines

1. Recommendation: A program of exercise training of the muscles of ambulation is recommended as a


mandatory component of pulmonary rehabilitation for patients with COPD. Grade of Recommendation: 1A

2. Recommendation: Pulmonary rehabilitation improves the symptom of dyspnea in patients with COPD. Grade
of Recommendation: 1A

3. Recommendation: Pulmonary rehabilitation improves health-related quality of life in patients with COPD.
Grade of Recommendation: 1A

4. Recommendation: Pulmonary rehabilitation reduces the number of hospital days and other measures of
healthcare utilization in patients with COPD. Grade of Recommendation: 2B

5. Recommendation: Pulmonary rehabilitation is cost-effective in patients with COPD. Grade of


Recommendation: 2C

6. Statement: There is insufficient evidence to determine if pulmonary rehabilitation improves survival in


patients with COPD. No recommendation is provided.

7. Recommendation: There are psychosocial benefits from comprehensive pulmonary rehabilitation programs
in patients with COPD. Grade of Recommendation: 2B

8. Recommendation: 6–12 weeks of pulmonary rehabilitation produces benefits in several outcomes that
decline gradually over 12–18 months. Grade of Recommendation: 1A
Some benefits, such as health-related quality of life, remain above control at 12–18 months. Grade of
Recommendation: 1C

9. Recommendation: Longer pulmonary rehabilitation programs (beyond 12 weeks) produce greater sustained
benefits than shorter programs. Grade of Recommendation: 2C

10. Recommendation: Maintenance strategies following pulmonary rehabilitation have a modest effect on long-
term outcomes. Grade of Recommendation: 2C

11. Recommendation: Lower extremity exercise training at higher exercise intensity produces greater
physiologic benefits than lower intensity training in patients with COPD. Grade of Recommendation: 1B

12. Recommendation: Both low- and high-intensity exercise training produce clinical benefits for patients with
COPD. Grade of Recommendation: 1A

13. Recommendation: Addition of a strength training component to a program of pulmonary rehabilitation


increases muscle strength and muscle mass. Strength of evidence: 1A

14. Recommendation: Current scientific evidence does not support the routine use of anabolic agents in
pulmonary rehabilitation for patients with COPD. Grade of Recommendation: 2C

15. Recommendation: Unsupported endurance training of the upper extremities is beneficial in patients with
COPD and should be included in pulmonary rehabilitation programs. Grade of Recommendation: 1A

16. Recommendation: The scientific evidence does not support the routine use of inspiratory muscle training as
an essential component of pulmonary rehabilitation. Grade of Recommendation: 1B

17. Recommendation: Education should be an integral component of pulmonary rehabilitation. Education


should include information on collaborative self-management and prevention and treatment of
exacerbations. Grade of Recommendation: 1B

18. Recommendation: There is minimal evidence to support the benefits of psychosocial interventions as a
single therapeutic modality. Grade of Recommendation: 2C

Evidence-Based Guidelines in Pulmonary Rehabilitation 199


Table 2. Continued

19. Statement: Although no recommendation is provided since scientific evidence is lacking, current practice
and expert opinion support the inclusion of psychosocial interventions as a component of comprehensive
pulmonary rehabilitation programs for patients with COPD.

20. Recommendation: Supplemental oxygen should be used during rehabilitative exercise training in patients
with severe exercise-induced hypoxemia. Grade of Recommendation: 1C

21. Recommendation: Administering supplemental oxygen during high-intensity exercise programs in patients
without exercise-induced hypoxemia may improve gains in exercise endurance. Grade of Recommendation:
2C

22. Recommendation: As an adjunct to exercise training in selected patients with severe COPD, noninvasive
ventilation produces modest additional improvements in exercise performance. Grade of Recommendation:
2B

23. Statement: There is insufficient evidence to support the routine use of nutritional supplementation in
pulmonary rehabilitation of patients with COPD. No recommendation is provided.

24. Recommendations: Pulmonary rehabilitation is beneficial for some patients with chronic respiratory diseases
other than COPD. Grade of Recommendation: 1B

25. Statement: Although no recommendation is provided since scientific evidence is lacking, current practice
and expert opinion suggest that pulmonary rehabilitation for patients with chronic respiratory diseases
other than COPD should be modified to include treatment strategies specific to individual diseases and
patients in addition to treatment strategies common to both COPD and non-COPD patients.

pulmonary rehabilitation intervention, post-re- C – low or very low) as well as the balance of ben-
habilitation maintenance strategies, intensity of efits to risks and burdens (grade 1, strong recom-
aerobic exercise training, strength training, ana- mendation – certainty that benefits do, or do not,
bolic drugs, upper extremity training, inspiratory outweigh risks and burdens; grade 2, weak rec-
muscle training, education, psychological and be- ommendation – evenly balanced or uncertainty
havioral components, oxygen supplementation, regarding benefits versus risks and burdens). The
noninvasive ventilation, nutritional supplementa- recommendations developed by the Panel are pre-
tion, and rehabilitation for patients with disorders sented in table 2, along with the rating for each.
other than COPD. The new document also makes
recommendations for future research in pulmo-
nary rehabilitation. Discussion
Through a thorough and systematic review of
the published literature from 1996 to 2004, the Overall, this new guideline provides an excellent
Panel developed recommendations on rehabilita- summary of the literature published over the past
tion for patients with chronic lung disease. Based decade. The increasingly solid base of scientific
on the published evidence systematically evalu- evidence further strengthens the justification for
ated, ratings of the recommendations followed including pulmonary rehabilitation as a stan-
guidelines developed by ACCP and are indicat- dard of care for patients with chronic lung dis-
ed in table 1 [10]. These ratings evaluate both the eases. These new guidelines clearly represent a
strength of the evidence (A – high; B – moderate; major step forward in advancing the practice of

200 Ries
pulmonary rehabilitation and provide a road map • Effective pulmonary rehabilitation can
for future research and needed areas for further improve symptoms, exercise tolerance, and
development in this field. health-related quality of life and reduce
hospitalization and healthcare utilization for
patients with chronic lung diseases.
Recommendations • Pulmonary rehabilitation should be considered
for patients with chronic respiratory diseases
• Pulmonary rehabilitation should be considered other than COPD.
for any patient with symptomatic, disabling
chronic lung disease.

References
1 American Thoracic Society: Pulmonary 5 ACCP-AACVPR Pulmonary Rehabilita- 8 Ries AL, Bauldoff GS, Carlin BW, et al:
rehabilitation. Am Rev Respir Dis 1981; tion Guidelines Panel: Pulmonary reha- Pulmonary rehabilitation: joint ACCP/
124:663–666. bilitation: joint ACCP/AACVPR evi- AACVPR evidence-based clinical prac-
2 American Thoracic Society: Pulmonary dence-based guidelines. Chest 1997; tice guidelines. Chest 2007;131(suppl
rehabilitation – 1999. Am J Respir Crit 112:1363–1396. 5):4S–42S.
Care Med 1999;159:1666–1682. 6 Global Initiative for Chronic Obstructive 9 American Association of Cardiovascular
3 American Thoracic Society/European Lung Disease: Workshop Report: Global and Pulmonary Rehabilitation: Guide-
Respiratory Society: ATS/ERS statement Strategy for Diagnosis, Management, lines for Pulmonary Rehabilitation Pro-
on pulmonary rehabilitation. Am J and Prevention of COPD – Updated grams, ed 3. Champaign/IL, Human
Respir Crit Care Med 2006;173:1390– 2008 (http://goldcopd.org). Kinetics, 2004.
1413. 7 American Thoracic Society-European 10 Guyatt G, Gutterman D, Baumann MH,
4 Ries AL: Scientific basis of pulmonary Respiratory Society Task Force: Stan- et al: Grading strength of recommenda-
rehabilitation. J Cardiopulm Rehabil dards for the diagnosis and management tions and quality of evidence in clinical
1990;10:418–441. of patients with COPD (Internet). Ver- guidelines: report from an American
sion 1.2. (http://www-test.thoracic.org/ College of Chest Physicians task force.
copd). New York, American Thoracic Chest 2006;129:174–181.
Society, 2005.

Andrew L. Ries, MD, MPH


Professor of Medicine and Family and Preventive Medicine
School of Medicine, University of California, San Diego
9500 Gilman Drive–0602, La Jolla, CA 92093 (USA)
Tel. +1 858 534 4877, Fax +1 858 534 0338
E-Mail aries@ucsd.edu

Evidence-Based Guidelines in Pulmonary Rehabilitation 201


Section TitleRehabilitation and Technology
Pulmonary
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 202–204

Ward Mortality in Patients Discharged from the


ICU with Tracheostomy
Rafael Fernandez Fernandez
Hospital Sant Joan de Deu, Fundacio Althaia, Manresa, Spain

Critically ill ventilated patients commonly need Main Topics


tracheostomy as an alternative method for air-
way management. Whereas some patients need We recently published our experience in this field
tracheostomy due to anatomical reasons (e.g. [1]. Our hypothesis was that the supposed mor-
upper airway obstruction, ORL surgery) with- bidity of remaining tracheotomized in the ward
out any choice, in the majority of ICU patients, will differently affect patients depending on their
tracheostomy is an elective method restricted to clinical conditions. Accordingly, we review clini-
patients needing long-term mechanical ventila- cal charts of patients admitted in our 16-bed ICU
tion. Despite the lower invasiveness of the today between 2003 and 2006. We excluded patients
commonly percutaneous approach, tracheos- who received <24 h mechanical ventilation in the
tomy still carries some morbidity. Physicians postoperative period. Variables recorded on ad-
should therefore balance the expected benefit mission included age, sex, diagnosis, APACHE II
(greater comfort, lower work of breathing, and score, and comorbidities. During the ICU stay, we
better clearance of secretions) against the risks recorded major procedures and adverse events.
(anesthetics, surgical wound, infection, and At ICU (or step-down unit, when applicable) dis-
hemorrhage). charge, the attending physician classified the pa-
In this scenario, tracheostomy is very com- tient according to the Sabadell score, a subjective
monly performed in the ICU, ranging from 8 to tool explained in detail elsewhere [2]. Briefly, it
35% of ventilated patients depending on the case has four levels of expected prognosis: score 0 is
mix and local practices. Whether tracheostomy for patients with good prognosis, score 1 is for
has an impact on the patient’s outcome remains patients with poor prognosis in the medium to
controversial. Some authors report faster taper- long term, score 2 is for patients with poor prog-
ing of sedatives with shortening in ICU length of nosis in the short term, and score 3 is for patients
stay, whereas others suggest that such short-term who are expected to die before discharge from the
improvements only transfer morbidity and mor- hospital.
tality to wards, with null advantages on medium Patients who remained tracheotomized at
and long-term outcomes. ICU discharge were placed in dedicated rooms
in ordinary wards. The ward team responsible for Due to the lower rate of death in the ward at-
their care always included dedicated laryngolo- tributable to airway care problems, we may sug-
gists and physiotherapists, but was not informed gest that the excess in mortality risk should be due
of the Sabadell score. We reviewed the wards’ to underlying conditions and progressive deterio-
clinical charts to determine the appropriateness ration. Then, efforts to avoid tracheotomized pa-
of airway care, characteristics of secretions, time tients in the ward by extending ICU stay or ac-
to decannulation, technical complications, and celerating decannulation may be futile, or not
cause of death in patients who died. cost-effective approaches.
From 3,065 admissions, 1,502 needed mechan- The clinical impact of our study will result
ical ventilation for at least 24 h. Only 936 (62%) of from a better definition of the population most
these patients survived the ICU and were trans- likely to benefit from outreach team surveillance
ferred to the ward; 130 (13.9%) patients had a tra- in the ward or from a prolonged stay in the step-
cheostomy cannula in place when transferred to down unit until decannulation, when feasible.
the ward. The overall ward mortality was 87/936 We conclude that lack of tracheostomy de-
(9.3%) and was higher in patients with a tracheo- cannulation in the ICU appears to be associated
stomy cannula in place at ICU discharge than in with ward mortality, but only in the group with
those without (34/130 (26%) vs. 53/806 (7%), p < a Sabadell score of 1. Whether discharge without
0.001). Patients that remained cannulated at dis- decannulation is a direct risk factor or a marker of
charge were sicker and had more complications sicker patients remains to be elucidated.
(blood transfusion, renal replacement therapy,
ICU infection, pneumothorax, acute renal failure,
upper gastrointestinal bleeding, and reintubation). Recommendations
A multivariate analysis found three factors associ-
ated with ward mortality: age, tracheostomy, and • Tracheostomy in the ICU remains common
Sabadell score, with a global accuracy of 93.1% for despite recent improvements in weaning
predicting ward mortality. We found that the mor- techniques and protocols.
tality increase associated with tracheostomy was • Despite optimal ward treatment, patients with
restricted to patients with intermediate Sabadell tracheostomy cannula in the ward are at higher
scores, whereas minimal or no difference was risk for death.
found between patients with tracheostomy can- • This additional risk is only important in
nulae and those without in the ‘good prognosis’ patients with medium and long-term bad
and ‘expected to die in hospital’ groups. prognosis, whereas patients with good progno-
sis were not affected.
• Whether a longer ICU stay for allowing safer
Discussion decannulation may improve outcome remains
an unanswered question.
Our study offers conclusions that should be care-
fully framed. First, treatment in the ward in our
center was optimal, and our results cannot be ex-
trapolated to any other institution without taking
into account this very sensitive aspect. Every hos-
pital should evaluate their outcome for tracheot-
omized patients in the ward before adopting any
new strategy.

Ward Mortality in Patients Discharged from the ICU with Tracheostomy 203
References
1 Fernandez R, Bacelar N, Hernandez G, 2 Fernandez R, Baigorri F, Navarro G,
Tubau I, Baigorri F, Gili G, Artigas A: Artigas A: A modified McCabe score for
Ward mortality in patients discharged stratification of patients after ICU dis-
from the ICU with tracheostomy may charge: the Sabadell score. Crit Care
depend on patient’s vulnerability. Inten- 2006;10:R179.
sive Care Med 2008;34:1878–1882.

Dr. Rafael Fernandez Fernandez


Hospital Sant Joan de Deu, Fundacio Althaia, Manresa
ES–08243 Manresa (Spain)
Tel. +34 938742112, Fax +34 938736204
E-Mail rfernandezf@althaia.cat

204 Fernandez
Pulmonary Medicine in Pediatrics and Neonatology Critical Care
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 205–209

Exogenous Surfactant in Respiratory Distress


Syndrome
Andrea Calkovskaa ⭈ Egbert Hertingb
a
Department of Physiology, Jessenius Faculty of Medicine, Comenius University and Martin Faculty Hospital, Martin, Slovakia, and
b
Department of Pediatrics, University of Lübeck, Lübeck, Germany

Abbreviations composed of several phospholipids, neutral lipids


ARDS Acute respiratory distress syndrome
and surfactant-specific proteins. The largest pro-
LA Large surfactant aggregates portion of phospholipids is phosphatidylcholine,
RDS Respiratory distress syndrome of which dipalmitoylphosphatidylcholine is the
rSP-C Recombinant SP-C major surface-active component. Four native SPs
SA Small surfactant aggregates have been identified: hydrophobic SP-B and SP-C
SP Surfactant protein are thought to have a role in the surface tension-
SP-A–D Surfactant proteins A–D lowering properties of pulmonary surfactant, and
SP-A and SP-D are hydrophilic and play a role in
surfactant metabolism and pulmonary host de-
Surfactant replacement therapy is an established fense [1].
part of routine clinical management of neonates
with RDS and this treatment may also be effective
in other forms of lung diseases including meco- Changes of Pulmonary Surfactant in ARDS
nium aspiration syndrome, neonatal pneumonia
and the ‘adult’ form of ARDS. In the present chap- RDS is the most common respiratory disorder of
ter we discuss the factors influencing the efficacy premature babies. Typically, idiopathic RDS affects
of exogenous surfactant therapy in the ARDS. infants below 35 weeks of gestational age and the
key element in the pathophysiology is the struc-
tural and functional immaturity of the lung with a
Pulmonary Surfactant primary deficiency of the endogenous surfactant
system. In contrast, the ‘adult’ form of ARDS is
Pulmonary surfactant is a lipoprotein complex characterized by secondary abnormalities of the
that covers the inner surface of the lungs. It pre- surfactant system. Several possible mechanisms
vents collapse of the alveoli and small airways responsible for surfactant changes in ARDS have
during expiration by reducing surface tension at been suggested [2]. The most important mecha-
the air-liquid interface and allows efficient gas ex- nism in the pathogenesis of ARDS seems to be al-
change at low transpulmonary pressures. It is syn- veolar flooding by plasma proteins which occurs
thesized in the type II alveolar epithelial cells and early in the course of the disease. Plasma proteins
such as albumin, fibrinogen and fibrin monomer of pulmonary surfactant in maintaining normal
inhibit surfactant adsorption, prevent the estab- lung function and the observation that alterations
lishment of low alveolar surface tension during in the endogenous surfactant system contribute to
surface compression and interfere with the sur- the pathophysiology of ARDS, provide a rationale
factant-related parameters of lung function. for treatment of ARDS patients with exogenous
As patients with ARDS exhibit an intense pul- surfactant. The effects of surfactant therapy are
monary inflammatory reaction, a variety of medi- however influenced by several factors.
ators may directly or indirectly interfere with sur-
factant function. Increased phospholipolytic and Timing of Surfactant Administration
proteolytic activities with a consequent decrease Both experimental and clinical studies demon-
in surfactant activity were demonstrated in the al- strated the superior physiological and clinical ef-
veolar compartment as a result of the inflamma- fects of early administration of surfactant (‘pro-
tory process. phylaxis’) in comparison to replacement at later
The change in the ratio between LA and SA is stages of the disease (‘rescue’). In contrast to neo-
another typical finding. Under physiological con- natal RDS, in the ‘adult’ form of ARDS there is no
ditions, up to 90% of the surfactant material in consensus on which patient would benefit most
bronchoalveolar lavage fluid is recovered in the from early surfactant therapy since there is a lack
fraction of LA with high surface activity and a of reliable and sensitive markers of lung injury.
relatively high SP-B content. The amount of the Theoretically, administration of surfactant earli-
functionally inferior SA fraction, which is consid- er in the course of ARDS may help to modulate
ered to be a degradation product of surfactant, is pulmonary inflammation before severe lung dys-
increased in bronchoalveolar lavage fluid in pa- function occurs.
tients with ARDS. Alterations in endogenous pul-
monary surfactant further include the decrease Mode of the Surfactant Administration
in total phospholipid content, changes in relative Commonly, exogenous surfactant is instilled as
distribution of the phospholipid classes, increased a bolus into the central airways. With this meth-
levels and altered neutral lipid profile, decreased od, large doses of the surface-active material can
levels of surfactant-specific proteins and inactiva- be given in a relatively short time and a rapid re-
tion of endogenous surfactant by incorporation sponse to the treatment is expected. Disadvantages
into hyaline membranes. of this technique include airway obstruction by
the liquid bolus and non-uniform distribution.
In comparison to the bolus administration, aero-
Surfactant Replacement Therapy solization of exogenous surfactant requires small-
er amounts of material and may facilitate a more
Taken together, beside neonatal RDS, surfactant uniform distribution. However, surfactant will
replacement therapy can also be helpful in oth- mainly be deposited in ventilated parts of the
er forms of lung disease, in which endogenous lungs. On the other hand, the loss of surfactant
surfactant is inactivated by aspirated material in the delivery system is high, and the response
or leakage of plasma proteins into the airspaces. may need several hours or even days. The prom-
Although the mortality associated with the ARDS ising technique of lung lavage may help to re-
in adults has decreased significantly in the past, move aspirated material, plasma inhibitors and/
it is still over 30%. Treatment of these patients is or inflammatory mediators from the lungs and at
mainly supportive and interventions improving the same time replace inactivated surfactant and
outcome are of great interest. The importance stabilize the lungs. Moreover, surfactant applied

206 Calkovska ⭈ Herting


by lavage technique spreads more homogenous- surfactant dose currently recommended ranges
ly throughout the lungs, and a higher volume of from 50 to 200 mg/kg b.w. In premature babies
diluted surfactant washes out the airspaces more with RDS this dose should be close to the esti-
effectively. Lavage methods of surfactant admin- mated pool size of alveolar surfactant in a normal
istration were shown to be effective in some res- full-term newborn, about 100 mg/kg b.w. In adult
piratory disorders like meconium aspiration syn- patients with ARDS the optimal dose is not yet
drome and may become an alternative in ARDS known. However, large quantities of exogenous
patients in the future. Bronchoscopic application of surfactant are needed to overcome the inhibitory
surfactant with or without lavage in patients with effects of plasma proteins in the airspaces and to
severe ARDS was also recommended as both fea- optimize surfactant distribution. In individual pa-
sible and safe resulting in an improvement of gas tients with ARDS, cumulative doses up to 800 mg/
exchange. kg b.w. have been applied. Based on clinical expe-
rience, it seems that more than 2–4 repeated doses
Ventilation Patterns of instilled surfactant have no additional benefit.
The goal of ventilatory management during the
early stages of RDS is to maintain adequate oxy- Type of Surfactant Preparation
genation and ventilation, while minimizing ven- The clinical response depends on the quality of
tilator-induced lung injury. The mode of venti- the exogenous surfactant preparation. Modified
lation preceding the surfactant administration natural surfactants containing the native hydro-
influences the physiological effects of exogenous phobic peptides SP-B and SP-C are more effective
surfactant as the surfactant-deficient lungs are than protein-free synthetic surfactants. However,
very susceptible to epithelial disruption due to surfactant preparations derived from animal lungs
e.g. large tidal volumes. In patients with ARDS the are expensive with a limited supply and they pose a
ventilation with low tidal volumes and sufficient- potential danger of viral or prion disease or allergic
ly high levels of positive end-expiratory pressure reactions. Thus, there is a need for synthetic sur-
after reopening of the collapsed alveoli will both factant substitutes, which can be produced in large
protect the lungs and reduce mortality. Moreover, quantities at (hopefully) reasonable costs [3].
an appropriate positive end-expiratory pressure is If the alveoli are flooded with protein-rich ede-
required for optimal function of some synthetic ma fluid (e.g. in ARDS), the inhibitory effects of
surfactant preparations. plasma proteins can only be counterbalanced by
relatively large amounts of surfactant and/or by
Dosing administration of surfactant preparations resis-
The individual dose of the surfactant depends on tant to plasma protein inhibition. Therefore, pre-
the disorder being treated, for example on wheth- venting surfactant inactivation is an important
er the exogenous material is given to compensate new approach that could increase the therapeutic
for primary surfactant deficiency in premature effects of exogenous surfactant in various forms
newborns, or administered to overcome the in- of lung disease. Sensitivity to inactivation varies
hibitory effects of aspirated meconium or plasma between different surfactant preparations and is
proteins leaking into the airspaces in the patients in part related to their protein content. Complete
with ARDS [1]. The initial dose, required for clin- natural surfactant containing all specific proteins
ical response and eventually the need for retreat- is much more resistant to inactivation than the
ment, depends on the quality of the surfactant modified natural surfactants isolated from mam-
preparation, the severity of the disease and the malian lungs. The latter contain only lipids and
clearance rate of exogenous surfactant. The initial small amounts of the hydrophobic SP-B and SP-C

Exogenous Surfactant in Respiratory Distress Syndrome 207


because the hydrophilic SP-A and SP-D are re- trials. Firstly, most animal studies have report-
moved by extraction procedures. The resistance ed on respiratory failure caused by direct injury
of commercially available modified natural sur- whereas clinical ARDS is caused more often by
factants to inactivation by plasma proteins or indirect injury. Secondly, in animals, surfactant is
meconium can be enhanced by enriching the ma- usually administered in optimal timing whereas
terial with SP-A and by addition of polymers such in patients this treatment tends to be late. Finally,
as dextran, polyethylene glycol, hyaluroran, or by most of the animal studies are terminated with-
polymyxin B. in several hours and thus, they cannot evaluate
Current research is focused on the develop- long-term effects of surfactant therapy in ARDS.
ment of a new generation of artificial surfactant Moreover, there are many differences among the
substitutes based on synthetic analogues of SP-B various clinical trials. Different types of surfactant
and/or SP-C [3]. These SP analogues have been and different delivery methods were used that may
produced by peptide synthesis or recombinant have resulted in varying concentrations of surfac-
technology to provide a new class of synthetic tant reaching the alveoli and altering the effective-
surfactants that may be a suitable alternative to ness of therapy. Other differences include differ-
animal-derived surfactants. Up to this time, two ent ventilation strategies, the timing of surfactant
synthetic preparations have been used in clinical administration as well as the cause of ARDS (pul-
trials, rSP-C surfactant (Venticute®) and KL4 sur- monary vs. extrapulmonary).
factant (Surfaxin®) with the peptide KL4 designed In a meta-analysis on surfactant treatment of
to mimic the function of SP-B. adults with ARDS published in 2006, the authors
identified 251 articles between 1966 and 2005 and
Surfactant Metabolism five studies met the inclusion criteria [4]. Only
The effects of therapy also depend on how the ex- studies that were randomized controlled clinical
ogenous surfactant is metabolized in the treated trials, that compared the use of exogenous sur-
lungs. The underlying lung injury (uniform vs. factant to an appropriate control group receiv-
non-uniform) which may influence the distribu- ing standard therapy and that evaluated mortality
tion of exogenous material and the speed of LA and/or pulmonary physiological parameters were
to SA conversion belong to the most important included in the analysis. It was concluded that
factors. If the damage of the alveolar epithelium exogenous surfactant may improve oxygenation
is not too extensive, an exogenous surfactant may but does not reduce mortality, and thus that ex-
become activated (‘recycled’) by entering the in- ogenous pulmonary surfactant cannot currently
tra-alveolar metabolism, combining with endog- be considered an effective adjunctive therapy in
enous surfactant and may be newly secreted into ARDS patients. The lack of effectiveness of exoge-
the alveolar space. Thus, the utilization of exoge- nous surfactant is explained firstly by the fact that
nous surfactant depends on interactions between the patients with ARDS usually die of multiorgan
the material delivered into the lungs and the host system failure rather than from respiratory fail-
alveolar environment. ure and thus the treatment of pulmonary disor-
ders may not affect overall mortality. In addition,
Perspectives of Surfactant Treatment an optimal regimen for surfactant treatment has
Taken together, the treatment of ARDS with ex- not yet been identified [4].
ogenous surfactant has a strong rationale, which It becomes evident that the success of surfac-
is firmly founded on experimental data. However, tant replacement therapy in ARDS depends on
stimulating results obtained in animal experi- the appropriate selection of the indication for this
ments were not always reproducible in clinical treatment. By pooled analysis of five multicenter

208 Calkovska ⭈ Herting


studies in which patients with ARDS treated with aspiration syndrome, pneumonia). Based on the
rSP-C surfactant, it was shown that rSP-C surfac- current knowledge, adults with ARDS are also a
tant improved oxygenation in patients with ARDS possible target group for surfactant replacement.
irrespective of the underlying disease. Reduced However, large doses of surfactant are necessary
mortality in association with surfactant treat- to overcome the inhibitory effects of plasma pro-
ment was only obtained in patients with direct teins leaking into the airspaces. As a consequence,
lung insult such as pneumonia or aspiration [5]. there is an urgent need to develop a new genera-
However, a randomized controlled study apply- tion of surfactant preparations that are more re-
ing rSP-C surfactant relatively early in the course sistant to inhibitors and can be produced in large
of ARDS in consequence of pneumonia or aspi- quantities at reasonable costs. At the same time, we
ration of gastric contents (VALID study) was re- need to find out the safe, effective and hopefully
cently stopped due to lack of effectiveness. non-invasive ways for surfactant administration.

Conclusion Acknowledgements

This work was supported by the National Scientific Grant


Surfactant replacement therapy is now an estab-
Agency (VEGA) (Project No. 1/0061/08), project ‘Center
lished part of routine clinical management of of Experimental and Clinical Respirology’ co-financed by
neonatal RDS and the indications are widening EC sources and a grant of the German Research Council
to other forms of neonatal diseases (meconium (DFG He 2072-2/2).

References
The number of references has been limited to five. A complete list is available from the authors on request.

1 Robertson B, Halliday H: Principles of 3 Curstedt T, Johansson J: New synthetic 5 Taut FJ, Rippin G, Schenk P, et al: A
surfactant replacement. Biochim Bio- surfactant – how and when? Biol Neo- search for subgroups of patients with
phys Acta 1998;1408:346–361. nate 2006;89:336–339. ARDS who may benefit from surfactant
2 Guenther A, Walmrath D, Grimminger 4 Davidson WJ, Dorscheid D, Spragg R, et replacement therapy: a pooled analysis
F, et al: Surfactant metabolism and al: Exogenous pulmonary surfactant for of five studies with recombinant surfac-
replacement in acute respiratory distress the treatment of adult patients with tant protein-C surfactant (Venticute®).
syndrome. Eur Respir Mon acute respiratory distress syndrome: Chest 2008;134:724–732.
2002;20:119–128. results of a meta-analysis. Criti Care
2006;10:R41.

Prof. Andrea Calkovska, MD, PhD


Department of Physiology, Jessenius Faculty of Medicine, Comenius University
Mala Hora 4, SK–037 54 Martin (Slovakia)
Tel./Fax +421 43 4131426
E-Mail calkovska@jfmed.uniba.sk

Exogenous Surfactant in Respiratory Distress Syndrome 209


Section TitleMedicine in Pediatrics and Neonatology Critical Care
Pulmonary
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 210–216

Hypercapnia and Hypocapnia in Neonates


Li Tao ⭈ Wei Zhou
Department of Neonatology, Guangzhou Children’s Hospital, Guangzhou Women and Children’s Medical Center,
Guangzhou, China

Abbreviations Influence of Hypocapnia on the Central


CBF Cerebral blood flow
Nervous System
IVH Intraventricular hemorrhage
PaCO2 Partial pressure of carbon dioxide In the last century it was reported that hypocap-
PAH Pulmonary artery hypertension nia induced by hyperventilation did not deflate
PbrO2 Brain tissue oxygen tension the infarcted area after middle cerebral artery
PVL Periventricular leukomalacia occlusion or increase CBF of the ischemic re-
VLBW Very low birth weight gion, but increased the size of the ischemic area.
VRLI Ventilator-related lung injury Subsequent trials demonstrated that hypocap-
nia could aggravate the decrease in energy-rich
phosphate, impair ischemic brain metabolism,
Arterial PaCO2 reflects the balance between and worsen cerebral ischemia [2]. The decrease
CO2 production and consumption. PaCO2 rang- in CBF reduces oxygen transport, raises ner-
es from 4.7 to 6.0 kPa (35–45 mm Hg) in nor- vous excitation, and limits cerebral metabolism,
mal neonates [1]. In newborn infants, hypercap- which further limits oxygen transport. The re-
nia caused by elevation of PaCO2 can have an sults of Ohyu et al. [3] suggested that hypocap-
adverse effect on the internal environment and nia under hypotension might cause neuronal cell
function; hypocapnia (also known as hypocarbia) death in the hippocampus of the neonatal rabbit.
can increase the incidence of brain injury. In re- Not only ischemia but also metabolic changes
cent years, researchers have focused on the per- induced by hypocapnia might contribute to this
missive hypercapnia ventilation strategy in neo- apoptotic neuronal cell damage. Alkali poison-
nates with respiratory disease and brain injury. ing of cerebrospinal fluid is the main adverse ef-
In this article, we review the latest progress in fect of hypocapnia on the nervous system. As
neonatal hypocapnia, hypercapnia and permis- an important component of the HCO3–/H2CO3
sive hypercapnia strategy, and discuss their clini- buffer system of extracellular fluid, CO2 chang-
cal implications. es initially alter pH and initiate cerebral ves-
sel contraction and reduce CBF through NO,
prostanoid, potassium channels, and intracel- associated with hypocapnia (PaCO2 <25 mm Hg).
lular Ca2+. It has been reported [4] that during Collins et al. [14] conducted a prospective cohort
end-tidal CO2 changes, PbrO2 is linearly corre- study and enrolled 1,105 newborns with a birth
lated with intracranial blood flow, and PbrO2 weight of 500–2,000 g. 657 two-year-old survi-
is correlated with end-tidal CO2 (PbrO2 >60 or vors had both neurodevelopmental assessments
<20 mm Hg). In addition, it was found that cere- and blood-gas data taken the first week after
bral hypoxic events can be reduced significantly birth. Disabling cerebral palsy was subsequently
by increasing cerebral perfusion pressure as re- diagnosed in 2.3% of the 257 unventilated new-
quired [5]. borns, 9.4% of the 320 ventilated newborns with-
Hypocapnia is a potential risk factor for PVL, out exposure to unusual levels of hypocapnia, and
cerebral palsy, cognition developmental disor- 27.5% of the 80 ventilated infants with exposure
ders, and auditory deficits [6, 7]. The studies of to significant hypocapnia. The researchers rec-
Erickson et al. [8] show that the risk of severe IVH/ ommend that neonatologists avoid arterial PCO2
PVL was significantly increased when PaCO2 was levels <35 mm Hg and arterial PO2 levels >60 mm
<30 mm Hg within 48 h after birth. There was Hg whenever possible in ventilated LBW infants.
also an association between duration of hypocap- There is extensive, albeit retrospective, evidence
nia and the risk of severe IVH/PVL. Fujimoto et that hypocapnia in premature infants is associat-
al. [9] retrospectively visited 167 survivors of ed with poor neurodevelopmental outcome, such
VLBW newborns <35 weeks’ gestational age. The as PVL, IVH, and cerebral palsy [14–16], possibly
results showed that the cystic PVL incidence in due to cerebral vasoconstriction, decreased CBF
infants with and without hypocapnia was 23 and and cerebral oxygen delivery. Therefore, preven-
5%, respectively. Seven years later, Okumura’s re- tion of hypocapnia must also be a primary objec-
search gained similar results as Fujimoto et al. tive in the management of these infants.
[10]. Hypocapnia within the first 2 h after birth
increased the risk of brain damage in severely as-
phyxiated newborns [11]. In an observational co- Effect of Hypocapnia on the Respiratory
hort study [12] performed in full-term asphyxia System
neonates with hypoxic ischemic encephalopathy
born from 1985 to 1995, mortality and incidence Neonatal respiratory failure was associated with
of severe cerebral palsy, blindness, deafness and PAH, right-left shunt, and severe hypoxemia.
hypoevolutism were found to be significant- After hyperventilation to maintain PaCO2 levels
ly increased in infants with severe hypocapnia. at 25–30 mm Hg, the pulmonary arterial pres-
In addition, 34 newborns with severe persistent sure of PAH newborns was significantly de-
pulmonary hypertension were treated by hyper- creased, and the right-left shunt was reversed, in-
ventilation or maintenance of PaCO2 >50 mm dicating that hyperventilation could lessen PAH
Hg [13]. The results in a group that received hy- and improve circulation oxygenation. Based on
perventilation showed that physical development the above theory, a hypocapnia strategy is rec-
delay and severe neurologic abnormalities were ommended to treat neonatal persistent PAH
highly correlated with the duration of hyperven- and congenital diaphragmatic hernia. However,
tilation, especially the incidence of sensorineural another experimental study suggests that hy-
hearing loss was the highest. However, none had pocapnia can deteriorate bronchial spasm, air-
sensorineural hearing loss in the group treated way pressure, microvascular permeability and
with maintenance of PaCO2 >50 mm Hg. These lung compliance, worsening the acute lung in-
data indicated that sensorineural hearing loss is jury following ischemia-reperfusion. Moreover,

Hypercapnia and Hypocapnia in Neonates 211


the degree of lung injury was proportional to the proteins in the cerebral cortex of newborn pig-
degree of hypocapnic alkalosis [17]. lets; therefore, it may be deleterious to the new-
born brain [21]. Hypercapnia in VLBW infants
during the first 3 postnatal days is associated
Hypercapnia with severe IVH [22]. In addition, a retrospec-
tive search analyzed blood gas data in the first 4
The influence of hypercapnia on neonates de- postnatal days for 849 infants weighing from 401
pends on the degree and speed of PaCO2 eleva- to 1,250 g and suggested that both extremes of
tion, hypoxia or ischemia condition, and intrac- arterial PaCO2 and the magnitude of fluctuations
ranial disease or other complications. Generally, in arterial PaCO2 are associated with severe IVH
the body has good compensation for and tol- in preterm infants, so it may be prudent to avoid
erance of acute hypercapnia or slowly elevat- extreme hypocapnia and hypercapnia during the
ed PaCO2. Many studies have suggested that risk period of IVH [23]. Severe hypercapnia can
acute hypercapnia with PaCO2 <10.67 kPa (80 inhibit the myocardial contractile force, expand
mm Hg) and pH >7.15 can hardly damage the vessels leading to blood pressure decrease, and
body; when saturation of blood oxygen is nor- stimulate gangliated nerve excitation, resulting
mal, PaCO2 slowly elevates to 10.0–14.67 kPa in arrhythmia. Hypercapnia complicates hypoxia
(75–110 mm Hg) and does not cause any notice- and can induce pulmonary vasoconstriction and
able clinical symptoms [2], and even has some increase pulmonary circulation resistance, lead-
benefits, such as increased gangliated nerve ex- ing to PAH, cardiac ejection fraction reduction,
citation, more catecholamine and improved cir- and worsening of cerebral functional lesions [24,
culation from vasodilation induced by hypercap- 25]. Acute hypercapnia can reduce skeletal mus-
nia. Mild intracellular acidosis can even protect cle contraction, especially diaphragmatic muscle
hypoxic cells [18]. However, PaCO2 >110 mm contraction, and exhaust respiratory muscles. By
Hg can lead to organ dysfunction. Hypoxia/isch- contrast, chronic hypercapnia contributes to an
emia and hypercapnia have an additive effect on increase in adrenal medulla secretion, adreno-
body injury. Neonate and preterm cerebral ves- corticotrophic hormone, aldosterone and antidi-
sels are more sensitive to PaCO2 elevation (ves- uretic hormone production.
sel expanding) than to PaCO2 decrease (vessel
contraction). Rapidly increased PaCO2 to 12.0–
14.67 kPa (90–110 mm Hg) can induce central Permissive Hypercapnia
nervous palsy (consciousness alterations, cata-
phora) and hyperspasmia [19], worsen blood- Traditional ventilation strategies cause adverse
brain barrier permeability, cerebral interstitial effects on newborn infants, such as barotraumas,
edema, CBF and CBF’s autoregulation, and fi- lung interstitial emphysema, and systemic gas em-
nally cause intracranial hypertension and even bolism. In our current understanding of the pa-
intracraninal hemorrhage. If complicated with thology and physiology of the respiratory system,
ischemia, intracellular acidosis from hypercap- permissive hypercapnia is used in clinic as a novel
nia can stimulate cell metabolism and oxygen ventilatory strategy. The term permissive hyper-
free radical production, and worsen brain dam- capnia defines a ventilatory strategy for acute re-
age, especially in reperfusion injury [18, 20]. spiratory failure, in which the lungs are ventilated
Hypercapnia alters neuronal energy metabolism, with a low inspiratory volume and pressure [26].
increases phosphorylation of transcription fac- The aim of permissive hypercapnia is to minimize
tors, and increases the expression of apoptotic lung damage during mechanical ventilation. The

212 Tao ⭈ Zhou


small tidal volume ventilation reduces peak in- infants. Permissive hypercapnia may reduce VRLI
spiratory pressure and mean airway pressure, and and bronchopulmonary dysplasia [31]. However,
minimizes the incidence of air leaks and the effect a meta-analysis from two trials involving 269
of mechanical ventilation on returned blood vol- newborns shows permissive hypercapnia can-
ume and cardiac output. not contribute to reduce the incidence of death
or chronic lung disease at 36 weeks, IVH grade
3 or 4, or PVL [32]. At present, the best ventila-
Permissive Hypercapnia and Disease of the tion strategy and the precision impact of permis-
Respiratory System sive hypercapnia on this strategy is not very clear,
however it is important to understand hypercap-
It was reported by Dariol and Perret [27] in 1984 nia’s mechanism which is helpful for determining
that controlled mechanical ventilation was per- the safety and therapeutic utility of hypercapnia
formed to maintain PaCO2 at 90 mm Hg to treat in protective lung ventilatory strategies [25]. An
severe acute asthma and the effect was favorable. animal experiment found that the pH and heart
Subsequent studies also suggest that, in the treat- rate of neonatal rabbits decreased and mean
ment of some respiratory tract diseases, such as blood pressure increased progressively as CO2
acute respiratory distress syndrome, CO2 eleva- was increased [33]. When PaCO2 was increased
tion to a certain extent can decrease complica- from 20 to 80 mm Hg, vessel diameter, blood flow
tions and improve survival rate. Some research- velocity, and blood flow rate increased marked-
ers treated newborns with persistent pulmonary ly. Cardiac output increased slightly. When CO2
hypertension or congenital diaphragmatic hernia exceeded 100 mm Hg, all of these variables de-
by elevating PaCO2 to 60 mm Hg, and found that creased. When PaCO2 exceeded 150 mm Hg, all
the newborns were tolerant of the treatment, with variables were significantly lower than the control
no adverse effects [28]. Recent studies have dem- values. Intravital microscopic visualization of rab-
onstrated that CO2 elevation can to a certain ex- bit ear microcirculation showed that 150 mm Hg
tent protect lung from ischemia/reperfusion in- is the permissive upper limit of acute hypercap-
jury [28]. Trials of animals with acute lung injury nia with respect to maintenance of the peripheral
showed that the lung interstitial edema and cardiac microcirculation.
load in the hypercapnic group were significantly
relieved compared with a high ventilation group,
indicating that hypercapnia could improve acute Permissive Hypercapnia and Central Nervous
pulmonary hemorrhage [29]. By the observation System Disease
of lung mechanical, blood gas and pathological
changes, it found that hypercapnia improved the Permissive hypercapnia can protect against ven-
lung mechanics index and dynamic compliance, tilation-induced brain injury. Hypoxemia associ-
decreased PIP level and protein content in bron- ated with acute hypercapnia (PaCO2 68 mm Hg)
choalveolar lavage fluid which reflex lung perme- can reduce 30% of cerebral oxygen consump-
ability [30]. All of the above-mentioned results tion, and induce synthesis of neuronal nitric ox-
suggest that permissive hypercapnia is of help in ide synthase-derived nitric oxide which increases
respiratory disease. the CBF and vasodilation response [34]. At mod-
Traditional mechanical ventilation often erate hypercapnia, CBF increases 6% as soon as
causes VRLI which prolongs the disease course PaCO2 increases 1 mm Hg [35], and such a re-
and is necessary for oxygen. The VRLI’s incidence sponse quickly reached a peak within 5–15 min.
is 13–35% in VLBW and ultra low birth weight Monitoring the mean CBF velocity, PaCO2, and

Hypercapnia and Hypocapnia in Neonates 213


mean arterial blood pressure of 43 ventilated who received a permissive hypercapnia strategy
VLBW infants during the postnatal first week were not more likely to have IVH than those with
showed that when PaCO2 was >45 mm Hg, mean normocapnia. There were no differences in any of
CBF velocity increased with increasing PaCO2, the behavioral or functional scores among chil-
suggesting that the progressive loss of cerebral au- dren according to the respiratory strategy. There
toregulation and impaired autoregulation during was a significant interaction between care strat-
this period may be associated with increased vul- egy and the 1-min Apgar score, indicating that
nerability to brain injury [20]. Collins et al. [36] infants with lower Apgar scores may be at higher
carried out a large-scale follow-up survey of 1,105 risk for IVH with permissive hypercapnia. On the
newborns who weighed 500–2,000 g. 777 of 902 other hand, inadvertent hypercapnia may result
cases received mechanical ventilation with assess- from this method, and hypercapnia has been as-
ment of neurological function for 2 years. The sociated with IVH [39–41]. In fact, in the initial
results show the incidence of cerebral palsy was pilot trial of permissive hypercapnia in premature
9.4% in cases treated with a permissive hyper- infants, many infants in the hypercapnic group
capnia strategy and 27.5% in those treated with had maximum PaCO2 >55 mm Hg, perhaps out
traditional ventilation characterized by hypocap- of the traditional ‘safe’ range of hypercapnia [42,
nia. These results indicate that permissive hyper- 43].
capnia in the neonatal period played a protective
role in the treatment of brain injury. By targeting
mild to moderate hypercapnia during ventilation Conclusion
of premature infants, it has been suggested that
permissive hypercapnia may be neuroprotective Permissive hypercapnia as a novel protective ven-
by avoidance of accidental hypocapnia [37], and tilation therapy may to a certain extent improve
it should be noted that the pursuit of permissive the survival rate of neonates with brain injury or
hypercapnia should not be at the expense of de- respiratory tract diseases. However, a large-sam-
creased lung expansion, i.e. an adequate PEEP is ple study needs to be conducted to assess its clini-
essential. Recently, Hagen et al. [38] studied the cal application. During permissive hypercapnia
effect of permissive hypercapnia on brain injury treatment, how to select the appropriate ventilato-
and developmental impairment. There were two ry index, whether or not permissive hypercapnia
groups: permissive hypercapnia in which PCO2 can worsen cerebral edema, and the potential risk
values fluctuated from 45 to 55 mm Hg, and nor- of intracranial hypertension, still need further ex-
mocapnia in which PCO2 values fluctuated from ploration and study.
35 to 45 mm Hg. The analysis shows that Infants

References
1 Brouillette RT, Waxman DH: Evaluation 3 Ohyu J, Endo A, Itoh M, Takashima S: 5 Meixensberger J, Jaeger M, Väth A, et al:
of the newborn’s blood gas status. Clin Hypocapnia under hypotension induces Brain tissue oxygen-guided treatment
Chem 1997;43:215–221. apoptotic neuronal cell death in the hip- supplementing ICP/CPP therapy after
2 Laffey JG, Kavanagh BP: Hypocapnia. N pocampus of newborn rabbits. Pediatr traumatic brain injury. J Neurol Neuro-
Engl J Med 2002;347:43–53. Res 2000;48:24–29. surg Psychiatry 2003;74:760–764.
4 Hemphill JC 3rd, Bleck T, Carhuapoma
JR, et al: Is neurointensive care really
optional for comprehensive stroke care?
Stroke 2005;36:2344–2345.

214 Tao ⭈ Zhou


6 Murase M, Ishida A: Early hypocarbia of 17 Laffey JG, Engelberts D, Kavanagh BP: 29 Laffey JG, Tanaka M, Engelberts D, Luo
preterm infants: its relationship to Injurious effects of hypocapnic alkalosis X, Yuan S, Tanswell AK, et al: Therapeu-
periventricular leukomalacia and cere- in the isolated lung. Am J Respir Crit tic hypercapnia reduces pulmonary and
bral palsy, and its perinatal risk factors. Care Med 2000;162:399–405. systemic injury following in vivo lung
Acta Paediatr 2005;94:85–91. 18 Fritz KI, Delivoria-Papadopoulos M: reperfusion. Am J Respir Crit Care Med
7 Ambalavanan N, Carlo WA: Hypocapnia Mechanisms of injury to the newborn 2000;162:2287–2294.
and hypercapnia in respiratory manage- brain. Clin Perinatol 2006;33:573–591. 30 Yang LL, Ji XP, Liu Z: Effects of hyper-
ment of newborn infants. Clin Perinatol 19 Wang X-L: Neonatal respiratory failure; capnia on nuclear factor-κB and tumor
2001;28:517–531. in Zhou X-G, Xiao X, Nong S-H (eds): necrosis factor-α in acute lung injury
8 Erickson SJ, Grauaug A, Gurrin L, et al: Therapeutics of Mechanical Ventilation models. Chin Med J (Engl) 2004;117:
Hypocarbia in the ventilated preterm in Neonates (in Chinese). Beijing, Peo- 1859–1861.
infant and its effect on intraventricular ple’s Medical Publishing House, 2004, pp 31 Ambalavanan N, Carlo WA: Ventilatory
haemorrhage and bronchopulmonary 75–89. strategies in the prevention and manage-
dysplasia. J Paediatr Child Health 2002; 20 Kaiser JR, Gauss CH, Williams DK: The ment of bronchopulmonary dysplasia.
38:560–562. effects of hypercapnia on cerebral auto- Semin Perinatol 2006;30:192–199.
9 Fujimoto S, Togari H, Yamaguchi N, et regulation in ventilated very low birth 32 Woodgate PG, Davies MW: Permissive
al: Hypocarbia and cystic periventricular weight infants. Pediatr Res 2005;58:931– hypercapnia for the prevention of mor-
leukomalacia in preterm infants. Arch 935 bidity and mortality in mechanically
Dis Child 1994;71:F107–F110. 21 Fritz KI, Zubrow A, Mishra OP, Delivo- ventilated newborn. Cochrane Database
10 Okumura A, Hayakawa F, Kato T, et al: ria-Papadopoulos M: Hypercapnia- Syst Rev 2001;CD002061.
Hypocarbia in preterm infants with induced modifications of neuronal func- 33 Komori M, Takada K, Tomizawa Y, et al:
periventricular leukomalacia: the rela- tion in the cerebral cortex of newborn Permissive range of hypercapnia for
tion between hypocarbia and mechani- piglets. Pediatr Res 2005;57:299–304. improved peripheral microcirculation
cal ventilation. Pediatrics 2001;107: 22 Kaiser JR, Gauss CH, Pont MM, Wil- and cardiac output in rabbits. Crit Care
469–475. liams DK: Hypercapnia during the first 3 Med 2007;35:2171–2175.
11 Klinger G, Beyene J, Shah P, Perlman M: days of life is associated with severe 34 Okamoto H, Hudetz AG, Roman RJ, Bos-
Do hyperoxaemia and hypocapnia add intraventricular hemorrhage in very low njak ZJ, Kampine JP: Neuronal NOS-
to the risk of brain injury after intrapar- birth weight infants. J Perinatol 2006;26: derived NO plays permissive role in
tum asphyxia? Arch Dis Child Fetal Neo- 279–285. cerebral blood flow response to hyper-
natal Ed 2005;90:F49–F52. 23 Fabres J, Carlo WA, Phillips V, Howard capnia. Am J Physiol 1997;272:
12 Foix-L’helias L, Baud O, Lenclen R, G, Ambalavanan N: Both extremes of H559–H566.
Kaminski M, Lacaze-Masmonteil T: arterial carbon dioxide pressure and the 35 Ito H, Kanno I, Ibaraki M, Hatazawa J,
Benefit of antenatal glucocorticoids magnitude of fluctuations in arterial Miura S: Changes in human cerebral
according to the cause of very premature carbon dioxide pressure are associated blood flow and cerebral blood volume
birth. Arch Dis Child Fetal Neonatal Ed with severe intraventricular hemorrhage during hypercapnia and hypocapnia
2005;90:F46–F48. in preterm infants. Pediatrics 2007;119: measured by positron emission tomog-
13 Marron MJ, Crisafi MA, Driscoll JM Jr, 299–305. raphy. J Cereb Blood Flow Metab 2003;
Wung JT, Driscoll YT, Fay TH, et al: 24 Ni Chonghaile M, Higgins B, Laffey JG: 23:665–670.
Hearing and neurodevelopmental out- Permissive hypercapnia: role in protec- 36 Collins MP, Lorenz JM, Jetton JR, et al:
come in survivors of persistent pulmo- tive lung ventilatory strategies. Curr Hypocapnia and other ventilation-
nary hypertension of the newborn. Pedi- Opin Crit Care 2005;11:56–62. related risk factors for cerebral palsy in
atrics 1992;90:392–396. 25 Dorrington KL, Talbot NP: Human pul- low birth weight infants. Pediatr Res
14 Collins MP, Lorenz JM, Jetton JR, Paneth monary vascular responses to hypoxia 2001;50:712–719.
N: Hypocapnia and other ventilation- and hypercapnia. Pflugers Arch 2004; 37 Thome UH, Carlo WA: Permissive
related risk factors for cerebral palsy in 449:1–15. hypercapnia. Semin Neonatol 2002;17:
low birth weight infants. Pediatr Res 26 Bigatello LM, Patroniti N, Sangalli F: 409–419.
2001;50:712–719. Permissive hypercapnia. Curr Opin Crit 38 Hagen EW, Sadek-Badawi M, Carlton D,
15 Greisen G, Munck H, Lou H: Severe Care 2001;7:34–40. et al: Permissive hypercapnia and risk
hypocarbia in preterm infants and neu- 27 Darioli R, Perret C: Mechanical con- for brain injury and developmental
rodevelopmental deficit. Acta Paediatr trolled hypoventilation in status asth- impairment. Pediatrics 2008;122:
Scand 1987;76:401–404. maticus. Am Rev Respir Dis 1984;129: e583–e589.
16 Okumura A, Hayakawa F, Kato T, Itomi 385–387. 39 Levene MI, Fawer CL, Lamont RF: Risk
K, Maruyama K, Ishihara N, et al: 28 Feihl F, Eckert P, Brimioulle S, Jacobs O, factors in the development of intraven-
Hypocarbia in preterm infants with Schaller MD, Mélot C, et al: Permissive tricular haemorrhage in the preterm
periventricular leukomalacia: the rela- hypercapnia impairs pulmonary gas neonate. Arch Dis Child 1982;57:410–
tion between hypocarbia and mechani- exchange in the acute respiratory dis- 417.
cal ventilation. Pediatrics 2001;107: tress syndrome. Am J Respir Crit Care
469–475. Med 2000;162:209–215.

Hypercapnia and Hypocapnia in Neonates 215


40 Van de Bor M, van Bel F, Lineman R, 41 Wallin LA, Rosenfeld CR, Laptook AR, 43 Mariani G, Cifuentes J, Carlo WA: Ran-
Ruys JH: Perinatal factors and periven- Maravilla AM, Strand C, Campbell N, et domized trial of permissive hypercapnia
tricular-intraventricular hemorrhage in al: Neonatal intracranial hemorrhage. II. in preterm infants. Pediatrics
preterm infants. Am J Dis Child Risk factor analysis in an inborn popula- 1999;104:1082–1088.
1986;140:1125–1130. tion. Early Hum Dev 1990;23:129–137.
42 Ambalavanan N, Carlo WA: Hypocapnia
and hypercapnia in respiratory manage-
ment of newborn infants. Clin Perinatol
2001;28:517–531.

Prof. Wei Zhou, MD, PhD


Department of Neonatology, Guangzhou Children’s Hospital
Guangzhou Women and Children’s Medical Center
Guangzhou 510120 (China)
Tel. +86 20 81330578, Fax +86 20 81861650, E-Mail zhouwei_pu002@126.com

216 Tao ⭈ Zhou


Respiratory Health Problems and Environment
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 217–222

Air Pollution and Health


Regiani Carvalho-Oliveiraa ⭈ Naomi Kondo Nakagawaa,b ⭈
Paulo Hilário Nascimento Saldivaa
aDepartment of Pathology, Laboratory of Experimental Air Pollution, Laboratory of Medical Investigation 05, and bDepartment of

Physiotherapy, Communication Science and Disorders and Occupational Therapy, Laboratory of Medical Investigation 34, Faculdade
de Medicina da Universidade de São Paulo, Brazil

Abbreviations out the populations at greatest risk in the USA [1].


CO Carbon monoxide
However, the determination of air quality stan-
COPD Chronic obstructive pulmonary disease dards for health-balancing risks varies according
NO2 Nitrogen dioxide to several factors, such as technical feasibility, so-
NOx Nitrogen oxide cial, political and economical factors (according
O3 Ozone to the World Health Organization), which in turn
PM Particulate matter depend, among others, on the level of national de-
SO2 Sulfur dioxide velopment and capacity to manage air quality. This
UFP Ultrafine particles heterogeneity and the local characteristics should
VOCs Volatile organic compounds
therefore be carefully considered before adopting
a range of pollutant levels as national standards
for air quality. Table 2 summarizes typical ranges
There is strong evidence that ambient air pollu- of major pollutant concentrations found in a se-
tion exposure causes adverse health effects. Long- lection of regions around the world [2].
and short-term air pollution exposure have been Assessment of human health risks due to ex-
related to noxious effects on the pulmonary and posure to air pollution requires the character-
cardiovascular system in children, elderly, preg- ization of pollutants, sources and emissions in a
nant women and baby birth outcomes, as well as specific area (named air quality background). By
in patients with chronic respiratory diseases such determination of the concentration of a specific
as asthma and COPD. In recent decades, many ef- pollutant in the atmosphere, the level of expo-
forts to understand and to reduce the effects of air sure of receptors (humans, other animals, plants
pollutants on human health have been employed. and equipment) is indicated as the final process
Around the world, environmental agencies have of emission of this specific pollutant in the atmo-
developed and set up air pollution indicator stan- sphere, considering the sources and atmospheric
dards and criteria which have been widely used to interactions. Conceptually, in this case, the re-
characterize air quality. Table 1 summarizes the ceiver or human exposure occurs along the ‘en-
health effects of the main air pollutants and points vironmental pathway’ between the concentration
Table 1. Health effects of air pollutants and populations at great risk

Pollutants Main source Groups at risk Clinical consequences Comments

CO Incomplete Healthy adults Decreased exercise Effects increased with


combustion of children capacity anemia or chronic
carbon-containing Patients with Decreased exercise lung disease
fuels ischemic heart capacity Angina pectoris
disease (Premature mortality)

NO2 Fuel and coal Healthy adults Increased airway Effects occur at levels
combustion reactivity found indoors with
Atmospheric unvented sources of
chemistry combustion

SO2 Combustion of fuels Healthy adults and Increased respiratory Highly soluble gas with
containing sulfur COPD patients symptoms little penetration to
(coal and diesel) distal airways

O3 Atmospheric Healthy adults Decreased lung function Effects found at or below


chemistry and children Increased airway reactivity current NAAQS; effects
(from primary Athletes, outdoor Lung inflammation increased with exercise
NOx and VOCs) workers Increased respiratory Effects seen in
Asthmatics (and symptoms combination
others with Decreased exercise capacity with acid aerosols
respiratory illnesses) Increased hospitalizations

PM10 Fuel and coal Children Increased respiratory Effects seen alone or in
combustion Patients with chronic symptoms combination with SO2
Road Traffic lung/heart disease Increased respiratory illness
Atmospheric Asthmatic Decreased lung function
chemistry; Premature mortality
Soil resuspension Increased asthma
exacerbations

NAAQS = National ambient air quality standards. The items in parentheses are associations that have been shown in
some studies; additional confirmation is suggested.

and dose of pollutants. Air pollution consists of air pollutants). The main source of primary air
a highly variable and complex mixture of differ- pollutants is from factory chimneys, vehicle ex-
ent substances, which may occur in the gas, liq- haust pipes, suspension of the crust by the ac-
uid or solid phase. In the troposphere, several tion of wind and vehicular traffic, volcanic activ-
hundred different components have been found ity and sea evaporation. Secondary air pollutants
and many of them are potentially harmful to hu- are produced by chemical reactions between pri-
man health and to the environment. Air pollu- mary pollutants and natural elements of the com-
tion components are generated by human and position of the atmosphere. Weather conditions
natural activities (primary air pollutants) and by directly affect air pollutant production and con-
chemical reactions in the atmosphere (secondary centration. Air quality deteriorates when weather

218 Carvalho-Oliveira ⭈ Nakagawa ⭈ Saldiva


Table 2. Ranges of annual concentrations of NO2, SO2, O3, PM10, and CO in some regions, standard maximum annual
and 24-hour averages

Annual average concentration Standard average time

Pollutants Africa Asia Australia/ Canada / Europe South Annual 24-hour


New Zeland United America (Arithmetic
States mean)

NO2 35–65 20–75 11–28 35–70 18–57 30–82 100 μg.m−3


SO2 10–100 6–65 3–17 9–35 8–36 40–70 0.03 ppm 0.14 ppm
O3 120–300 100–250 120–310 150–180 150–350 200–600 0.12 ppm*
PM10 40–150 35–220 28–127 20–60 20–70 30–120 150 μg.m−3 50 μg.m−3
CO – – – – – – 10 μg.m−3** 35 μg.m−3*

* One-hour average time.


** Eight-hour average time.

conditions are most unfavorable to pollutant dis- from the atmosphere). CO persists longer and its
persion, which can be observed during the winter removal is more difficult.
periods, that significantly increases CO, PM and (c) Regional scale: Some gas-phase pollutants
SO2 concentrations in the atmosphere. During and fine particles (<2.5 μm diameter, but not
spring and summer periods, concentrations of UFP) have atmospheric lifetimes of days or even
O3 are increased because the chemical reaction weeks, which facilitates their spread on a regional
that forms O3 in the atmosphere depends on the scale. For example, O3, sulfate particles, and black
intensity of sunlight. The association of weather carbon particles (from fossil fuels and biomass
conditions and concentration of pollutants al- burnings) readily travel thousands of kilometers
lows the development of dispersion models. crossing national boundaries in a process known
For a better understanding of the dynamics of as long-range transport.
pollutants dispersed in the atmosphere, the geo- (d) Hemispheric and global scale: Some pollut-
graphical location and distribution of sources ants, especially those associated with greenhouse-
should be taken into account [2, 3]: warming effects (CO2, nitrous oxide and methane)
(a) Local scale: By source or by having a very have atmospheric lifetimes of years and are capable
short atmospheric lifetime (typically of the order of distribution throughout a hemisphere and ulti-
of an hour during daytime), some pollutants such mately the world. One example of the hemispheri-
as UFP and volatile pollutants are encountered in cal transport of pollutants is the O3. Because the
elevated concentrations only in areas close to the atmospheric lifetime of O3 is 1–2 weeks in sum-
emission source. mer and 1–2 months in winter, O3 produced in a
(b) Urban scale: Higher concentrations of pol- polluted region of one continent can be transport-
lutants from urban sources, such as NOx and ed to another continent all year long [4]. The pol-
CO generated by road traffic, tend to be detected lutant concentrations are often marginally higher
throughout the city. The atmospheric lifetime of in locations close to the sources, unless the sourc-
these pollutants typically lasts hours (low removal es emit very large quantities [1].

Air Pollution and Health 219


The main classical indicators of air pollution in understood. However, it is widely accepted that
epidemiological studies are PM, measured as par- PM is like a mix container of toxicologically rel-
ticles (an aerodynamic diameter <10 μm, PM10), evant components and others. Thus, local char-
NO2, SO2 and O3. However, this selection does acteristics may influence the toxicological effect
not imply that other air pollutants do not have sig- of PM, and results from studies carried out in one
nificant effects on human health and the adverse region may not necessarily be applied somewhere
effects on the environment. For example, VOCs else. The Health Effects Institute has performed
and polycyclic aromatic hydrocarbons are report- an analysis of the quality of past epidemiologi-
ed to have deleterious effects on human health. cal studies. Factors such as population socioeco-
Based on the Assessment System for Population nomic status and the slope of the pollutant dose-
Exposure Nationwide (ASPEN) model, the US response relationships from time-series studies
Environmental Protect Agency (EPA) [5] showed needed adjustment due to some problems with
that almost 50% of the total estimated pollut- statistical analysis programs. Nevertheless, the
ant-induced cancer cases could be attributed to whole body of knowledge supports that PM plays
VOCs, with another 40% of the total estimate to an important role in pollution-related air distur-
polycyclic aromatic hydrocarbons. The charac- bances in human health [2, 6, 7].
teristics of pollutants (sources, distribution in the (b) Gas air pollutants are gases or vapors that
atmosphere) and the effects on human health are are not condensed and discharged into the atmo-
briefly discussed below: sphere. Gaseous air pollutants are readily taken
(a) PM varies in number, size, shape, surface into the human respiratory system, although if
area, chemical composition, solubility, and origin water-soluble they may very quickly be deposited
(dust, fumes and all sorts of solid and liquid ma- in the upper respiratory tract and not penetrate
terials that remain suspended in the atmosphere). deeply into the lungs. Currently the most impor-
Total suspended particles in the ambient air have tant gaseous pollutants are those from burning
trimodal size distribution, including coarse par- fossil fuels such as O3, SO2, NOx and VOCs. Both
ticles, fine particles, and UFP. They also show PM NO2 and VOCs are the precursors of O3 in the tro-
with a diameter of ≤2.5 μm (PM2.5), PM10, and posphere. The history of air pollution is directly
UFP fractions, which are typically those mea- linked to SO2 emissions. The first scientific man-
sured within the atmosphere for the purposes uscripts reporting air pollution-related problems
of epidemiological health effect studies; the first correlated with episodes of high concentrations
two fractions are also used for compliance moni- of SO2 and PM. From them, the severe episode
toring. The main sources of PM emission to the of air pollution that occurred at London in 1952,
atmosphere are automotive vehicles, industrial dubbed ‘London smog’ is one of the most known.
processes, biomass burning, and resuspension of Nowadays in developed countries, much of the
dust from the ground, among others. Depending sulfur element is removed from motor fuels in
on the compounds and processes involved during the refining process and from stack gases prior to
its formation, the emission can be classified either emission. However, in developing countries, un-
as primary or secondary main source with com- abated burning of coal and the use of fuel oils and
plex composition which may include sulfates, ni- automotive diesel with higher sulfur content are
trates, and, particularly, polar oxidized organics. the major sources of SO2, which is undoubtedly
The mechanisms by which particles influence hu- the main source of SO2 atmospheric emission and
man health are only poorly understood. The risk contains expressive amounts of sulfur (typically
assessment of coarse particles (i.e. the size frac- between 1 and 5%). Another major mechanism
tion between 2.5 and 10 μm) is not completely of SO2 production and emission is the sintering

220 Carvalho-Oliveira ⭈ Nakagawa ⭈ Saldiva


process used in metal smelting, which involves air quality guidelines a daily maximum 8-hour
roasting metal sulfide [2, 6]. NOx is produced by mean amount of 100 μg•m3 of O3. However, some
fuel combustion processes and by chemical atmo- health effects may occur below this level. In addi-
spheric reactions. From the fuel combustion pro- tion to the health effects, O3 damages ecosystems,
cess, the use of coal is the major nitrogen prod- agricultural crops and materials.
uct source compared with oil or gas combustions Air pollutants can be measured by several in-
(at the same temperature). However, during the struments that may vary greatly in complexity and
high-temperature combustion process, oil and costs. To determine the background of air pollu-
gases produce a great amount of NOx in the at- tion, simple to complex methods can be used to
mosphere by the combination of nitrogen and at- adequately estimate the short- and long-term av-
mospheric oxygen released. For this reason, road erage pollutant concentrations and trends. One of
traffic and generation of electricity are huge sourc- the simplest methods is the passive samplers of
es of NOx emission nowadays. In NOx products, pollutants that are used to screen air quality stud-
NO2 is the most important pollutant that adverse- ies. However, complex, more expensive and ad-
ly affects human health. Typically, 5% of the total vanced monitoring systems are needed to assess
NO2 is emitted directly into the atmosphere by the air pollution distributions and sources. Currently,
combustion process. The majority of NO2 is a sec- mathematics modeling, as well as modeling of
ondary product of an atmospheric chemical re- receptors, emissions and dispersions, have been
action (the oxidation of nitrogen monoxide, NO, used to determine emission sources and the en-
after dilution in air). The conversion of NO into vironmental distribution of pollutants. Although
NO2 occurs as part of the organic compound oxi- studies of mathematical modeling require long-
dation, initiated by reactive species (OH radical). term and adequate monitoring of pollutants and
Once this conversion occurs, there are a variety of climatic conditions, these models provide the de-
other chemical reaction pathways that may follow velopment of effective control strategies [8, 9].
[2, 6]. O3 is the most important photochemical
oxidant in the troposphere, and its production is
mostly due to human-related activities (fuel com- Conclusion
bustion vehicle-related). O3 is formed by photo-
chemical reactions in the presence of sunlight Despite all efforts to improve air quality, huge
and other pollutants, such as NOx and VOCs. problems related to atmospheric contamination
The complex chemical reaction involving VOCs are frequently found in urban areas. In addition,
and NOx (where NOx = NO + NO2) leads to the developing countries are currently facing other
formation of not only O3, but a variety of addi- major challenges to control the emission of pol-
tional oxidative species. There is evidence from lutants with the use of new fuels and their produc-
controlled human and animal exposure studies tion. According to Ott [10], human exposure can
of the potential for O3 to cause adverse health ef- be defined as ‘the event that a person comes into
fects. In short-term studies that evaluated pulmo- contact with a pollutant in a certain concentration
nary function, lung inflammation and permeabil- during a certain period of time’. Therefore, the ac-
ity, respiratory symptoms, quantity of medication, curacy of air quality data and representativeness
morbidity and mortality, O3 does appear to have in area and period are important factors for the
an independent effect (especially in the summer assessment of the relative risk of air pollution on
periods). However, the role of each independent human morbidity and mortality.
factor has not been fully determined [2, 6]. The
World Health Organization recommends in its

Air Pollution and Health 221


References
1 Bernard SM, Samet JM, Grambsch A, et 4 Akimoto H: Global air quality and pollu- 8 Lodge PJ: Methods of Air Sampling and
al: The potential impacts of climate vari- tion. Science 2003;302:1716–1719. Analysis, ed 3. Washington, Lewis Pub-
ability and change on air pollution- 5 EPA: Handbook for criteria pollutant lishers, 1989, pp 83–214.
related health effects in the United inventory development: A beginner’s 9 Krochmal D, Kalina A: A method of
States. Environ Health Perspect 2001; guide for point and area sources. Wash- nitrogen dioxide and sulphur dioxide
109:199–209. ington, Environmental Protection determination in ambient air by use of
2 World Health Organization: Air quality Agency, 1999. passive samplers and ion chromatogra-
guidelines, Global Update, 2005 (www. 6 Finlayson-Pitts BJ, Pitts JN Jr: Tropo- phy. Atmos Environ 1997;31:3473–3479.
who.int/phe/health_topics/outdoorair_ spheric air pollution: ozone, airborne 10 Ott WR: Concepts of human exposure to
agg/en/). toxics, polycyclic aromatic hydrocar- air pollutants. Environ Int 1982;7:179–
3 Holguin F, Flores S, Ross Z, et al: Traffic- bons, and particles. Science 1997;276: 196.
related exposures, airway function, 1045–1050.
inflammation and respiratory symptoms 7 Englert N: Fine particles and human
in children. Am J Respir Crit Care Med health: a review of epidemiological stud-
2007;176:1236–1242. ies. Toxicol Lett 2004;149:235–242.

Regiani Carvalho-Oliveira, PhD


Department of Pathology, Laboratory of Experimental Air Pollution, Laboratory of Medical Investigation 05
Faculdade de Medicina da Universidade de São Paulo
Av. Dr. Arnaldo 455, Rm 1150/1220, São Paulo 01246-903 (Brazil)
Tel. +55 11 3061 8520, Fax +55 11 3068 0072, E-Mail regiani@usp.br

222 Carvalho-Oliveira ⭈ Nakagawa ⭈ Saldiva


Respiratory Health Problems and Environment
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 223–230

Air Pollution and Non-Invasive Respiratory


Assessments
Naomi Kondo Nakagawaa,b ⭈ Mayumi Nakaob ⭈ Danielle Miyuki Gotoa,b ⭈
Beatriz Mangueira Saraiva-Romanholoc
a
Department of Physiotherapy, Communication Science and Disorders and Occupational Therapy, Laboratory of Medical
Investigation 34, bDepartment of Pathology, Laboratory of Medical Investigation 05, and cDepartment of Internal Medicine,
Laboratory of Medical Investigation 20, Faculdade de Medicina da Universidade de São Paulo, Brazil

Abbreviations relatively non-invasive methods for assessment of


COPD Chronic obstructive pulmonary disease
the normal biological processes and respiratory
EBC Exhaled breath condensate inflammation or dysfunction are used in current
FeNO Nitric oxide in exhaled air research. The biomarkers quantitatively or semi-
FEV1 Forced expiratory volume in 1 s quantitatively measure the extent of lung inflam-
FVC Forced vital capacity mation and/or damage without any major restric-
NL Nasal lavage tion related to the exposure conditions, age, or
NO Nitric oxide health status of the subjects. The analysis of lung
STT Saccharine transit time function, induced sputum, exhaled air, EBC and
STT test are feasible, repeatable and non-invasive
assessments to detect early lung damage induced
The respiratory system represents the route of by diseases and external agents. These methods
entry for many environmental and occupation- can also be useful to identify new biomarkers of
al pollutants. Inhalation of toxic particles and exposure or susceptibility in subjects/patients to
noxious gases affects the innate defense mech- enhance the understanding of airways chang-
anisms from the nose to the alveoli by increas- es due to diseases and noxious exposition to air
ing epithelial permeability, reducing mucociliary pollution.
clearance, activating neutrophils and depressing
macrophage function. Documentation of the ef-
fects of air pollution on the respiratory tract has Lung Function Testing
been performed in epidemiological studies and
established by a variety of methods and tech- The most important aspects of spirometry are: (a)
niques that acute episodes of air pollution are as- the FVC, which is the volume delivered during an
sociated with an increased risk of adverse pulmo- expiration made as forcefully and completely as
nary and cardiovascular events. Non-invasive or possible, which starts from full inspiration, and
A B

Fig. 1. Lung function testing (A) and induced sputum performance with an ultrasonic nebulizer (B).

(b) the FEV1, which is the volume delivered in the Adverse health effects and worsening of the clini-
first second of an FVC maneuver. FEV1 is an inde- cal status have also been reported after chronic air
pendent predictor of respiratory and cardiovascu- pollution exposition in the general population [9]
lar mortality [1]. Other spirometric variables de- and adult asthmatics [10].
rived from the FVC maneuver are also addressed
in epidemiological studies of pollutant effects in
children and adult asthmatics, such as peak expi- Sputum Induction
ratory flow. Spirometry can be undertaken with
many different types of equipment. However, this Sputum cells recovered from spontaneous cough-
method requires cooperation of the subject (fig. ing were first examined in stained smears in the
1A) and the results obtained will depend on tech- 1950s through the 1970s to study lung cancer and
nical as well as personal factors. The interpretation respiratory infection. Now, induced sputum is a
of lung function tests requires the classification of well-recognized, repeatable, useful and relatively
the derived values considering the reference pop- non-invasive sampling method for research and
ulation, and the integration of the results into the clinical use to diagnose and monitor clinical and
diagnosis for an individual patient [2]. It is the subclinical airway and bronchial inflammation,
gold standard method to measure airflow limita- as well as infection and other respiratory and sys-
tion [3, 4]. However, it does not completely de- temic diseases with lung involvement. There is
fine the extent and severity of the disease. Acute a large body of knowledge on sputum charac-
exposition to air pollutants (particulate matter teristics, particularly in relation to the inflam-
and noxious gases) is associated with respira- matory cells content [11–14] and mediators [15,
tory symptoms and a significant decline in lung 16], matrix metalloproteinases [17], and physi-
function in children [5], in young healthy volun- cal properties and appearance [18, 19]. Induced
teers (FEV1 and FVC, but not in the FEV1/FVC sputum tests (fig. 1B) are performed with 3, 3.5,
ratio parameter) [6, 7], and adult asthmatics [8]. 4, 4.5, 5 or 7% of saline solution that are given

224 Nakagawa ⭈ Nakao ⭈ Goto ⭈ Saraiva-Romanholo


A B

Fig. 2. Equipment and materials for collection of the exhaled breath for NO analysis (A); exhaled
breath collection for NO analysis (B).

sequentially in the amount of 5–7 ml in the form Exhaled Breath


of a spray produced by an ultrasonic nebulizer.
Following each dose of inhaled hypertonic saline The presence of NO in exhaled air, FeNO,
through a mouthpiece, the subject coughs and ex- of human and animals was first described in
pectorates the sputum into a sterile container or 1991. FeNO is the most extensively studied ex-
in special conditions; sputum can be suctioned. haled biomarker in respiratory diseases (fig. 2).
The induced sputum method with hypertonic sa- Measurements of FeNO can be performed on-
line has been shown to effectively and safely re- line and offline, with similar application in epide-
cruit the mucus from the alveoli of patients. Each miology. However, influence of ambient NO on
inhalation is followed by spirometry to check for FeNO should be carefully examined [24]. NO is
any FEV1 reduction. However, if a 20% reduc- a simple gaseous molecule composed of a single
tion from the baseline value of FEV1 occurs after nitrogen atom and one of oxygen atom, but it is
salbutamol, sputum induction should be discon- a highly reactive chemical agent that has a short
tinued. This method allows the examination of half-life when diluted (<10 s) due to its rapid oxi-
inflammatory events and can also be applied to dation. NO is a messenger of physiological pro-
assess the effects of inhaled toxic gases or parti- cess that stimulates the synthesis of cyclic GMP
cles in the airways and lungs in both normal sub- and promotes the relaxation of bronchial smooth
jects and patients with disease [20, 21]. However, muscle. NO is constitutively expressed in bron-
this method is relatively difficult to perform in chial epithelium and alveolar epithelial type II
the routine evaluation of children and adoles- cells are also found in human pulmonary blood
cents [15, 22] and it is time-consuming [13]. vessels and all types of arteries, particularly in
Interestingly, studies have raised the possibility the radial artery. The NO generated by NO syn-
to trace metals, such as zinc, manganese, and thase is responsible for maintaining low vascu-
copper, in induced sputum as biomarkers of in- lar tone and preventing the adhesion of leuko-
flammation in lung diseases [23]. cytes and platelets in the vascular wall. On the

Air Pollution and Non-Invasive Respiratory Assessments 225


contrary, it can also lead to vasodilatation in the a real-time assessment of pulmonary pathobiolo-
arterioles, leading to plasma extravasation and gy. Fluid pH and several biochemical components
edema. NO produced from induced NO syn- can be analyzed in EBC. EBC analysis is a simple,
thase may have cytostatic and cytotoxic effects. easy, safe, and non-invasive method to investigate
NO has pro- and antioxidant effects, depending the oxidative stress in many respiratory diseas-
on location and concentration. At low concentra- es and other conditions, also due to medication
tions, NO has protective actions, including neu- evaluation and air pollution exposure. Recently,
romodulation, smooth muscle relaxation and re- in adult patients with mild and moderate asthma,
duction of bronchial hyperresponsiveness caused McCreanor et al. [8] showed that breathing pol-
by bronchoconstrictor stimuli. However, in high luted air during a 2-hour walk on Oxford Street
concentrations, as observed in several pathologi- in London, where only diesel-powered vehicles
cal conditions, NO has deleterious effects, such are allowed, resulted in lung function reduction
as proinflammatory activity with increased gland accompanied by respiratory inflammatory activ-
secretion into the airways, activation and recruit- ity. They found airway acidification (a twofold in-
ment of numerous inflammatory cells, apopto- crease in hydrogen ions after 2 h of exposure) in
sis and necrosis. Significantly increased values EBC samples, increased levels of myeloperoxidase
of FeNO are observed in patients with asthma, and interleukin-8 in sputum samples and high-
COPD patients with sputum eosinophilia, and er fraction of NO in exhaled breath. The endog-
cystic fibrosis [25, 26]. However, FeNO mea- enous airway acidification assessed by pH of EBC
surement in COPD is of limited value due to the represents a biomarker associated with oxidative
smoking effect and the results are dependent of stress and sputum neutrophilia. There is evidence
exhalation rates. Recently, Saraiva-Romanholo et that oxidative stress may have an important role
al. [27] have reported a similar pattern to asth- in exacerbating COPD. The concentration of hy-
ma of increased FeNO in non-asthmatic patients drogen peroxide and 8-isoprostane are increased
during bronchospasm episodes in the surgical during COPD exacerbations. Many mechanisms
room. The concentration of FeNO has also been are raised to enhance inflammation and proteolyt-
used to measure human response to air pollut- ic injury, such as the activation of transcription
ants. Increased levels of FeNO can be found in factor nuclear factor-κB [29]. Recently, Salvi and
non-smoking subjects exposed to air pollution Barnes [30] reported evidence in the literature for
showing that airways inflammation is present the association of COPD with biomass fuel, oc-
with no clinical presentation [7]. cupational exposure to dusts and gases, and out-
door air pollution. EBC has limitations, such as
considerable variability due to the sample collec-
Exhaled Breath Condensate tion and analysis [31]. The EBC sample from tidal
breathing is obtained with the use of an appara-
EBC is a promising biological fluid that can be tus (‘breath condensate collector’). According to
easily and quickly obtained by cooling exhaled Koczulla et al. [32], there are no differences be-
air under condition of spontaneous breathing. tween EBC devices in healthy controls, asthmat-
Participants are asked to breathe tidally through ics and COPD patients for pH measurements. An
the mouthpiece connected to the tube during 10– example of the modified apparatus is showed in
15 min to collect approximately 2–3 ml of EBC, figure 2B. Another point is the exact site of ori-
which is aliquoted and frozen to –70°C. [28]. The gin of substances measured in EBC. Jackson et al.
analysis of EBC is increasingly studied as a non- [33] showed in a very elegant work that biomark-
invasive research method of sampling the lungs, as ers of inflammation and oxidative stress are easily

226 Nakagawa ⭈ Nakao ⭈ Goto ⭈ Saraiva-Romanholo


d a
c
b
A
B

Fig. 3. The exhaled breath condensator modified apparatus (A): (a) buccal, (b) saliva collector, (c) T-piece with unidirec-
tional valve, (d) filter (humid vent filter compact S, Louis Gibeck AB, Upplands Väsby, Sweden), (e) EBC tube connector,
(f ) EBC tube collector, (g) condenser reservoir, and (h) expiratory unidirectional valve. This apparatus was developed
by Dr. Paulo Hilário Nascimento Saldiva, Dr. Naomi Kondo Nakagawa, Simão Bacht, and Dr. Idágene Cestari, and built
by Bioengeering Division of Heart Institute (InCor) University of São Paulo Medical School; STT test (B).

detectable and measurable in EBC. However, com- (37°C) normal saline is instilled into each nostril
paring EBC with bronchoalveolar lavage, the bio- by a syringe. After 10 s, the subject brings the
markers do not correlate (8-isoprostane, nitrogen head forward and expels the liquid into a sterile
oxides, total phospholipid and pH were higher in plastic receptacle [34–36]. The lavage fluid from
EBC than in bronchoalveolar lavage, total protein both nostrils is pooled in the same receptacle and
was lower in EBC compared with bronchoalve- kept on ice until processed. Each lavage sample is
olar lavage, and hydrogen peroxide and keratin vigorously shaken to break up clumps of mucus
were similar). and then centrifuged (1,000 g at 4°C during 10
min). Cell counts are determined from the re-
suspended pellet and differential cell counts are
Nasal Lavage performed using the cytospin preparation at 450
rpm for 6 min [35]. Many cell types found in
NL is a well-established, safe, well-tolerated and the lining of the nasopharygeal region are simi-
low-cost technique that has been used to study lar to cells of the tracheal and bronchial lining.
the acute inflammatory response in the upper Therefore, it has been suggested that the cellular
respiratory tract for more than 20 years. It was responses in the nose to toxicants are likely to be
first described by Naclerio et al. [34]. NL is per- similar to the lower airway at the same dose of
formed by sitting the subject with the nasophar- the agent. If these pollutants are respiratory irri-
ynx closed while tilting the neck back 45° from tants, capable of causing cellular damage, effects
the vertical position. Five milliliters of warm may therefore be detected in the nasal passage.

Air Pollution and Non-Invasive Respiratory Assessments 227


However, a number of studies have reported con- or coughing. The normal mean value report-
troversial findings in NL of workers and subjects ed for this assay is 11–12 min in young healthy
exposed to several inhaled substances [37, 38]. adults [40–44]. However, despite its widespread
Blaski et al. [37] did not find a relationship be- use, there are factors that may affect STT. On the
tween NL cellularity and respiratory symptoms, one hand, age and smoking prolong STT, and on
airflow obstruction, and levels of short-term the other hand, exercise and acute rhinitis ac-
dust exposure in a cross-sectional field study celerate STT.
of workers exposed to organic dusts. However,
they found increased cellularity in NL in workers
exposed to high levels of ambient organic dusts Conclusions
compared with low levels. On the other hand,
Barraza-Vilarreal et al. [38] showed in a cohort Air pollutants may induce inflammation in the
study of 158 asthmatics and 50 non-asthmatic respiratory system. Inflammation is a fundamen-
school-age children significant associations of tal process in the pathophysiological cascade
increased inflammatory marker (interleukin-8) leading to respiratory disorders, including asth-
in NL, reduced pH of EBC and decreased FEV1 ma, COPD or interstitial lung disease. Several
and FVC after a short-term air pollutant expo- safe, repeatable, and non-invasive techniques of
sure. FeNO and FEV1 were inversely correlated inflammatory monitoring, and assessment of a
in asthmatic children. disease’s severity or host response to treatments
are available. The efficiency of these techniques
depends on the standardization and implemen-
Saccharin Transit Time Test tation of the procedures and technology, and ad-
equate interpretation of the results. Considering
STT test is a simple, inexpensive, repeatable and the adverse effects of air pollution, there is a spe-
non-invasive method for some purposes: (a) in cial interest for biomarkers that reflect or induce
vivo evaluation of the abnormal mucociliary oxidative stress, and inflammatory elements and
clearance in several conditions, (b) identifica- processes.
tion of subjects that other assessment studies
should be carried out, such as mucus properties
analysis, ciliary motility and structure, and (c) Acknowledgments
assessment of clinical and surgical interventions.
The authors would like to thank S. Fidalgo, T.M. Lima,
The STT test was first described by Andersen et
and A.S. Santos for providing the photographs shown of
al. [39] and modified by Rutland and Cole [40]. figures 1A, 3A, and 3B, respectively. The EBC-modified
To perform the STT test (fig. 2A), a 5-mg par- apparatus was developed by Dr. Paulo Hilário Nascimento
ticle of saccharine is placed 2 cm inside the non- Saldiva, Dr. Naomi Kondo Nakagawa, Simão Bacht, and
obstructed nostril, on the inferior turbinate un- Dr. Idágene Cestari, and built by Bioengeering Division of
Heart Institute (InCor) University of São Paulo Medical
der visual control while the subject is quiet and School.
seated. A timer is displayed to measure the tran-
sit time. STT is the elapsed time from the place-
ment of the particle into the nasal mucosa until
the subject reports the sweet taste of saccharine.
Subjects are allowed to swallow freely, and ori-
ented to maintain normal ventilation, avoiding
deep breaths, talking, sniffing, sneezing, eating,

228 Nakagawa ⭈ Nakao ⭈ Goto ⭈ Saraiva-Romanholo


References
1 Pellegrino R, Viegi G, Brusasco V, et al: 12 Drews AC, Pizzichini MM, Pizzichini E, 24 Linn WS, Berhane KT, Rappaport EB, et
Interpretative strategies for lung func- et al: Neutrophilic airway inflammation al: Relationships of online exhaled,
tion tests. Eur Respir J 2005;26:948–968. is a main feature of induced sputum in offline exhaled, and ambient nitric oxide
2 Miller MR, Crapo R, Hankinson J, et al: nonatopic asthmatic children. Allergy in an epidemiological survey of school-
General consideration for lung function 2009;64:1597–1601. children. J Expo Sci Environ Epidemiol
testing. Eur Respir J 2005;26:153–161. 13 Saraiva-Romanholo BM, Barnabé V, 2009;19:674–681.
3 Global Initiative for Chronic Obstructive Carvalho AL, Martins MA, Saldiva PH, 25 Baraldi E, Jongste JC: Measurement of
Lung Disease (GOLD), American Tho- Nunes MP: Comparison of three meth- exhaled nitric oxide in children, 2001.
racic Society 2006. Spirometry for ods for differential cell count in induced Eur Respir J 2002;20:1–15.
healthcare providers (http://www.gold- sputum. Chest 2003;124:1060–1066. 26 Ricciardolo FLM, Sterk PJ, Gaston B, et
copd.com/OtherResourcesItem. 14 Fireman E: Induced sputum as a diag- al: Nitric oxide in health and disease of
asp?l1=2&l2=2&intId=1836). nostic tactic in pulmonary diseases. Isr the respiratory system. Physiol Rev 2004;
4 American Thoracic Society: Standard- Med Assoc J 2003;5:524–527. 84:731–765.
ization of spirometry. Am J Respir Crit 15 Gagliardo R, La Grutta S, Chanez P, et al: 27 Saraiva-Romanholo BM, Machado FS,
Care Med 1995;152:1107–1136. Non-invasive markers of airway inflam- Almeida FM, et al: Non-asthmatic
5 Liu L, Poon R, Chen L, et al: Acute mation and remodeling in childhood patients show increased exhaled nitric
effects of air pollution on pulmonary asthma. Pediatr Allergy Immunol 2009; oxide concentrations. Clinics 2009;64:
function, airway inflammation, and oxi- 20:780–790. 5–10.
dative stress in asthmatic children. Envi- 16 Fireman E, Gilberto D, Marmor S: 28 Cepelak I, Dodig S: Exhaled breath con-
ron Health Perspect 2009;117:668–674. Angiogenic cytokines in induced spu- densate: a new method for lung disease
6 Steinvil A, Fireman E, Kordova-Biezuner tum of patients with sarcoidosis. Respi- diagnosis. Clin Chem Lab Med
L, et al: Environmental air pollution has rology 2009;14:117–123. 2007;45:945–952.
decremental effects on pulmonary func- 17 Fireman E, Kraiem Z, Sade O, et al: 29 Barnes PJ: Medical progress: chronic
tion test parameters up to one week after Induced sputum-retrieved matrix metal- obstructive pulmonary disease. N Engl J
exposure. Am J Med Sci 2009;338:273– loproteinase-9 and tissue metalloprotei- Med 2000;343:269–280.
279. nase inhibitor-1 in granulomatous dis- 30 Salvi SS, Barnes PJ: Chronic obstructive
7 Guarnieri G, Lodde V, Ferrazzoni S, et al: eases. Clin Exp Immunol 2002;130: pulmonary disease in non-smokers.
Acute effects of environmental pollution 331–337. Lancet 2009;374:733–743.
on the urban vigilants airways. G Ital 18 Murray MP, Pentland JL, Turnbull K, et 31 Leung TF, Li CY, Yung E, et al: Clinical
Med Lav Ergon 2007;29:838–840. al: Sputum colour: a useful clinical tool and technical factors affecting pH and
8 McCreanor J, Cullinan P, Nieuwenhui- in non-cystic fibrosis bronchiectasis. Eur other biomarkers in exhaled breath con-
jsen MJ, et al: Respiratory effects of Respir J 2009;34:361–364. densate. Pediatr Pulmonol 2006;41:
exposure to diesel traffic in persons with 19 Albertini-Yagi CS, Oliveira RC, Vieira JE, 87–94.
asthma. N Engl J Med 2007;357:2348– et al: Sputum induction as a research 32 Koczulla R, Dragonieri S, Schot R, et al:
2358. tool for the study of human respiratory Comparison of exhaled breath conden-
9 Ackerman-Liebrih U, Leuenberger P, mucus. Respir Physiol Neurobiol 2005; sate pH using two commercially avail-
Shwartz J, et al: Lung function and long- 145:101–110. able devices in healthy controls, asthma
term exposure to air pollutants in Swit- 20 Hogg JC, van Eeden S: Pulmonary and and COPD patients. Respir Res 2009;10:
zerland. Study on air pollution and lung systemic response to atmospheric pollu- 78.
diseases in adults (SAPALDIA) team. tion. Respirology 2009;14:336–346. 33 Jackson AS, Sandrini A, Campbell C, et
Am J Respir Crit Care Med 1009;155: 21 Mussafi H, Fireman EM, Mei-Zahav M, al: Comparison of biomarkers in exhaled
122–129. et al: Induced sputum in the very young. breath condensate and bronchoalveolar
10 Canova C, Torresan S, Simonato L, et al: Chest 2008;133:176–182. lavage. Am J Crit Care Med 2007;175:
Carbon monoxide pollution is associated 22 Paro-Heitor ML, Bussamra MH, Saraiva- 222–227.
with decreased lung function in asth- Romanholo BM, et al: Exhaled nitric 34 Naclerio RM, Meier HL, Kagey-Sobotka
matic adults. Eur Respir J 2009;35:266– oxide for monitoring childhood asthma A, et al: Mediator release after nasal
272. inflammation compared to sputum anal- challenge with allergen. Am Rev Respir
11 Pizzichini E, Pizzichini MMM, Kidney ysis, serum interleukins and pulmonary Dis 1983;128:597–602.
JC, et al: Induced sputum, bronchoalveo- function. Pediatr Pulmonol 2008;43: 35 Diaz-Sanches D, Dotson AR, Takanaka
lar lavage and blood from mild asthmat- 134–141. H, et al. Diesel exhaust particles induce
ics: inflammatory cells lymphocyte sub- 23 Gray RD, Duncan A, Noble D, et al: Spu- local IgE production in vivo and alter the
sets and soluble markers compared. Eur tum trace metals are biomarkers of pattern of IgE messenger RNA isoforms.
Respir J 1998;11:828–834. inflammatory and suppurative lung dis- J Clin Invest 1994;94:1417–1425.
ease. Chest 2010;37:635–641.

Air Pollution and Non-Invasive Respiratory Assessments 229


36 Diaz-Sanches D, Id RR, Gong H: Chal- 39 Andersen I, Camner P, Jensen PL, et al: 42 Persson C, Svensson C, Greiff L, et al:
lenge with environmental tobacco Nasal clearance in monozygotic twins. The use of the nose to study the inflam-
smoke exacerbates allergic airway dis- Am Rev Respir Dis 1974;110:301–305. matory response of the respiratory tract.
ease in human beings. J Allergy Clin 40 Rutland J, Cole PJ: Nasal mucociliary Thorax 1992;47:993–1000.
Immunol 2006;118:441–446. clearance and ciliary beat frequency in 43 Stanley PJ, Wilson R, Greenstone MA, et
37 Blaski CA, Watt JL, Quinn TJ, et al: Nasal cystic fibrosis compared with sinusitis al: Effect of cigarette smoking on nasal
lavage cellularity, grain dust, and airflow and bronchiectasis. Thorax 1981;36: mucociliary clearance and ciliary beat
obstruction. Chest 1996;109:1086–1092. 654–658. frequency. Thorax 1986;41:519–523.
38 Barraza-Villarreal A, Sunyer J, Hernan- 41 Nakagawa NK, Franchini ML, Driusso P, 44 Goto DM, Torres GM, Seguro AC, Sal-
dez-Cadena L, et al: Air pollution, air- et al: Mucociliary clearance is impaired diva PHN, Lorenzi-Filho G, Nakagawa
way inflammation, and lung function in in acutely ill patients. Chest 2005;128: NK: Furosemide impairs nasal mucocili-
a cohort study of Mexico City schoolchil- 2772–2777. ary clearance in humans. Respir Physiol
dren. Environ Health Perspect 2008;116: Neurobiol 2010;170:246–252.
832–838.

Naomi Kondo Nakagawa, BSc, PhD


Department of Physiotherapy, Communication Science and Disorders and Occupational Therapy
Laboratory of Medical Investigation 34, Faculdade de Medicina da Universidade de São Paulo
Av. Dr. Arnaldo 455, São Paulo 01246-903 (Brazil)
Tel. +55 11 3061 8529, Fax +55 11 3068 0072, E-Mail naomi.kondo@usp.br

230 Nakagawa ⭈ Nakao ⭈ Goto ⭈ Saraiva-Romanholo


Respiratory Health Problems and Environment
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 231–237

Air Pollution, Oxidative Stress and Pulmonary


Defense
Mariangela Macchionea ⭈ Maria Lúcia Bueno Garciab
a
Department of Pathology, Laboratory of Experimental Air Pollution, Laboratory of Medical Investigation 05, and bDepartment of
Internal Medicine, Laboratory of Experimental Therapeutics, Faculdade de Medicina da Universidade de São Paulo, Brazil

Abbreviations pathogens and particles. They also respond to at-


ARE Antioxidant response element
tacks of air pollutants, ranging from responses
ERK Extracellular signal-regulated protein kinases of extracellular stimuli and stress by activating
JNK Jun NH2-terminal kinases signaling pathways that lead to inflammation.
Keap1 Kelch-like ECH-associated protein Airway epithelial cells can produce a diverse ar-
MAPK Mitogen activation protein kinases ray of proinflammatory mediators, growth factors
MAPKK MAPK kinase and cytokines in response to environmental chal-
MAPKKK MAPK kinase kinase lenge, and are actively involved in different stages
NF-κB Nuclear factor-κB of epithelial repair. In vivo, the ability to generate
Nrf2 Nuclear factor erythroid-2-related factor
oxidants correlates with the ability to produce in-
PAH Polycyclic aromatic hydrocarbons
flammation [2]. In vitro, the acute inflammatory
PM Particulate matter
ROS Reactive oxygen species response to ambient particulate pollutants (PM)
seems to reflect oxidative activation of fundamen-
tal cell signaling pathways [3].
Many air pollutants exert their major effects by
Air Pollution and Oxidative Stress causing oxidative stress in cells and tissues. Gaseous
pollutants and PM are known to form ROS such as
Air pollutants affect different organs in mamma- superoxide anion, hydrogen peroxide and hydrox-
lians and cause several health problems, includ- yl radicals that may damage proteins, lipids and
ing respiratory, cardiovascular, neurological, re- DNA directly, and form distinct products.
productive and developmental ailments and even
cancer [1]. In addition, exposure to air pollutants
is reported to alter immune response and enhance Exogenous Oxidants
respiratory infection [1].
Respiratory epithelium cells are not a simple Several air pollution components have been asso-
barrier between the body and the external en- ciated to particulate toxicity. An important deter-
vironmental that protect against inhaled toxins, minant of the acute inflammatory response seems
to be the dose of bioavailability of transition met- inflammatory cells capable of generating ROS and
als (copper, vanadium, chromium, nickel, cobalt reactive nitrogen species as well as DNA damage
and iron) as well as organic components (PAH) [8]. Exposures to diesel exhaust particles, which
and biological fractions (endotoxins) [4]. contains polyaromatic hydrocarbons, quinones
Metals such as iron, copper, chromium, vana- and redox-active metals adsorbed to it, has shown
dium and cobalt are capable of redox cycling in to increase IL-6 and IL-8 in bronchial alveolar la-
which a single electron may be accepted or do- vage in humans and upregulate endothelial adhe-
nated by the metal. This action catalyzes reactions sion molecules P-selectin and vascular cell adhe-
that produce reactive radicals and can produce sion molecule-1 [9].
ROS. The most important reactions are probably In concern to toxic gaseous, ozone is a very
Fenton’s reaction and the Haber-Weiss reaction, reactive gas that forms secondary ROS directly
in which hydroxyl radical is produced from the by ozonization of the tract lining fluid lipids, in-
reduced iron and hydrogen peroxide. ducing lipid peroxidation as well as activation of
Exposures to oxidant air pollutants can promote transcription factors such as NF-κB and increased
lipid peroxidation that is caused by a free radical expression of several proinflammatory cytokines
reaction involving the polyunsaturated fatty acids and adhesion genes [10]. Nitrogen dioxide, like
of the membrane. This reaction can damage cell ozone, reacts with substrates present in the lung
membrane leading to cell death and pathological lining fluid compartment where the signaling cas-
consequences. In addition, one of these end-prod- cade of inflammatory cells into the lung occurs
ucts, 4-hydroxy-2-noneal, causes in vitro deple- [11].
tion of intracellular glutathione and induction of PAH air pollution originates mainly due to the
peroxide production, airway remodeling through incomplete combustion of wood or fuel used for
activation of the epidermal growth factor recep- residential heating, industrial or motor vehicle ex-
tor, providing evidence for the hypothesis that sec- haust. A number of studies have considered DNA
ondary mediators generated by oxidant reactions damage as the endpoint or the effects of pollu-
with lipids, proteins and others biomolecules con- tion in particular ‘bulky’ DNA adducts and also
tribute to toxic effects of pollutants [5]. The oxi- protein adducts which are related to exposure to
dant air pollutants can also promote modification aromatic compounds including PAH [12]. Select
in the proteins, particularly those that have amino PAH as quinones can also induce oxidative stress
acid composition with cysteine and methionine in cells and decrease the GSH concentration
residues, which are more susceptible to oxidation through electrophilic reaction [13]. Kikuno et al.
[6]. Studies also have shown that exposure to air [14] showed that 1,2-naphthoquinone, a PM qui-
pollutants can induce DNA damage, increasing none, may cause tyrosine kinase, phosphorylation
the risk of lung cancer [7]. by activating phospholipase A2/lipoxygenase/
Air pollution is made up of several compo- vanilloid receptor 1, signaling and increasing cal-
nents constantly reacting to each other as parti- cium levels which, in turn, resulted in contraction
cles and toxic gases. Both components are toxic of tracheal smooth muscle.
to healthy. PM contains a carbonaceous core with
adsorbed organic and inorganic materials as met-
als. Fine PM is within 2.5 μm of aerodynamic di- Oxidant-Induced Cell Signaling
ameter (PM <10 μm) as they can reach the dis-
tal lung tissue and blood circulation. PM may Oxidant pollutants through ROS may activate sig-
alter the function of mitochondria, reduce nic- nalization pathways that lead to cellular responses
otinamide adenine phosphate oxidase, activate which can trigger pathological responses in the

232 Macchione ⭈ Bueno Garcia


Stress, UV, heat-shock proteins

Protein synthesis inhibitors


Growth factor cytokines
Ligands/stimuli
Tyrosine kinase receptor

MAP4K GTP ras ras

raf MEKK
MAP3K

SER-PO4 THR-PO4 SER-PO4 THR-PO4

MAP2K MEK ?

THR-PO4 TYR-PO4 THR-PO4 TYR-PO4


MAPK ERKs JNK
SAPKs

Cellular differentiation or proliferation Stress effects

Fig. 1. MAPK cascades. Illustration of the three-tiered MAPK cascades for ERK and JNK family
members.

lung. The main types of signal transduction path- the specific response to the stimulus [16]. Other
ways in eukaryotic cells are protein kinase cas- studies have demonstrated the ability of air pollut-
cades that culminate in activation of a family of ants to activate NF-κB through of the epidermal
protein kinases known as MAPK [15]. They are growth factor receptor activation and the MAPK
a group of 38–110 kDA Ser/Thr that translate signaling pathway, which is involved in the stress
signals which lead to diverse cellular responses. response [17].
There are three major MAPK pathways which in-
cludes a Ser/Thr MAPK that is phosphorylated by
a Thr/Tyr MAPKK which is itself phosphorylated Endogenous Oxidants
by a Ser/Thr kinase known as a MAPKKK (fig.
1). There are three subtypes of MAPK: one is ac- Endogenous sources of ROS may have an indi-
tivated through ERK that mediates cell prolifera- rect role in the toxicity induced by exposure to air
tion, while the other two subtypes are p38 MAPK pollutants. The cells that produce ROS in the lung
and p46 to p54 MAPK (JNK) that mediate signals are neutrophils, eosinophils, alveolar macrophag-
in response to environmental stress and cytok- es, epithelial cells and endothelial cells. These cells
ines which results in the phosphorylation-depen- are recruited when air pollutants induce lung in-
dent activation of a variety of transcription fac- flammation and release ROS, enhancing inflam-
tors (e.g. Elk1, c-Jun, ATF-2). They also mediate mation with tissue damage and other pathological

Air Pollution, Oxidative Stress and Pulmonary Defense 233


Cell

O2

Mitochondria Enzymes of respiratory chain

P450 oxidase Cytosol enzymes


RE
Peroxisome
Oxidase
O2 –
NADPH oxidase
Fenton
Fe 2+ Fe 3+
Reaction
Superoxide H2O
O2– dismutase
H2O2 OH– + OH–
2OSH
Catalase
Glutathione Glutathione
peroxidase redutase

H2 O H2O
OOSG

Cell damage

Fig. 2. Glutathione peroxidase and oxidative stress.

effects. Inflammatory cells produce mainly super- to air pollutant-induced toxicity through an en-
oxide and hydrogen peroxide that can react with hancement of the oxidative burden within the
a number of substrates and biomolecules that lung.
generate radicals. ROS is also involved with reac-
tions of the electron transport chain within the
mitochondria and enzyme reactions involving cy- Pulmonary Antioxidants
clooxygenases, lipoxygenases, peroxygenases and
cytochrome P450. Alteration in these processes or There are several enzymatic and non-enzymatic
decrement in the antioxidants that offset the pro- systems that contribute to the inactivation of free
duction of ROS may also lead to tissue damage radicals. Most of them are in the airways, the main
and other pathological consequences [18]. and first barrier to external aggressors.
Peroxynitrite is another potential oxidant that The non-enzymatic antioxidant system blocks
is formed by the reaction of nitric oxide and super- the beginning of free radicals formation as well as
oxide, inducing lipid peroxidation, DNA damage their inactivation by scavengers and the final cel-
and protein oxidation. These studies demonstrate lular and tissue damage. Some examples of this
how endogenous sources of ROS can contribute category are vitamins: vitamin E (α-tocopherol)

234 Macchione ⭈ Bueno Garcia


Stressed cell and milieu

Oxidants
Pro-oxidants

Non-stressed cell
Oxidase
Oxidative stress

Keap1
Keap1 Nrf2

ROS
Nrf2
Antioxidants

P P P
Nrf2 Nrf2

ARE ARE

a
Neutrophil

b Blood vessel

Fig. 3. Nrf2-antioxidant defense system in airway cells. a In normal physiologic conditions, a bal-
ance between antioxidants and oxidants maintains cellular redox equilibrium. b Under stressed
conditions, oxidative stimuli could accelerate ROS production, directly or indirectly, and activate
Nrf2 for production of ARE-driven antioxidants, while overwhelming ROS over antioxidant capac-
ity may cause oxidative injury leading to pulmonary pathologic symptoms.

is secreted by type II pneumocyte with the sur- some radicals produced by the protein-copper or
factant, it is located in intra- and extracellular flu- iron complex, associated with iron compartmen-
ids, blocking the membrane lipid peroxidation by talization and homeostasis which facilitate the
the peroxyl radicals clearance; vitamin A (and its metal disponibility to interact with transferrin by
precursors β-carotene) and vitamin C (ascorbic the ceruloplasmin-ferroxidase activity.
acid) induce endothelial stabilization and trigger Several other enzymes are included as enzy-
vitamin E increment; uric acid, found in the nasal matic antioxidant systems which clear free radi-
mucus and in the respiratory epithelium, clears cals and lyse hydrogen peroxide and superoxide
oxidants in the mucus and superoxides; metallo- anion. These enzymes are the following: cata-
thionine, albumin and ceruloplasmin are extra- lase located in the peroxisomes, decomposes
cellular metal-binding proteins able of clearing H2O2 to O2 + H2O, enabling the membrane lipid

Air Pollution, Oxidative Stress and Pulmonary Defense 235


peroxidation through peroxyl radicals clearance; contain a cis-acting element called ARE [19]. ARE
superoxide dismutase group (MnSOD-SOD man- is activated by phenolic antioxidants, hydroper-
ganese in the mitochondria and copper-zinc-SOD oxides, quinones, heavy metals and other diverse
in the cytosol) is able to turn the superoxide into inducers that bind Nrf2 with ARE sequence and
H2O2 by blocking transition metals; glutathione activate the transcription of target genes. Under
peroxidase catalyzes and lyses free radicals (fig. homeostatic conditions, Nrf2 is in the cytoplasm
2) and the oxidized/reduced glutathione intrac- of the cell attached to an actin-binding cytosol-
ellular ratio reflects the oxidative state of the cell ic protein named Keap1 [20]. These proteins are
and is a key element in the cellular detoxification uncoupled when Nrf2-Keap1 is exposed to elec-
ability of ROS; heme oxygenase, ferritin, trans- trophiles and ROS, leading to the translocation of
ferrin and lactoferrin catalyze the heme degrada- Nrf2 to the nucleus where it can dimerize with
tion into biliverdin and may be induced by several other transcription factors binding to the ARE,
stimuli as hyperoxia, hypoxia, LPS and oxidative which causes the transcriptional activation of
stress, modulated by NF-κB and activator pro- phase II detoxifying enzymes and antioxidants
tein-1; transferrin, ferritin, lactoferrin and ce- (fig. 3).
ruloplasmin are metal protein ligands, diminish
transition metals storage levels and thus decrease
the OH• production; use of chelating agents as Conclusion
EDTA, DPTA, deferoxamine and d-penicilamine
(cuprimine). The metal chelation also improves Oxidative stress caused by air pollution may in-
the heme oxygenase and ferritin effects. duce several inflammatory signs in the lung in-
cluding exudates, cellular proliferation and influx,
matrix accumulation and airway obstruction.
Response to Oxidative Stress – Nrf2 Progress has been made to understand the mech-
anisms through which oxidants initiate and trig-
Responsible for the procedure of the antioxidant ger cell and tissue toxicity, however critical ques-
molecules is the promoter of the genes for phase tions still remain to be addressed.
II detoxifying enzymes and antioxidants that

References
1 Rubio V, Valverde M, Rojas E: Effects of 4 Soukup JM, Becker S: Human alveolar 7 Prahalad AK, Inmon J, Ghio AJ, et al:
atmospheric pollutants on the Nrf2 sur- macrophage responses to air pollution Enhancement of 2ʹ-deoxyguanosine
vival pathway. Environ Sci Pollut Res Int particulates are associated with insolu- hydroxylation and DNA damage by coal
2010;17:369–382. ble components of coarse material, and oil fly ash in relation to particulate
2 Schaumann F, Borm PJ, Herbrich A, et including particulate endotoxin. Toxicol metal content and availability. Chem Res
al: Metal-rich ambient particles (partic- Appl Pharmacol 2001;171:20–26. Toxicol 2000;13:1011–1019.
ulate matter 2.5) cause airway inflam- 5 Ciencewicki J, Trivedi S, Kleeberger SR: 8 González-Flecha B: Oxidant mecha-
mation in healthy subjects. Am J Respir Oxidants and the pathogenesis of lung nisms in response to ambient air par-
Crit Care Med 2004;170:898–903. diseases. J Allergy Clin Immunol ticles. Mol Aspects Med 2004;25:169–
3 Churg A, Xie C, Wang X, et al: Air pollu- 2008;122:456–468. 182.
tion particles activate NF-κB on contact 6 Kelly FJ, Mudway IS: Protein oxidation 9 Stenfors N, Nordenhäll C, Salvi SS, et al:
with airway epithelial cell surfaces. Toxi- at the air-lung interface. Amino Acids Different airway inflammatory
col Appl Pharmacol 2005;208:37–45. 2003;25:375–396. responses in asthmatic and healthy
humans exposed to diesel. Eur Respir J
2004;23:82–86.

236 Macchione ⭈ Bueno Garcia


10 Kelly FJ: Oxidative stress: its role in air 14 Kikuno S, Taguchi K, Iwamoto N, 17 Dagher Z, Garçon G, Billet S, et al: Role
pollution and adverse health effects. Yamano S, et al: 1,2-Naphthoquinone of nuclear factor-κB activation in the
Occup Environ Med 2003;60:612–616. activates vanilloid receptor-1 through adverse effects induced by air pollution
11 Janssen-Heininger YM, Persinger RL, increased protein tyrosine phosphoryla- particulate matter (PM2.5) in human
Korn SH, et al: Reactive nitrogen species tion, leading to contraction of guinea pig epithelial lung cells (L132) in culture.
and cell signaling: implications for death trachea. Toxicol Appl Pharmacol 2006; Appl Toxicol 2007;27:284–290.
or survival of lung epithelium. Am J 210:47–54. 18 Lee OH, Jain AK, Papusha V, et al: An
Respir Crit Care Med 2002;166:S9–S16. 15 Wu W, Graves LM, Jaspers I, et al: Acti- autoregulatory loop between stress sen-
12 Peluso M, Merlo F, Munnia A, et al: vation of the EGF receptor signaling sors INrf2 and Nrf2 controls their cellu-
32 pathway in human airway epithelial cells lar abundance. J Biol Chem 2007;282:
P-post-labeling detection of aromatic
adducts in the white blood cell DNA of exposed to metals. Am J Physiol 1999; 36412–36420.
nonsmoking police officers. Cancer Epi- 277:L924–931. 19 Ciencewicki J, Trivedi S, Kleeberger SR:
demiol Biomarkers Prev 1998;7:3–11. 16 Treisman R: Regulation of transcription Oxidants and the pathogenesis of lung
13 Bolton JL, Trush MA, Penning TM, et al: by MAP kinase cascades. Curr Opin Cell diseases. J Allergy Clin Immunol 2008;
Role of quinones in toxicology. Chem Biol 1996;8:205–215. 122:456–468.
Res Toxicol 2000;13:135–160. 20 Lee JM, Li J, Johnson DA, et al: Nrf2, a
multi-organ protector? FASEB J 2005;19:
1061–1066.

Mariangela Macchione, PhD


Department of Pathology, Laboratory of Experimental Air Pollution, Laboratory of Medical Investigation 05
Faculdade de Medicina da Universidade de São Paulo
Av. Dr. Arnaldo 455, São Paulo 01246-903 (Brazil)
Tel./Fax +55 11 3068 0072, E-Mail mmacchione@lim05.fm.usp.br

Air Pollution, Oxidative Stress and Pulmonary Defense 237


Section Title in Pulmonary Medicine
Organization
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 238–245

Mechanical Ventilation in Disaster Management


Richard Bransona ⭈ Thomas C. Blakemana ⭈ Dario Rodriquezb
a
Division of Trauma and Critical Care, University of Cincinnati, and bCincinnati CSTARS, United States Air Force,
University Hospital, Cincinnati, Ohio, USA

Abbreviations cess of the space to care for them and devices to


AARC American Association for Respiratory Care
provide ventilatory support [2–4].
ARDS Adult respiratory distress syndrome
CMV Continuous mandatory ventilation
FiO2 Inspired oxygen concentration History
HMEs Heat and moisture exchangers
MCRF Mass casualty respiratory failure History’s most instructive presentation of MCRF
PEEP Positive end-expiratory pressure occurred in the 1950s during the European po-
PPE Personal protective equipment liomyelitis epidemic. During the summer of 1952
SARS Severe acute respiratory syndrome
the hospital owned five ventilators, while treat-
ing 100 patients requiring mechanical ventila-
tion. This ‘surge’ of patients, relative to available
Recent history provides compelling tales of nat- ventilators, has not been seen since. Lassen and
ural disasters, the threat of terrorism, and out- colleagues [5] devised a clever solution to their
breaks of severe febrile respiratory illness includ- problem. Instead of negative pressure ventila-
ing H1N1 influenza and SARS. These threats tion they performed tracheostomy and enlisted
have focused healthcare planners, hospitals, and medical students to perform manual ventilation
communities on how to care for large numbers of in 4-hour shifts. Using a non-self-inflating bag
critically ill patients. Planning requires not only and carbon dioxide absorber, they were able to
space for the care of patients, but equipment and use very low flows of oxygen to sustain ventilation
staff [1]. MCRF is defined as an event resulting in and oxygenation. Interestingly, manual ventila-
patients requiring mechanical ventilation in ex- tion was also used following Hurricane Katrina,
when electricity failed at Charity Hospital in New
Orleans [6].
In 2009, while the world awaited an antici-
pated H5N1 (avian flu) outbreak, a novel H1N1
The work is solely that of the authors and does not reflect the official
position of the US Air Force, Department of Defense or US
virus originated in Mexico. The resulting pan-
Government. demic taxed ICUs around the world with severe
respiratory failure in pediatrics, obese, and obstet- Area Affected. In an explosion or fire, the af-
ric patients. The H1N1 epidemic is a lesson in the fected area is usually finite and managed locally.
unpredictability of viruses. The severity of ARDS A larger natural event may affect greater numbers
associated with H1N1 spurred a renewed interest of patients and result in damage to hospitals and
in rescue therapies for refractory hypoxemia [7]. transportation systems. In these instances, criti-
cally ill victims at the scene are likely to expire.

The Threat
Chemical Weapons
In the USA, the Department of Homeland Security
National Planning Guidelines are intended to co- Injuries following chemical weapons exposure
ordinate and prioritize emergency preparedness vary with the agent. Chemical agents are classi-
efforts at all response levels. Contained within the fied as lung-damaging agents, blood agents, blis-
Guidelines are 15 National Planning Scenarios, ter agents and nerve agents. These agents include
and at least two-thirds of these may result in MCRF chlorine, phosgene, and ammonia, all of which
[8]. The relevant scenarios are reviewed below. are commonly used in industrial processes and
readily available. Mustard gas is perhaps the best
known blister agent and cyanide the most likely
Traumatic Injury blood agent.
Expected Number of Victims. Under the appro-
Traumatic injury may result on a local level from priate environmental conditions, population den-
fire, explosion, or terrorist attack typically re- sity, and dispersion, chemical agents may result
sulting in <100 casualties. The experience from in thousands of victims. Despite this prediction
Israel with public explosions suggests that most however, to date the number of victims has been
incidents result in 20–30 casualties with half be- in the hundreds and the number of victims re-
ing hospitalized, half admitted to an ICU and the quiring mechanical ventilation has been less than
majority of those for life-saving mechanical ven- a dozen.
tilation [8]. Time from Injury to Need for Mechanical
Expected Number of Victims. In a local explo- Ventilation. The time until respiratory failure
sion or fire, typically <100 victims require hospi- requires mechanical ventilation varies with the
talization and fewer require mechanical ventila- agent and exposure. Pulmonary agents can cause
tion. In the instance of an earthquake or flood, sudden death as a result of laryngeal obstruction
many more victims may be expected, although and severe respiratory failure days after exposure.
the severity of injury is likely to be lower. Nerve agents causing paralysis may require venti-
Time from Injury to Need for Mechanical lation at the scene.
Ventilation. The severity of trauma may require Pathophysiology. Chemical weapons enter the
immediate airway management and ventilation body through the respiratory system and skin.
while others only require ventilation after opera- Blistering agents and choking agents result in
tive repair. bronchospasm and over time ARDS. Cyanide
Pathophysiology. Traumatic injuries result- poisons mitochondria and prevents cellular res-
ing in a need for mechanical ventilation include piration resulting in death from cellular hypoxia.
closed head injury, hemothorax, pneumothorax, Nerve agents result in flaccid paralysis and ap-
pulmonary contusion, flail chest, traumatic am- nea, but also produce significant bronchorrhea
putation, blood loss, and blast injury. and bronchospasm. So while patients exposed to

Mechanical Ventilation in Disaster Management 239


nerve agents may have normal lung compliance, entire regions depending on the length of the in-
airway resistance may be elevated. cubation period and the continued presence of
Area Affected. Optimum effectiveness of these the contagion. In the case of pandemic flu, entire
agents as a weapon includes exposure of a large portions of a country may be affected.
number of victims in a closed space. As such, these
exposures are limited to a well-defined area.
Planning for Mass Casualty Respiratory
Failure
Epidemics and Febrile Respiratory Illness
Staffing. Any MCRF scenario will result in an ex-
Epidemics and febrile illness may result from ponential need for mechanical ventilation and will
both natural and man-made causes. SARS and dramatically increase the need for ICU nurses, re-
pandemic flu are good examples resulting in ill- spiratory therapists, and physicians. Each hospital
ness around the world in a short time frame. and public health agency should have a plan for
Anthrax and botulism exposure are most like- support of healthcare workers and a disaster plan.
ly the result of bioterrorism although botulism Equipment is of no use if skilled caregivers are not
poisoning can occur from improperly preserved available to operate it [9–11].
foods. Anthrax, caused by Bacillus anthracis, has Personal Protective Equipment. Several of the
been used in the USA as a weapon, infecting 22 febrile respiratory illnesses associated with MCRF
people and killing 5. Anthrax is however not are highly contagious. PPE is a critical component
contagious. of mass casualty care in this setting. Availability
Expected Number of Victims. Epidemics can in- of PPE is important, as it allows caregivers to feel
fect people from all over the world affecting tens safe during patient interactions. In the absence of
of thousands of people. Weaponized botulism sufficient PPE, concerns about personal health
and anthrax have the ability to infect large num- may lead to employees avoiding the workplace.
bers of patients as well, but to date this has not Training caregivers in the use of PPE is also critical.
occurred. The risk of secondary transmission is likely greater
Time from Injury to Need for Mechanical in the ICU as a result of the number of interven-
Ventilation. Epidemics are likely to result in the tions causing aerosolization of infectious mate-
greatest number of casualties and typically the rial. Procedures such as endotracheal intubation,
time from exposure until respiratory failure de- open-circuit suctioning, and bronchoscopy are as-
velops is prolonged (days to weeks). In these cas- sociated with increased risk of transmission. The
es, patients may seek relief of early symptoms pri- use of filters in the ventilator circuit on the inspired
or to progression to respiratory failure. and/or expired side or inside an HME may make
Pathophysiology. Pandemic flu and SARS have sense but there is not data to support this practice.
caused severe ARDS requiring rescue therapies If filters are used, increased resistance may result in
for refractory hypoxemia. Botulism results in overdistension. Ventilators that compress room air
neuromuscular ventilatory failure from paralysis should have a filter on the inspiratory inlet.
and may require prolonged support. Anthrax re- Oxygen. Liquid oxygen is readily available at
sults in hemorrhagic mediastinitis, hemoptysis, most hospitals. Under normal circumstances, a
sepsis, profound hypoxemia, and acute respira- hospital has reserves to operate for 3 weeks. A typ-
tory failure. ical liquid system has 6,000–9,000 gallons of liq-
Area Affected. Natural epidemics and bioter- uid oxygen which evaporates to nearly 30,000,000
rorism agents in this class have the ability to infect gaseous liters of oxygen. Ritz and Privitera [12]

240 Branson ⭈ Blakeman ⭈ Rodriquez


estimated that their 500-bed hospital used 1.5 These data provide some standard requirements
million liters of oxygen per day. During a disaster, for the functional performance of ventilators for
oxygen conservation can be helpful. This includes use in MCRF. We could suggest that ventilators
use of reservoir or pendant cannulas, turning off for MCRF should be capable of setting PEEP from
flow to manual resuscitators, switching from heat- 6 to 13 cm H2O, FiO2 from 0.35 to 0.75, and a
ed aerosols to HMEs, and accepting lower levels minute ventilation of 10–18 l/min. Assuming pre-
of oxygen saturation in patients. dicted body weights of 62–90 kg, tidal volumes
Disposables. A frequently overlooked aspect of 375 up to 540 ml (6 ml/kg) should be capable
of MCRF are disposables. Ventilators require cir- of being set. These would be minimum require-
cuits, HMEs or humidifiers, and suction cathe- ments. Clearly some patients will require PEEP up
ters. Oxygen requires delivery tubing, cannulas, to or >20 cm H2O. Ventilators which are unable to
masks, and other appliances. In keeping with cost produce these settings at a minimum are not suit-
containment, these devices are commonly kept at able for in hospital MCRF.
a level sufficient for several weeks of supply. In Ventilator Performance Characteristics. Opera-
MCRF, most experts suspect that the disposables tional characteristics of ventilators for MCRF
will be among the first supplies to run out. Re- have been published by the AARC [13]. Some
use of circuits has been suggested, but this should explanation and clarification of these character-
only be considered as a last resort. istics are in order [14, 15]. Table 1 lists the desir-
able characteristics of ventilators for MCRF. The
optimal ranges for operation remain to be deter-
Ventilators mined. Characteristics of ventilators for MCRF
which are more difficult to quantify are clearly just
Ventilators may be needed following a disaster in as important. A ventilator for MCRF should be
three distinct environments. In the field to move rugged, portable, withstand shock and vibration,
patients from the scene of an accident to defini- and continue operation if dropped. Portability is
tive care, between facilities (decompressing a lo- important and ideally devices would be well ‘por-
calized event), and in-hospital care of critically ill table’. A weight of <10 kg is often a goal. In our
and injured patients. The following discussion re- minds, a portable device is one that a caregiver
views the requirements of mechanical ventilators can pick up with one hand and move it without
used for surge capacity in hospitals for definitive difficulty.
care. A ventilator for MCRF should have a low gas
Evidence-based management of the patient consumption. Equally important is battery life.
with ARDS is based on the results of the ARDSnet Battery life is affected by age of the battery, tem-
trial. The principles of ARDS management are perature, and charging history regardless of the
straightforward: (a) low tidal volume (6–8 ml/ ventilator. Ventilator characteristics which de-
kg of predicted body weight based on height); (b) crease battery life include the mode of ventila-
ability to give a constant tidal volume; (c) plateau tion, the FiO2, and PEEP. Patient characteristics
pressure ≤30 cm H2O; (d) stable FiO2 from 0.21 can also affect battery life, the greater the load the
to 1.0; (e) CMV – maintain minute ventilation; shorter the duration of operation.
(f) PEEP to prevent alveolar collapse and lung The ventilator should be easy to trigger and
injury. have an acceptable work of breathing. Most cur-
On day 1 of the ARDSnet trial, patients re- rent-generation portable ventilators meet this re-
ceived a PEEP of 6–13 cm H2O, an FiO2 of 0.35– quirement. Cost should be less than USD 10,000.
0.75, and a minute ventilation of 10–16 l/min. The ventilator should be intuitive and easy to use.

Mechanical Ventilation in Disaster Management 241


Table 1. Suggested performance characteristics of ventilators for MCRF

Characteristic Rationale Characteristics

mandatory desirable

FDA-approved Natural disasters, pandemics, Ventilate 10-kg Ventilate 5-kg


for adults and and chemical/bioterrorism will patient patient
pediatrics also affect children

Ability to operate The redundancy for electrical power Operate without 50 Operate with or
without 50 psig in hospitals far exceeds oxygen stores psig input without 50 psig
compressed gas and redundancy FiO2 from 0.21 to 1.0 input alone
In the absence of high-pressure
oxygen, low flow oxygen from a
flowmeter can be used to increase
FiO2
Battery life of 4 h Allow for transport from facility to 4 h of operation at >4 h operation at
of greater facility Provide continuous support nominal settings nominal settings
during intermittent power failure

Constant volume Meet guidelines for tidal volume delivery Volume control Pressure control and
delivery as dictated by ARDSnet protocol ventilation volume control
Reduce potential for ventilator-induced (350–600 ml) ventilation
lung injury
Provide age-appropriate settings

Mode: Meet ARDSnet guidelines CMV CMV,


CMV Assure minimum ventilation in a intermittent mandatory
situation of multiple patients and a ventilation, and
shortage of caregivers pressure support

PEEP Meet ARDSnet guidelines Adjustable from Adjustable from 5 to 20


Prevent ventilator-induced 5 to 15 cm H2O cm H2O
lung injury
Reverse hypoxemia
Separate controls Meet ARDSnet guidelines Respiratory rate from 6 Respiratory rate from 6
for respiratory rate Assure minute ventilation in to 35 breaths/min to 75 breaths/min (for
and tidal volume apneic patients pediatrics)

Monitor airway Meet ARDSnet guidelines Provide Monitor peak Monitor plateau
pressures and assessment of patient’s lung compliance inspiratory pressure and pressure and patient
tidal volume Patient safety – prevent overdistension delivered tidal volume tidal volumes

Appropriate Patient safety Alarms for Alarms for


alarms Improve ability to monitor large circuit disconnect high tidal
numbers of patients with High airway pressure volume in
reduced staff Low airway pressure pressure
(leak) modes
Loss of electric power Low minute
Loss of high pressure ventilation
source gas Remote alarms

242 Branson ⭈ Blakeman ⭈ Rodriquez


Fig. 1. A sophisticated portable ventilator capable of Fig. 2. Automatic resuscitator with limited capability.
ventilating critically ill patients. This device has an inter- These devices must be used with a caregiver present at
nal air source (compressor) and a suite of monitoring and each patient.
alarms allowing it to be used in a number of situations.

Ventilators. Classification of mechanical ven- 100% source gas or a lower concentration with the
tilators is a complex task. For the purposes of use of a Venturi. These devices are inexpensive,
describing ventilators for MCRF, operation and but fail to meet the demands of the patient with
application leads to description of types of ven- ARDS and are not suitable for stockpiling to treat
tilators which might be used based on character- MCRF. The best that can be said is that these de-
istics. This description includes automatic resus- vices are better than no support at all. An example
citators, EMS ventilators, pneumatically-powered of an automatic resuscitator is shown in figure 1.
portable ventilators, electrically-powered porta- EMS Portable Ventilators. An EMS portable
ble ventilators, and full-feature ICU ventilators. ventilator is used in patient transport, typically in
Automatic Resuscitators. An automatic resusci- emergency care via ambulance. These devices are
tator is designed to replace the need for hand-bag- more reliable, rugged, and have greater function-
ging. These devices are predominantly pneumat- ality than automatic resuscitators. The functional-
ically-powered and pressure-cycled. Automatic ity and cost in this group is variable. Some devices
resuscitators have few to no alarms, cannot pro- set tidal volume and respiratory rate via a single
vide a constant tidal volume, cannot set rate and control. Others have separate controls for both
tidal volume separately, and commonly provide settings. PEEP is usually supplied by an external

Mechanical Ventilation in Disaster Management 243


Table 2. Sources of additional ventilators for a MCRF scenario

Affected Cancel elective surgeries Number of anesthesia machines is


hospital Repurpose anesthesia limited
workstations as mechanical If the duration of mechanical
ventilators and ICU monitoring ventilation is prolonged, anesthesia
(during non-trauma disasters) machines will be needed when surgeries
and other procedures are reinitiated

Unaffected Redistribution of available There are few extra available


hospital equipment from unaffected ventilators at most hospitals even
hospitals to those in need during usual conditions
Delayed situational awareness may
reduce willingness of ‘unaffected’
hospitals to share equipment

Mechanical Provision of additional The same company may have


ventilator rental ventilators by a rental contracts with a number of affected
services company hospitals, so the total number of
additional ventilators may be limited
Logistical delays may be encountered
when sending ventilators from distant
geographic areas

Strategic Deployment of mechanical Delay in distribution since most states


National ventilators to states or still have limited capacity to distribute
Stockpile cities in need equipment from the Strategic National
Stockpile
Unclear how distribution will be
prioritized when multiple hospitals are
requesting ventilators at the same time

valve. FiO2 is commonly 100% source gas or a sin- Sophisticated Portable Ventilators (Electronic-
gle lower concentration with use of an air entrain- ally-Powered). These devices are often used
ment system. Monitoring and alarms are limited. in homecare and for in-hospital transport.
Sophisticated Portable Ventilators (Pneuma- Electronically-powered, sophisticated portable
tically-Powered). Sophisticated pneumatically- ventilators meet the performance characteristics
powered, portable ventilators have the ability to required of a ventilator for MCRF. There is some
provide CMV and intermittent mandatory ven- significant difference in weight of these devices
tilation, set PEEP, have a low work of breathing, (5–15 kg). Battery life and gas consumption vary
and allow separate control of tidal volume and depending on the driving system of the ventila-
respiratory rate. These devices meet most of the tor. Figure 2 depicts a commonly used portable
performance characteristics for MCRF. The limi- ventilator.
tations of these devices surround the pneumatic Critical Care Ventilators. Critical care venti-
power source. These devices cannot operate in the lators are capable of managing all types of respi-
absence of a 50-psig gas source. FiO2 is typically ratory failure. These devices have not been rec-
limited to 100% source gas which wastes oxygen. ommended for MCRF due to the large size, cost
Few alarms are also a weakness. (USD 30,000+), and complexity.

244 Branson ⭈ Blakeman ⭈ Rodriquez


Non-Invasive Ventilators. Non-invasive venti- most patients with the best possible outcome. All
lation is a standard of care for respiratory failure patients receive care in a mass casualty situation,
in the patient with chronic obstructive pulmo- even if it is comfort care.
nary disease, under normal circumstances. The
use of non-invasive ventilation in MCRF howev-
er has significant limitations. In a MCRF situa- Conclusion
tion, we suggest the repurposing of non-invasive
ventilators for use as invasive ventilators. Table 2 MCRF represents a significant concern for health-
lists the possible sources of additional ventilators care systems and governments around the world.
during MCRF. The best solution is thoughtful planning and coop-
Triage. No discussion of MCRF is complete eration between all the shareholders. Despite our
without mentioning triage of ventilators. This inability to predict when an event may occur and
is an ethical dilemma which the modern world how many patients will require mechanical ventila-
has not yet had to face. In MCRF, all patients will tion, we must plan using best evidence. Ventilators
receive care based on the likelihood of survival. purchased for MCRF must meet the demands of
These systems are being developed by national so- the patient and the skills of the operator.
cieties to allow the most good to be done for the

References
1 Hotchkin DL, Rubinson L: Modified crit- 6 DeBoisblanc BP: Black Hawk, please 11 Daugherty E, Branson RD, Desai A,
ical care and treatment space consider- come down: reflections on a hospital’s Rubinson L: Infection control in mass
ation for mass casualty critical illness struggle to survive in the wake of Hurri- respiratory failure: preparing to respond
and injury. Respir Care 2008;53:67–74. cane Katrina. Am J Respir Crit Care Med to H1N1. Crit Care Med
2 Rubinson L, Nuzzo JB, Talmor DS, 2005;172:1239–1240. 2010;38(suppl):e103–e109.
O’Toole T, Kramer BR, Inglesby TV: 7 Tang JW, Shetty N, Lam TT: Features of 12 Ritz RH, Privitera JE: Oxygen supplies
Augmentation of hospital critical care the new pandemic influenza A/ during a mass casualty situation. Respir
capacity after bioterrorist attacks or epi- H1N1/2009 virus: virology, epidemiol- Care 2008;53:215–224.
demics: recommendations of the Work- ogy, clinical and public health aspects. 13 AARC Guidelines for Acquisition of
ing Group on Emergency Mass Critical Curr Opin Pulm Med 2010;16:235–241. Ventilators to Meet Demands for Pan-
Care. Crit Care Med 2005;33:2393–2403. 8 Homeland Security: National Prepared- demic Flu and Mass Casualty Incidents
3 Rubinson L, O’Toole T: Critical care dur- ness Guidelines, October 2007. Accessed American Association Accessed April
ing epidemics. Crit Care 2005;9:311– April 24, 2010 (http://www.dhs.gov/xli- 14, 2008 (http://www.aarc.org/head-
313. brary/assets/National_Preparedness_ lines/ventilator_acquisitions/vent_
4 Rubinson L, Branson RD, Pesik N, Tal- Guidelines.pdf). guidelines.pdf).
mor D: Positive-pressure ventilation 9 Muskat P: Mass casualty chemical expo- 14 Branson RD, Johannigman JA, Daugh-
equipment for mass casualty respiratory sure and implications for respiratory erty EL, Rubinson L: Surge capacity
failure. Biosecur Bioterror 2006;4:183– failure. Respir Care 2008;53:58–63. mechanical ventilation. Respir Care
194. 10 Hanley ME, Bogdan GM: Mechanical 2008;53:78–90.
5 Lassen HCA: Management of Life- ventilation in mass casualty scenarios. 15 Daugherty EL, Branson RD, Rubinson L:
Threatening Poliomyelitis. Copenhagen, Augmenting staff: project XTREME. Mass casualty respiratory failure. Curr
1952–1956, with a Survey of Autopsy Respir Care 2008;53:176–188. Opin Crit Care 2007;13:51–56.
Findings in 115 cases. Edinburgh, Liv-
ingstone, 1956.

Richard Branson, MS, RRT


Division of Trauma and Critical Care, University of Cincinnati
231 Albert Sabin Way, Cincinnati, OH 45267-0558 (USA)
Tel. +1 513 558 6785, Fax +1 513 558 3747
E-Mail Richard.branson@uc.edu

Mechanical Ventilation in Disaster Management 245


Section Title in Pulmonary Medicine
Organization
Esquinas AM (ed): Applied Technologies in Pulmonary Medicine. Basel, Karger, 2011, pp 246–249

Postoperative Intensive and Intermediate Care?


Charles Weissman
Department of Anesthesiology and Critical Care Medicine, Hadassah-Hebrew University Medical Center, Hebrew University-
Hadassah School of Medicine, Jerusalem, Israel

Contemporary hospital medicine must cope with adequate facilities for postoperative care [3]. This
increasing numbers and proportions of very ill situation requires provisions for graduated lev-
patients requiring greater nursing and medical els of care (intensive, intermediate, routine floor
care than afforded on regular hospital floors. To care). The importance of graduated care was em-
care for such patients requires clinical areas with phasized in a report from hospitals without in-
increased nurse:patient ratios and specialized termediate care units so that postoperative inter-
medical personnel. Therefore, hospitals require mediate care patients were sent to regular wards.
increasing numbers of ICUs (intensive therapy The greater care requirements of such patients ad-
units), intermediate care units (high dependency versely affected the care of less ill patients [4]. The
units) and postoperative recovery rooms (post- advantages of postoperative intermediate care was
anesthesia care units). This situation has placed demonstrated by comparing major abdominal
hospitals in the perpetual quandary of how many surgery patients who were cared for on a general
and what proportion of beds should be intensive surgical ward because the hospital lacked an inter-
care, intermediate care and regular beds [1]. mediate unit with those who were initially man-
aged in an intermediate unit. Intermediate care
resulted in fewer cardiorespiratory complications,
Analysis of Main Topics without differences in mortality, but with a trend
towards shorter hospital stays [5, 6]. Alternately,
Increasingly complex surgery is being performed the introduction of a 4-bed intermediate unit in
on patients suffering from preexisting chronic dis- a teaching hospital failed to reduce postoperative
eases and who are elderly. Such patients frequent- serious events or mortality, nor did it decrease the
ly require enhanced care after surgery to monitor, mean duration of hospitalization [7].
prevent and, if necessary, treat life-threatening Graduated postoperative care requires that
complications as soon as they appear. In the UK, specific criteria be established for equipping, staff-
surgical patients make up 60–70% of the ICU ing and admissions to each type of unit. Surgical
workload [2]. The British National Confidential ICUs generally care for four types of patients: (1)
Enquiry into Perioperative Deaths for 1992/1993 Those after extensive elective surgery (e.g. open
recommended that surgery not be performed on abdominal aneurysm repairs, anterior-posterior
physiologically compromised patients without spine fusion) and those with severe preexisting
diseases undergoing major to moderate elective physiological monitors and staffed by specially
surgery [8]. (2) Emergency surgery patients who trained nurses but are not fully occupied during
have cardiopulmonary instability following sur- the evening and night. Among the disadvantages
gery. (3) Surgical patients who have cardiopulmo- of this arrangement is the need to discharge these
nary, septic or hemorrhagic deteriorations on sur- overnight patients early enough in the morning
gical floors or intermediate care units. (4) Trauma so as not to interfere with the elective surgical
victims admitted for non-operative management schedule.
(e.g. observation of splenic, renal or hepatic lac-
erations). Many institutions have subspecialty
ICUs to care for high volumes of specific patients. Discussion
The leading example is the cardiothoracic surgical
ICU which specializes in caring for postoperative The types of patients admitted to each kind of unit
cardiac and thoracic surgery patients. Such ICUs is institution-specific, i.e. depends on the patient
are unifocused and thus can optimize protocol population, number and types of ICU and inter-
and care plans for such patients. Neurosurgical mediate units, the number of such beds, the mon-
and burn ICUs serve much the same purpose. itoring, nursing and medical capabilities of each
Specialized units offload routine elective cardiac unit, the nursing capabilities on regular floors (i.e.
surgery and neurosurgery patients from the main nurse:patient ratio) and institutional administra-
surgical ICU allowing it to focus on caring for the tive and medical policies. For example, in our in-
remaining heterogeneous population of critically stitution, 12% of postoperative patients were ad-
ill surgical patients many of whom require exten- mitted directly to an ICU or intermediate unit
sive physiological monitoring, multiple diagnos- [10].
tic and therapeutic procedures and much medical A recent prospective observational study of
and nursing care. 1,233 adult postoperative patients transferred af-
Intermediate care units differ from ICUs by ter a short recovery room stay to a floor bed or
caring for lower acuity patients. They, thus, have the ambulatory surgery unit and 1,883 patients
higher nurse:patient ratios (ICU 1:1–1:2 vs. in- admitted to intermediate and ICU areas, exam-
termediate 1:4) and lower intensity of physician ined the effects of pre- and intraoperative factors
coverage. Surgical intermediate care units take a on receipt of postoperative ICU or intermediate
number of forms: (1) Postoperative/post-proce- care. There were distinct differences between the
dure units monitor (24–48 h) hemodynamically preoperative (preexisting systemic disease) and
stable patients after complex surgeries, patients intraoperative characteristics (complexity of sur-
with underlying cardiopulmonary disorders after gery and intraoperative care intensity) of postop-
moderate surgery and those receiving peripheral erative ICU/intermediate care patients and those
arterial thrombolytic therapy [1, 9]. (2) Step-down receiving routine floor care [11]. Moreover, there
units admit patients who no longer need ICU care were differences between elective and emergency
but require more intense nursing care than pro- surgery. Most patients undergoing elective opera-
vided on the surgical floors. (3) Mechanical ven- tions of the highest complexity (e.g. craniotomy,
tilator units care for patients requiring long-term cardiac surgery) received intermediate or ICU
mechanical ventilation. Alternately, the post-an- care including those without preexisting system-
esthesia care unit can be used as a postoperative/ ic illness (ASA class 1). Classic examples were
post-procedure intermediate unit for patients healthy teenagers undergoing elective orthog-
needing up to 24 h of such care. The rationale is nathic jaw surgery and anterior-posterior spine
that post-anesthesia care units are equipped with fusion for scoliosis. The former because they

Postoperative Intensive and Intermediate Care? 247


remained tracheally intubated overnight because Interestingly, 10% of the elective surgery pa-
of upper airway edema and the latter because tients unexpectedly received ICU or intermedi-
they were monitored for hemorrhage. Another ate care because of unanticipated intraoperative
example was elective neurosurgery patients with or immediate postoperative issues, such as up-
minimal (ASA class 2) underlying systemic dis- per airway problems or unexpected hemorrhage.
eases who underwent craniotomies for brain tu- Similar observations have been made by others,
mor excision. Alternately, patients undergoing who consider unexpected admissions a quality in-
less complex major elective surgery often tended dicator [12].
to receive intermediate or ICU care if they had
significant preexisting systemic disorders (ASA
classes 3 and 4). Recommendations
Emergency surgery patients differed substan-
tially from elective surgery ones with a greater • Modern in-patient surgical care systems
proportion receiving ICU rather than intermedi- require graduated levels of care (intensive,
ate care. ASA class was not significantly associ- intermediate and floor care).
ated with receipt of ICU rather than intermediate • Postoperative patients are major consumers
care. This was true even when trauma patients, of ICU and intermediate care as surgery is
many of whom were ASA classes 1 and 2, were increasingly performed on the elderly and on
removed from the analysis. Like the elective sur- patients with significant preexisting illness.
gery patients, high intraoperative care intensity • Receipt of postoperative intermediate and
and postoperative mechanical ventilation were intensive care is associated with distinct
associated with the receipt of ICU rather than patterns of both preoperative and intra-
intermediate care. This is in contrast to the elec- operative factors.
tive patients were ASA class, operative complexity • Postoperative mechanically ventilation is
and age were also so associated. This suggests that the major indication for ICU rather than
the intensity of non-surgical care received during intermediate care.
emergency surgery and the decision to leave the • The ASA class and complexity of the surgery are
patient mechanically ventilated after surgery re- important factors determining postoperative
sulted in ICU rather than intermediate admis- intensive/intermediate care after elective sur-
sion. Therefore, the severity of the acute illness gery. After emergency surgery the intensity of
or trauma dictates the need for significant inter- intraoperative interventions, and not surgical
ventions during surgery (e.g. blood products, he- complexity, is a major determining factor.
modynamic monitoring) resulting in a decision
to leave the patient mechanically ventilated after
surgery.

References
1 Heller J, Murch P: Development in ser- 2 Cuthbertson BH, Webster NR: The role 3 Campling EA, Devlin HB, Hoile RW, et
vice provision. Making major elective of the intensive care unit in the man- al: The report of the National Confi-
surgery happen. The development of a agement of the critically ill surgical dential Enquiry into Perioperative
postoperative surgical unit. Nurs Crit patient. J R Coll Surg Edinb 1999;44: Deaths 1992/1993. London, NCEPOD,
Care 2008;13:97–104. 294–300. 1995.

248 Weissman
4 Coggins RP: Delivery of surgical care in 7 Bellomo R, Goldsmith D, Uchino S, et al: 10 Weissman C, Klein N: The importance of
a district general hospital without high A before and after trial of the effect of a differentiating between elective and
dependency unit facilities. Postgrad Med high-dependency unit on post-operative emergency postoperative critical care
J 2000;76:223–226. morbidity and mortality. Crit Care patients. J Crit Care 2008;23:308–316.
5 Jones HJ, Coggins R, Lafuente J, et al: Resusc 2005;7:16–21. 11 Weissman C, Klein N: Who receives
Value of a surgical high-dependency 8 Cavaliere F, Conti G, Costa R, et al: postoperative intensive and intermedi-
unit. Br J Surg 1999;86:1578–1582. Intensive care after elective surgery: a ate care? J Clin Anesth 2008;20:263–
6 Armstrong K, Young J, Hayburn A, et al: survey on 30-day postoperative mortal- 270.
Evaluating the impact of a new high ity and morbidity. Minerva Anestesiol 12 Haller G, Myles PS, Langley M, et al:
dependency unit. Int J Nurs Pract 2003; 2008;74:459–468. Assessment of an unplanned admission
9:285–293. 9 Angel D, Sieunarine K, Finn J, et al: to the intensive care unit as a global
Comparison of short-term clinical post- safety indicator in surgical patients.
operative outcomes in patients who Anaesth Intensive Care 2008;36:190–
underwent carotid endarterectomy: 200.
intensive care unit versus the ward high-
dependency unit. J Vasc Nurs
2004;22:85–90.

Charles Weissman, MD
Department of Anesthesiology and Critical Care Medicine
Hadassah-Hebrew University Medical Center, Hebrew University-Hadassah School of Medicine
Jerusalem 91120 (Israel)
Tel. +972 2 677 7269, Fax +972 2 642 9392, E-Mail Charles@hadassah.org.il

Postoperative Intensive and Intermediate Care? 249


Author Index

Adam, A.K. 178 Hashizume, M. 185 Robson, A.G. 132


Aly, H. 151 Herff, H. 10 Rocco, G. 89
Anantham, D. 84 Herlihy, J.P. 19 Rodriquez, D. 238
Herting, E. 205
Baltopoulos, G.J. 163 Salati, M. 89
Barchfeld, T. 6 Kar, B. 19 Saldiva, P.H.N. 217
Bayrakci, B. 114 Kerl, J. 6 Saraiva-Romanholo, B.M. 223
Beck, J. 1 Koehler, D. 6 Scheuch, G. 60, 67, 77
Bergese, S.D. 102 Kolobow, T. 151 Schmölzer, G.M. 53
Berra, L. 151 Korupolu, R. 192 Shindo, Y. 172
Blakeman, T.C. 238 Siekmeier, R. 60, 67, 77
Boussignac, G. 51 Lepre, S. 192 Sinderby, C. 1
Branson, R. 238 Loyalka, P. 19 Siyue, M.K. 84
Bueno Garcia, M.L. 231 Luyt, C.E. 168 Soubani, A.O. 178
Swanson, J.M. 156
Calkovska, A. 205 Macchione, M. 231
Carvalho-Oliveira, R. 217 Manikandan, S. 96 Tacconi, F. 107
Connolly, T. 19 Matsuoka, Y. 185 Tao, L. 210
Mineo, T.C. 107 Tehrani, F.T. 28, 39
Davies, M.W. 15 Morley, C.J. 53 Truong, A. 192
De Keulenaer, B.L. 136 Murthy, J.N. 46
Dellweg, D. 6 Myrianthefs, P.M. 163 Vajrala, G. 192
Donnellan, M.E. 126 Vitacca, M. 119
Dunster, K.R. 15 Nakagawa, N.K. 217, 223
Nakao, M. 223 Weissman, C. 246
Epstein, S.K. 35 Needham, D.M. 192 Wood, G.C. 156
Esquinas, A.M. XIII Neema, P.K. 96
Evagelopoulou, P. 163 Zaitsu, A. 185
Papaioannou, V. 145 Zanni, J.M. 192
Fernandez, R.F. 202 Pavlidis, I. 46 Zhou, W. 210
Pneumatikos, I. 145
Gifford, J.M. 192 Pompeo, E. 107
Goto, D.M. 223 Puente, E.G. 102
Gregoric, I. 19
Rathod, R.C. 96
Hasegawa, Y. 172 Ries, A.L. 197

250
Subject Index

Abdominal compartment syndrome, see Intra- lung function testing 223, 224
abdominal pressure nasal lavage 227, 228
AbViser kit, intra-abdominal pressure nitric oxide in exhaled air 225, 226
measurement 137 saccharin transit time test 228
Acute lung collapse, see Cardiopulmonary bypass sputum induction 224, 225
Acute lung injury, intra-abdominal hypertension types and populations at risk 217, 218
142 Air travel
Acute respiratory distress syndrome chronic lung disease patients
capnography 127, 128 guidelines 133
chest wall compliance decrease 142, 143 oxygen supplementation 134
extracorporeal membrane oxygenation 22 preflight assessment 133, 134
intra-abdominal hypertension 142 recommendations 134
surfactant physiology at high altitude 132, 133
changes 205, 206 AKITA devices 64, 65
replacement therapy Anesthesia
administration 206, 207 epidural, see Thoracic epidural anesthesia
dosing 207 intraoperative magnetic resonance imaging, see
metabolism 208 Magnetic resonance imaging
outcomes 208, 209 α1-Antitrypsin, inhalation therapy 67
preparations 207, 208 Atelectasis
timing 206 intensive care unit readmission 185–190
ventilation patterns 207 intra-abdominal hypertension 141
Adaptive support ventilation 30, 31 Automated mechanical ventilation, see also specific
Air pollution techniques
annual concentration ranges 219 advanced technologies 29, 30
dispersion scales 219, 220 importance 29
gas pollutants and measurement 220, 221 overview 28, 29
oxidative stress recommendations for improvement 34
endogenous oxidants 233, 234 Automatic tube compensation 30
exogenous oxidants 231, 232
Nrf2 response 236 Bed rest, see Intensive care unit
pulmonary antioxidant defenses 234–236 Boussignac system 51, 52
signaling pathways 232, 233 Brain death, see Hyperbaric oxygen therapy
particulate matter 220 Bronchoalveolar lavage
respiratory system assessment culture and ventilation-associated pneumonia
exhaled breath condensate 226, 227 antibiotic therapy guidance 157–161

251
virus testing 169, 170 Dry powder inhaler 62, 63

Capnography Edema, see Lung edema


case reviews Edi 33
adult respiratory distress syndrome 127, 128 Endobronchial ultrasound
chronic obstructive pulmonary disease 127 linear ultrasound with transbronchial needle
neonates 128, 129 aspiration 85–87
pulmonary embolus 128 radial ultrasound applications 84, 85
respiratory distress non-intubated patient 128 recommendations 87, 88
end-tidal carbon dioxide and partial pressure End-tidal carbon dioxide, capnography gradient 126,
arterial carbon dioxide gradient 126, 127, 129, 127, 129, 130
130 Erythropoietin, inhalation therapy 73, 74
Carbon monoxide, see Air pollution Exhaled breath condensate, air pollution impact
Cardiopulmonary bypass, acute lung collapse assessment 226, 227
mechanisms 96, 97 Extracorporeal membrane oxygenation
pathophysiology of cardiovascular and respiratory acute respiratory distress syndrome and
complications 97, 98 respiratory failure 22
prevention 100 blood flow rate 24
recommendations 100, 101 complications 25
re-expansion 98, 99 congestive heart failure 22, 23
weaning 99, 100 historical perspective 19–22
Cardiopulmonary resuscitation prospects 25
Boussignac system 51, 52 pumps 21
guidelines 51 sedation 25
Catalase, antioxidant defense 235, 236 team 23, 24
Cerebral blood flow vascular access 20, 21, 24
hypercapnia effects in neonates 212, 213 wean trial 24
hypocapnia effects in neonates 210, 211
Chest wall, compliance 142, 143 Fc domain, inhalation therapy absorption
Chronic obstructive pulmonary disease enhancement 71, 72
air travel, see Air travel FLEX
capnography 127 flowchart of algorithm 40, 42
teleassistance for patients 122, 123 overview 40, 41
Clinical pulmonary infection score, ventilation- performance 43, 44
associated pneumonia resolution 164–166 recommendations 44
Community-acquired pneumonia, viral steps in procedure 40–43
infection 168–171 Foley manometer, intra-abdominal pressure
Computed tomography, pleural effusion 147 measurement 137–139
Congestive heart failure, extracorporeal membrane Forced expiratory volume in 1 s, air pollution impact
oxygenation 22, 23 assessment 224
Corticosteroids, post-extubation upper airway Forced vital capacity, air pollution impact
obstruction prevention 36–38 assessment 223, 224
Cuff leak volume 36–38
Cyclodextrins, inhalation therapy absorption Gram stain, ventilation-associated pneumonia
enhancement 71 antibiotic therapy guidance 157–161
Cyclosporin A, inhalation therapy 75 Granulocyte colony-stimulating factor, inhalation
therapy 67
Desoxyribonuclease, inhalation therapy 67 Granulocyte-macrophage colony-stimulating factor,
Diabetes, see Insulin inhalation therapy inhalation therapy 74

252 Subject Index


Growth hormone, inhalation therapy 67 macromolecules, see also Insulin inhalation
therapy
Healthcare-associated pneumonia absorption
clinical outcomes 173–175 barriers 67–70
definition 172, 173 factors affecting 70
diagnosis 175, 176 improvement 70–73
epidemiology 172, 173 prospects 75
pathogens 173, 174 safety 75
recommendations 177 stability 67
risk factors for potentially drug-resistant systemic therapies 73–75
pathogens 175 prospects 65
Heparin, inhalation therapy 67, 73 Inspiratory threshold device 11, 12
High-frequency jet ventilation 29 Insulin inhalation therapy
High-frequency oscillatory ventilation 29 absorption enhancers 78, 80
Home mechanical ventilation acceptance 82
costs 119 carriers 78
systems 119, 120 costs 82
teleassistance devices 79
benefits 122 historical perspective 77, 78
chronic obstructive pulmonary disease pharmacokinetics
outcomes 122, 123 normal individuals 80
overview 120, 121 pulmonary disease patients 81
recommendations 123, 124 smokers 81
Hyperbaric oxygen therapy prospects 82
indications 116 safety 81, 82
organ preservation from brain-dead donors Intensive care unit
acute cellular rejection 115 lung cancer patients, see Lung cancer
overview 114–116 mobilization
prospects 116, 117 benefits 194, 195
reperfusion injury 115 early mobilization goals 193
Hypercapnia, see Neonates feasibility 194
Hypocapnia, see Neonates incidence of physical medicine and
Hypovolemic shock rehabilitation therapy 192, 193
intrapulmonary pressure decrease and negative pathophysiology of bed rest and
side effects 12, 13 immobility 193
intrathoracic pressure effects on blood flow 10, safety 193, 194
11 pleural effusion management 148, 149
pressure controllers 12 postoperative care versus intermediate care unit
postoperative care 246–248
Immobility, see Intensive care unit readmission criteria for lung edema and
Inhalation therapy atelectasis 185–190
administration tracheostomy and ward mortality 202, 203
dry powder inhaler 62, 63 Interferon-α, inhalation therapy 67
metered dose inhaler 63, 64 Interleukin-2, inhalation therapy 74
nebulizer 64 Intra-abdominal pressure
novel devices 64, 65 body position effects 140
overview 62 hypertension
individualized controlled inhalation 65 acute lung injury 142
limitations 60, 61 acute respiratory distress syndrome 142

Subject Index 253


chest wall compliance decrease 142, 143 definition 238
compression atelectasis 141 historical perspective 238, 239
edema 142 planning
epidemiology 141, 142 disposables 242
respiratory mechanics 140, 141 oxygen 241, 242
measurement personal protective equipment 240
AbViser kit 137 staffing 240
Foley manometer 137–139 threats
overview 136, 137 chemical weapons 239, 240
Intraoperative magnetic resonance imaging, see epidemics 240
Magnetic resonance imaging traumatic injury 239
Intrathoracic pressure controller 12, 13 ventilator requirements and classification
Intrathoracic pressure, effects on blood flow 10, 11 241–245
Intraventricular hemorrhage Metered dose inhaler 63, 64
hypercapnia effects in neonates 212 Minimally invasive thoracic surgery, see Video-
hypocapnia effects in neonates 211 assisted thoracic surgery
Mitogen-activated protein kinase, oxidant-induced
Jet travel, see Air travel cell signaling 232, 233

Liposome, inhalation therapy absorption Nasal lavage, air pollution impact assessment 227, 228
enhancement 71 Nebulizer 64
Lung cancer Negative end-expiratory pressure, intrathoracic
intensive care unit pressure effects on blood flow 11
admission indications Neonates
cancer-related complications 179 capnography 128, 129
co-morbid illness 179, 180 hypercapnia
overview 178, 179 effects 212
treatment-related complications 179 permissive hypercapnia
outcomes and predictors 180–183 central nervous system disease 213, 214
recommendations 183 goals 213
mortality 178 respiratory disease 213
resection, see Video-assisted thoracic surgery hypocapnia effects
Lung collapse, see Cardiopulmonary bypass central nervous system 210, 211
Lung edema respiratory system 211, 212
intensive care unit readmission 185–190 Neurally adjusted ventilatory assist
intra-abdominal hypertension 142 patient-ventilator interaction 1, 2
principles 2, 3, 33
Magnetic resonance imaging, anesthesia during proportional assist ventilation
intraoperative imaging comparison 3, 4
electrocardiography 104 spontaneous breathing events 2
equipment 104, 105 Nitric oxide in exhaled air, air pollution impact
monitoring 104, 105 assessment 225, 226
recommendations 105 Nitrogen dioxide, see Air pollution
room design 102, 103 Non-invasive ventilation
safety 103, 104 rationale 6
tubing 104 TA mode
Mannequin teaching, see Simulation-based algorithm description 6–8
mannequin teaching clinical implications 8, 9
Mass casualty respiratory failure Nrf2, oxidative stress response 236

254 Subject Index


Open-heart surgery, see Cardiopulmonary bypass prevalence 35
Organ transplantation, see Hyperbaric oxygen therapy risk factors 35, 36
Oxidative stress, see Air pollution Proportional assist ventilation
Ozone, see Air pollution neurally adjusted ventilatory assist comparison 3, 4
patient-ventilator interaction 1, 2
Partial pressure arterial carbon dioxide gradient, principles 2, 31, 32
capnography gradient 126, 127, 129, 130 spontaneous breathing events 2
Particulate matter, see Air pollution Pulmonary arterial hypertension, hypocapnia effects
Peak inflation pressure, simulation-based mannequin in neonates 211
teaching 57–59 Pulmonary embolus, capnography 128
Perfluorocarbon immersion Pulmonary rehabilitation
adjunctive therapy 17, 18 definition 197, 198
skin gas exchange 15–17 indications 198
spontaneous liquid breathing 17 guidelines 198–201
Periventricular leukomalacia
hypercapnia effects in neonates 213 Respiratory function monitor, simulation-based
hypocapnia effects in neonates 211 mannequin teaching 55–58
Permissive hypercapnia, see Neonates
Peroxynitrite, air pollution induction 234 Saccharin transit time, air pollution impact
Personal protective equipment, mass casualty assessment 228
respiratory failure 240 Sepsis-related organ failure assessment, ventilation-
Pleural effusion associated pneumonia resolution 165
diagnosis Simulation-based mannequin teaching
chest film 146 mask hold and positioning techniques 56
computed tomography 147 peak inflation pressure 57–59
pleural fluid examination 147, 148 performance assessment 58
ultrasound 146, 147 positive end-expiratory pressure 57
etiology 146 positive pressure ventilation 53, 54, 56
management in intensive care unit 148, 149 respiratory function monitor 55–58
pathophysiology 145, 146 SmartCare 32, 33
Pneumonia, see Community-acquired pneumonia, Spontaneous breathing
Healthcare-associated pneumonia, Ventilation- events 2
associated pneumonia perfluorocarbon immersion 17
Pollution, see Air pollution Sputum induction 224, 225
Polycyclic aromatic hydrocarbons, air pollution 232 Sulfur dioxide, see Air pollution
Polysomnography Superoxide dismutase, antioxidant defense 236
sleep disordered breathing 46 Surfactant
thermal infrared imaging acute respiratory distress syndrome
integration with polysomnography 48 changes 205, 206
principles 47, 48 replacement therapy
prospects 47, 48 administration 206, 207
recommendations 49 dosing 207
Positive end-expiratory pressure metabolism 208
intrapulmonary pressure decrease and negative outcomes 208, 209
side effects 12, 13 preparations 207, 208
intrathoracic pressure effects on blood flow 10 timing 206
simulation-based mannequin teaching 57 ventilation patterns 207
Post-extubation upper airway obstruction synthesis 205
corticosteroids in prevention 36–38 types 205

Subject Index 255


TA mode overview 156, 157
algorithm description 6–8 guidance with Gram stain and bronchoalveolar
clinical implications in non-invasive ventilation 8, lavage culture 157–161
9 endotracheal tube role 151, 152
Teleassistance, see Home mechanical ventilation epidemiology 156
Thermal infrared imaging head position and gravity studies
polysomnography integration animal studies 152, 153
overview 48 clinical studies 154
prospects 47, 48 prospects for study 155
recommendations 49 mortality 156
principles 47, 48 recommendations for prevention 152
Thoracic epidural anesthesia resolution patterns 163–167
awake thoracic epidural anesthesia pulmonary Ventilator-related lung injury, neonates 213
resection Video-assisted thoracic surgery
lung cancer resection 112 advantages and limitations 91, 92, 94
metastasectomy 111, 112 applications 91
prospects 112 awake thoracic epidural anesthesia pulmonary
surgical pneumothorax physiological resection
effects 110 general anesthesia adverse effects 108, 109
undetermined pulmonary module infusion 109, 110
resection 110, 111 lung cancer resection 112
videothoracoscopy 107 metastasectomy 111, 112
general anesthesia adverse effects 108, 109 prospects 112
infusion 109, 110 surgical pneumothorax physiological
Time-adaptive mode, see TA mode effects 110
Timed-automated mode, see TA mode undetermined pulmonary module
Tracheostomy, ward mortality after intensive resection 110, 111
care 202, 203 videothoracoscopy 107
Training, see Simulation-based mannequin contraindications 90
teaching improvements 91–93
Transbronchial needle aspiration, endobronchial overview 89
ultrasound 85–87 recommendations 94
technical aspects 90, 91
Ultrasound, see Endobronchial ultrasound Viral infection
Upper airway obstruction, see Post-extubation upper community-acquired pneumonia 168–171
airway obstruction nosocomial viral infection 169–171
Vitamin E, antioxidant defense 234, 235
Ventilation-associated pneumonia Volume-assured pressure support 29, 30
antibiotic therapy

256 Subject Index

You might also like