Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
CURE PALLIATIVE E DI SUPPORTO IN ONCOLOGIA INDICE Volume di 336 pagine con 16 figure in nero e a colori e 100 tabelle ISBN 978-88-7711-864-6 INTRODUZIONE Cure palliative e di supporto Bioetica Comunicazione Fattori prognostici Qualità di vita Spiritualità DOLORE Neurofisiologia Caratteristiche del dolore Valutazione del dolore Trattamento farmacologico Oppioidi Aspetti specifici del trattamento con oppioidi Procedure invasive SINTOMI Sintomi gastrointestinali Sintomi respiratori Sintomi psichiatrici Sindromi sistemiche e situazioni specifiche Emergenze Cure di fine vita € 45,00 Le cure palliative sono state progressivamente adottate negli ospedali e nelle istituzioni accademiche di tutto il mondo, ma tale processo è stato lento e difficile. Le risorse umane ed economiche sono limitate rispetto ad altre discipline che si sono sviluppate nella corrente principale della medicina clinica e accademica. In questo volume l’autore introduce il lettore lungo un percorso in cui vengono trattati tutti gli aspetti delle cure palliative, dalla valutazione al trattamento del paziente oncologico. Questo testo sarà pertanto fonte di ispirazione per tutti i professionisti del settore nella loro pratica quotidiana e permetterà di migliorare le cure dei pazienti sofferenti. 3HUOHYRVWUHRUGLQD]LRQL COME ORDINARE LIBRI x ULWDJOLDWHHVSHGLWHODFHGRODD/LEUHULD0LQHUYD(GL]LRQL0LQHUYD0HGLFD&RUVR%UDPDQWH7RULQR x SHUXQFRQWDWWRGLUHWWR7HO)D[ x FROOHJDWHYLDOVLWR,QWHUQHWZZZPLQHUYDPHGLFDLW,QGLUL]]RHPDLOERRNGHSW#PLQHUYDPHGLFDLW ACQUISTA ON-LINE *OL(QWL3XEEOLFLVRQRSUHJDWLGLHIIHWWXDUH,HRUGLQD]LRQLXWLOL]]DQGRLOEXRQRG·RUGLQHGHOO·(QWHVWHVVR /·DFTXLUHQWHKDGLULWWRGLUHVWLWXLUHODPHUFHHQWURJLRUQLGDOODGDWDGLULFHYLPHQWRGHOSDFFRSRVWDOH/DUHVWLWX]LRQHVDUjDFDULFRGHOO·DFTXLUHQWHVWHVVRHODPHUFHQRQGRYUjHVVHUHLQ DOFXQPRGRGHWHULRUDWD'HFUHWROHJLVODWLYR1,GDWLWUDVPHVVLFLHFKH/HLDLVHQVLGHOOD/HJJHFLDXWRUL]]DDWUDWWDUHVDUDQQRXWLOL]]DWLDLVROLILQLFRPPHUFLDOLH SURPR]LRQDOLGHOODQRVWUDDWWLYLWj$JJLRUQDPHQWLRFDQFHOOD]LRQLGHLGDWLGRYUDQQRHVVHUHULFKLHVWLD(GL]LRQL0LQHUYD0HGLFD&RUVR%UDPDQWH7RULQR Accedi al sito www.minervamedica.it, clicca su LIBRI e procedi con l’ordine on-line. Metodi di pagamento: carta di credito e, solo per spedizioni in Italia, contrassegno. MODALITÀ E SPESE DI SPEDIZIONE 6HQG1RFRSLHVRI Spedizione tramite corriere espresso. Le spese di spedizione saranno calcolate in base al luogo di consegna, peso dei volumi e, solo per le spedizioni in Italia, metodo di pagamento. Contrassegno al ricevimento del volume Pagamento anticipato a mezzo assegno o su c /c postale n. 00279109 TEMPI DI SPEDIZIONE Carta diincredito Spedizioni Italia: 2-4 giorni lavorativi. Spedizioni fuori Italia: 3-7 giorni lavorativi a seconda della destinazione. Diners Club International Carta n. ® scadenza ALTRE MODALITÀ ACQUISTO Cognome e nome DI ............................................................................................................................................................................................................................................................ Per altre modalità di acquisto e pagamento contattaci: Indirizzo .................................................................................................................................................................................................................................................................................... - email: [email protected] - tel. 011 678282 int. 328. InCAP caso........................ di istituti, enti o ditte che effettuano le ordinazioni con buoni d’ordine contattaci: Città ............................................................................................................................................................................................................ Prov. ............................... - email: [email protected] - tel. 011 678282 int. 328. Partita IVA o Codice Fiscale ...................................................................................................................................................................................................................................... Ai sensi del D.Lgs 196/2003 Minerva Medica garantisce la riservatezza dei dati e la possibilità di modificarli o cancellarli con richiesta scritta al Data ........................ Email ............................................................................................. ............................................................................................................................... responsabile dei dati personali. Per ulteriori informazioni rivolgersi a: EdizioniFirma Minerva Medica - Privacy policy - Corso Bramante 83-85 10126 Torino (',=,21,0,1(59$0(',&$ MINERVA ANESTESIOLOGIC A . VOL. 82 . No. 11 . PAGES 1129 - 1240 . NOVEMBER 2016 S. MERCADANTE V O L U M E 8 2 · N o. 1 1 · N O V E M B E R 2 0 1 6 MINERVA ANESTESIOLOGICA ITALIAN JOURNAL OF ANESTHESIOLOGY AND ANALGESIA MONTHLY JOURNAL FOUNDED IN 1935 BY A. M. DOGLIOTTI OFFICIAL JOURNAL OF ITALIAN SOCIETY OF ANESTHESIOLOGY, ANALGESIA, RESUSCITATION AND INTENSIVE CARE (S.I.A.A.R.T.I.) EDITORIAL BOARD EDITOR IN CHIEF F. Cavaliere Roma, Italy CRITICAL CARE ANESTHESIA General Critical Care Associate Editor G. M. Albaiceta (Oviedo, Spain) Section Editor G. Biancofiore (Pisa, Italy) E. De Robertis (Napoli, Italy) General Anesthesia Associate Editor M. Rossi (Roma, Italy) Section Editor E. Cohen (New York, USA) P. Di Marco (Roma, Italy) J. T. Knape (Utrecht, The Netherlands) O. Langeron (Paris, France) P. M. Spieth (Dresden, Germany) Circulation Critical Care Section Editor S. Scolletta (Siena, Italy) E. Bignami (Milano, Italy), Pediatric Anesthesia Section Editor M. Piastra (Roma, Italy) Respiration Critical Care Section Editor S. Grasso (Bari, Italy) P. Terragni (Sassari, Italy), Obstetric Anesthesia Section Editor E. Calderini (Milano, Italy) Neurocritical Care Section Editor F. S. Taccone (Brussels, Belgium) Regional Anesthesia Section Editor A. Apan (Giresun, Turkey) M. Carassiti (Roma, Italy) ETHICS PAIN Section Editor A. Giannini (Milano, Italy) Section Editor M. Allegri (Parma, Italy) F. Coluzzi (Roma, Italy) MEDICAL STATISTIC Section Editor B. M. Cesana (Brescia, Italy) MANAGING EDITOR A. Oliaro Torino, Italy Vol. 82 - No. 11 MINERVA ANESTESIOLOGICA III GENERAL INFORMATION This journal is PEER REVIEWED and is quoted in: Current Contents, SciSearch, PubMed/MEDLINE, EMBASE, Scopus Impact factor: 2.036 Published by Edizioni Minerva Medica - Corso Bramante 83-85 - I-10126 Turin (Italy) Tel. +39 (011) 678282 - Fax +39 (011) 674502 Website: www.minervamedica.it Editorial office: [email protected] - Subscriptions: [email protected] Advertising: [email protected] Presidential Office: Antonio Corcione, UOC Anestesia e TIPO, Azienda Ospedaliera dei Colli, V. Monaldi, Via Leonardo Bianchi 1, 80131 Naples, Italy - Tel. 081 7065214. E-mail: [email protected] Technical Secretariat: S.I.A.A.R.T.I., Viale dell’Università 11, 00185 Rome, Italy - Tel. 06 4452816 E-mail: [email protected] - Website: www.siaarti.it Administrative Secretariat S.I.A.A.R.T.I. - Edizioni Minerva Medica - Corso Bramante 83-85 - 10126 Turin, Italy - Tel. +39 (011) 678282 - Fax +39 (011) 674502 - E-mail: [email protected] ANNUAL SUBSCRIPTIONS ITALY EUROPEAN UNION OUTSIDE THE EUROPEAN UNION Individual: Print € 130,00 Print+Online € 135,00 Online € 140,00 Individual: Print € 225,00 Print+Online € 235,00 Online € 245,00 Individual: Print € 250,00 Print+Online € 260,00 Online € 270,00 Institutional: Print € 180,00 Institutional: Print € 300,00 Institutional: Print € 335,00 Online Small Medium Large € 350,00 € 407,00 € 429,00 Online Small Medium Large € 351,00 € 409,00 € 429,00 Online Small Medium Large € 383,00 € 446,00 € 462,00 Print+Online Small Medium Large € 367,00 € 420,00 € 446,00 Print+Online Small Medium Large € 378,00 € 430,00 € 456,00 Print+Online Small Medium Large € 404,00 € 467,00 € 483,00 Subscribers: Payment to be made in Italy: a) by check; b) by bank transfer to: Edizioni Minerva Medica, INTESA SANPAOLO Branch no. 18 Torino. IBAN: IT45 K030 6909 2191 0000 0002 917 c) through postal account no. 00279109 in the name of Edizioni Minerva Medica, Corso Bramante 83-85, 10126 Torino; d) by credit card Diners Club International, Master Card, VISA, American Express. Foreign countries: a) by check; b) by bank transfer to: Edizioni Minerva Medica, INTESA SANPAOLO Branch no. 18 Torino. IBAN: IT45 K030 6909 2191 0000 0002 917; BIC: BCITITMM c) by credit card Diners Club International, Master Card, VISA, American Express. Members: for payment please contact the Administrative Secretariat SIAARTI. Notification of changes to mailing addresses, e-mail addresses or any other subscription information must be received in good time. Notification can be made by sending the new and old information by mail, fax or e-mail or directly through the website www.minervamedica.it at the section “Your subscriptions - Contact subscriptions department”. Complaints regarding missing issues must be made within six months of the issue’s publication date. Prices for back issues and years are available upon request. © Edizioni Minerva Medica - Torino 2016. All rights reserved. No part of this publication may be reproduced, stored or transmitted in any form or by any means, without the prior permission of the copyright owner. Monthly publication. Authorized by Turin Court no. 281 of 24-1-1953. This Journal is associated with IV This Journal complies with the Code of Self-Discipline of Medical/Scientific Publishers associated with FARMAMEDIA and may accept advertising MINERVA ANESTESIOLOGICA November 2016 INS TRUCT IONS TO AUT HORS Minerva Anestesiologica is the journal of the Italian National Society of Anaesthesia, Analgesia, Resuscitation, and Intensive Care (SIAARTI - http://www.siaarti.it). Minerva Anestesiologica publishes scientific papers on Anesthesiology, Intensive care, Analgesia, Perioperative Medicine and related fields. Manuscripts are expected to comply with the instructions to authors which conform to the Uniform Requirements for Manuscripts Submitted to Biomedical Editors by the International Committee of Medical Journal Editors (http://www.icmje.org). Articles not conforming to international standards will not be considered for acceptance. Submission of manuscripts Papers should be submitted directly to the online Editorial Office at the Edizioni Minerva Medica website: http://www.minervamedicaonlinesubmission.it Duplicate or multiple publication Submission of the manuscript means that the paper is original and has not yet been totally or partially published, is not currently under evaluation elsewhere, and, if accepted, will not be published elsewhere either wholly or in part. Splitting the data concerning one study in more than one publication could be acceptable if authors justify the choice with good reasons both in the cover letter and in the manuscript. Authors should state the new scientific contribution of their manuscript as compared to any previously published article derived from the same study. Relevant previously published articles should be included in the cover letter of the currently submitted article. Permissions to reproduce previously published material Material (such as illustrations) taken from other publications must be accompanied by the publisher’s permission. Copyright The Authors agree to transfer the ownership of copyright to Minerva Anestesiologica in the event the manuscript is published. Ethics committee approval All articles dealing with original human or animal data must include a statement on ethics approval at the beginning of the methods section, clearly indicating that the study has been approved by the ethics committee. This paragraph must contain the following information: the identification details of the ethics committee; the protocol number that was attributed by the ethics committee and the date of approval by the ethics committee. The journal adheres to the principles set forth in the Helsinki Declaration (http://www.wma.net/en/30publications/10policies/ b3/index.html) and states that all reported research concerning human beings should be conducted in accordance with such principles. The journal also adheres to the International Guiding Principles for Biomedical Research Involving Animals (http:// www.cioms.ch/index.php/12-newsflash/227-cioms-and-iclasrelease-the-new-international-guiding-principles-for-biomedical-research-involving-animals) recommended by the WHO and requires that all research on animals be conducted in accordance with these principles. Patient consent Authors should include at the beginning of the methods section of their manuscript a statement clearly indicating that patients have given their informed consent for participation in the research study. Every precaution must be taken to protect the privacy of patients. Conflicts of interest Authors must disclose possible conflicts of interest including financial agreements or consultant relationships with organizations involved in the research. All conflicts of interest must be declared both in the authors’ statement form and in the manuscript file. If there is no conflict of interest, this should also be explicitly stated as none declared. All sources of funding should be acknowledged in the manuscript. Authorship and contributorship All persons and organizations that have participated to the study must be listed in the byline of the article (authors) or in the notes (contributors). The manuscript should be approved by all coauthors, if any, as well as, tacitly or explicitly, by the responsible authorities of the institution where the work was carried out. Authors and contributors must meet the criteria for authorship and contributorship established by the Uniform Requirements for Manuscripts Submitted to Biomedical Editors by the International Committee of Medical Journal Editors (http://www.icmje.org). Authors’ statement Papers must be accompanied by the authors’ statement (http:// www.minervamedica.it/en/journals/minerva-anestesiologica/ index.php) relative to copyright, originality, authorship, ethics and conflicts of interest, signed by all authors. Disclaimer The Publisher, Editors, and Editorial Board cannot be held responsible for the opinions and contents of publications contained in this journal. The authors implicitly agree to their paper being peer-reviewed. All manuscripts will be reviewed by Editorial Board members who reserve the right to reject the manuscript without entering the review process in the case that the topic, the format or ethical aspects are inappropriate. In addition, the Editorial Board admits for submission manuscripts previously submitted for publication to another journal but rejected with reviewers’ comments. In such cases, authors are requested to submit the scientific comments of the other journal and their point-by-point reply to the reviewers’ comments in the manuscript file. This will help to speed up the peer review process. Once accepted, all manuscripts are subjected to copy editing. If modifications to the manuscript are requested, the corresponding author should send to the online Editorial Office the revised manuscript under two separate files, one file containing the revised clean version and another containing both a letter with pointby-point responses to the reviewers’ comments and the revised version with corrections highlighted. Correction of proofs should be limited to typographical errors. Substantial changes in content (changes of title and authorship, new results and corrected values, changes in figures and tables) are subject to editorial review. Changes that do not conform to the journal’s style are not accepted. Corrected proofs must be sent back within 3 working days to the online Editorial Office of Minerva Anestesiologica. In case of delay, the editorial staff of the journal may correct the proofs on the basis of the original manuscript. Publication of manuscripts is free of charge. Colour figures and excessive alterations to proofs will be charged to the authors. Authors will receive instructions on how to order reprints and a copy of the manuscript in PDF. For further information about publication terms please contact the Editorial Office of Minerva Anestesiologica, Edizioni Minerva Medica, Corso Bramante 83-85, 10126 Torino, Italy - Phone +39011-678282 - Fax +39-011-674502 E-mail: [email protected]. ARTICLE TYPES Original papers are reports of original researches on the topics of interest to the journal and do not exceed 3000 words, 6 figures or tables, and 40 references. Papers reporting randomized controlled trials should be in accordance with the CONSORT statement (http://www.consort-statement.org). Review articles are comprehensive papers that outline current knowledge, usually submitted after prior consultation with the Editors and do not exceed 4000 words, 6 figures or tables, 70 references. Reviews should be in accordance with the PRISMA statement (http://www.prisma-statement.org). Experts’ opinions are papers reviewing the literature, usually invited by the Editor in Chief and do not exceed 3000 and 2000 words with 50 and 40 references respectively. Editorials are brief comments, commissioned by the Editor-inChief and express author’s opinion about published papers or topical issues and do not exceed 1000 words and 20 references. Letters to the Editor (Correspondence) are brief comments on articles published in the journal or with other topics of interest and do not exceed 500 words, 5 references and 1 figure or table. PREPARATION OF MANUSCRIPTS Text file The formats accepted are Word (.DOC) and RFT. The text file must contain title, authors’ details, abstract, key words, text, references, notes and titles of tables and figures. Tables and figures should be submitted as separate files. The file should not contain active hyperlinks. Title and authors’ details Short title, with no abbreviations. First name in full, middle name’s initial, surname of the authors. Collective name, if any, as last author. Corresponding author marked with an asterisk. Affiliation (section, department and institution) of each author. Name, address, e-mail of the corresponding author. Abstract and key words Articles should include an abstract not exceeding 250 words. For original papers and technical notes, the abstract should be structured as follows: background (status of knowledge and aim of the study), methods (experimental design, patients and interventions), results (what was found), conclusion (meaning of the study). Key words should refer to the terms from Medical Subject Headings (MeSH) of the Index Medicus. No abstracts are required for editorials or letters to the Editor. Text Units of measurement, symbols and abbreviations must conform to international standards. Measurements of length, height, weight and volume should be given in metric units (meter, kilogram, liter) or their decimal multiples. Temperatures must be expressed in degrees Celsius. Blood pressure must be expressed in millimeters of mercury. All hematological and clinicochemical measurements should be expressed in metric units using the International System of Units (IS). The use of unusual symbols or abbreviations is strongly discour-aged. The first time an abbreviation appears in the text, it should be preceded by the words for which it stands. For original papers the text must be subdivided into the following sections: introduction, materials and methods, results, discussion, conclusions. The introduction should describe the present state of knowledge on the subject and clearly state the aim of the research. The Materials and methods section should provide enough information to allow other researchers to reproduce results and should include a statistics section. Detailed statistical methodology must be described in order to enable the reader to evaluate and verify the results. Please report power analysis, randomized procedure, tests of normality, descriptive parameters, statistical tests and the probability level chosen for statistical significance. Drugs should be identified by their international non-proprietary name. If special equipment is used, then the manufacturer’s details (including town and country) should be given in parentheses. The Results section should describe the outcome of the study. Data presented in tables and graphs should not duplicate those presented in the text. Very large tables should be avoided. The Discussion should include the interpretation of the results obtained, an analysis of the limits of the study, and references to work by other authors. Conclusions should highlight the significance of the results for clinical practice and experimental research. References The list of references should only include works that are cited in the text and that have been published or accepted for publication (a copy of the acceptation letter can be requested) in journals cited in peer-reviewed Index Medicus journals. Websites can also be cited providing the date of access. Bibliographical entries in the text should be quoted using superscripted Arabic numerals. References must be set out in the standard format approved by the International Committee of Medical Journal Editors (http:// www.icmje.org). Footnotes and endnotes must not be used in the preparation of references. Journals Each entry must specify the author’s surname and initials (list all authors when there are six or fewer; when there are seven or more, list only the first six and then “et al.”), the article’s original title, the name of the Journal (according to the abbreviations used by Index Medicus), the year of publication, the volume number and the number of the first and last pages. When citing references, please follow the rules for international standard punctuation carefully. Examples – Standard article Sutherland DE, Simmons RL, Howard RJ. Intracapsular technique of transplant nephrectomy. Surg Gynecol Obstet 1978;146:951-2. – Organization as author International Committee of Medical Journal Editors. Uniform requirements for manuscripts submitted to biomedical journals. Ann Int Med 1988;108:258-65. – Issue with supplement Payne DK, Sullivan MD, Massie MJ. Women’s psychological reactions to breast cancer. Semin Oncol 1996;23(1 Suppl 2): 89-97. Books and monographs For occasional publications the names of the Authors, the title, edition, place, publisher and year of publication must be given. Examples – Books by one or more Authors Rossi G. Manual of Otorhinolaryngology. Turin: Edizioni Minerva Medica; 1987. – Chapter from book De Meester TR. Gastroesophageal reflux disease. In: Moody FG, Carey LC, Scott Jones R, Ketly KA, Nahrwold DL, Skinner DB, editors. Surgical treatment of digestive diseases. Chicago: Year Book Medical Publishers; 1986. p. 132-58. – Congress proceedings Kimura J, Shibasaki H, editors. Recent advances in clinical neurophysiology. Proceedings of the 10th International Congress of EMG and Clinical Neurophysiology; 1995 Oct 15-19; Kyoto, Japan. Amsterdam: Elsevier; 1996. Notes Authors’ contribution statement; list of the members of the collective name (author’s name in full, middle name’s initial in capital letters and surname, with relevant affiliation); contributors’ names; mention of any funding, research contracts; conflicts of interest; dates of any congress where the paper has already been presented; acknowledgements. Titles of tables and figures Titles of tables and legends of figures should be included both in the text file and in the file of tables and figures. File of tables Each table should be submitted as a separate file. Formats accepted are Word (.DOC) and RTF. Each table should be created with the Table menu of the word processing software of the operating system employed, by selecting the number of rows and columns needed. Tabulations are not allowed. Each table must be typed correctly and prepared graphically in keeping with the page layout of the journal, numbered in Roman numerals and accompanied by the relevant title. Notes should be inserted at the foot of the table and not in the title. Tables should be referenced in the text sequentially. File of figures Each figure should be submitted as a separate file. Formats accepted: JPEG set at 300 dpi resolution preferred; other formats accepted are TIFF and PDF (high quality). Figures should be numbered in Arabic numerals and accompanied by the relevant legend. Figures should be referenced in the text sequentially. Histological photographs should always be accompanied by the magnification ratio and the staining method. If figures are in color, it should always be specified whether color or black and white reproduction is required. MINERVA ANESTESIOLOGICA ITALIAN JOURNAL OF ANESTHESIOLOGY AND ANALGESIA MONTHLY JOURNAL FOUNDED IN 1935 BY A. M. DOGLIOTTI OFFICIAL JOURNAL OF ITALIAN SOCIETY OF ANESTHESIOLOGY, ANALGESIA, RESUSCITATION AND INTENSIVE CARE (S.I.A.A.R.T.I.) Vol. 82 November 2016 No. 11 OFFICIAL JOURNAL OF ITALIAN SOCIETY OF ANESTHESIOLOGY, ANALGESIA, RESUSCITATION AND INTENSIVE CARE (SIAARTI) CONTENTS 1129 1149 EDITORIALS Propofol-remifentanil and spontaneous breathing, a magnificent pair Sbaraglia F., Sammartino M. Diaphragmatic ultrasonography as an adjunct predictor tool of weaning success in patients with difficult and prolonged weaning Flevari A., Lignos M., Konstantonis D., Armaganidis A. 1158 1132 Ultrasonography during weaning: a support to a comprehensive skilful strategy De Robertis E., Romano G. M., Piazza O. Non-invasive hemodynamic optimization in major abdominal surgery: a feasibility study Broch O., Carstens A., Gruenewald M., Nischelsky E., Vellmer L., Bein B., Aselmann H., Steinfath M., Renner J. 1170 1135 Perioperative hemodynamic therapy: goal-directed or meta-directed? Propofol versus midazolam for premedication: a placebo‑controlled, randomized double‑blinded study Uhlig C., Spieth P. M. Elvir Lazo O. L., White P. F., Tang J., Yumul R., Cao X., Yumul F., Hausman J., Hernandez Conte A., Anand K. K., Hemaya E. G., Zhang X., Wender R. H. 1138 1180 ORIGINAL ARTICLES Propofol-remifentanil anesthesia for upper airway endoscopy in spontaneous breathing patients: the ENDOTANIL Randomized Trial Besch G., Chopard-Guillemin A., Monnet E., Causeret A., Jurine A., Baudry G., Lasry B., Tavernier L., Samain E., Pili-Floury S. Vol. 82 - No. 11 The optimal time between clinical brain death diagnosis and confirmation using CT angiography: a retrospective study Kerhuel L., Srairi M., Georget G., Bonneville F., Mrozek S., Mayeur N., Lonjaret L., Sacrista S., Hermant N., Marhar F., Gaussiat F., Abaziou T., Osinski D., Le Gaillard B., Menut R., Larcher C., Fourcade O., Geeraerts T. MINERVA ANESTESIOLOGICA I CONTENTS 1189 Serum S100β as a prognostic marker in patients with non-traumatic intracranial hemorrhage Junttila E. K., Koskenkari J., Ohtonen P. P., Karttunen A., Ala-Kokko T. I. 1199 REVIEWS Perioperative hemodynamic goal-directed therapy and mortality: a systematic review and meta-analysis with meta-regression Giglio M., Manca F., Dalfino L., Brienza N. 1214 Targeting blood products transfusion in trauma: what is the role of thromboelastography? Figueiredo S., Tantot A., Duranteau J. 1235 LETTERS TO THE EDITOR High-sensitivity troponin and extubation failure after successful spontaneous breathing trial Mottard N., Renaudin P., Wallet F., Thiollière F., Bohe J., Friggeri A. 1236 Posterior reversible encephalopathy syndrome in acute pancreatitis Compagnone C., Bellantonio D., Pavan F., Tagliaferri F., Barbagallo M., Fanelli G. 1238 Successful treatment of life-threatening hemorrhaging due to amniotic fluid embolism Aurini L., Rainaldi M. P., White P. F., Borghi B. 1230 JOURNAL CLUB CRITIQUE Failure of statins in ARDS: the quest for the Holy Grail continues Grimaldi D., Durand A., Gleeson J., Taccone F. S. 1240 TOP 50 MINERVA ANESTESIOLOGICA REVIEWERS V O L U M E 8 2 · N o. 1 1 · N O V E M B E R 2 0 1 6 About the cover: The cover shows the Serge’s sphygmo-oscillograph produced by Viola Gaetano company, Milan, around 1930. From the SIAARTI collection of medical devices at Viale dell’Università, 11; Rome, Italy. II MINERVA ANESTESIOLOGICA November 2016 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1129-31 EDITORIAL Propofol-remifentanil and spontaneous breathing, a magnificent pair Fabio SBARAGLIA*, Maria SAMMARTINO Institute of Anesthesia and Intensive Care, Sacro Cuore Catholic University, Rome, Italy *Corresponding author: Fabio Sbaraglia, Institute of Anesthesia and Intensive Care, Sacro Cuore Catholic University, Largo F Vito 1, 00135 Rome, Italy. E-mail: [email protected] T he maintenance of spontaneous breathing during surgical/endoscopic procedures is a real challenge for the anesthesiologist. An increasing number of pathologies are treated with mini-invasive therapies; clinical status of our patients (poor conditions, neuromuscular diseases, high risk for weaning process, etc.) and new settings will force us to elaborate new strategies for the breathing control. For many years, the use of halogenated anesthetics has been mandatory but, the development of rapid off-set intravenous drugs seems to be a key source for the anesthesia community. Since the 1990s indeed, both propofol and remifentanil have gradually revolutionized our mindset, leading to experiment their potentiality in several fields. In this issue of Minerva Anestesiologica, Besch et al.1 have gone a step forward. The ENDOTANIL randomized trial he designed, assesses the efficacy of both propofol and remifentanil administered in target controlled infusion (TCI) in tubeless anesthesia with spontaneous breathing in patients undergoing pan-endoscopy. This procedure (a sequence of direct laryngoscopy, rigid bronchoscopy and esophagoscopy) is a high-risk challenge 2 and requires a close control over the respiratory trigger. The choice of this combination is Comment on p. 1138. Vol. 82 - No. 11 a pragmatic approach based on the synergistic interaction between propofol and remifentanil3 in a large number of patients, reporting good results and a safe profile. In the past, several papers looked into this matter with positive results. Anesthesiologists utilized propofol-remifentanil technique to achieve sedation while maintaining spontaneous ventilation in a large number of settings: awake intubation,4 bronchoscopy,5 complex digestive endoscopy,6 surgical procedures,7, 8 dentistry 9 and Monitored Anesthesia Care in adults 10 and children.11 The development of a more accurate administration by TCI models, further promoted the use of the intravenous technique in clinical practice. Despite the reluctance of the Food and Drug Administration in the United States 12 and the lack of a strong evidence of better outcomes,13 the number of papers published about this topic increased.14 TCI technique has been compared with traditional total intravenous anesthesia 15 and with manual controlled anesthesia too,16 but it does not provide yet sufficient evidence to allow firm recommendations about the TCI use. The approach of ENDOTANIL in the titration of propofol TCI is the paradigm of the success of this technique in the clinical practice, despite strong evidences are lacking. In that study infusion rate was increased step by step Minerva Anestesiologica 1129 SBARAGLIA Propofol-remifentanil and spontaneous breathing until the loss of consciousness (interindividual variability resulted in a range between 4.0 and 6.5 mg/mL), and, before starting with procedure, increased to reach a mild overdosage without affecting spontaneous breathing. Propofol administration is therefore tailored to the single patient, drawing attention from reached concentration to end results. We can apply the same remarks for remifentanil that, however, was maintained at fixed concentration in order to investigate the outcome of this study. The trend of clinical practice pushes toward an opening up of intravenous drugs, reaching the extreme in the patient controlled sedation (PCS). PCS seems to be the new frontier of sedation: patient, if well-trained, could dose the level of sedation thought the procedure. In this way, the risk of over-dosage, should be reduced, and the satisfaction of patient improved. First experiences in delivery room with remifentanil,17 and with propofol in endoscopy suite,18 reported positive results and the combination of these drugs has been satisfactory utilized in the early 2000s.19 Independently of who is the provider performing sedation, most papers emphasize the paramount importance of respiratory adverse events control by an adequate monitoring. Apnea, hypopnea and hypoventilation frequently occur during spontaneous breathing anesthesia and the use of SaO2 control is too late in detecting respiratory side effects. By contrast, end-expiratory carbon dioxide control (EtCO2), allows analyzing in real time the adequacy of alveolar ventilation and of respiratory rate. The immediate adverse events detection can help the provider in reducing the seriousness of them. After many authors confirmed these findings, the evidence of a need of an early EtCO2 monitoring has become so strong that even the American Society of Anesthesiologists modified its standards for basic anesthetic monitoring, including it as mandatory (July 2011).20 In this field, the clinical report by Besch finds its weakness. The number of respiratory adverse events is distorted by the absence of a reliable monitoring and the global amount of apneic events could not be accurately measured. The difference between the two arms 1130 of randomization (remifentanil vs. placebo) did match for this point, but only with the focus on the severe hypoxia. It is possible that a closer monitoring, increasing the number of light/moderate disventilation detected, could enhance the differences between these groups. Waiting for new trials about continuous transcutaneous measurement of pCO2 to guide sedation,21 EtCO2 monitoring must remain mandatory during spontaneous breathing sedation and anesthesia. Despite this bias, ENDOTANIL is a trial rich of positive suggestions. The combination of propofol and remifentanil in spontaneous breathing is a current reality, even if their potential is far from being understood. References 1. Besch G, Chopard-Guillemin A, Monnet E, Causeret A, Jurine A, Baudry G, et al. Propofol-remifentanil anesthesia for upper airway endoscopy in spontaneous breathing patients: the ENDOTANIL randomized trial. Minerva Anestesiol 2016;82:1138-48. 2. Passot S, Servin F, Allary R, Pascal J, Prades JM, Auboyer C, Pascal J, Prades JM, Auboyer C, et al. Target-controlled versus manually-controlled infusion of propofol for direct laryngoscopy and bronchoscopy. Anesth Analg 2002;94:1212-6. 3. Mertens MJ, Olofsen E, Engbers FH, Burm AG, Bovill JG, Vuyk J. Propofol reduces perioperative remifentanil requirements in a synergistic manner: response surface modeling of perioperative remifentanil-propofol interactions. Anesthesiology 2003;99:347-59. 4. Cafiero T, Esposito F, Fraioli G, Gargiulo G, Frangiosa A, Cavallo LM, et al. Remifentanil-TCI and propofol-TCI for conscious sedation during fibreoptic intubation in the acromegalic patient. Eur J Anaesthesiol 2008;25:670-4. 5.Chen L, Yu L, Fan Y, Manyande A. A comparison between total intravenous anaesthesia using propofol plus remifentanil and volatile induction/ maintenance of anaesthesia using sevoflurane in children undergoing flexible fibreoptic bronchoscopy. Anaesth Intensive Care 2013;41:742-9. 6.Borrat X, Valencia JF, Magrans R, Gimenez-Mila M, Mellado R, Sendino O, et al. Sedation-analgesia with propofol and remifentanil: concentrations required to avoid gag reflex in upper gastrointestinal endoscopy. Anesth Analg 2015;121:90-6. 7. Berkenstadt H, Perel A, Hadani M, Unofrievich I, Ram Z. Monitored anesthesia care using remifentanil and propofol for awake craniotomy. J Neurosurg Anesthesiol 2001;13:246-9. 8.Rosati M, Bramante S, Conti F, Rizzi M, Frattari A, Spina T. Laparoscopic salpingo-oophorectomy in conscious sedation. JSLS 2015;19. 9. Nagels AJ, Bridgman JB, Bell SE, Chrisp DJ. Propofolremifentanil TCI sedation for oral surgery. N Z Dent J 2014;110:85-9. 10.Höhener D, Blumenthal S, Borgeat A. Sedation and regional anaesthesia in the adult patient. Br J Anaesth 2008;100:8-16. Minerva Anestesiologica November 2016 Propofol-remifentanil and spontaneous breathingSBARAGLIA 11. Malherbe S, Whyte S, Singh P, Amari E, King A, Ansermino JM. Total intravenous anesthesia and spontaneous respiration for airway endoscopy in children--a prospective evaluation. Paediatr Anaesth 2010;20:434-8. 12.Egan TD, Shafer SL. Target-controlled infusions for intravenous anesthetics: surfing USA not! Anesthesiology 2003;99:1039-41. 13.Wilhelm W. Target-controlled infusion (TCI): clinical tool or scientific toy? Anaesthesist 2008;57:221-2. 14.Absalom AR, Glen JI, Zwart GJ, Schnider TW, Struys MM. Target-controlled infusion: a mature technology. Anesth Analg 2016;122:70-8. 15. Weninger B, Czerner S, Steude U, Weninger E. Comparison between TCI-TIVA, manual TIVA and balanced anaesthesia for stereotactic biopsy of the brain. Anasthesiol Intensivmed Notfallmed Schmerzther 2004;39:212-9. 16. Moerman AT, Herregods LL, De Vos MM, Mortier EP, Struys MM. Manual versus target-controlled infusion remifentanil administration in spontaneously breathing patients. Anesth Analg 2009;108:828-34. 17.Schnabel A, Hahn N, Broscheit J, Muellenbach RM, Rieger L, Roewer N, et al. Remifentanil for labour analgesia: a meta-analysis of randomised controlled trials. Eur J Anaesthesiol 2012;29:177-85. 18. Mazanikov M, Udd M, Kylänpää L, Mustonen H, Lindström O, Färkkilä M, et al. A randomized comparison of target-controlled propofol infusion and patient-controlled sedation during ERCP. Endoscopy 2013;45:915-9. 19. Joo HS, Perks WJ, Kataoka MT, Errett L, Pace K, Honey RJ. A comparison of patient-controlled sedation using either remifentanil or remifentanil-propofol for shock wave lithotripsy. Anesth Analg 2001;93:1227-32. 20. Weaver J. The latest ASA mandate: CO(2) monitoring for moderate and deep sedation. Anesth Prog 2011; 58:111-2. 21.Hannam JA, Borrat X, Trocóniz IF, Valencia JF, Jensen EW, Pedroso A, et al. Modeling respiratory depression induced by remifentanil and propofol during sedation and analgesia using a continuous noninvasive measurement of PCO2. J Pharmacol Exp Ther 2016;356:563-73 Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: August 31, 2016. - Manuscript accepted: August 24, 2016. - Manuscript received: July 14, 2016. (Cite this article as: Sbaraglia F, Sammartino M. Propofol-remifentanil and spontaneous breathing, a magnificent pair. Minerva Anestesiol 2016;82:1129-31) Vol. 82 - No. 11 Minerva Anestesiologica 1131 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1132-4 EDITORIAL Ultrasonography during weaning: a support to a comprehensive skilful strategy Edoardo DE ROBERTIS 1*, Giovanni M. ROMANO 1, Ornella PIAZZA 2 1Department of Neurosciences, Reproductive and Odontostomatologic Sciences, University of Naples Federico II, Naples, Italy; 2Department of Medicine and Surgery, University of Salerno, Salerno, Italy *Corresponding author: Edoardo De Robertis, Department of Neurosciences, Reproductive and Odontostomatologic Sciences, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy. E-mail: [email protected] M ost of critical ill patients can be extubated without complications, but 26-42% of those with mechanical ventilation (MV) fail the first attempt of weaning.1 Failure of extubation with subsequent reintubation is an independent risk factor for increased mortality.2 Therefore, deciding when a patient is ready for discontinuation of MV continues to be a challenge. Causes of weaning failure are: —— respiratory failure (increased ventilatory work; reduced compliance from cardiogenic or non-cardiogenic edema, etc.); —— cardiac failure (pre-existing cardiac dysfunction, increased metabolic demand, weaning-induced cardiac dysfunction); —— neuromuscular causes (depression of the respiratory center, muscular failure, including the diaphragm),3 ICU-acquired weakness;4 —— metabolic causes: malnutrition, hypoglicemia, electrolytes abnormalities, anemia; —— neuropsychological causes: anxiety, delirium. Early recognition and treatment of these adverse conditions help to reduce the risk of failure. Diaphragm is remarkably prone to dysfunction in critically ill patients receiving mechanical ventilation.5 Controlled mechanical ventiComment on p. 1149. 1132 lation induces an oxidative stress which leads to proteolysis and diaphragm atrophy.6, 7 Ultrasound is an excellent tool for anatomical and functional assessment of the diaphragm, with a valuable cost/ benefit ratio, since it is a non-invasive procedure. Diaphragmatic performance may be evaluated measuring the excursion of the muscle during inspiration with a 3- to 5-MHz probe (B- or M-mode), or by measuring the thickness with a 10-MHz probe (B- or M-mode).8 Both these methods have shown to correlate adequately with transdiaphragmatic pressure values in spontaneously breathing cardiac surgery patients.9 The study conducted by Flevari et al. in this issue of Minerva Anestesiologica adds new interesting insight on the potential value of ultrasonography evaluation of diaphragmatic function during weaning in critically ill patients.10 These authors examined 27 patients with difficult and/or prolonged weaning in a prospective cohort study. They found that during a spontaneous breathing trial, a left hemidiaphragm excursion at a cut-off 10 mm, assessed by M-mode ultrasonography, was the best index to predict weaning success (sensitivity 86%, specificity 85%, negative predictive value 94%). The cut-off values for predicting weaning failure as determined by the area under the receiver operating characteristic curve, were ≤10 mm for right hemidiaphragm excursion and ≤7 mm Minerva AnestesiologicaNovember 2016 Ultrasonography during weaning: a support to a comprehensive skilful strategy for left hemidiaphragm excursion. The authors concluded that ultrasonographic measurement of diaphragmatic excursion may be used as an adjunct predictor for the weaning process in patients with difficult and/or prolonged weaning. Indeed, focusing on the diaphragm excursion during the weaning process may be very intriguing. As a matter of fact, ultrasound method is by far easier to perform compared with more complex methods such as transdiaphragmatic pressure or maximal inspiratory pressure measurements. However, major drawbacks hide beyond the easiness and rapidity of the ultrasound technique. One is the operator-dependence and reproducibility of the results. In their study, Flevari et al., did not test interobserver and intraobserver variability, even though diaphragmatic excursion studied by M-mode ultrasonography seems to be a highly reproducible method.11 Moreover, the ultrasonographic evaluation of diaphragmatic function needs to take into account different variables: —— the precise identification of the phase of the respiratory cycle and its duration;12 —— the type and quality of breathing (quiet, deep, superficial); —— the patient’s position; —— the presence of PEEP (the increase in end-expiratory lung volume lowers the diaphragmatic dome and may decrease the diaphragmatic excursion);13 —— the presence of anatomic and/or pathologic alterations which can hinder the ultrasound scan. The measurement of diaphragmatic excursion requires a thorough understanding and identification of the various ultrasound diaphragmatic patterns in different illnesses and the corresponding changes in breathing mechanics. One strength of the study by Flevari et al., is the fact that the authors did measurements of both hemidiaphragmatic excursions, which may give a hint in the global diaphragmatic function evaluation and help understanding how the hemidiaphragms are working together. However, extrapolating the global respiratory function from the evaluation of the diaphragmatic excursion may be oversimplified. The interplay between thoracic cage, Vol. 82 - No. 11 DE ROBERTIS intercostal muscle and diaphragm contraction determine the force which generates flow through the airways, and the measurement of the diaphragmatic excursion is only one link of this complex biological network.14, 15 Nevertheless, we know that the causes of failure in retirement of MV are multifactorial and they cannot be related to the diaphragm dysfunction only. To remove mechanical ventilation the earliest is possible, it is necessary to reconsider the need for mechanical ventilation daily. As the authors suggest, ultrasonographic evaluation of the diaphragm is undoubtedly a powerful adjunct tool to predict weaning failure. This non-invasive technology offers the advantage of daily monitoring of the diaphragmatic function, assessing the progressive improvement over time of the patients and the readiness to wean from mechanical ventilation. However, ultrasonographic evaluation during the weaning needs to be integrated into defined protocols, which consider specific cutoff for predicting weaning success in patients with different illnesses, and in a comprehensive skillful strategy that envisions all possible causes of a difficult weaning. References 1. Boles JM, Bion J, Connors A, Herridge M, Marsh B, Melot C, et al. Weaning from mechanical ventilation. Eur Respir J 2007;29:1033-56. 2.Frutos-Vivar F, Esteban A, Apezteguia C, González M, Arabi Y, Restrepo MI, et al. Outcome of reintubated patients after scheduled extubation. J Crit Care 2011;26:502-9. 3. McCool FD, Tzelepis GE. Dysfunction of the diaphragm. N Engl J Med 2012;366:932-42. 4. Jung B, Moury PH, Mahul M, de Jong A, Galia F, Prades A, et al. Diaphragmatic dysfunction in patients with ICUacquired weakness and its impact on extubation failure. Intensive Care Med 2016;42:853-61. 5.Sieck GC, Mantilla CB. Effect of mechanical ventilation on the diaphragm. N Engl J Med 2008;358:1392-4. 6.Levine S, Nguyen T, Taylor N, Friscia ME, Budak MT, Rothenberg P, et al. Rapid disuse atrophy of diaphragm fibers in mechanically ventilated humans. N Engl J Med 2008;358:1327-35. 7. Dres M, Dubé B-P, Mayaux J, Delemazure J, Reuter D, Brochard L, et al. Coexistence and impact of limb muscle and diaphragm weakness at time of liberation from mechanical ventilation in medical icu patients. Am J Respir Crit Care Med 2016 [Epub ahead of print]. 8. Mayo P, Volpicelli G, Lerolle N, Schreiber A, Doelken P, Vieillard-Baron A. Ultrasonography evaluation during the weaning process: the heart, the diaphragm, the pleura and the lung. Intensive Care Med 2016;42:1107-17. Minerva Anestesiologica 1133 DE ROBERTIS Ultrasonography during weaning: a support to a comprehensive skilful strategy 9.Lerolle N, Guérot E, Dimassi S, Zegdi R, Faisy C, Fagon J-Y, et al. Ultrasonographic diagnostic criterion for severe diaphragmatic dysfunction after cardiac surgery. Chest 2009;135:401-7. 10. Flevari A, Lignos M, Konstantonis D, Armaganidis A. Diaphragmatic ultrasonography as an adjunct predictor tool of weaning success in patients with difficult and prolonged weaning. Minerva Anestesiol 2016;82:1149-57. 11. Boussuges A, Gole Y, Blanc P. Diaphragmatic motion studied by m-mode ultrasonography: methods, reproducibility, and normal values. Chest 2009;135:391400. 12. De Robertis E, Uttman L, Jonson B. Re-inspiration of CO2 from ventilator circuit: effects of circuit flushing and aspiration of dead space up to high respiratory rate. Crit Care 2010;14:R73. 13. Uttman L, Bitzén U, De Robertis E, Enoksson J, Johansson L, Jonson B. Protective ventilation in experimental acute respiratory distress syndrome after ventilatorinduced lung injury: a randomized controlled trial. Br J Anaesth 2012;109:584-94. 14. De Robertis E, Zito Marinosci G. The luxury of breathing oxygen. Minerva Anestesiol 2013;79:1324-5. 15.Iannuzzi M, De Sio A, De Robertis E, Piazza O, Servillo G, Tufano R. Different patterns of lung recruitment maneuvers in primary acute respiratory distress syndrome: effects on oxygenation and central hemodynamics. Minerva Anestesiol 2010;76:692-8. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: September 16, 2016. - Manuscript accepted: September 15, 2016. - Manuscript received: August 17, 2016. (Cite this article as: De Robertis E, Romano GM, Piazza O. Ultrasonography during weaning: a support to a comprehensive skilful strategy. Minerva Anestesiol 2016;82:1132-4) 1134 Minerva AnestesiologicaNovember 2016 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1135-7 EDITORIAL Perioperative hemodynamic therapy: goal-directed or meta-directed? Christopher UHLIG, Peter M. SPIETH* Department of Anesthesiology and Critical care Medicine, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany *Corresponding author: Peter Markus Spieth, Department of Anesthesiology and Critical Care Medicine, University Hospital Dresden, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany. E-mail: [email protected] P erioperative hemodynamic stability is one of the primary objectives during anesthesia. To achieve this goal, a blood pressure within a predefined range, fluid homeostasis as well as sufficient oxygen delivery is warranted to avoid impairment of organ function. To improve patient outcome different protocols have been developed focusing mainly on the use of fluids, red blood cells and vasoactive drugs to reach normal or in some cases supra-normal values of cardiac output and/or oxygen delivery. In the author’s opinion, many factors contribute to hemodynamic instability or impairment of postoperative organ function. The following factors are of paramount importance considering perioperative patient outcome: —— the patient. Individual preoperative assessment of the patients’ medical history, ASA status and preoperative risk factors are essential for a good outcome after surgery; —— the surgeon. Maybe the most important factor. The type of surgery and the resulting surgical trauma as well as the experience of the surgeon are key factors for patient survival. The type of surgery may force the anesthetists to be restrictive with fluids, since this influence postoperative outcome especially in surgical procedures where anastomoses of the Comment on p. 1199. Vol. 82 - No. 11 gastrointestinal tract are performed. The influence of the surgeon is often underestimated or underrepresented by evidence based medicine. Different types of surgery were often combined for trials performed by anesthetists and/ or the surgical procedures were performed by the same, usually experienced team. This may not reflect the reality especially in teaching hospitals. Since the surgeon is the main reason for intraoperative blood loss, duration of anesthesia and postoperative complications such as sepsis caused by impaired wound healing or insufficiency of anastomoses, these complications might by underrepresented in randomized controlled trials, or patients withdraw consent postoperatively due to the experience of a complication; —— the anesthesiologist. The experience of the anesthesiologist and his reaction to fast changing intraoperative conditions are also contributing to patient outcome. Like the surgeon, experience and patient centered decision making are the keys to a successful treatment of severe intraoperative hemorrhage or other complications like myocardial infarction, lung embolism or anaphylactic reaction; —— the team and resources. The experience of the whole team treating the patient intraoperatively, their interaction, their awareness and prediction of critical situations are crucial. Technical resources, such advanced he- Minerva Anestesiologica 1135 UHLIG PERIOPERATIVE HEMODYNAMIC THERAPY modynamic monitoring, devices for warming of infusion and blood products and access to adequate amount of blood products in case of severe hemorrhage; —— the goal. A standard established protocol for goal-directed therapy may help to guide the anesthesiologist during the procedure. Different protocols have been developed and investigated in randomized controlled trials over the last two decades. However, the definition of a goal for fluid therapy remains tricky since hemodynamic parameters like central venous pressure, pulse pressure variation or pulse contour derived cardiac output analysis are not reliable as single value or only valid in case of sinus rhythm in absence of cardiac valve pathology, respectively. In this issue of Minerva Anestesiologica, Giglio et al. performed a meta-analysis regarding the effects of goal-directed therapy protocols in a mixed surgical population consisting mainly of abdominal surgical patients on mortality.1 This meta-analysis was state of the art and is reported in accordance with the PRISMA statement.2 Giglio et al. analyzed 58 articles including the data of 8171 patients. Giglio et al. found that goal-directed protocols reduce perioperative mortality. In addition, a meta-regression model was used to determine the cut-off and the influence of risk of bias, mortality rate and publication year. Metaregression is a method which provides an extension of standard meta-analysis techniques, where the influence of statistical heterogeneity among results of multiple studies can be related to one or more characteristic of the trial.3 A subgroup analysis dividing the trials in two groups according to risk of bias and mortality in the control group, which were significant in the meta-regression, revealed that this effect is only consisting in the case of high risk of bias and a mortality in the control group greater than 10%. In trials with low risk of bias no statistic significant difference was detected. However, the analyses have several limitations. First of all, the risk of bias assessment was not correct according to the Cochrane collaboration, since the item “other risk of bias” was not assessed and the overall risk was not rated according to 1136 the GRADE approach.4, 5 Since most included trials had unclear risk of bias and only five trials fulfil the criteria for low risk of bias in all domains. The results of subgroup analysis related to risk of bias may be invalid. Second, mortality was not defined as a fixed time point, however the authors state that in-hospital mortality was considered in case of more than one type of mortality data. Furthermore, mortality might not be the favorable outcome parameter since pulmonary or extrapulmonary complications e.g. insufficiency of bowel anastomoses or cardiac events are more likely to be affected by goal directed therapy than mortality. Third, the analyses protocol was not registered before. Fourth, since death was a rare event in most trials the Peto odds ratio might be more appropriate for the analyses of binary outcomes.6, 7 Fifth, the goal parameters differ among trials as well as the type and amount of fluids and vasoactive drugs, which were no further analyzed. Sixth, no subgroup analysis among the different surgical procedures (e.g. cardiac surgery vs. non cardiac surgery). Since most trials had unclear or high risk of bias the results of the work performed by Giglio et al. should be interpreted with caution. In summary, we do not know whether goaldirected therapy improves patient survival since most randomized controlled trials are not powered for mortality as primary outcome. Interpreting the data of this meta-analysis is more likely that there is no effect on mortality. As pointed out by the authors there is a need for a large randomized controlled trial powered for mortality. Although goal-directed therapy may be a useful tool in high-risk patients, the experience and skills of surgeons and anesthetists are more likely to contribute to patient outcome. References 1. Giglio M, Manca F, Dalfino L, Brienza N. Perioperative hemodynamic goal-directed therapy and mortality: systematic review and meta-analysis with meta-regression. Minerva Anestesiol 2016;82:1199-213. 2. Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses. Br Med J 2009;339:b2535. Minerva Anestesiologica November 2016 PERIOPERATIVE HEMODYNAMIC THERAPYUHLIG 3. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Statistical heterogeneity in systematic reviews of clinical trials: A critical appraisal of guidelines and practice. J Health Serv Res Policy 2002;7:51-61. 4. Higgins JP, Altman DG, Gotsche PC, Jüni P, Moher D, Oxman AD, et al; Cochrane Bias Methods Group; Cochrane Statistical Methods Group. The Cochrane Collaboration’s tool for assessing risk of bias in randomized trials. Br Med J 2011;343:d5928. 5. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al; GRADE Working Group. GRADE: An emerging consensus on rating quality of evidence and strength of recommendations. Br Med J 2008;336:924-6. 6. Yusuf S, Peto R, Lewis J, Collins R, Sleight P. β-Blockade during and after myocardial infarction: An overview of the randomized trials. Prog Cardiovasc Dis 1985;27:335-71. 7. Brockhaus AC, Bender R, Skipka G. The Peto odds ratio viewed as a new effect measure. Stat Med J 2014;33:4861-74. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: September 16, 2016. - Manuscript accepted: September 15, 2016. - Manuscript revised: September 6, 2016. - Manuscript received: June 1, 2016. (Cite this article as: Uhlig C, Spieth PM. Perioperative hemodynamic therapy: goal-directed or meta-directed? Minerva Anestesiol 2016;82:1135-7) Vol. 82 - No. 11 Minerva Anestesiologica 1137 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1138-48 ORIGINAL ARTICLE Propofol-remifentanil anesthesia for upper airway endoscopy in spontaneous breathing patients: the ENDOTANIL Randomized Trial Guillaume BESCH 1, 2, Angeline CHOPARD-GUILLEMIN 1, Elisabeth MONNET 3, 4, Arnaud CAUSERET 1, Amelie JURINE 1, Gerald BAUDRY 1, Benjamin LASRY 1, Laurent TAVERNIER 5, 6, Emmanuel SAMAIN 1, 2, Sebastien PILI-FLOURY 1, 2 * 1Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, University of Franche-Comte, Besancon, France; 2EA 3920, INSERM SFR 133, Besancon, France; 3Clinical Investigation Center, University Hospital of Besancon, Besancon, France; 4SFR-FED 4234, University of Franche-Comte, Besancon, France; 5Department of ENT Surgery, University Hospital of Besancon, Besancon, France; 6EA 481 and SFR-FED 4234, University of Franche-Comte, Besancon, France *Corresponding author: Sebastien Pili-Floury, Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, 3 bvd Alexander Fleming, F-25000 Besancon, France. E-mail: [email protected] A B STRACT BACKGROUND: The ENDOTANIL Trial aimed at comparing an association of target-controlled infusion (TCI) of remifentanil and propofol to TCI of propofol alone on the clinical conditions during pan endoscopy for assessment of the upper airway (pan endoscopy) performed under tubeless general anesthesia. METHODS: This double-blind, single center, parallel, randomized, placebo-controlled trial was conducted in a French tertiary level of care, from June 2009 to February 2013. Patients scheduled for elective pan endoscopy were anesthetized using propofol TCI combined to either remifentanil TCI (effect-site concentration=1.5 ng.mL-1; remifentanil group) or placebo (control group). The main outcome measure was the percentage of clinically acceptable conditions for pan endoscopy, using a 5-criteria score (ease of laryngoscopy, position and movements of the vocal cords, cough and movements of the limbs to stimulation). The secondary outcomes were hemodynamic and respiratory safety. RESULTS: In this study 218 patients (mean±SD age 60 [10] yrs) were included. Clinically acceptable conditions were observed in 68% and 64% of the patients included in Remifentanil and Control group, respectively (P=0.39). None of the 5 parameters of the pan endoscopy score was significantly different between the 2 groups. Hemodynamic alterations were significantly lower in the Remifentanil as compared to the control group. Incidence of hypoxemia or need for rescue mechanical ventilation did not significantly differ between the 2 groups. CONCLUSIONS: The adjunction of remifentanil to propofol TCI, at a dose that maintain spontaneous breathing, did not improve the conditions for pan endoscopy, but attenuates the hemodynamic response induced by upper airway stimulation. (Cite this article as: Besch G, Chopard-Guillemin A, Monnet E, Causeret A, Jurine A, Baudry G, et al. Propofol-remifentanil anesthesia for upper airway endoscopy in spontaneous breathing patients: the ENDOTANIL Randomized Trial. Minerva Anestesiol 2016;82:1138-48) Key words: Anesthesia, General - Anesthesia, Intravenous - Endoscopy - Bronchoscopy - Laryngoscopy - Propofol. P an endoscopy for assessment of the upper aero digestive (pan endoscopy) tract consists in a three steps endoscopy: a direct larynComment in p. 1129. 1138 goscopy, a rigid bronchoscopy and an esophagoscopy, performed under general anesthesia. As endotracheal intubation is considered inadequate by many surgeons for both laryngoscopy and bronchoscopy, either high frequency Minerva AnestesiologicaNovember 2016 PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPYBESCH jet ventilation or spontaneous ventilation without endotracheal intubation (tubeless anesthesia) has been proposed during these first 2 steps of the procedure.1-3 Tubeless anesthesia requires cautious titration of short-acting anesthetic drugs to achieve a highly controlled anesthetic depth which warrants both optimal conditions for pan endoscopy and minimal risk of drug-induced apnea. The main challenge for anesthesiologists is to enable both optimal exposure of patient’s oropharyngeal tract and adequate oxygen supply. Total intravenous general anesthesia using target-controlled infusion (TCI) of propofol has been shown to be safe and efficient and is commonly used for the anesthetic management for pan endoscopy.4 However, the noxious stimulations observed during pan endoscopy may be similar to those described during endotracheal intubation performed by direct laryngoscopy and can be responsible of cough, movement of the patient, peri-procedure awareness, and/or haemodynamic alterations.5, 6 Coadministration of opioids could be considered to reduce the occurrence of these events,7, 8 and improve the conditions of pan endoscopy.9-12 Remifentanil is a powerful short-acting esterase-metabolized opioid whose pharmacokinetic profile could be well suited for the anesthetic management of pan endoscopy.13-15 Coadministration of remifentanil and propofol were reported to allow for titration of the anesthetic depth that enables adequate oxygen supply during spontaneous breathing.16, 17 The aim of the randomized, placebo-controlled ENDOTANIL Trial was to test the hypothesis that the addition of remifentanil to propofol TCI anesthesia improved the clinical conditions of the two first steps, laryngoscopy and bronchoscopy, in spontaneously breathing anaesthetized patients undergoing pan endoscopy. Materials and methods Design of the study The ENDOTANIL Trial was a randomized, placebo-controlled, double-blind, single- Vol. 82 - No. 11 center clinical study, conducted according to the French bioethics law (Art. L. 1121-1 of the law no. 2004-806, August 9th, 2004), at a University Hospital in France. The study was approved by the Institutional Review Board Est-II, University Hospital of Besancon, Besancon, France (no. 08/501, Chairperson Prof E. Haffen, MD, PhD) on April 9th, 2009. It was registered in the European Union Drug Regulating Authorities Clinical Trials (EudraCT no. 2008-007758-36) on November 18th, 2008. All patients gave written informed consent to participate to the study. Propofol (Diprivan®, AstraZeneca, London, UK) and remifentanil (Ultiva®, GlaxoSmithKline UK Ltd, Brentford, UK) used for anesthesia are drugs approved by the French Drug Regulatory Agency for human administration. All patients above 18 years, scheduled for pan endoscopy under general anesthesia between June 2009 and February 2013 were eligible. Non-inclusion criteria were: age <18 or >80 years, inability or refusal to give informed consent, pregnancy and/or breast feeding, long-term opioid treatment, known drug abuse or drug dependency, allergy to one of the study drugs, and chronic obstructive lung disease requiring oxygen therapy for more than 12 hours a day. Anesthetic management Upon arrival at operating room 1 hour after oral premedication using hydroxyzine 1 mg.kg-1, a standard monitoring was set up (General Electric Healthcare, Fairfield, CT, USA). Heart rate (HR), mean arterial pressure (MAP), respiratory rate (RR) and pulse oximetry (SpO2) values were collected before the induction of anesthesia and then every 3 min during the study period by an independent observer. After IRB approval on April 27th 2010, a BIS™ monitoring was added (Aspect BIS™ XP™ or BIS™ VISTA™, Aspect Medical Systems Inc., Newton, MA, USA) and BIS value measured before induction of anesthesia and just before laryngoscopy. The anesthesiologist in charge of the patient was blinded to BIS values throughout the study period. Minerva Anestesiologica 1139 BESCH PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPY A 20-gauge catheter was placed in a forearm vein and connected to a 3-way needle-free system (Smartsite®, Alaris Medical Systems, San Diego, CA, USA) allowing the infusion of both saline at a constant flow of 3 mL.kg-1.h-1 for hydration, and anesthetic drugs, delivered using a Base Primea™ infusion system (Fresenius Vial SAS, Brezins, France). After preoxygenation with 100% oxygen via a face mask (oxygen expiratory fraction >90%), anesthesia was induced with propofol TCI to achieve an initial propofol effect-site concentration (propofol-Ce) = 3 µg.mL-1, using Schnider pharmacokinetic model.18, 19 Infusion rate was then increased by step of 0.5 µg.mL-1 every 2 min, and the propofol-Ce at loss of consciousness (propofol-Ce LOC) was noted. Facial mask ventilation was tested, and the patient excluded in case of difficult or impossible facial mask ventilation. When stable spontaneous breathing was obtained, oxygen 3 L.min-1 was delivered via a nasal cannula, and maintained throughout the pan endoscopy. Then, the propofol-Ce was increased at a value equal to 1.3 * propofol-Ce LOC; and the infusion of the study drug (placebo or remifentanil according to randomization) was started. Study period ended when both direct laryngoscopy and rigid bronchoscopy were completed, and infusion of the study drug was stopped. Esophagoscopy was then performed after endotracheal intubation, using an anesthetic protocol managed by the attending anesthesiologist. Randomization and study drug administration Patients included in the study were randomly assigned the day before pan endoscopy into either Control or Remifentanil group. Randomization was performed in-block (ratio 1:1) using a computer-generated randomnumber table (nQuery Advisor® software v6.0, nQuery Advisor®, Los Angeles, CA, USA), prepared by our biostatistician (EM). Patients, investigators, practitioners in charge of patients, evaluators and data analysts were blinded to both block sizes of randomization and the study treatment administered. 1140 The study drug allocated by the randomization was prepared and delivered the day of the pan endoscopy by the Hospital pharmacist to the anesthesiologist in charge of the patient. The appearance and the volume of the syringes of placebo (40 mL of saline) and of remifentanil (2 mg of remifentanil diluted in 40 mL of saline, to obtain a remifentanil concentration of 50 µg.mL-1) were identical. Infusion rate was adjusted to reach a study drug effect-site concentration (study drug-Ce) = 1.5 ng.mL-1, calculated using Minto pharmacokinetic model.18 This study drug-Ce value was chosen as the median value allowing spontaneous breathing reported in the study by Murdoch et al., and was maintained steady throughout the study period.16 After steadystate effect-site concentrations were obtained for both drugs, the surgeon started the pan endoscopy. If a moderate hypoxemia and/or apnea lasting more than 60 sec occurred, both pan endoscopy and study drug infusion were stopped, propofol-Ce was decreased and oxygen delivery was increased to 5 L.min-1. If both a spontaneous breathing at a RR >8 c.min-1 and SpO2 value >95% were obtained in the following next minute, the study drug infusion was started again to reach a study drug-Ce = 0.75 ng.mL-1, i.e. half the former study drugCe. If the respiratory status worsened to severe hypoxemia and/or RR remained <8 c.min-1, emergency manual ventilation via a facial mask with 100% oxygen was performed. The decision for tracheal intubation or surgical tracheostomy was left to the discretion of the attending anesthesiologist. Data collected and endpoint measures Demographic data, past medical history, ASA physical status, Revised Cardiac Risk Index described by Lee et al.,20 were collected at inclusion. Tumor localization, extent and histology of the tumor were obtained after biopsies analysis. The condition for the pan endoscopy was assessed using a 5-criteria score (Table I), adapted from the clinical research practice Minerva AnestesiologicaNovember 2016 PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPYBESCH Table I.—Score to assess pan endoscopy* conditions during general anesthesia. Pan endoscopy conditions Variables Reactivity during pan endoscopy* Coughing during pan endoscopy* Movement of limbs during pan endoscopy* Initial vocal cords position at laryngoscopy Vocal cords movements during laryngoscopy Laryngoscopy Excellent Good Poor None None Abducted None Easy Diaphragm Slight Intermediate Moving Fair Sustained (>10 s) Vigorous Closed Closing Difficult Laryngoscopy: Easy = jaw relaxed, no resistance to blade during laryngoscopy; Fair = jaw not fully relaxed, slight resistance to blade; Difficult = poor jaw relaxation, active resistance by the patient. Condition for laryngoscopy and vocal cords position and movements were assessed by the surgeon, blinded to the study drug used. *Pan endoscopy consists in a three steps endoscopy for assessment of the upper aero digestive tract: a direct laryngoscopy, a rigid bronchoscopy and an esophagoscopy. Conditions were assessed during the first 2 steps of the pan endoscopy: the direct laryngoscopy and the rigid bronchoscopy. guidelines described by Viby-Mogensen et al.21 Condition of pan endoscopy was graded “excellent”, if all criteria were rated excellent; “good” if all criteria were at least good; or “poor” if at least one criteria was graded poor. The primary endpoint measure was the percentage of clinically acceptable conditions for pan endoscopy, defined as conditions graded as either excellent or good. The secondary endpoint measures were: 1) percentage of direct laryngoscopy or bronchoscopy interruption for inadequate anesthesia; 2) hemodynamic alterations observed during pan endoscopy, and 3) occurrence of respiratory adverse events, 4) complications related to pan endoscopy. Hemodynamic alterations were defined by the maximal and minimal variation (expressed in percent of preinduction value) of HR (∆-HRmax, and ∆-HRmin) and MAP (∆-MAPmax, and ∆-MAPmin) observed during the first 2 steps of pan endoscopy. Respiratory adverse events were defined by the number of episodes of moderate (SpO2 value <94%) or severe (SpO2 <90%) hypoxemia, and the need for rescue endotracheal intubation and mechanical ventilation. Pan endoscopy-related complications were defined as follows: significant hemorrhage and dental and/or mucosal injuries related to the endoscopic procedure or to the biopsy. A double data entry was done a posteriori by an independent data manager on a randomized sample of 22 patients to assess the reliability of the data collected. Vol. 82 - No. 11 Statistical analysis The Shapiro-Wilk Test was used to check whether each quantitative variable was normally distributed or not. Normally distributed and non-normally distributed quantitative variables, expressed respectively as mean (SD) and as median [interquartile range], have been compared between the Control and Remifentanil groups by using respectively the Student’s t-test and the Mann-Whitney U-Test. The intergroup comparisons for qualitative data, expressed as number of patients (percentage), were done by using the χ2 Test. All analyses were performed on an intention-to-treat basis. The reliability of the data collected by the two independent observers in the randomized sample of 22 patients was assessed using the non-parametric tests of agreement of the Cohen’s kappa coefficient for the qualitative variables and of the Kendall rank correlation coefficient for the quantitative variables (data not shown). A sample size of 222 patients was necessary to identify an increase in the percentage of clinically acceptable conditions for the pan endoscopy from 50% to 70% in the Remifentanil as compared to Control group, with a two-sided type I error of 0.05 and a power of 90%. We planned to include 256 patients in case of patients drop out. Two interim analysis were performed by an independent biostatistician (EM) after the inclusion of 80 and 160 patients, with stopping boundaries allowing early termination of the study, as described by O’Brien and Minerva Anestesiologica 1141 BESCH PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPY Fleming (P=0.0006 and P=0.016 for the 1st and the 2nd interim analysis, respectively).22 Statistical analyses were performed using SAS 9.3 software (SAS Institute Inc., Cary, NC, USA). All P-value are two-sided and P<0.05 was considered statistically significant. Results Four hundred and fifty eight patients were scheduled for pan endoscopy during the study period. As no interim analysis reached the statistical significance, the study was conducted until 256 patients, were randomized in either Control or Remifentanil groups. Two hundred and eighteen patients were finally analyzed (Figure 1). Demographic data, co-morbidities, and type and location of tumors of the 2 groups are given in Table II. Direct laryngoscopy and bronchoscopy could be completed in all patients while maintaining spontaneous breathing without endotracheal intubation, and a biopsy was done in 216 cases. The results of intraoperative measures are given in Table III. The values of propofol-Ce LOC, total dose Table II.—Demographic data of patients in the CONTROL and in the REMIFENTANIL groups. CONTROL group (N.=106 patients) REMIFENTANIL group (N.=112 patients) Age (yr) 60 (10) 60 (9) BMI¥† (kg.m-²) 23 [21-26] 24 [21-27] Male gender* 89 (84) 91 (81) Obesity* (defined as BMI† ≥ 11 (10) 9 (8) 30 kg.m-²) History of chronic alcohol 60 (57) 64 (57) abuse* ASA§ physical status 28 (26) 25 (22) III-IV* Revised cardiac risk index‡ 100 (94)/6 (6) 110 (98)/2 (2) value 0-1/≥ 2* Type of tumour Squamous cell carcinoma* 90 (86) 85 (75) Oral cavity or larynx* 58 (55) 43 (38) Naso-, oro- or 44 (42) 39 (35) hypo-pharynx* Esophagus* 1 (1) 1 (1) Multiple tumour sites* 6 (6) 5 (4) Other type of tumour* 13 (12) 24 (21) Negative biopsy* 5 (5) 7 (6) Values are mean (SD). ¥Values are median [interquartile range]. *Values are number (proportion). †BMI: Body Mass Index = weight/height². †‡Revised cardiac risk index is calculated according to Lee et al.19 §ASA: American Society of Anesthesiologists. Patients scheduled for pan endoscopy: N.=458 Patients randomized: N.=256 Patients allocated to control group: N.=128 Patients excluded: N.=22 — Consents withdrawal: N.=3 — Pan endoscopy cancelled: N.=9 — Preoperative death*: N.=0 — Incomplete data: N.=8 — Difficult face mask ventilation: N.=2 Patients analyzed: N.=106 Patients allocated to Remifentanil group: N.=128 Patients excluded: N.=16 — Consents withdrawal: N.=2 — Pan endoscopy cancelled: N.=5 — Preoperative death*: N.=1 — Incomplete data: N.=6 — Difficult face mask ventilation: N.=2 Patients analyzed: N.=112 Figure 1.—Flow chart of patient inclusions in the study. *preoperative pulmonary embolism. Pan endoscopy consists in a three steps endoscopy for assessment of the upper aero digestive tract consists: a direct laryngoscopy, a rigid bronchoscopy and an esophagoscopy. 1142 Minerva AnestesiologicaNovember 2016 PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPYBESCH Table III.—Intraoperative parameters of patients in the CONTROL and in the REMIFENTANIL groups. CONTROL group (N.=106) Anesthesia parameters Infusion of the study drug Total volume infused (mL) Total dose infused (µg.kg-1.min-1) Total dose infused (µg) Duration of the infusion (min) Infusion of propofol Total dose infused (mg.kg-1.min-1) Total dose infused (mg) Duration of the infusion (min) Propofol-Ce LOC ‡(µg.ml-1) BIS values**† BISt0† BISlaryngo† Safety parameters Hemodynamic safety§ HRmax (bpm) HRmin (bpm) HRt0 (bpm) Δ-HRmax (%) Δ-HRmin (%) MAPmax (mmHg) MAPmin (mmHg) MAPt0 (mmHg) Δ-MAPmax (%) Δ-MAPmin (%) Respiratory safety¶ Moderate hypoxemia* Severe hypoxemia* Surgical complications* REMIFENTANIL group (N.=112) P value 1.17 [0.95-1.43] 0 0 10 [8-14] 1.18 [0.96-1.44] 0.085 [0.074-0.102] 59.2 [48.0-72.0] 10 [7-13] 0.94 0.56 [0.45-0.71] 525 [393-700] 14 [11-17] 5.0 [4.0-6.5] 0.53 [0.42-0.67] 500 [379-674] 13 [10-17] 5.5 [4.0-6.3] 0.37 0.80 0.54 0.72 97 [96-98] 29 [23-40] 97 [94-98] 27 [24-37] 91 [80-100] 80 [67-87] 68 [62-81] 31 [13-44] 10 [0-24] 105 [95-122] 89 [77-100] 103 [93-113] 5 [(-6)-20] 13 [5-24] 90 [79-100] 78 [66-85] 74 [65-85] 17 [8-33] 1 [(-5)-11] 101 [90-115] 81 [71-94] 101 [92-115] 0 [(-13)-12] 21 [12 -29] 19 (18) 11 (11) 4 (4) 25 (22) 11 (10) 1 (1) 0.53 0.13 0.78 0.71 0.31 0.01 0.0016 0.0001 0.06 0.0022 0.94 0.02 0.002 0.44 0.87 0.20 Values are median [interquartile range]. *Values are number (proportion). †BIS: bispectral index. BISt0 and BISlaryngo are BIS values before induction of anesthesia and just before the laryngoscopy, respectively. ‡ propofol-Ce LOC: effect-site concentration of propofol predicted by the Schnider model at loss of consciousness during induction of anesthesia. §t0, max and min values for heart rate (heart rate) and MAP (mean arterial pressure) are values observed before induction of anesthesia, highest and lowest values during the procedure, respectively. Δ-HRmax, Δ-HRmin, Δ-MAPmax and Δ-MAPmin are the maximal and minimal variation (expressed in percent of preinduction value) of HR (∆-HRmax, and ∆-HRmin) and MAP (∆-MAPmax, and ∆-MAPmin) observed during the first 2 steps of the pan endoscopy: the direct laryngoscopy and the rigid bronchoscopy. ¶Moderate hypoxemia and severe hypoxemia are defined by a decrease in SpO2 <94% and <90%, respectively. **Data obtained in 87 and 95 patients in the CONTROL and REMIFENTANIL groups, respectively. Pan endoscopy consists in a three steps endoscopy for assessment of the upper aero digestive tract: a direct laryngoscopy, a rigid bronchoscopy and an esophagoscopy. of propofol infused, average dose of propofol, total volume of study-drug infused, were not different between the 2 groups (Table III). BIS value just before laryngoscopy, obtained in 182 patients (respectively 87 and 95 patients in the Control and in the Remifentanil groups) was not significantly different in the 2 groups (Table III). Clinically acceptable conditions for pan endoscopy (primary endpoint) were observed in 77 (68%) of the Remifentanil patients and 67 (64%) of Control patients, a difference not significant (P=0.39). None of the 5 parameters of Vol. 82 - No. 11 the score (cough, movement of limbs, position of the vocal cords, movements of the vocal cords, laryngoscopy) were significantly different between the groups (Figure 2). The hemodynamic response to endoscopyrelated noxious stimulation was significantly lower in the Remifentanil group as compared to the Control group (Table III). A significantly larger decrease in arterial blood pressure was observed in the Remifentanil group in comparison to the Control group (Table III). The incidence of respiratory adverse events or surgical complications was not different between Minerva Anestesiologica 1143 BESCH PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPY P=0.09 P=0.33 P=0.36 P=0.32 P=0.32 Figure 2.—Intergroup comparison of the 5 parameters of the score used to assess pan endoscopy conditions during general anesthesia. No significant difference between groups. Pan endoscopy consists in a three steps endoscopy for assessment of the upper aero digestive tract: a direct laryngoscopy, a rigid bronchoscopy and an esophagoscopy. Conditions were assessed during the first 2 steps of the pan endoscopy: the direct laryngoscopy and the rigid bronchoscopy. the 2 groups. No patient required emergency endotracheal intubation or surgical tracheostomy during the study period. Discussion The result of this study showed that adjunction of remifentanil to a TCI of propofol, at a dose that allow spontaneous breathing did not improve the clinical conditions for the pan endoscopy compared to the TCI of propofol alone. Hemodynamic response to the noxious stimulation of pan endoscopy was attenuated by remifentanil and the incidence of moderate or severe hypoxemia was not increased. ENDOTANIL trial aimed to assess the combination of both TCI propofol and remifentanil in tubeless anesthesia with spontaneous breathing in patients undergoing pan endoscopy. In our study, the incidence of clinically acceptable conditions for pan endoscopy was not increased by the addition of remifentanil. A beneficial effect was expected as a dose- 1144 dependent improvement in the clinical conditions of procedures such as laryngoscopy or endotracheal intubation has been described in several papers.8, 23 Several hypotheses could be raised to explain the lack of effect we observed in our study. First, the dose of remifentanil administrated to the patient may be too low to add a beneficial effect to propofol anesthesia. As a matter of fact, 1) the Ce of remifentanil reported in the literature to produce excellent conditions for endotracheal intubation, were higher than the Ce we used in the ENDOTANIL trial,9-12, 24 and 2) the continuous infusion of remifentanil was frequently preceded by an initial bolus dose.25-27 Other authors have reported that spontaneous breathing could be maintained during fiberoptic intubation or flexible bronchoscopy by using remifentanil alone at a mean effect-site concentration as high as 2.4 ng.mL-1 and 2.5 ng.mL-1 respectively.28, 29 However, we think that these higher doses of remifentanil could not be used safely for tubeless management of Minerva AnestesiologicaNovember 2016 PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPYBESCH general anesthesia for the pan endoscopy that requires spontaneous breathing.30 Remifentanil has depressive respiratory effect that is dose-dependent and the association of propofol and remifentanil increases the risk of respiratory depression in a synergistic manner.17, 30 Thus, we chose a remifentanil-Ce equal to 1.5 ng.mL-1, i.e. the median value reported by Murdoch et al. to perform spontaneous breathing sedation for flexible fiber optic bronchoscopy.16 In our study, this Ce of remifentanil allowed to obtain the same low rate of druginduced apnea and/or of facial mask ventilation requirement. We assume that half of the patients included in the study of Murdoch et al. maintained a spontaneous breathing with higher doses of remifentanil and that using higher dose of remifentanil could provide different results on both hemodynamics and quality of pan endoscopy. Second, the relative role of the analgesic and the hypnotic components of anesthesia on the response to several noxious stimuli has been studied graphically for several noxious procedures, using a three-dimension response surface modeling of the response/no response to a standardized stimulation (1st dimension) versus the corresponding remifentanil-Ce (2nd dimension: analgesia axis) versus propofolCe (3rd dimension: hypnotic axis).11, 12, 31 Interestingly, Kern et al. reported that the morphologic feature of the response surface for laryngoscopy was skewed toward the hypnotic axis and the difference in the probability of no response to laryngoscopy at a concentration of propofol equal to 5 µg.mL-1 (mean value in our study) appeared very low between patient receiving or not an infusion of remifentanil.11 Third, we used the pharmacokinetic models of Schnider and of Minto for the TCI of propofol and of remifentanil, respectively.18, 19 These pharmacokinetic models have been commonly used in clinical studies to assess the association of propofol and remifentanil in many clinical situations, including laryngoscopy and endotracheal intubation.31, 32 However, the models have been validated in healthy volunteers, and the environment of the operating room Vol. 82 - No. 11 may have generated a stress-induced increase in cardiac output that potentially may modify drug distribution.33, 34 The beneficial effect of either low or high doses of remifentanil on the cardiovascular responses observed during laryngoscopy, rigid bronchoscopy or endotracheal intubation has been previously described in elective surgical patients.8, 23, 35-39 In our study, the infusion of remifentanil lowered the increase of the heart rate during pan endoscopy without any clinically relevant hypotension. Thus, the use of opioids during pan endoscopy could have a potential interest to prevent perioperative myocardial ischemia in this population of smokers at risk of undiagnosed coronary artery disease.40, 41 As a continuous monitoring of capnography was not performed during the ENDOTANIL trial, the number of apneic events could not be reliably measured. We have chosen to focus on the clinically relevant apneic episodes that were responsible for hypoxemia. As none of the patients included in the study were hypoxic at baseline, we assume that all episodes of moderate or severe hypoxemia could potentially be related to drug-induced respiratory depressive effect. Considering the lack of difference between the 2 groups, we suppose that the moderate and severe hypoxic episodes were mainly related to the propofol infusion and to the presence of the endoscope in the upper airway during rigid bronchoscopy that could have been responsible for significant airway obstruction. Paralyzing patients during rigid bronchoscopy is matter of debate. The anesthetic protocol in the ENDOTANIL trial aimed to maintain spontaneous breathing during direct laryngoscopy and rigid bronchoscopy to provide adequate oxygen supply during the procedure. Therefore, neuromuscular blocking agents were not used to avoid apnea and its consequences. Limitations of the study In the absence of specific score developed to assess the conditions for the pan endos- Minerva Anestesiologica 1145 BESCH PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPY copy, the pan endoscopy-score was adapted from previously published tools that are based on the good clinical research practice guidelines.21 We hypothesized that the stimulations of the upper airway observed during the direct laryngoscopy or the rigid bronchoscopy of the pan endoscopy were close to those induced by laryngoscopy and endotracheal intubation. However, we cannot exclude that the absence of effect of remifentanil on the conditions for the pan endoscopy can only be explained by an inadequate assessment tool. In this regard, pan endoscopy-score may be considered too sensitive, as, although only 2/3 of the patients had clinically acceptable condition as assessed by the pan endoscopyscore, the endoscopic exploration of the upper airway and the biopsies during the procedure could be completed in all patients of the study. Nonetheless, movements of the limbs and/or vocal cords could occur in patients with clinically acceptable conditions. The results of our study could not support that therapeutic intervention, such as laser treatment or injection into the vocal cords, could be safely performed by using the same anesthetic protocol. Finally, the intra and inter observer variability of the pan endoscopy-score has not been assessed and could have impact the results of the study. The same Ce of remifentanil was used in all patients included in the Remifentanil group, for the reasons explained above. A dose adapted to each patient, using recently developed pain monitoring may have produced better results.41 The increase in the effect-site concentration of propofol just before the beginning of the pan endoscopy has been chosen arbitrarily and could have contributed to an overdosing. Considering that the same anesthetic protocol has been applied in the 2 groups, we assume that it has probably no impact on the results of the study. Our study was not designed to assess a reduction in intraoperative awareness by adding remifentanil to TCI of propofol. Even if a higher rate of this event has been reported in ENT surgery, it remains infrequent. Ran- 1146 domized controlled trials aimed to test this hypothesis should include a very large number of patients to reach a sufficient statistical power. Conclusions In this study, addition of remifentanil to propofol TCI anesthesia, at a dose that maintains spontaneous breathing did not improve clinical condition for laryngoscopy and bronchoscopy during pan endoscopy, although it blunted partially hemodynamic response during the procedure. Further studies are required to assess whether titration of remifentanil could improve condition for pan endoscopy. Key messages —— The adjunction of low doses of remifentanil to target-controlled infusion of propofol failed to significantly improve the conditions of direct laryngoscopy and rigid bronchoscopy during pan endoscopic assessment of the upper aero digestive tract. —— The infusion of low doses of remifentanil during pan endoscopy did not increase the risk for respiratory adverse events. —— Remifentanil could lower the hemodynamic response to direct laryngoscopy and rigid bronchoscopy. References 1.Bourgain JL, Desruennes E, Fischler M, Ravussin P. Transtracheal high frequency jet ventilation for endoscopic airway surgery: A multicentre study. Br J Anaesth 2001;87:870-5. 2. Jaquet Y, Monnier P, Van Melle G, Ravussin P, Spahn DR, Chollet-Rivier M. Complications of different ventilation strategies in endoscopic laryngeal surgery: A 10year review. Anesthesiology 2006;104:52-9. 3.Perrin G, Colt HG, Martin C, Mak MA, Dumon JF, Gouin F. Safety of interventional rigid bronchoscopy using intravenous anesthesia and spontaneous assisted ventilation. A prospective study. Chest 1992;102:1526-30. 4. Passot S, Servin F, Allary R, Pascal J, Prades JM, Auboyer C, et al. Target-controlled versus manually-controlled infusion of propofol for direct laryngoscopy and bronchoscopy. Anesth Analg 2002;94:1212-6. 5. Billard V, Moulla F, Bourgain JL, Megnigbeto A, Stan- Minerva AnestesiologicaNovember 2016 PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPYBESCH ski DR. Hemodynamic response to induction and intubation. Propofol/fentanyl interaction. Anesthesiology 1994;81:1384-93. 6. Kelly JS, Roy RC. Intraoperative awareness with propofol-oxygen total intravenous anesthesia for microlaryngeal surgery. Anesthesiology 1992;77:207-9. 7. Kazama T, Ikeda K, Morita K. Reduction by fentanyl of the cp50 values of propofol and hemodynamic responses to various noxious stimuli. Anesthesiology 1997;87:21327. 8.Thompson JP, Hall AP, Russell J, Cagney B, Rowbotham DJ. Effect of remifentanil on the haemodynamic response to orotracheal intubation. Br J Anaesth 1998;80:467-9. 9. Bouillon TW, Bruhn J, Radulescu L, Andresen C, Shafer TJ, Cohane C, et al. Pharmacodynamic interaction between propofol and remifentanil regarding hypnosis, tolerance of laryngoscopy, bispectral index, and electroencephalographic approximate entropy. Anesthesiology 2004;100:1353-72. 10.Grant S, Noble S, Woods A, Murdoch J, Davidson A. Assessment of intubating conditions in adults after induction with propofol and varying doses of remifentanil. Br J Anaesth 1998;81:540-3. 11. Kern SE, Xie G, White JL, Egan TD. A response surface analysis of propofol-remifentanil pharmacodynamic interaction in volunteers. Anesthesiology 2004;100:137381. 12. Mertens MJ, Olofsen E, Engbers FH, Burm AG, Bovill JG, Vuyk J. Propofol reduces perioperative remifentanil requirements in a synergistic manner: Response surface modeling of perioperative remifentanil-propofol interactions. Anesthesiology 2003;99:347-59. 13.Egan TD, Lemmens HJ, Fiset P, Hermann DJ, Muir KT, Stanski DR, et al. The pharmacokinetics of the new short-acting opioid remifentanil (gi87084b) in healthy adult male volunteers. Anesthesiology 1993;79:881-92. 14. Westmoreland CL, Hoke JF, Sebel PS, Hug CC, Jr., Muir KT. Pharmacokinetics of remifentanil (gi87084b) and its major metabolite (gi90291) in patients undergoing elective inpatient surgery. Anesthesiology 1993;79:893-903. 15. Mastronardi P, Dellacasa P. Observational study on the use of remifentanil in general anesthesia. Drug utilisation research. Minerva Anestesiol 2004;70:605-16. 16. Murdoch JA, Hyde RA, Kenny GN. Target-controlled remifentanil in combination with propofol for spontaneously breathing day-case patients. Anaesthesia 1999;54:1028-31. 17. Peacock JE, Luntley JB, O’Connor B, Reilly CS, Ogg TW, Watson BJ, et al. Remifentanil in combination with propofol for spontaneous ventilation anaesthesia. Br J Anaesth 1998;80:509-11. 18. Minto CF, Schnider TW, Shafer SL. Pharmacokinetics and pharmacodynamics of remifentanil. Ii. Model application. Anesthesiology 1997;86:24-33. 19.Schnider TW, Minto CF, Gambus PL, Andresen C, Goodale DB, Shafer SL, et al. The influence of method of administration and covariates on the pharmacokinetics of propofol in adult volunteers. Anesthesiology 1998;88:1170-82. 20.Lee TH, Marcantonio ER, Mangione CM, Thomas EJ, Polanczyk CA, Cook EF, et al. Derivation and prospective validation of a simple index for prediction of cardiac risk of major noncardiac surgery. Circulation 1999;100:1043-9. 21.Viby-Mogensen J, Engbaek J, Eriksson LI, Gramstad L, Jensen E, Jensen FS, et al. Good clinical research practice (gcrp) in pharmacodynamic studies of neuromuscular blocking agents. Acta Anaesthesiol Scand 1996;40:5974. 22. Fleming TR. One-sample multiple testing procedure for phase ii clinical trials. Biometrics 1982;38:143-51. Vol. 82 - No. 11 23.Hall AP, Thompson JP, Leslie NA, Fox AJ, Kumar N, Rowbotham DJ. Comparison of different doses of remifentanil on the cardiovascular response to laryngoscopy and tracheal intubation. Br J Anaesth 2000;84:1002. 24. Bouvet L, Stoian A, Rimmele T, Allaouchiche B, Chassard D, Boselli E. Optimal remifentanil dosage for providing excellent intubating conditions when co-administered with a single standard dose of propofol. Anaesthesia 2009;64:719-26. 25.Stevens JB, Wheatley L. Tracheal intubation in ambulatory surgery patients: Using remifentanil and propofol without muscle relaxants. Anesth Analg 1998;86:45-9. 26.Troy AM, Huthinson RC, Easy WR, Kenney GN. Tracheal intubating conditions using propofol and remifentanil target-controlled infusions. Anaesthesia 2002;57:1204-7. 27. Klemola UM, Mennander S, Saarnivaara L. Tracheal intubation without the use of muscle relaxants: Remifentanil or alfentanil in combination with propofol. Acta Anaesthesiol Scand 2000;44:465-9. 28.Lallo A, Billard V, Bourgain JL. A comparison of propofol and remifentanil target-controlled infusions to facilitate fiberoptic nasotracheal intubation. Anesth Analg 2009;108:852-7. 29.Chalumeau-Lemoine L, Stoclin A, Billard V, Laplanche A, Raynard B, Blot F. Flexible fiberoptic bronchoscopy and remifentanil target-controlled infusion in icu: A preliminary study. Intensive Care Med 2013;39:53-8. 30.Nieuwenhuijs DJ, Olofsen E, Romberg RR, Sarton E, Ward D, Engbers F, et al. Response surface modeling of remifentanil-propofol interaction on cardiorespiratory control and bispectral index. Anesthesiology 2003;98:312-22. 31. Johnson KB, Syroid ND, Gupta DK, Manyam SC, Egan TD, Huntington J, et al. An evaluation of remifentanil propofol response surfaces for loss of responsiveness, loss of response to surrogates of painful stimuli and laryngoscopy in patients undergoing elective surgery. Anesth Analg 2008;106:471-9. 32.Trabold F, Casetta M, Duranteau J, Albaladejo P, Mazoit JX, Samii K, et al. Propofol and remifentanil for intubation without muscle relaxant: The effect of the order of injection. Acta Anaesthesiol Scand 2004;48:35-9. 33.Avram MJ, Krejcie TC, Henthorn TK, Niemann CU. Beta-adrenergic blockade affects initial drug distribution due to decreased cardiac output and altered blood flow distribution. J Pharmacol Exp Ther 2004;311:617-24. 34. Kurita T, Uraoka M, Jiang Q, Suzuki M, Morishima Y, Morita K, et al. Influence of cardiac output on the pseudo-steady state remifentanil and propofol concentrations in swine. Acta Anaesthesiol Scand 2013;57:754-60. 35.Guignard B, Menigaux C, Dupont X, Fletcher D, Chauvin M. The effect of remifentanil on the bispectral index change and hemodynamic responses after orotracheal intubation. Anesth Analg 2000;90:161-7. 36.Prakash N, McLeod T, Gao Smith F. The effects of remifentanil on haemodynamic stability during rigid bronchoscopy. Anaesthesia 2001;56:576-80. 37.Albertin A, Casati A, Deni F, Danelli G, Comotti L, Grifoni F, et al. Clinical comparison of either small doses of fentanyl or remifentanil for blunting cardiovascular changes induced by tracheal intubation. Minerva Anestesiol 2000;66:691-6. 38.Iannuzzi E, Iannuzzi M, Cirillo V, Viola G, Parisi R, Cerulli A, et al. Peri-intubation cardiovascular response during low dose remifentanil or sufentanil administration in association with propofol tci. A double blind comparison. Minerva Anestesiol 2004;70:109-15. 39.Iannuzzi E, Iannuzzi M, Cirillo V, Viola G, Parisi R, Chiefari M. Small doses of remifentanil and alfetanil in continuous total intravenous anesthesia in major abdomi- Minerva Anestesiologica 1147 BESCH PROPOFOL-REMIFENTANIL ANESTHESIA FOR UPPER AIRWAY ENDOSCOPY nal surgery. A double blind comparison. Minerva Anestesiol 2003;69:127-36. 40. Warltier DC, Pagel PS, Kersten JR. Approaches to the prevention of perioperative myocardial ischemia. Anesthesiology 2000;92:253-9. 41. Boselli E, Bouvet L, Begou G, Torkmani S, Allaouchiche B. Prediction of hemodynamic reactivity during total intravenous anesthesia for suspension laryngoscopy using analgesia/nociception index (ani): A prospective observational study. Minerva Anestesiol 2015;81:288-97. Authors’ contributions.—Guillaume Besch had substantial contributions to 1) design of the study, and acquisition and interpretation of data; and 2) drafting the article manuscript. Angeline Chopard-Guillemin had substantial contributions to 1) conception and design of the study, and acquisition and interpretation of data; and 2) drafting the article manuscript. ������������������������������������� Elisabeth Monnet��������������������� had substantial contributions to 1) methodological design, and analysis and interpretation of data; and 2) revising the manuscript critically for important intellectual content. Arnaud Causeret had substantial contributions to 1) design of the study, acquisition and interpretation of data; and 2) revising the manuscript critically for important intellectual content. Amelie Jurine had substantial contributions to 1) acquisition and interpretation of data; and 2) revising the manuscript critically for important intellectual content. Gerald Baudry had substantial contributions to 1) acquisition and interpretation of data; and 2) revising the manuscript critically for important intellectual content. Benjamin Lasry had substantial contributions to 1) acquisition and interpretation of data; and 2) revising the manuscript critically for important intellectual content. Laurent Tavernier had substantial contributions to 1) conception and design of the study and interpretation of data; and 2) drafting the article. Emmanuel Samain had substantial contributions to 1) conception and design of the study and interpretation of data; and 2) drafting the article. Sebastien Pili-Floury had substantial contributions to 1) conception of the study, and analysis and interpretation of data; and 2) drafting the manuscript. Funding.—The work was not funded by: National Institutes of Health (NIH), Howard Hughes Medical Institute (HHMI), Medical Research Council (MRC), and/or Wellcome Trust. The work was not supported by official funding or a grant. The financial cost of the study was supported by the Department of Anesthesiology and Intensive Care Medicine, 3 Bvd Alexander Fleming, University Hospital of Besancon, F-25000 Besancon, France. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Congresses.—Data have been presented at the French Society of Anesthesia and Intensive Care Medicine Annual Meeting in Paris, France, on September 2014, and at the American Society of Anesthesiologists Annual Meeting in New Orleans, Louisiana, USA, on October 2014. Acknowledgements.—We would like to thank Dr Eviane Farah for her assistance in preparing the manuscript. Article first published online: June 17, 2016. - Manuscript accepted: June 15, 2016. - Manuscript revised: May 23, 2016. - Manuscript received: March 3, 2016. 1148 Minerva AnestesiologicaNovember 2016 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1149-57 ORIGINAL ARTICLE Diaphragmatic ultrasonography as an adjunct predictor tool of weaning success in patients with difficult and prolonged weaning Aikaterini FLEVARI *, Michael LIGNOS, Dimitrios KONSTANTONIS, Apostolos ARMAGANIDIS Second University Critical Care Clinic, Attikon University Hospital, Athens, Greece *Corresponding author: Aikaterini Flevari, 1 Rimini str., Chaidari, Athens, 12462, Greece. E-mail: [email protected] A B STRACT BACKGROUND: Diaphragmatic ultrasonography has been recently considered as a new weaning predictor method. Previous studies checked diaphragmatic excursion and thickness during quiet or deep breathing in unselected populations of critically ill patients. Our study aimed to investigate diaphragmatic excursion during quiet and unassisted breathing, in comparison to standard predictor tools, such as Rapid Shallow Breathing Index (RSBI) and Maximal Inspiratory Pressure (Pimax), in patients with difficult and/or prolonged weaning. METHODS: Patients with difficult and/or prolonged weaning, who met the criteria for spontaneous breathing trial, were assessed. The excursion of each hemidiaphragm (DEx) was evaluated by B-mode and M-mode ultrasonography while patient was on quiet breathing and at supine position. RSBI and Pimax were simultaneously recorded and weaning outcome was recorded. RESULTS: Thirteen male and fourteen female patients were included. DEx [median and interquartile range, mm] was 14 (8.5-22) for the right hemidiaphragm (RDEx) and 12 (7-23) for the left (LDEx). We found no difference in DEx between sexes. Among the four weaning predictor tools, LDEx at a cut-off 10 mm was the best index to predict weaning success (sensitivity 86%, specificity 85%, Negative Predictive Value 94%). The optimal cut-off values, as determined by the area under the receiver operating characteristic curve, were 10 mm for RDEx, 7 mm for LDEx, 57 breaths/min/L for RSBI and -20 cmH2O for Pimax. CONCLUSIONS: Our results suggest that DEx threshold of 10 mm and 7 mm for right and left hemidiaphragms respectively could be used as adjunct tool in the predictive algorithm of weaning in difficult to wean patients. (Cite this article as: Flevari A, Lignos M, Konstantonis D, Armaganidis A. Diaphragmatic ultrasonography as an adjunct predictor tool of weaning success in patients with difficult and prolonged weaning. Minerva Anestesiol 2016;82:1149-57) Key words: Ventilator weaning - Ultrasonography - Diaphragm. A lthough the process of discontinuing (weaning) mechanical ventilation (MV) remains one of the most challenging steps throughout the period of MV, weaning predictor tools often fail to attain their role. Weaning covers the entire process of liberating patients from MV until the final extubation. In 2005 a Task Force of experts came to a consensus Comment in p. 1132. Vol. 82 - No. 11 regarding guidelines for “Weaning from Mechanical Ventilation”. Tobin therein defined weaning as a six step process: treatment of the underlying condition (stage 1), suspicion that the patient is ready to wean (stage 2), assessment of the readiness to wean (stage 3), spontaneous breathing trial (SBT) (stage 4), extubation (stage 5) and assessment of probable reintubation (stage 6).1 In stage 3, apart from clinical stability which implies resolution Minerva Anestesiologica 1149 FLEVARI DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANING of the acute phase disease and cardiovascular, metabolic and neuropsychiatric stability, a series of objective measurements are continuously monitored in order to assess weaning capability. Commonly used parameters include Rapid Shallow Breathing Index, defined as the ratio of respiratory frequency to tidal volume (RSBI=f/VT), Maximum Inspiratory Pressure (Pimax), pulmonary gas exchange (e.g. PaO2/ FiO2, PaCO2), Vital Capacity, Minute Ventilation and static Compliance.2-4 Among them, RSBI has been acknowledged as the most important predictor of weaning failure with best cut-off value that of 105 breaths/min/L.2, 5 On the other hand, weaning process is not always straightforward and therefore Brochard in the same consensus statement introduced a classification of critically-ill patients based on the difficulty and length of weaning process.1 Difficult weaning may in fact be due to different or mixed etiologies, the diagnosis of which requires meticulous monitoring of all the aforementioned parameters. The commonly missed diagnosis of diaphragmatic dysfunction, from weakness to total paralysis, required until now strenuous methods, such as electrical or magnetic phrenic nerve stimulation, or thoracic fluoroscopy. During the last decade, portable ultrasonography has become an invaluable tool, since it is a simple, non-invasive and reproducible method of imaging, introducing a new and less studied weaning predictor tool: the diaphragmatic ultrasonography.5, 6 Several researches have studied diaphragmatic motion,7-10 but none of them has been performed in the intriguing sub-population of critically ill patients with difficult and prolonged weaning. Aim of this study was to test if ultrasonographic M-mode diaphragmatic excursion (DEx), during regular, quiet and effortless breathing, as compared to standard predictors, (RSBI and Pimax), could be a successful extubation tool in difficult to wean critically ill patients. Materials and methods Setting and ethical standards This is a prospective cohort clinical trial, conducted in a general university hospital 1150 adult Intensive Care Unit (ICU) from June to December 2014, in accordance with the principals enunciated in the World Medical Association Declaration of Helsinki. All data were part of routine treatment and their collection involved no risk for the subjects, whose anonymity has been preserved. The study was approved by the hospital’s Ethics Committee (No 9-13-16/May/14), before patients’ recruitment. For the aforementioned reasons the Committee waived the need for obtaining informed consent. Study population Demographic characteristics were recorded: sex, age, Body Mass Index (BMI), and Acute Physiology and Chronic Health Evaluation (APACHE II) Score on admission to ICU. Figure 1 shows flow diagram of patient recruitment. As per study protocol, among all patients who underwent SBT, only patients with difficult and prolonged weaning were eligible for inclusion. According to scientific data and definitions provided,1 70% of intubated critically ill patients can easily be extubated. Of the remaining 30%, 25% experience “Difficult Weaning” (DW-defined as failure of the first SBT, but success within one week of the first attempt), whereas 5% experience “Prolonged Weaning” (PW-defined as repeated failed attempts within more than a week from the first trial). In patients with weaning failure, mortality raises up to 25%.11 We therefore investigated patients which have had either more than one episode of failed SBT but successful extubation within a week (DW) or repeated failed episodes of SBT in more than a week time period (PW).1, 5 Patients were included in the study if they fulfilled all of the following criteria: age≥18 years; Glascow Coma Scale ≥14; hemodynamic stability; use of oxygen fraction ≤0.5; pressure support <12 cmH2O and positive end expiratory pressure (PEEP) <6 cmH2O; respiratory rate (RR) <35/min, tidal volume Vt >5 mL/kg Ideal Body Weight (IBW). The use of sedatives and/or analgesics was not contraindicated as long as the patient remained calm, cooperative and easily arous- Minerva Anestesiologica November 2016 DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANINGFLEVARI Figure 1.—Flow chart of patients for the study population. able (Ramsey score 2-3). Patients were excluded from the study if they had at least one of the following: lack of cooperation, incapability to initiate an inspiratory effort, intubation due to elective surgery or upper airway obstruction, ventilation via tracheostomy tube, hemodynamic instability. Weaning protocol In our clinic, a weaning protocol is being implemented: Patients if sedated receive assistcontrol ventilation. After gradual withdrawal of sedatives/ and or analgesics and if patients are clinically stable, (RR <25 bpm, PaO2 >80 mmHg on FiO2 <60%, PEEP <7 cmH2O, minute ventilation VE<10 L/min), we implement pressure-support ventilation (PSV). PSV is initially set at 15-20 cmH2O (above PEEP) and attempts to reduce this level of support by 2 to 4 cmH2O are made at least twice daily according to patient’s adaptation and recovery. When pressure-support and PEEP level are less than 10 and 6 cmH2O respectively and provided Vol. 82 - No. 11 that the patient is alert, calm, co-operative and has adequate cough, we withdraw the ventilator and allow him to breathe spontaneously through a T-tube circuit for up to two hours. In rare cases, patients may require small doses of analgosedation in order to be calm and cooperative (Ramsey score 2-3, absence of delirium), preferably propofol or remifentanyl. Per study protocol, we used remifentanil (at a dosage of ≤0.02 μgr/kg/min) which thanks to its favorable pharmacokinetics, allows rapid elimination from blood plasma. Remifentanil infusion was interrupted 10 minutes before any test performance, so as not to interfere with our measurements. Criteria for SBT discontinuation were any of the following: change in mental status, discomfort or diaphoresis, RR>35/ min, hemodynamic instability (systolic blood pressure >180 mmHg, or <90 mmHg, heart rate >130 or increase or 20% increase). The decision to discontinue SBT or to extubate was made by ICU-doctors, according to their clinical and parametrical judgement and so was the decision to implement NIV or re-intubate. All Minerva Anestesiologica 1151 FLEVARI DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANING SBTs were classified into a success group (SG) or a failure group (FG). Weaning failure was the trial after which the patient required mechanical ventilation (invasive or non-invasive) within forty-eight hours. Measurement of standard weaning parameters, namely RSBI and Pimax Pimax has long been used in ICU setting to assess inspiratory muscle strength. The maneuver consists of a maximum inspiratory effort via a unidirectional valve, which permits exhalation while inhalation is temporarily blocked, allowing patients after 3-4 breathing efforts to perform maximal effort at a lung volume approaching residual. Its measurement is noninvasive and simple to perform, but requires considerable degree of cooperation.12 Pimax was measured while the patient was on SBT on a T-piece trial. On the other hand, RSBI was measured with the patient on a continuous positive airway pressure of 5 cmH2O, an alternative to SBT method.13 This was provided by setting the ventilator at PSV mode and adjusting pressure support level at 0 cmH2O and PEEP at 5 cmH2O, enabling us to directly view tidal volume and respiratory frequency. Ultrasonography Diaphragmatic evaluation was performed with a multifrequency 5-MHz vector transducer placed immediately below costal margins. We used right midclavicular or anterior axillary line to visualize right DEx and left middle-posterior axillary line to visualize left DEx with liver and spleen serving as acoustic windows respectively. We started our exploration by B-mode sonography, a two dimensional image, essential to select the exploration line. Movement was recorded by M-mode sonography and diaphragmatic inspiratory amplitude (excursion) was measured along the selected line. The first endpoint was placed at the foot of the inspiration slope on the diaphragmatic echoic line and the second endpoint at the apex of the slope. A detailed description of the sonographic technique is provided by Matamis 1152 et al.6 In our study all patients were lying in 0-10° recumbent position and all measurements were performed during quiet breathing by two operators (among the three first writers). When patients were accustomed to our technique, the DEx of five consecutive normal respiratory cycles were recorded and the mean value was calculated. The mean value of the recordings made by the two operators was finally recorded. Apart from DEx, ultrasonography enabled us to visualize other pathologic entities, such as atelectasis, pleural effusion or diaphragmatic palsy, that would probably interpret weaning difficulty. Physicians responsible for weaning decisions were blinded to sonographic measurements. Statistical analysis All groups of data were tested for normality by Kolomogorov-Smirnov Test. If data were normally distributed, Student’s t-test tested the difference between the means; if not, non-parametric Mann-Whitney U Test was performed. Therefore descriptive statistics are presented as mean (±standard deviation) and median values (25-75th interquartile ranges) respectively. Sensitivity, specificity, positive and negative predictive value (PPV-NPV), positive and negative likelihood ratio (PLR-NLR) were calculated for all predictive variables, namely RDEx, LDEx, RSBI and Pimax. Sensitivity and specificity are dependent on the cut-off value used, above or below which a test is positive. For RDEx, we decided to test two cut-off points at 10 mm and 15 mm, whereas for LDEx we tested only 10 mm excursion during quiet breathing. This threshold is based on previous studies performed in normal subjects, which found that the lower normal limit of DE for both hemidiaphragms was 9 mm in women and 10mm in men.7 This cut-off limit has been adopted by other researchers as well.8 For RSBI and Pimax we adopted standard cutoff values, namely <100 breaths/min/L and ≤-30 cmH2O respectively.2, 14 Receiver Operative Characteristic (ROC) curves were plotted in order to examine the diagnostic accuracy of each variable and choose the optimal cut-off value for our study, based on our measurements. Minerva Anestesiologica November 2016 DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANINGFLEVARI Table I.—Characteristics and data of study population. No of patients % of patients No of patients Male/Female Etiology of ICU submission Cardiovasuclar surgery Non-surgical cardiac disease Trauma Non-traumatic post-surgical disease Sepsis - ARDS - MOF Miscellaneous 27 13/14 100 48/52 8 3 7 2 5 2 30 11 26 7 19 7 Data Median value 95% CI 65 7 54-73 5-10 3 3-4 Characteristics Age (years) Duration of MV before study inclusion (days) No of failed trials before study inclusion ICU: Intensive Care Unit; CABG: coronary artery bypass graft; COPD: chronic obstructive pulmonary disease; MOF: multiple organ failure; MV: mechanical ventilation. For all analyses, statistical significance was set at 5% (a=0.05) and testing was 2-tail. Statistical analysis was carried out by MedCalc Statistical Software version 16.4.3 (MedCalc Software bvba, Ostend, Belgium; 2016). Results In Table I, we present study population and primary reason for ICU admission. Among patients, eleven were admitted after elective or emergent surgical procedure and sixteen were admitted due to medical conditions. We also provide the duration of mechanical ventilation and the number of failed trials before study inclusion. Among the 27 patients enrolled, only two required remifentanil infusion, which as per study protocol was discontinued ten minutes before study performance. Trials were for both patients successful. Table II tabulates anthropometric characteristics, APACHE II score and results referring to the study population (A), as well as to male and female patients (B1-B2). Apart from results appearing in Tables, we state that we found no difference between RDEx and LDEx (P=0.6) in the overall study group, as well as between genders. In SG we found no difference between right and left DEx (P=0.86) and the same was found for FG as well (P=0.11). Findings from SG and FG are presented in Table II and Figure 2. As we can see, all four weaning predictors had statistically significant difference. Table IV tabulates sensitivity, specificity, PLR, NLR, PPV and NPV of all weaning predictors, standard (f/Vt and Pimax) and sonographic. We present data for two different RDEx cut-offs, namely 10 mm and 15 mm. The cut-off value of DEx for predicting successful extubation was found 10mm for the right hemidiaphragm and 7 mm for the left by receiver operating characteristic (ROC) curve analysis (Table V). For the other two traditional predictors, f/Vt and Pimax, the cut-off values were found at 57 br/min/L and -20 cmH2O respectively. At these cut-off limits, all four tests seem to have Table II.— Anthropometric characteristics (Age/BMI), Illness Severity Score (APACHE 2) and predictors of weaning success in A. Overall study group, B. Male and Female subpopulations (in median and 25-75th percentile values). Variables Age (years) BMI (kg/m2) APACHE 2 Days ventilated on inclusion Weaning predictors f/Vt (RSBI) (breaths/min/L) Pimax (cmH2O) RDEx (mm) LDEx (mm) A. All patients (100%) B1. Male patients (48%) B2. Female patients (52%) P value Median 25th- 75th Median 25th- 75th Median 25-75th 65 22 20 7 53-75 20-28 15-25 5-10 54 22 18 7 46-67 20-24 15-20 5-11 69 25 23 6 61-75 20-30 19-25 4-10 0.05* 0.13 0.08 0.36 70 25 14 12 53-100 20-34 8.5-22 7-23 70 25 15 18 46-80 19-31 12-30 10-25 74 28 13 10 57-119 21-35 7-20 5-19 0.19 0.9 0.11 0.24 BMI: Body Mass Index; APACHE II Score: Acute Physiology and Chronic Health Evaluation; f/Vt (RSBI): ratio of respiratory frequency to tidal volume, representing Rapid Shallow Breathing Index; Pimax: Maximal Inspiratory Pressure; RDEx: Right Diaphragmatic Excursion; LDEx: Left Diaphragmatic Excursion. Numbers with an asterisk (*) indicate difference of a statistical significance. Vol. 82 - No. 11 Minerva Anestesiologica 1153 FLEVARI DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANING A B Figure 2.—Right (A) and Left (B) Diaphragmatic Excursions (RDEx, LDEx) in Success and Failure groups. Table III.—Comparison of the characteristics (Age / BMI), APACHE II Score and predictors of weaning between Success and Failure groups (in median and 25-75th percentile values). SG Variables Median Age (years) BMI (kg/m2) APACHE II Weaning predictors f/Vt (RSBI) (breaths/min/L) Pimax (cmH2O) RDEx (mm) LDEx (mm) FG 25th- 75th Median 25-75th P value Median 60 22 21 48-74 20-28 16-25 70 28 20 67-77 20-32 13-25 0.06 0.45 0.64 64 30 19 19 45-85 25-35 13-27 11-25 116 15 7 5 68-120 11-24 5-10 2-6 0.03* 0.01* 0.004* 0.0004* SG: success group; FG: failure group; BMI: Body Mass Index; APACHE II Score: acute physiology and chronic health evaluation; f/Vt (RSBI): ratio of respiratory frequency to tidal volume, representing Rapid Shallow Breathing Index; Pimax: maximal inspiratory pressure; RDEx: right diaphragmatic excursion; LDEx: left diaphragmatic excursion. Numbers with an asterisk (*) indicate difference of a statistical significance. Table IV.—Sensitivity, specificity, AUC, positive/negative likelihood ratio, positive/negative predictive value of standard predictors of weaning failure, and of right and left diaphragmatic excursion during quiet breathing. RDEx was tested against two critical values. Prognostic variables RDEx <10 mm RDEx <15 mm LDEx <10 mm F/Vt >100 breaths/min/L Pimax <│-30 cmH2O│ Sensitivity (%) Specificity (%) AUC Positive Likelihood Ratio Negative Likelihood Ratio Positive Predictive Value (%) 71 100 86 71 85 85 65 85 85 65 0.78 0.83 0.85 0.78 0.75 4.8 2.85 5.7 4.8 2.4 0.3 0 0.16 0.33 0.2 63 50 67 63 46 Negative Predictive Value (%) 89 100 94 89 93 RDEx: right diaphragmatic excursion; LDEx: left diaphragmatic excursion; f/Vt: ratio of respiratory frequency to tidal volume; Pimax: maximal inspiratory pressure; AUC: area under the curve. comparable diagnostic accuracy as this is expressed by the area under the curve (AUC) of the ROC curve analysis. Pairwise Comparison of ROC curves showed no statistical difference between AUC. 1154 Discussion To our knowledge, this is the first study to investigate sonographic diaphragmatic excursion as a weaning tool in patients with Minerva Anestesiologica November 2016 DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANINGFLEVARI Table V.—Receiver operating characteristic (ROC) curves for all weaning predictor variables and optimal cut-off points for excluding weaning failure. Prognostic variables RDEx (mm) LDEx (mm) F/Vt (breaths/min/L) PImax (cmH2O) Optimal cut-off point AUC ≤10 <7 ≥57 <│-20│ 0.87 0.95 0.77 0.81 95% CI Lower Upper 0.69 0.79 0.57 0.61 0.97 0.99 0.91 0.93 P value 0.001* 0.0001* 0.006* 0.01* Sensitivity (%) Specificity (%) 86 86 100 71 85 95 60 85 AUC: area under the curve; CI: Confidence Interval; RDEx: right diaphragmatic excursion; LDEx: Left diaphragmatic excursion; F/Vt: ratio of respiratory frequency to tidal volume; PImax: maximal inspiratory pressure. Numbers with an asterisk (*) indicate difference of a statistical significance. prolonged and difficult weaning, in whom diaphragmatic contractile dysfunction is well described.15 We found that the excursion of both hemidiaphragms (RDEx-LDEx) performs well in terms of diagnostic accuracy. We also found that a cut-off limit of 10mm for the right hemidiaphragm and of 7 mm for the left can predict weaning success. The much lower left hemidiaphragmatic threshold (7 mm) was rather surprising. This could be probably attributed either to differences in probe positioning, or to the study population per se. As far as probe positioning is concerned, we handled the probe on the middle-posterior left axillary line, so as the angle of ultrasound beam to reach perpendicularly left diaphragm, whereas we placed the probe on the midclavicular or the anterior right axillary line in order to visualize right diaphragm. That is to say, we probably faced LDEx closer than RDEx. As far the study population is concerned, in patients with difficult weaning, who may have diminished DEx, this could be counterbalanced by an extra effort exerted by accessory inspiratory muscles, finally making weaning feasible. We found no gender difference in diaphragmatic excursion unlike the findings by previous research.7 Ultrasonographic indices as predictors of successful extubation are rather new in the scientific literature.5, 6, 8-10, 16 Our findings suggest that if we want to find all failed trials, RDEx <15 mm and LDEx<10 mm should be included (high sensitivity). On the other hand, if we want to correctly diagnose success (high NPV), we should rely on RDEx>15 mm, LDEx>10 mm and MIP above │-30 cmH2O│. Moreover based on optimal cut-off points, if LDEx<7 mm, RDEx≤ 10 Vol. 82 - No. 11 mm, and MIP<│-20 cmH2O│ weaning is highly probable to fail (high specificity). Among the researchers investigating sonographic indices as weaning predictor rules,8, 9, 16, 17 DiNino et al. 10 as well as Ferrari et al. 16 measured the alteration of diaphragmatic thickness on deep breathing, whereas Kim et al.,8 as well as Jiang et al.9 measured diaphragmatic excursion. All of them concluded that sonographic indices of the diaphragm could be used as weaning predictors, since they perform well, if not better, in comparison to standard predictors. Both studies investigating DEx included all patients eligible for a weaning trial and not only those with prolonged or/and difficult weaning. Jiang et al. investigated patients, already scheduled to be extubated, along the right anterior axillary and left posterior axillary line, to find the same cut-off threshold (11 mm) for both hemidiaphragms. As per study protocol, Jiang et al. measured real time liver and spleen displacements and not DEx, and as they stated, this may integrate the contraction of all inspiratory muscles both diaphragm and accessory inspiratory muscle. This study has a different concept of measuring, which along with the difference in probe positioning could explain the discrepancy between their LDEx threshold and ours. Kim et al. also included all ICU patients along the right anterior axillary and the left midaxillary line. Patients were classified according to a DEx cut-off of 10mm in two groups, with and without diaphragmatic dysfunction (DD), to find that 29% of the patients had indeed DD with an excursion of a median value of 3.0 mm for the right diaphragm and 2.6 mm for the left. Their cutoff limits for predicting primary (within 48 h) Minerva Anestesiologica 1155 FLEVARI DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANING weaning failure are higher than ours (14mm for RDEx and 12mm for LDEx), probably reflecting differences in methodology in terms of study population (all patients, instead of patients with difficult/prolonged weaning) and probe positioning. We therefore believe that patients with difficult and/or prolonged weaning, whose diaphragm has some degree of weakness due to prolonged mechanical ventilation,18 recruit all inspiratory muscles, making final extubation feasible. The concept that in this particular sub-population of patients, a lower diaphragmatic threshold would be acceptable, warrants further validation in future studies. As far as standard weaning predictors are concerned, we examined the performance of RSBI and Pimax. RSBI (with a threshold value of 105 breaths/min/L) was first introduced in 1991 by Yang and Tobin 2 as the best among many weaning predictors. Since then it has been re-examined in many studies and its value, though doubted,5, 14 remains high.19-21 These studies demonstrate a wide range of threshold values (60-105) and positive likelihood ratios (0.84-4.67),14 indicating that the index has great heterogeneity in posttest probability, probably due to differences in methodology and variations in pretest probability (Bayes’ theorem).4, 14, 21 In our study, RSBI was performed at CPAP 5 cmH2O, in order to be able to directly visualize tidal volume and respiratory frequency. Adding this minimal support is probably needed to compensate for the resistance and the dead space of the circuitry of the ventilator.22-24 Besides this method is considered as an alternative of SBT by many studies.13, 25 Nevertheless we are aware of the fact that differences in methodology would alter the cut-off threshold of the index, since even minimal support would affect work of breathing.25, 26 In our study, the RSBI cut-off value was 57 breaths/min/L, which lies at the lowest point of the aforementioned range. The fact that RSBI was recorded while the patient was on CPAP 5 cmH2O could probably explain the lower cut-off value, as has previously been stated.27 In terms of diagnostic accuracy (AUC, sensitivity and specificity (0.77, 100% and 1156 60% respectively), its low specificity diminishes its value. Nevertheless our results were comparable to those of other researches.2, 9 Finally, the standard index that determines inspiratory muscle strength is Pimax.12 Pimax is simple to perform but difficult to interpret. First of all, the lower cut-off limit (not uniformly accepted) is largely dependent on patient’s voluntary effort, which is very hard to obtain in critically ill patients. Moreover Pimax reflects the integrated pressure exerted by all inspiratory muscles and not by the diaphragm itself. We found the threshold of │-20 cmH2O│, as a good predictor of extubation failure, which lies in the range of │20-30 cmH2O│ described in literature.1 Limitations of the study There are some limitations for this study. First, interobserver and intraobserver variabilty were not tested. We accepted the fact that past studies have given good results in this field.7, 28 Second, we did not measure diaphragmatic thickness, since this requires both patient’s cooperation and an ultrasound probe of a higher frequency (>7.5 MHz). Finally the cut-off value of LDEx warrants validation in a new and larger cohort of patients. Conclusions Our results suggest that diaphragmatic excursion, as assessed by M-mode ultrasonography, can reliably estimate weaning failure in patients with difficult and prolonged weaning. In fact the sonographic thresholds of ≤10 mm for the right hemidiaphragm and <7 mm for the left could be used as adjunct tools in predicting failed extubation in the subpopulation of critically ill patients with difficult and prolonged weaning. Key messages —— Difficult and prolonged weaning is associated with increased mortality. —— Standard weaning parameters do not Minerva Anestesiologica November 2016 DIAPHRAGMATIC ULTRASONOGRAPHY IN PATIENTS WITH WEANINGFLEVARI suffice to predict weaning failure in patients with difficult and prolonged weaning. —— We suggest that if LDEx<7 mm and RDEx≤10 mm weaning trial is highly probable to fail. References 1. Boles JM, Bion J, Connors A, Herridge M, Marsh B, Melot C, et al. Weaning from mechanical ventilation. Eur Resp J 2007;29:1033-56. 2. Yang K, Tobin M. A prospective study of indexes predicting the outcome of trials of weaning from mechanical ventilation. N Engl J Med 1991;324:1445-50. 3. Nemer SN, Barbas CS, Caldeira JB, Cárias TC, Santos RG, Almeida LC, et al. A new integrative weaning index of discontinuation from mechanical ventilation. Critical Care 2009;13:R152. 4.Conti G, Montini L, Pennisi MA, Cavaliere F, Arcangeli A, Bocci MG, et al. A prospective, blinded evaluation of indexes proposed to predict weaning from mechanical ventilation. Intens Care Med 2004;30:830-6. 5.Sellarés J, Ferrer M, Torres A. Predictors of weaning after acute respiratory failure. Minerva Anestesiol 2012;78:1046-53. 6. Matamis D, Soilemezi E, Tsagourias M, Akoumianaki E, Dimassi S, Boroli F, et al. Sonographic evaluation of the diaphragm in critically ill patients. Technique and clinical applications. Intens Care Med 2013;39:801-10. 7. Boussuges A, Gole Y, Blanc P. Diaphragmatic motion studied by m-mode ultrasonography: methods, reproducibility, and normal values. Chest 2009;135:391-400. 8. Kim WY, Suh HJ, Hong SB, Koh Y, Lim CM. Diaphragm dysfunction assessed by ultrasonography: influence on weaning from mechanical ventilation. Critical Care Med 2011;39:2627-30. 9. Jiang JR, Tsai TH, Jerng JS, Yu CJ, Wu HD, Yang PC. Ultrasonographic evaluation of liver/spleen movements and extubation outcome. Chest 2004;126:179-85. 10. DiNino E, Gartman EJ, Sethi JM, McCool D. Diaphragm ultrasound as a predictor of succesful extubation from mechanical ventilation. Thorax 2014;69:423-7. 11.Esteban A, Alía I, Tobin MJ, Gil A, Gordo F, Vallverdú I, et al. Effect of Spontaneous Breathing Trial Duration on Outcome of Attempts to Discontinue Mechanical Ventilation. Am J Respir Crit Care Med 1999;159:512-8. 12. ATS/ERS Statement on respiratory muscle testing. Am J Respir Crit Care Med 2002;166:518-624. 13.Ely WE, Baker AM, Dunagan DP, Burke HL, Smith AC, Kelly PT, et al. Effect of the duration of mechanical ventilation of identifying patients capable of breathing spontaneously. N Engl J Med 1996;335:1864-9. 14. MacIntyre NR, Cook DJ, Ely EW, Epstein SK, Fink JB, Heffner JE, et al. Evidence-Based Guidelines for Weaning and Discontinuing Ventilatory Support. A Collective Task Force Facilitated by the American College of Chest Physicians; the American Association for Respiratory Care ; and the American College. Chest 2001;120:37595. 15. Powers SK, Kavazis AN, Levine S. Prolonged mechanical ventilation alters diaphragmatic structure and function. Crit Care Med 2009;37(10 Suppl):S347-353. 16. Ferrari G, Filippi G, De Elia F, Panero F, Volpicelli G, Aprà F. Diaphragm ultrasound as a new index of discontinuation from mechanical ventilation. Crit Ultrasound J 2014;6:8. 17.Umbrello, M, Formenti, P, Longhi, D, Galimberti, A, Piva, I, Pezzi, A, et al. Diaphragm ultrasound as indicator of respiratory effort in critically ill patients undergoing assisted mechanical ventilation : a pilot clinical study. Crit Care 2015;19:161. 18.Sassoon, C.S.H. Ventilator-associated Diaphragmatic Dysfunction. Am J Respir Crit Care Med 2002;166:10178. 19. de Souza LC, Lugon JR. The rapid shallow breathing index as a predictor of successful mechanical ventilation weaning : clinical utility when calculated from ventilator data. J Bras Pneumol 2015 41:530-5. 20. Epstein SK. Etiology of Extubation Failure and the Predictive Value of the Rapid Shallow Breathing Index. Am J Respir Crit Care Med 1995;152:545-9. 21.Tobin MJ, Jubran A. Variable performance of weaningpredictor tests : role of Bayes’ theorem and spectrum and test-referral bias. Intens Care Med 2006;32:2002-12. 22.Straus C, Louis B, Isabey D, Lemaire F, Harf A, Brochard L. Contribution of the Endotracheal Tube and the Upper Airway to Breathing Workload. Am J Respir Crit Care Med 1998;157:23-30. 23. Nathan SD, Ishaaya AM, Koerner SK, Belman MJ. Prediction of minimal Pressure Support during weaning from mechanical ventilation. Chest 1993;103:1215-9. 24. Pelosi P, Solca M, Ravagnan I, Tubiolo D, Ferrario L, Gattinoni L. Effects of heat and moisture exchangers on minute ventilation, ventilatory drive, and work of breathing during pressure-support ventilation in acute respiratory failure. Crit Care Med 1996;24:1184-8. 25. Desai NR, Myers L, Simeone F. Comparison of 3 different methods used to measure the rapid shallow breathing index. J Crit Care 2012;27:418.e1-418.e6. 26.Tobin MJ. Extubation and the myth of “minimal ventilator settings”. Am J Respir Crit Care Med 2012;185:34950. 27. El-Khatib MF, Zeineldine SM, Jamaleddine GW. Effect of pressure support ventilation and positive end expiratory pressure on the rapid shallow breathing index in intensive care unit patients. Intens Care Med 2008;34:50510. 28.Houston JG, Angus RM, Cowan MD, McMillan NC, Thomson NC. Ultrasound assessment of normal hemidiaphragmatic movement: relation to inspiratory volume. Thorax 1994;49:500-3. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Acknowledgements.—We would like to express our sincere thanks to all patients who patiently suffered our services. We would also like to gratefully acknowledge the critical reviews of anonymous reviewers made on an earlier, as well as on the present version of the manuscript. Article first published online: July 12, 2016. - Manuscript accepted: July 7, 2016. - Manuscript revised: June 16, 2016. - Manuscript received: March 5, 2016. Vol. 82 - No. 11 Minerva Anestesiologica 1157 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1158-69 ORIGINAL ARTICLE Non-invasive hemodynamic optimization in major abdominal surgery: a feasibility study Ole BROCH 1 *, Arne CARSTENS 2, Matthias GRUENEWALD 1, Edith NISCHELSKY 3, Lukas VELLMER 3, Berthold BEIN 4, Heiko ASELMANN 5, Markus STEINFATH 1, Jochen RENNER 1 1Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany; 2Department of Anesthesiology and Intensive Care Medicine, Imland Hospital, Rendsburg, Germany; 3Christian-Albrechts-University Kiel, Schleswig-Holstein, Germany; 4Department of Anesthesiology and Intensive Care Medicine, Asklepios Hospital St. Georg, Hamburg, Germany; 5Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Germany *Corresponding author: Ole Broch, University Hospital Schleswig-Holstein, Campus Kiel, Department of Anesthesiology and Intensive Care Medicine, Schwanenweg 21, D-24105 Kiel, Germany. E-mail: [email protected] ABSTRACT BACKGROUND: Today, most of the pre-emptive hemodynamic optimization algorithms are based on variables associated with invasive techniques like arterial cannulation. The non-invasive Nexfin™ technology is able to estimate continuous Cardiac Index (CI) and pulse pressure variation (PPV). However, the efficiency of an early goal directed therapy (EGDT) algorithm based on non-invasive variables has to be proven. The aim of our study was to investigate the feasibility of a non-invasive driven EGDT protocol and its impact on patient’s outcome. METHODS: Seventy-nine patients (ASA II-III) undergoing elective major abdominal surgery were randomized to either study group (SG, N.=39) or control group (CG, N.=40). The SG was treated according to an algorithm based on noninvasive CI and PPV, whereas the CG received standard of care. Postoperative complications up to 28 days and length of hospital stay (LOS) in both groups were recorded. RESULTS: There was no significant difference between the groups regarding demographics, hemodynamic variables, preoperative risk scores and duration of surgery. The total amount of complications was higher in the CG (SG 94 vs. CG 132 complications, P=0.22) without reaching statistical significance. LOS revealed no difference between both groups (SG, 9 [7-15] vs. CG, 9 [7-15.25] days, P=0.82). We have seen no impact of the non-invasive optimization protocol with respect to postoperative mortality. CONCLUSIONS: In this patient collective, we could demonstrate the feasibility of a non-invasive approach for hemodynamic optimization. However, EGDT based on non-invasive variables was not able to significantly improve outcome. (Cite this article as: Broch O, Carstens A, Gruenewald M, Nischelsky E, Vellmer L, Bein B, et al. Non-invasive hemodynamic optimization in major abdominal surgery: a feasibility study. Minerva Anestesiol 2016;82:1158-69) Key words: Anesthesia - Surgical procedures - Feasibility studies. M ajor surgical procedures include the threat of hypoperfusion and possible mismatch in oxygen delivery and demand. Several studies have demonstrated that a preemptive hemodynamic optimization by early goal directed fluid and catecholamine therapy (EDGT) was associated with beneficial impact on both morbidity and mortality.1 However, 1158 these beneficial effects are more pronounced in critically ill patients.2 It must be noted, that “basic” perioperative monitoring, i.e. electrocardiogram, oxygen saturation and non-invasive or invasive blood pressure measurements, is not able to sufficiently detect hypovolemia and ongoing organ hypoperfusion.3 Flow related variables like cardiac index (CI), Stroke Minerva AnestesiologicaNovember 2016 NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY Volume Index (SVI) and their surrogates, stroke volume variation (SVV) and pulse pressure variation (PPV) are claimed to be advantageous for guiding fluid and catecholamine therapy.4 Therefore, less invasive monitoring systems based on transpulmonary thermodilution or uncalibrated pulse contour analysis have gained increasing interest by gradually minimizing the reluctance of physicians to use advanced hemodynamic monitoring.5 Over the last years several studies focused on EGDT protocols, yielding a strong proof of feasibility of different algorithms and, moreover providing evidence that EDGT improves patient’s outcome in various clinical scenarios.1 The Nexfin™ monitoring system (previous manufacturer: BMEYE, Amsterdam, The Netherlands, current manufacturer: Clearsight™, Edwards Lifesciences LLC, Irvine, CA, USA) offers a completely non-invasive approach providing continuous beat-to-beat CI, SVI, SVV, PPV and arterial pressure by using an inflatable finger cuff. Several clinical investigations have demonstrated sufficient trending ability of the Nexfin™ technology.6-8 At present there is no evidence, whether an EGDT protocol based on a non-invasive monitoring technology in general is feasible in a wide range of patients on the one hand, and if it is able to reduce postoperative complications on the other hand. Therefore, the aim of our study was in first line to verify the feasibility of a non-invasive EDGT protocol in patients undergoing major open abdominal surgery and in second line to compare their postoperative outcome with a standard of care approach. Materials and methods The study was conducted as a single center, prospective randomized trial from March 2014 to October 2015 at the University Hospital Schleswig-Holstein, Campus Kiel, in accordance with the Helsinki declaration. After approval from institutional ethics committee (Ethikkomission UKSH Kiel, AZ B260/11, Christian Albrecht University Kiel, Schwanenweg 20, D 24105 Kiel), written informed consent for participation in the study Vol. 82 - No. 11 BROCH was obtained preoperatively from all patients. The trial was registered on ClinicalTrials.gov (NCT02559141). Eighty-six ASA II and III patients undergoing elective major abdominal surgery were included in the study. Seven patients have to be excluded due to various reasons. Patients undergoing major abdominal procedures with an estimated duration ≥120 minutes and a high transfusion probability due to an anticipated blood loss ≥1000 mL were screened for elegibility. Flow chart with enrolment, intervention allocation, follow-up and data analysis is represented in Figure 1. The Physiological and Operative Severity Score for the enumeration of Mortality and Morbidity (POSSUM) was used for risk evaluation. Exclusion criteria were patients less than 18 years old, ASA I or IV classification, heart rhythm disorders, advanced peripheral artery occlusive disease, arteriovenous shunts concerning upper extremities and laparoscopic abdominal procedures. Patients received midazolam 0.1 mg/kg orally 30 minutes before induction of anesthesia. A five lead electrocardiogram (ECG; S/5, GE Healthcare, Helsinki, Finland) and peripheral oxygen saturation (SpO2) were established as routine monitoring. As suggested by recent literature, non-invasive blood pressure was obtained on both upper arms to exclude a difference ≥10 mmHg.9 In case of a non-invasive blood pressure difference ≥10 mmHg, the Nexfin™ monitoring system was positioned on the ipsilateral side to the arterial line. Prior to induction of general anesthesia patients received a peripheral venous access and a radial arterial line (Arrow International, Inc. Reading, PA, USA) under local anesthesia. After installation of the pneumatic finger cuff, the Nexfin™ monitoring system was feeded with patient specific data as advised by the manufacturer. The underlying physiological background and functionality of this monitoring system is described elsewhere.10-13 Adjustment of the transducer was followed by zeroing at the height of the midaxillary position. The non-invasive device was established at the side contralateral to the indwelling radial catheter, followed by attachment of a temperature Minerva Anestesiologica 1159 BROCH NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY (N.=115) (N.=9) (N.=13) (N.=7) (N.=86) (N.=3) (N.=3) (N.=1) (N.=79) (N.=40) (N.=39) (N.=40) (N.=39) (N.=1) (N.=40) (N.=38) Analyzed (N.=38) Analyzed (N.=40) (N.=0) (N.=0) Figure 1.—Enrolment, allocation, follow-up and analysis of participants through the trail. probe (Siemens AG, S1138, Erlangen, Germany) for measuring skin temperature at the fingertip. Temperature was also measured with a nasopharyngeal probe. All variables were automatically indexed to body surface area. After induction of general anesthesia with 0.5 µg/ kg sufentanil and 1.5 mg/kg propofol, orotracheal intubation was facilitated with 0.6 mg/ 1160 kg rocuronium. Anesthesia was maintained in accordance with our standard operating procedure using sevoflurane and sufentanil. Thereafter, a central venous catheter was introduced in the right internal jugular vein. Patients were ventilated in a pressure controlled modus with an oxygen/air mixture using a tidal volume of 8 mL/kg ideal body weight and a positive end- Minerva AnestesiologicaNovember 2016 NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY expiratory pressure of 5-10 cmH2O. Respiratory rate was adjusted to achieve normocapnia (pCO2 35-40 mmHg). Ventilation parameters were documented every 60 minutes after intubation. During surgery, the patients received a continuous infusion of crystalloid solution (6 mL/kg/ h). Data collection Patients were randomized either into study group (SG, N.=39) or control group (CG, N.=40). Each patient received a central venous catheter and an arterial line. The Nexfin™ monitoring system was also established in each group but was strictly covered and inaccessible for the attending anesthetist in the CG. Another investigator who was not involved in management of anesthesia and hemodynamic therapy collected non-invasive hemodynamic variables estimated by the Nexfin™ monitoring system in the same manner in the CG as well in the SG. In accordance to the study algorithm, hemodynamic variables were recorded at baseline, after induction of anesthesia and subsequently every 15 minutes till the end of surgery. Central venous and arterial blood gas samples were drawn hourly after baseline. Patients in the CG were treated with a basic algorithm, targeting an invasive estimated mean arterial pressure (MAP) of ≥65 mmHg, a CVP ≤12 mmHg and a consistent hemoglobin level ≥8 g/dL. To preserve a MAP ≥65 mmHg, vasoactive agents like bolus injection of theodrenaline/cafedrine or continuous infusion of norepinephrine (0.01-0.08 µg/kg/min) were administered. Due to the absence of advanced hemodynamic monitoring and no possibility to control the therapeutic effect, dobutamine was not used in the CG. The SG was treated on the basis of an EDGT algorithm which among others, intended to maintain preload (Figure 2). In presence of a PPV >10%, volume substitution was initiated using 500 mL of crystalloids and/or colloids as long as CI increased ≥2.5 L/min/m2. Maintenance of CI ≥2.5 L/min/m2 and MAP ≥65 mmHg was achieved by using norepinephrine (0.01-0.08 µg/kg/min), theodrenaline/cafe- Vol. 82 - No. 11 BROCH drine and dobutamine (5-10 µg/kg/min). Catecholamine administration, blood loss, urine output, fluid input was recorded every 15 minutes up to the end of surgery in the SG as well as in the CG. Time between end of surgery and extubation was also recorded. Hemodynamic monitoring was continued in all patients in the post-anesthesia care unit (PACU) or intensive care unit (ICU) for at least 180 minutes. Again, data collection was performed every 15 minutes in all patients but there was no defined algorithm postoperatively. Central venous and arterial blood gas samples were drawn hourly after arrival in the PACU or ICU. Data on catecholamine use, estimated blood loss, urine output and fluid intake were obtained 24 hours postoperatively. Postoperative complications were divided into different categories: general (bleeding, reoperation), infection (wound, pneumonia, urinary tract, abdominal), cardiovascular (pulmonary edema, myocardial infarct, stroke, new onset atrial fibrillation), respiratory (respiratory support >24 h, pleural effusion) and abdominal (ascites, prolonged paralytic ileus, need of parenteral feeding, ongoing nausea and vomiting) complications. Length of hospital stay (LOS) and complications were followed up 28 days after surgery and documented by daily visits and aligning data from the patient charts. Documentation of LOS was carried out by the electronic chart. Statistical analysis Sample size calculation was based on our own hospital registry, which revealed an incidence of postoperative complications of 45% for a similar historical patient collective (unpublished data). For a decrease in morbidity from 45 to 30%, we calculated a sample size of 35 patients for each group with a 0.05 difference (two-sided) and a power of 80%. Considering a drop out of patients of 10-20% due to different reasons, we decided to include 43 patients in each group. Statistical analysis was performed by using commercially available statistics software (GraphPad Prism 5, GraphPad Software Inc., San Diego, CA, USA; SigmaPlot 13.0 Minerva Anestesiologica 1161 BROCH NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY Figure 2.—Fluid and catecholamine management in the study group based on hemodynamic variables. CI: Cardiac Index; PPV: pulse pressure variation; MAP: mean arterial pressure. for Windows version 7, Systat Software, Inc., San Jose, CA). Parametric data were analyzed with the student`s two-tailed t-test, whereas the Mann-Whitney U-test was used for analysis of non-parametric data. Categorical data were compared using χ2 and Fisher’s Exact Test and P<0.05 was considered statistical significant. Data are listed in mean, standard deviation (SD) or median, interquartile range (IQR) as appropriate. Comparison of data was restricted to the mean operation procedure time at 225 minutes chosen as the average. 1162 Results A total of 86 patients were randomized. Seven patients had to be excluded from the study for various reasons. Finally, data of 79 patients, 58 males and 21 females, were included and allocated; 39 patients in the SG and 40 patients in the CG. One patient in the SG has to be excluded from further analysis due to ongoing heart rhythm disorders. No patient has to be excluded due to persistent deficiencies regarding the non-invasive monitoring tool. Minerva AnestesiologicaNovember 2016 NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY We observed no side effects by the long lasting continuous non-invasive monitoring nor signal loss or any monitoring related complication. There were no significant differences between both groups regarding demographics, initial hemodynamic and laboratory variables and preoperative risk scores. Preoperative demographic, hemodynamic and laboratory data are represented in Table I. MAP did not differ significantly at any time from baseline up to 225 min. CI was comparable between both groups from baseline until 75 min, differences in CI between groups reached significance from 90 min until the end of data collection. At this time, CI was significant higher in the SG with the exception of CI at 105 min achieving no significance (Figure 1). PPV revealed significant differences at various time points from baseline to end of surgery (Figure 2). For lactate concentration analysis showed no significant difference between groups at any time from baseline. At baseline, lactate concen- BROCH tration was 0.88 (0.30) in the SG vs. 0.85 (0.23) mmol/L in the CG (P=0.51). After 60 min, lactate concentration was 0.78 (0.34) in the SG vs. 0.72 (0.18) mmol/L in the CG (P=0.46), at 120 min 0.84 (0.28) in the SG vs. 0.82 (0.26) mmol/L in the CG (P=0.42), at the end of operation 1.21 (1.39) in the SG vs. 0.97 (0.48) mmol/L in the CG (P=0.31) and 180 min after operation 1.11 (0.39) in the SG vs. 0.99 (0.43) mmol/L in the CG (P=0.33). During operation, temperature of the fingertip was 35.5 (0.7) °C in the study group and 35.7 (0.4) °C (P=0.39) in the CG. Values were given as mean (SD). Regarding the amount of administered fluid, packed red blood cells, urine output or blood loss intra- and postoperatively, no significant differences have been revealed. Fresh frozen plasma and noradrenaline have been administered more frequently in the CG. Dobutamine was preserved intraoperatively only for the SG. None of the patients in both groups received inotropes after the end of surgery (Table II). Table I.—Characteristics, demographic data, preoperative haemodynamic and laboratory parameters. Age (yr) Sex m/w Height (cm) ABW (kg) ASA II / III POSSUM (physiological score) POSSUM (operative score) NIBP SP (mmHg) NIBP DP (mmHg) NIBP MAP (mmHg) CI (L/min/m2) HR (min-1) SV (mL) O2 Sat (%) PPV (%) SVV (%) Eadyn WBC (103/µL) Lactate (mmol/L) Hb (g/dL) Epidural analgesia PONV history Study group N.=39 Control group N.=40 P value 67 (9) 28 / 11 174 (9.6) 79.7 (19.3) 28 / 11 18.0 (3.7) 17.4 (4.1) 135 (27) 72 (12) 95 (17) 2.8 (0.7) 71 (11) 79 (23) 95 (2) 10 (6) 8 (5) 1.25 (0.13) 7.78 (0.48) 0.88 (0.30) 12.9 (2.0) 6 1 65 (13) 30 / 10 176 (8.1) 80.8 (14.3) 28 / 12 17.5 (3.1) 18.0 (4.5) 138 (23) 75 (9) 98 (13) 2.9 (0.9) 72 (13) 79 (21) 96 (5) 11 (6) 9 (5) 1.22 (0.17) 7.42 (0.44) 0.85 (0.23) 13.0 (1.9) 6 2 =0.40 =0.49 =0.33 =0.78 n.a. =0.52 =0.53 =0.58 =0.17 =0.40 =0.65 =0.56 =0.95 =0.52 =0.43 =0.66 =0.59 =0.58 =0.56 =0.73 n.a. n.a. Data comparison performed by Student´s t-test, Mann Whitney U-Test, χ2-test or Fisher´s Exact Test for detection of preoperative group differences. Values are given as mean (SD). ABW: actual body weight; ASA: physical status classification by the American Society of Anesthesiologists; POSSUM: physiological and operative severity score for the enumeration of mortality and morbidity; NIBP: non-invasive blood pressure; SP: systolic pressure; DP: diastolic pressure; MAP: mean arterial pressure; CI: cardiac index; HR: heart rate; SV: stroke volume; O2Sat: oxygene saturation; PPV: pulse pressure variation; SVV: stroke volume variation; Eadyn: arterial elastance; WBC: white blood cell count; Hb: hemoglobin; PONV: postoperative nausea and vomiting; n.a.: not assessed. Vol. 82 - No. 11 Minerva Anestesiologica 1163 BROCH NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY Table II.—Intraoperative fluids: vasopressors and inotropes. Crystalloids (mL) Colloids (mL) PRBC (mL) FFP (mL) Blood loss (mL) Urine output (mL) Fluid Balance (mL) Noradrenaline (μg) Theo/Caf (mL) Dobutamine (µg) Study group N.=39 Control group N.=40 P value 3000 (2000-4000) 500 (250-1000) 0 (0-0) (N.=14) 0 (0-0) (N.=0) 800 (300-1500) 300 (200-700) 2300 (1500-3300) 0 (0-186) (N.=15) 0.5 (0-1.5) (N.=25) 11 (0-50) (N.=22) 3000 (2250-3875) 250 (0-1000) 0 (0-0) (N.=8) 0 (0-0) (N.=2) 675 (400-1150) 300 (150-637) 2350 (1500-3400) 60.6 (0-232) (N.=21) 0.8 (0-2) (N.=24) 0 (0-0) (N.=0) =0.99 =0.06 =0.22 =0.59 =0.44 =0.58 =0.95 =0.18 =0.40 =0.57 Data comparison by Student´s t-test (and nonparametric tests) to detect intraoperative group differences. PRBC: packed red blood cells; FFP: fresh frozen plasma; Theo/Caf: theodrenaline/cafedrine; *P<0.05 (vs. control group); Data are median (interquartile range [IQR]). Table III.—Length of stay in hospital and postoperative complications. Hospital stay (days) total ASA II Hospital stay (days) total Hospital stay (days) ASA II no complication Hospital stay (days) ASA II ≥1 complication ASA III Hospital stay (days) total Hospital stay (days) ASA III no complication Hospital stay (days) ASA III ≥1 complication Total amount of complications ASA II Complications ASA III Complications Study group N.=39 Control group N.=40 P value 9 (7-15) 9 (7-15.25) =0.82 8 (7-12.5) 8 (7-11.75) =0.71 7 (6-9.5) 7 (6-8) =0.50 9 (7-21) 10 (8-13) =0.65 13 (7-18) 14.5 (7.5-33.5) 6.5 (3.75-8.5) 16 (13-46) 94 6.5 (6-7) 17.5 (9.75-39.75) 132 =0.64 =0.81 =0.92 =0.22 61 (2.1 per patient) 71 (2.5 per patient) =0.43 33 (3.0 per patient) 61 (5.1 per patient) =0.17 Data analysis by Mann Whitney U-Test and Fisher´s Exact Test for detection of group differences. Data are median (interquartile range [IQR]). Duration of operation was comparable in both groups (SG: 227 (93) vs. CG: 230 (80) min, P=0.92) and time period from end of surgery until extubation differed not significantly (SG: 15 (7) vs. CG: 17 (8), P=0.15). Data were mean (SD). The total amount of complications was higher in the control group (SG: 94 vs. CG: 132, P=0.22). This difference in complication rate was also obtained for ASA II (SG: 61 vs. CG: 71, P=0.43) and ASA III patients 1164 (SG: 33 vs. CG: 61, P=0.17) without reaching statistical significance (Table III). Detailed analysis of complication rate in relation to the ASA classification is represented in Figure 3. In the SG, less patients developed ≥1 complication (SG, N.=29 vs. CG, N.=34, P=0.27). The number of complications for ASA III patients (SG, N.=7 vs. CG, N.=10, P=0.45) differed not significantly. Patients without any complication were higher in the SG (SG, Minerva AnestesiologicaNovember 2016 NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY BROCH Figure 3.—Intraoperative variance of CI. Hemodynamic variables for study group (grey dots) and control group (balck dots). *P<0.05 (vs. control group). Figure 4.—Intraoperative chronological sequence of pulse pressure variation (PPV). Hemodynamic variables for study group (grey dots) and control group (black dots). *P<0.05 (vs. control group). N.=10 versus CG, N.=6, P=0.27). Again, no significant differences for ASA II and ASA III patients were detected (Figure 3). LOS in both groups was comparable (SG, 9 (7-15) vs. CG, 9 (7-15.25) days, P=0.82). Looking at ≥1 complications and LOS, ASA III subgroup (SG, 16 (13-46) vs. CG, 17.5 (9.75–39.75) days, P=0.92) and ASA II subgroup (SG, 9 (7-21) vs. CG, 10 (8-13) days, P=0.48) showed no significant difference. Data were presented as median (IQR). With Vol. 82 - No. 11 respect to the patient collective without any complication and LOS, ASA III subgroup and ASA II subgroup again revealed no significant differences between SG and CG (Table III). Discussion The present single center study demonstrated that EGDT based on the non-invasive Nexfin™ technology was feasible in ASA II and III patients undergoing major open abdominal Minerva Anestesiologica 1165 BROCH NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY Figure 5.—Number of postoperative complications in the study group and the control group listed according to the ASA classification. *P<0.05 (vs. control group). surgery. Furthermore, preemptive hemodynamic optimization using an algorithm based on continuous non-invasive CI and PPV calculations was able to reduce the rate of postoperative complications, however without reaching statistical significance. Patients at higher risk (ASA III) benefited more from EGDT than patients at lower risk (ASA II). There was no difference in hospital LOS between the SG and the CG. To our best knowledge, this is the first study investigating the feasibility of a completely non-invasive approach based on the Nexfin™ technology for EGDT in a larger patient collective. The concept of early, preemptive algorithm-based titration of fluids and inotropes has been shown to reduce postoperative morbidity and mortality in different clinical scenarios.4 However, it is recognized that early implementation of GDT has a higher beneficial impact in patients exhibiting more severe comorbidities.2 Therefore, definition of high risk surgical procedures and identification of appropriate patients are indispensable prerequisites for performing efficient EGDT. Several years ago, Shoemaker and colleagues defined criteria for selecting surgery and patients associated with 1166 higher risk.14 These criteria have been slightly modified over the years and firmly established in daily clinical routine.4, 15 In this context, we obtained a reduced total amount of complications in ASA III classified patients in the SG compared to the ASA III patients of the CG, however without reaching statistical significance. Moreover, we observed a higher rate of patients in the SG without any complication. Again, these observations were statistically insignificant. A possible explanation of these results could be the composition of patients with less comorbidities in the present study. In this context, we observed considerable more ASA II patients in the SG as well in the CG. This is of crucial clinical importance as a recent investigation could demonstrate that EGDT performed in low risk patients could be associated with adverse effects on primary outcome.16 It must be noted, however, that we did not observe detrimental effects regarding our EGDT protocol in both patient collectives. On the contrary, by applying an EGDT algorithm based on non-invasive monitoring, we recognized a slight trend towards less or zero complications in ASA III patients of the SG compared to ASA III patients of the CG. Minerva AnestesiologicaNovember 2016 NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY Furthermore, by avoiding complications, ASA III patients revealed a similar hospital LOS compared to ASA II patients exhibiting less comorbidities. This potential benefit for the ASA III subgroup was even more pronounced considering that occurrence of ≥1 complication increased the hospital LOS more than three times in this patient population. Another explanation of our results could be related to the experience of the attending anesthesiologist. Anesthesia for both patient collectives was performed by a consultant with long-standing experience. Although patients in the CG received a hemodynamic therapy simply based on invasive variables like MAP and CVP we observed similar intraoperative lactate levels in both groups which revealed no statistic significant differences. As we used dynamic variables like PPV, some confounders should be noted. Several studies identified heart rhythm disturbances, respiratory rate17, spontaneous breathing efforts18, presence of intraabdominal hypertension19 and/ or right ventricular failure20 as clinical entities with a potentially relevant impact on the predictive accuracy of dynamic variables. To evaluate whether differences in arterial tone between both groups have influenced fluid responsiveness, dynamic arterial elastance was determined as suggested by recent literature.21 However, we observed no significant differences in arterial elastance between the study and the control group. Since none of these confounders has been identified in our study, one main requirement regarding the application of these variables was fulfilled. Moreover, several studies in recent times have demonstrated that Nexfin-based non-invasive assessment of PPV allows an acceptable prediction of fluid responsiveness.22 In contrast, another study reported that SVV and PPV given by the non-invasive Nexfin device were not able to predict fluid responsiveness.23 Both studies were performed in patients after cardiac surgery. However, two recently published studies could demonstrate that dynamic variables calculated by non-invasive finger cuff were able to adequately predict fluid responsiveness.24, 25 Consequently, and this was our as- Vol. 82 - No. 11 BROCH sumption, in combination with the literature proved sufficient CI-trending ability of the Nexfin technology,26 an EGDT protocol based on these non-invasive variables per se should be able to primarily avoid occult hypovolemia and secondarily maintain stable hemodynamics. Although our results are not significant enough to claim that non-invasive driven EDGT protocols per se have the potential to reduce postoperative outcome in this patient population, we have proven the feasibility. Obviously, the next reasonable step, based on our presented results, would be an appropriate powered multicenter study. We therefore performed a power analysis with a two sided 0.05 difference and a power of 80% to achieve a reduction in postoperative morbidity of 10%, yielding a sample size of 193 patients for each group. Limitations of the study Some limitations of our study should be noted. We investigated patients without heart rhythm disturbances and advanced peripheral artery occlusive disease or arteriovenous shunts. Keeping in mind, that pulse contour analysis could be impaired in presence of these conditions 27-29 we cannot extrapolate our results to patients exhibiting different comorbidities. With respect to our EGDT protocol, recent literature suggested that a threshold value of PPV ≥13% should be better used for volume therapy control.30 Further research is needed to more clearly define the indications and limitations of EGDT protocols based on non-invasive haemodynamic variables, especially in patients exhibiting more severe comorbidities. Conclusions In conclusion, our results suggest that an EGDT protocol based on non-invasive haemodynamic variables in patients undergoing open abdominal surgery is feasible. Although postoperative complications could be reduced, the EGDT protocol was not able to reach statistical significance and had no impact on LOS. Minerva Anestesiologica 1167 BROCH NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY Key messages —— Implementation of an EGDT protocol based on non-invasive haemodynamic variables in patients undergoing open abdominal surgery was feasible. —— The non-invasive driven EGDT algorithm induced a slight trend towards less or zero complications in ASA III patients. —— By avoidance of complications, ASA III patients revealed a similar hospital LOS compared to ASA II patients. —— Although postoperative complications could be reduced, the EGDT protocol was not able to reach statistical significance and had no impact on LOS. References 1. Hamilton MA, Cecconi M, Rhodes A. A systematic review and meta-analysis on the use of preemptive hemodynamic intervention to improve postoperative outcomes in moderate and high-risk surgical patients. Anesth Analg 2011;112:1392-402. 2. Kern JW, Shoemaker WC. Meta-analysis of hemodynamic optimization in high-risk patients. Crit Care Med 2002;30:1686-92. 3. Mythen MG, Webb AR. Intra-operative gut mucosal hypoperfusion is associated with increased post-operative complications and cost. Intensive Care Med 1994;20:99104. 4. Gurgel ST, do Nascimento P, Jr. Maintaining tissue perfusion in high-risk surgical patients: a systematic review of randomized clinical trials. Anesth Analg 2011;112:1384-91. 5. McGuinness S, Parke R. Using cardiac output monitoring to guide perioperative haemodynamic therapy. Curr Opin Crit Care 2015;21:364-8. 6. Bogert LW, van Lieshout JJ. Non-invasive pulsatile arterial pressure and stroke volume changes from the human finger. Exp Physiol 2005;90:437-46. 7. Stover JF, Stocker R, Lenherr R, Neff TA, Cottini SR, Zoller B, et al. Noninvasive cardiac output and blood pressure monitoring cannot replace an invasive monitoring system in critically ill patients. BMC Anesthesiol 2009;9:6. 8. Broch O, Renner J, Gruenewald M, Meybohm P, Schottler J, Caliebe A, et al. A comparison of the Nexfin(R) and transcardiopulmonary thermodilution to estimate cardiac output during coronary artery surgery. Anesthesia 2012;67:377-83. 9. Clark CE, Taylor RS, Shore AC, Ukoumunne OC, Campbell JL. Association of a difference in systolic blood pressure between arms with vascular disease and mortality: a systematic review and meta-analysis. Lancet 2012;379:905-14. 10. Eeftinck Schattenkerk DW, van Lieshout JJ, van den Meiracker AH, van den Meiracker AH, Wesseling KR, Blanc S, et al. Nexfin noninvasive continuous blood pressure validated against Riva-Rocci/Korotkoff. Am J Hypertens 2009;22:378-83. 1168 11. Penaz J, Voigt A, Teichmann W. Contribution to the continuous indirect blood pressure measurement. Z Gesamte Inn Med 1976;31:1030-3. 12. Wesseling KH, Jansen JR, Settels JJ, Schreuder JJ. Computation of aortic flow from pressure in humans using a nonlinear, three-element model. J Appl Physiol 1993;74:2566-73. 13. Bogert LW, Wesseling KH, Schraa O, Van Lieshout EJ, de Mol BA, van Goudoever J, et al. Pulse contour cardiac output derived from non-invasive arterial pressure in cardiovascular disease. Anaesthesia 2010;65:1119-25. 14. Shoemaker WC, Wo CC, Thangathurai D, Velmahos G, Belzberg H, Asensio JA, et al. Hemodynamic patterns of survivors and nonsurvivors during high risk elective surgical operations. World J Surg 1999;23:1264-70. 15. Lees N, Hamilton M, Rhodes A. Clinical review: Goaldirected therapy in high risk surgical patients. Crit Care 2009;13:231. 16. Challand C, Struthers R, Sneyd JR, Erasmus PD, Mellor N, Hosie KB, et al. Randomized controlled trial of intraoperative goal-directed fluid therapy in aerobically fit and unfit patients having major colorectal surgery. Br J Anaesth 2012;108:53-62. 17. De Backer D, Taccone FS, Holsten R, Ibrahimi F, Vincent JL. Influence of respiratory rate on stroke volume variation in mechanically ventilated patients. Anesthesiology 2009;110:1092-7. 18. Heenen S, De Backer D, Vincent JL. How can the response to volume expansion in patients with spontaneous respiratory movements be predicted? Crit Care 2006;10:R102. 19. Renner J, Gruenewald M, Quaden R, Hanss R, Meybohm P, Steinfath M, et al. Influence of increased intra-abdominal pressure on fluid responsiveness predicted by pulse pressure variation and stroke volume variation in a porcine model. Crit Care Med 2009;37:650-8. 20. Mahjoub Y, Pila C, Friggeri A, Zogheib E, Lobjoie E, Tinturier F, et al. Assessing fluid responsiveness in critically ill patients: False-positive pulse pressure variation is detected by Doppler echocardiographic evaluation of the right ventricle. Crit Care Med 2009;37:2570-5. 21. Monge Garcia MI, Gil Cano A, Gracia Romero M. Dynamic arterial elastance to predict arterial pressure response to volume loading in preload-dependent patients. Crit Care 2011;15:R15. 22. Lansdorp B, Ouweneel D, de Keijzer A, van der Hoeven JG, Lemson J, Pickkers P. Non-invasive measurement of pulse pressure variation and systolic pressure variation using a finger cuff corresponds with intra-arterial measurement. Br J Anaesth 2011;107:540-5. 23. Fischer MO, Coucoravas J, Truong J, Zhu L, Gérard JL, Hanouz JL, et al. Assessment of changes in cardiac index and fluid responsiveness: a comparison of Nexfin and transpulmonary thermodilution. Acta Anaesthesiol Scand 2013;57:704-12. 24. Stens J, Oeben J, Van Dusseldorp AA, Boer C. Noninvasive measurements of pulse pressure variation and stroke volume variation in anesthetized patients using the Nexfin blood pressure monitor. J Clin Monit Comput 2016;30:587-94. 25. Vos JJ, Poterman M, Salm PP, Van Amsterdam K, Struys MM, Scheeren TW, et al. Noninvasive pulse pressure variation and stroke volume variation to predict fluid responsiveness at multiple thresholds: a prospective observational study. Can J Anesth 2015;62:1153-60. 26. Ameloot K, Palmers PJ, Malbrain ML. The accuracy of noninvasive cardiac output and pressure measurements with finger cuff: a concise review. Curr Opin Crit Care 2015;21:232-9. 27. Maj G, Monaco F, Landoni G, Barile L, Nicolotti D, Pieri M, et al. Cardiac index assessment by the pressure re- Minerva AnestesiologicaNovember 2016 NON-INVASIVE HEMODYNAMIC OPTIMIZATION IN MAJOR ABDOMINAL SURGERY cording analytic method in unstable patients with atrial fibrillation. J Cardiothorac Vasc Anesth 2011;25:476-80. 28. Barile L, Landoni G, Pieri M, Ruggeri L, Maj G, Nigro Neto C, et al. Cardiac index assessment by the pressure recording analytic method in critically ill unstable patients after cardiac surgery. J Cardiothorac Vasc Anesth 2013;27:1108-13. 29. Talts J, Raamat R, Jagomagi K. Influence of pulse pres- BROCH sure variation on the results of local arterial compliance measurement: A computer simulation study. Comput Biol Med 2009;39:707-12. 30. Cannesson M, Le Manach Y, Hofer CK, Goarin JP, Lehot JJ, Vallet B, et al. Assessing the diagnostic accuracy of pulse pressure variations for the prediction of fluid responsiveness: a “gray zone” approach. Anesthesiology 2011;115:231-41. Authors’ contributions.—Broch O and Carstens A equally contributed to this manuscript. Funding.—The present study was supported by departmental funding only. Conflicts of interest.—Jochen Renner has received honoraria from Edwards Lifesciences (Irvine, CA, USA) for giving lectures. Berthold Bein is a member of the medical advisory board of Pulsion Medical Systems (Munich, Germany) and has received honoraria for consulting and giving lectures. Matthias Gruenewald has received honoraria from GE Healthcare (Munich, Germany) and Pulsion Medical Systems (Munich, Germany) for giving lectures. No other author has a conflict of interest with regard to any device employed in this study. Acknowledgements.—The authors wish to thank the anesthesiological nursing stuff for their help and assistance in conducting this study. Article first published online: June 28, 2016. - Manuscript accepted: June 23, 2016. - Manuscript revised: June 21, 2016. - Manuscript received: February 12, 2016. Vol. 82 - No. 11 Minerva Anestesiologica 1169 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1170-9 ORIGINAL ARTICLE Propofol versus midazolam for premedication: a placebo‑controlled, randomized double‑blinded study Ofelia L. ELVIR LAZO 1, Paul F. WHITE 1, 2, 3*, Jun TANG 1, Roya YUMUL 1, 4, Xuezhao CAO 1, 5, Firuz YUMUL 1, Jonathan HAUSMAN 1, Antonio HERNANDEZ CONTE 1, Kapil K. ANAND 1, Emad G. HEMAYA 1, Xiao ZHANG 1, Ronald H. WENDER 1 1Department of Anesthesiology, Cedars‑Sinai Medical Center, Los Angeles, CA, USA; 2White Mountain Institute, The Sea Ranch, CA, USA; 3Istituto Ortopedico Rizzoli, University of Bologna, Italy; 4David Geffen School of Medicine‑UCLA, Los Angeles, CA, USA; 5First Affiliated Hospital of China Medical University, Shenyang, China *Corresponding author: Paul F. White, The White Mountain Institute, 41299 Tallgrass, The Sea Ranch, CA 95497, USA. E‑mail: [email protected] A B STRACT BACKGROUND: It has been previously reported that subhypnotic doses of propofol could offer an advantage over midazolam for premedication. This study was designed to test the hypothesis that a 20 mg IV dose of propofol would be more effective than a standard 2 mg IV dose of midazolam for reducing acute anxiety prior to induction of anesthesia. METHODS: One hundred twenty outpatients scheduled to undergo orthopedic surgery were randomly assigned to one of three study groups: control (saline); propofol (20 mg); or midazolam (2 mg). Immediately before administering the study medication, each patient evaluated their level of acute anxiety and sedation on 11‑point verbal rating scales (VRSs) 0=none- 10=highest, and they were also shown a picture. Upon arrival in the OR ~5 min after administering the study medication, anxiety and sedation levels were reassessed and a second picture was shown. At discharge from the recovery area, anxiety and sedation levels and their ability to recall the two pictures were reassessed. RESULTS: Compared to the saline group, both propofol and midazolam produced significant increases in the patient’s level of sedation upon entering the OR (+2.5±2.4 vs. +4.6±2.5 and +5.2±2.3, respectively [p<0.001]). Propofol was ef‑ fective as midazolam compared to saline in reducing the patient’s level of preinduction anxiety (from 3.2±2.2 to1.8±1.8 vs. 3.1±2.2 to 2.3±2.1 and 2.7±1.8 to 2.8±2.1, respectively). Propofol produced more pain on injection and midazolam significantly reduced recall of the second picture. CONCLUSIONS: When administered ~5 min prior to entering the OR, propofol, 20mg IV, was as effective as midazolam 2mg IV in reducing anxiety. (Cite this article as: Elvir Lazo OL, White PF, Tang J, Yumul R, Cao X, Yumul F, et al. Propofol versus midazolam for pre‑ medication: a placebo‑controlled, randomized double‑blinded study. Minerva Anestesiol 2016;82:1170-9) Key words: Preoperative care - Anxiety - Amnesia - Propofol - Midazolam. E arlier studies 1‑3 have reported that the vast majority of outpatients would chose to receive a sedative‑anxiolytic drug prior to entering the OR. Administration of a small IV dose of midazolam for premedication prior to the patient entering the operating room (OR) is a common clinical practice in the ambula‑ tory setting. However, even small doses of IV 1170 midazolam can contribute to residual sedation and amnesia in the postanesthesia care unit (PACU).1‑2, 4 Propofol has become the most popular IV sedative‑hypnotic medication for both anes‑ thesia and sedation in the ambulatory setting because of its rapid onset and excellent recov‑ ery profile.5, 6 It has been previously reported Minerva AnestesiologicaNovember 2016 PROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATIONELVIR LAZO that subhypnotic doses of propofol could of‑ fer an economic advantage over midazolam for premedication.7 However, there is only one published study evaluating the use of propo‑ fol for IV premedication.7 In this earlier study, Quario and Thompson reported that propofol (0.4 mg/kg IV) had anxiolytic effects compa‑ rable to midazolam (40 µg/kg IV) with less memory impairment, respiratory depression and dizziness.7 The dose of midazolam used in the earlier study was almost twice the stan‑ dard IV premedication dosage and these inves‑ tigators failed to assess the effect of propofol and midazolam premedication on preoperative recall and the early postoperative recovery profile. Therefore, we designed a prospective, randomized, double‑blind, placebo‑controlled study to test the hypothesis that a 20 mg IV bolus dose of propofol would be more effec‑ tive than 2 mg IV bolus dose of midazolam in reducing acute anxiety prior to induction of anesthesia. The secondary objectives were to compare the effects of a small premedication dose of propofol or midazolam on the level of sedation and recall immediately prior to induc‑ tion of anesthesia. Materials and methods This clinical study was approved by Ce‑ dars‑Sinai Medical Center’s Institutional Re‑ view Board (IRB) with # Pro00025204 on 08/30/2013 (which was registered in ClinTri‑ als.gov, Reg. # NCT01976845). It also com‑ plied with all 25 items on the Consort 2010 checklist. After obtaining IRB approval and written informed consent, 120 ASA physical status I‑III adult patients (range: 18‑68 years) scheduled to undergo elective orthopedic sur‑ gery procedures were enrolled in this study and randomly assigned to one of three treatment groups (N.=40/group): control (Saline), pro‑ pofol (20 mg), or midazolam (2 mg). Random‑ ization assignment was generated with a 1:1:1 allocation ratio using a computer software pro‑ gram. The randomization number specifying the study medication (namely, saline, propofol or midazolam) was placed in sealed envelope and given to a non‑participating anesthesi‑ Vol. 82 - No. 11 ologist on the day of the scheduled procedure after completing the patient’s preoperative as‑ sessment. The study was conducted between November 2013 and December 2014. Prior to the day of surgery, potential study patients received a packet of materials from their surgeon describing the study. The pack‑ et contained an initial patient contact letter, a HIPAA information sheet, and the IRB‑ap‑ proved informed consent form. Patients who were interested in participating in the study were asked to bring the materi‑ als to the hospital on the day of surgery. After reviewing all the details of the study with the patient and verifying that they met all the in‑ clusionary criteria, the study coordinator col‑ lected the signed consent forms. The patients were informed that it would be important to complete all aspects of the study, including answering questions in the early postoperative period. Inclusion criteria included the following: Patients to undergo orthopedics procedures, Willingness and ability to sign an informed consent document, no allergies to midazolam or propofol, 18‑70 years of age, American Association of Anesthesiologists Class I‑III adults. Exclusion criteria included the following: Patients with known allergy, hypersensitivity or contraindications to midazolam, propofol, standardized anesthetic and analgesic medi‑ cations administered during the study period, unstable preexisting medical conditions in‑ volving the central nervous system (CNS), pregnant or lactating women, history of alco‑ hol or drug abuse within the past 3 months, chronic use of sedative‑hypnotic or anxiolytic medications prior to the operation, morbidity obesity (Body Mass Index [BMI] >40 kg/m2), unstable angina, diseases requiring oxygen therapy, obstructive sleep apnea (OSA) and sleep disordered breathing. Withdrawal criteria included the follow‑ ing: Allergies or hypersensitivity to propofol or midazolam, patients present any clinical‑ ly‑significant medical conditions involving the brain, heart, kidney, liver or endocrine diseas‑ es, patients agitated or confused with a level Minerva Anestesiologica 1171 ELVIR LAZOPROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATION of anxiety greater than 6 prior to receiving the study drug, patient who received additional premedication upon entering into the OR. In the preoperative holding area, consent‑ ing patients were asked to provide a detailed medical history including demographic infor‑ mation (e.g., age, weight, height, BMI), smok‑ ing history, chronic medications, recreational drug usage, and history of motion sickness or previous postoperative nausea and vomiting (PONV). Each patient was connected to moni‑ toring devices for recording their respiratory rate, oxygen saturation, heart rate, and blood pressure values as part of the standard of care. In the preoperative holding area immedi‑ ately before administering the study medi‑ cation each patient evaluated their level of acute anxiety and sleepiness (sedation) using 11‑point verbal rating scales (VRSs) (0=none to 10=extremely high). The VRSs have been used previously to assess sedation and anxiety levels.8‑11 In addition, each patient was evalu‑ ated using the Observer’s Assessment of Alert‑ ness/Sedation (OAA/S) Scale 12 (Appendix I) and the Ramsay Scale (Appendix II).13 After performing these assessments, the patients were shown a picture of an apple. The patients were then administered 2 mL IV of the study medication in an opaque syringe containing saline (2 mL), propofol (20 mg), or midazolam (2 mg) 3‑6 min prior to being transported to the OR by an anesthesiologist who was not in‑ volved in caring for the patient or performing the perioperative assessments. The syringes were covered with an opaque tape to blind the medication from the patient and the individual responsible for administering the study medi‑ cation. Each patient was asked whether or not they experienced any discomfort during the injec‑ tion of the study medication and a binary “yes or no” was recorded. The investigators in‑ (N.=161) (N.=0) (N.=0) (N.=22) (N.=0) (N.=139) (N.=48) (N.=48) (N.=48) (N.=48) (N.=43) (N.=43) (N.=0) (N.=0) (N.=0) (N.=0) (N.=8) (Analyzed N.=40) (N.=0) (N.=0) (N.=0) (N.=8) Analyzed (N.=40) (N.=0) (N.=3) Analyzed (N.=40) (N.=0) Figure 1.—Consort 2010 flow diagram. 1172 Minerva AnestesiologicaNovember 2016 PROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATIONELVIR LAZO volved in performing the perioperative assess‑ ments were blinded as to the patient’s response the question regarding pain on injection of the study medication. Upon arrival in the OR ~5 min after administering the study medication, anxiety and sedation levels were reassessed using the same 11‑point VRSs and a second picture (a dollar bill) was shown to the pa‑ tients. The patient’s level of consciousness was also reassessed using the OAA/S and Ramsay scales prior to induction of anesthesia. After applying the standard OR monitoring devices, general anesthesia was induced with a combi‑ nation of lidocaine 30‑50 mg IV and propofol 2‑2.5 mg/kg IV followed by desflurane 3‑6% or sevoflurane 1‑2% titrated to maintain he‑ modynamic stability. A laryngeal mask airway (LMA) device was used in all cases except in 3 cases where the anesthesiologists decided to perform tracheal intubation with an endotra‑ cheal tube. A local anesthetic infiltration was adminis‑ tered to all patients before performing the sur‑ gical incision and a peripheral nerve block at the end of the surgical procedural for periop‑ erative analgesia. After the operation, patients were transported to the PACU. Upon discharge from the recovery area, anxiety and sedation levels were again assessed using the VRSs and patients were queried regarding their ability to recall the two pictures. The standardized perioperative assessments included: 1) patient demographic data; 2) lev‑ el of anxiety (i.e., nervousness) and sedation (i.e., sleepiness) at three time points (In the preoperative area prior to being transported to the OR, before induction of anesthesia, and prior to discharge from the recovery area); 3) dosages of all anesthetic drugs, local anesthet‑ ics, and IV fluid therapy during the operation; 4) duration of surgery (from skin incision until closure) and anesthesia (from IV induction un‑ til discontinuation of the volatile anesthetic); 5) duration of the PACU stay; 6) requirements for “rescue” analgesic and antiemetic medica‑ tion in the PACU, 7) Side effects in the PACU prior to discharge home; 8) time from baseline assessment to study drug administration; 9) time from study drug administration until the Vol. 82 - No. 11 pre‑induction assessments were performed; and 10) ability to recall the two pictures shown before the start of surgery. None of the study patients received any type of sedative‑anxiolytic premedication prior to enrollment in this study. All consenting pa‑ tients received their premedication in the pre‑ operative holding area ~5 minutes prior to en‑ tering the operating room (OR). If the patient received additional premedication upon enter‑ ing into the OR, their participation in the study was terminated and their data was excluded from the final statistical analysis. Statistical analysis A sample size of 34 patients in each group was determined using a power analysis based on the presumption than both propofol and midazolam will decrease anxiety score by 25% or more compared to the Control (sa‑ line) group (VRS=3, 3, and 4, respectively), SD=1.5, with a power of 80% and significance level of 0.05 (using 2‑sided t‑test). However, to reduce the risk of a type II statistical er‑ ror, we decided before starting the study to increase the sample size to 40 patients per group.7 The analysis was performed using SAS 9.3 for Windows (SAS Institute, Cary, NC, USA) and R. 3.0.1. Our dataset contained both categorical and continuous measure‑ ments. For categorical measures, we presented total numbers (N.) with the percentages (%) and used χ2 test (or Fisher’s Exact Test) to conduct the group comparisons. For continu‑ ous measures, we presented mean values with their standard deviations and used the Kol‑ mogorov‑Smirnov test to check the normality. To conduct the multiple comparisons among those 3 groups, we performed the one‑way ANOVA if the measure is approximately nor‑ mal and the Kruskal Wallis test if the measure fails to pass the normality test. A Bonferroni correction was applied when multiple com‑ parisons were performed over time. All tests were two‑sided; and P values ≤0.05 were con‑ sidered statistically‑significant. Data are pre‑ sented as mean values±SD, numbers (N.), and percentages (%). Minerva Anestesiologica 1173 ELVIR LAZOPROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATION Results The three groups were comparable with re‑ spect to demographic characteristics, duration of anesthesia, surgery and recovery stay, type of surgery, vital signs, oxygen saturation and baseline sedation and anxiety scales among the three treatment groups (Table I, Figure 2). Compared to the saline group, both propofol and midazolam produced significant increases in the patient’s level of sedation (or sleepiness) upon entering the OR (+2.5±2.4 vs. +4.6±2.5 and +5.2±2.3, respectively [P<0.001] and compared to their respective baseline values). Propofol and midazolam were equivalent in effectiveness in reducing the patient’s level of preinduction anxiety compared to saline (from 3.2±2.2 to 1.8±1.8 vs. 3.1±2.2 to 2.3±2.1 and 2.7±1.8 to 2.8±2.1, respectively) (Figure 3). Although propofol produced more pain on injection than midazolam and saline (40% vs. 23% and 10%, respectively P≤ 0.007), mid‑ azolam significantly reduced recall of the sec‑ ond picture compared to propofol and saline (30% vs. 75% and 95%, respectively, P≤0.001) (Table I). Table II summarizes the intraoperative drug usage in the three premedication treatment Table I.—Demographic characteristics, duration of anesthesia, surgery and recovery stay, surgery type, OAAS and Ramsay sedation scale, picture recall and pain on administration of the study drug for the three premedication treatment groups. Age (yr) BMI (kg/m2) Gender (N.) F/M ASA (N.) I/II/III Race (N.) Asian/Black/Caucasian/Hispanic Smoker (N.) Alcohol user (N.) History of PONV (N.) yes/no Took sleeping or anti‑anxiety drugs the night before (N.) General anesthesia Sedation Peripheral nerve block at the end of surgery Anesthesia time (min) Surgery time (min) Time to discharge (min) to the postsurgical ward for arthroplasty cases Time to discharge (min) to home for the non‑arthroplasty cases Time from baseline assessment to study drug (min) Time from administration of study drug until the pre‑induction assess‑ ments were performed (min) Type of orthopedic surgery (N.): Arthroplasty Arthroscopy Open reduction internal fixation (ORIF) Tendon/ligament repair/biopsy OAAS§ (N.) (4 /5) baseline level (4/5) before induction level Ramsay (N.) baseline level Before induction level Memory [picture] recall: Pre‑picture # 1 (N.) yes/no Post‑picture # 2 (N.) yes/no Pain on administration (N.) Saline (N.=40) Propofol (N.=40) Midazolam (N.=40) P 49±14 28±5 15/25 16/19/5 1/4/35/0 2 21 7 12 13 27 18 107±45 79±41 172±92 116±70 28±28 3.8±2 52±13 28 ± 5 24/16 6/28/6 3/5/31/1 5 17 4 13 14 26 17 116±48 84±44 199±105 102±50 32±42 3.9±2 51 ±15 28 ± 6 17/23 12/24/4 3/6/31/0 5 21 9 11 16 24 23 130±61 102±54 184±100 109±30 24±26 4.6±2 0.7 0.9 0.1 0.2 0.8 0.4 0.6 0.3 0.9 0.8 0.8 0.6 0.2 0.07 0.8 0.7 0.5 0.1 21 7 6 5 0/40 2/37 40 40 25 7 4 4 0/40 6/34 40 40 23 6 6 5 0/40 7/33 40 40 0.9 0.9 0.8 0.9 NA 0.2 NA NA 40/0 38/2 4 39/1 30/10*† 16* 40/0 12/28*†‡ 9 NA <0.001 0.007 Values are mean±SD or numbers (N.) *Significantly different from Saline group, P<0.05. †Significantly different from the Baseline value, P<0.05. ‡Significantly different from the Propofol group, P<0.05. §OAAS: Observer Assessment of Alertness and Sedation Scale. 1174 Minerva AnestesiologicaNovember 2016 PROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATIONELVIR LAZO P value=0.6 P value=0.6 P value=0.8 P value=0.8 P value=0.7 P value=0.8 Figure 2.—Comparative effects of midazolam, propofol and saline on mean arterial pressure (MAP), the heart rate (HR), and oxygen saturation during the perioperative period for the three premedication treatment groups. Values are mean±SD or numbers (n). * Significantly different from Saline group, P<0.05. Baseline: before the administration of the premedication. Before Induction: approximately 5 minutes after the administration of the premedication. groups. There were no significant differences in intraoperative drug usage or emergence times from anesthesia among the three treat‑ ment groups. In addition to the length of the PACU stay; the incidence of side effects and requirements for rescue analgesic and anti‑ emetic medications were also similar among the three treatment groups (Table II). All patients were conscious and were re‑ sponsive to questions from the investigator at all times prior to induction of anesthesia. The patient’s level of consciousness was unchanged from their baseline values when assessed using Vol. 82 - No. 11 the OAA/S 12 and Ramsay 13 scales (Table I). Compared to saline, the administered dosag‑ es of propofol and midazolam failed to delay time to discharge to the postsurgical ward for arthroplasty cases or to discharge to home for the non‑arthroplasty cases. Discussion Premedication is considered to be a valuable part of the perioperative surgery experience by patients and anesthesia providers.14‑16 In the current study, premedication with propofol 20 Minerva Anestesiologica 1175 ELVIR LAZOPROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATION Anxiety Level Saline Propofol Baseline Level Sedation Level Midazolam Saline Before Induction Level Propofol Midazolam Postoperative Level Figure 3.—Comparative effects of propofol and midazolam (vs. saline) on the level of sedation and anxiety for the three premedication treatment groups. Values are mean±SD or numbers (n). * Significantly different from Saline group, P<0.05. † Significantly different from the Baseline value, P<0.05. Anxiety level (0=none to 10= extremely nervous) and Sedation level (0=none to 10= extremely se‑ dated/sleepy). Baseline: before the administration of the premedication. Before Induction: approximately 5 minutes after the administration of the premedication. mg IV, was found to produce significant anx‑ iolytic, sedative and amnestic effects without cardiorespiratory depression. The only disad‑ vantage of using propofol was more frequent mild discomfort upon IV injection. The use of a relatively small‑dose of propofol 20 mg IV avoided excessive CNS depression and avoid‑ ed delaying the early recovery processes after these minor (non‑arthroplasty) ambulatory orthopedic procedures. Both midazolam and propofol produced greater amnesia than saline; however, propofol predication was associated with less impairment of recall than midazolam (P<0.001). These findings are consistent with earlier studies documenting the significant an‑ terograde amnestic effects of midazolam pre‑ medication.1, 7 However, this is the first study documenting the amnestic effect of a subhyp‑ notic dose of propofol. 1176 The benzodiazepine drug class has been al‑ leged to produce dose‑dependent anxiolytic, sedative and amnestic effects when adminis‑ tered for premedication.10 However, the dos‑ age which is commonly administered for pre‑ medication, namely 2 mg IV (20‑25 µg/kg IV), failed to produce a statistically‑significant decrease in the patient’s level of preoperative anxiety compared to saline. Given its more fa‑ vorable anxiolytic effects (and lesser degrees of amnesia), low‑dose propofol may offer ad‑ vantages over midazolam for IV premedica‑ tion of anxious patients in the ambulatory set‑ ting. A larger scale follow‑up study is clearly needed to evaluate the impact of propofol pre‑ medication in the preoperative holding area and its impact on other clinical outcomes after ambulatory surgery. Propofol has the advantage of a more rapid Minerva AnestesiologicaNovember 2016 PROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATIONELVIR LAZO Table II.—Intraoperative Drugs Administered, Incidence of Side Effects, Need for Rescue Analgesic and Antiemetic Drugs Administered to the three premedication treatment groups. Intraoperative Drugs Lidocaine 50‑100 mg (n) Fentanyl µg (mcg) Propofol (mg) Glycopyrrolate 0.2‑1mg (n) Bupivacaine (mg) Ondansetron 4 mg (n) Ephedrine 10‑20 mg (n) Famotidine 20 mg (n) Hydromorphone (n) Morphine mg (n) Total Fluids (mL) Side Effects Nausea (n) Itching (n) Rescue analgesic requirement: Hydromorphone 0.2‑3 mg (n) Fentanyl 50‑100 mcg (n) Ketorolac 50 mg (n) Norco Tab (n) Lortab (n) Oxycodone (mg) Tramadol 50‑100 mg (n) Acetaminophen 650 mg‑1000 (n) Celebrex 200mg (n) Ondansetron (n) Saline (N.=40) Propofol (N.=40) Midazolam (N.=40) P value 6 88±40 500±357 4 31±23 20 1 12 0 0 1471±818 10 113±65 577±360 1 31±15 13 4 7 3 3 1646±708 3 125±82 681±505 5 40±19 18 2 9 3 0 1595±651 0.1 0.35 0.15 0.24 0.30 0.26 0.3 0.4 0.2 NA 0.54 5 0 6 1 4 0 0.8 NA 16 3 0 8 2 4 2 6 2 5 16 2 1 6 1 6 1 4 1 7 14 1 0 5 0 4 1 5 1 5 0.8 0.6 NA 0.6 0.4 0.7 0.8 0.8 0.8 0.2 Values are mean±SD or numbers (n). * Significantly difference from Saline and midazolam group, P<0.05 onset of action (30 seconds vs. 120 seconds for midazolam) and a shorter duration of ef‑ fect (biologic half‑life of 4 minutes vs. 30 minutes for midazolam), and can also produce dose‑dependent sedation.17 Consistent with the earlier study,7 we did not find any difference in the level of consciousness (OAA/S and Ram‑ say scales [Table I]) produced by propofol 20 mg and midazolam 2 mg prior to induction of anesthesia. Importantly, all patients were fully conscious, cooperative and responsive to ques‑ tions from the anesthesiologists immediately prior to induction of anesthesia with the doses of propofol and midazolam administered in the preoperative holding area prior to transferring the patient to the OR. One of the major concerns regarding the routine use of propofol for premedication re‑ lates to the possible need for additional moni‑ toring and/or supplemental oxygen in the pre‑ operative holding area. Although induction doses of propofol can produce dose‑dependent Vol. 82 - No. 11 decreases in blood pressure and heart rate, as well as apnea and respiratory depression, doses of propofol ranging from 40 to 150 mg (total dose) were not found to be associated with significant cardiovascular or respiratory changes.18 The findings of a meta‑analysis by Wang et al.19 are also consistent with our find‑ ings that low‑dose propofol sedation does not increase the risk of bradycardia, hypotension, or hypoxemia compared with midazolam seda‑ tion. Compared to saline, neither the anesthesia time nor the duration of the PACU stay were prolonged by use of propofol (20 mg e.v.) or midazolam (2 mg IV) for premedication. Fur‑ thermore, the postoperative pain scores, inci‑ dence of PONV, and need for rescue analge‑ sics and antiemetic medications did not differ among the three treatment groups, demonstrat‑ ing that these premedicant dosages of midazol‑ am or propofol did not increase postoperative side effects compared to saline. Minerva Anestesiologica 1177 ELVIR LAZOPROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATION Although the earlier study by Quario and Thompson 7 examining the use of propofol as an alternative to midazolam for premedication reported similar findings, these authors admin‑ istered larger doses of both propofol (0.4 mg/kg vs. 0.26 mg/kg) and midazolam (40 µg/kg vs. 24 µg/kg). The standard premedication dosage of midazolam is 20‑25 µg/kg e.v.). With larger dosages of these CNS depressant drugs, elderly and higher risk patients’ populations might ex‑ perience both cardiovascular and respiratory depression. In the earlier study,7 midazolam premedication in the absence of supplemental oxygen was associated with a decrease in SpO2 values <98% in 45% of the patients (vs. 10% in the propofol and saline groups, respectively). Despite breathing room air in the current study, SpO2 values did not show any significant differ‑ ences among the three treatment groups. In the earlier study, both the larger dosages of propo‑ fol and midazolam also produced small but sta‑ tistically significant decreases in MAP values. Although propofol was highly effective when administered for IV premedication, 16 patients (40%) in our study reported mild discomfort on injection of the low‑dose of propofol. The addi‑ tion of a small amount of lidocaine to the propo‑ fol formulation would have further reduced (or even eliminated) the discomfort on injection of the predication.20 Another potential criticism of this study is that fixed dosages of the two study drugs were administered to all patients irrespective of their body weight, they were chosen arbi‑ trarily based on standard clinical practices in North America to administer fixed doses of the medications which are commonly used for pre‑ medication. However, in the future, a proper dose‑response study should be performed to determine the optimal doses of each drug for endovenous premedication. Another limita‑ tion was the exclusion of patients with anxi‑ ety >6/10; this could biased the patient selec‑ tion, but this was a requirement from IRB in order to perform the study. Fortunately, none of the prospective patients were excluded from participating in the study due to high levels of anxiety. Finally, the absence of any compara‑ tive studies that analyze other types of memory 1178 (non‑declarative) testing, and the lack of ad‑ ditional memory evaluations in our study, are noteworthy study limitations. Conclusions When administered ~5 minutes prior to en‑ tering the OR, propofol, 20mg IV, appears to be an effective anxiolytic as midazolam, 2 mg IV. Compared to midazolam, propofol pro‑ duced a similar level of sedation but caused less impairment of recall prior to induction of anesthesia. Key messages —— Premedicant doses of propofol (20 mg) and midazolam (2 mg) which pro‑ duced comparable levels of sedation were associated with similar reductions in pre‑ operative anxiety. —— At equivalent levels of sedation, midazolam produced greater impairment of recall at induction of anesthesia com‑ pared to propofol. —— Premedicant dose of propofol (20 mg) or midazolam (2 mg) did not produce significant cardiorespiratory changes. —— When propofol (20 mg) or midazol‑ am (2 mg) were administered for IV pre‑ medication immediately prior to entering the operating room, these sedative-hypnot‑ ics failed to delay discharge home (outpa‑ tients) or to the surgical ward (inpatients). References 1.Shafer A, White PF, Urquhart ML, Doze VA. Outpatient premedication: use of midazolam and opioid analgesics. Anesthesiology 1989;71:495‑501. 2.Van Vlymen JM, Sá Rêgo MM, White PF. Benzodiaz‑ epine premedication: can it improve outcome in patients undergoing breast biopsy procedures? Anesthesiology. 1999;90:740‑7. 3.Raeder JC, Breivik H. Premedication with midazolam in out‑patient general anaesthesia: a comparison with morphine‑scopolamine and placebo. Acta Anaesthesiol Scand 1987;31:509‑14. 4. Kain ZN, Mayes LC, Bell C, Weisman S, Hofstadter MB, Rimar S. Premedication in the United States: a status re‑ port. Anesth Analg 1997;84:427‑32. Minerva AnestesiologicaNovember 2016 PROPOFOL VERSUS MIDAZOLAM FOR PREMEDICATIONELVIR LAZO 5.White PF. Ambulatory anesthesia and surgery: past, present and future. In: White PF, editors. Ambulatory an‑ esthesia & surgery. London: WB Saunders Co; 1997. p. 1‑34. 6. Doze VA, Westphal LM, White PF. Comparison of pro‑ pofol with methohexital for outpatient anesthesia. Anesth Analg 1986;65:1189‑95. 7. Quario Rondo L, Thompson C. Efficacy of propofol compared to midazolam as an intravenous premedication agent. Minerva Anestesiol 2008;74:173‑9. 8. White PF, Coe V, Shafer A, Sung ML. Comparison of al‑ fentanil with fentanyl for outpatient anesthesia. Anesthe‑ siology 1986;64:99‑106. 9.Shafer A, White PF, Urquhart ML, Doze VA. Outpatient premedication: use of midazolam and opioid analgesics. Anesthesiology 1989;71:495‑501. 10. White PF, Tufanogullari B, Taylor J, Klein K. The effect of pregabalin on preoperative anxiety and sedation levels:a dose‑ranging study. Anesth Analg. 2009;108:1140‑ 5. 11.Tanasale B, Kits J, Kluin PM, Trip A, Kluin‑Nelemans HC. Pain and anxiety during bone marrow biopsy. Pain Manag Nurs 2013;14:310‑7. 12.Chernik DA, Gillings D, Laine H, Hendler J, Silver JM, Davidson AB, et al. Validity and reliability of the Ob‑ server’s Assessment of Alertness/ Sedation Scale: study with intravenous midazolam. J Clin Psychopharmacol 1990;10:244‑51 13.Ramsay MAE, Savege TM, Simpson BRJ, Goodwin R. Controlled sedation with alpaxalone‑alphadolone. Br Med J 1974;2:656‑9. 14.Cressey DM, Claydon P, Bhaskaran NC, Reilly CS. Ef‑ fect of midazolam pretreatment on induction dose re‑ quirements of propofol in combination with fentanyl in younger and older adults. Anaesthesia 2001;56:108‑13. 15. Bergendahl H, Lonnqvist PA, Eksborg S. Clonidine: an alternative to benzodiazepines for premedication in chil‑ dren. Current Opin Anaesthesiol 2005;18:608‑13. 16. Bergendahl H, Lonnqvist PA, Eksborg S. Clonidine in paediatric anaesthesia: review of the literature and com‑ parison with benzodiazepines for premedication. Acta Anaesthesiol Scand 2006;50:135‑43. 17. Triantafillidis JK, Merikas E, Nikolakis D, Papalois AE. Sedation in gastrointestinal endoscopy: current issues. World J Gastroenterol 2013;19:463‑81. 18.Paspatis GA, Manolaraki M, Xirouchakis G, Papan‑ ikolaou N, Chlouverakis G, Gritzali A. Synergistic seda‑ tion with midazolam and propofol versus midazolam and pethidine in colonoscopies:a prospective, randomized study. Am J Gastroenterol 2002;97:1963‑7. 19. Wang D, Wang S, Chen J, Xu Y, Chen C, Long A, et al. Propofol combined with traditional sedative agents versus propofol- alone sedation for gastrointestinal endoscopy:a meta‑analysis. Scand J Gastroenterol 2013;48:101‑10. 20. Jalota L1, Kalira V, George E, Shi YY, Hornuss C, Radke O, et al. Perioperative Clinical Research Core. Preven‑ tion of pain on injection of propofol: systematic review and meta‑analysis. BMJ. 2011 15;342:d1110. Funding.—This study was funded by Department of Anesthesiology, Cedars‑Sinai Medical Center, Los Angeles, CA, USA. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Congresses.—This report was previously presented in part at the ASA and WARC meetings. Article first published online: September 9, 2016. - Manuscript accepted: September 7, 2016. - Manuscript revised: July 28, 2016. Manuscript received: February 4, 2016. Appendix I The observer’s assessment of alertness/sedation (OAAS) rating scale OAAS score 5—awake and responds readily to name spoken in normal tone. OAAS score 4—lethargic responses to name in normal tone. OAAS score 3—responds only after name is called loudly and/or repeatedly. OAAS score 2—responds only after name called loudly and mild shaking. OAAS score 1—does not respond when name is called loudly and mild shaking or prodding. OAAS score 0—does not respond to noxious stimulation. Appendix II Ramsey Sedation Scale Ramsey 1 Anxious, agitated, restless. Ramsey 2 Cooperative, oriented, tranquil. Ramsey 3 Responsive to commands only. Ramsey 4 Brisk response to light glabellar tap or loud auditory stimulus. Ramsey 5 Sluggish response to light glabellar tap or loud auditory stimulus. Ramsey 6 No response to light glabellar tap or loud auditory stimulus. Vol. 82 - No. 11 Minerva Anestesiologica 1179 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1180-8 ORIGINAL ARTICLE The optimal time between clinical brain death diagnosis and confirmation using CT angiography: a retrospective study Lionel KERHUEL 1, Mohamed SRAIRI 1 *, Gilles GEORGET 1, Fabrice BONNEVILLE 2, Ségolène MROZEK 1, Nicolas MAYEUR 1, Laurent LONJARET 1, Sandrine SACRISTA 1, Nathalie HERMANT 1, Fouad MARHAR 1, François GAUSSIAT 1, Timothée ABAZIOU 1, Diane OSINSKI 1, Benjamin LE GAILLARD 1, Rémi MENUT 1, Claire LARCHER 1, Olivier FOURCADE 1, Thomas GEERAERTS 1 1Department of Anaesthesia and Intensive Care, University Hospital of Toulouse, University Toulouse 3 Paul Sabatier, Toulouse, France; 2 Department of Neuroradiology, University Hospital of Toulouse, University Toulouse 3 Paul Sabatier, Toulouse, France *Corresponding author: Mohamed Srairi, Department of Anaesthesia and Intensive Care, University Hospital of Toulouse, 1 Place Baylac TSA 40031, 31059 Toulouse cedex 09, France. E‑mail: srairi.m@chu‑toulouse.fr A B STRACT BACKGROUND: In several countries, a computed tomography angiography (CTA) is used to confirm brain death (BD). A six‑hour interval is recommended between clinical diagnosis and CTA acquisition despite the lack of strong evidence to support this interval. The aim of this study was to determine the optimal timing for CTA in the confirmation of BD. METHODS: This retrospective observational study enrolled all adult patients admitted between January 2009 and De‑ cember 2013 to the intensive care units of a French university hospital with clinically diagnosed BD and at least one CTA performed as a confirmatory test. The CTAs were identified as conclusive (e.g. yielding confirmation of BD) or inconclusive (e.g. showing persistent brain circulation). RESULTS: One hundred and four patients (sex ratio M/F 1.8; age 55 years [41‑64]) underwent 117 CTAs. CTAs con‑ firmed cerebral circulatory arrest in 94 cases yielding a sensitivity of 80%. Inconclusive CTAs were performed earlier than conclusive ones (2 hours [1‑3] vs. 4 hours [2‑9], P=0.03) and were associated with decompressive craniectomy (5 cases [23%] vs. 6 cases [7%], P=0.05) and the failure to complete full neurological examination (5 cases [23%] vs. 4 cases [5%], P=0.02). Six hours after BD clinical diagnosis, the proportion of conclusive CTA was only 51%, with progressive increase overtime with more than 80% of conclusive CTA after 12 hours. CONCLUSIONS: A 12‑hour interval might be appropriate in order to limit the risk of inconclusive CTAs. (Cite this article as: Kerhuel L, Srairi M, Georget G, Bonneville F, Mrozek S, Mayeur N, et al. The optimal time between clin‑ ical brain death diagnosis and confirmation using CT angiography: a retrospective study. Minerva Anestesiol 2016;82:1180-8) Key words: Brain death - Tomography, X‑Ray computed - Angiography. I n 50% of European countries, the confirma‑ tion of a clinically diagnosed brain death (BD) by ancillary tests is mandatory before or‑ gan donation.1, 2 Brain computed tomography angiography (CTA) is the preferred confirma‑ tory test in approximately 40% of European countries,1 but also outside Europe.3 In coun‑ 1180 tries where confirmatory tests are not manda‑ tory like Canada, United States of America or United Kingdom, CTA remains recommended when uncertainty exists, and particularly when neurological examination cannot be fully com‑ pleted.4 BD can be affirmed using CTA when absence of contrast enhancement is found on a Minerva AnestesiologicaNovember 2016 OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD selected number of brain vessels.5 In France, a four‑vessel CTA interpretation model is cur‑ rently used, according to recent guidelines.6 The use of the four‑vessel score is supported by the study of Frampas et al.7 The French Society of Neuroradiology recommends a minimum six‑hour interval between clinically diagnosed BD and the ac‑ quisition of CTA, in order to allow the intra‑ cranial pressure (ICP) to exceed the mean arte‑ rial pressure (MAP) both in the infratentorial and the supratentorial compartments, leading to a cerebral circulatory arrest (CCA) 6 despite the fact that no strong evidence supports this six‑hour time interval. Interestingly, the rec‑ ommended time lag is the same regardless of BD etiology especially anoxic brain damage that is not managed differently from the other conditions. Determining the optimal timing for CTA may prevent unnecessary transport to the radiology facility and may improve the diagnostic performance of CTA for BD con‑ firmation. Moreover, several limits of CTA regarding its insufficient sensitivity (e.g. pre‑ served intracranial vascular filling in clinically brain‑dead patients) have been described, rais‑ ing serious concerns about its use as a compul‑ sory test in particular settings, such as posterior fossa primary lesion, skull fracture, decom‑ pressive craniectomy, ventricular drainage, and anoxic encephalopathy.8, 9 Most relevant studies assessing CTA sensitivity used clinical examination as a gold standard, have shown a sensitivity between 65% and 100%.7, 10‑14 This variability may preclude routine CTA use in common practice. The main objective of this study was to de‑ termine the optimal CTA timing for BD confir‑ mation. We also aimed to study the risk factors for inconclusive CTA (e.g. showing persistent brain circulation). Materials and methods Ethical considerations According to French law, informed consent from each patient and/or next of kin was not necessary because of the retrospective design Vol. 82 - No. 11 KERHUEL nor was the local ethical committee agreement. Data were anonymized before statistical analy‑ sis. Study population All adult patients admitted to the General Intensive Care Unit (ICU) or the Neurosur‑ gical ICU of a French university hospital be‑ tween 1 January 2009 and 31 December 2013, and subsequently developing clinical symp‑ toms of BD, were considered. Final inclusion in the study required at least one CTA acquisi‑ tion for BD confirmation during the ICU stay. Exclusion criteria were: a BD diagnosis prior admission to the ICU and substantial miss‑ ing data restricting statistical analysis. As the hospital is a referral center for organ procure‑ ment, some patients were referred from level II centers after BD was confirmed. ICU admis‑ sion policies in both units remained unchanged throughout the study period. BD diagnosis A nurse performed sequential clinical exam‑ inations at least every two hours (composed of the Glasgow Coma Scale, assessment of pupil diameter and their reactivity to light, urine out‑ put and urine density measurement) as part of our ICU clinical monitoring protocol. When a clinical modification was noticed, the intensive care physician was immediately called to per‑ form a full neurological examination to assess clinical BD. After the exclusion of confounding factors (hypothermia, mean arterial pressure less than 65 mmHg, residual sedation, electro‑ lyte or acid‑base disturbances), a clinical diag‑ nosis of BD was confirmed in patients with an identified critical brain injury compatible with a diagnosis of BD, a Glasgow Coma Scale of 3, and a cessation of all brainstem reflexes includ‑ ing spontaneous ventilation assessed by the Apnea Test (AT) according to practical guid‑ ance of the American Academy of Neurology for determination of BD.4 AT was performed as follows: a duration of 10‑15 minutes, preoxy‑ genation with 100% oxygen, initial equilibra‑ tion of arterial carbon dioxide, the absence of Minerva Anestesiologica 1181 KERHUEL OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD spontaneous breathing was confirmed by both a clinical examination and a partial pressure of carbon dioxide ≥ 60 mmHg.15 According to French legislation,2 either one CTA or two electroencephalograms (EEG) separated by a four‑hour interval are mandatory for BD confirmation before organ donation. Re‑ sidual opacified vessels on a CTA or residual electrical activity on an EEG precluded BD confirmation. In the case of an inconclusive an‑ cillary test, the French procedure requires the repetition of the EEG or CTA until BD confir‑ mation is reached. All CTAs were performed lo‑ cally, following the protocol recommended by the French Society of Neuroradiology.6 A pre‑ liminary unenhanced cerebral acquisition was used as a reference. Then, a contrast medium (Iomeprol; concentration: 400 mg/mL; volume: 90‑120 mL) was injected intravenously at a rate of 4 mL/s. A second acquisition was performed 60 seconds after the injection and was used to evaluate the contrast enhancement of the ves‑ sels. Local attending neuroradiologists inter‑ preted CTA images. Opacification of the super‑ ficial temporal arteries was assessed to confirm the correct injection of the contrast medium. Re‑ sidual vessel opacification was determined by a visual comparison between unenhanced and enhanced CT images. From 2009 to 2010, BD was confirmed on CTA when an absence of ves‑ sel contrast enhancement was noticed accord‑ ing to the seven‑point score outlined by Dupas et al.16 Since 2011, the analysis has been limited to the four‑point CTA score defined by Fram‑ pas et al.,7 as recommended by the most recent guidelines.6 CTAs showing a CCA according to the seven‑score or the four‑score scale were defined as “conclusive CTA”. CTAs showing a residual cerebral perfusion in at least one vessel were classified as “inconclusive CTA”. Patients were allocated into two subgroups depending on whether one CTA only was necessary to con‑ firm BD or at least one CTA was inconclusive during the BD confirmation procedure. Data collection Cases were identified through the Organs and Tissues Harvesting Department database. 1182 All data were obtained from medical records and patient charts. Baseline patient character‑ istics were collected, including demographics, causes for ICU admission, cause of death, and ICU length of stay (LoS). Variables recorded regarding BD diagnosis were: completeness of brainstem reflex assessment, AT achievement, and ancillary tests (e.g. EEG and/or CTA). We reviewed the time interval between the seda‑ tion withdrawal and the clinical BD diagnosis, the time interval between the clinical BD di‑ agnosis and the BD confirmation with an an‑ cillary test, and the time interval between the discontinuation of sedation and the final BD declaration. Statistical analysis The main outcome was the rate of con‑ clusive CTA over time (in other words, the probability to confirm BD with CTA). Un‑ fortunately, this probability has never been reported before, nor was the proportion of CTA to the total of ancillary tests. Therefore the needed sample size was impossible to de‑ termine. We assumed that a minimum of 100 confirmed BD cases using CTA would allow performing parametric statistical tests consid‑ ering the missing data expected to be around 5%. Data were analyzed using XLSTAT 2014.3.03 software and R software (R De‑ velopment Core Team 2008, Vienna, Austria. http://www.R‑project.org). Categorical vari‑ ables were reported as proportions and con‑ tinuous variables were reported as medians and interquartile ranges (IQR, 25‑75th percen‑ tile). Statistical analysis used the chi‑squared test or Fisher’s exact test for categorical vari‑ ables, Mann‑Whitney test for comparisons between two groups and Kruskal‑Wallis Rank Sum Test for comparison among three groups. A p‑value of 0.05 or less was considered sig‑ nificant. The Kaplan‑Meier estimator includ‑ ed all the performed CTAs in order measure the proportion of conclusive CTAs (Y‑axis) over time (X‑axis). The time of clinical BD diagnosis was considered as the origin of the X‑axis. Minerva AnestesiologicaNovember 2016 OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD Results KERHUEL BD diagnosis Study population During the study period, 104 out of 190 clin‑ ically brain‑dead patients were included (Fig‑ ure 1). The sex ratio M/F was 1.8 and the me‑ dian age was 55 years (41‑64). Demographic data are reported in Table I. The most common causes for ICU admission were brain trauma (36 cases, 35%), anoxic encephalopathy (22 cases, 21%) and aneurysmal subarachnoid hemorrhage (20 cases, 19%). The median ICU length of stay was 3 days (2‑6). The median time interval from sedation withdrawal to clinical BD diagnosis was 9 hours (6‑13). A complete examination of brain‑ stem reflexes was not possible in nine cases (9%). AT was contra‑indicated or prematurely stopped in 46 cases (44%) owing to thoracic trauma, ventilator acquired pneumonia and/or pre‑existing medical condition such as chronic obstructive lung disease or respiratory failure. CTA or EEG confirmed BD in 102 patients. CTA accounted for 90% of final BD diagnosis (94 patients). When a first CTA did not confirm Figure 1.—The diagram shows different brain death confirmation strategies. BD: Brain death; CTA: computed tomography angiography; EEG: electroencephalogram; ICU: Intensive Care Unit. Vol. 82 - No. 11 Minerva Anestesiologica 1183 KERHUEL OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD Table I.—Demographic data. Table II.—Characteristics of brain death diagnosis. Variables All patients (N.=104) Age (years) Sex ratio (male/female) Reasons for ICU admission –– Stroke –– Cardiac arrest –– Household accident –– Road accident –– Suicide attempt –– Other Causes of death –– Traumatic brain injury –– Anoxic encephalopathy –– Aneurysmal subarachnoid haemorrhage –– Haemorrhagic stroke –– Ischaemic stroke –– Brain infection Length of stay in the ICU (days) 55 [41‑64] 1.8 (67/37) 42 (40) 19 (18) 16 (15) 15 (14) 9 (9) 3 (3) 36 (35) 22 (21) 20 (19) 13 (12) 11 (11) 2 (2) 3 [2‑6] Data are reported as No (%) or median [IQR]. No: number; IQR: interquartile range; BD: brain death; ICU: Intensive Care Unit. BD, a second CTA was performed in 2 cases out of 3, otherwise EEG was chosen (Figure 1 and Table II). A withdrawal of life support was decided in 2 patients before an ancillary test could confirm BD. CTA sensitivity was not significantly different before and after 2011 (80% and 81% respectively; P=0.96). Inconclusive CTA One hundred and seventeen CTAs were conducted on the 104 clinically brain‑dead patients. Ninety‑four CTAs (80%) were con‑ clusive for BD, 21 CTAs (18%) showed a per‑ sistent cerebral blood flow in at least one ves‑ sel (e.g. inconclusive CTA), and 2 CTAs (2%) were not interpretable because of artefacts. Two inconclusive CTAs were reported for the same patient. The main causes of inconclusive CTAs were residual opacification of at least one M4‑segment of the middle cerebral artery (47% of inconclusive CTAs) or of the internal cerebral vein (34% of inconclusive CTAs) or both (Figure 2). In 12 patients out of the 94 patients who un‑ derwent CTA for the final BD confirmation, it was not possible to found the exact hour of BD clinical diagnosis. So, only 82 patients were included for the analysis of CTA performances 1184 All patients (N.=104) Variables Clinical BD determination Time from sedation withdrawal to clinical BD diagnosis (hours) Complete clinical examination not being possible Apnoea test failure Final ancillary test for BD confirmation Time from clinical BD diagnosis to confir‑ mation with an ancillary test (hours) Ancillary tests –– CTA confirmed BD –– EEG confirmed BD –– Inconclusive Time from sedation withdrawal to final BD declaration (hours) 9 [6‑13] 9 (9) 46 (44) 6 [2‑11] 94 (90) 8 (8) 2 (2) 21 [12‑35] Data are reported as No (%) or median [IQR]. No: number; IQR: interquartile range; BD: brain death; CTA: computed tomography angiography; EEG: electroencephalography. Artifact, N.=2 6% Not available, N.=4 13% Internal cerebral vein, N.=11 34% Terminal segment of the middle cerebral artery, N.=15 47% Great cerebral vein, N.=0 0% Figure 2.—Frequency of opacification for each vessel in case of inconclusive computed tomography angiography (CTA). According to guidelines 7 a four‑vessel CTA interpretation model is used in France to confirm cerebral circulatory arrest. It relies on the lack of opacification of the M4 segment of both middle cerebral arteries that is to say the terminal segment for the cortex (M4‑MCA) and also the lack of opacification of the internal cerebral vein (ICV) and the great cerebral vein (GCV). This figure represents the reason why CTAs were con‑ sidered as inconclusive in 22 patients. Two inconclusive CTAs were reported for the same patient so the results of 23 CTAs are showed. Residual opacification of both the M4‑MCA and the ICV was found in 9 CTAs. Technical artifact precluded the diagnosis of brain death in 2 cases. NA: not available. to confirm BD according to the delay from clinical BD (Figures 3, 4). The time needed to achieve BD confirmation in 50% of the popula‑ Minerva AnestesiologicaNovember 2016 OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD Figure 3.—Survival curve describing the probability of con‑ clusive CTA over time. The survival function describes the probability for each case to reach a final state (e.g. cerebral circulatory arrest confirmed by CTA) from an initial state (e.g. clinical brain death). The X‑axis represents the time between clinical brain death diagnosis (whether an apneoa testing was performed or not) and conclusive CTA. Dotted lines represent the lower and upper limits of the 95 % confidence interval of the survival function. Full line represents the study population. Abbreviation: CTA: Computed Tomography Angiography. KERHUEL tion using CTA was 5.5 hours (4‑7.5). Six hours after clinical BD, the proportion of conclusive CTA was 51%. Moreover, 12 hours after BD clinical diagnosis, CTA were conclusive in 65 cases (79%) and this proportion reached 85% and 89% respectively after 20 and 23.5 hours. CTA sensitivity was significantly associated with the time lapse from clinical diagnosis to CTA examination (P=0.007) when dividing our cohort into three subgroups based on this time lapse (Table III). Univariate analysis showed that inconclu‑ sive CTAs were acquired significantly ear‑ lier than conclusive CTAs (2 hours (1‑3) vs. 4 hours (2‑9), P=0.008). The failure to complete full neurological examination (P=0.02) and decompressive craniectomy (P=0.05) were associated with the occurrence of at least one inconclusive CTA (Table IV). The time from sedation withdrawal to final BD confirmation was significantly longer in the subgroup with at least one inconclusive CTA than in the con‑ clusive CTA group (48 hours (34‑84) vs. 17 Figure 4.—Computed tomography angiography sensitivity over time for brain death confirmation. This figure is derived from the survival analysis and represents the cumulated sensitivity of computed tomography angiography (CTA) over time. The X‑axis represents the time (expressed in hours) between clinical brain death diagnosis and the first CTA (if more than one were performed) showing cerebral circulatory arrest. The Y‑axis represents CTA sensitivity (expressed in percents) mean‑ ing the proportion of conclusive CTAs to the total number of CTAs performed at each time points. The statistical analysis included data from the 82 patients with a precise time for brain death clinical diagnosis and who underwent CTA for the final brain death confirmation. Vol. 82 - No. 11 Minerva Anestesiologica 1185 KERHUEL OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD Table III.—Effect of the delay from brain death on computed tomography angiography performance to confirm brain death. Time lapse (h) Total number of CTA performed –– Conclusive CTA –– Inconclusive CTA Sensitivity (%) Group 1 0‑6 hrs Group 2 6‑12 hrs Group 3 After 12 hrs 49 42 7 86 25 23 2 92 19 17 2 89 The statistical analysis included data from the 82 patients with a precise time for brain death clinical diagnosis and who underwent CTA for the final brain death confirmation. The three groups are significantly different in terms of proportion of conclusive and inconclusive CTAs using Kruskal Wallis Sum Test (group 1 vs. group 2 vs. group 3, P=0.007). h: hour; CTA: computed tomography angiography. Table IV.—Bivariate analysis of the association of inconclusive CTA occurrence with demographics and brain death diagnosis characteristics. No inconclusive CTA during BD assessment (N.=82) Variables Demographics Male gender Age (years) Cause of death –– Brain trauma –– Brain anoxia –– Ruptured brain aneurysm –– Hemorrhagic stroke –– Ischemic stroke –– Brain infection BD diagnosis Not able to complete clinical examination Apnoea testing failure Decompressive Craniectomy Time from sedation withdrawal to clinical BD diagnosis (hours) Time from clinical BD diagnosis to first CTA acquisition (hours) 54 (66) 55 [42‑63] 1 or more inconclusive CTA during BD assessment (N.=22) 13 (57) 54 [41‑63] P value 0.62 0.83 31 (38) 17 (21) 14 (17) 10 (12) 9 (11) 1 (1) 5 (23) 5 (23) 6 (27) 3 (14) 2 (9) 1 (4) 0.56 4 (5) 38 (46) 6 (7) 9 [5‑14] 4 [2‑9] 5 (23) 8 (36) 5 (23) 9 [3‑22] 2 [1‑3] 0.02 0.47 0.05 0.89 0.008 Data are reported as No (%) or median [IQR]. No: number. IQR: interquartile range. BD: brain death. CTA: computed tomography angiog‑ raphy. hours (9‑28), respectively; P<0.001). The con‑ sent for organ donation of the patients’ rela‑ tives decreased from 55% to 45% if an incon‑ clusive CTA occurred during the diagnostic procedure without reaching statistical signifi‑ cance (P=0.43). Discussion The main result of this study is that the rec‑ ommended time lapse of 6 hours from clini‑ cal BD seems to be insufficient since the pro‑ portion of conclusive CTA is only 51%, with progressive increase overtime with more than 80% of conclusive CTA after 12 hours. Incon‑ clusive CTAs (e.g. persistent cerebral flow) are associated with a shorter time from clinical BD 1186 diagnosis. The duration of the whole BD diag‑ nostic procedure was almost three times longer when an inconclusive CTA occurred. These re‑ sults highlight the fact that performing a CTA very early after BD significantly increase the likelihood of having a inconclusive CTA and increase the total time needed to confirm BD, possibly affecting relatives compliance for or‑ gan donation and grafts quality.17 Previous observational studies have fo‑ cused on time interval between clinical ex‑ amination and EEG: the median duration of the BD procedure varied from 4 hours to 27 hours.18‑20 Part of this variability was linked to differences between the diagnostic procedures across countries. For example, in a recent Spanish cohort study of 289 patients, Fernan‑ Minerva AnestesiologicaNovember 2016 OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD dez et al. reported a 3.5% rate of inconclusive EEG, and a time to BD diagnosis that varied strongly among patients.19 Notably, according to the Spanish legislation, an ancillary test is optional and may be used to shorten the rec‑ ommended six‑hour period of clinical observa‑ tion. In a recent US multicenter retrospective study of 1311 patients, Lustbader et al. found that the median duration time for BD diagnosis was 19 hours.18 In the present study, the me‑ dian time from clinical BD diagnosis to CTA is relatively short (6 hours). BD confirmation was obtained for 47 patients (40%) in the first six hours. Five CTAs that were performed be‑ yond the six‑hour time were inconclusive, and only 2 CTAs were still inconclusive beyond a 12‑hour time point. In the case of a 12‑hour in‑ terval, the rate of inconclusive CTA decreased to 2% (2 CTAs out of 104 patients). Clinical experience shows that an intensiv‑ ist faced with an inconclusive CTA is used to postponing the additional ancillary test to the next day, in order not to accumulate inconclu‑ sive results; this strategy may partly explain the time extension. Interestingly, Orban et al. 21 have published a prospective controlled study showing that the transcranial Doppler (TCD) was useful for identifying the precise time of brain circulation cessation, thus decreasing the time to CTA acquisition with a low risk of in‑ conclusive results. Univariate analysis confirmed that a decom‑ pressive craniectomy, complete clinical exami‑ nation not being possible, and an early CTA acquisition were significantly associated with the occurrence of an inconclusive CTA. These factors had already been described in expert opinions,9 but to our knowledge this is the first quantitative analysis of risk factors for incon‑ clusive CTA ever reported. In our study, AT was contra‑indicated or prematurely stopped in 44% of cases, a proportion twice as high as that re‑ ported in the literature.22 Our recruitment of se‑ verely injured patients with a high prevalence of chest trauma or ventilator‑acquired pneumonia may partly explain this result. Cause of death was not a significant risk factor for inconclusive CTA, although a primary anoxic lesion associ‑ ated with brain perfusion has been previously Vol. 82 - No. 11 KERHUEL reported.23 However, this may be in relation with lack of power given the small number of patients in this subgroup of the present study. Limitations of the study The present study has several limitations. Firstly, the recruitment of the patients through the Organs and Tissues Harvesting Department database may have led to selection bias. It is possible that several patients presenting with clinical BD were not reported to the department when the intensivist considered the patient not eligible for organ donation. Secondly, 51 pa‑ tients whose BD was confirmed by EEG only were excluded from the study. It is possible that some of these patients had a risk factor for an inconclusive CTA, guiding the choice of the intensivist to EEG. This exclusion could have influenced demographic data and have under‑ estimated the proportion of inconclusive CTAs. Finally the replacement of the seven‑point score with a four‑point CTA score in 2011 according to the French guidelines 6 may have influenced the rate of inconclusive CTAs. However, the reported sensitivity was not significantly dif‑ ferent before and after 2011. The impact of the variability in interpretation of CTA by radiolo‑ gists is probably low, since a study published in 2009 by Sawicki et al. showed an 89% and 95% agreement in the inter‑radiological inter‑ pretation regarding the seven‑point score and the four‑point score, respectively.24 Conclusions The recommended six‑hour interval between clinical BD diagnosis and CTA is probably in‑ sufficient, as a 12‑hour interval might be more appropriate in order to limit the risk of incon‑ clusive CTA. This study reveals sources of in‑ conclusive CTA (decompressive craniectomy, incomplete neurological examination, CTA ac‑ quisition before 6 hours after clinical BD). The choice of an optimal timing should nevertheless also take into account the medical complications and the ethical issues that may occur in the case of an extended time interval. These results have to be confirmed in a larger prospective study af‑ Minerva Anestesiologica 1187 KERHUEL OPTIMAL TIMING FOR CTA IN THE CONFIRMATION OF BD ter exclusion of the cases for which brainstem reflexes cannot be completely assessed. Key messages —— The recommended time lapse of 6 hours between clinical brain death and computed tomography angiography is probably insufficient. —— The optimal time for CT angiog‑ raphy from clinical signs of brain death might be 12 hours. —— Decompressive craniectomy, incom‑ plete clinical brain stem assessment and the timing of computed tomography angiogra‑ phy are predictors of inconclusive CTAs (e.g. false negative CTAs). References 1. Citerio G, Crippa IA, Bronco A, Vargiolu A, Smith M. Variability in brain death determination in europe: look‑ ing for a solution. Neurocrit Care 2014;21:376‑82. 2. [No authors listed]. Décret relatif au constat de la mort préalable a prélèvement d’organes, de tissus et de cellules à des fins thérapeutiques ou scientifiques et modifiant le code de la santé publique (deuxième partie: Décrets en Conseil d’Etat). JORF n°282 du 4 décembre 1996; 1996. 3. Gardiner D, Shemie S, Manara A, Opdam H. Interna‑ tional perspective on the diagnosis of death. Br J Anaesth 2012;108 Suppl 1:i14‑28. 4. Wijdicks EF, Varelas PN, Gronseth GS, Greer DM, American Academy Of N. Evidence‑based guideline update: determining brain death in adults: report of the Quality Standards Subcommittee of the American Acad‑ emy of Neurology. Neurology 2010;74:1911‑8. 5. Wijdicks EF. The diagnosis of brain death. N Engl J Med 2001;344:1215‑21. 6. Societe Francaise De N, Societe Francaise De R, Agence De La B. [Recommendations on diagnostic criteria of brain death by the technique of CT angiography]. J Neu‑ roradiol 2011;38:36‑9. 7. Frampas E, Videcoq M, De Kerviler E, Ricolfi F, Kuoch V, Mourey F, et al. CT angiography for brain death diag‑ nosis. AJNR Am J Neuroradiol 2009;30:1566‑70. 8. Sawicki M, Bohatyrewicz R, Walecka A, Solek‑Pastusz‑ ka J, Rowinski O, Walecki J. CT Angiography in the Di‑ agnosis of Brain Death. Pol J Radiol 2014;79:417‑21. 9. Flowers WM, Jr., Patel BR. Persistence of cerebral blood flow after brain death. South Med J 2000;93:364‑70. 10. Rieke A, Regli B, Mattle HP, Brekenfeld C, Gralla J, Schroth G, et al. Computed tomography angiography (CTA) to prove circulatory arrest for the diagnosis of brain death in the context of organ transplantation. Swiss Med Wkly 2011;141:w13261. 11. Bohatyrewicz R, Sawicki M, Walecka A, Walecki J, Rowinski O, Bohatyrewicz A, et al. Computed tomo‑ graphic angiography and perfusion in the diagnosis of brain death. Transplant Proc 2010;42:3941‑6. 12. Escudero D, Otero J, Marques L, Parra D, Gonzalo JA, Albaiceta GM, et al. Diagnosing brain death by CT per‑ fusion and multislice CT angiography. Neurocrit Care 2009;11:261‑71. 13. Leclerc X, Taschner CA, Vidal A, Strecker G, Savage J, Gauvrit JY, et al. The role of spiral CT for the assessment of the intracranial circulation in suspected brain‑death. J Neuroradiol 2006;33:90‑5. 14. Welschehold S, Kerz T, Boor S, Reuland K, Thomke F, Reuland A, et al. Detection of intracranial circulatory ar‑ rest in brain death using cranial CT‑angiography. Eur J Neurol 2013;20:173‑9. 15. Société Française D’anesthésie Réanimation ADLB, So‑ ciété Française De Réanimation De Langue Française, Conférence D’experts. Prise en charge des sujets en état de mort encéphalique dans l’optique d’un prélèvement d’organes. 2003. 16. Dupas B, Gayet‑Delacroix M, Villers D, Antonioli D, Veccherini MF, Soulillou JP. Diagnosis of brain death using two‑phase spiral CT. AJNR Am J Neuroradiol 1998;19:641‑7. 17. Inaba K, Branco BC, Lam L, Salim A, Talving P, Plurad D, et al. Organ donation and time to procurement: late is not too late. J Trauma 2010;68:1362‑6. 18. Lustbader D, O’hara D, Wijdicks EF, Maclean L, Tajik W, Ying A, et al. Second brain death examination may nega‑ tively affect organ donation. Neurology 2011;76:119‑24. 19. Fernandez‑Torre JL, Hernandez‑Hernandez MA, Mu‑ noz‑Esteban C. Non confirmatory electroencephalogra‑ phy in patients meeting clinical criteria for brain death: scenario and impact on organ donation. Clin Neurophysi‑ ol 2013;124:2362‑7. 20. Vicenzini E, Pro S, Pulitano P, Rocco M, Spadetta G, Zarabla A, et al. Current practice of brain death deter‑ mination and use of confirmatory tests in an Italian Uni‑ versity hospital: a report of 66 cases. Minerva Anestesiol 2013;79:485‑91. 21. Orban JC, El‑Mahjoub A, Rami L, Jambou P, Ichai C. Transcranial Doppler shortens the time between clinical brain death and angiographic confirmation: a randomized trial. Transplantation 2012;94:585‑8. 22. Goudreau JL, Wijdicks EF, Emery SF. Complications during apnea testing in the determination of brain death: predisposing factors. Neurology 2000;55:1045‑8. 23. Ala TA, Kuhn MJ, Johnson AJ. A case meeting clini‑ cal brain death criteria with residual cerebral perfusion. AJNR Am J Neuroradiol 2006;27:1805‑6. 24. Sawicki M, Bohatyrewicz R, Safranow K, Walecka A, Walecki J, Rowinski O, et al. Computed tomographic an‑ giography criteria in the diagnosis of brain death‑compar‑ ison of sensitivity and interobserver reliability of differ‑ ent evaluation scales. Neuroradiology 2014;56:609‑20. Authors’ contributions.—Lionel Kerhuel, Mohamed Srairi, and Thomas Geeraerts designed the study. Lionel Kerhuel collected the data and performed the statistical analysis. All others participated in collecting the data. Lionel Kerhuel, Mohamed Srairi, and Thomas Geeraerts wrote the paper. All authors read and approved the final manuscript. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: September 13, 2016. - Manuscript accepted: September 9, 2016. - Manuscript revised: September 6, 2016. - Manuscript received: March 20, 2016. 1188 Minerva AnestesiologicaNovember 2016 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1189-98 ORIGINAL ARTICLE Serum S100β as a prognostic marker in patients with non-traumatic intracranial hemorrhage Eija K. JUNTTILA* 1, 2, 3, Juha KOSKENKARI 2, 3, Pasi P. OHTONEN 4, Ari KARTTUNEN 5, Tero I. ALA-KOKKO 2, 3 1Department of Anesthesiology, Tampere University Hospital, Tampere, Finland; 2Division of Intensive Care, Department of Anesthesiology, Oulu University Hospital, Medical Research Center Oulu, Oulu, Finland; 3Research Unit of Surgery, Anesthesiology and Intensive Care, Oulu University, Oulu, Finland; 4Department of Anesthesiology and Surgery, Oulu University Hospital, Oulu, Finland; 5Department of Radiology, Oulu University Hospital, Oulu, Finland *Corresponding author: Eija Junttila, Department of Anesthesiology, Tampere University Hospital, PO BOX 2000, 33521 Tampere, Finland. E-mail: [email protected] ABSTRACT BACKGROUND: The serum concentration of S100β protein reportedly predicts outcomes after brain injury. We examined the prognostic accuracy of S100β in patients with non-traumatic intracranial hemorrhage. METHODS: This was a prospective, observational study of patients with non-traumatic intracranial hemorrhage treated in the intensive care unit at our university hospital. Computed tomography imaging findings and the level of consciousness on admission were recorded. Serum S100β concentration was measured serially during the first six days of admission. Patients with subarachnoid hemorrhage (SAH group) or intracerebral hemorrhage (ICH group) were analyzed separately. The 3-month and 1-year functional outcomes were assessed using the Glasgow Outcome Scale (GOS). RESULTS: Of 108 patients enrolled, 66 were included in the SAH group and 42 in the ICH group. High initial S100β concentration was associated with Glasgow Coma Score 3-6 on admission (SAH group 0.61 μg/L versus 0.15 μg/L, P=0.001 and ICH group 1.00 μg/L versus 0.42 μg/L, P=0.005). Initial S100β concentration correlated with ICH volume (rho=0.50, P<0.001) and IVH Sum Score (rho=0.30, P=0.013). The thresholds for the initial S100β concentration with 100% specificity for poor outcome (GOS 1-3) were 1.40 μg/L for SAH and 1.76 μg/L for ICH group. ORs varied between 3.1 and 6.1 for S100β on poor outcome in the SAH group. Increasing S100β level during study period was associated with poor outcome in the SAH group. CONCLUSIONS: Serum S100β concentration corresponds with the severity of neurological insult and predicts poor outcome in patients with non-traumatic intracranial hemorrhage. (Cite this article as: Junttila EK, Koskenkari J, Ohtonen PP, Karttunen A, Ala-Kokko TI. Serum S100β as a prognostic marker in patients with non-traumatic intracranial hemorrhage. Minerva Amestesiol 2016;82:1189-98) Key words: Glasgow Outcome Scale - Intracranial hemorrhages - S100 Calcium Binding Protein beta Subunit. N on-traumatic intracranial hemorrhage has high mortality and morbidity.1 The level of consciousness and the computed tomography (CT) imaging findings in the acute phase are strong predictors of outcome.2-5 Predicting outcome during intensive care unit (ICU) stay remains challenging. One prognostic marker of interest has been S100β protein, with the highest concentrations in astrocytes, oligodendrocytes and Schwann Vol. 82 - No. 11 cells.6 Necrosis of these brain parenchymal cells releases S100β into the extracellular fluid, which can pass through the disrupted bloodbrain barrier to the systemic circulation.6 Although the association between elevated serum S100β concentration and worse outcome has been reported in patients with different types of intracranial hemorrhage,7-9 few have considered S100β as a long-term predictor of outcome. In patients with SAH there have been Minerva Anestesiologica 1189 JUNTTILA SERUM S100β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE only few studies that have examined 1-year functional outcome as a primary endpoint 8, 10 and in patients with ICH there has been none. The aims of this study were to examine the association between serum S100β concentration and the level of consciousness and CT imaging findings on admission, and to evaluate the predictive value of serum S100β concentration for outcome 3 months and 1 year after in patients with non-traumatic intracranial hemorrhage. Materials and methods Patients and clinical management This was a subsidiary study using data collected from a prospectively enrolled cohort of consecutive patients admitted with non-traumatic intracranial hemorrhage to the tertiary level ICU of our institution between December 2007 and December 2009, the details of which are described in more detail elsewhere.11, 12 This study was approved by the Ethics Committee of the Oulu University Hospital. Informed consent was obtained from the patient or a legal surrogate in all cases. Patient characteristics including age, sex and medical history were recorded. The level of consciousness on hospital admission (or before sedation, if earlier) was assessed using the Glasgow Coma Scale (GCS) 13 and categorized into one of four groups: GCS 15; 13-14; 7-12 and 3-6. The data during the ICU stay was prospectively collected into an electronic database (Critical Care Information Management System, CCIMS, Clinisoft, GE, Helsinki, Finland). The diagnosis of intracranial hemorrhage was established by CT imaging of the head on admission. Images were retrospectively reviewed by a neuroradiologist (AK); SAH and IVH were judged against the Hijdra SAH CT score (SAH Sum Score 0-30 and IVH Sum Score 0-12) 14 and ICH volume (mL) was calculated ([d1×d2×d3]/2, [three perpendicular diameters, d]). The presence or absence of diffuse cerebral edema, acute hydrocephalus and midline shift were also noted. The etiology 1190 of the hemorrhage was recorded based on the data from head CT scans, CT-angiographies, digital subtraction angiographies, operation and autopsy reports. Based on radiological findings patients were divided into two groups: the SAH group included patients with aneurysmatic SAH and/or IVH/ICH or perimesencephalic SAH, while the ICH group was comprised of patients with primary and secondary ICH and/or IVH, which may have enlarged to the subarachnoid space. Criteria for ICU admission were the need for neurosurgical or endovascular intervention and/or derangement of physiologic functions in patients judged by the treating physician to have a credible chance of survival. During the ICU stay, all patients were treated according to our normal clinical practice, consistent with latest guidelines 15-17 and as previously described.11, 12 Blood samples for determination of serum S100β concentration Each patient’s participation in the study commenced on the day of ICU admission (day 0) and lasted for a further 5 days (days 1-5), unless the patient died or was transferred to another hospital. Study period was divided into three 2-day sections: days 0-1, days 2-3 and days 4-5. The first arterial blood sample for serum protein S100β analysis was taken after a consent was obtained shortly after recruitment on day 0 or 1, and thereafter once in every 2-day period. Serum protein S100β specimen collection, preparation and analysis were carried out according to the manufacturer’s instructions. Blood for serum specimen was collected using standard serum sampling tubes and preserved one hour in room temperature before 10 minutes centrifuging to separate blood cells and serum. S100β was measured using an immunochemiluminometric method (Elecsys 2010, Roche Diagnostics, Roche Diagnostics GmbH, Mannheim, Germany). The initial (on day 0 or 1), and the maximum (between days 0 and 5) S100β concentrations were used for analysis. In patients with two to three S100β values trends in blood concentra- Minerva Anestesiologica November 2016 SERUM S100Β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE JUNTTILA tions were categorized as 1) increasing or 2) decreasing or unchanged. Outcomes The 3-month and 1-year outcome was assessed using the Glasgow Outcome Scale (GOS). (18) Data were collected by means of a telephone interview by one of the authors (EJ) or a trained research assistant (MV) with either the patient or carer. Outcome was categorized as good (GOS 4–5: no disability to moderate disability) or poor (GOS 1–3: severe disability, vegetative state, death). Statistical analysis SPSS (IBM Corp. Released 2012. IBM SPSS Statistics for Windows, Version 21.0. Armonk, NY: IBM Corp.) software was used for statistical analysis. Summary measurements are expressed as a mean with standard deviation (SD) or as a median with 25th–75th percentile, unless otherwise stated. Continuous variables were analyzed using Student’s t-test or the Mann-Whitney U-test, the latter for non-normally distributed data. The χ² test or Fisher’s exact test were used for categorical variables. Spearman’s correlation coefficient (rho) was used to assess correlations between continuous variables. The area under the receiver operating characteristic (ROC) curve (AUC) was used as a measure of the diagnostic accuracy of S100β in predicting poor outcome. The sensitivity and specificity were calculated with exact 95% binominal confidence intervals. The lowest S100β value giving 100% specificity was chosen as a threshold value with the idea that with a highly specific test a positive test result one could be almost certain of the presence of the outcome, ie. GOS 1-3 in our study. A multivariable logistic regression model was built for SAH group using GOS 1-3 at 3-month as outcome variable. A maximum of two adjusting covariates was used due to small data (SAH group patients with GOS 1-3, N.=31) to assess the impact of S100β value on GOS, thus the variable S100β was forced into Vol. 82 - No. 11 each model. The adjusting covariates used in logistic regression model were age, sex, aneurysmatic bleeding, comorbidity, diffuse brain edema, ���������������������������������� ventriculostomy, neurogenic pulmonary edema, ischemic electrocardiography abnormalities, craniotomy, SAH sum score, IVH sum score and ICH volume. Since the linearity assumption of S100β did not hold, the S100β was categorized into two classes according to median value (≤0.225 μg/L versus >0.225 μg/L). Of the adjusting factors age was used as continuous variable and all other variables as dichotomous variables. Adjusting factors were chosen according to clinical interest or having P value <0.2 in univariable model. The results of these models are presented as minimum and maximum ORs. Multivariable logistic regression model was no created for ICH group patients due the small number of GOS 4-5 in this group (N.=7). Two-tailed P values were reported and P<0.05 was considered statistically significant. Results During the 2-year study period, 191 patients with non-traumatic intracranial hemorrhage were treated in our ICU, 108 of whom were included in our analysis. The detailed flowchart of this study is presented in Figure 1. The main reasons for exclusion were: unavailability of study personnel in 19 cases, treatment withdrawal in 17 cases, admission delay >48 hours in 14 cases and other reasons in 24 cases. Sixty-six patients were included to the SAH group and 42 to the ICH group. Patient characteristics, primary head CT findings and the neurosurgical interventions undertaken are presented in Table I, and in our previous study.12 Sixty-three patients had aneurysmal hemorrhage of which 38 (60%) underwent coiling. With the exception of more impaired level of consciousness on admission in ICH group patients, there were no significant differences in the clinical characteristics between the groups. Intraventricular hemorrhage and hydrocephalus were common in both groups. Diffuse cerebral edema was more common in SAH group and midline shift in Minerva Anestesiologica 1191 JUNTTILA SERUM S100β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE ICU admission for non-traumatic intracranial hemorrhage N.=191 Excluded, N.=81 age <18 y, N.=2 admission delay >48 h, N.=14 AVM bleeding, N.=2 decision to withdraw from the active treatment before recruitment, N.=17 decision to transfer to the ward before recruitment, N.=8 consent was not obtained, N.=7 study personnel absent, N.=19 other reason, N.=12 Eligible for the study N.=110 Excluded after review N.=2 AVM bleeding, N.=1 tumor bleeding, N.=1 Analyzed N.=108 SAH group N.=66 Follow-up interview N.=54 ICH group N.=42 Died N.=12 Follow-up interview N.=30 Died N.=12 Figure 1.—Flowchart, until 1-year follow-up. ICH group. Intracranial pressure (ICP) monitoring and ventriculostomy were often required in both patient groups, but the need for craniotomy was more frequent in ICH group patients. 1192 The 3-month and 1-year GOS are presented in Table II. Mortality rates were 15% and 18% in the SAH group and 26% and 29% in the ICH group, respectively. Functional outcome data were available for all 84 patients alive 1 year Minerva Anestesiologica November 2016 SERUM S100Β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE JUNTTILA Table I.—Clinical characteristics, initial imaging findings and neurosurgical interventions undertaken according to the study group. Clinical characteristics Age (Y), mean (range) Sex, male, N. (%) Co-morbidities, N. (%) The level of consciousness on admission, N. (%) GCS 15 GCS 13-14 GCS 7-12 GCS 3-6 APACHE II, mean (SD) Primary head CT findings Presence of SAH, N. (%) SAH Sum Score, med (25th-75th) Presence of IVH, N. (%) IVH Sum Score, med (25th-75th) Presence of ICH, N. (%) ICH volume, med (25th-75th) Hydrocephalus, N. (%) Diffuse brain edema, N. (%) Midline shift, N. (%) Performed neurosurgical interventions ICP monitoring, N. (%) Ventriculostomy, N. (%) Craniotomy*, N. (%) SAH group N.=66 ICH group N.=42 57 (25-80) 34 (52) 40 (61) 57 (28-75) 22 (52) 26 (62) 19 (29) 21 (32) 8 (12) 18 (27) 19.5 (6.6) 62 (94) 17 (10-22) 41 (62) 5 (2-6.5) 22 (33) 29 (5-46.25) 34 (52) 31 (47) 18 (27) 46 (70) 42 (64) 24 (36) 2 (5) 10 (24) 13 (31) 17 (41) 20.9 (6.2) 6 (14) 4.5 (1.75-11.75) 27 (64) 10 (2-10) 40 (95) 46.5 (17.75-82.5) 21 (50) 8 (19) 26 (62) 31 (74) 25 (60) 27 (64) SAH: subarachnoid hemorrhage; IVH: intraventricular hemorrhage; ICH: intracerebral hemorrhage; GCS: Glasgow Coma Scale; APACHE: Acute Physiology And Chronic Health Evaluation; ICP: intracranial pressure. *Craniotomy for aneurysm clipping and/or hematoma evacuation. Table II.—Functional outcome at 3 months and 1 year assessed by the Glasgow Outcome Scale according to the study group. 3-month GOS Good, N. (%) 5 (good recovery) 4 (moderate disability) Poor, N. (%) 3 (severe disability) 2 (vegetative state) 1 (dead) 1-year GOS, N. (%) Good, N. (%) 5 (good recovery) 4 (moderate disability) Poor, N. (%) 3 (severe disability) 2 (vegetative state) 1 (dead) SAH group N.=66 ICH group N.=42 35 (53) 22 13 31 (47) 20 1 10 7 (17) 2 5 35 (83) 23 1 11 39 (59) 27 12 27 (41) 14 1 12 13 (31) 6 7 29 (69) 17 0 12 GOS: Glasgow Outcome Scale; SAH: subarachnoid hemorrhage; IVH: intraventricular hemorrhage; ICH: intracerebral hemorrhage. Vol. 82 - No. 11 after the insult: outcomes were less favorable in the ICH group at both 3 months and 1 year. Between 3 months and 1 year, four patients in the SAH group and six in the ICH group recovered sufficiently for their GOS category to improve from poor to good. High initial S100β concentration was associated with GCS 3-6 on admission (0.61 [0.18-1.55] μg/L versus 0.15 [0.08-0.25] μg/L, P=0.001 in SAH group patients with and 1.00 [0.47-2.34] μg/L versus 0.42 [0.20-0.87] μg/L, P=0.005 in ICH group patients). There was a significant correlation between initial S100β concentration and ICH volume (rho=0.50, P<0.001), as well as IVH Sum Score (rho = 0.30, P=0.013). In contrast, there was no correlation between initial S100β concentration and SAH Sum Score (rho=0.17, P=0.17). Initial S100β concentration was higher in patients with midline shift (0.78 [0.29-1.68] μg/L ver- Minerva Anestesiologica 1193 JUNTTILA SERUM S100β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE sus 0.17 [0.09-0.44] μg/L, P<0.001), but not in patients with diffuse brain edema or hydrocephalus (0.31 [0.15-1.11] μg/L versus 0.27 [0.12-0.82] μg/L, P=0.60 and 0.38 [0.16-1.01] μg/L versus 0.23 [0.11-0.66] μg/L, P=0.061, respectively). The median delay from the onset of symptoms to the ICU admission was 7 (5-11) hours. It did not correlate with initial S100β values (Spearman´s correlation coefficient rho=0.017). Maximum S100β concentration was higher in patients with poor 1 year functional outcome compared with patients with good outcome in both SAH group (0.68 [0.20-2.22] versus 0.15 SAH group, 3-month [0.09-0.28], P<0.001) and ICH group (0.95 [0.69-1.98] versus 0.49 [0.24-0.81], P=0.002). The statistical powers of these results were 84.4% and 89.1%, respectively. The data with two to three S100β measurements was available in 57 SAH group and in 38 ICH group patients. Increasing trend in S100β concentration levels were more frequent in patients with poor outcome (SAH group, 16/24 versus 6/33, P<0.001 and ICH group 10/11 versus 16/27, P=0.059). Receiver operator characteristic curves for initial and maximum S100β concentration are shown in Figure 2. Maximum S100β concentration produced the best AUC ICH group, 3-month ROC Curve ROC Curve S100B max d 0-1 S100B max d 0-5 Sensitivity Sensitivity S100B max d 0-1 S100B max d 0-5 1-Specificity 1-Specificity SAH group, 1-year ICH group, 1-year ROC Curve ROC Curve S100B max d 0-1 S100B max d 0-5 Sensitivity Sensitivity S100B max d 0-1 S100B max d 0-5 1-Specificity 1-Specificity Figure 2.—Receiver operating characteristic curves of serum S100β concentration on initial (days 0 to 1) and maximum serum S100β concentration between days 0 and 5 predictive of poor outcome at 3-month and 1-year according to the type of intracranial hemorrhage. 1194 Minerva Anestesiologica November 2016 SERUM S100Β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE JUNTTILA Table III.—Prognostic accuracy according to the area under the curve of receiver operator characteristic curves in Figure 2, and thresholds for serum S100β concentration with 100% specificity and the corresponding sensitivity. Cox-Snell R2 (R2) values are given to assess the goodness of fit of the models. SAH group N.=66 3-month* S100β days 0-1, μg/L* days 0-5, max, μg/L 1-year S100β days 0-1, μg/L* days 0-5, max, μg/L ICH group N.=42 AUC 95% CI Threshold with 100% specificity Sensitivity R2 AUC 95% CI Threshold with 100% specificity Sensitivity R2 0.80 0.81 0.69-0.91 0.71-0.92 1.40 1.47 0.21 0.29 0.28 0.28 0.75 0.74 0.53-0.98 0.50-0.97 1.76 1.76 0.21 0.23 0.07 0.09 0.74 0.77 0.62-0.83 0.66-0.89 1.40 1.47 0.24 0.33 0.17 0.26 0.71 0.80 0.54-0.88 0.64-0.95 1.76 1.76 0.24 0.28 0.14 0.22 SAH: subarachnoid hemorrhage; IVH: intraventricular hemorrhage; ICH: intracerebral hemorrhage; AUC: area under the curve; CI: confidence interval. *N.=62 in SAH group. in SAH group patients at 3 months and for ICH group patients at 1 year (Table III). The threshold values for initial and maximum S100β concentrations with 100% specificity for unfavorable outcome at 3 months and 1 year were identical (Table III). According to adjusted logistic regression models the OR for the impact of S100β on poor outcome in SAH group at three months varied between 3.1 and 6.1 reaching statistical significance at 5% level in all tested models, except the one in which ICH volume and ventriculostomy were adjusting factors. In that model the OR for S100β was 3.1 (95% CI 0.87-11.1, P=0.082). Discussion We found that serum protein S100β concentration predicted poor outcome in both SAH and ICH groups. Cut-off values with 100% specificity for poor outcome at both 3 months and 1 year were 1.40 μg/L for SAH group and 1.76 μg/L for ICH group. Patients with poor outcome had more frequently increasing S100β values during the study period. Higher initial serum S100β concentration was associated with diminished conscious level on admission in both groups, and although serum S100β concentration correlated with ICH volume and IVH Sum Score, there was no correlation with the SAH Sum Score. Vol. 82 - No. 11 Higher serum S100β concentration was associated with poor outcome in both groups of patients at both follow-up time points, a finding that is consistent with studies on SAH patients,8, 9, 19 although in two previous studies outcome was only assessed after 6 months.9, 19 Delgado et al.7 reported a clear association between elevated S100β concentration and poor outcome 3 months after ICH, but their study population was substantially less neurologically impaired on admission than our cohort, and those who subsequently underwent surgical intervention were excluded from their analysis. Notably, 20% (6 out of 30) patients in our study with high-grade ICH recovered sufficiently well to improve from the poor to good outcome category between 3 months and 1 year, suggesting that longer-term assessments of recovery of function may better reflect outcome in these patient populations. In this study patients with increasing trend in S100β levels had more frequently poor outcome. In SAH group the result was clear and consistent with previous reports 8 despite the short sample collection time in our study. In ICH group it did not reach statistical significance, which might be explained just by small study group. Our study is the first to report thresholds for initial and maximum S100β concentrations giving 100% specificity for poor outcome at both 3 months and 1 year in these patient Minerva Anestesiologica 1195 JUNTTILA SERUM S100β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE populations. It is striking that these thresholds were identical for outcomes at both evaluation times, supporting the validity of our results and the usefulness of S100β as a long-term prognostic indicator. In patients with traumatic brain injury, threshold values for serum S100β have been shown to vary widely and are markedly higher for unfavorable neurological prognosis (GOS 1-3) than in this study.20 Potential explanations include more extensive and widespread central nervous cell injury or extracerebral release of S100β in patients with multiple injuries. However, our AUC analyses yielded plausible values for our patient population, and are in concordance with previous studies.8, 21 The level of consciousness and the computed tomography (CT) imaging findings in the acute phase are strong predictors of outcome in these patient populations.2-5 In agreement with previous reports,9, 21, 22 we also found that the initial low GCS scores were associated with elevated S100β concentration in both SAH and ICH groups. Previous investigators have used the Fisher Scale to report the radiological abnormalities in SAH,8, 9, 19, 22, 23 two of which identified an association between elevated protein S100β concentration and high Fisher grade.9, 22 We elected to use the Hijdra SAH CT score instead of the Fisher grading scale as it more precisely defines the site and amount of blood. Interestingly we did not find a correlation between S100β concentration and the amount of subarachnoid blood defined by the SAH Sum Score. This contrast between our findings and those of other investigators can likely be explained by the presence of IVH in patients with Fisher grade 4 hemorrhage. This conclusion is supported by reports that the presence of IVH increases S100β concentration in patients with ICH 7 and that IVH is associated with worse outcome in both SAH and ICH;4, 24 in our study there was a significant correlation between S100β concentration and IVH Sum Score. The correlation between S100β concentration and ICH volume that we identified concurs with previous studies and can likely be explained by the larger parenchymal injury.7, 21 1196 Limitations of the study Our study had some limitations. First, there is wide variation in delays between the onset of neurological symptoms and blood sampling for measurement of initial serum S100β concentration; in addition the delays caused by the patients and health care system and difficult to control, we were not able to collect research blood samples without first gaining the consent of the patient or the assent of a relative. However, the delay to sampling did not correlate with S100β levels. Second, S100β was not measured daily. All these may affect to the results and peak S100β values may be missed. Third, only critically ill patients with severe ICH requiring neurosurgical interventions were included in the study, and care must be exercised when extrapolating our findings to populations with less severe ICH. Instead, nearly all patients with aneurysmatic SAH arrive to the ICU prior the aneurysm securing. Fourth, the frequency of craniotomies in ICH group patients was almost double that of those in SAH group patients in this study, which must take into account when interpreting our findings. The difference is explainable by the ICU admission criteria described above and the priority of endovascular intervention for aneurysm securing in our hospital. Entering ICH volume and ventriculostomy into the multivariate model the OR for S100β loses statistical significance. This is explained by the small samples size as can been seen from the wide confidence interval (0.87-11.1). Finally, the period over which serum S100β concentration was measured may have been too short to account for the influence of vasospasm on outcome in patients with aneurysmal bleeding. Conclusions We found that serum protein S100β concentration predicted poor long-term functional outcome in both SAH and ICH group patients, and corresponded with the severity of neurological insult assessed by the level of consciousness and imaging findings on admission. These re- Minerva Anestesiologica November 2016 SERUM S100Β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE JUNTTILA sults suggest that prospective trials looking more precisely at the relationship between protein S100β and long-term neurologic outcome in hemorrhagic stroke would be useful. 8. 9. Key messages —— Outcome prediction during intensive care unit stay remains challenging in patients with non-traumatic intracranial hemorrhage and consequently there has been a great interest in finding a laboratory test for it. —— Studies evaluating the usability of blood S100β protein concentration as a prognostic marker have promising results, but the data considered it as a long-term predictor of outcome with these patients is scarce. —— In this study S100β concentration predicted poor long-term functional outcome and corresponded with the severity of neurological insult in this patient population. 10. 11. 12. 13. 14. 15. 16. References 1. Fernandes HM, Mendelow AD. Non-traumatic intracranial hemorrhage. In: Webb A, Shapiro MJ, Singer M, Suter PM, editors. Oxford textbook of critical care. New York: Oxford University Press; 1999. p. 464-73. 2. Christensen MC, Broderick J, Vincent C, Morris S, Steiner T. Global differences in patient characteristics, case management and outcomes in intracerebral hemorrhage: the Factor Seven for Acute Hemorrhagic Stroke (FAST) trial. Cerebrovasc Dis 2009;28:55-64. 3. Navarrete-Navarro P, Rivera-Fernandez R, Lopez-Mutuberria MT, Galindo I, Murillo F, Dominguez JM, et al. Outcome prediction in terms of functional disability and mortality at 1 year among ICU-admitted severe stroke patients: a prospective epidemiological study in the south of the European Union (Evascan Project, Andalusia, Spain). Intensive Care Med 2003;29:1237-44. 4. Rosengart AJ, Schultheiss KE, Tolentino J, Macdonald RL. Prognostic factors for outcome in patients with aneurysmal subarachnoid hemorrhage. Stroke 2007;38:2315-21. 5. Junttila EK, Koskenkari J, Romppainen N, Ohtonen PP, Karttunen A, Ala-Kokko TI. ���������������������������� Risk factors for 1-year mortality in patients with nontraumatic intracranial hemorrhage requiring intensive care. Acta Anaesthesiol Scand 2011;55:1052-60. 6. Brunkhorst R, Pfeilschifter W, Foerch C. Astroglial proteins as diagnostic markers of acute intracerebral hemorrhage-pathophysiological background and clinical findings. Transl Stroke Res 2010;1:246-51. 7. Delgado P, Alvarez Sabin J, Santamarina E, Molina CA, Vol. 82 - No. 11 17. 18. 19. 20. 21. 22. 23. Quintana M, Rosell A, et al. Plasma S100B level after acute spontaneous intracerebral hemorrhage. Stroke 2006;37:2837-9. Sanchez-Pena P, Pereira AR, Sourour NA, Biondi A, Lejean L, Colonne C, et al. S100B ���������������������������� as an additional prognostic marker in subarachnoid aneurysmal hemorrhage. Crit Care Med 2008;36:2267-73. Weiss N, Sanchez-Pena P, Roche S, Beaudeux JL, Colonne C, Coriat P, et al. Prognosis value of plasma S100B protein levels after subarachnoid aneurysmal hemorrhage. Anesthesiology 2006;104:658-66. Lai PM, Du R. Association between S100B Levels and Long-Term Outcome after Aneurysmal Subarachnoid Hemorrhage: Systematic Review and Pooled Analysis. PLoS One 2016;11:e0151853. Junttila E, Ala-Kokko T, Ohtonen P, Vaarala A, Karttunen A, Vuolteenaho O, et al. Neurogenic pulmonary edema in patients with nontraumatic intracerebral hemorrhage: predictors and association with outcome. Anesth Analg 2013;116:855-61. Junttila E, Vaara M, Koskenkari J, Ohtonen P, Karttunen A, Raatikainen P, et al. Repolarization abnormalities in patients with subarachnoid and intracerebral hemorrhage: predisposing factors and association with outcome. Anesth Analg 2013;116:190-7. Teasdale G, Jennett B. Assessment of coma and impaired consciousness. A practical scale. Lancet 1974;2:81-4. Hijdra A, Brouwers PJ, Vermeulen M, van Gijn J. Grading the amount of blood on computed tomograms after subarachnoid hemorrhage. Stroke 1990;21:1156-61. Connolly ES,Jr, Rabinstein AA, Carhuapoma JR, Derdeyn CP, Dion J, Higashida RT, et al. Guidelines for the management of aneurysmal subarachnoid hemorrhage: a guideline for healthcare professionals from the American Heart Association/american Stroke Association. Stroke 2012;43:1711-37. Diringer MN, Bleck TP, Claude Hemphill J,3rd, Menon D, Shutter L, Vespa P, et al. Critical care management of patients following aneurysmal subarachnoid hemorrhage: recommendations from the Neurocritical Care Society’s Multidisciplinary Consensus Conference. Neurocrit Care 2011;15:211-40. Morgenstern LB, Hemphill JC,3rd, Anderson C, Becker K, Broderick JP, Connolly ES,Jr, et al. Guidelines for the management of spontaneous intracerebral hemorrhage: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2010;41:2108-29. Jennett B, Bond M. Assessment of outcome after severe brain damage. Lancet 1975;1:480-4. Wiesmann M, Missler U, Hagenstrom H, Gottmann D. S-100 protein plasma levels after aneurysmal subarachnoid haemorrhage. Acta Neurochir (Wien) 1997;139:1155-60. Mercier E, Boutin A, Lauzier F, Fergusson DA, Simard JF, Zarychanski R, et al. Predictive value of S-100beta protein for prognosis in patients with moderate and severe traumatic brain injury: systematic review and metaanalysis. BMJ 2013;346:f1757. Hu YY, Dong XQ, Yu WH, Zhang ZY. Change in plasma S100B level after acute spontaneous basal ganglia hemorrhage. Shock 2010;33:134-40. Vos PE, van Gils M, Beems T, Zimmerman C, Verbeek MM. Increased GFAP and S100beta but not NSE serum levels after subarachnoid haemorrhage are associated with clinical severity. Eur J Neurol 2006;13:632-8. Moritz S, Warnat J, Bele S, Graf BM, Woertgen C. The prognostic value of NSE and S100B from serum and cerebrospinal fluid in patients with spontaneous subarachnoid hemorrhage. J Neurosurg Anesthesiol 2010;22:2131. Minerva Anestesiologica 1197 JUNTTILA SERUM S100β AS A PROGNOSTIC MARKER IN PATIENTS WITH NON-TRAUMATIC INTRACRANIAL HEMORRHAGE 24. Hallevi H, Albright KC, Aronowski J, Barreto AD, Martin-Schild S, Khaja AM, et al. ����������������������� Intraventricular hemor- rhage: Anatomic relationships and clinical implications. Neurology 2008;70:848-52. Authors’ contributions.—Eija K. Junttila, Juha Koskenkari, Tero I. Ala-Kokko designed the study, analyzed and interpreted the data, drafted and critically revised the manuscript. Ari Karttunen analyzed the CT findings, contributed to the interpretation of the data, provided critical revision of the article. Pasi �������������������������������������������������������������������������������������������������� P. Ohtonen����������������������������������������������������������������������������������� contributed substantially to the acquisition of the data, analysis and interpretation of the data, provided critical revision of the article. Funding.—This study was financially supported by a small project grant from the Oulu University Hospital EVO grant for Dr Junttila. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Acknowledgments.—The expert help of research assistant Maarika Vaara, MD, Elina Paloniemi, MD, and study nurse Sinikka Sälkiö in the collection of data, and Heikki Koskinen, MD in the figure editing was much appreciated. Article first published online: September 15, 2016. - Manuscript accepted: September 14, 2016. - Manuscript revised: September 13, 2016. - Manuscript received: February 1, 2016. 1198 Minerva Anestesiologica November 2016 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1199-213 REVIEW Perioperative hemodynamic goal-directed therapy and mortality: a systematic review and meta-analysis with meta-regression Mariateresa GIGLIO 1*, Fabio MANCA 2, Lidia DALFINO 1, Nicola BRIENZA 1 1Anaesthesia and Intensive Care Unit, Department of Emergency and Organ Transplantation, University of Bari, Policlinico, Bari, Italy; 2Department of Education, Psychology, Comunication studies, University of Bari, Bari, Italy *Corresponding author: Mariateresa Giglio, Anaesthesia and Intensive Care Unit, Department of Emergency and Organ Transplantation, University of Bari, Policlinico, Piazza G. Cesare 11, 70124 Bari, Italy. Email: [email protected] A B STRACT INTRODUCTION: Recent data found that perioperative goal directed therapy (GDT) was effective only in higher control mortality rates (>20%) with a relatively high heterogeneity that limited the strength of evidence. The aim of the present meta-analysis was to clearly understand which high risk patients may benefit of GDT. EVIDENCE ACQUISITION: Systematic review of randomized controlled trials with meta-analyses, including a metaregression technique. MEDLINE, EMBASE, and The Cochrane Library databases were searched (1980-January 2015). Trials enrolling adult surgical patients and comparing the effects of GDT versus standard hemodynamic therapy were considered. The primary outcome measure was mortality. Data synthesis was obtained by using Odds Ratio (OR) with 95% confidence interval (CI) by random-effects model. EVIDENCE SYNTHESIS: Fifty eight studies met the inclusion criteria (8171 participants). Pooled OR for mortality was 0.70 (95% CI 0.56-0.88, P=0.002, no statistical heterogeneity). GDT significantly reduced mortality when it is >10% in control group (OR 0.43, 95% CI 0.30-0.61, P<0.00001). The meta-regression model showed that the cut off of 10% of mortality rate in control group significantly differentiates 43 studies from the other 15, with a regression coefficient b of -0.033 and a P value of 0.0001. The significant effect of GDT was driven by high risk of bias studies (OR 0.48, 95% CI 0.34-0.67, P<0.0001). CONCLUSIONS: The present meta-analysis, adopting the meta-regression technique, suggests that GDT significantly reduces mortality even when the event control rate is >10%. (Cite this article as: Giglio M, Manca F, Dalfino L, Brienza N. Perioperative hemodynamic goal-directed therapy and mortality: a systematic review and meta-analysis with meta-regression. Minerva Anestesiol 2016;82:1199-213) Key words: Perioperative care - Mortality - Therapy. T Introduction he strategy of hemodynamic goal-directed therapy (GDT) refers to monitoring and manipulation of physiological hemodynamic parameters by means of therapeutic interventions,1 based mainly on fluids, red blood cells and inotropic drugs. This regimen was origiComment in p. 1135. Vol. 82 - No. 11 nally applied in surgical patients with the aim to reach normal or supranormal values of cardiac output and oxygen delivery 2 and later applied to critically ill patients. A first meta-analysis 3 including surgical and critically ill patients did not demonstrate any significant overall benefit. Some years later, a lower mortality was observed only in very severe surgical, trauma and medical patients in whom optimization treatment was performed before organ failure occurrence.4 A Minerva Anestesiologica 1199 GIGLIO GOAL DIRECTED THERAPY AND MORTALITY subsequent meta-analytic study,5 differentiating surgical patients from patients with sepsis and organ failure, showed that in septic patients no benefit was observed, although the matter is still under debate,6 while mortality was improved in perioperative subgroup, although the presence of statistical heterogeneity and inconsistency reduced the strength of evidence. The benefit was recently confirmed only in very high risk patients (control group mortality >20%) on the basis of a priori subgroup division.7, 8 In these papers, however, no clear explanation was provided about how this cut-off was obtained and a relatively high heterogeneity was still observed in all the analyses,7, 8 thus reducing the strength of evidence. We therefore conducted a meta-analysis with meta-regression to clearly evaluate which high risk patient can benefit from perioperative GDT. Information sources Evidence acquisition Eligibility criteria RCTs were selected according to the following inclusion criteria:9 —— types of participants. Adult patients (ages 18 years and older) undergoing major surgery were considered. Studies involving mixed populations of critically ill, nonsurgical patients, or postoperative patients with sepsis or organ failure were excluded; —— types of interventions. GDT was defined as monitoring and manipulation of hemodynamic parameters to reach normal or supranormal values by fluid infusion alone or in combination with inotropic therapy in the perioperative period within eight hours after surgery. Studies including late hemodynamic optimization treatment were excluded; —— types of comparisons. Trials comparing the beneficial and harmful effects of GDT versus standard hemodynamic therapy were considered. RCTs with no description or no difference in optimization strategies between groups, as well as RCTs in which therapy was titrated to the same goal in both groups or was not titrated to predefined end points were excluded; 1200 —— types of outcome measures. The primary outcome measure was mortality. For those RCTs providing more data on mortality (i.e. in-hospital, 30-day, 90-day), the inhospital mortality was considered. Sensitivity analysis was planned including only low risk of bias trials (see below). Moreover, another sub-group analysis was planned on the basis of the result of the meta-regression model (see below). A third subgroup analysis was planned combining the results of the previous two analyses (i.e. high mortality/high risk of bias, high mortality/low risk of bias, low mortality/high risk of bias, low mortality/low risk of bias); —— types of studies. RCTs on perioperative GDT in surgical patients were included. No language, publication date, or publication status restrictions were imposed. Different search strategies (last update January 2015) were performed to retrieve relevant randomized controlled trials (RCTs) by using MEDLINE, The Cochrane Library and EMBASE databases. No date restriction was applied for MEDLINE and The Cochrane Library databases, while the search was limited to 2008-2014 for EMBASE database.10 Additional RCTs were searched in The Cochrane Library and the Database of Abstracts of Reviews of Effects (DARE) databases and in the reference lists of previously published reviews and retrieved articles. Other data sources were hand-searched in the annual proceedings (2008-2014) of the Society of Critical Care Medicine, the European Society of Intensive Care Medicine, the Society of Cardiovascular Anesthesiologists, the Royal College of Anesthetists, the American Society of Anesthesiologists. In order to reduce publication bias, abstracts were searched.11 Publication language was not a search criterion. Search terms Trials selection was performed by using the following search terms: randomized con- Minerva Anestesiologica November 2016 GOAL DIRECTED THERAPY AND MORTALITYGIGLIO trolled trial, controlled clinical trial, surgery, goal directed, goal oriented, goal target, cardiac output, cardiac index, DO2, oxygen consumption, cardiac volume, stroke volume, fluid therapy, fluid loading, fluid administration, optimization, supranormal. The search strategies used for the MEDLINE, The Cochrane Library and EMBASE databases are reported in the Supplementary Appendix I, online content only. Study selection Two investigators (FM, NB) examined at first each title and abstract to exclude clearly irrelevant studies and to identify potentially relevant articles. Other two investigators (LD, MG) independently determined eligibility of full-text articles retrieved. The names of the author, institution, journal of publication and results were unknown to the two investigators at this time. Data abstraction and study characteristics Data were independently collected by two investigators (MG, NB), with any discrepancy resolved by re-inspection of the original article. To avoid transcription errors, the data were input into statistical software and rechecked by different investigators (LD, FM). RCT data gathered Data abstraction included surgical risk (defined by the authors on the basis of POSSUM score,12 ASA physical status classification, age >60 years, pre-operative morbidity, as previously adopted),13 mortality of control group, type of surgery (i.e., elective or emergent, abdominal, thoracic, vascular, etc.), anesthesiological management, hemodynamic goaldirected therapy (end-points, therapeutic intervention and monitoring tools). Risk of bias in individual studies A domain-based evaluation, as proposed by the Cochrane Collaboration, was used to eval- Vol. 82 - No. 11 uate the methodological quality of RCTs.14 This is a two-part tool, addressing seven specific domains (namely, sequence generation, allocation concealment, blinding of participants and personnel, blinding of outcome assessment, incomplete outcome data, selective outcome reporting) that are strongly associated with bias reduction.15, 16 Each domain in the tool includes one or more specific entries in a “Risk of bias” table. Within each entry, the first part of the tool describes what was reported to have happened in the study, in sufficient detail to support a judgement about the risk of bias. The second part of the tool assigns a judgement relating to the risk of bias for that entry. This is achieved by assigning a judgement of “Low risk”, “High risk”, or “Unclear risk” of bias. After each domain was completed, a “Risk of bias summary” figure presenting all of the judgements in a cross-tabulation of study by entry are generated. The green plus indicates low risk of bias, the red minus indicates high risk of bias, the white color indicates unclear risk of bias. For each study the number of green plus obtained for every domain was calculated: RCTs with 5 or 6 green plus were considered as having an overall low risk of bias. Summary measures and planned method of analysis Meta-analytic techniques (analysis software RevMan, version 5.3.5, Cochrane Collaboration, Oxford, England, UK) were used to combine studies using odds ratios (ORs) and 95% confidence intervals (CIs). A statistical difference between groups was considered to occur if the pooled 95% CI did not include 1 for the OR. An OR less than 1 favored GDT when compared with control group. Two-sided P values were calculated. A random-effects model was chosen for all analyses. Statistical heterogeneity and inconsistency were assessed by using the Q and I2 tests, respectively.17, 18 When the P value of the Q-Test was <0.10 17 and/or the I2 was >25%, heterogeneity and inconsistency were considered significant.19 Minerva Anestesiologica 1201 GIGLIO GOAL DIRECTED THERAPY AND MORTALITY Meta-regression. Assessing the impact of the slope In the present meta-analysis the chosen covariates were the mortality rate in control group, the risk of bias evaluation (considering how many green plus the study obtained) and the year of publication. The meta-regression model 20-22 was applied to all the included studies. The software used was Comprehensive Meta Analysis Version 3.0 Evidence synthesis Study selection Identification The search strategies identified 3244 (MEDLINE), 9948 (Cochrane Library) and 3054 (EMBASE) articles. Thirteen articles were identified through other sources (congress abstracts, ref- 16246 records identified through database searching erence lists). After initial screening and subsequent selection, a pool of 98 potentially relevant RCTs was identified. The subsequent eligibility process (Figure 1) excluded 40 articles and, therefore, 58 articles 23-80 with a total sample of 8171 patients, were considered for the analysis. Study characteristics All included articles evaluated the effects of hemodynamic optimization on mortality as primary or secondary outcome and had a population sample of adult surgical patients, undergoing both elective or emergent procedures (Supplementary Table I, online content only).23-80 The studies were performed in Australia, United States, Europe, Canada, Brazil, China, Israel and India from 1991 to 2014 (Supplementary Table I) and were all published in English. 13 additional records identified through other sources Screening 3516 records after duplicates removed 116 records excluded Eligibility 98 full-text articles assessed for eligibility Included 214 records screened 58 studies included in qualitative synthesis 40 full-text articles excluded: — 26: hemodynamic optimization titrated to the same end-point or not titrated to predefined end points, or no difference between groups in the optimization protocol; — 13: mixed population of critically ill, not surgical patients, with already established sepsis or organ failure and undergoing late optimization; — 1: only protocol 58 studies included in qualitative synthesis Figure 1.—Flow chart summarizing the studies selection procedure for the meta-analysis. 1202 Minerva Anestesiologica November 2016 GOAL DIRECTED THERAPY AND MORTALITYGIGLIO Data concerning RCTs morbidity/mortality risk definition, population and type of surgery are presented in Supplementary Table I. The risk of bias assessment for each trial is showed (in Supplementary Table II online content only). Figure 2.—Rates of mortality for each of the studies with Odds Ratios (ORs) and 95% Confidence Intervals (CI). The pooled OR and 95% CI are shown as the total. The size of the box at the point estimate of the OR gives a visual representation of the “weighting” of the study. The diamond represents the point estimate of the pooled OR and the length of the diamond is proportional to the CI. Vol. 82 - No. 11 Minerva Anestesiologica 1203 GIGLIO GOAL DIRECTED THERAPY AND MORTALITY Figure 3.—Rates of mortality with Odds Ratios (ORs) and 95% Confidence Intervals (CI) in subgroup according to risk of bias. Studies were divided in high risk of bias and low risk of bias according to the Cochrane domain-based evaluation (see text for details). The pooled OR and 95% CI are shown as the total. The size of the box at the point estimate of the OR gives a visual representation of the “weighting” of the study. The diamond represents the point estimate of the pooled OR and the length of the diamond is proportional to the CI. 1204 Minerva Anestesiologica November 2016 GOAL DIRECTED THERAPY AND MORTALITYGIGLIO Quantitative data synthesis In 58 RCTs, 482 patients died: 205 out of 4137 (5%) were randomized to perioperative goal-directed therapy, and 277 out of 4034 (7%) were randomized to control. Pooled OR for mortality was 0.70 and 95% CI was 0.560.88. No statistical heterogeneity and inconsistency were detected (Figure 2). Excluding the largest study,61 the result was confirmed: OR was 0.62 with 95% CI 0.48-0.80 (P=0.0002, 6177 pts), and no significant statistical heterogeneity (Q statistic P=0.56; I2=0%) was observed. The sensitivity analysis showed that the significant effect of GDT on mortality was driven by high risk of bias RCTs (OR 0.48, 95% CI 0.34-0.67, P<0.0001, Q statistic P=0.78; I2=0%, 32 RCTs), while no effect was demonstrated in low risk of bias trials (OR 0.94, 95% CI 0.73- 1.20, P=0.61, Q statistic P=0.57; I2=%, 26 RCTs) (Figure 3). Meta-regression Figure 4 showed the plot of log odds ratio on control group mortality: the meta-regression model identified, by inspection to the plot, a point (cut-off) on the upper confidence interval of the regression line that separates positive log odds ratios to negative log odds ratios: this cut-off coincides with the mortality rate in control group of 10%. In other words, the meta-regression model, applied to all 58 RCTs, showed that the cut off of 10% of mortality rate in control group significantly differentiated 43 studies from the other 15, with a regression coefficient b of -0.033 and a P value of 0.0001 (see Figure 4). Supplementary Table III (online content only), showed the results for meta-regression using control group mortality to predict the log odds ratio. The subgroup analysis including only studies in which the mortality rate in the control group was lower than 10% showed no significant results (OR 0.99, 95% CI 0.78-1.27, P=0.95, Q statistic P=0.97 I2=0%, 43 RCTs), while a statistical significant effect was observed in those RCTs with a mortality rate in Vol. 82 - No. 11 Figure 4.—Regression model applied to all 58 studies, basing on mortality rate in control group. The cut off of 10% of mortality rate in control group significantly differentiates 43 studies on the right side of the graph from 15 on the left side. The regression coefficient b=-0.033 with a P-value of 0.0001 confirms the analysis (see text for details). control group >10% (OR 0.43, 95% CI 0.300.61, P<0.00001, Q statistic P=0.41; I2=3%, 15 RCTs, (Figure 5). Figure 6 showed the plot of log odds ratio on risk of bias evaluation. The meta-regression model was applied to all 58 RCTs. The inspection to the plot showed that only studies with high risk of bias (<5 green plus obtained with the Cochrane domain-based evaluation for risk of bias) had a significant reduction in mortality rate, with a regression coefficient b of 0.225 and a P value<0.00001 (Figure 6), while no significant reduction was observed in low risk of bias RCTs. Supplementary Table IV (online content only) showed the results for meta-regression using risk of bias evaluation to predict the log odds ratio. Figure 7 showed the results of the combined 4 subgroups (i.e. mortality <10%/high risk of bias, mortality <10%/ low risk of bias, mortality >10%/high risk of bias, mortality >10%/low risk of bias): only the group with mortality rate >10% and high risk of bias reached statistical significance (OR 0.38, 95% CI 0.25-0.57, P<0.00001, Q statistic P=0.66; I2=0%, 9 RCTs). The group with mortality rate >10% and low risk of bias did not show any statistical significance (OR 0.53, 95% CI 0.25-1.13, P=0.10, Q statistic P=0.19; I2=33%, 6 RCTs). In order to look to the effect of time as a covariate, another meta-regression was per- Minerva Anestesiologica 1205 GIGLIO GOAL DIRECTED THERAPY AND MORTALITY Figure 5.—Rates of mortality with Odds Ratios (ORs) and 95% Confidence Intervals (CI) in subgroups defined according to the mortality rate in control group. RCTs were divided in studies with a control mortality rate < of 10% or > of 10%. The pooled OR and 95% CI are shown as the total. The size of the box at the point estimate of the OR gives a visual representation of the “weighting” of the study. The diamonds represent the point estimate of the pooled ORs and the length of the diamonds is proportional to the CI. 1206 Minerva Anestesiologica November 2016 GOAL DIRECTED THERAPY AND MORTALITYGIGLIO Figure 6.—Regression model applied to all 58 studies, basing on risk of bias evaluation (number of green plus obtained by each RCT according to the domain-based risk of bias evaluation, as proposed by the Cochrane Collaboration). The regression coefficient b=0.225 with a P-value of 0.0001 confirms the result of the sensitive analysis: the global effect of GDT on mortality was driven by high risk of bias studies, while the higher quality studies did not demonstrate any benefit in mortality reduction. (see text for details). formed, adopting the publication year as a covariate: this analysis (Figure 8) showed that there is no statistical dependence between the effect on mortality and the year of publication (regression coefficent 0.045). Discussion This systematic review and meta-analysis confirmed that perioperative GDT reduced mortality after surgery. This significant effect was maintained when the mortality rate in control group was >10% and when high risk of bias RCTs were considered. No significant effect was observed in low risk of bias trials. GDT has been originally applied in surgical patients in order to face the perioperative increase in oxygen demand and to prevent organ failure. When performed in patients with already established organ failure, no outcome improvement was found.5 These different results may rely on the basis that only in the early stage of systemic inflammatory response syndrome (i.e. in the early preoperative period) it is possible to prevent the deleterious effects of hypoperfusion and decreased oxygen delivery, while, when oxygen debt is no longer reversible, increasing oxygen transport is no more effective. Vol. 82 - No. 11 Expected mortality appears to be a very striking factor affecting effectiveness of GDT. Shoemaker et al.2 found that GDT was effective only when optimization treatment was performed in high risk medical, surgical and trauma patients (control group mortality >20%) before organ failure occurrence. Recent data 7, 8 have confirmed GDT benefits only in surgical patients, but have again limited its effectiveness only in patients with very high control mortality (i.e., >20%). This reported cut-off, however, may result from a priori classification and may not reflect the “real life” of every day practice.81, 82 The present meta-analysis, adopting the meta-regression technique, demonstrated that preoperative hemodynamic optimization significantly reduced mortality even when the event control rate is >10%. It should be underscored, however, that, in this meta-regression model, the relationship between effect estimates and the control group risk is complicated by a technical phenomenon known as regression to the mean. This arises because the control group risk forms an integral part of the effect estimate. A high risk in a control group, observed entirely by chance, will on average give rise to a higher than expected effect estimate, and vice versa. This phenomenon results in a false correlation between effect estimates and control group risks. This is the reason why we decided to perform also a subgroup analysis that is another way to investigate heterogeneous results or to answer specific questions about particular patient groups. The subgroup adopting the cut-off of mortality rate in control group >10% reinforced the meta-regression result. Moreover, despite the existence of methodological heterogeneity among studies dealing with GDT, such as timing, monitoring and protocols, a strong statistical homogeneity and consistency (even using conservative cut-off values) was still observed in the main as well as in the sensitive analyses, whereas moderate to high heterogeneity and inconsistency has limited precedent results.7 A reason for this discrepancy is that the present meta-analysis did not include two 83, 84 of the 32 studies of previous papers, because in these two papers dopex- Minerva Anestesiologica 1207 GIGLIO GOAL DIRECTED THERAPY AND MORTALITY Figure 7.—Rates of mortality with Odds Ratios (ORs) and 95% Confidence Intervals (CI) in subgroup according to the combination of mortality > or <10% and risk of bias. Four subgroup were obtained: mortality <10%/high risk of bias, mortality <10%/low risk of bias, mortality >10%/high risk of bias, mortality >10%/low risk of bias (see text for details). The pooled OR and 95% CI are shown as the total. The size of the box at the point estimate of the OR gives a visual representation of the “weighting” of the study. The diamond represents the point estimate of the pooled OR and the length of the diamond is proportional to the CI. 1208 Minerva Anestesiologica November 2016 GOAL DIRECTED THERAPY AND MORTALITYGIGLIO Figure 8.—Regression model applied to all 58 studies, basing on the publication year as a covariate: this analysis shows that there is no statistical dependence between the effect on mortality and the year of publication (regression coefficient 0.045). amine was used without predefined hemodynamic end points. Another paper 8 reviewed the use of GDT to reduce mortality, and again arbitrarily adopted a 20% cut-off of mortality rate in control group. Once again, heterogeneity reduced the strength of the evidence. One included paper 85 was not included in the present meta-analysis for methodological reason (both the control and the GDT group were treated with the same protocol). Moreover, the present meta-analysis was updated with new 19 studies published from 2010 to 2014. All these reasons may have reduced the variability in the outcome observed. The ineffectiveness of GDT in reducing mortality in the low control mortality subgroups could be due to a low statistical power needing much larger numbers of patients to show statistical significance. However, this subgroup analysis including 6900 patients was enough powered to exclude the latter hypothesis. An alternative hypothesis is that patients that are not very ill may not respond as clearly to increased hemodynamic. The quality analysis showed that the global effect of GDT on mortality was driven by high risk of bias studies, while higher quality studies did not demonstrate any benefit in mortality reduction. The meta-regression analysis further confirmed this figure. The risk of bias for each study was evaluated and studies were Vol. 82 - No. 11 classified as low and high risk of bias according to the domain-based evaluation, as that proposed by the Cochrane Collaboration. Out of 58 studies, 26 reached a low risk of bias evaluation. Many studies presented some important limitations since were conducted in single centers with limited patient samples, and only few RCTs were adequately randomized and double-blinded. The overall low quality of individual studies on GDT has been previously called into question,3 although the trial quality seemed to influence the outcome in the studies including perioperative patients less than in the subset of patients with established sepsis and multiple organ failure.5, 7 However, it is well established that studies with high risk of bias often overestimate the true effect, reducing the clinical significance of any result,13 and this could explain the results of the present study. Interestingly, most of the studies with low risk of bias are also low mortality studies: we therefore tried to combine the two subgroup analyses making high mortality/low bias, high mortality/high bias, low mortality/ low bias and low mortality/high bias groups. This analysis confirmed that no effect was seen in mortality rate <10%, both in low and in high risk of bias, while the benefit on mortality was driven by high risk of bias trials (9 RCTs), while the subgroup including mortality >10% and low risk of bias (6 RCTs) did not reach statistical significance. It has been proposed that the year of publication could affect the risk of bias, since older paper are more prone to high risk of bias, while newer ones are less affected by risk of bias. The meta-regression adopting the publication year as a covariate showed that there is no statistical dependence between the effect on mortality and the year of publication, suggesting that also older RCTs, if well planned and conducted, could be considered as low risk of bias, while recent ones, if not adequately designed, could be affected by high risk of bias. Therefore, another possible interpretation of the present meta-analysis could be that, when dealing with the effect of perioperative hemodynamic optimization on mortality, one should consider not only the “risk of bias” per se, but Minerva Anestesiologica 1209 GIGLIO GOAL DIRECTED THERAPY AND MORTALITY also the design of the study, and, maybe more important, the type of population enrolled, including co-morbidities, ASA class and mortality risk. This study had a number of limitations. No attempt was made to correct for the type or quantity of fluids or inotropes given, because they are inconsistently reported in the literature and have a demonstrable wide variability in their dosing across studies. Moreover, the studies included varied in terms of hemodynamic monitoring, the goals proposed and achieved, the timing of intervention: this could have introduced a relatively high clinical heterogeneity, although the results remained consistent across a number of subgroups and sensitivity analyses. However, the high heterogeneity among the tools and goals used to define GDT is still a major clinical problem. It is hard to believe that GDT by means of a Masimo pulse oxymeter can in anyway be equal to GDT conducted by a pulmonary artery catheter which is the goal standard to measure cardiac output. Additional well-designed randomized controlled studies are necessary to clarify these discrepancies and to determine whether mortality can be reduced through the maintenance of perioperative tissue perfusion in high-risk surgical patient. Moreover, several issues need to be clarified, such as timing, monitoring tools and protocols adopted, as well as the targets adopted, as recently underscored.86 Conclusions This meta-analysis, within the limitations of existing data, the high heterogeneity among adopted protocols, and the analytic approaches used, suggested that preoperative GDT significantly reduced mortality when the event control rate is >10%. In well conducted non-biased studies no mortality benefit was observed but the effect may be limited due to inclusion of small number of high mortality trials. Additional well-designed randomized controlled studies are still necessary to clarify several discrepancies among monitoring tools, goals and timing. 1210 Key messages —— The present meta-analysis, adopting the meta-regression technique, suggested that preoperative hemodynamic optimization significantly reduced mortality even when the event control rate is >10%. —— The global effect of GDT on mortality was driven by high risk of bias studies, while higher quality studies did not demonstrate any benefit in mortality reduction. —— When dealing with the effect of perioperative hemodynamic optimization on mortality, one should consider not only the “risk of bias” per se, but also the design of the study, and, maybe more important, the type of population enrolled, including comorbidities, ASA class and mortality risk. References 1. Harten J, Kinsella J. Perioperative optimisation. Scott Med J 2003;49:6-9. 2. Shoemaker WC, Appel PL, Kram HB. Hemodynamic and oxygen transport responses in survivors and nonsurvivors of high-risk surgery. Crit Care Med 1993;21:97790. 3. Heyland DK, Cook DJ, King D, Kernerman P, BrunBuisson C. Maximizing oxygen delivery in critically ill patients: a methodologic appraisal of the evidence. Crit Care Med 1996;24:517-24. 4. Kern JW, Shoemaker WC. Meta-analysis of hemodynamic optimization in high-risk patient. Crit Care Med 2002;30:1686-92. 5. Poeze M, Greve JWM, Ramsay G. Meta-analysis of hemodynamic optimization: relationship to methodological quality. Critical Care 2005;9:R 771-R779. 6.Dell’Anna AM, Taccone FS. Early-goal directed therapy for septic shock: is it the end? Minerva Anestesiol 2015;81:1138-43. 7. Hamilton, MA, Cecconi M, Rhodes A. A Systematic Review and Meta-Analysis on the Use of Preemptive Hemodynamic Intervention to Improve Postoperative Outcomes in Moderate and High-Risk Surgical Patients. Anesth Analg 2011,112:1392-40. 8.Gurgel ST, do Nascimento P. Maintaining Tissue Perfusion in High-Risk Surgical Patients: A Systematic Review of Randomized Clinical Trials. Anesth Analg 2011;112:1384-91. 9. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: the PRISMA statement. BMJ 2009;339:b2535. 10. Lefebvre C, Manheimer E, Glanville J. Searching for studies. In Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.1. Volume Chapter 6. Edited by: Higgins JPT, Green S. The Cochrane Collaboration (updated September 2008); [http://www.cochrane-handbook.org/]. 11. McAuley L, Pham B, Tugwell P, Moher D. Does the inclusion of grey literature influence estimates of inter- Minerva Anestesiologica November 2016 GOAL DIRECTED THERAPY AND MORTALITYGIGLIO vention effectiveness reported in meta-analyses? Lancet 2000;356:1228-31. 12. Copeland GP, Jones D, Walters M. POSSUM: a scoring system for surgical audit. Br J Surg 1991;78:355-60. 13. Brienza N, Giglio MT, Marucci M, Fiore T. Does perioperative hemodynamic optimization protect renal function in surgical patients? A meta-analytic study. Crit Care Med 2009;37:2079-90. 14. Higgins JPT, Altman DG, Sterne JAC. Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.cochrane-handbook.org. 15. Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. Assessing the quality of randomized clinical trials: is blinding necessary? Control Clin Trials 1996;17:1-12. 16. Juni P, Witschi A, Bloch R, Egger M. The hazards of scoring he quality of clinical trials for meta-analysis. JAMA 1999;282:1054-60. 17. Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003,327:557-60. 18. Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med 2002;21:1539-58. 19. Deeks JJ, Higgins JPT, Altman DG. Chapter 9: Analysing data and undertaking meta-analyses. In: Higgins JPT, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.1 (updated September 2008). The Cochrane Collaboration, 2008. Available from www.cochrane-handbook.org 20. Thompson S, Higgins J. How should meta-regression analyses by undertaken and interpreted? Stat Med 2002;21:1559-74. 21. Van Houwelingen HC, Arends LR, Stijnen T. Advanced methods in meta-analysis: multivariate approach and meta-regression. Stat Med 2002;21:589-624. 22.White IR. Multivariate random-effects meta-analysis. Stata J 2009;9:40-56. 23. Bartha E, Arfwedson C, Imnell A, Fernlund ME, Andersson LE, Kalman S. Randomized controlled trial of goaldirected hemodynamic treatment in patients with proximal femoral fracture. Br J Anaesth 2013;110:545-53. 24. Bender JS, Smith-Meek MA, Jones CE. Routine pulmonary artery catheterization does not reduce morbidity and mortality of elective vascular surgery: results of a prospective, randomized trial. Ann Surg 1997;226:229-36. 25. Benes J, Chytra I, Altmann P, Hluchy M, Kasal E, Svitak R, et al. Intraoperative fluid optimization using stroke volume variation in high risk surgical patients: results of prospective randomized study. Crit Care 2010;14:R118. 26. Berlauk JF, Abrams JH, Gilmour IJ, O’Connor SR, Knighton DR, Cerra FB. Preoperative optimization of cardiovascular hemodynamics improves outcome in peripheral vascular surgery: a prospective, randomized clinical trial. Ann Surg 1991;214:289-299. 27. Bisgaard J, Gilsaa T, Rønholm E, Toft P. Hemodynamic optimisation in lower limb arterial surgery: room for improvement? Acta Anaesthesiol Scand 2013;57:189-98. 28. Bisgaard J, Gilsaa T, Rønholm E, Toft P. Optimising stroke volume and oxygen delivery in abdominal aortic surgery: a randomised controlled trial. Acta Anaesthesiol Scand 2013;57:178-88. 29. Bishop MH, Shoemaker WC, Appel P, Meade P, Ordog GJ, Wasserberger J, et al. Prospective, randomized trial of survivor values of cardiac index, oxygen delivery, and oxygen consumption as resuscitation endpoints in severe trauma. Arch Surg 1992,127:1175-9. 30. Bonazzi M, Gentile F, Biasi GM, Migliavacca S, Esposti D, Cipolla M, et al. Impact of perioperative hemodynam- Vol. 82 - No. 11 ic monitoring on cardiac morbidity after major vascular surgery in low risk patients. A randomised pilot trial. Eur J Vasc Endovasc Surg 2002;23:445-51. 31. Boyd O, Grounds M, Bennett D. A randomized clinical trial of the effect of deliberate perioperative increase of oxygen delivery on mortality in high-risk surgical patients. JAMA 1993;270:2699-2708. 32. Brandstrup B, Svendsen PE, Rasmussen M, Belhage B, Rodt SÅ, Hansen B, et al. Which goal for fluid therapy during colorectal surgery is followed by the best outcome: near-maximal stroke volume or zero fluid balance? Br J Anaesth 2012;109:191-9. 33. Buettner M, Schummer W, Huettemann E, Schenke S, van Hout N, Sakka SG. Influence of systolic-pressurevariation-guided intraoperative fluid management on organ function and oxygen transport. Br J Anaesth 2008;101:194-9. 34.Cecconi M, Fasano N, Langiano N, Divella M, Costa MG, Rhodes A, et al. Goal-directed hemodynamic therapy during elective total hip arthroplasty under regional anaesthesia. Crit Care 2011;15:R132. 35. Challand C, Struthers R, Sneyd JR, Erasmus PD, Mellor N, Hosie KB, et al. Randomized controlled trial of intraoperative goal-directed fluid therapy in aerobically fit and unfit patients having major colorectal surgery. Br J Anaesth 2012;108:53-62. 36.Chytra I, Pradl R, Bosman R, Pelnár P, Kasal E, Zidková A. Esophageal Doppler-guided fluid management decreases blood lactate levels in multiple-trauma patients: a randomized controlled trial. Crit Care 2007;11:R24. 37. Conway DH, Mayall R, Abdul-Latif MS, Gilligan S, Tackaberry C. Randomised controlled trial investigating the influence ofintravenous fluid titration using Esophageal Doppler monitoring during bowel surgery. Anaesthesia 2002;57:845-9. 38. Donati A, Loggi S, Preiser JC, Orsetti G, Münch C, Gabbanelli V, et al. Goal-directed intraoperative therapy reduces morbidity and length of hospital stay in high-risk surgical patients. Chest 2007;132:1817-24. 39. Fleming A, Bishop M, Shoemaker W, Appel P, Sufficool W, Kuvhenguwha A, et al. Prospective trial of supranormal values as goals of resuscitation in severe trauma. Arch Surg 1992;127:1175-9. 40. Forget P, Lois F, de Kock M. Goal-directed fluid management based on the pulse oximeterderived pleth variability index reduces lactate levels and improves fluid management. Anesth Analg 2010;111:910-4. 41. Gan TJ, Soppitt A, Maroof M, el-Moalem H, Robertson KM, Moretti E, et al. Goal-directed intraoperative fluid administration reduces length of hospital stay after major surgery. Anesthesiology 2002;97:820-6. 42. Goepfert MS, Richter HP, Eulenburg CZ, Gruetzmacher J, Rafflenbeul E, Roeher K, et al. Individually Optimized Hemodynamic Therapy Reduces Complications and Length of Stay in the Intensive Care Unit: A Prospective, Randomized Controlled Trial. Anesthesiology 2013;119:824-36. 43. Harten J, Crozier JEM, McCreath B, Hay A, McMillan DC, McArdle CS, et al. Effect of intraoperative fluid optimisation on renal function in patients undergoing emergency abdominal surgery: a randomised controlled pilot study (ISRCTN 11799696). Int J Surg 2008;6:197204. 44. Jammer I, Ulvik A, Erichsen C, Lødemel O, Ostgaard G. Does central venous oxygen saturation-directed fluid therapy affect postoperative morbidity after colorectal surgery? A randomized assessor-blinded controlled trial. Anesthesiology 2010;113:1072-80. 45. Jhanii S, Vivian-Smith A, Lucena-Amaro S, Watson D, Hinds CJ, Pearse RM. Hemodynamic optimisation improves tissue microvascular flow and oxygenation after Minerva Anestesiologica 1211 GIGLIO 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. GOAL DIRECTED THERAPY AND MORTALITY major surgery: a randomised controlled trial. Crit Care 2010;14:R151. Jones C, Kelliher L, Dickinson M, Riga A, Worthington T, Scott MJ, et al. Randomized clinical trial on enhanced recovery versus standard care following open liver resection. Br J Surg 2013;100:1015-24. Kapoor M, Kakani M, Chowdhury U, Choudhury M, Lakshmy, Kiran U, et al. Early goal-directed therapy in moderate to high-risk cardiac surgery patients. Ann Card Anaesth 2008;11:27-34. Lobo SM, Salgado PF, Castillo VG, Borim AA, Polachini CA, Palchetti JC, et al. Effects of maximizing oxygen delivery on morbidity and mortality in high-risk surgical patients. Crit Care Med 2000;28:3396-404. Lopes MR, Oliveira MA, Pereira V, Lemos I, Auler J, Michard F. Goal-directed fluid management based on pulse pressure variation monitoring during high-risk surgery: a pilot randomized controlled trial. Crit Care 2007;11:R100. Mayer J, Boldt J, Mengistu A, Rohm K, Suttner S. Goaldirected intraoperative therapy based on autocalibrated arterial pressure waveform analysis reduces hospital stay in high-risk surgical patients: a randomized, controlled trial. Crit Care 2010;14:R18. McKendry M, McGloin H, Saberi D, Caudwell L, Brady AR, Singer M, et al. Randomised controlled trial assessing the impact of a nurse delivered, flow monitored protocol for optimisation of circulatory status after cardiac surgery. BMJ 2004;329:258. Mckenny M, Conroy P, Wong A, Farren M, Gleeson N, Walsh C, et al. A randomised prospective trial of intraoperative Esophageal Doppler-guided fluid administration in major gynaecological surgery. Anaesthesia. 2013;68:1224-31. Moppett IK, Rowlands M, Mannings A, Moran CG, Wiles MD. LiDCO-based fluid management in patients undergoing hip fracture surgery under spinal anaesthesia: a randomized trial and systematic review. Br J Anaesth 2015;114:444-59. Mythen MG, Webb AR. Perioperative plasma volume expansionreduces the incidence of gut mucosal hypoperfusionduring cardiac surgery. Arch Surg 1995;130:4239. Noblett SE, Snowden CP, Shenton BK, Horgan AF. Randomized clinical trial assessing the effect of Doppleroptimized fluid management on outcome after elective colorectal resection. Br J Surg 2006;93:1069-76. Pearse R, Dawson D, Fawcett J, Rhodes A, Grounds RM, Bennett ED. Early goal-directed therapy after major surgery reduces complications and duration of hospital stay. A randomised, controlled trial Crit Care 2005;9:687-93. Pearse R, Harrison DA, MacDonald N, Gillies MA, Blunt M, Ackland G, et al. Effect of a perioperative, cardiac output-guided hemodynamic therapy algorithm on outcomes following major gastrointestinal surgery: a randomized clinical trial and systematic review. JAMA 2014,311:2181-90. Peng K, Li J, Cheng H, Ji F. Goal-Directed Fluid Therapy Based on Stroke Volume Variations Improves Fluid Management and Gastrointestinal Perfusion in Patients Undergoing Major Orthopedic Surgery. Med Princ Pract 2014,23:413-20. Pestana D, Espinoza E, Eden A, Nájera D, Collar L, Aldecoa C, et al. Perioperative goal-directed hemodynamic optimization using noninvasive cardiac output monitoring in major abdominal surgery: a prospective, randomized, multicenter, pragmatic trial: POEMAS Study (PeriOperative goal-directed thErapy in Major Abdominal Surgery). Anesth Analg 2014;119:579-87. Polonen P, Ruokonen E, Hippelainen M, Pöyhönen M, 1212 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. Takala J. A prospective, randomized study of goal-oriented hemodynamic therapy in cardiac surgical patients. Anesth Analg 2000;90:1052-9. Poso T, Winso O, Aroch R, Kesek D. Perioperative Fluid Guidance with Transthoracic Echocardiography and Pulse-Contour Device in Morbidly Obese Patients. Obes Surg 2014;24:2117-25. Sandham JD, Hull RD, Brant RF, Knox L, Pineo GF, Doig CJ, et al. A Randomized, Controlled Trial of the Use of Pulmonary-Artery Catheters in High-Risk Surgical Patients. N Engl J Med 2003;348:5-14. Schereen TWL, Wiesenack C, Gerlach H, Marx G. Goaldirected intraoperative fluid therapy guided by stroke volume and its variation in high-risk surgical patients: a prospective randomized multicentre study. J Clin Monit Comput 2013;27:225-33. Senagore AJ, Emery T, Luchtefeld M, Kim D, Dujovny N, Hoedema R. Fluid management for laparoscopic colectomy: a prospective, randomized assessment of goal-directed administration of balanced salt solution or hetastarch coupled with an enhanced recovery program. Dis Colon Rectum 2009;52:1935-40. Shoemaker WC, Appel PL, Kram HB, Waxman K, Lee TS. Prospective trial of supranormal values of survivors as therapeutic goals in high-risk surgical patients. Chest 1988;94:1176-86. Sinclair S, James S, Singer M. Intraoperative intravascular volume optimisation and length of hospital stay after repair of proximal femoral fracture: randomised controlled trial. BMJ 1997;315:909-12. Smetkin AA, Kirov MY, Kuzkov VV, Lenkin AI, Eremeev AV, Slastilin VY, et al. Single transpulmonary thermodilution and continuous monitoring of central venous oxygen saturation during off-pump coronary surgery. Acta Anaesthesiol Scand 2009;53:505-14. Szakmany T, Toth I, Kovacs Z, Leiner T, Mikor A, Koszegi T, et al. Effects of volumetric vs. pressure-guided fluid therapy on postoperative inflammatory response: a prospective, randomized clinical trial. Intensive Care Med 2005;31:656-63. Ueno S, Tanabe G, Yamada H, Kusano C, Yoshidome S, Nuruki K, et al. Response of patients with cirrhosis who have undergone partial hepatectomy to treatment aimed at achieving supranormal oxygen delivery and consumption. Surgery 1998;123:278-86. Valentine RJ, Duke ML, Inman MH, Grayburn PA, Hagino RT, Kakish HB, et al. Effectiveness of pulmonary artery catheters in aortic surgery: a randomized trial. J Vasc Surg 1998;27:203-11. Van der Linden PJ, Dierick A, Wilmin S, Bellens B, De Hert SG. A randomized controlled trial comparing an intraoperative goal-directed strategy with routine clinical practice in patients undergoing peripheral arterial surgery. Eur J Anaesthesiol 2010;27:788-93. Velmahos GC, Demetriades D, Shoemaker WC, Chan LS, Tatevossian R, Wo CC, et al. Endpoints of resuscitation of critically injured patients: normal or supranormal? A prospective randomized trial. Ann Surg 2000;232:409-18. Venn R, Steele A, Richardson P, Poloniecki J, Grounds M, Newman P. Randomized controlled trial to investigate influence of the fluid challenge on duration of hospital stay and perioperative morbidity in patients with hip fractures. Br J Anaesth 2002;88:65-71 Wakeling HG, McFall MR, Jenkins CS, Woods WG, Miles WF, Barclay GR, et al. Intraoperative Esophageal Doppler guided fluid management shortens postoperative hospital stay after major bowel surgery. Br J Anaesth 2005;95:634-42. Wenkui Y, Ning L, Jianfeng G, Weiqin L, Shaoqiu T, Zhihui T, et al. Restricted peri-operative fluid administration adjusted by serum lactate level improved outcome after Minerva Anestesiologica November 2016 GOAL DIRECTED THERAPY AND MORTALITYGIGLIO 76. 77. 78. 79. 80. major elective surgery for gastrointestinal malignancy. Surgery 2010;147:542-52. Wilson J, Woods I, Fawcett J, Whall R, Dibb W, Morris C, et al. Reducing the risk of major elective surgery: Randomised controlled trial of preoperative optimisation of oxygen delivery. BMJ 1999,318:1099-103. Zakhaleva J, Tam J, Denoya PI, Bishawi M, Bergamaschi R. The impact of intravenous fluid administration on complication rates in bowel surgery within an enhanced recovery protocol: a randomized controlled trial. Colorectal Dis 2013;15:892-9. Zhang J, Chen CQ, Lei XZ, Feng ZY, Zhu SM. Goaldirected fluid optimization based on stroke volume variation and cardiac index during one-lung ventilation in patients undergoing thoracoscopy lobectomy operations: a pilot study. Clinics 2013;68:1065-70. Zheng H, Guo H, Ye JR, Chen L, Ma HP. Goal-Directed Fluid Therapy in Gastrointestinal Surgery in Older Coronary Heart Disease Patients: Randomized Trial. World J Surg 2013;37:2820-9. Ziegler DW, Wright JG, Coban PS, Flancbaum L. A prospective randomized trial of preoperative “optimization” of cardiac function in patients undergoing elective peripheral vascular surgery. Surgery 1997;122: 584-92. 81. Ghaferi AA, Birkmeyer JD, Dimick JB. Variation in hospital mortality associated with inpatient surgery. N Engl J Med 2009;361:1368-75. 82. Pearse RM, Moreno RP, Bauer P, Pelosi P, Metnitz P, Spies C, et al. Mortality after surgery in Europe: a 7 day cohort study. Lancet 2012;380:1059-65. 83. Stone MD, Wilson RJ, Cross J, Williams BT. Effect of adding dopexamine to intraoperative volume expansion in patients undergoing major elective abdominal surgery. Br J Anaesth 2003;91:619-24. 84. Takala J, Meier-Hellmann A, Eddleston J, Hulstaert P, Sramek V. Effect of dopexamine on outcome after major abdominal surgery: a prospective, randomized, controlled multicenter study. European Multicenter Study Group on Dopexamine in Major Abdominal Surgery. Crit Care Med 2000;28:3417-23. 85. Lobo SM, Lobo FR, Polachini CA, Patini DS, Yamamoto AE, de Oliveira NE, et al. Prospective, randomized trial comparing fluids and dobutamine optimization of oxygen delivery in high-risk surgical patients. Crit Care 2006;10:R72. 86. Berger MM, Gradwohl-Matis I, Brunauer A, Ulmer H, Dünser MW. Targets of perioperative fluid therapy and their effects on postoperative outcome: a systematic review and meta-analysis. Minerva Anestesiol 2015;81:794-808. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: April 14, 2016. - Manuscript accepted: April 12, 2016. - Manuscript revised: April 8, 2016. - Manuscript received: November 12, 2015. For supplementary materials, please see the online version of this article. Vol. 82 - No. 11 Minerva Anestesiologica 1213 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1214-29 REVIEW Targeting blood products transfusion in trauma: what is the role of thromboelastography? Samy FIGUEIREDO *, Audrey TANTOT, Jacques DURANTEAU 1Département d’Anesthésie et de Réanimation, Hôpitaux Universitaires Paris-Sud, Université Paris-Sud, Hôpital de Bicêtre, Assistance Publique – Hôpitaux de Paris, Le Kremlin-Bicêtre, France *Corresponding author: Samy Figueiredo, Département d’Anesthésie et de Réanimation, Hôpitaux Universitaires Paris-Sud, Université Paris-Sud, Hôpital de Bicêtre, Assistance Publique – Hôpitaux de Paris, 78 rue du Général Leclerc 94275 Le Kremlin-Bicêtre CEDEX, France. E-mail: [email protected]. A B STRACT Viscoelastic hemostatic assays (VHAs), mainly thromboelastography (TEG) and the rotational thromboelastometry (ROTEM), provide global information on clot formation and dissolution at patient bedside, allowing fast identification of coagulation disorders. In trauma patients, VHAs are able to predict massive transfusion and mortality. These devices might also be used for applying targeted administration of procoagulant factors (e.g. fibrinogen concentrate) as an alternative to or in addition to using predefined fixed ratios of red blood cells: platelets: fresh frozen plasma/cryoprecipitate. These goal-directed, individualized treatment algorithms seem to reduce blood product transfusion without deleterious effects on patient outcome. Nevertheless, a clear outcome benefit of using VHAs remains to be demonstrated in trauma patients. (Cite this article as: Figueiredo S, Tantot A, Duranteau J. Targeting blood products transfusion in trauma: what is the role of thromboelastography? Minerva Anestesiol 2016;82:1214-29) Key words: Blood coagulation disorders - Hemostatics - Thromboelastography - Blood transfusion - Blood coagulation. U ncontrolled hemorrhage is the leading cause of death in trauma patients.1 Hemorrhage may lead to coagulopathy and coagulopathy is able to exacerbate bleeding, thus creating a lethal vicious circle. Coagulation disorders are present in about 25-35% of trauma patients at their admission and impact significantly on morbidity and mortality.2-4 Current knowledge considers tissue injury and hypoperfusion as the main drivers of acute traumatic coagulopathy, leading to systemic anticoagulation, hypocoagulability and hyperfibrinolysis through the activation of the protein C pathway.4 Acidosis, hypothermia and hemodilution secondary to inadequate resuscitation will exacerbate coagulopathy. Fast and accurate diagnostic of coagulopathy is a key element for achieving 1214 adequate hemostatic resuscitation in bleeding trauma patients. Standard laboratory coagulation tests (SLCTs) including prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen concentration (Clauss method), D-dimer and platelets count are usually used to evaluate patients’ coagulation status. With the descriptions of the cell-based model of hemostasis 5 and the acute traumatic coagulopathy,4 the limitations of SLCTs have been increasingly recognized. First, SLCTs only evaluate plasmatic activation without taking into account the important interactions between clotting factors, platelets and other cellular components of whole blood involved in thrombin generation. Secondly, SCLTs do not evaluate the overall clot strength or the balance Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO between clot formation and fibrinolysis. Thirdly, the variable processing time, from blood sampling to obtaining results (median>80 minutes) 6 may impair the usefulness of SCLTS in emergency settings such as trauma. In recent years, there has been an increasing interest for viscoelastic hemostatic assays (VHAs), mainly thromboelastography (TEG; Hemoscope Corporation, Niles, OH, USA) and the rotational thromboelastometry (ROTEM; Tem International GmbH, Munich, Germany), to provide global and functional assessment of coagulation. These devices could enable clinicians to rapidly identify which component(s) of the coagulation process should be targeted during hemostatic resuscitation. This manuscript reviews recently published data on the use of TEG/ROTEM in trauma patients and discusses their contribution to coagulation management and their impact on morbidity and mortality. Materials and methods We searched Medline, Embase and Cochrane Library databases to identify studies in English on thromboelastography and/or rotational thromboelastometry in trauma. We used the MeSH terms and keywords “thromboelastography” AND “trauma”, “rotational thromboelastometry” AND “trauma”, “TEG” AND “trauma”, “ROTEM” AND “trauma”. Abstracts were screened for eligibility. The reference lists of all articles selected for detailed review and all relevant published reviews were searched to find any other studies potentially eligible for inclusion. The date of the last search was 31st January 2016. All original studies and major review articles concerning the use of TEG and/or ROTEM in trauma were included. �������������������������������������� The characteristics of the studies included in the present review are presented in Table I. Principles and theoretical advantages of thromboelastography VHAs such as TEG and ROTEM can be performed at patient bedside and give a graphic presentation of clot formation and subsequent lysis. Briefly, the collected wholeblood sample is placed in a special designed Mechanical-electrical transducer Torsion wire 4° 75 Pin Blood sample Cup 4°45 A B TEG ROTEM Figure 1.—Principles of thromboelastography (A) and rotational thromboelastometry (B). Citrated or native whole-blood is inserted in a specific cup with an activator of coagulation. A pin suspended by a torsion wire is immerged in the cup and connected to a detector system. Cup and pin are oscillated relative to each other with movement initiated from either the cup (TEG) or the pin (ROTEM). As fibrin forms between the cup and pin, the transmitted rotation from the cup to pin (TEG) or the impedance of the rotation of the pin (ROTEM) are detected and a tracing is generated. Vol. 82 - No. 11 Minerva Anestesiologica 1215 FIGUEIREDOTARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMA cup with an activator of coagulation. Inside the cup is suspended a pin connected to a detector system, and the cup and pin are oscillated relative to each other with movement initiated from either the cup (TEG) or the pin (ROTEM). As fibrin forms between the cup and pin, the transmitted rotation from the cup to pin (TEG) or the impedance of the rotation of the pin (ROTEM) are detected and a trace is generated as seen in Figure 1. This trace is divided into several parts, each one reflecting a different stage of the hemostatic process (Figure 1). Although TEG and ROTEM nomenclatures are different, both provide information on the speed of clot initiation (coagulation factors), kinetics of clot growth (thrombin generation and cleavage of fibrinogen), clot strength, and fibrinolysis (Table I). The contribution of fibrinogen to clot strength can be evaluated by adding platelet inhibition agents to the (Functional Fibrinogen assay for TEG and FIBTEM assay for ROTEM). Analysis of the tracings morphology and parameters will determine which component(s) of the hemostatic process needs to be targeted (Table II). COAGULATION Clot Clot initiation kinetics Thromboelastography-guided hemostatic resuscitation Given that hemorrhage-related mortality in trauma patients occurs within the 2 or 3 first hours after injury,7, 8 it is crucial to start the management of coagulopathy as soon as hemorrhage is clinically suspected. This management is part of an overall approach of damage-control resuscitation: correction of hypothermia and acidosis, restrictive fluid administration, permissive hypotension in selected populations and restoration of blood volume.9 Unbalanced transfusion strategies systematically lead to depletion and dilution of coagulation factors, increasing exsanguination. Consequently, hemostatic resuscitation combines repletion of red blood cells (RBC), platelets (PLT) and coagulation factors by transfusion (RBC, PLT, fresh frozen plasma (FFP)/cryoprecipitate) and/or administration of pharmaceutical hemostatic agents such as fibrinogen concentrate (FC), prothrombin complex concentrate (PCC), antifibrinolytic agents (mainly tranexamic acid) and less frequently desmopressin, recombinant factor VIIa and factor XIII. FIBRINOLYSIS Clot strength K TEG R MA LY30 MCF Li30 α angle α angle CT ROTEM CFT 10 20 50 Time (min) Figure 2.—Classical representation of TEG and ROTEM tracings. TEG: thrombelastography; ROTEM: rotational thromboelastometry; R-time: reaction time; CT: clotting time; K-time: coagulation time; CFT: clot formation time; MA: maximum amplitude; MCF: maximum clot firmness; LY 30: lysis at 30 min; Li30: clot lysis at 30 min. 1216 Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO Targeting red blood cell transfusion In bleeding trauma patients, RBC transfusion aims not only at improving perfusion and oxygen delivery but also at providing sufficient cellular phospholipid surfaces to enhance thrombin generation.10 RBCs also increase platelets activation and promote clotting at the bleeding site by pushing platelets to the vascular wall.11 In addition, logistic issues make it crucial to anticipate massive transfusion (MT) as soon as possible. VHAs are able to predict MT, usually defined as transfusion of ≥10 RBC units within 24 hours of trauma. Most of the studies showed that TEG/ROTEM had similar ability to predict MT than standard coagulation tests performed at the laboratory, with shorter turnaround times. The following TEG values were found to be predictive of MT: ACT>128 (odds ratio=5; 95% CI: 1-19; P=0.01),12 α-angle <56,13 G value 14 and hyperfibrinolysis defined by LY30>15% (OR=19) 15 or even >3% (with specificity and PPV > 90% but very low 31% sensibility).16 TEM values predictive of MT were: FIBTEM MCF and FIBTEM A10 (AUC-ROC curves >0.83),17 clot amplitude at 5 minutes [CA5] ≤35 mm, which had better sensitivity [71% vs. 42%, P<0.001] but worse specificity [85% vs. 94%] compared to a PTr>1.2,18 abnormal MCF (OR=8 and AUCROC curve=0.8 [95% CI: 0.7-0.9]; P<0.001) even though Hb<10g/dL was a stronger predictor of MT (OR=18). Besides MT prediction, rTEG ACT,12 TEG MA 19 and ROTEM MCF FIBTEM 20 were found to be correlated with the amount of blood products transfused whereas rTEG α-angle <74.7 was predictive of any transfusion.21 One study with a relatively small population of transfused patients (22 of 161) found that common TEG parameters (R, K, angle, MA, LY60) were not statistically different between transfused and not transfused patients; only MA-ADP from Platelet-Mapping® was statistically different (P=0.004).22 Studies focusing on the performances of the different VHAs to predict MT are summarized in Table III. Vol. 82 - No. 11 Targeting repletion of coagulation factors (PLT, FFP, cryoprecipitate or PCC): VHAs or massive transfusion protocols? Rapid and efficient blood product administration may be achieved either by massive transfusion protocols (MTPs) using predefined fixed ratios of RBC:PLT:FFP/cryoprecipitate or by applying goal-directed administration of coagulation factor concentrates guided by VHAs. Benefits reported from MTPs using fixed ratios are related to facilitation of communication in emergency settings between the blood transfusion service and the clinical team and to standardization of patient care between all medical providers in one center. However, optimal ratio is not so easy to be defined: if it is too low, treatment of coagulopathy is delayed and bleeding continues; if it is too high, indiscriminate administration of blood products may lead to transfusion-related complications (immunomodulation, acute lung injury, cardiac overload) or waste. Recently, the PROPPR Study 8 showed that among 680 patients predicted to receive MT and randomly assigned to a 1:1:1 or 1:1:2 FFP:PLT:RBC transfusion ratio, no significant difference in overall mortality at 24 hours or 30 days was detected. However, more patients achieved hemostasis and fewer patients died of exsanguination in the 1:1:1 group, without any increase in morbidity outcomes. It is noteworthy that the authors used a platelet first strategy for administering the 1:1:1 ratio whereas more than 80% of the patients had a platelet count ≥150.000/mm3. The recently recognized phenomenon of platelet dysfunction after trauma patients might be a rationale for this strategy.23 The “theragnostic approach” using point-ofcare VHAs allows an individualized and goaldirected hemostatic therapy with the use of coagulation factor concentrates such as FC, PCC and less frequently rFVIIa and Factor XIII. This strategy is primarily used in Austria and Switzerland. A first cohort study on 131 trauma patients described the ROTEM-guided administration of FC and PCC in addition to FFP and PLT, and found a favourable survival rate compared with a predicted survival rate by the Trauma Injury Severity Score (TRISS).24 Given that Minerva Anestesiologica 1217 FIGUEIREDOTARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMA Table I.—Characteristics of all the studies included in the present review. Reference Year Design Cotton 12 2011 Prospective, observational, single center Holcomb 13 2012 Pezold 14 Patients (n) Population Technique Trauma patients; Median ISS=14 rTEG citrated blood Retrospective, 1974 cohort study, single center Trauma patients; Median ISS=17 rTEG citrated blood 2012 Retrospective 80 Trauma patients with ISS>16 42 non-MT vs. 38 MT receiving more than 10U of RBC in the first 24 hours rTEG fresh whole blood Ives 15 2012 Prospective, single center 118 Trauma patients with an ISS>16 TEG citrated blood Chapman 16 2013 Prospective, single center 289 Trauma patients; TEG Median ISS=30 citrated blood MTP criteria at admission (N.=73) vs. all non-MTP trauma patients (N.=216) in the same period Schöchl 17 2011 Retrospective, observational 323 Trauma patients with an ISS >16 MT group (78) receiving more than 10U of RBC in 24h versus non MT group (245) ROTEM citrated blood Davenport 18 2011 Prospective, observational, cohort study 300 Trauma patients Median ISS=12 ROTEM citrated blood Nystrup 19 2011 Retrospective 89 Trauma patients Median ISS=21 TEG citrated blood Tauber 20 2011 Prospective, observational, single center 334 Trauma patients Median ISS=34 Polytrauma (274) and isolated head injury (60) ROTEM citrated blood Jeger 21 2012 Prospective, observational single center 76 Trauma patients Median ISS=29 rTEG citrated blood Carroll 22 2009 Prospective, observational, single center 161 Trauma patients Median ISS=20 TEG and PLT Mapping citrated blood Schöchl 24 2010 Retrospective 131 Trauma patients receiving more than 5U RBC/24h Median ISS=38 ROTEM citrated blood Schöchl 25 2011 Retrospective 681 Trauma patients ROTEM citrated blood 1218 272 Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO Objectives The ability of r-TEG to predict early blood transfusion (<2h) Main Results Linear regression demonstrated that ACT predicted RBC, plasma and PLT transfusions within the first 2 h of arrival. ACT>128 predicted MT (>10U of RBC) in the first 6 hours (OR 5; 95% CI, 1-19; P=0.01). The ability of r-TEG to reliably ACT-predicted RBC transfusion, and the α-angle predicted massive RBC transfusion better predict blood component trans- than PT, aPTT or INR (P<0.001). fusion compared to SLCTs The α-angle was superior to fibrinogen for predicting FFP transfusion (P<0.001). MA was superior to PLT count for predicting PLT transfusion (P<0.001). rTEG for prediction of MT and The predictive power for MT did not differ among INR (adjusted AUC ROC=0.92), aPTT mortality (AUC ROC=0.90, NS), or G (AUC ROC=0.89, NS). For MT-death, G had the greatest adjusted AUC ROC (0.93) compared with the AUC ROC for BD (0.87, P=0.05), INR (0.88, NS), and PTT (0.89; P=NS). MT and mortality associated with HF HF was present in 13 patients (11%). HF patients had a greater need for MT (77% vs. 9%; adjusted OR=19; 95% CI, 4-101; P<0.001) and had a greater early mortality (69% vs. 2%; adjusted OR=56; 95% CI, 7-432; P<0.001) and in-hospital mortality (92% vs. 9%; adjusted OR=56; 95% CI, 5-650; P=0.001). LY30 threshold associated with HF was present in 19 patients (26%) and 11 patients (15%) had LY30≥3%. MT and mortality LY30≥3%: performance for prediction of MT (in the first 6 hours): PPV=91%, Sens=31%, Spe=98%, NPV=65%. The ability of ROTEM for early The best predictive values for MT were provided by Hb and Quick value (AUC ROC=0.87 prediction of MT. for both parameters). Similarly high predictive values were observed for FIBTEM MCF (0.84) and FIBTEM A10 (0.83). Detection of ATC (defined by PTr> 1.2) with ROTEM compared with SLCTs, POC PT, and BD CA5 threshold ≤35mm had a detection rate of 77% for ATC with a false positive rate of 13%. Patients with CA5 ≤ 35 mm were more likely to receive RBC (46% vs. 17%, P<0.001) and FFP (37% vs. 11%, P<0.001) Better detection rate of MT with CA5=71% (vs. 43% for PTr>1.2, P<0.001). Transfusion requirement and mortality associated with low clot strength (MA) MA correlated with the amount of RBC (P=0.01), FFP (P=0.04) and PLT (P=0.03) transfused in the first 24 h. Patients with reduced MA demonstrated increased 30-day mortality (47% vs. 10%, P<0.001). Low MA is independently associated with mortality after adjusting for age and ISS. ROTEM threshold associated with transfusion requirement and mortality Significant differences in mortality were detected for defined ROTEM-thresholds: FIBTEM 7 mm (21% vs. 9%, P<0.01), EXTEM MCF 45 mm (25% vs. 9%, P<0.001). MCF FIBTEM was correlated with RBC transfusion (OR 0.9, 95% CI 0.9-0.98). HF increased fatality rates and occurred as frequently in isolated TBI as in polytrauma. Correlation between SLCTs and The r-TEG α-angle was the parameter with the greatest sensitivity (84%) and validity (77%) rTEG parameters in the predic- at a cut-off of 75 degrees. tion of any transfusion Physicians blinded to TEG: transfusion guided clinically and with SLCTs results TEG and Platelet mapping for prediction of transfusion and mortality No TEG parameter was significantly different for the 22 patients who required transfusion, except MA-ADP (P<0.01) R and MA were correlated with mortality (both P<0.001) ROTEM (FIBTEM, EXTEM)guided FC and PCC therapy Difference in mortality was 14% observed vs. 28% predicted by TRISS (P<0.01) and 24 % predicted by RISC (P=0.014). Standard FFP transfusion com- RBC transfusion avoided in 29% of patients in the FC-PCC group compared with only 3% in pared to PCC and FC guided by the FFP group (P<0.001). ROTEM PLT transfusion avoided in 91% of patients in the FC-PCC group, compared with 56% in the FFP group (P<0.001). Mortality was comparable between groups: 7.5% in the FC-PCC group and 10% in the FFP group (NS). (To be continued) Vol. 82 - No. 11 Minerva Anestesiologica 1219 FIGUEIREDOTARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMA Table I.—Characteristics of all the studies included in the present review (Continues). Reference Patients (n) Population Technique Tapia 26 2013 Year Retrospective, before/after study Design 289 TEG fresh whole blood Gonzalez 28 2015 Prospective, randomized control trial, single center 111 Moore 30 2014 Prospective, observational, single center 180 Rourke 38 2012 Prospective, cohort study, 2 centers 517 Patients receiving≥6U of RBC in the first 24h before (N.=165) and after (N.=124) MTP initiation Median ISS=25 PreMTP group (165) and standardized MTP group (N.=124) Trauma patients meeting criteria for MTP activation (SBP<70 mmHg and/or HR >108/min) Median ISS=30 TEG group (N.=56) vs. SLCT group (N.=55) Trauma patients Median ISS=29 3 groups according to fibrinolysis: HF (LY30≥3%); physiologic (LY30: 0.0812.9%), and shutdown (LY30: 0-0.08%) Trauma patients Meyer 40 2015 Prospective, observational, cohort study, single center 182 Trauma patients Median ISS=17 TEG and FIBTEM citrated blood Kornblith 41 2014 Prospective, observational, single center 251 Trauma patients Median ISS=9 TEG citrated blood Agren 44 2014 Prospective, observational, single center 101 TEG citrated blood Schlimp 46 2013 Retrospective, single center 157 Chapman 55 2015 Retrospective, cohort study 572 63 patients with trauma or surgery with ongoing bleeding and 38 healthy blood donors Trauma patients receiving more than 1 g Fibrinogen within 24h 3 groups: FC group, FC-PCC group and FCPCC-FFP group Trauma patients with≥1U of RBC Raza 56 2013 Prospective, cohort study, single center 288 Trauma patients Median ISS=10 ROTEM citrated blood Kashuk 68 2012 Retrospective, before (no rTEG)/after (with rTEG) study 68 Patients receiving more than 6 RBCs/6 hours rTEG 62% ISS >36 fresh whole blood rTEG fresh whole blood TEG citrated blood ROTEM citrated blood ROTEM citrated blood rTEG fresh whole blood ATC: acute traumatic coagulopathy; BD: ���������������������������������������������������������������������������������������������������� base deficit; FC: ���������������������������������������������������������������������������������� fibrinogen concentrate; FFP: fresh frozen plasma; Hb: hemoglobin; ������������ HF: Hyperfibrinolysis; MT: massive transfusion; MTP: massive transfusion protocol; NPV: negative predictive value; NS: non significant; OR: odds ratio; PAP: plasmin–antiplasmin complex; PCC: prothrombin complex concentrate; PLT: platelet; PPV: positive predictive value; PTr: PT ratio; RBC: red blood cell; Sens: sensibility; SLCT: Standard laboratory coagulation test; Spe: specificity. 1220 Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO Objectives Main Results TEG-guided resuscitation vs. standardized MTP TEG-guided resuscitation is equivalent to standardized MTP for patients receiving≥6U of RBC and for blunt trauma receiving≥10U of RBC. TEG-directed resuscitation is superior to MTP in patients with penetrating trauma receiving≥10U of RBC. MTP therapy worsened mortality in penetrating trauma patients receiving≥10U of RBC. rTEG-guided vs. SLCT-guided MTP 28-days survival in the rTEG-guided MTP group was significantly higher than the SLCT group (P=0.032). SLCT group required similar number of RBC units as the rTEG group (SLCT: 5 [2-11], TEG: 4.5 [2-8] (NS), but more plasma units and more PLT units in the first 2 hours of resuscitation). Fibrinolysis threshold associated with mortality rates Mortality rates were lower for the physiologic group (3%) compared with the HF (44%) and shutdown (17%) groups (P=0.001). U-shaped distribution of death related to the fibrinolysis system in response to major trauma, with a nadir in mortality, with level of fibrinolysis after 30 minutes between 0.81% and 2.9% Response and outcome associated with Fibrinogen replacement therapy, correlation to ROTEM parameters. Pre-fixed MTP including administration of RBC, FFP, PLT, CRYO and FC and ex-vivo FC. To compare TEG FF and FIBTEM with each other and with Clauss method Patients who received fibrinogen supplementation maintained their admission fibrinogen level (1.6 g.L-1). Fibrinogen level was an independent predictor of mortality at 24 h and 28 days (P<0.001). Patients who received cryoprecipitate maintained their fibrinogen levels, and had lower mortality rates than those who did not. FF MA and FIBTEM MCF had identical correlation coefficients (both ρ=0.64, P<0.001) and identical explained variances (both R2=0.41, P<0.001) Performance of FF MA and FIBTEM MCF versus Clauss fibrinogen levels of 1.5, 2.0, and 2.5 g/L by ROC analysis. For all three cut-offs, FF MA and FIBTEM MCF had AUROC curves above 0.8 with P<0.001. Patients requiring plasma transfusion had a significantly lower admission %MA(FF) (27% vs.. 31%, P<0.05). Higher admission %MA(FF) was predictive of reduced mortality (hazard ratio, 0.815, P<0.001). Level of FF and contribution of fibrinogen to clot strength associated with coagulopathy, transfusion requirements, and outcomes TEG-FF compared with Clauss TEG-FF was on average 1.0 g/L higher than the plasma fibrinogen concentration. method for fibrinogen concentration measurement Role of FC for rapidly increasing fibrinogen plasma levels. FFP given only to the most severely injured patients To determine which rTEG tracing pattern is exclusively found within the nonsurvivors Comparison of Fibrinolytic activation (FA) detected by ROTEM vs. plasmin–antiplasmin (PAP) complex and D-dimer levels. rTEG-guided resuscitation compared with patients admitted prior to the rTEG® period implementation. Vol. 82 - No. 11 FIBTEM-CF at 10 min (CA10) was maintained, with a small increase in the FC-PCC group. Fibrinogen concentration and FIBTEM CA10 were within the normal range in all groups at 24 hours. A “diamond-shaped” tracing, with short time to MA (≤ 14 min) and complete lysis before LY30 point, had 100% PPV for mortality. TEM detected clot lysis only when PAP complex levels were increased to 30 times normal (P<0.001) and antiplasmin levels were <75% of normal. Patients with FA had increased 28-day mortality as compared with those with no FA (12% vs. 1%, P<0.001). Patients with a MRTG > 9.2 received significantly less RBCs, FFP, and cryoprecipitate (all P values <0.05). G value from r-TEG was associated with survival (P=0.03). Minerva Anestesiologica 1221 FIGUEIREDOTARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMA Table II.—Main TEG and ROTEM parameters: definitions, representative coagulation process and therapeutic options. TEG parameter R-time (min) ROTEM parameter CT (min) ACT (sec) K-time (min) Coagulation parameter reflected Definition Time from the start of the test until 2 mm amplitude Enzymatic clotting factors Surrogate of R-time in the rapid-TEG Enzymatic clotting assay, which uses tissue factor to factors obtain a quicker tracing CFT (min) angle α (degrees) angle α (degrees) MA (mm) MCF (mm) G (dynes/cm2) MCE (dynes/cm2) LY30 (%) LI 30 (%) MA-FF (mm) FIBTEM (mm) Time from 2mm to 20mm amplitude Angle of a tangent line between the initial split point of the tracing and the growing curve Point at which clot strength reaches its maximum measure Overall total clot strength, calculated from MA Percentage of clot strength loss 30 min after reaching MA Contribution of fibrinogen to clot strength Enzymatic clotting factors Thrombin generation Therapeutic options to consider FFP or PCC if prolonged R-time (protamine if heparin present) FFP or PCC if prolonged ACT (protamine if heparin present) Fibrinogen, platelets, factor XIII All coagulation interactions Fibrinolysis FC and/or PLT if MA decreased Fibrinogen FC or cryoprecipitate if MA decreased TXA TEG: thrombelastography; ROTEM: rotational thromboelastometry; R-time: reaction time; ACT: activated clotting time; CT: clotting time; K-time: coagulation time; CFT: clot formation time; MA: maximum amplitude; MCF: maximum clot firmness; MCE: maximum clot elasticity; G: total clot strength; LY-30: lysis at 30 min; Li30: clot lysis at 30 min; MA-FF: maximum amplitude using Functional Fibrinogen assay; FFP: fresh frozen plasma; PCC: prothrombin complex concentrate; FC: fibrinogen concentrate; TXA: tranexamic acid; PLT: platelets. there was no control group, a second study compared these findings with the German Trauma Registry selected by transfusion of two units or more of FFP and then compared on transfusion therapy. RBC transfusion was avoided in 29% of patients in the FC-PCC group compared with only 3% in the FFP group (P<0.001). Transfusion of PLT was avoided in 91% of patients in the FC-PCC group, compared with 56% in the FFP group (P<0.001). Mortality was comparable between groups: 7.5% in the FC-PCC group and 10% in the FFP group (P=0.69).25 In a retrospective before/after study on 289 trauma patients without traumatic brain injury (TBI), Tapia et al. compared TEG-directed (=before) with MTP-directed (=after) resuscitation. The authors reported a survival benefit with TEG-directed resuscitation compared to a fixed 3RBC: 2PFC MTP in a subgroup of penetrating trauma patients requiring >10 units of RBC.26 All these before/after studies should be interpreted with caution since the results observed might be due to the use of TEG/ROTEM and/or only to the implementation of a protocol. Few randomized 1222 control trials (RCTs) have been conducted in order to determine the clinical impacts of VHAguided coagulation management on mortality and morbidity in trauma patients. Weber et al.27 included 100 cardiac surgical patients with coagulopathy in an RCT aiming at evaluating ROTEM-guided hemostatic therapy. The group treated according to a ROTEM algorithm had reduced chest tube drainage, reduced transfusion requirements and, importantly, reduced 30day mortality compared with the group treated according to SLCTs. Gonzalez et al. included 111 trauma patients in a very recently published RCT aiming at comparing MTP goal-directed by TEG and MTP guided by SLCTs. Patients resuscitated with TEG-guided MTP had significantly improved 6-hour and 28-day survivals and received less plasmas and platelets in the first 2 hours than patients receiving SLCTsguided MTP28. Some important points should be noted before generalizing the findings of this valuable study: 1) the first units of RBC and plasma were administered according to the clinician’s practice regardless of randomization Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO Table III.—Performances of the viscoelastic haemostatic assays to predict massive transfusion. Reference Year Cotton 12 2011 Prospective, observational, single center 2012 Retrospective, cohort study single center 2012 Retrospective Holcomb 13 Pezold 14 Design Patients (N.) Population Technique 272 Trauma patients; median ISS=14 rTEG citrated blood 1974 Trauma patients; median ISS=17 rTEG citrated blood 80 Trauma patients with rTEG ISS>16 fresh whole 42 non-MT vs. blood 38 MT receiving more than 10U of RBC in the first 24 hours Ives 15 2012 Prospective, single center 118 Trauma patients with an ISS>16 TEG citrated blood Chapman 16 2013 Prospective, single center 289 TEG citrated blood Schöchl 17 2011 Retrospective, observational 323 Davenport 18 2011 Prospective, observational, cohort study 300 Trauma patients; Median ISS=30 MTP criteria at admission (N.=73) vs. all non-MTP trauma patients (N.=216) in the same period Trauma patients with an ISS>16 MT group (78) receiving more than 10U of RBC in 24h vs. non MT group (245) Trauma patients Median ISS=12 Main Results ACT>128 predicted MT (>10U of RBC) in the first 6 hours (OR 5; 95%CI, 1-19; P=0.01) α-angle predicted massive RBC transfusion better than PT, aPTT or INR (P<0.001) The predictive power for MT did not differ among INR (adjusted AUC ROC=0.92), aPTT (AUC ROC=0.90, NS), or G (AUC ROC=0.89, NS) For MT-death, G had the greatest adjusted AUC ROC (0.93) compared with the AUC ROC for BD (0.87, P=0.05), INR (0.88, NS), and PTT (0.89; P=NS) HF was present in 13 patients (11%). HF patients had a greater need for MT (77% vs. 9%; adjusted OR=19; 95%CI, 4-101; P<0.001) and had a greater early mortality (69% vs. 2%; adjusted OR=56; 95% CI, 7-432; P<0.001) and in-hospital mortality (92% vs. 9%; adjusted OR=56; 95% CI, 5-650; P=0.001). LY30≥3%: performance for prediction of MT (in the first 6 hours): PPV=91%, Sens=31%, Spe=98%, NPV=65%. ROTEM citrated blood The best predictive values for MT were provided by Hb and Quick value (AUC ROC=0.87) ROTEM citrated blood Better detection rate of MT with CA5=71% (vs. 43% for PTr>1.2, P<0.001). ATC: acute traumatic coagulopathy; AUC: area under the ROC curve; BD: base deficit; FC: fibrinogen concentrate; FFP: fresh frozen plasma; Hb: hemoglobin; HF: Hyperfibrinolysis; MT: massive transfusion; MTP: massive transfusion protocol; NPV: negative predictive value; NS: non-significant; OR: odds ratio; PCC: prothrombin complex concentrate; PLT: platelet; PPV: positive predictive value; PTr: PT ratio; RBC: red blood cell; ROC: Receiver Operating characteristic; Sens: sensibility; SLCT: Standard laboratory coagulation test; Spe: specificity. group; 2) tranexamic acid was not widely used (12% of the whole cohort); 3) there are no data on the time and the methods used to achieve hemostasis. Additional studies, including RCTs, are warranted to confirm these beneficial effects of VHA-guided coagulation management on mortality and morbidity in trauma patients. Vol. 82 - No. 11 Noteworthy, using TEG/ROTEM devices does not necessarily imply using only FC and PCC. Thrombin generation does not represent a major issue in trauma patients. Since the administration of PCC will increase thrombin generation for several days PCC could increase the risk of thrombosis and should therefore be used with Minerva Anestesiologica 1223 FIGUEIREDOTARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMA caution.29 Factor replacement with FC and PCC is not considered equivalent to plasma or cryoprecipitate transfusion, as they do not contain important proteins present in these blood products, particularly Factors V, VIII and XIII. In addition, plasma transfusion may confer more benefits than FC and PCC beyond factor replacement.30 Plasma is an iso-osmolar resuscitation fluid with higher oncotic pressure than 0.9% saline and contains amount of proteins such as albumin, immunoglobulins, apolipoproteins and protease inhibitors. Interestingly, resuscitation with FFP compared to lactated Ringer’s or normal saline has been shown to attenuate endothelial glycocalyx disruption following hemorrhagic shock 31-33. Further studies are needed to better characterize potential benefits of plasma transfusion beyond restoration of coagulation. In summary, individualized hemostatic resuscitation guided by VHAs and use of MTPs with predefined FFP:PLT:RBC ratios have both strengths and weaknesses. VHAs allow goaldirected management of coagulopathy but it takes time to run the samples and to obtain the results. Activation of the MTP by the clinician leads to rapid and timely delivery of all predefined blood products but the optimal ratio is still being debated. Interestingly, it is not mandatory to choose a single strategy and to reject the other one: both strategies may be used at different times the resuscitation. Indeed, some authors advocate to initiate balanced transfusion therapy with transfusion packages using a 1:1:1 ratio in the early phase of massive bleeding, and then to use goal-directed administration of hemostatic blood components and coagulation factor concentrates during further resuscitation when information become available from laboratory or point-of-care tests (Copenhagen concept).34 Although this third strategy has recently been recommended,35 additional research is required in this field to precisely determine the respective role of VHAs and MTPs in coagulation resuscitation. Targeting fibrinogen supplementation Fibrinogen concentration decreases early after injury in coagulopathic trauma patients and 1224 low levels have been associated with worse outcome.36-38 Thus, fibrinogen supplementation is recommended in the management of trauma-induced coagulopathy.39 The optimal concentration of fibrinogen in this context has yet to be determined, but current consensus guidelines suggest a target level of 1.5-2.0 g/L.39 Clauss method is considered as a gold standard for measuring fibrinogen concentration in plasma. However, this method has slow turnaround times and do not reflect the qualitative (or functional) aspect of fibrinogen. Specific VHAs exist for estimating fibrinogen levels, including functional fibrinogen assay (FF) for TEG and FIBTEM for ROTEM. A recent study showed that both FF and FIBTEM correlated with fibrinogen level measured by Clauss method (both ρ=0.64, P<0.001), as well as with each other, 40 confirming previous results.41 Despite these correlations, absolute values obtained by using FF and FIBTEM are not equivalent. FF MA values have been reported to be higher than FIBTEM MCF values.42-44 Technological differences between the two devices along with differential effects of the platelets inhibitors used in the two tests (cytochalasin D for FIBTEM and abciximab for FF assay) may contribute to this phenomenon. Target values and thresholds for hemostatic therapy could consequently depend on the device used and clinicians should be aware of this issue. Fibrinogen supplementation may be provided by FFP, cryoprecipitate and/or FC. Plasma contains only low levels of fibrinogen 45 and some authors have shown that using plasma as the unique source of fibrinogen supplementation is not sufficient enough to correct fibrinogen depletion.38, 46 Cryoprecipitate contains variable amounts of fibrinogen (along with von Willebrand factor, factor VIII and factor XIII), is not virus inactivated and has to be pooled and thawed before administration. In contrast, FC contains a well-defined concentration of fibrinogen and is immediately available for use.47 In Europe, FC administration is authorized both in congenital and acquired coagulopathies, whereas its use is restricted to congenital coagulopathies in the United States. In summary, fibrinogen supplementation is Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO required in bleeding trauma patients and has probably to be done by administration of FC or cryoprecipitate guided by VHAs (initially 3-4 g or 50 mg/kg, ���������������������������������� respectively���������������������� ) �������������������� in addition to plas39 ma transfusion. Further studies are warranted to standardize TEG/ROTEM specific fibrinogen assays and to determine their clinical usefulness for guiding fibrinogen administration. Targeting antifibrinolytic therapy The CRASH-2 trial (Clinical Randomisation of Antifibrinolytic therapy in Significant Hemorrhage) 48 assessed the effects of early administration of tranexamic acid (TXA) on 28-day hospital mortality, vascular occlusive events, and transfusions in adult trauma patients. This trial randomized 20,211 patients with or at risk of significant bleeding to either TXA (loading dose 1 g over 10 minutes followed by infusion of 1 g over 8 hours) or placebo within 8 h of trauma. All-cause mortality was significantly reduced with TXA (1463 [14.5%] TXA vs. 1613 [16.0%] placebo; relative risk 0.91, 95% CI 0.85-0.97; P=0.0035), and the risk of death due to bleeding was also significantly reduced (489 [4.9%] vs. 574 [5.7%]; relative risk 0.85, 95% CI 0.76-0.96; P=0.0077). All cause mortality reduction was 1.5%, giving a number needed to treat (NNT) of 67 to save one life over 28 days. On the basis of the CRASH-2 trial results, it has been estimated that TXA use could save between 70,000 and 100,000 lives per year worldwide.49 Greatest impact of TXA on mortality was in patients with severe shock (systolic blood pressure [SBP]<75 mmHg).50 There was no difference in the rate of venous thrombotic events or stroke between groups, whereas there was a statistically significant reduced risk of myocardial infarction in the TXA group. Significant limitations of the CRASH-2 Trial have already been described: only 50% of study cohort received blood transfusion and TXA did not reduce RBC transfusion requirements; no data available regarding additional blood products (plasma, platelets) administered and whether damage-control resuscitation and/or MTP were used; fibrinolysis and coagulation assessments were not part of the study design; moreover, Vol. 82 - No. 11 TXA treatment given after 3 h after injury was associated with an increased risk of death caused by bleeding (4.4% vs. 3.1%; RR, 1.44; 95% CI, 1.12-1.84; p=0.004). For all these reasons, the mechanism by which TXA reduced mortality in the CRASH-2 trial remains unclear. Recently, studies evaluating the benefits of TXA administration in mature trauma systems with MTP protocols and early hemodynamic resuscitation have shown conflicting results.51-52 This calls for caution of indiscriminate use of antifibrinolytic drugs and raises the question of whether coagulation assessments should be performed prior to their administration. The traditional laboratory test for diagnosing pathologic fibrinolysis is the euglobulin lysis time (ELT) but it requires 4 hours to be prepared.50 Increased values of ROTEM LI30 or TEG LY30 can reflect hyperfibrinolysis. Thus, some authors have recommended using TEG or ROTEM to determine which patient to treat with TXA. However, definitions and thresholds values for pathologic fibrinolysis are not clearly defined. For example, TEG manufacturer reports the normal range of clot lysis at 30 min to be 0% to 7.5% whereas some authors use the cut-point of > 3% lysis since this value was associated with an increase in mortality.16,53 Interestingly, Moore et al. identified a Ushape distribution of death related to fibrinolysis in response to trauma, with an increased risk of death with level of fibrinolysis after 30 min above 2.9% and below 0.8%.54 The same team identified a particular “diamond-shaped” TEG tracing pattern (short time to MA and complete lysis before LY30 point) with 100% positive predictive value for mortality.55 First derivative velocity curve values generated by rTEG measuring lysis such as maximum rate of lysis (MRL) and total lysis (TL) were also described as a potential tool to define fibrinolysis.53 Another issue regarding the use of ROTEM or TEG to detect fibrinolysis is a possible lack of sensitivity to measure endogenous fibrinolytic activity. Indeed, Raza et al. showed that 5% of patients had TEM hyperfibrinolysis (defined as maximum clot lysis (ML) exceeding 15% of the maximum clot firmness) whereas 57% of these patients had evidence Minerva Anestesiologica 1225 FIGUEIREDOTARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMA of fibrinolysis according to plasmin-antiplasmin (PAP) complex and D-dimer levels.56 In summary, additional research is required to improve the ability of TEG/ROTEM to detect fibrinolysis, to standardize HF measurement and to determine whether these devices are useful to guide antifibrinolytic therapy in trauma patients. Pending further studies, TXA should be administered within 3 h after injury to the trauma patient who is bleeding or at risk of significant hemorrhage.39 Targeting whole blood transfusion? Whole blood (WB) was the primary resuscitation fluid in military settings through the beginning of the Vietnam War. After crucial advances in whole blood fractionation in the 1960s and the 1970s, blood donor centers began supplying hospitals with individual components (RBCs, plasma, PLT) and removed WB as a readily available product. Damage control resuscitation has recently emerged and promotes earlier and more aggressive use of plasma and PLT, in ratios approximating that of WB. This has renewed interest in the potential benefits of WB for some authors. A recent single-center randomized trial found that PLT-modified WB (MWB) was not superior to blood component therapy in reducing transfusion volumes or decreasing 24-hour and 30-day mortality in severely injured civilian patients predicted to receive massive transfusion. A benefit was seen only in a sensitivity analysis where patients with brain injury were excluded, where the use of MWB resulted in significantly reduced transfusion volumes compared to blood component therapy.57 However, many issues remain to be addressed: use of fresh warm or cooled WB, PLT-supplementation of WB requirement or not, maximal duration of WB storage? Targeting other hemostatic drugs: factor XIII, factor VIIa? Factor XIII (FXIII), which cross-links fibrin monomers, plays a key role in the stabilisation of the fibrin clot. Recent in vitro ROTEM studies using different models of hemodilution 1226 have shown that FXIII administration can reverse dilutional coagulopathy, although activated platelets and fibrinogen supplementation are required to observe this effect.58, 59 FXIII seems to exhibit some in vitro antifibrinolytic properties.60 Further studies with in vivo FXIII administration are needed to assess the clinical impact of FXIII supplementation in trauma settings. Thromboelastography can be used to monitor rFVIIa administration although there is no rFVIIa-specific parameter and moreover, recent European guidelines 39 suggest that the use of recombinant factor recombinant activated coagulation factor VII (rFVIIa) should be considered only if first-line treatment with a combination of surgical approaches, bestpractice use of blood products and antifibrinolytics, correction of acidosis, hypothermia and hypocalcemia fail to control bleeding. Limits of thromboelastography Although viscoelastic hemostatic assays (VHA) provide global information on clot formation and dissolution, these devices do not assess the endothelial contribution to hemostasis since an activator is directly added to initiate coagulation during the test.61 Consequently, diagnosis of conditions such as von Willebrand disease or endothelial dysfunction is not possible with VHAs. Additionally, platelet inhibition or dysfunction may not be identified with the standard assays, unless thrombin is inhibited and platelet agonists are utilized (TEGPlatelet Mapping assay for example). A recent study also showed that TEG results might be normal in patients receiving warfarin 62 and capacity of TEG to detect new oral anticoagulants remains to be determined in vivo.63 Some conditions have to be met for proper use of TEG/ROTEM as a POC device: the machines must be calibrated daily and, moreover, training and qualification must be put in place for anyone performing the test since running samples requires technical skills. Despite the material costs of all tests and devices, the use of VHAs has been proven to be cost-effective in cardiac surgery and in trauma settings.64, 65 Another limitation is the lack of standardiza- Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO tion of VHAs due to the considerable heterogeneity in methods, reagents and parameters measured. Indeed, TEG and ROTEM may not be fully equivalent, as the results provided by these devices do not seem to be interchangeable.66, 67 Different TEG/ROTEM-based transfusion algorithms for trauma patients have recently been proposed.14, 26, 28, 68 However, the data used to create these algorithms are often missing and no prospective validations or comparisons to other algorithms have been reported. Moreover, there is no consensus either on the threshold values that justify transfusion or on which hemostatic product should be administered for a given abnormal value (for example a low MA value on TEG may lead to PLT transfusion or desmopressin administration depending on which algorithm one is referring to). Finally, the lack of large and well-conducted RCTs assessing the real benefit of VHAs on patient outcomes represents the main limitation of these devices in trauma patients.69 Conclusions In trauma patients, TEG and ROTEM allow prediction of massive transfusion requirement and mortality, and some parameters show good correlation with the amount of blood products transfused. VHAs can identify at patient bedside which component(s) of the hemostatic process is (are) impaired and should be targeted: coagulation factors, fibrinogen, platelets and/or fibrinolysis. Lack of standardization among the different available devices/assays and the various therapeutic options (e.g. FFP or coagulation factors concentrates) limits the widespread use of VHAs-based coagulation algorithms. Goaldirected, individualized treatment algorithms may improve patient outcome, as it has been shown for cardiac surgery in prospective randomized trials. This outcome benefit remains to be clearly demonstrated in trauma settings. Key messages —— Viscoelastic hemostatic assays (VHAs), thromboelastography (TEG) and Vol. 82 - No. 11 rotational thromboelastometry (ROTEM), provide global and functional assessment of coagulation at patient bedside. —— In trauma patients, VHAs have been shown to predict massive transfusion and mortality. —— These devices allow goal-directed administration of hemostatic blood components (mainly coagulation factor concentrates such as fibrinogen concentrate) as an alternative to or in conjunction with transfusion protocols using fixed ratios of red blood cells, platelets and fresh frozen plasma. —— Standardization of the different TEG and ROTEM assays as well as high quality evidence of improvement in patient outcomes are mandatory to enable the widespread use of VHAs. References 1.Evans JA, van Wessem KJP, McDougall D, Lee KA, Lyons T, Balogh ZJ. Epidemiology of traumatic deaths: comprehensive population-based assessment. World J Surg 2010;34:158-63. 2.MacLeod JBA, Lynn M, McKenney MG, Cohn SM, Murtha M. Early coagulopathy predicts mortality in trauma. J Trauma 2003;55:39-44. 3.Maegele M, Lefering R, Yucel N, Tjardes T, Rixen D, Paffrath T, et al. Early coagulopathy in multiple injury: an analysis from the German Trauma Registry on 8724 patients. Injury 2007;38:298-304. 4.Brohi K, Singh J, Heron M, Coats T. Acute traumatic coagulopathy: The Journal of Trauma: injury, Infection, and Critical Care 2003;54:1127-30. 5.Roberts HR, Hoffman M, Monroe DM. A cell-based model of thrombin generation. Semin Thromb Hemost 2006;32(Suppl 1):32-8. 6.Toulon P, Ozier Y, Ankri A, Fléron M-H, Leroux G, Samama CM. Point-of-care versus central laboratory coagulation testing during haemorrhagic surgery. A multicenter study. Thromb Haemost 2009;101:394-401. 7. Holcomb JB, del Junco DJ, Fox EE, Wade CE, Cohen MJ, Schreiber MA, et al. The prospective, observational, multicenter, major trauma transfusion (PROMMTT) study: comparative effectiveness of a time-varying treatment with competing risks. JAMA Surg 2013;148:127-36. 8. Holcomb JB, Tilley BC, Baraniuk S, Fox EE, Wade CE, Podbielski JM, et al. Transfusion of plasma, platelets, and red blood cells in a 1:1:1 vs a 1:1:2 ratio and mortality in patients with severe trauma: the PROPPR randomized clinical trial. JAMA 2015;313:471-82. 9. Kaafarani HMA, Velmahos GC. Damage Control Resuscitation In Trauma. Scand J Surg 2014;103:81-8. 10. Whelihan MF, Mann KG. The role of the red cell membrane in thrombin generation. Thromb Res 2013;131:37782. 11. Du VX, Huskens D, Maas C, Al Dieri R, de Groot PG, de Minerva Anestesiologica 1227 FIGUEIREDOTARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMA Laat B. New insights into the role of erythrocytes in thrombus formation. Semin Thromb Hemost 2014;40:72-80. 12.Cotton BA, Faz G, Hatch QM, Radwan ZA, Podbielski J, Wade C, et al. Rapid Thrombelastography Delivers RealTime Results That Predict Transfusion Within 1 Hour of Admission: The Journal of Trauma: Injury, Infection, and Critical Care 2011;71:407-17. 13. Holcomb JB, Minei KM, Scerbo ML, Radwan ZA, Wade CE, Kozar RA, et al. ������������������������������ Admission rapid thrombelastography can replace conventional coagulation tests in the emergency department: experience with 1974 consecutive trauma patients. Ann Surg 2012;256:476-86. 14. Pezold M, Moore EE, Wohlauer M, Sauaia A, Gonzalez E, Banerjee A, et al. Viscoelastic clot strength predicts coagulation-related mortality within 15 minutes. Surgery 2012;151:48-54. 15.Ives C, Inaba K, Branco BC, Okoye O, Schochl H, Talving P, et al. Hyperfibrinolysis elicited via thromboelastography predicts mortality in trauma. J Am Coll Surg 2012;215:496-502. 16.Chapman MP, Moore EE, Ramos CR, Ghasabyan A, Harr JN, Chin TL, et al. Fibrinolysis greater than 3% is the critical value for initiation of antifibrinolytic therapy. J Trauma Acute Care Surg 2013;75:961-7;discussion 967. 17.Schöchl H, Cotton B, Inaba K, Nienaber U, Fischer H, Voelckel W, et al. FIBTEM provides early prediction of massive transfusion in trauma. Crit Care 2011;15:R265. 18. Davenport R, Manson J, De’Ath H, Platton S, Coates A, Allard S, et al. Functional definition and characterization of acute traumatic coagulopathy. Crit Care Med 2011;39:2652-8. 19.Nystrup, Kristin B, Nis A Windeløv, Annemarie B Thomsen, and Pär I Johansson, et al. Reduced clot strength upon admission, evaluated by thrombelastography (TEG), in trauma patients is independently associated with increased 30-day mortality. Scand J Trauma Resusc Emerg Med 2011;19:52. 20.Tauber H, Innerhofer P, Breitkopf R, Westermann I, Beer R, El Attal R, et al. Prevalence and impact of abnormal ROTEM(R) assays in severe blunt trauma: results of the “Diagnosis and Treatment of Trauma-Induced Coagulopathy (DIA-TRE-TIC) study.” Br J Anaesth 2011;107:37887. 21. Jeger V, Willi S, Liu T, Yeh DD, De Moya M, Zimmermann H, et al. The Rapid TEG α-Angle may be a sensitive predictor of transfusion in moderately injured blunt trauma patients. ScientificWorldJournal 2012;2012:821794. 22.Carroll RC, Craft RM, Langdon RJ, Clanton CR, Snider CC, Wellons DD, et al. Early evaluation of acute traumatic coagulopathy by thrombelastography. Transl Res 2009;154:34-9. 23. Wohlauer MV, Moore EE, Thomas S, Sauaia A, Evans E, Harr J, et al. Early platelet dysfunction: an unrecognized role in the acute coagulopathy of trauma. J Am Coll Surg 2012;214:739-46. 24.Schöchl H, Nienaber U, Hofer G, Voelckel W, Jambor C, Scharbert G, et al. Goal-directed coagulation management of major trauma patients using thromboelastometry (ROTEM)-guided administration of fibrinogen concentrate and prothrombin complex concentrate. Crit Care 2010;14:R55. 25.Schöchl H, Nienaber U, Maegele M, Hochleitner G, Primavesi F, Steitz B, et al. Transfusion in trauma: thromboelastometry-guided coagulation factor concentratebased therapy versus standard fresh frozen plasma-based therapy. Crit Care 2011;15:R83. 26.Tapia NM, Chang A, Norman M, Welsh F, Scott B, Wall MJ, et al. TEG-guided resuscitation is superior to standardized MTP resuscitation in massively transfused penetrating trauma patients. J Trauma Acute Care Surg 2013;74:378-85;discussion 385-6. 1228 27. Weber CF, Görlinger K, Meininger D, Herrmann E, Bingold T, Moritz A, et al. Point-of-care testing: a prospective, randomized clinical trial of efficacy in coagulopathic cardiac surgery patients. Anesthesiology 2012;117:53147. 28.Gonzalez E, Moore EE, Moore HB, Chapman MP, Chin TL, Ghasabyan A, et al. Goal-directed hemostatic resuscitation of trauma-induced coagulopathy: a pragmatic randomized clinical trial comparing a viscoelastic assay to conventional coagulation assays. Ann Surg 2016;263:1051-9. 29.Schöchl H, Voelckel W, Maegele M, Kirchmair L, Schlimp CJ. Endogenous thrombin potential following hemostatic therapy with 4-factor prothrombin complex concentrate: a 7-day observational study of trauma patients. Crit Care 2014;18:R147. 30. Moore EE, Chin TL, Chapman MC, Gonzalez E, Moore HB, Silliman CC, et al. Plasma first in the field for postinjury hemorrhagic shock. Shock 2014;41(Suppl 1):35-8. 31. Peng Z, Pati S, Potter D, Brown R, Holcomb JB, Grill R, et al. Fresh frozen plasma lessens pulmonary endothelial inflammation and hyperpermeability after hemorrhagic shock and is associated with loss of syndecan 1. Shock 2013;40:195-202. 32. Dekker SE, Sillesen M, Bambakidis T, Jin G, Liu B, Boer C, et al. Normal saline influences coagulation and endothelial function after traumatic brain injury and hemorrhagic shock in pigs. Surgery 2014;156:556-63. 33.Torres LN, Sondeen JL, Ji L, Dubick MA, Torres Filho I. Evaluation of resuscitation fluids on endothelial glycocalyx, venular blood flow, and coagulation function after hemorrhagic shock in rats. J Trauma Acute Care Surg 2013;75:759-66. 34.Stensballe J, Ostrowski SR, Johansson PI. Viscoelastic guidance of resuscitation. Curr Opin Anaesthesiol 2014;27:212-8. 35.Rossaint R, Bouillon B, Cerny V, Coats TJ, Duranteau J, Fernández-Mondéjar E, et al. The European guideline on management of major bleeding and coagulopathy following trauma: fourth edition. Crit Care 2016;20:100. 36.Inaba K, Karamanos E, Lustenberger T, Schöchl H, Shulman I, Nelson J, et al. Impact of fibrinogen levels on outcomes after acute injury in patients requiring a massive transfusion. J Am Coll Surg 2013;216:290-7. 37. Hagemo JS, Stanworth S, Juffermans NP, Brohi K, Cohen M, Johansson PI, et al. Prevalence, predictors and outcome of hypofibrinogenaemia in trauma: a multicentre observational study. Crit Care 2014;18:R52. 38.Rourke C, Curry N, Khan S, Taylor R, Raza I, Davenport R, et al. Fibrinogen levels during trauma hemorrhage, response to replacement therapy, and association with patient outcomes. J Thromb Haemost 2012;10:1342-51 39.Spahn DR, Bouillon B, Cerny V, Coats TJ, Duranteau J, Fernández-Mondéjar E, et al. Management of bleeding and coagulopathy following major trauma: an updated European guideline. Crit Care 2013;17:R76. 40. Meyer MAS, Ostrowski SR, Sørensen AM, Meyer ASP, Holcomb JB, Wade CE, et al. Fibrinogen in trauma, an evaluation of thrombelastography and rotational thromboelastometry fibrinogen assays. J Surg Res 2015;194:581-90. 41. Kornblith LZ, Kutcher ME, Redick BJ, Calfee CS, Vilardi RF, Cohen MJ. Fibrinogen and platelet contributions to clot formation: implications for trauma resuscitation and thromboprophylaxis. J Trauma Acute Care Surg 2014;76:255-6;discussion 262-3. 42.Solomon C, Sørensen B, Hochleitner G, Kashuk J, Ranucci M, Schöchl H. Comparison of whole blood fibrinbased clot tests in thrombelastography and thromboelastometry. Anesth Analg 2012;114:721-30. 43.Solomon C, Baryshnikova E, Schlimp CJ, Schöchl H, Minerva AnestesiologicaNovember 2016 TARGETING BLOOD PRODUCTS TRANSFUSION IN TRAUMAFIGUEIREDO Asmis LM, Ranucci M. FIBTEM PLUS provides an improved thromboelastometry test for measurement of fibrin-based clot quality in cardiac surgery patients. Anesth Analg 2013;117:1054-62. 44.Agren A, Wikman AT, Ostlund A, Edgren G. TEG® functional fibrinogen analysis may overestimate fibrinogen levels. Anesth Analg 2014;118:933-5. 45. Theusinger OM, Baulig W, Seifert B, Emmert MY, Spahn DR, Asmis LM. Relative concentrations of haemostatic factors and cytokines in solvent/detergent-treated and fresh-frozen plasma. Br J Anaesth 2011;106:505-11. 46.Schlimp CJ, Voelckel W, Inaba K, Maegele M, Schöchl H. Impact of fibrinogen concentrate alone or with prothrombin complex concentrate (+/- fresh frozen plasma) on plasma fibrinogen level and fibrin-based clot strength (FIBTEM) in major trauma: a retrospective study. Scand J Trauma Resusc Emerg Med 2013;21:74. 47. Maung AA, Kaplan LJ. Role of fibrinogen in massive injury. Minerva Anestesiol 2014;80:89-95. 48.CRASH-2 trial collaborators, Shakur H, Roberts I, Bautista R, Caballero J, Coats T, et al. Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a randomised, placebo-controlled trial. Lancet 2010;376:23-32. 49. Ker K, Kiriya J, Perel P, Edwards P, Shakur H, Roberts I. Avoidable mortality from giving tranexamic acid to bleeding trauma patients: an estimation based on WHO mortality data, a systematic literature review and data from the CRASH-2 trial. BMC Emerg Med 2012;12:3. 50.Napolitano LM, Cohen MJ, Cotton BA, Schreiber MA, Moore EE. Tranexamic acid in trauma: how should we use it? J Trauma Acute Care Surg 2013;74:1575-86. 51.Cole E, Davenport R, Willett K, Brohi K. Tranexamic acid use in severely injured civilian patients and the effects on outcomes: a prospective cohort study. Ann Surg 2015;261:390-4. 52. Harvin JA, Peirce CA, Mims MM, Hudson JA, Podbielski JM, Wade CE, et al. The impact of tranexamic acid on mortality in injured patients with hyperfibrinolysis. J Trauma Acute Care Surg 2015;78:905-9;discussion 90911. 53.Cotton BA, Harvin JA, Kostousouv V, Minei KM, Radwan ZA, Schöchl H, et al. Hyperfibrinolysis at admission is an uncommon but highly lethal event associated with shock and prehospital fluid administration. J Trauma Acute Care Surg 2012;73:365-70;discussion 370. 54. Moore HB, Moore EE, Gonzalez E, Chapman MP, Chin TL, Silliman CC, et al. Hyperfibrinolysis, physiologic fibrinolysis, and fibrinolysis shutdown: the spectrum of postinjury fibrinolysis and relevance to antifibrinolytic therapy. J Trauma Acute Care Surg 2014;77:811-7;discussion 817. 55.Chapman MP, Moore EE, Moore HB, Gonzalez E, Morton AP, Chandler J, et al. The “Death Diamond”: Rapid thrombelastography identifies lethal hyperfibrinolysis. J Trauma Acute Care Surg 2015;79:925-9. 56.Raza I, Davenport R, Rourke C, Platton S, Manson J, Spoors C, et al. The incidence and magnitude of fibrinolytic activation in trauma patients. J Thromb Haemost 2013;11:307-14. 57.Cotton BA, Podbielski J, Camp E, Welch T, del Junco D, Bai Y, et al. A randomized controlled pilot trial of modified whole blood versus component therapy in severely injured patients requiring large volume transfusions. Ann Surg 2013;258:527-32;discussion 532-3. 58.Schlimp CJ, Cadamuro J, Solomon C, Redl H, Schöchl H. The effect of fibrinogen concentrate and factor XIII on thromboelastometry in 33% diluted blood with albumin, gelatine, hydroxyethyl starch or saline in vitro. Blood Transfus 2013;11:510-7. 59. Kind SL, Spahn-Nett GH, Emmert MY, Eismon J, Seifert B, Spahn DR, et al. Is dilutional coagulopathy induced by different colloids reversible by replacement of fibrinogen and factor XIII concentrates? Anesth Analg 2013;117:1063-71.. 60. Dirkmann D, Görlinger K, Gisbertz C, Dusse F, Peters J. Factor XIII and tranexamic acid but not recombinant factor VIIa attenuate tissue plasminogen activator-induced hyperfibrinolysis in human whole blood. Anesth Analg 2012;114:1182-8. 61. Haas T, Görlinger K, Grassetto A, Agostini V, Simioni P, Nardi G, et al. Thromboelastometry for guiding bleeding management of the critically ill patient: a systematic review of the literature. Minerva Anestesiol 2014;80:132035. 62. Dunham CM, Rabel C, Hileman BM, Schiraldi J, Chance EA, Shima MT, et al. TEG® and RapidTEG® are unreliable for detecting warfarin-coagulopathy: a prospective cohort study. Thromb J 2014;12:4. 63. Dias JD, Norem K, Doorneweerd DD, Thurer RL, Popovsky MA, Omert LA. Use of Thromboelastography (TEG) for Detection of New Oral Anticoagulants. Arch Pathol Lab Med 2015;139:665-73. 64.Whiting P, Al M, Westwood M, Ramos IC, Ryder S, Armstrong N, Misso K, Ross J, Severens J, Kleijnen J. Viscoelastic point-of-care testing to assist with the diagnosis, management and monitoring of haemostasis: a systematic review and cost-effectiveness analysis. Health Technol Assess 2015;19:1-228. 65.Nardi G, Agostini V, Rondinelli B, Russo E, Bastianini B, Bini G, et al. Trauma-induced coagulopathy: impact of the early coagulation support protocol on blood product consumption, mortality and costs. Crit Care 2015;19, 83. 66.Venema LF, Post WJ, Hendriks HGD, Huet RCG, de Wolf JTW, de Vries AJ. An assessment of clinical interchangeability of TEG and RoTEM thromboelastographic variables in cardiac surgical patients. Anesth Analg 2010;111:339-44. 67.Sankarankutty A, Nascimento B, Teodoro da Luz L, Rizoli S. TEG® and ROTEM® in trauma: similar test but different results? World J Emerg Surg 2012;22;7(Suppl 1):S3. 68. Kashuk JL, Moore EE, Wohlauer M, Johnson JL, Pezold M, Lawrence J, et al. Initial experiences with pointof-care rapid thrombelastography for management of life-threatening postinjury coagulopathy. Transfusion 2012;52:23-33. 69. Hunt H, Stanworth S, Curry N, Woolley T, Cooper C, Ukoumunne O, et al. Thromboelastography (TEG) and rotational thromboelastometry (ROTEM) for trauma induced coagulopathy in adult trauma patients with bleeding. Cochrane Database Syst Rev 2015;CD010438. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: September 8, 2016. - Manuscript accepted: September 7, 2016. - Manuscript revised: August 30, 2016. Manuscript received: January 30, 2016. Vol. 82 - No. 11 Minerva Anestesiologica 1229 © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1230-4 JOURNAL CLUB CRITIQUE Failure of statins in ARDS: the quest for the Holy Grail continues David GRIMALDI, Arthur DURAND, James GLEESON, Fabio S. TACCONE * Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium *Corresponding author: Fabio S. Taccone, Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium. E‑mail: [email protected] A B STRACT Experimental and clinical observational studies have shown potential benefits of statin administration in the acute respiratory distress syndrome (ARDS) by modulating inflammation and preventing worsening respiratory function. More recently, two randomized clinical trials failed to demonstrate an improved survival of ARDS patients treated with statins. In the first study, conducted by the ARDS Network, 745 patients with sepsis‑associated ARDS were randomized within 48-hours of onset to receive either rosuvastatin or placebo. There was no significant difference between the rosuvastatin and placebo groups for hospital mortality (primary outcome, 29% vs. 25%, P=0.21) or ventilator‑free days (15±11 vs. 15±11, respectively; P=0.96). In rosuvastatin‑treated patients, renal and hepatic failure free‑days were significantly lower than in the placebo group, raising serious safety concerns. In the second study (HARP-2 trial), 540 patients with ARDS were randomized within 48-hours of onset to receive either simvastatin (80 mg/day) or placebo. There was no significant difference between the study groups for number of ventilator‑free days (primary outcome, 13±10 in the simvastatin vs. 12±10 in the placebo group, P=0.21) or 28-day mortality (22% vs. 27%, respectively; P=0.23). No significant difference in serious adverse events was reported between groups. Herein, we discuss the main reasons for these negative findings and consider where there could be a role for statins in ARDS patients. (Cite this article as: Grimaldi D, Durand A, Gleeson J, Taccone FS. Failure of statins in ARDS: the quest for the Holy Grail continues. Minerva Anestesiol 2016;82:1230-4) Key words: Adult respiratory distress syndrome - Hydroxymethylglutaryl‑CoA reductase inhibitors - Patient outcome assessment - Randomized controlled trial. A cute respiratory distress syndrome (ARDS) is a major cause of morbidity and mortality in the intensive care unit.1 ARDS is characterized by protein‑rich pulmonary edema due to alveolar‑capillary barrier injury caused by overwhelming inflammation.2 In particular, sepsis syndrome, either due to pneumonia or from an extra‑pulmonary source, is the leading cause of ARDS over all other inflammatory conditions. Protective ventilatory management, using low tidal volume and high positive end‑expiratory pressure ventilation to lower driving pressure, together with restrictive fluid management after hemodynamic sta- 1230 bilization, are the most effective therapeutic interventions to reduce length of mechanical ventilation and mortality in ARDS patients.3-5 To date, no targeted pharmacological therapy has shown substantial beneficial effects in this disease. Given the pathophysiology of ARDS, the modulation of the inflammatory response is an attractive and logical strategy. In particular, statins, which inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG‑CoA) reductase and are commonly used to treat dyslipidemia, have pleiotropic effects, including antioxidant, anti‑inflammatory and endothelial‑protective Minerva AnestesiologicaNovember 2016 STATINS AND ARDSGRIMALDI actions.6 These properties led to the hypothesis that statins could improve lung function and survival in ARDS patients. Several animal studies showed that statins were able to prevent experimentally‑induced ARDS.7, 8 For example, in mice receiving an intra‑tracheal administration of endotoxin, pre‑treatment with simvastatin decreased the severity of histological lung injury and alveolar‑capillary leakage.7 These results were later confirmed in a small cohort of healthy volunteers, who received oral simvastatin for 4 days before endotoxin inhalation.9 Moreover, observational data showed an association between previous statin treatment and a better outcome in ARDS and septic patients.10, 11 In a randomized phase‑II trial, Craig et al. observed that treatment with simvastatin (N.=30) decreased biomarkers of inflammation measured in the broncho‑alveolar lavage fluid of patients with ARDS without significant adverse events.12 Taken together, these promising data prompted the design of two large randomized multicenter studies (the SAIL study from the ARDS Network using rosuvastatin and the HARP-2 study using simvastatin) aiming to demonstrate a clinical benefit of statin treatment in ARDS patients.13, 14 Discussion Both trials failed to demonstrate a clinical benefit of statin administration during ARDS on mortality or ventilator free‑days; these negative results were in line with those from another recent study on simvastatin as an adjunctive therapy for ventilator‑associated pneumonia (VAP).15 A summary of the studies evaluating statin‑therapy in critically ill patients with severe respiratory failure is shown in Table I. Importantly, some issues deserve further discussion when interpreting these results. The biological rationale for using statins in ARDS is based on the presence of an acute systemic inflammatory response, which is enhanced by an increased pulmonary production of TNF-α and release of matrix metalloproteinases (MMPs), by the alteration of endothelium and leucocyte translocation.9 Indeed, damaged pulmonary vascular endothelium and Vol. 82 - No. 11 alveolar accumulation of neutrophils are histological hallmarks of ARDS.16 In pre‑clinical studies where statins have been shown to be protective, they were administered before the inflammatory insult, which could have given the drug a chance to “prepare” the endothelium. Furthermore, experimental lung‑injury models, such as those using an endotoxin challenge, do not mimic the full effects of a real infection neither reproduce the same inflammatory status. The choice of a hydrophilic statin (e.g. rosuvastatin) by the SAIL investigators, claiming a higher free drug‑fraction and less adverse effects, compromised the study as there is a lack of significant pre‑clinical data on this molecule in the ARDS setting. Also, it still remains unclear whether all statins have the same immune‑modulatory effects. Most pre‑clinical trials working on the anti‑inflammatory effects of these drugs in sepsis or in ARDS used lipophilic statins, such as atorvostatin or simvastatin.7, 9 Lipophilic molecules are known to have greater tissue penetration; moreover lipophilic statins demonstrate a stronger antibacterial effect.17 When the authors measured rosuvastatin concentrations in a subgroup of 404 patients, mean peak concentrations reached 7.3 ng/mL, which were largely below target drug‑levels (10-70 ng/mL) able to provide significant immune‑modulatory effects. These results could be explained by the use of a moderate daily dose (20 mg), similar to chronic therapy given in healthy subjects; however this therapeutic strategy did not take into account the altered drug pharmacokinetics recognised in septic patients, which may lead to inadequate concentrations in the early phase of therapy if standard regimens are used.18 In the HARP-2 study,14 the authors evaluated simvastatin at a higher dose than in chronic conditions, which had been previously shown to provide adequate concentrations in critically‑ill patients.12 Furthermore, no significant difference in ventilator‑free days and mortality was shown between groups, although drug levels were not specifically assessed. Neither trial considered the severity of ARDS in the inclusion criteria and mean Minerva Anestesiologica 1231 GRIMALDISTATINS AND ARDS Table I.—Clinical studies on statins and ARDS. Study Targeted pathology Methods Inclusion Criteria SAIL 13 Prospective Randomized Double‑blinded Placebo‑controlled Multicenter 100% sepsis Rosuvastatin 71% pneumonia (40 mg loading PaO2/ dose, 20 mg FiO2=170±70 daily) on inclusion* HARP-2 14 Prospective Randomized Double‑blinded Placebo‑controlled Multicenter ARDS<48 hours PaO2/FiO2<300 Sepsis (SIRS criteria) No treatment with statin in the last 48 hours ARDS<48 hours PaO2/FiO2<300 No treatment with statin in the last 2 weeks Papazian et al.15 Prospective Randomized Double‑blinded Placebo‑controlled Multicenter 100% pneumonia PaO2/FiO2=189 [140-240] on inclusion Simvastatin (60 mg daily) HARP 12 Prospective Randomized Double‑blinded Placebo‑controlled First VAP episode No PaO2/FiO2 criteria No treatment with statin since intubation ARDS PaO2/FiO2<300 PaO2/ FiO2=170±55 on inclusion* Simvastatin (80 mg daily) Primary outcome Intervention 42% sepsis Simvastatin 58% pneumonia (80 mg daily) PaO2/ FiO2=130±50 on inclusion* Results Hospital mortality N.=745 No effects on: –– Mortality –– MV‑free days –– CRP levels Less days free of hepatic or renal failure MV‑free days N.=540 (28 days) No effects on: –– Mortality –– MV‑free days –– CRP levels –– Non‑pulmonary organ failure 28-day N.=284 mortality No effects on: –– Mortality –– MV‑free days Physiological N.=60 and –– Decreased biological pulmonary outcomes inflammation –– Decrease CRP –– No increase in adverse events MV: mechanical ventilation; CRP: C‑reactive protein; VAP: ventilator‑associated pneumonia. *PaO2/FiO2 data were approximated by considering the two study groups (statins vs. placebo) altogether. PaO2/FiO2 ratios vary considerably across patients included in these studies. It would, however, make sense that the benefit of statins, if any, would be in patients with the greatest inflammation e.g. those with the most severe hypoxemia, as suggested by a recent observational study.19 Moreover, ARDS is not a well‑characterized disease but is rather a syndrome caused by numerous conditions; the HARP studies enrolled patients with ARDS whatever the cause, and the SAIL study enrolled sepsis‑related ARDS patients corresponding to a wide variety of conditions. It is reasonable to think that a given pharmacological intervention may be beneficial only in specific sub‑groups of ARDS patients while being detrimental in others, resulting in high statistical‑noise in large trials. As an example, patients de- 1232 veloping ARDS secondary to a VAP should be potentially excluded as in this particular condition, simvastatin has already been shown to be ineffective.15 Another important limitation is the possibility that some of the included patients were suffering from acute cardiogenic pulmonary edema rather than ARDS, as sometimes the diagnosis can be clinically challenging. Although in both studies the investigators cited elevated left atrial pressures as an exclusion criterion, it was not clearly indicated if or how this was assessed. Thus, the inclusion of non‑ARDS patients may have further diluted any effect of statins in this setting. Once again, disappointment arises from large clinical studies despite promising results in animal data, illustrating the difficulty to move from experimental studies to clinical Minerva AnestesiologicaNovember 2016 STATINS AND ARDSGRIMALDI outcomes.20 Overenthusiasm about suggestive results from observational studies needs to be tempered by the risk of overlooking unmeasured confounders. As an example, the trend towards statin protection in a prospective observational study reported by the Irish Critical Care Trials group did not reach statistical significance, even after multivariate analysis.12 Moreover, one potential confounder that has not been measured in recent observational statin/ARDS studies is the socio‑economic status, e.g. patients from lower socio‑economic groups are less likely to receive appropriate primary health care and are less likely to be taking statins than others.21 Finally, the concomitant prescription of macrolide antibiotics in these patients, which can inhibit statin metabolism and increase blood concentrations, went also unaccounted in these observational studies. The last point to underline is the fact that patients pre‑treated with statins were excluded from the trials (Table I). This is in contrast to the statin dosing approach taken in pre‑clinical and clinical observational studies. In the SAIL study, a pre‑specified small subgroup analysis of patients who previously took statins, but not during the 48 hours prior to enrolment, showed that continuation of therapy was not associated with a clinical benefit. However, as conflicting results have been reported in septic patients,22 it seems reasonable to pursue a statin treatment unless there is renal or hepatic failure (exclusion criteria in all trials), concern about rhabdomyolysis or the development of another recognized side‑effect. When considering the potential advantage of continuing a drug in critically ill patients it is also important to consider the potential negative impact of withdrawing the drug. Indeed, stopping statins in at‑risk hospitalized patients increases their risk of a cardiac event.23 The tolerance of statins in critically ill patients is uncertain, and few data exist. In the HARP and HARP-2 studies, simvastatin therapy appears to be well tolerated; however, in the SAIL study, rosuvastatin was associated with an increase duration of renal and hepatic dysfunction. Clinical relevance of these adverse ef- Vol. 82 - No. 11 fects needs to be clarified, but these safety concerns plead against the use of higher doses of rosuvastatin. Furthermore, statins may worsen sepsis‑induced immune dysfunction 24 and then increase the risk of ICU‑acquired infection. Conclusions In two prospective randomized trials, statin administration in the early inflammatory phase of ARDS was not associated with improved 60-day mortality. Statins did not show any benefit in ventilator‑free days or in reducing inflammatory biological markers. To date, statin treatment cannot be recommended in ARDS patients. Key messages —— Early statin therapy in patients with ARDS was not associated with any benefit on mortality or ventilator‑free days. —— Continuation of statin therapy in patients with early ARDS may be beneficial. —— Statins use in critically ill patients may induce renal and hepatic failure. —— The use of statin in other cause of sepsis remains to be studied by randomized trials. References 1. Schell‑Chaple HM, Puntillo KA, Matthay MA, Liu KD; National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome Network. Body temperature and mortality in patients with acute respiratory distress syndrome. Am J Crit Care 2015;24:15-23. 2. Fanelli V, Ranieri VM. Mechanisms and clinical consequences of acute lung injury. Ann Am Thorac Soc 2015;12(Suppl 1):S3-8. 3.The Acute Respiratory Distress Syndrome Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 2000;342:1301-8. 4.National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network: Wiedemann HP, Wheeler AP, Bernard GR, Thompson BT, Hayden D, deBoisblanc B, et al. Comparison of two fluid‑management strategies in acute lung injury. N Engl J Med 2006;354:2564-75. 5.Amato MB, Meade MO, Slutsky AS, Brochard L, Costa EL, Schoenfeld DA, et al. Driving pressure and survival in the acute respiratory distress syndrome. N Engl J Med 2015;372:747-55. Minerva Anestesiologica 1233 GRIMALDISTATINS AND ARDS 6. Ito MK, Talbert RL, Tsimikas S. Statin‑associated pleiotropy: possible beneficial effects beyond cholesterol reduction. Pharmacotherapy 2006;26:85S-97S. 7. Jacobson JR, Barnard JW, Grigoryev DN, Ma SF, Tuder RM, Garcia JG. Simvastatin attenuates vascular leak and inflammation in murine inflammatory lung injury. Am J Physiol Lung Cell Mol Physiol 2005;288:L1026-32. 8. Yao HW, Mao LG, Zhu JP. Protective effects of pravastatin in murine lipopolysaccharide‑induced acute lung injury. Clin Exp Pharmacol Physiol 2006;33:793-7. 9. Shyamsundar M, McKeown ST, O’Kane CM, Craig TR, Brown V, Thickett DR, et al. Simvastatin decreases lipopolysaccharide‑induced pulmonary inflammation in healthy volunteers. Am J Respir Crit Care Med 2009;179:1107-14. 10.Irish Critical Care Trials Group. Acute lung injury and the acute respiratory distress syndrome in Ireland: a prospective audit of epidemiology and management. Crit Care 2008;12:R30. 11. Mansur A, Steinau M, Popov AF, Ghadimi M, Beissbarth T, Bauer M, et al. Impact of statin therapy on mortality in patients with sepsis‑associated acute respiratory distress syndrome (ARDS) depends on ARDS severity: a prospective observational cohort study. BMC Med 2015;13:128. 12.Craig TR, Duffy MJ, Shyamsundar M, McDowell C, O’Kane CM, Elborn JS, et al. A randomized clinical trial of hydroxymethylglutaryl- coenzyme a reductase inhibition for acute lung injury (The HARP Study). Am J Respir Crit Care Med 2011;183:620-6. 13.National Heart, Lung, and Blood Institute ARDS Clinical Trials Network, Truwit JD, Bernard GR, Steingrub J, Matthay MA, Liu KD, Albertson TE, et al. Rosuvastatin for sepsis‑associated acute respiratory distress syndrome. N Engl J Med 2014;370:2191-200. 14. McAuley DF, Laffey JG, O’Kane CM, Perkins GD, Mullan B, Trinder TJ, et al.; HARP-2 Investigators; Irish Critical Care Trials Group. Simvastatin in the acute respiratory distress syndrome. N Engl J Med 2014;371:1695703. 15. Papazian L, Roch A, Charles PE, Penot‑Ragon C, Perrin G, Roulier P, et al.; STATIN‑VAP Study Group. Effect of statin therapy on mortality in patients with ventilator‑associated pneumonia: a randomized clinical trial. JAMA 2013;310:1692-700. 16. Preira P, Forel JM, Robert P, Nègre P, Biarnes‑Pelicot M, Xeridat F, et al. The leukocyte‑stiffening property of plasma in early acute respiratory distress syndrome (ARDS) revealed by a microfluidic single‑cell study: the role of cytokines and protection with antibodies. Crit Care 2016;20:8. 17. Masadeh M, Mhaidat N, Alzoubi K, Al‑Azzam S, Alnasser Z. Antibacterial activity of statins: a comparative study of atorvastatin, simvastatin, and rosuvastatin. Ann Clin Microbiol Antimicrob 2012;11:13. 18. Kruger PS, Freir NM, Venkatesh B, Robertson TA, Roberts MS, Jones M. A preliminary study of atorvastatin plasma concentrations in critically ill patients with sepsis. Intensive Care Med 2009;35:717-21. 19. Mansur A, Steinau M, Popov AF, Ghadimi M, Beissbarth T, Bauer M, Hinz J. Impact of statin therapy on mortality in patients with sepsis‑associated acute respiratory distress syndrome (ARDS) depends on ARDS severity: a prospective observational cohort study. BMC Med 2015;13:128. 20. Perrin S. Preclinical research: Make mouse studies work. Nature 2014;507:423-5. 21. Wallach‑Kildemoes H, Andersen M, Diderichsen F, Lange T. Adherence to preventive statin therapy according to socioeconomic position. Eur J Clin Pharmacol 2013;69:1553-63. 22. Thomas G, Hraiech S, Loundou A, Truwit J, Kruger P, Mcauley DF, et al. Statin therapy in critically‑ill patients with severe sepsis: a review and meta‑analysis of randomized clinical trials. Minerva Anestesiol 2015;81:92130. 23. Heeschen C, Hamm CW, Laufs U, Snapinn S, Böhm M, White HD; Platelet Receptor Inhibition in Ischemic Syndrome Management (PRISM) Investigators. Withdrawal of statins increases event rates in patients with acute coronary syndromes. Circulation 2002;105:1446-52. 24. Monneret G, Venet F. Statins and sepsis: do we really need to further decrease monocyte HLA‑DR expression to treat septic patients? Lancet Infect Dis 2007;7:697-9. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: May 27, 2016. - Manuscript accepted: May 25, 2016. - Manuscript revised: May 3, 2016. - Manuscript received: January 29, 2016. 1234 Minerva AnestesiologicaNovember 2016 LETTERS TO THE EDITOR © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it Minerva Anestesiologica 2016 November;82(11):1235-6 High-sensitivity troponin and extubation failure after successful spontaneous breathing trial Dear Editor, Myocardial dysfunction is one of the causes of failure to wean from mechanical ventilator support in intensive care. A multitude of clinical, biological and echocardiographic risk factors are associated with weaning failure linked to myocardial dysfunction. However, weaning failure must be differentiated from extubation failure, as the causes of failure are different.1 The incidence of extubation failure varies from 10-20% and is defined as the need for reintubation within 24-72 hours after properly conducted weaning.1 This failure is associated with increased morbidity and mortality. Numerous clinical risk factors are associated with extubation failure.2 The value of considering troponin levels in this context is a question that has already been asked.3 Weaning from mechanical ventilator support increases myocardial effort, respiratory effort, the myocardial oxygen demand and adrenergic stress.3 These conditions contribute to an imbalance between myocardial oxygen supply and demand. So, silent myocardial ischemia could occur during weaning.3 The dosage of high-sensitivity troponin (hs-troponin assays) allows hitherto undetectable troponin levels to be detected as well as faster variations. Our hypothesis is that an increase in hs-troponin during weaning is a sign of subclinical myocardial damage and could be related to early reintubation. We conducted a single-center study approved by the research ethics committee (Comité de Protection des Personnes SUD-EST IV) between February 2014 and September 2015. We prospectively included patients who were on ventilation for more than 48 hours and were extubated after successful respiratory weaning, Spontaneous breathing trial lasts actually one hour. The extubation was considered a failure if the patient was reintubated within 72 hours. Hs-troponin was measured within two hours before weaning (T1) and one hour after weaning (T2). The absolute value of this biomarker as well as its evolution between T1 and T2 were recorded (progression = [T2–T1]/T1). Seventy patients were included in the study. Twelve patients (17%) were reintubated within 72 hours. Both groups were not statistically different in terms of SAPS II score at admission, age, sex, number of days on ventilation before extubation and cardiac disease (Table I). In univariate analysis, troponin level at T1, at T2 or its variation between T1 and T2 were not significantly different between the group of patients with or without extubation failure at 72 hours. BNP was the only biomarker associated with a risk of reintubation after properly conducted weaning.4 In the context of myocardial ischemia, a significant increase in hs-troponin can be observed during the first hour.5 Spontaneous breathing trial could be considered as a cardio-pulmonary stress test. Despite a pathophysiological rationale for hs-troponin measure in the context of weaning, this study suggests that hs-troponin may not predict extubation failure. These results must Table I.—Clinical and biological characteristics. Age (years) SAPS II at admission Ventilation (days) Male sex Anterior cardiac disease Hs-Troponin (ng/L) Before weaning trial After weaning trial Progression (%) Entire cohort (N.=70) Success of weaning (N.=58) Failure of weaning (N.=12) 68±13 63±17 11.5±8.0 64% 32 (46%) 68±13 61±16 10.5±7.0 62% 25 (43%) 69±13 72±17 15.5±9.5 75% 8 (67%) 0.69* 0.053** 0.08** 0.39 0.14 196±602 184±593 -2.4±17.2 214±658 201±647 -1.8±18.2 110±170 102±159 -5.4±11.6 0.46** 0.49** 0.48** P value Values are expressed as mean±SD or as N. (%). Progression = (T2–T1)/T1 SAPS: Simplified Acute Physiologic Score. *t-test;**Mann-Whitney test. Vol. 82 - No. 11 Minerva Anestesiologica 1235 LETTERS TO THE EDITOR be interpreted with caution owing to the limited number of patients. Nicolas MOTTARD 1 *, Pauline RENAUDIN 1, Florent WALLET 1, Fabrice THIOLLIÈRE 1, Julien BOHE 1, 2, Arnaud FRIGGERI 1 1Service of Reanimation, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France; 2Université Claude-Bernard, Lyon, France *Corresponding author: Nicolas Mottard, Service de Réanimation, Centre Hospitalier Lyon-Sud, 165 chemin du grand Revoyet, 69495 Pierre Bénite, France. E-mail: [email protected] References 1.Epstein SK. Decision to extubate. Intensive Care Med 2002;28:535-46. 2.Sellarés J, Ferrer M, Torres A. Predictors of weaning after acute respiratory failure. Minerva Anestesiol 2012;78:1046-53. 3.Teboul J-L. Weaning-induced cardiac dysfunction: where are we today? Intensive Care Med 2014;40:1069-79. 4.Chien J-Y, Lin M-S, Huang Y-CT, Chien Y-F, Yu C-J, Yang P-C. Changes in B-type natriuretic peptide improve weaning outcome predicted by spontaneous breathing trial. Crit Care Med 2008;36:1421-6. 5.Reichlin T, Irfan A, Twerenbold R, Reiter M, Hochholzer W, Burkhalter H, et al. Utility of absolute and relative changes in cardiac troponin concentrations in the early diagnosis of acute myocardial infarction. Circulation 2011 Jul;124:136-45. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: May 27, 2016. - Manuscript accepted: May 25, 2016. - Manuscript revised: May 10, 2016. Manuscript received: January 5, 2016. (Cite this article as: Mottard N, Renaudin P, Wallet F, Thiollière F, Bohe J, Friggeri A. High-sensitivity troponin and extubation failure after successful spontaneous breathing trial. Minerva Anestesiol 2016;82:1235-6) © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it ;(): ;():;():(): ): Minerva Anestesiologica 2016 November;82(11):1236-7 Posterior reversible encephalopathy syndrome in acute pancreatitis Dear Editor, We report a case of posterior reversible encephalopathy syndrome (PRES) during the hospitalization of acute pancreatitis. 1236 PRES is clinical and radiologic syndrome characterized by a constellation of clinical signs and symptoms including headache, altered mental status, visual change, seizures and occasionally focal neurologic signs. Neuroimaging is essential to the diagnosis: typical findings are symmetrical white matter edema in the posterior cerebral hemispheres, particularly the parieto-occipital regions, most often visualized on magnetic resonance imaging.1 Many conditions could be associated with PRES development such as hypertension, preeclampsia, renal disease, sepsis or autoimmune diseases. A 73-year-old man with a medical history of hypertension, chronic ischemic heart disease and diabetes mellitus type 2 was first admitted with acute pancreatitis caused by gallstones. Endoscopic retrograde cholangiopancreatography with papillosphincterotomy were performed providing him with an overall wellness. Few days later, the patient showed a rapidly decreasing of consciousness and generalized seizures. After resolution of the seizures, a cerebral computed tomography (CT) scan was performed and the patient was admitted to Intensive Care Unit (ICU). Upon his arrival at the ICU, the patient was unconscious and able to localize painful stimuli without neurological deficits. He had uncontrolled hypertension, with systolic blood pressure more than 200 mmHg. The cerebral CT scan showed absence of ischemic or hemorrhagic lesions. Routine laboratory and cerebrospinal fluid parameters were normal. Cerebral magnetic resonance imaging was finally performed, showing areas of altered signal at the level of midbrain, cerebellum and posterior regions; no basilar artery occlusion was observed (Figure 1). With the achievement of blood pressure control, the neurologic function improved in few days, then the patient was discharged from the ICU in absence of neurological deficits. Follow-up MR imaging was performed two weeks later showing normal findings. The clinical features and the MR imaging findings of the reported case are strongly suggestive of PRES. Imaging is essential for the differential diagnosis of PRES, which includes strokes, cerebral venous thrombosis and encephalitis.2 The pathophysiologic mechanism of PRES is unknown, although it would result from a dysfunction of the cerebrovascular auto-regulatory mechanism due to hypertension. Severe acute pancreatitis is often associated with capillary leak syndrome and an increasing in endothelial permeability. These changes in vascular permeability could be the link between acute pancreatitis and PRES. Altered cerebral microcirculation is also the main mechanism underneath sepsis-related brain dysfunction which frequently shows clinical and radiological signs of PRES.³ The preferential involvement of the parietal and occipital lobes is thought to be related to the relatively poor sympathetic innervation of the vertebrobasilar circulation. In patients with PRES, the myogenic component of autoregulatory mechanism is blunted by passive Minerva Anestesiologica November 2016 LETTERS TO THE EDITOR Figure 1.—A T2-weighted MRI image of the patient shows bilateral, slightly asymmetric, multifocal T2 hyperintensities in the posterior temporo-occipital subcortical white matter. overdistension of the vessel due to elevations in blood pressure. Thus, autoregulation is strictly dependent on the neurogenic response: the more poorly innervated areas in the posterior circulation are most vulnerable. Prompt lowering of blood pressure, treatment of associated seizures and removal of causative agent are recommended in the management of PRES. The prognosis is usually benign with complete reversal of clinical symptoms within several days, when adequate treatment is immediately initiated. In the literature only few cases of association between acute pancreatitis and PRES with documentation of cerebral vasculopathy has been reported. In 2008 Shen et al.4 described a case of PRES after acute pancreatitis in a patient affected by acute intermittent porphyria. Later, Yamada et al.5 reported a case of recurrent PRES in a pediatric patient with pancreatitis subsequent to nephrotic syndrome. In all these cases posterior reversible encephalopathy was completely recovered without any neurological sequelae because of the early recognition of the symptoms and timely treatment. Christian COMPAGNONE, Daniele BELLANTONIO *, Federica PAVAN, Fernanda TAGLIAFERRI, Maria BARBAGALLO, Guido FANELLI Department of Anesthesia, Intensive Care and Pain Therapy, University Hospital of Parma, Parma, Italy Vol. 82 - No. 11 *Corresponding author: Daniele Bellantonio, Department of Anesthesia, Intensive Care and Pain Therapy, University Hospital of Parma, via Gramsci 14, Parma, Italy. E-mail: [email protected]. References 1. Fugate JE, Claassen DO, Cloft HJ, Kallmes DF, Kozak OS, Rabinstein AA. Posterior reversible encephalopathy syndrome: associated clinical and radiologic findings. Mayo Clin Proc 2010;85:427-32. 2. Bartynski WS. Posterior reversible encephalopathy syndrome, part 1: fundamental imaging and clinical features. AJNR Am J Neuroradiol 2008;29:1036-42. 3.Oddo M, Taccone FS. How to monitor the brain in septic patients? Minerva Anestesiol 2015;81:776-88. 4.Shen FC, Hsieh CH, Huang CR, Lui CC, Tai WC, Chuang YC. Acute intermittent porphyria presenting as acute pancreatitis and posterior reversible encephalopathy syndrome. Acta Neurol Taiwan 2008;17:177-83. 5. Yamada A, Atsumi M, Tashiro A, Hiraiwa T, Ueda N. Recurrent posterior reversible encephalopathy syndrome in nephrotic syndrome: case report and review of the literature. Clin Nephrology 2012;78:406-11. Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: June 7, 2016. - Manuscript accepted: June 6, 2016. - Manuscript revised: May 31, 2016. Manuscript received: March 20, 2016. (Cite this article as: Compagnone C, Bellantonio D, Pavan F, Tagliaferri F, Barbagallo M, Fanelli G. Posterior reversible encephalopathy syndrome in acute pancreatitis. Minerva������� �������������� Anestesiol 2016;82:1236-7) Minerva Anestesiologica 1237 LETTERS TO THE EDITOR © 2016 EDIZIONI MINERVA MEDICA Online version at http://www.minervamedica.it ;(): ;():;():(): ): Minerva Anestesiologica 2016 November;82(11):1238-9 Successful treatment of life-threatening hemorrhaging due to amniotic fluid embolism Dear Editor, The current report of a successful treatment of a case of amniotic fluid embolism (AFE) is of importance to the readership of this Journal because AFE is a rare but catastrophic event with a high mortality for both the mother and the fetus.1 Although the pathophysiology remains uncertain, an anaphylactic-type reaction is the most likely initiating factor. Unfortunately, AFE cannot be prevented and the diagnosis is based on the patient’s clinical presentation (and is essentially one of exclusion). During the second stage of labor, a healthy 39-year-old woman developed acute severe hypotension (systolic blood pressure [SBP]<30 mmHg), cyanosis (SpO2<70%), altered mental status (Glasgow Coma Scale [GCS] Score of 6 -opened her eyes in response to painful stimuli, made no verbal sounds, and displayed abnormal reflex responses to painful stimuli) and severe fetal bradycardia (HR of 40 bpm). A presumptive diagnosis of AFE was made and the patient was immediately transferred to the operating theatre for an emergency caesarean delivery. Following the extraction of the placenta, the patient developed uterine atony accompanied by severe haemorrhage which failed to respond to the infusion of uterotonic drugs (namely, oxytocin 20 and 10 mg IV bolus injections and sulprostone 8.3 ug/min IV) necessitating an emergency hysterectomy. The patient remained hypotensive with a SBP=70 mmHg, MAP=43 mmHg, DBP=30 mmHg despite aggressive fluid replacement (2 L of warmed crystalloid), intraoperative blood salvage reinfusion (washed red cells, 485 mL),2 packed red blood cells (5 units) and of fresh frozen plasma (4 units). Vasoactive therapy was initiated with a norepinephrine infusion at 0.09 ug/kg/min. The patient displayed signs of disseminated intravascular coagulation (DIC) and the routine clotting tests revealed a severe coagulation disorder (Table I). The coagulopathy was immediately treated using fibrinogen 2 g IV and recombinant activated factor VII (rFVIIa) 90 ug/kg IV,3 resulting in a prompt reversal of the life-��������������������������������������������� threatening bleeding. The ����������������������� mother and the newborn recovered promptly and completely without residual cardiac or neurological deficits. This case illustrates the importance of the early diagnosis of AFE followed by an aggressive treatment plan in order to achieve a good clinical outcome for both the mother and the child������������ . The availability of testing such as thromboelastometry and thrombelastography as an adjunct to conventional coagulation studies in the operating room may have facilitated the more precise treatment of the coagulation disorder (with hemostatic therapeutic agents) during post-partum hemorrhage. The use of rFVIIa remains controversial. However, current treatment guidelines for AFE suggest that rFVIIa only be used for hemostatic therapy in cases of ongoing obstetrical bleeding. Some experts have recommended that rFVIIa only be used after massive coagulation factors replacement has proven to be insufficient. 4 A large retrospective series presented by the Australian and New Zealand Haemostasis Registry suggested that earlier administration of rFVIIa might improve uterine preservation rates in this population. Despite an increased risk of thromboembolism in these patients, the complication has rarely been reported following rFVIIa use. This ���������������������������������������������� is a unique report involving the successful treatment of a parturient with a suspected AFE and life-threatening coagulopathy by early use of fibrinogen followed by rFVIIa. This approach may obviate the need for other more invasive and more costly therapeutic interventions. Both the parturient and the neonate survived without any residual morbidity. Table I.—Temporal changes in the patient’s perioperative laboratory test results. Hemoglobin (g/dL) Hematocrit (%) Platelets (n) Prothrombotin activity ratio (%) INR (%) Activated partial thromboplastin time (sec) Fibrinogen (mg/dL) 1238 Baseline Before procoagulation therapy After procoagulation therapy At 12 hours At 36 hours 11.6 35.0 359,000 1.1 1.0 1.0 469 04.5 13.1 247,000 4.00 3.84 Not coagulated Undetectable 08.7 25.5 138,000 0.98 1.00 1.61 101 07.8 23.9 114,000 0.98 1.00 1.13 140 08.4 24.7 133,000 Minerva Anestesiologica 1.01 0.96 168 November 2016 LETTERS TO THE EDITOR Lucia AURINI 1*, Maria Pia RAINALDI 2, Paul F. WHITE 3, Battista BORGHI 4 1Unit of Anesthesia, Department of Medical and Surgical Sciences, Sant’Orsola Hospital, University of Bologna, Bologna, Italy; 2Department of Maternal and Child Health, Unit of Anesthesia, Sant’Orsola Hospital, Bologna, Italy; 3Department of Anesthesiology at Cedars Sinai Medical Center, Los Angeles, CA, USA; White Mountain Institute, Tallgrass, The Sea Ranch, CA, USA; 4Department of Biomedical and Neuromotor Sciences, Unit of Anesthesia, Rizzoli Orthopedic Institute, University of Bologna, Bologna, Italy *Corresponding author: Lucia ������������������������������������� Aurini, Unit ����������������������� of Anesthesia, Department of Medical and Surgical Sciences, ����������������� University of Bologna, Sant’Orsola Hospital, via Massarenti 9, 40138 Bologna, Italy. E-mail: [email protected] References 1.Gist RS, Stafford IP, Leibowitz AB, Beilin Y. Amniotic fluid embolism. Anesth Analg 2009;108:1599-602. Vol. 82 - No. 11 2.Rainaldi MP, Tazzari PL, Scagliarini G, Borghi B, Conte R. Blood salvage during caesarean section. Br J Anaesth 1998;80:195-8. 3.Abdul-Kadir R, McLintock C, Ducloy AS, El-Refaey H, England A, Federici AB, et al. Evaluation and management of postpartum hemorrhage: consensus from an international expert panel. Transfusion 2014;54:1756-68. 4.Leighton BL, Wall MH, Lockhart EM, Phillips LE, Zatta AJ. Use of recombinant factor VIIa in patients with amniotic fluid embolism: a systematic review of case reports. Anesthesiology. 2011 115:1201-8. Authors’ contribution.—Lucia Aurini wrote the main part of the text (with the assistance of Dr. PF White); Maria Pia Rainaldi treated the patient, collected the data and revised the manuscript; Battista Borghi assisted with the revision of the manuscript Conflicts of interest.—The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Article first published online: July 22, 2016. - Manuscript accepted: July 6, 2016. - Manuscript revised: June 21, 2016. Manuscript received: May 3, 2016. (Cite this article as: Aurini L, Rainaldi MP, White PF, Borghi B. Successful treatment of life-threatening hemorrhaging due to amniotic fluid embolism. Minerva Anestesiol 2016;82:1238-9) Minerva Anestesiologica 1239 T O P 5 0 M I N E RVA A N E S T E S I O L O G I C A R E V I E W E R S Top 50 reviewers April 2016-September 2016 1240 Surname Name Savoia Bertini Agrò Ezzeldin Camporesi Biasucci Faraoni El Tahan Brazzi Aceto Benes Cattano Alston Brogly Ting Abad Gurumeta Caruselli Kinoshita Gottumukkala Montini Gómez-Ríos Vaida Deflandre Arnal Donadello Onutu Cavallone Riem Pabelick Donatelli Kayhan Borghi Turnbull De Blasi Tritapepe Gentili Max Dalfino Kim Bruder De Pascale Clendenen Lautrette Kakodkar Eden Beilin Sbaraglia Cinnella Lirk Guarracino Gennaro Pietro Eugenio Felice Ibrahim Saleh Enrico Mario Daniele Guerino David Mohamed Mohamed Paola Jan Davide Theodore Nicolas Chien-Kun Alfredo Marco Hiroyuki Vijaya Narasimha Raju Luca Manuel Ángel Sonia Eric P. Jean-Michel Katia Adela Hilda Laura Francesca Nicole Christina Maria Francesco Zeynep Battista David Roberto Alberto Luigi Andrea Martin Lidia Tae W. Nicolas Gennaro Steven R Alexandre Prashant Shivaji Arieh Yaakov Fabio Gilda Philipp Fabio MINERVA ANESTESIOLOGICA Number of revisions 10 9 9 9 8 8 8 7 7 7 7 7 6 6 6 6 6 5 5 5 5 5 5 5 5 4 4 4 4 4 4 4 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 November 2016