oulu 2015 d 1321 university of oulu p.o. box 8000 fi-90014...

102
UNIVERSITATIS OULUENSIS MEDICA ACTA D D 1321 ACTA Eila Sonkajärvi OULU 2015 D 1321 Eila Sonkajärvi THE BRAIN'S ELECTRICAL ACTIVITY IN DEEP ANAESTHESIA WITH SPECIAL REFERENCE TO EEG BURST-SUPPRESSION UNIVERSITY OF OULU GRADUATE SCHOOL; UNIVERSITY OF OULU, FACULTY OF MEDICINE; MEDICAL RESEARCH CENTER OULU; OULU UNIVERSITY HOSPITAL

Upload: others

Post on 11-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

UNIVERSITY OF OULU P .O. Box 8000 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

Professor Esa Hohtola

University Lecturer Santeri Palviainen

Postdoctoral research fellow Sanna Taskila

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Director Sinikka Eskelinen

Professor Jari Juga

University Lecturer Anu Soikkeli

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-0971-5 (Paperback)ISBN 978-952-62-0972-2 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1321

ACTA

Eila Sonkajärvi

OULU 2015

D 1321

Eila Sonkajärvi

THE BRAIN'S ELECTRICAL ACTIVITY IN DEEP ANAESTHESIAWITH SPECIAL REFERENCE TO EEGBURST-SUPPRESSION

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE;MEDICAL RESEARCH CENTER OULU;OULU UNIVERSITY HOSPITAL

Page 2: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2
Page 3: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

A C T A U N I V E R S I T A T I S O U L U E N S I SD M e d i c a 1 3 2 1

EILA SONKAJÄRVI

THE BRAIN'S ELECTRICAL ACTIVITY IN DEEP ANAESTHESIAWith special reference to EEG burst-suppression

Academic Dissertation to be presented with the assent ofthe Doctora l Train ing Committee of Health andBiosciences of the University of Oulu for public defence inAuditorium 1 of Oulu University Hospital,on 13 November 2015, at 12 noon

UNIVERSITY OF OULU, OULU 2015

Page 4: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

Copyright © 2015Acta Univ. Oul. D 1321, 2015

Supervised byProfessor Seppo AlahuhtaDocent Ville Jäntti

Reviewed byDocent Kaisa HartikainenProfessor Juhani V Partanen

ISBN 978-952-62-0971-5 (Paperback)ISBN 978-952-62-0972-2 (PDF)

ISSN 0355-3221 (Printed)ISSN 1796-2234 (Online)

Cover DesignRaimo Ahonen

JUVENES PRINTTAMPERE 2015

OpponentProfessor Leena Lindgren

Page 5: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

Sonkajärvi, Eila, The brain's electrical activity in deep anaesthesia. With specialreference to EEG burst-suppressionUniversity of Oulu Graduate School; University of Oulu, Faculty of Medicine; MedicalResearch Center Oulu; Oulu University HospitalActa Univ. Oul. D 1321, 2015University of Oulu, P.O. Box 8000, FI-90014 University of Oulu, Finland

Abstract

Several anaesthetics are able to induce a burst-suppression (B-S) pattern in theelectroencephalogram (EEG) during deep levels of anaesthesia. A burst-suppression patternconsists of alternating high amplitude bursts and periods of suppressed background activity. Allmonitors measuring the adequacy of anaesthesia recognize the EEG B-S as one criterion. A betterunderstanding of EEG burst-suppression is important in understanding the mechanisms ofanaesthesia. The aim of the study was to acquire a more comprehensive understanding of thefunction of neural pathways during deep anaesthesia.

The thesis is comprised of four prospective clinical studies with EEG recordings from 64patients, and of one experimental study of a porcine model of epilepsy with EEG registrationstogether with BOLD fMRI during isoflurane anaesthesia (II). In study I, somatosensory corticalevoked responses to median nerve stimulation were studied under sevoflurane anaesthesia at EEGB-S levels. In study III, The EEGs of three Parkinson`s patients were observed to describe thecharacteristics of B-S during propofol anaesthesia using scalp electrodes and depth electrodes inthe subthalamic nucleus. In study IV, EEG topography was observed in 20 healthy children underanaesthesia mask induction with sevoflurane. Twenty male patients were randomized to eithercontrolled hyperventilation or spontaneous breathing groups for anaesthesia mask induction withsevoflurane in study V. EEG alterations in relation to haemodynamic responses were examined instudies IV and V.

Somatosensory information reached the cortex even during deep anaesthesia at EEG burst-suppression level. Further processing of these impulses in the cortex was suppressed. The EEGslow wave oscillations were synchronous over the entire cerebral cortex, while spindles and sharpwaves were produced by the sensorimotor cortex. The development of focal epileptic activitycould be detected as a BOLD signal increase, which preceded the EEG spike activity. Theepileptogenic property of sevoflurane used at high concentrations especially duringhyperventilation but also during spontaneous breathing together with heart rate increase, wasconfirmed in healthy children and male. Spike- and polyspike waveforms concentrated in amultifocal manner frontocentrally.

Keywords: anaesthesia, BOLD, burst suppression, electroencephalography, epilepsy,epileptogenic, functional magnetic resonance imaging, hyperventilation, propofol,sevoflurane, sleep spindle, somatosensory evoked potentials

Page 6: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2
Page 7: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

Sonkajärvi, Eila, Aivojen sähköinen aktiivisuus syvässä anestesiassa. Erityisestiaivosähkökäyrän purskevaimentuman aikanaOulun yliopiston tutkijakoulu; Oulun yliopisto, Lääketieteellinen tiedekunta; Medical ResearchCenter Oulu; Oulun yliopistollinen sairaalaActa Univ. Oul. D 1321, 2015Oulun yliopisto, PL 8000, 90014 Oulun yliopisto

Tiivistelmä

Useat anestesia-aineet pystyvät aiheuttamaan aivosähkökäyrän (EEG) purskevaimentumansyvän anestesian aikana. Purskevaimentuma koostuu EEG:n suuriamplitudisten purskeiden sekävaimentuneen taustatoiminnan vaihtelusta. Kaikkien anestesian syvyyttä mittaavien valvontalait-teiden toiminta perustuu osaltaan EEG:n purskevaimentuman tunnistamiseen. Tämän ilmiönparempi tunteminen on tärkeää anestesiamekanismien ymmärtämiseksi. Tutkimuksen päämäärä-nä oli saada kattavampi käsitys hermoratojen toiminnasta syvässä anestesiassa.

Väitöskirjatyö koostuu neljästä prospektiivisesta yhteensä 64 potilaan EEG-rekisteröinnitsisältävästä tutkimuksesta sekä yhdestä kokeellisen epilepsiatutkimuksen koe-eläintyöstä, jossaporsailla käytettiin isofluraanianestesiassa sekä EEG-rekisteröintejä sekä magneettikuvantamis-ta (fMRI) samanaikaisesti (II). Ensimmäisessä osatyössä tutkittiin keskihermon stimulaationaiheuttamia somatosensorisia herätepotentiaaleja aivokuorella EEG:n purskevaimentumatasollasevofluraanianestesian aikana. Kolmannessa osatyössä selvitettiin propofolianestesian aiheutta-maa EEG:n purskevaimentumaa kolmelta Parkinsonin tautia sairastavalta potilaalta käyttäensekä pintaelektrodien että subtalamisen aivotumakkeen syväelektrodien rekisteröintejä. Neljän-nessä osatyössä tutkittiin EEG:n topografiaa 20:llä terveeellä lapsella indusoimalla anestesiasevofluraanilla. Kaksikymmentä miespotilasta nukutettiin sevofluraanilla ja heidät satunnaistet-tiin joko kontrolloidun hyperventilaation tai spontaanin hengityksen ryhmiin osatyössä V. EEG-muutoksia sekä niiden yhteyttä verenkiertovasteisiin selviteltiin molemmissa osatöissä IV ja V.

Omasta kehosta tuleviin tuntoärsykkeisiin liittyvä somatosensorinen informaatio saavuttiaivokuoren myös syvässä EEG:n purskevaimentumatasoisessa anestesiassa. Impulssien jatkokä-sittely aivokuorella oli kuitenkin estynyt. EEG:n hidasaaltotoiminta oli synkronista koko aivo-kuoren alueella, sen sijaan unisukkulat ja terävät aallot paikantuivat sensorimotoriselle aivokuo-relle. Paikallisen epileptisen toiminnan kehittyminen oli mahdollista havaita jo ennen piikikkäi-den EEG:n aaltomuotojen ilmaantumista edeltävänä BOLD-ilmiöön liittyvänä aivoverenkierronlisääntymisenä. Sevofluraanin epileptogeenisyys varmistui erityisesti hyperventilaation, muttamyös spontaanin hengityksen yhteydessä ja näihin liittyi sykkeen nousu sekä terveillä lapsillaettä miehillä. Piikkejä ja monipiikkejä käsittävien aaltomuotojen keskittymistä esiintyi otsaloh-kon keskialueilla.

Asiasanat: aivosähkökäyrä, anestesia, BOLD, epilepsia, epileptogeeninen,hyperventilaatio, magneettikuvaus, propofoli, purskevaimentuma, sevofluraani,somatosensoriset herätepotentiaalit, unisukkula

Page 8: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2
Page 9: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

7

Acknowledgements

The research for this thesis was carried out during the years 1996-2006 at the

Department of Anaesthesiology, in the Oulu University Hospital, in close

collaboration with the Department of Clinical Neurophysiology, Oulu University

Hospital. One of the studies was carried out at the Department of Diagnostic

Radiology, Oulu University Hospital.

I owe my deepest gratitude to my supervisors Professor Seppo Alahuhta,

M.D, Ph.D., and Docent Ville Jäntti, M.D, Ph.D. It has been a sincere delight and

privilege to have fruitful discussions and to share the moments of discovery

within their guidance. They have introduced me to the world of critical scientific

thinking. Seppo, you have given me the best possible support and encouridgement

in carrying out this project and you have patiently pushed me forward. Ville, you

have been the primus motor of this study and teached me the alphabet of clinical

neurophysiology, I am also grateful to the possibility you offered me to stay a few

months at the Department of Clinical Neurophysiology.

Docent Kaisa Hartikainen, M.D, Ph.D. and Professor Juhani Partanen, M.D,

Ph.D, who kindly agreed to be the official reviewers of this work, deserve my

sincere gratitude. Their valuable comments and constructive criticism have

improved the final outcome of this thesis.

I express my special thanks and respect to my co-author Minna Silfverhuth

nee Mäkiranta, Ph.D., for her encouraging example and scientific attitude. I also

want to thank all my other co-authors, especially Kalervo Suominen, Ph.Lic., Pasi

Puumala, M.D., Seppo Rytky, M.D., Docent Anne Vakkuri, M.D., Ph.D., Nita

Hakalax, specialised nurse in clinical neurophysiology, and Elina Karvonen,

M.D., for their professional assistance and contribution. The help by Professor

Heikki Löppönen, M.D, Ph.D, in making the fourth study in the

otorhinolaryngology unit possible is also acknowledged. I also want to owe my

warmest thanks to Raija Remes, specialised nurse in clinical neurophysiology, for

all her help and collaboration over this researh, it has meant a lot to me.

I am grateful to Professor Arvi Yli-Hankala, M.D., Ph.D., for his interest and

positive attitude for my project. He proposed the fourth study and also gave

valuable comments during the early stage of the writing process of the thesis.

I wish to thank Docent Kai Kiviluoma, M.D., Ph.D, and Docent Timo

Salomäki, M.D., Ph.D., for their valuable roles in the follow-up group.

My special thanks go to Pasi Ohtonen, for his excellent contribution on

biostatistics.

Page 10: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

8

I am sincerely grateful to Michael Spalding, M.D., Ph.D., for his efficient and

thorough language revision.

I am greatly indebted to Professor Esa Heikkinen, M.D., Ph.D., the pioneer in

deep-brain stimulation treatment at the Department of Neurosurgery, for his

positive attitude and help with the study of Parkinson`s patients.

I express my sincere thanks to Docent Päivi Laurila, M.D, Ph.D., Head of the

Department of Anaesthesiology, and Professor Tero Ala-Kokko, M.D., Ph.D.,

head of the Division of the Intensive Care medicine, for their positive and helpful

attitude towards my scientific work.

I owe my deep appreciation of all help given by the skillful anaesthesia staff

participating in the care of the study patients, especially by Arto Seljänperä,

specialised nurse in anaesthesiology.

I want to thank all my collegues for friendly and stimulating working

athmophere. I wish to thank Salli Pätsi, M.D., for her kind practical help with the

child patients of the study, and Vesa Pakanen, M.D., for his supportive way of

listening and sharing the problems in everyday patient care. I owe my special

thanks to Pirjo Ranta, M.D., Ph.D, and Risto Ahola, M.D., for their help with all

arrangements related to the dissertation day, planning with them has been fun.

Merja Vakkala, M.D., Ph.D., Hanna Rautiainen, M.D., and Tanja Forchini, M.D.,

are adknowledghed for their support and flexibility in organizing the time-table of

my clinical and scientific working periods.

I owe my warmest thanks to all my dear friends, who have given me plenty of

positive energy. I especially want to thank my `soul sister` Riitta Vesikukka for

her great source of woman power and sense of humour during all those moments

we have spent together.

Finally, I want to thank my dear mother Elvi for her lifelong support and

never failing love. I also want to thank my brother Risto for unlimited practical

help during these years. I am grateful to my family, Timo and our belowed

daughter Sara, for all the joy and happiness you have brought to my life.

This work was supported by EVO grants from Oulu University Hospital,

Pharmacy grants of Oulu University Hospital, and Graduate School of Finnish

Academy, all of which are gratefully acknowledged.

Oulu October 2015 Eila Sonkajärvi

Page 11: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

9

Abbreviations

AEP Auditory evoked potentials

AVDO2 Arterio-venous oxygen content difference

BIS Bispectral index

BOLD Blood oxygen level-dependent

bpm Beats per minute

B-S Burst-suppression

CBF Cerebral blood flow

CBFV Cerebral blood flow velocity

CH Controlled hyperventilation

CMR Cerebral metabolic rate

CNS Central nervous system

CO2 Carbon dioxide

CV Controlled ventilation

D Delta, < 4 Hz activity in EEG

DC Direct current

DS Delta slow, < 2 Hz activity in EEG

ECG Electrocardiogram

ECoG Electrocorticogram

EEG Electroencephalogram

EMG Electromyogram

EP Evoked potential

EPSP Excitatory postsynaptic potential

ET End-tidal

ETCO2 End-tidal carbon dioxide

ETN2O End-tidal nitrous oxide

ETsevo End-tidal sevoflurane concentration

FFT Fast Fourier transformation

FiO2 Fraction of inspired oxygen

fMRI Functional magnetic resonance imaging

GABA Gamma-amino butyric acid

GM Grand mal, epileptic seizure with tonic-clonic convulsions

Hz Hertz

HR Heart rate

IPSP Inhibitory postsynaptic potential

kPa kiloPascal

Page 12: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

10

MAC Minimal alveolar concentration

MAP Mean arterial pressure

MRI Magnetic resonance imaging

MV Minute ventilation

N20 Negative 20 ms wave in somatosensory evoked potentials

N2O Nitrous oxide

NMDA N-methyl-D-aspartic acid

NREM Non-REM or slow wave sleep

PaCO2 Arterial carbon dioxide concentration

PED Periodic epileptiform discharges

PS Polyspikes

PSP Postsynaptic potential

RCT Randomized controlled trial

RE Reticular nucleus of thalamus

REM Rapid eye movement sleep

S Suppression

SSEP Somatosensory evoked potentials

SB Spontaneous breathing

SAP Systolic arterial pressure

SSP Suppression with spikes

TR Thalamic reticular

Page 13: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

11

List of original publications

This thesis is based on the following publications, which are referred throughout

the text by their Roman numerals:

I Jäntti V, Sonkajärvi E, Mustola S, Rytky S, Kiiski P, Suominen K (1998) Single- sweep cortical somatosensory evoked potentials: N20 and evoked bursts in sevoflurane anaesthesia. Electroencephalogr clin Neurophysiol 108: 320–324.

II Mäkiranta M, Ruohonen J, Suominen K, Niinimäki J, Sonkajärvi E, Kiviniemi V, Seppänen T, Alahuhta S, Jäntti V, Tervonen O (2005) BOLD signal change precedes interictal spike activity in EEG – a dynamical model of experimental penicillin induced focal epilepsy in deep anesthesia. Neuroimage 27: 715–724.

III Sonkajärvi E, Puumala P, Erola T, Baer GA, Karvonen E, Suominen K, Jäntti V (2008) Burst-suppression during propofol anaesthesia recorded from scalp and subthalamic electrodes: report of three cases. Acta Anaesthesiol Scand 52: 274–279.

IV Sonkajärvi E, Alahuhta S, Suominen K, Hakalax N, Vakkuri A, Löppönen H, Ohtonen P, Jäntti V (2009) Topographic electroencephalogram in children during mask induction of anaesthesia with sevoflurane. Acta Anaesthesiol Scand 53; 77–84.

V Sonkajärvi E, Alahuhta S, Suominen K, Rytky S, Kumpulainen T, Ohtonen P, Karvonen E, Jäntti V. Epileptiform EEG activity induced by rapid sevoflurane anaesthesia induction. Manuscript.

Page 14: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

12

Page 15: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

13

Contents

Abstract

Tiivistelmä

Acknowledgements 7 Abbreviations 9 List of original publications 11 Contents 13 1 Introduction 15 2 Review of the literature 19

2.1 General anaesthesia ................................................................................. 19 2.1.1 Mechanisms of anaesthesia .......................................................... 19

2.2 EEG ......................................................................................................... 21 2.2.1 Physiological basis of EEG .......................................................... 21 2.2.2 Registering EEG ........................................................................... 22 2.2.3 Epileptiform EEG discharges and signs of epilepsy ..................... 23 2.2.4 Burst-suppression ......................................................................... 25 2.2.5 Arousal in EEG ............................................................................ 31 2.2.6 Somatosensory evoked potentials (SSEP) .................................... 32 2.2.7 EEG based monitoring ................................................................. 33

2.3 Anaesthetics and the CNS ....................................................................... 34 2.3.1 Sevoflurane ................................................................................... 34 2.3.2 Isoflurane ...................................................................................... 45 2.3.3 Propofol ........................................................................................ 45 2.3.4 Nitrous oxide ................................................................................ 47 2.3.5 Summary of the neurophysiological effects of anaesthetics ......... 48

2.4 EEG during physiological sleep and general anaesthesia ....................... 50 2.5 Epileptiform EEG, anaesthesia and sleep ............................................... 53

3 Aims of the study 55 4 Subjects and Methods 57

4.1 Subjects ................................................................................................... 57 4.2 Variables and measurements ................................................................... 58 4.3 Anaesthesia ............................................................................................. 58 4.4 EEG Recordings and analyses ................................................................ 60 4.5 Stimulation .............................................................................................. 61 4.6 Statistical analysis ................................................................................... 61

5 Results 63

Page 16: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

14

5.1 SSEP in sevoflurane anaesthesia (I) ........................................................ 63 5.2 Epileptiform EEG pattern in sevoflurane anaesthesia (IV, V) ................. 64 5.3 Focal seizure, fMRI and EEG (II) ........................................................... 68 5.4 Depth electrode recordings in propofol anaesthesia (III) ........................ 69

6 Discussion 71 6.1 Methodological aspects ........................................................................... 71 6.2 SSEP in sevoflurane anaesthesia (I) ........................................................ 71 6.3 Epileptiform EEG in sevoflurane anaesthesia (IV, V) ............................. 73 6.4 Focal seizure, fMRI and EEG (II) ........................................................... 76 6.5 Depth electrode recordings in propofol anaesthesia (III) ........................ 77

7 Conclusions 79 References 81 Original publications 97

Page 17: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

15

1 Introduction

General anaesthesia can be viewed as pharmacological actions administered to

prevent the adverse effects of surgical trauma and to create optimal conditions for

surgery. These pharmacological interventions include unconsciousness,

anxiolysis, amnesia, analgesia, autonomic stability and immobility (Kissin 1993).

Several studies suggest that different components of general anaesthesia result

from the actions of a single drug at separate molecular and anatomical sites in the

central nervous system (CNS), and it is now widely accepted that anaesthetic

agents produce immobility by acting primarily on the spinal cord (Rampil & King

1996), and amnesia and hypnosis by acting on neurons in the brain. Since the

introduction of inhaled anaesthetics in the 1840’s, anaesthetic practice has

changed significantly. Present-day anaesthetists employ a wide variety of drugs,

i.e. inhaled and intravenous anaesthetics, analgesics, muscle relaxants and

sedatives, to produce general anaesthesia (Rudolph & Antkowiak 2004,

Campagna et al. 2003). Consciousness is a complex entity comprising two major components: the

content of consciousness and the level of consciousness. The former means the

subject`s awareness of environment and of self and the latter wakefulness i.e. the

level of consciousness (Laureys 2005). Altered stages of consciousness are

produced via several different mechanisms such as physiological sleep, general

anaesthesia, intoxications, metabolic disorders, diffuse ischaemic brain damage

and epileptic seizures (Fig. 1). The electroencephalogram (EEG) pattern of burst-

suppression (B-S) may be observed in all of these states, but during sleep only the

discontinuous pattern of newborn babies resembles the B-S pattern (Jäntti 2012).

The safest way to produce unconsciousness is via the mechanisms of

physiological sleep. These include both sleep prompting mechanisms and arousal

mechanisms. This is obviously the reason why, with most anaesthetics, the EEG

closely resembles the patterns seen in physiological sleep. The most obvious

change is an increase in slow activity. This is an essentially local oscillation of the

cerebral cortex but in the healthy brain it is synchronized throughout the entire

cerebral cortex. During deep anaesthesia this is abruptly changed into a B-S

pattern which resembles the trace alternant, discontinuous pattern of neonatal

quiet sleep (Sainio 2006). In neonates, the postictal B-S pattern may sometimes

gradually transform into this physiological pattern after a focal seizure. Heart rate is mainly controlled by the autonomous nervous system. The

sympathetic system enhances, while the parasympathetic system inhibits the

Page 18: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

16

cardiac pacemaker sinoatrial node. These pathways are under the control of the

higher brain regions, such as the hypothalamic, thalamic, and cortical brain

regions. EEG burst-suppression during some periods of anaesthesia, especially during

induction, is not an uncommon phenomenon. Pulse variation and moderate

alterations in systemic circulation, as well as blood flow in the circulation of the

cerebral cortex are related to B-S. All monitors which measure the adequacy of

anaesthesia recognize B-S as one criterion. At present, B-S is accepted as a

definition for deep anaesthesia. A better understanding of B-S is important in

understanding the mechanisms of anaesthesia, as well as when treating epileptic

patients in intensive care units. This study was carried out to acquire a more comprehensive understanding of

the function of sensory and neural pathways during deep anaesthesia. The effects

of sevoflurane and propofol on somatosensory evoked potentials (SSEP) and EEG

during deep anaesthesia in adults and children were studied. Most previous

studies have reported four channel EEG registrations, while in the present study

an extensive 32 channel array, according to the full 10–20 system, including

orbitofrontal and ear electrodes, and depth electrode were used. This enabled

more detailed analyses of the EEG. The relationship between EEG and the blood-

oxygen-level-dependent (BOLD) signal during isoflurane burst-suppression

anaesthesia was evaluated using an experimental model of epilepsy.

Page 19: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

17

Fig. 1. An oversimplified illustration of the two major components of consciousness:

the level of consciousness (i.e. wakefulness or arousal) and the content of

consciousness (i.e. awareness or experience). In normal physiological states level

and content are positively correlated (with the exception of dream activity during

REM-sleep). Patients in pathological (e.g. intoxications, brain damage) or

pharmacological coma (that is, general anaesthesia) are unconscious and they cannot

be awakened. Dissociated states of consciousness (i.e. patients being seemingly

awake but lacking any behavioural evidence of voluntary or willed behaviour), such as

the vegetative state or much more transient equivalents such as absence and

complex partial seizures and sleepwalking, offer a unique opportunity to study the

neural correlates of awareness. (Modified from Laureys S. (2005) The neural correlate

of (un)awareness: lessons from vegetative state. TRENDS in Cognitive Sciences 9:

556-9.)

Sleepwalking Complex partial and

absence seizures Vegetative state

Cont

ent o

f con

scio

usne

ss(a

war

enes

s)

Level of consciousness (wakefulness)

General anaesthesia

Coma

REM sleep

ConsciousWakefulness

Drowsiness

Light sleep

Deep sleep

Page 20: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

18

Page 21: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

19

2 Review of the literature

2.1 General anaesthesia

Since the introduction of surgical anaesthesia one hundred and seventy years ago

the definition of anaesthesia has evolved and is now defined as unconsciousness

(hypnotic effect) and blockade of somatic motor response to noxious stimulation

(analgesic effect). It is difficult to separate the anaesthetic actions taking part in

these two subcomponents. Very few anaesthetics act exclusively on either of these

components, as they are closely related to each other. Unconsciousness prevents

the (conscious) perception of pain, and nociception may serve as an arousal

stimulus and change the level of sedation and hypnosis. The type of interaction

(synergism, antagonism, summation) may vary from that of other components.

(Kissin 1993).

2.1.1 Mechanisms of anaesthesia

In past decades, research on anaesthetic mechanisms has mainly focused on ion

channels located in the membranes of nerve cells. The identification of specific

binding sites for volatile anaesthetics on certain proteins was a dramatic departure

from the classic view that all general anaesthetics act non-specifically. Gamma-

aminobutyric acid type A (GABAA) receptors are involved in the actions of many

general anaesthetics. GABAA receptors are found throughout the central nervous

system, both at synapses and extrasynaptically, and they are thought to be one of

the main sites for the actions of anaesthetic agents. Anaesthetic agents which have

an effect on GABAA receptors include volatile anaesthetics and most intravenous

anaesthetics, e.g. sodium thiopental, propofol and etomidate (MacIver et al. 1988,

Krasowski et al. 1998). These drugs can produce three kinds of effects on

GABAA receptors: potentiation, i.e. markedly increase the current elicited by low

concentrations of GABA (Wakamori et al. 1991), direct gating, i.e. activate

channels in the absence of GABA (Wittmer et al. 1996), and inhibition, i.e.

prevent current flow through channels (Hall et al. 1994). Another important

molecular target for anaesthetic agents is the NMDA-type (N-methyl-D-aspartic

acid) receptor, a major postsynaptic, ionotropic receptor for the excitatory

neurotransmitter glutamate. These receptors are also found both presynaptically

and extrasynaptically. NMDA receptors are sensitive to ketamine (Zeilhofer et al.

Page 22: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

20

1992), nitrous oxide and xenon (Yamakura & Harris 2000), as well as to other

volatile anaesthetics to some extent - particularly with regards to their analgesic

effects (Aronstam et al. 1994). NMDA (glutamate) and GABA receptors together

constitute 90% of neurones in the mammalian brain. Potassium channels,

especially two-pore-domain potassium channels (2PK channels), have been

shown to be activated by volatile and gaseous anaesthetics, but not by clinically

relevant concentrations of intravenous anaesthetics. The anaesthesic activation of

2PK channels generally results in an inhibition of neuronal activity and a

reduction in excitatory currents (Patel et al. 1999). These channels are also found

presynaptically with either inhibitory or excitatory effects. There is growing

evidence that other receptors are also modulated by anaesthetics. These include

glycine receptors, where volatile anaesthetics and propofol increase the affinity of

the receptor for glycine, and neuronal nicotinic receptors, where channels are

inhibited by volatile anaesthetics. In addition, noradrenergic neurones and certain

Na+ channels modulate anaesthetic actions. The primary target for

dexmedetomidine is the alpha-2 receptors on neurons in the locus coeruleus

(Mizobe et al. 1996).

The fundamental mechanism which produces general anaesthesia is not fully

understood. Synaptic function is considered to be the major target of anaesthetic

drug action. It is widely considered that the deactivation of the thalamic nuclei

occurs during anaesthetic drug-induced unconsciousness. Current theories suggest

that disruption in information flow and integration within the corticothalamic and

corticocortical networks accounts for the mechanism of general anaesthesia. The

anaesthetic agents hyperpolarize neurons (create a more negative resting

membrane potential) by enhancing inhibitory or decreasing excitatory

transmission and alter neuronal activity depending on the site of action. Despite

different mechanisms and sites of action, the outcome produced by anaesthetic

agents is the same: i.e. unconsciousness. (Franks 2008, Alkire et al. 2008)

In the future, mathematical computational models advanced into more

sophisticated systems and replicating EEG patterns will enable the accurate

examination of anaesthesia mechanisms by offering directed simulations of

specific aspects of cortical function of neural responses to pharmacological

manipulation. A basic mean field method in which the mean activity of a small

region of the cortex is modelled, has already been used to study the mechanisms

of inhalation anaesthetics. The neural activity over roughly the extent of a cortical

macrocolumn of approximately one hundred inhibitory and excitatory neurons is

averaged in this model to produce a mean field description in space, while no

Page 23: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

21

averaging with respect to time is necessary (Liley & Bojak 2005, Wilson et al. 2006, Sceniak 2006). In order to better understand the neurophysiological

mechanisms behind the different alterations in consciousness induced by

numerous anaesthesia agents, further studies linking methods of cognitive

neuroscience such as EEG, magnetoencephalography (MEG), isotope methods

such as positron emission tomography (PET) and functional magnetic resonance

imaging are necessary (Jäntti 2005).

2.2 EEG

2.2.1 Physiological basis of EEG

The macroscopic electrical potentials recorded at the scalp are a summation of the

microscopic potentials generated by individual cellular elements. These cells

include neurons, glial cells, and muscle cells in the scalp (hence the artefacts that

can be generated from scalp electromyography EMG). The EEG is mainly

generated from the superficial layer of pyramidal neurons by changes in

postsynaptic potentials (PSP) in the dendrites oriented perpendicular to the

cortical surface (Young 2003). The PSP occurs when the presynaptic neurons

release neurotransmitters which alter the permeability of the ion channels in cell

membranes of the postsynaptic neurons, while the transmembrane ionic

concentrations change inducing an alteration in the transmembrane voltage

(Rampil 1998). The neuronal contribution to the EEG comes as a summation of

excitatory postsynaptic potentials (EPSPs) and inhibitory postsynaptic potentials

(IPSPs), not from action potentials (Young 2003).

Anaesthesia as well as other mechanisms such as reduced vigilance, both in

normal sleep and in pathological states which depress consciousness, are

associated with increased cortical synchrony and high-voltage waves of low

frequency. Higher cortical function, such as cognitive activity, is usually

associated with desynchronization and low-voltage waves of high frequency, as

more neurons act independently in the creation of conscious human behaviour.

Synchrony is also reduced by arousal during anaesthesia or normal sleep (Young

2003, Binnie & Prior 1994). Desynchronization occurs with the activation of

ascending cholinergic projections of the basal forebrain and brainstem nuclei of

raphe (an important supplier of serotonin) and locus coeruleus (the main supplier

of noradrenalin). Each of the states of awareness, awake, slow sleep, and deep

Page 24: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

22

sleep, has its characteristic rhythms of EEG oscillation. The rhythmicity is a result

of interactions between the thalamus and the cortex (John & Prichep 2005).

Most EEG signal activity manifests at below 30 Hz, at alpha, beta, theta and

delta frequencies. Alpha activity with a frequency of 8–13 Hz, mainly around

10 Hz, and an amplitude from 20 to 50 µV is distributed maximally in the

occipital regions bilaterally, while during the drowsy state this shifts anteriorly.

Alpha activity is blocked at eye opening when it becomes beta frequency (Berger

1929). By the age of three years, the alpha rhythm frequency is in the 8–9 Hz

range.

Beta rhythm is characteristic of an awake and alert person with a rapid

frequency of usually 13–30 Hz and an amplitude < 20 µV in most of adults. Beta

activity may exist during rapid eye movement (REM) and during light non-REM

(NREM) sleep (Steriade 2006), and at the beginning of anaesthesia.

Benzodiazepines are a group of sedatives known to potentiate beta activity.

Theta activity is rather slow in frequency, being between 4 and 7 Hz, with an

amplitude of 50–100 µV. This rhythm is observed in light sleep and may represent

the inhibitory action of GABAergic interneurons affecting the cortico-thalamic

network. It may be associated with limbic activity (John & Prichep 2005).

Delta rhythm has a slow frequency between 0.5 and 3–4 Hz, and a large

amplitude of 100–200 µV. This rhythm is observed in deep sleep, anaesthesia and

coma. It is generated in the cortical pyramidal cells with cortico-thalamic

dissociation in sensory input (Steriade 2006).

Gamma rhythm consists of rapid oscillations above 30 Hz. Reflecting the

interconnection of cortico-cortical and cortico-thalamo-cortical networks, this

rhythm may be involved in the processes of perception (John & Prichep 2005).

The EEG may contain special stereotypic waveforms, such as spikes or sharp

waves used in the diagnosis of epilepsy. They are sharply featured excursions in

the EEG and created by massive, but usually transient synchrony (Rampil 1998).

2.2.2 Registering EEG

The signal registered in an EEG is generated by the cerebral cortex. When a large

area of the cortex is active synchronously, it creates electrical currents which flow

through the skull to the scalp, then continue under the scalp to return to the inside

of the brain and close the current loop under the cortex (Fig. 2). Therefore the

EEG can be recorded both above the cortex as well as under the cortex. An EEG

is analyzed by measuring the frequency and amplitude of the electrical activity

Page 25: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

23

and by recognition of the wave patterns. The analysis of the change over time in

the original raw EEG voltages change is called time domain analysis, where

amplitude is presented along the y-axis and time horizontally along the x-axis.

The most commonly used method to register EEG activity is the international

10–20 system based on meridians crossing the scalp based on key landmarks with

additional lines drawn over the mid-frontal and mid-parietal lobes (Rampil 1998).

The amplitude of the EEG signals is 10 to 100 µV, which is 100 times weaker

than that of the electrocardiogram. The basic types of EEG activity are continuous

and rhythmic activity, transients, background activity and asymmetries between

cerebral hemispheres (Rampil 1998).

Fig. 2. The EEG is produced by electrical currents in a volume conductor, also

intracerebrally. The voltmeter (V) of the EEG recording machine does not measure

potential directly; it measures the current in the measuring circuit. This current always

depends on the location of both electrodes. (Nunez PL & Srinivasan R (2006) Electrical

fields of the brain. The neurophysics of EEG. Reprinted with the permission of the

copyright holder).

2.2.3 Epileptiform EEG discharges and signs of epilepsy

Epileptiform EEG discharges consist of spikes, spike- and slow wave complexes

and sharp waves (Mervaala 2006). Single spikes are generally negative transients

Page 26: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

24

clearly distinguished from the background EEG activity with variable amplitude

and duration, usually within a range of 20 to 70 ms (Chang et al. 2011). Sharp

waves have a longer duration of 70 to 200 ms with a prolonged descending phase

while the rising phase resembles that of single spikes (Chang et al. 2011). Sharp

waves are neurophysiologically closely related to spikes and both of these

paroxysmal discharges may take place during an interictal epilepsy period,

although they may also occur in persons without a history of seizure disorder

(Chang et al. 2011).

Rhythmic polyspikes consist of multiple spike complexes and occur more or

less rhythmically in bursts of variable duration. These paroxysmal waveforms

comprise more than two negative and positive deflections appearing at regular

intervals and are associated with a slow wave or mixed frequency EEG activity

between spike complexes. Polyspikes are usually bilateral. (Chang et al. 2011,

Nordli et al. 2011).

Periodic lateralized or focal epileptiform discharges (PLEDs) as described by

Chatrian et al. (1964), consist of simple and sharp waves or complex discharges

with mixed spiky and slower elements with amplitudes around 100 to 300 µV. It

is possible that PLED activity, however, is generated independently bilaterally

and synchronously over both hemispheres and the term generalized periodic

epileptiform discharges is often used to describe this activity (Chang et al. 2011).

PLEDs can be seen in the EEG in a variety of acute neurologic conditions such as

acute cerebral infarction, neoplastic or inflammatory central nervous diseases,

herpes simplex or infectious mononucleosis encephalitis (Mervaala 2006, Chang

et al. 2011).

Epileptogenesis refers to hypersynchrony involving either excitatory or

recurrent inhibitory neuronal networks (Binnie & Stefan 1999). Epileptogenesis is

a process which causes the brain to develop and extend the brain tissue capable to

generate spontaneous seizures resulting in the development of an epileptic

condition and, later, possible progression of established epilepsy (Pitkänen &

Engel 2014). Epileptogenic mechanismas are likely to be specific for many

different causes of epilepsy including genetic influences, malformations of

cortical development, autoimmune disorders, and cell loss and synaptic plasticity

e.g. hippocampal atrophy and sclerosis, the major cause of mesial temporal lobe

epilepsy (Pitkänen & Engel 2014). In epileptic current spontaneous seizures there

are paroxysmal hypersynchronous transient electrical discharges resulting from

too much activation or too little inhibition in the area of abnormal discharges

(Treiman 2001).

Page 27: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

25

Goddard (Goddard 1967, Goddard et al. 1969) used repeated electrical

stimulations of low intensity to the brain limbic structures when studying learning

mechanisms and noticed that some laboratory animals developed epileptic

seizures. This model - called kindling - is one example of secondary

epileptogenesis which has been extensively studied in the laboratory over the last

50 years; nevertheless its relevance to human epilepsy remains controversial

(Cibula & Gilmore 1997).

Temporal lobe epilepsy and hippocampal sclerosis have been found to

develop after prolonged febrile seizures, especially after febrile status epilepticus

during early childhood (Scott et al. 2003). In the FEBSTAT (Consequences of

Prolonged Febrile Seizures) study, focal slowing or attenuation 72 h after

convulsion were seen maximally over the temporal areas in a substantial

proportion of children and these alterations were highly associated with acute

hippocampal injury illustrated with MRI (Nordli et al. 2012). Febrile seizures are

associated with a febrile illness without CNS infection, electrolyte imbalance or

prior afebrile seizures in children older than one month (Anonymous 1993). Some

investigators believe that genetic factors or malformations of cortical brain

development such as brain dysplasias may predispose to febrile status epilepticus

(Patteson et al. 2014).

The EEG pattern of the classical absence seizure (petit mal) of genetic origin

most characteristically consists of a generalized slow 3 Hz spike-wave complex, a

dicrotic negative spike followed by a negative slow wave (Mervaala 2006, Nordli

et al. 2011). Activity with a large amplitude and maximum activity on the

superior frontal area is symmetrical and synchronous in both hemispheres and

commences abruptly with the onset of the attack and ceases at its end (Nordli et al. 2011). In the primarily generalized spike-wave seizure, burst firing of cortical

neurons occurs during the spike while the slow wave is accompanied by

hyperpolarization due to recurrent inhibition (Binnie & Stefan 1999).

2.2.4 Burst-suppression

Burst-suppression (B-S) is an EEG pattern usually consisting of bilateral

intermittent quasiperiodical sequences of high-voltage slow and sharp waves, i.e.

bursts, which mainly have a less than 15 Hz frequency (Kroeger & Amzica 2007),

alternating with periods of depressed background activity with an amplitude

usually < less than 5–10 µV (Kuroiwa & Celesia 1980, Quasha et al. 1981)

Page 28: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

26

lasting from a few seconds to minutes (suppression). B-S indicates a nonspecific

reduction in cerebral metabolic activity.

Derbyshire et al. (1936) initially reported on the B-S pattern and

demonstrated that it may appear under different anaesthetics and is reactive to

sensory stimulation. Because a large cortical response to mechanical nerve

stimulation was obtained even after the cortex had become completely quiescent

under pentobarbital or avertin anaesthesia, but under deep ether anaesthesia with

spontaneous waves still visible no response to stimulation appeared, they

concluded that the former two anaesthetics suppressed the cortex activity without

blocking the sensory paths, while ether blocked the sensory pathways before

cortical activity was wholly suspended. The name “burst suppression” was

introduced by Swank & Watson (1949) in deeply narcotized animals. They used

the term “suppression-burst”. Their discovery was that, during barbiturate

narcosis, spontaneous electrical activity periodically became decreased which was

then followed by bursts. According to their report, the phenomenon was found in

the frontal lobe cortex after isolation from the thalamus. Henry & Scoville (1952)

detected a B-S pattern by using subdural electrodes to register an EEG in the

surgically isolated cortex.

B-S is a phenomenon which can be induced by several inhalation anaesthetics

such as enflurane, isoflurane, desflurane and sevoflurane usually at concentrations

of 1.5–2 MAC. It is also induced with intravenous anaesthetics such as propofol,

thiopental and etomidate (Eger et al. 1971, Clark & Rosner 1973, Newberg et al. 1983, Jäntti & Yli-Hankala 1990, Yli-Hankala & Jäntti 1990, Rampil et al. 1991,

Hartikainen et al. 1995, Hartikainen et al. 1996, Arkawi et al. 1996). The B-S

pattern characteristic of deep anaesthesia varies with the anaesthetic used. During

enflurane anaesthesia, the spikes in the bursts are very sharp and during the

suppressions the EEG pattern is nearly isoelectric (Clark & Rosner 1973, Yli-

Hankala et al. 1990a). Hypocarbia lengthens the suppression time and decreases

the duration of bursts, but the amplitude and main frequency of the bursts increase

to resemble epileptoid spikes. Isoflurane bursts may have sharp waves but are not

as spiky as enflurane bursts and suppressions often have a low amplitude activity

of up to 10 µV (Clark & Rosner 1973). Hartikainen et al. (1996) used

somatosensory stimuli during isoflurane B-S anaesthesia. A local anaesthetic

nerve conduction block induced in a single finger in a double-blinded manner

caused a disappearance of the evoked EEG burst responses indicating a peripheral

conduction block in a somatosensory pathway. This test provided an example of

monitoring the integrity of the neural pathway during deep anaesthesia. Lipping et

Page 29: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

27

al. (1995) compared isoflurane and enflurane B-S patterns and found that bursts

and suppressions were longer during isoflurane anaesthesia and that the B-S ratio

decreased as the anaesthesia deepened, while deepening enflurane anaesthesia did

not change the B-S ratio. Enflurane bursts were also spikier (Lipping et al. 1995).

Propofol bursts are slow negative waves with a smooth onset and a superimposed

alpha frequency of 12–13 Hz spindle waves which begin and end smoothly (Jäntti

& Yli-Hankala 1993, Huotari et al. 2004). In some cases, minor stimuli may

trigger bursts after prolonged suppression during isoflurane anaesthesia (Yli-

Hankala et al. 1993b), and auditory, photic, and somatosensory stimuli all readily

produce these (Derbyshire et al. 1936, Eger et al. 1971, Porkkala et al. 1994

Hartikainen et al. 1995, Hartikainen & Rorarius 1999). Yli-Hankala et al. (1993b)

used vibration stimuli with a duration of 3 seconds applied to the hand of patients

under isoflurane anaesthesia. The EEG bursts had a latency of approximately

0.5 seconds and differed from spontaneously evoked bursts. They were also

associated with increased heart rate, which may indicate an activation of the

autonomic nervous system brought on by the stimulus (Yli-Hankala et al. 1993b)

Heart rate and cerebral blood flow velocity (CBFV) changes are associated

with B-S anaesthesia. During enflurane anaesthesia at B-S level in the EEG, heart

rate increased at burst onset and decreased at suppression onset. Researchers have

theorized that both the B-S cortical activity and heart rate fluctuations may

possibly be controlled by the same subcortical mechanism (Yli-Hankala et al. 1990a, Jäntti & Yli-Hankala 1990, Yli-Hankala et al. 1993c). The onset of EEG

suppression was associated with an abrupt decrease of CBFV in normoventilated

patients at a physiological steady-state under anaesthesia with a high dose of

inhalants (isoflurane and desflurane) at 1.5–2 MAC (Lam et al. 1995). The

change in EEG always preceded a change in flow velocity by 5–7 seconds. The

parallel alterations in cerebral blood flow and electrical activity of the brain

suggested that the flow-metabolism coupling mechanism is at least partially

preserved during high-dose inhalant anaesthesia. The changes in CBF induced by

inhalation anaesthetics may be mediated by a dual mechanism representing a

balance between two opposing mechanisms, the direct cerebral vasodilative

action and the indirect flow-metabolism mediated cerebral vasoconstriction (Lam

et al. 1995).

B-S is also associated with coma (Young 2000, Kaplan & Bauer 2011),

hypoxia (Pressler et al. 2001, Thordstein et al. 2004), cardiac arrest (Zaret 1985,

Berkhoff et al. 2000, Thömke et al. 2002, Rundgren et al. 2006), drug-related

intoxications, different childhood encephalopathies (Lenard et al. 1976, Ohtahara

Page 30: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

28

& Yamatogi 2003) and hypothermia (Quasha et al. 1981). Hofmeijer et al. (2014)

detected that identical bursts during B-S, with burst shapes highly similar and

bilaterally synchronous, were associated with a poor outcome in comatose

patients after cardiac arrest. Inter-burst intervals were variable in duration and

invariably flat. Widespread injury to the ascending neuronal systems normally

producing arousal and desynchronization of the EEG are observed in alpha coma

(Berkhoff et al. 2000). Postictal attenuation following epileptic seizure is in many

ways a phenomenon of the same type as B-S.

B-S usually occurs symmetrically, but in brain injury it may also appear

asynchronously or focally in brain hemispheres. In the Aicardi syndrome (Aicardi

et al. 1965), as well as in corpus callosum damage, asynchronic B-S is observed

with reference to B-S synchronization in hemispheres across this structure of the

brain. Lazar et al. (1999) demonstrated an interhemispheric asynchrony of

pentobarbital-induced B-S associated with a haemorrhage in the corpus callosum

in a child with status epilepticus.

The B-S level in the EEG can also be used as guided therapy level for epileptic

patients during status epilepticus treated with anaesthetics such as propofol or

barbiturates (Shorvon & Ferlisi 2012). Beydoun et al. (1991) used propofol for

treating status epilepticus patients by deepening anaesthesia until the suppression

time was approximately five seconds.

The neurophysiological mechanisms responsible for generating B-S patterns

are not fully understood. Burst may be developed in a small cortical area

synchronizing additional cortex or it may be generated via brainstem arousal

mechanisms. Focal or asymmetric bursts may represent the former description

while normal bursts during anaesthesia are examples of the latter. Using

intracellular recordings and extracellular calcium recordings in laboratory

animals, Kroeger & Amzica (2007) found that the brain becomes reactive to

subliminal stimuli (auditory, visual and somatosensory) during deep anaesthesia

at a B-S level in the EEG. The same stimuli did not provoke any overt responses

during varied depths of anaesthesia. They theorized that the increase in baseline

extracellular Ca2+ might constitute the main factor triggering cortical

hyperexcitability during B-S anaesthesia.

Figure 3 exemplifies bursts at a B-S anaesthesia level during

monoanaesthesia using the inhaled anaesthetic sevoflurane (Fig. 3) and the

intravenous anaesthetic propofol (Fig. 4).

Page 31: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

29

Fig. 3. EEG during B-S in sevoflurane anaesthesia. Sevoflurane burst registered with

scalp- and deep brain electrodes of a patient with Parkinson`s disease. Surface

electrodes referred to depth electrode. The four upper traces are the EEG recorded

between frontal midline (Fz), left parietal (P3), neck (C7), chin and one contact in the

depth electrode in the subthalamic nucleus. Depth 1 – Depth 4 is recorded between

two contacts of the subthalamic electrode. Note that it records the cortically

generated activity of bursts.

Page 32: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

30

Fig. 4. EEG during B-S in propofol anaesthesia. Propofol burst registered with scalp-

and deep brain electrodes of a patient with Parkinson`s disease (from study III). The

uppermost trace is the EEG from two scalp electrodes, the right parietal P4 and the

right frontal F4. The second trace is the EEG recorded between two contacts in the

depth electrode in the subthalamic nucleus. The lower traces are the EEG recorded

between scalp electrodes P4 and F4, and the electrode contact deepest in the

subthalamic nucleus (Jäntti et al. 2008. Reprinted with the permission of the copyright

holder).

Page 33: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

31

2.2.5 Arousal in EEG

The concept of brainstem arousal systems was introduced by Moruzzi et al. in the

1940`s (Moruzzi & Magoun 1949). By stimulating the reticular formation of the

brain stem of anaesthetized animals they demonstrated a cessation of

synchronized discharge in the EEG and its replacement with low voltage fast

activity, while the intensity of the alteration varied with the degree of background

synchrony present. Actually, arousal was first demonstrated by Hans Berger in

1929, when he observed the alpha rhythm blockade and shift to beta dominance

during eye-opening in a subject under EEG monitoring.

Several nuclei located in the pons, midbrain, hypothalamus, and basal

forebrain regulate normal sleep-wake cycles. These wake-ON/sleep-OFF and

sleep-ON/wake-OFF nuclei are thought to inhibit each another reciprocally. Some

arousal centers are active primarily during wakefulness, with cholinergic nuclei

also active during rapid eye movement sleep. Other centers, such as the

GABAergic ventrolateral preoptic nucleus of the thalamus, are active during sleep

(Mashour et al. 2011).

The noradrenergic locus coeruleus in the pons and the histaminergic

tuberomamillary nucleus in the posterior hypothalamus are active during waking,

whereas the ventrolateral preoptic nucleus is inhibited. As the homeostatic

pressure for sleep builds, the ventrolateral preoptic nucleus becomes active in

association with sleep and then inhibits the activity of the arousal-promoting

locus coeruleus and tuberomamillary nuclei (Mashour et al. 2011).

While the EEG pattern during deepening anaesthesia resembles the patterns

of deepening sleep, the patterns induced by arousals also resemble the EEG

patterns of arousal during sleep. These arousal patterns are usually rapid

transitions to fast low activities (beta arousal), but occasionally they are high

amplitude rhythmic delta patterns, which resemble the arousal patterns

occasionally seen in children during awakening, delta arousal (Aho et al. 2011).

Both the burst-suppression and the delta arousal patterns therefore seem to reflect

a return to neonatal EEG mechanisms. Delta arousal, “paradoxical arousal”, is a

condition presenting with a marked decrease of EEG frequency and a decrease in

bispectral index (BIS) induced by a noxious stimulus. It is observed during

anaesthesia induced with volatile anaesthetics in combination with nitrous oxide.

In a topographical EEG study, Kochs et al. (1994) demonstrated that surgical

stimulus resulted in an increase in delta activity during 0.6 MAC isoflurane

anaesthesia with nitrous oxide while delta shift was attenuated by using larger

Page 34: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

32

1.2 MAC concentrations of isoflurane. Pre-treatment with fentanyl suppressed the

paradoxical response of a decrease in BIS to intra-abdominal irrigation during

anaesthesia maintenance with sevoflurane and 50% N2O (Morimoto et al. 2005).

The study by Oda et al. (2006) confirmed these findings. BIS first increased after

intubation during isoflurane or sevoflurane anaesthesia. When 66% nitrous oxide

was added for intubation, BIS significantly decreased but pain medication with

fentanyl completely abolished the decrease in BIS.

Interestingly, a pattern similar to delta arousal has also been described in

deepening anaesthesia with sevoflurane and has been coined slow delta (Yli-

Hankala et al. 1999). The normal and pathological physiology of sleep and

arousal systems are essential keys for understanding unconsciousness during

anaesthesia, as is the level of EEG burst-suppression.

2.2.6 Somatosensory evoked potentials (SSEP)

Evoked potentials (EPs) are generated in the brain as electrophysiological

responses of the synchronized activation of the nervous system to external

sensory or motor stimulation. They are derived from the EEG in response to

auditory, somatosensory, nociceptive and visual stimuli. The evoked responses

reflect the functional integrity of specific peripheral and central nervous system

(CNS) regions (Thornton & Sharpe 1998). EPs are recorded at the scalp and

consist of precisely timed sequences of waves. The single cortical sensory evoked

response has a very low amplitude of 1–2 µV, compared with the much larger

EEG waves of 50–100 µV amplitude. Therefore, the EP wave has to be extracted

from concurrent spontaneous EEG activity and averaged by repetitive stimulation

and computer-signal techniques (Banoub & Tetzlaff 2003).

Intraoperative EP changes may result from surgical injury or ischaemia of the

specific neural pathway, or they may be due to nonspecific physiological or

pharmacological influences. Physiological factors influencing EPs include

temperature (Porkkala et al. 1997b), blood pressure, hematocrit, acid-base

balance, and oxygen and carbon dioxide tensions. Anaesthetic drugs and sedatives

are the most common pharmacologic causes of nonspecific EP changes (Porkkala

et al. 1997a). In addition to somatosensory EP, visual, auditory (brainstem) and

motor evoked potentials are also used to evaluate the integrity of the sensory

pathway. Sensory evoked potentials are classified as short-latency (< 30 ms),

intermediate-latency (30–75 ms), or long-latency (> 75 ms) waves (Banoub &

Tetzleff 2003).

Page 35: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

33

Halogenated inhalation anaesthetics have been demonstrated to produce dose-

dependent attenuation on short-latency SEP amplitude as well as latency delay

especially in cortical components (Peterson et al. 1986, Samra et al. 1987, Sebel

et al. 1986, Sebel et al. 1987, Sloan 1998). Although nitrous oxide stimulates the

EEG, it also causes an amplitude reduction in the somatosensory median nerve

precentral negative cortical component (N20) without an effect on latency (Sebel

et al. 1984, Sloan & Koht 1985, Thornton et al. 1992). Vandesteene et al. (1993)

demonstrated via topographic brain mapping that nitrous oxide did not change the

distribution of the precentral positive cortical component (P22) of median nerve

SEP despite its clear amplitude increase. The generator of the cortical component

was the same as that recorded in a person awake.

2.2.7 EEG based monitoring

The EEG provides a good indicator as to whether the brain is active or inactive as

well as affording a window to the level of brain activity, such as the depth of

hypnosis during general anaesthesia (Crosby & Culley 2012). Various algorithms

for EEG signal processing have been developed in commercially available depth-

of-anaesthesia devices, such as the bispectral index (BISR, Aspect Medical

Systems) (Rampil 1998, Sigl & Chamoun 1994), Entropy (M-EntropyTM module,

GE Healthcare) (Viertiö-Oja et al. 2004), Narcotrend, and Cerebral State IndexTM

(CSITM, Danmeter, Odense, Denmark). BIS® is an index derived empirically from

the EEG to estimate the effect of anaesthetics and unconsciousness. It reflects the

spectral properties of the signal as well as the burst-suppression pattern

(Drummond 2000, Sebel et al. 1997, Bruhn et al. 2000). To date, spectral entropy

is the only method being applied in commercial use from the entropy measures.

Approximate entropy responds better, however, to the visual impression of the

signal regularity. It is more adept at taking into account the patterns and

waveforms, the phase information in the signal, and is consequently better at

describing the underlying physiology, for example, epileptic spikes (Anier et al. 2012).

Of the two parameters generated by entropy, State entropy (SE) is calculated

over the 0.8–32 Hz frequency range including the EEG-dominant part of the

spectrum. It therefore primarily reflects the cortical state and is a reflection of the

patient`s level of hypnosis. Response entropy (RE) is calculated over a frequency

rate from 0.8 Hz to 47 Hz including both the EEG-dominant and the EMG-

dominant part of the spectrum (Viertiö-Oja et al. 2004). RE at least partly reflects

Page 36: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

34

the activation level of the upper facial EMG representing arousal and indirectly

provides a measurement of the adequacy of analgesia.

EEG-based depth-of-anaesthesia monitors reliably describe the hypnotic

effects of anaesthetics with a GABAergic (GABAA) mechanism of action such as

isoflurane, sevoflurane, desflurane, thiopental and propofol which cause a gradual

slowing of the EEG until burst suppression (Sebel et al. 1997, Ellerkmann et al. 2004, Vakkuri et al. 2004) whereas a correlation to discriminate unconsciousness

is poorer with anaesthetics acting mainly via NMDA receptors such as ketamine

(Hans et al. 2005, Maksimov et al. 2006) or nitrous oxide (Soto et al. 2006,

Ozcan et al. 2010). A correlation was found, however, with xenon anaesthesia

(Laitio et al. 2008, Fahlenkamp et al. 2010).

Using a BIS-monitor, Myles et al. (2004) reported a remarkable reduction in

risk of awareness during anaesthesia in high-risk patients. Depth-of-anaesthesia

monitoring also allows reduced anaesthetic consumption and faster emergence

(Vakkuri et al. 2005, Aime et al. 2006), postoperative nausea and vomiting

(Nelskylä et al. 2001) and cardiovascular stability during anaesthesia in elderly

patients (Riad et al. 2007).

Many authorities have highlighted the importance of interpreting the raw

EEG biosignal on the monitor while using a processed depth-of-anaesthesia

monitor to avoid under- or overdosing of anaesthetics (Jäntti 2005, Bennett et al. 2009, Mashour et al. 2011, Jäntti 2013).

2.3 Anaesthetics and the CNS

2.3.1 Sevoflurane

Sevoflurane (fluoromethyl 1, 1, 1, 3, 3, 3-heksafluoro-2-propyl-ether) is a methyl

isopropyl ether inhalational anaesthetic with a trifluorinated methyl group on the

alpha carbon atom. As a result of its chemical structure, sevoflurane is susceptible

to degradation by the hepatic microsomal enzymes P450 (specifically isoform

IIE1) with the release of an inorganic fluoride into the circulation. Sevoflurane

produces major potentiation in the GABAA and glycine receptors and major

inhibition in the NMDA and serotonin receptors.

Nonpungency and a rapid increase in alveolar anaesthetic concentration

makes sevoflurane the most popular agent for inhalation induction in paediatric

(Lerman et al. 1994, Goa et al. 1999) and adult patients (Yasuda et al. 1991, Eger

Page 37: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

35

1994). The rapid increase in the inspiratory concentration of sevoflurane,

however, has been demonstrated to be associated with transient hyperdynamic

reactions in adult patients during controlled mild hypocapneic hyperventilation

(Vakkuri et al. 1999). The same research group later observed that the

hyperdynamic circulatory response was associated with epileptiform EEG in

adults (Yli-Hankala et al. 1999), as well as paediatric patients without

hyperventilation (Vakkuri et al. 2001). Hyperventilation per se at a high

sevoflurane concentration produces an epileptiform EEG regardless of its timing

and hypocapnia seems to have an additive effect on the incidence of epileptiform

EEG during sevoflurane inhaled anaesthetic induction (Vakkuri et al. 2000).

Based on these results, the speed of induction of anaesthesia, high expired

concentration of sevoflurane, and hyperventilation have been proposed as risk

factors for the occurrence of sevoflurane-induced epileptiform EEG

abnormalities. Female gender has been shown to predispose to electrical seizure

during sevoflurane induction (Julliac et al. 2007).

The occurrence of sevoflurane-elicited epileptiform abnormalities seems to

be concentration dependent with a probable threshold around 3.2% in adults

(Jääskeläinen et al. 2003) and 4.3% in children (Gibert et al. 2012) expressed as

the expired fraction of sevoflurane. The risk of epileptiform EEG discharges can

be reduced, but not totally prevented, during the induction of anaesthesia with low

target concentrations of sevoflurane in normoventilated adult female patients

(Julliac et al. 2013). A non-randomized study found that epileptiform EEG

patterns were observed in 20% of normoventilated paediatric patients even though

the administration time of 8% sevoflurane was shortened by reducing the inspired

sevoflurane concentration to 4% immediately after loss of consciousness (Schultz

et al. 2012). The propensity for sevoflurane to produce epileptiform EEG activity

is greater in patients with epilepsy compared to normal controls (Iijima et al. 2000). The areas activated by sevoflurane in patients with epilepsy might not be

confined to the epileptogenic zones of spontaneous seizures (Hisada et al. 2001).

The combination of nitrous oxide with sevoflurane has been shown to decrease

the number of spikes in epileptic patients although it did not affect the extent of

areas with spikes (Kurita et al. 2005). Kaisti et al. (2002) detected in their PET

study that, at a surgical anaesthesia level of 1 MAC sevoflurane and an effective

concentration (EC50) of propofol, both anaesthetics caused a marked global

reduction of regional cerebral blood flow (rCBF), but propofol more than

sevoflurane. This effect was maintained with propofol even under deeper

anaesthesia, while at 2 MAC sevoflurane caused a noticeable redistribution.

Page 38: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

36

The relationships of sevoflurane anaesthesia and epileptiform episodes have

been widely debated by anaesthesiologists, as sevoflurane is the most commonly

used inhalation anaesthetic. Several reports of seizure-like phenomena in patients

under sevoflurane anaesthesia are available, but only a few of these observations

have accompanying EEG recordings. Many of these documents are clinical notes

including one to some patients interpreted as epileptic phenomena because of

seizure-like movements during either the induction or maintenance of anaesthesia

with sevoflurane without EEG monitoring (Table 1). Alternatively, there are also

prospective studies with EEG registration (Table 2). Without EEG registration, it

is very difficult to determine whether seizure-like muscle movements are due to

epileptiform activity or to non-epileptic myoclonia (Modica et al. 1990a).

Page 39: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

37

Ta

ble

1. S

ev

ofl

ura

ne

an

d e

pile

pti

form

EE

G,

ca

se

re

po

rts

(m

od

ifie

d f

rom

Co

ns

tan

t et

al.

200

5).

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Ada

chi e

t al.

1991

Cas

e re

port

(lette

r)

one

girl

9 ye

ars

Sev

o6 Fi1 4

% in

duct

ion

No

To

nic-

clon

ic m

ovem

ents

with

resp

alk

alos

is H

R, B

P o

k

Kom

atsu

et a

l.

1994

Cas

e re

port

two

epile

ptic

boy

s

aged

3 a

nd 8

yea

rs

Sev

o in

duct

ion

Fi 1

% to

7%

by in

crea

sing

0.5

-1.0

% d

urin

g

ever

y 1–

2 m

in

14-c

hann

el E

EG

(acc

ordi

ng to

10–

20)

Firs

t PS

2 , th

en R

PS

3 , B

-S5

with

incr

easi

ng s

evo

from

2 to

7%

in c

entra

l par

ieta

l,

occi

pita

l and

fron

tal a

reas

No

card

iova

sc s

ide-

effe

cts

No

mov

emen

t abn

orm

aliti

es

Tera

sako

& Is

hii

1996

Cas

e re

port

one

man

N

o E

EG

Sei

zure

-like

mov

emen

ts d

urin

g

emer

genc

y (la

sted

40

s)

Woo

dfor

th e

t al

1997

Cas

e re

port

one

girl

11 y

ears

Iv-in

duct

ion

(thio

pent

one)

mai

nten

ance

with

Fi s

evo

ad

7% in

O2/

air

No

prem

edic

atio

n

16-c

hann

el E

EG

(acc

ordi

ng to

10–

20)

PS

, RP

S, t

o lo

ng B

-S a

t

sevo

Fi 6

–7%

No

seiz

ure-

like

mov

emen

ts

Bös

enbe

rg e

t al.

1997

Cas

e re

port

(lette

r)

one

boy

12 y

ears

Sev

o in

duct

ion

(con

cent

ratio

n

not m

entio

ned)

No

S

eizu

re-li

ke m

ovem

ents

from

2 m

in o

f sta

rting

indu

ctio

n

Zach

aria

s et

al.

1997

Cas

e re

port

(lette

r)

two

child

ren

2 an

d

4 y

Sev

o Fi

7–8

% in

duct

ion

in

O2/a

ir

No

S

eizu

re-li

ke m

ovem

ents

Bai

nes

et a

l.

1998

Cas

e re

port

(lette

r)

child

ren

(num

ber n

ot

repo

rted)

Sev

o in

duct

ion

(% n

ot to

ld)

No

C

onvu

lsiv

e m

ovem

ents

inde

pend

ently

of w

heat

her o

r

not s

tepw

ise

incr

ease

of s

evo

Sch

ulz

& S

chul

z

1998

Cas

e re

port

(lette

r)

child

ren

and

adul

ts

(num

ber n

ot

repo

rted)

Sev

o in

duct

ion

Yes

E

pile

ptifo

rm c

hang

es w

ith

Fi s

evo

> 5%

(whe

n de

lta-

activ

ity c

hang

ed to

BS

)

Page 40: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

38

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Kai

sti e

t al.

1999

Cas

e re

port

of tw

o

youn

g ad

ults

Sev

o m

aint

enan

ce w

ith s

low

ly

incr

easi

ng to

2 M

AC

in O

2/air

No

prem

edic

atio

n

Four

EE

G m

onta

ges

in

both

sid

es (a

ccor

ding

to

10–2

0)

BS

, asy

mm

etric

al P

ED

s4

in b

oth

at 2

MA

C

One

had

asy

mm

etric

clo

nic

mov

emen

ts (p

artia

l mot

or

seiz

ure)

PE

T sh

owed

dec

reas

e to

40–

80%

of r

CB

F8 in

2 M

AC

sev

o

Hilt

y &

Dru

mm

ond

2000

Cas

e re

port

one

mal

e pa

tient

Indu

ctio

n w

ith s

evo

Fi 8

% in

N2O

/O2 5

0% ->

mai

nten

ance

sevo

2%

, SB

Mid

azol

am p

rem

idec

atio

n

No

D

urin

g re

cove

ry c

linic

al e

pile

ptic

seiz

ure

(cer

ebra

l im

agin

g

show

ed b

rain

tum

or)

Sch

ultz

2000

Cas

e re

port

two

child

ren

Indu

ctio

n w

ith s

evo

Fi 7

–8%

Y

es

Epi

lept

iform

EE

G-

chan

ges

Sch

ultz

2001

Cas

e st

udy

one

adul

t

Y

es

Spi

kes

unde

r sev

o 5–

6%

Koy

ama

et a

l.

2002

Cas

e re

port

an e

pile

ptic

man

36 y

ears

Mai

nten

ance

sev

o E

T7 1.5

2.5%

in O

2/air

Yes

M

ore

spik

es w

ith E

t 2.5

%

sevo

than

1.5

%.

Fent

anyl

dec

reas

ed s

pike

frequ

ency

und

er E

T 1.

5%

sevo

.

Åke

son

&

Did

rikss

en

2004

Cas

e re

port

two

child

ren

N

o

Sei

zure

-like

mov

emen

ts d

urin

g

indu

ctio

n w

ith s

evo

ET

3%

Chi

nzei

2004

Cas

e re

port

one

epile

ptic

adu

lt

E

EG

, BIS

E

pile

ptifo

rm E

EG

with

mai

nten

ance

of s

evo

1%

asso

ciat

ed w

ith

fluct

uatio

ns o

f BIS

Hsi

eh

2004

Cas

e re

port

one

neon

ate

N

o

Sei

zure

-like

mov

emen

ts in

all

extre

miti

es d

urin

g em

erge

nce

Page 41: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

39

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Lecl

erc

2006

Cas

e re

port

one

child

2 y

ears

Ana

esth

esia

indu

ctio

n w

ith

8% s

evo

Mid

azol

am p

rem

edic

atio

n

No

S

eizu

re-li

ke m

yocl

onic

mov

emen

ts

Yan

o

2008

Cas

e re

port

two

child

ren

with

hist

ory

of fe

brile

conv

ulsi

ons

Sev

o m

aint

enan

ce E

T 2–

3%

in 7

0% N

2O/O

2

No

D

urin

g m

aint

enan

ce o

ne h

ad

toni

c-cl

onic

sei

zure

and

the

othe

r one

dur

ing

reco

very

1 in

spira

tory

frac

tion,

2 po

lysp

ikes

and

–w

aves

, 3 rh

ythm

ic p

olys

pike

s, 4

perio

dic

epile

ptifo

rm d

isch

arge

s, 5

burs

t sup

pres

sion

, 6 se

voflu

rane

, 7 en

d-tid

al,

8 reg

iona

l cer

ebra

l blo

od fl

ow.

Page 42: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

40

Ta

ble

2. S

ev

ofl

ura

ne

an

d e

pile

pti

form

EE

G,

pro

sp

ec

tiv

e s

tud

ies

(m

od

ifie

d f

rom

Co

ns

tan

t et

al.

200

5).

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Hag

a et

al.

1992

Pro

spec

tive

stud

y

180

child

ren

1–6

y

Sev

o9 Fi1 4

–6%

indu

ctio

n N

o

Sei

zure

-like

mov

emen

ts

in 6

% o

f chi

ldre

n

Artr

u et

al.

1997

Pro

spec

tive

rand

omiz

ed s

tudy

,

four

teen

adu

lts

Iv-in

duct

ion

(thio

pent

one

/

etom

idat

e) 0

.5–1

.0–1

.5 M

AC

mai

nten

ance

with

sev

o (n

= 8

)

or is

o10 (n

= 6

) in

O2/a

ir

EE

G 1

6 or

5 e

lect

rode

mon

tage

s (a

ccor

ding

to

10–2

0)

No

inst

ance

s of

epi

lept

iform

activ

ity in

EE

G

ICP

12 d

id n

ot c

hang

e

Vm

ca d

ecre

ased

Wat

ts e

t al.

1999

Pro

spec

tive

stud

y

(mea

n 27

yea

rs o

ld)

elev

en e

pile

ptic

adul

ts

Mai

nten

ance

sev

o 1.

5 M

AC

in

O2/a

ir w

ith

1) n

orm

oven

tilat

ion

and

2) h

yper

vent

ilatio

n

For E

CoG

14 w

ith h

yper

-

vent

ilatio

n in

O2/a

ir

1) is

o 0.

3–1.

5 an

d

2) s

evo

1.5

MA

C

Eig

ht c

hann

el E

EG

band

pas

s 0.

5–30

Hz

8 ch

anne

l EC

oG

Epi

lept

iform

EE

G-c

hang

es

unde

r 1.5

MA

C s

evo

(incr

ease

of i

nter

icta

l spi

kes

in a

ll) >

1.5

MA

C is

o un

der

1.5

MA

C n

o m

ore

epile

ptifo

rm E

EG

in

hype

r/nor

mov

entil

atio

n

Con

stan

t et a

l.

1999

Pro

spec

tive

rand

omiz

ed s

tudy

four

ty-fi

ve c

hild

ren

of

2–12

yea

rs

Indu

ctio

n w

ith

1) s

evo

Fi 7

% in

100

% 0

2

2) s

evo

Fi 2

to 4

to 6

%

3) h

alo11

Fi 1

to 2

to 3

to 3

.5%

in N

2O/O

2 50%

4) la

ter o

n se

vo F

i 7%

in

N2O

/O2 5

0%

Pre

med

icat

ion

mid

azol

am

16 c

hann

els

(10–

20)

band

pas

s 0.

5–30

Hz

spec

tral a

naly

sis

No

epile

ptifo

rm E

EG

chan

ges,

sha

rp s

low

wav

es

with

sev

o

typi

cal h

alo

traci

ng s

imila

r to

barb

itura

te s

pind

les

(slo

w

wav

es s

uper

impo

sed

with

fast

rhyt

hms

alph

a an

d be

ta)

with

hal

o

Clin

ical

agi

tatio

n w

ith

sevo

in te

n ou

t of 1

1, in

seve

n ou

t of 1

3 w

ith h

alo

With

sev

o 10

0% O

2 mor

e

agita

tion

than

with

N2O

.

Page 43: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

41

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Yli-

Han

kala

et a

l.

1999

Pro

spec

tive,

rand

omiz

ed s

tudy

thirt

y he

alth

y w

omen

Indu

ctio

n w

ith s

evo

Fi 8

% in

N2O

/O2 5

0%

spon

tane

ous

vent

ilatio

n (S

V8 )

vs. c

ontro

lled

hype

rven

tilat

ion

(CH

V, E

TCO

213 2

6 m

mH

g)

Pre

med

icat

ion

diaz

epam

Four

bip

olar

ele

ctro

de

pairs

(10–

20)

(4 c

hann

els?

)

PS

2 13/

15 in

CH

V v

s 6/

15 in

SV

gro

up P

ED

4 in

13/1

5 C

HV

vs 1

/15

in S

V g

roup

B-S

6 with

spi

kes

3/15

in C

HV

grou

p

Ass

ocia

tion

betw

een

HR

rise

and

epile

ptifo

rm

EE

G (H

R ro

se >

30%

) in

CH

V p

atie

nts

Thre

e in

CH

V g

roup

had

jerk

ing

mov

emen

ts

Iijim

a et

al.

2000

Pro

spec

tive

cros

sove

r stu

dy s

evo

vs is

o (a

fter 3

mon

ths)

twel

ve e

pile

ptic

and

twel

ve n

on-e

pile

ptic

men

tally

hand

icap

ped

adul

ts

Indu

ctio

n se

vo/is

o 1.

0, 1

.5

and

2.0

MA

C

1) In

100

% O

2

norm

oven

tilat

ion

2) In

O2/

N2O

50%

norm

oven

tilat

ion

3) In

100

% O

2

hype

rven

tilat

ion

(HV

7 )

Pre

med

icat

ion

or a

nti-

conv

ulsa

nts

wer

e no

t giv

en

Hig

h ba

nd p

ass

120

Hz

In e

pile

ptic

pat

ient

s

epile

ptifo

rm E

EG

with

sev

o

1.5

MA

C >

iso

1.5

MA

C in

none

pile

ptic

pat

ient

s no

epile

ptifo

rm c

hang

es in

eith

er

grou

ps

Sup

plem

enta

tion

of N

2O o

r

HV

sup

pres

sed

occu

rren

ce

of s

pike

s

Vak

kuri

et a

l.

2000

Pro

spec

tive,

rand

omiz

ed s

tudy

thirt

y w

omen

Indu

ctio

n w

ith s

evo

Fi 8

% in

N2O

/O2 5

0%

1) im

med

iate

HV

ETC

O2

> 4%

2) d

elay

ed H

V (f

irst 2

min

SV

)

Four

bip

olar

ele

ctro

de

pairs

(acc

ordi

ng 1

0–20

)

band

pas

s 1–

50 H

z

sam

plin

g ra

te 1

28 H

z

Epi

lept

iform

EE

G in

13/

15

(gro

up 1

) vs

9/15

(gro

up 2

)

PE

D in

10/

15 in

gro

up 1

vs

5/15

in g

roup

2

Reg

ardl

ess

of ti

min

g, H

V

with

sev

o 8%

pro

duce

s

hype

rdyn

amic

circ

ulat

ory

resp

onse

Vak

kuri

et a

l.

2001

Pro

spec

tive,

rand

omiz

ed s

tudy

31 h

ealth

y ch

ildre

n of

1–12

yea

rs (m

ean

age

6 ye

ars)

Indu

ctio

n w

ith s

evo

Fi 8

% in

N2O

/O2 2

:1

1) c

ontro

lled

vent

ilatio

n

ETC

O2 4

.3–5

.3%

)

2) s

pont

aneo

us b

reat

hing

ETC

O2 a

d 6–

6.7%

Pre

med

icat

ion

mid

azol

am

Bip

olar

four

-cha

nnel

EE

G

Ban

d pa

ss 1

–50

Hz

Sam

plin

g ra

te 1

28 H

z

RP

S3 i

n 7/

16 in

CV

vs

3/15

in

SB

gro

up

PE

D in

7/1

6 in

CV

vs

none

in

SB

gro

up

SS

P5 i

n 4/

16 v

s no

ne in

SB

grou

p

No

mot

or m

anife

stat

ions

Epi

lept

iform

EE

G w

as

asso

ciat

ed w

ith

incr

ease

d H

R (m

ore

than

60%

from

bas

elin

e) a

nd

BP

Page 44: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

42

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Con

reux

et a

l.

2001

Pro

spec

tive

stud

y

Twen

ty c

hild

ren

of 1

8 ye

ars

and

AS

A I-

II

Indu

ctio

n w

ith s

evo

Fi 8

% in

O2 1

00%

No

prem

edic

atio

n

Eig

ht b

ipol

ar m

onta

ges

Sam

plin

g ra

te 1

28 H

z

2/20

chi

ldre

n ha

d S

,PS

4/20

had

B-S

with

out s

pike

s

14/2

0 ha

d sh

arp

mon

opha

sic

slow

del

ta d

urin

g

lary

ngos

copy

2/20

chi

ldre

n ha

d

myo

clon

ic s

eizu

re-li

ke

mov

emen

ts

Sch

ultz

et a

l.

2001

Pro

spec

tive

stud

y

seve

n ad

ults

Indu

ctio

n w

ith s

evo

Yes

6/

7 ha

d sp

ikes

in E

EG

with

sevo

Fi 5

–6%

His

ada

et a

l.

2001

Pro

spec

tive

cros

s-

over

stu

dy

six

epile

ptic

adu

lts

Pro

pofo

l ind

uctio

n ->

mai

nten

ance

with

sev

o

1.5

MA

C (s

tepw

ise

incr

ease

)

in O

2/ N

2O 3

3% ->

cha

nged

to

iso

1.5

MA

C

No

prem

edic

atio

n or

antie

pile

ptic

s gi

ven

EC

oG s

ubdu

ral g

rid

elec

trode

s

Epi

lept

iform

cha

nges

far

mor

e th

an w

ith is

o, n

ot

conf

ined

to z

one

of

spon

tane

ous

seiz

ures

Sat

o et

al.

2002

Pro

spec

tive

stud

y

seve

n no

nepi

lept

ic

adul

ts m

ean

age

59

year

s

Iv-in

duct

ion

Mai

nten

ance

1, 1

.5 a

nd

2 M

AC

sev

o in

= 2

/air

Pre

med

icat

ion

diaz

epam

EC

oG g

rid e

lect

rode

s

front

ally

and

tem

pora

lly

l.a b

and

pass

50

Hz

Spi

ke-a

ctiv

ity m

ax/m

in

anal

ysed

Epi

lept

iform

spi

ke a

ctiv

ity

and

B-S

in a

ll w

ith 1

.5–

2 M

AC

sev

o

End

o et

al.

2002

Pro

spec

tive

stud

y

ten

epile

ptic

adu

lts

Iv-in

duct

ion

(pro

pofo

l)

mai

nten

ance

with

0.5

1.5

MA

C s

evo

in O

2/air

EC

oG 1

6–64

ele

ctro

des

in g

rid th

roug

h se

izur

e

focu

s

Ban

d ba

ss 6

0 H

z

Spi

ke a

ctiv

ity m

ax/m

in

anal

ysed

Dur

ing

1.5

MA

C s

evo

spik

e

activ

ity d

ecre

ased

to 1

4/m

in

com

pare

d w

ith b

asel

ine

38/m

in

Page 45: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

43

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Nie

min

en e

t al.

2002

Pro

spec

tive

stud

y

thirt

y he

alth

y ch

ildre

n

aged

3–8

yea

rs

Iv-in

duct

ion

thio

pent

al

Mai

nten

ance

with

sev

o Fi

2%

in O

2/ai

r

Pre

med

icat

ion

iv m

idaz

olam

5-ch

anne

l EE

G

(acc

ordi

ng to

10–

20)

Sam

plin

g 27

9 H

z

No

epile

ptifo

rm E

EG

-

chan

ges

Jääs

kelä

inen

et a

l.

2003

Pro

spec

tive

rand

omiz

ed s

tudy

sixt

een

heal

thy

men

Mai

nten

ance

with

1, 1

.5,

2.0

MA

C/E

C50

of e

ither

sev

o

or p

ropo

fol

4-ch

anne

l EE

G

SE

F 95

% a

nd p

eak

frequ

ency

Maj

or e

pile

ptifo

rm E

EG

-

chan

ges

and

B-S

in a

ll un

der

sevo

1.5

–2 M

AC

No

epile

ptifo

rm E

EG

chan

ges

but B

-S in

all

unde

r

prop

ofol

1–1

.5–2

EC

50.

Kur

ita e

t al.

2005

Pro

spec

tive

cros

sove

r stu

dy

11 e

pile

ptic

pat

ient

s

Mai

nten

ance

with

sev

o

1.5

MA

C w

ith 5

0% N

2O o

r

with

out N

2O

EC

oG

N2O

sig

nific

antly

dim

inis

hed

the

spik

e fre

quen

cy in

EC

oG

Vak

kuri

2005

P

rosp

ectiv

e

rand

omiz

ed s

tudy

31 a

dults

Ane

st in

d:pr

opo

Rap

id in

crea

se

1) s

evo

7% o

r

2) d

es 1

8%

Dia

zepa

m p

rem

edic

atio

n

BIS

E

pile

ptifo

rm E

EG

(S, P

S,

PE

D) w

ith 8

/15

in s

evo

grou

p

but n

one

in d

es g

roup

Sha

rp in

crea

se o

f HR

in

des

grou

p, g

radu

al

incr

ease

in s

evo

grou

p

San

din

et a

l.

2008

Pro

spec

tive

stud

y

ten

youn

g he

alth

y

adul

ts

Mai

nten

ance

with

1, 1

.5 a

nd

2 M

AC

sev

o in

oxy

gen-

air

BIS

A

t 2 M

AC

sev

o hi

gh B

IS

valu

es in

dica

ting

wak

eful

ness

due

to

epile

ptog

enic

act

ivity

At 2

MA

C s

evo

HR

chan

ged

sign

ifica

ntly

, but

not B

P, a

nd o

ne p

atie

nt

had

conv

ulsi

ons

Fuku

i et a

l.

2010

Pro

spec

tive

rand

omiz

ed s

tudy

twen

ty-tw

o ep

ilept

ic

brai

n tu

mor

pat

ient

s

aged

13

to 6

7 ye

ars

Mai

nten

ance

of s

evo

or

isof

lura

ne 1

.5–2

Mac

in O

2-ai

r

nitra

zepa

m p

rem

edic

atio

n

EC

oG g

rids

on fr

onta

l

corte

x

In is

oflu

rane

gro

up n

o

rem

arka

ble

findi

ngs

whi

le in

sevo

gro

up p

arox

ysm

al E

EG

activ

ity in

thre

e pa

tient

s

toge

ther

with

rCB

F15 in

crea

se

Page 46: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

44

Stu

dy

Type

of s

tudy

S

tudy

pro

toco

l M

onito

ring

of E

EG

E

EG

find

ings

C

linic

al/o

ther

find

ings

Sch

ultz

et a

l.

2012

Pro

spec

tive

stud

y

seve

nty

AS

A I-

II

child

ren

aged

7 to

96

mon

ths

Indu

ctio

n w

ith 8

% s

evo

ad

LOC

(mea

n 1–

2 m

in)

mai

nten

ance

with

4%

insp

ired

conc

entra

tion

Mid

azol

am p

rem

edic

atio

n

EE

G m

onito

r Nar

cotre

nd

(two

elec

trode

s fro

ntal

ly,

from

whi

ch ra

w E

EG

)

Four

teen

chi

ldre

n ha

d

epile

ptifo

rm E

EG

pat

tern

s

(del

ta w

ith s

pike

s, rh

ythm

ic

PS

, PE

D) d

espi

te o

f sho

rt

indu

ctio

n tim

e

No

clin

ical

man

ifest

atio

ns

Gib

ert e

t al.

2012

Pro

spec

tive

rand

omiz

ed s

tudy

seve

nty-

nine

chi

ldre

n

aged

3 to

11

year

s

Mai

nten

ance

sev

o in

100

% O

2

or in

50%

N2O

-air

to

dete

rmin

e M

AC

of s

evo

asso

ciat

ed w

ith m

ajor

epile

ptifo

rm E

EG

sig

ns (M

ES

)

BIS

(with

fron

tal r

aw E

EG

cana

l, ba

nd p

ass

of 0

.5–

47.5

Hz,

sam

plin

g ra

te o

f

128

Hz)

ET-

sevo

for M

AC

(rhy

thm

ic

S/P

S e

tc.)

with

100

% O

2 w

as

4.3%

and

with

50%

N2O

-O2

was

4.6

%

One

chi

ld h

ad to

nic-

clon

ic c

onvu

lsio

n

Julli

ac e

t al.

2013

Pro

spec

tive

rand

omiz

ed s

tudy

thirt

y-th

ree

fem

ale

patie

nts

A: s

evo

indu

ctio

n w

ith 8

% fo

r

2 m

in, m

aint

enan

ce E

T se

vo

2.5%

B: t

arge

t con

trolle

d E

T se

vo

2.5%

no p

rem

edic

atio

n

EE

G o

f Fp1

, Fp2

, T3,

T4,

C3,

C4,

O1

and

O2

filte

red

0.1–

70 H

z,

sam

plin

g 25

6 H

z

Epi

lept

iform

dis

char

ges

durin

g in

duct

ion

in o

ne

patie

nt in

gro

up A

and

in fo

ur

patie

nts

in g

roup

B

Fifte

en p

atie

nts

had

abno

rmal

mov

emen

ts

with

out s

imul

tane

ous

EE

G a

bnor

mal

ity

Kre

uzer

et a

l.

2014

Pro

spec

tive

stud

y

100

child

ren

> 1y

AS

A I-

III

A: s

evo

indu

ctio

n w

ith 8

% in

100%

O2 fo

r 3 m

in, t

hen

4%

sevo

unt

il in

tub

B: s

evo

indu

ctio

n w

ith 6

% in

100%

O2 f

or 6

min

, the

n 4%

sevo

unt

il in

tub

Mid

azol

am p

rem

edic

atio

n

Nar

cotre

nd®

Epi

lept

iform

EE

G in

76%

(38)

with

sev

o 8%

and

in 5

2%(2

6)

with

6%

Epi

lept

iform

EE

G e

arlie

r with

8% s

evo

No

clin

ical

man

ifest

atio

ns

1 in

spira

tory

frac

tion,

2 po

lysp

ikes

and

–w

aves

, 3 rh

ythm

ic p

olys

pike

s, 4

perio

dic

epile

ptifo

rm d

isch

arge

s, 5

supp

ress

ion

with

spi

kes,

6 bu

rst s

uppr

essi

on,

7 hy

perv

entil

atio

n, 8

spon

tane

ous

vent

ilatio

n, 9

sevo

flura

ne, 10

isof

lura

ne, 11

hal

otha

ne, 12

intra

cran

ial p

ress

ure,

13 e

nd-ti

dal,

14 e

lect

roco

rtico

gram

, 15

regi

onal

cer

ebra

l blo

od fl

ow

Page 47: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

45

2.3.2 Isoflurane

Isoflurane (1-chloro-2, 2, 2-trifuoroethyl difluoromethyl ether) is a chemical

isomer of enflurane with a pungent ethereal odour. The major targets for the effect

of isoflurane are GABAA, two-pore potassium channels, glycine and serotonin

receptors in which it potentiates currents. Unlike enflurane, isoflurane is

considered to not possess convulsive properties. It has no organ toxicity.

Experimental and human studies suggest that isoflurane has neuroprotective

effects (Berg-Johnsen et al. 1992, Larsen et al. 1998). Fraga et al. (2003)

compared 1 MAC isoflurane and desflurane in normocapnic patients undergoing

removal of supratentorial brain tumors without midline shift and did not detect

any variations in ICP. Case reports of tremor, clonus and clinical seizures during

isoflurane have been documented but no clinical study has reported EEG

abnormalities during isoflurane anaesthesia. In epileptic patients, isoflurane was

associated with spike activity in a small number of patients, while no epileptic

pattern was observed in the control group (Iijima et al. 2000). A study using an

animal model of temporal lobe epilepsy (kindling) suggested that isoflurane has

an intensifying effect on electrically evoked seizures (Veronesi et al. 2008). The

authors speculated that isoflurane has the potential to impact on the secondary

generalization of epilepsy during the anaesthesia recovery period. In a study of

epileptic patients with electrocorticogram (ECoG) monitoring, however,

isoflurane decreased the mean frequency of epileptiform spikes compared to N2O

in oxygen alone, while enflurane produced increasing paroxysms of high-voltage

spikes compared to N2O in oxygen alone (Ito el al. 1988).

2.3.3 Propofol

Propofol (2, 6-diisopropylphenol), which belongs to the alkylphenol group, is the

most frequently used intravenous anaesthetic agent with a rapid onset of action

and quick recovery. Alkylphenols are not water-soluble but highly liposoluble.

Clinically propofol is available in 1% and 2% formulations.

Unconsciousness following propofol administration results from a

potentiation of the gamma-aminobutyric acid (GABA)-induced chloride current

in the GABAergic interneurons of the cortex, the thalamic reticular nucleus, and

arousal centers in the midbrain and pons. Atonia associated with propofol

administration has been suggested to be attributed to actions on GABAergic

neural systems in the spinal cord and in the pontine and medullary reticular nuclei

Page 48: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

46

(Kungys et al. 2009). In addition to the GABAA receptors, propofol potentiates

synaptic transmission in glycine receptors and has a minor inhibitive effect on

NMDA channels, voltage-gated potassium channels, and on nicotinic and

muscarinic channels (Kungys et al. 2009).

Alkire et al. (1995) reported in a positron emission tomography (PET) study

that propofol produced a global metabolic depression of the central nervous

system (CNS) by decreasing cortical metabolism more than subcortical

metabolism.

Auditory input is believed to be the final sensory modality to be blunted

during anaesthesia. Dueck et al. (2005) observed using propofol anaesthesia in a

fMRI study that propofol bilaterally attenuates the auditory-induced BOLD signal

of the auditory cortex in a dose-dependent manner. Complex analysis of the

auditory input was impaired even at low propofol concentrations, whereas basic

auditory information was still processed at high levels of sedation up to the

2.0 µg/ml plasma target concentrations (Dueck et al. 2005).

Propofol is considered to be an anticonvulsant, as modulation of neuronal

activity by propofol results from a depression of the excitatory mechanism, i.e. a

prolongation of GABAA-receptor function at GABAergic synapses, and an

enhancement of inhibitory transmission, i.e. the selective suppression of persistent

sodium channels and high-voltage-activated calcium channel conductance in

cortical neurons (Martella et al. 2005). Experimental studies in models of cerebral

ischaemia have provided evidence of the possible neuroprotective effects of

propofol through the prevention of an increase in neuronal mitochondrial swelling

(Adembri et al. 2006).

Propofol has been widely used for the treatment of patients with status

epilepticus. Spontaneous dystonic movements were found, however, during

propofol anaesthesia induction in children by Borgeat et al. (1991). They used

EEG registration but did not observe any cortical epileptiform activity, while the

movements occurred coincident with the appearance of slow delta waves in the

EEG. These movements may rather be related to subcortical structures. In their

systematic review of 512 case reports suggesting the use of propofol may be

associated with seizure-like phenomena, Walder et al. (2002) accepted 11 patients

with epilepsy and 70 patients without epilepsy to the final analysis and found

mainly seizure-like phenomena (SLP) such as tonic-clonic seizures, involuntary

movements, opisthotonus mainly on induction or emergence from anaesthesia. It

Page 49: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

47

was noteworthy, however, that none of the patients underwent EEG registration

during SLP and EEG registration was only performed afterwards in 32% of cases.

The painful electrical stimulation of a peripheral nerve during deep propofol

anaesthesia induced a cortical response with four successive components, each

representing an at least partly independent cerebral mechanism. The B-S pattern

evoked by propofol was found to be different from that seen during isoflurane or

sevoflurane anaesthesia. Evoked potentials characteristic of propofol were also

recorded (Huotari et al. 2004). When propofol is administered in doses sufficient

to produce a B-S pattern in the EEG, there is a concern as to the haemodynamic

consequences. Doses of propofol sufficient to silence the EEG are associated with

venodilatation and myocardial depression, but the haemodynamic risk in healthy

patients is minimal if filling pressures are maintained with a crystalloid infusion

(Illevich et al. 1993).

2.3.4 Nitrous oxide

Nitrous oxide is frequently used as an adjunct to general anaesthesia. It

potentiates the depressant effects of both volatile and intravenous anaesthetics.

Nitrous oxide, similar to another NMDA receptor antagonist, ketamine, activates

the EEG (Maksimov et al. 2006), however, and increases cerebral blood flow

velocity (CBFV) by increasing the cerebral metabolic rate (CMR) (Matta & Lam

1995, Wilson-Smith et al. 2003). During isoflurane B-S steady-state anaesthesia,

supplementation with 65% nitrous oxide activated the EEG by decreasing

suppression periods and by increasing the duration of bursts (Yli-Hankala et al. 1993a). When nitrous oxide at 70% was added to propofol at steady-state EEG

suppression anaesthesia, the CBFV of the middle cerebral artery increased and

occasional burst activities of the EEG were also observed (Matta & Lam 1995).

Furthermore, in children aged 1.5 to 6 years anaesthetized with propofol in 35%

oxygen in air, the CBFV of the middle cerebral artery increased by 12% when air

was replaced with nitrous oxide (Wilson-Smith et al. 2003). Porkkala et al. (1997)

recognized activation of the EEG during stable anaesthesia maintenance with 1.9

ET isoflurane concentrations in a 60% air-oxygen mixture at B-S level when the

air was replaced with nitrous oxide. The proportion of EEG suppressions

decreased from 53% to 34%. At the same time, nitrous oxide induced a reduction

in the amplitude of cortical N20 component by 69% while the amplitude and the

latency of earlier components remained unchanged. Lam et al. (1994) observed

that the use of 0.5 MAC isoflurane together with 0.6 MAC (60%) nitrous oxide

Page 50: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

48

was associated with a higher CBFV of the middle cerebral artery and an increased

cerebral arterio-venous oxygen content difference (AVDO2) reflecting an

increased cerebral metabolic rate compared to equipotent 1.1 MAC isoflurane

alone in healthy males.

Nitrous oxide has been shown to decrease epileptogenic patterns in the EEG

of epileptic patients (Kurita et al. 2005). An earlier study (Hosain et al. 1996) did

not demonstrate any difference in spike activity with or without nitrous oxide 60–

70% at low concentrations of isoflurane 0.25–0.5%.

2.3.5 Summary of the neurophysiological effects of anaesthetics

The effects of anaesthetics on neurophysiological information such as the

electroencephalogram, sensory evoked potentials, auditory evoked potentials and

motor evoked potentials are summarized and expressed in Table 3. The idea

behind neurophysiological monitoring is to improve the safety of the patient in

the demanding conditions of special operations.

Page 51: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

49

Ta

ble

3. S

um

ma

ry o

f th

e n

eu

rop

hy

sio

log

ica

l eff

ects

of

an

aes

the

tics

. S

um

ma

ry o

f th

e n

eu

rop

hys

iolo

gic

al

eff

ec

ts o

f h

yp

no

tics

in

mo

no

an

ae

sth

es

ia a

t s

urg

ica

l le

ve

l (1

MA

C o

r h

igh

er

for

vo

lati

le a

nae

sth

eti

cs

). S

low

wa

ve

sle

ep

is

in

clu

de

d f

or

co

mp

ari

so

n,

as

th

e

EE

G p

att

ern

s a

nd

th

e e

ffe

ct

on

so

ma

tos

en

so

ry a

nd

mo

tor

res

po

ns

es o

f s

pec

ific

GA

BA

A a

go

nis

ts a

nd

alp

ha-2

ag

on

ists

are

pro

ba

bly

ca

use

d p

art

ly b

y t

he

sam

e m

ech

an

ism

s.

On

th

e o

the

r h

an

d,

aro

us

al

ch

an

ge

s a

ll t

he

se

neu

rop

hy

sio

log

ica

l m

eas

ure

s

tow

ard

s a

wa

ke p

att

ern

s,

alt

ho

ug

h w

e o

nly

wa

ke

up

fro

m p

hy

sio

log

ica

l s

lee

p.

SE

P r

efe

rs m

ain

ly t

o t

he

sh

ort

-la

ten

cy

co

rtic

all

y

ge

ne

rate

d w

ave

s a

nd

au

dit

ory

ev

ok

ed

po

ten

tia

l (A

EP

) m

ain

ly t

o c

ort

ica

l m

id-l

ate

nc

y A

EP

s.

B-S

, B

urs

t-s

up

pre

ss

ion

. (S

loa

n &

ntt

i (

20

08

) A

ne

sth

eti

c e

ffe

cts

on

evo

ke

d p

ote

nti

als

, in

Han

db

oo

k o

f C

lin

ica

l N

eu

rop

hy

sio

log

y.

Re

pri

nte

d w

ith

th

e p

erm

iss

ion

of

the

co

py

rig

ht

ho

lde

r).

Mec

hani

sm o

f act

ion

Hyp

notic

E

EG

S

EP

A

EP

M

EP

Spe

cific

GA

BA

ago

nist

P

ropo

fol

Spi

ndle

s, v

erte

x-w

ave,

B-S

↓↓

E

tom

idat

e S

pind

les,

ver

tex-

wav

e, B

-S

↑ ↓↓

GA

BA

and

oth

ers

Hal

otha

ne

B-S

var

iabl

e ↓↓

↓↓

↓↓

Is

oflu

rane

B

-S

↓↓

↓↓

↓↓

E

nflu

rane

B

-S, s

eizu

res

↓↓

↓↓

↓↓

S

evof

lura

ne

B-S

, sei

zure

s ↓↓

↓↓

↓↓

D

esflu

rane

B

-S

↓↓

↓↓

↓↓

B

arbi

tura

tes

B-S

, epi

lept

iform

pat

tern

s ↓↓

↓↓

↓↓

Alp

ha 2

ago

nist

C

loni

dine

S

low

? ↓

D

exm

edet

omid

ine

Slo

w

↓ ?

NM

DA

ant

agon

ist

Nitr

ous

oxid

e Fr

onta

l bet

a ↓↓

-

↓↓

K

etam

ine

Thet

a ↑

- ↓

X

enon

C

entra

l slo

w

↓ ↓

Slo

w w

ave

slee

p

Spi

ndle

s, v

erte

x-w

ave

↓ ↓

Page 52: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

50

2.4 EEG during physiological sleep and general anaesthesia

Loomis et al. (1935a, 1935b) first described the EEG patterns during sleep states

and during disturbance of sleep including the K-complex (Loomis et al. 1938), a

large potential change as a result of tone stimulation. Furthermore, Davis et al. (1939) precisely described the single components of the K-complex.

Sleep and anaesthesia may have many common mechanisms, but distinct

behavioural and physiological differences clearly distinguish these two states.

Sleep is a state associated with loss of reactivity to surroundings or

unconsciousness regulated both homeostatically and via circadian rhythm and

may be disturbed by environmental factors, but the depth and duration of general

anaesthesia is influenced by anaesthesia agent dosage and duration of

administration (Tung & Mendelson 2004). In general anaesthesia, arousal

mechanisms become weaker, whereas physiological sleep allows arousal and is

broken by external stimuli (Tung & Mendelson 2004).

GABAergic anaesthetics have many EEG effects similar to that of

physiological non-rapid eye movement (NREM) or slow wave sleep. EEG

oscillations mimicking NREM physiological sleep such as sleep spindles, delta-

waves and slow waves have been observed during propofol anaesthesia (Huotari

et al. 2004). Nelson et al. (2002) emphasized the role of the tuberomamillary

nucleus in the sedative response to GABAergic anaesthetics. Dexmedetomidine, a

selective alpha2-adrenoceptor agonist, caused slow wave sleep-like EEG patterns

mimicking physiological sleep-promoting factors (Nelson et al. 2003). The

researchers observed that the sedative properties of dexmedetomidine involve an

inhibition of the locus ceruleus, which inhibits ventrolateral preoptic nucleus

firing, thus increasing GABA release and inhibiting tuberomamillary nucleus

firing (Nelson et al. 2003). Huupponen et al. (2007) also used dexmedetomidine

and found that at a sedation dose it produced a state closely resembling

physiological light sleep in humans. Sleep and anaesthesia share common

regulatory mechanisms, and interactions between them produce the possibility

that certain anaesthetics may facilitate sleep in environments where sleep

deprivation is common (Tung et al. 2004b, Drouot et al. 2008).

At subanaesthetic doses, both intravenous and inhaled hypnotics induce

rapid, mainly beta frequency EEG oscillations: initially mainly in the frontal

regions, but then spreading to the more posterior regions. As anaesthesia is

deepened, an increase in the amplitude and a decrease in the frequency of the

Page 53: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

51

EEG occur as slow theta and delta waves proliferate. Under deep anaesthesia with

supra-anaesthetic doses, most hypnotics suppress cortical activity and induce B-S

in the EEG and when anaesthesia is further deepened, suppression alone. (Clark

& Rosner 1973, John & Prichep 2005).

Non-rapid eye movement (NREM) sleep is associated with synchronized

activity across large areas of the brain, including well-defined EEG oscillations

such as sleep spindles, delta-waves, and slow cortical oscillations. The transition

from wakefulness to NREM sleep is associated with typical signs of brain

electrical activity, characterised by prolonged periods of hyperpolarization and

increased membrane conductance in thalamocortical (TC) neurons (Sinha 2011).

The early stage of quiescent sleep is associated with sleep spindles in the

EEG, waxing and waning spindle waves occurring at a frequency of 7 to 14 Hz

typically lasting for 2–3 seconds and repeated once every 3 to 10 seconds during

NREM sleep, especially in stages 2 and 3. In adults, sleep spindles are most

commonly seen over the central regions, near electrodes C3 and C4 of the

international 10–20 system. In children, slower 10–12 Hz spindles with a more

anterior prominence are observed. The reticular nucleus (RE) of the thalamus and

its connection to the dorsal thalamus appears to be responsible for generating

sleep spindles. The spindle oscillation generated in the RE cells is transferred to

thalamocortical relay cells and drives the cortical cells which produce the EEG

spindles (Steriade et al. 1994, Contreras et al. 1996, Sinha 2011).

As sleep deepens, waves with slower frequencies of 1 to 4 Hz appear on the

EEG. These delta waves were shown to arise between cortical layers 2 to 3 and 5

(Steriade et al. 1993b). The thalamus is also involved in the generation of this

rhythm. The hyperpolarization of thalamocortical cells is a critical factor in the

generation of delta oscillations. Delta oscillations occur when the thalamocortical

cells are more hyperpolarized than they are during the spindle oscillations

(Steriade et al. 1993c).

The low-frequency sleep oscillations of 0.5–1 Hz of NREM sleep originate in

the cortex and were first described by Steriade and co-workers (1993b, 1993c) in

anaesthetized cats. The slow oscillations consist of rhythmic alternating

membrane potentials between depolarizing components of 0.4–0.8 seconds

duration separated by prolonged hyperpolarization levels lasting 0.2–0.8 seconds.

Prolonged hyperpolarizations are seen in the surface EEG as high amplitude

negative field potentials. Acherman & Borbely (1997) and Amzica & Steriade

(1997) investigated the slow oscillation in human sleep. Using high-density EEG

recordings in humans Massimini et al. (2004) showed that sleep slow oscillations

Page 54: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

52

are travelling waves which sweep the human cerebral cortex up to once per

second as sleep deepens. Each slow oscillation has a definite site of origin and

direction of propagation, which vary from one cycle to the next. Slow oscillations

originate more frequently at anterior cortical regions and propagate in an

anteroposterior direction (Massimini et al. 2004). The electrodes with the highest

probability of detecting a slow oscillation were concentrated in a scalp region

anterior to the central midline electrode, and posterior to the anteriofrontal

midline electrode, projecting onto cortical areas 8 and 9. Slow oscillations can

occur during any stage of NREM sleep (Massimini et al. 2004). The increase in

slow wave activity after sleep deprivation is highest in the anterior prefrontal

regions (Finelli et al. 2000).

The K-complex, which is unique to NREM sleep, consists of an ample

surface-positive transient followed by a slow surface-negative component, which

may be followed by a spindle wave (Davis et al. 1939, Amzica & Steriade 1997).

The K-complex emerges from a cortically generated slow oscillation, occurs

spontaneously, but can also be evoked by sensory stimuli (Amzica & Steriade

1997, Sinha 2011). The same circuit that generates sleep spindles is also thought

to play a role in the 1–4 Hz delta oscillation which has both a cortical and a

thalamic component. The slow oscillation is thought to form the basis for the K

complex, which occurs at periodic intervals in all stages of NREM sleep. K

complexes are thought to represent fluctuating arousals and can be provoked by

external sensory stimuli and intrinsic sleep oscillations (Sinha 2011).

REM sleep, associated with rapid eye movements and dreaming episodes, is

characterised by an abolition of low-frequency oscillations with a relatively

desynchronized EEG resembling the waking state and an increase in cellular

excitability, although motor output is markedly inhibited. Activation of the

thalamo-cortical system by ascending brainstem cholinergic arousal systems

during the steady depolarisation of cortical cells in waking and REM sleep

suppresses the autonomous slow oscillations and increases activities within both

beta and gamma frequencies (Steriade et al. 1993a, Steriade 2006). These fast

oscillations may also appear during light anaesthesia and, sometimes, during

NREM sleep (Steriade 2006).

Decreased blood flow in the thalamus and in the prefrontal and multimodal

parietal association cortices accompanies the onset and deepening of NREM

sleep.

Page 55: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

53

REM sleep is associated with an increase in blood flow in the pons, midbrain

and thalamus, amygdala, hypothalamus and basal ganglia. Activated thalamic

nuclei, which occupy key sites in sensory-relay and other brain circuits, transmit

endogenous stimuli, which results in the sensory phenomena of dreaming

(Hobson & Pace-Schott 2002).

The EEG oscillations which occur during sleep and arousal states are

generated in the thalamus and cerebral cortex, two regions intimately linked by

means of reciprocal projections (Steriade 2006). The thalamus is the major

gateway for the flow of information toward the cerebral cortex and is the first

station at which incoming signals can be blocked by synaptic inhibition during

sleep (Steriade 2006). This mechanism contributes to the shift that the brain

undergoes as it changes from an aroused state, open to signals from the outside

world, to the closed state of sleep (Hobson & Pace-Schott 2002).

2.5 Epileptiform EEG, anaesthesia and sleep

While decreasing activity in the central nervous system (CNS), some general

anaesthetics can also paradoxically provoke cortical seizures in the EEG in deeply

anaesthetized patients (Modica 1990a, 1990b, Faizo et al. 2014). It has been

proposed that proconvulsant drugs may act by decreasing the amplitude of

miniature inhibitory postsynaptic currents (IPSCs) or by eliciting a greater

calcium-induced presynaptic mobilization of excitatory neurotransmitters (Wilson

et al. 2006). Epileptiform EEGs have been detected, for instance, during

enflurane (Neigh et al. 1971, Rosen & Söderberg 1975, Burchiel et al. 1977,

Jäntti & Yli-Hankala 1990) and sevoflurane anaesthesia (Yli-Hankala et al. 1999,

Kaisti et al. 1999, Vakkuri et al. 2000, Vakkuri et al. 2001, Jääskeläinen et al. 2003). Vakkuri et al. (2005) also found epileptiform EEGs and tachycardia during

deep sevoflurane anaesthesia, but tachycardia was not associated with

epileptiform EEG during desflurane anaesthesia. Compared with isoflurane, the

proconvulsant properties of enflurane may be due to its potency to reduce the

peak magnitude of synaptically induced IPSCs to a greater extent than isoflurane

(Banks & Pearce 1999).

Epileptic seizures mainly develop during slow wave sleep. During sleep

initiation, thalamic activity changes are a response to the characteristic cortical

synchronization observed in sleep. Such thalamic neuronal activity changes are

related to the action of GABA systems from the thalamic reticular nucleus

inducing a bursting pattern, which might account for the cortical

Page 56: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

54

hypersynchronization observed during sleep initiation. The experiments of

Steriade et al. (2003) using multi-site, extra- and intracellular recordings, showed

a transformation without discontinuity from sleep patterns to seizures. The

spindle oscillations of physiological sleep were related to the development of the

spike-and-wave EEG complexes, which were associated with absence (petit mal)

epileptic seizures. Steriade et al. (1993a) stated that while the reticular thalamic

nucleus is central to the genesis of spindle oscillations, decreasing or abolishing

the inhibitory efficacy of the RE upon thalamocortical cells would decrease the

incidence of epileptic spike-and-wave discharges. In their prospective study using

continuous EEG monitoring with automated seizure detection in patients with

partial-onset seizures, Herman et al. (2001) found that frontal lobe seizures were

most likely to occur during sleep while seizures arising from the parietal or

occipital area rarely occurred during sleep. Temporal lobe complex partial

seizures were more likely to secondarily generalize during sleep than during

wakefulness. The researchers concluded that hypersynchrony - particularly during

light stage 2 NREM sleep - facilitated both the initiation and propagation of

partial seizures, but the synchronizing and activating effects of sleep on various

brain regions could differ. No seizures occurred during REM sleep and they were

also rare during the deeper NREM sleep stages.

Arousal state transitions are also known to promote the occurrence of

epileptiform phenomena or seizures in patients with epilepsy (Contreras &

Steriade 1995). The transitional state occurs during drowsiness, especially

between waking and NREM sleep. It is of remarkable interest that spike- and

wave seizure patterns are totally absent or significantly decreased during REM

sleep (Steriade 2006).

Page 57: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

55

3 Aims of the study

1. To examine whether median nerve SSEP cortical short latency waveforms

can be detected without averaging at EEG burst-suppression level during

sevoflurane anaesthesia and to compare those with SSEPs recorded awake.

2. To analyse temporal and spatial relationships between EEG and BOLD

signals during the dynamic induction of focal epileptic seizures at EEG burst-

suppression level of deep isoflurane anaesthesia in an experimental epilepsy

model in pigs.

3. To obtain a better understanding of electric fields during EEG burst-

suppression of propofol anaesthesia in humans by using both scalp and depth

electrode recordings.

4. To describe the topography of EEG features during sevoflurane mask

induction anaesthesia in children and to evaluate, whether BIS can recognize

these waveforms.

5. To describe the electric fields of burst-suppression and epileptiform patterns

in sevoflurane anaesthesia during spontaneous breathing and controlled

hyperventilation in male patients and to compare these patterns to

cardiovascular parameters.

Page 58: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

56

Page 59: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

57

4 Subjects and Methods

Study I was a prospective monoanaesthesia study with sevoflurane in adult

patients. Cortical evoked responses to median nerve stimulation were recorded

under EEG B-S anaesthesia levels.

Study II was an experimental epilepsy model with EEG registration together

with fMRI under EEG B-S level of isoflurane anaesthesia in piglets.

Study III was a prospective study where EEG was registered under

anaesthesia maintenance with an intravenous anaesthetic propofol during EEG B-

S levels. Both scalp electrodes and deep brain electrodes in the subthalamic

nucleus were used in patients with Parkinson`s disease.

Study IV was a prospective anaesthesia induction study with EEG

registration under sevoflurane anaesthesia using normoventilation in children.

Study V was a prospective randomized study in adult males with EEG

registration under induction of sevoflurane anaesthesia. The patients were

randomized either to normoventilation or hyperventilation groups. The closed

envelopes used for randomization were opened immediately prior to the induction

of anaesthesia in the operation room. The neurophysiologist (V.J.) who examined

the EEG was blinded to the study groups.

The summary of subjects and methods are summarised in Table 4.

4.1 Subjects

Study I was based on 21 ASA I-II risk classification patients, aged 22 to 52 years,

scheduled for routine surgery. In study III, three Parkinson’s patients underwent

stimulator implantation for electrical stimulation of the subthalamic nucleus for

treatment of Parkinson`s disease. Study IV involved 20 children aged 4–10 years

undergoing otolaryngological or orthopaedic surgery. The children were of the

ASA I risk classification group with no previous history of febrile convulsions or

other neurological disorders. Twenty ASA I risk group men aged 23–52 years

undergoing cervical or lumbar disc operation participated in study V. Study II was

performed on seven 2–3 month-old female pigs.

The clinical studies I, III, IV and V were approved by the Oulu University

Ethics Committee and written informed consent was obtained from all patients (I,

III, V) or from the parents of the children (IV). In study II, all experiments were

performed in compliance with the guidelines of the European Convention for the

Protection of Vertebrate Animals used for Experimental and Other Scientific

Page 60: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

58

Purposes (1986) and in compliance with the European Union Directive

86/609/EEC (1997). Study II was approved by the Animal Care and Use

Committee of the University of Oulu.

Table 4. Summary of the subjects and methods.

Study Subjects N Age EEG monitoring Other measurements

I 5 females

16 males

21 22–52 years,

mean 35

8-channel HR, blood pressure, SaO2, ETsevo,

ETCO2, inspiratory O2

II piglets 7 2 – 3 months 4-channel,

fMRI

HR, blood pressure, SaO2,

ETisoflurane, MV, ETCO2,

III Parkinso

n’s

patients

3 40, 44, 67

years

18-channel, 1–2

depth electrodes

with 4 contacts

each

HR, blood pressure, SaO2, ETCO2,

inspiratory O2, MV

IV children 20 4–10 years,

mean 6.9

32-channel

HR, blood pressure, SaO2, ETsevo,

ETCO2, inspiratory O2

V males 20 23–52 years,

mean 42

32-channel HR, blood pressure, SaO2, ETsevo,

ETCO2, inspiratory O2 and N2O,

blood-gas analysis

4.2 Variables and measurements

Haemodynamic and ventilatory monitoring was performed using the Datex-

Ohmeda AS/3 Anaesthesia Monitor (Datex-Ohmeda division, Instrumentarium,

Helsinki, Finland), and consisted of heart rate, electrocardiography, non-invasive

blood pressure, oxygen saturation with pulse oximetry, end-tidal carbon dioxide

pressure, end-tidal sevoflurane concentration, end-tidal isoflurane concentration,

inspiratory oxygen and nitrous oxide concentrations. Ventilatory and anaesthesia

gas measurements were performed from the ventilatory circuit, with the

connecting piece close to the face mask. Haemodynamic and gas measurements

were performed once a minute during anaesthesia.

4.3 Anaesthesia

In study I, anaesthesia of the adult patients was induced by mask with sevoflurane

in 100% oxygen and they were intubated after induction under deep sevoflurane

anaesthesia at a level of steady-state burst-suppression achieved at 1.5–2.5 MAC.

Anaesthesia was maintained with sevoflurane in 40% oxygen in air. Four patients

Page 61: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

59

required muscle relaxation with succinyl choline due to spontaneous ventilation

after a precurarisation dose of vecuronium.

In study II, after a fasting period of 12 hours and premedication using

1.5 mg/kg midazolam and 15 mg/kg ketamine, the animals were anaesthetised

with thiopental 25 mg/kg. After intubation, anaesthesia was maintained using

isoflurane at an end-tidal concentration of 1.4–1.8% in 40% oxygen/air, with the

EEG burst-suppression pattern used as an end-point. Normocapnia was

maintained during mechanical ventilation.

Patients in study III were pre-loaded with 500 ml of hydroxyethyl starch and

NaCl 0.9% 1 000 ml in order to prevent vasodilation-induced hypotension.

Muscle relaxation was induced and maintained with rocuronium bromide.

Anaesthesia was maintained and adjusted with a propofol infusion at a rate of 12–

20 mg/kg/h to maintain the EEG at the burst-suppression level. Ventilation was

controlled with air/oxygen 60% /40%. Normocapnia (end-tidal CO2 between 4.8

and 5.5 kPa) was maintained during mechanical ventilation. Phenylephrine was

administered either as boluses or as an infusion if mean arterial pressure dropped

under 60 mmHg.

The children in study IV were premedicated with oral midazolam 0.4 mg/kg,

up to a maximum dose of 10 mg, and 0.005 mg/kg glycopyrrolate was

administered i.v. after peripheral vein cannulation. Anaesthesia was induced with

sevoflurane inhalation using a semi-open anaesthesia system primed with a fresh

gas flow of at least 6 L/min (nitrous oxide 3 L/min and oxygen 3 L/min) and the

sevoflurane vaporizer (Sevorane Abbott Vapor 19,3; Draegerwerk AG, Lubeck,

Germany) set at a maximum of 8%. The induction phase lasted for 8 min at

maximum. Ventilation was manually assisted after spontaneous ventilation had

ceased. End-tidal CO2 was kept above 4.5 kPa in order to maintain

normoventilation. Mivacuronium was given to facilitate intubation. Thereafter the

sevoflurane vaporizer was adjusted to 2% with an oxygen-nitrous oxide ratio at

1:2 and EEG registration continued for a further three minutes.

The patients in study V were randomly allocated to receive either controlled

hyperventilation (CH) or spontaneous ventilation (SP) during induction.

Anaesthesia was induced via a face mask with sevoflurane (8% in nitrous oxide

50% in oxygen) using a semi-open system primed with a fresh gas flow of

10 L/min. Ventilation of the patients in the hyperventilation group was manually

assisted to maintain an end-tidal CO2 concentration below 4 kPa while in the

normoventilation group spontaneous ventilation was allowed but assisted if

needed to keep end-tidal CO2 below 6.5 kPa. At the end of the 5 min induction

Page 62: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

60

phase, a blood gas sample was obtained from a radial artery using a single-

puncture method. Thereafter the trachea was intubated without muscle relaxation.

4.4 EEG Recordings and analyses

In study I, the EEG data was collected with a Nicolet Viking IV P

electromyography, using the IOM programme, electrodes at Fp1, F3, FC3, C3,

CP3, P3, O1, F4, C4, P4, O2, ear electrode A2 and extracerebral electrodes at

neck C7 or the shoulder contralateral to the stimulus. Due to the limited number

of channels, different recording montages were used for different patients. Eight

channels were recorded: one was used to monitor EEG on screen to detect burst-

suppression pattern, one recorded the electrical stimulation at the wrist, and 6

were used to continuously record the EEG on to a hard disk at a sampling

frequency of 1 000 Hz. The amplifier bandwidths were 0.2–100 Hz. A custom-

made analysis program was used to mark bursts and suppressions onsets and

classify responses. This program also configurated montages and averaged

selected sweeps.

EEG registration using two electrodes and a ground electrode was employed

in study II. The fMRI terminology and methods visualizing function, i.e.

perfusion, diffusion and BOLD imaging were covered in detail by Mäkiranta in

her dissertation in 2004. The EEG data was collected on the hard disk of the

NeuroScan™ recorder with the following amplifier set-up parameters: DC-

recording, 0–200 Hz bandwidth, gain 150, sampling frequency 1 000 Hz, range

37 mV, accuracy of 0.559 μV. The EEG electrodes were attached with tissue glue

on the following positions: the surface of the pig skull, the right (contralateral to

the lesion) and left (ipsilateral to the lesion) side posteriorly to the coronal suture.

The equipment, cables and electrodes were compatible with the MRI

environment. The EEG was recorded continuously during the MRI session.

In study III, a Nervus™ digital EEG recorder (Taugagreining, Reykjavik,

Iceland) was used to record the EEG together with the ECG. EEG data was

registered from electrodes Fp1, Fp2, F3, Fz, F4, T3, Cz, P3, Pz, P4, O1, O2, A1,

A2, the four depth electrodes, and an electrode on the spinous process of C7.

Bandpass was 0.016–100 Hz with a sampling rate of 256 Hz (patient 1) and

512 Hz (patient 2). The EEG for patient 3 was recorded with the NeuroScan

Synamp amplifier™ using a sampling frequency of 8 000 Hz, down-sampled to

200 Hz and filtered 0–30 Hz for illustration.

Page 63: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

61

In study IV the EEG was registered together with an ECG using a digital

EEG device Nervus™ (Taugagreining, Reykjavik, Iceland) from the surface

electrodes of the international 10–20 system, with added orbitofrontal and ear

electrodes. The sampling frequencies were 256 Hz in eleven patients and 512 Hz

in nine patients. A bandpass of 5.0 Hz–70 Hz was used for spike evaluation with

an ear reference and 0.016 Hz–70 Hz was used for evaluation of slow activity,

also with an ear reference. Placing the EEG surface electrodes in the paediatric

patients took time (approximately 20–30 minutes) and was done in the operation

room. A baseline EEG was recorded with the patient awake for at least two

minutes before the onset of induction, and then continued for approximately three

minutes after the induction period. The topographies of the waveforms were

analysed with different band-pass filters and montages of the EEG. The beat-to-

beat heart rate was analysed offline.

In study V, the EEG data was collected together with an ECG using the

NeuroscanTM digital EEG device with surface electrodes according to the 10–20

system, including orbitofrontal and ear electrodes at a frequency of 2 000 Hz. A

bandpass was 0.016–134 Hz. The EEG data was stored on a computer for later

analyses. The location and topography of the waveforms were analysed by

adjusting the EEG filtrations and couplings.

4.5 Stimulation

In study I, somatosensory evoked potential (SSEP) registration was used.

Electrical stimulation during burst-suppression level was applied to the right

median nerve in 20 patients and to the left in one patient. The intensity was

adjusted to three times the sensory threshold, which was enough to produce

movement of the thumb with a duration of 0.2 ms. Trains of stimuli at frequencies

of 20, 10, 5, 4, 3, 2 and 1 Hz were applied in addition to single stimuli, and

different stimulation trains were studied in different patients due to the limited

recording time prior to the start of the operation.

4.6 Statistical analysis

In the statistical analysis of study IV, summaries of the variables were expressed

as means and standard deviation (SD) and range, or median with range. A Linear

Mixed Model (LMM) was utilized for repeatedly measured data (MAP and heart

rate) with SAS procedure mixed; the Toeplitz covariance structure was defined in

Page 64: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

62

repeated statements, and independence between subjects (children) was assumed

by random statement. In the analysis of repeatedly measured data, heart rate and

MAP were compared with children who displayed a suppression (short and/or

long suppression), and those who did not display this suppression. The p-values

were reported as follows: p-group, indicates a level of difference between groups,

p-time*group, indicates interaction between group and time. The T-test was used

to compare demographic data and values before anaesthesia induction. Statistical

analysis was performed using SPSS (SPSS, version 12.0.1, SPSS Inc, Chicago,

IL) and SAS (version 8.02, SAS Institute Inc., Cary, NC) statistical programs.

Two-tailed p-values are reported.

In the statistical analysis of study V, summaries of the variables were

presented as mean standard deviation (SD) unless otherwise stated. Repeatedly

measured data was analysed using LMM using a combination of random effects

model and covariance pattern model. If time x group interaction was significant

(p<0.05) the differences in separate time points were estimated by LMM: Simple

between group comparisons were performed using Student`s t-test or Mann-

Whitney U-test (continuous data) and Fisher`s exact test (categorical data). Two

tailed p-values were presented. Analyses were performed using SPSS (IBM Corp.

Released 2011. IBM SPSS Statistics for Windows. Version 20.0. Armonk, NY:

IBM Corp. and SAS for Windows version 9.3. SAS Institute Inc., Cary, NC,

USA).

Page 65: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

63

5 Results

5.1 SSEP in sevoflurane anaesthesia (I)

Two types of responses to median nerve stimulation could be recorded without

averaging during sevoflurane-induced EEG burst-suppression: the short-latency

response N20/P20 and partly overlapping-P22, and the long-latency burst (Fig. 5).

The percentage of stimuli evoking bursts in individual patients changed from 0 to

54%, mean 17%. The short latency N20/P20 and partly-overlapping P22 could be

recorded particularly with short derivations across the central sulcus after every

stimulus in all our patients. They could often be reliably evaluated from two or

three single sweeps with a stimulation interval exceeding one second, with

amplitudes in the higher frequencies decreasing rapidly. Even with a stimulus rate

of one per second, there was a 30% decrease in amplitude from the first to the

second stimulus (Fig. 6). The other response was a burst, which involved the

entire cortex, but did not follow every stimulus. No cortical activity was visible

between the two responses, after averaging several responses.

Fig. 5. Two types of responses without averaging to right median nerve stimulation:

the short-latency response N20 and the long-latency burst.

Page 66: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

64

Fig. 6. Adaptation of the cortical component of median nerve SSEP in sevoflurane B-S

level anaesthesia during different stimulation frequencies. This recording is from P3

to C3, presented as an average over six patients.

5.2 Epileptiform EEG pattern in sevoflurane anaesthesia (IV, V)

The rapid induction of anaesthesia using high 8% concentrations of sevoflurane in

children (study IV) produced an initial increase in beta activity, followed by a

dominancy of alpha activity and thereafter slow delta wave activity with a very

high amplitude (ad 2 050 µV, mean 1 447), which sometimes developed into

sharper waveforms. Thereafter, polyspike and -wave activity appeared in the

EEG, in which the spikes were mainly located frontally and were multifocal with

frontocentral maxima. The increase in heart rate appeared before epileptogenic

spikes. Four patients had long suppressions. The examples of EEG patterns are

expressed in Fig. 7. No symptoms of epilepsy were detected clinically. The time

interval to appearance and duration and also location of the main EEG events in

children during anaesthesia induction with sevoflurane are presented in Table 5

and Table 6.

Adult male patients with controlled hyperventilation (study V) under rapid

induction of anaesthesia with high sevoflurane concentration had epileptiform

EEG patterns (EEG gr. 2–4) in 90% of cases, such as polyspikes (PS), periodic

epileptiform discharges occurring bilaterally, or PEDs during EEG suppression

(S). Furthermore, 40% of spontaneously breathing (SB) patients also displayed

Page 67: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

65

epileptiform EEGs. No focal or generalized epileptic activity was seen in the

EEG.

Fig. 7. Examples of EEG patterns during anaesthesia induction with sevoflurane in

children.

A. Onset of suppression after polyspike and wave activity,

B. High-amplitude spike during suppression,

C. Onset of activity after long suppression,

D. Multifocal spikes frontally,

E. Short suppression during high amplitude slow wave activity,

F. High-amplitude delta activity.

Page 68: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

66

Ta

ble

5. T

ime

in

terv

al

(ap

pe

ara

nc

e)

in s

ec

on

ds (

s)

an

d l

oc

ati

on

of

ma

in e

lec

tro

en

ce

ph

alo

gra

m (

EE

G)

ev

en

ts a

nd

ag

ita

tio

n i

n

ch

ild

ren

du

rin

g i

nd

uc

tio

n o

f an

ae

sth

es

ia w

ith

se

vo

flu

ran

e. P

ati

en

ts 1

–1

0.

Pat

ient

N:o

1

2 3

4 5

6 7

8 9

10

Age

(yea

rs)

7.1

6.5

8.4

6.3

6.3

6.3

10.7

5.

7 6.

5 6.

3

Agi

tatio

n

JM1

M

2 M

M

M

M

Slo

w d

elta

appe

aran

ce

24

122

96

98

73

86

11

2 11

4 46

max

am

plitu

de (n

V)

704

1525

2048

16

02

1570

13

53

1760

15

40

1543

frequ

ency

(Hz)

2.

3 1

1.

3 1.

3 1.

3 1.

3 1.

3 1.

5 1.

5

Firs

t irr

itatio

n S

S3

SS

S

S

MS

4 S

S

MS

M

S

MS

M

S

MS

appe

aran

ce

60

180

120

60

120

60

240

180

120

60

Max

irrit

atio

n S

S

SS

S

S

S

S

M

S

S

S

PE

D

appe

aran

ce

65

212

318

184

155

300

452

473

420

120

loca

tion

F3, F

4 F3

, F4

Fp2

front

. F3

, F4

Fp1,

Fp2

Fp

i, Fp

2 fro

nt.

F3, F

4 fro

nt.

Fz

F7

F3

, F4

Fp

1, F

p2

F3, F

4, F

8 F3

, F7

Fp

1, F

p2

O1,

O2

Fz

, Cz,

C3

Sho

rt su

ppre

ssio

ns

appe

aran

ce

12

0

300

18

0

60

num

ber

2

2

10

6

dura

tion

(ms)

Long

sup

pres

sion

appe

aran

ce

42

0

480

17

7

dura

tion

29

8

299

16

1

PE

D5

P

ED

Spi

kes

durin

g lo

ng s

uppr

.

Yes

1 je

rkin

g m

ovem

ent,

2 m

ovem

ent,

3 si

ngle

spi

kes,

4 m

ultis

pike

s an

d –w

aves

, 5 pe

riodi

c ep

ilept

iform

dis

char

ges.

Page 69: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

67

Ta

ble

6. T

ime

in

terv

al

(ap

pe

ara

nc

e)

in s

ec

on

ds (

s)

an

d l

oc

ati

on

of

ma

in e

lec

tro

en

ce

ph

alo

gra

m (

EE

G)

ev

en

ts a

nd

ag

ita

tio

n i

n

ch

ild

ren

du

rin

g i

nd

uc

tio

n o

f an

an

esth

esia

wit

h s

evo

flu

ran

e. P

ati

en

ts 1

1–

20

.

Pat

ient

N:o

11

12

13

14

15

16

17

18

19

20

Age

(yea

rs)

8 9.

7 4.

6 4.

8 7.

7 8.

1 5.

5 6.

4 7

5.6

Agi

tatio

n H

1 H

, M2

H

M

M

M

Slo

w d

elta

appe

aran

ce

88

86

51

53

71

176

133

11

5 22

2

max

am

plitu

de (n

V)

1386

18

63

1663

12

54

781

1679

16

48

1032

16

17

1719

frequ

ency

(Hz)

1

1.3

1.3

1.8

2 1

1.3

1.5

1.3

1

Firs

t irr

itatio

n M

S3

MS

M

S

SS

4 S

S

MS

M

S

SS

M

S

MS

appe

aran

ce

60

90

120

60

120

240

120

120

180

150

Max

irrit

atio

n

P

ED

S

S

SS

P

ED

M

S

SS

M

S

MS

appe

aran

ce

240

30

0

420

300

16

2

180

loca

tion

F3

, F4

F3, F

4 F3

, F4

F3, F

4 Fp

1, F

p2

front

. F3

, F4

Fp1,

F3

F3, F

4 fro

nt.

Fp

1, F

p2

Fp1,

Fp2

Fz

sp

read

s

F4, F

7 Fp

1, F

7

Sho

rt su

ppre

ssio

ns

appe

aran

ce

60

120

60

300

num

ber

7 13

3

1

dura

tion

(ms)

840

Long

sup

pres

sion

appe

aran

ce

292

dura

tion

159

PE

D5

PE

D

PE

D

Spi

kes

durin

g lo

ng s

uppr

.

1 sp

onta

neou

s hy

perv

entil

atio

n te

nden

cy, 2

mov

emen

t, 3 m

ultis

pike

s an

d –w

aves

, 4 si

ngle

spi

kes,

5 pe

riodi

c ep

ilept

iform

dis

char

ges

Page 70: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

68

5.3 Focal seizure, fMRI and EEG (II)

Penicillin, administered in the cortex while the EEG was in continuous

suppression during isoflurane anaesthesia with ETisoflurane 1.4–1.7%, induced focal

epileptic activity. This was associated with focal EEG discharge and a fMRI

signal increase in the swine brain. The most important result was that the initial

continuous blood-oxygen-level-dependent (BOLD) signal increased from 9.4% to

21.3%, which was clear prior to the appearance of spike activity in the scalp EEG.

This appeared 49 s–2 min 40 s after the penicillin injection (Fig. 8).

Fig. 8. Comparison of fMRI and EEG signals. A. Average signal changes of BOLD

activation map and background gray matter (GM) region of interest (ROI). B. Ipsilateral

EEG signal, 1 500 ms window plotted and the respective EEG total power C.

Dotted line represents the interrupted continuity (approximately 20 s pause

between two scans). Arrowheads point to the time points of ipsilateral spikes (at

1 min), biphasic spikes (at 3 min), double spikes (at 8 min) to high amplitude

monophasic spikes at 13 min).

Page 71: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

69

5.4 Depth electrode recordings in propofol anaesthesia (III)

Propofol burst suppression characteristics were studied by describing electrical

fields of three EEG patterns, the sharp wave, the burst and the spindle (Fig. 9 and

Fig. 10) Simultaneous recording of the EEG from both depth and scalp electrodes

showed that bursts and their slow wave oscillations were synchronous throughout

the entire cortex, as well as in the basal regions of the brain, while spindles and

sharp waves were produced by the sensorimotor cortex.

Sharp waves maximal up to 170 μV with the depth electrode reference at

vertex were negative. Bursts were positive, resembling the delta waves of non-

REM sleep. Burst-suppression could also be recorded between two contacts in the

depth electrode, although with a much lower amplitude than between two widely

spaced scalp electrodes, while the highest amplitude was recorded between the

scalp and depth electrode.

Fig. 9. The burst consists of slow waves with mixed-frequency fast activity and a

rhythmic β spindle on it. The spindle is enlarged on the right. Cz = central,

Fp1 = frontopolar, C7 = skin electrode on the spine of the cervical vertebra C7,

D1 = depth electrode of the subthalamic nucleus.

Page 72: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

70

Fig. 10. Sharp wave and bursts. The sharp wave is enlarged on the right. Cz = central,

Fp2 = frontopolar, A2 = ear electrode, C7 = skin electrode on spine of cervical vertebra

C7. D1 = depth electrode reference. D1-D2 is recorded between the electrode at tip and

the most proximal of the four electrode points of the deep electrode.

Page 73: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

71

6 Discussion

6.1 Methodological aspects

The EEG in studies IV and V were recorded using extensive 32 channel

recordings according to the full 10–20 system used in the EEG laboratory,

including orbitofrontal and ear electrodes and the topography of the EEG could

therefore be examined more extensively than in earlier published examinations.

For example, Yli-Hankala et al. (1999) used four channels (four bipolar electrode

pairs: Fp1-left mastoid, Fp2-right mastoid, Fpz-F7, and Fpz-F8) with adult

patients randomly allocated to spontaneous breathing or controlled

hyperventilation groups in their sevoflurane anaesthesia induction study. When

studying EEG phenomena in sevoflurane anaesthesia induction with

normoventilated children, Vakkuri et al. (2001) also used a four-channel EEG

registration (four bipolar electrode pairs (Fp1-left mastoid, Fp2-right mastoid,

Fpz-left temporal, Fpz-right temporal). Constant (1999) used a 16-channel EEG

registration in sevoflurane and halothane anaesthesia induction study with

children. The EEG was analysed in the frequency domain. Spectral analysis of the

EEG signals was performed using fast Fourier transformation (FFT). Julliac et al. (2007) studied EEG alterations during sevoflurane anaesthesia induction with

adults using four channels (positions Fp1, Fp2, T3, T4, C3, C4, O1 and O2).

6.2 SSEP in sevoflurane anaesthesia (I)

As anaesthesia deepens and activation of the thalamic nuclei decreases,

information is still relayed forward as the cortex remains active for an extended

period. This was demonstrated in study I, in which somatosensory information

was received in the cerebral cortex while under deep sevoflurane anaesthesia with

burst-suppression patterns observed in the EEG, similar to the manner in which it

is in subjects who are awake. Our observation is in agreement with findings

presented in the study by Hartikainen et al. (1995), who reported that all auditory,

visual and somatosensory (median nerve) stimulation modalities readily evoked

bursts, decreased the duration of suppression and increased the total duration of

bursts during steady-state B-S anaesthesia at a 1.5% end-tidal isoflurane

concentration (ETiso). Under deeper anaesthesia (at 1.8 ETiso concentrations) the

Page 74: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

72

stimuli created less of a reaction and there was a predominance of stimulus offset-

evoked bursts over onset bursts (Hartikainen et al. 1995).

It has been widely debated whether the thalamus blocks the flow of

information during anaesthesia. It would appear, however, that it does not block

that flow, but rather that the subsequent processing of information is prevented.

Hudetz & Imas (2007) compared the spatiotemporal properties and frequency

spectra of discrete flash stimuli-induced cortical field potential responses with

spontaneous bursts during isoflurane 1.4–1.8% B-S anaesthesia in laboratory

animals. Each flash produced a visual evoked potential in the primary visual

cortex followed by secondary bursting activity in the more anterior regions while

spontaneous and flash-induced bursts did not differ in frequency, duration or

spatial distribution, with maximal power in the frontal (primary motor) cortex.

Electrocortical suppression of the cortex during anaesthesia did not prevent a

response to visual stimuli. Anaesthetic unconsciousness may not be due to

cortical deafferentation, but may arise from an inability of the brain to integrate or

perceive, rather than receive, sensory information (Hudetz & Imas 2007).

Vandesteen et al. (1991) evaluated the effects of isoflurane, enflurane and

halothane anaesthesia on SSEP by stimulation of the median nerve. Of the median

nerve SSEP components, the pre-central P22 was enhanced while the parietal N20

potential did not increase. In the present study I, however, N20 increased.

Halothane does not induce B-S, nor does it increase P22 (Vandesteene et al. 1991). One reason for the enhancement of the early components of the median

nerve-evoked potential P22 and N20/P20 during B-S anaesthesia under

sevoflurane, isoflurane and enflurane may be that by reflecting an increased

tendency to suppression of the inhibitory activity these anaesthetics permit

thalamocortical excitatory activity.

The stimulation frequency is of significance in the registration of SSEP under

deep inhalation anaesthesia. We could regularly record a high amplitude N20/P22

wave by stimulating the median nerve under sevoflurane anaesthesia at B-S level

and a reduction in stimulus frequency from 5 Hz to 1 Hz resulted in a marked

increase in the N20 amplitude without averaging in single sweeps. Porkkala

(1994) used a stimulation frequency from 4 to 5 Hz on the median nerve during

isoflurane anaesthesia and found a reduction in N20 amplitude and an increase in

latency at isoflurane concentrations over 1 MAC. In some cases the N20 response

was almost entirely abolished during suppression of the EEG.

Cortical tibial nerve SSEP with a high amplitude (up to 7.2 µV) was

registered under deep sevoflurane suppression anaesthesia and was usually visible

Page 75: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

73

without averaging when the stimulation frequency was less than one per second

(Rytky et al. 1999). Stimulation frequencies higher than 1 Hz produced a

significant decrease in amplitude, which is in agreement with study I.

Westeren-Punnonen et al. (2008) used a sevoflurane end-tidal concentration

of 2% (corresponds to 1 MAC) for the maintenance of anaesthsia after induction

with thiopental while studying median nerve SSEP responses in children aged

three to eight years. The study group used a stimulation frequency of 5 Hz and

found that the somatosensory cortical N20 component latency was prolonged by

25% as a result of central conduction time (interpeak latency N13-N20)

prolongation, while no or minimal changes were recorded in the peripheral or

cervical SEP latencies. The mean cortical N20-P22 amplitude was decreased by

32%.

6.3 Epileptiform EEG in sevoflurane anaesthesia (IV, V)

High concentrations of sevoflurane (8%) in 50% nitrous oxide and oxygen were

used for anaesthesia induction in healthy children with spontaneous and

subsequently controlled normoventilation during the deepening of anaesthesia

(study IV). The study confirmed epileptiform EEG patterns, which began as early

as one to four minutes after the start of anaesthesia induction, with multifocal

single spikes and polyspikes with frontal maxima. Later on, PEDs developing

from polyspikes and waves in some subjects were recorded synchronously over

the entire cortex. Gibert et al. (2012) found major epileptiform signs in the EEG

in 50% of healthy normoventilated children receiving 1.7 MAC sevoflurane in

100% oxygen at steady-state maintenance anaesthesia. Nitrous oxide 50% in

oxygen or alfentanil raised the threshold for epileptiform EEG transients.

No focal epileptiform EEG seizures were observed in studies IV and V. Focal

epileptiform EEG seizures have not been detected in other anaesthesia induction

studies with high concentrations of sevoflurane, using either hyper- or

normoventilation (Yli-Hankala et al. 1999, Vakkuri 2000 et al, Vakkuri 2001 et al., Constant et al. 1999). In an earlier study with enflurane, EEG bursts gradually

transformed into an epileptic discharge in children (Rosen & Söderberg 1975) and

in adults (Neigh et al. 1971, Lebowitz et al. 1972, Yli-Hankala & Jäntti 1990a)

being hyperventilated. Enflurane and sevoflurane seem to share, at least, some

common mechanisms in producing epileptiform discharges in the EEG during

deep anaesthesia.

Page 76: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

74

In contrast to the above, focal epileptiform EEG transitions with motoric

seizures were reported at high concentrations of sevoflurane maintenance

anaesthesia in healthy young adults (Kaisti et al. 1999, Jääskeläinen et al. 2003).

Seizure-like activity was observed in all patients under steady-state anaesthesia

with a maintenance sevoflurane concentration over 1.5 MAC (Sato et al. 2002,

Jääskeläinen et al. 2003). Nieminen et al. (2002) used intravenous anaesthesia

induction and sevoflurane maintenance with an end-tidal concentration of 2% in

40% oxygen in air for healthy children aged three to eight years and EEG

registration was carried out with a five-channel measure according to the 10–20

system. They did not detect any epileptiform EEG alterations with this

sevoflurane maintenance concentration.

Epileptiform EEG patterns related to the use of sevoflurane are observed

during the rapid increase in sevoflurane concentrations at anaesthesia induction

(Yli-Hankala et al. 1999, Vakkuri et al. 2000, Vakkuri et al. 2001, Julliac et al. 2007, Schultz et al. 2012, Julliac et al. 2013, Kreuzer et al. 2014), during long-

lasting maintenance of anaesthesia with high sevoflurane concentrations (Kaisti et al. 1999, Jääskeläinen et al. 2003), and when sevoflurane is used in mentally

disabled patients with lowered seizure thresholds, as well as in epileptic patients

(Iijima et al. 2000).

The heart rate rise observed in study IV appeared before epileptogenic spikes

were seen anywhere in the cortex, suggesting a complex relationship with

epileptiform EEG. The polyspike and -wave activity yields erroneous BIS values,

as the depth-of-anaesthesia cannot be defined during seizure activity.

Risk factors for epileptiform discharges among healthy adult patients during

anaesthesia induction with sevoflurane have been found to be the speed of

anaesthesia induction, a high alveolar sevoflurane concentration, and female sex

(Julliac et al. 2007). Rapid sevoflurane induction may induce a too rapid sleep

initiation and thus trigger epileptiform discharges, as epileptiform phenomena

corresponding to pathologic cortical EEG hypersynchronization and EEG

abnormalities can be more frequent during sleep initiation (Julliac et al. 2007).

Even after decreasing the speed of the increase in the sevoflurane CNS

concentration by delaying the onset of hyperventilation for two minutes at high

sevoflurane concentrations during anaesthesia induction in healthy women, severe

epileptiform EEGs with a hyperdynamic response were still observed (Vakkuri et al. 2000). PED, however, tended to occur more often with an immediate onset of

hyperventilation (Vakkuri et al. 2000). In addition, anaesthesia induction with a

Page 77: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

75

low target for sevoflurane end-tidal concentration of 2.5% failed to totally prevent

epileptiform EEGs in young healthy female patients (Julliac et al. 2013)

In study V, the controlled hyperventilated (CH) male adults had more

epileptiform EEG alterations (90%) than did spontaneously breathing (SB) ones

(40%). The deviations of heart rate values in CH patients were significant in study

V, but the mean HR was higher than in the SB group. Mean HR increased during

the first two minutes in both groups and thereafter decreased in SB patients. Heart

rate rose abruptly at burst onset and dropped at suppression onset during deep

enflurane or isoflurane anaesthesia (Yli-Hankala et al. 1990a, Yli-Hankala &

Jäntti 1990b, Jäntti & Yli-Hankala 1990). An increase in heart rate up to 54% as

well as a smaller increase in blood pressure was detected by Yli-Hankala et al. (1999) in hyperventilated patients, but not in spontaneously breathing patients

during high concentration sevoflurane anaesthesia induction, and a 30% increase

in heart rate was associated with epileptiform EEGs, while no such connection

was seen between EEG and mean arterial pressure (MAP). The higher level of

mean HR in study V resembles the observation by Yli-Hankala et al. (1999), but

the lack of statistical significance may be due to the smaller number of patients

(n = 10 compared to n = 15). In study V, spontaneous breathing, allowing relative

carbon dioxide retention, maintained both EEG and HR alterations to a lesser

degree than by using controlled normoventilation (Vakkuri et al. 2001).

Premedication with a benzodiazepine - midazolam in study IV and diazepam

in study V - was used, both of which increase the threshold for convulsion (Rivera

et al. 1993, Scott et al. 1998). Nitrous oxide was used in 50% oxygen during

anaesthesia induction for both children and adults (studies IV and V). Nitrous

oxide reduces the epileptogenic effects in the EEG. Kurita et al. (2005) used an

electrocorticogram (ECoG) in epileptic patients below 1.5 MAC of sevoflurane

anaesthesia concentration with or without 50% nitrous oxide and found that the

numbers of spikes with nitrous oxide were significantly lower than those without

nitrous oxide, although the areas displaying spike activity did not differ.

Regardless of the use of both midazolam premedication and nitrous oxide,

however, the first spikes in the EEG were observed as early as from one to four

minutes from the beginning of anaesthesia induction in normoventilated children

in study IV.

In study IV, the location of spikes and polyspikes were concentrated mainly

frontally and were sometimes limited entirely to the frontocentral region. They

might therefore not be recognized in the fronto-temporal recordings used by

depth-of-anaesthesia monitors (Vakkuri et al. 2004).

Page 78: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

76

The BIS calculated offline from the EEG recordings of patients in study IV

increased due to epileptogenic EEG activity (Jäntti et al. 2013). The BIS first

dropped under 10 in 11 children during high amplitude monophasic delta activity,

the average being 13.7, which ordinarily suggests a B-S pattern. After a slow delta

pattern, the EEG changed to epileptiform activity in all patients and the BIS

jumped to 95 in two patients and over 60 in two others. Sandin et al. (2008) also

demonstrated a similar pattern of BIS increase at a 2 MAC sevoflurane

concentration in healthy young adults and regarded it as being associated with the

epileptogenic activity of sevoflurane. One of their patients suffered a concomitant

short convulsive episode.

6.4 Focal seizure, fMRI and EEG (II)

The basic principle of the BOLD signal depends on the association of neuronal

activation with an increase in regional cerebral blood flow and a proportional

reduction in oxygen extraction resulting in a regional decrease of

deoxyhemoglobin. This change of regional deoxyhemoglobin concentration can

be detected by fMRI, thus representing an indirect, secondary measure of

neuronal activation (Dueck et al. 2005).

A remarkable BOLD increase prior to spike activity in the EEG was found in

the experimental penicillin-induced epileptic focus study in pigs anaesthetized

with isoflurane to a deep burst-suppression level of anaesthesia (study II). Thus,

this remarkable cerebral blood flow elevation was related to epileptiform activity

at an EEG burst suppression level (study II). Burst-suppression did not protect

from focal discharge. Hawko et al. (2007) examined epileptic patients in their

EEG-fMRI study and also detected a BOLD response occurring several seconds

prior to interictal scalp EEG discharges. This finding may imply that some

epileptic discharges are preceded by an energy-consuming phenomenon which

lasts several seconds and may not consist of a synchronized neuronal discharge

(Hawko et al. 2007). A study by Jacobs et al. (2009) also confirmed the

occurrence of BOLD changes prior to spikes in patients with idiopathic or

symptomatic focal epilepsy. Early BOLD changes were strongly related to the

spike field and were more local in comparison with later spike-related BOLD

changes, thus resulting in a better localizing value. Pittau et al. (2011) came to the

same conclusion in their study of focal epilepsy patients in which they also used

depth electrodes. They speculated that, in some cases, the early BOLD activity

was explained by a synchronized neural discharge which was detectable with

Page 79: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

77

stereo-EEG but not visible in the scalp EEG, while in other cases the early BOLD

response possibly reflected a metabolic phenomenon not resulting from a

synchronized neuronal discharge, but rather from non-neuronal mechanisms

including the glia. In another study (Moeller et al. 2008), patients with

generalized epilepsy displayed a BOLD increase several seconds prior to

generalized spike wave discharges, with the earliest haemodynamic changes in

the medial thalamus. Faizo et al. (2014) studied functional connectivity changes

in a neural network prior to interictal spikes with simultaneous EEG and fMRI in

patients with mesial temporal lobe focal epilepsy. They found a significant loss of

connectivity between the hippocampi several seconds before the appearance of

spikes on EEG signifying a decoupling of hippocampal inhibition which may

predispose to spike generation.

Isoflurane exerts a more potent anticonvulsant effect as compared to

sevoflurane and halothane (Murao et al. 2000), so the focal penicillin-induced

findings were not manipulated by anaesthetic agent-induced epileptogenic

findings. Alternatively, isoflurane at an anaesthesia level close to a B-S pattern

did not suppress epileptic spikes in humans (Fiol et al. 1993). Isoflurane

anaesthesia also enabled the localization of the primary epileptogenic focus by

limiting the spread of epileptic activity.

6.5 Depth electrode recordings in propofol anaesthesia (III)

Study III elucidated the biophysics of the EEG. In that study, it was demonstrated

that EEG bursts are positive waves like the delta waves of deep, non-REM sleep.

The vertex-wave was negative. The slow waves of bursts were synchronous over

the entire cortex, even in the basal regions.

The reticular nucleus of the thalamus is a small structure and unlikely to

produce an EEG field visible at the scalp. It has been deduced through

thalamocortical projections that it probably drives cortical neurons, which then

produce the EEG spindles recorded off the scalp (Sinha 2011). The results of

study III show that spindles recorded from the depth electrodes are very low in

amplitude but high in the transcortical recording, i.e. between the scalp and the

depth electrode. This means that the scalp-recorded signal must be generated by

the cerebral cortex and that the reticular nucleus is only a pacemaker, contrary to

what has been claimed by Velly et al. (2007).

Page 80: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

78

Touching, skin pinching or sound stimuli did not evoke bursts in propofol

burst-suppression anaesthesia. This was in agreement with the results presented in

the study by Huotari et al. (2004).

Due to synchronous slow activity over the entire cortex (including the basal

areas) resulting in a partly closed field, the EEG patterns and their generators

cannot be described precisely with scalp-recordings alone. The amplitude of EEG

patterns can only be measured using both surface and depth electrodes. In study

III, the recordings were made from scalp and neck electrodes referred to the depth

electrode at the subthalamic nucleus.

Wennberg & Lozano (2003) showed that the slow waves in the K-complex

during physiological NREM sleep were in the same phase in all surface electrodes

when referred to the depth electrode in a protocol similar to that used in the

present study III during propofol anaesthesia at burst-suppression level. The slow

waves of the K-complex were also more than twice the amplitude in any of the

scalp-depth recordings compared with the maximal amplitude of scalp-recording,

as with the burst slow waves in study III.

Page 81: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

79

7 Conclusions

1. The amplitude of the short latency N20 wave of the median nerve SSEP

enhanced and was detected without averaging during sevoflurane-induced

EEG suppression. The latter components adapted and disappeared. This

showed that the somatosensory information reached the cortex and was not

totally blocked by the thalamus. Further processing of these impulses in the

cortex was suppressed.

2. Locally administered penicillin induced typical focal seizures during EEG

suppression in an experimental focal epilepsy model in pigs and provided

additional insight into the localization of epileptic foci with fMRI. The

development of focal epileptic activity could be detected as a BOLD signal

increase, which preceded the scalp EEG spike activity in deep isoflurane

anaesthesia at continuous EEG suppression.

3. Bursts and their slow wave oscillations were observed synchronously over

the entire cortex during propofol anaesthesia at EEG burst-suppression level

in Parkinson`s patients, while spindles and sharp waves were produced by the

sensorimotor cortex. The slow waves were surface positive and the vertex

wave was negative. Cortically generated burst-suppression could also be

recorded using depth electrodes.

4. Spike- and polyspike waveforms during 8% sevoflurane anaesthesia mask

induction in children were concentrated in a multifocal manner fronto-

centrally. These epileptiform waveforms may be underestimated when using

the typical processed depth-of-anaesthesia monitoring devices.

5. High sevoflurane concentrations during anaesthesia mask induction caused

epileptiform EEG activity concomitant with dynamic cardiovascular

phenomena in controlled hyperventilated adult male patients. Both

spontaneously breathing and controlled normoventilated subjects displayed

these alterations.

Page 82: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

80

Page 83: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

81

References

Achermann P & Borbely AA (1997) Low-frequency (<1 Hz) oscillations in the human sleep electroencephalogram. Neuroscience 81: 213–22.

Adachi M, Ikemoto Y, Kubo K, Takuma C (1992) Seizure-like movements during induction of anaesthesia with sevoflurane. Br J Anaesth 68: 214–5.

Adembri C, Venturi L, Tani A, Chiarugi A, Gramigni E, Cozzi A, Pancani T, De Gaudio RA & Pellegrini-Giampietro DE (2006) Neuroprotective effects of propofol in models of cerebral ischemia. Anesthesiology 104: 80–9.

Aho AJ, Lyytikäinen L-P, Yli-Hankala A, Kamata K, Jäntti V (2011) Explaining entropy responses after a noxious stimulus, with or without neuromuscular blocking agents, by means of the raw electroencephalographic and electromyographic characteristics. Br J Anaesth 106: 69–76.

Aicardi J, Lefebvre J, Lerique-Koechlin AA (1965) New syndrome: spasms in flexion, callosal agenesis, ocular abnormalities. Electroencephalogr Clin Neurophysiol 19: 609–10.

Aime` I, Verroust N, Masson-Lefoll C, Taylor G, Laloe P-A, Liu N & Fischler M (2006) Does monitoring of bispectral index or spectral entropy reduce sevoflurane use? Anesth Analg 103: 1469–77.

Alkire MT, Haier RJ, Barker SJ, Shah NK, Wu JC & Kao YJ (1995) Cerebral metabolism during propofol anesthesia in humans studied with positron emission tomography. 82: 393–403.

Alkire MT, Hudetz AG & Tononi G (2008) Consciousness and anesthesia. Science 322: 876–80.

Amzica F & Steriade M (1997) The K-complex: its slow (< 1-Hz) rhythmicity and relation to delta waves. Neurology 49: 952–9.

Anier A, Lipping T, Ferenets R, Puumala P, Sonkajärvi E, Rätsep I & Jäntti V (2012) Relationship between approximate entropy and visual inspection of irregularity in the EEG signal, a comparison with spectral entropy. Br J Anaesth 109: 928–34.

Anonymous (1993) Guidelines for epidemiologic studies on epilepsy. Commission on epidemiology and prognosis of international league against epilepsy. Epilepsia 34: 592–6.

Antognini JF, Buonocore MH, Disbrow EA & Carstens E (1997) Isoflurane anesthesia blunts cerebral responses to noxious and innocuous stimuli: a fMRI study. Life Sciences 61: pp. PL 349–54.

Arkawi WP, Drummond JC, Kalkman CJ & Patel PM (1996) A comparison of electrophysiologic characteristics of EEG burst-suppression as produced by isoflurane, thiopental, etomidate, and propofol. J Neurosurg Anesthesiol 8: 40–6.

Aronstam RS, Martin DC & Dennison RL (1994) Volatile anesthetics inhibit NMDA-stimulated 45Ca uptake by rat brain microvesicles. Neurochem Res 19: 1515–1520

Artru AA (1994) Intracranial volume/pressure relationship during desflurane anesthesia in dogs: comparison with isoflurane and thiopental/halothane. Anesth Analg 79: 751–60.

Baines D (1998) Convulsive movements with sevoflurane. Anaesth Int Care 26: 329.

Page 84: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

82

Banks MI & Pearce RA (1999) Dual actions of volatile anesthetics on GABAA IPSCs. Dissociation of blocking and prolonging effects. Anesthesiology 90: 120–34.

Banoub M, Tetzlaff JE & Schubert A (2003) Pharmacologic and physiologic influences affecting sensory evoked potentials. Implications for perioperative monitoring. Anesthesiology 2003; 99: 716–37.

Bennett C, Voss LJ, Barnard JPM & Sleigh JW (2009) Practical use of the raw electroencephalogram waveform during general anesthesia: the art of science. Anesth Analg 109: 539–50.

Berger H (1929) Uber das elektroenkephalogram des menschen. Arch Psychiatr Nervenkr 87: 527–70.

Berg-Johansen J & Langmoen IA (1992) The effect of isoflurane on excitatory synaptic transmission in the rat hippocampus. Acta Anaesthesiol Scand 36: 350–5.

Berkhoff M, Donati F & Bassetti C (2000) Postanoxic alpha (theta) coma: a reappraisal of its prognostic significance. Clin Neurophysiol 111: 297–304.

Beydoun A, Yen CE & Drury I (1991) Variance of interburst intervals in burst suppression. Electroencephalogr Clin Neurophysiol 79: 435–9.

Binnie CD & Prior PF (1994) Electroencephalography. J Neurol Neurosurg Psychiatry 57: 1308–19.

Binnie CD & Stefan H (1999) Modern electroencephalography: its role in epilepsy management. Clin Neurophysiol 110: 1671–97.

Borgeat A, Dessibourg C, Popovic V, Meier D, Blanchard M & Schwander D (1991) Propofol and spontaneous movements: an EEG study. Anesthesiology 74: 24–7.

Brown EN, Lydic R & Schiff ND (2010) General anesthesia, sleep and coma. N Engl J Med 363: 2638–50.

Bruhn J, Bouillon TW & Shafer SL (2000) Bispectral index (BIS) and burst suppression: revealing a part of the BIS algorithm. J Clin Monit Comput 16: 593–6.

Burchiel KJ, Stockard JJ, Roderick KC & Smith NT (1977) Relationship of pre- and postanesthetic EEG abnormalities to enflurane-induced seizure activity. Anesth Analg 56: 509–14.

Bösenberg AT (1997) Convulsions and sevoflurane. Paediatr Anaesth 7: 476–8. Campagna JA, Miller KW & Forman SA (2003) Mechanisms of actions of inhaled

anesthetics. N Engl J Med 348: 2110–24. Chang BS, Schomer DL & Niedermeyer E (2011) Epilepsy in adults and the elderly. In

Schomer DL & Lopes da Silva FH (eds) Niedermayer`s electroencephalography. Basic principles, clinical applications, and related fields. 6th edition. Lippincott Williams & Wilkins, Philadelphia. 541–62.

Chatrian GE, Shaw CM & Leffman H (1964) The significance of periodic epileptiform discharges in EEG: an electrographic, clinical and pathological study. Electroencephalogr Clin Neurophysiol 17: 177–93.

Ching S, Purdon PL, Vijayan S, Kopell NJ & Brown EN (2011) A neurophysiological-metabolic model for burst suppression. PNAS 109: 3095–100.

Page 85: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

83

Chinzei M, Sawamura S, Hayashida M et al (2004) Change in bispectral index during epileptiform electrical activity under sevoflurane anesthesia in a patient with epilepsy. Anesth Analg 98: 1734–6.

Cibula JE & Gilmore RL (1997) Secondary epileptogenesis in humans. J Clin Neurophysiol 14: 111–27.

Clark DL & Rosner BS (1973) Neurophysiologic effects of general anesthetics: I. The electroencephalogram end sensory evoked responses in man. Anesthesiology 38: 564–82.

Conreux F, Best O, Preckel MP et al (2001) Electroencephalographic effects of sevoflurane in pediatric anesthesia: a prospective study of 20 cases. Ann Fr Anesth Reanim 20: 438–45.

Constant I, Dubois M-C, Piat V, Moutard M-L, McCue M & Murat I (1999) Changes in electroencephalogram and autonomic cardiovascular activity during induction of anesthesia with sevoflurane compared with halothane in children. Anesthesiology 91: 1604–15.

Constant I, Seeman R & Murat I (2005) Sevoflurane and epileptiform EEG changes. Paediatr Anaesth 15: 266–74.

Contreras & Steriade (1995) Cellular basis of EEG slow rhythms: a study of dynamic corticothalamic relationships. J Neuroscience 15: 604–22.

Contreras D, Destexhe A, Sejnowski TJ & Steriade M (1997) Spatiotemporal patterns of spindle oscillations in cortex and thalamus. J Neuroscience 17: 1179–96.

Crosby G & Culley DJ (2012) Processed electroencephalogram and depth of anesthesia. Window to nowhere or into the brain? Editorial Anesthesiology 116:235–7.

Derbyshire AJ, Rempel B, Forbes A & Lambert EF (1936) The effects of anesthetics on action potentials in the cerebral cortex of the cat. Am J Physiol 116: 577–96.

Drouot X, Cabello B, d`Ortho & Brochard L (2008) Sleep in the intensive care unit. Sleep Med Rev 12: 391–403.

Drummond JC (2000) Monitoring depth of anesthesia - with emphasis on the application of the bispectral index and the middle latency auditory evoked response to the prevention of recall. Anesthesiology 93: 876–882.

Dueck MH, Petzke F, Gerbershagen HJ, Paul M, Hesselman V, Girnus R, Krug B, Goebel R, Lehrke R, Sturm V & Boerner U (2005) Propofol attenuates responses of the auditory cortex to acoustic stimulation in a dose-dependent manner: a fMRI study. Acta Anaesthesiol Scand 49: 784–91.

Eger EI II, Stevens WC & Cromwell TH (1971) The electroencephalogram in man anesthetized with forane. Anesthesiology 35: 504–8.

Eger EI (1994) New inhaled anesthetics. Anesthesiology 80: 906–22. Ellerkmann RK, Liermann V-M, Alves TM, Wenningmann I, Kreuer S, Wilhelm W,

Roepcke H & Hoeft A (2004) Spectral entropy and bispectral index as measures of the electroencephalographic effects of sevoflurane. Anesthesiology 101: 1275–82.

Endo T, Sato K, Shamoto H & Yoshimoto T (2002) Effects of sevoflurane on electrocorticography in patients with intractable temporal lobe epilepsy. J Neurosurg Anesthesiol 14: 59–62.

Page 86: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

84

Fahlenkamp AV, Peters D, Biener IA, Billoet C, Apfel CC, Rossaint R & Coburn M (2010) Evaluation of bispectral index and auditory evoked potentials for hypnotic depth monitoring during balanced xenon anaesthesia compared with sevoflurane. Br J Anaesth 105: 334–41.

Faizo NL, Burianova H, Gray M, Hocking J, Galloway G & Reutens D (2014) Identification of pre-spike network in patients with mesial temporal lobe epilepsy. Front Neurol 5. doi: 103389/fneur 2014.00222.

Finelli LA, Baumann H, Borbely AA & Achermann P (2000) Dual electroencephalogram markers of human sleep homeostasis: correlation between theta activity in waking and slow-wave activity in sleep. Neuroscience 101: 523–9.

Fiol ME, Boening JA, Cruz-Roriguez R & Maxwell R (1993) Effect of isoflurane (Forane) on intraoperative electrocorticogram. Epilepsia 34: 897–900.

Fraga M, Rama-Maceiras P, Rodino S, Aymerich H, Pose P & Belda J (2003) The effects of isoflurane and desflurane on intracranial pressure, cerebral perfusion pressure and cerebral arteriovenous oxygen content difference in normocapnic patients with supratentorial brain tumors. Anesthesiology 98: 1085–90.

Franks NP (2008) General anaesthesia: from molecular targets to neuronal pathways of sleep and arousal. Nature Reviews Neuroscience 9: 370–86.

Fukui K, Morioka T, Hashiguchi K, Kawamura T, Irita K, Hoka S, Sasaki T & Takahashi S (2010) Relationship between regional cerebral blood flow and electrocorticographic activities under sevoflurane and isoflurane anesthesia. J Clin Neurophysiol 27: 110–115.

Gibert S, Sabourdin N, Louvet N, Moutard M-L, Piat V, Guye M-L, Rigouzzo A & Constant I (2012) Epileptogenic effect of sevoflurane. Determination of the minimal alveolar concentration of sevoflurane associated with major epileptoid signs in children. Anesthesiology 117: 1253–61.

Goa KL, Noble S & Spencer CM (1999) Sevoflurane in paediatric anaesthesia. A review. Paediatr Drugs 1: 127–53.

Goddard GV (1967) Development of epileptic seizures through brain stimulation at low intensity. Nature 214: 1020–1.

Goddard GV, McIntyre DC & Leech CK (1969) A permanent change in brain function resulting from daily electrical stimulation. Experimental Neurology 25: 295–330.

Haga S, Shima T, Momose K et al (1992) Anesthetic induction of children with high concentrations of sevoflurane. Masui 41: 1951–5.

Hall AC, Lieb WR, Franks NP. Stereoselective and non-stereoselective actions of isoflurane on the GABAA receptor. Br J Pharmacol 1994; 112: 906.

Hans P, Dewandre P-Y, Brichant JF & Bonhomme V (2005) Comparative effects of ketamine on Bispectral Index and spectral entropy of the electroencephalogram under sevoflurane anaesthesia. Br J Anaesth 94: 336–40.

Hartikainen K, Rorarius M, Mäkelä K, Peräkylä J, Varila E & Jäntti V (1995) Visually evoked bursts during isoflurane burst suppression anaesthesia. Br J Anaest 74: 681–5.

Page 87: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

85

Hartikainen K, Rorarius M, Mäkelä K, Yli-Hankala A & Jäntti V (1995) Propofol and isoflurane induced EEG burst suppression patterns in rabbits. Acta Anaesthesiol Scand 39: 814–8.

Hartikainen KM, Rorarius M, Peräkylä JJ, Laippala PJ & Jäntti V (1995) Cortical reactivity during isoflurane burst-suppression anesthesia. Anesth Analg 81: 1223–8.

Hartikainen KM, Rorarius M & Jäntti V (1996) Monitoring the integrity of somatosensory pathways with evoked electroencephalographic bursts. Anesth Analg 83: 454–8.

Hartikainen K & Rorarius MG (1999) Cortical responses to auditory stimuli during isoflurane burst suppression anaesthesia. Anaesthesia 54: 210–4.

Hawko CS, Bagshaw AP, Lu Y, Dubeau F & Gotman J (2007) Bold changes occur prior to epileptic spikes seen on scalp EEG. Neuroimage 35: 1450–8.

Henry CE & Scoville WB (1952) Suppression-burst activity from isolated cerebral cortex in man. Electroenceph Clin Neurophysiol 4:1–22.

Herman ST, Walczak TS & Bazil CW (2001) Distribution of partial seizures during the sleep-wake cycle. Differences by seizure onset site. Neurology 56: 1453–9.

Hilty CA & Drummond JC (2000) Seizure-like activity on emergence from sevoflurane anesthesia. Anesthesiology 93: 1357–9.

Hisada K, Morioka T, Fukui K, Nishio S, Kuruma T, Irita K, Takahashi S & Fukui M (2001) Effects of sevoflurane and isoflurane on electrocorticographic activities in patients with temporal lobe epilepsy. J Neurosurg Anesth 13: 333–337.

Hobson JA & Pace-Schott EF (2002) The cognitive neuroscience of sleep: neuronal systems, consciousness and learning. Nature Reviews/Neuroscience 3: 679–93.

Hofmeijer J, Tjepkema-Cloostermans MC and Putten MJAM (2014) Burst-suppression with identical bursts: a distinct EEG pattern with poor outcome in postanoxic coma. Clin Neurophysiol 125: 947–54.

Hosain S, Nagarajan L, Fraser R, Van Poznak A & Labar D (1996) Electroencephalogr Clin Neurophysiol 102: 340–2.

Hsieh S-W, Lan K-M, Luk H-N & Jawan B (2004) Postoperative seizures after sevoflurane anaesthesia in a neonate. Acta Anaesthesiol Scand 48: 663.

Hudetz AG & Imas OA (2007) Burst activation of the cerebral cortex by flash stimuli during isoflurane anesthesia in rats. Anesthesiology 107: 983–91.

Huotari A-M, Koskinen M, Suominen K, Alahuhta S, Remes R, Hartikainen KM & Jäntti V (2004) Evoked EEG patterns during burst suppression with propofol. Br J Anaesth 92: 18–24.

Huupponen E, Maksimow A, Lapinlampi P, Särkelä M, Saastamoinen A, Snapir A, Scheinin H, Scheinin M, Meriläinen P, Himanen S-L & Jääskeläinen S (2008) Electroencephalogram spindle activity during dexmedetomidine sedation and physiologic sleep. Acta Anaesthesiol Scand 52: 289–94.

Iijima T, Nakamura Z, Iwao Y & Sankawa H (2000) The epileptic properties of the volatile anesthetics sevoflurane and isoflurane in patients with epilepsy. Anesth Analg 91: 989–95.

Page 88: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

86

Illievich UM, Petricek W, Schramm W, Weindlmayr-Goettel M, Czech T & Spiss C (1993) Electroencephalographic burst suppression by propofol infusion in humans: hemodynamic consequences. Anesth Analg 77: 155–60.

Ito BM, Sato S, Kufta CV & Tran D (1988) Effect of isoflurane and enflurane on the electrocorticogram of epileptic patients. Neurology 38: 924–8.

Jääskeläinen SK, Kaisti K, Suni L, Hinkka S & Scheinin H (2003) Sevoflurane is epileptogenic in healthy subjects at surgical levels of anesthesia. Neurology 61: 1073–8.

Jacobs J, Levan P, Moeller F, Boor R, Stephani U, Gotman J & Siniatchkin M (2009) Hemodynamic changes preceding the interictal EEG spike in patients with focal epilepsy investigated using simultaneous EEG-fMRI. Neuroimage 45: 1220–35.

Jäntti V & Yli-Hankala A (1990) Correlation of instantaneous heart rate and EEG suppression during enflurane anaesthesia: a synchronous inhibition of heart rate and cortical electrical activity? Electroencephalogr Clin Neurophysiol 76: 476–9.

Jäntti V, Yli-Hankala A, Baer GA & Porkkala T (1993) Slow potentials of EEG burst suppression pattern during anaesthesia. Acta Anaesthesiol Scand 37: 121–3.

Jäntti VHK & Sonkajärvi E (2004) Sevoflurane is epileptogenic in healthy subjects at surgical levels of anesthesia. Neurology 62: 2335.

Jäntti V (2005) From crystal ball towards cognitive anaesthesiology. Acta Anaesthesiol Scand 49: 273–6.

Jäntti V (2012) Unconsciousness and EEG burst suppression. Br J Anaesth 108: 334–67P, 8th International symposium of memory and awareness in anesthesia (MAA8), Milwaukee, WI, USA 2–5 June 2011. abnormalities. Ann Fr Anesth Reanim 32: e143–8.

Jäntti V (2013) Do we need more anesthesia EEG indexes? J Clin Monit Comput 27: 105–6.

Jäntti V, Sonkajärvi E, Särkelä M, Zabihi M, Sompa U, Kulkas A & Lipping T (2013) Effect of slow delta EEG on BIS-index in sevoflurane induction with children. Acta Anaesth Scand 57 supplement S120: P3.6: 10, 32nd congress of Scandinavian Society of Anaesthesiologists and Intensive Care Medicine, focusing on the brain, Turku, Finland 26–29 August 2013.

John ER & Prichep LS (2005) The anesthetic cascade. A theory of how anesthesia suppresses consciousness. Anesthesiology 102: 447–71.

Julliac BJ, Guehl D, Chopin F, Arne P, Burbaud P, Sztark F & Cros A-M (2007) Risk factors for occurrence of electroencephalogram abnormalities during induction of anesthesia with sevoflurane in nonepileptic patients. Anesthesiology 106: 243–51.

Julliac B, Cotillon P, Guehl D, Richez B & Sztark F (2013) Target-controlled induction with 2.5% sevoflurane does not avoid the risk of electroencephalographic abnormalities. Ann Fr Anesth Reanim 32: e143-e148.

Kaisti KK, Jääskeläinen SK, Rinne JO, Metsähonkala L & Scheinin H (1999) Epileptiform discharges during 2 MAC sevoflurane anesthesia in two healthy volunteers. Anesthesiology 99: 1952–5.

Page 89: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

87

Kaisti KK, Metsähonkala L, Teräs M, Oikonen V, Aalto S, Jääskeläinen S, Hinkka S & Scheinin H (2002) Effects of surgical levels of propofol and sevoflurane anaesthesia on cerebral blood flow in healthy subjects studied with positron emission tomography. Anesthesiology 96: 1358–70.

Kaplan PW & Bauer G (2011) Anoxia, coma and brain death. In Schomer DL & Lopes da Silva FH (eds) Niedermeyer`s Electroencephalography. Basic principles, clinical applications and related fields. Lippincott Williams & Wilkins, Philadelphia: 435–56.

Kissin I (1993) General anesthetic action: an obsolete notion? Anesth Analg 76: 215–8. Kochs E, Bischoff P, Pichlmeyer U & am Esch JS (1994) surgical stimulation induces

changes in brain electrical activity during isoflurane/nitrous oxide anesthesia: A topographic electroencephalographic analysis. Anesthesiology 80: 1026–34.

Komatsu H, Taie S, Endo S, Fukuda K, Ueki M, Nogaya J & Ogli K (1994) Electrical seizures during sevoflurane anesthesia in two pediatric patients with epilepsy. Anesthesiology 81: 1535–7.

Koyama S, Makino Y, Tanaka K, Morino M, Nishikawa K & Asada A (2002) Fentanyl administration during sevoflurane anesthesia suppresses spike waves from epileptic focus on electrocorticogram. Masui 51: 755–8.

Krasowski MD, Koltchine VV, Rick CE, Qing YE, Finn SE, Harrison NL. Propofol and other intravenous anesthetics have sites of action on the gamma aminobutyric acid type A receptor distinct from that for isoflurane. Mol Phamacol 1998; 53: 530–538.

Kreuzer I, Osthaus WA, Schultz A & Schultz B (2014). Influence of the sevoflurane concentration on the occurrence of epileptiform EEG patterns. PLoS one ; 9: e89191.

Kroeger D & Amzica F (2007) Hypersensitivity of the anesthesia-induced comatose brain. J Neuroscience 27: 10597–607.

Kungys G, Kim JB, Jinks SL, Atherley RJ & Antognini JF (2009) Propofol produces immobility via action in the ventral horn of the spinal cord by a GABAergic mechanism. Anesth Analg 108: 1531–7.

Kurita N, Kavaguchi M, Hoshida T, Nakase H, Sakaki T & Furuya H (2005) Effects of nitrous oxide on spike activity on electrocorticogram under sevoflurane anesthesia in epileptic patients. J Neurosurg Anesthesiol 17: 199–202.

Laitio RM, Kaskinoro K, Särkelä MOK, Kaisti KK, Salmi E, MaksimowA, Långsjö JW, Aantaa R, Kangas K, Jääskeläinen S & Scheinin H (2008) Bispectral index, entropy, and quantitative electroencephalogram during single-agent xenon. Anesthesiology 108: 63–70.

Lam AM, Mayberg TS, Eng CC, Cooper JO, Bachenberg KL & Mathisen TL (1994) Nitrous-oxide-isoflurane anesthesia causes more cerebral vasodilatation than equipotent dose of isoflurane in humans. Anesth Analg 78: 462–8.

Lam AM, Matta BF, Mayberg TS & Strebel S (1995) Change in cerebral blood flow velocity with onset of EEG silence during inhalation anesthesia in humans: evidence of flow-metabolism coupling? J Cereb Blood Flow Metab 15: 714–7.

Långsjö JW, Alkire MT, Kaskinoro K, Hayama H, Maksimow A, Kaisti KK, Aalto S, Aantaa R, Jääskeläinen SK, Revonsuo A & Scheinin H (2012) Returning from oblivion: imaging the neural core of consciousness. J Neuroscience 32(14): 4935–43.

Page 90: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

88

Larsen M, Haugstad TS, Berg-Johnsen J & Langmoen IA (1998) Effect of isoflurane on release and uptake of gamma-aminobutyric acid from rat cortical synaptosomes. Br J Anaesth 80: 634–8.

Laureys S (2005) The neural correlate of (un)awareness: lessons from the vegetative state. TRENDS in Cognitive Sciences 9: 556–9.

Lazar LM, Milrod LM, Solomon GE & Labar DR (1999) Asynchronous pentobarbital-induced burst suppression with corpus callosum hemorrhage. Clin Neurophysiol 110: 1036–40.

Lebowitz MH, Blitt CD & Dillon JB (1972) Enflurane-induced central nervous system excitation and its relation to carbon dioxide tension. Anesth Analg 51: 355–63.

Lenard HG, Yaneza PL & Reimer M (1976) Polygraphic recordings in subacute sclerosing panencephalitis. A study of the pathophysiology of the periodic EEG complexes. Neuropädiatrie 7: 52–65.

Lerlerc C & Vauguet E (2006) Like-seizure movements in a child under sevoflurane anaesthesia. Ann Fr Anesth Reanim 25: 903.

Lerman J, Sikich N, Kleinman S & Yentis S (1994) The pharmacology of sevoflurane in children. Anesthesiology 80: 814–24.

Liley DTJ & Bojak I (2005) Understanding the transition to seizure by modelling the epileptiform activity of general anesthetic agents. J Clin Neurophysiol 22: 300–13.

Lipping T, Jäntti V, Yli-Hankala A & Hartikainen K (1995) Adaptive segmentation of burst-suppression pattern in isoflurane and enflurane anesthesia. Int J Clin Monit Comput 12: 161–7.

Logothetis NK, Pauls J, Augath M, Trinath T & Oeltermann A (2001) Neurophysiological investigation of the basis of the fMRI signal. Nature 412: 150–7.

Loomis AL, Harwey EN & Hobart G (1935a) Potential rhythms of the cerebral cortex during sleep. Science 81: 597–8.

Loomis AL, Harvey NE & Hobart G (1935b) Further observations on the potential rhythms of the cerebral cortex during sleep. Science 82: 198–200.

Loomis AL, Harvey EN & Hobart G (1938) Distribution of disturbance patterns in the human electroencephalogram, with special reference to sleep. J Neurophysiol 1: 413–40.

Lydic R & Baghdoyan HA (2005) Sleep, anesthesiology, and the neurobiology of arousal state control. Anesthesiology 103: 1268–95.

MacIver HB, Roth SH (1988) Inhalational anaesthetics exhibit pathway-specific and differential actions on hippocampal synaptic responses in vitro. Br J Anaesth 60: 680–691.

Mäkiranta Minna (2004) EEG and BOLD-contrast fMRI in brain. Academic dissertation, Faculty of Medicine, University of Oulu. Acta Universitatis Ouluensis.

Maksimov A, Särkelä M, Långsjö JW, Salmi E, Kaisti KK, Yli-Hankala A, Hinkka-Yli-Salomäki S, Scheinin H & Jääskeläinen SK (2006) Increase in high-frequency EEG activity explains the poor performance of EEG spectral entropy monitor during S-ketamine anesthesia. Clin Neurophysiol 117: 1660–8.

Page 91: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

89

Martella G, DePersis C, Bonsi P, Natoli S, Cuomo D, Bernardi G, Calabresi P & Pisani A (2005) Inhibition of persistent sodium current fraction and voltage-gated L-type calcium current by propofol in cortical neurons: implications for its antiepileptic activity. Epilepsia 46: 624–35.

Mashour GA, Orser BA & Avidan MS (2011) Intraoperative awareness. From neurobiology to clinical practice. Anesthesiology 114: 1218–33.

Massimini M, Huber R, Ferrarelli F, Hill S & Tononi G (2004) The sleep slow oscillation as a traveling wave. J Neuroscience 24(31): 6862–70.

Massimini M, Ferrarelli F, Huber R, Esser SK, Singh H & Tononi G (2005) Breakdown of cortical effective connectivity during sleep. Science 309: 2228–32.

Matta B & Lam AM (1995) Nitrous oxide increases cerebral blood flow velocity during pharmacologically induced EEG silence in humans. J Neurosurg Anesthesiol 7: 89–93.

Mervaala E (2006) Aikuisten epilepsia. In Partanen J, Falck B, Hasan J, Jäntti V, Salmi T & Tolonen U (eds) Kliininen neurofysiologia. 1th ed. Duodecim, Helsinki: 155–72.

Mizobe T, Maghsoudi K, Sitwala K, Tianzhi G, Ou J, Maze M. Antisense technology reveals the α2A adrenoreceptor to be the subtype mediating the hypnotic response to the highly selective agonist, dexmedetomidine, in the locus coeruleus of the rat. J Clin Invest 1996; 98: 1076–80.

Modica PA, Tempelhoff R & White PF (1990a) Pro- and anticonvulsant effects of anesthetics (part I). Anesth Analg 70: 303–15.

Modica PA, Tempelhoff R & White PF (1990b) Pro- and anticonvulsant effects of anesthetics (part II). Anaesth Analg 70: 433–44.

Moeller F, Siebner HR, Wolff S, Muhle H, Boor R, Granert O, Jansen O, Stephani U & Siniatchkin M (2008) Changes in activity of striato-thalamo-cortical network precede generalized spike wave discharges. NeuroImage 39: 1839–49.

Morimoto Y, Matsumoto A, Koizumi Y, Gohara T, Sakabe T & Hagihira S (2005) Changes in the bispectral index during intraabdominal irrigation in patients anesthetized with nitrous oxide and sevoflurane. Anesth Analg 100: 1370–4.

Moruzzi G & Magoun HW (1949) Brain stem reticular formation and activation of the EEG. Electroencephalogr Clin Neurophysiol 1: 455–73.

Murao K, Shingu K, Tsushima K, Takahira K, Ikeda S, Matsumoto H, Nakao S & Asai T (2000) The anticonvulsant effect of volatile anesthetics on penicillin-induced status epilepticus. Anesth Analg 90: 142–7.

Myles PS, Leslie K, McNeil J, Forbes A & Chan MT (2004) Bispectral index monitoring to prevent awareness during anaesthesia: the B-Aware randomized controlled trial. Lancet 363: 1757–63.

Neigh JL, Garman JK & Harp JR (1971) The electroencephalic pattern during anesthesia with ethrane: effects of depth of anesthesia, PaCO2, and nitrous oxide. Anesthesiology 35: 482–7.

Nelskylä KA, Yli-Hankala AM, Puro PH & Korttila KT (2001) Sevoflurane titration using bispectral index decreases postoperative vomiting on phase II recovery after ambulatory surgery. Anesth Analg 93: 1165–9.

Page 92: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

90

Nelson LE, Guo TZ, Lu J, Saper CB, Franks NP & Maze M (2002) The sedative component of anesthesia is mediated by GABAA receptors in an endogenous sleep pathway. Nature Neuroscience 5: 979–84.

Nelson LE, Lu J, Guo T, Saper CB, Franks NP & Maze M (2003) The alpha2-adrenoceptor agonist dexmedetomidine converges on an endogenous sleep-promoting pathway to exert its sedative effects. Anesthesiology 98: 428–36.

Newberg LA, Milde JH & Michenfelder JD (1983) The cerebral metabolic effects of isoflurane at and above concentrations that suppress cortical electrical activity. Anesthesiology 59: 23–8.

Nieminen K, Westeren-Punnonen S, Kokki H, Yppärilä H, Hyvärinen A & Partanen J (2002) Sevoflurane anaesthesia in children after induction of anaesthesia with midazolam and thiopental does not cause epileptiform EEG. Br J Anaesth 89: 853–6.

Nordli DR, Riviello JJ & Niedermeyer E (2011) Seizures and epilepsy in infants to adolescents. In Schomer DL & Lopes da Silva FH (eds) Niedermeyer`s electroencephalography. Basic principles, clinical applications, and related fields. 6th ed. Lippincott Williams &Wilkins: 479–540.

Nordli DR Jr, Moshe SL, Shinnar S, Hesdorffer DC, Sogawa Y, Pellock JM, Lewis DV, Frank LM, Shinnnar RC & Sun S (2012) Acute EEG findings in children with febrile status epilepticus. Results of the FEBSTAT study. Neurology 79: 2180–6.

Nunez PL & Srinivasan R eds. (2006) Eletric fields of the brain. The neurophysics of EEG. Oxford University Press.

Oda Y, Tanaka K, Matsuura T, Hase I, Nishikawa K & Asada A (2006) Nitrous oxide induces paradoxical electroencephalographic changes after tracheal intubation during isoflurane and sevoflurane anesthesia. Anesth Analg 102: 1094–1102.

Ohtahara S & Yamatogi Y (2003) Epileptic encephalopathies in early infancy with suppression-burst, J Clin Neurophysiol 20: 398–407.

Patel AJ, Honoré E, Lesage F, Fink M, Romey G, Lazdunski M. Inhalational anesthetics activate two-pore-domain background K+ channels. Nature Neurosci 1999: 2; 422–426.

Ozcan SO, Ozcan MD, Khan QS, Thompson DM & Chetty PK (2010) Does nitrous oxide affect bispectral index and state entropy when added to a propofol versus sevoflurane anesthetic? J Neurosurg Anesthesiol 22: 309–15.

Patteson KP, Baram TZ & Shinnar S (2014) Origins of temporal lobe epilepsy: febrile seizures and febrile status epilepticus. Neurotherapeutics 11. 242–50.

Peterson DO, Drummond JC & Todd MM (1986) Effects of halothane, enflurane, isoflurane, and nitrous oxide on somatosensory evoked potentials on humans. Anesthesiology 65: 35–40.

Pitkänen A & Engel J Jr (2014) Past and present definitions of epileptogenesis and its biomarkers. Neurotherapeutics 11: 231–41.

Pittau F, LeVan P, Moeller F, Gholipour T, Haegelen C, Zelman R, Dubeau F & Gotman J (2011) Changes preceding interictal epileptic EEG abnormalities: comparison between EEG/fMRI and intracerebral EEG. Epilepsia 52: 1120–9.

Page 93: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

91

Porkkala T, Jäntti V, Kaukinen S & Häkkinen V (1994) Somatosensory evoked potentials during isoflurane anaesthesia. Acta Anaesthesiol Scand 38: 206–10.

Porkkala T, Jäntti V, Kaukinen S & Häkkinen V (1997a) Nitrous oxide has different effects on the EEG and somatosensory evoked potentials during isoflurane anaesthesia in patients. Acta Anaesthesiol Scand 41: 497–501.

Porkkala T, Kaukinen S, Häkkinen V & Jäntti V (1997b) Effects of hypothermia and sternal retractors on median nerve somatosensory evoked potentials. Acta Anaesthesiol Scand 41: 843–8.

Pressler RM, Boylan GB, Morton M, Binnie CD & Rennie JM (2001) Early serial EEG in hypoxic ischaemic encephalopathy. Clin Neurophysiol 112: 31–7.

Quasha AL, Tinker JH & Sharbrough FW (1981) Hypothermia plus thiopental. Prolonged electroencephalographic suppression. Anesthesiology 55: 636–40.

Rampil IJ, Lockhart SH, Eger EI, Yasuda N, Weiskopf RB & Cahalan MK (1991) The electroencephalographic effects of desflurane in humans. Anesthesiology 74: 434–9.

Rampil IJ (1998) A primer for EEG signal processing in anesthesia. Anesthesiology 89: 980–1002.

Rampil IJ (2001) Monitoring depth of anesthesia. Curr Opin Anaesthesiol 14: 649–53. Riad W, Schreiber M, Saeed AB (2007) Monitoring with EEG entropy decreases propofol

requirement and maintains cardiovascular stability during induction of anaesthesia in elderly patients. Eur J Anaesthesiol 24: 684–8.

Rivera R, Segini M, Baltadano A, Perez V (1993) Midazolam in the treatment of status epilepticus in children Crit Care Med 21: 991–4.

Rosen I & Söderberg M (1975) Electroencephalographic activity in children under enflurane anesthesia. Acta Anaesthesiol Scand 19: 361–9.

Rowney DA, Fairgrieve R & Bissonnette B (2004) The effect of cerebral blood flow velocity in children anaesthetized with sevoflurane. Anaesthesia 59: 10–14.

Rudolph U & Antkowiak B (2004) Molecular and neuronal substrates for general anaesthetics. Nature Reviews Neuroscience 5: 709–20

Rundgren M, Rosen I & Friberg H (2006) Amplitude-integrated EEG (aEEG) predicts outcome after cardiac arrest and induced hypothermia. Intensive Care Med 32: 836–42.

Rytky S, Huotari A-M, Alahuhta S, Remes R, Suominen K & Jäntti V (1999) Tibial somatosensory evoked potentials during EEG suppression in sevoflurane anaesthesia. Clin Neurophysiol 110: 1655–8.

Sainio K (2006) Vastasyntyneen normaali EEG. In Partanen J, Falck B, Hasan J, Jäntti V, Salmi T & Tolonen U (eds) Kliininen neurofysiologia. 1st ed. Duodecim: 129–135.

Samra SK, Vanderzant CW, Domer PA & Sackellares JC (1987) Differential effects of isoflurane on human median nerve somatosensory evoked potentials. Anesthesiology 66: 29–35.

Sandin M, Thörn S-E, Dahlqvist A, Wattwil L, Axelsson K & Wattwil M (2008) Effects of pain stimulation on bispectral index, heart rate and blood pressure at different minimal alveolar concentration values of sevoflurane. Acta Anaesthesiol Scand 52: 420–6.

Sato K, Shamoto H & Kato M (2002) Effect of sevoflurane on electrocorticogram in normal brain. J Neurosurg Anesthesiol 14: 63–5.

Page 94: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

92

Sceniak MP (2006) Anesthesia in silico. Anesthesiology 104: 400–2. Schultz B & Schultz A (1998) Epileptiform EEG potentials with sevoflurane. Anaesth Int

Care 26: 329. Schultz A, Schultz B, Grouven U et al (2000) Epileptiform activity in the EEGs of two

nonepileptic children under sevoflurane anaesthesia. Anaesth Int Care 28: 205–7. Schultz B, Schultz A, Grouven U et al (2001) Epileptoform EEG activity: occurrence

under sevoflurane and not during propofol application. Anaesthesist 50: 43–5. Schultz A, Schultz B, Grouven U et al (2001) Sharp transients in the EEGs of non-epileptic

adult patients receiving sevoflurane. Pharm World Sci 23: 82–5. Schultz B, Otto C, Schultz A, Osthaus WA, Krauss T, Dieck T, Sander B, Rahe-Meyer N

& Raymondos K (2012) Incidence of epileptiform EEG activity in children during mask induction of anaesthesia with brief administration of 8% sevoflurane. PLoS ONE 7: e40903. doi:10.1371/journal.pone.0040903.

Schwilden H, Kochs E, Daunderer M, Jeleazcov Ch, Scheller B, Schneider G, Schuttler J, Schwender D, Stockmanns G & Pöppel E (2005) Concurrent recording of AEP, SSEP and EEG parameters during anaesthesia: a factor analysis. Br J Anaesth 95: 197–206.

Scott RC, Besag FMC, Boyd SG, Berry D & Neville BGR (1998) Buccal absorption of midazolam: Pharmacokinetics and EEG pharmacodynamics. Epilepsia 39: 290–4.

Scott RC, King MD, Gadian DG, Neville BGR & Connelly A (2003) Hippocampal abnormalities after prolonged febrile convulsion: a longitudinal MRI study. Brain 126: 2551–7.

Sebel PS, Flynn PJ & Ingram DA (1984) Effects of nitrous oxide on visual, auditory and somatosensory evoked potentials. Br J Anaesth 56: 1403–7.

Sebel PS, Ingram DA, Flynn PJ, Rutherfoord CF & Rogers H (1986) Evoked potentials during isoflurane anaesthesia. Br J Anaesth 58: 580–5.

Sebel PS, Erwin CW & Neville WK (1987) Effects of halothane and enflurane on far and near field somatosensory evoked potentials. Br J Anaesth 59: 1492–6.

Sebel PS, Lang E, Rampil IJ, White PF, Cork R, Jopling M, Smith NT, Glass PSA & Manberg P (1997) A multicenter study of bispectral electroencephalogram analysis for monitoring anesthetic effect. Anesth Analg 84: 891–9.

Sewell JC & Sear JW (2006) Determinants of volatile general anesthetic potency: a preliminary three-dimensional pharmacophore for halogenated anesthetics. Anesth Analg 102: 764–71.

Shorvon S & Ferlisi M (2012) The outcome of therapies in refractory and super-refractory convulsive status epilepticus and recommendations for therapy. Brain 135: 2314–28.

Sigl JC & Chamoun NG (1994) An introduction to bispectral analysis for the electroencephalogram. J Clin Monit 10: 392–404.

Sinha SR (2011) Basic mechanisms of sleep and epilepsy. J Clin Neurophysiol 28: 103–10. Sloan TB & Koht A (1985) Depression of cortical somatosensory evoked potentials by

nitrous oxide. Br J Anaesth 57: 849–52. Sloan TB (1998) Anesthetic effects on electrophysiologic recordings. J Clin Neurophysiol

15: 217–26.

Page 95: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

93

Sloan TB & Jäntti V (2008) Anesthetic effects on evoked potentials. In Nuwer MR (ed) Handbook of clinical neurophysiology. Intraoperative monitoring of neural function. Vol 8. Elsevier, Amsterdam: 94–126.

Sonner JM, Antognini JF, Dutton RC, Flood P, Gray AT, Harris RA, Homanics GE, Kendig J, Orser B, Raines DE, Trudell J, Vissel B & Eger II EI (2003) Inhaled anesthetics and immobility: mechanisms, mysteries and minimum alveolar anesthetic concentration. Anesth Analg 97: 718–40.

Soto RG, Smith RA, Zaccaria AL & Miguel RV (2006) The effect of addition of nitrous oxide to a sevoflurane anesthetic on BIS, PSI and entropy. J Clin Monit Comput 20: 145–50.

Steriade M, McCormick DA & Sejnowski TJ (1993a) Thalamocortical oscillations in the sleeping and aroused brain. Science 262: 679–85.

Steriade M, Nunez A & Amzica F (1993b) A novel slow (<1 Hz) oscillation of neocortical neurons in vivo: depolarizing and hyperpolarizing components. J Neuroscience 13: 3252–65.

Steriade A, Nunez A & Amzica F (1993c) Intracellular analysis of relations between the slow neocortical oscillation and other sleep rhythms of the electroencephalogram. J Neuroscience 13: 3266–83.

Steriade M, Amzica F & Contreras D (1994) Cortical and thalamic cellular correlates of electroencephalographic burst-suppression. Electroenceph Clin Neurophysiol 90: 1–16.

Steriade M & Amzica F (2003) Sleep oscillations developing into seizures in corticothalamic systems. Epilepsia 44 (suppl 12): 9–20

Steriade M (2005) Sleep, epilepsy and thalamic reticular inhibitory neurons. Trends Neurosci 28: 317–24.

Steriade M (2006) Grouping of brain rhythms in corticothalamic systems. Neuroscience 137: 1087–1106.

Swank RL & Watson W (1949) Effects of barbiturates and ether on spontaneous electrical activity of dog brain. J Neurophysiol 12: 137–60.

Terasako K &Ishii S (1996) Postoperative seizure-like activity following sevoflurane anesthesia. Acta Anaesthesiol Scand 40: 953–4.

Thordstein M, Flisberg A, Löfgren N, Bågenholm R, Lindecrantz K, Wallin BG & Kjellmer I (2004) Spectral analysis of burst periods in EEG from healthy and post-asphyctic full-term neonates. Clin Neurophysiol 115: 2461–6.

Thornton C, Creagh-Barry P, Jordan C, Luff NP, Dore CJ, Henley M & Newton DEF (1992) Somatosensory and auditory evoked responses recorded simultaneously: differential effects of nitrous oxide and isoflurane. Br J Anaesth 68: 508–14.

Thornton C & Sharpe RM (1998) Evoked responses in anaesthesia. Br J Anaesth 81: 771–81.

Thömke F, Brand A & Weilemann SL (2002) The temporal dynamics of postanoxic burst-suppression EEG.. J Clin Neurophysiol 19: 24–31.

Treiman DM (1995) Electroclinical features of status epilepticus. J Clin Neurophysiol 12: 343–62.

Page 96: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

94

Treiman DM (2001) GABAergic mechanisms in epilepsy. Epilepsia 42 (suppl 3): 8–12. Tung A & Mendelson WB (2004a) Anesthesia and sleep. Sleep Med Rev 8: 213–25. Tung A, Bergmann B, Herrera S, Cao D & Mendelson WB (2004b) Recovery from sleep

deprivation occurs during propofol anesthesia. Anesthesiology 100: 1419–26. Wakamori M, Ikemoto Y, Akaike N. Effects of two volatile anesthetics and a volatile

convulsant on the excitatory and inhibitory amino acid responses in dissociated CNS neurons in the rat. Journal of Neurophysiol 1991; 66: 2014–2012.

Vakkuri AP, Lindgren L, Korttila KT & Yli-Hankala AM (1999) Transient hyperdynamic response associated with controlled hypocapneic hyperventilation during sevoflurane-nitrous oxide mask induction in adults. Anesth Analg 88: 1384–1388.

Vakkuri A, Jäntti V, Särkelä M, Lindgren L, Korttila K & Yli- Hankala A (2000) Epileptiform EEG during sevoflurane mask induction: effect of delaying the onset of hyperventilation. Acta Anaesthesiol Scand 44: 713–719.

Vakkuri AP, Yli-Hankala AM, Särkelä M, Lindgren L, Mennander S, Korttila KT, Saarnivaara L & Jäntti V (2001) Sevoflurane mask induction of anaesthesia is associated with epileptiform EEG in children. Acta Anaesthesiol Scand 45: 805–811.

Vakkuri A, Yli-Hankala A, Talja P, Mustola S, Tolvanen-Laakso H, Sampson T & Viertiö-Oja H (2004) Time-frequency balanced spectral entropy as a measure of anesthetic drug effect in central nervous system during sevoflurane, propofol, and thiopental anesthesia. Acta Anaesthesiol Scand 48: 145–153.

Vakkuri A, Yli-Hankala A, Sandin R, Mustola S, Hoymork S, Nyblom S, Talja P, Sampson T, van Gils M & Viertiö-Oja H (2005) Spectral entropy monitoring is associated with reduced propofol use and faster emergence in propofol-nitrous oxide-alfentanil anesthesia. Anesthesiology 103: 274–9.

Vakkuri AP, Seitsonen ER, Jäntti VH, Särkelä M, Korttila KT, Paloheimo MPJ & Yli-Hankala AM (2005) A rapid increase in the inspired concentration of desflurane is not associated with epileptiform encephalogram. Anesth Analg 101: 396–400.

Walder B, Tramer MR, Seeck M (2002) Seizure-like phenomena and propofol. Neurology 58: 1327–32.

Vandesteene A, Nogueira MC, Mavroudakis N, Defevrimont M, Brunko E & Zegers de Beyl D (1991) Synaptic effects of halogenated anesthetics on short-latency SEP. Neurology 41: 913–8.

Vandesteene A, Mavroudakis N, Defevrimont M, Brunko E & Zegers de Beyl D (1993) Topographic analysis of the effects of isoflurane anesthesia on SEP. Electroenceph Clin Neurophysiol 88: 77–81.

Watts ADJ, Herrick IA, McLachlan RS, Craen RA & Gelb AW (1999) The effect of sevoflurane and isoflurane anesthesia on interictal spike activity among patients with refractory epilepsy. Anesth Analg 89: 1275–1281.

Velly LJ, Rey MF, Bruder NJ, Gouvitsos FA, Witjas T, Regis JM, Peragut JC & Gouin FM (2007) Differential dynamic of action on cortical and subcortical structures of anesthetic agents during induction of anesthesia. Anesthesiology 107: 202–12.

Page 97: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

95

Wennberg RA, Quesney LF & Villemure J-G (1997) Epileptiform and non-epileptiform activity from isolated cortex after functional hemispherectomy. Electroenceph Clin Neurophysiol 102: 437–42.

Wennberg RA & Lozano AM (2003) Intracranial volume conduction of cortical spikes and sleep potentials recorded with deep brain stimulating electrodes. Clin Neurophysiol 114: 1403–18.

Veronesi MC, Kubek DJ & Kubek MJ (2008) Isoflurane exacerbates electrically evoked seizures in amygdala-kindled rats during recovery. Epilepsy Research 82: 15–20.

Westeren-Punnonen S, Yppärilä-Wolters H, Partanen J, Nieminen K, Hyvärinen A & Kokki H (2008) Somatosensory evoked potentials by median nerve stimulation in children during thiopental/sevoflurane anesthesia and the additive effects of ketoprophen and fentanyl. Paediatr Anaesth 107: 799–805.

White NS & Alkire MT (2003) Impaired thalamocortical connectivity in humans during general-anesthetic-induced unconsciousness. Neuroimage 19: 402–11.

Viertiö-Oja H, Maja V, Särkelä M, Talja P, Tenkanen N, Tolvanen-Laakso H, Paloheimo M, Yli-Hankala A & Meriläinen P (2004) Description of the EntropyTM algorithm as applied in the Datex-Ohmeda S/5TM Entropy module. Acta Anaesthesiol Scand 48: 154–61.

Wilson MT, Sleigh JW, Steyn-Ross A & Steyn-Ross ML (2006) General anesthetic-induced seizures can be explained by a mean-field model of cortical dynamics. Anesthesiology 104: 588–93.

Wilson-Smith E, Karsli C, Luginbuehl IA & Bissonnette B (2003) The effect of nitrous oxide on cerebral blood flow velocity in children anesthetized with propofol. Acta Anaesthesiol Scand 47: 307–11.

Wittmer LL, Hu Y, Kalkbrenner M, Evers AS, Zorumski CF, Covey DF. Enantioselectivity of steroid-induced gamma-aminobutyric acid A receptor modulation and anesthesia. Mol Pharmacol 1996; 50: 1581–1586.

Wolter S, Friedel C, Böhler K, Hartmann U, Kox WJ & Hensel M (2006) Presence of 14 Hz spindle oscillation in the human EEG during sleep. Clin Neurophysiol 117: 157–68.

Woodforth IJ, Hicks RG, Crawford MR, Stephen JPH & Burke DJ (1997) Electroencephalographic evidence of seizure activity under deep sevoflurane anesthesia in a nonepileptic patient. Anesthesiology 87: 1579–82.

Yamakura T, Harris RA. Effects of gaseous anesthetics nitrous oxide and xenon on ligand-gated ion channels. Anesthesiology 2000; 93: 1095–1100.

Yano T, Okubo S, Naruo H, Iwasaki T, Takasaki M & Tsuneyoshi I (2008) Two cases with past and family history of febrile convulsion developed seizure-like movements during sevoflurane anesthesia. Anesthesiology 109: 571.

Yasuda N, Lockhart SH, Eger EI, Weiskopf RB, Liu J, Laster M, Taheri S & Peterson NA (1991) Comparison of kinetics of sevoflurane and isoflurane in humans. Anesth Analg 72: 316–24.

Page 98: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

96

Yli-Hankala A, Heikkilä H, Värri A & Jäntti V (1990a) Correlation between EEG and heart rate variation in deep enflurane anaesthesia. Acta Anaesthesiol Scand 34: 138–43.

Yli-Hankala A & Jäntti V (1990b) EEG burst suppression pattern correlates with the instantaneous heart rate under isoflurane anaesthesia. Acta Anaesthesiol Scand 34: 665–8.

Yli-Hankala A, Lindgren L, Porkkala T & Jäntti V (1993a) Nitrous oxide-mediated activation of the EEG during isoflurane anaesthesia in patients. Br J Anaesth 70: 54–7.

Yli-Hankala A, Jäntti V, Pyykkö I & Lindgren L (1993b) Vibration stimulus induced EEG bursts in isoflurane anesthesia. Electroenceph Clin Neurophysiol 87: 215–20.

Yli-Hankala A, Loula P, Annila P, Lindgren L & Jäntti V (1993c) Atropine abolishes electroencephalogram-associated heart rate changes without an effect on respiratory sinus arrhythmia during anaesthesia in humans. Acta Physiol Scand 149: 435–40.

Yli-Hankala A, Vakkuri A, Särkelä M, Lindgren L, Korttila K & Jäntti V (1999) Epileptiform electroencephalogram during mask induction of anesthesia with sevoflurane. Anesthesiology 91: 1596–1603.

Young GB (2000) The EEG in coma. J Clin Neurophysiol 17: 473–85. Young GB (2003) Mechanisms of consciousness with emphasis on the cerebral cortical

component. In: Neural mechanisms of anesthesia pp 13–20. Eds. Antognini JF, Carstens EE & Raines DE, Humana Press, New Jersey

Zacharias M (1997) Convulsive movements with sevoflurane in children. Anaesth Int Care 25: 727.

Zaret BS (1985) Prognostic and neurophysiological implications of concurrent burst suppression and alpha patterns in the EEG of post-anoxic coma. Electroencephalogr Clin Neurophysiol 61: 199–209.

Zeilhofer HU, Swandulla D, Geisslinger G, Brune K. Differential effects of ketamine enantiomers on NMDA receptor currents in cultured neurons. Eur J Pharmacol 1992; 213: 155–158.

Åkeson J & Didriksson I (2004) Convulsions on anaesthetic induction with sevoflurane in young children. Acta Anaesthesiol Scand 48: 405-407.

Page 99: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

97

Original publications

I Jäntti V, Sonkajärvi E, Mustola S, Rytky S, Kiiski P & Suominen K (1998) Single-sweep cortical somatosensory evoked potentials: N20 and evoked bursts in sevoflurane anaesthesia. Electroencephalogr clin Neurophysiol 108: 320–324.

II Mäkiranta M, Ruohonen J, Suominen K, Niinimäki J, Sonkajärvi E, Kiviniemi V, Seppänen T, Alahuhta S, Jäntti V & Tervonen O (2005) BOLD signal change preceeds interictal spike activity in EEG – a dynamical model of experimental penicillin induced focal epilepsy in deep anesthesia. Neuroimage 27: 715–724.

III Sonkajärvi E, Puumala P, Erola T, Baer GA, Karvonen E, Suominen K & Jäntti V (2008) Burst-suppression during propofol anaesthesia recorded from scalp and subthalamic electrodes: report of three cases. Acta Anaesthesiol Scand 52: 274–279.

IV Sonkajärvi E, Alahuhta S, Suominen K, Hakalax N, Vakkuri A, Löppönen H, Ohtonen P & Jäntti V (2009) Topographic electroencephalogram in children during mask induction of anaesthesia with sevoflurane. Acta Anaesthesiol Scand 53: 77–84.

V Sonkajärvi E, Alahuhta S, Suominen K, Rytky S, Kumpulainen T, Ohtonen P, Karvonen E, & Jäntti V (2015) Epileptiform EEG activity induced by rapid sevoflurane anaesthesia induction. Manuscript.

Reprinted with permission from John Wiley & Sons Ltd (III, IV) and Elsevier

(I, II)

Original publications are not included in the electronic version of the dissertation.

Page 100: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

98

Page 101: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

A C T A U N I V E R S I T A T I S O U L U E N S I S

Book orders:Granum: Virtual book storehttp://granum.uta.fi/granum/

S E R I E S D M E D I C A

1306. Kujala, Tiia (2015) Acute otitis media in young children : randomized controlledtrials of antimicrobial treatment, prevention and quality of life

1307. Kämppi, Antti (2015) Identifying dental restorative treatment need in healthyyoung adults at individual and population level

1308. Myllymäki, Satu-Marja (2015) Specific roles of epithelial integrins in chemical andphysical sensing of the extracellular matrix to regulate cell shape and polarity

1309. Antonoglou, Georgios (2015) Vitamin D and periodontal infection

1310. Valtokari, Maria (2015) Hoitoon pääsyn moniulotteisuus erikoissairaanhoidossa

1311. Toljamo, Päivi (2015) Dual-energy digital radiography in the assessment of bonecharacteristics

1312. Kallio-Pulkkinen, Soili (2015) Effect of display type and room illuminance inviewing digital dental radiography : display performance in panoramic andintraoral radiography

1313. Roivainen, Eka (2015) Validity in psychological measurement : an investigation oftest norms

1314. Puhto, Ari-Pekka (2015) Prosthetic joint infections of the hip and knee :treatment and predictors of treatment outcomes

1315. Pentikäinen, Ilkka (2015) Distal chevron osteotomy for hallux valgus surgery :role of fixation and postoperative regimens in the long-term outcomes of distalchevron osteotomy – a randomised, controlled, two-by-two factorial trial of 100patients

1316. Mikkonen, Paula (2015) Low back pain and associated factors in adolescence : acohort study

1317. Hakalahti, Antti (2015) Efficacy and safety of radiofrequency catheter ablation inthe treatment of atrial fibrillation

1318. Hannula, Virva (2015) The prevalence of diabetic retinopathy and its effect onsocial well-being and health related quality of life in children and young adultswith type 1 diabetes

1319. Leppänen, Virpi (2015) Epävakaan persoonallisuuden hoitomallitutkimus Oulunmielenterveyspalveluissa.

1320. Piira, Olli-Pekka (2015) Effects of emotional excitement on cardiovascularregulation

Page 102: OULU 2015 D 1321 UNIVERSITY OF OULU P.O. Box 8000 FI-90014 …jultika.oulu.fi/files/isbn9789526209722.pdf · 2015-12-16 · isbn 978-952-62-0971-5 (paperback) isbn 978-952-62-0972-2

UNIVERSITY OF OULU P .O. Box 8000 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

Professor Esa Hohtola

University Lecturer Santeri Palviainen

Postdoctoral research fellow Sanna Taskila

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Director Sinikka Eskelinen

Professor Jari Juga

University Lecturer Anu Soikkeli

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-0971-5 (Paperback)ISBN 978-952-62-0972-2 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1321

ACTA

Eila Sonkajärvi

OULU 2015

D 1321

Eila Sonkajärvi

THE BRAIN'S ELECTRICAL ACTIVITY IN DEEP ANAESTHESIAWITH SPECIAL REFERENCE TO EEGBURST-SUPPRESSION

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE;MEDICAL RESEARCH CENTER OULU;OULU UNIVERSITY HOSPITAL