Uploaded by gwendy801

Essential Neuroanesthesia

advertisement
https://t.me/Anesthesia_Books
ESSENTIALS OF NEUROANESTHESIA
This page intentionally left blank
ESSENTIALS OF
NEUROANESTHESIA
Edited by
HEMANSHU PRABHAKAR
Department of Neuroanaesthesiology and Critical Care
All India Institute of Medical Sciences
New Delhi, India
Academic Press is an imprint of Elsevier
125 London Wall, London EC2Y 5AS, United Kingdom
525 B Street, Suite 1800, San Diego, CA 92101-4495, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
Copyright © 2017 Elsevier Inc. All rights reserved.
No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical,
including photocopying, recording, or any information storage and retrieval system, without permission in writing from
the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our
arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be
found at our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be
noted herein).
Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding,
changes in research methods, professional practices, or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any
information, methods, compounds, or experiments described herein. In using such information or methods they should be
mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury
and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of
any methods, products, instructions, or ideas contained in the material herein.
Library of Congress Cataloging-in-Publication Data
A catalog record for this book is available from the Library of Congress
British Library Cataloguing-in-Publication Data
A catalogue record for this book is available from the British Library
ISBN: 978-0-12-805299-0
For information on all Academic Press publications visit our website at
https://www.elsevier.com/books-and-journals
Publisher: Mara Conner
Acquisition Editor: Melanie Tucker
Editorial Project Manager: Kristi Anderson
Production Project Manager: Edward Taylor
Designer: Maria Ines Cruz
Typeset by TNQ Books and Journals
Dedicated to my parents—Avinash and Kanti Prabhakar
The best gifts they stored for me—Kavita and Hemant, who in turn gifted me Sunil and Deepali
To those who mean the world to me—Pallavi, Anavi, and Amyra
To Aishwarya, Avi, and Anav
This page intentionally left blank
Contents
II
List of Contributors
xvii
Foreword
xix
Preface
xxi
Acknowledgments
xxiii
Introduction and Brief History of Neuroanesthesia
W. S. Jellish
xxv
NEUROPHYSIOLOGY
4. Neurophysiology
M. SETHURAMAN
Intracranial Pressure
Introduction
Normal Intracranial Pressure
Cerebral Compliance
Importance of Intracranial Pressure
Summary
Cerebral Blood Flow
Introduction
Vascular Anatomy
Summary
Brain Metabolism
Introduction
Normal Cerebral Metabolism
Summary
Cerebrospinal Fluid
Introduction
Ventricular System
Summary
The Spinal Cord
Introduction
Anatomy
Organization of the Spinal Cord
Summary
References
I
NEUROANATOMY
1. Neuroanatomy
D. GUPTA
Introduction
Embryological Differentiation of Different Parts of Brain
Anatomy of Brain
Vascular Supply of the Brain
The Meninges and Cerebrospinal Fluid
Acknowledgment
References
3
4
4
30
33
39
40
2. Neuroembryology
G.P. SINGH
Formation of Zygote
Formation of Blastocyst
Formation of Embryonic or Germ Disc
Formation of Definitive Notochord
Development of Nervous System
References
41
41
42
44
45
50
5. Brain Protection in Neurosurgery
H. EL BEHEIRY
Introduction
Nonpharmacological Strategies
Mild Hypothermia
Blood Pressure Control
Induced Arterial Hypertension
Normoglycemia
Target Hemoglobin Concentration
Pharmacological Strategies
Nonanesthetic Agents
Anesthetic Agents
Conclusion
References
3. Blood–Brain Barrier
A.K. KHANNA AND E. FARAG
Introduction
Permeability at the Blood–Brain Barrier
Cellular and Molecular Effects of Anesthetics on the
Blood–Brain Barrier
Clinical and Experimental Implications of Anesthetics
on the Blood–Brain Barrier
Conclusion
References
62
62
62
62
63
68
68
68
68
74
74
74
74
79
79
79
79
83
83
83
84
84
89
89
51
51
52
54
56
56
vii
91
91
92
93
94
94
95
96
97
97
98
98
viii
CONTENTS
III
Cerebral Microdialysis
Conclusion
References
NEUROPHARMACOLOGY
158
159
159
9. Multimodal Monitoring
6. Neuropharmacology
A. DEFRESNE AND V. BONHOMME
P. GANJOO AND I. KAPOOR
Anesthetic Drugs and Sedatives
Intravenous Anesthetic Agents
Inhalational Anesthetic Agents
Neuromuscular Blocking Agents
Local Anesthetic Agents
Miscellaneous Drugs
Future Directions in Neuropharmacology
Conclusion
References
104
104
111
115
116
116
116
116
118
7. Anesthetic Agents: Neurotoxics or
Neuroprotectives?
Introduction
Temperature
Oxygen Transport, Hemodynamics, and Brain
Metabolism
Intracranial Pressure Monitoring
Electroencephalography and Depth of Anesthesia
Monitoring
Miscellaneous
Integration of Information and DecisionHelping Systems
Clinical Pearls
References
161
162
162
171
173
174
175
176
176
J. FIORDA-DIAZ, N. STOICEA AND S.D. BERGESE
Introduction
Pharmacological Considerations
Anesthesia Practice: Clinical Outcomes
Anesthesia and Fragile Brain
Conclusion
Abbreviations
References
123
124
126
127
127
128
128
IV
NEUROMONITORING
8. Neuromonitoring
V.J. RAMESH AND M. RADHAKRISHNAN
Introduction
Cerebral Blood Flow
Transcranial Sonography
Thermal Diffusion Flowmetry
Laser Doppler Flowmetry
Intra-Arterial 133Xenon
CT Perfusion
Xenon Enhanced CT
Positron Emission Tomography
Single Photon Emission Computed Tomography
Magnetic Resonance Imaging
Intracranial Pressure
Electroencephalogram
Evoked Potential Monitoring
Motor Evoked Potentials
Depth of Anesthesia
Cerebral Oxygenation Monitoring
Jugular Venous Oximetry
Regional Cerebral Oximetry
Brain Tissue Oxygen Monitoring
134
134
139
139
139
139
139
139
140
140
140
140
143
145
149
150
152
152
154
156
V
POSITIONS IN NEUROSURGERY
10. Positioning in Neurosurgery
G. SINGH
Introduction
Historical Background
Principles of Positioning
The Conduct of Positioning
Surgical Approach for Craniotomies
Positioning for Craniotomy
Positions Used for Craniotomies
Surgical Approach for Procedures of the Spine
Patient Positioning For Spinal Procedures
Conclusion
Abbreviations
References
184
184
184
185
186
187
189
195
195
203
203
204
VI
PREANESTHETIC EVALUATION
11. Preanesthetic Evaluation of Neurosurgical
Patients
R. MARIAPPAN
Introduction
Preoperative Evaluation of Patient-Related Risk Factors
Preoperative Evaluation of Specific
Neurosurgical Conditions
References
209
210
217
225
CONTENTS
VII
16. Anesthesia for Epilepsy Surgery
N. GUPTA
NEUROSURGERY
12. Supratentorial Lesions
H. BHAGAT AND S. MAHAJAN
Introduction
Classification
Pathophysiology and Clinical Correlations
Clinical Features
Neuroimaging
Intraoperative Considerations: The Team
Approach
Anesthetic Management
Intraoperative Management
Emergence From Anesthesia
Postoperative Management
Awake Craniotomy
Conclusions
Acknowledgment
References
231
232
233
235
235
236
236
238
240
241
242
245
245
245
13. Emergence From Anesthesia
Introduction
Surgical Management of Epilepsy
Types of Surgical Treatment
Presurgical Evaluation
Anesthesia for Epilepsy Surgery
Effect of Anesthetic Agents in Patients With Epilepsy
Antiepileptic Drug Interactions
Preanesthetic Evaluation and Preparation
Anesthetic Management of Preoperative Procedures
Anesthesia for Intracranial Electrode Insertion
Anesthetic Management of Resection of Seizure Focus
Awake Craniotomy
Resection of Epileptogenic Focus Under General
Anesthesia
Neurostimulation for Drug-Resistant Epilepsy
Anesthetic Management of the Patient With
Epilepsy for Incidental Surgery
Abbreviations
References
247
248
248
250
251
252
252
Introduction
Epidemiology
Classification
Cause
Pathophysiology
Diagnosis
Management
Treatment
Conclusions
References
14. Anesthesia for Posterior Fossa Surgery
18. Aneurysmal Subarachnoid Hemorrhage
C. MAHAJAN
255
255
256
256
264
271
272
272
273
15. Transesophageal Echocardiography
A. LELE AND V. KRISHNAMOORTHY
Introduction
Basics of Transesophageal Echocardiography
Summary
References
300
301
302
303
304
17. Refractory Status Epilepticus
K. SANDHU AND N. GUPTA
Introduction
Anatomy
Clinical Presentation
Perioperative Management of Patients for Posterior
Fossa Surgery
Venous Air Embolism
Postoperative Management
Complications
Abbreviations
References
285
286
286
287
288
288
290
291
292
294
295
295
M. PANEBIANCO AND A. MARSON
M. ECHEVERRÍA, J. FIORDA-DIAZ, N. STOICEA AND S.D. BERGESE
Introduction
Neurophysiological Response During Emergence in
Neurosurgical Patients
Specific Perioperative Considerations
Delayed Emergence and Arousal
Complications
Conclusion
References
ix
277
277
283
283
History
Introduction
Clinical Presentation and Diagnosis
Grading of Subarachnoid Hemorrhage
Initial Management Concerns in Neurocritical
Care Unit
Timing of Surgery
Clipping or Coiling
Evaluation of a Patient With Subarachnoid
Hemorrhage for Anesthesia
Anesthetic Management
Temporary Clipping and Brain Protection Strategy
Intraoperative Aneurysm Rupture
Giant Aneurysms and Circulatory Arrest
Endovascular Management for Aneurysm Ablation
Postoperative Management of Patients
Conclusion
References
309
309
310
310
311
311
311
312
313
314
316
316
317
319
321
327
327
328
328
330
330
331
331
333
333
333
x
CONTENTS
19. Circulatory Arrest
Rapid Ventricular Pacing–Assisted Cerebral Blood
Flow Arrest
References
D.E. TRAUL
Introduction
Deep Hypothermic Circulatory Arrest
Anesthesia Management
Complications
Adenosine-Induced Circulatory Arrest
Anesthesia Considerations
Complications
Summary
References
339
339
340
341
341
342
342
342
343
20. Cerebrovascular Disease
M. ABRAHAM AND M. MARDA
Intracerebral Hemorrhage
Incidence and Risk Factors
Imaging
Clinical Presentation
Management of Intracerebral Hemorrhage
Arteriovenous Malformations
Cause and Incidence
Natural History
Pathophysiologic Effects and Clinical Presentation
Grading of Arteriovenous Malformations
Imaging
Cerebral Hemodynamics in Arteriovenous
Malformation
Management
Surgical Resection of Arteriovenous Malformation
Anesthetic Considerations for Resection of
Arteriovenous Malformation
Postoperative Management
Anesthetic Considerations for Arteriovenous
Malformation Embolization
Complications During Arteriovenous Malformation
Embolization
Pediatric Arteriovenous Malformations
Pregnancy and Arteriovenous Malformations
Vein of Galen Aneurysmal Malformations
Dural Arteriovenous Fistula
Clinical Presentation
Management
Carotid Endarterectomy
Preoperative Evaluation
Management of Carotid Artery Disease
Monitoring
Intraoperative Management
Postoperative Complications and Outcomes
Coronary Angioplasty and Stenting
Moyamoya Disease
Management of Moyamoya Disease
References
346
346
346
346
348
352
352
353
353
353
354
354
354
355
355
356
356
357
357
358
358
360
360
360
360
362
362
362
363
363
363
363
364
364
21. Flow Arrest in Cerebrovascular Surgery
M.L. JAMES, M.-A. BABI AND S.A. KHAN
Deep Hypothermic Circulatory Arrest
Adenosine-Assisted Cerebral Blood Flow Arrest
367
370
372
373
22. Neuroendocrine Lesions
P.K. BITHAL
Hypothalamic-Pituitary–Adrenal
Axis Evaluation
Neuroendocrine Response Related to
Anesthesia and Surgery
Pituitary Gland Adenomas
Physiology of Pituitary Gland
Endocrine Diseases
Nonfunctioning Tumors
Intraoperative Considerations
Advantages of Endoscopic Endonasal
Approach
Relative Contraindications to
Transsphenoidal Approach
Intraoperative Issues
Disorder of Water and Electrolytes
References
376
377
377
379
380
382
383
384
384
384
387
389
23. Pituitary Apoplexy
S.S. THOTA
Clinical Features
Management
References
395
395
397
24. Spinal Surgery
M.S. TANDON AND D. SAIGAL
Introduction
Spine
Types of Spine Surgeries
Surgical Approaches to the Spine
Common Spine Disorders
Imaging in Spine Lesions
Positioning for Spine Surgeries
Neurophysiological Intraoperative Monitoring
During Spine Surgeries
Preanesthetic Assessment and Optimization
Anesthesia Management
Postoperative Management
Special Considerations
Conclusion
References
400
400
401
403
403
417
417
418
420
423
430
431
437
437
25. Postoperative Visual Loss
K.M. KLA AND L.A. LEE
Introduction
Central Retinal Artery Occlusion
Ischemic Optic Neuropathy
Cortical Blindness
Recent Advances
Conclusion
References
441
441
442
442
443
445
445
xi
CONTENTS
26. Neuroendoscopy
S. MONINGI AND D.K. KULKARNI
Introduction
Anesthetic Goals and Management
Anesthetic Management of Specific
Neuroendoscopic Procedures
Advances in Neuroendoscopy
Conclusion
Clinical Pearls
References
447
450
453
466
467
467
468
N. FÀBREGAS AND L. SALVADOR
513
514
515
516
516
517
517
F. RABAI AND R. RAMANI
471
471
472
473
475
477
477
28. Anesthesia for Functional Neurosurgery
S.K. DUBE
Introduction
Procedure
Anesthetic Consideration
Anesthetic Techniques
Complications
Anesthesia in Patients With Deep Brain Stimulator In Situ
Conclusion
References
510
31. Magnetic Resonance Imaging: Anesthetic
Implications
27. Pressure Inside the Neuroendoscope
Introduction
Indications and Procedures
How Do Neurosurgeons Perform an
Intraventricular Endoscopic Procedure?
Anesthetic Procedure: What to Take Into Account?
Perioperative Complications
Conclusion
References
Anesthetic Management of Endovascular Coiling
Anesthetic Management of Endovascular Embolization of
Arteriovenous Malformation, Arteriovenous
Fistula, and Vein of Galen Malformation
Anesthesia for Stroke Interventions
Issues Related to Radiation During Neurointervention
Anesthesia for Stereotactic Radiosurgery
Pregnancy and Neuroradiology
Clinical Pearls
References
479
479
481
484
485
486
486
486
29. Awake Craniotomy
P.H. MANNINEN AND T. Y. YEOH
Introduction: The Road From X-Ray to Magnetic
Resonance Imaging
Principles of Nuclear Magnetic Resonance and Magnetic
Resonance Imaging
Various Types of Signals Recorded
Hazards Related to Magnetic Resonance Imaging
Magnetic Resonance Imaging Safety:
General Considerations
Magnetic Resonance Imaging Safety: Management of Cardiac
Implantable Electronic Devices and Other
Implantable Devices
Anesthesia for Magnetic Resonance Imaging
Research Applications/Emerging Clinical
Applications of Magnetic Resonance Imaging
References
519
521
522
524
526
527
530
531
532
IX
NEUROTRAUMA
32. Neurotrauma
Introduction
Patient Selection
Awake Craniotomy for Tumor Surgery
Awake Craniotomy for Epilepsy
Conclusion
References
489
490
490
496
499
499
VIII
NEURORADIOLOGY
30. Anesthesia for Neuroradiology
K. SRIGANESH AND B. VINAY
Introduction
Issues Relating to Anesthesia Care in Neuroradiology
Anesthesia for Computed Tomographic Study
Anesthesia for Magnetic Resonance Imaging Study
Anesthesia for Diagnostic Angiography
505
506
506
506
510
D. PADMAJA, A. LUTHRA AND R. MITRA
Traumatic Brain Injury
Introduction
Definition
Epidemiology
Classification of Traumatic Brain Injury
Physiologic Response to Brain Injury
Neuroimaging
Severity of Traumatic Brain Injury
Management of Traumatic Brain Injury
Outcome
Emerging Treatment Modalities
Conclusion
Spine and Spinal Cord Trauma
Introduction
Epidemiology
Classification of Spinal Injury
Pathophysiology of Spinal Cord Trauma
Systemic Complications of Spinal
Cord Injuries
536
536
536
536
537
543
545
549
549
559
559
560
560
560
560
561
563
565
xii
CONTENTS
Management of Spine and Spinal Cord Injury
Emerging Treatment Modalities
References
567
578
582
33. Biomarkers in Traumatic Brain Injury
J. ŽUREK
Introduction
Conclusion
References
587
590
590
X
NEUROINTENSIVE CARE
34. Neurological Critical Care
G.S. UMAMAHESWARA RAO AND S. BANSAL
Introduction
History of Neurocritical Care
Design of a Neurocritical Care Unit
Clinical Conditions Requiring Admission to
Neurocritical Care Unit
Justification for Neurological Critical Care Units
Pathophysiological Issues in Neurological Critical Care
Management of Patients in a Neurological
Intensive Care Unit
Management of General Systemic Physiology
Specific Therapeutic Issues in Individual
Clinical Conditions
Advanced Neuromonitoring
Outcomes of Neurological Intensive Care Unit
End-of-Life Issues in Neurological Critical Care
Clinical Pearls
References
595
596
596
596
596
597
598
598
603
603
606
606
608
608
Intraoperative Management
Postoperative Considerations
Management of Specific Conditions
Conclusion
References
37. Fluid and Blood Transfusion in
Pediatric Neurosurgery
S. RAJAN AND S. RAO
Introduction
Fluid and Electrolyte Choices
Type of Fluids for Perioperative Administration in
Pediatric Patients
Fluid Management in Pediatric Neurosurgery
Osmotherapy
Fluid and Electrolyte Disturbances in Pediatric
Neurosurgery
Blood Transfusion
Blood Components
Special Situations
Epilepsy Surgery
Scoliosis
Conclusion
References
Introduction
Implications of Surgical Stress and Anesthesia
on the Elderly
Neurosurgical Concerns Unique to the Elderly
Conclusion
References
Introduction
Definitions, Epidemiology, and Pathophysiology
Risk Factors
Prevention
Screening Tools
Treatment
Outcome
References
653
653
654
658
658
661
661
663
664
664
665
666
666
40. Pregnancy
XI
V. SINGHAL
SPECIAL CONSIDERATIONS
36. Pediatric Neuroanesthesia
G.P. RATH
Overview
Pediatric Neurophysiology
General Principles of Pediatric Neuroanesthesia
647
647
648
649
649
649
650
650
39. Postoperative Cognitive Dysfunction
35. Antibiotics: Prophylactic and Therapeutics
616
620
623
645
645
646
38. Geriatric Neuroanesthesia
S. ERB, L.A. STEINER AND C. OETLIKER
613
613
643
644
S. TRIPATHY
A. BOROZDINA, L. PORCELLA AND F. BILOTTA
Introduction
Principles of Antimicrobial Therapy in Neurosurgery
Treatment of Central Nervous System Infections in the
Neurosurgical Patient
Antimicrobial Prophylaxis in Neurosurgery
References
631
633
633
641
641
629
629
630
Requirement of Neurosurgery During Pregnancy
Physiological Alterations During Pregnancy
Effect of Anesthetic Agents on Fetal Outcome
Uteroplacental Drug Transfer and Neonatal Depression
Timing and Method of Delivery
Anesthetic Considerations During Pregnancy
Induction: Rapid Sequence Versus Slow Neuroinduction
Combined Cesarean Delivery and Neurosurgery
670
671
673
674
675
675
676
678
CONTENTS
Intracranial Pressure and Regional Anesthesia
Postoperative Management
Anesthesia for Interventional Neurosurgical Procedures
References
678
678
679
679
41. Cerebral Venous Thrombosis
E.E. SHARPE AND J.J. PASTERNAK
Definition
Venous Anatomy
Incidence of Cerebral Venous Thrombosis
Risk Factors
Pathophysiology
Clinical Manifestations
Diagnostic Evaluation
Treatment
Anesthetic Management
Prognosis
Conclusion
References
681
681
681
683
684
685
687
688
689
690
690
690
42. Neurosurgical Anesthesia in Patients With
Coexisting Cardiac Disease
S. SRIVASTAVA AND A. KANNAUJIA
Introduction
Preoperative Evaluation
Risk Stratification
Perioperative Monitoring
Ischemic Heart Disease
Valvular Heart Disease
Tumors of the Heart
Congenital Heart Disease
Hypertension
Conclusion
References
693
694
694
695
695
697
699
700
700
701
701
43. Intraoperative Cardiopulmonary Resuscitation
R. GORJI AND M. SIDANI
Introduction
Incidence, Morbidity, and Mortality
Survival From Intraoperative Cardiac Arrest
Predictors
Cause of Intraoperative Cardiac Arrest
Cardiopulmonary Resuscitation Quality
Cardiac Arrest and Cardiopulmonary Resuscitation in
Neurosurgical Patients
Prognosis
Conclusion
References
703
703
704
704
705
706
706
709
709
710
44. Coexisting Diabetes Mellitus in Neurosurgical
Patients
N.B. PANDA, S. SAHU AND A. SWAIN
Introduction
Incidence of Diabetes Mellitus
Glycemic Indices
714
714
714
Modes of Glucose Measurement
Pathophysiology of Diabetes Mellitus
Cerebral Glucose Metabolism
Hyperglycemia and the Brain
Hyperglycemic Neuropathy
Diabetic Dysautonomia
Hypoglycemia and the Brain
Evidence of Glycemic Control in Important
Neurosurgical Subsets
Traumatic Brain Injury
Subarachnoid Hemorrhage
Cerebrovascular Accidents
Tumor Surgery
Spine Surgery
Blood Sugar Management in Perioperative
Period and Neurocritical Care
Intraoperative Management
Anesthetic Management
Postoperative Glycemic Management
Blood Sugar Control in Emergency
Neurosurgical Patient
Blood Sugar Control in Intensive Care Setup
Nutrition
Conclusions
Coexisting Hypertension in Neurosurgical Patients
Introduction
Physiology of Cerebral Circulation
Pathophysiology of Arterial Hypertension
Hypertension in Patients With Traumatic
Brain Injury
Perioperative Management
Preoperative Evaluation
Antihypertensive Drugs
Intraoperative Management
Monitoring
Induction of Anesthesia
Maintenance of Anesthesia
Recovery From Anesthesia
Postoperative Care
Neurocritical Care
Conclusion
References
xiii
714
715
715
715
716
716
716
717
717
717
717
718
718
718
719
719
719
720
720
720
720
721
721
721
722
723
724
724
725
725
725
725
726
726
726
727
727
727
45. Neuromuscular Disorders
P.U. BIDKAR AND M.V.S. SATYA PRAKASH
Introduction
Myasthenia Gravis
Myasthenic Crisis
Lambert–Eaton Myasthenic Syndrome
Guillain–Barré Syndrome
Periodic Paralysis
Myotonias
Muscular Dystrophies
Motor Neuron Diseases
Multiple Sclerosis
Parkinson’s Disease
Alzheimer’s Disease
Huntington’s Disease
References
734
735
743
747
748
753
755
758
759
760
761
763
764
765
xiv
CONTENTS
46. Neuromuscular Electrical Stimulation in
Critically Ill Patients
Conclusion
Clinical Pearls
References
N. LATRONICO, N. FAGONI AND M. GOBBO
Introduction
Neuromuscular Electrical Stimulation: Basic
Concepts and Practical Considerations
Neuromuscular Electrical Stimulation in the
Intensive Care Unit
Contraindications and Adverse Effects
Recommendations for the Use of Neuromuscular
Electrical Stimulation in the Intensive Care Unit
References
771
772
775
776
777
780
Crystalloid Fluids
Colloid Fluids
Which Fluid to Choose?
Conclusions
References
827
829
830
831
832
XIII
PAIN MANAGEMENT
K. JANGRA, V.K. GROVER AND H. BHAGAT
784
788
791
793
794
797
800
48 Anesthesia for Electroconvulsive Therapy
U. GRUNDMANN
Background
Technique of Electroconvulsive Therapy
Contraindications
Preprocedure Management
Anesthesia for Electroconvulsive Therapy
Side Effects
Special Conditions
Conclusion
References
50. Crystalloid and Colloid Fluids
R.G. HAHN
47. Neurological Patients for Nonneurosurgeries
Neurodegenerative Diseases
Demyelinating Disease
Neuromuscular Disease: Myasthenia Gravis
Epilepsy
Intracranial Tumors
Traumatic Brain Injury
References
824
825
825
805
805
806
806
806
809
809
810
810
XII
FLUIDS AND ELECTROLYTE
MANAGEMENT
51. Pain Management
Z. ALI, S. SINGH, N. HASSAN AND I. NAQASH
Postcraniotomy Pain
Introduction
Incidence
Anatomical and Physiological Basis of Pain
Following Craniotomy
Pain-Sensitive Structures of Cranium
Pathogenesis of Postcraniotomy Pain
Factors Affecting Postcraniotomy Pain
Classification and Assessment of
Postcraniotomy Pain
Preemption of Pain
Treatment of Acute Pain
Postcraniotomy Pain Management in the
Pediatric Population
Conclusion
Acute Pain Management After Spinal Surgery
Pathophysiology
Treatment Modalities for Acute Postoperative
Spinal Pain
Conclusion
References
836
836
836
836
838
838
838
839
840
840
843
843
843
844
844
848
849
XIV
49. Fluids and Electrolyte Management
BRAIN DEATH AND ETHICAL ISSUES
J.N. MONTEIRO
Introduction
Anatomy and Physiology
Pathophysiology
Definitions
Choice of Fluids
Hypertonic Fluids
Isotonic Fluids
Colloids
Hypotonic Fluids
Fluid Management
Commonly Encountered Fluid Abnormalities
815
815
817
817
818
818
820
820
821
822
823
52. Brain Death and Ethical Issues in Neuroanesthesia
Practice
M. RADHAKRISHNAN AND S. LALWANI
Part A: Brain Death
Introduction
Criteria for Diagnosing Death
Need for Brain Death Diagnosis
Rules Regulating Diagnosis of Brain Death
Criteria for Certifying Brain Stem Death
Pitfalls/Controversies
856
856
856
856
856
857
859
xv
CONTENTS
Conclusion
Appendix I
Part B: Ethical Issues in Neuroanesthesia Practice
Introduction
Ethical Issues in Clinical Care
Ethical Issues Related to Research
Ethical Issues Related to Team Work
Ethical Issues Related to Training
Ethical Issues Related to Innovative Neurosurgery
Conclusion
References
861
861
863
863
863
867
868
868
869
869
869
53. Organ Donation
M.J. SOUTER
Introduction
Conclusion
References
871
876
876
901
902
903
57. Stem Cell Therapy
S. SHARMA AND R. AGGARWAL
Hypothesis of Stem Cell Research
Stem Cell
Historical Background
Types of Stem Cells
Sources of Stem Cells
Mesenchymal Stem Cells
Stem Cells in Neurological Diseases
Mode of Action of Stem Cell Therapy
Ethical Issues
Recent Advances
References
907
907
908
908
908
909
909
910
910
911
911
58. Pharmacogenomics
XV
Y.N. MARTIN AND W.T. NICHOLSON
EVIDENCE-BASED PRACTICE
54. Evidence-Based Practice of Neuroanesthesia
I. KAPOOR AND H. PRABHAKAR
Introduction
Evidence-Based Practice and Neuroanesthesia
Evidence and the Brain Trauma Foundation Guidelines
Unresolved Issues in the Practice of Neuroanesthesia
Conclusion
Clinical Pearls
References
Brain Monitoring
New Assays for Creutzfeldt–Jakob Disease
References
881
883
887
887
887
889
889
55. Translational Research
M. IDA AND M. KAWAGUCHI
Introduction
Basic Genetic Principles
Basic Pharmacologic Principles
Anesthesia Contribution to the History
of Pharmacogenomics
Pharmacogenomics: Current Application
to Clinical Anesthesia
Conclusion
References
913
914
914
915
916
921
922
XVII
STERILIZATION TECHNIQUES
59. Sterilization and Disinfection
Introduction
Definition
In Neuroanesthesia
Why Not Lead to Clinical
To Be a Successful Translation
Conclusion
References
891
891
893
893
894
894
894
XVI
RECENT ADVANCES
56. Recent Advances in Neuroanesthesiology
T.L. WELCH AND J.J. PASTERNAK
Introduction
Endovascular Treatment of Stroke and Perioperative Stroke
Indications for Deep Brain Stimulation
Anesthetic Neurotoxicity
Pre- and Postconditioning
897
897
898
899
900
S. MOHAPATRA
Background
Recommendation of Preferred Methods for
Various Medical Devices
Recommendation for the Cleaning and
Decontamination of Environmental Surfaces
Recommendation for Blood Spill on the Surface
Cleaning and Disinfection of Medical Instruments
Cleaning and Reprocessing of Patient
Care Equipment
Reprocessing of Respiratory Apparatus and
Endoscopes
Reprocessing of Endoscopes
Specific Issues
Special Precaution for Inactivation of
Creutzfeldt–Jakob Disease
Health Care–Associated Infections
Infections in Operating Rooms and Intensive
Care Units
Conclusion
References
930
931
931
932
933
933
935
936
938
939
940
940
943
943
xvi
CONTENTS
60. Universal Precautions in the Intensive
Care Unit
A.YU LUBNIN AND K.A. POPUGAEV
Introduction
Prophylactics of Health Care–Associated Infections
in the Intensive Care Unit
Early Diagnosis of Pathogens and Infection
Complications in the Intensive Care Unit
Rational Antibiotic Therapy
Systemic Approach
Conclusion
References
945
946
947
947
948
948
948
XVIII
PALLIATIVE CARE
61. Palliative Care to Neurological and
Neurosurgical Patients
S. BHATNAGAR AND S.J. BHARTI
Introduction
References
953
961
62. Quality of Life and Health-Related Issues
L. VENKATRAGHAVAN AND S. BHARADWAJ
Introduction
Quality of Life and Health-Related Quality of Life
Utility of Health-Related Quality of Life
Tools for Measuring Health-Related Quality of Life
Uses of Measuring Health-Related Quality of Life
Health-Related Quality of Life in Relation to
Neurosurgical/Neurological Conditions
Conclusion
References
963
963
964
964
965
965
970
970
XIX
BIOSTATISTICS
63. Biostatistics
M. KALAIVANI, S. AMUDHAN, A.D. UPADHYAY AND V.K. KAMAL
Introduction to Biostatistics
Definition of Statistics
Biostatistics and Its Applications
976
976
976
Uses of Statistical Methods in Medical Sciences
Some Basic Statistical Concepts
Population and Sample
Scale of Measurements
Constant
Variables
Parameter and Statistic
Ratio, Proportion, and Rate
Statistical Inference
Estimation
Hypothesis Testing
Steps in Hypothesis Testing or Testing the
Statistical Significance
Defining the Null and Alternative Hypotheses
Calculating the Test Statistic
Obtaining, Using, and Interpreting the p-Value
Errors in Hypothesis Testing
The Possible Mistakes We Can Make
Other Important Concepts That Are Essential
in Statistical Inference
Parametric and Nonparametric Statistical Methods
Basic Principles of Statistics
Probability Distributions
Study Design
Sample Size
Data Collection and Preparing Data for Analysis
Analysis and Presentation of Data
Summarizing Data
Comparing Groups: Continuous Data
Comparing Groups: Categorical Data
Comparing Groups: Time to Event Data
Relation Between Two Continuous Variables
Multivariable Analysis
Conclusion
References
Index
976
976
977
977
977
977
978
978
979
979
979
979
980
980
980
980
980
981
981
981
982
982
985
987
989
989
989
991
991
992
994
995
995
997
List of Contributors
M. Abraham Max Hospital Panchsheel, New Delhi, India
R. Aggarwal All India Institute of Medical Sciences, New
Delhi, India
D. Gupta Sanjay Gandhi Post Graduate Institute of Medical
Sciences, Lucknow, India
N. Gupta Indraprastha Apollo Hospital, New Delhi, India
Z. Ali SKIMS, Srinagar, India
R.G. Hahn Södertälje Hospital, Södertälje, Sweden
S. Amudhan NIMHANS, Bengaluru, India
N. Hassan Government Gousia Hospital, Srinagar, India
M.-A. Babi Duke University, Durham, NC, United States
M. Ida Nara Medical University, Kashihara, Japan
S. Bansal National Institute of Mental Health and
NeuroSciences (NIMHANS), Bangalore, India
M.L. James Duke University, Durham, NC, United States
S.D. Bergese Ohio State University, Columbus, OH,
United States
H. Bhagat Postgraduate Institute of Medical Education
and Research, Chandigarh, India
S. Bharadwaj NIMHANS, Bangalore, India
S.J. Bharti AIIMS, New Delhi, India
K. Jangra Postgraduate Institute of Medical Education and
Research, Chandigarh, India
M. Kalaivani AIIMS, New Delhi, India
V.K. Kamal AIIMS, New Delhi, India
A. Kannaujia Sanjay Gandhi Post Graduate Institute of
Medical Sciences, Lucknow, India
S. Bhatnagar AIIMS, New Delhi, India
I. Kapoor All India Institute of Medical Sciences, New
Delhi, India
P.U. Bidkar JIPMER, Puducherry, India
M. Kawaguchi Nara Medical University, Kashihara, Japan
F. Bilotta Sapienza University of Rome, Rome, Italy
A.K. Khanna Cleveland Clinic Foundation, Cleveland, OH,
United States
P.K. Bithal AIIMS, New Delhi, India
V. Bonhomme CHR Citadelle, Liege, Belgium
A. Borozdina I.M. Sechenov First Moscow Medical
University, Moscow, Russia
A. Defresne CHR Citadelle, Liege, Belgium
S.K. Dube All India Institute of Medical Sciences, New
Delhi, India
M. Echeverría Centro Médico Docente Paraíso, Maracaibo,
Venezuela
H. El Beheiry University of Toronto, Toronto, ON, Canada;
Trillium Health Partners, Toronto, ON, Canada
S. Erb University Hospital Basel, Basel, Switzerland
N. Fàbregas Hospital Clinic Universitari, Barcelona, Spain
N. Fagoni University of Brescia, Brescia, Italy
E. Farag Cleveland Clinic Foundation, Cleveland, OH,
United States
S.A. Khan Duke-NUS Medical School, Singapore,
Singapore
K.M. Kla Vanderbilt University Medical Center, Nashville,
TN, United States
V. Krishnamoorthy University of Washington, Seattle, WA,
United States
D.K. Kulkarni Nizam’s Institute of Medical Sciences,
Hyderabad, India
S. Lalwani All India Institute of Medical Sciences,
New Delhi, India
N. Latronico University of Brescia, Brescia, Italy
L.A. Lee Kadlec Regional Medical Center, Richland, WA,
United States
A. Lele University of Washington, Seattle, WA,
United States
J. Fiorda-Diaz Ohio State University, Columbus, OH,
United States
A.Yu Lubnin Neurocritical Care of Burdenko Research
Neurosurgical Institute, Ministry of Health, Moscow,
Russia
P. Ganjoo GB Pant Hospital, New Delhi, India
A. Luthra PGIMER, Chandigarh, India
M. Gobbo University of Brescia, Brescia, Italy
C. Mahajan AIIMS, New Delhi, India
R. Gorji Upstate Medical University, Syracuse, NY,
United States
S. Mahajan Postgraduate Institute of Medical Education
and Research, Chandigarh, India
V.K. Grover Postgraduate Institute of Medical Education
and Research, Chandigarh, India
P.H. Manninen Toronto Western Hospital, Toronto, ON,
Canada
U. Grundmann Saarland University Medical Center,
Homburg/Saar, Germany
M. Marda Max Hospital Panchsheel, New Delhi, India
R. Mariappan Christian Medical College, Vellore, India
xvii
xviii
LIST OF CONTRIBUTORS
A. Marson University of Liverpool, Liverpool, United
Kingdom
K. Sandhu Max Superspeciality Hospital, New Delhi, India
Y.N. Martin Mayo Clinic, Rochester, MN, United States
M. Sethuraman Sree Chitra Tirunal Institute for Medical
Sciences and Technology, Trivandrum, India
R. Mitra Care Hospital, Bhubhaneswar, India
S. Mohapatra AIIMS, New Delhi, India
S. Moningi Nizam’s Institute of Medical Sciences,
Hyderabad, India
J.N. Monteiro P.D. Hinduja Hospital and Medical Research
Centre, Mumbai, India
M.V.S. Satya Prakash JIPMER, Puducherry, India
S. Sharma All India Institute of Medical Sciences, New
Delhi, India
E.E. Sharpe Mayo Clinic College of Medicine, Rochester,
MN, United States
I. Naqash SKIMS, Srinagar, India
M. Sidani Upstate Medical University, Syracuse, NY,
United States
W.T. Nicholson Mayo Clinic, Rochester, MN, United States
V. Singhal Medanta (The Medicity), Gurgaon, India
C. Oetliker University Hospital Basel, Basel, Switzerland
G. Singh Christian Medical College, Vellore, India
D. Padmaja Nizam’s Institute of Medical Sciences,
Hyderabad, India
G.P. Singh AIIMS, New Delhi, India
N.B. Panda Post Graduate Institute of Medical Education
and Research, Chandigarh, India
M.J. Souter University of Washington, Seattle, WA, United
States
M. Panebianco University of Liverpool, Liverpool, United
Kingdom
K. Sriganesh NIMHANS, Bangalore, India
J.J. Pasternak Mayo Clinic College of Medicine, Rochester,
MN, United States
S. Singh SKIMS, Srinagar, India
S. Srivastava Sanjay Gandhi Post Graduate Institute of
Medical Sciences, Lucknow, India
L.A. Steiner University Hospital Basel, Basel, Switzerland
K.A. Popugaev Federal Medical-Biological Agency,
Ministry of Health, Moscow, Russia
N. Stoicea Ohio State University, Columbus, OH, United
States
L. Porcella Spedali Civili University Hospital, Brescia, Italy
A. Swain Tata Main Hospital, Jamshedpur, India
H. Prabhakar All India Institute of Medical Sciences,
New Delhi, India
M.S. Tandon University of Delhi, New Delhi, India
F. Rabai University of Florida, Gainesville, FL, United
States
M. Radhakrishnan National Institute of Mental Health
and NeuroSciences, Bengaluru, India
S. Rajan Cleveland Clinic, Cleveland, OH, United States
R. Ramani University of Florida, Gainesville, FL,
United States
V.J. Ramesh National Institute of Mental Health and
NeuroSciences, Bengaluru, India
S. Rao Yale New Haven Hospital, New Haven, CT,
United States
G.P. Rath All India Institute of Medical Sciences (AIIMS),
New Delhi, India
S. Sahu Tata Main Hospital, Jamshedpur, India
D. Saigal University of Delhi, New Delhi, India
L. Salvador Consorcio Hospital General Universitario
de Valencia, Valencia, Spain
S.S. Thota Upstate Medical University, State University of
New York, Syracuse, NY, United States
D.E. Traul Cleveland Clinic, Cleveland, OH, United States
S. Tripathy All India Institute of Medical Sciences
Bhubaneswar, Bhubaneswar, India
G.S. Umamaheswara Rao National Institute of Mental
Health and NeuroSciences (NIMHANS), Bangalore, India
A.D. Upadhyay AIIMS, New Delhi, India
L. Venkatraghavan University of Toronto, Toronto, ON,
Canada
B. Vinay Gulf Medical University, Ajman, United Arab
Emirates
T.L. Welch Mayo Clinic College of Medicine, Rochester,
MN, United States
T.Y. Yeoh
Toronto Western Hospital, Toronto, ON, Canada
J. Žurek University Hospital Brno, Brno, Czech Republic
Foreword
There has been substantial flux in the field of neuroanesthesia over the past two decades. This followed what
could be viewed as a relatively quiescent and narrowly focused period in neuroanesthesia. During the latter period
much of the focus was on the roles of hypotension in aneurysm surgery, hyperventilation for head injury, anesthetics
as cerebral protectants, and endless debates about intravenous versus inhaled anesthetics. More recently the purview
of neuroanesthesia broadened substantially partly reflecting the huge expansion in the way patients with neurological diseases are managed. Patients are cared for not only in the traditional operating theater and intensive care unit
but also in more complex ways inside and outside the operating theater. Examples include endovascular treatment
of aneurysms, magnetic resonance imaging (MRI)- and computed tomography (CT)-guided surgery, minimally invasive approaches such as deep brain stimulation (DBS), the growth in neurological monitoring from the awake patient
to complex electrophysiology, and the ever increasingly aggressive spine reconstructions. The neuroanesthesiologist
of today is not only a traveler going to different parts of the hospital but needs to be an expert in patient management
in all the newer scenarios. Furthermore, this expansion of the repertoire requires greater refinement in our intimate
knowledge of how drugs and techniques may enhance or adversely affect the nuanced neurosurgical outcomes.
Given the above changes in practice, the novice and experienced neuroanesthesia practitioners now, more than
ever, need an authoritative text not just full of “book knowledge” but written by those who on a daily basis meld
the academic with the practical. To his credit, Hemanshu Prabhakar has brought together an accomplished group of
international experts to contribute to this excellent volume. Their writing is authoritative and up to date while being
practical and easy to understand. There is no doubt that this book is a very useful contribution to the modern practice
of neuroanesthesia.
Adrian W. Gelb
Distinguished Professor
Department of Anesthesia and Perioperative Care
University of California
San Francisco
xix
This page intentionally left blank
Preface
Neuroanesthesia is growing fast as a superspecialty as more and more research is being conducted to improve
the practice. The focus is now not restricted to the bench but has also extended to the bedside. There is a need to have
a volume that provides a comprehensive view of various topics and issues related to neuroanesthesia. This book
provides easy understanding of anesthesia related to neurological sciences. This book will be useful for any medical practitioner associated with neurosurgical and allied branches such as neurology and neuroradiology. This book
also caters to the needs of all those anesthetists who practice neuroanesthesia but do not have a formal training in
it. It will provide a quick and easy access to understand neuroanesthesia. This book will provide an insight into all
possible aspects of anesthetic management of neurosurgical and neurologic patients. This book has been written
mainly for the residents and students appearing for examination and anesthetists practicing neuroanesthesia. This
book includes the basic sciences such as anatomy, physiology, and pharmacology related to brain and spinal cord.
This book also provides an understanding of related issues such as palliative care, evidence-based practice of neuroanesthesia, sterilization techniques, and ethical issues.
This book covers all topics related to neuroanesthesia and provides complete knowledge about brain and spinal
cord. The book includes chapters related to allied specialties such as critical care, neurology, and neuroradiology.
This book also contains a section on biostatistics, which would be extremely useful to residents and trainees who
have to submit dissertation or thesis during their course.
This book contains pieces of information that have been brought together, which may have otherwise been available in different books.
I am grateful to all my authors across the globe, from as many as 14 different countries. The knowledge and information shared by the authors through different chapters is the representation of the global practice of neuroanesthesia and not limited to geographical boundaries. I sincerely hope this endeavor will improve our knowledge in the
management of neurologically compromised patients and bring about an improved patient care.
Hemanshu Prabhakar
xxi
This page intentionally left blank
Acknowledgments
I wish to acknowledge the support of the administration of the All India Institute of Medical Sciences (AIIMS),
New Delhi, in allowing me to conduct this academic task.
Words are not enough to express my gratitude for the constant support and encouragement from Prof. P.K. Bithal
(Former Head of Neuroanesthesiology and Critical Care, AIIMS, New Delhi). I thank the faculty and staff of the
department of Neuroanesthesiology and Critical Care, for their support.
Special thanks are due to the production team at Elsevier.
xxiii
This page intentionally left blank
Introduction and Brief History of
Neuroanesthesia
W.S. Jellish
Loyola University Medical Center, Maywood, IL, United States
Neuroanesthesia has evolved as a subspecialty of anesthesiology and has continued to evolve in association with
further surgical advancements such as minimally invasive techniques and three-dimensional imaging using neuronavigation. Neuroradiology has also advanced with invasive neurovascular procedures, once done in the operating
rooms, now performed in the neurointerventional radiology suite with the support of the anesthesiologists.
Early neuroanesthesia was performed as a method to support the practice of trephination. It was uncertain
whether brain surgery had been performed during this trephination or if it was part of a religious or social ritual
(Fig. 1). Some were obviously related to injury, but the presence of multiple defects in the absence of other apparent
injuries in both young and old suggested a possible therapeutic purpose. A popular aspect was that these openings
were used to alleviate pain or allow the escape of evil spirits and humors or to drain pus or reduce inflammation. No
matter what the reason for the trephination, skulls with these types of defects have been found all over the world.
There are several documented examples, which point out that these skull defects were produced as part of a neurosurgical procedure. The Edwin Smith Papyrus is one of the earliest written records of surgical practice (Fig. 2). The
text may well represent the first neurosurgical practitioner’s manual as it describes 48 cases that consist of 15 head
injuries, 12 facial wounds and fractures, and 7 vertebral injuries. Several other written works produced around 400
BC have been linked to Hippocrates. One of the texts on injuries of the head describes trephination for skull fractures, epilepsy, blindness, and headaches. The practitioner was advised to avoid suture lines and the temporal areas
because of fear of damaging the anatomy that would lead to contralateral convulsions.1 It was also noted that the
inner table should be preserved to protect the dura and this bone fragment would later be extracted by suppuration.
Despite the advanced neurosurgical and neuroanatomical knowledge for the times in Greece and Rome, use and
understanding of neuroanesthesia did not appear to be much different from those of fellow practitioners elsewhere
in the world. In the prehistoric period, anesthesia was probably done by chewing or locally applying a mixture of
coca and yucca.2 Daturas had also been used with its anesthetic effect thought to be produced by its contents of scopolamine, hyoscyamine, and atropine. In early cultures, wine making was highly developed. In Egypt the soporific
effects of alcohol had been well documented in hieroglyphic writings. It is considered likely that some analgesia and
amnesic effects were derived from this source. Sometimes just compression of the carotid artery was used to induce
unconsciousness.3
Progress in neurosurgery slowed considerable during the Middle Ages. The Roman Catholic Church became very
influential in medical care and monasteries were the centers for science and knowledge. With the edict of AD 1163
entitled, “Ecclesia abhoret a sanguine,” there were restrictions placed on the use of human bodies for anatomical
studies, and progress in the field of surgery was almost halted. At the beginning of the 10th century, Rhazes compiled the Liber Continens, a collection of all literature belonging to the Arab world including medicine, philosophy,
religion, mathematics, and astronomy. Among the several important observations he made with this work, one was
particularly remarkable. He wrote that the pressure on the brain, rather than the presence of the skull fracture itself,
was more important in determining the outcome after head injury.4 It was not until later in the 10th century with the
establishment of a medical school in Salerno, Italy, that this and other medical principles were brought forward to
revive European medicine.
Few references were made on the use of anesthesia, perhaps because pain was mentioned so many times in religious teachings and was felt to be a noble state that served God’s purpose. When anesthesia was attempted, opium,
hyoscyamine, and sometimes wine were used as agents to alleviate pain.5 On occasion Cannabis indica and henbane
were used, along with a sponge containing opium, marberry, water hemlock, and ivy which was boiled and then
applied to the patient’s nose during the surgery.6 It was thought that wine was added to the sponge during boiling
to enhance its sedative power. The physicians of Myddavi, herbalist from Wales, further advised that to improve
xxv
xxvi
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
FIGURE 1 The trephined skulls discovered in paracus, Peru (≈500 BC), and obsidian blades. The surgical holes are covered with roles of cotton
dressing (A). The trepanation had been performed using obsidian blade (B).
FIGURE 2 The Edwin Smith Papyrus. The terms of brain, as original term “iesh,” pointed by an arrow (and depicted below) can be seen. From
James Henry Breasted. The Edwin Smith Surgical Papyrus. Chicago: University Chicago Press; 1930 (reproduced from Eric R Kandel, James H Schwartz,
Thomas M Jessell, editors Principles of Neural Science. 4th ed. New York: McGraw-Hill; 2000).
anesthetic efficiency, when you prepare to operate on a patient, direct them to avoid sleep as long as possible then let
some of the potion be poured into the nostril of the patient who will fall asleep without fail.7
The first account of inducing anesthesia and reversing sedation was attempted by two Hindu brothers who performed a craniotomy in AD 927 to remove an unidentified tumor from the brain of the King of Dhar. They induced
anesthesia with a drug called samohine and reversed the anesthetic effects by pouring an onion compounded with
vinegar into the subject’s mouth.7 Numerous other concoctions both oral and topical were administered to patients
for better outcomes. Some combined puppies boiled in an oil of lilies and earthworms and prepared in turpentine
of Venice.2 Other mixtures used were rose oil, egg yolk, and turpentine, which were either heated in cold weather or
cooled in warm weather to maintain temperature. This was done to produce optimal wound healing.
The major improvements and overall advancement in anesthesia for neurosurgery occurred in the latter half of
the 18th century. The discovery of carbon dioxide, hydrogen, and nitrogen along with experiments by Priestly and
others using several gases including oxygen and nitrous oxide created interest in the use of these agents to support
patients who were sedated or anesthetized for procedures.8 However, the reluctance of surgeons to perform cranial
operations slowed the implementation of anesthesia for neurosurgery. Nevertheless, the accumulation of knowledge
of functional neuroanatomy, establishment of concepts of asepsis, and the discovery of general anesthesia all moved
the process of neurosurgery accompanied by anesthesia forward.
William Macewen was the first neurosurgeon to excise a brain tumor under endotracheal intubation. He was the
first to show the necessity of controlling the airway and ventilation during craniotomy. At the beginning of the 20th
century there was a great deal of controversy as to what was the best inhalational anesthetic, chloroform, or either.
Victor Horsley performed a series of experiments in animals in 1883–85 and concluded that, although ether was the
safer drug, it was not to be recommended in favor of chloroform because it produced a rise in blood pressure and an
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
xxvii
FIGURE 3 The Vernon Harcourt vaporizer arranged with a cylinder of compressed oxygen.
increase in blood viscosity with a potential for hemorrhage.9 He also noted a propensity for postoperative vomiting
and concluded it should not be used for neurosurgery. Also, since morphine constricted blood flow, he suggested
that a combination of morphine and chloroform be used. He subsequently abandoned morphine use because of its
recognized depressant effect on respiration.9
Death related to the administration of chloroform was not uncommon, and several commissions were set up to
study the effects of the drug. In 1901, The British Medical Association appointed a special chloroform committee to
study its use. It was known that approximately 2% chloroform vapor in air was sufficient to induce anesthesia with
much less required for maintenance. Some believed that the concentration should be strictly controlled with a vaporizer, while others thought it could be administered strictly by sprinkling onto a cloth. Horsley was of the opinion
that the concentration should be controlled and used a vaporizer designed by Vernon Harcourt, which delivered
chloroform at a 2% maximum (Fig. 3). In patients undergoing craniotomy, Horsley felt that chloroform administration should be reduced to 0.5% once the bone flap was removed.10 Determination of concentration was particularly
important in patients with raised intracranial pressure (ICP) since higher concentrations in these patients could be
fatal.
Horsley also contributed to neuroscience and neurosurgery by his support and defense of surgery done on animals for scientific research and to advance clinical knowledge. His work and testimony against the Anti-Vivisection
Society helped to defend the use of animal models to advance neurosurgery.11
Around the same time, the use of local anesthesia began to gain prominence. Cocaine had been formally discovered in 1860 and was introduced in surgery in 1884. Procaine was first synthesized in 1905 and immediately became
commonplace among surgical anesthesia. Most neurosurgeons used local anesthetics selectively. However, in 1913
deMartel popularized its use, and it became common practice for use in all craniotomies. By 1917, Harvey Cushing,
considered the founder of neurosurgery, recommended the use of local anesthesia for all neurosurgical cases.7
Besides being one of the leading neurosurgeons of the early 20th century, Cushing introduced numerous new
methods of anesthesiology. His first contracts in providing anesthesia were not entirely successful. He had several
notable patient deaths while providing ether for procedural anesthesia. In all instances, he was told the deaths
occurred due to the patient’s condition, but he remained unconvinced that adverse reactions to anesthesia were
due only to the condition of the patients. At the coaxing of Dr. F. B. Harrington, Cushing and fellow student Amory
Codman tried to determine who gave the best anesthetics. To make the decision objective, they documented their
anesthesia in the form of “ether charts” (Fig. 4).12 Their most important parameters measured were the pulse rate,
xxviii
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
FIGURE 4 An example of the ether charts made by Cushing.
breathing, and temperature of the patients. According to Cushing, “a perfect anesthetic was supposed to be one
in which the patient was sufficiently conscious to respond when left in the ward with the nurse and did not subsequently vomit.” The use of these either charts was a major step toward improvement in what had been a casual
administration of a very dangerous drug.
In 1900, Cushing began an extended tour of Europe, and while in Bern, he recognized the association between
raised ICP and systemic arterial hypertension.13 While in Padua, Italy, at the Ospidale di St. Matteo, he learned the
use of Riva-Rocci’s method of blood pressure measurement (Fig. 5). After returning to Baltimore, he adopted this
method of blood pressure measurement into clinical practice. He gave a lecture in 1903 titled, “Considerations of
Blood Pressure,” only to have a committee of the Harvard Medical School state that, “the skilled finger was of much
greater value clinically for determination of the state of circulation than any pneumatic instrument, and the work
should be put aside, as of no significance.” Even though blood pressure monitoring was not supported by thought
leaders at that time, Cushing still supported the recording of blood pressure during surgery. He also attached great
importance to continuous auscultation of the heart and lungs, a technique he learned from his anesthesiologist,
Dr. S. Griffith Davis.14 The precordial auscultation device used a transmitter of the phonendoscope secured by adhesive strips over the precordium and was connected with a long tube to the anesthesiologist’s ear. The receiver was
held in place by a device similar to a telephone operator’s headgear.15
Cushing remained skeptical about general anesthesia for neurosurgery. Mortality was still high and many of the
anesthetics were performed by students. He began to experiment on work started by Halsted with block anesthesia
using circumferential cocaine infiltration.16 He popularized the use of several local anesthetic techniques and coined
the term “regional anesthesia.” From his first work dealing with regional anesthesia, it was noted that the purpose of
administering it was to avoid side effects in patients with full stomachs and to ensure better cardiovascular stability
in elderly patients.
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
xxix
FIGURE 5 Dr. Harvey Cushing’s sketch of Riva-Rocci’s blood pressure device.
Dr. Fedor Krause, the founder of German neurosurgery, was exposed to combination morphine–chloroform anesthesia but was not convinced of its worth for neurosurgery. He felt that controlled hypotension produced by higher
concentrations of chloroform alone were beneficial to reduce bleeding.17 He also noted that sudden death could
occur in patients with intracranial tumors who stopped breathing. He used the Roth-Drager oxygen–chloroform
apparatus which allowed high concentrations of oxygen to be administered. He also noted the brain was insensitive
to pain, and only very light planes of anesthesia were needed to perform these surgeries.18 He concluded a good
neurosurgical outcome required a rapid aseptic technique, minimal blood loss, normothermia, and general narcosis.
During the early 20th century, new delivery systems for anesthesia were being developed. The Junker bottle used
a hand bellows to blow air through a vaporizer and the Ombrédanne ether inhaler could be used in the prone position (Fig. 6). Airway management became less supportive and passive and more active with endotracheal intubation
and insufflation of air to maintain oxygenation without ventilation. By 1930, endotracheal anesthesia was recommended for neurosurgery. Inhalational anesthetics such as trichloroethylene with nitrous oxide were developed
as a neuroanesthetic technique,18 while other physicians such as Hershenson used low concentrations of closed
circuit cyclopropane and reported this method in 1942.19 Volwiler and Tabern developed thiopental in 1930 and was
introduced into clinical practice 4 years later.20 Halothane was synthesized in 1956 and introduced into practice that
same year.21 Though popular, its propensity to increase ICP and brain size made it a concern to anesthesiologists
and neurosurgeons alike. In 1932, most fluids were given rectally along with a wide variety of anesthetic techniques
including ether-based anesthesia, rectal ether in oil, ethylene, and nitrous oxide.
Certain large centers began to publish their outcomes for neurosurgical procedures and many included a description of the anesthetic techniques used. The Montreal Neurologic Institute showed that of 1000 cases, 700 were performed under general anesthesia and 300 with local anesthetics.22 The major concerns were still airway management
and fluid replacement. A wide variety of agents were used. However, by 1949 and the early 1950s, pentothal was the
induction agent of choice for oral intubation. Throat packing and the use of a nonrebreathing valve to prevent the
buildup of carbon dioxide was also common.
After World War II, great advances in neuroanesthetic techniques were brought on by the development of new
anesthetic agents and advanced knowledge of neurophysiology and pharmacology. Lundy in 1942 noted that in
the presence of hypoventilation it was difficult to reduce ICP unless the patient was artificially ventilated.23 The
effects of hypoventilation on intracranial and systemic dynamics were further elucidated by Keaty and Schmidt
(Fig. 7). They also described methods to measure cerebral blood flow using inhaled N2O.24 Another group used the
injection of intraarterial krypton to measure blood flow in the brain and speed of washout.25 The measurement of
ICP had been discovered by Cannon in 1901 but continuous ICP measurement was not described until 1960.26 The
link between CO2, O2 tension and cerebral blood flow was also developed at this time. Dr. Thomas Langfitt further
xxx
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
FIGURE 6 (A) Ombrédanne inhaler. (B) Ombrédanne inhaler in use.
5
Arterial
N2O Conc Vol%
4
3
A-V
Venous
2
1
1
2
3
4
5
6
Time (minutes)
7
8
9
10
FIGURE 7 Kety–Schmidt method of arterial and venous nitrous oxide concentration for calculating cerebral blood flow.
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
xxxi
defined intracranial dynamics based on previous work and developed the pressure volume curves along with the
concept of intracranial compliance.27
Three major anesthesia groups contributed to the development of the specialty, the Glascow Group, Pennsylvania
Group, and the Mayo Clinic Group. All the three centers were considered think tanks in the development of neuroanesthetic principles and techniques to help improve intracranial surgery. Most of the principles for neuroanesthesia
have been developed over the last 70 years. The term, “neuroanesthesia” was coined by John Michenfelder from the
Mayo Clinic.28
In the 1960s, researchers at the University of Pennsylvania studied the cerebral effects of nitrous oxide, cyclopropane, halothane, enflurane, hypotension, and hyperventilation.29–31 These studies formed many of the basics for neuroanesthesia principles today. At the same time, researchers at the University of Glascow studied anesthetic effects
on intracranial dynamics. This group was one of the first to confirm that anesthetic agents did indeed exert a measurable effect on cerebral blood flow and metabolism in patients with intracranial mass lesions.32,33 It was some of these
studies that were used to justify the abandonment of the use of halothane because of its vasodilating properties, especially in patients with mass lesions in the brain. At the Mayo Clinic the only emphasis of research by Michenfelder
was of providing cerebral protection, especially with hypothermia during neurovascular procedures. These same
three centers continued to expand their influence in neuroanesthesia, and at the University of Pennsylvania the role
of the neuroanesthesiologist was expanded to the intensive care unit.
In the 1980s, research continued on glucose control and again on the effects of modest hypothermia. A wide range
of drugs emerged as possible cerebral protective agents. A much clearer understanding of pathways involved in
cell damage was also achieved. Research on brain trauma and survival also became important because of the effects
of war on traumatic brain injury. Despite much of this research, the morbidity and mortality from subarachnoid
hemorrhage, stroke, trauma, and neoplastic lesions have remained largely unchanged in the past several decades.
Nevertheless, neuroanesthesia practice has appreciably changed over the past few decades.
Hyperventilation has long been known to reduce ICP. At the beginning of the 1990s it was widely held that this
had therapeutic value for intracranial procedures. However, with the use of oximetric pulmonary artery catheters,
investigators have been able to do retrograde cannulation of the jugular vein up to the bulb to examine venous Hb
saturation in response to therapeutic hyperventilation in head trauma patients. In some patients, this hyperventilation resulted in increased hypoxia that has resulted in abandonment of hyperventilation unless surgical conditions
dictate. However, other recent studies have demonstrated that hyperventilation, especially for supratentorial brain
tumors, was associated with reduced ICP and a 45% reduction in brain bulk, once again demonstrating its worth,
especially with supratentorial surgery.34
The early 1990s also demonstrated a surge of new anesthetic techniques and drugs. Both desflurane and sevoflurane were introduced and were found to have cerebral metabolic properties similar to isoflurane. However, some
questions still remain regarding desflurane causing an insidious increase in ICP. There are also questions concerning
the metabolism of sevoflurane and possible renal toxicity, especially with the effect of anticonvulsants on hepatic
function. Since both drugs have been accepted and widely used as neuroanesthetics, these concerns do not seem
to hold major importance. Remifentanil was also introduced in the 1990s and was found to be essentially identical
to other opioids with respect to µ-opioid agonist-mediated events. It does have a much more rapid and predictable
emergence compared to other opioid-based techniques. However, the drug produces hypertension and tachycardia
with increased sympathetic activity during emergence which could be especially problematic with large tumor resections where hemodynamic stability during emergence to prevent bleeding is imperative.
The administration of proper IV fluids and correct fluid replacement therapy was better developed in the 1990s.
There was a widespread acceptance that glucose-containing solutions were not essential in patients undergoing
neurosurgical procedures. Perioperative glycemic control is one of the important topics that have been investigated
in the 1980s and 1990s. Hyperglycemia in ischemic conditions has been proven to be detrimental, and strict control
of plasma glucose has been shown to produce better outcomes in critically ill patients. Many studies have demonstrated that plasma glucose levels are well maintained at close to normal ranges with nonglucose-containing solutions, while patients who received glucose had high plasma levels which fluctuated dramatically during their care.
Therefore, routine use of glucose-containing solutions should be avoided during neurosurgery.
The use of crystalloid solutions has also been altered by studies from the last 20 years. In the 1980s it remained a
standard practice to dehydrate patients with intracranial pathology under the assumption that brain volume would
be decreased. This was often performed at the expense of stable hemodynamics and cerebral perfusion pressure.
However, extensive studies have demonstrated that fluid restriction for neurosurgical procedures may be detrimental. In addition, many large trials evaluating colloid versus crystalloid solutions, especially with trauma-related injury,
showed that saline resuscitation may be of greater benefit than treatment with albumin. In fact, studies demonstrated
xxxii
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
a detrimental effect of albumin treatment, especially with the relative risk of death within 24 months.35 This has led
to a recommendation that colloids not be used for resuscitation in patients with brain injury.
The early 1990s also provided consistent evidence that small changes in brain temperature could have a major
impact on outcome from ischemic or traumatic brain injury.36 These changes did not require overt cooling or the use
of extracorporeal perfusions but were well within the range of routine manipulation by anesthesiologists. The practice of the use of mild hypothermia increased, but there were other warnings about inadvertent hypothermia from
other patient populations, which caused concerns. In the late 1990s, a multinational consortium of investigators was
formed to examine the risks and benefits of mild hypothermia in patients undergoing aneurysm clipping and was
inconclusive as to the benefit of hypothermia on survival and outcome after a neurologic event producing ischemia.
Based on clinical data, mild hypothermia may still have beneficial effects in patients with good grade subarachnoid
hemorrhage (SAH).37 The latest American Heart Association and American Stroke Association guidelines for the
management of aneurysmal SAH recommend induced hypothermia as a reasonable option in selected cases.38
At the clinical level, no progress has been made in pharmacologic neuroprotection. Despite successful experimental studies, by far no anesthetic technique has been convincingly shown to provide profound neuroprotection in
humans. Barbiturates remain the gold standard, although clinical evidence of efficacy from this class of compounds
is suspected on methodologic grounds. In a post hoc analysis of IHAST (intraoperative hypothermia for aneurysm
surgery trial) data administration of thiopentanil or etomidate was not found to have a demonstrable effect on
postoperative neurological outcomes in patients undergoing temporary clipping.39 Use of etomidate has waned as
a neuroprotective agent as a result of absence of clinical evidence of benefit in both clinical and laboratory studies.
Some of the most exciting and important clinical advances in neuroanesthesia have been in the area of monitoring, both in the operating room and neurointensive care units. With the advancement of near-infrared spectroscopy (NIRS) and transcranial Doppler, the detection of cerebral ischemia, especially under general anesthesia, has
improved tremendously. Multimodal intraoperative monitoring of spinal cord sensory and motor function during
surgical correction of adult spinal deformity is feasible and provides useful neurophysiological data with an overall
sensitivity of 100% and specificity of 84.3%.40 In the neurointensive care unit, insertion of microdialysis and multiparameter biochemical probes into traumatized human brains has confirmed findings. Cytokine production and proapoptotic markers have been detected during oxidative stress, and these markers have recovered during enhanced
perfusion. Ischemic events have been associated with tissue acidosis, and spontaneous depolarizations have been
observed. Use of these monitors in the operating room has occurred. What remains to be totally defined is what values constitute thresholds for interventions. These thresholds have been characterized mostly for NIRS monitoring,
and much of this has revolutionized the treatment of patients with the possibility of cerebral hypoperfusion.
The development of neuroanesthesia has paralleled advances in neurosurgery. As techniques and procedures
have become advanced, so have the techniques and protocols to anesthetize and monitor neurologic function. The
overall goal is to provide a good surgical outcome and better quality of life. Procedures will become less invasive
with more functional neurosurgery requiring a cooperative patient. This makes the anesthetic management for these
procedures even more complex. It is also likely that there will be greater integration of neurosurgery and neuroradiology, with greater emphasis on maintaining cerebral vascular function without the effects of inhalational anesthetics and opioids. Neurosurgery is ever evolving; the practice of anesthesiology for these procedures will also have to
evolve to accommodate the demands of the surgeon and improve patient outcomes. Neuroanesthesia practice will
shadow neurosurgical breakthroughs. These changes will accelerate over the next 10 years as scientific advances,
and the understanding of the diseases we treat enhance the capacity of the anesthesiologist to develop techniques to
provide an ideal surgical environment with rapid awakening to assess neurologic function.
References
1. Hippocrates on injuries of the head [Adams F, Trans.]. London: The Genuine Works of Hippocrates, in 2 vols.; 1849.
2. Frost EAM. A history of neuroanesthesia. In: Eger E, Saidman LJ, Westhope RN, editors. The wondrous story of anesthesia. New York: Springer;
2014. p. 871–85.
3. Gunther RT. Dioscorides Pedanius. The greek herbal of dioscorides. Oxford: Oxford University Press; 1934.
4. Cooper A. Lectures on the principles and practice of surgery. London: Westley; 1829.
5. Raper HR. Man against pain: the epic of anesthesia. New York: Prentice-Hall; 1945. p. 8.
6. Robinson V. Victory over pain: a history of anesthesia. New York: Henry Schumann; 1945. p. 29.
7. Walker AE. A history of neurological surgery. New York: Hafner; 1967.
8. Priestley J. Experiments and observations on different kinds of air. London: Thomas Pearson; 1790.
9. Horsley V. On the technique of operations on the central nervous system. BMJ 1906;2:411–23.
10. Horsley V. On the technique of operations on the central nervous system. Address in Surgery. Toronto Lancet 1906;2:484.
INTRODUCTION AND BRIEF HISTORY OF NEUROANESTHESIA
xxxiii
11. Lyons JB. Citizen surgeon. London: Peter Downay; 1966.
12. Beecher HK. The first anesthesia records (Codman Cushing). Surg Gynecol Obstet 1940;71:689.
13. Cushing HW. Concerning a definitive regulatory mechanism of the vasomotor center which controls blood pressure during cerebral compression. Bull Johns Hopkins Hosp 1901;12:290.
14. Cushing HW. Some principles of cerebral surgery. JAMA 1909;52:184.
15. Shephard DA. Harvey Cushing and anesthesia. Can Anaesth Soc J 1965;12:431–2.
16. Halsted WS. Surgical papers. Baltimore: Johns Hopkins Press; 1924. p. 167.
17. Krause F. [Haubold H, Thorek M, Trans.]. Surgery of the brain and spinal cord based on personal experiences, vol. 1. New York: Rebman & Co.; 1912.
p. 137.
18. Jackson DE. A study of analgesia and anesthesia with special reference to such substances as trichloroethylene and vinesthene together with
apparatus for their administration. Anesth Analg (Curr Res) 1934;13:198.
19. Hershenson BB. Some observations on anesthesia for neurosurgery. NY State J Med 1942;42:2111.
20. Lundy JS. Intravenous anesthesia: preliminary report of the use of two new thiobarbiturates. Mayo Clin Proc 1935;10:536.
21. Johnstone M. The human cardiovascular response to flurothane anaesthesia. Br J Anesth 1956;28:392.
22. Stephen CR, Pasquet A. Anesthesia for neurosurgical procedures. Analysis of 1000 cases. Anesth Analg 1949;28:77.
23. Lundy JS. Clinical anesthesia. Philadelphia: WB Saunders; 1942. p. 3.
24. Kety SS, Schmidt CF. Determination of cerebral blood flow in man by the use of nitrous oxide in low concentrations. Am J Physiol 1945;143:53.
25. Lassen NA, Ingvar DH. The blood flow of the cerebral cortex determined by radioactive krypton. Experientia 1961;17:42.
26. Lundberg N. Continuous recording and control of ventricular fluid pressure in neurosurgical practice. Acta Psychiatr Neurol Scand 1960;36
(Suppl. 149).
27. Langfitt TW. Increased intracranial pressure. Clin Neurosurg 1969;16:438.
28. Michenfelder JD, Gronert VA, Rehder K. Neuroanesthesia. Anesthesiology 1969;30:65–100.
29. Alexander SC, Wollman H, Cohen PJ, Chase PE, Behar M. Cerebral vascular responses to PaCO2 during halothane anesthesia in man. J Appl
Physiol 1964;19:561.
30. Smith AL, Wollman J. Cerebral blood flow and metabolism: Effects of anesthetic drugs and techniques. Anesthesiology 1972;36:378.
31. Pierce Jr EC, Lambertsen CJ, Deutsch S, Chase PE, Linde HW, Dripps RD, et al. Cerebral circulation and metabolism during thiopental anesthesia and hyperventilation in man. J Clin Invest 1962;41:1664.
32. Okuda Y, McDowall DG, Ali MM, Lane JR. Changes in CO2 responsiveness and in autoregulation of the cerebral circulation during and after
halothane induced hypotension. J Neurol Neurosurg Psychiatry 1972;39:221.
33. Harper AM, Bell RA. The effect of metabolic acidosis and alkalosis on the blood flow through the cerebral cortex. J Neurol Neurosurg Psychiatry
1963;26:341.
34. Gelb AW, Craen RA, Rao GS, Reddy KR, Megvesi J, Mohanty B, et al. Does hyperventilation improve operating condition during supratentorial craniotomy? A multicenter randomized crossover trial. Anesth Analg 2008;106:585–94.
35. SAFE Study Investigators, Australian and New Zealand Intensive Care Society Clinical Trials Group, Australian Red Cross Blood Service,
George Institute for International Health, Myburgh J, Cooper DJ, Finfer S, Bellomo R, Norton R, Bishop N, et al. Saline or albumin for fluid
resuscitation in patients with traumatic brain injury. N Engl J Med 2007;357:874–84.
36. Clifton GL, Valadka A, Zygun D, Coffey CS, Drever P, Fourwinds S, et al. Very early hypothermia induction in patients with severe brain
injury (the National Acute Brain Injury Study: Hypothermia II): a randomized trial. Lancet Neurol 2011;10:131–9.
37. Li LR, You C, Chaudhary B. Intraoperative mild hypothermia for postoperative neurological deficits in intracranial aneurysm patients.
Cochrane Database Syst Rev 2012;2:CD008–445.
38. Connolly Jr ES, Rabinstein AA, Carhuapoma JR, Derdeyn CP, Dion J, Higashida RT, et al. Guidelines for the management of aneurysmal
subarachnoid hemorrhage: a guideline for healthcare professionals from the American Heart Association. Stroke 2012;43:1711–37.
39. Hindman BJ, Bayman EO, Pfisterer WK, Torner JC, Todd MM. IHAST Investigators. No association between intraoperative hypothermia of
supplemental protective drug and neurologic outcomes in patients undergoing temporary clipping during cerebral aneurysm surgery: findings from the intraoperative hypothermia for aneurysm surgery trial. Anesthesiology 2010;112(1):86–101.
40. Quraishi NA, Lewis SJ, Kelleher MO, Sarjeant R, Rampersaud UR, Fehlings MG. Intraoperative multimodality monitoring in adult spinal
deformity: analysis of a prospective series of one hundred two cases with independent evaluation. Spine (Phil PA 1976) 2009;34:1504–12.
This page intentionally left blank
S E C T I O N
I
NEUROANATOMY
This page intentionally left blank
C H A P T E R
1
Neuroanatomy
D. Gupta
Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
O U T L I N E
Introduction
3
Embryological Differentiation of Different
Parts of Brain
4
Anatomy of Brain
Cerebrum
Frontal Lobe
Temporal Lobes
Parietal Lobe
Functional Areas (of Cerebral Cortex)
Premotor Area
Sensory Areas
Visual Area
Acoustic (Auditory) Area
Association Areas
Diencephalon
The Thalamus
Hypothalamus
Epithalamus
Habenular Nucleus
Afferent Fibers
Efferent Fibers
Nucleus Subthalamicus
Zona Incerta
Basal Ganglia
4
4
6
6
7
8
10
10
11
11
11
11
11
13
15
15
16
16
16
16
16
Internal Capsule
White Matter
Corpus Callosum
Ventricular System
Lateral Ventricles
Third Ventricle
Fourth Ventricle
Limbic System
Midbrain (Mesencephalon)
Pons
Medulla
Reticular Formation
Cerebellum
17
18
19
19
20
21
22
22
23
25
26
29
29
Vascular Supply of the Brain
Arterial System
Cerebral Venous System
30
30
32
The Meninges and Cerebrospinal Fluid
The Meninges
Dura Mater
The Spinal Cord
Ascending Tracts of Spinal Cord (Sensory Tracts)
33
33
33
34
36
Acknowledgment
39
References
40
INTRODUCTION
Why should a well-established neuroanesthetist study clinical neuroanatomy? This question, albeit a vexing
one, is very pertinent in the present day scenario. The answer is evident. A tower of knowledge built on broad
and diverse information helps one to prepare for all the eventualities that one may encounter. Anatomy is the
basis of every procedure that we perform. An anesthetist who embarks on a new journey into the anatomical
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00001-4
3
© 2017 Elsevier Inc. All rights reserved.
4
1. NEUROANATOMY
basis of his or her clinical practice has adapted well to a vista that emphasizes fundamental sciences as the basis
of all medical education. It helps the person improve his or her procedural skills. Finally, it helps him or her to
be better equipped to deal with a changing and unpredictable world where knowledge empowers and also acts
as a haven of safety.
About 100 billion neurons and 10–50 trillion neuroglias make up the brain, which has a mass of about 1300–
1500 g in adult. On average each neuron forms 1000 synapses with other neurons1. The total number of synapses, about 1000 trillion, is larger than the number of stars in the galaxy. The central nervous system (CNS)
included the brain and the spinal cord and is composed of (1) cerebral hemisphere, (2) diencephalon, (3) basal
ganglion, (4) midbrain, (5) pons, (6) medulla, (7) cerebellum, and (8) spinal cord. This chapter will provide
information of these parts individually, that is integrated, informative, and relevant to educational need of the
neuroanesthesiologists.
EMBRYOLOGICAL DIFFERENTIATION OF DIFFERENT PARTS OF BRAIN
Knowledge of the embryological development of the brain is necessary to understand the terminology used for
the principal part of the adult brain. The development of the brain is dealt with details in the following chapter.
ANATOMY OF BRAIN
Cerebrum
The cerebrum consists of two cerebral hemispheres that are partially connected with each other by corpus callosum. Each hemisphere contains a cavity called the lateral ventricle. The cerebrum is arbitrarily divided into five
lobes: frontal, parietal, temporal, occipital, and insula.2 On the lateral surface three sulci (central, lateral or Sylvian,
and parietooccipital sulci) and two imaginary lines divide the cerebrum into four lobes (Fig. 1.1). The first imaginary
line (lateral parietotemporal line) is drawn from parietooccipital sulcus to preoccipital notch and second (temporooccipital line) backward continuation of posterior ramus of lateral sulcus before it turns upward to meet first line.
The central sulcus and posterior ramus of Sylvian fissure (SF) separate frontal lobe from parietal lobe and temporal
lobe. Posteriorly parietooccipital sulcus and lateral parietotemporal line separate occipital lobe from parietal lobe
and temporal lobe. Temporal and parietal lobes are separate by posterior ramus of SF and temporo-occipital line
(Fig. 1.1).
The cerebral cortex is the outermost sheet of neural tissue of the cerebrum whereas white matter lies in the center.
Cerebral cortex is folded into sulci and gyri, which actually increases the surface area of cortex. Sulci include the
central lateral and parietooccipital.
The central sulcus begins by cutting the superomedial border of the hemisphere a little behind the midpoint between
the frontal and parietal lobe. It runs on the superolateral surface obliquely downward and forward for about 8–10 cm
and ends a slight above the posterior ramus of lateral sulcus. It separates precentral gyrus (motor area) from postcentral gyrus (sensory area) (Figs. 1.2 and 1.3). It was originally called the fissure of Rolando or the Rolandic fissure.
FIGURE 1.1 Schematic diagram of lateral aspect of left cerebral hemisphere. Line 1. Lateral parietotemporal line; Line 2. Temporooccipital line.
I. NEUROANATOMY
ANATOMY OF BRAIN
5
FIGURE 1.2 Brain anatomy. Superior view. 1. Longitudinal fissure of cerebrum. 2. Frontal pole. 3. Superior margin of cerebrum. 4. Superior
frontal sulcus. 5. Inferior frontal sulcus. 6. Precentral sulcus. 7. Central sulcus. 8. Postcentral sulcus. 9. Intraparietal sulcus. 10. Parietooccipital
sulcus. 11. Transverse occipital sulcus. 12. Occipital pole. 13. Superior parietal lobule. 14. Inferior parietal lobule. 15. Paracentral lobule. 16.
Postcentral gyrus. 17. Precentral gyrus. 18. Inferior frontal gyrus. 19. Middle frontal gyrus. 20. Superior frontal gyrus.
FIGURE 1.3 Brain anatomy. Lateral view of right hemisphere. 1. Central sulcus. 2. Precentral sulcus. 3. Precentral gyrus. 4. Superior frontal
gyrus. 5. Superior frontal sulci. 6. Middle frontal gyrus. 7. Middle frontal sulcus. 8. Frontal pole. 9. Orbital gyri. 10. Olfactory bulb. 11. Olfactory
tract. 12. Anterior ramus of lateral sulcus (Sylvian fissure (SF)). 13. Frontal operculum. 14. Ascending ramus lateral sulcus (SF). 15. Frontoparietal
operculum. 16. Posterior ramus lateral sulcus (SF). 17. Superior temporal gyrus. 18. Middle temporal gyrus. 19. Superior temporal sulcus. 20.
Inferior temporal sulcus. 21. Inferior temporal gyrus. 22. Pons. 23. Pyramid (medulla oblongata). 24. Olive. 25. Flocculus. 26. Cerebellar hemisphere. 27. Preoccipital notch. 28. Occipital pole. 29. Postcentral gyrus. 30. Supramarginal gyrus. 31. Angular gyrus. 32. Transverse occipital sulcus.
33. Inferior parietal lobule. 34. Intraparietal sulcus. 35. Superior parietal lobule. 36. Postcentral sulcus.
I. NEUROANATOMY
6
1. NEUROANATOMY
The lateral sulcus or Sylvian fissure (SF) is one of the earliest-developing sulci of the human brain. It first appears
around the 14th gestational week.3 It is the deepest and most prominent of the cortical sulci. The lateral sulcus (SF)
separates frontal and parietal lobes from temporal lobe. It begins on the superomedial margin. The SF starts on basal
and extends to the lateral surface of the brain. It has both a superficial part and a deep part. Superficial part has a
stem and three rami (Figs. 1.2 and 1.3). The anterior portion of the deep part (Sylvian cistern) is called the sphenoidal
compartment and the posterior part is called the operculoinsular compartment. SF is an important corridor in neurosurgery as it connects the surface of anterior part of brain to its depth with all the neural and vascular components
along the way. The structures within the reach through the Transylvian approach include middle cerebral artery; optic
nerves; internal carotid artery; and its branched lamina terminalis, insula, basal ganglia, and interpeduncular fossa.
Parietooccipital sulcus begins on the medial surface of hemisphere nearly 5 cm in front of the occipital pole
(Fig. 1.4). The upper end of the sulcus reaches the superomedial border to meet the calcarine sulcus, and a small
part of it is seen on the superolateral surface.
Frontal Lobe
The frontal lobe is an area in the brain of mammals, located at the front of each cerebral hemisphere and positioned
anterior to (in front of) the parietal lobe and superior and anterior to the temporal lobes. A prefrontal sulcus runs
downward and forward parallel to the central sulcus. The area between it and central sulcus is the precentral gyrus.
Two sulcus run horizontally anterior to precentral gyrus, i.e., superior and inferior frontal sulcus and divide the region
into superior, middle, and inferior frontal gyri (Fig. 1.3). The frontal lobe contains most of the dopamine-sensitive
neurons in the cerebral cortex associated with reward, attention, short-term memory tasks, planning, and motivation.
Temporal Lobes
Temporal lobes are bounded by SF superiorly and temporo-occipital and lateral parietotemporal line posteriorly
(Fig. 1.3). The temporal lobe has two sulci, superior and inferior, that run parallel to the posterior ramus of the lateral
sulcus and divide superiolateral surface into superior, middle, and inferior temporal gyri (Fig. 1.3). The temporal
lobes are involved in the retention of visual memories, processing sensory input, comprehending language, storing
new memories, emotion, and deriving meaning.4
FIGURE 1.4 Brain surface anatomy, view of medial surface of right hemisphere. 1. Frontal pole of frontal lobe. 2. Medial frontal gyrus. 3.
Cingulate sulcus. 4. Sulcus of corpus callosum. 5. Cingulate gyrus. 6. Paracentral lobule. 7. Precuneus. 8. Subparietal sulcus. 9. Parietooccipital sulcus.
10. Cuneus. 11. Calcarine fissure. 12. Occipital pole of occipital lobe. 13–16. Corpus callosum (cut surface). (Parts of Corpus callosum 13. Splenium.
14. Trunk. 15. Genu. 16. Rostrum.). 17. Lamina terminalis. 18. Anterior commissure. 19. Septum pellucidum. 20. Fornix. 21. Tela choroidea of the third
ventricle. 22. Choroid plexus of the third ventricle. 23. Transverse cerebral fissure. 24. Thalamus. 25. Interthalamic adhesion. 26. Interventricle foramen
of Monro. 27. Hypothalamus. 28. Suprapineal recess and pineal body. 29. Vermis of cerebellum. 30. Cerebral hemisphere. 31. Choroid plexus of the
fourth ventricle. 32. Medulla oblongata. 33. Pons. 34. Fourth ventricle. 35. Tectal lamina and mesencephalic aqueduct of Sylvius. 36. Mammillary body.
37. Oculomotor nerve. 38. Infundibular recess. 39. Temporal lobe of lateral occipitotemporal gyrus. 40. Rhinal fissure. 41. Hypophysis with adenohypophysis (anterior lobe) and neurohypophysis (posterior lobe) of pituitary gland. 42. Optic chiasma. 43. Optic nerve. 44. Olfactory bulb and tract.
I. NEUROANATOMY
ANATOMY OF BRAIN
7
Parietal Lobe
The parietal lobe is positioned superior to the occipital lobe and posterior to the frontal lobe. The parietal lobe
is bounded anteriorly by central sulcus, inferiorly by SF and temporo-occipital line, medially by interhemispheric
fissure, and posteriorly by parietotemporal line. The two main sulci are postcentral sulcus, which run downward
and forward parallel to central sulcus, and intraparietal sulci, which are directed posteriorly and inferiorly toward
occipital pole. Thus divide the parietal lobe into postcentral gyrus, superior parietal lobule, and inferior parietal
lobule (Fig. 1.3). The upturn posterior end of the posterior ramus of lateral sulcus or SF extends into inferior parietal
lobule, also superior and inferior temporal sulci turn upward to enter into this lobule and constitute supramarginal,
angular gyri, and arcus temporo-occipitalis (Fig. 1.3).
The parietal lobe integrates sensory information from different modalities, particularly determining spatial sense
and navigation. For example, it comprises somatosensory cortex and the dorsal stream of the visual system. This
enables regions of the parietal cortex to map objects perceived visually into body coordinate positions. Several portions of the parietal lobe are important in language processing. Just posterior to the central sulcus lies the postcentral
gyrus. This area of the cortex is responsible for somatosensation.5
The occipital lobule occupies space behind the lateral parietotemporal line. It has a number of short lobules
divided by short sulci. A horizontal sulci, lateral to occipital sulcus divides the lobe into superior and inferior occipital gyri (Fig. 1.3). A vertical strip anterior to curved lunate sulcus is the gyrus descendens. The transverse occipital
sulcus is located in the uppermost part of the occipital lobe. A strip superiolateral to this sulcus is arcus parietooccipitalis. The occipital lobe is the visual processing center of the brain containing most of the anatomical region of
the visual cortex.6
Insula is a portion of the cerebral cortex folded deep within the lateral sulcus. This area grows less than its surrounding areas during development and thus lies deep and not seen from surface view (Fig. 1.5). The surrounding
cortical areas are called opercula such as frontal opercula, frontoparietal opercula, and temporal opercula. The insula
are believed to be involved in consciousness and play a role in diverse functions usually associated to emotion or the
regulation of the body’s homeostasis. These functions include perception, self-awareness, cognitive functioning, and
interpersonal experience.
Two hemispheres are attached with each other by corpus callosum. On the medial surface above the corpus callosum there are many sulci and gyri (Fig. 1.4). The most prominent sulcus is the cingulate sulcus, which follows the
curve course parallel of corpus callosum. The area between the cingulate sulcus and corpus callosum is the gyrus
cinguli. Above the cingulate sulcus, large anterior part is medial frontal gyrus and posteriorly paracentral lobule (Fig.
1.4). Behind the paracentral lobule, two major sulci, parietooccipital sulcus and calcarine sulcus, cut the area into a
triangular area called the cuneus. Between parietooccipital sulcus and paracentral lobule, a quadrangular area is
called precuneus, which is anteriorly separated from gyrus cinguli by suprasplenial sulcus.
FIGURE 1.5 Coronal section through the brain. 1. Longitudinal fissure of the cerebrum. 2. Cingulate sulcus. 3. Cingulate gyrus. 4. Corpus
callosum. 5. Sulcus of corpus callosum. 6. Caudate nucleus. 7. Claustrum. 8. Putamen. 9. Lateral sulcus (Sylvian fissure). 10. Globus pallidus. 11.
Thalamus. 12. Subthalamic nucleus. 13. Mammillary body. 14. Amygdala. 15. Optic tract. 16. Third ventricle and choroid plexus. 17. Body of fornix.
18. Lateral ventricle and choroid plexus. 19. Cortex of insula. 6, 8, and 10. Corpus striatum. 8 and 10. Lentiform nucleus.
I. NEUROANATOMY
8
1. NEUROANATOMY
The majority of the space of cerebral hemisphere deep to the cortex is full of the white matter. There are some
important structures that are embedded within the white matter. On coronal section the corpus callosum is seen as
a strip connecting both the hemispheres (Fig. 1.5). Third ventricle is situated in midline just below corpus callosum.
Thalamus and hypothalamus, which are derived from diencephalon, lie adjacent to lateral wall of the third ventricle.
Caudate nucleus is situated above and lateral to thalamus. Another gray matter mass lentiform nucleus lies more
lateral and just deep to insula. There is a strip of gray matter between insula and lentiform nucleus called claustrum
(Fig. 1.5). The caudate nucleus, lentiform nucleus, claustrum along with some other gray matter nucleus are (derived
from telencephalon) collectively mentioned as basal nuclei or basal ganglia. There is a white matter, an internal capsule that lies between thalamus and lentiform nucleus (Fig. 1.5). The white matter that radiates from the upper part
of internal capsule to the cortex is called corona radiate.
Functional Areas (of Cerebral Cortex)
Korbinian Brodmann was a German neurologist who studied the brain in the early part of the 20th century.7
Brodmann originally defined and numbered (from 1 to 52) different areas of cerebral cortex based on cytoarchitecture
or how the cells were functionally organized (Box 1.1). Brodmann areas have been discussed, debated, refined, and
renamed exhaustively for nearly a century and remain the most widely used and frequently cited cytoarchitectural
organization of the human cortex.8
On the basis of function, regions of the cerebral cortex are divided into three functional categories of areas
(Fig. 1.6). (1) Primary sensory areas, which receive signals from the sensory nerves and tracts by way of relay
nuclei in the thalamus. Primary sensory areas include the somatosensory cortex in the parietal lobe, visual
area of the occipital lobe, and the auditory area in parts of the temporal lobe and insular cortex. (2) Primary
BOX 1.1
I M P O R TA N T B R O D M A N N A R E A S
Frontal lobe contains areas that Brodmann identified
as involved in cognitive functioning and in speech and
language (Fig. 1.8).
t Area 4 corresponds to the precentral gyrus or primary
motor area.
t Area 6 is the premotor or supplemental motor area.
t Area 8 is anterior of the premotor cortex. It facilitates
eye movements and is involved in visual reflexes as
well as pupil dilation and constriction.
t Areas 9, 10, and 11 are anterior to area 8. They are
involved in cognitive processes such as reasoning and
judgment which may be collectively called biological
intelligence.
t Area 44 is Broca’s area.
Parietal lobe plays a role in somatosensory processes
(Fig. 1.8).
t Areas 3, 2, and 1 are located on the primary sensory
strip, with area 3 being superior to the other two.
These are somastosthetic areas, meaning that they are
the primary sensory areas for touch and kinesthesia.
t Areas 5, 7, and 40 are found posterior to the primary
sensory strip and correspond to the presensory to
sensory association areas.
t Area 39 is the angular gyrus.
Temporal lobe: Areas that are involved in the processing of auditory information and semantics as well as the
appreciation of smell (Fig. 1.8).
t Area 41 is the primary auditory area.
t Area 42 is immediately inferior to area 41 and also
involved in the detection and recognition of speech.
The processing done in this area of the cortex
provides a more detailed analysis than that done in
area 41.
t Areas 21 and 22 are the auditory association areas.
Both areas are divided into two parts; one half of each
area lies on either side of area 42.
t Area 37 is found on the posterior–inferior part of the
temporal lobe.
Occipital lobe contains areas that process visual
stimuli (Fig. 1.8).
t Area 17 is the primary visual area.
t Areas 18 and 19 are the secondary visual areas.
I. NEUROANATOMY
ANATOMY OF BRAIN
9
motor cortex, which sends axons down to motor neurons in the brain stem and spinal cord and finally innervate voluntary skeletal muscles; (3) remaining parts of the cortex, which are called the association areas. These
areas receive input from the sensory areas and lower parts of the brain which integrate sensory information
with emotional states, memories, learning, and rational thought processes that we call perception, thought, and
decision-making.
Motor areas—The motor area is classically located in precentral gyrus on the superiolateral surface of the hemisphere and in anterior part of paracentral lobule. It is shaped like a pair of headphones stretching from ear to ear
(Fig. 1.6). Specific area within the motor cortex controls voluntary muscle activity on the opposite part of body. The
body is represented on the motor strip in an upside–down fashion (Fig. 1.7). The lower parts of the body, such as the
FIGURE 1.6 Traditional concept of functional areas on the superolateral aspect of the cerebral hemisphere (left sided).
FIGURE 1.7 The motor homunculus in primary motor cortex. Coronal section anterior view of the left hemisphere.
I. NEUROANATOMY
10
1. NEUROANATOMY
feet and the legs, receive motor movement commands from the superior part of the precentral gyrus (motor strip).
Parts of the face, on the other hand are innervated by the inferior part of the motor strip. The motor strip extends
down some distance into the longitudinal cerebral fissure. The portion inside this fissure is its medial aspect. The
part on the lateral surface of the hemisphere is called its lateral aspect. The medial cortex controls the movements of
the body from the hips on down while the lateral aspect sends commands to the upper body including the larynx,
face, hands, shoulders, and trunk (Fig. 1.7). The medial and lateral aspects of the motor strip have different blood
supplies. Blood comes to the medial area from the anterior cerebral artery while the lateral cortex is supplied by the
middle cerebral artery.
Premotor Area
There is supplementary motor area on and above the superior part of cingulate sulcus on the medial aspect hemisphere that reaches to the premotor cortex (Brodmann areas 6 and 8) on the lateral surface of brain. The cortical area
in inferior frontal gyrus corresponds to motor speech area or speech area of Broca (Brodmann areas 44 and 45) and
frontal eye area (Fig. 1.8). Lesion into the motor speech area of Broca results in aphasia even the muscles concerned
are intact. In 95% of right-handers do have left-hemisphere dominance for language functions, only around 19% of
left-handers have right-hemisphere language dominance, with another 20% or so processing language functions in
both hemispheres.9
Sensory Areas
From the specific nuclei of the thalamus, neurons are projected into two somatosensory areas of the cortex: somatosensory area I in postcentral gyrus and somatosensory area II in the wall of the SF (Fig. 1.6). The arrangement of the
thalamic fibers in somatosensory area I is such that the part of the body is represented in order along the postcentral
gyrus, with leg on the top and head at the foot of the gyrus. The area of the cortex that receives sensation from a part
FIGURE 1.8 Brodmann areas in the neocortex. A number of important Brodmann areas have been marked out in the figure.
I. NEUROANATOMY
ANATOMY OF BRAIN
11
of the body is not proportional to the size of that part rather the complexity of sensation received from it, as cortical
areas for the sensation from the trunk and back are small whereas, hand and part of mouth concerned in speech are
very large.
Visual Area
The primary visual receiving area (visual cortex, Brodmann area 17) is located primarily in occipital lobe on the
sides of the calcarine fissure (Fig. 1.6). This area also extends into the cuneus and into the lingual gyrus. The visual
area is continuous, above and below, with area 18 and beyond this 19. These areas often described as psychovisual
areas are responsible mainly for interpretation of visual impulses reaching visual area (Fig. 1.6).
Acoustic (Auditory) Area
The primary auditory cortex (Brodmann area 41) is in the superior portion of temporal lobe. It is located in the part
of temporal gyrus which forms the inferior wall of the posterior ramus of the lateral sulcus (Fig. 1.6). The auditory
associated areas adjacent to the primary auditory receiving areas are widespread and extend into insula.
Association Areas
Specific areas of the cerebral cortex integrate sensory information with emotional states, memories, learning, and
rational thought processes. Primary motor cortex (precentral gyrus of frontal lobe) is located just anterior to the
central sulcus in the frontal lobe of the cerebral cortex, of the gray matter motor neurons, which initiates impulses
routed through the medulla and spinal cord. It represents the conscious voluntary commands to the prime movers
of skeletal muscle groups for specific actions; it is highly organized with specific regions representing each part of
the body. Primary sensory cortex (postcentral gyrus of parietal lobe) is located just posterior to the central sulcus
in the parietal lobe of the cerebral cortex, of the somatic sensory neurons and receives impulses from the thalamus,
medulla, and spinal cord. It responds with the first conscious perceptions/awareness of cutaneous sensations arriving from stimulated receptors in the skin and subcutaneous tissues; it is highly organized with specific regions
representing each part of the body. Frontal eye field—a specific motor area within the frontal cortex which controls
the voluntary scanning movements of the eyes, such as tracking a bird in flight, by sending impulses to the extrinsic
muscles of the eyes.
Diencephalon
The diencephalon is midline structure and embedded in the cerebrum. The third ventricle is considered as the
cavity of diencephalon. Diencephalon is bounded anteriorly from the plane through the optic chiasm and anterior
commissure; caudally from plane through the posterior commissure and the caudal edge of the mammillary bodies;
medially from wall of the third ventricle, stria medullaris thalami, and mass intermedia; laterally from the internal
capsule, tail of caudate nucleus, and stria terminalis; and dorsally by the fornix and floor of the lateral ventricles
(Figs. 1.4 and 1.5).
Diencephalon consists of the following parts:
1.
2.
3.
4.
Thalamus
Hypothalamus
Subthalamus
Epithalamus
The Thalamus
It is a large, egg-shaped (ovoid), 4 × 1.5-cm nuclear mass. It makes up about 80% of the mass of the diencephalon. It consists mainly of gray matter, but its superior and lateral surfaces are covered by thin layers of white matter termed the stratum zonale and the external medullary lamina, respectively. The gray matter is incompletely
divided into anterior, medial, and ventrolateral nuclei by a Y-shaped lamina of white matter called the internal
medullary lamina. It has two ends (anterior and posterior) and four surfaces (superior, inferior, medial, and lateral)
(Fig. 1.9).
I. NEUROANATOMY
12
1. NEUROANATOMY
FIGURE 1.9 Schematic representation of thalamic nuclei and their projections. CM, centromedial; LD, lateral dorsal; LGB, lateral geniculate
body; LP, lateral posterior; MD, mediodorsal; MGB, medial geniculate body; VA, ventroanterior; VL, ventrolateral; VPI, ventroposterior inferior;
VPL, ventroposterior lateral; VPM, ventroposterior medial; VPMpc, ventroposterior medial part mammillothalamic.
It extends anteriorly to the interventricular foramen; superiorly to the transverse cerebral fissure (between corpus
callosum and fornix); inferiorly to the hypothalamic sulcus; and posteriorly it overlaps the midbrain (pulvinar).
Anterior end of thalamus is smaller than posterior end and lies behind interventricular foramen which connects
the lateral ventricle and the third ventricle. Posterior end is large and expanded, called as pulvinar (Fig. 1.9). It is
projected backwards and laterally over superior colliculus of midbrain. There are two small swellings on inferior
surface of pulvinar called the medial and lateral geniculate bodies. Superior surface is not clearly demarcated from
lateral surface. Stria medullaris thalami marks the junction between the superior and medial surfaces. It is separated from the ventricular surface of caudate nucleus by the stria terminalis and thalamostriate vein. It is divided
into two areas by an impression produced by the lateral margin of fornix. The lateral area is covered by ependyma
and forms part of the floor of the body of lateral ventricle (Fig. 1.5). The median area is covered by the tela choroidea
I. NEUROANATOMY
ANATOMY OF BRAIN
13
of the third ventricle (double fold of pia matter) (Fig. 1.4). Inferior surface lies upon the subthalamic tegmental
region [i.e., hypothalamus, subthalamus, and midbrain (from before backwards)]. Medial surface forms part of
lateral wall of the third ventricle separated from corresponding surface of opposite thalamus by a narrow interval.
The two thalami are connected by a short band called the interthalamic adhesion (Fig. 1.4). Lateral surface separated from lentiform nucleus by posterior limb of internal capsule. Many fibers stream out of this surface and enter
internal capsule en route for cerebral cortex and form the thalamic radiation.
Functions of Thalamus
1. Relay station
a. Most somatic sensory pathways except olfaction
b. Few motor pathways (e.g., cerebellar)
2. Integrating center
For impulses from many sources (e.g., somatic sensory; visual; visceral; some motor, e.g., cerebellar, corpus
striatum)
3. Maintenance and regulation of state of:
Consciousness, alertness, attention (through influence upon cerebral cortex)
4. Emotional connotations
(Which accompany most sensory experiences?)
5. Crude sensations
(For example, pain which may reach consciousness at this level even when all connections between thalamus
and cortex are destroyed.)
Thalamus is not simply a relay station where information is passed onto the neocortex but thalamus acts as receptionist for information to the cerebral cortex, preventing or enhancing the passage of specific information depending
on behavioral state of the individual. Though it has more than 50 nuclei, however, classically they are divided into
four groups depending on their position in relation to the internal medullar lamina.
Anterior group is connected from mammillary bodies and subiculum of the hippocampal formation and closely
associated with the limbic system (Fig. 1.9). This connection is concerned with emotional tone and mechanism of
recent memory. Stimulation or ablation of mammillothalamic tract causes alteration in autonomic control and loss
of recent memory.
The medial group receives input from basal ganglion, amygdala, and midbrain and its major output is to the
frontal cortex (Fig. 1.9). It provides mechanisms for the integration of certain somatovisceral impulses projecting
to prefrontal cortex. It mediates impulses of an affective nature which contributes to the formation of personality.
Stimulation, disease, or surgical ablation of medial nuclei results in changes in (1) motivational drive, (2) ability to
solve problems, (3) consciousness level, (4) general personality, (5) subjective feeling status (affective tone), (6) pain
perception (indifference to pain), (7) emotional content.
The ventral anterior and anterior lateral nuclei transmit information basal ganglia and cerebellum to the motor
cortex (Fig. 1.9). These make important contribution to initiation of movements, control of muscle tone, regulation
of cortical reflexes.
The posterior group includes medial and lateral geniculate nucleus, lateral posterior nuclei, and pulvinar
(Fig. 1.9). Lateral geniculate nucleus receives information from the retina and conveys to the primary visual cortex;
medial geniculate nucleus is a component of the auditory system.
The nonspecific projecting nuclei are located either in the midline or within the internal medullary lamina. The
largest intralaminar nuclei, centromedial nucleus, is projected to amygdala, hippocampus, and basal ganglia.
Hypothalamus
Hypothalamus is a part of the diencephalon which forms lateral wall and floor of the third ventricle. Laterally it
is in contact with internal capsule and ventral thalamus; posteriorly it merges with ventral thalamus and tegmentum
of the midbrain; anteriorly it extends up to lamina terminalis; and inferiorly it is related with structures in the floor
of the third ventricle (tuber cinereum, infundibulum, and mammillary bodies).
Hypothalamus receives afferent from retina, frontal lobe, hippocampus, corpus striatum, and reticular formation of brain stem. It has efferent to supraopticohypophyseal tract from optic nuclei to the posterior pituitary, pars
tuberalis, and pars intermedius. Thus posterior pituitary is brought under the retinal control. It also has efferent to
mammillothalamic tract and mammillotegmental tract. The Hypothalamus is also subdivided anterior-posteriorly
into three regions (Box 1.2).
I. NEUROANATOMY
14
1. NEUROANATOMY
BOX 1.2
HYPOTHALAMIC NUCLEI (FIG. 1.10)
Zone
1. Preoptic
2. Supraoptic (anterior)
3. Infundibulo- tubular (middle)
4. Mammillary (caudal)
Nuclei
Preoptic nuclei
1.
2.
3.
Supraoptic nuclei
Paraventricular nuclei
Suprachiasmatic nuclei
1.
2.
3.
Dorsomedial nuclei
Ventromedial nuclei
Arcuate nuclei (infundibular)
1.
2.
3.
Posterior nuclei
Mammillary nuclei
Mammilloinfundibular nuclei
FIGURE 1.10 Nuclei in the right hypothalamus: midsagittal section of the right hemisphere viewed from the medial side.
I. NEUROANATOMY
ANATOMY OF BRAIN
15
Functions of Hypothalamus
1. Endocrine control:
i. Release factors
Anterior pituitary
ii. Release inhibiting factors
2. Neurosecrtion:
i. Vasopressin (supraoptic nucleus)
Posterior pituitary
ii. Oxytocin (paraventricular nucleus)
3. Autonomic control:
Higher center for control of lower autonomic centers in brainstem & spinal cord
i. Anterior region
Influence parasypathetic activity
ii. Preoptic region
iii. Posterior region
Influence sympathetic activity
iv. Lateral region
4. Temperature regulation
a. Anterior region controls dissipation of heat
b. Posterior region controls conservation of heat
5. Food intake regulation
a. Lateral region (hunger center) initiates eating and increases food intake
b. Medial region (satiety center) inhibits eating and reduces food intake
6. Water intake and balance
Lateral region (thirst center) increases water intake. Vasopressin effects on distal convoluted tubules and
collecting tubules of kidney.
7. Emotion and behavior
Hypothalamus, limbic system and prefrontal cortex interconnect intensively and responsible for
emotional state and behavior however, hypothalamus generates behaviors involved in rage, aggression
and escape.
8. Circadian rhythms (daily rhythm of a biological function)
Circadian rhythms include body temperature, adrenocortical activity, sleep, and wakefulness (anterior region).
Suprachiasmatic nucleus (afferent from retina) plays a role in control of circadian rhythms (variation in intensity
of light is transmitted by this nucleus to many hypothalamic nuclei).
Epithalamus
Epithalamus is composed of the following:
1. Pineal body
2. Habenula
3. Posterior and habenular commissures
It is the most dorsal, smallest, and oldest part of diencephalon. Epithalamus is functionally and anatomically
linked to the limbic system. It is implicated in the following functions:
1. Autonomic functions (e.g., respiratory, cardiovascular, etc.)
2. Endocrine functions (e.g., thyroid functions)
3. Reproductive functions (e.g., mating behavior)
Habenular Nucleus
The habenular nuclei are situated in relation to a triangular depression in the wall of the third ventricle called
habenular trigone. It is the center for integration of olfactory, visceral, and somatic afferent pathways (correlation of
olfactory and somatic afferent impulses). Ablation of these nuclei produces changes in metabolism, endocrine regulation, and thermoregulation.
I. NEUROANATOMY
16
1. NEUROANATOMY
Most of the fibers travel through the stria medullaris thalami.
Afferent Fibers
1.
2.
3.
4.
5.
6.
7.
Amygdaloid complex (via stria terminalis)
Hippocampal formation (via fornix)
Olfactory tubercle
Preoptic and septal areas (via stria medullaris thalami)
Anterior perforated substance
Various hypothalamic nuclei
Globus pallidus
Efferent Fibers
1. Interpeduncular nuclei (via fasciculus retroflexus)
2. Medial nucleus of thalamus
3. Tectum and reticular formation of midbrain
The stria medullaris thalami, habenula, and fasciculus retroflexes form segments of visceral efferent pathways
which carry impulses to parts of brain stem and spinal cord (e.g., Tectotegmentospinal tracts and dorsal longitudinal
fasciculus), which connect with autonomic preganglionic centers.
Nucleus Subthalamicus
It lies medial to internal capsule and is continuous with substantia nigra. Important site for integration of
a number of motor control centers especially through its connections with the corpus striatum and midbrain
tegmentum. Lesion of one subthalamic nuclei results in a condition called hemiballismus (subthalamic dyskinesia). It receives afferent fibers from globus pallidus, motor cortex (precentral), and pedunculopontine nucleus. It
has efferent fibers to globus pallidus, contralateral globus pallidus, substantia nigra, and opposite subthalamic
nucleus.
Zona Incerta
This is a thin strip of gray matter that lies between thalamic and lenticular fasciculi. It is continuous with
thalamic reticular nuclei. Functionally associated with the zona incerta are the nuclei of prerubral field and
entopeduncular nuclei. It receives signals from precentral motor cortex and sends impulses to midbrain reticular
formation.
Basal Ganglia
The basal ganglia (or basal nuclei) are a group of nuclei (mostly of telencephalic origin) in the brains of vertebrates that act as a cohesive functional unit. They are situated at the base of the forebrain and are strongly connected
with the cerebral cortex, thalamus, and other brain areas. The four main components of the basal ganglion are (1) the
striatum (caudate nucleus, putamen, and nucleus accumbens), (2) the globus pallidus, (3) the substantia nigra, and
(4) the subthalamic nucleus (Fig. 1.5).
The basal ganglia play a major role in voluntary motor movement, although they do not have direct input or
output with spinal cord. The largest component, the striatum, receives input from many brain areas but sends output only to other components of the basal ganglia. Inputs to striatum are from entire cerebral cortex (glutamatergic
fibers), intralaminar nuclei of the thalamus, pars compacta (dopaminergic fibers), raphe nuclei of reticular formation (noradrenergic fibers), and locus coeruleus (serotonergic fibers). The pallidum gets input from the striatum and
sends inhibitory output to a number of motor-related areas. The substantia nigra receives striatal input of the neurotransmitter dopamine, which plays an important role in basal ganglia function. The subthalamic nucleus receives
input mainly from the striatum and cerebral cortex, and projects to the globus pallidus. Each of these areas has a
complex internal anatomical and neurochemical organization.
The disturbance of basal ganglia causes a number of movement disorders including Parkinson’s disease, which
involves degeneration of the dopamine-producing cells in the substantia nigra pars compacta, and Huntington’s
I. NEUROANATOMY
ANATOMY OF BRAIN
17
disease, which primarily involves damage to the striatum.10 The lesion in basal ganglia causes hypertonicity (lead
pipe type), loss of automatic associated movement, and involuntary movement.
Internal Capsule
A massive layer (8–10 mm thick) of white matter situated between the caudate nucleus and thalamus (medial)
from the more laterally situated lentiform nucleus (globus pallidus and putamen) (Fig. 1.5). In axial (horizontal) section it appears in the form of a V opening out laterally. It is a narrow gate through which fibers are densely crowded
and consequently a small pinpoint lesion causes widespread neurological deficit.
It consists of five parts: an anterior limb, genu, posterior limb, retrolentiform (or retrolenticular) limb, and
sublentiform (or sublenticular) limb (Fig. 1.11A). Internal capsule consists of two types of fiber populations: fibers
ascending from the thalamus to the cerebral cortex that comprises of anterior thalamic radiation, among others, the
visual, auditory, and somatic sensory radiations, and fibers descending from the cerebral cortex to the thalamus,
subthalamic region, midbrain, hindbrain, and spinal cord (Fig. 1.11B).
1. Anterior limb lies between head of caudate nucleus and lentiform nucleus. It contains descending tract:
frontopontine fibers and ascending tract: anterior thalamic radiation.
2. Genu is a bend of internal capsule and lies close to ventricular surface immediately lateral to the foramen
of Monro. It contains descending tract: corticonuclear fiber and ascending tract: anterior fiber of thalamic
radiation.
3. Posterior limb lies between thalamus and lentiform nucleus and contains descending tracts: corticospinal tract,
frontopontine, corticorubral fibers and ascending tract: fibers of the superior thalamic radiation.
4. Retrolenticular part lies behind the lentiform nucleus and contains descending tracts: parietopontine,
occipitopontine, occipitocollicular, occipitotectal fibers and ascending tract: optic radiation.
5. Sublenticular part lies below lenticular nucleus and contains descending tracts: temporopontine, parietopontine
and ascending tract: acoustic radiation from medial geniculate body to superior temporal and transverse
temporal gyri.
As these ascending and descending fibers are densely passed through a narrow space in internal capsule, a lesion in
the internal capsule causes widespread paralysis on the opposite half of the body, which may also involve lower part
of the face and tongue. The lesions in internal capsule result from thrombosis or rupture of the arteries supplying it
(Box 1.3).
FIGURE 1.11A Schematic representation of the horizontal view showing the three parts of the internal capsule: anterior limb, genu,
and posterior limb.
I. NEUROANATOMY
18
1. NEUROANATOMY
FIGURE 1.11B Schematic representation of the horizontal view showing the ascending pathways and descending tracts passing through
the internal capsule.
BOX 1.3
B L O O D S U P P LY O F I N T E R N A L C A P S U L E
Anterior limb
Upper part is supplied by lenticulostriate branches of
middle cerebral artery, and lower part is supplied by recurrent branch (Heubner artery) of anterior cerebral artery.
Genu
Upper part is supplied by lenticulostriate branches
of middle cerebral artery, and lower part is supplied by
recurrent branch (Heubner artery) of anterior cerebral
artery.
Posterior limb
Upper part is supplied by lenticulostriate branches
of middle cerebral artery, and lower part is supplied
by anterior choroidal artery (branch of internal carotid
artery).
White Matter
The white matter is mainly myelinated nerve fibers, occupy deeper part of cerebrum, and connect various parts of
the cortex to one another and also to other part of the CNS. These may be classified as association fibers, projection
fibers, and commissural fibers (Fig. 1.12).
Association fibers: These are the fibers connecting different cortical areas of the same side to one another. Short
association fibers connect adjacent gyri. Long association fibers connect distant part of the cerebral cortex in the same
side (Fig. 1.12).
1. Uncinate fasciculus: temporal pole to motor speech area and orbital cortex
2. Cingulum fasciculus: cingulum gyrus to parahippocampal gyrus
I. NEUROANATOMY
ANATOMY OF BRAIN
19
FIGURE 1.12 Schematic diagram showing the short and long association fibers of the cerebrum. Fasc., fasciculus.
3. Superior longitudinal fasciculus: frontal to occipital and temporal lobes
4. Inferior longitudinal fasciculus: temporal to occipital lobe
Projection fibers: These are the fibers which connect the cerebral cortex to other parts of the CNS such as brain
stem and spinal cord in both directions, e.g., corticospinal and corticopontine fibers.
Commissural fibers: These fibers connect corresponding areas of the two hemispheres. The commissural fibers
include the following:
1.
2.
3.
4.
5.
6.
7.
Corpus callosum
Anterior commissure
Posterior commissure
Hippocampal commissure
Habenular commissure
Hypothalamic commissure
Cerebellar commissure
Corpus Callosum
The corpus callosum is the largest commissure, which connects two hemispheres. The corpus callosum has two
anterior parts, rostrum and genu, a central part, trunk, and posterior part, splenium (Fig. 1.4). Corpus callosum is
closely related to lateral ventricle. The fibers of the genu run forward into the frontal lobes and make the forklike
structure called the forceps minor. This forms the anterior wall of frontal horn of lateral ventricle. The rostrum is
located below and forms the floor of the frontal horn. The genu and trunk form roof of both of the frontal horns
and body of lateral ventricles. Fibers of the trunk run laterally and intersect the fiber of corona radiate. As some
fibers of trunk and splenium run laterally they form a flattened band called the tapetum. The tapetum is closely
related to posterior and inferior horn of the lateral ventricle. Tapetum separates the fibers of the optic radiation
from temporal horn and the atrium. Large fibers of splenium run backward into occipital lobe to form forceps
major.
Ventricular System
The ventricles of the brain are a communicating network of cavities filled with cerebrospinal fluid (CSF) and
located within the brain parenchyma. The ventricular system is composed of two lateral ventricles, one third ventricle, the cerebral aqueduct, and the fourth ventricle (Fig. 1.13). It is continuous with the central canal of the spinal
cord. The ventricle lining consists of a specialized epithelial membrane called ependymal layer. The choroid plexuses
located in the ventricles produce CSF, which fills the ventricles and subarachnoid space, following a cycle of constant
production and reabsorption.
CSF flows from the lateral ventricles via the foramina of Monro into the third ventricle, and then it reaches into
the fourth ventricle through the cerebral aqueduct in the brain stem (Fig. 1.13). From there it passes into the central
I. NEUROANATOMY
20
1. NEUROANATOMY
FIGURE 1.13 Human ventricular system (left lateral view).
canal of the spinal cord or into the cisterns of the subarachnoid space via three small foramina: the central foramen
of Magendie and the two lateral foramina of Luschka. The fluid then flows around the superior sagittal sinus to be
reabsorbed via the arachnoid villi into the venous system. CSF within the spinal cord flows down to the lumbar cistern at the end of the cord around the cauda equine and bath whole of the spinal cord.
Lateral Ventricles
Lateral ventricles are two c-shaped cavities one on each side of cerebral hemisphere. It consists of a central part
called body and three extensions: anterior (frontal horn), posterior (occipital horn), and inferior (temporal horn).
Each lateral ventricle communicates with the third ventricle with a common interventricular foramen or foramen of
Monro (Fig. 1.13).
Central part of the lateral ventricle extended anteroposteriorly from interventricular foramen to splenium of corpus callosum. Their boundaries are roof: trunk of corpus callosum, floor: superior surface of the thalamus medially
and body of caudate nucleus laterally and in between these two stria terminalis and thalamostrial vein, medial surface: septum pallucidum and body of fornix. There is space between fornix and upper surface of the thalamus called
choroid fissure.
The frontal horn is located in front of interventricular foramen or foramina of Monro. It is triangular in shape with
boundaries anterior part of trunk of corpus callosum as roof, head of caudate nucleus as head, septum pellucidum
as medial wall.
The occipital horn of the lateral ventricle extends backward into occipital lobe with its roof and lateral wall formed
by tapetum, optic radiation, and inferior longitudinal fasciculus; and bulb of posterior horn (floor and medial wall)
is raised by forcep major and calceravis.
I. NEUROANATOMY
ANATOMY OF BRAIN
21
FIGURE 1.14 Detailed system limbic structure and connections.
The temporal horn or inferior horn projecting form the posterior end of central part run downward and forward
into temporal lobe and reach the uncus. In cross section it is a narrow cavity with boundaries: roof and lateral wall
formed by tapetum, tail of caudate lobe, stria terminalis and amygdaloid body and floor by hippocampus.
Third Ventricle
This is a narrow funnel-shaped cavity of the diencephalon which lies between the thalamus (Fig. 1.5). It communicates anteriosuperiorly on each side with lateral ventricles by foramen of Monro or interventricular foramen.
Posteriorly, it communicates with the fourth ventricle through aqueduct of Sylvius (Fig. 1.13). It is bounded by an
anterior wall, a posterior wall, roof, floor, and two lateral walls.
The lateral wall is formed superiorly by thalamus and below by hypothalamus. Hypothalamus is separated
from thalamus by hypothalamic sulcus, a groove that extends from foramen of Monro anteriorly to aqueduct
posteriorly. The interventricular foramen of Monro is seen on lateral wall just behind the column of fornix. The
anterior wall is formed by lamina terminalis. Lamina terminalis is a thin layer of gray matter in the telencephalon
that extends backward from the corpus callosum above the optic chiasma and forms the median portion of the
rostral wall of the third ventricle of the cerebrum (Fig. 1.4). Posterior wall is formed by pineal body and posterior
commissure. The roof is extended from foramen of Monro anteriorly to suprapineal recess posteriorly. The roof is
formed by ependyma that stretches across the two thalami. The floor extends from optic chiasma anteriorly to the
orifice of aqueduct of Sylvius posteriorly. It is formed by optic and infundibular recess, tuber cinereum, mammillary bodies, and posterior perforated substance of tegmentum of midbrain. The cavity of the third ventricle shows
number of prolongations or recesses, e.g., infundibular recess, optic recess, pineal recess, and supraspinal recess.
Endoscopic third ventriculostomy is a surgical procedure in which an opening is created in the floor of the third
ventricle using an endoscope placed within the ventricular system through a burr hole. This allows the CSF to flow
directly to the basal cisterns, thereby shortcutting any obstruction. It is used as an alternative to a cerebral shunt to
treat certain forms of obstructive hydrocephalus, such as aqueductal stenosis.
I. NEUROANATOMY
22
1. NEUROANATOMY
Fourth Ventricle
The fourth ventricle is a cavity of hindbrain connected to the third ventricle by a narrow cerebral aqueduct. The
fourth ventricle is a diamond-shaped cavity located dorsal to the pons and upper medulla oblongata and anterior to
the cerebellum (Fig. 1.13). Fourth ventricle connected to the third ventricle above and central canal below. Through
medial aperture, foramen of Magendie, it communicates with subarachnoid space. Laterally on either side it communicated with subarachnoid space through foramen of Luschka.
The superior cerebellar peduncles and the anterior and posterior medullary vela form the roof of the fourth ventricle. The apex or fastigium is the extension of the ventricle up into the cerebellum. The floor of the fourth ventricle
is named the rhomboid fossa. The lateral recess is an extension of the ventricle on the dorsal inferior cerebellar
peduncle.
Inferiorly, it extends into the central canal of medulla. The fourth ventricle communicates with the subarachnoid space through the lateral foramen of Luschka, located near the flocculus of the cerebellum, and through the
median foramen of Magendie, located in the roof of the ventricle. Most of the CSF outflow passes through the
medial foramen. The cerebral aqueduct contains no choroid plexus. The tela choroidea of the fourth ventricle,
which is supplied by branches of the posterior inferior cerebellar arteries, is located in the posterior medullary
velum.11,12
The lateral wall of fourth ventricle on the upper side is formed by superior cerebellar peduncle and lower part
is formed by inferior cerebellar peduncle and gracile and cuneate tubercle. The roof is tent in shape and projected
into cerebellum (Fig. 1.13). Roof is formed superiorly by superior cerebellar peduncle and superior medullary
velum and inferiorly by membrane consisting of ependymal and double layer of pia meter which constitute tela
choroidea of the fourth ventricle. Floor of the fourth ventricle is rhomboid in shape and thus called as rhomboid
fossa. Upper triangular part is formed by pons and lower triangular part by medulla. Intermediate part prolonged
laterally to form the lateral recess. The floor of the fourth ventricle is divided into two symmetrical halves. Each
half contains facial colliculus, hypoglossal triangle, sulcus limitans, vestibular area, stria medullaris, and vagal
triangle. The vital centers are situated in vagal triangle and injury during surgery into the fourth ventricle to these
areas can be fatal.
Limbic System
The limbic system (or paleomammalian brain) is a complex set of brain structures that involves with learning,
memory, and emotion. It is affected in many neuropsychiatric diseases including schizophrenia, Alzheimer disease,
and some forms of epilepsy.
It is a collection of structures from the telencephalon, diencephalon, and mesencephalon.13
The limbic system includes the olfactory bulbs, hippocampus, amygdala, anterior thalamic nuclei, fornix, and
column of fornix, mammillary body, septum pellucidum, habenular commissure, cingulate gyrus, parahippocampal
gyrus, limbic cortex, limbic midbrain areas, and pons (Fig. 1.14).
The limbic lobe is a ring of cortex on the medial aspect of the cerebral hemisphere (Fig. 1.14). This ring of cortex
consists of the cingulate gyrus, parahippocampal gyrus, and septal cortex. These cortical areas are connected via
the cingulum (Fig. 1.12). The cortical areas within the limbic lobe, together with certain adjacent deep structures,
are known as the limbic system. The areas that are usually included within the limbic system include the following:
1.
2.
3.
4.
5.
6.
Limbic lobe
Hippocampal formation and fornix
Amygdala
Septal area
Mammillary bodies (or in some accounts, the entire hypothalamus)
Anterior nuclei of the thalamus
The fornix connects the hippocampus to the mammillary bodies, which in turn is connected to the anterior nuclei
of the thalamus by mammillothalamic tract. The anterior nuclei of thalamus are projected to the cingulate cortex.
From the cingulate cortex, these are connected to the hippocampus, completing a complex closed circuit. This circuit
was originally described by Papez and later named as Papez circuit.
The complexity of the behavioral responses presumably explains the complexity of the limbic system. Connections
with sensory, motor, and autonomic systems are required. The presence of these connections may give rise to misleading results when different parts of the limbic system are stimulated electrically in an attempt to discern their
I. NEUROANATOMY
ANATOMY OF BRAIN
23
functions. For example, stimulation of most components of the limbic system produces autonomic effects such as
changes in blood pressure and respiration. Similarly, movement can be obtained from stimulation at many points.
This does not mean that the limbic system is primarily involved with autonomic control and movement, but, rather,
that it has connections with the hypothalamus and motor areas of the brain for integrating the output of these
systems in whatever ways are necessary for the production of visceral or emotional behavioral patterns. This also
explains why emotions and visceral sensations have a strong effect on the learning process. It is therefore not too
surprising that the part of the brain that appears to control our emotions and regulate visceral functions also plays a
central role in learning and memory.
Midbrain (Mesencephalon)
The midbrain or mesencephalon (from the Greek mesos—middle, and enkephalos—brain14) is a portion of the
CNS that connects hindbrain with forebrain and associated with vision, hearing, motor control, sleep/wake, arousal
(alertness), and temperature regulation.15
Midbrain passes through the tentorial notch and is related to each side to parahippocampal gyri, optic tract,
trocheal nerve, and posterior cerebral artery and geniculate bodies (Figs. 1.15–1.17). Interpeduncular structures are
anterior to it whereas great cerebral vein, pineal body, splenium of corpus callosum, and pulvinar are posterior to it
(Figs. 1.4 and 1.17).
The part that lies posterior to cerebral aqueduct is called the tectum. It consists of superior and inferior colliculi of the two sides (Fig. 1.18). The part anterior to cerebral aqueduct is made up of the right and left halves
called cerebral peduncles. Each peduncle consists of three parts: (from anterior to posterior side) crus cerebri (or
basal peduncle), the substantia nigra, and tegmentum (Fig. 1.18). The crus cerebri consists of vertically running
a large mass of fibers. These fibers descend from the cerebral cortex, some reach to the pons while other extents
FIGURE 1.15 Lateral view of the brain stem. 1. Medial geniculate body. 2. Lateral geniculate body. 3. Optic tract. 4. Dorsal part (mesencephalic tegmentum). 5. Ventral part (Crus cerebri). 6. Mammillary body. 7. Infundibulum. 8. Hypophysis. 9. Trigeminal nerve. 10. Pons. 11.
Abducens nerve. 12. Olive. 13. Pyramids (medulla oblongata). 14. Hypoglossal nerve. 15. Ventrolateral sulcus. 16. Ventral root of first cervical
nerve. 17. Spinal root of accessory nerve. 18. Dorsal root of first cervical nerve (retracted). 19. Dorsolateral sulcus (medulla oblongata). 20. Cranial
root of accessory and vagus nerve. 21. Tenia of the fourth ventricle. 22. Glossopharyngeal and vagus nerve. 23. Middle cerebellar peduncle.
24. Facial nerve with nervous intermedius and vestibulocochlear nerve. 25. Inferior cerebellar peduncle. 26. Superior cerebellar peduncle. 27.
Trochlear nerve. 28. Inferior colliculus and brachium of inferior colliculus. 29. Superior colliculus. 30. Pulvinar.
I. NEUROANATOMY
24
1. NEUROANATOMY
FIGURE 1.16 Dorsal view of brain stem. (Neocortex and cerebellum has been removed.) 1. Caudate nucleus. 2. Lamina affixa. 3. Terminal
stria and superior thalmostriate vein in terminal sulcus. 4. Tenia choroidea. 5. Pulvinar (thalamus). 6. Habenular trigone. 7. Pineal body. 8–11.
Mesencephalon. 8. Brachium of superior colliculus. 12. Brachium of inferior colliculus. 9. 10. Tectum. 9. Superior colliculus. 10. Inferior colliculus. 11. Superior medullary velum. 13. Trochlear nerve. 14. Superior cerebellar peduncle. 15. Median eminences. 16. Facial colliculus. 17. Middle
cerebellar peduncle. 18. Inferior cerebellar peduncle. 19. Stria medullaris (fourth ventricle) and lateral recess of the fourth ventricle. 20. Tenia
of the fourth ventricle. 21. Trigone of hypoglossal nerve. 22. Trigone of vagus nerve (ala cinerea). 23. Obex. 24. Dorsal intermediate sulcus. 25.
Dorsolateral sulcus. 26. Dorsal median sulcus. 27. Lateral funiculus. 28. Fasciculus gracilis. 29. Fasciculus cuneatus. 30. Tuberculum gracile. 31.
Tuberculum cuneatum. 32. Vestibular area. 33. Median sulcus. 34. Sulcus limitans. 35. Cerebral peduncle. 36. Lateral geniculate body. 37. Medial
geniculate body.
to the spinal cord. The two crura are separated by a notch on anterior aspect of midbrain. The substantia nigra
consists of pigmented gray matter and therefore appears dark. The tegmentums of the two sides are continuous
across the midline and contain important mass of gray matter and fiber bundle. The red nucleus is the largest of
the nuclei of midbrain and lies in upper half of midbrain (Fig. 1.18). The tegmentum also contains the reticular
formation which is continuous below with that of the pons and medulla. The medial lemniscus is a fiber bundle
of tegmentums that lie behind the substantia nigra and lateral to red nucleus. In the lower part of tegmentum,
the fibers of superior cerebellar peduncles decussate before ending into red nucleus. Trochlear nucleus lies in
ventral part of midbrain; its fibers run dorsally and decussate before emerging from dorsal surface brain stem
(Figs. 1.15–1.17).
Mesencephalic nucleus of trigeminal nerve lies in the lateral part of gray matter and receives proprioceptive
impulses from the muscles of mastication, face, facial muscles, ocular muscle and from teeth (Fig. 1.18). Superior colliculus receives afferents from retina (visual), spinal cord (tactile), inferior colliculus (auditory), and occipital cortex
(modulating pathway). Efferent goes to retina, spinal cord, brain stem nuclei, and tegmentum. Inferior colliculus
receives afferent impulses from lateral lemniscus and efferent to medial geniculate body. It controls auditory reflex
and helps in localizing the source of sound.
Red nucleus lies in anterior part of tegmentum dorsomedial to substantia nigra (Fig. 1.20). It receives afferent from
superior cerebellar peduncle, globus pallidus, subthalamic nucleus, and cerebral cortex. It supplies efferent to spinal
cord as rubrospinal tract, reticular formation, thalamus, subthalamic nucleus, and olivary nucleus. It has inhibitory
influence over muscle tone.
Oculomotor nucleus of two sides closes together, forms a single complex. The Edinger–Westphal nucleus (supplies the sphincter pupillae and ciliaris muscles) forms part of oculomotor complex.
I. NEUROANATOMY
ANATOMY OF BRAIN
25
FIGURE 1.17 Ventral view of brain stem comprising of midbrain, pons, and medulla oblongata. 1. Corpus callosum in depths of anterior
interhemispheric or longitudinal cerebral fissure. 2. Olfactory bulb. 3. Olfactory tract. 4. Medial olfactory stria. 5. Lateral olfactory stria. 6. Olfactory
trigone. 7. Anterior perforated substance. 8. Diagonal band of Broca. 9. Optic tract. 10. Cut surface of left temporal lobe. 11. Infundibulum with
hypophyseal stalk. 12. Mammillary body. 13. Interpeduncular fossa with interpeduncular perforated substance. 14. Ventral part of cerebral peduncle. 15. Pons. 16. Basilar sulcus. 17. Pyramid (medulla oblongata). 18. Middle cerebellar peduncle. 19. Olive. 20. Ventrolateral sulcus. 21. Ventral
root of first cervical nerve. 22. Ventral median sulcus. 23. Decussation of pyramids. 24. Hypoglossal nerve. 25. Spinal root of accessory nerve. 26.
Accessory nerve and cranial root. 27. Glossopharyngeal and vagus nerve. 28. Facial nerve with nervous intermedius and vestibulocochlear nerve.
29. Abducens nerve. 30. Motor and sensory root of trigeminal nerve. 31. Trochlear nerve. 32. Oculomotor nerve. 33. Optic chiasm.
Pons
Pons is the forepart of the hindbrain situated in front of the cerebellum. Pons is continuous behind and below with
the medulla oblongata. Pons is separated from medulla in front by a furrow in which the abducent, facial, and acoustic
nerves appear (Figs. 1.15 and 1.17). Trigeminal nerves emerge at the junction of pons and middle cerebellar peduncle.
Anterior surface of pons is convex and has a sulcus in midline called basilar sulcus which is occupied by basilar artery
(Fig. 1.17). Superior surface of pons is related to superior cerebellar artery and lower surface is related to anterior inferior
cerebellar artery. Posterior surface of pons is hidden by cerebellum and bounded laterally by superior cerebellar peduncle (Fig. 1.17). On either side of the lower part of the pons there is a region called cerebellopontine angle. The facial,
vestibulocochlear, glossopharyngeal nerves; nervous intermedius; and labyrinthine arteries lie in this region (Fig. 1.19).
On transverse section, pons can be divided into two parts: ventral part and dorsal part (Fig. 1.20). Ventral part
of pons contains (1) the pontine nuclei, (2) vertically running corticopontine and corticospinal fibers, and (3) transversely running arising from pontine nuclei and projecting to opposite half of cerebellum through middle cerebellar
peduncle. The bulk of ventral part is due to pontine nuclei and their connection (corticopontocerebellar) (Fig. 1.20).
Dorsal part (tegmentum) of pons is occupied mainly by reticular formation. The dorsal part is bounded laterally
by the inferior cerebellar peduncle in the lower part of the pons and by the superior cerebellar peduncle in the upper
part (Fig. 1.20). Structures present at the level of upper pons are medial longitudinal fasciculus, cerebellar peduncle,
locus ceruleus, parabrachial nucleus, and pediculopontine nucleus (Fig. 1.20). Structures present at the level of middle cerebellar peduncle are medial lemniscus, lateral lemniscus, trapezoid body, and trigeminal nucleus (sensory and
motor). Structures present at the level of facial nucleus are cranial nerve (CN) VI nucleus (abducens nerve), CN VII
nucleus (facial nerve), and CN VIII (vestibular nuclei) (Figs. 1.15 and 1.17).
I. NEUROANATOMY
26
1. NEUROANATOMY
FIGURE 1.18 Transverse section through the midbrain at the level of superior colliculi.
FIGURE 1.19 Schematic representation of left cerebellopontine angle (CP angle) as in a suboccipital approach showing various nerves and
PICA. CN, cranial nerve; PICA, posterior inferior cerebellar artery.
Medulla
Medulla is the lowest part of the brain stem and with other part of hindbrain occupies infratentorial space of the
skull. It is the direct and expanded upward continuation of spinal cord and includes important fiber tracts (Fig. 1.4).
The medulla oblongata extends from the lower margin of the pons to a plane passing transversely below the pyramidal decussation and above the first pair of cervical nerves. This plane corresponds with the foramen magnum, i.e.,
I. NEUROANATOMY
ANATOMY OF BRAIN
27
FIGURE 1.20 Transverse section through the mid-pons.
upper border of the atlas behind and the middle of the odontoid process of the axis in front. Anteriorly it is related
with clivus and meninges; posteriorly, to the vallecula of the cerebellum.
Its caudal part is like the spinal cord, while its cranial half is split open to form lower part of the floor of
the fourth ventricle. Thus medulla is often thought of as being in two parts: open and closed. An open part or
superior part where the dorsal surface of the medulla is formed by the fourth ventricle (Fig. 1.21) whereas a
closed part or inferior part where the central canal lies within the medulla. The bulbopontine sulcus separates
oblongata and pons ventrally; the sixth, seventh, and eighth CNs arise from the bulbopontine sulcus (Figs. 1.15
and 1.17).
Two median fissures (anterior and posterior) divide medulla into two symmetrical halves (Fig. 1.21). Anterior
median fissure is crossed by pyramidal decussation. Each half of medulla is marked by two longitudinal sulci,
anterolateral sulci between pyramids and olive, and posterolateral between olive and inferior cerebellar peduncle.
Thus these two sulci divide each half of medulla into three regions: anterior, lateral, and posterior region.
Anterolateral sulcus lies in line with ventral root of spinal nerves and the 12th CN emerges from it (Fig. 1.21).
Posterolateral sulcus lies in line with dorsal root of spinal nerve and gives attachment to the 11th, 10th, and 9th CNs.
The region (anterior region) between the anterior median sulcus and the anterolateral sulcus is occupied by an elevation on either side known as the pyramid (Fig. 1.21). This elevation is caused by corticospinal tract. In the lower part
of the medulla some of these fibers cross each other thus obliterating the anterior median fissure. This is known as
the decussation of the pyramids. Some other fibers that originate from the anterior median fissure above the decussation of the pyramids and run laterally across the surface of the pons are known as the external arcuate fibers. The
region (lateral region) between the anterolateral and posterolateral sulci in the upper part of the medulla is marked
by a swelling known as the olivary body. It is caused by a large mass of gray matter known as the inferior olivary
nucleus (Fig. 1.21). The posterior part (posterior region) of the medulla between the posterior median sulcus and the
posterolateral sulcus contains tracts that enter it from the posterior funiculus of the spinal cord. These are the fasciculus gracilis, lying medially next to the midline, and the fasciculus cuneatus, lying laterally (Fig. 1.21). These fasciculi
end in rounded elevations known as the gracile and the cuneate tubercles. They are caused by masses of gray matter
known as the nucleus gracilis and the nucleus cuneatus. Just above the tubercles, the posterior aspect of the medulla
is occupied by a triangular fossa, which forms the lower part of the floor of the fourth ventricle (Fig. 1.21). The fossa
is bounded on either side by the inferior cerebellar peduncle, which connects the medulla to the cerebellum.
The lower part of the medulla, immediately lateral to the fasciculus cuneatus, is marked by another longitudinal
elevation known as tuberculum cinereum. It is caused by an underlying collection of gray matter known as the spinal
nucleus of the trigeminal nerve (Fig. 1.21). The gray matter of this nucleus is covered by a layer of nerve fibers that
I. NEUROANATOMY
28
1. NEUROANATOMY
FIGURE 1.21 Transverse section of the medulla oblongata (lower portion).
TABLE 1.1 Medulla Oblongata at Different Levels
Level
Cavity
Nuclei
Motor Tract
Sensory Tract
Junction of pons and Fourth ventricle
medulla
Lateral vestibular nucleus,
cochlear nucleus
Pyramids
Medial longitudinal fasciculus, tectospinal
tract, medial lemniscus, lateral spinothalamic
tract, anterior spinothalamic tract, spinal tract
of the fifth cranial nerve
Olives, inferior
cerebellar peduncles
Fourth ventricle
Inferior olivary nucleus,
Pyramids
spinal nucleus of 5th nerve,
nucleus of the 8th–11th
nerves, nucleus ambiguous,
nucleus solitarius
Medial longitudinal fasciculus, tectospinal
tract, medial lemniscus, lateral spinothalamic
tract, anterior spinothalamic tract, spinal tract
of the fifth cranial nerve
Decussation of
medial lemniscus
Central canal
Nucleus gracilis and
cuneatus, spinal nucleus of
the fifth nerve, accessory
nucleus, hypoglossal
nucleus
Pyramids
Decussation of medial lemniscus, fasciculus
gracilis, and cuneatus, spinal tract of the fifth
nerve, posterior and lateral spinothalamic tract,
anterior spinocerebellar tract
Decussation of
pyramids
Central canal
Nucleus gracilis and
cuneatus, spinal nucleus of
the fifth nerve, accessory
nucleus
Decussation of
Spinal tract of the fifth nerve, posterior
corticospinal tracts and lateral spinothalamic tract, anterior
spinocerebellar tract
form the spinal tract of the trigeminal nerve. The base of the medulla is defined by the commissural fibers; crossing
over from the ipsilateral side in the spinal cord to the contralateral side in the brain stem. Below this lies the spinal cord.
The medulla oblongata controls autonomic functions and relays nerve signals between the brain and spinal cord.
It is also responsible for controlling several major autonomic functions of the body such as respiration via dorsal
respiratory nucleus and ventral respiratory nucleus, vital reflex arcs, vomiting, swallowing, reflexes, etc. (Table 1.1).
I. NEUROANATOMY
ANATOMY OF BRAIN
29
Lateral medullary syndrome, or “Wallenberg’s syndrome.” The most commonly affected artery is the vertebral
artery, followed by the posterior inferior cerebellar artery (PICA), superior middle and inferior medullary arteries.20
The spinothalamic tract is damaged, resulting in loss of pain and temperature sensation to the opposite side of the
body. There is sensory deficit affecting the trunk and extremities on the opposite side of the infarction and sensory
deficits affecting the face and CNs on the same side. The damage to the cerebellum or the inferior cerebellar peduncle
can cause ataxia. Other clinical symptoms and findings are swallowing difficulties (dysphagia) slurred speech, facial
pain, vertigo, nystagmus, Horner syndrome, diplopia, and possibly palatal myoclonus.
Medial medullary syndrome. It results from occlusion of the vertebral artery or of a branch of the vertebral or
lower basilar artery.21 This results in the infarction of medial part of the medulla oblongata. The infarction leads to
infarction of the ipsilateral medullary pyramid, the ipsilateral medial lemniscus, and hypoglossal nerve fibers that
pass through the medulla. It results into contralateral hemiparesis sparing the face, hemisensory loss of the posterior
column type (contralateral), and deviation of the tongue.
Reticular Formation
In addition to the distinct nuclei, much of the brain stem consists of small bunches of neuronal cell bodies (gray
matter) interspersed among small bundle of myelinated axons (white matter). The broad region where gray matter
and white matter form a network-like arrangement is known as reticular formation. Reticular formation network
extends from the upper part of spinal cord, throughout the brain stem and into the lower part of the diencephalon.
Reticular formation have both ascending (sensory) and descending (motor) fibers. Part of the reticular formation
called the reticular activating system consists of sensory axons that project to sensory cortex. The reticular activating
system helps in maintaining consciousness and activates awakening from sleep.21 The reticular formation descending tracts help in regulating posture and muscle tone.22, 23
Cerebellum
The term cerebellum literally means “little brain.” It is located dorsal to the brain stem and is connected to the
brain stem by three pairs of cerebellar peduncles. The cerebellum consists of primarily of white matter surrounded
by a thin layer of gray matter (cerebellar cortex) and four pairs of deep nuclei.
Cerebellum has three surfaces: tentorial, suboccipital, and petrosal. Superior surface or tentorial surface is convex
and related superiorly to tentorium cerebelli, the suboccipital surface is in relation to squamous pat of occipital bone,
and petrosal surface is related anteriorly to petrous part of the temporal bone. The fourth ventricle is intimately
in relation with the cerebellum. The fourth ventricle is a tent-shaped midline structure surrounded by vermian of
cerebellum.
Morphologically cerebellum consists of three parts: a part lying in midline called vermis and of two large lateral
cerebellar hemispheres. Cerebellum (both vermis and hemisphere) is divided by fissure and sulci, into lobules. Thus,
parts of cerebellar hemisphere represent lateral extension of vermis (except the lingual which has no extension). The
connections of the cerebellum are grouped into three cerebellar peduncles which are named according to their position. (1) Inferior cerebellar peduncle connects the cerebellum with the medulla, contains afferent and efferent axons;
(2) middle cerebellar peduncle connects cerebellum with the pons, contains only afferent axons from pontine nuclei;
and (3) superior cerebellar peduncle connects cerebellum with the midbrain, it is predominantly efferent axons. Gray
matter of cerebellar cortex consists of four pairs of nuclei and all the output come from them. These are (1) nucleus
dentatus (of neocerebellum), (2) nucleus globosus, (3) nucleus emboliformis (of paleocerebellum), and (4) nucleus
fastigi (of archicerebellum).
From the functional viewpoint, the cerebellum represents three distinct regions: one is the vermis and the other
two regions are located in intermediate and lateral parts of the cerebellar hemisphere. The functional cerebellum can
be divided into three regions.
The smallest region, the flocculonodular lobe (Fig. 1.22) is the oldest part in evolutionary terms (archicerebellum)
and participates mainly in balance and spatial orientation; its primary connections are with the vestibular nuclei,
although it also receives visual and other sensory input. Damage to it causes disturbances of balance and gait.16
The medial zone of the anterior and posterior lobes (Fig. 1.22) constitutes the spinocerebellum, also known as
paleocerebellum. It receives proprioception input from the dorsal columns of the spinal cord (including the spinocerebellar tract) and from the trigeminal nerve, as well as from visual and auditory systems. It sends fibers to deep
cerebellar nuclei that, in turn, project to both the cerebral cortex and the brain stem, thus providing modulation of
descending motor systems thus control tone, posture, and crude movements.16
I. NEUROANATOMY
30
1. NEUROANATOMY
FIGURE 1.22 Schematic representation of the major anatomic subdivisions of the cerebellum.
The lateral zone (Fig. 1.22), which in humans is by far the largest part, constitutes the cerebrocerebellum, also
known as neocerebellum. It receives input exclusively from the cerebral cortex (especially the parietal lobe) via the
pontine nuclei (forming corticopontocerebellar pathways) and project mainly to the motor areas of the premotor
cortex and primary motor area of the cerebral cortex and to the red nucleus.16 It is involved in planning and mental
rehearsal of complex motor actions, conscious assessment of movement error17 in evaluating sensory information for
action,16 and in a number of purely cognitive functions.18,19
VASCULAR SUPPLY OF THE BRAIN
Arterial System
Brain is supplied by branches of two main pairs of arteries, i.e., internal carotid arteries and vertebral arteries. The
left carotid artery is branch of the aortic arch whereas; the right is branching from the brachiocephalic trunk. The
common carotid artery branches into the internal and external carotid arteries at the level of cervical vertebra 3–4.
The internal carotid artery enters the cranial cavity at the base of the skull through the carotid canal, then forms the
S shape siphon and passes through the sinus cavernous.
The internal carotid artery ends as a bifurcation into anterior and middle cerebral arteries. Ophthalmic artery is
the first branch of the internal carotid artery (Fig. 1.23). In addition, two small branches of internal carotid artery
are anterior choroidal artery and recurrent branch of anterior cerebral artery also called as artery of Heubner. The
anterior choroidal artery which arises directly forms the internal carotid artery, runs backward in relation to optic
tract, and enters the inferior horn of the lateral ventricle through choroid fissure (Fig. 1.23). Artery of Heubner runs
backward and laterally to enter the anterior perforated substances. Thrombosis of artery of Heubner results in contralateral paralysis of the face and upper extremity. Large branches of the internal carotid artery are anterior cerebral
artery, middle cerebral artery, posterior communicating artery.
The two vertebral arteries in their extravertebral course (after arising from subclavial artery) are in close relation
to the cervical vertebrae. The vertebral arteries run in the transverse foramen of the cervical vertebrae C6 and above.
Vertebral arteries ascend on the anterolateral aspect of medulla and unit at the lower border of pons to form basilar
artery. Basilar artery is then divided into two posterior cerebral arteries at the upper end of the pons (Fig. 1.23). They
are connected to the carotid field by joining a posterior communicating artery.
The internal carotid artery and vertibrobasilar system are connected by the posterior communicating arteries. The
anterior communicating artery connects both anterior cerebral arteries. As a result of these anastomoses an arterial
ring, the circulus arteriosus (or circle of Willis), is formed in relation to the base of the brain, and through this circle
it is possible to compensate the obliteration of any mentioned artery (Fig. 1.23).
The anterior, middle, and posterior cerebral arteries give rise to two sets of branches: cortical and central. The
cortical branches divide on the surface of the cerebral hemisphere and supply the cortex. The central (or perforating)
branches pass deep into the substance of the cerebral hemisphere to supply white matter and masses of gray matter.
I. NEUROANATOMY
VASCULAR SUPPLY OF THE BRAIN
31
FIGURE 1.23 Circle of Willis (viewed from below the brain). a., artery; A-comm a., anterior communicating artery; ACAs, anterior cerebral
arteries; AICA, anterior inferior cerebellar artery; Cr., cranial nerve; ICA, internal carotid artery; MCA, middle cerebral artery; n., nerve; P-comm a.,
posterior communicating artery; PCA, posterior cerebral artery; PICA, posterior inferior cerebellar artery.
The central branches are end arteries. The amount of blood supply is more in the gray matter than the white matter;
this is related to the intensity of the metabolic rate.
Arterial supply to important structure of brain.
Middle cerebral artery supplies greater part of superiolateral surface of the cerebral cortex. A band 0.5–1 inch
wide extended from frontal pole to the parietooccipital sulcus is supplied by anterior cerebral artery. The area
of occipital lobe is supplied by the posterior cerebral artery. The inferior temporal gyrus is also supplied by the
posterior cerebral artery (Fig. 1.24).
Medial surface of the cerebral cortex is mainly supplied by anterior cerebral artery except occipital lobe which is
supplied by posterior cerebral artery (Fig. 1.24).
Orbital surface on the lateral part is supplied by middle cerebral artery and medial part by the anterior cerebral
artery. Tentorial surface is supplied by the posterior cerebral artery. The temporal pole is supplied by middle cerebral
artery (Fig. 1.24).
Thrombosis of the anterior cerebral artery causes paralysis of the muscles of the leg and foot of the opposite side,
loss of the sensation from the leg and foot of the opposite side, and personality changes.
Thrombosis of the middle cerebral artery causes hemiplegia and loss of the sensation of the opposite side of the
body mainly face and arm, homonymous hemianopia of opposite side, and aphasia (by involvement of the Broca’s
and Wernickes’s area) specially in left side thrombosis in right-handed person.
Thrombosis of the posterior cerebral artery causes visual disturbance mainly homonymous hemianopia of the
opposite side.
Choroid plexuses of the lateral and the third ventricles are formed by anterior choroidal (branch of internal carotid)
and posterior choroidal artery (branch of posterior cerebral artery). The choroid plexus of the fourth ventricle is
formed by a branch from the posterior inferior cerebellar artery.
Internal capsule is supplied by central branches of middle cerebral artery, the lenticulostriate branches, anterior
cerebral artery, the Huebner recurrent branch, posterior communicating artery, and anterior choroidal artery.
Thalamus is supplied mainly by perforating branches of the posterior cerebral artery (posteromedial) and partly
anteromedial central branches (Fig. 1.24).
I. NEUROANATOMY
32
1. NEUROANATOMY
FIGURE 1.24 Vascular supply of cerebral hemispheres. AChA, anterior choroidal artery; MCA, middle cerebral artery; P-comm a., posterior
communicating artery; RAH, recurrent artery of Heubner.
Hypothalamus is supplied by central branches of anterior medial group and posterior medial group arising from
posterior cerebral and posterior communicating arteries.
Corpus striatum (caudate and putamen) is mainly supplied by anterolateral central branches of the middle cerebral artery and partly by the anteromedial central branches from the anterior and anterior communicating artery. The
main supply of the globus pallidus is from the anterior choroidal artery.
Medulla is supplied by the various branches of vertebral artery including anterior and posterior spinal artery,
posterior inferior cerebellar artery, and small direct branches.
Pons is supplied by the branches from the basilar artery (paramedian branches and short and long circumferential
branches) (Fig. 1.23).Midbrain is supplied by branches from basilar artery (posterior cerebral, superior cerebellar
arteries, and direct branch from basilar artery).
Cerebellum is supplied by superior cerebellar, anterior inferior cerebellar of basilar artery, and posterior cerebellar
branches of vertebral artery.
Cerebral Venous System
Venous system of the brain can be divided into a superficial system and a deep system. The superficial system
comprises of cortical veins and sagittal sinuses (Fig. 1.25). These drain superficial surfaces of both cerebral hemispheres. The superficial cerebral veins can be divided into three groups.24 A mediodorsal group draining into superior sagittal sinus and the straight sinus; a lateroventral group draining into the lateral sinus; and an anterior group
draining into the cavernous sinus. The veins of the posterior fossa may again be divided into three groups: (1) superior group draining into the galenic system, (2) anterior group draining into petrosal sinus, and (3) posterior group
draining into the torcular. The superior sagittal sinus drains major part of the cerebral hemispheres (Fig. 1.25). The
cavernous sinuses drain blood from the orbits, the inferior parts of the frontal and parietal lobe, and the superior and
inferior petrosal sinuses. Blood from them flow into the internal jugular veins.
The inferior sagittal sinus runs in the free edge of falx cerebri and unites with the vein of Galen to form the straight
sinus. Straight sinus runs backward in the center of the tentorium cerebelli at the attachment of the falx cerebri,
emptying into the torcular Herophili at the internal occipital protuberance (Fig. 1.25). The lateral sinuses extend from
torcular Herophili to jugular bulbs and consist of a transverse and sigmoid portion. They receive blood from the
cerebellum, the brain stem, and posterior parts of the hemisphere.
The deep cerebral veins are more important than superficial veins from the angiographic point of view.25 The
deep system comprises of lateral sinus, straight sinus, and sigmoid sinus along with draining deeper cortical veins.
I. NEUROANATOMY
THE MENINGES AND CEREBROSPINAL FLUID
33
FIGURE 1.25 Dural sinus tributaries from the cerebral veins (right lateral view). Venous blood collected deep within the brain drains to the
dural sinuses through superficial and deep cerebral vein. The red arrows in the diagram show the principal directions of venous blood flow in the
major sinuses.
Three veins (choroid vein, septal vein, and thalamostriate vein) unite just behind the interventricular foramen of
Monro to form the internal cerebral vein. The internal cerebral veins of each side run posteriorly in the roof of the
third ventricle and unite beneath the splenium of the corpus callosum to form the great cerebral vein. The great
cerebral vein of Galen is a short (1–2 cm long), thick vein that passes posterosuperiorly behind the splenium of corpus callosum in the quadrigeminal cistern. It receives the basal veins and the posterior fossa veins and drains to the
anterior end of the straight sinus where this unites with the inferior sagittal sinus (Fig. 1.25).
Both these venous systems mostly drain into internal jugular veins (Fig. 1.25). The cerebral veins and sinuses neither have valves nor tunica muscularis. Because they lack valves, blood flow is possible in different directions. The
cortical veins are linked by numerous anastomosis, allowing the development of a collateral circulation and probably explaining the good prognosis of some cerebral venous thrombosis. Lack of tunica muscularis permits veins
to remain dilated and prone for venous air embolism during intracranial surgery. The dural sinuses especially the
superior sagittal sinus contains most of the arachnoid villi and granulations, in which absorption of CSF takes place.
So dural sinus thrombosis blocks villi and leads to intracranial hypertension and papilledema.
THE MENINGES AND CEREBROSPINAL FLUID
The Meninges
The meninges consists of (from outside to inside) dura mater, arachnoid mater, and pia mater.
Dura Mater
It consists of two layers over the brain, named periosteal and meningeal or investing. The two layers are fused to
each other all over, except where the cranial venous sinuses are enclosed between them.
The outer periosteal layer: The outer periosteal layer is the periosteum of the inner surface of the skull. It is continuous with the pericranium through the suture and foramina. It is firmly adhered over base, less over the vault
except at the suture, where it is attached with pericranium by suture membrane. It ends at foramen magnum. It provides sheath for CNs. Owing to its firm fixation to the base of the skull, it usually is torn in the skull base fracture.
Since it forms the wall of basal venous sinuses, a fracture of skull base is often associated with bleeding from ear,
nose, or pharynx.
I. NEUROANATOMY
34
1. NEUROANATOMY
Meningeal layer: This lies close to the brain. It forms four folds by projecting inward to form the folds and thus compartmentalized the cranial cavity into many freely communicating compartments which lodge different parts of brain.
The large midline fold separates the two hemispheres and is called the falx. A smaller fold separates the cerebral hemispheres from the cerebellum and is known as the tentorium cerebelli. Where the edges of the falx and tentorium meet
the skull, the dura mater encloses large venous sinuses that are responsible for draining venous blood from the brain.26
Falx cerebri: This is a sickle shaped reduplication which intervenes between medial surfaces of two cerebral hemispheres. It contains three venous sinuses: (1) the superior sagittal along its upper border; (2) the inferior sagittal along
its lower free border; (3) the straight sinus along its line of attachment to the tentorium cerebelli.
Tentorium cerebelli: This is a semilunar reduplication of the dura mater that separates cerebellum from occipital
lobes of cerebrum. It therefore forms the roof of posterior fossa of cranium. Its outer convex border is attached to the
lips of the transverse sinuses on the occipital bone, mastoid angle of parietal bone, and the superior border of petrous
part of temporal bone; and end by attaching posterior clinoid process. It is attached anteriorly to the anterior clinoid
process. This border bound to an oval space which is occupied by midbrain. The transverse sinuses lie between the
two layers of the tentorium along its posterior border. Superior petrosal sinus lies along its anterior border.
Falx cerebelli: It is a small sickle-shaped fold intervening posteriorly between the two halves of cerebellum. Its
base is attached to the posterior part of inferior surface of the tentorium cerebelli. Anterior margin is concave and free
whereas posterior margin is convex and contains the occipital sinus.
Diaphragm sella: It is a fold of dura mater which forms the roof of the pituitary fossa. It has a central opening for
emergence of stalk of the pituitary. It encloses intercavernous sinuses.
The Spinal Cord
The spinal cord is a long cylindrical lower part of CNS confined within the vertebral canal. Spinal cord extends
from the level of foramen magnum to end (by conus medullaris) at the first lumbar. So, the rest of lumbar, sacral, and
coccygeal part of vertebral canal are free from spinal cord, i.e., the spinal cord is shorter than vertebral canal. The
spinal cord itself has thick cervical region (origin of brachial plexus), thin thoracic, and thick lumbosacral (origin of
lumbosacral plexus). The lowest part of the spinal cord is conical called conus medullaris. Spinal cord below become
fibrous and is called filum terminalis.
The spinal cord is developed from the caudal cylindrical part of the neural tube. During intrauterine life, the cord
fills the whole length of the vertebral canal. At birth, lower end of the cord is found at the level of the third lumbar
vertebrae (L3). In adulthood, the lower end of spinal cord recedes to the first lumbar vertebral (L1). Its adult length
is about 45 cm in males and 42 cm in females.
Spinal cord is divided into two halves by an anterior median fissure and a posterior median sulcus (Fig. 1.26).
The two anterolateral sulci and two posterolateral sulci further divide each halve. Inside the spinal cord, there is a
FIGURE 1.26 Spinal cord pathways.
I. NEUROANATOMY
35
THE MENINGES AND CEREBROSPINAL FLUID
central canal which contains fluid called CFS. The canal is surrounded by gray matter in the form of H-shaped horns.
So in each halves the gray matter has larger ventral mass (anterior or ventral) gray column or thin elongated posterior (dorsal) gray column (Fig. 1.26). In some part of the spinal cord a small lateral projection of gray matter is seen.
Therefore, there are six horns present in the spinal cord: two dorsal horns, two lateral horns, and two ventral horns
(Table 1.2). The gray matter of both halves is connected in the midline by a band of gray matter called gray commissure which is traversed by central canal.
Spinal cord gives attachment to series of the spinal nerves on each side. Each spinal nerve is formed by two roots:
ventral and dorsal root. Just proximal to the junction of two nerve roots, the dorsal root is marked by a swelling
called the dorsal nerve root ganglion or spinal ganglion. There are 31 pairs of spinal nerve (8 cervical, 12 thoracic, 5
lumbar, 5 sacral, 1 coccygeal). As a result of upward migration of cord the roots of the spinal nerve have to follow an
oblique downward course to reach appropriate intervertebral foramen. The oblique course and length of the roots is
most marked in the lower nerves, and many of these roots occupy the vertebral canal below the level of spinal cord.
These roots collectively constitute cauda equine.
The spinal cord is also surrounded by the meninges (dura mater, arachnoid mater, and pia mater) in the same way
as brain. The dura mater is a thick membrane whereas pia and arachnoid maters are thin. The space between pia
mater and arachnoid mater is called as subarachnoid space, which is filled by CSF.
The gray matter can also be divided into layers of axon termination, based on cytological criteria. This was first
done by the Swedish neuroanatomist Bror Rexed (1914–2002), who divided the gray matter in to laminae I–X. This
laminar architecture is especially well defined in the posterior (dorsal) horn, where primary sensory axon makes
synapses in specific layers. In the spinal cord, nerve cells are arranged into 10 laminae, which have different properties (Fig. 1.27). Some nuclei (columns of cell bodies) are present throughout the spinal cord gray matter, other nuclei
have more restricted segmental distributions (Table 1.3).
1.
2.
3.
4.
5.
Lamina I—at the tip of dorsal horns
Lamina II until VI—along dorsal horns
Lamina VII and VIII—at ventral horns
Lamina IX—at anterior part of ventral horns
Lamina X—around central canal
TABLE 1.2 Horns of the Spinal Cord
2 Dorsal Horns (Sensory Horns)
2 Ventral Horns (Motor Horns)
2 Lateral Horns (Autonomic Horns)
Position in
spinal cord
Along the whole segment of spinal
cord
Along the whole segment of spinal cord
Thoracic segment and lumbosacral
segments
Functions
Sensory functions
Motor functions
Autonomic functions
Nuclei
Receive exteroceptive and
proprioceptive.
The nuclei are:
1. Substantia gelatinosa of Rolandi
2. Main sensory nucleus
3. Nucleus dorsalis of Clarke
Supply skeletal muscle. The nuclei are:
1. Anteromedial nucleus
2. Anterolateral nucleus
3. Posteromedial nucleus
4. Posterolateral nucleus
5. Central nucleus
Supply visceral structures. The nuclei are:
1. Intermediomedial nucleus
2. Intermediolateral nucleus
FIGURE 1.27 Synaptic layers in the gray matter. (A) Cervical cord. (B) Thoracic cord. (C) Lumbar cord. Motor neurons are shown in red and
sensory neurons in blue.
I. NEUROANATOMY
36
1. NEUROANATOMY
In spite of highly organized and complexed structure, spinal nuclei play in the 3 most important basic functions
of individual: motor, sensory and autonomic control (Table 1.4).
Whole of the gray matter is surrounded by white matter. On each side, there are three columns separated by sensory
and motor horns. These are (1) dorsal column, (2) ventral column, and (3) lateral column. Through these columns, there
are nerve bundles called tracts running, which are classified into two groups: ascending and descending tracts (Table 1.5).
Ascending Tracts of Spinal Cord (Sensory Tracts)
Type of Ascending Tracts (Fig. 1.26)
1. Gracile and cuneate (posterior white column)
2. Spinothalamic
Lateral and ventral
3. Spinocerebellar
Posterior and ventral
Pathway of the Ascending Tract
Each sensory pathway, from body to the brain is made up of three principal neurons. The cell body of the first neuron is situated in the sensory ganglion of the spinal nerve and extends in the CNS to varying level for different sensibilities. The second neuron crosses to the opposite side and forms fiber bundles known as lemnisci that ascend up to the
thalamus. The third-order neuron extends from the thalamus to the sensory cortex, in the form of radiation (Box 1.4).
TABLE 1.3 Sensory and Motor Nuclei of Spinal Cord
Nuclei
Site
Functions
Substantia gelatinosa of Rolandi
At tip of sensory horn of all segments
For pain and temperature sensation
t G
ive first-order neuron of lateral spinothalamic tract.
Main sensory nucleus
(nucleus proprius)
At middle of sensory nucleus in all segments
Receive crude and pressure sensation
t P
rojects first-order neuron of ventral spinothalamic
tract
Nucleus dorsalis
(Clark’s column)
At base of sensory horns of all thoracic segment
and upper 3 lumbar
Receive proprioceptive sensations from collateral branch
of gracile tract.
t S
tarts dorsal spinothalamic tract of same side
t S
tarts ventral spinothalamic tract of same and opposite
side
Lateral nucleus
(autonomic)
At lateral horn of all thoracic segment and
upper 3 lumbar, and appear again at sacral 2–4.
Autonomic (parasympathetic and sympathetic)
Ventromedial motor nucleus
At middle part of motor horns in all segment.
Effect axial musculature
Dorsal-medial motor nucleus
At thoracic and upper 3 lumbar
Supply axial muscle
Ventrolateral and dorsolateral
nuclei
Along lateral plane of motor horns in cervical
and lumbosacral on.
Supply axial muscle
Central motor
In cervical and lumbosacral motor
Supply axial muscle
TABLE 1.4 Functions of Spinal Cord
Sensory
t R
eceives superficial general sensations from skin and mucous membrane from all of the body except face and other body
organs
t S
uperficial external sensations is called exteroceptive sensations
t P
roprioceptive sensations receive deep types of sensation from tendons and muscles
Motor
t M
otor nuclei convey efferent fibers that pass through spinal nerves to control all muscles of body except muscles of head
and neck
Autonomic
t S
ympathetic nuclei are found at thoracolumbar region of spinal cord which control arrector pili muscle, vasomotor, and
dilates the pupil
t T
hey may join spinal or cranial nerves or may pass directly
t P
arasympathetic nuclei are located at sacral segments of spinal cord and control sphincters.
t T
hey give pelvic splanchnic nerve which carries parasympathetic outflow to derivatives of the hind gut
I. NEUROANATOMY
37
THE MENINGES AND CEREBROSPINAL FLUID
TABLE 1.5 Descending Tracts
Tract
Origin
Site and Course
End
Lateral corticospinal tract
Cortex (premotor and
sensory)
Posterior limb of internal
capsule, middle 2/3 of
crus cerebri, pyramid in
medulla, decussation at
medulla
Anterior gray column cells Fine-skilled motor activity,
(interneurons and lower
modulation of sensory
motor neurons)
function
Anterior corticospinal
tract
Cortex (premotor and
sensory)
Posterior limb of Internal
capsule, middle 2/3 of
crus cerebri, pyramid in
medulla, decussation at
spinal level they innervate
Anterior gray column cells Gross and postural motor
(interneurons and lower
motor neurons)
Corticonuclear
Cortex (premotor and
sensory)
Genu of internal capsule,
Cranial nerve nuclei
middle 2/3 of crus cerebri,
decussation at brainstem
Fine-skilled motor activity,
modulation of sensory
function
Rubrospinal tract
Red nucleus in midbrain
Descends into lateral
column of spinal cord just
ventral corticospinal tract
Facilitator to flexors of
opposite limbs
Tectospinal tract
Superior colliculus nuclei
Descends and crosses to
Cervical anterior horn cells Visuospinal reflex to move
locate on surface of ventral of opposite side
eyes and neck toward
column. It relays on
stimulus reflexly
anterior horn nuclei
Olivospinal tract
Inferior alivary nucleus in
medulla
Descends without crossing Cervical anterior horn cells Equilibrium and
of same side
proprioceptive
Medial vestibular spinal
tract
Medial, lateral, and inferior Into medial column
vestibular nuclei
of same side along
anterior median fissure
(sulcomarginal)
Anterior horn cells of
cervical and thoracic
regions of same side
Equilibrium
Lateral vestibular spinal
tract
Lateral vestibular nucleus
in pons
Anterior horn cells of all
segments of spinal cord of
same side
Equilibrium
Descending on same
side on surface of ventral
column of all spinal
segments
Anterior horn motor
nuclei of opposite side
Function
Lateral reticulospinal tract Reticular formation nuclei
in medulla of opposite side
Lateral column just medial Anterior horn cells of
to lateral corticospinal
opposite side and lateral
tract and in all segments of horn cells (autonomic)
spinal cord
Facilitatory to extensor
muscles through
its connection with
extrapyramidal center
(corpus striatum) and also
has pressor and depressor
effects on respiration
and circulation through
its connection with
hypothalamus
Medial reticulospinal
Reticular formation nuclei
of pons of same side
Descends on same side
along ventral white
column
Anterior horn cells all over
the cord of same side and
also lateral horn of same
side
Facilitatory to extensor
muscles through
its connection with
extrapyramidal center
(corpus striatum) and also
has pressor and depressor
effects on respiration
and circulation through
its connection with
hypothalamus (same side
like lateral reticulospinal
tract)
Medial longitudinal
fasciculus
Vestibular nucleus
Anterior column
Cervical and midthoracic
Coordination of head and
anterior column (extending eye movements
to upper point of brainstem)
I. NEUROANATOMY
38
1. NEUROANATOMY
BOX 1.4
O R G A N I Z A T I O N O F A S C E N D I N G P A T H W AY S
First-order neuron: dorsal root ganglion (spinal ganglion)
Second-order neuron: spinal cord
Third-order neuron: posterolateral ventral nucleus of
the thalamus
Termination: cerebral cortex “postcentral gyrus”
FIGURE 1.28 Schematic diagram of spinal cord arterial supply.
Function of Ascending Tract
1. Gracile and cuneate tracts: Discriminative touch, vibratory sense, and conscious muscle joint sense (sense of
position)
2. Lateral spinothalamic tract: Pain—Temperature
3. Anterior spinothalamic tract: Crude touch—pressure
4. Spinotectal tract: Provides afferent information for spinovisual reflexes and brings movements of the eyes and
head toward the source of the stimulation.
5. Spinoolivary tract: Carries unconscious proprioceptive and exteroceptive sensation.
6. Spinocerebellar tract (dorsal and ventral): Carries unconscious proprioceptive sensation.
7. Lissuar’s gelatinosa tract: Links the spinal segments.
Both the anterior and posterior spinal arteries are reinforced by the anastomotic arteries entering along the nerve
roots. These anastomotic arteries are the special importance at the level of T1 and T11 vertebrae and called the arteries of Adamkiewicz which correspond to the enlarged spinal cord (Fig. 1.28) (Table 1.6).
I. NEUROANATOMY
THE MENINGES AND CEREBROSPINAL FLUID
39
TABLE 1.6 Arterial Supply of the Spinal Cord (Figs. 1.28 and 1.29)
Arteries
Origin and Site
Course and Supply
Single anterior spinal
artery
From each vertebral artery
They unite forming single anterior spinal artery.
t S
upply anterior column and anterior horn
Two posterior spinal
arteries
From each vertebral artery
They did not unite.
t P
osterior arteries supply posterior column and posterior horn.
t T
he anterior artery shares in formation of arterial corona (supply
lateral column)
Lateral spinal arteries
From vertebral artery, ascending and
deep cervical, and descending aorta at
interventricular foramina
Each run along the spinal nerve trunk to divide into anterior and
posterior radicular arteries.
t T
hese arteries anastomos with arterial corona to supply lateral column.
FIGURE 1.29 Diagrammatic representation of blood supply of spinal cord at single level.
Veins Around the Surface of Spinal Cord
These six channels are freely connected with each other to encircle the spinal cord by what is called “Vena Corona.”
It drains interior of the cord. Then venous blood goes to epidural venous plexus. Obstruction of venous return
causes edema of spinal cord with subsequent paralysis.
Venous return of the spinal cord is through:
1.
2.
3.
4.
5.
One anterior median vein
One posterior median vein
Two anterior lateral veins
Two posterior lateral veins
Vena corona
Acknowledgment
I am thankful to Mr. Anil Kumar, a senior artist from SGPGIMS, Lucknow, for drawing all diagrams given in this chapter.
I. NEUROANATOMY
40
1. NEUROANATOMY
References
1. Tortora G, Derrickson B, Principle of Anatomy and Physiology: organization, support and movement, and Control system of Human body.
13th ed. Asia: John Wiley and Sons; 2009.
2. Ono M, Kubik S, Aberbathey CD. Atlas of the cerebral sulci. Stuttgart: Georg TheimeVerlag; 1990.
3. Chi JG, Dooling EC, Gilles FH. Gyral development of the human brain. Ann Neurol January 1977;1(1):86–93.
4. Kosslyn S. Cognitive psychology: mind and brain. New Jersey: Prentice Hall; 2007. p. 21. 194–9, 349.
5. http://www.ruf.rice.edu/∼lngbrain/cglidden/parietal.html.
6. SparkNotes: brain anatomy: parietal and occipital lobes. Archived from the original on 2007-12-31.
7. Loukas M, Pennell C, Groat C, Tubbs RS, Cohen-Gadol AA. Korbinian Brodmann (1868–1918) and his contributions to mapping the cerebral
cortex. Neurosurgery January 2011;68(1):6–11. discussion 11.
8. Garey LJ. Brodmann’s localisation in the cerebral cortex. New York: Springer; 2006.
9. Taylor I, Taylor MM, Psycholinguistics: Learning and using Language, Lincolnshire: Anybook Ltd; 1990, 362.
10. Stocco A, Lebiere C, Anderson JR. Conditional routing of information to the cortex: a model of the basal ganglia’s role in cognitive coordination. Psychol Rev April 2010;117(2):541–74.
11. FitzGerald MJT, Folan-Curran J. Clinical neuroanatomy and related neuroscience. 4th ed. Philadelphia (PA): WB Saunders; 2002.
12. Waxman SG. Ventricles and coverings of the brain. Correlative neuroanatomy. 24th ed. New York (NY): Lange Medical Books/McGraw-Hill; 2000.
p. 153–68.
13. Princeton Review. Anatomy coloring workbook. 2nd ed. The Princeton Review; July 29, 2003. p. 120.
14. Mosby’s medical, nursing and allied health dictionary. 4th ed. Mosby-Year Book; 1994. p. 981.
15. Breedlove W, Rosenzweig. Biological psychology. 6th ed. 2010. p. 45–6.
16. Ghez C, Fahn S. The cerebellum. In: Kandel ER, Schwartz JH, editors. Principles of neural science. 2nd ed. New York: Elsevier; 1985. p. 502–22.
17. Kingsley RE. Concise text of neuroscience. 2nd ed. Lippincott Williams and Wilkins; 2000.
18. Timmann D, Daum I. Cerebellar contributions to cognitive functions: a progress report after two decades of research. Cerebellum 2007;6(3):159–62.
19. Lenhoff HM, Wang PP, Greenberg F, Bellugi U. Williams syndrome and the brain. Sci Am December 1997;277(6):72.
20. Barkar AB. Cerebrovascular disease. IX. The medullary blood supply and the lateral medullary syndrome. Neurology October 1961;11:852–61.
21. Gan R, Noronha A. The medullary vascular syndromes revisited. J Neurol March 1995;242(4):195–202.
22. Olson DM, Graffagnino C. Consciousness, coma, and caring for the brain-injured patient. AACN Clin Issues October–December 2005;16(4):441–55.
23. Skinner RD, Homma Y, Garcia-Rill E. Arousal mechanisms related to posture and locomotion: 2. Ascending modulation. Prog Brain Res
2004;143:291–8. Review.
24. Einhaupl KM, Masuhr F. Cerebral venous and sinus thrombosis – an update. Eur J Neurol 1994;1:109–26.
25. Dora F, Zileli T. Common variations of the lateral and occipital sinuses at the confluence sinuum. Neuroradiology 1980;20:23–7.
26. Sinnatamby CS, Last RJ. Last’s anatomy: regional and applied. 11th ed. Churchill Livingston; 2006.
I. NEUROANATOMY
C H A P T E R
2
Neuroembryology
G.P. Singh
AIIMS, New Delhi, India
O U T L I N E
Formation of Zygote
41
Formation of Blastocyst
41
Formation of Embryonic or Germ Disc
42
Formation of Definitive Notochord
44
Development of Nervous System
Development of Brain
45
46
Prosencephalon
Mesencephalon
Rhombencephalon
46
47
47
Development of Spinal Cord
Development of Peripheral Nervous System
Spinal Nerve
Autonomic Nervous System
References
48
49
49
49
50
Embryology is a branch of science that is related to the formation, growth, and development of embryo. It deals
with the prenatal stage of development beginning from formation of gametes, fertilization, formation of zygote,
development of embryo and fetus to the birth of a new individual.1,2 Two basic processes involved during conversion of a single-celled zygote to a complex, multicellular organism are growth and differentiation. Growth occurs
by increase in cell number (cell division and multiplication) or cell size. On the other hand, cell differentiation is a
complicated process in which the cell acquires special characteristics to perform specific functions. These lead to the
formation of various tissues and organs assigned to perform specific functions.2
FORMATION OF ZYGOTE
The germ cells or gametes (sperm and ovum) are specialized haploid cells (with 23 unpaired chromosomes in human).
Fertilization results in union of the gametes (i.e., fusion of sperm with ovum, Fig. 2.1) to form an undifferentiated, mononucleated, diploid cell (with 23 pairs or 46 chromosomes) called zygote. The fertilization usually takes place in the ampulla
or lateral third of fallopian tube. After fertilization, the fertilized egg travels down the fallopian tube to reach the uterus.2,3
FORMATION OF BLASTOCYST
The single-celled zygote (Fig. 2.2) divides repeatedly by mitotic division thereby retaining the same number of chromosomes (i.e., 46 chromosomes) in each of the two daughter cells. The cells so formed are called blastomeres, and the process
of division is called cleavage (Fig. 2.2). Thus a single-celled zygote results in the formation of a mass of cell called morula
(16- to 32-celled stage). The inner cells of the morula called the inner cell mass gives rise to embryo proper, and the outer
layer of cells called the outer cell mass forms the covering of embryo and contributes to formation of placenta. As the cells of
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00002-6
41
© 2017 Elsevier Inc. All rights reserved.
42
2. NEUROEMBRYOLOGY
FIGURE 2.1 Fertilization of an ovum by sperm.
FIGURE 2.2 A single-celled zygote results in formation of morula (16- to 32-cell stage) and blastocele by the process of multiple cell division
called cleavage.
morula continue to divide, fluid from uterine cavity enters the intercellular spaces between the inner and outer cell mass.
Later the intercellular spaces fuse to form a single cavity called blastocele, and this stage of embryo is called blastocyst.2 The
cells of the inner cell mass are pushed to one side of blastocyst and this side of blastocyst is known as the embryonic pole.
The cells of the inner cell mass are called embryoblast cells. The cells of outer cell mass flatten and form the wall of blastocyst
and are called trophoblast cells (Fig. 2.2). The trophoblast cells covering the embryonic pole have the property to invade the
epithelial cells of uterine mucosa and thus get attached to uterus.
After fertilization in the fallopian tube, as the fertilized egg (zygote) divides repeatedly to form morula, it travels
down the fallopian tube to reach the uterine cavity. The morula reaches the uterine cavity on the third to fourth day
of fertilization. On day 5, blastocyst is formed, which adheres to uterine mucosa on the sixth day of fertilization and
gets implanted in the uterus.
FORMATION OF EMBRYONIC OR GERM DISC
During the second week of development, the cells of the inner cell mass (embryoblasts) differentiate and organize into
two epithelial layers—the inner layer of cuboidal cells or hypoblast on the ventral surface that faces the blastocyst cavity
and the outer layer of columnar cells or epiblast on the dorsal surface. These layers together form the bilaminar germ disc or
embryonic disc (Fig. 2.3).2,4,5 The trophoblasts start forming the placenta. Fluid begins to collect between the cells of outer
layer (epiblast cells) and the trophoblasts and forms a fluid-filled cavity known as amniotic cavity. The epiblast cells proliferate and migrate to line the roof of amniotic cavity. These cells are called amniogenic cells. Similarly, the hypoblast cells
facing the blastocyst cavity proliferate and migrate to line the blastocyst cavity to form the yolk sac (Fig. 2.3).5
I. NEUROANATOMY
FORMATION OF EMBRYONIC OR GERM DISC
43
FIGURE 2.3 Formation of bilaminar germ disc.
FIGURE 2.4 Germ disc or embryonic disc as seen from dorsal aspect after opening the amniotic cavity. (A) Appearance of primitive streak
and primitive node. (B) Growth of embryonic disc due to migration of cells from primitive streak. Formation of prechordal plate and notochordal
process.
FIGURE 2.5 Formation of trilaminar germ disc.
During the third week of embryonic development, bilaminar germ disc is converted to trilaminar germ disc
with the formation of the three primary germ layers—ectoderm, mesoderm, and endoderm.4,5 This process is called
gastrulation, which begins with the appearance of primitive streak (characterized by narrow median groove with
slight raised margins) on the outer surface (epiblast) of the embryonic disc. At the cranial end of this streak there is a
primitive node (Hensen’s node), the center of which presents a depression called primitive pit (Fig. 2.4A). The cells of
the epiblast migrate toward the primitive streak, get detached from the epiblast layer, and come to lie underneath it
(Fig. 2.5). This is called invagination. Some of these invaginated cells displace the hypoblast cells from the endoderm
I. NEUROANATOMY
44
2. NEUROEMBRYOLOGY
FIGURE 2.6 Formation of definitive notochord. (A) Sagittal section of the embryonic disc showing the notochordal process (hollow) between
the ectoderm and endoderm layers. The base of this process fuses with endoderm cells and then both disappear thus forming a communication
between the amniotic cavities and the yolk sac called the neurenteric canal. (B) Cross-section through the embryonic disc which shows roof of
opened notochordal process forming the notochordal plate. (C) Notochordal plate detaches from the endoderm to form the solid cord-like structure called definitive notochord (lying in the intraembryonic mesoderm).
while others migrate to occupy the space between the epiblast and hypoblast (endoderm) cell layers to form the third
germ layer—the intraembryonic or secondary mesoderm.2,5 The remaining cells of epiblast forms the ectoderm. The
three germ layers thus formed give rise to all the tissues and organs in the embryo.
FORMATION OF DEFINITIVE NOTOCHORD
At the cephalic end of the germ or embryonic disc, some of the endodermal cells thicken to form an oval plate
called the prechordal plate (Fig. 2.4B).2,4 The prechordal plate decides the cephalic end of the embryo. The intraembryonic mesoderm extends between the ectoderm and endoderm over the entire embryonic disc except at two
sites—one in the region of prechordal plate and the other caudal to primitive streak. At these sites, the endoderm
is closely adherent to overlying ectoderm without mesoderm in between forming two bilayered membranes—the
buccopharyngeal membrane (cranially) and the cloacal membrane (caudally) (Fig. 2.4). Buccopharyngeal membrane is the site for future oral opening and cloacal membrane for anal opening.4
The embryonic disc grows more at the cephalic end than the caudal end because of continuous migration of
cells from primitive streak and primitive node in the cephalic direction. This causes the rounded embryonic disc
to become elongated with broad cephalic and narrow caudal end (Fig. 2.4B).4 The primitive streak regresses after
the third week and finally disappears. The primitive pit surrounded by cord of cells extends in cephalic direction from primitive node to the prechordal plate in midline and lies between the ectodermal and endodermal
layers. This canalized cellular cord is called notochordal process (Fig. 2.4B). This process cannot extend beyond the
prechordal plate as the endoderm and ectoderm are firmly adherent to each other here.2 The cells in the floor
of the notochord canal fuse with the endoderm cells beneath it (which forms roof of yolk sac) and subsequently
both group of cells disappears in craniocaudal direction. Thus, the yolk sac communicates with amniotic cavity
through primitive pit. This temporary communication between the two cavities is called neurenteric canal, which
later gets closed (Fig. 2.6A). The notochord process now forms a notochordal plate along the roof of the yolk sac
(Fig. 2.6B). Later this plate folds along its long axis and separates from the roof of yolk sac, which is now lined
by endoderm. This chord of cells is known as definitive notochord (Fig. 2.6C).5,6
I. NEUROANATOMY
DEVELOPMENT OF NERVOUS SYSTEM
45
FIGURE 2.7 Stages in the formation of neural tube and neural crest cells. Formation of (A) neural plate (B,C) neural fold, neural groove and
neural crest cells and (D) neural tube with neural canal.
FIGURE 2.8 Differentiation of neural tube into different layers and formation of alar and basal plates.
DEVELOPMENT OF NERVOUS SYSTEM
The nervous system develops from the ectodermal cell layer. During the fourth week, the individual differentiation of the three germ layers and formation of the folds of embryo occur. The ectoderm cell overlying the notochord
thickens to form neural plate (Fig. 2.7A). The cells of neural plate are called neuroectodermal cells, which later give
rise to nervous system.5,7,8 The lateral margins of the neural plate become raised to form the neural folds forming
a longitudinal groove in between known as neural groove (Fig. 2.7B). The margins of the neural fold are lined by
special neuroectodermal cells called the neural crest cells (Fig. 2.7B,C). Gradually, the neural folds come close to
each other and fuse dorsally to form a hollow tune known as neural tube (Fig. 2.7D). The fusion begins at the future
cervical region and then proceeds both in cranial and caudal direction. The process of conversion of neural plate
into the neural tube is known as neurulation. The neural tube initially has the openings at the cephalic and caudal
ends known as anterior and posterior neuropores, respectively. These openings get closed by the end of 4 weeks,
thus giving rise to a completely closed, hollow neural tube from which the central nervous system (brain and spinal
cord) develops. Later, the dorsal surface of the neural tube gets detached from the surface ectoderm, and the neural
tube comes to lie underneath the surface ectoderm (Fig. 2.7D). The neural crest cells dissociate from the neural tube
(Fig. 2.7D) and migrate to form melanocytes in skin and hair, dorsal root ganglion (DRG), sympathetic ganglion,
enteric neurons, cells of adrenal medulla, and Schwann cells. The cranial part of the neural tube enlarges and gives
rise to the brain while the narrow caudal part forms the spinal cord. The cavity of the neural tube, known as neural
canal, gives rise to the ventricles of the brain and the central canal of spinal cord. The peripheral nervous system (PNS)
I. NEUROANATOMY
46
2. NEUROEMBRYOLOGY
FIGURE 2.9 Development of central nervous system (brain and spinal cord) from the neural tube. (A) Anterior part enlarges to form the three
primary vesicles of brain while the narrow posterior part forms the spinal cord. (B) Primary and secondary vesicles of the brain develop into
various parts of brain. The cavity of the neural tube gives rise to ventricles of brain and central canal of spinal cord. AS, aqueduct of Sylvius; FV,
fourth ventricle; LV, lateral ventricle; TV, third ventricle.
(which includes spinal, cranial and autonomic nerves, and their ganglia) is derived partly from the neural tube and
partly from the neural crest cells.5
As the growth occurs, the lateral walls of the neural tube thicken by proliferation of the cells lining the tube called
neuroepithelial cells. However, the roof and floor of neural tube remain thin. These neuroepithelial cells differentiate
into two types of cell—the neuroblasts that form the neurons and gliablasts that forms the glial cells (astrocytes and oligodendrocytes). The neuroblasts form a layer around the neuroepithelium called the mantle layer that forms the gray
matter of brain and spinal cord. The axons of these neuroblasts form the marginal layer outside the mantle layer that
forms the white matter. The neuroepithelial cells layer around the neural canal form the ependymal or germinal layer
(Fig. 2.8). The thickened lateral wall of neural tube gets divided into an alar plate dorsally and a basal plate ventrally
by a longitudinal groove called sulcus limitans (Fig. 2.8).5
Development of Brain
The cephalic portion of the neural tube enlarges to form three successive dilatation (separated by two circular
constrictions) known as primary brain vesicles (Fig. 2.9). These are prosencephalon or forebrain vesicle, mesencephalon or
midbrain vesicle, and rhombencephalon or hindbrain vesicle, which later form the forebrain, midbrain, and hindbrain,
respectively. Two evaginations appear one on each side of forebrain vesicle (prosencephalon) and divide it into an
anterior part called telencephalon (which includes the two evaginations and the area intervening between them) and
a posterior part called diencephalon. The hindbrain (rhombencephalon) vesicle is also subdivided into an anterior
part called metencephalon and a posterior part called myelencephalon. The cavities of the telencephalon, diencephalon,
mesencephalon, and rhombencephalon form the lateral ventricles, third ventricle, aqueduct of Sylvius, and the fourth
ventricle, respectively (Fig. 2.9).2–7 All these cavities are connected with each other and caudally with the cavity of the
spinal cord. Each lateral ventricle is connected to the third ventricle through the interventricular foramen of Monro. The
third ventricle in turn is connected to the fourth ventricle through aqueduct of Sylvius. The fourth ventricle communicates caudally with the central canal of spinal cord and subarachnoid space around the brain through foramen of
Luschka and Magendie. This forms a continuous channel for the flow of cerebrospinal fluid (CSF).
Prosencephalon
It includes telencephalon and diencephalon and forms the forebrain.
Telencephalon
It gives rise to the two cerebral hemispheres and corpus striatum. The telencephalon consists of two lateral outbulgings or evaginations (telencephalic vesicles), which form the right and left cerebral hemispheres and (a median
portion between the two evaginations) lamina terminalis. As the telencephalic vesicles increase in size, they completely
I. NEUROANATOMY
DEVELOPMENT OF NERVOUS SYSTEM
47
cover the lateral aspect of diencephalon and eventually fuse with it. With further expansion of telencephalic vesicles
(in upward, forward, and backward direction), the two vesicles forming cerebral hemisphere come to lie in opposition
to each other. Due to the growth of the telencephalic vesicles in the anteroposterior direction, the frontal and occipital
lobes are formed taking the cavity along with it (forming anterior and posterior horn of lateral ventricle). The upward
expansion forms the parietal lobe. The posterior part of the telencephalic vesicle also grows downward and forward
forming the temporal lobe into which extends the inferior horn of the lateral ventricle. Thus the lateral ventricle now
becomes C-shaped. Due to enlargement of telencephalon, the medial surfaces of the two cerebral hemispheres lie
opposite to each other with a groove in between. The floor of this groove is formed by the roof of the third ventricle
(diencephalon). Just above the floor of this groove, the medial wall of each hemisphere invaginates into the lateral
ventricle forming the choroid fissure. A fold of piamater extends into this fissure and forms the telachoroidea in which
lies bundle of capillaries forming choroid plexus. Immediately above the choroid fissure, the wall of each cerebral hemisphere thickens to form hippocampus, which bulges into the lateral ventricle on each side.5
Corpus striatum develops from the wall of the telencephalon. The floor of the developing hemispheres thickens to
form corpus striatum that bulges into the floor of the lateral ventricles. The growth of the temporal lobe carries the
caudal part of the corpus striatum along with it into the roof of inferior horn of the lateral ventricle. Thus, the corpus
striatum becomes a C-shaped structure. As the axons of developing neurons grow to make connections between the
cerebral hemisphere and other areas of brain and spinal cord, they pass through the corpus striatum and divide it into
dorsomedial part (caudate nucleus) and ventrolateral part (lentiform nucleus). The developing axon collectively forms
the fiber bundle known as internal capsule. The caudate nucleus is C-shaped and consists of head, body, and tail. The
head and body lie in the floor of the lateral ventricle and the tail in the roof of the inferior horn of lateral ventricle. The
tail ends in an enlargement known as amygdaloid body. The lentiform nucleus is later divided into putamen and globus
pallidus. The cerebral cortex overlying the corpus striatum grows at a relatively slower rate, so that it gets completely
buried by the adjoining lobes. This area is called as insula, with the overlying lobes forming the operculum.5
A groove appears on the under surface of the telencephalic vesicles in the anteromedial part, which evaginates
rostrally. It outgrows as a solid structure forming the olfactory tract and dilates at the distal end to form the olfactory
bulb. The area where the olfactory tract is attached to the under surface of brain is known as the piriform area.2 The
growth of the surface of cerebral hemisphere (i.e., cerebral cortex) is more than the hemisphere as a whole. Thus, the
cortex is thrown into folds which form the sulci and gyri on the surface.
The axons of the cortical neurons grow toward the other areas of the same cortex (association fibers) or opposite cortex
(commissural fibers) or to other regions of brain such as brain stem or spinal cord (projection fibers). Axons also connect basal
ganglia, hypothalamus, and thalamus to each other and to the cerebral cortex. Also there are axons projecting from the spinal cord and brain stem to thalamus and cerebral cortex. All these constitute the white matter of the cerebral hemisphere.
The corpus callosum is the largest bundle of commissural fibers connecting the two cerebral hemispheres.5–8
Diencephalon
It gives rise to thalamus, hypothalamus, epithalamus, optic cup, and stalk and pars nervosa of pituitary gland. The cavity of diencephalon forms the third ventricle of brain. Diencephalon gets hidden from the surface due to growth of telencephalic vesicles which covers it completely. The roof of diencephalon is a thin plate formed by single layer of ependymal
cells. The posterior of the roof plate thickens in midline to form the pineal gland or epiphysis. Above the roof lies the mesoderm into which the capillary vessels grow. This plexus of capillaries together with single layer of ependymal cells of the
diencephalon forms the choroid plexus which projects from the roof into the third ventricle. The alar plate forms the lateral
wall of diencephalon. Two grooves (epithalamic and hypothalamic sulci) appear and divide the lateral wall (alar plate) into
three regions. Region above the epithalamus sulcus forms the epithalamus, region dorsal to hypothalamic sulcus (between
the epi- and hypothalamic sulcui) forms the thalamus, and region ventral to the hypothalamic sulcus forms the hypothalamus. On ventral surface of hypothalamus, a group of cells form a midline structure called mammillary body on each side.2–5
Mesencephalon
It gives rise to midbrain and its cavity forms the aqueduct of Sylvius. Midbrain mainly contains axons (fiber tracts)
connecting the forebrain to the hindbrain or spinal cord and some group of cell bodies called nuclei. The basal and
alar plates of mesencephalon give rise to important nuclei. The basal plate gives rise to nucleus of oculomotor, trochlear
nerves, and the Edinger–Westphal nucleus while the alar plate gives rise to superior and inferior colliculus, red nucleus,
and substantia nigra. The marginal layer of basal plate expands to form the crus cerebri.2,4
Rhombencephalon
It gives rise to hindbrain and consists of two parts—metencephalon cranially and myelencephalon caudally. The
cavity of rhombencephalon forms the fourth ventricle.
I. NEUROANATOMY
48
2. NEUROEMBRYOLOGY
Metencephalon
It forms the pons and cerebellum. Pons develops from ventral part of metencephalon. The lateral wall of metencephalon becomes everted so that the alar plate comes to lie on the dorsolateral aspect of basal plate and the roof
of the metencephalon becomes wide and thin. These plates give rise to various nuclei in the region of pons. From
the basal plate develops the abducent nerve nucleus, motor nuclei of trigeminal and facial nerves, and the superior
salivatory nucleus of facial nerve while the alar plate gives rise to cranial part of dorsal nucleus of vagus, pontine
part of nucleus tractus solitaries, pontine part of sensory nucleus of trigeminal nerve, and nuclei of vestibulocochlear
nerve. The nuclei of basal and alar plates are arranged on the dorsal aspect of pons and together they form the tegmentum of pons. Some neurons from alar plate migrate ventrally and form pontine nuclei. Marginal layer of basal
plate expands through which fibers connecting the cerebral cortex and cerebellum to medulla and spinal cord cross.
Hence the name pons, meaning bridge.
The cerebellum develops from alar plates of metencephalon. The margins of the alar plates that attach to the roof of
metencephalon bend medially to form the rhombic lip bilaterally.2–6 The rhombic lips are wide apart in the caudally
but are close to each other cranially. The cells of rhombic lip proliferate to form the cerebellar plates. These cerebellar
plates grow to form the cerebellum. The median portion forms the vermis, and the lateral portion forms the cerebellar hemispheres. As further growth occurs, numerous fissures appear on its surface and divide it into lobes and folia.
Axon (white fibers) connecting the cerebellum to the cerebral cortex, pons, and medulla or spinal cord form the
superior, middle, and inferior cerebellar peduncles, respectively.5
Myelencephalon
Myelencephalon gives rise to medulla oblongata. Medulla oblongata has a closed lower part with a central canal and
an open upper part forming the caudal area of the fourth ventricle. Like pons, the lateral wall is everted so that the
alar plate comes to lie dorsolateral to basal plate and the roof is stretched. The cells of the basal and alar plates give
rise to various nuclei. The basal plate forms the hypoglossal nucleus, nucleus ambiguous (which contributes fibers to
glossopharyngeal, vagus, and accessory nerves), dorsal nucleus of vagus nerve, and inferior salivatory nucleus of glossopharyngeal nerve. The alar plate contributes to dorsal nucleus of vagus, nucleus of tractus solitarius, spinal nucleus
of trigeminal nerve, cochlear and vestibular nuclei.2 The roof plate of myelencephalon is thin, which is formed by single
layer of ependymal cells. Over it lies the piamater derived from vascular mesenchyme. The pia along with ependymal
cells forms the telachoroidea into which tuft of capillaries grow. This plexus of capillaries is called choroid plexus that
bulges from roof of the fourth ventricle and produces CSF. At three areas the roof the fourth ventricle bulges and finally
ruptures forming the foramen of Magendie in the middle and foramen of Luschka on each side.2
Development of Spinal Cord
The caudal part of the neural tube develops into the spinal cord (Fig. 2.9). The thickened lateral wall of neural
tube gets divided into an alar plate dorsally and a basal plate ventrally by a longitudinal groove called sulcus limitans
(Fig. 2.10A). The basal plate forms the motor area of spinal cord containing the motor horn cells ventrally. The alar
plate forms the sensory area of the spinal cord containing sensory horn cells dorsally. Another group of neurons
appear in the thoracolumbar region of the spinal cord (T1-L3) between the ventral and dorsal horn cells. These form
the intermediate horn and are concerned with sympathetic nervous system. The enlargement of the basal plate on
either side forms a furrow ventrally in the midline called the anterior or ventral median fissure.5
FIGURE 2.10 Development of spinal cord from the lower part of the neural tube. (A) The neural tube is divided into alar and basal plates by
sulcus limitans. (B) The alar plate forms the dorsal or sensory horn while the basal plate forms the ventral or motor horn of the spinal cord.
I. NEUROANATOMY
DEVELOPMENT OF NERVOUS SYSTEM
49
FIGURE 2.11 Transverse section of spinal cord. Each spinal nerve is attached to the spinal cord through the dorsal (sensory) root and ventral
(motor) root. Both the spinal nerve roots join to form the trunk of spinal nerve which then divide into dorsal and ventral primary rami.
Initially, the spinal cord extends throughout the length of developing vertebral column. However, the vertebral
column grows faster in length than the spinal cord, so that the spinal cord ends at the level of the third lumbar vertebrae at birth and at lower border of the first or upper border of the second lumbar vertebrae in adults.5 Below this
level the nerve roots of spinal nerve extends toward the corresponding intervertebral foramina as a bundle of nerve
roots known as cauda equina.
Development of Peripheral Nervous System
The PNS consists of the spinal and cranial nerves and the autonomic (sympathetic and parasympathetic) nervous
system.
Spinal Nerve
Each spinal nerve is connected to the spinal cord by the dorsal (sensory) and ventral (motor) nerve roots (Fig. 2.11).
The axons of the basal plate neurons (motor neurons) pass through the marginal layer and form the ventral or motor
root of spinal nerve. The dorsal or sensory root of spinal nerve is formed by the axon of neurons located in DRG of each
spinal nerve. The DRG is formed by the neural crest cells. The cells in the DRG give two processes. The central process
migrates toward the spinal cord and reaches the spinal cord along the dorsal root of spinal nerve. The peripheral
process moves peripherally and joins the fibers of the ventral root to form the nerve trunk of spinal nerve which then
divides into the dorsal and ventral primary rami and supplies the skin, joints. and muscles of a specific region of
the body (Fig. 2.11). The peripheral process of DGR neurons thus reaches the sensory receptors in the distribution of
spinal nerve. These neurons carry sensory impulse from periphery to spinal cord.5,6
The fibers that form the peripheral nerves and constitute the PNS, are covered by a sheath around them called
neurilemma. This sheath is formed by the Schwann cells which are derivatives of neural crest cells. The Schwann cells
form the myelin sheath around the axons in the peripheral nerves.2,4
The axons of DRG, which reach the spinal cord through the dorsal root either synapse with neurons of alar plate
or ascend without synapsing through the marginal layer. The axons of the alar plate neurons also ascend through
the marginal layer. The axons (ascending in the marginal layer) form the ascending tracts. The descending tracts are
formed by the axons of neurons in the brain, which descends in the marginal layer. These axons synapse with the
neurons of ventral basal plate and carry motor impulse from the brain. Due to formation of dorsal and ventral horns
by the mental layer (gray matter), the marginal layer (white matter) gets organized into the anterior, lateral, and
posterior columns having the fibers of ascending and descending spinal tracts.5
Autonomic Nervous System
Sympathetic Nervous System
It consists of a pair of sympathetic trunks which are elongated chains of sympathetic nerve fibers running along
each side of vertebral column with number of sympathetic ganglia along its length.9 The cells of sympathetic ganglia
arise from the neural crest cells. The neural crest cells migrate to lie posterior to aorta (to form sympathetic ganglia) or
in front of aorta (to form preaortic ganglia such as celiac ganglia). The axon from the intermediate horn of spinal cord
migrates toward the sympathetic ganglia by passing through the ventral root of spinal nerve to reach the sympathetic
ganglia via white rami communicantes. These are known as preganglionic fibers and are myelinated. These axons either
I. NEUROANATOMY
50
2. NEUROEMBRYOLOGY
FIGURE 2.12 Arrangement of pre- and postganglionic sympathetic nerve fibers.
make connections with the neurons in the same sympathetic ganglia or travel up or down through the sympathetic
chain without synapsing to other sympathetic ganglia and synapse there. The axons of the neurons in the sympathetic
ganglion pass back through gray rami communicantes to reach the spinal nerves (Fig. 2.12). These are the nonmyelinated
postganglionic sympathetic fibers which supply blood vessels, hairs, sweat glands of skin through spinal nerves. A
few preganglionic sympathetic fibers may leave the sympathetic trunk, without synapsing, via the visceral branches
of sympathetic trunk such as cardiac, pulmonary, or splanchnic nerves to reach the autonomic nerve plexuses and synapse in these plexuses. The postganglionic fibers supply the various visceral organs such as heart, lungs, intestine, etc.
(Fig. 2.10).5,9,10
Parasympathetic Nervous System
The parasympathetic fibers emerge from the brain and sacral segment of spinal cord along with the cranial and
sacral spinal nerves (craniosacral outflow). The parasympathetic neurons are located in the brain stem associated
with the nuclei of origin of cranial nerves III, VII, IX, and X.9,10 The axons of these neurons pass through the corresponding cranial nerves to synapse in the peripheral parasympathetic ganglia. These axons constitute the preganglionic parasympathetic fibers. The postganglionic fibers from these ganglions are short, nonmyelinated and supply
various glands, eye, thoracic, and abdominal viscera. In the sacral region, the parasympathetic neurons are located
in the gray matter of the second, third, and fourth sacral segment of spinal cord. The preganglionic parasympathetic
fibers (myelinated) emerge through the ventral root of corresponding sacral spinal nerves and reach the pelvic autonomic plexuses passing through the pelvic splanchnic nerves and synapse here. The postganglionic (nonmyelinated)
fibers innervate the pelvic viscera and part of large intestine.11
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
Balinsky BI, Fabian BC, editors. An introduction to embryology. 5th ed. Philadelphia: WB Saunders Co.; 1981.
Dutta AK, editor. Essentials of human embryology. 3rd ed. Calcutta: Current Books International; 1995.
Austin CR, Short RV, editors. Reproduction in mammals. 2nd ed. Cambridge: Cambridge University Press; 1984.
Singh I, editor. Human embryology. 10th ed. New Delhi: Jaypee Brothers Medical Publishers Ltd; 2014.
Kumar R, editor. Human embryology. 1st ed. New Delhi: Top Publishing Company; 2011.
Carlson BM, editor. Human embryology and developmental biology. Philadelphia (PA): Elsevier Saunders; 2013.
Schoenwolf GC, Bleyl SB, Brauer PR, Francis-West, editors PH. Larsen’s human embryology. 5th ed. Philadelphia (PA): Elsevier Saunders; 2015.
Sadler TW, editor. Langman’s medical embryology. 12th ed. Philadelphia: Lippincott Williams and Wilkins; 2012.
Snell RS, editor. Clinical anatomy for medical students. 5th ed. Boston: Little Brown and Company (inc.); 1995.
Young PA, Young PH, Tolbert DL, editors. Basic clinical neurosciences. 2nd ed. Philadelphia: Lippincott Williams and Wilkins; 2008.
Singh, editor I. Textbook of anatomy. 5th ed. New Delhi: Jaypee Brothers Medical Publishers; 2011.
I. NEUROANATOMY
C H A P T E R
3
Blood–Brain Barrier
A.K. Khanna, E. Farag
Cleveland Clinic Foundation, Cleveland, OH, United States
O U T L I N E
Introduction
51
Permeability at the Blood–Brain Barrier
51
Anesthetic Neuroprotection in Perioperative
Neurological Injury
54
Antiinflammatory Effects of General Anesthetics in the
Treatment of Refractory Status Epilepticus
55
Antiinflammatory Considerations in Barbiturate
Induced Coma for Traumatic Brain Injury
55
Cellular and Molecular Effects of Anesthetics on the
Blood–Brain Barrier
52
Anesthesia and Nitric Oxide Signaling
52
Anesthesia Effects on Tight Junctions
52
Anesthesia Effects on Endothelial Cells
53
Anesthesia and Neuroinflammation
53
Clinical and Experimental Implications of
Anesthetics on the Blood–Brain Barrier
Conclusion
56
References
56
54
INTRODUCTION
The blood–brain barrier (BBB) maintains the brain parenchyma and blood components in separate compartments.
In addition, by allowing glucose transport it helps fuel neuronal function. Maintenance of the integrity of this closed
compartment comprises a dynamic combination of vascular, cellular, molecular, and ionic factors.1 Structurally, this
barrier is composed of endothelial cells supported mainly by astrocytes and pericytes. BBB endothelial cells also
have a transport function that acts to maintain a constant parenchymal milieu. Endothelial cells transport amino
acids, participate to ionic homeostasis, and allow a controlled exchange of solute and water. Importantly, a variety
of traumatic and nontraumatic inflammatory insults to the BBB may lead to a loss of the closed compartment and
consequences of such.
The anesthesiologist must be aware that interventions such as cardiopulmonary bypass,2,3 cerebral arteriography,4
and osmotic BBB opening5 have all been linked to impairment of cerebral homeostasis in patients. We will focus our
text to the effects of anesthetics on the BBB and clinical implications of the same.
PERMEABILITY AT THE BLOOD–BRAIN BARRIER
Structurally, the brain microvasculature is lined with endothelial cells that are secured together by tight junctions
(TJs). These TJs provide a means to regulate movement of substances into and out of the brain. The lipid bilayer
of these endothelial cells allows movement and determines permeability across the BBB. Substances do not cross
through the alternative paracellular route.6
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00003-8
51
© 2017 Elsevier Inc. All rights reserved.
52
3. BLOOD–BRAIN BARRIER
Translated into clinical practice, this unique structure means that the BBB does not allow a majority of CNS drugs to
enter the brain parenchyma.7 Interestingly anesthetics are an important exception in that they freely exert CNS effects,
and it has been proven that the lipophilicity of these agents drives them to cross the BBB. The log octanol/water partition coefficient has a significant role in predicting how and if compounds will cross the BBB.
Typically this coefficient is determined using an aqueous substance (water) and a hydrophobic substance (octanol). Compounds with a high log (P) favor hydrophobic compartments and will cross a lipid bilayer while compounds with a low log (P) will tend to stay in hydrophilic compartments (e.g., serum) and will not cross the BBB.8 As
a general rule, compounds with a log (P) > 0 will cross the BBB rapidly with the major limiting factor being supply of
the drug. On the other hand, compounds with a log (P) < −1 are limited in their ability to cross the BBB.9 Importantly,
log (P) is a velocity, and therefore a higher value is necessary for a clinically relevant effect. It is indeed in actuality a
pharmacokinetic property that allows anesthetic agents to move from blood to brain.
Also, compounds with similar log (P)s can have differing ability to cross the BBB.9 To cross the BBB efficiently, a
number of conditions must be met: (1) Larger compounds (greater than 400 Da) require some additional transport
mechanism to cross the BBB. These are too large to pass through TJs or directly across the lipid bilayer. (2) Many
drugs have changes in ionization states that affect the ability of a drug to cross the BBB. (3) Hyperthermia enhances
while hypothermia impedes BBB permeability. (4) Highly regulated mechanisms of transport by BBB endothelial
cells. This type of facilitated transport is dependent on membrane receptors and is the major mechanism of transport
regulation across the BBB (most notably glucose).10
CELLULAR AND MOLECULAR EFFECTS OF ANESTHETICS ON THE BLOOD–BRAIN
BARRIER
Anesthetic agents may interact in several different ways with the BBB. This is because the same anesthetic agent
establishes a different relationship with the target, signaling pathway, and pathology involved. Both direct and indirect effects of anesthetic agents on the molecular components of BBB integrity may affect these signaling pathways.
At the cellular level several changes may be important such as tight and adherens junctions, vasodilation, endothelial
cell survival, and neuroinflammation.11–15 Therefore, anesthetic action at the BBB depends upon the extent to which
the agent modulates these pathways of signal transmission and in addition the systemic cerebral milieu in which it
is administered.
Anesthesia and Nitric Oxide Signaling
Anesthetics have a varied effect on nitric oxide (NO) signaling pathways. NO is a signaling molecule and potent
vasodilator that has been effective in ischemic preconditioning and, at low levels, shown to induce neuroprotection
mediated by the BBB. NO has important downstream effects such as vascular regulation, mediation of neuroinflammation, and regulation of endothelial cell survival.15–17
Volatile anesthetics result in potent cerebral vasodilation that in turn can induce endothelial stretching, increasing
the gap between neighboring endothelial cells, and hence BBB permeability.12,13 These agents act in part via modulation of NO signaling (via nitric oxide synthase iNOS) and regulation of ATP-sensitive K+ channels in vascular smooth
muscle cells.11
Let us look at isoflurane as a specific example. Isoflurane can induce nitric oxide synthase (iNOS)-dependent
cerebral vasodilation and also neuroprotection via NO downstream effects. But as both the agents, on the flip side,
NO and isoflurane are known to stimulate prostanoid production, this may potentially result in increased inflammation.15 Some other studies have also noted the proapoptotic effects of NO donors in vitro that lead to disruption
of the cerebral endothelial cell monolayer and increased leukocyte adhesion in the presence of NO.16 Some of these
effects may be attenuated by propofol administration.17
Volatile anesthetics are important regulators of cardiovascular output, vascular tone, and cerebral blood flow. This
may be a mechanism of BBB perturbation (via endothelial stretching) and a potential experimental confounder when
assessing other molecular mechanisms influencing permeability.18
Anesthesia Effects on Tight Junctions
Animal models have shown isoflurane results in an increased edema in comparison to other anesthetics such
as sevoflurane or pentobarbital.12,19–21 While vasodilation-induced cell starching and separation may play a role,
I. NEUROANATOMY
CELLULAR AND MOLECULAR EFFECTS OF ANESTHETICS ON THE BLOOD–BRAIN BARRIER
53
other mechanisms are important in determining the direct effects on TJs.12 TJs are redistributed following ischemia, inflammation, and TBI.22–24 Sevoflurane compared to isoflurane, may alter the balance between TJ proteins
that are modulated by anesthetics specifically by greater induction of zonula occludens-1 (ZO-1) as compared
to occludin.13,25,26 This coincides with a reduction in cerebral edema in sevoflurane-treated animals. The other
important TJ protein, occludin, is regulated in part by glutamate signaling, which is altered by certain anesthetics.
Glutamatergic signaling alters the phosphorylation of certain occludin residues, resulting in its redistribution and a
consequent increase in BBB permeability. The clinically important implication for this mechanism is the reversal of
this increased permeability via NMDA antagonists including the commonly used ketamine.25,27 An increase in circulating amino acids levels results in elevated plasma and cerebrospinal fluid (CSF) glutamate. This presents another
possible mechanism by which anesthetic administration may modulate BBB TJs via glutamatergic signaling.28,29
As a direct effect, isoflurane downregulates the expression of occludin in human brain vascular endothelial cell
cultures in a hypoxia inducible factor-1α (HIF-1α)–dependent manner.30
In addition to its role in occludin expression, HIF-1α acts in biphasic manner with BB integrity to both protect against
and exacerbate damaging pathways of cerebral ischemia and consequently plays a significant role in mediating cerebral water content.31–33 Propofol and isoflurane have both been implicated in modulation of this signaling pathway,
which regulates expression of vascular endothelial growth factor, aquaporin-1, aquaporin-4, and matrix metalloprotease expression, among many others.32 Importantly, for the traumatized brain, HIF-1α has a biphasic expression, with a
trough at 24 h postreperfusion that corresponds to a decrease in BBB permeability.31 While the protective effects of early
HIF-1α inhibition with respect to increased BBB integrity have been confirmed by several studies, biological reality is
likely to be far more complex.26,31,33 The reader is referred to the bibliography to find more reading of the HIF pathways,
which is beyond the scope of inclusion in this text.
Anesthesia Effects on Endothelial Cells
Endothelial cell survival, structure, and expression of adherens or TJ proteins represent another set of pathways
by which anesthetics, particularly isoflurane and propofol, alter the integrity of the BBB.17,34,35 In addition to its aforementioned roles, isoflurane posttreatment upregulates the activity of sphingosine kinase 1 (SphK1), which catalyzes
production of sphingosine 1 phosphate (S1P). One such downstream signaling receptor of S1P is S1P1, signaling of
which has been shown to decrease BBB permeability by inducing changes in expression of junction proteins such
as VE-cadherins and altering cytoskeletal structure that decreases space between neighboring endothelial cells via
GTPases Rho/Rac.36–38 Endothelial cell proliferation and migration is mediated via another such S1P receptor, S1P3.
Furthermore, S1P3 alters calcium signaling and induces vasoconstriction in vascular smooth muscle, while S1P1
activation upregulates endothelial nitric oxide synthase activity, resulting in vasodilation.39,40 Isoflurane activates
the antiapoptotic Akt pathway, increasing endothelial cell survival and improving cerebral edema via SphK1.14,35
The long-term protective effects of isoflurane postconditioning appear to depend upon the model of cerebral injury
(e.g., hemorrhagic versus ischemic stroke), outcomes measured, and anesthetic protocol.14,35,41 This reemphasizes
that clinical context remains of paramount importance in that the effects of anesthetics such as isoflurane in different
experimental neurological pathologies are different, where the mechanism of early brain injury may be similar, but
the sequelae distinct.42
Anesthesia and Neuroinflammation
Neuroinflammation plays a significant role in the damage that follows a cerebral insult by mediating acute
increases in permeability leading to delayed cell death.41 The role of inflammation via NFκB, IL-1β, TNF-α, and
other reactive oxygen species in oxidative stress, endothelial dysfunction, and apoptosis has been described in many
studies with potential clinical implications including cerebral edema, neuronal death, and short- and long-term cognitive impairment.35,43–45 Significantly, anesthesia has mostly antiinflammatory or rather minimal proinflammatory
effects in healthy animals.46,47 Pretreatment or postconditioning with isoflurane reduces expression of the proinflammatory cytokines NFκB, IL-1β, and IL-6, while ketamine consistently demonstrates similar antiinflammatory
properties, decreasing expression of NFκB, COX-2, iNOS, TNF-α, IL-1β, and other important inflammatory signaling
molecules.41,43,48 Propofol exhibits similar antiinflammatory effects in endothelial cells in vitro through inhibition of
NFκB, iNOS, and IL-1β.17 Extending beyond endothelial cells, astrocytes (an integral part of the BBB) exposed to midazolam and corticosterone challenge produced pregnenolone and progesterone, both of which are cytoprotective.49,50
Clinical effects of delayed cognitive dysfunction that are not immediately evident during acute-phase reactions may
be secondary to the role of neuroinflammation acting in conjunction with delayed apoptotic signaling.14,46 This also
I. NEUROANATOMY
54
3. BLOOD–BRAIN BARRIER
may translate into timed administration of specific anesthetics that may protect the BBB and thus reduce cerebral
edema and neuronal death in common inflammatory states such as infection, malignancies, etc.41,46
Accumulating evidence of the effect of surgery on neuroinflammation and neurodegeneration suggests a potentially important role in anesthetic selection for vulnerable populations such as children, the elderly, and those with
preexisting neurologic conditions such as Alzheimer disease.46,51 Induction of neuroinflammation by anesthetics is of
particular concern in pediatric populations due to the documented inhibitory effect of proinflammatory cytokines on
long-term potentiation.46 Indeed, consistent administration of sevoflurane or isoflurane to young mice results in cognitive impairment, although the causal mechanisms have yet to be fully elucidated.51,52 In young mice, sevoflurane
administration upregulates the production inflammatory cytokines IL-6 and TNF-α, possibly as a downstream result
of NFκB induction.52 Moderate induction of neuroinflammation by sevoflurane may have a particularly significant
effect during periods of neurodevelopment due to the documented inhibitory effect of proinflammatory cytokines
on long-term potentiation. This would support the clinical findings that repeated exposures to anesthesia during
childhood can raise the risk for cognitive impairment as well as experimental results identifying neurodegeneration
in neonatal rats exposed to isoflurane.12,53 In light of the evidence in favor of an antiinflammatory role of volatile
anesthetics under pathologic conditions, it is unclear whether this is related to an age-dependent difference or to
experimental variability in models (e.g., TBI vs healthy animals) or dosage protocol. The much talked about general
anesthesia compared to spinal anesthesia (GAS trial) will examine outcomes at 2 and 5 years of age after exposure of
the infant brain to volatile anesthesia. This will help establish whether general anesthesia in infancy has any effect
on neurodevelopmental outcome. Outcomes at 2 years of age were accessed using the Bayley Scales of Infant and
Toddler Development III during this multicenter, randomized controlled trial. These outcomes that have since been
published found no evidence that just less than 1 h of sevoflurane anesthesia in infancy increases the risk of adverse
neurodevelopmental outcome at 2 years of age compared with awake-regional anesthesia.54
In summary, anesthetic agents differentially regulate several pathways directly and indirectly involved in BBB
integrity including TJ formation, endothelial cell survival, vasodilation, reactive oxygen species production and
signaling, and neuroinflammation. The varied effects of volatile anesthetics on inflammation and permeability
underscore the potential role of variability in administration protocols and the presence and type of underlying
neuropathology. These are important factors to account for in interpreting the above highlighted data by the bedside clinician. It is safe to conclude at this moment of time that the bulk of available data suggest a possible role for
anesthetic agents in pre- or postconditioning of the BBB, though some other studies have also identified potentially
detrimental effects including increased BBB permeability and neuroinflammation.
CLINICAL AND EXPERIMENTAL IMPLICATIONS OF ANESTHETICS ON
THE BLOOD–BRAIN BARRIER
The proceeding text has shown us that the interpretation of clinical and animal research to the operating room,
and intensive care units is guarded to say the least. The clinician would specifically like to know if anesthetic neuroprotection exists in common clinical scenarios.
The following section highlights some such common clinical scenarios, such as perioperative neurological injury
in cardiac surgery, the use of general anesthetics to treat refractory status epilepticus (SE), and high-dose barbiturate
therapy to treat refractory intracranial hypertension.
Anesthetic Neuroprotection in Perioperative Neurological Injury
Perioperative neurological injury is one of the most serious adverse complications of general anesthesia.
Manifestations are on a diverse spectrum ranging from dense coma and brain death to the relatively subtle (though
importantly very disabling) postoperative cognitive dysfunction (POCD).55 Thus, perioperative brain damage is a
major concern after cardiac surgery and is one such surgical insult where both cerebral ischemia and the POCD have
a significantly higher incidence compared to noncardiac surgery.56–58
While the etiology of perioperative neurological injury is incompletely understood, it is believed that in
addition to ischemia due to hypoperfusion and cerebral microembolization, followed by reperfusion injury,
cardiac surgery induces a systemic inflammatory response that may act in concert to disrupt the BBB and
cause cerebral edema and inflammation, thereby leading to neurocognitive dysfunction.59,60 This is supported
by animal models demonstrating that cardiopulmonary bypass leads to opening of the BBB.61,62 Importantly,
more recent studies have also corroborated these results in patients undergoing cardiac surgery. Reinsfelt et al.
I. NEUROANATOMY
CLINICAL AND EXPERIMENTAL IMPLICATIONS OF ANESTHETICS ON THE BLOOD–BRAIN BARRIER
55
demonstrated a pronounced cerebral inflammatory response in patients following “on-pump” surgical aortic
valve replacement. Their results show increased levels of proinflammatory cytokines in CSF as well as BBB disruption and glial cell injury as assessed using biomarkers of BBB dysfunction and astrocytic damage in CSF.63
Further, using MRI-DTI and FLAIR, others found subclinical BBB disruption in both on- and off-pump cardiac
surgery patients.64
Nussmeier et al. conducted a randomized, clinical trial in patients undergoing cardiac surgery with normothermic
cardiopulmonary bypass, in which they compared the infusion of thiopental with fentanyl in control subjects.65 They
concluded that thiopental offers neuroprotection in patients undergoing cardiopulmonary bypass. However, in a
previous randomized trial in patients undergoing coronary artery bypass grafting comparing thiopental to placebo,
Zaidan et al. reported that, contrary to the Nussmeier et al. results, there was no significant difference in either the
incidence of neurological deficits or rate of recovery among the two groups.66 It was concluded that thiopental does
not offer neuroprotection in patients undergoing coronary artery surgery. While many now regard thiopental as “an
agent of the past,” this example highlights the difficulty in ascertaining a clear outcome signal in these types of studies. The heterogeneity of the population and the methods used are the biggest problems when comparing these and
other studies.
Similarly a review of numerous other clinical trials investigating the neuroprotective properties of other anesthetics
has led to ambiguous conclusions.67 As a result, the neuroprotective statuses of both lidocaine58,68–71 and ketamine72,73
are currently still controversial. Propofol does not appear to have significant neuroprotective properties in patients
undergoing cardiac surgery.74,75
It is important to note that the follow-up period for a number of these studies was quite short (5–10 days postsurgery), which may have contributed to the significant findings. Thus, the question of whether any anesthetic neuroprotection is observed for a long term still remains. One randomized trial investigating the influence of propofol
versus desflurane on the long-term incidence of POCD in patients undergoing coronary artery bypass surgery found
that although desflurane was associated with decreased incidences of POCD early in recovery (4–7 days postsurgery), there was no significant difference in incidence between the two groups at 3 months postsurgery.93 This result
is in agreement with others who also looked at long-term follow-up and found no evidence for neuroprotection on
POCD incidence.69–71,74
Antiinflammatory Effects of General Anesthetics in the Treatment of Refractory Status Epilepticus
SE refers to the state in which a patient has continuous or rapidly repeating seizures most commonly generalized
tonic–clonic. To treat SE, benzodiazepines are used as first-line therapy (first 30 min) and intravenous antiepileptic
drugs, such as phenytoin, are used as second-line therapy (30–120 min).76 If SE does not respond to either first- or
second-line therapy, it is considered refractory SE and requires the application of general anesthetics.
While the neuronal molecular mechanisms by which general anesthetics work as antiepileptic drugs are widely
accepted—propofol and thiopental are γ-aminobutyric acid (GABA) receptor agonists and ketamine is an
NMDA receptor antagonist—these anesthetic drugs also have important immunomodulatory effects that partially overlap with those of corticosteroids.9 Thiopental, propofol, and ketamine exert potent antiinflammatory effects that are
mediated through inhibition of the activation of NFκB, a transcription factor essential for the expression of proinflammatory cytokines, in the experimental setting.77–82 Studies in patients have also corroborated the antiinflammatory
effects of ketamine.82 Sevoflurane lacks the antiinflammatory effects of general anesthetics and thus may actually have an
epileptogenic effect.77,80
BBB breach has been shown to decrease seizure threshold and promote seizure development in both animals and
humans.83,84 While general anesthetics have direct antiepileptic neuronal effects, they may be more effective in treating refractory SE because of their antiinflammatory properties that prevent BBB disruption and promote the repair
of the cerebrovasculature.23,78,79,81,85,86
Antiinflammatory Considerations in Barbiturate Induced Coma for Traumatic Brain Injury
Increased intracranial pressure (ICP) or intracranial hypertension occurs commonly in 25–40% of patients with
severe traumatic brain injury usually as a consequence of both vasogenic and cytotoxic edema.87–89 Vasogenic edema
is secondary to BBB disruption and results in the extravasation of proteins and fluid from the cerebrovasculature into
the extracellular space. On the other hand, cytotoxic edema—the accumulation of intracellular fluid—is believed to
occur as a result of loss of the cell’s ability to regulate ionic gradients. The Monroe–Kellie doctrine determines that
due to the rigidity of the skull, cerebral edema causes a significant increase in ICP and cerebral ischemia once the
I. NEUROANATOMY
56
3. BLOOD–BRAIN BARRIER
compensatory mechanisms are overwhelmed. However, despite aggressive management strategies, refractory intracranial hypertension may persist in approximately 10–15% of patients with severe TBI.90,91
High-dose barbiturates are recognized as an effective therapy for controlling refractory intracranial hypertension.92 One randomized, multicenter trial found that inducing barbiturate coma in such patients resulted in double
the chance of achieving ICP control.93 High-dose barbiturates, when used in hemodynamically stable patients (to
account for the effects on mean arterial pressure), are believed to lower ICP through the suppression of cerebral
metabolism, which reduces cerebral blood volume and ICP due to the coupling of cerebral blood flow to regional
metabolic demands.94
Thiopental exerts potent antiinflammatory effects through the inhibition of NFκB activation.79,85 Importantly, it has
been demonstrated that it is the thio-group of thiopental that is of functional importance for this inhibitory effect, as
the oxibarbiturate analogs of thiobarbiturates (such as pentobarbital vs thiopental) fail to inhibit NFκB in equimolar
amounts.78 Considering that vasogenic edema is secondary to BBB disruption, thiopental may thus help to decrease
cerebral edema by promoting the repair of the BBB through its antiinflammatory effects—although it is important to
note that it is actually cytotoxic edema that is believed to be the more insidious of the two types of edema involved in
intracranial hypertension.22
CONCLUSION
Anesthetics influence the BBB via a multitude of different mechanisms and roles. While some anesthetics appear
to have a negative effect on BBB permeability, others can provide protection to the BBB and decrease inflammation.
Molecular mechanisms of anesthetic influences on the BBB tailoring anesthesia to specific needs and personalization of anesthetics to individual patients undergoing a specific procedure are all areas of future research. The bulk
of evidence points to a neuroprotective and antiinflammatory effect of anesthetics on the brain. There is a limitation
in comparing studies with a lack of standardization of experimental practices. Once standardization is achieved, the
true nature and impact of anesthesia on the BBB and inflammation may be revealed.
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
Janigro D. Are you in or out? Leukocyte, ion, and neurotransmitter permeability across the epileptic blood–brain barrier. Epilepsia 2012;53:26–34.
Scott DAEL, Silbert BS. Cardiac surgery, the brain, and inflammation. J Extra Corpor Technol 2014;1:15–22.
Silbert B, Evered LA, Scott DA. Dementia after anesthesia and non-cardiac surgery. Alzheimer’s Dementia 2010;6:S373–4.
Numaguchi Y, Fleming MS, Hasuo K, Puyau FA, Nice CM. Blood–brain barrier disruption due to cerebral arteriography. J Computer Assisted Tomogr 1984;8:936–9.
Marchi N, Granata T, Alexopoulos A, Janigro D. The blood–brain barrier hypothesis in drug resistant epilepsy. Brain 2012;135:e211.
Abbott NJ, Bundgaard M, Cserr HF. Tightness of the blood–brain barrier and evidence for brain interstitial fluid flow in the cuttlefish, Sepia officinalis. J Physiol
1985;368:213–26.
Rajadhyaksha M, Boyden T, Liras J, El-Kattan A, Brodfuehrer J. Current advances in delivery of biotherapeutics across the blood–brain barrier. Curr Drug Discovery
Technol 2011;8:87–101.
Berthod A, Carda-Broch S. Determination of liquid–liquid partition coefficients by separation methods. J Chromatogr A 2004;1037:3–14.
Greig NH, Yu Q-S, Utsuki T, et al. Optimizing drugs for brain action. Blood—Brain Barrier 2001:281–309.
Goldstein GW, Betz AL. The blood–brain barrier. Sci Am 1986;255:74–83.
Matta BF, Heath KJ, Tipping K, Summors AC. Direct cerebral vasodilatory effects of sevoflurane and isoflurane. Anesthesiology 1999;91:677.
Tétrault S, Chever O, Sik A, Amzica F. Opening of the blood–brain barrier during isoflurane anaesthesia. Eur J Neurosci 2008;28:1330–41.
Thal SC, Luh C, Schaible E-V, et al. volatile anesthetics influence blood–brain barrier integrity by modulation of tight junction protein expression in traumatic
brain injury. PLoS One 2012;7:e50752.
Altay O, Hasegawa Y, Sherchan P, et al. Isoflurane delays the development of early brain injury after subarachnoid hemorrhage through sphingosine-related
pathway activation in mice. Crit Care Med 2012;40:1908–13.
Zhao P, Zuo Z. Isoflurane preconditioning induces neuroprotection that is inducible nitric oxide synthase-dependent in neonatal rats. Anesthesiology
2004;101:695–703.
Lehmberg J, Waldner M, Baethmann A, Uhl E. Inflammatory response to nitrous oxide in the central nervous system. Brain Res 2008;1246:88–95.
Chen R-M, Tai Y-T, Chen T-G, et al. Propofol protects against nitrosative stress-induced apoptotic insults to cerebrovascular endothelial cells via an intrinsic mitochondrial mechanism. Surgery 2013;154:58–68.
Sakabe T, Matsumoto M. Effects of anesthetic agents and other drugs on cerebral blood flow, metabolism, and intracranial pressure. In: Cottrell and Young’s neuroanesthesia. Elsevier BV; 2010. p. 78–94.
Ritz M-F, Schmidt P, Mendelowitsch A. Effects of isoflurane on glutamate and taurine releases, brain swelling and injury during transient ischemia and reperfusion. Int J Neurosci 2006;116:191–202.
Hu Q, Ma Q, Zhan Y, et al. Isoflurane enhanced hemorrhagic transformation by impairing antioxidant enzymes in hyperglycemic rats with middle cerebral artery
occlusion. Stroke 2011;42:1750–6.
Stover JF, Kempski OS. Anesthesia increases circulating glutamate in neurosurgical patients. Acta Neurochir 2005;147:847–53.
Unterberg AW, Stover J, Kress B, Kiening KL. Edema and brain trauma. Neuroscience 2004;129:1019–27.
Sandoval KE, Witt KA. Blood–brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis 2008;32:200–19.
Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood–brain barrier. Nat Med 2013;19:1584–96.
András IE, Deli MA, Veszelka S, Hayashi K, Hennig B, Toborek M. The NMDA and AMPA/KA receptors are involved in glutamate-induced alterations of occludin
expression and phosphorylation in brain endothelial cells. J Cereb Blood Flow Metab 2007;27:1431–43.
I. NEUROANATOMY
REFERENCES
57
26. Colgan OC, Collins NT, Ferguson G, et al. Influence of basolateral condition on the regulation of brain microvascular endothelial tight junction properties and
barrier function. Brain Res 2008;1193:84–92.
27. Miller RD, Monsul NT, Vender JR, Lehmann JC. NMDA- and endothelin-1-induced increases in blood–brain barrier permeability quantitated with Lucifer yellow.
J Neurol Sci 1996;136:37–40.
28. Carli F, Ronzoni G, Webster J, Khan K, Elia M. The independent metabolic effects of halo thane and isoflurane anaesthesia. Acta Anaesthesiol Scand 1993;37:672–8.
29. Horber FF, Krayer S, Miles J, Cryer P, Rehder K, Haymond MW. Isoflurane and whole body leucine, glucose, and fatty acid metabolism in dogs. Anesthesiology
1990;73:82–92.
30. Zhao J, Hao J, Fei X, Wang X, Hou Y, Deng C. Isoflurane inhibits occludin expression via up-regulation of hypoxia-inducible factor 1α. Brain Res 2014;1562:1–10.
31. Lee JH, Cui HS, Shin SK, et al. Effect of propofol post-treatment on blood–brain barrier integrity and cerebral edema after transient cerebral ischemia in rats.
Neurochem Res 2013;38:2276–86.
32. Engelhardt S, Al-Ahmad AJ, Gassmann M, Ogunshola OO. Hypoxia selectively disrupts brain microvascular endothelial tight junction complexes through a
hypoxia-inducible factor-1 (HIF-1) dependent mechanism. J Cell Physiol 2014;229:1096–105.
33. Engelhardt S, Patkar S, Ogunshola OO. Cell-specific blood–brain barrier regulation in health and disease: a focus on hypoxia. Br J Pharmacol 2014;171:1210–30.
34. Dittmar MS, Petermichl W, Schlachetzki F, Graf BM, Gruber M. Isoflurane induces endothelial apoptosis of the post-hypoxic blood–brain barrier in a transdifferentiated human umbilical vein edothelial cell model. PLoS One 2012;7:e38260.
35. Altay O, Suzuki H, Hasegawa Y, et al. Isoflurane attenuates blood–brain barrier disruption in ipsilateral hemisphere after subarachnoid hemorrhage in mice.
Stroke 2012;43:2513–6.
36. Gaengel K, Niaudet C, Hagikura K, et al. The sphingosine-1-phosphate receptor S1PR1 restricts sprouting angiogenesis by regulating the interplay between
VE-cadherin and VEGFR2. Dev Cell 2012;23:1264.
37. Garcia JGN, Liu F, Verin AD, et al. Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest
2001;108:689–701.
38. Lee M-J, Thangada S, Claffey KP, et al. Vascular endothelial cell adherens junction assembly and morphogenesis induced by sphingosine-1-phosphate. Cell
1999;99:301–12.
39. Nofer J-R, van der Giet M, Tölle M, et al. HDL induces NO-dependent vasorelaxation via the lysophospholipid receptor S1P3. J Clin Invest 2004;113:569–81.
40. Salomone S, Potts EM, Tyndall S, et al. Analysis of sphingosine 1-phosphate receptors involved in constriction of isolated cerebral arteries with receptor null mice
and pharmacological tools. Br J Pharmacol 2008;153:140–7.
41. Li H, Yin J, Li L, Deng J, Feng C, Zuo Z. Isoflurane postconditioning reduces ischemia-induced nuclear factor-κB activation and interleukin 1β production to
provide neuroprotection in rats and mice. Neurobiol Dis 2013;54:216–24.
42. Sabri M, Lass E, Macdonald RL. Early brain injury: a common mechanism in subarachnoid hemorrhage and global cerebral ischemia. Stroke Res Treat 2013;2013:1–9.
43. Blum FE, Zuo Z. Volatile anesthetics-induced neuroinflammatory and anti-inflammatory responses. Med Gas Res 2013;3:16.
44. Lakhan SE, Kirchgessner A, Hofer M. Inflammatory mechanisms in ischemic stroke: therapeutic approaches. J Transl Med 2009;7:97.
45. Xu X, Kim JA, Zuo Z. Isoflurane preconditioning reduces mouse microglial activation and injury induced by lipopolysaccharide and interferon-γ. Neuroscience
2008;154:1002–8.
46. Tang JX, Eckenhoff MF, Eckenhoff RG. Anesthetic modulation of neuroinflammation in Alzheimer’s disease. Curr Opin Anaesthesiol 2011;24:389–94.
47. Wu X, Lu Y, Dong Y, et al. The inhalation anesthetic isoflurane increases levels of proinflammatory TNF-α, IL-6, and IL-1β. Neurobiol Aging 2012;33:1364–78.
48. Adams SD, Radhakrishnan RS, Helmer KS, Mercer DW. Effects of anesthesia on lipopolysaccharide-induced changes in serum cytokines. J Trauma Inj Infect Crit
Care 2008;65:170–4.
49. Guo W-Z, Miao Y-L, An L-N, et al. Midazolam provides cytoprotective effect during corticosterone-induced damages in rat astrocytes by stimulating steroidogenesis. Neurosci Lett 2013;547:53–8.
50. Abbott NJ. Astrocyte-endothelial interactions and blood–brain barrier permeability. J Anat 2002;200:629–38.
51. Liu Y, Gao M, Ma L, Zhang L, Pan N. Sevoflurane alters the expression of receptors and enzymes involved in Aβ clearance in rats. Acta Anaesthesiol Scand
2013;57:903–10.
52. Hirschy JC, Thorpe JJ, Cortese AF. Meckel’s stones. Radiology 1976;119:19–20.
53. Shen X, Dong Y, Xu Z, et al. Selective anesthesia-induced neuroinflammation in developing mouse brain and cognitive impairment. Anesthesiology 2013;118:502–15.
54. Davidson AJ, Disma N, de Graaff JC, et al. Neurodevelopmental outcome at 2 years of age after general anaesthesia and awake-regional anaesthesia in infancy
(GAS): an international multicentre, randomised controlled trial. Lancet 2016;387:239–50.
55. Eagle KA, Guyton RA, Davidoff R, et al. ACC/AHA 2004 guideline update for coronary artery bypass graft surgery: summary article. J Am Coll Cardiol
2004;44:1146–54.
56. Selim M. Perioperative stroke. N Engl J Med 2007;356:706–13.
57. Coburn M, Fahlenkamp A, Zoremba N, Schaelte G. Postoperative kognitive dysfunktion. Der Anaesthesist 2010;59:177–85.
58. Newman MF, Mathew JP, Grocott HP, et al. Central nervous system injury associated with cardiac surgery. The Lancet 2006;368:694–703.
59. Rinder C. Cellular inflammatory response and clinical outcome in cardiac surgery. Curr Opin Anaesthesiol 2006;19:65–8.
60. Grocott HP. Cognitive dysfunction after cardiac surgery: revisiting etiology. Semin Cardiothorac Vasc Anesth 2005;9:123–9.
61. Okamura T, Ishibashi N, Kumar TS, et al. Hypothermic circulatory arrest increases permeability of the blood–brain barrier in watershed areas. Ann Thorac Surg
2010;90:2001–8.
62. Cavaglia M, Seshadri SG, Marchand JE, Ochocki CL, Mee RBB, Bokesch PM. Increased transcription factor expression and permeability of the blood–brain barrier
associated with cardiopulmonary bypass in lambs. Ann Thorac Surg 2004;78:1418–25.
63. Reinsfelt B, Ricksten S-E, Zetterberg H, Blennow K, Fredén-Lindqvist J, Westerlind A. Cerebrospinal fluid markers of brain injury, inflammation, and blood–brain
barrier dysfunction in cardiac surgery. Ann Thorac Surg 2012;94:549–55.
64. Merino JG, Latour LL, Tso A, et al. Blood–brain barrier disruption after cardiac surgery. Am J Neuroradiol 2012;34:518–23.
65. Nussmeler NA, Arlund C, Slogoff S. Neuropsychiatric complications after cardiopulmonary bypass. Anesthesiology 1986;64:165–70.
66. Zaidan JR, Klochany A, Martin WM, Ziegler JS, Harless DM, Andrews RB. Effect of thiopental on neurologic outcome following coronary artery bypass grafting.
Anesthesiology 1991;74:406–11.
67. Bilotta F, Gelb AW, Stazi E, Titi L, Paoloni FP, Rosa G. Pharmacological perioperative brain neuroprotection: a qualitative review of randomized clinical trials. Br
J Anaesth 2013;110:i113–20.
68. Wang D, Wu X, Li J, Xiao F, Liu X, Meng M. The effect of lidocaine on early postoperative cognitive dysfunction after coronary artery bypass surgery. Anesth Analg
2002;95:1134–41.
69. Mitchell SJ, Merry AF, Frampton C, et al. Cerebral protection by lidocaine during cardiac operations: a follow-up study. Ann Thorac Surg 2009;87:820–5.
70. Mitchell SJ, Pellett O, Gorman DF. Cerebral protection by lidocaine during cardiac operations. Ann Thorac Surg 1999;67:1117–24.
71. Mathew JP, Mackensen GB, Phillips-Bute B, et al. Randomized, double-blinded, placebo controlled study of neuroprotection with lidocaine in cardiac surgery.
Stroke 2009;40:880–7.
72. Nagels W, Demeyere R, Van Hemelrijck J, Vandenbussche E, Gijbels K, Vandermeersch E. Evaluation of the neuroprotective effects of S(+)-ketamine during openheart surgery. Anesth Analg 2004:1595–603.
I. NEUROANATOMY
58
3. BLOOD–BRAIN BARRIER
73. Hudetz JA, Iqbal Z, Gandhi SD, et al. Ketamine attenuates post-operative cognitive dysfunction after cardiac surgery. Acta Anaesth Scand 2009;53:864–72.
74. Roach GW, Newman MF, Murkin JM, et al. Ineffectiveness of burst suppression therapy in mitigating perioperative cerebrovascular dysfunction. Anesthesiology
1999;90:1255–64.
75. Kanbak M, Saricaoglu F, Avci A, Ocal T, Koray Z, Aypar U. Propofol offers no advantage over isoflurane anesthesia for cerebral protection during cardiopulmonary bypass: a preliminary study of S-100ß protein levels. Can J Anesth/J Can Anesth 2004;51:712–7.
76. Shorvon S, Ferlisi M. The treatment of super-refractory status epilepticus: a critical review of available therapies and a clinical treatment protocol. Brain
2011;134:2802–18.
77. Sánchez-Conde P, Rodríguez-López JM, Nicolás JL, et al. The comparative abilities of propofol and sevoflurane to modulate inflammation and oxidative stress in
the kidney after aortic cross-clamping. Anesth Analg 2008;106:371–8.
78. Loop T, Humar M, Pischke S, et al. Thiopental inhibits tumor necrosis factor α-induced activation of nuclear factor κB through suppression of IκB kinase activity.
Anesthesiology 2003;99:360–7.
79. Loop T, Liu Z, Humar M, et al. Thiopental inhibits the activation of nuclear factor κB. Anesthesiology 2002;96:1202–13.
80. Schneemilch CE, Hachenberg T, Ansorge S, Ittenson A, Bank U. Effects of different anaesthetic agents on immune cell function in vitro. Eur J Anaesth 2005;22:616–23.
81. Hsing C-H, Lin M-C, Choi P-C, et al. Anesthetic propofol reduces endotoxic inflammation by inhibiting reactive oxygen species-regulated Akt/IKKβ/NF-κB
signaling. PLoS One 2011;6:e17598.
82. Dale O, Somogyi AA, Li Y, Sullivan T, Shavit Y. Does intraoperative ketamine attenuate inflammatory reactivity following surgery? A systematic review and
meta-analysis. Anesth Analg 2012;115:934–43.
83. van Vliet EA, Aronica E, Gorter JA. Role of blood–brain barrier in temporal lobe epilepsy and pharmacoresistance. Neuroscience 2014;277:455–73.
84. Seiffert E, Dreier JP, Ivens S, et al. Lasting blood–brain barrier disruption induces epileptic focus in the rat somatosensory cortex. J Neurosci 2004;24:7829–36.
85. Ichiyama T, Nishikawa M, Lipton JM, Matsubara T, Takashi H, Furukawa S. Thiopental inhibits NF-κB activation in human glioma cells and experimental brain
inflammation. Brain Res 2001;911:56–61.
86. Welters ID, Hafer G, Menzebach A, et al. Ketamine inhibits transcription factors activator protein 1 and nuclear factor-κB, interleukin-8 production, as well as
CD11b and CD16 expression: studies in human leukocytes and leukocytic cell lines. Anesth Analg 2010;110:934–41.
87. Miller JD, Becker DP, Ward JD, Sullivan HG, Adams WE, Rosner MJ. Significance of intracranial hypertension in severe head injury. J Neurosurg 1977;47:503–16.
88. Marshall LF, Smith RW, Shapiro HM. The outcome with aggressive treatment in severe head injuries. Part II: acute and chronic barbiturate administration in the
management of head injury. J Neurosurg 1979;50:26–30.
89. Marshall LF, Smith RW, Shapiro HM. The outcome with aggressive treatment in severe head injuries. Part I: the significance of intracranial pressure monitoring.
J Neurosurg 1979;50:20–5.
90. Miller JD, Butterworth JF, Gudeman SK, et al. Further experience in the management of severe head injury. J Neurosurg 1981;54:289–99.
91. Rea GL, Rockswold GL. Barbiturate therapy in uncontrolled intracranial hypertension. Neurosurgery 1983;12:401–4.
92. Brain Trauma Foundation, American Association of Neurological Surgeons, Congress of Neurological Surgeons, et al. Guidelines for the management of severe
traumatic brain injury. XI. Anesthetics, analgesics, and sedatives. J Neurotrauma 2007;24(Suppl. 1):S71–6.
93. Eisenberg HM, Frankowski RF, Contant CF, Marshall LF, Walker MD. High-dose barbiturate control of elevated intracranial pressure in patients with severe head
injury. J Neurosurg 1988;69:15–23.
94. Roberts I, Sydenham E. Barbiturates for acute traumatic brain injury. Cochrane Database Syst Rev 2012;12:CD000033.
I. NEUROANATOMY
S E C T I O N
I I
NEUROPHYSIOLOGY
This page intentionally left blank
C H A P T E R
4
Neurophysiology
M. Sethuraman
Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
O U T L I N E
Intracranial Pressure
62
Introduction
62
Normal Intracranial Pressure
62
Cerebral Compliance
62
Importance of Intracranial Pressure
Intracranial Waves
Intracranial Wave Analysis
Factors Influencing the Intracranial Pressure
Waveform
Pathological Intracranial Pressure Waves
Intracranial Pressure–Derived Indices
Pressure–Volume Compensatory Curve
Cerebrovascular Pressure Reactivity
Intracranial Pressure Monitoring Methods
63
64
64
Summary
68
Cerebral Blood Flow
Effects of Temperature and Anesthetic Agents
Measurement of Cerebral Blood Flow
73
73
Summary
74
Brain Metabolism
74
Introduction
74
68
Normal Cerebral Metabolism
Brain Energy is Utilized for the Following Process
Brain Metabolism in Presence of Oxygen
Cerebral Metabolism in Hypoxic State
Cerebral Metabolism in Hypoglycaemic States
Control of Cerebral Metabolism
Metabolic Coupling
Metabolic Uncoupling
Cerebral Metabolism in Pathological States
Apoptosis
Monitoring of Cerebral Metabolism
Cerebral Microdialysis
74
74
75
75
75
76
76
77
77
78
78
78
Introduction
68
Summary
79
Vascular Anatomy
Arterial System
Venous System
Control of Venous Circulation
Regulation of Cerebral Blood Flow
Cerebral Autoregulation
Mechanisms of Autoregulation
Myogenic Control
Neurogenic Control
Flow Metabolism Coupling
Factors Affecting Cerebral Blood Flow
Partial Pressure of Arterial CO2
Effects of Partial Pressure of Arterial Oxygen
Effects of Hematocrit
Effects of Age and Gender
68
68
69
70
70
70
71
71
71
72
72
72
72
73
73
Cerebrospinal Fluid
79
Introduction
79
Ventricular System
Formation of Cerebrospinal Fluid
Cerebrospinal Fluid Circulation
Cerebrospinal Fluid Absorption
Cerebrospinal Fluid Volume and Composition
Regulation of Cerebrospinal Fluid Dynamics
Effects of Anesthetic Agents
Effects of Pathologies on Cerebrospinal Fluid Dynamics
Imaging of Cerebrospinal Fluid Pathways
CT Scan
Magnetic Resonance Imaging
79
79
80
81
81
82
82
82
83
83
83
Summary
83
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00004-X
64
66
66
66
67
68
61
© 2017 Elsevier Inc. All rights reserved.
62
4. NEUROPHYSIOLOGY
The Spinal Cord
83
Introduction
83
Anatomy
84
Organization of the Spinal Cord
White Matter of Spinal Cord
Intrinsic Pathways
Ascending Pathways
Descending Pathways
Spinal Cord Interneurons
Glial Cells of Spinal Cord
Blood Supply
84
84
84
84
84
86
86
86
Intrinsic Supply
Extrinsic Blood Supply
Venous System of Spinal Cord
Pathophysiology
Autoregulation and CO2 Reactivity of the Spinal Cord
Functions of the Spinal Cord
Sensory Function
Motor Function
Autonomic Function
87
87
88
88
88
88
89
89
89
Summary
89
References
89
INTRACRANIAL PRESSURE
INTRODUCTION
Brain is enclosed in the skull which is ought to be a protective layer. However, the rigid skull can be damaging the
brain in case of pathological lesion. The relationship between the skull contents was initially proposed by Alexander
Munro in 1783 and his colleague George Kellie. A modified Munro–Kellie doctrine was proposed by George Burrows
in 1846 by including the cerebrospinal fluid (CSF) component along with brain and blood.1 The current doctrine was
introduced by Harvey Cushing who states that in an intact skull, the volume of brain, blood and CSF remains constant.
Any increase in the volume of one compartment needs to be compensated by the reduction in either one or both
of the other compartments.2 In normal circumstances the intracranial pressure (ICP) is kept in its normal range,
maintaining the relationship between the CSF, blood, and brain tissue constant. Any increase in the ICP can lead to
reduced cerebral perfusion and can be harmful. This chapter describes the normal ICP, factors affecting the ICP, and
overview of the monitoring of ICP.
NORMAL INTRACRANIAL PRESSURE
The ICP is derived from the circulating intracranial blood and CSF (ICPtotal = ICPvascular + ICPCSF). The vascular
component is difficult to quantify due to variation in cerebral blood volume, the autoregulation. The circulatory CSF component may be expressed using Davson’s equation; ICP = (resistance to CSF outflow) × (CSF formation) + (pressure in sagittal sinus).3 The normal values of ICP in different age groups in horizontal position
are 10–15 mmHg for adults and 3–7 mmHg for children. The ICP is lower in newborn (1.5–6 mmHg) due to the
opened cranial sutures and fontanels. In the vertical position it is negative with a mean of around −10 mmHg,
but not exceeding −15 mmHg. ICP is considered to be high if the value exceeds 20 mmHg and needs to be
treated.4 When the ICP exceeds 60 mmHg, cerebral perfusion ceases and there will be irreversible brain damage
and death.
CEREBRAL COMPLIANCE
Since the intracranial volume is inversely proportional to the pressure, a curve can be plotted for change
in pressure for a change in volume.5 This measures the intracranial compliance (Fig. 4.1). The first part of the
curve is characterized by a very limited increase in pressure with increase in volume because compensatory
reserve is large enough to accommodate the extra volume. With further increasing volume, the compensatory
reserve is eventually exceeded, causing a rapid increase in pressure. The normal intracranial compliance is
II. NEUROPHYSIOLOGY
63
IMPORTANCE OF INTRACRANIAL PRESSURE
FIGURE 4.1 Pressure–volume relationship of intracranial pressure.
TABLE 4.1 Stages of Increase in Intracranial Pressure (ICP) With Clinical Symptoms6
Stage
Changes in ICP
Clinical Symptoms
1
Increase in tumor volume, compensatory reduction in cerebrospinal
fluid and blood volume, no raise in ICP
Usually asymptomatic
2
Compensatory mechanisms exhausted, gradual rise in ICP with
increase in volume
Drowsiness, headache
3
Rapid rise in ICP, falling cerebral perfusion pressure
Deterioration of consciousness,
raised blood pressure, bradycardia
4
Cerebral vasomotor paralysis, ICP equals mean arterial blood pressure,
cerebral perfusion ceases
Coma, dilated fixed pupils, and death
60–80 mL in young adults and 100–140 mL in older people due to cerebral atrophy. Depending on the compliance the increase in ICP produces various clinical symptoms. Theoder Koher described four stages of raised
ICP6 (Table 4.1).
IMPORTANCE OF INTRACRANIAL PRESSURE
ICP can be raised due to various pathological processes such as brain tumor, trauma, hemorrhage, ischemia producing a stroke or cerebral edema. The raised ICP has two important consequences:
1. The cerebral perfusion pressure (CPP) is denoted by CPP = MAP−ICP, where MAP is the mean arterial pressure
and ICP is the intracranial pressure. When the ICP increases, the CPP decreases. In addition, there can be
vasomotor paralysis caused by the raised ICP and the intracranial pathological process. Both these factors result
in the reduction of CPP causing ischemia and damage to the neurons.
2. The skull is divided into various partial compartments by the folds of dura mater (right and left hemispheres,
supratentorial and infratentorial compartments). This leads to compartmentalization of the pressure in case of
any pathology. The increase in pressure in one of the compartments pushes the surrounding brain to the other
compartments below the dural folds causing herniation of brain. The various herniation syndromes are given in
Table 4.2 (Fig. 4.2).7
II. NEUROPHYSIOLOGY
64
4. NEUROPHYSIOLOGY
TABLE 4.2 Various Features of Brain Herniation Syndromes
Herniation
Structures Involved
Clinical Feature
Subfalcine herniation
Cingulate gyrus is pushed below the falx
Dilation of contralateral ventricle
With collapse of ipsilateral ventricle
Uncal herniation
Medial temporal lobe structures are displaced
against tentorial edge
Unilateral pupillary dilation with contralateral
hemiplegia
Central herniation
Downward displacement of brain stem and
diencephalon
Cheyne Stokes respiration, bilateral pinpoint pupils,
rapidly progressing to death
Tonsillar herniation
Cerebellar tonsils are pushed downward through
foramen magnum
Hydrocephalus, coma, hemodynamic disturbances,
and respiratory arrest
Transcalvarial
Brain is pushed against a defect in the skull
Effects depend on the brain area involved
FIGURE 4.2 Diagrammatic representation of various brain herniation syndromes.
Intracranial Waves
Monitoring of ICP is important as a guide to diagnosis as well as therapeutic interventions. No universally
accepted guidelines exist for ICP monitoring, and the indications vary considerably between hospitals and physicians. Some of the indications of ICP monitoring include severe head injury, intracerebral hemorrhage, acute stroke,
metabolic encephalopathy, etc.
Intracranial Wave Analysis
Intracranial waveform analysis provides wealth of information regarding the ICP and compliance. It is important to understand the waveform morphology and its changes in different conditions. The pulsations of the CSF
are predominant because of transmission of the arterial pulsations. Jugular venous pulsations and respiration
also affect the waveform. However, in pathological raise it is mainly the arterial waveform that influences the ICP
waves.
Under normotension, normocarbia, and normal PO2, the ICP waveform shows two distinct patterns. A slow,
large sinusoidal wave and a rapid, small peak wave (Fig. 4.3). The slow sinusoidal pattern corresponds to the
phase of respiration (Fig. 4.3A and B). The rapid waves correspond to arterial pulse. The rapid wave has an initial peak, followed by dicrotic peak, and few other peaks after the dicrotic peak. These are referred to P1 (percussion wave corresponds to choroid plexus pulsations), P2 (dicrotic wave), and P3 (tidal wave–venous pulsations)
waves. In normal persons, P1 amplitude will be higher than P2; however, in patients with raised ICP, P2 will
have more amplitude than P1 (Fig. 4.4).
Factors Influencing the Intracranial Pressure Waveform
Various systemic factors can influence the ICP waveforms.8 It is important to understand the effects of these factors while interpreting the waveforms.
II. NEUROPHYSIOLOGY
IMPORTANCE OF INTRACRANIAL PRESSURE
65
FIGURE 4.3 Relationship between intracranial pressure (ICP) waveform (A) with arterial tracing and central venous tracing and changes in
ICP with respiration (B). ABP, arterial blood pressure; CVP, central venous pressure.
FIGURE 4.4 Normal intracranial pressure (ICP) waveform (A) as well as in poor brain compliance (B).
1. Blood pressure: The effects of the changes in blood pressure (BP) depend on the cerebral autoregulation and
intracranial compliance. A transient fall in BP causes reduced cerebral blood flow (CBF), fall in CSF production
and ICP. However, if the fall is severe or sustained, there will be cerebral vasodilation, brain ischemia, cerebral
edema, and rise in ICP. A transient high BP does not affect the ICP within autoregulation. If the BP increases
beyond autoregulation, then the CBF will be pressure passive and the ICP will increase (Fig. 4.3A). The
morphology of the ICP wave changes and the amplitude increases with the shape like arterial pulse waveform.
2. Central venous pressure (CVP): Elevated CVP causes increase in ICP (Fig. 4.3B). The morphology shows more
prominent “a” wave peaks.
3. Respiration: During inspiration there is fall in intrathoracic pressure, CVP and is reflected by fall in ICP and vice
versa in expiration. In addition, the ICP waveform shows sinusoidal pattern corresponding to respiration
(Fig. 4.3B).
4. ICP: In case of increase in ICP two distinct changes occur in ICP waveform. The waveform assumes the shape
of arterial waves and the venous pulsation disappear. The amplitude of the ICP wave increases. In addition,
there will be increase in P2 amplitude compared to P1 wave, and later there will be rounding off without distinct
II. NEUROPHYSIOLOGY
66
4. NEUROPHYSIOLOGY
waves. This is called blunting of waves. P1/P2 ratio of more than 0.8 along with an ICP value of more than 10
has been associated with poor compliance of the intracranial compartment. However, there are differences of
opinion regarding the predictive value of P1/P2 ratio regarding reduced compliance.
5. Partial pressure of O2 (PaO2): In the presence of hypoxia, there was cerebral hyperemia and moderate increase
in ICP. The amplitude of the CSF wave increases and the venous pulsations disappear.
6. Partial pressure of CO2 (PaCO2): The effects of hypercarbia on the ICP waveform is similar to the pattern seen in
increased BP. There will be increased CBF, ICP and the amplitude of the wave increases.
Pathological Intracranial Pressure Waves
The ICP waveform shows three different patterns during increase in ICP. They are of clinical significance.
1. A waves: These are also called plateau waves. A wave comprises a steep rise in ICP from near normal values to
50 mmHg or more, persisting for 5–20 min and then falling sharply. These waves are always pathological and
indicate greatly reduced compliance. They are frequently accompanied by neurological deterioration. As the
baseline ICP increases the magnitude of A wave also increases. A wave is seen when there are sudden painful
stimuli such as endotracheal suctioning to the patient. A waves are associated with severe fall in CPP and needs
to be avoided in the ICU care (Fig. 4.5).
2. B waves: These are rhythmic oscillations that occur every 1–2 min. ICP rises in a crescendo manner to levels
20–30 mmHg higher than baseline and then falls abruptly. These waves were originally always associated with
Cheyne–Stokes respiration. However, they also occur in ventilated patients and are probably related to changes in
cerebrovascular tone and cerebral blood volume. B waves are also indicative of failing intracranial compensation.
3. C waves: These oscillations occur with a frequency of 4–8 per minute and are of smaller amplitude than B waves.
They are synchronous with spontaneous Traub–Hering–Meyer type variations in BP and are probably of limited
pathological significance.
Intracranial Pressure–Derived Indices
There are two indices which are derived from ICP monitoring: (1) pressure–volume compensatory curve and (2)
cerebrovascular pressure reactivity.
Pressure–Volume Compensatory Curve
The pressure–volume compensatory curve measures the degree of compensation that can be achieved due to change in
intracranial volume.9,10 This measures the relationship between the amplitude (A) of the ICP wave and absolute ICP (P).
When the ICP increases the amplitude of the wave also increases; however, the degree of increase in amplitude depends on
the intracranial compliance (Fig. 4.6). The curve to the left of the line indicates good compliance, and the change of amplitude to pressure will be nonlinear whereas to the right on the line there is linear increase in both amplitude and pressure.
The index called RAP [correlation coefficient (R) between AMP amplitude (A) and mean pressure (P); which is
the index of compensatory reserve] can be derived by calculating the linear correlation between consecutive, time
averaged data points of AMP and ICP (usually 40 of such samples are used) acquired over a reasonably long period
FIGURE 4.5 The Lundberg waves of intracranial pressure (ICP).
II. NEUROPHYSIOLOGY
IMPORTANCE OF INTRACRANIAL PRESSURE
67
FIGURE 4.6 The relation between the changes in the amplitude of intracranial pressure (ICP) waves and ICP values.
FIGURE 4.7 The positive PRx (top panel) and negative PRx (bottom panel) along with the curve of compensatory stages. ICP, intracranial
pressure; ABP, arterial blood pressure; PRx, pressure reactivity index.
to average over respiratory and pulse waves (usually 6–10 s periods). This index indicates the degree of correlation
between AMP and mean ICP over short periods of time usually 4 min.
Interpretation: If RAP = 0, there is a good intracranial reserve; if RAP = 1, then the compliance is reduced; if RAP
<0, the compliance is in steeper part of the curve and autoregulation is lost, the amplitude decreases.
Cerebrovascular Pressure Reactivity
The cerebrovascular reactivity is measured using pressure reactivity index (PRx).11 It measures the relationship between
the slow changes in the arterial BP and ICP. It indirectly measures the autoregulatory state. When the cerebrovascular reactivity is intact, changes in BP cause inverse change in cerebral blood volume and ICP. When the autoregulation is lost, then
II. NEUROPHYSIOLOGY
68
4. NEUROPHYSIOLOGY
the increase in BP increases the CBF with a corresponding increase in ICP. PRx is determined by calculating the correlation
coefficient between 40 consecutive, time averaged data points of ICP and arterial blood pressure (Fig. 4.7).
Interpretation: When PRx is negative (−1), it indicates good cerebrovascular reserve (Stage 1).
When PRx = 0, the patient is in compensatory part of the ICP curve (Stages 2 and 3).
When PRx is positive (+1), it implies poor reserve (Stage 4).
Intracranial Pressure Monitoring Methods
ICP can be monitored invasively or noninvasively.12 Invasive methods are more common in use due to their
reliability and continuous monitoring. Intraventricular catheter is the gold standard method, whereas fiber optic
catheters are more advanced techniques of monitoring. The details of different methods of ICP monitoring will be
beyond the scope of this chapter.
SUMMARY
ICP is a key component of the measurement of the intracranial volume/compliance. Many pathological conditions present with features of raised ICP. The symptoms and signs depend on the level of compensation and the
duration of the change. Long-standing and slow-growing lesions may produce mild symptoms, whereas severe and
acute lesions will rapidly progress to various herniation syndromes and death. Hence it is important to understand
the pathophysiology of the disease process and its effect on the ICP so that methods to monitor and treatment plan
can be made accordingly.
CEREBRAL BLOOD FLOW
INTRODUCTION
The brain is 2% of body weight. However, it receives 15–20% of cardiac output, utilizes 20% of the oxygen,
and 25% of the glucose used by the whole body at rest. The high blood flow is due to high metabolic demand
and lack of substrate storage in brain. This high metabolic function is devoted to synaptic activity (50%), maintenance of ionic gradient (25%), and biosynthesis of cellular material and proteins (25%). Hence precise control
of CBF is essential for survival.18 Hence understanding of various factors affecting the CBF becomes important
to prevent its fall. Alterations in CBF can be either etiology or contributing factor in many brain pathological
conditions.19 In addition, various drugs and anesthetic agents alter the CBF differently. This chapter reviews
the normal blood supply of the brain, factors governing the blood supply, and the effect of various pathological
process and drugs.
VASCULAR ANATOMY
Arterial System
Brain is supplied by two internal carotid arteries (anterior circulation) and two vertebral arteries that join to form
basilar artery (posterior circulation). The arteries form anastomoses called circle of Willis (CW). The CW is the pyramidal shaped anastomoses in the interpeduncular cistern formed between anterior and posterior circulation as well
as between arteries of two cerebral hemispheres (Fig. 4.8). The anastomosis was thought to prevent ischemia if blood
supply of any of the major vessels is compromised. However, a recent view on the existence of the CW is it reduces
the pressure within the arterial system. Pressure gradient exists because pulse wave and blood flow arrive into the
skull through different cerebral arteries asynchronously, due to arterial tree asymmetry. Therefore CW and its communicating arteries protect cerebral artery and blood–brain barrier from hemodynamic stress. The CW is formed by
anterior cerebral arteries on either side, anterior communicating artery, internal carotid artery bifurcation, posterior
communicating artery, posterior cerebral artery (PCA) on either side. The PCA is a part of posterior circulation. Rest
of the arteries mentioned above belongs to the anterior circulation.
II. NEUROPHYSIOLOGY
69
VASCULAR ANATOMY
Anterior Cerebral
Artery
Anterior
Communicating
Artery
ICA Bifurcation
Middle
Cerebral
Artery
Posterior
Cerebral
Artery
Posterior
Communicating
Artery
Basilar Artery
Vertebral
Artery
Extracranial
Internal Carotid
Artery (ICA)
FIGURE 4.8 The arterial supply of the brain along with circle of Willis.
Venous System
The cranial veins are classified into three different systems:
1. Superficial—draining the scalp, muscles, and tendons.
2. Intermediate—draining the bone, diploe, and dura mater and consists of emissary veins, diploid veins,
meningeal veins, and venous sinuses.
3. Deep system—veins draining the brain.
Normally the flow scalp veins drain mostly to extracranial venous system and partly through emissary veins to
dural venous sinus. In case of raised intracranial pressure, there will be retrograde flow from intracranial to extracranial venous system resulting in dilation of scalp veins and emissary veins. The diploid and scalp veins serve as
collateral between superior sagittal sinuses to extracranial veins.
The veins of the brain form the superficial and deep venous system. The superficial veins include cortical vein,
and they drain to the superior sagittal and transverse sinus. The veins of the medial and inferior cortical surface
drain to vein of Galen. There are anastomotic channels between superficial and deep venous systems, which
include occipital vein, posterior Calloway vein, and basal vein of Rosenthal. The deep venous system consists of
internal cerebral vein, thalamo striate veins, and vein of Galen. The deep venous system drains of inferior sagittal
II. NEUROPHYSIOLOGY
70
4. NEUROPHYSIOLOGY
FIGURE 4.9 The description of flow metabolism coupling. CBF, cerebral blood flow.
sinus to form straight sinus. The veins of the infratentorial compartment form a network which drains to straight
sinus or transverse sinus.
Control of Venous Circulation
The transmural pressure across the intracranial veins is very low, and as the ICP increases the pressure in the
cortical veins also increases. There is a cuff system at the junction of the venules where it drains to venous sinuses.
The venous sinus pressure remains lower as a result of cuffing of the venules. Animal experiments have found
that among the myogenic, metabolic, and neurogenic control, the venous flow is influenced by the sympathetic
system especially alpha 1 adrenergic mechanism. During resting conditions 70–80% of the cerebral blood volume
is located in the venous system, and it is controlled by cervical sympathetic system. When the ICP increases, the
activation of sympathetic system results in veno constriction and drives the blood away thereby offsetting the
effects of raised ICP. There is little evidence of metabolic and myogenic control of the venous system. In the presence of hypercapnia, the pial venules dilate in response to increased arterial flow produced by arterial dilation.
However, as the PaCO2 raises further, the increased sympathetic activity causes pial–veno constriction and tries
to offset the increased ICP. Acute arterial hypertension causes areas of cortical venous dilation due to increase in
the blood flow. The venous transmural pressure may be higher than ICP and can lead to perivenular disruption
of blood–brain barrier.
Regulation of Cerebral Blood Flow
The normal CBF is 50–55 mL/100 g/min.18 The distribution of CBF is not uniform. The gray matter receives
approximately 80 mL/100 g/min, whereas white matter receives 20 mL/100 g/min (4:1). Blood flow of less than
20 mL/100 g/min is considered to be critical below which irreversible cellular death can occur. The CBF is affected
by many factors (Fig. 4.9). Among them partial pressure of arterial carbon dioxide (PCO2), the MAP, cerebral metabolism, and the autonomic nervous system are the principal regulators of CBF.19 They do not act independently. There
is usually an integration of various factors, which precisely determine the CBF.
Cerebral Autoregulation
Experiments have proved the CBF remains constant despite variation in many local and systemic factors that affect
the blood flow. This ability to maintain a constant CBF with wide variation in CPP is called as cerebral autoregulation
(Fig. 4.10). Beyond the limits of the autoregulation, the CBF depends on the MAP. However, the limits of autoregulation have been questioned.20 This questioning is because of the fact that the autoregulation measurement assumes a
II. NEUROPHYSIOLOGY
VASCULAR ANATOMY
71
FIGURE 4.10 The normal cerebral autoregulation.
linear relationship between the CBF and the BP. However, other factors such as baroreceptor reflex, PaCO2 levels can
also affect the validity of the data. In an analysis of transcranial doppler (TCD), MRI, positron emission tomography
(PET), etc. based on 41 studies, the authors have found that the cerebrovasculature does have autoregulatory capacity, but its efficacy is not perfect and is dependent on the severity and direction of change in perfusion pressure.21
Recent data have found that cerebral autoregulation derived from pressure–flow recordings needs to consider not
only absolute BP as an input to the cerebral circulation, but also whether BP is accelerating or decelerating and rising
or falling.22 It was found that the compliance of the cerebral vessels also is a major factor in the autoregulation. The
brain vessels respond well to rapidly rising BP than a rapidly falling BP.23
There are two types of autoregulation described: static and dynamic. “Static” autoregulation refers to a steadystate relationship between MAP and CBF. It is usually assessed using TCD either by changes in the CBF or by changes
in MAP by administering phenylephrine. “Dynamic” refers to the cerebral pressure–flow relationship during transient changes in MAP. It is usually measured by transient hyperemic response test. However, many studies have found
that the difference between the two is little.21 Recent methods for assessing the continuous autoregulation include
transfer function analysis based on the principle that higher frequency BP fluctuations are more linearly transferred
to the cerebral circulation than lower frequency fluctuations.
Mechanisms of Autoregulation
The principle mechanism by which the cerebral vessels respond to change in the MAP is by changing the cerebral vascular resistance (CVR). The initial thought was that the pial arterioles were primarily responsible for CVR;
however, it was found that the large intracranial arteries are responsible for changing CVR in response to changes in
MAP.23 The postulated mechanism of autoregulation includes neurogenic, myogenic, and local factors.
Myogenic Control
The myogenic theory described by Bayliss in 1902 proposes that autoregulation is caused by the stretching of the
smooth muscle cells of the vessel wall leading to vasoconstriction.24 The mechanism behind it is by the calciummediated pathway in the vascular muscle since it is inhibited by the Ca+2 channel inhibitors. There is an opening
of voltage-gated Ca+2 channel following the stretch leading to entry of calcium and vasoconstriction.25 A calciumindependent mediated theory is also proposed for myogenic action called calcium sensitization, which occurs in the
absence of calcium entry.
Neurogenic Control
A large body of data has been published on the role of autonomic nervous system in the control of CBF. The
cerebral blood vessels, perivasculature innervations, and astrocyte foot process have been identified as a single unit
called “neurovascular unit.” The neurogenic theory suggests that two distinct mechanisms exist in the neural control of CBF. The extrinsic mechanisms are mediated by the superior cervical ganglion (sympathetic), sphenopalatine
ganglion (vagus–parasympathetic), and trigeminal ganglion that innervate the cerebral blood vessels from neck to
II. NEUROPHYSIOLOGY
72
4. NEUROPHYSIOLOGY
Virchow Robin spaces. The sympathetic stimulation leads to the cerebral vasoconstriction up to the upper limit of
CA. The parasympathetic activity has not been fully elucidated. In addition, stimulation of trigeminal nerve leads
calcitonin gene-related peptide (CGRP)–mediated vasodilation.26
The intrinsic mechanism is caused after the vessels leave the Virchow Robin spaces. The nerves originate from the
distal regions of the brain as well as local interneurons which connect to the astrocyte foot process rather than the
blood vessels. These nerves control constriction or dilation of the vessels depending on the stimuli. The exact mechanism is not understood. GABA-mediated vasodilation occurs in the interneurons.
Flow Metabolism Coupling
Even though the brain is only 2% of body weight, it consumes 20% of energy of the whole body. Another unique
feature of the brain energy consumption is the regional differences in the synaptic activity. Much of the energy expenditure is used maintaining the ion homeostasis. Sherrington in the 19th century proposed that the brain possesses an
intrinsic ability to maintain its local blood flow depending on its need which is otherwise its activity.27 This intrinsic
ability is called “flow metabolism coupling” (Fig. 4.9).
Brain is unique in that different function is represented in specific areas. Initiation of a particular function
such as motor movements, speech, etc. results in increase in the blood flow in that area subserving the function.
This is the principle used in functional MRI using blood-oxygen-level dependent contrast imaging signals. The
increase in blood flow was thought to be due to initiation of action potentials.28 However, studies have shown
that increased synaptic transmission during activity leads to increase product of metabolism either adenosine
or changes in ions like K+ and H+ ions. Potassium ions released during synaptic transmission have been shown
to act via K+-dependent ATP channels in the vascular smooth muscle causing vasodilation. Hydrogen ions
also produce vasodilation, but the exact mechanism is not known. It is probably thought to be similar to CO2induced vasodilation or via nitric oxide pathways.29 Among the metabolites, adenosine appears to be a potent
vasodilator. Extracellular levels of adenosine were found to increase with neuronal activity, and topical application of adenosine to cerebral microcirculation causes vasodilation.30
Factors Affecting Cerebral Blood Flow
Partial Pressure of Arterial CO2
Changes in the PaCO2 affect the CBF as cerebral circulation is very sensitive to the levels of PaCO2. Studies have
shown an approximate 3–6% increase and/or 1–3% decrease in flow per mmHg in PaCO2 above and below eucapnoeic PaCO2, respectively.31 All the vessels of cerebral circulation such as large arteries of the neck, intracranial vessels, pial arterioles are sensitive to changes in PaCO2. The CO2 reactivity of the microvasculature in gray matter is
greater than that of white matter because of relatively less vascularization of white matter. The mechanism by which
PaCO2 acts is not known. It is thought to exert action by directly acting on the smooth muscle of the blood vessels or
by changes in local tissue pH. The actions of PaCO2 occur regardless of blood pH.
An increase in PaCO2 causes cerebral vasodilation and hypocapnia causes vasoconstriction.32 The lower limit of
CA has been found to shift upwards in acute hypercapnia. The lower limit of CA of 50–60 mmHg under normocapnia (PaCO2 = 30–50 mmHg) was shifted to 80–100 mmHg when hypercapnia (PaCO2 = 70–95 mmHg) was instituted.33
The upper limit in hypercapnia was found to be limited leading to a situation in which the CA limits were shorter in
hypercapnia conditions.33 In severe hypercapnia, the blood flow is pressure dependent. It is also found that PaCO2
of more than 55 mmHg abolishes CA (Fig. 4.11).
During hypocapnia, there is vasoconstriction of cerebral blood vessels. The effect on CA is well established than
hypercapnia. Hypocapnia has been found to decrease the plateau of CA curve; the change in the lower limit was not
significant compared to normocapnia; however, data on the upper limit has not been elucidated.32
Effects of Partial Pressure of Arterial Oxygen
The changes in PaO2 under normoxic conditions are less pronounced compared to PaCO2. However, when the
PaO2 is below 50 mmHg, there is cerebral vasodilation. The effects of the changes in PaO2 depend on the PaCO2 levels.
Studies have shown that hypercapnia increases and hypocapnia decreases cerebrovascular sensitivity to hypoxia.34
The mechanism by which hypoxia acts is thought to be by (1) products of metabolism in the ECF, (2) stimulus from
local neurovascular coupling, or (3) adenosine.
Isocapnic hypoxia below 50 mmHg produces cerebral vasodilation. However, in clinical situations, when there is low
CBF, there is excessive ventilatory drive to wash out the PaCO2 leading to vasoconstriction. This hypocapnia opposes
the hypoxia-induced vasodilation and the net results in minimal change or further reduction in CBF.35 Compared to
II. NEUROPHYSIOLOGY
VASCULAR ANATOMY
73
FIGURE 4.11 The effect of partial pressure of CO2 (PaCO2) (hypocapnia and hypercapnia) on the autoregulatory curve.
acute hypoxia, effects of chronic hypoxia on CBF are complex. The data are obtained from high altitude climbing.
Initially (3–5 days), there is an increase in CBF of approximately 20% after which it tends to fall. After 2 weeks the CBF
returns to normal level, which is thought to be due to adaptation of hypoxia by increase in hematocrit, and mild.36
Effects of Hematocrit
Level of hemoglobin (Hb) or hematocrit (Hct) is one of the factors affecting the CBF. Since oxygen content is a
major factor in the delivery of oxygen changes, the level of Hct can impact the oxygen delivery and cerebral metabolism. The optimum level of Hct is a matter of big debate. Higher Hct >44 has been shown to increase the blood viscosity and reduce CBF.37 Hct <28 has been shown to increase the CBF.38 The mechanism of increase in CBF is thought to
be due to vasodilation induced by either low viscosity or low oxygen content. It has been found in a recent experiment that combination of low Hct and hypercapnia is detrimental as the autoregulation is abolished.39 In such cases,
occurrence of hypotension in patients with low Hct can lead to severe ischemia. Various studies have shown that low
Hct of less than 25 or Hb <7 g% has been associated with poor outcome.40
Effects of Age and Gender
Age has been found to be an important factor in the CBF. Studies have shown that there is a decrease in the mean
FV in TCD as the age advances. The mean FV was found to decrease from 74 cm/s at 20–39 years to 58 cm/s in persons
older than 60 years.41 In addition to the peak systolic flow, diastolic flow also decreases, and the pulsatality index and
resistance index increase as age advances.41 Moreover the difference in FV gets exaggerated in patients with SAH.
Females upto 60 years have been found to have higher FV compared to males after which the differences are abolished.41
Effects of Temperature and Anesthetic Agents
Hypothermia causes reduction in the CBF, and maximum reduction is seen at 18°C whereas the suppression of
metabolism has been found to continue below 18°C. Temperatures below 18°C are associated with loss of autoregulation and vulnerable to ischemia.42 Hyperthermia during exercise has been shown to reduce global CBF without
reduction in cerebral metabolism. Hence hyperthermia is detrimental to brain.43 The anesthetic agents have variable effects on the CBF depending on the type (intravenous vs. inhalational) and concentration used and has been
described elsewhere.
Measurement of Cerebral Blood Flow
Since CBF and cerebral oxygenation are major determinants of brain function, it is essential that a technique be
available to monitor in various situations. CBF measurement has been advocated in traumatic brain injury, stroke,
deep circulatory arrest, cardiopulmonary bypass, etc. Such techniques need to be simple, accurate, capable of measuring both global as well as regional CBF, noninvasive, and cost effective. Various techniques are available to measure the CBF.44 Unfortunately, the accurate estimation of CBF is done by invasive methods and requires complex
II. NEUROPHYSIOLOGY
74
4. NEUROPHYSIOLOGY
equipment, more expensive, and cannot be used bedside. Currently CT- and MRI-based techniques such as PET,
SPECT scan are widely used for clinical use compared to radiolabeled techniques. Methods which are noninvasive,
such as TCD, optical, or laser systems, do not measure the CBF directly, but only the FV which is a surrogate marker of
CBF. Moreover, the values are affected by various local and systemic factors to be used for clinical/research purpose.
SUMMARY
CBF is one of the major determinants of brain function, and various factors can affect the autoregulation, metabolism, and function. Both elevated as well as reduced blood flow can be detrimental to the brain function. It is affected
by both intrinsic as well as extrinsic factors. The measurement of CBF is a challenging process due to limited availability of bedside techniques. It is important to understand the changes in CBF in different pathological conditions
so that optimum CBF can be ensured.
BRAIN METABOLISM
INTRODUCTION
Brain is one of the organs in the body with high blood flow as well as metabolism. In addition to basic needs for
cellular energy, large amount of energy is needed to maintain the neurological function especially ion homeostasis.
Brain utilizes glucose as a main substrate for energy production.45 Alternate sources of energy include ketone bodies
in case of starvation and lactate in case of high physical activity.46 The cerebral metabolism can be altered in various
physiological as well as pathological states. It is important to understand the normal cerebral metabolism, its monitoring, and various factors that alter the metabolism. In this overview, we will discuss the normal metabolism of the
brain and how it is altered in pathological conditions.
NORMAL CEREBRAL METABOLISM
Unlike other organs of the body, brain is totally dependent on the glucose for its energy.45 No other substances
have been found as energy source under normal conditions. The understanding of the cerebral metabolism comes
from the measurements of arteriovenous differences of glucose, oxygen, CO2. The normal, conscious human brain
consumes oxygen at a rate of 156 µmol/100 g tissue/min. CO2 production is the same, leading to a respiratory quotient of 1.0, which shows that the carbohydrate is the ultimate substrate for oxidative metabolism. For complete oxidation of glucose, the theoretical ratio of O2:glucose utilization is 6.0; but the measured ratio in brain is 5.5. The actual
glucose utilization measured is 31 µmol/100 g/min, which indicates that glucose consumption is not only sufficient
to account for total O2 consumption but is in excess by 5 µmol/100 g/min. The excess amount of glucose is thought
to be utilized for synthesis of chemical constituents.47
Brain Energy is Utilized for the Following Process
1. In the resting state, there are more Na+ ion outside the cell and more K+ ion inside the cell. There is a tendency
for the ions to move down the electrochemical gradient. In the resting state, there is leakage of K+ ion outside
the cell and Na+ inside the cells. The inside of the cell is kept at an electronegativity of −70 mV by means of the
ion transporters. If there were no ion transporters there will be depolarization to 0 mV, and there will be cell
death. During the period of activity, there is opening of the Na+ and Ca+2 ion channels which leads to rapid
depolarization of the cell membrane and generation of the action potentials. After the spread of the action
potential, the cell should come back to its hyper polarization state. This is possible only by moving the Na+ and
Ca+2 ions against concentration gradient to ECF. The ion transporters and cotransporters pump out these ions
to bring the cell back to its resting state. This is an energy-dependent process. Effective functioning of the ion
transporters and cotransporters require large amount of energy.48
2. Various cellular organelles such as vesicles, mitochondria, endoplasmic reticulum are involved in the catabolism
and synthesis of various proteins, enzymes, neurotransmitters that require energy.
3. Many of the neurotransmitters are synthesized in the cell body of the neuron and need to be transported to the
axons for release, which require energy.
II. NEUROPHYSIOLOGY
NORMAL CEREBRAL METABOLISM
75
FIGURE 4.12 The normal metabolic pathway of glucose utilization by the brain.
Brain Metabolism in Presence of Oxygen
Glucose from the blood is transported via carrier-mediated mechanism via Glut 1 transporter to the brain ECF.
From the brain ECF it is transported into the neuronal cells via Glut 3 cotransporter. Inside the cell the glucose is
converted to glucose 6 phosphate, which is utilized in one of the three pathways (Fig. 4.12):
1. It enters the glycolytic pathway which is the most common destination of the glucose.
2. It enters pentose phosphate pathway.
3. It enters the glycogenesis in astrocytes.
In the presence of oxygen, glucose enters the glycolytic pathway and the end product is generation of pyruvate
molecules as well as two molecules of ATP. The pyruvate enters the TCA cycle in which additional 34 molecules of
ATP are generated.
Cerebral Metabolism in Hypoxic State
If there is lack of oxygen the pyruvate enters anaerobic glycolysis and lactate and H+ ion is produced. During this
process two ATP is released compared to 34 ATP in oxidative glycolysis. The resultant H+ ions cause a cellular acidosis and block the enzymatic process and ion transport. Therefore the end result of oxygen deprivation is intracellular
acidosis and cell death. It should be borne in mind in case of severe ischemia that supply of both oxygen as well as
glucose is limited resulting in accelerated cell death.
Cerebral Metabolism in Hypoglycaemic States
In case of hypoglycemia, the brain tries to utilize alternate source of energy. During periods of low glucose levels,
the body tries to produce glucose from alternate sources such as stored glycogen from liver, amino acids, and fat.
Once these stores are exhausted a state of ketosis occurs, where the blood levels of ketone bodies especially beta
II. NEUROPHYSIOLOGY
76
4. NEUROPHYSIOLOGY
hydroxy butyrate and acetoacetate increase. Normal brain does not utilize these ketone bodies due to the presence
of glucose. However, in states of ketosis, the ketones enter the neurons from blood via cotransport mechanisms. The
brain has enzymes to utilize the ketone bodies, and acetyl CoA is formed. Acetyl CoA enters the citric acid cycle and
produces ATPs. Although it produces ATPs, it is an inefficient way of metabolism for the brain. If the hypoglycemia
resolves, the metabolism of brain shifts back to normal.
Additional source of energy for the brain in hypoglycemic states is derived from lactate. Normal blood lactate
levels are 0.5–1 mmol/dL. Under normal conditions lactate is exported from the brain to blood. The brain utilizes
lactate less than 10% as energy source. In case of low energy supply as in hypoglycemia, lactate enters the neurons
of the brain via astrocyte, and the lactate can be up to 60% as substrate for oxidative phosphorylation. The entry of
lactate is called astrocyte-to-neuron lactate shuttle.48
Control of Cerebral Metabolism
Metabolic Coupling
One of the most important features of the cerebral metabolism is the flow-metabolism coupling and the neuron-glia
metabolic coupling.49 Neuronal activity is both spatial and temporal in nature. Whenever there is increased activity of
specific neuronal population, there is increase in regional blood flow, increased uptake of oxygen and glucose. This
is called flow-metabolism coupling. The mechanism by which the metabolism is coupled with activity is described by
the neuron-glia coupling (Fig. 4.13).
Major proportion of brain energy is needed to maintain the ion balance and neurotransmitter uptake. Synaptic
potentials, rather than action potentials, appear to represent by far the main energetic cost related to maintenance of
excitability. Excitatory synapses largely dominate in the gray matter, glutamatergic ones alone represent at least 80%
of cortical synapses suggesting that excitatory neurotransmission accounts for most of the energy requirements at the
cortical level. There is a close relationship between brain activity, glutamatergic neurotransmission, energy requirements, and glucose utilization.
Glutamate is the major neurotransmitter of the brain which is released from the presynaptic terminals to synaptic
cleft. The glutamate is stored in vesicles and present in high concentration in the cytoplasm of neurons. The glutamate inside the cell is nontoxic to neurons. However, once they are released in synaptic cleft, it can potentiate the
excitatory pathway and cause Ca+2 influx which can damage the neurons. Hence it needs to be removed from synaptic cleft. Once released, the neurons are incapable of glutamate uptake. The released glutamate is taken up by the
astrocytes and converted to glutamine. The glutamine is released into brain ECF and taken up by neurons, which will
convert it to glutamate for future release. This process is called glutamate–glutamine cycle and anaplerotic pathway.
The astrocytes are the only structures which can generate glutamate from glucose because of the presence of enzyme
pyruvate carboxylase. This process is an energy-dependent process. Another important role in the neuron-glia coupling is the lactate shuttle50 (Fig. 4.14).
FIGURE 4.13 The neuron-glial coupling in the energy metabolism.
II. NEUROPHYSIOLOGY
NORMAL CEREBRAL METABOLISM
77
As already mentioned, the intracellular glucose can be utilized by three pathways in neurons. However, the neurons lack capacity for formation of fructose-2,6-biphosphate, which is a key product in the glycolytic pathway. Hence
the glycolytic pathway is slow inside the neurons, and majority of glucose is diverted via the “pentose phosphate
pathway” which is important in forming NADPH from NADP. NADPH is a potent antioxidant that protects the
neurons from overexpression of oxygen and apoptosis. Hence for energy source the neurons rely on the astrocytes.
Astrocytes produce lactate from the glycolytic pathway, and the lactate is taken by the neurons. The lactate is utilized
in the citric acid cycle to produce 30–34 ATPs. Thus the energy of the neurons is spent efficiently by slowing a glycolytic process which yields only two ATPs as well as reducing the oxygen utility.
Metabolic Uncoupling
During physical activity increased amount of lactate is produced from the muscle that raises the serum lactate
levels. Coupled with increased physical activity there is increased metabolism in the brain. The sympathetic
stimulation associated with exercise increases the CBF, CMRO2, and cerebral glucose uptake. It was found that
there is a metabolic uncoupling in these states, i.e., uptake of carbohydrates is more than the CMRO2. Cerebral
metabolic uncoupling is a consequence of increased lactate availability leading both to passive lactate influx
and increased cerebral dependence on lactate as fuel. The uptake of lactate depends on the cerebral metabolic
activity and is independent of sympathetic stimulation. The lactate is used as a substitute for glucose by the
brain.
Cerebral Metabolism in Pathological States
When the CBF reduces to critically low levels, there is reduction in substrates such as glucose and oxygen delivery to the neurons. The cells will switch over from aerobic to anerobic metabolism. This leads to reduction in the
number of ATPs produced in anerobic (2) compared to that in aerobic (32) metabolism. The energy required for the
ionic homeostasis is not matched with the supply. The uptake of glutamate released from the presynaptic neurons is
inhibited, and the glutamate accumulates in the synaptic vesicles. The glutamate causes stimulation of the postsynaptic neurons via the AMPA and NMDA receptor leading to persistent depolarization. The Ca+2 and Na+ channels
opening for longer duration cause massive influx of calcium and sodium into the cell. Moreover, the lower energy
prevents the membrane-bound ion exchangers to function adequately. The high cytoplasmic calcium level is thought
to trigger a number of events that lead to the ischemic damage. These include increasing the activity of proteases
and phospholipases. These enzymes convert membrane lipid molecules to products of arachnoid acid metabolism.
FIGURE 4.14 The astrocyte–neuron lactate shuttle.
II. NEUROPHYSIOLOGY
78
4. NEUROPHYSIOLOGY
In addition there is release of oxygen-free radicals. All these events lead to cell death and disruption of blood–brain
barrier, edema formation. Later stages lead to infiltration of the ischemic area by the macrophages causing necrosis.
Apoptosis
The process of necrosis described above is a severe form of neuronal damage. There are other forms of cell injury
in which the neurons die, but do not disintegrate. There is no inflammation and immune activation. This process is
called apoptosis. In apoptosis there is expression of genes such as c-Jun and c-Fos. These genes express the formation of new proteins that can be either apoptotic or antiapoptotic. The final fate of the neurons to survive or to die
depends on which of the protein action dominates.
Monitoring of Cerebral Metabolism
Normal cerebral metabolism is affected in various pathological conditions that can result in deficiency in substrate
supply such as glucose or oxygen which in turn depend on the CBF. This can result in anaerobic metabolism and if
severe, results in cell death. It is therefore important to monitor the cerebral metabolism. The monitoring modalities
include global as well as regional techniques. Global metabolism is usually assessed using jugular venous oximetry
whereas regional metabolism can be assessed noninvasively by near-infrared spectroscopy (NIRS) and invasively
cerebral microdialysis.
Jugular Venous Oximetry
Jugular venous oximetry is a measure of global oxygen utilization of the brain.51 The basic principle lies on the fact
that the blood draining the jugular vein is from both the hemispheres (70% ipsilateral, 30% from contralateral side),
there is a dominance usually on the right side; blood sampled from the jugular bulb is predominantly drained from
brain whereas anything below can be contaminated from an extracranial source. When there is increased demand of
oxygen, brain extracts more oxygen resulting in reduced jugular venous oximetry.
Normal values for jugular venous oxygen saturation (SjVO2) are 55–75%. If the value falls less than 55%, it shows
increased oxygen extraction due to imbalance in supply–demand ratio. However a value of more than 75% can be
due to either excess blood supply or decreased demand as in a metabolically suppressed state such as hypothermia,
barbiturate coma. A SjVO2 of less than 50% has been associated with cerebral anaerobic metabolism, <40% with
electroencephalogram slowing, <30% with unconsciousness or low Glasgow coma scale score.
In addition to the SjvO2 values, the information obtained from jugular venous oximetry helps in deriving other
values of cerebral circulation such as cerebral metabolic demand for oxygen (CMRO2), arteriovenous oxygen difference (AjvDO2) = CMRO2/CBF.
Near-Infrared Spectroscopy
NIRS provides a real-time continuous measurement of regional cerebral blood oxygenation and indirect blood
flow. NIRS uses the principle of reflectance spectrophotometry.52
NIRS has been used to measure rSO2 in a variety of clinical conditions. In patients with SAH it is used during clipping of aneurysm, early detection of vasospasm. It is also used for managing head-injured patients,
carotid endarterectomy, cardiac surgery during cardiopulmonary bypass. There are limitations of NIRS use.
The absorption by extra skull tissues, presence of hematomas, other pigments such as melanin, bilirubin can
give false values. The skull thickness as well as hemoglobin concentration also interferes with the accuracy of
monitoring.
Cerebral Microdialysis
The concept of cerebral microdialysis originated in 1960 for assessment of brain ECF composition, especially
amino acids. Progression in science has led to development of commercially available cerebral microdialysis catheter
and dialyzate analyzer.53
Cerebral microdialysis is used to monitor brain ischemia.
1. Head injury: Microdialysis has been found to be useful in assessing the patients, as a guide for lower limit of
autoregulation and optimization of CPP, adjusting the hyperventilation, guiding the surgical planning.
2. Subarachnoid hemorrhage: Microdialysis is useful in early detection of vasospasm and delayed ischemia
neurological damage and guiding the triple H therapy.
3. Other potential benefits for cerebral microdialysis includes drug and substrate delivery. This application is in
research and yet to be utilized to its potential.
II. NEUROPHYSIOLOGY
VENTRICULAR SYSTEM
79
Positron Emission Tomography
Positron emission tomography is useful in assessing the cerebral metabolism for glucose, CMRO2, and CBF. The
neurometabolic and flow metabolic coupling form the basis of the PET scan. The PET/CT or PET/MRI can be used;
many of them prefer the later investigation. The glucose utilization can be assessed using 18F-fluoro-2-deoxyglucose.
The tracer is injected into the vein and after approximately 30 min the scan is performed. PET scan for glucose metabolism is useful in epilepsy and degenerative conditions such as Parkinsonism, Alzheimer disease, etc.
SUMMARY
The cerebral metabolism is unique that it is continuously dependent of the substrate supply from the brain.
Advances in technology have helped us to understand the basic process of cerebral metabolism. At present
many of these monitors are limited to research tools. However, there are limited studies on the ways to improve
the metabolism in ICU as well as OT setting and how the therapeutic interventions improve the outcome in
these patients.
CEREBROSPINAL FLUID
INTRODUCTION
The CSF is the fluid covering the brain and spinal cord. The main function of the CSF initially was thought to
reduce the buoyancy of the brain. However, evidence shows it has many other important functions, especially the
regulation of neuronal function.13 The CSF composition and secretion is altered in various disease processes. Hence
it is important to understand the physiology of CSF which will be elaborated in this chapter.
VENTRICULAR SYSTEM
The interior of the brain contains cavities filled with CSF. The lateral ventricle is present one in each cerebral hemisphere and a median cavity, the third ventricle between the hemispheres. The fourth ventricle is located dorsal to
pons and medulla. The ventricles communicate with each other through various foramen as well as to central canal
of spinal cord and subarachnoid spaces. The cavity of ventricle is covered with ependymal cells.
Formation of Cerebrospinal Fluid
CSF is formed by several regions of the central nervous system (CNS). The choroid plexus of the ventricles forms
the main bulk (two-thirds) of the CSF. The remaining is formed from extrachoroidal sources.
Choroidal secretion: Microscopically the choroid plexus consists of layer of capillary endothelium, matrix, and
the lining epithelium (Fig. 4.15). The choroid plexuses have granular villous protrusions into the ventricular lumen,
the epithelial surface of which is continuous with the ependyma. Each villus is composed of a single layer of epithelial cells overlying a core of connective tissue and blood capillaries. The capillaries in the choroid plexus, unlike those
in the majority of the cerebral circulation, are fenestrated and hence provide little resistance to the movement of small
molecules, ions, and water. They comprise a tuft of fenestrated capillaries.
A barrier is formed by the junctional complexes between the epithelial cells, which restrict the passage of molecules
and ions freely into the CSF. Choroidal cells are epithelial cells which have microvilli at their apical pole and are interconnected by tight junctions with a variable distribution according to the site on the ventricular wall. The choroid
plexus epithelial cells form the “blood–CSF barrier.”
CSF formation occurs in two stages: passive filtration of fluid across choroidal capillary endothelium along
the pressure gradient into the choroidal interstitium, followed by a regulated active secretion across a singlelayered epithelium from the interstitium into ventricles.14 The solute movement from blood to CSF occurs by
diffusion, facilitated transport, and active transport. A blood flow of about 4 mL/min/g is converted to a CSF
formation of approximately 0.4 mL/min/g. CSF formation begins as plasma is filtered across permeable choroidal capillaries. Net filtration is proportional to the hydrostatic pressure gradient between blood and choroid
II. NEUROPHYSIOLOGY
80
4. NEUROPHYSIOLOGY
FIGURE 4.15 The structure of the choroid plexus.
interstitial fluid. Hence a lower BP as well as increased interstitial pressure as occurs in raised ICP reduces the
formation of CSF.
The choroid plexus epithelium is polarized both structurally and functionally. Actively formed CSF stems from
coordinated secretion of solutes across the thin epithelial interface between the inner choroidal plasma and outer
ventricular fluid. Although bidirectional transport of ions and water can occur, CSF formation is mainly by the passage of the Na+, K+, HCO3− from interstitium to ventricles. Ions and water are taken up by facilitating mechanisms
at the basolateral membrane, convected through cytoplasm, and then released or actively secreted by the epithelium
into the ventricles on the apical side (Fig. 4.16).
Cytoplasmic carbonic anhydrase catalyzes the formation of H + and HCO3− ions from water and CO2. The carrier proteins of basolateral membranes of choroidal cells exchange H+ and HCO3− ions for Na+ and Cl− ions. ATPdependent ion pumps of the apical membrane expel Na+, Cl−, HCO3−, and K+ ions toward the ventricular lumen.
Water transport is facilitated by aquaporins I of the apical membrane, and it follows the osmotic gradients generated
by these pumps. The NaK2Cl cotransporter of the apical membrane generates ion transport in both directions and
participates in regulation of CSF secretion and composition.14
Extrachoroidal secretion: Extrachoroidal secretion of CSF is derived from the brain extracellular fluid (ECF), cerebral capillaries across blood–brain barrier and ependymal cells. Under normal conditions, this pathway appears to
be less significant source of CSF formation.
Cerebrospinal Fluid Circulation
The CSF flows from the lateral to the third ventricle via the cerebral aqueduct and the fourth ventricle to subarachnoid space in cisterna magna and subarachnoid spaces. The flow of CSF across the ventricles is pulsatile, unidirectional, and is caused by the transmission of the arterial pulsations. Ventricular CSF also flows by another route
to the basal and midbrain cisterns, i.e., into subarachnoid extensions of the velum interpositum (from dorsal third
ventricle) and superior medullary velum (rostral fourth ventricle). Thereafter fluid is convected to the subarachnoid
spaces of the spinal cord and brain convexities. As CSF flows through the ventriculosubarachnoid system, there are
diffusional and bulk flow exchanges between CSF and brain depending upon region-specific gradients for concentration and hydrostatic pressure that promote widespread distribution of CSF-borne materials. From the subarachnoid
II. NEUROPHYSIOLOGY
VENTRICULAR SYSTEM
81
FIGURE 4.16 The secretion of various substances into the cerebrospinal fluid.
spaces the CSF moves cranially over the surface of the brain to the superior sagittal sinus from where it is absorbed
and caudally to the spinal subarachnoid spaces.
Cerebrospinal Fluid Absorption
In humans, major portion of CSF is absorbed through the arachnoid villi into superior sagittal sinus especially
in its posterior part. The arachnoid villi are projections of the arachnoid matter through defects in the dura of the
sinus at the embryonic life. The arachnoid villi consist of thin layer of endothelium. The pressure gradient between
subarachnoid spaces and the venous sinus necessary to ensure CSF drainage is between 3 and 5 mmHg.15 Each
arachnoid villus was thought to have a one-way (CSF outward) valve-like mechanism that opened in response to a
positive hydrostatic pressure gradient between CSF and dural venous blood. This belief has been questioned with
recent research showing that bulk of CSF flow is absorbed through olfactory and optic nerve sheath, via cribriform
plate to nasal mucosa and cervical lymph in animals.14 The absorption via arachnoid villi occurs only in case of raised
CSF pressure. However, evidence is lacking in humans to this type of absorption.
Spinal arachnoid villi in contact with the epidural venous plexus represent a pathway of CSF absorption especially during effort. In man, arachnoid villi in lumbosacral nerve roots increase CSF absorption in the upright position in response to gravity, and the absorbed CSF then enters the lymphatic system.15
Cerebrospinal Fluid Volume and Composition
The CSF volume is estimated to be about 150 mL in adults. It is distributed between 125 mL in cranial and spinal
subarachnoid spaces and 25 mL in the ventricles. CSF secretion in adults varies between 400 and 600 mL/day or 0.3–
0.6 mL/min/g. CSF is renewed four to five times per 24 h in young adults. In old age the CSF volumes are more due
to atrophy of the brain and there is low turnover of CSF. The composition of CSF is different than plasma in which it
has less potassium, calcium, glucose, and proteins, whereas it contains more chloride and magnesium compared to
plasma (Table 4.3).
II. NEUROPHYSIOLOGY
82
4. NEUROPHYSIOLOGY
TABLE 4.3 Composition of the Cerebrospinal Fluid (CSF) Compared to Plasma
Component
Plasma
CSF
Na+ (mmol/dL)
140
141
K+ (mmol/dL)
4.6
2.9
Cl− (mmol/dL)
101
124
Ca+2
5
2.5
1.7
2.4
23
21
Glucose (mg/dL)
90
54
Proteins (mg/dL)
700
4.3
Osmolarity
305
298
pH
7.4
7.3
PaCO2 (mmHg)
41
51
(mmol/dL)
Mg+2
(mmol/dL)
HCO3−
(mmol/dL)
Regulation of Cerebrospinal Fluid Dynamics
CSF production, circulation, and absorption can be affected by pathological process or by administration of
the drugs. The choroid plexus receives cholinergic, adrenergic autonomic innervation. The sympathetic stimulation reduces CSF secretion whereas the cholinergic stimulation causes increased CSF secretion. In addition to
autonomic innervation, choroid plexus has seretonergic and peptigergic systems. Monoamines and neuropeptide
factors have also been shown to play a role. Dopamine, serotonin, melatonin, atrial natriuretic peptide (ANP),
and arginine vasopressin receptors are present on the surface of choroidal epithelium. Alteration in the ANP and
vasopressin receptors has been thought to be involved in the pathophysiology of hydrocephalus and dementia of
Alzheimer type.
Drugs such as loop diuretics and carbonic anhydrase inhibitors act on the transporter mechanisms and reduce
the CSF secretion. In patients with raised ICP, especially hydrocephalus, the pressure gradient between the plasma
and CSF is reduced leading to reduced secretion of CSF as well as diversion of CSF from intracranial to spinal
compartments.
Effects of Anesthetic Agents
Anesthetic agents have variable effects on the CSF rate of formation (Vf) as well as resistance to absorption (Ra).
Among the intravenous agents ketamine, in the anesthetic doses, increases Ra, but has no effect on Vf. Other intravenous anesthetic agents such as thiopentone, etomidate, and propofol tend to decrease both Vf and Ra, especially
in higher dose range.
Inhalational agents have different effects compared to intravenous agents. Nitrous oxide has no effect on Vf and
Ra. Isoflurane has variable effects on Vf and Ra depending on the concentration. At higher doses it decreases Ra.
In other situations they have either no effect on Vf and Ra or decrease Vf. Sevoflurane reduces Vf and increases Ra.
Halothane decreases Vf and increases Ra. Desflurane has variable effects depending on PaCO2 and ICP levels. Under
normocapnia, desflurane increases CSF pressure whereas in hypocapnia it decreases CSF pressure.
Midazolam appears to have variable effects on the CSF dynamic depending on the dose. Low-dose fentanyl has
no effect on Vf and it decreases Ra, whereas high-dose fentanyl reduced Vf. Sufentanil was found to have no effect
on Vf, but reduced Ra.
Effects of Pathologies on Cerebrospinal Fluid Dynamics
1. Head injury: Patients with head injury may have high ICP; however, the Vf has not changed in head-injured
patients, whereas the Ra was increased contributing to additional cause of raised ICP in 20% of patients.
II. NEUROPHYSIOLOGY
INTRODUCTION
83
2. Intracranial tumors: The intracranial mass lesion can compress the CSF spaces and shift the CSF to spinal from
intracranial compartment. Depending on the speed with which the ICP increases the effects on CSF dynamics are
affected. Since CSF rate of formation depends on CBF, acute increase in ICP can compromise CBF and can reduce CSF
production whereas Ra is increased. A slow growing mass lesion has limited effects on Vf, whereas it can increase Ra.
3. Bacterial meningitis: In bacterial meningitis there is increased Ra which is a major factor in the increase in ICP
seen in meningitis.
4. Pseudotumor cerebri: There are multiple mechanisms for raised ICP in this condition. An increase in Vf, Ra,
CBV, and CBF all have been thought to be responsible. Prednisolone treatment has been found to reduce Ra.
5. Subarachnoid hemorrhage (SAH): Patients with SAH can present with hydrocephalus. Various studies have
shown that the presence of blood in the subarachnoid space increases Ra leading to raised ICP.16
Imaging of Cerebrospinal Fluid Pathways
Various pathological lesions can affect the CSF pathways. The earliest one used was cisternal puncture and injecting air or contrast agents and using X-rays to assess the ventricular size and pathology. This is no more in vogue.
Currently the CT scans and magnetic resonance imaging (MRI) are used for assessing the CSF pathways.
CT Scan
CT scan of the brain is the initial imaging of choice for identifying the ventricle. The normal CSF spaces will
appear dark in the CT films. The size of the different ventricles, the cisterns can be seen well with CT. Lesions such
as dilated ventricles, slit ventricles, tumors, etc., can be readily delineated in CT imaging. Contrast enhanced CT will
help in visualizing the choroid plexus and its pathology.
Magnetic Resonance Imaging
In addition to the size and pathology of the ventricles, MRI will help in identifying site of obstruction of CSF pathways, tumors in the ventricles as well as surrounding the pathways, CSF flow dynamics using phase contrast MRI.
In T1-weighted images, CSF will appear darker and in T2-weighted images CSF will appear as bright structures.
The phase contrast MRI generates signal contrast between flowing and stationary nuclei by sensitizing the phase of
transverse magnetization to the velocity of motion.17 Axial and sagittal images are obtained for CSF flow analysis.
The gray scale intensity of each pixel is directly related to the velocity of CSF. Caudal flow of CSF is conventionally
represented as shades of white on phase images, whereas cranial flow is by shades of black. Phase contrast MRI
is used to differentiate communicating and noncommunicating hydrocephalus. In addition, it helps in identifying
patients with normal pressure hydrocephalus who will benefit from shunt surgery. Patients with CSF flow velocity
(FV) of more than 18 mL/min have been shown to benefit from shunt surgery. Phase contrast MRI is also useful in
assessing the function of ventriculoperitoneal shunts.
SUMMARY
The CSF is an important component of the skull having multitude of functions ranging from protecting the brain, supplying nutrients, maintaining the cerebral homeostasis, and removal of waste materials. The CSF dynamics is a complex
process which is affected by various physiological, pathological mechanisms as well as by the drugs including anesthetic
agents. The CSF pathway is also a mechanism for the spread of some brain metastasis as well as infections. The major disorder affecting the CSF dynamics is the hydrocephalus which is also a commonest clinical situation in practice. Advanced
flow sequences based on MRI technique help us to study the CSF dynamics in various pathological states.
THE SPINAL CORD
INTRODUCTION
The spinal cord is a neural structure that connects the brain to the peripheral nerve system. The major function
of the spinal cord is to transmit action potentials between the brain and the peripheral structures via the peripheral
nerves. The spinal cord is enclosed in the vertebral column with intervening intervertebral discs. Similar to the brain
II. NEUROPHYSIOLOGY
84
4. NEUROPHYSIOLOGY
there are three protective meningeal coverings. Since the spinal cord is enclosed in a closed space like brain, it is
vulnerable for injury due to various diseases. This chapter enumerated the basic anatomy, physiology, and various
pathophysiological aspects of the spinal cord.
ANATOMY
The spinal cord is a part of CNS which extends from the lower end of medulla oblongata to the caudal equina
which are the tuft of spinal nerves. In adults the spinal cord ends at L1-2 vertebra and in children at L2-3 vertebra.
The difference is due to the rapid growth of the vertebral column compared to the spinal cord. The spinal cord is
divided into four segments, from cranial to caudal as cervical, thoracic, lumbar, and sacral. There are two enlargements, cervical and lumbar. There are 31 bilateral spinal nerves that consist of cervical (8), thoracic (12), lumbar (5),
sacral (5), and coccyx (1) nerves.
ORGANIZATION OF THE SPINAL CORD
The spinal cord is made up of central gray matter that consists of cell bodies of neurons and the outer white
matter that consists of fiber tracts. The spinal cord is divided incompletely into two symmetrical hemispheres
by the anterior median fissure which is much deeper than the posterior median fissure with the intervening
undivided central gray matter. On either side of the spinal cord, the anterior lateral and posterior lateral fissures
are present which represent the area where the ventral and dorsal rootlets (later roots) emerge from the cord to
form the spinal nerves. Each spinal nerve has two roots, a dorsal and ventral. The dorsal root emerges from the
dorsal gray horn, dorsolateral funiculus and enters a dorsal root ganglion located in the intervertebral foramen.
Outside the foramen it joins the ventral root that emerges from the ventral gray horn and ventrolateral funiculus
to form the spinal nerve. The spinal nerves in the cervical and lumbar areas form plexus from where peripheral
nerve emerges.
The white matter circumferentially surrounds the gray matter. The gray matter has three horns namely, the
ventral, intermediate, and dorsal horn. The white matter is conventionally divided into the dorsal, dorsolateral,
lateral, ventral, and ventrolateral funiculi. The spinal cord consists of neurons and nonneuronal cells. The neurons of the gray matter have specific laminar architectural pattern which was described by Rexed54 and Scheibel
and Scheibel.55
White Matter of Spinal Cord
White matter of the spinal cord has various interconnecting fibers. The connections of spinal cord can be classified
as intrinsic pathways, ascending pathways, and descending pathways (Fig. 4.17).
Intrinsic Pathways
Intrinsic pathways establish connections between spinal interneurons and various ascending and descending
fibers and also between different segments of the spinal cord. They may be important for proprioception and reflex
pathways. Examples of intrinsic pathways include Lissauer’s tract, comma tract, septomarginal tract, cornucommissural tract, etc.
Ascending Pathways
Table 4.4A shows various ascending pathways in the spinal cord. The ascending pathways consist of axons of the
dorsal root ganglion that enters the spinal cord via dorsal root.
Descending Pathways
Table 4.4B shows various descending pathways. The major descending pathway of the spinal cord is the
corticospinal tract that originates in the motor cortex. Majority of the fibers cross in the medulla and descends
as lateral corticospinal tract. Few uncrossed fibers descend as ventral corticospinal tract and cross at the level
of spinal cord.
II. NEUROPHYSIOLOGY
85
ORGANIZATION OF THE SPINAL CORD
ASCENDING TRACTS
DESCENDING TRACTS
Fasciculus Gracilis
septomarginal
Comma
Fasciculus Cuneatus
Dorsal spinocerebellar
Lateral corticospinal
Rubrospinal
Lateral Spinothalamic
Olivospinal
Ventral spinocerebellar
Ventral Spinothalamic
Lateral reticulospinal
Vestibulospinal
Spinotectal
Medial reticulospinal
Tectospinal
Ventral corticospinal
Spino olivary
FIGURE 4.17 The various ascending and descending pathways of spinal cord.
TABLE 4.4 The Important Ascending and Descending Pathways for Spinal Cord
A. Ascending Pathways
Funiculus
Tracts
Destination
Function
Posterior
Fasciculus gracilis
Ventral posterolateral nucleus of thalamus
Touch, proprioception from lower
limbs, lumbar, sacral, coccygeal, and
lower thoracic
t F
asciculus cuneatus
Ventral posterolateral nucleus of thalamus
Touch, proprioception from upper
limbs, upper thorax, neck
Posterior spinocerebellar Anterior
spinocerebellar
Cerebellum
Unconscious proprioception (posture
control)
Lateral spinothalamic
Ventral posterolateral nucleus of thalamus
Pain and temperature
t S
pinotectal
Superior colliculus
Connection between eye and body
parts
t S
pinoreticular
Reticular formation of medulla and pons
Lateral
t D
orsolateral (Lissauer’s tract)
Continued
II. NEUROPHYSIOLOGY
86
4. NEUROPHYSIOLOGY
TABLE 4.4 The Important Ascending and Descending Pathways for Spinal Cord—Cont'd
A. Ascending Pathways
Funiculus
Tracts
Destination
Function
Anterior
Anterior spinothalamic
Ventral posterolateral nucleus of thalamus
Crude touch
Spino olivary
Superior olivary nucleus
t S
pinotectal
Superior colliculus
t S
pinoreticular
Reticular formation of medulla and pons
Connection between eye and body parts
B. Descending Pathways
Funiculus
Tracts
Origin
Posterior
Fasciculus interfascicularis
(semilunar tract/comma tract of
Schultz)
To cervical and thoracic cord Located
between fasciculus gracilis and cuneatus
Function
Septomarginal tract
Lateral
Anterior
Lateral corticospinal
Motor area
Muscle innervation
Rubrospinal
Red nucleus
Facilitates flexors, inhibitory to
extensors
Lateral reticulospinal
Reticular system from medulla
Facilitates flexors, breathing control
Olivospinal
Inferior olivary nucleus to cervical cord
Control of neck in equilibrium
Descending autonomic bres
Hypothalamus
Anterior corticospinal
Motor area
Muscle innervation
Lateral vestibulospinal
Lateral vestibular nucleus
Equilibrium
Medial vestibulospinal
Medial vestibular nucleus
Equilibrium
Tectospinal
Superior colliculus
Connection between eye and body
parts
Medial reticulospinal
Reticular system from medulla
Facilitates extensors, posture control
Spinal Cord Interneurons
Spinal interneurons are distributed widely and they are responsible for the fine control of the spinal cord function.
They receive sensory inputs from various areas (both peripheral and central), and the final output depends on the type
of inputs by which the functions are modified. The interneurons express various neurotransmitters such as glycine.
Glial Cells of Spinal Cord
In addition to the neurons, glial cells are also present in the spinal cord. The cell types are similar to those in brain:
astrocyte, oligodendrocyte, ependyma, and microglia. Microglia are derived from monocytes whereas the other
glial cells are derived from neuroectodermal cells. Astrocytes during development helps in migration of neurons,
whereas in mature stage they contribute to the structural support of other cells. Astrocyte foot process contributes
to the formation of blood spinal cord barrier formation. Oligodendrocytes produce myelin which cover the multiple
axons within CNS, whereas Schwann cells contribute myelin to peripheral nerves. They are the targets in diseases
that cause demyelination. Microglia are immune cells of the spinal cord. They are dendritic antigen-presenting cells
expressing major histocompatibility extraspinal antigens.
Blood Supply
The blood supply of the spinal cord is unique in that it receives blood supply from multiple extraspinal arteries.
Despite these abundant interconnections, the spinal cord is vulnerable to ischemia due to the variability of the blood
supply. The blood supply can be classified as intrinsic system and extrinsic system.
II. NEUROPHYSIOLOGY
87
ORGANIZATION OF THE SPINAL CORD
Posterior Spinal Artery
Dorsal Nerve roots
Ventral Nerve Roots
Spinal Nerve
Central artery
Radiculo medullary
Artery
Anterior
Spinal Artery
Artery of Ademkiewicz
FIGURE 4.18 The diagrammatic representation of spinal cord blood supply.
Intrinsic Supply
The intrinsic system of blood supply to the spinal cord is by single anterior spinal artery (ASA) and two posterior
spinal arteries (PSAs), which are located on the surface of the cord and the pial anastomotic system (Fig. 4.18). The
intrinsic system has been classified to be a central and a peripheral system based on the supply.56 The central system
predominantly supplies two-thirds of the spinal cord and is derived from the ASA. The blood flow is centrifugal.
The central system supplies the anterior gray matter, anterior portion of the posterior gray matter and posterior
white columns, inner half of the anterior and lateral white columns, and base of the posterior white columns. In the
peripheral system, the blood flows centripetally from the PSAs and pial arterial plexus. This system supplies the
outer portion of the anterior and lateral white columns and the posterior portion of the posterior gray matter and
posterior white columns.56
The ASA arises from the branches of the vertebral arteries that are intracranially proximal to basilar artery. The
diameter of the ASA is larger in the cervical region, and it decreases as it descends down the cord. At the lower end of
the sacral or coccygeal region, branches from the ASA loop caudally around the conus medullaris and join each limb
of the PSA forming anastomoses. The ASA receives segmental arteries along the length of the cord.
The PSA arises from the vertebral or posterior inferior cerebellar arteries, winds around the brain stem, and
descends on the dorsomedial surface of the spinal cord. The PSA ends at the conus medullaris by forming anastomosis with the terminal branches of the ASA.
Throughout the surface of the spinal cord the branches of the ASA and PSA form a pial arterial plexus. Most of the
branches from the pial arterial plexus penetrate the dorsal midline of the spinal cord. All of the penetrating branches
run directly inward, perpendicular to the surface of the spinal cord. These branches mostly supply the outer portions
of the spinal cord, including the greater part of the posterior horns up to the substantia gelatinosa.
The central arteries, which are the branches of the ASA, supply the central portion of the spinal cord. The central
arteries feed the central area of the spinal cord, which consists of the white matter bordering the central sulcus, the
gray matter of anterior horn, and the deep gray matter of posterior horn.57
Extrinsic Blood Supply
The extrinsic supply refers to the blood supply of the spinal cord which feeds the ASA and PSA. The radicular
arteries which are usually 31 pairs are the main arteries that feed the ASA and PSA. Radicular arteries supply blood
to the dura mater, nerve roots that they accompany, dorsal spinal ganglia, and ASA and PSA.57 The radicular arteries
arise from anterior cervical arteries, thoracic aorta, and lumbar arteries. The artery of Ademkiewicz is the largest of
II. NEUROPHYSIOLOGY
88
4. NEUROPHYSIOLOGY
the radicular artery. It usually joins the ASA. In 75% people, this artery travels with the T9–12 roots. In 10% it follows
roots L-1 or L-2, and in 15% it has a high origin at posterior roots T5–8. Ligation of artery of Ademkiewicz in animals
results in paraplegia indicating that this is the major source of blood to lower parts of spinal cord.57
Despite abundant blood supply, watershed regions are common in spinal cord especially between the areas where
the radicular arteries join the ASA. The blood flow at the watershed area is both upward and downward making the
spinal cord vulnerable to ischemia. The mid thoracic area where the distance between radicular arteries is the greatest has the maximum watershed effect.
Venous System of Spinal Cord
The veins of the spinal cord have extrinsic and intrinsic veins. The intrinsic veins (sulcal and radial veins) drain the
substance of the cord to the extrinsic system. The extrinsic system consists of anterior spinal vein which accompanies
the ASA till the filum terminale and continues as filum terminale vein, the greatest spinal vein which traverses in the
posterior median sulcus along with two posterolateral veins on either sides.
The spinal veins drain into the anterior and posterior radiculomedullary veins, which in turn drain into the paravertebral and intervertebral plexuses. Finally, the venous plexuses drain into the segmental veins, draining into the
ascending lumbar veins, azygos system, and pelvic venous plexuses.
Pathophysiology
Various disease processes such as tumors, atherosclerosis, trauma, vascular malformations, aortic surgeries, etc.
can affect the vascular supply and can cause cord ischemia. It is important to recognize the specific pattern of vascular involvement. Two types of infarct can occur:
1. Anterior spinal artery syndrome: It is caused by reduced supply of the ASA. The clinical manifestations include,
bilateral loss of motor function and pain/temperature sensation, with relative sparing of proprioception and
vibratory senses below the level of the lesion. Autonomic disturbance also can occur.
2. Posterior spinal artery syndrome: It is caused by reduced blood supply in the territory of posterior spinal artery.
It is characterized by sensory symptoms such as loss of proprioception and vibratory senses below the level of
the injury and total anesthesia at the level of the injury. Motor weakness is usually mild.
3. Other infarcts include incomplete infarcts, central cord infarct (bilateral spinothalamic lesion with sparing of
motor and posterior columns), transverse medullary infarct (paraplegia or tetraplegia with loss of sensations).
Autoregulation and CO2 Reactivity of the Spinal Cord
Autoregulation of the spinal cord blood flow is similar to that seen in the brain. The studies on the autoregulation
are derived from animal experiments. Spinal cord receives 40–60% of the CBF (50–55 mL/100 g/min). The tissue
oxygen levels are the same (35–39 mmHg).58 The blood flow was found to be constant over a MAP of 40–100 mmHg
in animal studies.59 Hitchon et al. have found wide variation in the blood flow between gray matter and white matter (approximate ratio of 5:1) and also differences in the blood flow between cervical and the lumbar levels.60 There
was a drop in spinal cord blood flow below MAP of 50 mmHg in their study. In another animal study, an MAP of
>135 mmHg was associated with increased blood flow and risk of disruption of the blood spinal cord barrier and
edema and neuronal damage.61 The mechanism of the spinal cord autoregulation has not been fully understood.
Experimental evidence suggests that blockade of dorsal sympathetic ganglion distal to the medulla has been found
to abolish the autoregulation. The sensors for the autoregulation have been thought to be in the glomeruli of the
capillaries in and around anterior and posterior horns in the spinal cord.
The CO2 reactivity of the spinal cord also appears to be similar to brain with vasodilation in response to hypercapnia and vice versa.62 The spinal cord blood flow was found to be constant between 10 and 50 mmHg of CO2.62 Beyond
50 mmHg there was an increase in the spinal cord blood flow.
Functions of the Spinal Cord
The spinal cord is a complex structure that plays a major role in three important functions. They are sensory control, motor, and autonomic functions. The spinal cord is not a simple relay system of the signals to and fro between
brain and the organ system via peripheral nerves as thought to be. In spinal cord there is a complicated network
II. NEUROPHYSIOLOGY
REFERENCES
89
of neurons that normally operates in conjunction with the rest of the CNS to allow perfect control of sensory, autonomic, and motor functions.
Sensory Function
Different sensations arising from various areas are transmitted via the peripheral nerves which are the axons
whose cell body is located in the dorsal root ganglion. The dorsal root ganglion represents the first-order neurons
that project into the spinal cord. Table 4.4 shows the various spinal processing of different sensory modalities.
Motor Function
The spinal cord is involved in two major motor activities by itself without the concurrent activity of the brain.
1. Monosynaptic stretch reflex: Stimuli originating from the muscle spindles cause contraction of the muscle in
which the stimuli originated via 1A afferent fibers.
2. Patterned movements: The spinal cord can generate patterned responses that involve movement of several
joints. Examples of reflex of this type are the flexor or withdrawal reflex in response to various sensory stimuli,
and in particular in response to pain.
Autonomic Function
Spinal cord function is important for the autonomic control of the viscera such as bladder, GI tract, and sexual
function. The sympathetic and parasympathetic neurons are located in the spinal cord. The preganglionic neurons
of the sympathetic system are localized in the thoracic and lumbar part of the spinal cord, while neurons that control
the parasympathetic ganglia originate in the sacral region. Spinal cord damage can result in dysfunction of the autonomic functions such as bladder dysfunction, defecation, and sexual dysfunction.
SUMMARY
The spinal cord is a part of CNS connecting between the brain and the peripheral nervous system and subserving
the integration of various functions. The spinal cord is vulnerable to various injuries and can lead to long-lasting
morbidity. It is important to know the anatomy and physiology to aid in the monitoring, management of patients
with spinal cord problems. Newer therapies such as stem cell implantation, immunomodulatory therapy, neuroplasticity, electrostimulation are emerging in the management of these patients.
References
1. Burrows G. On disorders of the cerebral circulation and on the connection between affections of the brain and diseases of the heart. London: Longman, Brown, Green and
Longman; 1846. p. 55–6.
2. Cushing H. The third circulation in studies in intracranial physiology and surgery. London (UK): Oxford University Press; 1926.
3. Malm J, Kristrensen B, Karlsson T, Fagerlund M, Elfverson J, Ekstead J. The predictive value of cerebro spinal fluid dynamic tests in patients with idiopathic adult
hydrocephalus syndrome. Arch Neurol 1995;52:783–9.
4. Sorrentino E, Diedler J, Kasprowicz M, et al. Critical thresholds for cerebrovascular reactivity after traumatic brain injury. Neurocrit Care 2012;16:258–66.
5. Steiner LA, Balestreri M, Johnston AJ, et al. Predicting the response of intracranial pressure to moderate hyperventilation. Acta Neurochir (Wien) 2005;147:477–83.
6. Dunn LT. Raised intracranial pressure. J Neurol Neurosurg Psychiatry 2002;73(Suppl. I):i23–7.
7. Barlow A, Stewart W. The pathology of raised intracranial pressure. ACNR 2007;7:25–7.
8. Hamer J, Alberti E, Hoyer S, Wiedmann K. Influence of systemic and cerebral vascular factors on the cerebro spinal fluid pulse waves. J Neurosurg 1977;46:36–45.
9. Lofgren J, von Essen C, Zwetnow NN. The pressure-volume curve of the cerebrospinal fluid space in dogs. Acta Neurol Scand 1973;49:557–74.
10. Avezaat CJ, van Eijndhoven JH, Wyper DJ. Cerebrospinal fluid pulse pressure and intracranial volume-pressure relationships. J Neurol Neurosurg Psychiatry
1979;42:687–700.
11. Muizelaar JP, Ward JD, Marmarou A, et al. Cerebral blood flow and metabolism in severely head-injured children. Part 2: Autoregulation. Neurosurg 1989;71:72–6.
12. Abraham M, Singhal V. Intracranial pressure monitoring. J Neuroanesthesiol Crit Care 2015;2:193–203.
13. Sakka L, Coll G, Chazal J. Anatomy and physiology of cerebrospinal fluid. Eur Ann Otorhinolaryngol Head Neck Dis 2011;128:309–16.
14. Pollay M. The function and structure of the cerebrospinal fluid system. Cereb Fluid Res 2010;7:9.
15. Brierley JB, Field EJ, Yoffey JM. Passage of Indian ink particles from the cranial subarachnoid space. J Anat 1949;83:77.
16. Johnson RN, Maffeo CJ, Dacey RG, et al. Mechanism for intracranial hypertension during experimental subarachnoid hemorrhage: Acute malfunction of arachnoid villi by components of plasma. Trans Am Neurol Assoc 1978;103:138–42.
17. Battal B, Kocaoglu M, Bulakbasi N, Husman G, Sanal HT, Tayfun C. Cerebrospinal fluid flow imaging by using phase-contrast MR technique. Br J Radiol
2011;84:758–65.
18. Peterson EC, Wang Z, Britz G. Regulation of cerebral blood flow. Int J Vasc Med Vol 2011:823525. http://dx.doi.org/10.1155/2011/823525. 8 pages.
19. Willie KC, Tzeng YC, Fisher JA, Ainslie PN. Integrative regulation of human brain blood flow. J Physiol 2014;592:841–59.
20. Drummond JC. The lower limit of autoregulation: time to revise our thinking? Anesthesiology 1997;86:1431–3.
21. Birch AA, Dirnhuber MJ, Hartley-Davies R, Iannotti F, Neil-Dwyer G. Assessment of autoregulation by means of periodic changes in blood pressure. Stroke
1995;26:834–7.
II. NEUROPHYSIOLOGY
90
4. NEUROPHYSIOLOGY
22. Chan GSH, Ainslie PN, Willie CK, Taylor CE, Atkinson G, Jones H, Lovell NH, Tzeng YC. Contribution of arterial Windkessel in low-frequency cerebral hemodynamics during transient changes in blood pressure. J Appl Physiol 2011;110:917–25.
23. Heistad DD, Marcus ML, Abboud FM. Role of large arteries in regulation of cerebral blood flow in dogs. J Clin Invest 1978;62:761–8.
24. Bayliss WM. On the local reactions of the arterial wall to changes of internal pressure. J Physiol 1902;28:220–31.
25. Gokina NI, Park KM, McElroy-Yaggy K, Osol G. Effects of Rho kinase inhibition on cerebral artery myogenic tone and reactivity. J Appl Physiol 2005;98:1940–8.
26. Cauli B, Tong XK, Rancillac A, et al. Cortical GABA interneurons in neurovascular coupling: relays for subcortical vasoactive pathways. J Neurosci 2004;24:8940–9.
27. Roy CS, Sherrington CS. On the regulation of the blood-supply of the brain. J Physiol 1890;11:85–158.
28. Nielsen AN, Lauritzen M. Coupling and uncoupling of activity-dependent increases of neuronal activity and blood flow in rat somatosensory cortex. J Physiol
2001;533:773–85.
29. Iadecola C. Does nitric oxide mediate the increases in cerebral blood flow elicited by hypercapnia? Proc Natl Acad Sci U.S.A. 1992;89:3913–6.
30. Hoehn K, White TD. Role of excitatory amino acid receptors in K+- and glutamate-evoked release of endogenous adenosine from rat cortical slices. J Neurochem
1990;54:256–65.
31. Battisti-Charbonney A, Fisher J, Duffin J. The cerebrovascular response to carbon dioxide in humans. J Physiol 2011;589:3039–48.
32. Meng L, Gelb AW. Regulation of cerebral autoregulation by carbon Dioxide. Anesthesiology 2015;122:196–205.
33. Häggendal E, Johansson B. Effects of arterial carbon dioxide tension and oxygen saturation on cerebral blood flow autoregulation in dogs. Acta Physiol Scand Suppl
1965;258:27–53.
34. Ogoh S, Sato K, Nakahara H, Okazaki K, Subudhi AW, Miyamoto T. Effect of acute hypoxia on blood flow in vertebral and internal carotid arteries. Exp Physiol
2013;98:692–8.
35. Mardimae A, Balaban DY, Machina MA, Battisti-Charbonney A, Han JS, Katznelson R, Minkovich LL, Fedorko L, Murphy PM, Wasowicz M, Naughton F, Meineri
M, Fisher JA, Duffin J. The interaction of carbon dioxide and hypoxia in the control of cerebral blood flow. Pflugers Arch 2012;464:345–51.
36. Ogoh S, Ainslie PN. Regulation of cerebral blood flow in mammals during chronic hypoxia: a matter of balance. Exp Physiol 2009;95:251–62.
37. Massik J, Tang YL, Hudak ML, Koehler RC, Traystman RJ, Jones Jr MD. Effect of hematocrit on cerebral blood flow with induced polycythemia. J Appl Physiol
1987;62:1090–6.
38. Crystal GJ, Salem MR. Myocardial and systemic hemodynamics during isovolemic hemodilution alone and combined with nitroprusside induced controlled
hypotension. Anesth Analg 1991;72:227–37.
39. Crystal GJ, Czinn EA, Salem MR. The mechanism of increased blood flow in the brain and spinal cord during hemodilution. Anesth Analg 2014;118:637–43.
40. Le Roux P. Haemoglobin management in acute brain injury. Curr Opin Crit Care 2013;19:83–91.
41. Krejza J, Mariak Z. Effect of age on cerebral blood flow velocity in patients after aneurysmal subarachnoid hemorrhage. Stroke 2002;33:640–2.
42. Ehrlich MP, McCullough JN, Zhang N, Weisz DJ, Juvonen T, Bodian CA, Griepp RB. Effect of hypothermia on cerebral blood flow and metabolism in the pig. Ann
Thorac Surg 2002;73:191–7.
43. Nybo L, Møller K, Volianitis S, Nielsen B, Secher NH. Effects of hyperthermia on cerebral blood flow and metabolism during prolonged exercise in humans. J Appl
Physiol 1985;93:58–64.
44. Sethuraman M. Cerebral blood flow monitoring. J Neuroanaesthesiol Crit Care 2015;2:204–14.
45. Magistretti PJ, Pellerin L. Cellular mechanisms of brain energy metabolism and their relevance to functional brain imaging. Phil Trans R Soc Lond B 1999;354:1155–63.
46. Larrabee MG. Lactate metabolism and its effects on glucose metabolism in an excised neural tissue. J Neurochem 1995;64:1734–41.
47. Gjedde A, Marrett S, Vafee M. Oxidative and non oxidative metabolism of excited neurons and astrocytes. J Cereb Blood Flow Metab 2002;22:1–14.
48. Boumezbeur F, Peterson KF, Cline GW, Mason GF, Behar KL, Shulman GL, Rothman DL. The contribution of blood lactate to brain energy metabolism in humans
measured by dynamic 13C nuclear magnetic resonance spectroscopy. J Neurosci 2010;30:13983–91.
49. Magistretti PJ. Neuron–glia metabolic coupling and plasticity. J Exp Biol 2006;209:2304–11.
50. Belanger M, Allaman I, Magistretti PJ. Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation. Cell Metab 2011;14:724–38.
51. Schell RM, Cole DJ. Cerebral monitoring: jugular venous oximetry. Anesth Analg 2000;90:559–66.
52. Murkin JM, Arango M. Near-infrared spectroscopy as an index of brain and tissue oxygenation. Br J Anaesth 2009;103(Suppl.1):i3–13.
53. Tisdall MM, Smith M. Cerebral microdialysis: research tool or clinical tool. Br J Anaesth 2006;97:18–25.
54. Rexed B. A cytoarchitectonic atlas of the spinal cord in the cat. J Comp Neurol 1954;100:297–379.
55. Scheibel ME, Scheibel AB. Spinal motor neurons, interneurons and Renshaw cells. A Golgi study. Arch Ital Biol 1966b;104:328–53.
56. Tveten L. Spinal cord vascularity. III. The spinal cord arteries in man. Acta Radiol Diagn (Stockh) 1976;17:257–73.
57. Lazorthes G, Gouaze A, Zadeh JO, Santini JJ, Lazorthes Y, Burdin P. Arterial vascularization of the spinal cord: recent studies of anastomotic substitution pathways. J Neurosurg 1971;35:253–62.
58. Ducker TB, Perot Jr PL. Spinal cord oxygen and blood flow in trauma. Surg Forum 1971;22:413–5.
59. Lobosky JM, Hitchon PW, Torner JC, Yamada T. Spinal cord autoregulation in the sheep. Curr Surg 1984;41:264–7.
60. Hitchon PW, Lobosky JM, Yamada T, Johnson G, Girton RA. Effect of hemorrhagic shock upon spinal cord blood flow and evoked potentials. Neurosurgery
1987;21:849–57.
61. Kobrine AI, Doyle TF, Martins AN. Local spinal cord blood flow in experimental traumatic myelopathy. J Neurosurg 1975;42:144–9.
62. Kobrine AI, Doyle TF, Martins AN. Auto regulation of spinal cord blood flow. Clin Neurosurg 1975;22:573–81.
II. NEUROPHYSIOLOGY
C H A P T E R
5
Brain Protection in Neurosurgery
H. El Beheiry1,2
1University
of Toronto, Toronto, ON, Canada; 2Trillium Health Partners, Toronto, ON, Canada
O U T L I N E
Introduction
91
Target Hemoglobin Concentration
95
Nonpharmacological Strategies
91
Pharmacological Strategies
96
Mild Hypothermia
92
Nonanesthetic Agents
97
Blood Pressure Control
93
Anesthetic Agents
97
Induced Arterial Hypertension
94
Conclusion
98
Normoglycemia
94
References
98
INTRODUCTION
Neuroprotection describes strategies to protect neuronal elements against damage and impairment of neurologic
function. One of the essentials of neuroanesthesia practice is to provide the patient with neuroprotective measures.
It is hoped that these measures will reduce poor neurologic outcomes, i.e., motor and sensory deficits and cognitive
dysfunction resulting from inevitable surgical brain injury during neurosurgical procedures. The most common
forms of brain injury during neurosurgical procedures are (1) brain retraction, (2) incising and removing brain tissue,
and (3) temporary vascular occlusion.1 For instance, eliminating pathological brain tissue and brain retraction will
inevitably lead to injury of normal brain structures. Moreover, clamping of a carotid artery during carotid endarterectomy or temporary clipping of intracerebral arteries can simulate unilateral global ischemia or acute ischemic
stroke, respectively (Table 5.1).
Neuroprotective strategies can be classified into nonpharmacological and pharmacological (Table 5.2). Some
of these strategies are based on laboratory evidence and are either target specific or with indeterminate targets.2,3
Other neuroprotective approaches are “empiric” meaning that they are guided by experience not precepts or
theory.
NONPHARMACOLOGICAL STRATEGIES
Nonpharmacological strategies signify the manipulation of homeostatic processes in a manner that will have
neuroprotective effects (Table 5.2).
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00005-1
91
© 2017 Elsevier Inc. All rights reserved.
92
5. BRAIN PROTECTION IN NEUROSURGERY
TABLE 5.1 Surgical Brain Injury During Neurosurgical Procedures
Forms of Surgical Brain Injury
Effects
Surgical brain incisions
Neuronal death, brain edema, disruption BBBa
Application of thermal and ultrasonic energy
Neuronal death, disruption BBB
Retraction of brain tissue with brain retractors
Decreased CBFb, rebound brain edema
Temporary or permanent vascular occlusion
Cessation of CBF or decreased CBF
Local surgical bleeding and brain contusion
Brain edema, disruption BBB
Major air embolism or surgical bleeding
Global brain hypoperfusion
aBBB
bCBF
indicates blood–brain barrier.
indicates cerebral blood flow.
TABLE 5.2 Strategies for Neuroprotection During Neurosurgical Procedures
Nonpharmacological Strategies
Pharmacological Strategies
Anesthesia related
Agents with specific site of action
Hypothermia
Antiexcitotoxicity
Normoglycemia
Ca2+ channel blockers
Maintenance of adequate CBFa
(Normotension, induced hypertension)
Antioxidants
Target hemoglobin concentration
Antiinflammatory
Respiratory gases manipulation
(PaO2 optimization, PaCO2 control)
Antiapoptosis
Osmotherapy
Agents with nonspecific site of action
Surgery related
Cell membrane stabilizers
Decrease brain tissue injury
(Micro- and image-guided neurosurgery)
Erythropoietin
CSFb drainage
Antithrombotics and thrombolytics
Limit ischemic time
Anesthetics
Embolic load reduction
Inert gases
aCBF
bCSF
indicates cerebral blood flow.
indicates cerebrospinal fluid.
MILD HYPOTHERMIA
Hypothermia has been commonly classified into three levels: mild from 32 to 35°C, moderate from 32 to 28°C,
and deep under 28°C. Deep hypothermia associated with circulatory arrest was previously used during clipping
of giant complex intracerebral aneurysms without favorable but detrimental outcomes.4,5 Such disappointing
experience has steered the evolution of mild hypothermia as a neuroprotective strategy based on encouraging
results shown in many laboratory investigations.6,7 The mechanisms of the presumed hypothermic neuroprotection are multifaceted and include changes in various cellular processes including its ability to decrease the
cerebral metabolic rate by about 10% for every degree Celsius.8–10 Hypothermia maintains the integrity of the
blood–brain barrier after ischemic insults and constricts cerebral blood vessels and thus reduces brain edema
and cerebral blood volume and decreases intracranial pressure (ICP). Additionally, it inhibits excitotoxicity by
decreasing glutamate release resulting in decrease of cellular depolarization and inhibition of deleterious calcium influx through voltage- and receptor-operated calcium channels. Also, hypothermia depresses the delayed
responses to brain injury, namely reactive oxygen species production and mitochondrial dysfunction that
II. NEUROPHYSIOLOGY
BLOOD PRESSURE CONTROL
93
triggers neuronal tissue inflammation and programmed cell death (apoptosis), respectively. Finally, hypothermia can ameliorate secondary neuronal damage by downregulating certain gene-induced proteomic responses
leading to cell damage and upregulating a small subset of cold-shock proteins that depress apoptosis and promote cell proliferation.
Despite the various neuroprotective mechanisms of mild hypothermia reported in the laboratory, its clinical efficacy is still indefinable. A recent carefully conducted metaanalysis11 showed that among patients undergoing craniotomy for various neurosurgical indications including aneurysm clipping,12,13 traumatic brain injury,14 and ischemic
stroke15 there was no evidence that intraoperative or postoperative hypothermia significantly reduces or increases
mortality or significantly alters the risk of severe neurologic disability. Application of mild hypothermia did not alter
the risk for postoperative complications, i.e., intracranial hemorrhage, ischemic stroke, congestive cardiac failure,
or myocardial infarction. There was some weak evidence that postoperative hypothermia may increase the risk of
infective complications. Such lack of efficacy and relative safety of mild hypothermia has been in agreement with
other systematic reviews.16,17 It should be noted that mild hypothermia might be beneficial in the case of comatose
survivors of out-of-hospital cardiac arrest18,19 and peripartum asphyxia-induced brain injury.20,21
To date, there is no convincing clinical evidence to establish the value of mild hypothermia as a neuroprotective
strategy during neurosurgery. Anesthesiologists who opt to use mild hypothermia for their patients because of its
favorable safety profile and efficacy in nonneurosurgical situations of global brain ischemia should consider the
following precautions: core temperature should be monitored at two sites to avoid inadvertent excessive cooling,
target temperature has to be reached before opening the dura, rewarming should start after brain tissue handling
has ended, rewarming should continue in the postoperative period until core temperature normalizes, and active
cooling equipment should be calibrated and tested before use.
BLOOD PRESSURE CONTROL
During neurosurgical procedures, regional and global cerebral blood flow (CBF) is compromised mainly due
to brain retraction and surgical bleeding, respectively (Table 5.2). Regional CBF was shown to decrease during
brain retraction in swine animal models by about 50% of baseline during normoventilation or hyperventilation.22 Global CBF decrements are not uncommon during neurosurgery and can lead to surgical brain injury and
unfavorable outcomes.23–25 CBF is autoregulated, i.e., sustained within a normal range (50–60 mL per 100 g brain
tissue per minute) provided that the mean arterial pressure (MAP) is between 60 and 150 mmHg and the ICP is
about 10 mmHg. CBF is correlated to the cerebral perfusion pressure (CPP) and CPP = MAP-ICP (normal CPP
is 70–90 mmHg). During neurosurgery, the autoregulatory mechanisms for CBF maintenance are not optimal
and the extent of its dysfunction is unknown. In fact, the relationship between CBF and CPP may become linear
due to loss of vessel reactivity that is responsible for autoregulation. Therefore, decrements in MAP should be
avoided and kept above 80 mmHg to maintain CPP near 70 mmHg during neurosurgery. Recent reports provide
important indirect evidence of the deleterious effects of intraoperative regional and global brain hypoperfusion.
In patients with severe traumatic brain injury, who had neurosurgical intervention in the form of decompressive
craniotomies, the autoregulatory curves were determined.26 Data from these patients revealed constant brain
perfusion over a wide CPP range (50–90 mmHg) and a 100% incidence of ischemia when CPPs fell below the
lower limit of autoregulation. As CPPs increased there was a corresponding decrease in the incidence of ischemia, potentially mediated through an associated force-dependent dilation of the cerebral vessels. In another
population of patients who undergo endovascular treatment of acute ischemic stroke, a relatively consistent
conclusion from studies is that general anesthesia appears to be associated with higher mortality and morbidity
compared with monitored anesthesia care.27,28 This has been attributed to the hypotensive actions of general
anesthesia resulting from anesthetic agents and positive pressure ventilation, and such depressive cardiovascular effects will lead to decrements in brain perfusion. Additionally, data from the International Stroke Trial
suggest that for every 10 mmHg below asystolic blood pressure of 150 mmHg early death increased by about
18%.29 Finally, acute reductions in MAP in patients with intracerebral hemorrhage caused diminishing cerebral
tissue diffusion on MRI and were associated with cerebral ischemia, disability, and mortality.30
In conclusion, during neurosurgical procedures moderate and severe intraoperative hypotension should be
avoided and MAP should be maintained close to the patient’s baseline pressure. Elevation of MAP could be achieved
by using a combination of measures including volume resuscitation, decreasing anesthetic levels and vasopressors,
i.e., alpha agonists and ephedrine.
II. NEUROPHYSIOLOGY
94
5. BRAIN PROTECTION IN NEUROSURGERY
INDUCED ARTERIAL HYPERTENSION
Induced (or permissive) hypertension is a technique that can achieve stable and adequate collateral CPP
during neurosurgical procedures associated with localized ischemic compromise of brain tissue. Such technique uses vasopressors to raise the arterial blood pressure by 20–40% to recruit cerebral collateral networks
including the leptomeningeal circulation particularly in patients who have incomplete circle of Willis.31 Hence,
induced hypertension will be potentially beneficial to reduce the incidence of perioperative cerebral ischemic
events during: (1) interventional neuroradiology procedures, (2) temporary clipping or clamping during cerebral aneurysmal surgery or carotid endarterectomy, respectively, (3) extracranial to intracranial bypass surgery,
(4) surgery in patients with cerebral vasospasm and (5) surgery in patients with conditions leading to significant
cerebral autoregulation dysfunction, e.g., intracranial pathology with mass effect, severe systemic hypertensive
disease and traumatic brain injury.32
Several reports allude to the potential benefits of induced hypertension. Actually, increased middle cerebral
artery mean blood flow velocity by intentional hypertension during dissection of the carotid artery in carotid endarterectomy prevented the postoperative development of new cerebral ischemic lesions as detected by diffusion
weighted MRI imaging.33 In another recent study, reactionary approach to malperfusion, i.e., selective shunting
or elevation of blood pressure was compared to a preemptive routine protocol for induced hypertension during
carotid clamping to maintain adequate collateral CBF during carotid endarterectomy. The risk of temporary neurologic malperfusion was 18.1% in the groups where a reactionary approach to malperfusion was addressed by
shunt, or the elevation of blood pressure, as compared with 0.86% of patients with pretreated collateral CPP with a
standard induced hypertension protocol.34 Additionally, the safety of induced hypertension has been described in
several reports. For example, induced hypertension did not cause rupture in small, intact, unprotected intracranial
aneurysms in subarachnoid hemorrhage patients.35 Similarly, there were no reported events of myocardial infarction, congestive heart failure, intracranial hemorrhage, or hyperperfusion syndrome.33–35
Phenylephrine (50–200 µg IV boluses) and ephedrine (5–10 mg IV boluses) are the primary vasoconstrictors used
to raise blood pressure during induced hypertension. Phenylephrine is a pure α1-adrenergic receptor agonist and
causes reflex bradycardia. It does not have chronotropic or inotropic effect. It has an immediate onset of action and
a short half-life of approximately 5 min that makes it suitable for use as a variable rate infusion. If severe bradycardia occurs, anticholinergic agents can be administered to antagonize the baroreceptor-mediated vagotonic effect of
phenylephrine. Ephedrine is a sympathomimetic amine. It indirectly stimulates the adrenergic receptor system by
increasing the activity of norepinephrine at the postsynaptic α and β receptors thus causing vasoconstriction and
week chronotropic and inotropic effects. Other sympathomimetic agents have been used to induce hypertension
particularly in the ICU, e.g., dopamine, dobutamine, and vasopressin.
NORMOGLYCEMIA
Perioperative hyperglycemia may occur in diabetic as well as nondiabetic patients. In nondiabetic patients, perioperative hyperglycemia exists in two forms: stress hyperglycemia and glucose variability.36 The latter is a measure
of the magnitude of glucose excursions over time and can occur in diabetic patients. Perioperative hyperglycemia
results from neuroendocrine responses activated by surgery or trauma that includes an increase in the stress hormones such as catecholamines, cortisol, and glucagon providing conditions for tissue recuperation. Nonetheless,
the intensification of such homeostatic response mechanisms can trigger organ damage of its own accord.37 In fact,
the evidence consistently suggests that perioperative hyperglycemia leads to unfavorable neurologic and nonneurologic outcomes after various neurosurgical procedures. A recent study showed that after surgical intervention in
diabetics with cervical spondylosis myelopathy, perioperative glucose levels were linearly associated with impaired
improvement in Nurick score that is based on the extent of walking disability.38 In carotid endarterectomy, patients
with operative day glucose more than 11.1 mmol/L (200 mg/dL) were 2.8-, 4.3-, and 3.3-fold more likely to experience perioperative stroke or transient ischemic attack, myocardial infarction, or death, respectively.39 Additionally,
patients with elevated fasting blood sugar 6.1 mmol/L (>110 mg/dL) undergoing carotid artery stenting are at a
greater risk for worse major acute events, namely stroke, myocardial infarction, and death in both the short and
long term.39 The Intraoperative Hypothermia for Aneurysm Surgery Trial (IHAST) concluded that blood glucose
levels more than 7.2 and 8.4 mmol/L (130 and 150 mg/dL) at the time of clipping of a ruptured cerebral aneurysm
are associated with long-term changes in cognition and gross neurologic dysfunction, respectively.40 In the pediatric
population, patients who developed postoperative complications exhibited higher mean blood glucose levels on
II. NEUROPHYSIOLOGY
TARGET HEMOGLOBIN CONCENTRATION
95
TABLE 5.3 Suggested Intraoperative Therapy for Hyperglycemia in Neurosurgical Patients
Intraoperative Blood Sugar Control [Target: 7.8–10.0 mmol/L (140–180 mg/dL)]
1. Patients who are undergoing elective neurosurgery do not use insulin and whose blood glucose is controlled (normal finger-stick readings
and A1C values) in the outpatient setting do not require an insulin drip and can be managed with subcutaneous (SC) supplemental insulin
therapy guided by the following sliding scale:
Glucose value
Insulin dose
mmol/L
mg/dL
IU
≤8.3
≤ 150
—
8.4–11.1
151–200
5
11.2–13.9
201–250
10
13.9–16.7
251–300
15
16.7–19.4
301–350
20
2. Patients who are undergoing urgent surgery or using insulin to control their blood sugar or having uncontrolled diabetics:
Prepare insulin drip: 100 IU of regular insulin in 100 mL normal saline (1 IU/mL)
Start an empiric infusion 0.02 IU/kg/h 2–3 h prior to the surgery
Measure blood sugar after 1 h and adjust the infusion according to a sliding scale
In the operating room, continue insulin infusion
Measure blood sugar every hour
Adjust the insulin infusion according to the following sliding scale:
Glucose value
Insulin infusion rate
mmol/L
mg/dL
IU/h
8.3–9.4
150–169
2
9.4–11.0
170–199
3
11.1–13.8
200–249
4
13.9–16.6
250–299
6
16.7–22.1
300–399
8
22.2 +
400 +
10
If hypoglycemia is detected (blood sugar < 60 mg/dL or <3.3 mmol/L):
Administer hypertonic dextrose (50%) according to the following formula:
(100 − glycemia or 7.8 mmol/L) × 0.3 = dextrose 50% in mL IV bolus
Check blood glucose every 30 min
If glucose < 60 mg/dL (3.3 mmol/L), repeat the IV bolus step as necessary
“This approach will prevent over correction of hypoglycemia”
In the PACU, the insulin infusion is continued as mentioned above with hourly measurement of blood sugar
Then patient is transferred to a step-down or intensive care unit where the infusion will be transitioned to SC insulin or oral hypoglycemic
medications according to the intensivist discretion
admission to the intensive care unit 9 mmol/L (162.0 mg/dL) and mean peak blood glucose levels on postoperative
day one 9.6 mmol/L (171.9 mg/dL).41
Because the brain tissue is vulnerable at the extremes of blood sugar values, a consensus pertaining to the
blood sugar target is still controversial in neurosurgical patients. However, to date most evidence cannot support intensive insulin therapy with tight glucose control in the neurosurgical population during the perioperative period.42–44 Tight glucose control has been shown to cause much higher incidence of severe hypoglycemic
episodes, stroke, myocardial infarction, and death as well as brain energy crisis that correlates with increased
mortality.45,46 Consequently, a consensus statement by the American Association of Clinical Endocrinologists
and the American Diabetes Association47 has recommended that treatment should be initiated at a threshold of
>10.0 mmol/L (>180 mg/dL), preferably with IV insulin therapy, and maintain the glucose level between 7.8 and
10.0 mmol/L (140 and 180 mg/dL). The consensus also conveyed that (1) greater benefit may be obtained at the
lower end of this range, (2) glucose concentrations <6.0 mmol/L (110 mg/dL) are not recommended, and (3) the
suggested glucose targets should be flexible and individualized to the patient and the clinical situations relating
to the speed of achieving normoglycemia and the insulin regimen used. Several insulin regimens have been proposed. A modified guideline is suggested in Table 5.3.
TARGET HEMOGLOBIN CONCENTRATION
Anemia causes activation of compensatory physiologic cardiovascular and cellular mechanisms to optimize
tissue oxygen delivery. These include (1) increased cardiac output, (2) preferential blood flow to the brain, and
(3) induction of neuronal nitric oxide synthase that is necessary for the function of hypoxia inducible factor-1α
II. NEUROPHYSIOLOGY
96
5. BRAIN PROTECTION IN NEUROSURGERY
to stimulate the cellular responses to anemia to maintain adequate tissue oxygenation. Such cellular responses to
anemia are enhancement of angiogenesis, promotion of erythropoiesis, increase in cellular glucose, and prompting of glycolytic anaerobic metabolism.48 Despite the activation of these potent responses, tissue oxygen tension
in the brain decreases in proportion to the hemoglobin level when these mechanisms become overwhelmed and
fail. A previous report showed that packed red blood cell transfusion in subarachnoid hemorrhage patients with
initial hemoglobin of 8 g/dL resulted in brain tissue oxygen (PbtO2) improvement and a positive and independent
association between hemoglobin concentration and PbtO2 as verified by univariate and multivariate analyses.49
While there is an abundance of experimental studies showing the harmful effects of anemia on the brain, the
causation link has yet to be established in neurosurgical patients. Nonetheless, a large retrospective cohort study
including about 8000 elective neurosurgical patients provided evidence that anemia is an independent risk factor
for increased postoperative mortality and neurologic morbidity in the form of stroke or coma. In addition, a specific
level of anemia corresponding to hemoglobin below 11 g/dL was shown to be associated with increased morbidity.50 In other large retrospective studies, hemoglobin concentration <11 g/dL was an independent risk factor for
symptomatic postoperative cerebral vasospasm in spontaneous subarachnoid hemorrhage patients. Furthermore, a
hemoglobin concentration of <11 g/dL during the hospital stay was associated with unfavorable outcomes including
increased mortality, and brain infarction.51,52
Although anemia is associated with poor neurologic outcome, especially in the presence of cerebral ischemia,
there is a lack of level I evidence by which transfusion thresholds can be suggested. Furthermore, packed red cell
transfusion was not always associated with improved outcome, e.g., in traumatic brain injury and acute ischemic
stroke.53 Hence, based on the evidence currently available, it may be prudent to recommend more higher transfusion
thresholds for patients undergoing neurosurgical procedures: preoperative and intraoperative hemoglobin levels in
neurosurgical patients can be maintained at a minimum of 12 and 9 g/dL, respectively.53–55
PHARMACOLOGICAL STRATEGIES
Pharmacological strategies comprise the use of pharmaceutical agents to oppose or block a pathologic process in
the brain injury cascade of events leading to cell death by necrosis or apoptosis (Figs. 5.1 and 5.2).
Energy Failure
Depolarization
Glutamate Release
& Excitotoxicity
Opening Ca Channels
Increased [Ca2+]i
Enzyme Induction
Formation of
O2 free radicals
Membrane Damage
Inflammation
Necrosis
Mitochondrial damage
DNA damage
Apoptosis
FIGURE 5.1 A flow diagram shows the cascade that is activated after an ischemic insult to the brain. The pathways within the cascade lead to
cell death by necrosis or apoptosis. The immediate response to brain injury is represented by the necrosis pathway and the delayed responses are
represented by the inflammatory and the apoptotic pathways.
II. NEUROPHYSIOLOGY
ANESTHETIC AGENTS
97
FIGURE 5.2 The integration of injury mechanisms excitotoxicity, inflammation, and apoptosis that lead to cell death are shown in a temporal representation. Excitotoxicity causes neuronal depolarization, excessive presynaptic glutamate release, increased intraneuronal calcium and
increased production of free radicals. Inflammatory mechanisms lead to increased secretion of injurious interleukins and adhesion molecules.
Apoptosis is initiated by activation of cellular destructive enzymes, endonucleases, and caspases.
NONANESTHETIC AGENTS
Countless pharmaceutical compounds targeting the cascade of neuronal death were successful when tested in
the laboratory to achieve neuroprotection. However, phase II and phase III single and multicenter clinical trials of
many of those strategies failed to show any neurological benefit in the case of brain injury resulting from cardiac
and vascular surgery, stroke, head trauma, intracerebral hemorrhage, or subarachnoid hemorrhage56–59 (Table 5.4).
In cardiac surgery the use of magnesium and atorvastatin in single-center trials showed potential neuroprotective
effects, but the small number of studies, methodological inconsistencies, and weakness of the evidence do not allow
any firm conclusions.56
To date, there are no randomized controlled studies that determined the efficacies of pharmacologic agents as neuroportectants in neurosurgical patients undergoing intracranial procedures. Thus, the hope is to develop a “blanket neuroprotection” against surgical brain injury.60 This concept depends on the fact that failures of many neuroprotective agents
may have resulted from their administration outside the narrow therapeutic window which is usually when the majority
of the patients arrive at the hospital. In elective neurosurgery, the timing of surgical brain injury is known and the neuroprotective agents can be administered before, during, and after the neurosurgical intervention to prevent inevitable
brain injuries associated with routine neurosurgical procedures. A therapeutic regimen “blanket neuroprotection” can be
designed using apparently neuroprotective agents. This concept has not been studied in patients and has to be initially
validated in animal models of surgical brain injury.60
ANESTHETIC AGENTS
Many laboratory studies have shown neuroprotective effects of various volatile and intravenous anesthetics.
However, recent reviews of the topic have shown conflicting evidence pertaining to their clinical neuroprotective
effects. Anesthetics may have temporary cerebrodynamic benefits in decreasing cerebral metabolism, ICP, and brain
volume but their role in improving neurologic mortality or morbidity is still elusive.56,61–63 In the IHAST study, anesthetic agents did not impact the odds of favorable neurologic function in patients undergoing clipping of ruptured
cerebral aneurysms who had temporary arterial clamping during the procedure.64 Such uncertainty about the neuroprotective effects of anesthetic agents has been shown in another large multicenter trial, the GALA trial for comparing
the use of general anesthesia versus local anesthesia for carotid endarterectomy.65 The incidence of stroke at postoperative day 30 was similar in patients having general or local anesthesia.
Anesthetic preconditioning is a phenomenon of transient organ exposure to anesthetic clinical concentrations that
triggers endogenous cellular neuroprotective processes. Similarly, anesthetic postconditioning describes the neuroprotection induced by introducing short episodes of anesthetic exposure during the early phase of reperfusion after a
prolonged episode of ischemia. The molecular processes stimulated during these cellular events include activation of
II. NEUROPHYSIOLOGY
98
5. BRAIN PROTECTION IN NEUROSURGERY
TABLE 5.4 Examples of Completed Multicenter Randomized Controlled Trials for Neuroprotection as Determined by Improvement in
Neurologic Function. Pharmaceutical Agents with Diverse Molecular Targets were Tested
Drug
Clinical Setting
Target and Action
Result
Gavestinel (GAIN trial)
Acute ischemic stroke
NMDA receptor antagonist
Failed
Mg2+
Acute ischemic stroke
NMDA receptor antagonist
Failed
Flunatrizine (FIST trial)
Acute ischemic stroke
Ca2+
channel blocker
Failed
Nimodipine (VENUS trial)
Acute ischemic stroke
Ca2+ channel blocker
Failed
Clomethiazole (CLASS trial)
Acute ischemic stroke
GABAA receptor agonist
Failed
Citalopram (TALOS trial)
Acute ischemic stroke
Selective serotonin reuptake inhibitor
Failed
Repinotan (mRECT trial)
Acute ischemic stroke
Serotonin receptor agonist
Failed
NXY-059 (SAINT I and II trials)
Acute ischemic stroke
Antioxidant
Failed
Trilazad (RANTASS trial)
Subarachnoid hemorrhage
Antioxidant
Failed
Progesterone (ProTECT III & SyNAPSE trials)
Traumatic brain injury
Antioxidant and antiinflammatory
Failed
Citicoline trial
Traumatic brain injury
Nonspecific (cell membrane stabilizer)
Failed
Erythropoietin (EPO stroke trial)
Acute ischemic stroke
Nonspecific (hemopoietic agent)
Failed
Erythropoietin (TENPEAKS trial)
Coronary artery bypass
Nonspecific (hemopoietic agent)
Failed
(IMAGES and FAST-MAG trials)
sarcolemmal and mitochondrial ATP-potassium dependent channels, stimulation of adenosine receptors and the initiation of cellular signaling cascades. This will prevent acute and delayed neuronal death by necrosis and apoptosis,
respectively.66 There are no clinical data that convincingly support the occurrence of anesthetic pre- or postconditioning in the clinical setting despite the numerous laboratory reports showing their benefit.
Xenon recently reemerged as an anesthetic agent, and its neuroprotective properties have been shown in the laboratory. The presumed mechanism is competitive inhibition of the NMDA receptor. Consequently, xenon suppresses
the increase in intraneuronal calcium concentration thus preventing cell death67 (Fig. 5.1). Clinical trials are underway to elicit the neuroprotection efficacy of xenon in several clinical situations including cardiopulmonary bypass,
neonatal asphyxia, and cardiac arrest. To date, none of these trials have been completed. Nonetheless, small clinical
studies did not show any benefit in using xenon to decrease the incidence of neurologic morbidity.68
CONCLUSION
There is no convincing evidence that pharmaceutical agents including anesthetics can protect the brain during
vulnerable clinical situation including neurosurgical procedures. Hence, the anesthesiologist is compelled to use
nonpharmacological approaches that may be based on physiologic and pharmacologic empiric evidence to protect
the brain during surgery. Accordingly, CPP should be maintained by controlling the systemic mean arterial pressure
above 80 mmHg. Additionally, induced hypertension should be used in certain neurosurgical situations in which
there is arterial vessel occlusion or a deleterious change in the cerebral autoregulation mechanism. Blood glucose levels should be kept within a liberal normal range (7.8–10 mmol/L; 140–180 mg/L) and intensive insulin therapy protocols avoided. Adequate hemoglobin levels preoperatively (≥120 g/dL) and intraoperatively (≥90 g/dL) will ensure
optimum oxygen delivery to the brain. To date there is no conclusive evidence that mild hypothermia (32–35°C) has
neuroprotective benefit. However, because of its relative safety, the anesthesiologist who would like to extrapolate its
beneficial effects from asphyxiated neonates and cardiac arrest patients may use it cautiously.
References
1. Jadhav V, Solaroglu I, Obenaus A, Zhang JH. Neuroprotection against surgically induced brain injury. Surg Neurol 2007;67:15–20.
2. Badenes R, Gruenbaum SE, Bilotta F. Cerebral protection during neurosurgery and stroke. Curr Opin Anaesthesiol 2015;28(5):532–6.
3. Turner RC, Dodson SC, Rosen CL, Huber JD. The science of cerebral ischemia and the quest for neuroprotection: navigating past failure to future success. J
Neurosurg 2013;118(5):1072–85.
4. Drake CG, Barr HW, Coles JC, Gergely NF. The use of extracorpeal circulation and profound hypothermia in the treatment of ruptured intracranial aneurysms. J
Neurosurg 1964;21:575–81.
II. NEUROPHYSIOLOGY
REFERENCES
99
5. Spetzler RF, Hadley MN, Rigamonti D, Carter LP, Raudzens PA, Shedd SA, et al. Aneurysms of the basilar artery treated with circulatory arrest, hypothermia, and
barbiturate cerebral protection. J Neurosurg 1988;68(6):868–79.
6. Frantseva MV, Carlen PL, El Beheiry H. A submersion method to induce hypoxic damage in organotypic hippocampal cultures. J Neurosci Methods 1999;89:25–31.
7. Krieger DW, Yenari MA. Therapeutic hypothermia for acute ischemic stroke: what do laboratory studies teach us? Stroke 2004;35(6):1482–9.
8. Laptook AR, Corbett RJ, Sterett R, Garcia D, Tollesfbol G. Quantitative relationship between brain temperature and energy utilization rate measured in vivo using
31P and 1H magnetic resonance spectroscopy. Pediatr Res 1995;38:919–25.
9. Zhao H, Steinberg GK, Sapolsky RM. General versus specific actions of mild-moderate hypothermia in attenuating cerebral ischemic damage. J Cereb Blood Flow
Metab 2007;27(12):1879–94.
10. Yenari MA, Han HS. Neuroprotective mechanisms of hypothermia in brain ischaemia. Nat Rev Neurosci 2012;13(4):267–78.
11. Galvin IM, Levy R, Boyd JG, Day AG, Wallace MC. Cooling for cerebral protection during brain surgery. Cochrane Database Syst Rev 2015;(1). ArtNo.: CD006638.
12. Hindman BJ, Todd MM, Gelb AW, Loftus CM, Craen RA, Schubert A, et al. Mild hypothermia as a protective therapy during intracranial aneurysm surgery: a
randomized prospective pilot trial. Neurosurgery 1999;44(1):23–32.
13. Todd MM, Hindman BJ, Clarke WR, Torner JC, Intraoperative Hypothermia for Aneurysm Surgery Trial (IHAST) Investigators. Mild intraoperative hypothermia
during surgery for intracranial aneurysm. N Engl J Med 2005;352(2):135–45.
14. Qiu W, Zhang Y, Sheng H, Zhang J, Wang W, Liu W, et al. Effects of therapeutic mild hypothermia on patients with severe traumatic brain injury after craniotomy.
J Crit Care 2007;22(3):229–35.
15. Els T, Oehm E, Voigt S, Klisch J, Hetzel A, Kassubek J. Safety and therapeutical benefit of hemicraniotomy combined with mild hypothermia in comparison with
hemicraniotomy alone in patients with malignant ischemic stroke. Cerebrovasc Dis 2006;21(1–2):79–85.
16. Li LR, You C, Chaudhary B. Intraoperative mild hypothermia for postoperative neurological deficits in people with intracranial aneurysm. Cochrane Database Syst
Rev March 22, 2016;3:CD008445.
17. Zhao ZX1, Wu C, He M. A systematic review of clinical outcomes, perioperative data and selective adverse events related to mild hypothermia in intracranial
aneurysm surgery. Clin Neurol Neurosurg 2012;114(7):827–32.
18. Arrich J, Holzer M, Havel C, Müllner M, Herkner H. Hypothermia for neuroprotection in adults after cardiopulmonary resuscitation. Cochrane Database Syst Rev
February 15, 2016;2:CD004128.
19. Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Gutteridge G, et al. Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med 2002;346(8):557–63.
20. Shankaran S, Laptook AR, Ehrenkranz RA, Tyson JE, McDonald SA, Donovan EF, et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med 2005;353:1574–84.
21. Jacobs SE, Berg M, Hunt R, Tarnow-Mordi WO, Inder TE, Davis PG. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev
January 31, 2013;(1):CD003311.
22. Andrews RJ. Neuroprotection in surgery. Development of a pharmacologic cocktail for intraoperative use. Ann N Y Acad Sci 1997;15(825):288–304.
23. Moore LE, Sharifpour M, Shanks A, Kheterpal S, Tremper KK, Mashour GA. Cerebral perfusion pressure below 60 mm Hg is common in the intraoperative setting.
J Neurosurg Anesthesiol 2012;24(1):58–62.
24. Rasulo FA, Girardini A, Lavinio A, De Peri E, Stefini R, Cenzato M, et al. Are optimal cerebral perfusion pressure and cerebrovascular autoregulation related to
long-term outcome in patients with aneurysmal subarachnoid hemorrhage? J Neurosurg Anesthesiol 2012;24(1):3–8.
25. Juul N, Morris GF, Marshall SB, Marshall LF. Intracranial hypertension and cerebral perfusion pressure: influence on neurological deterioration and outcome in
severe head injury. The Executive Committee of the International Selfotel Trial. J Neurosurg 2000;92(1):1–6.
26. Tackla R, Hinzman JM, Foreman B, Magner M, Andaluz N, Hartings JA. Assessment of cerebrovascular autoregulation using regional cerebral blood flow in surgically managed brain trauma patients. Neurocrit Care 2015;23:339–46.
27. Abou-Chebl A, Yeatts SD, Yan B, Cockroft K, Goyal M, Jovin T, et al. Impact of general anesthesia on safety and outcomes in the endovascular arm of Interventional
Management of Stroke (IMS) III trial. Stroke 2015;46(8):2142–8.
28. LöwhagenHendén P, Rentzos A, Karlsson JE, Rosengren L, Sundeman H, Reinsfelt B, et al. Hypotension during endovascular treatment of ischemic stroke is a risk
factor for poor neurological outcome. Stroke 2015;46(9):2678–80.
29. Leonardi-Bee J, Bath PM, Phillips SJ, Sandercock PA, IST Collaborative Group. Blood pressure and clinical outcomes in the International Stroke Trial. Stroke
2002;33(5):1315–20.
30. Garg RK, Liebling SM, Maas MB, Nemeth AJ, Russell EJ, Naidech AM. Blood pressure reduction, decreased diffusion on MRI, and outcomes after intracerebral
hemorrhage. Stroke 2012;43(1):67–71.
31. Schubert A. Cardiovascular therapy of neurosurgical patients. Best Pract Res Clin Anaesthesiol 2007;21:483–96.
32. El Beheiry H. Protecting the brain during neurosurgical procedures: strategies that can work. Curr Opin Anaesthesiol 2012;25(5):548–55.
33. Kobayashi M, Ogasawara K, Yoshida K, Sasaki M, Kuroda H, Suzuki T, et al. Intentional hypertension during dissection of carotid arteries in endarterectomy
prevents postoperative development of new cerebral ischemic lesions caused by intraoperative microemboli. Neurosurgery 2011;69(2):301–7.
34. LeSar CJ, Sprouse LR, Harris WB. Permissive hypertension during awake eversion carotid endarterectomy: a physiologic approach for cerebral protection. J Am
Coll Surg 2014;218:760–7.
35. Reynolds MR, Buckley RT, Indrakanti SS, Turkmani AH, Oh G, Crobeddu E, et al. The safety of vasopressor-induced hypertension in subarachnoid hemorrhage
patients with coexisting unruptured, unprotected intracranial aneurysms. J Neurosurg 2015;123:862–71.
36. Sebranek JJ, Lugli AK, Coursin DB. Glycaemic control in the perioperative period. Br J Anaesth 2013;111(Suppl. 1):i18–34.
37. Godoy DA, Di Napoli M, Biestro A, Lenhardt R. Perioperative glucose control in neurosurgical patients. Anesthesiol Res Pract 2012;2012:690362.
38. Kusin DJ, Ahn UM, Ahn NU. The influence of diabetes on surgical outcomes in cervical myelopathy. Spine (Philadelphia, PA, 1976) March 10, 2016. [Epub ahead of
print]. PMID: 26974831.
39. McGirt MJ, Woodworth GF, Brooke BS, Coon AL, Jain S, Buck D, et al. Hyperglycemia independently increases the risk of perioperative stroke, myocardial infarction, and death after carotid endarterectomy. Neurosurgery 2006;58(6):1066–73.
40. Pasternak JJ, McGregor DG, Schroeder DR, Lanier WL, Shi Q, Hindman BJ, et al. Hyperglycemia in patients undergoing cerebral aneurysm surgery: its association
with long-term gross neurologic and neurophysiological function. Mayo Clin Proc 2008;83(4):406–17.
41. Filho NO, Alves RL, Fernandes AT, Castro FS, Melo JR, Módolo NS. Association of increased morbidity with the occurrence of hyperglycemia in the immediate
postoperative period after elective pediatric neurosurgery. J Neurosurg Pediatr 2016;17(5):625–9.
42. Atkins JH, Smith DS. A review of perioperative glucose control in the neurosurgical population. J Diabetes Sci Technol 2009;3(6):1352–64.
43. Tiemessen CA, Hoedemaekers CW, Van lersel FM, Rösken GH, van der Hoeven J, Biessels GJ, et al. Intensive insulin therapy increases the risk of hypoglycaemia
in neurocritical care patients. J Neurosurg Anesthesiol 2011;23(3):206–14.
44. The NICE-SUGAR study investigators. Intensive versus conventional glucose control in critically ill patients. N Engl J Med 2009;360:1283–97.
45. Gandhi GY, Nuttall GA, Abel MD, Mullany CJ, Schaff HV, O’Brien PC, et al. Intensive intraoperative insulin therapy versus conventional glucose management
during cardiac surgery: a randomized trial. Ann Intern Med 2007;146(4):233–43.
46. Oddo M, Schmidt JM, Carrera E, Badjatia N, Connolly ES, Presciutti M, et al. Impact of tight glycemic control on cerebral glucose metabolism after severe brain
injury: a microdialysis study. Crit Care Med 2008;36(12):3233–8.
II. NEUROPHYSIOLOGY
100
5. BRAIN PROTECTION IN NEUROSURGERY
47. Moghissi ES, Korytkowski MT, DiNardo M, Einhorn D, Hellman R, Hirsch IB, et al. American Association of Clinical Endocrinologists and American Diabetes
Association consensus statement on inpatient glycemic control. Diabetes Care 2009;32(6):1119–31.
48. Tsui AK, Marsden PA, Mazer CD, Sled JG, Lee KM, Henkelman RM, et al. Differential HIF and NOS responses to acute anemia: defining organ-specific hemoglobin thresholds for tissue hypoxia. Am J Physiol Regul Integr Comp Physiol 2014;307(1):R13–25.
49. Kurtz P, Helbok R, Claassen J, Schmidt JM, Fernandez L, Stuart RM, et al. The effect of packed red blood cell transfusion on cerebral oxygenation and metabolism
after subarachnoid hemorrhage. Neurocrit Care 2016;24(1):11821.
50. Bydon M, Abt NB, Macki M, Brem H, Huang J, Bydon A, et al. Preoperative anemia increases postoperative morbidity in elective cranial neurosurgery. Surg Neurol
Int 2014;5:156.
51. Stein M, Brokmeier L, Herrmann J, Scharbrodt W, Schreiber V, Bender M, et al. Mean hemoglobin concentration after acute subarachnoid hemorrhage and the
relation to outcome, mortality, vasospasm, and brain infarction. J Clin Neurosci 2015;22(3):530–4.
52. Sun J, Tan G, Xing W, He Z. Optimal hemoglobin concentration in patients with aneurysmal subarachnoid hemorrhage after surgical treatment to prevent symptomatic cerebral vasospasm. Neuroreport 2015;26(5):263–6.
53. Gruenbaum SE, Ruskin KJ. Red blood cell transfusion in neurosurgical patients. Curr Opin Anaesthesiol 2014;27(5):470–3.
54. Sheth KN, Gilson AJ, Chang Y, Kumar MA, Rahman RM, Rost NS, et al. Packed red blood cell transfusion and decreased mortality in intracerebral hemorrhage.
Neurosurgery 2011;68(5):1286–92.
55. Alan N, Seicean A, Seicean S, Neuhauser D, Weil RJ. Impact of preoperative anemia on outcomes in patients undergoing elective cranial surgery.
J Neurosurg 2014;120(3):764–72.
56. Bilotta F, Gelb AW, Stazi E, Titi L, Paoloni FP, Rosa G. Pharmacological perioperative brain neuroprotection: a qualitative review of randomized clinical trials. Br
J Anaesth 2013;110(Suppl. 1):i113–20.
57. Minnerup J, Sutherland BA, Buchan AM, Kleinschnitz C. Neuroprotection for stroke: current status and future perspectives. Int J Mol Sci 2012;13(9):11753–72.
58. Stocchetti N, Taccone FS, Citerio G, Pepe PE, Le Roux PD, Oddo M, et al. Neuroprotection in acute brain injury: an up-to-date review. Crit Care 2015;19(1):186.
59. Kellner CP, Connolly Jr ES. Neuroprotective strategies for intracerebral hemorrhage: trials and translation. Stroke 2010;41:S99–102.
60. Jadhav V, Zhang JH. Surgical brain injury: prevention is better than cure. Front Biosci 2008;13:3793–7.
61. Deng J, Lei C, Chen Y, Fang Z, Yang Q, Zhang H, et al. Neuroprotective gases – fantasy or reality for clinical use? Prog Neurobiol 2014;115:210–45.
62. Schifilliti D, Grasso G, Conti A, Fodale V. Anaesthetic-related neuroprotection: intravenous or inhalational agents? CNS Drugs 2010;24:893–907.
63. Kruger T, Hoffmann I, Blettner M, Borger MA, Schlensak C, Weigang E, et al. Intraoperative neuroprotective drugs without beneficial effects? Results of the
German Registry for Acute Aortic Dissection Type A (GERAADA). Eur J Cardiothorac Surg 2013;44(5):939–46.
64. Hindman BJ, Bayman EO, Pfisterer WK, Torner JC, Todd MM, IHAST Investigators. No association between intraoperative hypothermia or supplemental protective drug and neurologic outcomes in patients undergoing temporary clipping during cerebral aneurysm surgery: findings from the Intraoperative Hypothermia
for Aneurysm Surgery Trial. Anesthesiology 2010;112(1):86–101.
65. GALA Trial Collaborative Group. General anaesthesia versus local anaesthesia for carotid surgery (GALA): a multicentre, randomised controlled trial. Lancet
2008;372(9656):2132–42.
66. Zwerus R, Absalom A. Update on anesthetic neuroprotection. Curr Opin Anaesthesiol 2015;28(4):424–30.
67. Banks P, Franks NP, Dickinson R. Competitive inhibition at the glycine site of the N-methyl-D-aspartate receptor mediates xenon neuroprotection against
hypoxia-ischemia. Anesthesiology 2010;112:614–22.
68. Maze M. Preclinical neuroprotective actions of xenon and possible implications for human therapeutics: a narrative review. Can J Anaesth 2016;63(2):212–26.
II. NEUROPHYSIOLOGY
S E C T I O N
I I I
NEUROPHARMACOLOGY
This page intentionally left blank
C H A P T E R
6
Neuropharmacology
P. Ganjoo1, I. Kapoor2
1GB
Pant Hospital, New Delhi, India; 2All India Institute of Medical Sciences, New Delhi, India
O U T L I N E
Anesthetic Drugs and Sedatives
104
Intravenous Anesthetic Agents
Barbiturates
Cerebral Effects of Barbiturates
Other Effects of Barbiturates
Current Status
Recent Research
Propofol
Cerebral Effects of Propofol
Other Effects of Propofol
Current Status
Recent Research
Etomidate
Cerebral Effects of Etomidate
Other Effects of Etomidate
Current Status
Recent Research
Ketamine
Cerebral Effects of Ketamine
Other Effects of Ketamine
Current Status
Recent Research
Benzodiazepines
Cerebral Effects of Benzodiazepines
Other Effects of Benzodiazepines
Current Status
Recent Research
Opioids
Cerebral Effects of Opioids
Other Effects of Opioids
104
105
105
105
107
107
107
107
107
107
107
108
108
108
108
108
108
109
109
109
109
109
110
110
110
110
110
110
111
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00006-3
Current Status
Recent Research
111
111
Dexmedetomidine
Recent Research
111
111
Inhalational Anesthetic Agents
Desflurane
Cerebral Effects of Desflurane
Other Effects of Desflurane
Current Status
Recent Research
Sevoflurane
Cerebral Effects of Sevoflurane
Other Effects of Sevoflurane
Current Status
Recent Research
Isoflurane
Cerebral Effects of Isoflurane
Other Effects of Isoflurane
Current Status
Recent Research
Nitrous Oxide
Cerebral Effects of N2O
Other Effects of N2O
Current Status
Recent Research
Xenon
Cerebral Effects of Xenon
Other Effects of Xenon
Current Status
103
111
111
112
112
112
113
113
113
113
113
113
113
114
114
114
114
114
114
114
115
115
115
115
115
115
© 2017 Elsevier Inc. All rights reserved.
104
6. NEUROPHARMACOLOGY
Neuromuscular Blocking Agents
Depolarizing Neuromuscular Blocking Agents
Nondepolarizing Neuromuscular Blocking Agents
115
115
116
Local Anesthetic Agents
116
Miscellaneous Drugs
116
Future Directions in Neuropharmacology
116
Conclusion
116
References
118
With the advent of newer neurosurgeries like minimally invasive procedures, endovascular neurosurgeries
and complex revascularization operations, increased use of sophisticated intraoperative neurological monitors,
and significant advances in neurocritical care, the scope of neuroanesthesia has also expanded tremendously.
This has necessitated a quest for more capable, more rapidly titratable, and safer drugs for anesthesia and sedation. Drugs used in neuroanesthesia should be able to provide optimal brain conditions for surgery and also help
maintain adequate brain tissue perfusion to meet increased regional metabolic demands. The ability to not only
rapidly achieve deeper intraoperative anesthesia levels but also allow quick postoperative recovery of consciousness, good analgesic effects, no epileptogenic effects, minimal interference with neuromonitoring modalities, no
adverse effects on other body organs, and capability of preserving systemic and cerebral hemodynamic stability
are some of the desirable attributes in a neuroanesthetic drug. The beneficial cerebral effects of these drugs would
be maintenance of cerebral autoregulation, vasoreactivity to carbon dioxide (CO2) and coupling of cerebral blood
flow (CBF) and cerebral metabolic rate (CMR), and prevention of increases in intracranial pressures (ICPs) and
cerebral blood volume (CBV).
The correct choice of drugs and their doses for anesthesia and sedation is thus vital in preventing further worsening of
the intracranial pathology or introduction of a new cerebral insult. This necessitates a better understanding of the cerebral
effects and other important issues related to the commonly used anesthetics and sedatives, and is the main scope of this
chapter. Discussion on other drugs and adjuvants used in neurosurgical practice can be found elsewhere in this book.
ANESTHETIC DRUGS AND SEDATIVES
Anesthetic drugs cause their cerebral effects by producing metabolic and functional changes in the central nervous
system (CNS). Broadly, intravenous agents tend to reduce both CBF and CMR in a parallel manner and maintain their
coupling, while inhalational agents decrease the CMR and increase the CBF and appear not to maintain coupling.
However, anesthesia-related CBF–CMR coupling may vary under different brain conditions as the effects of anesthetics
on CBF is influenced by both a direct effect on the cerebral vascular tone (vasoconstriction or vasodilatation) and
indirect changes in the CMR. Furthermore, this dual mechanism of action makes it difficult to predict whether
anesthetics can cause an “intracerebral steal” or the beneficial “reverse intracerebral steal” phenomenon in the
pathological brain in which CO2 reactivity and autoregulation may be lost. Anesthetics also produce changes in
the ICP by changing the CBF and thereby the CBV, and by their influence on cerebrospinal fluid (CSF) dynamics, i.e.,
the rate of production and reabsorption of CSF. The cerebral effects of anesthetics are also governed by their systemic
effects, primarily on the blood pressure, arterial CO2, and body temperature.
A promising attribute of anesthetic drugs that has been identified lately is that some of them have the potential
for neuroprotective effects and may even be able to reduce neuronal damage from ischemic insults. These effects
are attributed to their ability to reduce neuronal activity and metabolic rates. Lidocaine, thiopental, and sevoflurane
have shown to be protective against ischemia in animal studies, particularly when given at the beginning of an ischemic insult due to their proposed “preconditioning effect.” However, the clinical utility of anesthetics in preventing
and ameliorating ischemic damage needs further investigation. The neuroprotective effects of various anesthetic
drugs are discussed in a separate chapter.
Recent suggestions that anesthetic drugs can cause neurotoxicity and postoperative cognitive dysfunction (POCD) is an
area of great concern for the anesthetists. Detailed discussion on this important subject can be found elsewhere in this book.
INTRAVENOUS ANESTHETIC AGENTS
Intravenous anesthetic agents are small hydrophobic compounds that when injected, enter the highly perfused and
lipophilic tissues in the brain and spinal cord where they produce anesthesia in a single circulation time. Termination
of anesthesia with these drugs does not reflect metabolism but their redistribution out of the CNS into the blood and
III. NEUROPHARMACOLOGY
105
INTRAVENOUS ANESTHETIC AGENTS
then into the lesser perfused tissues like muscles and viscera. Drug redistribution can cause accumulation and slower
recovery from its effects.
Barbiturates
Barbiturates are CNS depressant drugs commonly used in neurological practice for providing mild sedation to total
anesthesia, and also as anticonvulsants, hypnotics, anxiolytics and analgesics. While sodium thiopental (thiobarbiturates), thiamylal (thiobarbiturates), and methohexital (oxybarbiturates) are used for induction of anesthesia, amobarbital
is mainly used for performing “the Wada test,” also known as the “intracarotidsodium amobarbital procedure” used for
testing cerebral language and memory representation of the cerebral hemispheres. Barbiturates are derivatives of barbituric acid (2,4,6-trioxohexahydropyrimidine) (Fig. 6.1)1 and act primarily as γ-aminobutyric acid (GABA) receptor agonists (http://en.wikipedia.org/wiki/Barbiturate).2 They also act on the glutamate, adenosine, and nicotinic acetylcholine
receptors. The clinically recommended dosages and pharmacokinetics of barbiturates are summarized in Table 6.1.3–6
Cerebral Effects of Barbiturates (Table 6.2)
Barbiturates produce cerebral function depression and cause a dose-dependent decrease in cerebral metabolic rate
for oxygen consumption (CMRO2) and CBF till the electroencephalograph (EEG) becomes isoelectric.7 The induction
dose of thiopental causes a 25–30% decrease in CMRO2 with a maximum 55% decrease occurring at 2–5 times that
dose.8 They cause a reduction in ICP9 due to decreases in CBF and CBV, and also maintain cerebral autoregulation
and CO2 reactivity. In low doses, thiopental sodium causes no change in the rate of CSF formation, and either no
change or an increase in the resistance to reabsorption of CSF, but in higher doses, it causes decrease in CSF formation rate with either no change or a decrease in the resistance to resorption resulting in a raised ICP. As autoregulation is similar in both brain and spinal cord,10 high-dose barbiturate therapy causes a significant decrease in spinal
cord blood flow (SCBF) suggestive of a protective effect of barbiturates on spinal cord injury,11 although spinal cord
metabolism seems to be less sensitive to depression by barbiturates.12
Other Effects of Barbiturates
Barbiturates cross the blood–brain barrier (BBB) very rapidly. Methohexital has been shown to reduce the seizure threshold,13 and hence seizure activity may be a concern on emergence from barbiturate anesthesia. Cognitive
impairment on chronic use of barbiturates is well known; both propofol and barbiturates were shown to cause
severe cognitive side effects, but the result was confounded by the differences in age distribution in the two study
groups.14 Curcumin, a substance in turmeric, is being considered as a safe and effective adjuvant to barbiturates in
preventing cognitive impairment due to its antioxidant, antiinflammatory, and neuroprotective properties.15 Recent
literature has demonstrated that drugs that antagonize N-methyl-d-aspartate (NMDA) receptors and agonize GABA
receptors produce widespread neurodegeneration in the developing brain.16 Fredriksson et al. observed a reduction
in cognitive function in rodents, after a combination of thiopental or propofol and ketamine at postnatal day 10 and
at 8–10 weeks of age.17 Significant systemic effects of barbiturates include hypotension and respiratory depression.
FIGURE 6.1 Barbituric acid.
TABLE 6.1 Physical Properties of Intravenous Anesthetic Agents
Anesthetic Agents
Thiopentone Sodium
Propofol
Etomidate
Ketamine
Induction dose (mg/kg)
3–5
2.0–2.5
0.2–0.4
0.5–1.5
Induction duration (mins)
5–8
4–8
4–8
10–15
t1/2 (h)
12.1
1.8
2.9–5.3
3
Clearance (mL/kg/min)
3.4
23–50
18–25
19.1
Protein binding(%)
85
95–99
76
12
t1/2, half-life.
III. NEUROPHARMACOLOGY
III. NEUROPHARMACOLOGY
↑↑
↑↑
↓↓
↑/−
↓/−
↓/−
−
−
↓/−
↑
↓↓
↓↓
↑↑
↓/−
↑/−/↓
↑/−/↓
↑/−/↓
−
−
↑/−
↓
Propofol
Etomidate
Ketamine
Midazolam
Fentanyl
Sufentanil
Remifentanil
Vecuronium
Rocuronium
Succinylcholine
Dexmedetomidine
↓↓
↑/−
−
−
↓/−/↑
↓/−/↑
↓/−/↑
↓
↑↑
↓↓
↓↓
↓↓↓
↑
↑
↑
↑
↓
CBF
↓↓
↑/−
−
−
↓/−
↓/−
↓/−
↓
↑
↓↓
↓↓
↓↓↓
↑
↓↓
↓↓
↓↓
↓↓
CMRO2
−
−
−
−
−
−
−/↓
−/↓
−
−/↓
−
−/↓
−
−
↑
↓/−/↑
?
Resistance to Resorption
−
−
−
−
−
↑/−/↓
↑/−/↓
−/↑
↑
−/↓
−
↑/−/↓
−
−/↑
↓
−
?
CSF Formation
CSF Dynamics
+
/
/
/
+
+
+
+
+
+
+
+
+
+
+
+
+
BBB
Yes
No
No
No
Yes
Yes
Yes
No
Yes
Yes
No
No
Yes
No
Yes
No
No
Epileptogenic
BBB, blood–brain barrier; CBF, cerebral blood flow; CMRO2, cerebral metabolic rate of oxygen consumption; CPP, cerebral perfusion pressure; CSF, cerebrospinal fluid; ICP, intracranial pressure; ↑, increase;
↓, decrease; −, no effect; +, cross; /, does not cross; ?, unknown.
−
↑↑
↓↓
↓/−
↑/−−
Desflurane
Thiopentone sodium
↓/−
↑/−
Sevoflurane
↓↓
↓/−
↑/−
Isoflurane
↑↑
↓/−
↓
Xenon
Nitrous oxide
CPP
ICP
Anesthetic Agents/Muscle
Relaxants
TABLE 6.2 Cerebral Effects of Anesthetic Agents and Muscle Relaxants
106
6. NEUROPHARMACOLOGY
INTRAVENOUS ANESTHETIC AGENTS
107
FIGURE 6.2 2,6-Diisopropylphenol.
Current Status
Due to its ICP-reducing and possibly neuroprotective effects, barbiturates continue to be used widely in neurosurgical anesthesia, especially in patients with raised ICP. However, barbiturates may require vasopressor support to
maintain cerebral perfusion pressure (CPP) and may cause delayed recovery due to accumulated effects.
Recent Research
At present, there is no evidence to prove that the administration of barbiturates in patients with acute severe head
injury improves the overall outcome.18 A systematic review in 2012 has also found inefficient evidence in favor of its
effectiveness as an anxiolytic drug.19
Propofol
The chemical formulation of propofol is 2,6-diisopropylphenol (Fig. 6.2). It is used for induction and maintenance
of general anesthesia as well as for sedation. Propofol is also known as “milk of amnesia,” because of its milklike
appearance.20 The presently available preparation of propofol is 1% (10 mg/mL), which contains 2.25% of glycerol
as a tonicity-stabilizing agent, 10% soybean oil, and 1.2% purified egg phospholipid as an emulsifier, with sodium
hydroxide to adjust the pH. The mechanism of action of propofol is either though activation of GABA receptors21,22
or blocking action on sodium channels.23,24 A 2004 research also suggests that the endocannabinoid system may also
contribute significantly to the anesthetic action of propofol.25 The recommended clinical dosage and pharmacokinetics of propofol is summarized in Table 6.1.26–28
Cerebral Effects of Propofol (Table 6.2)
Propofol causes decreases in CMRO2 and CBF similar to barbiturates,26 the reduction in CMRO2 being less than
decreases in CBF. It also causes decreases in ICP by decreasing the CBF; the ICP is lowered while maintaining the
CPP, unlike with barbiturates and inhalational anesthetic agents like sevoflurane and isoflurane.29 In clinical dosages, it does not affect cerebral autoregulation.30 The CO2 vasoreactivity is preserved, and hence, hyperventilation
will decrease the ICP under propofol anesthesia. Propofol has no effect on the production and resorption of CSF.31
The SCBF autoregulation is maintained with low- and high-dose propofol infusion.32 It induces depression of metabolic activity in spinal cord gray matter also.33 Propofol may also have direct cerebral vasoconstrictive activity.
Other Effects of Propofol
Propofol as a highly lipophilic drug crosses BBB and placenta and distributes into the breast milk too. The anticonvulsant effect of propofol is not clear as some data suggest a proconvulsant effect when used with other drugs.34
A measurable postoperative memory impairment has been observed in patients who have received 1–2 h of anesthesia with propofol and remifentanil.35 No differences in the incidence of POCD has been demonstrated in patients
anesthetized with xenon, propofol, desflurane, or sevoflurane.36–39 Propofol anesthesia for prolonged period (5 h)
can cause death of neurons and oligodendrocytes in both the fetal and neonatal brain.40 Hence prolonged infusion in
small children is best avoided as it can cause acidosis, heart failure, and even death.
Current Status
It is useful anesthetic for neurosurgery due to favorable cerebral effects, rapid onset and recovery, and minimal
interference with neurophysiological monitoring. Cerebral vasoconstriction makes it a suitable drug for vascular
neurosurgeries.41 It is useful in patients with intracranial hypertension, but caution is required as it can decrease CPP
due to associated hypotension.
Recent Research
Propofol is widely used in pediatric and adult populations its safety in neonates has not been defined and at
present, there is no evidence supporting its use in neonates.42 Both thiopental sodium and propofol are used for the
treatment of refractory status epilepticus, but there is no clear evidence supporting the efficacy of either of the two
drugs in terms of clinical outcome.43
III. NEUROPHARMACOLOGY
108
6. NEUROPHARMACOLOGY
Etomidate
Etomidate is a short-acting anesthetic agent used for induction of general anesthesia and for sedation.44 The chemical formulation of etomidate is ethyl 3-[(1R)-1-phenylethyl] imidazo 5-carboxylate (Fig. 6.3). Etomidate has limited
suppression of ventilation and lack of histamine release and protects from myocardial and cerebral ischemia.45 The
“etomidate speech and memory test” is used to determine speech lateralization in patients prior to performing
lobectomies to remove epileptogenic centers in the brain. The drug acts primarily on GABA receptors46 and is highly
protein bound. It is metabolized by hepatic and plasma esterases to inactive products.47,48 The pharmacological characteristics of etomidate are described in Table 6.1.49–51
Cerebral Effects of Etomidate (Table 6.2)
Etomidate reduces CMRO2 and CBF in a parallel manner to produce an isoelectric EEG.52 The maximal fall in CMRO2 is
achieved after a fall in CBF, and this effect is possibly due to a direct effect causing cerebral vasoconstriction.53 It also causes
a dose-dependent fall in ICP following reduction in CBF. In pediatric patients with severe traumatic brain injury, singledose etomidate administration results in significant reductions in ICP and improvement in CPP.54 The reactivity to CO2
is maintained well under etomidate anesthesia. Its effect on cerebral autoregulation has not been evaluated. Etomidate
in low dose causes no change in the rate of CSF formation and resistance to CSF resorption. However, in higher doses
etomidate causes reduction in rate of CSF formation with either decrease or no change in resistance to CSF resorption.55
Other Effects of Etomidate
It is a hydrophobic drug and crosses BBB very rapidly like barbiturates; the CNS effect lasts only for few minutes.
Etomidate has been used to protect against cerebral ischemia in high risk patients, however no human trials are
available to support the evidence. Also its role in seizure control has not been proven.34 Etomidate can cause POCD
in elderly patients.56 Prolonged infusion of etomidate can cause propylene glycol toxicity that can clinically present
as hyperosmolality with an increased osmolal gap, hemolysis, hemoglobinuria, and metabolic acidosis.57 It can cause
adrenocortical suppression and involuntary muscle activity.
Current Status
Lack of cardiovascular side effects makes etomidate a useful neuroanesthetic. It can also be used safely for neurophysiological monitoring as it maintains both somatosensory evoked potential (EP) and motor EP threshold.58,59 It
should be avoided or used cautiously in patients with seizure history.
Recent Research
Etomidate provides more stable hemodynamic parameters as compared to propofol. It can be used safely without
serious cortisol suppression lasting more than 24 h.60 In comparison with ketamine for rapid sequence induction,
etomidate does not provide superior intubating conditions and more favorable hemodynamic response to laryngoscopy and tracheal intubation.61
Ketamine
Ketamine is a phencyclidine derivative, and its chemical formulation is arylcyclohexylamine (Fig. 6.4). It produces
a state called “dissociated anesthesia,” which is characterized by the presence of dissociation between thalamocortical and limbic system.62 It also provides intense analgesia as well as amnesia. Because of the possibility of increased
airway secretions and emergence delirium, it is advised to give an antisialagogue (glycopyrrolate) and midazolam as
premedication in patients receiving ketamine. Ketamine mainly binds to NMDA receptors. It acts on other receptors
O
O
N
N
FIGURE 6.3 Ethyl 3-[(1R)-1-phenylethyl] imidazo 5-carboxylate.
III. NEUROPHARMACOLOGY
INTRAVENOUS ANESTHETIC AGENTS
109
FIGURE 6.4 Arylcyclohexylamine.
FIGURE 6.5 C9H8N2.
like opioid receptors, GABA receptors, muscarinic receptors, voltage-sensitive sodium channels, and calcium channels.63 The pharmacological characteristics of ketamine are described in Table 6.1.
Cerebral Effects of Ketamine (Table 6.2)
Unlike other intravenous anesthetic agents, ketamine increases the CBF and CMRO2. At subanesthetic doses,
ketamine acts as a potent cerebral vasodilator and increases the CBF by 60% in normal situations.64 In patients with
brain tumor and aneurysmal resection, 1 mg/kg ketamine administration does not cause increase in middle cerebral
artery velocity.65 It was believed earlier that an induction dose of ketamine significantly increases the ICP and hence
was considered contraindicated in patients with a raised ICP. However, some studies have found its use safe when
accompanied with hyperventilation.66,67 Cerebral autoregulation and CO2 reactivity are well maintained with ketamine. It increases the rate of CSF formation but either decreases or causes no change in resistance to CSF resorption.
Ketamine has protective effects on the spinal cord. It prevents loss of antioxidant activity in spinal cord tissue in cord
injury cases.68
Other Effects of Ketamine
Ketamine is highly lipid soluble and rapidly crosses the BBB producing quick onset of action and rapid recovery from anesthesia. Ketamine would be unlikely to have proconvulsant action; however, myoclonic and seizure
like activities may occur in normal patients.34 It is known to cause emergence delirium, which occurs more frequently within an hour and is less frequent in children.69 Ketamine increases the amplitude of somatosensory EPs
but decreases the auditory and visual evoked response in humans.70
Current Status
It is not the first choice in neuroanesthesia, and is avoided in patients with raised ICP or decreased intracranial
compliance.
Recent Research
According to Schreiberova et al., sedation by dexmedetomidine–ketamine–midazolam combination is a safe
and suitable method for endovascular neurointerventions. It provides hemodynamic stability without respiratory
depression.71
Benzodiazepines
The chemical formula of benzodiazepine is C9H8N2 (Fig. 6.5). Benzodiazepines exert their CNS effects by escalating the effect of GABA neurotransmitter leading to hypnosis, sedation, anxiolysis, anticonvulsant effect, and
anterograde amnesia.72 Side effects of this central action includes dizziness, sedation, weakness, loss of orientation,
headache, irritability, aggression, sleep disturbances, and confusions.
III. NEUROPHARMACOLOGY
110
6. NEUROPHARMACOLOGY
Cerebral Effects of Benzodiazepines (Table 6.2)
Benzodiazepines (midazolam, diazepam, lorazepam) cause decrease in CBF and CMRO2 in all doses. Midazolam
also produces dose-related changes in regional CBF.73 These agents may produce EEG slowing but cannot completely
eliminate EEG activity. They cause little or no increase in ICP but do not blunt the reflex increase in ICP during direct
laryngoscopy.74 The benzodiazepine antagonist (flumazenil) can increase the ICP when used in large doses to reverse
midazolam action. CBF autoregulation and CO2 reactivity are well preserved with benzodiazepines.75 At low doses,
benzodiazepines cause no change in the rate of CSF formation, but at higher doses, they decrease CSF formation; the
resistance to resorption is either increased or there is no change.
Other Effects of Benzodiazepines
Low doses of midazolam are shown to preserve SCBF but higher doses can cause a decrease.76 Benzodiazepines
cross the BBB. They also have potent anticonvulsant effects. Prolonged benzodiazepine infusions can cause encephalopathy.77 Seizures can be precipitated by the administration of large doses of benzodiazapine antagonist flumazenil.
POCD can also be induced by benzodiazapines.78 The metabolism of benzodiazepine in neonates is very slow, and its
effect can persist up to 2 weeks after birth if consumed during pregnancy resulting in the “floppy infant syndrome”
characterized by hypotonia, CNS depression, and failure to suck.79
Current Status
They are used as supplemental drugs in neuroanaesthesia. Use of flumazenil to reverse benzodiazepine-induced
sedation must be done cautiously in patients with impaired intracranial compliance.
Recent Research
Although benzodiazepines are used to treat muscle spasms due to their muscle relaxant effects, there is no current
evidence to support their efficacy for treatment of muscle spasm, especially in patients suffering from rheumatoid
arthritis.80 Midazolam is commonly used as infusion to sedate children or neonates in the intensive care unit (ICU),
but again, at present there is insufficient data to promote this use of midazolam and further research is required to
evaluate the effectiveness of midazolam in neonates.81
Opioids
All opioids have variable effects on cerebral circulation and metabolism, which is attributed to the other anesthetics used in combination. With a vasodilating drug, opioids produce cerebral vasoconstriction, and with drug having
vasoconstriction properties, it produces vasodilation in cerebral circulation. In animal studies it has been found that
fentanyl and sufentanil along with a volatile anesthetic agent CBF and CMRO2.82 However, sufentanil with no volatile agent in background produces an increase in CBF.83 There are some reports that show that both fentanyl and sufentanil increase ICP in patients with severe head injury.84,85 According to Werner et al., under well-controlled mean
arterial blood pressure (MABP), sufentanil had significant effects on ICP in patients with head-injury, whereas with a
low MABP it produced only transient increases in ICP.86 In one study there was no difference in ICP-elevating effect
of fentanyl in patients with impaired and preserved autoregulation.87 Alfentanil and remifentanil produce little effect
on ICP and CBF and middle cerebral artery velocity.88–90 These two drugs are considered ideal for neuroanesthesia
and have shown satisfactory results in neurosurgical patients.91,92
Cerebral Effects of Opioids (Table 6.2)
Opioids have minimal effects on CBF and CMRO2 in low doses, while in higher doses, there is a progressive
reduction in both CBF and CMRO2. Like benzodiazepines, opioids also cause slowing of EEG activity but cannot
eliminate it. Opioids directly produce either minimal reduction or no change in the ICP but can markedly raise the
ICP secondary to respiratory depression causing hypercapnia. Large doses of opioids can cause hypotension and
decrease the CPP and hence, should be used cautiously in neurosurgical patients. The opioid antagonist (naloxone)
has minimal effects on CBF and ICP if given in titrated doses, but large doses of naloxone can cause intracranial
hemorrhage and arrhythmias. Cerebral autoregulation and CO2 reactivity are well preserved with all opioids. In low
doses, they cause no change in rate of CSF formation and decrease the resistance to resorption of CSF. Fentanyl, at
higher doses, decreases CSF formation with either an increase or no change in resistance to resorption, while alfentanil at higher doses causes no change in CSF formation and resistance to resorption. High doses of sufentanil cause
no change in CSF formation and either no change or increase in resistance to resorption.
III. NEUROPHARMACOLOGY
INHALATIONAL ANESTHETIC AGENTS
111
Other Effects of Opioids
All opioids cross the BBB. High doses of narcotics have been shown to produce seizures in laboratory animals
but rarely in humans. Normeperidine, a metabolite of meperidine, is a known convulsant. Opioids and associated
disturbances of calcium, sodium, and glucose homeostasis can cause POCD.93 Severe neonatal CNS depression is
reported after maternal consumption of opioids.94 They have minimal effects on the somatosensory EPs.
Current Status
As clinical doses of most opioids produce minimal effects on cerebral circulation and metabolism, these are used
widely in neurosurgical patients in combination with other anesthetics. However, caution is required when using
them in patients with raised or unstable ICP.
Recent Research
No significant difference in extubation time was observed between patients receiving remifentanil and sufentanil
by target control infusion during elective intracranial surgery.95 In comparison to continuous remifentanil infusion,
a single dose of dexmedetomidine (0.5 µg/kg) provided smooth emergence with hemodynamic stability in patients
undergoing cerebral aneurysm clipping.96
Dexmedetomidine
Dexmedetomidine is an agonist of α2-adrenergic receptors in certain parts of the brain and acts as an anxiolytic,
sedative, and analgesic. It provides sedation without causing risk of respiratory depression and is often used in the
ICU for providing light to moderate sedation to critically ill patients. Its use is associated with less delirium. Data on
the cerebral effects of dexmedetomidine are limited. It decreases the CBF, CMRO2, and ICP97,98 and can impair cerebral autoregulation, but it does not abolish CO2 reactivity.99 Dexmedetomidine has an important role during awake
craniotomy surgeries. Along with scalp block, dexmedetomidine provides an effective and safe anesthetic approach
in these surgeries.100 Dexmedetomidine during general anesthesia may effectively inhibit or reduce perioperative
stress responses in children with brain tumors.101 Both in vitro and in vivo studies have shown neuroprotective
effects of dexmedetomidine102; however, the mechanism of neuroprotection is not yet clear. It does not cause any
significant alteration in sensory or motor EPs.
Recent Research
A 2015 randomized controlled trial reports better controlled postoperative arterial pressures and superior analgesia with the use of dexmedetomidine during craniotomy.103 When used as an adjunct to total intravenous anesthesia,
dexmedetomidine does not seem to alter EPs and therefore can be safely used during surgeries requiring neurophysiological monitoring.104 Infusion of low-dose ketamine and dexmedetomidine both provide good postoperative
analgesia with minimal side effects. Both of the tested analgesic regimes can be used safely and effectively for postoperative pain relief in patients after spine surgery.105
INHALATIONAL ANESTHETIC AGENTS
The commonly used inhaled anesthetic agents in clinical practice are nitrous oxide and volatile liquids like desflurane, sevoflurane, and isoflurane.106 Halothane and enflurane are used less frequently.107 The inhalational anesthetic
agents have very narrow margin of safety, and each of them have a distinctive side effect. An ideal inhaled anesthetic
agent will produce rapid induction and rapid recovery from anesthesia. At higher concentrations, ICP may increase
due to increase in CBF secondary to loss of cerebral autoregulation. This may get limited by reduction in CPP due to
decrease in mean arterial pressure due to systemic vasodilation (Fig. 6.6). The physical properties of various inhaled
anesthetic agents are described in Table 6.3. Cost is an important factor in adopting these agents. Xenon, an inert gas
with anesthetic property, is hardly used because of its high cost.108
Desflurane
Desflurane (1,2,2,2-tetrafluoroethyl difluoromethyl ether) is a highly fluorinated inhalational anesthetic agent
(Fig. 6.7). Desflurane is a highly pungent inhalational agent with low potency. Because of its odor, desflurane can cause
III. NEUROPHARMACOLOGY
112
6. NEUROPHARMACOLOGY
1.5 MAC
Cerebral blood flow
(ml/100 gm/min)
1 MAC
0.5 MAC
Unanesthetized
0
50
150
Cerebral perfusion pressure (mm Hg)
FIGURE 6.6 Cerebral autoregulatory response attenuation by inhaled anesthetic agents. Cerebral blood flow relates to cerebral perfusion
pressure at >1.5 minimal alveolar concentration.
TABLE 6.3 Physical Properties of Inhalational Anesthetic Agents
Xenon
Desflurane
Sevoflurane
Isoflurane
Nitrous Oxide
Odor
Odorless
Pungent
Sweet
Pungent
Sweet
Minimum alveolar
concentration
71
6.6
1.8
1.2
104
Molecular weight
131.29
168
200
184
44
Blood–gas partition
coefficient
0.115
0.42
0.69
1.46
0.46
Soda lime stability
Yes
Yes
No
Yes
Yes
FIGURE 6.7 1,2,2,2-Tetrafluoroethyl difluoromethyl ether.
airway irritation leading to its infrequent use to induce anesthesia. Another drawback of desflurane anesthesia is
its high cost. The precise mechanisms of action by which volatile anesthetic agent produce unconsciousness has not
been clearly defined. These agents bind to membrane proteins and alter its function109 and potentiate the activity of
the inhibitory neurotransmitter GABA. The pharmacological characteristics of desflurane are described in Table 6.3.
Cerebral Effects of Desflurane (Table 6.2)
Desflurane causes increase in CBF resulting in increase in ICP in patients with decreased intracranial compliance
with normal end tidal carbon dioxide and blood pressure. At 1 minimal alveolar concentration (MAC), desflurane
reduces CMRO2 by half and CBF by 22%.110 Desflurane impairs the cerebral autoregulation. The cerebrovascular
reactivity to carbon dioxide is well maintained at desflurane concentrations between 0.5 and 1.5 MAC. Desflurane
causes no change in resistance to CSF resorption and it either increases or has no effects on CSF formation. Desflurane
(7%) in patients with midline shift causes increase in CSF pressure despite hypocapnia.111
Other Effects of Desflurane
The neuroprotective effect of desflurane is almost similar to that of isoflurane; however, because desflurane causes
increase in ICP to a greater extent than isoflurane or sevoflurane, it is used cautiously in patients with low intracranial compliance. Desflurane crosses the BBB but does not have epileptogenic property.
Current Status
Desflurane provides rapid onset and recovery of anesthesia and facilitates early neurological evaluation, hence is
useful for shorter neuroprocedures and not suitable in unstable ICP situations.
III. NEUROPHARMACOLOGY
INHALATIONAL ANESTHETIC AGENTS
113
Recent Research
According to a 2014 clinical trial, desflurane has greater hypnotic effect and produces low bispectral index than sevoflurane during general anesthesia.112 In patients who were undergoing elective supratentorial craniotomy, both sevoflurane and desflurane had similar intraoperative brain conditions, hemodynamics, and postoperative recovery profile.113
Sevoflurane
The chemical formula of sevoflurane is fluoromethyl 2,2,2-trifluoro-1-[trifluoromethyl]ethyl ether (Fig. 6.8).
Sevoflurane is one of the most widely used volatile anesthetic agents. It is commonly used in children because of
sweet odor and rapid induction and recovery of anesthesia. It acts on many receptors, causing activation of GABA
receptors and glycine receptors and inhibition of NMDA receptors, nicotinic acetylcholine (nACh) receptors and
5-hydroxytryptamine 3 receptors.114,115 The pharmacological characteristics of sevoflurane are described in Table 6.3.
Cerebral Effects of Sevoflurane (Table 6.2)
Sevoflurane causes increase in CBF, ICP, and decrease in CMRO2. The rise in ICP is less than isoflurane and desflurane and can be blocked by hyperventilation. It maintains cerebral autoregulation and the CO2 reactivity is well preserved. Sevoflurane decreases the rate of CSF formation and increases the resistance to CSF resorption. It is observed
that in patients undergoing pituitary surgery through the transsphenoidal approach show minimal increase in lumbar CSF pressure comparable to desflurane and isoflurane.116
Other Effects of Sevoflurane
There is evidence that sevoflurane can also induce in vitro preconditioning due to alteration of biochemical pathways
before an ischemic insult.117 Due to the supposed neuroprotective effects of sevoflurane in animal experiments, it may
be prudent to choose sevoflurane for anesthesia, although its clinical benefits are yet unestablished. Some investigators
have reported epileptogenic activity with sevoflurane anesthesia mostly during the induction phase.118 It is biodegradable and can produce toxic metabolites, which can cause renal toxicity, although toxicity is not yet reported in humans.
Current Status
Used widely for neuroanesthesia in small children. Less preferred than propofol in patients requiring tight control of ICP.
Prolonged use of sevoflurane should be avoided, especially in low-flow circuits and in patients with prior renal disease.
Recent Research
According to the Cochrane systemic review, high initial concentration sevoflurane technique offers rapid induction of anesthesia but can cause apnea.119 Cerebral cortical oxygenation measured by near-infrared spectroscopy is
better preserved with sevoflurane than with propofol.120
Isoflurane
Isoflurane is a halogenated volatile agent, 2-chloro-2-(difluoromethoxy)-1,1,1-trifluoro-ethane (Fig. 6.9). Its pungent odor can irritate the airway leading to laryngospasm, and so it is not considered for induction of anesthesia in
pediatric patients. Isoflurane binds to GABA and glycine receptors and enhances the activity of glycine receptors
FIGURE 6.8 Fluoromethyl 2,2,2-trifluoro-1-[trifluoromethyl]ethyl ether.
FIGURE 6.9 2-Chloro-2-(difluoromethoxy)-1,1,1-trifluoroethane.
III. NEUROPHARMACOLOGY
114
6. NEUROPHARMACOLOGY
resulting in decreased motor function. It also has inhibitory action on NMDA receptors and potassium channels. The
pharmacological characteristics of isoflurane are described in Table 6.3.
Cerebral Effects of Isoflurane (Table 6.2)
Isoflurane causes cerebral vasodilation leading to increase in CBF and ICP and markedly reduces the CMRO2. However,
at MAC values below 1, isoflurane produces no changes in CBF.121 Increases in ICP are mild and can be reduced satisfactorily by hyperventilation.122 Cerebral autoregulation is well maintained at low (0.5%) and high concentrations, with
moderate hypocapnia.123 At low or high concentrations, isoflurane causes no change in rate of CSF formation and there is
no change (increase or decrease) in resistance to resorption of CSF. Isoflurane produces an increase in SCBF and an attenuation of autoregulation at 1 and 2 MAC. The changes seen at 2 MAC are greater for the spinal cord than for the cortex.
Other Effects of Isoflurane
There is evidence that isoflurane improves recovery from cerebral ischemia by preconditioning of neurons in male
mice.124 Isoflurane crosses the BBB but does not produce epileptogenic activity.
Current Status
Although the cerebral protective effects of isoflurane (mainly due to pronounced decreases in cerebral metabolism) are not proven clinically, it may be a useful anesthetic for neurosurgeries where the threat of cerebral ischemia
is large like carotid endarterectomy. Propofol is preferred over isoflurane in patients with raised ICP.
Recent Research
A 2015 systematic review supports the use of isoflurane in treatment of status epilepticus to produce burst suppression.125 It can also be used to sedate the patients with stroke and trauma in the neuro-ICU. Isoflurane decreases
the occurrence of cortical spreading depolarization, which is a major mechanism of delayed brain injury in stroke
and brain trauma.126
Nitrous Oxide
Nitrous oxide (N2O) is also known as laughing gas because of its euphoric effect. Its chemical formula is “dinitrogen monoxide” (Fig. 6.10). Due to its poor blood solubility, alveolar and brain concentrations are achieved very
rapidly (Table 6.3). It also has analgesic and anesthetic effects. Its mechanism of action includes partial blockade
of NMDA receptors, nAch receptors, GABA receptors, and histamine (5-HT3) receptors and partial potentiation of
GABA and glycine receptors (http://en.wikipedia.org/wiki/Nitrous_oxide).127,128 The pharmacological characteristics of N2O are described in Table 6.3.
Cerebral Effects of N2O (Table 6.2)
N2O is no longer considered an inert gas as it has significant cerebral effects. When administered alone or with
minimal background inhalational anesthesia it causes increases in CBF. However, when administered with certain
intravenous anesthetics, its effects on CBF may be reduced. The CBF is higher with 1 MAC combination of 0.5 MAC
volatile agent and 0.5 MAC N2O, compared to 1 MAC of volatile agent alone. N2O may increase or produce no
change in CMRO2. N2O may cause increased ICP in patients with mass lesions. This increase in ICP can be reduced
by intravenous drugs like barbiturates or propofol. N2O can impair cerebral autoregulation but preserves CO2 reactivity and has no effect on the rate of CSF formation and resistance to CSF resorption. N2O increases the spinal cord’s
utilization of glucose similar to the effect in the brain (approximately 25%). Both neurotoxic and neuroprotective
properties have been demonstrated with N2O use.
Other Effects of N2O
N2O diffuses rapidly into closed air-filled spaces and expands. In case of venous air embolism, N2O can increase
the size of the air bubble and worsen the consequences of air embolism. N2O crosses the BBB and can produce epileptogenic activity.
FIGURE 6.10 Di nitrogen monoxide.
III. NEUROPHARMACOLOGY
NEUROMUSCULAR BLOCKING AGENTS
115
Current Status
Use of N2O in neuroanesthesia has generated a lot of debate. Due to its ICP-raising effects and potential to enlarge
air spaces, and a higher incidence of nausea and vomiting associated with its use, N2O has been discontinued in
neurosurgical practice at many centers.
Recent Research
There is very little evidence that N2O as an inhalation agent is also effective. Furthermore, well-designed randomized controlled trials to evaluate this effect of N2O are needed.129 Avoiding N2O during craniotomies in neurosurgical
patients will not affect the overall outcome.130
Xenon
Xenon is a colorless, dense, and odorless gas. Currently, its use as a general anesthetic agent is limited because of
its high cost and scarcity. However, it proposes many advantageous properties for its use in neuroanesthesia. It has
been shown to have neuroprotective properties through its antagonist action on NMDA receptors, has exceptional
cardiovascular stability, and has a rapid induction and rapid recovery from anesthesia due to its low blood gas
coefficient.
Cerebral Effects of Xenon (Table 6.2)
Xenon decreases CBF leading to decreases in ICP and CMRO2 more than the CBF.131,132Cerebrovascular autoregulation and CO2 reactivity are well maintained with xenon at 1 MAC concentration.133 Its effect on rate of CSF production and resistance to resorption is unknown.
Other Effects of Xenon
Xenon crosses the BBB and does not induce seizures. Subanesthetic levels of xenon may have an anticonvulsant
effect. Inhaled xenon may be a valuable new therapy in neonatal asphyxial seizures.134 It does not cause POCD or
delirium in elderly patients.135
Current Status
Due to its low blood/gas partition coefficient, xenon allows rapid anesthetic recovery and may be useful in neuroanesthesia. Its potential for causing cerebral neuroprotection is also attractive. However, the unclear clinical benefits
and high cost preclude widespread use of xenon in neurosurgical practice.
NEUROMUSCULAR BLOCKING AGENTS
The depolarizing agents mimic the action of acetylcholine, while the nondepolarizing drugs interfere with the
action of acetylcholine. These drugs do not cross the BBB and hence do not cause any CNS side effects.
Depolarizing Neuromuscular Blocking Agents
Succinylcholine (SCh) produces a very rapid paralysis and recovery from muscle relaxation, and makes it
a muscle relaxant of choice for rapid sequence induction in emergency situations.136 It can release potassium
ions from the cells resulting in increase in serum potassium levels by 0.5 mEq/L, and hence should be used cautiously in small children and posttrauma patients with electrolyte imbalance. The cerebral effects of SCh (Table
6.2) mainly include increases in ICP from muscle fasciculations, increased EEG activity, and increases in CBF.137
The rise in ICP can be blunted by prior administration of nondepolarizing neuromuscular blocking agents.138
Cerebral autoregulation and CO2 reactivity is preserved, and it causes no change in CSF dynamics. SCh does
not cross the BBB and does not induce seizures. A recent study has found magnesium to be effective in preventing SCh-induced fasciculations.139 Succinylcholine should be used cautiously with propranolol, a nonselective
β-blocker that also affects the intracellular redistribution of potassium resulting in hyperkalemia.140 Use of
SCh has declined in neuroanesthesia except in emergency situations and in patients with anticipated difficult
intubation.
III. NEUROPHARMACOLOGY
116
6. NEUROPHARMACOLOGY
Nondepolarizing Neuromuscular Blocking Agents
Nondepolarizing muscle relaxants combine with nAch receptors at postsynaptic junction and antagonize the
action of acetylcholine.141,142 They have minimal or no effects on CBF, CMRO2, and ICP. However, large doses of
pancuronium can cause rapid increases in blood pressure that could adversely affect the ICP. Release of histamine
with high doses of atracurium can cause increases in ICP. Cerebral autoregulation and CO2 reactivity is well preserved with nondepolarizing muscle relaxants. These drugs do not alter the CSF dynamics. There is no change in
rate of CSF formation or resistance to resorption. They are also unable to cross the BBB and do not induce seizurelike
activity, although laudanosine, a metabolite of atracurium, can cause seizures but is produced in low levels only.
Cis-atracurium produces less laudanosine and histamine than atracurium. Rocuronium is also a preferred drug for
rapid sequence induction due to its rapid onset of action. Spine surgery under general anesthesia without muscle
relaxants provides adequate working conditions, earlier eye opening, earlier tracheal extubation, and higher levels
of consciousness on emergence.143
LOCAL ANESTHETIC AGENTS
Lidocaine is considered as the standard local anesthetic agent. It acts mainly through blockade of sodium ion
channel.144 Local anesthetics produce a dose-related fall in CBF and CMRO2. Lidocaine in large doses cause 30%
decrease in CMRO2 in dogs. These drugs also decrease the ICP following fall in CBF. Lignocaine 1.5 mg/kg given
intravenously is useful in preventing ICP fluctuations during endotracheal intubation, suctioning, and skin incision.
Cerebral autoregulation and CO2 reactivity is maintained with local anesthetics, and the rate of CSF formation is
reduced. Lidocaine causes sedation at low concentrations, while it may result in seizures at higher doses. Although
neuroprotective properties have been demonstrated with lidocaine, it has not been seen in severe ischemic conditions. A clinical trial reported long-term improvement in neuropsychological condition with lidocaine infusion during cardiac surgery,145 but examination of its role in cerebral ischemia needs further investigation. Present evidence
does not support the use of local anesthetic sympathetic blockade for complex regional pain syndrome as it is not
found effective in reducing pain.146 There is also no supportive evidence available to prove the role of local anesthetics in treating meralgia paraesthetica.147
MISCELLANEOUS DRUGS
Description of pharmacology of various other drugs used in the practice of neuroanesthesia is beyond the scope
of this chapter and is incorporated in different chapters focusing on their clinical use. However, the cerebrovascular
effects of the commonly used vasoactive drugs are tabulated here (Table 6.4).
FUTURE DIRECTIONS IN NEUROPHARMACOLOGY
There is a need for measurement of continuous blood levels of intravenous anesthetics. Although online monitoring of propofol levels has been tried, its common use is not supported by enough evidence.148 Future pharmacologic
work also needs to be directed toward development of methods for achieving faster, reliable emergence after neuroanesthesia, pharmacologic therapies specific for patients with spinal cord injury, and development of anesthesia
robots that would assist in administering anesthetic drugs based on the effects of drugs like hypnosis, analgesia,
neuromuscular blockade, etc.149
CONCLUSION
The main emphasis in neuroanesthesia in patients undergoing neurosurgery and those with cerebrovascular disease is to attenuate further brain tissue damage, prevent new damage, and provide a good surgical field to the neurosurgeon. An accurate pharmacologic manipulation is necessary to help achieve a good neurologic outcome and
patient safety in patients with brain injury.
III. NEUROPHARMACOLOGY
III. NEUROPHARMACOLOGY
+
?
+
+
++
+
0
+
−
?
?
?
0
0/−
Nimodipine
Nicardipine
Milrinone
Dobutamine
Dopamine
Epinephrine
Norepinephrine
Ephedrine
Phenylephrine
Vasopressin
Nitroglycerine
Sodium nitroprusside
Labetalol
Esmolol
?
?
?
?
+
+
?
+
+
?
+/0/−
?
?
+
CPP
?
?
+
+
?
?
?
0
0
+
?
?
?
−
ICP
?
?
?
−
?
?
0
0
0
?
0
?
–
+
Cerebral Autoregulation
0
0
?
?
?
?
0
?
?
?
0
0
−
−
CO2 Reactivity
CBF, cerebral blood flow; CPP, cerebral perfusion pressure; ICP, intracranial pressure; + = increase or improve; − = decrease; 0 = no effect, ? = not known.
CBF
Drugs
TABLE 6.4 Cerebral Effects of Miscellaneous Drugs
−/0
−/0
−/0
−/0
+/0
+/−
+
++
+++
+/−
++
0
0
0
Cerebral Oxygen
Delivery
0
−
—
—
+++
++
+
+++
+++
+
−
–
–
—
Vasomotor Tone
CONCLUSION
117
118
6. NEUROPHARMACOLOGY
References
1. Garrett ER, Bojarski JT, Yakatan GJ. Kinetics of hydrolysis of barbituric acid derivatives. J Pharm Sci 2006;60:1145–54.
2. Li X, Pearce RA. Effects of halothane on GABA (A) receptor kinetics: evidence for slowed agonist unbinding. J Neurosci 2000;20:899–907.
3. Clarke RS, Dundee JW, Barron DW, McArdle L. Clinical studies of induction agents. XVXVI. The relative potencies of thiopentone, methohexitone and propanidid. Br J Anesth 1968;40:593–601.
4. Burch PG, Stanski DR. The role of metabolism and protein binding in thiopental anesthesia. Anesthesiology 1983;58:146–52.
5. Hung OR, Varvel JR, Shafer SL, Stanski DR. Thiopental pharmacodynamics. II. Quantitation of clinical and electroencephalographic depth
of anesthesia. Anesthesiology 1992;77:237–44.
6. McMurry TJ, Robinson FP, Dundee JW, Riddle JG, McClean E. A method for producing constant plasma concentration of drugs. Application
to methohexitone. Br J Anaesth 1986;58:1085–90.
7. Young WL, Prohovnik I, Correll JW, Ostapkovich N, Ornstein E. Thiopental effect on cerebral blood flow during carotid endarterectomy.
J Neurosurg Anesthesiol 1991;3:265–9.
8. Stullken Jr EH, Milde JH, Michenfelder JD, Tinker JH. The nonlinear response of cerebral metabolism to low concentration of halothane,
enflurane, isoflurane, and thiopental. Anesthesiology 1977;46:28–34.
9. Shapiro HM, Galindo A, Wyte SR, Haris AB. Rapid intra-operative reduction of intracranial pressure with thiopentone. Br J Anaesth
1973;45:1057–62.
10. Hickey R, Albin MS, Bunegin L, Gelineau AJ. Autoregulation of spinal cord blood flow: is the cord a microcosm of the brain? Stroke 1986;17:6.
11. Hitchon PW, Kassell NF, Hill TR, Gerk MK, Sokoll MD. The response of spinal cord blood flow to high-dose barbiturates. Spine 1982;7:41–5.
12. Crosby G, Crane AM, Sokoloff L. A comparison of local rates of glucose utilization in spinal cord and brain in conscious and nitrous oxide- or
pentobarbital-treated rats. Anesthesiology 1984;61:434–8.
13. Gumpert J, Paul R. Activation of the electroencephalogram with intravenous Brietal (methohexitone): the findings in 100 cases. J Neurol
Neurosurg Psychiatry 1971;34:646–8.
14. Bauer J, Hageman I, Dam H, Báez A, Bolwig T, Roed J, Olsen NV, Jørgensen MB. Comparison of propofol and thiopental as anesthetic agents
for electroconvulsive therapy: a randomized, blinded comparison of seizure duration, stimulus charge, clinical effect, and cognitive side
effects. J ECT 2009;25:85–90.
15. Reeta KH, Mehla J, Gupta YK. Curcumin ameliorates cognitive dysfunction and oxidative damage in phenobarbitone and carbamazepine
administered rats. Eur J Pharmacol 2010;644:106–12.
16. Olney JW, Ishimaru MJ, Bittigau P, Ikonomidou C. Ethanol-induced apoptotic neurodegeneration in the developing brain. Apoptosis
2000;5:515–21.
17. Fredriksson A, Pontén E, Gordh T, Eriksson P. Neonatal exposure to a combination of N-methyl-d-aspartate and gamma-aminobutyric acid
type A receptor anesthetic agents potentiates apoptotic neurodegeneration and persistent behavioral deficits. Anesthesiology 2007;107:427–36.
18. Roberts I, Syndenham E. Barbiturates for acute traumatic brain injury (review). Cochrane Libr 2012;(12):CD000033.
19. Candy B, Jackson KC, Jones L, Tookman A, King M. Drug therapy for symptoms associated with anxiety in adult palliative care patients
(review). Cochrane Libr 2012;(10):CD004596.
20. Euliano TY, Gravenstein JS. A brief pharmacology related to anesthesia. Essential anesthesia: from science to practice. Cambridge (UK): Cambridge
University Press; 2009. p. 173.
21. Trapani G, Latrofa A, Franco M, Altomare C, Sanna E, Usala M, Biggio G, Liso G. Propofol analogues. Synthesis, relationships between structure and affinity at GABAA receptor in rat brain, and differential electrophysiological profile at recombinant human GABAA receptors. J Med
Chem 1998;41:1846–54.
22. Krasowski MD, Jenkins A, Flood P, Kung AY, Hopfinger AJ, Harrison NL. General anesthetic potencies of a series of propofol analogs correlate with potency for potentiation of gamma-aminobutyric acid (GABA) current at the GABA(A) receptor but not with lipid solubility.
J Pharmacol Exp Ther 2001;297:338–51.
23. Haeseler G, Leuwer M. High-affinity block of voltage-operated rat IIA neuronal sodium channels by 2,6 di-tert-butylphenol, a propofol
analogue. Eur J Anaesthesiol 2003;20:220–4.
24. Haeseler G, Karst M, Foadi N, Gudehus S, Roeder A, Hecker H, Dengler R, Leuwer M. High-affinity blockade of voltage-operated skeletal
muscle and neuronal sodium channels by halogenated propofol analogues. Br J Pharmacol 2008;155:265–75.
25. Fowler CJ. Possible involvement of the endocannabinoid system in the actions of three clinically used drugs. Trends Pharmacol Sci 2004;25:59–61.
26. Langley MS, Propofol HRC. A review of its pharmacodynamic and pharmacokinetic properties and use as an intravenous anaesthetic. Drugs
1988;35:334–72.
27. Shafer A, Doze VA, Shafer SL, White PF. Pharmacokinetics and pharmacodynamics of propofol infusions during genral anesthesia.
Anesthesiology 1988;69:348–56.
28. Kirkpatrick T, Cockshot ID, Douglas EJ, Nimmo WS. Pharmacokinetics of propofol (diprivan) in elderly patients. Br J Anaesth 1988;60:146–50.
29. Petersen KD, Landsfeldt U, Cold GE, et al. Intracranial pressure and cerebral hemodynamic in patients with cerebral tumor: a randomized prospective study of patients subjected to craniotomy in propofol-fentanyl, isoflurane-fentanyl, or sevoflurane-fentanyl anesthesia.
Anesthesiology 2003;98:329–36.
30. Lagerkranser M, Stånge K, Sollevi A. Effects of propofol on cerebral blood flow, metabolism, and cerebral autoregulation in the anesthetized
pig. J Neurosurg Anesthesiol 1997;9:188–93.
31. Artru AA. Propofol combined with halothane or with fentanyl/halothane does not alter the rate of CSF formation or resistance to reabsorption of CSF in rabbits. J Neurosurg Anesthesiol 1993;5:250–7.
32. Werner C, Hoffman WE, Kochs E, Esch JSam, Albrecht RF. The effects of propofol on cerebral and spinal cord blood flow in rats. Anesth Analg
1993;76:971–5.
33. Cavazzuti M, Porro CA, Barbieri A, Galetti A. Brain and spinal cord metabolic activity during propofol anaesthesia. Br J Anaesth 1991;66:490–5.
34. Modica PA, Tempelhoff R, White PF. Pro and anticonvulsant effects of anesthetics. Anesth Analg 1990;70:433–44.
35. Dressler I, Fritzsche I, Spies C, Cortina K, Rundshagen I. Psychomotor dysfunction after remifentanil/propofol anaesthesia. Eur J Anaesthesiol
2007;24:347–54.
III. NEUROPHARMACOLOGY
REFERENCES
119
36. Rasmussen LS, Schmehl W, Jakobsson J. Comparison of xenon with propofol for supplementary general anaesthesia for knee replacement: a
randomized study. Br J Anaesth 2006;97:154–9.
37. Coburn M, Baumert HJ, Roertgen D, et al. Emergence and early cognitive function in the elderly after xenon or desflurane anaesthesia: a
double-blinded randomized controlled trial. Br J Anaesth 2007;98:756–62.
38. Hoecker J, Stapelfeldt C, Leiendecker J, et al. Postoperative neurocognitive dysfunction in elderly patients after xenon versus propofol anesthesia for major noncardiac surgery. Anesthesiology 2009;110:1068–76.
39. Cremer J, Stoppe C, Fahlenkamp AV, et al. Early cognitive function, recovery and well-being after sevoflurane and xenon anaesthesia in the
elderly: a double-blinded randomized controlled trial. Med Gas Res 2011;1:9.
40. Creeley C, Dikranian K, Dissen G, Martin L, Olney J, Brambrink A. Propofol-induced apoptosis of neurones and oligodendrocytes in fetal
and neonatal rhesus macaque brain. Br J Anaesth 2013;110:29–38.
41. Kikuta K, Takagi Y, Nozaki K, Yamada K, Miyamoto S, Kataoka H, Arai T, Hashimoto N. Effects of intravenous anesthesia with propofol on
regional cortical blood flow and intracranial pressure in surgery for moyamoya disease. Surg Neurol 2007;68:421–4.
42. Shah PS, Shah VS. Propofol for procedural sedation/anaesthesia in neonates (review). Cochrane Database Syst Rev 2011;(3):CD007248.
43. Prabhakar H, Kalaivani M. Propofol versus thiopental sodium for the treatment of refractory status epilepticus. Cochrane Database Syst Rev
2015;(6):CD009202.
44. Vinson DR, Bradbury DR. Etomidate for procedural sedation in emergency medicine. Ann Emerg Med 2002;39:592–8.
45. Hohl CM, Kelly-Smith CH, Yeug TC, Sweet DD, Doyle-Waters MM, Schulzer M. The effect of a bolus dose of etomidate on cortisol levels,
mortality, and health services utilization: a systematic review. Ann Emerg Med 2010;56:105–13.
46. Vanlersberghe C, Camu F. Etomidate and other non-barbiturates. Handb Exp Pharmacol 2008;182:267–82. http://en.wikipedia.org/wiki/
Digital_object_identifier.
47. Gooding JM, Crossen G. Etomidate: an ultra-short acting nonbarbiturate agent for anesthesia induction. Anesth Analg 1976;55:286–9.
48. Heykant JJ, Meuldermans WE, Michiels LJ, Lewi PJ, Jansen PA. Distribution, metabolism and excretion of etomidate, a short-acting hypnotic
drug, in the rat. Comparative study of (R)-(+)-(−)-Etomidate. Arch Int Pharmacodyn Ther 1975;216:113–29. http://en.wikipedia.org/wiki/
Digital_object_identifier.
49. Doenicke A. Etomidate, a new intravenous hypnotic. Acta Anaesthesiol Belg 1974;25:307–15.
50. Meuldermans WE, Heykants JJ. The plasma protein binding and distribution of etomidate in dog, rat and human blood. Arch Int Pharmacodyn
Ther 1976;221:150–62.
51. Fragen RJ, Avram MJ, Henthorn TK, Caldwell NJ. A pharmacokinetically designed etomidate infusion regimn for hypnosis. Anesth Analg
1983;62:654–60.
52. Modica PA, Tempelhoff R. Intracranial pressure during induction of anesthesia and tracheal intubation with etomidate induced EEG burst
suppression. Can J Anaesth 1992;39:236–41.
53. Milde LN, Milde JH, Michenfelder JD. Cerebral functional metabolic and hemodynamic effect of etomidate in dogs. Anesthesiology
1985;63:371–7.
54. Bramwell KJ, Haizlip J, Pribble C, VanDerHeyden TC, Witte M. The effect of etomidate on intracranial pressure and systemic blood pressure
in pediatric patients with severe traumatic brain injury. Pediatr Emerg Care 2006;22:90–3.
55. Artru AA. Dose-related changes in the rate of cerebrospinal fluid formation and resistance to reabsorption of cerebrospinal fluid following
administration of thiopental, midazolam, and etomidate in dogs. Anesthesiology 1988;69:541–6.
56. Thompson 2nd TL, Moran MG. Nies AS–Psychotropic drug use in the elderly. N Engl J Med 1983;308:194–9.
57. Van de Wiele B, Rubinstein E, Peacock W, Martin N. Propylene glycol toxicity caused by prolonged infusion of etomidate. J Neurosurg
Anesthesiol 1995;7:259–62.
58. Ghaly RF, Lee JJ, Ham JH, Stone JL, George S, Raccforte P. Etomidate dose-response on somatosensory and transcranial magnetic induced
spinal motor evoked potentials in primates. Neurol Res 1999;21:714–20.
59. Ubags LH, Kalkman CJ, Been HD, Porsius M, Drummond JC. The use of ketamine or etomidate to supplement sufentanil/N2O anesthesia
does not disrupt monitoring of myogenic transcranial motor evoked responses. J Neurosurg Anesthesiol 1997;9:228–33.
60. Kaushal RP, Vatal A, Pathak R. Effect of etomidate and propofol induction on hemodynamic and endocrine response in patients undergoing coronary artery bypass grafting/mitral valve and aortic valve replacement surgery on cardiopulmonary bypass. Ann Card Anaesth 2015;18:172–8.
61. Lyon RM, Perkins ZB, Chatterjee D, Lockey DJ, Russell MQ, et al. Significant modification of traditional rapid sequence induction improves
safety and effectiveness of pre-hospital trauma anaesthesia. Crit Care 2015;19:134.
62. Kohrs R, Durieux ME. Ketamine: teachoing on old drug new tricks. Anesth Analg 1998;87:1186–93.
63. Hirota K, lambert DG. Ketamine: its mechanism(s) of action and unusual clinical use. Br J Anaesth 1996;77:441–4.
64. Takeshita H, Okuda Y, Sari A. The effects of ketamine on cerebral circulation and metabolism in man. Anesthesiology 1972;36:69–75.
65. Mayberg TS, Lam AM, Matta BF, et al. Ketamine does not increase cerebral blood flow velocity or intracranial pressure during isoflurane/
nitrous oxide anesthesia in patient undergoing anesthesia. Anesth Analg 1995;81:84–9.
66. Bar-Joseph G, Guilburd Y, Tamir A, Guilburd JN. Effectiveness of ketamine in decreasing intracranial pressure in children with intracranial
hypertension. J Neurosurg Pediatr 2009;4:40–6.
67. Himmelseher S, Durieux ME. Revising a dogma: ketamine for patients with neurological injury? Anesth Analg 2005;101:524–34.
68. Yu QJ, Zhou QS, Huang HB, Wang YL, Tian SF, Duan DM. Protective effect of ketamine on ischemic spinal cord injury in rabbits. Ann Vasc
Surg 2008;22:432–9.
69. Sussman DR. A comparative evaluation of ketamine anesthesia in children and adults. Anesthesiology 1974;40:459–64.
70. Schubert A, Licine MG, Lineberry PJ. The effect of ketamine on human somatosensory evoked potentials and its modification by nitrous
oxide. Anesthesiology 1990;72:33–9.
71. Schreiberova J, Hess L, Krajina A, Lojik M. Dexmedetomidine-ketamine-midazolam combination for sedation in endovascular treatment of
cerebral arterio-venousmalformations and carotid artery stenosis. Ceska a Slovenska Neurologie a Neurochirurgie 2008;71:446.
72. Page C, Michael C, Sutter M, Walker M, Hoffman BB. Integrated pharmacology. 2nd ed. 2002.
73. Veselis RA, Reinsel RA, Beattie BJ, et al. Midazolam changes regional cerebral blood flow in discrete brain regions: an H215O positron tomography study. Anesthesiology 1997;87:1106–17.
III. NEUROPHARMACOLOGY
120
6. NEUROPHARMACOLOGY
74. Giffin JP, Cottrell JE, Shwiry B, et al. Intrcranial pressure, mean arterial pressure, and heart rate following midazolam or thiopental in
humans with brain tumors. Anesthesiology 1984;60:491–4.
75. Foster A, Juge O, Morel D. Effects of midazolam on cerebral hemodynamics and cerebral vasomotor responsiveness to carbon dioxide. J Cereb
Blood Flow Metab 1983;3:246–9.
76. Nishiyama T. Spinal cord blood flow change by intravenous midazolam during isoflurane anesthesia. Anesth Analg 2005;101:242–5.
77. Bergman I, Steeves M, Burckart G, et al. Reversible neurologic abnormalities associated with prolonged intravenous midazolam and fentanyl
administration. J Pediatr 1991;119:644–9.
78. Richardson JS, Miller PS, Lemay JS, et al. Mental dysfunction and the blockade of muscarinic receptors in the brains of the normal elderly.
Prog Neuropsychopharmacol Biol Psychiatry 1985;9:651–4.
79. Laegreid L, Olegård R, Conradi N, Hagberg G, Wahlstrom J, Abrahamsson L. Association between congenital malformations and maternal
consumption of benzodiazepines. A case-control study. Deu Med Child Neurol 1990;32:432–41.
80. Richards BL, Whittle SL, Buchbinder R. Muscle relaxants for pain management in rheumatoid arthritis (review). Cochrane Database Syst Rev
2012;(1):CD008922.
81. Ng E, Taddio A, Ohlsson A. Intravenous midazolam infusion for sedation of infants in the neonatal intensive care unit (review). Cochrane
Database Syst Rev 2012;(6):CD002052.
82. Werner C, Hoffman WE, Baughman VL, Albrecht RF, Schulte J. Effects of sufentanil on cerebral blood flow, cerebral blood flow velocity, and
metabolism in dogs. Anesth Analg 1991;72:177–81.
83. Milde LN, Milde JH, Gallagher WJ. Effects of sufentanil on cerebral circulation and metabolism in dogs. Anesth Analg 1990;70:138–46.
84. Albanese J, Durbec O, Viviand X, Potie F, Alliez B, Martin C. Sufentanil increases intracranial pressure in patients with head trauma.
Anesthesiology 1993;79:493–7.
85. Sperry RJ, Bailey PL, Reichman MV, Peterson JC, Petersen PB. Pace NLFentanyl and sufentanil increase intracranial pressure in head trauma
patients. Anesthesiology 1992;77:416–20.
86. Werner C, Kochs E, Bause H, Hoffman WE, Schulte am Esch J. Effects of sufentanil on cerebral hemodynamics and intracranial pressure in
patients with brain injury. Anesthesiology 1995;83:721–6.
87. de Nadal M, Munar F, Poca MA, Sahuquillo J, Garnacho A, Rosselló J. Cerebral hemodynamic effects of morphine and fentanyl in patients
with severe head injury: absence of correlation to cerebral autoregulation. Anesthesiology 2000;92:11–9.
88. Souter MJ, Andrews PJ, Piper IR, Miller JD. Effects of alfentanil on cerebral haemodynamics in an experimental model of traumatic brain
injury. Br J Anaesth 1997;79:97–102.
89. Mayberg TS, Lam AM, Eng CC, Laohaprasit V, Winn HR. The effect of alfentanil on cerebral blood flow velocity and intracranial pressure
during isoflurane-nitrous oxide anesthesia in humans. Anesthesiology 1993;78:288–94.
90. Warner DS, Hindman BJ, Todd MM, Sawin PD, Kirchner J, Roland CL, Jamerson BD. Intracranial pressure and hemodynamic effects of remifentanil versus alfentanil in patients undergoing supratentorial craniotomy. Anesth Analg 1996;83:348–53.
91. From RP, Warner DS, Todd MM, Sokoll MD. Anesthesia for craniotomy: a double-blind comparison of alfentanil, fentanyl, and sufentanil.
Anesthesiology 1990;73:896–904.
92. Ostapkovich ND, Baker KZ, Fogarty-Mack P, Sisti MB, Young WL. Cerebral blood flow and CO2 reactivity is similar during remifentanil/
N2O and fentanyl/N2O anesthesia. Anesthesiology 1998;89:358–63.
93. Fines DP, Severn AM. Anaesthesia and cognitive disturbance in the elderly. Contin Educ Anaesth Crit Care Pain 2006;6:37–40.
94. Madadi P, Ross CJ, Hayden MR, Carleton BC, Gaedigk A, Leeder JS, Koren G. Pharmacogenetics of neonatal opioid toxicity following maternal use of codeine during breastfeeding: a case-control study. Clin Pharmacol Ther 2009;85:31–5.
95. Djian MC, Blanchet B, Pesce F, Sermet A, Disdet M, Vazquez V, Gury C, Roux FX, Raggueneau JL, Coste J, Joly LM. Comparison of the time
to extubation after use of remifentanil or sufentanil in combination with propofol as anesthesia in adults undergoing nonemergency intracranial surgery: a prospective, randomized, double-blind trial. Clin Thera 2006;28:560–8.
96. Kim H, Choi SH, Min KT, Byon HJ. Randomized trial to compare the effect of a single dose of dexmedetomidine and continuous infusion
of remifentanil on airway reflex and hemodynamic response during emergence in patients undergoing cerebral aneurysm clipping. Eur J
Anaesthesiol 2012;29:112–3.
97. Drummond JC, Dao AV, Roth DM, Cheng CR, Atwater BI, Minokadeh A, Pasco LC, Patel PM. Effect of dexmedetomidine on cerebral blood
flow velocity, cerebral metabolic rate, and carbon dioxide response in normal humans. Anesthesiology 2008;108:225–32.
98. Prielipp RC, Wall MH, Tobin JR, Groban L, Cannon MA, Fahey FH, Gage HD, Stump DA, James RL, Bennett J, Butterworth J. Dexmedetomidineinduced sedation in volunteers decreases regional and global cerebral blood flow. Anesth Analg 2002;95:1052–9.
99. Ogawa Y, Iwasaki K, Aoki K, et al. Dexmedatomidine weakens dynamic cerebral autoregulation as assessed by transfer function analysis and
the thigh cuff method. Anesthesiology 2008;109:642–50.
100. Garavaglia MM, Das S, Cusimano MD, Crescini C, Mazer CD, Hare GM, Rigamonti A. Anesthetic approach to high-risk patients and prolonged awake craniotomy using dexmedetomidine and scalp block. J Neurosurg Anesthesiol 2014;26:226–33.
101. Wu L, Lv H, Luo W, Jin S, Hang Y. Effects of dexmedetomidine on cellular immunity of perioperative period in children with brain neoplasms. Int J Clin Exp Med 2015;8:2748–53.
102. Ma D, Hossain M, Rajakumaraswamy N, Arshad M, Sanders RD, Franks NP, Maze M. Dexmedetomidine produces its neuroprotective effect
via the alpha 2A-adrenoceptor subtype. Eur J Pharmacol 2004;502:87–97.
103. Rajan S, Hutcherson MT, Sessler DI, Kurz A, Yang D, Ghobrial M, Liu J, Avitsian R. The effects of dexmedetomidine and remifentanil on
hemodynamic stability and analgesic requirement after craniotomy: a randomized controlled trial. J Neurosurg Anesthesiol 2016;28:282–90.
104. Rozet I, Metzner J, Brown M, Treggiari MM, Slimp JC, Kinney G, Sharma D, Lee LA, Vavilala MS. Dexmedetomidine does not affect evoked
potentials during spine surgery. Anesth Analg 2015;121:492–501.
105. Garg N, Panda NB, Gandhi KA, Bhagat H, Batra YK, Grover VK, Chhabra R. Comparison of small dose ketamine and dexmedetomidine infusion for postoperative analgesia in spinesurgery-a prospective randomized double-blind placebo controlled study. J Neurosurg
Anesthesiol 2016;28:27–31.
106. Smith I, Nathanson M, White PF. Sevoflurane-a long awaited volatile anesthetic. Br J Anaesth 1996;76:435–45.
III. NEUROPHARMACOLOGY
REFERENCES
121
107. Eger II EI. New inhaled anesthetics. Anesthesiology 1994;80:906–22.
108. Goto t, Nakata Y, Morita S. Will xenon be a stranger or a friend? The cost, benefit, and future of xenon anesthesia. Anesthesiology
2003;98:1–2.
109. Koblin DD. Mechanisms of action. In: Miller RD, editor. Anesthesia. 4th ed New York: Churchill Livingston, Inc.; 1994. p. 67–99.
110. Mielck F, Stephan H, Buhre W, et al. Effects of 1 MAC desflurane on cerebral metabolism, blood flow and carbon dioxide reactivity in
humans. Br J Anaesth 1998;81:155–60.
111. Muzzi DA, Losasso TJ, Dietz NM, et al. The effect of desflurane and isoflurane on cerebrospinal fluid pressure in humans with supratentorial
mass lesions. Anesthesiology 1992;76:720–4.
112. Kim JK. Relationship of bispectral index to minimum alveolar concentration during isoflurane, sevoflurane or desflurane anaesthesia. J Int
Med Res 2014;42:130–7.
113. Dube SK, Pandia MP, Chaturvedi A, Bithal P, Dash HH. Comparison of intraoperative brain condition, hemodynamics and postoperative
recovery between desflurane and sevoflurane in patients undergoing supratentorial craniotomy. Saudi J Anaesth 2015;9:167–73.
114. Brosnan, Robert J, Thiesen R. Increased NMDA receptor inhibition at an increased Sevoflurane MAC. BMC Anesthesiol 2012;12:9.
115. Suzuki T, Koyama H, Sugimoto M, Uchida I, Mashimo T. The diverse actions of volatile and gaseous anesthetics on human-cloned 5-hydroxytryptamine3 receptors expressed in Xenopus oocytes. Anesthesiology 2002;96:699–704.
116. Talke p, Caldwell J, Dodsont B, et al. Desflurane and isoflurane increase lumbar cerebrospinal fluid pressure in normocapnic patients undergoing transsphenoidal hypophysectomy. Anesthesiology 1996;85:999–1004.
117. Wang J, Lei B, Popp S, Meng F, Cottrell JE, Kass IS. Sevoflurane immediate preconditioning alters hypoxic membrane potential changes
in rat hippocampal slices and improves recovery of CA1 pyramidal cells after hypoxia and global cerebral ischemia. Neuroscience
2007;145:1097–107.
118. Constant I, Seeman R, Murat I. Sevoflurane and epileptiform EEG changes. Paediatr Anaesth 2005;15:266–74.
119. Boonmak P, Boonmak S, Pattanittum P. High initial concentration versus low initial concentration sevoflurane for inhalational induction of
anaesthesia (review). Cochrane Syst Rev 2012;9:CD006837.
120. Valencia L, Rodríguez-Pérez A, Kühlmorgen B, Santana RY. Does sevoflurane preserve regional cerebral oxygen saturation measured by
near-infrared spectroscopy better than propofol? Ann Fr Anesth Reanim 2014;33:59–65.
121. Schlunzen L, Cold GE, Rasmussen M, et al. Effects of dose dependent levels of isoflurane on cerebral blood flow in healthy subjects studied
using positron emission tomography. Acta Anaesthesiol Scand 2006;50:306–12.
122. McPherson RW, Briar JE, traystman RJ. Cerebraovascular responsiveness to carbon dioxide in dogs 1ith 1.4% and 2.8% isoflurane.
Anesthesiology 1989;70:843–50.
123. McCulloch TJ, Boesel TW, Lam AM. The effect of hypocapnia on the autoregulation of CBF during administration of isoflurane. Anesth Analg
2005;100:1463–7.
124. Kitano H, Young JM, Cheng J, Wang L, Hurn PD, Murphy SJ. Gender-specific response to isoflurane preconditioning in focal cerebral ischemia. J Cereb Blood Flow Metab 2007;27:1377–86.
125. Zeiler FA, Zeiler KJ, Teitelbaum J, Gillman LM, West M. Modern inhalational anesthetics for refractory status epilepticus. Can J Neurol Sci
2015;42:106–15.
126. Takagaki M, Feuerstein D, Kumagai T, Gramer M, Yoshimine T, Graf R. Isoflurane suppresses cortical spreading depolarizations compared
to propofol–implications for sedation of neurocritical care patients. Exp Neurol 2014;252:12–7.
127. Yamakura T, Harris RA. Effects of gaseous anaesthetics nitrous oxide and xenon on ligand-gated ion channels. Comparison with isoflurane
and ethanol. Anesthesiology 2000;93:1095–101.
128. Mennerick S, Jevtovic-Todorovic V, Todorovic SM, Shen W, Olney JW, Zorumski CF. Effect of nitrous oxide on excitatory and inhibitory
synaptic transmission in hippocampal cultures. J Neurosci 1998;18:9716–26.
129. Lourenço-Matharu L, Ashley PF, Furness S. Sedation of children undergoing dental treatment (review). Cochrane Syst Rev 2012;(3):CD003877.
130. Singh GP, Prabhakar H, Bithal PK, Dash HH. A comparative evaluation of nitrous oxide-isoflurane vs isoflurane anesthesia in patients
undergoing craniotomy for supratentorial tumors: a preliminary study. Neurol India 2011;201(59):18–24.
131. Laitio RM, Kaisti KK, Låangsjö JW, Aalto S, Salmi E, Maksimow A, Aantaa R, Oikonen V, Sipilä H, Parkkola R, Scheinin H. Effects of xenon
anesthesia on cerebral blood flow in humans: a positron emission tomography study. Anesthesiology 2007;106:1128–33.
132. Rex S, Meyer PT, Baumert JH, Rossaint R, Fries M, Büll U, Schaefer WM. Positron emission tomography study of regional cerebral blood flow
and flow-metabolism coupling during general anaesthesia with xenon in humans. Br J Anaesth 2008;100:667–75.
133. Schmidt M, Marx T, Papp Jambor C, et al. Effect of xenon on cerebral autoregulation in pigs. Anesthesia 2002;57:960–6.
134. Azzopardi D, Robertson NJ, Kapetanakis A, Griffiths J, Rennie JM, Mathieson SR, Edwards AD. Anticonvulsant effect of xenon on neonatal
asphyxial seizures. Arch Dis Child Fetal Neonatal Ed 2013;98:437–9.
135. Sanfilippo M, Wefki Abdelgawwad Shousha AA, Paparazzo A. Emergence in elderly patient undergoing general anesthesia with xenon. Case
Rep Anesthesiol 2013;2013:736–90.
136. Griffith HR, Johnson GE. The use of curare in general anesthesia. Anesthesiology 1942;3:418–20.
137. Lanier WL, Iaizzo PA, Milde JH. Cerebral blood flow and afferent muscle activity following IV succinylcholine in dogs. Anesthesiology Rev
1987;14:60–6.
138. Minton MD, Grosslight K, Stirt JA, et al. Increase in intracranial pressure from succinylcholine: prevention by prior nondepolarizing blockade. Anesthesiology 1986;65:165–9.
139. Ahsan B, Rahimi E, Moradi A, Rashadmanesh N. The effects of magnesium sulphate on succinylcholine-induced fasciculation during induction of general anaesthesia. J Pak Med Assoc 2014;6:1151–3.
140. Ganigara A, Ravishankar C, Ramavakoda C, Nishtala M. Fatal hyperkalemia following succinylcholine administration in a child on oral
propranolol. Drug Metabol Personal Ther 2015;30:69–71.
141. Bowman WC. Neuromuscular block. Br J Pharmacol 2006;147:277–86.
142. Waud BE, Waud DR. The relation between tetanic fade and receptor occlusion in the presence of competitive neuromuscular block.
Anesthesiology 1971;35:456–64.
III. NEUROPHARMACOLOGY
122
6. NEUROPHARMACOLOGY
143. Li Y-L, Liu Y-L, Xu C-M, Lv X-H, Wan Z-H. The effects of neuromuscular blockade on operating conditions during general anesthesia for
spinal surgery. J Neurosurg Anesthesiol 2014;26:45–9.
144. Butterworth JF, Strichartz GR. Molecular mechanisms of local anesthetics: a review. Anesthesiology 1990;72:722–34.
145. Mitchell SJ, Pellett O, Gorman DF. Cerebral protection by lidocaine during cardiac operations. Ann Thorac Surg 1999;67:1117–24.
146. Stanton TR, Wand BM, Carr DB, Birklein F, Wasner GL, O’Connell NE. Local anaesthetic sympathetic blockade for complex regional pain
syndrome (review). Cochrane Database Syst Rev 2013;8:CD004598.
147. Khalil N, Nicotra A, Rakowicz W. Treatment for meralgia paraesthetica. Cochrane Database Syst Rev 2012;12:CD004159.
148. Takita A, Masui K, Kazama T. Online monitoring of end-tidal propofol concentration in anesthetized patients. Anesthesiology 2007;106:659–64.
149. Rossignol S, Scwab M, Schwartz M, Fehlings MG. Spinal cord injury: time to move? J Neurosci 2007;27:11782–92.
III. NEUROPHARMACOLOGY
C H A P T E R
7
Anesthetic Agents: Neurotoxics or
Neuroprotectives?
J. Fiorda-Diaz, N. Stoicea, S.D. Bergese
Ohio State University, Columbus, OH, United States
O U T L I N E
Introduction
123
Anesthesia Practice: Clinical Outcomes
126
Pharmacological Considerations
Neurotoxicity of Anesthetic Drugs
Inhaled Anesthetics
Intravenous Anesthetics
Neuroprotection of Anesthetic Drugs
Inhaled Anesthetics
Intravenous Anesthetics
124
124
124
124
125
125
126
Anesthesia and Fragile Brain
127
Conclusion
127
Abbreviations
128
References
128
INTRODUCTION
The National Health Statistics Report published in 2010 estimated that around 45 million inpatient surgical procedures are carried out annually in the United States with an increasing rate among older population (≥65 years
old).1 Newborns’ data was excluded from this survey although around 1.5 million interventions per year have
been reported.2 Therefore, a considerable number of patients are annually being exposed to potential perioperative
complications.
Central nervous system (CNS) functioning is affected during anesthesia. Neurons in eloquent areas are commonly
targeted by local and general anesthetic drugs, interfering with their physiologic mechanisms and autoregulation.3,4
An extensive body of evidence assessed the neuroprotective and neurotoxic effects of anesthetics.
Different individual factors should be considered to assess the actual risk of developing perioperative neurological complications. Cerebral metabolic rate is decreased as a result of anesthetic drugs use. However, correlation between cerebral blood flow (CBF) and oxygen consumption is usually conserved. Disruption of the cerebral
autoregulation involves several variables related to anesthesia (exposition to anesthetics, depth of anesthesia, high
plasmatic concentration of anesthetics), surgery (cardiovascular or neurologic surgery), or patient demographics and
medical history (aging, comorbidities, etc.).
Molecular mechanisms involving anesthetic-induced neurotoxicity were extensively studied in animal models.5
Nevertheless, because of the natural limitations imposed by the clinical research it is difficult to conclude whether
these processes similarly occur within human physiopathological conditions.6–9 Studies published in more than
five decades have reported on neuroprotective effects of anesthetics. Decreased CBF, intracranial pressure (ICP), and
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00007-5
123
© 2017 Elsevier Inc. All rights reserved.
124
7. ANESTHETIC AGENTS: NEUROTOXIC OR NEUROPROTECTIVE?
rate of oxygen consumption were the first neuroprotective mechanisms described for both volatile and intravenous
anesthetics.10,11 Paradoxically, anesthetics’ neuroprotective mechanisms are linked to mitochondria, the same organelle in which anesthetics’ neurotoxic effects are carried out.
PHARMACOLOGICAL CONSIDERATIONS
Neurotoxicity of Anesthetic Drugs
In general, the molecular effect of anesthetics is studied in different species including nonhuman primates.
Relevant findings explaining potential neurotoxic effects in fragile brain models have been reported.12,13
A pioneer study published by Ikonomidou et al. in 1999 associated anesthesia with neurotoxicity.14 Glutamate is
one of the major neurotransmitter with an important role during developing brain stages. Therefore, N-methyl-daspartate (NMDA) antagonists (+MK801, ketamine, phencyclidine, and carboxypiperazin-4-yl-propyl-1-phosphonic
acid) are able to trigger dose- and time-dependent proapoptotic responses leading to cellular degeneration in rats.
Similar consequences may be responsible for neurological impairments observed in children whose mothers were
exposed to comparable substances during pregnancy.14
Inhaled Anesthetics
Retinal cells have shown neurodegeneration and apoptosis after isoflurane exposure in rats. Cheng et al. studied two groups of 7-day-old rats comparing mitochondrial responses in retinal cells. One of the groups was
exposed for 1 h to air (control group), and the second group was exposed for 1 h to isoflurane 2% inhalation
(interventional group) in 8–12 L of fresh gas flow mixture.15 Caspase 3 activation is the final stage after intrinsic and extrinsic apoptotic cascade initiation. Isoflurane is capable of acting on caspase 3 and initializing final
apoptotic cascade throughout several mechanisms such as inactivation of proteins with antiapoptotic activity
(BCL-xL and BCL-2), increase of the mitochondrial membrane permeability, and activation of caspases 8 and
9.15,16 Retinal cells seem to be a good starting point to assess the potentially deleterious effects of some anesthetics on the CNS as they may be directly observed using noninvasive techniques.15 Additionally, the effect of
inhaled anesthetics in the progression of neurodegenerative disorders in humans is supported by studies done
in animal models experiencing an increase in proinflammatory cytokines levels, such as tumor necrosis factor-α
(TNF-α), after isoflurane exposure.17
Wang et al. reported that sevoflurane administration for a 6-h period in 7-day-old rats causes a significant impairment in astrocytes functioning. An important reduction in GLAST (glutamate-aspartate transporter) and JAK/STAT
(Janus kinase/signal transducer and activator of transcription) activity was noticed after sevoflurane administration
and considered to be associated with impaired astrocytic proliferation.18 Neuronal cells exposure to sevoflurane triggered endoplasmic reticulum dysfunction and dysregulation of intracellular Ca2+ homeostasis.19
Schallner et al. studied the effects of sevoflurane, isoflurane, and desflurane in neuronal cells after induced hypoxia.
Activation of a protein linked to cellular stress response (NF-κB) with concomitant inactivation of p75NTR, was seen
under isoflurane exposure but not after the use of sevoflurane or desflurane. In other words, isoflurane is associated
with the progression of neuronal death under hypoxic conditions. This mechanism was not associated with sevoflurane or desflurane during in vitro and in vivo studies, promoting the use of these agents in patients with a history of
stroke and cardiovascular interventions.20
Intravenous Anesthetics
Komuro and Rakic also studied the role of the NMDA receptor and its blockade during neurological cells development. A significant disruption in cell migration was noticed using NMDA receptor antagonists (MK-801 and D-AP5),
but not γ-aminobutyric acid (GABA) receptors antagonists. Cellular migration is a major stage necessary for CNS
formation and functionality. These findings may explain how ketamine triggers some neurotoxic mechanisms associated with its use.21
Upregulation of NMDA receptor with a consequent increase in receptor expression and activity is a well-described
neuronal mechanism occurring after its exposure to several antagonists. Ketamine is responsible for an overstimulation of NMDA receptors through their natural ligand (glutamate), explaining other mechanisms related to subsequent activation of the proapoptotic cascade with a greater influx of sodium, calcium, and chloride into the cytosol.22
Anesthesia-related neurodegeneration in rats is characterized by an accelerated rate of cellular genesis commonly
found during developmental phases. However, with respect to clinical studies several variables might interfere.12,23
III. NEUROPHARMACOLOGY
PHARMACOLOGICAL CONSIDERATIONS
125
Therefore, it is important to differentiate and identify important factors involving basic research (drug-related information such as dose and length of exposure, vitals monitoring, oxygenation, and others) and general anesthesia in
children undergoing surgery.23
This “controlled setting” characterizing clinical research may explain some of the remaining controversies between
research findings in animal models and clinical outcomes in humans. Slikker et al. compared the response of rhesus
monkeys to anesthetic doses of ketamine with a control group, monitoring hemodynamics, and other physiological
variables (oxygen saturation, capnography, noninvasive blood pressure, and temperature).12 Even under “physiological conditions,” caspase 3 expression in cortical neurons was found to be significantly increased in ketamine
group, indicating stimulation of the apoptotic cascade.12
Comparable in vitro results were published by Bai et al. who found that ketamine induced caspase 3 activation and increased reactive oxygen species (ROS) in human stem cells.24 However, cell death and degeneration also
depend on some other factors such as age, dose, and duration of exposure. Even during a single exposure to ketamine, anesthetic concentrations achieved in critical periods of brain cell development may be responsible for longterm cognitive impairment.25
Propofol is a well-known anesthetic drug used in pediatrics either for sedation or general anesthesia. Similar to
ketamine, in vitro animal model studies using different propofol concentrations, comparable with in vivo plasmatic
values reached during general anesthesia, reported unfavorable reactions of immature neuronal tissue. In fact,
increased influx of calcium and chloride, mediated by GABAA receptor activation, may stimulate proapoptotic pathways leading to programmed cell death in immature neuronal cells.26,27
A comparable mechanism for neuronal degeneration and apoptosis in animal models has also been described
with benzodiazepines use.28 Diazepam used at 10 mg/kg was associated with neuronal cell damage in rats during
early stages of brain development. These proapoptotic reactions were not triggered when diazepam was antagonized by flumazenil administration.29 Additionally, cortical neurons and the caudate/putamen nuclei seem to be
the most affected after midazolam use, a short-acting benzodiazepine, with greater apoptotic effects obtained during concomitant ketamine administration.28 The combination of these two anesthetic drugs is common in pediatric
anesthesia.
Opioids have also been associated with neurotoxicity and brain cell dysfunction. Fentanyl, remifentanil, sufentanil, and other µ-agonists have been associated with brain cells damage with consequent seizure activity in animal
models.30,31
Neuroprotection of Anesthetic Drugs
The neuroprotective effects of intravenous and inhaled anesthetics consist of preserving cerebral autoregulation
and normal ICP, avoiding seizure activities, and providing at the same time an adequate anesthetic and analgesic
level during surgery. In general, these effects are dose dependent. Hence, administration of more than 1.0 minimal
alveolar concentration (MAC) of most inhaled anesthetics is associated with cerebral vasodilation with a subsequent
increase in CBF.32
Coupling between CBF and neurological cells metabolism is a well-described physiologic and protective mechanism. Brain pathologies and pharmacological effects of some drugs may result in cerebral coupling disruption.33
Inhaled Anesthetics
There is no consensus regarding the preference of inhaled anesthetic use during neurosurgery, although particular
effects have been described for all of them. Sevoflurane has shown the ability to maintain cerebral coupling in a better
way than isoflurane at anesthetic doses without significantly increasing ICP.34 Reduction in CBF as a result of reduced
metabolism (coupling mechanism) has been reported with low doses of sevoflurane, isoflurane, and halothane.35
However, specific reactions may occur simultaneously within anterior and posterior cerebral circulation depending
on the type of inhaled anesthetic used.36
Neurological cell impairment has been described as a result of the activation of NMDA receptors. Partial inhibition of these receptors during inhalational anesthesia is linked to the neuroprotective effect conferred by isoflurane,
sevoflurane, and desflurane. In vitro, isoflurane is responsible for a greater percentage of NMDA receptor inhibition
when compared to other halogenated anesthetics, in preference to sevoflurane and desflurane.37 Moreover, studies
conducted in animal models and using inhalational anesthesia with sevoflurane and isoflurane reported enhanced
mitochondrial activity by decreasing the activation of crucial proapoptotic mechanisms and preconditioning to cerebral ischemia.38–40
III. NEUROPHARMACOLOGY
126
7. ANESTHETIC AGENTS: NEUROTOXIC OR NEUROPROTECTIVE?
Intravenous Anesthetics
Systemic pathophysiological responses including exacerbation of sympathomimetic reactions and generation of
ROS are triggered by the ischemia/reperfusion phenomenon in neurological cells. ROS generated during hypoxic
stages will cause peroxidation of cells’ membranes during reperfusion. Malondialdehyde (MDA) is one of the metabolic markers used to quantify the intensity of these events. Propofol seems to have a significant neuroprotective role
by preventing peroxidation of the membranes, its antioxidant effect being described in ischemic animal models.41
Propofol administration is responsible for regulation of proapoptotic and antiapoptotic genes, decreased caspase 3
activity, and regulation of other proteins such as p53 involved in programmed death of cells.42
Ischemic insult in neurological cells is associated with downregulation of GABA receptors. In vitro studies have
shown that stimulation of GABA receptors may be linked to cellular death. However, diazepam exerts a positive allosteric effect on GABA receptors enhancing the affinity for its ligand, and providing a neuroprotective effect in vivo
by attenuating cells death.43,44 This attenuation has been explained as a result of increased local blood flow in the
penumbra areas or decreased nitric oxide (NO) synthesis.44
Dexmedetomidine, a well-known α2 agonist, has the ability to mitigate sympathomimetic effects by mediating
the norepinephrine and dopaminergic effects on hippocampal cells avoiding neuronal cell death after sustainable
ischemia.45,46 Moreover, inflammatory markers, ROS, and their consequent deleterious effects during ischemiareperfusion phenomena have been found to be reduced in hippocampal and dentate gyrus cells after dexmedetomidine administration.47 Eser et al. reported the effects of dexmedetomidine on ischemia/reperfusion animal models
by describing significant differences in levels of TNF-α, NO, MDA, and apoptotic neuronal cells with concomitant
increased activity in superoxide dismutase and catalase.47 Moreover, an increased activation of antiapoptotic pathways involving the modulation of Bcl-2 proteins activity has also been reported in ischemia/reperfusion models
after dexmedetomidine exposure.48 Li et al. reported an increase of 217% in caspase 3 activity in fetal rats whose
mothers were exposed to propofol during pregnancy.49 This proapoptotic response was significantly attenuated
when dexmedetomidine was used and explained by a decrease of microglial stimulation.49
ANESTHESIA PRACTICE: CLINICAL OUTCOMES
During the past decades, a considerable body of scientific evidence supports the association between general
anesthesia and behavioral changes within the first year of life in infants undergoing surgeries. Most of these associations were described in retrospective studies. Levy (1945) named these outcomes “emotional sequelaes” and linked
the behavioral changes to surgery and anesthesia. Levy observed that children exposed to surgery and anesthesia
within the first months of life were characterized by learning impairments, poor social skills, behavioral changes,
and common habits such as nail biting, masturbation, hair pulling, enuresis, and soiling. Surprisingly, he found that
50% of children who underwent surgery (usually tonsillectomy) at the age of 3 years or earlier had one or more emotional sequelaes.50 However, several anesthetic drugs have been created since then and several research trials have
been developed to determine these correlations.
Even though human anesthetic neurotoxicity cannot be assessed by reproducing studies from animal models,
different clinical outcomes such as postoperative cognitive dysfunction (POCD), delirium, and impaired learning
abilities in children are identified as consequences of neurological cell dysfunction after anesthesia exposure.
Zhang et al. carried out a pilot study to analyze the cognitive changes after general anesthesia with different hypnotics. The authors randomized 45 subjects undergoing lower extremity or abdominal surgery to three groups. One
group received spinal anesthesia and isoflurane general anesthesia, the second group received spinal anesthesia and
desflurane general anesthesia, and the third group received spinal anesthesia (control). Although with considerable
limitations, this study showed a significant increased incidence of POCD in the isoflurane group (27%) when compared with control group (0%), and desflurane (0%).51 Therefore, stimulation of proapoptotic pathways observed in
animal models after isoflurane exposure may be comparable with neurological outcomes such as cognitive dysfunction reported in patients who underwent general anesthesia with isoflurane. However, decreased dynamic cerebral
autoregulation has been noticed at 0.5 MAC for both isoflurane and desflurane, with absence of autoregulation at
1.5 MAC.52
Evaluation of the cognitive function in children who underwent general anesthesia in critical stages of brain development has been a major concern during the past decades among physicians. To avoid the limitations entailed by
studies with retrospective design, a multidisciplinary group has initiated a longitudinal multicenter study to evaluate
the cognitive outcomes in children who received general anesthesia before the age of 3 years. The PANDA (Pediatric
III. NEUROPHARMACOLOGY
CONCLUSION
127
Anesthesia and Neurodevelopment Assessment) study will compare the cognitive functionality and neuropsychological characteristics between siblings where one of them was exposed to general anesthesia due to inguinal hernia repair.
A pilot study published by the PANDA group showed the feasibility of developing an ambidirectional project, collecting previous data from parents, electronic medical (anesthesia) records, and by performing neuropsychological examinations in children at different stages of the life.53 The Mayo Anesthesia Safety in Kids study is also expected to offer
valuable data by enrolling patients who were exposed to general anesthesia before they were 3 years old. They planned
to assess neurocognitive status in children between ages of 8–12 and 15–19 years and exposed to multiple, single or no
anesthesia through an extensive battery of tests performed one time during a period 2012–16.54
Flick et al. reported no difference in learning ability between patients who had a unique general anesthesia episode before the age of 2 years and those who did not receive anesthesia. Nevertheless, receiving general anesthesia
two or more times was identified as a risk factor for learning disabilities. Additionally, individualized education programs were more commonly used for patients who received anesthesia two or more times.55 On the other hand, the
GAS study assessed the neurological development in 2-year-old children who underwent inguinal hernia repair by
receiving either general anesthesia with sevoflurane, or regional anesthesia within the first 60 postmenstrual weeks
(gestational + chronological age). Quality data was collected from more than 500 children showing that general anesthesia with sevoflurane lasting less than 1 h was not associated with developmental neurological impairment.56
Decreased CBF and brain metabolism has been described during propofol anesthesia in patients undergoing cardiopulmonary bypass.57,58 Although these encouraging results have been considered as neuroprotective effects in
animal models, current clinical data do not show conclusive results.59,60 Randomized clinical trials using propofol
during coronary artery bypass grafting and brain aneurysm clipping surgery found no significant differences in
POCD incidence or neurological outcomes.61,62
ANESTHESIA AND FRAGILE BRAIN
POCD, delirium, and dementia are commonly diagnosed among elderly patients who underwent cardiac and
noncardiac major surgery. Age, nutritional state, and preoperative cognitive impairment are the main risk factors for
the aforementioned postoperative complications.63,64
Perioperative management of hemodynamic parameters such as oxygen saturation and mean arterial pressure
(MAP) may not play a significant role in the onset of postoperative cognitive impairment as it was thought. The
International Study of Post-Operative Cognitive Dysfunction analyzed the data collected from 1218 elderly patients
(more than 60 years old) who underwent major noncardiac surgical procedures. Patients with low Mini–Mental State
Examination scores (≤23) and those with neurological comorbidities were excluded during screening. Continuous
monitoring of oxygen saturation was performed along with periodic monitoring of the blood pressure. POCD was
diagnosed in 266 patients (25.8% vs. 3.4% in control group) in the first postoperative week. After 3 months, POCD
was diagnosed in 94 patients (9.9% vs. 2.8% in control group). Finally, authors could not find a significant correlation
between low oxygen saturation or MAP and POCD incidence.65
Certainly, apoptosis is linked to the onset and progression of many neurological diseases. Apoptotic mechanisms
along with an increase of β-amyloid protein play an essential role in the development of the Alzheimer disease. In
addition to the proapoptotic effects, isoflurane has been linked to an increase in β-amyloid protein synthesis by
modulating enzymatic reactions.66,67 This positive enzymatic modulation is dose dependent, being attenuated at
lower doses and explained by a potential protective cellular mechanism.68
CONCLUSION
Human research describing potential mechanisms of neurodegeneration is limited. Studies conducted in animal
models are difficult to reproduce in the clinical setting. Neuroprotective and neurotoxic effects of anesthetics are currently under exhaustive investigation, especially for extreme age populations (children and older patients).
Surgery and anesthesia trigger several molecular responses reflected in patient clinical outcomes. Most of the
studies concluded that neurotoxic effects are usually found at higher doses, neuroprotective effects commonly being
achieved under regular plasmatic concentration. Currently, plenty of anesthetic drugs with different mechanisms of
action are available. Taking advantage of synergic effects of hypnotic and opioid medication should be considered
based on patient comorbidities.
III. NEUROPHARMACOLOGY
128
7. ANESTHETIC AGENTS: NEUROTOXIC OR NEUROPROTECTIVE?
Data collected from ongoing clinical trials will contribute to the general effort of establishing guidelines for anesthesia management during early stages of human neurological development. Whether or not these trials will find a
direct association between anesthesia, neurotoxicity, and impaired neurological development in children, they will
definitely bring new valuable information for parents and physicians.
Inflammation-induced reactions, cellular dysfunction, and apoptosis studied in vivo and in vitro after anesthesia
exposure are strongly related to persistent cognitive dysfunction diagnosed in older patients. In addition, patients’
comorbidities and current psychosocial status may impact postoperative neurological outcomes. Although new
anesthetic drugs with less associated neurotoxic effects seem to be an attractive option, future research encourages
various techniques of brain preconditioning to improve patient preoperative cognitive status. However, an important role is played by translational research through its ability to create bridging designs by applying animal model
concepts to clinical trial protocols.
ABBREVIATIONS
BCL-xL and BCL-2 B-cell lymphoma-extra large and B-cell lymphoma 2
CPP Carboxypiperazin-4-yl-propyl-1-phosphonic acid
D-AP5 d-2-Amino-5-phosphono-pentanoic acid
MK-801 (+)-5-Methyl-10,11-dihydro-5H-dibenzo{a,d}cyclohepten-5,10-imine hydrogen maleate
NF-κB Nuclear factor-kappa light chain enhancer of activated B cells
p75NTR p75 Neutrophin receptor
PCP Phencyclidine
References
1. Hall MJ, DeFrances CJ, Williams SN, Golosinskiy A, Schwartzman A. National hospital discharge survey: 2007 summary. Natl Health Stat Rep 2007;(29):1–20, 24.
2. Anand KJ, Soriano SG. Anesthetic agents and the immature brain: are these toxic or therapeutic? J Am Soc Anesthesiol 2004;101(2):527–30.
3. Pittson S, Himmel AM, MacIver MB. Multiple synaptic and membrane sites of anesthetic action in the CA1 region of rat hippocampal slices. BMC Neurosci
2004;5(1):52.
4. Franceschini MA, Radhakrishnan H, Thakur K, et al. The effect of different anesthetics on neurovascular coupling. Neuroimage 2010;51(4):1367–77.
5. Stratmann G. Neurotoxicity of anesthetic drugs in the developing brain. Anesth Analg 2011;113(5):1170–9.
6. Thomas J, Crosby G, Drummond JC, Todd M. Anesthetic neurotoxicity: a difficult dragon to slay. Anesth Analg 2011;113(5):969–71.
7. McGowan Jr FX, Davis PJ. Anesthetic-related neurotoxicity in the developing infant: of mice, rats, monkeys and, possibly, humans. Anesth Analg 2008;106(6):1599–602.
8. Mellon RD, Simone AF, Rappaport BA. Use of anesthetic agents in neonates and young children. Anesth Analg 2007;104(3):509–20.
9. Armstrong R, Xu F, Arora A, Rasic N, Syed NI. General anesthetics and cytotoxicity: possible implications for brain health. Drug Chem Toxicol 2016:1–9.
10. Smith AL, Hoff JT, Nielsen SL, Larson CP. Barbiturate protection in acute focal cerebral ischemia. Stroke 1974;5(1):1–7.
11. Yatsu F, Diamond I, Graziano C, Lindquist P. Experimental brain ischemia: protection from irreversible damage with a rapid-acting barbiturate (methohexital).
Stroke 1972;3(6):726–32.
12. Slikker W, Zou X, Hotchkiss CE, et al. Ketamine-induced neuronal cell death in the perinatal rhesus monkey. Toxicol Sci 2007;98(1):145–58.
13. Durieux ME. Anesthetic neurotoxicity: it’s not just for children anymore. Anesth Analg 2010;110(2):291–2.
14. Ikonomidou C, Bosch F, Miksa M, et al. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science 1999;283(5398):70–4.
15. Cheng Y, He L, Prasad V, Wang S, Levy RJ. Anesthesia-induced neuronal apoptosis in the developing retina: a window of opportunity. Anesth Analg
2015;121(5):1325–35.
16. Wang H, Dong Y, Zhang J, et al. Isoflurane induces endoplasmic reticulum stress and caspase activation through ryanodine receptors. Br J Anaesth 2014:aeu053.
17. Wu X, Lu Y, Dong Y, et al. The inhalation anesthetic isoflurane increases levels of proinflammatory TNF-α, IL-6, and IL-1β. Neurobiol Aging 2012;33(7):1364–78.
18. Wang W, Lu R, Feng D-y, Zhang H. Sevoflurane inhibits glutamate-aspartate transporter and glial fibrillary acidic protein expression in hippocampal astrocytes
of neonatal rats through the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. Anesth Analg 2016;123(1):93–102.
19. Komita M, Jin H, Aoe T. The effect of endoplasmic reticulum stress on neurotoxicity caused by inhaled anesthetics. Anesth Analg 2013;117(5):1197–204.
20. Schallner N, Ulbrich F, Engelstaedter H, et al. Isoflurane but not sevoflurane or desflurane aggravates injury to neurons in vitro and in vivo via p75NTR-NF-ĸB
activation. Anesth Analg 2014;119(6):1429–41.
21. Komuro H, Rakic P. Modulation of neuronal migration by NMDA receptors. Science 1993;260(5104):95–7.
22. Wang C, Sadovova N, Hotchkiss C, et al. Blockade of N-methyl-d-aspartate receptors by ketamine produces loss of postnatal day 3 monkey frontal cortical neurons
in culture. Toxicol Sci 2006;91(1):192–201.
23. Loepke AW, McGowan Jr FX, Soriano SG. CON: the toxic effects of anesthetics in the developing brain: the clinical perspective. Anesth Analg 2008;106(6):1664–9.
24. Bai X, Yan Y, Canfield S, et al. Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated
mitochondrial pathway. Anesth Analg 2013;116(4):869.
25. Paule M, Li M, Allen R, et al. Ketamine anesthesia during the first week of life can cause long-lasting cognitive deficits in rhesus monkeys. Neurotoxicol Teratol
2011;33(2):220–30.
26. Kahraman S, Zup SL, McCarthy MM, Fiskum G. GABAergic mechanism of propofol toxicity in immature neurons. J Neurosurg Anesthesiol 2008;20(4):233.
27. Sinner B, Becke K, Engelhard K. General anaesthetics and the developing brain: an overview. Anaesthesia 2014;69(9):1009–22.
28. Young C, Jevtovic-Todorovic V, Qin YQ, et al. Potential of ketamine and midazolam, individually or in combination, to induce apoptotic neurodegeneration in the
infant mouse brain. Br J Pharmacol 2005;146(2):189–97.
29. Bittigau P, Sifringer M, Genz K, et al. Antiepileptic drugs and apoptotic neurodegeneration in the developing brain. Proc Natl Acad Sci USA 2002;99(23):15089–94.
30. Kofke WA, Garman RH, Stiller RL, Rose ME, Garman R. Opioid neurotoxicity: fentanyl dose-response effects in rats. Anesth Analg 1996;83(6):1298–306.
31. Kofke WA, Attaallah AF, Kuwabara H, et al. The neuropathologic effects in rats and neurometabolic effects in humans of large-dose remifentanil. Anesth Analg
2002;94(5):1229–36.
III. NEUROPHARMACOLOGY
REFERENCES
129
32. Conti A, Iacopino D, Fodale V, Micalizzi S, Penna O, Santamaria L. Cerebral haemodynamic changes during propofol–remifentanil or sevoflurane anaesthesia:
transcranial Doppler study under bispectral index monitoring. Br J Anaesth 2006;97(3):333–9.
33. Hansen TD, Warner DS, Todd MM, Vust LJ. The role of cerebral metabolism in determining the local cerebral blood flow effects of volatile anesthetics: evidence
for persistent flow-metabolism coupling. J Cereb Blood Flow Metab 1989;9(3):323–8.
34. Matta BF, Heath KJ, Tipping K, Summors AC. Direct cerebral vasodilatory effects of sevoflurane and isoflurane. J Am Soc Anesthesiol 1999;91(3):677.
35. Engelhard K, Werner C. Inhalational or intravenous anesthetics for craniotomies? Pro inhalational. Curr Opin Anesthesiol 2006;19(5):504–8.
36. Rozet I, Vavilala MS, Lindley AM, Visco E, Treggiari M, Lam AM. Cerebral autoregulation and CO2 reactivity in anterior and posterior cerebral circulation during
sevoflurane anesthesia. Anesth Analg 2006;102(2):560–4.
37. Solt K, Eger EI, Raines DE. Differential modulation of human N-methyl-d-aspartate receptors by structurally diverse general anesthetics. Anesth Analg
2006;102(5):1407–11.
38. Ye R, Yang Q, Kong X, et al. Sevoflurane preconditioning improves mitochondrial function and long-term neurologic sequelae after transient cerebral ischemia:
role of mitochondrial permeability transition. Crit Care Med 2012;40(9):2685–93.
39. Zhang H-p, Sun Y-y, Chen X-m, et al. The neuroprotective effects of isoflurane preconditioning in a murine transient global cerebral ischemia–reperfusion model:
the role of the notch signaling pathway. Neuromol Med 2014;16(1):191–204.
40. Wang H, Shi H, Yu Q, Chen J, Zhang F, Gao Y. Sevoflurane preconditioning confers neuroprotection via anti-apoptosis effects. Brain Edema XVI: Springer 2016:55–61.
41. Ergün R, Akdemir G, Șen S, Tașçı A, Ergüngör F. Neuroprotective effects of propofol following global cerebral ischemia in rats. Neurosurg Rev 2002;25(1–2):95–8.
42. Fan W, Zhu X, Wu L, et al. Propofol: An anesthetic possessing neuroprotective effects. Eur Rev Med Pharmacol Sci 2015;19:1520–9.
43. Sarnowska A, Beręsewicz M, Zabłocka B, Domańska-Janik K. Diazepam neuroprotection in excitotoxic and oxidative stress involves a mitochondrial mechanism
additional to the GABA A R and hypothermic effects. Neurochem Int 2009;55(1):164–73.
44. Aerden L, Kessels F, Rutten B, Lodder J, Steinbusch H. Diazepam reduces brain lesion size in a photothrombotic model of focal ischemia in rats. Neuroscience Lett
2004;367(1):76–8.
45. Kuhmonen J, Pokorny J, Miettinen R, Haapalinna A, Jolkkonen J, Sivenius J. Neuroprotective effects of dexmedetomidine in the gerbil hippocampus after transient
global ischemia. J Am Soc Anesthesiol 1997;87(2):371–7.
46. Farag E, Argalious M, Sessler DI, Kurz A, Ebrahim ZY, Schubert A. Use of α2-agonists in neuroanesthesia: an overview. Ochsner J 2011;11(1):57–69.
47. Eser O, Fidan H, Sahin O, et al. The influence of dexmedetomidine on ischemic rat hippocampus. Brain Res 2008;1218:250–6.
48. Engelhard K, Werner C, Eberspächer E, et al. The effect of the α2-agonist dexmedetomidine and the N-methyl-d-aspartate antagonist S (+)-ketamine on the expression of apoptosis-regulating proteins after incomplete cerebral ischemia and reperfusion in rats. Anesth Analg 2003;96(2):524–31.
49. Li J, Xiong M, Nadavaluru PR, et al. Dexmedetomidine attenuates neurotoxicity induced by prenatal propofol exposure. J Neurosurg Anesthesiol 2016;28(1):51–64.
50. Levy DM. Psychic trauma of operations in children: and a note on combat neurosis. Am J Dis Child 1945;69(1):7–25.
51. Zhang B, Tian M, Zhen Y, et al. The effects of isoflurane and desflurane on cognitive function in humans. Anesth Analg 2012;114(2):410.
52. Strebel S, Lam A, Matta B, Mayberg TS, Aaslid R, Newell DW. Dynamic and static cerebral autoregulation during isoflurane, desflurane, and propofol anesthesia.
J Am Soc Anesthesiol 1995;83(1):66–76.
53. Sun LS, Li G, DiMaggio CJ, et al. Feasibility and pilot study of the Pediatric Anesthesia NeuroDevelopment Assessment (PANDA) project. J Neurosurg Anesthesiol
2012;24(4):382.
54. Gleich SJ, Flick R, Hu D, et al. Neurodevelopment of children exposed to anesthesia: design of the Mayo Anesthesia Safety in Kids (MASK) study. Contemp Clin
Trials 2015;41:45–54.
55. Flick RP, Katusic SK, Colligan RC, et al. Cognitive and behavioral outcomes after early exposure to anesthesia and surgery. Pediatrics 2011;128(5):e1053–61.
56. Davidson AJ, Disma N, de Graaff JC, et al. Neurodevelopmental outcome at 2 years of age after general anaesthesia and awake-regional anaesthesia in infancy
(GAS): an international multicentre, randomised controlled trial. Lancet 2016;387(10015):239–50.
57. Newman MF, Murkin JM, Roach G, et al. Cerebral physiologic effects of burst suppression doses of propofol during nonpulsatile cardiopulmonary bypass. Anesth
Analg 1995;81(3):452–7.
58. Souter M, Andrews P, Alston R. Propofol does not ameliorate cerebral venous oxyhemoglobin desaturation during hypothermic cardiopulmonary bypass. Anesth
Analg 1998;86(5):926–31.
59. Zwerus R, Absalom A. Update on anesthetic neuroprotection. Curr Opin Anesthesiol 2015;28(4):424–30.
60. Bilotta F, Stazi E, Zlotnik A, Gruenbaum SE, Rosa G. Neuroprotective effects of intravenous anesthetics: a new critical perspective. Curr Pharm Des 2014;20(34):5469.
61. Mahajan C, Chouhan RS, Rath GP, et al. Effect of intraoperative brain protection with propofol on postoperative cognition in patients undergoing temporary clipping during intracranial aneurysm surgery. Neurol India 2014;62(3):262.
62. Kanbak M, Saricaoglu F, Avci A, Ocal T, Koray Z, Aypar U. Propofol offers no advantage over isoflurane anesthesia for cerebral protection during cardiopulmonary bypass: a preliminary study of S-100ß protein levels. Can J Anesth 2004;51(7):712–7.
63. Chen CCH, Saczynski J, Inouye SK. The modified Hospital Elder Life Program: adapting a complex intervention for feasibility and scalability in a surgical setting.
J Gerontol Nurs 2014;40(5):16–22.
64. Bergese S. Fragile brains,,The Brain of the Elderlyʼ. J Neuroanaesth Crit Care 2016;3(4):41.
65. Moller J, Cluitmans P, Rasmussen L, et al. Long-term postoperative cognitive dysfunction in the elderly: ISPOCD1 study. Lancet 1998;351(9106):857–61.
66. Wei H. The role of calcium dysregulation in anesthetic-mediated neurotoxicity. Anesth Analg 2011;113(5):972.
67. Wei H, Xie Z. Anesthesia, calcium homeostasis and Alzheimer’s disease. Curr Alzheimer Res 2009;6(1):30.
68. Pan C, Xu Z, Dong Y, et al. The potential dual effects of anesthetic isoflurane on hypoxia-induced caspase-3 activation and increases in β-site amyloid precursor
protein-cleaving enzyme levels. Anesth Analg 2011;113(1):145.
III. NEUROPHARMACOLOGY
This page intentionally left blank
S E C T I O N
I V
NEUROMONITORING
This page intentionally left blank
C H A P T E R
8
Neuromonitoring
V.J. Ramesh, M. Radhakrishnan
National Institute of Mental Health and NeuroSciences, Bengaluru, India
O U T L I N E
Introduction
134
Cerebral Blood Flow
Transcranial Doppler
Assumptions
Technique
Pulsatility Index
Uses of Transcranial Doppler
Limitations of Transcranial Doppler
134
134
135
135
135
135
138
Transcranial Sonography
139
Thermal Diffusion Flowmetry
139
Laser Doppler Flowmetry
139
Intra-Arterial 133Xenon
139
CT Perfusion
139
Xenon Enhanced CT
139
Positron Emission Tomography
140
Single Photon Emission Computed Tomography
140
Magnetic Resonance Imaging
140
Intracranial Pressure
Technology
Values
Pathophysiology
Waveform Analysis
Pressure–Volume Relationship
Pressure Reactivity Index
Indications for Intracranial Pressure Monitoring
140
140
141
141
141
142
142
142
Electroencephalogram
Recording
Normal EEG
Analysis
143
143
144
144
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00008-7
Electrocorticogram
Uses of Electroencephalogram
145
145
Evoked Potential Monitoring
Somatosensory Evoked Potentials
Variables Affecting Somatosensory Evoked
Potentials Recording
Uses
Limitations
Brain Stem Auditory Evoked Potential
Stimulus Characteristics
Normal Waveforms
Factors Affecting Brain Stem Auditory Evoked
Potentials
Uses
Visual Evoked Potential
Stimulus Characteristics
Normal Waveform
Variables Affecting Visual Evoked Potential
145
145
Motor Evoked Potentials
Changes Considered Significant
Uses
Complications
Contraindications
Limitations
149
150
150
150
150
150
Depth of Anesthesia
Bispectral Index
Uses
Limitations of Bispectral Index Monitoring
Spectral Entropy
Uses of Entropy
150
150
151
151
151
151
Cerebral Oxygenation Monitoring
152
Jugular Venous Oximetry
Technology
152
153
133
146
147
147
147
147
148
148
148
149
149
149
149
© 2017 Elsevier Inc. All rights reserved.
134
8. NEUROMONITORING
Normal Values
Indications
Limitations
Complications
Contraindications
153
153
154
154
154
Regional Cerebral Oximetry
Equipment
Normative Values (Based on INVOS Device)
Factors Influencing rSO2 Values
Indications
Limitations
154
155
156
156
156
156
Brain Tissue Oxygen Monitoring
Technology
Probe Placement
PbtO2 Values
156
156
157
157
O2 Reactivity Index
Uses
Limitations
Complications
157
157
157
158
Cerebral Microdialysis
Probe Placement
Uses
Limitations
158
158
158
158
Conclusion
159
References
159
INTRODUCTION
It is important to monitor continuously the organ that we want to see perform correctly at all the times. Brain is
a very complex organ to monitor, and in fact we can call the brain as an organ of organs. There are many structures
that have to be monitored separately and continuously, if we have to ensure the correct functioning of all those structures. However, with so many independent structures, it is impossible to monitor all of them continuously. Thus,
the neuromonitoring is always a challenging task and the information gained may not always transform into clinical utility. However, with so many modalities available, and in combination, we are able to gain some insight into
the pathophysiology of the brain. We are now able to use the neuromonitoring in clinically managing the patients.
CEREBRAL BLOOD FLOW
Transcranial Doppler
TCD is the most commonly used technique to measure the cerebral blood flow (CBF).
The TCD uses a principle of Doppler sound waves where the waves reflected from a moving object are at a higher
frequency than the origin frequency (Fig. 8.1). Similarly, reflected waves from the object moving away will be at a
FIGURE 8.1 Doppler principle.
IV. NEUROMONITORING
CEREBRAL BLOOD FLOW
135
lower frequency than the origin frequency. The ultrasound waves were used to measure the CBF velocity at basal
arteries. The RBCs in the vessels act as a moving object toward the probe or away from the probe.
Assumptions
1. TCD is measuring the blood flow velocity (FV) in the vessel and not the actual flow. However, we presume the
FV is equivalent to flow.
2. The diameter of the vessel remains constant. This is debatable but few studies have shown that vessel diameter
indeed remains constant in many conditions.2
3. The angle of insonation has to remain constant for comparing the measurements.
Technique
The technique commonly used is 2 MHz probe as the waves have to penetrate through the bone. Higher the frequency, lower is the capability to penetrate the bone. Most commonly used site is transpterional, where the bone
thickness is the least. It is just above the zygus, about 1–2 cm anterior to tragus. Keeping the probe perpendicular
to the skin and directed slightly anterior will frequently encounter the middle cerebral artery (MCA). The MCA is
identified with flow toward the probe at a depth of 50–65 mm and with characteristic flow sound (Fig. 8.2). From that
point, with slight manipulation, it is possible to trace back to internal carotid artery (ICA) bifurcation: anterior cerebral artery (ACA) and posterior cerebral artery. Transorbital (carotid artery) and suboccipital (basilar and vertebral
arteries) approaches are also used in some situations (Table 8.1).
Pulsatility Index
Pulsatility refers to peak systolic to lowest diastolic FV. With constant cerebral perfusion pressure (CPP), any
change in pulsatility reflects the change in the cerebrovascular resistance (CVR), i.e., higher the CVR, higher is the
pulsatility index (PI). The normal PI is 0.5–1.0, which is a dimensionless number.
0) &6SYS å &6DIA
&6MEAN
Uses of Transcranial Doppler
1. Subarachnoid hemorrhage (SAH): Commonly used to diagnose the vasospasm to quantify the degree of
vasospasm and its response to treatment. Any FV more than 120 cm/s with PI of more than 1.5 is considered
indicative of vasospasm (Table 8.2 & Fig. 8.3). Many centers use TCD regularly to monitor SAH patients.
FIGURE 8.2 Transcranial Doppler recording from middle cerebral artery (A) and anterior cerebral artery (B).
IV. NEUROMONITORING
136
8. NEUROMONITORING
TABLE 8.1 Normal Transcranial Doppler Parameters in Different Vessels
Artery
FV (cm/s)
Depth of Insonation (mm)
Direction of Flow
Effect of Ipsilateral Carotid
Compression
Middle cerebral artery
40–60
45–60
Toward
Decreases
Anterior cerebral artery
40–50
60–75
Away
Decreases
Posterior cerebral artery
30–45
60–75
Toward
No change
Carotid art (orbital)
35–50
60–80
Toward
Decreases
Basilar
35–50
60–80
Away
No change
Vertebral
30–45
80–110
Away
No change
TABLE 8.2 Transcranial Doppler Classification of the Severity of Vasospasm
Severity of
Vasospasm
MFV Value (cm/s)
MCA/ICA Ratio
Normal
<85
<3
Mild
<120
<3
Moderate
120–150
3–5.9
Severe
151–200
>6
Critical
>200
>6
ICA, anterior cerebral artery; MCA, middle cerebral artery.
From Kassab MY, Majid A, Farooq MU, Azhary H, Hershey LA, Bednarczyk EM, Graybeal DF,
Johnson MD. Transcranial Doppler: an introduction for primary care physicians. J Am Board Fam
Med 2007;20:65–71.
FIGURE 8.3 Middle cerebral artery in cerebral vasospasm with increased pulsatility index of 1.83 and peak FV of 92 cm/s.
2. Carotid endarterectomy (CEA): TCD is used to identify many things:
a. Ischemia: During cross clamping, FV decrease <40% of baseline is considered as ischemia. Simultaneous
monitoring of electroencephalogram (EEG) will help in better delineation of ischemia. Generally, surgeon
considers placement of shunt if the FV < 40%. There is a study further classified the decrease as mild (16–40%
of baseline value) and ≤15% as severe.3
b. Emboli: Detection of emboli is easy with TCD. Surgeon can modify their technique to decrease emboli.
Detection of emboli will also help in the prediction of postoperative cognitive deficits.4
IV. NEUROMONITORING
CEREBRAL BLOOD FLOW
137
c. Identification of the shunt malfunction.
d. Hyperemia: Postoperatively patient may develop sudden hyperemia due to vasoparalysis in the ischemic
area. This can lead to cerebral edema and hemorrhage. TCD can identify the patients early and thus,
preventive measures can be instituted.
e. Postoperative ischemia: Carotid occlusion at operative site is a lethal complication. TCD can help in
identifying these patients before total occlusion by identifying decrease in the FV.
3. Head Injury: It is useful in identifying cerebral vasospasm. Cerebral vasospasm develops in 20–30% of patients
with head injury. TCD is used to measure intracranial pressure (ICP) and CPP noninvasively.5 It is also used to
assess the presence or absence of autoregulation and carbon dioxide reactivity. It is also used to diagnose the
brain death where typical oscillatory flow is seen with intact skull (Fig. 8.4).
4. Other uses:
a. Cardiac surgery: TCD is used to detect emboli during cardiac surgery and also to measure the CBF during
cardiopulmonary bypass.
b. Hepatic encephalopathy: TCD is used to assess the ICP and CPP noninvasively because of bleeding risks with
invasive monitoring.
c. Eclampsia: TCD is used to assess the ICP and CPP noninvasively.
d. TCD is used in noninvasive assessment of CBF in diverse conditions.6,7
TCD is also used in:
1. Testing of pressure autoregulation: With intact autoregulation, any changes to the arterial pressure does not affect any
change in CBF (measured by FV with TCD). Both the static and dynamic autoregulation can be tested using TCD.8
a. Dynamic tests:
i. Dynamic autoregulation: Sudden hypotension is induced by deflation of a large thigh cuff [at least 20 mm Hg
drop in mean arterial pressure (MAP)]. The FV also drops immediately, but recovers within few seconds.
With continuous TCD monitoring this drop and recovery are mapped. This map will be compared with
standardized graphs and the degree of autoregulation would be said [autoregulatory index (ARI)].
Normally the return of autoregulation is complete within 5 s, i.e., dynamic rate of autoregulation (dRoR)
is 20%/s. This measurement has to be done within 10 s of deflation of thigh cuff to avoid confusion due to
carbon dioxide (CO2) changes.
ii. Transient hyperemic response test: This is the most commonly used test due to ease of testing. The baseline
FV is recorded and the carotid artery is compressed for 5–8 s and released. With the compression the FV
would decrease and after the release, FV would increase due to cerebral vascular dilation in response
to ischemia. This increase would return back to baseline within 5 s with intact autoregulation. If the
autoregulation is impaired, there would be no hyperemia as there would be no cerebral vasodilation
in response to ischemia (Fig. 8.5). The success of the test depends on the adequate compression of the
carotid artery.
b. Static tests: The MAP is raised by 20 mm Hg by vasopressor (phenylephrine) infusion. The change in
the FV would be minimal if the autoregulation is intact. The FV would increase if the autoregulation is
impaired.
FIGURE 8.4 Transcranial Doppler in brain death. Only little flow is seen entering the middle and anterior cerebral arteries (near carotid bifurcation) with each beat during systole.
IV. NEUROMONITORING
138
8. NEUROMONITORING
FIGURE 8.5 Transient hyperemic response test in a patient with normal autoregulation.
3TATIC RATE OF REGULATION (S2O2) ° %
#62E
-!0
where CVRe = MAP/FV; sRoR of “1” indicates perfect autoregulation and “0” indicates an absence of
autoregulation.
2. Cerebrovascular CO2 reactivity: With every mm Hg change in CO2, there will be 3–4% change in the CBF in the same
direction until limitation/saturation develops, i.e., within a range of 20–80 mm Hg CO2 levels. TCD FV can be
used instead of CBF measurements in assessing the change in CBF to change on CO2 levels. It is considered that
the diameter of basal arteries remains constant with the changes in CO2 levels. It is also assumed that only the
distal vessel diameter is altered with CO2.2 Therefore TCD is used to assess the CBF changes to CO2 levels, i.e.,
percentage of change in the FV to percentage of change in CO2 levels. T is commonly used in head-injured patients
to assess the CO2 reactivity9 and to prognosticate these patients.
3. Noninvasive assessment of ICP: One of the important uses of TCD is noninvasive assessment of ICP and CPP.
Many techniques of assessment have been described but still they have not achieved the perfection to be used in
clinical management of patients.
The estimated CPP by TCD is calculated using the formula
#00E -!0 ° &6D&6M
MM(G
It is calculated both the sides and averaged. Authors were able to achieve <15 mm Hg error in 92% (<10 mm Hg
error in 89%) of measurements.5
Initially, Aaslid et al.10 described
#00E &6M ° !&
(A1—amplitude of fundamental frequency component of arterial pressure, F1—amplitude of fundamental frequency component of FV.) Fundamental frequency is calculated by fast Fourier transformation of the waveform.
Other techniques are based on the following:
1. Increased pulsatility11
2. Decreased diastolic FV12
3. Decreased ratio of diastolic to mean FV13
Limitations of Transcranial Doppler
1. It is a blind procedure. The accuracy depends on the individual doing it.
2. In 5–10% of patients, insonation is not possible because of thick bone.
3. Difficult to detect the distal branches.
IV. NEUROMONITORING
XENON ENHANCED CT
139
TRANSCRANIAL SONOGRAPHY
Recently transcranial B-mode ultrasound is used to monitor brain parenchyma. Repeated measurements of ventricular size, midline shift, intracerebral hemorrhage size, optical nerve sheath diameter to monitor raised ICP is
possible. The technology is used in the early diagnosis of Parkinson disease, other movement disorders, sleep disorders, treatment of stroke, etc. Even though at present the utility is very limited, the technology holds great promise
to future.14–16
THERMAL DIFFUSION FLOWMETRY
The principle is that two sensors are placed nearby and one sensor is heated and the temperature is measured by the
other sensor. The temperature difference between the sensors is inversely proportional to the thermal conductivity of
the brain tissue between the sensors. There are many types of sensors and many techniques of heating and measuring
the temperature difference. The assumption is thermal conductivity is constant in all the individuals. The probe is about
1 mm thickness, which is placed deep into the brain tissue in the arterial territory of interest. It gives the absolute values
of CBF and almost instantaneous (1–2 s) and continuous measurement. Operative lights, irrigation of surgical fields,
febrile patients can cause problems in measuring. However, thermal diffusion flowmetry is slowly getting popular and
many feel that there is a greater role for this modality in monitoring the CBF in neurological patients.17
LASER DOPPLER FLOWMETRY
Laser Doppler flowmetry (LDF) measures the CBF noninvasively (with open skull), semi quantitatively, but continuously over the cortical surface. It detects the Doppler shift of laser light reflected from the RBCs in a small volume
of cortical tissue. It can be used to scan a large surface of cortical tissue. It has been used intraoperatively to detect
both ischemia and hyperemia.18,19
INTRA-ARTERIAL 133XENON
Kety–Schmidt technique using the Fick principle perfected the measurement of CBF. The method quantifies the
difference between cerebral washin and washout of freely diffusible inert gas (N2O) by serial measurements of arterial and jugular bulb blood concentrations of the tracer. Now it is well known that the N2O is not an inert gas, thus
133Xe is used. The tracer is injected into carotid artery and washout is recorded by multiple external scintillation
counters. The rate at which the tracer is washed out is proportional to CBF. With appropriate mathematical equations, gray and white matter blood flow calculation is possible.
Carotid puncture is not possible every time and thus, noninvasive techniques have been developed, i.e., inhalational 133Xe and intravenous 133Xe. These noninvasive techniques also provide reproducible results with a reasonable
spatial resolution.20
CT PERFUSION
The iodinated contrast is injected and simultaneously images are acquired using a helical CT multislice scanner in
a cine mode which allows for the measurement of CBF and cerebral blood volume (CBV). This technique is relatively
fast and can be done in most of the CT scanners. Clinically this method can be used to measure the perfusion of the
brain in many clinical scenarios, e.g., perfusion of the brain in severe traumatic brain injury (TBI) and hypoperfused
areas in the SAH patients.21
XENON ENHANCED CT
The technique involves inhalation of nonradioactive xenon and simultaneous acquisition of CT images. Similar to
intra-arterial xenon technique with modified Kety–Schmidt equation, CBF is calculated.
IV. NEUROMONITORING
140
8. NEUROMONITORING
POSITRON EMISSION TOMOGRAPHY
Both CBF and metabolism can be measured with positron emission tomography (PET) scan. Regional CBF,
regional CBV, regional oxygen extraction fraction (rOEF), and regional cerebral metabolic rate of oxygen
(rCMRO2) from the whole brain can be obtained. Kety–Schmidt technique is used to measure CBF. Resolution is
about 4–6 mm. However, it is not useful in emergency settings and used mainly in the research settings because
of high cost.
SINGLE PHOTON EMISSION COMPUTED TOMOGRAPHY
Single photon emission computed tomography is similar to 133Xe technique described earlier but is reconstructed in three dimensions with a rotating camera. Whole brain is covered and takes about 10–15 min for
the study. Absolute values are difficult to obtain. It has slightly less resolution but much cheaper than the PET
scan.
MAGNETIC RESONANCE IMAGING
Many magnetic resonance imaging (MRI) techniques measuring CBF have been developed. The most successful
approaches are dynamic tracking of a bolus of a paramagnetic contrast agent (dynamic susceptibility contrast) or on
arterial spin labeling. Whole brain is covered and anatomical localization is possible. Good resolution is possible but
absolute values are difficult to obtain.
INTRACRANIAL PRESSURE
It is defined as the pressure within cranial cavity relative to the atmospheric pressure.
Technology
ICP can be measured by either invasive or noninvasive techniques.
1. Invasive
a. Fluid-filled external pressure transducer: This is similar to arterial pressure and central venous pressure
monitoring. A catheter is inserted in to the lateral ventricle and is connected to the transducer (Wheatstone
bridge) through fluid-filled tubing.
b. Miniature strain gauge transducer (Codman): In this, the sensor is placed in the tip of the catheter. Changes in
pressures cause change in resistance of the circuit within the sensor and is interpreted as a waveform. Zeroing
has to be done preinsertion and once placed cannot be rezeroed in vivo. Hence, zero drifting can occur over a
period of days and results in false ICP values.
c. Fiberoptic (Camino): The sensor at the catheter tip uses a light source. Pressure changes cause change in the
light reflection and this is quantified as pressure change. Zeroing has to be done preinsertion and once placed
cannot be rezeroed in vivo.
d. Spielberg ICP system: In this system, a fluid-filled catheter has an air balloon pouch at the tip of the catheter. A
fluctuation in balloon pressure is interpreted as ICP change.
2. Noninvasive:
a. Tympanic membrane displacement.
b. TCD: Various formulae have been described to estimate CPP noninvasively (see the detailed discussion
earlier).
c. Optic nerve sheath diameter: Measurement is taken 3 mm behind the globe. It is qualitative. Diameter >6 mm
is highly indicative of raised ICP.
IV. NEUROMONITORING
141
INTRACRANIAL PRESSURE
As ICP measures the pressure inside the cranial cavity, the catheter can be placed in the epidural/subdural/
intraparenchymal or ventricular space (Table 8.3). Ventricular measurement is considered as gold standard. Epidural
(Richmond, Gaeltec) and subdural sites are less commonly used. For intraparenchymal monitoring (Codman,
Camino), a burr hole is placed separately or as a part of triple bolt system into the nondominant frontal region. The
tip is usually placed in to the white matter.
Values
Normal: 5–15 mm Hg (healthy adults, supine); 3–7 mm Hg (children); 1–5 mm Hg (infants). Cutoff values for
treating ICP depends on the intracranial pathology. For head injury, treatment is initiated when the ICP exceeds
20–25 mm Hg.
Pathophysiology
High ICP is an indicator of brain injury and also results in secondary brain injury by reducing CBF (Fig. 8.6).
Waveform Analysis
ICP is made up of three components: arterial vascular component, cerebrospinal fluid (CSF) circulatory component, and cerebral venous outflow component.22 Normal waves have three peaks: percussion (P1), tidal (P2), and
dicrotic (P3) (Fig. 8.7).
During reduced compliance, the P2 merges with P1 or exceeds P1.
TABLE 8.3 Different Sites of Intracranial Pressure Measurement
Site of Catheter Placement
Advantages
Disadvantages
Intraventricular
t S
imple, cost-effective
t D
ifficult placement in slitlike ventricles
t Z
ero calibration possible
t I nfection
t T
herapeutic—cerebrospinal fluid
t H
emorrhage—along catheter pathline,
withdrawal, antibiotic instillation, studying
intraventricular
pressure volume index curves
Intraparenchymal
t E
asier to place
t Z
ero drifting
t A
ccurate measurement
t E
xpensive
t L
ess complications including infections and t T
herapeutic options not possible
hemorrhage
FIGURE 8.6 High intracranial pressure (ICP) and the unfavorable consequences. CBF, cerebral blood flow; CPP, cerebral perfusion pressure.
IV. NEUROMONITORING
142
8. NEUROMONITORING
FIGURE 8.7 Intracranial pressure waveforms.
Three types of waveforms (Lundberg) has been reported:
t A waves (plateau waves): In patients with reduced intracranial compliance, systemic hypotension results in
cerebral vasodilation leading to increase in CBV and hence ICP. The ICP values reach up to 40 mm Hg and stays
there for 5–15 min. When the duration of plateau waves exceeds >30 min, there are high chances for cerebral
ischemia. Due to intact autoregulation, blood pressure rises that reverses the phenomenon. This condition
should always be treated to prevent cerebral ischemia and herniation syndromes.
t B waves: The frequency is around 0.5–2/min with amplitudes going up to 20–30 mm Hg. These waves indicate
vasomotor center instability due to low CPP or at the lower end of cerebral autoregulation.
t C waves: Frequency is around 4–8/min with amplitudes around 20 mm Hg. This wave has been documented in
healthy individuals.
Pressure–Volume Relationship
Pressure and volume relationship within the intracranial compartment is nonlinear. Intracranial compliance is
defined as the changes in volume for a given change in pressure. The inverse of compliance is called elastance and
the relationship is nonlinear. The slope of this relationship in the logarithmic scale is linear and is described in terms
of pressure–volume index (PVI). PVI is the volume required to change the ICP by 10-fold (PVI = ∆V/log 10 Po/pm).
Normal value is around 20–25 mL. This is calculated by withdrawal of around 2 mL of CSF and noting the pressure
change. This procedure is repeated multiple times with aspirating and injecting saline into the catheter, and the pressure changes noted. Reduced compliance occurs even before the ICP values go high. On the other hand, high PVI
values can be seen in patients with normal CPP but defective cerebral autoregulation. Caution should be exercised
when interpreting PVI inpatients whose CPP is below the autoregulatory threshold.
Pressure Reactivity Index
Pressure reactivity index (PRx) describes the changes in smooth muscle tone of the arterial walls in relation to the
changes in transmural pressure. It is calculated as a linear correlation coefficient between averaged ICP and arterial blood pressure over a 3–5 min period. Negative PRx index indicates good pressure reactivity index. The index
helps in identifying patients who might benefit from targeted CPP therapy as increasing CPP will reduce ICP only in
patients with intact vasomotor reactivity. CPP at which PRx is lowest is the optimal CPP.
Indications for Intracranial Pressure Monitoring
t Severe head injury with abnormal CT head scan
t Severe head injury with normal head CT with any of the two—age >40 years, systolic blood pressure <90 mm Hg,
and abnormal motor posturing
IV. NEUROMONITORING
ELECTROENCEPHALOGRAM
143
t Systemic diseases causing raised ICP, e.g., Rye’s syndrome, hepatic failure
t In patients with head injury where clinical neurological examination is not possible for prolonged periods of
time (e.g., patients undergoing general surgery)
t In intracerebral and SAH
t Malignant cerebral infarction
t Hydrocephalus, meningitis
ELECTROENCEPHALOGRAM
The EEG is a recording of spontaneous electrical activity of the cerebral cortex and is recorded from the surface of
scalp. EEG is a continuous, noninvasive indicator of cerebral function, even when the patient’s consciousness is not
adequate. It is a summation of excitatory (EPSP) and inhibitory (IPSP) postsynaptic potentials produced from the
pyramidal cells of the cerebral cortex.
Recording
EEG is recorded from the cup electrodes or needle electrodes from the scalp. Meticulous cleaning of the area is
important to remove the oil and dead cells from the surface. Thus the electrical resistance (impedance) is decreased
and signal conduction is increased. A pair of electrodes is called as a montage. The montage can be bipolar or referential. In the bipolar montage both the electrodes are active (i.e., electrodes lie on the underlying cerebral cortex and
are capable of recording the electrical activity of the cortex), and the voltage difference between the two electrodes is
recorded. In referential montage only one electrode is active and the other one is common referential electrode which
is commonly on the mastoid, ear lobe, or the shoulder. The focal lesions/changes are better picked up by the bipolar
and diffuse changes are better picked up by referential (unipolar) montages. Intraoperative monitoring is more commonly done with bipolar montage.
The 10–20 system of electrode placement is commonly employed. It is 10% or 20% of the nasion–inion line or
preauricular line or hemi-circumference line. The midline electrodes are designated as “z” and the right-sided electrodes as even numbers and left-sided electrodes as odd numbers. There are frontal prominence, frontal, central,
parietal, and occipital electrodes. Artifacts are common during EEG recording. Many filters are used to decrease
these artifacts. However, most of the filters are frequency filters which will filter low-frequency and high-frequency
waveforms. Artifacts within clinically useful range of frequency are difficult to eliminate [electromyogram (EMG),
blinking, motion, cautery, etc.] (Fig. 8.8).
FIGURE 8.8 Electrode placement for electroencephalogram (EEG) recording in 10–20 system (A) and morphology of EEG waveforms (B).
IV. NEUROMONITORING
144
8. NEUROMONITORING
Normal EEG
It is a plot of voltage versus time of 1–35 Hz frequency. Based on the frequency, the waveforms are classified into
different bands. Normally higher is the frequency, lower is the amplitude (Table 8.4).
Analysis
The raw EEG is looked for frequency, amplitude, sleep spindles, spikes, spike and wave pattern, burst suppression,
etc. and changes occurring in them. As the waveform is different in each individual and also from time to time in the
same individual, the analysis requires training and dedicated person to look for any changes happening intraoperatively.
Therefore, the automated analysis has come into vogue, and the anesthesiologists prefer them. Synchronized EEG activity
with higher amplitude is seen during sleep or general anesthesia. However, deep anesthesia produces burst suppression.
Awakening produces desyncronization with higher frequency and lower amplitude. Normally, higher the complexity of
the function, higher will be the frequency and lower will be the amplitude with lesser synchronization.
Initial step in automated analysis is breaking the EEG waveform into small segments called as “epoch.” The epoch
length varies from 2 to 16 s. Each epoch is scanned for the presence of artifacts and any epoch with artifact is rejected.
Commonest analysis is power spectrum analysis. The waveform is transformed into frequency components by fast
Fourier analysis. The amplitude at each frequency is noted and the power is calculated (amplitude2). The power is
displayed as a compressed spectral array (CSA) or density spectral array (DSA).
The CSA is obtained by creating a histogram of power at each frequency. By smoothening of the histogram a nice
line of the power versus frequency in the epoch is obtained. By stacking up these power lines we can see the changes
happening over a long duration and thus create a z axis (time). The same can be displayed in a two-dimensional form
as DSA, i.e., the time as x axis, frequency as y axis, and the power as density. In the same analysis we can measure
median frequency, spectral edge frequency. Also various ratios can be calculated, e.g., beta ratio, alpha/delta ratio
(ADR, 8–13 Hz/1–4 Hz), mean amplitude, total power (TP, 0–23 Hz), burst suppression percent, etc. with specific utility.23,24 Further advanced analysis includes coherence/bispectral analysis. This analysis measures the phasic relationship between two or more frequencies in a single channel or two channels25 (Fig. 8.9).
TABLE 8.4 Electroencephalogram Classification and Characteristics
Waves
Frequency (Hz)
Normal Amplitude (µV)
Characteristics
Beta (β)
>13
20
With mental activity, mainly frontal
Alpha (α)
8–12.5
40–100
Adults with eyes closed, mainly seen in the occipital
Theta (θ)
4–7.5
>50
Sedation/anesthesia/sleep
Delta (δ)
0.5–3.5
>50
Sedation/anesthesia/sleep/ischemia
FIGURE 8.9 Intraoperative electroencephalogram (EEG) monitoring showing various measured parameters (A) and compressed spectral
array (B).
IV. NEUROMONITORING
EVOKED POTENTIAL MONITORING
145
FIGURE 8.10 Electrocorticogram: strip electrode with four electrodes placed on the gyrus adjacent to the lesion on the brain surface.
Electrocorticogram
Recording of the spontaneous electrical activity of the brain from the cortical surface or depth of the brain is called
electrocorticogram. The waveforms are better localized and better delineated with electrocorticogram. It is recorded
intraoperatively to delineate the seizure focus and area of surgical resection. Sometimes a grid electrode is placed
on the cortical surface by the surgery, and electrocorticogram is recorded postoperatively to locate the seizure focus
(Fig. 8.10).
Uses of Electroencephalogram
EEG is a reflection of cortical neuronal function. It can be used wherever the cortical neuronal function has to be
monitored continuously. EEG is used with the intension of picking up cerebral ischemia before the cellular structure
is damaged and to identify the seizures. The EEG shows abnormal changes at CBF of less than 20 mL/100 g/min
much before the level at which cellular integrity is threatened (12 mL/100 g/min).26
CEA: Used to detect cerebral ischemia during cross clamping.
SAH: Used to detect cerebral ischemia during temporary occlusion of the vessel during dissection of the aneurysm. The quantitative EEG has also been used for early prediction of delayed cerebral ischemia.27
Cerebral protection: In surgeries where severe metabolic suppression is required for cerebral protection, EEG is used
to titrate the drugs.
Intensive care unit: EEG is used to titrate sedation, detect subclinical seizures, monitor the adequacy of metabolic
suppression. EEG can also be used to assess the degree of anoxic damage and to prognosticate the ICU patients.28,29
EVOKED POTENTIAL MONITORING
Evoked potentials are recordings of electrical activity of specific neuronal pathways in response to external stimulus. The recordings involve either the sensory pathway or motor pathway. When neural tissues are stimulated, the
electrical activity ascends along specific neuronal pathway. Based on stimulation site and recording location, characteristic waveform of the traveling impulses can be recorded.
t Sensory evoked potentials
t Somatosensory evoked potentials (SSEP)
t Brain stem auditory evoked potentials (BAEP)
t Visual evoked potentials (VEP)
t Motor evoked potentials
Somatosensory Evoked Potentials
Following peripheral stimulation, electrical activity travels along the posterior column (proprioception, mechanoception) and spinothalamic pathways (nociception, thermal). The pathways are supplied by the posterior spinal
artery. At the level of brain stem, it is supplied by the vertebral arteries and perforators of the basilar artery. At
the level of sensory cortex, vascular supply is from ACA (lower limb) and MCA (upper limb, face and trunk). The
IV. NEUROMONITORING
146
8. NEUROMONITORING
electrical activity is recorded, along the sensory pathways via the dorsal column in the spinal cord and the scalp
overlying the sensory cortex.
The stimulation site should be below the area at risk of ischemia from surgery and the recording site should be
above, so that the neuronal pathway should travel through the area at risk from surgery30 (Table 8.5).
Variables Affecting Somatosensory Evoked Potentials Recording
Anesthetic agents: As anesthetics act predominantly on the synapses and to some extent on the axonal conduction,
cortical potentials are more affected than the subcortical potentials.
Volatile: Halogenated compounds produce dose-dependent increase in latency and decrease in amplitude. This
effect is pronounced with the addition of nitrous oxide. Nitrous oxide per se can delay latency and depress amplitude
of SSEP.
Intravenous: At low doses, barbiturates and propofol cause minimal changes in SSEP. At higher doses, their cause
delayed latency and suppressed amplitude. Etomidate and ketamine augment amplitude of SSEP. Opioids when
given as a bolus cause depression, while low-dose continuous infusion causes minimal changes. Alpha agonists are
compatible with intraoperative SSEP recordings. Benzodiazepines have minimal depressant effects. Neuromuscular
blocking agents directly do not affect SSEP.
Temperature: Mild hypothermia produces increased latency of cortical SSEP. With profound hypothermia, cortical
potentials disappear followed by increased latency and reduced amplitude of subcortical potentials, finally leading
to disappearance of potentials.
Hemodynamics: Perfusion of the neuronal pathways can affect the SSEP recordings. Ischemia can result in prolonged latency and reduced amplitude. Severe anemia can affect cortical latency and amplitude. Increased ICP can
reduce amplitude and prolong latency of cortical SSEP.
Ventilation: Severe hypoxemia results in changes in amplitude of cortical SSEP. Hypercapnia has no effect on SSEP.
Though hypocapnia augments amplitude and reduce latency of SSEP in awake volunteers, these effects are not seen
in anesthetized patients.
Changes are considered significant in the following conditions:
t latency: prolongation by ≥10%
t amplitude: decrease by ≥50%
Values are lower if the initial waveforms are of low signal quality (Fig. 8.11).
TABLE 8.5 Somatosensory Evoked Potential Monitoring Parameters
Nerves stimulated
Upper limb: median, ulnar
Lower limb: posterior tibial
Stimulus specifications
Square wave pulses
Intensity: 20–35 mA (for lower limb up to 50 mA)
Duration: 200–300 ms
Rate: 2–7 Hz
Average: 250–500 stimulus
Filters: 2 Hz–3 KHz
Recording areas
(Upper limb stimulation): Erb’s point, C5, C2, C3’/4′
(Lower limb stimulation): T12, C5, C2, Cpz
Electrodes
Stimulating: skin/subdermal needles
Recording: skin, subdermal needles
Normal waveforms (neural
generators)
Upper limb:
N9: Erb’s point
N13: dorsal column (cervical spine region)
N18—thalamus
N20, P22: cortex origin
Central conduction time (N13–N20): 5–8 ms
IV. NEUROMONITORING
Lower limb:
Lumbar potentials: depending on level of
recording
N30: dorsal column
N37: cortex origin
EVOKED POTENTIAL MONITORING
147
FIGURE 8.11 Somatosensory evoked potential following right median nerve stimulation.
Uses
t For spinal procedures: intramedullary tumors, scoliosis surgery, spine stabilization
t For cerebral vascular surgeries where the specific pathways are affected by ischemia:
t Intracranial aneurysm (median/ulnar nerve for MCA territory; posterior tibial nerve for ACA territory)
t CEA
t In tumor surgeries: to locate the junction of sensory and motor cortex (phase reversal)
t To identify patient position related nerve injury (ulnar, median)
Limitations
t Does not monitor spinal cord supplied by the anterior spinal artery. A normal intraoperative SSEP does not rule
out occurrence of postoperative paraplegia.
t Artifacts by OT table, cautery machine, convection warmer, anesthesia machine, drill, CUSA, etc. can pose
problems in SSEP recording.
Brain Stem Auditory Evoked Potential
BAEP is the recording of the activities of the neural generators involved in the auditory pathways following stimulation of the cochlear nerve. The first 10 ms recordings represent the activities up to the brain stem and are termed
early latency; up to 80 ms represent cortical activities and are termed middle latency; beyond 80 ms, waveforms represent the cortical association areas and are termed late latency potentials.
Following stimulation of the ear, cochlea gets activated. The electrical impulses travel along the vestibulocochlear
nerve to the brain stem, midbrain and reach the primary auditory cortex. Auditory stimuli have bilateral representation in the cortex.
Stimulus Characteristics
Filter: 30 Hz–3 KHz
Amplitude: 0.3–3 microV
Stimulus averaging: around 2000
Frequency: 10–40 Hz
Intensity: click, square wave electrical pulse, 100–110 dBpeSPL (decibel peak equivalent sound pressure
level)
IV. NEUROMONITORING
148
8. NEUROMONITORING
Normal Waveforms (Fig. 8.12)
Waves I–VII: early latency BAEP
Interpeak latency: I–III: 2.1 ms
III–V: 1.9 ms
Changes considered significant: V waveform: latency prolongation >0.5 ms; 50% decrease in amplitude; prolonged
interpeak latency III–V (Table 8.6).
Factors Affecting Brain Stem Auditory Evoked Potentials
Early latency BAEP are resistant to the anesthetic effects. Use of drill during surgeries can interfere with the auditory stimuli and waveforms get distorted. Damage to the cochlear vessel (or vasospasm) will abolish all the waveforms and will not be helpful in brain stem monitoring. Mid latency potential are very sensitive to the anesthetic
agents and are used as a depth of anesthesia monitor.
Uses
t Surgeries at the cerebellopontine angle
t Brain stem procedures
t Posterior circulation vascular surgeries
FIGURE 8.12 Early latency brain stem auditory evoked potential (BAEP) response depicting five waveforms (A) and middle latency BAEP
response (B).
TABLE 8.6 Generators of Brain Stem Auditory Evoked Potential Waveforms
Waves
Latency (ms)
Origin
I
1.7
Cochlear nerve
II
2.8
Cochlear nucleus
III
3.9
Superior olivary complex
IV
5.1
Lateral lemniscus
V
5.7
Inferior colliculus
VI
Medial geniculate body, auditory cortex
VII
Cortex
Middle latency
No, Po, Na, Pa, Nb, N1
Pa <12 ms, Nb < 44.5 ms (indicate awareness during anesthesia)
Late latency
P2, N2
IV. NEUROMONITORING
149
MOTOR EVOKED POTENTIALS
(A)
(B)
Nasion
Nasion
FPZ
FPZ
Ground
A1
CZ
Ground
A2
A1
CZ
O1
A2
O2
Inion
FIGURE 8.13 Electrode placement for recording brain stem auditory evoked potential response (A) and visual evoked potential response (B).
t Surgeries in the posterior fossa
t Microvascular decompression of the lower cranial nerves.
Visual Evoked Potential
VEP records the electrical activity from neural generators along the visual pathway. Following light stimuli, retinal
receptors get activated. The electrical activity travels along the optic nerve, optic chiasm, optic tracts, lateral geniculate body and reach visual projection areas in the occipital cortex (Fig. 8.13).
Stimulus Characteristics
Light source: white or red LED-loaded goggles; 500–2000 lumens
Stimulus rate: 1–2.5 Hz (for steady state responses: 8–30 Hz)
Filter: 5–100 Hz
Signal averaging: 50–200
Recording electrodes: Oz–Fz; Oz–A1; Oz–A2; Ground Cz
Normal Waveform
t Triphasic waveform response
t P40 (lateral geniculate body)
t N70 (striatum)
t P100 (areas 17–19 of visual cortex in occipital lobe)
Variables Affecting Visual Evoked Potential
Halogenated compounds depress VEP to a greater extent. Low-dose opioids, benzodiazepines, and low-dose
propofol infusion do not affect VEP responses. Opioids cause pupillary constriction resulting in reduced light
transmission into the retina, and this can reduce VEP response. It is better to use mydriatics at the beginning of
surgery. Reduced core temperature results in prolonged latency of VEP (20% at 35°C).
MOTOR EVOKED POTENTIALS
Following stimulation (electrical or magnetic) of the motor cortex, electrical impulses travel along the cortical
spinal tract, descend at the level of brain stem, travel down to the anterior funiculi of the spinal cord, and result in
muscle activity (Table 8.7).
IV. NEUROMONITORING
150
8. NEUROMONITORING
TABLE 8.7 Stimulus Characteristics for Motor Evoked Potential
Myogenic
D Waves
Intensity
200–1000 V
100–500 V
Amplitude
24–2000 microV
1–30 mmicroV
Filter
10–5 KHz
10–2 KHz
Duration
0.05–1 ms (usually 0.2–0.5 ms)
Rate
Trains of 4–8 with interstimulus latency 2–4 ms
Stimulus site
C1–C2 or C3–C4
Recording site
Muscle: abductor hallucis, gastrocnemius, abductor pollicis, tibialis anterior
Spinal cord (epidural/subdural)
Changes Considered Significant
The dictum is that unchanged stimulus parameters should produce similar responses in a particular group of
muscles (provided there is no change in anesthetic depth and no use of muscle relaxants). So, changes are considered
significant when there is:
t an increased requirement of stimulus strength (>50 V) to produce the same initial response
t increased number of stimuli to achieve the same initial response
t decrease in amplitude by more than 80%.
Uses
t
t
t
t
For spinal procedures as a supplement to SSEP, e.g., scoliosis correction surgery, intramedullary tumors
Spinal decompression procedures
Intracranial tumors where motor cortex is involved
To map motor cortex when tumor is situated close to the motor cortex (stimulus strength required is minimal
when cortex is directly stimulated: 2–10 mA).
Complications
t Tongue injury
t Bone fractures including mandible
t Patient fall from OT table
Contraindications
t Patients with implantable deep brain stimulator, clips
t Recent craniotomy (relative)—possibility of stimulating needle penetrating brain through skull defect
t Cardiac pacemaker (arrhythmias)
Limitations
t Not sensitive to individual root injury
t Highly sensitive to anesthetic agents
DEPTH OF ANESTHESIA
Bispectral Index
Most commonly used and extensively studied monitor to assess depth of anesthesia. BIS examines the relationship/synchronization among the waves. The monitor utilizes various subparameters in time domain, frequency
domain, and bispectral domain.25
IV. NEUROMONITORING
151
DEPTH OF ANESTHESIA
&REQUENCY DOMAIN "ETA RATIO ,OG
"ISPECTRAL $OMAIN 3YNCH &AST 3LOW (P å (Z)
(P å (Z)
(3UM OF ALL BISPECTRUM PEAKS (. å (Z))
(3UM OF BISPECTRUM IN THE AREA ( å (Z))
Time domain: Burst suppression ratio and QUAZI—both are burst suppression calculated by different
algorithm.
All the above parameters are fitted in an equation with different weightings to obtain a dimensionless number
called BIS. Also, the given weighting to a parameter is altered according to the patient’s consciousness level. The values range from 100 (awake) to 0 (isoelectric EEG). As the patient is sedated the BIS value drops to <90 and the values
of 60 to 40 are considered adequate hypnosis component of general anesthesia. A value of less than 40 is considered
as deeply anesthetized (Fig. 8.14).
Uses
The BIS monitoring was developed to assess the depth of hypnosis during general anesthesia and to prevent the
awareness under anesthesia. Later, it was used to assess adequacy of sedation in various clinical situations, to assess
and to monitor the depth of coma. BIS is used to detect cerebral ischemia and in the diagnosis of brain death. The
indications for BIS monitoring during diagnostic and therapeutic procedures are ever increasing.
Limitations of Bispectral Index Monitoring
In general, all the factors that affect the EEG can cause inaccuracy in the BIS measurement even though the technological advances are decreasing the inaccuracies.
Ketamine: May increase BIS due to EEG activation.
Nitrous oxide: Seen to decrease the BIS values when used with other general anesthetic agents. But when used
as a sole agent it does not decrease the BIS values.
Opioids: They decrease the BIS values to some extent but not to levels suggestive of general anesthesia.31
Serious medical conditions: Cardiac arrest, hypovolemia, hypotension, cerebral ischemia/hypoperfusion,
hypoglycemia, hypothermia have shown very low BIS values presumably due to decreased metabolism.
Neurological disorders: Disorders having EEG changes and metabolic suppression would show abnormal
BIS values, e.g., severe brain injury, postictal state, dementia, cerebral palsy, etc. Patients who have received
electroconvulsive therapy have shown low BIS values even much later than postictal phase.32
Different anesthetic agents may have different BIS values: It has been shown that decrease in BIS values with
halothane is much less in comparison with isoflurane. There is a mild difference between isoflurane and
propofol also.31,33
Technical: It is important to remember that the value displayed is not real time. There is a 10–15 s delay. EMG
activity of the frontalis muscle and electrocautery can produce artifacts.
Spectral Entropy
This mode of assessing depth of anesthesia was introduced by the Datex company. Entropy means randomness. The principle in measuring entropy is that as the depth of anesthesia increases the randomness in the EEG
comes down and rhythmic oscillations and predictability increases. The EEG and EMG are recorded from the sensor
attached to forehead. It calculates two parameters: state entropy (SE) and response entropy (RE). SE is calculated
from the 0.8 to 32 Hz frequency and RE is calculated from 0.8 to 47 Hz. SE is a more stable number and ranges from 0
to 91 (isoelectric EEG to awake state). For the calculation of RE, the EMG activity is also taken into calculation. Thus,
in entropy assessment, the EMG signal is not discarded. The RE ranges from 0 to 100 and for anesthesia, the recommended range for both is 40–60. The RE is always equal to or higher than the SE.
Uses of Entropy
The entropies are similar to BIS monitoring, although few studies suggest that response to pain is better detected
by entropy (RE) as the EMG signal is included into the analysis.
Other depth of anaesthesia monitors are:
IV. NEUROMONITORING
152
8. NEUROMONITORING
FIGURE 8.14 Bispectral index (BIS) monitor. BIS value, electroencephalogram, signal quality index, electromyogram, burst suppression ratio,
(SR) and trend graph are seen.
Narcotrend
Patient state analyzer
Index of consciousness
Auditory evoked potential index (ARx index)
CEREBRAL OXYGENATION MONITORING
Brain is highly dependent on oxygen as its fuel. The purpose of monitoring is to identify ischemia at an early
(reversible) stage, so that interventions can be planned. When the oxygen supply is less than consumption, initially
oxygen extraction increases (resulting in fall in venous oxygen level) and when the supply falls below the critical
level, ischemia sets in. Monitors of cerebral oxygenation include jugular venous oximetry, cerebral oximetry, and
brain tissue oxygen.
JUGULAR VENOUS OXIMETRY
Jugular venous oximetry (SjvO2) measures the oxygen saturation of the venous blood at jugular bulb. SjvO2 represents the global oxygen extraction of the cerebral tissues. Oxygen delivery to the cerebral tissues is dependent on CBF
and arterial oxygen content. CBF is dependent on MAP, CVR, and cerebral autoregulation. Arterial oxygen content
is dependent on hemoglobin, saturation, arterial partial pressure of oxygen. Cerebral oxygen consumption (CMRO2)
is calculated as a product of CBF and arteriovenous oxygen difference (A-VO2).
CMRO2 = CBF × A-VO2
A-VO2 = Hb × 1.39 (SaO2 − SjvO2) + 0.003 × (PaO2 − PjvO2)
Considering Hb to be a constant, dissolved oxygen to be negligent (except in hypothermia), and SaO2 equivalent
to 1, the equation can be rewritten as
A-VO2 α 1 − SjvO2
CMRO2/CBF α 1 − SjvO2
The above equation shows that SjvO2 is inversely proportional to CMRO2 and A-VO2 and has direct relationship with CBF. In healthy situations, when there is increased demand (CMRO2), CBF increases by autoregulatory
IV. NEUROMONITORING
JUGULAR VENOUS OXIMETRY
153
mechanisms thereby keeping arteriovenous oxygen difference (oxygen extraction) a constant. In brain injury,
there is defective autoregulation and CBF does not compensate demand resulting in increased oxygen extraction
leading to decreased SjvO2. Thus, reduced SjvO2 indicates imbalance in supply–demand ratio irrespective of
the cause. On the other hand, high SjvO2 indicates reduced consumption either because of infarct or hyperemia.
Technology
The dominant jugular vein is cannulated retrograde and catheter tip is placed at the jugular bulb. Correct
position is confirmed by X-ray of the skull. In lateral X-ray skull view, catheter tip should lie cranial to C1–C2
interspace. In AP X-ray skull view, catheter tip should lie cranial to the line joining the two mastoid processes
and caudal to the lower margin of the orbit. The dominant side of the jugular vein is identified by the size of the
jugular foramen in CT head and ICP response to jugular vein compression. If the catheter tip lies within 2.5 cm
of the jugular bulb, the chances for extracerebral contamination is minimal.34 A standard central venous catheter
can be placed and blood can be sampled intermittently or dedicated fiberoptic catheters can be placed, which give
continuous measurements.
Normal Values
A-V O2: 2.2–3.3 µmol/mL (5–7.5 vol%)
SjvO2: 60–75%
Right to left side difference ranges from 5% to 15% (Tables 8.8 and 8.9).
Indications
t Head injury patients to identify secondary neuronal injury.
t SAH patients, to differentiate vasospasm from hyperemia (TCD shows high velocity in both conditions).
t To titrate hyperventilation therapy (2 tier therapy to manage intracranial hypertension).
TABLE 8.8 Classification of SjvO2 Values.
Value (in %)
Conditions
>90
Brain death, severe hypothermia, arteriovenous malformations (AVM)
75–90
Hyperemia, AVM
60–75
Normal
50–60
Increased O2 extraction
45–50
Mild–moderate cerebral ischemia
<45
Severe ischemia, anaerobic metabolism associated with EEG changes
TABLE 8.9 Factors Affecting SjvO2 Values
Low SjvO2 (<50%)
High SjvO2 (>75%)
t D
ecreased O2 supply:
Decreased cerebrospinal fluid (low cerebral perfusion pressure,
high intracranial pressure)
Low cardiac output
Anemia
Hyperventilation
Low PaO2, low FIO2
t I ncreased demand
Hyperthermia
Seizures
t I ncreased supply
Arteriovenous malformations
Cerebral hyperemia
Hypercapnia
Bohr effect (pH > 7.6, oxygen dissociation curve shift to left prevents
dissociation of O2)
Cerebral infarction
Brain death
t R
educed demand
Hypothermia
IV. NEUROMONITORING
154
8. NEUROMONITORING
Limitations
t It is a global indicator of oxygen imbalance and focal ischemic changes can be readily missed out.
t The amount of venous drainage into each jugular vein is unknown. The amount of contralateral contamination
can result in abnormal values.
t Low values indicate only oxygen imbalance status, not the cause for the imbalance. Other systemic monitors are
required to identify possible causes.
Complications
t Injury to brachial plexus and carotid artery during catheter insertion.
t Infection
t Jugular vein thrombosis (possibly raised ICP)
Contraindications
t Bleeding diathesis
t Cervical spine injuries
REGIONAL CEREBRAL OXIMETRY
Cerebral oximetry noninvasively and continuously measures the regional cerebral tissue oxygen saturation (rSO2)
by near-infrared (NIR) spectroscopic technique.
NIR light has the ability to penetrate tissue including bone. Oxygen binding to the hemoglobin affects the absorption spectrum. By measuring light absorption at two or more wavelengths, it is possible to measure the concentration
of oxy and deoxyhemoglobin. The peak absorption for reduced hemoglobin is around 740 nm, for oxy hemoglobin it
is around 850–1000 nm and at 810 nm, the absorption spectrum of oxy and deoxyhemoglobin are similar (isosbestic
point). The principle behind the NIR technology is based on the Lambert Beer equation. This equation calculates the
absolute concentration of the chromophore provided the sampling volume and the path length of the light source
are known. In biological tissues such as the skull, light (photons) has the tendency to scatter resulting in increased
and variable path length and some photons do not reach the detector. To overcome this, different types of reflectance
spectroscopic techniques are employed.36
Differential spectroscopy: In this technique, path length and the amount of scattered light are assumed to be
constant. The monitor is useful as a trend monitor starting from an arbitrary baseline value. Currently available
commercial monitors do not employ this technique.
Spatially resolved spectroscopy (Invos, Equanox): The light (from diodes) reflecting from the extracranial and
intracranial tissues is differentiated by using two detectors. The depth of penetration and reflection of photons
are dependent on the distance between the light emitter and the detector. The detector (silicone photodiode/
photomultiplier tube) closer to the diode (shallow) receives photons from extracranial tissues, and the detector
which is farther (deeper) receives signals from the intracranial tissues (Figs. 8.15 and 8.16).
Frequency-resolved spectroscopy (Oxiplex): Light intensity with known radio frequency is used as the source.
This technique measures the absolute chromophore concentration. Multiple wavelength technology facilitates
measurements of other chromophores such as cytochrome a–a3 and with dyes, such as indocyanine green,
facilitates CBV measurements.
For a given cortical tissue, cerebral vascular bed is made of 70–85% venous and capillary and the remaining arterial. The measurement made by the cerebral oximetry predominantly reflects the concentration/saturation of hemoglobin in the venous and capillary bed. Cerebral oximetry provides a continuous, bedside, noninvasive measurement
for focal oxygenation status of the brain tissues. Any intervention to improve oxygen delivery can be assessed by the
corresponding changes in the rSO2 values.
The sensor is usually placed over the forehead on either side of the midline few centimeters above the eye brow
to avoid contamination by the sagittal sinus and the frontal sinus. It is better to have a look at the X-ray skull and CT
head to have knowledge on the extent of the frontal sinus before placing the sensors. The sensors must be protected
IV. NEUROMONITORING
REGIONAL CEREBRAL OXIMETRY
155
from external light source to prevent contamination. Perspiration can result in displacement of the sensors. Cerebral
oximeters are generally resistant to motion artifacts and cautery. The pressure on the forehead by the sensors might
cause damage to the skin.
Equipment
1.
2.
3.
4.
5.
6.
7.
8.
Invos 5100 (Somenetics corporation) (735- and 810 nm-light source)
CAS Medical Fore sight (Branford, CT)
CerOx (OrNim Inc. LosGatos, CA)
EquanoX 7600 Nonin Inc. (Plymouth, MN)
NIRO 300 (Hamamastsu Photonics, Japan)
Oxymon (Ortinis, the Netherlands)
OxiplexTS (ISS, IL)
TRS-20 (Hamamatsu, Japan)
FIGURE 8.15 Equanox cerebral oximetry monitor (Nonin Inc.) and Equanox sensor. It contains two LED light sources on either end of the
sensor. There are two detectors: 2 and 4 cm away from the light source on either side.
FIGURE 8.16 Diagram representing light pathway into brain tissue.
IV. NEUROMONITORING
156
8. NEUROMONITORING
Normative Values (Based on INVOS Device)
Normative values differ between different equipments. The values given below are from the INVOS device.
Adult: 71 ± 6%
Children: 71 ± 7%
Neonate: 76 ± 8%
Normative values are generally greater than jugular venous saturation. The values are independent for hematocrit
values greater than 30%. The value is age dependent. Right to left differences is generally less than 10% in majority
of the patients. Values less than 40–50% or fall >20–25% of the baseline values are generally considered as cutoff for
development of cerebral ischemia.36
Factors Influencing rSO2 Values
Blood pressure
Systemic oxygenation
Hemoglobin
Arterial PO2, PCO2, pH
Temperature
Anesthesia
Seizure
Indications
t
t
t
t
t
Surgeries such as CEA, cardiac surgeries
Neuroradiology suite during balloon occlusion testing of ICA
ICU, to titrate blood pressure in patients with shock
Detect cerebral vasospasm following SAH
During surgery in elderly patients under general anesthesia. Maintaining normal rSO2 values reduces the
incidence of perioperative strokes and cognitive dysfunction
Limitations
t Presence of abnormal hemoglobins and bilirubin can give erroneous rSO2 values.
t Presence of extracranial collection of CSF or blood can give inaccurate results (e.g., extradural hematoma). At the
same time the abnormal values on one side can give a clue to the presence of abnormal collection and facilitate
surgical decision.
t Placement over the infarcted brain tissue (frontal cortex) can give falsely high values.
t In patients who have undergone surgeries such as decompressive craniectomy, the light is excessively reflected
and the values are unreliable.
t rSO2 measurement (regional) is restricted to the focal prefrontal cortex and infarct on other areas can be easily missed.
BRAIN TISSUE OXYGEN MONITORING
This technique measures (focal) oxygen tension at the brain’s interstitial space. It represents the oxygen available
for mitochondrial oxidative phosphorylation. Brain tissue oxygen monitoring (PbtO2) is determined by the CBF and
the difference in cerebral arterial and venous oxygen tension.
Technology
PbtO2 is measured by two different techniques:
1. Modified Clark electrode (e.g., Licox, Neurovent-P)
2. Optical fluorescence (e.g., Neurotrend, Codman, Oxylab)
In Clark electrode, a membrane surrounds an electrolyte layer and two electrodes made of noble metal. Oxygen
diffuses in to the membrane, electrochemically gets reduced, and causes a voltage difference which is proportional to
IV. NEUROMONITORING
BRAIN TISSUE OXYGEN MONITORING
157
the oxygen tension. This process is temperature dependent, and brain temperature must be simultaneously measured
for PbtO2 calibration. Brain area monitored by Licox probe is around 14–18 mm2 while with that of Neurovent-P is
around 24 mm2. Hence, values by different probes cannot be compared. Clark-type electrodes are precalibrated and
following insertion requires at least an hour for stabilization before actual measurements take place.
In optical fluorescence technology, optode sensors are coated with colored markers that emit color when
comes in contact with oxygen. The intensity of color depends upon the oxygen tension and is picked up by
fluorescent sensors. However, this probe requires calibration with the known oxygen concentration prior to its
insertion.
Probe Placement
PbtO2 probes are fine catheters, approximately 0.5–0.8 mm in diameter, placed in to the white matter close (roughly
3.5 cm below the dura) to the injured brain, preferably in the penumbral region. In diffuse injury, the probe is usually
placed in the nondominant frontal subcortical white matter. The position of the probe must be confirmed with the CT
head. If brain temperature is not monitored, the probe must be calibrated with body temperature at regular intervals.
PbtO2 Values
PbtO2 is a focal measurement. In diffuse injury, values correlate with global monitors such as jugular venous oximetry. Values depend on probe location and oxygen diffusion from capillaries. Brain hypoxia is not only dependent on
the values but also on the duration of low values. Values are listed as follows:
Normal
20–35 mm Hg
Compromised
20 mm Hg
Brain hypoxia
<15 mm Hg
O2 Reactivity Index
It is the ability of the brain to maintain a constant PbtO2 in spite of varying CPP. Impaired reactivity predicts worse
outcomes in TBI and in SAH (Table 8.10).
Uses
Cerebral hypoxia has been detected [using cerebral microdialysis (CMD), PbtO2] in spite of normal ICP and CPP.
PbtO2 monitoring is useful to titrate individualized therapy such as osmotherapy, hyperventilation, CPP, patient positioning, and deciding on timing of decompressive craniectomy. It is useful in identifying delayed cerebral ischemia in
SAH before ICP rise or clinical deterioration. PbtO2 monitoring is also useful to determine transfusion trigger, and blood
product administration can be rationalized. As secondary neuronal injury is caused by multiple mechanisms, multimodal monitoring is required to pick up the changes, and PbtO2 monitoring is a useful component to this multimodality.
Limitations
PbtO2 is a focal measure of oxygen tension. In conditions such as severe head injury, there will be greater regional
differences in blood flow and metabolism.
TABLE 8.10 Factors Affecting PbtO2 Values
Systemic Factors
Local Factors
Mean arterial pressure, intracranial pressure
Capillaries—number, length, diameter, flow pattern, perfusion rate
PaO2 and PaCO2
Oxygen consumption—neurons and glial cells
Hemoglobin, oxygen dissociation, viscosity
Oxygen diffusion gradients
pH, temperature
IV. NEUROMONITORING
158
8. NEUROMONITORING
Complications
t Infection
t Hemorrhage during probe placement
t Probe malfunction, displacement
CEREBRAL MICRODIALYSIS
This monitoring modality measures the brain’s metabolic substrate, both aerobic and anaerobic, in the interstitial
tissue space.
The probe consists of a coaxial double lumen tube (outer diameter 0.6 mm) covered by a dialysis membrane in the
distal 10 mm. The tube is continuously infused with an artificial CSF with known concentrations using a microinjector pump at a rate 0.3 µl/min. Depending on the concentration gradients and the pore size of membrane, metabolic
substrates diffuse through the dialysis membrane into the probe and are collected in to a sampling bottle. The sample
is then analyzed (hourly) for substrates using photometric enzyme-kinetic analyzer or by high-performance liquid
chromatography.
Currently three cutoff values (in KD) for membrane pore size are available. 20 and 100 KD are commercially
available for human use, and 10 KD is available for animal research purpose. 20 KD is used for assessing metabolic
substrates such as glucose, lactate, pyruvate, glycerol, and glutamate. 100 KD is used for assessing cytokines, interleukins, neurotrophic factors and neuronal biomarkers such as s-beta-100 protein. 10 KD is used for assessing neurotransmitters such as acetylcholine and norepinephrine.
Metabolic substrate measurements done by CMD are not absolute. It depends on various factors such as pore
size, rate of diffusion, and flow rate. Hence, the measurement is called as relative recovery. For a CMD probe
of 10-mm membrane length, 20 KD cutoff, and with a flow rate of 0.3 micro L/min, the relative recovery rate is
around is 70%.
In ischemia, there is accumulation of lactate and increase in lactate/pyruvate ratio (>25, significant). Also the
glucose level falls to <0.8 mmol/L (critical). Glutamate increase during ischemia indicates excitotoxicity. Glycerol is
a component of cell membrane and its accumulation indicates cell death.37
Probe Placement
CMD probe can be placed separately or as a component of triple bolt system. The probe is placed into the
white matter. Site depends on the lesion and the etiology. For diffuse injury, the probe is placed into the nondominant frontal subcortical white matter and the values are representative of global changes. The probe can
also be placed in the pericontusional area (in TBI) or in the vasospastic territory (in SAH), and the values are
representative of focal changes in the penumbra. The CMD measures the metabolic substrates in the surrounding 1-cm brain area.
Uses
t CMD helps in monitoring metabolic substrates to pick up neuronal ischemia at a reversible stage, e.g., in severe
head injury.
t In SAH, metabolic changes occur much before clinical manifestations of vasospasm and provide a window of
opportunity to treat this condition.
t CMD allows for glucose management in the ICU.
Limitations
t
t
t
t
Localized measurement
Expensive
Intermittent
Comparison between different studies using different threshold values is difficult to interpret as recovery rates
differ
IV. NEUROMONITORING
REFERENCES
159
CONCLUSION
Any one modality of monitoring is very unlikely to give complete picture of the cerebral functioning. At best it
can give complete information on one aspect of the cerebral functioning. Therefore, it is essential to have multimodal
monitoring and obtain as much information as possible and assimilate that information to come to some decision.
The neuromonitoring depends on the area of the brain at risk during the surgery and needs to decide how best to
monitor that part using various combination of modalities.
References
1. Kassab MY, Majid A, Farooq MU, Azhary H, Hershey LA, Bednarczyk EM, Graybeal DF, Johnson MD. Transcranial Doppler: an introduction
for primary care physicians. J Am Board Fam Med 2007;20:65–71.
2. Huber P, Handa J. Effect of contrast material, hypercapnia, hyperventilation, hypertonic glucose and papaverine on the diameter of the cerebral arteries. Angiographic determination in man. Invest Radiol 1967;2:17–32.
3. Halsey Jr JH. Risks and benefits of shunting in carotid endarterectomy. The International Transcranial Doppler Collaborators. Stroke
1992;23:1583–7.
4. Jansen C, Ramos LM, van Heesewijk JP, Moll FL, van Gijn J, Ackerstaff RG. Impact of microembolism and hemodynamic changes in the brain
during carotid endarterectomy. Stroke 1994;25:992–7.
5. Schmidt EA, Czosnyka M, Gooskens I, Piechnik SK, Matta BF, Whitfield PC, Pickard JD. Preliminary experience of the estimation of cerebral
perfusion pressure using transcranial Doppler ultrasonography. J Neurol Neurosurg Psychiatry 2001;70:198–204.
6. Salgado AS, Zângaro RA, Parreira RB, Kerppers II. The effects of transcranial LED therapy (TCLT) on cerebral blood flow in the elderly
women. Lasers Med Sci 2015;30(1):339–46.
7. Alpaidze M, Beridze M. Reversible cerebral vasoconstriction syndrome and migraine: sonography study. Georgian Med News March
2014;228:28–36.
8. Tiecks FP, Lam AM, Aaslid R, Newell DW. Comparison of static and dynamic cerebral autoregulation measurements. Stroke 1995;26:1014–9.
9. Puppo C, Fariña G, López FL, Caragna E, Biestro A. Cerebral CO2 reactivity in severe head injury. A transcranial Doppler study. Acta Neurochir
Suppl 2008;102:171–5.
10. Aaslid R, Lundar T, Lindegaard KF, Nornes H. Estimation of cerebral perfusion pressure from arterial blood pressure and transcranial recordings. In: Miller J, Teasdale G, Rowan J, Galbraith SL, Mendelow A, editors. Intracranial pressure VI. Berlin: Springer; 1986. p. 226–9.
11. Chan KH, Miller JD, Dearden NM, Andrews PJ, Midgley S. The effect of changes in cerebral perfusion pressure upon middle cerebral artery
blood flow velocity and jugular bulb venous oxygen saturation after severe brain injury. J Neurosurg 1992;77:55–61.
12. Nagai H, Moritake K, Takaya M. Correlation between transcranial Doppler ultrasonography and regional cerebral blood flow in experimental
intracranial hypertension. Stroke 1997;28:603–7.
13. Czosnyka M, Matta BF, Smielewski P, Kirkpatrick PJ, Pickard JD. Cerebral perfusion pressure in head-injured patients: a noninvasive assessment using transcranial Doppler ultrasonography. J Neurosurg 1998;88:802–8.
14. Soltani A, Singhal R, Obtera M, Roy RA, Clark WM, Hansmann DR. Potentiating intra-arterial sonothrombolysis for acute ischemic stroke by
the addition of the ultrasound contrast agents (Optison™ & SonoVue(®)). J Thromb Thrombolysis 2011;31:71–84.
15. Harrer JU, Eyding J, Ritter M, Schminke U, Schulte-Altedorneburg G, Köhrmann M, Nedelmann M, Schlachetzki F. The potential of neurosonography in neurological emergency and intensive care medicine: basic principles, vascular stroke diagnostics, and monitoring of strokespecific therapy – Part 1. Ultraschall Med 2012;33:218–32.
16. Harrer JU, Eyding J, Ritter M, Schminke U, Schulte-Altedorneburg G, Köhrmann M, Nedelmann M, Schlachetzki F. The potential of neurosonography in neurological emergency and intensive care medicine: monitoring of increased intracranial pressure, brain death diagnostics,
and cerebral autoregulation – Part 2. Ultraschall Med 2012;33:320–31.
17. Vajkoczy P, Roth H, Horn P, Lucke T, Thomé C, Hubner U, Martin GT, Zappletal C, Klar E, Schilling L, Schmiedek P. Continuous monitoring
of regional cerebral blood flow: experimental and clinical validation of a novel thermal diffusion microprobe. J Neurosurg 2000;93:265–74.
18. Nakase H, Kaido T, Okuno S, Hoshida T, Sakaki T. Novel intraoperative cerebral blood flow monitoring by laser-Doppler scanner. Neurol Med
Chir (Tokyo) 2002;42:1–4.
19. Kawamata T, Kawashima A, Yamaguchi K, Hori T, Okada Y. Usefulness of intraoperative laser Doppler flowmetry and thermography to predict a risk of postoperative hyperperfusion after superficial temporal artery-middle cerebral artery bypass for moyamoya disease. Neurosurg
Rev 2011;34:355–62.
20. Young WL, Prohovnik I, Schroeder T, Correll JW. OstapkovichN.Intraoperative 133Xe cerebral blood flow measurements by intravenous
versus intracarotid methods. Anesthesiology 1990;73:637–43.
21. Wintermark M, Sesay M, Barbier E, Borbély K, Dillon WP, Eastwood JD, Glenn TC, Grandin CB, Pedraza S, Soustiel JF, Nariai T, Zaharchuk
G, Caillé JM, Dousset V, Yonas H. Comparative overview of brain perfusion imaging techniques. Stroke 2005;36:e83–99.
22. Zweifel C, Hutchinson P, Czosnyka M. Intracranial pressure monitoring. In: Matta BF, Menon DK, Smith M, editors. Core topics in neuroanaesthesia and neurointensive care. Cambridge University Press; 2011. p. 45–62.
23. Leon-Carrion J, Martin-Rodriguez JF, Damas-Lopez J, Barroso y Martin JM, Dominguez-Morales MR. Delta-alpha ratio correlates with level
of recovery after neurorehabilitation in patients with acquired brain injury. Clin Neurophysiol 2009;120:1039–45.
24. Foreman B, Claassen J. Quantitative EEG for the detection of brain ischemia. Crit Care 2012;16:216.
25. Rampil IJ. A primer for EEG signal processing in anesthesia. Anesthesiology 1998;89:980–1002.
26. Sundt Jr TM, Sharbrough FW, Piepgras DG, Kearns TP, Messick Jr JM, O’Fallon WM. Correlation of cerebral blood flow and electroencephalographic changes during carotid endarterectomy: with results of surgery and hemodynamics of cerebral ischemia. Mayo Clin Proc 1981;56:533–43.
IV. NEUROMONITORING
160
8. NEUROMONITORING
27. Gollwitzer S, Groemer T, Rampp S, et al. Early prediction of delayed cerebral ischemia in subarachnoid hemorrhage based on quantitative
EEG: A prospective study in adults. Clin Neurophysiol 2015;126(8):1514–23.
28. Guerit JM. Neurophysiological testing in neurocritical care. Curr Opin Crit Care 2010;16:98–104.
29. Sivaraju A, Gilmore EJ, Wira CR, et al. Prognostication of post-cardiac arrest coma: early clinical and electroencephalographic predictors of
outcome. Intensive Care Med 2015;41:1264–72.
30. Zouridakis G, Papanicolaou AC, editors. A concise guide to intraoperative monitoring. New York: CRC Press; 2001.
31. Glass PS, Bloom M, Kearse L, Rosow C, Sebel P, Manberg P. Bispectral analysis measures sedation and memory effects of propofol, midazolam, isoflurane, and alfentanil in healthy volunteers. Anesthesiology 1997;86:836–47.
32. Thimmiah R, Thirthalli J, Ramesh VJ, et al. Effect of a course of Electroconvulsive therapy on interictal bispectral index values. JECT
2012;28:20–3.
33. Umamaheswara Rao GS, Ali Z, Ramamoorthy M, Patil J. Equi-MAC concentrations of halothane and isoflurane do not produce similar bispectral index values. J Neurosurg Anesthesiol 2007;19:93–6.
34. Sarrafzadeh AS, Kienin KL, Unterberg AW. Neuromonitoring: brain oxygenation and microdialysis. Curr Neurol Neurosci Rep 2003;3:517–23.
35. Deleted in review.
36. Ghosh A, Elwell C, Smith M. Cerebral near-infrared spectroscopy: A work in progress. Anesth Analg 2012;115:1373–83.
37. Rey JR, Guerrero JJE, Sanchez MAM, Cabezas FM. Cerebral microdialysis in the current clinical setting. Med Intensiva 2012;36:213–9.
IV. NEUROMONITORING
C H A P T E R
9
Multimodal Monitoring
A. Defresne, V. Bonhomme
CHR Citadelle, Liege, Belgium
O U T L I N E
Introduction
161
Intracranial Pressure Monitoring
Temperature
162
Electroencephalography and Depth of Anesthesia
Monitoring
Electroencephalography, Electrocorticography,
and Evoked Potentials for Monitoring Nervous
System Integrity
Hypnotic Component of Anesthesia Monitoring
Nociception Monitoring
Oxygen Transport, Hemodynamics, and Brain
Metabolism
Arterial Blood Pressure
Cardiac Output
Intravascular Volume Status
Hemoglobin Concentration
Jugular Venous Oxygen Saturation
Near Infrared Spectroscopy
Intraoperative Indications of Near Infrared
Spectroscopy
Limitations of Near Infrared Spectroscopy
Transcranial Doppler Ultrasonography
Ultrasound Basic Physics
Practical Applications
162
162
163
164
164
166
166
166
168
168
169
169
171
173
173
173
174
Miscellaneous
174
Integration of Information and Decision-Helping
Systems
175
Clinical Pearls
Inaccessible Forehead for Electroencephalographic
Monitoring
176
References
176
176
INTRODUCTION
The intraoperative anesthetic management of neurosurgical patients, particularly in case of intracranial
neurosurgery, necessitates close attention to maintaining perfect homeostasis, to avoid secondary lesions to
eventually suffering neural tissues.1 This involves rapid and smooth anesthetic depth transitions, adequate
hypnotic and antinociceptive depth, hemodynamic stability, minimal interference with cerebral circulation,
control of intracranial pressure (ICP) and cerebral perfusion pressure (CPP), temperature, hematosis, and,
ideally, neuroprotection.2 Specific situations also require avoidance of a too marked interference with recordings that aim at assessing nervous system integrity. In addition to classical anesthesia monitoring, including
electrocardiography, pulse oximetry, noninvasive blood pressure, and respiratory gas concentration, flow and
pressure monitoring, several dedicated monitors may be of utility in achieving these specific goals of neuroanesthesia and eventually help improving patient outcome. They will be reviewed in this chapter, with emphasis
on their basic principles, their routine use, the additive value of multimodality, and their demonstrated
clinical benefits.
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00009-9
161
© 2017 Elsevier Inc. All rights reserved.
162
9. MULTIMODAL MONITORING
TEMPERATURE
Temperature control during neuroanesthetic procedures is mandatory. Hyperthermia is known to worsen computed tomography–evidenced lesions and the clinical status of brain-injured patients,3 and hypothermia is associated with undesirable side effects including alterations of pharmacokinetics and pharmacodynamics of anesthetic
agents, shivering, discomfort upon recovery, sympathetic nervous system activation, increase of myocardial
workload, arrhythmias, coagulation disorders, facilitated wound infection, delayed healing, and immune deficit.4 Active warming and tight temperature control lower the occurrence of hypothermia-related adverse events.5
Hence, maintenance of normothermia is the rule for most neurosurgical procedures and is a requirement for a
good quality early recovery after intracranial neurosurgery and adequate subsequent neurological follow-up.6
Apart from its deleterious effects, hypothermia has been advocated as a potential intraoperative neuroprotective
measure in specific neurosurgical procedures but has never proved to be beneficial in terms of outcome,7,8 except
for deep hypothermia during the rarely occurring surgical treatment of giant aneurism involving prolonged circulatory arrest.9
Temperature can be reliably measured using either thermistors or thermocouples. Thermistors are composed of a
semiconductor, whose impedance increases with temperature. Thermocouples contain two metals, and the electric
potential difference between them changes with temperature. Other devices are based on infrared emission and
measure temperature in the external auditory canal, or they contain a liquid crystal sensor that can be applied on the
skin,4 but they are less frequently used in the operating room.
Temperatures of interest during neurosurgical procedures are mainly core and brain temperatures, which are
not necessarily identical. Routinely monitored sites generally allow obtaining only estimates of those temperatures.
Tympanic temperature is probably the closest to brain temperature, but the risk of tympanic lesion by the probe
is not negligible. Gold standard assessment of core temperature is the central blood temperature measured at the
level of a pulmonary artery catheter. However, those catheters are rarely necessary for hemodynamic control during
anesthesia for neurosurgery. Reliable alternatives for measuring core temperature are esophageal, bladder, and rectal
temperatures. Temperature monitoring and active control for all intracranial procedures and procedures that last for
more than 1 h sound reasonable.
OXYGEN TRANSPORT, HEMODYNAMICS, AND BRAIN METABOLISM
To fulfill metabolism requirements, adequate oxygen delivery to all parts of the central nervous system must be
constantly insured. This requires effective hematosis in the lungs, high enough blood hemoglobin concentration, and
adequate cardiac output. Aside from global systemic transport, oxygen delivery to the brain also depends on global
and regional cerebral hemodynamics, which is under the dependency of physiological regulating systems, and may
be impeded by pathological processes, iatrogenic interventions, or by medications. Raised ICP and decreased CPP,
vasospasms, or hypocarbia are examples of such. Several monitoring devices, either invasive or noninvasive, may be
used to control for the adequacy of all intervening elements.
Arterial Blood Pressure
Intermittent noninvasive blood pressure monitoring is standard during all anesthetic procedures and is considered to be enough control for most moderately invasive spine surgeries. A beat-to-beat control of blood pressure is
necessary when managing fragile patients or during potentially bleeding surgeries, as well as during all intracranial
procedures. Indeed, such a tight control and invasive monitoring allows maintaining adequate cerebral perfusion10
and limiting the risk of bleeding, either during the course of the procedure or upon recovery.6 Invasive arterial blood
pressure monitoring may also be of help when assessing the intravascular volume status and for drawing arterial
blood samples and performing gas analysis.
The entire set for invasive blood pressure monitoring includes an arterial cannula, a continuous irrigation system
through infusion of saline under pressure, a pressure transducer that transforms the mechanical signal into an electrical one, and a monitor with a display screen. Zeroing is required at starting of use, with the transducer placed at
the horizontal level of the heart. Site for arterial cannula insertion may vary, the most frequent being the radial artery
at the wrist. Ulnar, pedal, and femoral arteries can also be used. Humeral artery should be avoided because its distal
territory cannot be supplied by collateral vascularization. Strict sterile conditions must be respected during insertion,
and echo guiding may be of help for difficult cases11 (Fig. 9.1).
IV. NEUROMONITORING
OXYGEN TRANSPORT, HEMODYNAMICS, AND BRAIN METABOLISM
163
FIGURE 9.1 Example of an echo-guided arterial cannula insertion. Localization of the radial artery is made easy using echography and color
Doppler, allowing visualization of flux pulsatility and direction (1). After puncture of the artery, the insertion of the metallic guide can be followed
online (2) and the adequate positioning of the catheter into the artery can be checked thereafter (3).
Continuous beat-to-beat measurement of blood pressure is also possible noninvasively, using volume clamp or
arterial tonometry methods.12 The volume clamp derives blood pressure according to pressure variations in a finger
cuff, so as to keep finger artery volume constant. Arterial tonometry uses fingertip plethysmography and pulse volume amplitude. Some authors have evaluated the use of such systems during neurosurgical procedures, in patients
where arterial blood sampling is not expected to be necessary. The idea was to avoid the well-known complications
of an arterial cannula, including vessel or nerve trauma, pseudo-aneurysm formation, bleeding due to unnoticed
disconnection, local hematoma, infection, arterial thrombosis, distal embolism, and ischemia.13 Although some studies have reported acceptable bias as compared to invasive intra-arterial measurement,14 a recent meta-analysis has
warned about out of acceptable range inaccuracy and imprecision.12
Blood pressure management during a neurosurgical procedure must take account of patient comorbidities,
risk of bleeding, and brain perfusion. Large hemodynamic variations frequently occur during induction of and
recovery from general anesthesia, and practitioners should seek after stability as much as possible. However,
perfusion pressure is not the only determinant of cerebral blood flow. In addition to CPP, autonomic regulation, metabolism coupling, and cerebrovascular reactivity to CO2 and O2, acute or chronic changes in global
cardiac output play a role. Cerebral blood flow autoregulation should be regarded as a dynamic integrated
process, displacing the lower limit, upper limit, and plateau height of the autoregulatory perfusion pressure
curve as a function of circumstances. Hence, combining blood pressure monitoring with estimates of cardiac
output and cerebral circulation seems to be sound and might reveal efficient at ameliorating patient outcome
in the future.10
The respiratory variation of the blood pressure curve also offers simple means for assessing the intravascular volume status. This point is developed later in the chapter.
Cardiac Output
Assessment of cardiac output during intracranial neurosurgery has recently been advocated as a potential interesting monitoring to help optimizing cerebral hemodynamics.10 However, systematic cardiac output monitoring in
a neurosurgical context is not the rule, except for very specific and rare procedures such as giant aneurysm clipping
under circulatory arrest and in patients with severe cardiovascular comorbidities.
IV. NEUROMONITORING
164
9. MULTIMODAL MONITORING
Gold standard methods of cardiac output measurement are the classical thermodilution techniques through a
pulmonary artery catheter and transthoracic or transesophageal echocardiography. For neurosurgical procedures,
and in otherwise healthy patients, transesophageal echocardiography is mainly used during surgery in the sitting
position15 in case of patent oval foramen to detect eventual paradoxical air embolism.16 Transesophageal Doppler of
the aorta can also estimate the stroke volume from the aortic velocity waveform and is sometimes considered as a reference measure of cardiac output.17 Its accuracy highly depends on probe placement and on any modification of the
aortic diameter such as in case of intra-aortic balloon or coarctation. Several noninvasive devices are also proposed
with variable performance, accuracy, and precision.18 Electrical thoracic bioimpedance relies on the assumption that
thoracic impedance changes parallel to those of stroke volume. The limitations of the technique include electrode
positioning, electrical interference, abnormal fluid inside the thorax, changes in peripheral vascular resistance, age,
gender, arrhythmias, and movement. Thoracic bioreactance is a variant, which takes account of thoracic voltage
phase shift and has almost the same limitations, except that it can still provide an estimate of cardiac output in case
of cardiac arrhythmia because it averages values over several seconds. Cardiac output may also be derived from
the arterial pressure contour, obtained either directly or from the noninvasive methods described above, and using
variable algorithms.17 All these techniques are highly dependent on arterial pressure waveform signal quality, and
therefore on eventual finger edema, intense vasoconstriction, sensor position, and movement. Some of them necessitate calibration and others not.19
The clinical utility and reliability of cardiac output measurement in neuroanesthesia have been relatively poorly
studied. Thoracic bioimpedance has been demonstrated to reliably mirror the hemodynamic changes associated with
crucial events during neurosurgical procedures such as induction, scalp infiltration, changes in anesthetic depth,20
or mannitol administration.21 Stroke volume monitoring is also sometimes used to guide fluid administration during intracranial neurosurgery.22 Up to now, scientific evidence does not support equivalence between invasive and
noninvasive methods of hemodynamic monitoring and the lasts cannot replace the formers when close monitoring
is needed, such as during surgery in the sitting position.15
Intravascular Volume Status
Substantial fluid shifts can occur during neurosurgical procedures, either as a consequence of blood loss or following the administration of medications aiming at improving brain relaxation, such as mannitol or furosemide. Hence,
controlling for the intravascular volume status is essential. Several methods are available of which some are simple
and easy to implement. Indeed, central venous pressure and pulmonary capillary wedge pressure are no longer considered as gold standards,23 at least in patients under positive pressure ventilation.
The invasive arterial pressure waveform allows measuring the delta pulse pressure (DPP) or the delta-down
(DD) intermittently (Fig. 9.2). To do so, the patient must be in cardiac sinus rhythm and under mechanical ventilation. The tidal volume should be set at least at 8 mL/kg, and the frequency rate reduced to values as low as 4
per min to mimic respiratory pauses. DPP corresponds to the difference between the maximal and minimal pulse
pressure during one breathing cycle, divided by the mean of those two pulse pressures,24 while DD is the difference between the systolic arterial pressure at the end of a 5-s respiratory pause, immediately before lung inflation,
and its minimal value during the course of one respiratory cycle.25 Values of DPP higher than 13%, and values
of DD higher than 5 mmHg are indicative of an increased probability of preload cardiac output dependency and
a positive hemodynamic response to fluid loading. Both indices have been demonstrated to be accurate in the
context of intracranial neurosurgery.26 Their main disadvantages are their intermittent nature and susceptibility
to confounding factors, including nonsinus cardiac rhythm, ventilation volume and pressure, vasopressor administration, abdominal pressure, and cardiac failure.27 Dynamic parameters may have additive value, insofar as they
offer a continuous assessment of the intravascular volume. They are based on systolic pressure or stroke volume
variability as assessed by pulse contour analysis of an arterial or plethysmographic waveform. Using such technology, several commercially available devices provide indexes of susceptibility to fluid loading.23,28 Closed-loop
systems of fluid administration have even been proposed.29 Accumulating evidence suggests that goal-directed
fluid administration is beneficial for enhanced recovery after specific types of surgeries,30,31 but this question has
still not been fully addressed for neurosurgical procedures.
Hemoglobin Concentration
The main oxygen transporter in the blood is hemoglobin. During potentially bleeding neurosurgical procedures and
in patients with preexisting anemia, it is necessary to have prompt access to rapid, accurate, and reliable hemoglobin
IV. NEUROMONITORING
OXYGEN TRANSPORT, HEMODYNAMICS, AND BRAIN METABOLISM
165
FIGURE 9.2 Illustration of delta-down (DD) and delta pulse pressure (DPP) calculation. For this measurement, respiratory rate should be set
at 4 per min or lower to mimic a respiratory pause. The patient should be in cardiac sinus rhythm, and the respiratory tidal volume should be
8 mL/kg or higher. DPP corresponds to the difference between maximum and minimum pulse pressure over a respiratory cycle, divided by their
mean. DD is the difference between maximum and minimum systolic blood pressure over a respiratory cycle.
TABLE 9.1 Characteristics of Point-of-Care Hemoglobin Concentration Measuring Devices
Commercial Devices
Technology
Blood Sample Needed
Comparison With Automated
Laboratory Measurements MD (LA)
Hemocue
Reagent-based photometry
Yes
0.08 (−1.3–1.4)32
Diaspect
Nonreagent-based photometry
Yes
−0.18 (−2.05–1.68)165
i-Stat
Conductivity
Yes
0.05 (−1.16–1.25)166
CO-Oxi
Multiple wavelength co-oximetry
No
−0.03 (−3.0–2.9)32
OrSense
Occlusion spectroscopy
No
−0.66 (−3.39–2.09)165
Haemospect
Transcutaneous reflection spectroscopy
No
−0.22 (−2.64–2.21)167
An example for each type of technology is presented. Mean difference (MD), 95% limits of agreement (LA) in g/dL compared to automated laboratory measurements are indicated, as well as the source from which those data were driven.
CO-Oxi, Pulse CO-Oximetry (Masimo Corp., Irvine, CA, USA); Diaspect, Diaspect Hemoglobinometry (Diaspect Medical GmbH, Germany); Haemospect,
Haemospect™ (MBR Optical Systems, Herdecke, Germany); Hemocue, Hemocue (HemoCue, Angelholm, Sweden); i-Stat, i-Stat (Abbott Laboratories, Abbott Park,
IL, USA); OrSense, OrSense™ (Ness Ziona, Israel).
concentration measuring devices. Aside from automated laboratory hemoglobin measurement systems, which may be
too time demanding particularly in the context of an emergency, several point-of-care devices are proposed.32
Blood sample–requiring systems at the bedside use reagent-based33 or nonreagent-based34 absorption photometry at multiple wavelengths or conductivity.35 Other noninvasive, and hence non–blood sample–requiring devices
involve multiple wavelength co-oximetry,36 occlusion spectroscopy,37 or transcutaneous reflection spectroscopy38
technologies. Several studies have been performed to compare the results obtained with those devices and those
obtained using automated laboratory systems or between them (Table 9.1), not necessarily in the context of neurosurgery. Although bias as compared to laboratory values is generally small, the limits of agreement may be large
with a significant amount of outliers. In addition, discrepancies may vary according to real hemoglobin level39 and
several confounding factors. Pulse co-oximetry may be confounded by ambient light, bilirubin, hemoglobin variants, intravascular dyes, motion, sensor positioning, temperature, venous congestion, edema, high tissue lipid content, and hypovolemia.40 Although having been demonstrated to be indicative of hemoglobin level during complex
spine surgery,41 pulse co-oximetry remains less accurate than conventional laboratory measurements.42 Accuracy
may improve when calibrating with a baseline laboratory testing at the beginning of the procedure, particularly in
children undergoing neurosurgery.39 Those noninvasive monitors provide more timely information, but should be
considered as trend monitors rather than absolute indicators of transfusion need.
IV. NEUROMONITORING
166
9. MULTIMODAL MONITORING
Jugular Venous Oxygen Saturation
Near infrared spectroscopy (NIRS) combined with transcranial Doppler ultrasonography (TCDU) and jugular
venous oxygen saturation (SjvO2) monitoring are means for appreciating brain hemodynamics and oxygen delivery.
SjvO2 monitoring requires the retrograde placement of a catheter into a jugular vein, up to the jugular bulb, to monitor the mixed cerebral venous blood and above the C1/C2 level to limit contamination by the facial venous blood.43
Side for placement should preferably be the dominant side of the brain or the side with the most prominent pathology, if any. Oxygen saturation may be measured in regularly drawn blood samples or using a fiber-optic catheter and
light absorption in the red/infrared wavelength range. Although generally quite easily placed, the catheter may be
the source of rare complications, including hematoma, infection, thrombosis, raised ICP, or incidental wrong vessel
puncture.44
SjvO2 reflects the balance between oxygen delivery to the brain and cerebral metabolic consumption of oxygen.
Normal values range between 50% and 75%. Decreases in SjvO2 can be seen in case of systemic or local oxygen supply
deficiency, including hypoxia, low blood pressure, decreased CPP, embolism, or vasospasm (Table 9.2), and in case of
increased oxygen consumption including hyperthermia and seizure. Causes of SjvO2 values above normal range can
be classified into those related to a decrease in cerebral metabolism, restricted oxygen diffusion or extraction, shunting, increased oxygen supply, or a combination of these. For example, a global decrease in cerebral metabolism can
be observed during hypothermia. Infarction or inflammation causes restriction in oxygen diffusion and extraction,
as well as microvascular shunting. Increased oxygen supply is observed during hyperoxia, and hypercarbia can be
responsible for substantial shunting.
Interpretation of SjvO2 values may be uneasy in some instances. It is a global measure and it may miss eventually
occurring focal brain ischemia.45 When brain tissue death is massive, SjvO2 can be high as a consequence of shunting.
Classical causes of wrong SjvO2 values are displacement of the catheter, thrombus around its tip, and contamination
by facial venous blood. Anesthesia may also have an influence on SjvO2 values. Transient desaturations have been
reported under propofol anesthesia, more frequently in normothermic than in hypothermic patients,46 while SjvO2
increases have been observed under sevoflurane anesthesia.47 This is due to the different properties of those agents
on cerebral vasculature, propofol being vasoconstrictive, and sevoflurane vasodilating. Side of recording may also
be important, insofar as cerebral venous drainage usually predominate on one side through the dominant jugular
bulb.48
SjvO2 has mostly been used to guide therapy in the intensive care unit for traumatic brain injury and subarachnoid
hemorrhage patients. However, its intraoperative use during intracranial neurosurgery allows commonly detecting desaturation episodes,48 and its combination with TCDU49 may impact on clinical decision-making to optimize
cerebral physiology. Large randomized clinical trials investigating the impact of intraoperative SjvO2 monitoring on
patient outcome are lacking.
Near Infrared Spectroscopy
NIRS consists in measuring near infrared light absorption by the superficial part of brain tissues. Light is emitted
through the skin and the skull, entering the brain tissue to a depth of approximately 1–2 cm. Absorption at different wavelengths allows estimating the mixed capillary–venous regional cerebral oxygen saturation (rScO2).50 rScO2
depends on the balance between local metabolism and oxygen delivery, which in turn depends on regional cerebral
blood flow and blood oxygen content. If metabolism is constant, rScO2 can be regarded as a surrogate measure of
regional cerebral blood flow changes. The system necessitates the use of a noninvasive scalp probe and allows continuous monitoring of rScO2 over a restricted area of approximately 1 cm2.
Normal rScO2 values at atmospheric oxygen levels range from 62% ± 6% in cardiac surgery patients51 to 71% ± 6%
in healthy young men.52 Low preoperative baseline rScO2 values are predictive of increased 30-day morbidity and
mortality.50,51 There is no consensus in the literature on the rScO2 threshold value indicating significant desaturation,
but a decline of more than 20% as compared to baseline is generally accepted as a predictor of cerebral ischemia. It
is also generally agreed that both depth and duration of desaturation are important.50,53 A less extreme threshold of
10% should probably prompt therapeutic intervention to raise rScO2.54
Intraoperative Indications of Near Infrared Spectroscopy
At the extremes of age, in the very young and in the elderly over 60 years, the brain is at risk of injury during surgery
and anesthesia.55 In addition to the sensitivity of their brain to the effects of anesthetic agents, aged patients are prone
to experience postoperative cognitive decline and develop delirium or stroke. Evidence accumulates demonstrating
IV. NEUROMONITORING
t S
pO2
t B
GA
t I CP monitor
t T
CDU
Hypoxia
↓ CPP
t C
O monitoring
Low CO
Embolism
Thrombosis
Cause
t I mprove
hemodynamics
t T
ransfuse
t E
EG monitoring t L
ighten
t D
HOA monitoring
anesthesia if
appropriate
Deep sedation
IV. NEUROMONITORING
Hypercarbia
Cause
Correction
t R
espiratory CO2 t O
ptimize ventilation
monitoring
t B
lood gas
analysis
Diagnosis
t I dentify cause
t T
reat cause
t E
ventual reperfusion
strategies if not too late
Shunting
t I maging
t E
EG
monitoring
Inflammation t I maging
Infarction
Correction
Hyperoxia
Cause
Inflammation
Infarction
Cause
t C
alcium antagonist
Seizure
t A
ngiography and
endovascular reperfusion
strategies
t I ncrease CPP
Restricted Oxygen Diffusion
Diagnosis
Cause
t A
ntiepileptic
medications
t C
ooling
Correction
Correction
t B
GA
Diagnosis
t O
ptimize
ventilation and
FiO2
Correction
Increase in Oxygen Supply
Same as restricted oxygen diffusion
Diagnosis
Restricted Oxygen Extraction
t E
EG monitoring
t T
emperature
monitoring
Diagnosis
Increased Cerebral Oxygen Consumption
t I dentify cause
Hyperthermia
t S
ystemic or endovascular
reperfusion strategies
Correction
High SjvO2 ( > 75%)
t E
EG
t T
CDU
t E
EG
Diagnosis
Local Oxygen Supply Deficiency
ABP, arterial blood pressure; BGA, blood gas analysis; CO, cardiac output; CPP, cerebral perfusion pressure; EEG, electroencephalography; FiO2, inspired oxygen fraction; ICP, intracranial pressure;
SjvO2, jugular venous oxygen saturation; SpO2, peripheral saturation in oxygen; TCDU, transcranial Doppler ultrasonography.
Same as restricted oxygen diffusion
Infarction
Inflammation
Correction
Diagnosis
Cause
Microvascular Shunting
t W
arming
t T
emperature
monitoring
Hypothermia
Correction
Diagnosis
Cause
Cause
t L
ower ICP
Vasospasm
t I mprove
hemodynamics
t O
ptimize
ventilation
t ↑ FiO2
Correction
Decrease in Cerebral Metabolism
t H
emoglobin
measurement
Anemia
t ↑ ICP
t ↓ ABP
Diagnosis
Cause
Systemic Oxygen Supply Deficiency
Low SjvO2 (<50%)
TABLE 9.2 Causes of Abnormal Jugular Venous Oxygen Saturation Values and Proposed Diagnosis Help, and Corrective Measures
OXYGEN TRANSPORT, HEMODYNAMICS, AND BRAIN METABOLISM
167
168
9. MULTIMODAL MONITORING
that cerebral oxygen deprivation episodes lead to adverse clinical outcomes including cognitive decline, major organ
dysfunction, and prolonged hospital length of stay, both in cardiac and noncardiac surgery patients.54,56
The intraoperative use of NIRS was initially described for aortic arch surgeries and carotid endarterectomies.
Despite an increasingly large use of rScO2 monitoring during cardiac surgery, Zheng et al. were not able, in their
systematic review, to strongly link cerebral desaturations, and interventions to improve it, with the incidence of postoperative neurological complications.57 Indeed, most of NIRS observational studies in aortic arch surgery patients
have small sample sizes, and a very low incidence of strokes, rendering the assessment of the relationship between
cerebral desaturation and neurological outcome uneasy. Some case reports, as well as observational studies, have
shown the usefulness of NIRS to warn about cannula malposition and failure of cardiopulmonary bypass.58–60 During
carotid endarterectomy, the most efficient approach to cure reduced rScO2 is the insertion of a shunt. NIRS has a sensitivity of approximately 75% and a specificity of 98% at correctly indicating the need for shunting.61,62 After clamp
release, an increase in rScO2 of more than 20% predicts the occurrence of a cerebral hyper-perfusion syndrome.63,64
Regarding nonvascular surgical procedures, a review performed by Nielsen identified surgeries where a decrease
in rScO2 is reported. These surgeries include procedures in the Fowler position, which are characteristic of shoulder
and laparoscopic surgeries, for example. Hip surgery and single lung ventilation thoracic surgery are also at risk of
decrease in rScO2.
The link between repeated occurrences of cerebral desaturation and worse postoperative outcome is, however,
not straightforward. A pronounced intraoperative cerebral desaturation does not necessarily lead to postoperative
cognitive decline after shoulder surgery in the beach chair position. In addition, an association between cerebral
desaturation and outcome parameters such as postoperative wound infection, kidney failure, and myocardial infarction remains to be established.54 Current evidence points toward worse outcome, including postoperative cognitive
decline, kidney dysfunction, and prolonged hospital length of stay in cardiac surgery patients having experienced
severe cerebral desaturation, as well as in patients having been submitted to certain types of other noncardiac surgeries. Even if more studies are needed to demonstrate a clear association, the overall risk to benefit ratio goes in favor
of the intraoperative utility of NIRS, all the more since it is noninvasive, continuous, and has a moderate cost.
Limitations of Near Infrared Spectroscopy
Several confounding factors may impede the adequate interpretation of rScO2 values. A contamination by blood
originating from the external carotid artery territory may occur, and the extent to which this contamination may
skew NIRS accuracy varies from a device to another. External carotid contamination may be as high as 20%.50,56 An
influence of anatomic variability in skull shape, for example enlarged frontal sinus, or uncommon cerebral venous
drainage through superior sagittal veins must be considered.54 When vasopressors such as noradrenaline are administered to maintain arterial pressure, up to one-third of changes in rScO2 can be accounted for by changes in skin
blood flow. Ephedrine, by increasing mean arterial pressure without affecting cardiac output, does not change rScO2,
while pure α1-adrenergic agonists such as phenylephrine do. In that case, the decrease in rScO2 is secondary to a
drop in cardiac output. It may therefore be advised to check for an adequate intravascular volume status before
interpreting changes in rScO2 following the administration of a vasopressor.54,56 In patients with a liver disease, high
bilirubin plasma concentrations alter NIRS measurements through the competitive absorption of light by this pigment.54,56 Another limiting factor is the limited frontal brain area where NIRS can measure oxygenation. Embolism
or ischemia occurring in distant areas can therefore be undetected53. Finally, commercial cerebral oximetry devices
assume a fixed value of either 70:30 or 75:25 for the venous to arterial blood volume ratio. Relative changes in the
volume of one compartment or the other will change the overall cerebral hemoglobin saturation, without any real
change in the saturation of each individual compartment.53
Transcranial Doppler Ultrasonography
In 1982, Aaslid was the first to describe the use of transcranial Doppler for measuring cerebral blood flow velocity.65 Ultrasound brain exploration has long been limited to Doppler exploration, but technological advances now
allow grossly imaging the main brain structures using echography. Through the visual recognition of the insonated
vessel, it has considerably facilitated Doppler use and has added morphological elements that can be useful to diagnosis. TCDU is the only noninvasive and cost-effective bedside tool that provides real-time information on cerebral
hemodynamics. It has no contraindications, and its technical limitations can often be overcome by the intravenous
administration of a sonography contrast agent.66 In adults, insonation usually occurs through the four commonly
defined acoustic windows, including the temporal, orbital, suboccipital, and submandibular window. These windows are characterized by a thinner bone layer, permitting ultrasound penetration.
IV. NEUROMONITORING
OXYGEN TRANSPORT, HEMODYNAMICS, AND BRAIN METABOLISM
169
Ultrasound Basic Physics
Echography necessitates an ultrasound transducer, which acts both as a transmitter and a receiver. Reflection
of ultrasound waves occurs at the interface between different tissue types, and depends on the resistance to ultrasound breakthrough, or acoustic impedance. When ultrasounds are reflected, and thus sent back to the transducer,
the piezoelectric crystals convert the mechanical energy of the returning echoes into an electrical current, which is
processed by the ultrasound machine to produce a two-dimensional gray scale image. During their travel into the
tissues, ultrasound waves are attenuated.67 Attenuation depends on three factors, namely the attenuation coefficient
of the tissue, the traveled distance, and the ultrasound frequency. Insofar as bones are substantially impervious to
ultrasounds, and that attenuation increases with ultrasound frequency, a low-frequency transducer in the order of
2 MHz is necessary for transcranial ultrasonography.
The flow within vascular structures can be studied using color flow Doppler. Red blood cells moving toward the
transducer return waves at a higher frequency than the emitted signal. The corresponding image of flow is displayed
in red on the device. Contrarily, when those cells are moving away from the transducer, the returned waves have a
lower frequency and are displayed in blue.68 Color flow Doppler helps identifying arteries through the integration
of information coming from the anatomical view, as well as direction and pulsatility of flow. Pulse wave Doppler
is based on the Doppler principle described by Christian Doppler in 1843. Exactly like in color flow Doppler, the
reflected ultrasound wave has a higher or a lower frequency than the emitted one. This is termed as a positive
or negative shift in frequency, respectively. The velocity of red blood cells as measured by a pulse wave Doppler
depends on the cosine of the angle of insonation (Fig. 9.3). As a consequence, when the angle of insonation is 0°, the
measured velocity equals the true velocity. When the angle is 90°, the measured velocity is 0, irrespective of the actual
velocity.67–69
Practical Applications
In the operating room and during neurosurgery, TCDU can be used to seek for hyperemia, vasospasm, microembolic signals, vasomotor reactivity (VMR), and to estimate ICP. These indications are also common in the neurointensive care unit. Other common indications of TCDU are the identification of sickle cell disease children at the highest
risk of first-ever stroke,70 and those in need of blood transfusion.71 TCDU is also very sensitive and specific to detect
right-to-left shunt and to quantify the functional repercussions of such a shunt. The last indication of TCDU is the
diagnosis of a cerebral circulatory arrest.69,72 This paragraph will be limited to the indications that are of potential
utility in the operating room.
Vasospasm and Hyperemia
Vasospasm is a suddenly occurring and rapidly evolving pathology. TCDU is therefore a key tool for its identification and diagnosis, as well as for daily monitoring, appreciation of severity, and circumscription of incriminated
territory. A change in mean flow velocity (MFV) often precedes an eventual neurological deficit. Detecting those
changes in velocity authorizes to adapt the treatment precociously, and prevent complications.73
The threshold for deciding on an increased MFV varies according to the studied artery (Table 9.3).74 More than an
absolute number, the magnitude of a change in mean velocity is also important. An increase of more than 50 cm/s
in 24 h is predictive of the occurrence of a vasospasm-related ischemic lesion. In the middle cerebral artery, when
the MFV remains between 120 and 200 cm/s, the sensitivity for diagnosing a vasospasm is weak (67%).75 In that
case, only arteriography may provide certainty. Another useful parameter for diagnosing vasospasm in the middle
FIGURE 9.3 Illustration of the importance of the insonation angle in pulse wave Doppler.
IV. NEUROMONITORING
170
9. MULTIMODAL MONITORING
TABLE 9.3 Proposed Criteria for Diagnosing Vasospasm on Most Frequently Studied Cerebral Arteries
Concerned Artery
MCA
MFV
Lindegaard’s ratio for the MCA
Moderate vasospasm
>120 and <200 cm/s
Severe vasospasm
>200 cm/s
Hyperemia: <3
Moderate vasospasm: 3–6
Severe vasospasm: >6
ACA
MFV
Discreet vasospasm
>80 cm/s
Significant vasospasm
>130 cm/s
PCA
MFV
Discreet vasospasm
>90 cm/s
Significant vasospasm
>110 cm/s
Vertebral artery
MFV
Vasospasm
>80 cm/s
Basilar artery
MFV
Vasospasm
>95 cm/s
ACA, anterior cerebral artery; Lindegaard’s ratio, ratio between the mean flow velocity in the MCA and the mean flow velocity in the submandibular internal carotid artery; MCA, middle cerebral artery; MFV, mean flow velocity; PCA, posterior cerebral artery.
cerebral artery is the Lindegaard’s ratio. It corresponds to the ratio between the MFV in the middle cerebral artery
and the MFV in the submandibular internal carotid artery. The Lindegaard’s ratio may also help differentiating
hyperemia from vasospasm, which have completely opposite treatments.72,76
Microembolic Signals or High Intensity Transient Signals
During carotid surgery, monitoring an intracranial artery distal to the steno-occlusive site allows detecting
spontaneous embolic signals and quantifying the embolic potential of the atherosclerotic plaque. According to
the International Cerebral Hemodynamics Society, those signals are characterized by a high intensity, that is
more than 10 dB higher than blood flow, a random occurrence during the cardiac cycle, a unidirectional nature,
a short duration (10–100 ms), and an audible component (explosive signal) (Fig. 9.4). This kind of surveillance
is useful not only during carotid surgery, but also during aortic, cardiac, or even orthopedic surgery. It helps
identifying the critical phase of surgery, when the risk of embolism is important. Detecting such a high risk
should prompt corrective measures at the level of the equipment (e.g., extracorporeal circulation) or the surgical technique.72
Vasomotor Reactivity
VMR represents the ability of cerebral vessels to maintain a near constant blood flow in response to a vasomotor
stimulus. TCDU allows a real-time observation of vasomotor changes in response to various stimuli. Concerned
stimuli can be hemodynamic stimuli such as a rapid leg cuff deflation or a Valsalva maneuver or the transient hyperemia response to medications such as acetazolamide, breath-holding, and CO2 inhalation.77 CO2 is the strongest
vasodilation stimulus to the cerebral circulation. Several factors may influence VMR, including gender and level
of estrogenic impregnation, as well as chronic cardiovascular pathologies such as chronic high blood pressure and
diabetes. In those two last cases, VMR is reduced.
A simple method for measuring VMR has been described by Markus et al. It consists in calculating the breathholding index (BHI) as follows:
-&6END å -&6BASELINE
-&6BASELINE
°
SECONDS OF BREATH HOLDINGS
where the middle cerebral artery MFV is measured during an at least 30 s breath-holding. MFVbaseline corresponds to
the MFV before breath-holding and MVFend to the MFV 4 s after restarting breathing. When BHI is lower than 0.69,
VMR is impaired. In patients with asymptomatic carotid stenosis, BHI can be used to identify those at a higher risk
of stroke.78
IV. NEUROMONITORING
INTRACRANIAL PRESSURE MONITORING
171
FIGURE 9.4 Illustration of microembolic signals or high intensity transient signals as evidenced by transcranial Doppler ultrasonography.
Estimation of Intracranial Pressure
To avoid invasiveness of direct ICP monitoring, TCDU can serve as a noninvasive alternative. This bedside tool
may be used to study the hemodynamic changes induced by an increase in ICP. There exists a qualitative relationship
between the progressive increase in ICP and the occurrence of abnormal TCDU waveform, while assuming a constant arterial CO2 content and a constant degree of distal vasoconstriction. The most sensitive parameters to detect an
increase in ICP, and hence a decrease in CPP, are the diastolic velocity (Vd) and the pulsatility index (PI) measured at
the level of the middle cerebral artery. Vd reflects the distal resistance to flow and decreases proportionally to CPP.
Contrarily, PI is inversely proportional to a decrease in CPP. A PI higher than 1.4 and a Vd lower than 20 cm/s are
indicative of severity.77,79
PI is not dependent on the angle of insonation cosine (Cos α), as indicated in its calculation formula:
#OSȗ (6S å 6D)
6S å 6D
, where Vs = systolic velocity, Vd = diastolic velocity, and Vm = mean velocity. For that
#OSȗ6M
6M
reason, PI is the most important variable to take account of when analyzing the Doppler signal. A low flow velocity
with a normal PI means an open insonation angle. These values should not be considered as pathological and should
motivate new measurements with a different insonation angle. Of note, PI is dependent on arterial CO2 partial
pressure.74
TCDU and direct measures of ICP are not interchangeable. In this indication, TCDU should rather be considered a
guide to identify critical patients, to orient therapeutic options while waiting for the insertion of an invasive monitoring device, and to unravel intraoperative doubts when not having a real ICP number.80 The information derived from
TCDU should always be integrated with several other available data, including mean arterial pressure, arterial CO2
partial pressure, peripheral saturation in oxygen, hemoglobin concentration, and the clinical status of the patient.
Other TCDU limitations are the operator skills, depth and angle of insonation accuracy, impermeable insonation
window, and eventual arterial stenosis or microangiopathy.81 Some of these limitations may be overcome by targeting the ophthalmic artery through the eye, using extracranial and intracranial segments of it as pressure sensors. In
that case, no patient-specific calibration is necessary.82
0) INTRACRANIAL PRESSURE MONITORING
Contrarily to neurointensive care, direct ICP and CPP monitoring is not often available intraoperatively during
intracranial neurosurgical procedures. The reasons are that patients often come to surgery without any implanted
monitoring device and intracranial neurosurgery involves skull and dura opening, hence compromising any pressure measurement. As a consequence, ICP monitoring will only be useful during the beginning of the procedure,
before skull opening, in patients already holding a probe. Otherwise, estimations of ICP can be obtained through
indirect means such as TCDU or those that are described hereafter.
IV. NEUROMONITORING
172
9. MULTIMODAL MONITORING
Classical invasive methods of ICP monitoring include intraventricular or lumbar catheters connected to a
pressure transducer, as well as intraparenchymal transducers.81 Subdural and epidural systems are less accurate (Fig. 9.5). Several annoyances may impede the adequate use of those invasive devices, including infection,
hemorrhage, malfunction, obstruction, wrong positioning, or impossibility of insertion.83
There exist several noninvasive indirect methods of ICP estimation (Fig. 9.5), but none of them are frankly
validated for routine clinical use.84 Before any intracranial surgical procedure, it is recommended to have a look at
cerebral imagery. Signs of elevated ICP on the computerized tomography or magnetic resonance imaging (MRI)
include a midline shift, ventricles and cisterns of reduced size, reduced sulci size, and presence of a mass of any
origin. However, normal imagery does not exclude elevated ICP.81 Some have proposed an MRI-based measurement of ICP, through the estimation of ICP/volume changes over cardiac cycles, but with doubtful reliability.85,86
Aside from TCDU (see Section 3.7.3.4.), ultrasounds, and, more precisely, ocular echography may serve to estimate
ICP. This technique consists in measuring the optic nerve sheath diameter that correlates with ICP.87–89 Others have
proposed the use of optical coherence tomography, a noninvasive infrared light–based method, to detect changes
in optical nerve head shape according to ICP.90 Chen et al. have used the electroencephalogram power spectrum
analysis to derive ICP, but the technique still needs to be validated.91 Audiological methods of ICP estimation study
the tympanic membrane displacement in response to sound. This displacement varies as a function of ICP, necessitates a patent stapedial reflex, and no middle ear dysfunction.92 Another method consists in recording phase-shift
changes in evoked otoacoustic emissions in response to sounds of low frequencies but interindividual variability is
high.93 A novel method uses transcranial acoustic signals and head-generated sounds with rather good accuracy.94
Mean ICP is usually considered normal when ranging between 5 and 15 mmHg. Continuous ICP monitoring provides more information than a simple static number, showing variations of the ICP value as a function of the cardiac
cycle (frequency of 40–160 per min), the respiratory cycle (8–20 per min), or the vasogenic regulation of cerebral blood
volume (0.3–3 per min).95 Current concepts now advocate the individualized adjustment of ICP and CPP rather than
a fixed threshold, to take account of the underlying pathophysiology and external influencing factors such as sedation, anesthesia, or cardiac output. For example, it is recommended to individually estimate the range of CPP where
cerebral blood flow autoregulation persists. This can be done through the calculation of the correlation coefficient
between the mean arterial pressure and ICP. When this correlation coefficient is negative, it means that autoregulation
is preserved. The automatic calculation of the mean arterial pressure/ICP correlation coefficient is possible through
the use of dedicated software.96 Ideally, the individualized approach should be completed by the use of monitors of
brain oxygenation and metabolism, such as brain tissue partial pressure of oxygen monitoring or microdialysis.97
FIGURE 9.5 Summary of the different types of direct (red) and indirect (blue) means for estimating intracranial pressure. CT, computerized
X-ray tomography; EEG, electroencephalography; MRI, magnetic resonance imaging; TCDU, transcranial Doppler ultrasonography.
IV. NEUROMONITORING
ELECTROENCEPHALOGRAPHY AND DEPTH OF ANESTHESIA MONITORING
173
ELECTROENCEPHALOGRAPHY AND DEPTH OF ANESTHESIA MONITORING
Recording of central nervous system electrical activity constitutes the most direct insights into function exploration. Clinical applications of electrophysiological recordings are numerous, ranging, for example, from the identification of epileptogenic sources to in-depth recordings for the implantation of deep brain stimulation electrodes,
to the assessment of spinal cord integrity during scoliosis repair surgery, and to the monitoring of the depth of the
hypnotic component of anesthesia. The most frequently encountered modalities of electroencephalography (EEG)
are described in the following paragraphs.
Electroencephalography, Electrocorticography, and Evoked Potentials for Monitoring Nervous
System Integrity
The intraoperative use of multiple channel EEG, with electrodes placed on the scalp, is seldom practiced during
intracranial neurosurgery. The main reason is a conflicting territory with the surgical field. EEG may, however, be
of utility during several types of procedures to detect the occurrence of seizure activity or focal ischemia. During
carotid surgery, its interpretation may be limited by the user expertise, the anesthetic regimen, the temperature, and
the hemodynamic stability.98 In addition, electrodes may not cover all territories at risk of ischemia.99 Processed EEG
and the analysis of parameters such as total power, power spectrum, spectral edge frequency, or bispectral index is
generally not of supplementary help.98,100–103 Changes may occur in varying directions, the most alarming tracing
being a flat EEG.104 In that respect, neurological testing in the wake patient remains the most reliable.
In epilepsy surgery, electrocorticography is the reference method for identifying seizure foci. It consists in recording surface electrical activity through electrodes directly placed on the cortical surface. Most of hypnotic agents,
including propofol, barbiturates, halogenated vapors, and benzodiazepines, dose-dependently affect interictal epileptiform activities, therefore impeding the identification of foci.105 Ketamine and etomidate have a nonspecific facilitating effect on those activities, while opioids and dexmedetomidine have minimal effects. As a consequence, the
preferred anesthetic technique for managing those patients will be a wake technique or sedation with dexmedetomidine and/or opioids. Should general anesthesia be preferred, doses of hypnotic agents will be set at their minimum
during mapping, with the inherent risk of intraoperative awareness.
Somatosensory evoked potentials are mainly used to monitor the sensory dorsal column–medial lemniscus pathway integrity during scoliosis repair.106 They necessitate repeated and regular peripheral skin stimulations distal
to the surgical site. The cortical and subcortical signals evoked by the stimulations are recorded through scalp electrodes, and then averaged. A 50% decrease in amplitude or a 10% increase in latency as compared to baseline is considered alarming. A persistent decrease in the amplitude of somatosensory-evoked potentials can also be observed
during an intraoperative cerebral ischemia.107 Transcranial stimulation–evoked motor potentials are equally of utility
during scoliosis repair, to monitor the motor system integrity,106 as is spontaneous or triggered electromyography
for the integrity of nerve roots. Of note, evoked potentials are sensitive to the action of anesthetic agents, and particularly of halogenated vapors and nitrous oxide. Again, propofol and benzodiazepines have fewer effects, while
ketamine rather increases responses. Opioids and dexmedetomidine remain best choice in that respect. Muscle relaxants impede recording of motor-evoked potentials and electromyography.106
Hypnotic Component of Anesthesia Monitoring
During the 1990s, considerable effort has been made to develop EEG-derived dimensionless indices of the depth
of the hypnotic component of anesthesia to appreciate the degree to which consciousness is altered. The general
principle governing the calculation of those indices is first the extraction of EEG parameters that are known to be
dose-dependently influenced by hypnotic anesthetic agents and second their treatment through a mathematical
algorithm whose output is generally a number between 0 and 100.108 At least seven of those EEG-derived indices are
currently commercially available, the most famous being the Bispectral Index. Despite initial expectancies regarding
their ability to prevent unexpected intraoperative awareness with recall, they are relatively poor performant in that
respect.109 The main reasons of this lack of efficacy are first the low incidence of awareness with recall, rendering
the demonstration of a preventive effect difficult. Second, EEG-derived index parameters may be affected by several confounding factors, including electrical artifacts, interindividual variability, site of recording, delay for value
display, eventual hypothermia or hypoglycemia, cortical atrophy, age, seizures, carotid clamping, cerebral ischemia,
and interactions with other medications. Third, the parameters entering the calculation algorithm were arbitrarily
chosen and may not adequately reflect the presence or the absence of consciousness and/or connectedness to the
IV. NEUROMONITORING
174
9. MULTIMODAL MONITORING
environment.108,110 At present, EEG-derived indices are proven to prevent awareness with recall when guiding total
intravenous anesthesia, at least in patients at risk of such an event, but they are not superior to a low concentration
alarm during inhaled anesthesia.109 Nevertheless, they help individually adjusting hypnotic agent administration,
avoiding episodes of overdose,111 and predicting the moment of recovery.112 Their use in the context of intracranial
neurosurgery may not always be possible, because of surgical site inaccessibility to the adhesive scalp electrodes, but
acceptable alternatives exist (see the Clinical Pearls later).113–116
Recent advances in the understanding of the mechanisms of consciousness and its alteration by anesthetic agents
now offer the perspective of improving depth of the hypnotic component of anesthesia monitoring. It appears that
mental content is sustained by the activity of brain networks, which are composed of circumscribed brain regions
that communicate with each other.117 The functioning of these higher-order cognitive networks is differentially, specifically, and dose-dependently altered by hypnotic anesthetic agents. For example, integrate frontoparietal communication seems necessary for having conscious thoughts, with or without connectedness to the environment.118,119
New EEG-derived parameters are now proposed to specifically monitor this frontoparietal communication.120,121
Nociception Monitoring
Several parameters are now proposed to intraoperatively assess the balance between the intensity of noxious
stimulation and antinociception provided by anesthesia, but they are not necessarily derived from the EEG.108
Insofar as noxious stimulation processing involves complex pathways and multiple brain regions, in addition to
connections with the autonomic and motor nervous systems, and as the conscious aspect of painful stimulation
is lost during anesthesia, no direct witness mirroring all elements of this processing exists. Aside from the indicators that are derived from pharmacodynamic modeling,122,123 the proposed parameters can be classified into three
categories according to the response to stimulation they are markers of: the autonomic nervous system response,124
the motor response or the brain responses in the form of evoked potentials,125,126 processed EEG parameters,127–130
or the EEG and electromyogram variability.131 Examples of such parameters for the autonomic nervous system are
the pupil diameter,132 the heart rate variability,133 the pulse amplitude,134 the skin conductance,135 the skin vasomotor reflex,136,137 or a combination of them.138 Devices for the monitoring of the motor response are based on spinal
reflexes.139,140 To date, clinical utility has been demonstrated for only a few of those monitors, in terms of prediction
of movement in response to stimulation,141 better intraoperative adjustment of opioid administration,142 improved
hemodynamic stability,142 and prediction of the intensity of pain felt upon recovery.143–145 For most of them, clinical
validation is still in progress.
Again, confounding factors may impede the interpretation of recorded data. Autonomic nervous system–derived
parameters may be influenced by cardiovascular medications146 and cardiac pacing,147 regional anesthesia,148 the
intravascular volume status,149,150 the temperature, dysrhythmia, and the ventilation mode. Factors influencing the
EEG are the same as those described for the depth of hypnosis monitoring, and muscle relaxants are the main confounders of the motor response to stimulation.
MISCELLANEOUS
Other monitoring devices are part of the anesthesiologist’s armamentarium, but may be of lower utility or provide
less readily available information in the context of a neurosurgical procedure in the operating room, as compared
to the intensive care unit. For example, muscle relaxation for the management of neurosurgical procedures usually
occurs at induction of anesthesia, to facilitate tracheal intubation, but is seldom necessary thereafter. The main reason
is that the ability to observe a motor response to various kinds of stimulations is often necessary.151 As a consequence,
muscle relaxation monitoring is often omitted, except for situations where doubt on muscle relaxation exists, for
endovascular embolization procedures, where total immobility and profound neuromuscular blockade is necessary,
or for procedures of less than 2 h, where residual muscle relaxation is possible upon recovery.152
Microdialysis has been introduced into clinical practice in 1992 by Persson.153 This technology uses a flexible
catheter, whose tip contains a semipermeable membrane. The catheter is infused with a dialysate and is inserted
into the white matter. After equilibration between the dialysate and the brain, the concentration of lactate, pyruvate,
glutamate, glycerol, and glucose can be measured. The use of a 100-kDa molecular weight membrane, instead of
the traditional 20-kDa membrane, permits the study of larger molecules, including cytokines, concomitantly to the
smaller ones. Hourly sampling appears adequate to detect metabolic changes, although more frequent sampling
is possible.154,155 Insofar as microdialysis is a focal technique, brain chemistry should be interpreted according to
IV. NEUROMONITORING
INTEGRATION OF INFORMATION AND DECISION-HELPING SYSTEMS
175
the position of the catheter within the brain. Brain injury is frequently heterogeneous, and the catheter position
relative to the injury location is important. Cerebral microdialysis provides information on the underlying cerebral
metabolism and oxygen delivery. In this respect, measuring glucose, lactate, and the lactate/pyruvate ratio is now
considered more useful than measuring glutamate and glycerol.156 Microdialysis is primarily a research tool, and
it has been mainly used to acquire knowledge on some diseases pathophysiology such as traumatic brain injury or
subarachnoid hemorrhage. It has evolved into a complementary clinical tool for the management of patients on an
individual intention-to-treat basis, mainly in the intensive care unit, but its utility in the operating room has not been
evaluated yet.
The invasive measurement of oxygen partial pressure into brain parenchyma (PbtO2) allows a continuous
and real-time evaluation of the brain tissue oxygenation balance. PbtO2 values identify brain tissue at risk of
secondary injury. It can serve to optimize hemodynamics, and transfusion thresholds, and it can be a trigger for
a systemic evaluation. The Brain Trauma Foundation recommends the use of a brain oxygenation monitoring
whenever strategies of hyperventilation are employed in traumatic brain-injured patients,157 and more recent
guidelines from the Neurocritical Care Society advocate to placement of such a monitoring in any patients at
risk of cerebral ischemia.158 The PbtO2 probe is a Clark electrode and is placed within the white matter. It looks
at a small volume of brain tissue. Similarly to microdialysis, PbtO2 probe location relative to brain injury is
important and should be taken into account when interpreting PbtO2 data. Brain oxygenation relies on adequate
oxygen supply by the blood and on adequate oxygen diffusion to the tissue. Hence, PbtO2 monitoring provides
information on adequate oxygen delivery when targeting optimal CPP and can identify brain hypoxia that is
not related to inadequate perfusion when the CPP target is attained.159 Normal PbtO2 values range between 20
and 35 mmHg.160,161 Sustained PbtO2 values lower than 15 mmHg lead to cerebral ischemia and poor neurological outcome. The most recent guidelines suggest a threshold of 20 mmHg for setting up treatment.158 PbtO2 is a
complex variable because it is sensitive to the inspired oxygen fraction, the arterial oxygen partial pressure, and
the cerebral blood flow, which in turn depends on CPP and CO2 partial pressure. It can also be modified during
the hypermetabolic states such as fever, shivering, or seizures.162,163 PbtO2 must always be interpreted according
to several clinical and physiologic factors.
INTEGRATION OF INFORMATION AND DECISION-HELPING SYSTEMS
In face of devices multiplicity and huge afflux of information coming from the monitoring station (Fig. 9.6), there
might sometimes be difficulties of information integration by the practitioner, particularly in emergency situations
or in the context of fatigue. Although no existing machine surpass the human brain in its capacity of integration,
dedicated software may help identifying converging changes in physiological variables to end up with propositions
of a diagnosis. The best therapeutic option can then be chosen by the practitioner according to his/her own experience and sagacity. Such softwares are currently being developed and show promising abilities,164 but they still need
to be improved.
FIGURE 9.6 Photograph illustrating the multiplicity of monitors and screens displaying the information to the anesthesiologist during an
anesthetic procedure.
IV. NEUROMONITORING
176
9. MULTIMODAL MONITORING
FIGURE 9.7 Alternative position for the adhesive depth of the hypnotic component of anesthesia monitoring (here a spectral entropy electrode) as proposed by Nelson et al.113
CLINICAL PEARLS
Inaccessible Forehead for Electroencephalographic Monitoring
During intracranial neurosurgical procedures, the forehead is frequently not accessible to the placement of EEG
electrodes, and this may be a problem to obtain recordings for several purposes, including depth of anesthesia monitoring. The main reasons are that the surgical field may be frontal or one of the pins of the head holder is placed
there. Some authors have proposed alternative positions for the commercially available adhesive electrodes and have
compared the numbers obtained from that position with numbers obtained classically. They report reasonable concordance and acceptable bias and limits of agreement. Hence, these alternatives may be a solution, keeping in mind
that they do not offer exactly the same as when those devices are used according to the manufacturer recommendations. Alternative positions can be on the upper part of the face (Fig. 9.7),113 on the lower part of the face following
a curved line joining the lateral temporal cheek and the tip of the chin,114 more posterior, with one electrode on the
lateral temporal cheek and the others posterior to the ear,115 or even totally occipital.116
References
1. Hans P, Bonhomme V. Why we still use intravenous drugs as the basic regimen for neurosurgical anaesthesia. Curr Opin Anaesthesiol 2006;19:498–503.
2. El Beheiry H. Protecting the brain during neurosurgical procedures: strategies that can work. Curr Opin Anaesthesiol 2012;25:548–55.
3. Algarra NN, Lele AV, Prathep S, et al. Intraoperative secondary insults during orthopedic surgery in traumatic brain injury. J Neurosurg Anesthesiol 2016. epub
ahead of print.
4. Kurz A. Thermal care in the perioperative period. Best Pract Res Clin Anaesthesiol 2008;22:39–62.
5. Madrid E, Urrutia G, Roque IF, et al. Active body surface warming systems for preventing complications caused by inadvertent perioperative hypothermia in
adults. Cochrane Database Syst Rev 2016;4:CD009016.
6. Fabregas N, Bruder N. Recovery and neurological evaluation. Best Pract Res Clin Anaesthesiol 2007;21:431–47.
7. Todd MM, Hindman BJ, Clarke WR, Torner JC. Mild intraoperative hypothermia during surgery for intracranial aneurysm. N Engl J Med 2005;352:135–45.
8. Li LR, You C, Chaudhary B. Intraoperative mild hypothermia for postoperative neurological deficits in people with intracranial aneurysm. Cochrane Database Syst
Rev 2016;3:CD008445.
9. Levati A, Tommasino C, Moretti MP, et al. Giant intracranial aneurysms treated with deep hypothermia and circulatory arrest. J Neurosurg Anesthesiol
2007;19:25–30.
10. Meng L, Hou W, Chui J, Han R, Gelb AW. Cardiac output and cerebral blood flow: the integrated regulation of brain perfusion in adult humans. Anesthesiology
2015;123:1198–208.
11. Troianos CA, Hartman GS, Glas KE, et al. Special articles: guidelines for performing ultrasound guided vascular cannulation: recommendations of the American
Society of Echocardiography and the Society of Cardiovascular Anesthesiologists. Anesth Analg 2012;114:46–72.
12. Kim SH, Lilot M, Sidhu KS, et al. Accuracy and precision of continuous noninvasive arterial pressure monitoring compared with invasive arterial pressure: a
systematic review and meta-analysis. Anesthesiology 2014;120:1080–97.
13. Lin WQ, Wu HH, Su CS, et al. Comparison of continuous noninvasive blood pressure monitoring by TL-300 with standard invasive blood pressure measurement
in patients undergoing elective neurosurgery. J Neurosurg Anesthesiol 2015. epub ahead of print.
14. Ilies C, Grudev G, Hedderich J, et al. Comparison of a continuous noninvasive arterial pressure device with invasive measurements in cardiovascular postsurgical intensive care patients: a prospective observational study. Eur J Anaesthesiol 2015;32:20–8.
15. Schramm P, Tzanova I, Goock T, et al. Noninvasive hemodynamic measurements during neurosurgical procedures in sitting position. J Neurosurg Anesthesiol
2016. epub ahead of print.
IV. NEUROMONITORING
REFERENCES
177
16. Gracia I, Fabregas N. Craniotomy in sitting position: anesthesiology management. Curr Opin Anaesthesiol 2014;27:474–83.
17. Caillard A, Gayat E, Tantot A, et al. Comparison of cardiac output measured by oesophageal doppler ultrasonography or pulse pressure contour wave analysis.
Br J Anaesth 2015;114:893–900.
18. Saugel B, Cecconi M, Wagner JY, Reuter DA. Noninvasive continuous cardiac output monitoring in perioperative and intensive care medicine. Br J Anaesth
2015;114:562–75.
19. Sangkum L, Liu GL, Yu L, Yan H, Kaye AD, Liu H. Minimally invasive or noninvasive cardiac output measurement: an update. J Anesth 2016;30:461–80.
20. Ali Z, Umamaheswara Rao GS, Jaganath A. Haemodynamic changes during craniotomy monitored by a bioimpedance plethysmographic noninvasive cardiac
output monitor. J Clin Monit Comput 2009;23:157–61.
21. Sabharwal N, Rao GS, Ali Z, Radhakrishnan M. Hemodynamic changes after administration of mannitol measured by a noninvasive cardiac output monitor.
J Neurosurg Anesthesiol 2009;21:248–52.
22. Lindroos AC, Niiya T, Randell T, Niemi TT. Stroke volume-directed administration of hydroxyethyl starch (HES 130/0.4) and Ringer’s acetate in prone position
during neurosurgery: a randomized controlled trial. J Anesth 2014;28:189–97.
23. Singh S, Kuschner WG, Lighthall G. Perioperative intravascular fluid assessment and monitoring: a narrative review of established and emerging techniques.
Anesthesiol Res Pract 2011;2011:231493.
24. Michard F, Chemla D, Richard C, et al. Clinical use of respiratory changes in arterial pulse pressure to monitor the hemodynamic effects of PEEP. Am J Respir Crit
Care Med 1999;159:935–9.
25. Perel A, Pizov R, Cotev S. Systolic blood pressure variation is a sensitive indicator of hypovolemia in ventilated dogs subjected to graded hemorrhage.
Anesthesiology 1987;67:498–502.
26. Deflandre E, Bonhomme V, Hans P. Delta down compared with delta pulse pressure as an indicator of volaemia during intracranial surgery. Br J Anaesth
2008;100:245–50.
27. Ho KM. Pitfalls in haemodynamic monitoring in the postoperative and critical care setting. Anaesth Intensive Care 2016;44:14–9.
28. Forget P, Lois F, de Kock M. Goal-directed fluid management based on the pulse oximeter-derived pleth variability index reduces lactate levels and improves
fluid management. Anesth Analg 2010;111:910–4.
29. Rinehart J, Lilot M, Lee C, et al. Closed-loop assisted versus manual goal-directed fluid therapy during high-risk abdominal surgery: a case-control study with
propensity matching. Crit Care 2015;19:94–104.
30. Cannesson M, Gan TJ. PRO: Perioperative goal-directed fluid therapy is an essential element of an enhanced recovery protocol. Anesth Analg 2016;122:1258–60.
31. Yu Y, Dong J, Xu Z, Shen H, Zheng J. Pleth variability index-directed fluid management in abdominal surgery under combined general and epidural anesthesia.
J Clin Monit Comput 2015;29:47–52.
32. Hiscock R, Kumar D, Simmons SW. Systematic review and meta-analysis of method comparison studies of Masimo pulse co-oximeters (Radical-7 or Pronto-7)
and HemoCue(R) absorption spectrometers (B-Hemoglobin or 201+) with laboratory haemoglobin estimation. Anaesth Intensive Care 2015;43:341–50.
33. Lardi AM, Hirst C, Mortimer AJ, McCollum CN. Evaluation of the HemoCue for measuring intra-operative haemoglobin concentrations: a comparison with the
Coulter Max-M. Anaesthesia 1998;53:349–52.
34. Goldman M, Uzicanin S, Yi QL, Acker J, Ramirez-Arcos S. Validation and implementation of a new hemoglobinometer for donor screening at Canadian Blood
Services. Transfusion 2012;52:1607–13.
35. Rudolf J, Douglass J, Baron J, Lewandrowski K. Evaluation of the i-STAT point-of-care capillary whole blood hematocrit and hemoglobin: comparison to the
siemens RAPIDLab 1200, Sysmex XE5000, and manual spun hematocrit. Clin Chim Acta 2015;446:37–42.
36. Lamhaut L, Apriotesei R, Combes X, Lejay M, Carli P, Vivien B. Comparison of the accuracy of noninvasive hemoglobin monitoring by spectrophotometry
(SpHb) and HemoCue(R) with automated laboratory hemoglobin measurement. Anesthesiology 2011;115:548–54.
37. Kim SH, Lilot M, Murphy LS, et al. Accuracy of continuous noninvasive hemoglobin monitoring: a systematic review and meta-analysis. Anesth Analg
2014;119:332–46.
38. Crowley C, Montenegro-Bethancourt G, Solomons NW, Schumann K. Validity and correspondence of non-invasively determined hemoglobin concentrations by
two trans-cutaneous digital measuring devices. Asia Pac J Clin Nutr 2012;21:191–200.
39. Park YH, Lee JH, Song HG, Byon HJ, Kim HS, Kim JT. The accuracy of noninvasive hemoglobin monitoring using the radical-7 pulse CO-Oximeter in children
undergoing neurosurgery. Anesth Analg 2012;115:1302–7.
40. Kost GJ, Tran NK. Continuous noninvasive hemoglobin monitoring: the standard of care and future impact. Crit Care Med 2011;39:2369–71.
41. Berkow L, Rotolo S, Mirski E. Continuous noninvasive hemoglobin monitoring during complex spine surgery. Anesth Analg 2011;113:1396–402.
42. Miller RD, Ward TA, Shiboski SC, Cohen NH. A comparison of three methods of hemoglobin monitoring in patients undergoing spine surgery. Anesth Analg
2011;112:858–63.
43. Rohlwink UK, Figaji AA. Methods of monitoring brain oxygenation. Childs Nerv Syst 2010;26:453–64.
44. Macmillan CS, Andrews PJ. Cerebrovenous oxygen saturation monitoring: practical considerations and clinical relevance. Intensive Care Med 2000;26:1028–36.
45. Coles JP. Regional ischemia after head injury. Curr Opin Crit Care 2004;10:120–5.
46. Iwata M, Kawaguchi M, Inoue S, et al. Effects of increasing concentrations of propofol on jugular venous bulb oxygen saturation in neurosurgical patients under
normothermic and mildly hypothermic conditions. Anesthesiology 2006;104:33–8.
47. Liao R, Li J, Liu J. Volatile induction/maintenance of anaesthesia with sevoflurane increases jugular venous oxygen saturation and lumbar cerebrospinal fluid
pressure in patients undergoing craniotomy. Eur J Anaesthesiol 2010;27:369–76.
48. Sharma D, Siriussawakul A, Dooney N, Hecker JG, Vavilala MS. Clinical experience with intraoperative jugular venous oximetry during pediatric intracranial
neurosurgery. Paediatr Anaesth 2013;23:84–90.
49. Sharma D, Ellenbogen RG, Vavilala MS. Use of transcranial Doppler ultrasonography and jugular oximetry to optimize hemodynamics during pediatric posterior fossa craniotomy. J Clin Neurosci 2010;17:1583–4.
50. Kampf S, Schramm P, Klein KU. Transcranial doppler and near infrared spectroscopy in the perioperative period. Curr Opin Anaesthesiol 2013;26:543–8.
51. Heringlake M, Garbers C, Kabler JH, et al. Preoperative cerebral oxygen saturation and clinical outcomes in cardiac surgery. Anesthesiology 2011;114:58–69.
52. Kim MB, Ward DS, Cartwright CR, Kolano J, Chlebowski S, Henson LC. Estimation of jugular venous O2 saturation from cerebral oximetry or arterial O2 saturation during isocapnic hypoxia. J Clin Monit Comput 2000;16:191–9.
53. Bevan PJ. Should cerebral near-infrared spectroscopy be standard of care in adult cardiac surgery? Heart Lung Circ 2015;24:544–50.
54. Nielsen HB. Systematic review of near-infrared spectroscopy determined cerebral oxygenation during non-cardiac surgery. Front Physiol 2014;5:93–107.
55. Cottrell JE, Hartung J. Developmental disability in the young and postoperative cognitive dysfunction in the elderly after anesthesia and surgery: do data justify
changing clinical practice? Mt Sinai J Med 2012;79:75–94.
56. Sorensen H, Grocott HP, Secher NH. Near infrared spectroscopy for frontal lobe oxygenation during non-vascular abdominal surgery. Clin Physiol Funct Imaging
2015. epub ahead of print.
57. Zheng F, Sheinberg R, Yee MS, Ono M, Zheng Y, Hogue CW. Cerebral near-infrared spectroscopy monitoring and neurologic outcomes in adult cardiac surgery
patients: a systematic review. Anesth Analg 2013;116:663–76.
58. Orihashi K, Sueda T, Okada K, Imai K. Near-infrared spectroscopy for monitoring cerebral ischemia during selective cerebral perfusion. Eur J Cardiothorac Surg
2004;26:907–11.
IV. NEUROMONITORING
178
9. MULTIMODAL MONITORING
59. Prabhune A, Sehic A, Spence PA, Church T, Edmonds Jr HL. Cerebral oximetry provides early warning of oxygen delivery failure during cardiopulmonary
bypass. J Cardiothorac Vasc Anesth 2002;16:204–6.
60. Vernick WJ, Oware A. Early diagnosis of superior vena cava obstruction facilitated by the use of cerebral oximetry. J Cardiothorac Vasc Anesth 2011;25:1101–3.
61. Ali AM, Green D, Zayed H, Halawa M, El-Sakka K, Rashid HI. Cerebral monitoring in patients undergoing carotid endarterectomy using a triple assessment
technique. Interact Cardiovasc Thorac Surg 2011;12:454–7.
62. Ritter JC, Green D, Slim H, Tiwari A, Brown J, Rashid H. The role of cerebral oximetry in combination with awake testing in patients undergoing carotid endarterectomy under local anaesthesia. Eur J Vasc Endovasc Surg 2011;41:599–605.
63. Pennekamp CW, Immink RV, den Ruijter HM, et al. Near-infrared spectroscopy can predict the onset of cerebral hyperperfusion syndrome after carotid endarterectomy. Cerebrovasc Dis 2012;34:314–21.
64. Matsumoto S, Nakahara I, Higashi T, et al. Near-infrared spectroscopy in carotid artery stenting predicts cerebral hyperperfusion syndrome. Neurology
2009;72:1512–8.
65. Aaslid R, Markwalder TM, Nornes H. Noninvasive transcranial doppler ultrasound recording of flow velocity in basal cerebral arteries. J Neurosurg 1982;57:769–74.
66. Gomez-Choco M, Schreiber SJ, Weih M, Doepp F, Valdueza JM. Delayed transcranial echo-contrast bolus arrival in unilateral internal carotid artery stenosis and
occlusion. Ultrasound Med Biol 2015;41:1827–34.
67. Arbona FL, Khabiri B, Norton JA. Ultrasound basics for the busy novice practitioner. Int Anesthesiol Clin 2011;49:34–51.
68. Bathala L, Mehndiratta MM, Sharma VK. Transcranial doppler: technique and common findings (Part 1). Ann Indian Acad Neurol 2013;16:174–9.
69. Naqvi J, Yap KH, Ahmad G, Ghosh J. Transcranial doppler ultrasound: a review of the physical principles and major applications in critical care. Int J Vasc Med
2013;2013:629378.
70. Adams R, McKie V, Nichols F, et al. The use of transcranial ultrasonography to predict stroke in sickle cell disease. N Engl J Med 1992;326:605–10.
71. Adams RJ, McKie VC, Hsu L, et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial doppler
ultrasonography. N Engl J Med 1998;339:5–11.
72. Sharma AK, Bathala L, Batra A, Mehndiratta MM, Sharma VK. Transcranial doppler: techniques and advanced applications: part 2. Ann Indian Acad Neurol
2016;19:102–7.
73. Kumar G, Shahripour RB, Harrigan MR. Vasospasm on transcranial doppler is predictive of delayed cerebral ischemia in aneurysmal subarachnoid hemorrhage:
a systematic review and meta-analysis. J Neurosurg 2016;124:1257–64.
74. Gautier C, Deklunder G. Echodoppler transcranien: methodologie et applications cliniques. 1 ed. Sauramps Medical; 2010.
75. Vora YY, Suarez-Almazor M, Steinke DE, Martin ML, Findlay JM. Role of transcranial doppler monitoring in the diagnosis of cerebral vasospasm after subarachnoid hemorrhage. Neurosurgery 1999;44:1237–47.
76. Geeraerts T, Ract C, Duranteau J, Vigue B. Transcranial doppler in neurosurgery. Neurochirurgie 2008;54:731–8.
77. Sloan MA, Alexandrov AV, Tegeler CH, et al. Assessment: transcranial doppler ultrasonography: report of the therapeutics and technology assessment subcommittee of the American Academy of Neurology. Neurology 2004;62:1468–81.
78. Markus HS, Harrison MJ. Estimation of cerebrovascular reactivity using transcranial doppler, including the use of breath-holding as the vasodilatory stimulus.
Stroke 1992;23:668–73.
79. Vigue B, Tazaroute K, Geeraerts T, Ract C, Duranteau J. Transcranial doppler in the intensive care unit. Reanimation 2007;16:538–45.
80. Cardim D, Robba C, Bohdanowicz M, et al. Non-invasive monitoring of intracranial pressure using transcranial doppler ultrasonography: is it possible? Neurocrit
Care 2016. epub ahead of print.
81. Kristiansson H, Nissborg E, Bartek Jr J, Andresen M, Reinstrup P, Romner B. Measuring elevated intracranial pressure through noninvasive methods: a review
of the literature. J Neurosurg Anesthesiol 2013;25:372–85.
82. Krakauskaite S, Petkus V, Bartusis L, et al. Accuracy, precision, sensitivity, and specificity of noninvasive ICP absolute value measurements. Acta Neurochir Suppl
2016;122:317–21.
83. Dimitriou J, Levivier M, Gugliotta M. Comparison of complications in patients receiving different types of intracranial pressure monitoring: a retrospective study
in a single center in Switzerland. World Neurosurg 2016;89:641–6.
84. Kawoos U, McCarron RM, Auker CR, Chavko M. Advances in intracranial pressure monitoring and its significance in managing traumatic brain injury. Int J Mol
Sci 2015;16:28979–97.
85. Alperin NJ, Lee SH, Loth F, Raksin PB, Lichtor T. MR-Intracranial pressure (ICP): a method to measure intracranial elastance and pressure noninvasively by
means of MR imaging: baboon and human study. Radiology 2000;217:877–85.
86. Marshall I, MacCormick I, Sellar R, Whittle I. Assessment of factors affecting MRI measurement of intracranial volume changes and elastance index. Br J
Neurosurg 2008;22:389–97.
87. Cammarata G, Ristagno G, Cammarata A, Mannanici G, Denaro C, Gullo A. Ocular ultrasound to detect intracranial hypertension in trauma patients. J Trauma
2011;71:779–81.
88. Geeraerts T, Merceron S, Benhamou D, Vigue B, Duranteau J. Non-invasive assessment of intracranial pressure using ocular sonography in neurocritical care
patients. Intensive Care Med 2008;34:2062–7.
89. Geeraerts T, Duranteau J, Benhamou D. Ocular sonography in patients with raised intracranial pressure: the papilloedema revisited. Crit Care 2008;12:150.
90. Anand A, Pass A, Urfy MZ, et al. Optical coherence tomography of the optic nerve head detects acute changes in intracranial pressure. J Clin Neurosci 2016;29:73–6.
91. Chen H, Wang J, Mao S, Dong W, Yang H. A new method of intracranial pressure monitoring by EEG power spectrum analysis. Can J Neurol Sci 2012;39:483–7.
92. Shimbles S, Dodd C, Banister K, Mendelow AD, Chambers IR. Clinical comparison of tympanic membrane displacement with invasive ICP measurements. Acta
Neurochir Suppl 2005;95:197–9.
93. Buki B, Avan P, Lemaire JJ, Dordain M, Chazal J, Ribari O. Otoacoustic emissions: a new tool for monitoring intracranial pressure changes through stapes displacements. Hear Res 1996;94:125–39.
94. Levinsky A, Papyan S, Weinberg G, Stadheim T, Eide PK. Non-invasive estimation of static and pulsatile intracranial pressure from transcranial acoustic signals.
Med Eng Phys 2016;38:477–84.
95. Kasprowicz M, Lalou DA, Czosnyka M, Garnett M, Czosnyka Z. Intracranial pressure, its components and cerebrospinal fluid pressure-volume compensation.
Acta Neurol Scand 2015. epub ahead of print.
96. Steiner LA, Czosnyka M, Piechnik SK, et al. Continuous monitoring of cerebrovascular pressure reactivity allows determination of optimal cerebral perfusion
pressure in patients with traumatic brain injury. Crit Care Med 2002;30:733–8.
97. Makarenko S, Griesdale DE, Gooderham P, Sekhon MS. Multimodal neuromonitoring for traumatic brain injury: a shift towards individualized therapy. J Clin
Neurosci 2016;26:8–13.
98. Bonhomme V, Desiron Q, Lemineur T, Brichant JF, Dewandre PY, Hans P. Bispectral index profile during carotid cross clamping. J Neurosurg Anesthesiol
2007;19:49–55.
99. Kresowik TF, Worsey MJ, Khoury MD, et al. Limitations of electroencephalographic monitoring in the detection of cerebral ischemia accompanying carotid
endarterectomy. J Vasc Surg 1991;13:439–43.
100. Rampil IJ, Correll JW, Rosenbaum SH, Quest DO, Holzer JA. Computerized electroencephalogram monitoring and carotid artery shunting. Neurosurgery
1983;13:276–9.
IV. NEUROMONITORING
REFERENCES
179
101. Rampil IJ, Holzer JA, Quest DO, Rosenbaum SH, Correll JW. Prognostic value of computerized EEG analysis during carotid endarterectomy. Anesth Analg
1983;62:186–92.
102. Fiori L, Parenti G. Electrophysiological monitoring for selective shunting during carotid endarterectomy. J Neurosurg Anesthesiol 1995;7:168–73.
103. Visser GH, Wieneke GH, van Huffelen AC. Carotid endarterectomy monitoring: patterns of spectral EEG changes due to carotid artery clamping. Clin Neurophysiol
1999;110:286–94.
104. Merat S, Levecque JP, Le GY, Diraison Y, Brinquin L, Hoffmann JJ. BIS monitoring may allow the detection of severe cerebral ischemia. Can J Anaesth
2001;48:1066–9.
105. Chui J, Manninen P, Valiante T, Venkatraghavan L. The anesthetic considerations of intraoperative electrocorticography during epilepsy surgery. Anesth Analg
2013;117:479–86.
106. Glover CD, Carling NP. Neuromonitoring for scoliosis surgery. Anesthesiol Clin 2014;32:101–14.
107. Manninen PH, Tan TK, Sarjeant RM. Somatosensory evoked potential monitoring during carotid endarterectomy in patients with a stroke. Anesth Analg
2001;93:39–44.
108. Marchant N, Sanders R, Sleigh J, et al. How electroencephalography serves the anesthesiologist. Clin EEG Neurosci 2014;45:22–32.
109. Avidan MS, Mashour GA. Prevention of intraoperative awareness with explicit recall: making sense of the evidence. Anesthesiology 2013;118:449–56.
110. Sanders RD, Tononi G, Laureys S, Sleigh JW. Unresponsiveness not equal unconsciousness. Anesthesiology 2012;116:946–59.
111. Klopman MA, Sebel PS. Cost-effectiveness of bispectral index monitoring. Curr Opin Anaesthesiol 2011;24:177–81.
112. White PF. Use of cerebral monitoring during anaesthesia: effect on recovery profile. Best Pract Res Clin Anaesthesiol 2006;20:181–9.
113. Nelson P, Nelson JA, Chen AJ, Kofke WA. An alternative position for the BIS-Vista montage in frontal approach neurosurgical cases. J Neurosurg Anesthesiol
2013;25:135–42.
114. Lee SY, Kim YS, Lim BG, Kim H, Kong MH, Lee IO. Comparison of bispectral index scores from the standard frontal sensor position with those from an alternative mandibular position. Korean J Anesthesiol 2014;66:267–73.
115. Akavipat P, Hungsawanich N, Jansin R. Alternative placement of bispectral index electrode for monitoring depth of anesthesia during neurosurgery. Acta Med
Okayama 2014;68:151–5.
116. Shiraishi T, Uchino H, Sagara T, Ishii N. A comparison of frontal and occipital bispectral index values obtained during neurosurgical procedures. Anesth Analg
2004;98:1773–5.
117. Bonhomme VL, Boveroux P, Brichant JF, Laureys S, Boly M. Neural correlates of consciousness during general anesthesia using functional magnetic resonance
imaging (fMRI). Arch Ital Biol 2012;150:155–63.
118. Lee U, Kim S, Noh GJ, Choi BM, Hwang E, Mashour GA. The directionality and functional organization of frontoparietal connectivity during consciousness and
anesthesia in humans. Conscious Cogn 2009;18:1069–78.
119. Hudetz AG. Feedback suppression in anesthesia. Is it reversible? Conscious Cogn 2009;18:1079–81.
120. Jordan D, Stockmanns G, Kochs EF, Pilge S, Schneider G. Electroencephalographic order pattern analysis for the separation of consciousness and unconsciousness: an analysis of approximate entropy, permutation entropy, recurrence rate, and phase coupling of order recurrence plots. Anesthesiology
2008;109:1014–22.
121. Ku SW, Lee U, Noh GJ, Jun IG, Mashour GA. Preferential inhibition of frontal-to-parietal feedback connectivity is a neurophysiologic correlate of general anesthesia in surgical patients. PLoS One 2011;6:e25155.
122. Luginbuhl M, Schumacher PM, Vuilleumier P, et al. Noxious stimulation response index: a novel anesthetic state index based on hypnotic-opioid interaction.
Anesthesiology 2010;112:872–80.
123. von Dincklage F, Correll C, Schneider MH, Rehberg B, Baars JH. Utility of nociceptive flexion reflex threshold, bispectral index, composite variability index and
noxious stimulation response index as measures for nociception during general anaesthesia. Anaesthesia 2012;67:899–905.
124. De Jonckheere J, Bonhomme V, Jeanne M, et al. Physiological signal processing for individualized anti-nociception management during general anesthesia: a
review. Yearb Med Inform 2015;10:95–101.
125. Kochs E, Treede RD, Schulte am EJ, Bromm B. Modulation of pain-related somatosensory evoked potentials by general anesthesia. Anesth Analg 1990;71:225–30.
126. Bonhomme V, Llabres V, Dewandre PY, Brichant JF, Hans P. Combined use of bispectral index and A-Line autoregressive index to assess anti-nociceptive component of balanced anaesthesia during lumbar arthrodesis. Br J Anaesth 2006;96:353–60.
127. Liley DT, Sinclair NC, Lipping T, Heyse B, Vereecke HE, Struys MM. Propofol and remifentanil differentially modulate frontal electroencephalographic activity.
Anesthesiology 2010;113:292–304.
128. MacKay EC, Sleigh JW, Voss LJ, Barnard JP. Episodic waveforms in the electroencephalogram during general anaesthesia: a study of patterns of response to
noxious stimuli. Anaesth Intensive Care 2010;38:102–12.
129. Jensen EW, Valencia JF, Lopez A, et al. Monitoring hypnotic effect and nociception with two EEG-derived indices, qCON and qNOX, during general anaesthesia.
Acta Anaesthesiol Scand 2014;58:933–41.
130. Melia U, Vallverdu M, Borrat X, et al. Prediction of nociceptive responses during sedation by linear and non-linear measures of EEG signals in high frequencies.
PLoS One 2015;10:e0123464.
131. Mathews DM, Clark L, Johansen J, Matute E, Seshagiri CV. Increases in electroencephalogram and electromyogram variability are associated with an increased
incidence of intraoperative somatic response. Anesth Analg 2012;114:759–70.
132. Barvais L, Engelman E, Eba JM, Coussaert E, Cantraine F, Kenny GN. Effect site concentrations of remifentanil and pupil response to noxious stimulation. Br J
Anaesth 2003;91:347–52.
133. Jeanne M, Clement C, De JJ, Logier R, Tavernier B. Variations of the analgesia nociception index during general anaesthesia for laparoscopic abdominal surgery.
J Clin Monit Comput 2012;26:289–94.
134. Bergmann I, Gohner A, Crozier TA, et al. Surgical pleth index-guided remifentanil administration reduces remifentanil and propofol consumption and shortens
recovery times in outpatient anaesthesia. Br J Anaesth 2013;110:622–8.
135. Shimoda O, Ikuta Y, Sakamoto M, Terasaki H. Skin vasomotor reflex predicts circulatory responses to laryngoscopy and intubation. Anesthesiology 1998;88:297–304.
136. Gjerstad AC, Storm H, Hagen R, Huiku M, Qvigstad E, Raeder J. Skin conductance or entropy for detection of non-noxious stimulation during different clinical
levels of sedation. Acta Anaesthesiol Scand 2007;51:1–7.
137. Gjerstad AC, Storm H, Hagen R, Huiku M, Qvigstad E, Raeder J. Comparison of skin conductance with entropy during intubation, tetanic stimulation and emergence from general anaesthesia. Acta Anaesthesiol Scand 2007;51:8–15.
138. Martini CH, Boon M, Broens SJ, et al. Ability of the nociception level, a multiparameter composite of autonomic signals, to detect noxious stimuli during propofol-remifentanil anesthesia. Anesthesiology 2015;123:524–34.
139. von Dincklage F, Hackbarth M, Mager R, Rehberg B, Baars JH. Monitoring of the responsiveness to noxious stimuli during anaesthesia with propofol and remifentanil by using RIII reflex threshold and bispectral index. Br J Anaesth 2010;104:201–8.
140. Baars JH, Mager R, Dankert K, Hackbarth M, von DF, Rehberg B. Effects of sevoflurane and propofol on the nociceptive withdrawal reflex and on the H reflex.
Anesthesiology 2009;111:72–81.
141. Gruenewald M, Herz J, Schoenherr T, Thee C, Steinfath M, Bein B. Measurement of the nociceptive balance by analgesia nociception index and surgical pleth
index during sevoflurane-remifentanil anesthesia. Minerva Anestesiol 2015;81:480–9.
IV. NEUROMONITORING
180
9. MULTIMODAL MONITORING
142. Chen X, Thee C, Gruenewald M, et al. Comparison of surgical stress index-guided analgesia with standard clinical practice during routine general anesthesia: a
pilot study. Anesthesiology 2010;112:1175–83.
143. Boselli E, Bouvet L, Begou G, et al. Prediction of immediate postoperative pain using the analgesia/nociception index: a prospective observational study. Br J
Anaesth 2014;112:715–21.
144. Boselli E, Daniela-Ionescu M, Begou G, et al. Prospective observational study of the non-invasive assessment of immediate postoperative pain using the analgesia/nociception index (ANI). Br J Anaesth 2013;111:453–9.
145. Aissou M, Snauwaert A, Dupuis C, Atchabahian A, Aubrun F, Beaussier M. Objective assessment of the immediate postoperative analgesia using pupillary reflex
measurement: a prospective and observational study. Anesthesiology 2012;116:1006–12.
146. Ahonen J, Jokela R, Uutela K, Huiku M. Surgical stress index reflects surgical stress in gynaecological laparoscopic day-case surgery. Br J Anaesth 2007;98:456–61.
147. Hocker J, Broch O, Grasner JT, et al. Surgical stress index in response to pacemaker stimulation or atropine. Br J Anaesth 2010;105:150–4.
148. Ilies C, Gruenewald M, Ludwigs J, et al. Evaluation of the surgical stress index during spinal and general anaesthesia. Br J Anaesth 2010;105:533–7.
149. Bonhomme V, Uutela K, Hans G, et al. Comparison of the surgical pleth indexTM with haemodynamic variables to assess nociception-anti-nociception balance
during general anaesthesia. Br J Anaesth 2010;106:101–11.
150. Hans P, Verscheure S, Uutela K, Hans G, Bonhomme V. Effect of a fluid challenge on the surgical pleth index during stable propofol-remifentanil anaesthesia.
Acta Anaesthesiol Scand 2012;56:787–96.
151. Hans P, Bonhomme V. Muscle relaxants in neurosurgical anaesthesia: a critical appraisal. Eur J Anaesthesiol 2003;20:600–5.
152. Hans P, Welter P, Dewandre PY, Brichant JF, Bonhomme V. Recovery from neuromuscular block after an intubation dose of cisatracurium and rocuronium in
lumbar disc surgery. Acta Anaesthesiol Belg 2004;55:129–33.
153. Persson L, Hillered L. Chemical monitoring of neurosurgical intensive care patients using intracerebral microdialysis. J Neurosurg 1992;76:72–80.
154. Skjoth-Rasmussen J, Schulz M, Kristensen SR, Bjerre P. Delayed neurological deficits detected by an ischemic pattern in the extracellular cerebral metabolites in
patients with aneurysmal subarachnoid hemorrhage. J Neurosurg 2004;100:8–15.
155. Bhatia R, Hashemi P, Razzaq A, et al. Application of rapid-sampling, online microdialysis to the monitoring of brain metabolism during aneurysm surgery.
Neurosurgery 2006;58:ONS-20.
156. Hutchinson PJ, Jalloh I, Helmy A, et al. Consensus statement from the 2014 International Microdialysis Forum. Intensive Care Med 2015;41:1517–28.
157. Bratton SL, Chestnut RM, Ghajar J, et al. Guidelines for the management of severe traumatic brain injury. X. Brain oxygen monitoring and thresholds.
J Neurotrauma 2007;24(Suppl. 1):S65–70.
158. Le Roux P, Menon DK, Citerio G, et al. The International Multidisciplinary Consensus Conference on multimodality monitoring in neurocritical care: a list of
recommendations and additional conclusions: a statement for healthcare professionals from the neurocritical care society and the European society of intensive
care medicine. Neurocrit Care 2014;21(Suppl. 2):S282–96.
159. Roh D, Park S. Brain multimodality monitoring: updated perspectives. Curr Neurol Neurosci Rep 2016;16:56.
160. Pennings FA, Schuurman PR, van den Munckhof P, Bouma GJ. Brain tissue oxygen pressure monitoring in awake patients during functional neurosurgery: the
assessment of normal values. J Neurotrauma 2008;25:1173–7.
161. Hoffman WE, Charbel FT, Edelman G. Brain tissue oxygen, carbon dioxide, and pH in neurosurgical patients at risk for ischemia. Anesth Analg 1996;82:582–6.
162. Stiefel MF, Udoetuk JD, Spiotta AM, et al. Conventional neurocritical care and cerebral oxygenation after traumatic brain injury. J Neurosurg 2006;105:568–75.
163. Jaeger M, Schuhmann MU, Soehle M, Nagel C, Meixensberger J. Continuous monitoring of cerebrovascular autoregulation after subarachnoid hemorrhage by
brain tissue oxygen pressure reactivity and its relation to delayed cerebral infarction. Stroke 2007;38:981–6.
164. Defresne A, Harrison M, Bonhomme V. Clinical pertinence and diagnostic accuracy of an evidence-based monitoring system: custos(c). Eur J Anaesthesiol 2016.
epub ahead of print.
165. Singh A, Dubey A, Sonker A, Chaudhary R. Evaluation of various methods of point-of-care testing of haemoglobin concentration in blood donors. Blood Transfus
2015;13:233–9.
166. Maslow A, Bert A, Singh A, Sweeney J. Point-of-Care hemoglobin/hematocrit testing: comparison of methodology and technology. J Cardiothorac Vasc Anesth
2016;30:352–62.
167. Ardin S, Stormer M, Radojska S, Oustianskaia L, Hahn M, Gathof BS. Comparison of three noninvasive methods for hemoglobin screening of blood donors.
Transfusion 2015;55:379–87.
IV. NEUROMONITORING
S E C T I O N
V
POSITIONS IN NEUROSURGERY
This page intentionally left blank
C H A P T E R
10
Positioning in Neurosurgery
G. Singh
Christian Medical College, Vellore, India
O U T L I N E
Introduction
184
Historical Background
184
Principles of Positioning
184
The Conduct of Positioning
185
Surgical Approach for Craniotomies
186
Positioning for Craniotomy
Positioning of the Head
Alignment of the Head and Neck
Monitoring During Head and Neck Positioning
Fixation of the Head for Craniotomy
187
187
187
187
188
Andrew’s Hinder–Binder Frame (OSI,
Union City, CA, USA)
Wilson Supporting Frame (OSI, Union City,
CA, USA)
Equipment for Stabilizing the Head
Dough Nut–Shaped Foam/Gel Pads
Prone Pillows
Gardner-Wells Tongs, Traction Systems
Pulmonary Compliance in the Prone Position
Position-Related Factors Affecting Blood Loss in
Spinal Procedures in Prone Position
Alignment of the Spine for Procedures of the Spine
Positioning of the Head for Procedures of the Spine
Hemodynamic Monitoring in the Prone Position
Problems Associated With Prone Position
Increased Intra-Abdominal Pressure in the
Prone Position
Nerve Palsies/Neuropraxia
Pressure Sores
Edema of the Face
Venous Air Embolism
Endotracheal Tube Displacement
Perioperative Vision Loss
Ischemic Optic Neuropathy
Central Retinal Artery Occlusion (Headrest
Syndrome)
Cortical Blindness
Acute Angle Closure Glaucoma
Positions Used for Craniotomies
189
Supine Position or Dorsal Decubitus Position
189
Lateral Position
190
Park Bench Position
190
Semilateral Position (Janetta Position)
191
Prone Position
191
Concorde Position
191
Three-Quarters Prone (Lateral Oblique
or Semiprone)
192
Transoral Approach
192
Approach for Transnasal Transsphenoidal Surgery
192
Sitting Position
193
Complications Associated With Sitting Position 194
Contraindications to Sitting Position
194
Surgical Approach for Procedures of the Spine
195
Patient Positioning for Spinal Procedures
Equipment Required for Prone Positioning
Frames Used for Positioning the Body in Prone
Relton and Hall Four Poster Frame (1969,
Imperial Surgicals, Quebec, Canada)
195
195
195
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00010-5
196
196
197
197
197
197
198
198
198
198
199
199
199
199
200
201
201
201
201
201
201
202
202
Conclusion
203
Abbreviations
203
References
204
196
183
© 2017 Elsevier Inc. All rights reserved.
184
10. POSITIONING IN NEUROSURGERY
INTRODUCTION
Positioning is an important aspect of the perioperative care in a neurosurgical patient. The prolonged duration
of neurosurgical procedures and the need for precise localization of pathological lesions demands that the patient
is in a physiologically optimal, physically safe and comfortable position which offers the best approach to the neurosurgeon and access to the anesthesiologist. It must also ensure that the neurosurgeon is able to approach the area
of interest with ease and minimal injury to the normal brain. Often it is achieved by positions, which are a compromise between a comfortable position for the patient and the surgeon. It is also desirable that the intended position
complements the anesthetic and surgical goals such as minimizing intracranial pressure (ICP), reduction of brain
retraction, providing a clear and bloodless field, and avoiding venous obstruction. Meticulous positioning is critical
in the overall planning and successful outcome of any neurosurgical procedure. Inadequate planning or execution
during positioning can have serious consequences.
HISTORICAL BACKGROUND
Sir Victor Horsley, the pioneer of neurological surgery described the use of a separate headrest for immobilizing
the head for positioning during neurosurgical procedures. Although Harvey Cushing the father of neurosurgery did
not feel the need for a separate headrest, he emphasized the need for elevating the head so as to decrease the venous
pressure and described the use of pillows and loosely filled long sandbags to elevate the head end. Krause the
German Neurosurgeon described the need for the head of the table to be able to change its position on its transverse
axis in such a way should there be an excessive bleeding, it is possible to elevate the head end. Frazier introduced the
use of headrest attached to the operating table for surgeries performed for cerebellar tumors in the prone position.
Later, Cushing developed a horse shoe headrest for use in the prone position. In 1916, Dr. Martel, who pioneered in
sitting position, introduced the special chair and head fixation holder for surgeries performed in the sitting position.1
Over the past century, a lot of positions have been tried by trial and error. Various adjuncts to positioning have also
been tried and many refinements of these further the cause of safety. In 1950, Moore and Edmunds2 described the
first prone position frame for use in spinal procedures following which multiple frames have been designed. Frank
Mayfield and George Kees developed the Mayfield horseshoe and general purpose headrest to stabilize the head.
Subsequently in 1973, the Mayfield Kees (MFK), three pins skull clamp which is the most common and widely used
in neurosurgical practice to rigidly fix the patient’s head during craniotomy was introduced.
PRINCIPLES OF POSITIONING
A precise and scrupulous approach should be undertaken during positioning of the patient for neurosurgical
procedures to avoid potential complications. Since neurosurgical procedures often involve positions which have
multiple effects on the cardiopulmonary systems, it should be proceeded to in a gradual fashion with vigilant monitoring of the hemodynamic status of the patient. Moreover, the prolonged duration, varying hemodynamic status,
mechanical pressure, preexisting weakness, and immobility makes the patient more vulnerable to tissue damage.3 It
is also essential to assess patients at risk to see if they will tolerate the intended position. Preoperative radiographs
and MRI may be helpful in deciding position of the neck.
Although the responsibility of positioning the patient is primarily shared by the neurosurgeon and the attending
anesthesiologist, every member of the surgical team plays a vital role. Positioning is usually attended to by the circulating nurse, neurosurgeon, anesthesiologist, theater technologist, and anesthetic assistants. It is initiated soon after
induction of anesthesia and placement of adequate venous access and essential monitoring devices.
The neurosurgeon decides the approach to the surgical site and position required for the same depending on the
location of the tumor and the intended route for its approach. The anesthesiologist ensures adequate depth of anesthesia, hemodynamic stability, adequacy of oxygenation, securing the vascular and invasive accesses during and
after positioning. They also ensure that all the detached monitoring cables are reconnected and checked to verify
optimal performance. The circulating nurse safeguards the dignity and safety while ensuring comfort of the anesthetized patient. Although, these individual responsibilities may vary with each institution, it is usually the circulating
nurse who is responsible for keeping the positioning devices and equipment ready and ensuring adequate personnel
are available for the change in position. Each member of the team has a distinctive role, and it is important that there
is a close coordination between the different members of the team to avoid potential mishaps during positioning.3
V. POSITIONS IN NEUROSURGERY
THE CONDUCT OF POSITIONING
185
THE CONDUCT OF POSITIONING
Once all the preparations are made and lines, catheter, monitors are secured, members of the team take their
respective positions. IV lines, A-lines, endotracheal tube (ETT), monitoring cables are disconnected creating a blackout state and a period of hypoventilation. It is necessary that the period of blackout state is kept to the minimum and
there is at least pulse oximetry ECG and blood pressure monitoring during this period. Patency of external ventricular drains and chest tubes should be maintained. The transfer should be swift, smooth, and coordinated.
After achieving the final desired position, adequate access, optimal function of the IV lines, A -lines and monitors
are checked and confirmed. Special attention is paid to the hemodynamic stability and respiratory compliance in the
newly achieved position. Normal alignment of the head and neck with the body should be maintained. The endotracheal tube migration (either in to the bronchus or outside) should be ruled out and then stabilized to avoid kinking.
The cuff pressure needs to be rechecked in the new position as there may be changes and kept within the normal
range.4 To ensure optimal safety, a repositioning checklist is ideal.5
Table 10.1 describes a comprehensive repositioning checklist to be performed after achieving the desired position.
The anesthesiologist ensures that the eyes are free from direct pressure to minimize ocular damage. Optimal head
end elevation is provided to reduce venous pressure. All potential pressure points (sacrum, ischium, trochanter, and
heel) are well padded by the placement of viscoelastic (gel) pads so as to redistribute mechanical pressure. Any contact of the patient’s skin with metal surfaces should be avoided to prevent burns related to the use of diathermy. Since
there is a rapid fall of temperature following induction of general anesthesia, active efforts should be undertaken to
reduce fall in temperature by limiting the exposure of the skin to the minimum and use of temperature-regulating
blankets or forced air warming devices. Warm and humidified inspired gas and fluids should be used combined
with temperature monitoring. Since many of the neurosurgical patients have weakness related to immobility and
use of anticoagulants is not preferred except under special circumstances, it is important to prevent thromboembolic complications. Thromboembolic deterrent stockings and sequential compression devices are applied in patients
undergoing prolonged procedures. The operating table is an important accessory. The position and stability of the
operating table should be checked by the operating nurse before the patient is placed on the table. Optimal functioning of the remote control should be confirmed since this is a vital adjunct for subsequent changes in position.6 It is
important to ensure that no body part extends beyond the edge of the OR table or positioning devices. The OR table
safety strap must not be secured in such a manner that it is not too tightly placed across the patient in order to avoid
pressure. It must be possible to comfortably insert two fingers under the mid-section of the safety strap to ensure
that it is safely applied. The final position of the patient should be acceptable to the surgeon and anesthesiologist,
providing easy access to operating microscope, navigation systems, and radiological imaging.
TABLE 10.1 Repositioning Checklist
A
Airway
ETT—Connected and incorrect position Confirmation of capnography trace
Auscultation of both axilla
B
Breathing
Check PAP, VTE (compliance)
Ventilation
C
Circulation
Heart rate, blood pressure, ECG
D
Devices
All monitoring devices connected and working, SPO2, ETCO2, temperature, BIS, Warmers, NM monitor,
intraoperative neuromonitoring devices
E
Extremity and eyes
Pulses are checked, eyes taped and free of pressure
F
Flows
Oxygen concentration optimized and anesthetic gas levels are checked
G
Gel pads
Padding of pressure points
H
Head
Head end elevation. Ensure two fingerbreadths between chin and mentum
Alignment of head and neck checked
I
Intravenous access
IV lines checked and extensions obtained
Infusions restarted
J
Joints
Appropriately positioned
BIS, bispectral index; ETCO2, end tidal carbon dioxide; ETT, endotracheal tube; IV, intravenous; NM, neuromuscular; PAP, pulmonary arterial pressure;
SPO2, peripheral saturation in oxygen; VTE, venous thromboembolism.
V. POSITIONS IN NEUROSURGERY
186
10. POSITIONING IN NEUROSURGERY
SURGICAL APPROACH FOR CRANIOTOMIES
There are about six standard craniotomies described.
t
t
t
t
t
t
Pterional craniotomy (frontotemporal)
Temporal and subtemporal craniotomy
Anterior parasagittal and subfrontal approach
Posterior parasagittal craniotomy
Midline suboccipital craniotomy
Lateral suboccipital approach
Table 10.2 describes the standard surgical approach for craniotomies, their variants, and the various options of
body positions for each approach.
TABLE 10.2 Surgical Approach for Craniotomies
Type of Craniotomy
Location of Lesion
Body Position
Pterional
Lesions in anterior and
Supine
middle cranial fossa, anterior
circulation aneurysms
Head Position
Variants
Flexion, 45° of head rotation
toward contralateral shoulder,
shoulder roll under ipsilateral
shoulder
Cranio–orbito–zygomatic
approach
Fronto temporal
craniotomy
Subfrontal approach
Temporal and
subtemporal
Petrous apex pathology,
basilar top aneurysms,
middle cranial fossa lesions
Supine
Flexion, 90° rotation, roll under
ipsilateral shoulder
Lateral
Lateral flexion of neck,
dependent ear toward ipsilateral
shoulder (gravity facilitates
retraction of temporal lobe)
DACAs
Third and lateral ventricular
tumors
Intraventricular diseases
Interhemispheric approach
Supine
Neutral midline with degrees
of flexion depending upon the
surgical target
Lateral decubitus
Head tilted up
Anterior cranial fossa
Supine
Neutral with extension until the
brow is at the superior point (to
facilitate frontal lobe retraction)
Supine oblique
More flexion than that required
for anterior sagittal
Prone/Lateral
Neck flexion so that tumor is at
highest point of operating field
(nose pointing down)
Fourth ventricular tumors,
midline cerebellar, pineal
lesions, posterior third
ventricular tumors
Prone/Concorde
Neck flexion not exceeding
two fingerbreadths head lifted
upwards
Sitting (supracerebellar
infratentorial approach)
Neck flexion back elevated
elevation of thighs flexion of
knees
Lateral suboccipital
Cerebellopontine angle
tumors lateral cerebellar
tumors
Concorde/prone/lateral
park bench sitting
Varying degrees of neck flexion
Transsphenoidal
approach
Pituitary, suprasellar lesions
Supine with head on
Head rotated with flexion until
horseshoe headrest, body the bridge of the nose is 45° from
close to right side
horizontal axis
Anterior parasagittal
Subfrontal
Posterior parasagittal
Midline suboccipital
DACA, distal artery aneurysm; PICA, posterior inferior cerebellar artery.
V. POSITIONS IN NEUROSURGERY
Park bench position
Lateral transcondylar
approach for lesions of the
anterior foramen magnum,
aneurysm of PICA
POSITIONING FOR CRANIOTOMY
187
POSITIONING FOR CRANIOTOMY
Positioning of the Head
Ideal position of the head is one which provides optimal surgical approach to the target area with minimal trespass of the normal brain.
It is based on two principles.
t The imaginary trajectory from the highest point of the skull surface to the area of interest in the brain should be
the shortest distance.
t The exposed surface of the skull and an imaginary perimeter of the skull should be parallel to the floor.7
Alignment of the Head and Neck
Rotation of the Head
While extreme care is taken that the surgical target is reached, it is important to remember that the neck is in line
with the head and the body. The head can be safely turned between zero and 45° laterally to the right and left from
the body’s sagittal access. If more than 45 degrees of head rotation is required, the ipsilateral shoulder is raised on a
pillow or roll to maintain the axis.1 Though trivial complications such as cervical strain may frequently occur with
extreme rotation of the head, occasionally it may have extremely deleterious effects on the vascular structures of
the neck. It decreases the blood flow in the ipsilateral vertebral arteries as they traverse the narrow foramina in the
transverse process along the cervical spine. It also impairs venous return from the internal jugular veins leading to
increased ICP, brain swelling, thereby increasing the bleeding. Although there is conflicting evidence regarding the
side that develops compromised blood flow, when the head is placed in rotation, it is evident from a recent metaanalysis that the flow is more often reduced on the contralateral side of rotation and more in the intracranial part
as compared to the cervical part.8 Mechanical compression of the extracranial vertebral artery during neck rotation
has also been described.9 Patients with associated risk factors such as cervical spondylosis, vertigo, atherosclerosis,
osteoarthritis, elderly, etc. are more likely to have compromised vertebral blood flow with lateral rotation of the neck.
Preoperatively, identifying the patients with signs of possible vertebrobasilar insufficiency (VBI) such as vertigo will
better enable the neurosurgeons and anesthesiologists to optimally position the neck. In such individuals, evaluation
of VBI with a transcranial Doppler USG will be beneficial.10
Flexion and Extension of the Head
Hyperflexion of the head also leads to a decrease in blood flow in both the vertebral and carotid arteries which
may possibly lead to brain stem and cervical spine ischemia. It also reduces the anteroposterior size of the hypopharynx causing ischemia of the base of tongue leading to pharyngeal and tongue edema. This phenomenon may be
accentuated by the placement of the other devices in the oropharynx such as the transesophageal echocardiography
(TEE) probe, oral airway. Hence it is recommended that at least the neck flexion should not be less than two to three
fingerbreadths of thyromental distance.11 Extension of the head may cause dislodgement of ETT.
External pressure on the neck due to tight tapes for endotracheal tube fixation, securement of oral airway, neck
collar also impair venous drainage resulting in poor surgical conditions. Any obvious neck vein distension is a sign
of inadequate head and neck positioning which will contribute to raised ICP.
Elevation of the Head
The head is often positioned above the heart to facilitate venous return. In supine patients, 10° reverse Trendelenberg
produces a significant decrease in ICP while the cerebral perfusion pressure (CPP) is unchanged.12 This occurs 1 min
after the position change and remains stable. This is true in the prone position as well.13 As per Munroe–Kelly doctrine, the decrease in ICP due to the reverse Trendelenberg position is due to the displacement of the cerebrospinal
fluid (CSF) into the spinal segment.14
In patients positioned prone, increases in the intra-abdominal pressure often contribute to the rise in ICP and
hence elevating the head end is vital to reduce the ICP.
Monitoring During Head and Neck Positioning
Although in practice no additional routine monitoring is utilized during head and neck positioning, in vulnerable patients, ICP monitoring may be helpful aiming at a target of 20 mmHg. Jugular bulb pressure and jugular
venous saturation are surrogates to ICP monitoring. It can be easily monitored using a retrograde jugular catheter. Simultaneous measurement of the central venous pressure (CVP) and jugular venous pressure (JVP) with the
V. POSITIONS IN NEUROSURGERY
188
10. POSITIONING IN NEUROSURGERY
appropriate reference points (CVP at right atrium and JVP at the level of tragus) should show no change in pressures
as compared to the baseline with optimal positioning. Partial obstruction of venous outflow should be considered
should there be an increase in JVP, and prompt repositioning of the head should be undertaken.15
Fixation of the Head for Craniotomy
Fixation of the head after deciding on the final position of the head is a crucial step in positioning the neurosurgical procedure. The head may be positioned on a variety of fixation devices depending upon the surgery. The common devices used are MFK head clamp, Sugita head frame assembly, horseshoe headrest.
Mayfield Kees Skull Fixation Device (Ohio Medical Instrument Co., Cincinnati, OH)
The Mayfield head holder (Fig. 10.1A) consists of a clamp with three sterile pins. It is inserted into the skull in a
band-like area just above the orbits and the pinna. Special care is taken to avoid the frontal sinus and the temporal
bone. It should be positioned such that it does not interfere with the cranial incision but facilitates the attachment
of the halo self-retaining retractor. When the clamps are squeezed together, the gears slide until the pins are seated
in the skull. The knob housing the tension spring and gauze is tightened. Each ring exerts a pressure of 20 lb. In the
adults, up to 80 lb of pressure is allowable whereas in the pediatric population, 30–40 lb is the preferable limit. It is
then clamped on to the head frame assembly which is attached to the table (Fig. 10.1B). The manufacturer recommends the use of specially designed pediatric pins for children less than 10 years of age but it is preferable to avoid
using MFK in children less than 3 years.6
Problems associated with the use of Mayfield Kees
t
t
t
t
t
t
t
t
t
t
Pressure necrosis
Perforation/Fracture of skull
Injury to middle meningeal artery leading to hematoma or arteriovenous fistula
Extradural hematoma remote from pin site
Scalp and eye laceration due to slippage of the head holder
Bleeding from the pin site
Air embolism
Malposition, poor fixation
Pin site infection
Cervical spine injuries due to inadvertent patient movement
Sugita Multipurpose Head Frame
Sugita multipurpose head frame devised by Dr. Sugita uses four head pins to position the patient’s head (Fig.
10.1C). It is ideal for procedures where maximum support is necessary. It allows for a full 360° rotation and adjustment of the angle of patient’s head during surgery if required. It consists of a robust head frame assembly and a head
holder.
The head holder consists of four pins. The head frame assembly consists of a basal frame which is mounted on to
the head holder (Fig. 10.1D). The self-retaining retractors are attached to the base frames using thumb screws which
are spaced at 35° angles. At each end of the basal frame, there are two holes to support the hand rest and instrument
receptacles. The angle and height of the hand rest are all adjustable to suit individual requirements. The semicircular
bar mounted on the basal frame is used to attach the scalp hooks or the self-retaining retractors. There is an additional provision of a quarter frame such that it provides enough space to facilitate lateral suboccipital approach.
FIGURE 10.1 (A) Mayfield Kees three-pin head holder, (B) Mayfield Kees with head frame assembly, (C) Sugita four-pin head holder,
(D) Sugita pins with head frame assembly.
V. POSITIONS IN NEUROSURGERY
189
POSITIONS USED FOR CRANIOTOMIES
Horse Shoe Head Rest
It is a horseshoe-shaped headrest with both vertical and lateral adjustments which provide flexibility in patient
positioning both in supine and prone position. It also has gel pads which make it comfortable for use. The pulley rod
attachment for skeletal traction makes it an attractive option for cervical spine procedures. Precaution must be taken
so as to avoid pressure on the face in the prone positions which may lead to necrosis of the forehead, injury to the eye.
Scalp alopecia is a known complication in patients positioned supine on horseshoe headrest for prolonged duration.1
POSITIONS USED FOR CRANIOTOMIES
Supine Position or Dorsal Decubitus Position
It is the most commonly used position in neurosurgery. It does not require any special equipment for positioning
and is often easily achievable because the patient is able to move into the bed by themselves thereby allowing most
of the positioning to be completed before the induction of anesthesia. It is safe since it does not require disconnection
of the ETT and other monitoring devices.
In this position, the stroke volume (SV), cardiac output (CO), and venous return are optimal, and there is minimal
decrease of the mean arterial blood pressure. The functional residual capacity (FRC) and total lung capacity (TLC) are
decreased due to atelectasis of the dependent lung zones causing ventilation–perfusion (V/Q) mismatch. Manikandan
et al.16 have shown that the PaCO2 levels, 30 minutes after induction of anesthesia and positioning in the supine position,
were elevated due to increased alveolar dead space. The CPP is maintained but the CSF drainage may be impaired. The
head should be positioned above the level of the heart to promote venous drainage and to reduce cerebral edema. The head
may be rotated up to 45° relative to the body but if more rotation is needed, a roll or pillow should be placed under the contralateral shoulder. This also causes mild displacement of the abdominal viscera downwards, improving the ventilation.
After the final positioning of the head either in the flexion or extension position, it is important to rule out endobronchial migration of the endotracheal tube. It is preferable to fix the ETT on the side opposite to the surgery to
avoid accidental disconnections. Certain procedures such as the transoral approach to the odontoid requires tube
placement at the center of lips. A bar across the table enables access to the tubes and allows for observation of the
face after draping. Direct pressure on the globe is avoided by the placement of an attachment placed over the patients
face to prevent the drapes falling over the eyes. It is important to avoid skin to metal contact. The head is positioned
above the heart to facilitate venous return. The arm should be positioned so as to minimize the pressure on the ulnar
groove. Neutral forearm position should be maintained when the arms are tucked inside. There must be adequate
padding at the ulnar nerve to avoid ulnar neuropathy. Brachial plexus injury can occur if the arms are abducted
more than 90°, where the head of the humerus acts as a fulcrum around which the nerves of the brachial plexus are
stretched.17 Prolonged pressure on the peroneal nerve at the fibular head should be minimized by protective padding. Bony contact points at elbow, knee, occiput, sacrum, and heel must be padded.18 The patient is firmly secured
to the operating table with safety straps, which are across the patient’s thigh so that the vessels are not occluded. The
classical supine position leads to the loss of lateral lumbar lordosis and may cause postoperative back pain.
The supine positioning is often slightly modified either into a contoured position (lawn chair position) or reverse
Trendelenberg position. The lawn chair or contoured position (Fig. 10.2) is physiologically more favorable for the
lumbosacral spine. A 15° angulation and flexion at the trunk, thigh, and knee is provided. The knee is kept flexed by
a pillow under it. This position is also associated with improvement of the venous return from the lower extremities
with optimal CSF and lymph drainage.
The head-up tilt or reverse Trendelenberg position involves 10–15° tilt from the horizontal axis, and it improves
the venous drainage from the heart.
15 °
15 °
FIGURE 10.2 Lawn chair position.
V. POSITIONS IN NEUROSURGERY
15 °
190
10. POSITIONING IN NEUROSURGERY
Post positioning, the nurse assesses and documents that the pressure points such as the occiput, scapula, olecranon, elbows, popliteal space, and calcaneum are free of pressure. It is also important that these are rechecked and
documented at the end of the procedure.
Lateral Position
In the lateral position, hemodynamic parameters are minimally changed with mild decrease in SV, CO, and increase in
systemic vascular resistance (SVR) and pulmonary vascular resistance (PVR). This leads to modest decreases in systolic
blood pressure and mean arterial pressure (MAP) as compared to supine position. With regard to ventilation, there is mild
increase in PaO2 as compared to the supine position with a normal value of PaCO2.16 Perfusion is best in the dependent
lung zone while the nondependent lung is better ventilated thereby causing a mild V/Q mismatch. However, since the
abdominal excursions are free as compared to the supine position, its impact on oxygenation is limited.
With extreme neck flexion, there is a possibility of decreased jugular venous flow (JVF), jugular venous resistance
(JVR), and ICP. A chest or axillary roll is placed under the thorax below the axilla to prevent axillary compression.
When the head is on the MFK, the dependent shoulder is brought beyond the cephalad edge of the operating theatre
table and the dependent arm is rested on a low padded arm rest which is inserted between the table and head fixation. In these cases, there is no need for an axillary roll. The nondependent arm is usually placed over the trunk on
a pillow. It may also be positioned on the airplane arm rest. To monitor the blood flow to the dependent arm, it is
prudent to place the saturation probe or arterial line on the dependent arm (Fig. 10.3A and B). The trunk is supported
on lateral bolsters and secured by tapes. When the head is not fixed on MFK, it is important to support the patient’s
head with a pillow to avoid angulation of the cervical spine. The lower limbs are positioned with pillow between
the legs and the dependent extremities are flexed to avoid pressure over the fibula head and the peroneal nerve.
Complications anticipated with lateral positioning are brachial plexus injury, vascular compromise to the dependent
upper extremity, ear and eye injuries, and injury to the suprascapular nerve of the dependent shoulder.
Park Bench Position
It is a modification of the lateral position, and it provides better access to the low lying cranial lesions and provides
the surgeon with access to the anterior brain stem and foramen magnum as well as cerebellopontine angle tumors.
In this position, the trunk is rotated 15° from the lateral position. The upper arm is positioned along the lateral trunk
and the upper shoulder is taped toward the table. The dependent shoulder and arm are positioned outside the OR
bed and the arm is supported by a sling.3 The lower extremity is slightly flexed and a pillow is placed between the
knees. It is important not to tape the shoulder tightly or to drop the neck too much (Fig. 10.4).
FIGURE 10.3 (A) Left lateral position, hand on padded arm rest, (B) lateral position, view from behind.
FIGURE 10.4 Park bench position.
V. POSITIONS IN NEUROSURGERY
POSITIONS USED FOR CRANIOTOMIES
191
Semilateral Position (Janetta Position)
This position has been named after the neurosurgeon who popularized its use for microvascular decompression
of the fifth cranial nerve. It is achieved by semilateral tilting of the table combined with a generous shoulder roll.
Extreme head rotation which may cause compression of the contralateral jugular by the chin should be avoided.
Head is rotated slightly away from the affected side and flexed to approximately two fingerbreadths from the sternum. The vertex of the head is kept parallel to the floor.19
Prone Position
In the prone position, there are significant cardiovascular and respiratory changes. Venous return and SV are
significantly reduced due to increased intra-abdominal pressure and pooling of blood at the extremities leading to a
decreased CO, decreased left ventricular ejection fraction, and cardiac index (CI). To compensate for the significantly
reduced CO, heart rate increases along with increases in SVR and PVR.
As compared to the supine position, the FRC and TLC increase and there is improved perfusion. Moreover, there is a
decrease in atelectasis due to opening up of the dorsal zone of the lungs, thus minimizing V/Q mismatch and improving oxygenation. Although there is decrease in chest wall compliance due to increase in intra-abdominal pressure, this
is usually overcome by positive pressure ventilation. On the whole, the oxygenation improves with prone positioning.16
The cerebral hemodynamics varies with the position of the head. When the head is maintained neutral, the JVF is
increased and JVR is decreased. However, if the head is lower than the heart, there is venous congestion leading to
increase in ICP.15
For positioning prone, the patient is usually anesthetized on the trolley and then turned prone on to the operating
table which is prepared with chest rolls placed longitudinally/transversely. The head is fixed on to the head fixation
device such as MFK before turning prone. There are multiple potential problems with the turning of the patient such
as disconnection/dislodgement of monitors, IV accesses, ETT, etc. All these may contribute to either delayed recognition of anesthetic incidents such as hypotension, hypoventilation, desaturation, arrhythmias, or cardiac arrest.
Extreme vigilance is required during turning the patient, and it is prudent to keep the pulse oximeter and invasive
arterial line/ECG monitoring for early recognition of these events. All catheters [urinary, central venous, chest drain,
external ventricular drainage (EVD)] should be firmly secured to the body before turning.
Once the patient is positioned prone, the patient’s chin must be free from the table/frame. Extreme flexion, rotation, or extension of the neck may impair venous return thereby increasing ICP. Pressure points such as knees, groins
should be padded. The breasts should be medially displaced and pressure on the nipples should be avoided. The
lower extremity peripheral pulses should be checked to rule out vascular compromise of the femoral vessels. The
upper extremities are positioned by the patient’s side with the hands facing the patient. The eyes, nose, ears should
be protected and eyelids should be closed. It is important to secure the endotracheal tube firmly to avoid accidental
extubation in the prone position. Antisialogogues may be used to decrease secretion which may loosen the fixation
of endotracheal tubes. A soft gauze roll is placed as a bite block to prevent inadvertent tongue bite during monitoring
of motor-evoked potentials and to securely hold the endotracheal tube in place.
Reinforced ETT may also be considered to avoid kinking. The anesthesiologist must be prepared for sudden loss in the
airway in the prone position. Reports of laryngeal mask airway (LMA) being secured in the prone position as an emergency
alternative to endotracheal intubation have been described.20 Since prone positioning has been associated with significant
hemodynamic changes, it is prudent to preload the patient adequately before turning and also be prepared to handle sudden hemodynamic instability in the prone position. Patients who are susceptible to significant hemodynamic compromise
such as those with uncontrolled diabetes or hypertension may require vasopressor treatment while proning.21 Backup
plans should be made for sudden cardiac arrest in the prone position in patients with significant comorbidities, and the
need for the placement of transcutaneous pads in patients with significant cardiovascular risk should be considered before
proning. It is also important that the trolley is readily accessible in case of a need to turn supine (Fig. 10.5).
Concorde Position
It is preferred for the lateral suboccipital approach for the posterior fossa tumors, especially for the occipito transtentorial and supracerebellar infratentoreal approach. The patient is positioned as for a midline suboccipital position. The
head is secured in MFK before turning the patient prone. It is then flexed with the thoracolumbar region extended such
that the head is elevated above the heart. It is slightly modified to make the craniotomy or craniectomy site more prominent by rotating the patient’s head approximately 45° to the shoulder ipsilateral to the lesion. Reverse Trendelenberg
positioning is done to improve the venous return (Fig. 10.6). There is a lower incidence of air embolism22 and less
fatigue for the surgeon. Moreover, complicated instrumentation is not needed. The complications are the same as that
V. POSITIONS IN NEUROSURGERY
192
10. POSITIONING IN NEUROSURGERY
FIGURE 10.5 Prone position, head on Mayfield Kees.
FIGURE 10.6 Concorde position.
of prone position.23 In the modified concorde position, the patient’s arm at the surgeon’s side hangs down over the
head end of the operating table, with elbow flexion supported by an arm holder. This arm-down Concorde position
provides good access for the surgeon in muscular- or broad-shouldered, short-necked, or obese patients.24
Three-Quarters Prone (Lateral Oblique or Semiprone)
The three-quarter prone positioning is very similar to the lateral position and is used for approaching both the
posterior fossa and the parietooccipital region. The head is placed on the MFK and the nondependent arm is placed
behind the body in a coma or sleeping position. However, if a suboccipital approach is used, the hand may be taped
down toward the foot. The upper shoulder may be pulled away from the head and neck area toward the foot of the bed
with a tape. The patient’s trunk is supported by straps, tapes, or braces. The dependent lower extremity is extended
with the knee padded to prevent pressure on the peroneal nerve, and the upper lower extremity is flexed at the hip
and knee. The advantages are that it is comfortable for the surgeon and decreased risk of venous air embolism (VAE).
Transoral Approach
Most direct access to pathologies is located on the ventral side of the craniovertebral junction such as foramen
magnum, atlas, axis, atlantoaxial instability.25
The transoral approach is achieved by placing the patient in the supine position on the horseshoe headrest or MFK
with the head in extension. Oral intubation with an armored ETT is preferred. The arms are placed by the side. The
Crockard retractor is placed over the tongue and the counterfoil is placed against the incisor teeth.1
Approach for Transnasal Transsphenoidal Surgery
The patient is usually positioned supine with the head on a horseshoe headrest and hands by the side. The head
is turned to the patient’s right with the bridge of the nose parallel to the floor. The head is rotated in such a way that
the nostrils and nasal cavities are easily accessible for endoscopy. Reverse Trendelenberg position is performed to
decrease venous congestion. Incorrect positioning of patients for the transsphenoidal approach makes the surgery
V. POSITIONS IN NEUROSURGERY
POSITIONS USED FOR CRANIOTOMIES
193
difficult and dangerous, since a small alteration in the planned trajectory may result in disastrous consequences such
as injury to the carotid or the brain stem.26
Sitting Position
Sitting position offers optimum access to the midline lesions. It also improves cerebral venous decompression
lowering ICP, decreases the need for cerebellar retraction, promotes gravity-assisted drainage of blood and CSF
thereby enabling a clean surgical field and visualization of bleeding points and unobstructed view of the patient’s
face permitting observation of motor responses to cranial nerve stimulation.
Although sitting position offers distinct advantages to the surgeon, the extent of removal of tumor, neurological
outcomes, and facial nerve preservation have not been shown to be very different as compared to the lateral position
in a recent study.27 Instrumented cervical surgeries have also been done in the sitting position.28
In the sitting position, there is significant venous pooling of the lower extremities due to the effect of gravity leading to decrease in CO and hence arterial hypotension. Heart rate and SVR are increased. SV and CI are decreased.
PCWP is decreased, and SVR is increased.
Increase in FRC and TLC have been observed. However, since perfusion is limited, no major benefit in oxygenation
is observed. Ventilation is unimpeded as the diaphragmatic movement is improved, lowering the airway pressures.
Since arterial blood pressure reduces by 0.77 mmHg for each centimeter gradient above the heart, decrease in CPP
occurs after positioning the patient in sitting position leading to the possible development of cerebral ischemia. The
ICP is also reduced in the sitting position. A modified semisitting (lounging) position aiming to create a positive pressure in the transverse and sigmoid sinuses, with lower head and higher legs positioned above the top of the head,
decreases the incidence and severity of VAE.29
The patient is initially positioned supine on the operating table and anesthetized. The head is placed in head fixation. The
patient is placed in the sitting position by slowly elevating the back of the operating table incrementally over minutes while
maintaining hemodynamic stability. It is important to avoid hyperventilation as it may compromise cerebral blood flow.30
As the patient is made to sit, the operating table is flexed, elevating the thighs and the foot of the operating table is
dropped, flexing the knees to prevent stretch on the sciatic nerve. A pillow may be placed under the knees. The operating table is tilted backward as the table is flexed. The patient’s neck is kept in neutral or flexed. The arms should be
secured across the body or on arm rest to prevent drooping of the shoulder downward compressing on the brachial
plexus. The feet should not be allowed to hang off the table and the ankles should be rested to prevent injury to the
Achilles tendon.1 The frame of the head holder is properly clamped to the side rails of the back section so that it is
possible to lower the head end in the event of a significant air embolism. If improperly attached to the thigh section,
it will require that the head be disengaged from the head holder when the head needs to be lowered31 (Fig. 10.7).
FIGURE 10.7 Schematic representation of sitting position.
V. POSITIONS IN NEUROSURGERY
194
10. POSITIONING IN NEUROSURGERY
To avoid injury during neck flexion, it is preferable to monitor somatosensory evoked potential (SSEP) while positioning. It is recommended that the patient be checked preoperatively to determine the degree of neck flexion possible.
The hemodynamic changes during positioning may be influenced by patient’s factors such as intravascular volume
status, preexisting hypotension, BMI. Positioning the patient with flexion of hips and elevating the knees to the level of
the heart help to minimize hypotension. Slow-staged positioning after adequate hydration over a period of 5–10 min
with intermittent boluses of vasopressors as required will prevent any abrupt fall in blood pressures. Wrapping up of
the lower extremities, placing the patient in G suit also promotes venous return from the lower extremities.
Complications Associated With Sitting Position
Venous Air Embolism
Incidence ranges from 25% to 50% in a precordial Doppler study32 and 76% in a TOE study.33 The negative venous
pressure and the exposure of bony venous sinuses to air are responsible for the VAE. Various monitoring modalities have
been described with varying sensitivities such as capnography, fractional excretion of nitrogen, transthoracic precordial
Doppler, right heart catheter, TOE, and transcutaneous oxygen measurement. VAE reduces CO by causing an air lock
and subsequent mechanical overdistention of the right ventricle and obstruction to the pulmonary circulation. Precordial
Doppler is the best noninvasive device to monitor VAE and is placed from the third to sixth intercostal space, right sternal
border where the highest pitch over the right upper sternal border with intravenous injection of agitated saline.
Carbon dioxide field flooding has been used to reduce the hemodynamic effects of VAE.34 Intrajugular balloon
catheter is also used to prevent VAE.35 Turgut et al. have shown that the use of paramedian approach leads to
increased incidence of VAE as compared to the median approach.27 Usefulness of preoperative contrast ECHO as a
screening technique to detect patent foramen ovale (PFO) has been described.36
Paradoxical Air Embolism
The gradient between the atria is an important factor in the pathophysiology of VAE. Any condition which increases
right atrial pressure relative to left atrial pressure may predispose to paradoxical air embolism (PAE). Application and
release of positive end-expiratory pressure increases right atrial pressures thereby promoting right to left shunting and
PAE. Patients at risk of PAE such as those with right to left shunts, PFO must be preoperatively identified.
Pneumocephalus
The incidence of pneumocephalus is almost 100%. Diminution of the brain volume secondary to mannitol administration, hyperventilation, intraoperative drainage of CSF predispose to pneumocephalus. Avoiding intracranial dehydration,
hyperventilation during dural closure to facilitate the expansion of brain helps to decrease the incidence of pneumocephalus. High index of suspicion is required to diagnose potentially life-threatening complication, tension pneumocephalus.
Confusion, headache, convulsions, neurological deficits in the immediate postoperative period should be evaluated with
a prompt CT scan. Rapid evacuation of the tension pneumocephalus results in prompt recovery.
Quadriplegia
Extreme flexion of the neck stretches the cord at C5 leading to decreased regional cord perfusion, especially during
periods of hypotension. SSEP may be used to monitor the adequacy of regional cord perfusion.
Nerve Injuries
Common peroneal neuropathy often occurs secondary to nerve compression as it courses around the neck of the
fibula. Hyperflexion of thigh may lead to stretching of the peroneal division off the sciatic nerve. Recurrent laryngeal
nerve injury also may be noticed due to extreme flexion.
Other Complications
Macroglossia may occur leading to airway obstruction. This is more pronounced in infants where there is an anterior
larynx, small tracheal diameter, and large tongue. Injury to the soft palate and posterior pharyngeal wall and tongue
has also been described. Sustained neck flexion, prolonged duration, and use of oral airway have been implicated in the
pathogenesis of macroglossia. Bite blocks and smaller diameter TEE probes will minimize the incidence of macroglossia.
Contraindications to Sitting Position
Absolute
t
t
t
t
PFO
Cerebral ischemia when upright and awake
Patent ventriculoatrial shunt
Right atrial pressure more than left atrial pressure
V. POSITIONS IN NEUROSURGERY
195
PATIENT POSITIONING FOR SPINAL PROCEDURES
Relative
t
t
t
t
Extremes of age
Uncontrolled hypertension
Chronic obstructive pulmonary disease
Degenerative disease of the spine
Careful patient selection, intraoperative techniques, prompt diagnosis and treatment decrease the incidence of
VAE and the risks associated with sitting position.
SURGICAL APPROACH FOR PROCEDURES OF THE SPINE
PATIENT POSITIONING FOR SPINAL PROCEDURES
There are wide varieties of spinal procedures ranging from simple laminectomies to major instrumentation, performed in various positions. Positioning appropriately is an important component of successful postoperative outcome
in spinal procedures. The surgeon decides on the optimal surgical approach and the position required. Since procedures
of the spine are associated with extensive use of fluoroscopy, radiography, and navigation systems intraoperatively, these
should be taken into consideration while positioning. Surgical objectives during any spine procedure are to facilitate
exposure, minimize bleeding, prevent injury to vital structures, and have a good functional outcome postoperatively.
The anesthesiologist aims at providing optimal ventilation and oxygenation together with hemodynamic stability. In
some situations, the associated comorbidities in a patient demand modifications to the commonly used approach.
Table 10.3 describes the various approaches for procedures of the spine.
Equipment Required for Prone Positioning
t Basic operating table with appropriate prone positioning frames
t Jackson’s spinal table (Misuho OSI, Union City, CA, USA)
Although a basic operating table may be used for most procedures, the modular spine-specific operating tables
such as Jackson’s are adjustable offering greater vertical range providing ergonomic advantage to surgeons. The
radiolucent carbon fiber frame allows for the use of fluoroscopy, intraoperative CT scanning, and navigation systems which precisely locate the levels and to course the trajectory of pedicle screws. The ability to rotate 360° allows
intraoperative repositioning for combined anterior and posterior procedures. The accessories allow customization
to suit individual patient and surgical needs. The head may be supported either with foam headrest, rigid fixator or
traction, and the legs may be supported in a sling or on a rigid frame.37
Frames Used for Positioning the Body in Prone
Various techniques and frames have been designed to position the patient in the prone position so as to have optimal ventilation and oxygenation while facilitating exposure and minimizing the blood loss.
TABLE 10.3 Approaches for Surgery of the Spine
Approach
Procedures
Body Position
Immobilization of Head
Anterior
ACDF
Cervical corpectomies
Anterior lumbar fusion
Supine
Tapes
Gardner-Wells tongs
Lateral
Thoracic corpectomies
Thoracolumbar lesions
Anterior lumbar lesions
Lateral Jackson’s table
Gel headrest
Posterior
Posterior spine fusions
Lumbar and thoracic discectomies, laminectomies
Scoliosis correction, CVJ anomalies
Prone on chest rolls, Wilson’s frame,
Relton and Hall frame, Jackson’s
table
Prone headrest
MFK for cervical laminectomies
Transoral
Atlanto occipital dislocation
Supine
Head on horseshoe headrest with
hyperextension
ACDF, anterior cervical discectomy and fusion; CVJ, craniovertebral junction; MFK, Mayfield Kees.
V. POSITIONS IN NEUROSURGERY
196
10. POSITIONING IN NEUROSURGERY
Chest rolls are the most effective and inexpensive techniques used for prone surgery. They may be placed either
transversely or longitudinally.
Relton and Hall Four Poster Frame (1969, Imperial Surgicals, Quebec, Canada)
It consists of four padded supports in two “V” shaped pads, which support the lateral parts of the upper chest
rostrally and the lateral aspect of the hip caudally in such a manner to allow free abdominal excursion. The supports are angled 45° inwardly and are adjustable (Fig. 10.8). The hyperextension of the vertebral column is limited
by lowering the legs. As compared to the use of chest rolls, the use of four poster frame is associated with decreased
intra-abdominal pressure, inferior vena cava (IVC) pressure, and preservation of lumbar lordosis.38
Andrew’s Hinder–Binder Frame (OSI, Union City, CA, USA)
It has an adjustable tibial support to allow for a varying range of hip flexion and it allows for the use of C-arm.
Patients are positioned in a modified knee chest position on the Andrew’s table. It allows C-arm integration for both
A/P and lateral intraoperative views.38
Wilson Supporting Frame (OSI, Union City, CA, USA)
It consists of two curved full length pads to provide support to the chest and pelvis and adjust laterally to improve
ventilation and to relieve pressure from the abdomen. Blood loss and intra-abdominal pressure were lower in patients
positioned on wider Wilson frame support.39 More recently, a newer version of this frame, Wilson Plus offers 360° of
unobstructed radiolucency, for easily obtainable images by either C-arm or X-ray is available38 (Fig. 10.9).
FIGURE 10.8 Relton hall frame.
FIGURE 10.9 Wilson’s frame.
V. POSITIONS IN NEUROSURGERY
PATIENT POSITIONING FOR SPINAL PROCEDURES
197
Equipment for Stabilizing the Head
It is important to maintain neutral position of the cervical spine to avoid postoperative cervical myeloradiculopathy.
t
t
t
t
t
Dough nut–shaped foam/gel pads
Prone pillows
Gardner-Wells tongs
MFK
Horseshoe headrest
Dough Nut–Shaped Foam/Gel Pads
This may be utilized to provide head support and to maintain neck in neutral position for patients undergoing
procedures in the supine and lateral procedures. Appropriate sizes must be chosen to avoid neck flexion, extension,
or hyperextension.
Prone Pillows
Different prone head positioners are available to position the head for spine procedures in the prone position
(Fig. 10.10). They are usually made of polyurethane foam with preformed slits designed such that the eyes and nose
are free of pressure, and there is a provision for the exit of the endotracheal tube. Recently a dry flotation device with
multiple adjustable rubber-filled bladders is being used (ROHO group, Belle Vue, IL).40 The prone view helmet system (Dupaco, Oceanside, CA) was developed to address visual loss caused by direct pressure. It consists of a rigid
helmet to support the head, soft foam inserts to disperse pressure while avoiding pressure on eyes, nose, mouth,
and a mirror to reflect the patient’s eyes.41
Geordie et al. have described the use of a prone view helmet system with a modified table platform for open
access to the eyes during prone position allowing them to measure intraocular pressure, ocular perfusion pressure, and ocular perfusion (using digital retinal or optic disc imaging).40 Mc Michael, in a study comparing
polyurethane foam, prone view, and neoprene rubber pillows, has shown that the prone view positioner was
considered the most comfortable for patients.42 The prone view positioner and the ROHO neoprene pillow demonstrated significantly lower face pillow interface pressures as compared to the commonly used polyurethane
foam.
Gardner-Wells Tongs, Traction Systems
This provides two ropes for the traction. The alignment of the spine can be altered during surgery because of the
availability of dual vectors for traction. Placement of the upper thoracic pedicle screw causes movement which may
result in dislodgement of head from even a rigid skull fixation device. In such cases, it is beneficial to use a properly
placed traction system.37
FIGURE 10.10 Prone pillow.
V. POSITIONS IN NEUROSURGERY
198
10. POSITIONING IN NEUROSURGERY
Pulmonary Compliance in the Prone Position
By compressing the abdomen and limiting the chest wall movement, there is a significant compromise of the pulmonary compliance in the prone position.
In a study comparing the three commonly used prone positioning devices, longitudinally placed chest rolls,
Jackson spinal table, and Wilson’s frame, Sally et al. have shown that patients undergoing procedures in the prone
position have an increase in peak airway pressure and decrease in lung compliance when moved from the supine
to prone position in both the group with chest rolls and the Wilson’s frame, but not while using the Jackson’s table,
suggesting that the pulmonary mechanisms are frame dependent.43
Free movement of the abdomen improves the excursion of the diaphragm, thereby improving the oxygenation
and ventilation. It also decreases bleeding due to decrease in the intra-abdominal pressure and improves venous
return from the lower extremities.15
Position-Related Factors Affecting Blood Loss in Spinal Procedures in Prone Position
There are several plexuses of thin-walled valveless veins that contain blood at low pressure, and the direction of
flow of blood is reversible.44
The vertebral veins are connected to those in the chest through the vertebral canal and the ones in the abdomen
and pelvis through intercostal, lumbar, and other connecting veins.38 With the obstruction of IVC, blood from the
lower part of the body could be diverted into the vertebral venous system. Batson’s plexus consists of the following:
1. Internal venous system, which is continuous from the base of skull to the sacrococcygeal region. It is formed
by the anterior internal venous veins (on the posterior surface of the vertebral body), posterior internal venous
veins (on the surface of the lamina), and the anastomotic veins between the two.
2. External venous system which consists of the longitudinal traversing veins anterior to the vertebral bodies on
the outer aspect of the lamina.
3. Anastomotic or connecting veins which are a group of veins connecting the internal to the external venous
system and the systemic and vertebral venous systems.
During prone positioning, multiple reasons are responsible for IVC obstruction, and increase in IVC pressure
results in diversion of blood into the vertebral veins. Pressure on the anterior abdominal wall due to sandbags, bolsters, or excessive abdominal muscle tension is transmitted to the IVC.45 Even moderate increase in abdominal pressure is known to cause rise in caval pressure. The altered respiratory compliance associated with prone positioning
can cause elevated airway pressures which impair venous return to the heart thereby decreasing CO and increasing
systemic venous pressure. This often leads to decreased perfusion pressure of the spinal cord as well (SEPP = MAP—
spinal venous pressure) leading to potential for increased neurological complications. Hypercarbia with increase in
airway pressure during expiration also causes an increase in venous pressure. All these factors contribute to blood
loss in the prone position. Optimal positioning is hence essential to minimize the blood loss in this position.
Alignment of the Spine for Procedures of the Spine
It is important to achieve proper spinal alignment during positioning for the best clinical outcome. When arthrodesis is not performed, the aim in intraoperative positioning is to provide optimal neural decompression without
excessive bleeding. But if arthrodesis is planned, positioning must include the placement of spine in anatomic alignment to avoid iatrogenic deformity. Improper alignment of the occipitocervical region can have various complications. In the extremely extended position, fusion of the spine leads to the inability of the patient to see his own body,
whereas in the excessive flexion, there is difficulty with swallowing. Rotational malalignment will lead to head tilt.
In lumbar arthrodesis, maintaining adequate lumbar lordosis is crucial to prevent flat back syndrome, and hence it
is preferable to avoid Wilson’s frame for fusion procedures of the lumbar spine.37
Positioning of the Head for Procedures of the Spine
In cases of unstable cervical spine, the head needs to be stabilized with a rigid immobilization device such as
Philadelphia collar or cervical traction. Careful and meticulous transfer from the supine to the prone position
should be done while maintaining the alignment of the axis of the body and head. In cases of suspected myelopathy, motor evoked potential monitoring should be done before and after positioning to confirm integrity of the
V. POSITIONS IN NEUROSURGERY
PATIENT POSITIONING FOR SPINAL PROCEDURES
199
neural pathways.46 In the absence of such monitoring, awake proning may also be done.47 Neural integrity may be
confirmed by the patient and then anesthetized for the procedure. Recurrent laryngeal nerve palsy has been associated with procedures involving the anterior approach to the lower cervical spines specially from the right side.48
Endotracheal tube–mediated compression of the recurrent laryngeal nerve due to retraction for anterior cervical
spine surgery is a significant cause of vocal cord paralysis. Monitoring the endotracheal cuff pressure frequently
and keeping it within the normal range during the procedure is recommended. Deflating the endotracheal tube cuff
and reinflating just to seal the pressure after the placement of retractors helps to reduce the incidence of temporary
recurrent laryngeal nerve palsies.49
In procedures of the lumbar, lower thoracic, and sacral spine, the use of head fixation devices is not required, and
head is positioned on a gel or foam headrest. The upper extremities are usually positioned either upwards—superman position for procedures on the lumbar spine or by the side for thoracic cases. Care must be taken that the brachial plexus is not stretched.
Hemodynamic Monitoring in the Prone Position
Since there is a decrease in venous return leading to decreased CO, it is essential to maintain euvolemic status.
CVP monitoring may be used to monitor intravascular volume. Since the CVP values are not accurate, the trends
should be monitored rather than absolute values. Dynamic indices of preload assessment such as pulse pressure
variation, stroke volume variation, and systolic pressure variation are reliable in the prone positioning. Beat to beat
monitoring of the blood pressure with an arterial line enables the anesthesiologist to pick up unanticipated sudden
decreases in CO due to VAE, massive bleeding, etc. and allows for frequent blood sampling. High-risk patients
should be identified and optimized prior to surgery and adhesive defibrillator pads are placed. The least change
in hemodynamic status due to proning has been observed with Jackson’s table and it should be used in high-risk
cases. In case of sudden arrest, chest compressions may have to be initiated while in the prone position itself.56
Prolonged hypotension in the prone position may lead to ischemic hepatitis. Maintaining the neck in the neutral
nonextended position helps reducing pressure on the neurovascular bundle. Kinking, intimal injury, and thrombosis have been observed to cause carotid and vertebrobasilar dissections in patients with nonneutral neck position.
Any foreign body in the internal jugular vein such as cannulas may predispose to thrombosis and stroke. It is preferable to use subclavian catheters since they have been noted to be less thrombogenic.57 Increased age, malignancy,
obesity, and recent major surgery predispose to thrombosis and stroke in these patients.
Problems Associated With Prone Position
Brachial plexus stretch injury is known to occur in the prone position. Extreme degrees of extension or rotation of
the head to the contralateral side causes stretching of the nerve roots leading to brachial plexus injury.
Excessive pressure on the clavicle can compress the neurovascular bundle against the first rib. Head of humerus
can press on the brachial plexus when the arms are hyperabducted (abducted more than 90 degrees) and the shoulder is not sufficiently mobile and relaxed.
Increased Intra-Abdominal Pressure in the Prone Position
With improper positioning and in obese patients, the intra-abdominal pressure increases with prone positioning
which leads to abdominal compartment syndrome and increased bleeding in procedures of the spine. An increase in
intra-abdominal pressure of more than 12 mmHg from the supine position is a high risk for developing abdominal
compartment syndrome (ACS). Due to tight abdominal closure, patients with previous abdominal surgery are at
high risk for developing ACS. Prolonged elevation of intra-abdominal pressures may lead to decreased perfusion
pressure predisposing to multiorgan failure. Identifying patients who are at risk to develop high intra-abdominal
pressure and to monitoring intra-abdominal pressure in them should be considered. Any decrease in blood pressure,
urine output or persistence of abnormal base deficit, increase in PCO2 may point toward multiorgan failure. Prompt
abdominal decompression should be done if ACS is suspected.50
Nerve Palsies/Neuropraxia
In the prone position, there is an increased risk of injury to the cervical spine and the brachial plexus. Impaired
axoplasmic transmissions due to increased intraneural venous pressure, local edema are the pathophysiologic mechanisms leading to nerve palsies and neuropraxias. This may be due to stretch, compression, generalized ischemia, or
metabolic causes. Several risk factors have been identified such as male gender, extremely thin individuals, obese,
V. POSITIONS IN NEUROSURGERY
200
10. POSITIONING IN NEUROSURGERY
TABLE 10.4 Practice Advisory for the Prevention of Perioperative Peripheral Neuropathies
SUMMARY OF ADVISORY STATEMENTS
I. Preoperative history and physical assessment
When judged appropriate, it is helpful to ascertain that patients can comfortably tolerate the anticipated operative position.
II. Specific positioning strategies for the upper extremities
t A
rm abduction in supine patients should be limited to 90°.
t P
atients who are positioned prone may comfortably tolerate arm abduction greater than 90°.
Supine patient with arm on an arm board
The upper extremity should be positioned to decrease pressure on the postcondylar groove of the humerus (ulnar groove). Either supination
or the neutral forearm positions facilitate this action.
Supine patient with arms tucked at side
t T
he forearm should be in a neutral position.
t F
lexion of the elbow may increase the risk of ulnar neuropathy, but there is no consensus on an acceptable degree of flexion during the
perioperative period.
t P
rolonged pressure on the radial nerve in the spiral groove of the humerus should be avoided.
t E
xtension of the elbow beyond the range that is comfortable during the preoperative assessment may stretch the median nerve.
t P
eriodic perioperative assessments may ensure maintenance of the desired position.
III. Specific positioning strategies for the lower extremities
t S
tretching of the hamstring muscle group—Positions that stretch the hamstring muscle group beyond the range that is comfortable during
the preoperative assessment may stretch the sciatic nerve.
t L
imiting hip flexion—Because the sciatic nerve or its branches cross both the hip and the knee joints, extension and flexion of these joints,
respectively, should be considered when determining the degree of hip flexion.
t N
either extension nor flexion of the hip increases the risk of femoral neuropathy.
t P
rolonged pressure on the peroneal nerve at the fibular head should be avoided.
IV. Protective padding
t P
added arm boards—Padded arm boards may decrease the risk of upper extremity neuropathy.
t C
hest rolls—The use of chest rolls in the laterally positioned patient may decrease the risk of upper extremity neuropathy.
t P
adding at the elbow—Padding at the elbow may decrease the risk of upper extremity neuropathy.
t P
adding to protect the peroneal (fibular) nerve—The use of specific padding to prevent pressure of a hard surface against the peroneal nerve
at the fibular head may decrease the risk of peroneal neuropathy.
t C
omplications from the use of padding—The inappropriate use of padding (e.g., padding too tight) may increase the risk of perioperative
neuropathy.
V. Equipment
t T
he use of properly functioning automated blood pressure cuffs on the arm (i.e., placed above the antecubital fossa) does not change the risk
of upper extremity neuropathy.
t T
he use of shoulder braces in a steep head-down position may increase the risk of perioperative neuropathies.
VI. Postoperative assessment
t A
simple postoperative assessment of extremity nerve function may lead to early recognition of peripheral neuropathies.
An updated report by the American Society of Anesthesiologists Task Force on prevention of perioperative peripheral neuropathies.
hypotension, diabetes mellitus, hypothermia, anatomic variations, and malnutrition.51,52. Positioning the humerus
anterior to the trunk and placement of supportive padding for the arm is beneficial to prevent brachial plexus injury.
Palpation of the pectoralis major muscle tendon may be used to monitor tension on the brachial plexus.51,52 Anterior
shoulder dislocation is higher in the prone position and may lead to ischemia and compartment syndrome that may
cause rhabdomyolysis, raised myoglobin, and the development of limb compartment syndrome. Long-duration procedures, presence of peripheral vascular disease, obesity, increased muscularity predispose to compartment syndrome.
Table 10.4 describes the task force recommendations for prevention of neuropathy.53
Pressure Sores
In the prone position, extreme pressure is exerted on the chin, forehead, supraorbital, malar eminence, and ears.
On an average, pressure on the face is 30 mmHg which is above the capillary perfusion pressure. It may be even up
to 50 mmHg.54 Although supportive padded headrests do reduce the pressures, the risk increases with prolonged
duration of surgery, head-down position, and excessive use of crystalloids for replacement. The head end improves
the venous drainage and reduces the venous congestion. 15 pounds of upward traction at 45° angle produced by
Gardner-Wells tong alleviates the pressure on the face.55 Pressure on the chest ranges from mild erythema to skin
V. POSITIONS IN NEUROSURGERY
PATIENT POSITIONING FOR SPINAL PROCEDURES
201
FIGURE 10.11 (A) Gel pads for positioning, (B) gel sacral pads and axillary rolls, (C) knee pads and heel rests.
peeling. Patients with larger breasts are more susceptible to injury. Breast rupture and necrosis are possible in those
with implants.52 Hematoma, chondritis, ischemia, and necrosis have been reported in the ear. All pressure points
need to be padded with appropriate padding such as gel pads (Fig. 10.11A–C).
Edema of the Face
Oropharyngeal swelling, macroglossia, and sublingual hematoma have been observed in patients positioned
prone. Kinking and stretching of the salivary ducts, blood vessels, and lymphatics when the neck is flexed
predispose to salivary gland swellings.58 Temporomandibular joint dislocation during intubation, poor oral
hygiene, dehydration, malnutrition, repeated laryngoscopy, use of angiotensin converting enzyme inhibitors
are known risk factors for salivary gland swellings. Use of soft bite block to prevent tongue compression is
preferred over hard oral airways. It is important to monitor the head and neck positioning in the prone position
every hour. Cuff leak should be checked before extubation, and extubation should be postponed if there is no
cuff leak or if there is significant facial swelling. Patient should be monitored in the first few hours for postextubation stridor.59
Venous Air Embolism
Risk of air entrainment is higher if the venous pressure is low or if the surgical site is positioned above the heart.
Central venous catheter should be placed in those at higher risk for VAE.
Endotracheal Tube Displacement
Patients in prone position are at higher risk for endotracheal tube displacement due to the weight of the breathing
circuits. LMA may be used as a rescue airway disease in case of accidental extubation.20 To avoid displacement of the
LMA in the prone position, it is essential to maintain higher cuff pressures.60
Perioperative Vision Loss
The incidence of perioperative vision loss following procedures of the spine is 0.05–1%.61 Faulty positioning
accounts for only a small percentage of postoperative vision loss. Direct pressure on the eye causes trauma resulting
in conjunctival edema, hemorrhage, chemosis, and pain.
Ischemic Optic Neuropathy
Increased intraocular pressure and increased orbital venous pressure leads to decreased perfusion pressure on
the optic nerve head. Ischemic optic neuropathy (ION) may be either anterior to the lamina cribrosa or posterior.
Posterior ION is more common postoperatively and is more severe than the anterior.62 They are known to occur
even with sufficient support to facial padding. Risk factor for ION is longer operative time in the prone position in a
patient with anemia, hypotension, and blood transfusion.63.
Central Retinal Artery Occlusion (Headrest Syndrome)
This may be due to direct or indirect pressure and is characterized by periorbital and scleral edema and a
cherry red spot appearance on the fundus. The risk for central retinal artery occlusion (CRAO) is usually due
to improper positioning during the surgery.63 Pathogenesis may be due to vasospasm, emboli, compression, or
V. POSITIONS IN NEUROSURGERY
202
10. POSITIONING IN NEUROSURGERY
hypotension.61 Dislodgement of plaques from the carotid artery may cause CRAO. Direct pressure from the headrest may be higher in patients with exophthalmos or low nasal bridge.
Cortical Blindness
This is due to infarction of the posterior cerebral artery or significant hypotension. This may recover slowly but
not completely. Risk factors for cortical blindness include cardiac arrest, air embolism, significant hypotension, or
prolonged hypoxia. Prone positioning and obesity were found to be most commonly associated with development
of cortical blindness.63
Acute Angle Closure Glaucoma
The prone position shifts the lens iris diaphragm forward and obstructs the flow of aqueous humor. This causes
sudden increase in intraocular pressure. In high-risk patients, even short surgical procedures may precipitate acute
angle closure glaucoma.
Most POVL is ION which has been associated with duration of procedure more than 6 h, blood loss more than
1000 ml, DM, hypertension, smoking, and small cup to disc ratio. ION occurs irrespective of the frame and frequency
of eye checks. Short-staged procedures with careful positioning of the head (10–15° head up), avoiding swings of
blood pressure, maintaining MAP ≥ 70 mmHg, hematocrit > 25 mmHg are strategies which will reduce the incidence
of POVL. Continuous real-time monitoring of the eye has been described by Woodruff et al. as an alternative to frequent monitoring of eye position.
Table 10.5 summarizes the task force recommendation for prevention of perioperative vision loss.64
TABLE 10.5 Practice Advisory for Perioperative Visual Loss Associated With Spine Surgery
SUMMARY OF ADVISORY STATEMENTS
I. Preoperative patient evaluation and preparation
Although the consultants and specialty society members agree that there are identifiable preoperative risk factors, at this time the Practice
Advisory Task Force does not believe that there are identifiable preoperative patient characteristics that predispose patients to perioperative
ION.
t F
urther, the Task Force believes that there is no evidence that an ophthalmic or neuro-ophthalmic evaluation would be useful in identifying
patients at risk for perioperative visual loss.
t T
he Task Force believes that the risk of perioperative ION may be increased in patients who undergo prolonged procedures, have
substantial blood loss, or both.
t C
onsider informing patients in whom prolonged procedures, substantial blood loss, or both are anticipated that there is a small,
unpredictable risk of perioperative visual loss.
t B
ecause the frequency of visual loss after spine surgery of short duration is very low, the decision to inform patients who are not anticipated
to be “high risk” for visual loss should be determined on a case-by-case basis.
II. Intraoperative management blood pressure management
t S
ystemic blood pressure should be monitored continually in high-risk patients.
t T
he Task Force believes that the use of deliberate hypotensive techniques during spine surgery has not been shown to be associated with
the development of perioperative visual loss.
Therefore, the use of deliberate hypotension for these patients should be determined on a case-by-case basis.
Management of intraoperative fluids
t C
entral venous pressure monitoring should be considered in high-risk patients.
t C
olloids should be used along with crystalloids to maintain intravascular volume in patients who have substantial blood loss.
Management of anemia
t H
emoglobin or hematocrit values should be monitored periodically during surgery in high-risk patients who experience substantial blood
loss.
t T
he Task Force believes that there is no documented lower limit of hemoglobin concentration that has been associated with the
development of perioperative visual loss.
Therefore, the Task Force believes a transfusion threshold that would eliminate the risk of perioperative visual loss related to anemia cannot be
established at this time.
Use of vasopressors
t T
he Task Force consensus is that there is insufficient evidence to provide guidance for the use of adrenergic agonists in high-risk patients
during spine surgery.
Therefore, the decision to use adrenergic agonists should be made on a case-by-case basis.
V. POSITIONS IN NEUROSURGERY
ABBREVIATIONS
203
TABLE 10.5 Practice Advisory for Perioperative Visual Loss Associated With Spine Surgery—cont’d
Patient positioning
t T
he Task Force believes that there is no pathophysiologic mechanism by which facial edema can cause perioperative ION.
t T
here is no evidence that ocular compression causes isolated perioperative anterior ION or posterior ION.
However, direct pressure on the eye should be avoided to prevent CRAO.
t T
he high-risk patient should be positioned so that the head is level with or higher than the heart when possible.
t T
he high-risk patient’s head should be maintained in a neutral forward position (e.g., without significant neck flexion, extension, lateral
flexion, or rotation) when possible.
III. Staging of surgical procedures
t A
lthough the use of staged spine surgery procedures in high-risk patients may entail additional costs and patient risks (e.g., infection,
thromboembolism, or neurologic injury), it also may decrease these risks and the risk of perioperative visual loss in some patients.
Therefore, consideration should be given to the use of staged spine procedures in high-risk patients.
IV. Postoperative management
t T
he consensus of the Task Force is that a high-risk patient’s vision should be assessed when the patient becomes alert (e.g., in the recovery
room, intensive care unit, or nursing floor).
t I f there is concern regarding potential visual loss, an urgent ophthalmologic consultation should be obtained to determine its cause.
t A
dditional management may include optimizing hemoglobin or hematocrit values, hemodynamic status, and arterial oxygenation.
t T
o rule out intracranial causes of visual loss, consider magnetic resonance imaging.
t T
he Task Force believes that there is no role for antiplatelet agents, steroids, or intraocular pressure-lowering agents in the treatment of
perioperative ION.
An updated report by the American Society of Anesthesiologists Task Force on perioperative visual loss.
CRAO, central retinal artery occlusion; ION, ischemic optic neuropathy.
CONCLUSION
Surgical procedures of the brain and spine require complex positioning so that the surgical target is easily accessible to the surgeon. It is a delicate balance between the best surgical approach and the physiologically optimal
position for the patient. Within physiological limits, positioning should be modified so as to assist the surgical goals
such as reduction of bleeding, ICP, minimal brain retraction. Careful attention and meticulous protection should be
offered to vulnerable tissues especially during prolonged duration of surgery. Optimal positioning is the key to prevent morbidity and improve surgical outcomes following complex neurosurgical procedures.
ABBREVIATIONS
A-line Arterial line
CI Cardiac index
CO Cardiac output
CPP Cerebral perfusion pressure
CSF Cerebrospinal fluid
CVJ Craniovertebral junction
ECG Electrocardiogram
EDH Extradural hemorrhage
ETT Endotracheal tube
EVD External ventricular drainage
FRC Functional residual capacity
ICP Intracranial pressure
IV Intravenous
JVF Jugular venous flow
JVR Jugular venous resistance
LVEF Left ventricular ejection fraction
MFK Mayfield Kees
OT Operating theater
PAP Pulmonary arterial pressure
PVR Pulmonary vascular resistance
SCD Sequential compression device
SV Stroke volume
SVR Systemic vascular resistance
TED Thromboembolic deterrent
TLC Total lung capacity
V/Q Ventilation–perfusion
VTE Venous thromboembolism
V. POSITIONS IN NEUROSURGERY
204
10. POSITIONING IN NEUROSURGERY
References
1. Richard Winn H. General principles of operative positioning. In: YOUMANS neurological surgery. 5th ed. Philadelphia, PA: Saunders, Elsevier;
2004. p. 595–621.
2. Moore DC, Edmunds LH. Prone position frame. Surgery February 1950;27(2):276–9.
3. St-Arnaud D, Paquin M-J. Safe positioning for neurosurgical patients. AORN J June 2008;87(6):1156–68. quiz 1169–72.
4. Athiraman U, Gupta R, Singh G. Endotracheal cuff pressure changes with change in position in neurosurgical patients. Int J Crit Illn Inj Sci
2015;5(4):237–41.
5. David JW, Knight RP, Mahajan. Patient positioning in anaesthesia [Internet]. Available from: http://ceaccp.oxfordjournals.org/content/
4/5/160.abstract.
6. Tew Jr JM, Scodary DJ. Basic techniques and surgical positioning. In: Brain Surgery, Complication avoidance and management. New York: Churchill
Livingston Inc.; 1993. p. 31–51.
7. Clatterbuck R, Tamargo R. Surgical positioning and exposure for cranial procedures. In: YOUMANS neurological surgery. 5th ed. Philadelphia:
Saunsers, Elsevier Inc.; 2004. p. 595–691.
8. Mitchell J. Vertebral artery blood flow velocity changes associated with cervical spine rotation: a meta-analysis of the evidence with implications for professional practice. J Man Manip Ther 2009;17(1):46–57.
9. Sakaguchi M, Kitagawa K, Hougaku H, Hashimoto H, Nagai Y, Yamagami H, et al. Mechanical compression of the extracranial vertebral
artery during neck rotation. Neurology September 23, 2003;61(6):845–7.
10. Alnaami I, Siddiqui M, Saqqur M. The diagnosis of vertebrobasilar insufficiency using transcranial Doppler ultrasound. Case Rep Med
2012;2012:894913.
11. Brambrink AM, Kirsch JR. Positioning the patient for neurosurgical operations essentials of neurosurgical anesthesia & critical care. In:
Essentials of neurosurgical anesthesia & critical care. New York: Springer; 2012. p. 151–8.
12. Haure P, Cold GE, Hansen TM, Larsen JR. The ICP-Lowering effect of 10° reverse Trendelenburg position during craniotomy is stable during
a 10-minute period. J Neurosurg Anaesthesiol October 2003;15(4):297–301.
13. Tankisi A, Rolighed Larsen J, Rasmussen M, Dahl B, Cold GE. The effects of 10 degrees reverse trendelenburg position on ICP and CPP in
prone positioned patients subjected to craniotomy for occipital or cerebellar tumours. Acta Neurochir (Wien) July 2002;144(7):665–70.
14. Tankisi A, Cold GE. Optimal reverse trendelenburg position in patients undergoing craniotomy for cerebral tumors. J Neurosurg February
2007;106(2):239–44.
15. Rozet I, Vavilala MS. Risks and benefits of patient positioning during neurosurgical care. Anesthesiol Clin September 2007;25(3):631–53. x.
16. Manikandan S, Umamaheswara Rao GS. Effect of surgical position on pulmonary gas exchange in neurosurgical patients. Indian J Anaesth
2002;46(5):356–9.
17. Winfree CJ, Kline DG. Intraoperative positioning nerve injuries. Surg Neurol January 2005;63(1):5–18. discussion 18.
18. Schubert A. Positioning injuries in anesthesia: an update. Adv Anesth October 1, 2008;26:31–65.
19. Jannetta PJ, McLaughlin MR, Casey KF. Technique of microvascular decompression. Technical note. Neurosurg Focus 2005;18(5):E5.
20. Raphael J, Rosenthal-Ganon T, Gozal Y. Emergency airway management with a laryngeal mask airway in a patient placed in the prone position. J Clin Anesth November 2004;16(7):560–1.
21. Edgcombe H, Carter K, Yarrow S. Anaesthesia in the prone position. Br J Anaesth November 23, 2007;100(2):165–83.
22. Albin MS, Carroll RG, Maroon JC. Clinical considerations concerning detection of venous air embolism. Neurosurgery December 1978;3(3):380–4.
23. Kobayashi S, Sugita K, Tanaka Y, Kyoshima K. Infratentorial approach to the pineal region in the prone position: concorde position. Technical
note. J Neurosurg January 1983;58(1):141–3.
24. Kyoshima K. Arm-down Concorde position: a technical note. Surg Neurol June 2002;57(6):443–5. discussion 445–6.
25. Landeiro JA, Boechat S, Christoph Dde H, Gonçalves MB, Castro Ide, Lapenta MA, et al. Transoral approach to the craniovertebral junction.
Arq Neuropsiquiatr December 2007;65(4B):1166–71.
26. Crowley RW, Dumont AS, McKisic MS, Jane Sr JA. Positioning for cranial surgery. In: YOUMANS neurological surgery. 6th ed. Philadelphia,
PA: Saunders, Elsevier; 2011. p. 442–6.
27. Turgut N, Ali A, Turkmen A, Altan A, Bikmaz K, Iplikcioglu AC. Comparison of surgical approaches in neurosurgical patients experiencing
venous air embolism in the sitting position. West Indian Med J August 2014;63(4):388.
28. Gan C, King JA, Maartens NF. The role and safety of the sitting position in instrumented cervical surgery. J Clin Neurosci March 2016;25:75–8.
29. Gracia I, Fabregas N. Craniotomy in sitting position: anesthesiology management. Curr Opin Anaesthesiol October 2014;27(5):474–83.
30. Tindall GT, Craddock A, Greenfield JC. Effects of the sitting position on blood flow in the internal carotid artery of man during general anesthesia. J Neurosurg April 1967;26(4):383–9.
31. Martin JT, Warner MA, editors. Positioning in anesthesia and surgery. 3rd ed. Philadelphia, PA: Saunders, Elsevier; 1997. p. 72.
32. Voorhies RM, Fraser RA, Van Poznak A. Prevention of air embolism with positive end expiratory pressure. Neurosurgery May 1983;12(5):503–6.
33. Papadopoulos G, Kuhly P, Brock M, Rudolph KH, Link J, Eyrich K. Venous and paradoxical air embolism in the sitting position. A prospective
study with transoesophageal echocardiography. Acta Neurochir (Wien) 1994;126(2–4):140–3.
34. Longatti P, Marton E, Feletti A, Falzarano M, Canova G, Sorbara C. Carbon dioxide field flooding reduces the hemodynamic effects of venous
air embolism occurring in the sitting position. Childs Nerv Syst August 2015;31(8):1321–6.
35. Eckle VS, Neumann B, Greiner TO, Wendel HP, Grasshoff C. Intrajugular balloon catheter reduces air embolism in vitro and in vivo. Br J
Anaesth June 2015;114(6):973–8.
36. Cucchiara RF, Seward JB, Nishimura RA, Nugent M, Faust RJ. Identification of patent foramen ovale during sitting position craniotomy by
transesophageal echocardiography with positive airway pressure. Anesthesiology July 1985;63(1):107–9.
37. Angevine PD, Gigante PR. Patient positioning for spinal surgery. In: YOUMANS neurological surgery. 6th ed. Philadelphia, PA: Saunders,
Elsevier; 2011. p. 447–52.
38. Schonauer C, Bocchetti A, Barbagallo G, Albanese V, Moraci A. Positioning on surgical table. Eur Spine J October 2004;13(Suppl. 1):S50–5.
39. Park CK. The effect of patient positioning on intraabdominal pressure and blood loss in spinal surgery. Anesth Analg September 2000;91(3):552–7.
V. POSITIONS IN NEUROSURGERY
REFERENCES
205
40. Grisell M, Place HM. Face tissue pressure in prone positioning: a comparison of three face pillows while in the prone position for spinal surgery. Spine December 15, 2008;33(26):2938–41.
41. Grant GP, Turbin RE, Bennett HL, Szirth BC, Heary RF. Use of the Proneview Helmet system with a modified table platform for open access
to the eyes during prone spine surgery. Anesth Analg August 2006;103(2):499–500.
42. McMichael JC, Place HM. Face tissue pressures in prone positioning: a comparison of 3 pillows. J Spinal Disord Tech October 2008;21(7):508–13.
43. Palmon SC, Kirsch JR, Depper JA, Toung TJ. The effect of the prone position on pulmonary mechanics is frame-dependent. Anesth Analg
November 1998;87(5):1175–80.
44. Batson OV. The function of the vertebral veins and their role in the spread of metastases. Ann Surg July 1940;112(1):138–49.
45. Pearce DJ. The role of posture in laminectomy. Proc R Soc Med February 1957;50(2):109–12.
46. Ofiram E, Lonstein JE, Skinner S, Perra JH. “The disappearing evoked potentials”: a special problem of positioning patients with skeletal
dysplasia: case report. Spine June 15, 2006;31(14):E464–70.
47. Douglass J, Fraser J, Andrzejowski J. Awake intubation and awake prone positioning of a morbidly obese patient for lumbar spine surgery.
Anaesthesia February 2014;69(2):166–9.
48. Kriskovich MD, Apfelbaum RI, Haller JR. Vocal fold paralysis after anterior cervical spine surgery: incidence, mechanism, and prevention of
injury. Laryngoscope September 2000;110(9):1467–73.
49. Tan TP, Govindarajulu AP, Massicotte EM, Venkatraghavan L. Vocal cord palsy after anterior cervical spine surgery: a qualitative systematic
review. Spine July 1, 2014;14(7):1332–42.
50. Shih P, Slimack NP, Roy A, Fessler RG, Koski TR. Abdominal complications following posterior spinal fusion in patients with previous
abdominal surgeries. Neurosurg Focus October 2011;31(4):E16.
51. Akhavan A, Gainsburg DM, Stock JA. Complications associated with patient positioning in urologic surgery. Urology December
2010;76(6):1309–16.
52. Mobley SR, Miller BT, Astor FC, Fine B, Halliday NJ. Prone positioning for head and neck reconstructive surgery. Head Neck November
2007;29(11):1041–5.
53. American Society of Anesthesiologists Task Force on Prevention of Perioperative Peripheral Neuropathies. Practice advisory for the prevention of perioperative peripheral neuropathies: an updated report by the American Society of Anesthesiologists Task Force on prevention of
perioperative peripheral neuropathies. Anesthesiology April 2011;114(4):741–54.
54. Leibovitch I, Casson R, Laforest C, Selva D. Ischemic orbital compartment syndrome as a complication of spinal surgery in the prone position.
Ophthalmology January 2006;113(1):105–8.
55. Koreckij J, Price N, Schwend RM. Vectored cranial-cervical traction limits facial contact pressure from prone positioning during posterior
spinal deformity surgery. Spine July 1, 2011;36(15):E993–7.
56. Brown J, Rogers J, Soar J. Cardiac arrest during surgery and ventilation in the prone position: a case report and systematic review. Resuscitation
August 2001;50(2):233–8.
57. Minami K, Mimami K, Iida M, Iida H. Case report: central venous catheterization via internal jugular vein with associated formation of perioperative venous thrombosis during surgery in the prone position. J Anesth June 2012;26(3):464–6.
58. Hans P, Demoitié J, Collignon L, Bex V, Bonhomme V. Acute bilateral submandibular swelling following surgery in prone position. Eur J
Anaesthesiol January 2006;23(1):83–4.
59. Sinha A, Agarwal A, Gaur A, Pandey CK. Oropharyngeal swelling and macroglossia after cervical spine surgery in the prone position.
J Neurosurg Anesthesiol July 2001;13(3):237–9.
60. Yano T, Imaizumi T, Uneda C, Nakayama R. Lower intracuff pressure of laryngeal mask airway in the lateral and prone positions compared
with that in the supine position. J Anesth 2008;22(3):312–6.
61. Stambough JL, Dolan D, Werner R, Godfrey E. Ophthalmologic complications associated with prone positioning in spine surgery. J Am Acad
Orthop Surg March 2007;15(3):156–65.
62. Lee LA, Newman NJ, Wagner TA, Dettori JR, Dettori NJ. Postoperative ischemic optic neuropathy. Spine April 20, 2010;35(Suppl. 9):S105–16.
63. Li A, Swinney C, Veeravagu A, Bhatti I, Ratliff J. Postoperative visual loss following lumbar spine surgery: a review of risk factors by diagnosis. World Neurosurg December 2015;84(6):2010–21.
64. American Society of Anesthesiologists Task Force on Perioperative Visual Loss. Practice advisory for perioperative visual loss associated with
spine surgery: an updated report by the American Society of Anesthesiologists Task Force on perioperative visual loss. Anesthesiology February
2012;116(2):274–85.
V. POSITIONS IN NEUROSURGERY
This page intentionally left blank
S E C T I O N
V I
PREANESTHETIC EVALUATION
This page intentionally left blank
C H A P T E R
11
Preanesthetic Evaluation of Neurosurgical
Patients
R. Mariappan
Christian Medical College, Vellore, India
O U T L I N E
Introduction
Preoperative Evaluation of Patients With
Diabetes and Hypertension
Preoperative Medication Evaluation
Comprehensive Neurological Examination for
Neuroanesthesiologist
209
Preoperative Evaluation of Patient-Related
Risk Factors
210
Preoperative Evaluation of Cardiac Risk in
Neurosurgical Patients
210
Risk Assessment Tools for Estimating
Cardiac Risk
210
Assessment of Functional Capacity
211
Indication for Cardiac Testing During Preoperative
Evaluation
211
Evaluation of Pulmonary Risk in Patients Undergoing
Neurosurgical Procedure
211
Preoperative Pulmonary Risk Stratification
212
Indication for Preoperative Pulmonary Testing
212
Preoperative Evaluation of Patients With Obstructive
Sleep Apnea
213
Various Screening Tools for the Diagnosis of
Obstructive Sleep Apnea
213
Specific Issues to Be Considered During
Preoperative Evaluation of Patients With
Obstructive Sleep Apnea
213
Preoperative Evaluation of Specific Neurosurgical
Conditions
Preoperative Evaluation of Patients With
Supratentorial and Posterior Fossa Tumor
Preoperative Evaluation of Patients for
Awake Craniotomy
Preoperative Evaluation of Patients With
Pituitary Diseases
Preoperative Evaluation of Patients With Suprasellar
Lesions
Preoperative Evaluation of Patients With Epilepsy
Preoperative Evaluation of Patients With
Subarachnoid Hemorrhage
Preoperative Evaluation of Patients With
Arteriovenous Malformation
References
213
214
214
217
217
219
220
220
222
223
224
225
INTRODUCTION
A routine preanesthetic evaluation is a process that includes clinical assessment, risk stratification, and optimization before surgery. The primary aim of the preanesthetic evaluation is to reduce the perioperative morbidity and
mortality. This involves many steps, which include:
1. Establishing rapport with the patient and family, explaining the process of surgery, anesthesia, and recovery.
2. Reviewing the past medical, surgical, personal and family history and history of medication allergy and the lists
of current medication.
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00011-7
209
© 2017 Elsevier Inc. All rights reserved.
210
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
3. A through general and clinical examination focusing on cardiopulmonary system and nervous system.
4. Review of investigations and to assess whether further investigations or multidisciplinary care is needed, which
would help to improve the patient condition.
5. Risk-stratifying the patient for perioperative morbidity and mortality using clinical and laboratory data for the
proposed surgical procedure.
6. Optimization of the patient using the risk reduction strategies.
7. To plan the type of anesthesia after assessing the risk/benefit of each anesthetic technique for that particular
procedure.
8. Utilization of best available resources to maximize the safety, to reduce the morbidity and mortality, and to
speed up the recovery by providing adequate postoperative pain management.
9. Getting an informed consent after explaining the risk and benefits of anesthesia technique and the need for
invasive monitoring, blood transfusion (if applicable), and its associated risks.
Preanesthetic evaluations should be performed well ahead of the surgical procedure especially for high-risk
patients [American Society of Anesthesiologists (ASA) grades 3 and 4] thereby allowing time for preoperative optimization, which has been shown to reduce the perioperative morbidity and mortality.1,2 Patients with ASA grades
1 and 2 can undergo preoperative evaluation either on the day of surgery or the day before surgery. ASA classification of physical status is a universally accepted grading system to stratify the patient’s preexisting health condition.
Although it was not designed for outcome prediction, there has been a good correlation between the grading and
perioperative morbidity and mortality.3–5
Most patients undergoing neurosurgical procedures will have an underlying medical comorbidities such as
diabetes, hypertension, ischemic heart diseases (IHD), obstructive sleep apnea (OSA), and chronic obstructive
pulmonary disease (COPD), or bronchial asthma and seizures. These medical conditions require more intense
scrutiny than the pathological process to prevent perioperative morbidity and mortality. Each neurosurgical
procedure carries its inherent risk and requires specific preoperative evaluation and optimization. So, during
the preoperative evaluation both patient- and procedure-related factors should be kept in mind and optimized
accordingly.
PREOPERATIVE EVALUATION OF PATIENT-RELATED RISK FACTORS
In this section, the preoperative evaluation of patient-related risk factors focusing on cardiac and respiratory conditions, OSA, diabetes, and hypertension are discussed.
Preoperative Evaluation of Cardiac Risk in Neurosurgical Patients
Most neurosurgical procedures are considered to be intermediate- to high-risk surgeries. Patients with cardiac diseases undergoing neurosurgical procedure can have aggravation of cardiac dysfunction due to the systemic effect of
raised intracranial pressure (ICP) (tumor, head injury) or catecholamine surge [in subarachnoid hemorrhage (SAH)]
or autonomic dysfunction (Parkinson disease, cervical myelopathy, and brainstem lesion) leading to increased morbidity. Long-standing hypertension and diabetes are common in patients undergoing carotid endarterectomy and
spine surgery, and these patients are more prone for IHD. Identification of cardiac risk associated with each neurosurgical procedure provides information to both the patient and the surgeon, which in turn helps them to understand
the benefit vs. risk of a procedure. Cardiac interventions even before the surgery in certain high-risk cases decreases
the perioperative morbidity and mortality.
Risk Assessment Tools for Estimating Cardiac Risk
There are various risk assessment tools available to assess the cardiac risk in patient undergoing neurosurgery.
These tools use the information obtained from the history, physical examination, electro- and echocardiogram, and
type of surgery. The various tools are given below.
1. Revised Cardiac Risk Index (Lee’s) tool6: This is the most commonly used tool and was originally published in
1999; it has been used to assess the risk for >15 years. It uses six factors such as high-risk surgery, history of IHD,
congestive heart failure (CHF), cerebrovascular diseases, diabetes or insulin, and presence of high creatinine
to estimate the cardiac risk. Using the aforementioned predictors, the estimated risk of cardiac death, nonfatal
cardiac arrest, and nonfatal myocardial infarction (MI) following the elective surgical procedure are 0.4% [95%
VI. PREANESTHETIC EVALUATION
PREOPERATIVE EVALUATION OF PATIENT-RELATED RISK FACTORS
211
confidence interval (CI): 0.1–0.8] if there is no risk factor and 1.0% (95% CI: 0.5–1.4) if there is one risk factor. The
risk increases to 2.4% (95% CI: 1.3–3.5) and 5.4% (95% CI: 2.8–7.9) if there are two and three or more risk factors,
respectively.7
2. American College of Surgeons-National surgical Quality Improvement Program (ACS-NSQIP) universal surgical risk
calculator8: This surgical risk calculator model is a Web-based tool consisting of 20 patient factors such as
body mass index (BMI), age, sex, ASA classification, functional status, prior cardiac history, and so on. The
calculator then provides a risk of a major adverse cardiac event for the patient. This model had excellent
performance for mortality, morbidity, and six additional complications. Limitations of this tool are that it is
more cumbersome and its external validity is still questionable.
3. Gupta myocardial infarction/cardiac arrest (MICA)-NSQIP database risk model9: This risk model uses the following
five factors to assess the risk of perioperative MI and cardiac arrest. They are (1) type of surgery, (2) dependent
functional status, (3) abnormal creatinine, (4) American Society of Anesthesiologists’ class, and (5) increased age.
Assessment of Functional Capacity
Assessment of patients functional capacity is the next step in cardiac evaluation, expressed in metabolic
equivalents (METs). One MET is defined as 3.5 mL of O2 uptake/kg/min in a sitting position, which is a resting
oxygen uptake. Duke activity state Index is one of the most frequently used scales to assess the functional status.10 Ability to take care of oneself, such as eating, dressing, or using the toilet is considered 1 MET. Walking up
a flight of steps or a hill or walking on level ground at 3–4 mph is considered as 4 METs. Participating in strenuous sports such as swimming, singles tennis, football, basketball, and skiing is equivalent to >10 METs. Ability
to do an activity that requires >4 METs indicates good functional activity. Assessing the functional capacity in
patients with walking disability such as paraplegia or hemiplegia can be done by using bicycle or arm ergometry stress testing.11 After determining the cardiac risk and functional capacity, one can follow the multistep
algorithm provided by the American College of Cardiology (ACC) and American Heart Association (AHA) for
determining the need for further preoperative cardiac evaluation before proceeding with surgery. Refer ACC
and AHA 2014 guideline for details.12
Indication for Cardiac Testing During Preoperative Evaluation
Electrocardiogram (ECG): All patients with cardiac problems such as IHD, arrhythmias, valvular heart diseases
(VHD), and peripheral arterial diseases should obtain a baseline ECG to compare the changes that occur during the
perioperative period. Patients with neurological problem such as raised ICP or SAH have underlying cardiac dysfunction, they often need baseline ECG.
Echocardiography: It is indicated to assess the ventricular function in symptomatic patients (dyspnea or heart failure), patients with VHD, and patients with past history of previously documented cardiac dysfunction with no
assessment within 1 year.13
Stress echocardiography: Although there is a correlation between the degree of MI and the prognosis, there is no evidence that prophylactic revascularization at the time of surgery improves outcomes. So, further cardiac evaluation
(stress echocardiography or 24-h ambulatory monitoring) is only indicated in patients with known IHD with recent
deterioration.14
Evaluation of Pulmonary Risk in Patients Undergoing Neurosurgical Procedure
The incidence of perioperative pulmonary complications (POPCs) are high in patients with preexisting lung
(obstructive, restrictive) diseases. Perioperative hypoxia and hypercapnia not only affect the cardiorespiratory status
but also can aggravate the existing neurological illness. Neurosurgical patients are more prone to pulmonary complications because of low Glasgow Coma Scale (GCS) score (tumor, head injury, seizure, SAH), lower cranial nerve
dysfunction in posterior fossa tumors or cranio vertebral junction anomalies causing aspiration pneumonia, or the
presence of a high cervical or thoracic spine lesion causing cord compression leading to restrictive lung diseases.
POPCs contribute significantly to overall perioperative morbidity and mortality in the neurosurgical population. According to the NSQIP report, POPCs are the costliest of all postoperative medical complications (including cardiac, thromboembolic, and infectious) and resulted in the longest length of hospital stay.15,16 POPC is
defined as any pulmonary disease or dysfunction that is clinically significant or adversely affects the clinical
course of the patient.17,18 They are (1) atelectasis, (2) infection including bronchitis and pneumonia, (3) prolonged mechanical ventilation (>48 h), (4) respiratory failure, (5) exacerbation of underlying chronic lung disease, and (5) bronchospasm.
VI. PREANESTHETIC EVALUATION
212
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
Preoperative Pulmonary Risk Stratification
A complete history and physical examination are the most important elements of preoperative pulmonary risk
assessment. Based on the available literature, the risk factors for POPC are classified into definite and probable,
which are listed in Table 11.1.19,20 There are many risk prediction tools available to stratify the postoperative pulmonary risk21,22; they are (1) ARISCAT (Canet) risk index used for predicting POPC,21 (2) Arozullah index for predicting
postoperative pneumonia23 and respiratory failure,24 (3) Gupta calculator for predicting postoperative pneumonia25
and respiratory failure,26 and (4) ASA grading for predicting POPC.5
1. ARISCAT (Canet) risk index: It predicts the overall incidence of postoperative pulmonary complications using the
patient’s age, preoperative oxygen saturation, recent respiratory infection, preoperative anemia, type of surgical
incision, and duration of surgery. It has a total of 165 points; patients with low scores (<26 points) are considered
as low risk and patients with high score (>45 points) are considered as high risk for POPC. According to this
risk index the incidence of POPC is 1.6%, 13.3%, and 42.2% for low-, intermediate-, and high-risk classes. It is a
very simple tool that calculates the risk manually at the bedside using the readily available clinical information.
The limitation of this tool is it that includes even the minor complications such as wheezing treated with
bronchodilators, which is not clinically significant.
2. ASA classification: ASA classification has been shown to predict the postoperative pulmonary complication.5 The
incidence of POPC for ASA grades 1, 2, 3, and 4 are 1.2%,5.4%, 11.4%, and 10.9%, respectively.
Indication for Preoperative Pulmonary Testing
1. Chest X-ray: Recent chest X-ray (within 6 months) is warranted for patients older than 50 years with
cardiopulmonary diseases undergoing major surgery.27
2. Pulmonary function test (PFT): Based on a systematic review, the American College of Physicians guideline
recommends that PFT should not be used for risk stratification. It should be reserved to determine the clinical
cause only in patients with COPD and asthma, in whom clinical evaluation cannot determine the reduction in
airflow obstruction, and in patients with unexplained dyspnea or poor exercise tolerance.28
3. Arterial blood gas (ABG) analysis: In patients with severe COPD, PaCO2 >45 mmHg is one of the probable risk
factors for POPC. However, routine ABG is not warranted for risk stratification.29
Preoperative risk reduction strategies to reduce POPCs are as follows28:
1. Smoking cessation for more than 8 weeks prior to surgery has been shown to reduce the perioperative morbidity
and mortality.30,31
2. Preoperative administration of inhaled bronchodilators and glucocorticoids in patients with COPD and
asthma.32,33
3. Treating the exacerbation of asthma and COPD with systemic glucocorticoids (e.g., prednisone 40 mg/day for
5 days) and treating the lower respiratory tract infection with antibiotics and postponing the elective surgery.34
4. Preoperative education regarding lung expansion maneuvers and initiation of chest physiotherapy such as
aerobic exercises, breathing exercises, and inspiratory muscle training.35
TABLE 11.1 Risk Factors for POPC
Definite Risk Factors
Probable Risk Factors
Patient related
1. Age >65 years
2. Poor general health status (ASA class >2)
3. Functional dependence
4. Heart failure
5. Chronic obstructive lung disease
Procedure related
6. Upper abdominal, open thoracic, head and neck, neurosurgical,
and abdominal aortic aneurysm surgery
7. Emergency surgery
8. Anesthesia lasting >3 h,
9. Use of long-acting muscle relaxants
1. General anesthesia (compared to spinal, epidural anesthesia, or
other regional anesthetic techniques)
2. Arterial tension of carbon dioxide (PaCO2) >45 mmHg (5.99 kPa)
3. Postoperative nasogastric tube placement
4. Abnormal chest radiograph
5. Cigarette use within the previous 8 weeks
6. Current upper respiratory tract infection
Test predictors
10. Albumin level <3 g/dL
ASA, American Society of Anesthesiologists; POPC, postoperative pulmonary complication.
VI. PREANESTHETIC EVALUATION
PREOPERATIVE EVALUATION OF PATIENT-RELATED RISK FACTORS
213
Preoperative Evaluation of Patients With Obstructive Sleep Apnea
OSA is a sleep-related breathing disorder characterized by repetitive episodes of apnea or hypopnea due to upper
airway obstruction during sleep. It is classified into mild, moderate, and severe according to the Apnea–Hypopnea
Index (AHI). AHI of 5–15 per hour is mild OSA, AHI of 15–30 per hour is considered as moderate OSA, and AHI >30
per hour is considered as severe OSA.
Over the past two decades, with the parallel increase in obesity, the prevalence of OSA is rapidly increasing.36
Studies have reported that the incidence of OSA is >30% in neurosurgical population. Patients with Cushing
disease, acromegaly, intractable epilepsy, and intracranial tumors are more prone to OSA.37–39 Studies and metaanalysis have concluded that the incidence of postoperative desaturation, respiratory failure, postoperative cardiac events, and intensive care unit (ICU) transfers were higher in patients with OSA.40,41 Over half of patients
with OSA who present for surgery are undiagnosed42; the complication rates are two- to fourfold higher when
compared with those who are optimized preoperatively.43 OSA-related postoperative complications are common
because of the following reasons:
1. Administration of perioperative medication (sedatives, opioids, neuromuscular blocking drugs)
2. Worsening of airway edema (endotracheal tube placement, throat pack application, tracheal retraction, headdown positioning, prone position)
3. Discontinuation of continuous positive airway pressure (CPAP) therapy in patients undergoing transsphenoidal
surgery
4. Sleep deprivation due to pain, anxiety, and ICU environment can all lead to increased rapid eye movement
sleep, which increases the OSA episodes44
5. Presence of OSA-related comorbidities [obesity systemic hypertension, obesity hypoventilation syndrome
(OHS), pulmonary hypertension, cardiac arrhythmias, coronary artery disease, and heart failure]
Various Screening Tools for the Diagnosis of Obstructive Sleep Apnea
1. STOP-BANG questionnaire45,46: It is a very simple tool to screen, and it is the most frequently used
questionnaire in our practice. It requires “Yes” or “No” responses to eight questions about snoring,
tiredness, observed apnea, and blood pressure; body mass index>35 kg/m2, age >50 years, neck
circumference >40 cm, and male gender. Patients with zero to two positive responses are considered “low
risk,” those with three to four are considered “intermediate risk,” and those with five to eight positive
responses are considered “high risk for OSA.” Patients with raised serum bicarbonate level (≥28 mmol/L)
with a STOP-BANG score of ≥3 are equals to STOP-BANG score of ≥5. These patients are considered to have
moderate to severe OSA.47
2. Sleep Apnea Clinical Score—Flemons’ screening tool48: It is four-item questionnaire (habitual snoring, nocturnal
gasping/choking, neck circumference, hypertension) with the score ranging from 0 to 100. Values >15 indicate
that the patient is at high risk for OSA.
3. Berlin Questionnaire49: This uses items such as snoring, excessive daytime sleepiness, sleepiness while driving,
apnea during sleep, hypertension, and BMI to stratify patients as having a high or low risk for OSA.
Specific Issues to Be Considered During Preoperative Evaluation of Patients With Obstructive
Sleep Apnea
While evaluating patients with OSA, the symptoms and signs of OSA; the duration, severity, and presence of
OSA-related comorbidities, type of treatment given (CPAP or bilevel positive airway pressure); response to treatment; and the details of current airway pressure setting all, need to be documented. Patients with long-standing
OSA are more prone for OHS and pulmonary hypertension.50 Elevated bicarbonate level and hypoxemia are
the indicators OHS. Presence of OHS and pulmonary hypertension indicates the need for echocardiography.
Patients with severe OSA have to be admitted and optimized at least 1 week prior to surgery. Practice Guidelines
for the Perioperative Management of Patients With OSA—an updated report by the American Society of
Anesthesiologists Task Force—was published in 2014 based on expert opinion, literature review, and consensus.51
Preoperative Evaluation of Patients With Diabetes and Hypertension
Detailed discussion on preoperative evaluation on both these conditions are beyond the scope of this chapter. Specific
concerns regarding diabetes and hypertension and their anesthetic implications are given in Tables 11.2 and 11.3.
VI. PREANESTHETIC EVALUATION
214
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
TABLE 11.2 Specific Concerns While Evaluating Patients With DM
Specific Concerns
Details Regarding Specific Points
1. Type and duration of diabetes
Patients with type I DM are more prone for perioperative complications. Longer
duration of diseases is associated with increased perioperative complications
2. Treatment history
Insulin: Type of insulin, dose, frequency, timing. OHA: Type, dose, frequency,
complication of drugs (lactic acidosis)
3. Adequacy of glycemic control
Trends and ranges of blood glucose level during the immediate preoperative
period including HbA1C (reveals 3 months’ control). Incidence of hypoglycemia,
and its severity
Perioperative administration of dexamethasone can cause aggravation of
hyperglycemia
4. Presence of long-term complications
Look for signs of neuropathy, retinopathy, nephropathy, peripheral vascular
diseases, and IHD—increases the perioperative morbidity and mortality
DM, diabetes mellitus; HbA1C, glycosylated hemoglobin; IHD, ischemic heart diseases; OHA, oral hypoglycemic agents.
TABLE 11.3 Specific Concerns While Evaluating a Patient With Hypertension
Specific Concerns
Details Regarding Specific Points
1. Duration of hypertension
Long-standing hypertension is associated with LVH and its associated
complication (systolic and diastolic dysfunction), which increases the
perioperative morbidity
2. Type of antihypertensive
β-Blocker/calcium channel blocker/ACE inhibitor/ARB/central sympatholytic
drugs
Drug effects and its anesthetic implications are discussed in detail in medication
evaluation section
3. Adequacy of blood pressure control
Blood pressure trends during the perioperative period should be noted
especially in the presence of white coat hypertension
4. Presence of long-term complications
Look for LVH as well as systolic and diastolic dysfunction. Presence of these
complications are associated with increased perioperative morbidity
ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker; LVH, left ventricular hypertrophy.
Preoperative Medication Evaluation
Patients undergoing neurosurgical procedures will be on multiple medication, either for their neurological
diseases itself (anticonvulsants, antiedema drugs, H2 blockers, or proton pump inhibitors) or for their associated
medical comorbidities. Most medications are known to cause interaction with anesthetics. There are large variations in recommendation among the anesthesiologist in regard to continuing/discontinuing certain drugs during the perioperative period. Every physician should balance the risks vs. benefits of continuing/discontinuing
the drug during the perioperative period. Commonly used drugs in neurosurgical practice and their anesthetic
interactions and the recommendation regarding whether they need to be continued or stopped before surgery are
given in Table 11.4. These recommendations are based on expert opinions and their reviews and the theoretical
considerations.
Comprehensive Neurological Examination for Neuroanesthesiologist
If an anesthesia technique (general or regional) can affect an organ system, then that organ system should be
evaluated and documented preoperatively by the anesthesiologist. Although most patients’ neurological status is
assessed and documented by the neurosurgeon before coming to the preanesthesia clinic, it is better that every
neuroanesthesiologist know how to perform a comprehensive neurologic screening examination. While doing a
preoperative evaluation of neurosurgical patients, the pulmonary and cardiac examination should be done prior
to neurological examination to integrate other physical findings with the presenting neurological abnormality. For
VI. PREANESTHETIC EVALUATION
215
PREOPERATIVE EVALUATION OF PATIENT-RELATED RISK FACTORS
TABLE 11.4 Showing the Commonly Used Medications in Neurosurgical Practice and the Benefits/Risk of Continuing These Drugs in
the Perioperative Settings
Drug
Strategy to Continue
Yes/No
Β$Blockers
Yes
Perioperative Anesthetic Implications
Benefits/Anesthetic Drug Interaction
Risks
Decreases the myocardial ischemia by
reducing the oxygen demand
Increases the risk of perioperative
bradycardia and hypotension
Controls and prevents arrhythmias
Nonselective β-blockers can interact with
epinephrine or ephedrine and cause
hypertensive crisis due to unopposed
alpha stimulation
Perioperative initiation reduces the
cardiovascular morbidity but increases the
risk of stroke (POISE I trial) in high-risk
cardiac patients.
Abrupt withdrawal increases the
cardiovascular morbidity
α-Blocker (clonidine)
Calcium channel blocker
Yes
Yes
Improves the perioperative outcome
(proven only in a smaller studies)
Increases the incidence of hypotension and
nonfatal cardiac arrest. (POISE II trial)
Abrupt withdrawal can cause rebound
hypertension and myocardial ischemia
Perioperative initiation is not warranted
No interaction with anesthetic drugs
Abrupt withdrawal can cause coronary
spasm
Since it decreases the platelet aggregation,
there is a conflicting opinion regarding its
bleeding risk
ACE inhibitor and
angiotensin II receptor
blocker
Yes/no
Decision to continue or discontinue
depends on the patient’s condition
Increased incidence of perioperative
hypotension and the need for inotropes
According to ACC/AHA 2014 guidelines— Withdrawal can cause postoperative
to continue if the patient is taking for
hypertension
hypertension or CHF
Risk vs. benefits of intraoperative
hypotension should be kept in mind
Diuretics
Yes/no
No consensus available regarding
discontinuation
HMG CoA reductase
inhibitors (statins)
Yes
Reduces the cardiovascular and
cerebrovascular morbidity because of
its cholesterol-lowering effect, plaque
stabilization, antiinflammatory, and
decreased thrombogenesis effects
Increased incidence of perioperative
hypotension and hypokalemia
No interaction with anesthetic drug
OHAs
No
No interaction with anesthetic drugs
Intraoperative hypoglycemia with
sulfonylureas
Increased incidence of lactic acidosis with
metformin
Thiazolidinediones worsen the peripheral
edema and fluid retention and HF
Sodium–glucose cotransporter 2 inhibitors
increase the risk of hypovolemia
DPP-IV inhibitors and GLP-1 analogs
alter the GI motility and worsen the
postoperative state
AEDs
Yes
Continued
VI. PREANESTHETIC EVALUATION
216
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
TABLE 11.4 Showing the Commonly Used Medications in Neurosurgical Practice and the Benefits/Risk of Continuing These Drugs in
the Perioperative Settings—cont’d
Drug
Strategy to Continue
Yes/No
Carbamazepine, phenytoin,
Hepatic microsomal
isoenzyme (CYP 450)
inducer
Perioperative Anesthetic Implications
Benefits/Anesthetic Drug Interaction
Risks
Drugs that causes enzyme induction
accelerate the metabolism of drugs
metabolized by this enzyme system,
thereby increasing the requirement of
propofol, opioid, and muscle relaxants
Oxcarbamazepine,
eslicarbazepine: weak
enzyme inducer
Sodium valproate: enzyme
inhibition
Drugs that cause enzyme inhibition
decrease the metabolism of drugs
metabolized by this enzyme system,
thereby decreasing the requirement of
propofol, opioid, and muscle relaxants
Gabapentin, lamotrigine,
levetiracetam, tiagabine,
vigabatrin: do not affect
hepatic enzyme system
Aspirin (low dose
75–300 mg)
No
Neurosurgical operations are closed space
surgery and considered to be moderateto high-risk surgeries, aspirin has to be
stopped 7 days prior to surgery
Platelet P2Y12 receptor
blockers
Increases the bleeding risk if it is
continued.
Increased bleeding risk
Clopidogrel
No
Clopidogrel to be stopped 5–7 days prior to
surgery
Ticlopidine
No
Ticlopidine had to be discontinued 10 days
prior to surgery
Dipyridamole
No
Dipyridamole has to be discontinued 2 days
prior to surgery
Aggrenox
(Aspirin + dipyridamole)
No
Aggrenox had to be stopped 7–10 days
prior to surgery
H2 blockers and proton
pump inhibitors
Yes
Decreases the mucosal damage, decreases
Cimetidine is the only drug that interferes
the gastric secretion, and increases the
with many drug metabolism
gastric pH thereby decreasing the chance of
aspiration pneumonitis
TCAs
No
(Imipramine, amitriptyline,
nortriptyline, desipramine,
and clomipramine)
Abrupt withdrawal causes insomnia,
Increases the risk of perioperative
nausea, headache, increased salivation, and arrhythmia in combination with volatile
sweating and should be avoided
anesthetics and sympathomimetics
SSRIs
No
Abrupt discontinuation can lead to
dizziness, chills, muscle aches, and anxiety
SSRIs increases the bleeding risk so, it
has to be tapered and stopped 2–3 weeks
earlier. It has to be replaced with another
regime of antidepressant to avoid
exacerbation of mood disorder
No
Causes accumulation of biogenic amines in
central and autonomic system neurons by
causing irreversible inhibition of MAO
Concomitant administration of
sympathomimetic agents can result in
hypertensive crisis
(Paroxetine, fluvoxamine,
sertraline, fluoxetine)
MAO inhibitors
With pethidine and tramadol, it can cause
serotonin syndrome
VI. PREANESTHETIC EVALUATION
PREOPERATIVE EVALUATION OF SPECIFIC NEUROSURGICAL CONDITIONS
217
Table 11.4 Showing the Commonly Used Medications in Neurosurgical Practice and the Benefits/Risk of Continuing These Drugs in the
Perioperative Settings—cont’d
Strategy to Continue
Yes/No
Drug
(Isocarboxazid, pargyline,
phenelzine, and
tranylcypromine)
Perioperative Anesthetic Implications
Benefits/Anesthetic Drug Interaction
Risks
Recovery of MAO function takes 2 weeks
after discontinuation of the drug. So, drug
should be tapered and stopped 2 weeks
before elective surgery
Coadministration of pethidine can cause
type I serotonin syndrome
By inhibiting the hepatic microsomal
enzyme system responsible for opioid
metabolism, it can cause type II reaction.
(sedation, respiratory depression, and
cardiovascular collapse)
Antipsychotics
Yes/no
Antipsychotic effectively controls the
psychoses in vulnerable patients.
Both typical and atypical antipsychotic
drugs are associated with an increased
risk for sudden death because of QT
prolongation and arrhythmogenic effect
Should be used cautiously in the
perioperative settings. Frequent ECG
monitoring is needed especially in the
perioperative settings
Potentiates the effect of sedatives,
anaesthetics, and analgesics
ACC, American College of Cardiology; ACE, angiotensin-converting enzyme; AED, antiepileptic drugs; AHA, American Heart Association; CHF, congestive heart
failure; DPP-IV, dipeptidyl peptidase IV; ECG, electrocardiogram; GLP-1, glucagon-like peptide 1; GI, gastrointestinal; HMG-CoA, 3-hydroxy-3-methyl-glutarylcoenzyme A; MAO, monoamine oxidase; OHA, oral hypoglycemic agents; SSRIs, selective serotonin reuptake inhibitor; TCA, tricyclic antidepressants.
example, raised ICP decreases the pulmonary compliance and causes cardiac conduction abnormality. Similarly,
SAH can cause myocardial dysfunction and pulmonary edema due to severe catecholamine surge.
The purpose of the neurological examination is:
1.
2.
3.
4.
To determine the general location and extent of the neurological lesion.
To document the nervous system malfunction so that perioperative comparison is easier.
To determine and document the patient’s preoperative physical status and stability.
To develop an appropriate anesthesia management plan according to their physical status.
Neurological examination can be accomplished within minutes if it is performed in an organized and systematic
fashion. Based on the patient’s chief complaint or findings on the screening examination, components of the evaluation are then elaborated upon. For example, a full mental status examination is not necessarily warranted in the
patient who is awake, oriented, and conversant, while it must be fully investigated in patients with altered mental
status or a history of a change in behavior. Likewise, in a patient with no sensory complaints, determination of the
ability to distinguish sharp from dull bilaterally is sufficient, while a patient complaining of vague sensory deficits
needs to be tested for extinction on simultaneous sensory stimulation on both the limbs for assessing the sensory
level, or for astereognosis (inability to identify an object by palpation). A comprehensive method to perform a quick
neurological examination is listed in Table 11.5.
PREOPERATIVE EVALUATION OF SPECIFIC NEUROSURGICAL CONDITIONS
Preoperative Evaluation of Patients With Supratentorial and Posterior Fossa Tumor
Brain tumors are the leading cause of cancer-related death in children and young adults with an estimated
prevalence of 222/100,000.52 While evaluating a patient with supra- or infratentorial tumor one should focus
on history including the current medication, neurological examination, relevant general and systemic examination, radiological examination, review of laboratory investigation, and the probable diagnosis. The main
aim of preoperative evaluation is to find out the extent of impairment in cerebral autoregulation and intracranial compliance. Patients who are at the edge of intracranial compliance have to be recognized and treated
VI. PREANESTHETIC EVALUATION
218
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
TABLE 11.5 Methods of Performing a Comprehensive Neurological Examination
Systems
Performing a Quick Neurological Examination
Mental status
Appearance, mood, thought processes, cognitive function, and speech can be examined while
interacting with the patient
Altered mental status can be examined using “AVPU” score (A: Alert; V: response to Verbal stimuli; P:
response to Painful stimuli U: Unresponsive) and glasgow coma S score
Motor system
Observe the patient’s gait, ability to perform toe and heel walk.
Pronator drift and hand grasp test on the ulnar side: for checking the upper limb weakness
Lower limb drift and ability to perform dorsiflexion and plantarflexion: for checking the lower limb
weakness
If there is an abnormal test result with motor testing as described earlier, more formal testing of the
extremity
Motor power testing: using 0–5 point score. Grade 0 being no movement and grade 5 being full range of
movements against gravity and resistance
Reflex testing: biceps (C5, C6), triceps (C7, C8), and patellar (L2, L3, L4) and tendoachilles reflex (S1, S2)
and plantar reflex (Babinski sign for UMN lesion) to be tested. Response is graded 0 (no response) to 4+
(hyperreflexia)
Note: UMN lesion: spasticity, hyperreflexia, no atrophy or fasciculation on muscle. LMN lesion: flaccidity,
hyporeflexia, atrophy or fasciculation of muscle will be present
Sensory system
Sensory examination is done by testing the pain and light touch on patient’s hands, feet, and limbs
bilaterally. Simultaneous sensory extinction testing helps to identify the subtle central nervous system
sensory deficits
Posterior column testing is generally reserved for patients with suspected neuropathies or patients who
present with sensory symptoms
Note: Anterior spinothalamic tract carries temperature, pain, and light touch fibers, while the posterior column
carries fibers for crude touch, vibration, two-point discrimination, and proprioception
Cranial nerve examination
By history taking and by observing the patient, the cranial nerves are examined. Cranial nerves II, III,
IV, and VI are critical components of the screening examination and must be carefully assessed in all
patients with neurologic complaints
Cranial nerve II: testing of visual acuity, field, and ophthalmoscopic examination
Cranial nerve III: bilateral pupil size and symmetry. Adduction and vertical gaze is checked
Cranial nerve IV: internal depression via the superior oblique
Cranial nerve VI: abduction via lateral rectus is tested
Cerebellar function (coordination
and balance)
Balance requires an integration of cerebellar, visual, vestibular, and proprioception input. It is evaluated
by the Romberg test
Romberg test: The patient is asked to stand with his feet together (proprioceptive input), without
support; first with eyes open (visual input) and then with eyes closed. Closing the eyes eliminates
vision, but with proprioception and vestibular sense intact, the patient will not sway. If there is a
proprioceptive deficit, the patient will keep his balance with the eyes open and lose his balance when
the eyes are closed. When this occurs, the Romberg test is positive. This indicates posterior spinal
column diseases
If the patient sways with the eyes open or closed, this is suggestive of a cerebellar lesion that is not
compensated by sensory input from the other systems
Vertical nystagmus is always central in origin and it persists even at rest, does not fatigue, and does not
change with position. (Nylen–Barany maneuver)
appropriately to avoid the permanent neurological deficit. The details of preoperative evaluation in patients
undergoing supratentorial and infratentorial tumor surgery are given in Tables 11.6 and 11.7. Each tumor type
has its own anesthetic significance, so it is important to know the tumor type, its vascularity, and the position of
the patient during the surgery. This will help us to plan the anesthetic technique and the type of vascular access
needed during surgery.
VI. PREANESTHETIC EVALUATION
PREOPERATIVE EVALUATION OF SPECIFIC NEUROSURGICAL CONDITIONS
219
TABLE 11.6 Preoperative Evaluation of Patients With Supratentorial Tumor
History Focusing on
1. Symptoms of raised ICP: headache, nausea, vomiting, blurring of vision, somnolence
2. Symptoms of focal neurological deficit: motor, sensory and cranial nerve deficit
3. Seizure: type, frequency, antiepileptic use, duration of treatment, response to medication
4. Steroid use: duration, side effects of steroids (hyperglycemia), long-term steroid use warrants perioperative steroid cover
5. Diuretic use: associated hypovolemia. Intraoperative hypotension is severe; can be detrimental to patients with raised ICP
6. Past history of chemotherapy or radiation: certain chemotherapeutic medications are associated with complications such as
cardiomyopathy (doxorubicin) or inhibition of plasma cholinesterase activity (cyclophosphamide) and renal or hepatotoxicity
Physical Examination
1. Signs of raised ICP: low GCS, hypertension, bradycardia, and papilledema: indicates low intracranial compliance
Pupillary asymmetry, hemiparesis, aphasia, deep shallow breathing: indicates decompensated state
2. Other systemic examination focusing the CVS and RS and renal system and risk stratification (discussed separately)
3. Estimation of volume status: because of associated vomiting, diuretic use, SIADH, DI
Radiological Examination Using MRI or CT Brain
(1) Location (eloquent or noneloquent); (2) size of the tumor and surrounding edema; (3) presence of contrast enhancement (increased
vascularity) and involvement of major blood vessel (high risk for bleeding); (4) signs of raised ICP such as midline shift (>5 mm), presence
of ventricular effacement or hydrocephalus, and transtentorial or subfalcine herniation; (5) probable diagnosis: meningioma and high-grade
glioma; metastasis: bleeding risk is high
Others
(1) Type of craniotomy planned (frontal, parietal, temporal); (2) position of the patient (supine, supine oblique, lateral, or prone); (3) the need for
neuromonitoring technique such as SSEP, MEP, cortical mapping (tumor arising from eloquent areas); (4) need for lumbar drain in patients with
deep-seated tumors to avoid excessive brain retraction
CT, computed tomography; CVS, cardiovascular system; DI, diabetes insipidus; GCS, Glasgow Coma Scale; ICP, intracranial pressure; MEP, motor evoked potential;
MRI, magnetic resonance imaging; RS, respiratory system; SIADH, syndrome of inappropriate antidiuretic hormone; SSEP, somatosensory evoked potential.
TABLE 11.7 Preoperative Evaluation in Patients With Posterior Fossa Lesion
Symptoms
1. Headache, nausea, vomiting (raised ICP or direct compression of vagal nucleus or area postrema), papilledema (raised ICP)
2. H/o gait disturbance (cerebellar compression)
3. H/o vertigo (brainstem compression)
4. H/o facial deviation or asymmetry (seventh cranial nerve involvement)
5. H/o hearing loss, tinnitus (eighth cranial nerve involvement)
6. H/o dysphagia, and frequent aspiration (lower cranial nerve IX, X, and XI involvement)
7. H/o hoarseness of voice (10th cranial nerve involvement)
Signs
1. Cushing response due to raised ICP as well as brainstem compression.
2. Ataxia, tremor, dysmetria, (cerebellar hemisphere), truncal ataxia, titubation, broad based gait (cerebellar vermis) ipsilateral cranial nerve
deficit and opposite sensory and motor deficit, rotatory or vertical nystagmus (brainstem involvement).
3. Signs of cranial nerve dysfunction (facial, auditory, glossopharyngeal, vagus): need for postoperative ventilation.
4. Assessment of volume status: to avoid precipitous drop in BP after induction and positioning.
5. CVS examination: to identify the PFO; PFO is one of the relative contraindication for sitting positioning. Presence of long-standing diabetes,
Hypertension with LVH, autonomic dysfunction, cervical canal stenosis, presence of a functioning VP shunt are contraindications for sitting
position.
6. Cerebellar hemangioblastoma has strong association with pheochromocytoma and VHL syndrome: This has to be evaluated and treated
before surgery. Vestibular schwannoma is associated with neurofibromatosis type II.
Radiological Examination of MRI or CT
(1) Location; (2) size of the tumor and surrounding edema and degree of brainstem compression; (3) features of raised ICP: severity of
hydrocephalus, presence of transtentorial (upward), or Tonsillar herniation; (4) probable diagnosis using the radiological findings
Other Considerations
1. Type of craniotomy (approach), position of the patient (lateral, prone, park bench position, and sitting position), need for neuromonitoring
technique such as SSEP and MEP, facial and other lower cranial nerve monitoring
BP, blood pressure; CT, computed tomography; CVS; ICP, intracranial pressure; LVH, left ventricular hypertrophy; MEP, motor evoked potential; MRI, magnetic
resonance; PFO; SSEP, somatosensory evoked potential; VHL; VP.
Preoperative Evaluation of Patients for Awake Craniotomy
Awake craniotomy is the technique of choice for tumors involving eloquent cortical areas and procedures
for epilepsy and movement disorders. It is a safe, well-tolerated procedure; careful patient selection is the key
to success. Benefits of awake craniotomy are (1) better preservation of eloquent function in tumor surgery,53,54
VI. PREANESTHETIC EVALUATION
220
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
(2) better localization of seizure foci with complete resection in seizure surgery,55 and (3) reduces the hospitalization
time and cost when it is performed routinely for some neurosurgical procedure such as small tumor resection,
ventriculostomy, and for functional neurosurgery.56 Presence of alcohol or drug abuse, chronic pain disorders, low
tolerance to pain, anxiety, and psychiatric disorders are known to cause sedation failure during the procedure.
Presence of difficult airway, OSA, morbid obesity, poor seizure control even with multiple medication, and inability to lie flat (orthopnea) are some of the relative contraindications for awakecraniotomy.57,58 Although there are
reports of safe use of awake craniotomy in pediatric patients, case selection should be more rigorous and based
on the patients’ maturity and the individual risk–benefit assessment.59 Good preoperative psychological preparation, explaining about the level of cooperation needed, and the realistic description of the entire procedure with
expected discomforts at certain time period (during craniotomy and dural opening) will help the patient to tolerate the procedure well. A well-conducted preoperative consultation can alleviate the anxiety and improve their
cooperation during awake craniotomy.
Preoperative Evaluation of Patients With Pituitary Diseases
Pituitary gland is a master endocrine gland which controls the other endocrine glands of the body. Pituitary
tumors are classified as either micro or macroadenoma according to the size of the tumor (<10 mm: microadenoma, >10 mm: macroadenoma) or it is classified as functioning or nonfunctioning tumor according to presenting features. Microadenomas are often present with features of hormonal hypersecretion and macroadenomas
are nonfunctioning tumors that often present with pressure symptoms such as headache, visual field defect, or
features of raised ICP due to obstruction of cerebrospinal fluid flow at the level of third ventricle. Sometime macro
adenomas can bleed into the tumor and present with hormonal hyposecretion symptoms, otherwise called pituitary apoplexy. These patients often need supplements of steroid and thyroid hormone. The common presentation
of pituitary tumors, their associated problems, and their anesthetic significance and specific investigations are
discussed in detail in Table 11.8.
Preoperative Evaluation of Patients With Suprasellar Lesions
Common suprasellar lesions are craniopharyngioma, diaphragma sella meningioma, epidermoid, optic chiasmal
glioma, Rathke cleft cyst. Its clinical presentations depend upon the location of the tumor and the involvement of
(compressive symptoms) of neighboring structures. Preoperative investigation: In the presence of endocrine dysfunction, apart from routine laboratory investigation, full endocrine workup needs to be done to know the degree of
dysfunction and for optimization. In the presence of diabetes insipidus, serum sodium, serum and urine osmolality,
and urine specific gravity all, need to be checked and the patient had to be optimized before surgery. Computed
tomography (CT) and magnetic resonance imaging (MRI) of the brain had to be seen to know the size and extent of
the tumor such as cavernous sinus invasion and carotid artery encasement. Adequate blood needs to be arranged in
the presence of vascular involvement
TABLE 11.8 Showing the Common Presentations, the Associated Problems, Its Anesthetic Implications, and the Relevant
Investigations of Patients With Pituitary Tumor
Pituitary Tumor
Associated Problems and Its Anesthetic Implications
Investigations
GH-secreting pituitary
adenoma—acromegaly
1. Excess peripheral soft tissue deposition, nerve
entrapment syndrome. Enlargement of jaw, hand,
feet—difficult IV cannulation and arterial cannulation,
difficult mask ventilation
2. Macrognathia, macroglossia, expansion of upper
airway soft tissues, hypertrophy of aryepiglottic fold,
recurrent laryngeal nerve injury—Difficult airway
OSA: need for postoperative respiratory support
3. Hypertension, eccentric LVH, ischemic heart diseases,
arrhythmia, heart block, cardiomyopathy—perioperative
MI and heart failure
4. Diabetes: already discussed
5. Kyphoscoliosis and proximal myopathy—postoperative
respiratory support
Routine investigations:
CBC, basic metabolic panel (Na, K, creatinine,
AC, PC)
ECG, echo, chest X-ray, CT brain, MRI brain,
ophthalmic examination
Specific investigations
VI. PREANESTHETIC EVALUATION
1. GH >5 ng/mL
2. Elevated IGF-1
3. Failure to suppress the GH to <1 ng/mL
after administration of 75 g of oral glucose
4. Other endocrine workup—TFT, cortisol,
LH, FSH, or testosterone
PREOPERATIVE EVALUATION OF SPECIFIC NEUROSURGICAL CONDITIONS
221
TABLE 11.8 Showing the Common Presentations, the Associated Problems, Its Anesthetic Implications, and the Relevant
Investigations of Patients With Pituitary Tumor
Pituitary Tumor
Associated Problems and Its Anesthetic Implications
Investigations
ACTH-secreting tumour:
Cushing diseases
Truncal obesity, moon facies, buffalo hump,
supraclavicular pad of fat—difficult mask ventilation and
intubation
Routine investigations: same as mentioned
earlier
Specific investigations:
1. Elevated serum and urinary cortisol
2. Loss of diurnal variation of cortisol
3. Low-dose dexamethasone suppression
test (1 mg administered night before
sampling)—ACTH undetectable, adrenal
cause; ACTH 10–100 ng/mL, pituitary
cause; ACTH >200 ng/mL, ectopic origin
4. High-dose dexamethasone suppression test
(2 mg every 6 h for 48 h) suppresses both
serum and urinary cortisol
5. Inferior petrosal sinus sampling of ACTH
6. CRH stimulation test: increased ACTH
level will be seen in pituitary diseases
7. Other endocrine workup—TFT, GH, LH,
FSH, or testosterone
Friable skin and thin peripheral veins—difficult IV access,
extravasation of IVF
Osteoporotic joints, especially vertebral joints, vertebral
collapse—cervical spine fracture during laryngoscopy and
intubation. Fracture of spine during positioning for lumbar
drain
Retrobulbar fat deposit—Exophthalmos: corneal abrasion
and ulcer, proper eye covering is needed
Hypertension, eccentric LVH, ischemic heart diseases,
diastolic dysfunction—discussed earlier
Diabetes—discussed earlier proximal myopathy,
hypernatremia, hypokalemia, alkalosis—muscle weakness
TSH-secreting tumor
Very rare. Presents with features of hyperthyroidism—
tachycardia, hypertension, arrhythmia, loss of weight,
increased appetite, diarrhea, heat intolerance, mental
changes—need to be euthyroid before surgery, possibility of
intraoperative thyrotoxicosis crisis
Routine: as mentioned earlier
Specific investigations: TFT: TSH, T4, FTC with
other endocrine workup
Prolactin-secreting
tumours—prolactinoma
Women: galactorrhea, menstrual dysfunction; men: loss of
libido, erectile dysfunction—no anesthetic implications
Routine: same as mentioned earlier
Specific investigations:
Elevated serum prolactin level; >20 ng/
mL is considered elevated; 50–100 ng/mL,
microadenoma; >300 ng/mL, macroadenoma;
>150 ng/mL, pituitary stalk effect
Other endocrine workup
LH-, FSH-, and testosteronesecreting tumor
Very rare. Menstrual disturbance in woman, increased
sperm production in men, and precocious puberty in
children
Routine: as mentioned earlier
Specific: full endocrine workup
Hyposecretion syndrome
due to sudden bleeding into
the pituitary gland (pituitary
apoplexy) or compression of
normal gland by macroadenoma
Hypothyroidism: increased sensitivity, delayed
metabolism of anesthetic drugs, delayed emergence from
anesthesia, abnormal ventilatory response to hypoxia,
hypercapnia, bradycardia, postoperative ventilatory
support
Same as mentioned earlier
Adrenocortical insufficiency, but the rennin-angiotensinaldosterone axis is preserved—perioperative
hemodynamic instability. Need for perioperative steroid
cover
DI: severe dehydration—hemodynamic instability.
Hypernatremia and its associated problems
Nonfunctioning macroadenoma
Headache, visual field defect (bitemporal hemianopia),
prolactinemia (due to stalk effect), obstructive
hydrocephalus
Same as mentioned earlier
AC, ante cibum (before meal); ACTH, adrenocorticotrophic hormone; CBC, complete blood count; CRH, corticotropin-releasing hormone; CT, computed tomography; DI, diabetes insipidus; ECG, electrocardiogram; FSH, follicle-stimulating hormone; FTC, fractional thyroxine concentration; GH, growth hormone; IGF, insulinlike growth factor; IV, intravenous; LH, luteinising hormone; LVH, left ventricular hypertrophy; MI, myocardial infarction; MRI, magnetic resonance imaging; OSA,
obstructive sleep apnea; PC, post cibum (after meal); TFT, thyroid function test; TSH, thyroid stimulating hormone.
VI. PREANESTHETIC EVALUATION
222
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
Preoperative Evaluation of Patients With Epilepsy
Epilepsy is a common neurological disorder with a prevalence of 0.5–1% of the population. With the introduction
of newer drugs, there has been an improvement in pharmacotherapy for controlling epilepsy with less side effects.
About one-third of patients have persistent seizure, which are refractory to medical therapy and need surgery for
controlling the epilepsy. Before undergoing epilepsy surgery, all patients undergo extensive presurgical evaluation
to identify both the seizure foci and the functional areas of the brain near the seizure foci so that the seizure foci can
be excised without causing damage to the eloquent area, which improves the surgical and functional outcomes.
Anesthesiologists are involved in both, during the presurgical evaluation and in the anesthetic management of an
epilepsy surgery. Apart from routine history and physical examination these patients undergo neuropsychological
examination, followed by series of noninvasive and invasive testing (Table 11.9).60,61 These tests are done with or
without sedation. Studies have shown that the presence of an anesthesiologist during presurgical evaluation testing
will definitely improve the reliability and safety.60,62 After seizure foci localization, definitive surgery will be carried
out. Common surgical techniques performed are amygdalohippocampectomy, hemispherectomy, hemispherotomy,
corpus callosotomy, and multiple subpial transection and vagus nerve stimulation and deep brain stimulation of
hippocampus.
During the preoperative evaluation all the details of seizure, which include type of seizure (simple partial or
generalized tonic clonic seizure), frequency, presence or absence of aura, the time of occurrence of last episode, the
medication history, side effects of medication, need to be documented. Awareness of pharmacological properties of
antiepileptic drugs and their potential interaction with anesthetic drugs should be kept in mind63 (see medication
TABLE 11.9 Noninvasive and Invasive Tests Performed During the Presurgical Evaluation and the Anesthesiologist’s Role
Noninvasive Testing
Role in Epilepsy
Anesthesiologist Role
Video EEG
Commonly done noninvasive test for foci
localization. Patients are admitted and
continuous EEG recording will be done for
2–3 days
Not needed
CT scan
Identifies the gross structural lesion, e.g., tumor,
cavernoma
If the patient is uncooperative, the procedure will be
done under sedation
MRI
Identifies the structural lesion such as
hippocampal sclerosis, focal cortical dysplasia,
gliosis, low-grade tumors, and vascular
malformation
If the patients need GA, anesthesiologist will be
involved
SPECT
99mTc-labeled
Anesthesiologists are involved in pharmacoactivation
compounds are injected after the
seizure
Increased tracer uptake in the seizure foci due
to increased CBF. Scan can be taken up to 4 h of
tracer injection
Activation of focal epileptiform discharges by
pharmacological measures is called pharmacoactivation
Methohexital, Etomidate, Clonidine are used for this
purpose.
PET scan
The scan will be taken during the interictal
period. Neuronal uptake of glucose is measured
by injecting 18fluorodeoxygenase tracer. An area
of decreased tracer indicates the seizure foci
Same as mentioned earlier
MEG
MEG signals are derived from the intracellular
electrical current flowing from sulci and fissures
Patient may or may not need sedation
Grid, needle, depth, strip electrodes are placed
over the cortex
GA is administered for placing the electrode
through the burr hole or craniotomy. Patients will be
monitored for 2–7 days. While removing the electrode
pharmacoactivation is done using methohexitol,
etomidate, alfentanil, or sedation dose of propofol
Invasive Testing
EcoG
These electrodes are biologically inert, radioopaque, and MRI compatible such as platinum,
stainless steel, and silastic material
VI. PREANESTHETIC EVALUATION
PREOPERATIVE EVALUATION OF SPECIFIC NEUROSURGICAL CONDITIONS
223
TABLE 11.9 Noninvasive and Invasive Tests Performed During the Presurgical Evaluation and the Anesthesiologist’s Role
Noninvasive Testing
Role in Epilepsy
Anesthesiologist Role
Neuropsychological testing
Memory, language, motor skills, attention,
concentration, visual spatial skills, executive
abilities, and emotional function are tested
Not needed
fMRI
Used for identifying the eloquent cortex using
Not needed
the cerebral blood oxygenation level in the brain
while assessing the different task.
Language fluency, semantsc task: for Brocas area
Stress comprehension, story listening: for
Wernicke area
WADA test (intracarotid
amobarbital procedure)
Short-acting intravenous anaesthetics are
injected into one cerebral hemisphere to
anesthetize the particular side and then
the language and memory of contralateral
hemisphere will be tested using different
questionnaire to lateralize the language and
memory function
Functional Assessment Testing
In many centers anesthesiologist often involved for this
procedure
Traditionally sodium amobarbital was used. Because
of nonavailability of this drug various drugs such as
methohexitol, pentobarbital, secobarbital, propofol, and
etomidate are used to perform a WADA test
CBF; CT, computed tomography; ECoG, electrocorticography; EEG, electroencephalography; fMRI, functional MRI; GA, general anesthesia; MEG, magnetoencephalography; MRI, magnetic resonance imaging; PET, positron emission tomography; SPECT, single photon emission computed tomography.
evaluation section). Some patients with seizure disorder will have an associated genetic disorder such as tuberous
sclerosis (arrhythmia, cardiac tumor, renal and pulmonary dysfunction), Huntington chorea (abnormal response
to thiopentone), or von Recklinghausen disease (atlanto axial dislocation, airway compromise), which are associated with many anesthetic implications.62
Preoperative Evaluation of Patients With Subarachnoid Hemorrhage
SAH is a devastating clinical event with significant neurological morbidity and mortality (50%). Most SAHs are
due to rupture of an intracranial aneurysm (75–80%), About 20–25% are due to rupture of an arteriovenous malformation (AVM) or rupture of a blood vessel due to dural sinus thrombosis, vasculitis, or cerebral arterial dissection.
Preoperative evaluation and optimization depends on the cause. The three main predictors of morbidity and mortality after SAH are (1) advanced age, (2) impaired level of consciousness (higher grade, bad prognosis), and (3) volume
of blood in the CT imaging (more blood, bad prognosis).64–66
The anesthesiologist is involved at various stages while managing the patients with SAH: initial resuscitation
and stabilization, therapeutic management in the ICU, and providing anesthesia for aneurysm clipping or coiling.
Preanesthetic evaluation involves the following steps;
1. History and physical examination focusing on neurological examination and grading the SAH using the various
grading systems available. Commonly used grading are Hunt and Hess grading, Fisher grading, and World
Federation of Neurological Surgeons grading67–69 (details of grading are discussed in respective chapters in
detail).
2. Reviewing the intracranial pathology using CT of the brain, CT angiography, CT perfusion scan, and
digital subtraction angiography. The location, size, type of the aneurysm and extent of bleed, presence of
hydrocephalus, cerebral edema, and midline shift are assessed.
3. Evaluation of systemic functions focusing on the systems that are affected by the SAH (Table 11.10).
4. Reviewing the physiological and biochemical disturbances associated with SAH using the available
investigations and optimization of patient’s condition before surgery.
5. Since SAH is common in patients with hypertension, polycystic kidney disease, coarctation of aorta,
fibromuscular dysplasia, and chronic smoking, all these factors should be kept in mind during the preoperative
evaluation.
6. Communicate with the neurosurgeon regarding the complexity of aneurysm, hemodynamic goals, need
for temporary clipping, neurophysiological monitoring (somatosensory evoked potential) and the need for
VI. PREANESTHETIC EVALUATION
224
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
TABLE 11.10 Systemic Effects of SAH and Its Anesthetic Implications and the Relevant Investigations
System Involved
Systemic Effects and Its Anesthetic Implications
Investigations Needed
CNS
Raised ICP, cerebral edema, obstructive hydrocephalus
CT brain
Anesthetic implication: Raised ICP and its concerns, TIVA may
be better compared to inhalational anaesthetics
CT angiography, CT perfusion, or DSA to assess
the cause
Rate and rhythm disturbances in ECG, QTc prolongation,
myocardial injury, catecholamine surge induced Takatsubo
cardiomyopathy with severe LV dysfunction
ECG, echo, cardiac enzymes
Anesthetic implication: Perioperative hemodynamic instability,
MI, worsening of myocardial function
Patients with MI/hemodynamically unstable/
CHF/poor LV function (EF <30%) need
preoperative optimization before surgery
Pulmonary edema due to cardiac dysfunction
Chest X-ray, USG chest—Kerley B line
CVS
RS
Aspiration pneumonia, basal atelectasis in patients with low
GCS
Anesthetic implication: Perioperative hypoxia and hypercarbia
causes secondary neurological injury and increases POPCs
Renal
Acute renal failure: may be due to hypovolemia or contrast
induced
BUN, serum creatinine
Anesthetic implication: Fluid overload, prolonged drug effect
because of decreased renal elimination
Vascular
Intravascular volume depletion due to supine diuresis,
mannitol effect, negative nitrogen balance, iatrogenic blood
loss, and decreased erythropoiesis
CBC, basic electrolyte panel (sodium, potassium,
magnesium, calcium, glucose)
Electrolyte disturbances: hyponatremia due to SIADH, CSW.
Hypokalemia, hypocalcemia, and hypomagnesaemia
Coagulation parameters
Anesthetic implications: Severe hemodynamic instability, delayed
awakening, cardiac arrhythmia
BUN, blood urea nitrogen; CBC, complete blood count; CHF, congestive heart failure; CNS, central nervous system; CSW, cerebral salt wasting; CT, computed
tomography; CVS, cardiovascular system; DSA, digital subtraction angiography; ECG, electrocardiogram; EF, ejection fraction; GCS, Glasgow Coma Scale; ICP,
intracranial pressure; LV, left ventricular; MI, myocardial infarction; POPCs, perioperative pulmonary complications; RS, respiratory system; SAH, subarachnoid
haemorrhage; SIADH, syndrome of inappropriate antidiuretic hormone; TIVA, total intravenous anesthesia; USG, ultrasonography.
lumbar drain placement for brain relaxation, as well as indocyanine green injection to see the relationship of an
aneurysm to parent vessel and to confirm the complete obliteration after clipping.
7. In case of a complex aneurysm, the anesthesiologist has to be prepared to administer adenosine for producing
transient asystole to facilitate clipping.
Patients with high-grade SAH often present with vasospasm, obstructive hydrocephalus, impaired autoregulation, and cerebrovascular CO2 reactivity. The incidence of hypovolemia, hyponatremia, arrhythmia, myocardial
dysfunctions, and mortality are higher in these patients.70 SAH affects almost every organ system of the body, the
systemic effects, its anesthetic implication, and the relevant preoperative investigations are mentioned in Table 11.10.
Preoperative Evaluation of Patients With Arteriovenous Malformation
AVM is an abnormal (nidus) communication between small arteries and veins without intervening capillaries producing low resistance and high-flow shunt. Cerebral AVM is a congenital malformation involving the
cerebral vasculature; although it is congenital, it usually manifests between the second and fourth decades. It
commonly presents with signs and symptoms of intracranial bleed, seizure, focal neurological deficits, hydrocephalus, or rarely CHF (in large AVMs). Most AVMs are supratentorial in origin (70–97%); some are located
in infratentorial (3–30%) or deeper brain structures (5–18%). Anesthesiologists often deal with these patients
either for diagnostic investigation (angiography, MRI) or for therapeutic embolization and/or for excision/
stereotactic radiosurgery.
VI. PREANESTHETIC EVALUATION
REFERENCES
225
There are various studies looking at several factors for stratifying the surgical risk. Pasqualin et al.71 studied
several anatomic and hemodynamic factors such as (1) volume of AVM (>20 mL), (2) presence of deep feeding vessels and deep draining system, (3) shunt flow >120 cm/s and pulsatility index <0.5, using transcranial Doppler, (4)
involvement of eloquent area of brain, and (5) history of previous bleed, to stratify the surgical risk. Langer et al.72 had
suggested that the presence of hypertension and small-size and deep venous drainage are associated with increased
risk of hemorrhage. The Spetzler and Martin grading system73 uses three factors, namely, (1) size, (2) involvement of
eloquent area of the brain, and (3) pattern of venous drainage to stratify the risk. It is the most frequently used grading system to stratify the surgical risk.
The steps of preoperative evaluation are very similar to aneurysmal SAH: signs and symptoms of presenting
features, neurological status, and physical condition of the patient [congestive cardiac failure (CCF), pulmonary
edema, renal dysfunction]; history of transient ischemic attack or cerebrovascular accident, which are the relative
contraindications for giving controlled hypotension during embolization; history of allergy to protamine (protamine
insulin use, fish allergy, vasectomy) and contrast (general atopy/selfish allergy); as well as history of prior anticoagulation or coagulation disorders, which changes the coagulation management during embolization, all should be
documented. In female patients of reproductive age group, pregnancy test should be done. Some patients with large
AVM can have sacular aneurysms inside the nidus (incidence 3.7–8.7%); this should be ruled out before commencing
treatment for AVM. Anticonvulsant and antiedema measures should be continued throughout the surgery.
Embolization can cause redistribution of cerebral blood flow into adjacent regions and hyperemia of normal brain
leading to “normal perfusion pressure breakthrough” syndrome. Patients coming for surgery after embolization can
have raised ICP and new neurological insult, which need to be documented before surgery. Volume status has to
be assessed in view of contrast-induced diuresis causing hypovolemia. Large volume of contrast load can worsen
preexisting renal dysfunction. Since AVM is a high flow shunt and can cause torrential intraoperative bleeding, it
necessitates arrangement of adequate blood and blood products especially for a high-grade AVMs. For patients who
present with CCF or low GCS, elective ventilation should be planned to control the postoperative hemodynamic
status and to prevent cardiopulmonary complications.
References
1. Davies SJ. Preoperative optimization of the high-risk surgical patient. Br J Anaesth July 1, 2004;93(1):121–8.
2. Tote SP, Grounds RM. Performing perioperative optimization of the high-risk surgical patient. Br J Anaesth July 1, 2006;97(1):4–11.
3. Menke H, John KD, Klein A, Lorenz W, Junginger T. Preoperative risk assessment with the ASA classification. A prospective study of morbidity and mortality in
various ASA classes in 2,937 patients in general surgery. Chir Z Für Alle Geb Oper Medizen December 1992;63(12):1029–34.
4. Sankar A, Johnson SR, Beattie WS, Tait G, Wijeysundera DN. Reliability of the American Society of Anesthesiologists physical status scale in clinical practice. Br J
Anaesth April 2014:aeu100. http://dx.doi.org/10.1093/bja/aeu100.
5. Owens WD, Felts JA, Spitznagel EL. ASA physical status classifications: a study of consistency of ratings. Anesthesiology October 1978;49(4):239–43.
6. Lee TH, Marcantonio ER, Mangione CM, Thomas EJ, Polanczyk CA, Cook EF, et al. Derivation and prospective validation of a simple index for prediction of
cardiac risk of major noncardiac surgery. Circulation September 7, 1999;100(10):1043–9.
7. Ford MK, Beattie WS, Wijeysundera DN. Systematic review: prediction of perioperative cardiac complications and mortality by the revised cardiac risk index. Ann
Intern Med January 5, 2010;152(1):26–35.
8. Bilimoria KY, Liu Y, Paruch JL, Zhou L, Kmiecik TE, Ko CY, et al. Development and evaluation of the universal ACS NSQIP surgical risk calculator: a decision aid
and informed consent tool for patients and surgeons. J Am Coll Surg November 2013;217(5):833–42. e1–3.
9. Gupta PK, Gupta H, Sundaram A, Kaushik M, Fang X, Miller WJ, et al. Development and validation of a risk calculator for prediction of cardiac risk after surgery. Circulation July 26, 2011;124(4):381–7.
10. Hlatky MA, Boineau RE, Higginbotham MB, Lee KL, Mark DB, Califf RM, et al. A brief self-administered questionnaire to determine functional capacity (the Duke
Activity Status Index). Am J Cardiol September 15, 1989;64(10):651–4.
11. Orr JL, Williamson P, Anderson W, Ross R, McCafferty S, Fettes P. Cardiopulmonary exercise testing: arm crank vs cycle ergometry. Anaesthesia May 1,
2013;68(5):497–501.
12. Goff DC, Lloyd-Jones DM, Bennett G, O’Donnell CJ, Coady S, Robinson J. 2013 ACC/AHA guideline on the assessment of cardiovascular risk. [Internet] J Am Coll
Cardiol 2014. Available from: http://content.onlinejacc.org/article.aspx?articleid=1879711.
13. /data/revues/18752136/01010004/284/[Internet]. Indications for echocardiography in coronary risk stratification before non-cardiac surgery. June 24, 2008. Available
from: http://www.em-consulte.com/en/article/174401.
14. Das MK, Pellikka PA, Mahoney DW, Roger VL, Oh JK, McCully RB, et al. Assessment of cardiac risk before nonvascular surgery: dobutamine stress echocardiography in 530 patients. J Am Coll Cardiol May 2000;35(6):1647–53.
15. Lawrence VA, Hilsenbeck SG, Mulrow CD, Dhanda R, Sapp J, Page CP. Incidence and hospital stay for cardiac and pulmonary complications after abdominal
surgery. J Gen Intern Med December 1995;10(12):671–8.
16. Dimick JB, Chen SL, Taheri PA, Henderson WG, Khuri SF, Campbell DA. Hospital costs associated with surgical complications: a report from the private-sector
National Surgical Quality Improvement Program. J Am Coll Surg October 2004;199(4):531–7.
17. O’Donohue WJ. Postoperative pulmonary complications. When are preventive and therapeutic measures necessary? Postgrad Med February 15, 1992;91(3):167–70.
173–5.
18. Hall JC, Tarala RA, Hall JL, Mander J. A multivariate analysis of the risk of pulmonary complications after laparotomy. Chest April 1991;99(4):923–7.
19. Smetana GW, Lawrence VA, Cornell JE. American College of Physicians. Preoperative pulmonary risk stratification for noncardiothoracic surgery: systematic
review for the American College of Physicians. Ann Intern Med April 18, 2006;144(8):581–95.
20. McAlister FA, Bertsch K, Man J, Bradley J, Jacka M. Incidence of and risk factors for pulmonary complications after nonthoracic surgery. Am J Respir Crit Care Med
March 1, 2005;171(5):514–7.
VI. PREANESTHETIC EVALUATION
226
11. PREANESTHETIC EVALUATION OF NEUROSURGICAL PATIENTS
21. Canet J, Gallart L, Gomar C, Paluzie G, Vallès J, Castillo J, et al. Prediction of postoperative pulmonary complications in a population-based surgical cohort.
Anesthesiology December 2010;113(6):1338–50.
22. Mazo V, Sabaté S, Canet J, Gallart L, de Abreu MG, Belda J, et al. Prospective external validation of a predictive score for postoperative pulmonary complications.
Anesthesiology August 2014;121(2):219–31.
23. Arozullah AM, Khuri SF, Henderson WG, Daley J. Participants in the National Veterans Affairs Surgical Quality Improvement Program. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Ann Intern Med November 20, 2001;135(10):847–57.
24. Arozullah AM, Daley J, Henderson WG, Khuri SF. Multifactorial risk index for predicting postoperative respiratory failure in men after major noncardiac surgery.
Ann Surg August 2000;232(2):242–53.
25. Gupta H, Gupta PK, Schuller D, Fang X, Miller WJ, Modrykamien A, et al. Development and validation of a risk calculator for predicting postoperative pneumonia. Mayo Clin Proc November 2013;88(11):1241–9.
26. Gupta H, Gupta PK, Fang X, Miller WJ, Cemaj S, Forse RA, et al. Development and validation of a risk calculator predicting postoperative respiratory failure. Chest
November 2011;140(5):1207–15.
27. Archer C, Levy AR, McGregor M. Value of routine preoperative chest x-rays: a meta-analysis. Can J Anaesth November 1993;40(11):1022–7.
28. Qaseem A, Snow V, Fitterman N, Hornbake ER, Lawrence VA, Smetana GW, et al. Risk assessment for and strategies to reduce perioperative pulmonary complications for patients undergoing noncardiothoracic surgery: a guideline from the American College of Physicians. Ann Intern Med April 18, 2006;144(8):575–80.
29. Tisi GM. Preoperative evaluation of pulmonary function. Validity, indications, and benefits. Am Rev Respir Dis February 1979;119(2):293–310.
30. Warner DO. Perioperative abstinence from cigarettes: physiologic and clinical consequences. Anesthesiology February 2006;104(2):356–67.
31. Wong J, Lam DP, Abrishami A, Chan MTV, Chung F. Short-term preoperative smoking cessation and postoperative complications: a systematic review and metaanalysis. Can J Anaesth March 2012;59(3):268–79.
32. Woods BD, Sladen RN. Perioperative considerations for the patient with asthma and bronchospasm. Br J Anaesth December 2009;103(Suppl. 1):i57–65.
33. Silvanus M-T, Groeben H, Peters J. Corticosteroids and inhaled salbutamol in patients with reversible airway obstruction markedly decrease the incidence of
bronchospasm after tracheal intubation. Anesthesiology May 2004;100(5):1052–7.
34. Lawrence VA, Cornell JE, Smetana GW. American College of Physicians. Strategies to reduce postoperative pulmonary complications after noncardiothoracic
surgery: systematic review for the American College of Physicians. Ann Intern Med April 18, 2006;144(8):596–608.
35. Makhabah DN, Martino F, Ambrosino N. Peri-operative physiotherapy. Multidiscip Respir Med January 23, 2013;8(1):4.
36. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol May 1,
2013;177(9):1006–14.
37. Rahimi E, Mariappan R, Tharmaradinam S, Manninen P, Venkatraghavan L. Perioperative management and complications in patients with obstructive sleep
apnea undergoing transsphenoidal surgery: our institutional experience. J Anaesthesiol Clin Pharmacol 2014;30(3):351–4.
38. Malow BA, Levy K, Maturen K, Bowes R. Obstructive sleep apnea is common in medically refractory epilepsy patients. Neurology October 10, 2000;55(7):1002–7.
39. Pollak L, Shpirer I, Rabey JM, Klein C, Schiffer J. Polysomnography in patients with intracranial tumors before and after operation. Acta Neurol Scand January
2004;109(1):56–60.
40. Kaw R, Chung F, Pasupuleti V, Mehta J, Gay PC, Hernandez AV. Meta-analysis of the association between obstructive sleep apnoea and postoperative outcome.
Br J Anaesth December 2012;109(6):897–906.
41. Lockhart EM, Willingham MD, Abdallah AB, Helsten DL, Bedair BA, Thomas J, et al. Obstructive sleep apnea screening and postoperative mortality in a large
surgical cohort. Sleep Med May 2013;14(5):407–15.
42. Singh M, Liao P, Kobah S, Wijeysundera DN, Shapiro C, Chung F. Proportion of surgical patients with undiagnosed obstructive sleep apnoea. Br J Anaesth April
2013;110(4):629–36.
43. Abdelsattar ZM, Hendren S, Wong SL, Campbell DA, Ramachandran SK. The impact of untreated obstructive sleep apnea on cardiopulmonary complications in
general and vascular surgery: a cohort study. Sleep August 2015;38(8):1205–10.
44. Persson HE, Svanborg E. Sleep deprivation worsens obstructive sleep apnea. Comparison between diurnal and nocturnal polysomnography. Chest March
1996;109(3):645–50.
45. Chung F, Yegneswaran B, Liao P, Chung SA, Vairavanathan S, Islam S, et al. STOP questionnaire: a tool to screen patients for obstructive sleep apnea. Anesthesiology
May 2008;108(5):812–21.
46. Chung F, Subramanyam R, Liao P, Sasaki E, Shapiro C, Sun Y. High STOP-Bang score indicates a high probability of obstructive sleep apnoea. Br J Anaesth May
2012;108(5):768–75.
47. Chung F, Chau E, Yang Y, Liao P, Hall R, Mokhlesi B. Serum bicarbonate level improves specificity of STOP-Bang screening for obstructive sleep apnea. Chest May
2013;143(5):1284–93.
48. Flemons WW, Whitelaw WA, Brant R, Remmers JE. Likelihood ratios for a sleep apnea clinical prediction rule. Am J Respir Crit Care Med November 1994;150(5 Pt
1):1279–85.
49. Netzer NC, Stoohs RA, Netzer CM, Clark K, Strohl KP. Using the Berlin Questionnaire to identify patients at risk for the sleep apnea syndrome. Ann Intern Med
October 5, 1999;131(7):485–91.
50. Chau EHL, Lam D, Wong J, Mokhlesi B, Chung F. Obesity hypoventilation syndrome: a review of epidemiology, pathophysiology, and perioperative considerations. Anesthesiology July 2012;117(1):188–205.
51. Gross JB, Bachenberg KL, Benumof JL, Caplan RA, Connis RT, Coté CJ, et al. Practice guidelines for the perioperative management of patients with obstructive sleep apnea: a report by the American Society of Anesthesiologists Task Force on Perioperative Management of patients with obstructive sleep apnea.
Anesthesiology May 2006;104(5):1081–93. quiz 1117–8.
52. Porter KR, McCarthy BJ, Freels S, Kim Y, Davis FG. Prevalence estimates for primary brain tumors in the United States by age, gender, behavior, and histology.
Neuro-Oncol June 2010;12(6):520–7.
53. Nguyen HS, Sundaram SV, Mosier KM, Cohen-Gadol AA. A method to map the visual cortex during an awake craniotomy. J Neurosurg April 2011;114(4):922–6.
54. Taylor MD, Bernstein M. Awake craniotomy with brain mapping as the routine surgical approach to treating patients with supratentorial intraaxial tumors: a
prospective trial of 200 cases. J Neurosurg January 1999;90(1):35–41.
55. Chui J, Manninen P, Valiante T, Venkatraghavan L. The anesthetic considerations of intraoperative electrocorticography during epilepsy surgery. Anesth Analg
August 2013;117(2):479–86.
56. Serletis D, Bernstein M. Prospective study of awake craniotomy used routinely and nonselectively for supratentorial tumors. J Neurosurg July 2007;107(1):1–6.
57. Chui J. Anesthesia for awake craniotomy: an update. Rev Colomb Anestesiol Engl Ed January 1, 2015;43:22–8.
58. Burnand C, Sebastian J. Anaesthesia for awake craniotomy. Contin Educ Anaesth Crit Care Pain June 19, 2013. mkt024.
59. Klimek M, Verbrugge SJC, Roubos S, van der Most E, Vincent AJ, Klein J. Awake craniotomy for glioblastoma in a 9-year-old child. Anaesthesia June 1,
2004;59(6):607–9.
60. Chui J, Venkatraghavan L, Manninen P. Presurgical evaluation of patients with epilepsy: the role of the anesthesiologist. Anesth Analg April 2013;116(4):881–8.
61. Van Paesschen W, Dupont P, Sunaert S, Goffin K, Van Laere K. The use of SPECT and PET in routine clinical practice in epilepsy. Curr Opin Neurol April
2007;20(2):194–202.
62. Bhagat H, Dash HH. Anaesthesiologist’s role in the management of an epileptic patient. Indian J Anaesth 2006;50(1):20–2.
63. Perks A, Cheema S, Mohanraj R. Anaesthesia and epilepsy. Br J Anaesth April 1, 2012;108(4):562–71.
VI. PREANESTHETIC EVALUATION
REFERENCES
227
64. Ko S-B, Choi HA, Carpenter AM, Helbok R, Schmidt JM, Badjatia N, et al. Quantitative analysis of hemorrhage volume for predicting delayed cerebral ischemia
after subarachnoid hemorrhage. Stroke J Cereb Circ March 2011;42(3):669–74.
65. Rosengart AJ, Schultheiss KE, Tolentino J, Macdonald RL. Prognostic factors for outcome in patients with aneurysmal subarachnoid hemorrhage. Stroke J Cereb
Circ August 2007;38(8):2315–21.
66. de Oliveira Manoel AL, Goffi A, Marotta TR, Schweizer TA, Abrahamson S, Macdonald RL. The critical care management of poor-grade subarachnoid haemorrhage. [Internet] Crit Care 2015. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4724088/.
67. Hunt WE, Hess RM. Surgical risk as related to time of intervention in the repair of intracranial aneurysms. J Neurosurg January 1968;28(1):14–20.
68. Report of World Federation of Neurological Surgeons Committee on a Universal Subarachnoid Hemorrhage Grading Scale. J Neurosurg June 1988;68(6):985–6.
69. Fisher CM, Kistler JP, Davis JM. Relation of cerebral vasospasm to subarachnoid hemorrhage visualized by computerized tomographic scanning. Neurosurgery
January 1980;6(1):1–9.
70. Pong RP, Lam AM. Anaesthetic management of cerebral aneurysm surgery. In: Cottrell JE, Young WL, editors. Cottrell and Young’s neuroanesthesia. 5th ed.
Philadelphia: Elsevier Health Sciences; 2010. 481 pp.
71. Pasqualin A, Barone G, Cioffi F, Rosta L, Scienza R, Da Pian R. The relevance of anatomic and hemodynamic factors to a classification of cerebral arteriovenous
malformations. Neurosurgery March 1991;28(3):370–9.
72. Langer DJ, Lasner TM, Hurst RW, Flamm ES, Zager EL, King JT. Hypertension, small size, and deep venous drainage are associated with risk of hemorrhagic
presentation of cerebral arteriovenous malformations. Neurosurgery March 1998;42(3):481–6. Discussion 487–9.
73. Spetzler RF, Martin NA. A proposed grading system for arteriovenous malformations. J Neurosurg October 1986;65(4):476–83.
VI. PREANESTHETIC EVALUATION
This page intentionally left blank
S E C T I O N
V I I
NEUROSURGERY
This page intentionally left blank
C H A P T E R
12
Supratentorial Lesions
H. Bhagat, S. Mahajan
Postgraduate Institute of Medical Education and Research, Chandigarh, India
O U T L I N E
Introduction
231
Classification
232
Pathophysiology and Clinical Correlations
233
Clinical Features
235
Neuroimaging
235
Intraoperative Considerations: The Team Approach 236
Anesthetic Management
Emergency Management of Patients With
Supratentorial Tumors
Nonemergent Management of Patients With
Supratentorial Tumors
236
Intraoperative Management
Monitoring
Induction and Maintenance of Anesthesia
Hemodynamics
Preservation of Systemic Milieu
238
238
238
239
239
236
237
Intraoperative Medications
Intraoperative Surgical Field
239
239
Emergence From Anesthesia
240
Postoperative Management
Postcraniotomy Pain
Postoperative Seizures
Postcraniotomy Nausea and Vomiting
241
241
242
242
Awake Craniotomy
Indications
Techniques for Awake Craniotomy
Steps in the Conduct of Awake Craniotomy
Postoperative Care
Complications
242
243
243
243
244
245
Conclusions
245
Acknowledgment
245
References
245
INTRODUCTION
The supratentorial region accounts for major intracranial distribution in the intracranial compartment. The
supratentorial region is bound superiorly and laterally by the tense dura mater and skull. The inferior boundary consists of the anterior cranial fossa, middle cranial fossa, posterior cranial fossa, and tentorium cerebelli.
The supratentorial region is divided into the right and left halves by the falx cerebri. The contents include the
subarachnoid space containing cerebrospinal fluid and major vessels of the two cerebral hemispheres and lateral ventricles.
The supratentorial region consists of the part of the brain that lies above the tentorium cerebelli. It consists
of two cerebral hemisphere, ventricles and blood vessels. The two cerebral hemispheres are separated by falx
cerebri (Fig. 12.1). Each cerebral hemisphere consists of four lobes: frontal, parietal, temporal, and occipital.
Individual lobes have specific neurological functions. The supratentorial lesions may be associated with either
a compromise or loss of neurological functions depending on the site and size of the lesion.
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00012-9
231
© 2017 Elsevier Inc. All rights reserved.
232
12. SUPRATENTORIAL LESIONS
FIGURE 12.1 Sketch of the coronal section of supratentorial compartment.
CLASSIFICATION
The supratentorial lesions can be broadly classified as follows (Box 12.1).
Although supratentorial lesions can occur due to causes related to tumors, trauma, and infections, the present
chapter will focus on supratentorial tumors only.
BOX 12.1
C L A S S I F I C AT I O N O F S U P R AT E N T O R I A L L E S I O N S
Intra-Axial Lesions (Within Brain
Parenchyma)
1. Neoplasm
a. Primary
i. Glioblastoma multiforme
ii. Astrocytoma
iii. Primary central nervous system lymphoma
iv. Ganglioma
v. Oligodendroglioma
b. Metastasis
2. Infections
a. Abscess
b. Neurocysticercosis
3. Hemorrhage (intracerebral hemorrhage)
Extra-Axial Lesions (External to Brain
Parenchyma)
1. Neoplasm
a. Meningioma
b. Hemangiopericytoma
c. Pituitary tumors
2. Hemorrhage
a. Subarachnoid hemorrhage
b. Subdural hematoma
c. Extradural hematoma
3. Arachnoid cysts
Intraventricular Lesions
1. Neoplasms
a. Ventricular wall and septum pellucidum
i. Ependymoma
ii. Central neurocytoma
b. Choroid plexus
i. Choroid plexus papilloma/carcinoma
c. Others
i. Intraventricular meningioma
2. Colloid cysts
3. Infections
a. Hydatid cysts
b. Tuberculoma
c. Neurocysticercosis
VII. NEUROSURGERY
233
PATHOPHYSIOLOGY AND CLINICAL CORRELATIONS
PATHOPHYSIOLOGY AND CLINICAL CORRELATIONS
INTRACRANIAL PRESSURE
(mmHg)
The supratentorial region is well protected within the confines of the tough dura mater and skull. The pressure
within the right and left sides of the supratentorial compartment is in equilibrium as is the pressure within the supratentorial and infratentorial compartments. The supratentorial compartment follows the Monro–Kellie doctrine that
states that the sum total of the volume of the compartment at any point remains constant. This would translate into
a realization that the compensation for any expanding mass lesion would be limited and could lead to an increase in
intracranial pressure (ICP).
During the initial phase of a slow-growing mass lesion there would be compensation in terms of egress of cerebrospinal fluid and reduction of blood volume (phase I, Fig. 12.2). At this point of time surgery can be planned as an
elective procedure. As the mass lesion grows further, there comes a point whereby the compensatory mechanisms
are exhausted (phase II, Fig. 12.2). With further growth of mass lesion, there is increase in ICP. Discordance in ICP
between the two halves in the supratentorial compartment leads to shift of brain parenchyma from one half to the
other, which is referred to as subfalcine herniation or midline shift (phase III, Fig. 12.2). A midline shift of more than
5 mm is usually considered to be associated with significant mass effect (Fig. 12.3). At this point of time surgery is
80
70
60
50
40
30
20
10
IV
HERNIATION
SPATIAL
EXHAUSATION
III
SPATIAL
COMPENSATION
I
II
INTRACRANIAL VOLUME
FIGURE 12.2 Intracranial pressure–volume curve.
FIGURE 12.3 Computed tomograpgic scan showing significant midline shift due to a supratentorial tumor.
VII. NEUROSURGERY
234
12. SUPRATENTORIAL LESIONS
considered as an urgent procedure. The unabated growth of mass lesion may lead to differential pressure between
the supratentorial and infratentorial compartments. The transtentorial pressure gradient may lead to herniation of
the medial portion of the temporal lobe into the infratentorial compartment, called “uncal herniation/central herniation” (phase IV, Figs. 12.2 and 12.4). This leads to compression of the ipsilateral third cranial nerve, which clinically
manifest as anisocoria. Radiologically this is manifested as obliteration of basal cisterns. This is the time when surgical procedure for decompression of mass lesion is an absolute emergency.
The enlarging supratentorial mass in animal studies has been associated with loss of autoregulation in the
regions of brain ipsilateral and contralateral to the lesion. However, there is relative preservation of midbrain
autoregulation.1 The understanding of the effect of supratentorial tumors with regard to the integrity of blood–
brain barrier (BBB) is limited. The integrity of BBB as studied by contrast enhanced scanning will depend on the
degree of the malignancy of the supratentorial tumors.2 The effect on the BBB depends on the type and stage of
tumor development.3,4 BBB permeability is heterogeneous within the tumor site and depends on the type of tumor
and stage of tumor development. Low-grade gliomas usually have preservation of the BBB. It has been found that
BBB is intact in the initial steps of tumor development with significant increase in BBB permeability only during
the later stages as tumor mass increases. Low-grade gliomas and tumor in early stages are least likely to impact the
BBB.5 Nevertheless, even in later stages, BBB is still functional and limiting in terms of solute and drug permeability in and around the tumor.6 The regional blood cerebral flow can be affected ipsilateral as well as contralateral to
the supratentorial mass lesion.7 The blood flow to the midbrain is preserved till the point of total decompensation
and herniation.
Various animals studies have shown that the progressive expansion of a supratentorial epidural space gave rise to
a series of pathophysiological reactions.8,9 The vital physiological variables and sequential magnetic resonance (MR)
images were recorded simultaneously using intracranial expanding mass. When the expanding mass was increased
into 4–5% of the intracranial volume (ICV), there was a progressive slow rise in systemic arterial pressure, along
with changes in pulse and respiratory frequency. This volume is known as reaction volume. At a volume of 8–10% of
ICV, apnea (referred to as apneic volume) and an isoelectric electroencephalogram were observed. Following apnea
the systemic arterial pressure increased as part of the Cushing triad. The changes in vital physiological variables
progressed in a rostrocaudal direction. Reaction volume associated with a marked transtentorial pressure gradient and magnetic resonance imaging (MRI) changes were consistent with tentorial herniation. Respiratory arrest
(apneic arrest) was associated with occlusion of the cistern magna, consistent with some degree of foramen magnum
herniation.
Epileptogenesis associated with tumors can be due to changes in peritumoral microenvironment, alterations of
synaptic neurotransmitter release and reuptake and the exitotoxic effect of glutamate.10,11
FIGURE 12.4 Transtentorial herniation.
VII. NEUROSURGERY
NEUROIMAGING
235
CLINICAL FEATURES
The clinical features of the patients with supratentorial lesions will depend on the following three factors(S3): (1) site of
lesion (localizing signs), (2) size of lesion and mass effect (nonlocalizing signals), and (3) speed/rate of growth of lesion.
The clinical manifestations of a supratentorial lesion may be site specific and may produce localizing clinical manifestation. The lesion affecting the motor frontal cortex may be associated with contralateral hemiparesis. The tumors of speech
area will result in aphasia. Affection of visual pathway at a particular site will help in localizing features (e.g., bitemporal
hemianopia) in case of suprasellar lesion. Presence of a focal seizure can help in precise localization of a tumor.
Large tumors and associated mass effect may produce generalize signs and symptoms, which are referred as nonlocalizing clinical manifestations. Raised ICP is a feature of many supratentorial lesions. Raised ICP may manifest
as headache and/or nausea and vomiting. Papilledema may be a common accompaniment. There may be elevation
of blood pressure with increase or decrease in pulse rate. A slow pulse rate appears to be due to vagal excitation or a
response of carotid sinus to acute rise in blood pressure. The rise of blood pressure has been hypothesized to be due
to compensatory autoregulatory responses to decreased cerebral blood flow caused by the raised ICP.
An expanding mass lesion causes third nerve palsy due to compression by herniating temporal lobe at the edge
of the tentorium. Sixth nerve palsy can occur as a consequence of stretching of the abducens nerve due to rostrocaudal displacement of brain stem. Compression of foramen of Monro or third ventricle can result in hydrocephalus. A
supratentorial mass lesion may result in cognitive decline.
The speed of growth of the lesions determine the nature of onset and progress of the signs and symptoms. Slowgrowing tumors have an insidious onset of presentation and are gradually progressive. Rapidly expanding masses
have fast progression. Similarly, a bleed into the tumor may result in an acute presentation. A transtentorial herniation (TTH) is representative of an acute neurological deterioration. An acute neurological deterioration may manifest
as sudden loss of consciousness.
NEUROIMAGING
Neuroimaging is a routine diagnostic procedure in patients with suspected intracranial lesions. This helps in
precise localization as well as differentiating the type of lesion. The type of radiological imaging will depend
upon the clinical presentation and urgency for evaluation and management. Patients who have slow onset and
progression of neurological symptoms are more commonly subjected to MRI, which is more sensitive in the
detection and differentiation of intracranial lesions as compared to computed tomographic (CT) scans. Various
sequences of MRI can help in delineating the intracranial tumor and the associated mass effect (Fig. 12.5). MR
spectroscopy evaluates the chemical spectrum of the mass, and it helps in differentiating tumors from various
FIGURE 12.5 Magnetic resonance imaging sequences of brain (T1, T2, and FLAIR, from left to right; showing ill-defined supratentorial mass
in insular region with midline shift).
VII. NEUROSURGERY
236
12. SUPRATENTORIAL LESIONS
mass lesions like abscess. Patients who present with acute neurological deterioration are usually subjected to
CT scan as this procedure is faster and can help in emergency management of these groups of patients. Cerebral
angiography may define the arterial supply of the tumor and help in planning tumor resection. In case of highly
vascular tumor preoperative radiologically guided intravascular embolization helps in reducing the vascularity
and intraoperative bleeding. Angiography is also helpful in distinguishing arteriovascular malformations and
giant aneurysm, which may present as mass lesions.
It is important for the neuroanesthesiologist to have a working knowledge of the imaging scans so as to estimate
the mass effect and take appropriate intraoperative steps to optimize the brain conditions. In case of giant tumors it is
possible that despite all the physiological, pharmacological, and mechanical methods advocated by the anesthesiologist to reduce the ICP, the brain may still be bulging. It is important to interact with the surgeons prior to the surgery
regarding the possibility of refractory brain bulge and the need to drain the CSF if required. Imaging scans can also
help to triage patients who may otherwise need an urgent surgery. Positron emission tomographic (PET) scan gives
the amount of metabolic activity in the brain, thus differentiating the tumors from inflammatory lesions. PET scans
are promising in differentiating between recurrent tumors and treatment-induced changes.12 PET scan can be superior to MRI for follow-up of patients who have been treated surgically or by radiation for brain tumors.
INTRAOPERATIVE CONSIDERATIONS: THE TEAM APPROACH
The conduct of neurosurgery is a team work, which includes the anesthesiologist, surgeons, operating room technicians, nursing staff, and at time radiographers and neurophysiologists. There should be good understanding and
communication among the team members so as to conduct smooth anesthesia, perioperative management, optimal
patient positioning, and maximal safe tumor resection.
The surgery for intracranial lesions is challenging and can be associated with variable intraoperative patient dynamics.
Consequently it is important to be familiar not only with the disease process and presentation but also with the important surgical steps so as to anticipate complications and appropriately manage them. It is important in this regard for
the anesthesiologist to interact with the surgeon prior to surgery and discuss various concerns regarding the patient. An
understanding of the crucial surgical steps and anticipating blood loss keeps one vigilant and prepared to handle a critical
situation. The surgical site should always to be confirmed and should be kept free from interference of monitoring sensors/electrodes like those used for monitoring the depth of anesthesia. Similarly the endotracheal tubes, drug delivery
systems, intravenous fluids, various monitoring devices, and anesthesia equipment should be adequately secured and
should be placed in such a way that they do not affect the working in the operation theater. It is important to advocate
measures to facilitate maximal brain relaxation, which will enable the surgeons to conduct maximal tumor resection.
The anesthesiologist should brief the surgeons from time to time the hemodynamic changes during surgical stimulation so as to warn him against damage to vital brain structures. In situations in which intraoperative evoked
potential studies are conducted there needs to be good communication between the anesthesiologist, neurophysiologist, and the surgeons. The surgical approaches and craniotomy depend on the tumor location. Most of the surgeries anterior to coronal suture are conducted in supine position. Pterional and frontal approaches are the most
common surgical approaches. The lesion behind the coronal suture may require the patient to be positioned in the
semsitting or lateral decubitus position. The lesion in the pituitary gland and the sellar and suprasellar regions can
be approached through the transsphenoidal route. The nursing staff should ensure adequate sterility and absolute
cleanliness of the operating room environment. The operation theater technicians should be well trained, should be
alert, and should be ready with all the equipment required for the conduct of anesthesia and surgery.
ANESTHETIC MANAGEMENT
An anesthesiologist may encounter a patient with supratentorial lesions in three scenarios: (1) emergency medical and surgical management, which poses threat to the life of the patient and needs immediate action, (2) urgent
surgery, which does not pose immediate threat to the life of patient but has to be performed within 24–48 h, and (3)
elective surgery, which can be taken up at the convenience of the patient and availability of hospital resources.
Emergency Management of Patients With Supratentorial Tumors
The patients may present with acute neurological deterioration in the emergency room or in the neurosurgical
ward following fluid and electrolyte imbalance, bleed in the tumor bed, or have a herniation following a seizure/an
VII. NEUROSURGERY
ANESTHETIC MANAGEMENT
237
FIGURE 12.6 Schema of management of patient with acute neurological deterioration. CT, computed tomography.
expanding mass lesion. The patient may be unconscious with an abnormal breathing pattern, hemodynamic fluctuations, pupillary and localizing signs.
An AB-5C protocol would be a good option for the emergency management of patients with acute neurological
deterioration.
1. Secure the airway. The patients who suffer acute neurological deterioration and become unresponsive may have
an obstructed airway and are also at risk of aspiration. The consequent hypoxemia can lead to aggravation of
ongoing neurological insult. It is imperative to secure the airway with an endotracheal tube. The technique
to facilitate tracheal incubation is best judged by the onsite anesthesiologists depending on expertise and
experience. It is important to emphasize that in the setting of hypoxemia the airway management would take
precedence; it would be prudent to avoid further rise in ICP.
2. Breathe the patient. Correction of hypoxemia and hypercapnia as initial temporizing measures would minimize
the risk of any impending herniation.
3. Check for circulation. Any hemodynamic instability should be meticulously diagnosed and managed. Acute
rise in ICP is associated with systemic catecholamine surge and myocardial dysfunction.13 In the presence of
hemodynamic instability, raised central venous pressure (CVP) and electrocardiographic (ECG) changes should
arouse a suspicion of myocardial dysfunction.
4. Correct physiological factors (blood sugar, arterial blood gases, and serum electrolytes).
5. Cerebral resuscitation should be initiated if there are signs suggestive of raised ICP. A 20–30 degrees head-up
position would help in cerebral venous drainage and reducing ICP. Once the hemodynamic stability is
ensured, the use of hyperventilation, cerebral decongestant (mannitol, hypertonic saline), and intravenous
anesthetics (thiopentone/propofol) has been shown to be associated with reversal of raised ICP and TTH
in significant number of patients with supratentorial lesion with clinically defined TTH (decrease in level
of consciousness and pupillary dilation).14 This may help in extending the window for CT scanning and
adjunctive treatment.
6. Catheterize the patient as a full bladder can lead to raised ICP.
7. Shift the patient for CT scanning. If a patient has a lesion that needs surgical intervention, then the patient
should be taken for emergency surgery. If there is no indication for surgery then the patient should be medically
managed in a neuro–intensive care unit (Fig. 12.6).
Nonemergent Management of Patients With Supratentorial Tumors
Patients with supratentorial pathology can have dynamic intracranial as well as systemic physiology depending on the site, size, and speed of growth of the tumor. The anesthetic management of these patients should be
VII. NEUROSURGERY
238
12. SUPRATENTORIAL LESIONS
comprehensive, beginning at the preoperative phase and continuing in the postoperative period. The active and keen
participation of an anesthesiologist in perioperative management can be an important factor in the critical outcome
of neurosurgical patients. An anesthetic evaluation should entail detailed neurosurgical and systemic evaluation of
the patient. The anesthesiologist should develop good rapport with the patient and his family members.
The concept of conduct of a preanesthetic evaluation should be to have a comprehensive systemic evaluation and
the need for optimization of any existing medical illness. Following the assessment of risk we need to plan our perioperative anesthesia technique and discuss it with the patient and/or relatives of the patient. The possible complications as well as the need for invasive monitoring and postoperative mechanical ventilation must be discussed and
informed consent must be sought. The need to administer sedative premedication will depend on the surgery, site of
lesion, as well as the neurological and systemic illness of the patient. Similarly, the administration of antisialogogues
will have to be considered in patients who are operated in prone position.
INTRAOPERATIVE MANAGEMENT
The goals of anesthetic management are to maintain CPP, avoid secondary systemic insults (maintain the systemic
milieu of the body), provide a slack brain, institute neuroprotective strategies, and to facilitate early awakening.
A patient who is in the operating room should undergo assessment as regard to the status of hydration. The moistness of the tongue, color of urine, and heart rate can give some idea regarding the hydration. Neurological patients
with raised ICP are on diuretics, have high incidence of vomiting, and the concomitant reduced desire to drink orally
predisposes them to a dehydrated state. The consequence may be a falsely elevated hematocrit and postinduction
hemodynamic instability. It is important to rehydrate them prior to induction of anesthesia. The choice of fluids
for resuscitation and maintenance in neurosurgical patients is debatable. Normal saline has the advantage of being
mildly hyperosmolar but has the risk of hyperchloremic metabolic acidosis. Ringer lactate is a balanced salt/solution
with the disadvantage of being mildly hypoosmolar. It is important to avoid dextrose-containing fluids. A 2014 study
has indicated that intraoperative use of half strength (0.45%) normal saline in patients undergoing surgery for craniopharyngioma was associated with better outcome when compared with normal saline or 5% dextrose solution.15
Monitoring
Standard American Society of Anesthesiologists’ monitoring guidelines have to be followed in all patients undergoing craniotomy for supratentorial lesions. Besides, these patients should have an invasive arterial pressure monitoring for beat-to-beat monitoring of heart rate and blood pressure. This also allows for obtaining an arterial blood
sample for blood gas analysis, electrolyte, and hematocrit estimation. The need for central venous cannulation and
CVP monitoring will be dictated by the nature of the lesion, patient position during expected blood loss and hemodynamic perturbations, and the need for postoperative fluid and hemodynamic management. The patient being
operated in sitting or semisitting position for occipital lesions should be monitored for venous air embolism using a
precordial Doppler or transesophageal echocardiography.
Induction and Maintenance of Anesthesia
Amnesia, analgesia, and muscle relaxants are the cornerstones of anesthesia management. However, it is important to understand its effect on systemic and cerebral physiology and consequently on the cerebral perfusion. An
ideal anesthetic agent could be the one that should maintain systemic hemodynamics, maintain cerebral blood flow
(CBF), and cause reduction of cerebral metabolic requirement of oxygen (CMRO2) and ICP. The effect of the various
anesthetic agents on hemodynamics, CBF, CMRO2, and ICP is described in the chapter relating to pharmacological
effects of anesthetics.
The induction of anesthesia in neurosurgical patients is usually a coinduction with intravenous anesthetic agents and
opioids. The popular choice for anesthetic induction is propofol and thiopentone sodium. Etomidate has the advantage of providing hemodynamic stability but can result in adrenal suppression. Ketamine is best avoided as it has the
potential to cause increase in ICP and postoperative hallucination and delirium. The commonly used muscle relaxants
to facilitate intubation are rocuronium and vecuronium. Rocuronium enables faster intubating conditions when compared to vecuronium. Succinylcholine has the potential to cause/increase in ICP and should preferably be avoided.
The commonly used opioids for neurosurgical procedures are fentanyl and morphine. The effect of opioids for
intraoperative analgesia as well on cerebral physiology appears to be similar. Opioids have minimal effect on cerebral
VII. NEUROSURGERY
INTRAOPERATIVE MANAGEMENT
239
blood flow, metabolism, and ICP, provided that normocapnia and normotension are maintained. The choice of agent
for the maintenance of anesthesia is either propofol or an inhalational agent. A 2015 study comparing propofol, sevoflurane, and desflurane along with nitrous oxide found all the three agents similar and acceptable for the conduct of
anesthesia in patients undergoing craniotomy for supratentorial tumors.16
Hemodynamics
The brain tissue meets its metabolic need and substance on cerebral perfusion, which depends on mean arterial
blood pressure. Consequently, hypotension is an important factor to be avoided at all costs. What should be an optimal mean arterial pressure (MAP) will vary individually depending on the lesion volume, site, and mass effect as
well as the status of the autoregulation. The cerebral perfusion pressure is very dynamic depending on the MAP and
ICP. Avoidance of intraoperative hypertension is also important as it can increase the cerebral blood volume (CBV)
beyond the autoregulation capacity and increased ICP. The two important time points when hypertension occurs
are during skull pin insertion of head holders and during periosteum elevation. Increasing the depth of anesthesia
along with supplementary agents like intravenous opioids, β-blockers, and lignocaine has shown reduction in the
hypertensive response to noxious stimulus.
Preservation of Systemic Milieu
Paramount importance has to be given to maintaining normal systemic physiology. The perilesional area has compromised perfusion due to mass effect and raised ICP. Hence systemic hypoxemia can lead to impaired neuronal
oxygenation and death. Ventilation should be adjusted to maintain normocapnia. Hypercapnia leads to cerebral vasodilation and increase in cerebral blood volume and further rise in ICP. Glycemic control needs to be advocated in the
modest range of 110–160 mg/dL as hyperglycemia predisposes to anaerobic metabolism and normal acidosis, which
has deleterious effects on the neurological outcome of the patients.17 The hematocrit values have to be optimally balanced, and anemia can jeopardize oxygen delivery and preservation of neurons in the penumbral area around the
lesion. Preoperative iron supplementation may be considered to correct preoperative anemia.18 The need for intraoperative blood transfusion should take into account the balance of expected blood loss and allowable blood loss. The
transfusion trigger will have to be individualized based on the physiological end points of oxygen delivery and oxygen
requirements: base deficit, bicarbonate ions, and pH, as well as clinical end points, i.e., hemodynamics. The intraoperative hematocrit estimation may not be reliable due to either hemoconcentration due to dehydration or hemodilution
resulting from overzealous fluid administration. Normothermia should be maintained and hyperthermia has to be corrected as it predisposes to increase in CMRO2. This can be deleterious in the face of raised ICP and reduced neuronal
perfusion.19
Intraoperative Medications
There are different types of practices with regard to the use of intraoperative medications. The antibiotic prophylaxis is administered following inductions in patients who have not received them preoperatively. Use of diuretics
has been the conventional method of reduction of ICP. 20% Mannitol in the dose of 0.25–1 g/kg has been seen to be
associated with reduction of ICP. Coadministration of loop diuretics like furosemide have been supported by few
studies in being additives in reduction of ICP, although the clinical reduction of ICP has not been convincing to
strongly advocate its use.20 Steroids have been used to reduce the perilesional interstitial edema associated with the
tumor. It reduces the leakage of fluids through the neocapillaries that form around tumors. They are usually started
once the patient has been diagnosed to have a tumor and are continued in the perioperative period. Seizure is a common symptom in patients who have supratentorial lesion. The lesions in and around the cortex have increased seizure potential and are usually on anticonvulsant medications. Phenytoin sodium is commonly used in these patients.
Patients who do not tolerate phenytoin receive either phenobarbitone or sodium valproate. Emerging drug for seizure prophylaxis is second-generation antiepileptics (levetiracetam).21 The administration of these medications is
popularly remembered in our institution as MAAD (mannitol, antibiotic, antiepileptic, dexamethasone) regime.
Intraoperative Surgical Field
One of the important goals of anesthetic management is to provide a slack surgical field so as to allow the neurosurgeon to have maximal exposure to the lesion into minimal brain retraction and perilesional neuronal injury. It
VII. NEUROSURGERY
240
12. SUPRATENTORIAL LESIONS
FIGURE 12.7 Intraoperative brain bulge.
is also important to understand the fact that the role of the anesthesiologist in controlling the brain bulge depends
on CBF management. The arterial blood flow accounts for 15% of CBV, while venous accounts for 40% of CBV.
Management of intraoperative brain bulge should encompass manipulation of cerebral arterial and venous blood
flow. The venous outflow can be improved by head-up position. A modest 15–20 degrees head-up position helps
in cerebral venous outflow and reduction of ICP. It is also important to avoid extreme head rotation, which may
impede cerebral venous outflow resulting in raised ICP. Manipulation of arterial blood flow by various methods like
modification of anesthesia technique and hyperventilation should be instituted. Consequently the anesthesiologist
can achieve about 55% reduction of CBV and ICP, which may be significant in numbers but actually may not achieve
clinical translation when the lesions are large with significant midline shifts and mass effects. In such cases when the
craniotomy is achieved, the tense brain follows the pathway of least resistance leading to transcalvarial herniation
referred to as “brain bulge” (Fig. 12.7). One of the important causes of intraoperative bulge can be surgical bleeding.
In such cases the surgeon needs to control bleeding and evacuate hematoma to achieve sufficient brain relaxation.
Suggested stepwise management for intraoperative brain bulge:
1. Head-up position
2. Hyperventilation
3. Repeat a bolus of cerebral decongestants: mannitol
4. Switch it over to intravenous anesthetic agents
5. Ventriculostomy and cerebrospinal fluid drainage
EMERGENCE FROM ANESTHESIA
Emergence from anesthesia following craniotomy closure is an important aspect that needs to carried out with
proper planning and skills. The goals of emergence in neurosurgical patients are twofold: an early awakening and
good quality of awakening. An awake patient is the best neurological monitor. Early awakening from anesthesia
allows to conduct a neurological examination and detect a postoperative complication like postoperative hematoma. Early reintervention in management of prospective complication will result in better neurological outcome
of patients. However, fast-track awakening from anesthesia has its own pitfalls like emergence hypertension, which
can lead to postoperative hematoma formation. The anesthesia technique that can lead to early awakening from
anesthesia have been addressed from time to time. Use of propofol, sevoflurane, and desflurane along with nitrous
oxide did not show any difference in the awakening times,16 although a few studies have demonstrated a benefit in
awakening times with use of desflurane and sevoflurane.22
It would be prudent to allow 15 min for awakening following cessation of anesthesia, considered as “early awakening.” Early examination and detection with radiological confirmation will allow to do a reintervention. Most of the
commonly used anesthetics have demonstrated acceptable emergence times, and there appears no clinically significant difference with the use of either propofol, sevoflurane, or desflurane.16
One of the important beliefs in neuroanesthesia practice is that use of morphine may be associated with delayed
emergence when compared to fentanyl. A study compared the effect of morphine to that of fentanyl in patients
VII. NEUROSURGERY
POSTOPERATIVE MANAGEMENT
241
undergoing surgery for supratententorial tumors in two different tertiary care centers of India. No difference was
noted in the emergence times.23
Fast-track awakening from anesthesia has its own drawbacks. Emergence hypertension is a common phenomenon
associated with early awakening, which can predispose to intracranial hematoma formation. Various pharmacological agents like lignocaine, esmolol, nicardipine, etc., have been used to manage emergence hypertension. One clinical
trial tried to address the issue of facilitating early emergence along with limiting the consequence hypertension. They
conclude that low dose of fentanyl along with nitrous oxide during craniotomy closure was effective in facilitating
early emergence along with control of emergence hypertension.24
Coughing during emergence from anesthesia is an unpleasant experience for the patients. It leads to sympathetic
stimulation and increase in blood pressure as well as metabolic requirement of the body. It increases the intrathoracic
pressure and cerebral venous pressure leading to the disruption of venous hemostasis and intracranial hematoma
formation. The effect of intravenous agents as well as inhalational agents appears to be similar on the incidence of
coughing during emergence. Use of lignocaine in the pilot balloon of the endotracheal tube has been found to be
associated with reduced incidence of emergence coughing.25
Emergence agitation is another undesirable response following neurosurgery. An agitated patient will not allow
for an adequate neurological examination. Moreover, it may be difficult to ascertain the cause due to anesthetics or
neurosurgical factors. However, the incidence of emergence agitation has been found to be similar with use of propofol, sevoflurane, and desflurane.16
POSTOPERATIVE MANAGEMENT
The management of a patient following supratentorial craniotomy should be carried out in a specialized neuro–
intensive care unit. These patients require continuous neurological and systematic physiological monitoring. Any
neurological deterioration can have systemic manifestation, while a compromise in systemic condition like hypertension, hypoxemia, hyperthermia, and hyperglycemia can adversely affect the neurological outcome of the patients.
Constant vigilance with immediate detection of any postoperative complications is the key to good outcome in these
groups of patients. Meticulous care has to be taken to maintain fluid and electrolyte balance in the patients along
with nutritional supplementation. Patients who are mechanically ventilated in the postoperative period should be
adequately sedated so as to allow them to tolerate the endotracheal tube and ventilatory support. The sedative agent
used should be easily titrable with fast onset and offset so as to allow for adequate neurological assessment. Seizure
prophylaxis, corticosteroids, and cerebral decongestants should be continued in the postoperative period.
Postcraniotomy Pain
Postcraniotomy pain is an important aspect of postoperative management and needs to be dealt with utmost care.
The incidence is around 60%, with the highest incidence observed 12 h after surgery, the majority being moderate in
nature and intensity.26 Patients of our institute experience mild pain within 48 h of craniotomy (Figs. 12.8 and 12.9).
FIGURE 12.8 Incidence of postcraniotomy pain (PGIMER Chandigarh data, 2009). Figures represent percentage of patients.
VII. NEUROSURGERY
242
12. SUPRATENTORIAL LESIONS
FIGURE 12.9 Incidence and duration of postcraniotomy pain (PGIMER Chandigarh data, 2009). Figures represent percentage of patients.
The management can be pre-emptive or based on symptoms. Scalp block administrated prior to craniotomy has
been found to reduce intraoperative analgesic requirement as well as delay the onset of postcraniotomy pain. Use
of bupivacaine or ropivacaine has been found to be effective. Paracetamol alone is not effective. Combination of
paracetamol with other analgesic medication may be effective. Opioids have been commonly advocated with oral
codeine being the most common. Patient-controlled analgesia with fentanyl, remifentanil, and morphine has been
found to be useful in ameliorating postcraniotomy pain. It is common belief that use of nonsteroidal antiinflammatory drugs in patients with postcraniotomy can lead to intracranial hematoma formation due to its effect on platelet
function. This is yet to be proven. Our personal experience is to use diclofenac 2 mg/kg as intravenous dose during
craniotomy closure, which is effective in reducing postoperative pain.
Postoperative Seizures
The occurrence of seizures following craniotomy has been found to be present in 15–20% patients.27 Postoperative
seizure has the potential to increase CMRO2 and CBF leading to raised ICP and neurological deterioration. In case
of inability to maintain the airway or an unconscious patient, there will be a need to secure the airway and maintain
oxygenation. Single focal seizures can be managed with reassurance. However, generalized tonic-clonic seizure need
to be managed with anticonvulsant drug. The cause could be postoperative edema or hematoma or pneumocephalus
mass effect. Significant pneumocephalus or hematoma will require evacuation, and presence of edema will need antiedema measures. Beside this proper oxygenation and maintenance of hemodynamics are also important. If the patient
is already on antiepileptic drugs, the dose should be increased after repeating loading dose or newer antiepileptic
agent is added to the current medications. If it is new-onset seizure, then loading dose of antiepileptics followed by
maintenance dose should be started. Antiepileptics that are effective include phenytoin and levetiracetam. Sodium
valproate and phenobarbital have not proven beneficial in preventing postoperative seizures. Moreover, these drugs
are also associated with significant side effects such as hepatotoxicity and hematological abnormalities.28,29
Postcraniotomy Nausea and Vomiting
Postcraniotomy nausea and vomiting (PONV) is one of the most important preventable complication. Its incidence is up to 45–70% without treatment.30 Vomiting and retching cause increase in intra-abdominal intrathoracic pressure, which leads to raised ICP. In addition, it increases chances of pulmonary aspiration and cause
fluid electrolyte imbalance. All these factors deteriorate patient neurological status. Antiemetics are the mainstay
of prophylaxis against PONV and 5-HT3 antagonists are considered as agent of choice. 5-HT3 antagonists have
acceptable side effects such as sedation, no extrapyramidal reactions, and no drug interactions. Dexamethasone
routinely administered in neurosurgical patients for reduction of peritumoral edema increases the antiemetic efficacy of 5-HT3 antagonists.
AWAKE CRANIOTOMY
Awake craniotomy refers to conduct of intracranial tumor surgery under conscious sedation so as to enable
clinical neuromonitoring, which allows the neurosurgeon to conduct tumor resection with no or minimal neurological deficit. The conduct of awake craniotomy must ensure the patients’ comfort, which in turn help in seeking
VII. NEUROSURGERY
AWAKE CRANIOTOMY
243
patient cooperation. Awake craniotomy has been traditionally used from ancient times and is a challenge for the
anesthesiologist. With the advent of new monitoring gadgets and availability of short-acting drugs, the conduct
of awake craniotomy has become comfortable for the anesthesiologist and a pleasant experience for the patient.
Indications
1. Supratentorial tumors close to eloquent areas like motor strips and speech area.
2. Epilepsy surgery.
Techniques for Awake Craniotomy
There are conventionally three techniques for the conduct of awake craniotomy. Although there are no clear-cut
guidelines for preference of each over the other, but the clinical experience and patients clinical profile will help the
anesthesiologist in identifying the technique that suits the patient.
Awake technique: This particular technique refers to conduct of entire intracranial surgery under conscious
sedation and monitoring. The process of conscious sedation is initiated, and scalp block is administered. The
skin, periosteal layer of cranium, and the dura matter are infiltrated by local anesthetics as the surgeon conduct
surgery.
Our experience indicates that this technique is good for patients who have some degree of cognitive impairment
due to neurological condition, alcoholism, and drug withdrawal. Similarly, patients with alcoholic liver disease may
fair better in this technique as awakening after general anesthesia (GA) may have residual anesthetic effects and
patients may not be cooperative. There are few limitation with awake techniques. The patients may perceive pain
during periosteal elevation and dural incision despite optimal-level sedation and presumed analgesia.
Sleep–awake technique: This technique encompasses the use of GA with a supraglottic airway device (SAD) till
the raising of dural flap. Muscle relaxants are preferably avoided. Subsequently, the GA is switched of and SAD
removed. The tumor is resected with patients under conscious sedation to enable neurological examination. This
technique is comfortable for the patients as the painful part of the procedures like periosteal elevation and dural incision are not perceived by the patient. However, some patients may experience pain during dural and skin closure.
This can be limited by increasing the depth of sedation and adequate supplementation of analgesia. Care must be
taken to ensure adequate awakening in obese patients and those with history suggestive of obstructive sleep apnea
(OSA) before removal of SAD following GA.
Sleep–awake–sleep technique: The initial phase is similar as described under sleep–awake technique. Following
tumor resection and hemostasis the patients are readministered GA and airway is secured with a SAD. This procedure mitigates any chances of pain during craniotomy closure. The issue that needs to be addressed is resecuring of
airway during craniotomy closure. It has to be preoperatively ensured that the patient has adequate mouth opening and there should be adequate access of the airway. Use of proseal laryngeal mask airway, which comes with an
inserter, is usually handy in resecuring the airway.
Steps in the Conduct of Awake Craniotomy
Once the patient has been identified for awake craniotomy, the patient should be counseled extensively by the
surgeon and the anesthesiologist. The surgeon should explain the need and advantage of this surgery under awake
condition. The anesthesiologist should first develop the support with the patient by introducing himself and having a general discussion. Then the patient should be explained about the various steps in the conduct of awake
craniotomy assured from time to time that he will be kept comfortable in the entire procedure. It has to be emphasized to the patients with due care the need to remain calm and cooperative during the procedure. They have to
be assured that should any pain discomfort or complication arise, it will be adequately taken care of. The patients
should be subsequently explained about the various techniques of conduct craniotomy and the neurological and
psychometric testing to be conducted interoperatively. After proper explanation, the patients should be allowed
time to understand and have their queries answered. Subsequent visits would boost patient’s confidence, and this
is one important factor for the success of awake craniotomy The preanesthetic evaluation should specifically take
into consideration airway and mouth opening. History should be sought regarding snoring, seizures, and motion
sickness. Preoperative cognitive function assessment has to be done to assess the anesthesia technique to be adopted
for awake craniotomy, and the neurological and cognitive function for motor power, speech, and vision has to be
explained and rehearsed in the preoperative period.
VII. NEUROSURGERY
244
12. SUPRATENTORIAL LESIONS
The operation theater has to be specifically prepared for the smooth conduct of awake craniotomy. Special arrangements have to made to allow visualization and access to patients during conduct of surgery (Fig. 12.10). The environment should be calm, and the patient should feeling relaxed (Fig. 12.11). Premedication includes antianxiety and
antiemetic measures.
Standard monitoring like ECG, peripheral capillary oxygen saturation, blood pressure, and capnography should be
done. Invasive arterial blood pressure in the limbs, which may not require extensive neurological examination, may
be considered as use of non-invasive blood pressure may be a source of discomfort due to repeated inflation. During
conscious sedation, the capnography can be monitored by placing the sampling line near to one of the nostrils of the
patient; monitoring of the depth of anesthesia would allow for easy titrability of anesthetics and sedative agents.
The intravenous cannula should be placed in the limbs, preferably not in the primary limb to be examined. Patient
should be covered with a warming blanket. Scalp block should be given to all patients (Fig. 12.12). Neurological
examination should be carried out prior to and time to time during tumor resection. Any alteration in neurological
and cognitive function should be promptly informed to the surgeon. The patient has to be emotionally supported
and assured about the surgery. Any discomfort has to be promptly attended.
Postoperative Care
The standard of postoperative care should be the same as is with any patient undergoing neurosurgical procedures. Beside it is important to make a postoperative visit to the patient to understand the experience of the patient
and make necessary modifications related to anesthetic management.
FIGURE 12.10 Arrangement of operation theater for awake craniotomy.
FIGURE 12.11 Canopy for easy accessibility of patient for communication.
VII. NEUROSURGERY
REFERENCES
245
FIGURE 12.12 Nerves for scalp block.
Complications
1. Oversedation can occur, which may result in lack of patient cooperation for conduct of intraoperative
neurological examination. Use of bispectral index or entropy monitors may help in titrating the sedative drugs
and avoid oversedation. Oversedation can also lead to tongue fall and obstruction of airway especially in
patients who are obese and those who have history suggestive of OSA.
2. Undersedation can lead to patients’ anxiety, restlessness, and movements, which can affect the conduct of surgery. A
calculated bolus dose and stepping up of infusion of seductive drugs can help in providing comfort to the patients.
3. Pain may be perceived during various stages of awake craniotomy. Reassurance and use of bolus dose of shortacting opioids can help in ameliorating pain during procedure.
4. Nausea and vomiting can occur during the procedure. Antiemetic prophylaxis can be effecting in reducing the
incidence of intraoperative nausea and vomiting.
5. Seizures can occur intraoperatively, which can jeopardize the procedure. In our experience seizure prophylaxis
has shown to be effective in preventing the intraoperative convulsions. Nevertheless, if seizures occur, the first
concern should be to ensure adequate airway and breathing. Simultaneously the surgeon should instill ice cold
saline on the brain surface. Intravenous midazolam can help in limiting the seizure episode. If the patient is
comfortable and cooperative after the seizure episode, the surgery can proceed under conscious sedation. If not,
then the airway will need to be secured and the surgery has to be conducted under GA.
6. Patient discomfort like dry mouth, itching around the face, heaviness of head, etc., has to be adequately addressed.
CONCLUSIONS
Supratentorial lesions account for the major bulk of case load at neurosurgical centers. The anesthesiologists who
are involved in the management of neurosurgical patients should have working knowledge of anatomy and radiology related to supratentorial lesions. The anesthesiologist should develop clinical skills in the assessment of patients
with supratentorial lesion and accord them due care with respect to the changes in cerebral and systemic physiology.
Active participation of an anesthesiologist in the perioperative management of patients is important in the critical
outcome of these groups of patients.
Acknowledgment
We are thankful to Mr. Munish Kumar, Junior Research Fellow in the Department of Anaesthesia and Intensive Care, for the sketches of Figs 12.1,
12.2, and 12.4.
References
1. Smith DR, Jacobson J, Kobrine AI, Rizzoli HV. Regional cerebral blood flow with intracranial mass lesions. Part II: autoregulation in localized
mass lesion. Surg Neurol 1977;7:238–40.
2. Butler AR, Passalaqua AM, Berenstein A, Kricheff II . Contrast enhanced CT scan and radionuclide brain scan in supratentorial gliomas. Am J
Roentgenol 1979;132:607–11.
VII. NEUROSURGERY
246
12. SUPRATENTORIAL LESIONS
3. Wiranowska M, Gonzalvo AA, Saporta S, Gonzalez OR, Prockop LD. Evaluation of blood–brain barrier permeability and the effect of interferon in mouse glioma model. J Neurooncol 1992;14:225–36.
4. Fidler IJ, Yano S, Zhang RD, Fujimaki T, Bucana CD. The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol
2002;3:53–7.
5. Finizio FS. CT and MRI aspects of supratentorial hemispheric tumors of childhood and adolescence. Childs Nerv Syst 1995;11:559–67.
6. On NH, Mitchell R, Savant SD, Bachmeier CJ, Hatch GM, Miller DW. Examination of blood–brain barrier (BBB) integrity in a mouse brain
tumor model. J Neurooncol 2013;111:133–43.
7. Smith DR, Jacobson J, Kobrine AI, Rizzoli HV. Regional cerebral blood flow with intra cranial mass lesions. Part I: local alterations in cerebral
blood flow. Surg Neurol 1977;7:233–40.
8. Zwetnow NN, Schrader H, Löfgren J. Effects of continuously expanding intracranial lesions on vital physiological parameters. An experimental animal study. Acta Neurochir (Wien) 1986;80:47–56.
9. Thuomas KA, Vlajkovic S, Ganz JC, Nilsson P, Bergström K, Pontén U, et al. Progressive brain compression, changes in vital physiological variables, correlated with brain tissue water content and brain tissue displacement, experimental MR imaging in dogs. Acta Radiol 1993;34:289–95.
10. Liubinas SV, O’Brien TJ, Moffat BM, Drummond KJ, Morokoff AP, Kaye AH. Tumour associated epilepsy and glutamate excitotoxicity in
patients with gliomas. J Clin Neurosci 2014;21:899–908.
11. Buckingham SC, Campbell SL, Haas BR, Montana V, Robel S, Ogunrinu T, et al. Glutamate release by primary brain tumors induces epileptic
activity. Nat Med 2011;17:1269–74.
12. Chen W. Clinical applications of PET in brain tumors. J Nucl Med 2007;48:1468–81.
13. Kolin A, Norris JW. Myocardial damage from acute cerebral lesions. Stroke 1984;15:990–3.
14. Qureshi AI, Geocadin RG, Suarez JI, Ulatowski JA. Long-term outcome after medical reversal of transtentorial herniation in patients with
supratentorial mass lesions. Crit Care Med 2000;28:1556–64.
15. Mukherjee KK, Dutta P, Singh A, Gupta P, Srinivasan A, Bhagat H. Choice of fluid therapy in patients of craniopharyngioma in the perioperative period: a hospital-based preliminary study. Surg Neurol Int 2014;5:105.
16. Bastola P, Bhagat H, Wig J. Comparative evaluation of propofol, sevoflurane and desflurane for neuroanaesthesia: a prospective randomised
study in patients undergoing elective supratentorial craniotomy. Indian J Anaesth 2015;59:287–94.
17. Sieber F, Smith DS, Kupferberg J, Crosby L, Uzzell B, Buzby G, et al. Effects of intraoperative glucose on protein catabolism and plasma glucose levels in patients with supratentorial tumors. Anesthesiology 1986;64:453–9.
18. Kozek-Langenecker SA, Afshari A, Albaladejo P, Santullano CAA, De Robertis E. Management of severe perioperative bleeding. Guidelines
from the European Society of Anaesthesiology. Eur J Anaesthesiol 2013;30:270–382.
19. Dietrich WD. The importance of brain temperature in cerebral injury. J Neurotrauma 1992;9:S475–85.
20. Eccher M, Suarez JI. Cerebral edema and intracranial pressure. Monitoring and intracranial dynamics. In: Suarez JI, editor. Critical care neurology and neurosurgery. Totowa (NJ): Humana Press; 2004. p. 47–100.
21. Gokhale S, Khan SA, Agrawal A, Friedman AH, McDonagh DL. Levetiracetam seizure prophylaxis in craniotomy patients at high risk for
postoperative seizures. Asian J Neurosurg 2013;8:169–73.
22. Magni G, Rosa IL, Melillo G, Savio A, Rosa G. A comparison between sevoflurane and desflurane anesthesia in patients undergoing craniotomy for supratentorial intracranial surgery. Anesth Analg August 2009;109:567–71.
23. Bhagat H, bukhal I, bastola P, Bithal PK, Dash HH. A47 does morphine prolong emergence in neurosurgical patients. Eur J Anaesthesiol
2012;29:S-14.
24. Bhagat H, Dash HH, Bithal PK, Chouhan RS, Pandia MP. Planning for early emergence in neurosurgical patients: a randomized prospective
trial of low-dose anesthetics. Anesth Analg 2008;107:1348–55.
25. George SE, Singh G, Mathew BS, Fleming D, Korula G. Comparison of the effect of lignocaine instilled through the endotracheal tube and
intravenous lignocaine on the extubation response in patients undergoing craniotomy with skull pins: a randomized double blind clinical
trial. J Anaesthesiol Clin Pharmacol 2013;29:168–72.
26. De Benedittis G, Lorenzetti A, Spagnoli D, Migliore M, Tiberio F, Villani R. Postoperative pain in neurosurgery: a pilot study in brain surgery.
Neurosurgery 1996;38:466–70.
27. Foy PM, Copeland GP, Shaw MD. The incidence of postoperative seizures. Acta Neurochir 1981;55:253–64.
28. Glantz MJ, Cole BF, Friedberg MH, et al. A randomized, blinded, placebo-controlled trial of divalproex sodium prophylaxis in adults with
newly diagnosed brain tumors. Neurology 1996;46:985–91.
29. Franceschetti S, Binelli S, Casazza M, et al. Influence of surgery and antiepileptic drugs on seizures symptomatic of cerebral tumours. Acta
Neuro Chirurgica 1990;103:47–51.
30. Latz B, Mordhorst C, Kerz T, Schmidt A, Schneider A, Wisser G, et al. Postoperative nausea and vomiting in patients after craniotomy: incidence and risk factors. J Neurosurg 2011;114:491–6.
VII. NEUROSURGERY
C H A P T E R
13
Emergence From Anesthesia
M. Echeverría1, J. Fiorda-Diaz2, N. Stoicea2, S.D. Bergese2
1Centro
Médico Docente Paraíso, Maracaibo, Venezuela; 2Ohio State University, Columbus, OH, United States
O U T L I N E
Introduction
247
Neurophysiological Response During Emergence
in Neurosurgical Patients
248
Specific Perioperative Considerations
Surgical Site and Lesion Location
Anesthetic Techniques
Balanced Anesthesia
Total Intravenous Anesthesia
Anesthetic Technique and Recovery of
Cognitive Function
Dexmedetomidine
248
248
248
249
249
249
249
Delayed Emergence and Arousal
Agitation During Emergence From Anesthesia
Delayed Emergence
Planned Delayed Emergence
Causes of Delayed Emergence
250
250
250
250
251
Complications
Intracranial Hemorrhage or Hematoma
Hypertension and Cerebral Hyperperfusion
Postoperative Nausea and Vomiting
251
251
251
252
Conclusion
252
References
252
INTRODUCTION
Multiple historical factors may be related directly or indirectly with the emergence from anesthesia. William
Harvey’s statement in Exercitatio anatomica de motu cordis et sanguinis in animalibus (On the Movement of the Heart
and Blood in Animals) regarding the blood circulating around the body (1628) took down Galenic dogma (130 BC)
about blood circulatory mechanism1 and created the basis of physiological and pharmacological principles. In 1846,
William T. Morton performed the first successful administration of anesthesia but little was known about emergence
from anesthesia and patients’ reaction after ether administration. Prior to this successful experience, Morton tried
couple of times to anesthetize a patient with unexpected reactions: agitation and disorganized speech.2
Cerebral autoregulation and physiological adaptation in the immediate postoperative period are linked to intraoperative efforts to maintain homeostasis. Induced hypocapnia, use of osmotic fluids, and placement of drains are
examples of perioperative variables with an important impact in outcomes during the emergence from anesthesia.3
An increasing body of evidence regarding anesthesia management supports different techniques based on physicians and institutions’ preferences such as total intravenous anesthesia (TIVA), balanced anesthesia, local anesthesia,
or use of adjuvants and their combinations.4
Postoperative stress response during extubation may entail increased catecholamine response, oxygen consumption (VO2), blood pressure, and heart rate due to laryngeal stimulation, although other mechanisms are described.5
Variations in metabolism, hemodynamic parameters, cerebral blood flow (CBF), and intracranial pressure may be
developed as a result of shivering or pain with consequent impairment in patients’ outcomes.5
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00013-0
247
© 2017 Elsevier Inc. All rights reserved.
248
13. EMERGENCE FROM ANESTHESIA
Neurological examination should be carried out in the operating room during emergence to identify a potential
neurological deficit or emergence delirium. Some patients undergoing complex procedures involving eloquent areas
of the brain may be suitable for a planned delay in emergence from anesthesia, avoiding life-threatening complications such as intracranial hemorrhage (ICH), respiratory impairment, and aspiration due to unprotected airway.6
NEUROPHYSIOLOGICAL RESPONSE DURING EMERGENCE IN NEUROSURGICAL
PATIENTS
Several physiological changes take place during the emergence from anesthesia including metabolic variations
and those affecting brain homeostasis. Even with slightly normal PaCO2 levels, bicarbonate levels in the cerebrospinal fluid (CSF) may decrease significantly.7 Therefore, consequent pH reduction in brain perivascular areas will lead
to vasodilation and hyperemia.7
Apparently, these events occur regardless of the anesthetic agent used during maintenance. Bruder et al. reported
a significant increment of 60% from baseline in CBF velocity during extubation and within the first hour after neurosurgery, with no relation found between these values and the surgery or anesthesia techniques.8
Increased catecholamine blood levels have been reported after neurological surgery.7,8 However, this condition
by itself may interfere with the CBF only in the presence of a defective blood–brain barrier or when autoregulation
is compromised.8 Different options are available to treat catecholamine hemodynamic effects such as β-blocker use
(e.g., esmolol)9,10 and hypothermia prevention.11
On the other hand, sympathetic activation may play a crucial role on the central nervous system (CNS) responses
during emergence from anesthesia. Reuptake inhibition of dopamine and norepinephrine by methylphenidate
administration will trigger several mechanisms within the CNS leading to a faster recovery after isoflurane and
propofol exposure.12,13
Surgery and anesthesia may also increase the incidence of emesis during the emergence. The “emesis center”
includes the area postrema (AP), nucleus tractus solitarius, and the dorsal motor nucleus of the vagus nerve. No less
than 17 neurotransmitters exert their action on this center throughout the interaction with several receptors such as
dopamine, substance P/neurokinin-1, cannabinoid, histamine, among others.14 The aforementioned responses may
be generated during extubation as a result of the autonomic nervous system activation.
SPECIFIC PERIOPERATIVE CONSIDERATIONS
Diagnosis of potential postoperative complications may be delayed due to a slow return to consciousness making neurological examination difficult to perform after neurosurgery. Postanesthesia arousal is a variable considered
when assessing anesthesia quality.15,16 Preoperative neurologic status, location and size of the lesion, and anesthesia
technique are some of the variables interfering with the early postoperative recovery of cognition, hemodynamics,
and nociception during emergence.
Surgical Site and Lesion Location
Brain herniation, ischemia, and poor surgical field are the expected consequences of increased intracranial volume.
These events have been associated with delayed arousal after intracranial surgery.10 Predictive factors for delayed
postsurgical emergence are the following: mass effect of space-occupying lesion (over 30 mm size), cerebral structures shifting from midline >3 mm with perilesional edema, and prolonged retraction pressure needed to expose the
surgical field during tumor resection.10
Anesthetic Techniques
Definitive criteria for quality of recovery related to the anesthesia technique during neurosurgical procedures
have not been established.15 However, a targeted and neurological evaluation is performed at the end of surgery to
assess outcomes.17
The Short Orientation Memory Concentration Test (SOMCT) and the Aldrete score are commonly used when
comparing the time and quality of recovery of patients undergoing supratentorial craniotomy, either under balanced
or TIVA technique.18,19
VII. NEUROSURGERY
SPECIFIC PERIOPERATIVE CONSIDERATIONS
249
TIVA based on propofol–remifentanil has shown slightly shorter extubation time, faster return to consciousness, and faster recovery in comparison with balanced anesthesia.17,20 However, most of the studies have demonstrated no statistical significance in quality of recovery among patients either when TIVA or balanced anesthesia was
performed.5,18,21–24
The NeuroMorfeo Study was carried out in 411 patients reporting no difference in time after extubation to achieve
an Aldrete score ≥9 between both balanced and TIVA groups. Hemodynamics and brain surgical conditions were
also similar among groups. Nevertheless, a reduced endocrine response to surgical stress was linked to propofol/
remifentanil combination.24
Balanced Anesthesia
Desflurane provides shorter extubation and recovery time than sevoflurane in patients undergoing craniotomy
under balanced anesthesia.25,26 Magni et al. compared the effects of sevoflurane (1.5–2%) and desflurane (6–7%) on
emergence analyzing the data collected from 120 neurosurgical patients. No significant difference was found regarding the time for emergence (12.2 ± 4.9 min versus 10.8 ± 7.2 min), recorded as the time when eye opening occurred
since the inhaled agent administration was suspended. On the other hand, the sevoflurane group needed more time
for tracheal extubation in contrast to the desflurane group (18.2 ± 2.3 min versus 11.3 ± 3.9 min). Similarly, the time of
recovery (defined in this study as the time that patients’ consciousness allowed them to repeat their name and date
of birth) was longer in the sevoflurane group (12.4 ± 7.7 min versus 1.3 ± 3.9 min).25 The characteristic of the surgical
field, such as state of the brain tissue and the intracranial pressure (ICP) and postoperative cognitive function are also
comparable between both inhaled anesthetics during supratentorial surgeries.27 Cognitive impairment commonly
occurs in both groups within the first 15 min after extubation25 or after Aldrete score was ≥9, usually returning to
baseline during the next 30 min.26
Moreover, when compared with opioids such as fentanyl, alfentanil, and sufentanil in combination with isoflurane
showed no clinical relevance regarding the time for obtaining a satisfactory neurological evaluation after surgery.28
Total Intravenous Anesthesia
Despite the advantages of propofol use in neurosurgery such as reduction of cerebral metabolic rate of oxygen and
ICP, increase of cerebral perfusion pressure (CPP), and antiemetic effect,17,29–32 adverse events like shivering, high
blood pressure, and postoperative nausea and vomiting (PONV) are not uncommon.4
The effects and outcomes of several combinations of opioids and propofol infusions have been compared during
neurosurgery with different results. Gerlach et al. compared propofol/remifentanil versus propofol/sufentanil in
supratentorial surgeries and reported a reduced time for extubation in the remifentanil group (6.4 min vs. 14.3 min).33
When remifentanil/propofol combination was used, SOMCT and Rancho Los Amigos Scale scores are comparable to
baseline sooner than the interval of time reported by sufentanil/propofol group.19 Nevertheless, Djian et al. showed
similar results among groups regarding extubation time and reported an increase in costs in the remifentanil group
when compared to the sufentanil group.34
On the other hand, remifentanil is characterized by a faster onset in reducing cardiovascular responses during
intubation when compared with fentanyl. However, hemodynamic parameters, ICP, and CPP levels are similar during the maintenance of anesthesia with both, remifentanil and fentanyl, whereas naloxone administration after neurosurgery is importantly increased in patients who received fentanyl.35
Moreover, remifentanil/propofol seems to be a common option for transsphenoidal procedures as it may offer a
reduced incidence of postoperative coughing during and after extubation by providing an ideal emergence.36
In fact, these studies provide more evidence on the convenience of using opioid drips in neuroanesthesia as discontinuation at an appropriate time will result in a shorter extubation and recovery.
Anesthetic Technique and Recovery of Cognitive Function
Recovery of cognitive functions after surgery has become one of the major concerns among neuroanesthesiologists and also an important parameter when comparing different anesthetic techniques in neurosurgical patients.37
In fact, a connection between the pathogenesis of neurodegenerative diseases and the development of postoperative
cognitive dysfunction has been described.37
Dexmedetomidine
Some adjuvants such as the alpha-2 adrenoreceptor agonist dexmedetomidine have been widely used in neuroanesthesia with potential intra- and postoperative advantages. Dexmedetomidine produces dose-dependent sedation, anxiolysis, and analgesia without any concomitant respiratory depression along with decreased anesthetic
VII. NEUROSURGERY
250
13. EMERGENCE FROM ANESTHESIA
requirements (hypnotics and opioids).38,39 These characteristics make dexmedetomidine a suitable drug for
neuroanesthesia.38,39
Smoother arousal and recovery after neurological surgery has been reported with the use of dexmedetomidine as
an adjunct during neuroanesthesia. Bekker et al. carried out a prospective, randomized, double-blinded study and
concluded that dexmedetomidine use during sevoflurane/remifentanil general anesthesia was associated with less
perioperative antihypertensive drugs administration, when compared with placebo (43% vs. 86%).38
Dexmedetomidine is also associated with a reduction in CBV, leading to minimization of intraoperative brain
retraction, optimization of oxygen supply/demand relation, and inhibition of hypercapnia-induced cerebral
vasodilation.39,40
DELAYED EMERGENCE AND AROUSAL
Agitation During Emergence From Anesthesia
Agitation during and after emergence is a clinical situation commonly seen in neurosurgical patients (up to 30% of incidence) and usually described within the first hours after intensive care unit admission. The sedation-agitation scale (SAS)
and the Richmond Agitation-Sedation Scale are useful clinical assessing tools for both physicians and nurses, to detect a
potential postoperative agitation or delirium event.41,42 Around 80% of neurosurgical patients who experienced agitation
during the emergence received a score of ≥6 based on SAS classification (very agitated or dangerous agitation).41
Systemic high blood pressure, increased ICP, hemorrhage, and PONV are the most relevant postoperative complications associated with a fast emergence from anesthesia in neurosurgical patients.43 In addition, extremely agitated
patients may unintentionally pull out their catheters or even the endotracheal tube.41
Randomized controlled trials have compared the influence of different anesthetics on emergence agitation after
intracranial surgery, revealing that the incidence of agitation during the early phase of recovery varied from 2.5% to
13.3%.17,24,44 Chen et al. identified several independent factors associated with emergence agitation after craniotomy
under general anesthesia such as patient-related factors (male gender or patients taking antidepressant drugs or
benzodiazepines), anesthesia-related factors (anesthesia management, time exposed to anesthesia, and existence of
endotracheal tube), and surgery-related factors (frontal approach).41
Surgery and anesthesia time, pain, and anesthesia management have been also reported as relevant predictors
for delirium and agitation during emergence in nonneurosurgical patients.45–47 In this patient setting, TIVA using
propofol seems to decrease the incidence of agitation during the emergence from anesthesia.45,46,48
In summary, preemptive techniques and pharmacological interventions could be applied to decrease the onset of
agitation during the emergence or within the first postoperative hours.
Delayed Emergence
Early neurological assessments after surgery entail a fast recovery from the moment of emergence from general
anesthesia. Nevertheless, rapid emergence is not an adequate option for every patient. Baseline assessed neurological status, surgical concerns and prognosis, hemodynamics, and respiratory support, among other factors, may play
an important role on delaying emergence (planned or unexpected).43 Hypothermia or pain may contribute to the
postoperative physiologic stress responses to emergence.43,49
Delayed emergence is considered when patients failed to awake being unable to respond to simple verbal commands. The time when a delayed emergence should be diagnosed is 15 min after anesthesia discontinuation, although
up to 30 min has also been considered.6,50
Planned Delayed Emergence
In patients with compromised clinical conditions, risks of early extubation may outweigh the benefits. Impaired
baseline level of consciousness, poor airway control, prolonged (>6 h) or complicated surgery, unstable intraoperative hemodynamics or ventilation difficulties, and increased brain swelling are some of the clinical situations in
which planned delayed recovery should be considered.5 Other considerations may include surgery involving eloquent areas of the brain, significant brain ischemia, and posterior fossa lesions.5
Cai et al. reported a 49.8% rate of delayed extubation after intracranial surgery (398 out of 800 patients).51 Although
data may be limited, the criteria for delayed extubation have been described in reviews and observational prospective studies.43,51
VII. NEUROSURGERY
COMPLICATIONS
251
If a planned emergence delay has been decided, the possibility of a brief awakening to perform a short neurological assessment is a common practice among physicians. Complementary options to an early neurologic evaluation
are postoperative computed tomography or ICP monitoring.6
In patients undergoing posterior fossa or infratentorial surgery, the feasibility of postoperative extubation will
be determined by (but not limited to) the location of the lesion, the extent of surgery (especially if the brainstem is
involved), and brain edema.52,53 PONV is also more frequent in posterior fossa surgery.53 Unprotected airway combined with PONV may result in life-threatening situations such as aspiration of gastric content, increased ICP, among
others.
Surgical positioning may also interfere with patient outcomes during emergence. Macroglossia and consequent
upper airway obstruction is usually related with the sitting and prone position probably due to a regional disruption in venous and lymphatic drainage.54 Some authors suggest that patients undergoing posterior fossa surgery
should be extubated at least 1–2 h after they are considered to be fully recovered from anesthesia. However, increased
incidence of pneumonia coexisting with other comorbidities, higher incidence of tracheostomy, prolonged hospital
admission, and increased costs have been associated with delayed emergence after neurosurgery.51
Causes of Delayed Emergence
Scientific evidence incriminates multiple factors in delayed emergence such as perioperative opiate analgesia,50
hypothermia,5 anxiolytics, metabolic or electrolyte disturbances, drug clearance impairment, stroke, pneumocephalus, CSF hypotension, and seizures.10
COMPLICATIONS
Even though PONV and hypertension are commonly diagnosed complications in patients recovering in postanesthesia care unit, preventing their onset is one of the main goals in neurosurgical patients due to associated
life-threatening consequences.5 The most common signs of deterioration after intracranial surgery are decreased
level of consciousness and focal neurologic deficit. Cerebral hemorrhage usually occurs in the first 6 h after surgery,
justifying the importance of closed clinical monitoring within the first postoperative hours.5 PONV occurs more frequently after neurosurgical procedures than after general surgical procedures.53 ICH and seizures are also described
as complications after neurosurgery.5
Intracranial Hemorrhage or Hematoma
Physiological responses to stimulation induced by extubation and recovery are associated with ICH and tissue
edema.8 Overall the incidence of ICH after neurosurgery may vary between 0.77% and 3.9% among studies, with
a maximum value of 10.8% reported by Fukamachi et al. in the 1980’s.55 A retrospective study conducted by Basali
et al. in 11,214 craniotomy surgeries found that 62% of patients with ICH experienced intraoperative hypertension,
acute episodes of postoperative hypertension being linked to increased morbidity and mortality by cerebral edema
or ICH.56
Hypertension and Cerebral Hyperperfusion
Increased blood pressure is an expected postoperative event after brain surgery,9,57 probably due to activation of
autoregulatory mechanisms. However, postoperative hypertension has been associated with an increased risk of
postoperative cerebral hyperemia.8,56
Autoregulation may be impaired after neurosurgery, especially in retracted areas and its surroundings. Therefore,
any postoperative increase in blood pressure will overwhelm autoregulatory responses and compromise cerebral
perfusion. Although blood pressure control may be achieved during the emergence from anesthesia and reduce the
incidence of postoperative intracranial bleeding, it cannot prevent cerebral hyperperfusion.55,56,58–60
Bruder et al. considered that cerebral hyperemia occurs as a result of cerebral metabolic stress.7 Additionally,
CBF reestablished after prolonged hypocapnia may be also considered a contributing factor to cerebral hyperemia in neurosurgical patients.7,8,61 In terms of postoperative hemodynamic changes, when comparing TIVA to
inhalational anesthesia, differences in blood pressure and heart rate seem to have no statistical significance.20,21,23
However, Magni et al. concluded that remifentanil is associated with more frequent hypertension than sevoflurane/
fentanyl.18
VII. NEUROSURGERY
252
13. EMERGENCE FROM ANESTHESIA
Esmolol, a selective β-adrenergic receptor antagonist, is considered an effective pharmacological option to attenuate hyperdynamic responses occurring during extubation and postextubation stages.62,63 Moreover, blocking the β1adrenergic receptor may reduce cerebral stress responses and hyperperfusion without any relevant change on ICP.27,63
Postoperative Nausea and Vomiting
The overall incidence of PONV has been reported with high variability from 38% to 80% in some patient populations.64,65 Fabling et al. found an important incidence of PONV in patients who underwent infratentorial surgery even when prophylactic medication was administered. Therefore, multiple variables not related to anesthesia
management must be taken into consideration to achieve a successful control of PONV in neurosurgical patients,65
improving outcomes, and avoiding hemorrhagic complications associated with this complication.32
Intracranial surgery entails a higher risk for PONV when compared to general surgery. In addition to general
PONV risk factors such as gender and smoking status, surgical site (higher incidence in infratentorial procedures),
and manipulation around the AP have a specific weight in the onset of PONV after neurosurgery.66
Triple therapy for PONV prophylaxis (droperidol, promethazine, and dexamethasone) is widely accepted among
physicians. Moreover, other pharmacological options such as aprepitant and fosaprepitant have demonstrated longer effects,67 but supporting clinical literature is limited.
CONCLUSION
Hemodynamic and pathologic responses to complications occurring during emergence from anesthesia are
potentially life threatening in neurosurgical patients, as they will disrupt the physiology of an overwhelmed central
nervous system. Certainly, anesthetics and adjuvants play an important role in maintaining perioperative cerebral
homeostasis.
Cardiovascular, neurologic, and respiratory effects of the anesthetics are dose dependent in the vast majority of
the cases. Therefore, a combination of drugs with different mechanisms of action offers synergistic effects by reducing the plasmatic concentrations of every single one, avoiding the onset of adverse events usually linked to higher
concentrations. Although no definitive criteria have been established for anesthesia management during neurosurgery, there are general considerations that should be addressed based on current clinical conditions of the patient and
expected surgery outcomes.
Attenuation of neurological responses to local or systemic stimulation during emergence should be the paramount
to the neuroanesthesia management, improving patients’ outcomes and decreasing postoperative complications.
References
1. Wright T. William Harvey goes back to the future. Lancet 2013;381(9867):620–1.
2. Haridas RP. “Gentlemen! This is no humbug” Did John Collins Warren, MD, proclaim these words on October 16, 1846, at Massachusetts
General Hospital, Boston? J Am Soc Anesthesiol March 1, 2016;124(3):553–560.
3. Yan L-M, Chen H, Yu R-G, et al. Emergence agitation during recovery from intracranial surgery under general anaesthesia: a protocol and
statistical analysis plan for a prospective multicentre cohort study. BMJ Open 2015;5(4):e007542.
4. Wong A, O’Regan A, Irwin M. Total intravenous anaesthesia with propofol and remifentanil for elective neurosurgical procedures: an audit of
early postoperative complications. Eur J Anaesthesiol 2006;23(7):586–90.
5. Bruder N, Ravussin P. Recovery from anesthesia and postoperative extubation of neurosurgical patients: a review. J Neurosurg Anesthesiol
1999;11(4):282–93.
6. Munis JR, Marcukaitis AW, Sprung J. Delayed emergence from anesthesia associated with absent brainstem reflexes following suboccipital
craniotomy. Neurocritical Care 2006;5(3):206–9.
7. Schubert A. Cerebral hyperemia, systemic hypertension, and perioperative intracranial morbidity: is there a smoking gun? Anesth Analg
2002;94(3):485–7.
8. Bruder N, Pellissier D, Grillot P, Gouin F. Cerebral hyperemia during recovery from general anesthesia in neurosurgical patients. Anesth Analg
2002;94(3):650–4.
9. Gibson B, Oliver S, Maass L, Cucchiara R. Esmolol for the control of hypertension following neurologic surgery. Anesth Analg 1988;67(2):71.
10. Jellish WS. Arousal from anesthesia after neurosurgical operations. In: Essentials of neurosurgical anesthesia & critical care. Springer; 2012. p. 409–17.
11. Frank SM, Fleisher LA, Olson KF, et al. Multivariate determinants of early postoperative oxygen consumption in elderly patients. Effects of
shivering, body temperature, and gender. J Am Soc Anesthesiol 1995;83(2):241–9.
12. Solt K, Cotten JF, Cimenser A, Wong KF, Chemali JJ, Brown EN. Methylphenidate actively induces emergence from general anesthesia. J Am
Soc Anesthesiol 2011;115(4):791–803.
13. Chemali JJ, Van Dort CJ, Brown EN, Solt K. Active emergence from propofol general anesthesia is induced by methylphenidate. J Am Soc
Anesthesiol 2012;116(5):998–1005.
VII. NEUROSURGERY
REFERENCES
253
14. Bergese S, Viloria A, Uribe A, Antor A, Fernandez S. Aprepitant versus ondansetron in preoperative triple-therapy treatment of nausea and
vomiting in neurosurgery patients: study protocol for a randomized controlled trial. Trials 2012;13(1):130.
15. Leslie K, Troedel S, Irwin K, et al. Quality of recovery from anesthesia in neurosurgical patients. J Am Soc Anesthesiol 2003;99(5):1158–65.
16. Royse CF, Newman S, Chung F, et al. Development and feasibility of a scale to assess postoperative recovery: the post-operative quality
recovery scale. J Am Soc Anesthesiol 2010;113(4):892–905.
17. Todd MM, Warner DS, Sokoll MD, et al. A prospective, comparative trial of three anesthetics for elective supratentorial craniotomy. Propofol/
fentanyl, isoflurane/nitrous oxide, and fentanyl/nitrous oxide. Anesthesiology 1993;78(6):1005–20.
18. Magni G, Baisi F, La Rosa I, et al. No difference in emergence time and early cognitive function between sevoflurane-fentanyl and propofolremifentanil in patients undergoing craniotomy for supratentorial intracranial surgery. J Neurosurg Anesthesiol 2005;17(3):134–8.
19. Bilotta F, Caramia R, Paoloni F, et al. Early postoperative cognitive recovery after remifentanil–propofol or sufentanil–propofol anaesthesia
for supratentorial craniotomy: a randomized trial. Eur J Anaesthesiol 2007;24(2):122–7.
20. Ravussin P, Tempelhoff R, Modica PA, Bayer-Berger M-M. Propofol vs. thiopental-isoflurane for neurosurgical anesthesia: comparison of
hemodynamics, CSF pressure, and recovery. J Neurosurg Anesthesiol 1991;3(2):85–95.
21. Lauta E, Abbinante C, Del Gaudio A, et al. Emergence times are similar with sevoflurane and total intravenous anesthesia: results of a multicenter RCT of patients scheduled for elective supratentorial craniotomy. J Neurosurg Anesthesiol 2010;22(2):110–8.
22. Bhagat H, Dash HH, Bithal PK, Chouhan RS, Pandia MP. Planning for early emergence in neurosurgical patients: a randomized prospective
trial of low-dose anesthetics. Anesth Analg 2008;107(4):1348–55.
23. Sneyd J, Andrews C, Tsubokawa T. Comparison of propofol/remifentanil and sevoflurane/remifentanil for maintenance of anaesthesia for
elective intracranial surgery. Br J Anaesth 2005;94(6):778–83.
24. Citerio G, Pesenti A, Latini R, et al. A multicentre, randomised, open-label, controlled trial evaluating equivalence of inhalational and intravenous anaesthesia during elective craniotomy. Eu J Anaesthesiol 2012;29(8):371–9.
25. Magni G, La Rosa I, Melillo G, Savio A, Rosa G. A comparison between sevoflurane and desflurane anesthesia in patients undergoing craniotomy for supratentorial intracranial surgery. Anesth Analg 2009;109(2):567–71.
26. Bilotta F, Doronzio A, Cuzzone V, Caramia R, Rosa G, Group PS. Early postoperative cognitive recovery and gas exchange patterns after balanced anesthesia with sevoflurane or desflurane in overweight and obese patients undergoing craniotomy: a prospective randomized trial.
J Neurosurg Anesthesiol 2009;21(3):207–13.
27. Dube SK, Pandia MP, Chaturvedi A, Bithal P, Dash HH. Comparison of intraoperative brain condition, hemodynamics and postoperative
recovery between desflurane and sevoflurane in patients undergoing supratentorial craniotomy. Saudi J Anaesth 2015;9(2):167.
28. From RP, Warner DS, Todd MM, Sokoll MD. Anesthesia for craniotomy: a double-blind comparison of alfentanil, fentanyl, and sufentanil.
Anesthesiol 1990;73(5):896–904.
29. Hans P, Bonhomme V. Why we still use intravenous drugs as the basic regimen for neurosurgical anaesthesia. Curr Opin Anesthesiol
2006;19(5):498–503.
30. Petersen KD, Landsfeldt U, Cold GE, et al. Intracranial pressure and cerebral hemodynamic in patients with cerebral tumors: a randomized
prospective study of patients subjected to craniotomy in propofol-fentanyl, isoflurane-fentanyl, or sevoflurane-fentanyl anesthesia. J Am Soc
Anesthesiol 2003;98(2):329–36.
31. Kahveci FS, Kahveci N, Alkan T, Goren B, Korfali E, Ozluk K. Propofol versus isoflurane anesthesia under hypothermic conditions: effects on
intracranial pressure and local cerebral blood flow after diffuse traumatic brain injury in the rat. Surg Neurol 2001;56(3):206–14.
32. Gupta A, Stierer T, Zuckerman R, Sakima N, Parker SD, Fleisher LA. Comparison of recovery profile after ambulatory anesthesia with propofol, isoflurane, sevoflurane and desflurane: a systematic review. Anesth Analg 2004;98(3):632–41.
33. Gerlach K, Uhlig T, Hüppe M, et al. Remifentanil–propofol versus sufentanil–propofol anaesthesia for supratentorial craniotomy: a randomized trial. Eur J Anaesthesiol 2003;20(10):813–20.
34. Djian M-C, Blanchet B, Pesce F, et al. Comparison of the time to extubation after use of remifentanil or sufentanil in combination with
propofol as anesthesia in adults undergoing nonemergency intracranial surgery: a prospective, randomized, double-blind trial. Clin Ther
2006;28(4):560–8.
35. Guy J, Hindman BJ, Baker KZ, et al. Comparison of remifentanil and fentanyl in patients undergoing craniotomy for supratentorial spaceoccupying lesions. J Am Soc Anesthesiol 1997;86(3):514–24.
36. Choi SH, Min KT, Lee J-R, et al. Determination of EC95 of remifentanil for smooth emergence from propofol anesthesia in patients undergoing
transsphenoidal surgery. J Neurosurg Anesthesiol 2015;27(2):160–6.
37. Bilotta F, Doronzio A, Stazi E, et al. Postoperative cognitive dysfunction: toward the Alzheimer’s disease pathomechanism hypothesis.
J Alzheimer’s Dis 2010;22(s3):81–9.
38. Bekker A, Sturaitis M, Bloom M, et al. The effect of dexmedetomidine on perioperative hemodynamics in patients undergoing craniotomy.
Anesth Analg 2008;107(4):1340–7.
39. Schwarz A, Nossaman B, Carollo D, Ramadhyani U. Dexmedetomidine for neurosurgical procedures. Curr Anesthesiol Rep 2013;3(3):205–9.
40. Farag E, Argalious M, Sessler DI, Kurz A, Ebrahim ZY, Schubert A. Use of α2-agonists in neuroanesthesia: an overview. Ochsner J 2011;11(1):57–69.
41. Chen L, Xu M, Li G-Y, Cai W-X, Zhou J-X. Incidence, risk factors and consequences of emergence agitation in adult patients after elective
craniotomy for brain tumor: a prospective cohort study. PLoS One 2014;9(12):e114239.
42. Barr J, Fraser GL, Puntillo K, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the
intensive care unit. Crit Care Med 2013;41(1):263–306.
43. Bruder NJ. Awakening management after neurosurgery for intracranial tumours. Curr Opin Anesthesiol 2002;15(5):477–82.
44. Kanaya N, Kuroda H, Nakayama M, Homma Y, Namiki A. Does propofol anesthesia increase agitation in neurosurgical patients?—a pilot
study. Can J Anesth 2002;49(6):638–9.
45. Lepouse C, Lautner C, Liu L, Gomis P, Leon A. Emergence delirium in adults in the post-anaesthesia care unit. Br Journal Anaesth
2006;96(6):747–53.
46. Yu D, Chai W, Sun X, Yao L. Emergence agitation in adults: risk factors in 2000 patients. Can J Anesth 2010;57(9):843–8.
47. Radtke F, Franck M, Hagemann L, Seeling M, Wernecke K, Spies C. Risk factors for inadequate emergence after anesthesia: emergence delirium and hypoactive emergence. Minerva Anestesiol 2010;76(6):394–403.
VII. NEUROSURGERY
254
13. EMERGENCE FROM ANESTHESIA
48. Liang C, Ding M, Du F, Cang J, Xue Z. Sevoflurane/propofol coadministration provides better recovery than sevoflurane in combined general/epidural anesthesia: a randomized clinical trial. J Anesth 2014;28(5):721–6.
49. Bruder N, Stordeur J-M, Ravussin P, et al. Metabolic and hemodynamic changes during recovery and tracheal extubation in neurosurgical
patients: immediate versus delayed recovery. Anesth Analg 1999;89(3):674.
50. Schubert A, Mascha EJ, Bloomfield EL, DeBoer GE, Gupta MK, Ebrahim ZY. Effect of cranial surgery and brain tumor size on emergence from
anesthesia. J Am Soc Anesthesiol 1996;85(3):513–21.
51. Cai YH, Zeng H-Y, Shi Z-H, et al. Factors influencing delayed extubation after infratentorial craniotomy for tumour resection: a prospective
cohort study of 800 patients in a Chinese neurosurgical centre. J Int Med Res 2013;41(1):208–17.
52. Artru AA, Cucchiara RF, Messick JM. Cardiorespiratory and cranial-nerve sequelae of surgical procedures involving the posterior fossa. J Am
Soc Anesthesiol 1980;52(1):83–5.
53. Jagannathan S, Krovvidi H. Anaesthetic considerations for posterior fossa surgery. Contin Educ Anaesth, Crit Care Pain 2014;14(5):202–6.
54. Lam AM, Vavilala MS. Macroglossia: compartment syndrome of the tongue? J Am Soc Anesthesiol 2000;92(6):1832.
55. Fukamachi A, Koizumi H, Nukui H. Postoperative intracerebral hemorrhages: a survey of computed tomographic findings after 1074 intracranial operations. Surg Neurol 1985;23(6):575–80.
56. Basali A, Mascha EJ, Kalfas I, Schubert A. Relation between perioperative hypertension and intracranial hemorrhage after craniotomy. J Am
Soc Anesthesiol 2000;93(1):48–54.
57. Günes Y, Türktan M, Erman T, Özcengiz D. Comparison of dexmedetomidine, remifentanil, and esmolol for the control of hypertension during tracheal extubation and emergence from anesthesia after a craniotomy. Neurosurg Q 2013;23(4):294–8.
58. Felding M, Jakobsen CJ, Gold G, Davidsen B, Jensen K. The effect of metoprolol upon blood pressure, cerebral blood flow and oxygen consumption in patients subjected to craniotomy for cerebral tumours. Acta Anaesthesiol Scand 1994;38(3):271–5.
59. Felding M, Cold G, Jagobsen CJ, Stjernholm P, Voss K. The effect of ketanserin upon postoperative blood pressure, cerebral blood flow and
oxygen metabolism in patients subjected to craniotomy for cerebral tumours. Acta Anaesthesiol Scand 1995;39(5):582–5.
60. Dalman J, Beenakkers I, Moll F, Leusink J, Ackerstaff R. Transcranial Doppler monitoring during carotid endarterectomy helps to identify
patients at risk of postoperative hyperperfusion. Eur J Vasc Endovasc Surg 1999;18(3):222–7.
61. Albrecht RF, Miletich DJ, Ruttle M. Cerebral effects of extended hyperventilation in unanesthetized goats. Stroke 1987;18(3):649–55.
62. Hosseinzadeh H, Eydi M, Ghaffarlou M, Ghabili K, Golzari S. Esmolol: a unique beta-blocker in maintaining cardiovascular stability following neurosurgical procedures. Adv Pharm Bull 2012;2(2):249–52.
63. Bilotta F, Lam AM, Doronzio A, Cuzzone V, Delfini R, Rosa G. Esmolol blunts postoperative hemodynamic changes after propofol-remifentanil total intravenous fast-track neuroanesthesia for intracranial surgery. J Clin Anesth 2008;20(6):426–30.
64. Gan TJ, Diemunsch P, Habib AS, et al. Consensus guidelines for the management of postoperative nausea and vomiting. Anesth Analg
2014;118(1):85–113.
65. Fabling JM, Gan TJ, El-Moalem HE, Warner DS, Borel CO. A randomized, double-blind comparison of ondansetron versus placebo for prevention of nausea and vomiting after infratentorial craniotomy. J Neurosurg Anesthesiol 2002;14(2):102–7.
66. Latz B, Mordhorst C, Kerz T, et al. Postoperative nausea and vomiting in patients after craniotomy: incidence and risk factors. J Neurosurg
2011;114(2):491–6.
67. Tsutsumi YM, Kakuta N, Soga T, et al. The effects of intravenous fosaprepitant and ondansetron for the prevention of postoperative nausea
and vomiting in neurosurgery patients: a prospective, randomized, double-blinded study. BioMed Res Int 2014:2014.
VII. NEUROSURGERY
C H A P T E R
14
Anesthesia for Posterior Fossa Surgery
K. Sandhu1, N. Gupta2
1Max
Superspeciality Hospital, New Delhi, India; 2Indraprastha Apollo Hospital, New Delhi, India
O U T L I N E
Introduction
255
Anatomy
255
Clinical Presentation
256
Perioperative Management of Patients for Posterior
Fossa Surgery
256
Preoperative Evaluation
256
Surgical Approach
257
Anesthetic Considerations During Posterior Fossa
Craniotomy
257
Premedication
258
Patient Positioning
258
Supine
258
Lateral
258
Park-Bench (Semiprone)
258
Prone
258
Sitting or Semisitting Position
259
Technical Considerations
259
Anesthetic Management
Respiratory Management
Hemodynamic Management
Intraoperative Monitoring
261
261
262
263
Venous Air Embolism
Incidence
Pathophysiology
Clinical Presentation of Venous Air Embolism
Monitoring
Grading of Venous Air Embolism
Paradoxical Air Embolism
Management of Venous Air Embolism
264
264
264
265
265
268
268
270
Postoperative Management
271
Complications
272
Abbreviations
272
References
273
INTRODUCTION
Posterior fossa surgery poses significant challenges to both the anesthesiologist and surgeons with a wider variety
of complications than surgery in the supratentorial compartment. Apart from the general perioperative considerations involving any intracranial lesion, highlights of posterior fossa lesions include unusual surgical positioning
and its complications, potential for brain stem injury, lengthy surgical procedures, perioperative cardiovascular and
respiratory embarrassment, and acute obstructive hydrocephalus.
A thorough understanding of the patient’s history, neurological findings, imaging studies, operative anatomy, as well
as all potential adverse events associated with the procedure is thus of paramount importance to minimize complications.
ANATOMY
Posterior cranial fossa is the largest and deepest cranial fossa. It houses the brain stem (midbrain, pons, and upper
medulla), the cerebellum, 3rd to 12th cranial nerve nuclei, the ascending and descending tracts, and the vertebrobasilar vascular system.
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00014-2
255
© 2017 Elsevier Inc. All rights reserved.
256
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
The presence of structures vital for control of airway, cardiovascular and respiratory systems within the narrow
confines of this rigid and compact space makes the surgical anatomy unique and challenging. The presence of
any space occupying pathology may lead to mass effect on vital brain stem structures. In addition, the cerebrospinal fluid (CSF) pathway is very narrow through the cerebral aqueduct, and a minor obstruction can cause acute
hydrocephalus with significant increase in intracranial pressure (ICP). Presence of multiple large venous sinuses
contained within the dural folds of the tentorium further adds to the risk of bleeding and air entrainment during
surgery.
CLINICAL PRESENTATION
Symptomatology of lesions in the posterior cranial fossa differs from supratentorial tumors in terms of presentation and rapid worsening, as seen with acute hydrocephalus. Most posterior fossa tumors present with signs and
symptoms of increased ICP including headache, nausea, or vomiting and papilledema.
Signs and symptoms typical to the site of the lesion include movement disorders, altered tonicity, and ocular
signs such as nystagmus, strabismus, diplopia, and pupillary abnormality. Additionally, there may be cranial
nerve dysfunction, bulbar palsy, bradycardia, respiratory embarrassment, and sudden brain stem herniation
leading to death. The classic triad of symptoms referable to a mass in the posterior fossa is said to be headache,
vomiting, and ataxia. Unlike supratentorial tumors, seizures are rare. From pathological perspective, lesion in the
posterior fossa may be neoplastic (most common), developmental, vascular, and traumatic (Table 14.1).
PERIOPERATIVE MANAGEMENT OF PATIENTS FOR POSTERIOR FOSSA SURGERY
Preoperative Evaluation
As a routine, preoperative evaluation should include a thorough history and clinical evaluation of respiratory, cardiovascular, and neurologic systems; airway anatomy and necessary investigations based on patient’s
requirements; and institutional protocols. Specific considerations for posterior fossa lesions include evaluation
based on surgical approach and the intended patient positioning.
The focus of preoperative evaluation should be on the identification as well as optimization of any coexisting
medical conditions. Quantification and risk stratification of patients with known coronary artery disease and
carotid disease is essential as it poses excessive risks in certain surgical positions. Hypertension resets autoregulatory range and might result in significant perfusion deficits due to hypotension associated with positions such as
sitting and prone.
A decrease in the level of consciousness and altered respiratory pattern may indicate the presence of elevated
ICP. External ventricular drainage or other shunt procedures may be indicated to manage hydrocephalus before
surgery or intraoperatively. Raised ICP may be associated with vomiting and inadequate intake resulting in
hypovolemic status, which may give rise to significant hemodynamic perturbations on induction of anesthesia
and positioning. In addition, presence of diabetes insipidus, administration of diuretics, and use of intravenous (IV) contrast agents to facilitate imaging may contribute to dehydration and electrolyte disturbances.
Preoperative administration of IV fluid and optimization of electrolytes should be considered on an individual
basis. Cerebellar hemangioblastomas often secrete erythropoietin, resulting in polycythemia, which should be
taken into consideration during preoperative evaluation.
Preoperative evaluation and documentation of dysphagia, cough, gag, and other cranial nerve dysfunctions; evaluation of cerebellar functions; vision and auditory functions may be needed in specific types of tumor. In patients
with bulbar dysfunction, loss of gag and cough reflex increases the risk of aspiration pneumonitis and extubation
failure. Hence, possibility of need for postoperative ventilation or tracheostomy and extended intensive care unit
(ICU) stay should be explained preoperatively.
Patients with atlantoaxial subluxation and lack of neck movement secondary to craniocervical fusion can present
challenges during airway management and positioning, especially in sitting and prone position surgery. Hence, a
preoperative assessment of cervical spine by dynamic flexion and extension views and Doppler study of neck vessels to look out for carotid insufficiency should be done in all patients where extreme neck flexion is anticipated,
especially in the elderly.1,2
VII. NEUROSURGERY
257
PERIOPERATIVE MANAGEMENT OF PATIENTS FOR POSTERIOR FOSSA SURGERY
TABLE 14.1 Location and Types of Various Posterior Fossa Pathologies
Intraaxial tumors
Cerebellum
Astrocytoma
Hemangioblastoma
Metastasis
Fourth ventricle and pons
Medulloblastoma
Ependymoma
Choroid plexus papilloma
Hemangioblastoma
Extraaxial tumors
Cerebellopontine angle
Vestibular schwanoma
Meningioma
Epidermoid tumor
Glomus jugulare tumor
Skull base
Metastasis
Chordoma
Chondrosarcoma
Brain stem
Glioma
Hemangioblastoma
Vascular malformations
Posterior cerebellar artery aneurysm
Vertebral/vertebrobasillar aneurysm
Basillar tip aneurysm
AV malformations
Cerebellar hematoma
Cerebellar infarction
Cysts
Epidermoid cyst
Arachnoid cyst
Cranial nerve lesion
Trigeminal neuralgia
(cranial nerve V)
Hemifacial spasm
(cranial nerve VII)
Glossopharyngeal neuralgia
(cranial nerve IX)
Craniocervical abnormalities
Atlanto-occipital instability
Congenital
Acquired
Atlantoaxial instability
Congenital
Acquired
Arnold–Chiari malformation
For patients to be operated on in a sitting position, there is not a uniform approach on what special preoperative evaluation is necessary. However, detailed evaluation should be conducted to minimize complications that are
preventable if known. If sitting position is planned, it is prudent to rule out a patent foramen ovale (PFO) using a
transthoracic echocardiography3–6 and perform PFO closure if present,7 to prevent paradoxical air embolism (PAE)
(discussed in detail later).
Surgical Approach
There are several surgical approaches to the posterior fossa, which include suboccipital (retrosigmoid) approach
and midline posterior approach, which can be subtentorial or transtentorial. There are less common ones such as
translabyrinthine, subtemporal/middle cranial fossa approaches or combinations of the above.
Anesthetic Considerations During Posterior Fossa Craniotomy
The surgical complexity of the posterior fossa and the hazards of different patient positioning make the intraoperative management of a patient posted for posterior fossa craniotomy quite challenging and unique. In addition
VII. NEUROSURGERY
258
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
to the basic neuroanesthetic considerations inherent to any neurosurgical procedure, the major intraoperative goals
during posterior fossa craniotomy are:
1. to provide optimal patient positioning and surgical access, with minimum possibility of positioning-related
hazards to the patient;
2. maintaining adequate depth of anesthesia while avoiding hemodynamic instability;
3. to provide optimum conditions for intraoperative neurophysiological monitoring (IONM);
4. prevention, early identification, and effective management of venous air embolism (VAE); and
5. to allow smooth emergence with early awakening so as to facilitate neurological assessment.
Premedication
Premedication should include all regular medications, including steroids (dexamethasone). The role of sedative
premedication is limited in patients with posterior fossa lesions with their inherent risk of hypoventilation and
potential to increase ICP. However, short-acting benzodiazepine given under supervision may be reserved for anxious patients who are neurologically intact.6
Patient Positioning
Proper patient positioning during posterior fossa surgery is one of the most important factors for success or failure
of the procedure. All positions have advantages and disadvantages, assessed either from the surgical or anesthetic
perspective. The greatest challenge for the anesthesiologist is to choose the most appropriate surgical position that
provides the best surgical exposure as well as pose minimum positioning related risks to the patient. Great attention
should thus be paid to the physical and physiologic consequences of different surgical positions to help prevent serious adverse events and associated complications.8
Depending on the planned surgical approach and the lesion, the most common positions for posterior fossa surgery are supine, lateral, park bench (semiprone), prone, and semisitting. (Discussed in detail elsewhere in the book.)
The surgical approach must be individualized for each patient because the risk of postoperative complications may
vary greatly with patient’s age, neurological status, and lesion location.
Supine
Acoustic neuroma and cerebellopontine angle (CPA) tumors may be carried out in the supine position with the
head turned to the opposite side and placement of a sandbag under the ipsilateral shoulder to minimize stretching
of the brachial plexus.
Lateral
The lateral position facilitates gravity-assisted drainage of blood and CSF and provides good surgical access
for unilateral procedures. Patient instability and brachial plexus injury are potential positioning hazards of this
position.
Park-Bench (Semiprone)
The park-bench position is a modification of lateral position with back elevated and head turned about 30% facing down with maximum possible neck flexion. It provides better access of the posterior fossa, as compared with
the lateral position and can be attained quickly. It may be used to gain rapid access to cerebellar hemispheres, for
example, need for rapid evacuation of a posterior fossa bleed. It gives lesser hemodynamic perturbations but the
surgical orientation of the neck may be lost after draping.
Prone
The prone position is the oldest and most suited for midline approach during infratentorial craniotomies. Over a
period of time, it gained popularity because of much lower incidence of clinically significant VAE when compared
with the sitting position (discussed in VAE section). However, a retrospective survey comparing the semisitting and
prone positions for posterior fossa surgery in children found more intraoperative and postoperative complications in
the prone position, with increased length of intensive care and hospital stay.9 In addition, the amount of blood loss is
more than in sitting craniotomies.10 Logistically, it is the most difficult positioning due to challenges associated with
VII. NEUROSURGERY
PERIOPERATIVE MANAGEMENT OF PATIENTS FOR POSTERIOR FOSSA SURGERY
259
providing adequate oxygenation, ensuring adequate ventilation, maintaining hemodynamics, and securing intravenous lines and the tracheal tube.8
Sitting or Semisitting Position
In neurosurgical practice, sitting position remained popular during the 1960s and 1970s for procedures involving
the cervicodorsal spine and posterior and lateral cranial fossae. Later, its use declined throughout the neurosurgical
community because of its potential severe consequences and associated litigations, with an only absolute indication
being the supracerebellar infratentorial surgical approach for pineal gland tumors. Indeed, there has been a great difference between countries in its use since its introduction into clinical practice, being popular mainly in Europe and
India and is still a subject of controversy.10–13
At present, however, there is no hard scientific evidence justifying the abandonment of this patient position
with the recent literature proving its safety in experienced hands, with appropriate monitoring.4,6 Feigl et al. have
demonstrated that under meticulous anesthesia and neurosurgical management, even patients with a PFO can be
operated on in the semisitting position with only a very low risk for VAE.4 Henceforth, by keeping a high index of
suspicion for possible VAE and managing them promptly to limit their progression, the sitting position is a safe
alternative to the prone and lateral positions for posterior fossa and posterior cervical spine surgery.
Nonetheless, the potential advantages of the sitting position in emergency cases (reduction of cerebellar swelling)
may be annihilated by the additional time spent for positioning. Therefore alternative techniques of positioning (e.g.,
concorde, semiprone, etc.) should be considered whenever applicable. The extra positioning time to put a patient
into the sitting position takes approximately 15–20 min longer and consists of placing the transesophageal echocardiography (TEE) probe and bringing the patient in an upright position.
Technical Considerations
Sitting or semisitting position offers a number of technical advantages for the neurosurgeon and neuroanesthesiologist compared to other positions, especially for surgeries of large and vascularized tumors in the posterior cranial fossa and CPA1,4,6,10,11,14,15 (Table 14.2). Various studies have shown reduced operating time, less intraoperative
venous bleeding with lower transfusion rates, and better preservation of cranial nerve function in the sitting position
compared with horizontal positions, despite having a higher incidence of VAE.10,14,16
Sitting position, however, poses unique physiological challenges for the neuroanesthesiologist. Changing from
the supine to the sitting position induces a significant decrease of cardiac index, stroke volume index, right atrial
TABLE 14.2 Advantages of the Sitting Position1,4,6,10,11,14,15
For the neurosurgeon:
1. Improved surgical exposure
2. Improved anatomical orientation
3. Improved venous drainage from the surgical field
4. Cleaner operating field due to gravitational drainage of the cerebrospinal fluid, blood, or irrigation fluid from the operative field
permitting more rapid access to bleeding points
5. Reduced intraoperative blood loss and reduced blood transfusion requirements
6. Reduces need for coagulation allowing a well-defined tumor–brain interface
7. Provides optimum surgical access to midline structures
8. Significant reduction of cerebellar swelling
9. Better surgical teaching due to the nonrotated anatomical situation
10. Shorter surgical time
11. Minimizes the amount of cerebellar retraction needed to gain access to deeper structures during supracerebellar infratentorial approaches
to the pineal gland
12. Improved postoperative cranial nerve function compared to
other positions
13. Technically easier procedure than is possible in the prone position
For the neuroanesthesiologist:
1. Improved access to the tracheal tube, chest wall, and arms
2. Improved ventilation with lower airway pressure
3. Free diaphragmatic movements
4. Provides an unobstructed view of the patient’s face, enabling observation of motor responses to cranial nerve stimulation
5. Decreased intracranial pressure
6. In the event of cardiac arrest, cardiopulmonary resuscitation is easier, with decreased intrathoracic pressure permitting easier ventilation
VII. NEUROSURGERY
260
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
pressure, mean arterial pressure (MAP), mean pulmonary arterial pressure (PAP) and pulmonary wedge pressure,
and an increase in pulmonary and systemic vascular resistance.17 Sitting may also affect brain arterial and venous
pressure and alter the venous or arterial ratio with different blood distributions.18 Cerebral perfusion pressure (CPP)
decreases in the sitting position in nonanesthetized patients and could further decrease under anesthesia because of
vasodilation and impaired venous return. This can be further aggravated by jugular venous obstruction caused by
unfavorable head and neck position.
Sitting position has the potential for serious complications10,19–22 (Table 14.3). The increased risk of VAE, with
its most feared sequela of PAE, is by and large the most feared complication20–22 (discussed in detail later). Paying
meticulous attention during patient positioning is thus of paramount importance to prevent positioning related
complications. Prerequisite for a safe and routine adaption of the sitting position is an interdisciplinary dialogue
between neurosurgeons and neuroanesthesiologist considering the relative risk–benefit of sitting position surgery
for the individual patient, based on physical status and specific intracranial pathology, absence of contraindications
(Table 14.4), anticipation of potential complications; level of comfort, both with the procedure, and with each other
and careful perioperative monitoring.
Since its introduction, this patient position has been modified for neurosurgery to a modified semisitting or lounging position as it is used today to reduce the risk of an air embolism.1,4,5,24,25 This position aims to achieve a positive
venous pressure at the operation site by a combination of adjustments. The upper body and legs are elevated by
bending the operating table to a position in which the hip is flexed to a maximum of 90°. A 30° flexion of the knees is
maintained to avoid overstretching of the tendons and nerves of the leg. The patient’s head is flexed anteriorly and a
two-finger space between the sternal notch and the chin is left to avoid venous outflow obstruction. The inclination
of the whole operating table is then changed to a lower head and higher leg position, in which the legs of the patients
are as high as the vertex. Arms are supported to avoid traction of the shoulders; and all pressure points including
legs, arms, and heels are adequately padded.
The patient positioning should be done incrementally to avoid any hemodynamic instability. Positioning of the
head should ideally be performed under electrophysiologic neuromonitoring to minimize cervical cord compression. Finally, it must be ensured that the anesthesiologist has adequate access to the patient with minimum possible
disturbance to the surgical field.
TABLE 14.3 Potential Complications of the Sitting Position10,19–22
1.
2.
3.
4.
5.
6.
7.
8.
Increased incidence of venous air embolism and paradoxical air embolism
Hemodynamic instability, particularly if intravascular volume is inadequate
Increased incidence of pneumocephalus
Lingual and laryngeal trauma (including trauma from a TEE probe if used)
Quadriplegia due to excessive neck flexion
Macroglossia
Pressure points sores
Compressive peripheral neuropathy
TABLE 14.4 Contraindications to the Sitting Position8,22,23
Absolute
1. Patent ventriculoatrial shunt
2. Signs of cerebral ischemia when upright and awake
Relative
1. Right to left intracardiac shunt or other pulmonary-systemic shunt
2. Cervical spine instability
3. Severe cervical canal stenosis
4. Hemodynamic instability
5. Anesthesia or surgical team not familiar with the position
6. Extremes of age
7. Severe autonomic neuropathy
VII. NEUROSURGERY
PERIOPERATIVE MANAGEMENT OF PATIENTS FOR POSTERIOR FOSSA SURGERY
261
Anesthetic Management
During anesthetic induction, care should be taken to avoid hypotension, hypoxia, and hypercapnia with attendant
cerebral ischemia and brain stem herniation in view of low compliance of the posterior fossa.
Considering the choice of anesthetic drugs during posterior fossa craniotomies, total intravenous anesthesia has been reported as the most commonly employed anesthetic maintenance technique in recent studies.26
Anesthesia is usually maintained by using a continuous infusion of propofol (6–8 mg/kg/h) with supplemental
administration of opioids (either repetitive boluses of fentanyl or continuous infusion of remifentanil or sufentanil titrated to effect). Few studies report on the use of sevoflurane with a maximum concentration of 1 MAC
during anesthetic maintenance.5,6
The use of nitrous oxide (N2O) is the most controversial in posterior fossa interventions, especially in sitting craniotomies.27–38 During supratentorial craniotomy, N2O has potential advantages in terms of stable hemodynamics,28
good surgical conditions,28,29 reduction of awareness with recall, and use in neurologically and cardiovascularly “atrisk” patients.30,31 However, the classic adverse characteristics, such as unfavorable effects on intracranial dynamics,
expansion of gas-filled spaces, and postoperative nausea and vomiting (PONV) are often cited as reasons to avoid
N2O-based anesthetic regimen during posterior fossa surgery.32,33
Nitrous oxide has been hypothesized to convert a pneumocephalus into a tension pneumocephalus.34 However,
there appears to be no difference in the volume of intracranial gas postcraniotomy in patients who have received
N2O versus those who have had a nitrous-free anesthetic.35 In fact it may be advantageous to maintain anesthesia
with high-inspired concentrations of N2O until dural closure so that the rapid washout of N2O may actually decrease
the pneumocephalus when it is discontinued.36 It has been postulated that N2O equilibrates with the intracranial
air-containing cavity while the dura is open, such that after dural closure, no further volume expansion and/or significant ICP increase will occur.37 Hence, it is not necessary to discontinue N2O prior to dural closure for reasons of
avoiding expansion of intracranial air and increasing ICP.
Also of concern during posterior fossa surgery is the risk that N2O, being 34 times more soluble in blood than
nitrogen, can dramatically increase the size of venous air emboli. However, Losasso and colleagues38 found no evidence that N2O increased the risk, volume, or clinical consequences of VAE, if its administration is discontinued
immediately upon Doppler detection of VAE. Nonetheless, N2O administration in presence of VAE results in its
volume augmentation and intensifies the hemodynamic alterations thus allowing for earlier detection and, consequently, prompter treatment of VAE.
Although it is not rational then to omit N2O solely on the fear of a VAE or tension pneumocephalus, it is still
prudent to avoid it during a repeat craniotomy within 6–8 weeks after dural opening and stop its administration as
soon as an air embolus is suspected intraoperatively.33 The emetogenic effect of N2O, although undesirable after a
craniotomy, can be controlled with antiemetic prophylaxis.31
Respiratory Management
The polyvinyl endotracheal tube (ETT) may kink during posterior fossa surgery from overbending of the softened
tube (due to oral temperature) and neck flexion required to improve surgical access.39 Manual straightening of the
tube may be helpful to relieve kinking of ETT. In a recent report the placement of Berman intubating airway was
found helpful to relieve the kinking of the ETT in a prone patient.40 Nonetheless, most neuroanesthesiologist prefer
reinforced ETT to prevent kinking in view of varied patient positioning during posterior fossa craniotomies. A gap
of at least two-finger space should always be present between the chin and the chest, and head rotation should be
minimized.
Patients are mechanically ventilated to maintain either normocapnia5,6,25,41 or mild hypercapnia [to allow for a
change in end-tidal carbon dioxide (EtCO2) to be more prominent].1 A higher arterial partial pressure of carbon dioxide (PaCO2) level of about 35 mmHg (slightly greater than during neurosurgery in other positions) may be acceptable
in sitting position craniotomy because ICP is usually less in this position.
Fraction of inspired oxygen (FiO2) is usually maintained between 0.4 and 1.26 Nonetheless, when administering
anesthesia for operations involving risk of intrapulmonary right-to-left transmission, higher levels of FiO2 should be
maintained as hyperoxia may prevent or reduce blood flow through arteriovenous pathways bypassing the capillary
system when they are exercise induced.41 It, however, remains unknown whether FiO2 or oxygen tension specifically
regulates these recruited anastomoses or opens them indirectly.
VII. NEUROSURGERY
262
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
The use of positive end-expiratory pressure (PEEP) during sitting position craniotomies is controversial. PEEP
has been proposed to lower the incidence of VAE essentially by increasing the central venous pressure (CVP)42 and
has been found safe up to 10 cm H2O in terms of non-alteration of interatrial pressure difference.43 Nonetheless, VAE
has been shown to occur during release of PEEP and repositioning after sitting position surgery.44 Earlier studies
have documented that PEEP is potentially detrimental during sitting craniotomies as it does not decrease the incidence of VAE, impairs hemodynamic performance, and might predispose patients with a probe PFO to the risk of
PAE.45,46 However, current literature gives mixed results in the usage of PEEP with many groups using PEEP from 6
to 10 cm H2O1,25,47 while some avoid it all together.5 It is generally contraindicated in patients with PFO.4 However,
Ammirati et al. have established biphasic PEEP (7–10 cm H2O) in patients with proven PFO to increase the intrathoracic pressure.1
Use of spontaneous ventilation for the time period of tumor excision has been advocated by some authors to monitor the structural and functional integrity of vital brain stem structures but is no longer in vogue.48,49 During spontaneous ventilation, changes in the respiratory pattern would provide the surgeon with a warning signal for potential
damage of these structures; thus avoiding any iatrogenic injury. However, it is necessary to maintain adequate depth
of anesthesia to avoid coughing and patient movement.
Hemodynamic Management
In addition to the effects of anesthetic agents, patient’s cardiovascular system is exposed to the effects of gravity
with venous pooling of blood in lower extremities during sitting position craniotomy. Intraoperative VAE or cranial
nerve manipulation further adds to hemodynamic instability, thereby jeopardizing cerebral blood flow (CBF), especially in patients with disturbed autoregulation. Hence, any hemodynamic instability during induction and positioning should be avoided and aggressively managed. Normovolemia must be maintained at all times as dehydration
exacerbates the low venous pressure and increases the risk of VAE. To combat positioning related hypotension, prepositioning controlled fluid loading5,25,50 and use of antigravity devices3,5 have been advised. Fluid loading should be
done meticulously in patients with reduced cardiovascular reserves (e.g., elderly patients) taking into account their
existing intravascular volume status.
In the first randomized study focusing on fluid therapy in neurosurgical patients operated on in sitting position with a stroke volume–guided therapy, Lindroos et al. found that 6% hydroxyethyl starch (HES) boluses
resulted in 34% smaller infusion volume and less positive fluid balance than crystalloid while significantly
increasing cardiac and stroke volume indexes.5 Furthermore, no difference was observed in thromboelastometry coagulation analysis between Ringer’s acetate and HES groups. Authors thus suggested that use of stroke
volume–guided HES therapy might be advantageous during sitting craniotomies, especially in patients with
decreased brain compliance. Later, similar study was done by the same author group in patients undergoing
neurosurgery in prone position utilizing the same protocol of stroke volume–directed administration of HES
(130/0.4) and Ringer’s acetate.51 Although, the amount of HES needed for comparable hemodynamics was 24%
less, a slight disturbance in coagulation parameters was observed, and hence authors suggested caution while
using colloids in neurosurgical patients.
Use of intermittent sequential compression device on the lower extremities is another simple and effective method
to decrease intraoperative hypotensive episodes and improve cerebral oxygen saturation.52
With hypotension induced by the upright sitting position, both intracranial blood flow velocity53 and cerebral
oxygen saturation are reported to decrease.54 Arterial pressure should thus be maintained close to preinduction
values, to preserve cerebral perfusion and reduce any risk of cerebral injury or postoperative cognitive dysfunction.
Lindroos et al. have suggested maintaining a target MAP of 60 mmHg or higher at the brain level during sitting craniotomies.5 Unfortunately, a single targeted value of MAP may not suffice in all the patients. Moreover, a reported
CPP of 60 mmHg may vary from a true head-level value of 43–60 mmHg, depending on reference point, head-of-bed
elevation and height of the patient.55 Emphasis should thus be given on use of individualized targeted MAP with the
help of bedside autoregulation testing, to continuously adjust the “better MAP” to maintain CBF and brain oxygenation without increasing cerebral blood volume.
Ephedrine and phenylephrine are the most frequently used vasopressors to treat intraoperative hypotension during sitting craniotomies. However, when comparing both agents, cerebral oxygenation was found to decrease significantly after phenylephrine bolus treatment and remained unchanged after ephedrine bolus treatment, even though
MAP was significantly increased by both agents.56 Phenylephrine infusion has also been associated with cerebral
oxygen desaturation, possibly caused by cerebral vasoconstriction, despite preventing hypotension in the upright
position.57 Norepinephrine also negatively affects cerebral oxygenation.58
VII. NEUROSURGERY
PERIOPERATIVE MANAGEMENT OF PATIENTS FOR POSTERIOR FOSSA SURGERY
263
Intraoperatively, various frequent changes in cardiovascular responses including bradycardia, tachycardia,
hypotension, or hypertension and arrhythmia occur during surgical manipulation of the lower pons, upper
medulla, floor of the fourth ventricle, and the cranial nerve nuclei.59,60 Hence, drugs that would mask these
sentinel cardiovascular responses, including anticholinergic medications and/or long acting beta-adrenergic
blockers, should be avoided. The surgeon should be notified immediately, with most of these changes subsiding
immediately after the surgical stimulus is withdrawn and pharmacological treatment is generally not required.
Transcutaneous pacing should, however, be considered in high-risk patients in view of bradycardia and risk of
asystole during the surgery.
In a rare cause of bradycardia during posterior fossa surgery, Prabhakar et al. have reported reproducible bradycardia following hydrogen peroxide irrigation at the end of surgery, which resolved following aspiration of the
effervescent solution.61
Intraoperative Monitoring
During posterior fossa surgeries, patients are susceptible to intraoperative hemodynamic instability, blood loss,
cardiac arrhythmias, VAE, and specific positioning related complications. Hence, in addition to “routine” neuroanesthesia monitoring, such as electrocardiography (ECG), pulse oximetry, capnography, temperature, urine output,
invasive arterial blood pressure (ABP), arterial blood gases and CVP, additional specific VAE (discussed in detail
in VAE section) and neurophysiological monitoring, with minimal interruptions during positioning, is required to
increase the safety of the procedure.
Invasive arterial monitoring allows continuous ABP monitoring and repeated blood gas analysis. During sitting
craniotomies, the arterial line transducer should be located and zeroed at the level of tragus to estimate the CPP correctly.4,5,47,62 Central venous catheter is essential as it is helpful for aspirating air during VAE, in addition to monitoring CVP (discussed in detail in VAE section).
Electroencephalography-based monitors can be used to detect cerebral hypoperfusion as well as to determine the
depth of anesthesia, especially when spontaneous ventilation is planned during tumor resection. IONM techniques
including somatosensory evoked potentials (SSEPs); transcranial electrical motor evoked potentials; brain stem auditory evoked responses (BAERs); and spontaneous electromyography (EMG) offer a great tool for live monitoring of
the integrity of central nervous structures. Thus, any dysfunction can be identified early and prompt modification of
the surgical technique or operating conditions helps to avoid permanent structural damage.5,25,63 SSEPs help to monitor spinal cord ischemia (related to hypotension in the sitting position) and should be opted during neck positioning
whenever feasible to reduce the risk of midcervical flexion myelopathy.2,64 However, the incidence of false positives
during SSEP monitoring is very high, particularly for cases of brain stem monitoring.65
The vestibulocochlear nerve (CN VIII) and, to a greater extent, the auditory pathways (as they pass through the
brain stem) are especially at risk during CPA, posterior/middle fossa, or brain stem surgery. The CN VIII can be
damaged by several mechanisms, from vascular compromise to mechanical injury by stretch, compression, dissection, and heat injury. Additionally, cochlea itself can be significantly damaged during temporal bone drilling, by
noise, mechanical destruction, or infarction, and because of rupture, occlusion, or vasospasm of the internal auditory
artery. Intraoperative monitoring of CN VIII can be successfully achieved by live recording of the function of one of
its parts, using the BAERs, electrocochleography (ECochG), and compound nerve action potentials of the cochlear
nerve.66
The BAERs is the most widely used method for hearing preservation as it has high sensitivity and reliability to
detect cochlear nerve damage. Latency increase of wave V of 1.0 s and amplitude decrease of 50% are the most widely
used criteria to warn the surgeon about potential cranial nerve damage and thus encourage redirection of the operative plan of action.67 However, BAERs are prone to presenting false-positive results and there is a significant time
delay of several seconds to minutes to deliver reliable response of wave changes. In this interim, a permanent damage to the cochlear nerve could happen, preventing the surgical team from detecting and avoiding it. In comparison,
ECochG and direct stimulation of CN VIII are “near-field” techniques with shorter latency periods and provide
immediate feedback on the state of the auditory system.
Facial nerve injury is a complication of major concern after posterior fossa surgery due to severe negative
impact on patient’s quality of life. Continuous intraoperative facial nerve monitoring helps to minimize accidental
damage to the nerve during CPA surgery and skull base tumor surgery. Current standard facial nerve monitoring
modalities include direct electrical stimulation, free-running continuous EMG, and facial MEP. However, a lack of
standardization in electrode montage and stimulation parameters precludes a definite conclusion regarding the
best method of monitoring.68
VII. NEUROSURGERY
264
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
In addition to the seventh and the eighth nerve monitoring, lower cranial nerves (CN IX–XII) can be monitored similarly by EMG, using needle electrodes within their respective musculature.69 Use of these monitors requires modification of the anesthetic technique to minimize interference with the monitoring (discussed elsewhere in the book).
VENOUS AIR EMBOLISM
Neurosurgical procedures carry the highest risk of VAE among surgical procedures as the surgical site is always
at a higher level relative to the heart creating a negative venous pressure at the surgical wound level, irrespective
of the patient position. The risk exists for all kinds of neurosurgical procedures; however, its incidence is higher for
procedures requiring the patient to be in a sitting or semisitting position.1,6
The presence of numerous large, noncollapsible venous channels in the surgical field—especially involving cervical procedures and craniotomies that breach the dural sinuses—leads to air entrainment in the venous system. Air
entrainment usually occurs through the diploic veins tethered patent by their bony and dural attachment or through
open venous sinuses. High degree of vascularity of lesions as in tumors or malformations or compromised vessel
(like in trauma) further increases the risk. In a recent study, surgical tumor resection was found to have a higher incidence of VAE opposed to decompressive surgery alone.6
Incidence
The reported incidence of VAE varies widely in literature, ranging from 1.6% to 76%.6,7,9,20,23,25,70–73 The variability exists in view of different monitoring modalities used for its detection and different definitions for VAE and its
associated hypotension. Nonetheless, most detected VAE are reported not to be clinically relevant. In a recent metaanalysis of neurosurgical procedures performed in the sitting position, Fathi et al. reported an overall incidence of
VAE of 39% and 12% for posterior fossa and cervical procedures, respectively.7
In their retrospective review of 600 cases, Ganslandt et al. found an overall incidence of VAE in 19% of all patients,
whereas the rate of severe complications associated with VAE, such as a decline of arterial partial pressure of oxygen
(PaO2) or a drop of blood pressure, was only 3.3% in all patients.6 However, in only 0.5% of cases, a termination of the
surgical procedure became necessary, which were operated on uneventfully afterward. In all other cases, the cause
of air embolism could be found and eliminated during surgery.
Considering pediatric patients undergoing surgery in sitting position, Harrison and coworkers reported a low
incidence of VAE (9.3%) and VAE associated hypotension (in only 2% of 407 operations) without any perioperative morbidity or mortality directly attributed to it.72 The age of the patients ranged from 6 weeks to 17 years with
a median age of 5 years. When compared with adults, Bithal et al. have reported an equal incidence of capnometry
detected VAE (28% versus 22%) and VAE with consequent hypotension (37% versus 33%) in adults and children
undergoing neurosurgery in the sitting position.73 Venous air embolism during neurosurgery is not exclusive to the
sitting position and has frequently been reported in the prone and supine positions.10,14,16
Pathophysiology
The severity of symptoms resulting from VAE varies according to the amount and rate at which air enters the
venous system and the end location of the air bubble. Local factors such as the presence of venous plexus may
be equally important. Patients may be asymptomatic or may have complete cardiovascular collapse. In adults,
approximately 100 mL of air in the venous system may trigger clinical manifestations. The adult lethal volume
has been described as 3–4 mL/kg.19,74 Small built and pediatric patients, however, poorly tolerate similar volumes of air. Furthermore, closer the vein of entrainment is to the right heart, the smaller the required lethal
volume.
Several pathophysiologic pathways may be elucidated after a substantive volume of air entrainment. The pathway
manifested is greatly dependent on the volume of air accumulated within the right ventricle (RV). If entrainment is slow,
the heart may be able to withstand large quantities of air despite entrainment over a prolonged period. Continuous
entrainment of low volume air will lead to breaking up into microbubbles in turbulent flows. These microbubbles on
reaching the pulmonary vasculature, cause increase in pulmonary vascular resistance by pulmonary vasoconstriction
and mechanical obstruction. Obstruction of the pulmonary circulation results in an increase in physiological dead space.
Thus EtCO2 falls and arterial PaCO2 rises. Lack of treatment at this stage leads to hypoxia. Further, air entrainment into
VII. NEUROSURGERY
VENOUS AIR EMBOLISM
265
the pulmonary circulation causes gaps between endothelial cells and consequential mediator release, which in turn can
lead to platelet or complement activation, thus facilitating pulmonary edema.
Intravascular air also induces direct binding of platelets to air bubbles, forming air–platelet conglomerates. The
formation of platelet–air–clots affects air reabsorption and thus is likely to contribute to a sustained increase in right
ventricular afterload, and prolonged occlusion of pulmonary arterioles or capillaries which might aggravate cardiopulmonary complications of VAE. In addition, the microbubbles formed due to turbulent flow in the circulation
precipitate platelet aggregation and the release of platelet activator inhibitors, resulting in platelet dysfunction that
contributes to the bleeding diathesis.50
If the embolism is large (approximately 5 mL/kg), a gas air-lock scenario immediately occurs. There may be complete outflow obstruction from the RV. This rapidly leads to right-sided heart failure and immediate cardiovascular
collapse. Moderate volume of air goes through to the right atrium (RA), RV, and finally into the pulmonary circulation causing an increase in pulmonary vascular resistance and pulmonary hypertension. This results in elevated
right heart pressure and the risk of PAE.
Large VAEs also result in an increase in pulmonary shunt. Under certain conditions, air bubbles may pass through
lung capillaries and reach the pulmonary veins. However, the transpulmonary passage of air into the systemic circulation is a rare clinical event during VAE as lung is a very good filter.
Clinical Presentation of Venous Air Embolism
Venous air embolism may produce a broad array of catastrophic cardiovascular, pulmonary, neurological, and
coagulation sequela. Elevated right atrial pressure results in decreased venous return, elevated CVP measurements,
jugular venous distension followed by hypotension and shock. ECG demonstrates peaking of “P” waves, tachyarrythmias, right heart strain pattern, and new onset ST-T changes. Massive VAE or a large embolus obstructing the
outlet of the RV can result in a sudden onset right heart failure and cardiac arrest. The classic finding of a “mill wheel
murmur” indicates a near fatal entity.
After an episode of VAE, the progressive decrease in lung perfusion leads to an increased physiological dead space,
which is reflected by a sudden decrease in EtCO2 and an increase in EtN2. With resolution of VAE, the EtCO2 returns to
normal, often with an overshoot due to the raised arterial PaCO2. Other pulmonary signs of VAE include wheeze and/
or crepitations. An arterial blood gas would reveal respiratory acidosis with hypercarbia and hypoxia.25,47
Neurological manifestations include cerebral hypoperfusion as a result of shock and stroke in the event of a PAE.
Postoperative mental status changes, presence of focal deficits or seizures should raise the suspicion of cerebral ischemia secondary to cerebral air embolism in at-risk individuals.
Coagulation abnormalities are under-recognized complications of VAE. The interaction of Factor VIII with the prostaglandin system and possibly other blood/tissue factors initiate a coagulation derangement.75 VAE is also associated with
a significant decrease in platelet count. Nonlethal VAE induces an almost one-third decrease in platelet count, and even
grade 1 VAE evokes marked thrombocytopenia.50 There is a relationship between the grade of VAE and the severity of
thrombocytopenia, with a greater fall in platelet count associated with a higher grade of VAE. Thus, patients with a major
VAE who already have bleeding disorders or preexisting thrombocytopenia may be particularly at risk for perioperative
bleeding. Based on these findings, it appears prudent to reassess blood coagulation and platelet count intraoperatively,
following VAE. Near-patient tests such as rotational thrombelastometry and impedance aggregometry allows immediate
and sophisticated analysis of platelet function and whole blood coagulation.76
Monitoring
For VAE detection, it is recommended to use more than one monitoring modality during posterior fossa surgery
(especially during sitting position), as no single monitor is completely reliable. In addition, timely anticipation of
VAE during critical portions of a procedure is as vital to patient well-being as any detection device. In order of
decreasing sensitivity, detection of VAE intraoperatively may be done through TEE, precordial Doppler (PCD), pulmonary artery catheter, EtCO2 by capnography, EtN2 by mass spectrometry, right heart catheterization, and esophageal stethoscope (Table 14.5).
Although intracardiac transvenous echocardiography is reportedly superior to TEE, it has not yet become
routine.81 Currently, TEE is considered the gold standard monitoring modality to detect VAE as well as PAE.4,25,41
In clinical studies, the incidence of VAE in patients monitored with TEE was significantly higher compared to
the patients monitored with PCD (25.6% versus 9.4%).6 However, TEE is too sensitive and not necessarily specific for VAE.25 With use of continuous intraoperative TEE, the risk of false positive increases, which can be
VII. NEUROSURGERY
VII. NEUROSURGERY
Placement of such an invasive device
should have other cardiac indication and
not risk of VAE alone
Pulmonary artery High (0.25 mL/kg)
catheter
Basic anesthesia monitor used
intraoperatively in all cases
Probe is placed at the of SVC-RA junction
on the chest wall (corresponds with the
third or fourth intercostal space just to
the right of the sternum)
Probe should be placed when the patient
is already in the final surgical position
Appropriate placement should be
verified by injecting a small bolus of
saline through the RAC, which should
cause detectable changes in sound78
High (0.05 mL/kg)
Precordial
Doppler
Moderate (0.15 mL/kg)
The TEE probe is placed with the patient
in supine position and is then readjusted
so that the probe is in the mid-esophagus
for mid-esophageal four-chamber view
or mid-esophageal bicaval view visible
on the TEE monitor, after the patient is
put in the semisitting position (Fig. 14.1)
Excessive neck flexion should be avoided
A bolus of cold saline solution is injected
to test whether it is shown as a reflex
pattern on the monitor and to verify that
the system is adjusted correctly
Highest
(0.02 mL/kg)7
Transoesophageal
echocardiography
End-tidal CO2
Technical Considerations
Sensitivity
Modality
TABLE 14.5 Detection of Vascular Air Embolism19,21,47
Most convenient
Semiquantitative
VAE detected by capnography is always
clinically significant79
Noninvasive
Offers continuous objective monitoring
Continuous monitoring
Objective
Semiquantitative
The pressure gradient between the left
and right atria may help in assessing the
risk of PAE
Considered standard of care for VAE
(along with EtCO2 monitoring)
Noninvasive
High sensitivity (small emboli can be
detected)
Detects air before it enters the
pulmonary circulation
Gold standard for VAE monitoring
Most sensitive
Objective monitoring of VAE
Gives information about the size of the
embolus and whether it is ongoing
Useful for assisting with optimal
positioning of a RAC
Also useful for detection of a PFO or
other possible pulmonary-systemic
shunt before placing the patient in the
sitting position
Detects PAE that may result in ischemic
cerebral complications (observation of
air appearing in the left atrium is by far
the best way of confirming PAE)
Advantages
Less sensitive
Nonspecific for air
Not reliable in the event of systemic hypotension,
tachypnea, and patients with chronic obstructive
pulmonary disease
An acute reduction in EtCO2 may occur as an artifact
during partial blockade in the gas sampling line80
Less sensitive
Invasive
Nonspecific for air
Of limited ability to withdraw air because of its fixed
distance and small orifice
Use is restricted to those patients who have
significant comorbidities that may benefit from its
use as a monitoring tool for cardiac output or mixed
venous saturation
Placement done for optimal air aspiration may
not allow pulmonary capillary wedge pressure
measurement
Subjective
Nonquantitative
Interference by diathermy
False negative results in approximately 10% of cases
where air does not pass beneath ultrasonic beam
False positive in presence of mannitol crystals
Continuous embolism is easily missed, as the ear
detects changes in sound more readily than an
“abnormal” sound
Difficult to position the probe accurately in obese
patients, patients with chest wall deformity, and
during prone and lateral patient positioning
Invasive
Expensive
Requires operator expertise
Requires constant vigilance
Nonquatitative
Can cause laryngeal and oral trauma
Rapid intravenous infusion, however, can be confused
with VAE, but differentiation is easily done by slowing
the infusion
Almost too sensitive (detecting virtually any amount
of air in the circulation, most leading to no adverse
sequela)
Despite high sensitivity, PFO cannot be ruled out 100%
and it can even fail to detect some VAE incidents77
Disadvantages
266
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
Low (1.7 mL /kg)
Air in the RV produces a loud churning
sound called the “mill wheel murmur”
Not readily available
Not useful if N2O is used as a carrier gas
The presence of EtN2 may also indicate air clearance
from the pulmonary circulation prematurely
Routinely used
Noninvasive
Useful in confirming VAE in equivocal
cases
Routinely used during all craniotomies
Late changes
Associated with signs of cardiovascular collapse
Of little clinical use
Late changes
Late indicator
Has therapeutic value during VAE19,20,71 Complications associated with central venous
cannulation (depending upon the route of insertion)
Most specific monitoring modality
EtCO2, end-tidal carbon dioxide; EtN2, end-tidal nitrogen; N2O, nitrous oxide; PaCO2, arterial partial pressure of carbon dioxide; PAE, paradoxical air embolism; PaO2, arterial partial pressure of oxygen;
PFO, patent foramen ovale; RA, right atrium; RAC, right atrial catheter; RV, right ventricle; SVC, superior vena cava; TEE, transesophageal echocardiography; VAE, venous air embolism.
Esophageal
stethoscope
ST–T changes are noted first, followed
by supraventricular and ventricular
tachyarrythmias
An increase in arterial PaCO2 and a
reduction in arterial PaO2, associated
with no change in ventilation, may be
due to VAE
Low
Oxygen
saturation
Electrocardiogram Low (1.25 mL/kg)
Accurate placement of a right atrial
catheter tip is quintessential for
aspirating air
Catheter tip should be readjusted and
confirmed after final patient positioning
If appropriately
positioned, then
aspiration of air will
confirm VAE
The sensitivity compares to or exceeds
that of EtCO2 during large-bolus VAE
but may be less sensitive during slower
entrained volumes
Air aspiration
via a central vein
catheter
End-tidal N2 by
Moderate (0.5 mL/kg)
mass spectrometry
VENOUS AIR EMBOLISM
VII. NEUROSURGERY
267
268
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
FIGURE 14.1 Transesophageal echocardiogram showing entrained air emboli (white arrow) in the right atrium.
interpreted as the cases found to have a positive TEE and negative air aspiration and/or no instability in vital
signs. PCD is the most sensitive noninvasive monitor for VAE and is preferred over TEE in many centers1,3,47
and during emergency cases.6
Capnography is an efficient monitoring technique for detection of all episodes of VAE severe enough to cause
hemodynamic disturbances.79 On the basis of EtCO2 monitoring, VAE has been defined variedly in the literature
with some authors considering that a VAE episode has occurred when EtCO2 decreases suddenly by more than
3 mmHg6 whereas others use a value of more than 5 mmHg, without a change in minute volume ventilation.21,46,73,79
The right atrial catheter (RAC) can be inserted via the antecubital, the subclavian, or internal jugular veins.
Accurate placement of a RAC is quintessential for aspirating air from the circulation should an air embolism
occur. The ideal location for multiorifice catheters is 2 cm below the junction of the superior vena cava (SVC)
and RA at an inclination of 80° for maximal efficacy (up to 80%) in aspirating air.82 A single-orificed catheter
gives a maximum yield of 45–50% aspiration when the tip is positioned 3.0 cm above the SVC and arterial chamber junction.82
Appropriate placement of RAC can be confirmed either by plain chest X-ray, real-time X-ray imaging, intravascular
ECG (point of large negative P complex),82 TEE-guided placement83 or by withdrawing the catheter after eliciting right
ventricular waveform on pressure transducer. The relationship of the heart to the thoracic contents changes when a patient
is changed from the supine to the sitting position, thereby resulting in migration of the catheter tip. Hence, catheter tip
should be readjusted and confirmed after final patient positioning.
Grading of Venous Air Embolism
Literature till date have several comparable scales for VAE grading, which differ in their definition and details,
and hence cause confusion when comparing the data in systematic reviews4, 20,25,70,84 (Table 14.6). New scales should
be introduced sparingly and preferably after validation for either an outcome or a management decision.
Paradoxical Air Embolism
PAE occurs when VAE passes into the systemic arterial circulation, for example, through PFO. Air in the systemic
circulation can cause catastrophic consequences. Blockage of coronary arteries can cause myocardial infarction of
various degrees and that in the cerebral vessels may cause cerebral infarction.
The incidence of PFO in the general population seems to be relatively high (about 10–30%).85 Hence, preoperative
screening to determine the presence of a right-to-left cardiac shunt should be performed in all patients who are to be operated in a sitting position.4–6 Few centers, however, perform PFO search only after anesthesia induction.1,47 A PFO has been
detected in up to 5–33% of the neurosurgical patients while the reported rate of clinical and TEE detected PAE is between
0% and 14%.7
VII. NEUROSURGERY
269
VENOUS AIR EMBOLISM
TABLE 14.6 Grading of Venous Air Embolism
Traditional According to
Volume of Air84
Lobato VAE Grading70
Tübingen VAE Grading4
Girard Scale20
Jadik Scale25
Grade I: PCD changes only
Grade 0: No air bubbles
visible, no air embolism
Grade 1: Positive
PCD signal without
hemodynamic alterations
Minor clinical
VAE: Positive TEE
accompanied by an
EtCO2 decrease >3 mmHg
Moderate VAE (10–50 mL):
Grade II: PCD changes
t I n addition to detection
plus a decrease in EtCO2
by PCD and TEE, there is >3 mmHg
a fall in EtCO2 and a rise
in PAP
t U
sually accompanied by
a sympathetic response,
and heart rate and ABP
often increase
Grade I: Air bubbles
visible on TEE
Grade 2: Positive PCD
signal + increase in systolic
PAP >5 mmHg and/
or a decrease of EtCO2
≥3 mmHg
Moderate clinical VAE:
Positive TEE with ABP
decrease or heart rate
increase
Large VAE (>50 mL):
Grade III: PCD changes and
t M
ajor changes are
a decrease in systolic blood
detected on all monitors pressure > l0% of baseline
t D
ysrhythmia,
tachycardia, bradycardia,
hypotension, right
ventricular failure, and
cardiac arrest may occur
Grade II: Air bubbles
visible on TEE with
decrease of EtCO2 ≤3
mmHg
Grade 3: ABP decreases
>20% or >20% increase
in heart rate, plus one
positive “grade 2”
criterion
Severe clinical VAE:
Positive TEE and
decrease ABP >40%
or heart rate increase>
40%; including
situations requiring
cardiopulmonary
resuscitation
Grade III: Air bubbles
visible on TEE with
decrease of EtCO2 >3
mmHg
Grade 4: Sudden ABP
decrease of at least 40%
or a 40% increase in heart
rate in the presence of at
least one positive “grade
2” criterion
Grade IV: Air bubbles
visible on TEE with
decrease of EtCO2 >3 mm
Hg and decrease of MAP
≥20% or increase of heart
rate ≥40% (or both)
Grade 5: Cardio circulatory
collapse in the presence of
at least one positive “grade
2” criterion
Small VAE (<10 mL):
t D
etected with PCD or
TEE
t N
o changes in EtCO2,
oxygen saturation, or
PAP
Grade IV: PCD changes
and cardiovascular collapse
Grade V: VAE causing
arrhythmia with
hemodynamic instability
requiring cardiopulmonary
resuscitation
ABP, arterial blood pressure; EtCO2, end-tidal carbon dioxide; MAP, mean arterial pressure; PAP, pulmonary arterial pressure; PCD, precordial doppler;
TEE, transesophageal echocardiography; VAE, venous air embolism.
Paradoxical embolism can occur even in the absence of a PFO, commonly via intrapulmonary transmission
of air.41 Flow across a PFO is more typically functional and therefore dependent on relative pressures in the two
atria. Right-to-left interatrial shunting may occur only when the pressure in the RA exceeds the pressure in the
left atrium (minimum pressure gradient of 5 mmHg), allowing paradoxical embolism of thrombus.86
Usually left atrial pressure is slightly higher than RA pressure, but the opposite may occur transiently during part
of the normal respiratory cycle,87 after release of PEEP,44 after about half an hour in upright sitting position86 and
during any episode of VAE and hypovolemia.84 However, in a study done in anesthetized neurosurgical patients
(positioned both supine and seated prior to operation), Zasslow et al. concluded that levels of PEEP up to 10 cm H2O
does not alter the interatrial pressure difference in seated neurosurgical patients, and, therefore, would not predispose these patients to PAE.43
VII. NEUROSURGERY
270
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
For the detection of right-to-left shunting across a PFO, agitated saline contrast medium is typically injected into a
peripheral vein during the strain phase of the valsalva maneuver (or coughing) and the atrial septum is imaged during the release phase of this maneuver. Currently, semi-invasive bubble contrast TEE is accepted as the gold standard
to detect right-to-left shunt across a PFO, but the use of sedation makes the performance of the valsalva maneuver
more difficult along with incomplete glottis closure.
Agitated saline is generated by mixing 9 mL of 0.9% saline with approximately 0.75 mL of the patient’s venous
blood and 0.25 mL of air, which is then agitated repeatedly through two 10 mL syringes connected via a 3-way stopcock. The size of the PFO has been categorized (somewhat arbitrarily) according to the amount of agitated venous
saline contrast bubble passage at rest or with the valsalva maneuver within the first three to five cardiac cycles: small
PFO ≤ 20 bubbles, moderate PFO = 20–50 bubbles, and large PFO ≥50 bubbles.88 Any cause of right-to-left shunt,
however, including atrial or ventricular septal defect, sinus venous defect, or intrapulmonary arteriovenous malformation, can lead to a positive bubble study, thus lowering the specificity of this test.
Other imaging modalities utilizing contrast such as second-harmonic transthoracic echocardiogram with a valsalva maneuver, transcranial Doppler sonography, cardiac computed tomography (CT), and magnetic resonance
imaging have been shown to have similar sensitivity and specificity in detecting a PFO when compared with TEE.88
However, TEE has an added advantage of confidently measuring PFO size and morphologic details, and also assisting in PFO closure device deployment.
Many neurosurgeons consider PFO an absolute contraindication for neurosurgery in the semisitting position because
of the high risk of PAE.25 However, there is no hard scientific evidence to prove a significantly higher risk for these
patients.4,89 Sitting position cannot always be avoided during posterior fossa surgeries, as some neurosurgeons feel
uncomfortable with the horizontal position for certain surgical procedures (surgeries of large and vascularized tumors in
the posterior cranial fossa) and are prepared to accept the increased risk for PAE.4,10 Feigl et al. have studied the incidence
of VAE in a retrospective study of 52 patients who had PFO and underwent a neurosurgical procedure in a semisitting
position with meticulous specific monitoring (TEE with intermittent jugular compression to detect any bleeding and
evoked potentials).4 In 29 patients, a total of 63 events occurred during which air bubbles appeared in the TEE, and only
1 patient experienced clinically relevant VAE without any clinical consequence.
Despite clinical studies proving safety of semisitting position surgery in the presence of PFO, the potential risk
of an air embolism remains and should by no means be underestimated. Percutaneous PFO closure using dedicated
devices under intracardiac echocardiography guidance involves low risk with a high success rate and should be
considered prior to sitting position neurosurgery.7,90
Management of Venous Air Embolism
The optimum management of VAE includes prevention and prompt recognition of this event to avoid or minimize
the serious complications. Preoperative risk evaluation; intraoperative real-time monitoring; adequate hemostasis;
strict adherence to a standardized protocol; and a collaborative approach by neurosurgeons, neuroanesthesiologists,
and cardiologists are prerequisites to minimize the risk of VAE and its complications.25
The most important measure to prevent VAE is to avoid injury to the venous system in the first place. Special care
has to be taken during the craniectomy, where an opening in the venous system might be difficult to close because
of the limited access at that stage of the surgery. Careful preoperative study of the cranial CT scan is mandatory for
the neurosurgeon to be aware of and prepared for the quick closure of emissary veins. Burr holes are not made anywhere close to the transverse or sigmoid sinus. Any necessary exposure of the transverse or sigmoid sinus is always
performed as the last step to ensure an optimal access in case of an injury to the venous system.
Other preventive measures include optimal intravascular volume loading, avoiding drugs that dilate the venous
capacitance vessels, use of antigravity devices, and the final positioning of the patient with the toes above the heart.
Application of PEEP to increase CVP to prevent air entrainment is controversial (discussed earlier in Anesthetic
Management).
Once diagnosed, the management of VAE is generally supportive. Treatment requires simultaneous management
by both neurosurgeon and neuroanesthesiologist to stop air entrainment and reduce the size of embolism. Immediate
measures taken by neuroanesthesiologist include alerting the surgeon, administering 100% oxygen, ceasing N2O
administration, IV fluids to increase venous pressure, and aspiration of air from the RAC.19,84
Myocardial perfusion is optimized with IV fluid loading and a vasopressor, if required to augment blood pressure.
The neurosurgeon should immediately flood the surgical field with moist cotton strips or a veil of irrigation fluid using a
pressurized irrigation system to prevent further air entrainment, and lower the head if possible. Any suspected air entry
point is identified and closed. The possible sources of air emboli include the muscles, in the diploe of the bones, and along
VII. NEUROSURGERY
POSTOPERATIVE MANAGEMENT
271
the venous channels, especially around the draining intracranial sinuses. The open veins are coagulated or plugged with
either moist cotton pledget, gelatin foam, or fibrin glue and open diploic channels may be sealed with bone wax.
If a RAC is in place, attempts should be made to aspirate air from RA as it is probably the only management strategy with demonstrated clinical efficacy.19 Two large series of patients have found a success rate of 43–52%.20,71
During craniectomies, bilateral compression of the jugular veins has been described to increase the intracranial
venous pressure quickly, thereby checking for a potential venous leak and in case of a VAE to prevent air bubbles
from reaching the RV via the noncompressed jugular vein.91 In their study in patients with known PFO and operated
on semisitting position, Feigl et al. employed frequent bilateral jugular compression for early detection of any bleeding that could imply venous air entrance.4 However, compression of the external jugular vein for anterior scalp procedures is still logical, but the majority of neck and scalp incidences of air embolism occur via the posterior venous
complexes.19 In addition, bilateral compression of the jugular veins also increases ICP, decreases CBF, and increases
the risk of carotid artery atheromatous plaque rupture because of the simultaneous compression of the carotid artery.
Other complications include severe bradycardia because of carotid sinus stimulation and venous engorgement leading to cerebral edema. Hence, it may only be considered (with utmost caution) on an emergent basis situations when
high volume and rapid entrainment of air occurs.
Some authors recommend immediate lowering of the operative site to below heart level if possible thereby preventing air entry into the pulmonary artery based on the work of Durant et al. (i.e., placing the patient in Trendelenburg
and left lateral decubitus position also known as Durant’s maneuver).92,93 This positioning is proposed to allow the
entrapped air in the heart to be stabilized within the apex of the ventricle. At present, however, there is little scientific
evidence to support special patient positioning as a means to enhance air dispersion. Moving the patient into left
lateral decubitus position obscures the surgeon’s view of the operative field and also allows a very grave potential
for contamination of the operative field. Moreover, the lateral position would interrupt or delay cardiac massage
and vasopressor therapy that are evidence-based interventions. In the event of massive VAE with cardiovascular
collapse, immediate initiation of chest compression can result in the breaking down of a large air bubble obstructing
the right ventricular outflow tract with return of spontaneous circulation.
In patients who survive the initial insult of a left-sided air embolism, hyperbaric oxygen therapy has shown some
benefit in reversing neurologic deficits.94 High oxygen tension promotes the absorption of nitrogen from the bubble
and the elevated ambient pressure reduces the size of the bubbles in accordance with Boyle’s law.
POSTOPERATIVE MANAGEMENT
Extubation depends on the preoperative neurological condition of the patient and the intraoperative course. In a
neurologically intact patient with an uneventful surgery, smooth emergence and extubation is feasible, with the patient
monitored postoperatively in the neurosurgical ICU for signs of neurological deterioration.95 Caution should, however,
be taken in patients with poor physical status undergoing vascular surgery and long procedures with potential significant
fluid shifts. Patients who have had small intraoperative VAE or PAE can also be extubated at the end of surgery.
Patients with symptomatic VAE, who have been managed adequately, tolerate spontaneous ventilation and extubation well. Ganslandt et al. found no difference in the rate of immediate postoperative extubation, postoperative
duration of ventilation, length of ICU stay, length of hospital stay, reoperation rate, and in-hospital mortality in
patients with and without VAE operated upon in sitting position for different posterior fossa and cervical spine
pathologies.6 However, in patients with clinical evidence of PAE, the management decision is more difficult and
prolonged ventilation and hyperbaric oxygenation may be required.
Delayed emergence after posterior fossa surgery is not uncommon and multifactorial. Tension pneumocephalus,
air embolism, and intracranial hemorrhage have been cited as reasons for nonreversal of patients after posterior fossa
craniotomies.14,96–98 Patients with preoperative bulbar dysfunction and extensive intraoperative dissection, particularly in the floor of the fourth ventricle and near the brain stem, may precipitate central respiratory dysfunction and
hence, may require elective ventilation.95
ICP monitoring should be considered if postoperative ventilation is required because hydrocephalus remains
a risk. Postoperative hypertension should be carefully managed to avoid operative site hematoma. Postoperative
pain and PONV are important considerations after posterior fossa surgery. Infratentorial procedures are associated
with severe postoperative pain (both at rest and with movement), due to extensive muscle damage from resection
and reflection of the temporalis and posterior cervical muscles and subsequent spasm.99,100 Patients are also at high
risk of PONV because of the proximity of the vomiting center to the surgical site and postoperative use of opioids to
control the pain.
VII. NEUROSURGERY
272
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
COMPLICATIONS
Posterior cranial fossa surgery can be performed in most patients with acceptable morbidity and mortality.
Flexman et al. have reported that infratentorial craniotomy is associated with an increased risk of postoperative
respiratory failure and death when compared with supratentorial craniotomy, and death is the most important contributor to the elevated risk experienced by this population.101
Dubey et al. reported an overall complication rate of 31.8% in their 10-year retrospective study of 500 patients who
underwent posterior fossa surgery.102 CSF leaks were the most frequently encountered, presenting in 13% patients
followed by meningitis (9.2%), wound infection (7%), and cranial nerve palsies (4.8%). Other complications that were
observed to develop almost exclusively in patients undergoing cerebellar parenchymal tumor resection included
cerebellar edema (5%), hydrocephalus (4.6%), cerebellar hematoma (3%), and cerebellar mutism (1.2%). The overall
mortality rate related to surgery was 2.6% occurring in 13 patients.
Venous air embolism and its sequelae PAE are the most feared complications during posterior fossa surgery
(discussed in detail earlier). Other complications commonly associated with the sitting position include hemodynamic instability (concerning patients especially at extreme ages), pneumocephalus and its complication tension pneumocephalus, peripheral neuropathies, macroglossia, subdural hematoma, central cord syndrome, and
quadriplegia.8,14,72,98,103–106
Complications associated with prone positioning in neurosurgery includes hemodynamic instability; poor access
to patient’s airway; macroglossia risking post extubation airway obstruction; brachial plexus injuries; axillary, ulnar,
and radial neuropathies; retinal ischemia from orbital compression; inferior vena caval compression; pressure sores,
VAE; and cervical cord compression with quadriplegia.14,107 Rath et al. compared two groups of patients who were
either operated in the sitting position or in the prone position with regard to intra and postoperative complications.
While the incidence of VAE was higher in the sitting position (15.2 vs. 1.4%) the incidences of postoperative complications were equal in both groups.14
Depending on the location of the lesions and the surgical approaches, the postoperative cranial nerve dysfunction
may involve CN III to XII, which may be temporary or permanent. Lower cranial nerve dysfunction can result in
increased risk of reintubation or failure to wean from mechanical ventilation. Hence, patients may require feeding
tube placement and a tracheostomy to prevent aspiration pneumonia, until they sufficiently recover function.
Supratentorial pneumocephalus is a known complication of posterior fossa neurosurgery with incidence varying
from 57% to 73% in horizontal positions to 100% in sitting position.103,104 In recently published studies on sitting craniotomy, pneumocephalus was found to cause transient postoperative lethargy with a 3.7% incidence in one study47
and 31% in the other.25 For tension pneumocephalus, immediate aspiration of air with twist drill burr hole evacuation is required to prevent rapid and irreversible neurological deterioration.97,98 High-flow oxygen is recommended
for less severe cases.
Macroglossia after neurosurgical procedures in sitting, prone, and lateral positions is presumed to be caused by an
extreme neck flexion (often employed to increase surgical access) and a resultant bilateral lingual vein thrombosis,
lymphatic obstruction, or arterial compromise.108,109 Macroglossia may occur in conditions without neck flexion due
to abnormal brain stem signaling.109
Quadriplegia is a rare but potentially disastrous complication of posterior fossa surgery that can be caused either
by prolonged focal pressure on the spinal cord secondary to the acute flexion of the head in the sitting position or by
ischemic damage to the spinal cord during episodes of significant hypotension.23
ABBREVIATIONS
ABP Arterial blood pressure
BAERs Brain stem auditory evoked responses
CBF Cerebral blood flow
CN VIII Vestibulocochlear nerve
CPA Cerebellopontine angle
CPP Cerebral perfusion pressure
CSF Cerebrospinal fluid
CT Computed tomography
CVP Central venous pressure
ECG Electrocardiography
ECochG Electrocochleography
EMG Electromyography
EtCO2 End-tidal carbon dioxide
VII. NEUROSURGERY
REFERENCES
273
EtN2 End-tidal nitrogen
ETT Endotracheal tube
FiO2 Fraction of inspired oxygen
HES Hydroxyethyl starch
ICP Intracranial pressure
ICU Intensive care unit
IONM Intraoperative neurophysiological monitoring
IV Intravenous
MAP Mean arterial pressure
MEPs Motor evoked potentials
N2O Nitrous oxide
PaCO2 Arterial partial pressure of carbon dioxide
PAE Paradoxical air embolism
PaO2 Arterial partial pressure of oxygen
PAP Pulmonary arterial pressure
PCD Precordial Doppler
PEEP Positive end-expiratory pressure
PFO Patent foramen ovale
PONV Postoperative nausea and vomiting
RA Right atrium
RAC Right atrial catheter
RV Right ventricle
SSEPs Somatosensory evoked potentials
SVC Superior vena cava
TEE Transesophageal echocardiography
VAE Venous air embolism
References
1. Ammirati M, Lamki TT, Shaw AB, Forde B, Nakano I, Mani M. A streamlined protocol for the use of the semi-sitting position in neurosurgery: a report on 48 consecutive procedures. J Clin Neurosci 2013;20(1):32–4.
2. Kumar A, Bhattacharjee S, Sahu BP, Kulkarni D. Brain death after concorde positioning for supracerebellar-infratentorial approach: unanswered
questions and lessons learned. Surg Neurol Int 2012;3:37.
3. Jürgens S, Basu S. The sitting position in anaesthesia: old and new. European J Anaesthesiol 2014;31(5):285–7.
4. Feigl GC, Decker K, Wurms M, Krischek B, Ritz R, Unertl K, Tatagiba M. Neurosurgical procedures in the semisitting position: evaluation of
the risk of paradoxical venous air embolism in patients with a patent foramen ovale. World Neurosurg 2014;81(1):159–64.
5. Lindroos AC, Niiya T, Silvasti-Lundell M, Randell T, Hernesniemi J, Niemi TT. Stroke volume-directed administration of hydroxyethyl
starch or Ringer’s acetate in sitting position during craniotomy. Acta Anaesthesiol Scand 2013;57(6):729–36.
6. Ganslandt O, Merkel A, Schmitt H, Tzabazis A, Buchfelder M, Eyupoglu I, Muenster T. The sitting position in neurosurgery: indications,
complications and results. a single institution experience of 600 cases. Acta Neurochirur (Wien) 2013;155(10):1887–93.
7. Fathi AR, Eshtehardi P, Meier B. Patent foramen ovale and neurosurgery in sitting position: a systematic review. Br J Anaesth 2009;102(5):588–96.
8. Rozet I, Vavilala MS. Risks and benefits of patient positioning during neurosurgical care. Anesthesiol Clin 2007;25(3):631–53.
9. Orliaguet GA, Hanafi M, Meyer PG, Blanot S, Jarreau MM, Bresson D, Zerah M, Carli PA. Is the sitting or the prone position best for surgery
for posterior fossa tumours in children? Pediatr Anesth 2001;11(5):541–7.
10. Black S, Ockert DB, Oliver Jr WC, Cucchiara RF. Outcome following posterior fossa craniectomy in patients in the sitting or horizontal positions. Anesthesiology 1988;69(1):49–56.
11. Leonard IE, Cunningham AJ. The sitting position in neurosurgery—not yet obsolete!. Br J Anaesthes 2002;88(1):1–3.
12. Schaffranietz L1, Grothe A, Olthoff D. Use of the sitting position in neurosurgery. Results of a 1998 survey in Germany. Anaesthesist
2000;49(4):269–74.
13. Kida H, Nishikawa N, Matsunami K, Kawahito M, Ota M, Miyao S. Sitting position in the neurosurgery: the results of a questionnaire sent
to neurosurgeons of medical colleges. Masui 2000;49(5):566–9.
14. Rath GP, Bithal PK, Chaturvedi A, Dash HH. Complications related to positioning in posterior fossa craniectomy. J Clin Neurosci
2007;14(6):520–5.
15. Misra BK, Purandare HR, Ved RS, Bagdia AA, Mare PB. Current treatment strategy in the management of vestibular schwannoma. Neurol
India 2009;57(3):257–63.
16. Duke DA, Lynch JJ, Harner SG, Faust RJ, Ebersold MJ. Venous air embolism in sitting and supine patients undergoing vestibular schwannoma resection. Neurosurgery 1998;42(6):1282–6. discussion 1286–7.
17. Tsaousi GG, Karakoulas KA, Amaniti EN, Soultati ID, Zouka MD, Vasilakos DG. Correlation of central venous-arterial and mixed venousarterial carbon dioxide tension gradient with cardiac output during neurosurgical procedures in the sitting position. Eur J Anaesthesiol
2010;27(10):882–9.
18. Closhen D, Berres M, Werener C, Engelhard K, Schramm P. Influence of beach chair position on cerebral oxygen saturation: a comparison of
INVOS and FORE-SIGHT cerebral oximeter. J Neurosurg Anesthesiol 2013;25(4):414–9.
19. Mirski MA, Lele AV, Fitzsimmons L, Toung TJ. Diagnosis and treatment of vascular air embolism. Anesthesiology 2007;106(1):164–77.
20. Girard F, Ruel M, McKenty S, Boudreault D, Chouinard P, Todorov A, Molina-Negro P, Bouvier G. Incidences of venous air embolism and
patent foramen ovale among patients undergoing selective peripheral denervation in the sitting position. Neurosurgery 2003;53(2):316–9.
discussion 319–20.
VII. NEUROSURGERY
274
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
21. Mammoto T, Hayashi Y, Ohnishi Y, Kuro M. Incidence of venous and paradoxical air embolism in neurosurgical patients in the sitting position: detection by transesophageal echocardiography. Acta Anaesthesiol Scand 1998;42(6):643–7.
22. Porter JM, Pidgeon C, Cunningham AJ. The sitting position in neurosurgery: a critical appraisal. Br J Anaesthes 1999;82(1):117–28.
23. Matjasko J, Petrozza P, Cohen M, Steinberg P. Anesthesia and surgery in the seated position: analysis of 554 cases. Neurosurgery
1985;17(5):695–702.
24. von Gösseln HH, Samii M, Suhr D, Bini W. The lounging position for posterior fossa surgery: anesthesiological considerations regarding air
embolism. Childs Nerv Sys 1991;7(7):368–74.
25. Jadik S, Wissing H, Friedrich K, Beck J, Seifert V, Raabe A. A standardized protocol for the prevention of clinically relevant venous air embolism during neurosurgical interventions in the semi-sitting position. Neurosurgery 2009;64(3):533–8.
26. Gracia I, Fabregas N. Craniotomy in sitting position: anesthesiology management. Curr Opin Anaesthesiol 2014;27(5):474–83.
27. Munson ES, Merrick HC. Effect of nitrous oxide on venous air embolism. Anesthesiology 1966;27(6):783–7.
28. Singh GP, Prabhakar H, Bithal PK, Dash HH. A comparative evaluation of nitrous oxide-isoflurane vs isoflurane anesthesia in patients
undergoing craniotomy for supratentorial tumors: a preliminary study. Neurol India 2011;59(1):18–24.
29. Todd MM, Warner DS, Sokoll MD, Maktabi MA, Hindman BJ, Scamman FL, Kirschner J. A prospective, comparative trial of three anesthetics for elective supratentorial craniotomy. Propofol/fentanyl, isoflurane/nitrous oxide, and fentanyl/nitrous oxide. Anesthesiology
1993;7896:1005.
30. Pasternak JJ, McGregor DG, Lanier WL, Schroeder DR, Rusy DA, Hindman B, Clarke W, Torner J, Todd MM, IHAST Investigators. Effect of
nitrous oxide use on long-term neurologic and neuropsychological outcome in patients who received temporary proximal artery occlusion
during cerebral aneurysm clipping surgery. Anesthesiology 2009;110(3):563–73.
31. Myles PS, Leslie K, Chan MT, Forbes A, Peyton PJ, Paech MJ, Beattie WS, Sessler DI, Devereaux PJ, Silbert B, Schricker T, Wallace S, ANZCA
Trials Group for the ENIGMA-II investigators. The safety of addition of nitrous oxide to general anaesthesia in at-risk patients having major
non-cardiac surgery (ENIGMA-II): a randomised, single-blind trial. Lancet 2014;384(9952):1446–54.
32. Culley DJ, Crosby G. Nitrous oxide in neuroanesthesia: tried and true or toxin? Anesthesiology 2008;108(4):553–4.
33. Pasternak JJ, Lanier WL. Is nitrous oxide use appropriate in neurosurgical and neurologically at-risk patients? Curr Opin Anaesthesiol
2010;23(5):544–50.
34. Artru AA. Nitrous oxide plays a direct role in the development of tension pneumocephalus intraoperatively. Anesthesiology 1982;57(1):59–61.
35. Hernandez-Palazon J, Martinez-Lage JF, de la Rosa-Carrillo VN, Tortosa JA, Lopez F, Poza M. Anesthetic technique and development of
pneumocephalus after posterior fossa surgery in the sitting position. Neurocirugia (Asturias) 2003;14(3):216–21.
36. Friedman GA, Norfleet EA, Bedford RF. Discontinuance of nitrous oxide does not prevent tension pneumocephalus. Anesth Analg
1981;60(1):57–8.
37. Domino KB, Hemstad JR, Lam AM, Laohaprasit V, Mayberg TA, Harrison SD, Grady MS, Winn HR. Effect of nitrous oxide on intracranial
pressure after cranial-dural closure in patients undergoing craniotomy. Anesthesiology 1992;77(3):421–5.
38. Losasso TJ, Muzzi DA, Dietz NM, Cucchiara RF. Fifty percent nitrous oxide does not increase the risk of venous air embolism in neurosurgical patients operated upon in the sitting position. Anesthesiology 1992;77(1):21–30.
39. Hübler M, Petrasch F. Intraoperative kinking of polyvinyl endotracheal tubes. Anesth Analg 2006;103(6):1601–2.
40. Ogden LL, Bradway JA. Maneuver to relieve kinking of the endotracheal tube in a prone patient. Anesthesiology 2008;109(1):159.
41. Schlundt J, Tzanova I, Werner C. A case of intrapulmonary transmission of air while transitioning a patient from a sitting to a supine position
after venous air embolism during a craniotomy. Can J Anaesth 2012;59(5):478–82.
42. Meyer PG, Cuttaree H, Charron B, Jarreau MM, Perie AC, Sainte-Rose C. Prevention of venous air embolism in paediatric neurosurgical
procedures performed in the sitting position by combined use of MAST suit and PEEP. Br J Anaesth 1994;73(6):795–800.
43. Zasslow MA, Pearl RG, Larson CP, Silverberg G, Shuer LF. PEEP does not affect left atrial-right atrial pressure difference in neurosurgical
patients. Anesthesiology 1988;68(5):760–3.
44. Schmitt HJ, Hemmerling TM. Venous air emboli occur during release of positive end-expiratory pressure and repositioning after sitting position surgery. Anesth Analg 2002;94(2):400–3.
45. Perkins NA, Bedford RF. Hemodynamic consequences of PEEP in seated neurological patients–implications for paradoxical air embolism.
Anesth Analg 1984;63(4):429–32.
46. Giebler R, Kollenberg B, Pohlen G, Peters J. Effect of positive end-expiratory pressure on the incidence of venous air embolism and on the
cardiovascular response to the sitting position during neurosurgery. Br J Anaesth 1998;80(1):30–5.
47. Hervias A, Valero R, Hurtado P, Gracia I, Perelló L, Tercero FJ, González JJ, Fàbregas N. Detection of venous air embolism and patent foramen
ovale in neurosurgery patients in sitting position. Neurocirugia (Astur) 2014;25(3):108–15.
48. Manninen PH. Spontaneous ventilation is a useful monitor of brain stem function during posterior fossa surgery. J Neurosurg Anesthesiol
1995;7(1):63–5.
49. Pandia MP, Bithal PK, Sharma MS, Bhagat H, Prasanna B. Use of spontaneous ventilation to monitor the effects of posterior fossa surgery in
the sitting position. J Clin Neurosci 2009;16(7):968–9.
50. Schäfer ST, Sandalcioglu IE, Stegen B, Neumann A, Asgari S, Peters J. Venous air embolism during semi- sitting craniotomy evokes thrombocytopenia. Anaesthesia 2011;66(1):25–30.
51. Lindroos AC, Niiya T, Randell T, Niemi TT. Stroke volume-directed administration of hydroxyethyl starch (HES 130/0.4) and Ringer’s
acetate in prone position during neurosurgery: a randomized controlled trial. J Anesth 2014;28(2):189–97.
52. Kwak HJ, Lee D, Lee YW, Yu GY, Shinn HK, Kim JY. The intermittent sequential compression device on the lower extremities attenuates the
decrease in regional cerebral oxygen saturation during sitting position under sevoflurane anesthesia. J Neurosurg Anesthesiol 2011;23(1):1–5.
53. McCulloch TJ, Liyanagama K, Petchell J. Relative hypotension in the beach-chair position: effects on middle cerebral artery blood velocity.
Anaesth Intensive Care 2010;38(3):486–91.
54. Murphy GS, Szokol JW, Marymont JH, Greenberg SB, Avram MJ, Vender JS, Vaughn J, Nisman M. Cerebral oxygen desaturation events
assessed by near-infrared spectroscopy during shoulder arthroscopy in the beach chair and lateral decubitus positions. Anesth Analg
2010;111(2):496–505.
VII. NEUROSURGERY
REFERENCES
275
55. Kosty JA, Leroux PD, Levine J, Park S, Kumar MA, Frangos S, Maloney-Wilensky E, Kofke WA. Brief report: a comparison of clinical and
research practices in measuring cerebral perfusion pressure: a literature review and practitioner survey. Anesth Analg 2013;117(3):694–8.
56. Meng L, Cannesson M, Alexander BS, Yu Z, Kain ZN, Cerussi AE, Tromberg BJ, Mantulin WW. Effect of phenylephrine and ephedrine bolus
treatment on cerebral oxygenation in anaesthetized patients. Br J Anaesth 2011;107(2):209–17.
57. Soeding PF, Hoy S, Hoy G, Evans M, Royse CF. Effect of phenylephrine on the haemodynamic state and cerebral oxygen saturation during
anaesthesia in the upright position. Br J Anaesth 2013;111(2):229–34.
58. Brassard P, Seifert T, Secher NH. Is cerebral oxygenation negatively affected by infusion of norepinephrine in healthy subjects? Br J Anaesth
2009;102(6):800–5.
59. Prabhakar H, Anand N, Chouhan RS, Bithal PK. Sudden asystole during surgery in the cerebellopontine angle. Acta Neurochirurg (Wien)
2006;148(6):699–700. discussion 700.
60. Goyal K, Philip FA, Rath GP, Mahajan C, Sujatha M, Bharti SJ, Gupta N. Asystole during posterior fossa surgery: report of two cases. Asian J
Neurosurg 2012;7(2):87–9.
61. Prabhakar H, Rath GP, Dash HH. Bradycardia following hydrogen peroxide irrigation during posterior fossa surgery. Anaesthesia
2006;61(9):914.
62. Bijker JB, Gelb AW. Review article: the role of hypotension in perioperative stroke. Can J Anesth 2013;60(2):159–67.
63. Jahangiri FR, Minhas M, Jane Jr J. Preventing lower cranial nerve injuries during fourth ventricle tumor resection by utilizing intraoperative
neurophysiological monitoring. Neurodiagn J 2012;52(4):320–32.
64. Deinsberger W, Christophis P, Jödicke A, Heesen M, Böker DK. Somatosensory evoked potential monitoring during positioning of the
patient for posterior fossa surgery in the semisitting position. Neurosurgery 1998;43(1):36–40. discussion 40–2.
65. Wiedemayer H, Sandalcioglu IE, Armbruster W, Regel J, Schaefer H, Stolke D. False negative findings in intraoperative SEP neurosurgical
cases and review of published monitoring: analysis of 658 consecutive reports. J Neurol Neurosurg Psychiatry 2004;75(2):280–6.
66. Simon MV. Neurophysiologic intraoperative monitoring of the vestibulocochlear nerve. J Clin Neurophysiol 2011;28(6):566–81.
67. Loiselle DL, Nuwer MR. When should we warn the surgeon? Diagnosis based warning criteria for BAEP monitoring. Neurology
2005;65(10):1522–3.
68. Acioly MA, Liebsch M, de Aguiar PH, Tatagiba M. Facial nerve monitoring during cerebellopontine angle and skull base tumor surgery: a
systematic review from description to current success on function prediction. World Neurosurg 2013;80(6):e271–300.
69. Topsakal C, Al-Mefty O, Bulsara KR, Williford VS. Intraoperative monitoring of lower cranial nerves in skull base surgery: technical report
and review of 123 monitored cases. Neurosurg Rev 2008;31(1):45–53.
70. Lobato EB, Black S, De Soto H. Venous air embolism and selective denervation for torticollis. Anesthes Analg 1997;84(3):551–3.
71. Young ML, Smith DS, Murtagh F, Vasquez A, Levitt J. Comparison of surgical and anesthetic complications in neurosurgical patients experiencing venous air embolism in the sitting position. Neurosurgery 1986;18(2):157–61.
72. Harrison EA, Mackersie A, McEwan A, Facer E. The sitting position for neurosurgery in children: a review of 16 years’ experience. Br J
Anaesth 2002;88(1):12–7.
73. Bithal PK, Pandia MP, Dash HH, Chouhan RS, Mohanty B, Padhy N. Comparative incidence of venous air embolism and associated hypotension in adults and children operated for neurosurgery in the sitting position. Eur J Anaesth 2004;21(7):517–22.
74. Toung TJ, Rossberg MI, Hutchins GM. Volume of air in a lethal venous air embolism. Anesthesiology 2001;94(2):360–1.
75. Moningi S, Kulkarni D, Bhattacharjee S. Coagulopathy following venous air embolism: a disastrous consequence -a case report. Korean J
Anesth 2013;65(4):349–52.
76. Flisberg P, Rundgren M, Engstrom M. The effects of platelet transfusions evaluated using rotational thromboelastometry. Anesth Analg
2009;108(5):1430–2.
77. Wong AY, Irwin MG. Large venous air embolism in the sitting position despite monitoring with transoesophageal echocardiography.
Anaesthesia 2005;60(8):811–3.
78. Schubert A, Deogaonkar A, Drummond JC. Precordial Doppler probe placement for optimal detection of venous air embolism during craniotomy. Anesth Analg 2006;102(5):1543–7.
79. Pandia MP, Bithal PK, Dash HH, Chaturvedi A. Comparative incidence of cardiovascular changes during venous air embolism as detected by
transesophageal echocardiography alone or in combination with end tidal carbon dioxide tension monitoring. J Clin Neurosci 2011;18(9):1206–9.
80. Vinay B, Sriganesh K, Gopala Krishna KN. An abrupt reduction in end-tidal carbon dioxide during neurosurgery is not always due to venous
air embolism: a capnograph artefact. J Clin Monit Comput 2014;28(2):217–9.
81. Schäfer ST, Lindemann J, Brendt P, Kaiser G, Peters J. Intracardiac transvenous echocardiography is superior to both precordial doppler
and transesophageal echocardiography techniques for detecting venous air embolism and catheter-guided air aspiration. Anesth Analg
2008;106(1):45–54.
82. Bunegin L, Albin MS, Helsel PE, Hoffman A, Hung TK. Positioning the right atrial catheter: a model for reappraisal. Anesthesiology
1981;55(4):343–8.
83. Reeves ST, Bevis LA, Bailey BN. Positioning a right atrial air aspiration catheter using transesophageal echocardiography. J Neurosurg
Anesthesiol 1996;8(2):123–5.
84. Domaingue CM. Anaesthesia for neurosurgery in the sitting position: a practical approach. Anaesth Intensive Care 2005;33(3):323–31.
85. Meissner I, Khandheria BK, Heit JA, Petty GW, Sheps SG, Schwartz GL, Whisnant JP, Wiebers DO, Covalt JL, Petterson TM, Christianson TJ,
Agmon Y. Patent foramen ovale: innocent or guilty? Evidence from a prospective population-based study. J Am Coll Cardiol 2006;47(2):440–5.
86. Perkins-Pearson NA, Marshall WK, Bedford RF. Atrial pressures in the seated position: implication for paradoxical air embolism.
Anesthesiology 1982;57(6):493–7.
87. Black S, Cucchiara RF, Nishimura RA, Michenfelder JD. Parameters affecting occurrence of paradoxical air embolism. Anesthesiology
1989;71(2):235–41.
88. Buchholz S, Shakil A, Figtree GA, Hansen PS, Bhindi R. Diagnosis and management of patent foramen ovale. Postgrad Med J 2012;88(1038):217–25.
89. Engelhardt M, Folkers W, Brenke C, Scholz M, Harders A, Fidorra H, Schmieder K. Neurosurgical operations with the patient in sitting position: analysis of risk factors using transcranial doppler sonography. Br J Anaesth 2006;96(4):467–72.
VII. NEUROSURGERY
276
14. ANESTHESIA FOR POSTERIOR FOSSA SURGERY
90. Wahl A, Kunz M, Moschovitis A, Nageh T, Schwerzmann M, Seiler C, Mattle HP, Windecker S, Meier B. Long-term results after fluoroscopyguided closure of patent foramen ovale for secondary prevention of paradoxical embolism. Heart 2008;94(3):336–41.
91. Losasso TJ, Muzzi DA, Cucchiara RF. Jugular venous compression helps to identify the source of venous air embolism during craniectomy
in patients in the sitting position. Anesthesiology 1992;76(1):156–7.
92. Durant TM, Long J, Oppenheimer MJ. Pulmonary (venous) air embolism. Am Heart J 1947;33(3):269–81.
93. Shaffner DH, Heitmiller ES, Deshpande JK. Pediatric perioperative life support. Anesth Analg 2013;117(4):960–79.
94. Jørgensen TB, Sørensen AM, Jansen EC. Iatrogenic systemic air embolism treated with hyperbaric oxygen therapy. Acta Anasthesiol Scand
2008;52(4):566–8.
95. Cata JP, Saager L, Kurz A, Avitsian R. Successful extubation in the operating room after infratentorial craniotomy: the Cleveland clinic experience. J Neurosurg Anesthesiol 2011;23(1):25–9.
96. Tondon A, Mahapatra AK. Supratentorial intracerebral hemorrhage following infratentorial surgery. J Clin Neurosci 2004;11(7):762–5.
97. Vimala S, Reddy MK, Rao UG. Non-awakening from anesthesia following posterior fossa surgery due to skull pin-induced tension pneumocephalus. Neurol India 2011;59(4):641–2.
98. Gupta N, Rath GP, Mahajan C, Dube SK, Sharma S. Tension pneumoventricle after excision of third ventricular tumor in sitting position. J
Anaesthesiol Clin Pharmacol 2011;27(3):409–11.
99. Thibault M, Girard F, Moumdjian R, Chouinard P, Boudreault D, Ruel M. Craniotomy site influences postoperative pain following neurosurgical procedures: a retrospective study. Can J Anaesth 2007;54(7):544–8.
100. Gottschalk A, Berkow LC, Stevens RD, Mirski M, Thompson RE, White ED, Weingart JD, Long DM, Yaster M. Prospective evaluation of pain
and analgesic use following major elective intracranial surgery. J Neurosurg 2007;106(2):210–6.
101. Flexman AM, Merriman B, Griesdale DE, Mayson K, Choi PT, Ryerson CJ. Infratentorial neurosurgery is an independent risk factor for
respiratory failure and death in patients undergoing intracranial tumor resection. J Neurosurg Anesthesiol 2014;26(3):198–204.
102. Dubey A, Sung WS, Shaya M, Patwardhan R, Willis B, Smith D, Nanda A. Complications of posterior cranial fossa surgery—an institutional
experience of 500 patients. Surg Neurol 2009;72(4):369–75.
103. Sloan T. The incidence, volume, absorption, and timing of supratentorial pneumocephalus during posterior fossa neurosurgery conducted
in the sitting position. J Neurosurg Anesthesiol 2010;22(1):59–66.
104. Toung T, Donham RT, Lehner A, Alano J, Campbell J. Tension pneumocephalus after posterior fossa craniotomy: report of four additional
cases and review of postoperative pneumocephalus. Neurosurgery 1983;12(2):164–8.
105. Hindman BJ, Palecek JP, Posner KL, Traynelis VC, Lee LA, Sawin PD, Tredway TL, Todd MM, Domino KB. Cervical spinal cord, root, and
bony spine injuries a closed claims analysis. Anesthesiology 2011;114(4):729–31.
106. Haisa T, Kondo T. Midcervical flexion myelopathy after posterior fossa surgery in the sitting position: case report. Neurosurgery
1996;38(4):819–21.
107. Edgcombe H, Carter K, Yarrow S. Anaesthesia in the prone position. Br J Anaesth 2008;100(2):165–83.
108. Bhardwaj A, Long DM, Ducker TB, Toung TJ. Neurologic deficits after cervical laminectomy in the prone position. J Neurosurg Anesthesiol
2001;13(4):314–9.
109. Chowdhury T, Gupta N, Rath GP. Macroglossia in a child undergoing posterior fossa surgery in sitting position. Saudi J Anaesth 2012;6(1):85–6.
VII. NEUROSURGERY
C H A P T E R
15
Transesophageal Echocardiography
A. Lele, V. Krishnamoorthy
University of Washington, Seattle, WA, United States
O U T L I N E
Introduction
277
Basics of Transesophageal Echocardiography
277
Common Indications for Utilization of Transesophageal
Echocardiography in Neuroanesthesia
278
Screening for Venous Air Embolism During
Neurosurgical Procedures
278
Absolute Contraindications
279
Relative Contraindications
279
Steps in Preparation for Placement of
Transesophageal Echocardiography Probe
Under Anesthesia
279
Screening, Risk Stratification, and Preparation of
Patients at High Risk for Vascular Air Embolism
Prior to Proposed Neurosurgical Procedures
280
Verification of Multiorifice Catheter Placement
for High-Risk Neurosurgical Procedures and
Ventriculoatrial Shunts
Monitoring Intraoperative Cardiac Function in
Patients With Cardiomyopathy
280
281
Complications Associated With Transesophageal
Echocardiography
282
Advantages and Disadvantages of Using Transesophageal
Echocardiography as an Intraoperative Monitor
282
Summary
283
References
283
INTRODUCTION
The ability to visualize the chambers of the heart during neurosurgical procedures provides the anesthesiologist
with data regarding cardiac function, valvular abnormalities, and presence or absence of intracardiac shunts; helps
to visualize intracardiac extraneous material such as clots and air; and helps facilitate certain bedside hemodynamic
and neurosurgical procedures.
Via a detailed literature search, this chapter reviews the utilization of transesophageal echocardiography (TEE) in
the management of patients during neuroanesthesia. Upon reading the chapter, the reader will be able to understand
the common indications, procedural characteristics, and practical applications of this technique.
BASICS OF TRANSESOPHAGEAL ECHOCARDIOGRAPHY
Unlike surface cardiography, TEE allows real-time visualization of various cardiac chambers. It is facilitated via
placement of a TEE probe into the esophagus aimed anteriorly at the cardiac chambers. Due to the proximity to the
heart, TEE examination generally provides images with a high degree of spatial resolution. With the probe in place,
cardiac walls, valves, interatrial and interventricular septum, pericardial structures, and left and right outflow tract
structures can be easily visualized. A traditional complete TEE examination covers such views.
Essentials of Neuroanesthesia
http://dx.doi.org/10.1016/B978-0-12-805299-0.00015-4
277
© 2017 Elsevier Inc. All rights reserved.
278
15. TRANSESOPHAGEAL ECHOCARDIOGRAPHY
Common Indications for Utilization of Transesophageal Echo
Download