12.07.2015 Views

Respiratory Management in Critical Care

Respiratory Management in Critical Care

Respiratory Management in Critical Care

SHOW MORE
SHOW LESS
  • No tags were found...

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Respiratory</strong> <strong>Management</strong><strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Edited by M J D Griffiths and T W Evans


___________________________________________________


<strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Edited byMJD GriffithsUnit of <strong>Critical</strong> <strong>Care</strong>, Imperial College of Science, Technology and Medic<strong>in</strong>e,Royal Brompton Hospital, London, UKTW EvansUnit of <strong>Critical</strong> <strong>Care</strong>, Imperial College of Science, Technology and Medic<strong>in</strong>e,Royal Brompton Hospital, London, UKiii


© BMJ Publish<strong>in</strong>g Group 2004BMJ Books is an impr<strong>in</strong>t of the BMJ Publish<strong>in</strong>g GroupAll rights reserved. No part of this publication may be reproduced, stored <strong>in</strong> a retrievalsystem, or transmitted, <strong>in</strong> any form or by any means, electronic, mechanical, photocopy<strong>in</strong>g,record<strong>in</strong>g and/or otherwise, without the prior written permission of the publishers.First published <strong>in</strong> 2004by BMJ Books, BMA House, Tavistock Square,London WC1H 9JRwww.bmjbooks.comBritish Library Catalogu<strong>in</strong>g <strong>in</strong> Publication DataA catalogue record for this book is available from the British LibraryISBN 0 7279 1729 3Typeset by BMJ Electronic ProductionPr<strong>in</strong>ted and bound <strong>in</strong> Spa<strong>in</strong> by GraphyCems, Navarraiv


ContributorsK AtabaiLung Biology Center, Department of Medic<strong>in</strong>e, University of California, San Francisco, USASV Baudo<strong>in</strong>Department of Anaesthesia, Royal Victoria Infirmary, Newcastle upon Tyne, UKGJ Bell<strong>in</strong>ganDepartment of Intensive <strong>Care</strong> Medic<strong>in</strong>e, University College London Hospitals, The Middlesex Hospital, London, UKRM du BoisInterstitial Lung Disease Unit, Royal Brompton Hospital, London, UKRJ BoytonHost Defence Unit, Royal Brompton Hospital, London, UKS BrettDepartment of Anaesthesia and Intensive <strong>Care</strong>, Hammersmith Hospital, London, UKJJ Cord<strong>in</strong>gleyDepartment of Anaesthesia and Intensive <strong>Care</strong>, Royal Brompton Hospital, London, UKPA CorrisDepartment of <strong>Respiratory</strong> Medic<strong>in</strong>e, Cardiothoracic Block, Freeman Hospital, Newcastle upon Tyne, UKJ CranshawUnit of <strong>Critical</strong> <strong>Care</strong>, NHLI Division, Imperial College of Science, Technology and Medic<strong>in</strong>e, Royal Brompton Hospital, London, UKJ Dak<strong>in</strong>Unit of <strong>Critical</strong> <strong>Care</strong>, NHLI Division, Imperial College of Science, Technology and Medic<strong>in</strong>eRoyal Brompton Hospital, London, UKAC DavidsonDepartments of <strong>Critical</strong> <strong>Care</strong> and <strong>Respiratory</strong> Support (Lane Fox Unit), Guys & St Thomas’ Hospital, London, UKSC DaviesDepartment of Haematology and Sickle Cell Unit, Central Middlesex Hospital, London, UKJ Dunn<strong>in</strong>gPulmonary Vascular Diseases Unit, Papworth Hospital, Cambridge and Department of Medic<strong>in</strong>e, University of Cambridge Schoolof Cl<strong>in</strong>ical Medic<strong>in</strong>e, Addenbrooke’s Hospital, Cambridge, UKTW EvansUnit of <strong>Critical</strong> <strong>Care</strong>, NHLI Division, Imperial College of Science, Technology and Medic<strong>in</strong>e, Royal Brompton Hospital, London, UKS EwigInstitut Cl<strong>in</strong>ic De Pneumologia i Cirurgia Toracica, Hospital Cl<strong>in</strong>ic, Servei de Pneumologia i Al.lergia Respiratoria, Barcelona,Spa<strong>in</strong>CS GarrardIntensive <strong>Care</strong> Unit, John Radcliffe Hospital, Oxford, UKA GascoigneDepartment of <strong>Respiratory</strong> Medic<strong>in</strong>e and Intensive <strong>Care</strong>, Royal Victoria Infirmary, Newcastle upon Tyne, UKJ GoldstoneDepartment of Intensive <strong>Care</strong> Medic<strong>in</strong>e, University College London Hospitals, The Middlesex Hospital, London, UKP GoldstrawDepartment of Thoracic Surgery, Royal Brompton Hospital, London, UK.JT GrantonUniversity Health Network, Mount S<strong>in</strong>ai Hospital and the Interdepartmental Division of <strong>Critical</strong> <strong>Care</strong>, University of Toronto,Toronto, Ontario, CanadaME GriffithDepartment of Renal Failure, St Mary’s Hospital NHS Trust, London, UKMJD GriffithsUnit of <strong>Critical</strong> <strong>Care</strong>, NHLI Division, Imperial College of Science, Technology and Medic<strong>in</strong>e, Royal Brompton Hospital, London, UKN HartSleep and Ventilation Unit, Royal Brompton and Harefield NHS Trust, London, UKAT JonesAdult Intensive <strong>Care</strong> Unit, Royal Brompton Hospital, London, UKBF KeoghDepartment of Anaesthesia and Intensive <strong>Care</strong>, Royal Brompton Hospital, London, UKOM KonChest and Allergy Department, St Mary’s Hospital NHS Trust, London, UKvii


SE Lap<strong>in</strong>skyMount S<strong>in</strong>ai Hospital and the Interdepartmental Division of <strong>Critical</strong> <strong>Care</strong>, University of Toronto, Toronto, Ontario, CanadaRM LeachDepartment of Intensive <strong>Care</strong>, Guy’s & St Thomas’ NHS Trust, London, UKJL LordanDepartment of <strong>Respiratory</strong> Medic<strong>in</strong>e, Cardiothoracic Block, Freeman Hospital, Newcastle upon Tyne, UKV MakDepartment of <strong>Respiratory</strong> and <strong>Critical</strong> <strong>Care</strong> Medic<strong>in</strong>e, Central Middlesex Hospital, London, UKMA MatthayCardiovascular Research Institute and Departments of Medic<strong>in</strong>e and Anesthesia, University of California, San Francisco, USAK McNeilPulmonary Vascular Diseases Unit, Papworth Hospital, Cambridge and Department of Medic<strong>in</strong>e, University of Cambridge Schoolof Cl<strong>in</strong>ical Medic<strong>in</strong>e, Addenbrooke’s Hospital, Cambridge, UKDM MitchellChest and Allergy Department, St Mary’s Hospital NHS Trust, London, UKED MoloneyImperial College School of Medic<strong>in</strong>e at the National Heart and Lung Institute,Royal Brompton Hospital, London, UKNW MorrellPulmonary Vascular Diseases Unit, Papworth Hospital, Cambridge and Department of Medic<strong>in</strong>e, University of Cambridge Schoolof Cl<strong>in</strong>ical Medic<strong>in</strong>e, Addenbrooke’s Hospital, Cambridge, UKP PhippsDepartment of Intensive <strong>Care</strong>, Royal Pr<strong>in</strong>ce Alfred Hospital, Sydney, AustraliaAK SimondsSleep and Ventilation Unit, Royal Brompton and Harefield NHS Trust, London, UKAS SlutskyDepartment of <strong>Critical</strong> <strong>Care</strong> and Department of Medic<strong>in</strong>e, St MichaelÆs Hospital, Interdepartmental Division of <strong>Critical</strong> <strong>Care</strong>,University of Toronto, Toronto, Ontario, CanadaSR ThomasDepartment of <strong>Respiratory</strong> Medic<strong>in</strong>e, St George’s Hospital, London, UKA TorresInstitut Cl<strong>in</strong>ic De Pneumologia i Cirurgia Toracica, Hospital Cl<strong>in</strong>ic, Servei de Pneumologia i Al.lergia Respiratoria, Barcelona,Spa<strong>in</strong>DF TreacherDepartment of Intensive <strong>Care</strong>, Guy’s & St Thomas’ NHS Trust, London, UKAU WellsInterstitial Lung Disease Unit, Royal Brompton Hospital, London, UKT WhiteheadDepartment of <strong>Respiratory</strong> Medic<strong>in</strong>e, Central Middlesex Hospital, London, UKviii


ContentsContributorsviiIntroductionMJD Griffiths, TW Evans 11. Pulmonary <strong>in</strong>vestigations for acute respiratory failureJ Dak<strong>in</strong>, MJD Griffiths 32. Oxygen delivery and consumption <strong>in</strong> the critically illRM Leach, DF Treacher 113. <strong>Critical</strong> care management of community acquired pneumoniaSV Baudou<strong>in</strong> 194. Nosocomial pneumoniaS Ewig, A Torres 245. Acute lung <strong>in</strong>jury and the acute respiratory distress syndrome: def<strong>in</strong>itions and epidemiologyK Atabai, MA Matthay 316. The pathogenesis of acute lung <strong>in</strong>jury/acute respiratory distress syndromeGJ Bell<strong>in</strong>gan 387. <strong>Critical</strong> care management of severe acute respiratory syndrome (SARS)JT Granton, SE Lap<strong>in</strong>sky 458. Ventilator <strong>in</strong>duced lung <strong>in</strong>juryT Whitehead, AS Slutsky 529. Ventilatory management of acute lung <strong>in</strong>jury/acute respiratory distress syndromeJJ Cord<strong>in</strong>gley, BF Keogh 6010. Non-ventilatory strategies <strong>in</strong> acute respiratory distress syndromeJ Cranshaw, MJD Griffiths, TW Evans 6611. Difficult wean<strong>in</strong>gJ Goldstone 7412. <strong>Critical</strong> care management of respiratory failure result<strong>in</strong>g from chronic obstructive pulmonary diseaseAC Davidson 8013. Acute severe asthmaP Phipps, CS Garrard 8614. The pulmonary circulation and right ventricular failureK McNeil, J Dunn<strong>in</strong>g, NW Morrell 9315. Thoracic trauma, <strong>in</strong>halation <strong>in</strong>jury and post-pulmonary resection lung <strong>in</strong>jury <strong>in</strong> <strong>in</strong>tensive careED Moloney, MJD Griffiths, P Goldstraw 9916. Illustrative case 1: cystic fibrosisSR Thomas 10617. Illustrative case 2: <strong>in</strong>terstitial lung diseaseAT Jones, RM du Bois, AU Wells 11018. Illustrative case 3: pulmonary vasculitisME Griffith, S Brett 11419. Illustrative case 4: neuromusculoskeletal disordersN Hart, AK Simonds 11720. Illustrative case 5: HIV associated pneumoniaRJ Boyton, DM Mitchell, OM Kon 12021. Illustrative case 6: acute chest syndrome of sickle cell anaemiaV Mak, SC Davies 12522. Illustrative case 7: the assessment and management of massive haemoptysisJL Lordan, A Gascoigne, PA Corris 128Index 135v


___________________________________________________


IntroductionM J D Griffiths, T W Evans.............................................................................................................................The care of the critically ill has changedradically dur<strong>in</strong>g the past 10 years. Technologicaladvances have improved monitor<strong>in</strong>g,organ support, and data collection, while smallsteps have been made <strong>in</strong> the development of drugtherapies. Conversely, new challenges (e.g. severeacute respiratory syndrome [SARS], multipleantimicrobial resistance, bioterrorism) cont<strong>in</strong>ueto arise and public expectations are elevated,sometimes to an unreasonable level. In this bookwe summarize some of the most important medicaladvances that have emerged, concentrat<strong>in</strong>gparticularly on those relevant to the grow<strong>in</strong>gnumbers of respiratory physicians who pursue asubspecialty <strong>in</strong>terest <strong>in</strong> this cl<strong>in</strong>ical arena.EVOLUTION OF INTENSIVE CAREMEDICINE AS A SPECIALTYIn Europe <strong>in</strong>tensive care medic<strong>in</strong>e (ICM) hasbeen one of the most recent cl<strong>in</strong>ical discipl<strong>in</strong>es toemerge. Dur<strong>in</strong>g a polio epidemic <strong>in</strong> Denmark <strong>in</strong>the early 1950s mortality was dramaticallyreduced by the application of positive pressureventilation to patients who had developed respiratoryfailure and by concentrat<strong>in</strong>g them <strong>in</strong> adesignated area with medical staff <strong>in</strong> constantattendance. This focus on airway care andventilatory management led to the gradual <strong>in</strong>troductionof <strong>in</strong>tensive care units (ICU), pr<strong>in</strong>cipallyby anaesthesiologists, throughout Western Europe.The development of sophisticated physiologicalmonitor<strong>in</strong>g equipment <strong>in</strong> the 1960s facilitatedthe diagnostic role of the <strong>in</strong>tensivist,extend<strong>in</strong>g their skill base beyond anaesthesiologyand attract<strong>in</strong>g cl<strong>in</strong>icians tra<strong>in</strong>ed <strong>in</strong> general <strong>in</strong>ternalmedic<strong>in</strong>e <strong>in</strong>to the ICU. Moreover, because respiratoryfailure was (and still is) the mostcommon cause of ICU admission, pulmonaryphysicians, particularly <strong>in</strong> the USA, were frequently<strong>in</strong>volved <strong>in</strong> patient care.ARE INTENSIVE CARE UNITS EFFECTIVE?Does <strong>in</strong>tensive care work and does the way <strong>in</strong>which it is provided affect patients’ outcomes? Ahigher rate of attributable mortality has beendocumented <strong>in</strong> patients who are refused <strong>in</strong>tensivecare, particularly on an emergency basis. 1 Cl<strong>in</strong>icaloutcome is improved by the conversion ofso-called “open” ICU to closed facilities <strong>in</strong> whichpatient management is directed primarily by<strong>in</strong>tensive care specialists. 23Superior organisationalpractices emphasis<strong>in</strong>g strong medical andnurs<strong>in</strong>g leadership can also improve outcome. 4The emergence of <strong>in</strong>termediate care, high dependency,or step down facilities has attempted tofill the grow<strong>in</strong>g gap between the level of care thatmay be provided <strong>in</strong> the ICU and that <strong>in</strong> thegeneral wards. Worry<strong>in</strong>gly, the time at whichpatients are discharged from ICU <strong>in</strong> the UK has ademonstrable effect on their outcome. 5Earlyidentification of patients at risk of death—bothbefore admission and after discharge from theICU—may decrease mortality. 6Patients can beidentified who have a low risk of mortality andwho are likely to benefit from a brief period ofmore <strong>in</strong>tensive supervision and care. 7 Designatedteams that are equipped to transfer critically illpatients between specialist units have a crucialrole to play <strong>in</strong> ensur<strong>in</strong>g that patient care and theuse of resources are optimized. 8 F<strong>in</strong>ally, long termfollow up of the critically ill as outpatientsfollow<strong>in</strong>g discharge from hospital may identifyproblems of chronic ill health that require activemanagement and rehabilitation. 9TRAINING IN INTENSIVE CARE MEDICINEImproved tra<strong>in</strong><strong>in</strong>g of medical and nurs<strong>in</strong>g staffand organisational changes have undoubtedlyplayed their part <strong>in</strong> improv<strong>in</strong>g the outcome ofcritical illness. ICM is now a recognised specialty<strong>in</strong> two European Union member states, namelySpa<strong>in</strong> and the UK. Where available, tra<strong>in</strong><strong>in</strong>g <strong>in</strong>ICM is of variable duration and is accessible variablyto cl<strong>in</strong>icians of differ<strong>in</strong>g base specialties. InSpa<strong>in</strong> 5 years of tra<strong>in</strong><strong>in</strong>g are required to achievespecialist status, 3 years of which are <strong>in</strong> ICM. InFrance, Germany, Greece, and the UK, 2 years oftra<strong>in</strong><strong>in</strong>g <strong>in</strong> ICM are required, <strong>in</strong> addition to thosneeded for the base specialty (usually anaesthesiology,respiratory or general <strong>in</strong>ternal medic<strong>in</strong>e).In Italy, only anaesthesiologists may practiceICM. There is considerable variation betweenmembers states of the European Union regard<strong>in</strong>gthe amount of exposure to ICM <strong>in</strong> the tra<strong>in</strong><strong>in</strong>g ofpulmonary physicians as a mandatory (M) oroptional (O) requirement: France and Greece 6months (O), Germany 6 months (M, as part ofgeneral <strong>in</strong>ternal medic<strong>in</strong>e), UK 3 months (O),and Italy and Spa<strong>in</strong> none.TRAINING IN INTENSIVE CARE MEDICINEIN THE UKAn <strong>in</strong>creas<strong>in</strong>g number of appo<strong>in</strong>tments <strong>in</strong> ICMare now available to tra<strong>in</strong>ees <strong>in</strong> general <strong>in</strong>ternalmedic<strong>in</strong>e at senior house officer level, usually fora period of 3 months. For specialist registrars, anumber of options have emerged. First, <strong>in</strong> somespecialties (e.g. respiratory medic<strong>in</strong>e, <strong>in</strong>fectiousdiseases) specialist registrars are already encouragedto undertake a period of tra<strong>in</strong><strong>in</strong>g <strong>in</strong> ICM.Second, 6 months of tra<strong>in</strong><strong>in</strong>g <strong>in</strong> anaesthesia plus6 months of ICM (<strong>in</strong> addition to 3 months ofexperience as a senior house officer) <strong>in</strong> approvedprogrammes confers <strong>in</strong>termediate accreditationby the Inter-Collegiate Board for Tra<strong>in</strong><strong>in</strong>g <strong>in</strong> ICM(http://www.ics.ac.uk/ibticm_board.html). F<strong>in</strong>allya further 12 months of experience <strong>in</strong> recognisedunits can lead to the award of a Certificate ofCompletion of Specialist Tra<strong>in</strong><strong>in</strong>g (CCST) comb<strong>in</strong>edwith base specialty. Importantly, up to 12months of such experience can be substituted for6 months <strong>in</strong> general <strong>in</strong>ternal medic<strong>in</strong>e (foranaesthesia) and respiratory medic<strong>in</strong>e (for ICM).


2 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Table 1 Proposed classification of critical illness 10Level 0Level 1Level 2Level 3Patients whose needs can be met through normal ward care <strong>in</strong> an acute hospitalPatients at risk of their condition deteriorat<strong>in</strong>g, or those recently relocated from higher levels of care, whose needs can be met onan acute ward with additional advice and support from the critical care teamPatients requir<strong>in</strong>g more detailed observations or <strong>in</strong>tervention <strong>in</strong>clud<strong>in</strong>g support for a s<strong>in</strong>gle fail<strong>in</strong>g organ system or postoperativecare and those “stepp<strong>in</strong>g down” from higher levels of carePatients requir<strong>in</strong>g advanced respiratory support alone or basic respiratory support together with support of at least two organsystems. This level <strong>in</strong>cludes all complex patients requir<strong>in</strong>g support for multiorgan failureThus, a period of 5 years is needed for <strong>in</strong>termediate accreditation<strong>in</strong> ICM plus a CCST <strong>in</strong> general <strong>in</strong>ternal and respiratorymedic<strong>in</strong>e, and 6 for the award of a treble CCST. Programmesare now becom<strong>in</strong>g available <strong>in</strong> all regions to enable tra<strong>in</strong>eeswith National Tra<strong>in</strong><strong>in</strong>g Numbers from all base specialties toachieve these tra<strong>in</strong><strong>in</strong>g requirements and the proscribed competencies<strong>in</strong> ICM.THE FUTURE FOR INTENSIVE CARE MEDICINE: A UKPERSPECTIVEThe chang<strong>in</strong>g requirements and <strong>in</strong>creased need for provisionof <strong>in</strong>tensive care were recognised <strong>in</strong> the UK <strong>in</strong> the late 1990sby the Department of Health which commissioned the reportentitled “Comprehensive <strong>Critical</strong> <strong>Care</strong>” produced by an expertgroup to provide a blue pr<strong>in</strong>t for the future development ofICM with<strong>in</strong> the NHS. 10 A central tenet of the report is the ideathat the service should extend to the provision of critical carethroughout the hospital, and not merely to patients locatedwith<strong>in</strong> the traditional conf<strong>in</strong>es of the ICU. To this end, theadoption of a new classification of illness severity based ondependency rather than location was recommended. Traditionally,the critically ill were def<strong>in</strong>ed accord<strong>in</strong>g to their needfor <strong>in</strong>tensive care (delivered at a ratio of one nurse to onepatient) and those requir<strong>in</strong>g high dependency care (deliveredat a ratio of one nurse to two or more patients). The newclassification is based on the severity of the patient’s illnessand on the level of care needed (table 1). The report thereforerepresents a “whole systems” approach encompass<strong>in</strong>g theprovision of care, both before and after the acute episodewith<strong>in</strong> an <strong>in</strong>tegrated system.To <strong>in</strong>itiate and oversee the implementation of this policy, 29local “networks” have been established, with an adm<strong>in</strong>istrativeand cl<strong>in</strong>ical <strong>in</strong>frastructure. Networks will be used to pilotnational <strong>in</strong>itiatives and enable groups of hospitals to establishlocally agreed practices and protocols. <strong>Critical</strong>ly ill patientswill be transferred between network hospitals if facilities orexpertise with<strong>in</strong> a s<strong>in</strong>gle <strong>in</strong>stitution are <strong>in</strong>adequate to providethe necessary care, thereby obviat<strong>in</strong>g the problems associatedwith mov<strong>in</strong>g such patients over long distances to access asuitable bed.CONCLUSIONHow should the respiratory physician react to these developments?We suggest that an attachment <strong>in</strong> ICM for all respiratorytra<strong>in</strong>ees is necessary. Indeed, specialty recognition andthe <strong>in</strong>creased availability of tra<strong>in</strong><strong>in</strong>g opportunities shouldencourage some tra<strong>in</strong>ees from respiratory medic<strong>in</strong>e to seek aCCST comb<strong>in</strong>ed with ICM. Second, we suggest that changes <strong>in</strong>the organisational and adm<strong>in</strong>istrative structure of <strong>in</strong>tensivecare services heralded by the publication of “Comprehensive<strong>Critical</strong> <strong>Care</strong>” are likely to impact most heavily on respiratoryphysicians. For example, respiratory support services us<strong>in</strong>gnon-<strong>in</strong>vasive ventilation are particularly attractive <strong>in</strong> provid<strong>in</strong>gboth “step up” (from the general wards) and “step down”(from the ICU) facilities. In the USA, respiratory physicianshave for a long time been the major providers of critical care.In the UK and the rest of Europe, given appropriate resourcesand tra<strong>in</strong><strong>in</strong>g, the pulmonary physician is ideally suited tobecome an <strong>in</strong>tegral component of the critical care servicewith<strong>in</strong> all hospitals.REFERENCES1 Metcalfe MA, Sloggett A, McPherson K. Mortality among appropriatelyreferred patients refused admission to <strong>in</strong>tensive-care units. Lancet1997;350:7–11.2 Carson SS, Stock<strong>in</strong>g C, Podsadecki T, et al. Effects of organizationalchange <strong>in</strong> the medical <strong>in</strong>tensive care unit of a teach<strong>in</strong>g hospital: acomparison of ‘open’ and ‘closed’ formats. JAMA 1996;276:322–8.3 Ghorra S, Re<strong>in</strong>ert SE, Cioffi W, et al. Analysis of the effect of conversionfrom open to closed surgical <strong>in</strong>tensive care unit. Ann Surg1999;229:163–71.4 Zimmerman JE, Shortell SM, Rousseau DM, et al. Improv<strong>in</strong>g <strong>in</strong>tensivecare: observations based on organizational case studies <strong>in</strong> n<strong>in</strong>e <strong>in</strong>tensivecare units: a prospective, multicenter study. Crit <strong>Care</strong> Med1993;21:1443–51.5 Goldfrad C, Rowan K. Consequences of discharges from <strong>in</strong>tensive careat night. Lancet 2000;355:1138–42.6 Jakob SM, Rothen HU. Intensive care 1980–1995: change <strong>in</strong> patientcharacteristics, nurs<strong>in</strong>g workload and outcome. Intensive <strong>Care</strong> Med1997;23:1165–70.7 Kilpatrick A, Ridley S, Plenderleith L. A chang<strong>in</strong>g role for <strong>in</strong>tensivetherapy: is there a case for high dependency care? Anaesthesia1994;49:666–70.8 Bell<strong>in</strong>gan G, Olivier T, Batson S, Webb A. Comparison of a specialistretrieval team with current United K<strong>in</strong>gdom practice for the transport ofcritically ill patients. Intensive <strong>Care</strong> Med 2000;26:740–4.9 Angus DC, Musthafa AA, Clermont G, et al. Quality-adjusted survival <strong>in</strong>the first year after the acute respiratory distress syndrome. Am J RespirCrit <strong>Care</strong> Med 2001;163:1389–94.10 Department of Health. Comprehensive critical care: review of adultcritical care services. London: Department of Health, 2000.


Pulmonary <strong>in</strong>vestigations for acute respiratory failure 5Table 1.2 Typical bronchoalveolar lavage differential cell counts <strong>in</strong> conditions associated with acute respiratory failureand diffuse pulmonary <strong>in</strong>filtratesCondition Cell differential counts CommentsMacrophage Lymphocyte Neutrophil Eos<strong>in</strong>ophilNormal 90% 10%


6 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>fluid neutrophilia had adverse prognostic significance while ahigher macrophage count was associated with a betteroutcome. 54 The fibroproliferative phase of ARDS may be amenableto treatment with steroids 55 and it is recommended thateither BAL or PSB is performed before start<strong>in</strong>g treatment toexclude <strong>in</strong>fection.For patients with suspected or confirmed ARDS a sensitiveand specific marker of disease would have several benefits.Firstly, it might improve the ability to predict which patientswith risk factors develop ARDS 56 so that potentially protectivemeasures could be assessed and developed. Secondly, it mayhelp to quantify the severity of disease and to predict complicationssuch as fibrosis and superadded <strong>in</strong>fection. Most studieshave <strong>in</strong>volved assays on plasma samples or BAL fluid. 56Analysis may provide <strong>in</strong>formation about soluble <strong>in</strong>flammatorymediators and by-products of <strong>in</strong>flammation (such as shedadhesion molecules, elastase, peroxynitrite) <strong>in</strong> the distalairways and air spaces. Analysis of samples from patients atrisk has revealed <strong>in</strong>creased alveolar levels of the potentneutrophil chemok<strong>in</strong>e <strong>in</strong>terleuk<strong>in</strong> 8 (IL-8) <strong>in</strong> those patientswho progress to ARDS. 57The development of establishedfibrosis conveys a poor prognosis <strong>in</strong> ARDS. 58 Type III procollagenpeptide is present from the day of tracheal <strong>in</strong>tubation <strong>in</strong>the pulmonary oedema fluid of patients with <strong>in</strong>cipient lung<strong>in</strong>jury, and the concentration correlates with mortality. 59 Less<strong>in</strong>vasive methods of sampl<strong>in</strong>g distal lung l<strong>in</strong><strong>in</strong>g fluid us<strong>in</strong>gexhaled breath 60 61 or exhaled breath condensates 62 63 are be<strong>in</strong>gexam<strong>in</strong>ed <strong>in</strong> critically ill patients. The assay of potentialbiomarkers is currently used exclusively as a research tool.RADIOLOGY64 65Chest radiographyThe cost effectiveness of a daily chest radiograph <strong>in</strong> the66 67mechanically ventilated patient has been debated but isrecommended by the American College of Radiology 68 basedon series highlight<strong>in</strong>g the <strong>in</strong>cidence (15–18%) of unsuspectedf<strong>in</strong>d<strong>in</strong>gs lead<strong>in</strong>g directly to changes <strong>in</strong> management. 69–71 Filmacquisition <strong>in</strong> the ICU is technically demand<strong>in</strong>g but guidel<strong>in</strong>eshave been published. 72 Digital imag<strong>in</strong>g techniques permit theuse of lower radiation doses and manipulate images toproduce, <strong>in</strong> effect, a standard exposure as well as an edgeenhanced image to facilitate visualisation of, for example,<strong>in</strong>travenous l<strong>in</strong>es and pneumothoraces.Endotracheal tubes and central venous catheters 73A radiograph is recommended after placement or reposition<strong>in</strong>gof all central venous catheters, pleural dra<strong>in</strong>s, nasogastric,and endotracheal tubes. 68 The tip of the endotracheal tube maymove up to 4 cm with neck flexion and extension, 74 and theend should be 5–7 cm from the car<strong>in</strong>a or project on a pla<strong>in</strong>chest radiograph to the level of T3–T4. 75 Tracheal rupture maybe reflected <strong>in</strong> radiological evidence of overdistension of theendotracheal tube or tracheostomy balloon to a greater diameterthan that of the trachea. Surpris<strong>in</strong>gly, the presentation ofthis potentially catastrophic complication is often gradual,with surgical emphysema and pneumomediast<strong>in</strong>um develop<strong>in</strong>gover 24 hours. 76Central venous catheters should be positioned <strong>in</strong> the superiorvena cava (SVC) at the level of or slightly above the azygosve<strong>in</strong>. Caudal to this, the SVC lies with<strong>in</strong> the pericardium mak<strong>in</strong>gtamponade likely if the atrial wall is perforated. Position<strong>in</strong>gof left sided l<strong>in</strong>es with their ends abutt<strong>in</strong>g the wall of theSVC is a risk factor for perforation. Encroachment of l<strong>in</strong>es <strong>in</strong>tothe atrium may cause arrhythmia and be associated with ahigher <strong>in</strong>cidence of endocarditis. 77The ideal radiologicalplacement of pulmonary artery catheters has not beenstudied. To m<strong>in</strong>imize the risk of <strong>in</strong>farction or perforation, theballoon should be sited rout<strong>in</strong>ely <strong>in</strong> the largest diameter pulmonaryartery that will provide a wedge trace on <strong>in</strong>flation, andplacement should be reviewed frequently to prevent migrationof the catheter tip more away from the hilum. 78Radiographic appearances <strong>in</strong> ARFThe radiographic appearance of ARDS is a cornerstone of itsdiagnosis (see chapter 5). However, dist<strong>in</strong>guish<strong>in</strong>g betweencardiogenic and high permeability pulmonary oedema onradiographic signs alone is unreliable. 79 The cardiac size andvascular pedicle width reflect the haemodynamic state of thepatient, 80 but this sign relies on exact and often unachievablepatient position<strong>in</strong>g. Pleural effusions and Kerley’s l<strong>in</strong>esreflect<strong>in</strong>g lymphatic engorgement are not characteristic ofARDS because the high prote<strong>in</strong> content and viscosity of theoedema fluid prevents it from spread<strong>in</strong>g <strong>in</strong>to the peripheral<strong>in</strong>terstitial and pleural spaces. Air bronchograms are seen <strong>in</strong>up to one third of cases as the airways rema<strong>in</strong> dry <strong>in</strong> ARDS,thereby contrast<strong>in</strong>g with the surround<strong>in</strong>g parenchyma.In contrast to hydrostatic pulmonary oedema, the radiographicsigns of ARDS are frequently not visible on the pla<strong>in</strong>chest radiograph for 24 hours after the onset of symptoms.Early changes comprise patchy ill def<strong>in</strong>ed densities thatbecome confluent to form ground glass shadow<strong>in</strong>g. Inventilated patients air space shadow<strong>in</strong>g commonly resultsfrom pneumonia or atelectasis; other causes are ARDS, haemorrhage,and lung contusion. The detection and quantificationof pleural fluid by the sup<strong>in</strong>e chest radiograph is81 82<strong>in</strong>accurate.Thoracic ultrasoundThe presence of fluid with<strong>in</strong> the pleural space has an adverseeffect on ventilation-perfusion match<strong>in</strong>g 83 ; removal improvesoxygenation and pulmonary compliance. 83 84 Dra<strong>in</strong>age may beperformed safely by ultrasound guided thoracocentesis <strong>in</strong> the85 86ventilated patient.Thoracic computed tomography (CT)Transportation to and monitor<strong>in</strong>g of a critically ill patient forCT scann<strong>in</strong>g <strong>in</strong>volves a team effort from medical, nurs<strong>in</strong>g, andtechnical support staff. There are no published data describ<strong>in</strong>gthe risks and benefits of this <strong>in</strong>vestigation <strong>in</strong> a well def<strong>in</strong>edgroup of critically ill patients. However, <strong>in</strong> a retrospectivereview of 108 thoracic CT scans performed on patients <strong>in</strong> ageneral ICU, at least one new cl<strong>in</strong>ically significant f<strong>in</strong>d<strong>in</strong>g(most commonly abscess, malignancy, unsuspected pneumonia,or pleural effusion) was identified <strong>in</strong> 30% of cases and <strong>in</strong>22% led to a change <strong>in</strong> management. 87 The normal standardsand precautions for transport<strong>in</strong>g critically ill patients apply, 88<strong>in</strong>clud<strong>in</strong>g a period of stabilisation on the transport ventilatorprior to movement. Despite the added risk of complicationssuch as pneumothorax, haemodynamic <strong>in</strong>stability and lungderecruitment associated with transportation, we rout<strong>in</strong>elyscan patients with ARDS if their gas exchange on thetransport ventilator is acceptable. Portable CT scannersprovide mediast<strong>in</strong>al images of comparable quality to thoseobta<strong>in</strong>ed <strong>in</strong> the radiology department, but the images of thelung parenchyma are <strong>in</strong>ferior. 89Thoracic CT <strong>in</strong> specific conditionsARDSInsight <strong>in</strong>to the nature of ARDS has been obta<strong>in</strong>ed from CTscann<strong>in</strong>g, for example, by def<strong>in</strong><strong>in</strong>g the disease distribution anddemonstrat<strong>in</strong>g ventilator <strong>in</strong>duced lung <strong>in</strong>jury (see chapter 8). 90CT scans of the lung parenchyma show that the diffuse opacificationon the pla<strong>in</strong> radiograph is not homogenous; classically,there is a gradient of decreas<strong>in</strong>g aeration pass<strong>in</strong>g from ventral todorsal dependent regions. 91 Tidal volume is therefore directedexclusively to the overly<strong>in</strong>g anterior regions which areconsequently overdistended. This may account for the anteriordistribution of reticular damage seen on CT scans <strong>in</strong>survivors. 92 The improvement <strong>in</strong> oxygenation of patients with


Pulmonary <strong>in</strong>vestigations for acute respiratory failure 7Figure 1.3. Radiology of a case of left lower lobe pneumonia complicated by ARDS. (A) Chest radiograph and CT scan taken on the sameday 3 weeks after the onset of respiratory failure. An abscess is obvious <strong>in</strong> the apical segment of the left lower lobe on the CT scan. There isdense dependent consolidation bilaterally but elsewhere the lungs are affected <strong>in</strong> a patchy distribution. (B) Chest radiograph and CT scantaken on the same day 5 months after the onset of respiratory failure. Bilateral loculated pneumothoraces are evident despite the placement ofseveral <strong>in</strong>tercostal chest dra<strong>in</strong>s on both sides. (C) Chest CT scan taken 6 months after discharge from hospital show<strong>in</strong>g diffuse emphysema andpatchy areas of fibrosis.ARDS follow<strong>in</strong>g prone position<strong>in</strong>g suggests improvedventilation-perfusion match<strong>in</strong>g. However, microsphere CT studies<strong>in</strong> animal models of ARDS have failed to demonstrate redirectionof perfusion with prone position<strong>in</strong>g 93 ; redirection of ventilationto the consolidated dorsal regions may therefore be themechanism responsible.Recovery from ARDS is commonly complicated by pneumothoraceswhich are often loculated. If a pneumothorax does notextend to the lateral thoracic wall, it will not be readily apparenton a chest radiograph. Its presence may be <strong>in</strong>ferred from a rangeof <strong>in</strong>direct signs such as a vague radiolucency or undue clarity ofthe diaphragm, but this gives no <strong>in</strong>formation as to whether thecollection of air is located anteriorly or posteriorly. Similarly,empyema and abscess formation may cause treatment failure <strong>in</strong>patients with pneumonia and ARDS and are not <strong>in</strong>frequentlymissed on the pla<strong>in</strong> film (fig 1.3). 94CT guided percutaneousdra<strong>in</strong>age may be required for loculated pneumothoraces andmay be an alternative to surgery for lung abscesses.Pulmonary embolusMassive pulmonary embolus is a treatable cause of rapid cardiorespiratorydeterioration which is frequently not diagnosedbefore death (see chapter 14). Radionuclide scann<strong>in</strong>g has along image acquisition time and assays for detect<strong>in</strong>g D-dimersare unduly sensitive <strong>in</strong> this sett<strong>in</strong>g, mak<strong>in</strong>g both unsuitablefor the critically ill patient. CT pulmonary angiography is the


8 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong><strong>in</strong>vestigation of choice and may provide an alternativediagnosis to account for the presentation.TraumaRout<strong>in</strong>e CT scann<strong>in</strong>g of all victims of serious trauma uncoverslesions (pneumothorax, haemothorax, pulmonary contusion)not detected on cl<strong>in</strong>ical exam<strong>in</strong>ation and pla<strong>in</strong> radiography. 95However, there is no evidence to suggest that a better patientoutcome follows rout<strong>in</strong>e scann<strong>in</strong>g. Different trauma centresfavour aggressive 96 and conservative 97 98 management of smallpneumothoraces <strong>in</strong> the ventilated patient.LUNG FUNCTIONFormal assessment of lung function is most commonlyrequired for patients who experience difficulty <strong>in</strong> wean<strong>in</strong>gwhere measurements of peak flow, vital capacity, and respiratorymuscle strength may be useful (see chapters 11 and 19).An airtight connection between the endotracheal tube and ahand held spirometer can give accurate and reproducibleresults. A vital capacity of 10 ml/kg is usually required to susta<strong>in</strong>spontaneous ventilation. If respiratory muscle weaknessis suspected, measurements should be performed sitt<strong>in</strong>g andsup<strong>in</strong>e. A sup<strong>in</strong>e reduction of 25% or more <strong>in</strong>dicatesdiaphragm weakness. Direct measurement of diaphragmstrength is useful where borderl<strong>in</strong>e results are obta<strong>in</strong>ed fromspirometric test<strong>in</strong>g, <strong>in</strong> uncooperative patients, or <strong>in</strong> those withlung disease that impairs spirometric measurements.Transdiaphragmatic pressure, an <strong>in</strong>dex of the strength of diaphragmaticcontractility, is measured by peroral passage ofballoon manometers <strong>in</strong>to the oesophagus and stomach. Avolitional measurement is made by ask<strong>in</strong>g the patient to sniffforcefully from functional residual capacity. A non-volitionalmeasurement can be made reproducibly by magnetic stimulationof the phrenic nerves us<strong>in</strong>g a coil directly applied to thesk<strong>in</strong> of the neck. 99 A low maximal <strong>in</strong>spiratory pressure (PI max)predicts failure to wean, although it is <strong>in</strong>sensitive <strong>in</strong> predict<strong>in</strong>gsuccess. 100In the mechanically ventilated patient gas exchange andventilation are assessed rout<strong>in</strong>ely by arterial blood gas analysisand cont<strong>in</strong>uous oxygen saturation monitor<strong>in</strong>g. Refractoryhypoxia that is characteristic of ARDS is almost entirelycaused by <strong>in</strong>trapulmonary shunt<strong>in</strong>g. 101 Oxygenation is quantified<strong>in</strong> the American-European Consensus Conference(AECC) def<strong>in</strong>ition of ARDS and ALI by the ratio of the arterialpartial pressure and the <strong>in</strong>spired oxygen concentration (PaO 2/FiO 2). 1 This <strong>in</strong>itial value does not predict survival 102 but is a reasonablepredictor of shunt fraction 103 and has epidemiologicalimportance as it is used to dist<strong>in</strong>guish patients with severe(ARDS) and less severe (ALI) lung <strong>in</strong>jury. The PaO 2/FiO 2ratio issimple to calculate but does not take <strong>in</strong>to account other factorsthat affect oxygenation such as the mean airway pressure(mPaw). 104 The oxygenation <strong>in</strong>dex (OI = mPaw × FiO 2× 100/PaO 2) benefits from <strong>in</strong>clud<strong>in</strong>g this variable; similarly, therespiratory severity <strong>in</strong>dex (PO 2alveolar − PO 2arterial/PO 2alveolar + 0.014PEEP) is more cumbersome but the value<strong>in</strong> the first 24 hours did dist<strong>in</strong>guish survivors and nonsurvivors<strong>in</strong> a study of 56 consecutive patients with ARDSdef<strong>in</strong>ed us<strong>in</strong>g the AECC criteria. 105 As a compromise the PaO 2/FiO 2ratio may be calculated at a standardised level of PEEP.Assessment of respiratory physiology has undergone arecent resurgence as novel adjuncts to ventilator therapy (e.g.prone position<strong>in</strong>g and <strong>in</strong>haled vasodilators) have been <strong>in</strong>vestigatedand the importance of mitigat<strong>in</strong>g ventilator <strong>in</strong>ducedlung <strong>in</strong>jury has been recognised. 106 Most ventilators cont<strong>in</strong>uouslydisplay airway pressures, delivered and exhaledvolumes, and compliance. The compliance of the respiratorysystem is def<strong>in</strong>ed by the relationship:change <strong>in</strong> volume/change <strong>in</strong> elastic recoil pressure =tidal volume/plateau pressure – PEEP (ml/cm H 2O)This gives the total compliance of the lung and chest wallassum<strong>in</strong>g that the patient is mak<strong>in</strong>g no spontaneous respiratoryeffort. Values are commonly halved or lower <strong>in</strong> ARDS(normal range 50–80 ml/cm H 2O), although measurement ofthis variable is not required by the standard def<strong>in</strong>ition. 1Study<strong>in</strong>g pressure-volume curves of patients with ARDShighlighted the risk of overdistension at what would beconsidered a “normal” tidal volume, 107 and the results of therecent ARDS network study confirmed the benefit of ventilationat a restricted volume. 106While the optimum balancebetween PEEP and FiO 2and the role of the pressure-volumecurve <strong>in</strong> sett<strong>in</strong>g the optimum level of PEEP rema<strong>in</strong> to bedeterm<strong>in</strong>ed, we cannot recommend that generat<strong>in</strong>g pressurevolumecurves <strong>in</strong> patients with lung <strong>in</strong>jury is required otherthan for research. 108SUMMARYWhen <strong>in</strong>vestigat<strong>in</strong>g patients with ARF and pulmonary<strong>in</strong>filtrates, one must achieve a balance between the necessityof rapid diagnosis and the early <strong>in</strong>stigation of effectivetherapy, aga<strong>in</strong>st the potential harm caused by <strong>in</strong>vasivetechniques <strong>in</strong> patients with very limited reserves. Because ofthe pressure on <strong>in</strong>tensive care beds <strong>in</strong> the UK, our facilitiesand expertise <strong>in</strong> provid<strong>in</strong>g temporary support are probablyunder-used <strong>in</strong> the <strong>in</strong>vestigation of such cases before mechanicalventilation is mandatory. We suggest a scheme for the<strong>in</strong>vestigation of patients present<strong>in</strong>g with ARF and discuss theeffects of mechanical ventilation and critical illness oncommonly used <strong>in</strong>vestigations of the respiratory system.REFERENCES1 Bernard GR, Artigas A, Brigham KL, et al. The American-EuropeanConsensus Conference on ARDS. Def<strong>in</strong>itions, mechanisms, relevantoutcomes, and cl<strong>in</strong>ical trial coord<strong>in</strong>ation. Am J Respir Crit <strong>Care</strong> Med1994;149(3 Pt 1):818–24.2 Macnaughton P, Evans T. Pulmonary function <strong>in</strong> the <strong>in</strong>tensive care unit.In: Hughes J, Pride N, eds. Lung function tests. Physiological pr<strong>in</strong>ciplesand cl<strong>in</strong>ical applications, 1st ed. London: Saunders, 1999:185–201.3 British Thoracic Society. British Thoracic Society guidel<strong>in</strong>es ondiagnostic flexible bronchoscopy. Thorax 2001;56(Supplement 1):i1–22.4 L<strong>in</strong>dholm CE, Ollman B, Snyder JV, et al. Cardiorespiratory effects offlexible fiberoptic bronchoscopy <strong>in</strong> critically ill patients. Chest1978;74:362–8.5 Jolliet P, Chevrolet JC. Bronchoscopy <strong>in</strong> the <strong>in</strong>tensive care unit. Intensive<strong>Care</strong> Med 1992;18:160–9.6 Marx WH, Ciaglia P, Graniero KD. Some important details <strong>in</strong> thetechnique of percutaneous dilatational tracheostomy via the modifiedSeld<strong>in</strong>ger technique. Chest 1996;110:762–6.7 Barrett CR Jr. Flexible fiberoptic bronchoscopy <strong>in</strong> the critically ill patient.Methodology and <strong>in</strong>dications. Chest 1978;73(5 Suppl):746–9.8 Olopade CO, Prakash UB. Bronchoscopy <strong>in</strong> the critical-care unit. MayoCl<strong>in</strong> Proc 1989;64:1255–63.9 Niederman M, Torres A. Bronchospopy for pneumonia: <strong>in</strong>dications,methodology and applications. In: Fe<strong>in</strong>silver SFA, ed. Textbook ofbronchoscopy. Baltimore: Williams and Wilk<strong>in</strong>s, 1995:221–41.10 Jimenez P, Saldias F, Meneses M, et al. Diagnostic fiberopticbronchoscopy <strong>in</strong> patients with community-acquired pneumonia.Comparison between bronchoalveolar lavage and telescop<strong>in</strong>g pluggedcatheter cultures. Chest 1993;103:1023–7.11 Ruiz M, Torres A, Ewig S, et al. Non<strong>in</strong>vasive versus <strong>in</strong>vasive microbial<strong>in</strong>vestigation <strong>in</strong> ventilator-associated pneumonia: evaluation of outcome.Am J Respir Crit <strong>Care</strong> Med 2000;162:119–25.12 Papazian L, Thomas P, Garbe L, et al. Bronchoscopic or bl<strong>in</strong>d sampl<strong>in</strong>gtechniques for the diagnosis of ventilator-associated pneumonia. Am JRespir Crit <strong>Care</strong> Med 1995;152(6 Pt 1):1982–91.13 Marik PE, Brown WJ. A comparison of bronchoscopic vs bl<strong>in</strong>d protectedspecimen brush sampl<strong>in</strong>g <strong>in</strong> patients with suspected ventilator-associatedpneumonia. Chest 1995;108:203–7.14 Pap<strong>in</strong> TA, Grum CM, Weg JG. Transbronchial biopsy dur<strong>in</strong>gmechanical ventilation. Chest 1986;89:168–70.15 P<strong>in</strong>cus PS, Kallenbach JM, Hurwitz MD, et al. Transbronchial biopsydur<strong>in</strong>g mechanical ventilation. Crit <strong>Care</strong> Med 1987;15:1136–9.16 Trulock EP, Ett<strong>in</strong>ger NA, Brunt EM, et al. The role of transbronchial lungbiopsy <strong>in</strong> the treatment of lung transplant recipients. An analysis of 200consecutive procedures. Chest 1992;102:1049–54.17 Scott JP, Fradet G, Smyth RL, et al. Prospective study of transbronchialbiopsies <strong>in</strong> the management of heart-lung and s<strong>in</strong>gle lung transplantpatients. J Heart Lung Transplant 1991;10(5 Pt 1):626–36; discussion636–7.18 Higenbottam T, Stewart S, Penketh A, et al. Transbronchial lung biopsyfor the diagnosis of rejection <strong>in</strong> heart-lung transplant patients.Transplantation 1988;46:532–9.


Pulmonary <strong>in</strong>vestigations for acute respiratory failure 919 Yousem SA, Berry GJ, Cagle PT, et al. Revision of the 1990 work<strong>in</strong>gformulation for the classification of pulmonary allograft rejection: LungRejection Study Group. J Heart Lung Transplant 1996;15(1 Pt 1):1–15.20 McGregor CG, Daly RC, Peters SG, et al. Evolv<strong>in</strong>g strategies <strong>in</strong> lungtransplantation for emphysema. Ann Thorac Surg 1994;57:1513–20;discussion 1520–1.21 Trulock EP. <strong>Management</strong> of lung transplant rejection. Chest1993;103:1566–76.22 Bulpa PA, Dive AM, Mertens L, et al. Comb<strong>in</strong>ed bronchoalveolar lavageand transbronchial lung biopsy: safety and yield <strong>in</strong> ventilated patients.Eur Respir J 2003;21:489–94.23 Flabouris A, Myburgh J. The utility of open lung biopsy <strong>in</strong> patientsrequir<strong>in</strong>g mechanical ventilation. Chest 1999;115:811–7.24 Papazian L, Thomas P, Bregeon F, et al. Open-lung biopsy <strong>in</strong> patientswith acute respiratory distress syndrome. Anesthesiology1998;88:935–44.25 Nelems JM, Cooper JD, Henderson RD, et al. Emergency open lungbiopsy. Ann Thorac Surg 1976;22:260–4.26 Warner DO, Warner MA, Divertie MB. Open lung biopsy <strong>in</strong> patientswith diffuse pulmonary <strong>in</strong>filtrates and acute respiratory failure. Am RevRespir Dis 1988;137:90–4.27 Canver CC, Mentzer RM Jr. The role of open lung biopsy <strong>in</strong> early andlate survival of ventilator-dependent patients with diffuse idiopathic lungdisease. J Cardiovasc Surg 1994;35:151–5.28 Mo<strong>in</strong>e P, Vercken JB, Chevret S, et al. Severe community-acquiredpneumonia. Etiology, epidemiology, and prognosis factors. French StudyGroup for Community-Acquired Pneumonia <strong>in</strong> the Intensive <strong>Care</strong> Unit.Chest 1994;105:1487–95.29 Sorensen J, Forsberg P, Hakanson E, et al. A new diagnostic approachto the patient with severe pneumonia. Scand J Infect Dis1989;21:33–41.30 Potgieter PD, Hammond JM. Etiology and diagnosis of pneumoniarequir<strong>in</strong>g ICU admission. Chest 1992;101:199–203.31 Pachon J, Prados MD, Capote F, et al. Severe community-acquiredpneumonia. Etiology, prognosis, and treatment. Am Rev Respir Dis1990;142:369–73.32 Torres A, Serra-Batlles J, Ferrer A, et al. Severe community-acquiredpneumonia. Epidemiology and prognostic factors. Am Rev Respir Dis1991;144:312–8.33 Sole Violan J, Fernandez JA, Benitez AB, et al. Impact of quantitative<strong>in</strong>vasive diagnostic techniques <strong>in</strong> the management and outcome ofmechanically ventilated patients with suspected pneumonia. Crit <strong>Care</strong>Med 2000;28:2737–41.34 Crawford SW, Petersen FB. Long-term survival from respiratory failureafter marrow transplantation for malignancy. Am Rev Respir Dis1992;145:510–4.35 Gal AA, Klatt EC, Koss MN, et al. The effectiveness of bronchoscopy <strong>in</strong>the diagnosis of Pneumocystis car<strong>in</strong>ii and cytomegalovirus pulmonary<strong>in</strong>fections <strong>in</strong> acquired immunodeficiency syndrome. Arch Pathol Lab Med1987;111:238–41.36 Golden JA, Hollander H, Stulbarg MS, et al. Bronchoalveolar lavage asthe exclusive diagnostic modality for Pneumocystis car<strong>in</strong>ii pneumonia. Aprospective study among patients with acquired immunodeficiencysyndrome. Chest 1986;90:18–22.37 Broaddus C, Dake MD, Stulbarg MS, et al. Bronchoalveolar lavage andtransbronchial biopsy for the diagnosis of pulmonary <strong>in</strong>fections <strong>in</strong> theacquired immunodeficiency syndrome. Ann Intern Med1985;102:747–52.38 Jules-Elysee KM, Stover DE, Zaman MB, et al. Aerosolizedpentamid<strong>in</strong>e: effect on diagnosis and presentation of Pneumocystis car<strong>in</strong>iipneumonia. Ann Intern Med 1990;112:750–7.39 Lev<strong>in</strong>e SJ, Kennedy D, Shelhamer JH, et al. Diagnosis of Pneumocystiscar<strong>in</strong>ii pneumonia by multiple lobe, site-directed bronchoalveolar lavagewith immunofluorescent monoclonal antibody sta<strong>in</strong><strong>in</strong>g <strong>in</strong> humanimmunodeficiency virus-<strong>in</strong>fected patients receiv<strong>in</strong>g aerosolizedpentamid<strong>in</strong>e chemoprophylaxis. Am Rev Respir Dis 1992;146:838–43.40 Yung RC, We<strong>in</strong>acker AB, Steiger DJ, et al. Upper and middle lobebronchoalveolar lavage to diagnose Pneumocystis car<strong>in</strong>ii pneumonia. AmRev Respir Dis 1993;148(6 Pt 1):1563–6.41 Salomon N, Perlman DC. Cytomegalovirus pneumonia. Sem<strong>in</strong> RespirInfect 1999;14:353–8.42 Clelland C, Higenbottam T, Stewart S, et al. Bronchoalveolar lavageand transbronchial lung biopsy dur<strong>in</strong>g acute rejection and <strong>in</strong>fection <strong>in</strong>heart-lung transplant patients. Studies of cell counts, lymphocytephenotypes, and expression of HLA-DR and <strong>in</strong>terleuk<strong>in</strong>-2 receptor. AmRev Respir Dis 1993;147(6 Pt 1):1386–92.43 Rawl<strong>in</strong>son WD. Diagnosis of human cytomegalovirus <strong>in</strong>fection anddisease. Pathology 1999;31:109–15.44 Denn<strong>in</strong>g DW. Invasive aspergillosis. Cl<strong>in</strong> Infect Dis 1998;26:781–803;quiz 804–5.45 Stevens DA, Kan VL, Judson MA, et al. Practice guidel<strong>in</strong>es for diseasescaused by Aspergillus. Infectious Diseases Society of America. Cl<strong>in</strong> InfectDis 2000;30:696–709.46 McWh<strong>in</strong>ney PH, Kibbler CC, Hamon MD, et al. Progress <strong>in</strong> thediagnosis and management of aspergillosis <strong>in</strong> bone marrowtransplantation: 13 years’ experience. Cl<strong>in</strong> Infect Dis 1993;17:397–404.47 Denn<strong>in</strong>g DW, Evans EG, Kibbler CC, et al. Guidel<strong>in</strong>es for the<strong>in</strong>vestigation of <strong>in</strong>vasive fungal <strong>in</strong>fections <strong>in</strong> haematological malignancyand solid organ transplantation. British Society for Medical Mycology.Eur J Cl<strong>in</strong> Microbiol Infect Dis 1997;16:424–36.48 Kahn FW, Jones JM, England DM. The role of bronchoalveolar lavage <strong>in</strong>the diagnosis of <strong>in</strong>vasive pulmonary aspergillosis. Am J Cl<strong>in</strong> Pathol1986;86:518–23.49 Levy H, Horak DA, Tegtmeier BR, et al. The value of bronchoalveolarlavage and bronchial wash<strong>in</strong>gs <strong>in</strong> the diagnosis of <strong>in</strong>vasive pulmonaryaspergillosis. Respir Med 1992;86:243–8.50 Maertens J, Verhaegen J, Demuynck H, et al. Autopsy-controlledprospective evaluation of serial screen<strong>in</strong>g for circulat<strong>in</strong>g galactomannanby a sandwich enzyme-l<strong>in</strong>ked immunosorbent assay for hematologicalpatients at risk for <strong>in</strong>vasive aspergillosis. J Cl<strong>in</strong> Microbiol1999;37:3223–8.51 Klech H, Hutter C. Cl<strong>in</strong>ical guidel<strong>in</strong>es and <strong>in</strong>dications forbronchoalveolar lavage (BAL): report of the European Society ofPneumonology Task Group on BAL. Eur Respir J 1990;3:938–69.52 Allen JN, Davis WB, Pacht ER. Diagnostic significance of <strong>in</strong>creasedbronchoalveolar lavage fluid eos<strong>in</strong>ophils. Am Rev Respir Dis1990;142:642–7.53 Velay B, Pages J, Cordier JF, et al. Hypereos<strong>in</strong>ophilia <strong>in</strong> bronchoalveolarlavage. Diagnostic value and correlations with blood eos<strong>in</strong>ophilia. RevMal Respir 1987;4:257–60.54 Ste<strong>in</strong>berg KP, Milberg JA, Mart<strong>in</strong> TR, et al. Evolution of bronchoalveolarcell populations <strong>in</strong> the adult respiratory distress syndrome. Am J RespirCrit <strong>Care</strong> Med 1994;150:113–22.55 Meduri GU, Headley AS, Golden E, et al. Effect of prolongedmethylprednisolone therapy <strong>in</strong> unresolv<strong>in</strong>g acute respiratory distresssyndrome: a randomized controlled trial. JAMA 1998;280:159–65.56 Pittet JF, Mackersie RC, Mart<strong>in</strong> TR, et al. Biological markers of acutelung <strong>in</strong>jury: prognostic and pathogenetic significance. Am J Respir Crit<strong>Care</strong> Med 1997;155:1187–205.57 Donnelly SC, Strieter RM, Kunkel SL, et al. Interleuk<strong>in</strong>-8 anddevelopment of adult respiratory distress syndrome <strong>in</strong> at-risk patientgroups. Lancet 1993;341:643–7.58 Mart<strong>in</strong> C, Papazian L, Payan MJ, et al. Pulmonary fibrosis correlateswith outcome <strong>in</strong> adult respiratory distress syndrome. A study <strong>in</strong>mechanically ventilated patients. Chest 1995;107:196–200.59 Chesnutt AN, Matthay MA, Tibayan FA, et al. Early detection of type IIIprocollagen peptide <strong>in</strong> acute lung <strong>in</strong>jury. Pathogenetic and prognosticsignificance. Am J Respir Crit <strong>Care</strong> Med 1997;156(3 Pt 1):840–5.60 Adrie C, Monchi M, Tuan D<strong>in</strong>h-Xuan A, et al. Exhaled and nasal nitricoxide as a marker of pneumonia <strong>in</strong> ventilated patients. Am J Respir Crit<strong>Care</strong> Med 2001;163:1143–9.61 Brett SJ, Evans TW. Measurement of endogenous nitric oxide <strong>in</strong> thelungs of patients with the acute respiratory distress syndrome. Am J RespirCrit <strong>Care</strong> Med 1998;157(3 Pt 1):993–7.62 Carpenter CT, Price PV, Christman BW. Exhaled breath condensateisoprostanes are elevated <strong>in</strong> patients with acute lung <strong>in</strong>jury or ARDS.Chest 1998;114:1653–9.63 Schubert JK, Muller WP, Benz<strong>in</strong>g A, et al. Application of a new methodfor analysis of exhaled gas <strong>in</strong> critically ill patients. Intensive <strong>Care</strong> Med1998;24:415–21.64 Toc<strong>in</strong>o I. Chest imag<strong>in</strong>g <strong>in</strong> the <strong>in</strong>tensive care unit. Eur J Radiol1996;23:46–57.65 Maffessanti M, Berlot G, Bortolotto P. Chest roentgenology <strong>in</strong> the<strong>in</strong>tensive care unit: an overview. Eur Radiol 1998;8:69–78.66 Helfaer M. To chest x-rays and beyond. Crit <strong>Care</strong> Med1999;27:1676–7.67 Price MB, Grant MJ, Welkie K. F<strong>in</strong>ancial impact of elim<strong>in</strong>ation of rout<strong>in</strong>echest radiographs <strong>in</strong> a pediatric <strong>in</strong>tensive care unit. Crit <strong>Care</strong> Med1999;27:1588–93.68 American College of Radiology. ACR appropriateness criteria: rout<strong>in</strong>eportable daily X-ray. 1999.69 Bekemeyer WB, Crapo RO, Calhoon S, et al. Efficacy of chestradiography <strong>in</strong> a respiratory <strong>in</strong>tensive care unit. A prospective study.Chest 1985;88:691–6.70 Greenbaum DM, Marschall KE. The value of rout<strong>in</strong>e daily chest x-rays<strong>in</strong> <strong>in</strong>tubated patients <strong>in</strong> the medical <strong>in</strong>tensive care unit. Crit <strong>Care</strong> Med1982;10:29–30.71 Stra<strong>in</strong> DS, K<strong>in</strong>asewitz GT, Vereen LE, et al. Value of rout<strong>in</strong>e daily chestx-rays <strong>in</strong> the medical <strong>in</strong>tensive care unit. Crit <strong>Care</strong> Med 1985;13:534–6.72 American College of Radiology. ACR standards for the performance ofpediatric and adult bedside (portable) chest radiography. 1997.73 Dunbar RD. Radiologic appearance of compromised thoracic catheters,tubes, and wires. Radiol Cl<strong>in</strong> North Am 1984;22:699–722.74 Conrardy PA, Goodman LR, La<strong>in</strong>ge F, et al. Alteration of endotrachealtube position. Flexion and extension of the neck. Crit <strong>Care</strong> Med1976;4:7–12.75 Goodman LR, Conrardy PA, La<strong>in</strong>g F, et al. Radiographic evaluation ofendotracheal tube position. AJR 1976;127:433–4.76 Satyadas T, Nasir N, Erel E, Mudan SS. Latrogenic tracheal rupture: anovel approach to repair and a review of the literature. J Trauma2003;54:369–71.77 Tsao MM, Katz D. Central venous catheter-<strong>in</strong>duced endocarditis: humancorrelate of the animal experimental model of endocarditis. Rev Infect Dis1984;6:783–90.78 Gomez CM, Palazzo MG. Pulmonary artery catheterization <strong>in</strong>anaesthesia and <strong>in</strong>tensive care. Br J Anaesth 1998;81:945–56.79 Thomason JW, Ely EW, Chiles C, et al. Apprais<strong>in</strong>g pulmonary edemaus<strong>in</strong>g sup<strong>in</strong>e chest roentgenograms <strong>in</strong> ventilated patients. Am J Respir Crit<strong>Care</strong> Med 1998;157(5 Pt 1):1600–8.80 Milne EN, Pistolesi M, M<strong>in</strong>iati M, et al. The radiologic dist<strong>in</strong>ction ofcardiogenic and noncardiogenic edema. AJR 1985;144:879–94.81 Emamian SA, Kaasbol MA, Olsen JF, et al. Accuracy of the diagnosis ofpleural effusion on sup<strong>in</strong>e chest X-ray. Eur Radiol 1997;7:57–60.82 Eibenberger KL, Dock WI, Ammann ME, et al. Quantification of pleuraleffusions: sonography versus radiography. Radiology 1994;191:681–4.


10 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>83 Agusti AG, Cardus J, Roca J, et al. Ventilation-perfusion mismatch <strong>in</strong>patients with pleural effusion: effects of thoracentesis. Am J Respir Crit<strong>Care</strong> Med 1997;156(4 Pt 1):1205–9.84 Talmor M, Hydo L, Gershenwald JG, et al. Beneficial effects of chesttube dra<strong>in</strong>age of pleural effusion <strong>in</strong> acute respiratory failure refractory topositive end-expiratory pressure ventilation. Surgery 1998;123:137–43.85 Lichtenste<strong>in</strong> D, Hulot JS, Rabiller A, et al. Feasibility and safety ofultrasound-aided thoracentesis <strong>in</strong> mechanically ventilated patients.Intensive <strong>Care</strong> Med 1999;25:955–8.86 Keske U. Ultrasound-aided thoracentesis <strong>in</strong> <strong>in</strong>tensive care patients.Intensive <strong>Care</strong> Med 1999;25:896–7.87 Miller WT Jr, T<strong>in</strong>o G, Friedburg JS. Thoracic CT <strong>in</strong> the <strong>in</strong>tensive careunit: assessment of cl<strong>in</strong>ical usefulness. Radiology 1998;209:491–8.88 Intensive <strong>Care</strong> Society. Guidel<strong>in</strong>es for the transport of the critically illadult. 1997.89 White CS, Meyer CA, Wu J, et al. Portable CT: assess<strong>in</strong>g thoracicdisease <strong>in</strong> the <strong>in</strong>tensive care unit. AJR 1999;173:1351–6.90 Gatt<strong>in</strong>oni L, Presenti A, Torres<strong>in</strong> A, et al. Adult respiratory distresssyndrome profiles by computed tomography. J Thorac Imag<strong>in</strong>g1986;1:25–30.91 Desai SR, Wells AU, Suntharal<strong>in</strong>gam G, et al. Acute respiratory distresssyndrome caused by pulmonary and extrapulmonary <strong>in</strong>jury: acomparative CT study. Radiology 2001;218:689–93.92 Pelosi P, Crotti S, Brazzi L, et al. Computed tomography <strong>in</strong> adultrespiratory distress syndrome: what has it taught us? Eur Respir J1996;9:1055–62.93 Wiener CM, Kirk W, Albert RK. Prone position reverses gravitationaldistribution of perfusion <strong>in</strong> dog lungs with oleic acid-<strong>in</strong>duced <strong>in</strong>jury. JAppl Physiol 1990;68:1386–92.94 Snow N, Berg<strong>in</strong> KT, Horrigan TP. Thoracic CT scann<strong>in</strong>g <strong>in</strong> critically illpatients. Information obta<strong>in</strong>ed frequently alters management. Chest1990;97:1467–70.95 Guerrero-Lopez F, Vazquez-Mata G, Alcazar-Romero PP, et al.Evaluation of the utility of computed tomography <strong>in</strong> the <strong>in</strong>itial assessmentof the critical care patient with chest trauma. Crit <strong>Care</strong> Med2000;28:1370–5.96 Enderson BL, Abdalla R, Frame SB, et al. Tube thoracostomy for occultpneumothorax: a prospective randomized study of its use. J Trauma-InjuryInfect Crit <strong>Care</strong> 1993;35:726–9; discussion 729–30.97 Wolfman NT, Gilp<strong>in</strong> JW, Bechtold RE, et al. Occult pneumothorax <strong>in</strong>patients with abdom<strong>in</strong>al trauma: CT studies. J Comput Assist Tomogr1993;17:56–9.98 Garramone RR Jr, Jacobs LM, Sahdev P. An objective method tomeasure and manage occult pneumothorax. Surg Gynecol Obstet1991;173:257–61.99 Watson AC, Hughes PD, Louise Harris M, et al. Measurement of twitchtransdiaphragmatic, esophageal, and endotracheal tube pressure withbilateral anterolateral magnetic phrenic nerve stimulation <strong>in</strong> patients <strong>in</strong>the <strong>in</strong>tensive care unit. Crit <strong>Care</strong> Med 2001;29:1325–31.100 Yang KL, Tob<strong>in</strong> MJ. A prospective study of <strong>in</strong>dexes predict<strong>in</strong>g theoutcome of trials of wean<strong>in</strong>g from mechanical ventilation. N Engl J Med1991;324:1445–50.101 Dantzker DR, Brook CJ, Dehart P, et al. Ventilation-perfusiondistributions <strong>in</strong> the adult respiratory distress syndrome. Am Rev Respir Dis1979;120:1039–52.102 Krafft P, Fridrich P, Pernerstorfer T, et al. The acute respiratory distresssyndrome: def<strong>in</strong>itions, severity and cl<strong>in</strong>ical outcome. An analysis of 101cl<strong>in</strong>ical <strong>in</strong>vestigations. Intensive <strong>Care</strong> Med 1996;22:519–29.103 Covelli HD, Nessan VJ, Tuttle WK 3rd. Oxygen derived variables <strong>in</strong>acute respiratory failure. Crit <strong>Care</strong> Med 1983;11:646–9.104 Mar<strong>in</strong>i JJ, Ravenscraft SA. Mean airway pressure: physiologicdeterm<strong>in</strong>ants and cl<strong>in</strong>ical importance – part 2: cl<strong>in</strong>ical implications. Crit<strong>Care</strong> Med 1992;20:1604–16.105 Villar J, Perez-Mendez L, Kacmarek RM. Current def<strong>in</strong>itions of acute lung<strong>in</strong>jury and the acute respiratory distress syndrome do not reflect their trueseverity and outcome. Intensive <strong>Care</strong> Med 1999;25:930–5.106 The Acute <strong>Respiratory</strong> Distress Syndrome Network. Ventilation withlower tidal volumes as compared with traditional tidal volumes for acutelung <strong>in</strong>jury and the acute respiratory distress syndrome. N Engl J Med2000;342:1301–8.107 Roupie E, Dambrosio M, Servillo G, et al. Titration of tidal volume and<strong>in</strong>duced hypercapnia <strong>in</strong> acute respiratory distress syndrome. Am J RespirCrit <strong>Care</strong> Med 1995;152:121–8.108 Dreyfuss D, Saumon G. Pressure-volume curves. Search<strong>in</strong>g for the grailor lay<strong>in</strong>g patients with adult respiratory distress syndrome on Procrustes’bed? Am J Respir Crit <strong>Care</strong> Med 2001;163:2–3.


2 Oxygen delivery and consumption <strong>in</strong> the critically illR M Leach, D F Treacher.............................................................................................................................Although traditionally <strong>in</strong>terested <strong>in</strong> conditionsaffect<strong>in</strong>g gas exchange with<strong>in</strong> thelungs, the respiratory physician is <strong>in</strong>creas<strong>in</strong>gly,and appropriately, <strong>in</strong>volved <strong>in</strong> the care ofcritically ill patients and therefore should beconcerned with systemic as well as pulmonaryoxygen transport. Oxygen is the substrate that cellsuse <strong>in</strong> the greatest quantity and upon which aerobicmetabolism and cell <strong>in</strong>tegrity depend. S<strong>in</strong>ce thetissues have no storage system for oxygen, acont<strong>in</strong>uous supply at a rate that matches chang<strong>in</strong>gmetabolic requirements is necessary to ma<strong>in</strong>ta<strong>in</strong>aerobic metabolism and normal cellular function.Failure of oxygen supply to meet metabolic needs isthe feature common to all forms of circulatory failureor “shock”. Prevention, early identification, andcorrection of tissue hypoxia are therefore necessaryskills <strong>in</strong> manag<strong>in</strong>g the critically ill patient and thisrequires an understand<strong>in</strong>g of oxygen transport,delivery, and consumption.OXYGEN TRANSPORTOxygen transport describes the process by whichoxygen from the atmosphere is supplied to thetissues as shown <strong>in</strong> fig 2.1 <strong>in</strong> which typical valuesare quoted for a healthy 75 kg <strong>in</strong>dividual. Thephases <strong>in</strong> this process are either convective or diffusive:(1) the convective or “bulk flow” phasesare alveolar ventilation and transport <strong>in</strong> the bloodfrom the pulmonary to the systemic microcirculation:these are energy requir<strong>in</strong>g stages that rely onwork performed by the respiratory and cardiac“pumps”; and (2) the diffusive phases are themovement of oxygen from alveolus to pulmonarycapillary and from systemic capillary to cell: thesestages are passive and depend on the gradient ofoxygen partial pressures, the tissue capillary density(which determ<strong>in</strong>es diffusion distance), andthe ability of the cell to take up and use oxygen.This chapter will not consider oxygen transportwith<strong>in</strong> the lungs but will focus on transport fromthe heart to non-pulmonary tissues, deal<strong>in</strong>g specificallywith global and regional oxygen delivery,the relationship between oxygen delivery andconsumption, and some of the recent evidencerelat<strong>in</strong>g to the uptake and utilisation of oxygen atthe tissue and cellular level.OXYGEN DELIVERYGlobal oxygen delivery (DO 2) is the total amountof oxygen delivered to the tissues per m<strong>in</strong>ute irrespectiveof the distribution of blood flow. Underrest<strong>in</strong>g conditions with normal distribution ofcardiac output it is more than adequate to meetthe total oxygen requirements of the tissues (VO 2)and ensure that aerobic metabolism is ma<strong>in</strong>ta<strong>in</strong>ed.Recognition of <strong>in</strong>adequate global DO 2can bedifficult <strong>in</strong> the early stages because the cl<strong>in</strong>icalfeatures are often non-specific. Progressive metabolicacidosis, hyperlactataemia, and fall<strong>in</strong>gmixed venous oxygen saturation (SvO 2), as well asorgan specific features such as oliguria andimpaired level of consciousness, suggest <strong>in</strong>adequateDO 2. Serial lactate measurements can <strong>in</strong>dicateboth progression of the underly<strong>in</strong>g problemand the response to treatment. Raised lactate levels(>2 mmol/l) may be caused by either <strong>in</strong>creasedproduction or reduced hepatic metabolism.Both mechanisms frequently apply <strong>in</strong> thecritically ill patient s<strong>in</strong>ce a marked reduction <strong>in</strong>DO 2produces global tissue ischaemia and impairsliver function.Table 2.1 illustrates the calculation of DO 2fromthe oxygen content of arterial blood (CaO 2) andcardiac output (Qt) with examples for a normalsubject and a patient present<strong>in</strong>g with hypoxaemia,anaemia, and a reduced Qt. The effects ofprovid<strong>in</strong>g an <strong>in</strong>creased <strong>in</strong>spired oxygen concentration,red blood cell transfusion, and <strong>in</strong>creas<strong>in</strong>gcardiac output are shown. This emphasises that:(1) DO 2may be compromised by anaemia, oxygendesaturation, and a low cardiac output, eithers<strong>in</strong>gly or <strong>in</strong> comb<strong>in</strong>ation; (2) global DO 2dependson oxygen saturation rather than partial pressureand there is therefore little extra benefit <strong>in</strong><strong>in</strong>creas<strong>in</strong>g PaO 2above 9 kPa s<strong>in</strong>ce, due to the sigmoidshape of the oxyhaemoglob<strong>in</strong> dissociationcurve, over 90% of haemoglob<strong>in</strong> (Hb) is alreadysaturated with oxygen at that level. This does notapply to the diffusive component of oxygentransport that does depend on the gradient ofoxygen partial pressure.Although blood transfusion to polycythaemiclevels might seem an appropriate way to <strong>in</strong>creaseDO 2, blood viscosity <strong>in</strong>creases markedly above100 g/l. This impairs flow and oxygen delivery,particularly <strong>in</strong> smaller vessels and when the perfusionpressure is reduced, and will thereforeexacerbate tissue hypoxia. 1 Recent evidencesuggests that even the traditionally accepted Hbconcentration for critically ill patients of approximately100 g/l may be too high s<strong>in</strong>ce an improvedoutcome was observed if Hb was ma<strong>in</strong>ta<strong>in</strong>edbetween 70 and 90 g/l with the exception ofpatients with coronary artery disease <strong>in</strong> whom alevel of 100 g/l rema<strong>in</strong>s appropriate. 2With theappropriate Hb achieved by transfusion, and s<strong>in</strong>cethe oxygen saturation (SaO 2) can usually bema<strong>in</strong>ta<strong>in</strong>ed above 90% with supplemental oxygen(or if necessary by <strong>in</strong>tubation and mechanicalventilation), cardiac output is the variable that ismost often manipulated to achieve the desiredglobal DO 2levels..................................................Abbreviations: SO 2, oxygen saturation (%); PO 2, oxygenpartial pressure (kPa); PIO 2, <strong>in</strong>spired PO 2 ;PEO 2, mixedexpired PO 2 ;PECO 2, mixed expired PCO 2 ;PAO 2, alveolarPO 2;PaO 2, arterial PO 2;SaO 2, arterial SO 2;SvO 2, mixedvenous SO 2; Qt, cardiac output; Hb, haemoglob<strong>in</strong>; CaO 2,arterial O 2content; CvO 2, mixed venous O 2content; VO 2,oxygen consumption; VCO 2,CO 2production; O 2R, oxygenreturn; DO 2, oxygen delivery; Vi/e, m<strong>in</strong>ute volume,<strong>in</strong>spiratory/expiratory.


12 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Inspired (dry)PIO 2 (21)Expired (dry)PEO 2 (15.9)PECO 2 (4.2)PaO 2 (P 50 )PAO 2 RALVLAVi/e(5)Lungs ‘shunt’RV(14)(13) SaO 2 (97) CaO 2 (200)DO 2Hb (150) Q t (5)(1000)Diffusion of O 2 <strong>in</strong> tissuesvenous(5.3)CapillaryInterstitial(5.3–2.7)Intracellular(2.7–1.3)arterial(13)Mitochondrial(1.3–0.7)VO 2(250)VCO 2(200)Q t(5)Q tHb (150)Q tPvO 2 (5.3) (P 50 ) SvO 2 (75) CvO 2 (150)O 2 R(750)Figure 2.1 Oxygen transport from atmosphere to mitochondria. Values <strong>in</strong> parentheses for a normal 75 kg <strong>in</strong>dividual (BSA 1.7 m 2 ) breath<strong>in</strong>gair (FIO 20.21) at standard atmospheric pressure (P B101 kPa). Partial pressures of O 2and CO 2(PO 2 ,PCO 2) <strong>in</strong> kPa; saturation <strong>in</strong> %; contents(CaO 2,CvO 2) <strong>in</strong> ml/l; Hb <strong>in</strong> g/l; blood/gas flows (Qt, Vi/e) <strong>in</strong> l/m<strong>in</strong>. P 50= position of oxygen haemoglob<strong>in</strong> dissociation curve; it is PO 2at which50% of haemoglob<strong>in</strong> is saturated (normally 3.5 kPa). DO 2= oxygen delivery; VO 2= oxygen consumption, VCO 2= carbon dioxide production;PIO 2 ,PEO 2= <strong>in</strong>spired and mixed expired PO 2 ;PEC O 2= mixed expired PCO 2 ;PAO 2= alveolar PO 2.OXYGEN CONSUMPTIONGlobal oxygen consumption (VO 2) measures the total amountof oxygen consumed by the tissues per m<strong>in</strong>ute. It can bemeasured directly from <strong>in</strong>spired and mixed expired oxygenconcentrations and expired m<strong>in</strong>ute volume, or derived fromthe cardiac output (Qt) and arterial and venous oxygencontents:VO 2=Qt×(CaO 2–CvO 2)Directly measured VO 2is slightly greater than the derivedvalue that does not <strong>in</strong>clude alveolar oxygen consumption. It isimportant to use the directly measured rather than thederived value when study<strong>in</strong>g the relationship between VO 2andDO 2to avoid problems of mathematical l<strong>in</strong>kage. 3The amount of oxygen consumed (VO 2) as a fraction of oxygendelivery (DO 2) def<strong>in</strong>es the oxygen extraction ratio (OER):OER=VO 2/DO 2In a normal 75 kg adult undertak<strong>in</strong>g rout<strong>in</strong>e activities, VO 2is approximately 250 ml/m<strong>in</strong> with an OER of 25% (fig 2.1),which <strong>in</strong>creases to 70–80% dur<strong>in</strong>g maximal exercise <strong>in</strong> thewell tra<strong>in</strong>ed athlete. The oxygen not extracted by the tissuesreturns to the lungs and the mixed venous saturation (SvO 2)measured <strong>in</strong> the pulmonary artery represents the pooledvenous saturation from all organs. It is <strong>in</strong>fluenced by changes<strong>in</strong> both global DO 2and VO 2and, provided the microcirculationand the mechanisms for cellular oxygen uptake are <strong>in</strong>tact, avalue above 70% <strong>in</strong>dicates that global DO 2is adequate.A mixed venous sample is necessary because the saturationof venous blood from different organs varies considerably. Forexample, the hepatic venous saturation is usually 40–50% butthe renal venous saturation may exceed 80%, reflect<strong>in</strong>g theconsiderable difference <strong>in</strong> the balance between the metabolicrequirements of these organs and their <strong>in</strong>dividual oxygendeliveries.CLINICAL FACTORS AFFECTING METABOLIC RATEAND OXYGEN CONSUMPTIONThe cellular metabolic rate determ<strong>in</strong>es VO 2. The metabolic rate<strong>in</strong>creases dur<strong>in</strong>g physical activity, with shiver<strong>in</strong>g, hyperthermiaand raised sympathetic drive (pa<strong>in</strong>, anxiety). Similarly,certa<strong>in</strong> drugs such as adrenal<strong>in</strong>e 4and feed<strong>in</strong>g regimensconta<strong>in</strong><strong>in</strong>g excessive glucose <strong>in</strong>crease VO 2. Mechanical ventilationelim<strong>in</strong>ates the metabolic cost of breath<strong>in</strong>g which,although normally less than 5% of the total VO 2, may rise to30% <strong>in</strong> the catabolic critically ill patient with respiratorydistress. It allows the patient to be sedated, given analgesiaand, if necessary, paralysed, further reduc<strong>in</strong>g VO 2.Table 2.1 Relative effects of changes <strong>in</strong> PaO 2, haemoglob<strong>in</strong> (Hb), and cardiac output (Qt) on oxygen delivery (DO 2)FIO 2 PaO 2 (kPa) SaO 2 (%) Hb (g/l)Dissolved O 2(ml/l) CaO 2 (ml/l) Qt (l/m<strong>in</strong>) DO 2 (ml/m<strong>in</strong>) DO 2 (% change)‡Normal* 0.21 13.0 96 130 3.0 170 5.3 900 0Patient† 0.21 6.0 75 70 1.4 72 4.0 288 – 68↑FIO 2 0.35 9.0 92 70 2.1 88 4.0 352 + 22↑↑FIO 2 0.60 16.5 98 70 3.8 96 4.0 384 + 9↑Hb 0.60 16.5 98 105 3.8 142 4.0 568 +48↑Qt 0.60 16.5 98 105 3.8 142 6.0 852 +50DO 2 =CaO 2 ×Qt ml/m<strong>in</strong>, CaO 2 = (Hb × SaO 2 × 1.34) + (PaO 2 × 0.23) ml/l where FIO 2 = fractional <strong>in</strong>spired oxygen concentration; PaO 2 ,SaO 2 ,CaO 2 =partial pressure, saturation and content of oxygen <strong>in</strong> arterial blood; Qt = cardiac output. 1.34 ml is the volume of oxygen carried by 1gof100%saturated Hb. PaO 2 (kPa) × 0.23 is the amount of oxygen <strong>in</strong> physical solution <strong>in</strong> 1lofblood, which is less than


Oxygen delivery and consumption <strong>in</strong> the critically ill 13Oxygen consumption (VO 2 ) (ml/m<strong>in</strong>)200100ADB400 800 1200Figure 2.2 Relationship between oxygen delivery andconsumption.RELATIONSHIP BETWEEN OXYGEN CONSUMPTIONAND DELIVERYThe normal relationship between VO 2and DO 2is illustrated byl<strong>in</strong>e ABC <strong>in</strong> fig 2.2. As metabolic demand (VO 2) <strong>in</strong>creases or DO 2dim<strong>in</strong>ishes (C–B), OER rises to ma<strong>in</strong>ta<strong>in</strong> aerobic metabolismand consumption rema<strong>in</strong>s <strong>in</strong>dependent of delivery. However,at po<strong>in</strong>t B—called critical DO 2(cDO 2)—the maximum OER isreached. This is believed to be 60–70% and beyond this po<strong>in</strong>tany further <strong>in</strong>crease <strong>in</strong> VO 2or decl<strong>in</strong>e <strong>in</strong> DO 2must lead to tissuehypoxia. 5 In reality there is a family of such VO 2/DO 2relationshipswith each tissue/organ hav<strong>in</strong>g a unique VO 2/DO 2relationshipand value for maximum OER that may vary withstress and disease states. Although the technology currentlyavailable makes it impracticable to determ<strong>in</strong>e these organspecific relationships <strong>in</strong> the critically ill patient, it is importantto realise that conclusions drawn about the genesis of<strong>in</strong>dividual organ failure from the “global” diagram are potentiallyflawed.In critical illness, particularly <strong>in</strong> sepsis, an altered globalrelationship is believed to exist (broken l<strong>in</strong>e DEF <strong>in</strong> fig 2.2).The slope of maximum OER falls (DE v AB), reflect<strong>in</strong>g thereduced ability of tissues to extract oxygen, and the relationshipdoes not plateau as <strong>in</strong> the normal relationship. Henceconsumption cont<strong>in</strong>ues to <strong>in</strong>crease (E–F) to “supranormal”levels of DO 2, demonstrat<strong>in</strong>g so called “supply dependency”and the presence of a covert oxygen debt that would berelieved by further <strong>in</strong>creas<strong>in</strong>g DO 2. 6The relationship between global DO 2and VO 2<strong>in</strong> critically illpatients has received considerable attention over the past twodecades. Shoemaker and colleagues demonstrated a relationshipbetween DO 2and VO 2<strong>in</strong> the early postoperative phase thathad prognostic implications such that patients with highervalues had an improved survival. 7 A subsequent randomisedplacebo controlled trial <strong>in</strong> a similar group of patients showedimproved survival if the values for DO 2(>600 ml/m<strong>in</strong>/m 2 ) andSvO 2(>70%) that had been achieved by the survivors <strong>in</strong> theearlier study were set as therapeutic targets (“goal directedtherapy”). 8This evidence encouraged the use of “goal directed therapy”<strong>in</strong> patients with established (“late”) septic shock and organdysfunction <strong>in</strong> the belief that this strategy would <strong>in</strong>crease VO 2and prevent multiple organ failure. DO 2was <strong>in</strong>creased us<strong>in</strong>gvigorous <strong>in</strong>travenous fluid load<strong>in</strong>g and <strong>in</strong>otropes, usually dobutam<strong>in</strong>e.The mathematical l<strong>in</strong>kage caused by calculat<strong>in</strong>gboth VO 2and DO 2us<strong>in</strong>g common measurements of Qt andCaO 23and the “physiological” l<strong>in</strong>kage result<strong>in</strong>g from the metaboliceffects of <strong>in</strong>otropes <strong>in</strong>creas<strong>in</strong>g both VO 2and DO 2wereconfound<strong>in</strong>g factors <strong>in</strong> many of these studies. 9 This approachwas also responsible for a considerable <strong>in</strong>crease <strong>in</strong> the use ofpulmonary artery catheters to direct treatment. However, aftera decade of conflict<strong>in</strong>g evidence from numerous small, oftenmethodologically flawed studies, two major randomisedEOxygen delivery (DO 2 ) (ml/m<strong>in</strong>)FCcontrolled studies f<strong>in</strong>ally showed that there was no benefitand possibly harm from apply<strong>in</strong>g this approach <strong>in</strong> patientswith established “shock”. 10 11 Interest<strong>in</strong>gly, these studies alsofound that those patients who neither <strong>in</strong>creased their DO 2spontaneously nor <strong>in</strong> response to treatment had a particularlypoor outcome. This suggested that patients with late “shock”had “poor physiological reserve” with myocardial and otherorgan failure caused by fundamental cellular dysfunction.These changes would be unresponsive to Shoemaker’s goalsthat had been successful <strong>in</strong> “early” shock. Indeed, one mightpredict that, <strong>in</strong> patients with the <strong>in</strong>creased endothelialpermeability and myocardial dysfunction that typifies late“shock”, aggressive fluid load<strong>in</strong>g would produce widespreadtissue oedema impair<strong>in</strong>g both pulmonary gas exchange andtissue oxygen diffusion. The reported <strong>in</strong>crease <strong>in</strong> mortalityassociated with the use of pulmonary artery catheters 12 mayreflect the adverse effects of their use <strong>in</strong> attempt<strong>in</strong>g to achievesupranormal levels of DO 2.SHOULD GOAL DIRECTED THERAPY BEABANDONED?Recent studies exam<strong>in</strong><strong>in</strong>g perioperative “optimisation” <strong>in</strong>patients, many of whom also had significant pre-exist<strong>in</strong>g cardiopulmonarydysfunction, have confirmed that identify<strong>in</strong>gand treat<strong>in</strong>g volume depletion and poor myocardial performanceat an early stage is beneficial. 13–16 This was the messagefrom Shoemaker’s studies 20 years ago, but unfortunately itwas over<strong>in</strong>terpreted and applied to <strong>in</strong>appropriate patientpopulations caus<strong>in</strong>g the confusion that has only recently beenresolved. Thus, adequate volume replacement <strong>in</strong> relatively volumedepleted perioperative patients is entirely appropriate.However, the strategy of us<strong>in</strong>g aggressive fluid replacementand vasoactive agents <strong>in</strong> pursuit of supranormal “global” goalsdoes not improve survival <strong>in</strong> patients present<strong>in</strong>g late with<strong>in</strong>cipient or established multiorgan failure.This saga highlights the difference between “early” and“late” shock and the concept well known to traumatologists asthe “golden hour”. Of the various forms of circulatory shock,two dist<strong>in</strong>ct groups can be def<strong>in</strong>ed: those with hypovolaemic,cardiogenic, and obstructive forms of shock (group 1) have theprimary problem of a low cardiac output impair<strong>in</strong>g DO 2; thosewith septic, anaphylactic, and neurogenic shock (group 2)have a problem with the distribution of DO 2between andwith<strong>in</strong> organs—that is, abnormalities of regional DO 2<strong>in</strong> additionto any impairment of global DO 2. Sepsis is also associatedwith cellular/metabolic defects that impair the uptake andutilisation of oxygen by cells. Prompt effective treatment of“early” shock may prevent progression to “late” shock andorgan failure. In group 1 the peripheral circulatory response isphysiologically appropriate and, if the global problem iscorrected by <strong>in</strong>travenous fluid adm<strong>in</strong>istration, improvement<strong>in</strong> myocardial function or relief of the obstruction, the peripheraltissue consequences of prolonged <strong>in</strong>adequacy of globalDO 2will not develop. However, if there is delay <strong>in</strong> <strong>in</strong>stitut<strong>in</strong>geffective treatment, then shock becomes established andorgan failure supervenes. Once this late stage has beenreached, manipulation of the “global” or convective componentsof DO 2alone will be <strong>in</strong>effective. Global DO 2should nonethelessbe ma<strong>in</strong>ta<strong>in</strong>ed by fluid resuscitation to correcthypovolaemia and <strong>in</strong>otropes to support myocardial dysfunction.REGIONAL OXYGEN DELIVERYHypoxia <strong>in</strong> specific organs is often the result of disorderedregional distribution of blood flow both between and with<strong>in</strong>organs rather than <strong>in</strong>adequacy of global DO 2. 17 The importanceof regional factors <strong>in</strong> determ<strong>in</strong><strong>in</strong>g tissue oxygenation shouldnot be surpris<strong>in</strong>g s<strong>in</strong>ce, under physiological conditions ofmetabolic demand such as exercise, alterations <strong>in</strong> local vasculartone ensure the necessary <strong>in</strong>crease <strong>in</strong> regional and overall


14 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Figure 2.3 Example of tissue ischaemia and necrosis fromextensive microvascular and macrovascular occlusion <strong>in</strong> a patientwith severe men<strong>in</strong>gococcal sepsis.blood flow—that is, “consumption drives delivery”. It istherefore important to dist<strong>in</strong>guish between global andregional DO 2when consider<strong>in</strong>g the cause of tissue hypoxia <strong>in</strong>specific organs. Loss of normal autoregulation <strong>in</strong> response tohumoral factors dur<strong>in</strong>g sepsis or prolonged hypotension cancause severe “shunt<strong>in</strong>g” and tissue hypoxia despite both globalDO 2and SvO 2be<strong>in</strong>g normal or raised. 18 In thesecircumstances, improv<strong>in</strong>g peripheral distribution and cellularoxygen utilisation will be more effective than further <strong>in</strong>creas<strong>in</strong>gglobal DO 2. Regional and microcirculatory distribution ofcardiac output is determ<strong>in</strong>ed by a complex <strong>in</strong>teraction ofendothelial, neural, metabolic, and pharmacological factors.In health, many of these processes have been <strong>in</strong>tensively<strong>in</strong>vestigated and well reviewed elsewhere. 19Until recently the endothelium had been perceived as an<strong>in</strong>ert barrier but it is now realised that it has a profound effecton vascular homeostasis, act<strong>in</strong>g as a dynamic <strong>in</strong>terfacebetween the underly<strong>in</strong>g tissue and the many components offlow<strong>in</strong>g blood. In concert with other vessel wall cells, theendothelium not only ma<strong>in</strong>ta<strong>in</strong>s a physical barrier betweenthe blood and body tissues but also modulates leucocytemigration, angiogenesis, coagulation, and vascular tonethrough the release of both constrictor (endothel<strong>in</strong>) andrelax<strong>in</strong>g factors (nitric oxide, prostacycl<strong>in</strong>, adenos<strong>in</strong>e). 20 Thedifferential release of such factors has an important role <strong>in</strong>controll<strong>in</strong>g the distribution of regional blood flow dur<strong>in</strong>g bothhealth and critical illness. The endothelium is both exposed toand itself produces many <strong>in</strong>flammatory mediators that <strong>in</strong>fluencevascular tone and other aspects of endothelial function.For example, nitric oxide production is <strong>in</strong>creased <strong>in</strong> septicshock follow<strong>in</strong>g <strong>in</strong>duction of nitric oxide synthase <strong>in</strong> the vesselwall. Inhibition of nitric oxide synthesis <strong>in</strong>creased vascularresistance and systemic blood pressure <strong>in</strong> patients with septicshock, but no outcome benefit could be demonstrated. 21 Similarly,capillary microthrombosis follow<strong>in</strong>g endothelial damageand neutrophil activation is probably a more common cause oflocal tissue hypoxia than arterial hypoxaemia (fig 2.3).Manipulation of the coagulation system, for example, us<strong>in</strong>gactivated prote<strong>in</strong> C may reduce this thrombotic tendency andimprove outcome as shown <strong>in</strong> a recent randomised, placebocontrolled, multicentre study <strong>in</strong> patients with severe sepsis. 22The cl<strong>in</strong>ical implications of disordered regional blood flowdistribution vary considerably with the underly<strong>in</strong>g pathologicalprocess. In the critically ill patient splanchnic perfusion isreduced by the release of endogenous vasoconstrictors and thegut mucosa is frequently further compromised by failure toma<strong>in</strong>ta<strong>in</strong> enteral nutrition. In sepsis and experimental endotoxaemiathe oxygen extraction ratio is reduced and the criticalDO 2<strong>in</strong>creased to a greater extent <strong>in</strong> splanchnic tissue than<strong>in</strong> skeletal muscle. 23This tendency to splanchnic ischaemiarenders the gut mucosa “leaky”, allow<strong>in</strong>g translocation ofendotox<strong>in</strong> and possibly bacteria <strong>in</strong>to the portal circulation.This toxic load may overwhelm hepatic clearance produc<strong>in</strong>gwidespread endothelial damage. Treatment aimed at ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>gor improv<strong>in</strong>g splanchnic perfusion reduces the<strong>in</strong>cidence of multiple organ failure and mortality. 24Although <strong>in</strong>creas<strong>in</strong>g global DO 2may improve blood flow toregionally hypoxic tissues by rais<strong>in</strong>g blood flow through allcapillary beds, this is an <strong>in</strong>efficient process and, if achievedus<strong>in</strong>g vasoactive drugs, may adversely affect regional distribution,particularly to the kidneys and splanchnic beds. Thepotent α receptor agonist noradrenal<strong>in</strong>e is frequently used tocounteract sepsis <strong>in</strong>duced vasodilation and hypotension. The<strong>in</strong>crease <strong>in</strong> blood pressure may improve perfusion to certa<strong>in</strong>hypoxia sensitive vital organs but may also compromise bloodflow to other organs, particularly the splanchnic bed. The roleof vasodilators is less well def<strong>in</strong>ed: tissue perfusion isfrequently already compromised by systemic hypotension anda reduced systemic vascular resistance, and their effect onregional distribution is unpredictable and may impair bloodflow to vital organs despite <strong>in</strong>creas<strong>in</strong>g global DO 2. In a group ofcritically ill patients prostacycl<strong>in</strong> <strong>in</strong>creased both DO 2and VO 2and this was <strong>in</strong>terpreted as <strong>in</strong>dicat<strong>in</strong>g that there was a previouslyunidentified oxygen debt. However, there is no conv<strong>in</strong>c<strong>in</strong>gevidence that vasodilators improve outcome <strong>in</strong> critically illpatients. An alternative strategy that attempts to redirectblood flow from overperfused non-essential tissues such assk<strong>in</strong> and muscle tissues to underperfused “vital” organs byexploit<strong>in</strong>g the differences <strong>in</strong> receptor population and densitybetween different arteries is theoretically attractive. Whiledobutam<strong>in</strong>e may reduce splanchnic perfusion, dopexam<strong>in</strong>ehydrochloride has dopam<strong>in</strong>ergic and β-adrenergic but noα-adrenergic effects and may selectively <strong>in</strong>crease renal andsplanchnic blood flow. 25OXYGEN TRANSPORT FROM CAPILLARY BLOOD TOINDIVIDUAL CELLSThe delivery of oxygen from capillary blood to the cell dependson:• factors that <strong>in</strong>fluence diffusion (fig 2.4);• the rate of oxygen delivery to the capillary (DO 2);• the position of the oxygen-haemoglob<strong>in</strong> dissociationrelationship (P 50);• the rate of cellular oxygen utilisation and uptake (VO 2).The sigmoid oxygen-haemoglob<strong>in</strong> dissociation relationshipis <strong>in</strong>fluenced by various physicochemical factors and its positionis def<strong>in</strong>ed by the PaO 2at which 50% of the Hb is saturated(P 50), normally 3.5 kPa. An <strong>in</strong>crease <strong>in</strong> P 50or rightward shift <strong>in</strong>this relationship reduces the Hb saturation (SaO 2) for anygiven PaO 2, thereby <strong>in</strong>creas<strong>in</strong>g tissue oxygen availability. This iscaused by pyrexia, acidosis, and an <strong>in</strong>crease <strong>in</strong> <strong>in</strong>tracellularphosphate, notably 2,3-diphosphoglycerate (2,3-DPG). Theimportance of correct<strong>in</strong>g hypophosphataemia, often found <strong>in</strong>diabetic ketoacidosis and sepsis, is frequently overlooked. 26Mathematical models of tissue hypoxia show that the fall <strong>in</strong>cellular oxygen result<strong>in</strong>g from an <strong>in</strong>crease <strong>in</strong> <strong>in</strong>tercapillarydistance is more severe if the reduction <strong>in</strong> tissue DO 2is causedby “hypoxic” hypoxia (a fall <strong>in</strong> PaO 2) rather than “stagnant” (afall <strong>in</strong> flow) or “anaemic” hypoxia (fig 2.5). 27Studies <strong>in</strong>patients with hypoxaemic respiratory failure have also shownthatitisPaO 2rather than DO 2—that is, diffusion rather thanconvection—that has the major <strong>in</strong>fluence on outcome. 9Thus, tissue oedema due to <strong>in</strong>creased vascular permeabilityor excessive fluid load<strong>in</strong>g may result <strong>in</strong> impaired oxygendiffusion and cellular hypoxia, particularly <strong>in</strong> cl<strong>in</strong>ical situationsassociated with arterial hypoxaemia. In these situations,avoid<strong>in</strong>g tissue oedema may improve tissue oxygenation.


Oxygen delivery and consumption <strong>in</strong> the critically ill 15SlowdiffusionLong diffusion distance( )Low pressure gradientRed cellRapiddiffusionShort diffusion distance( )High pressure gradientCapillaryPaO 25.3 kPaPaO 27 kPaPaO 213 kPaDiffusion distancePressure gradient5.3 – 1.3 = 4.0 kPaIntracellularPaO 2 2.7 kPaDiffusion distancePressure gradient13 – 2.7 = 10.3 kPaMitochondrialPaO 2 >1.3 kPaIntracellularPO 2 1.3 kPaMitochondrialPaO 2


16 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Intercapillarydistance = 80 µm80 µm80 µm80 µm80 µmINTERSTITIALOEDEMAOxygen consumption (ml/m<strong>in</strong>/kg)6543Hypoxia21AnaemiaLow flow0 0 5 10 15 20Oxygen delivery (ml/m<strong>in</strong>/kg)Intercapillarydistance = 160 µm160 µm160 µm160 µm160 µmOxygen consumption (ml/m<strong>in</strong>/kg)6543Hypoxia21AnaemiaLow flow0 0 5 10 15 20Oxygen delivery (ml/m<strong>in</strong>/kg)Figure 2.5 Influence of <strong>in</strong>tercapillary distance on the effects of hypoxia, anaemia, and low flow on the oxygen delivery-consumptionrelationship. With a normal <strong>in</strong>tercapillary distance illustrated <strong>in</strong> the top panels the DO 2/VO 2relationship is the same for all <strong>in</strong>terventions.However, <strong>in</strong> the lower panels an <strong>in</strong>creased <strong>in</strong>tercapillary distance, as would occur with tissue oedema, reduc<strong>in</strong>g DO 2by progressive falls <strong>in</strong>arterial oxygen tension results <strong>in</strong> a change <strong>in</strong> the DO 2/VO 2relationship with VO 2fall<strong>in</strong>g at much higher levels of global DO 2. This alteredrelationship is not seen when DO 2is reduced by anaemia or low blood flow.same relationship may be found <strong>in</strong> patients with normal lactateconcentrations. 35Serial lactate measurements, particularlyif corrected for pyruvate, may be of greater value.Measurement of <strong>in</strong>dividual organ and tissue oxygenation isan important goal for the future. These measurements are difficult,require specialised techniques, and are not widely available.At present only near <strong>in</strong>frared spectroscopy and gastrictonometry have cl<strong>in</strong>ical applications <strong>in</strong> the detection of organhypoxia. 24 In the future NMR spectroscopy may allow directnon-<strong>in</strong>vasive measurement of tissue energy status and oxygenutilisation. 36CELLULAR OXYGEN UTILISATIONIn general, eukaryotic cells are dependent on aerobic metabolismas mitochondrial respiration offers greater efficiency forextraction of energy from glucose than anaerobic glycolysis.The ma<strong>in</strong>tenance of oxidative metabolism is dependent oncomplex but poorly understood mechanisms for microvascularoxygen distribution and cellular oxygen uptake. Teleologically,the response to reduced blood flow <strong>in</strong> a tissue is likely to haveevolved as an energy conserv<strong>in</strong>g mechanism when substrates,particularly molecular oxygen, are scarce. Pathways that useATP are suppressed and alternative anaerobic pathways forATP synthesis are <strong>in</strong>duced. 37This process <strong>in</strong>volves oxygensens<strong>in</strong>g and transduction mechanisms, gene activation, andprote<strong>in</strong> synthesis.CELLULAR METABOLIC RESPONSE TO HYPOXIAAlthough cellular metabolic responses to hypoxia rema<strong>in</strong>poorly understood, the importance of understand<strong>in</strong>g andmodify<strong>in</strong>g the cellular responses to acute hypoxia <strong>in</strong> the criticallyill patient has recently been appreciated. In isolatedmitochondria the partial pressure of oxygen required togenerate high energy phosphate bonds (ATP) that ma<strong>in</strong>ta<strong>in</strong>aerobic cellular biochemical functions is only about 0.2–0.4 kPa. 17 28 However, <strong>in</strong> <strong>in</strong>tact cell preparations hypoxia<strong>in</strong>duced damage may result from failure of energy dependentmembrane ion channels with subsequent loss of membrane<strong>in</strong>tegrity, changes <strong>in</strong> cellular calcium homeostasis, and oxygendependent changes <strong>in</strong> cellular enzyme activity. 28 The sensitivityof an enzyme to hypoxia is a function of its PO 2<strong>in</strong> mm Hgat which the enzyme rate is half maximum (KmO 2), 28 and thewide range of values for a variety of cellular enzymes is shown<strong>in</strong> table 2.2, illustrat<strong>in</strong>g that certa<strong>in</strong> metabolic functions aremuch more sensitive to hypoxia than others. Cellular toleranceto hypoxia may <strong>in</strong>volve “hibernation” strategies that reducemetabolic rate, <strong>in</strong>creased oxygen extraction from surround<strong>in</strong>gtissues, and enzyme adaptations that allow cont<strong>in</strong>u<strong>in</strong>gmetabolism at low partial pressures of oxygen. 37Anaerobic metabolism is important for survival <strong>in</strong> some tissuesdespite its <strong>in</strong>herent <strong>in</strong>efficiency: skeletal muscle <strong>in</strong>creasesglucose uptake by 600% dur<strong>in</strong>g hypoxia and bladder smoothmuscle can generate up to 60% of total energy requirement byanaerobic glycolysis. 38 In cardiac cells anaerobic glucose utilisationprotects cell membrane <strong>in</strong>tegrity by ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g energydependent K + channels. 39 Dur<strong>in</strong>g hypoxic stress endothelialand vascular smooth muscle cells <strong>in</strong>crease glucose transportthrough the expression of membrane glucose transporters(GLUT-1 and GLUT-4) and the production of glycolyticenzymes, thereby <strong>in</strong>creas<strong>in</strong>g anaerobic glycolysis and ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>genergy production. 38High energy functions like ion


Oxygen delivery and consumption <strong>in</strong> the critically ill 17Table 2.2 Oxygen aff<strong>in</strong>ities of cellular enzymesexpressed as the partial pressure of oxygen <strong>in</strong> mm Hgat which the enzyme rate is half maximum (KmO 2)Enzyme Substrate KmO 2Glucose oxidase Glucose 57Xanth<strong>in</strong>e oxidase Hypoxanth<strong>in</strong>e 50Tryptophan oxygenase Tryptophan 37Nitric oxide synthase L-arg<strong>in</strong><strong>in</strong>e 30Tyros<strong>in</strong>e hydroxylase Tyros<strong>in</strong>e 25NADPH oxidase Oxygen 23Cytochrome aa3 Oxygen 0.05transport and prote<strong>in</strong> production are downregulated tobalance supply and demand.Cellular oxygen utilisation is <strong>in</strong>hibited by metabolic poisons(cyanide) and tox<strong>in</strong>s associated with sepsis such as endotox<strong>in</strong>and other cytok<strong>in</strong>es, thereby reduc<strong>in</strong>g energy production. 29 Itis yet to be established whether there are importantdifferences <strong>in</strong> the response to tissue hypoxia result<strong>in</strong>g fromdamage to mitochondrial and other <strong>in</strong>tracellular functions asoccurs <strong>in</strong> poison<strong>in</strong>g and sepsis, as opposed to situations suchas exercise and altitude when oxygen consumption exceedssupply.OXYGEN SENSING AND GENE ACTIVATIONThe molecular basis for oxygen sens<strong>in</strong>g has not beenestablished and may differ between tissues. Current evidencesuggests that, follow<strong>in</strong>g activation of a “hypoxic sensor”, thesignal is transmitted through the cell by second messengerswhich then activate regulatory prote<strong>in</strong> complexes termed40 41transcription factors. These factors translocate to thenucleus and b<strong>in</strong>d with specific DNA sequences, activat<strong>in</strong>gvarious genes with the subsequent production of effector prote<strong>in</strong>s.It has long been postulated that the “hypoxic sensor”may <strong>in</strong>volve haem-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>s, redox potential ormitochondrial cytochromes. 42 Recent evidence from vascularsmooth muscle suggests that hypoxia <strong>in</strong>duced <strong>in</strong>hibition ofelectron transfer at complex III <strong>in</strong> the electron transport cha<strong>in</strong>may act as the “hypoxic sensor”. 43 This sens<strong>in</strong>g mechanism isassociated with the production of oxygen free radicals (ubiqu<strong>in</strong>onecycle) that may act as second messengers <strong>in</strong> the activationof transcription factors.Several transcription factors play a role <strong>in</strong> the response totissue hypoxia <strong>in</strong>clud<strong>in</strong>g hypoxia <strong>in</strong>ducible factor 1 (HIF-1),early growth response 1 (Erg-1), activator prote<strong>in</strong> 1 (AP-1),nuclear factor kappa-B (NF-κB), and nuclear factor IL-6 (NF-IL-6). HIF-1 <strong>in</strong>fluences vascular homeostasis dur<strong>in</strong>g hypoxiaby activat<strong>in</strong>g the genes for erythropoiet<strong>in</strong>, nitric oxidesynthase, vascular endothelial growth factor, and glycolyticenzymes and glucose transport thereby alter<strong>in</strong>g metabolicfunction. 40Erg-1 prote<strong>in</strong> is also rapidly <strong>in</strong>duced by hypoxialead<strong>in</strong>g to transcription of tissue factor, which triggersprothrombotic events. 41REFERENCES1 Harrison MJG, Kendall BE, Pollock S, et al. Effect of haematocrit oncarotid stenosis and cerebral <strong>in</strong>farction. Lancet 1981;ii:114–5.2 Hebert PC, Wells G, Blajchman MA, et al. A multicentre, randomized,controlled cl<strong>in</strong>ical trial of transfusion requirements <strong>in</strong> critical care. N EnglJ Med 1999;340:409–17.3 Archie JP. Mathematic coupl<strong>in</strong>g of data. A common source of error. AnnSurg 1981;193:296–303.4 Fellows IW, Bennett T, MacDonald IA. The effects of adrenal<strong>in</strong>e uponcardiovascular and metabolic functions <strong>in</strong> man. Cl<strong>in</strong> Sci1985;69:215–22.5 Schumacker PT, Ca<strong>in</strong> SM. The concept of a critical DO 2 . Intensive <strong>Care</strong>Med 1987;13:223.6 Bihari D, Smithies M, Gimson A, et al. The effect of vasodilation withprostacycl<strong>in</strong> on oxygen delivery and uptake <strong>in</strong> critically ill patients. NEngl J Med 1987;317:397–403.Key po<strong>in</strong>ts• Restoration of global oxygen delivery is an important goal <strong>in</strong>early resuscitation but thereafter circulatory manipulation tosusta<strong>in</strong> “supranormal” oxygen delivery does not improvesurvival and may be harmful.• Regional distribution of oxygen delivery is vital: if sk<strong>in</strong> andmuscle receive high blood flows but the splanchnic bed doesnot, the gut may become hypoxic despite high globaloxygen delivery.• Microcirculatory, tissue diffusion, and cellular factors<strong>in</strong>fluence the oxygen status of the cell and global measurementsmay fail to identify local tissue hypoxaemia.• Supranormal levels of oxygen delivery cannot compensatefor diffusion problems between capillary and cell, nor formetabolic failure with<strong>in</strong> the cell.• When assess<strong>in</strong>g DO 2/VO 2relationships, direct measurementsshould be used to avoid errors due to mathematicall<strong>in</strong>kage.• Strategies to reduce metabolic rate to improve tissueoxygenation should be considered.7 Shoemaker WC, Appel PL, Waxman K, et al. Cl<strong>in</strong>ical trial of survivors’cardiorespiratory patterns as therapeutic goals <strong>in</strong> critically illpostoperative patients. Crit <strong>Care</strong> Med 1982;10:398–403.8 Shoemaker WC, Appel PL, Kram HB, et al. Prospective trial ofsupranormal values of survivors as therapeutic goals <strong>in</strong> high-risk surgicalpatients. Chest 1988;94:1176–86.9 Leach RM, Treacher DF. The relationship between oxygen delivery andconsumption. Disease-a-Month 1994;40:301–68.10 Hayes MA, Timm<strong>in</strong>s AC, Yau E, et al. Elevation of systemic oxygendelivery <strong>in</strong> the treatment of critically ill patients. N Engl J Med1994;330:1712–22.11 Gatt<strong>in</strong>oni L, Brazzi L, Pelosi P, et al. A trial of goal-orientatedhaemodynamic therapy <strong>in</strong> critically ill patients. N Engl J Med1995;333:1025–32.12 Connors AF, Speroff T, Dawson NV, et al. The effectivenss of right heartcatheterisation <strong>in</strong> the <strong>in</strong>itial care of critically ill patients. JAMA1996;276:889–97.13 Boyd O, Grounds M, Bennett ED. A randomised cl<strong>in</strong>ical trial of the effectof deliberate peri-operative <strong>in</strong>crease of oxygen delivery on mortality <strong>in</strong>high-risk surgical patients. JAMA 1993;270:2699–708.14 S<strong>in</strong>clair S, James S, S<strong>in</strong>ger M. Intraoperative <strong>in</strong>travascular volumeoptimisation and length of hospital stay after repair of proximal femoralfracture; randomised control trial. BMJ 1997;315:909–12.15 Wilson J, Woods I, Fawcett J, et al. Reduc<strong>in</strong>g the risk of major electivesurgery: randomised control trial of preoperative optimis<strong>in</strong>g of oxygendelivery. BMJ 1999;318:1099–103.16 Lobo SMA, Salgado PF, Castillo VGT, et al. Effects of maximiz<strong>in</strong>goxygen delivery on morbidity and mortality <strong>in</strong> high-risk surgical patients.Crit <strong>Care</strong> Med 2000;28:3396–404.17 Dul<strong>in</strong>g BR. Oxygen, metabolism, and microcirculatory control. In: KaleyG, Altura BM, eds. Microcirculation. Volume 2. Baltimore: University ParkPress, 401–29.18 Curtis SE, Ca<strong>in</strong> SM. Regional and systemic oxygen delivery/uptakerelations and lactate flux <strong>in</strong> hyperdynamic, endotox<strong>in</strong>-treated dogs. AmRev Respir Dis 1992;145:348–54.19 Mulvany MJ, Aalkjaer C, Heagerty AM, et al, eds. Resistance arteries,structure and function. Amsterdam: Elsevier Science, 1991.20 Karimova A, P<strong>in</strong>sky DJ. The endothelial response to oxygen deprivation:biology and cl<strong>in</strong>ical implications. Intensive <strong>Care</strong> Med 2001;27:19–31.21 Petros A, Bennett A, Vallance P. Effect of nitric oxide synthase <strong>in</strong>hibitorson hypotension <strong>in</strong> patients with septic shock. Lancet 1991;338:1557–8.22 Bernard GR, V<strong>in</strong>cent J-L, Laterre P-F, et al. Efficacy and safety ofrecomb<strong>in</strong>ant human activated prote<strong>in</strong> C for severe sepsis. N Engl J Med2001;344:699–709.23 Nelson DP, Samsel RW, Wood LDH, et al. Pathological supplydependence of systemic and <strong>in</strong>test<strong>in</strong>al O 2 uptake dur<strong>in</strong>g endotoxaemia. JAppl Physiol 1988;64:2410–9.24 Gutierrez G, Palizas F, Doglio G, et al. Gastric <strong>in</strong>tramucosal pH as atherapeutic <strong>in</strong>dex of tissue oxygenation <strong>in</strong> critically ill patients. Lancet1992;339:195–9.25 Stephan H, Sonnatag H, Henn<strong>in</strong>g H, et al. Cardiovascular and renalhaemodynamic effects of dopexam<strong>in</strong>e: comparison with dopam<strong>in</strong>e. Br JAnaesth 1990;65:380–7.26 Clebaux T, Detry B, Reynaert M, et al. Re-estimation of the effects of<strong>in</strong>organic phosphates on the equilibrium between oxygen andhemoglob<strong>in</strong>. Intensive <strong>Care</strong> Med 1992;18:222–5.27 Schumacker PT, Samsel RW. Analysis of oxygen delivery and uptakerelationships <strong>in</strong> the Krogh tissue model. J Appl Physiol1989;67:1234–44.28 Rob<strong>in</strong> ED. Of men and mitochondria: cop<strong>in</strong>g with hypoxic dysoxia. AmRev Respir Dis 1980;122:517–31.29 Bradley SG. Cellular and molecular mechanisms of action of bacterialendotox<strong>in</strong>s. Ann Rev Microbiol 1979;33:67–94.


18 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>30 V<strong>in</strong>cent J-L, Roman A, De Backer D, et al. Oxygen uptake/supplydependency. Am Rev Respir Dis 1990;142:2–7.31 Ca<strong>in</strong> SM, Curtis SE. Experimental models of pathologic oxygen supplydependence. Crit <strong>Care</strong> Med 1991;19:603–11.32 Vary TC, Siegel JH, Nakatani T, et al. Effects of sepsis on activity ofpyruvate dehydrogenase complex <strong>in</strong> skeletal muscle and liver. Am JPhysiol 1986;250:E634–9.33 Wilson DF, Erec<strong>in</strong>ska M, Drown C, et al. Effect of oxygen tension oncellular energetics. Am J Physiol 1973;233:C135–40.34 Silverman HJ. Lack of a relationship between <strong>in</strong>duced changes <strong>in</strong>oxygen consumption and changes <strong>in</strong> lactate levels. Chest1991;100:1012–5.35 Mohsenifar Z, Am<strong>in</strong> D, Jasper AC, et al. Dependence of oxygenconsumption on oxygen delivery <strong>in</strong> patients with chronic congestive heartfailure. Chest 1987;92:447–56.36 Leach RM, Sheehan DW, Chacko VP, et al. Energy state, pH, andvasomotor tone dur<strong>in</strong>g hypoxia <strong>in</strong> precontracted pulmonary and femoralarteries. Am J Physiol 2000;278:L294–304.37 Hochachka PW, Buck LT, Doll CJ, et al. Unify<strong>in</strong>g theory of hypoxiatolerance: molecular/metabolic defense and rescue mechanisms forsurviv<strong>in</strong>g oxygen lack. Proc Natl Acad Sci USA 1996;93:9493–8.38 Cartee GD, Dounen AG, Ramlai T, et al. Stimulation of glucose transport<strong>in</strong> skeletal muscle by hypoxia. J Appl Physiol 1991;70:1593–600.39 Paul RJ. Smooth muscle energetics. Ann Rev Physiol 1989;51:331–49.40 Semenza GL. Hypoxia-<strong>in</strong>ducible factor 1: master regulator of O 2homeostasis. Curr Op<strong>in</strong> Genet Dev 1998;8:588–94.41 Yan S-F, Lu J, Zou YS, et al. Hypoxia-associated <strong>in</strong>duction of earlygrowth response-1 gene expression. J Biol Chem 1999;274:15030–40.42 Archer SL, Weir EK, Reeve HL, et al. Molecular identification of O 2sensors and O 2 -sensitive potassium channels <strong>in</strong> the pulmonary circulation.Adv Exp Med Biol 2000;475:219–40.43 Leach RM, Hill HS, Snetkov VA, et al. Divergent roles of glycolysis andthe mitochondrial electron transport cha<strong>in</strong> <strong>in</strong> hypoxic pulmonaryvasoconstriction of the rat: identity of the hypoxic sensor. J Physiol2001;536:211–24.


3 <strong>Critical</strong> care management of community acquiredpneumoniaS V Baudou<strong>in</strong>.............................................................................................................................Community acquired pneumonia (CAP) is acommon illness with an estimated <strong>in</strong>cidenceof 2–12 cases/1000 population peryear. 12 The majority of cases of CAP are successfullymanaged outside hospital, but approximately20% require hospital admission. Out ofthis group about 10% develop severe CAP andneed treatment <strong>in</strong> an <strong>in</strong>tensive care unit (ICU).The mortality of these patients can exceed 50%,and the purpose of this chapter is to review themanagement of severe CAP. Excellent guidel<strong>in</strong>esfor the management of CAP have been producedby several organisations <strong>in</strong>clud<strong>in</strong>g the BritishThoracic Society (BTS), the American ThoracicSociety (ATS), European and Infectious DiseaseWork<strong>in</strong>g Groups. 3–6Revised BTS guidel<strong>in</strong>es haverecently been published 4 and previous ATS recommendationsare be<strong>in</strong>g revised. Any practitionerwho is responsible for patients with CAP shouldconsult one of these documents. This chapter willdiscuss both general approaches to CAP and alsohighlight specific areas of critical care management.ASSESSMENT OF SEVERITYFor the purposes of epidemiological studies, thedef<strong>in</strong>ition of severe CAP as “CAP need<strong>in</strong>g ICUadmission” is adequate. In practical managementterms, however, a more detailed method ofassessment is needed. Severe CAP is almostalways a multiorgan disease and patients withsevere CAP at presentation will either alreadyhave, or will be rapidly develop<strong>in</strong>g, multiple organfailure. It is important that respiratory and other“front l<strong>in</strong>e” physicians appreciate this aspect ofthe disease. Apparent stability on high flowoxygen can rapidly change to respiratory, circulatory,and renal failure. Progressive loss of tissueoxygenation needs to be anticipated, recognisedquickly, and rapid action taken to prevent its progressionto established organ failure.The BTS guidel<strong>in</strong>es def<strong>in</strong>e severe pneumonia(“rule 1”) as the presence of two or more of thefollow<strong>in</strong>g features on hospital admission 4 :• <strong>Respiratory</strong> rate >30/m<strong>in</strong>ute• Diastolic blood pressure 7 mmol/lThe guidel<strong>in</strong>es <strong>in</strong>clude three additional assessmentrecommendations. The presence of any oneof these approximately doubles the rate of death:• Altered mental status, confusion or an AbbreviatedMental Test score of


20 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>a wide range of both medical and surgical admissions <strong>in</strong>clud<strong>in</strong>gpatients with severe CAP. The study found that suboptimalcare had been given to 54% and, importantly, that hospitalmortality <strong>in</strong> this group was significantly higher than <strong>in</strong> thosemanaged well (56% v 35%). Errors <strong>in</strong> the management of theairway, breath<strong>in</strong>g, circulation, monitor<strong>in</strong>g, and oxygentherapy were common.Correct management of severe CAP before admission to theICU is therefore essential. Recognition of the severity of illnessis the first vital step, <strong>in</strong> which application of the BTS severityrules and screen<strong>in</strong>g pulse oximetry are useful tools. Repeatedregular assessment by the same observer <strong>in</strong> the <strong>in</strong>itial stagesof the illness is necessary and rapid review by a critical carepractitioner should be arranged for any patient who meets theBTS or similar severity criteria or who is deteriorat<strong>in</strong>g. Theneed for <strong>in</strong>creas<strong>in</strong>g FIO 2, altered mental state (confusion,aggression), and the onset of either respiratory or metabolicacidosis are all signs of disease progression and the need forfurther <strong>in</strong>tervention.In the UK the recent publication of the Department ofHealth document “Comprehensive <strong>Critical</strong> <strong>Care</strong>” 11 suggestsexpand<strong>in</strong>g high dependency or—<strong>in</strong> the new term<strong>in</strong>ology—level 2 care. This would provide a suitable environment for the<strong>in</strong>itial treatment of patients with severe CAP who do not needimmediate mechanical ventilation. These patients are likely tobenefit from more <strong>in</strong>tensive monitor<strong>in</strong>g (arterial l<strong>in</strong>e, centralvenous l<strong>in</strong>e, ur<strong>in</strong>ary catheter) and treatment (rapid correctionof hypovolaemia, <strong>in</strong>otropic support, cont<strong>in</strong>uous positiveairway pressure (CPAP), non-<strong>in</strong>vasive ventilation (NIV)).Level 2 care also allows the rapid <strong>in</strong>itiation of <strong>in</strong>vasivemechanical ventilation when needed.CO-MORBIDITYThe orig<strong>in</strong>al BTS study on severe CAP po<strong>in</strong>ted to the importanceof pre-exist<strong>in</strong>g co-morbidity 8 : 63% of this group had seriouspre-morbid conditions <strong>in</strong>clud<strong>in</strong>g chronic obstructive pulmonarydisease (COPD, 32%), asthma (13%), and cardiacproblems (15%). Other significant conditions <strong>in</strong>cludeddiabetes, chronic liver disease, chronic renal failure, and alcoholdependency. Immunosuppression was also a risk factor forsevere CAP. The <strong>in</strong>cidence of severe CAP <strong>in</strong>creases with ageand <strong>in</strong>creas<strong>in</strong>g age probably adversely affects outcome; analysisof 11 studies of CAP <strong>in</strong> the elderly 12 showed that more than90% of pneumonia deaths occurred <strong>in</strong> patients over the age of70.MICROBIOLOGYIn the last decade a number of important facts have beenestablished about the microbiology of CAP 12 : (1) a relativelysmall number of pathogens account for the majority of <strong>in</strong>fections;(2) Streptococcus pneumoniae has been consistently shownto be the commonest pathogen <strong>in</strong> Europe and North America;and (3) <strong>in</strong> at least one third of cases no def<strong>in</strong>ite causativepathogen can be isolated. However, the relative importance ofpathogens varies considerably worldwide. For example, <strong>in</strong> areport from S<strong>in</strong>gapore, Burkholderia pseudomallei was the mostcommon cause of severe CAP. 13In addition to S pneumoniae, other important pathogens <strong>in</strong>CAP <strong>in</strong>clude Haemophilus <strong>in</strong>fluenza, Legionella species, Staphylococcusaureus, Gram negative organisms, Mycoplasma, Coxiellaspecies, and respiratory viruses. European and North Americanstudies have found similar <strong>in</strong>cidences of specificpathogens. In a survey of 16 studies of severe CAP the follow<strong>in</strong>gpathogens were isolated: S pneumoniae 12–38%; Legionellaspp 0–30%; Staph aureus 1–18%; and Gram negative entericbacilli 2–34%. 1 There is an important change <strong>in</strong> the frequencyof these pathogens depend<strong>in</strong>g on the severity of the illness (fig3.1). In the UK there is a high relative frequency of Legionellaand Staph aureus <strong>in</strong> severe CAP compared with cases cared forPrecentage isolated4035302520151050CommunityHospitalICUS pneumonia Staph aureus LegionellaFigure 3.1 Percentage isolation of S pneumoniae, Staph aureus,and Legionella species from patients with CAP treated <strong>in</strong> thecommunity, general medical wards, and <strong>in</strong>tensive care units. Spneumoniae rema<strong>in</strong>s the commonest pathogen isolated <strong>in</strong> thecritically ill but the frequency of Staph aureus and Legionella<strong>in</strong>fections significantly <strong>in</strong>creases <strong>in</strong> this group. Data adapted from theBTS guidel<strong>in</strong>es. 4<strong>in</strong> the community or the general wards. The relative frequencyof S pneumoniae is reduced <strong>in</strong> severe CAP, but it rema<strong>in</strong>s themost frequent pathogen isolated.MICROBIOLOGICAL INVESTIGATION ANDDIAGNOSISAt least three strategies have been used <strong>in</strong> the microbiologicaldiagnosis of severe CAP. These can be summarised as (1) thesyndrome approach; (2) the laboratory based approach; and(3) the empirical approach. 1 The strengths or weaknesses ofeach of these strategies will be reviewed <strong>in</strong> the follow<strong>in</strong>g sections.The syndrome approachThis is based on the assumption that different pathogenscause dist<strong>in</strong>ct and non-overlapp<strong>in</strong>g cl<strong>in</strong>ical syndromes. Theterms “typical” and “atypical” pneumonia were adopted todescribe these syndromes. Typical pneumonia was caused bythe pneumococcus and was said to present with pyrexia ofgreater than 39°C, pleuritic chest pa<strong>in</strong>, a lobar distribution ofconsolidation, and an <strong>in</strong>crease <strong>in</strong> immature granulocytes. Featuresof atypical pneumonia <strong>in</strong>cluded a more gradual onsetand a diffuse <strong>in</strong>terstitial or alveolar pattern on the pla<strong>in</strong> chestradiograph.Numerous studies, however, have shown that cl<strong>in</strong>ical overlapbetween the different pathogens is great and that no s<strong>in</strong>gleor comb<strong>in</strong>ation of symptoms and pla<strong>in</strong> chest radiology willreliably differentiate between the different pathogens. 1Insevere CAP the situation is even more difficult; case series ofsevere pneumococcal, staphylococcal, and legionella pneumoniashow no reliable dist<strong>in</strong>guish<strong>in</strong>g features. In a recent seriesof 84 patients requir<strong>in</strong>g ICU admission for severe legionellapneumonia, 39% had only unilateral radiographic changes atpresentation. 14Hyponatraemia is often quoted as a sign oflegionella pneumonia but <strong>in</strong> this series 14 hyponatraemia wasstrongly associated with poor outcome, suggest<strong>in</strong>g that it is amarker of disease severity rather than disease type.The laboratory based approachThere are a number of reasons for attempt<strong>in</strong>g to identify preciselythe pathogen <strong>in</strong> severe CAP: to confirm the diagnosis, toguide antibiotic choice, to def<strong>in</strong>e antibiotic sensitivities, and toprovide epidemiological <strong>in</strong>formation. All current guidel<strong>in</strong>es


<strong>Critical</strong> care management of community acquired pneumonia 21Box 3.1 BTS guidel<strong>in</strong>es for rout<strong>in</strong>e <strong>in</strong>vestigations <strong>in</strong>hospital for all patients with severe CAP• Blood cultures• Sputum or lower respiratory tract sample for Gram sta<strong>in</strong>,rout<strong>in</strong>e culture, and sensitivity tests• Pleural fluid analysis, if present• Pneumococcal antigen test on sputum, blood, or ur<strong>in</strong>e• Investigations for legionella pneumonia <strong>in</strong>clud<strong>in</strong>g (a) ur<strong>in</strong>efor legionella antigen, (b) sputum or lower respiratory tractsamples for legionella culture and direct immunofluorescence,and (c) <strong>in</strong>itial and follow up legionella serology• <strong>Respiratory</strong> samples for direct immunofluorescence to respiratoryviruses, Chlamydia species, and possibly Pneumocystis• Initial and follow up serology for atypical pathogensrecommend <strong>in</strong>tensive microbiological <strong>in</strong>vestigation. The BTSrecommendations for rout<strong>in</strong>e <strong>in</strong>vestigations <strong>in</strong> severe CAP aresummarised <strong>in</strong> box 3.1. 4 While it is difficult to disagree withthis thorough approach to diagnosis, a number of practicalproblems require discussion. Firstly, there is no good evidencethat this strategy alters the outcome of severe CAP and retrospectivestudies disagree about the impact of laboratory basedmicrobiological test<strong>in</strong>g on outcome. 15 16 In at least 30% of casesno pathogen can be isolated and this group has as good aprognosis. Outcome <strong>in</strong> severe CAP is also strongly related tosecondary factors <strong>in</strong>clud<strong>in</strong>g the number of failed organs andco-morbidities. For these reasons the precise identification ofthe respiratory pathogen may have little impact on recovery.Secondly, current diagnostic tests are neither sensitive norspecific <strong>in</strong> severe CAP. 17One difficulty is that isolation of apathogen <strong>in</strong> severe CAP does not necessarily <strong>in</strong>dicatecausation unless the pathogen is never isolated from healthy<strong>in</strong>dividuals—for example, Mycobacterium tuberculosis. <strong>Respiratory</strong>tract specimens conta<strong>in</strong><strong>in</strong>g few squamous epithelial cells,numerous neutrophils, and large numbers of Gram positive,lancet-shaped diplococci are highly specific for pneumococcalpneumonia. However, sensitivity is much lower. Poorlyobta<strong>in</strong>ed or processed specimens and lack of observer experiencecan dramatically alter the yield. Sputum culture suffersfrom similar problems of low sensitivity and specificity, withthe quality of the sample and prior antibiotic treatment hav<strong>in</strong>ga major impact on yield. Blood cultures are positive <strong>in</strong> only4–18% of hospitalised patients with CAP. 17 Pneumococcus isthe most common pathogen isolated but prior antibiotic treatmentsignificantly reduces yield.Pneumococcal polysaccharide antigen can be detected <strong>in</strong>respiratory or other fluids by a variety of methods. It has theadvantage of be<strong>in</strong>g less strongly <strong>in</strong>fluenced by prior antibiotics,but sensitivity and specificity are very variable betweenstudies. Ur<strong>in</strong>ary antigen test<strong>in</strong>g for legionella serogroup 1 ismore than 95% specific for <strong>in</strong>fection, but sensitivity is low andthe test does not detect other Legionella species. There iscurrent <strong>in</strong>terest <strong>in</strong> the detection of specific microbiologicalnucleic acids by amplification techniques such as reversetranscriptase polymerase cha<strong>in</strong> reaction (RT-PCR). Thesetechniques are likely to suffer from similar sensitivity andspecificity problems that affect conventional tests.Most patients with severe CAP require endotracheal<strong>in</strong>tubation and mechanical ventilatory support. In thesecircumstances, fibreoptic bronchoscopy becomes relativelystraightforward and safe. Should all <strong>in</strong>tubated patients withsevere but microbiologically undiagnosed CAP be bronchoscoped?An evidence based approach cannot be taken as randomisedcontrolled trials have not been performed. Theadvantages are that other pathology such as endobronchialobstruction may be discovered and that a targeted sample oflower respiratory tract secretions may be obta<strong>in</strong>ed. However,samples obta<strong>in</strong>ed us<strong>in</strong>g standard techniques are alwayscontam<strong>in</strong>ated by upper airway flora and are probably no betterthan standard sputum samples. Protected specimen brush(PSB) and bronchoalveolar lavage (BAL) are techniqueswhich attempt to overcome some of these obstacles. PSB techniquesuse a telescoped plugged catheter that is passedthrough the bronchoscope. It conta<strong>in</strong>s a brush protected by aplug which is used to obta<strong>in</strong> the sample and then placed <strong>in</strong>culture medium. Quantification of the subsequent culture isusually performed to improve diagnosis. Studies on non<strong>in</strong>tubatedpatients with CAP report potential pathogens <strong>in</strong>54–85% of cases. However, the yield <strong>in</strong> three series of<strong>in</strong>tubated patients with severe CAP already receiv<strong>in</strong>g antibioticswas reduced to 13–48%. 18–20 BAL samples a larger lung volumethan PSB and the yield appears to be comparable to PSB,although the evidence is very limited. In patients with CAPwho fail to respond to <strong>in</strong>itial treatment, BAL identifies pathogens<strong>in</strong> 12–30%. Hence, while the yield <strong>in</strong> severe CAP is21 22relatively low, it is recommended that bronchoscopy isperformed where the diagnosis is not established or wheretreatment is fail<strong>in</strong>g.Empirical approach to microbiological treatmentAll major guidel<strong>in</strong>es take the view that cl<strong>in</strong>ical syndromes arenon-specific and that diagnostic tests are either too slow or<strong>in</strong>sufficiently reliable to help <strong>in</strong> the <strong>in</strong>itial choice of treatment.An empirical approach relies on a good knowledge of therange of likely local pathogens and the fact that a smallnumber of antibiotics (or a s<strong>in</strong>gle agent) will usually be effective.It has the added advantage of prevent<strong>in</strong>g long delays <strong>in</strong>treatment while the results of laboratory tests are awaited. Theperformance of diagnostic tests is encouraged as a guide tomodify antibiotic treatment if a pathogen is identified.ANTIMICROBIAL TREATMENTDetailed reviews of candidate antibiotics for the treatment of23 24severe CAP are available <strong>in</strong> recently published articles. Ifthe specific pathogen has been isolated, then the choice isrelatively straightforward. The optimal choice of antibiotics forthe empirical treatment of severe CAP is less clear. This will bedeterm<strong>in</strong>ed by local surveillance data but <strong>in</strong> Europe and NorthAmerica must <strong>in</strong>clude effective treatment for S pneumoniae,Legionella spp, Haemophilus spp and Staphylococcus spp. Gramnegative bacilli are a rare cause of severe CAP <strong>in</strong> most series,although they may be found <strong>in</strong> patients with pre-exist<strong>in</strong>g lungdisease or on steroid therapy.Antibiotic resistance is becom<strong>in</strong>g an <strong>in</strong>creas<strong>in</strong>g problemwith a number of reports of penicill<strong>in</strong> resistant S pneumoniae.In the UK, however, cl<strong>in</strong>ically relevant S pneumoniae resistanceis rare and the BTS guidel<strong>in</strong>es cont<strong>in</strong>ue to recommend amoxicill<strong>in</strong>alone for non-severe home based CAP treatment.The severely ill patient with CAP requires a broaderantibiotic coverage that must <strong>in</strong>clude the pathogens mostcommonly caus<strong>in</strong>g severe CAP. The BTS guidel<strong>in</strong>es 4recommendthe comb<strong>in</strong>ation of amoxicill<strong>in</strong>/clavulanate with clarithromyc<strong>in</strong>and the optional addition of rifampic<strong>in</strong>. Theamoxicill<strong>in</strong>/clavulanate comb<strong>in</strong>ation will cover both thepneumococcus and beta-lactamase produc<strong>in</strong>g pathogens suchas H <strong>in</strong>fluenzae. Clarithromyc<strong>in</strong> is a macrolide antibiotic that iseffective aga<strong>in</strong>st “atypical” organisms <strong>in</strong>clud<strong>in</strong>g Legionella sppand aga<strong>in</strong>st S pneumoniae. Rifampic<strong>in</strong> is effective aga<strong>in</strong>stLegionella spp and provides antistaphylococcal cover. Otherantibiotic regimens have been suggested for the empiricaltreatment of severe CAP but there is little objective evidence tosupport one approach over another. Alternatives for patients<strong>in</strong>tolerant of the preferred comb<strong>in</strong>ation <strong>in</strong>clude:• Substitution of cefuroxime, cefotaxime, or ceftriaxone foramoxicill<strong>in</strong>/clavulanate; clarithromyc<strong>in</strong> and rifampic<strong>in</strong> rema<strong>in</strong>


22 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>• Use of a s<strong>in</strong>gle fluoroqu<strong>in</strong>olone with Gram positive cover(e.g. levofloxac<strong>in</strong>)INTENSIVE CARE TREATMENTPublished case series of severe CAP emphasise that the ICUtreatment of this group of patients <strong>in</strong>volves the support ofmultiple fail<strong>in</strong>g organ systems. Most patients die of the complicationsof multiorgan failure rather than from respiratoryfailure alone. In the BTS severe CAP study 32% developedacute renal failure and 55% septic shock; 25% developed centralnervous system problems <strong>in</strong>clud<strong>in</strong>g vascular events andconvulsions. 8Patients with severe CAP have sepsis from a respiratorysource and are optimally managed by a team with experienceof the complications of sepsis. These patients often requirehaemofiltration for renal replacement therapy, <strong>in</strong>vasive circulatorymonitor<strong>in</strong>g, and the use of vasopressors and <strong>in</strong>otropes.Survivors of severe CAP tend to have prolonged ICUadmissions and complications are frequent. In the BTS study12 of the 18 patients who still required ventilatory support at14 days ultimately survived. Most patients who needprolonged ventilatory support will require a tracheostomy towean from ventilation.RESPIRATORY MANAGEMENTAll patients with severe CAP require high flow oxygen therapy.In all except those with a background of chronic respiratoryfailure, FIO 2can be rapidly titrated aga<strong>in</strong>st non-<strong>in</strong>vasive SaO 2measurements with regular arterial blood gas analysis used tocheck calibration. Hypercapnia is a sign of ventilatory failureand <strong>in</strong>dicates the need for more <strong>in</strong>tensive support (usually<strong>in</strong>tubation and mechanical ventilation). Increas<strong>in</strong>g metabolicacidosis <strong>in</strong>dicates the development of circulatory shock andthe requirement for fluid resuscitation and <strong>in</strong>otropic support.CPAP can improve oxygenation <strong>in</strong> diffuse lung disease byrecruit<strong>in</strong>g and stabilis<strong>in</strong>g collapsed alveolar units. It is astandard treatment <strong>in</strong> severe pneumocystis pneumonia and afew case reports describe its successful use <strong>in</strong> severe CAP. 25However, a recent randomised controlled trial of CPAP <strong>in</strong>patients at high risk of develop<strong>in</strong>g acute respiratory distresssyndrome (ARDS) was negative. 26 In the study 123 consecutiveadult patients with marked impairment of gas exchange(PaO 2 /FIO 2


<strong>Critical</strong> care management of community acquired pneumonia 233 European <strong>Respiratory</strong> Society Task Force Report. Guidel<strong>in</strong>es formanagement of adult community-acquired lower respiratory tract<strong>in</strong>fections. Eur Respir J 1998;11:986–91.4 British Thoracic Society. BTS guidel<strong>in</strong>es for the management ofcommunity acquired pneumonia <strong>in</strong> adults. Thorax 2001;56(SupplIV):iv1–64.5 Niederman MS, Bass JB, Campbell GD, et al. Guidel<strong>in</strong>es for the <strong>in</strong>itialmanagement of adults with community-acquired pneumonia: diagnosis,assessment of severity, and <strong>in</strong>itial antimicrobial therapy. AmericanThoracic Society. Am Rev Respir Dis 1993;148:1418–26.6 Mandell LA, Marrie TJ, Grossman RF, et al. Canadian guidel<strong>in</strong>es for the<strong>in</strong>itial management of community-acquired pneumonia: anevidence-based update by the Canadian Infectious Diseases Society andthe Canadian Thoracic Society. The Canadian Community-AcquiredPneumonia Work<strong>in</strong>g Group. Cl<strong>in</strong> Infect Dis 2000;31:383–421.7 Woodhead M. Predict<strong>in</strong>g death from pneumonia. Thorax 1996;51:970.8 British Thoracic Society Research Committee and The Public HealthLaboratory Service. The aetiology, management and outcome of severecommunity-acquired pneumonia on the <strong>in</strong>tensive care unit. Respir Med1992;86:7–13.9 Tang CM, Macfarlane JT. Early management of younger adults dy<strong>in</strong>g ofcommunity acquired pneumonia. Respir Med 1993;87:289–94.10 McQuillan P, Pilk<strong>in</strong>gton S, Allan A, et al. Confidential <strong>in</strong>quiry <strong>in</strong>toquality of care before admission to <strong>in</strong>tensive care. BMJ1998;316:1853–8.11 Department of Health. Comprehensive critical care. A review of adultcritical care services. London: Department of Health, 2000.12 Woodhead M. Pneumonia <strong>in</strong> the elderly. J Antimicrob Chemother1994;34(Suppl A):85–92.13 Tan YK, Khoo KL, Ch<strong>in</strong> SP, et al. Aetiology and outcome of severecommunity-acquired pneumonia <strong>in</strong> S<strong>in</strong>gapore. Eur Respir J1998;12:113–5.14 El Ebiary M, Sarmiento X, Torres A, et al. Prognostic factors of severeLegionella pneumonia requir<strong>in</strong>g admission to ICU. Am J Respir Crit <strong>Care</strong>Med 1997;156:1467–72.15 Leroy O, Santre C, Beuscart C, et al. A five-year study of severecommunity-acquired pneumonia with emphasis on prognosis <strong>in</strong> patientsadmitted to an <strong>in</strong>tensive care unit. Intensive <strong>Care</strong> Med 1995;21:24–31.16 Torres A, Serra-Batlles J, Ferrer A, et al. Severe community-acquiredpneumonia. Epidemiology and prognostic factors. Am Rev Respir Dis1991;144:312–8.17 Skerrett SJ. Diagnostic test<strong>in</strong>g for community-acquired pneumonia. Cl<strong>in</strong>Chest Med 1999;20:531–48.18 Mo<strong>in</strong>e P, Vercken JB, Chevret S, et al. Severe community-acquiredpneumonia. Etiology, epidemiology, and prognosis factors. French StudyGroup for Community-Acquired Pneumonia <strong>in</strong> the Intensive <strong>Care</strong> Unit.Chest 1994;105:1487–95.19 Sorensen J, Forsberg P, Hakanson E, et al. A new diagnostic approachto the patient with severe pneumonia. Scand J Infect Dis1989;21:33–41.20 Potgieter PD, Hammond JM. Etiology and diagnosis of pneumoniarequir<strong>in</strong>g ICU admission. Chest 1992;101:199–203.21 Fe<strong>in</strong>silver SH, Fe<strong>in</strong> AM, Niederman MS, et al. Utility of fiberopticbronchoscopy <strong>in</strong> nonresolv<strong>in</strong>g pneumonia. Chest 1990;98:1322–6.22 Ortqvist A, Kal<strong>in</strong> M, Lejdeborn L, et al. Diagnostic fiberopticbronchoscopy and protected brush culture <strong>in</strong> patients withcommunity-acquired pneumonia. Chest 1990;97:576–82.23 Mandell LA. Antibiotic therapy for community-acquired pneumonia. Cl<strong>in</strong>Chest Med 1999;20:589–98.24 Ortqvist A. In-hospital management of adults who havecommunity-acquired pneumonia. Sem<strong>in</strong> Respir Infect 1999;14:135–50.25 Brett A, S<strong>in</strong>clair DG. Use of cont<strong>in</strong>uous positive airway pressure <strong>in</strong> themanagement of community acquired pneumonia. Thorax1993;48:1280–1.26 Delclaux C, L’Her E, Alberti C, et al. Treatment of acute hypoxemicnonhypercapnic respiratory <strong>in</strong>sufficiency with cont<strong>in</strong>uous positive airwaypressure delivered by a face mask: a randomized controlled trial. JAMA2000;284:2352–60.27 Plant PK, Elliott MW. Non-<strong>in</strong>vasive positive pressure ventilation. J R CollPhysicians Lond 1999;33:521–5.28 Confalonieri M, Potena A, Carbone G, et al. Acute respiratory failure <strong>in</strong>patients with severe community-acquired pneumonia. A prospectiverandomized evaluation of non<strong>in</strong>vasive ventilation. Am J Respir Crit <strong>Care</strong>Med 1999;160:1585–91.29 Wood KA, Lewis L, Von Harz B, et al. The use of non<strong>in</strong>vasive positivepressure ventilation <strong>in</strong> the emergency department: results of arandomized cl<strong>in</strong>ical trial. Chest 1998;113:1339–46.30 Hillman KM, Barber JD. Asynchronous <strong>in</strong>dependent lung ventilation(AILV). Crit <strong>Care</strong> Med 1980;8:390–5.31 Carlon GC, Ray C, Kle<strong>in</strong> R, et al. Criteria for selective positiveend-expiratory pressure and <strong>in</strong>dependent synchronized ventilation ofeach lung. Chest 1978;74:501–7.32 Dreyfuss D, Djeda<strong>in</strong>i K, Lanore JJ, et al. A comparative study of theeffects of almitr<strong>in</strong>e bismesylate and lateral position dur<strong>in</strong>g unilateralbacterial pneumonia with severe hypoxemia. Am Rev Respir Dis1992;146:295–9.33 The Acute <strong>Respiratory</strong> Distress Syndrome Network. Ventilation withlower tidal volumes as compared with traditional tidal volumes for acutelung <strong>in</strong>jury and the acute respiratory distress syndrome. N Engl J Med2000;342:1301–8.34 Fagon JY, Chastre J, Wolff M, et al. Invasive and non<strong>in</strong>vasive strategiesfor management of suspected ventilator-associated pneumonia. Arandomized trial. Ann Intern Med 2000;132:621–30.35 F<strong>in</strong>e MJ, Smith MA, Carson CA, et al. Prognosis and outcomes ofpatients with community-acquired pneumonia. A meta-analysis. JAMA1996;275:134–41.36 Knaus WA, Draper EA, Wagner DP, et al. APACHE II: a severity ofdisease classification system. Crit <strong>Care</strong> Med 1985;13:818–29.


4 Nosocomial pneumoniaS Ewig, A Torres.............................................................................................................................Nosocomial pneumonia is the second mostfrequent hospital acquired <strong>in</strong>fection andthe most frequently acquired <strong>in</strong>fection <strong>in</strong>the <strong>in</strong>tensive care unit (ICU). The <strong>in</strong>cidence is agedependent, with about 5/1000 cases <strong>in</strong> hospitalisedpatients aged under 35 and up to 15/1000 <strong>in</strong>those over 65 years of age. 1–3 Death fromnosocomial pneumonia <strong>in</strong> ventilated patientsreaches 30–50%, with an estimated attributablemortality of 10–50%. 4–9Increas<strong>in</strong>g microbial resistanceworldwide imposes an additional challengefor prevention and antimicrobial treatmentstrategies. 10In the last two decades efforts have been madeto improve outcomes by establish<strong>in</strong>g valid diagnosticand therapeutic strategies. Nevertheless,controversy persists <strong>in</strong> many issues regard<strong>in</strong>g themanagement of nosocomial pneumonia. Thischapter focuses on the ma<strong>in</strong> controversies <strong>in</strong>diagnosis and treatment.DEFINITIONSNosocomial pneumonia usually affects mechanicallyventilated patients, hence the term “ventilatorassociated pneumonia (VAP)” is used synonymously.However, nosocomial pneumonia mayoccur <strong>in</strong> non-ventilated patients, creat<strong>in</strong>g adist<strong>in</strong>ct entity (table 4.1). Notably, all concepts ofnosocomial pneumonia refer to the nonimmunosuppressedhost, with absence of “immunosuppression”def<strong>in</strong>ed as absence of risk for<strong>in</strong>fection with opportunistic pathogens.Divid<strong>in</strong>g patients with VAP <strong>in</strong>to groups withearly and late onset has been shown to be ofparamount importance. 11 Early onset pneumoniacommonly results from aspiration of endogenouscommunity acquired pathogens such as Staphylococcusaureus, Streptococcus pneumoniae, and Haemophilus<strong>in</strong>fluenzae, with endotracheal <strong>in</strong>tubationand impaired consciousness be<strong>in</strong>g the ma<strong>in</strong> riskfactors. 12–15 Conversely, late onset pneumoniafollows aspiration of oropharyngeal or gastricsecretions conta<strong>in</strong><strong>in</strong>g potentially drug resistantnosocomial pathogens. Only late onset VAP isassociated with an attributable excess mortality. 9The def<strong>in</strong>itions of early and late onset VAP havenot been standardised. Firstly, the start<strong>in</strong>g po<strong>in</strong>tfor early onset pneumonia has varied considerably,<strong>in</strong>clud<strong>in</strong>g time of hospital admission, ofadmission to the ICU, or of endotracheal <strong>in</strong>tubation.If the time of admission to the ICU is chosenas the start<strong>in</strong>g po<strong>in</strong>t, patients may already havebeen colonised <strong>in</strong> hospital and consequentlydifferences between early and late onset pneumoniawill no longer be evident. In accordance14 16with the American Thoracic Society (ATS) guidel<strong>in</strong>es,we advocate us<strong>in</strong>g the time of hospitaladmission. Secondly, the cut off time separat<strong>in</strong>gearly and late onset VAP has not been standardised.The ATS suggested us<strong>in</strong>g the fifth day afterhospital admission. 11 We have shown that colonisationof patients after head <strong>in</strong>jury markedlychanged between the third and fourth day <strong>in</strong>favour of nosocomial pathogens. 13Whereas theoropharynx, nose, tracheobronchial tree were <strong>in</strong>itiallycolonized with endogenous communityacquired pathogens, this pattern was subsequentlychanged by an <strong>in</strong>creas<strong>in</strong>g number oftypical nosocomial pathogens. Trouillet et al haveshown that isolation of drug resistant microorganismscan be predicted by the duration of <strong>in</strong>tubationand antimicrobial treatment 17 ; the cut offbetween early and late onset VAP used was 7 days.Traditionally, nosocomial pneumonia is def<strong>in</strong>edas occurr<strong>in</strong>g <strong>in</strong> patients admitted to hospital (or<strong>in</strong>tubated) for at least 48 hours. 18However, thisdef<strong>in</strong>ition is no longer adequate at least for VAPbecause a significant number of cases occurwith<strong>in</strong> 48 hours of hospital admission as a consequenceof <strong>in</strong>tubation, particularly emergency<strong>in</strong>tubation. In these patients cardiopulmonaryresuscitation and cont<strong>in</strong>uous sedation were <strong>in</strong>dependentrisk factors for the development of VAPwhile antimicrobial treatment was protective. 19Key features of the current def<strong>in</strong>itions of nosocomialpneumonia are summarized <strong>in</strong> box 4.1.ANTIMICROBIAL TREATMENTSeveral <strong>in</strong>vestigations have addressed the efficacyof antimicrobial treatment as well as its impact onmicrobial resistance. Such studies have resolvedmany of the controversies surround<strong>in</strong>g the use ofantimicrobial agents <strong>in</strong> the hospital sett<strong>in</strong>g (fig4.1). The immediate adm<strong>in</strong>istration of treatmentTable 4.1 Differences <strong>in</strong> nosocomial pneumonia affect<strong>in</strong>g non-ventilated and ventilated patients (ventilator associatedpneumonia, VAP)Non-ventilated patientsVentilated patientsIncidence Relatively low HighAetiology GNEB, Legionella spp Core pathogens; PDRMMortality Probably relatively low 30–50%Diagnosis Cl<strong>in</strong>ical; TTA; virtually no data on bronchoscopy Cl<strong>in</strong>ical; TBAS; bronchoscopyAntibiotics Monotherapy Early onset: monotherapy. Late onset: comb<strong>in</strong>ation therapyPrevention General measures of <strong>in</strong>fection control Additionally, measures to reduce risk factors associated with <strong>in</strong>tubationGNEB = Gram negative enteric bacteria; PDRM = potentially drug resistant microorganisms; TTA = transthoracic aspiration; TBAS = tracheobronchialsecretions.


Nosocomial pneumonia 25Adequate antimicrobialtreatmentFigure 4.1treatment.IncreasedMortalityDecreasedRapid reduction of bacterial loadLimitation of <strong>in</strong>flammatory responseOngo<strong>in</strong>g bacterial proliferationand <strong>in</strong>flammationSelection of drug resistantmicroorganismsInadequate antimicrobialtreatmentImportance of adequate and appropriate antimicrobialis crucial and <strong>in</strong>appropriate treatment is associated with an<strong>in</strong>creased risk of death from pneumonia. 20–22 Moreover, even ifthe <strong>in</strong>itially <strong>in</strong>appropriate antimicrobial treatment is correctedaccord<strong>in</strong>g to diagnostic test results, there rema<strong>in</strong>s an excessmortality compared with patients treated appropriately fromthe beg<strong>in</strong>n<strong>in</strong>g. 23Conversely, antimicrobial treatment is not without risk,particularly prolonged broad spectrum antimicrobial treatment.Rello and coworkers showed that antimicrobialpretreatment was the only adverse prognostic factor <strong>in</strong> a multivariatemodel. However, if pneumonia due to high riskorganisms (P aerug<strong>in</strong>osa, A calcoaceticus, S marcescens, P mirabilis,and fungi) was <strong>in</strong>cluded <strong>in</strong> the model, the presence of thesehigh risk organisms was the only <strong>in</strong>dependent predictor andantimicrobial pretreatment dropped out. 20 Thus, antimicrobialpretreatment imposes considerable microbial selection pressure,and is associated with excess mortality due to pneumoniacaused by drug resistant microorganisms.It has become <strong>in</strong>creas<strong>in</strong>gly clear that each antimicrobialtreatment policy imposes specific selection pressures, andtherefore microbial and resistance patterns <strong>in</strong> each sett<strong>in</strong>g canto some extent be regarded as the footpr<strong>in</strong>ts of pastantimicrobial treatment policies. With this <strong>in</strong> m<strong>in</strong>d, it is clearthat recommendations for <strong>in</strong>itial empirical antimicrobialtreatment must be flexible so that they may be modified <strong>in</strong>accordance with local circumstances. 24–26 Accord<strong>in</strong>gly, chang<strong>in</strong>gmicrobial patterns and <strong>in</strong>creas<strong>in</strong>g rates of microbial resistancemust be recognized at the local level so that correspond<strong>in</strong>gchanges <strong>in</strong> general antimicrobial treatment policies maybe <strong>in</strong>stituted. 27DIAGNOSTIC STRATEGIESCl<strong>in</strong>ical observations, laboratory results, and chest radiographsare of limited value <strong>in</strong> diagnos<strong>in</strong>g VAP, and so greateffort has been made to establish <strong>in</strong>dependent microbiologicalcriteria. In our view these efforts have not yet succeeded.Despite its limitations, cl<strong>in</strong>ical assessment is the start<strong>in</strong>g po<strong>in</strong>tfor diagnos<strong>in</strong>g VAP and alternative strategies must be<strong>in</strong>terpreted with regard to their ability to decrease the rate offalse positive cl<strong>in</strong>ical judgements (about 10–25%). 28On theother hand, the 20–40% false negative cl<strong>in</strong>ical judgementsrema<strong>in</strong> undetected. 29 Moreover, although it is generally recognizedthat qualitative culture of tracheobronchial secretions ishighly sensitive but poorly specific, preclud<strong>in</strong>g its use forestablish<strong>in</strong>g the diagnosis of VAP <strong>in</strong> the <strong>in</strong>dividual patient,only rarely has it been used for exclusion of VAP and as a toolfor local surveillance. 28 Qualitative tracheobronchial aspirationhas a high negative predictive value, and a negative cultureresult <strong>in</strong> the absence of antimicrobial treatment virtuallyexcludes VAP. Surveillance based on potential pathogenspresent <strong>in</strong> patients with suspected VAP is an <strong>in</strong>creas<strong>in</strong>glyBox 4.1 Key features of the def<strong>in</strong>ition of nosocomialpneumoniaNon-immunosuppressed host (no risk of opportunistic<strong>in</strong>fections) Pneumonia acquired after hospital admission atany time (48 hour threshold no longer adequate)Pneumonia may occur <strong>in</strong>:• Non-ventilated patients• Ventilated patients (i.e. ventilator associated pneumonia,VAP)Pneumonia may present as:• Early onset pneumonia (5 days after hospital admission or<strong>in</strong>tubation)Late onset VAP has a particular risk for potentially drugresistant microorganisms <strong>in</strong> the case of:• More than 7 days of mechanical ventilation• Broad spectrum antimicrobial pretreatmentattractive tool to direct local empirical antimicrobial treatmentpolicies. Can quantitative culture overcome the limitations ofqualitative tracheobronchial aspirates and allow for an<strong>in</strong>dividual diagnostic approach to VAP?The technique of quantitative culture of bronchoscopicallyretrieved protected specimen brush (PSB) and bronchoalveolarlavage (BAL) specimens has been evaluated by a varietyof approaches. Early animal studies established a relationshipbetween histological pneumonia and bacterial loads, butmore recent studies have highlighted limitations of quantitativecultures. In ventilated m<strong>in</strong>i-pigs the severity of bronchialand pulmonary <strong>in</strong>flammatory lesions and bacterial load wereclearly associated. However, there was a large overlap, such thatthreshold bacterial loads could not differentiate between samplesfrom unaffected pigs, those with bronchitis, and thosewith pneumonia. 30 Similarly, <strong>in</strong> a subsequent study evaluat<strong>in</strong>gdiagnostic tools, none had a satisfactory diagnostic yield. 31Studies <strong>in</strong> healthy non-<strong>in</strong>tubated patients have shown ahigh specificity for PSB and BAL. In mechanically ventilatedpatients without suspected VAP the results were lessimpressive, yield<strong>in</strong>g false positive results <strong>in</strong> 20–30%, althoughno strictly <strong>in</strong>dependent reference was used. 32–35In patientswith suspected VAP a variety of diagnostic tools have beenevaluated with conflict<strong>in</strong>g results. 36–39 These studies providedseveral general <strong>in</strong>sights, although references and thresholdsfor the calculation of diagnostic <strong>in</strong>dices varied considerably.Firstly, PSB and BAL had generally comparable diagnosticyields; secondly, tracheobronchial aspirates had comparableyields to PSB and BAL, with a tendency towards a lower specificity;and thirdly, all tools exhibited a rate of false negativeand false positive results rang<strong>in</strong>g from 10% to 30%. A studyfocus<strong>in</strong>g on the variability of PSB showed that the qualitativerepeatability was 100%, while <strong>in</strong> 59% of the patients thequantitative results varied more than tenfold. 40 Based on thesestudies, several <strong>in</strong>vestigations were performed us<strong>in</strong>g postmortemhistological results or lung culture as an <strong>in</strong>dependentreference or gold standard. 41–47Despite several importantmethodological limitations, these studies revealed importantclues to the relationships between histology, microbiology, andthe diagnosis of VAP: (1) limited correlation betweenhistological f<strong>in</strong>d<strong>in</strong>gs and the bacterial load of lung cultures;(2) the recognition that no s<strong>in</strong>gle technique would be irrefutable;(3) a surpris<strong>in</strong>gly high rate of false negative and falsepositive results of 10–50% regardless of the technique used;and (4) a comparable yield from non-<strong>in</strong>vasive and <strong>in</strong>vasivediagnostic tools. Reasons for false negative f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong>cludedsampl<strong>in</strong>g errors, antimicrobial pretreatment, and the presence


26 <strong>Respiratory</strong> <strong>Management</strong> of <strong>Critical</strong> <strong>Care</strong>of stage specific bacterial loads dur<strong>in</strong>g the evolution of pneumonia(develop<strong>in</strong>g as well as resolv<strong>in</strong>g pneumonia). Conversely,false positive results were attributable to contam<strong>in</strong>ationof the samples and bronchiolitis or bronchitis,particularly <strong>in</strong> patients with structural lung disease.Studies evaluat<strong>in</strong>g the <strong>in</strong>fluence of diagnostic techniqueson outcome have a number of limitations: (1) the usefulnessof diagnostic techniques may vary with<strong>in</strong> different populations;(2) this approach ignores the long term effects onmicrobial resistance; (3) the presence of excess mortality hasonly been shown for late onset VAP and was low (0–10%) <strong>in</strong>some studies 4–9 ; and (4) outcome measures are most consistentlyevaluated when antimicrobial treatment is stopped <strong>in</strong>patients without positive culture results which, <strong>in</strong> our view, isunethical. 48Four randomised studies have been publishedevaluat<strong>in</strong>g non-<strong>in</strong>vasive and <strong>in</strong>vasive diagnostic tools, threefrom Spa<strong>in</strong> and one from France. 49–52The Spanish studiesfound no difference <strong>in</strong> outcome measures such as mortality,cost, duration of hospitalisation, ICU stay, and<strong>in</strong>tubation. 49 51 52 The multicentre French study found abronchoscopic strategy <strong>in</strong>clud<strong>in</strong>g quantitative cultures of PSBand/or BAL specimens to be superior to a cl<strong>in</strong>ical strategyus<strong>in</strong>g qualitative tracheobronchial aspirates <strong>in</strong> terms of 14 daymortality, morbidity, and use of antimicrobial treatment. 50Each study had limitations, however, and the results of theFrench study raise the follow<strong>in</strong>g concerns: firstly, the cl<strong>in</strong>icalstrategy did not necessarily reflect rout<strong>in</strong>e practice; secondly, itis not clear from the data how the <strong>in</strong>vasive strategy accountedfor the better outcome; and, thirdly, the cl<strong>in</strong>ical group had asignificantly higher rate of <strong>in</strong>adequate antimicrobial treatment.In a response to our correspond<strong>in</strong>g critique, 53 the authors<strong>in</strong>dicated that the latter was accounted for by the greaternumbers of pathogens detected <strong>in</strong> tracheobronchial secretionsfrom the cl<strong>in</strong>ical group. 54 Although this is plausible, it is contradictoryto assume that the higher detection rate of resistantmicroorganisms <strong>in</strong> tracheobronchial aspirates was associatedwith a worse outcome. Thus, it renders even less clear the issueof how the <strong>in</strong>vasive strategy could translate <strong>in</strong>to lowermortality. Moreover, the study does not allow one to draw anyconclusion regard<strong>in</strong>g the value of <strong>in</strong>vasive bronchoscopic toolsas compared with quantitative tracheobronchial aspirates. Inour randomized study evaluat<strong>in</strong>g the impact of diagnostictechniques on outcome, we could not f<strong>in</strong>d any difference <strong>in</strong>outcome when quantitative tracheobronchial aspirates werecompared with a bronchoscopic strategy.In view of these data, we draw the follow<strong>in</strong>g conclusions:• Quantitative culture cannot confirm a diagnosis of VAP <strong>in</strong>the <strong>in</strong>dividual case.• Non-<strong>in</strong>vasive and <strong>in</strong>vasive bronchoscopic tools have comparablediagnostic yields and share similar methodologicallimitations.• The <strong>in</strong>troduction of microbiological criteria to correct forfalse positive cl<strong>in</strong>ical judgements does not result <strong>in</strong> moreconfident diagnoses of VAP ; the microbiological correctionof false positive judgements is countered by the misclassificationof correctly positive cl<strong>in</strong>ical judgements. 29STATEMENTS FROM A CONSENSUS CONFERENCEA consensus conference, sponsored by four societies, on pneumoniaacquired <strong>in</strong> the ICU was held <strong>in</strong> May 2002, and a summarydocument was published. 55 Although we do not totallyagree with the recommendations, those regard<strong>in</strong>g diagnosisare summarized here.(1) Microbiological samples must be collected before <strong>in</strong>itiationof antimicrobial agents.(2) Reliance on qualitative cultures of endotracheal aspiratesleads to both over-diagnosis and under-diagnosis ofpneumonia.Identification of local epidemiologyDef<strong>in</strong>ition of <strong>in</strong>itial antimicrobial treatment policyAdaptation accord<strong>in</strong>g to results of qTBSCureTreatment failureIndividual diagnostic work up(preferably by bronchoscopy)Figure 4.2 Suggested approach to the management of a patientwith suspected VAP. qTBS = quantitative tracheobronchial secretions.(3) The available evidence favours the use of <strong>in</strong>vasive quantitativeculture techniques over tracheal aspirates whenestablish<strong>in</strong>g an <strong>in</strong>dication for antimicrobial therapy.(4) The available data suggest that the accuracy of nonbronchoscopictechniques for obta<strong>in</strong><strong>in</strong>g quantitativecultures of lower respiratory tract samples is comparableto that of bronchoscopic techniques.(5) The cost effectiveness of <strong>in</strong>vasive as compared with that ofnon-<strong>in</strong>vasive diagnostic strategies has not been established.NEW DEVELOPMENTS IN ANTIMICROBIALTREATMENTThe general limitations of diagnostic criteria for the diagnosisof VAP <strong>in</strong> the <strong>in</strong>dividual patient have fundamental consequencesfor any antimicrobial treatment strategy. We suggesta change <strong>in</strong> perspective away from the <strong>in</strong>dividual and towardsan epidemiological approach, as elaborated <strong>in</strong> the ATSguidel<strong>in</strong>es. 11Important components of such an approach<strong>in</strong>clude the follow<strong>in</strong>g.(1) Initial antimicrobial treatment must always be empirical.(2) Empirical antimicrobial treatment can be guided by threecriteria: severity of pneumonia, time of onset, and specific riskfactors. All pneumonias acquired <strong>in</strong> the ICU are severe bydef<strong>in</strong>ition <strong>in</strong> the guidel<strong>in</strong>es.(3) The selection of antimicrobial agents must be adapted toregional or even local microbial and resistance patterns.(4) The diagnostic work up may offer additional clues thatmust be <strong>in</strong>terpreted <strong>in</strong> the context of the patient’s condition.However, it is generally conf<strong>in</strong>ed to suggest<strong>in</strong>g potentialpathogens and their resistance, which may be particularly relevantwhen there is no response to empirical antibiotics. It istherefore our practice to use quantitative tracheobronchialaspirates regularly, and bronchoscopy with PSB and BAL <strong>in</strong>patients who are not respond<strong>in</strong>g to treatment (fig 4.2).When can antimicrobial treatment be withheld or stopped?Firstly, patients exhibit<strong>in</strong>g signs of severe sepsis or septicshock must receive empirical treatment. Secondly, patientswith cl<strong>in</strong>ically suspected VAP, yield<strong>in</strong>g borderl<strong>in</strong>e colonycounts (>10 2but


Nosocomial pneumonia 27Table 4.2General framework for empirical <strong>in</strong>itial antimicrobial treatment of VAPClass of antimicrobial agentsExamplesVentilated patients:Early onset, no risk factors Cephalospor<strong>in</strong> II • CefuroximeorCephalospor<strong>in</strong> IIIorAm<strong>in</strong>openicill<strong>in</strong>/β-lacatamase <strong>in</strong>hibitorThird or fourth generation qu<strong>in</strong>oloneorCl<strong>in</strong>damyc<strong>in</strong>/aztreonam• Cefotaxime• Ceftriaxone• Amoxicill<strong>in</strong>/clavulanic acid• Levofloxac<strong>in</strong>• Cl<strong>in</strong>damyc<strong>in</strong>• AztreonamLate onset, no risk factors Qu<strong>in</strong>olone • Ciprofloxac<strong>in</strong>orAm<strong>in</strong>oglycoside• Gentamic<strong>in</strong>• Tobramyc<strong>in</strong>• Amikac<strong>in</strong>plusAntipseudomonal β-lactam/β-lactamase <strong>in</strong>hibitor • Piperacill<strong>in</strong>/tazobactamorCeftazidime• CeftazidimeEarly or late onset, risk factorsNon-ventilated patients:Early onset, no risk factorsLate onset, no risk factorsEarly or late onset, risk factorsorCarbapenemsplus/m<strong>in</strong>usVancomyc<strong>in</strong>Risk factors for P aerug<strong>in</strong>osa: see late onsetRisk factors for MRSA: + vancomyc<strong>in</strong>Risk factor for legionellosis: macrolideSee ventilated patientSee ventilated patient; possibly monotherapy <strong>in</strong> theabsence of severe pneumoniaSee ventilated patient, early or late onset, riskfactors• Imipenem/cilastat<strong>in</strong>• Meropenem• Vancomyc<strong>in</strong>• Vancomyc<strong>in</strong>• Erythromyc<strong>in</strong>or• Azithromyc<strong>in</strong>or• Clarithromyc<strong>in</strong>or• Levofloxac<strong>in</strong>or• Moxifloxac<strong>in</strong><strong>in</strong>filtrate on the chest radiograph plus one to three of the follow<strong>in</strong>g:fever or hypothermia, leucocytosis or leucopenia, andpurulent tracheobronchial secretions) are outdated. Inparticular, it is <strong>in</strong>appropriate to ignore changes <strong>in</strong> oxygenation,and the criteria for severe sepsis and/or septic shock.Pug<strong>in</strong> et al 56 have suggested a scor<strong>in</strong>g system for VAP, <strong>in</strong>clud<strong>in</strong>gthe follow<strong>in</strong>g six weighted cl<strong>in</strong>ical and microbiologicalvariables: temperature, white blood cell count, mean volumeand nature of tracheobronchial aspirate, gas exchange ratio,and chest radiograph <strong>in</strong>filtrates. This score achieved a sensitivityof 72% and a specificity of 85% <strong>in</strong> a necroscopic study. 45It is tedious to calculate and <strong>in</strong>cludes microbiological criteria,but it <strong>in</strong>dicates that criteria may be developed thatsignificantly improve the predictive value of cl<strong>in</strong>ical judgement.A second way <strong>in</strong> which our ability to diagnosepneumonia could be improved is by develop<strong>in</strong>g valid severitycriteria. Somewhat surpris<strong>in</strong>g is that, <strong>in</strong> contrast to communityacquired pneumonia, 57severity assessment of VAP hasnot received much attention. However, it is clear that validseverity criteria may be of great help <strong>in</strong> determ<strong>in</strong><strong>in</strong>g whenantimicrobial treatment may safely be withheld or stopped.F<strong>in</strong>ally, valid markers of the <strong>in</strong>flammatory response associatedwith VAP could work as surrogate markers for VAP andthereby be of help <strong>in</strong> guid<strong>in</strong>g antimcrobial treatmentdecisions.In the meantime, our approach is to judge the condition ofthe patient <strong>in</strong> view of all available cl<strong>in</strong>ical, laboratory, andradiographical <strong>in</strong>formation <strong>in</strong> order to improve our ability topredict the presence or absence of VAP. Culture results are then<strong>in</strong>terpreted with<strong>in</strong> this context and decisions are not madeexclusively on the basis of thresholds.A recent multicenter French study has shown similar cl<strong>in</strong>icalefficacy treat<strong>in</strong>g VAP with 7 days compared to 14. The confirmationof these f<strong>in</strong>d<strong>in</strong>gs would be of extreme importance toreduce the amount of antibiotics given <strong>in</strong> ICUs. 58Another approach to reduc<strong>in</strong>g the microbial selection pressureimposed by empirical antimicrobial treatment is toreduce exposure by m<strong>in</strong>imis<strong>in</strong>g the duration of treatment.The challenge would be to identify low risk groups withoutdrug resistant microorganisms. In an elegant study by S<strong>in</strong>ghet al 59 patients with suspected nosocomial pneumonia (58%VAP) with a Pug<strong>in</strong> score of


28 <strong>Respiratory</strong> <strong>Management</strong> of <strong>Critical</strong> <strong>Care</strong>8070S aureus P aerug<strong>in</strong>osa Ac<strong>in</strong>etobacter spp S maltophiliaFigure 4.3 Proportion ofmicroorganisms account<strong>in</strong>g forantimicrobial treatment failures of23 63–65VAP <strong>in</strong> four studies.Percentage of bacterial isolates6050403020100Alvarez-Lerma(1996)Rello(1997)Luna(1997)Kollef(1998)In patients with suspected VAP due to Gram negative pathogens,a controlled rotation of one antimicrobial regimen(ceftazidime) to another (ciprofloxac<strong>in</strong>) was associated with asignificant reduction <strong>in</strong> the <strong>in</strong>cidence of VAP (12% v 7%), the<strong>in</strong>cidence of resistant Gram negative pathogens (4% v 1%), andthe <strong>in</strong>cidence of Gram negative bacteraemia (2% v 0.3%). 25Similarly, controlled rotation of antibiotics <strong>in</strong>clud<strong>in</strong>g restricteduse of ceftazidime and ciprofloxac<strong>in</strong> over 2 years wasassociated with a significant reduction <strong>in</strong> VAP cases from 231 to161 (70%), of potentially drug resistant microorganisms from140 to 79 (56%), but with an <strong>in</strong>crease from 40% to 60% ofmethicill<strong>in</strong> resistant Staphylococcus aureus (MRSA) isolates. 26Itshould be stressed that these studies do not practise rotation <strong>in</strong>its strict sense, but simply strategies of controlled antimicrobialtreatment. The role of antimicrobial rotation cannot thereforebe determ<strong>in</strong>ed yet, either as a fixed (or bl<strong>in</strong>ded) rotation or asa flexible (or controlled) rotation based on local microbial andresistance patterns. 60RECOMMENDATIONS FOR EMPIRICALANTIMICROBIAL TREATMENTBased on the ATS guidel<strong>in</strong>es, 11the follow<strong>in</strong>g recommendationscan be made (table 4.2):(1) Patients with early onset VAP and no risk factors: coreorganisms such as community endogenous pathogens (Staphylococcusaureus, Streptococcus pneumoniae, and Haemophilus <strong>in</strong>fluenzae)and non-resistant Gram negative enterobacteriaceae(GNEB, <strong>in</strong>clud<strong>in</strong>g Escherichia coli, Klebsiella pneumoniae, Enterobacterspp, Serratia spp, Proteus spp) should be appropriatelycovered. This can be achieved by monotherapy with a secondor third generation cephalospor<strong>in</strong> (cefotaxime or ceftriaxone),or by an am<strong>in</strong>openicill<strong>in</strong> plus a β-lactamase <strong>in</strong>hibitor.Qu<strong>in</strong>olones or a comb<strong>in</strong>ation of cl<strong>in</strong>damyc<strong>in</strong> and aztreonamare alternatives.(2) Patients with late onset VAP and no risk factors: potentiallydrug resistant microorganisms must also be taken <strong>in</strong>toaccount. This is particularly true when mechanical ventilationis required for more than 7 days and aga<strong>in</strong>st a background ofbroad spectrum antimicrobial treatment. 17 These <strong>in</strong>clude multiresistantMRSA, GNEB, and Pseudomonas aerug<strong>in</strong>osa, Ac<strong>in</strong>etobacterspp, as well as Stenotrophomonas maltophilia. Although notproven by randomized studies, it seems prudent to adm<strong>in</strong>istercomb<strong>in</strong>ation treatment, <strong>in</strong>clud<strong>in</strong>g an antipseudomonal penicill<strong>in</strong>(plus a β-lactamase <strong>in</strong>hibitor) or a cephalospor<strong>in</strong> or acarbapenem, and a qu<strong>in</strong>olone (ciprofloxac<strong>in</strong>) or an am<strong>in</strong>oglycoside.Vancomyc<strong>in</strong> may be added where MRSA is a concern.(3) Patients with early or late onset VAP and risk factors:treatment is identical to that for late onset VAP without riskfactors, except when Legionella spp are suspected <strong>in</strong> which casethese pathogens must also be covered.The guidel<strong>in</strong>es do not make specific recommendations fornon-ventilated patients with nosocomial pnuemonia. Instead,patients not meet<strong>in</strong>g severity criteria are treated as early onsetVAP with modifications <strong>in</strong> the presence of additional risk factors.In our view it would be useful to compare this severitybased approach with an algorithm that separates pneumonia<strong>in</strong> the non-<strong>in</strong>tubated and <strong>in</strong>tubated patient, differentiatesearly and late onset, and considers the presence of risk factors.This is the direction of the recently published German guidel<strong>in</strong>esfor the treatment and prevention of nosocomialpneumonia. 61This general framework for empirical <strong>in</strong>itial antimicrobialtreatment must be modified accord<strong>in</strong>g to local requirements.Regular updates of data on potential pathogens of VAP<strong>in</strong>dicat<strong>in</strong>g trends <strong>in</strong> microbial and resistance patterns aremandatory. 62 Although data on antimicrobial treatmentfailures are scarce, we recommend <strong>in</strong>vestigat<strong>in</strong>g each case. Theseparate record of these data is particularly useful <strong>in</strong> detect<strong>in</strong>gpatients at risk, as well as microorganisms typically associatedwith treatment failures. Although few microorganisms areresponsible for the vast majority of antimicrobial treatmentfailures, the distribution of pathogens is widely divergent23 63–65between centres (fig 4.3).CONCLUSIONMuch progress has been made <strong>in</strong> the understand<strong>in</strong>g of nosocomialpneumonia and this has <strong>in</strong>fluenced managementguidel<strong>in</strong>es. Nevertheless, important issues <strong>in</strong> diagnosis andtreatment rema<strong>in</strong> unresolved. We argue that the controversyover diagnostic tools should be closed. Instead, every effortshould be made to <strong>in</strong>crease our ability to make valid cl<strong>in</strong>icalpredictions about the presence of VAP and to establish criteriato guide restrict<strong>in</strong>g empirical antimicrobial treatment withoutcaus<strong>in</strong>g harm to patients. At the same time, more emphasismust be put on local <strong>in</strong>fection control measures such asrout<strong>in</strong>e surveillance of pathogens, def<strong>in</strong>ition of controlledpolicies of antimicrobial treatment, and effective implementationof strategies of prevention.


Nosocomial pneumonia 29REFERENCES1 Fagon JY, Chastre J, Domart Y, et al. Nosocomial pneumonia <strong>in</strong> patientsreceiv<strong>in</strong>g cont<strong>in</strong>uous mechanical ventilation. Prospective analysis of 52episodes with use of a protected specimen brush and quantitative culturetechnique. Am Rev Respir Dis 1989;139:877–84.2 Torres A, Aznar R, Gatell JM, et al. Incidence, risk, and prognosticfactors of nosocomial pneumonia <strong>in</strong> mechanically ventilated patients. AmRev Respir Dis 1990;142:523–8.3 Rello J, Qu<strong>in</strong>tana E, Aus<strong>in</strong>a V, et al. Incidence, etiology, and outcome ofnosocomial pneumonia <strong>in</strong> mechanically ventilated patients. Chest1991;100:439–44.4 Fagon JY, Chastre J, Vuagnat A, et al. Nosocomial pneumonia andmortality among patients <strong>in</strong> <strong>in</strong>tensive care units. JAMA1996;275:866–9.5 Kollef MH. Ventilator-associated pneumonia. A multivariate analysis.JAMA 1993;270:1965–70.6 Papazian L, Bregeon F, Thirion X, et al. Effect of ventilator-associatedpneumonia on mortality and morbidity. Am J Respir Crit <strong>Care</strong> Med1996;154:91–7.7 Leu HS, Kaiser DL, Mori M, et al. Hospital-acquired pneumoniaattributable mortality and morbidity. Am J Epidemiol 1989;129:1258–67.8 Fagon JY, Chastre J, Hance AJ, et al. Nosocomial pneumonia <strong>in</strong>ventilated patients: a cohort study evaluat<strong>in</strong>g attributable mortality andhospital stay. Am J Med 1993;94:281–8.9 Kollef M, Silver P, Murphy DM, et al. The effect of late-onset pneumonia<strong>in</strong> determ<strong>in</strong><strong>in</strong>g patient mortality. Chest 1995;108:1655–62.10 Jones RN, Pfaller MA. Bacterial resistance: a worldwide problem. DiagnMicrobiol Infect Dis 1998;31:379–88.11 American Thoracic Society. Hospital-acquired pneumonia <strong>in</strong> adults;diagnosis, assessment, <strong>in</strong>itial severity, and prevention. A consensusstatement. Am J Respir Crit <strong>Care</strong> Med 1996;153:1711–25.12 Rello J, Aus<strong>in</strong>a V, Castella J, et al. Nosocomial respiratory tract<strong>in</strong>fections <strong>in</strong> multiple trauma patients. Influence of level of consciousnesswith implications for therapy. Chest 1992;102:525–9.13 Ewig S, Torres A, El-Ebiary M, et al. Bacterial colonization patterns <strong>in</strong>mechanically ventilated patients with traumatic and medical head <strong>in</strong>jury.Incidence, risk factors, and association with ventilator-associatedpneumonia. Am J Respir Crit <strong>Care</strong> Med 1999;159:188–98.14 Akca O, Koltka K, Uzel S, et al. Risk factors for early-onset,ventilator-associated pneumonia <strong>in</strong> critical care patients: selectedmultiresistant versus nonresistant bacteria. Anesthesiology2000;93:638–45.15 Sirvent JM, Torres A, Vidaur L, et al. Tracheal colonisation with<strong>in</strong> 24 hof <strong>in</strong>tubation <strong>in</strong> patients with head trauma: risk factor for develop<strong>in</strong>gearly-onset ventilator-associated pneumonia. Intensive <strong>Care</strong> Med2000;26:1369–72.16 Ibrahim EH, Ward S, Sherman G, et al. A comparative analysis ofpatients with early-onset vs late-onset nosocomial pneumonia <strong>in</strong> the ICUsett<strong>in</strong>g. Chest 2000;117:1434–42.17 Trouillet JL, Chastre J, Vugnat A, et al. Ventilator-associated pneumoniacaused by potentially drug-resistant bacteria. Am J Respir Crit <strong>Care</strong> Med1998;157:531–9.18 Woodhead M, Torres A. Def<strong>in</strong>ition and classification ofcommunity-acquired and nosocomial pneumonias. Eur Respir Mon1997;3:1–12.19 Rello J, Diaz E, Roque M, et al. Risk factors for develop<strong>in</strong>g pneumoniawith<strong>in</strong> 48 hours of <strong>in</strong>tubation. Am J Respir Crit <strong>Care</strong> Med1999;159:1742–6.20 Rello J, Aus<strong>in</strong>a V, Ricart M, et al. Impact of previous antimicrobialtherapy on the etiology and outcome of ventilator-associated pneumonia.Chest 1993;104:1230–5.21 Kollef M. Inadequate antimicrobial treatment: an important determ<strong>in</strong>antof outcome for hospitalized patients. Cl<strong>in</strong> Infect Dis 2000;31(Suppl 4):S131–8.22 Kollef M, Ward S, Sherman G, et al. Inadequate treatment ofnosocomial <strong>in</strong>fections is associated with certa<strong>in</strong> empiric antibioticchoices. Crit <strong>Care</strong> Med 2000;28:3456–64.23 Luna C, Vujachich P, Niederman MS, et al. Impact of BAL data on thetherapy and outcome of ventilator-associated pneumonia. Chest1997;111:676–87.24 Rello J, Sa-Borges M, Correa H, et al. Variations <strong>in</strong> etiology ofventilator-associated pneumonia across four treatment sites: implicationsfor antimicrobial prescrib<strong>in</strong>g practices. Am J Respir Crit <strong>Care</strong> Med1999;160:608–13.25 Kolleff MH, Vlasnik J, Sharpless L, et al. Scheduled change of antibioticclasses: a strategy to decrease the <strong>in</strong>cidence of ventilator-associatedpneumonia. Am J Respir Crit <strong>Care</strong> Med 1997;156:1040–8.26 Gruson D, Hilbert G, Vargas F, et al. Rotation and restricted use ofantibiotics <strong>in</strong> a medical <strong>in</strong>tensive care unit. Impact on the <strong>in</strong>cidence ofventilator-associated pneumonia caused by antibiotic-resistantgram-negative bacteria. Am J Respir Crit <strong>Care</strong> Med 2000;162:8378–43.27 Walger P, Post K, Mayershofer R, et al. Initial antimicrobial policies <strong>in</strong>the ICU should be based on surveillance rather than on crop rotation.Abstracts of the 40th Interscience Conference on Antimicrobial Agentsand Chemotherapy, 401, abstract 89.28 Torres A, Carlet J, and members of the Task Force. ERS task force onventilator-associated pneumonia. Eur Respir J 2001;17:1034–45.29 Fabregas N, Ewig S, Torres A, et al. Cl<strong>in</strong>ical diagnosis ofventilator-associated pneumonia revisited: comparative evaluation us<strong>in</strong>gimmediate postmortem biopsies. Thorax 1999;54:867–73.30 Marquette CH, Wallet F, Cop<strong>in</strong> MC, et al. Relationship betweenmicrobiologic and histiologic features <strong>in</strong> bacterial pneumonia. Am JRespir Crit <strong>Care</strong> Med 1996;154:1784–7.31 Wermert D, Marquette CH, Cop<strong>in</strong> MC, et al. Influence of pulmonarybacteriology and histology on the yield of diagnostic procedures <strong>in</strong>ventilator-acquired pneumonia. Am J Respir Crit <strong>Care</strong> Med1998;158:139–47.32 Wimberley N, Fal<strong>in</strong>g SJ, Bartlett JG. A fiberoptic bronchoscopytechnique to obta<strong>in</strong> uncontam<strong>in</strong>ated lower airway secretion for bacterialculture. Am Rev Respir Dis 1979;119:337–43.33 Kahn FW, Jones JM. Diagnos<strong>in</strong>g bacterial respiratory <strong>in</strong>fection bybronchoalveolar lavage. J Infect Dis 1987;155:862–9.34 Kirkpatrick MB, Bass JB. Quantitative bacterial cultures ofbronchoalveolar lavage fluids and protected brush catheter specimensfrom normal subjects. Am Rev Respir Dis 1989;139:546–8.35 Torres A, Martos A, Puig de la Bellacasa J, et al. Specificity ofendotracheal aspiration, protected specimen brush, and bronchoalveolarlavage <strong>in</strong> mechanically ventilated patients. Am Rev Respir Dis1993;147:952–7.36 Fagon JY, Chastre J, Hance AJ, et al. Detection of nosocomial lung<strong>in</strong>fection <strong>in</strong> ventilated patients. Use of a protected specimen brush andquantitative culture techniques <strong>in</strong> 147 patients. Am Rev Respir Dis1988;138:110–6.37 Torres A, De La Bellacasa JP, Xaubet A, et al. Diagnostic value ofquantitative cultures of bronchoalveolar lavage and telescop<strong>in</strong>g pluggedcatheters <strong>in</strong> mechanically ventilated patients with bacterial pneumonia.Am Rev Respir Dis 1989;140:306–10.38 El-Ebiary M, Torres A, Gonzalez J, et al. Quantitative cultures ofendotracheal aspirates for the diagnosis of ventilator-associatedpneumonia. Am Rev Respir Dis 1993;148:1552–7.39 Marquette CH, Georges H, Wallet F, et al. Diagnostic efficency ofendotracheal aspirates with quatitative bacterial cultures <strong>in</strong> <strong>in</strong>tubatedpatients with suspected pneumonia. Am Rev Respir Dis1993;148:138–44.40 Marquette CH, Herengt F, Mathieu D, et al. Diagnosis of pneumonia <strong>in</strong>mechanically ventilated patients. Repeatability of the protected specimenbrush. Am Rev Respir Dis 1993;147:211–4.41 Rouby JJ, Mart<strong>in</strong> De Lassal E, Poete P, et al. Nosocomialbronchopneumonia <strong>in</strong> the critically ill. Histologic and bacteriologicaspects. Am Rev Respir Dis 1992;146:1059–66.42 Torres A, El-Ebiary M, Padró L, et al. Validation of different techniquesfor the diagnosis of ventilator-associated pneumonia. Am J Respir Crit<strong>Care</strong> Med 1994;149:324–31.43 Chastre J, Fagon JY, Bornet-Lesco M, et al. Evaluation of bronchoscopictechniques for the diagnosis of nosocomial pneumonia. Am J Respir Crit<strong>Care</strong> Med 1995;152:231–40.44 Marquette CH, Cop<strong>in</strong> MC, Wallet F, et al. Diagnostic tests forpneumonia <strong>in</strong> ventilated patients: prospective evaluation of diagnosticaccuracy us<strong>in</strong>g histology as a diagnostic gold standard. Am J Respir Crit<strong>Care</strong> Med 1995;151:1878–88.45 Papazian L, Thomas P, Garbe L, et al. Bronchoscopic or bl<strong>in</strong>d sampl<strong>in</strong>gtechniques for the diagnosis of ventilator-associated pneumonia. Am JRespir Crit <strong>Care</strong> Med 1995;152:1982–91.46 Fabregas N, Torres A, El-Ebiary M, et al. Histopathologic andmicrobiologic aspects of ventilator-associated pneumonia. Anesthesiology1996;84:760–71.47 Kirtland SH, Corley DE, W<strong>in</strong>terbauer RH, et al. The diagnosis ofventilator-associated pneumonia. A comparison of histologic,microbiologic, and cl<strong>in</strong>ical criteria. Chest 1997;112:445–57.48 Dreyfuss D, Mier L, Le Bourdelles G, et al. Cl<strong>in</strong>ical significance ofborderl<strong>in</strong>e quantitative protected brush specimen culture results. Am RevRespir Dis 1993;147:946–51.49 Sanchez-Nieto JM, Torres A, Garcia-Cordoba F, et al. Impact of<strong>in</strong>vasive and non<strong>in</strong>vasive quantitative culture sampl<strong>in</strong>g on outcome ofventilator-associated pneumonia: a pilot study. Am J Respir Crit <strong>Care</strong>Med 1998;157:371–6.50 Fagon JY, Chastre J, Wolff M, et al. Invasive and non<strong>in</strong>vasive strategiesfor management of suspected ventilator-associated pneumonia. Arandomized trial. Ann Intern Med 2000;132:621–30.51 Ruiz M, Torres A, Ewig S, et al. Non<strong>in</strong>vasive versus <strong>in</strong>vasive microbial<strong>in</strong>vestigation <strong>in</strong> ventilator-associated pneumonia: evaluation of outcome.Am J Respir Crit <strong>Care</strong> Med 2000;162:119–25.52 Sole Violan J, Fernandez JA, Benitez AB, et al. Impact of quantitative<strong>in</strong>vasive diagnostic techniques <strong>in</strong> the management and outcome ofmechanically ventilated patients with suspected pneumonia. Crit <strong>Care</strong>Med 2000;28:2737–41.53 Ewig S, Niederman MS, Torres A. <strong>Management</strong> of suspectedventilator-associated pneumonia. Ann Intern Med 2000;133:1008–9.54 Fagon JY, Chastre J. <strong>Management</strong> of suspected ventilator-associatedpneumonia. Ann Intern Med 2000;133:1009.55 Hubmayr RD, Burchardi H, Elliot M, et al. Statement of the 4thInternational Consensus Conference <strong>in</strong> <strong>Critical</strong> <strong>Care</strong> on ICU-AcquiredPneumonia: Chicago, Ill<strong>in</strong>ois, May 2002. Intensive <strong>Care</strong> Med2002;28:1521–36.56 Pug<strong>in</strong> J, Auckenthaler R, Mili N, et al. Diagnosis of ventilator-associatedpneumonia by bacteriologic analysis of bronchoscopic andnonbronchoscopic “bl<strong>in</strong>d” bronchoalveolar lavage fluid. Am Rev RespirDis 1991;143:1121–9.57 Ewig S, Schäfer H, Torres A. Severity assessment <strong>in</strong> community-acquiredpneumonia. Eur Respir J 2000;16:1193–201.58 Chastre J, Wolff M, Fagon JY, et al. Comparisons of two durations ofantibiotic therapy to treat ventilator-associated pneumonia. Am J RespirCrit <strong>Care</strong> Med 2003;167:(Suppl) A21.


30 <strong>Respiratory</strong> <strong>Management</strong> of <strong>Critical</strong> <strong>Care</strong>59 S<strong>in</strong>gh N, Rogers P, Atwood CW, et al. Short-course empiric antibiotictherapy for patients with pulmonary <strong>in</strong>filtrates <strong>in</strong> the <strong>in</strong>tensive care unit. Aproposed solution for <strong>in</strong>discrim<strong>in</strong>ate antibiotic prescription. Am J RespirCrit <strong>Care</strong> Med 200; 162:505–11.60 Niederman MS. Is “crop rotation” of antibiotics the solution to a“resistant” problem <strong>in</strong> the ICU? Am J Respir Crit <strong>Care</strong> Med1997;156:1029–31.61 Ewig S, Dalhoff K, Lorenz J, et al. Deutsche Geselschaft für Pneumologie:Empfehlungen zur Therapie und Prävention der nosokomilaen Pneumonie.Pneumologie 2000;54:525–38.62 Niederman MS. An approach to empiric therapy of nosocomialpneumonia. Med Cl<strong>in</strong> North Am 1994;78:1123–41.63 Alvarez-Lerma F. Modification of empiric antibiotic treatment <strong>in</strong> patientswith pneumonia acquired <strong>in</strong> the <strong>in</strong>tensive care unit. ICU-AcquiredPneumonia Study Group. Intensive <strong>Care</strong> Med 1996;22:387–94.64 Rello J, Gallego M, Mariscal D, et al. The value of rout<strong>in</strong>e microbial<strong>in</strong>vestigation <strong>in</strong> ventilator-associated pneumonia. Am J Respir Crit <strong>Care</strong>Med 1997;156:196–200.65 Kollef MH, Ward S. The <strong>in</strong>fluence of m<strong>in</strong>i-BAL cultures on patientoutcomes: implications for the antibiotic management ofventilator-associated pneumonia. Chest 1998;113:412–20.


5 Acute lung <strong>in</strong>jury and the acute respiratory distresssyndrome: def<strong>in</strong>itions and epidemiologyK Atabai, M A Matthay.............................................................................................................................The acute respiratory distress syndrome(ARDS) is a common cl<strong>in</strong>ical disorder characterisedby <strong>in</strong>jury to the alveolar epithelial andendothelial barriers of the lung, acute <strong>in</strong>flammation,and prote<strong>in</strong> rich pulmonary oedema lead<strong>in</strong>gto acute respiratory failure. S<strong>in</strong>ce its first descriptionby Ashbaugh et al, 1 a considerable volume ofboth basic and cl<strong>in</strong>ical research has led to a moresophisticated appreciation of the pathogenesisand pathophysiology of the syndrome. 2 However,our understand<strong>in</strong>g of the epidemiology andeffects of treatments have been hampered by thelack of uniform def<strong>in</strong>itions. Several attempts havebeen made to provide workable def<strong>in</strong>itions thatwould be useful <strong>in</strong> both cl<strong>in</strong>ical management andresearch. This chapter reviews the def<strong>in</strong>itions andepidemiology of ARDS, with particular attentionto how changes <strong>in</strong> def<strong>in</strong><strong>in</strong>g the syndrome haveaffected our understand<strong>in</strong>g of the natural historyand treatment options.DEFINITIONSBasic def<strong>in</strong>itionIn 1967 Ashbaugh and colleagues described acl<strong>in</strong>ical syndrome of tachypnoea, hypoxaemiaresistant to supplemental oxygen, diffuse alveolar<strong>in</strong>filtrates, and decreased pulmonary compliance<strong>in</strong> 12 patients who required positive pressuremechanical ventilation. The onset of the syndromewas acute, typically with<strong>in</strong> hours of the<strong>in</strong>cit<strong>in</strong>g cl<strong>in</strong>ical disorder. The majority of patientsdid not have a history of pulmonary disease.Adequate oxygenation required the use of cont<strong>in</strong>uouspositive pressure with end expiratorypressures (PEEP) of 5–10 cm H 2O. The earliestradiographic f<strong>in</strong>d<strong>in</strong>gs were patchy <strong>in</strong>filtrates<strong>in</strong>dist<strong>in</strong>guishable from cardiogenic pulmonaryoedema that usually became confluent with progressivecl<strong>in</strong>ical deterioration. Lung compliancewas substantially decreased. Gross lung specimensresembled hepatic tissue with large airwaysbe<strong>in</strong>g free from obstruction. Histological exam<strong>in</strong>ationrevealed hyal<strong>in</strong>e membranes <strong>in</strong> the alveoliwith microscopic atelectasis and <strong>in</strong>tra-alveolarhaemorrhage similar to the <strong>in</strong>fant respiratorydistress syndrome. 1In a subsequent paper Petty and Ashbaughref<strong>in</strong>ed and elaborated on what they co<strong>in</strong>ed the“adult respiratory distress syndrome”. 3 In a reviewof 40 cases the mechanism of lung <strong>in</strong>jury waseither direct (chest trauma, aspiration) or <strong>in</strong>direct(pancreatitis, sepsis) and, <strong>in</strong> some cases, wasattributed to mechanical ventilation. Despite theheterogeneity of <strong>in</strong>cit<strong>in</strong>g events, the physiologicaland pathological response of the lung wasuniform. The use of PEEP was critical <strong>in</strong>ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g acceptable oxygen saturation byreduc<strong>in</strong>g the right to left <strong>in</strong>trapulmonary shuntand <strong>in</strong>creas<strong>in</strong>g the functional residual capacity.Recovery from lung <strong>in</strong>jury could be rapid andcomplete or could progress to <strong>in</strong>terstitial fibrosisand progressive respiratory failure. Fatalities wereprimarily due to septic complications. 3Expanded def<strong>in</strong>itionOver the next two decades the basic def<strong>in</strong>itionwas thought by many experts to be a h<strong>in</strong>drance tounderstand<strong>in</strong>g the syndrome. The def<strong>in</strong>ition wasnot sufficiently specific, was open to vary<strong>in</strong>g<strong>in</strong>terpretations, and did not require the cl<strong>in</strong>icalaetiology of the syndrome to be specified. Investigatorsused different criteria to enrol patients <strong>in</strong>cl<strong>in</strong>ical studies mak<strong>in</strong>g comparison of resultsacross trials difficult. In 1988 Murray andcolleagues proposed an expanded def<strong>in</strong>ition ofARDS <strong>in</strong>tended to describe whether the syndromewas <strong>in</strong> an acute or chronic phase, thephysiological severity of pulmonary <strong>in</strong>jury, andthe primary cl<strong>in</strong>ical disorder associated with thedevelopment of lung <strong>in</strong>jury (table 5.1). 4 The firstpart of the def<strong>in</strong>ition addressed the cl<strong>in</strong>ical courseseparat<strong>in</strong>g acute from chronic cases; patientswith a prolonged course (chronic) were presumablymore likely to develop pulmonary fibrosisand to have poor outcomes. The second part, thelung <strong>in</strong>jury score (LIS), quantified the severity oflung <strong>in</strong>jury from the degree of arterial hypoxaemia,the level of PEEP, the respiratory compliance,and the radiographic abnormalities (table 5.2).F<strong>in</strong>ally, the cause or associated medical conditionwas to be specified. 4 This proposal was accompaniedby an editorial by Petty endors<strong>in</strong>g the newdef<strong>in</strong>ition.The expanded def<strong>in</strong>ition had several advantages.By describ<strong>in</strong>g whether patients had anacute course with rapid resolution or a morechronic course, the def<strong>in</strong>ition differentiated betweenthe rapidly resolv<strong>in</strong>g course typical ofARDS secondary to drug overdoses or pulmonarycontusion and the complicated and protractedcourse of many patients with severe pneumoniaor sepsis syndrome. The LIS quantified the severityof lung <strong>in</strong>jury separat<strong>in</strong>g patients with severelung <strong>in</strong>jury (LIS >2.5) from those with mild lung<strong>in</strong>jury (LIS 0.1). Most importantly, theidentification of the cause or associated medicalcondition addressed the aetiology of lung <strong>in</strong>jury.As the authors argued, group<strong>in</strong>g all causes ofARDS under an umbrella classification potentiallyprevented the discovery of beneficial treatmentsaimed at a particular cause. 4.................................................Abbreviations: ALI, acute lung <strong>in</strong>jury; ARDS, acuterespiratory distress syndrome; PEEP, positive endexpiratory pressure; PaO 2, arterial oxygen tension; FiO 2,fractional <strong>in</strong>spired oxygen; PAO 2, alveolar oxygen tension;LIS, lung <strong>in</strong>jury score; PAOP, pulmonary artery occlusionpressure.


32 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Table 5.1 Three part expanded def<strong>in</strong>ition of cl<strong>in</strong>icalacute lung <strong>in</strong>jury (ALI) and the acute respiratorydistress syndrome (ARDS) proposed by Murray andcolleagues 4Part 1Part 2Part 3Acute or chronic, depend<strong>in</strong>g on courseSeverity of physiological lung <strong>in</strong>jury as determ<strong>in</strong>ed bythe lung <strong>in</strong>jury score (see table 5.2)Lung <strong>in</strong>jury caused by or associated with known riskfactor for ARDS such as sepsis, pneumonia, aspiration,or major traumaTable 5.3 1994 consensus conference def<strong>in</strong>ition ofacute lung <strong>in</strong>jury (ALI) and the acute respiratorydistress syndrome (ARDS)OnsetOxygenation criteriaExclusion criteriaRadiographic criteria+Acute and persistent• PaO 2 /FiO 2 300 0PAO 2 /FiO 2 225–299 1PaO 2 /FiO 2 175–224 2PaO 2 /FiO 2 100–174 3PaO 2 /FiO 2 80 ml/cm H 2 O 060–79 ml/cm H 2 O 140–59 ml/cm H 2 O 220–39 ml/cm H 2 O 32.5NAECC def<strong>in</strong>itionIn 1994 the North American-European Consensus Conference(NAECC) on ARDS proposed a revised def<strong>in</strong>ition for acutelung <strong>in</strong>jury (ALI) and ARDS (table 5.3). 5 The panel recognisedthat accurate estimates of the <strong>in</strong>cidence and outcomes ofARDS were h<strong>in</strong>dered by the lack of a simple uniformdef<strong>in</strong>ition, especially one that could be used to enrol patients<strong>in</strong> cl<strong>in</strong>ical studies. The panel changed “adult” back to “acuterespiratory distress syndrome”, recognis<strong>in</strong>g that the syndromewas not limited to adults (the orig<strong>in</strong>al Ashbaugh report<strong>in</strong>cluded one 11 year old patient). Mechanical ventilation wasnot a requirement, although it was anticipated that mostcl<strong>in</strong>ical trials would only enrol <strong>in</strong>tubated patients. In order toexclude chronic lung disease, the def<strong>in</strong>ition required an acuteonset of respiratory failure.The physiological severity of lung <strong>in</strong>jury was addressed byus<strong>in</strong>g the term ALI to refer to patients with a PaO 2/FiO 2ratio of


Acute lung <strong>in</strong>jury and ARDS 33Table 5.4Strengths and limitations of the different def<strong>in</strong>itions of ARDSDef<strong>in</strong>ition Strengths LimitationsPetty and Ashbaugh (1971) 3Murray et al (1988) 4• Detailed cl<strong>in</strong>ical description of thehallmarks of ARDS which rema<strong>in</strong>srelevant today• Three part def<strong>in</strong>ition evaluateschronicity, severity, and cause of lung<strong>in</strong>juryNorth American-European • Simple criteria which are easy toConsensus Conferenceapply <strong>in</strong> the cl<strong>in</strong>ical sett<strong>in</strong>g(1994) 5 • Disease spectrum recognised byseparation of ARDS from ALI• No formal criteria foridentification of patients• Lung <strong>in</strong>jury score has not beenpredictive of mortality• No formal criteria to excludecardiogenic pulmonary oedema• Cause of lung <strong>in</strong>jury not required• Radiographic criteria notsufficiently specificdiagnoses were followed prospectively for the development ofARDS. The diagnostic accuracy <strong>in</strong> the “at risk” population(true positive + true negative/total number of patients) was90% for the LIS def<strong>in</strong>ition and 97% for both the modified LISand the NAECC def<strong>in</strong>itions (p=0.03). The authors concludedthat the three scor<strong>in</strong>g systems identified similar populationswhen applied to patients with clearly def<strong>in</strong>ed at riskdiagnoses.A more recent study assessed the agreement between thedef<strong>in</strong>itions of Murray et al and the NAECC <strong>in</strong> diagnos<strong>in</strong>g ARDS<strong>in</strong> a prospective trial of 118 patients compar<strong>in</strong>g ventilationstrategies. 10 The <strong>in</strong>cidence us<strong>in</strong>g the LIS was 62% while thatus<strong>in</strong>g the NAECC def<strong>in</strong>ition was 55%. Statistical agreementbetween the two def<strong>in</strong>itions was moderate and improvedwhen analysis was limited to patients who had undergonecompliance measurements (65%). A more liberal PaO 2/FiO 2ratio decreased agreement, as did <strong>in</strong>creas<strong>in</strong>g the LIS thresholddiagnostic of ARDS to 3 or decreas<strong>in</strong>g it to 2. Omitt<strong>in</strong>g data onoxygenation, chest radiography, PAOP, PEEP, or respiratorycompliance either decreased agreement or left it unchanged.While agreement between the def<strong>in</strong>itions was only moderate,there was no difference <strong>in</strong> mortality between the two groupsof patients identified. The authors concluded that, for <strong>in</strong>vestigativepurposes, the two criteria could be used <strong>in</strong>terchangeably.Significance of def<strong>in</strong>itions <strong>in</strong> cl<strong>in</strong>ical trials11 12The NAECC updated its recommendations <strong>in</strong> 1998.Although no formal changes were made, the Committeeemphasised the importance of address<strong>in</strong>g epidemiologicaland aetiological differences between patients when design<strong>in</strong>gcl<strong>in</strong>ical trials. Several prospective studies had identified riskfactors present at the onset of lung <strong>in</strong>jury that predictedpoorer outcomes 13–16 ; cl<strong>in</strong>ical trial organisers would need toensure an equal distribution of these risk factors <strong>in</strong> theirexperimental and control arms. They also encouraged furtherresearch focused on identify<strong>in</strong>g markers predictive of progressionto or poor outcomes from lung <strong>in</strong>jury. 11Previousdef<strong>in</strong>itions had relied on abnormalities of lung physiology tograde <strong>in</strong>jury 1 3–5 ; however, neither the <strong>in</strong>itial LIS nor the <strong>in</strong>itialPaO 2/FiO 2ratio was predictive of mortality <strong>in</strong> cl<strong>in</strong>icaltrials. 13 14 16 17 Some biological markers, on the other hand, hadalready been proved to be useful <strong>in</strong> identify<strong>in</strong>g patients at riskfor poor outcomes. For example, raised levels of procollagen IIIpeptide <strong>in</strong> early bronchoalveolar lavage and pulmonaryoedema fluid samples predicted a protracted cl<strong>in</strong>ical coursewith progression to pulmonary fibrosis <strong>in</strong> patients with ALI/18 19ARDS, and <strong>in</strong>creased levels of von Willebrand factorantigen <strong>in</strong> plasma predicted the development of lung <strong>in</strong>jury <strong>in</strong>patients with non-pulmonary sepsis syndrome. 20 Also, a recentstudy reported that higher levels of von Willebrand factorantigens <strong>in</strong> the plasma of ALI/ARDS patients <strong>in</strong>dependentlypredicted mortality early <strong>in</strong> the cl<strong>in</strong>ical course. 21The importance of standard def<strong>in</strong>itions and a mechanisticapproach to enroll<strong>in</strong>g patients <strong>in</strong> cl<strong>in</strong>ical trials are apparent <strong>in</strong>the results of two recent randomised multicentre trials. Thefirst, conducted by the ARDS Network, evaluated the benefitsof a low tidal volume strategy of mechanical ventilation andfound an absolute reduction <strong>in</strong> the primary outcome of deathprior to hospital discharge of 9% us<strong>in</strong>g lower tidal volumes(22% relative reduction <strong>in</strong> mortality). 22 Patients were enrolledus<strong>in</strong>g the 1994 def<strong>in</strong>ition and the benefit of a protective ventilationstrategy was ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> all subsets of patients withALI/ARDS. 23 This was the first large trial to show the benefit ofany <strong>in</strong>tervention <strong>in</strong> ALI/ARDS. 224The importance of identify<strong>in</strong>g the mechanism of lung<strong>in</strong>jury is borne out by a recent trial of recomb<strong>in</strong>ant humanactivated prote<strong>in</strong> C <strong>in</strong> severe sepsis. 25A 96 hour <strong>in</strong>fusion ofprote<strong>in</strong> C resulted <strong>in</strong> an absolute reduction <strong>in</strong> mortality of6.1% (20% relative reduction) <strong>in</strong> a large <strong>in</strong>ternationalmulticentre trial; 54% of the 1690 patients had a pulmonarysource of sepsis and 75% required mechanical ventilation onentry <strong>in</strong>to the study. Although the study did not report thenumber of patients who met the criteria for ALI/ARDS, it islikely that most did s<strong>in</strong>ce ARDS is the most common cause ofrespiratory failure <strong>in</strong> sepsis and sepsis is the most commonpredispos<strong>in</strong>g factor for the development of ARDS. 226 Based onthis study, it is likely that a trial of prote<strong>in</strong> C <strong>in</strong> patients withlung <strong>in</strong>jury due to sepsis will show a treatment benefit whilethe same trial <strong>in</strong> patients with ARDS due to trauma or fatemboli may not. As more is learned about the epidemiologyand pathophysiology of ARDS, identify<strong>in</strong>g the cause of <strong>in</strong>jury<strong>in</strong> design<strong>in</strong>g treatment trials will become <strong>in</strong>creas<strong>in</strong>gly critical.EPIDEMIOLOGYIncidenceThe <strong>in</strong>cidence of ALI and ARDS has been difficult to establish.Most studies were conducted before the NAECC def<strong>in</strong>itionwas proposed and used different criteria to enrol patients.Def<strong>in</strong><strong>in</strong>g the population at risk <strong>in</strong> a given study has beenequally problematic. Accurate measurement of disease <strong>in</strong>cidencerequires knowledge of the number of people with thedisease with<strong>in</strong> a def<strong>in</strong>ed population at risk for develop<strong>in</strong>g it.Prospective trials must account for the catchment area of thehospitals studied; each hospital’s catchment area may overlapwith that of several other hospitals.In 1972 the National Heart and Lung Institute (NHLI) taskforce estimated an <strong>in</strong>cidence of 75 cases of ARDS per 100 000population per year. 27 Several subsequent studies haveestimated a much lower annual <strong>in</strong>cidence of 1.5–13.5 cases per100 000 population (table 5.5). 28–32 There are several reasonswhy the number of cases may have been overestimated. Thetask force predated the widespread acceptance of thedef<strong>in</strong>ition of ARDS and used a broad def<strong>in</strong>ition of lung <strong>in</strong>jurythat <strong>in</strong>cluded conditions such as renal failure and volumeoverload. In addition, the population at risk was not clearly


34 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Table 5.5 Annual <strong>in</strong>cidence of the acute respiratory distress syndrome (ARDS) and acute lung <strong>in</strong>jury (ALI) <strong>in</strong> differentcl<strong>in</strong>ical studiesStudyCriteria used to diagnoseALI/ARDSIncidence(cases/100 000population/year)Study limitationsNHLI task force 27 75 • Broad def<strong>in</strong>ition of respiratory distress syndrome <strong>in</strong>clud<strong>in</strong>g patients withvolume overloadCanary Islands 31 PaO 2 /FiO 2


Acute lung <strong>in</strong>jury and ARDS 35The second study from Seattle def<strong>in</strong>ed ARDS as a PaO 2/FiO 2ratio of


36 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong><strong>in</strong>jury represented a turn<strong>in</strong>g po<strong>in</strong>t towards a more quantitativeapproach. The NAECC 1994 def<strong>in</strong>ition simplified thediagnostic criteria proposed by Murray and colleagues byelim<strong>in</strong>at<strong>in</strong>g the level of PEEP from the oxygenation criteriaand the measurement of respiratory compliance, and reduc<strong>in</strong>gthe chest radiographic <strong>in</strong>clusion criteria to the presence ofbilateral opacities consistent with pulmonary oedema. As withthe def<strong>in</strong>ition of Murray et al, the NAECC def<strong>in</strong>ition gradedlung <strong>in</strong>jury by def<strong>in</strong><strong>in</strong>g different oxygenation criteria for ALIand ARDS. In the past decade the application of these twodef<strong>in</strong>itions has led to substantial progress <strong>in</strong> the understand<strong>in</strong>gof the natural history of lung <strong>in</strong>jury. Although the two criteriadef<strong>in</strong>e overlapp<strong>in</strong>g populations with a similar prognosis,it is not clear whether conclusions generated us<strong>in</strong>g one def<strong>in</strong>itioncan be extrapolated to populations def<strong>in</strong>ed by the other. Itis hoped that future trials will use the NAECC def<strong>in</strong>itionexclusively.Several aspects of the def<strong>in</strong>ition of ALI/ARDS need furtherref<strong>in</strong>ement. Although <strong>in</strong>itial oxygenation <strong>in</strong>dices have littleprognostic value, the more liberal oxygenation criteriadescrib<strong>in</strong>g ALI appear to identify patients with similarbasel<strong>in</strong>e characteristics and prognosis at an earlier stage of theillness. If there is no prognostic or epidemiological differencebetween ALI and ARDS patients, then the two categoriesshould be comb<strong>in</strong>ed. On the other hand, <strong>in</strong> certa<strong>in</strong> subsetssuch as trauma, patients with ALI may have better a prognosisthan those with ARDS. While the radiographic criteria ofthe NAECC def<strong>in</strong>ition are easy to apply, recent data suggestthat there is significant <strong>in</strong>terobserver variability. Although amore standardised approach is desirable, complex schemasquantify<strong>in</strong>g the severity of <strong>in</strong>filtrates <strong>in</strong> different quadrantshave no prognostic value. It therefore appears that anapproach that comb<strong>in</strong>es the simplicity of the NAECCdef<strong>in</strong>ition with more standardised and specific criteria wouldbe ideal.The risk of an <strong>in</strong>dividual develop<strong>in</strong>g lung <strong>in</strong>jury and itsprognosis will be more predictable as more accurate physiologicalmarkers are identified. Interest<strong>in</strong>gly, a recent largeprospective cl<strong>in</strong>ical study of 179 patients has found that amarkedly raised dead space fraction (0.58) occurs early <strong>in</strong> thecourse of ARDS and is <strong>in</strong>dependently associated withmortality. 48As the pathogenesis and epidemiology of lung<strong>in</strong>jury are elucidated, treatment may be <strong>in</strong>dividualised aroundthe mechanism of <strong>in</strong>jury and the cl<strong>in</strong>ical characteristics ofeach patient.REFERENCES1 Ashbaugh DG, Bigelow DB, Petty TL, et al. Acute respiratory distress <strong>in</strong>adults. Lancet 1967;ii:319–23.2 Ware LB, Matthay MA. The acute respiratory distress syndrome. N EnglJ Med 2000;342:1334–49.3 Petty TL, Ashbaugh DG. The adult respiratory distress syndrome. Cl<strong>in</strong>icalfeatures, factors <strong>in</strong>fluenc<strong>in</strong>g prognosis and pr<strong>in</strong>ciples of management.Chest 1971;60:233–9.4 Murray JF, Matthay MA, Luce JM, et al. An expanded def<strong>in</strong>ition of theadult respiratory distress syndrome. Am Rev Respir Dis1988;138:720–3; erratum 1989;139:1065.5 Bernard GR, Artigas A, Brigham KL, et al. The American-EuropeanConsensus Conference on ARDS. Def<strong>in</strong>itions, mechanisms, relevantoutcomes, and cl<strong>in</strong>ical trial coord<strong>in</strong>ation. Am J Respir Crit <strong>Care</strong> Med1994;149:818–24.6 Sloane PJ, Gee MH, Gottlieb JE, et al. A multicenter registry of patientswith acute respiratory distress syndrome. Physiology and outcome. AmRev Respir Dis 1992;146:419–26.7 Rubenfeld GD, Caldwell E, Granton J, et al. Interobserver variability <strong>in</strong>apply<strong>in</strong>g a radiographic def<strong>in</strong>ition for ARDS. Chest 1999;116:1347–53.8 Meade MO, Cook RJ, Guyatt GH, et al. Interobserver variation <strong>in</strong><strong>in</strong>terpret<strong>in</strong>g chest radiographs for the diagnosis of acute respiratorydistress syndrome. Am J Respir Crit <strong>Care</strong> Med 2000;161:85–90.9 Moss M, Goodman PL, He<strong>in</strong>ig M, et al. Establish<strong>in</strong>g the relativeaccuracy of three new def<strong>in</strong>itions of the adult respiratory distresssyndrome. Crit <strong>Care</strong> Med 1995;23:1629–37.10 Meade MO, Guyatt GH, Cook RJ, et al. Agreement between alternativeclassification of acute respiratory distress syndrome. Am J Respir Crit<strong>Care</strong> Med 2001;163:490–3.11 Abraham E, Matthay MA, D<strong>in</strong>arello CA, et al. Consensus conferencedef<strong>in</strong>itions for sepsis, septic shock, acute lung <strong>in</strong>jury, and acuterespiratory distress syndrome: time for a reevaluation. Crit <strong>Care</strong> Med2000;28:232–5.12 Artigas A, Bernard GR, Carlet J, et al. The American-EuropeanConsensus Conference on ARDS, Part 2. Ventilatory, pharmacologic,supportive therapy, study design strategies and issues related to recoveryand remodel<strong>in</strong>g. Intensive <strong>Care</strong> Med 1998;24:378–98.13 Doyle RL, Szaflarski N, Mod<strong>in</strong> GW, et al. Identification of patients withacute lung <strong>in</strong>jury. Predictors of mortality. Am J Respir Crit <strong>Care</strong> Med1995;152:1818–24.14 Monchi M, Bellenfant F, Cariou A, et al. Early predictive factors ofsurvival <strong>in</strong> the acute respiratory distress syndrome. A multivariateanalysis. Am J Respir Crit <strong>Care</strong> Med 1998;158:1076–81.15 Moss M, Bucher B, Moore FA, et al. The role of chronic alcohol abuse <strong>in</strong>the development of acute respiratory distress syndrome <strong>in</strong> adults. JAMA1996;275:50–4.16 Zilberberg MD, Epste<strong>in</strong> SK. Acute lung <strong>in</strong>jury <strong>in</strong> the medical ICU:comorbid conditions, age, etiology, and hospital outcome. Am J RespirCrit <strong>Care</strong> Med 1998;157:1159–64.17 Krafft P, Fridrich P, Pernerstorfer T, et al. The acute respiratory distresssyndrome: def<strong>in</strong>itions, severity and cl<strong>in</strong>ical outcome. An analysis of 101cl<strong>in</strong>ical <strong>in</strong>vestigations. Intensive <strong>Care</strong> Med 1996;22:519–29.18 Chesnutt AN, Matthay MA, Tibayan FA, et al. Early detection of type IIIprocollagen peptide <strong>in</strong> acute lung <strong>in</strong>jury. Pathogenetic and prognosticsignificance. Am J Respir Crit <strong>Care</strong> Med 1997;156:840–5.19 Clark JG, Milberg JA, Ste<strong>in</strong>berg KP, et al. Type III procollagen peptide<strong>in</strong> the adult respiratory distress syndrome. Association of <strong>in</strong>creasedpeptide levels <strong>in</strong> bronchoalveolar lavage fluid with <strong>in</strong>creased risk fordeath. Ann Intern Med 1995;122:17–23.20 Rub<strong>in</strong> DB, Wiener-Kronish JP, Murray JF, et al. Elevated von Willebrandfactor antigen is an early plasma predictor of acute lung <strong>in</strong>jury <strong>in</strong>nonpulmonary sepsis syndrome. J Cl<strong>in</strong> Invest 1990;86:474–80.20 Pug<strong>in</strong> J, Verghese G, Widmer MC, et al. The alveolar space is the siteof <strong>in</strong>tense <strong>in</strong>flammatory and profibrotic reactions <strong>in</strong> the early phase ofacute respiratory distress syndrome. Crit <strong>Care</strong> Med 1999;27:304–12.21 Ware LB, Conner ER, Matthay MA. Von Willebrand factor antigen is an<strong>in</strong>dependent marker of poor outcome <strong>in</strong> patients with early acute lung<strong>in</strong>jury. Crit <strong>Care</strong> Med 2001;29:2325–31.22 ARDS Network. Ventilation with lower tidal volumes as compared withtraditional tidal volumes for acute lung <strong>in</strong>jury and the acute respiratorydistress syndrome. The Acute <strong>Respiratory</strong> Distress Syndrome Network. NEngl J Med 2000;342:1301–8.23 Eisner MD, Thompson T, Hudson LD, et al. Efficacy of low tidal volumeventilation <strong>in</strong> patients with different cl<strong>in</strong>ical risk factors for acute lung<strong>in</strong>jury and the acute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong>Med 2001 (<strong>in</strong> press).24 McIntyre RC Jr, Pulido EJ, Bensard DD, et al. Thirty years of cl<strong>in</strong>icaltrials <strong>in</strong> acute respiratory distress syndrome. Crit <strong>Care</strong> Med2000;28:3314–31.25 Bernard GR, V<strong>in</strong>cent J, Laterre P, et al. Efficacy and safety ofrecomb<strong>in</strong>ant human sctivated prote<strong>in</strong> C for severe sepsis. N Engl J Med2001;344:699–709.26 Matthay MA. Severe sepsis: a new treatment with both anticoagulantand anti<strong>in</strong>flammatory properties. N Engl J Med 2001;344:759–61.27 National Heart and Lung Institute. Task force on problems, researchapproaches, needs: the lung program. Wash<strong>in</strong>gton, DC: Department ofHealth, Education, and Welfare, 1972: 165–80.28 Lewandowski K, Metz J, Deutschmann C, et al. Incidence, severity, andmortality of acute respiratory failure <strong>in</strong> Berl<strong>in</strong>, Germany. Am J Respir Crit<strong>Care</strong> Med 1995;151:1121–5.29 Luhr OR, Antonsen K, Karlsson M, et al. Incidence and mortality afteracute respiratory failure and acute respiratory distress syndrome <strong>in</strong>Sweden, Denmark, and Iceland. The ARF Study Group. Am J Respir Crit<strong>Care</strong> Med 1999;159:1849–61.30 Thomsen GE, Morris AH. Incidence of the adult respiratory distresssyndrome <strong>in</strong> the state of Utah. Am J Respir Crit <strong>Care</strong> Med1995;152:965–71.31 Villar J, Slutsky AS. The <strong>in</strong>cidence of the adult respiratory distresssyndrome. Am Rev Respir Dis 1989;140:814–6.32 Webster NR, Cohen AT, Nunn JF. Adult respiratory distress syndrome:how many cases <strong>in</strong> the UK? Anaesthesia 1988;43:923–6.33 Hudson LD, Ste<strong>in</strong>berg KP. Epidemiology of acute lung <strong>in</strong>jury and ARDS.Chest 1999;116:74–82S.34 Ste<strong>in</strong>berg KP, Hudson LD. Acute lung <strong>in</strong>jury and acute respiratorysyndrome. Cl<strong>in</strong> Chest Med 2000;21:401–17.35 Fowler AA, Hamman RF, Good JT, et al. Adult respiratory distresssyndrome: risk with common predispositions. Ann Intern Med1983;98:593–7.36 Hudson LD, Milberg JA, Anardi D, et al. Cl<strong>in</strong>ical risks for developmentof the acute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong> Med1995;151:293–301.37 Pepe PE, Potk<strong>in</strong> RT, Reus DH, et al. Cl<strong>in</strong>ical predictors of the adultrespiratory distress syndrome. Am J Surg 1982;144:124–30.38 Hyers TM. Prediction of survival and mortality <strong>in</strong> patients with adultrespiratory distress syndrome. New Horiz 1993;1:466–70.39 Hallengren B, Forsgren A. Effect of alcohol on chemotaxis, adherenceand phagocytosis of human polymorphonuclear leucocytes. Acta MedScand 1978;204:43–8.40 Moss M, Guidot DM, Ste<strong>in</strong>berg KP, et al. Diabetic patients have adecreased <strong>in</strong>cidence of acute respiratory distress syndrome. Crit <strong>Care</strong>Med 2000;28:2187–92.


Acute lung <strong>in</strong>jury and ARDS 3741 Frank JA, Nuckton TJ, Matthay MA. Diabetes mellitus: a negativepredictor for the development of acute respiratory distress syndrome fromseptic shock. Crit <strong>Care</strong> Med 2000;28:2645–6.42 Bell RC, Coalson JJ, Smith JD, et al. Multiple organ system failure and<strong>in</strong>fection <strong>in</strong> adult respiratory distress syndrome. Ann Intern Med1983;99:293–8.43 Heffner JE, Brown LK, Barbieri CA, et al. Prospective validation of anacute respiratory distress syndrome predictive score. Am J Respir Crit<strong>Care</strong> Med 1995;152:1518–26.44 Ferr<strong>in</strong>g M, V<strong>in</strong>cent JL. Is outcome from ARDS related to the severity ofrespiratory failure? Eur Respir J 1997;10:1297–300.45 Roupie E, Lepage E, Wysocki M, et al. Prevalence, etiologies andoutcome of the acute respiratory distress syndrome among hypoxemicventilated patients. SRLF Collaborative Group on Mechanical Ventilation.Société de Réanimation de Langue Française. Intensive <strong>Care</strong> Med1999;25:920–9.46 Suchyta MR, Clemmer TP, Elliott CG, et al. The adult respiratory distresssyndrome. A report of survival and modify<strong>in</strong>g factors. Chest1992;101:1074–9.47 Ely WE, Wheeler AP, Thompson BT, et al. Recovery rate and prognosis<strong>in</strong> older persons who develop lung <strong>in</strong>jury and the acute respiratorydistress syndrome. Ann Intern Med 2002;136:25–36.48 Nuckton TJ, Alonso J, Kallet RH, et al. Pulmonary dead space fractionas a risk factor for mortality <strong>in</strong> the acute respiratory distress syndrome. NEngl J Med 2002;346:1281–6.


6 The pathogenesis of acute lung <strong>in</strong>jury/acuterespiratory distress syndromeG J Bell<strong>in</strong>gan.............................................................................................................................Lung <strong>in</strong>jury is the term used to describe the pulmonaryresponse to a broad range of <strong>in</strong>juriesoccurr<strong>in</strong>g either directly to the lung or as theconsequence of <strong>in</strong>jury or <strong>in</strong>flammation at othersites <strong>in</strong> the body. Acute respiratory distresssyndrome (ARDS) represents the more severe endof the spectrum of this condition <strong>in</strong> which thereare widespread <strong>in</strong>flammatory changes throughoutthe lung, usually accompanied by aggressivefibrosis. 1–5 The pathogenesis of lung <strong>in</strong>jury is notwell understood. 15We do not know why somepeople progress to ARDS while others whosusta<strong>in</strong> <strong>in</strong>dist<strong>in</strong>guishable <strong>in</strong>juries rema<strong>in</strong> relativelyunaffected. 6 ARDS is unique among pulmonaryfibrotic conditions <strong>in</strong> that the fibrosisresolves almost completely <strong>in</strong> many cases; onceaga<strong>in</strong> the mechanisms for this are notunderstood. 7 It is now well recognised that someof the damage is created and exacerbated bymechanical ventilation. 8 However, mortality fromARDS has improved <strong>in</strong> certa<strong>in</strong> centres over thelast 10 years, 910 predat<strong>in</strong>g major changes <strong>in</strong> ventilatorypractice; the reasons for this improvement<strong>in</strong> mortality are thus also not clear.ARDS often occurs as part of a wider picture ofmultiorgan dysfunction syndrome (MODS). 11Central to the pathogenesis are an explosive<strong>in</strong>flammatory process and the reparative responses<strong>in</strong>voked <strong>in</strong> an attempt to heal this. Thischapter will outl<strong>in</strong>e the pathological processeswhich occur dur<strong>in</strong>g ARDS and their evolution asour understand<strong>in</strong>g of the <strong>in</strong>flammatory, immune,and fibroproliferative responses has grown. It willdescribe the underly<strong>in</strong>g cellular and molecularprocesses, correlate these with the cl<strong>in</strong>ical picture,and highlight how such <strong>in</strong>sights can lead to noveltherapeutic approaches.DEFINITIONSLung <strong>in</strong>jury (acute lung <strong>in</strong>jury (ALI) and ARDS)is currently def<strong>in</strong>ed cl<strong>in</strong>ically by gas exchangeand chest radiographic abnormalities whichoccur shortly after a known predispos<strong>in</strong>g <strong>in</strong>juryand <strong>in</strong> the absence of heart failure. 1 The def<strong>in</strong>itionand range of predispos<strong>in</strong>g conditions are discussed<strong>in</strong> chapter 5.The pathophysiology of ARDS is driven by anaggressive <strong>in</strong>flammatory reaction. Indirect <strong>in</strong>juryoccurs as part of a systemic <strong>in</strong>flammatoryresponse syndrome (SIRS), which can be due to<strong>in</strong>fective or non-<strong>in</strong>fective causes such as pancreatitisor trauma; when SIRS is caused by <strong>in</strong>fectionit is called sepsis. SIRS with organ dysfunction iscalled severe sepsis and, <strong>in</strong> the presence ofsignificant hypotension, septic shock. 12 ARDS canthus occur <strong>in</strong> conjunction with failure of otherorgans, the multiorgan dysfunction syndrome(MODS). A number of endogenous anti<strong>in</strong>flammatorymechanisms are also <strong>in</strong>itiated tocounterbalance the effects of such an aggressive<strong>in</strong>flammatory response and this is termed thecompensatory anti-<strong>in</strong>flammatory response syndrome(CARS), although these responses too maybe excessive and contribute to a state of13 14immunoparesis.PATHOLOGYLung <strong>in</strong>jury is an evolv<strong>in</strong>g condition and thepathological features of ARDS are typicallydescribed as pass<strong>in</strong>g through three overlapp<strong>in</strong>gphases (table 6.1)—an <strong>in</strong>flammatory or exudativephase, a proliferative phase and, lastly, a fibroticphase. 2–5These phases are complicated by othervariables—for example, episodes of nosocomialpneumonia and the deleterious effects of ventilator<strong>in</strong>duced lung <strong>in</strong>jury. Moreover, the <strong>in</strong>itiat<strong>in</strong>g<strong>in</strong>sults themselves may <strong>in</strong>fluence the pathophysiologicalpicture. Radiographic differences havebeen identified between patients with ARDS aris<strong>in</strong>gfrom direct pulmonary <strong>in</strong>juries comparedwith those from <strong>in</strong>direct <strong>in</strong>juries. 15Likewise, asmall study has suggested greater areas of alveolarcollapse and oedema <strong>in</strong> patients dy<strong>in</strong>g withARDS from direct rather than <strong>in</strong>direct <strong>in</strong>juries. 16Although these differences may translate <strong>in</strong>todifferences <strong>in</strong> outcome, no clear mechanistic differencesbetween these groups have beenidentified. 17A recent study has suggested, however,that lung cyclo-oxygenase-2 (COX-2) geneexpression (a gene implicated <strong>in</strong> the early pro<strong>in</strong>flammatoryresponse) is only <strong>in</strong>duced by <strong>in</strong>directmechanisms related to the systemic response toendotox<strong>in</strong> rather than directly <strong>in</strong> response to<strong>in</strong>haled endotox<strong>in</strong>. 18Exudative phaseTypically, this lasts for the first week after theonset of symptoms. The histological changes aretermed diffuse alveolar damage. 5At necroscopicexam<strong>in</strong>ation the lungs are heavy, rigid and, whensectioned, do not exude fluid because of its highprote<strong>in</strong> content. Bachofen and Wiebel wereamong the first to study the histopathologicalchanges <strong>in</strong> detail <strong>in</strong> patients who died withARDS. 19 An acute stage commenc<strong>in</strong>g with<strong>in</strong> thefirst 24 hours of symptoms was marked bysignificant prote<strong>in</strong>aceous and often markedlyhaemorrhagic <strong>in</strong>terstitial and alveolar oedemawith hyal<strong>in</strong>e membranes. The hyal<strong>in</strong>e membranesare eos<strong>in</strong>ophilic conta<strong>in</strong><strong>in</strong>g fibr<strong>in</strong>, immunoglobul<strong>in</strong>,and complement. The microvascularand alveolar barriers have focal areas of damageand the alveolar wall is oedematous with areas ofnecrosis with<strong>in</strong> the epithelial l<strong>in</strong><strong>in</strong>g, although thebasal lam<strong>in</strong>a is <strong>in</strong>tact <strong>in</strong>itially. The early endotheliallesions are more subtle, conta<strong>in</strong><strong>in</strong>g areas ofnecrosis and denuded spaces usually filled withfibr<strong>in</strong> clot. Neutrophils are found <strong>in</strong>creas<strong>in</strong>glydur<strong>in</strong>g the <strong>in</strong>itial phases <strong>in</strong> capillaries, <strong>in</strong>terstitialtissue, and progressively with<strong>in</strong> airspaces. 20


The pathogenesis of ALI/ARDS 39Table 6.1Summary of some histopathological changes <strong>in</strong> ARDSExudative phase Proliferative phase Fibrotic phaseMacroscopic • Heavy, rigid, dark • Heavy, grey • CobblestonedMicroscopic • Hyal<strong>in</strong>e membranes • Barrier disruption • Fibrosis• Oedema • Oedema • Macrophages• Neutrophils• Alveolar type II cell • Lymphocytesproliferation• Epithelial>endothelial • Myofibroblast <strong>in</strong>filtration • Matrix organisationdamage• Neutrophils• Deranged ac<strong>in</strong>ararchitecture• Alveolar collapse • Patchy emphysematouschange• Alveoli filled with cells andorganis<strong>in</strong>g matrix• Epithelial apoptosis• FibroproliferationVasculature • Local thrombus • Loss of capillaries • Myo<strong>in</strong>timal thicken<strong>in</strong>g• Pulmonary hypertension • Tortuous vesselsProliferative phaseTypically occupy<strong>in</strong>g the second 2 weeks after the onset of respiratoryfailure, the proliferative phase is characterised by2–5 19 21organisation of the exudates and by fibrosis. The lungrema<strong>in</strong>s heavy and solid, and microscopically the <strong>in</strong>tegrity ofthe lung architecture becomes steadily more deranged. Thecapillary network is damaged and there is a progressivedecl<strong>in</strong>e <strong>in</strong> the profile of capillaries <strong>in</strong> tissue sections; later,<strong>in</strong>timal proliferation is evident <strong>in</strong> many small vessels furtherreduc<strong>in</strong>g the lum<strong>in</strong>al area. The <strong>in</strong>terstitial space becomesgrossly dilated, necrosis of type I pneumocytes exposes areasof epithelial basement membrane, and the alveolar lumen fillswith leucocytes, red cells, fibr<strong>in</strong>, and cell debris. Alveolar typeII cells proliferate <strong>in</strong> an attempt to cover the denuded epithelialsurfaces and differentiate <strong>in</strong>to type I cells. 22Fibroblastsbecome apparent <strong>in</strong> the <strong>in</strong>terstitial space and later <strong>in</strong> thealveolar lumen. 23 These processes result <strong>in</strong> extreme narrow<strong>in</strong>gor even obliteration of the airspaces. Fibr<strong>in</strong> and cell debris areprogressively replaced by collagen fibrils. The ma<strong>in</strong> site offibrosis is the <strong>in</strong>tra-alveolar space, but it also occurs with<strong>in</strong> the<strong>in</strong>terstitium.Fibrotic phaseThis can beg<strong>in</strong> from day 10 after <strong>in</strong>itiat<strong>in</strong>g <strong>in</strong>jury. Macroscopically,the lungs have a cobblestoned character due toscarr<strong>in</strong>g. 2–5The vasculature is grossly deranged with vesselsnarrowed by myo<strong>in</strong>timal thicken<strong>in</strong>g and mural fibrosis. Themicroscopic events occurr<strong>in</strong>g <strong>in</strong> the repair phase are not welldocumented, ma<strong>in</strong>ly because of a paucity of histological dataas patients recover. Some <strong>in</strong>sights have been obta<strong>in</strong>ed frombronchoalveolar lavage (BAL), radiology, and from animalmodels. 24 BAL confirms a marked decl<strong>in</strong>e <strong>in</strong> neutrophils and arelative accumulation of lymphocytes and macrophages. Themost dramatic, although unpredictable, changes are those oflung collagen. 25 Total lung collagen content may double with<strong>in</strong>the first 2 weeks, 26but this burden can be elim<strong>in</strong>ated andmany, albeit small, studies show that survivors can return torelatively normal lung function. 27 Interest<strong>in</strong>gly, although theseverity of pathological changes <strong>in</strong> the first few weeks (hyal<strong>in</strong>emembrane, airspace organisation, or cellularity) do not seemto correlate with late functional recovery, the degree of fibrosisis a key predictor of outcome. 28 29 High levels of procollagenpeptides detected early <strong>in</strong> ARDS have been repeatedly shown30 31to predict a poor outcome. Moreover, established fibrosisreduces lung compliance thereby <strong>in</strong>creas<strong>in</strong>g the work ofbreath<strong>in</strong>g, decreas<strong>in</strong>g the tidal volume, and result<strong>in</strong>g <strong>in</strong> CO 2retention. Also, because of the alveolar obliteration and <strong>in</strong>terstitialthicken<strong>in</strong>g, gas exchange is reduced which contributesto hypoxia and ventilator dependence. Although late deathsfrom ARDS have been ascribed ma<strong>in</strong>ly to sepsis rather thanprogressive hypoxia, sepsis is a common complication <strong>in</strong> thesepatients. It is usually the consequence of ventilator associatedpneumonia or other nosocomial <strong>in</strong>fections related to their32 33ventilator dependence.Recent evidence suggests that there is a much greater overlapof the <strong>in</strong>flammatory and fibroproliferative phases thanpreviously thought, 7 and many mediators are common to bothprocesses. 24 The fibroproliferative response beg<strong>in</strong>s remarkablyearly with N-term<strong>in</strong>al procollagen III peptide levels, a markerfor collagen turnover, be<strong>in</strong>g raised <strong>in</strong> BAL fluid with<strong>in</strong> 24hours of ventilation for ARDS. 30 Myofibroblast cells also showan early <strong>in</strong>crease <strong>in</strong> the alveolar walls, and BAL fluid fromARDS patients with<strong>in</strong> 48 hours of diagnosis is <strong>in</strong>tenselymitogenic for fibroblasts. 31 This all suggests that the fibrosischaracteristic of ARDS may not be a late event but is switchedon at a very early stage. This is particularly important as the<strong>in</strong>flammatory and fibrotic processes, although closely overlapp<strong>in</strong>g,appear to be separately regulated, thus offer<strong>in</strong>g thepossibility for early directed treatments aga<strong>in</strong>st fibrosis<strong>in</strong>dependent of the effects on <strong>in</strong>flammation. 34PATHOGENESISLung <strong>in</strong>jury is <strong>in</strong>itiated by a specific <strong>in</strong>sult but can be exacerbatedby <strong>in</strong>appropriate mechanical ventilatory strategies(reviewed <strong>in</strong> chapter 8). Briefly, alveolar overdistension cangenerate a pro<strong>in</strong>flammatory response which is exacerbated byrepetitive open<strong>in</strong>g and clos<strong>in</strong>g of alveoli as occurs through theuse of <strong>in</strong>appropriately low levels of positive end expiratorypressure (PEEP). 8 Indeed, overdistension or recurrentopen<strong>in</strong>g/clos<strong>in</strong>g of alveoli can also <strong>in</strong>duce structural damageto the lung. 35 36 The effect of high <strong>in</strong>spired concentrations ofoxygen on the disease process is uncerta<strong>in</strong>, particularly <strong>in</strong>humans. However, prolonged exposure to 100% oxygen is fatal<strong>in</strong> most animal models, produc<strong>in</strong>g neutrophil <strong>in</strong>flux andalveolar oedema that can be blocked <strong>in</strong> rodents us<strong>in</strong>ganti-<strong>in</strong>flammatory strategies such as <strong>in</strong>haled low dose carbon37 38monoxide.The ma<strong>in</strong> players <strong>in</strong> the <strong>in</strong>flammatory process are neutrophilsand multiple mediator cascades. 39–41 The fibroblast iskey <strong>in</strong> the fibroproliferative response and is the target of regulatorsof matrix deposition. 42 A complex <strong>in</strong>terplay of regulatorycytok<strong>in</strong>es counteracts the <strong>in</strong>flammatory mediators; similarly,matrix deposition is balanced by the actions of the metalloproteases.There is no uniform response to <strong>in</strong>jury: some patients


40 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>develop ARDS, some ALI, and some do not develop pulmonarysymptoms at all. The reasons for this are not clear but may bepartly genetic. There is evidence for a genetic susceptibility tosepsis and, recently, to ARDS itself. 43–45Because of the difficulties <strong>in</strong> obta<strong>in</strong><strong>in</strong>g histological samples<strong>in</strong> humans, studies have been undertaken <strong>in</strong> animals and haveprovided a valuable understand<strong>in</strong>g of the mechanisms driv<strong>in</strong>glung <strong>in</strong>jury. 46 It is important to be aware of the limitations ofthese models as animals differ <strong>in</strong> their sensitivity to the <strong>in</strong>itiat<strong>in</strong>g<strong>in</strong>sult (especially endotox<strong>in</strong>) and <strong>in</strong> their pulmonaryresponses. The tim<strong>in</strong>g and severity of the <strong>in</strong>sults and,especially, the degree of subsequent resuscitation also do notmirror the cl<strong>in</strong>ical condition. Models <strong>in</strong>clude direct challengeto the lung such as bleomyc<strong>in</strong>, endotox<strong>in</strong> or acid aspiration,surfactant washout and oxygen toxicity, or <strong>in</strong>travenouschallenges <strong>in</strong>clud<strong>in</strong>g endotox<strong>in</strong>, complement or microemboli:all have features <strong>in</strong> common with the human counterpart<strong>in</strong>clud<strong>in</strong>g <strong>in</strong>flammatory cell <strong>in</strong>flux and endothelial38 47 48damage. Animals challenged with endotox<strong>in</strong> developendothelial and epithelial barrier dysfunction although theendothelium appears to be more sensitive than the epithelium,whereas <strong>in</strong> humans it is the epithelium that showsgreater damage. 49Animal models have also been used toexam<strong>in</strong>e the later stages of ARDS. The best are studies <strong>in</strong> primatesexposed to high concentrations of oxygen whichshowed enlarged airspaces, thickened alveolar walls and<strong>in</strong>terstitial fibrosis, and <strong>in</strong> the survivors there was decreasedalveolarisation and bronchopulmonary dysplasia. 38INFLAMMATIONWith <strong>in</strong>itiation of <strong>in</strong>flammation there is <strong>in</strong>creased leucocyteproduction and rapid recruitment to the <strong>in</strong>flamed site. There isalso activation of mediator cascades <strong>in</strong>clud<strong>in</strong>g the productionof cytok<strong>in</strong>es, chemok<strong>in</strong>es, acute phase prote<strong>in</strong>s, free radicals,complement, coagulation pathway components, and focalupregulation of adhesion molecule expression. The “anti<strong>in</strong>flammatory”response <strong>in</strong>cludes the glucocorticoids, cytok<strong>in</strong>es(<strong>in</strong>terleuk<strong>in</strong> (IL)-4, IL-10 and IL-1 receptor antagonist(IL-1ra)) and other mechanisms such as shedd<strong>in</strong>g of adhesionmolecules. 39–41The neutrophilThis is the dom<strong>in</strong>ant leucocyte found both <strong>in</strong> BAL fluid and <strong>in</strong>histological specimens from patients with ARDS. 20In manyanimal models the degree of lung <strong>in</strong>jury is reduceddramatically if neutrophil <strong>in</strong>flux is ablated, although this is50 51not a universal f<strong>in</strong>d<strong>in</strong>g. However, ARDS can develop <strong>in</strong>neutropenic patients, hence neutrophils are believed to be animportant but not essential component of the <strong>in</strong>juriousresponse. 52 Neutrophils cause cell damage though the productionof free radicals, <strong>in</strong>flammatory mediators, and proteases.Excessive quantities of neutrophil products <strong>in</strong>clud<strong>in</strong>g elastase,collagenase, reactive oxygen species, and cytok<strong>in</strong>es such astumour necrosis factor α (TNF-α) have been found <strong>in</strong> patientswith ARDS. 20 41 53–56 Recent studies with mice deficient <strong>in</strong> neutrophilelastase suggest that this enzyme may be an importantmediator of damage to the alveolar epithelium and theprogression to fibrosis. 57Adhesion molecules, notably β 2<strong>in</strong>tegr<strong>in</strong>s, mediate neutrophilb<strong>in</strong>d<strong>in</strong>g to the pulmonary endothelium. This process isbelieved to promote leucocyte <strong>in</strong>duced lung <strong>in</strong>jury althoughsome <strong>in</strong>vestigators suggest that adhesion molecules have adim<strong>in</strong>ished role <strong>in</strong> the pulmonary compared with the systemic58 59circulation. Adhesion molecules also modulate activationand mediator release by neutrophils. In a landmark studyFolkesson and Matthay <strong>in</strong>duced pulmonary <strong>in</strong>jury <strong>in</strong> thepresence of β 2<strong>in</strong>tegr<strong>in</strong> block<strong>in</strong>g antibodies and demonstratedthat, while neutrophil migration still occurred, a number of<strong>in</strong>dices of <strong>in</strong>flammation were reduced. 60 Neutrophil activationleads to cytoskeletal changes that reduce cell deformabilityand slow their transit time through the lung capillary bed,provid<strong>in</strong>g an <strong>in</strong>tegr<strong>in</strong> <strong>in</strong>dependent mechanism whereby neutrophilcontact with the pulmonary endothelium is<strong>in</strong>creased. 61 Other <strong>in</strong>flammatory cells <strong>in</strong>clud<strong>in</strong>g macrophagesand, later, lymphocytes are <strong>in</strong>volved, while platelets may exacerbatethe vascular <strong>in</strong>jury and endothelial cells themselves arecapable of produc<strong>in</strong>g many damag<strong>in</strong>g mediators of <strong>in</strong>flammation.Inflammatory mediatorsThe <strong>in</strong>flammatory process is driven <strong>in</strong> part by cytok<strong>in</strong>es<strong>in</strong>clud<strong>in</strong>g TNF-α and IL-1β, IL-6, and IL-8. All have been56 62 63found <strong>in</strong> BAL fluid and plasma of patients with ARDS.TNF-α and IL-1β can both produce an ARDS-like conditionwhen adm<strong>in</strong>istered to rodents. They are produced by<strong>in</strong>flammatory cells and can promote neutrophil-endothelialadhesion, microvascular leakage, and amplify other pro<strong>in</strong>flammatoryresponses. Despite their profile <strong>in</strong> the septicresponse, the importance of these cytok<strong>in</strong>es <strong>in</strong> the pathogenesisof ARDS is unclear. Levels of TNF-α are not uniformly<strong>in</strong>creased <strong>in</strong> patients with lung <strong>in</strong>jury and anti-TNF-α andIL-1 therapies have been disappo<strong>in</strong>t<strong>in</strong>g. The <strong>in</strong>crease <strong>in</strong> TNF-αlevels occurs very early <strong>in</strong> the cl<strong>in</strong>ical course and may bemissed by the time of presentation, although anti-TNF-αtherapies can still be of benefit <strong>in</strong> some cases of sepsis. 64 Thehuge redundancy <strong>in</strong> the pro<strong>in</strong>flammatory mediator systemssuggests that the search for a “common pathway” susceptibleto <strong>in</strong>hibition may be too simplistic. 39 Many pro<strong>in</strong>flammatorymediators <strong>in</strong>clud<strong>in</strong>g endotox<strong>in</strong>, pro<strong>in</strong>flammatory cytok<strong>in</strong>es,vascular endothelial growth factor (VEGF), high mobilitygroup-1 prote<strong>in</strong>, and thromb<strong>in</strong> are implicated <strong>in</strong> the <strong>in</strong>creasedvascular permeability that contributes to oedema <strong>in</strong> lung<strong>in</strong>jury. 65The balance of “anti-<strong>in</strong>flammatory” cytok<strong>in</strong>es andmediators must also be considered. There is now good experimentalevidence that mediators such as IL-1ra, soluble TNFreceptors (sTNF-R), IL-4, IL-10, and even other moleculessuch as carbon monoxide at very low concentrations are powerfuldownregulators of the <strong>in</strong>flammatory response.37 64The <strong>in</strong>tense neutrophilic <strong>in</strong>filtrate has led to a search for thechemotactic factors responsible. Early studies implicatedcomplement; however, more recently, the focus has been onchemok<strong>in</strong>es. IL-8 levels are raised <strong>in</strong> the BAL fluid of patientsat risk who ultimately develop ARDS. 63It is a powerfulneutrophil chemoattractant derived from alveolar macrophagesand other cells that is regulated by hypoxia/hyperoxia. 66Another neutrophil chemoattractant, ENA-78,may account for IL-8 <strong>in</strong>dependent neutrophil adhesion <strong>in</strong>ARDS. MIF, a neuropeptide, is <strong>in</strong>creased <strong>in</strong> ARDS but its roleis unclear.Animal model work suggests that free radicals arefundamental to the tissue damage result<strong>in</strong>g from pro<strong>in</strong>flammatorystimuli and that antioxidants, <strong>in</strong>clud<strong>in</strong>g glutathioneand superoxide dismutase, are important protective mechanisms.Similarly, <strong>in</strong> humans oxidant stress is <strong>in</strong>creased andplasma antioxidant levels are reduced <strong>in</strong> patients withARDS. 55 67 Nitric oxide may play a role <strong>in</strong> septic lung <strong>in</strong>jury asnitrotyros<strong>in</strong>e, a product derived from peroxynitrite, is found <strong>in</strong><strong>in</strong>creased amounts <strong>in</strong> patients with ARDS. 68 The lipid mediatorplatelet activat<strong>in</strong>g factor (PAF) can activate both neutrophilsand platelets and adm<strong>in</strong>istration can mimic manyfeatures of lung <strong>in</strong>jury. Other mechanisms for the generationof barrier dysfunction dur<strong>in</strong>g the <strong>in</strong>flammatory phase <strong>in</strong>cludepathological changes <strong>in</strong> the regulation of apoptosis. In thisregard soluble Fas ligand (sFasL) has been shown to drivealveolar epithelial cell apoptosis <strong>in</strong> vitro, and to cause lung<strong>in</strong>jury with <strong>in</strong>creased airway cell apoptosis <strong>in</strong> vivo, to bereleased <strong>in</strong> the airspaces of and be localised to the lungepithelial cells <strong>in</strong> patients with ARDS. 69 In addition to excessapoptosis <strong>in</strong> the parenchymal cells, delayed apoptosis <strong>in</strong> the<strong>in</strong>filtrat<strong>in</strong>g neutrophils may contribute to the pro<strong>in</strong>flammatoryload. 70


The pathogenesis of ALI/ARDS 41Mediators of pulmonary hypertensionMild pulmonary arterial hypertension is frequently seen <strong>in</strong>patients with ARDS, and loss of normal control overpulmonary vasomotor tone is an important mechanismunderly<strong>in</strong>g refractory hypoxaemia. Hypoxic pulmonary vasoconstrictionis lost <strong>in</strong> sepsis <strong>in</strong>duced lung <strong>in</strong>jury as <strong>in</strong>halationof 100% oxygen before and dur<strong>in</strong>g endotox<strong>in</strong> challenge doesnot prevent pulmonary hypertension and hypoxic vasoconstrictionis <strong>in</strong>hibited for several hours after endotox<strong>in</strong>challenge. 71 Studies with knockout mice have demonstrated acentral role for nitric oxide (NO) <strong>in</strong> the normal modulation ofpulmonary vascular tone. The mechanisms whereby thisregulation is lost <strong>in</strong> lung <strong>in</strong>jury are not clear, but it is knownthat endotox<strong>in</strong> <strong>in</strong>duces COX-2 and <strong>in</strong>ducible nitric oxide synthase(iNOS) expression <strong>in</strong> the pulmonary vasculature. 72 Thesituation is complicated, however, as endotox<strong>in</strong> contributes toan early marked pulmonary hypertension despite the <strong>in</strong>ductionof iNOS and irrespective of the pulmonary arteryocclusion pressure or cardiac output. 73 Increased expression ofthe powerful vasoconstrictor endothel<strong>in</strong>-1 is associated withpulmonary hypertension <strong>in</strong> sepsis and ARDS. 74 ThromboxaneB 2, another pulmonary vasoconstrictor, may also be an importantmediator of pulmonary hypertension <strong>in</strong> ARDS as COX<strong>in</strong>hibitors reduce the early pulmonary hypertension <strong>in</strong>ducedby endotox<strong>in</strong>. Other pulmonary vasoconstrictors may also bereleased <strong>in</strong> ARDS and other mechanisms of pulmonary hypertensionsuch as microthromboembolism probably contribute.Inflammation leads to a procoagulant state and dissem<strong>in</strong>ated<strong>in</strong>travascular coagulation which is well recognised <strong>in</strong> ARDSand sepsis. Thromb<strong>in</strong> can also potentiate <strong>in</strong>flammation andlead to endothelial barrier dysfunction, <strong>in</strong> addition to itsprofibrotic effects (see below).Surfactant dysfunctionInflammation leads to surfactant dysfunction <strong>in</strong> ARDS. 75 Surfactantis secreted ma<strong>in</strong>ly by alveolar type II cells and consistsof phospholipids (predom<strong>in</strong>antly phosphatidylchol<strong>in</strong>e) andsurfactant specific prote<strong>in</strong>s, SP-A, SP-B, SP-C and SP-D. 76 Theability of surfactant to lower surface tension is criticallydependent on both the phospholipid and the prote<strong>in</strong> components,especially the hydrophobic prote<strong>in</strong>s SP-B and SP-C. Thephospholipids are stored <strong>in</strong> the lamellar bodies of type II cellsand <strong>in</strong>teract with surfactant prote<strong>in</strong>s upon release from thecells, form<strong>in</strong>g large aggregates called tubular myel<strong>in</strong>. Dur<strong>in</strong>gthe normal cycle of breath<strong>in</strong>g these functional large surfactantaggregates become dissipated, reduc<strong>in</strong>g to smalleraggregates which do not have the same surface tension lower<strong>in</strong>gproperties. Type II cells take up these small aggregates andrecycle them <strong>in</strong>to new surfactant. The hydrophilic surfactantprote<strong>in</strong> SP-A, quantitatively the major surfactant prote<strong>in</strong>,plays a key role <strong>in</strong> this process. The other hydrophilicsurfactant prote<strong>in</strong> is SP-D, which may also have a function <strong>in</strong>phospholipid recycl<strong>in</strong>g. SP-A and SP-D are members of thecollect<strong>in</strong> family and form part of the <strong>in</strong>nate immune system <strong>in</strong>the lung; <strong>in</strong>terest<strong>in</strong>gly, both have significant antibacterialactivity and <strong>in</strong>hibit neutrophil apoptosis. 77Surfactant levels are dramatically decreased <strong>in</strong> the <strong>in</strong>fantrespiratory distress syndrome due to immaturity of the type IIcells. By contrast, <strong>in</strong> ARDS surfactant deficiency is not aprimary causal event; rather, the <strong>in</strong>flammatory processes leadto surfactant dysfunction as a secondary factor. Damage andloss of type II cells leads to decreased synthesis and recirculationof surfactant. This defect <strong>in</strong> turnover can lead to accumulationof small aggregates, while overall surfactant performancefollows a reduction <strong>in</strong> the functional large surfactantaggregates and damage to surfactant prote<strong>in</strong>s. 78 In addition,the prote<strong>in</strong> rich oedema <strong>in</strong> ARDS “contam<strong>in</strong>ates” surfactant,further reduc<strong>in</strong>g its functional capacity. Furthermore, <strong>in</strong> lung<strong>in</strong>jury the ratio of m<strong>in</strong>or phospholipids to phosphatidylchol<strong>in</strong>e<strong>in</strong>creases, possibly <strong>in</strong>dicat<strong>in</strong>g damage and release of cellmembrane lipids. The degree to which surfactant dysfunctioncontributes to the pathogenesis of ARDS is currently not clear.THE FIBROPROLIFERATIVE RESPONSEFibroproliferation is a stereotypical part of the normal repairprocess which, if not closely regulated, can have seriousconsequences. The fibrotic response is fuelled by mediatorsthat stimulate local fibroblasts to migrate, replicate, andproduce excessive connective tissue. 72442 Animal models havesuggested a number of potential profibrotic factors. For example,the expression of TNF-α <strong>in</strong> the lung, us<strong>in</strong>g an SP-Cpromoter for tissue specificity, led to the development of a Tlymphocyte predom<strong>in</strong>ant alveolitis which progressed steadilyto a histological picture resembl<strong>in</strong>g idiopathic pulmonaryfibrosis. 79 These data suggest that TNF-α and other pro<strong>in</strong>flammatorymediators, <strong>in</strong>clud<strong>in</strong>g IL-1β, play important roles <strong>in</strong> thedevelopment of pulmonary fibrosis. Similarly, expression oftransform<strong>in</strong>g growth factor α (TGF-α) <strong>in</strong> the distal pulmonaryepithelium <strong>in</strong>duced pulmonary fibrosis, the extent ofwhich is related to the level of gene expression. 46The Th2cytok<strong>in</strong>es have been implicated <strong>in</strong> fibroproliferative disordersand both IL-4 and IL-13 are <strong>in</strong>creased <strong>in</strong> a bleomyc<strong>in</strong> model offibrosis; <strong>in</strong>terest<strong>in</strong>gly, <strong>in</strong>hibition of IL-13 significantly abrogatedthis fibrotic response. 80 Although many mediators regulatecollagen metabolism <strong>in</strong> pulmonary fibrosis, studies ofthese <strong>in</strong> ARDS are very limited. There is evidence that the levelsof TGF-α and a platelet derived growth factor (PDGF)-like81 82factor are <strong>in</strong>creased <strong>in</strong> BAL fluid from patients with ARDS.A number of products of the coagulation cascade—particularly fibr<strong>in</strong>, thromb<strong>in</strong>, and factor Xa—are importantmediators of the pulmonary profibrotic response and are<strong>in</strong>creased <strong>in</strong> patients with ARDS. 24The archetypal profibrotic cytok<strong>in</strong>e is TGF-β, of which thereare three closely related isoforms (TGF-β1, 2 and 3) that exertnearly identical effects as modulators of <strong>in</strong>flammation, <strong>in</strong>hibitorsof growth and differentiation, and regulators of extracellularmatrix production. 83 Studies <strong>in</strong> animals and <strong>in</strong> humansstrongly suggest that TGF-β is important <strong>in</strong> the pathogenesisof pulmonary fibrosis. 84 It has been shown be mitogenic andchemotactic for fibroblasts, to <strong>in</strong>crease the synthesis of extracellularmatrix prote<strong>in</strong>s, and to <strong>in</strong>hibit the production ofmatrix degrad<strong>in</strong>g enzymes. 24TGF-β1 should not be simplyviewed as a profibrotic cytok<strong>in</strong>e, however, as it has multipleother actions—it is anti-<strong>in</strong>flammatory, decreases epithelialproliferation, and can be pro-apoptotic for many cell types. Itis also a powerful chemotactic agent for monocytes and macrophages,essential cells <strong>in</strong> the process of wound heal<strong>in</strong>g. Inthe normal repair process the secretion of TGF-β1 is thus apowerful effector for resolution and it is only when its expressionis persistent or excessive that it leads to pathologicalfibrosis. TGF-β1 is produced as a latent precursor that is converted<strong>in</strong>to the mature bioactive form after cleavage of theN-term<strong>in</strong>al portion, termed the latency associated peptide(LAP). In animal models the transfection of active (but notlatent) TGF-β1 results <strong>in</strong> severe fibrosis. 85 More recently it hasbeen shown that the <strong>in</strong>tegr<strong>in</strong> αvβ6, an adhesion molecule thatb<strong>in</strong>ds matrix and anchors cells, can b<strong>in</strong>d LAP and activateTGF-β1. 86 The expression of this <strong>in</strong>tegr<strong>in</strong> is very low or absenton most normal adult epithelial cells but is upregulateddramatically after <strong>in</strong>jury. 46Failure to express this adhesionmolecule confers almost complete protection aga<strong>in</strong>st bleomyc<strong>in</strong><strong>in</strong>duced lung <strong>in</strong>jury and fibrosis <strong>in</strong> mice. This highlightsthe role of the alveolar epithelium, <strong>in</strong>tegr<strong>in</strong>s, and TGF-β1 <strong>in</strong>the regulation of the <strong>in</strong>flammatory and repair processes.RESOLUTION OF ARDSLiquid is gradually cleared from airspaces by ion pumps thattransport sodium with osmotically driven water movementacross the alveolar epithelium via membrane water


42 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>channels. 87Catecholam<strong>in</strong>es may <strong>in</strong>crease this while propranololor amiloride <strong>in</strong>hibit sodium transport and impedeclearance of alveolar fluid. Repair of the <strong>in</strong>jured lung requiresan <strong>in</strong>tact epithelial basal lam<strong>in</strong>a which facilitates restorationof the epithelial barrier and may thus further promoteclearance of oedema. 88 Lung lymph flow, a much ignored subject,may also contribute to the clearance of pulmonaryoedema.Histologically, the resolution phase of ARDS has been theleast clearly documented. Apoptosis is essential to theclearance of neutrophils and has been clearly demonstrated <strong>in</strong>the lung <strong>in</strong> patients with ARDS. 70 89 Apoptosis is also responsiblefor remov<strong>in</strong>g surplus alveolar type II cells while survivorsdifferentiate <strong>in</strong>to a type I phenotype. BAL fluid recovered frompatients with ALI dur<strong>in</strong>g the repair phase can <strong>in</strong>ducefibroblast and endothelial cell death. This provides a mechanismto clear excess cells while reta<strong>in</strong><strong>in</strong>g underly<strong>in</strong>g normallung structure. Resolution of ARDS requires more than theclearance of leucocytes; for successful heal<strong>in</strong>g the fibroproliferativeresponse must be term<strong>in</strong>ated and excess mesenchymalcells cleared. As the <strong>in</strong>flammatory and fibroproliferative processesare <strong>in</strong>timately l<strong>in</strong>ked and many mediators such asthromb<strong>in</strong> and IL-1 are central to both, it is likely that the resolutionof <strong>in</strong>flammation makes a major contribution to theresolution of fibroproliferation. 34 Other mediators will be producedthat promote resolution—for example, <strong>in</strong>terferon(IFN)γ produced by activated T cells downregulates the transcriptionof the TGF-β1 gene. The generation of pulmonaryfibrosis <strong>in</strong>volves a complex <strong>in</strong>terplay between collagen depositionand degradation, with the early balance shifted dramaticallytowards deposition. In ARDS type III collagen is <strong>in</strong>itiallydeposited which is more flexible and susceptible to breakdown;later this is remodelled to the thicker and more resistanttype I collagen. 26The mechanisms <strong>in</strong>volved <strong>in</strong> theclearance of the fibrotic matrix are not well established but arelikely to <strong>in</strong>volve matrix metalloproteases (MMPs) and gelat<strong>in</strong>asesthat digest collagens. At least two of these—MMP-2 andMMP-9—are <strong>in</strong>creased <strong>in</strong> the lungs of patients with ARDS. 90The MMPs are further regulated by tissue <strong>in</strong>hibitors of metalloproteases(TIMPs), although the relative balance of thesesystems dur<strong>in</strong>g ARDS is unknown.The pulmonary fibrosis <strong>in</strong> ARDS does not necessarily completelyresolve and can lead to persist<strong>in</strong>g pulmonary problemsboth dur<strong>in</strong>g wean<strong>in</strong>g and after patients leave the <strong>in</strong>tensivecare unit. Recovery of lung function can be slow, with somepatients tak<strong>in</strong>g up to 12 months to return to basel<strong>in</strong>e whileothers have persist<strong>in</strong>g abnormalities. In the majority, however,lung mechanics fully recover suggest<strong>in</strong>g that the pulmonaryfibrosis <strong>in</strong> ARDS is reversible. 227PATHOPHYSIOLOGY RELATED TO PATHOLOGYRefractory hypoxaemia may be multifactorial, but <strong>in</strong>trapulmonaryshunt<strong>in</strong>g with ventilation-perfusion mismatch<strong>in</strong>g isbelieved to be the primary cause. Studies on patients withARDS have shown that the degree of <strong>in</strong>trapulmonaryshunt<strong>in</strong>g is sufficient to account for the entire alveolar-arterialoxygen gradient, suggest<strong>in</strong>g that a decrease <strong>in</strong> transfer factormay be of secondary importance. 2Microscopic studies haveshown that the alveolar airspaces are filled with oedema,debris, hyal<strong>in</strong>e membranes, and matrix or lost through alveolarcollapse, creat<strong>in</strong>g a huge physiological deadspace. Theeffects of this on gas exchange are maximised by loss ofhypoxic pulmonary vasoconstriction and by the widespreadpatchy vascular defects so common <strong>in</strong> ARDS. Pulmonaryoedema will also lead to a significant diffusion block. Thecauses of pulmonary oedema are multiple and <strong>in</strong>clude mediator<strong>in</strong>duced vascular permeability, <strong>in</strong>creased pulmonary pressures,and alterations to the oncotic pressure as suggested <strong>in</strong>the sem<strong>in</strong>al studies by Guyton <strong>in</strong> 1959. 91• There is much greater overlap of the <strong>in</strong>flammatory and fibroproliferativephases of ARDS than previously imag<strong>in</strong>ed, withthe fibroproliferative response beg<strong>in</strong>n<strong>in</strong>g with<strong>in</strong> 24 hoursand its severity be<strong>in</strong>g directly related to the outcome.• An exaggerated <strong>in</strong>flammatory response underlies the pathogenesisof ARDS. The neutrophil is the dom<strong>in</strong>ant leucocyte,while many pro<strong>in</strong>flammatory agents <strong>in</strong>clud<strong>in</strong>g endotox<strong>in</strong>,pro<strong>in</strong>flammatory and chemotactic cytok<strong>in</strong>es, vascular endothelialgrowth factor, high mobility group-1 prote<strong>in</strong> andthromb<strong>in</strong>, together with oxidant stress, are all implicated <strong>in</strong>vascular leakage and lung damage.• Surfactant deficiency is not a primary causal event <strong>in</strong> ARDS;rather, the <strong>in</strong>flammatory processes lead to surfactantdysfunction as a secondary factor.• Alveolar type II cell proliferation and enhanced fibroproliferationare key steps <strong>in</strong> attempts at repair <strong>in</strong> the lung; bothoffer potential targets for future therapies.Lung compliance is markedly decreased <strong>in</strong> ARDS. This willbe related <strong>in</strong>itially to flood<strong>in</strong>g of the alveoli and the <strong>in</strong>terstitialspaces with fluid <strong>in</strong>flammatory cells and debris. Progressively,this <strong>in</strong>flammatory response is organised and replaced bymatrix that further reduces compliance. The effects of this onthe pressure-volume curve and on mechanical ventilation willbe reviewed <strong>in</strong> greater detail <strong>in</strong> chapter 9.CONCLUSIONSUnderstand<strong>in</strong>g the pathogenesis of ARDS is essential both tochoos<strong>in</strong>g effective management strategies and to look<strong>in</strong>g fornovel treatments. We have seen that mechanical ventilationcan <strong>in</strong>duce <strong>in</strong>flammation if applied <strong>in</strong>appropriately or reducemortality if correctly used, and that this has a clearpathophysiological rationale. Similar concepts have led toongo<strong>in</strong>g studies of prone ventilation and to the <strong>in</strong>vestigationof other ventilatory strategies such as high frequency ventilationand lung rest with extracorporeal membrane oxygenation.This chapter has outl<strong>in</strong>ed the key mediators <strong>in</strong>volved <strong>in</strong>both the <strong>in</strong>flammatory and fibroproliferative responses andhighlighted some of the important mechanisms throughwhich these responses are regulated. Many of these mediatorswill be exam<strong>in</strong>ed <strong>in</strong> more detail <strong>in</strong> other chapters <strong>in</strong> this bookthat deal with treatment options.Funded by the Medical Research Council (MRC) UK.REFERENCES1 Bernard GR, Artigas A, Brigham KL, et al. The American-EuropeanConsensus Conference on ARDS. Def<strong>in</strong>itions, mechanisms, relevantoutcomes, and cl<strong>in</strong>ical trial co-ord<strong>in</strong>ation. Am J Respir Crit <strong>Care</strong> Med1994;149:818–24.2 Griffiths MJD, Evans TW. Adult respiratory distress syndrome. In: BrewisRAL, Corr<strong>in</strong> B, Geddes DM, Gibson GJ, eds. <strong>Respiratory</strong> medic<strong>in</strong>e. 2nded. Philadelphia: WB Saunders, 1995: 605–29.3 Luce JM. Acute lung <strong>in</strong>jury and the acute respiratory distress syndrome.Crit <strong>Care</strong> Med 1998;26:369–76.4 Deby-Dupont G, Lamy M. Pathophysiology of acute respiratory distresssyndrome and acute lung <strong>in</strong>jury. In: Webb A, Shapiro MJ, S<strong>in</strong>ger M,Suter PM, eds. Oxford textbook of critical care. Oxford: OxfordUniversity Press, 1999: 55–9.5 Tomashefski JF Jr. Pulmonary pathology of the adult respiratory distresssyndrome. Cl<strong>in</strong> Chest Med 1990;11:593–619.6 Hudson LD, Milberg JA, Anardi D, et al. Cl<strong>in</strong>ical risks for developmentof the acute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong> Med1995;151:293–301.7 Marshall R, Bell<strong>in</strong>gan G, Laurent G. The acute respiratory distresssyndrome: fibrosis <strong>in</strong> the fast lane. Thorax 1998;53:815–7.8 Ranieri VM, Suter PM, Tortorella C, et al. Effect of mechanicalventilation on <strong>in</strong>flammatory mediators <strong>in</strong> patients with acute respiratorydistress syndrome: a randomised controlled trial. JAMA1999;282:54–61.9 Milberg JA, Davis DR, Ste<strong>in</strong>berg KP, et al. Improved survival of patientswith acute respiratory distress syndrome (ARDS):1983–1993. JAMA1995;273:306–9.10 Abel SJ, F<strong>in</strong>ney SJ, Brett SJ, et al. Reduced mortality <strong>in</strong> association withthe acute respiratory distress syndrome (ARDS). Thorax 1998;53:292–4.


The pathogenesis of ALI/ARDS 4311 Bion J. Pathophysiology of acute respiratory distress syndrome and acutelung <strong>in</strong>jury. In: Webb A, Shapiro MJ, S<strong>in</strong>ger M, Suter PM, eds. Oxfordtextbook of critical care. Oxford: Oxford University Press, 1999: 923–6.12 Bone RC, Balk RA, Cerra FB, et al. Def<strong>in</strong>itions for sepsis and organfailure and guidel<strong>in</strong>es for the use of <strong>in</strong>novative therapies <strong>in</strong> sepsis. TheAmerican College of Chest Physicians/Society of <strong>Critical</strong> <strong>Care</strong> Medic<strong>in</strong>e(ACCP/SCCM) Consensus Conference Committee. Chest1992;101:1644–55.13 Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit <strong>Care</strong> Med1996;24:1125–8.14 Docke WD, Randow F, Syrbe U, et al. Monocyte deactivation <strong>in</strong> septicpatients: restoration by IFN-gamma treatment. Nature Med1997;3:678–81.15 Desai SR, Wells AU, Suntharal<strong>in</strong>gam G, et al. Acute respiratory distresssyndrome caused by pulmonary and extrapulmonary <strong>in</strong>jury: acomparative CT study. Radiology 2001;218:689–93.16 Hoelz C, Negri EM, Lichtenfels AJ, et al. Morphometric differences ofpulmonary lesions <strong>in</strong> primary and secondary ARDS: a prelim<strong>in</strong>ary study<strong>in</strong> autopsies. Am J Respir Crit <strong>Care</strong> Med 2001;163:450.17 F<strong>in</strong>ney SJ, Robb JD, Suntharal<strong>in</strong>ngam G, et al. ARDS due to direct and<strong>in</strong>direct pulmonary <strong>in</strong>juries: ICU survival, broncho-alveolar lavageneutrophil counts and oxygenation. Am J Respir Crit <strong>Care</strong> Med2001;163:139.18 Sadikot RT, Hao C, Jansen D, et al. Biolum<strong>in</strong>escent measurement ofmultiorgan Cox-2 gene expression <strong>in</strong> transgenic mice treated with LPS.Am J Respir Crit <strong>Care</strong> Med 2001;163:208.19 Bachofen M, Wiebel ER. Structural alteration of lung parenchyma <strong>in</strong> theadult respiratory distress syndrome. Cl<strong>in</strong> Chest Med 1982;3:35–6.20 Weiland JE, Davis WB, Holter JF, et al. Lung neutrophils <strong>in</strong> the adultrespiratory distress syndrome. Cl<strong>in</strong>ical and pathophysiologic significance.Am Rev Respir Dis 1986;133:218–25.21 Matsubara O, Tamura A, Ohdama S, et al. Alveolar basementmembrane breaks down <strong>in</strong> diffuse alveolar damage: animmunohistochemical study. Pathol Int 1995;45:473–82.22 Anderson WR, Thielen K. Correlative study of adult respiratory distresssyndrome by light, scann<strong>in</strong>g and transmission electron microscopy.Ultrastruct Pathol 1992;l16:615–28.23 Snyder LS, Hertz MI, Peterson MS, et al. Acute lung <strong>in</strong>jury. Pathogenesisof <strong>in</strong>traalveolar fibrosis. J Cl<strong>in</strong> Invest 1991;88:663–73.24 Marshall RP, Bell<strong>in</strong>gan GJ, Laurent GJ. The fibroproliferative response toacute lung <strong>in</strong>jury. In: Bell<strong>in</strong>gan GJ, Laurent GJ, eds. Acute lung <strong>in</strong>jury:from <strong>in</strong>flammation to repair. Amsterdam: IOS Press, 2000: 104–14.25 Zapol WM, Trelstad R, Coffey JW, et al. Pulmonary fibrosis <strong>in</strong> severeacute respiratory failure. Am J Respir Crit <strong>Care</strong> Med 1979;119:547–54.26 Raghu G, Striker LJ, Hudson LD, et al. Extracellular matrix <strong>in</strong> normal andfibrotic human lungs. Am Rev Respir Dis 1985;131:281–9.27 McHugh LG, Milberg JA, Whitcomb ME, et al. Recovery of function <strong>in</strong>survivors of the acute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong>Med 1994;150:90–4.28 Suchyta MR, Elliott CG, Colby T, et al. Open lung biopsy does notcorrelate with pulmonary function after the adult respiratory distresssyndrome. Chest 1991;99:1232–7.29 Mart<strong>in</strong> C, Papazian L, Payan MJ, et al. Pulmonary fibrosis correlateswith outcome <strong>in</strong> adult respiratory distress syndrome. A study <strong>in</strong>mechanically ventilated patients. Chest 1995;107:196–200.30 Chesnutt AN, Matthay MA, Tibayan FA, et al. Early detection of type IIIprocollagen peptide <strong>in</strong> acute lung <strong>in</strong>jury. Pathogenic and prognosticsignificance. Am J Respir Crit <strong>Care</strong> Med 1997;156:840–5.31 Marshall RP, Bell<strong>in</strong>gan GJ, Gunn S, et al. Fibroproliferation occurs early<strong>in</strong> the acute respiratory distress syndrome and impacts on outcome. Am JRespir Crit <strong>Care</strong> Med 2000;162:1783–8.32 Montgomery AB, Stager MA, Carrico CJ, et al. Causes of mortality <strong>in</strong>patients with the adult respiratory distress syndrome. Am Rev Respir Dis1985;132:485–9.33 Bell RC, Coalson JJ, Smith JD, et al. Multiple organ system failure and<strong>in</strong>fection <strong>in</strong> adult respiratory distress syndrome. Ann Intern Med1983;99:293–8.34 Bell<strong>in</strong>gan GJ, Marshall RP, Laurent GJ. Fibrosis <strong>in</strong> ARDS: how close isthe l<strong>in</strong>k between <strong>in</strong>flammation and fibroproliferation? In: V<strong>in</strong>cent JL, ed.Yearbook of <strong>in</strong>tensive care and emergency medic<strong>in</strong>e. Berl<strong>in</strong>: Spr<strong>in</strong>ger,2000: 217–24.35 Acute <strong>Respiratory</strong> Distress Syndrome Network. Ventilation with lowertidal volumes as compared with traditional tidal volumes for acute lung<strong>in</strong>jury and the acute respiratory distress syndrome. N Engl J Med2000;342:1301–8.36 Rouby JJ, Lherm T, Mart<strong>in</strong> de Lassale E, et al. Histologic aspects ofpulmonary barotrauma <strong>in</strong> critically ill patients with acute respiratoryfailure. Intensive <strong>Care</strong> Med 1993;19:383–9.37 Otterbe<strong>in</strong> LE, Mantell LL, Choi AM. Carbon monoxide providesprotection aga<strong>in</strong>st hyperoxic lung <strong>in</strong>jury. Am J Physiol1999;276:L688–94.38 Kapanci Y, Wiebel ER, Kaplan HP, et al. Pathogenesis and reversibilityof the pulmonary lesions of oxygen toxicity <strong>in</strong> monkeys. 2. Ultrastructuraland morphometric studies. Lab Invest 1969;20:101–8.39 Bell<strong>in</strong>gan G. Inflammatory cell activation <strong>in</strong> Sepsis. In: Evans TW,Bennett D Bion JF, et al, eds. Intensive care medic<strong>in</strong>e. Br Med Bull1999;55:12–29.40 Bell<strong>in</strong>gan GJ. Leukocyte: friend or foe? Intensive <strong>Care</strong> Med2000;26:111–8.41 Chollet-Mart<strong>in</strong> S, Jourda<strong>in</strong> B, Gibert C, et al. Interactions betweenneutrophils and cytok<strong>in</strong>es <strong>in</strong> blood and alveolar spaces dur<strong>in</strong>g ARDS. AmJ Respir Crit <strong>Care</strong> Med 1996;154:594–601.42 Coker RK, Laurent GJ. Pulmonary fibrosis: cytok<strong>in</strong>es <strong>in</strong> the balance. EurRespir J 1998;11:1218–21.43 Wax RS, Angus DC. The molecular genetics of sepsis. In: V<strong>in</strong>cent JL, ed.Yearbook of <strong>in</strong>tensive care and emergency medic<strong>in</strong>e. Berl<strong>in</strong>: Spr<strong>in</strong>ger,2000: 3–17.44 Marshall RP, Webb S, Bell<strong>in</strong>gan GJ, et al. Angiotens<strong>in</strong> convert<strong>in</strong>genzyme I/D polymorphism <strong>in</strong> the acute respiratory distress syndrome. AmJ Respir Crit <strong>Care</strong> Med 2000;161:209.45 Marshall RP, Webb S, Brull D, et al. Interleuk<strong>in</strong>-6 (-174G>C)polymorphism predicts outcome <strong>in</strong> acute respiratory failure. Am J RespirCrit <strong>Care</strong> Med 2001;163:305.46 Griffiths M. Transgenic and knockout mouse models of pulmonary<strong>in</strong>flammatory diseases. In: Bell<strong>in</strong>gan GJ, Laurent GJ, eds. Acute lung<strong>in</strong>jury: from <strong>in</strong>flammation to repair. Amsterdam: IOS Press, 2000:93–104.47 Meyrick B, Brigham KL. Acute effects of Eschericia coli endotox<strong>in</strong> on thepulmonary microcirculation of anaesthetised sheep structure:functionrelationships. Lab Invest 1983;48:458–70.48 Adler KB, Callahan LM, Evans JN. Cellular alterations <strong>in</strong> the alveolarwall <strong>in</strong> bleomyc<strong>in</strong>-<strong>in</strong>duced pulmonary fibrosis <strong>in</strong> rats. An ultrastructuralmorphometric study. Am Rev Respir Dis 1986;133:1043–8.49 Wiener-Kronish JP, Albert<strong>in</strong>e KH, Matthay MA. Differential responsesof the endothelial and epithelial barriers of the lung <strong>in</strong> sheep toEsherichia coli endotox<strong>in</strong>. J Cl<strong>in</strong> Invest 1991;88:864–75.50 Hefl<strong>in</strong> AC, Brigham KL. Prevention by granulocyte depletion of <strong>in</strong>creasedlung vascular permeability of sheep follow<strong>in</strong>g endotoxaemia. J Cl<strong>in</strong> Invest1981;68:1253–60.51 W<strong>in</strong>n R, Maunder R, Chi E, et al. Neutrophil depletion does not preventlung edema after endotox<strong>in</strong> <strong>in</strong>fusion <strong>in</strong> goats. J Appl Physiol1987;62:116–21.52 Braude S, Apperley J, Krausz T, et al. Adult respiratory distresssyndrome after allogenic bone-marrow transplantation; evidence for aneutrophil-<strong>in</strong>dependent mechanism. Lancet 1985;i:1239–42.53 Lee CT, Fe<strong>in</strong> AM, Lippmann M, et al. Elastolytic activity <strong>in</strong> pulmonarylavage fluid from patients with adult respiratory-distress syndrome. N EnglJ Med 1981;304:192–6.54 Ward PA, Till GO, Kunkel R, et al. Evidence for the role of hydroxylradical <strong>in</strong> complement and neutrophil dependent tissue <strong>in</strong>jury. J Cl<strong>in</strong>Invest 1983;72:789–801.55 Zhang H, Slutsky AS, V<strong>in</strong>cent JL. Oxygen free radicals <strong>in</strong> ARDS, septicshock and organ dysfunction. Intensive <strong>Care</strong> Med 2000;26:474–6.56 Armstrong L, Millar AB. Relative production of tumour necrosis factoralpha and <strong>in</strong>terleuk<strong>in</strong> 10 <strong>in</strong> adult respiratory distress syndrome. Thorax1997;52:442–6.57 Chua F, Dunsmore SE, Mutsaers SE, et al. Characterisation of theresponse to mice deficient <strong>in</strong> neutrophil elastase or catheps<strong>in</strong> G tobleomyc<strong>in</strong>. Am J Respir Crit <strong>Care</strong> Med 2001;163:231.58 Donnelly SC, Haslett C, Dransfield I, et al. Role of select<strong>in</strong>s <strong>in</strong>development of adult respiratory distress syndrome. Lancet1994;344:215–9.59 Doershuk CM, W<strong>in</strong>n RK, Coxson HO, et al. CD18 dependent and<strong>in</strong>dependent mechanisms of neutrophil emigration <strong>in</strong> pulmonary andsystemic microcirculation <strong>in</strong> rabbits. J Immunol 1990;144:2327–33.60 Folkesson HG, Matthay MA. Inhibition of CD18 or CD11b attenuatesacute lung <strong>in</strong>jury after acid <strong>in</strong>stillation <strong>in</strong> rabbits. J Appl Physiol1997;82:1743–50.61 Worthen GS, Schwab B III, Elson EL, et al. Mechanics of stimulatedneutrophils: cell stiffen<strong>in</strong>g <strong>in</strong>duces retention <strong>in</strong> capillaries. Science1989;245:183–6.62 Pug<strong>in</strong> J, Ricou B, Ste<strong>in</strong>berg KP, et al. Pro-<strong>in</strong>flammatory activity <strong>in</strong>bronchoalveolar lavage fluids from patients with ARDS, a prom<strong>in</strong>ent rolefor <strong>in</strong>terleuk<strong>in</strong>-1. Am J Respir Crit <strong>Care</strong> Med 1996;153:1850–6.63 Donnelly SC, Strieter RM, Kunkel SL, et al. Interleuk<strong>in</strong>-8 anddevelopment of adult respiratory distress syndrome <strong>in</strong> at-risk patientgroups. Lancet 1993;341:643–7.64 Panacek EA. Results of the “MONARCS” phase III cl<strong>in</strong>ical trial of ananti-TNF-α antibody (afelimomab) <strong>in</strong> sepsis. Am J Respir Crit <strong>Care</strong> Med2000;161:318.65 Albert<strong>in</strong>e KH. Histopathology of pulmonary oedema and the acuterespiratory distress syndrome. In: Matthay MA, Ingbar DH, eds.Pulmonary edema. Lung Biology <strong>in</strong> Health and Disease. Volume 116.New York: Marcel Dekker, 1998: 37–84.66 Hirani N, Clay, M, Sri-Pathmanathan, R, et al. A role for acutehypoxia/hyperoxia <strong>in</strong> the pathogenesis of the acute respiratory distresssyndrome – an <strong>in</strong> vivo model. Thorax 1999;54(Suppl 3):A13.67 Metnitz PG, Bartens C, Fischer M, et al. Antioxidant status <strong>in</strong> patientswith acute respiratory distress syndrome. Intensive <strong>Care</strong> Med1999;25:180–5.68 Haddad IY, Pataki G, Hu P, et al. Quantitation of nitrotyros<strong>in</strong>e levels <strong>in</strong>lung sections of patients and animals with acute lung <strong>in</strong>jury. J Cl<strong>in</strong> Invest1994;94:2407–13.69 Matute-Bello G, W<strong>in</strong>n RK, Jonas M, et al. Fas (CD95) <strong>in</strong>duces alveolarepithelial cell apoptosis <strong>in</strong> vivo: implications for acute pulmonary<strong>in</strong>flammation. Am J Pathol 2001;158:153–6170 Matute-Bello G, Liles WC, Radella F, et al. Neutrophil apoptosis <strong>in</strong> theacute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong> Med1997;156:1969–77.71 Robb<strong>in</strong>s IM, Newman JH, Brigham KL. Increased-permeabilitypulmonary edema from sepsis/endotoxemia. In: Matthay MA, IngbarDH, eds. Pulmonary edema. Lung Biology <strong>in</strong> Health and Disease. Volume116. New York: Marcel Dekker, 1998: 203–46.72 Griffiths MJ, Curzen NP, Mitchell JA, et al. In vivo treatment withendotox<strong>in</strong> <strong>in</strong>creases rat pulmonary vascular contractility despite NOS<strong>in</strong>duction. Am J Respir Crit <strong>Care</strong> Med 1997;156:654–8.


44 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>73 Sibbald WJ, Paterson NA, Holliday RL, et al. Pulmonary hypertension <strong>in</strong>sepsis: measurement by the pulmonary artery diastolic wedge pressuregradient and the <strong>in</strong>fluence of passive and active factors. Chest1978;73:583–91.74 Druml W, Steltzer H, Waldhausl W, et al. Endothel<strong>in</strong>-1 <strong>in</strong> adultrespiratory distress syndrome. Am Rev Respir Dis 1993;148:1169–73.75 Baudou<strong>in</strong> SV. Surfactant medication for acute respiratory distresssyndrome. Thorax 1997;52:S9–15.76 Rooney SA, Young SL, Mendelson CR. Molecular and cellularprocess<strong>in</strong>g of lung surfactant. FASEB J 1994;8:957–67.77 Greene KE, Gardai S, Henson PM. SP-A and SP-D <strong>in</strong>hibit spontaneousneutrophil apoptosis via activation of the PI-3 k<strong>in</strong>ase/AKT. Am J RespirCrit <strong>Care</strong> Med 2001;163:23.78 Baker CS, Evans TW, Randle BJ, et al. Damage to surfactant-specificprote<strong>in</strong> <strong>in</strong> acute respiratory distress syndrome. Lancet 1999;353:1232–7.79 Miyazaki Y, Araki K, Ves<strong>in</strong> C, et al. Expression of a tumor necrosisfactor-alpha transgene <strong>in</strong> mur<strong>in</strong>e lung causes lymphocytic and fibros<strong>in</strong>galveolitis. A mouse model of progressive pulmonary fibrosis. J Cl<strong>in</strong> Invest1995;96:250–9.80 Keane MP Belperio JA, Burdick MD, et al. IL-13 is an importantcytok<strong>in</strong>e <strong>in</strong> the pathogenesis of pulmonary fibrosis. Am J Respir Crit <strong>Care</strong>Med 2001;163:43.81 Madtes DK, Rubenfeld G, Klima LD, et al. Elevated transform<strong>in</strong>g growthfactor-alpha levels <strong>in</strong> bronchoalveolar lavage fluid of patients with acuterespiratory distress syndrome. Am J Respir Crit <strong>Care</strong> Med 1998;158:424–30.82 Henke C, Mar<strong>in</strong>eili W, Jessurun J, et al. Macrophage production ofbasic fibroblast growth factor <strong>in</strong> the fibroproliferative disorder of alveolarfibrosis after lung <strong>in</strong>jury. Am J Pathol 1993;143:1189–99.83 Clark DA, Coker R. Transform<strong>in</strong>g growth factor-beta (TGF-beta). Int JBiochem Cell Biol 1998;30:293–8.84 Khalil N, Greenberg AH. The role of TGF-beta <strong>in</strong> pulmonary fibrosis.Ciba Found Symp 1991;157:194–20785 Sime PJ, X<strong>in</strong>g Z, Graham FL, et al. Adenovector-mediated gene transferof active transform<strong>in</strong>g growth factor-beta1 <strong>in</strong>duces prolonged severefibrosis <strong>in</strong> rat lung. J Cl<strong>in</strong> Invest 1997;100:768–76.86 Munger JS, Huang X, Kawakatsu H, et al. The <strong>in</strong>tegr<strong>in</strong> alpha v beta 6b<strong>in</strong>ds and activates latent TGF beta 1: a mechanism for regulat<strong>in</strong>gpulmonary <strong>in</strong>flammation and fibrosis. Cell 1999;96:319–28.87 Folkesson HG, Matthay MA, Hasegawa H, et al. Transcellular watertransport <strong>in</strong> lung alveolar epithelium through mercury sensitive waterchannels Proc Natl Acad Sci USA 1994;91:4970–4.89 Matthay MA, Wiener-Kronish JP. Intact epithelial barrier function iscritical for the resolution of alveolar oedema <strong>in</strong> humans. Am Rev RespirDis 1990;142:1250–7.89 Haslett C. Granulocyte apoptosis and its role <strong>in</strong> the resolution andcontrol of lung <strong>in</strong>flammation. Am J Respir Crit <strong>Care</strong> Med1999;160:S5–11.90 Ricou B, Nicod L, Lacraz S, et al. Matrix metalloprote<strong>in</strong>ases and TIMP <strong>in</strong>acute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong> Med1996;154:346–52.91 Guyton AC, L<strong>in</strong>dsey AW. Effect of elevated left atrial pressure anddecreased plasma prote<strong>in</strong> concentration on the development ofpulmonary oedema. Circ Res 1959;VII:649–57.


7 <strong>Critical</strong> care management of severe acute respiratorysyndromeJ T Granton, S E Lap<strong>in</strong>sky.............................................................................................................................Severe acute respiratory syndrome (SARS) isa viral illness characterised by a syndrome offever and respiratory symptoms that canprogress to respiratory failure and death. Initialreports of a highly contagious atypical pneumoniaorig<strong>in</strong>ated from the Guangdong prov<strong>in</strong>ce ofthe People’s Republic of Ch<strong>in</strong>a <strong>in</strong> November 2002.The condition rema<strong>in</strong>ed isolated to Ch<strong>in</strong>a untilFebruary 2003 when an <strong>in</strong>fected physiciantravelled to Hong Kong. S<strong>in</strong>ce that time thedisease has spread, with 8437 probable cases <strong>in</strong> 32countries and 813 deaths provid<strong>in</strong>g a case fatalityrate of 9.6% (WHO website http://www.who.<strong>in</strong>t/csr/sars/en/). 1 The largest outbreaks have been <strong>in</strong>Ch<strong>in</strong>a, Toronto (Canada), and S<strong>in</strong>gapore. Thisreview describes the current state of knowledgeof SARS, with particular reference to the managementof the critically ill patient and the safety andprotection of the staff <strong>in</strong> <strong>in</strong>tensive care units(ICU). The recommendations are based on thepublished data available at the time of writ<strong>in</strong>g,collaborations between physicians <strong>in</strong> many affectedcentres, recommendations from the WHOand Centres for Disease Control (CDC), and localexperience. Given the novelty of this illness, it is<strong>in</strong>evitable that treatments will evolve. The readershould therefore refer to more current sources toguide patient management.AETIOLOGICAL AGENTThe worldwide cooperative effort to identify theaetiological agent for SARS has been summarisedelsewhere. 1The speed at which the aetiologicalagent was identified is a testimony to the progress<strong>in</strong> advances <strong>in</strong> molecular biology, the automationof molecular methods, and the power of genomic<strong>in</strong>formation available <strong>in</strong> databases accessible onthe Internet. Initial efforts focused on severalpotential viruses—most notably, metapneumovirusesand paramyxovirus. On 21 March 2003 anagent isolated from patients meet<strong>in</strong>g the casedef<strong>in</strong>ition for SARS was found to be capable ofproduc<strong>in</strong>g cytopathic changes <strong>in</strong> a Vero and amur<strong>in</strong>e cell l<strong>in</strong>e from a laboratory <strong>in</strong> Germany,and <strong>in</strong> rhesus monkey renal cell l<strong>in</strong>es <strong>in</strong> a laboratory<strong>in</strong> Hong Kong. Us<strong>in</strong>g electron microscopy,coronavirus-like particles were identified (fig7.1). Subsequent sequenc<strong>in</strong>g us<strong>in</strong>g reversetranscription-polymerase cha<strong>in</strong> reaction (RT-PCR) of the isolated product identified the virusas a novel coronavirus. 23Further study of thisagent and sequenc<strong>in</strong>g its 29 751 base genomicstructure supported the notion that this virus isnot merely a mutation of an exist<strong>in</strong>g stra<strong>in</strong>, butthat it represents a novel <strong>in</strong>fectious agent. 4The SARS coronavirus (SARS-CoV) is an RNAvirus belong<strong>in</strong>g to the family Coronaviridae. Thegenome sequence revealed that SARS-CoV wasnot significantly related to other coronaviruses,<strong>in</strong>clud<strong>in</strong>g two human coronaviruses (HCoV-OC43and HCoV-229E). Marra and coworkers reportedthat the virus did not closely resemble any of thethree previously known groups of coronaviruses. 4Coronaviridae are capable of produc<strong>in</strong>g bothenteric and respiratory disease <strong>in</strong> a variety ofhosts. Indeed, their cellular tropism has beenexploited to develop models of gastroenteritis andhepatitis (G Levy, personal communication). Aglycoprote<strong>in</strong> projection on the surface of the virus(S prote<strong>in</strong>; <strong>in</strong> effect, a cellular anchor) is thoughtto provide the virus with its cellular and hostpreferences. Interest<strong>in</strong>gly (and of some concern),two dist<strong>in</strong>ct genotypes of the SARS-CoV havebeen described. 5The different genotypes werepartitioned between two epidemiologically dist<strong>in</strong>ctepidemics. Disturb<strong>in</strong>gly, they also reportedthat there was a common variant with an am<strong>in</strong>oacid substitution <strong>in</strong> the sequence cod<strong>in</strong>g for the Sprote<strong>in</strong> of the SARS coronavirus. They suggestedthat this substitution may have resulted fromimmunological pressure mounted by the host.Given the purported role of the S prote<strong>in</strong>, changes<strong>in</strong> its structure have the potential both to affectthe cellular tropism and, <strong>in</strong> turn, the cl<strong>in</strong>ical featuresof the illness, and further to complicatefuture vacc<strong>in</strong>e development. Until changes <strong>in</strong> thestructure of the virus divert its attention to othermammalian hosts, SARS is likely to rema<strong>in</strong> a globalhealth concern to humans.CLINICAL FEATURESIn the absence of a rapid early diagnostic assay,case def<strong>in</strong>itions of SARS are based on thepresence of epidemiological risk factors (closecontact with SARS cases or travel to SARS“affected” areas) with a comb<strong>in</strong>ation of fever andrespiratory symptoms, with or without hypoxiaand chest radiographic changes. However, as theSARS epidemic spread, the ability to dist<strong>in</strong>guish itfrom other community acquired pneumoniasbased on such epidemiological clues became<strong>in</strong>creas<strong>in</strong>gly tenuous. Although more recent def<strong>in</strong>itionshave <strong>in</strong>corporated serological assays oridentification of viral RNA to confirm cases (seebelow), at this time SARS must be considered <strong>in</strong>the differential diagnosis of any communityacquired or nosocomial pneumonia. The sensitivityof the WHO def<strong>in</strong>ition of a SARS case has beenshown to be limited when applied as a screen<strong>in</strong>gmethod for <strong>in</strong>dividuals at risk of <strong>in</strong>fection. Us<strong>in</strong>gcl<strong>in</strong>ical criteria and progression of the disease <strong>in</strong>the face of conventional therapy as a gold standard,the current WHO def<strong>in</strong>ition of SARS had aspecificity of 96% and negative predictive value of86%. 6 These f<strong>in</strong>d<strong>in</strong>gs were attributed to theabsence of respiratory symptoms <strong>in</strong> many cases atpresentation and because radiographic featureswere not used <strong>in</strong> the WHO def<strong>in</strong>ition of a case.However, their results are not surpris<strong>in</strong>g as theWHO def<strong>in</strong>ition was not <strong>in</strong>tended as a screen<strong>in</strong>g


46 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Log 10 copies/mL nasopharyngeal aspirate9876543Patient APatient BPatient CPatient DPatient EPatient FPatient GPatient HPatient IPatient JPatient KPatient LPatient MPatient N20 5 10 15 20Time after onset of symptoms (days)Figure 7.2 Sequential quantitative reverse transcriptionpolymerase cha<strong>in</strong> reaction (RT-PCR) for SARS coronavirus <strong>in</strong> thenasopharyngeal secretions of 14 patients reported by Peiris andcoworkers. A peak <strong>in</strong> the viral loads was seen on the 10th day ofsampl<strong>in</strong>g. Reproduced with permission from Ksiazek et al. 2 [AQ:3]Figure 7.1 Electron micrographs of the SARS coronavirus. (A) Th<strong>in</strong>section electron micrograph of viral nucleocapsids aligned along themembrane of the rough endoplasmic reticulum (arrow) as particlesbud <strong>in</strong>to the cisternae. (B) A methylam<strong>in</strong>e tungstate penetratedcoronavirus particle with an <strong>in</strong>ternal helical nucleocapsid-likestructure and club shaped surface projections surround<strong>in</strong>g theperiphery of the particle. Reproduced with permission from Ksiazeket al. 2tool. Screen<strong>in</strong>g methods should <strong>in</strong>corporate questions surround<strong>in</strong>gpotential exposure, travel history, and an evaluationof the presence of fever, both respiratory and non-respiratorycompla<strong>in</strong>ts such as malaise, headache, fatigue, and abdom<strong>in</strong>alsymptoms. The importance of proper screen<strong>in</strong>g, a rational andaggressive local health policy, sweep<strong>in</strong>g quarant<strong>in</strong>es, and<strong>in</strong>fection control measures <strong>in</strong> the management of theepidemic cannot be overemphasised. 7–10It is through theseefforts that affected regions have been able to conta<strong>in</strong> thespread of this disease. In the absence of specific treatment forSARS, such measures are (and will rema<strong>in</strong>) the cornerstone ofglobal therapy. The current case def<strong>in</strong>itions can be found atthe websites for the Centre for Disease Control (http://www.cdc.gov/ncidod/sars/) and WHO (http://www.who.<strong>in</strong>t/csr/sars/en/).DIAGNOSTIC TESTINGAt present, specific test<strong>in</strong>g for SARS-CoV <strong>in</strong>fection has limitationsthat prohibit def<strong>in</strong>itive early confirmation of disease.Diagnostic tests for the SARS virus <strong>in</strong>clude (a) detection ofantibodies produced <strong>in</strong> response to SARS <strong>in</strong>fection; (b)molecular tests—for example, PCR to detect genetic materialof the SARS-CoV <strong>in</strong> blood, stool or respiratory secretions; and(c) cell culture techniques which allow the identification oflive virus. Immunofluorescence assays become positive later <strong>in</strong>the illness, but may be useful to confirm undifferentiatedcases or to assist with epidemiological studies. IgG seroconversionoccurs <strong>in</strong> most patients by day 20. 11 Positive antibody testresults, seroconversion, or a fourfold rise <strong>in</strong> titre <strong>in</strong>dicate previous<strong>in</strong>fection with the SARS virus. A negative antibody testresult more than 21 days after the onset of illness is likely toexclude <strong>in</strong>fection with SARS-CoV. At the time of writ<strong>in</strong>g, antibodytest<strong>in</strong>g is unsuitable dur<strong>in</strong>g the acute illness. Clearlythere is a need to develop an assay that will allow the earlierdetection of <strong>in</strong>fected <strong>in</strong>dividuals to facilitate <strong>in</strong>fection control.PCR based assays have been developed by different groupsaround the world and, although such assays are availablecommercially, results of these tests should still not be used toexclude the diagnosis of SARS. Studies evaluat<strong>in</strong>g viral loads<strong>in</strong> respiratory secretions us<strong>in</strong>g quantitative PCR describe an“<strong>in</strong>verted V” pattern with peak viral load occurr<strong>in</strong>g near day10 (fig 7.2). 11 The presence of the <strong>in</strong>fectious virus can be confirmedby <strong>in</strong>oculat<strong>in</strong>g suitable cell cultures (such as Vero cells)with respiratory secretions, blood or stool, and propagat<strong>in</strong>gthe virus <strong>in</strong> vitro. Once isolated, the virus must be identified asSARS-CoV us<strong>in</strong>g further tests performed under biosafety precautionsthat are not rout<strong>in</strong>ely available.Initial diagnostic test<strong>in</strong>g should also <strong>in</strong>clude a search forother respiratory pathogens <strong>in</strong>clud<strong>in</strong>g blood cultures, sputumGram sta<strong>in</strong> and culture, and serological tests. Bronchoscopy isvaluable to exclude other diagnoses but is not recommended<strong>in</strong> patients with a typical cl<strong>in</strong>ical picture and clear epidemiologicall<strong>in</strong>k because of the high risk of transmitt<strong>in</strong>g the <strong>in</strong>fection.In patients who are immunosuppressed and <strong>in</strong> whomconcerns regard<strong>in</strong>g other diagnoses are high, the risk of bronchoscopymay be acceptable. Cl<strong>in</strong>icians should save any availablecl<strong>in</strong>ical specimens (respiratory, blood, and serum) foradditional test<strong>in</strong>g with RT-PCR.Lymphopenia commonly complicates SARS. Additionallaboratory f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> patients with SARS <strong>in</strong>clude thrombocytopenia,leucocytosis, and raised levels of creat<strong>in</strong>e k<strong>in</strong>ase,lactate dehydrogenase (LDH), bilirub<strong>in</strong>, andtransam<strong>in</strong>ases. 12–14A high peak LDH level and an <strong>in</strong>creased


<strong>Critical</strong> care management of severe acute respiratory syndrome 47white cell count at presentation may carry a poor prognosis.Anaemia has been reported <strong>in</strong> up to 60% of patients 14 and maybe secondary to ribavir<strong>in</strong> mediated haemolysis. 12 Haemolysismay also contribute to the hyperbilirub<strong>in</strong>aemia and high LDHlevels seen <strong>in</strong> some patients.NATURAL HISTORYThe reported <strong>in</strong>cubation period of SARS varies from 2 to 7 days(http://www.cdc.gov/mmwr). However, based on the <strong>in</strong>fectiouscharacteristics of other members of the Coronaviridae, there isconcern that the <strong>in</strong>cubation period may be up to 21 days.Indeed, <strong>in</strong> a recent case <strong>in</strong> Toronto a medical student developedsymptoms beyond the 10 day quarant<strong>in</strong>e period. The meanduration of symptoms to time of admission to hospital is 3–5days and <strong>in</strong> one study was observed to shorten throughout theepidemic. 9The viral load may play a role <strong>in</strong> both thetransmission and severity of subsequent disease. The notion of“super spreaders” has been suggested to describe the occasionalpatient who is associated with spread to large numbers of contacts.At present it is unknown if asymptomatic <strong>in</strong>dividuals arecapable of spread<strong>in</strong>g the disease. The mode of transmission isthought to be droplet spread. Consequently, protective measureshave focused on barrier methods, the <strong>in</strong>tensity of which varieswith the risk of aerosolisation of secretions.To date, several studies have provided valuable <strong>in</strong>formationregard<strong>in</strong>g the natural progression of the disease. 9111516TheHong Kong group has provided an excellent summary of theirprospective evaluation of the temporal progression of the disease<strong>in</strong> a cohort of <strong>in</strong>dividuals who were <strong>in</strong>fected follow<strong>in</strong>gexposure to sewage <strong>in</strong> a local hous<strong>in</strong>g complex, AmoyGardens. 11 A three phase illness was described.The first stage was characterised by <strong>in</strong>fluenza-like symptomssuch as fever, myalgia and headache. Our current understand<strong>in</strong>gis that fever eventually occurs <strong>in</strong> all patients and isoften the present<strong>in</strong>g symptom. However, we also recognisethat fever may occasionally be absent on presentation <strong>in</strong> theelderly. 17 Patients often have mild respiratory symptoms at theonset, and gastro<strong>in</strong>test<strong>in</strong>al manifestations are relativelyuncommon <strong>in</strong>itial features. Patients may deffervesce at theend of this phase. Peiris et al correlated this first phase with an<strong>in</strong>crease <strong>in</strong> viral load and ascribed the symptoms to direct viral<strong>in</strong>fection and cytolysis. 11 The chest radiograph is abnormal atpresentation <strong>in</strong> up to 70% of patients, 18 19 featur<strong>in</strong>g patchy orfocal airspace disease with a predilection for the periphery andbases of the lungs (fig 7.3). Half of the opacities are <strong>in</strong>itiallyunilateral. In addition to a progression of the focal opacitiesdur<strong>in</strong>g the course of the illness, the opacities are also oftenmigratory (fig 7.3). Spontaneous pneumomediast<strong>in</strong>um has11 19been reported <strong>in</strong> some patients. The studies that have<strong>in</strong>cluded CT evaluation reveal a pattern that shares remarkablesimilarities with cryptogenic organis<strong>in</strong>g pneumonia orbronchiolitis obliterans organis<strong>in</strong>g pneumonia (COP/18 20 21BOOP). CT changes may be present <strong>in</strong> the absence ofdetectable abnormalities on chest radiographs (fig 7.4).Ground glass opacification and the peripheral distribution ofthe opacities on the CT scan are characteristic. 21Pleuraleffusions are notably absent.Recurrence of fever occurs <strong>in</strong> up to 85% of patients after 5–7days and heralds the onset of the second phase. Dur<strong>in</strong>g thisphase respiratory symptoms are often <strong>in</strong>tensified withworsen<strong>in</strong>g breathlessness and dry cough. 11 In one cohort 73%of patients developed diarrhoea a week <strong>in</strong>to their illness. Diarrhoeamay pose special risks for healthcare providers as thevirus can probably spread by the faecal-oral route. Theobserved decrease <strong>in</strong> viral load dur<strong>in</strong>g this phase supports thenotion that this phase may be related to the immunologicalresponse by the host. In some cases these symptoms arefollowed by hypoxaemia and progression <strong>in</strong> the severity anddistribution of the pulmonary <strong>in</strong>filtrates.Figure 7.3 Progression <strong>in</strong> appearances on the chest radiograph ofa patient with SARS. (A) Radiograph taken on presentation to theemergency department with fever and cough. Ill def<strong>in</strong>ed airspacefill<strong>in</strong>g disease is seen <strong>in</strong> the left base (arrow). (B) 48 hours afteradmission the patient developed progressive hypoxaemia andworsen<strong>in</strong>g of the chest radiograph with <strong>in</strong>volvement of both lungs.(C) 72 hours after admission the patient was mechanically ventilatedvia an endotracheal tube. This chest radiograph taken 96 hours afteradmission demonstrates bilateral patchy airspace disease consistentwith the acute respiratory distress syndrome (ARDS). Evidence ofextrapulmonary air is also seen (arrow).


48 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Figure 7.4 Computed tomographic images of two patients withSARS. The first images (A and B) were taken of the patient described<strong>in</strong> fig 7.3 on the same day as the first chest radiograph (fig 7.3A).Peripheral ground glass opacities (arrow) were seen on multiplesections affect<strong>in</strong>g both lung fields. The second patient (C and D) alsoshows the peripheral opacities and ground glass pattern thatcharacterise SARS.Figure 7.5 (A) Necroscopic lung specimen show<strong>in</strong>g early(“exudative phase”) diffuse alveolar damage with vascularcongestion and airspaces (alveoli/alveolar ducts) l<strong>in</strong>ed by fibr<strong>in</strong>ousfluid exudate (“hyal<strong>in</strong>e membranes”, arrow). These changes arenon-specific with respect to the cause of <strong>in</strong>jury (H&E; orig<strong>in</strong>almagnification ×200). (B) Necroscopic lung specimen show<strong>in</strong>g moreadvanced and more severe acute lung <strong>in</strong>jury reflected by fibr<strong>in</strong>ousexudates <strong>in</strong> alveoli undergo<strong>in</strong>g early organisation with <strong>in</strong>growth ofmesenchymal cells. This pattern of acute lung <strong>in</strong>jury has been called“acute fibr<strong>in</strong>ous and organis<strong>in</strong>g pneumonia”(AFOP). 27 This is nonspecificwith respect to cause and implies that the process will evolve<strong>in</strong>to a more classical picture of organis<strong>in</strong>g pneumonia (formerlycalled BOOP: H&E; orig<strong>in</strong>al magnification ×200). Photograph courtesyof Dr Dean Chamberla<strong>in</strong>.<strong>Respiratory</strong> failureAbout 20–25% of patients with SARS become critically ill andrequire ICU admission. 22 23 The mean duration from the onset ofillness to ICU admission is 5–10 days and respiratory failure isthe usual <strong>in</strong>dication. A recent report from the S<strong>in</strong>gapore groupfound that 45 of 199 <strong>in</strong>fected patients eventually fulfilled thecriteria for the diagnosis of acute lung <strong>in</strong>jury (PaO 2/FiO 2


<strong>Critical</strong> care management of severe acute respiratory syndrome 49Table 7.1High risk procedures for transmission of SARS <strong>in</strong> the ICUProcedure Concern Possible solutionNasopharyngeal swabs Cough<strong>in</strong>g Use nasal swabsBag-valve-mask ventilation Difficult to seal at face Limit as much as possibleTracheal <strong>in</strong>tubation Cough<strong>in</strong>g, agitation Sedation and neuromuscular blockade, PAPRhoodsBronchoscopy Cough<strong>in</strong>g, aerosolisation Limit use of PAPR hoods, neuromuscular blockadeSuction<strong>in</strong>g Cough<strong>in</strong>g, aerosolisation M<strong>in</strong>imise, <strong>in</strong>-l<strong>in</strong>e suctionNon-<strong>in</strong>vasive ventilation Unfiltered aerosolised exhalation ProhibitedHigh frequency oscillation Unfiltered exhalation, uncontrolled secretions Avoid at present until an <strong>in</strong>-l<strong>in</strong>e filter is developedPAPR=powered air purification respirators.disease there was a significantly lower rate of mortality (0 v10.4%, p=0.04) and mechanical ventilation (0 v 12.2%, p=0.03)<strong>in</strong> the group treated with Lop<strong>in</strong>avir compared with historicalcontrols (n=690; http://www.who.<strong>in</strong>t/csr/sars/conference/june_2003/). Lop<strong>in</strong>avir may also have had benefit as rescuetherapy when used with pulsed corticosteroids.Anecdotal evidence suggests that corticosteroids showsome benefit, particularly <strong>in</strong> patients with progressive pulmonary<strong>in</strong>filtrates and hypoxaemia. Various methylprednisoloneregimens have been used at different centres with doses rang<strong>in</strong>gfrom 40 mg twice daily (similar to treatment ofPneumocystis pneumonia) to 2 mg/kg/day (similar to treatment28 29of late phase ARDS), to pulsed doses of 500 mg IV daily.Physicians must search for other causes for fever and <strong>in</strong>fectionas secondary nosocomial <strong>in</strong>fections may be responsible for thecl<strong>in</strong>ical progression seen <strong>in</strong> some critically ill patients later <strong>in</strong>their illness. Indeed, early reports <strong>in</strong> coronaviral models ofhepatitis suggest that the use of corticosteroids may worsenoutcome. 30 31 The use and correct tim<strong>in</strong>g of steroids <strong>in</strong> patientswith SARS may be important and needs to be prospectivelyevaluated.More recently the use of <strong>in</strong>terferon beta has beenadvocated. The rationale for its use stems from the concernthat the later phases of the illness are driven by the host’simmune response. Anecdotal reports on the use of <strong>in</strong>terferonappear promis<strong>in</strong>g and a cl<strong>in</strong>ical study is currently underway.Plasmapheresis as rescue therapy has also been used <strong>in</strong> somecentres with uncerta<strong>in</strong> benefit (http://www.who.<strong>in</strong>t/csr/sars/conference/june_2003/).SURVIVALThe case fatality rate varies from 3% to 12%, depend<strong>in</strong>g onwhether the denom<strong>in</strong>ator <strong>in</strong>cludes suspected and probable11 12 15 16 32cases (3%) or probable cases alone (12%). Olderpatients or those with pre-exist<strong>in</strong>g disease (diabetes, cardiacdisease) have a higher mortality rate. 1 12–13 16 22 23 33 In one studythe mortality rate of patients over 60 years was 43% comparedwith 13.2% <strong>in</strong> their younger counterparts. 9 While themortality rate is higher <strong>in</strong> older patients, particularly thosewith pre-exist<strong>in</strong>g co-morbidity, young previously healthypeople succumb to SARS, possibly because of higher viralloads or their vigorous host response. In multivariate analysisage, LDH level, and an <strong>in</strong>creased neutrophil count at presentationwere associated with a higher odds ratio for a pooroutcome (death or ICU admission). Children appear to have amuch less severe course than adults. 32 For patients admitted tothe ICU, the mortality rates at 28 days have been reported to be34–37%. In the series reported by Fowler and coworkers, 23 ICUoutcome was worse <strong>in</strong> patients over 65 years, <strong>in</strong> diabetics, and<strong>in</strong> those with a higher heart rate and creat<strong>in</strong>e k<strong>in</strong>ase level onadmission. Lew and colleagues 22 correlated a delayed recovery(requir<strong>in</strong>g ventilation beyond 14 days) or death with both theAPACHE II score (odds ratio for each 1 unit <strong>in</strong>crease of 0.87, CI0.78 to 0.97) and PaO 2/FiO 2ratio (odds ratio for each 1 unit<strong>in</strong>crease of 1.02, CI 1.0 to 1.04) on admission to the ICU. TheFigure 7.6 Photograph of a tra<strong>in</strong><strong>in</strong>g session of healthcare workersperform<strong>in</strong>g an <strong>in</strong>tubation on a patient simulator system. Photographcourtesy of Dr Randy Wax.majority of deaths (75%) occurred late <strong>in</strong> the course of the ICUadmission from ARDS, multisystem organ failure, thrombosis,or septic shock.MANAGEMENT OF RESPIRATORY FAILUREThe risk of droplet spread is <strong>in</strong>creased by various procedures(table 7.1). Efforts to avoid viral spread <strong>in</strong>clude the avoidanceof nebulisers for drug adm<strong>in</strong>istration and limitation or avoidanceof the use of non-<strong>in</strong>vasive ventilation. Nebulised humidificationfor oxygen therapy may carry similar risks, and ourpractice is to provide non-humidified oxygen us<strong>in</strong>g nasalprongs or a Venturi mask. A non-rebreather mask withexpiratory port allow<strong>in</strong>g for gas filtration is available and maybe of value. Dur<strong>in</strong>g bag-mask-valve ventilation a filter shouldbe used on the expiratory port.Tracheal <strong>in</strong>tubation poses a special risk to healthcareproviders. Indeed, several of the outbreaks among healthcareworkers <strong>in</strong> Toronto occurred follow<strong>in</strong>g <strong>in</strong>tubation of patientswith SARS. As a result, strict guidel<strong>in</strong>es for <strong>in</strong>tubation and themanagement of cardiac arrest have been developed. 34 Recommendationsfor general anaesthesia have also beenpublished. 35In all <strong>in</strong>stances, when a patient with known orsuspected SARS requires tracheal <strong>in</strong>tubation, perfect executionof <strong>in</strong>fection control measures and donn<strong>in</strong>g of protectiveequipment is required (fig 7.6). In Toronto mock cardiacarrests and patient simulators with relevant cl<strong>in</strong>ical scenarioshave been used. Intubation should be performed by the mostskilled person available us<strong>in</strong>g the method with which they aremost comfortable. Awake <strong>in</strong>tubation may be associated withagitation and cough<strong>in</strong>g which can severely compromise <strong>in</strong>fectioncontrol precautions. We therefore recommend rapidsequence <strong>in</strong>duction to facilitate airway stabilisation. Apowered air purification respirator (PAPR) such as 3MAirmate can be used for these procedures.Designated mechanical ventilators should have two filters(for example, Conserve 50 PALL filters) placed to elim<strong>in</strong>ate the


50 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>exhalation of viral particles <strong>in</strong>to the environment and to protectthe <strong>in</strong>side of the ventilator from contam<strong>in</strong>ation. One filtershould be <strong>in</strong>terposed between the distal end of the expiratorytub<strong>in</strong>g and the ventilator itself, and the second should beplaced on the exhalation outlet of the ventilator. Ideally, theexhalation port should then be connected to a centralscaveng<strong>in</strong>g system that would elim<strong>in</strong>ate release of viral particles<strong>in</strong>to the ICU. High frequency oscillation may be associatedwith <strong>in</strong>creased risk of droplet spread and exposure to respiratorysecretions, and our practice is to avoid this <strong>in</strong>tervention <strong>in</strong>patients with SARS. High frequency jet ventilation for thosefail<strong>in</strong>g conventional ventilation may be used safely (ow<strong>in</strong>g tothe ability to place <strong>in</strong>-l<strong>in</strong>e filters).If a ventilated patient desaturates requir<strong>in</strong>g manualbag-valve-mask ventilation, it is important to turn theventilator to standby before disconnection to avoid dropletspray. In fact, <strong>in</strong> an <strong>in</strong>tubated patient with SARS werecommend avoid<strong>in</strong>g this <strong>in</strong>tervention unless there is an obviousmechanical ventilator failure, even <strong>in</strong> the event of acardiac arrest. Similarly, tracheobronchial suction<strong>in</strong>g ofpatients should be m<strong>in</strong>imised and an <strong>in</strong>-l<strong>in</strong>e suction cathetersystem should be used. Because of the effect of <strong>in</strong>fection controlmeasures on bedside care, the use of k<strong>in</strong>etic beds may beconsidered for patients who are unable to move withoutassistance.The severity of ARDS complicat<strong>in</strong>g SARS has beens<strong>in</strong>gularly impressive. Aside from the advanced <strong>in</strong>fection controlmeasures, the ventilatory management of these patients isno different from others with ARDS. We therefore use a lowtidal volume strategy to m<strong>in</strong>imise the risk of ventilator associatedlung <strong>in</strong>jury and adopt an open lung approach us<strong>in</strong>grecruitment manoeuvres and higher levels of PEEP toma<strong>in</strong>ta<strong>in</strong> alveolar patency. The use of rout<strong>in</strong>e recruitmentmanoeuvres is offset by the desire to m<strong>in</strong>imise exposure torespiratory therapists and nurses. Little experience exists withthe use of <strong>in</strong>terventions such as prone position<strong>in</strong>g or <strong>in</strong>halednitric oxide <strong>in</strong> patients with SARS. In the report by Lew et al 22three of seven patients had a significant improvement <strong>in</strong> oxygenationwith prone position<strong>in</strong>g. Anecdotally, the experience<strong>in</strong> Toronto and S<strong>in</strong>gapore has been that nitric oxide offers littlebenefit.The development of air leaks appears to be high <strong>in</strong> patientswith SARS related ARDS, rang<strong>in</strong>g between 20% and 34%.Eleven episodes of venous thromboembolism and sevenepisodes of proven or suspected pulmonary embolism werereported by Lew and colleagues, 22although the mode ofprophylaxis was not stated.INFECTION CONTROL PRECAUTIONSInitial unfamiliarity and, later <strong>in</strong> the SARS outbreak, the failureto adhere to <strong>in</strong>fection control procedures resulted <strong>in</strong> thespread of SARS to many healthcare personnel. 23 The organismappears to be transmitted by droplet spread, although surfacecontam<strong>in</strong>ation and possibly airborne spread may play a role.Recent data suggest that the virus may rema<strong>in</strong> viable for considerableperiods (up to 24 hours) on a dry surface. 36As aresult, staff education and cont<strong>in</strong>ued vigilance are essential.Infection of healthcare workers <strong>in</strong>volved <strong>in</strong> high riskprocedures is a very real threat. Despite the use of <strong>in</strong>fectioncontrol precautions, n<strong>in</strong>e healthcare workers developed SARSfollow<strong>in</strong>g a prolonged <strong>in</strong>tubation procedure, eight of whomrequired hospitalisation. 36Droplet aerosolisation also occursdur<strong>in</strong>g bronchoscopy and similar precautions should beemployed, <strong>in</strong>clud<strong>in</strong>g sedation and paralysis to prevent cough<strong>in</strong>g.Some recommendations for ICU staff have been publishedand are shown <strong>in</strong> box 7.1. 34 To avoid repeatedly break<strong>in</strong>g thenegative pressure barrier, <strong>in</strong>dividual rooms should be stockedwith basic supplies and modified cardiac arrest carts conta<strong>in</strong><strong>in</strong>gemergency drugs should be available <strong>in</strong> the room. StaffBox 7.1 Infection control precautions <strong>in</strong> the ICUStaff education• High risk procedures, alternatives and precautions.• Ways of m<strong>in</strong>imis<strong>in</strong>g exposure and effective use of timewhen <strong>in</strong> the room.• Instructions to staff on how to “undress” and “re-dress”without contam<strong>in</strong>ation.• Importance of vigilance and adherence to all <strong>in</strong>fection controlprecautions.• Importance of monitor<strong>in</strong>g own health.• Information on SARS as it evolves.Dress precautions• Airborne precautions us<strong>in</strong>g an N-95 mask or equivalent.• Contact precautions.• Eye protection with a non-reusable goggles or face shield.• Pens, paper, other personal items should not be allowed<strong>in</strong>to or removed from the room.• Powered air purification respirator (PAPR) hoods should beused dur<strong>in</strong>g high risk procedures.Environment/equipment• Negative pressure isolation rooms with antechambers, anddoors closed at all times.• Individual isolation rooms stocked with basic supplies andemergency drugs.• Alcohol based hand and equipment dis<strong>in</strong>fectants.• Gloves, gowns, masks and disposal units readily available.• Use of video camera equipment or w<strong>in</strong>dows to monitorpatients.• <strong>Care</strong>ful and frequent clean<strong>in</strong>g of surfaces with disposableclothes and alcohol based detergents.• No equipment should be shared.Transport• Avoid patient transport where possible.• Reflect on need for <strong>in</strong>vestigations and whether the benefitsjustify the transportation risks.• Intubated patients should have a filter (Conserve PALL 50)<strong>in</strong>serted between the bag-valve and the swivel connector.• Infection control should be alerted.should rema<strong>in</strong> outside the negative pressure rooms as much aspossible. This means tim<strong>in</strong>g venesection and adm<strong>in</strong>istration oftreatment to m<strong>in</strong>imise entries and the use of video cameraequipment or w<strong>in</strong>dows to monitor patients without expos<strong>in</strong>gstaff. An antechamber (preferably with a s<strong>in</strong>k) helps to ma<strong>in</strong>ta<strong>in</strong>strict <strong>in</strong>fection control precautions. Pens, paper, and otherpersonal items should not be allowed <strong>in</strong>to or removed from theroom. Airborne precautions us<strong>in</strong>g a N-95 mask or equivalentshould be taken; it is important that manufacturers’ specificationsare adhered to—for example, some N-95 masksma<strong>in</strong>ta<strong>in</strong> protection for 8 hours and some only for 4 hours.Touch<strong>in</strong>g the mask or lift<strong>in</strong>g it to wipe the face or nose shouldbe avoided. It is crucial to ma<strong>in</strong>ta<strong>in</strong> a close seal to the sk<strong>in</strong> andto ensure a proper fit. Sessions to determ<strong>in</strong>e optimum fit andmask type should be provided to all personnel. In addition,contact precautions—<strong>in</strong>clud<strong>in</strong>g the use of double gowns (atleast one of which is waterproof) and double gloves, hats andshoe covers—should be used. Gowns, gloves, hats, boots,masks, and goggles should be changed after see<strong>in</strong>g eachpatient. Eye protection with non-reusable goggles or a faceshield is required and staff should change <strong>in</strong>to hospital scrubsupon arrival and change <strong>in</strong>to their own cloth<strong>in</strong>g at the end ofthe day to avoid formite spread. Scrubs should not leave thehospital and should be sterilised after each use. Theimportance of proper removal of protective equipment cannotbe overemphasised as contact with droplets on the surface ofmasks and gowns may occur.Transportation of a patient with SARS is an <strong>in</strong>fection controlchallenge that should be avoided whenever possible. SARS


<strong>Critical</strong> care management of severe acute respiratory syndrome 51patients should never be transported while be<strong>in</strong>g supported bybag-valve-mask ventilation and should preferably be <strong>in</strong>tubated.If bag ventilation is used, a filter should be placedbetween the bag and the endotracheal tube. The <strong>in</strong>fectioncontrol department should be consulted for advice on properprecautions.An important component of <strong>in</strong>fection control is the limitationof personnel and visitors hav<strong>in</strong>g contact with the patient.It is crucial that staff do not work if they are ill, even if thediagnosis is not clear. After unprotected contact with a SARSpatient, staff are subject to a compulsory 10 day quarant<strong>in</strong>eperiod. Visitors were restricted <strong>in</strong> Toronto hospitals dur<strong>in</strong>g theoutbreak. Indeed, several of the restrictions rema<strong>in</strong> <strong>in</strong> placeand will cont<strong>in</strong>ue to do so for an <strong>in</strong>def<strong>in</strong>ite period. All visitorsare screened for symptoms of SARS and adhere to the sameprecautions as hospital staff. Visits with SARS patients areprohibited even on compassionate grounds.CONCLUSIONSSARS has resulted <strong>in</strong> significant challenges for critical caremedic<strong>in</strong>e. The ability of this disease to <strong>in</strong>capacitate staff hasresulted <strong>in</strong> staff safety becom<strong>in</strong>g a priority to ma<strong>in</strong>ta<strong>in</strong>adequate critical care services. Indeed, the impact of this<strong>in</strong>fection on the healthcare system and regional economycannot be understated. Resources of <strong>in</strong>dividual hospitalswere rapidly outstripped as scores of adm<strong>in</strong>istrative and frontl<strong>in</strong>e care providers were quarant<strong>in</strong>ed or became ill. In Toronto18% of the critically ill patients were healthcare workers. Theability of this <strong>in</strong>fection to spread is s<strong>in</strong>gularly impressive anddevastat<strong>in</strong>g. Our understand<strong>in</strong>g of the virus, its diagnosis,and treatment cont<strong>in</strong>ues to evolve. Infection control measuresrema<strong>in</strong> the ma<strong>in</strong>stay of regional and global health. Theconcept of “universal precautions” has expanded beyondpolicies regard<strong>in</strong>g blood borne <strong>in</strong>fections and now <strong>in</strong>cludesstrict respiratory and contact precautions. The effect of thesestr<strong>in</strong>gent policies on patients without SARS was devastat<strong>in</strong>g.As a result of the outbreak, hospitals were closed andadvanced surgical and medical care programmes such astransplantation and organ donation were shut down. In one<strong>in</strong>stance 35 critical care beds were closed (represent<strong>in</strong>g 38%of our tertiary ICU beds) because of an <strong>in</strong>advertent exposureof ICU staff to a patient with SARS. The secondary morbidityand mortality from this disease on patients who were placedon hold or whose surgery was delayed rema<strong>in</strong>s to bedeterm<strong>in</strong>ed. The guidel<strong>in</strong>es and recommendations discussedhere will change as our knowledge grows. No doubt<strong>in</strong>formation technology played an important role <strong>in</strong> allow<strong>in</strong>gcollaboration and rapid transfer of <strong>in</strong>formation throughoutthe SARS pandemic. Indeed, through such collaboration wecan hope to improve the mortality and morbidity of ourpatients and, <strong>in</strong>deed, ourselves.REFERENCES1 Anon. Update: severe acute respiratory syndrome—United States, June18, 2003. MMWR 2003;52:570.2 Ksiazek TG, Erdman D, Goldsmith CS, et al. A novel coronavirusassociated with severe acute respiratory syndrome. N Engl J Med2003;348:1953–66.3 Drosten C, Gunther S, Preiser W, et al. Identification of a novelcoronavirus <strong>in</strong> patients with severe acute respiratory syndrome. N Engl JMed 2003;348:1967–76.4 Marra MA, Jones SJ, Astell CR, et al. The genome sequence of theSARS-associated coronavirus. Science 2003;300:1399–404.5 Ruan YJ, Wei CL, Ee AL, et al. Comparative full-length genomesequence analysis of 14 SARS coronavirus isolates and commonmutations associated with putative orig<strong>in</strong>s of <strong>in</strong>fection. Lancet2003;361:1779–85.6 Ra<strong>in</strong>er TH, Cameron PA, Smit D, et al. Evaluation of WHO criteria foridentify<strong>in</strong>g patients with severe acute respiratory syndrome out ofhospital: prospective observational study. BMJ 2003;326:1354–8.7 Yang W. Severe acute respiratory syndrome (SARS): <strong>in</strong>fection control.Lancet 2003;361:1386–7.8 Zhong NS, Zeng GQ. Our Strategies for fight<strong>in</strong>g severe acuterespiratory syndrome (SARS). Am J Respir Crit <strong>Care</strong> Med 2003;168:7–9.9 Donnelly CA, Ghani AC, Leung GM, et al. Epidemiologicaldeterm<strong>in</strong>ants of spread of causal agent of severe acute respiratorysyndrome <strong>in</strong> Hong Kong. Lancet 2003;361:1761–6.10 Pearson H, Clarke T, Abbott A, et al. SARS: what have we learned?Nature 2003;424:121–6.11 Peiris JS, Chu CM, Cheng VC, et al. Cl<strong>in</strong>ical progression and viral load<strong>in</strong> a community outbreak of coronavirus-associated SARS pneumonia: aprospective study. Lancet 2003;361:1767–72.12 Booth CM, Matukas LM, Toml<strong>in</strong>son GA, et al. Cl<strong>in</strong>ical features andshort-term outcomes of 144 patients with SARS <strong>in</strong> the greater Torontoarea. JAMA 2003;289:2801–9.13 Tan YM, Chow PK, Soo KC. Severe acute respiratory syndrome: cl<strong>in</strong>icaloutcome after <strong>in</strong>patient outbreak of SARS <strong>in</strong> S<strong>in</strong>gapore. BMJ2003;326:1394.14 Wong RS, WuA,ToKF,et al. Haematological manifestations <strong>in</strong> patientswith severe acute respiratory syndrome: retrospective analysis. BMJ2003;326:1358–62.15 Avendano M, Derkach P, Swan S. Cl<strong>in</strong>ical course and management ofSARS <strong>in</strong> health care workers <strong>in</strong> Toronto: a case series. Can Med Assoc J2003;168:1649–60.16 Lee N, Hui D, Wu A, et al. A major outbreak of severe acute respiratorysyndrome <strong>in</strong> Hong Kong. N Engl J Med 2003;348:1986–94.17 Fisher DA, Lim TK, Lim YT, et al. Atypical presentations of SARS. Lancet2003;361:1740.18 Gr<strong>in</strong>blat L, Shulman H, Glickman A, et al. Severe acute respiratorysyndrome: radiographic review of 40 probable cases <strong>in</strong> Toronto,Canada. Radiology 2003;228:802–9.19 Wong KT, Antonio GE, Hui DS, et al. Severe acute respiratorysyndrome: radiographic appearances and pattern of progression <strong>in</strong> 138patients. Radiology 2003;228:401–6.20 Wong KT, Antonio GE, Hui DS, et al. Th<strong>in</strong>-section CT of severe acuterespiratory syndrome: evaluation of 74 patients exposed to or with thedisease. Radiology 2003;228:395–400.21 Muller NL, Ooi GC, Khong PL, et al. Severe acute respiratory syndrome:radiographic and CT f<strong>in</strong>d<strong>in</strong>gs. AJR 2003;181:3–8.22 Lew TW, Kwek TK, Tai D, et al. Acute respiratory distress syndrome <strong>in</strong>critically ill patients with severe acute respiratory syndrome. JAMA2003;290:374–80.23 Fowler RA, Lap<strong>in</strong>sky SE, Hallett D, et al. <strong>Critical</strong>ly ill patients with severeacute respiratory syndrome. JAMA 2003;290:367–73.24 Nicholls JM, Poon LL, Lee KC, et al. Lung pathology of fatal severe acuterespiratory syndrome. Lancet 2003;361:1773–8.25 D<strong>in</strong>g Y, Wang H, Shen H, et al. The cl<strong>in</strong>ical pathology of severe acuterespiratory syndrome (SARS): a report from Ch<strong>in</strong>a. J Pathol2003;200:282–9.26 Franks TJ, Chong PY, Chui P, et al. Lung pathology of severe acuterespiratory syndrome (SARS): a study of 8 autopsy cases from S<strong>in</strong>gapore.Hum Pathol 2003;34:729.27 Beasley MB, Franks TJ, Galv<strong>in</strong> JR, et al. Acute fibr<strong>in</strong>ous and organiz<strong>in</strong>gpneumonia: a histological pattern of lung <strong>in</strong>jury and possible variant ofdiffuse alveolar damage. Arch Pathol Lab Med 2002;126:1064–70.28 Gagnon S, Boota AM, Fischl MA, et al. Corticosteroids as adjunctivetherapy for severe Pneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> the acquiredimmunodeficiency syndrome. A double-bl<strong>in</strong>d, placebo-controlled trial. NEngl J Med 1990;323:1444–50.29 Meduri GU, Headley AS, Golden E, et al. Effect of prolongedmethylprednisolone therapy <strong>in</strong> unresolv<strong>in</strong>g acute respiratory distresssyndrome: a randomized controlled trial. JAMA 1998;280:159–65.30 F<strong>in</strong>gerote RJ, Leibowitz JL, Rao YS, et al. Treatment of resistant A/J micewith methylprednisolone (MP) results <strong>in</strong> loss of resistance to mur<strong>in</strong>ehepatitis stra<strong>in</strong> 3 (MHV-3) and <strong>in</strong>duction of macrophage procoagulantactivity (PCA). Adv Exp Med Biol 1995;380:89–94.31 F<strong>in</strong>gerote RJ, Abecassis M, Phillips MJ, et al. Loss of resistance tomur<strong>in</strong>e hepatitis virus stra<strong>in</strong> 3 <strong>in</strong>fection after treatment with corticosteroidsis associated with <strong>in</strong>duction of macrophage procoagulant activity. J Virol1996;70:4275–82.32 Hon KL, Leung CW, Cheng WT, et al. Cl<strong>in</strong>ical presentations andoutcome of severe acute respiratory syndrome <strong>in</strong> children. Lancet2003;361:1701–3.33 Chan JW, Ng CK, Chan YH, et al. Short term outcome and risk factorsfor adverse cl<strong>in</strong>ical outcomes <strong>in</strong> adults with severe acute respiratorysyndrome (SARS). Thorax 2003;58:686–9.34 Lap<strong>in</strong>sky SE, Hawryluck L. ICU management of severe acute respiratorysyndrome. Intensive <strong>Care</strong> Med 2003;29:870–5.35 Kamm<strong>in</strong>g D, Gardam M, Chung F. Anaesthesia and SARS. Br J Anaesth2003;90:715–8.36 Anon. Cluster of severe acute respiratory syndrome cases amongprotected health-care workers—Toronto, Canada, April 2003. MMWR2003;52:433–6.


8 Ventilator <strong>in</strong>duced lung <strong>in</strong>juryT Whitehead, A S Slutsky.............................................................................................................................Mechanical ventilation has become an<strong>in</strong>dispensable tool, facilitat<strong>in</strong>g generalanaesthesia and support<strong>in</strong>g life <strong>in</strong> thecritically ill. However, its application has adverseeffects <strong>in</strong>clud<strong>in</strong>g an <strong>in</strong>creased risk of pneumonia,impaired cardiac performance, and neuromuscularproblems relat<strong>in</strong>g to sedation and musclerelaxants. Moreover, it has become clear thatapply<strong>in</strong>g pressure—whether positive ornegative—to the lung can cause damage knownas ventilator <strong>in</strong>duced lung <strong>in</strong>jury (VILI). Thisconcept is not new. In his treatise on resuscitationof the apparently dead, John Fothergill <strong>in</strong> 1745suggested that mouth to mouth <strong>in</strong>flation of thevictim’s lungs might be preferable to us<strong>in</strong>g a pairof bellows as “the lungs of one man may bear,without <strong>in</strong>jury, as great a force as another mancan exert; which by the bellows cannot always bedeterm<strong>in</strong>’d”. 1 Although not specifically addressed,Forthergill’s admonition aga<strong>in</strong>st the useof the bellows probably related to gross air leaksproduced by large pressures. This type of <strong>in</strong>jury isnow called barotrauma and was the first widelyrecognised manifestation of VILI. The cl<strong>in</strong>ical andradiological manifestations of barotrauma <strong>in</strong>cludepneumothorax, pneumomediast<strong>in</strong>um, andsurgical emphysema.Later, evidence accumulated to suggest thatventilation causes more subtle morphological andfunctional changes and can excite an <strong>in</strong>flammatoryresponse with<strong>in</strong> the lung. This type of <strong>in</strong>jurywas not recognised for many years as the patternof damage is often <strong>in</strong>dist<strong>in</strong>guishable from thatseen <strong>in</strong> other forms of lung <strong>in</strong>jury such as theacute respiratory distress syndrome (ARDS), forwhich mechanical ventilation is an <strong>in</strong>dispensabletreatment. Studies us<strong>in</strong>g animal models werenecessary to def<strong>in</strong>e key aspects of VILI. Based onthese, many cl<strong>in</strong>icians <strong>in</strong> the 1990s began toadopt ventilatory strategies designed to m<strong>in</strong>imiselung <strong>in</strong>jury, although the cl<strong>in</strong>ical importance ofVILI has only recently been established. 2In this chapter we summarise the ma<strong>in</strong> riskfactors for VILI, its possible mechanisms, and itscl<strong>in</strong>ical relevance. Specific ventilation techniquesfor ARDS are addressed <strong>in</strong> chapter 9, and will onlybe alluded to here for the purpose of illustrat<strong>in</strong>ggeneral pr<strong>in</strong>ciples.MAJOR DETERMINANTS OF VILIMost research <strong>in</strong>to VILI is based on positive pressureventilation delivered via an endotrachealtube, although the pr<strong>in</strong>ciples are equally relevantto non-<strong>in</strong>vasive or negative pressure ventilation. Ithas become clear that the degree of VILI is determ<strong>in</strong>edby the <strong>in</strong>teraction of the ventilator sett<strong>in</strong>gsand patient related factors, particularly thecondition of the ventilated lung.Ventilator determ<strong>in</strong>ants of VILIAirway pressure and lung distensionConceptually, it seems obvious that <strong>in</strong>flation ofthe lung will cause damage if airway pressures arehigh enough. The important issues for thecl<strong>in</strong>ician have been (1) what levels of airwaypressure are dangerous and (2) can they beavoided <strong>in</strong> the mechanical ventilation of patientswith stiff lungs, as <strong>in</strong> ARDS?The association between high airway pressureand air leaks has long been recognised. 34However,this relationship does not necessarily implycausality as damaged, stiff lungs that require highairway pressures for ventilation may be <strong>in</strong>tr<strong>in</strong>sicallymore prone to air leaks. Recent large studies<strong>in</strong> patients with ARDS have, <strong>in</strong> fact, shown a poorcorrelation between airway pressure (or tidal volume)and the occurrence of air leaks, whichoccurred <strong>in</strong> 8–14% of the patients. 2 5–7However,these data should not be <strong>in</strong>terpreted as demonstrat<strong>in</strong>gthat the degree of lung distension isunimportant <strong>in</strong> the development of barotrauma.In these studies airway pressures and tidalvolumes were lower than those used <strong>in</strong> the pastwhen barotrauma rates as high as 39% werereported <strong>in</strong> patients ventilated for acute respiratoryfailure. 8More subtle lung damage was first unequivocallyshown by Webb and Tierney who ventilatedrats for 1 hour us<strong>in</strong>g different airway pressureswith and without positive end expiratory pressure(PEEP). 9 Animals ventilated with a peak airwaypressure of 14 cm H 2O had no histologicalchanges <strong>in</strong> the lung, while those ventilated with apressure of 30 cm H 2O had mild perivascularoedema. In contrast, all rats ventilated at45 cm H 2O (without PEEP) developed severehypoxia and died before the end of the hour. Theirlungs had marked perivascular and alveolaroedema. Similar f<strong>in</strong>d<strong>in</strong>gs have been observed <strong>in</strong>other species, although there is considerable variation<strong>in</strong> the susceptibility to VILI. Whereas <strong>in</strong> ratsventilation at high peak <strong>in</strong>spiratory pressure forjust 2 m<strong>in</strong>utes is sufficient to <strong>in</strong>duce pulmonaryoedema, larger animals such as rabbits and sheeprequire much longer periods of ventilation forchanges to be evident. 10–12 This is clearly importantwhen extrapolat<strong>in</strong>g data from animal studies tohumans.Pressure or volume?Although the term barotrauma is commonly usedwhen discuss<strong>in</strong>g VILI, the evidence <strong>in</strong>dicates thatthe degree of lung <strong>in</strong>flation is a more importantdeterm<strong>in</strong>ant of lung <strong>in</strong>jury than airway pressureper se. This may be <strong>in</strong>ferred from the observationthat trumpet players commonly achieve airwaypressures of 150 cm H 2O without develop<strong>in</strong>grepetitive episodes of gross air leakage. 13 The relativecontribution of pressure and volume to lung<strong>in</strong>jury was first studied by ventilat<strong>in</strong>g rats whosetidal excursion was limited by strapp<strong>in</strong>g the chestand abdomen. 14High airway pressure without ahigh tidal volume did not produce lung <strong>in</strong>jury. Bycontrast, animals ventilated without thoracicrestriction us<strong>in</strong>g high tidal volumes, achievedeither with high positive <strong>in</strong>spiratory pressure or


Ventilator <strong>in</strong>duced lung <strong>in</strong>jury 53negative pressure <strong>in</strong> an iron lung, developed severe <strong>in</strong>jury.15 16These results have been confirmed <strong>in</strong> other species.The term volutrauma is therefore more accurate thanbarotrauma, although <strong>in</strong> practice the two are closely related.The degree of alveolar distension is determ<strong>in</strong>ed by thepressure gradient across the alveoli, approximated by thetranspulmonary pressure, the difference between the staticairway pressure (estimated cl<strong>in</strong>ically by the plateau airwaypressure) and the pleural pressure. Peak airway pressure is notnecessarily a reflection of alveolar pressure and is greatly<strong>in</strong>fluenced by the resistance to flow <strong>in</strong> the airways. Recentguidel<strong>in</strong>es have therefore emphasised the importance of limit<strong>in</strong>gplateau pressures and of be<strong>in</strong>g aware of factors that<strong>in</strong>crease (or decrease) the degree of alveolar distension for agiven alveolar pressure. 17 For example, conditions associatedwith <strong>in</strong>creased chest wall compliance such as immaturity<strong>in</strong>crease lung distension, and hence damage, for a givenalveolar pressure. Conversely, chest wall compliance iscommonly reduced by abdom<strong>in</strong>al distension, 18which canresult <strong>in</strong> lower alveolar <strong>in</strong>flation, derecruitment, and hypoxaemiaif the plateau pressure is <strong>in</strong>appropriately low.Lung <strong>in</strong>jury at low lung volumesThere is evidence that lung damage may also be caused byventilation at low lung volume (mean<strong>in</strong>g low absolute lungvolume rather than low tidal volume). This has been welldef<strong>in</strong>ed <strong>in</strong> animal models, but the relevance to humans is notfirmly established. Several studies have shown that the <strong>in</strong>juriouseffects of mechanical ventilation can be attenuated by theapplication of PEEP. 9141920 Ventilation with high tidal volumeand low or zero PEEP appears to be more damag<strong>in</strong>g than lowtidal volume and high PEEP, even though both strategiesresult <strong>in</strong> similar high levels of end <strong>in</strong>spiratory pressure andalveolar distension. Ventilation of isolated lavaged rat lungswith small tidal volumes (5–6 ml/kg) and low or zero PEEPcaused lung <strong>in</strong>jury that could be reduced by the application ofhigher levels of PEEP. 21A number of mechanisms may expla<strong>in</strong> lung <strong>in</strong>juryassociated with ventilation at low absolute lung volumes.Cyclic open<strong>in</strong>g and clos<strong>in</strong>g (recruitment-derecruitment) ofsmall airways/lung units may lead to <strong>in</strong>creased local shearstress—so called atelectrauma. PEEP effectively spl<strong>in</strong>ts open thedistal airways, ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g recruitment throughout theventilatory cycle. The static pressure-volume curve (fig 8.1) isoften used to illustrate the balance between overdistensionand recruitment. As airway pressure is <strong>in</strong>creased fromfunctional residual capacity (FRC), an abrupt change <strong>in</strong> thelung compliance is often evident, particularly <strong>in</strong> <strong>in</strong>jured orsurfactant deficient lungs. This lower <strong>in</strong>flection po<strong>in</strong>t (LIP)may represent the approximate pressure (volume) at whichlung units are recruited. The upper <strong>in</strong>flection po<strong>in</strong>t (UIP) atwhich lung compliance decreases at higher airway pressurewas thought to reflect the po<strong>in</strong>t at which alveoli are becom<strong>in</strong>goverdistended, and therefore potentially damaged. 23 Based onthese concepts, an ideal ventilatory strategy would be one <strong>in</strong>which all the tidal ventilation would take place on the “steep”portion of the pressure-volume curve where the lung is mostcompliant. The value of PEEP would be sufficient to preventderecruitment but not so great as to lead to overdistension.High frequency oscillatory ventilation (HFOV) potentiallyoffers the ideal comb<strong>in</strong>ation of m<strong>in</strong>imum tidal volume whilema<strong>in</strong>ta<strong>in</strong><strong>in</strong>g maximal recruitment (the “open lung”), providedsufficient end expiratory lung volume is ma<strong>in</strong>ta<strong>in</strong>ed.24 25Although theoretically sound, the explanation of VILIaccord<strong>in</strong>g to the pressure-volume curve is certa<strong>in</strong>ly a grossoversimplification. Recruitment is not complete at the LIP andcont<strong>in</strong>ues at higher <strong>in</strong>flat<strong>in</strong>g pressures. 26 27 Similarly, the UIPdoes not necessarily reflect the onset of overdistension.Instead, it may represent the po<strong>in</strong>t at which recruitment iscomplete and therefore compliance decreases. Furthermore,VolumeFRCDeflationFigure 8.1 Pressure-volume curve derived from a patient withARDS. FRC=functional residual capacity; LIP=lower <strong>in</strong>flection po<strong>in</strong>t;UIP=upper <strong>in</strong>flection po<strong>in</strong>t. Adapted from Matamis et al 22 with permission.<strong>in</strong>flation may be expand<strong>in</strong>g alveoli without necessarily overdistend<strong>in</strong>gthem. 27In addition, it is difficult to show thatrecruitment-derecruitment actually occurs, 28and dynamicventilation may not follow the pattern of the static pressurevolumecurve. Indeed, a recent study us<strong>in</strong>g sal<strong>in</strong>e lavaged rabbitssuggested that ventilation follows the deflation limb ofthe pressure-volume curve, provided an adequate recruitmentmanoeuvre is performed. 29 Thus, theoretically, a lung could bema<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> a recruited state at lower airway pressures thanthe <strong>in</strong>flation limb of the pressure-volume curve would <strong>in</strong>dicateand, conversely, pressures applied on the basis of the <strong>in</strong>flationlimb would cause overdistension. F<strong>in</strong>ally, but of perhapsgreatest importance, the damaged lung <strong>in</strong> the cl<strong>in</strong>ical sett<strong>in</strong>gis not homogeneously affected, as detailed below. Applied airwaypressure that may be ideal to recruit and ventilate somelung units may be <strong>in</strong>adequate to open the most densely atelectaticregions, and yet simultaneously cause overdistension30 31<strong>in</strong> the most compliant areas.Other ventilator parametersFew studies have addressed the effect of ventilator parametersother than tidal volume, airway pressure, and PEEP <strong>in</strong> VILI.Increased respiratory frequency may augment lung <strong>in</strong>jurythrough greater stress cycl<strong>in</strong>g, a phenomenon well described<strong>in</strong> eng<strong>in</strong>eer<strong>in</strong>g, or through the deactivation of surfactant. Isolatedperfused rabbit lungs ventilated with a frequency of 20breaths/m<strong>in</strong> show greater oedema and perivascular haemorrhagethan those ventilated at 3 breaths/m<strong>in</strong>. 32 The relevanceof these f<strong>in</strong>d<strong>in</strong>gs to cl<strong>in</strong>ical practice is unclear.Inspired oxygen fractionOxidant stress is believed to be an important mechanism <strong>in</strong>mediat<strong>in</strong>g lung <strong>in</strong>jury <strong>in</strong> a variety of lung diseases <strong>in</strong>clud<strong>in</strong>gARDS, and exposure of animals or humans to a high <strong>in</strong>spiredoxygen fraction (FiO 2) leads to lung damage, probably through33 34the <strong>in</strong>creased generation of reactive oxygen species.Current practice <strong>in</strong> ARDS is to use the lowest FiO 2giv<strong>in</strong>g anoxygen saturation of around 90%.Carbon dioxideArterial carbon dioxide tensions (PaCO 2) reflect m<strong>in</strong>ute ventilation.The advent of protective ventilatory strategies <strong>in</strong> ARDS,with lower tidal volumes and m<strong>in</strong>ute ventilation, was accompaniedby the acceptance of higher levels of PaCO 2(permissivehypercapnia) and mild respiratory acidosis. 35Rather thanbe<strong>in</strong>g harmful, some animal studies suggest that hypercapniaexerts a protective effect <strong>in</strong> lung <strong>in</strong>jury, although to date thereis no clear evidence from cl<strong>in</strong>ical studies. 36Patient determ<strong>in</strong>ants of VILIThe condition of the ventilated lung is of considerable importance<strong>in</strong> determ<strong>in</strong><strong>in</strong>g susceptibility to VILI. At one extreme,LIP00PressureInflationUIP


54 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>portions of the lung are more severely affected (fig 8.2A), adistribution determ<strong>in</strong>ed largely by gravity. The greatercompliance of less affected areas means that they receive amuch greater proportion of the delivered tidal volume. 41 Thismay result <strong>in</strong> substantial regional overdistension and hence<strong>in</strong>jury.In a recent study, piglets with multifocal pneumonia wereventilated us<strong>in</strong>g a tidal volume of 15 ml/kg for 43 hours. 42Approximately 75% of the lung volume was consolidated, sothe residual 25% of normally ventilated lung may havereceived a tidal volume equivalent to 40–50 ml/kg. Histologicalexam<strong>in</strong>ation showed emphysema-like lesions <strong>in</strong> these areas,whereas <strong>in</strong> consolidated areas the alveoli were “protected”aga<strong>in</strong>st overdistension, but the bronchioles that rema<strong>in</strong>edpatent were <strong>in</strong>jured through overdistension and by the forcesgenerated through <strong>in</strong>terdependence and recruitmentderecruitment(fig 8.3). Lesions similar to those describedabove have been reported <strong>in</strong> a necropsy series of patients withARDS 43 ; furthermore, CT scans of ARDS survivors have showngreatest residual abnormality <strong>in</strong> the anterior parts of the lung,even though the posterior areas had been most abnormal <strong>in</strong>the acute phase (fig 8.2B). 44These changes may be due to<strong>in</strong>jury caused by overdistension.Other factors that may promote VILI <strong>in</strong> already <strong>in</strong>juredlungs are surfactant abnormalities and the presence of anactivated <strong>in</strong>flammatory <strong>in</strong>filtrate which may be furtherstimulated by mechanical ventilation.Figure 8.2 (A) CT scan of a 25 year old man with ARDS show<strong>in</strong>gthe typically heterogeneous distribution of opacification with<strong>in</strong> thelungs, mostly <strong>in</strong> the posterior dependent regions. (B) CT scan of thesame patient 8 months later show<strong>in</strong>g remarkably little abnormality <strong>in</strong>the posterior regions but with reticular changes anteriorly (largearrows). Reproduced with permission from Desai et al. 44VILI does not appear to be a cl<strong>in</strong>ical problem <strong>in</strong> patients withnormal lungs who can undergo prolonged periods ofmechanical ventilation without detrimental effect. 37 In these<strong>in</strong>stances the pressures and flows with<strong>in</strong> the lung closelyresemble the physiological situation. At the other extreme, thegrossly abnormal lungs of patients with ARDS are highly susceptibleto VILI, and it may be that <strong>in</strong> some patients nomechanical ventilation strategy is entirely devoid of detrimentaleffects.Ventilat<strong>in</strong>g the <strong>in</strong>jured lungAnimal studies us<strong>in</strong>g isolated lungs and <strong>in</strong>tact animals have<strong>in</strong>dicated that <strong>in</strong>jured lungs are more susceptible to VILI. 38–40An important factor underly<strong>in</strong>g this predisposition to VILI isthe uneven distribution of disease and <strong>in</strong>flation seen <strong>in</strong><strong>in</strong>jured lungs. Based on the diffuse relatively homogeneousdistribution of shadow<strong>in</strong>g on a pla<strong>in</strong> chest radiograph, it wasthought that the lung was uniformly affected <strong>in</strong> ARDS. However,CT scann<strong>in</strong>g showed that the posterior dependentLung immaturityThe immature lung may be particularly susceptible to VILI. 45The volume of the lung relative to body weight and thenumber of alveoli are lower <strong>in</strong> premature <strong>in</strong>fants, mak<strong>in</strong>g atidal volume based on weight potentially more <strong>in</strong>jurious. Theresilience of the lung tissue is lower, due to less well developedcollagen and elast<strong>in</strong> elements, and surfactant deficiency leadsto alveolar <strong>in</strong>stability and favours airway closure. At deliverythe fluid filled, surfactant deficient airways of the pretermneonate require high <strong>in</strong>flation pressures to establish patency,with potential generation of high shear stress. Preterm lambsshow evidence of lung <strong>in</strong>jury after only six high volume<strong>in</strong>sufflations. 46MANIFESTATION OF VILIPulmonary oedemaPulmonary oedema is a prom<strong>in</strong>ent feature of experimentalmodels of VILI, particularly those us<strong>in</strong>g small animals. 91247The high prote<strong>in</strong> content of the oedema fluid suggests that itis, at least <strong>in</strong> part, due to <strong>in</strong>creased permeability, andexperimental studies have implicated changes at both the epithelialand microvascular endothelial barriers. Increased static<strong>in</strong>flation of fluid filled lung lobes <strong>in</strong> sheep led to the passage oflarger solutes across the epithelium, a f<strong>in</strong>d<strong>in</strong>g observed <strong>in</strong>48 49other models. Increased microvascular permeability hasbeen shown by the redistribution of 125 I-labelled album<strong>in</strong> <strong>in</strong>tothe extravascular space <strong>in</strong> mechanically ventilated rats, 50 withFigure 8.3 Histological specimens of lung from (A) control piglets and (B) and (C) piglets with experimental pneumonia follow<strong>in</strong>g ventilationfor 2 days. (A) shows normal lung architecture and bronchioles (arrow 1). In piglets with pneumonia there are emphysematous changes <strong>in</strong> theventilated regions (B), but bronchioles are of normal size (arrow 2). In consolidated areas (C) there is bronchiolar dilatation (arrow 3).Reproduced with permission from Goldste<strong>in</strong> et al. 42


Ventilator <strong>in</strong>duced lung <strong>in</strong>jury 55parallel f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> other species. 16 51 In contrast to the clearevidence for <strong>in</strong>creased permeability, relatively little is knownabout the contribution of hydrostatic pressures to thedevelopment of pulmonary oedema <strong>in</strong> mechanical ventilation.This stems largely from the great difficulty <strong>in</strong> assess<strong>in</strong>g transmuralpressures at the microvascular level. Studies <strong>in</strong> lambshave <strong>in</strong>dicated that high tidal volume ventilation causes arelatively modest <strong>in</strong>crease <strong>in</strong> transmural pulmonary vascularpressures. 16 Hydrostatic forces may still be important. Firstly,regional differences <strong>in</strong> lung perfusion and atelectasis maygenerate far greater filtration forces <strong>in</strong> some areas 52and,secondly, <strong>in</strong> the <strong>in</strong>jured lung even small <strong>in</strong>creases <strong>in</strong> transmuralpressure may greatly <strong>in</strong>crease oedema formation.Morphological changesThe acute structural changes of “<strong>in</strong>jurious” mechanical ventilationhave been best def<strong>in</strong>ed us<strong>in</strong>g small animal models.Under the light microscope the development of oedema is firstevident as perivascular cuff<strong>in</strong>g which progresses to florid<strong>in</strong>terstitial oedema and alveolar flood<strong>in</strong>g with cont<strong>in</strong>ued ventilationat high pressure. 950 Changes <strong>in</strong> the endothelium aredetectable by electron microscopy with<strong>in</strong> only a few m<strong>in</strong>utesof high airway pressure ventilation of rats, and appear to precedealterations <strong>in</strong> the epithelium. Some endothelial cellsbecome focally separated from their basement membraneform<strong>in</strong>g <strong>in</strong>tracapillary blebs. Eventually, diffuse alveolar damageis evident; the epithelial surface becomes grosslydisrupted <strong>in</strong> some areas with destruction of type I but spar<strong>in</strong>g14 47of type II cells.Larger animals have been used to study the longer termeffects of mechanical ventilation. In piglets with experimentalpneumonia, alveolar damage is seen <strong>in</strong> the ventilated regionsafter 2–3 days, with bronchiolar dilatation <strong>in</strong> the consolidatedregions (fig 8.3). 42An <strong>in</strong>flammatory reaction also becomesprom<strong>in</strong>ent. Ventilation of piglets with high peak airway pressuresleads to neutrophil recruitment with<strong>in</strong> 24 hours andfibroproliferative changes after 3–6 days. 53 Similarly, conventionalventilation (tidal volume 12 ml/kg) of sal<strong>in</strong>e lavagedrabbits led to accumulation of neutrophils <strong>in</strong> the lungs, as wellas severe epithelial damage and hyal<strong>in</strong>e membrane formation.By contrast, animals ventilated with HFOV had m<strong>in</strong>imal54 55<strong>in</strong>jury.Increased vascular permeability, diffuse alveolar damage,<strong>in</strong>flammatory cell <strong>in</strong>filtrates, and later fibroproliferativechanges are not specific to VILI. ARDS and other forms of lung<strong>in</strong>jury are associated with identical pathological appearances.The pert<strong>in</strong>ent question for the cl<strong>in</strong>ician is the degree to whichthe changes classically ascribed to ARDS are <strong>in</strong> factattributable to VILI?MECHANISMS OF VILIThe mechanical forces applied through ventilation may havedeleterious effects <strong>in</strong> at least two ways: (1) through physicaldisruption of the tissues and cells, which depends not only onthe magnitude and pattern of the applied stress but also onthe resilience of the lung tissue, and (2) through the aberrantactivation of cellular mechanisms lead<strong>in</strong>g to <strong>in</strong>appropriateand harmful responses. Both certa<strong>in</strong>ly occur, although it is notclear which is more important cl<strong>in</strong>ically.Physical disruption (stress failure)How forces are generated with<strong>in</strong> the lungMackl<strong>in</strong> and Mackl<strong>in</strong> proposed that air leaks are caused by amomentary high pressure gradient between the alveolus andthe bronchovascular sheath. 3 Air ruptures across the epithelialsurface and tracks along the bronchovascular sheath. It maythen pass <strong>in</strong>to the <strong>in</strong>terstitium caus<strong>in</strong>g pulmonary <strong>in</strong>terstitialemphysema, <strong>in</strong>to the pleural space caus<strong>in</strong>g pneumothorax,<strong>in</strong>to the pericardial cavity lead<strong>in</strong>g to pneumopericardium, andso on. The endothelium, <strong>in</strong> close apposition to the epithelialsurface, is subject to stress failure due to forces derived bothfrom transpulmonary and <strong>in</strong>travascular pressures. The extremelyth<strong>in</strong> blood gas barrier (0.2–0.4 µM) permits free gasexchange by diffusion but exposes the capillary to high wallstress, determ<strong>in</strong>ed by the ratio of the wall tension tothickness. 56 In rabbits stress failure occurs at capillarytransmural pressures of 52.5 cm H 2O(∼40 mm Hg) or greaterand the microscopic lesions of endothelial and epithelialdisruption are similar to those caused by high volumeventilation. 56–58Importantly, the blood-gas barrier is moreprone to stress failure at higher lung volumes, probably due to<strong>in</strong>creased longitud<strong>in</strong>al forces act<strong>in</strong>g on the blood vessel. 59 Theforces generated by mechanical ventilation may therefore<strong>in</strong>teract with those due to pulmonary vascular perfusion tomagnify lung <strong>in</strong>jury. Isolated rabbit lungs ventilated with apeak static pressure of 30 cm H 2O exhibit greater oedema andhaemorrhage when perfused with high flow rates correspond<strong>in</strong>gto higher pulmonary artery pressures. 60InterdependenceAdjacent alveoli and term<strong>in</strong>al bronchioles share commonwalls so that forces act<strong>in</strong>g on one lung unit are transmitted tothose around it. This phenomenon, known as <strong>in</strong>terdependence,is believed to be important <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g uniformity ofalveolar size and surfactant function. 61Under conditions ofuniform expansion all lung units will be subject to a similartransalveolar pressure, approximately equal to the alveolarm<strong>in</strong>us the pleural pressure. However, if the lung is unevenlyexpanded, such forces may vary considerably. When an alveoluscollapses the traction forces exerted on its walls byadjacent expanded lung units <strong>in</strong>crease and these are appliedto a smaller area. These forces will promote re-expansion atthe expense of greatly <strong>in</strong>creased and potentially harmfulstress at the <strong>in</strong>terface between collapsed and expanded lungunits. At a transpulmonary pressure of 30 cm H 2O it has beencalculated that re-expansion pressures could reach140 cm H 2O. 61 62 In a necroscopic study of patients who haddied with ARDS, expanded cavities and pseudocysts werefound particularly around atelectatic areas, suggest<strong>in</strong>g thatthese forces do <strong>in</strong>deed play a role <strong>in</strong> VILI. 43Recruitment-derecruitmentTheoretically, small airways may become occluded by exudate63 64or apposition of their walls. In either event, the airwaypressure required to restore patency greatly exceeds that <strong>in</strong> anunoccluded passage. The result<strong>in</strong>g shear stress may damagethe airways, particularly if the cycle is repeated with eachbreath (∼20 000 times per day). The pressure required to reopenan occluded airway is <strong>in</strong>versely proportional to itsdiameter, 65 which is consistent with the observation that smallairway damage <strong>in</strong> isolated lungs ventilated at zero PEEPoccurs more distally as PEEP is applied. 21Airway collapse is favoured by surfactant deficiency or conditions<strong>in</strong> which the <strong>in</strong>terstitial support of the airways is65 66weakened or underdeveloped. Conversely, recruitmentderecruitmentmay not occur at all <strong>in</strong> normal lungs, whichtolerate periods of negative end expiratory pressure withoutevident harm. 67Importance of surfactantSurfactant plays a role <strong>in</strong> VILI <strong>in</strong> two related ways; firstly, surfactantdysfunction or deficiency appears to amplify the <strong>in</strong>juriouseffects of mechanical ventilation and, secondly, ventilationitself can impair surfactant function thereby favour<strong>in</strong>gfurther lung damage.Abnormalities of the surfactant system may contribute to<strong>in</strong>jury <strong>in</strong> the mature lung as <strong>in</strong> preterm <strong>in</strong>fants. Surfactantisolated from patients with ARDS and experimental models ofpneumonia is functionally impaired. 68 69 This is associated witha decrease <strong>in</strong> the functionally active large aggregate (LA)


56 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>component of the surfactant pool relative to the less activesmall aggregate (SA) component. Although stretch of type 2cells <strong>in</strong> vitro and a modest degree of hyperventilationstimulate surfactant production, 70 more <strong>in</strong>jurious ventilatorystrategies of high tidal volume and low PEEP lead to a reducedpool of functional surfactant and a decreased LA:SA ratio,particularly <strong>in</strong> <strong>in</strong>jured lungs. 71 Large cyclical alterations <strong>in</strong> thealveolar surface area and the presence of serum prote<strong>in</strong>s <strong>in</strong> theairspace may be responsible for these changes.Lungs can be rendered experimentally deficient <strong>in</strong> surfactantby sal<strong>in</strong>e lavage or detergent aerosolisation, and theymay behave <strong>in</strong> a manner similar to those of premature <strong>in</strong>fants.Strategies that ma<strong>in</strong>ta<strong>in</strong> lung recruitment, such as HFOV(with recruitment manoeuvres) and conventional mechanicalventilation at high levels of PEEP, appear to be particularlyeffective <strong>in</strong> m<strong>in</strong>imis<strong>in</strong>g lung <strong>in</strong>jury <strong>in</strong> these models. 72–75Surfactant abnormalities may contribute to VILI <strong>in</strong> severalways relat<strong>in</strong>g to the <strong>in</strong>crease <strong>in</strong> surface tension:(1) Alveoli and airways are more prone to collapse withgeneration of shear stress as they are reopened.(2) The uneven expansion of lung units <strong>in</strong>creases regionalstress forces through <strong>in</strong>terdependence.(3) The transvascular filtration pressure is <strong>in</strong>creased, promot<strong>in</strong>goedema formation. 76In addition, surfactant is thought to have important immunoregulatoryfunctions 77 which may become impaired throughmechanical ventilation.From the preced<strong>in</strong>g discussion it is logical to propose that<strong>in</strong>creas<strong>in</strong>g the pool of function<strong>in</strong>g surfactant might lessenlung <strong>in</strong>jury. Surfactant therapy reduces mortality <strong>in</strong> the neonatalrespiratory distress syndrome and may decrease lung<strong>in</strong>jury. 78 In ARDS a role for surfactant supplementation is notestablished, 79partly because of difficulties <strong>in</strong> deliver<strong>in</strong>gadequate amounts of active surfactant to damaged andcollapsed lung regions.Activation of aberrant cellular pathwaysPhysical forces such as stretch play an important role <strong>in</strong>physiological processes. In fetal life breath<strong>in</strong>g is essential forlung development and <strong>in</strong> the mature lung ventilationstimulates surfactant production by type II pneumocytes. 70Central to this is the concept of mechanotransduction, wherebyphysical forces are detected by cells and converted <strong>in</strong>tobiochemical signals. There is now good evidence thatsignall<strong>in</strong>g events activated by <strong>in</strong>jurious ventilation play a role80 81<strong>in</strong> VILI.The <strong>in</strong>crease <strong>in</strong> lung vascular permeability <strong>in</strong>duced <strong>in</strong>isolated perfused rat lungs by high airway pressure ventilationcan be blocked by gadol<strong>in</strong>ium <strong>in</strong> the perfusate. 82 Gadol<strong>in</strong>iumprobably exerts this effect through its <strong>in</strong>hibition of stretchactivated cation channels. This <strong>in</strong>dicates that the oedema seen<strong>in</strong> <strong>in</strong>jurious ventilation is, at least <strong>in</strong> part, due to the activationof specific cellular processes rather than simply be<strong>in</strong>g a reflectionof physical disruption of the alveolar-capillary barrier(the “stretched pore” phenomenon).Considerable attention has focused recently on the releaseof <strong>in</strong>flammatory mediators from lung tissue exposed tomechanical forces. A number of studies <strong>in</strong>volv<strong>in</strong>g isolatedlungs or <strong>in</strong>tact lung <strong>in</strong>jured animals of different species haveshown that <strong>in</strong>jurious ventilatory strategies are associated withthe release of a variety of pro<strong>in</strong>flammatory mediators, <strong>in</strong>clud<strong>in</strong>gthromboxane B 2, platelet activat<strong>in</strong>g factor, and severalcytok<strong>in</strong>es. 83–87 This humoral <strong>in</strong>flammatory response canprecede overt histological damage and appears to be due tostretch activation of specific pathways, <strong>in</strong> addition to an<strong>in</strong>flammatory reaction to non-specific <strong>in</strong>jury. 81 87 The importanceof these mediators <strong>in</strong> caus<strong>in</strong>g lung <strong>in</strong>jury is unknown,and it is conceivable that they exert a beneficial effect <strong>in</strong> the<strong>in</strong>jured lung. 88 However, studies us<strong>in</strong>g rabbit models of VILIBiochemical <strong>in</strong>juryCytok<strong>in</strong>es, complementprostanoids, leukotrienesreactive oxygen speciesproteasesNeutrophilsmΦMechanical ventilationBacteriaBiophysical <strong>in</strong>jury• shear• overdistention• cyclic stretch• <strong>in</strong>trathoracic pressurealveolar-capillary permeabilitycardiac outputorgan perfusionFigure 8.4 Mechanisms by which mechanical ventilation mightcontribute to multiple system organ failure (MSOF). Reproduced withpermission from Slutsky and Tremblay. 93have shown that lung damage can be attenuated by adm<strong>in</strong>istrationof anti-tumour necrosis factor (TNF)-α antibodies or<strong>in</strong>terleuk<strong>in</strong> (IL)-1 receptor antagonists, suggest<strong>in</strong>g that these89 90cytok<strong>in</strong>es exert a deleterious effect. In the preterm lungcytok<strong>in</strong>es generated by mechanical ventilation may <strong>in</strong>terferewith lung development. 45 The term biotrauma has been co<strong>in</strong>edto describe this potentially <strong>in</strong>jurious <strong>in</strong>flammatory response tophysical stress.Mediators generated <strong>in</strong> response to <strong>in</strong>jurious ventilation donot rema<strong>in</strong> compartmentalised with<strong>in</strong> the lung. Experimentswith perfused mouse lungs and with lung <strong>in</strong>jured rats <strong>in</strong> vivohave <strong>in</strong>dicated that <strong>in</strong>jurious ventilation leads to <strong>in</strong>creasedcytok<strong>in</strong>e levels <strong>in</strong> the systemic circulation, and recent studies85 86 91 92suggest that the same applies <strong>in</strong> the cl<strong>in</strong>ical sett<strong>in</strong>g.This has led to the hypothesis that mechanical ventilation canfuel the systemic <strong>in</strong>flammatory response commonly seen <strong>in</strong>ARDS and contribute to the development of multiple systemorgan failure (MSOF). 93Ventilation may also <strong>in</strong>fluence the systemic <strong>in</strong>flammatoryresponse through translocation of bacteria or their productsfrom the air spaces <strong>in</strong>to the circulation. In dogs and ratsbacteraemia is more likely to develop when lungs that havebeen <strong>in</strong>oculated with bacteria are ventilated with high tidal94 95volume/zero PEEP compared with less <strong>in</strong>jurious strategies.An analogous effect has been observed <strong>in</strong> ventilated rabbitsfollow<strong>in</strong>g <strong>in</strong>tratracheal adm<strong>in</strong>istration of lipopolysaccharide(LPS). Injurious ventilation resulted <strong>in</strong> much higher levels ofcirculat<strong>in</strong>g LPS accompanied by a rise <strong>in</strong> TNF-α. 96 The possibleways <strong>in</strong> which ventilation impacts on systemic <strong>in</strong>flammationand distal organs are summarised <strong>in</strong> fig 8.4.CLINICAL CONSEQUENCES OF VILIDespite the difficulties <strong>in</strong> dist<strong>in</strong>guish<strong>in</strong>g the effects ofmechanical ventilation from those of the underly<strong>in</strong>g condition,there are now clear data show<strong>in</strong>g the cl<strong>in</strong>ical impact of•••Distal organs• tissue <strong>in</strong>jury secondary to<strong>in</strong>flammatory mediators/cells• impaired oxygen delivery• bacteremiaMSOF


Ventilator <strong>in</strong>duced lung <strong>in</strong>jury 57VILI <strong>in</strong> two conditions—neonatal respiratory distress syndromeand ARDS.Neonatal chronic lung diseaseMost cases of neonatal chronic lung disease (CLD), alsoknown as bronchopulmonary dysplasia, occur <strong>in</strong> the aftermathof neonatal respiratory distress syndrome. Hyperoxiaand mechanical ventilation have been implicated <strong>in</strong> itsaetiology. 97 Evidence for the role of mechanical ventilation <strong>in</strong>this respect <strong>in</strong>cludes the observation that neonatal <strong>in</strong>tensivecare units with high rates of <strong>in</strong>tubation and ventilation alsohave high rates of CLD without improvements <strong>in</strong> mortality orother morbidity. 98 99 At two centres with very different rates ofmechanical ventilation of low birth weight <strong>in</strong>fants (75% v29%) and prevalence of CLD (22% v 4%) multivariateregression analysis <strong>in</strong>dicated that the development of CLDwas strongly associated with the <strong>in</strong>itiation of mechanicalventilation. 99 Several trials have addressed the use of differentventilatory strategies, particularly HFOV, <strong>in</strong> neonatal respiratorydistress. Despite its theoretical advantages over conventionalventilation <strong>in</strong> terms of reduc<strong>in</strong>g lung <strong>in</strong>jury, the role ofHFOV <strong>in</strong> neonatal respiratory distress rema<strong>in</strong>s25 100controversial.ARDSThe magnitude of the cl<strong>in</strong>ical burden of VILI was shown bythe recent ARDSnet trial <strong>in</strong> which 861 patients with ARDSwere randomised to receive either a “traditional” tidal volume(12 ml/kg predicted body weight) or a low tidal volume strategy(6 ml/kg). 2 Mortality was 39.8% <strong>in</strong> the traditional groupand 31% <strong>in</strong> the low volume group. In other words, at least 8.8%of the absolute mortality from ARDS is attributable to VILI. Aconsiderable amount of attention is currently directed at thepotential cl<strong>in</strong>ical benefits of improv<strong>in</strong>g and ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g lungrecruitment, based on the theories outl<strong>in</strong>ed above, us<strong>in</strong>ghigher levels of PEEP or HFOV.It is <strong>in</strong>terest<strong>in</strong>g to speculate on precisely how VILI <strong>in</strong>creasesmortality. The <strong>in</strong>jury to the lung described <strong>in</strong> experimentalmodels probably occurs <strong>in</strong> humans, to a greater or lesserdegree. However, most deaths <strong>in</strong> ARDS are from MSOF ratherthan respiratory failure. 101 It is therefore likely that mechanicalventilation can <strong>in</strong>fluence the development of MSOF, possiblythrough the release of pro<strong>in</strong>flammatory mediators, asdescribed above. Two recent cl<strong>in</strong>ical studies add weight to thishypothesis. Ranieri and coworkers exam<strong>in</strong>ed the effect of twoventilatory strategies on cytok<strong>in</strong>e levels <strong>in</strong> ARDS. Forty fourpatients were randomised either to a “protective” strategy, <strong>in</strong>which the PEEP and tidal volume were set such that tidalventilation occurred exclusively between the lower and upper<strong>in</strong>flection po<strong>in</strong>ts of the pressure volume curve (see fig 8.1), ora “control” strategy <strong>in</strong> which the tidal volume was set toobta<strong>in</strong> normal values of arterial CO 2and the PEEP set to producethe greatest improvement <strong>in</strong> arterial oxygen saturationwithout worsen<strong>in</strong>g haemodynamics. The protective group hadsignificantly lower levels of plasma and bronchoalveolarlavage cytok<strong>in</strong>es and significantly less organ failure. 92 102 Alongsimilar l<strong>in</strong>es, the ARDSnet study found that plasma levels ofIL-6 fell significantly more <strong>in</strong> patients ventilated with thelower than the traditional tidal volume. 2It therefore seemslikely that mechanical ventilation <strong>in</strong> ARDS can promotesystemic <strong>in</strong>flammation and multiorgan failure. Crucially,however, it is not known which factor(s) are responsible formediat<strong>in</strong>g this detrimental effect and how they exert a toxiceffect on distal organs.In addition to <strong>in</strong>creas<strong>in</strong>g mortality <strong>in</strong> ARDS, VILI may contributeto the persistent lung function abnormalities (pr<strong>in</strong>cipallya restrictive defect with abnormal transfer factor) seen <strong>in</strong>a m<strong>in</strong>ority of survivors. 103However, no studies to date haveshown that protective ventilatory strategies <strong>in</strong> ARDS are associatedwith improved long term lung function. 104CONCLUSIONExtensive research over the past 30 years has identified keydeterm<strong>in</strong>ants of VILI. From this, practice has been modified <strong>in</strong>an attempt to m<strong>in</strong>imise lung damage. In the case of ARDS,ventilation with lower tidal volumes has been shown to reducemortality.One of the most excit<strong>in</strong>g developments has been the realisationthat VILI may be caused not only by the mechanicaldisruption of lung tissue, but also by the <strong>in</strong>appropriate activationof cellular pathways. Such mechanisms may contribute tonon-pulmonary organ damage. Future treatments to m<strong>in</strong>imisethe impact of VILI may target these mechanisms at themolecular level, <strong>in</strong> addition to develop<strong>in</strong>g less <strong>in</strong>jurious ventilationstrategies.ACKNOWLEDGEMENTSTW is supported by the Scadd<strong>in</strong>g-Morriston-Davies Trust. AS is supportedby the Canadian Institutes of Health Research (grant # 8558).REFERENCES1 Fothergill J. Observations on a case published <strong>in</strong> the last volume of themedical essays, &c of recover<strong>in</strong>g a man dead <strong>in</strong> appearance, bydistend<strong>in</strong>g the lungs with air. Phil Trans R Soc Med 1745;43:275–81.2 The Acute <strong>Respiratory</strong> Distress Syndrome Network. Ventilation withlower tidal volumes as compared with traditional tidal volumes for acutelung <strong>in</strong>jury and the acute respiratory distress syndrome. N Engl J Med2000;342:1301–8.3 Mackl<strong>in</strong> MT, Mackl<strong>in</strong> CC. Malignant <strong>in</strong>terstitial emphysema of the lungsand mediast<strong>in</strong>um as an important occult complication <strong>in</strong> many respiratorydiseases and other conditions: an <strong>in</strong>terpretation of the cl<strong>in</strong>ical literature <strong>in</strong>the light of laboratory experiment. Medic<strong>in</strong>e 1944;23:281–352.4 Petersen GW, Baier H. Incidence of pulmonary barotrauma <strong>in</strong> a medicalICU. Crit <strong>Care</strong> Med 1983;11:67–9.5 Weg JG, Anzueto A, Balk RA, et al. The relation of pneumothorax andother air leaks to mortality <strong>in</strong> the acute respiratory distress syndrome. NEngl J Med 1998;338:341–6.6 Stewart TE, Meade MO, Cook DJ, et al. Evaluation of a ventilationstrategy to prevent barotrauma <strong>in</strong> patients at high risk of acute respiratorydistress syndrome. N Engl J Med 1998;338:355–61.7 Brochard L, Roudot-Thoraval F, Roupie E, et al. Tidal volume reductionfor prevention of ventilator-<strong>in</strong>duced lung <strong>in</strong>jury <strong>in</strong> acute respiratorydistress syndrome. The Multicenter Trial Group on Tidal Volume reduction<strong>in</strong> ARDS. Am J Respir Crit <strong>Care</strong> Med 1998;158:1831–8.8 Downs JB, Chapman RL Jr. Treatment of bronchopleural fistula dur<strong>in</strong>gcont<strong>in</strong>uous positive pressure ventilation. Chest 1976;69:363–6.9 Webb HH, Tierney DF. Experimental pulmonary edema due to<strong>in</strong>termittent positive pressure ventilation with high <strong>in</strong>flation pressures.Protection by positive end-expiratory pressure. Am Rev Respir Dis1974;110:556–65.10 Dreyfuss D, Soler P, Saumon G. Spontaneous resolution of pulmonaryedema caused by short periods of cyclic over<strong>in</strong>flation. J Appl Physiol1992;72:2081–9.11 John E, Ermocilla R, Golden J, et al. Effects of <strong>in</strong>termittentpositive-pressure ventilation on lungs of normal rabbits. Br J Exp Pathol1980;61:315–23.12 Kolobow T, Moretti MP, Fumagalli R, et al. Severe impairment <strong>in</strong> lungfunction <strong>in</strong>duced by high peak airway pressure dur<strong>in</strong>g mechanicalventilation. An experimental study. Am Rev Respir Dis 1987;135:312–5.13 Bouhuys A. Physiology and musical <strong>in</strong>struments. Nature1969;221:1199–204.14 Dreyfuss D, Soler P, Basset G, et al. High <strong>in</strong>flation pressure pulmonaryedema. Respective effects of high airway pressure, high tidal volume,and positive end-expiratory pressure. Am J Respir Crit <strong>Care</strong> Med1988;137:1159–64.15 Hernandez LA, Peevy KJ, Moise A, et al. Chest wall restriction limitshigh airway pressure-<strong>in</strong>duced lung <strong>in</strong>jury <strong>in</strong> young rabbits. J Appl Physiol1989;66:2364–8.16 Carlton DP, Cumm<strong>in</strong>gs JJ, Scheerer RG. Lung overexpansion <strong>in</strong>creasespulmonary microvascular prote<strong>in</strong> permeability <strong>in</strong> young lambs. J ApplPhysiol 1990;69:577–83 .17 Slutsky AS. Mechanical ventilation. American College of ChestPhysicians’ Consensus Conference. Chest 1993;104:1833–59.18 Ranieri VM, Brienza N, Santostasi S, et al. Impairment of lung and chestwall mechanics <strong>in</strong> patients with acute respiratory distress syndrome: roleof abdom<strong>in</strong>al distension. Am J Respir Crit <strong>Care</strong> Med 1997;156:1082–91.19 Argiras EP, Blakeley CR, Dunnill MS, et al. High PEEP decreases hyal<strong>in</strong>emembrane formation <strong>in</strong> surfactant deficient lungs.Br J Anaesth1987;59:1278–85.20 Sandhar BK, Niblett DJ, Argiras EP, et al. Effects of positiveend-expiratory pressure on hyal<strong>in</strong>e membrane formation <strong>in</strong> a rabbitmodel of the neonatal respiratory distress syndrome. Intensive <strong>Care</strong> Med1988;14:538–46.


58 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>21 Muscedere JG, Mullen JB, Gan K, et al. Tidal ventilation at low airwaypressures can augment lung <strong>in</strong>jury. Am J Respir Crit <strong>Care</strong> Med1994;149:1327–34.22 Matamis D, Lemaire F, Harf A, et al. Total respiratory pressure-volumecurves <strong>in</strong> the adult respiratory distress syndrome. Chest 1984;86:58–66.23 Amato MB, Barbas CS, Medeiros DM, et al. Beneficial effects of the“open lung approach” with low distend<strong>in</strong>g pressures <strong>in</strong> acute respiratorydistress syndrome. A prospective randomized study on mechanicalventilation. Am J Respir Crit <strong>Care</strong> Med 1995;152:1835–46.24 McCulloch PR, Forkert PG, Froese AB. Lung volume ma<strong>in</strong>tenanceprevents lung <strong>in</strong>jury dur<strong>in</strong>g high frequency oscillatory ventilation <strong>in</strong>surfactant-deficient rabbits. Am Rev Respir Dis 1988;137:1185–92.25 Riphagen S, Bohn D. High frequency oscillatory ventilation. Intensive<strong>Care</strong> Med 1999;25:1459–62.26 Radford PR. Static mechanical properties of mammalian lungs. In: FennWO, Rahn H, eds. Handbook of physiology. Wash<strong>in</strong>gton DC: AmericanPhysiological Society, 1964: 429–49.27 Hickl<strong>in</strong>g KG. The pressure-volume curve is greatly modified byrecruitment. A mathematical model of ARDS lungs. Am J Respir Crit <strong>Care</strong>Med 1998;158:194–202.28 Martynowicz MA, M<strong>in</strong>or TA, Walters BJ, et al. Regional expansion ofoleic acid-<strong>in</strong>jured lungs. Am J Respir Crit <strong>Care</strong> Med 1999;160:250–8.29 Rimensberger PC, Cox PN, Frndova H, et al. The open lung dur<strong>in</strong>gsmall tidal volume ventilation: concepts of recruitment and “optimal”positive end-expiratory pressure. Crit <strong>Care</strong> Med 1999;27:1946–52.30 Gatt<strong>in</strong>oni L, D’Andrea L, Pelosi P, et al. Regional effects and mechanismof positive end-expiratory pressure <strong>in</strong> early adult respiratory distresssyndrome. JAMA 1993;269:2122–7.31 Dreyfuss D, Saumon G. Pressure-volume curves. Search<strong>in</strong>g for the grailor lay<strong>in</strong>g patients with adult respiratory distress syndrome on Procrustes’bed? Am J Respir Crit <strong>Care</strong> Med 2001;163:2–3.32 Hotchkiss JR, Blanch L, Murias G, et al. Effects of decreased respiratoryfrequency on ventilator-<strong>in</strong>duced lung <strong>in</strong>jury. Am J Respir Crit <strong>Care</strong> Med2000;161:463–8.33 Chabot F, Mitchell JA, Gutteridge JM, et al. Reactive oxygen species <strong>in</strong>acute lung <strong>in</strong>jury. Eur Respir J 1998;11:745–57.34 Davis WB, Rennard SI, Bitterman PB, et al. Pulmonary oxygen toxicity.Early reversible changes <strong>in</strong> human alveolar structures <strong>in</strong>duced byhyperoxia. N Engl J Med 1983;309:878–83.35 Hickl<strong>in</strong>g KG, Henderson SJ, Jackson R. Low mortality associated withlow volume pressure limited ventilation with permissive hypercapnia <strong>in</strong>severe adult respiratory distress syndrome. Intensive <strong>Care</strong> Med1990;16:372–7.36 Laffey JG, Kavanagh BP. Biological effects of hypercapnia. Intensive<strong>Care</strong> Med 2000;26:133–8.37 Nash G, Bowen JA, Langl<strong>in</strong>ais PC. “Respirator lung”: a misnomer. ArchPathol 1971;91:234–40.38 Bowton DL, Kong DL. High tidal volume ventilation produces <strong>in</strong>creasedlung water <strong>in</strong> oleic acid-<strong>in</strong>jured rabbit lungs. Crit <strong>Care</strong> Med1989;17:908–11.39 Hernandez LA, Coker PJ, May S, et al. Mechanical ventilation <strong>in</strong>creasesmicrovascular permeability <strong>in</strong> oleic acid-<strong>in</strong>jured lungs. J Appl Physiol1990;69:2057–61.40 Dreyfuss D, Soler P, Saumon G Mechanical ventilation-<strong>in</strong>ducedpulmonary edema. Interaction with previous lung alterations. Am J RespirCrit <strong>Care</strong> Med 1995;151:1568–75.41 Gatt<strong>in</strong>oni L, Pesenti A, Avalli L, et al. Pressure-volume curve of totalrespiratory system <strong>in</strong> acute respiratory failure. Computed tomographicscan study. Am Rev Respir Dis 1987;136:730–6.42 Goldste<strong>in</strong> I, Bughalo MT, Marquette CH, et al. Mechanicalventilation-<strong>in</strong>duced air-space enlargement dur<strong>in</strong>g experimentalpneumonia <strong>in</strong> piglets. Am J Respir Crit <strong>Care</strong> Med 2001;163:958–64.43 Rouby JJ, Lherm T, Mart<strong>in</strong> de Lassale E, et al. Histologic aspects ofpulmonary barotrauma <strong>in</strong> critically ill patients with acute respiratoryfailure. Intensive <strong>Care</strong> Med 1993;19:383–9.44 Desai SR, Wells AU, Rubens MB, et al. Acute respiratory distresssyndrome: CT abnormalities at long-term follow-up. Radiology1999;210:29–35.45 Jobe AH, Ikegami M. Lung development and function <strong>in</strong> preterm <strong>in</strong>fants<strong>in</strong> the surfactant treatment era. Ann Rev Physiol 2000;62:825–46.46 Bjorklund LL, Ingimarsson J, Curstedt T, et al. Manual ventilation with afew large breaths at birth compromises the therapeutic effect ofsubsequent surfactant replacement <strong>in</strong> immature lambs. Pediatr Res1997;42:348–55.47 Dreyfuss D, Saumon G. Ventilator-<strong>in</strong>duced lung <strong>in</strong>jury: lessons fromexperimental studies. Am J Respir Crit <strong>Care</strong> Med 1998;157:294–323.48 Egan EA. Lung <strong>in</strong>flation, lung solute permeability, and alveolar edema. JAppl Physiol 1982;53:121–5.49 KimKJ, Crandall ED. Effects of lung <strong>in</strong>flation on alveolar epithelialsolute and water transport properties. J Appl Physiol 1982;52:1498–505.50 Dreyfuss D, Basset G, Soler P, et al. Intermittent positive-pressurehyperventilation with high <strong>in</strong>flation pressures produces pulmonarymicrovascular <strong>in</strong>jury <strong>in</strong> rats. Am Rev Respir Dis 1985;132:880–4.51 Parker JC, Townsley MI, Rippe B, et al. Increased microvascularpermeability <strong>in</strong> dog lungs due to high airway pressures. J Appl Physiol1984;57:1809–16.52 Albert RK, Lakshm<strong>in</strong>arayan S, Kirk W, et al. Lung <strong>in</strong>flation can causepulmonary edema <strong>in</strong> zone I of <strong>in</strong> situ dog lungs. J Appl Physiol1980;49:815–9.53 Tsuno K, Miura K, Takeya M, et al. Histopathologic pulmonary changesfrom mechanical ventilation at high peak airway pressures. Am RevRespir Dis 1991;143:1115–20.54 Hamilton PP, Onayemi A, Smyth JA, et al. Comparison of conventionaland high-frequency ventilation: oxygenation and lung pathology. J ApplPhysiol 1983;55:131–8.55 Kawano T, Mori S, Cybulsky M, et al. Effect of granulocyte depletion <strong>in</strong>a ventilated surfactant-depleted lung. J Appl Physiol 1987;62:27–33.56 Mathieu-Costello OA, West JB. Are pulmonary capillaries susceptibleto mechanical stress? Chest 1994;105:102–7S.57 West JB, Tsukimoto K, Mathieu-Costello O, et al. Stress failure <strong>in</strong>pulmonary capillaries. J Appl Physiol 1991;70:1731–42.58 Tsukimoto K, Mathieu-Costello O, Prediletto R, et al. Ultrastructuralappearances of pulmonary capillaries at high transmural pressures. JAppl Physiol 1991;71:573–82.59 Fu Z, Costello ML, Tsukimoto K, et al. High lung volume <strong>in</strong>creases stressfailure <strong>in</strong> pulmonary capillaries. J Appl Physiol 1992;73:123–33.60 Broccard AF, Hotchkiss JR, Kuwayama N, et al. Consequences ofvascular flow on lung <strong>in</strong>jury <strong>in</strong>duced by mechanical ventilation. Am JRespir Crit <strong>Care</strong> Med 1998;157:1935–42.61 Mead J, Takishima T, Leith D. Stress distribution <strong>in</strong> lungs: a model ofpulmonary elasticity. J Appl Physiol 1970;28:596–608.62 Amato MBP Mar<strong>in</strong>i JJ. Barotrauma, volutrauma, and ventilation of acutelung <strong>in</strong>jury. In: Mar<strong>in</strong>i JJ Slutsky AS, eds. Physiological basis of ventilatorysupport. New York: Marcel Dekker, 1998: 1187–245.63 Kamm RD, Schroter RC. Is airway closure caused by a liquid film<strong>in</strong>stability? Respir Physiol 1989;75:141–56.64 Macklem PT, Proctor DF, Hogg JC. The stability of peripheral airways.Respir Physiol 1970;8:191–203.65 Naureckas ET, Dawson CA, Gerber BS, et al. Airway reopen<strong>in</strong>gpressure <strong>in</strong> isolated rat lungs. J Appl Physiol 1994;76:1372–7.66 Gaver DP, Samsel RW, Solway J. Effects of surface tension and viscosityon airway reopen<strong>in</strong>g. J Appl Physiol 1990;69:74–85.67 Taskar V, John J, Evander E, et al. Healthy lungs tolerate repetitivecollapse and reopen<strong>in</strong>g dur<strong>in</strong>g short periods of mechanical ventilation.Acta Anaesthesiol Scand 1995;39:370–6.68 Lewis JF, Jobe AH. Surfactant and the adult respiratory distresssyndrome. Am Rev Respir Dis 1993;147:218–33.69 Vanderzwan J, McCaig L, Mehta S, et al. Characteriz<strong>in</strong>g alterations <strong>in</strong>the pulmonary surfactant system <strong>in</strong> a rat model of Pseudomonasaerug<strong>in</strong>osa pneumonia. Eur Respir J 1998;12:1388–96.70 Chander A, Fisher AB. Regulation of lung surfactant secretion. Am JPhysiol 1990;258:L241–53.71 Malloy J, McCaig L, Veldhuizen R, et al. Alterations of the endogenoussurfactant system <strong>in</strong> septic adult rats. Am J Respir Crit <strong>Care</strong> Med1997;156:617–23.72 Coker PJ, Hernandez LA, Peevy KJ, et al. Increased sensitivity tomechanical ventilation after surfactant <strong>in</strong>activation <strong>in</strong> young rabbit lungs.Crit <strong>Care</strong> Med 1992;20:635–40.73 Imai Y, Kawano T, Miyasaka K, et al. Inflammatory chemical mediatorsdur<strong>in</strong>g conventional ventilation and dur<strong>in</strong>g high frequency oscillatoryventilation. Am J Respir Crit <strong>Care</strong> Med 1994;150:1550–4.74 Taskar V, Wollmer P, Evander E, et al. Effect of detergent comb<strong>in</strong>ed withlarge tidal volume ventilation on alveolocapillary permeability. Cl<strong>in</strong>Physiol 1996;16:103–14.75 Taskar V, John J, Evander E, et al. Surfactant dysfunction makes lungsvulnerable to repetitive collapse and reexpansion. Am J Respir Crit <strong>Care</strong>Med 1997;155:313–20.76 Albert RK, Lakshm<strong>in</strong>arayan S, Hildebrandt J, et al. Increased surfacetension favors pulmonary edema formation <strong>in</strong> anesthetized dogs’ lungs. JCl<strong>in</strong> Invest 1979;63:1015–8.77 Wright JR. Immunomodulatory functions of surfactant. Physiol Rev1997;77:931–62.78 Jobe AH. Pulmonary surfactant therapy. N Engl J Med1993;328:861–8.79 Anzueto A, Baughman RP, Guntupalli KK, et al. Aerosolized surfactant<strong>in</strong> adults with sepsis-<strong>in</strong>duced acute respiratory distress syndrome. ExosurfAcute <strong>Respiratory</strong> Distress Syndrome Sepsis Study Group. N Engl J Med1996;334:1417–21.80 Liu M, Tanswell AK, Post M. Mechanical force-<strong>in</strong>duced signaltransduction <strong>in</strong> lung cells. Am J Physiol 1999;277:L667–83.81 Dos Santos CC, Slutsky AS. Invited review: mechanisms ofventilator-<strong>in</strong>duced lung <strong>in</strong>jury: a perspective. J Appl Physiol2000;89:1645–55.82 Parker JC, Ivey CL, Tucker JA. Gadol<strong>in</strong>ium prevents high airwaypressure-<strong>in</strong>duced permeability <strong>in</strong>creases <strong>in</strong> isolated rat lungs. J ApplPhysiol 1998;84:1113–8.83 Imai Y, Kawano T, Miyasaka K, et al. Inflammatory chemical mediatorsdur<strong>in</strong>g conventional ventilation and dur<strong>in</strong>g high frequency oscillatoryventilation. Am J Respir Crit <strong>Care</strong> Med 1994;150:1550–4.84 Tremblay L, Valenza F, Ribeiro SP, et al. Injurious ventilatory strategies<strong>in</strong>crease cytok<strong>in</strong>es and c-fos m-RNA expression <strong>in</strong> an isolated rat lungmodel. J Cl<strong>in</strong> Invest 1997;99:944–52.85 von Bethmann AN, Brasch F, Nus<strong>in</strong>g R, et al. Hyperventilation <strong>in</strong>ducesrelease of cytok<strong>in</strong>es from perfused mouse lung. Am J Respir Crit <strong>Care</strong>Med 1998;157:263–72.86 Chiumello D, Prist<strong>in</strong>e G, Slutsky AS. Mechanical ventilation affects localand systemic cytok<strong>in</strong>es <strong>in</strong> an animal model of acute respiratory distresssyndrome. Am J Respir Crit <strong>Care</strong> Med 1999;160:109–16.87 Held H, Boettcher S, Hamann L, et al. Ventilation-<strong>in</strong>duced chemok<strong>in</strong>eand cytok<strong>in</strong>e release is associated with activation of NFκB and isblocked by steroids. Am J Respir Crit <strong>Care</strong> Med 2001;163:711–6.88 Plotz FB, van Vught H, Heijnen CJ. Ventilator-<strong>in</strong>duced lung <strong>in</strong>flammation:is it always harmful? Intensive <strong>Care</strong> Med 1999;25:236.89 Imai Y, Kawano T, Iwamoto S, et al. Intratracheal anti-tumor necrosisfactor-alpha antibody attenuates ventilator-<strong>in</strong>duced lung <strong>in</strong>jury <strong>in</strong> rabbits.J Appl Physiol 1999;87:510–5.


Ventilator <strong>in</strong>duced lung <strong>in</strong>jury 5990 Narimanbekov IO, Rozycki HJ. Effect of IL-1 blockade on <strong>in</strong>flammatorymanifestations of acute ventilator-<strong>in</strong>duced lung <strong>in</strong>jury <strong>in</strong> a rabbit model.Exp Lung Res 1995;21:239–54.91 Haitsma JJ, Uhlig S, Goggel R, et al. Ventilator-<strong>in</strong>duced lung <strong>in</strong>juryleads to loss of alveolar and systemic compartmentalization of tumornecrosis factor-alpha. Intensive <strong>Care</strong> Med 2000;26:1515–22.92 Ranieri V, Suter P, Tortorella C, et al. Effect of mechanical ventilation on<strong>in</strong>flammatory mediators <strong>in</strong> patients with acute respiratory distresssyndrome: a randomized controlled trial. JAMA 1999;282:54–61.93 Slutsky AS, Tremblay LN. Multiple system organ failure. Is mechanicalventilation a contribut<strong>in</strong>g factor? Am J Respir Crit <strong>Care</strong> Med1998;157:1721–5.94 Nahum A, Hoyt J, Schmitz L, et al. Effect of mechanical ventilationstrategy on dissem<strong>in</strong>ation of <strong>in</strong>tratracheally <strong>in</strong>stilled Escherichia coli <strong>in</strong>dogs. Crit <strong>Care</strong> Med 1997;25:1733–43.95 Verbrugge SJ, Sorm V, van‘t Veen A, et al. Lung over<strong>in</strong>flation withoutpositive end-expiratory pressure promotes bacteremia after experimentalKlebsiella pneumoniae <strong>in</strong>oculation. Intensive <strong>Care</strong> Med 1998;24:172–7.96 Murphy DB, Cregg N, Tremblay L, et al. Adverse ventilatory strategycauses pulmonary-to-systemic translocation of endotox<strong>in</strong>. Am J Respir Crit<strong>Care</strong> Med 2000;162:27–33.97 Northway WH Jr. Bronchopulmonary dysplasia: twenty-five years later.Pediatrics 1992;89:969–73.98 Poets CF, Sens B. Changes <strong>in</strong> <strong>in</strong>tubation rates and outcome of very lowbirth weight <strong>in</strong>fants: a population-based study. Pediatrics 1996;98:24–7.99 Van Marter LJ, Allred EN, Pagano M, et al. Do cl<strong>in</strong>ical markers ofbarotrauma and oxygen toxicity expla<strong>in</strong> <strong>in</strong>terhospital variation <strong>in</strong> rates ofchronic lung disease? The Neonatology Committee for the DevelopmentalNetwork. Pediatrics 2000;105:1194–201.100 Thome U, Kossel H, Lipowsky G, et al. Randomized comparison ofhigh-frequency ventilation with high-rate <strong>in</strong>termittent positive pressureventilation <strong>in</strong> preterm <strong>in</strong>fants with respiratory failure. J Pediatr1999;135:39–46.101 Ferr<strong>in</strong>g M, V<strong>in</strong>cent JL. Is outcome from ARDS related to the severity ofrespiratory failure? Eur Respir J 1997;10:1297–300.102 Ranieri VM, Giunta F, Suter PM, et al. Mechanical ventilation as amediator of multisystem organ failure <strong>in</strong> acute respiratory distresssyndrome. JAMA 2000;284:43–4.103 Hudson LD, Ste<strong>in</strong>berg KP. Epidemiology of acute lung <strong>in</strong>jury and ARDS.Chest 1999;116:74–82S.104 Cooper AB, Ferguson ND, Hanly PJ, et al. Long-term follow-up ofsurvivors of acute lung <strong>in</strong>jury: lack of effect of a ventilation strategy toprevent barotrauma. Crit <strong>Care</strong> Med 1999;27:2616–21.


9 Ventilatory management of acute lung <strong>in</strong>jury/acuterespiratory distress syndromeJ J Cord<strong>in</strong>gley, B F Keogh.............................................................................................................................The ventilatory management of patients withacute lung <strong>in</strong>jury (ALI) and acute respiratorydistress syndrome (ARDS) has evolved <strong>in</strong>conjunction with advances <strong>in</strong> understand<strong>in</strong>g ofthe underly<strong>in</strong>g pathophysiology. In particular,evidence that mechanical ventilation has an<strong>in</strong>fluence on lung <strong>in</strong>jury and patient outcome hasemerged over the past three decades. 1 The presentunderstand<strong>in</strong>g of optimal ventilatory managementis outl<strong>in</strong>ed and other methods of respiratorysupport are reviewed.PATHOPHYSIOLOGYThe pathophysiology of ARDS is reviewed byBell<strong>in</strong>gan <strong>in</strong> chapter 6. However, it is useful tohighlight important features relevant to ventilatorymanagement, <strong>in</strong> particular the anatomicaldistribution of pulmonary pathology and thepotential for ventilator <strong>in</strong>duced lung <strong>in</strong>jury.The orig<strong>in</strong>al description of ARDS <strong>in</strong>cluded thepresence of bilateral <strong>in</strong>filtrates on the chestradiograph. 2 Computerised tomographic (CT)scann<strong>in</strong>g has shown that parenchymal consolidation,far from be<strong>in</strong>g evenly distributed, is concentrated<strong>in</strong> dependent lung regions leav<strong>in</strong>g nondependentlung relatively spared. Thispathological distribution of aerated lung ly<strong>in</strong>gover areas of dense consolidation has led to comparisonswith ventilation of a much smaller or“baby lung” 3 and has important implications forventilatory management. Thus, the application ofnormal physiological tidal volumes can lead tooverdistension of the small volume of normallyaerated lung, while fail<strong>in</strong>g to recruit consolidateddependent regions.Ventilator <strong>in</strong>duced lung <strong>in</strong>jury can occur byseveral mechanisms: oxygen toxicity from the useof high FiO 2, 4 (see chapter 8) overdistension of thelung caus<strong>in</strong>g local damage and further<strong>in</strong>flammation, 5<strong>in</strong>jurious cyclical open<strong>in</strong>g andclos<strong>in</strong>g of alveoli from ventilation at low lungvolumes, 6and by <strong>in</strong>creas<strong>in</strong>g systemic levels of<strong>in</strong>flammatory cytok<strong>in</strong>es. 7Ventilatory strategies must therefore be tailoredto m<strong>in</strong>imise the risk of <strong>in</strong>duc<strong>in</strong>g or exacerbat<strong>in</strong>glung <strong>in</strong>jury.RESPIRATORY MECHANICSDecreased lung compliance is a prom<strong>in</strong>ent featureof ARDS. The static compliance of the respiratorysystem (lung + chest wall) <strong>in</strong> a ventilated patientis calculated by divid<strong>in</strong>g the tidal volume (Vt) byend <strong>in</strong>spiratory plateau pressure (Pplat) m<strong>in</strong>usend expiratory pressure + <strong>in</strong>tr<strong>in</strong>sic PEEP(PEEPi). As the pathology of ARDS is heterogeneous,calculat<strong>in</strong>g static compliance does notprovide <strong>in</strong>formation about regional variations <strong>in</strong>lung recruitment and varies accord<strong>in</strong>g to lungvolume. Much attention has therefore focused onanalysis of the pressure-volume (PV) curve.The static PV curve of the respiratory systemcan be obta<strong>in</strong>ed by <strong>in</strong>sert<strong>in</strong>g a pause dur<strong>in</strong>g an<strong>in</strong>flation-deflation cycle. A number of differentmethods have been described <strong>in</strong>clud<strong>in</strong>g the use ofa large syr<strong>in</strong>ge (super-syr<strong>in</strong>ge), or hold<strong>in</strong>g amechanical ventilator at end <strong>in</strong>spiration ofvary<strong>in</strong>g tidal volumes. The pr<strong>in</strong>ciples and methodsof PV curve measurement have recently beenreviewed. 8The PV curves thus obta<strong>in</strong>ed are sigmoidal andhave an <strong>in</strong>spiratory limb that usually <strong>in</strong>cludes apo<strong>in</strong>t above which the curve becomes steeper (fig9.1). 3 Identification of the lower <strong>in</strong>flection po<strong>in</strong>tby cl<strong>in</strong>icians us<strong>in</strong>g PV curves is subject to largevariability, but is improved by curve fitt<strong>in</strong>g. 9Insome patients the lower <strong>in</strong>flection po<strong>in</strong>t may beabsent. At higher lung volumes the curvebecomes flatter aga<strong>in</strong> (upper <strong>in</strong>flection po<strong>in</strong>t),above which further <strong>in</strong>creases <strong>in</strong> pressure causelittle <strong>in</strong>crease <strong>in</strong> volume. Currently, ventilatorsused rout<strong>in</strong>ely <strong>in</strong> <strong>in</strong>tensive care units do not haveautomated functions to obta<strong>in</strong> a static PV curve.Moreover, the static PV curve only provides <strong>in</strong>formationabout accessible lung 3and also <strong>in</strong>cludeschest wall compliance. Separat<strong>in</strong>g the lung andchest wall components requires the use ofoesophageal pressure measurement. 10Despite these limitations, many advances <strong>in</strong>cl<strong>in</strong>ical management <strong>in</strong> patients with ALI/ARDShave been based on consideration of static PVcurves. It has recently been suggested that use ofthe lower and upper <strong>in</strong>flection po<strong>in</strong>ts of the staticPV curve as <strong>in</strong>dicators of recruitment and overdistension<strong>in</strong> order to adjust ventilator sett<strong>in</strong>gs <strong>in</strong>patients with ARDS is unreliable. 11It is arguedthat alveolar recruitment occurs beyond the lower<strong>in</strong>flection po<strong>in</strong>t and that further <strong>in</strong>formation,<strong>in</strong>clud<strong>in</strong>g the deflation PV curve, is required todeterm<strong>in</strong>e optimal ventilator sett<strong>in</strong>gs for an <strong>in</strong>dividualpatient. Analysis of the <strong>in</strong>spiratorypressure-time curve under conditions of constantflow may also provide useful <strong>in</strong>formation aboutlung recruitment. 12VENTILATORY STRATEGIES IN ARDSThe goals of ventilat<strong>in</strong>g patients with ALI/ARDSare to ma<strong>in</strong>ta<strong>in</strong> adequate gas exchange and avoidventilator <strong>in</strong>duced lung <strong>in</strong>jury.Ma<strong>in</strong>tenance of adequate gas exchangeOxygenHigh concentrations of <strong>in</strong>spired oxygen should beavoided to limit the risk of oxygen toxicity and toavoid reabsorption atelectasis. Arterial oxygensaturation (SaO 2) is used as a target <strong>in</strong> preferenceto arterial oxygen tension (PaO 2) because oxygendelivery determ<strong>in</strong>es tissue oxygenation. SaO 2values of around 90% are commonly accepted butoxygen delivery decreases quickly below 88%


Ventilatory management of ALI/ARDS 61VolumeExpirationInspirationLower <strong>in</strong>flection po<strong>in</strong>tPressureUpper <strong>in</strong>flection po<strong>in</strong>tFigure 9.1 Schematic representation of a static pressure-volumecurve of the respiratory system from a patient with ARDS. Note thelower and upper <strong>in</strong>flection po<strong>in</strong>ts of the <strong>in</strong>spiratory limb.because of the shape of the oxyhaemoglob<strong>in</strong> dissociationcurve. However, if a higher SaO 2can only be obta<strong>in</strong>ed by<strong>in</strong>creas<strong>in</strong>g airway pressure to levels that result <strong>in</strong> haemodynamiccompromise, then a lower SaO 2may have to be accepted.There is no cl<strong>in</strong>ical evidence to support the use of specificFiO 2thresholds, but it is common cl<strong>in</strong>ical practice to decreaseFiO 2below 0.6 as quickly as possible.Oxygenation can be improved by <strong>in</strong>creased alveolar recruitmentthrough the application of higher airway pressureprovided that ventilation-perfusion (V/Q) match<strong>in</strong>g is notadversely affected by the haemodynamic consequences of<strong>in</strong>creased <strong>in</strong>trathoracic pressure. Lung recruitment is usuallyobta<strong>in</strong>ed by apply<strong>in</strong>g extr<strong>in</strong>sic PEEP, <strong>in</strong>creas<strong>in</strong>g the <strong>in</strong>spiratory:expiratory(I:E) ratio, or by specific recruitment manoeuvres(discussed below).Carbon dioxideLimit<strong>in</strong>g tidal volume and peak pressure to reduce ventilator<strong>in</strong>duced lung <strong>in</strong>jury may cause hypercapnia. Strategies used tomanage hypercapnia have <strong>in</strong>cluded <strong>in</strong>creas<strong>in</strong>g tidal volumeand airway pressure, or <strong>in</strong>creas<strong>in</strong>g CO 2removal withtechniques such as tracheal gas <strong>in</strong>sufflation or extracorporealCO 2removal. In 1990 it was reported that the alternative ofsimply allow<strong>in</strong>g CO 2to rise to a higher level (permissivehypercapnia) and ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g limits on tidal volume and airwaypressure was associated with a significantly lower thanpredicted mortality from ARDS. 13The physiological consequences of hypercapnia are respiratoryacidosis, <strong>in</strong>creased cardiac output, and pulmonary hypertension.Neurological changes <strong>in</strong>clude <strong>in</strong>creased cerebralblood flow, and cerebral oedema and <strong>in</strong>tracranial haemorrhagehave been reported. 14 With severe acidosis there may bemyocardial depression, arrhythmias, and decreased responseto exogenous <strong>in</strong>otropes. Renal compensation for the respiratoryacidosis occurs slowly.Unfortunately there are no data to confirm the degree ofrespiratory acidosis that is safe. Recent studies have allowedhypercapnia as part of lung protective ventilatoryprotocols. 1 15–18Arterial pH was lower <strong>in</strong> the lung protectivegroups and the ARDSNet study <strong>in</strong>cluded the use of sodiumbicarbonate to correct arterial pH to normal. 1At present norecommendations can be made concern<strong>in</strong>g the managementof respiratory acidosis <strong>in</strong>duced by permissive hypercapnia.However, if bicarbonate is <strong>in</strong>fused, it should be adm<strong>in</strong>isteredslowly to allow CO 2excretion and avoid worsen<strong>in</strong>g of<strong>in</strong>tracellular acidosis.One method used to <strong>in</strong>crease CO 2clearance is <strong>in</strong>sufflation ofgas <strong>in</strong>to the trachea to flush out dead space CO 2and reducerebreath<strong>in</strong>g. 19Tracheal gas <strong>in</strong>sufflation has been used bothcont<strong>in</strong>uously and dur<strong>in</strong>g expiration only. As no commerciallyavailable ventilator <strong>in</strong>cludes this technique, modifications arerequired to the ventilator circuit and sett<strong>in</strong>gs to prevent <strong>in</strong>advertentand potentially dangerous <strong>in</strong>creases <strong>in</strong> <strong>in</strong>tr<strong>in</strong>sic PEEP,Vt, and peak airway pressure.In adult patients with ARDS, managed us<strong>in</strong>g pressure controlventilation, the <strong>in</strong>troduction of cont<strong>in</strong>uous tracheal gas<strong>in</strong>sufflation allowed a decrease <strong>in</strong> <strong>in</strong>spiratory pressure of5cmH 2O without <strong>in</strong>creas<strong>in</strong>g arterial carbon dioxide tension(PaCO 2). 20Tracheal gas <strong>in</strong>sufflation may therefore be usefulwhen permissive hypercapnia is contra<strong>in</strong>dicated. However,manag<strong>in</strong>g the appropriate ventilator sett<strong>in</strong>gs and adjustmentis complicated, with real potential for iatrogenic <strong>in</strong>jury.In practice, PaCO 2is allowed to rise dur<strong>in</strong>g lung protectivevolume and pressure limited ventilation. PaCO 2levels of 2–3times normal seem to be well tolerated for prolonged periods.Renal compensation for respiratory acidosis occurs overseveral days. Many cl<strong>in</strong>icians <strong>in</strong>fuse sodium bicarbonateslowly if arterial pH falls below 7.20.Avoidance of ventilator <strong>in</strong>duced lung <strong>in</strong>juryTraditional mechanical ventilation (as applied dur<strong>in</strong>g rout<strong>in</strong>egeneral anaesthesia) <strong>in</strong>volves tidal volumes that are relativelylarge (10–15 ml/kg) <strong>in</strong> order to reduce atelectasis. PEEP levelsare adjusted to ma<strong>in</strong>ta<strong>in</strong> oxygenation but high levels are generallyavoided to prevent cardiovascular <strong>in</strong>stability related to<strong>in</strong>creased <strong>in</strong>trathoracic pressure. Present understand<strong>in</strong>g ofventilator <strong>in</strong>duced lung <strong>in</strong>jury suggests that traditionalmechanical ventilation, us<strong>in</strong>g high tidal volumes and lowPEEP, is likely to enhance lung <strong>in</strong>jury <strong>in</strong> patients with ARDS.Five randomised studies of “lung protective” ventilation <strong>in</strong>ARDS have recently been published, four of which <strong>in</strong>vestigatedlimitation of tidal volume to prevent <strong>in</strong>jury from overdistension(table 9.1).In these studies the protective ventilatory strategy wasdirected at prevent<strong>in</strong>g lung overdistension and was notdesigned to look at differences <strong>in</strong> ventilation at low lung volumes.Only the largest study (ARDSNet) 1 showed anadvantage of such a protective strategy. The ARDSNet studyhad the largest difference <strong>in</strong> Vt and Pplat between the groups,the highest power, and was the only study to correct respiratoryacidosis (table 9.2).Other studies have addressed the issue of adjustment ofventilatory support based on PV curve characteristics. Amatoet al 18randomised 53 patients with early ARDS to eithertraditional ventilation (volume cycled, Vt 12 ml/kg, m<strong>in</strong>imumPEEP guided by FiO 2, normal PaCO 2) or a lung protective strategy(PEEP adjusted to above the lower <strong>in</strong>flection po<strong>in</strong>t of astatic PV curve, Vt


62 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Table 9.1Randomised prospective studies of ventilatory strategies to limit lung overdistension <strong>in</strong> patients with ARDSReference n “Protective” Control MortalityStewart (1998) 15 120 • Vt


Ventilatory management of ALI/ARDS 63Table 9.3Reported lung recruitment manoeuvresReference n Pressure (cm H 2 O) Time (s) Effective DurationAmato (1998) 18 29 35–40 40 – –Lap<strong>in</strong>sky (1999) 53 14 30–45 20 Yes 4 hoursMedoff (2000) 54 1 40+20 PS 120 Yes –PS=pressure support.Recruitment manoeuvres may be more effective <strong>in</strong> patientsventilated with relatively low levels of PEEP. Conversely, theymay be less effective and cause lung overdistension <strong>in</strong> patientswith already optimally recruited lungs—that is, with higherlevels of PEEP. Recruitment manoeuvres all <strong>in</strong>volve <strong>in</strong>creas<strong>in</strong>g<strong>in</strong>trathoracic pressure and therefore the risk of barotraumaand cardiovascular <strong>in</strong>stability. At present there are nopublished data from randomised studies to <strong>in</strong>dicate whetherrecruitment manoeuvres, of whatever form, <strong>in</strong>fluence outcome.Spontaneous breath<strong>in</strong>g dur<strong>in</strong>g positive pressureventilation (BiPAP, APRV)Two modes of ventilation commonly available on mechanicalventilators—biphasic airway pressure (BiPAP) and airwaypressure release ventilation (APRV)—allow spontaneousbreath<strong>in</strong>g to occur at any stage of the respiratory cycle. Inthese modes the ventilator cycles between an upper and lowerpressure at preset time <strong>in</strong>tervals. Spontaneous breath<strong>in</strong>g dur<strong>in</strong>gmechanical ventilation decreases <strong>in</strong>trathoracic pressureand improves V/Q match<strong>in</strong>g and cardiac output. 30 These theoreticalbenefits have resulted <strong>in</strong> more widespread use of theBiPAP mode, which provides a range of I:E ratios (APRVapplies a very short expiratory time), but aga<strong>in</strong> no data existconcern<strong>in</strong>g any <strong>in</strong>fluence on outcome.PRONE VENTILATIONProne position was reported to improve oxygenation <strong>in</strong>patients with ARDS as long ago as 1976. 31 The mechanism ofthe improvement <strong>in</strong> oxygenation on turn<strong>in</strong>g prone, seen <strong>in</strong>about two thirds of patients with ARDS, is complex. The<strong>in</strong>tuitive explanation that regional lung perfusion is primarilydependent on gravity lead<strong>in</strong>g to improved perfusion ofnon-consolidated lung on turn<strong>in</strong>g is not substantiated byresearch. In fact, perfusion to dorsal lung regions predom<strong>in</strong>ateswhatever the patient’s position, 32 and gravity accountsfor less than half the perfusion heterogeneity seen <strong>in</strong> eitherthe sup<strong>in</strong>e or prone position. 33Changes <strong>in</strong> regional pleuralpressure are more important. The gradient of pleural pressurefrom negative ventrally to positive dorsally <strong>in</strong> the sup<strong>in</strong>e positionis not completely reversed on turn<strong>in</strong>g prone, so that thedistribution of positive pressure ventilation is more homogenous<strong>in</strong> the prone position. 34Thus, recruitment of dorsallung appears to be the predom<strong>in</strong>ant mechanism of improvedoxygenation.Potential problems associated with prone position<strong>in</strong>g arepressure-<strong>in</strong>duced sk<strong>in</strong> damage, <strong>in</strong>creased venous pressure <strong>in</strong>the head (facial oedema), eye damage (corneal abrasions,ret<strong>in</strong>al and optic nerve ischaemia), dislodgment of endotrachealtubes and <strong>in</strong>travascular catheters, and <strong>in</strong>creased<strong>in</strong>tra-abdom<strong>in</strong>al pressure.A multicentre prospective randomised study of the proneposition for adult patients with acute respiratory failure wasundertaken <strong>in</strong> Italy. 35 Patients randomised to prone position<strong>in</strong>gwere assessed daily for the first 10 days and turned pronefor at least 6 hours if severity criteria were met. There were nodifferences <strong>in</strong> cl<strong>in</strong>ical outcome.Prone position<strong>in</strong>g is a useful adjunct to ventilation and mayhelp to improve oxygenation and pulmonary mechanics but,as yet, has not been shown to alter outcome <strong>in</strong> ARDS.HIGH FREQUENCY VENTILATIONThere has been a resurgence of <strong>in</strong>terest <strong>in</strong> high frequency ventilation(HFV, rate >60/m<strong>in</strong>) over the last few years. Initialenthusiasm had been tempered by practical difficulties andthe lack of cl<strong>in</strong>ical outcome data show<strong>in</strong>g any advantage overconventional mechanical ventilation. The recent cl<strong>in</strong>ical studiesof conventional ventilation demonstrat<strong>in</strong>g the advantagesof limited Vt and ma<strong>in</strong>tenance of lung volume have helped topromote <strong>in</strong>terest <strong>in</strong> HFV. The very low Vt (1–5 ml/kg) providedby HFV offers the possibility of ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g lung volume at ahigher po<strong>in</strong>t on the PV curve with less risk of caus<strong>in</strong>goverdistension. 36High frequency jet ventilation (HFJV) andhigh frequency oscillatory ventilation (HFOV) have been thetwo most commonly used methods.High frequency jet ventilation (HFJV)HFJV uses a high pressure gas jet delivered <strong>in</strong>to anendotracheal tube at high frequency (1–10 Hz). Other gas <strong>in</strong>the ventilator circuit is entra<strong>in</strong>ed produc<strong>in</strong>g a Vt of 2–5 ml/kgthat can be adjusted by alter<strong>in</strong>g the <strong>in</strong>spiratory time and/ordriv<strong>in</strong>g pressure. Dur<strong>in</strong>g HFJV, expiration occurs passively.Practical problems encountered are <strong>in</strong>adequate humidification,potential for gas trapp<strong>in</strong>g, difficulty <strong>in</strong> adjust<strong>in</strong>g ventilatorsett<strong>in</strong>gs, and the need for a specialised endotracheal tube.HFJV has been <strong>in</strong>vestigated <strong>in</strong> two large prospectiverandomised studies. In a study of 309 patients be<strong>in</strong>g ventilatedfor different causes of respiratory failure, the use of HFJVresulted <strong>in</strong> no significant outcome differences. 37Similarly, astudy of 113 patients at risk of ARDS had similar cl<strong>in</strong>ical outcomes<strong>in</strong> both patients ventilated conventionally and <strong>in</strong> those<strong>in</strong> whom HFJV was used. 38These studies did not <strong>in</strong>cluderecruitment manoeuvres that are now recognised to beimportant 39and were underpowered with respect to cl<strong>in</strong>icaloutcomes such as mortality. 40High frequency oscillatory ventilation (HFOV)HFOV differs from HFV <strong>in</strong> a number of important aspects.Tidal volume (1–3 ml/kg) is generated by the excursion of anoscillator with<strong>in</strong> a ventilator circuit similar to that used forCPAP and is varied by alter<strong>in</strong>g the frequency, I:E ratio, andoscillator amplitude. The use of an oscillator to generate Vtresults <strong>in</strong> active expiration. Mean airway pressure is adjustedby alter<strong>in</strong>g the fresh gas flow (bias flow) <strong>in</strong>to the circuit or theexpiratory pressure valve. Oxygenation is controlled byalter<strong>in</strong>g mean airway pressure or FiO 2.On <strong>in</strong>itiation of HFOV, lung recruitment is achieved by<strong>in</strong>creas<strong>in</strong>g mean airway pressure and monitor<strong>in</strong>g arterial oxygenation.Once optimal recruitment has occurred, meanairway pressures are reduced, tak<strong>in</strong>g advantage of the hysteresisof the lung pressure-volume relationship <strong>in</strong> order to preventalveolar overdistension. This process needs to be repeatedafter each episode of derecruitment. 40HFOV has been used extensively <strong>in</strong> neonates, and studiessuggest that it is associated with a lower <strong>in</strong>cidence of chroniclung disease than conventional ventilation. 41HFOV (with arecruitment protocol) was compared with conventionalmechanical ventilation <strong>in</strong> 70 paediatric patients with respiratoryfailure secondary to diffuse alveolar disease or large airleaks us<strong>in</strong>g a crossover (for treatment failure) study design. 42Overall outcomes were similar with the exception that


64 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>patients randomised to HFOV had a lower requirement forsupplemental oxygen at 30 days. After subgroup analysis,mortality was lower <strong>in</strong> patients treated with HFOV than <strong>in</strong>those treated with conventional mechanical ventilation only(6% v 40%).There are few data on the use of HFOV <strong>in</strong> adult patients. Inan observational study of 17 patients with ARDS, HFOV wasreported to be effective and safe. 43 A prospective randomisedcontrolled trial (Multicentre Oscillator ARDS Trial, MOAT) ofHFOV versus conventional ventilation <strong>in</strong> 148 adults withARDS was recently reported. 44The HFV group had an earlynon-susta<strong>in</strong>ed improvement <strong>in</strong> Pao 2/Fio 2ratio but there wereno significant differences <strong>in</strong> mortality at 30 days or 6 months.The authors concluded that HFOV was a safe and effectivealternative to conventional ventilation.LIQUID VENTILATIONARDS is associated with loss of surfactant, a consequent rise<strong>in</strong> surface tension, and alveolar collapse. Fill<strong>in</strong>g the lung withliquid removes the air-liquid <strong>in</strong>terface and supports alveoli,thus prevent<strong>in</strong>g collapse. Perfluorocarbons have been used <strong>in</strong>this approach because they have low surface tension and dissolveoxygen and carbon dioxide readily.Total liquid ventilation <strong>in</strong>volves fill<strong>in</strong>g the entire lung withliquid and us<strong>in</strong>g a special ventilator to oxygenate theperfluorocarbon, a technique that is both difficult and expensive.Partial liquid ventilation is a much more practicalalternative. The lung is filled to FRC with liquid and ventilatedwith a conventional mechanical ventilator. Although partialliquid ventilation is practical and safe, no randomisedprospective studies aga<strong>in</strong>st conventional management haveyet been published. 45 Further <strong>in</strong>formation on liquid ventilationcan be obta<strong>in</strong>ed from a recent review by Leonard. 46OTHER RESPIRATORY SUPPORTInhaled vasodilatorsThe use of <strong>in</strong>haled vasodilators <strong>in</strong> patients with ALI/ARDS isdescribed <strong>in</strong> chapter 10 and is not discussed further here.Extracorporeal gas exchangeDur<strong>in</strong>g extracorporeal membrane oxygenation (ECMO) venousblood is removed via a cannula <strong>in</strong> the <strong>in</strong>ferior vena cavaor right atrium, passed through a heart/lung mach<strong>in</strong>e, and isreturned to either the right atrium (veno-venous bypass) oraorta (veno-arterial bypass). In veno-venous bypass, pulmonaryand systemic haemodynamics are ma<strong>in</strong>ta<strong>in</strong>ed by thepatient’s own cardiovascular function. Veno-arterial bypassallows systemic haemodynamic support as well as gasexchange. Institution of ECMO allows ventilator pressuresand volumes to be decreased to prevent further ventilator<strong>in</strong>duced lung <strong>in</strong>jury. In addition, the reduction <strong>in</strong> <strong>in</strong>trathoracicpressure allows fluid removal to be carried out with less risk ofhaemodynamic <strong>in</strong>stability. A pumpless form of extracorporealgas exchange us<strong>in</strong>g arteriovenous cannulation has recentlybeen described. 47ECMO has proven mortality benefit <strong>in</strong> neonatal ARDS. Inadults a s<strong>in</strong>gle prospective randomised study failed to show asurvival advantage over conventional support. 48However,overall survival <strong>in</strong> both groups was extremely low and theresults are not applicable to current practice. ExtracorporealCO 2removal (ECCOR) <strong>in</strong>volves use of an extracorporeal venovenouscircuit with lower blood flows and oxygenation stilloccurr<strong>in</strong>g via the patient’s lungs. A randomised prospectivestudy of ECCOR compared with conventional support <strong>in</strong>patients with severe ARDS reported no significant difference<strong>in</strong> survival. 49 Several centres have recently reported observationalstudies show<strong>in</strong>g high survival rates <strong>in</strong> adult patientsmanaged with extracorporeal support (table 9.4). Theseencourag<strong>in</strong>g survival rates should be <strong>in</strong>terpreted, however, <strong>in</strong>50 51the context of improved survival without ECMO. ATable 9.4 Recent observational studies report<strong>in</strong>gsurvival of patients with ARDS managed withextracorporeal membrane oxygenation (ECMO)Reference n Survival (%)Lewandowski (1997) 55 122 75*Ullrich (1999) 56 13 62*Bartlett (2000) 57 86 61L<strong>in</strong>den (2000) 58 16 76*Patients managed with a protocol that <strong>in</strong>cluded ECMO if necessary.randomised prospective controlled study of ECMO <strong>in</strong> adultpatients is currently underway <strong>in</strong> Leicester, UK (CESAR trial).CONCLUSIONThe current data relat<strong>in</strong>g to conventional ventilation <strong>in</strong> ARDSsuggest that high tidal volumes (12 ml/kg) with high plateaupressure (more than 30–35 cm H 2O) are deleterious and that astrategy aimed at prevent<strong>in</strong>g overdistension by decreas<strong>in</strong>gtidal volume to 6 ml/kg and limit<strong>in</strong>g peak pressure to


Ventilatory management of ALI/ARDS 6517 Brower RG, Shanholtz CB, Fessler HE, et al. Prospective, randomised,controlled cl<strong>in</strong>ical trial compar<strong>in</strong>g traditional versus reduced tidal volumeventilation <strong>in</strong> acute respiratory distress syndrome patients. Crit <strong>Care</strong> Med1999;27:1492–8.18 Amato MBP, Barbas CSV, Medeiros DM, et al. Effect of aprotective-ventilation strategy on mortality <strong>in</strong> the acute respiratory distresssyndrome. N Engl J Med 1998;338:347–54.19 Nahum A, Ravenscraft SA, Nakos G, et al. Tracheal gas <strong>in</strong>sufflationdur<strong>in</strong>g pressure-control ventilation. Effect of catheter position, diameter,and flow rate. Am Rev Respir Dis 1992;146:965–73.20 Hoffman LA, Miro AM, Tasota FJ, et al. Tracheal gas <strong>in</strong>sufflation. Limitsof efficacy <strong>in</strong> adults with acute respiratory distress syndrome. Am J RespirCrit <strong>Care</strong> Med 2000;162:387–92.21 Eichacker PQ, Gerstenberger EP, Banks SM, et al. Meta-analysis ofacute lung <strong>in</strong>jury and acute respiratory distress syndrome trials test<strong>in</strong>glow tidal volumes. Am J Respir Crit <strong>Care</strong> Med 2002;166:1510–4.22 Brower RG, Mathay M, Schoenfeld D. Meta-analysis of acute lung <strong>in</strong>juryand acute respiratory distress syndrome trials [letter]. Am J Respir Crit<strong>Care</strong> Med 2002;166:1515–7.23 Rappaport SH, Shp<strong>in</strong>er R, Yoshihara G, et al. Randomized, prospectivetrial of pressure-limited versus volume-controlled ventilation <strong>in</strong> severerespiratory failure. Crit <strong>Care</strong> Med 1994;22:22–32.24 Lessard MR, Guerot E, Lor<strong>in</strong>o H, et al. Effects of pressure-controlledventilation on respiratory mechanics, gas exchange and haemodynamics<strong>in</strong> patients with adult respiratory distress syndrome. Anesthesiology1994;80:983–91.25 Esteban A, Alia I, Gordo F, et al. Prospective randomised trialcompar<strong>in</strong>g pressure-controlled ventilation and volume-controlledventilation <strong>in</strong> ARDS. Spanish Lung Failure Collaborative Group. Chest2000;117:1690–6.26 Ranieri VM, Eissa NT, Corbeil et al. Effects of positive end-expiratorypressure on alveolar recruitment and gas exchange <strong>in</strong> patients with theadult respiratory distress syndrome. Am Rev Respir Dis1991;144:544–51.27 Richard JC, Brochard L, Vandelet P, et al. Respective effects ofend-expiratory and end-<strong>in</strong>spiratory pressures on alveolar recruitment <strong>in</strong>acute lung <strong>in</strong>jury. Crit <strong>Care</strong> Med 2003;31:89–92.28 Puybasset L, Gusman P, Muller JC, et al. Regional distribution of gasand tissue <strong>in</strong> acute respiratory distress syndrome. III. Consequences forthe effects of positive end-expiratory pressure. CT Scan ARDS StudyGroup. Adult <strong>Respiratory</strong> Distress Syndrome. Intensive <strong>Care</strong> Med2000;26:1215–27.29 Pelosi P, Cadr<strong>in</strong>gher P, Bott<strong>in</strong>o N, et al. Sigh <strong>in</strong> acute respiratory distresssyndrome. Am J Respir Crit <strong>Care</strong> Med 1999;159:872–80.30 Putensen C, Mutz NJ, Putensen-Himmer G, et al. Spontaneous breath<strong>in</strong>gdur<strong>in</strong>g ventilatory support improves ventilation-perfusion distributions <strong>in</strong>patients with acute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong>Med 1999;159:1241–8.31 Piehl MA, Brown RS. Use of extreme position changes <strong>in</strong> respiratoryfailure. Crit <strong>Care</strong> Med 1976;4:13–4.32 Glenny RW, Lamm WJ, Albert RK, et al. Gravity is a m<strong>in</strong>or determ<strong>in</strong>antof pulmonary blood flow distribution. J Appl Physiol 1991;71:620–9.33 Jones AT, Hansell DM, Evans TW. Pulmonary perfusion <strong>in</strong> sup<strong>in</strong>e andprone positions: an electron-beam computed tomography study. J ApplPhysiol 2001;90:1342–8.34 Lamm WJ, Graham MM, Albert RK. Mechanism by which the proneposition improves oxygenation <strong>in</strong> acute lung <strong>in</strong>jury. Am J Respir Crit <strong>Care</strong>Med 1994;150:184–93.35 Gatt<strong>in</strong>oni L. Presentation at the European Society of Intensive <strong>Care</strong>Medic<strong>in</strong>e 13th Annual Congress, Rome 2000.36 Froese AB, McCullough PR, Siguria M, et al. Optimiz<strong>in</strong>g alveolarexpansion prolongs the effectiveness of exogenous surfactant therapy <strong>in</strong>the adult rabbit. Am Rev Respir Dis 1993;148:569–77.37 Carlon GC, Howland WS, Ray C, et al. High frequency jet ventilation: aprospective, randomised evaluation. Chest 1983;84:551–9.38 Hurst JM, Branson RD, Davis K, et al. Comparison of conventionalmechanical ventilation and high-frequency ventilation: a prospectiverandomised trial <strong>in</strong> patients with respiratory failure. Ann Surg1990;211:486–91.39 Herridge MS, Slutsky AS, Colditz GA. Has high-frequency ventilationbeen <strong>in</strong>appropriately discarded <strong>in</strong> adult acute respiratory distresssyndrome? Crit <strong>Care</strong> Med 1998;26:2073–7.40 Froese AB. High-frequency oscillatory ventilation for adult respiratorydistress syndrome: let’s get it right this time. Crit <strong>Care</strong> Med1997;25:906–8.41 Riphagen S, Bohn D. High frequency oscillatory ventilation. Intensive<strong>Care</strong> Med 1999;25:1459–62.42 Arnold JH, Hanson JH, Toro-Figuero LO, et al. Prospective, randomisedcomparison of high-frequency oscillatory ventilation and conventionalmechanical ventilation <strong>in</strong> pediatric respiratory failure. Crit <strong>Care</strong> Med1994;22:1530–9.43 Fort P, Farmer C, Westerman J, et al. High-frequency oscillatoryventilation for adult respiratory distress syndrome: a pilot study. Crit <strong>Care</strong>Med 1997;25:937–47.44 Derdak S, Mehta S, Stewart TE, et al. High-frequency oscillatoryventilation for acute respiratory distress syndrome <strong>in</strong> adults: arandomised, controlled trial. Am J Respir Crit <strong>Care</strong> Med 2002;6:801–8.45 Hirschl RB, Conrad S, Kaiser R, et al. Partial liquid ventilation <strong>in</strong> adultpatients with ARDS: a multicenter phase I-II trail. Ann Surg1998;228:692–700.46 Leonard RC. Liquid ventilation. Anaesth Int <strong>Care</strong> 1998;26:11–21.47 Reng M, Philipp A, Kaiser M, et al. Pumpless extracorporeal lung assistand adult respiratory distress syndrome. Lancet 2000;356:219–20.48 Zapol WM, Snider MT, Hill JD, et al. Extracorporeal membraneoxygenation <strong>in</strong> severe acute respiratory failure. A randomisedprospective study. JAMA 1979;242:2193–6.49 Morris AH, Wallace CJ, Menlove RL, et al. Randomized cl<strong>in</strong>ical trial ofpressure-controlled <strong>in</strong>verse ratio ventilation and extracorporeal CO 2removal for adult respiratory distress syndrome. Am J Respir Crit <strong>Care</strong>Med 1994;149:295–305.50 Abel SJ, F<strong>in</strong>ney SJ, Brett SJ, et al. Reduced mortality <strong>in</strong> association withthe acute respiratory distress syndrome (ARDS). Thorax 1998;53:292–4.51 Millberg JA, Davis DR, Ste<strong>in</strong>berg KP, et al. Improved survival of patientswith acute respiratory distress syndrome (ARDS): 1983–1993. JAMA1995;273:306–9.52 International consensus conferences <strong>in</strong> <strong>in</strong>tensive care medic<strong>in</strong>e.Ventilator-associated lung <strong>in</strong>jury <strong>in</strong> ARDS. Intensive <strong>Care</strong> Med1999;25:1444–52.53 Lap<strong>in</strong>sky SE, Aub<strong>in</strong> M, Mehta S, et al. Safety and efficacy of asusta<strong>in</strong>ed <strong>in</strong>flation for alveolar recruitment <strong>in</strong> adults with respiratoryfailure. Intensive <strong>Care</strong> Med 1999;25:1297–301.54 Medoff BD, Harris RS, Kesselman H, et al. Use of recruitment maneuversand high-positive end-expiratory pressure <strong>in</strong> a patient with acuterespiratory distress syndrome. Crit <strong>Care</strong> Med 2000;28:1210–6.55 Lewandowski K, Rossa<strong>in</strong>t R, Pappert D, et al. High survival rate <strong>in</strong> 122patients managed accord<strong>in</strong>g to a cl<strong>in</strong>ical algorithm <strong>in</strong>clud<strong>in</strong>gextra-corporeal membrane oxygenation. Intensive <strong>Care</strong> Med1997;23:819–35.56 Ullrich R, Larber C, Roder G, et al. Controlled airway pressure therapy,nitric oxide <strong>in</strong>halation, prone position, and ECMO as components of an<strong>in</strong>tegrated approach to ARDS. Anesthesiology 1999;91:1577–86.57 Bartlett RH, Roloff DW, Custer JR, et al. Extracorporeal life support. TheUniversity of Michigan Experience. JAMA 2000;283:904–8.58 L<strong>in</strong>den V, Palmer K, Re<strong>in</strong>hard J, et al. High survival <strong>in</strong> adult patients withacute respiratory syndrome treated by extracorporeal membraneoxygenation, m<strong>in</strong>imal sedation, and pressure supported ventilation.Intensive <strong>Care</strong> Med 2000;26:1630–7.


10 Non-ventilatory strategies <strong>in</strong> acute respiratory distresssyndromeJ Cranshaw, MJDGriffiths, T W Evans.............................................................................................................................Our understand<strong>in</strong>g of the pathophysiologyand management of the acute respiratorydistress syndrome (ARDS) has improvedimmensely s<strong>in</strong>ce its orig<strong>in</strong>al description, butpharmacotherapies have proved disappo<strong>in</strong>t<strong>in</strong>g <strong>in</strong>cl<strong>in</strong>ical trials. Several reasons have been proposedfor this failure.• There are no good experimental models ofARDS so that drugs may not have the desiredeffect or produce unacceptable side effectswhen used cl<strong>in</strong>ically.• The <strong>in</strong>flammatory cascades that cause sepsisand ARDS are characterised by widespreadredundancy so it is unlikely that a s<strong>in</strong>gle agentcould reverse or term<strong>in</strong>ate such complex processes.• Although a drug may improve pulmonaryfunction, it may not alter outcome. Fewer than5% of patients with ARDS die of respiratoryfailure; the majority suffer from multiple organfailure and succumb after withdrawal ofsupport.• Enroll<strong>in</strong>g patients with ARDS <strong>in</strong> cl<strong>in</strong>ical trialsus<strong>in</strong>g the American-European Consensus Conferencedef<strong>in</strong>ition 1 ignores the heterogeneity ofthe disease. In pathological terms, the acuteexudative and fibroproliferative phases presentdist<strong>in</strong>ct targets for <strong>in</strong>tervention, mak<strong>in</strong>g thetim<strong>in</strong>g of drug adm<strong>in</strong>istration (after diseaseonset) crucial. The primary cause(s) of ARDS,the patient’s age and medical history all affectprognosis and possibly drug responsiveness ofthe condition.Recent reports suggest that the mortality associatedwith ARDS may be fall<strong>in</strong>g, probablybecause of advances <strong>in</strong> support<strong>in</strong>g critically illpatients. 2 This trend may <strong>in</strong>crease the number ofpotential survivors and the w<strong>in</strong>dow of opportunityfor pharmacological manipulation of lung<strong>in</strong>jury. Thus, drugs that have been apparentfailures <strong>in</strong> terms of mortality may still have auseful role to play, either <strong>in</strong> comb<strong>in</strong>ation withother agents or <strong>in</strong> subgroups of ARDS patientsdef<strong>in</strong>ed either by their underly<strong>in</strong>g condition or bytheir stage of lung <strong>in</strong>jury. This chapter reviews themajor pharmacological approaches to treat<strong>in</strong>gARDS <strong>in</strong> the context of modern supportive care(fig 10.1). We conclude with a proposal for futurestrategies <strong>in</strong> the non-ventilatory management ofARDS.INTRAVENOUS FLUID MANAGEMENTPatients with ARDS often have cardiovasculardysfunction caused by systemic <strong>in</strong>flammationthat is commonly associated with sepsis. Hence,myocardial depression, abnormal vascular tone,and permeability contribute to abnormal tissueoxygenation and ultimately organ failure. Inpractice, achiev<strong>in</strong>g adequate organ perfusion mayoccur at the cost of <strong>in</strong>creas<strong>in</strong>g extravascular watermanifest<strong>in</strong>g as an exacerbation of pulmonaryoedema. Low extravascular lung water levels areassociated with better oxygenation and a lowermortality <strong>in</strong> patients with ARDS <strong>in</strong> retrospectivestudies. 34There is limited prospective evidencethat target<strong>in</strong>g lower extravascular lung waterus<strong>in</strong>g diuretics with vasopressors to supportorgan perfusion reduces the time required on aventilator. 56 Our policy is to keep the <strong>in</strong>travascularvolume as low as possible while ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>gan adequate cardiac <strong>in</strong>dex and mean arterialpressure.INHALED VASODILATORSNitric oxide (NO) is a free radical gas producedconstitutively <strong>in</strong> the lung by nitric oxide synthasefrom L-arg<strong>in</strong><strong>in</strong>e, NADPH, and oxygen. Endothelialcells constitutively release NO, caus<strong>in</strong>g pulmonaryvasodilation primarily via the secondarymessenger cyclic guanos<strong>in</strong>e monophosphate.ARDS is characterised by ventilation-perfusionmismatch<strong>in</strong>g which produces arterial hypoxaemiathat may <strong>in</strong> part be caused by disorderedendogenous NO activity. Patients with ARDScommonly have mild pulmonary hypertension.Any <strong>in</strong>haled vasodilator can augment hypoxicpulmonary vasoconstriction by selectively vasodilat<strong>in</strong>gvessels associated with ventilated alveoli toimprove oxygenation (fig 10.2).Improved oxygenation and direct vascularsmooth muscle relaxation by NO also reduce pulmonaryvascular resistance (PVR). Vasoconstrictionby hypoxia, hypercapnia, thromboxane A 2,and angiotens<strong>in</strong> II can all be partially reversed by<strong>in</strong>haled NO, although the PVR of normal volunteersis not affected. 7Reduc<strong>in</strong>g PVR and consequentialimprovements <strong>in</strong> right ventricular functionmay benefit some patients with ARDS.However, NO does not <strong>in</strong>crease cardiac output <strong>in</strong>the majority. 8 Reduced arteriolar and venous tonemay lower capillary pressure, reduc<strong>in</strong>g leakageand further improv<strong>in</strong>g gas exchange. Unfortunately,<strong>in</strong> patients with pulmonary hypertensionassociated with impaired left ventricular function,pulmonary vascular relaxation may also<strong>in</strong>crease pulmonary oedema.Endothelial NO also <strong>in</strong>hibits platelet aggregationand neutrophil adhesion that are likelymediators of lung <strong>in</strong>jury. Although the importanceof these actions <strong>in</strong> ARDS is uncerta<strong>in</strong>,<strong>in</strong>haled NO has been used immediately after lungtransplantation to reduce ischaemia-reperfusion<strong>in</strong>jury. 9NO quickly scavenges reactive oxygenspecies (ROS) <strong>in</strong>clud<strong>in</strong>g the superoxide anion toproduce a less reactive but still potentially harmfulproduct, namely peroxynitrite. Although ROS


Non-ventilatory strategies <strong>in</strong> ARDS 67AECC def<strong>in</strong>itionPrimary diagnosisDef<strong>in</strong>itive treatment ofprimary pathologyVentilatory strategyFluid balanceHaemodynamic managementNutritionPAC/echocardiogramCT thoraxBAL, <strong>in</strong>fection screenNoOxygenationat FiO 2 0.5SaO 2 >88%YesProne position<strong>in</strong>gInhaled vasodilatorStabiliseConsiderSurfactant replacementLiquid ventilationExtracorporeal gas exchangeTransplantationMethylprednisolone(exclude <strong>in</strong>fection)Wean<strong>in</strong>g protocolFigure 10.1 Suggested treatment algorithm for acute lung <strong>in</strong>jury (ALI) and ARDS. AECC=American-European Consensus Conference 1993;PAC=pulmonary artery catheter; CT=computed tomography; BAL=bronchoalveolar lavage; FiO 2=fractional <strong>in</strong>spired oxygen concentration;SaO 2=arterial oxygen saturation.are usually kept at low levels <strong>in</strong> lung tissue by antioxidantsand dismutases, these protective systems may be overwhelmeddur<strong>in</strong>g ARDS. 10Peroxynitrite oxidises and nitrosylatesprote<strong>in</strong>s, nucleic acids and lipids, <strong>in</strong>clud<strong>in</strong>g essentialcomponents of the surfactant system. However, thecl<strong>in</strong>ical significance of peroxynitrite production is unknown.Approximately 60% of patients with ARDS or acute lung<strong>in</strong>jury (ALI) of all causes respond to <strong>in</strong>haled NO, <strong>in</strong>creas<strong>in</strong>gtheir PaO 2by more than 20%. 11 The effect can frequently be12 13seen <strong>in</strong> less than 10 m<strong>in</strong>utes or may take several hours.However, <strong>in</strong> several trials the oxygenation of control groupshas risen to meet that of NO treated patients between 24 hours11 13 14and 4 days. The dose-response relationship between<strong>in</strong>haled NO and arterial oxygenation shows considerable<strong>in</strong>ter<strong>in</strong>dividual variation. 15Currently there are no <strong>in</strong>dicatorsthat will predict the response. Maximal improvement <strong>in</strong> oxygenationis sometimes achieved with 1–2 parts per million(ppm) and occurs at less than 10 ppm <strong>in</strong> most patients. Maximalreduction <strong>in</strong> pulmonary artery pressure is usuallyobta<strong>in</strong>ed between 10 and 40 ppm, with no benefit and possibletoxicity at doses greater than 80 ppm. United K<strong>in</strong>gdomguidel<strong>in</strong>es suggest a maximum dose of 40 ppm. 16Intra<strong>in</strong>dividualvariation <strong>in</strong> response with time is also significantand may be <strong>in</strong>fluenced by lung recruitment, co-existentpathology, or the resolution of <strong>in</strong>flammation. Cl<strong>in</strong>ically, it issometimes difficult to stop <strong>in</strong>haled NO without “rebound”pulmonary hypertension and hypoxaemia. The last 1–2 ppmmay have to be weaned especially slowly.The systemic effects of <strong>in</strong>haled NO are negligible due to therapid strong comb<strong>in</strong>ation of NO with haemoglob<strong>in</strong> to formmethaemoglob<strong>in</strong>. This is normally reduced to functionalhaemoglob<strong>in</strong> and NO is ultimately converted to soluble NO 3.Methaemoglob<strong>in</strong>aemia produces a functional anaemia and aleft shift <strong>in</strong> the dissociation curve, but rarely causes a cl<strong>in</strong>icalproblem. Normal levels of methaemaglob<strong>in</strong> are less than 2%and values less than 5% usually do not need treatment. NOreacts slowly with oxygen and water to form toxic NO 2, nitrousand nitric acids. These damage the lung at concentrations aslow as 2 ppm. The reaction rate is proportional to thefractional <strong>in</strong>spired oxygen concentration (FiO 2) and the squareof the NO concentration. Thus, the contact time andconcentrations of the gases should be kept to a m<strong>in</strong>imum.With proper monitor<strong>in</strong>g, delivery systems, and NO doses ofless than 40 ppm, NO 2is not a significant problem. Deliverysystems that add NO “upstream” of the ventilator allow longermix<strong>in</strong>g with oxygen and are not recommended. Cont<strong>in</strong>uous“downstream” addition of NO may allow NO to collect <strong>in</strong> the<strong>in</strong>spiratory limb of the circuit dur<strong>in</strong>g the expiratory phase ofsome systems. Synchronised NO delivery dur<strong>in</strong>g <strong>in</strong>spirationmay be the optimum mode of delivery. NO conta<strong>in</strong>ed <strong>in</strong>exhaled gas should be absorbed before release.Randomised controlled trials <strong>in</strong> patients with ARDS haveshown that, while <strong>in</strong>haled NO temporarily improves oxygenationand reduces pulmonary artery pressure <strong>in</strong> the majority, itsuse is not associated with an improved outcome (table 10.1).Inhaled NO is therefore not a standard treatment for ARDS.However, patients with severe refractory hypoxaemia and<strong>in</strong>adequate right ventricular function secondary to pulmonaryhypertension may benefit from <strong>in</strong>haled NO. NO may also protectpatients whose oxygenation might otherwise dependupon a potentially damag<strong>in</strong>g ventilatory strategy. In smallstudies the pulmonary vasoconstrictor almitr<strong>in</strong>e improvedoxygenation <strong>in</strong> patients with ARDS. 17It has been suggestedthat low dose <strong>in</strong>travenous almitr<strong>in</strong>e potentiates hypoxicpulmonary vasoconstriction and the comb<strong>in</strong>ation of almitr<strong>in</strong>eand <strong>in</strong>haled NO may improve oxygenation synergistically <strong>in</strong>patients with ARDS. 18Prostacycl<strong>in</strong> (PGI 2) is an endothelium-derived prostagland<strong>in</strong>vasodilator that <strong>in</strong>hibits platelet aggregation andneutrophil adhesion. Its mechanism of action differs from NO<strong>in</strong> that smooth muscle relaxation is associated with a rise <strong>in</strong>cytoplasmic cyclic adenos<strong>in</strong>e monophosphate. Its half life isonly 2–3 m<strong>in</strong>utes but it is not metabolised by the lung, sowhen adm<strong>in</strong>istered <strong>in</strong>travenously, PGI 2lowers pulmonaryvascular resistance but may also <strong>in</strong>crease <strong>in</strong>trapulmonaryshunt<strong>in</strong>g and cause systemic hypotension. 15However, nebulisedPGI 2(0–50 ng/kg/m<strong>in</strong>) 19 20 or alprostadil (PGE 1,20–80 µg/h) 21 produce equivalent effects to <strong>in</strong>haled NO withm<strong>in</strong>imal systemic side effects and without measurable plateletdysfunction, but there have been no large randomised trials of


68 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>↓ V/Q↓ SaO 2Hypoxic pulmonary vasodilationIntravenous vasodilatorInhaled vasodilator↓ V/Q↓ SaO 2NOPGI 2PGI 2SNPFigure 10.2 Effect of <strong>in</strong>travenous and <strong>in</strong>haled vasodilators <strong>in</strong> lung <strong>in</strong>jury. V/Q=ventilation:perfusion ratio; SaO 2=arterial oxygen saturation;PGI 2=prostacycl<strong>in</strong>; SNP=sodium nitroprusside; NO=nitric oxide.these therapies. Nevertheless, the relatively simple deliverysystem, harmless metabolites, and no requirement for specialmonitor<strong>in</strong>g make nebulised PGI 2an attractive alternative to<strong>in</strong>haled NO, despite its expense.CORTICOSTEROIDSCorticosteroids reduce the production of a great number of<strong>in</strong>flammatory and profibrotic mediators by many mechanisms.The importance of steroid therapy to the resolution ofTable 10.1AuthorRandomised controlled trials of <strong>in</strong>haled nitric oxide (NO) <strong>in</strong> patients with ARDSNo ofpatients Diagnosis Bl<strong>in</strong>dedInhaledNO dose(ppm) Duration OutcomeLund<strong>in</strong> et al(1999) 76 260 ALI (American-European ConsensusConference) and 18–96 hours ventilationwith PaO 2 /FiO 2 10cm H 2 O and I:E 1:2–2:1Dell<strong>in</strong>ger et al(1998) 11 177 ARDS (American-European ConsensusConference) with<strong>in</strong> 72 hours of onsetand PEEP at least 8 cm H 2 O and FiO 2>0.5Michael et al40 ARDS (American-European Consensus(1998) 13 Conference) and FiO 2 at least 0.8 for12 hours or 0.65 for 24 hoursNo 2–40 30 days 180 randomised responded to NO. Thefrequency of reversal of ALI did notdiffer from controls. Development ofsevere respiratory failure less (2.2% v10.3%) <strong>in</strong> NO treated group. Mortalitynot altered (44% v 40% control).Yes 1.25–80 28 days PaO 2 <strong>in</strong>creased >20% <strong>in</strong> first 4 hours <strong>in</strong>60% of patients treated with NO and24% of controls. FiO 2 and <strong>in</strong>tensity ofventilation could be reduced <strong>in</strong> first 4days. No difference <strong>in</strong> mortality (30% v32–38% <strong>in</strong> NO treated groups).No 5–20 3 days NO improved PAO 2 /FiO 2 by at least20% and allowed a decrease <strong>in</strong> FiO 2 ofat least 0.15 only <strong>in</strong> the first 24 hours <strong>in</strong>more treated patients than controls.Troncy(1998) 14 30 Murray score at least 2.5 No 0.5–40 30 days NO improved oxygenation only <strong>in</strong> thefirst 24 hours <strong>in</strong> more treated patientsthan controls. Mortality (60% v 67% <strong>in</strong>control) not altered.


Non-ventilatory strategies <strong>in</strong> ARDS 69Table 10.2 Published trials of short term, high dose steroid therapy <strong>in</strong> patients with ARDSControlgroup Randomised Prospective Bl<strong>in</strong>ded Methylprednisolone dose Duration OutcomeNo ofpatients DiagnosisAuthor32 days Improvement <strong>in</strong> LIS, MODS, reducedmortalityMeduri et al (1998) 28 24 Late ARDS Yes Yes Yes Yes 2 mg/kg load then 0.5 mg/kg 6 hrlyreduced weekly to 1 mg/kg/day, 0.5mg/kg/day then 0.15 mg/kg/day3–6 weeks 81% survivalNo No Yes No 125–250 mg 6 hrly for 3–4 days reduc<strong>in</strong>gby 50% every 2–3 daysHooper et al (1996) 77 26 Established ARDS of >3days with cause resolvedReduction <strong>in</strong> plasma and BAL<strong>in</strong>flammatory cytok<strong>in</strong>esMeduri et al (1995) 78 9 Late ARDS No No Yes No 200 mg bolus, 2–3 mg/kg/day Until extubation(average 6 weeks)Biffl et al (1995) 79 6 Prolonged ARDS fail<strong>in</strong>g No No Yes No 1–2 mg/kg 6 hrly 13–42 days 83% survival; improved LIS andconventional therapyPaO 2 /FiO 2Meduri et al (1994) 80 25 Late ARDS No No Yes No 200 mg bolus, 2–3 mg/kg/day Until extubation Reduction <strong>in</strong> LIS, improved PaO 2 /FiO 2Meduri et al (1991) 81 8 Late ARDS No No No No 2 mg/kg bolus, 2–3 mg/kg 6 hrly Until extubation Reduction <strong>in</strong> LISYes Yes Yes 30 mg/kg 6 hrly 24 h Term<strong>in</strong>ated early after publication of26 ; no alteration <strong>in</strong> LISYes (mannitolplacebo)Luce et al (1988) 82 75 Culture positive septicshockYes Yes Yes Yes 30 mg/kg 6 hrly 24 h No change <strong>in</strong> <strong>in</strong>cidence of ARDS;mortality of ARDS higher; lessfrequent ARDS reversalBone et al (1987) 26 381 Severe sepsis and septicshockBernard et al (1987) 25 99 ARDS Yes Yes Yes Yes 30 mg/kg 6 hrly 24 h No mortality benefitWeigelt et al (1985) 24 81 ARDS Yes Yes Yes Yes 30 mg/kg 6 hrly 48 h No mortality benefit; <strong>in</strong>creased<strong>in</strong>fection rateYes Yes Yes No 30 mg/kg 6 hrly 3 days Increased mortalityLucas et al (1981) 23 114 Post-<strong>in</strong>jury hypovolaemicshock lungSladen (1976) 22 10 Shock lung No No Yes No 30 mg/kg 6 hrly 48 h Improved mortalityLIS=lung <strong>in</strong>jury score; MODS=multiple organ dysfunction syndrome.lung <strong>in</strong>flammation <strong>in</strong> animal models became apparent <strong>in</strong> the1980s. Unfortunately, trials of short term, high dose steroidtherapy (for example, methylprednisolone 30 mg/kg 6 hourlyfor 24 hours) failed to show an improvement <strong>in</strong> mortality ofpatients at risk of or with early ARDS associated with sepsis,aspiration, and trauma (table 10.2). 22–26In fact, some trialsshowed <strong>in</strong>creased risk of <strong>in</strong>fection, lower rates of reversal ofARDS, and <strong>in</strong>creased mortality associated with the use of highdose steroids. Meta-analyses of available trials emphasise theadverse effects of these agents <strong>in</strong> patients with sepsis.However, the use of steroids <strong>in</strong> patients with late ARDS(7–14 days from diagnosis) has not been abandoned. Recentdata suggest that <strong>in</strong>flammation and fibrosis <strong>in</strong> the lung aredist<strong>in</strong>ct and thus <strong>in</strong>dependently manipulable processes. 27There is also cl<strong>in</strong>ical evidence that steroids favourably modifythe fibroproliferative phase of ARDS. In the 1990s lower dose(2–8 mg/kg/day methyprednisolone), longer term (2–6 weeks)corticosteroid treatment was used <strong>in</strong> patients with ARDS ofover 10 days duration. Mortality fell to approximately 20% <strong>in</strong>some uncontrolled series but complications attributable tosteroids were not <strong>in</strong>frequent and <strong>in</strong>cluded sepsis, pneumonia,wound <strong>in</strong>fection, gastric ulceration, and diabetes. A trial us<strong>in</strong>gthis approach randomised 24 patients with “unresolv<strong>in</strong>g”ARDS of more than 7 days duration to methylprednisolone(2 mg/kg load then 2 mg/kg/day <strong>in</strong> four divided dosesreduc<strong>in</strong>g weekly to 1 mg/kg/day, then 0.5 mg/kg/day, then0.15 mg/kg/day). 28 None of the 16 patients <strong>in</strong> the steroid groupdied compared with five of eight orig<strong>in</strong>ally given placebo.Steroid therapy was associated with improved oxygenationand successful extubation. Eligible patients were exam<strong>in</strong>ed forpulmonary <strong>in</strong>fection by bronchoalveolar lavage at day 5 andall febrile patients received a broad septic screen before trialentry. Documented <strong>in</strong>fections were treated with appropriateantibiotics for at least 3 days before steroids were adm<strong>in</strong>istered.Despite these precautions, 75% of patients <strong>in</strong> both armsof the trial suffered new sepsis. Although the results arepromis<strong>in</strong>g, the design and <strong>in</strong>terpretation of this trial haveproved contentious. 29The larger NIH ARDS Network LateSteroid Rescue Study may provide sounder evidence forprescrib<strong>in</strong>g low dose steroids <strong>in</strong> late ARDS.SURFACTANTType II alveolar cells synthesise and recycle surfactantphospholipids and prote<strong>in</strong>s. Surfactant lowers alveolar surfacetension and prevents collapse at low lung volumes. The sameeffects reduce the hydrostatic pressure gradient favour<strong>in</strong>gfluid movement <strong>in</strong>to the alveolar space. Surfactant also hasanti-<strong>in</strong>flammatory and antimicrobial properties. Dur<strong>in</strong>gARDS, surfactant activity may be deficient because of reducedproduction, <strong>in</strong>creased removal with recurrent alveolar collapsedur<strong>in</strong>g ventilation, abnormal composition and, importantly,dysfunction caused by plasma prote<strong>in</strong>s, 30 ROS, and proteases<strong>in</strong> the flooded alveolar space. 31Various preparations, doses, adm<strong>in</strong>istration regimens, anddelivery techniques have been proposed. Phospholipids aloneare <strong>in</strong>ferior to composites of lipid and surfactant prote<strong>in</strong>s. Theamount of drug required varies with the type of adm<strong>in</strong>istrationtechnique—<strong>in</strong>tratracheal delivery, aerosolisation <strong>in</strong> ventilatorgas, and direct bronchoscopic <strong>in</strong>stillation. Simultaneoussegmental lavage has been comb<strong>in</strong>ed with the latter approachand this process itself may benefit some patients. Theoptimum tim<strong>in</strong>g and duration of surfactant therapy is still tobe determ<strong>in</strong>ed.A phase III cl<strong>in</strong>ical trial of one aerosolised preparation <strong>in</strong>sepsis <strong>in</strong>duced ARDS has been conducted on a background ofimproved oxygenation <strong>in</strong> previous reports. 32No significanteffect was seen on oxygenation, duration of ventilation, orsurvival with up to 5 days of cont<strong>in</strong>uous treatment beg<strong>in</strong>n<strong>in</strong>gwith<strong>in</strong> 48 hours of ventilation. However, this study has beencriticised because the compound used conta<strong>in</strong>ed no prote<strong>in</strong>


70 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>and aerosolisation may have delivered less than 5 mg/kg/dayof phospholipid when <strong>in</strong>vestigations suggest <strong>in</strong>stillation of300 mg/kg/day may be required. Bov<strong>in</strong>e surfactant adm<strong>in</strong>istered<strong>in</strong>tratracheally <strong>in</strong> a smaller trial has shown that higherdoses (100 mg/kg qds) are required to alter alveolar surfactantcomposition. 33 This dose improved oxygenation over 120 hoursand a trend was observed towards reduced mortality at 28days. Total bronchopulmonary segmental lavage with 30 mlper segment of a synthetic prote<strong>in</strong> conta<strong>in</strong><strong>in</strong>g surfactant wassafe and effective at improv<strong>in</strong>g oxygenation with<strong>in</strong> 72 hours ofthe onset of sepsis <strong>in</strong>duced ARDS. 34 Despite these trials, theuse of surfactant <strong>in</strong> ARDS rema<strong>in</strong>s experimental. Successfuluse of surfactant <strong>in</strong> animal models of lung <strong>in</strong>jury and <strong>in</strong> neonatalrespiratory distress syndrome suggests that efficientadm<strong>in</strong>istration of an effective substitute could be beneficial <strong>in</strong>ARDS. The goals of treatment would <strong>in</strong>clude both improvedgas exchange and protection aga<strong>in</strong>st ventilator <strong>in</strong>duced lung<strong>in</strong>jury.IMMUNONUTRITIONAvoid<strong>in</strong>g nutritional depletion while deliver<strong>in</strong>g a high fat, lowcarbohydrate diet to reduce carbon dioxide production andventilatory demand is appropriate for patients with ARDS. 35Immunonutrition aims to <strong>in</strong>fluence <strong>in</strong>flammation positivelyand to protect gastro<strong>in</strong>test<strong>in</strong>al <strong>in</strong>tegrity. However, supply<strong>in</strong>genteral nutrition of any type may stimulate gastro<strong>in</strong>test<strong>in</strong>aland pulmonary IgA defence mechanisms. 36In vitro, dietaryadditives improve depressed immune cell function from somecritically ill patients and attenuate the production ofpro<strong>in</strong>flammatory mediators <strong>in</strong> others.The am<strong>in</strong>o acids glutam<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e may be usefuldietary additives for patients at risk of or with establishedARDS. 37 Enterocytes metabolise glutam<strong>in</strong>e <strong>in</strong> a manner thatenhances <strong>in</strong>test<strong>in</strong>al mucosal <strong>in</strong>tegrity and reduces translocationof bacteria and tox<strong>in</strong>s <strong>in</strong>to the portal circulation that mayfuel a systemic <strong>in</strong>flammatory response. Glutam<strong>in</strong>e andarg<strong>in</strong><strong>in</strong>e also augment lymphocyte function, and arg<strong>in</strong><strong>in</strong>eimproves monocyte function <strong>in</strong> critically <strong>in</strong>jured patients.L-arg<strong>in</strong><strong>in</strong>e supplementation may also <strong>in</strong>crease NO production,alter vascular tone, and augment free radical mediatedantibacterial defences. Similarly, omega-3 fatty acids such aseicosapentaenoic acid and the unsaturated oil gammal<strong>in</strong>olenicacid reduce pro<strong>in</strong>flammatory cytok<strong>in</strong>e and eicosanoidproduction. 38 Less biologically active eicosanoids such asprostagland<strong>in</strong> PGE 1, thromboxane TXA 3, and leukotriene LTB 5are produced from these unsaturated fats by cyclo-oxygenase(COX) and 5-lipoxygenase dur<strong>in</strong>g <strong>in</strong>flammation. Animalexperiments suggest that polyunsaturated fatty acids canreduce pulmonary vascular resistance, lung neutrophil <strong>in</strong>filtration,and microvascular permeability, thereby improv<strong>in</strong>ggas exchange.In patients with ARDS, enteral immunonutrition supplementedwith antioxidants for at least 4 days was associatedwith reduced pulmonary neutrophil recruitment, improvedoxygenation, a shortened duration of mechanical ventilation,and reduced morbidity <strong>in</strong> terms of new organ failure. 39 However,there was no difference <strong>in</strong> mortality between the controland treatment groups. A meta-analysis of 12 randomised controlledtrials compar<strong>in</strong>g critically ill medical, surgical, andtrauma patients given standard enteral nutrition with patientsreceiv<strong>in</strong>g immunonutrition suggested reduced rates of <strong>in</strong>fection<strong>in</strong>clud<strong>in</strong>g nosocomial pneumonia, but aga<strong>in</strong> no effect onmortality. 40With some reservations, the authors concludedthat their data suggested real benefits of immunonutrition <strong>in</strong>surgical and trauma patients, but that a large double bl<strong>in</strong>d,multicentre, randomised controlled trial was still required.PROSTAGLANDIN E 1Intravenous PGE 1causes both pulmonary and systemicvasodilation and, <strong>in</strong> some critically ill patients, <strong>in</strong>creasescardiac output and oxygen delivery. 41 Although the effect onthe pulmonary circulation is usually small, vasodilation ismore marked under hypoxic conditions, and the nebuliseddrug improves ventilation-perfusion match<strong>in</strong>g. 21 PGE 1also<strong>in</strong>hibits platelet aggregation and neutrophil adhesion. The42<strong>in</strong>itial trial of PGE 1showed improved survival <strong>in</strong> traumapatients with respiratory failure. However, this benefit couldnot be reproduced <strong>in</strong> a subsequent multicentre trial 43<strong>in</strong>patients suffer<strong>in</strong>g from lung <strong>in</strong>jury precipitated by surgery,trauma, or sepsis. The dose of PGE 1was limited by side effects,particularly systemic hypotension. More recent trials haveused liposome technology to <strong>in</strong>crease drug delivery whilemitigat<strong>in</strong>g side effects. 44 45 The use of a liposome itself is associatedwith immune modulat<strong>in</strong>g effects <strong>in</strong>clud<strong>in</strong>g downregulationof neutrophil adhesion molecules. A comb<strong>in</strong>ed PGE 1-liposome preparation <strong>in</strong> a rodent model of ALI reducedpulmonary neutrophil <strong>in</strong>filtration and capillary leak. 46However,although the phase II and III trials of liposomal PGE 1showed that patients with ARDS receiv<strong>in</strong>g the drug had morerapid improvements <strong>in</strong> the PaO 2/FiO 2ratio, neither a survivalbenefit nor a reduced requirement for ventilatory support was44 45found <strong>in</strong> the treatment group. Retrospective subgroupanalysis suggested that high dose therapy might reduce thetime to extubation.THROMBOXANE SYNTHASE AND 5-LIPOXYGENASEINHIBITORSThromboxane and leukotrienes are <strong>in</strong> part responsible for thepulmonary hypertension and hypoxaemia of ARDS. Pulmonaryvascular smooth muscle cells, endothelial cells, platelets,and neutrophils all release TXA 2on stimulation. TXA 2can <strong>in</strong>itiatemicrovascular thromboses consist<strong>in</strong>g of neutrophil andplatelet aggregates that are responsible for perfusion abnormalitiesand recurrent ischaemia-reperfusion <strong>in</strong>jury to thelung. The vasoconstrictive effect of TXA 2similarly contributesto impaired gas exchange. 47 In animal models of lung <strong>in</strong>jurythromboxane synthase <strong>in</strong>hibition reduced pulmonary oedemaformation and <strong>in</strong>hibited microembolism, but pulmonaryhypertension was only partially relieved. 48 Similarly, improvedoxygenation and reduced pulmonary hypertension have beenfound <strong>in</strong> small trials of a thromboxane receptor antagonist <strong>in</strong>patients with ARDS.Leukotrienes (LT) are derived from arachidonic acid by5-lipoxygenase. LTB 4is a potent neutrophil chemok<strong>in</strong>e whileLTC 4and LTD 4cause pulmonary vasoconstriction, capillaryleak, and pulmonary oedema. The role of leukotrienes <strong>in</strong>ARDS has been less well researched but bronchoalveolarlavage fluid from patients with ARDS conta<strong>in</strong>s <strong>in</strong>creased concentrationsof LTB 4,LTC 4and LTD 4, which may be markers fordevelop<strong>in</strong>g ARDS. 49Ketoconazole is an imidazole antifungalagent that <strong>in</strong>hibits thromboxane synthase and 5-lipoxygenasewithout <strong>in</strong>hibit<strong>in</strong>g COX. Ketoconazole may therefore have adual anti-<strong>in</strong>flammatory action <strong>in</strong> ARDS by <strong>in</strong>hibit<strong>in</strong>g <strong>in</strong>flammatoryeicosanoid synthesis and direct<strong>in</strong>g COX productsdown other less <strong>in</strong>flammatory metabolic paths such as thosesynthesis<strong>in</strong>g prostacycl<strong>in</strong> or PGE 2. 50Four trials have usedenteral ketoconazole <strong>in</strong> patients at risk of or with ARDS. The<strong>in</strong>cidence of acute respiratory failure was reduced <strong>in</strong> high risksurgical patients and other critically ill patients. 51–53 However,an ARDS Network trial (http://hedwig.mgh.harvard.edu/ardsnet) <strong>in</strong> patients with established ARDS of medical andsurgical aetiology found no differences <strong>in</strong> <strong>in</strong>-hospital mortality,ventilator free days at day 28, organ failure-free days, ormarkers of gas exchange between patients given ketoconazoleor placebo. 54 This trial achieved plasma levels of ketoconazolehigher than targeted previously but could not demonstrate areduction <strong>in</strong> thromboxane production <strong>in</strong> vivo. The effect ofdecreas<strong>in</strong>g thromboxane synthesis <strong>in</strong> ARDS is therefore stillunknown. It is still possible that ketoconazole <strong>in</strong> surgical andtrauma patients at risk of or with <strong>in</strong>cipient pulmonary <strong>in</strong>jurymay be beneficial.


Non-ventilatory strategies <strong>in</strong> ARDS 71ANTIOXIDANTSThe damage caused by ROS to matrix and cellular prote<strong>in</strong>s,lipids and nucleic acids and ROS mediated signall<strong>in</strong>g contributeto the pathogenesis of ARDS. 55The thiol groups ofglutathione concentrated <strong>in</strong> the lower respiratory tractnormally provide physiological antioxidant protection. However,the concentration and activity of glutathione <strong>in</strong> bronchoalveolarlavage fluid of patients with ARDS is reduced. 56 Intravenousadm<strong>in</strong>istration does not reliably raise glutathionelevels, but glutathione synthesis is stimulated byN-acetylcyste<strong>in</strong>e (NAC) and procyste<strong>in</strong>e (L-2-oxothiazolid<strong>in</strong>e-4-carboxylate). Adm<strong>in</strong>istration of these precursors <strong>in</strong>creasesplasma, erythrocyte, neutrophil, and BAL fluid concentrationsof glutathione <strong>in</strong> patients with ARDS, although a completeeffect may require 10 days of treatment. 57 Early results of NACtherapy were promis<strong>in</strong>g, but several trials have found no difference<strong>in</strong> mortality, length of ventilatory support, orimprovement <strong>in</strong> oxygenation <strong>in</strong> patients with establishedARDS, 58–61and a large, as yet unpublished, phase III trial ofprocyste<strong>in</strong>e was stopped early because of concerns about mortality<strong>in</strong> the treatment arm of the study. Currently, there islittle evidence that <strong>in</strong>travenous NAC or procyste<strong>in</strong>e are ofbenefit to patients with ARDS.Dietary antioxidants such as ascorbic acid, tocopherol, andflavonoids have ROS scaveng<strong>in</strong>g ability and the capacity toreduce oxidised antioxidants as well as b<strong>in</strong>d<strong>in</strong>g ROSproduc<strong>in</strong>g catalysts such as free iron. Although reports of asignificant action of these supplements on pulmonary <strong>in</strong>flammationare lack<strong>in</strong>g, they are already components of some“immunonutrition” preparations. 39PHOSPHATIDIC ACID INHIBITIONPhosphatidic acids are liberated <strong>in</strong> response to <strong>in</strong>flammatorystimuli by lysophosphatidic acyl transferase and, like arachidonicacid, are a source of <strong>in</strong>flammatory mediators. Pentoxifyll<strong>in</strong>eand its more potent metabolite lisofyll<strong>in</strong>e are lysophosphatidicacyl transferase <strong>in</strong>hibitors. These compounds reduceserum free fatty acids <strong>in</strong> humans and lower cytok<strong>in</strong>e production,neutrophil activation, pulmonary neutrophil sequestration,and attenuate lung <strong>in</strong>jury <strong>in</strong> animal models. However, astudy of lisofyll<strong>in</strong>e <strong>in</strong> 235 patients with ALI and ARDS wasstopped at the first <strong>in</strong>terim analysis because the pre-specifiedlevel of improvement for the treatment arm of the trial wasnot achieved. 62PHOSPHOLIPASE INHIBITIONThe enzymes of the secretory phospholipase A 2group arecapable of releas<strong>in</strong>g biologically active polyunsaturated lipids,<strong>in</strong>clud<strong>in</strong>g arachidonic acid, from cell membranes and circulat<strong>in</strong>gplasma lipoprote<strong>in</strong> complexes. The liberated free fattyacids and their metabolites, <strong>in</strong>clud<strong>in</strong>g prostagland<strong>in</strong>s andother eicosanoids, are pro<strong>in</strong>flammatory and may contribute tothe pathophysiology of the sepsis syndrome and multipleorgan failure. 63Furthermore, secretory phospholipases havethe capacity to damage type II alveolar cells directly anddegrade surfactant. 64 65 Raised secretory phospholipase A 2activity is found <strong>in</strong> bronchoalveolar lavage fluid from patientswith ARDS and correlates with the severity of lung <strong>in</strong>jury asmeasured us<strong>in</strong>g the Murray lung <strong>in</strong>jury score. 66 Serum secretoryphospholipase A 2concentrations are elevated <strong>in</strong> patientswith sepsis and correlate with the development of ARDS <strong>in</strong>67 68patients with septic shock and trauma.The positive effects of secretory phospholipase A 2<strong>in</strong>hibition<strong>in</strong> animal models of sepsis and lung <strong>in</strong>jury led to prelim<strong>in</strong>arytrials of two concentrations of a selective <strong>in</strong>hibitor of group IIasecretory phospholipase A 2, namely LY315920Na/S-5920,<strong>in</strong>fused for 7 days <strong>in</strong> patients present<strong>in</strong>g with<strong>in</strong> 36 hours withsevere sepsis. 69In the phase II study the treatment groupshowed a non-significant reduction <strong>in</strong> the development ofARDS and time spent on the ventilator. The pr<strong>in</strong>cipalmortality benefit occurred <strong>in</strong> patients who received the drugwith<strong>in</strong> 18 hours of their first organ failure. Prospective trialsare required to confirm that early adm<strong>in</strong>istration ofLY315920Na/S-5920 protects lung function and improvesmortality <strong>in</strong> patients with severe sepsis.CONCLUSIONMost pharmacological strategies used <strong>in</strong> ARDS have targetedthe <strong>in</strong>flammatory response. Many agents not mentionedhere—cytok<strong>in</strong>e <strong>in</strong>hibitors, anti-<strong>in</strong>flammatory cytok<strong>in</strong>es, antiproteases,and anti-endotox<strong>in</strong> agents—alone and <strong>in</strong> comb<strong>in</strong>ationare <strong>in</strong> the early phases of drug development. Between 18%and 41% of patients with sepsis will develop ARDS and postmortem exam<strong>in</strong>ation reveals evidence of <strong>in</strong>fection, particularlypneumonia, <strong>in</strong> almost all patients with ARDS. 70Thisoverlap between sepsis and ARDS means that improvements<strong>in</strong> the treatment of sepsis may <strong>in</strong>fluence the <strong>in</strong>cidence andoutcome of ARDS. Large cl<strong>in</strong>ical trials of anti-<strong>in</strong>flammatoryagents for sepsis such as ibuprofen, 71 anti-endotox<strong>in</strong>, 72 73 andanti-tumour necrosis factor alpha antibodies 74have had nohuman impact on the mortality associated with ARDS so far.At the time of writ<strong>in</strong>g recomb<strong>in</strong>ant activated prote<strong>in</strong> C hasbeen shown to decrease the mortality of patients with sepsisand may improve the outlook of patients with sepsis <strong>in</strong>ducedARDS. 75If the future of ARDS treatment lies <strong>in</strong> improvements <strong>in</strong> themanagement of multiorgan failure, then the pharmacologicalapproach to treat<strong>in</strong>g lung <strong>in</strong>jury may change. Previousattempts at simply dampen<strong>in</strong>g or halt<strong>in</strong>g the whole acute<strong>in</strong>flammatory process may be replaced by targeted therapiesdirected at elements of the pathological process that producespecific cl<strong>in</strong>ical problems. For example, reduc<strong>in</strong>g pathologicalfibrosis may be possible when more is understood about theregulation of collagen turnover <strong>in</strong> the normal and <strong>in</strong>juredlung. Similarly, the protection, stimulation or suppression ofalveolar cell division, migration, and secretion may bepossible. This may enhance the repair of the alveolar epithelialcell barrier, the clearance of <strong>in</strong>tra-alveolar exudate, and thenormal turnover and function of surfactant.REFERENCES1 Bernard GR, Artigas A, Brigham KL, et al. The American-EuropeanConsensus Conference on ARDS. Def<strong>in</strong>itions, mechanisms, relevantoutcomes, and cl<strong>in</strong>ical trial coord<strong>in</strong>ation. Am J Respir Crit <strong>Care</strong> Med1994;149(3 Pt 1):818–24.2 Abel SJ, F<strong>in</strong>ney SJ, Brett SJ, et al. Reduced mortality <strong>in</strong> association withthe acute respiratory distress syndrome (ARDS). Thorax 1998;53:292–4.3 Davey-Qu<strong>in</strong>n A, Gedney J, Whiteley S, et al. Extravascular lung waterand acute respiratory distress syndrome: oxygenation and outcome.Anaesth Intensive <strong>Care</strong> 1999;27:357–62.4 Humphrey H, Hall J, Sznajder I, et al. Improved survival <strong>in</strong> ARDSpatients associated with a reduction <strong>in</strong> pulmonary capillary wedgepressure. Chest 1990;97:1176–80.5 Mitchell JP, Schuller D, Calandr<strong>in</strong>o FS, et al. Improved outcome basedon fluid management <strong>in</strong> critically ill patients requir<strong>in</strong>g pulmonary arterycatheterization. Am Rev Respir Dis 1992;145:990–8.6 Schuller D, Mitchell JP, Calandr<strong>in</strong>o FS, et al. Fluid balance dur<strong>in</strong>gpulmonary edema. Is fluid ga<strong>in</strong> a marker or a cause of poor outcome?Chest 1991;100:1068–75.7 Payen D. Inhaled nitric oxide and acute lung <strong>in</strong>jury. Cl<strong>in</strong> Chest Med2000;21:519–29.8 Rossa<strong>in</strong>t R, Slama K, Steudel W, et al. Effects of <strong>in</strong>haled nitric oxide onright ventricular function <strong>in</strong> severe acute respiratory distress syndrome.Intensive <strong>Care</strong> Med 1995;21:197–203.9 Murakami S, Bacha EA, Mazmanian GM, et al. Effects of varioustim<strong>in</strong>gs and concentrations of <strong>in</strong>haled nitric oxide <strong>in</strong> lungischemia-reperfusion. The Paris-Sud University Lung TransplantationGroup. Am J Respir Crit <strong>Care</strong> Med 1997;156(2 Pt 1):454–8.10 Chabot F, Mitchell J, Gutteridge J, et al. Reactive oxygen species <strong>in</strong>acute lung <strong>in</strong>jury. Eur Respir J 1998;11:745–57.11 Dell<strong>in</strong>ger RP, Zimmerman JL, Taylor RW, et al. Effects of <strong>in</strong>haled nitricoxide <strong>in</strong> patients with acute respiratory distress syndrome: results of arandomized phase II trial. Inhaled Nitric Oxide <strong>in</strong> ARDS Study Group.Crit <strong>Care</strong> Med 1998;26:15–23.


72 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>12 Gerlach H, Rossa<strong>in</strong>t R, Pappert D, et al. Time-course and dose-responseof nitric oxide <strong>in</strong>halation for systemic oxygenation and pulmonaryhypertension <strong>in</strong> patients with adult respiratory distress syndrome. Eur JCl<strong>in</strong> Invest 1993;23:499–502.13 Michael JR, Barton RG, Saffle JR, et al. Inhaled nitric oxide versusconventional therapy: effect on oxygenation <strong>in</strong> ARDS. Am J Respir Crit<strong>Care</strong> Med 1998;157(5 Pt 1):1372–80.14 Troncy E, Collet JP, Shapiro S, et al. Inhaled nitric oxide <strong>in</strong> acuterespiratory distress syndrome: a pilot randomized controlled study. Am JRespir Crit <strong>Care</strong> Med 1998;157(5 Pt 1):1483–8.15 Rossa<strong>in</strong>t R, Falke KJ, Lopez F, et al. Inhaled nitric oxide for the adultrespiratory distress syndrome. N Engl J Med 1993;328:399–405.16 Cuthbertson BH, Dell<strong>in</strong>ger P, Dyar et al. UK guidel<strong>in</strong>es for the use of<strong>in</strong>haled nitric oxide therapy <strong>in</strong> adult ICUs. American-European ConsensusConference on ALI/ARDS. Intensive <strong>Care</strong> Med 1997;23:1212–8.17 Reyes A, Roca J, Rodriguez-Rois<strong>in</strong> R, et al. Effect of almitr<strong>in</strong>e onventilation-perfusion distribution <strong>in</strong> adult respiratory distress syndrome.Am Rev Respir Dis 1988;137:1062–7.18 Gallart L, Lu Q, Puybasset L, et al. Intravenous almitr<strong>in</strong>e comb<strong>in</strong>ed with<strong>in</strong>haled nitric oxide for acute respiratory distress syndrome. The NOAlmitr<strong>in</strong>e Study Group. Am J Respir Crit <strong>Care</strong> Med 1998;158:1770–7.19 Walmrath D, Schneider T, Schermuly R, et al. Direct comparison of<strong>in</strong>haled nitric oxide and aerosolized prostacycl<strong>in</strong> <strong>in</strong> acute respiratorydistress syndrome. Am J Respir Crit <strong>Care</strong> Med 1996;153:991–6.20 Zwissler B, Kemm<strong>in</strong>g G, Habler O, et al. Inhaled prostacycl<strong>in</strong> (PGI2)versus <strong>in</strong>haled nitric oxide <strong>in</strong> adult respiratory distress syndrome. Am JRespir Crit <strong>Care</strong> Med 1996;154(6 Pt 1):1671–7.21 Meyer J, Theilmeier G, Van Aken H, et al. Inhaled prostagland<strong>in</strong> E1 fortreatment of acute lung <strong>in</strong>jury <strong>in</strong> severe multiple organ failure. AnesthAnalg 1998;86:753–8.22 Sladen A. Methylprednisolone. Pharmacologic doses <strong>in</strong> shock lungsyndrome. J Thorac Cardiovasc Surg 1976;71:800–6.23 Lucas CE, Ledgerwood AM. Pulmonary response of massive steroids <strong>in</strong>seriously <strong>in</strong>jured patients. Ann Surg 1981;194:256–61.24 Weigelt JA, Norcross JF, Borman KR, et al. Early steroid therapy forrespiratory failure. Arch Surg 1985;120:536–40.25 Bernard GR, Luce JM, Sprung CL, et al. High-dose corticosteroids <strong>in</strong>patients with the adult respiratory distress syndrome. N Engl J Med1987;317:1565–70.26 Bone RC, Fisher CJ Jr, Clemmer TP, et al. A controlled cl<strong>in</strong>ical trial ofhigh-dose methylprednisolone <strong>in</strong> the treatment of severe sepsis and septicshock. N Engl J Med 1987;317:653–8.27 Kam<strong>in</strong>ski N, Allard JD, Pittet JF, et al. Global analysis of geneexpression <strong>in</strong> pulmonary fibrosis reveals dist<strong>in</strong>ct programs regulat<strong>in</strong>g lung<strong>in</strong>flammation and fibrosis. Proc Natl Acad Sci USA 2000;97:1778–83.28 Meduri GU, Headley AS, Golden E, et al. Effect of prolongedmethylprednisolone therapy <strong>in</strong> unresolv<strong>in</strong>g acute respiratory distresssyndrome: a randomized controlled trial. JAMA 1998;280:159–65.29 Brun-Buisson C, Brochard L. Corticosteroid therapy <strong>in</strong> acute respiratorydistress syndrome: better late than never? JAMA 1998;280:182–3.30 Manalo E, Merritt TA, Kheiter A, et al. Comparative effects of someserum components and proteolytic products of fibr<strong>in</strong>ogen on surfacetension-lower<strong>in</strong>g abilities of beractant and a synthetic peptide conta<strong>in</strong><strong>in</strong>gsurfactant KL4. Pediatr Res 1996;39:947–52.31 Amirkhanian JD, Merritt TA. Inhibitory effects of oxyradicals onsurfactant function: utiliz<strong>in</strong>g <strong>in</strong> vitro Fenton reaction. Lung1998;176:63–72.32 Anzueto A, Baughman RP, Guntupalli KK, et al. Aerosolized surfactant<strong>in</strong> adults with sepsis-<strong>in</strong>duced acute respiratory distress syndrome. ExosurfAcute <strong>Respiratory</strong> Distress Syndrome Sepsis Study Group. N Engl J Med1996;334:1417–21.33 Gregory TJ, Ste<strong>in</strong>berg KP, Spragg R, et al. Bov<strong>in</strong>e surfactant therapy forpatients with acute respiratory distress syndrome. Am J Respir Crit <strong>Care</strong>Med 1997;155:1309–15.34 Wiswell TE, Smith RM, Katz LB, et al. Bronchopulmonary segmentallavage with Surfax<strong>in</strong> (KL(4)-surfactant) for acute respiratory distresssyndrome. Am J Respir Crit <strong>Care</strong> Med 1999;160:1188–95.35 Al-Saady NM, Blackmore CM, Bennett ED. High fat, low carbohydrate,enteral feed<strong>in</strong>g lowers PaCO 2 and reduces the period of ventilation <strong>in</strong>artificially ventilated patients. Intensive <strong>Care</strong> Med 1989;15:290–5.36 K<strong>in</strong>g BK, Kudsk KA, Li J, et al. Route and type of nutrition <strong>in</strong>fluencemucosal immunity to bacterial pneumonia. Ann Surg 1999;229:272–8.37 Chang DW, DeSanti L, Deml<strong>in</strong>g RH. Anticatabolic and anabolicstrategies <strong>in</strong> critical illness: a review of current treatment modalities.Shock 1998;10:155–60.38 Zaloga GP. Dietary lipids: ancestral ligands and regulators of cellsignal<strong>in</strong>g pathways. Crit <strong>Care</strong> Med 1999;27:1646–8.39 Gadek JE, DeMichele SJ, Karlstad MD, et al. Effect of enteral feed<strong>in</strong>gwith eicosapentaenoic acid, gamma-l<strong>in</strong>olenic acid, and antioxidants <strong>in</strong>patients with acute respiratory distress syndrome. Enteral Nutrition <strong>in</strong>ARDS Study Group. Crit <strong>Care</strong> Med 1999;27:1409–20.40 Beale RJ, Bryg DJ, Bihari DJ. Immunonutrition <strong>in</strong> the critically ill: asystematic review of cl<strong>in</strong>ical outcome. Crit <strong>Care</strong> Med 1999;27:2799–805.41 Silverman HJ, Slotman G, Bone RC, et al. Effects of prostagland<strong>in</strong> E1 onoxygen delivery and consumption <strong>in</strong> patients with the adult respiratorydistress syndrome. Results from the prostagland<strong>in</strong> E1 multicenter trial. TheProstagland<strong>in</strong> E1 Study Group. Chest 1990;98:405–10.42 Holcroft JW, Vassar MJ, Weber CJ. Prostagland<strong>in</strong> E1 and survival <strong>in</strong>patients with the adult respiratory distress syndrome. A prospective trial.Ann Surg 1986;203:371–8.43 Bone RC, Slotman G, Maunder R, et al. Randomized double-bl<strong>in</strong>d,multicenter study of prostagland<strong>in</strong> E1 <strong>in</strong> patients with the adult respiratorydistress syndrome. Prostagland<strong>in</strong> E1 Study Group. Chest1989;96:114–9.44 Abraham E, Park YC, Cov<strong>in</strong>gton P, et al. Liposomal prostagland<strong>in</strong> E1 <strong>in</strong>acute respiratory distress syndrome: a placebo-controlled, randomized,double-bl<strong>in</strong>d, multicenter cl<strong>in</strong>ical trial. Crit <strong>Care</strong> Med 1996;24:10–5.45 Abraham E, Baughman R, Fletcher E, et al. Liposomal prostagland<strong>in</strong> E1(TLC C-53) <strong>in</strong> acute respiratory distress syndrome: a controlled,randomized, double-bl<strong>in</strong>d, multicenter cl<strong>in</strong>ical trial. TLC C-53 ARDS StudyGroup. Crit <strong>Care</strong> Med 1999;27:1478–85.46 Leff JA, Baer JW, Kirkman JM, et al. Liposome-entrapped PGE1posttreatment decreases IL-1 alpha-<strong>in</strong>duced neutrophil accumulation andlung leak <strong>in</strong> rats. J Appl Physiol 1994;76:151–7.47 Petrak RA, Balk RA, Bone RC. Prostagland<strong>in</strong>s, cyclo-oxygenase<strong>in</strong>hibitors, and thromboxane synthetase <strong>in</strong>hibitors <strong>in</strong> the pathogenesis ofmultiple systems organ failure. Crit <strong>Care</strong> Cl<strong>in</strong> 1989;5:303–14.48 Hechtman HB, Valeri CR, Shepro D. Role of humoral mediators <strong>in</strong> adultrespiratory distress syndrome. Chest 1984;86:623–7.49 Pittet JF, Mackersie RC, Mart<strong>in</strong> TR, et al. Biological markers of acutelung <strong>in</strong>jury: prognostic and pathogenetic significance. Am J Respir Crit<strong>Care</strong> Med 1997;155:1187–205.50 Beetens JR, Loots W, Somers Y, et al. Ketoconazole <strong>in</strong>hibits thebiosynthesis of leukotrienes <strong>in</strong> vitro and <strong>in</strong> vivo. Biochem Pharmacol1986;35:883–91.51 Slotman GJ, Burchard KW, D’Arezzo A, et al. Ketoconazole preventsacute respiratory failure <strong>in</strong> critically ill surgical patients. J Trauma1988;28:648–54.52 Yu M, Tomasa G. A double-bl<strong>in</strong>d, prospective, randomized trial ofketoconazole, a thromboxane synthetase <strong>in</strong>hibitor, <strong>in</strong> the prophylaxis ofthe adult respiratory distress syndrome. Crit <strong>Care</strong> Med1993;21:1635–42.53 S<strong>in</strong>uff T, Cook DJ, Peterson JC, et al. Development, implementation, andevaluation of a ketoconazole practice guidel<strong>in</strong>e for ARDS prophylaxis. JCrit <strong>Care</strong> 1999;14:1–6.54 The ARDS Cl<strong>in</strong>ical Trials Network. Ketoconazole for early treatment ofacute lung <strong>in</strong>jury and acute respiratory distress syndrome: a randomizedcontrolled trial. The ARDS Network. JAMA 2000;283:1995–2002.55 Qu<strong>in</strong>lan G, Upton R. Oxidant-antioxidant balance <strong>in</strong> acute respiratorydistress syndrome. In: Evans T, Griffiths M, Keogh B, eds. European<strong>Respiratory</strong> Monograph: ARDS. Sheffield: European <strong>Respiratory</strong> JournalsLtd, 2002.56 Pacht ER, Timerman AP, Lykens MG, et al. Deficiency of alveolar fluidglutathione <strong>in</strong> patients with sepsis and the adult respiratory distresssyndrome. Chest 1991;100:1397–403.57 Bernard GR. N-acetylcyste<strong>in</strong>e <strong>in</strong> experimental and cl<strong>in</strong>ical acute lung<strong>in</strong>jury. Am J Med 1991;91:54–9S.58 Ortolani O, Conti A, De Gaudio AR, et al. Protective effects ofN-acetylcyste<strong>in</strong>e and rut<strong>in</strong> on the lipid peroxidation of the lung epitheliumdur<strong>in</strong>g the adult respiratory distress syndrome. Shock 2000;13:14–8.59 Jepsen S, Herlevsen P, Knudsen P, et al. Antioxidant treatment withN-acetylcyste<strong>in</strong>e dur<strong>in</strong>g adult respiratory distress syndrome: aprospective, randomized, placebo-controlled study. Crit <strong>Care</strong> Med1992;20:918–23.60 Domenighetti G, Suter PM, Schaller MD, et al. Treatment withN-acetylcyste<strong>in</strong>e dur<strong>in</strong>g acute respiratory distress syndrome: arandomized, double-bl<strong>in</strong>d, placebo-controlled cl<strong>in</strong>ical study. J Crit <strong>Care</strong>1997;12:177–82.61 Bernard GR, Wheeler AP, Arons MM, et al. A trial of antioxidantsN-acetylcyste<strong>in</strong>e and procyste<strong>in</strong>e <strong>in</strong> ARDS. The Antioxidant <strong>in</strong> ARDSStudy Group. Chest 1997;112:164–72.62 The ARDS Cl<strong>in</strong>ical Trials Network. Randomized, placebo-controlledtrial of lisofyll<strong>in</strong>e for early treatment of acute lung <strong>in</strong>jury and acuterespiratory distress syndrome. Crit <strong>Care</strong> Med 2002;30:1–6.63 Anderson BO, Moore EE, Banerjee A. Phospholipase A 2 regulatescritical <strong>in</strong>flammatory mediators of multiple organ failure. J Surg Res1994;56:199–205.64 Durham SK, Selig WM. Phospholipase A 2 -<strong>in</strong>duced pathophysiologicchanges <strong>in</strong> the gu<strong>in</strong>ea pig lung. Am J Pathol 1990;136:1283–91.65 Kuwabara K, Furue S, Tomita Y, et al. Effect of methylprednisolone onphospholipase A 2 activity and lung surfactant degradation <strong>in</strong> acute lung<strong>in</strong>jury <strong>in</strong> rabbits. Eur J Pharmacol 2001;433:209–16.66 Kim DK, Fukuda T, Thompson B, et al. Bronchoalveolar lavage fluidphospholipase A 2 activities are <strong>in</strong>creased <strong>in</strong> human adult respiratorydistress syndrome. Am J Physiol 1995;269:109–18.67 Endo S, Inada K, Nakae H, et al. Plasma levels of type II phospholipaseA 2 and cytok<strong>in</strong>es <strong>in</strong> patients with sepsis. Res Commun Mol PatholPharmacol 1995;90:413–21.68 Buchler M, Deller A, Malferthe<strong>in</strong>er P, et al. Serum phospholipase A 2 <strong>in</strong><strong>in</strong>tensive care patients with peritonitis, multiple <strong>in</strong>jury, and necrotiz<strong>in</strong>gpancreatitis. Kl<strong>in</strong> Wochenschr 1989;67:217–21.69 Abraham E, Naum C, Bandi V, et al. Efficacy and safety ofLY315920Na/S-5920, a selective <strong>in</strong>hibitor of 14-kDa group IIAsecretory phospholipase A 2 , <strong>in</strong> patients with suspected sepsis and organfailure. Crit <strong>Care</strong> Med 2003;31:718–28.70 Fe<strong>in</strong> AM, Calalang-Colucci MG. Acute lung <strong>in</strong>jury and acute respiratorydistress syndrome <strong>in</strong> sepsis and septic shock. Crit <strong>Care</strong> Cl<strong>in</strong>2000;16:289–317.71 Bernard GR, Wheeler AP, Russell JA, et al. The effects of ibuprofen onthe physiology and survival of patients with sepsis. The Ibuprofen <strong>in</strong>Sepsis Study Group. N Engl J Med 1997;336:912–8.72 McCloskey RV, Straube RC, Sanders C, et al. Treatment of septic shockwith human monoclonal antibody HA-1A. A randomized, double-bl<strong>in</strong>d,placebo-controlled trial. CHESS Trial Study Group. Ann Intern Med1994;121:1–5.


Non-ventilatory strategies <strong>in</strong> ARDS 7373 Ziegler EJ, Fisher CJ, Jr., Sprung CL, et al. Treatment of gram-negativebacteremia and septic shock with HA-1A human monoclonal antibodyaga<strong>in</strong>st endotox<strong>in</strong>. A randomized, double-bl<strong>in</strong>d, placebo-controlled trial.The HA-1A Sepsis Study Group. N Engl J Med 1991;324:429–36.74 Cohen J, Carlet J. INTERSEPT: an <strong>in</strong>ternational, multicenter,placebo-controlled trial of monoclonal antibody to human tumor necrosisfactor-alpha <strong>in</strong> patients with sepsis. International Sepsis Trial StudyGroup. Crit <strong>Care</strong> Med 1996;24:1431–40.75 Bernard GR, V<strong>in</strong>cent JL, Laterre PF, et al. Efficacy and safety ofrecomb<strong>in</strong>ant human activated prote<strong>in</strong> C for severe sepsis. N Engl J Med2001;344:699–709.76 Lund<strong>in</strong> S, Mang H, Smithies M, et al. Inhalation of nitric oxide <strong>in</strong> acutelung <strong>in</strong>jury: results of a European multicentre study. The European StudyGroup of Inhaled Nitric Oxide. Intensive <strong>Care</strong> Med 1999;25:911–9.77 Hooper RG, Kearl RA. Established ARDS treated with a susta<strong>in</strong>ed courseof adrenocortical steroids. Chest 1990;97:138–43.78 Meduri GU, Headley S, Tolley E, et al. Plasma and BAL cytok<strong>in</strong>eresponse to corticosteroid rescue treatment <strong>in</strong> late ARDS. Chest1995;108:1315–25.79 Biffl WL, Moore FA, Moore EE, et al. Are corticosteroids salvage therapyfor refractory acute respiratory distress syndrome? Am J Surg1995;170:591–5; discussion 595–6.80 Meduri GU, Ch<strong>in</strong>n AJ, Leeper KV, et al. Corticosteroid rescue treatmentof progressive fibroproliferation <strong>in</strong> late ARDS. Patterns of response andpredictors of outcome. Chest 1994;105:1516–27.81 Meduri GU, Belenchia JM, Estes RJ, et al. Fibroproliferative phase ofARDS. Cl<strong>in</strong>ical f<strong>in</strong>d<strong>in</strong>gs and effects of corticosteroids. Chest1991;100:943–52.82 Luce JM, Montgomery AB, Marks JD, et al. Ineffectiveness of high-dosemethylprednisolone <strong>in</strong> prevent<strong>in</strong>g parenchymal lung <strong>in</strong>jury and improv<strong>in</strong>gmortality <strong>in</strong> patients with septic shock. Am Rev Respir Dis1988;138:62–8.


11 Difficult wean<strong>in</strong>gJ Goldstone.............................................................................................................................Difficulty <strong>in</strong> wean<strong>in</strong>g from mechanical ventilationis associated with <strong>in</strong>tr<strong>in</strong>sic lungdisease and/or a prolonged critical illness.After critical illness the <strong>in</strong>cidence of wean<strong>in</strong>gfailure varies with 20% of all admissions fail<strong>in</strong>g<strong>in</strong>itial wean<strong>in</strong>g. 12 The <strong>in</strong>cidence of wean<strong>in</strong>gfailure <strong>in</strong>creases <strong>in</strong> patients who have been ventilatedfor many weeks but is low (


Difficult wean<strong>in</strong>g 75Table 11.2 The three determ<strong>in</strong>ants of ventilation andcommon pathophysiological conditions associated withfailure to weanCentral drive<strong>Respiratory</strong> muscle strengthLoad applied to the muscles<strong>Respiratory</strong> musclestrength• Sedation, analgesia or anaesthesia• Coma• Raised <strong>in</strong>tracranial pressure• Hypercapnia• Hypophosphataemia• Disuse atrophy• Sepsis• Polyneuropathy/myopathy• Hyper<strong>in</strong>flation• Left ventricular failure• Bronchospasm• Lung fibrosisCNS driveApplied loadFailure to generateforce (fatigue)Figure 11.1 Key components of spontaneous breath<strong>in</strong>g. Drivefrom the central nervous system acts on the peripheral respiratorymuscles. The balance between the components can be disordered,lead<strong>in</strong>g to fatigue of the respiratory muscles, failure to generateforce, and a decrease <strong>in</strong> alveolar ventilation.Yang and Tob<strong>in</strong> 8 devised the CROP <strong>in</strong>dex ((Cdyn × PImax ×[PaO 2 /PAO 2])/rate) which consisted of dynamic compliance(Cdyn), maximum mouth pressure (PImax), oxygenation(PaO 2 /PAO 2), and respiratory rate. This was no better than f/Vtalone when assessed prospectively. Measurements <strong>in</strong>tegrat<strong>in</strong>gventilatory endurance and the efficiency of gas exchange yieldthe most successful results but are complex and difficult touse. 10COMPONENTS OF WEANING FAILUREWean<strong>in</strong>g from mechanical ventilation depends on thestrength of the respiratory muscles, the load applied to themuscles, and the central drive (table 11.2). <strong>Respiratory</strong> failuremay result from disorders <strong>in</strong> one of these three areas—forexample, a myopathy reduc<strong>in</strong>g strength, acute bronchospasmsuddenly <strong>in</strong>creas<strong>in</strong>g load, or opiates act<strong>in</strong>g on the centralnervous system. However, it is also possible that disorders ofstrength and load occur together.The relationship between these three key components ofspontaneous breath<strong>in</strong>g may be visualised as a balance (fig11.1). If the muscles are heavily loaded, spontaneous contractioncannot be ma<strong>in</strong>ta<strong>in</strong>ed and the muscles may fail acutely.Such acute reversible failure of force generation is termedfatigue. This has been shown <strong>in</strong> studies of both electromyography(EMG) 11 12 and changes <strong>in</strong> the relaxation rate of respiratorymuscles dur<strong>in</strong>g wean<strong>in</strong>g. 13 The pathophysiology of wean<strong>in</strong>gfailure has been studied <strong>in</strong> small groups of patients. 13–15 Itseems likely that the dom<strong>in</strong>ant feature is high levels of loadrelative to the strength of the respiratory muscles. As wean<strong>in</strong>gprogresses, load <strong>in</strong>creases compared with those who succeed awean<strong>in</strong>g trial. In most cases the drive to breathe is high. 15<strong>Respiratory</strong> muscle strengthOrig<strong>in</strong>ally the tension of the respiratory muscles was tested <strong>in</strong>normal subjects by tak<strong>in</strong>g maximum pressure measurements16 17at the mouth (PImax), while oesophageal and gastricballoon catheters allow the study of diaphragmatic strength.Contractions of the diaphragm can be obta<strong>in</strong>ed by electric or18 19magnetic stimulation of the phrenic nerves.In the <strong>in</strong>tubated patient maximal pressure generation canbe assessed dur<strong>in</strong>g occluded maximal manoeuvres and thiscan be simply performed as the endotracheal tube is easilyaccessible. PImax was orig<strong>in</strong>ally measured <strong>in</strong> <strong>in</strong>tubatedpatients be<strong>in</strong>g weaned from mechanical ventilation by Sahnand Lakshm<strong>in</strong>arayan. 1 Patients with severe weakness (PImax


76 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>rest between tra<strong>in</strong><strong>in</strong>g exercises. 28 In addition, strengthtra<strong>in</strong><strong>in</strong>g differs conceptually and <strong>in</strong> practice from endurancetra<strong>in</strong><strong>in</strong>g. 29 For the respiratory muscles, tra<strong>in</strong><strong>in</strong>g is ill def<strong>in</strong>edand although it is felt that the respiratory muscles shouldbehave <strong>in</strong> a similar manner to other muscle groups, def<strong>in</strong>itivestudies have yet to show how they may be tra<strong>in</strong>ed. It is likelythat the response to tra<strong>in</strong><strong>in</strong>g will <strong>in</strong> part be genetically determ<strong>in</strong>ed.The general observation that some <strong>in</strong>dividuals areresponsive to and have ability at certa<strong>in</strong> types of exercise hasled to studies show<strong>in</strong>g genetic differences <strong>in</strong> the response totra<strong>in</strong><strong>in</strong>g accord<strong>in</strong>g to genotype. 30 A genetic polymorphism ofthe angiotens<strong>in</strong> convert<strong>in</strong>g enzyme (ACE) gene has beendescribed with a 256 base pair deletion or <strong>in</strong>sertion, termedDD or II. 31 In de-tra<strong>in</strong>ed subjects there is an 11-fold differencebetween homozygous subgroups <strong>in</strong> response to perform<strong>in</strong>g arepetitive biceps exercise. 30 Recently, respiratory musclestrength and endurance was studied <strong>in</strong> de-tra<strong>in</strong>ed subjectswho underwent general non-specific tra<strong>in</strong><strong>in</strong>g. <strong>Respiratory</strong>muscle endurance was <strong>in</strong>creased fivefold <strong>in</strong> the II subgroup. 32AObstruction150AlveolusTrachea_ 16Distalairway00Pleural pressureProximalbranch<strong>in</strong>gairwaysCentral nervous system driveAlthough central respiratory drive is not often measured onthe ICU, it is possible to measure P 0.1, an <strong>in</strong>dex of drive. 33 P 0.1israised when respiratory drive is artificially <strong>in</strong>creased dur<strong>in</strong>g ahypercapnic challenge and is also high <strong>in</strong> patients suffer<strong>in</strong>gventilatory failure. 34 In <strong>in</strong>tubated patients it is often the casethat little or no gas flows <strong>in</strong> the early part of <strong>in</strong>spiration, ifvalves are required to open and the speed of response is slow.In such circumstances, patients may be mak<strong>in</strong>g occlud<strong>in</strong>gbreath<strong>in</strong>g efforts and P 0.1may be measured with<strong>in</strong> the airwayautomatically by the ventilator. 35Can P 0.1be used to assess wean<strong>in</strong>g from mechanical ventilation?It is easy to apply the technique to ventilated patientsand, when respiratory drive is raised, the measured pressureexceeds 5.5 cm H 2O. A raised P 0.1is associated with failure towean. 36 Interest<strong>in</strong>gly, patients who are able to breathe dur<strong>in</strong>gwean<strong>in</strong>g trials not only have a low P 0.1but are also able to<strong>in</strong>crease drive and m<strong>in</strong>ute ventilation dur<strong>in</strong>g a hypercapnicchallenge. 37 Patients who are able to breathe spontaneously doso with a lower central drive and also have some ventilatoryreserve, contrast<strong>in</strong>g with the fixed capacity of patients whofail to wean.<strong>Respiratory</strong> drive has been measured <strong>in</strong> patients receiv<strong>in</strong>gpressure support ventilation where the level of pressuresupport was decreased <strong>in</strong> stages. 38 It would follow that, as theamount of support decreases, there will be a moment whendrive to the muscles <strong>in</strong>creases. In patients who were able tobreathe spontaneously, the level of drive rema<strong>in</strong>ed low.Conversely, <strong>in</strong> patients who fail to wean, drive <strong>in</strong>creased, oftenabove the level seen previously. It is possible that the level ofventilatory support could be titrated <strong>in</strong> this manner, keep<strong>in</strong>gthe level of drive with<strong>in</strong> the “normal” range for patients on theICU. A similar approach was used to adjust the level of externalPEEP applied to patients with vary<strong>in</strong>g degrees of <strong>in</strong>tr<strong>in</strong>sicPEEP. 39 As the level of external PEEP <strong>in</strong>creased, the ability ofthe patients to achieve gas flow reduced until the optimumbalance of <strong>in</strong>ternal and external PEEP was reached (fig 11.2).<strong>Respiratory</strong> drive <strong>in</strong>creased if external PEEP achievedhyper<strong>in</strong>flation, enabl<strong>in</strong>g the adjustment of external PEEP tothe correct level without requir<strong>in</strong>g the difficult measurementof <strong>in</strong>ternal PEEP <strong>in</strong> the spontaneously breath<strong>in</strong>g patient.Load applied to the musclesWork is performed when a force moves through a distance andis termed “external” as it may be easily measured. Internalwork is performed when there is no movement, when themuscle contracts and produces tension and heat. To calculateexternal work <strong>in</strong> the respiratory system, the tidal volume mustbe <strong>in</strong>tegrated with respect to the transpleural pressure generateddur<strong>in</strong>g the breath. This requires some measure of pleuralBObstruction15Gas flow15AlveolusTrachea_ 115Distalairway_ 1Pleural pressureProximalbranch<strong>in</strong>gairwaysFigure 11.2 The threshold load effect of <strong>in</strong>tr<strong>in</strong>sic or auto-positiveend expiratory pressure (PEEP). When auto-PEEP is high, no gas flowwill occur at the mouth until the pressure generated with<strong>in</strong> the chestexceeds the level of <strong>in</strong>tr<strong>in</strong>sic PEEP. (A) The pressure with<strong>in</strong> thealveolus cannot fall to zero because of the obstruction to expiratoryflow. The pressure to beg<strong>in</strong> gas flow must be less than the level of<strong>in</strong>tr<strong>in</strong>sic PEEP, <strong>in</strong> this case –16 cm H 2O. (B) External PEEP is applied,balanc<strong>in</strong>g the <strong>in</strong>tr<strong>in</strong>sic PEEP. This has the effect of reduc<strong>in</strong>g thepressure required to beg<strong>in</strong> <strong>in</strong>spiratory flow, <strong>in</strong> this case from –16 to–1 cm H 2O.pressure, usually obta<strong>in</strong>ed from oesophageal balloon catheters,and simultaneous measurement of volume at themouth. Internal work can be imag<strong>in</strong>ed if there is no gas flow,as occurs <strong>in</strong> complete obstruction. In this circumstance,energy is dissipated aga<strong>in</strong>st distortions of the chest wall andno ventilation occurs.Work is often <strong>in</strong>creased <strong>in</strong> wean<strong>in</strong>g failure 40 41 and successfulwean<strong>in</strong>g occurs when work is reduced. It is possible tomonitor work cont<strong>in</strong>uously and, <strong>in</strong> those who fail to wean,pressure generation is significantly higher at the end of thewean<strong>in</strong>g trial, <strong>in</strong>spiration as a fraction of the respiratory cyclelengthens, and patients are tachypnoeic.CLINICAL IMPLICATIONSExhaustive breath<strong>in</strong>g may damage skeletal muscle fibres andcause a reduction <strong>in</strong> the ability to generate pressure. Indeed, <strong>in</strong>healthy volunteers the strength of the diaphragm as judged bymagnetic twitch transdiaphragmatic pressure is substantiallyreduced up to 24 hours after breath<strong>in</strong>g to exhaustion throughan <strong>in</strong>spiratory resistance of 60% of maximal. 4215PEEP valve


Difficult wean<strong>in</strong>g 77Failure to wean from mechanical ventilation does notexclusively affect respiratory muscle performance. The oxygenconsumption of muscles can rise considerably and changes <strong>in</strong>gut mucosal pH <strong>in</strong>dicate that an oxygen debt occurs dur<strong>in</strong>g43 44failed wean<strong>in</strong>g attempts. More organ specific disordersoccur when the level of respiratory work is high. In patients atrisk of coronary artery disease, wean<strong>in</strong>g precipitatesischaemia 45which may not be detected at the bedside,particularly if three lead ECG monitor<strong>in</strong>g is used. Wean<strong>in</strong>g isless likely to succeed if myocardial ischaemia occurs. 46Furthermore, mechanical ventilation supports left ventricularfunction <strong>in</strong> patients with <strong>in</strong>cipient heart failure. 47Hence,<strong>in</strong>vasive haemodynamic measurements and radionuclideimag<strong>in</strong>g <strong>in</strong> patients showed decreased left ventricularperformance and oesophageal pressure, and a 2–3-fold<strong>in</strong>crease <strong>in</strong> pulmonary artery occlusion pressure when wean<strong>in</strong>gfailed. Re-ventilation reversed this effect and subsequenttreatment to support the myocardium led to successful wean<strong>in</strong>g.Heart failure <strong>in</strong> patients who fail to disconnect frommechanical ventilation is an important differential diagnosis.Trigger<strong>in</strong>g mechanical ventilation is an important aspect ofsett<strong>in</strong>g the ventilator when patients are breath<strong>in</strong>g spontaneously.Trigger<strong>in</strong>g from the <strong>in</strong>spiratory flow reduces work<strong>in</strong>volved <strong>in</strong> trigger<strong>in</strong>g compared with pressure trigger<strong>in</strong>g. 48 Ifthe trigger sensitivity is set <strong>in</strong>appropriately, it is difficult tobreathe through the ventilator and, <strong>in</strong> weak patients, it is possiblethat no breath is delivered and de-synchrony occurs.Improvements <strong>in</strong> trigger methodology have decreased <strong>in</strong>spiratorywork, with flow trigger<strong>in</strong>g becom<strong>in</strong>g the standard. Inhealth, ventilators can now be set such that almost no work isperformed to <strong>in</strong>itiate a breath. Mechanical ventilation <strong>in</strong>severe lung disease is a greater challenge, especially <strong>in</strong>obstructive lung diseases where the transmission of the<strong>in</strong>spiratory effort to the upper airway may be delayed. Whenthe time delay is prolonged, the ventilator senses <strong>in</strong>spiration ata po<strong>in</strong>t when the <strong>in</strong>spiratory muscles are contract<strong>in</strong>g. Thus,persistent respiratory muscle contraction leads to occult <strong>in</strong>ternalwork be<strong>in</strong>g performed. Instead of conventional trigger<strong>in</strong>gat the ventilator end of the airway, sens<strong>in</strong>g <strong>in</strong>spiration at thedistal tip of the endotracheal tube would avoid some of thetime delay <strong>in</strong> patients with chronic obstructive pulmonarydisease (COPD). Experimentally, it is possible to compare conventionaltrigger<strong>in</strong>g with trigger<strong>in</strong>g at the ventilator, and substantialreductions <strong>in</strong> the work can be achieved. It is possibleto move the <strong>in</strong>spiratory trigger even closer to the respiratorymuscles, offsett<strong>in</strong>g the delay <strong>in</strong> trigger<strong>in</strong>g if pressure is sensedwith<strong>in</strong> the chest. Oesophageal trigger<strong>in</strong>g has recently beenfound to reduce total <strong>in</strong>spiratory work <strong>in</strong> normal volunteers. 49RECENT ADVANCES IN MECHANICAL VENTILATIONProportional assist ventilation (PAV)A conventional ventilator has one variable that is determ<strong>in</strong>edby the user. For example, <strong>in</strong> the pressure control mode the airwaypressure can be manipulated. Dur<strong>in</strong>g the breath the volumedelivered to the patient depends on the mechanics of thelung and chest wall. With PAV the ventilator measures thecompliance and resistance of the system dur<strong>in</strong>g each breath.The ventilator can then be set to deliver the pressure requiredfor a given tidal volume, or a proportion of it, depend<strong>in</strong>g on thega<strong>in</strong> of the system set by the operator. PAV can unload the respiratorymuscles to a greater extent than other modes of ventilation.It can compensate for dynamic changes <strong>in</strong> resistanceand compliance and allow the patient to vary tidal breath<strong>in</strong>g,ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g the amount of <strong>in</strong>tr<strong>in</strong>sic muscle effort set by the50 51operator. Similar technology can be applied to the workrequired to breathe through an endotracheal tube. By measur<strong>in</strong>gthe resistance and compliance of a standard endotrachealtube, automatic tube compensation (the amount of assistancerelative to the <strong>in</strong>spiratory flow rate) can be provided. 52Hyper<strong>in</strong>flationExpiratory flow limitation causes hyper<strong>in</strong>flation and <strong>in</strong>creasedrest<strong>in</strong>g end expiratory pressure. This is termed auto or<strong>in</strong>tr<strong>in</strong>sic PEEP, 53 and it acts as a load dur<strong>in</strong>g <strong>in</strong>spiration as thepatient must generate a negative pressure equal to the level ofauto-PEEP <strong>in</strong> order to generate gas flow at the mouth thattriggers <strong>in</strong>spiration. Asynchrony with the ventilator may becaused by excessive auto-PEEP and may be resolved bymatch<strong>in</strong>g the external applied PEEP to balance the system. 54In normal subjects, such a load would be easily borne. Forexample, an average level of <strong>in</strong>tr<strong>in</strong>sic PEEP of 11 cm H 2Oisasmall fraction of the total pressure generat<strong>in</strong>g ability.However, many <strong>in</strong>tubated patients generate a maximum of–30 cm H 2O. In this context, overcom<strong>in</strong>g the threshold loadeffect of <strong>in</strong>tr<strong>in</strong>sic PEEP uses 33% of available pressure generationand may contribute to fatigue.Techniques of wean<strong>in</strong>gImportant studies <strong>in</strong> the 1990s established that the mode ofventilation has a major <strong>in</strong>fluence on the success of wean<strong>in</strong>g.Brochard et al 55 compared wean<strong>in</strong>g by pressure support (PS),T-piece trials, and synchronised <strong>in</strong>termittent mandatoryventilation (SIMV) <strong>in</strong> a group of patients who had failed towean and <strong>in</strong> whom it was predicted that wean<strong>in</strong>g would beproblematic. Over a period of 28 days SIMV was clearly<strong>in</strong>ferior to the other techniques, with an advantage <strong>in</strong> favourof the PS group. This study also emphasised the importance ofa wean<strong>in</strong>g protocol. The Spanish Lung Failure CollaborativeGroup reported contrast<strong>in</strong>g f<strong>in</strong>d<strong>in</strong>gs. 56 While SIMV was clearlyless favourable, T-piece wean<strong>in</strong>g was advantageous overall.Although both studies showed that SIMV was disadvantageous,the explanation for the differ<strong>in</strong>g f<strong>in</strong>d<strong>in</strong>gs between PSand T-piece wean<strong>in</strong>g may relate to differences <strong>in</strong> study design.For example, the duration of mechanical ventilation wasdifferent between the two studies, with fewer longer termpatients <strong>in</strong> the Spanish study.Non-<strong>in</strong>vasive ventilation (NIV)NIV has several advantageous features for wean<strong>in</strong>g patients,<strong>in</strong>clud<strong>in</strong>g the absence of sedative drugs, early removal of theendotracheal tube, a decrease <strong>in</strong> ventilator associated pneumonia,and better compliance with chest physiotherapy. 57These advantages have seldom been studied <strong>in</strong> a controlledmanner and to date the majority of studies have beenperformed <strong>in</strong> patients with chronic respiratory failure <strong>in</strong> aneffort to avoid endotracheal <strong>in</strong>tubation.In 22 patients referred to a specialist chronic ventilationunit for wean<strong>in</strong>g from mechanical ventilation, NIV wasrapidly tolerated <strong>in</strong> 20 and many were extubated quickly. 58 Tenpatients required nasal ventilation at night when dischargedhome. Although this study was not controlled, it certa<strong>in</strong>lyshows that patients who are difficult to wean can be extubatedand may be managed <strong>in</strong> a high dependency area us<strong>in</strong>g NIVsupport.Psychological supportPsychological disturbance occurs dur<strong>in</strong>g wean<strong>in</strong>g trials andfeel<strong>in</strong>gs of hopelessness affect performance. 59 Cl<strong>in</strong>ically, thereappears to be a gap between the physiological test<strong>in</strong>gperformed at the bedside and the actual performance of thepatient, some of which may be attributable to other factors<strong>in</strong>clud<strong>in</strong>g personality, fear, agitation, depression, andempowerment. 60Wean<strong>in</strong>g is associated with depression andtreatment may be helpful. 61 An important element of the careof patients dur<strong>in</strong>g wean<strong>in</strong>g is to devise methods of psychologicalsupport. 62 One example is to ask the patient to imag<strong>in</strong>e aparticularly strong memory and through rehearsal thisfantasy is strengthened. The memory is then used dur<strong>in</strong>gwean<strong>in</strong>g to allay anxiety and <strong>in</strong>crease tolerance of reductions<strong>in</strong> ventilatory support.


78 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Specialist wean<strong>in</strong>g unitsThe demand for ICU beds has led to the development of facilitiesspecialis<strong>in</strong>g <strong>in</strong> wean<strong>in</strong>g. 63 64 Wean<strong>in</strong>g units tend to admitpatients with s<strong>in</strong>gle organ failure who do not require complexorgan support. Such units are more cost effective, withdramatic reductions <strong>in</strong> both the fixed overheads and the consumablecost associated with wean<strong>in</strong>g. Moreover, by concentrat<strong>in</strong>geffort <strong>in</strong> a specialist area and through the developmentof protocols to guide the wean<strong>in</strong>g effort, it is possible to63 65 66decrease the time spent on mechanical ventilation.REFERENCES1 Sahn SA, Lakshm<strong>in</strong>arayan S. Bedside criteria for discont<strong>in</strong>uation ofmechanical ventilation. Chest 1973;63:1002–5.2 Tahvana<strong>in</strong>en J, Salmenpera M, Nikki P. Extubation criteria afterwean<strong>in</strong>g from <strong>in</strong>termittent mandatory ventilation and cont<strong>in</strong>uous positiveairway pressure. Crit <strong>Care</strong> Med 1983;11:702–7.3 Deml<strong>in</strong>g RH, Read T, L<strong>in</strong>d LJ, et al. Incidence and morbidity ofextubation failure <strong>in</strong> surgical <strong>in</strong>tensive care patients. Crit <strong>Care</strong> Med1988;16:573–7.4 Ely EW, Baker AM, Evans GW, et al. The prognostic significance ofpass<strong>in</strong>g a daily screen of wean<strong>in</strong>g parameters. Intensive <strong>Care</strong> Med1999;25:581–7.5 Tob<strong>in</strong> MJ, Peres W, Guenther SM, et al. The pattern of breath<strong>in</strong>g dur<strong>in</strong>gsuccessful and unsuccessful trials of wean<strong>in</strong>g from mechanical ventilation.Am Rev Respir Dis 1986;134:1111–8.6 Yang KL. Reprodcibility of wean<strong>in</strong>g parameters a need forstanderdization. Chest 1992;102:1829–32.7 Yang KL, Tob<strong>in</strong> MJ. Measurement of m<strong>in</strong>ute ventilation <strong>in</strong> ventilatordependent patients: need for standardization. Crit <strong>Care</strong> Med1991;19:49–53.8 Yang KL, Tob<strong>in</strong> MJ. A prospective study of <strong>in</strong>dexes predict<strong>in</strong>g theoutcome of trials of wean<strong>in</strong>g from mechanical ventilation. N Engl J Med1991;324:1445–50.9 Sassoon CSH, Mahutte CK. Airway occlusion pressure and breath<strong>in</strong>gpattern as predictors of wean<strong>in</strong>g outcome. Am Rev Respir Dis1993;148:860–6.10 Jabour ER, Rabil DM, Truwit J, et al. Evaluation of a new wean<strong>in</strong>g <strong>in</strong>dexbased on ventilatory endurance and the efficiency of gas excahnge. AmRev Respir Dis 1991;144:531–7.11 Cohen CA, Zagelbaum G, Gross D, et al. Cl<strong>in</strong>ical manifestations of<strong>in</strong>spiratory muscle fatigue. Am J Med 1982;73:308–16.12 Brochard L, Harf A, Lor<strong>in</strong>o H, et al. Inspiratory pressure support preventsdiaphragmatic fatigue dur<strong>in</strong>g wean<strong>in</strong>g from mechanical ventilation. AmRev Respir Dis 1989;139:513–21.13 Goldstone JC, Green M, Moxham J. Maximum relaxation rate of thediaphragm dur<strong>in</strong>g wean<strong>in</strong>g from mechanical ventilation. Thorax1994;49:54–6014 Jubran A, Tob<strong>in</strong> MJ. Pathophysiologic basis of acute respiratory distress<strong>in</strong> patients who fail a trial of wean<strong>in</strong>g from mechanical ventilation. Am JRespir Crit <strong>Care</strong> Med 1997;155:906–15.15 Purro A, Append<strong>in</strong>i L, de Gaetano A, et al. Pathophysiologicdeterm<strong>in</strong>ants of wean<strong>in</strong>g. Am J Respir Crit <strong>Care</strong> Med 2000;161:1115–23.16 Black LF, Hyatt RE. Maximal respiratory pressures: normal values andrelationship to age and sex. Am Rev Respir Dis 1969;99:696–702.17 Kim MJ, Druz WS, Danon J, et al. Mechanics of the can<strong>in</strong>e diaphragm. JAppl Physiol 1976;41:369–82.18 Similowski TB, Fleury S, Launois HP, et al. Cervical magneticstimulation: a new pa<strong>in</strong>less method for bilateral phrenic nerve stimulation<strong>in</strong> conscious humans. J Appl Physiol 1989;67:1311–8.19 Polkey MI, Duguet A, Luo Y, et al. Anterior magnetic phrenic nervestimulation: laboratory and cl<strong>in</strong>ical evaluation. Intensive <strong>Care</strong> Med2000;26:1065–75.20 Moxham J, Goldstone JC. Assessment of respiratory muscle strength <strong>in</strong>the <strong>in</strong>tensive care unit. Eur Respir J 1994;7:2057–61.21 Mar<strong>in</strong>i JJ, Smith TC, Lamb V. Estimation of <strong>in</strong>spiratory muscle strength <strong>in</strong>mechanically ventilated patients: measurement of maximum <strong>in</strong>spiratorypressure. J Crit <strong>Care</strong> 1988;1:32–8.22 Multz AS, Aldrich TK, Prezant DJ, et al. Maximal <strong>in</strong>spiratory pressure isnot a reliable test of <strong>in</strong>spiratory muscle strength <strong>in</strong> mechanically ventilatedpatients. Am Rev Respir Dis 1990;142:529–32.23 Watson AC, Hughes PD, Harris L, et al. Measurement of twitchtransdiaphragmatic, esophageal and endo-tracheal tube pressure withbilateral anterolateral magnetic phrenic nerve stimulation <strong>in</strong> patients <strong>in</strong>the <strong>in</strong>tensive care unit. Crit <strong>Care</strong> Med 2001;29:1476–8.24 Anzueto A, Peters JI, Tob<strong>in</strong> MJ, et al. Effects of prolonged controlledmechanical ventilation on diaphragmatic function <strong>in</strong> healthy adultbaboons. Crit <strong>Care</strong> Med 1997;25:1187–90.25 Bolton CF. Neuromuscular complications of sepsis. Intensive <strong>Care</strong> Med1993;19:S58–63.26 Coakley JH, Nagendran K, Yarwood GD, et al. Patterns ofneurophysiological abnormality <strong>in</strong> prolonged critical illness. Intensive<strong>Care</strong> Med 1998;24:801–7.27 Kennedy DD, Fletcher SN, Ghosh IR, et al. Reversible tetraplegia due topolyneuropathy <strong>in</strong> a diabetic patient with hyperosmolar non-ketoticcoma. Intensive <strong>Care</strong> Med 1999;25:1437–9.28 Rochester DF, Arora NS. <strong>Respiratory</strong> muscle failure. Med Cl<strong>in</strong> North Am1983;67:573–97.29 Leith DE, Bradley M. Ventilatory muscle strength and endurance tra<strong>in</strong><strong>in</strong>g.J Appl Physiol 1976;41:508–16.30 Williams AG, Rayson MP, Jubb M, et al. The ACE gene and muscleperformance. Nature 2000;403:614.31 Woods DR, Humphries SE, Montgomery HE. The ACE I/D polymorphismand human physical performance. Trends Endocr<strong>in</strong>ol Metab2000;11:416–20.32 Chieveley-Williams S, Field D, Woods D, et al. Genetic determ<strong>in</strong>ants ofrespiratory muscle tra<strong>in</strong><strong>in</strong>g. Am J Respir Crit <strong>Care</strong> Med 2001;163:A287.33 Whitelaw WA, Derenne JP, Milic-Emili J. Occlusion pressure as ameasure of respiratory center output <strong>in</strong> conscious man. Respir Physiol1975;23:181–99.34 Murciano D, Boczkowski J, Lecocguic Y, et al. Tracheal occlusionpressure: a simple <strong>in</strong>dex to monitor respiratory muscle failure <strong>in</strong> patientswith chronic obstructive pulmonary disease. Ann Intern Med1988;108:800–5.35 Kuhlen R, Hausmann S, Pappert D, et al. A new method for P 0.1measurement us<strong>in</strong>g standard respiratory equipment. Intensive <strong>Care</strong> Med1995;21:545–6.36 Sassoon CSH, Te TT, Mahutte CK, et al. Airway occlusion pressure: animportant <strong>in</strong>dicator for successful wean<strong>in</strong>g <strong>in</strong> patients with chronicobstructive pulmonary disease. Am Rev Respir Dis 1987;135:107–13.37 Montgomery AB, Holle RHO, Neagley SR, et al. Prediction of successfulventilator wean<strong>in</strong>g us<strong>in</strong>g airway occlusion pressure and hypercapnicchallenge. Chest 1987;91:496–9.38 Alberti A, Gallo F, Fongaro A, et al. P 0.1 is a useful parameter <strong>in</strong> sett<strong>in</strong>gthe level of pressure support ventilation. Intensive <strong>Care</strong> Med1995;21:547–53.39 Mancebo J, Albaladejo P, Touchard D, et al. Airway occlusion pressureto titrate positive end-expiratory pressure <strong>in</strong> patients with dynamichyper<strong>in</strong>flation. Anesthesiology 2000;93:81–90.40 Fiastro JF, Habib MP, Shon BY, et al. Comparison of standard wean<strong>in</strong>gparameters and the mechanical work of breath<strong>in</strong>g <strong>in</strong> mechanicallyventilated patients. Chest 1988;94:232–8.41 Shikora A, Bistrian BR, Borlase BC, et al. Work of breath<strong>in</strong>g: reliablepredictor of wean<strong>in</strong>g and extubation. Crit <strong>Care</strong> Med 1990;18:157–62.42 Laghi F, D’Alfonso N, Tob<strong>in</strong> MJ. Pattern of recovery from diaphragmaticfatigue over 24 hours. J Appl Physiol 1995;79:539–46.43 Hurtado FJ, Beron M, Olivera W, et al. Gastric <strong>in</strong>tramucosal pH and<strong>in</strong>tralum<strong>in</strong>al PCO 2 dur<strong>in</strong>g wean<strong>in</strong>g from mechanical ventilation. Crit <strong>Care</strong>Med 2001;29:70–6.44 Maldonado A, Bauer TT, Ferrer M, et al. Capnometric recirculation gastonometry and wean<strong>in</strong>g from mechanical ventilation. Am J Respir Crit<strong>Care</strong> Med 2000;161:171–6.45 Abalos A, Leibowitz AB, Distefano D, et al. Myocardial ischemia dur<strong>in</strong>gthe wean<strong>in</strong>g period. Am J Crit <strong>Care</strong> 1992;1:32–6.46 Srivastava S, Chatila W, Amoateng-Adjepong Y, et al. Myocardialischemia and wean<strong>in</strong>g failure <strong>in</strong> patients with coronary artery disease:an update. Crit <strong>Care</strong> Med 1999;27:2109–12.47 Lemaire F, Teboul JL, C<strong>in</strong>otti L, et al. Acute left ventricular dysfunctiondur<strong>in</strong>g unsuccessful wean<strong>in</strong>g from mechanical ventilation. Anesthesiology1988;69:171–9.48 Polese G, Massara A, Poggi R, et al. Flow trigger<strong>in</strong>g reduces <strong>in</strong>spiratoryeffort dur<strong>in</strong>g wean<strong>in</strong>g from mechanical ventilation. Intensive <strong>Care</strong> Med1995;21:682–6.49 Barnard M, Shukla A, Lovell T, et al. Esophageal-directed pressuresupport ventilation <strong>in</strong> normal volunteers. Chest 1999;115:482–9.50 Ranieri VM, Giuliani R, Mascia L, et al. Patient-ventilator <strong>in</strong>teractiondur<strong>in</strong>g acute hypercapnia: pressure-support vs. proportional assistventilation. J Appl Physiol 1996;81:426–36.51 Grasso S, Puntillo F, Mascia L, et al. Compensation for <strong>in</strong>crease <strong>in</strong>respiratory workload dur<strong>in</strong>g mechanical ventilation: pressure-supportversus proportional-assist ventilation. Am J Respir Crit <strong>Care</strong> Med2000;161:819–26.52 Fabry B, Haberthur C, Zappe D, et al. Breath<strong>in</strong>g pattern and additionalwork of breath<strong>in</strong>g <strong>in</strong> spontaneously breath<strong>in</strong>g patients with differentventilatory demands dur<strong>in</strong>g <strong>in</strong>spiratory pressure support ventilation andautomatic tube compensation. Intensive <strong>Care</strong> Med 1997;23:545–52.53 Pepe PE, Mar<strong>in</strong>i JJ. Occult positive end-expiratory pressure <strong>in</strong>mechanically ventilated patients with airflow obstruction: the auto-PEEPeffect. Am Rev Respir Dis 1982;126:166–70.54 Tob<strong>in</strong> M. PEEP, auto-PEEP and waterfalls. Chest 1989;96:449–51.55 Brochard L, Rauss A, Benito S, et al. Comparison of three methods ofgradual withdrawal from ventilatory support dur<strong>in</strong>g wean<strong>in</strong>g frommechanical ventilation. Am J Respir Crit <strong>Care</strong> Med 1994;150:896–903.56 Esteban A, Frutos F, Tob<strong>in</strong> MJ, et al. A comparison of four methods ofwean<strong>in</strong>g patients from mechanical ventilation. N Engl J Med1995;332:345–50.57 Goldstone JC. Non <strong>in</strong>vasive ventilation <strong>in</strong> the <strong>in</strong>tensive care unit andoperat<strong>in</strong>g theatre. In: Simonds AK, ed. Non <strong>in</strong>vasive respiratory support.London: Chapman & Hall, 1996.58 Udwadia ZF, Santis GK, Steven MH, et al. Nasal ventilation to facilitatewean<strong>in</strong>g <strong>in</strong> patients with chronic respiratory <strong>in</strong>sufficiency. Thorax1992;47:715–8.59 Moody LE, Lowry L, Yarandi H, et al. Psychophysiologic predictors ofwean<strong>in</strong>g from mechanical ventilation <strong>in</strong> chronic bronchitis andemphysema. Cl<strong>in</strong> Nurs Res 1997;6:311–30.60 Engstrom CP, Persson LO, Larsson S, et al. Health-related quality of life<strong>in</strong> COPD: why both disease-specific and generic measures should beused. Eur Respir J 2001;18:69–76.


Difficult wean<strong>in</strong>g 7961 Rothenhausler HB, Ehrentrault S, von Gegenfeld G, et al. Treatment ofdepression with methylphenidate <strong>in</strong> patients difficult to wean frommechanical ventilation <strong>in</strong> the <strong>in</strong>tensive care unit. J Cl<strong>in</strong> Psychiatry2000;61:750–5.62 Miller JF. Hope-<strong>in</strong>spir<strong>in</strong>g strategies of the critically ill. Appl Nurs Res1989;2:23–9.63 Dasgupta A, Rice R, Mascha E, et al. Four-year experience with a unitfor long-term ventilation (respiratory special care unit) at the ClevelandCl<strong>in</strong>ic Foundation. Chest 1999;116:447–55.64 Seneff MG Wagner D, Thompson D, et al. The impact of long-termacute-care facilities on the outcome and cost of care for patientsundergo<strong>in</strong>g prolonged mechanical ventilation. Crit <strong>Care</strong> Med2000;28:342–50.65 Henneman E, Dracup K, Ganz T, et al. Effect of a collaborativewean<strong>in</strong>g plan on patient outcome <strong>in</strong> the critical care sett<strong>in</strong>g. Crit <strong>Care</strong>Med 2001;29:297–303.66 Marelich GP, Mur<strong>in</strong> S, Battistella F, et al. Protocol wean<strong>in</strong>g ofmechanical ventilation <strong>in</strong> medical and surgical patients by respiratorycare practitioners and nurses: effect on wean<strong>in</strong>g time and<strong>in</strong>cidence of ventilator-associated pneumonia. Chest 2000;118:459–67.


12 <strong>Critical</strong> care management of respiratory failureresult<strong>in</strong>g from chronic obstructive pulmonary diseaseA C Davidson.............................................................................................................................Acute episodes of respiratory failure <strong>in</strong>patients with chronic obstructive pulmonarydisease (COPD) account for 5–10% ofemergency medical admissions to hospital andfailure of first l<strong>in</strong>e treatment is a common reasonfor referral to the <strong>in</strong>tensive care unit (ICU). Inrecent years such patients have become bettercharacterised and the driv<strong>in</strong>g force <strong>in</strong> this hasbeen the need to def<strong>in</strong>e those suitable fortreatment by non-<strong>in</strong>vasive ventilation (NIV)rather than <strong>in</strong>tubation. Bacterial <strong>in</strong>fection hastraditionally been considered aetiologically dom<strong>in</strong>antbut its importance has been over stressed.Heart failure, cardiac arrhythmia, pulmonaryembolism, and “uncerta<strong>in</strong> causes” are common. 1Acute deterioration precipitated by viral <strong>in</strong>fectionis <strong>in</strong>creas<strong>in</strong>gly recognised. 2Consideration of allthe ways <strong>in</strong> which the co-morbidity of COPD<strong>in</strong>fluences ICU management is beyond the scopeof this chapter. Its presence affects both ventilatorstrategy and the outcome of patients after electiveor emergency surgery. COPD also contributes todelay <strong>in</strong> the wean<strong>in</strong>g of patients from mechanicalventilation. 3–5This chapter focuses on the commonproblem of the patient with respiratory failurearis<strong>in</strong>g from an exacerbation of chronicairflow obstruction.In the past the perception that survival ofpatients with COPD was poor, especially longterm, comb<strong>in</strong>ed with <strong>in</strong>sufficient provision ofcritical care facilities <strong>in</strong> the UK has limited accessto the ICU. This was especially so when “endstage” COPD was considered to be present. Thismight be <strong>in</strong>ferred if there is no apparent precipitat<strong>in</strong>gcause such as pneumonia or pneumothorax.In these circumstances, as there is no apparentreversible cause, it could be argued thatrecovery is unlikely. Survival follow<strong>in</strong>g mechanicalventilation (MV) is, however, better <strong>in</strong> theabsence of a major precipitat<strong>in</strong>g cause. 1Thisapparent paradox probably arises because patientswho require a longer period of ventilatorysupport—which will be the case if, for <strong>in</strong>stance,pneumonia is present—are exposed to the secondarycomplications of ICU admission. Just assurvival <strong>in</strong> the acute respiratory distress syndromeis more closely related to associated multiorganfailure or nosocomial <strong>in</strong>fection than to theseverity of the <strong>in</strong>itial lung <strong>in</strong>jury, 6 so thecomplications that arise dur<strong>in</strong>g the ICU stay of apatient with COPD may have a greater <strong>in</strong>fluenceon outcome than the severity of airflow obstruction.Nevertheless, age, severity of airflow obstruction,co-morbidity, and general preadmissionhealth status are important <strong>in</strong>determ<strong>in</strong><strong>in</strong>g survival. 1 7–10There are national and <strong>in</strong>ternational differences<strong>in</strong> both the <strong>in</strong>stitution and withdrawal ofMV <strong>in</strong> COPD. The prevalence of COPD <strong>in</strong> thecommunity and admission practices will determ<strong>in</strong>ehow costly ICU management of COPD willbe locally. For <strong>in</strong>stance, <strong>in</strong> one patient simulationstudy there was considerable variation <strong>in</strong> prognosticestimates by respondents for identicalcl<strong>in</strong>ical scenarios. 11 The prognosis predicted markedlyaffected will<strong>in</strong>gness to offer hypothetical ICUadmission and the estimates were uniformlyworse than US outcome prediction data wouldsuggest. In one UK report withdrawal of treatmentwas the most common cause of death, 12while <strong>in</strong> an Italian study two thirds of patientswere still be<strong>in</strong>g actively weaned 60 days afteradmission. 13 The European Human Rights Act 14might <strong>in</strong>crease the pressure to admit patients tothe ICU and there is some evidence that this isoccurr<strong>in</strong>g. This is probably desirable <strong>in</strong> the UKwhere, <strong>in</strong> the past, respiratory physicians may nothave sufficiently championed the cause of thepatient with COPD. Short term survival follow<strong>in</strong>g1 7–10 15<strong>in</strong>vasive MV can be expected <strong>in</strong> 63–86%, afigure well above that for unplanned medicaladmissions. Although long term survival is lessgood—<strong>in</strong> one study 52%, 42%, and 37% at 1, 2 and3 years, respectively 15 —this is similar to survivalfollow<strong>in</strong>g myocardial <strong>in</strong>farction when left ventriculardysfunction is present. A better long termoutcome is reported follow<strong>in</strong>g an episode ofrespiratory failure managed with NIV. 16–18 It is alsopossible that survival may subsequently beimproved by domiciliary NIV <strong>in</strong> selectedpatients, 19 although <strong>in</strong>terim results of controlledtrials of domiciliary ventilation have been negative.Despite reasonable survival to hospital discharge,the decision to admit to the ICU <strong>in</strong>advanced cases is frequently difficult 20and <strong>in</strong>volvesbalanc<strong>in</strong>g health status with an estimate ofexpectation of survival and quality of life issues.This often needs to be established on the basis ofscant <strong>in</strong>formation and <strong>in</strong> the face of sometimesunreasonable expectations from distraught relatives.Furthermore, these difficult decisions commonlyfall on the least experienced doctors ashospital presentation is often “out of hours”. Arecent report found that co-morbidity, need forMV beyond 72 hours, and failure follow<strong>in</strong>g extubationwere strong predictors of a pooroutcome. 10Survival to discharge for the wholegroup (166 patients) was 72% and <strong>in</strong>creased to88% <strong>in</strong> those without co-morbidity. This reporttherefore suggests that an active policy, with earlyreview once MV has been <strong>in</strong>itiated, may beappropriate. Ideally, the value and complicationsof MV should be discussed prior to the medicalemergency. 21 Such discussion may be difficult to<strong>in</strong>itiate <strong>in</strong> the outpatient cl<strong>in</strong>ic and primary careis probably a better sett<strong>in</strong>g. The recovery periodfollow<strong>in</strong>g a period of MV is an ideal opportunityand it is well suited for <strong>in</strong>clusion <strong>in</strong> rehabilitationprogrammes, 22 but resistance to such discussion iscommon, at least <strong>in</strong> the UK. 23


<strong>Critical</strong> care management of respiratory failure result<strong>in</strong>g from COPD 81Box 12.1 Mechanisms <strong>in</strong>volved <strong>in</strong> decompensatedCOPDIncreased resistive load• Widespread airflow obstructionDecreased respiratory system compliance• High lung volumeDynamic hyper<strong>in</strong>flation• Shortened expiratory time• Poorly empty<strong>in</strong>g lung unitsReduced power of respiratory pump• Impaired mechanical efficiency• Effects of acidosis and hypoxaemiaImpaired drive• Sleep deprivation•CO 2narcosisRECOGNISING THE NEED FOR VENTILATORYSUPPORTThe recognition that MV is required is commonly an “end ofthe bed” assessment by an experienced cl<strong>in</strong>ician. No one cl<strong>in</strong>icalfeature or <strong>in</strong>vestigation is absolute except respiratoryarrest or loss of consciousness. 20In most cases failure toimprove with medical treatment <strong>in</strong> the hours follow<strong>in</strong>gadmission triggers ICU referral. Late failure several days afteradmission to hospital is less common and may <strong>in</strong>dicate aworse prognosis. 24 In many, a downward spiral of <strong>in</strong>creas<strong>in</strong>gcarbon dioxide retention and sleep deprivation eventuallyleads to impaired consciousness as the ventilatory pump failsto cope with the <strong>in</strong>creased respiratory “load”. The mechanisms<strong>in</strong>volved <strong>in</strong> decompensated COPD (box 12.1) are an<strong>in</strong>crease <strong>in</strong> airflow resistance related to widespread bronchialwall <strong>in</strong>flammation and progressive dynamic hyper<strong>in</strong>flationthat maximises expiratory flow at the cost of <strong>in</strong>creas<strong>in</strong>g25 26<strong>in</strong>spiratory muscle work.In addition to this resistive work, reduced respiratorysystem compliance associated with operat<strong>in</strong>g towards the topof the pressure-volume curve is comb<strong>in</strong>ed with decreasedmechanical efficiency of the diaphragm at high lung volumes.Premature expiratory closure of small airways, either becauseof lack of support <strong>in</strong> emphysema or functional narrow<strong>in</strong>gfrom airway <strong>in</strong>flammation or smooth muscle contraction,results <strong>in</strong> impaired gas exchange. Positive end expiratory<strong>in</strong>trathoracic pressure—so called <strong>in</strong>tr<strong>in</strong>sic PEEP—furtherloads the <strong>in</strong>spiratory muscles. Recruitment of abdom<strong>in</strong>almuscles dur<strong>in</strong>g expiration is common. This may not <strong>in</strong>creaseexpiratory airflow as dynamic expiratory resistance, the chokeeffect, may occur and will then only accentuate gas trapp<strong>in</strong>g.Sudden relaxation of abdom<strong>in</strong>al muscle contraction at endexpiration, a feature of the fail<strong>in</strong>g patient, may be employed tounload the <strong>in</strong>spiratory muscles by natural recoil at the start of<strong>in</strong>spiration. 25Additionally, as respiratory rate <strong>in</strong>creases, gasexchange is further impaired by <strong>in</strong>creased dead space ventilationand further muscle load<strong>in</strong>g is the result of additionaldynamic hyper<strong>in</strong>flation as expiratory time shortens. Increasedpulmonary vascular resistance and reduced venous returnimpair right heart function and decrease cardiac output. Inadequatesystemic oxygen delivery to meet energy requirementsthen adds a metabolic component to the respiratory acidosis.Hypoxaemia and acidosis further impair respiratory musclefunction. 27 Unless controlled oxygen therapy, bronchodilators,and fluid replacement can both improve gas exchange andreduce the load on the respiratory muscles, mechanical ventilatorysupport will be required.At what stage <strong>in</strong> this process should <strong>in</strong>tervention occur andhow can this state be recognised? The need for MV is betterpredicted by arterial pH and carbon dioxide (PaCO 2) levels thanthe degree of hypoxaemia. For <strong>in</strong>stance, <strong>in</strong> a study <strong>in</strong>vestigat<strong>in</strong>gthe value of NIV <strong>in</strong> acute COPD, 74% of patientsBox 12.2 Contra<strong>in</strong>dications to NIV• Impaired consciousness (except O 2<strong>in</strong>duced)• Uncooperative patient• Significant vomit<strong>in</strong>g risk• Cardiac arrythmia or hypotension (if severe)• Profound hypoxaemia (unless <strong>in</strong> ICU)• Excessive secretionsrandomised to management without NIV (mean pH 7.26)reached the a priori criteria for tracheal <strong>in</strong>tubation. 16 The useof uncontrolled oxygen therapy may have precipitated furtherdeterioration <strong>in</strong> this study, result<strong>in</strong>g <strong>in</strong> a high frequency ofventilatory support. In a similar study which <strong>in</strong>cluded lessseverely affected patients, 1727% of patients with a wardadmission pH of 7.25–7.35 progressed to fulfil <strong>in</strong>tubation criteriacompared with 36% of those with a pH of


82 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Flow PawFigure 12.1 Progressive hyper<strong>in</strong>flation results from either excessivetidal volume or <strong>in</strong>sufficient expiratory time, or both. If mach<strong>in</strong>edelivered breath occurs before flow has ceased (positive endexpiratory pressure, PEEP), peak airway pressure (Paw) will <strong>in</strong>creaseand tidal volume will fall as progressive hyper<strong>in</strong>flation develops. Ifthis results from premature airway collapse, externally applied PEEPwill <strong>in</strong>crease tidal volume without an <strong>in</strong>crease <strong>in</strong> Paw.usually required but leak<strong>in</strong>g is then more problematic and thismay also affect ventilator trigger<strong>in</strong>g. Not uncommonly,apparent early success is not matched by a fall <strong>in</strong> the PaCO 2.Rebreath<strong>in</strong>g with the <strong>in</strong>creased dead space of a face mask maybe the cause, but an <strong>in</strong>effective cough and reta<strong>in</strong>ed bronchialsecretions are more commonly responsible. In these situationsa nose mask and ch<strong>in</strong> strap may be beneficial by allow<strong>in</strong>gspontaneous cough<strong>in</strong>g. Excessive secretions may also causeimpairment of gas exchange result<strong>in</strong>g <strong>in</strong> refractoryhypoxaemia.Monitor<strong>in</strong>g the impact of NIV is essential. A greater expansionof the chest dur<strong>in</strong>g assisted breath<strong>in</strong>g should be the primaryaim with good match<strong>in</strong>g of the patient’s breath<strong>in</strong>g effortwith the ventilator or effective ventilation with mach<strong>in</strong>e timedbreaths. Whichever mode is employed, a reduction <strong>in</strong> respiratorydistress is an important prognostic feature and both cardiacand respiratory rate will fall with a gradual reversal ofrespiratory acidosis when NIV is effective. In our experiencethe need for frequent arterial blood gas analysis and appropriatemonitor<strong>in</strong>g of physiological variables is best provided <strong>in</strong>the HDU or level 2 facility. In some hospitals, where specialistmedical wards are available, NIV may be provided <strong>in</strong> level 1beds. This is particularly the case when used <strong>in</strong> patients withless physiological disturbance such as a higher pH, us<strong>in</strong>gspontaneous mode only ventilators. 17 With <strong>in</strong>creased recognitionof the value of NIV <strong>in</strong> such patients, greater availability ofequipment and the necessary skill mix of staff required, NIVwill hopefully be effectively used outside the ICU. Excellentreviews and comprehensive guidel<strong>in</strong>es for NIV are29 30available.Tracheal <strong>in</strong>tubation and mechanical ventilationImpend<strong>in</strong>g cardiorespiratory arrest is <strong>in</strong>dicated by profoundhypoxaemia on disconnection from oxygen or NIV, significanthypotension, or an altered mental state. Immediate <strong>in</strong>tubationmay then be required. As cardiovascular collapse is commonafter <strong>in</strong>tubation, transfer of the spontaneously breath<strong>in</strong>gpatient to the ICU may, however, be safer. Collapse arises froma comb<strong>in</strong>ation of reduced venous return secondary to positive<strong>in</strong>trathoracic pressure, and direct vasodilation and reducedsympathetic tone <strong>in</strong>duced by sedative agents. Before <strong>in</strong>tubationpre-oxygenation is essential. Intubation with the rapidsequence <strong>in</strong>duction and cricoid pressure to reduce the risk ofaspiration should ideally be performed by an experienced cl<strong>in</strong>ician.Suxamethonium is classically used for muscle relaxationas its short effect makes it safer <strong>in</strong> the event of a failureto <strong>in</strong>tubate. Concerns about hyperkalaemic cardiac arrest 31have led to the <strong>in</strong>creased use of short act<strong>in</strong>g non-depolaris<strong>in</strong>gagents such as rocuronium. Doubts about the effectiveness ofcricoid pressure <strong>in</strong> prevent<strong>in</strong>g aspiration 32 have also resulted <strong>in</strong>a move to “head up” non-paralytic <strong>in</strong>tubation. This is a highrisk period <strong>in</strong> which profound hypotension may result <strong>in</strong> cardiacarrhythmia or arrest. Unless hypotension resolves rapidlywith fluid replacement, cardiac tamponade <strong>in</strong>duced by hyper<strong>in</strong>flation(bagg<strong>in</strong>g) should be suspected. In these circumstances,temporary disconnection of the endotracheal tubefrom positive pressure will lead to a return <strong>in</strong> cardiac output.Controlled mechanical ventilationHav<strong>in</strong>g secured the airway and corrected hypoxaemia,management is aimed at correct<strong>in</strong>g the respiratory acidosiswhile avoid<strong>in</strong>g further hyper<strong>in</strong>flation. This is best achieved bya comb<strong>in</strong>ation of slow MV with a prolonged expiratory timeand a limited tidal volume. A degree of permissive hypercapniais well tolerated, 33 while bronchial toilet andbronchodilation—usually with a comb<strong>in</strong>ation of <strong>in</strong>travenousand nebulised agents—will improve alveolar ventilation. Thebenefit of steroids has been established <strong>in</strong> acute COPD, 34 butthese probably take hours to effect an improvement. Inotropessuch as ep<strong>in</strong>ephr<strong>in</strong>e (adrenal<strong>in</strong>e) are well known to cause ametabolic acidosis but this may also occur with β 2stimulants,largely by stimulat<strong>in</strong>g metabolism. 35 In the first 12–24 hours ofMV, paralysis is normally required. This reduces the chest andabdom<strong>in</strong>al wall contributions to the reduced respiratorysystem compliance and prevents patient ventilator dysynchronyor fight<strong>in</strong>g, which will impair alveolar ventilation andresult <strong>in</strong> high airway pressures. Airflow resistance and hyper<strong>in</strong>flationboth contribute to the need for high <strong>in</strong>flationpressures to achieve an effective tidal volume and these mayprogressively <strong>in</strong>crease if the set ventilatory parameters arecaus<strong>in</strong>g further hyper<strong>in</strong>flation (see fig 12.1). The immediatecomplications of high airway pressures are impaired cardiacoutput, pneumothorax, and mediast<strong>in</strong>al and subcutaneousemphysema.The ventilator may be set either to control volume or pressure.In volume controlled ventilation, conventional sett<strong>in</strong>gswould be a tidal volume of 8–12 ml/kg at a frequency of 10–14breaths/m<strong>in</strong>ute and an <strong>in</strong>spiratory:expiratory (I: E) ratio of1:2.5 or 3.0. The disadvantage of volume control is the potentialfor high airway pressures; pressure limitation providesprotection and is available on most modern mach<strong>in</strong>es.Alternatively, pressure controlled ventilation may be preferredas high airway pressures are avoided and the <strong>in</strong>spiratory flowpattern, which better resembles normal breath<strong>in</strong>g, tends toequalise ventilation between lung units rather than preferentiallyventilat<strong>in</strong>g, and possibly over<strong>in</strong>flat<strong>in</strong>g, the less obstructed(or faster fill<strong>in</strong>g and empty<strong>in</strong>g) lung units (fig 12.2).This mode of ventilation has ga<strong>in</strong>ed favour as it has becomerecognised that additional lung <strong>in</strong>jury may result fromrelatively high tidal volumes that accompany the use of highventilatory pressures rather than from high airway pressureper se. 36 Although the importance of this concept has so faronly been demonstrated <strong>in</strong> ARDS where a reduced mortalityaccompanies limited tidal volume ventilation, 37the samemechanisms probably operate <strong>in</strong> other <strong>in</strong>dications for MV.The use of PEEP when ventilat<strong>in</strong>g patients with airflowobstruction is controversial. It was argued that externallyapplied positive airway pressure (PEEPe) would be harmful asit would <strong>in</strong>crease hyper<strong>in</strong>flation. When small airway collapsedevelops dur<strong>in</strong>g expiration from the structural changes associatedwith emphysema, the application of PEEPe will reducegas trapp<strong>in</strong>g by stent<strong>in</strong>g open the airways. The value of PEEPeto offset <strong>in</strong>tr<strong>in</strong>sic PEEP is also important when support<strong>in</strong>gspontaneous breath<strong>in</strong>g and is considered below. In controlledventilation, a practical method to judge its use is to monitortidal volume and airway pressure (PEEPi). As PEEPe isapplied, tidal volume will <strong>in</strong>crease without an <strong>in</strong>crease <strong>in</strong> airwaypressure until PEEPe exceeds PEEPi . Intr<strong>in</strong>sic PEEP canbe measured by measur<strong>in</strong>g plateau pressure follow<strong>in</strong>g aprolonged expiratory pause, so called static PEEPi (see fig12.3). 38 Intr<strong>in</strong>sic PEEP will, however, be overestimated if thereis active abdom<strong>in</strong>al expiratory effort. Accurate measurementof dynamic PEEP (PEEPi dyn) <strong>in</strong> spontaneously breath<strong>in</strong>g


<strong>Critical</strong> care management of respiratory failure result<strong>in</strong>g from COPD 83End expirationEnd <strong>in</strong>spirationNormal lung(equal ventilation to lung units)Partially obstructed lung unitreceives less ventilation andhas higher end expiratoryvolume with volume control.Normal lung may beover<strong>in</strong>flatedMore equal ventilation to bothunits with pressure controlFigure 12.2Improved distribution of ventilation with pressure controlled mandatory ventilation.A 215B 1515Figure 12.3 Measurement of <strong>in</strong>tr<strong>in</strong>sic positive end expiratorypressure (PEEPi). (A) Tracheal end expiratory pressure is low withopen expiratory port. (B) Measurement of auto-PEEP or <strong>in</strong>tr<strong>in</strong>sic PEEPby occlusion of expiratory port at end expiration. The balloonrepresents the ventilated lung with expiratory flow obstruction.Intr<strong>in</strong>sic PEEP will be overestimated if there is active abdom<strong>in</strong>almuscle contraction.patients is more difficult and requires simultaneous measurementof gastric pressure (Pga) when PEEPi = PEEPi dyn –Pga. 39Assisted modes of ventilatory supportIn many patients correction of acidosis and the need for a high<strong>in</strong>spired oxygen concentration (FiO 2) rapidly resolves. Spontaneousbreath<strong>in</strong>g may still be <strong>in</strong>adequate but partial ventilatorysupport is possible with synchronised <strong>in</strong>termittent mandatoryventilation (SIMV). It provides a background of mach<strong>in</strong>edelivered breaths whilst spontaneous breath<strong>in</strong>g effort isenhanced by positive pressure (pressure support) act<strong>in</strong>g to<strong>in</strong>crease the tidal volume of such triggered breaths. Thesebreaths then delay the next mach<strong>in</strong>e delivered breath(synchronisation). It would seem an attractive mode dur<strong>in</strong>gthe wean<strong>in</strong>g period. Excessive amounts of respiratory workmay, however, occur with SIMV 40 unless attention is paid tooptimise trigger<strong>in</strong>g by adjustment of PEEPe, and to titrate thedegree of pressure support. At this po<strong>in</strong>t, know<strong>in</strong>g the level ofPEEPi is useful but more difficult to measure. 39 By adjust<strong>in</strong>gPEEPe to approximate PEEPi, the <strong>in</strong>spiratory pressurerequired to trigger a breath can be reduced (gas flow cannotbeg<strong>in</strong> until a negative deflection <strong>in</strong> airway pressure isregistered by the ventilator). Flow triggers are more sensitivethan pressure triggers but are only available on newer ventilators.A bias flow, usually 1–5 l/m<strong>in</strong>, is provided by the ventilatordur<strong>in</strong>g expiration. When the flow signal changes with theonset of <strong>in</strong>spiration, the ventilator is triggered to deliver pressuresupport.An additional cause of patient distress may, however, occurbefore the ventilator beg<strong>in</strong>s to provide flow. If the <strong>in</strong>spiratoryflow rate, which commonly has a default sett<strong>in</strong>g of 60–80l/m<strong>in</strong>, is <strong>in</strong>sufficient for patient demand (which may be up to120 l/m<strong>in</strong>), a sense of “air hunger” occurs which may result <strong>in</strong>premature cessation of <strong>in</strong>spiratory effort. On the other hand, ifthe mandatory mach<strong>in</strong>e delivered breaths are too large or toolong, expiratory effort will occur before the end of <strong>in</strong>spirationand result <strong>in</strong> unnecessary work and patient distress. This phenomenonalso occurs if the level of support is excessive (toensure a “normal” tidal volume). Disentangl<strong>in</strong>g the primaryproblem lead<strong>in</strong>g to patient-ventilator dysynchrony versusmore straightforward causes such as the discomfort of theendotracheal tube or anxiety may be difficult. 41Accord<strong>in</strong>gly,accept<strong>in</strong>g a high respiratory rate and small tidal volume withpressure support may be preferable to SIMV. With eithermode, exam<strong>in</strong>ation of the real time pressure and volumetraces, available on modern ventilators, will provide clues tothe sett<strong>in</strong>g of PEEPe, the presence of <strong>in</strong>spiratory effort thatfails to produce trigger<strong>in</strong>g or of expiratory effort before theend of <strong>in</strong>spiration. Occasionally, however, direct measurementof the oesophageal or gastric pressure is necessary.One disadvantage of pressure support occurs dur<strong>in</strong>g sleepwhen prolonged apnoeic periods, potentiated by lower<strong>in</strong>gPaCO 2below normal, may result <strong>in</strong> repeated ventilator alarms.It is our preference to ensure adequate ventilatory support andallow restorative sleep at night us<strong>in</strong>g a controlled mode andthen progressively reduce the degree of pressure support dur<strong>in</strong>gthe day. An alternative is to use timed bi-level pressuresupport 42which ensures adequate ventilation dur<strong>in</strong>g sleepand, if adjusted appropriately, comfortable pressure supportby day. As this method does not <strong>in</strong>volve triggered breath<strong>in</strong>g (itcan be conceptualised as CPAP with a timed higher pressureperiod superimposed), <strong>in</strong>advertent trigger<strong>in</strong>g dur<strong>in</strong>g suction<strong>in</strong>gor cough<strong>in</strong>g is avoided—another mechanism for patientsbecom<strong>in</strong>g distressed. With bi-level pressure support (BiPAP)there is the potential for <strong>in</strong>creas<strong>in</strong>g hyper<strong>in</strong>flation if <strong>in</strong>appropriatetim<strong>in</strong>g results <strong>in</strong> expiratory effort dur<strong>in</strong>g the high pressureperiod.Although conventional extubation criteria 43 such as an FiO 2of 10 ml/kg can be encountered soonafter admission to the ICU, up to 30% of patients with COPDmeet<strong>in</strong>g such criteria fail <strong>in</strong> the period follow<strong>in</strong>gextubation. 44A significant delay <strong>in</strong> the wean<strong>in</strong>g process orfailure follow<strong>in</strong>g extubation may result from airflow obstruction,cont<strong>in</strong>ued hypersecretion, impaired left ventricular function,or over-sedation. 45 Propofol, a short act<strong>in</strong>g sedative, mayallow good titration of sedation <strong>in</strong> the period lead<strong>in</strong>g up toextubation and permit good synchrony between patient and


84 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>ventilator, an essential requirement when decid<strong>in</strong>g upon thelikelihood of successful extubation. Nava et al 13 have providedevidence that early extubation is possible <strong>in</strong> patients whowould be at high risk of post extubation failure by us<strong>in</strong>g NIVas a bridge. Although another study was unable to confirmmore successful wean<strong>in</strong>g with this approach, the use of NIVhad some benefits. 46 It is our practice to aim for extubation atthe 48–72 hours “w<strong>in</strong>dow of opportunity” before secondary<strong>in</strong>fections or other complications occur. Should this then fail,especially if stridor from glottic or supraglottic oedema ispresent after extubation, we proceed immediately to percutaneoustracheostomy on days 3–4 (see below).NON-VENTILATORY CONSIDERATIONSSteroids are useful <strong>in</strong> speed<strong>in</strong>g the resolution of airway<strong>in</strong>flammation but are implicated <strong>in</strong> the myopathy associatedwith critical illness 47 and our practice is to taper the dose rapidly.The value of nebulised steroids has not been established<strong>in</strong> this situation. Adequate nutritional support is essential butshould not be excessive. There is no conv<strong>in</strong>c<strong>in</strong>g evidence thatmanipulation of the metabolic costs of feed<strong>in</strong>g by energy substitutionwith fats speeds wean<strong>in</strong>g. The risk of nosocomialpneumonia <strong>in</strong>creases with longer ventilatory support. Nurs<strong>in</strong>g<strong>in</strong> the head up position may reduce the <strong>in</strong>cidence, 48 while therisk/benefits of ulcer prophylaxis 49 and gut sterilisation 50 cont<strong>in</strong>ueto be debated. Adequate hydration is clearly important<strong>in</strong> mobilis<strong>in</strong>g tenacious secretions. Inhaled or nebulised β 2stimulants are more effective than sal<strong>in</strong>e <strong>in</strong> aid<strong>in</strong>g sputumclearance, and mucolytics such as N-acetyl cyste<strong>in</strong>e or DNasemay occasionally be helpful. High <strong>in</strong>spired oxygen (>50%)<strong>in</strong>activates N-acetyl cyste<strong>in</strong>e but is rarely required <strong>in</strong> COPD.The value of cough assist devices (Exsufflator; Emerson & Co)is <strong>in</strong>creas<strong>in</strong>gly recognised <strong>in</strong> neuromuscular causes of respiratoryfailure when cough is <strong>in</strong>effective and may prove to be ofuse <strong>in</strong> COPD.In the past the morbidity and <strong>in</strong>convenience of surgical tracheostomiesoften resulted <strong>in</strong> prolonged ventilation with anendotracheal tube. The advantages of the percutaneous techniqueand the recognition that the result<strong>in</strong>g comfort of a tracheostomyallows less sedation has resulted <strong>in</strong> percutaneoustracheostomy be<strong>in</strong>g performed earlier <strong>in</strong> the cl<strong>in</strong>ical course. Itallows <strong>in</strong>termittent ventilatory support and access to thelower respiratory tract for suction<strong>in</strong>g when ventilatorysupport is no longer required. A further advantage is thatrehabilitation can be more active without the risk of <strong>in</strong>advertentextubation. Fenestrated tracheostomy tubes will providephonation, which improves communication and is an importantmilestone when wean<strong>in</strong>g. One-way speak<strong>in</strong>g valves (PasseyMuir) provide an even better voice and can be <strong>in</strong>serted <strong>in</strong>tothe s<strong>in</strong>gle lumen ventilation circuits employed with bi-levelventilators when used to support patients dur<strong>in</strong>g the wean<strong>in</strong>gprocess.WEANING FAILUREThis aspect of management is considered <strong>in</strong> the chapter byGoldstone. Wean<strong>in</strong>g protocols 51 may be helpful, pr<strong>in</strong>cipally byidentify<strong>in</strong>g patients who no longer require ventilatorysupport. COPD accounts for approximately 25% of wean<strong>in</strong>gfailures, def<strong>in</strong>ed as those still ventilator dependent 3 weeks ormore after recovery from the condition precipitat<strong>in</strong>g ICUadmission. 3–5 The negative aspects of a cont<strong>in</strong>ued stay <strong>in</strong> themodern ICU environment, especially when only s<strong>in</strong>gle organ(respiratory) failure persists, justifies consider<strong>in</strong>g referral tospecialist wean<strong>in</strong>g centres 45 which may be regionally provided<strong>in</strong> the future. On the other hand, sensitivity to the wishes ofpatients and/or judicious withhold<strong>in</strong>g of an escalation <strong>in</strong>therapy when deterioration occurs is also good practice <strong>in</strong> theirreversibly ventilator dependent patient. 14REFERENCES1 Seneff MG, Wagner DP, Wagner RP, et al. Hospital and 1 year survivalof patients admitted to <strong>in</strong>tensive care units with acute exacerbations ofchronic obstructive disease. JAMA 1995;274:1852–7.2 Seemungal TA, Harper-Owen R, Bhowmik A, et al. Detection ofrh<strong>in</strong>ovirus <strong>in</strong> <strong>in</strong>duced sputum at exacerbations of chronic obstructivepulmonary disease. Eur Respir J 2000;16:677–83.3 Hamid S, Noonan YM, Williams AJ, et al. An audit of wean<strong>in</strong>g frommechanical ventilation <strong>in</strong> a UK wean<strong>in</strong>g centre. Thorax 1999;54:86P.4 Sche<strong>in</strong>horn DJ, Chao DC, Stearn-Hassenpflug M, et al. Outcomes <strong>in</strong>post ICU mechanical ventilation. Treatment of 1123 patients at aregional wean<strong>in</strong>g centre. Chest 1997;111:1654–9.5 Pilcher DV, Hamid S, Williams AJ, et al. Outcomes, cost and long termsurvival of patients referred to a regional centre for wean<strong>in</strong>g frommechanical ventilation. Br J Anaesth 2002;89:361–2P.6 Squara P, Dha<strong>in</strong>aut JF, Artigas A, et al. Hemodynamic profile <strong>in</strong> severeARDS: results of the European Collaborative ARDS study. Intensive <strong>Care</strong>Med 1998;24:1018–28.7 Hudson LD. Survival data <strong>in</strong> patients with acute or chronic lung diseaserequir<strong>in</strong>g mechanical ventilation. Am Rev Respir Dis 1989;140:519–24.8 Gracey DR, Naessens JM, Krishan I, et al. Hospital and post hospitalsurvival among patients with COPD who require mechanical ventilationfor acute respiratory failure. Chest 1989;101:211–4.9 Soo Hoo GW, Hakiman I, Santiago SM. Hypercapnic respiratory failure<strong>in</strong> COPD patients. Response to therapy. Chest 2000;117:169–77.10 Nev<strong>in</strong>s ML, Epste<strong>in</strong> SK. Predictors of outcome for patients with COPDrequir<strong>in</strong>g <strong>in</strong>vasive mechanical ventilation. Chest 2001;119:1840–9.11 Wildman MJ, Odea J, Kostopoulou O, et al. Variation <strong>in</strong> <strong>in</strong>tubationdecisions for patients with COPD <strong>in</strong> one critical care network. QJM2003;96:583–91.12 Hill AT, Hopk<strong>in</strong>son RB, Stapleforth DE. Ventilation <strong>in</strong> a Birm<strong>in</strong>gham<strong>in</strong>tensive care unit 93–95: outcome for patients with chronic obstructivepulmonary disease. Respir Med 1998;92:156–61.13 Nava S, Ambros<strong>in</strong>o N, Cl<strong>in</strong>i E, et al. Non <strong>in</strong>vasive mechanicalventilation <strong>in</strong> the wean<strong>in</strong>g of patients with respiratory failure due tochronic obstructive pulmonary disease. A randomised controlled trial.Ann Intern Med 1998;128:721–8.14 Withhold<strong>in</strong>g and withdraw<strong>in</strong>g life prolong<strong>in</strong>g treatment. Guidance fordecision-mak<strong>in</strong>g. London: BMJ Publish<strong>in</strong>g, 2001.15 Breen D, Churches T, Hawker F, et al. Acute respiratory failuresecondary to chronic obstructive pulmonary disease treated <strong>in</strong> the<strong>in</strong>tensive care unit: a long term follow up study. Thorax 2002;57:29–33.16 Brochard L, Moncebo J, Wysocki M, et al. Non <strong>in</strong>vasive ventilation foracute exacerbations of chronic obstructive pulmonary disease. N Engl JMed 1995;333:817–22.17 Plant P, Owen J, Elliott M. A multi centre randomised control trial of theearly use of non <strong>in</strong>vasive ventilation for acute exacerbations of chronicobstructive pulmonary disease. Lancet 2000;355:1931–5.18 Confalonieri M, Perigi P, Scartabellati A, et al. Non <strong>in</strong>vasivemechanical ventilation improves the immediate and long term outcome ofCOPD patients with acute respiratory failure. Eur Respir J1996;9:422–30.19 Jones SE, Packham S, Hebden M, et al. Domicilary nocturnal <strong>in</strong>termittentpositive pressure ventilation <strong>in</strong> patients with respiratory failure due tosevere COPD: long term follow up and effect on survival. Thorax1998;53:495–8.20 Aldrich TK, Prezant DJ. Indications for mechanical ventilation. In: Tob<strong>in</strong>MJ, ed. Pr<strong>in</strong>ciples and practice of mechanical ventilation. New York:McGraw Hill, 1994: 155–89.21 Dales RE, O’Connor A, Hebert P, et al. Intubation and mechanicalventilation for COPD: development of an <strong>in</strong>strument to elicit patientpreferences. Chest 1999;116:792–800.22 British Thoracic Society Standards of <strong>Care</strong> Subcommittee onPulmonary Rehabilitation. Pulmonary rehabilitation. Thorax2001;56:827–35.23 Morgan D, Evans S, Steele K, et al. Prognosis <strong>in</strong> COPD. Ignorance isbliss? ERJ 2002;20(Suppl 38):169424 Moretti C, Cilione, C, Tampieri A, et al. Incidence and causes ofnon-<strong>in</strong>vasive mechanical ventilation failure after <strong>in</strong>itial success. Thorax2000;55:819–25.25 N<strong>in</strong>ane V, Yernault JC, De Troyer A. Intr<strong>in</strong>sic PEEP <strong>in</strong> patients withchronic obstructive pulmonary disease: role of expiratory muscles. AmRev Respir Dis 1993;148:1037–42.26 Rochester DF. <strong>Respiratory</strong> muscle weakness, pattern of breath<strong>in</strong>g andCO 2 retention <strong>in</strong> chronic obstructive pulmonary disease. Am Rev RespirDis 1991;143:901–3.27 Decramer M, Aubier M. The respiratory muscles: cellular and molecularphysiology. Eur Respir J 1997;10:1943–5.28 Plant PK, Owen JL, Elliott MW. Non-<strong>in</strong>vasive ventilation <strong>in</strong> acuteexacerbations of chronic obstructive pulmonary disease: long termsurvival and predictors of <strong>in</strong>-hospital outcome. Thorax 2001;56:708–12.29 British Thoracic Society Standards of <strong>Care</strong> Subcommittee.Non-<strong>in</strong>vasive ventilation <strong>in</strong> acute respiratory failure. Thorax2002;57:192–211.30 Brochard L. Non <strong>in</strong>vasive ventilation for acute exacerbations of COPD:a new standard of care. Thorax 2000;55:817–8.31 Yentis SM. Suxamethonium and hyperkalaemia. Anaesth Intensive <strong>Care</strong>1990;18:92–101.32 Brimacombe JR, Berry AM. Cricoid pressure: a review article. Can JAnaesth 1997;44:414–25.33 Feihl F, Perrett C. Permissive hypercapnoea. How permissive should webe? Am J Respir Crit <strong>Care</strong> Med 1994;150:1722–37.


<strong>Critical</strong> care management of respiratory failure result<strong>in</strong>g from COPD 8534 Davies L, Angus RM, Calverley PMA. Oral steroids <strong>in</strong> patients admittedto hospital with exacerbations of chronic obstructive pulmonary disease:a prospective randomised controlled trial. Lancet 1999;354:456–60.35 Rabbat A, Laaban JP, Boussairi A, et al. Hyperlactatemia dur<strong>in</strong>g acutesevere asthma. Intensive <strong>Care</strong> Med 1998;24:304–12.36 Ranieri VM, Suter PM, Tortorella C, et al. Effect of mechanicalventilation on <strong>in</strong>flammatory mediators <strong>in</strong> patients with acute respiratorydistress syndrome. A randomised controlled trial. JAMA1999;282:54–61.37 Adult <strong>Respiratory</strong> Distress Syndrome Network. Ventilation with lowertidal volumes compared with traditional lung <strong>in</strong>jury <strong>in</strong> ARDS. N Engl JMed 2000;342:1301–8.38 Rossi A, Gottfried SB, Zocchi L, et al. Measurement of static complianceof the total respiratory system <strong>in</strong> patients with acute respiratory failuredur<strong>in</strong>g mechanical ventilation: the effect of <strong>in</strong>tr<strong>in</strong>sic positiveend-expiratory pressure. Am Rev Respir Dis 1985;131:672–7.39 Zakynth<strong>in</strong>os SG, Vassilakopoulos T, Zakynth<strong>in</strong>os E, et al. Measur<strong>in</strong>gdynamic PEEP. Am J Respir Crit <strong>Care</strong> Med 2000;162:1633–40.40 Mar<strong>in</strong>i JJ, Smith TC, Lamb VJ. External work output and force generationdur<strong>in</strong>g synchronised <strong>in</strong>termittent mechanical ventilation. Effect of mach<strong>in</strong>eassistance on breath<strong>in</strong>g effort. Am Rev Respir Dis 1988;138:1169–79.41 Rossi RA, Append<strong>in</strong>i L. Wasted effort and dyssynchrony: is thepatient-ventilator battle back? Intensive <strong>Care</strong> Med 1995;21:867–70.42 Hormann CH, Baum M, Putensen CH, et al. Bi-phasic positive airwaypressure (BIPAP) a new mode of ventilatory support. Eur J Anaesthesiol1993;11:37–42.43 Yang KL, Tob<strong>in</strong> MJ. A prospective study of <strong>in</strong>dices predict<strong>in</strong>g theoutcome of trials of wean<strong>in</strong>g from mechanical ventilation. N Engl J Med1991;324:1445–50.44 Jubran A, Tob<strong>in</strong> MJ. Pathophysiologic basis of acute respiratory distress<strong>in</strong> patients who fail a trial of wean<strong>in</strong>g from mechanical ventilation. Am JRespir Crit <strong>Care</strong> Med 1997;155:906–15.45 Kollef MH, Levy NT, Ahrens TS, et al. The use of cont<strong>in</strong>uous IV sedationis associated with prolongation of mechanical ventilation. Chest1998;114:541–8.46 Girault C, Daudenthoni, Chevron V, et al. Non <strong>in</strong>vasive ventilation as asystematic extubation and wean<strong>in</strong>g technique <strong>in</strong> acute or chronicrespiratory failure. Am J Respir Crit <strong>Care</strong> Med 1999;160:186–92.47 Hanson P, Dive A, Brucher JM, et al. Acute corticosteroid myopathy <strong>in</strong><strong>in</strong>tensive care patients. Muscle Nerve 1997;20:1371–80.48 Drakulovic MB, Torres A, Bauer TT, et al. Sup<strong>in</strong>e body position as a riskfactor for nosocomial pneumonia <strong>in</strong> mechanically ventilated patients, arandomised trial. Lancet 1999: 354:1851–8.49 Messori A, Trippoli S, Vaiani M, et al. Breath<strong>in</strong>g and pneumonia <strong>in</strong><strong>in</strong>tensive care patients given r<strong>in</strong>itad<strong>in</strong>e and sucralfate for prevention ofstress ulcer: meta-analysis of randomised trials. BMJ 2000;321:1103–6.50 D’Amico R, Pifferi S, Leuretti C, et al. Effectiveness of antibioticprophylaxis <strong>in</strong> critically ill adult patients: systematic review of randomisedcontrolled trials. BMJ 1998;316:1275–85.51 Ely EW, Baker A, Dungan D, et al. Effect of the duration of mechanicalventilation of identify<strong>in</strong>g patients capable of breath<strong>in</strong>g spontaneously. NEngl J Med 1996;335:1864–9.


13 Acute severe asthmaP Phipps, C S Garrard.............................................................................................................................Most asthma exacerbations are managed <strong>in</strong>the community or emergency departmentwhile the more severe cases that fail torespond to bronchodilator and anti-<strong>in</strong>flammatorytherapy require admission to high dependency(HDU) or <strong>in</strong>tensive care units (ICU).Worldwide asthma prevalence is <strong>in</strong>creas<strong>in</strong>g,and with that the total number of admissions tohospital and <strong>in</strong>tensive care. Although the timebetween the onset of symptoms and the requirementfor ventilation is becom<strong>in</strong>g shorter, the outcomeis improv<strong>in</strong>g with fewer deaths and lowercomplication rates. 1MORTALITYA history of mechanical ventilation or ICUadmission is a well documented <strong>in</strong>dicator of subsequentnear fatal asthma. 23 Women and smokersare also over-represented <strong>in</strong> both life threaten<strong>in</strong>gattacks and asthma deaths. 3–5It is believed thatpatients who have had a life threaten<strong>in</strong>g attackand those who die are from a similar demographicgroup. In a large study of patients admittedwith a near fatal episode, two thirds of subsequentsevere attacks or deaths had occurredwith<strong>in</strong> a year. 25Interest<strong>in</strong>gly, the associationbetween asthma deaths and β agonist use is stilldebated and there has been concern that the useof long act<strong>in</strong>g β agonists may <strong>in</strong>crease asthmamortality. 6 This has not been confirmed <strong>in</strong> studiesmonitor<strong>in</strong>g their use. 78 In contrast, there is a consensusthat underuse of anti-<strong>in</strong>flammatory treatment<strong>in</strong> the period lead<strong>in</strong>g up to the acute severeattack worsens prognosis. 9Unfortunately, a proportion of asthmatic patientsdie despite reach<strong>in</strong>g hospital alive. Suchdeaths can usually be attributed either to<strong>in</strong>adequate observation or treatment. Sadly, anumber of patients suffer unobserved respiratoryfailure that progresses to cardiac arrest andanoxic bra<strong>in</strong> damage. 10 A small number ofpatients are resistant to the most aggressivetreatments and <strong>in</strong>terventions.Necroscopic studies of patients dy<strong>in</strong>g of acutesevere asthma have found extensive mucus plugg<strong>in</strong>gof bronchi that has been termed “endobronchialmucus suffocation” (fig 13.1). 11 Microscopicexam<strong>in</strong>ation reveals extensive <strong>in</strong>flammatorychanges that <strong>in</strong>volve all airway wall components12 13and the pulmonary arterioles. The degree ofbronchial occlusion is much greater than <strong>in</strong> controlasthmatic subjects, with mucus, desquamatedepithelium, <strong>in</strong>flammatory cells and plasmaexudate all contribut<strong>in</strong>g. 11 Sudden asphyxicasthma may be a dist<strong>in</strong>ct pathological subtype <strong>in</strong>which <strong>in</strong>tense bronchoconstriction causes respiratoryfailure, often over the course of 1–2hours. 14Recovery appears to be rapid, whichsuggests that bronchoconstriction may be thepredom<strong>in</strong>ant pathophysiological factor. 15INTENSIVE THERAPY AND MONITORINGPatients who fail to improve with optimal medicaltreatment <strong>in</strong> the emergency department shouldbe considered for HDU or ICU admission to facilitatecont<strong>in</strong>uous monitor<strong>in</strong>g of physiological parameterssuch as pulse oximetry, ECG, andarterial and central venous pressure. Equipmentand experienced staff are also available for urgentprocedures such as endotracheal <strong>in</strong>tubation orthe <strong>in</strong>sertion of thoracostomy tubes.Cl<strong>in</strong>ical, physiological and laboratoryassessmentThe immediate assessment of patients withasthma should <strong>in</strong>clude the degree of respiratorydistress (ability to speak, respiratory rate, use ofaccessory muscles, air entry), degree of hypoxia(cyanosis, pulse oximetry, level of consciousness),and cardiovascular stability (arrhythmias, bloodpressure). Accessory muscle use, wheeze, paradox,and tachypnoea may dim<strong>in</strong>ish as the patienttires. 16Forced expiratory volume <strong>in</strong> 1 second (FEV 1)and peak expiratory flow rate are the most usedand convenient measures of airflow obstructionthat support cl<strong>in</strong>ical f<strong>in</strong>d<strong>in</strong>gs and quantify theresponse to treatment. 17 Occasionally patients aretoo distressed to perform forced expiratorymanoeuvres or there is a risk of precipitat<strong>in</strong>g furtherbronchoconstriction. 18 Expiratory airflowlimitation results <strong>in</strong> a dynamic <strong>in</strong>crease <strong>in</strong> endexpiratory lung volume which <strong>in</strong>terferes with<strong>in</strong>spiratory muscle function, both of the diaphragmand the chest wall. The positive alveolarpressure at end expiration (PEEP) due to residualelastic recoil has been termed <strong>in</strong>tr<strong>in</strong>sic (PEEPi)and its presence is suggested by residual expiratoryflow at the onset of <strong>in</strong>spiration. In spontaneouslybreath<strong>in</strong>g patients the magnitude of PEEPican be estimated by the change <strong>in</strong> <strong>in</strong>trapleuralpressure (usually measured by an oesophagealpressure probe) between the onset of <strong>in</strong>spiratoryeffort and the onset of <strong>in</strong>spiratory flow (fig 13.2).The work of breath<strong>in</strong>g is <strong>in</strong>creased <strong>in</strong> the presenceof PEEPi because the residual alveolar pressuremust be overcome by muscle effort before<strong>in</strong>spiratory flow commences. Many patients alsouse expiratory muscles to aid expiration, whichmay paradoxically worsen dynamic airway collapseand PEEPi. Inspiratory muscle activity mayalso persist dur<strong>in</strong>g expiration, 19–21 which contributesto <strong>in</strong>creased expiratory work of breath<strong>in</strong>g. Inmechanically ventilated paralysed patients themagnitude of PEEPi is estimated by perform<strong>in</strong>gan end expiratory breath hold and measur<strong>in</strong>g theairway pressure with reference to that of theatmosphere. In the presence of PEEPi a shortexpiratory time may lead to “breath stack<strong>in</strong>g” andprogressive hyper<strong>in</strong>flation as the next breath is<strong>in</strong>itiated before the previous tidal volume hasbeen completely exhaled. The pathological effectsof PEEPi <strong>in</strong>clude hypotension due to reducedvenous return and an <strong>in</strong>creased risk ofpneumothorax. 22


Acute severe asthma 87AExpiration InspirationResidual expiratory flow atonset of <strong>in</strong>spiratory effort+Flow0_BOnset of <strong>in</strong>spiratory effortOnset of <strong>in</strong>spiratory flowFigure 13.1 Mucus cast of bronchial tree coughed up by anasthmatic patient dur<strong>in</strong>g an exacerbation. Reproduced withpermission of E Klatt, Utah.Poes+0PEEPiPulse oximetry is an <strong>in</strong>valuable adjunct to monitor<strong>in</strong>gs<strong>in</strong>ce the avoidance or abolition of hypoxia is a prime goal oftreatment. Regular arterial blood gas measurements providea measure of gas exchange and facilitate the monitor<strong>in</strong>g ofserum potassium levels. The arterial carbon dioxide tension(PaCO 2) and acid-base status help to identify the presence ofpre-exist<strong>in</strong>g respiratory or metabolic acidosis, and the trend<strong>in</strong> PaCO 2is helpful when assess<strong>in</strong>g the response to treatment.The degree of hypokalaemia and lactic acidosis may alsoguide treatment. 23 A chest radiograph is <strong>in</strong>dicated to identifypneumothorax, areas of segmental or lobar collapse, or <strong>in</strong>filtratesthat may suggest pneumonia. However, the yield islow. 24 25 An ECG may detect myocardial ischaemia or identifyarrhythmias, especially <strong>in</strong> older patients. 26 Right axis deviationand right heart stra<strong>in</strong> are common f<strong>in</strong>d<strong>in</strong>gs. Potassium,magnesium, calcium and phosphate deficiencies should becorrected to reduce the risk of arrhythmia and respiratorymuscle weakness.There are other causes of wheeze and respiratory distressthat must be considered <strong>in</strong> the differential diagnosis. These<strong>in</strong>clude left ventricular failure, upper airway obstruction,<strong>in</strong>haled foreign body, and aspiration of stomach contents.TreatmentIntensive care treatment of the poorly responsive asthmaticpatient should <strong>in</strong>clude high concentrations of <strong>in</strong>spiredoxygen, cont<strong>in</strong>uous nebulisation of β agonists, <strong>in</strong>travenouscorticosteroids, and respiratory support. 27–29 Cl<strong>in</strong>icians must beaware of the need to optimise oxygenation and avoiddehydration and hypokalaemia. Unrestricted high concentrationsof oxygen (60–100%) must be adm<strong>in</strong>istered to abolish27 30hypoxaemia, unlike the patient with chronic obstructivelung disease where controlled limited oxygen is <strong>in</strong>dicated.Hypokalaemia is common and may be exaggerated by fluidresuscitation and the adm<strong>in</strong>istration of β agonist bronchodilators.Repeated <strong>in</strong>fusions of potassium chloride may berequired with careful monitor<strong>in</strong>g of serum levels and cont<strong>in</strong>uousECG monitor<strong>in</strong>g.Specific asthma drug treatmentOn admission to the ICU there should be a rapid review ofearlier asthma treatment to identify elements that can be<strong>in</strong>tensified or deficiencies remedied. Drugs contra<strong>in</strong>dicated <strong>in</strong>asthma <strong>in</strong>clude β blockers, aspir<strong>in</strong>, non-steroidal anti<strong>in</strong>flammatorydrugs, and adenos<strong>in</strong>e.CorticosteroidsEvidence cont<strong>in</strong>ues to accumulate that early treatment withadequate doses of corticosteroid improves outcome <strong>in</strong> severeacute asthma. There does not appear to be any benefit fromhigh doses of hydrocortisone exceed<strong>in</strong>g 400 mg/day, and noCLung volume_+0_TimeIncreas<strong>in</strong>gFRCFigure 13.2 (A) Schematic diagram of an asthmatic patientexhibit<strong>in</strong>g significant residual flow at end expiration. (B) Theoesophageal pressure (Poes), an estimate of <strong>in</strong>trapleural pressure,shows the degree of pressure change required to overcome <strong>in</strong>tr<strong>in</strong>sicpressure (PEEPi) and <strong>in</strong>itiate <strong>in</strong>spiratory flow. (C) A progressive<strong>in</strong>crease <strong>in</strong> lung volume (breath stack<strong>in</strong>g) occurs if expiratory time is<strong>in</strong>sufficient to allow complete exhalation of the tidal volume.particular advantage of the <strong>in</strong>travenous over the oral routeprovided there is reliable gastro<strong>in</strong>test<strong>in</strong>al absorption. 31 Inhaledcorticosteroids have not been fully evaluated <strong>in</strong> this sett<strong>in</strong>g.β agonistsSalbutamol (albuterol <strong>in</strong> North America) is the mostcommonly prescribed β agonist for the treatment of acuteasthma. 29It appears to be more effective and <strong>in</strong>duces lesshypokalaemia when delivered by the <strong>in</strong>haled route, althoughthere is a theoretical rationale for adm<strong>in</strong>ister<strong>in</strong>g salbutamol<strong>in</strong>travenously to bypass obstructed airways. 32Concerns overblood levels and potential cardiotoxicity may <strong>in</strong>hibit moreaggressive use of nebulised salbutamol. 33 However, cont<strong>in</strong>uousadm<strong>in</strong>istration of nebulised salbutamol <strong>in</strong> doses approach<strong>in</strong>g20 mg/h can achieve bronchodilation without toxicity; <strong>in</strong>deed,the patient’s heart rate may fall with alleviation of airwayobstruction. 34Interpretation of the literature on cont<strong>in</strong>uous nebulisedsalbutamol is hampered by differences <strong>in</strong> the def<strong>in</strong>itions of“cont<strong>in</strong>uous” (length of time) and the delivered doses used <strong>in</strong><strong>in</strong>dividual studies. One study compared 27.5 mg salbutamolby either cont<strong>in</strong>uous or <strong>in</strong>termittent nebulisation over 6hours 35 and, not surpris<strong>in</strong>gly, showed little difference between<strong>in</strong>termittent and cont<strong>in</strong>uous regimens. Another study suggestedbenefit from prolonged cont<strong>in</strong>uous aerosol use only <strong>in</strong>severe asthma. 36Doses of 0.3 mg/kg/h nebulised salbutamolhave been safely used <strong>in</strong> children without significant


88 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>toxicity. 37 Higher <strong>in</strong>haled doses may be required <strong>in</strong> mechanicallyventilated patients due to aerosol losses <strong>in</strong> the ventilatorcircuit.Salbutamol delivered by metered dose <strong>in</strong>haler (MDI) with aspacer device is at least as effective as nebulised drug <strong>in</strong> themanagement of acute asthma <strong>in</strong> the emergencydepartment. 38 In patients with hypoxia and respiratorydistress, however, nebulised drugs may be easier to adm<strong>in</strong>ister.Even <strong>in</strong>tranasal and <strong>in</strong>tratracheal <strong>in</strong>stillation of β agonistsmay be effective <strong>in</strong> an emergency situation. 39 40 Salbutamol iscurrently a racemic mixture of R and L forms. TheS-enantiomer does not have β agonist effects and competes forthe b<strong>in</strong>d<strong>in</strong>g sites of the R form, levosalbutamol. 41Formulationsof levosalbutamol have fewer side effects and greaterefficacy than the racemic mixture, suggest<strong>in</strong>g that it may bepreferable <strong>in</strong> acute severe asthma. 42 Terbutal<strong>in</strong>e is an alternativeβ agonist that has been less widely studied but is effectiveby the <strong>in</strong>haled, <strong>in</strong>travenous, and subcutaneous routes. Longact<strong>in</strong>g β agonists such as salmeterol and eformoterol are notrecommended because of their slow onset of action.Both nebulised and <strong>in</strong>travenous adrenal<strong>in</strong>e (ep<strong>in</strong>ephr<strong>in</strong>e)are effective <strong>in</strong> the treatment of acute asthma. The putativebenefits of the α-adrenergic component of adrenal<strong>in</strong>e <strong>in</strong>cludereduced microvascular permeability and airway wall oedema,and less impairment of ventilation/perfusion match<strong>in</strong>g thanwith more selective β agonists. 43However, there does notappear to be any cl<strong>in</strong>ical benefit over β agonists such assalbutamol. 44Some of the metabolic and cardiovascular complications ofacute severe asthma may be exacerbated by high dose β agonisttherapy. Lactic acidosis as a result of parenteral β agonistuse, anaerobic metabolism due to high work of breath<strong>in</strong>g, tissuehypoxia, <strong>in</strong>tracellular alkalosis, and reduced lactate clearancedue to liver congestion all contribute to the complexmetabolic disturbances of acute severe asthma. Haemodynamicallysignificant arrhythmias are relatively <strong>in</strong>frequent,even with the comb<strong>in</strong>ation of methylxanth<strong>in</strong>es and β receptoragonists 45 ; however, β agonist <strong>in</strong>duced hypokalaemia heightensthe risk.IpratropiumIpratropium bromide has a mild additional bronchodilat<strong>in</strong>geffect when added to β agonists that may only be significant <strong>in</strong>severe asthma. 46 The safety profile and the fact that <strong>in</strong>dividualpatients may obta<strong>in</strong> benefit have resulted <strong>in</strong> aerosolised ipratropium(500 µg 6 hourly) be<strong>in</strong>g recommended for thetreatment of acute severe asthma. 47Am<strong>in</strong>ophyll<strong>in</strong>eThe addition of am<strong>in</strong>ophyll<strong>in</strong>e does not add to the bronchodilat<strong>in</strong>geffect of optimal doses of β agonists. 48 Other reportedbenefits of am<strong>in</strong>ophyll<strong>in</strong>e such as improv<strong>in</strong>g diaphragmaticendurance, stimulat<strong>in</strong>g ventilatory drive, and anti<strong>in</strong>flammatoryeffect do not seem to improve outcome <strong>in</strong> acutesevere asthma. 49–53Currently, am<strong>in</strong>ophyll<strong>in</strong>e is not recommendedas a first l<strong>in</strong>e drug <strong>in</strong> acute asthma management andits <strong>in</strong>clusion as a second l<strong>in</strong>e agent is still debated. 46 However,when other agents fail to achieve bronchodilation, am<strong>in</strong>ophyll<strong>in</strong>ecan be used provid<strong>in</strong>g dos<strong>in</strong>g regimens are adhered to.Typically, a load<strong>in</strong>g dose of 5 mg/kg by slow <strong>in</strong>travenous <strong>in</strong>fusionover 20 m<strong>in</strong>utes is followed by an <strong>in</strong>fusion of 500 µg/kg/h. If prolonged adm<strong>in</strong>istration is required, daily monitor<strong>in</strong>g ofblood theophyll<strong>in</strong>e levels is essential. The therapeutic range is55–110 µmol/l (10–20 mg/l).ASSISTED VENTILATIONIf there is <strong>in</strong>adequate response to drug treatment or if thepatient is <strong>in</strong> extremis at presentation, mechanical ventilationmay be required.Box 13.1 Contra<strong>in</strong>dications to a trial of mask CPAPor NIV <strong>in</strong> acute severe asthma• Need for immediate endotracheal <strong>in</strong>tubation• Poor patient cooperation• Inability of the ventilator to supply high FiO 2• Hypercapnia (CPAP less likely to benefit than NIV)• Excess respiratory secretions• Lack of experienced staff and/or a high dependency areaMask cont<strong>in</strong>uous positive airway pressure (CPAP) andnon-<strong>in</strong>vasive ventilation (NIV)In spontaneously breath<strong>in</strong>g patients the application of lowlevels of mask CPAP (3–8 cm H 2O) may improve respiratoryrate, dyspnoea, and work of breath<strong>in</strong>g <strong>in</strong> asthma, particularlyif there is evidence of smok<strong>in</strong>g related lung disease. 20 54 55 Thereis a danger that CPAP may worsen lung hyper<strong>in</strong>flation. Ifpatients are <strong>in</strong>tolerant of the mask or do not derive benefit,CPAP should be withdrawn. In hypercapnic patients CPAPalone may not improve ventilation.Few studies have looked specifically at NIV <strong>in</strong> asthma. Lowlevels of CPAP and pressure support of 10–19 cm H 2O <strong>in</strong> acutesevere asthma improved gas exchange and prevented endotracheal<strong>in</strong>tubation <strong>in</strong> all but two of 17 hypercapnic patients. 56However, the rate of <strong>in</strong>tubation <strong>in</strong> patients with acute asthma,even <strong>in</strong> the presence of hypercapnia, is low at 3–8%. 28 57 It isreasonable to give asthmatic patients a trial of NIV over 1–2hours <strong>in</strong> an HDU or ICU if there are no contra<strong>in</strong>dications (box13.1). 56 Decid<strong>in</strong>g when to <strong>in</strong>itiate NIV, when a trial of NIV hasfailed, and optimis<strong>in</strong>g NIV <strong>in</strong> this sett<strong>in</strong>g require considerableexpertise. In our experience, high flow ventilators specificallydesigned for NIV (such as the BiPAP Vision; Respironics, Pittsburg,USA) that allow significant mask and mouth leaks arebetter tolerated than many conventional ICU ventilators.Endotracheal <strong>in</strong>tubationCardiopulmonary arrest and deteriorat<strong>in</strong>g consciousness areabsolute <strong>in</strong>dications for <strong>in</strong>tubation and assisted ventilation.Hypercapnia, acidosis, and cl<strong>in</strong>ical signs of severe disease atpresentation may not require immediate <strong>in</strong>tubation before an57 58aggressive trial of conventional bronchodilator therapy.Conversely, progressive deterioration with <strong>in</strong>creas<strong>in</strong>g distressor physical exhaustion may warrant <strong>in</strong>tubation and mechanicalventilation without the presence of hypercapnia.Once it has been decided that mechanical ventilation isrequired, the necessary medications, suitable monitor<strong>in</strong>gequipment, and expert help should be sought. An understand<strong>in</strong>gof the pathophysiology and anticipation of difficulties canm<strong>in</strong>imise the complications associated with endotracheal<strong>in</strong>tubation and ventilation. The best technique for <strong>in</strong>tubationis generally that most familiar to the cl<strong>in</strong>ician perform<strong>in</strong>g theprocedure.The process of <strong>in</strong>tubation beg<strong>in</strong>s with explanation andreassurance for the patient, followed by pre-oxygenation. Theasthmatic patient is often dehydrated and the comb<strong>in</strong>ation ofPEEPi, the loss of endogenous catecholam<strong>in</strong>es, and thevasodilat<strong>in</strong>g properties of the anaesthetic agents can causecatastrophic hypotension. 59 60 Volume resuscitation before<strong>in</strong>duction of anaesthesia can limit the degree of hypotensionbut vasoconstrictors such as ephedr<strong>in</strong>e or metaram<strong>in</strong>ol shouldbe at hand.Intubation is best performed by direct laryngoscopy after<strong>in</strong>duction of general anaesthesia. Endoscopic methods have16 58been advocated with either oral or nasal <strong>in</strong>tubation, butlaryngeal spasm and further bronchoconstriction may occur.Satisfactory local anaesthesia of the oropharynx, nasopharynx,and larynx is therefore essential. Longer term sedation isrequired once the airway has been secured. Some recommendationsfor successful and safe endotracheal <strong>in</strong>tubation aresummarised <strong>in</strong> box 13.2.


Acute severe asthma 89Box 13.2 Summary of recommendations for theprocess of <strong>in</strong>tubation• Performed/supervised by experienced anaesthetists or<strong>in</strong>tensivists• Skilled assistants <strong>in</strong> an appropriate environment• Good preparation and understand<strong>in</strong>g of the pathophysiology• Correct electrolyte disturbances and rehydrate• Obta<strong>in</strong> reliable large bore venous access• Cont<strong>in</strong>uous ECG and pulse oximetry• Cont<strong>in</strong>uous arterial monitor<strong>in</strong>g not essential, but helpful• Pre-oxygenate• Use familiar method of <strong>in</strong>tubation• Use familiar sedatives and muscle relaxants• Prepare for the rapid correction of hypotension, arrhythmiasand barotrauma.• Ventilator set up and ready to monitor airway pressuresearly• Get aerosol delivery system for the ventilator connected orcommence parenteral bronchodilator therapy• Plan ongo<strong>in</strong>g sedation/paralysis before <strong>in</strong>tubationDrug therapy for <strong>in</strong>tubation and mechanical ventilationAnaesthetic agents and sedativesEtomidate and thiopentone are short act<strong>in</strong>g imidazole andbarbiturate drugs, respectively, that are commonly used for<strong>in</strong>tubation although rarely bronchospasm and anaphylactoidreactions have been reported. Longer term sedation may beobta<strong>in</strong>ed by <strong>in</strong>fusion of midazolam (2–10 mg/h); metabolitesmay accumulate <strong>in</strong> renal and hepatic impairment. Propofol isa useful drug for <strong>in</strong>tubation and <strong>in</strong>termediate term sedation,ma<strong>in</strong>ly because of its rapid onset and offset of action. It is easilytitratable for <strong>in</strong>tubation, provid<strong>in</strong>g deep sedation rapidly,although it has no analgesic properties. However, vasodilatationand hypotension occur, especially <strong>in</strong> dehydrated patients.Relatively little literature regard<strong>in</strong>g its specific use <strong>in</strong> asthma isavailable. The doses of all the above agents need to be adjustedfor patient size and pre-exist<strong>in</strong>g level of consciousness.Ketam<strong>in</strong>e is a general anaesthetic agent that has been usedbefore, dur<strong>in</strong>g, and after <strong>in</strong>tubation <strong>in</strong> patients with acutesevere asthma. 61–63 It has sympathomimetic and bronchodilat<strong>in</strong>gproperties. The usual dose for <strong>in</strong>tubation is 1–2 mg/kggiven <strong>in</strong>travenously over 2–4 m<strong>in</strong>utes. It may <strong>in</strong>crease bloodpressure and heart rate, lower seizure threshold, alter mood,and cause delirium. Inhalational anaesthetics used for gas<strong>in</strong>duction have the advantage of bronchodilation and maymake muscle relaxation unnecessary. However, specialisedanaesthetic equipment is required for this approach.Opioids are a useful addition to sedatives and provide analgesiadur<strong>in</strong>g <strong>in</strong>tubation and mechanical ventilation. Morph<strong>in</strong>e<strong>in</strong> large boluses causes histam<strong>in</strong>e release, which may worsenbronchoconstriction and hypotension. Some <strong>in</strong>travenouspreparations also conta<strong>in</strong> metabisulphite, to which someasthmatics are sensitive. Fentanyl is a better choice of opioidfor <strong>in</strong>tubation as it <strong>in</strong>hibits airway reflexes and is short act<strong>in</strong>g.It causes less histam<strong>in</strong>e release than morph<strong>in</strong>e but largeboluses may cause bronchospasm and chest wall rigidity.Neuromuscular block<strong>in</strong>g drugsRapid sequence <strong>in</strong>duction with cricoid pressure should beused to prevent aspiration of gastric contents. Suxamethonium,a depolaris<strong>in</strong>g muscle relaxant, is widely used. It has arapid onset and short duration of action but may cause hyperkalaemiaand <strong>in</strong>creased <strong>in</strong>tracranial pressure. Rocuronium, anon-depolaris<strong>in</strong>g muscle relaxant with an acceptably rapidonset, offers an alternative. Allergic sensitivity may occur toany neuromuscular block<strong>in</strong>g agent and most may also causehistam<strong>in</strong>e release and the potential for bronchospasm,particularly <strong>in</strong> bolus doses. Atracurium boluses should beBox 13.3 Initial ventilator sett<strong>in</strong>gs <strong>in</strong> paralysedpatients (adapted from F<strong>in</strong>fer and Garrard 109 )•FiO 2= 1.0 (<strong>in</strong>itially)• Long expiratory time (I:E ratio >1:2)• Low tidal volume 5–7 ml/kg• Low ventilator rate (8–10 breaths/m<strong>in</strong>)• Set <strong>in</strong>spiratory pressure 30–35 cm H 2O on pressure controlventilation or limit peak <strong>in</strong>spiratory pressure to


90 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Humidification of <strong>in</strong>spired gas is particularly important <strong>in</strong>asthmatic patients to prevent thicken<strong>in</strong>g of secretions anddry<strong>in</strong>g of airway mucosa, a stimulus for bronchospasm <strong>in</strong>itself. 69Ventilator alarmsThese <strong>in</strong>clude peak pressure and low tidal volume/low m<strong>in</strong>uteventilation alarms. If exceptionally high airway pressuresoccur or there is a sudden fall <strong>in</strong> VT, blockage of the endotrachealtube, pneumothorax, or lobar collapse should beexcluded. Plateau rather than peak airway pressure may providethe best measure of alveolar pressure and provide the bestpredictor of barotrauma, together with measures of hyper<strong>in</strong>flationsuch as PEEPi. 70Extr<strong>in</strong>sic PEEPLow level CPAP may be beneficial <strong>in</strong> spontaneously breath<strong>in</strong>g,mechanically ventilated patients, especially if expiratory muscleactivity is contribut<strong>in</strong>g to dynamic airways collapse. However,<strong>in</strong> mechanically ventilated paralysed patients extr<strong>in</strong>sicPEEP was of no benefit at low levels and was detrimental athigh levels because the fall <strong>in</strong> gas trapp<strong>in</strong>g was outweighed bythe rise <strong>in</strong> functional residual capacity (FRC). 22However, <strong>in</strong>this study large VT were used (up to 18 ml/kg); furthermorePEEPi and arterial blood gases were not measured. Changes <strong>in</strong>FRC and gas trapp<strong>in</strong>g may guide the level of PEEP. Appliedextr<strong>in</strong>sic PEEP should not exceed PEEPi.Topical drug delivery to the ventilated patientMechanical ventilation, whether <strong>in</strong>vasive or non-<strong>in</strong>vasive, maycompromise the delivery of bronchodilator aerosols. Theamount of nebulised drug reach<strong>in</strong>g the airways depends onthe nebuliser design, driv<strong>in</strong>g gas flow, characteristics of the47 71ventilator tub<strong>in</strong>g, and the size of the endotracheal tube.Drug delivery may vary from 0% to 42% <strong>in</strong> ventilatedpatients. 72The presence of humidification alone may reducedrug deposition by as much as 40%, but may be reversed by theaddition of a spacer device. 73 74 Both ultrasonic and jet nebulisersare effective <strong>in</strong> ventilated patients. 75 Nebulisers may, however,be a source of bacterial contam<strong>in</strong>ation. 76Metered dose <strong>in</strong>halers have been widely used and may provideat least as good drug delivery as nebulisers, depend<strong>in</strong>g onactuator design and the presence of humidification and spacerdevices. 77 78 The recommended characteristics of aerosol deliverysystems used <strong>in</strong> ventilated patients are shown <strong>in</strong> box 13.4.Ideally, each aerosol delivery system should be evaluated foreach type of ventilator circuit used. 73THERAPEUTIC OPTIONS IN THE NON-RESPONDINGPATIENTA proportion of patients improve rapidly follow<strong>in</strong>g the<strong>in</strong>troduction of mechanical ventilation, and wean<strong>in</strong>g shouldoccur <strong>in</strong> l<strong>in</strong>e with this improvement. Unfortunately, for somethere is difficulty <strong>in</strong> achiev<strong>in</strong>g adequate ventilation or there ispersistent hypoxia. Difficulty <strong>in</strong> ventilation may be due torefractory bronchospasm, extreme hyper<strong>in</strong>flation, or mucusplugg<strong>in</strong>g.Manual compressionThis technique was first described anecdotally by Watts <strong>in</strong>1984. 79 Hyper<strong>in</strong>flation is relieved by manual compression ofthe chest wall dur<strong>in</strong>g expiration. 80The technique has beenadvocated and used with success <strong>in</strong> both <strong>in</strong>tubated and non<strong>in</strong>tubatedpatients, although it has not been fully evaluated by81 80a controlled cl<strong>in</strong>ical study <strong>in</strong> humans.MucolyticsThere is often a strik<strong>in</strong>g degree of mucus impaction <strong>in</strong> bothlarge and small airways that contributes to hyper<strong>in</strong>flation,segmental and lobar collapse with shunt<strong>in</strong>g, <strong>in</strong>creased airwayBox 13.4 Recommendations for aerosol delivery tomechanically ventilated patientsMetered dose <strong>in</strong>haler (MDI) system• Spacer or hold<strong>in</strong>g chamber• Location <strong>in</strong> <strong>in</strong>spiratory limb rather than Y piece• No humidification (briefly discont<strong>in</strong>ue)• Actuate dur<strong>in</strong>g lung <strong>in</strong>flation• Large endotracheal tube <strong>in</strong>ternal diameter• Prolonged <strong>in</strong>spiratory timeJet nebuliser system• Mount nebuliser <strong>in</strong> <strong>in</strong>spiratory limb• Delivery may be improved by <strong>in</strong>spiratory trigger<strong>in</strong>g• Increase <strong>in</strong>spiratory time and decrease respiratory rate• Use a spacer• High flow to generate aerosol• High volume fill• Stop humidification• Consider cont<strong>in</strong>uous nebulisationUltrasonic nebulisers• Position <strong>in</strong> <strong>in</strong>spiratory limb prior to a spacer device• Use high power sett<strong>in</strong>g• Use a high volume fill• Maximise <strong>in</strong>spiratory time• Drugs must be stable dur<strong>in</strong>g ultrasonic nebulisationpressure, and barotrauma. Chest physiotherapy and mucolyticshave no proven benefit. Bronchoscopic lavage with locallyapplied acetylcyste<strong>in</strong>e may be used to help clear impactedsecretions <strong>in</strong> selected refractory patients but its rout<strong>in</strong>e use isnot advocated. 82Recently, recomb<strong>in</strong>ant DNase, a mucolyticprescribed for sputum liquefaction <strong>in</strong> cystic fibrosis, has beenused to treat mucus impaction <strong>in</strong> asthma but there are nocl<strong>in</strong>ical trials. 83Inhalational anaesthetic agentsHalothane, isoflurane, and sevoflurane are potent bronchodilators<strong>in</strong> asthmatic patients receiv<strong>in</strong>g mechanical ventilationwho have failed to respond to conventional β adrenergicagents. 84Experimental evidence <strong>in</strong>dicates a direct effect onbronchial smooth muscle mediated via calcium dependentchannels as well as by modulat<strong>in</strong>g vagal, histam<strong>in</strong>e, allergen,85 86and hypoxia <strong>in</strong>duced bronchoconstrictor mechanisms.Furthermore, these agents reduce pulmonary vascular toneresult<strong>in</strong>g <strong>in</strong> lower pulmonary artery pressures <strong>in</strong> acuteasthma. 87Bronchodilator responses are seen <strong>in</strong> the form ofreduced peak airway pressures with<strong>in</strong> m<strong>in</strong>utes, associatedwith improved ventilation distribution (lower PaCO 2) andreduced air trapp<strong>in</strong>g. 88Although bronchodilator effects areseen at sub-anaesthetic concentrations, these agents alsooffer a relatively expensive method of sedation. A few ICUventilators, such as the Seimens Servo 900 series, can be fittedwith a vaporiser allow<strong>in</strong>g anaesthetic gases to be adm<strong>in</strong>istered.Effective exhaled gas scaveng<strong>in</strong>g systems are requiredwhen us<strong>in</strong>g <strong>in</strong>halational anaesthetics <strong>in</strong> the ICU. If this facilityis not available a Cardiff canister can be added to theexpiratory port of the ventilator to remove effluent anaestheticgases. Significant side effects such as hypotension andmyocardial irritability exist, and prolonged adm<strong>in</strong>istration ofsome agents may result <strong>in</strong> bromide or fluoride toxicity. 89Sevoflurane, a halogenated ether, is largely devoid ofcardiorespiratory side effects and may be the preferred agent.Adm<strong>in</strong>istration of sub-anaesthetic concentrations of theseagents via face mask may relieve bronchospasm refractory toconventional treatment. 90One of the difficult aspects of mechanical ventilation of theacute asthmatic patient is the wean<strong>in</strong>g and extubationprocess. The presence of the endotracheal tube with<strong>in</strong> the larynxand trachea <strong>in</strong>duces bronchoconstriction which becomes


Acute severe asthma 91troublesome as the sedation is withdrawn <strong>in</strong> preparation forextubation. 91 92 Use of an <strong>in</strong>halational anaesthetic agent allowsthe endotracheal tube to be removed under anaesthesia withthe confident expectation of rapid recovery once the anaestheticis discont<strong>in</strong>ued.HeliumA mixture of helium and oxygen (heliox) may reduce the workof breath<strong>in</strong>g and improve gas exchange because of its lowdensity that reduces airway resistance and hyper<strong>in</strong>flation.However, the benefits are marg<strong>in</strong>al and the concentration of<strong>in</strong>spired oxygen is consequently decreased. Flow meters andnebuliser generator systems must be adapted for heliox use <strong>in</strong>ventilated patients. 93 The use of heliox to prevent <strong>in</strong>tubationhas not been studied, but dyspnoea scores were improved <strong>in</strong>one study, possibly by reduc<strong>in</strong>g the work of breath<strong>in</strong>g. 94Magnesium sulphateEarly anecdotal reports suggested benefit from <strong>in</strong>travenousmagnesium sulphate, which has been <strong>in</strong>consistently supportedby randomised studies. 95–99A significant benefit wasrecently observed <strong>in</strong> children receiv<strong>in</strong>g <strong>in</strong>travenous magnesiumsulphate (40 mg/kg) dur<strong>in</strong>g acute asthma attacks. 100Overall, the case for magnesium sulphate <strong>in</strong> acute asthmarequires further evaluation <strong>in</strong> both adults and children.Leukotriene <strong>in</strong>hibitorsLeukotrienes are <strong>in</strong>flammatory mediators known to be active<strong>in</strong> the airway <strong>in</strong>flammation of asthma. Leukotriene receptorantagonists (zafirlukast, montelukast) and synthesis blockers(zilueton) currently have a relatively m<strong>in</strong>or role <strong>in</strong> themanagement of poorly controlled and aspir<strong>in</strong> sensitiveasthma. 101 However, recent work has suggested a role for leukotrieneantagonists <strong>in</strong> acute asthma. 102–105Platelet activat<strong>in</strong>g factor (PAF) <strong>in</strong>hibitorsPAF <strong>in</strong>hibitors attenuate the late response <strong>in</strong> asthma but havelimited cl<strong>in</strong>ical efficacy. 106Nitric oxide (NO)NO exerts a weak bronchodilator effect. 107 It dilates pulmonaryarteries and, when <strong>in</strong>haled, may improve ventilation/perfusionmatch<strong>in</strong>g.OUTCOME AND FOLLOW UPICU admission identifies an asthmatic patient as a member ofa poor prognostic group. 3 5 108 Follow up should <strong>in</strong>clude a focuson anti-<strong>in</strong>flammatory therapy and a written managementplan that may <strong>in</strong>clude the emergency use of <strong>in</strong>tramuscularadrenal<strong>in</strong>e. Issues such as access to health care services, compliancewith treatment, avoidance of triggers, socioeconomicand psychosocial factors also need to be addressed.ACKNOWLEDGEMENTThe authors would like to thank Dr Duncan Young for his review of themanuscript and helpful suggestions.REFERENCES1 Kearney SE, Graham DR, Atherton ST. Acute severe asthma treated bymechanical ventilation: a comparison of the chang<strong>in</strong>g characteristics overa 17 yr period. Respir Med 1998;92:716–21.2 Richards GN, Kolbe J, Fenwick J, et al. Demographic characteristics ofpatients with severe life threaten<strong>in</strong>g asthma: comparison with asthmadeaths. Thorax 1993;48:1105–9.3 Turner MO, Noertjojo K, Vedal S, et al. Risk factors for near-fatalasthma. A case-control study <strong>in</strong> hospitalized patients with asthma. Am JRespir Crit <strong>Care</strong> Med 1998;157:1804–9.4 Sandford AJ, Pare PD. The genetics of asthma. The important questions.Am J Respir Crit <strong>Care</strong> Med 2000;161:S202–6.5 Marquette CH, Saulnier F, Leroy O, et al. Long-term prognosis ofnear-fatal asthma. A 6-year follow-up study of 145 asthmatic patientswho underwent mechanical ventilation for a near-fatal attack of asthma.Am Rev Respir Dis 1992;146:76–81.6 Abramson MJ, Bailey MJ, Couper FJ, et al. Are asthma medications andmanagement related to deaths from asthma? Am J Respir Crit <strong>Care</strong> Med2001;163:12–8.7 Castle W, Fuller R, Hall J, et al. Serevent nationwide surveillance study:comparison of salmeterol with salbutamol <strong>in</strong> asthmatic patients whorequire regular bronchodilator treatment. BMJ 1993;306:1034–7.8 Donohue JF. The expand<strong>in</strong>g role of long-act<strong>in</strong>g beta-agonists. Chest2000;118:283–5.9 Kallenbach JM, Frankel AH, Lap<strong>in</strong>sky SE, et al. Determ<strong>in</strong>ants of nearfatality <strong>in</strong> acute severe asthma. Am J Med 1993;95:265–72.10 Shugg AW, Kerr S, Butt WW. Mechanical ventilation of paediatricpatients with asthma: short and long term outcome. J Paediatr ChildHealth 1990;26:343–6.11 Saetta M, Di Stefano A, Ros<strong>in</strong>a C, et al. Quantitative structural analysisof peripheral airways and arteries <strong>in</strong> sudden fatal asthma. Am Rev RespirDis 1991;143:138–43.12 Carroll N, Elliot J, Morton A, et al. The structure of large and smallairways <strong>in</strong> nonfatal and fatal asthma. Am Rev Respir Dis1993;147:405–10.13 Faul JL, Tormey VJ, Leonard C, et al. Lung immunopathology <strong>in</strong> cases ofsudden asthma death. Eur Respir J 1997;10:301–7.14 Reid LM. The presence or absence of bronchial mucus <strong>in</strong> fatal asthma. JAllergy Cl<strong>in</strong> Immunol 1987;80:415–6.15 Wasserfallen JB, Schaller MD, Feihl F, et al. Sudden asphyxic asthma:a dist<strong>in</strong>ct entity? Am Rev Respir Dis 1990;142:108–11.16 Corbridge TC, Hall JB. The assessment and management of adults withstatus asthmaticus. Am J Respir Crit <strong>Care</strong> Med 1995;151:1296–316.17 Rodrigo G, Rodrigo C. Assessment of the patient with acute asthma <strong>in</strong>the emergency department. A factor analytic study. Chest1993;104:1325–8.18 Lim TK. Status asthmaticus <strong>in</strong> a medical <strong>in</strong>tensive care. S<strong>in</strong>gapore Med J1989;30:334–8.19 Hill AR. <strong>Respiratory</strong> muscle function <strong>in</strong> asthma. J Assoc Acad M<strong>in</strong>or Phys1991;2:100–8.20 Lougheed DM, Webb KA, O’Donnell DE. Breathlessness dur<strong>in</strong>g <strong>in</strong>ducedlung hyper<strong>in</strong>flation <strong>in</strong> asthma: the role of the <strong>in</strong>spiratory threshold load.Am J Respir Crit <strong>Care</strong> Med 1995;152:911–20.21 De Troyer A, Pride NB. The chest wall and respiratory muscles. In:Roussos C, ed. The thorax. New York: Dekker, 1995: 1975–2006.22 Tuxen DV. Detrimental effects of positive end-expiratory pressure dur<strong>in</strong>gcontrolled mechanical ventilation of patients with severe airflowobstruction. Am Rev Respir Dis 1989;140:5–9.23 Appel D, Rubenste<strong>in</strong> R, Schrager K, et al. Lactic acidosis <strong>in</strong> severeasthma. Am J Med 1983;75:580–4.24 F<strong>in</strong>dley LJ, Sahn SA. The value of chest roentgenograms <strong>in</strong> acute asthma<strong>in</strong> adults. Chest 1981;80:535–6.25 White CS, Cole RP, Lubetsky HW, et al. Acute asthma. Admission chestradiography <strong>in</strong> hospitalized adult patients. Chest 1991;100:14–6.26 Myrianthefs MM, Nicolaides EP, Pitiris D, et al. False positiveST-segment depression dur<strong>in</strong>g exercise <strong>in</strong> subjects with short PR segmentand angiographically normal coronaries: correlation withexercise-<strong>in</strong>duced ST depression <strong>in</strong> subjects with normal PR and normalcoronaries. J Electrocardiol 1998;31:203–8.27 British Thoracic Society, British Paediatric Association, Research Unit ofthe Royal College of Physicians of London, et al. Guidel<strong>in</strong>es on themanagement of asthma. Thorax 1993;48(Suppl):S1–24.28 Garrard CS, Nolan K. Intensive care management of asthma. In: WebbAR, Shapiro MJ, S<strong>in</strong>ger M, et al, eds. Oxford textbook of <strong>in</strong>tensive care.Oxford: Oxford University Press, 1999:90–5.29 Garrard CS, Benham S. Bronchodilators <strong>in</strong> <strong>in</strong>tensive care. In: Webb AR,Shapiro MJ, S<strong>in</strong>ger M, et al, eds. Oxford textbook of <strong>in</strong>tensive care.Oxford: Oxford University Press, 1999:1241–4.30 Lipworth BJ, Jackson CM, Ziyaie D, et al. An audit of acute asthmaadmissions to a respiratory unit. Health Bull (Ed<strong>in</strong>b) 1992;50:389–98.31 Manser R, Reid D, Abramson M. Corticosteroids for acute severe asthma<strong>in</strong> hospitalised patients. Cochrane Database Syst Rev 2000;2.32 Salmeron S, Brochard L, Mal H, et al. Nebulized versus <strong>in</strong>travenousalbuterol <strong>in</strong> hypercapnic acute asthma. A multicenter, double-bl<strong>in</strong>d,randomized study. Am J Respir Crit <strong>Care</strong> Med 1994;149:1466–70.33 L<strong>in</strong> RY, Smith AJ, Hergenroeder P. High serum albuterol levels andtachycardia <strong>in</strong> adult asthmatics treated with high-dose cont<strong>in</strong>uouslyaerosolized albuterol. Chest 1993;103:221–5.34 L<strong>in</strong> RY, Sauter D, Newman T, et al. Cont<strong>in</strong>uous versus <strong>in</strong>termittentalbuterol nebulization <strong>in</strong> the treatment of acute asthma. Ann Emerg Med1993;22:1847–53.35 Besbes-Ouanes L, Nouira S, Elatrous S, et al. Cont<strong>in</strong>uous versus<strong>in</strong>termittent nebulization of salbutamol <strong>in</strong> acute severe asthma: arandomized, controlled trial. Ann Emerg Med 2000;36:198–203.36 Rudnitsky GS, Eberle<strong>in</strong> RS, Schoffstall JM, et al. Comparison of<strong>in</strong>termittent and cont<strong>in</strong>uously nebulized albuterol for treatment of asthma<strong>in</strong> an urban emergency department. Ann Emerg Med 1993;22:1842–6.37 Papo MC, Frank J, Thompson AE. A prospective, randomized study ofcont<strong>in</strong>uous versus <strong>in</strong>termittent nebulized albuterol for severe statusasthmaticus <strong>in</strong> children. Crit <strong>Care</strong> Med 1993;21:1479–86.38 Idris AH, McDermott MF, Raucci JC, et al. Emergency departmenttreatment of severe asthma. Metered-dose <strong>in</strong>haler plus hold<strong>in</strong>g chamberis equivalent <strong>in</strong> effectiveness to nebulizer. Chest 1993;103:665–72.39 Weksler N, Brill S, Tarnapolski A, et al. Intranasal salbutamol <strong>in</strong>stillation<strong>in</strong> asthma attack. Am J Emerg Med 1999;17:686–8.40 Steedman DJ, Robertson CE. Emergency endotracheal drugadm<strong>in</strong>istration us<strong>in</strong>g aerosol. Resuscitation 1987;15:135–9.41 Boulton DW, Fawcett JP. The pharmacok<strong>in</strong>etics of levosalbutamol: whatare the cl<strong>in</strong>ical implications? Cl<strong>in</strong> Pharmacok<strong>in</strong>et 2001;40:23–40.


92 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>42 Handley DA. S<strong>in</strong>gle-isomer beta-agonists. Pharmacotherapy2001;21:21–7S.43 Baldw<strong>in</strong> DR, Sivardeen Z, Pavord ID et al. Comparison of the effects ofsalbutamol and adrenal<strong>in</strong>e on airway smooth muscle contractility <strong>in</strong> vitroand on bronchial reactivity <strong>in</strong> vivo. Thorax 1994;49:1103–8.44 Abroug F, Nouira S, Bchir A, et al. A controlled trial of nebulizedsalbutamol and adrenal<strong>in</strong>e <strong>in</strong> acute severe asthma. Intensive <strong>Care</strong> Med1995;21:18–23.45 Molf<strong>in</strong>o NA, Nann<strong>in</strong>i LJ, Martelli AN, et al. <strong>Respiratory</strong> arrest <strong>in</strong>near-fatal asthma. N Engl J Med 1991;324:285–8.46 Silverman R. Treatment of acute asthma. A new look at the old and atthe new. Cl<strong>in</strong> Chest Med 2000;21:361–79.47 The COPD Guidel<strong>in</strong>es Group of the Standards of <strong>Care</strong> Committeeof the BTS. BTS guidel<strong>in</strong>es for the management of chronic obstructivepulmonary disease. Thorax 1997;52(Suppl 5):S1–28.48 Parameswaran K, Belda J, Rowe BH. Addition of <strong>in</strong>travenousam<strong>in</strong>ophyll<strong>in</strong>e to beta2-agonists <strong>in</strong> adults with acute asthma (CochraneReview). Cochrane Database Syst Rev 2000;4.49 Murciano D, Auclair MH, Pariente R, et al. A randomized, controlledtrial of theophyll<strong>in</strong>e <strong>in</strong> patients with severe chronic obstructive pulmonarydisease. N Engl J Med 1989;320:1521–5.50 Lakshm<strong>in</strong>arayan S, Sahn SA, Weil JV. Effect of am<strong>in</strong>ophyll<strong>in</strong>e onventilatory responses <strong>in</strong> normal man. Am Rev Respir Dis1978;117:33–8.51 Ward AJ, McKenniff M, Evans JM, et al. Theophyll<strong>in</strong>e: animmunomodulatory role <strong>in</strong> asthma? Am Rev Respir Dis1993;147:518–23.52 Rodrigo C, Rodrigo G. Treatment of acute asthma. Lack of therapeuticbenefit and <strong>in</strong>crease of the toxicity from am<strong>in</strong>ophyll<strong>in</strong>e given <strong>in</strong> additionto high doses of salbutamol delivered by metered-dose <strong>in</strong>haler with aspacer. Chest 1994;106:1071–6.53 Za<strong>in</strong>ud<strong>in</strong> BM, Ismail O, Yusoff K. Effect of add<strong>in</strong>g am<strong>in</strong>ophyll<strong>in</strong>e<strong>in</strong>fusion to nebulised salbutamol <strong>in</strong> severe acute asthma. Thorax1994;49:267–9.54 Mansel JK, Stogner SW, Norman JR. Face-mask CPAP and sodiumbicarbonate <strong>in</strong>fusion <strong>in</strong> acute, severe asthma and metabolic acidosis.Chest 1989;96:943–4.55 Shivaram U, Miro AM, Cash ME, et al. Cardiopulmonary responses tocont<strong>in</strong>uous positive airway pressure <strong>in</strong> acute asthma. J Crit <strong>Care</strong>1993;8:87–92.56 Meduri GU, Turner RE, Abou-Shala N, et al. Non<strong>in</strong>vasive positivepressure ventilation via face mask. First-l<strong>in</strong>e <strong>in</strong>tervention <strong>in</strong> patients withacute hypercapnic and hypoxemic respiratory failure. Chest1996;109:179–93.57 Mounta<strong>in</strong> RD, Sahn SA. Cl<strong>in</strong>ical features and outcome <strong>in</strong> patients withacute asthma present<strong>in</strong>g with hypercapnia. Am Rev Respir Dis1988;138:535–9.58 Leatherman J. Life-threaten<strong>in</strong>g asthma. Cl<strong>in</strong> Chest Med1994;15:453–79.59 Mansel JK, Stogner SW, Petr<strong>in</strong>i MF, et al. Mechanical ventilation <strong>in</strong>patients with acute severe asthma. Am J Med 1990;89:42–8.60 Zimmerman JL, Dell<strong>in</strong>ger RP, Shah AN, et al. Endotracheal <strong>in</strong>tubationand mechanical ventilation <strong>in</strong> severe asthma. Crit <strong>Care</strong> Med1993;21:1727–30.61 L’Hommedieu CS, Arens JJ. The use of ketam<strong>in</strong>e for the emergency<strong>in</strong>tubation of patients with status asthmaticus. Ann Emerg Med1987;16:568–71.62 Hemm<strong>in</strong>g A, MacKenzie I, F<strong>in</strong>fer S. Response to ketam<strong>in</strong>e <strong>in</strong> statusasthmaticus resistant to maximal medical treatment. Thorax1994;49:90–1.63 Howton JC, Rose J, Duffy S, et al. Randomized, double-bl<strong>in</strong>d,placebo-controlled trial of <strong>in</strong>travenous ketam<strong>in</strong>e <strong>in</strong> acute asthma. AnnEmerg Med 1996;27:170–5.64 Griff<strong>in</strong> D, Fairman N, Cours<strong>in</strong> D, et al. Acute myopathy dur<strong>in</strong>g treatmentof status asthmaticus with corticosteroids and steroidal muscle relaxants.Chest 1992;102:510–4.65 Leatherman JW, Fluegel WL, David WS, et al. Muscle weakness <strong>in</strong>mechanically ventilated patients with severe asthma. Am J Respir Crit<strong>Care</strong> Med 1996;153:1686–90.66 Margolis BD, Khachikian D, Friedman Y, et al. Prolonged reversiblequadriparesis <strong>in</strong> mechanically ventilated patients who received long-term<strong>in</strong>fusions of vecuronium. Chest 1991;100:877–8.67 Darioli R, Perret C. Mechanical controlled hypoventilation <strong>in</strong> statusasthmaticus. Am Rev Respir Dis 1984;129:385–7.68 Shapiro MB, Kleaveland AC, Bartlett RH. Extracorporeal life support forstatus asthmaticus. Chest 1993;103:1651–4.69 Scharf SM, Heimer D, Walters M. Bronchial challenge with roomtemperature isocapnic hyperventilation. A comparison with histam<strong>in</strong>echallenge. Chest 1985;88:586–93.70 Tuxen DV, Williams TJ, Sche<strong>in</strong>kestel CD, et al. Use of a measurement ofpulmonary hyper<strong>in</strong>flation to control the level of mechanical ventilation <strong>in</strong>patients with acute severe asthma. Am Rev Respir Dis 1992;146:1136–42.71 Thomas SH, O’Doherty MJ, Fidler HM, et al. Pulmonary deposition of anebulised aerosol dur<strong>in</strong>g mechanical ventilation. Thorax1993;48:154–9.72 Dhand R, Tob<strong>in</strong> MJ. Inhaled bronchodilator therapy <strong>in</strong> mechanicallyventilated patients. Am J Respir Crit <strong>Care</strong> Med 1997;156:3–10.73 Lange CF, F<strong>in</strong>lay WH. Overcom<strong>in</strong>g the adverse effect of humidity <strong>in</strong>aerosol delivery via pressurized metered-dose <strong>in</strong>halers dur<strong>in</strong>g mechanicalventilation. Am J Respir Crit <strong>Care</strong> Med 2000;161:1614–8.74 Harvey CJ, O’Doherty MJ, Page CJ, et al. Effect of a spacer onpulmonary aerosol deposition from a jet nebuliser dur<strong>in</strong>g mechanicalventilation. Thorax 1995;50:50–3.75 Harvey CJ, O’Doherty MJ, Page CJ, et al. Comparison of jet andultrasonic nebulizer pulmonary aerosol deposition dur<strong>in</strong>g mechanicalventilation. Eur Respir J 1997;10:905–9.76 Craven DE, Lichtenberg DA, Goularte TA, et al. Contam<strong>in</strong>atedmedication nebulizers <strong>in</strong> mechanical ventilator circuits. Source ofbacterial aerosols. Am J Med 1984;77:834–8.77 Gay PC, Patel HG, Nelson SB, et al. Metered dose <strong>in</strong>halers forbronchodilator delivery <strong>in</strong> <strong>in</strong>tubated, mechanically ventilated patients.Chest 1991;99:66–71.78 Duarte AG, Momii K, Bidani A. Bronchodilator therapy withmetered-dose <strong>in</strong>haler and spacer versus nebulizer <strong>in</strong> mechanicallyventilated patients: comparison of magnitude and duration of response.Respir <strong>Care</strong> 2000;45:817–23.79 Watts JI. Thoracic compression for asthma. Chest 1984;86:505.80 Fisher MM, Bowey CJ, Ladd-Hudson K. External chest compression <strong>in</strong>acute asthma: a prelim<strong>in</strong>ary study. Crit <strong>Care</strong> Med 1989;17:686–7.81 Eason J, Tayler D, Cottam S, et al. Manual chest compression for totalbronchospasm. Lancet 1991;337:366.82 Lang DM, Simon RA, Mathison DA, et al. Safety and possible efficacy offiberoptic bronchoscopy with lavage <strong>in</strong> the management of refractoryasthma with mucous impaction. Ann Allergy 1991;67:324–30.83 Patel A, Harrison E, Durward A, et al. Intratracheal recomb<strong>in</strong>ant humandeoxyribonuclease <strong>in</strong> acute life-threaten<strong>in</strong>g asthma refractory toconventional treatment. Br J Anaesth 2000;84:505–7.84 Eger EI. The pharmacology of isoflurane. Br J Anaesth1984;56:71–99S.85 Hirshman CA, Edelste<strong>in</strong> G, Peetz S, et al. Mechanism of action of<strong>in</strong>halational anesthesia on airways. Anesthesiology 1982;56:107–11.86 Korenaga S, Takeda K, Ito Y. Differential effects of halothane on airwaynerves and muscle. Anesthesiology 1984;60:309–18.87 Saulnier FF, Durocher AV, Deturck RA, et al. <strong>Respiratory</strong> andhemodynamic effects of halothane <strong>in</strong> status asthmaticus. Intensive <strong>Care</strong>Med 1990;16:104–7.88 Maltais F, Reissmann H, Navalesi P, et al. Comparison of static anddynamic measurements of <strong>in</strong>tr<strong>in</strong>sic PEEP <strong>in</strong> mechanically ventilatedpatients. Am J Respir Crit <strong>Care</strong> Med 1994;150:1318–24.89 Echeverria M, Gelb AW, Wexler HR, et al. Enflurane and halothane <strong>in</strong>status asthmaticus. Chest 1986;89:152–4.90 Padk<strong>in</strong> AJ, Baigel G, Morgan GA. Halothane treatment of severeasthma to avoid mechanical ventilation. Anaesthesia 1997;52:994–7.91 Gal TJ. Pulmonary mechanics <strong>in</strong> normal subjects follow<strong>in</strong>g endotracheal<strong>in</strong>tubation. Anesthesiology 1980;52:27–35.92 Habre W, Scalfaro P, Sims C, et al. <strong>Respiratory</strong> mechanics dur<strong>in</strong>gsevoflurane anesthesia <strong>in</strong> children with and without asthma. AnesthAnalg 1999;89:1177–81.93 Gluck EH, Onorato DJ, Castriotta R. Helium-oxygen mixtures <strong>in</strong> <strong>in</strong>tubatedpatients with status asthmaticus and respiratory acidosis. Chest1990;98:693–8.94 Kass JE, Terreg<strong>in</strong>o CA. The effect of heliox <strong>in</strong> acute severe asthma: arandomized controlled trial. Chest 1999;116:296–300.95 McNamara RM, Spivey WH, Skobeloff E, et al. Intravenous magnesiumsulfate <strong>in</strong> the management of acute respiratory failure complicat<strong>in</strong>gasthma. Ann Emerg Med 1989;18:197–9.96 Green SM, Rothrock SG. Intravenous magnesium for acute asthma:failure to decrease emergency treatment duration or need forhospitalization. Ann Emerg Med 1992;21:260–5.97 Bloch H, Silverman R, Mancherje N, et al. Intravenous magnesiumsulfate as an adjunct <strong>in</strong> the treatment of acute asthma. Chest1995;107:1576–81.98 Rowe BH, Bretzlaff JA, Bourdon C, et al. Magnesium sulfate for treat<strong>in</strong>gexacerbations of acute asthma <strong>in</strong> the emergency department. CochraneDatabase Syst Rev 2000;2.99 Chande VT, Skoner DP. A trial of nebulized magnesium sulfate toreverse bronchospasm <strong>in</strong> asthmatic patients. Ann Emerg Med1992;21:1111–5.100 Ciarallo L, Brousseau D, Re<strong>in</strong>ert S. Higher-dose <strong>in</strong>travenous magnesiumtherapy for children with moderate to severe acute asthma. Arch PediatrAdolescent Med 2000;154:979–83.101 Drazen JM, Israel E, O’Byrne PM. Treatment of asthma with drugsmodify<strong>in</strong>g the leukotriene pathway. N Engl J Med 1999;340:197–206.102 Kuitert LM, Barnes NC. Leukotriene receptor antagonists: useful <strong>in</strong> acuteasthma? Thorax 2000;55:255–6.103 Drazen JM, O’Brien J, Sparrow D, et al. Recovery of leukotriene E4 fromthe ur<strong>in</strong>e of patients with airway obstruction. Am Rev Respir Dis1992;146:104–8.104 Dworski R, Fitzgerald GA, Oates JA, et al. Effect of oral prednisone onairway <strong>in</strong>flammatory mediators <strong>in</strong> atopic asthma. Am J Respir Crit <strong>Care</strong>Med 1994;149:953–9.105 Dockhorn RJ, Baumgartner RA, Leff JA, et al. Comparison of the effectsof <strong>in</strong>travenous and oral montelukast on airway function: a double bl<strong>in</strong>d,placebo controlled, three period, crossover study <strong>in</strong> asthmatic patients.Thorax 2000;55:260–5.106 Evans DJ, Barnes PJ, Cluzel M, et al. Effects of a potentplatelet-activat<strong>in</strong>g factor antagonist, SR27417A, on allergen-<strong>in</strong>ducedasthmatic responses. Am J Respir Crit <strong>Care</strong> Med 1997;156:11–6.107 Hogman M, Frostell CG, Hedenstrom H, et al. Inhalation of nitric oxidemodulates adult human bronchial tone. Am Rev Respir Dis1993;148:1474–8.108 Molf<strong>in</strong>o NA, Nann<strong>in</strong>i LJ, Rebuck AS, et al. The fatality-prone asthmaticpatient. Follow-up study after near-fatal attacks. Chest 1992;101:621–3.109 F<strong>in</strong>fer SR, Garrard CS. Ventilatory support <strong>in</strong> asthma. Br J Hosp Med1993;49:357–60.


14 The pulmonary circulation and right ventricular failureK McNeil, J Dunn<strong>in</strong>g, N W Morrell.............................................................................................................................The lungs are the only organs that receive theentire cardiac output which is delivered at amean rest<strong>in</strong>g pulmonary arterial pressure of15 mm Hg. The capacitance pulmonary arteriesare larger <strong>in</strong> calibre and have th<strong>in</strong>ner walls thantheir systemic counterparts. Moreover, the pulmonarycirculation possesses little rest<strong>in</strong>g vasculartone and has a large reserve for recruitment ofvascular segments that are normally nonperfused.1Thus, the pulmonary circulation is alow pressure, low resistance circuit capable ofhandl<strong>in</strong>g large <strong>in</strong>creases <strong>in</strong> pulmonary blood flow(up to sixfold with strenuous exercise) with onlysmall changes <strong>in</strong> pressure. The ma<strong>in</strong>tenance of alow pulmonary capillary pressure is vital <strong>in</strong>preserv<strong>in</strong>g the function of the blood-gas barrier. 2In accordance with this low pressure circuit, theright ventricle (RV) is a th<strong>in</strong> muscle with limitedcontractile reserve, which has significant implicationsfor both the prognosis associated withsevere pulmonary hypertension (PHT) and for thepr<strong>in</strong>ciples underly<strong>in</strong>g the cl<strong>in</strong>ical management ofPHT and RV failure.Both PHT and RV dysfunction are commoncomplications of the complex medical disordersexperienced <strong>in</strong> the <strong>in</strong>tensive care unit. In mostcircumstances the PHT is mild or moderate <strong>in</strong>degree and associated with RV dysfunction ratherthan frank right heart failure. Occasionally, however,patients do present with life threaten<strong>in</strong>gPHT and associated RV failure requir<strong>in</strong>g promptand appropriate <strong>in</strong>tervention.Right heart dysfunction and PHT of vary<strong>in</strong>gseverity are commonly encountered <strong>in</strong> patientswith chronic lung disease and left ventricularfailure, but these specific entities will not be consideredfurther. This chapter will rather concentrateon the management of severe PHT <strong>in</strong> thesett<strong>in</strong>g of RV failure. In addition, we will discussthe relevance and treatment of PHT <strong>in</strong> the contextof acute respiratory distress syndrome (ARDS) <strong>in</strong>adults. Def<strong>in</strong>itions of PHT and calculations formean pulmonary artery pressure (PAP) areshown <strong>in</strong> table 14.1.In the <strong>in</strong>tensive care unit haemodynamics areusually measured us<strong>in</strong>g a flow directed, balloonTable 14.1 Def<strong>in</strong>itions of pulmonaryhypertension (PHT) and calculations formean pulmonary artery pressure (PAP)PHT def<strong>in</strong>itionsRestExerciseMildModerateSevereMean PAP>25 mm Hg>30 mm Hg45 mm HgCalculation of mean PAP: (1) mPAP = dPAP + (sPAP –dPAP)/3; (2) mPAP = (2 × dPAP + sPAP)/3 (these arethe same equation expressed <strong>in</strong> two commonly usedforms); d = diastolic, s = systolic.tipped pulmonary artery catheter. Cardiac outputis most commonly and conveniently determ<strong>in</strong>edby thermodilution techniques. It can also bederived via the Fick pr<strong>in</strong>ciple but this is not usedfrequently <strong>in</strong> cl<strong>in</strong>ical practice. Transoesophagealechocardiography can also be used to estimatecardiac output us<strong>in</strong>g Doppler imag<strong>in</strong>g. The procedurerequires sedation, however, and is not thereforeusually performed <strong>in</strong> cases with severe PHTunless the patient is <strong>in</strong>tubated (or undergo<strong>in</strong>ganother essential procedure).AETIOLOGY OF PULMONARYHYPERTENSIONThe WHO consensus conference <strong>in</strong> 1998 3 reclassifiedPHT accord<strong>in</strong>g to cl<strong>in</strong>icopathological criteria.Both the site of pathology (arterial or venous) andcausative factors (association with respiratorydisease/hypoxia, thromboembolic disease, or primaryvascular pathology) were <strong>in</strong>cluded. Thisclassification adds to our understand<strong>in</strong>g of themechanisms <strong>in</strong>volved and provides a good start<strong>in</strong>gpo<strong>in</strong>t <strong>in</strong> develop<strong>in</strong>g a rational cl<strong>in</strong>ical approachto the management of severe PHT.By this classification, the cause of PHT isdivided <strong>in</strong>to either an <strong>in</strong>tr<strong>in</strong>sic disease of the pulmonaryvessels or a vascular response to anotherdisease process. In general, treatment for this secondgroup should be directed at the underly<strong>in</strong>gdisease rather than at the pulmonary vasculatureper se. Probably the commonest causes of PHT <strong>in</strong>the ITU are raised left atrial pressure and hypoxaemia.Treatment should be directed at theunderly<strong>in</strong>g cardiac or respiratory disease, respectively.THE PULMONARY CIRCULATION IN ARDSThe acute respiratory distress syndrome (ARDS)is characterised by non-hydrostatic pulmonaryoedema and refractory hypoxaemia, and complicatesup to 25% of cases of the systemic<strong>in</strong>flammatory response syndrome The consensusdef<strong>in</strong>itions of ARDS and acute lung <strong>in</strong>jury (ALI)are shown <strong>in</strong> table 14.2. PHT with <strong>in</strong>creased pulmonaryvascular resistance is common, evenwhen systemic vascular resistance is low. Thedegree of pulmonary arterial hypertension isusually mild to moderate but promotes the accumulationof extravascular lung water and cancause right ventricular dysfunction, reduc<strong>in</strong>gejection fraction and cardiac output. The presenceof PHT has been shown to be an adverse prognostic<strong>in</strong>dicator <strong>in</strong> patients with ARDS. 45Initially, a number of factors may contribute tothe <strong>in</strong>crease <strong>in</strong> PAP <strong>in</strong> ARDS. 6 Increased circulat<strong>in</strong>glevels of vasoactive mediators such asseroton<strong>in</strong>, endothel<strong>in</strong>-1, thromboxane and leukotrienesmay contribute to the <strong>in</strong>crease <strong>in</strong> pulmonaryvascular tone. There may also be animportant contribution from <strong>in</strong>creased dischargefrom the sympathetic nervous system. Althoughhypoxic pulmonary vasoconstriction may play


94 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Table 14.2 Recommended criteria for def<strong>in</strong>ition ofacute lung <strong>in</strong>jury (ALI) and acute respiratory distresssyndrome (ARDS). Modified from Bernard et al 42ALIARDSTim<strong>in</strong>gAcuteonsetAcuteonsetOxygenationPaO 2 /FiO 2


Pulmonary circulation and right ventricular failure 95<strong>in</strong> these situations it is usually possible at least to amelioratethe added oxygen desaturation associated with sleep andexercise. As <strong>in</strong> any low cardiac output state, anticoagulation isdesirable.With <strong>in</strong>tr<strong>in</strong>sic disease of the pulmonary arteries, severalvasodilator strategies can be considered. For patients requir<strong>in</strong>gmechanical ventilation <strong>in</strong>haled NO can be used to maximiseV/Q match<strong>in</strong>g, as vasodilation only occurs <strong>in</strong> ventilatedareas. 19Prostacycl<strong>in</strong> (PGI 2) is also an effective pulmonaryvasodilator but, when adm<strong>in</strong>istered <strong>in</strong>travenously, can result<strong>in</strong> worsen<strong>in</strong>g of V/Q match<strong>in</strong>g. 20 The short biological half lifeof <strong>in</strong>haled NO (seconds) leads to a greater effect on thepulmonary than the systemic circulation, its biological effectsbe<strong>in</strong>g rapidly elim<strong>in</strong>ated by reaction with haemoglob<strong>in</strong>. Prostacycl<strong>in</strong>has a rather longer half life <strong>in</strong> the circulation (1–2m<strong>in</strong>utes) and usually lowers pulmonary and systemic vascularresistance, although the systemic effects can be m<strong>in</strong>imised by<strong>in</strong>haled treatment. Calcium channel blockers should not beused <strong>in</strong> patients with significant cardiac dysfunction as theirnegative <strong>in</strong>otropic effects may further impair RVperformance. 21Nitric oxide donors such as nitroprusside ornitrates are rarely effective <strong>in</strong> this sett<strong>in</strong>g, and usually exacerbatethe systemic hypotension associated with the low cardiacoutput syndrome.Right heart dysfunctionThe RV is designed to work with a low pressure circuit and, assuch, has limited contractile reserve. For this reason RVsupport is aimed at reduc<strong>in</strong>g afterload. Severe acute RVdysfunction associated with PHT may present dur<strong>in</strong>g heart orlung transplantation or surgery for pulmonary embolus orsevere mitral valve disease. Inhaled NO may be useful <strong>in</strong> thissett<strong>in</strong>g to decrease pulmonary vascular resistance withoutreduc<strong>in</strong>g systemic arterial pressure, which is essential for thema<strong>in</strong>tenance of coronary perfusion to the right ventricle.Inhaled NO <strong>in</strong> the dose range 20–40 ppm may benefit thesepatients. 14Unless the pulmonary vascular resistance can bereduced, <strong>in</strong>otropes are <strong>in</strong>effective, impos<strong>in</strong>g more work on astruggl<strong>in</strong>g RV. Inotropes can transiently improve cardiacoutput for 6–12 hours but <strong>in</strong>evitably the RV fails, result<strong>in</strong>g <strong>in</strong>a lethal downward spiral of <strong>in</strong>creas<strong>in</strong>g <strong>in</strong>otrope requirementsbut dim<strong>in</strong>ish<strong>in</strong>g effect. Inotropes are, however, useful for support<strong>in</strong>gor augment<strong>in</strong>g RV contractility <strong>in</strong> situations where thepulmonary vascular resistance can be reduced throughconcomitant adm<strong>in</strong>istration of vasodilator therapy. In thisrespect, a phosphodiesterase <strong>in</strong>hibitor such as enoximone 22 isour preferred choice as its vasodilator properties contrast withthe pulmonary vasoconstrictor effects characteristic of catecholam<strong>in</strong>es.Intra-aortic balloon counterpulsation is useful for shortterm RV support, 23augment<strong>in</strong>g coronary blood flow and<strong>in</strong>creas<strong>in</strong>g central systemic blood pressure, thereby reduc<strong>in</strong>gthe need for pressor agents such as noradrenal<strong>in</strong>e which arepotent pulmonary vasoconstrictors. Most devices used <strong>in</strong> thecontext of RV support are modified from those used to supportthe fail<strong>in</strong>g left heart. When the lung function rema<strong>in</strong>sadequate to allow sufficient oxygenation and carbon dioxideremoval, mechanical assist devices alone may be used to supportthe RV. When severe lung <strong>in</strong>jury accompanies RV failure,mechanical RV support can be <strong>in</strong>corporated <strong>in</strong>to either extracorporealmembrane oxygenation (ECMO) or extracorporealcarbon dioxide removal (ECCOR) circuits.Paracorporeal devices such as the Abiomed are implantedthrough a sternotomy us<strong>in</strong>g right atrial or ventricular cannulationfor dra<strong>in</strong>age to the pump chamber, with return to thepulmonary artery through a vascular conduit sutured to thepulmonary trunk. Such a procedure is highly <strong>in</strong>vasive and thesubsequent management of the pump is complex. In particular,ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g a balance between vasodilator agents,<strong>in</strong>otropes, and fill<strong>in</strong>g pressures may be difficult to achieve.Overflow<strong>in</strong>g the pump can result <strong>in</strong> gross pulmonary oedema,compound<strong>in</strong>g an already difficult situation. A further sourceof difficulty may be experienced with the management ofanticoagulation. While it is important to achieve adequateanticoagulation, <strong>in</strong>tracranial haemorrhage may result if tightcontrol is not ma<strong>in</strong>ta<strong>in</strong>ed. Success is most likely <strong>in</strong> centreswhere there is familiarity with the management of thesedevices and their complications.Although the use of ECMO is more complex, cannulationand the establishment of the ECMO circuit is less <strong>in</strong>vasivethan that required for most paracorporeal pulsatile ventricularassist devices. This technique is only of value when the <strong>in</strong>sultnecessitat<strong>in</strong>g its use is reversible. Although well established <strong>in</strong>children, its adoption <strong>in</strong> adult practice has been lesswidespread. Key components to successful <strong>in</strong>tervention<strong>in</strong>clude early <strong>in</strong>stitution (ventilated less than 5 days), aflexible approach to cannulation (established percutaneouslyvia femoral and jugular routes), and read<strong>in</strong>ess to supportother systems <strong>in</strong>clud<strong>in</strong>g the kidneys and liver.Any attempt to support the RV with a mechanical device <strong>in</strong>the face of PHT is complicated. A multidiscipl<strong>in</strong>ary approachmust be adopted, and there is no simple unify<strong>in</strong>g technique.The advent of small axial and rotary blood pumps currentlyundergo<strong>in</strong>g trials <strong>in</strong> left ventricular dysfunction may improvethe outlook <strong>in</strong> this area <strong>in</strong> years to come.Atrial septostomy has been used <strong>in</strong> the treatment ofpatients with severe PHT and RV compromise. 24 This treatmentevolved from the observation that patients with PPH and apatent foramen ovale lived longer than those with no septaldefect. 25 Creat<strong>in</strong>g a shunt at the atrial level decompresses theright sided chambers and augments left atrial fill<strong>in</strong>g with aconcomitant <strong>in</strong>crease <strong>in</strong> cardiac output and systemic oxygentransport. The result<strong>in</strong>g reduction <strong>in</strong> RV end diastolic pressureand wall tension are postulated to improve Starl<strong>in</strong>g haemodynamicsand RV contractility. Although the result<strong>in</strong>g right toleft shunt causes systemic oxygen desaturation (which isespecially marked with exercise when PAP rises), this canusually be controlled with supplemental oxygen.The cl<strong>in</strong>ical benefits reported follow<strong>in</strong>g septostomy <strong>in</strong>cluderesolution of syncopal and pre-syncopal episodes, decreasedcough, decreased systemic venous congestion, and improvedexercise tolerance. Our own experience of this proceduresuggests it is most effective before the onset of severe RV dysfunctionand, conversely, as reported by others, 26it has notbeen effective <strong>in</strong> patients with end stage right heart failure oracute right heart failure severe enough to require admission tothe ITU. Atrial septostomy has been used primarily <strong>in</strong>advanced PPH as a bridge to heart-lung transplantation, 24although its exact role (<strong>in</strong> particular the optimal tim<strong>in</strong>g of theprocedure) is not known. As our own experience with thisprocedure has developed, it is be<strong>in</strong>g used earlier <strong>in</strong> the courseof disease. Experience of the procedure <strong>in</strong> any form of severePHT is very limited and at present there are no controlledcl<strong>in</strong>ical studies <strong>in</strong> any disease group.Thromboembolic diseaseThe most common cause of acute severe PHT is massive centralpulmonary thromboembolism. Some of the mechanisms<strong>in</strong>volved <strong>in</strong> the development of shock <strong>in</strong> the sett<strong>in</strong>g of acutemassive pulmonary embolism (PE) are shown <strong>in</strong> fig 14.1. Theoverall mortality from massive PE is 6–8%, <strong>in</strong>creas<strong>in</strong>g to 30%if complicated by systemic hypotension. 27Of those patientswho fail to survive, 67% die with<strong>in</strong> 1 hour of the onset ofsymptoms. 28The diagnosis of massive PE is suggested by the cl<strong>in</strong>icalpresentation of right ventricular failure, a normal or oligaemicchest radiograph, and a suggestive ECG (right ventricularstra<strong>in</strong>). 29 In more stable patients there may be time to organisespiral CT pulmonary angiography. Echocardiography17 30(either transthoracic or transoesophageal) may reveal thrombus<strong>in</strong> the pulmonary outflow tract or show signs of right ventricular18 31dysfunction/hypok<strong>in</strong>esis.


96 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Reduced cross sectional areapulmonary vascular bedRelease of vasoactivesubstancesRaised MPAPHypoxaemiaLV fill<strong>in</strong>gRV afterloadmyocardial wall tensionmyocardial oxygen consumptionReduced COReduced PvO 2ShockFigure 14.1 Factors <strong>in</strong>volved <strong>in</strong> the generation of acute right ventricular failure follow<strong>in</strong>g massive pulmonary embolism. MPAP=meanpulmonary artery pressure; CO=cardiac output; LV=left ventricle; PvO 2=ventricular oxygen tension.Oxygen and analgesia should be given to all patients immediately.Invasive monitor<strong>in</strong>g of the central venous pressure willguide cautious fluid and colloid replacement to optimise rightsided fill<strong>in</strong>g pressures. The central venous pressure should bema<strong>in</strong>ta<strong>in</strong>ed at 15–20 cm H 2O. Overfill<strong>in</strong>g worsens right ventricularfunction, but <strong>in</strong>adequate fill<strong>in</strong>g (or <strong>in</strong>deed overlyaggressive diuresis) also compromises RV haemodynamics. Ifhaemodynamic compromise is present and there are nocontra<strong>in</strong>dications (shown <strong>in</strong> box 14.1), thrombolysis shouldbe considered for acute massive PE. 32 33 The rationale for this isthe greater mortality <strong>in</strong> patients with right ventriculardysfunction follow<strong>in</strong>g acute PE. 34 Thrombolysis leads to morerapid restoration of RV function than hepar<strong>in</strong> alone. 28However, the potential benefits must justify the 1% risk of cerebraland fatal bleed<strong>in</strong>g, 35 and the effects of thrombolysis onmortality still need to be confirmed by a prospectiverandomised trial. Two hour <strong>in</strong>fusion regimens of streptok<strong>in</strong>ase(1.5 million units), urok<strong>in</strong>ase and recomb<strong>in</strong>ant tissueplasm<strong>in</strong>ogen activator (rt-PA; 100 mg) followed by a hepar<strong>in</strong><strong>in</strong>fusion have similar efficacy and safety profiles. 36 Thrombolysismay be considered <strong>in</strong> all age groups and <strong>in</strong> postoperativepatients. The risk of major haemorrhage with these agents<strong>in</strong>creases with <strong>in</strong>creas<strong>in</strong>g age and body mass <strong>in</strong>dex. Bolus andfront loaded regimens (adm<strong>in</strong>istered over


Pulmonary circulation and right ventricular failure 97Massive PEHaemodynamiccompromiseHaemodynamicallystableEchocardiographyRV dysfunctionNo RV dysfunctionConsiderthrombolysisConsiderthrombolysisCardiogenicshockContra<strong>in</strong>dicatedPatientstableNo contra<strong>in</strong>dicationContra<strong>in</strong>dicatedSurgicalembolectomyTransvenousembolectomyorCatheterfragmentationThrombolysisAnticoagulationFigure 14.2Therapeutic approach to massive PE.Appropriate treatment h<strong>in</strong>ges on identification of the underly<strong>in</strong>gcause and effective reduction <strong>in</strong> RV afterload. The commonestcause of acute severe PHT is massive pulmonarythromboembolism and, if no contra<strong>in</strong>dications exist, thrombolytictherapy is the treatment of choice. Pulmonary vasodilatorssuch as <strong>in</strong>travenous prostacycl<strong>in</strong> or <strong>in</strong>haled NO areoften effective <strong>in</strong> other cases where <strong>in</strong>creased pulmonary vasculartone is present. Whatever the underly<strong>in</strong>g cause, withouteffective afterload reduction the RV will <strong>in</strong>evitably fail and it isthus of the utmost importance that <strong>in</strong>otropes are not relied onto support the RV without effective treatment of theunderly<strong>in</strong>g problem.REFERENCES1 Rodman DM, Voelkel NF. Regulation of vascular tone. In: Crystal RG,West JB, et al, eds. The lung. Scientific Foundations. Philadelphia:Lipp<strong>in</strong>cott-Raven, 1997: 1473–92.2 West JB. Pulmonary capillary stress failure. J Appl Physiol2000;89:2483–9.3 Rich S, ed. Primary pulmonary hypertension: executive summary of aWHO meet<strong>in</strong>g. Geneva: World Health Organisation, 1998.4 Villar J, Blazquez MA, Lubillo S, et al. Pulmonary hypertension <strong>in</strong> acuterespiratory failure. Crit <strong>Care</strong> Med 1989;17:523–6.5 Leeman M. Pulmonary hypertension <strong>in</strong> acute respiratory distresssyndrome. Monaldi Arch Chest Dis 1999;54:146–9.6 Wort SJ, Evans TW. The role of the endothelium <strong>in</strong> modulat<strong>in</strong>g vascularcontrol <strong>in</strong> sepsis and related conditions. Br Med Bull 1999;55:30–48.7 Leeman M. The pulmonary circulation <strong>in</strong> acute lung <strong>in</strong>jury: a review ofsome recent advances. Intensive <strong>Care</strong> Med 1991;17:254–60.8 Santos C, Ferrer M, Roca J, et al. Pulmonary gas exchange response tooxygen breath<strong>in</strong>g <strong>in</strong> acute lung <strong>in</strong>jury. Am J Respir Crit <strong>Care</strong> Med2000;161:26–31.9 Tomashefski JF, Davies P, Boggis C, et al. The pulmonary vascularlesions of the adult respiratory distress syndrome. Am J Pathol1983;112:112–26.10 Naeije R. Medical treatment of pulmonary hypertension <strong>in</strong> acute lungdisease. Eur Respir J 1993;6:1521–8.11 Radermacher P, Santak B, Wust HJ, et al. Prostacycl<strong>in</strong> and rightventricular function <strong>in</strong> patients with pulmonary hypertension associatedwith ARDS. Intensive <strong>Care</strong> Med 1990;16:227–32.12 Rossa<strong>in</strong>t R, Falke KJ, Lopez F, et al. Inhaled nitric oxide for the adultrespiratory distress syndrome. N Engl J Med 1993;328:399–405.13 Fierobe L, Brunet F, Dha<strong>in</strong>aut JF, et al. Effect of <strong>in</strong>haled nitric oxide onright ventricular function <strong>in</strong> adult respiratory distress syndrome. Am JRespir Crit <strong>Care</strong> Med 1995;151:1414–9.14 Cuthbertson BH, Dell<strong>in</strong>ger P, Dyar OJ, et al. UK guidel<strong>in</strong>es for the useof <strong>in</strong>haled nitric oxide therapy <strong>in</strong> adult ICUs. Intensive <strong>Care</strong> Med1997;23:1212–8.15 Lund<strong>in</strong> S, Mang H, Smithies M, et al. Inhalation of nitric oxide <strong>in</strong> acutelung <strong>in</strong>jury: results of a European multicentre study. The European StudyGroup of <strong>in</strong>haled nitric oxide. Intensive <strong>Care</strong> Med 1999;25:911–9.16 McIntyre KM, Sasahara AA. Determ<strong>in</strong>ants of right ventricular functionand haemodynamics after pulmonary embolism. Chest1974;65:534–43.17 Remy-Jard<strong>in</strong> M, Remy J, Deschildre F, et al. Diagnosis of pulmonaryembolism with spiral CT: comparison with pulmonary angiography andsc<strong>in</strong>tigraphy. Radiology 1996;200:699–706.18 Jard<strong>in</strong> F, Dubourg O, Bourdarias JP. Echocardiographic pattern of acutecor pulmonale. Chest 1997;111:209–17.19 Hayward CS, Kelly RP, MacDonald PS. Inhaled nitric oxide <strong>in</strong>cardiology practice. Cardiovasc Res 1999;43:628–38.20 Otulana B, Higenbottam T. The role of physiological deadspace andshunt <strong>in</strong> the gas exchange of patients with pulmonary hypertension: astudy of exercise and prostacycl<strong>in</strong> <strong>in</strong>fusion. Eur Respir J 1988;1:732–7.21 Ga<strong>in</strong>e SP, Rub<strong>in</strong> LJ. Primary pulmonary hypertension. Lancet1998;352:719–25.22 Bauer J, Dapper F, Demirakca S, et al. Perioperative management ofpulmonary hypertension after heart transplantation <strong>in</strong> childhood. J HeartLung Transplant 1997;16:1238–47.23 Arafa OE, Geiran OR, Andersen K, et al. Intraaortic balloon pump<strong>in</strong>gfor predom<strong>in</strong>antly right ventricular failure after heart transplantation. AnnThorac Surg 2000;70:1587–93.24 Rothman A, Sklansky MS, Lucas VW, et al. Atrial septostomy as abridge to lung transplantation <strong>in</strong> patients with severe pulmonaryhypertension. Am J Cardiol 1999;84:682–6.25 Rozkovec A, Montanes P, Oakley CM. Factors that <strong>in</strong>fluence theoutcome of primary pulmonary hypertension. Br Heart J1986;55:449–58.26 Rich S, Dod<strong>in</strong> E, McLaughl<strong>in</strong> VV. Usefulness of atrial septostomy as atreatment for primary pulmonary hypertension and guidel<strong>in</strong>es for itsapplication. Am J Cardiol 1997;80:369–71.27 Alpert JS, Smith R, Carlson CJ, et al. Mortality <strong>in</strong> patients treated forpulmonary embolism. JAMA 1976;236:1477–80.28 Dalen JE, Alpert JS. Natural history of pulmonary embolism. ProgCardiovasc Dis 1975;17:259–70.29 British Thoracic Society Standards of <strong>Care</strong> Committee. Suspectedpulmonary embolism: a practical approach. Thorax 1997;52(Suppl4):S1–24.30 Blum AG, Delfau F, Grignon B, et al. Spiral-computed tomographyversus pulmonary angiography <strong>in</strong> the diagnosis of acute massivepulmonary embolism. Am J Cardiol 1994;74:96–8.31 McConnell MV, Solomon SD, Rayan ME, et al. Regional right ventriculardysfunction detected by echocardiography <strong>in</strong> acute pulmonary embolism.Am J Cardiol 1996;78:469–73.32 Jerjes-Sanchez C, Ramirez-Rivera A, Garcia ML, et al. Streptok<strong>in</strong>aseand hepar<strong>in</strong> versus hepar<strong>in</strong> alone <strong>in</strong> massive pulmonary embolism: arandomised controlled trial. J Thromb Thrombolysis 1995;2:227–9.33 Goldhaber SZ. Pulmonary embolism. N Engl J Med 1998;339:93–104.34 Ribeiro A, L<strong>in</strong>dmarker P, Juhl<strong>in</strong>-Dannfelt A, et al. EchocardiographyDoppler <strong>in</strong> pulmonary embolism: right ventricular dysfunction as apredictor of mortality rate. Am Heart J 1997;134:479–87.35 Konstant<strong>in</strong>ides S, Geibel A, Kasper W. Submassive and massivepulmonary embolism: a target for thrombolytic therapy. Thromb Haemost1999;82(Suppl 1):104–8.


98 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>36 Meneveau N, Schiele F, Metz D, et al. Comparative efficacy of atwo-hour regimen of streptok<strong>in</strong>ase versus alteplase <strong>in</strong> acute massivepulmonary embolism: immediate cl<strong>in</strong>ical and hemodynamic outcome andone-year follow up. J Am Coll Cardiol 1998;31:1057–63.37 Layish DT, Tapson VF. Pharmacologic hemodynamic support <strong>in</strong> massivepulmonary embolism. Chest 1997;111:218–24.38 Konstant<strong>in</strong>ides S, Geibel A, Olschewski M, et al. Association betweenthrombolytic treatment and the prognosis of hemodynamically stablepatients with major pulmonary embolism: results of a multicentre registry.Circulation 1997;96:882–8.39 Elliott CG. Embolectomy, catheter extraction, or disruption of pulmonaryemboli: editorial review. Curr Op<strong>in</strong> Pulm Med 1995;1:298–302.40 Doerge H, Schoendube FA, Voss M, et al. Surgical therapy of fulm<strong>in</strong>antpulmonary embolism: early and late results. Thorac Cardiovasc Surg1999;47:9–13.41 Gulba DC, Schmid C, Borst HG, et al. Medical compared with surgicaltreatment for massive pulmonary embolism. Lancet 1994;343:576–7.42 Bernard GR, Artigas A, Brigham KL, et al. The American-Europeanconsensus conference on ARDS. Am J Respir Crit <strong>Care</strong> Med1994;149:818–24.43 Archibald CJ, Auger WR, Fedullo PF, et al. Long-term outcome afterpulmonary thromboendarterectomy. Am J Respir Crit <strong>Care</strong> Med1999;160:523–8.


15 Thoracic trauma, <strong>in</strong>halation <strong>in</strong>jury andpost-pulmonary resection lung <strong>in</strong>jury <strong>in</strong> <strong>in</strong>tensive careE D Moloney, MJDGriffiths, P Goldstraw.............................................................................................................................Trauma to the respiratory tract may cause respiratoryfailure and critical illness by severalmechanisms. In this article we discuss threesuch conditions that are best managed by amultidiscipl<strong>in</strong>ary approach between physiciansand surgeons. Thoracic trauma and <strong>in</strong>halation<strong>in</strong>jury are common causes of respiratory failureand critical illness often <strong>in</strong> young patients whohave a good prognosis if the orig<strong>in</strong>al <strong>in</strong>sult is survivable.The pathogenesis of post-pulmonaryresection lung <strong>in</strong>jury is discussed as a welldef<strong>in</strong>ed human model of the acute respiratorydistress syndrome (ARDS) whose management isdiscussed <strong>in</strong> detail elsewhere <strong>in</strong> this book.Despite the disparate nature of these conditions,common themes emerge <strong>in</strong> the supportivemanagement of such patients while they recover(fig 15.1). For example, atelectasis and sputumretention follow all major pulmonary <strong>in</strong>sults,<strong>in</strong>clud<strong>in</strong>g surgery, as a consequence of impairedcough and shallow respiration caused by pa<strong>in</strong> andweakness. The tendency to develop respiratoryfailure and pneumonia may be m<strong>in</strong>imised byadequate pa<strong>in</strong> relief, mobilisation, and physiotherapy.Intermittent non-<strong>in</strong>vasive ventilatorysupport and tracheal aspiration facilitated by anasopharyngeal airway or a m<strong>in</strong>i-tracheostomyhelp a patient with an <strong>in</strong>adequate cough.THORACIC TRAUMAEpidemiologyThoracic trauma accounts for 20–25% of deathsfollow<strong>in</strong>g trauma and contributes to another 25%of trauma-related deaths. Blunt thoracic <strong>in</strong>jurieshave a higher mortality than penetrat<strong>in</strong>g <strong>in</strong>juriesbecause of a higher <strong>in</strong>cidence of <strong>in</strong>jury to otherorgans. However, <strong>in</strong> both adults and children thepredom<strong>in</strong>ant cause of death <strong>in</strong> patients withblunt thoracic trauma is head <strong>in</strong>jury. 12 Approximatelyone third of all patients admitted totrauma centres have susta<strong>in</strong>ed serious <strong>in</strong>juries tothe chest and the lung parenchyma is <strong>in</strong>jured <strong>in</strong> ahigh proportion of these. 3 The lead<strong>in</strong>g causes ofblunt and penetrat<strong>in</strong>g thoracic trauma are roadtraffic accidents, and stabb<strong>in</strong>gs and gun shotwounds, respectively.Blunt thoracic trauma can produce a spectrumof <strong>in</strong>juries—<strong>in</strong>clud<strong>in</strong>g rib fractures, pneumothorax,flail segments and pulmonary contusion (fig15.2)—all of which can impair pulmonaryfunction. 4Mortality for patients with three ormore rib fractures is double that of patients withoutrib fractures (1.8% versus 3.9%), 2and thepresence of thoracic trauma <strong>in</strong>creases the overallrisk of death from 27% to 33%. 1 The presence ofthree or more rib fractures <strong>in</strong> an <strong>in</strong>jured adultidentifies a small group with an <strong>in</strong>creased risk ofsplenic and liver <strong>in</strong>jury. 1 Similarly, the presence ofpneumothorax, haemothorax, pulmonary contusion,and a flail segment are each associated withan <strong>in</strong>creased risk of death. 1Thoracic trauma is a significant aetiologicalfactor <strong>in</strong> up to 20% of the 1 500 000 cases per yearof ARDS <strong>in</strong> the United States. 5 ARDS may occur asa result of direct lung <strong>in</strong>jury or from the systemicsequelae of extrathoracic <strong>in</strong>jury. 67In mechanicallyventilated trauma patients, 18% developedARDS when pulmonary contusion was their onlyrisk factor. However, the <strong>in</strong>cidence rose to 35%when other risk factors were present. 8 Pulmonarycontusion is the most common <strong>in</strong>jury seen <strong>in</strong>association with thoracic trauma, occurr<strong>in</strong>g <strong>in</strong>30–75% of patients suffer<strong>in</strong>g major thoracic<strong>in</strong>jury. 9 Contusion occurs more frequently follow<strong>in</strong>gblunt thoracic <strong>in</strong>jury and is an important factorlead<strong>in</strong>g to respiratory failure. It causes disruptionof the alveolar-capillary membrane and<strong>in</strong>creased pulmonary shunt<strong>in</strong>g, which may beexacerbated by a reduction <strong>in</strong> compliance causedby rib fracture or by attendant pa<strong>in</strong> and musclespasm. In one series 11% of patients with seriousisolated pulmonary contusions died, whereas themortality was much higher (22%) <strong>in</strong> patientswith associated <strong>in</strong>juries. 10<strong>Management</strong>The first priorities <strong>in</strong> manag<strong>in</strong>g thoracic traumaare secur<strong>in</strong>g an adequate airway and ventilation,controll<strong>in</strong>g bleed<strong>in</strong>g, and restor<strong>in</strong>g adequatetissue perfusion. Mechanical ventilation is <strong>in</strong>dicatedwhen gas exchange is <strong>in</strong>adequate, despiteaggressive pa<strong>in</strong> management and pulmonary11 12toilet. The cervical sp<strong>in</strong>e should be stabiliseddur<strong>in</strong>g tracheal <strong>in</strong>tubation <strong>in</strong> cases of severe blunttrauma. If an arrhythmia, hypotension, and/orchanges <strong>in</strong> the central nervous system occur dur<strong>in</strong>gor immediately after endotracheal <strong>in</strong>tubation,air embolism should be suspected. Air embolismis a rare cause of <strong>in</strong>tractable shock and cardiacarrest follow<strong>in</strong>g thoracic trauma. 13Decreased breath sounds on one side, hypotension,neck ve<strong>in</strong> distension, and deviation of thetrachea suggest the presence of a tension pneumothoraxwarrant<strong>in</strong>g immediate needle decompressionor tube thoracostomy. 14 A small well toleratedpneumothorax <strong>in</strong> a spontaneouslybreath<strong>in</strong>g patient may suddenly be converted <strong>in</strong>toa tension pneumothorax after positive pressure isapplied to the airway. Nearly all air leaks willeventually seal if the lung is fully expanded andthe chest tube is placed on suction. Generally,surgical <strong>in</strong>tervention is considered if an air leakpersists for 14 days. The exception to this rule isthe patient with a major bronchial <strong>in</strong>jury, which issuggested by a very large air leak, an <strong>in</strong>ability tore-expand the lung with closed tube thoracostomy,and usually confirmed by bronchoscopy. Ingeneral, <strong>in</strong>juries to the trachea or major bronchirequire operative repair. 14Hypotension without neck ve<strong>in</strong> distension, trachealdeviation and dullness to percussion


100 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Thoracic trauma/surgery or <strong>in</strong>halation <strong>in</strong>jury/burnsNeurological Chest wall AirwayAlveolar-capillarymembrane↓Ventilation & cough*↑Mucus secretion*↓Gas exchangeAtelectasis<strong>Respiratory</strong> failureHyperoxia ßVentilator-associated pneumoniaVentilator-associated lung <strong>in</strong>juryTracheal <strong>in</strong>tubation &Mechanical ventilationFigure 15.1 Common pathways to respiratory failure follow<strong>in</strong>gthoracic trauma or <strong>in</strong>halation <strong>in</strong>jury. *Optimis<strong>in</strong>g pa<strong>in</strong> relief, sputumclearance and respiratory mechanics are crucial <strong>in</strong> prevent<strong>in</strong>grespiratory failure at an early stage <strong>in</strong> the recovery period.§Hyperoxia causes reabsorption atelectasis.suggests a significant haemothorax. In most cases bleed<strong>in</strong>gwill stop once the lung has been re-expanded us<strong>in</strong>g tube thoracostomy.Indications for thoracotomy are an <strong>in</strong>itial loss of atleast 1500 ml of blood upon placement of the chest tube, orcont<strong>in</strong>ued bleed<strong>in</strong>g of 200–300 ml/h. 14Haemorrhage is themajor preventable cause of trauma deaths <strong>in</strong> hospital, andhypotension follow<strong>in</strong>g <strong>in</strong>jury should be considered to be dueto hypovolaemia until proved otherwise. Patients who have areta<strong>in</strong>ed haemothorax are at risk of empyema or a fibrothoraxwith lung entrapment. Non-operative management us<strong>in</strong>gclosed tube thoracostomy has a lower likelihood of successwhen there is a significant amount of reta<strong>in</strong>ed clot, if the fluidis viscous, or if it has a low pH. A larger empyema generallyrequires thoracotomy with decortication and dra<strong>in</strong>age, particularlyif an extensive pleural peel has developed. 14Sternal fracture may be associated with cardiac <strong>in</strong>jury andis an <strong>in</strong>dication for a 12 lead electrocardiogram (ECG) andcardiac monitor<strong>in</strong>g for 12 hours. Acute cardiac tamponadeshould be suspected <strong>in</strong> patients who have refractory shock andevidence of raised central venous pressure. Pericardiocentesisand emergency thoracotomy may be life sav<strong>in</strong>g <strong>in</strong> this groupof patients. 14 Echocardiography can be used as a non-<strong>in</strong>vasivemeans of evaluat<strong>in</strong>g stable patients with a possible myocardialcontusion or tamponade. Disruption of the aorta and greatvessels, suggested by fracture of the first and second ribs,requires operative management that needs to be performedimmediately if the patient is haemodynamically unstable.Aortography or chest CT scann<strong>in</strong>g are def<strong>in</strong>itive tests for bluntaortic <strong>in</strong>jury and should be obta<strong>in</strong>ed <strong>in</strong> patients who have awide mediast<strong>in</strong>um. Transoesophageal echocardiography canbe performed <strong>in</strong>stead of aortography <strong>in</strong> selected patients,allow<strong>in</strong>g good views of the descend<strong>in</strong>g aorta but not the upperascend<strong>in</strong>g aorta or aortic arch. 14Penetrat<strong>in</strong>g <strong>in</strong>juries to theoesophagus are relatively uncommon, and blunt <strong>in</strong>juries to theoesophagus are rare. In patients not undergo<strong>in</strong>g surgicalexploration of a penetrat<strong>in</strong>g wound, the diagnosis of oesophageal<strong>in</strong>jury can be made from radiographic studies. In general,oesophageal <strong>in</strong>juries are managed operatively. If the <strong>in</strong>jury ismore than 24 hours old, mediast<strong>in</strong>al <strong>in</strong>flammation may makeit impossible to close the perforation; <strong>in</strong> this case, dra<strong>in</strong>agewith or without diversion may be the only option. 14Patients with rib fractures are more likely to require thoracotomyand their length of stay <strong>in</strong> the ICU is also significantly<strong>in</strong>creased. Multiple rib fractures and flail chest (at least threecontiguous ribs, each fractured <strong>in</strong> two places) suggest associated<strong>in</strong>trathoracic <strong>in</strong>jury 15 and pneumothorax orhaemothorax. 16 The mortality rate for patients with flail chestFigure 15.2 Radiographic features of thoracic trauma. (A) Rightsided flail segment associated with ipsilateral pulmonary contusion,surgical emphysema and fractured clavicle. (B) The result of bluntthoracic trauma <strong>in</strong> a child caus<strong>in</strong>g bilateral pneumothoraces,pulmonary contusion and pneumopericardium; note the absence ofrib fractures. The possible mediast<strong>in</strong>al displacement to the leftsuggests that the undra<strong>in</strong>ed right sided pneumothorax is undertension.<strong>in</strong>juries is 30–33%, 16 17 with 30% of deaths related to <strong>in</strong>trathoracic<strong>in</strong>jury, 30% to extrathoracic <strong>in</strong>jury (most commonlyclosed head <strong>in</strong>jury), and 30% related to multiple organ failureand ARDS. 16 In a study of 144 consecutive patients with bluntchest trauma, the mortality rate for pulmonary contusionalone or flail chest alone was 16%, whereas the comb<strong>in</strong>ation ofpulmonary contusion and flail chest resulted <strong>in</strong> a mortalityrate of 42%. 17 Flail chest can lead to paradoxical ventilation,generat<strong>in</strong>g <strong>in</strong>efficient ventilation with progressive respiratorydistress and failure. <strong>Respiratory</strong> failure <strong>in</strong> flail chest may alsoresult from the <strong>in</strong>creased work of breath<strong>in</strong>g, pa<strong>in</strong>, compromisedcough, and atelectasis. A flail chest alone is not an <strong>in</strong>dicationfor mechanical ventilation if the patient has adequategas exchange. Thus, not all patients require tracheal <strong>in</strong>tubationbut rather alleviation of pa<strong>in</strong> so that effective tidalvolumes and vital capacities can be generated. 18However,patients with flail chest require mechanical ventilation more16 17frequently than those without flail segment. The use ofnon-<strong>in</strong>vasive positive pressure ventilation (NIPPV) <strong>in</strong> patientswith a flail segment and lesser degrees of pulmonarycontusion has also been described. 19 The decision to stabilise


Thoracic trauma, <strong>in</strong>halation <strong>in</strong>jury and post-pulmonary resection lung <strong>in</strong>jury <strong>in</strong> <strong>in</strong>tensive care 101the chest wall surgically <strong>in</strong> flail chest is based on theexperience and judgement of the surgeon. To date, the goal ofm<strong>in</strong>imis<strong>in</strong>g <strong>in</strong>tubation time by <strong>in</strong>ternal fixation of ribfractures has not been proven to be effective <strong>in</strong> a randomisedtrial. However, a patient who either fails to wean or manifestsa persistent chest wall deformity may benefit from surgicalmanagement. In addition, if thoracotomy is <strong>in</strong>dicated forother reasons <strong>in</strong> a patient with a flail segment, it would appearreasonable to take the opportunity to stabilise the ribfractures.Traditionally, the effect of the flail segment on pulmonarymechanics was thought to be the most significant aspect ofthe <strong>in</strong>jury. However, pa<strong>in</strong> is now recognised as the primaryfactor affect<strong>in</strong>g pulmonary function. 20 By enabl<strong>in</strong>g the patientto take adequate tidal volumes and cough effectively, relief ofpa<strong>in</strong> from rib fractures can prevent the development ofatelectasis, pneumonia, and sepsis, and obviate the need formechanical ventilation <strong>in</strong> patients with severe chesttrauma. 20 21 The standard of care has evolved from <strong>in</strong>tubationand mechanical ventilation for all patients to optimisation ofpa<strong>in</strong> control comb<strong>in</strong>ed with chest physiotherapy. 21Systemicanalgesics may be helpful <strong>in</strong> reliev<strong>in</strong>g pa<strong>in</strong> but detrimental tothe patient with marg<strong>in</strong>al pulmonary mechanics <strong>in</strong> whom anyreduction <strong>in</strong> respiratory drive may lead to hypoxia, atelectasis,and the need for mechanical ventilation. 22 The development ofregional techniques has revolutionised the management ofblunt thoracic trauma, provid<strong>in</strong>g a range of options foranalgesia that should be tailored to <strong>in</strong>dividual needs. 23 The useof epidural catheters for cont<strong>in</strong>uous adm<strong>in</strong>istration of opiatesor local anaesthetics is the preferred technique for pa<strong>in</strong> control<strong>in</strong> severe thoracic trauma and rarely produces any cl<strong>in</strong>icallysignificant alteration <strong>in</strong> pulmonary function. 24–27 Usefulalternatives are <strong>in</strong>tercostal nerve blocks and <strong>in</strong>trapleural localanaesthetic <strong>in</strong>fusions. 28 The primary disadvantage of <strong>in</strong>tercostalnerve blocks, however, is their relatively short duration ofaction (12.3 hours <strong>in</strong> one study 29 ), requir<strong>in</strong>g repeated<strong>in</strong>jections for adequate pa<strong>in</strong> control after trauma. Themechanism of action of <strong>in</strong>trapleural analgesia is unclear, butmay be the simultaneous blockade of multiple <strong>in</strong>tercostalnerves or nerve end<strong>in</strong>gs <strong>in</strong> the pleura. 30 A retrospective reviewof outcome factors among elderly patients after thoracictrauma showed clear improvement when an epidural catheterwas used for analgesia compared with systemic narcoticalone. 31There was a decreased <strong>in</strong>cidence of pulmonarycomplications <strong>in</strong> the epidural group, and the <strong>in</strong>cidence ofpneumonia, ARDS, and death was significantly lower <strong>in</strong> thesepatients. 31 In a further study <strong>in</strong> which the efficacy of epiduraland <strong>in</strong>trapleural catheters for pa<strong>in</strong> relief and improvements <strong>in</strong>pulmonary function was compared, cont<strong>in</strong>uous epiduralanalgesia was clearly superior. 32The only negative effect ofepidural analgesia consistently found <strong>in</strong> this study was hypotensionthat was easily corrected. 32Most patients with penetrat<strong>in</strong>g chest <strong>in</strong>juries can be treatednon-operatively, with only 10–15% requir<strong>in</strong>g surgery. 33 34 In aseries of 373 patients with penetrat<strong>in</strong>g lung <strong>in</strong>juries, chesttube <strong>in</strong>sertion was found to be the only treatment required <strong>in</strong>76% of patients and, of the rema<strong>in</strong><strong>in</strong>g 24% who underwentexploratory thoracotomy, half required pulmonary repair orresection. 35The majority of patients with mild to moderatechest wall <strong>in</strong>jury who survive the acute phase recover withlittle or no pulmonary disability. 36INHALATION INJURYEpidemiologyInhalation <strong>in</strong>jury can be thermal and/or chemical. Itaccompanies severe burns <strong>in</strong> up to 35% of those admitted toburn centres and accounts for 50–70% of burn relateddeaths. 37–39 The first comprehensive description of <strong>in</strong>halation<strong>in</strong>jury resulted from the 1942 Boston Coconut Grove fire <strong>in</strong>which 491 people died. 40 Furthermore, <strong>in</strong> the aftermath of the2001 World Trade Center terrorist attack <strong>in</strong> New York, <strong>in</strong>halation<strong>in</strong>jury was the most frequent reason that survivors soughtmedical attention. 41 Inhalation <strong>in</strong>jury predisposes burnpatients to pneumonia, respiratory failure, and death. 42 Mostburn patients who die have multiple organ failure 43 ; <strong>in</strong> onestudy mortality was 67% <strong>in</strong> patients with isolated pulmonaryfailure but 92% when pulmonary failure was complicated byfailure of at least one other system. 44 The <strong>in</strong>cidence of ARDScomplicat<strong>in</strong>g burns is 2–7%, 45but is significantly higher <strong>in</strong>patients with <strong>in</strong>halation <strong>in</strong>jury. 42In a review of 529 burnpatients admitted over a 4 year period, patients with<strong>in</strong>halation <strong>in</strong>jury had a 73% <strong>in</strong>cidence of respiratory failureand a 20% <strong>in</strong>cidence of ARDS. 42 Advances <strong>in</strong> the treatment ofburn victims have reduced mortality to 2–3% <strong>in</strong> those withoutrespiratory complications compared with 46% <strong>in</strong> those withconcomitant <strong>in</strong>halation <strong>in</strong>jury. 46Burn size is an important predictor of the development ofpulmonary complications. 47Smoke <strong>in</strong>halation with m<strong>in</strong>imalor no cutaneous burn is associated with chemical tracheobronchitisthat does not add significantly to the morbidity andmortality of thermal <strong>in</strong>jury. Patients with an <strong>in</strong>halation <strong>in</strong>juryand a medium sized burn often develop significant sequelae oftheir pulmonary <strong>in</strong>jury which can add up to 20% to theexpected mortality. 47The importance of <strong>in</strong>halation <strong>in</strong>jurywithout pulmonary complications is controversial. Whilesome have found that this does not add to mortality, 42 othershave reported that <strong>in</strong>halation <strong>in</strong>jury <strong>in</strong>creased mortality by60% when complicated by pneumonia, and by as much as 20%even <strong>in</strong> the absence of <strong>in</strong>fection. 47 It is possible, however, that<strong>in</strong> some patients without proven pneumonia, <strong>in</strong>halation<strong>in</strong>jury produced other complications that adversely affectedoutcome such as prolonged ventilator dependence, conf<strong>in</strong>ementto bed, and atelectasis.PathogenesisInhalation <strong>in</strong>juries can produce a wide spectrum of cl<strong>in</strong>icaleffects. Smoke <strong>in</strong>halation is a particularly challeng<strong>in</strong>g cl<strong>in</strong>icalproblem because, apart from thermal <strong>in</strong>jury, patients are oftenexposed to a large number of <strong>in</strong>haled tox<strong>in</strong>s such as pyrolysisproducts of plastics and other chemicals. 48 Death from smoke<strong>in</strong>halation may be caused by direct pulmonary <strong>in</strong>jury orasphyxia caused by oxygen deprivation or carbon monoxideexposure. Simple asphyxia may occur dur<strong>in</strong>g a fire because theambient oxygen level <strong>in</strong> a burn<strong>in</strong>g room may fall to less than10%. Older <strong>in</strong>dividuals and those with pre-exist<strong>in</strong>g lungdiseases may be more susceptible to the effects of <strong>in</strong>halation<strong>in</strong>jury. 49 In <strong>in</strong>dividuals with no past history of airway disease,the emergence of significant bronchial hyperreactivity after<strong>in</strong>halation <strong>in</strong>jury from irritat<strong>in</strong>g agents such as chlor<strong>in</strong>e gas 5051 52is known as reactive airways disease syndrome (RADS).Irritant gases may produce a variety of cl<strong>in</strong>ical problems,<strong>in</strong>clud<strong>in</strong>g upper airway mucosal irritation and <strong>in</strong>flammation,laryngospasm, bronchoconstriction, atelectasis, and ARDS.Water solubility plays a key role <strong>in</strong> determ<strong>in</strong><strong>in</strong>g where <strong>in</strong>haledgases deposit <strong>in</strong> the respiratory tract. Because the respiratorytract is l<strong>in</strong>ed with mucus, gases that are highly water solublesuch as ammonia, sulphur dioxide, and hydrogen chloridegenerally cause acute irritant <strong>in</strong>jury to mucous membranes,<strong>in</strong>clud<strong>in</strong>g the eyes and the l<strong>in</strong><strong>in</strong>g of the nose and upper airway,and spare the lower respiratory tract. However, high solubilitygases are also capable of caus<strong>in</strong>g lower tract <strong>in</strong>jury atsufficiently high doses. Less soluble gases such as phosgene,ozone, and nitrogen oxides often have no effect on the upperairway, but they penetrate <strong>in</strong>to the lower airway and irritatethe small airways and gas exchange surface. 53 Gases of <strong>in</strong>termediatesolubility such as chlor<strong>in</strong>e may exert irritant effectswidely throughout the respiratory tract. Furthermore, theparticle size of <strong>in</strong>haled substances determ<strong>in</strong>es the site andnature of the <strong>in</strong>jury. Most particles that are smaller than100 µm can enter the airway. Particles with a diameter of


102 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>


Thoracic trauma, <strong>in</strong>halation <strong>in</strong>jury and post-pulmonary resection lung <strong>in</strong>jury <strong>in</strong> <strong>in</strong>tensive care 103Ischaemia-reperfusion <strong>in</strong>juryAtelectotraumaSurgical traumaVolutraumaOxygen toxicityHyperperfusionAcute lung <strong>in</strong>juryFigure 15.4 Both the lungs suffer damage that may contribute toacute lung <strong>in</strong>jury dur<strong>in</strong>g one lung ventilation and resection.Figure 15.3 Radiographic features of acute lung <strong>in</strong>jury follow<strong>in</strong>g aright sided pneumonectomy. The CT scan shows dense dependentcollapse and consolidation with overly<strong>in</strong>g patchy consolidation andground glass opacification.surgery and the development of ALI/ARDS. 72 78 However, menover the age of 60 years, especially when undergo<strong>in</strong>g lungresection for lung cancer, form a high risk group. 73 Furthermore,no correlation has been found between the side ofresection and the development of ALI/ARDS, but the risk73 75 79<strong>in</strong>creases progressively with more extensive resections.73 77ALI may present up to 7 days after surgery, but most75 76patients present between 1 and 3 days postoperatively.Excessive perioperative adm<strong>in</strong>istration of crystalloid may precipitaterespiratory failure, 74 77 although recently the perceived75 76role of fluid overload has dim<strong>in</strong>ished. The high prote<strong>in</strong>content of the alveolar oedema fluid and the frequent delay <strong>in</strong>presentation suggest that perioperative fluid overload is notthe primary cause of post-pulmonary resection lung <strong>in</strong>jury.The differential diagnosis <strong>in</strong>cludes lower respiratory tract<strong>in</strong>fection and cardiogenic pulmonary oedema (fig 15.3]). Bothcan be <strong>in</strong>vestigated <strong>in</strong> the <strong>in</strong>tubated patient by tracheal aspirationor bronchoscope guided sampl<strong>in</strong>g and pulmonary arterialcatheterisation.PathogenesisDur<strong>in</strong>g pulmonary resection one lung is mechanicallyventilated while clamp<strong>in</strong>g one lumen of a double lumenendotracheal tube collapses the lung be<strong>in</strong>g operated on.Repeated collapse and re<strong>in</strong>flation occur dur<strong>in</strong>g the course ofthoracotomy as the surgeon carries out the many steps <strong>in</strong> theoperation. These cycles cause ischaemia-reperfusion (IR)<strong>in</strong>jury that are likely to resemble the <strong>in</strong>sult suffered by a80 81transplanted lung. IR <strong>in</strong>jury comb<strong>in</strong>ed with the oxygentoxicity required to counterbalance the effects of shunt <strong>in</strong> thedeflated lung <strong>in</strong>duce the formation of reactive oxygen species(ROS) and reactive nitrogen species (RNS). Similarly, the gasexchange surface <strong>in</strong> the ventilated lung may be damaged byhyperperfusion, hyperoxia, and overdistension. Cl<strong>in</strong>ical observationssuggest that <strong>in</strong>jury to the non-operated side may bemore important, as shown by a significant <strong>in</strong>crease <strong>in</strong> densityon the CT scan of the non-operated lung <strong>in</strong> eight out of n<strong>in</strong>epatients follow<strong>in</strong>g pulmonary resection. 82Conversely, whenthe other lung that has been subjected to surgical trauma isre-expanded, IR leads to neutrophil recruitment andactivation 80 83 84 and the formation of ROS, 85–87 and may causesystemic effects through the return of toxic metabolites to thecirculation (fig 15.4). 88Under normal circumstances endogenous antioxidantsclosely regulate redox balance. However, if these defencemechanisms become overwhelmed, high levels of ROS/RNSmay directly damage lipids, prote<strong>in</strong>s and DNA, while lowerlevels affect signal transduction caus<strong>in</strong>g a change <strong>in</strong> cellbehaviour without necessarily damag<strong>in</strong>g or kill<strong>in</strong>g the cell.ROS have been implicated <strong>in</strong> the onset and progression ofARDS, both <strong>in</strong> animal models and cl<strong>in</strong>ical studies, <strong>in</strong> whichmarkers of oxidative damage have been identified. 89 90 Iron is acatalyst for hydroxyl radical formation, and <strong>in</strong> patients withARDS aberrant iron metabolism has been identified. Levels ofchelatable redox active iron <strong>in</strong> bronchoalveolar lavage (BAL)fluid are greater <strong>in</strong> survivors of ARDS than <strong>in</strong> non-survivors. 91Paradoxically, non-survivors had <strong>in</strong>creased levels of transferr<strong>in</strong>and iron b<strong>in</strong>d<strong>in</strong>g antioxidant activity. 92 After lobectomy the<strong>in</strong>crease <strong>in</strong> hydrogen peroxide <strong>in</strong> exhaled breath condensatewas greater than <strong>in</strong> breath from patients follow<strong>in</strong>gpneumonectomy. 93 This may be expla<strong>in</strong>ed by the <strong>in</strong>creased tissuehandl<strong>in</strong>g and dissection <strong>in</strong>volved <strong>in</strong> do<strong>in</strong>g a lobectomycompared with a pneumonectomy. Others have reportedchanges <strong>in</strong> several markers <strong>in</strong> the plasma <strong>in</strong>dicat<strong>in</strong>g oxidativestress follow<strong>in</strong>g lung resection (prote<strong>in</strong> thiol, prote<strong>in</strong> carbonyl,and myeloperoxidase levels), but no differences were observedbetween patients undergo<strong>in</strong>g lobectomy and those undergo<strong>in</strong>gpneumonectomy. 94Activated neutrophils <strong>in</strong> the lungs of patients with ALI produceROS. 95–97 However, neutrophil depletion does not attenuateexperimental IR mediated lung <strong>in</strong>jury. 80Follow<strong>in</strong>g IR <strong>in</strong>the isolated perfused rat lung model, non-leucocyte derivedROS appear before neutrophil-endothelial cell adhesion andactivation occur. 98In humans an <strong>in</strong>crease <strong>in</strong> pulmonaryvascular permeability persists throughout the course of ARDSand correlates with the severity of lung <strong>in</strong>jury and the neutrophilcontent of the BAL fluid. 99 Studies <strong>in</strong> rodents have looked


104 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>at the effects of IR on hypoxic pulmonary vasoconstrictionus<strong>in</strong>g album<strong>in</strong> escape as a marker of endothelial <strong>in</strong>tegrity. 100The <strong>in</strong>tegrity of the endothelium was ma<strong>in</strong>ta<strong>in</strong>ed for up to 30m<strong>in</strong>utes of ischaemia alone. If, however, a shorter period ofischaemia was followed by reperfusion, the tendency foroedema formation was dramatically <strong>in</strong>creased. 100 This suggeststhat short periods of ischaemia followed by reperfusion maycause as much damage as much longer periods of ischaemiaalone.CONCLUSIONThoracic trauma and <strong>in</strong>halation <strong>in</strong>jury are common causes ofrespiratory failure and critical illness often <strong>in</strong> young patientswho have a good prognosis if the orig<strong>in</strong>al <strong>in</strong>sult is survivable.Blunt thoracic trauma can produce a spectrum of <strong>in</strong>juries<strong>in</strong>clud<strong>in</strong>g rib fractures, pneumothorax, flail segments andpulmonary contusion, all of which can impair pulmonaryfunction. Inhalation <strong>in</strong>jury can be both thermal and/orchemical, and predisposes burn patients to pneumonia, respiratoryfailure, and death. ALI/ARDS is the most commoncause of death follow<strong>in</strong>g pulmonary resection, the pathogenesisof which results from IR <strong>in</strong>jury. The mortality rate rema<strong>in</strong>shigh despite advances <strong>in</strong> supportive techniques.Despite the disparate nature of these conditions, commonthemes emerge <strong>in</strong> the supportive management of suchpatients. For example, atelectasis and sputum retention followall major pulmonary trauma, <strong>in</strong>clud<strong>in</strong>g surgery, as a consequenceof impaired cough and shallow respiration caused bypa<strong>in</strong> and weakness. The tendency to develop respiratoryfailure and pneumonia may be m<strong>in</strong>imised by adequate pa<strong>in</strong>relief, mobilisation, and physiotherapy. Intermittent non<strong>in</strong>vasiveventilatory support and tracheal aspiration facilitatedby a nasopharyngeal airway or a m<strong>in</strong>i-tracheostomy may berequired to help a patient with an <strong>in</strong>adequate cough. Theseconditions are best managed by a multidiscipl<strong>in</strong>ary approach.REFERENCES1 Gaillard M, Herve C, Mand<strong>in</strong> L, et al. Mortality prognostic factors <strong>in</strong>chest <strong>in</strong>jury. J Trauma 1990;30:93–6.2 Lee RB, Bass SM, Morris JA Jr, et al. Three or more rib fractures as an<strong>in</strong>dicator for transfer to a level I trauma center: a population-based study.J Trauma 1990;30:689–94.3 Tom<strong>in</strong>aga GT, Waxman K, Scannell G, et al. Emergency thoracotomywith lung resection follow<strong>in</strong>g trauma. Am Surg 1993;59:834–7.4 Webb WR. Thoracic trauma. Surg Cl<strong>in</strong> North Am 1974;54:1179–92.5 Bass TL, Miller PK, Campbell DB, et al. Traumatic adult respiratorydistress syndrome. Surg Cl<strong>in</strong> North Am 1997;7:429–42.6 Ashbaugh DG, Bigelow DB, Petty TL, Lev<strong>in</strong>e BE. Acute respiratorydistress <strong>in</strong> adults. Lancet 1967;ii:319–23.7 Russell GB, Campbell DB. Thoracic trauma and the adult respiratorydistress syndrome. Sem<strong>in</strong> Thorac Cardiovasc Surg 1992;4:241–6.8 Maunder RJ, Hudson LD. Adult respiratory distress syndrome. NewYork: Marcel Dekker 1991: 1–21.9 Chopra PS, Kroncke GM, Berkoff HA, et al. Pulmonary contusion aproblem <strong>in</strong> blunt chest trauma. Wiscons<strong>in</strong> Med J 1977;76:S1–3.10 DeMuth WE, Smith JM. Pulmonary contusion. Am J Surg1965;109:819.11 Richardson JD, Adams L, Fl<strong>in</strong>t LM. Selective management of flail chestand pulmonary contusion. Ann Surg 1982;196:481–7.12 Majeski JA. <strong>Management</strong> of flail chest after blunt trauma. South Med J1981;74:848–9.13 Saada M, Goar<strong>in</strong> JP, Riou B, et al. Systemic gas embolism complicat<strong>in</strong>gpulmonary contusion. Diagnosis and management us<strong>in</strong>g transesophagealechocardiography. Am J Respir Crit <strong>Care</strong> Med 1995;152:812–5.14 American College of Surgeons. Advanced trauma life supportprogram for doctors. Chicago, IL: American College of Surgeons, 1997.15 Freedland M, Wilson RF, Bender JS, et al. The management of flail chest<strong>in</strong>jury: factors affect<strong>in</strong>g outcome. J Trauma 1990;30:1460–8.16 Ciraulo DL, Elliott D, Mitchell KA, et al. Flail chest as a marker forsignificant <strong>in</strong>juries. J Am Coll Surg 1994;178:466–70.17 Clark GC, Schecter WP, Trunkey DD. Variables affect<strong>in</strong>g outcome <strong>in</strong>blunt chest trauma: flail chest vs. pulmonary contusion. J Trauma1988;28:298–304.18 Cicala RS, Voeller GR, Fox T, et al. Epidural analgesia <strong>in</strong> thoracictrauma: effects of lumbar morph<strong>in</strong>e and thoracic bupivaca<strong>in</strong>e onpulmonary function. Crit <strong>Care</strong> Med 1990;18:229–31.19 Garfield MJ, Howard-Griff<strong>in</strong> RM. Non-<strong>in</strong>vasive positive pressureventilation for severe thoracic trauma. Br J Anaesth 2000;85:788–90.20 Rank<strong>in</strong> AP, Comber RE. <strong>Management</strong> of fifty cases of chest <strong>in</strong>jury with aregimen of epidural bupivaca<strong>in</strong>e and morph<strong>in</strong>e. Anaesth Intensive <strong>Care</strong>1984;12:311–4.21 Tr<strong>in</strong>kle JK, Richardson JD, Franz JL, et al. <strong>Management</strong> of flail chestwithout mechanical ventilation. Ann Thorac Surg 1975;19:355–63.22 Egbert LD, Bendixen HH. Effect of morph<strong>in</strong>e on breath<strong>in</strong>g pattern: apossible factor <strong>in</strong> atelectasis. JAMA 1964;188:485–8.23 Karmakar MK, Ho AM. Acute pa<strong>in</strong> management of patients withmultiple fractured ribs. J Trauma 2003;54:615–25.24 Dittmann M, Steenblock U, Kranzl<strong>in</strong> M, et al. Epidural analgesia ormechanical ventilation for multiple rib fractures? Intensive <strong>Care</strong> Med1982;8:89–92.25 Johnston JR, McCaughey W. Epidural morph<strong>in</strong>e. A method ofmanagement of multiple fractured ribs. Anaesthesia 1980;35:155–7.26 Ullman DA, Fortune JB, Greenhouse BB, et al. The treatment of patientswith multiple rib fractures us<strong>in</strong>g cont<strong>in</strong>uous thoracic epidural narcotic<strong>in</strong>fusion. Reg Anesth 1989;14:43–7.27 Rawal N, Sjostrand UH, Dahlstrom B, et al. Epidural morph<strong>in</strong>e forpostoperative pa<strong>in</strong> relief: a comparative study with <strong>in</strong>tramuscular narcoticand <strong>in</strong>tercostal nerve block. Anesth Analg 1982;61:93–8.28 Haenel JB, Moore FA, Moore EE, et al. Extrapleural bupivaca<strong>in</strong>e foramelioration of multiple rib fracture pa<strong>in</strong>. J Trauma 1995;38:22–7.29 Nunn JF, Slav<strong>in</strong> G. Posterior <strong>in</strong>tercostal nerve block for pa<strong>in</strong> relief aftercholecystectomy. Anatomical basis and efficacy. Br J Anaesth1980;52:253–60.30 Shafei H, Chamberla<strong>in</strong> M, Natrajan KN, et al. Intrapleural bupivaca<strong>in</strong>efor early post-thoracotomy analgesia: comparison with bupivaca<strong>in</strong>e<strong>in</strong>tercostal block and cryofreez<strong>in</strong>g. Thorac Cardiovasc Surg1990;38:38–41.31 Wisner DH. A stepwise logistic regression analysis of factors affect<strong>in</strong>gmorbidity and mortality after thoracic trauma: effect of epiduralanalgesia. J Trauma 1990;30:799–804.32 Luchette FA, Radafshar SM, Kaiser R, et al. Prospective evaluation ofepidural versus <strong>in</strong>trapleural catheters for analgesia <strong>in</strong> chest wall trauma. JTrauma 1994;36:865–9.33 Siemens R, Polk HC Jr, Gray LA Jr, et al. Indications for thoracotomyfollow<strong>in</strong>g penetrat<strong>in</strong>g thoracic <strong>in</strong>jury. J Trauma 1977;17:493–500.34 McSwa<strong>in</strong> NE Jr. Blunt and penetrat<strong>in</strong>g chest <strong>in</strong>juries. World J Surg1992;16:924–9.35 Graham JM, Mattox KL, Beall AC Jr. Penetrat<strong>in</strong>g trauma of the lung. JTrauma 1979;19:665–9.36 Landercasper J, Cogbill TH, L<strong>in</strong>desmith LA. Long-term disability afterflail chest <strong>in</strong>jury. J Trauma 1984;24:410–4.37 Herndon DN, Langner F, Thompson P, et al. Pulmonary <strong>in</strong>jury <strong>in</strong> burnedpatients. Surg Cl<strong>in</strong> North Am 1987;67:31–46.38 Herndon DN, Barrow RE, L<strong>in</strong>ares HA, et al. Inhalation <strong>in</strong>jury <strong>in</strong> burnedpatients: effects and treatment. Burns Incl Therm Inj 1988;14:349–56.39 Zawacki BE, Jung RC, Joyce J, et al. Smoke, burns, and the naturalhistory of <strong>in</strong>halation <strong>in</strong>jury <strong>in</strong> fire victims: a correlation of experimentaland cl<strong>in</strong>ical data. Ann Surg 1977;185:100–10.40 Saffle JR. The 1942 fire at Boston’s Coconut Grove nightclub. Am J Surg1993;166:581–91.41 Centers for Disease Control and Prevention. Rapid assessment of<strong>in</strong>juries among survivors of the terrorist attack on the World TradeCenter—New York City, September 2001. JAMA 2002;287:835–8.42 Holl<strong>in</strong>gsed TC, Saffle JR, Barton RG, et al. Etiology and consequencesof respiratory failure <strong>in</strong> thermally <strong>in</strong>jured patients. Am J Surg1993;166:592–6.43 Hamit HF. Factors associated with deaths of burned patients <strong>in</strong> acommunity hospital. J Trauma 1978;18:405–18.44 Marshall WG Jr, Dimick AR. The natural history of major burns withmultiple subsystem failure. J Trauma 1983;23:102–5.45 Fowler AA, Hamman RF, Zerbe GO, et al. Adult respiratory distresssyndrome. Prognosis after onset. Am Rev Respir Dis 1985;132:472–8.46 Haponik EF, Crapo RO, Herndon DN, et al. Smoke <strong>in</strong>halation. Am RevRespir Dis 1988;138:1060–3.47 Shirani KZ, Pruitt BA Jr, Mason AD Jr. The <strong>in</strong>fluence of <strong>in</strong>halation <strong>in</strong>juryand pneumonia on burn mortality. Ann Surg 1987;205:82–7.48 Kimmel EC, Still KR. Acute lung <strong>in</strong>jury, acute respiratory distresssyndrome and <strong>in</strong>halation <strong>in</strong>jury: an overview. Drug Chem Toxicol1999;22:91–128.49 Rajpura A. The epidemiology of burns and smoke <strong>in</strong>halation <strong>in</strong>secondary care: a population-based study cover<strong>in</strong>g Lancashire and SouthCumbria. Burns 2002;28:121–30.50 Leroyer C, Malo JL, Girard D, et al. Chronic rh<strong>in</strong>itis <strong>in</strong> workers at risk ofreactive airways dysfunction syndrome due to exposure to chlor<strong>in</strong>e.Occup Environ Med 1999;56:334–8.51 Alberts WM, Brooks SM. Reactive airways dysfunction syndrome. CurrOp<strong>in</strong> Pulm Med 1996;2:104–10.52 Bardana EJ Jr. Reactive airways dysfunction syndrome (RADS):guidel<strong>in</strong>es for diagnosis and treatment and <strong>in</strong>sight <strong>in</strong>to likely prognosis.Ann Allergy Asthma Immunol 1999;83:583–6.53 Schwartz DA. Acute <strong>in</strong>halational <strong>in</strong>jury. Occup Med 1987;2:297–318.54 LaLonde C, Nayak U, Hennigan J, et al. Plasma catalase andglutathione levels are decreased <strong>in</strong> response to <strong>in</strong>halation <strong>in</strong>jury. J Burn<strong>Care</strong> Rehabil 1997;18:515–9.55 Guha SC, Herndon DN, Evans MJ, et al. Is the CD18 adhesion complexof polymorphonuclear leukocytes <strong>in</strong>volved <strong>in</strong> smoke-<strong>in</strong>duced lungdamage? A morphometric study. J Burn <strong>Care</strong> Rehabil 1993;14:503–11.56 Isago T, Noshima S, Traber LD, et al. Analysis of pulmonarymicrovascular permeability after smoke <strong>in</strong>halation. J Appl Physiol1991;71:1403–8.


Thoracic trauma, <strong>in</strong>halation <strong>in</strong>jury and post-pulmonary resection lung <strong>in</strong>jury <strong>in</strong> <strong>in</strong>tensive care 10557 Ischiropoulos H, Mendiguren I, Fisher D, et al. Role of neutrophils andnitric oxide <strong>in</strong> lung alveolar <strong>in</strong>jury from smoke <strong>in</strong>halation. Am J RespirCrit <strong>Care</strong> Med 1994;150:337–41.58 Wright JL. Inhalational lung <strong>in</strong>jury caus<strong>in</strong>g bronchiolitis. Cl<strong>in</strong> Chest Med1993;14:635–44.59 Hunt JL, Agee RN, Pruitt BA Jr. Fiberoptic bronchoscopy <strong>in</strong> acute<strong>in</strong>halation <strong>in</strong>jury. J Trauma 1975;15:641–49.60 Nakae H, Tanaka H, Inaba H. Failure to clear casts and secretionsfollow<strong>in</strong>g <strong>in</strong>halation <strong>in</strong>jury can be dangerous: report of a case. Burns2001;27:189–91.61 Agee RN, Long JM 3rd, Hunt JL, et al. Use of 133-xenon <strong>in</strong> earlydiagnosis of <strong>in</strong>halation <strong>in</strong>jury. J Trauma 1976;16:218–24.62 Zikria BA, Weston GC, Chodoff M, et al. Smoke and carbon monoxidepoison<strong>in</strong>g <strong>in</strong> fire victims. J Trauma 1972;12:641–5.63 Grube BJ, Marv<strong>in</strong> JA, Heimbach DM. Therapeutic hyperbaric oxygen:help or h<strong>in</strong>drance <strong>in</strong> burn patients with carbon monoxide poison<strong>in</strong>g? JBurn <strong>Care</strong> Rehabil 1988;9:249–52.64 Cox CS Jr, Zwischenberger JB, Traber DL, et al. Hepar<strong>in</strong> improvesoxygenation and m<strong>in</strong>imizes barotrauma after severe smoke <strong>in</strong>halation <strong>in</strong>an ov<strong>in</strong>e model. Surg Gynecol Obstet 1993;176:339–49.65 Tasaki O, Moz<strong>in</strong>go DW, Dubick MA, et al. Effects of hepar<strong>in</strong> andlisofyll<strong>in</strong>e on pulmonary function after smoke <strong>in</strong>halation <strong>in</strong>jury <strong>in</strong> an ov<strong>in</strong>emodel. Crit <strong>Care</strong> Med 2002;30:637–43.66 Tanaka H, Matsuda T, Miyagantani Y, et al. Reduction of resuscitationfluid volumes <strong>in</strong> severely burned patients us<strong>in</strong>g ascorbic acidadm<strong>in</strong>istration: a randomized, prospective study. Arch Surg2000;135:326–31.67 Nieman GF, Clark WR, Hakim T. Methylprednisolone does not protectthe lung from <strong>in</strong>halation <strong>in</strong>jury. Burns 1991;17:384–90.68 Bernard GR, Artigas A, Brigham KL, et al. The American-EuropeanConsensus Conference on ARDS. Def<strong>in</strong>itions, mechanisms, relevantoutcomes, and cl<strong>in</strong>ical trial coord<strong>in</strong>ation. Am J Respir Crit <strong>Care</strong> Med1994;149:818–24.69 Williams EA, Evans TW, Goldstraw P. Acute lung <strong>in</strong>jury follow<strong>in</strong>g lungresection: is one lung anaesthesia to blame? Thorax 1996;51:114–6.70 Romano PS, Mark DH. Patient and hospital characteristics related to<strong>in</strong>-hospital mortality after lung cancer resection. Chest 1992;101:1332–7.71 Wada H, Nakamura T, Nakamoto K, et al. Thirty-day operative mortalityfor thoracotomy <strong>in</strong> lung cancer. J Thorac Cardiovasc Surg1998;115:70–3.72 Ruff<strong>in</strong>i E, Parola A, Papalia E, et al. Frequency and mortality of acutelung <strong>in</strong>jury and acute respiratory distress syndrome after pulmonaryresection for bronchogenic carc<strong>in</strong>oma. Eur J Cardiothorac Surg2001;20:30–6.73 Kutlu CA, Williams EA, Evans TW, et al. Acute lung <strong>in</strong>jury and acuterespiratory distress syndrome after pulmonary resection. Ann Thorac Surg2000;69:376–80.74 Zeld<strong>in</strong> RA, Normand<strong>in</strong> D, Landtw<strong>in</strong>g D, et al. Postpneumonectomypulmonary edema. J Thorac Cardiovasc Surg 1984;87:359–65.75 Turnage WS, Lunn JJ. Postpneumonectomy pulmonary edema. Aretrospective analysis of associated variables. Chest 1993;103:1646–50.76 Waller DA, Gebitek<strong>in</strong> C, Saunders NR, et al. Noncardiogenicpulmonary edema complicat<strong>in</strong>g lung resection. Ann Thorac Surg1993;55:140–3.77 Mathru M, Blakeman B, Dries DJ, et al. Permeability pulmonary edemafollow<strong>in</strong>g lung resection. Chest 1990;98:1216–8.78 Hayes JP, Williams EA, Goldstraw P, et al. Lung <strong>in</strong>jury <strong>in</strong> patientsfollow<strong>in</strong>g thoracotomy. Thorax 1995;50:990–1.79 Keagy BA, Lores ME, Starek PJ, et al. Elective pulmonary lobectomy:factors associated with morbidity and operative mortality. Ann ThoracSurg 1985;40:349–52.80 Lu YT, Hellewell PG, Evans TW. Ischemia-reperfusion lung <strong>in</strong>jury:contribution of ischemia, neutrophils, and hydrostatic pressure. Am JPhysiol 1997;273:L46–54.81 De Perrot M, Liu M, Waddell TK, et al. Ischaemia-reperfusion-<strong>in</strong>ducedlung <strong>in</strong>jury. Am J Respir Crit <strong>Care</strong> Med 2003;167:490–511.82 Padley SP, Jordan SJ, Goldstraw P, et al. Asymmetric ARDS follow<strong>in</strong>gpulmonary resection: CT f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong>itial observations. Radiology2002;223:468–73.83 Anderson BO, Moore EE, Moore FA, et al. Hypovolemic shockpromotes neutrophil sequestration <strong>in</strong> lungs by a xanth<strong>in</strong>e oxidase-relatedmechanism. J Appl Physiol 1991;71:1862–5.84 Terada LS, Dormish JJ, Shanley PF, et al. Circulat<strong>in</strong>g xanth<strong>in</strong>e oxidasemediates lung neutrophil sequestration after <strong>in</strong>test<strong>in</strong>alischemia-reperfusion. Am J Physiol 1992;263:L394–401.85 McCord JM. Oxygen-derived radicals: a l<strong>in</strong>k between reperfusion <strong>in</strong>juryand <strong>in</strong>flammation. Fed Proc 1987;46:2402–6.86 Adkison D, Hollwarth ME, Benoit JN, et al. Role of free radicals <strong>in</strong>ischemia-reperfusion <strong>in</strong>jury to the liver. Acta Physiol Scand Suppl1986;548:101–7.87 Williams EA, Qu<strong>in</strong>lan GJ, Ann<strong>in</strong>g PB, et al. Lung <strong>in</strong>jury follow<strong>in</strong>gpulmonary resection <strong>in</strong> the isolated, blood-perfused rat lung. Eur Respir J1999;14:745–50.88 Parks DA, Granger DN. Contributions of ischemia and reperfusion tomucosal lesion formation. Am J Physiol 1986;250:G749–53.89 Pittet JF, Mackersie RC, Mart<strong>in</strong> TR, et al. Biological markers of acutelung <strong>in</strong>jury: prognostic and pathogenetic significance. Am J Respir Crit<strong>Care</strong> Med 1997;155:1187–205.90 Metnitz PG, Bartens C, Fischer M, et al. Antioxidant status <strong>in</strong> patientswith acute respiratory distress syndrome. Intensive <strong>Care</strong> Med1999;25:180–5.91 Gutteridge JM, Mumby S, Qu<strong>in</strong>lan GJ, et al. Pro-oxidant iron is present<strong>in</strong> human pulmonary epithelial l<strong>in</strong><strong>in</strong>g fluid: implications for oxidativestress <strong>in</strong> the lung. Biochem Biophys Res Commun 1996;220:1024–7.92 Gutteridge GMC, Qu<strong>in</strong>lan G, Evans TW. Primary plasma antioxidantsare secreted <strong>in</strong> patients with adult respiratory distress syndrome. J LabCl<strong>in</strong> Med 1994;124:263–73.93 Lases EC, Duurkens VA, Gerritsen WB, et al. Oxidative stress after lungresection therapy: a pilot study. Chest 2000;117:999–1003.94 Williams EA, Qu<strong>in</strong>lan GJ, Goldstraw P, et al. Postoperative lung <strong>in</strong>juryand oxidative damage <strong>in</strong> patients undergo<strong>in</strong>g pulmonary resection. EurRespir J 1998;11:1028–34.95 Tate RM, Rep<strong>in</strong>e JE. Neutrophils and the adult respiratory distresssyndrome. Am Rev Respir Dis 1983;128:552–9.96 Lamb NJ, Gutteridge JM, Baker C, et al. Oxidative damage to prote<strong>in</strong>sof bronchoalveolar lavage fluid <strong>in</strong> patients with acute respiratory distresssyndrome: evidence for neutrophil-mediated hydroxylation, nitration, andchlor<strong>in</strong>ation. Crit <strong>Care</strong> Med 1999;27:1738–44.97 Grace PA. Ischaemia-reperfusion <strong>in</strong>jury. Br J Surg 1994;81:637–47.98 Seibert AF, Haynes J, Taylor A. Ischemia-reperfusion <strong>in</strong>jury <strong>in</strong> theisolated rat lung. Role of flow and endogenous leukocytes. Am Rev RespirDis 1993;147:270–5.99 S<strong>in</strong>clair DG, Braude S, Haslam PL, et al. Pulmonary endothelialpermeability <strong>in</strong> patients with severe lung <strong>in</strong>jury. Cl<strong>in</strong>ical correlates andnatural history. Chest 1994;106:535–9.100 Messent M, Griffiths MJ, Evans TW. Pulmonary vascular reactivity andischaemia-reperfusion <strong>in</strong>jury <strong>in</strong> the rat. Cl<strong>in</strong> Sci 1993;85:71–5.


16 Illustrative case 1: cystic fibrosisS R Thomas.............................................................................................................................CASE REPORTA 26 year old man was admitted to the <strong>in</strong>tensivecare unit (ICU) on two occasions. Cystic fibrosis(CF) had been diagnosed at 2 months when hewas fail<strong>in</strong>g to thrive and he was subsequentlyfound to be homozygous for the ∆F508 mutation.At 19 years of age his respiratory tract secretionswere colonised by Burkholderia cepacia. He hadrecurrent pneumothoraces requir<strong>in</strong>g two surgicalpleurodeses. Immediately before his ICU admissionshis forced expiratory volume <strong>in</strong> 1 second(FEV 1) was 31% predicted. He had pancreatic<strong>in</strong>sufficiency at the time of diagnosis anddeveloped diabetes at the age of 23. He also hadCF related liver disease, portal hypertension, andoesophageal varices.He was admitted to the CF centre with ahistory of <strong>in</strong>creased breathlessness and sputumproduction with haemoptysis. Intravenous antibioticswere commenced. Staphylococcus aureus andB cepacia were cultured <strong>in</strong> the sputum. On thenight after admission he had a massive haematemesisrequir<strong>in</strong>g resuscitation, endotracheal <strong>in</strong>tubation,and transfer to the ICU. An emergencyendoscopy identified bleed<strong>in</strong>g oesophagealvarices that were banded. Bronchoscopic exam<strong>in</strong>ationshowed aspirated blood throughout thebronchial tree. He was successfully weaned frommechanical ventilation and transferred back to ahigh dependency area. The subsequent <strong>in</strong>patientstay was complicated by decompensation of hisliver disease and ascites. He also developed B cepaciasepticaemia and was eventually allowed to gohome almost 1 month after discharge from theICU.Three weeks after discharge from the CF centrehe presented to his local hospital with haematemesis.After a further episode of haemorrhage, anendotracheal tube and subsequently a Sengstaken-Blakemore tube were <strong>in</strong>serted. Endoscopy thefollow<strong>in</strong>g day did not suggest variceal haemorrhage,but a bronchoscopy showed blood predom<strong>in</strong>antly<strong>in</strong> the right bronchial tree. He required amassive blood transfusion and was transferredback to the ICU at the CF centre. Bronchial angiographydemonstrated abnormal bronchial arteries<strong>in</strong> the right upper lobe and some abnormal vessels<strong>in</strong> the left lower lobe, which were embolised. Aftera failed extubation, a percutaneous tracheostomywas <strong>in</strong>serted. He cont<strong>in</strong>ued to have episodes ofhaemoptysis and developed worsen<strong>in</strong>g ascites thatcompromised diaphragmatic function. The ascitesdid not improve with conservative managementand was dra<strong>in</strong>ed on two occasions. Hypernatraemiadeveloped probably as a result of a sodiumconta<strong>in</strong><strong>in</strong>g elemental feed, <strong>in</strong>travenous antibiotics,and hyperaldosteronism associated with liver disease.A change to a low sodium non-elemental feed<strong>in</strong> comb<strong>in</strong>ation with pancreatic enzymes correctedthe hypernatraemia. He subsequently developed Bcepacia septicaemia and the <strong>in</strong>travenous antibioticswere changed based on new sensitivities. Gradualwean<strong>in</strong>g from pressure support ventilation wasachieved and he was breath<strong>in</strong>g spontaneously via atracheostomy 25 days after admission. He wastransferred to the respiratory ward on day 28 anddischarged home 18 days later. He died suddenlyafter only 12 days at home follow<strong>in</strong>g what wasthought to be a massive haematemesis.MANAGEMENT OF CF PATIENTS IN THEICUMost critically ill patients with CF have end stagedisease and <strong>in</strong>tensive care is not considered; however,when the possibility of <strong>in</strong>tensive care arisesfor <strong>in</strong>dividual patients it is often difficult to determ<strong>in</strong>ewhether this is appropriate. When a patientwith CF is admitted to the ICU a number of specialistissues arise—such as the management ofhaemoptysis, the treatment of <strong>in</strong>fective exacerbations,and nutritional support—which will bediscussed below. Close liaison between the CF andICU teams is required. Non-<strong>in</strong>vasive ventilation ofpatients with CF has been found to be highlyeffective, but does not usually occur <strong>in</strong> the ICU <strong>in</strong>the UK and will not be discussed extensively here.Select<strong>in</strong>g CF patients for ICU careThe selection of patients with CF who are suitablefor admission to the ICU is not easy, partlybecause there are relatively few published data onwhich to base the decision. One retrospectivemulticentre study published <strong>in</strong> 1978 exam<strong>in</strong>edthe outcomes of 46 patients who developed respiratoryfailure between the ages of 1 month and 32years 1 ; 21 were aged over 15 years. In 35 episodes(69%) the patients died while receiv<strong>in</strong>g mechanicalventilation, and only eight patients (16%) survivedfor more than 6 weeks after discharge. Inthis small study age did not appear to <strong>in</strong>fluencethe probability of survival. Another study exam<strong>in</strong>edthe outcomes of five mechanically ventilatedpatients under 1 year of age. 2 All were successfullyweaned from mechanical ventilation and dischargedhome, and were alive 2–6 years later.Two more recent studies, one <strong>in</strong> the USA 3 andone <strong>in</strong> the UK, 4 have reached somewhat differentconclusions. This is partly related to differences <strong>in</strong>approach and differences <strong>in</strong> the def<strong>in</strong>ition of“survivors”.The US study 3 exam<strong>in</strong>ed the outcomes of 76adult patients with CF admitted to an ICU at a CFand transplant centre. The difference between UKand USA practice is exemplified by the observationthat, of a total of 136 admissions, only 32episodes required endotracheal <strong>in</strong>tubation and 30were admitted for antibiotic desensitisation.Thirty three episodes were precipitated by massivehaemoptysis and 11 of these patients (73%)were alive 1 year after discharge from the ICU. Theauthors did not state whether any of thesepatients required endotracheal <strong>in</strong>tubation, butthis seems unlikely as all of the <strong>in</strong>tubationepisodes appear to be accounted for by <strong>in</strong>fectiveexacerbations. There were a total of 65 admissions


Illustrative case 1: cystic fibrosis 107for <strong>in</strong>fective exacerbations, 32 of which required endotracheal<strong>in</strong>tubation. A total of 71% of the respiratory failure episodesresulted <strong>in</strong> survival to ICU discharge. This <strong>in</strong>cluded 15episodes that did not require any ventilatory support and 18episodes that required non-<strong>in</strong>vasive ventilation only. Twenty(62%) of the patients who received endotracheal <strong>in</strong>tubationsurvived to be discharged from the ICU. Two patients werealive without transplantation 1 year after discharge and 10had successful transplants. Although 17 subjects who wereadmitted with respiratory failure received transplants and 14were alive 1 year later, the proportion of the patients who weretransplanted from the ventilator and were alive 1 year laterwas not stated.The UK study 4 exam<strong>in</strong>ed the outcomes of 31 patients withCF admitted over 8 years to the ICU who requiredendotracheal <strong>in</strong>tubation. In the UK most patients <strong>in</strong> the ICUrequire endotracheal <strong>in</strong>tubation, and restrictions on bed availabilitymean that other patients are cared for <strong>in</strong> high dependencyunits or elsewhere. The patients were divided <strong>in</strong>to twogroups. The first group <strong>in</strong>cluded 12 patients admitted on 13occasions for respiratory failure, either due to aspiration ofblood (three episodes) or <strong>in</strong>fective exacerbations. Five subjects(38%) survived to hospital discharge, a figure similar to theAmerican study described above, but only two patients (16%)survived beyond 6 months. The second group <strong>in</strong>cluded 16patients admitted to the ICU after surgical procedures on 18occasions, 16 of which followed surgical pleurodesis. Fourteenof the subjects survived to hospital discharge and 11 (65%)survived beyond 6 months.What conclusions can be drawn from these studies? Itwould appear that outcomes are good <strong>in</strong> <strong>in</strong>fants requir<strong>in</strong>gmechanical ventilation because of respiratory failure and <strong>in</strong>patients requir<strong>in</strong>g ventilation after surgical pleurodesis. Theoutcome <strong>in</strong> patients admitted to the ICU after massivehaemoptysis and haematemesis also appears relatively good.The outcome <strong>in</strong> the patients <strong>in</strong> the US study 3 also appearedfavourable with the majority alive at 1 year, although (asmentioned above) it appears that these subjects did notrequire endotracheal <strong>in</strong>tubation. Although the outcomes ofpatients with <strong>in</strong>fective exacerbations <strong>in</strong> the US study appearedfavourable, the population studied was different from that <strong>in</strong>the UK study <strong>in</strong> that only a m<strong>in</strong>ority required endotracheal<strong>in</strong>tubation. In addition, many of the patients receiv<strong>in</strong>gmechanical ventilation subsequently had lung transplants,whereas none of the UK patients who were <strong>in</strong>tubated weretransplanted. Both the recent UK and US studies found thatFEV 1was not predictive of survival.The role of transplantation for endotracheally <strong>in</strong>tubatedpatients rema<strong>in</strong>s controversial. Although one study 5 has documenteda1yearsurvivalrateof50%<strong>in</strong>10patients whoreceived mechanical ventilation for up to 42 days, manycentres do not consider these patients for transplantation. Atthe Royal Brompton and Harefield Hospitals 6 only two of fivepatients who had been <strong>in</strong>tubated and mechanically ventilatedsurvived transplantation, and both survivors were ventilatedfor less than 24 hours preoperatively. The poorer outcome ofventilated patients receiv<strong>in</strong>g transplants coupled with severelyrestricted organ availability discourages transplantation <strong>in</strong>this group. The different ways <strong>in</strong> which centres prioritisepatients for transplantation will also <strong>in</strong>fluence whetherorgans will become available with<strong>in</strong> a reasonable time scalefor ventilated patients. In contrast, non-<strong>in</strong>vasive ventilation iswell established as a bridg<strong>in</strong>g technique to transplantation.There are probably many reasons for the poor outcome from<strong>in</strong>vasive ventilation <strong>in</strong> these patients. They usually have severepre-exist<strong>in</strong>g pulmonary disease and may also have associatedliver disease. Even with optimal physiotherapy, sputum clearancewill be less effective than <strong>in</strong> a conscious patient who isable to cough effectively and cooperate with physiotherapy.Reversible factors need to be identified before decid<strong>in</strong>g to<strong>in</strong>tubate and ventilate a patient with CF, and those with endstage disease should not normally receive this form ofsupport. The relative lack of published evidence makes it difficultto make decisions for <strong>in</strong>dividual patients and, where possible,both the patient and the family need to be <strong>in</strong>volved <strong>in</strong>the decision mak<strong>in</strong>g process. It is only rarely appropriate toventilate patients with CF who develop respiratory failure, andthis is well illustrated by the experience at the Royal BromptonHospital where only 16 such episodes have occurred over an 8year period. 4<strong>Management</strong> of massive haemoptysisGuidel<strong>in</strong>es have been published for the management of massivehaemoptysis (>240 ml/day). 78Medical treatment <strong>in</strong>cludesthe correction of coagulation defects with vitam<strong>in</strong> Kand fresh frozen plasma. An <strong>in</strong>fective exacerbation is often aprecipitant and <strong>in</strong>travenous antibiotics should be commenced.The affected lung should be kept dependent <strong>in</strong> an attempt toavoid contam<strong>in</strong>ation of the other lung. There is case reportevidence for the use of tranexamic acid. 9 An attempt should bemade to localise the site of bleed<strong>in</strong>g. This can be achieved <strong>in</strong> anumber of ways. Many patients experience a sensation of gurgl<strong>in</strong>g<strong>in</strong> a particular part of the chest 81011 and a recent studyhas shown that this is a reliable guide. 8 The chest radiographmay also show unilateral air space shadow<strong>in</strong>g. If doubtrema<strong>in</strong>s, bronchoscopy should be performed. Bronchialembolisation is effective 8 and carries a low risk of complicationsthat <strong>in</strong>clude chest pa<strong>in</strong>, dysphagia, bronchial necrosis,10 12 13bowel ischaemia, and, very rarely, paraplegia.<strong>Management</strong> of <strong>in</strong>fective exacerbationsAntibiotics should be selected based on the most recentsputum culture. Pseudomonas aerug<strong>in</strong>osa is usually treated withan am<strong>in</strong>oglycoside <strong>in</strong> comb<strong>in</strong>ation with another antipseudomonalantibiotic. Comb<strong>in</strong>ation therapy is thought to reducethe risk of emergence of resistance. Although a recent studyhas suggested that monotherapy with tobramyc<strong>in</strong> iseffective, 14 it cannot be recommended <strong>in</strong> the ICU sett<strong>in</strong>g. Thechoice of antipseudomonal antibiotic <strong>in</strong>cludes carbapenemssuch as meropenem, carboxypenicill<strong>in</strong>s such as ticarcill<strong>in</strong>,ureidopenicill<strong>in</strong>s such as azlocill<strong>in</strong>, or third generation cephalospor<strong>in</strong>ssuch as ceftazidime. Piperacill<strong>in</strong> appears to be associatedwith febrile reactions <strong>in</strong> patients with CF 15 and shouldbe avoided. Once daily tobramyc<strong>in</strong> does appear to be effective<strong>in</strong> patients with CF, 16 but this conclusion is based on a smallstudy and confirmation is required. Burkholderia cepacia is usuallymultiresistant but may be sensitive to chloramphenicol,cotrimoxazole, ceftazidime, temocill<strong>in</strong>, or meropenem. 17 If thepulmonary pathogen is unknown, antipseudomonal antibioticsshould always be given <strong>in</strong> conjunction with an antistaphylococcalantibiotic such as flucloxacill<strong>in</strong>. Am<strong>in</strong>oglycosideshave a higher volume of distribution <strong>in</strong> CF patients than <strong>in</strong>non-CF <strong>in</strong>dividuals so a higher dose is usually required. Monitor<strong>in</strong>gof am<strong>in</strong>ogylcoside drug levels is essential.Regular physiotherapy is crucial <strong>in</strong> these patients as they areunable to clear their secretions spontaneously. Whether toiletbronchoscopy is better than bl<strong>in</strong>d endobronchial suction isunknown, but <strong>in</strong>spection of the bronchi and suction is usedwhen ventilatory problems arise and sputum plugg<strong>in</strong>g issuspected.The role of recomb<strong>in</strong>ant human deoxyribonuclease (DNase)is uncerta<strong>in</strong> <strong>in</strong> ventilated patients. In patients with CF it18 19reduces the frequency of <strong>in</strong>fective exacerbations and hasbeen shown to be safe and effective <strong>in</strong> those with severedisease. 20 However, efficacy is thought to be dependent uponeffective sputum clearance which is more difficult <strong>in</strong> the ventilatedpatient. The effect of DNase on sputum clearance <strong>in</strong>ventilated patients has not been studied directly but, when apatient has been receiv<strong>in</strong>g DNase before admission to the ICU,it is generally cont<strong>in</strong>ued.


108 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Table 16.1 Common cystic fibrosis related pathologies, complications andpr<strong>in</strong>cipal elements of management <strong>in</strong> <strong>in</strong>tensive care (for details see text)CF related pathology Complication <strong>Management</strong>Lung diseaseBacterial colonisation of airways Infective exacerbations • Antibiotics• Physiotherapy• DNaseMultiresistant pathogens • Antibiotics selected accord<strong>in</strong>gto sensitivitiesSputum plugg<strong>in</strong>g• Endobronchial suction• Bronchoscopy• DNaseObstructive lung disease Difficult ventilation • BronchodilatorsProlonged wean<strong>in</strong>g • Tracheostomy• NIVOther Haemoptysis • Tranexamic acid• Bronchial artery embolisationGastro<strong>in</strong>test<strong>in</strong>al disease Pancreatic <strong>in</strong>sufficiency • Pancreatic enzymes• Elemental feedDistal <strong>in</strong>test<strong>in</strong>al obstructionsyndrome• Ma<strong>in</strong>ta<strong>in</strong> good hydration• Lactulose• N-acetyl cyste<strong>in</strong>e• Gastrograf<strong>in</strong> enemaLiver disease Coagulopathy • Vitam<strong>in</strong> K• Fresh frozen plasmaThrombocytopenia• Platelet transfusionAltered drug metabolism • <strong>Care</strong>ful drug prescrib<strong>in</strong>gpracticeVariceal haemorrhage • Variceal band<strong>in</strong>gDiabetes Hyperglycaemia • Intravenous <strong>in</strong>sul<strong>in</strong>OtherCF upper airway disease S<strong>in</strong>usitis • Avoid nasal <strong>in</strong>tubation• AntibioticsTracheostomies and wean<strong>in</strong>gWean<strong>in</strong>g patients with CF from mechanical ventilation is frequentlyprolonged and tracheostomies are often used to facilitatethis process. A percutaneous rather than a surgical techniquemay be preferred for convenience and a lower <strong>in</strong>cidenceof complications. 21There is limited experience <strong>in</strong> the use ofpercutaneous tracheostomies <strong>in</strong> patients with CF, and it isunknown whether the <strong>in</strong>fected bronchial secretions <strong>in</strong> thesepatients predispose to a higher <strong>in</strong>cidence of postoperative<strong>in</strong>fections.Non-<strong>in</strong>vasive ventilationAs described above, <strong>in</strong>vasive ventilation is associated withpoor outcomes. Non-<strong>in</strong>vasive ventilation has therefore been<strong>in</strong>vestigated as an alternative and has been shown to be effective<strong>in</strong> selected cases. It may also be used to facilitate22 23wean<strong>in</strong>g from mechanical ventilation.Gastro<strong>in</strong>test<strong>in</strong>al pathology and liver diseaseEarly nutritional support should be commenced. An elementalfeed may be used or, alternatively, a standard feed withpancreatic enzyme supplementation. In those with liverdisease a feed with a low sodium content should be used toavoid hypernatraemia.Patients with CF who are acutely unwell, especially aftersurgery, 24are at risk of develop<strong>in</strong>g the distal <strong>in</strong>test<strong>in</strong>alobstruction syndrome (DIOS); fever, dehydration, 25 and opioidanalgesia may all contribute. Preventative strategies <strong>in</strong>cludethe avoidance of dehydration, cont<strong>in</strong>uation of pancreaticenzyme supplementation, use of lactulose, and a carefullyconsidered approach to the use of opioids. Treatment options<strong>in</strong>clude nasogastric or rectal N-acetyl cyste<strong>in</strong>e, Gastrograf<strong>in</strong>enemas, and <strong>in</strong>test<strong>in</strong>al lavage with a balanced electrolytesolution conta<strong>in</strong><strong>in</strong>g polyethylene glycol. Surgery can usuallybe avoided.The <strong>in</strong>cidence of liver disease <strong>in</strong> patients with CF dependson how it is def<strong>in</strong>ed, but it may occur <strong>in</strong> 25% of subjects 26 andsymptomatic liver disease occurs <strong>in</strong> less than 5% of cases.Many patients with CF have a small <strong>in</strong>crease <strong>in</strong> the liverisoenzyme of alkal<strong>in</strong>e phosphatase and γ-glutamyltranspeptidasebut, if these enzymes are raised to more thanfour times normal, then liver disease is usually present. Thepresence of liver disease undoubtedly <strong>in</strong>creases the risk ofcomplications occurr<strong>in</strong>g dur<strong>in</strong>g an ICU admission because ofan <strong>in</strong>creased risk of bleed<strong>in</strong>g due to thrombocytopenia andcoagulopathy. Oesophageal varices may be present. <strong>Care</strong>should be taken with the use of drugs that are metabolised bythe liver or excreted <strong>in</strong> the bile. Ascites may causediaphragmatic spl<strong>in</strong>t<strong>in</strong>g and may need to be dra<strong>in</strong>ed to facilitateventilation.DiabetesA significant proportion of patients have CF related diabetes(CFRD). The prevalence appears to <strong>in</strong>crease with age and onestudy has reported a prevalence of about 15%. 27 Tight controlof blood glucose levels is associated with improved survival <strong>in</strong>critically ill non-CF patients. 28CONCLUSIONIntensive care is only appropriate for a small number ofpatents with CF with reversible complications of their disease.Limited evidence is available to help <strong>in</strong> decid<strong>in</strong>g whichpatients should be selected for support of organ failures <strong>in</strong> theICU and how such patients should be managed. There is aneed for more research <strong>in</strong> this area. A summary of some of theproblems that may be experienced is given <strong>in</strong> table 16.1.


Illustrative case 1: cystic fibrosis 109Where possible, severely ill CF patients should be transferredto their CF specialist centre and this will facilitate optimaltreatment of their multisystem disease. It is also easier fordecisions about cont<strong>in</strong>uation of treatment to be made wherepre-exist<strong>in</strong>g trust<strong>in</strong>g relationships have developed betweenpatients, their relatives, and their cl<strong>in</strong>icians.REFERENCES1 Davis PB, Di Sant’Agnese PA. Assisted ventilation for patients with cysticfibrosis. JAMA 1978; 239:1851–4.2 Garland JS, Chan YM, Kelly KJ, et al. Outcome of <strong>in</strong>fants with cysticfibrosis requir<strong>in</strong>g mechanical ventilation for respiratory failure. Chest1989;96:136–8.3 Sood N, Paradowski LJ, Yankaskas JR. Outcomes of <strong>in</strong>tensive care unitcare <strong>in</strong> adults with cystic fibrosis. Am J Respir Crit <strong>Care</strong> Med200;163:335–8.4 Thomas SR, Evans TW, Geddes TW, et al. CF <strong>in</strong> adults: outcome afteradmission to <strong>in</strong>tensive care (abstract). Pediatr Pulmonol2000;20(Suppl):292.5 Massard G, Shennib H, Metras D, et al. Double-lung transplantation <strong>in</strong>mechanically ventilated patients with cystic fibrosis. Ann Thorac Surg1993;55:1087–91.6 Swami A, Evans TW, Morgan CJ, et al. Ventilation of patients with endstage cystic fibrosis (abstract). J Cardiothorac Vasc Anaesth1992;6(Suppl l):47.7 Schidlow DV, Taussig LM, Knowles MR. Cystic Fibrosis Foundationconsensus conference report on pulmonary complications of cysticfibrosis. Pediatr Pulmonol 1993;15:187–98.8 Br<strong>in</strong>son GM, Noone PG, Mauro MA, et al. Bronchial arteryembolization for the treatment of hemoptysis <strong>in</strong> patients with cysticfibrosis. Am J Respir Crit <strong>Care</strong> Med 1998;157:1951–8.9 Graff GR. Treatment of recurrent severe hemoptysis <strong>in</strong> cystic fibrosis withtranexamic acid. Respiration 2001;68:91–4.10 Tonk<strong>in</strong> IL, Hanissian AS, Boulden TF, et al. Bronchial arteriography andembolotherapy for hemoptysis <strong>in</strong> patients with cystic fibrosis. CardiovascIntervent Radiol 1991;14:241–6.11 Cohen AM, Doershuk CF, Stern RC. Bronchial artery embolization tocontrol hemoptysis <strong>in</strong> cystic fibrosis. Radiology 1990;175:401–5.12 Ivanick, MJ, Thorwarth W, Donohue J, et al. Infarction of the leftma<strong>in</strong>-stem bronchus: a complication of bronchial artery embolization. AmJ Roentgenol 1983;141:535–7.13 Lemoigne F, Rampal P, Petersen R. Fatal ischemic colitis after bronchialartery embolization. Presse Med 1983;12:2056–7.14 Master V, Roberts GW, Coulthard KP, et al. Efficacy of once-dailytobramyc<strong>in</strong> monotherapy for acute pulmonary exacerbations of cysticfibrosis: a prelim<strong>in</strong>ary study. Pediatr Pulmonol 2001;31:367–76.15 Stead RJ, Kennedy HG, Hodson ME, et al. Adverse reactions topiperacill<strong>in</strong> <strong>in</strong> adults with cystic fibrosis. Thorax 1985;40:184–6.16 Whitehead A, Conway SP, Ether<strong>in</strong>gton C, et al. Once-daily tobramyc<strong>in</strong><strong>in</strong> the treatment of adult patients with cystic fibrosis. Eur Respir J2002;19:303–9.17 Ciofu O, Jensen T, Pressler T, et al. Meropenem <strong>in</strong> cystic fibrosis patients<strong>in</strong>fected with resistant Pseudomonas aerug<strong>in</strong>osa or Burkholderia cepaciaand with hypersensitivity to beta-lactam antibiotics. Cl<strong>in</strong> Microbiol Infect1996; 2:91–8.18 Fuchs HJ, Borowitz DS, Christiansen DH, et al. Effect of aerosolizedrecomb<strong>in</strong>ant human DNase on exacerbations of respiratory symptomsand on pulmonary function <strong>in</strong> patients with cystic fibrosis. The PulmozymeStudy Group. N Engl J Med 1994;331:637–42.19 Quan JM, Tiddens HA, Sy JP, et al. A two-year randomized,placebo-controlled trial of dornase alfa <strong>in</strong> young patients with cysticfibrosis with mild lung function abnormalities. J Pediatr2001;139:813–20.20 Shah PI, Bush A, Canny GJ, et al. Recomb<strong>in</strong>ant human DNase I <strong>in</strong> cysticfibrosis patients with severe pulmonary disease: a short-term, double-bl<strong>in</strong>dstudy followed by six months open-label treatment. Eur Respir J1995;8:954–8.21 Freeman BD, Isabella K, L<strong>in</strong> N, et al. A meta-analysis of prospectivetrials compar<strong>in</strong>g percutaneous and surgical tracheostomy <strong>in</strong> critically illpatients. Chest 2000;118:1412–8.22 Hodson ME, Madden BP, Steven MH, et al. Non-<strong>in</strong>vasive mechanicalventilation for cystic fibrosis patients: a potential bridge totransplantation. Eur Respir J 1991;4:524–7.23 Madden BP, Kariyawasam H, Siddiqi AJ, et al. Non<strong>in</strong>vasive ventilation<strong>in</strong> cystic fibrosis patients with acute or chronic respiratory failure. EurRespir J 2002;19:310–3.24 M<strong>in</strong>kes RK, Langer JC, Sk<strong>in</strong>ner MA, et al. Intest<strong>in</strong>al obstruction after lungtransplantation <strong>in</strong> children with cystic fibrosis. J Pediatr Surg1999;34:1489–93.25 Hodson ME, Mearns MB, Batten JC. Meconium ileus equivalent <strong>in</strong> adultswith cystic fibrosis of pancreas: a report of six cases. BMJ1976;2:790–1.26 Nagel RA, Westaby D, Javaid A, et al. Liver disease and bile ductabnormalities <strong>in</strong> adults with cystic fibrosis. Lancet 1989;ii:1422–5.27 Lanng S, Thorste<strong>in</strong>sson B, Lund-Andersen C, et al. Diabetes mellitus <strong>in</strong>Danish cystic fibrosis patients: prevalence and late diabeticcomplications. Acta Paediatr 1994;83:72–7.28 Van den Berghe G, Wouters P, Weekers F, et al. Intensive <strong>in</strong>sul<strong>in</strong>therapy <strong>in</strong> the critically ill patients. N Engl J Med 2001;345:1359–67.


17 Illustrative case 2: <strong>in</strong>terstitial lung diseaseA T Jones, RMduBois, A U Wells.............................................................................................................................CASE REPORTA 31 year old man with a 5 month history of Jo-1negative dermatomyositis was admitted to the<strong>in</strong>tensive care unit (ICU) with respiratory failure.Five months previously he had developed severemyositis which responded to corticosteroid treatment(prednisolone 1 mg/kg) with symptomaticimprovement and a fall <strong>in</strong> creat<strong>in</strong>e k<strong>in</strong>ase. Sixweeks later he developed chest radiographic <strong>in</strong>filtrates,extensive ground glass opacification onhigh resolution computed tomographic (HRCT)scann<strong>in</strong>g (fig 17.1), and hypoxaemic respiratoryfailure despite ma<strong>in</strong>tenance treatment with prednisolone20 mg daily. He deteriorated despiteantimicrobial and <strong>in</strong>creased corticosteroid treatment,requir<strong>in</strong>g mechanical ventilation. A thoracoscopicbiopsy specimen taken while on the ventilatordisclosed diffuse alveolar damage admixedwith organis<strong>in</strong>g pneumonia. Intravenous methylprednisolone(750 mg daily for 3 days) allowedwean<strong>in</strong>g from ventilatory support and eventualdischarge from hospital on prednisolone (0.5 mg/kg) and azathiopr<strong>in</strong>e (200 mg once daily). Onemonth later he was readmitted with <strong>in</strong>creas<strong>in</strong>gdyspnoea and was treated for cytomegalovirus(CMV) pneumonitis diagnosed on the basis of apositive ur<strong>in</strong>ary detection of early antigen fluorescentfoci (DEAFF) test and bronchoalveolarlavage (BAL) immunofluorescence. After a good<strong>in</strong>itial response, progression of <strong>in</strong>terstitial lungdisease became evident radiologically and a s<strong>in</strong>gledose of <strong>in</strong>travenous cyclophosphamide (1.4 g)was adm<strong>in</strong>istered.One week later he developed <strong>in</strong>creas<strong>in</strong>g dyspnoeaassociated with <strong>in</strong>creas<strong>in</strong>g oxygen requirementsand a low grade fever, but there were nomajor changes <strong>in</strong> chest radiographic abnormalitiesor <strong>in</strong>flammatory <strong>in</strong>dices which were onlymildly <strong>in</strong>creased. Broad spectrum antibiotic treatmentwas <strong>in</strong>stituted with cotrimoxazole to coverpneumocystis pneumonia (PCP) and <strong>in</strong>travenousganciclovir to cover recrudescence of CMV pneumonitis.However, all cultures, <strong>in</strong>clud<strong>in</strong>g specifictest<strong>in</strong>g for CMV antigen, were negative. Cont<strong>in</strong>ueddeterioration prompted transfer to the ICU 48hours after admission.Exam<strong>in</strong>ation of bronchoalveolar lavage (BAL)samples revealed no evidence of <strong>in</strong>fection. Hav<strong>in</strong>gfailed to identify an <strong>in</strong>fective agent and <strong>in</strong> thepresence of broad spectrum antimicrobial treatment,the patient’s cont<strong>in</strong>ued decl<strong>in</strong>e was treatedwith three further daily doses of <strong>in</strong>travenousmethylprednisolone and a further dose of <strong>in</strong>travenouscyclophosphamide. The transplantation<strong>in</strong>vestigation protocol was <strong>in</strong>itiated and cyclospor<strong>in</strong>was added <strong>in</strong> the hope of decreas<strong>in</strong>g steroidrequirements. However, <strong>in</strong>termittent non<strong>in</strong>vasivesupport was <strong>in</strong>creas<strong>in</strong>gly necessary andtracheal <strong>in</strong>tubation and mechanical ventilationwere required 7 days after admission to the ICU.Despite vasopressor support, adjustment of antimicrobialtreatment (<strong>in</strong>clud<strong>in</strong>g the empiricaladdition of liposomal amphoteric<strong>in</strong>), and the useof granulocyte colony stimulat<strong>in</strong>g factor to treatpancytopenia, he cont<strong>in</strong>ued to deteriorate anddied 30 days after admission to the ICU. No clearevidence of underly<strong>in</strong>g <strong>in</strong>fection was obta<strong>in</strong>ed.Overall, the balance of probability strongly favoured<strong>in</strong>exorable progression of underly<strong>in</strong>g<strong>in</strong>terstitial lung disease.MANAGEMENT OF PATIENTS WITHDIFFUSE INTERSTITIAL LUNG DISEASE INTHE ICUUse of diagnostic techniquesThis case illustrates important managementdifficulties <strong>in</strong> patients with diffuse <strong>in</strong>terstitiallung disease (DILD) who progress to respiratoryfailure. Cl<strong>in</strong>ically, the differential diagnosis usuallyconsists of deterioration of the underly<strong>in</strong>gdisease demand<strong>in</strong>g <strong>in</strong>creased immunosuppression,and <strong>in</strong>fection requir<strong>in</strong>g antimicrobial treatmentand a reduction <strong>in</strong> immunosuppressivetreatment. The dist<strong>in</strong>ction is important, whateverthe likely outcome. Young patients with connectivetissue disease may have an excellent longterm outcome if they survive an acuteepisode—be it <strong>in</strong>fective or due to <strong>in</strong>flammatoryDILD—and prolonged aggressive <strong>in</strong>tervention isappropriate. By contrast, major progression offibrotic disease generally denotes a very poor outcomeonce mechanical ventilation has been <strong>in</strong>stituted<strong>in</strong> idiopathic and connective tissue diseasealike; prolonged ventilation is <strong>in</strong>appropriate.Unfortunately, <strong>in</strong> most connective tissue diseasesand other forms of DILD no serologicalmarker correlates closely with pulmonary diseaseactivity. The dist<strong>in</strong>ction between the onset of<strong>in</strong>fection and progression of disease is complicatedby the marked similarities <strong>in</strong> cl<strong>in</strong>icalpresentation (fever, cough, <strong>in</strong>creased breathlessness,and <strong>in</strong>creased radiographic shadow<strong>in</strong>g).Similarly, laboratory <strong>in</strong>dices of <strong>in</strong>fection (whiteblood cell count, erythrocyte sedimentation rate,C reactive prote<strong>in</strong>) lack sensitivity or specificity asall may be <strong>in</strong>fluenced by the underly<strong>in</strong>g pulmonary<strong>in</strong>flammation or systemic disease activity.The development of organ dysfunction or nosocomial<strong>in</strong>fection <strong>in</strong> patients requir<strong>in</strong>g <strong>in</strong>tensive caremay add to the diagnostic difficulties. Ideal markersthat dist<strong>in</strong>guish between <strong>in</strong>fection and <strong>in</strong>flammation<strong>in</strong> patients requir<strong>in</strong>g critical care and<strong>in</strong> those with autoimmune disease are not yetavailable for rout<strong>in</strong>e cl<strong>in</strong>ical use. 12Chest radiography is often unhelpful <strong>in</strong> discrim<strong>in</strong>at<strong>in</strong>gbetween <strong>in</strong>fection and progression ofDILD <strong>in</strong> patients requir<strong>in</strong>g critical care. 3HRCTscann<strong>in</strong>g is central to the diagnosis and managementof DILD 4 and is a safe means of obta<strong>in</strong><strong>in</strong>gcl<strong>in</strong>ical and physiological <strong>in</strong>formation <strong>in</strong> criticallyill patients. 5It may be diagnostic <strong>in</strong> advancedDILD, 6 obviat<strong>in</strong>g the need for <strong>in</strong>vasive <strong>in</strong>vestigation.When it is <strong>in</strong>conclusive diagnostically, CTmay def<strong>in</strong>e the optimal site for surgical biopsy.However, although fairly accurate <strong>in</strong> the exclusionof ventilator associated pneumonia <strong>in</strong>


Illustrative case 2: <strong>in</strong>terstitial lung disease 111Presentation with DILD and ARFReversible diseaseYesNoPalliationAdmit to ICU for respiratory supportIs the nature of DILD def<strong>in</strong>ed?NoConsider SLBYesCan the diagnosis of <strong>in</strong>fection or disease progression be made?NoYesFigure 17.1 Th<strong>in</strong> section CT image through the upper lobesshow<strong>in</strong>g patchy consolidation, some of which is peribronchial. Thereis a generalised non-specific <strong>in</strong>crease <strong>in</strong> attenuation of the lungparenchyma. The consolidation could represent autoimmuneorganis<strong>in</strong>g pneumonia associated with connective tissue disease (<strong>in</strong>view of its bronchocentric distribution), but an <strong>in</strong>fective cause for thechanges (or a coexist<strong>in</strong>g <strong>in</strong>fective component) cannot be excluded.Poor quality images are also a major constra<strong>in</strong>t <strong>in</strong> the patient withsevere dyspnoea at rest. Some of the ground glass attenuation maybe technical.ARDS, 7CT scann<strong>in</strong>g has not been evaluated as a diagnostictest for opportunistic <strong>in</strong>fection <strong>in</strong> the ventilated patient withDILD. As illustrated by the present case, the <strong>in</strong>terpretation ofground glass opacification on the CT scan is not alwaysstraightforward, especially <strong>in</strong> connective tissue diseases whereopportunistic <strong>in</strong>fection may coexist with a number of primarypulmonary disease processes. In this sett<strong>in</strong>g, <strong>in</strong>vasive or semi<strong>in</strong>vasive<strong>in</strong>vestigation is often required. In the present casethere was a compell<strong>in</strong>g need to def<strong>in</strong>e the underly<strong>in</strong>g <strong>in</strong>terstitiallung disease and an immediate diagnostic surgical lungbiopsy (SLB) was performed, obviat<strong>in</strong>g semi-<strong>in</strong>vasive lessdef<strong>in</strong>itive procedures. However, <strong>in</strong> cases <strong>in</strong> which theunderly<strong>in</strong>g diagnosis is known and the problem is one of dist<strong>in</strong>guish<strong>in</strong>gbetween <strong>in</strong>fection and disease progression, a morecalibrated approach is usually appropriate <strong>in</strong>clud<strong>in</strong>g BAL andtransbronchial biopsy (TBB) before SLB (fig 17.2).In a case where pulmonary <strong>in</strong>filtrates are associated withimmunosuppressive therapy, BAL makes a crucial contributionto the detection of opportunistic <strong>in</strong>fection. 89 Thespectrum of likely <strong>in</strong>fective organisms depends on a variety offactors <strong>in</strong>clud<strong>in</strong>g the presence of neutropenia, 10 the nature ofthe underly<strong>in</strong>g disease process and immunosuppressivetherapy, 11 12 the prior adm<strong>in</strong>istration of antimicrobialtreatment, 13and the tim<strong>in</strong>g of the BAL relative to hospitaladmission and the onset of ventilation. 14 15 Bacterial pathogensare isolated most commonly, but sta<strong>in</strong><strong>in</strong>g and cultures shouldbe undertaken to exclude fungal, mycobacterial, and viral<strong>in</strong>fections. In addition, non-<strong>in</strong>fectious causes of diffuse radiographicshadow<strong>in</strong>g <strong>in</strong>clud<strong>in</strong>g malignancy and alveolar haemorrhagemay be identified. New diagnostic techniquesapplicable to BAL fluid <strong>in</strong>clude antigen detection (e.g. Aspergillusspp, Cryptococcus neoformans, Legionella pneumophilia),antibody detection (e.g. antipneumolys<strong>in</strong> for pneumococcalpneumonia), special methods for culture (BACTEC radiometricculture for mycobacteria), and techniques from molecularbiology such as the polymerise cha<strong>in</strong> reaction. However, theBALInconclusiveConsider TBB and/or SLBDiagnosticTreat appropriatelyFigure 17.2 An algorithm for the management of patients withdiffuse <strong>in</strong>terstitial lung disease (DILD) and acute respiratory failure(ARF). BAL=bronchoalveolar lavage; TBB=transbronchial biopsy;SLB=surgical lung biopsy.appropriate use of new diagnostic tests is often difficult torationalise; their cl<strong>in</strong>ical usefulness is likely to be heavilydependent upon the quality of specimen, BAL technique, andpopulation studied. 16It is advisable for cl<strong>in</strong>icians to seekmicrobiological advice before perform<strong>in</strong>g BAL if the use ofnovel diagnostic procedures is contemplated.BAL is generally safe <strong>in</strong> immunosuppressed patients<strong>in</strong>clud<strong>in</strong>g those with haematological dysfunction 17and <strong>in</strong>18 19critically ill patients requir<strong>in</strong>g mechanical ventilation.However, deterioration <strong>in</strong> respiratory mechanics and gasexchange is well recognised and may be cl<strong>in</strong>icallysignificant. 20 21 Thus, BAL should be performed <strong>in</strong> the ICU <strong>in</strong>high risk patients; occasionally it is appropriate to <strong>in</strong>stitutemechanical ventilation before BAL is undertaken.The diagnosis of CMV pneumonitis was unexpected butillustrates the diagnostic usefulness of BAL. CMV pneumonitisis probably rare outside transplant patients and those<strong>in</strong>fected with HIV. 22–24 In patients <strong>in</strong>fected with HIV the cl<strong>in</strong>icalsignificance of positive CMV serological test<strong>in</strong>g isuncerta<strong>in</strong>. 24 There are few data <strong>in</strong> other patient groups 25 butCMV pneumonitis is associated with a high rate of mortality(>80%) if treatment is delayed. 26 27 Serological evidence ofprevious CMV <strong>in</strong>fection is common <strong>in</strong> adults, especially afterthe age of 40. As CMV pneumonitis typically presents withfever, dyspnoea and diffuse <strong>in</strong>filtrates on the chest radiograph,it may be very difficult to make the crucial dist<strong>in</strong>ction between<strong>in</strong>fection and progression of disease. 28 Diagnostic viralcytopathic changes <strong>in</strong> TBB or open lung biopsy specimenstend to occur <strong>in</strong> advanced <strong>in</strong>fection by which time antiviraltreatment may be less efficacious, re<strong>in</strong>forc<strong>in</strong>g cont<strong>in</strong>ued <strong>in</strong>terest<strong>in</strong> non-<strong>in</strong>vasive techniques that promise rapid24 25diagnosis.The added value of TBB <strong>in</strong> patients undergo<strong>in</strong>g BALrema<strong>in</strong>s contentious. In a retrospective study of immunosuppressedpatients TBB was more sensitive than BAL (77% v 48%


112 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong><strong>in</strong> HIV disease, 55% v 20% <strong>in</strong> haematological malignancy, 57%v 27% <strong>in</strong> renal transplant recipients) and there were few seriouscomplications. 29 In patients with HIV <strong>in</strong>fection it has beenargued that a negative BAL result should prompt a repeat BALwith TBB at the most abnormal site; this approach has a diagnosticyield of 90% for nodules or focal <strong>in</strong>filtrates. 30 In a retrospectivestudy of mechanically ventilated patients, TBB wasdiagnostic <strong>in</strong> 35% of cases and led to a change <strong>in</strong> management<strong>in</strong> 60% of “medical” patients and <strong>in</strong> 25% of patients follow<strong>in</strong>glung transplantation. 31The frequency of pneumothorax washigher (10.4%) than is generally reported <strong>in</strong> non-ventilatedsubjects (5%), but there were no serious complications. Atsubsequent open lung biopsy or necroscopic exam<strong>in</strong>ationthere was a concordance of 85% with TBB f<strong>in</strong>d<strong>in</strong>gs. Theauthors argue that TBB is safe <strong>in</strong> mechanically ventilatedpatients with undiagnosed pulmonary <strong>in</strong>filtrates and mayobviate the need for SLB.When BAL and TBB are non-diagnostic, SLB may bewarranted. Although widely accepted <strong>in</strong> DILD <strong>in</strong> general, 4 therole of SLB has been questioned <strong>in</strong> ventilated subjects becauseof a perception of higher risk and reduced benefit <strong>in</strong> criticallyill patients. However, it can be argued that diagnostic accuracy<strong>in</strong> this patient group (which is central to confident management)justifies the <strong>in</strong>creased <strong>in</strong>cidence of perioperativecomplications. The diagnostic yield of SLB <strong>in</strong> ventilatedpatients has varied from 46% to 100%, 32–35 but the <strong>in</strong>fluence ofdiagnosis on management is not always easy to quantify. Inone study the atta<strong>in</strong>ment of a def<strong>in</strong>itive diagnosis resulted <strong>in</strong>cont<strong>in</strong>uation of exist<strong>in</strong>g treatment <strong>in</strong> 33% of patients,<strong>in</strong>creased immunosuppression <strong>in</strong> 26%, <strong>in</strong>itiation of immunosuppression<strong>in</strong> 22%, and a change <strong>in</strong> antimicrobial treatment<strong>in</strong> 19%. 32In ventilated patients the mortality rate with SLBmay be as high as 10% and operative complications, occurr<strong>in</strong>g<strong>in</strong> approximately 20%, may <strong>in</strong>fluence survival. 32–35However,mortality after the <strong>in</strong>itial postoperative period is probablylargely ascribable to progression of the underly<strong>in</strong>g conditionrather than to the surgical procedure, although controlledcl<strong>in</strong>ical data are lack<strong>in</strong>g. Factors predict<strong>in</strong>g mortality <strong>in</strong> ventilatedpatients with pulmonary <strong>in</strong>filtrates undergo<strong>in</strong>g SLBhave <strong>in</strong>cluded an immunocompromised status at the onset ofrespiratory failure or current immunosuppressive treatment,severe hypoxia, multiorgan failure, and older age. 33–35In immunocompromised patients <strong>in</strong> the ICU, high <strong>in</strong>patientand 1 year mortality rates (50% and 90%, respectively) areoften cited to suggest that SLB adds little to the managementprovided that broad spectrum antibiotic cover (<strong>in</strong>clud<strong>in</strong>g cotrimoxazoleto cover PCP) and a trial of corticosteroidtreatment are <strong>in</strong>stituted. 36 37 However, “patient benefit” is notalways synonymous with eventual survival. Inappropriateimmunosuppressive therapy may be associated with <strong>in</strong>fectivecomplications. SLB may identify irreversible disease, allow<strong>in</strong>g<strong>in</strong>appropriate support to be m<strong>in</strong>imised 33 34 and withdrawal of38 39care issues to be discussed def<strong>in</strong>itively with the relatives.F<strong>in</strong>ally, immunocompromised patients with underly<strong>in</strong>g DILDcan be viewed as “special cases” with regard to theperformance of SLB because of the unique difficulties <strong>in</strong>dist<strong>in</strong>guish<strong>in</strong>g between <strong>in</strong>fection and progression of theprimary disease.Role of lung transplantationMechanical ventilation is widely regarded as a strong relativecontra<strong>in</strong>dication to lung transplantation because of the highrisk of pneumonia due to airway microbial colonisation, severemuscular decondition<strong>in</strong>g due to immobility, and othercomplications such as sepsis, deep venous thrombosis, gastro<strong>in</strong>test<strong>in</strong>alhaemorrhage, altered gut motility, and nutritionalproblems. 40 The International Society of Heart LungTransplantation/United Network for Organ Shar<strong>in</strong>g has documenteda threefold <strong>in</strong>crease <strong>in</strong> 1 year mortality <strong>in</strong> mechanicallyventilated patients compared with those who are nonventilated.41However, <strong>in</strong> small populations from selectedcentres, outcomes have been similar to those <strong>in</strong> non-ventilatedrecipients. In a recent report of 21 patients who weretransplanted while be<strong>in</strong>g mechanically ventilated, six developedacute allograft dysfunction and all died with<strong>in</strong> a yearwith three not surviv<strong>in</strong>g the postoperative period. 42 However,there were no postoperative deaths among the rema<strong>in</strong><strong>in</strong>g 15patients and their long term survival (40%) did not differ fromnon-ventilated patients undergo<strong>in</strong>g transplantation. Thus,mechanical ventilation should not be viewed as an absolutecontra<strong>in</strong>dication to transplantation. In our patient the factorsfavour<strong>in</strong>g attempted transplantation <strong>in</strong>cluded his age, relativelybrief history (m<strong>in</strong>imis<strong>in</strong>g severe muscular decondition<strong>in</strong>g),and the lack of evidence of relentless systemic diseaseactivity.Non-<strong>in</strong>vasive ventilation decreases the need for tracheal<strong>in</strong>tubation and <strong>in</strong>creases the likelihood of successful wean<strong>in</strong>gfrom mechanical ventilation, both result<strong>in</strong>g <strong>in</strong> a lower<strong>in</strong>cidence of nosocomial <strong>in</strong>fection. 43The successful use ofnon-<strong>in</strong>vasive ventilation as a bridge to transplantation <strong>in</strong>44 45patients develop<strong>in</strong>g respiratory failure has been reported.In view of the general scarcity of donor organs, the <strong>in</strong>dicationsfor transplantation <strong>in</strong> patients receiv<strong>in</strong>g mechanical ventilationare necessarily imprecise and controversial. Decisionsshould not be subject to generic guidel<strong>in</strong>es but must be <strong>in</strong>dividualised,tak<strong>in</strong>g <strong>in</strong>to account such factors as the presence ofreversible superimposed processes (with a realistic chance ofbridg<strong>in</strong>g to surgery with treatment) and the likelihood ofexpeditious transplantation. Immediate transplantation dur<strong>in</strong>gan acute episode is seldom practicable.CONCLUSIONIn patients with <strong>in</strong>terstitial lung disease who deteriorate torespiratory failure, the dist<strong>in</strong>ction between <strong>in</strong>fection and progressionof disease is often difficult and sometimes, as <strong>in</strong> thepresented case, the two may coexist. Accurate managementrequires BAL which is best performed <strong>in</strong> the ICU <strong>in</strong> those withborderl<strong>in</strong>e respiratory failure; TBB may be a useful adjunct. Inselected cases SLB may be <strong>in</strong>valuable both diagnostically andas an aid to confident management. Although seldom justifiable,transplantation of a ventilated patient is not absolutelycontra<strong>in</strong>dicated, especially <strong>in</strong> young non-deconditioned patients.REFERENCES1 Brunkhorst R, Eberhardt OK, Haubitz M, et al. Procalciton<strong>in</strong> fordiscrim<strong>in</strong>ation of systemic autoimmune disease and systemic bacterial<strong>in</strong>fection. Intensive <strong>Care</strong> Med 2000;26:S199–201.2 Re<strong>in</strong>hart K, Karsai W, Meisner M. Procalciton<strong>in</strong> as a marker of thesystemic <strong>in</strong>flammatory response to <strong>in</strong>fection. Intensive <strong>Care</strong> Med2000;26:1193–200.3 Hansell DM. Imag<strong>in</strong>g the <strong>in</strong>jured lung. In: Evans TW, Haslett C, eds.Acute respiratory distress <strong>in</strong> adults. London: Chapman and Hall, 1996:361–79.4 British Thoracic Society. BTS guidel<strong>in</strong>es on the diagnosis, assessmentand treatment of diffuse parenchymal lung disease <strong>in</strong> adults. Thorax1999;54(Suppl 1):S4–14.5 Pesenti A, Tagliabue P, Patroniti N, et al. Computerised tomographyscan imag<strong>in</strong>g <strong>in</strong> acute respiratory distress syndrome. Intensive <strong>Care</strong> Med2001;27:631–9.6 Primack SL, Hartman TE, Hansell DM, et al. End stage lung disease:f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> 61 patients. Radiology 1993;189:681–6.7 W<strong>in</strong>er-Muram HT, Ste<strong>in</strong>er RM, Gurney JW, et al. Ventilator-associatedpneumonia <strong>in</strong> patients with adult respiratory distress syndrome: CTevaluation. Radiology 1998;208:193–9.8 Klech H, Hutter C. Cl<strong>in</strong>ical guidel<strong>in</strong>es for bronchoalveolar lavage (BAL):Report of the European Society of Pneumonology Task Force Group onBAL. Eur Respir J 1990;3:937–74.9 Stover DE, Zaman MB, Hajdu SI, et al. Bronchoalveolar lavage <strong>in</strong> thediagnosis of diffuse pulmonary <strong>in</strong>filtrates <strong>in</strong> the immunocompromisedhost. Ann Intern Med 1984;101:1–7.10 Cordonnier C, Escudier E, Verra F, et al. Bronchoalveolar lavage dur<strong>in</strong>gneutropenic episodes: diagnostic yield and cellular pattern. Eur Respir J1994;7:114–20.11 Ewig S, Torres A, Riquelme R, et al. Pulmonary complications <strong>in</strong>patientswith haematological malignancies treated at a respiratory ICU. Eur RespirJ 1998;12:116–22.


Illustrative case 2: <strong>in</strong>terstitial lung disease 11312 Dunagan DP, Baker AM, Hurd DD, et al. Bronchoscopic evaluation ofpulmonary <strong>in</strong>filtrates follow<strong>in</strong>g bone ,marrow transplantation. Chest1997;111:135–41.13 Hohenadel IA, Kiworr M, Genitsariotis R, et al. Role of bronchoalveolarlavage <strong>in</strong> immunocompromised patients with pneumonia treated withbroad spectrum antibiotic and antifungal regimen. Thorax2001;56:115–20.14 Fagon J, Chastre J, Wolf M, et al. Invasive and non<strong>in</strong>vasive strategiesfor management of suspected ventilator-associated pneumonia. AnnIntern Med 2000;132:621–30.15 Mehta R, Niederman MS. Adequate empirical therapy m<strong>in</strong>imizes theimpact of diagnostic methods <strong>in</strong> patients with ventilator-associatedpneumonia. Crit <strong>Care</strong> Med 2000;28:3092–4.16 Mayaud C, Cadranel J. A persistent challenge: the diagnosis ofrespiratory disease <strong>in</strong> the non-AIDS immunocompromised host. Thorax2000;55:511–7.17 Saito H, Anaissie EJ, Morice RC, et al. Bronchoalveolar lavage <strong>in</strong> thediagnosis of pulmonary <strong>in</strong>filtrates <strong>in</strong> patients with acute leukaemia. Chest1988;94:745–9.18 Ste<strong>in</strong>berg KP, Mitchel DR, Maunder RJ, et al. Safety of bronchoalveolarlavage <strong>in</strong> patients with adult respiratory distress syndrome. Am RevRespir Dis 1993;148:556–61.19 Montravers P, Gauzit R, Dombret MC, et al. Cardiopulmonary effects ofbronchoalveolar lavage <strong>in</strong> critically ill patients. Chest 1993;106:1541–7.20 Kle<strong>in</strong> U, Karzai W, Zimmermann P, et al. Changes <strong>in</strong> pulmonarymechanics after fibreoptic bronchoalveolar lavage <strong>in</strong> mechanicallyventilated patients. Intensive <strong>Care</strong> Med 1998;12:1289–93.21 Verra F, Hmouda H, Rauss A, et al. Bronchoalveolar lavage <strong>in</strong>immunocompromised patients. Cl<strong>in</strong>ical and functional consequences.Chest 1992;101:1215–20.22 Mann M, Shelhamer JH, Masur H, et al. Lack of cl<strong>in</strong>ical utility ofbronchoalveolar lavage cultures for cytomegalovirus <strong>in</strong> HIV <strong>in</strong>fection. AmJ Respir Crit <strong>Care</strong> Med 1997;155:1723–8.23 Mera JR, Whimbey E, Elt<strong>in</strong>g L, et al. Cytomegalovirus pneumonia <strong>in</strong>adult nontransplantation patients with cancer: review of 20 casesoccurr<strong>in</strong>g from 1964 through 1990. Cl<strong>in</strong> Infect Dis 1996;22:1046–50.24 Baughman RP. Cytomegalovirus: the monster <strong>in</strong> the closet. Am J RespirCrit <strong>Care</strong> Med 1997;156:1–2.25 Tamm M, Traenkle P, Grilli B, et al. Pulmonary cytomegalovirus <strong>in</strong>fection<strong>in</strong> immunocompromised patients. Chest 2001;119:838–43.26 Reed EC, Bowden RA, Dandliker PS, et al. Treatment of cytomegaloviruswith ganciclovir and <strong>in</strong>travenous cytomegalovirus immunoglobul<strong>in</strong> <strong>in</strong>patients with bone marrow transplants. Ann Intern Med1988;109:783–8.27 Emmanuel D, Cunn<strong>in</strong>gham I, Jules-Elysee K, et al. Cytomegaloviruspneumonia after bone marrow transplantation successfully treated withcomb<strong>in</strong>ation of ganciclovir and high-dose <strong>in</strong>travenous immune globul<strong>in</strong>.Ann Intern Med 1988;109:777–82.28 Smith CB. Cytomegalovirus pneumonia: state of the art. Chest1989;95:182–7S.29 Cazzadori A, Di Perri G, Todesch<strong>in</strong>i G, et al. Transbronchial biopsy <strong>in</strong>the diagnosis of pulmonary <strong>in</strong>filtrates <strong>in</strong> immunocompromised patients.Chest 1995;107:101–6.30 Cadranel J, Gillet-Juv<strong>in</strong> K, Anto<strong>in</strong>e M. Site directed bronchoalveolarlavage and transbronchial biopsy <strong>in</strong> HIV-<strong>in</strong>fected patients withpneumonia. Am J Respir Crit <strong>Care</strong> Med 1995;152:1103–6.31 O’Brien JD, Ett<strong>in</strong>ger NA, Shevl<strong>in</strong> D, et al. Safety and yield oftransbroncial biopsy <strong>in</strong> mechanically ventilated patients. Crit <strong>Care</strong> Med1997;25:440–6.32 Canver CC, Menttzer RM. The role of open lung biopsy <strong>in</strong> early and latesurvival of ventilator-dependent patients with diffuse idiopathic lungdisease. J Cardiovasc Surg 1994;35:151–5.33 Flabouris A, Myburgh J. The utility of open lung biopsy <strong>in</strong> patientsrequir<strong>in</strong>g mechanical ventilation. Chest 1999;115:811–7.34 Warner DO, Warner MA, Divertie MB. Open lung biopsy <strong>in</strong> patientswith diffuse pulmonary <strong>in</strong>filtrates and acute respiratory failure. Am RevRespir Dis 1988;137:90–4.35 Poe RH, Wahl GW, Qazi R, et al. Predictors of mortality <strong>in</strong> theimmunocompromised patient with pulmonary <strong>in</strong>filtrates. Arch Intern Med1986;146:1304–8.36 Potter D, Pass HI, Brower S, et al. Prospective randomized study of openlung biopsy versus empirical antibiotic therapy foracute pneumonitis <strong>in</strong>non-neutropenic cancer patients. Ann Thorac Surg 1985;40:422–8.37 McKenna RJ, Mounta<strong>in</strong> CF, McMurtrey MJ. Open lung biopsy <strong>in</strong>immunocompromised patients. Chest 1984;86:671–4.38 Ferrand E, Robert R, Ingrand P, et al. Withold<strong>in</strong>g and withdrawal of lifesupport therapy <strong>in</strong> <strong>in</strong>tensive care units <strong>in</strong> France: a prospective survey.Lancet 2001;357:9–14.39 Abbott KH, Breen CM, Abernethy AP, et al. Families look<strong>in</strong>g back: oneyear after discussion of withdraw or withhold<strong>in</strong>g life-susta<strong>in</strong><strong>in</strong>g therapy.Crit <strong>Care</strong> Med 2001;29:197–201.40 American Thoracic Society. International giudel<strong>in</strong>es for the selection oflung transplant candidates. Am J Respir Crit <strong>Care</strong> Med1998;158:335–9.41 O’Brien GD, Cr<strong>in</strong>er GJ. Mechanical ventilation as a bridge to lungtransplantation. J Heart Lung Transplant 1999;18:255–65.42 Meyers BF, Lynch JP, Battafarano RJ, et al. Lung transplant is warranted<strong>in</strong> stable, ventilator-dependent patients. Ann Thorac Surg2000;70:1675–8.43 Evans TW, Albert RK, Angus DC, et al. International ConsensusConferences <strong>in</strong> Intensive <strong>Care</strong> Medic<strong>in</strong>e: Non<strong>in</strong>vasive positive pressureventilation <strong>in</strong> acute respiratory failure. Am J Respir Crit <strong>Care</strong> Med2001;163:283–91.44 Hodson ME, Madden BP, Steven MH. Nonivasive mechanical ventilationfor cystic fibrosis patients: a potential bridge to transplantation. EurRespir J 1991;4:524–7.45 Quarata AJ, Roy B, O’Brien GD, et al. Non<strong>in</strong>vasive positive pressureventilation as bridge therapy to lung transplantation or volume reductionsurgery. Am J Respir Crit <strong>Care</strong> Med 1996;153:A608.


18 Illustrative case 3: pulmonary vasculitisME Griffith, S Brett.............................................................................................................................CASE REPORTA 28 year old man was admitted from a local hospitalto the <strong>in</strong>tensive care unit (ICU) at HammersmithHospital via the operat<strong>in</strong>g theatre. He hadpresented with a 3 month history of nasal stuff<strong>in</strong>ess,epistaxis and haemoptysis, and had recentlynoticed <strong>in</strong>creas<strong>in</strong>g shortness of breath. He hadlost 6 kg <strong>in</strong> weight and reported night sweats. His<strong>in</strong>itial chest radiograph showed diffuse peripheralshadow<strong>in</strong>g and a subsequent computed tomographic(CT) scan confirmed the presence of multiple<strong>in</strong>filtrative lesions (fig 18.1). Bronchoscopyshowed evidence of recent diffuse haemorrhage;bronchoalveolar lavage cytology revealed an eos<strong>in</strong>ophiliabut was negative for acid fast bacilli,legionella, and fungi. Over the next few days hedeteriorated, requir<strong>in</strong>g endotracheal <strong>in</strong>tubationand ventilation. He was transferred for an openlung biopsy. Lung histology was non-specificshow<strong>in</strong>g diffuse alveolar haemorrhage with fibr<strong>in</strong>deposition, fibroblastic proliferation, and a predom<strong>in</strong>antlyneutrophilic <strong>in</strong>flammatory <strong>in</strong>filtrate.Follow<strong>in</strong>g the lung biopsy the patient was admittedto the ICU with severe hypoxaemic respiratoryfailure.The patient had no significant past medical history.He was born <strong>in</strong> India and raised <strong>in</strong> the UK;his last visit abroad was to India 2 years beforeadmission. He was a sales manager for a build<strong>in</strong>gcompany and did not smoke. On admission to ICUhe was pyrexial at 38°C. He was paralysed andventilated requir<strong>in</strong>g 95% oxygen. There were norashes, eyes and jo<strong>in</strong>ts were normal, but he wasnoted to have a slight crust<strong>in</strong>g of his nares. Theblood pressure was 114/54 mm Hg supported bynorep<strong>in</strong>ephr<strong>in</strong>e and dopexam<strong>in</strong>e <strong>in</strong>fusions. Cl<strong>in</strong>icalbiochemistry results were: C reactive prote<strong>in</strong>99 mg/l, Na 135 mmol/l, K 4.3 mmol/l, urea6.1 mmol/l, creat<strong>in</strong><strong>in</strong>e 52 µmol/l, album<strong>in</strong> 16 g/l,ionised calcium 1.88 mmol/l, bilirub<strong>in</strong> 8 µmol/l,alkal<strong>in</strong>e phosphatase 134 U/l (normal range 30–130), and alan<strong>in</strong>e am<strong>in</strong>otransferase 76 U/l (0–31).The blood film showed a leucoerythroblasticpicture; the counts were haemoglob<strong>in</strong> 7.5 g/dl,mean cell volume 89 fl, white cell count 15 × 10 9 /l(neutrophil leucocytosis), platelets 324 × 10 9 /l.Ur<strong>in</strong>e microscopy showed multiple red cell casts.Urgent ANCA (ant<strong>in</strong>eutrophil cytoplasmic antibodies)immunofluorescence was strongly positivefor C-ANCA; anti-PR3 ELISA was also stronglypositive at 89% (0–15%). This confirmed the cl<strong>in</strong>icaldiagnosis of Wegener’s granulomatosis andtreatment was <strong>in</strong>itiated with prednisolone andcyclophosphamide. In view of the severity of hislung haemorrhage he underwent five episodes ofplasma exchange. His renal function deterioratedover 6 days and he required cont<strong>in</strong>uous renalreplacement therapy for fluid balance and metaboliccontrol. The aetiology of the renal failure wasmultifactorial with sepsis and nephritis contribut<strong>in</strong>g.As he was receiv<strong>in</strong>g heavy immunosuppression,he was given anti-<strong>in</strong>fective prophylaxis withco-trimoxazole, fluconazole, and isoniazid. Inorder to m<strong>in</strong>imise further lung haemorrhage, his<strong>in</strong>travascular volume was kept as low as possibleby tightly controll<strong>in</strong>g fluid balance. He susta<strong>in</strong>edone episode of l<strong>in</strong>e sepsis treated with vancomyc<strong>in</strong>.He required high concentrations of oxygen(FiO 2>0.5) for 10 days and then his conditiongradually improved; a tracheostomy was performedand his trachea was ultimately decannulated15 days after ICU admission. His renal functionrecovered and he was discharged fromhospital 46 days after admission with a creat<strong>in</strong><strong>in</strong>elevel of 86 µmol/l. His pulmonary function wascl<strong>in</strong>ically good and was not assessed formally.DISCUSSIONPresentation and diagnosisThis case illustrates the importance of the aggressivepursuit and treatment of the cause of acuterespiratory failure. The history <strong>in</strong> this case providedimportant clues, but <strong>in</strong> some cases of acute severerespiratory failure a cl<strong>in</strong>ical picture of hypoxaemicrespiratory failure with non-specific changes onthe chest radiograph is all that is available on presentationto the ICU. The <strong>in</strong>itial differential diagnosisis wide and <strong>in</strong>cludes most obviously <strong>in</strong>fectiveagents, but also <strong>in</strong>terstitial lung disease and vasculitis.High resolution CT scann<strong>in</strong>g can be helpfulbut, unless it is performed <strong>in</strong> held static <strong>in</strong>spirationwith arms out of the field, can be mislead<strong>in</strong>g. Serologicaltests are extremely valuable but may not berapidly available.Systemic vasculitis is characteristically a multifocaldisease with a variety of cl<strong>in</strong>ical manifestationsaccord<strong>in</strong>g to the size of the blood vessel<strong>in</strong>volved and the organs affected. It is classifiedaccord<strong>in</strong>g to the size of the smallest vessel<strong>in</strong>volved. 1Alveolar haemorrhage results fromsmall vessel vasculitides <strong>in</strong>volv<strong>in</strong>g the lungs;these <strong>in</strong>clude Wegener’s granulomatosis, microscopicpolyangiitis and, rarely, Churg-Strausssyndrome. They can present at any age but occurmost commonly <strong>in</strong> patients <strong>in</strong> their 50s and 60sand there is a slight male predom<strong>in</strong>ance. 23 Thereare reports of vasculitis occurr<strong>in</strong>g <strong>in</strong> families, 4–7but most cases arise sporadically and, unlike mostautoimmune diseases, so far no consistent HLAassociations have been identified, 8 although thereare associations with certa<strong>in</strong> complement allelesand alleles of α 1-antitryps<strong>in</strong>. 9–12Environmentalfactors such as silica exposure, 13–15 drugs, 316 and<strong>in</strong>fections 17–19 may play a part; however, althoughthese may be important <strong>in</strong> elucidat<strong>in</strong>g the pathogenesisof vasculitis, <strong>in</strong> practice most cases haveno obvious genetic or environmental precipitant.As can be seen from this case presentation,prompt diagnosis of vasculitis is vital, not only to<strong>in</strong>stigate appropriate treatment but also to avoidunnecessary <strong>in</strong>vasive diagnostic procedures. Inretrospect, the previously arranged open lungbiopsy <strong>in</strong> this case was probably not required andwas precipitated by the deteriorat<strong>in</strong>g state of thepatient and the need to secure a diagnosis. The


Illustrative case 3: pulmonary vasculitis 115Figure 18.1 CT scan of a ventilated patient with pulmonaryhaemorrhage. Scattered dense airspace shadow<strong>in</strong>g can be seenwith some background ground glass shadow<strong>in</strong>g. Small effusions arealso visible. Courtesy of Dr Jeremy Levy.ANCA result arrived on the ITU at the same time as the patientreturned from theatre.Small vessel vasculitis <strong>in</strong> the lung <strong>in</strong>volves destruction ofarterioles, capillaries and venules by an <strong>in</strong>filtration ofactivated neutrophils. This results <strong>in</strong> <strong>in</strong>terstitial oedema anddiffuse alveolar haemorrhage. Repeated episodes of lunghaemorrhage may result <strong>in</strong> pulmonary fibrosis. 20 Haemoptysisis a common present<strong>in</strong>g feature, although it can be late or evenabsent even <strong>in</strong> the face of significant alveolar bleed<strong>in</strong>g. 21 Othersymptoms <strong>in</strong>clude dyspnoea, fever, and weight loss. Patientsare anaemic, hypoxaemic, and have diffuse <strong>in</strong>filtrativeshadow<strong>in</strong>g on the chest radiograph. Alveolar haemorrhagecan sometimes be confirmed on bronchoscopy, withhaemosider<strong>in</strong>-laden macrophages seen on cytological exam<strong>in</strong>ationof lavage fluid. However, patients may be too hypoxicto undergo this procedure. Carbon monoxide transfer coefficientcorrected for lung volume (KCO) will be raised. In theITU patients are often ventilated, and methods of measur<strong>in</strong>gKCO <strong>in</strong> ventilated patients have been reported and validated,although they are often only available <strong>in</strong> specialist centreswhere operators are familiar with their <strong>in</strong>terpretation. 22 23 ACTscan of the chest is often unhelpful <strong>in</strong> determ<strong>in</strong><strong>in</strong>g theaetiology of alveolar haemorrhage, although it may revealcavitat<strong>in</strong>g lesions typical of granulomatous <strong>in</strong>filtration. Lungbiopsies frequently demonstrate necrotic tissue or show nonspecific<strong>in</strong>flammation and haemorrhage, particularly if obta<strong>in</strong>edvia the transbronchial route, but the yield can beimproved by open lung biopsy. If there is evidence of renal<strong>in</strong>volvement a renal biopsy is more likely to be diagnostic andwill usually show a focal necrotis<strong>in</strong>g glomerulonephritis.Patients with vasculitis may present with isolated pulmonaryhaemorrhage, but usually there is also systemic <strong>in</strong>volvement.Most patients with Wegener’s granulomatosis havegranulomatous lesions <strong>in</strong> the upper respiratory tract whichresult <strong>in</strong> chronic s<strong>in</strong>usitis, epistaxis, chronic otitis media, anddeafness. Involvement of the trachea can present with lifethreaten<strong>in</strong>g stridor. Granulomas are also found <strong>in</strong> many sitesoutside the respiratory system <strong>in</strong>clud<strong>in</strong>g the kidney, centralnervous system, prostate, parotid and orbit. 3Patients withmicroscopic polyangiitis and Wegener’s granulomatosis alsopresent with systemic symptoms secondary to the small vesselvasculitis. Ur<strong>in</strong>e abnormalities <strong>in</strong>clude microscopic haematuriaand prote<strong>in</strong>uria, with red cell casts on microscopy. Theserum creat<strong>in</strong><strong>in</strong>e may <strong>in</strong>itially be <strong>in</strong> the normal range, butacute renal failure tends to develop quite rapidly. Renal andrespiratory <strong>in</strong>volvement both have a significant effect onmortality. 32425Other organs <strong>in</strong> which vasculitis commonlyoccurs are sk<strong>in</strong>, jo<strong>in</strong>ts, muscles, gastro<strong>in</strong>test<strong>in</strong>al system,peripheral and central nervous system, and the eye.Rout<strong>in</strong>e blood counts often show a normocytic or microcyticanaemia with a neutrophil leucocytosis, although there can alsobe eos<strong>in</strong>ophilia (especially <strong>in</strong> Churg-Strauss syndrome) andthrombocythaemia. Thrombocytopenia suggests alternativecauses of lung haemorrhage such as primary haematologicalabnormalities or systemic lupus erythematosus, <strong>in</strong> which casethere will often be accompany<strong>in</strong>g low complement levels andpositive ant<strong>in</strong>uclear antibodies. The erythrocyte sedimentationrate and C reactive prote<strong>in</strong> level will both be raised. Biochemistrywill often show a low serum album<strong>in</strong>, raised alkal<strong>in</strong>e phosphatase,and raised urea and creat<strong>in</strong><strong>in</strong>e. These results may po<strong>in</strong>tto a diagnosis of vasculitis but are relatively non-specific. Themost important serological tests <strong>in</strong> the diagnosis of vasculitisare ant<strong>in</strong>eutrophil cytoplasmic antibodies (ANCA). ANCA werefirst described <strong>in</strong> 1982 26 and subsequently was found to be associatedwith both Wegener’s granulomatosis and microscopicpolyangiitis. There are two ma<strong>in</strong> patterns of ANCA—cytoplasmic (C) and per<strong>in</strong>uclear (P)—def<strong>in</strong>ed by their appearanceon <strong>in</strong>direct immunofluorescence us<strong>in</strong>g ethanol fixed neutrophils.The ma<strong>in</strong> target antigen of C-ANCA is prote<strong>in</strong>ase 3(PR3). 27–30P-ANCA have a wider range of specificities but, <strong>in</strong>systemic vasculitis, they are usually specific for myeloperoxidase(MPO). 31Most patients with Wegener’s granulomatosis haveanti-PR3 specific ANCA, patients with Churg-Strauss syndromeand polyarteritis nodosa have anti-MPO ANCA, and patientswith microscopic polyangiitis have either anti-PR3 or anti-MPOANCA. If immunofluorescence is used alone, then C-ANCA ismore specific than p-ANCA for vasculitis s<strong>in</strong>ce per<strong>in</strong>uclearsta<strong>in</strong><strong>in</strong>g is not only produced by anti-myeloperoxidase antibodiesbut also by ant<strong>in</strong>uclear antibodies and antibodies to neutrophilenzymes not associated with vasculitis. All serum that isANCA positive should therefore be tested <strong>in</strong> PR3 and MPOELISA as this greatly <strong>in</strong>creases their specificity. 32In a largeEuropean trial for the standardisation of ANCA assays, the specificityof immunofluorescence alone was found to be 97% forC-ANCA and 81% for a P-ANCA pattern. The comb<strong>in</strong>ation ofC-ANCA with anti-PR3 and P-ANCA with anti-MPO both had aspecificity of 99%. 33 However, a negative ANCA assay does notexclude the possibility of vasculitis as there are a few cases of34 35small vessel vasculitides that are ANCA negative. In thesecases a tissue diagnosis should be sought.<strong>Management</strong>Systemic vasculitis should be treated with cyclophosphamide(we use 3 mg/kg/day, reduced <strong>in</strong> the elderly to 2 mg/kg/day)comb<strong>in</strong>ed with prednisolone. Historically, cyclophosphamidewas cont<strong>in</strong>ued for at least a year after remission. 36 37 However,<strong>in</strong> view of the side effect profile of cyclophosphamide, manycl<strong>in</strong>icians now substitute azathiopr<strong>in</strong>e <strong>in</strong> stable patients at 3months. The European Vasculitis Study Group has set up aseries of multicentre randomised controlled trials to <strong>in</strong>vestigatethe roles of different immunosuppressive regimes <strong>in</strong> vasculitis.The first of these trials to be completed has recentlyconfirmed that azathiopr<strong>in</strong>e is as effective as cyclophosphamidefor ma<strong>in</strong>tenance of remission after 3 months, 38Morerecently mycophenylate mofetil, levamisole, and TNF directedtreatment have also been used. 39 In patients with fulm<strong>in</strong>antdisease such as dialysis dependent renal failure and alveolarhaemorrhage, there may be an additional benefit from plasma40 41exchange or <strong>in</strong>travenous methylprednisolone and theresults of the current European trials should clarify this. Inpatients who fail to respond to this treatment or who cannottolerate cyclophosphamide due to marrow suppression, theremay also be a role for immunoabsorption, 42 43 pooledimmunoglobul<strong>in</strong>, 44 45 or monoclonal antibodies, 46 although sofar much of the evidence is anecdotal. Lung haemorrhage is amajor cause of early mortality and aggressive supportivetherapy is important. Patients should be kept relatively <strong>in</strong>travascularlydepleted until the lung haemorrhage is controlled,


116 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>and clearly exacerbat<strong>in</strong>g pulmonary oedema must be avoided.Although acute renal failure may be exacerbated by this regimen,this is of secondary importance as the patient can besupported with renal replacement therapy and acute tubularnecrosis caused by <strong>in</strong>itial volume depletion is likely to recover<strong>in</strong> the long term. However, care must be taken to avoid hypoperfusionof the splanchnic circulation with consequent gutand liver dysfunction that may result <strong>in</strong> multiple organ failure.There is little evidence on which to base ventilatorystrategies for adults. A rational approach is to limit excessivetidal volume excursions or pressure changes which are likelyto damage further the fragile vasculature and exacerbatehaemorrhage. Modest gas exchange targets should thereforebe set. However, profound hypoxaemia and hypercapniashould be avoided as this may exacerbate pulmonaryhypertension. A high <strong>in</strong>spired oxygen fraction may be harmfulon theoretical grounds concern<strong>in</strong>g oxidant stress, but a compromiseneeds to be found until the patient responds tospecific treatment. Extracorporeal membrane oxygenation hasbeen used successfully <strong>in</strong> a small number of cases 47 but cannotcurrently be recommended.In summary, patients admitted to the ICU with suspectedpulmonary vasculitis have a high mortality of around 25–50%,depend<strong>in</strong>g upon aetiology. These patients should be discussedearly with a centre which has specialist expertise <strong>in</strong> this areato ensure prompt diagnosis and access to adjuvant therapiessuch as plasma exchange. ANCA assays are vital <strong>in</strong> thediagnosis, and patients with suspected lung haemorrhageshould be tested with<strong>in</strong> 24 hours of presentation. This willhelp to m<strong>in</strong>imise morbidity and mortality from these diseases.Good long term survival can be achieved with prompttreatment with appropriate immunosuppressive agents. Cont<strong>in</strong>uedlow dose immunosuppression and follow up arerequired long term as there is a high <strong>in</strong>cidence of relapse.REFERENCES1 Jennette JC, Falk RJ, Andrassy K, et al. Nomenclature of systemicvasculitides. Proposal of an <strong>in</strong>ternational consensus conference. ArthritisRheum 1994;37:187–92.2 Luqmani RA, Bacon PA, Beaman M, et al. Classical versus non-renalWegener’s granulomatosis. Q J Med 1994;87:161–7.3 Gask<strong>in</strong> G, Pusey CD. Systemic vasculitis. In: Davison AM, Cameron JS,Grunfeld JP, et al, eds. Oxford textbook of cl<strong>in</strong>ical nephrology. Oxford:Oxford University Press, 1996: 877–911.4 Franssen CF, ter Maaten JC, Hoorntje SJ. Brother and sister withmyeloperoxidase associated autoimmune disease. Ann Rheum Dis1994;53:213.5 Heuze-Claudot L, Leroy B, Chevailler A, et al. A familial ANCAassociated pulmonary renal syndrome. Cl<strong>in</strong> Exp Immunol 1993;93(Suppl1):40.6 Knudsen BB, Joergensen T, Munch-Jensen B. Wegener’s granulomatosis<strong>in</strong> a family. A short report. Scand J Rheumatol 1988;17:225–7.7 Munia<strong>in</strong> MA, Moreno JC, Gonzalez Campora R. Wegener’sgranulomatosis <strong>in</strong> two sisters. Ann Rheum Dis 1986;45:417–21.8 Griffith ME, Pusey CD. HLA genes <strong>in</strong> ANCA-associated vasculitides. ExpCl<strong>in</strong> Immunogenet 1997;14:196–205.9 F<strong>in</strong>n JE, Zhang L, Agrawal S, et al. Molecular analysis of C3 allotypes <strong>in</strong>patients with systemic vasculitis. Nephrol Dial Transplant1994;9:1564–7.10 Esnault VL, Testa A, Audra<strong>in</strong> M, et al. Alpha 1-antitryps<strong>in</strong> geneticpolymorphism <strong>in</strong> ANCA-positive systemic vasculitis. Kidney Int1993;43:1329–32.11 Elzouki AN, Segelmark M, Wieslander J, et al. Strong l<strong>in</strong>k between thealpha 1-antitryps<strong>in</strong> PiZ allele and Wegener’s granulomatosis. J InternMed 1994;236:543–8.12 Griffith ME, Lovegrove JU, Gask<strong>in</strong> G, et al. C-ant<strong>in</strong>eutrophil cytoplasmicantibody positivity <strong>in</strong> vasculitis patients is associated with the Z allele ofalpha-1-antitryps<strong>in</strong>, and P- ant<strong>in</strong>eutrophil cytoplasmic antibody positivitywith the S allele. Nephrol Dial Transplant 1996;11:438–43.13 Gregor<strong>in</strong>i G, Ferioli A, Donato F, et al. Association between silicaexposure and necrotiz<strong>in</strong>g crescentic glomerulonephritis with p-ANCA andanti-MPO antibodies: a hospital- based case-control study. Adv Exp MedBiol 1993;336:435–40.14 Hogan SL, Satterly KK, Dooley MA, et al. Silica exposure <strong>in</strong>anti-neutrophil cytoplasmic autoantibody-associated glomerulonephritisand lupus nephritis. J Am Soc Nephrol 2001;12:134–42.15 Nuyts GD, Van Vlem E, De Vos A, et al. Wegener granulomatosis isassociated to exposure to silicon compounds: a case-control study.Nephrol Dial Transplant 1995;10:1162–5.16 Devogelaer JP, Pirson Y, Vandenbroucke JM, et al. D-penicillam<strong>in</strong>e<strong>in</strong>duced crescentic glomerulonephritis: report and review of the literature.J Rheumatol 1987;14:1036–41.17 F<strong>in</strong>kel TH, Torok TJ, Ferguson PJ, et al. Chronic parvovirus B19 <strong>in</strong>fectionand systemic necrotis<strong>in</strong>g vasculitis: opportunistic <strong>in</strong>fection or aetiologicalagent? Lancet 1994;343:1255–8.18 Mandell BF, Calabrese LH. Infections and systemic vasculitis. Curr Op<strong>in</strong>Rheumatol 1998;10:51–7.19 Guillev<strong>in</strong> L, Visser H, Noel LH, et al. Ant<strong>in</strong>eutrophil cytoplasmantibodies <strong>in</strong> systemic polyarteritis nodosa with and without hepatitis Bvirus <strong>in</strong>fection and Churg-Strauss syndrome: 62 patients. J Rheumatol1993;20:1345–9.20 Schwarz MI, Mortenson RL, Colby TV, et al. Pulmonary capillaritis. Theassociation with progressive irreversible airflow limitation andhyper<strong>in</strong>flation. Am Rev Respir Dis 1993;148:507–11.21 Schwarz MI, Brown KK. Small vessel vasculitis of the lung. Thorax2000;55:502–10.22 Macnaughton PD, Morgan CJ, Denison DM, et al. Measurement ofcarbon monoxide transfer and lung volume <strong>in</strong> ventilated subjects. EurRespir J 1993;6:231–6.23 Macnaughton PD, Evans TW. Measurement of lung volume and DLCO<strong>in</strong> acute respiratory failure. Am J Respir Crit <strong>Care</strong> Med1994;150:770–5.24 Cohen BA, Clark WF. Pauci-immune renal vasculitis: natural history,prognostic factors, and impact of therapy. Am J Kidney Dis2000;36:914–24.25 Lauque D, Cadranel J, Lazor R, et al. Microscopic polyangiitis withalveolar hemorrhage. A study of 29 cases and review of the literature.Groupe d’Etudes et de Recherche sur les Maladies “Orphel<strong>in</strong>es”Pulmonaires (GERM”O”P). Medic<strong>in</strong>e (Baltimore) 2000;79:222–33.26 Davies DJ, Moran JE, Niall JF, et al. Segmental necrotis<strong>in</strong>gglomerulonephritis with ant<strong>in</strong>eutrophil antibody: possible arbovirusaetiology? BMJ (Cl<strong>in</strong> Res Ed) 1982;285:606.27 Goldschmed<strong>in</strong>g R, van der Schoot CE, ten Bokkel Hu<strong>in</strong><strong>in</strong>k D, et al.Wegener’s granulomatosis autoantibodies identify a noveldiisopropylfluorophosphate-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> <strong>in</strong> the lysosomes of normalhuman neutrophils. J Cl<strong>in</strong> Invest 1989;84:1577–87.28 Jenne DE, Tschopp J, Ludemann J, et al. Wegener’s autoantigendecoded. Nature 1990;346:520.29 Jennette JC, Hoidal JR, Falk RJ. Specificity of anti-neutrophil cytoplasmicautoantibodies for prote<strong>in</strong>ase 3. Blood 1990;75:2263–4.30 Niles JL, McCluskey RT, Ahmad MF, et al. Wegener’s granulomatosisautoantigen is a novel neutrophil ser<strong>in</strong>e prote<strong>in</strong>ase. Blood1989;74:1888–93.31 Falk RJ, Jennette JC. Anti-neutrophil cytoplasmic autoantibodies withspecificity for myeloperoxidase <strong>in</strong> patients with systemic vasculitis andidiopathic necrotiz<strong>in</strong>g and crescentic glomerulonephritis. N Engl J Med1988;318:1651–7.32 Savige J, Gillis D, Benson E, et al. International Consensus Statement onTest<strong>in</strong>g and Report<strong>in</strong>g of Ant<strong>in</strong>eutrophil Cytoplasmic Antibodies (ANCA).Am J Cl<strong>in</strong> Pathol 1999;111:507–13.33 Hagen EC. Development of solid-phase assays for the detection ofanti-neutrophil cytoplasmic antibodies (ANCA) for cl<strong>in</strong>ical application:report of a large cl<strong>in</strong>ical evaluation study. Cl<strong>in</strong> Exp Immunol1995;101(Suppl 1):29.34 Adu D, Savage COS, Lockwood CM, et al. ANCA positive and ANCAnegative microscopic polyarteritis. Cl<strong>in</strong> Exp Immunol 1995;101(Suppl1):62.35 B<strong>in</strong>di P, Mougenot B, Mentre F, et al. Necrotiz<strong>in</strong>g crescenticglomerulonephritis without significant immune deposits: a cl<strong>in</strong>ical andserological study. Q J Med 1993;86:55–68.36 Fauci AS, Haynes BF, Katz P, et al. Wegener’s granulomatosis:prospective cl<strong>in</strong>ical and therapeutic experience with 85 patients for 21years. Ann Intern Med 1983;98:76–85.37 Hoffman GS, Kerr GS, Leavitt RY, et al. Wegener granulomatosis: ananalysis of 158 patients. Ann Intern Med 1992;116:488–98.38 Jayne D. Update on the European Vasculitis Study Group trials. CurrOp<strong>in</strong> Rheumatol 2001;13:48–55.39 Kallenberg CG, Cohen Tervaert JW. New treatments ofANCA-associated vasculitis. Sarcoid Vasc Diffuse Lung Dis2000;17:125–9.40 Levy JB, W<strong>in</strong>earls CG. Rapidly progressive glomerulonephritis: whatshould be first-l<strong>in</strong>e therapy? Nephron 1994;67:402–7.41 Pusey CD, Rees AJ, Evans DJ, et al. Plasma exchange <strong>in</strong> focalnecrotiz<strong>in</strong>g glomerulonephritis without anti-GBM antibodies. Kidney Int1991;40:757–63.42 Matic G, Michelsen A, Hofmann D, et al. Three cases ofC-ANCA-positive vasculitis treated with immunoadsorption: possiblebenefit <strong>in</strong> early treatment. Ther Apher 2001;5:68–72.43 Palmer A, Cairns T, Dische F, et al. Treatment of rapidly progressiveglomerulonephritis by extracorporeal immunoadsorption, prednisoloneand cyclophosphamide. Nephrol Dial Transplant 1991;6:536–42.44 Jordan SC, Toyoda M. Treatment of autoimmune diseases and systemicvasculitis with pooled human <strong>in</strong>travenous immune globul<strong>in</strong>. Cl<strong>in</strong> ExpImmunol 1994;1:31–8.45 Jayne DR, Chapel H, Adu D, et al. Intravenous immunoglobul<strong>in</strong> forANCA-associated systemic vasculitis with persistent disease activity.Q J Med 2000;93:433–9.46 Lockwood CM, Thiru S, Stewart S, et al. Treatment of refractoryWegener’s granulomatosis with humanized monoclonal antibodies.Q J Med 1996;89:903–12.47 Matsumoto T, Ueki K, Tamura S, et al. Extracorporeal membraneoxygenation for the management of respiratory failure due toANCA-associated vasculitis. Scand J Rheumatol 2000;29:195–7.


19 Illustrative case 4: neuromusculoskeletal disordersN Hart, A K Simonds.............................................................................................................................Congenital or acquired disorders affect<strong>in</strong>gthe respiratory muscles and/or caus<strong>in</strong>gchest wall deformity can precipitate ventilatory<strong>in</strong>sufficiency either through the developmentof <strong>in</strong>spiratory muscle weakness or by amarked <strong>in</strong>crease <strong>in</strong> the work of breath<strong>in</strong>g due tolow thoracic compliance. In some congenital disorderssuch as Duchenne muscular dystrophy and<strong>in</strong>termediate sp<strong>in</strong>al muscular atrophy respiratorymuscle <strong>in</strong>volvement is almost <strong>in</strong>evitable; <strong>in</strong> otherssuch as limb girdle muscular dystrophy andfacioscapulohumeral muscular dystrophy respiratorymuscle weakness is highly variable. Expiratorymuscle weakness reduces cough efficiencyand <strong>in</strong>creases the tendency to atelectasis. Bulbarmuscle <strong>in</strong>volvement predisposes the <strong>in</strong>dividual toaspiration. Risk factors for ventilatory decompensation<strong>in</strong> patients with idiopathic scoliosis <strong>in</strong>cludeearly onset scoliosis (before the age of 5 years), ahigh (cephalad) thoracic curve, and a vital capacityof less than 30% predicted. Some acquiredneuromuscular disorders—for example, motorneurone disease, Guillla<strong>in</strong> Barré syndrome—maypresent with ventilatory failure due to respiratorymuscle weakness; other patients with a precariousbalance between ventilatory load and capacitymay decompensate dur<strong>in</strong>g a chest <strong>in</strong>fection orafter surgical <strong>in</strong>tervention. In some neuromusculardisorders—for example, Duchenne musculardystrophy and acid maltase deficiency—cardiomypathy may complicate the picture. Inaddition, it should be remembered that congenitalscoliosis is associated with an <strong>in</strong>creased<strong>in</strong>cidence of congenital heart disease. Intensivistsshould be able to identify patients at high risk ofventilatory failure from neuromusculoskeletaldisorders and be prepared for wean<strong>in</strong>g problems.They should also be aware of advances <strong>in</strong>non-<strong>in</strong>vasive ventilation (NIV) that may be ofvalue <strong>in</strong> avoid<strong>in</strong>g the need for endotracheal <strong>in</strong>tubationand conventional ventilation, and helpfacilitate early discharge from the <strong>in</strong>tensive careunit (ICU).Figure 19.1 Chest radiograph show<strong>in</strong>g elevatedright hemidiaphragm and bibasal atelectasis, moremarked at the right lung base.CASE REPORTA 40 year old woman was found to be anaemic ata rout<strong>in</strong>e blood donor session. Shortly after shedeveloped jo<strong>in</strong>t pa<strong>in</strong>s and pruritus, and autoimmunehaemolytic anaemia was diagnosed. Amediast<strong>in</strong>al mass was observed on her chestradiograph which was shown to be a thymoma onneedle biopsy. The autoimmune haemolytic anaemiawas complicated by red cell aplasia. Follow<strong>in</strong>gprednisolone therapy the haemoglob<strong>in</strong> rose from6.2 to 12.8 g/dl. There were no symptoms ofmyasthenia such as diplopia, limb weakness ordyspnoea, and the FEV 1/FVC was 2.39/2.98 litres.The patient underwent a thoracotomy dur<strong>in</strong>gwhich the thymic tumour was found to be adherentto the right hilum, right phrenic nerve, andpericardium. The tumour was resected with stripsof the right lung at the hilum. Histologically, theA100.020.0B100.020.090.018.090.018.080.016.080.016.070.014.070.014.0SpO 2 (%)60.050.040.012.010.08.0PtcCO 2 (kPa)SpO 2 (%)60.050.040.012.010.08.0PtcCO 2 (kPa)30.06.030.06.020.04.020.04.010.02.010.02.00.00.00 2:00:00 4:00:00 6:00:00 8:00:00Hours0.00 2:00:00 4:00:00Hours0.06:00:00 8:00:00Figure 19.2 Overnight monitor<strong>in</strong>g of arterial oxygen saturation (SpO 2) and transcutaneous CO 2(PtcCO 2) (A) after surgery breath<strong>in</strong>gspontaneously and (B) dur<strong>in</strong>g NIV.


118 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Table 19.1Tests of respiratorymuscle strength<strong>Respiratory</strong> muscle strength resultsPredicted value(cm H 2 O)Basel<strong>in</strong>e(cm H 2 O)6 months(cm H 2 O)Sniff Poes >70 20.6 29.8Sniff Pdi >70 2.4 13.8Cough Pgas >120 77.3 143.1Bilateral TwPdi >20 0.0 5.0Right TwPdi >7 0.0 0.0Left TwPdi >8 0.0 5.0Sniff Poes=maximum sniff oesphageal pressure; sniff Pdi=maximumsniff trandiaphragmatic pressure; cough Pgas=maximum coughgastric pressure; TwPdi=twitch transdiagphragmatic pressurefollow<strong>in</strong>g unilateral and bilateral magnetic stimulation of the phrenicnerve.20 cm H 2 ONormal twitch PdiPdiPgasPoes100 mstumour was a cortical thymoma. Initially the patient made agood recovery on the ICU and was rapidly extubated. However,over the follow<strong>in</strong>g 2 weeks she became progressively morebreathless and orthopnoeic. The chest radiograph showedelevation of the right hemidiaphragm and bibasal consolidation(fig 19.1). A ventilation-perfusion scan confirmedmatched defects at both lung bases. Little response was seen toseveral courses of antibiotics and physiotherapy, and thepatient reported cont<strong>in</strong>ued orthopnoea and fragmented sleep.She was therefore referred to the wean<strong>in</strong>g programme at theRoyal Brompton Hospital for further assessment.On arrival, breath<strong>in</strong>g spontaneously she was unable to lieflat and the FEV 1/FVC while sitt<strong>in</strong>g was 1.05/1.2 litres. Arterialblood gas tensions on air were PO 210.6 kPa, PCO 25.9 kPa, HCO 330.3 mmol/l. Overnight monitor<strong>in</strong>g dur<strong>in</strong>g which the patientslept very lightly showed a rise <strong>in</strong> transcutaneous (Tc) CO 2to12 kPa with dips <strong>in</strong> arterial oxygen saturation (SaO 2)to60%breath<strong>in</strong>g air (fig 19.2A). A CT scan showed patchysubsegmental atelectasis affect<strong>in</strong>g the right and left lowerlobes with marked elevation of the right hemidiaphragm.There was no evidence of thromboembolism. Basel<strong>in</strong>e respiratorymuscle test results are shown <strong>in</strong> table 19.1 and <strong>in</strong>dicate amarked reduction <strong>in</strong> <strong>in</strong>spiratory muscle strength with a modestreduction <strong>in</strong> expiratory muscle strength. Stimulation of thephrenic nerves generated no transdiaphragmatic pressurespike bilaterally. A tensilon test us<strong>in</strong>g the diaphragm as thetest muscle was negative (fig 19.3).Thyroid function tests demonstrated hypothyroidism (freethyrox<strong>in</strong>e 7.6 pmol/l (NR 9–23), thyroid stimulat<strong>in</strong>g hormone17.7 mU/l (NR 0.32–5)).MSPre tensilon twitch PdiPdiPgasPoesPost tensilon twitch PdiPdiPgasPoesFigure 19.3 Twitch transdiaphragmatic pressure before and afterthe tensilon test. Pdi=transdiaphragmatic pressure; Pgas=gastricpressure; MS=magnetic stimulation. A normal twitch Pdi is givenabove for reference.DiagnosisThe patient had developed bilateral basal atelectasis and consolidationafter thoracotomy for thymic resection because ofmarked diaphragmatic weakness. Ventilatory decompensationoccurred dur<strong>in</strong>g sleep due to loss of <strong>in</strong>tercostal muscle tone <strong>in</strong>REM sleep leav<strong>in</strong>g ventilation dependent on the already weakdiaphragm. Ventilation is further compromised dur<strong>in</strong>g sleepby a reduction <strong>in</strong> central drive and basal ventilation-perfusionmismatch. In this patient the differential diagnosis of respiratorymuscle weakness lies between:• myasthenia gravis;• phrenic nerve <strong>in</strong>jury caused by thymic tumour <strong>in</strong>volvementand/or surgery;• a comb<strong>in</strong>ation of myasthenia and phrenic palsy;• respiratory muscle weakness associated withhypothyroidism. 1The tensilon test was negative and respiratory muscle testsshowed absent conduction down both phrenic nerves <strong>in</strong>dicat<strong>in</strong>gthat the ma<strong>in</strong> problem was phrenic nerve <strong>in</strong>jury. The situationwas probably exacerbated by hypothyroidism <strong>in</strong>ducedmyopathy. Acetylchol<strong>in</strong>esterase (Ach) antibody was subsequentlyshown to be positive <strong>in</strong> this patient, but the comb<strong>in</strong>ationof red cell aplasia, thymoma and Ach antibodies withoutcl<strong>in</strong>ical features of myasthenia has been reported previously.In a Japanese series of 17 cases of red cell aplasia andthymoma, only two patients had myasthenia. 2<strong>Management</strong>The patient was started on nocturnal NIV via a nasal mask andthyrox<strong>in</strong>e replacement therapy. She also received physiotherapydur<strong>in</strong>g NIV to facilitate cough<strong>in</strong>g and secretion clearance.Theoretically, <strong>in</strong> this situation bilevel non-<strong>in</strong>vasive positivepressure support may be more beneficial <strong>in</strong> address<strong>in</strong>gatelectasis than volume preset ventilation or <strong>in</strong>spiratory pressuresupport alone. 3 The patient’s sleep quality improved, andovernight monitor<strong>in</strong>g on NIV showed improvements <strong>in</strong> SaO 2and TcCO 2(fig 19.2B). Persistent anaemia was treated bytransfusion to raise the haemoglob<strong>in</strong> above 8 g/dl. Basal atelectasisresolved over 1 week but the right hemidiaphragmrema<strong>in</strong>ed elevated. The patient was discharged home after 16days and cont<strong>in</strong>ued to use nocturnal NIV.Two months later the sniff <strong>in</strong>spiratory pressure was27.6 cm H 2O compared with a value of 16.2 cm H 2O on arrival,and the patient had returned to work hav<strong>in</strong>g completed acourse of radiotherapy. <strong>Respiratory</strong> muscle test resultsobta<strong>in</strong>ed 6 months after surgery are given <strong>in</strong> table 19.1. Theseshow a further improvement <strong>in</strong> <strong>in</strong>spiratory muscle strengthwith some recovery <strong>in</strong> conduction down the left phrenic nerve(fig 19.4), <strong>in</strong>dicat<strong>in</strong>g that it was probably traumatised dur<strong>in</strong>gthe difficult surgical resection. There was no recovery <strong>in</strong> rightphrenic nerve function, presumably because of partial


Illustrative case 4: neuromusculoskeletal disorders 1195 cm H 2 O100 msNovember 1999PdiPgasPoesMay 2000PdiPgasIn this non-smoker, ventilatory decompensation only occurredat night due to the effects of sleep on respiration, but sleepfragmentation exacerbated her daytime symptoms. <strong>Respiratory</strong>muscle tests 8were used to help differentiate a truemyasthenic syndrome from phrenic nerve damage.NIV is a useful wean<strong>in</strong>g tool. A randomised trial compar<strong>in</strong>grapid extubation on to NIV with cont<strong>in</strong>ued <strong>in</strong>tubation andpressure support ventilation <strong>in</strong> COPD patients showed morerapid wean<strong>in</strong>g with fewer complications <strong>in</strong> the NIV group. 9 Inpatients with restrictive ventilatory defects due to neuromuscularor chest wall disease, case series data 10 suggest that NIVcan shorten wean<strong>in</strong>g and reduce the time spent on the ICU.NIV can also be used to prevent the need for re<strong>in</strong>tubation ifventilatory failure recurs after extubation. 11In the casepresented here, re<strong>in</strong>troduction of <strong>in</strong>vasive ventilation follow<strong>in</strong>gextubation was not <strong>in</strong>dicated but NIV is likely to havefacilitated recovery from basal atelectasis. NIV was successfullycarried out on a high dependency unit and subsequentlyon a general respiratory ward, thereby elim<strong>in</strong>at<strong>in</strong>g the needfor cont<strong>in</strong>ued ICU bed occupancy.MSPoesFigure 19.4 Recovery of left twitch transdiaphragmatic pressureafter 6 months. Pdi=transdiaphragmatic pressure; Poes=oesophagealpressure; Pgas=gastric pressure; MS=magnetic stimulation.resection. A sleep study with the patient breath<strong>in</strong>g spontaneouslyshowed normal TcCO 2and SaO 2values, so the patient wasweaned from nocturnal ventilatory support.DISCUSSIONPhrenic nerve <strong>in</strong>jury occurs variably after cardiothoracicsurgery 4and ranges from complete nerve ablation to the“frostbitten phrenic” seen after procedures <strong>in</strong>volv<strong>in</strong>g topicalcool<strong>in</strong>g to produce cardioplegia. 5 If the nerve is notirretrievably damaged, recovery is usually seen over a numberof months. 6 Patients with bilateral phrenic nerve <strong>in</strong>jury oftenpresent with bibasal atelectasis. In those with underly<strong>in</strong>g respiratorycompromise overt ventilatory failure is precipitated. 7REFERENCES1 Siafakas NM, Salesiotou V, Filaditaki V, et al. <strong>Respiratory</strong> musclestrength <strong>in</strong> hypothyroidism. Chest 1992;102:189–94.2 Masaoka A, Hashimoto T, Yamakawa Y, et al. Thymomas associatedwith pure red cell aplasia. Histologic and follow-up studies. Cancer1989;64:1872–8.3 Elliott MW, Simonds AK. Nocturnal assisted ventilation us<strong>in</strong>g bilevelpositive airway pressure: the effect of expiratory positive airwaypressure. Eur Respir J 1995;8:436–40.4 Tripp HF, Bolton JW. Phrenic nerve <strong>in</strong>jury follow<strong>in</strong>g cardiac surgery: areview. J Cardiac Surg 1998;13:218–23.5 Mills GH, Khan ZP, Moxham J, et al. Effects of temperature on phrenicnerve and diapragmatic function dur<strong>in</strong>g cardiac surgery. Br J Anaesth1997;79:726–32.6 Olopade CO, Staats BA. Time course of recovery from frostbittenphrenics after coronary artery bypass. Chest 1991;99:1112–5.7 Burgess RW, Boyd AF, Moore PG, et al. Post-operative respiratoryfailure due to bilateral phrenic nerve palsy. Postgrad Med J1989;65:39–41.8 Polkey MI, Moxham J. Cl<strong>in</strong>ical aspects of respiratory muscle dysfunction<strong>in</strong> the critically ill. Chest 2001;119:926–9.9 Nava S, Ambros<strong>in</strong>o N, Cl<strong>in</strong>i E, et al. Non-<strong>in</strong>vasive mechanicalventilation <strong>in</strong> the wean<strong>in</strong>g of patients with respiratory failure due tochronic obstructive pulmonary disease. A randomized controlled trial.Ann Intern Med 1998;128:721–8.10 Udwadia ZF, Santis GK, Steven MH, et al. Nasal ventilation to facilitatewean<strong>in</strong>g <strong>in</strong> patients with chronic respiratory <strong>in</strong>sufficiency. Thorax1992;47:715–8.11 Hilbert G, Gruson D, Portel L, et al. Non<strong>in</strong>vasive pressure supportventilation <strong>in</strong> COPD patients with postextubation hypercapnic respiratory<strong>in</strong>sufficiency. Eur Respir J 1998;11:1349–53.


20 Illustrative case 5: HIV associated pneumoniaR J Boyton, D M Mitchell, OMKon.............................................................................................................................An estimated 36 million people worldwideare currently <strong>in</strong>fected with HIV, about 1.46million <strong>in</strong> North America and WesternEurope and a further 25.3 million <strong>in</strong> sub-SaharanAfrica. 1 An estimated 30 000 adults and childrenbecame <strong>in</strong>fected with HIV <strong>in</strong> Western Europedur<strong>in</strong>g the year 2000. The rate of <strong>in</strong>fection,coupled with longer survival due to primary andsecondary prophylaxis aga<strong>in</strong>st opportunistic <strong>in</strong>fectionand highly active antiretroviral therapy(HAART), has resulted <strong>in</strong> the prevalence cont<strong>in</strong>u<strong>in</strong>gto <strong>in</strong>crease. 12Infection with HIV is associated with <strong>in</strong>creasedsusceptibility to opportunistic <strong>in</strong>fection withmore than 100 viruses, bacteria, protozoa andfungi. 3 Primary and secondary prophylaxisaga<strong>in</strong>st opportunistic <strong>in</strong>fections and HAART hasled to changes <strong>in</strong> the nature, <strong>in</strong>cidence, and presentationof opportunistic <strong>in</strong>fections such as Pneumocystispneumonia (PCP), Mycobacterium avium<strong>in</strong>tracellulare (MAI), and cytomegalovirus (CMV)ret<strong>in</strong>itis. 24 New challenges are presented tophysicians <strong>in</strong> medical high dependency units(HDUs) and <strong>in</strong>tensive care units (ICUs). Wereport a patient who presented with HIV associatedpneumonia and discuss the issues concern<strong>in</strong>gadmission to HDU/ICU of HIV <strong>in</strong>fected<strong>in</strong>dividuals <strong>in</strong> the PCP prophylaxis and post-HAART era, draw<strong>in</strong>g together current views ofprognostic <strong>in</strong>dicators and outcomes.CASE REPORTA 39 year old white man presented with a3weekhistory of <strong>in</strong>creas<strong>in</strong>g shortness of breath accompaniedby a non-productive cough, fever, and 5 kgweight loss. A diagnosis of HIV <strong>in</strong>fection with alow CD4 count of 30 cells/mm 3 had been made 6months earlier. He was homosexual with nohistory of recreational <strong>in</strong>travenous drug use. Hewas not tak<strong>in</strong>g PCP prophylaxis or HAART but<strong>in</strong>stead took homeopathic treatment. On physicalexam<strong>in</strong>ation oral candidiasis, oral herpes <strong>in</strong>fection,axillary and <strong>in</strong>gu<strong>in</strong>al lymphadenopathywere identified. He had fever (38°C), tachypnoea,tachycardia, and oxygen saturation on air of 85%.There were no chest signs. A pla<strong>in</strong> chestradiograph showed diffuse bilateral shadow<strong>in</strong>g.Arterial blood gas measurements on air were asfollows: PaO 25.8 kPa, PCO 23.54 kPa, O 2saturation82%. The erythrocyte sedimentation ratio wasraised at 119 mm/h and the C reactive prote<strong>in</strong>(CRP) level was raised at 144 mg/l. Liver and renalfunction tests were normal. The patient wasunable to tolerate a diagnostic bronchoscopy.A cl<strong>in</strong>ical diagnosis of PCP/community acquiredpneumonia was made and he was startedon high dose <strong>in</strong>travenous co-trimoxazole withadjunctive corticosteroid therapy, oral fluconazole,and <strong>in</strong>travenous cefuroxime/oral clarithromyc<strong>in</strong>.Cont<strong>in</strong>uous positive airway pressure(CPAP) ventilation was started. Initially there wascl<strong>in</strong>ical improvement. In particular, the oxygensaturation improved to 93% on air and the CRPlevel fell to 7 mg/l. However, on day 9 he becameunwell with fever (38°C), tachypnoea, and tachycardia.A chest radiograph showed <strong>in</strong>creased diffusebilateral change with a nodular appearanceand patchy consolidation. Arterial blood gasmeasurements on air were as follows: PaO 24.48 kPa, PCO 24.39 kPa, O 2saturation 71%. CRPhad risen to 163 mg/l. He was treated for hospitalacquired pneumonia with piperacill<strong>in</strong>/tazobactam and vancomyc<strong>in</strong> <strong>in</strong> addition to thePCP treatment. Ganciclovir therapy was started.He was transferred to the ICU for <strong>in</strong>creased respiratorysupport with bilevel positive airway pressure(BiPAP) ventilation via a nasal mask andsubsequently improved cl<strong>in</strong>ically.DISCUSSIONPneumonia and HIVThe case described was <strong>in</strong>itially treated empiricallyfor community acquired bacterial pneumonia andPCP. Table 20.1 outl<strong>in</strong>es common HIV associatedpulmonary <strong>in</strong>fections. In the absence of confirmatorytests, a diagnosis of PCP was most likely basedon the cl<strong>in</strong>ical presentation and chest radiographicappearance <strong>in</strong> this at risk patient. PCP is nowadaysmost commonly seen <strong>in</strong> newly diagnosed HIV<strong>in</strong>fected patients with advanced disease or HIV<strong>in</strong>fected <strong>in</strong>dividuals not tak<strong>in</strong>g PCP prophylaxis orHAART. In the case described the patient hadrecently been diagnosed with advanced disease(CD4 count 30 cells/mm 3 ) and was not tak<strong>in</strong>g PCPprophylaxis or HAART. PCP typically presentswhen the CD4 count falls below 200 cells/mm 3 andis one of the most common opportunistic <strong>in</strong>fectionsprecipitat<strong>in</strong>g admission to the HDU and ICUfor respiratory support. 4 10–13The risk of a firstepisode of <strong>in</strong>fection below a CD4 count of200 cells/mm 3 (<strong>in</strong> patients not tak<strong>in</strong>g PCP prophylaxisor HAART) is estimated to be 18% at 12months <strong>in</strong> asymptomatic <strong>in</strong>dividuals, ris<strong>in</strong>g to 44%<strong>in</strong> those with early symptomatic disease such asoral candidiasis as <strong>in</strong> the case described. 14PCPprophylaxis with co-trimoxazole is recommendedwhen the CD4 count falls to 200 cells/mm 3orbelow. Patients with HIV <strong>in</strong>fection on HAART witha CD4 count consistently improved to >200 cells/mm 3 have had PCP primary and secondaryprophylaxis stopped without significant risk ofsubsequent PCP. 15–20Methods of diagnosis range from sputum<strong>in</strong>duction to open lung biopsy. The diagnostic testof choice is fibreoptic bronchoscopy with lavage,provid<strong>in</strong>g the patient can tolerate the procedure.Transbronchial biopsy is useful but is occasionallycomplicated by haemorrhage and pneumothorax.Sputum <strong>in</strong>duction with nebulised sal<strong>in</strong>e has alower diagnostic sensitivity and should be carriedout <strong>in</strong> a negative pressure facility. Patients unableto tolerate bronchoscopy should be treatedempirically, based on cl<strong>in</strong>ical judgement andexpert advice, as was the case here. The case discussedwas treated with high dose co-trimoxazole


Illustrative case 5: HIV associated pneumonia 121Table 20.1HIV associated pulmonary <strong>in</strong>fectionsBacteria Mycobacteria Fungi Parasites VirusesStreptococcus pneumoniae* M tuberculosis** Pneumocystis car<strong>in</strong>ii† Toxoplasma gondii, InfluenzaHaemophilus <strong>in</strong>fluenzae* M avium <strong>in</strong>tracellulare Cryptococcus neoformans*** Cryptosporidium spp Para<strong>in</strong>fluenzaStaphylococcus aureus* M kansasii Candida albicans Microsporidium spp <strong>Respiratory</strong> syncytial virusKlebsiella pneumoniae* Aspergillus spp Leishmania spp Rh<strong>in</strong>ovirusPseudomons aerug<strong>in</strong>osa* Penicillium marneffei Strongyloides stercoralis AdenovirusNocardia asteroides Histoplasma capsulatum CytomegalovirusRochalimaea henselae Coccidiodes immitis Herpes simplex virusBlastomyces dermatitidisHerpes varicella-zoster virusBacterial pneumonia occursmore frequently <strong>in</strong> HIVpositive patients at all CD4counts than HIV negativecontrols. The risk <strong>in</strong>creases asthe CD4 count falls below200 cells/mm 3 and <strong>in</strong><strong>in</strong>travenous drug users 5HIV positive <strong>in</strong>dividuals are Pulmonary <strong>in</strong>fections withat <strong>in</strong>creased risk of <strong>in</strong>fectionwith M tuberculosis, whateverthe CD4 count, and shouldbe offered an HIV test. 7Extrapulmonary tuberculosistends to occur at CD4 counts45 years, and CD4 count100 cells/mm 3to 100% <strong>in</strong>those with CD4 counts


122 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>14 21Table 20.2 Treatment of Pneumocystis pneumonia (PCP)Drug**First l<strong>in</strong>e treatment*Co-trimoxazole 120 mg/kg daily <strong>in</strong>2–4 divided doses po/iv (480mgco-trimoxazole consists ofsulfamethoxazole 400 mg andtrimethoprim 80 mg)Severe disease:*Adjuvant high dose steroids (e.g.prednisolone 40–80 mg daily po.Alternatively, hydrocortisone may begiven iv)Duration oftreatment Side effects Comments21 days Nausea, vomit<strong>in</strong>g, fever, rash(<strong>in</strong>clud<strong>in</strong>g Stevens-Johnson’s syndrome,toxic epidermal necrolysis,photosensitivity), blood disorders(<strong>in</strong>clud<strong>in</strong>g neutropenia,thrombocytopenia, rarelyagranulocytosis and purpura), rarelyallergic reactions, diarrhoea, glossitis,stomatitis, anorexia, arthralgia,myalgia, liver damage, pancreatitis,antibiotic associated colitis,eos<strong>in</strong>ophilia, aseptic men<strong>in</strong>gitis,headache, depression, convulsions,ataxia, t<strong>in</strong>nitus, megaloblastic anaemiadue to trimethoprim, crystaluria, renaldisorders <strong>in</strong>clud<strong>in</strong>g <strong>in</strong>terstitial nephritisIntolerance common. Initial treatment with ivpreparation. Comes <strong>in</strong> ampoule conta<strong>in</strong><strong>in</strong>g 480mg; these should be diluted <strong>in</strong> at least 75 ml of 5%dextrose. Infuse over 60 m<strong>in</strong>utes5 days; reducedose over 14–21daysIndicated <strong>in</strong> severe disease. Optimal dose notdeterm<strong>in</strong>ed. Consult HIV specialist for adviceSecond l<strong>in</strong>e treatmentMild to moderate disease:*Trimethoprim 20 mg/kg/day po/iv <strong>in</strong>2–3 divided doses and dapsone 100mg po daily*Cl<strong>in</strong>damyc<strong>in</strong> 600 mg 6 hourly po/ivand primaqu<strong>in</strong>e 15 mg daily poAtovaquone suspension 750 mg twicedailySevere disease:*Pentamid<strong>in</strong>e isethionate 4 mg/kg/dayas a slow <strong>in</strong>travenous <strong>in</strong>fusion21 days Trimethoprim: gastro<strong>in</strong>test<strong>in</strong>aldisturbance, pruritus, rash, depressionof haematopoiesis; rarely erythemamultiforme, toxic epidermal necrolysis;aseptic men<strong>in</strong>gitisDapsone: haemolysis,methaemoglob<strong>in</strong>aemia, neuropathy,allergic dermatitis, anorexia, nausea,vomit<strong>in</strong>g, <strong>in</strong>somnia, psychosis,agranulocytosis; dapsone syndrome(rash with fever and eos<strong>in</strong>ophilia) -stop immediately (may progress toexfoliative dermatitis, hepatitis,hypoalbum<strong>in</strong>aemia, psychosis anddeath)21 days Cl<strong>in</strong>damyc<strong>in</strong>: diarrhoea, nausea andvomit<strong>in</strong>g; jaundice, abnormal liverfunction tests; neutropenia,eos<strong>in</strong>ophilia, agranulocytosis andthrombocytopenia; rashPrimaqu<strong>in</strong>e: nausea and vomit<strong>in</strong>g,abdom<strong>in</strong>al pa<strong>in</strong>;methaemoglob<strong>in</strong>aemia, heamolyticanaemia.21 days Nausea, vomit<strong>in</strong>g and diarrhoea;headache, <strong>in</strong>somnia; rash, fever;elevated liver enzymes and amylase;anaemia, neutropenia; hyponatraemia21 days Severe reactions, sometimes fatal, dueto hypotension, hypoglycaemia,pancreatitis and arrythmias; alsoleucopenia, thrombocytopenia, acuterenal failure, hypocalcaemia; alsoreported azotaemia, abnormal liverfunction tests, anaemia,hyperkalaemia, nausea and vomit<strong>in</strong>g,dizz<strong>in</strong>ess, syncope, flush<strong>in</strong>g,hyperglycaemia, rash, tastedisturbance; Stevens-Johnson’ssyndrome reported; on <strong>in</strong>halation,bronchoconstriction, cough, shortnessof breath and wheeze; discomfort,pa<strong>in</strong>, <strong>in</strong>duration, abscess formation,and muscle necrosis at <strong>in</strong>jection site.*Trimetrexate 45 mg/m 2 iv and fol<strong>in</strong>icacid 80 mg/m 2 21 days Blood disorders (thrombocytopenia,granulocytopenia and anaemia);diarrhoea and vomit<strong>in</strong>g, oral andgastro<strong>in</strong>test<strong>in</strong>al mucosal ulceration;fever; confusion, rarely seizures;disturbed liver function tests, plasmacalcium, potassium and magnesiumreported; rash, anaphylaxis and localirritation at the <strong>in</strong>jection site.Avoid <strong>in</strong> G6PD deficiencyClostridium difficile tox<strong>in</strong> associated diarrhoea is acomplication of cl<strong>in</strong>damyc<strong>in</strong> therapyPrimaqu<strong>in</strong>e: caution <strong>in</strong> G6PD deficiencyConsider comb<strong>in</strong>ation with iv pentamid<strong>in</strong>e asresistance reported with monotherapyGive over at least 1 hour with patient ly<strong>in</strong>g flat.Monitor blood pressure closely. Important sideeffects <strong>in</strong>clude severe hypotension andhypoglycaemia. Monitor BMstix dur<strong>in</strong>g and after<strong>in</strong>fusion for 12 hours. If chang<strong>in</strong>g fromco-trimoxazole to pentamid<strong>in</strong>e due to poor cl<strong>in</strong>icalresponse, cont<strong>in</strong>ue co-trimoxazole for 3 days. If<strong>in</strong>tolerant, give nebulised pentamid<strong>in</strong>e 600 mgdaily for first 3 days.Used as an alternative for patients <strong>in</strong>tolerant ofco-trimoxazole and pentamid<strong>in</strong>e isethionate or whodo not respond to these drugs. Trimetrexate is apotent dihydrofolate reductase <strong>in</strong>hibitor and mustbe give with calcium fol<strong>in</strong>ate. Adm<strong>in</strong>ister calciumfol<strong>in</strong>ate dur<strong>in</strong>g treatment and for 72 hours after lastdose (to avoid potentially serious bone marrowsuppression, oral and gastro<strong>in</strong>test<strong>in</strong>al ulceration,and renal and hepatic dysfunction); suspendmyelosuppressive drugs (e.g. zidovud<strong>in</strong>e)po=by mouth; iv=<strong>in</strong>travenously.* Consult HIV specialist for advice.**Treatment of PCP <strong>in</strong>fections should be undertaken where facilities for appropriate monitor<strong>in</strong>g are available; consult a microbiologist/HIV specialist andthe product literature before adm<strong>in</strong>ister<strong>in</strong>g these drugs.


Illustrative case 5: HIV associated pneumonia 123Table 20.3 ICU admission, mechanical ventilation, and mortality <strong>in</strong> epidsodes of HIV related Pneumocystis pneumonia(PCP) studied between 1985 and 1997HIV related PCPepisodes studied (n)Country ofstudyPeriod of study% of patientsadmitted to ICU% of patients requir<strong>in</strong>gmechanical ventilationMortality (%) of patientsmechanically ventilatedReference348 USA 1985–89 6.3 5.7 60 312174 USA 1987–90 18 * 62–46** 32110 France 1989–94 100*** 31 79 33257 USA 1990–95 8.2 4.7 50 311660 USA 1995–97 14 9 62 34*Data not available.**Episodes were stratified <strong>in</strong>to patients receiv<strong>in</strong>g care <strong>in</strong> an ICU with (first figure) or without (second figure) a prior AIDS def<strong>in</strong><strong>in</strong>g illness. Data relat<strong>in</strong>g tomechanical ventilation status not available for this study.***This study was limited to ICU admissions.Table 20.4 Prognostic markers significantly associated with mortality <strong>in</strong> ICU admissions of HIV positive patients withPneumocystis pneumonia (PCP) requir<strong>in</strong>g mechanical ventilationHIV related PCP episodes studied (n) Period of study Prognostic markers associated with ICU mortality* Reference110 1989–94 <strong>Respiratory</strong> status deterioration requir<strong>in</strong>g delayed 33mechanical ventilation; mechanical ventilation for 5days or more; nosocomial <strong>in</strong>fection, pneumothorax48* 1993–96 Low CD4 cell count with<strong>in</strong> 2 weeks of admission** 40176 1990–99 Low CD4 cell count; prior PCP prophylaxis, CMV <strong>in</strong> 9BAL fluid, age, <strong>in</strong>itial anti-PCP therapy155 1995–97 Prior PCP prophylaxis 3421 1993–98 High APACHE II score >17, low serum album<strong>in</strong>


124 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>respiratory failure secondary to pneumonia is associated witha positive outcome. HIV and <strong>in</strong>tensive care physicians need towork <strong>in</strong> close collaboration to deliver optimal care.ACKNOWLEDGEMENTThe authors thank Dr Daniel Altmann for his critical review of themanuscript. Department of Infectious Diseases, Hammersmith Campus,Imperial College, London, UK.REFERENCES1 Piot P, Bartos M, Ghys PD, et al. The global impact of HIV/AIDS. Nature2001;410:968–73.2 Palella FJ, Delaney KM, Moorman AC, et al. Decl<strong>in</strong><strong>in</strong>g mortality amongpatients with advanced human immunodeficiency virus <strong>in</strong>fection. N Engl JMed 1998;338:853–60.3 Weiss RA. Gulliver’s travels <strong>in</strong> HIVland. Nature 2001;410:963–7.4 Kovacs JA, Masur H. Prophylaxis aga<strong>in</strong>st opportunistic <strong>in</strong>fections <strong>in</strong>patients with human immunodeficiency virus <strong>in</strong>fection. N Engl J Med2000;342:1416–29.5 Hirschtick RE, Glassroth J, Jordan MC, et al. Bacterial pneumonia <strong>in</strong>persons <strong>in</strong>fected with the human immunodeficiency virus. N Engl J Med1995;333:845–51.6 Afessa B, Green, B. Bacterial pneumonia <strong>in</strong> hospitalized patients withHIV <strong>in</strong>fection. Chest 2000;117:1017–22.7 Bowen EF, Rice PS, Cooke NT, et al. HIV seroprevalence by anonymoustest<strong>in</strong>g <strong>in</strong> patients with Mycobacterium tuberculosis and <strong>in</strong> tuberculosiscontacts. Lancet 2000;356:1488–9.8 Saag MS, Graybill RJ, Larsen RA, et al. Practice guidel<strong>in</strong>es for themanagement of cryptococcal disease. Cl<strong>in</strong> Infect Dis 2000;30:710–18.9 Benfield TL, Helweg-Larsen J, Bang D, et al. Prognostic markers ofshort-term mortality <strong>in</strong> AIDS-associated Pneumocystis car<strong>in</strong>ii pneumonia.Chest 2001;119:844–51.10 De Palo VA, Millste<strong>in</strong> BH, Mayo PH, et al. Outcome of <strong>in</strong>tensive care <strong>in</strong>patients with HIV <strong>in</strong>fection. Chest 1995;107:506–10.11 Casal<strong>in</strong>o E, Mendoza-Sassi G, Wolff M, et al. Predictors of short andlong term survival <strong>in</strong> HIV <strong>in</strong>fected patients admitted to the ICU. Chest1998;113:421–9.12 Afessa B, Green B. Cl<strong>in</strong>ical course, prognostic factors, and outcomeprediction for HIV patients <strong>in</strong> ICU. Chest 2000;118:138–45.13 Gill JK, Greene L, Miller R, et al. ICU admission <strong>in</strong> patients <strong>in</strong>fected withhuman immunodeficiency virus: a multicentre survey. Anaesthesia1999;54:727–32.14 Weller V, Williams I. ABC of AIDS. Treatment of <strong>in</strong>fections. BMJ2001;322:1350–4.15 Schneider MME, Borleffs JCC, Stolk RP, et al. Discont<strong>in</strong>uation ofprophylaxis for Pneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> HIV-1 <strong>in</strong>fected patientstreated with highly active antiretroviral therapy. Lancet1999;353:201–3.16 Furrer H, Egger M, Opravil M, et al. Discont<strong>in</strong>uation of primaryprophylaxis aga<strong>in</strong>st Pneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> HIV-1 <strong>in</strong>fectedadults treated with comb<strong>in</strong>ation antiretroviral therapy. N Engl J Med1999;340:1301–6.17 Kirk O, Lundgren JD, Pedersen C, et al. Can chemoprophylaxisaga<strong>in</strong>st opportunistic <strong>in</strong>fections be discont<strong>in</strong>ued after an <strong>in</strong>crease <strong>in</strong> CD4cells <strong>in</strong>duced by highly active antiretroviral therapy? AIDS1999;13:1647–51.18 Weverl<strong>in</strong>g GJ, Mocroft A, Ledergerber B, et al. Discont<strong>in</strong>uation ofPneumocystis car<strong>in</strong>ii pneumonia prophylaxis after start of highly activeantiretroviral therapy <strong>in</strong> HIV-1 <strong>in</strong>fection. Lancet 1999;353:1293–8.19 Lopez Bernaldo De Quiros JC, Miro JM, Pena JM, et al. A randomizedtrial of the discont<strong>in</strong>uation of primary and secondary prophylaxis aga<strong>in</strong>stPneumocystis car<strong>in</strong>ii pneumonia after highly active antiretroviral therapy<strong>in</strong> patients with HIV <strong>in</strong>fection. N Engl J Med 2001;344:159–67.20 Ledergerber B, Mocroft A, Reiss P, et al. Discont<strong>in</strong>uation of secondaryprophylaxis aga<strong>in</strong>st Pneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> patients with HIV<strong>in</strong>fection who have a response to antiretroviral therapy. N Engl J Med2001;344:168–74.21 British National Formulary. British Medical Association and the RoyalPharmaceutical Society of Great Brita<strong>in</strong>, March 2000: 39.22 Millar A, Patou G, Miller R, et al. Cytomegalovirus <strong>in</strong> the lungs ofpatients with AIDS: respiratory pathogen or passenger? Am Rev RespirDis 1990;141:1474–7.23 Miles P, Baughman R, L<strong>in</strong>neman C. Cytomegalovirus <strong>in</strong> BAL fluid ofpatients with AIDS. Chest 1990;97:1072–6.24 Nelson M, Ersk<strong>in</strong>e D, Hawk<strong>in</strong>s D, et al. Treatment with corticosteroids: arisk factor for the development of cl<strong>in</strong>ical cytomegalovirus disease <strong>in</strong>AIDS. AIDS 1993;7:375–8.25 Lathey J, Spector S. Unrestricted replication of human cytomegalovirus<strong>in</strong> hydrocortisone treated macrophages. J Virol 1991;65:6371.26 Dwork<strong>in</strong> M, Hanson D, Nav<strong>in</strong> T. Survival of patients with AIDS after adiagnosis of Pneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> the United States. J InfectDis 2001;183:1409–12.27 Curtis JR. ICU outcomes for patients with HIV <strong>in</strong>fection: a mov<strong>in</strong>g target(editorial). Chest 1998;113:269–70.28 Rosen M, Cucco R, Teirste<strong>in</strong> A. Outcome of <strong>in</strong>tensive care <strong>in</strong> patientswith the acquired immunodeficiency syndrome. J Intensive <strong>Care</strong> Med1986;1:55–60.29 Wachter RM, Luce JM, Turner J, et al. Intensive care of patients with theacquired immunodeficiency syndrome: outcome and chang<strong>in</strong>g patterns ofutilization. Am Rev Respir Dis 1986;134:891–6.30 Murry J, Felton C, Garay M, et al. Pulmonary complications of theacquired immunodeficiency syndrome: report of the National Heart,Lung, and Blood Institute workshop. N Engl J Med 1984;310:1682–8.31 Naresh G, Mansharamani M, Garland R, et al. <strong>Management</strong> andoutcome patterns for adult Pneumocystis car<strong>in</strong>ii pneumonia, 1985 to1995. Chest 2000;118:704–11.32 Curtis J, Randall M, Bennett C, et al. Variations <strong>in</strong> <strong>in</strong>tensive care unitutilization for patients with human immunodeficiency virus-relatedPneumocystis car<strong>in</strong>ii pneumonia: Importance of hospital characteristicsand geographic location. Crit <strong>Care</strong> Med 1998;26:668–75.33 Bedos JP, Dumoul<strong>in</strong> JL, Gachot B, et al. Pneumocystis car<strong>in</strong>ii pneumoniarequir<strong>in</strong>g <strong>in</strong>tensive care management: survival and prognostic study <strong>in</strong>110 patients with human immunodeficiency virus. Crit <strong>Care</strong> Med1999;27:1109–15.34 Randall Curtis J, Yarnold PR, Schwartz RA, et al. Improvements <strong>in</strong>outcomes of acute respiratory failure for patients with humanimmunodeficiency virus related Pneumocystis car<strong>in</strong>ii pneumonia. Am JRespir Crit <strong>Care</strong> Med 2000;162:393–8.35 El-Sadr W, Simberkoff MS. Survival and prognostic factors <strong>in</strong> severePneumocystis car<strong>in</strong>ii pneumonia requir<strong>in</strong>g mechanical ventilation. Am RevRespir Dis 1988;137:1264–7.36 Montaner JS, Russell JA, Lawson L, et al. Acute respiratory failuresecondary to Pneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> the acquiredimmunodeficiency syndrome: a potential role for corticosteroids. Chest1989;95:881–4.37 Efferen LS, Nadarajah D, Palat DS. Survival follow<strong>in</strong>g mechanicalventilation for Pneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> patients with theacquired immunodeficiency syndrome: a different perspective. Am J Med1989;87:401–4.38 Friedman Y, Frankl<strong>in</strong> C, Rackow C, et al. Improved survival <strong>in</strong> patientswith AIDS, Pneumocystis car<strong>in</strong>ii pneumonia, and severe respiratoryfailure. Chest 1989;96:862–6.39 Wachter RM, Russi MB, Bloch DA, et al. Pneumocystis car<strong>in</strong>ii pneumoniaand respiratory failure <strong>in</strong> AIDS. Improved outcomes and <strong>in</strong>creased use of<strong>in</strong>tensive care units. Am Rev Respir Dis 1991;143:251–6.40 Kumar SD, Krieger BP. CD4 lymphocyte counts and mortality <strong>in</strong> AIDSpatients requir<strong>in</strong>g mechanical ventilator support due to Pneumocystiscar<strong>in</strong>ii pneumonia. Chest 1998;113:430–3.41 Arozullah AM, Yarnold PR, We<strong>in</strong>ste<strong>in</strong> RA, et al. A new preadmissionstag<strong>in</strong>g system for predict<strong>in</strong>g <strong>in</strong>patient mortality from HIV-associatedPneumocystis car<strong>in</strong>ii pneumonia <strong>in</strong> the Early Highly Active AntiretroviralTherapy (HAART) era. Am J Respir Crit <strong>Care</strong> Med 2000;161:1081–6.42 Alves C, Nicolas J, Miro J, et al. Reapprasial of the aetiology andprognostic factors of severe acute respiratory failure <strong>in</strong> HIV patients. EurRespir J 2001;17:87–93.43 Richman DD. HIV chemotherapy. Nature 2001;410:995–1001.44 Ledergerber B, Egger M, Oppravil, et al. Cl<strong>in</strong>ical progression andvirological failure on highly active antiretroviral therapy <strong>in</strong> HIV patients: aprospective cohort study: Swiss HIV Cohert Study. Lancet1999;353:863–8.45 Whitcup SM, Fort<strong>in</strong> E, L<strong>in</strong>dbald AS, et al. Discont<strong>in</strong>uation ofanticytomegalovirus therapy <strong>in</strong> persons with HIV <strong>in</strong>fection andcytomegalovirus ret<strong>in</strong>itis. JAMA 1999;282:1633–7.46 Macdonald JC, Torriani FJ, Morse LS, et al. Lack of reactivation ofcytomegalovirus (CMV) ret<strong>in</strong>itis after stopp<strong>in</strong>g CMV ma<strong>in</strong>tenance therapy<strong>in</strong> AIDS patients with susta<strong>in</strong>ed elevations <strong>in</strong> CD4 T cells <strong>in</strong> response tohighly active antiretroviral therapy. J Infect Dis 1998;177:1182–7.47 Tural C, Romeu J, Sirera G, et al. Long last<strong>in</strong>g remission ofcytomegalovirus ret<strong>in</strong>itis without ma<strong>in</strong>tenance therapy <strong>in</strong> humanimmunodeficiency virus <strong>in</strong>fected patients. J Infect Dis 1998;177:1080–3.48 Vrabec TR, Baldassano VF, Whitcup SM, et al. Discont<strong>in</strong>uation ofma<strong>in</strong>tenance therapy <strong>in</strong> patients with quiescent cytomegalovirus ret<strong>in</strong>itisand elevated CD4+ counts. Ophthalmology 1998;105:1259–64.49 Jones JL, Hanson DL, Dwork<strong>in</strong> MS, et al. Surveillance for AIDS-def<strong>in</strong><strong>in</strong>gopportunistic illnesses, 1992–1997. MMWR CDC Surveill Summ1999;48:1–22.50 Miller V, Mocroft A, Reiss P, et al. Relations among CD4 lymphocytecount nadir, antiretroviral therapy, and HIV-1 disease progression: resultsfrom the EuroSIDA study. Ann Intern Med 1999;130:570–7.51 Wislez M, Bergot E, Anto<strong>in</strong>e M, et al. Acute respiratory failure follow<strong>in</strong>gHAART <strong>in</strong>troduction <strong>in</strong> patients treated for Pneumocystis car<strong>in</strong>iipneumonia. Am J Respir Crit <strong>Care</strong> Med 2001;164:847–51.


21 Illustrative case 6: acute chest syndrome of sickle cellanaemiaV Mak, S C Davies.............................................................................................................................CASE REPORTA 21 year old Afro-Caribbean man with knownsickle cell disease (SCD) was admitted to hospitalwith pa<strong>in</strong>ful chest, thighs, and generalisedabdom<strong>in</strong>al pa<strong>in</strong>. This was his second admission tohospital with a sickle cell crisis. He was on noregular medications apart from analgesia taken athome dur<strong>in</strong>g crises. Apart from anaemia, therewas noth<strong>in</strong>g abnormal to f<strong>in</strong>d on exam<strong>in</strong>ation.The haemoglob<strong>in</strong> (Hb) was 7.5 g/dl (normal forhim), white cell count 22.5 × 10 9 , and the electrolyteswere normal apart from a slightly raisedC-reactive prote<strong>in</strong> (CRP) level of 30 mg/l. Radiographsof his abdomen and thighs were normal,but his chest radiograph showed a small degree ofbasal atelectasis bilaterally. His oxygen saturationwas 96% on air. He was treated with a subcutaneousopiate <strong>in</strong>fusion us<strong>in</strong>g a syr<strong>in</strong>ge pump, <strong>in</strong>travenousfluids, oxygen, and encouraged to dr<strong>in</strong>k.Over the next 24 hours his pa<strong>in</strong> was not wellcontrolled and required an <strong>in</strong>creas<strong>in</strong>g dose of opiates.He became pyrexial (38.5°C), his oxygensaturation fell to 92% on air, and antibiotics tocover community acquired pneumonia were commenced.On the third day, however, he becamemore drowsy with arterial blood gases of pH 7.35,PaO 213.5 kPa, and PaCO 27.5 kPa on 40% oxygen. Arepeat chest radiograph showed new <strong>in</strong>filtrates <strong>in</strong>both lower zones and a diagnosis of acute sicklechest syndrome (ACS) was made. Despite anexchange transfusion, he cont<strong>in</strong>ued to deteriorateand was eventually <strong>in</strong>tubated and ventilated. On<strong>in</strong>tensive care his FiO 2was reduced from the<strong>in</strong>itial 50% to 28% with<strong>in</strong> 24 hours. Sputum samplesobta<strong>in</strong>ed by tracheobronchial suction showedno significant bacterial growth, but his CRP hadrisen to 150 mg/l so the antibiotic spectrum wasbroadened. After 3 days of mechanical ventilationhis chest radiograph showed significant clear<strong>in</strong>gof the lower zones, he was extubated without<strong>in</strong>cident, and discharged from hospital after afurther few days. Subsequent atypical respiratoryserological exam<strong>in</strong>ation did not show any rise <strong>in</strong>titres.DISCUSSIONACS consists of a comb<strong>in</strong>ation of signs and symptoms<strong>in</strong>clud<strong>in</strong>g dyspnoea, chest pa<strong>in</strong>, fever,cough, multifocal pulmonary <strong>in</strong>filtrates on thechest radiograph, and a raised white cell count. Itis a form of lung <strong>in</strong>jury that can progress to adultrespiratory distress syndrome (ARDS). It isestimated that half of all patients with sickle cellanaemia will develop ACS at least once <strong>in</strong> theirlives, and ACS is the second most common causeof admission after pa<strong>in</strong>ful vaso-occlusive crises.The most recent statistics from the USA suggestthat 13% of patients with ACS require mechanicalventilation with a mortality rate of 3%, mostlyaffect<strong>in</strong>g adults 1 ; the average length of stay was10.5 days. ACS is the most common cause ofdeath <strong>in</strong> these patients 2–4 and may be the cause ofthe chronic pulmonary abnormalities seen onhigh resolution CT scann<strong>in</strong>g. 5Although this illustrative case with a typicalpresentation of ACS had a favourable outcome,the question is raised of whether anyth<strong>in</strong>g couldhave been done to avoid respiratory failure.Pathophysiology of the acute chestsyndrome <strong>in</strong> sickle cell diseaseThe genetic defect <strong>in</strong> sickle cell anaemia causes asubstitution of val<strong>in</strong>e for glyc<strong>in</strong>e <strong>in</strong> the β-glob<strong>in</strong>subunit of haemoglob<strong>in</strong> to form HbS. 6 HbS is lesssoluble than normal haemoglob<strong>in</strong> (HbA) whendeoxygenated, as a result of which HbS polymeriseswith<strong>in</strong> the cell. This stiffens the erythrocyteand changes it from its normal biconcave form toa sickle shaped cell. The sickle cell also loses theflexibility required to traverse capillary beds.Hypoxia also enhances adhesion of red cells to thevascular endothelium, a process mediated by<strong>in</strong>teraction between very late activation antigen 4(VLA-4) expressed on sickle cells and vascular celladhesion molecule 1 (VCAM-1) on endothelialcells. 7 As a consequence, the sickle cells occludesmall and sometimes larger vessels caus<strong>in</strong>gvascular <strong>in</strong>jury, especially to organs with sluggishcirculation such as the spleen and bone marrowand <strong>in</strong> atelectatic areas of lung.There are four precipitants of ACS: <strong>in</strong>fection,atelectasis, fat embolism, and true thromboembolism(fig 21.1). Each may progress to a commonf<strong>in</strong>al pathway of reduced ventilation with hypoxiaand <strong>in</strong>creased sickl<strong>in</strong>g. The most common f<strong>in</strong>d<strong>in</strong>g<strong>in</strong> the lung dur<strong>in</strong>g an acute pa<strong>in</strong>ful crisis affect<strong>in</strong>gthe chest wall is atelectasis from a comb<strong>in</strong>ation ofpoor chest expansion caused by pa<strong>in</strong>ful rib andvertebral <strong>in</strong>farction and suppressed respiratorydrive due to opiates. Atelectasis promotes sickl<strong>in</strong>glocally due to hypoxia caus<strong>in</strong>g <strong>in</strong>flammation,<strong>in</strong>travascular coagulation, and vascular obstructionwith eventual micro-<strong>in</strong>farction. In adults,particularly, vaso-occlusion <strong>in</strong> the bone marrowcauses marrow <strong>in</strong>farction and fat embolism. Atpost mortem exam<strong>in</strong>ation many patients havebony spicules and marrow fat <strong>in</strong> the lung, andlipid laden macrophages can be found <strong>in</strong>bronchoalveolar lavage (BAL) fluid of patientswith ACS. 8There is also <strong>in</strong>creased circulat<strong>in</strong>gsecretory phospholipase A2, a potent <strong>in</strong>flammatorymediator orig<strong>in</strong>at<strong>in</strong>g from the bone marrow,both <strong>in</strong> patients with ACS and those who are atrisk of develop<strong>in</strong>g ACS, but not <strong>in</strong> those withuncomplicated vaso-occlusive crises. 910Nitric oxide (NO) is an endothelium derivedvasodilator and a modulator of diverse <strong>in</strong>flammatoryprocesses. Plasma concentrations of secretoryVCAM-1 are raised <strong>in</strong> patients with ACS and are<strong>in</strong>versely related to plasma levels of NO


126 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>AtelectasisPa<strong>in</strong> due to rib andvertebral <strong>in</strong>farctionReduced respiratorydrive and coughTrue thromboembolismActivated endotheliumReduced red cell flexibilityIncreased red cell adherenceVaso-occlusionFigure 21.1cell disease.Acute chestsyndromeFat embolismBone marrow <strong>in</strong>farctionRelease of phospholipase A2InfectionChalmydia pneumoniaeMycoplasma pneumoniae/hom<strong>in</strong>isStaphylococcus aureusStreptococcus pneumoniae<strong>Respiratory</strong> syncytial virusParvovirusRh<strong>in</strong>ovirusInfluenza and para<strong>in</strong>fluenzavirusPathophysiology of the acute chest syndrome <strong>in</strong> sicklemetabolites. 11NO <strong>in</strong>hibits VCAM-1 expression, thus reducedendothelial synthesis of NO dur<strong>in</strong>g vaso-occlusive crises andhypoxia may contribute to red cell adhesion with<strong>in</strong> the lung. NOb<strong>in</strong>ds and <strong>in</strong>creases HbS avidity for oxygen both <strong>in</strong> vitro and <strong>in</strong>vivo, and hence may reduce its tendency for polymerisation 12and thus has a potential therapeutic role <strong>in</strong> ACS.NO is thought to play a significant role <strong>in</strong> the regulation ofhypoxic pulmonary vasoconstriction. Exhaled concentrationsof NO are directly related to the <strong>in</strong>haled concentration ofoxygen, 13suggest<strong>in</strong>g that oxygen may be a rate limit<strong>in</strong>gsubstrate for NO synthesis. Normal Hb has an aff<strong>in</strong>ity for NOthat is 3000 times that of oxygen, so Hb may act as a “biologicals<strong>in</strong>k” for NO 14 and rapid clearance of NO by Hb may contributeto hypoxic pulmonary vasoconstriction. 15 However, <strong>in</strong>the presence of anaemia there may be failure of hypoxic vasoconstrictiondue to <strong>in</strong>creased local levels of NO, despitereduced local production. Failure of hypoxic vasoconstrictionworsens ventilation-perfusion match<strong>in</strong>g, giv<strong>in</strong>g ideal conditionsfor further sickl<strong>in</strong>g and eventually ACS.Hydroxyurea has made a big impact on the management ofSCD. In a double bl<strong>in</strong>d, placebo controlled trial <strong>in</strong> 299 adultswith sickle cell anaemia who had at least three pa<strong>in</strong>ful episodes<strong>in</strong> the preced<strong>in</strong>g year, hydroxyurea reduced the frequency ofpa<strong>in</strong>ful episodes, the <strong>in</strong>cidence of ACS, and reduced the need forhospitalisation and transfusion. 16Hydroxyurea <strong>in</strong>creases theconcentration of fetal Hb (HbF) <strong>in</strong> erythrocytes, reduc<strong>in</strong>g thetendency for polymerisation of HbS. 17 18 However, some cl<strong>in</strong>icalimprovements are seen before the HbF concentration<strong>in</strong>creases. 19 In other studies hydroxyurea reduced adhesion ofsickle cells to the vascular endothelium <strong>in</strong> vitro by reduc<strong>in</strong>gVCAM-1 expression. 20Hydroxyurea is also oxidised by hemegroups to produce NO, 21 and <strong>in</strong>creased plasma NO metabolitescan be detected dur<strong>in</strong>g treatment. 22The beneficial effects ofhydroxyurea may therefore be mediated via its properties as anNO donor as well as its effects on HbF.<strong>Management</strong> of acute chest syndromeMost adult patients are admitted with vaso-occlusive crisesand develop ACS after a few days, whereas children are morelikely to have a preced<strong>in</strong>g febrile illness or <strong>in</strong>fection. 123 Oncethe process of lung <strong>in</strong>jury has started it may be difficult tostop, and thus the aim of management should be to preventACS. Although the causes, cl<strong>in</strong>ical presentation, and outcomesof ACS have been well documented, 1223 less is known about itsprevention or management.General managementThe most common cause for emergency admission of adultswith SCD is a vaso-occlusive crisis. The ma<strong>in</strong>stays of managementare pa<strong>in</strong> control, rehydration, oxygenation, and treatmentof any identifiable precipitat<strong>in</strong>g cause. Common precipitantsare <strong>in</strong>fection, cold, stress, hypoxia and dehydration, butvery often no obvious cause can be found. Infection—bothbacterial and viral—is more common <strong>in</strong> children.Adequate pa<strong>in</strong> control usually requires <strong>in</strong>itially high dosesof opiates given by subcutaneous or <strong>in</strong>tramuscular <strong>in</strong>jection.The aim is to control pa<strong>in</strong> rather than treat<strong>in</strong>g it as required. Itis not uncommon for the doses required to suppressrespiration and cough, caus<strong>in</strong>g atelectasis, reta<strong>in</strong>ed secretions,and hypoxaemia. On the other hand, <strong>in</strong>adequate pa<strong>in</strong> controlmay limit expansion and cough with the same consequences.In patients requir<strong>in</strong>g opiates, <strong>in</strong>travenous rehydration ispreferred aim<strong>in</strong>g for 3–4 litres a day as the patient may not beable to dr<strong>in</strong>k adequate amounts ow<strong>in</strong>g to pa<strong>in</strong> or drows<strong>in</strong>essfrom excessive analgesia. <strong>Care</strong> should be taken not to causefluid overload, especially <strong>in</strong> patients with impaired renal orcardiac function as a long term complication of SCD. Fluidoverload will exacerbate pulmonary oedema associated withlung <strong>in</strong>jury. Patients should be encouraged to dr<strong>in</strong>k freely.There is a trend to give oxygen rout<strong>in</strong>ely to all patients with avaso-occlusive crisis. However, if the oxygen saturations are normal(>97% on air), there is little to be ga<strong>in</strong>ed from supplementaloxygen although it may reduce sickl<strong>in</strong>g <strong>in</strong> poorly ventilatedareas of the lung. In patients with reduced respiratory drive orventilation-perfusion mismatch, oxygen saturations may bereassur<strong>in</strong>gly normal while the patient is on supplementaloxygen, mask<strong>in</strong>g the onset of acute lung <strong>in</strong>jury. We recommendthat pulse oximetry should be monitored regularly and allmeasurements should be performed after breath<strong>in</strong>g room air for10 m<strong>in</strong>utes. If there is a fall of more than a few percent fromnormal values (patients with SCD may have low basel<strong>in</strong>e levelsdue to chronic sickle lung disease), the cause should be soughtand treated aggressively to prevent progression to ACS.Specific management<strong>Respiratory</strong> <strong>in</strong>fection is a common precipitant of a sickle crisisbut pathogens are rarely detected. In one study an identifiablepathogen was isolated <strong>in</strong> just over 30% of episodes, 1 but thisfigure is dependent on how hard one looks. The two mostcommon organisms were Chlamydia pneumoniae and Mycoplasma(mostly M pneumoniae and occasionally M hom<strong>in</strong>is).Children suffered more <strong>in</strong>fections with respiratory syncytialvirus and parvovirus. Pneumococcus or Staphylococcus were lesscommon, even though most patients are hyposplenic. However,<strong>in</strong> the reported study patients with chlamydial <strong>in</strong>fectionswere less likely to be tak<strong>in</strong>g prophylactic antibiotics. Many ofthe cases of <strong>in</strong>fection also had evidence of marrow<strong>in</strong>farction. 1S<strong>in</strong>ce atypical organisms predom<strong>in</strong>ate, a strongcase can be made for treatment with macrolide antibiotics asthe first l<strong>in</strong>e treatment when <strong>in</strong>fection is thought to be thecause. However, caution should be exercised as the pattern of<strong>in</strong>fectious agents <strong>in</strong> the UK may be different and furtherstudies are required.About 25% of patients with ACS wheeze and may respondto bronchodilators. Obstructive spirometry and small airwaysdisease is not an uncommon f<strong>in</strong>d<strong>in</strong>g <strong>in</strong> patients with SCD. 1Thus, <strong>in</strong> patients on mechanical ventilation, high airway pressuremay be due to airway obstruction rather than reducedlung compliance, and gas trapp<strong>in</strong>g and <strong>in</strong>tr<strong>in</strong>sic positive endexpiratory pressure should be monitored. Rout<strong>in</strong>e use of<strong>in</strong>centive spirometry has recently been recommended toprevent atelectasis and ACS <strong>in</strong> patients with SCD admittedwith chest or bone pa<strong>in</strong>. 24 In this randomised control study on


Illustrative case 6: acute chest syndrome of sickle cell anaemia 12729 patients, 10 maximal <strong>in</strong>spirations with the <strong>in</strong>centive spirometerevery 2 hours while the patient was awake significantlyreduced the <strong>in</strong>cidence of pulmonary complications. We preferto use CPAP when the saturations fall below 93% on air orthere is atelectasis on the chest radiograph. This is becausepa<strong>in</strong> and opiate sedation limits effort and compliance withactive breath<strong>in</strong>g techniques.Blood transfusion and exchange transfusion are not requireddur<strong>in</strong>g uncomplicated pa<strong>in</strong>ful episodes, but may be necessarywhen haemolysis is severe, if a large amount of blood is sequestrated<strong>in</strong> the spleen, or if there is an aplastic crisis caused byparvovirus <strong>in</strong>fection. However, exchange transfusion candramatically alter the course of ACS by replac<strong>in</strong>g sickle cellswith those conta<strong>in</strong><strong>in</strong>g HbA and by improv<strong>in</strong>g anaemia. 25Inaddition, transfusion also rapidly improves oxygenation, whichsuggests that anaemia may lead to <strong>in</strong>creased local pulmonaryNO accumulation <strong>in</strong>creas<strong>in</strong>g shunt by counteract<strong>in</strong>g hypoxicvasoconstriction. 26 Whether exchange transfusion is superior tosimple transfusion is unclear, but <strong>in</strong> the US study of ACS bothwere effective with a low risk of alloimmunisation if phenotypicallymatched blood was used. 1 Our practice is to use exchangetransfusion aim<strong>in</strong>g for HbS


22 Illustrative case 7: assessment and management ofmassive haemoptysisJ L Lordan, A Gascoigne, P A Corris.............................................................................................................................Haemoptysis may be the present<strong>in</strong>g symptomof a number of diseases, 12with anassociated mortality rang<strong>in</strong>g from 7% to30%. 3–5 Although fewer than 5% of patientspresent<strong>in</strong>g with haemoptysis expectorate largevolumes of blood, the explosive cl<strong>in</strong>ical presentationand the unpredictable course of life threaten<strong>in</strong>ghaemoptysis demands prompt evaluation andmanagement. We have reviewed the aetiology ofmassive haemoptysis and alveolar haemorrhage,with particular reference to current diagnosticand therapeutic strategies.CASE HISTORYA 69 year old woman was an emergencyadmission with large volume haemoptysis whichdid not settle spontaneously. She had previouslyundergone a left mastectomy for breast carc<strong>in</strong>oma.Alveolar shadow<strong>in</strong>g was noted <strong>in</strong> the leftmid zone on the chest radiograph, consistent withrecent pulmonary haemorrhage (fig 22.1A). Athoracic computed thoracic (CT) scan confirmedconsolidation and volume loss <strong>in</strong> the left upperlobe and l<strong>in</strong>gula, but also showed a massanteriorly erod<strong>in</strong>g through the chest wall, consistentwith local recurrence of the breast neoplasm(fig 22.1B). Pulmonary angiography showed noabnormality, but bronchial angiography identifieda trunk that supplied a moderate pathologicalcirculation anteriorly <strong>in</strong> the left upper lobe <strong>in</strong> theregion of the abnormality on the CT scan. Theartery was successfully embolised us<strong>in</strong>g polyv<strong>in</strong>ylalcohol (PVA) foam granules (500–700 µm <strong>in</strong>diameter, fig 22.2). The <strong>in</strong>ternal mammary arterywas also catheterised and a pathological circulationwas noted that was occluded us<strong>in</strong>g plat<strong>in</strong>umcoils (fig 22.1A) and PVA granules, with no complicationsand no recurrence of haemoptysis.DEFINITIONAlthough there is no generally accepted def<strong>in</strong>itionof the volume of blood that constitutes a massivehaemoptysis, studies have quoted volumes rang<strong>in</strong>gfrom 100 ml up to or more than 1000 ml perday. 2 As the anatomical dead space of the majorairways is 100–200 ml, a more relevant def<strong>in</strong>itionof massive haemoptysis is the volume that is lifethreaten<strong>in</strong>g by virtue of airway obstruction orblood loss. 56AETIOLOGYIt is important to establish that the lung is thesource of bleed<strong>in</strong>g, <strong>in</strong> part by exclud<strong>in</strong>g thenasopharynx or gastro<strong>in</strong>test<strong>in</strong>al tract. The mostcommon causes of massive haemoptysis are listed<strong>in</strong> box 22.1. Haemoptysis orig<strong>in</strong>ates from thebronchial and pulmonary circulation <strong>in</strong> 90% and5% of cases, respectively. 7 Bleed<strong>in</strong>g from thebronchial arteries has the propensity to causemassive haemoptysis as it is a circulation atsystemic pressure. Alveolar haemorrhage is a recognisedcause of haemoptysis, but rarely causesmassive bleed<strong>in</strong>g as the alveoli have the capacityto accommodate a large volume of blood. 8 A morecommon presentation is mild haemoptysis, pulmonary<strong>in</strong>filtrates, and anaemia. 2Chronic <strong>in</strong>flammatory conditions (<strong>in</strong>clud<strong>in</strong>gbronchiectasis, tuberculosis, lung abscess) andlung malignancies are the most common causesof massive haemoptysis. 910Similarly, bleed<strong>in</strong>gmay occur from a mycetoma <strong>in</strong> the presence ofcavitat<strong>in</strong>g lung disease. 11 12 The concurrent developmentof haemoptysis and menstruation po<strong>in</strong>tsto a diagnosis of catamenial haemoptysis. Thepresence of haemoptysis and spontaneous pneumothorax<strong>in</strong> a woman of childbear<strong>in</strong>g age withdiffuse <strong>in</strong>terstitial abnormalities on the chestradiograph should raise the suspicion oflymphangioleiomyomatosis. 16The presence of a saddle nose, rh<strong>in</strong>itis, orperforated nasal septum may suggest a diagnosisof Wegener’s granulomatosis. 17Features of Behcet’sdisease <strong>in</strong>clude oral or genital ulceration,uveitis, cutaneous nodules, and pulmonary arteryaneurysm which is associated with a 30% 2 yearmortality rate. 18Although haematuria may bepresent <strong>in</strong> association with Goodpasture’s disease,5–10% of patients present without cl<strong>in</strong>icalevidence of renal disease. 8DIAGNOSTIC PROCEDURESSputum should be sent for microbiological <strong>in</strong>vestigation,<strong>in</strong>clud<strong>in</strong>g sta<strong>in</strong><strong>in</strong>g and culture for mycobacteria,and cytological exam<strong>in</strong>ation if thepatient is a smoker and over 40 years of age. Chestradiography may help to identify causative lesionsor <strong>in</strong>filtrates result<strong>in</strong>g from pulmonary haemorrhage,but fails to localise the lesion <strong>in</strong> 20–46% ofpatients with haemoptysis. 19 A CT scan may showsmall bronchial carc<strong>in</strong>omas or localisedbronchiectasis. 13 20 21 The use of contrast may helpto identify vascular abnormalities such as arteriovenousmalformations or aneurysms. 14 22 Despiteall <strong>in</strong>vestigative procedures, the aetiology of haemoptysisis unknown <strong>in</strong> up to 5–10% of patients. 7MANAGEMENT OF MASSIVEHAEMOPTYSISThe <strong>in</strong>itial approach to manag<strong>in</strong>g life threaten<strong>in</strong>ghaemorrhage <strong>in</strong>volves resuscitation and protect<strong>in</strong>gthe airway (fig 22.3), the second step isdirected at localis<strong>in</strong>g the site and cause of bleed<strong>in</strong>g,and the f<strong>in</strong>al step <strong>in</strong>volves the application ofdef<strong>in</strong>itive and specific treatments to preventrecurrent bleed<strong>in</strong>g.Airway protection and resuscitationAll patients with massive haemoptysis should bemonitored <strong>in</strong> an <strong>in</strong>tensive care unit (ICU) or highdependency unit (HDU) and the patient’s fitness


Illustrative case 7: assessment and management of massive haemoptysis 129for surgery established. Attempts should be made todeterm<strong>in</strong>e the side of bleed<strong>in</strong>g and the patient positioned withthe bleed<strong>in</strong>g side down to prevent aspiration <strong>in</strong>to theunaffected lung. Blood loss should be treated with volumeresuscitation, blood transfusion, and correction of coagulopathy.If large volume bleed<strong>in</strong>g cont<strong>in</strong>ues or the airway iscompromised, the patient’s trachea should be <strong>in</strong>tubated withas large an endotracheal tube as is possible to allow adequatesuction<strong>in</strong>g and access for bronchoscopy. 2 If the bleed<strong>in</strong>g canonly be localised to the right or left lung, unilateral lung <strong>in</strong>tubationmay protect the non-bleed<strong>in</strong>g lung. 23For right sidedbleed<strong>in</strong>g a bronchoscope may be directed <strong>in</strong>to the left ma<strong>in</strong>bronchus which can then be selectively <strong>in</strong>tubated over thebronchoscope with the patient ly<strong>in</strong>g <strong>in</strong> the right lateralposition (fig 22.4). The left lung is then protected from aspirationand selectively ventilated. For a left sided bleed<strong>in</strong>g sourcethe patient is placed <strong>in</strong> the left lateral position and selective<strong>in</strong>tubation of the right lung may be performed, but this maylead to occlusion of the right upper lobe bronchus. 2Analternative strategy is to pass an endotracheal tube over thebronchoscope <strong>in</strong>to the trachea. A Fogarty catheter (size 14French/100 cm length) may then be passed through the vocalcords beside the endotracheal tube, directed by the bronchoscope<strong>in</strong>to the left ma<strong>in</strong> bronchus and <strong>in</strong>flated (fig 22.5). Thisprevents aspiration of blood from the left lung and theendotracheal tube positioned <strong>in</strong> the trachea allows ventilationof the unaffected right lung.An alternative strategy for unilateral bleed<strong>in</strong>g is to pass adouble lumen endotracheal tube, which allows isolation andventilation of the normal lung and prevents aspiration fromthe side <strong>in</strong>volved by bleed<strong>in</strong>g (fig 22.6). 2However, <strong>in</strong>sert<strong>in</strong>gdouble lumen tubes should only be performed by experiencedoperators to avoid the serious consequences of poorposition<strong>in</strong>g. 24Figure 22.1 (A) Chest radiograph show<strong>in</strong>g previous leftmastectomy and left upper lobe and l<strong>in</strong>gular <strong>in</strong>filtrates due to airwaybleed<strong>in</strong>g. A plat<strong>in</strong>um embolisation coil is noted on thispost-embolisation radiograph (arrow).(B) CT scan show<strong>in</strong>g a masslesion (arrow) <strong>in</strong>volv<strong>in</strong>g the left anterior chest wall with associatedleft upper zone consolidation, consistent with recent haemoptysisand local tumour recurrence follow<strong>in</strong>g the previous mastectomy.Identify<strong>in</strong>g the site and cause of bleed<strong>in</strong>gPrecise localisation of the bleed<strong>in</strong>g site directs def<strong>in</strong>itive treatment.Fibreoptic bronchoscopy and angiography are themodalities of choice to localise the site of bleed<strong>in</strong>g and to allowtherapeutic <strong>in</strong>tervention, although the tim<strong>in</strong>g of bronchoscopyis controversial. 25 26 Early compared with delayedbronchoscopy gives a higher yield for localis<strong>in</strong>g the site ofbleed<strong>in</strong>g. 26 In contrast to mild haemoptysis, localisation of theAPre-embolisation bronchial angiogramBPost-embolisation bronchial angiogramRightLeftAbnormal circulationEmbolised abnormalleft circulationFigure 22.2 Use of selective bronchial artery embolisation to control massive haemoptysis. (A) Bronchial angiogram show<strong>in</strong>g common trunkand left sided abnormal circulation pre-embolisation, and (B) post-embolisation angiogram show<strong>in</strong>g the left bronchial artery and successfulembolisation of abnormal vessels.


130 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Box 22.1 Causes of massive haemoptysis andalveolar haemorrhageInfections• Mycobacteria, particularly tuberculosis• Fungal <strong>in</strong>fections (mycetoma)• Lung abscess• Necrotis<strong>in</strong>g pneumonia (Klebsiella, Staphylococcus, Legionella)Iatrogenic• Swan-Ganz catheterisation• Bronchoscopy• Transbronchial biopsy• Transtracheal aspirateParasitic• Hydatid cyst• ParagonimiasisTrauma• Blunt/penetrat<strong>in</strong>g <strong>in</strong>jury• Suction ulcers• Tracheoarterial fistulaNeoplasm• Bronchogenic carc<strong>in</strong>oma• Bronchial adenoma• Pulmonary metastases• SarcomaHaemoptysis <strong>in</strong> children• Bronchial adenoma• Foreign body aspiration• Vascular anomaliesVascular• Pulmonary <strong>in</strong>farct, embolism• Mitral stenosis• Arteriobronchial fistula• Arteriovenous malformations• Bronchial telangiectasia• Left ventricular failureCoagulopathy• Von Willebrand’s disease• Haemophilia• Anticoagulant therapy• Thrombocytopenia• Platelet dysfunction• Dissem<strong>in</strong>ated <strong>in</strong>travascular coagulationVasculitis• Behcet’s disease• Wegener’s granulomatosisPulmonary• Bronchiectasis (<strong>in</strong>clud<strong>in</strong>g cystic fibrosis)• Chronic bronchitis• Emphysematous bullaeMiscellaneous• Lymphangioleiomatosis• Catamenial (endometriosis)• Pneumoconiosis• Broncholith• IdiopathicSpurious• Epistaxis• Haematemesissite of bleed<strong>in</strong>g is essential <strong>in</strong> the management of massivehaemoptysis and urgent bronchoscopy should be considered. 7Fibreoptic bronchoscopy can be performed at the bedsideand allows visualisation of more peripheral and upper lobelesions, but has a limited suction capacity. 25 26 Rigid bronchoscopyprovides superior suction to ma<strong>in</strong>ta<strong>in</strong> airway patency,but it has a limited ability to identify peripheral lesions anddoes not permit good views of the upper lobes. 3 It is usuallyperformed under general anaesthetic but can be performedunder local anaesthesia and sedation <strong>in</strong> experienced hands. 27The techniques can be comb<strong>in</strong>ed when the fibreopticbronchoscope is passed through the lumen of the rigid bronchoscope.Bronchoscopic treatmentInstillation of ep<strong>in</strong>ephr<strong>in</strong>e (1:20 000) is advocated to controlbleed<strong>in</strong>g, although its efficacy <strong>in</strong> life threaten<strong>in</strong>g haemoptysisis uncerta<strong>in</strong>. 2 The topical application of thromb<strong>in</strong> andthromb<strong>in</strong>-fibr<strong>in</strong>ogen solutions has also had some success, butfurther study is required before widespread use can berecommended. 28In massive haemoptysis, isolation of a bleed<strong>in</strong>g segmentwith a balloon catheter may prevent aspiration of blood <strong>in</strong>tothe large airways, thereby ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g airway patency andoxygenation. Hav<strong>in</strong>g identified the segmental bronchus that isthe source of bleed<strong>in</strong>g, the bronchoscope is wedged <strong>in</strong> the orifice.A size 4–7 Fr 200 cm balloon catheter is passed throughthe work<strong>in</strong>g channel of the bronchoscope and the balloon is<strong>in</strong>flated <strong>in</strong> the affected segment, isolat<strong>in</strong>g the bleed<strong>in</strong>g site (fig7). 2 A double lumen balloon catheter (6 Fr, 170 cm long) witha detachable valve at the proximal end has recently beendesigned that passes through the bronchoscope channel andallows the removal of the bronchoscope without anymodification of the catheter. 29 The second channel of the cathetermay also be used to <strong>in</strong>stil vasoactive drugs to help controlbleed<strong>in</strong>g. The bronchoscope can then be removed over thecatheter, which is left <strong>in</strong> place for 24 hours. The balloon may bedeflated under controlled conditions with bronchoscopic visualisationand the catheter removed if the bleed<strong>in</strong>g has stopped.The prolonged use of balloon tamponade catheters should beavoided to prevent ischaemic mucosal <strong>in</strong>jury and postobstructivepneumonia. Endobronchial tamponade shouldonly be applied as a temporary measure until a more def<strong>in</strong>itivetherapeutic procedure can be deployed.Neodymium-yttrium-alum<strong>in</strong>ium-garnet (Nd-YAG) laserphotocoagulation has been used with some success <strong>in</strong> themanagement of massive haemorrhage associated with directlyvisualised endobronchial lesions. 30 However, target<strong>in</strong>g the culpritvessel with the laser beam can be difficult <strong>in</strong> the presenceof ongo<strong>in</strong>g bleed<strong>in</strong>g.Bronchial artery embolisation (BAE)This was first reported by Remy and colleagues <strong>in</strong> 1977 31 andis <strong>in</strong>creas<strong>in</strong>gly used <strong>in</strong> the management of life threaten<strong>in</strong>ghaemoptysis. 20 The procedure <strong>in</strong>volves the <strong>in</strong>itial identificationof the bleed<strong>in</strong>g vessel by selective bronchial artery cannulation,and the subsequent <strong>in</strong>jection of particles (polyv<strong>in</strong>yl alcoholfoam, isobutyl-2-cyanoacrylate, Gianturco steel coils orabsorbable gelat<strong>in</strong> pledgets) <strong>in</strong>to the feed<strong>in</strong>g vessel (fig 22.2).A number of features provide clues to the bronchial artery asthe source of bleed<strong>in</strong>g, <strong>in</strong>clud<strong>in</strong>g the <strong>in</strong>frequent identificationof extravasated dye or the visualisation of tortuous vessels of<strong>in</strong>creased calibre or aneurysmal dilatation. 32The immediatesuccess rates for control of massive haemoptysis is excellent,rang<strong>in</strong>g from 64% to 100%, although recurrent non-massivebleed<strong>in</strong>g has been reported <strong>in</strong> 16–46% of patients. 32–35 Technicalfailure of BAE occurs <strong>in</strong> up to 13% of cases and is largelycaused by non-bronchial artery collaterals from systemic vesselssuch as the phrenic, <strong>in</strong>tercostal, mammary, or subclavianarteries. 35Complications of BAE <strong>in</strong>clude vessel perforation,


Illustrative case 7: assessment and management of massive haemoptysis 131Massive haemoptysisInvestigationsFBC, U&Es, COAG,ABG, CXR,group & crossmatch*Admit to HDU/ITU<strong>Respiratory</strong> consultResuscitationOxygen supplementationCorrect any coagulopathyConsider tranexamic acidUnstable patientIntubationTransfusionThoracic surgery consultEarly bronchoscopySuspectedpulmonary embolusSp<strong>in</strong>al CT angiogramStable patientCT thoraxBleed<strong>in</strong>g sitelocalisedBleed<strong>in</strong>g sitenot localisedBronchoscopyEndobronchialtamponadeAngiographgyInvestigation for<strong>in</strong>terstitial lung diseaseGoodpasture’s, vasculitisInterstitialreticularpatternBleed<strong>in</strong>g sitelocalisedBleed<strong>in</strong>g sitenot localisedSputum culture, AFBfungal cultureInfiltrateConservativemanagementTB, aspergilloma, abscessAppropriate antibioticsCavityNodule orcystic lesionBronchial arteryembolisation orsurgery if <strong>in</strong>dicatedPersistentbleed<strong>in</strong>gBronchoscopy, <strong>in</strong>stillationof antifungal agents, as<strong>in</strong>dicatedFigure 22.3malignancy.Algorithm for management of massive haemoptysis. *Palliative measures may be appropriate <strong>in</strong> the sett<strong>in</strong>g of advanced<strong>in</strong>timal tears, chest pa<strong>in</strong>, pyrexia, haemoptysis, systemicembolisation, and neurological complications. When the anteriorsp<strong>in</strong>al artery is identified as orig<strong>in</strong>at<strong>in</strong>g from thebronchial artery, embolisation is often deferred ow<strong>in</strong>g to therisk of <strong>in</strong>farction and paraparesis. 32The development andapplication of coaxial microcatheter systems allows moreselective catheterisation and embolisation of branches of thebronchial arteries, thereby reduc<strong>in</strong>g the risk of occlud<strong>in</strong>gbranches such as the anterior sp<strong>in</strong>al artery. 34Selective left lungventilationCuffed endotracheal tubepositioned <strong>in</strong> tracheaEndotracheal tubepositioned <strong>in</strong> leftma<strong>in</strong>stem bronchusInflated cuffof endotrachealtubeFogarty catheter passed asan endobronchial blockerRight sidedairway filledwith bloodBloodFigure 22.4 Selective <strong>in</strong>tubation of left ma<strong>in</strong> bronchus <strong>in</strong> a case ofright sided massive haemoptysis.Figure 22.5 Control of left sided massive haemoptysis by tracheal<strong>in</strong>tubation, placement, and <strong>in</strong>flation of a Fogarty catheter <strong>in</strong> the leftma<strong>in</strong> bronchus.


132 <strong>Respiratory</strong> <strong>Management</strong> <strong>in</strong> <strong>Critical</strong> <strong>Care</strong>Double lumenendotracheal tubeTracheal lumenVentilationSuctionInflated trachealcuffInflatedbronchial cuffBronchiallumenFigure 22.6 Application of a double lumen endotracheal tube forthe control of massive haemorrhage. The bronchial lumen ispositioned <strong>in</strong> the left ma<strong>in</strong> bronchus to ventilate the left lung and thetracheal lumen is positioned above the car<strong>in</strong>a, allow<strong>in</strong>g ventilation ofthe right lung while prevent<strong>in</strong>g occlusion of the right upper lobeorifice.Surgical managementSurgery is considered for the management of localised lesions.Surgical mortality ranges from 1% to 50% <strong>in</strong> different seriesdepend<strong>in</strong>g on selection criteria, but bias <strong>in</strong> the selection ofcandidates for surgery limits a direct comparison with medicaltreatment. 2 Surgery is contra<strong>in</strong>dicated <strong>in</strong> patients with<strong>in</strong>adequate respiratory reserve or those with <strong>in</strong>operable lungcancer due to direct thoracic spread. Surgical resection is <strong>in</strong>dicatedwhen BAE is unavailable or the bleed<strong>in</strong>g is unlikely to becontrolled by embolisation. It rema<strong>in</strong>s the treatment of choicefor the management of life threaten<strong>in</strong>g haemoptysis due to aleak<strong>in</strong>g aortic aneurysm, selected cases of arteriovenous malformations,hydatid cyst, iatrogenic pulmonary rupture, chest<strong>in</strong>juries, bronchial adenoma, or haemoptysis related tomycetoma resistant to other treatments. 723 Pulmonary arteryrupture related to the use of pulmonary artery catheters maybe temporarily controlled by withdraw<strong>in</strong>g the catheter slightlyand re<strong>in</strong>flat<strong>in</strong>g the balloon to compress the bleed<strong>in</strong>g vesselmore proximally. 36 However, surgical resection of the bleed<strong>in</strong>gvessel is the def<strong>in</strong>itive management.Fogarty catheter can bepassed via suction channelof fibreoptic bronchoscopeor rigid bronchoscope and<strong>in</strong>flated <strong>in</strong> a segmentalbronchus to isolate thesource of bleed<strong>in</strong>gBleed<strong>in</strong>g source localisedto segmental orificeFigure 22.7 Placement of a Fogarty catheter guided by fibreopticbronchoscopy to control massive bleed<strong>in</strong>g from a segmentalbronchus.The onset of massive haemoptysis <strong>in</strong> a patient with atracheostomy may be associated with the development of atracheal-arterial fistula, usually the <strong>in</strong>nom<strong>in</strong>ate artery. 37 Theprompt application of anterior and downward pressure on thetracheal cannula and over<strong>in</strong>flation of the tracheostomyballoon may help to tamponade the bleed<strong>in</strong>g vessel, andimmediate surgical review should be requested. Deflation ofthe tracheostomy balloon and removal of the tracheal cannulashould be performed <strong>in</strong> a controlled environment.Other treatmentThe oral antifibr<strong>in</strong>olytic agent tranexamic acid, an <strong>in</strong>hibitor ofplasm<strong>in</strong>ogen activation, is frequently used to control recurrenthaemoptysis. Intravenous vasopress<strong>in</strong> has also been used butcaution is advised <strong>in</strong> patients with coexistent coronary arterydisease or hypertension. Vasoconstriction of the bronchialartery may also hamper effective BAE by obscur<strong>in</strong>g the site ofbleed<strong>in</strong>g, lead<strong>in</strong>g to difficulties <strong>in</strong> cannulation of the artery. 2Systemic antifungal agents have been tried <strong>in</strong> the managementof haemoptysis related to mycetoma, but the resultshave been poor. By contrast, the direct <strong>in</strong>stillation ofantifungal drugs such as amphoteric<strong>in</strong> B with or withoutN-acetylcyste<strong>in</strong>e or iod<strong>in</strong>e by means of a percutaneous ortransbronchial catheter <strong>in</strong> the cavity has resulted <strong>in</strong> satisfactorycontrol of haemoptysis <strong>in</strong> some cases. 12 38 This techniqueshould be considered <strong>in</strong> patients with ongo<strong>in</strong>g bleed<strong>in</strong>gfollow<strong>in</strong>g attempted BAE who are not otherwise fit for surgicalresection.Invasive therapeutic procedures have no role <strong>in</strong> themanagement of pulmonary haemorrhage related to coagulopathy,blood dyscrasias, or immunologically mediated alveolarhaemorrhage. Appropriate medical treatment is usuallysufficient. 39On the rare occasion when an immunologicallymediated alveolar haemorrhage leads to massive haemoptysis,the adm<strong>in</strong>istration of systemic corticosteroids, cytotoxicagents, or plasmapheresis may be useful. 8The long termadm<strong>in</strong>istration of danazol or gonadotroph<strong>in</strong> releas<strong>in</strong>g hormoneagonists may prove useful <strong>in</strong> the management ofcatamenial haemoptysis. 40 Radiation therapy has been used <strong>in</strong>the management of massive haemoptysis associated with vasculartumours or mycetoma by <strong>in</strong>duc<strong>in</strong>g necrosis of feed<strong>in</strong>gblood vessels and vascular thrombosis due to perivascularoedema. 41OUTCOMEMortality has been closely correlated with the volume of bloodexpectorated, the rate of bleed<strong>in</strong>g, the amount of bloodreta<strong>in</strong>ed with<strong>in</strong> the lungs and premorbid respiratory reserve,<strong>in</strong>dependent of the aetiology of bleed<strong>in</strong>g. 14 The mortality rateis 58% when the rate of blood loss exceeds 1000 ml/24 hours,compared with 9% if bleed<strong>in</strong>g is less than 1000 ml/hour. 739 Themortality rate <strong>in</strong> patients with malignancy is 59%, which<strong>in</strong>creases to 80% <strong>in</strong> the presence of a comb<strong>in</strong>ation ofmalignant aetiology and a bleed<strong>in</strong>g rate of more than1000 ml/24 hours. A better outcome has been noted formassive haemorrhage due to bronchiectasis, lung abscess, ornecrotis<strong>in</strong>g pulmonary <strong>in</strong>fections, with a mortality rate of lessthan 1% <strong>in</strong> some series. 39SUMMARYThe unpredictable and potentially lethal course of massivehaemoptysis requires prompt resuscitation, airway protectionand correction of coagulopathy. Early <strong>in</strong>vestigation with bronchoscopyis recommended for localisation and control ofbleed<strong>in</strong>g by the application of topical adrenal<strong>in</strong>e, balloon tamponadeor selective lung <strong>in</strong>tubation. There is <strong>in</strong>creas<strong>in</strong>gacceptance of bronchial artery embolisation as the treatmentof choice to control acute massive haemoptysis that cont<strong>in</strong>uesdespite conservative treatment when a bronchial artery can beidentified as the source of bleed<strong>in</strong>g. Surgical resection rema<strong>in</strong>s


Illustrative case 7: assessment and management of massive haemoptysis 133the treatment of choice for particular conditions, where thebleed<strong>in</strong>g site is localised and the patient is fit for lungresection.ACKNOWLEDGEMENTSThe authors thank Dr Leslie Mitchell for provid<strong>in</strong>g the radiographicand bronchial embolisation images and Dr Stephen Cook and JohnSternman for review<strong>in</strong>g the manuscript.REFERENCES1 Cahill BC, Ingbar DH. Massive hemoptysis. Assessment andmanagement. Cl<strong>in</strong> Chest Med 1994;15:147–67.2 Dweik RA, Stoller JK. Role of bronchoscopy <strong>in</strong> massive hemoptysis. Cl<strong>in</strong>Chest Med 1999;20:89–105.3 Conlan AA, Hurwitz SS, Krige L, et al. Massive hemoptysis. Review of123 cases. J Thorac Cardiovasc Surg 1983;85:120–4.4 Yeoh CB, Hubaytar RT, Ford JM, et al. Treatment of massive hemorrhage<strong>in</strong> pulmonary tuberculosis. J Thorac Cardiovasc Surg 1967;54:503–10.5 Holsclaw DS, Grand RJ, Shwachman H. Massive hemoptysis <strong>in</strong> cysticfibrosis. J Pediatr 1970;76:829–38.6 Garzon AA, Cerruti MM, Gold<strong>in</strong>g ME. Exsangu<strong>in</strong>at<strong>in</strong>g hemoptysis. JThorac Cardiovasc Surg 1982;84:829–33.7 Jean-Baptiste E. Cl<strong>in</strong>ical assessment and management of massivehemoptysis. Crit <strong>Care</strong> Med 2000;28:1642–7.8 Schwarz MI, Brown KK. Small vessel vasculitis of the lung. Thorax2000;55:502–10.9 Santiago S, Tobias J, Williams AJ. A reappraisal of the causes ofhemoptysis. Arch Intern Med 1991;151:2449–51.10 Hirshberg B, Biran I, Glazer M, et al. Hemoptysis: etiology, evaluation,and outcome <strong>in</strong> a tertiary referral hospital. Chest 1997;112:440–4.11 Jewkes J, Kay PH, Paneth M, et al. Pulmonary aspergilloma: analysis ofprognosis <strong>in</strong> relation to haemoptysis and survey of treatment. Thorax1983;38:572–8.12 Rumbak M, Kohler G, Eastrige C, et al. Topical treatment of lifethreaten<strong>in</strong>g haemoptysis from aspergillomas. Thorax 1996;51:253–5.13 McGu<strong>in</strong>ness G, Beacher JR, Hark<strong>in</strong> TJ, et al. Hemoptysis: prospectivehigh-resolution CT/bronchoscopic correlation. Chest 1994;105:1155–62.14 Ference BA, Shannon TM, White RI Jr, et al. Life-threaten<strong>in</strong>g pulmonaryhemorrhage with pulmonary arteriovenous malformations and hereditaryhemorrhagic telangiectasia. Chest 1994;106:1387–90.15 Kalra S, Bell MR, Rihal CS. Alveolar hemorrhage as a complication oftreatment with abciximab. Chest 2001;120:126–31.16 Johnson S. Rare diseases—1: Lymphangioleiomyomatosis: cl<strong>in</strong>icalfeatures, management and basic mechanisms. Thorax 1999;54:254–64.17 Langford CA, Hoffman GS. Rare diseases—3: Wegener’sgranulomatosis. Thorax 1999;54:629–37.18 Erkan F, Gul A, Tasali E. Pulmonary manifestations of Behcet’s disease.Thorax 2001;56:572–8.19 Marshall TJ, Flower CD, Jackson JE. The role of radiology <strong>in</strong> the<strong>in</strong>vestigation and management of patients with haemoptysis. Cl<strong>in</strong> Radiol1996;51:391–400.20 Haponik EF, Fe<strong>in</strong> A, Ch<strong>in</strong> R. Manag<strong>in</strong>g life-threaten<strong>in</strong>g hemoptysis: hasanyth<strong>in</strong>g really changed? Chest 2000;118:1431–5.21 Millar AB, Boothroyd AE, Edwards D, et al. The role of computedtomography (CT) <strong>in</strong> the <strong>in</strong>vestigation of unexpla<strong>in</strong>ed haemoptysis. RespirMed 1992;86:39–44.22 Penn<strong>in</strong>gton DW, Gold WM, Gordon RL, et al. Treatment of pulmonaryarteriovenous malformations by therapeutic embolization. Rest andexercise physiology <strong>in</strong> eight patients. Am Rev Respir Dis1992;145:1047–51.23 Gour<strong>in</strong> A, Garzon AA. Control of hemorrhage <strong>in</strong> emergency pulmonaryresection for massive hemoptysis. Chest 1975;68:120–1.24 Kle<strong>in</strong> U, Karzai W, Bloos F, et al. Role of fiberoptic bronchoscopy <strong>in</strong>conjunction with the use of double-lumen tubes for thoracic anesthesia: aprospective study. Anesthesiology 1998;88:346–50.25 Gong H Jr, Salvatierra C. Cl<strong>in</strong>ical efficacy of early and delayedfiberoptic bronchoscopy <strong>in</strong> patients with hemoptysis. Am Rev Respir Dis1981;124:221–5.26 Saumench J, Escarrabill J, Padro L, et al. Value of fiberopticbronchoscopy and angiography for diagnosis of the bleed<strong>in</strong>g site <strong>in</strong>hemoptysis. Ann Thorac Surg 1989;48:272–4.27 Helmers RA, Sanderson DR. Rigid bronchoscopy. The forgotten art. Cl<strong>in</strong>Chest Med 1995;16:393–9.28 Tsukamoto T, Sasaki H, Nakamura H. Treatment of hemoptysis patientsby thromb<strong>in</strong> and fibr<strong>in</strong>ogen-thromb<strong>in</strong> <strong>in</strong>fusion therapy us<strong>in</strong>g a fiberopticbronchoscope. Chest 1989;96:473–6.29 Freitag L, Tekolf E, Stamatis G, et al. Three years experience with a newballoon catheter for the management of haemoptysis. Eur Respir J1994;7:2033–7.30 Edmondstone WM, Nanson EM, Woodcock AA, et al. Life threaten<strong>in</strong>ghaemoptysis controlled by laser photocoagulation. Thorax1983;38:788–9.31 Remy J, Arnaud A, Fardou H, et al. Treatment of hemoptysis byembolization of bronchial arteries. Radiology 1977;122:33–7.32 Uflacker R, Kaemmerer A, Picon PD, et al. Bronchial artery embolization<strong>in</strong> the management of hemoptysis: technical aspects and long-termresults. Radiology 1985;157:637–44.33 Br<strong>in</strong>son GM, Noone PG, Mauro MA, et al. Bronchial arteryembolization for the treatment of hemoptysis <strong>in</strong> patients with cysticfibrosis. Am J Respir Crit <strong>Care</strong> Med 1998;157:1951–8.34 Tanaka N, Yamakado K, Murashima S, et al. Superselective bronchialartery embolization for hemoptysis with a coaxial microcatheter system. JVasc Interv Radiol 1997;8:65–70.35 Keller FS, Rosch J, Lofl<strong>in</strong> TG, et al. Nonbronchial systemic collateralarteries: significance <strong>in</strong> percutaneous embolotherapy for hemoptysis.Radiology 1987;164:687–92.36 Thomas R, Siproudhis L, Laurent JF, et al. Massive hemoptysis fromiatrogenic balloon catheter rupture of pulmonary artery: successful earlymanagement by balloon tamponade. Crit <strong>Care</strong> Med 1987;15:272–3.37 Schaefer OP, Irw<strong>in</strong> RS. Tracheoarterial fistula: an unusual complicationof tracheostomy. J Intensive <strong>Care</strong> Med 1995;10:64–75.38 Shapiro MJ, Albelda SM, Mayock RL, et al. Severe hemoptysisassociated with pulmonary aspergilloma. Percutaneous <strong>in</strong>tracavitarytreatment. Chest 1988;94:1225–31.39 Corey R, Hla KM. Major and massive hemoptysis: reassessment ofconservative management. Am J Med Sci 1987;294:301–9.40 Matsubara K, Ochi H, Ito M. Catamenial hemoptysis treated with along-act<strong>in</strong>g GnRH agonist. Int J Gynaecol Obstet 1998;60:289–90.41 Shneerson JM, Emerson PA, Phillips RH. Radiotherapy for massivehaemoptysis from an aspergilloma. Thorax 1980;35:953–4.


134Index ..........................................................................................................Page numbers <strong>in</strong> bold text refer to figures <strong>in</strong> the text; those <strong>in</strong> italics to tables or boxed materialacetylchol<strong>in</strong>esterase (Ach) antibody 118acetylcyste<strong>in</strong>e 90acid maltase deficiency 117acidosis, respiratory 61, 81, 82, 89activated prote<strong>in</strong> C, recomb<strong>in</strong>ant human 33acute lung <strong>in</strong>jury (ALI)def<strong>in</strong>itions 32, 94follow<strong>in</strong>g lung resection 102–4acute respiratory distress syndrome (ARDS)cl<strong>in</strong>ical trials 33conditions mimick<strong>in</strong>g/caus<strong>in</strong>g 3def<strong>in</strong>itions 31–3, 94epidemiology and risk factors 33–5<strong>in</strong>vestigations 5–8mortality 57, 66non-ventilatory management 66–71pathogenesis 39–41, 60pathological features 38–9pulmonary circulation 41, 93–4resolution 41–2ventilatory management 57, 60–3acute sickle chest syndrome (ACS) 125–7adhesion molecules 40, 125–6adrenal<strong>in</strong>e (ep<strong>in</strong>ephr<strong>in</strong>e) 82, 88Adult/Adolescent Spectrum of HIV DiseaseCohort Study 123“adult respiratory distress syndrome” 31air embolism 99“air hunger” 83airway pressure 52–3airway pressure release ventilation (APRV) 63alcohol abuse 35almitr<strong>in</strong>e 67alprostadil 67altitude acclimatisaton 15ALVEOLI study 61American Thoracic Society 24am<strong>in</strong>oglycosides 107am<strong>in</strong>ophyll<strong>in</strong>e 88amphoteric<strong>in</strong> B 132anaemia, autoimmune haemolytic 117–18anaerobic metabolism 16–17anaesthesia, regional 101anaesthetic agents 89, 90–1angiotens<strong>in</strong> convert<strong>in</strong>g enzyme (ACE) gene76antifungal agents, systemic 132antimicrobials 24–5, 26–8, 107ant<strong>in</strong>eutrophil cytoplasmic antibodies(ANCA) 114–16antioxidants 71antiretroviral therapy 48, 123aorta, <strong>in</strong>jury 100apoptosis 40, 42ARDSNet study 57, 61, 62ARDS Network 32, 33arg<strong>in</strong><strong>in</strong>e 70arrhythmias 88arterial oxygen saturation 60–1arterial partial pressure:<strong>in</strong>spired oxygenconcentration ratio (Pa02:Fi02) 8, 31, 32,33, 34ascorbic acid 71, 102aspergillosis 5asphyxia 101assisted ventilatory support 77, 83–4, 88, 89asthma, acute severeassessment 86–7assisted ventilation 88–90drug treatment 87–8mortality 86therapy of non-respond<strong>in</strong>g patient 90–1atelectasis 102, 125, 126, 126bibasal 117, 118, 119atelectrauma 53atovaquone 122atrial septostomy 95autoimmune haemolytic anaemia 117–18azathiopr<strong>in</strong>e 110, 115β agonists 86, 87 8bag-valve-mask ventilation 50balloon catheter 130barbiturate drugs 89Behçet s disease 128bi-level pressure support 83biotrauma 56biphasic airway pressure (BiPAP) 63blood-gas barrier 93blood transfusion 11, 127bone marrow transplant 5Boston Coconut Grove fire 101breast neoplasm 128“breath stack<strong>in</strong>g” 86, 87British Thoracic Society 3, 19–20, 21bronchial arteryembolisation 107, 128, 129, 130–1, 132haemorrhage 128–30bronchial hyperreactivity 101bronchial <strong>in</strong>tubation 129, 131bronchial secretions 82, 84analysis 25, 26bronchoalveolar lavage (BAL) 4, 5, 25–6, 111,125bronchoconstriction 86bronchodilators 87–8, 126bronchopulmonary dysplasia 57bronchoscopy 3–5haemoptysis 129–30, 132<strong>in</strong>halation <strong>in</strong>jury 102SARS <strong>in</strong>fection 46<strong>in</strong> ventilated patient 4bronchosegmental lavage 69, 70Burkholderia cepacia 106, 107Burkholderia pseudomallei 20burnsmortality 101see also <strong>in</strong>halation <strong>in</strong>jurycalcium channel blockers 95capillary oxygen tension 14, 15carbon dioxide exchange 53, 61carbon monoxide poison<strong>in</strong>g 102carboxyhaemoglob<strong>in</strong> 102carboxypenicill<strong>in</strong>s 107cardiac output, determ<strong>in</strong>ation 93cardiac tamponade 82, 100cardiovascular collapse 82cast formation 102catamenial haemoptysis 128, 132cavitat<strong>in</strong>g lung disease 128CD4 lymphocyte count 120, 121, 123ceftazidime 27, 28, 107cellular metabolism 16–17central respiratory drive 76central venous catheter 6cephalospor<strong>in</strong>s 27, 107cerebral venous hypertension 94chemical <strong>in</strong>jury 102chest compression, manual 90chest radiograph 5–6ARDS 32massive haemoptysis 129post-pulmonary resection <strong>in</strong>jury 103SARS 47Chlamydia pneumoniae 126chronic obstructive pulmonary disease(COPD) 80–4decisions to admit to ICU 80non-ventilatory treatment 84outcome of ICU care 80recognition of ventilatory support need 81ventilatory support 81–4wean<strong>in</strong>g failure 77, 84Churg-Straus syndrome 5, 115ciprofloxac<strong>in</strong> 28cl<strong>in</strong>damyc<strong>in</strong> 122collagen 39community acquired pneumonia 19–22assessment of severity 19–20comorbidity 20management guidel<strong>in</strong>es 19microbiological <strong>in</strong>vestigations/diagnosis4–5, 20–1outcome and prognosis 22treatment 21–2compensatory anti-<strong>in</strong>flammatory responsesyndrome (CARS) 38compliance 8, 53, 60Comprehensive <strong>Critical</strong> <strong>Care</strong> (DoH) 2, 20computed tomography (CT) 6–8high resolution 110–11spiral angiography 94connective tissue disease 110–12cont<strong>in</strong>uous positive airway pressure (CPAP)22, 62, 88, 127controlled mechanical ventilation 82–3pressure 61–2, 82–3volume 82COPD see chronic obstructive pulmonarydiseasecoronaviruses 45corticosteroids 49, 68–9, 84, 87, 102, 110, 121,122co-trimoxazole 107, 110, 114, 120–1, 122cough 82cough assist devices 84cricoid pressure 82critical illness, classification 2CROP <strong>in</strong>dex 75cyclophosphamide 110, 115cystic fibrosis 106–9management 107–8non-pulmonary pathologies 108patient selection for ICU care 106–7cytok<strong>in</strong>es 40, 41, 56cytomegalovirus (CMV) <strong>in</strong>fection 5, 110, 111,121, 123cytoplasmic ant<strong>in</strong>eutrophil cytoplasmicantibodies (C-ANCA) 115danazol 132dapsone 122dead space, pulmonary 36deoxyribonuclease (DNase), recomb<strong>in</strong>ant 90,107dermatomyositis, Jo-1 negative 110–12detection of early antigen fluorescent foci(DEAFF) test 110detection of early antigen fluorescent foci(DEAFF) 5diabetes mellitus 35, 108diaphragm strength 76, 118–19measurement 8, 75diffuse <strong>in</strong>terstitial lung disease (DILD)110–122,3-diphosphoglycerate (2, 3 DPG) 14dobutam<strong>in</strong>e 14dopexam<strong>in</strong>e hydrochloride 14


Index 135drug resistance 25, 26–7, 28, 123Duchenne muscular dystrophy 117echocardiography 94, 96, 100transoesophageal 100eformoterol 88eicosanoids 70eicosapentaenoic acid 70electrocardiogram (ECG) 87, 100electrolyte disturbances 75embolectomy, surgical 96embolisation, bronchial artery 107, 128, 129,130–1empyema 100endobronchial suction 107endothelial <strong>in</strong>tegrity, ischaemia-reperfusion<strong>in</strong>jury 104endothelium, vascular 14endotracheal <strong>in</strong>tubationasthma 88–9COPD 82haemoptysis 129, 131, 132SARS patient 49thoracic trauma 99endotracheal tube 6enoximone 95epidural analgesia 101ep<strong>in</strong>ephr<strong>in</strong>e 130eschar, chest wall 102etomidate 89European Human Rights Act 80European Vasculitis Study Group 115EuroSIDA study 123exchange transfusion 127exhaustive breath<strong>in</strong>g 76extracorporeal carbon dioxide removal(ECCOR) 64, 95extracorporeal membrane oxygenation(ECMO) 64, 95, 116extravascular lung water 66extubation 83–4, 90–1see also wean<strong>in</strong>gface mask 81–2fat metabolism 125fatty acids 70fentanyl 89fibrosis, ARDS 39, 41, 42filters, ventilator 49–50flail chest 100–1flavonoids 71flow trigger<strong>in</strong>g 77, 83flucloxacill<strong>in</strong> 107fluconazole 114fluid management 66, 126Fogarty catheter 129, 131, 132forced expiratory volume <strong>in</strong> 1 second (FEV1)86foscarnet 121free radicals 40“frostbitten phrenic” 119gadol<strong>in</strong>ium 56gamma-l<strong>in</strong>olenic acid 70gancyclovir 110, 120, 121gastrograf<strong>in</strong> enema 108glutam<strong>in</strong>e 70glutathione 71, 102goal directed therapy 13gonadotroph<strong>in</strong> releas<strong>in</strong>g hormone agonists132Goodpasture’s disease 128Gram negative enterobacteria 28great vessel <strong>in</strong>jury 100Guilla<strong>in</strong> Barré syndrome 117haemoglob<strong>in</strong> 11, 125, 126haemoptysis, massive 106, 107, 128–32haemorrhagealveolar 4, 114, 115, 130bronchial artery 128–30thoracic trauma 100haemothorax 100halothane 90Harefield Hospital 107heart failure 77helium 91herpar<strong>in</strong>, aerosolised 102high frequency jet ventilation (HFJV) 50,63–4high frequency oscillatory ventilation (HFOV)49–50, 53, 63–4highly active antiretroviral therapy (HAART)123HIV 111, 120–4human rights 80humidification, <strong>in</strong>spired gas 90hydrocortisone 87hydrogen peroxide, exhaled 103hydroyurea 126hyperbaric oxygen therapy 102hypercapnia 53, 61, 89hyper<strong>in</strong>flation 77, 82, 86, 87hypertension, systemic venous 94hypokalaemia 87hypophosphataemia 14hypothyroidism 118hypoxiacellular response 16–17tissues/organs 13–16hypoxia tolerance 14hypoxic pulmonary vasoconstriction 126iatrogenic <strong>in</strong>juries 6, 132imidazole 89immunonutrition 70immunosuppression 5, 111, 115, 116<strong>in</strong>centive spirometry 126–7<strong>in</strong>fection control 49, 50–1<strong>in</strong>flammatory mediators 40, 56<strong>in</strong>halation <strong>in</strong>jury 101–2<strong>in</strong>otropes 82, 95<strong>in</strong>spiratory time 62<strong>in</strong>tensive care medic<strong>in</strong>e 1–2<strong>in</strong>tensive care unit, efficacy 1<strong>in</strong>terdependence 55<strong>in</strong>terferon 49<strong>in</strong>terleuk<strong>in</strong>-8 40<strong>in</strong>termediate sp<strong>in</strong>al muscular atrophy 117International Society of Heart LungTransplantation 112<strong>in</strong>terstitial lung disease 110–12ipratropium bromide 88iron metabolism 103irritant gases 101ischaemia-reperfusion <strong>in</strong>jury 103–4isoflurane 90isoniazid 114ketam<strong>in</strong>e 89ketoconazole 70lactate, blood concentration 15–16lactate dehydrogenase (LDH) 46lactic acidosis 87, 88latency associated peptide 41left ventricular performance 77legionella pneumonia 20leukotriene <strong>in</strong>hibitors 91leukotrienes 70levamisole 115levosalbutamol 885-lipoxygenase <strong>in</strong>hibitors 70liquid ventilation 64lisofyll<strong>in</strong>e 71, 102lobectomy 102–3Lop<strong>in</strong>avir 48lospor<strong>in</strong>s 107lung abscess 7lung biopsysurgical (SLB) 4, 112transbronchial (TBB) 4, 111–12lung function assessment 8lung <strong>in</strong>juryafter lung resection 102–4<strong>in</strong>halation 101–2thoracic trauma 99–101see also acute lung <strong>in</strong>jury; ventilator <strong>in</strong>ducedlung <strong>in</strong>jurylung <strong>in</strong>jury score 31–3lung <strong>in</strong>tubation 129, 131lung resection 102–4LY315920Na/S-5920 71lymphangioleiomyomatosis 128lymphopenia 46lysophosphatidic acyl transferase <strong>in</strong>hibitors71magnesium sulphate 91malignancy 3, 128, 129, 132matrix metalloproteases 42maximum <strong>in</strong>spiratory pressure 74, 75meropenem 107metabisulphite 89metered dose <strong>in</strong>haler (MDI) 88, 90methaemoglob<strong>in</strong>aemia 67methylprednisolone 49, 69, 110methylxanth<strong>in</strong>es 88midazolam 89m<strong>in</strong>ute ventilation 74mitochondria 16morph<strong>in</strong>e 89mucolytics 71, 84, 90, 108, 132mucus, bronchial 86, 87,90multiple organ dysfunction syndrome(MODS) 38multiple organ failure 13, 14, 19, 56, 57muscular dystrophy 117myasthenia gravis 118mycetoma 128, 132Mycobacterium avium 123Mycobacterium avium <strong>in</strong>tracellulare 120mycophenylate mofetil 115Mycoplasma hom<strong>in</strong>is 126Mycoplasma pneumoniae 126myocardial ischaemia 77, 87myopathy 75, 89N-acetylcyste<strong>in</strong>e (NAC) 71, 84, 108, 132National Heart and Lung Institute (NHLI) 33nebulised drugs 87–8, 90, 102neodymium-yttrium-alum<strong>in</strong>ium-garnet (Nd-YAG) laser photocoagulation 130neonate 57, 63–4nerve block, <strong>in</strong>tercostal 101neuromuscular block<strong>in</strong>g drugs 89neuromuscular disorders 117–19neutrophils 38, 40, 103–4nitric oxide (NO)endothelial 14, 41, 66, 125–6<strong>in</strong>haled 50, 66–7, 68, 91, 127nitrotyros<strong>in</strong>e 40nocturnal ventilatory support 118–19non-<strong>in</strong>vasive positive pressure ventilation(NIPPV) 100–1non-<strong>in</strong>vasive ventilation (NIV) 22, 108, 112,117acute severe asthma 88COPD 80, 81failure 81–2nocturnal 118–19wean<strong>in</strong>g from mechanical ventilation 77noradrenal<strong>in</strong>e 95North American-European ConsensusConference (NAECC) 32–3nosocomial pneumonia 24–8, 84antimicrobial treatment 24–5, 26–8


136 Index<strong>in</strong> COPD 84def<strong>in</strong>itions 24, 25diagnosis 4–5, 25–6, 26–7non-ventilated patient 24, 28nutritional support 70, 71, 84, 108occlusion pressure 74oesophageal <strong>in</strong>jury 100opioids 89, 126overdistension, lungcauses 8, 39, 52–3prevention 61–2oxidant stress 40, 53, 66–7, 71, 102, 103oxygencellular utilisation 16–17<strong>in</strong>spired 53, 60–1oxygenation, abnormality <strong>in</strong> ARDS 31, 32, 33,34oxygenation abnormality, ARDS 35oxygenation <strong>in</strong>dex 8oxygen consumption 12, 13oxygen deliveryregional 13–14to tissues 11, 12oxygen extraction ratio (OER) 12oxygen-haemoglob<strong>in</strong> dissociationrelationship 14, 15oxygen sens<strong>in</strong>g 17oxygen supplementation 87, 126oxygen transport 11, 12, 14, 15pa<strong>in</strong> control 101, 126Papworth Hospital 96partial (assisted) ventilatory support 77, 83–4,88, 89parvovirus 126patient distress 83pentamid<strong>in</strong>e isethionate 122pentoxifyll<strong>in</strong>e 71perfluorocarbons 64pericardiocentesis 100per<strong>in</strong>uclear ant<strong>in</strong>eutrophil cytoplasmicantibodies (P-ANCA) 115peroxynitrite 66–7pH, arterial 81, 82, 89phosphatidic acid <strong>in</strong>hibition 71phosphodiesterase <strong>in</strong>hibitors 95phospholipase <strong>in</strong>hibition 71phrenic nerve <strong>in</strong>jury 118–19piperacill<strong>in</strong> 107plasmapheresis 49platelet activat<strong>in</strong>g factor (PAF) <strong>in</strong>hibitors 91Pneumocystis pneumonia 5, 120–4pneumonectomy 102–3pneumoniabronchoscopy 4–5cytomegalovirus 5nosocomial 4–5, 24–8, 84Pneumocystis 5, 120–4pneumonitisCMV 110, 111diffuse 102pneumothorax 4, 7, 8, 99, 100polyangitis, microscopic 115polyarteritis nodosa 115polymerase cha<strong>in</strong> reaction (PCR) 46polyneuropathy 75polyv<strong>in</strong>yl alcohol (PVA) foam granules 128positive end expiratory pressure (PEEP) 62–3,76, 77, 82–3, 90<strong>in</strong>tr<strong>in</strong>sic (auto) 62, 76, 77, 82–3, 86, 87post-mortem studies, SARS 48pre-oxygenation 82pressure controlled ventilation 61–2, 82–3, 89pressure support ventilation 83, 89pressure trigger<strong>in</strong>g 77, 83pressure-volume (PV) curve 8, 60, 61primaqu<strong>in</strong>e 122procollagen III peptide 33, 39procyste<strong>in</strong>e 71prone position<strong>in</strong>g 5–6, 50, 63propofol 83–4, 89proportional assist ventilation (PAV) 77prostacycl<strong>in</strong> (PGI 2) 14, 67–8, 95prostagland<strong>in</strong> E1 (PGE 1)70protected specimen brush (PSB) 4, 25–6prote<strong>in</strong> C 33Pseudomonas aerug<strong>in</strong>osa 28, 107pulmonary artery, rupture 132pulmonary artery catheter 6, 132pulmonary artery catheterisation 94pulmonary artery occlusion pressure (PAOP)32pulmonary artery pressure (PAP) 93pulmonary contusion 99, 100pulmonary embolismdiagnosis 7–8, 95treatment 96, 97pulmonary endarterectomy 96pulmonary hypertension 41, 93–5pulmonary oedema 42, 64–5, 66pulmonary vascular remodell<strong>in</strong>g 94quality of care 19–20qu<strong>in</strong>olone 27,28rapid shallow breath<strong>in</strong>g 74reactive airways disease syndrome (RADS)101reactive nitrogen species (RNS) 103reactive oxygen species (ROS) 66–7, 71, 102,103rebreath<strong>in</strong>g 82recruitment, alveolar 5, 61manoeuvres to <strong>in</strong>crease 62–3red cell aplasia 118renal dysfunction 94respiratory musclesassessment 8<strong>in</strong> asthma 86strength tra<strong>in</strong><strong>in</strong>g 75–6weakness 77, 117, 118–19ribavir<strong>in</strong> 48rib fractures 99, 100–1right ventricular dysfunction 93, 94, 95, 96right ventricular support 95rocuronium 82, 89Royal Brompton Hospital 107salbutamol 87–8salmeterol 88SARS see severe acute respiratory syndromeSARS coronavirus 45tests for 46scoliosis 117sedatives 83–4, 89sepsis 13–14, 33, 35, 38septic shock 13, 38septostomy, atrial 95severe acute respiratory syndrome (SARS) 45cl<strong>in</strong>ical features 45–6diagnostic test<strong>in</strong>g 46impact on healthcare systems 51<strong>in</strong>fection control 50–1natural history 47–8survival 49treatment and respiratory management48–50sevoflurane 90shock 13, 95, 96sickle cell anaemia 125–7SIMV see synchronised <strong>in</strong>termittentmandatory ventilationskeletal muscle 15, 75–6sleep 83, 118–19smoke <strong>in</strong>halation 101–2Spanish Lung Failure Collaborative Group 77spiral CT angiography 94spirometry<strong>in</strong>centive 126–7prediction of wean<strong>in</strong>g 74splanchnic circulation 14, 94spontaneous breath<strong>in</strong>gcomponents 75<strong>in</strong> mechanical ventilation 63, 77, 83Staphylococcus aureus 20, 28, 126methicill<strong>in</strong>-resistant (MRSA) 28sternal fracture 100Streptococcus pneumoniae 20, 28, 126streptok<strong>in</strong>ase 96stress failure, lungs 55–6surfactantdysfunction 41, 55–6supplementation 56, 69–70suxamethonium 82, 89synchronised <strong>in</strong>termittent mandatoryventilation (SIMV) 77, 83, 89systemic <strong>in</strong>flammatory response syndrome38, 56systemic vasculitis 114–16, 128temocill<strong>in</strong> 107tensilon test 118terbutal<strong>in</strong>e 88thermal <strong>in</strong>jury 101–2thiopentone 89thoracic trauma 99–101thoracostomy, closed tube 100thoracotomy 100, 117–18thromb<strong>in</strong> 41, 130thromb<strong>in</strong>-fibr<strong>in</strong>ogen solution 130thrombocytopenia 115thrombolysis 96thromboxane synthase <strong>in</strong>hibitors 70, 70–1thymoma 117–18tidal volume, ventilator 52–3, 61–2tissue hypoxia 13–16tissue oedema 14, 15–16tissue plasm<strong>in</strong>ogen activator, recomb<strong>in</strong>ant(rt-PA) 96tobramyc<strong>in</strong> 107tocopherol 71Toronto, SARS outbreak 49, 51T-piece wean<strong>in</strong>g 77tracheal-arterial fistula 132tracheal gas <strong>in</strong>sufflation 61tracheal rupture 6tracheobronchial aspirates 25, 26tracheobronchial suction<strong>in</strong>g 50tracheostomy 84, 108, 132tra<strong>in</strong><strong>in</strong>g, <strong>in</strong> <strong>in</strong>tensive care 1–2tranexamic acid 107, 132transcription factors 17transdiaphragmatic pressure 8transform<strong>in</strong>g growth factor-β1 41transplantationheart-lung 95lung 107, 112trauma 8, 99 101triggered mechanical ventilation 77, 83, 89trimethoprim 122trimetrexate 122ultrasound, thoracic 6United Network for Organ Shar<strong>in</strong>g 112urok<strong>in</strong>ase 96vancomyc<strong>in</strong> 27,28vascular cell adhesion molecule 1 (VCAM-1)125–6vasculitis, pulmonary 114–16, 128vasodilation, sepsis-<strong>in</strong>duced 14vasodilators 68,94<strong>in</strong>haled 66–8vaso-occlusive crisis 126vasopress<strong>in</strong>, <strong>in</strong>travenous 132vena caval filter 96ventilation-perfusion match<strong>in</strong>g 94, 126


Index 137ventilator alarms 90ventilator associated pneumonia (VAP) 4–5,24–8ventilator <strong>in</strong>duced lung <strong>in</strong>jury (VILI) 52avoidance 61cl<strong>in</strong>ical consequences 56–7manifestations 54–5mechanisms 55–6, 60ventilator and patient determ<strong>in</strong>ants 52–4ventricular assist devices 95very late activation antigen 4 (VLA-4) 125visual disturbance 94volume controlled ventilation 61–2, 82volutrauma 53von Willebrand factor antigen 33wean<strong>in</strong>gacute severe asthma 90–1cystic fibrosis 108non-<strong>in</strong>vasive ventilation 77, 119obstructive lung disease 77, 83–4predictors of success 74–5psychological support 77specialist unit 78techniques 77wean<strong>in</strong>g failure 74–7, 83–4Wegener’s granulomatosis 114–16, 128wheeze 87, 126work of breath<strong>in</strong>g 76, 86xenon-133 ventilation-perfusion lungsc<strong>in</strong>tiphotography 102


___________________________________________________

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!